You are on page 1of 411

World Health Organization Classification of Tumours

Delellis R.A., Lloyd RV, Heitz P.U., Bosman F.T., Carneiro F., Travis WO., Brambilla E., Burke A.P.,
Eng C. (Eds): World Health Hruban R.H., Theise N.D. (Eds): Marx A., Nicholson A.G. (Eds)
Organization Classification of WHO Classification of Tumours of the WHO Classification of Tumours of the
Tumours. Pathology and Genetics of Digestive System (4th edition). Lung, Pleura, Thymus and Heart
Tumours of Endocrine Organs IARC: Lyon 2010. ( 4th edition). IARC: Lyon 2015.
(3rd edition). IARC Press: Lyon 2004. ISBN 978-92-832-2432-7 ISBN 978-92-832-2436-5
ISBN 978-92-832-2416-7

Barnes L., Eveson J.W., Reichart P., Lakhani S.R., Ellis 10., Schnitt S.J . Moch H., Humphrey P.A.,
Sidransky D. (Eds): World Health Tan P.H., van de Vijver M.J. (Eds): Ulbright T.M., Reuter V.E. (Eds):
Organization Classification of WHO Classification of Tumours of the WHO Classification of Tumours of
Tumours. Pathology and Genetics of Breast (4th edition). IARC: Lyon 2012. the Urinary System and Male Genital
Head and Neck Tumours (3rd ISBN 978-92-832-2433-4 Organs (4th edition).
edition). IARC Press: Lyon 2005. IARC: Lyon 2016.
ISBN 978-92-832-2417-5 ISBN 978-92-832-2437-2

LeBoit P.E., Burg G .. Weedon D . Fletcher C.D M . Bridge J.A., Louis D.N., Ohgaki H.,
Sarasin A. (Eds): World Health Hogendoorn P.C.W., Mertens F. Wiestler O.D., Cavenee W.K. (Eds):
Organization Classification of (Eds): WHO Classification of Tumours WHO Classification of Tumours of the
Tumours. Pathology and Genetics of of Soft Tissue and Bone (4th edition). Central Nervous System (Revised 4th
Skin Tumours (3rd edition). IARC: Lyon 2013. edition). IARC: Lyon 2016.
IARC Press: Lyon 2006. ISBN 978-92-832-2434-1 ISBN 978-92-832-4492-9
ISBN 978-92-832-2414-0

Swerdlow S.H.. Campo S .. Kurman R.J., Carcangiu M.L.,


Harris N.L., Jaffe E.S. Pileri S.A., Herrington C S .. Young R.H. (Eds):
Stein H., Thiele J. Vardiman J.W. WHO Classification of Tumours of
(Eds): WHO Classification of Tumours Female Reproductive Organs
of Haematopoietic and Lymphoid (4th edition). IARC: Lyon 2014.
Tissues (4th edition) ISBN 978-92-832-2435-8
IARC: Lyon 2008.
ISBN 978-92-832-2431-0

This book and all other volumes of the series can be purchased:

From all countries From USA From Canada

WHO Press Stylus Publishing, LLC Renouf Publishing Company Limited


World Health Organization 22883 Quicksilver Drive PO Box 605 22-1010 Polytek Street
CH-1211 Geneva 27 Herndon, VA 20172-05 Ottawa, ON K 1 J 9J 1
Switzerland Tel. + 1 800 232 0223 Tel. + 1 866 767 6766
Tel. +4122791 3264 or + 1 703 661 1581 Fax +1 613 745 7660
Fax +41 22 791 4857 Fax +17036611501 orders@renoufbooks.com
bookorders@who.int sty I usmai l@presswarehouse.com www.renoufbooks.com
www.who.int/bookorders/ www.styluspub.com
�··
World Health Organization Classification of Tumours

WHO
-�
�� , �-1� IJJI
��
OMS

International Agency for Research on Cancer (IARC)

Revised 4th Edition

WHO Classification of Tumours of


the Central Nervous System

Edited by

David N. Louis
Hiroko Ohgaki
Otmar D. Wiestler
Webster K. Cavenee

International Agency for Research on Cancer


Lyon, 2016
World Health Organization Classification of Tumours

Series Editors Fred T. Bosman, MD PhD


Elaine S. Jaffe, MD
Sunil R. Lakhani, MD FRCPath
Hiroko Ohgaki, PhD

WHO Classification of Tumours of the Central Nervous System


Revised 4th Edition

Editors David N. Louis, MD


Hiroko Ohgaki, PhD
Otmar D. Wiestler, MD
Webster K. Cavenee, PhD

Senior Advisors David W. Ellison, MD PhD


Dominique Figarella-Branger, MD
Arie Perry, MD
Guido Reifenberger, MD
Andreas von Deimling, MD

Project Coordinator Paul Kleihues, MD

Project Assistant Asiedua Asante

Technical Editor Jessica Cox

Database Kees Kleihues-van Toi

Layout Stefanie Brottrager

Printed by Maestro
38330 Saint-lsmier, France

Publisher International Agency for


Research on Cancer (IARC)
69372 Lyon Cedex 08, France
This volume was produced with support from and in collaboration with the

German Cancer Research Center

dkfz. GERMAN
CANCER RESEARCH CENTER
IN THE HELMHOLTZ ASSOCIATION

The WHO Classification of Tumours of the Central Nervous System


presented in this book reflects the views of a Working Group that convened for a
Consensus and Editorial Meeting at the German Cancer Research Center,
Heidelberg, 21-24 June 2015.

Members of the Working Group are indicated


in the list of contributors on pages 342-348.
Published by the International Agency for Research on Cancer (IARC)
150 Cours Albert Thomas, 69372 Lyon Cedex 08, France

© International Agency for Research on Cancer, 2016

Distributed by
WHO Press, World Health Organization, 20 Avenue Appia, 1211 Geneva 27, Switzerland
Tel.: +41 22 791 3264; Fax: +41 22 791 4857; email: bookorders@who int

Publications of the World Health Organization enjoy copyright protection in accordance with the
provisions of Protocol 2 of the Universal Copyright Convention. All rights reserved.

The designations employed and the presentation of the material in this publication do not imply the
expression of any opinion whatsoever on the part of the Secretariat of the World Health Organization
concerning the legal status of any country, territory, city, or area or of its authorities, or concerning the
delimitation of its frontiers or boundaries.

The mention of specific companies or of certain manufacturers' products does not imply that they are
endorsed or recommended by the World Health Organization in preference to others of a similar nature
that are not mentioned. Errors and omissions excepted, the names of proprietary products are
distinguished by initial capital letters.

The authors alone are responsible for the views expressed in this publication.

The copyright of figures and charts remains with the authors.


(See Sources of figures and tables, pages 351-355.)

First print run {10 000 copies)

Format for bibliographic citations:


David N. Louis, Hiroko Ohgaki, Otmar 0. Wiestler, Webster K. Cavenee (Eds):
WHO Classification of Tumours of the Central Nervous System (Revised 4th edition).

J
IARC: Lyon 2016.

I
I
IARC Library Cataloguing in Publication Data

II
WHO classification of tumours of the central nervous system I edited by David N. Louis, Hiroko Ohgaki, Otmar 0. Wiestler,
Webster K. Cavenee. - Revised 4th edition.

(World Health Organization classification of tumours)

1. Central Nervous System Neoplasms - genetics


j:.
2. Central Nervous System Neoplasms - pathology
I. Louis David N I
ISBN 978-92-832-4492-9 (NLM Classification: WJ 160)
#'

Contents
WHO classification 10 Anaplastic ganglioglioma 141
Introduction: WHO classification and grading of tumours Dysplastic cerebellar gangliocytoma
of the central nervous system 12 (Lhermitte-Duclos disease) 142
Desmoplastic infantile astrocytoma and ganglioglioma 144
Diffuse astrocytic and oligodendroglial tumours 15 Papillary glioneuronal tumour 147
Introduction 16 Rosette-forming glioneuronal tumour 150
Diffuse astrocytoma, IDH-mutant 18 Diffuse leptomeningeal glioneuronal tumour 152
Gemistocytic astrocytoma, IDH-mutant 22 Central neurocytoma 156
Diffuse astrocytoma, IDH-wildtype 23 Extraventricular neurocytoma 159
Diffuse astrocytoma, NOS 23 Cerebellar liponeurocytoma 161
Anaplastic astrocytoma, IDH-mutant 24 Paraganglioma 164
Anaplastic astrocytoma, IDH-wildtype 27
Anaplastic astrocytoma, NOS 27 7 Tumours of the pineal region 169
Glioblastoma, IDH-wildtype 28 Pineocytoma 170
Giant cell glioblastoma 46 Pineal parenchymal tumour of intermediate differentiation 173
Gliosarcoma 48 Pineoblastoma 176
Epithelioid glioblastoma 50 Papillary tumour of the pineal region 180
Glioblastoma, IDH-mutant 52
Glioblastoma, NOS 56 8 Embryonal tumours 183
Diffuse midline glioma, H3 K27M-mutant 57 Medulloblastoma 184
Oligodendroglioma, IDH-mutant and 1p/19q-codeleted 60 Medulloblastoma, NOS 186
Oligodendroglioma, NOS 69 Meduf/oblastomas. genetically defined 188
Anaplastic oligodendroglioma, IDH-mutant and Medulloblastoma, WNT-activated 188
1 p/19q-codeleted 70 Medulloblastoma, SHH-activated and TP53-mutant 190
Anaplastic oligodendroglioma, NOS 74 Medulloblastoma, SHH-activated and TP53-wildtype 190
Oligoastrocytoma, NOS 75 Medulloblastoma, non-WNT/non-SHH 193
Anaplastic oligoastrocytoma, NOS 76 Medulloblastomas, histological/y defined 194
Medulloblastoma, classic 194
2 Other astrocytic tumours 79 Desmoplastic/nodular medulloblastoma 195
Pilocytic astrocytoma 80 Medulloblastoma with extensive nodularity 198
Pilomyxoid astrocytoma 88 Large cell I anaplastic medulloblastoma 200
Subependymal giant cell astrocytoma 90 Embryonal tumour with multilayered rosettes,
Pleomorphic xanthoastrocytoma 94 C 19MC-altered 201
Anaplastic pleomorphic xanthoastrocytoma 98 Embryonal tumour with multilayered rosettes. NOS 205
Other CNS embryonal tumours 206
3 Ependymal tumours 101 Medulloepithelioma 207
Subependymoma 102 CNS neuroblastoma 207
Myxopapillary ependymoma 104 CNS ganglioneuroblastoma 207
Ependymoma 106 CNS embryonal tumour, NOS 208
Papillary ependymoma 111 Atypical teratoid/rhabdoid tumour 209
Clear cell ependymoma 111 CNS embryonal tumour with rhabdoid features 212
Tanycytic ependymoma 111
Ependymoma, RELA fusion-positive 112 9 Tumours of the cranial and paraspinal nerves 213
Anaplastic ependymoma 113 Schwannoma 214
Cellular schwannoma 216
4 Other gliomas 115 Plexiform schwannoma 217
Chordoid glioma of the third ventricle 116 Melanotic schwannoma 218
Angiocentric glioma 119 Neurofibroma 219
Astroblastoma 121 Atypical neurofibroma 220
Plexiform neurofibroma 220
5 Choroid plexus tumours 123 Perineurioma 222
Choroid plexus papilloma 124 Hybrid nerve sheath tumours 224
Atypical choroid plexus papilloma 126 Malignant peripheral nerve sheath tumour (MPNST) 226
Choroid plexus carcinoma 128 MPNST with divergent differentiation 227
Epithelioid MPNST 228
6 Neuronal and mixed neuronal-glial tumours 131 MPNST with perineurial differentiation 228
Dysembryoplastic neuroepithelial tumour 132
Gangliocytoma 136 10 Meningiomas 231
Ganglioglioma 138 Meningioma 232
.,,.

Meningioma variants 237 lntravascular large B-cell lymphoma 276


Meningothelial meningioma 237 Miscellaneous rare lymphomas in the CNS 276
Fibrous meningioma 237 Low-grade B-cell lymphomas 276
Transitional meningioma 238 T-cell and NK!T-cell lymphomas 276
Psammomatous meningioma 238 Anaplastic large cell lymphoma (ALK+/ALK-) 277
Angiomatous meningioma 238 MALT lymphoma of the dura 277
Microcystic meningioma 239
Secretory meningioma 239 14 Histiocytic tumours 279
Lymphoplasmacyte-rich meningioma 240 Langerhans cell histiocytosis 280
Metaplastic meningioma 240 Erdheim-Chester disease 281
Chordoid meningioma 240 Rosai-Dorfman disease 282
Clear cell meningioma 241 Juvenile xanthogranuloma 282
Atypical meningioma 241 Histiocytic sarcoma 283
Papillary meningioma 242
Rhabdoid meningioma 243 15 Germ cell tumours 285
Anaplastic (malignant) meningioma 244 Germinoma 288
Embryonal carcinoma 289
11 Mesenchymal, non-meningothelial tumours 247 Yolk sac tumour 290
Solitary fibrous tumour I haemangiopericytoma 249 Choriocarcinoma 290
Haemangioblastoma 254 Teratoma 291
Haemangioma 258 Mature teratoma 291
Epithelioid haemangioendothelioma 258 Immature teratoma 291
Angiosarcoma 259 Teratoma with malignant transformation 291
Kaposi sarcoma 259 Mixed germ cell tumour 291
Ewing sarcoma I peripheral primitive
neuroectodermal tumour 259 16 Familial tumour syndromes 293
Lipoma 260 Neurofibromatosis type 1 294
Angiolipoma 260 Neurofibromatosis type 2 297
Hibernoma 260 Schwannomatosis 301
Liposarcoma 260 Von Hippel-Lindau disease 304
Desmoid-type fibromatosis 260 Tuberous sclerosis 306
Myofibroblastoma 260 Li-Fraumeni syndrome 310
Inflammatory myofibroblastic tumour 261 Cowden syndrome 314
Benign fibrous histiocytoma 261 Turco! syndrome 317
Fibrosarcoma 261 Mismatch repair cancer syndrome 317
Undifferentiated pleomorphic sarcoma I malignant Familial adenomatous polyposis 318
fibrous histiocytoma 261 Naevoid basal cell carcinoma syndrome 319
Leiomyoma 262 Rhabdoid tumour predisposition syndrome 321
Leiomyosarcoma 262
Rhabdomyoma 262 17 Tumours of the sellar region 323
Rhabdomyosarcoma 262 Craniopharyngioma 324
Chondroma 262 Adamantinomatous craniopharyngioma 327
Chondrosarcoma 263 Papillary craniopharyngioma 328
Osteoma 264 Granular cell tumour of the sellar region 329
Osteochondroma 264 Pituicytoma 332
Osteosarcoma 264 Spindle cell oncocytoma 334

12 Melanocytic tumours 265 18 Metastatic tumours 337


Meningeal melanocytosis 267
Meningeal melanomatosis 267 Contributors 342
Meningeal melanocytoma 268 Declaration of interest statements 349
Meningeal melanoma 269 IARC/WHO Committee for ICD-0 350
Sources of figures and tables 351
13 Lymphomas 271 References 356
Diffuse large B-cell lymphoma of the CNS 272 Subject index 402
Corticoid-mitigated lymphoma 275 List of abbreviations 408
Sentinel lesions 275
Immunodeficiency-associated CNS lymphomas 275
AIDS-related diffuse large B-cell lymphoma 275
EBV+ diffuse large B-cell lymphoma, NOS 276
Lymphomatoid granulomatosis 276
,

WHO classification of tumours of the central nervous system


Diffuse astrocytic and oligodendroglial tumours Neuronal and mixed neuronal-glial tumours
Diffuse astrocytoma, IDH-mutant 9400/3 Dysembryoplastic neuroepithelial tumour 9413/0
Gemistocytic astrocytoma, IDH-mutant 9411/3 Gangliocytoma 9492/0
Diffuse astrocytoma, IDH-wildtype 9400/3 Ganglioglioma 9505/1
Diffuse astrocytoma, NOS 9400/3 Anaplastic ganglioglioma 9505/3
Dysplastic cerebellar gangliocytoma
Anaplastic astrocytoma, IDH-mutant 9401/3 (Lhermitte-Duclos disease) 9493/0
Anaplastic astrocytoma, IOH-wildtype 9401/3 Desmoplastic infantile astrocytoma and
Anaplastic astrocytoma, NOS 9401/3 ganglioglioma 9412/1
Papillary glioneuronal tumour 9509/1
Glioblastoma, I DH-wildtype 9440/3 Rosette-forming glioneuronal tumour 9509/1
Giant cell glioblastoma 9441/3 Diffuse leptomeningeal glioneuronal tumour
Gliosarcoma 9442/3 Central neurocytoma 9506/1
Epithelioid glioblastoma 9440/3 Extraventricular neurocytoma 9506/1
Glioblastoma, IDH-mutant 9445/3* Cerebellar liponeurocytoma 9506/1
Glioblastoma, NOS 9440/3 Paraganglioma 8693/1

Diffuse midline glioma, H3 K27M-mutant 9385/3* Tumours of the pineal region


Pineocytoma 9361/1
Oligodendroglioma, IDH-mutant and Pineal parenchymal tumour of intermediate
1 p/19q-codeleted 9450/3 differentiation 9362/3
Oligodendroglioma, NOS 9450/3 Pineoblastoma 9362/3
Papillary tumour of the pineal region 9395/3
Anaplastic oligodendroglioma, IDH-mutant
and 1 p/19q-codeleted 9451/3 Embryonal tumours
Anaplastic oligodendroglioma, NOS 9451/3 Medulloblastomas, genetically defined
Medulloblastoma, WNT-activated 9475/3*
0/igoastrocytoma, NOS 9382/3 Medulloblastoma, SHH-activated and
Anaplastic oligoastrocytoma, NOS 9382/3 TP53-mutant 9476/3*
Medulloblastoma, SHH-activated and
Other astrocytic tumours TP53-wildtype 9471/3
Pilocytic astrocytoma 9421/1 Medulloblastoma, non-WNT/non-SHH 9477/3*
Pilomyxoid astrocytoma 9425/3 Medulloblastoma, group 3
Subependymal giant cell astrocytoma 9384/1 Medulloblastoma, group 4
Pleomorphic xanthoastrocytoma 9424/3 Medulloblastomas, histologically defined
Anaplastic pleomorphic xanthoastrocytoma 9424/3 Medulloblastoma, classic 9470/3
Medulloblastoma, desmoplastic/nodular 9471/3
Ependymal tumours Medulloblastoma with extensive nodularity 9471/3
Su bependymoma 9383/1 Medulloblastoma, large cell I anaplastic 9474/3
Myxopapillary ependymoma 9394/1 Medulloblastoma, NOS 9470/3
Ependymoma 9391/3
Papillary ependymoma 9393/3 Embryonal tumour with multilayered rosettes,
Clear cell ependymoma 9391/3 C 19MC-altered 9478/3*
Tanycytic ependymoma 9391/3 Embryonal tumour with multilayered
Ependymoma, RELA fusion-positive 9396/3* rosettes, NOS 9478/3
Anaplastic ependymoma 9392/3 Medulloepithelioma 9501/3
CNS neuroblastoma 9500/3
Other gliomas CNS ganglioneuroblastoma 9490/3
Chordoid glioma of the third ventricle 9444/1 CNS embryonal tumour, NOS 9473/3
Angiocentric glioma 9431/1 Atypical teratoid/rhabdoid tumour 9508/3
Astroblastoma 9430/3 CNS embryonal tumour with rhabdoid features 9508/3

Choroid plexus tumours Tumours of the cranial and paraspinal nerves


Choroid plexus papilloma 9390/0 Schwannoma 9560/0
Atypical choroid plexus papilloma 9390/1 Cellular schwannoma 9560/0
Choroid plexus carcinoma 9390/3 Plexiform schwannoma 9560/0

10 WHO classification of tumours of the central nervous system


Melanotic schwannoma 9560/1 Osteochondroma 9210/0
Neurofibroma 9540/0 Osteosarcoma 9180/3
Atypical neurofibroma 9540/0
Plexiform neurofibroma 9550/0 Melanocytic tumours
Perineurioma 9571/0 Meningeal melanocytosis 8728/0
Hybrid nerve sheath tumours Meningeal melanocytoma 8728/1
Malignant peripheral nerve sheath tumour 9540/3 Meningeal melanoma 8720/3
Epithelioid MPNST 9540/3 Meningeal melanomatosis 8728/3
MPNST with perineurial differentiation 9540/3
Lymphomas
Meningiomas Diffuse large B-cell lymphoma of the CNS 9680/3
Meningioma 9530/0 Immunodeficiency-associated CNS lymphomas
Meningothelial meningioma 9531/0 AIDS-related diffuse large B-cell lymphoma
Fibrous meningioma 9532/0 EBV-positive diffuse large B-cell lymphoma, NOS
Transitional meningioma 9537/0 Lymphomatoid granulomatosis 9766/1
Psammomatous meningioma 9533/0 lntravascular large B-cell lymphoma 9712/3
Angiomatous meningioma 9534/0 Low-grade B-cell lymphomas of the CNS
Microcystic meningioma 9530/0 T-cell and NK/T-cell lymphomas of the CNS
Secretory meningioma 9530/0 Anaplastic large cell lymphoma, ALK-positive 9714/3
Lymphoplasmacyte-rich meningioma 9530/0 Anaplastic large cell lymphoma, ALK-negative 9702/3
Metaplastic meningioma 9530/0 MALT lymphoma of the dura 9699/3
Chordoid meningioma 9538/1
Clear cell meningioma 9538/1 Histiocytic tumours
Atypical meningioma 9539/1 Langerhans cell histiocytosis 9751/3
Papillary meningioma 9538/3 Erdheim-Chester disease 9750/1
Rhabdoid meningioma 9538/3 Rosai-Dorfman disease
Anaplastic (malignant) meningioma 9530/3 Juvenile xanthogranuloma
Histiocytic sarcoma 9755/3
Mesenchymal, non-meningothelial tumours
Solitary fibrous tumour I haemangiopericytoma** Germ cell tumours
Grade 1 8815/0 Germinoma 9064/3
Grade 2 8815/1 Embryonal carcinoma 9070/3
Grade 3 8815/3 Yolk sac tumour 9071/3
Haemangioblastoma 9161/1 Choriocarcinoma 9100/3
Haemangioma 9120/0 Teratoma 9080/1
Epithelioid haemangioendothelioma 9133/3 Mature teratoma 9080/0
Angiosarcoma 9120/3 Immature teratoma 9080/3
Kaposi sarcoma 9140/3 Teratoma with malignant transformation 9084/3
Ewing sarcoma I PNET 9364/3 Mixed germ cell tumour 9085/3
Lipoma 8850/0
Angiolipoma 8861/0 Tumours of the sellar region
Hibernoma 8880/0 Craniopharyngioma 9350/1
Liposarcoma 8850/3 Adamantinomatous craniopharyngioma 9351/1
Desmoid-type fibromatosis 8821/1 Papillary craniopharyngioma 9352/1
Myofibroblastoma 8825/0 Granular cell tumour of the sellar region 9582/0
Inflammatory myofibroblastic tumour 8825/1 Pituicytoma 9432/1
Benign fibrous histiocytoma 8830/0 Spindle cell oncocytoma 8290/0
Fibrosarcoma 8810/3
Undifferentiated pleomorphic sarcoma I Metastatic tumours
malignant fibrous histiocytoma 8802/3
Leiomyoma 8890/0 The morphology codes are from the International Classification of Diseases
for Oncology (ICD-0) (742AJ. Behaviour is coded /0 for benign tumours;
Leiomyosarcoma 8890/3 /1 for unspecified, borderline, or uncertain behaviour; /2 for carcinoma in
Rhabdomyoma 8900/0 situ and grade Ill intraepithelial neoplasia; and /3 for malignant tumours.
Rhabdomyosarcoma 8900/3 The classification is modified from the previous WHO classification, taking
into account changes in our understanding of these lesions.
Chondroma 9220/0
*These new codes were approved by the IARC/WHO Committee for ICD-0.
Chondrosarcoma 9220/3 Italics: Provisional tumour entities. '*Grading according to the 2013
Osteoma 9180/0 WHO Classification of Tumours of Soft Tissue and Bone.

WHO classification of tumours of the central nervous system 11


WHO classification and grading of Louis O.N

tumours of the central nervous system

Combined histological-molecular molecular genetic alterations suggest a genetic finding is present. leaving that
classification that such challenges will be readily over- to individual practitioners and institutions,
For nearly a century, the classification of come in the near future {2105). Many of but the commentary sections do clarify
brain tumours has been based on con- the genetic parameters included in the the implication of certain genetic features;
cepts of histogenesis, hinging on the idea 2016 WHO classification can be as- for example, in what situations IOH status
that tumours can be classified according sessed using immunohistochemistry can be designated as wildtype.
to their microscopic similarities with puta- or FISH, but it is recognized that some
tive cells of origin and their developmental centres may not have the ability to carry Histological grading
differentiation states. These histological out molecular analyses and that some Histological grading is a means of pre-
similarities have been characterized pri- molecular results may not be conclusive. dicting the biological behaviour of a
marily on the basis of the light microscop- With this in mind, an NOS diagnostic des- neoplasm. In the clinical setting, tumour
ic appearance of H&E-stained sections, ignation has been included in the 2016 grade is a key factor influencing the
the immunohistochemical expression of WHO classification wherever such issues choice of therapies. Since its first pub-
proteins, and the electron microscopic may apply. The NOS designation indi- lication in 1979, the WHO classification
assessment of ultrastructural features. cates that there is insufficient informa- of tumours of the central nervous system
The 2000 and 2007 WHO classifications tion to assign a more specific code. In has included a grading scheme that es-
considered histological features along this context. the NOS category includes sentially constitutes a malignancy scale
with the rapidly increasing knowledge both tumours that have not been tested (ranging across a wide variety of neo-
of the genetic changes that underlie the for the genetic parameter(s) and tumours plasms) rather than a strict histological
tumorigenesis of CNS tumours. Many that have been tested but did not show grading system (1290,1291,2878} WHO
of the canonical genetic alterations had the diagnostic genetic alterations. In grading is widely used, and it has incor-
been identified by the time the 2007 other words, the NOS designation does porated or largely replaced other previ-
WHO classification was published, but at not refer to a specific entity; instead, it ously published grading systems for
the time the consensus opinion was that designates the lesions that cannot be brain tumours. Although grading is not
such changes could not yet be used to classified into any of the more precisely a requirement for the application of the
define neoplasms; instead, genetic sta- defined groups. WHO classification for some tumours,
tus served as supplementary information including gliomas and meningiomas,
within the framework of diagnostic cat- Definitions, disease summaries, and numerical WHO grades are useful addi-
egories established by standard, histolo- commentaries tions to the diagnoses. The WHO Work-
gy-based means. In contrast, the present Each entity-specific section in the 2016 ing Group responsible for this update of
update (the 2016 classification) breaks WHO classification begins with a short the 4th edition has expanded the classifi-
with this nearly century-old tradition and disease definition that describes the es- cation to include additional entities; how-
incorporates well-established molecular sential diagnostic criteria. This initial ever, since the number of cases of some
parameters into the classification of dif- definition is followed by a description of of these newly defined entities is limited,
fuse gliomas. characteristic associated findings; for the assignment of grades to such entities
Changing the classification to include example, although a delicate branching is still provisional, pending publication of
diagnostic categories that depend on vasculature and calcospherites are not additional data and long-term follow-up.
genotype may create certain challenges essential for the diagnosis of oligoden-
with respect to testing and reporting. droglioma, they are highly characteristic. Histological grading across tumour
These challenges include the availability The diagnostic criteria and characteris- entities
and choice of genotyping and surrogate tic features are then followed by the rest Grade I lesions are generally tumours
genotyping assays, the approaches that of the disease summary, in which other with low proliferative potential and the
may need to be taken by centres without notable clinical, pathological. and mo- possibility of cure after surgical resec-
genotyping (or surrogate genotyping) ca- lecular findings are described. Finally, tion alone. Grade II lesions are usually
pabilities, and the actual formats used to for some entities, there is also additional infiltrative in nature and often recur, de-
report these integrated diagnoses {1535\ commentary that provides information on spite having low levels of proliferative
However, an important consideration for classification, clarifying the nature of the activity. Some grade 11 entities tend to
the 2016 WHO classification was that the genetic parameters to be evaluated and progress to higher grades of malignancy;
implementation of combined phenotyp- providing genotyping information on pos- for example, grade II diffuse astrocytoma
ic-genotypic diagnostics in some large sibly overlapping histological entities. The tends to transform to grade Ill anaplas-
centres and the increasing availability classification does not specify the type tic astrocytoma and glioblastoma. The
of immunohistochemical surrogates for of testing required to establish whether grade Ill designation is applied to lesions

12 WHO classification and grading


with clear histological evidence of ma- which is the more common practice in implications. For each tumour entity, the
lignancy, including nuclear atypia and non-nervous system tumours. Specifi- combination of these parameters contrib-
sometimes brisk mitotic activity. In most cally, this principle is applied to solitary fi- utes to an overall estimate of prognosis.
settings, patients with grade Ill tumours brous tumour I haemangiopericytoma, for Patients with WHO grade 11 tumours typi-
receive radiation and/or chemotherapy. which three different grades can be as- cally survive for> 5 years, and those with
The grade IV designation is applied to signed within the same tumour designa- grade Ill tumours survive for 2-3 years.
cytologically malignant, mitotically ac- tion. It is anticipated that the greater flex- For patients with WHO grade IV tumours,
tive, necrosis-prone neoplasms that are ibility afforded by grading within tumour prognosis depends heavily on whether
often associated with rapid pre- and entities may be an advantage for future effective treatment regimens are avail-
postoperative disease evolution and WHO classifications of brain tumours. able. Most glioblastoma patients, and in
fatal outcome. Glioblastoma and most particular elderly patients, succumb to
embryonal neoplasms are examples of Tumour grade as a prognostic factor the disease within a year. For those with
grade IV lesions. Widespread infiltration WHO grade is based on histological other grade IV neoplasms, the outlook
of surrounding tissue and a propensity features and is one component of a set may be considerably better. For exam-
for craniospinal dissemination character- of criteria used to predict response to ple, grade IV cerebellar medulloblasto-
ize many grade IV neoplasms, although therapy and outcome 11535). Other cri- mas and germ cell tumours such as ger-
these features are not essential. teria include clinical findings (e.g. patient minomas are rapidly fatal if left untreated,
age, performance status, and tumour but state-of-the-art radiation and chemo-
Histological grading within tumour location), radiological features (e.g. con- therapy result in 5-year survival rates ex-
entities trast enhancement), extent of surgical ceeding 60% and 80%, respectively. For
To date, the WHO classifications of CNS resection, proliferation index values, some tumour types, the presence of cer-
tumours have assigned grades as de- and genetic alterations. Genetic profile tain genetic alterations can shift prognos-
scribed above (i.e. across tumour enti- has become increasingly important be- tic estimates markedly within the same
ties). For the first time, the 2016 WHO cause some genetic changes (e.g. IDH grade, in some cases outweighing the
classification has also adopted the prin- mutation in diffuse gliomas) have been prognostic strength of grade itself 1952,
ciple of grading within a tumour entity, found to have important prognostic 1535,2105)

WHO grades of select CNS tumours Desmoplastic infantile astrocytoma and ganglioglioma I
Papillary glioneuronal tumour I
Diffuse astrocytic and oligodendroglial tumours Rosette-forming glioneuronal tumour I
Diffuse astrocytoma, I DH-mutant II Central neurocytoma 11
Anaplastic astrocytoma, I DH-mutant Ill Extraventricular neurocytoma II
Glioblastoma, IDH-wildtype IV Cerebellar liponeurocytoma II
Glioblastoma, I DH-mutant IV
IV
Tumours of the pineal region
Diffuse mid line glioma, H3 K27M-mutant
Pineocytoma I
Oligodendroglioma, I DH-mutant and 1 p/19q-codeleted II
Pineal parenchymal tumour of intermediate differentiation II or Ill
Anaplastic oligodendroglioma, IDH-mutant and
Pineoblastoma IV
1 p/19q-codeleted Ill
Papillary tumour of the pineal region II or Ill
Other astrocytic tumours
Pilocytic astrocytoma I
Embryonal tumours
Medulloblastoma (all subtypes) IV
Subependymal giant cell astrocytoma I
Embryonal tumour with multilayered rosettes, C19MC-altered IV
Pleomorphic xanthoastrocytoma II
Medulloepithelioma IV
Anaplastic pleomorphic xanthoastrocytoma Ill
CNS embryonal tumour, NOS IV
Ependymal tumours Atypical teratoid/rhabdoid tumour IV
Subependymoma I CNS embryonal tumour with rhabdoid features IV
Myxopapillary ependymoma I
Ependymoma II Tumours of the cranial and paraspinal nerves
Schwannoma I
Ependymoma, RELA fusion-positive II or Ill
Neurofibroma I
Anaplastic ependymoma Ill
Perineurioma I
Other gliomas Malignant peripheral nerve sheath tumour (MPNST) 11, Ill or IV
Angiocentric glioma I
Chordoid glioma of third ventricle II Meningiomas
Meningioma I
Choroid plexus tumours Atypical meningioma II
Choroid plexus papilloma I Anaplastic (malignant) meningioma Ill
Atypical choroid plexus papilloma II
Choroid plexus carcinoma Ill
Mesenchymal, non-meningothelial tumours
Solitary fibrous tumour I haemangiopericytoma I, II or Ill
Neuronal and mixed neuronal-glial tumours Haemangioblastoma I
Dysembryoplastic neuroepithelial tumour I
Gangliocytoma I
Tumours of the sellar region
Craniopharyngioma
Ganglioglioma I
Granular cell tumour
Anaplastic ganglioglioma Ill
Pituicytoma
Dysplastic gangliocytoma of cerebellum (Lhermitte-Duclos) I
Spindle cell oncocytoma

WHO classification and grading of tumours of the central nervous system 13


CHAPTER 1

Diffuse astrocytic and oligodendroglial tumours


Diffuse astrocytoma, IDH-mutant
Anaplastic astrocytoma, I DH-mutant
Glioblastoma, IDH-wildtype
Glioblastoma, IDH-mutant
Diffuse midline glioma, H3 K27M-mutant
Oligodendroglioma, I DH-mutant and 1 p/19q-codeleted
Anaplastic oligodendroglioma, I DH-mutant and 1 p/19q-codeleted
Diffuse astrocytic and Louis D.N.
van Deimling A.
oligodendroglial tumours - Introduction Cavenee W.K.

This 2016 update of the 2007 WHO clas- Histology Astrocytoma Oligoastrocytoma Oligodendroglioma Glloblastoma
sification incorporates well-established
molecular parameters into the classifica- i------�-------1-' ------�
IDH,tatus
tion of diffuse gliomas, and this nosologi-

!
IDHmutant IDHwlld-type I /DHmutant /DHwild-type
cal shift has impacted the classification
in several ways. Most notably, whereas !
l
lp/l9qond ATRXI� Gliob:stoma,/DHmutant
all astrocytic tumours were previously other genetic 1p/19q codeletion I
I
l ;
TPS3 mutation"
parameters I Glioblastoma, IDH wild-type
grouped together, now all diffuse gliomas
(whether astrocytic or not) are grouped i
together, on the basis of not only their I Diffuse astrocytoma, IDH mutant
,.--�������������
growth pattern and behaviours, but more
.
Oligodendroglioma, IDH mutant and lp/19q codeleted Genetic testing not done

pointedly of their shared IDH1 and IDH2 or inconclusive

genetic status. From a pathogenetic After exclusion of other entities:


I
I
I
point of view, this provides a dynamic Diffuse astrocytoma, /DH wild-type I
Oli odendro ioma, NOS I
classification based on both phenotype I
I

and genotype; from a prognostic point of t


Diffuse astrocytoma, NOS
view, it groups tumours that share similar Oligodendroglioma, NOS
prognostic markers; and from the even- • =characteristic butnot
Oligoastrocytoma, NOS
Glloblastoma, NOS
tual therapeutic point of view, it will pre- required for diagnosis

sumably guide the treatment of biologi- Fig. 1.01 Diffuse gliomas: from histology, !DH status, and other genetic parameters to WHO diagnosis.
cally similar entities.
In this new classification, the diffuse glio- a reasonably narrowly defined group placed into categories biologically com-
ma category includes the WHO grade II of tumours characterized by K27M mu- patible with either astrocytoma or oligo-
and Ill astrocytic tumours, the grade II tations in histone H3 genes, a diffuse dendroglioma 12220,2751}. with only rare
and Ill oligodendrogliomas, the rare growth pattern, and midline location: the cases having histological and molecular
grade II and Ill oligoastrocytomas, the newly defined H3 K27M-mutant diffuse features of both { 1067,2754) As a result,
grade IV glioblastomas, and the related midline glioma. the more common diagnoses of astrocy-
diffuse gliomas (e.g. those of childhood) The use of integrated phenotypic and toma and oligodendroglioma both be-
This approach more sharply separates genotypic parameters in the 2016 WHO come more homoqeneously defined, as
astrocytomas that have a more circum- classification provides an increased does the very uncommon diagnosis of
scribed growth pattern, lack IDH gene level of objectivity. This will presumably oligoastrocytoma.
alterations, and sometimes have BRAF yield more homogeneous and narrowly The above example of classifying as-
mutations (i.e. pilocytic astrocytoma, defined diagnostic entities than in prior trocytomas, oligodendrogliomas, and
pleomorphic xanthoastrocytoma. and classifications, which in turn should lead oligoastrocytomas raises the question of
subependymal giant cell astrocytoma) to better correlations with prognosis and whether classification can proceed on
from diffuse gliomas. In other words, dif- treatment response It will also result in the basis of genotype alone, i.e. without
fuse astrocytoma and oligodendroglioma potentially larger groups of tumours that histology. At this point in time, it is not
are now nosologically more similar than do not fit into any of the more narrowly possible; in the classification process, a
are diffuse astrocytoma and pilocytic defined entities - groups that themselves diagnosis of diffuse glioma (rather than
astrocytoma; the family trees have been will be more homogeneous and therefore some other tumour type) must first be
redrawn. more amenable to subsequent study and established histologically in order for the
Similarly, paediatric diffuse gliomas were improved classification. nosological and clinical significance of
previously grouped together with their One compelling example of this im- specific genetic changes to be under-
overall (generally adult) counterparts, de- proved objectivity relates to the diagno- stood in the appropriate context. Another
spite known differences in behaviour be- sis of oligoastrocytoma - a difficult-to- reason phenotype remains essential is
tween paediatric and adult gliomas with define category of tumours that has been that some individual tumours do not meet
similar histological appearances. More subject to high interobserver variability in the more narrowly defined genotypic cri-
recent information on the distinct under- diagnosis, with some centres diagnosing teria; for example. a phenotypically clas-
lying genetic abnormalities in paediatric these lesions frequently and others only sic diffuse astrocytoma that lacks the
diffuse gliomas has since prompted the rarely. The use of both genotype and signature genetic characteristics of IDH1/
separation of some of these entities from phenotype in diagnosing these tumours IDH2 and ATRX mutation. However, it is
histologically similar adult tumours - with results in essentially all of them being possible that future WHO classifications

16 Diffuse gliomas
I
of diffuse gliomas, in the setting of deep-
A Mutations B Frequency of Mutations
er and broader genomic capabilities, will J/J 7/7
100 3</36
require less histological evaluation - per- 27/30
'3/51 11/ll
• IDH2 mutated
• IDHl mutated
haps only an initial diagnosis of diffuse 80 3&/52
Rl72G GGG N•2
glioma. For the time being, the current Rl72M ATG N•l
R172K AAG N•4
WHO classification is predicated on the t
concept of combined phenotypic and IDH2
An : GGC AGG CAC GCC

genotypic classification, and on the gen- 1110 :Gm Rm Hin Au•

eration of so-called integrated diagnoses 1uo :Gm Rm ..... 20 1/7

I
Hill
6/123
o,, ••

IOHl
ATA: GGT CGT CAT GCT 0,21 0,10 • 0/15 0/55
(1535)
It is important to acknowledge that Rll2H CAT N•l42
RlllC TGT N•7
changing the classification to include Rl32L en N•7
Rll2S AGT
diagnostic categories that depend on Rl32G GGT
N•4
N•I
genotype may create certain challenges
with respect to testing and reporting,
which have been discussed elsewhere
(1535). These challenges include the Fig. 1.02 IDH1 and IDH2 mutations in human gliomas, histologically diagnosed according to the 2007 WHO classi-
availability and choice of genotyping and fication. Reprinted from Yan H et al. {2810).
surrogate genotyping assays, the ap-
proaches that may need to be taken by prognostic information in addition to that or morphology. The finding of a solitary
centres without genotyping (or surrogate provided by histological grade. Never- mitosis in an ample specimen is not suf-
genotyping) capabilities, and the actual theless, in the 2016 WHO classification, ficient proof of WHO grade Ill behaviour,
formats used to report these integrated diffuse astrocytomas are graded using but the separation of grade II tumours
diagnoses (1535). However, the imple- a three-tiered system similar to the Ring- from grade Ill tumours may be facilitated
mentation of combined phenotypic-gen- ertz system (2130). the St. Anne-Mayo by determination of the Ki-67 proliferation
otypic diagnostics in some large centres system (533). and the previously pub- index {1056). Microvascular proliferation
(2105) and the increasing availability of lished WHO schemes (1293,1534). Ac- is defined as apparent multilayering of
immunohistochemical surrogates for mo- cording to the WHO's current histological endothelium (rather than simple hyper-
lecular genetic alterations suggest that definition, tumours with cytological atypia vascularity) or glomeruloid vasculature.
such challenges will be readily overcome alone (i.e. diffuse astrocytomas) are con- Necrosis may be of any type; perinecrotic
in the near future {251 OA). sidered grade 11, those that also show an- palisading need not be present. Simple
aplasia and mitotic activity (i.e. anaplas- apposition of cellular zones with interven-
Histological grading of diffuse tic astrocytomas) are considered WHO ing pallor suggestive of incipient necrosis
astrocytic tumours grade 111, and tumours that additionally is insufficient. The aforementioned crite-
In neuro-oncology, histological grading show microvascular proliferation and/or ria make their appearance in a predict-
has been most systematically evaluated necrosis are grade IV. Atypia is defined able sequence: atypia is followed in turn
and successfully applied to diffusely in- as variation in nuclear shape or size with by mitotic activity, then increased cellu-
filtrative astrocytic tumours, although accompanying hyperchromasia. Mitoses larity, and finally microvascular prolifera-
recent studies suggest that molecu- must be unequivocal, but no special sig- tion and/or necrosis.
lar parameters may provide powerful nificance is accorded to their number

Diffuse astrocytic and oligodendroglial tumours - Introduction 17


Diffuse astrocytoma, IDH-mutant von Deimling A.
Huse JT
Berger M.S.
Weller M.
Yan H. Nakazato Y.
Brat D.J. Burger P.C.
Reifenberger G. Ellison D.W.
Ohgaki H. Louis D.N.
Kleihues P

Definition Epidemiology those for I DH-mutant anaplastic astrocy-


A diffusely infiltrating astrocytoma with a toma (2103).
mutation in either the IDH1 or IDH2 gene. Incidence
/DH-mutant diffuse astrocytoma typically Tumour registries have not distinguished Localization
features moderately pleomorphic cells diffuse astrocytomas on the basis of IDH /DH-mutant diffuse astrocytomas can
and is characterized by a high degree of mutation status. However, because most be located in any region of the CNS, but
cellular differentiation and slow growth. cases carry an IDH mutation, the avail- most commonly develop supratentorially
The diagnosis is supported by the pres- able data reflect this genetically defined in the frontal lobes (24281 This is similar
ence of ATRX and TP53 mutation. The tumour entity to some extent. Diffuse to the preferential localization of IDH-
presence of a component morphologi- astrocytomas account for approximately mutant and 1p/19q-codeleted oligoden-
cally resembling oligodendroglioma is 11-15% of all astrocytic brain tumours droglioma (1418,28711 and supports the
compatible with this diagnosis in the ab- (1862,1863}. Annual incidence rates have hypothesis that these gliomas develop
sence of 1 p/19q codeletion. This tumour been estimated at 0.55 and 0.75 new from a distinct population of precursor
most commonly affects young adults and cases per 100 000 population (1862, cells (1830}.
occurs throughout the CNS, but is prefer- 1863,2797).
entially located in the frontal lobes (2428}. Some reports suggest that the incidence Clinical features
Diffuse astrocytomas have an intrinsic of astrocytomas in children has slightly Seizures are a common presenting symp-
capacity for malignant progression to increased in recent decades in several tom; however, subtle abnormalities such
/DH-mutant anaplastic astrocytoma and Scandinavian countries and North Amer- as speech difficulties, changes in sen-
eventually to I DH-mutant glioblastoma. ica (989,1015,2471). There is a male pre- sation or vision, and some form of motor
dominance, with a male-to-female ratio of change may be present earlier. Symptom
ICD-0 code 9400/3 1.3:1 (1863}. onset is rarely abrupt, and some tumours
are diagnosed incidentally (2390,2511 I.
Grading Age distnbution With frontal lobe tumours, changes in
Diffuse astrocytoma corresponds histo- The median and mean ages of patients behaviour or personality may be the pre-
logically to WHO grade II. with /DH-mutant diffuse astrocytoma are senting feature. Such changes may be
in the mid-30s. According to one study present for months before diagnosis.
Synonyms of adult patients only, the median age of
Low-grade astrocytoma (discouraged); patients with /DH-mutant diffuse astro- Imaging
fibrillary astrocytoma (no longer cytoma is 36 years, and the mean age Like clinical features, the results of neu-
recommended) is 38 years. These values are similar to roimaging studies can be extremely vari-

-
able. On CT, diffuse astrocytomas most
100 often present as poorly defined, homo-
90
� geneous masses of low density, without
80
� ........ contrast enhancement. However, calcifi-
cation and cystic change may be present
� ff
VI

....0u"'
70
). , early in the evolution of the tumour. MRI
60 studies usually show T1-hypodensity and
* QJ so /I T2-hyperintensity, with enlargement of
>
+'
.!'! 40 // the areas involved early in the evolution
:,
E 30
:,
XI of the tumour. Gadolinium enhancement
is not common in low-grade diffuse as-
u
20 /I trocytomas, but tends to appear during
// tumour progression.
10

0
v
0 10 20 30 40 so 60 70 80 90 100 Macroscopy
Age at diagnosis --All --Alli
Because of their infiltrative nature, these
tumours usually show blurring of the gross
Fig. 1.03 Cumulative age distribution of astrocytomas at diagnosis, both sexes. Based on 613 cases of I DH-mutant diffuse
astrocytoma (All) and 674 cases of IDH-mutant anaplastic astrocytoma (Alli). Note that in this aggregate of patients,
anatomical boundaries. There is enlarge-
despite the difference in grade, the age distribution is similar, with a mean age of 35 years for All and 37 years for Alli. This ment and distortion (but not destruction)
corresponds to similar data published by Reuss DE et al. (2103), showing that I DH-mutant All and Alli astrocytomas differ of the invaded anatomical structures (e.q.
little with respect to age at diagnosis and survival in adult patients. Includes data from Reuss DE et al. (2103). the cortex and the compact myelinated

18 Diffuse gliomas
pathways). Local mass lesions may
be present in either grey or white mat-
ter, but they have indistinct boundaries,
and changes such as smaller or larger
cysts, granular areas, and zones of firm-
ness or softening may be seen. Cystic
change most commonly presents as a
focal spongy area, with multiple cysts of
various sizes. Extensive microcyst forma-
tion may cause a gelatinous appearance. A B
Occasionally, a single large cyst filled Fig. 1.04 A Large diffuse astrocytoma occupying the left temporal lobe, with extension to the Sylvian fissure. Note the
with clear fluid is present. Tumours with homogeneous surface and the enlargement of local anatomical structures. B Large diffuse astrocytoma originating
prominent gemistocytes sometimes have from the pericallosal cortex of the right hemisphere. The tumour extends into the interhemispheric fissure and shifts the
single, large, smooth-walled cysts. Focal mid line towards the left hemisphere. Macroscopically, this lesion is well delineated and still shows structures resembling
calcification may also be present, and a cortical architecture.
more diffuse grittiness may be observed.
Extension into contralateral structures, filaments. The pattern may vary markedly for R132H-mutant IDH1 (sometimes in
particularly in the frontal lobes, is rarely in different regions of the neoplasm. His- combination with p53 immunohistochem-
observed. tological recognition of neoplastic astro- istry and rarely with assessment for tri-
cytes using H&E staining on sectioned somy 7) is a powerful means to distinguish
Microscopy material depends mainly on nuclear neoplastic from reactive astrocytes (3471.
Diffuse astrocytoma is composed of characteristics. The normal astrocytic nu- In other situations, however, the differen-
well-differentiated fibrillary astrocytes in cleus is oval to elongated, but on section- tial diagnosis can be challenging and may
a background of a loosely structured, of- ing, occasional round cross-sections are rely on standard histological differences.
ten microcystic tumour matrix. Fibrillary seen. The nucleus is typically vesicular, Diffuse astrocytoma contains astrocytes
astrocytoma is the classic type of diffuse with intermediate-sized masses of chro- that are increased in number and usually
astrocytoma and is no longer listed as a matin and often with a distinct nucleolus. in size. but are otherwise difficult to distin-
variant (1533}. Normal human astrocytes show no H&E- guish on an individual basis from normal
The cellularity is moderately increased stainable cytoplasm that is distinct from or reactive cells. In minor degrees of ana-
compared with that of normal brain, and the background neuropil. Reactive as- plasia, it is the number of astrocytes and
nuclear atypia is a characteristic feature. trocytes are defined by enlarged nuclei (most commonly) the uniformity of their
Mitotic activity is generally absent, but a and the presence of stainable, defined morphology that is most helpful in recog-
single mitosis does not justify the diagno- cytoplasm, culminating in the gemisto- nizing their neoplastic nature. Reactive
sis of anaplastic astrocytoma unless ob- cyte, which has a mass of eosinophilic astrocytes are rarely all in the same stage
served in a small biopsy or in the setting cytoplasm, often an eccentric nucleus, of reactivity at the same time, so reac-
of obvious nuclear anaplasia. The pres- and cytoplasm that extends into fine tions reveal mixtures of astrocytes; some
ence of necrosis or microvascular prolif- processes. with enlarged nuclei, others with varying
eration is incompatible with the diagnosis amounts of cytoplasm. most often on a
of diffuse astrocytoma. Phenotypically, Differential diagnosis somewhat rarefied background. In diffuse
neoplastic astrocytes may vary consider- The major entity in the differential diagno- astrocytoma, almost all of the nuclei look
ably with respect to their size, the promi- sis is reactive astrocytosis. Because most identical, and the background is of at least
nence and disposition of cell processes, !OH-mutant diffuse astrocytomas have normal density or shows increased num-
and the abundance of cytoplasmic glial R132H mutations, immunohistochemistry bers of cellular processes. Microcystic

... .. . . . - ...
Fig. 1.05 Diffuse astrocytoma. A Moderately cellular tumour composed of uniform neoplastic fibrillary astrocytic cells. B Extensive microcyst formation.

Diffuse astrocytoma, I DH-mutant 19


reveals identical TP53 mutations in both
gemistocytes and non-gemistocytic tu-
mour cells (2094). Although it has been
reported that the gemistocytic variant
may be particularly prone to progression
to anaplastic astrocytoma and glioblas-
toma (1377,2204,2273). this does not jus-
tify a general classification as anaplastic
astrocytoma (320,2273). nor is this im-
pression based on current molecular
Fig. 1.06 Diffuse astrocytoma. Low cellularity and characterization, in particular knowledge
nuclear atypia. of IDH mutation status.

change may be present, but most cells /mmunophenotype


look like one another, without the admix- Diffuse astrocytomas reliably express distinction of true neoplasia from reac-
ture of gemistocytes more often seen in GFAP, although to various degrees and tive gliosis (347,359). Strong nuclear p53
reactions to injury. Pre-existing cell types not in all tumour cells. In particular, small expression is also frequently observed,
(e.g. neurons) are often entrapped. round cells with scanty cytoplasm and consistent with the high incidence of
processes tend not to label avidly for TP53 mutations in diffuse astrocytoma
lntraoperative diagnosis GFAP. In these cases, immunopositiv- (915). However, the use of p53 immuno-
The smear/squash technique is often ity may be restricted to a small perinu- positivity to reflect TP53 mutation is not
used during stereotaxic biopsies and clear rim and to admixed neoplastic cell entirely sensitive or specific (1530,1771).
yields similar findings, although this processes in the fibrillary tumour back- In contrast, ATRX expression is almost
method is highly unreliable for estimating ground (1292) Vimentin is typically im- invariably lost in the setting of ATRX mu-
cellularity. Many histological features are munopositive as well, with a labelling pat- tations, which also feature prominently in
exaggerated and amplified (e.g. nuclear tern approximating that of GFAP (995). diffuse astrocytoma (see Genetic profile)
folds, abnormal chromatin pattern, and The signature molecular characteristics (361,1160,1215,2105). ATRX typically
astrocytic processes). The presence of of diffuse astrocytoma (see Genetic pro- demonstrates strong nuclear expression
many round to oval nuclei with smooth file) can often be demonstrated immuno- in normal, unmutated tissue; therefore,
chromatin can indicate the presence of histochemically. For example, expression retention of immunolabelling in non-
an apparent oligodendroglial component of R132H-mutant IDH1 (the IDH1 R132H neoplastic vasculature and admixed
or (if the nuclei are less prominent) back- mutation accounts for about 90% of all neuronal, glial, and microglial elements
ground white matter. Histologically, there glioma-associated IDH mutations) can serves as a necessary internal control for
may be significant variation between tu- be detected using a mutation-specific the accurate interpretation of a negative
mours and within the same lesion. antibody (360) In mutant tumours, all ATRX immunostaining pattern. Finally,
neoplastic cells typically exhibit some consistent with its inapparent mitotic ac-
Growth fraction degree of cytoplasmic (stronger) and tivity, diffuse astrocytoma nearly always
The growth fraction as determined by the nuclear (weaker) labelling, provided the has a Ki-67 proliferation index of < 4%
Ki-67 proliferation index is usually < 4%. staining preparation used is technically (492,1137,1223,2059).
The gemistocytic neoplastic astrocytes adequate (359). For this reason, R132H-
show a significantly lower rate of prolif- mutant IDH1 immunohistochemistry has Cell of origin
eration than does the intermingled small- become an invaluable diagnostic ad- The available evidence suggests that
cell component (1046,1372,1377,1847, junct, not only in the molecular stratifi- IDH-mutant and 1p/19q-codeleted oli-
2706,2812). However, microdissection cation of diffuse glioma, but also in the godendrogliomas, IDH-mutant diffuse

- .
/

B The Ki-67 proliferation index is low.

20 Diffuse gliomas
astrocytomas, IOH-mutant anaplastic protein, and its deficiency has been as- which also predisposes patients to chon-
astrocytomas, and IOH-mutant glioblas- sociated with epigenomic dysregulation drosarcoma (734,1027).
tomas develop from a distinct population and telomere dysfunction (473) In par-
of precursor cells that differ from the pre- ticular, ATRX mutations seem to induce Prognosis and predictive factors
cursor cells of IDH-wildtype glioblastoma an abnormal telomere maintenance
(870,18301. mechanism known as alternative length- Clinical prognostic factors
ening of telomeres (977). ATRXmutations In the pre-lOH era, the median survival
Genetic profile and alternative lengthening of telomeres time was reported to be in the range of
Diffuse gliomas of WHO grades II and are mutually exclusive with activating mu- 6-8 years, with marked individual vari-
Ill, including diffuse astrocytoma, are tations in the TERT gene, which encodes ation. The total length of disease is in-
nearly all characterized by mutations in the catalytic component of telomerase. fluenced mainly by the dynamics of
IDH genes either IDH1 or /OH2 /118,953, Interestingly, TERT mutations are found malignant progression, which had been
1895,2810). Diffuse gliomas that occur in in the vast majority of oligodendroglio- reported to occur after a median time
adults and that do not harbour IDH muta- mas and most IDH-wildtype glioblasto- of 4-5 years {254,1826,18341. The Eu-
tion, regardless of their WHO grade, tend mas (349,622,1270). Distinct telomere ropean Organisation for Research and
to exhibit more aggressive clinical behav- maintenance mechanisms, mediated by Treatment of Cancer (EORTC) trials
iour (870,22381. either activated telomerase or alternative 22844 and 22845 showed that patient
Glioma-associated IOH1 and /OH2muta- lengthening of telomeres, seem to be re- age 2: 40 years, astrocytoma histology,
tions impart a gain-of-function phenotype quired for the pathogenesis of all diffuse largest tumour diameter 2: 6 cm, tumour
to the respective metabolic enzymes gliomas. crossing the midline. and neurological
IDH1 and IDH2, which overproduce ATRX deficiency has also been associ- deficits prior to surgery were associated
the oncometabolite 2-hydroxyglutarate ated with generalized genomic instability, with inferior outcome {1975) However,
{5231. The physiological consequences which can induce p53-dependent cell these prognostic estimates must be re-
of 2-hydroxyglutarate overproduction are death in some contexts {488). Therefore, evaluated in the context of IDH mutation
widespread. including profound effects TP53 mutations in diffuse astrocytoma status; one study that included 683 IOH-
on cellular epigenomic states and gene may enable tumour cell survival in the mutant diffuse astrocytoma cases from
regulation. Specifically, !OH mutations in- setting of ATRX loss. The genomic insta- three series showed a median survival of
duce G-CIMP, by which widespread hy- bility of I DH-mutant diffuse astrocytomas 10.9 years (2103)
permethylation in gene promoter regions is reflected in characteristic DNA copy
silences the expression of several impor- number abnormalities, which include Proliferation
tant cellular differentiation factors {1540, low-level amplification events involv- Low to absent proliferation rates as esti-
25891 In this way, IDH mutation and ing the oncogenes MYC and CCND2 in mated by mitotic count or the Ki-67 prolif-
G-CIMP are thought to maintain glioma mutually exclusive subsets (349). Copy eration index have traditionally been con-
cells of origin in stem cell-like physiologi- number events typically associated with sidered a diagnostic criterion for grading
cal states inherently more prone to self- IDH-wildtype glioblastoma. such as a diffuse astrocytoma as WHO grade II.
renewal and tumorigenesis. In particular, EGFR amplification and homozygous Among histologically diagnosed diffuse
it appears that IOH mutations promote COKN2A deletion, are rarely encoun- astrocytomas, the level of proliferation
glioma formation by disrupting chromo- tered, emphasizing the biological differ- has not been associated with outcome.
somal topology and allowing aberrant ences between !OH-mutant and IOH-
chromosomal regulatory interactions that wildtype astrocytomas {349,622,870) Histopathological factors
induce oncogene expression, including On the basis of expression profiling, Gemistocytic astrocytoma has been
glioma oncogenes such as POGFRA multiple diffuse astrocytoma subclasses associated with early progression and
(713AI Consistent with this concept, IDH have been designated, stratified by !DH inferior outcome {1834), but data on
mutations seem to be among the first ge- mutation status as well as neuroglial lin- larger contemporary patient cohorts with
netic alterations that occur in WHO grade eage markers {349,870). The transcrip- known IDH mutation status are lacking.
II diffuse glioma {2709). MGMT promoter tional profiles of diffuse astrocytomas Other histological factors associated with
methylation was reported in about 50% indicate distinct cells of origin in addition outcome have not yet been identified.
of diffuse astrocytomas in the pre-lOH to specific genomic features.
era, but this proportion may be higher Genetic alterations
among !DH-mutant diffuse astrocytomas Genetic susceptibility IDH1/2 mutations distinguish astrocyto-
and does not correlate consistently with Diffuse astrocytoma can occur in pa- mas with a more favourable course from
G-CIMP {1753,2589). tients with inherited TP53 germline mu- IDH-wildtype tumours, which have a less
The vast majority of IDH-mutant dif- tations I Li-Fraumeni syndrome (see favourable course {951). Among IDH-
fuse astrocytomas, as well as the WHO Li-Fraumeni syndrome, p. 310), although wildtype tumours, a genotype of 7q gain
grade 111 anaplastic astrocytomas and affected family members more frequently and 10q loss is associated with a par-
grade IV glioblastomas that evolve from develop anaplastic astrocytoma and glio- ticularly poor outcome (2731 I. However,
them, also harbour class-defining loss- blastoma. Lower-grade astrocytoma has as noted earlier (see Clinical prognostic
of-function mutations in TP53 and ATRX been diagnosed in patients with inherited factors), a study that included 683 IOH-
11160,1215,1834,2092,27041. ATRX en- Oiiier-type multiple enchondromatosis, mutant diffuse astrocytomas from three
codes an essential chromatin-binding series reported a median survival of

Diffuse astrocytoma, !DH-mutant 21


. . ... -- '• .
Fig. 1.09 Gemistocytic astrocytoma. A Tumour cells have abundant eosinophilic cytoplasm, with nuclei displaced to the periphery. B Perivascular lymphocytic infiltrates are common.

10.9 years. IDH mutations may be use- For a diagnosis of gemistocytic astro- Localization
ful as a predictive biomarker when IDH- cytoma, gemistocytes should constitute Gemistocytic astrocytomas can develop
targeted therapies such as small-mole- more than approximately 20% of all tu- in any region of the CNS, but they most
cule inhibitors (2685) or vaccines (2301) mour cells. The presence of occasional commonly develop in the frontal and tem-
become available. Comprehensive gen- gemistocytes in a diffuse astrocytoma poral lobes.
otyping studies have shown correlations does not justify the diagnosis (1533). Re-
between IDH mutation status and other ports have suggested that gemistocytic Macroscopy
molecular parameters; in particular, there astrocytoma may progress more rapidly Macroscopically, gemistocytic astrocy-
are strong associations between IDH mu- than standard diffuse astrocytoma to tomas are not substantially different from
tation and TP53 mutation (present in 94% anaplastic astrocytoma and secondary other low-grade diffuse gliomas. They
of cases) and ATRX inactivation (present glioblastoma, but these reports are from are often characterized by expansion of
in 86% of cases) (349). the pre-lDH era and it remains unclear the infiltrated brain areas without clear
whether JOH-mutant gemistocytic astro- delineation of the neoplasm. The involved
Mutant IDH catalyses the formation of cytomas have an increased tendency for areas may show greyish discolouration,
2-hydroxyglutarate, which could poten- anaplastic progression (319,1533) granularity, firmer or softer consistency,
tially be monitored by MR spectroscopy and microcystic change (1533).
(449) or in body fluids {358). However, ICD-0 code 9411/3
the clinical value of these approaches Microscopy
has yet to be validated. Grading The histopathological hallmark of gemis-
IDH-mutant gemistocytic astrocytoma cor- tocytic astrocytoma is the presence of a
responds histologically to WHO grade II. conspicuous proportion of gemistocytic
Gemistocytic astrocytoma, neoplastic astrocytes. Gemistocytes
/DH-mutant Epidemiology should account for more than approxi-
Gemistocytic astrocytomas account for mately 20% of all tumour cells; the pres-
Definition approximately 10% of all WHO grade 11 ence of occasional gemistocytes in a
A variant of /OH-mutant diffuse astrocy- diffuse astrocytomas (981). The mean diffuse astrocytoma does not justify the di-
toma characterized by the presence of reported patient age at diagnosis is agnosis of gemistocytic astrocytoma. The
a conspicuous {though variable) propor- 40 years {2703}, the median age is mean proportion of gemistocytes is ap-
tion of gemistocytic neoplastic astrocytes 42 years (1377), and the male-to-female proximately 35% (2703). The cut-off point
(gemistocytes). ratio is 2:1 (2703). of 20% is somewhat arbitrary, but a useful

f
..
.,:,
. ,
• .,; .........,
.... ...,..,. :
...
'
'.
.

'I
Fig. 1.10 Gemistocytic astrocytoma. A Tumour cells have enlarged, glassy, eosinophilic cytoplasm and eccentric nuclei. B lmmunostaining shows a marked and consistent
accumulation of GFAP in the cytoplasm of neoplastic gemistocytes. Interspersed are small tumour cells with little cytoplasm; proliferation is largely restricted to this inconspicuous cell
population. C p53 accumulation is present in nuclei of small and gemistocytic tumour cells.

22 Diffuse gliomas
Paediatric diffuse astrocytoma Genetic aspects remains to be determined whether the
Although the histopathology of paedi- Diffuse astrocytomas in children and prognosis of !DH-mutant gemistocytic
atric diffuse astrocytoma resembles adults have distinct genetic profiles. astrocytoma differs significantly from that
that of adult diffuse astrocytoma, there However, diffuse astrocytomas with ge- of IDH-mutant diffuse astrocytoma.
are many important distinctions be- netically defined so-called adult-type
tween the disease in children and in disease can present in adolescents,
adults. and so-called paediatric-type disease
can present in young adults. Paediatric Diffuse astrocytoma,
C!inicopathologica/ aspects diffuse astrocytomas are characterized IDH-wildtype
The annual incidence of paediatric dif- mainly by alterations in MYB and BRAF
fuse astrocytoma (defined by patient Amplification or rearrangements of MYB Definition
age < 20 years at diagnosis) is 0.27 cas- are detected in approximately 25% of A diffusely infiltrating astrocytoma without
es per 100 000 population; lower than paediatric diffuse astrocytomas {2518, mutations in the /OH genes.
that of adult diffuse astrocytoma, which 2855). Rearrangements of MYBL1 have IDH-wildtype diffuse astrocytoma is rare.
is 0,58 per 100 000 {1863). Most pae- also been described {2068}. Other pae- Most gliomas with a histological appear-
diatric diffuse astrocytomas are located diatric diffuse astrocytomas harbour ance resembling that of diffuse astrocy-
in the cerebral hemispheres, but a sig- BRAF V600E mutations, FGFR1 altera- toma but without IDH mutation can be re-
nificant proportion present in the thala- tions, or KRAS mutations {28551. Rare classified in adults as other tumours with
mus. which is an unusual site for adult paediatric diffuse astrocytomas contain additional genetic analyses. Tumours
diffuse astrocytoma. Anaplastic pro- the H3 K27M mutation usually found in that conform to this diagnosis most likely
gression occurs in approximately 75% paediatric high-grade gliomas {2855). constitute a variety of entities, and can
of adult lesions, but is rare in paediatric The mutations in IDH1, IDH2, TP53, and therefore follow a broad range of clini-
tumours (284). ATRX that are frequently found in adult cal courses. Thus. IDH-wildtype diffuse
diffuse astrocytomas are not present in astrocytoma is considered a provisional
the paediatric tumours (2443}. entity.

G/iomatosis cerebrigrowth pattern


criterion in borderline cases (1377,2556). displacement of nuclei to the periphery of Like other diffuse gliomas, diffuse astro-
The gemistocytes are characterized by the cell body. Expression of p53 protein cytoma can manifest at initial clinical pre-
plump, glassy, eosinophilic cell bodies of is also frequently seen in gemistocytes sentation with a gliomatosis cerebri pat-
angular shape. Stout, randomly oriented 127061 tern of extensive involvement of the CNS.
processes form a coarse fibrillary network. with the affected area ranging from most
These processes are often useful in dis- Genetic profile of one cerebral hemisphere (three lobes
tinguishing the tumour cells from the mini- Gemistocytic astrocytoma is a variant of or more) to both cerebral hemispheres
gemistocytes found in oligodendroglioma. I DH-mutant diffuse astrocytoma. Reports with additional involvement of the deep
Gemistocytic neoplastic astrocytes con- have noted that gemistocytic astrocyto- grey matter structures, brain stem, cer-
sistently express GFAP in their perikarya mas are characterized by a particularly ebellum, and spinal cord. See Anaplastic
and cell processes. The nuclei are usually high frequency of TP53 mutations, which astrocytoma, IOH-wildtype (p. 27) for ad-
eccentric, with small, distinct nucleoli and are present in > 80% of all cases (1834, ditional detail.
densely clumped chromatin. Perivascular 2703). and likely in nearly all cases of
lymphocyte cuffing is frequent {3221. Elec- IDH-mutant gemistocytic astrocytoma.
tron microscopy confirms the presence of The fact that TP53 mutations are present
abundant, compact glial filaments in the in both gemistocytes and non-gemis- Diffuse astrocytoma, NOS
cytoplasm and in cell processes. Enlarged tocytic tumour cells indicates that the
mitochondria have also been noted 1609}. gemistocytes are neoplastic and not re- Definition
active in nature (2094). This interpretation A tumour with morphological features of
Pro//feration is also supported by immunoreactivity to diffuse astrocytoma, but in which JOH
The gemistocytic neoplastic astrocytes mutant IDH1 protein {3591. mutation status has not been not fully
show a significantly lower rate of prolif- assessed.
eration than in the intermingled small- Prognosis and predictive factors Full assessment of !DH mutation status in
cell component {1046,1372,1377,27061 Gemistocytic astrocytomas have been diffuse astrocytomas involves sequence
However, microdissection has revealed reported to undergo progression to analysis for IOH1 codon 132 and /OH2
identical TP53 mutations in both gemi- anaplastic (gemistocytic) astrocytoma codon 172 mutations in cases that are
stocytes and non-gemistocytic tumour and I DH-mutant glioblastoma more com- immunohistochemically negative for the
cells 120941. monly than do other diffuse astrocytomas IOH1 R132H mutation.
{1377,1834,1929,1930}. However, these
lmmunophenotype data pertain to histologically diagnosed ICD-0 code 9400/3
The cytoplasm of neoplastic gemis- gemistocytic astrocytomas irrespective
tocytes is filled with GFAP, causing of the presence of an IDH mutation. It

Dittuse astrocytoma, IDH-wildtype 23


Anaplastic astrocytoma, von Deimling A. Berger M.S.
Huse JT Weller M.
Paulus W.
Wesseling P.
I DH-mutant Yan H.
Brat D.J.
Burger P.C.
Ellison D.W
Aldape KO.
Louis ON
Ohgaki H. Rosenblum M.K.
Kleihues P. Reifenberger G.

Definition Synonym diffuse astrocytomas, and IDH-mutant


A diffusely infiltrating astrocytoma with High-grade astrocytoma (discouraged) glioblastomas), are preferentially located
focal or dispersed anaplasia, significant in the frontal lobe.
proliferative activity, and a mutation in ei- Epidemiology
ther the IDH1 or IDH2 gene. The peak incidence of IDH-mutant ana- Clinical features
Anaplastic astrocytomas can arise from plastic astrocytoma occurs at a mean The symptoms are similar to those of
lower-grade diffuse astrocytomas, but patient age of 38 years {2103) However, WHO grade II diffuse astrocytoma. In
are more commonly diagnosed without until the discovery of IDH mutation as a some patients with a history of a diffuse
indication of a less-malignant precursor molecular marker, the diagnosis of ana- WHO grade II astrocytoma, there are in-
lesion. The presence of a component plastic astrocytoma was based only on creasing neurological deficits, seizures,
morphologically resembling oligodendro- histological evidence {1823,1827). Hospi- and signs of intracranial pressure (de-
glioma is compatible with this diagnosis tal-based data from the University of Zu- pending on the location of the tumour,
in the absence of 1p/19q codeletion. rich in the pre-lDH era showed a mean the degree of oedema, and the mass
Anaplastic astrocytomas have an intrin- patient age at diagnosis of approximately effect surrounding the lesion). Patients
sic tendency for malignant progression to 45 years, with a male-to-female ratio of with anaplastic astrocytoma commonly
I DH-mutant glioblastoma. 1.6:1. In one population-based study, the present after a history of a few months,
mean patient age at biopsy was 46 years with no evidence of a preceding WHO
ICD-0 code 9401/3 {1826). Population-based registry data grade 11 astrocytoma.
from the USA for the period 2007-2011
Grading show an annual incidence of 0.37 cas- Imaging
IDH-mutant anaplastic astrocytoma es per 100 000 population, a male-to- IDH-mutant anaplastic astrocytoma pre-
corresponds histologically to WHO female ratio of 1.39:1, and median patient sents as a poorly defined mass of low
grade Ill. Some retrospective studies age at diagnosis of 53 years {1863) In density. Unlike in WHO grade II diffuse
have shown that, in the setting of cur- a study that incorporated IDH mutation astrocytomas, partial contrast enhance-
rent therapy, IDH-mutant anaplastic as- data and included 562 IDH-mutant ana- ment is usually observed, but the central
trocytomas may follow clinical courses plastic astrocytomas, the median patient necrosis with ring enhancement typical
only somewhat worse than those of IDH- age at presentation was 36 years and of glioblastomas is absent. More rapid
mutant diffuse astrocytomas (WHO the mean age 38 years, similar to that for tumour growth with development of peri-
grade II) {870,2103,2464). but this was IDH-mutant WHO grade II diffuse astro- tumoural oedema can lead to mass shifts
not found in other studies {1268}. Al- cytoma {2103) and increased intracranial pressure.
though IDH-mutant anaplastic astro-
cytoma is assigned a WHO grade of Ill Localization Spread
based on histological appearance, grad- I DH-mutant anaplastic astrocytomas can Like other diffuse gliomas, anaplastic as-
ing algorithms for distinguishing between develop in any region of the CNS but most trocytomas are generally characterized
IDH-mutant anaplastic astrocytoma and frequently occur in the cerebrum. These by diffuse infiltrative growth in the brain
IDH-mutant diffuse astrocytoma may tumours, like other IDH-mutant diffuse {463). Occasionally, leptomeningeal
need to be refined. gliomas (including oligodendrogliomas, spread is evident {1059).

A B
Fig. 1.11 A Anaplastic astrocytoma in the right frontotemporal region. Note the ill-defined borders with the adjacent brain structures and focal cysts. B Frontotemporal anaplastic
astrocytoma containing a large cyst but no macroscopically discernible necrosis. C Anaplastic astrocytoma with diffuse, bilateral infiltration of the corpus callosum, the caudate
nucleus, and the fornices. The fornices are grossly enlarged and show petechial haemorrhages.

24 Diffuse gliomas
Macroscopy astrocytomas (i.e. gliomas that lack IDH
Like WHO grade II diffuse astrocytoma, mutations) (953f.
anaplastic astrocytomas tend to infiltrate
the surrounding brain without causing Genetic profile
frank tissue destruction. This often leads The molecular features of anaplastic as-
to a marked enlargement of invaded trocytoma largely recapitulate those of
structures, such as adjacent gyri and WHO grade II IDH-mutant diffuse astro-
basal ganglia. On cut surface, the higher cytoma. By definition, mutations in IOH1
cellularity of the anaplastic astrocytoma or IDH2 are present in all tumours, and


results in a discernible tumour mass, TP53 and ATRX alterations are found in
which in some cases is distinguished Fig. 1.12 Anaplastic astrocytoma. Smear preparations the majority of tumours (118,953,1160,
more clearly from the surrounding brain show various degrees of nuclear atypia. 1215,1834,1895,2092, 2704, 281 Of.
structures than is seen in WHO grade II Robust molecular correlates of anaplastic
diffuse astrocytomas. Macroscopic cysts astrocytoma at one end of the range and progression within diffuse astrocytoma
are uncommon, but there are often areas with glioblastoma at the other (492,1137, lineages have yet to be established, be-
of granularity, opacity, and soft consist- 1223,2059!. The index may vary con- cause the biological distinctions between
ency. It is often difficult to grossly dis- siderably, however, even within a single IDH-mutant and IDH-wildtype tumours
tinguish between a WHO grade Ill ana- tumour. have emerged only recently. However,
plastic astrocytoma and a WHO grade II compared with WHO grade II I DH-mutant
diffuse astrocytoma. lmmunophenotype astrocytomas, WHO grade Ill tumours
In general, the immunohistochemical have higher frequencies of chromosome
Microscopy features of anaplastic astrocytoma re- arm 9p and 19q losses {349,12691
The principal histopathological features capitulate those of WHO grade II diffuse
are those of a diffusely infiltrating astro- astrocytoma, consistent with their shared Prognosis and predictive factors
cytoma with increased mitotic activity histogenetic and molecular foundations.
compared with the WHO grade II equiva- IDH-mutant anaplastic astrocytomas are Clinical prognostic factors
lent, usually accompanied by distinct nu- typically positive for GFAP and frequently Historically, median survival has been in
clear atypia and high cellularity. Mitotic exhibit strong and diffuse nuclear expres- the range of 3-5 years, but with marked
activity should be evaluated in the con- sion of p53. The majority express R132H- differences in cases with older patient
text of sample size. In small specimens, mutant IDH1 (reflecting their underlying age and low performance status, both
such as those obtained at stereotactic IDH mutation status) and display nega- of which are associated with inferior
biopsy, a single mitosis suggests signifi- tive immunostaining for nuclear ATRX. outcome (2740f. In the era of subtyping
cant proliferative activity; in such cases, anaplastic astrocytomas by IDH mutation
Ki-67 labelling may be helpful. In large Cell of origin status, survival estimates now vary more
resection specimens, a few mitoses are The cell of origin is unknown. The fact widely (see Genetic alterations). The ex-
not sufficient for WHO grade Ill designa- that diffuse WHO grade II and Ill astro- tent of surgical resection at diagnosis
tion (826l Regional or diffuse hypercellu- cytomas, IDH-mutant glioblastomas, also seems to impact outcome (2740f.
larity is an important diagnostic criterion; and oligodendrogliomas all carry an IDH
but even in the setting of low cellularity, mutation suggests that they may share a Prol!feration
the diagnosis is still appropriate if there cell of origin different from those of IDH- Proliferative activity, as estimated by
is significant mitotic activity. With pro- wildtype glioblastomas and pilocytic mitotic count or the Ki-67 proliferation
gressive anaplasia, nuclear morphology index, is not prognostic for anaplastic
becomes more atypical, with increasing astrocytomas.
variation in nuclear size, shape, coarse-
ness, and dispersion of chromatin and Histopathological factors
increasing nucleolar prominence and Histological factors are not associated
quantity. Additional signs of anaplasia with outcome of anaplastic astrocytoma,
include multinucleated tumour cells and but they have not yet been carefully eval-
abnormal mitoses, but these are not ob- uated within the context of IDH-mutant
ligatory for WHO grade Ill. By definition, anaplastic astrocytoma.
microvascular proliferation (multilayered
vessels) and necrosis are absent. Genetic alterations
IDH1/2 mutations are associated with
Prol!feration better outcome, whereas IDH-wildtype
Unlike WHO grade II diffuse astrocy- anaplastic astrocytoma has an outcome
toma, anaplastic astrocytoma displays similar to that of IDH-wildtype glioblasto-
mitotic activity. The growth fraction as de- ma (952l. A study that included 562 IDH-
termined by the Ki-67 proliferation index mutant anaplastic astrocytomas from
is usually in the range of 5-10%, but can Fig. 1.13 Anaplastic astrocytoma. Moderate cellularity, three series showed a median survival
overlap with values for low-grade diffuse marked nuclear atypia, and mitoses. of 9.3 years (2103). EGFR amplification

Anaplastic astrocytoma, IDH-mutant 25


.,
. - -
Fig. 1.14 Anaplastic astrocytoma. A Marked nuclear pleomorphism. Note the atypical mitosis in the centre. B Hypercellularity and hyperchromatic, irregular, so-called naked nuclei
appearing within a fibrillary background. Two mitotic figures are present.

;.,,.,...,. ..
.
,:: � --\.
7.- c • � - ,o; .....
Fig. 1.15 IDH-mutant anaplastic astrocytoma. A Well-delineated focus with higher cellularity, mitotic activity, and a lack of GFAP expression (left). Such foci are encountered in
anaplastic astrocytomas and glioblastomas and may represent new clones resulting from the acquisition of additional genetic alterations, indicating progression to a higher grade of
dedifferentiation and malignancy (750). B GFAP immunoreactivity. C lmmunoreactivity for the proliferation marker MIB1, including a cell in mitosis.

• SNV or indel • Amplification • Deletion • SV • Fusion • Two or more aberrations and a genotype of 7q gain and 10q loss
have been associated with worse out-
LGG with /OH Mutation LGG with /OH Mutation
and lp/19q Codeletion and No lp/19q Codeletion LGG with Wild-Type /DH GBM with Wild-Type /OH come. !DH mutations may be useful as a
0 0.5 1 0 0.5 1 0 0.5 l O 0.5 1
predictive biomarker when !DH-targeted
TPSJ
therapies such as small-molecule inhibi-
RBl tors (2685} or vaccines (2301} become
PTEN available.
PIKJR
PIKJC
PIKJC2 Mutant !DH catalyses the formation of
POGFR 2-hydroxyglutarate, which could poten-
NFl
MET tially be monitored by MR spectroscopy
MOM4 {449) or in body fluids {358f. However,
MOM2
the clinical value of these approaches
MOMJ
IDHl has yet to be validated.
FGFRJ
FGFR2
FGFRl
EGFR
COKN2C
COKNlA
COK6
CDK4
BRAF
ATRX
0 0.5 1 0 0.5 1 0 0.5 1 0 0.5
Fraction of Samples with Specific Alteration in Gene
Fig. 1.16 Mutations in 'lower-grade' (i.e. WHO grade II and Ill) gliomas (LGGs), detected in The Cancer Genome
Atlas (TCGA) series (349). Note that the mutational pattern in LGG with wildtype IDH is similar to that of glioblastoma
(GBM) with wildtype IDH. LGGs with IDH mutation are divided into oligodendroglial tumours with 1p/19q codeletion and
astrocytic tumours with frequent TP53 and ATRX mutations but no 1 p/19q codeletion. SNV, single nucleotide variant;
SV, structural variant. Reprinted from: Brat DJ et al., Cancer Genome Atlas Research Network (349).

26 Diffuse gliomas
······························································
Anaplastic astrocytoma, G!iomatosis cerebri growth pattern Anaplastic astrocytoma, NOS
tDH-wildtype Like other diffuse gliomas, anaplastic
astrocytoma can manifest at initial clini- Definition
Definition cal presentation with a gliomatosis cer- A tumour with morphological features
A diffusely infiltrating astrocytoma with ebri pattern of extensive involvement of of anaplastic astrocytoma, but in which
focal or dispersed anaplasia and signif- the CNS, with the affected area ranging /OH mutation status has not been fully
icant proliferative activity but without mu- from most of one cerebral hemisphere assessed.
tations in the !OH genes. (three lobes or more) to both cerebral Full assessment of IOH mutation status
IDH-wildtype anaplastic astrocytoma is hemispheres with additional involvement in anaplastic astrocytomas involves se-
uncommon and accounts for about 20% of the deep grey matter structures, brain quence analysis for /OH1 codon 132 and
of all anaplastic astrocytomas. Nonethe- stem, cerebellum, and spinal cord. A IOH2 codon 172 mutations in cases that
less. histologically defined anaplastic similar extensive, diffuse involvement of are immunohistochemically negative for
astrocytomas have the highest incidence the deep grey matter structures (i.e. ba- the !OH1 R132H mutation.
of wildtype !OH1 and IOH2 among the sal ganglia and thalamus), brain stem,
WHO grade II and Ill diffuse glioma vari- cerebellum, and spinal cord can also be ICD-0 code 9401/3
ants 11269,2810}. Most gliomas with a seen in the absence of cerebral corti-
histological appearance resembling that cal involvement. Gliomatosis was once Grading
of anaplastic astrocytoma but without thought to be a distinct nosological en- Anaplastic astrocytoma, NOS, corre-
IDH mutation share molecular features tity, but is now considered to be a pat- sponds histologically to WHO grade Ill.
with IDH-wildtype glioblastoma, and tern of exceptionally widespread involve-
sometimes with H3 K27M-mutant glio- ment of the neuraxis. It can be seen in
mas if located preferentially in midline any of the diffuse glioma subtypes, but
locations. Tumours in this category are is most common in anaplastic astrocy-
more clinically aggressive than are IDH- toma. There are no unique molecular
mutant anaplastic astrocytomas and may signatures that distinguish gliomatosis
follow a clinical course more similar to from the well-characterized subtypes of
that of glioblastoma. diffuse glioma, but IDH mutations seem
to be restricted to tumours with distinct
Grading solid tumour components (2313}.
IDH-wildtype anaplastic astrocytoma
corresponds histologically to WHO
grade Ill.

Anaplastic astrocytoma, IDH-wildtype 27


Glioblastoma, IDH-wildtype Louis D.N.
Suva M.L.
Brat D.J.
Biernat W.
Ohgaki H.
Cavenee W.K.
Stupp R.
Hawkins C.
Burger P.C. Bigner DO. Wick W. Verhaak R.G.W.
Perry A. Nakazato Y. Barnholtz-Sloan J. Ellison D.W.
Kleihues P. Plate K.H. Rosenblum M.K. van Deimling A.
Aldape K.D. Giangaspero F. Hegi M.

Definition Genetic parameters H3 K27M mutation has also been ex-


A high-grade glioma with predominantly The genetic alterations typical of IDH- cluded) and with no history of a pre-
astrocytic differentiation; featuring nucle- wildtype glioblastoma include TERT existing lower-grade glioma. Although
ar atypia, cellular pleomorphism (in most promoter mutations (present in -80% it is not possible to establish an exact
cases), mitotic activity, and typically a dif- of cases), homozygous deletion of patient age cut-off point, one algorithm
fuse growth pattern, as well as microvas- CDKN2A/COKN2B (-60%), loss of has suggested that, in the setting of
cular proliferation and/or necrosis; and chromosomes 10p (-50%) and 10q negative R132H-mutant IDH1 immuno-
which lacks mutations in the !DH genes. (-70%), EGFR alterations (i.e. mutation, histochemistry in a glioblastoma from a
IDH-wildtype glioblastoma is the most rearrangement, altered splicing, and/ patient without prior lower-grade glio-
common and most malignant astrocytic or amplification; -55%), PTEN muta- ma, the probability of an alternative IDH
glioma, accounting for about 90% of all tions/deletion (-40%), TP53 mutations mutation is < 6% in a 50-year-old pa-
glioblastomas and typically affecting (25-30%), and Pl3K mutations (-25%) tient and decreases to < 1% in patients
adults, with a mean patient age at diag- {277,1830). aged > 54 years {425). The designation
nosis of 62 years and a male-to-female The prognosis of IDH-wildtype glio- "IDH-wildtype" can therefore be safely
ratio of about 1.35:1. As the synonymous blastoma with current therapies is poor. applied in this setting, even in the ab-
designation "IDH-wildtype primary glio- Determining MGMT promoter methyla- sence of IDH sequencing. However, in
blastoma" indicates, this glioblastoma tion status provides information in these younger patients, certain findings more
typically arises de nova, with no recog- tumours on response to alkylating and strongly suggest the need for IDH se-
nizable lower-grade precursor lesion. methylating chemotherapies. quencing before designating a tumour
A preferentially supratentorial location Ideally, the designation "IDH-wildtype" as IDH-wildtype. These include a his-
is characteristic. The tumour diffusely should be applied to a glioblastoma tory of a lower-grade glioma and the
infiltrates adjacent and distant brain when both R132H-mutant IDH1 im- absence of nuclear ATRX expression
structures. munohistochemistry and subsequent (particularly if p53 immunohistochem-
!OH1/2 sequencing reveal wildtype se- istry shows strong and diffuse nuclear
ICD-0 code 9440/3 quences at IDH1 codon 132 and IDH2 positivity and in the absence of stain-
codon 172. However, in some situations ing for K27M-mutant H3.3). In such a
Grading it may, for practical purposes, be suffi- setting, if IDH sequencing cannot be
Glioblastoma and its variants correspond cient to rely on negative R132H-mutant performed, a diagnosis of glioblastoma,
histologically to WHO grade IV However, IDH1 immunohistochemistry alone, NOS, should be rendered, with a note
in the setting of current therapy, IDH- most notably in older patients with a stating that R132H-mutant IDH1 immu-
mutant glioblastomas may follow a clini- histologically classic glioblastoma that nohistochemistry was negative.
cal course that is less aggressive than is is not in a midline location (unless an
typical of WHO grade IV tumours.

Synonym

-r: i..---
Primary glioblastoma, IDH-wildtype 100
90
Epidemiology 80
/ /
/ �/
70
Incidence
Glioblastoma is the most frequent malig- ;f!. 60
I /
nant brain tumour in adults, accounting 1 so I /
for approximately 15% of all intracranial
..!!!
E 40
I /
neoplasms and approximately 45-50% ::,
u 30 / /

-
of all primary malignant brain tumours
20
/ /
{1826,1863). In most European and North _/ /
American countries and in Australia, the
annual incidence is about 3-4 cases per
10
0
v
100 000 population {1863}. whereas the 0 10 20 30 40 so 60 70 80 90 100
incidence is relatively low in eastern Asia, Age at diagnosis Female - Male
with 0.59 cases per 100 000 population Fig. 1.17 Cumulative age distribution of patients with IDH-wildtype glioblastoma. There is a tendency for an earlier
per year in the Republic of Korea, for manifestation in women. Based on 371 cases from Nobusawa Set al. {1797).

28 Diffuse gliomas
example (1861 }. The annual incidence A series of environmental and genetic rostral lateral ventricles {1417). In general,
of glioblastoma in the USA, adjusted to factors have been studied as potential tumour infiltration extends into the adja-
the United States Standard Population, causes of glioblastoma. To date, these cent cortex and through the corpus cal-
is 3.19 cases per 100 000 population investigations have yielded inconclusive losum into the contralateral hemisphere.
(1863). The corresponding rate in a popu- or negative results. including results on Glioblastoma of the basal ganglia and
lation-based study in Switzerland (adjust- the potential influence of non-ionizing ra- thalamus is common, especially in chil-
ed to the European Standard Population) diation (e.g. from mobile phones) and oc- dren. Glioblastoma of the brain stem
was 3.55 cases per 100 000 population cupational exposures. The only validated is uncommon and most often affects
{1826). Significantly lower rates have risk factor associations are an increased children {595) (see also Diffuse midline
been reported in Asian and African coun- risk after ionizing radiation to the head glioma, H3 K27M-mutant, p. 57). The
tries. but this may be due in large part to and neck and a decreased risk among cerebellum and spinal cord are rare sites.
underascertainment. individuals with a history of allergies and/
or atopic disease(s) {1861 ). Genome- Clinical features
Age and sex distnbution wide association studies have identified Glioblastomas develop rapidly. The
Glioblastoma can manifest in patients some specific heritable risk variants as- symptoms depend largely on the tumour
of any age, but preferentially affects sociated with glioblastoma (see Genetic location, primarily manifesting as focal
older adults. with peak incidence occur- susceptibility, p. 42). As our understand- neurological deficits (e.g. hemiparesis
ring in patients aged 55-85 years. Glio- ing of the heterogeneity of glioblastoma and aphasia) and tumour-associated
blastoma is the second most common increases with the use of genomic tech- oedema with increase in intracranial
type of intracranial neoplasm in adults nologies, our ability to discover and vali- pressure. As many as half of all patients
aged � 55 years {1863}. Glioblastomas date glioblastoma subtype-specific risk are diagnosed after an inaugural seizure.
are rare in individuals aged < 40 years. factors will probably improve. Other common symptoms are behaviour-
In the USA. the median age of patients al and neurocognitive changes. nausea
with glioblastoma is 64.0 years, and the Localization and vomiting, and occasionally severe
annual incidence rate in the 0-19 years Glioblastoma is most often centred in pulsating headaches {557,2640,2733) In
age group, adjusted to the United States the subcortical white matter and deeper a study of 677 patients with IDH-wildtype
Standard Population, is 0.14 new cases grey matter of the cerebral hemispheres. glioblastoma, the time from first symp-
per 100 000 population. The median In a series of 987 glioblastomas from toms to diagnosis was < 3 months in 68%
patient age at diagnosis of IDH-wildtype the University Hospital Zurich, the most of cases and < 6 months in 84% {1827).
glioblastomas is 62 years. The male-to- frequently affected sites were the tem- In patients with a significantly longer
female ratio for glioblastoma is 1.60:1 in poral lobe (affected in 31% of cases). duration of symptoms, the possibility of
the USA {1863) and 1.28:1 in Switzerland the parietal lobe (in 24%), the frontal an IDH-mutant glioblastoma that has
{1825). lobe (in 23%), and the occipital lobe (in evolved from a lower-grade astrocytoma
16%). Similar location trends are seen in should be considered.
Etiology the USA {1863). Whereas primary, IDH-
A very small proportion of glioblastomas wildtype glioblastomas have a wide- Imaging
are inherited as part of specific Mende- spread anatomical distribution. second- Glioblastomas are irregularly shaped and
lian syndromes (see Genetic suscep- ary, IDH-mutant glioblastomas have a have a ring-shaped zone of contrast en-
tibility, p. 42) {1861). but the etiology of striking predilection for the frontal lobe, hancement around a dark, central area
most glioblastomas remains unknown. particularly in the region surrounding the of necrosis. They may extend widely

Fig. 1.18 IDH-wildtype glioblastoma. A This tumour appears multifocal on postcontrast T1-weighted MRI. B The corresponding FLAIR image of this 'multifocal' glioma shows an
abnormal signal connecting the seemingly separate foci of contrast enhancement. C This postcontrast T1-weighted image shows the typical features of a 'butterfly glioblastoma' with
extensive involvement of the corpus callosum leading to bihemispheric spread.

Glioblastoma. IDH-wildtype 29
treated with antiangiogenic therapies,
presumably due to vascular normaliza-
tion (542).

Gliomatosis cerebri growth pattern


Like other diffuse gliomas, glioblastoma
can manifest at initial clinical presenta-
tion with a gliomatosis cerebri pattern of
extensive involvement of the CNS, with

68 days the affected area ranging from most of
one cerebral hemisphere (three lobes
or more) to both cerebral hemispheres
with additional involvement of the deep
grey matter structures, brain stem, cer-
ebellum, and spinal cord. See Anap/astic
astrocytoma, IDH-wildtype (p. 27) for ad-
ditional detail.
Fig. 1.19 Rare case in which the rapid development of a primary glioblastoma, IDH-wildtype, could be followed by
neuroimaging. After a seizure, the 79-year-old man was hospitalized and MRI showed a small cortical lesion of 1 cm
in diameter. Only 2 months later, the patient presented with a full-blown glioblastoma with ring enhancement, central Metastasis
necrosis, and perifocal oedema {1533, 1822}. Despite its rapid, infiltrative growth, glio-
blastoma does not commonly extend
into adjacent lobes, the opposite hemi- Other infiltrative patterns give rise to into the subarachnoid space or spread
sphere, and the brain stem. In the set- secondary structures of Scherer, includ- through the cerebrospinal fluid, although
ting of a ring-enhancing mass, biopsies ing perineuronal satellitosis, perivascular this may be more frequent in children (90,
showing high-grade astrocytoma but not aggregation, and subpial spread (2838} 880) Similarly, penetration of the dura,
demonstrating frank histological features Infiltrative cells are located both inside venous sinus, and bone is exceptional
of glioblastoma should be suspected to and outside of the contrast-enhancing (812,2002). Although extension within
have been inadequately sampled. rim of glioblastoma and generally create and along perivascular spaces is typi-
a gradient of decreasing cell density with cal, invasion of the vessel lumen is not a
Spread increasing distance from the tumour cen- frequent histological finding. Extracranial
Infiltrative spread is a defining feature of tre. Individual infiltrating tumour cells can metastasis of glioblastoma is uncommon
all diffuse gliomas, but glioblastoma is be histologically identified several centi- in patients without previous surgical in-
particularly notorious for its rapid invasion metres from the tumour epicentre, both in tervention, but has been documented in
of neighbouring brain structures {316}. regions that are T2-hyperintense on MRI patients who have undergone interstitial
Infiltration occurs most readily along and in regions that appear uninvolved. therapies and in patients with ventricular
white matter tracts, but can also involve These infiltrating cells are the most likely shunts (935,1546,2676}. More recently,
cortical and deep grey structures. When source of local recurrence after initial circulating tumour cells have been found
infiltration extends through the corpus therapy, because they escape surgi- in the blood of some patients with glio-
callosum, with subsequent growth in the cal resection, do not receive the highest blastoma, suggesting that immune mech-
contralateral hemisphere, the result can dose of radiotherapy, and involve regions anisms or inhospitable environments of
be a bilateral, symmetrical lesion (so- with an intact blood-brain barrier (which distant organs suppress metastatic im-
called butterfly glioma). Similarly, rapid diminishes chemotherapeutic bioavail- plantation and growth (2454). The find-
spread is observed along white matter ability) (834). Interestingly, a pattern of ing that immunosuppressed recipients of
tracts of the internal capsule, fornix, an- increased infiltration has been observed organ transplants from donors with glio-
terior commissure, and optic radiation. in a subset of patients with glioblastoma blastoma have developed glioblastoma
in their transplanted organ suggests that
the immune system normally suppresses
the metastatic potential of circulating tu-
mour cells (1161).

Mechanisms of invasion
Mechanisms that promote the invasive
properties of glioblastoma cells include
those involved with cell motility, cell-ma-
trix and cell-cell interactions, and remod-
elling of the extracellular matrix, as well
A as microenvironmental influences (160,
Fig. 1.20 A Glioblastoma with bilateral, symmetrical invasion of the corpus callosum and adjacent white matter of the 573). Tumour cells produce migration-
cerebral hemispheres (butterfly glioblastoma). B Unusual case of a glioblastoma with focal infiltration of the cerebral enhancing extracellular matrix compo-
cortex and the adjacent subarachnoid space, macroscopically presenting as greyish thickening of the meninges. nents and secrete proteolytic enzymes

30 Diffuse gliomas
tumours and that in approximately 3%
of these, the tumour foci differ in histo-
logical appearance (128). True multifocal
glioblastomas are most likely polyclonal if
they occur infratentorially and supratento-
rially (i.e. outside easily accessible routes
such as the cerebrospinal fluid pathways
or the median commissures) {2228}. By
definition, multiple independently arisinq
gliomas must be of polyclonal origin, and
A their existence can only be proven by ap-
plication of molecular markers, which in
informative cases enable the distinction
between tumours of common or inde-
pendent origin {175,197,1359).

Macroscopy
Despite the short duration of symptoms
in many cases, glioblastomas are often
surprisingly large at the time of presenta-
c <, tion, and can occupy much of a lobe. The
Fig. 1.21 A Glioblastoma of the right frontotemporal region with infiltration of the basal ganglia, compression of the lesions are usually unilateral, but those in
right lateral ventricle, and midline shift towards the contralateral (left) cerebral hemisphere. B Large, diffusely infiltrating the brain stem and corpus callosum can
glioblastoma of the left frontal lobe with typical coloration: whitish-grey tumour tissue in the periphery, yellow areas be bilaterally symmetrical. Supratentorial
of necrosis, and extensive haemorrhage. Note the extension through the corpus callosum into the right hemisphere.
bilateral extension occurs as a result of
c Symmetrical glioblastoma infiltrating the lateral ventricles and adjacent brain structures. D Large glioblastoma of
the brain stem (pons), causing compression of the fourth ventricle. This location is typical of H3 K27M-mutant diffuse growth along myelinated structures, in
midline gliomas. particular across the corpus callosum
and the commissures. Most glioblas-
tomas of the cerebral hemispheres are
that permit invasion, including the matrix angiogenic mechanisms and direct ef- clearly intraparenchymal with an epicen-
metalloproteinases MMP2 and MMP9, fects that enhance glioma cell migra- tre in the white matter. Infrequently, they
uPA and its receptor uPAR, and cath- tion {1239,2840). Hypoxic tumour cells are largely superficial and in contact with
epsins. Gliomas also express a variety display elevated expression of extracel- the leptomeninges and dura and may
of integrin receptors that mediate interac- lular matrix components and intracellu- be interpreted by the neuroradiologist or
tions with molecules in the extracellular lar proteins associated with cell motility surgeon as metastatic carcinoma, or as
space and lead to alterations of the cel- (264,2170). Activation of a pro-migration an extra-axial lesion such as meningi-
lular cytoskeleton and activation of intra- transcriptional programme seems to be oma. Cortical infiltration may produce a
cellular signalling networks such as the associated with a decrease in prolifera- preserved gyriform rim of thickened grey
AKT, mTOR, and MAPK pathways. Many tion, which may have therapeutic conse- cortex overlying a necrotic zone in the
growth factors expressed in glioblasto- quences {834,835). white matter.
ma, such as hepatocyte growth factor, fi- Glioblastomas are poorly delineated;
broblast growth factor, epidermal growth Mult1foca/ g/ioblastoma the cut surface is variable in colour, with
factor, and VEGF, also stimulate migra- Although multifocality is not unusual when peripheral greyish tumour masses and
tion by activation of corresponding re- defined radiologically, the incidence of central areas of yellowish necrosis from
ceptor tyrosine kinases and downstream truly multiple, independent gliomas oc- myelin breakdown. The peripheral hyper-
mediators that more directly promote mi- curring outside the setting of inherited cellular zone presents macroscopically
gration, including FAK and the Rho fam- neoplastic syndromes is unknown. Even
ily GTPases Rae, RhoA, and CDC42. In careful postmortem studies on whole-
EGFR-amplified glioblastomas, cells with brain sections do not always reveal a
amplification are preferentially located at connection between apparently multifo-
the infiltrating edges, suggesting a role in cal gliomas, because the cells infiltrat-
peripheral expansion (2385). The overall ing along myelinated pathways are often
mass migration of glioblastoma is radially small, polar, and largely undifferentiated.
outward, away from central necrosis and One careful histological analysis {143}

.�
associated severe hypoxia, with rates concluded that 2.4% of glioblastomas
substantially greater than those of pre- are truly multiple independent tumours,
necrotic gliomas (2170,2467). Hypoxia
promotes invasion through the activa-
a value similar to that reported by others
(2.3%) (2204). A postmortem study found
_,
Fig. 1.22 IDH-wildtype glioblastoma. This intraoperative
tion of HIF1 and other hypoxia-inducible that 7.5% of gliomas (including oligoden- smear preparation shows small, elongated bipolar cells,
transcription factors, due to both pro- drogliomas) are multiple independent a characteristic component of glioblastomas (1956).

Glioblastoma, IDH-wildtype 31
.,

The presence of highly anaplastic glial


cells, mitotic activity, and microvascular
proliferation and/or necrosis is required.
The distribution of these key features
within the tumour is variable, but large
necrotic areas usually occupy the tumour
centre, whereas viable tumour cells tend
to accumulate in the periphery. The cir-
cumferential region of high cellularity
and abnormal vessels corresponds to
the contrast-enhancing ring seen radio-
logically, and is an appropriate target for
needle biopsy. Microvascular prolifera-
tion is seen throughout the lesion, but is
usually most marked around necrotic foci
and in the peripheral zone of infiltration.

Cellular composition and


histological patterns
Few human neoplasms are as heteroge-
neous in composition as glioblastoma.
Fig. 1.23 Diagnostic hallmarks of IDH-wildtype glioblastoma. A focus of ischaemic necrosis (NE) is surrounded by
palisading tumour cells and hyalinized vascular proliferation (VP).
Poorly differentiated, fusiform, round, or
pleomorphic cells may prevail, but bet-
ter-differentiated neoplastic astrocytes
are usually discernible, at least focally
!317). This is particularly true of glioblas-
tomas resulting from the progression of
WHO grade II diffuse astrocytomas, but
these are typically IDH-mutant glioblas-
tomas. The transition between areas that
still have recognizable astrocytic differ-
entiation and highly anaplastic cells may
be either continuous or abrupt. In the
Fig. 1.24 Glioblastoma, IDH-wildtype. A Microvascular proliferation in glioblastomas often have a glomeruloid case of gemistocytic lesions, anaplas-
appearance. B Palisading necrosis is characterized by irregular zones of necrosis surrounded by dense accumulations tic tumour cells may be diffusely mixed
of tumour cells. with the differentiated gemistocytes. An
abrupt change in morphology may reflect
as a soft, grey to pink rim or a grey band tumour cells with nuclear atypia and brisk the emergence of a new tumour clone
of tumour tissue. However, necrotic tissue mitotic activity. Prominent microvascular through the acquisition of one or more
may also border adjacent brain structures proliferation and/or necrosis is an essen- additional genetic alterations (see Primi-
without an intermediate zone of macro- tial diagnostic feature. tive neuronal cells and glioblastoma with
scopically detectable tumour tissue. The As the outdated term 'glioblastoma rnul- a primitive neuronal component, below)
central necrosis can occupy as much as tiforme' suggests, the histopathology of !750). Cellular pleomorphism includes
80% of the total tumour mass. Glioblas- this tumour is extremely variable. Some the formation of small, undifferentiated,
tomas are typically stippled with red and lesions show a high degree of cellular lipidized, granular, and giant cells. There
brown foci of recent and remote haem- and nuclear polymorphism with numer- are also often areas where bipolar, fusi-
orrhage. Extensive haemorrhages can ous multinucleated giant cells; others form cells form intersecting bundles,
occur and cause stroke-like symptoms, are highly cellular but relatively mono- and fascicles prevail. The accumulation
which are sometimes the first clinical sign morphic. The astrocytic nature of the of highly polymorphic tumour cells with
of the tumour. Macroscopic cysts, when neoplasms is easily identifiable (at least well-delineated plasma membranes and
present, contain a turbid fluid and con- focally) in some tumours, but difficult to a lack of cell processes may mimic meta-
stitute liquefied necrotic tumour tissue, in recognize in tumours that are poorly dif- static carcinoma or melanoma.
contrast to the well-delineated retention ferentiated. The regional heterogeneity Several cellular morphologies appear
cysts present in WHO grade II diffuse of glioblastoma is remarkable, making commonly in glioblastomas. Some glio-
astrocytomas. histopathological diagnosis difficult on blastomas have well-recognized pat-
specimens obtained by stereotaxic nee- terns that are characterized by a great
Microscopy dle biopsies !317) predominance of a particular cell type.
Glioblastoma is typically a highly cellu- The diagnosis of glioblastoma is often These morphologies are discussed in
lar glioma, usually composed of poorly based on the identification of the tissue the following subsections, along with the
differentiated, sometimes pleomorphic pattern rather than of specific cell types. corresponding glioblastoma patterns that

32 Diffuse gliomas
can be established if a particular cellu- mitotic activity. In the zone of infiltration, IDH mutations are absent {1183,1937).
lar morphology predominates. Because tumour cells can be difficult to identify In one series, mutations of TP53 were
most of these glioblastoma patterns are as neoplastic, given their small size and found to be slightly less common in this
found in IDH-wildtype glioblastomas, bland cytology. GFAP immunoreactivity subtype, but the difference did not reach
they are discussed here, but it is recog- variably highlights delicate processes, statistical significance (1031 ). The clini-
nized that some of these variants (e.g. and the Ki-67 proliferation index is typi- cal behaviour of the small-cell subtype is
gemistocytic astrocytomas progressing cally high. Due to their nuclear regular- similar to that of other primary glioblas-
to glioblastoma) are more characteristic ity, clear haloes, microcalcifications, tomas in general, with a median survival
of !DH-mutant glioblastoma. and chicken wire-like microvasculature, time of 11 months in one series (1937).
these tumours overlap with anaplastic In the same series, about a third of the
Small cells and small cell glioblastoma oligodendroglioma (1937). But unlike cases presented as non-enhancing or
This subtype features a predominance oligodendrogliomas, small cell glioblas- minimally enhancing masses with no
of highly monomorphic small, round to tomas frequently have EGFR amplifi- evidence of microvascular proliferation or
slightly elongated, hyperchromatic nu- cation (present in -70% of cases) and necrosis on histology. However, follow-up
clei with minimal discernible cytoplasm, chromosome 10 losses (in > 95%). As in imaging 2-3 months later often showed
little nuclear atypia, and (often) brisk other primary glioblastomas in general, ring enhancement, and survival times
were shorter for these cases (median:
6 months), suggesting that these cases
constitute early presentations of WHO
grade IV glioblastoma rather than WHO
grade Ill anaplastic astrocytoma {1937).

Prirmtive neuronal cells and glioblastoma


with a primitive neuronal component
This subtype constitutes an otherwise
classic diffuse glioma with one or more
solid-looking primitive nodules showing
neuronal differentiation. The glioma com-
ponent is typically astrocytic, although
rare primitive neuronal foci have also been
reported in oligodendrogliomas {1946)
The primitive foci are sharply demar-
cated from the adjacent glioma and dis-
play markedly increased cellularity and
a high nuclear-to-cytoplasmic ratio and
mitosis-karyorrhexis index. More variable
features include Homer Wright rosettes,
cell wrapping, and anaplastic cytol-
ogy similar to that of medulloblastoma or
other CNS embryonal neoplasms. Addi-
tional primitive neuronal features include
immunoreactivity for neuronal markers
such as synaptophysin, reduction or loss
Fig. 1.26 Glioblastoma with a primitive neuronal component. A Diffuse astrocytoma component on the right and of GFAP expression, and a markedly ele-
primitive neuronal component on the left. B Homer Wright rosettes within the primitive neuronal component of a vated Ki-67 proliferation index compared
glioblastoma. C Strong synaptophysin positivity in the primitive cells. with adjacent foci of glioma. This subtype

Glioblastoma, IDH-wildtype 33
.,

they may have a better prognosis than


standard glioblastoma (975,1031,1360).
More recent studies suggest that this is
a heterogeneous tumour group, and that
some cases are IDH1- or /OH2-mutant
glioblastomas. The current WHO classi-
fication does not consider glioblastoma
with an oligodendroglioma component
to be a distinct diagnostic entity; with
genetic analysis, it should be possible to
classify such tumours as IDH-wildtype
glioblastoma (in particular the small-cell
variant, given the morphological overlap
with oligodendroglial cells), IDH-mutant
glioblastoma, or I DH-mutant and 1p/19q-
codeleted anaplastic oligodendroglioma.

Gemistocytes and gemistocytic


astrocytic neoplasms
Gemistocytes are cells with copious,
glassy, non-fibrillary cytoplasm that dis-
Fig. 1.27 Granular cell glioblastoma. A Eosinophilic cytoplasm reminiscent of a granular cell tumour of the pituitary, but places the dark, angulated nucleus to
with cytologically more atypical cells. B GFAP immunoreactivity. C The strong nuclear OLIG2 immunoreactivity helps the periphery of the cell. Processes ra-
to establish its glial lineage. D In contrast to most glioblastomas, the granular cell variant often shows immunoreactivity diate from the cytoplasm, but are stubby
for EMA.
and not long-reaching. GFAP staining is
presents either de novo or during pro- patients, the typically strong and exten- largely confined to the periphery of the
gression from a known diffuse glioma. In sive tumoural p53 immunoreactivity, and cell, with the central hyaline organelle-
both settings, the survival time and ge- the presence of IDH1 R132H mutations in rich zone remaining largely unstained.
netic background are similar to those of 15-20% of cases {1183,2394) Perivascular lymphocytes frequently
glioblastoma in general {1946). However, populate gemistocytic regions, but of-
this subtype is distinctive in its high rate Oligodendroglioma components ten avoid other regions in the same neo-
(30-40%) of cerebrospinal fluid dissemi- Occasional glioblastomas contain foci plasm. When present in large numbers,
nation and frequency (-40%) of MYCN that resemble oligodendroglioma. These particularly in a patient known to have a
or MYC gene amplification MYC ampli- areas are variable in size and frequency, pre-existing glioma (e.g. an IDH-mutant
fication is found only in the primitive-ap- and individual pathologists' thresholds gemistocytic astrocytoma), these cells
pearing nodules, and it is likely that such for identifying oligodendroglioma fea- may constitute a lower-grade precursor
alterations drive the primitive-appearing tures vary. Two large studies of malignant lesion within a .secondary IDH-mutant
clonal transformation at least in part, gliomas suggest that necrosis is associ- glioblastoma. Better-differentiated areas
given that a similar phenotype has been ated with a significantly worse prognosis can sometimes be identified radiologi-
observed in N-myc-driven murine fore- in the setting of anaplastic glioma with cally as non-contrast-enhancing periph-
brain tumours {2468). In some cases, ei- both oligodendroglial and astrocytic eral regions, and in whole-brain sections,
ther new 10q losses or expanded regions components (1667,2617); patients whose as WHO grade II to Ill astrocytomas
of 10q loss are also found in the primi- tumours had necrosis had a substantially clearly distinct from foci of glioblastoma
tive neuronal focus {750). Evidence that shorter median overall survival than did {317,2266). lmmunohistochemical stud-
some examples of this subtype are sec- patients whose tumours did not. Such tu- ies have emphasized the low proliferation
ondary glioblastomas includes the his- mours were classified as glioblastomas rate of the neoplastic gemistocyte itself,
tory of a lower-grade precursor in some with an oligodendroglial component, and despite the reported tendency of WHO
grade II or Ill gemistocytic astrocytoma
lesions to progress more rapidly to glio-
blastoma than non-gemistocytic coun-
terparts of the same grade {2706) The
proliferating component presents as a
population of cells with larger hyperchro-
matic nuclei and scant cytoplasm {2706).

Multinucleated giant cells


Large, multinucleated tumour cells are
often considered a hallmark of glio-
Fig. 1.28 Glioblastoma, IDH-wildtype. blastomas and occur with a spectrum
oligodendroglioma-like component. of increasing size and pleomorphism.

34 Diffuse gliomas
Although common, the presence of multi- lesion such as demyelinating disease. 2506) The lipidized cells may be grossly
nucleated giant cells is neither an obliga- Given their lysosomal content, granular enlarged (811 ]. If such a lesion is super-
tory feature nor associated with a more tumour cells may be immunoreactive for ficially located in a young patient, the
aggressive clinical course {315). Despite macrophage markers such CD68, but diagnosis of pleomorphic xanthoastrocy-
their appearance, these cells are con- not for specific markers such as CD163. toma should be considered, particularly
sidered a type of regressive change. If Occasional cells may have peripheral im- if the xanthomatous cells are surrounded
multinucleated giant cells dominate the munopositivity for GFAP, but most cells by basement membranes staining posi-
histopathological picture, the designation are negative {271,793). Some diffuse as- tively for reticulin and accompanied by
of giant cell glioblastoma is justified (see trocytic tumours feature extensive granu- eosinophilic granular bodies (1248).
Giant cell g!ioblastoma, p. 46). lar cell change and have been termed Other lipid-rich lesions have epithelioid
"granular cell astrocytoma" or "granular cytological features (2180). Lobules of
Granular cells and granular cell cell glioblastoma". These lesions have a juxtaposed fully lipidized (i.e not foamy)
astrocytoma/glioblastoma distinct histological appearance and are cells can simulate adipose tissue.
Large cells with a granular, periodic acid- typically characterized by aggressive
Schiff-positive cytoplasm may be scat- glioblastoma-like clinical behaviour {271). Metaplasia and gliosarcoma
tered within glioblastoma. In rare cases, even when the histology otherwise sug- In general, 'metaplasia' refers to the ac-
they dominate and create the impression gests only a WHO grade II or Ill desig- quisition by a differentiated cell of mor-
of a morphologically similar but unrelated nation; one review of 59 reported cases phological features typical of another dif-
granular cell tumour of the pituitary stalk found median survival times of 11 months ferentiated cell type. However, the term is
{569,948). In the cerebral hemispheres, for WHO grade II cases and 9 months for also used to designate aberrant differen-
transitional forms between granular cells WHO grade Ill-IV cases {2283). tiation in neoplasms. In glioblastoma, this
and neoplastic astrocytes can be identi- is exemplified by foci displaying features
fied in some cases, but in others it is dif- Liptdized cells and heavily liptdized of squamous epithelial cells (i.e. epithelial
ficult to identify any conventional astro- glioblastoma whorls with keratin pearls and cytokeratin
cytoma component. Although larger and Cells with foamy cytoplasm are another expression) (320,1734).
more coarsely granular, the tumour cells feature occasionally observed in glio- Occasionally, glioblastomas contain foci
also resemble macrophages. Especially blastoma. The rare lesions in which they with glandular and ribbon-like epithelial
in the context of perivascular chronic predominate have been designated structures {2180). These elements have
inflammation, the tumour cells may be malignant gliomas with heavily lipidized a large oval nucleus, prominent nucleo-
misinterpreted as a macrophage-rich (foamy) tumour cells {1247,1253,2180, lus, and round well-defined cytoplasm.

Glioblastoma, IDH-wildtype 35
for the formation of bone and cartilage,
which predominates in gliosarcoma and
in a variety of childhood CNS neoplasms
(1608).

Secondary structures
The migratory capacity of glioblastoma
cells within the CNS becomes readily
apparent when they reach a border that
constitutes a barrier: tumour cells line up
�:..
Fig. 1.31 IDH-wildtype glioblastoma. GFAP immunohisto-
and accumulate in the subpial zone of Fig. 1.32 Subpial, perivascular, and perineuronal
chemistry typically labels only some cells in glioblastoma, the cortex, in the subependymal region, accumulation of glioblastoma cells. Asterisk indicates
highlighting the tumour cell bodies and processes. and around neurons (so-called satellito- uninvolved subarachnoid space.
sis) and blood vessels. These patterns,
They are also referred to as adenoid called secondary structures (2267), re- One feature of many glioblastomas, es-
glioblastomas. Expression of GFAP in sult from the interaction of glioma cells pecially the small-cell variant, is exten-
these areas may be diminished, and re- with host brain structures, and are highly sive involvement of the cerebral cortex.
placed by expression of epithelial mark- diagnostic of an infiltrating glioma. Sec- Secondary structures and most of the
ers. Small cells with even more epithelial ondary structures may also be present apparently multifocal glioblastomas arise
features and cohesiveness are less com- in other highly infiltrative gliomas, such essentially as a result of the pathways
mon {1250). When there is an extensive as oligodendroglioma (2266,2267). This of migration of glioma cells in the CNS
mesenchymal component, in particular concept also extends to the adaptation (1440). The subependymal region may
a spindle cell sarcoma-like component, of tumour cells to myelinated pathways, also be diffusely infiltrated, especially in
a diagnosis of gliosarcoma should be which often acquire a fusiform, polar the terminal stages of disease.
considered. A mucinous background shape as a result. Identifying neoplas-
and a mesenchymal component (gliosar- tic astrocytes in the perifocal zone of Prol!feration
coma) are not uncommon in metaplastic oedema and at more distant sites can Proliferative activity is usually prominent,
glioblastomas. Adenoid and squamous be challenging for pathologists, in par- with detectable mitoses in nearly every
epithelial metaplasia are more common ticular when dealing with stereotaxic case. Atypical mitoses are frequently
in gliosarcoma than in ordinary glioblas- biopsies (535). Small cell glioblastomas present. However, there is great intertu-
toma (1250,1734). This is similarly true pose a particular problem in this regard. moural and intratumoural variation in mi-
totic activity. The growth fraction as de-
termined by the Ki-67 proliferation index
can thus show great regional variation.
Typical values are 15-20%, but some
tumours have a proliferation index of
> 50% focally. Rare tumours have a low
proliferation index despite other histo-
logical features of malignancy. Tumours
with small, undifferentiated, fusiform cells
often show marked proliferative activ-
ity, in contrast to tumours composed of
neoplastic gemistocytes, which typically
have a lesser degree of proliferation
(2706). Despite the wide range in the pro-
liferation index observed in glioblastoma,
an association between proliferation in-
dex and clinical outcome has not been
demonstrated ( 1715).

Microvascular proliferation and


angiogenesis
The presence of microvascular prolif-
eration is a histopathological hallmark
of glioblastoma. On light microscopy,
microvascular proliferation typically pre-
sents as so-called glomeruloid tufts of
multilayered mitotically active endothelial
cells together with smooth muscle cells I
pericytes {920,1741,2734). Another (less

36 Diffuse gliomas
pericytes {which are negative for CD31 of VEGFA by monoclonal antibodies,
and CD34, positive for SMA, and positive used for the treatment of recurrent glio-
for PDGFRB) (2264). Morphologically blastoma (1998). seems to target pri-
inconspicuous vessels have a Ki-67 pro- marily small, immature vessels and lead
liferation index of 2-4%, whereas prolif- to vascular normalization accompanied
erating tumour vessels have an index of by improved perfusion and oxygenation
> 10% (2702). (2398). Other signalling pathways impor-
Glioblastomas are among the most vascu- tant for glioblastoma angiogenesis in-
larized of all human tumours. Vasculariza- clude angiopoietin/Tie2 receptor signal-
tion occurs through several mechanisms, ling, IL8/IL8R signalling, platelet-derived
including vessel cooption (adoption of growth factor (PDGF) I PDGF receptor
pre-existing vessels by migrating tumour signalling, WNT/beta-catenin signalling,
cells {708,1559)), classic angiogenesis Eph/ephrin signalling, and transforming
(sprouting of capillaries from pre-existing growth factor beta signalling. Pericytes I
vessels by endothelial cell proliferation/ smooth muscle cells and perivascular
migration), and vasculogenesis (homing bone marrow-derived cells (in addition
of bone marrow-derived cells that sup- to endothelial cells) also participate in the
port vessel growth in a paracrine man- vascular remodelling processes typically
,. � ner (6,708,1559)). lntussusception and observed in glioblastoma.
Fig. 1.34 Potential mechanism of palisade formation.
cancer stem cell-derived vasculogen-
A Endothelial injury and the expression of procoagulant
factors result in intravascular thrombosis and increasing esis have also been described (944,1127, Necrosis
perivascular hypoxia (light blue). Tumour cells begin 1989). Hypoxia is a major driving force Tumour necrosis is a fundamental feature
to migrate away, creating a peripherally moving of glioblastoma angiogenesis (6) and of glioblastoma, and its presence is one
wave of palisading cells. B The zone of hypoxia and leads to intracellular stabilization of the of the strongest predictors of aggres-
central necrosis expands. Hypoxic tumour cells of master regulator HIF1A. HIF1A accumu- sive behaviour among diffuse astrocytic
palisades secrete proangiogenic factors (VEGF, IL8). lation leads to transcriptional activation of tumours {315,1031,2079). Presenting on
c Microvascular proliferation in regions adjacent to > 100 hypoxia-regulated genes encod- neuroimaging as a hypodense core within
central hypoxia causes an accelerated outward migration
of tumour cells towards a new vasculature. Illustration © ing proteins that control angiogenesis a contrast-enhancing rim, necrosis con-
2005 Mica Duran. Adapted from Rong Yet al. {2170). (e.g. VEGFA, angiopoietins, erythropoi- stitutes areas of non-viable tumour tissue
etin, and IL8), cellular metabolism (e.g. that can range from extremely small to
common) form is hypertrophic proliferat- carbonic anhydrase and lactate dehydro- accounting for > 80% of the total tumour
ing endothelial cells within medium-sized genase), survival/apoptosis (e.g. BNIP), mass. Higher proportions of necrosis on
vessels. Microvascular proliferation of and migration (e.g. hepatocyte growth MRI have been associated with shorter
the glomeruloid type is most commonly factor receptor, CXCR4, and ACKR3) survival, and the extent of necrosis is also
located in the vicinity of necrosis and is VEGFA seems to be the most important related to the tumour's transcriptional
directionally oriented to it, reflecting the mediator of glioma-associated vascu- profile (904,1969). Grossly, necrosis pre-
response to vasostimulatory factors re- lar functions; it is primarily produced by sents as a yellow or white granular co-
leased from the ischaemic tumour cells. perinecrotic palisading cells as a conse- agulum. Microscopically, glioma cells in
Vascular thrombosis is common and quence of cellular stress such as hypoxia various stages of degeneration are seen
may be apparent to the neurosurgeon and hypoglycaemia {1239,1988,2357). within necrobiotic debris, together with
as so-called black veins. It may play a VEGFA is regulated by transcription fac- faded contours of large, dilated necrotic
role in the pathogenesis of ischaemic tu- tors, oncogenes, tumour suppressor tumour vessels. Occasionally, preserved
mour necrosis (2170). The hyperplastic genes, cytokines, and certain hormones. tumour vessels with a corona of viable tu-
endothelial cells {which are positive for VEG FA induces tumour angiogenesis, in- mour cells can be seen within extensive
CD31 and CD34, negative for SMA, and creases vascular permeability (oedema), areas of necrosis.
positive for VEGFR2) are surrounded by and regulates homing of bone marrow- A second form of necrosis that is a his-
basal lamina and an incomplete layer of derived cells (6). Therapeutic blocking tological hallmark of glioblastoma is the

Fig. 1.35 Extensive coagulative ischaemic necrosis Fig. 1.36 A Longitudinal cut of perinecrotic palisades, presenting as long, serpiginous pattern. B Reticulin stain of a
(right). Note several large thrombosed tumour vessels. perinecrotic garland of proliferated tumour vessels.

Glioblastoma, IDH-wildtype 37
palisading form (historically called the the release of mitochondrial factors glioblastomas. In poorly differentiated
pseudopalisading form), which consists or by death receptor ligation by mem- tumours, the expression of OLIG2 may
of multiple, small, irregularly shaped bers of the tumour necrosis factor fam- be of diagnostic utility, being strongly
band-like or serpiginous foci surround- ily, including TNFSF10/TNFRSF10B and positive far more commonly in astrocy-
ed by radially oriented, densely packed TNFSF6/TNFRSF6 1943,1872). The high- tomas and oligodendrogliomas than in
glioma cells {2170l. The outdated term er levels of apoptosis seen in palisading ependymomas and non-glial tumours
"pseudopalisading" implied that the tu- cells surrounding necrosis may be due 11101,1865). The expression of cytokerat-
mour cells did not truly aggregate around to increased expression or ligation of ins is determined by the class of these
necrosis, but only created this impres- death receptors {264,2485l. TNFSF10 intermediate filaments and antibodies
sion, because they were believed to be induces apoptosis in glioblastoma by used, some of which may indicate cross-
a rim of hypercellular tumour cells that binding to TNFRSF10B and ultimately reactivity with GFAP; keratin positivity is
remained after central degeneration of a activating caspase-8 {943l. Levels of most often detected with the keratin anti-
highly proliferative clone. However. this both TNFRSF6 and TNFSF6 are higher body cocktail AE1/AE3, in contrast to the
is likely not actually the case, given that in astrocytomas than in normal brain lack of positivity for many other keratins
the palisading cells have lower rates of and correlate with tumour grade 12485. [2535l. Nestin is frequently expressed in
proliferation than adjacent glioma cells 2563l Most TNFRSF6 expression in glio- glioblastoma and can be diagnostically
and the central area of smaller palisading blastoma is within palisading cells; physi- useful to distinguish glioblastoma from
structures often consists of a fine fibrillary cal interactions between tumour cells other high-grade gliomas l88l.
network without viable or necrotic glioma expressing TNFRSF6 and TNFSF6 may Glioblastomas with missense TP53 mu-
cells {264,1847,2170l. Palisading cells promote apoptosis. In malignant gliomas, tations show strong and diffuse immu-
are hypoxic and strongly express HIF1A the overall levels of cell death due to apo- nohistochemical overexpression of p53
and other hypoxia-inducible transcription ptosis are low (compared with coagula- {2496). with such overexpression evident
factors l2841,2861l. Downstream hypox- tive necrosis). and apoptotic rates do not in 21-53% of cases {276,1443,2007,
ic upregulation of VEGF. IL8, and other correlate with prognosis l1663,2272l. 2455). This may facilitate discrimina-
proangiogenic factors is responsible for tion between neoplastic astrocytes and
the microvascular proliferation that oc- Inflammation those in reactive gliosis in treated cases
curs immediately adjacent to palisading The number of inflammatory cells in glio- of glioblastoma {268). Detection of WT1
cells. providing a biological link between blastomas varies. Notable perivascular expression, which is sometimes pres-
the histological hallmarks of necrosis and lymphocyte cuffing occurs in a minority ent in both low-grade and high-grade
microvascular hyperplasia in glioblasto- of glioblastomas, most typically in areas gliomas, may serve a similar purpose
ma 11989,2170). The initiating necrogenic with a homogeneous gemistocytic com- [2281}. EGFR expression occurs in about
events have yet to be firmly established, ponent. Inflammatory cells are scant if 40-98% of glioblastomas and correlates
but vascular cooption by neoplastic present at all in small cell glioblastomas. (to some extent) with the presence of
cells has been hypothesized to induce The inflammatory cells have been char- gene amplification [198,678,1023,1443}.
vascular regression through endothelial acterized primarily as CDS+ T lympho- EGFRvlll expression is less common (oc-
angiopoetin-2 expression 1708,1030). It cytes, with CD4+ lymphocytes present curring in 27-33% of cases) [198,678).
has been speculated that microscopic in smaller numbers {227,2184l and with Tumours harbouring H3 K27M mutations
vaso-occlusion and intravascular throm- B lymphocytes detectable in < 10% of (see Diffuse midline glioma, H3 K27M-
bosis may initiate or propagate the devel- cases l2184l. Extensive CDS+ T-cell mutant, p. 57) can be detected using an
opment of hypoxia and necrosis, given infiltrates may be more common in the antibody specific for K27M-mutant H3,
that thrombi are present near regions of tumours of long-term glioblastoma sur- which can be useful in distinguishing
necrosis in nearly all glioblastomas (but vivors 12813}. Microglia and histiocytes these neoplasms from other astrocytic
not in lower-grade, non-necrotic astro- are also present in glioblastomas [1494, tumours {2644}. Some glioblastomas
cytomas) and are often observed within 2162). although lipid-laden histiocytes are (i.e. !DH-mutant glioblastomas) express
or emerging from palisades l264,2529l. uncommon in untreated glioblastomas. R132H-mutant IDH1, but the presence
In this proposed sequence, hypoxia- of R132H-mutant IDH1 expression is
induced cell migration away from cen- lmmunophenotype not compatible with a diagnosis of IDH-
tral hypoxia establishes the palisading Glioblastomas often express GFAP. but wildtype glioblastoma.
structures, which subsequently develop the degree of reactivity differs markedly
into increasingly larger regions of central between cases; for example, gemisto- Cell of origin
necrosis and continue to expand radially cytic areas are frequently strongly posi- The cellular origin of glioblastoma re-
outward 12170). tive, whereas primitive cellular compo- mains unknown. The expression of mark-
nents are often negative. The gliomatous ers of differentiated astrocytes by glio-
Apoptosis component of gliosarcoma may show blastoma cells has long been considered
Apoptosis, the programmed death of expression of GFAP, as opposed to ab- to indicate the dedifferentiation of the
individual cells, is a cell-intrinsic process sent or meagre focal expression in the cells after transformation. More recently,
characterized by nuclear fragmentation sarcomatous component. which may the cellular. biochemical, and genetic
and condensation, with packaging of express alpha-1-antitrypsin, alpha-1-an- heterogeneity that typify glioblastoma, to-
apoptotic bodies within an intact mem- tichymotrypsin, actin, and EMA. S100 gether with the distinct clinical responses
brane. The process is initiated through protein is also typically expressed in of histologically similar tumours, has led

38 Diffuse gliomas
to the hypothesis that the tumours arise Table 1.01 Genetic profile of IDH-wildtype glioblastomas
from the malignant transformation of ei- Gene Change % of tumours References
ther a bipotential precursor cell !1794) or TERT Mutation 72-90% (622,1268,1270)
an even more primordial cell: the neural
EGFR Amplification 35-45% (350, 1823, 1895)
stem cell !1616). This interpretation is
supported by the coincident anatomical CDKN2A Deletion 35-50% (350, 1823, 1895)
position in the subventncular zone of the TP53 Mutation 28-35% (350, 1823, 1895)
brain of dividing cells with stem cell-like PTEN Mutation 25-35% (350, 1823, 1895)
properties and the development of g/io-
NFKB1A Deletion 25% {273A}
blastoma. Moreover, cells with stem cell-
like properties have been isolated from NF1 Mutation 15-18% (350, 1895)
glioblastoma tumours and cell lines and P/C3CA Mutation 5-15% (350, 1288A,1895}
can be produced by the expression of a POGFRA Amplification 13% (350)
set of developmental transcription factors
PTPRD Mutation 12% (350)
in glioblastoma cells !2461). These cells,
called brain tumour stem cells, are only RB1 Mutation 8-12% (350)
a minor subpopulation, but they have the PIK3R1 Mutation 8% (1895)
capacity of self-renewal, express mark-
MDM2 Mutation 5-15% {350,2848A}
ers of developmental regulation, and
are tumorigenic in animals. Therefore, MDM4 Amplification 7% (350)
brain tumour stem cells may be the de- MET Amplification 4% (350)
scendants of neural stem cells that had IDH1 Mutation 0% (1797)
unrepaired DNA damage leading to
IDH2 Mutation 0% (2810)
mutations in cancer genes or that were
subject to an environmental carcinogenic
insult !1112,1756,2230,2365,2650). Ei-
ther of these initiating events could seed Chromosome 7p gain in combination with Various truncations of EGFR can occur
a tumour, given the unlimited growth po- 10q loss is the most frequent genetic al- within the same tumour !732,1906).
tential of the neoplastic cells. teration in gliob/astoma !1079,2072). This
combination is associated with EGFR Receptor tyrosine kinase I Pl3K I PTEN I
Genetic profile amplification. Allelic loss of the chromo- AKTIm TOR pathway
somal region containing the PTEN gene Alterations in receptor tyrosine kinase
Genetics occurs in 75-95% of glioblastomas, signalling pathways occur in nearly 90%
Malignant transformation of neuroepithe- whereas PTEN mutations are present in of glioblastomas !350} In addition to
lial cells is a multistep process driven by 30-44% of cases. Not only is loss of 10q EGFR alterations, alternate routes to me-
the sequential acquisition of genetic and the most frequent genetic alteration, but diate aberrant receptor tyrosine kinase
epigenetic alterations. Of the astrocytic it also typically co-presents with any of signalling include PDGFRA amplification
neoplasms, glioblastoma contains the the other genetic alterations. Another le- (present in 15% of cases), MET ampli-
greatest number of genetic changes. The sion frequently observed in glioblastoma fication (in 5%) and (in rare cases) the
following sections focus on so-called pri- is the combined gain of 19 and 20 !277). fusion protein FGFR1-TACC1 or FGFR3-
mary IDH-wildtype glioblastoma, which TACC3 !2364,2645). Amplification of the
differs in its genetic profile from so-called Epidermal growth factor receptor POGFRA gene is often associated with
secondary /DH-mutant glioblastoma (see EGFR is the most frequently amplified gene truncations, the most common of
G/iob/astoma, /OH-mutant, p. 52). Many gene in glioblastomas !764) and is as- which is an in-frame deletion of exons
of the genetic alterations that are char- sociated with overexpression; 70-90% 8 and 9 (POGFRA!J.8,9) !1869). EGFR,
acteristic of IDH-wildtype glioblastomas of g/ioblastomas with EGFR overexpres- PDGFRA, and MET amplifications and
are also present in the majority of WHO sion show EGFR amplification !198, truncations can occur in different cells
grade II and Ill IDH-wildtype gliomas, 2562). EGFR amplification occurs in in the same tumour, which could pose
suggesting that these various grades approximately 40% of primary glioblas- a problem for targeted therapies !2385).
of IDH-wildtype tumours constitute a tomas !277,627,1823,2780} but rarely in Mutations and amplifications in Pl3K
continuum of disease and further re- secondary glioblastomas !1823,2705). genes (e.q. PIK3CA and PIK3R1) are in-
inforcing the necessity of distinguishing Amplification of the EGFR gene is often frequent in glioblastomas (occurring in
IDH-wildtype from /DH-mutant diffuse associated with gene truncations, most < 10% of cases) !350,1309,1692,1720}.
gliomas j349}. commonly truncation of the gene encod- The PTEN gene is involved in cell prolifer-
ing EGFRv/11 !2753}, which is present in ation, tumour cell migration, and tumour
Cytogenetics and numerical 20-50% of glioblastomas with EGFR am- cell invasion, and is mutated in 15-40%
chromosome alterations plification !198,277,2306,2446}. The pro- of glioblastomas, almost exclusively in
The most common chromosomal imbal- tein is structurally and functionally similar primary glioblastomas !277,2562). The
ances are gain of 7 and loss of 9, 10, and to v-erbB and is constitutively activated mutation pattern suggests that PTEN
13 [http://www. progenetix. net/194403]. in a ligand-independent manner !458). truncation at any site and PTEN missense

Glroblastorna, IDH-wildtype 39
mutations in the region homologous to rare (993). and gene promoter methyla- Genetic parameters (p. 28) for discus-
tensin/auxilin and dual-specificity phos- tion correlated with loss of RB1 expres- sion of the steps necessary to designate
phatases are associated with a malignant sion is more common in secondary glio- a glioblastoma as IDH-wildtype.
phenotype. Mutations in the NF1 gene blastomas (occurring in 43% of cases)
are present in approximately 20% of glio- than in primary glioblastomas (occurring Chromatin-related genes
blastomas (350). in 14% of cases) (1755). Mutations in chromatin-remodelling
genes are common in glioblastomas.
p53/MDM2/p 14ARFpathway TERT promoter mutations Paediatric high-grade gliomas also bear
Alterations in the p53 pathway occur in The promoter region of TERT contains signature mutations directly affecting the
nearly 90% of glioblastomas (3501. TP53 two hotspots for point mutations, with histone gene H3F3A and less commonly
mutations are more often genetic hall- most glioblastomas (-80% in one study) HIST1H3B/C These histone genes have
marks of clinicopathologically defined carrying these mutations (1270) Within two mutation hotspots, in codons K27
secondary glioblastomas and, in almost IDH-wildtype adult diffuse gliomas, TERT and G34, with K27 mutations more often
all cases, are already present in precur- promoter mutations are inversely corre- found in midline, i.e. brain stem, thala-
sor lower-grade or anaplastic astrocyto- lated with TP53 mutations (1801) They mus, and spinal cord tumours (see Dif-
mas (2705). They are significantly less are frequent in /OH1-wildtype glioblasto- fuse midline glioma, H3 K27M-mutant,
common in clinicopathologically defined mas but rare in secondary (/OH1-mutant) p. 57) and G34 alterations in hemispheric
primary glioblastomas (present in -25% glioblastomas and astrocytomas TERT lesions (1263,24441. In paediatric high-
of cases) (1823}. G:C->A:T transitions at mutations are also frequent in oligoden- grade gliomas, H3 mutations are asso-
CpG sites are most common (18231 drogliomas. TERT promoter mutations ciated with mutations in its chaperone
Amplification of MDM2or overexpression lead to the recruitment of multimeric GA- ATRX and are inversely related to IDH
of MDM2 is an alternative mechanism for binding protein (GABP) transcription fac- mutations. ATRX mutations also occur
escaping p53-regulated control of cell tor specifically to the mutant promoter. in adult astrocytomas and glioblastomas
proliferation. Amplification is observed in leading to aberrant TERT expression (277). particularly in those with IDH muta-
< 10% of glioblastomas without TP53 mu- (159A). In /OH1-mutant glioblastomas tions {1160,2304,2792).
tations (2084). Overexpression of MDM2 and astrocytomas, telomere mainte-
has been observed in > 50% of primary nance preferentially uses the alternative Epigenetics, chromatin, and promoter
but only 11% of secondary glioblastomas lengthening of telomeres pathway, which methylation
(199). is activated by mutations in the ATRX The interplay between epigenetic regu-
The CDKN2A locus gives rise to several gene (see Epigenetics, chromatin, and lation and gliomagenesis has several
splice variants encoding distinct proteins promoter methylation). modalities. Epigenetic modifiers can be
(CDKN2A and p14ARF) with tumour- bona tide oncogenes or tumour sup-
suppressing function. The p14ARF pro- IDH mutations pressors that are directly affected by
tein binds directly to MDM2 and inhibits Mutations of the /OH1 and IDH2 genes, gain- or loss-of-function genetic muta-
MDM2-mediated p53 degradation. Loss which encode IDH1 and IDH2 (1895). tions, resulting in the disruption of epige-
of p14ARF expression is frequent in glio- are frequent in diffuse astrocytomas, netic regulatory processes by affecting
blastomas (occurring in 76% of cases), anaplastic astrocytomas, oligodendro- histone modifications, DNA methylation,
and correlates with homozygous deletion gliomas, anaplastic oligodendroglio- and chromatin remodelling (2462) In
or promoter methylation of the CDKN2A mas, oligoastrocytomas. and anaplastic one study, nearly half of the 291 IDH-
gene (1752). oligoastrocytomas (occurring in > 70% wildtype glioblastoma samples sub-
of cases) {2810). These mutations are jected to whole-exome sequencing har-
CDKN2A I CDK4 I retinoblastoma present in nearly all glioblastomas that boured one or more non-synonymous
protein pathway have progressed from astrocytomas (i.e. mutations affecting chromatin organiza-
Alterations in the retinoblastoma pro- clinicopathologically defined secondary tion (277). Even in the absence of direct
tein pathway occur in nearly 80% of all glioblastomas), but they are exceptional genetic alterations, epigenetic modifiers
glioblastomas (350). In glioblastomas, in primary glioblastomas and absent in can modulate gene expression to directly
COKN2A deletion and RB1 altera- pilocytic astrocytomas (118,277,2709, regulate glioma-relevant processes such
tions are mutually exclusive (2594). The 2810). A clinically primary glioblastoma as glioma stem cell programmes, senes-
COKN2A locus encodes the CDK4 in- with an IDH1 mutation may be misclas- cence, genome stability, and invasion
hibitor CDKN2A as well as the alternate sified and may actually constitute an (2443,2461 ).
reading frame protein p14ARF. Inactiva- asymptomatic lower-grade glioma that One of the key functions of chromatin
tion of genes in this pathway is common has progressed and has only become regulation is to maintain inactive portions
in both primary and secondary glioblas- symptomatic as a secondary glioblas- of the genome in repressive chromatin
tomas (200,1752}. The CDK4 gene is am- toma (1797). Thus, IDH1 mutations con- structures with a compact organization
plified in approximately 15% of all high- stitute a reliable molecular signature of a refractory to regulatory activity. Canoni-
grade gliomas (1789,2086). particularly separate group of glioblastomas that may cal repressive states include classic het-
in those without CDKN2A homozygous be synonymous with the secondary type erochromatin marked by H3K27me3, a
deletion (1789}. These homozygous de- {1797). Notably, the presence of IDH1 mark deposited by PRC2 and its catalytic
letions also involve the nearby COKN2B mutation is incompatible with the diag- subunit, EZH2. EZH2 is overexpressed in
gene on 9p (2397} RB1 mutations are nosis of IDH-wildtype glioblastoma. See IDH-wildtype glioblastoma and various

40 Diffuse gliomas
other cancer types, presumably con-
tributing to the silencing of key tumour
Active chromatin Repressive chromatin
suppressor genes (556). Loss of func-
tion of EZH2 can also promote cancer in
a context-dependent manner. Although
loss-of-function mutations of EZH2 in
IDH-wildtype glioblastoma are rare (oc-
curring in < 1% of cases), studies have
strongly implicated an inhibition of its en-
zymatic activity through a mutation of H3
genes in paediatric glioblastoma (277,
2304,2791 ). Apparently, a fine-tuning of
PRC2 activity must be maintained, with O H3K9me3 0 H3K27me3 0 H3K27ac
O H3KJ6me3 0 HJK4me1.2,3
both gain and loss of activity linked to
gliomagenesis. Mutations in ATRX, which Fig. 1.37 Pathways of chromatin organization. DNA methylation, histone modifications, and numerous chromatin
encodes a chromatin remodeller that de- regulators determine the global structure of chromatin and are frequently altered in glioblastoma. Active chromatin (left)
posits H3.3 in pericentromeric and sub- is globally accessible for transcriptional regulation. Repressive chromatin (right) sequesters portions of the genome, is
enriched for characteristic histone modifications, and is refractory to regulatory activity. DNMTs, DNA methyltransferases;
telomeric regions, were observed in 13 of
HDMs, histone demethylases; HMTs, histone methyltransferases; MLL, mixed-lineage leukaemia protein; UTX, histone
291 IDH-wildtype glioblastomas (277) demethylase UTX (also called KDM6A). Adapted from Suva ML et al. (2462).
ATRX mutations are observed in about
60-70% of IDH-mutant gliomas and in
about 30% of paediatric glioblastomas promutagenic alkyl groups from the 06 pilocytic astrocytoma (2124). anaplastic
(1160,2304,2792) position of guanine in DNA, thereby astrocytoma (765). and oligodendroglio-
Within chromatin, both genes and regu- blunting the treatment effects of alkylat- ma (765); primary glioblastoma from sec-
latory elements are associated with ing agents {653,798). MGMT promoter ondary glioblastoma {2580); and IDH-
characteristic chromatin modifications. methylation is common in glioblastoma mutant glioma from IDH-wildtype glioma,
Actively transcribed gene bodies are (present in 40-50% of cases) {277,654, across grades and histology {277,870,
marked by H3K36me3, a mark deposit- 982). with the percentage varying de- 1810,2444) Unsupervised analysis of
ed by the methyltransferase SETD2. Rare pending on the assay used {2051). It is expression profiles can be used to clus-
mutations in SETD2 occur in about 2% of predictive of benefit from therapy with ter glioma into groups that correlate with
IDH-wildtype glioblastoma and are more alkylating agents such as temozolomide histology and grade (886,1812) and may
common in paediatric and IDH-mutant in patients with glioblastoma {982,1577, be a better predictor of patient outcome
gliomas (277,722). Enhancers and pro- 2742). A higher frequency of MGMT pro- {645). A commonly used gene expres-
moters are marked by histone acetylation moter methylation (> 75%) is associated sion-based classification of glioblastoma
and H3K4 methylation. The methylation with glioblastomas that have G-CIMP, defines proneural, neural, classic, and
mark is catalysed by complexes that con- which is characteristic of I DH-mutant mesenchymal subtypes, which correlate
tain the mixed-lineage leukaemia (MLL) gliomas {105,277,1830). Distinct DNA with genomic alterations including TP53
homologues. Missense mutations in methylation subclasses (three of which mutation, EGFR mutation/amplification,
KMT2B, KMT2C, and KMT20 have been are linked with specific mutations) have and NF1 deletion/mutation {2645). How-
detected in some cases (2-3%) of IDH- been identified, which is suggestive of di- ever, individual cells characteristic of dif-
wildtype glioblastoma. Histone deacety- verse developmental and pathogenetic/ ferent subtypes can be found within the
lases (HDAC2 and HDAC9) and a range pathoepigenetic origins {277,2444) Mu- same glioblastoma {1906)
of histone demethylases (e.g. KDM40, tations in IDH1 have been causally linked
KDMSA/B/C, and KDM6AIB) are also in- with G-CIMP and longer survival (1810, Genotype-phenotype correlations
frequently mutated in IDH-wildtype glio- 2589). Two methylation subtypes are re- Most cases (> 90%) are glioblastomas
blastoma, broadly affecting chromatin lated to hotspot mutations in H3 genes that develop rapidly and de novo (so-
activity. Both histone and DNA demethyl- (H3 K27 and H3 G34), which are most called primary glioblastoma), with no
ases are inhibited by the IDH mutations, prevalent in paediatric glioblastomas known less-malignant precursor lesion,
suggesting that different classes of glio- (2105,2444) H3F3A G34 is associated often in middle-aged or elderly patients
mas use different modalities to inactivate with a CpG hypomethylator phenotype (as opposed to the so-called secondary
key epigenetic regulators (2443). {2444). Cancer-relevant pathways (e.g. glioblastomas that have IDH1 mutations;
In addition to chromatin regulation, epi- the WNT pathway) that contribute to see Glioblastoma, /DH-mutant, p. 52).
genetic gene silencing by DNA meth- malignant behaviour and resistance to Glioblastomas in which multinucleated
ylation of their promoters is a common therapy are frequently deregulated by giant cells constitute > 5% of the tumour
mechanism of inactivating genes or non- aberrant promoter methylation of genes are associated with frequent TP53 muta-
coding RNAs with tumour suppressing encoding inhibitory factors (914,1425) tions but infrequent EGFR amplification
functions (2010). The MGMT gene is the (1931). whereas small cell glioblastomas
most commonly methylated gene across Expression proftles often have EGFR amplification (318). In
tumour types (462) and encodes a DNA Gene expression patterns can be younger patients, high-grade gliomas
repair protein. It specifically removes used to distinguish glioblastoma from (including glioblastomas) of the brain

Glioblastoma, IDH-wildtype 41
Paediatric high-grade diffuse Clinicopathological aspects are found exclusively in diffuse midline
astrocytic tumours The annual incidence of paediatric gliomas, whereas H3.3 G34R or (rarely)
Paediatric high-grade diffuse astrocytic glioblastoma (defined by patient age G34V mutations are found exclusively
tumours (WHO grade Ill/IV) should be < 20 years at diagnosis) is 0.14 cases in tumours of the cerebral hemispheres
considered, for therapeutic purposes, per 100 000 population; lower than that {2443,2792}. which are more frequently
as a single category encompassing of adult glioblastoma, which is approxi- seen in teenagers and young adults. In
both paediatric glioblastoma and pae- mately 4 per 100 000 {282,18631. Most contrast, hemispheric glioblastomas in
diatric anaplastic astrocytoma. This ap- diffuse WHO grade II astrocytomas pre- infants harbour NTRK fusions in approx-
proach is supported by our understand- senting in adults eventually progress to imately 40% of cases, and histone mu-
ing of these childhood tumours' similar high-grade astrocytomas (anaplastic tations have not been reported. Other
genetic profiles and clinical courses. astrocytoma or glioblastoma). In con- commonly altered genes in hemispheric
Although the histopathologies of paedi- trast, nearly all paediatric high-grade high-grade astrocytomas of childhood
atric high-grade astrocytomas overlap diffuse astrocytic tumours arise de nova, are TP53 (present in 30-50% of cas-
with those of their adult counterparts, rarely deriving from a WHO grade II es). ATRX (in -25%), SETD2 (in -15%),
the two groups have distinct genetic al- astrocytoma. CDKN2A (deletion; in -30%), and
terations {2304,2444,2792}. Unlike adult PDGFRA (amplification and/or mutation;
glioblastomas, paediatric high-grade Genetic aspects in -30%) {27921. In contrast, IDH muta-
diffuse astrocytic tumours frequently Recurrent mutations in paediatric tions, TERT promoter mutations, and
arise in the midline of the neuraxis, high-grade diffuse astrocytic tumours EGFR mutations/amplifications are rare.
commonly in the pons (as diffuse pon- involve genes coding for proteins in- Several hereditary tumour syndromes
tine glioma) or the thalamus and rarely volved in chromatin and transcription predispose paediatric patients to diffuse
in the spinal cord or cerebellum. These regulation, or the receptor tyrosine ki- astrocytic tumours, including Li-Frau-
diffuse midline gliomas share genetic al- nase I RAS I MAPK and/or retinoblas- meni syndrome (associated with TP53),
terations and are discussed separately toma protein I p53 pathways. Many of neurofibromatosis type 1, and constitu-
(see Diffuse midline glioma, H3 K27M- these genes are also mutated in the tional mismatch repair deficiency.
mutant, p. 57). Distinct from these tu- equivalent adult tumours, but some al-
mours and from their adult counterparts terations are particularly associated
are the cerebral hemispheric high-grade with paediatric or adult disease. H3
diffuse astrocytic tumours of childhood. variant (H3.1/H3.3) K27M mutations

Table 1.02 Frequent genetic alterations in diffuse


astrocytic tumours: paediatric versus adult disease stem and thalamus often harbour muta- neurofibromatosis type 1, as well as less
Gene class Paediatric tions in histone genes, particularly K27M common syndromes such as Oiiier-type
mutations in H3.3 and H3.1 {1263,2444) multiple enchondromatosis {7341. In rare
H3F3AK27M'
Histone protein (see Diffuse midline glioma, H3 K27M- cases, patients with L-2-hydroxyglutar-
H3F3A G34RN2
variants mutant, p. 57). Rare glioblastomas or low- ic aciduria have developed malignant
HIST1H38 K27M
er-grade astrocytic neoplasms have been brain tumours, including glioblastoma
Histone modification SETO:?
reported with focal losses of SMARCB1 {9281. Five independent genome-wide
Growth factor ACVR13 expression, often with monosomy 22 and association studies have identified eight
BRAFV600E2 epithelioid or rhabdoid cytology {1013, specific heritable risk variants in seven
Cell signalling POGFRA mutation/ 1299}; these can be called SMARCB1- genes (TERT, EGFR, CCDC26, CDKN2B,
amplification deficient glioblastoma. Epithelioid glio- PHLDB1, TP53, and RTEL1) associated
Gene class Adult blastomas often have BRAF V600E mu- with an increased risk of glioma {18611.
Cell signalling EGFR mutation/amplification tations. Overall, glioblastomas in children
have a genetic profile distinct from that Prognosis and predictive factors
Telomere
maintenance
TERT promoter mutation of glioblastomas in adult patients; the dif- Glioblastoma is an almost invariably fatal
ferences between adult- and paediatric- disease, with most patients dying within
Intermediate
IDH1 R132H type glioblastomas are discussed sepa- 15-18 months after diagnosis, and « 5%
metabolism
rately (see the box Paediatric high-grade of patients alive after 5 years {5961. Even
PTEN mutation I biallelic diffuse astrocytic tumours). in clinical trials with somewhat more fa-
Phosphatase
deletion
vourable patient selection, the 5-year
Specific chromo-
Gain of chromosome 7 and Genetic susceptibility survival rates do not exceed 10% {2442}.
some copy number Glioblastomas sometimes occur in more Younger age (< 50 years) and complete
loss of chromosome 10
alteration than one member of a family. This is macroscopic tumour resection are asso-
'In tumours of midline (mutation in -80% of diffuse most often the case within the setting ciated with longer survival; on a molecu-
intrinsic pontine gliomas), thalamus, and spinal cord. of inherited tumour syndromes such as lar level, tumours associated with longer
2ln tumours of cerebral hemispheres.
Turcot syndrome (in particular Turcot survival often exhibit two favourable
3
ln diffuse intrinsic pontine gliomas only (-20%).
'Both alternative lengthening of telomeres and
syndrome type 1, which is character- molecular aberrations: MGMT promoter
telomerase re-expression reported. ized by non-polyposis colorectal car- methylation {2088,24421 and/or IDH mu-
cinoma), Li-Fraumeni syndrome, and tation {950).

42 Diffuse gliomas
Table 1.03 IDH-wildtype glioblastoma: genetic susceptibility because of partial blood-brain barrier
syndrome Gene Chromosome OMIM preservation and high interstitial pressure
17p13.1 151623 in the tumour tissue; (2) invasive proper-
Li-Fraumeni* TP53
ties of glioblastoma cells that enable
L-2-hydroxyglutaric aciduria L2HGDH 14q21.3 236792
them to spread distantly within the CNS
Turco! MLH1, PMS2, MSH2, MSH6 3p21.3, 7p22, 2p22-p21, 2p16 276300 and remain behind an intact blood-brain
Neurofibromatosis type 1 NF1 7q12 162200 barrier; (3) retention of DNA repair ma-
3p21-22 166000 chinery that abrogates the effectiveness
Oiiier/Maffucci PTHR1
of chemotherapy and radiotherapy; (4)
*Glioblastomas associated with Li-Fraumeni syndrome are IDH-mutant.
intratumoural heterogeneity and genomic
instability resulting in clonal populations
Age that these uncertainties will be clarified of resistant cells, making it necessary to
A prospective series {2728) and numer- by molecular-based classifications of identify the driving events on an ongoing
ous clinical trials {439,837,838,2442} such tumours. basis; (5) the presence of a population
have shown that younger patients with of tumour-initiating or stem cell-like cells
glioblastoma (those aged < 50 years Genetic alterations and biomarkers that may harbour resistance mechanisms
at diagnosis) have a substantially bet- MGMT promoter methylation is a strong distinct from those of the majority of bulk
ter prognosis. In one large population- predictive marker for the efficacy of and tumour cells and that may contribute to
based study, patient age was a signifi- response to alkylating and methylating cellular heterogeneity; and (6) secondary
cant prognostic factor; the correlation chemotherapy agents in glioblastoma oncogenic changes induced by tumour
persisted through all of the age groups in {982,2729,2741,2742,2480}. More than progression.
a linear manner {1823). Because elderly 90% of long-term surviving patients with Besides MGMT promoter methylation,
patients have an inferior prognosis over- glioblastoma have a methylated MGMT there are no predictive biomarkers {2743)
all, distinct and de-escalated treatment promoter {2442). versus only 35% of the that facilitate the tailoring of primary {982,
strategies favouring a short therapeutic general population of patients with glio- 2741,2742) or secondary {2480,2729)
course and the best quality of life pos- blastoma {2441). IDH1 and IOH2 muta- therapies, and MGMT promoter methyla-
sible are commonly proposed to patients tions are positive prognostic factors and tion is only identified in less than half of
aged > 65-70 years. However, there is are closely associated with secondary all patients using current assays {2441,
no prospectively validated age cut-off glioblastoma {2810) (see Glioblastoma, 2743). Cellular responses to DNA dam-
for clinical decisions. Although the fre- /OH-mutant, p. 52). There is no consist- age involve distinct DNA repair path-
quency of MGMT promoter methylation ent correlation between EGFR amplifica- ways, such as mismatch repair and base
(see Genetic alterations and biomarkers) tion and survival, regardless of patient excision repair. Mismatch repair is criti-
is stable across the various age groups, age at clinical manifestation {1823). Allel- cal for mediating the cytotoxic effect of
recent research has identified molecular ic loss of 10q was associated with shorter 6-0-methylguanine. The mismatch repair
profiles associated with paediatric glio- survival in one study {1823}; however, the pathway consists of several proteins (i.e.
blastoma, younger patient age, and sec- presence of PTEN mutations is not clear- MLH1, PMS2, MSH2, MSH3, and MSH6)
ondary glioblastoma, and these profiles ly associated with prognosis {1823,2289, and corrects errors in DNA base pairing
may suggest novel therapeutic targets 2374,2728). that occur during DNA replication. De-
{2444,2728). fects in this system arise in the setting
Mechanisms of treatment response and of alkylating chemotherapy and may re-
Histopathology resistance sult in resistance to such therapy {342,
In general, histopathological features do Glioblastoma is highly resistant to thera- 2826). potentially by causing tolerance of
not confer significant prognostic informa- py, with only modest survival increases the mispairing of 6-0-methylguanine with
tion, but in some studies, giant cell glio- achieved in a minority of patients, even thymine {2244). but also through chemo-
blastoma (see Giant cell glioblastoma, after aggressive surgical resection, ex- therapy-induced mutagenesis {11671. Te-
p. 46) has been noted to have a some- ternal beam radiation therapy, and maxi- mozolomide treatment of WHO grade II
what better prognosis than ordinary glio- mum tolerated doses of chemotherapy gliomas induces a hypermutational state
blastoma {1356,18531. with agents such as temozolomide or ni- and causes driver mutations in the ret-
Greater extent of necrosis is associated trosoureas, including in conjunction with inoblastoma protein and AKT/mTOR
with shorter survival {126,315,1031) The blood stem cell transplantation or use pathways {2627). In one small series,
presence of necrosis in a high-grade of transgenic haematopoietic stem cells 5 of 6 hypermutated tumours showed
oligoastrocytic neoplasm confers a pro- {8}. Over the past several decades, hun- de novo mutations in mismatch repair
gnosis consistent with a WHO grade IV dreds of clinical trials have achieved only genes {2627).
designation {1667). but whether the limited therapeutic success; responders Molecular abnormalities in glioblastoma
presence of oligodendroglial features have been very rare and trial designs also provide specific mechanisms of
in a glioblastoma has prognostic value have limited the lessons learned from resistance and susceptibility to therapy.
relative to other glioblastomas remains the failed approaches, including those Some of the signature genetic alterations
uncertain, with some studies showing a of targeted therapies. The therapeutic in glioblastoma (e.g. mutations in PTEN,
positive association {85,1667} and others resistance is due to a variety of factors, TP53, EGFR, NF1, RB1, PIK3CA, PIK3R1,
having negative results {983}. It is likely including: (1) uncertain drug delivery and IDH1) may be related to resistance

Glioblastoma, IDH-wildtype 43
100 may be seen (in particular, decreases
in enhancement and oedema). These
90
changes reflect the effect of the agents
80 Overall Wald test: p <0.0001 (df=3) on the tumour vasculature and may ben-
"-�>
70
efit patients symptomatically, but antian-
giogenic agents have not clearly been
.
;;J
C/1
60
shown to improve survival (141,1543). By
...e
c 50
Meth, TMZ/ RT normalizing the tumour vasculature to
-�. 40
some extent, antiangiogenic agents may
also facilitate perfusion and oxidation of
=:..
0 30 tumours.
.t
Cellular immunotherapy I tumour vaccine
20
approaches were initiated decades ago,
10 but to date have had little effect on the
L------:=1.-----==:..._ _,_ months survival of patients with glioblastoma.
4 12 16 20 24 28 32 36 40 However, recent vaccine/immunotherapy
0 I I umbtr of patltnts at risk: treatment of glioblastoma may be more
54
53
54
60
2S
44
9
lS
0
s
- Unmeth,RT 1Jone
- Unmoth, TMZ/ RT
promising; for example, against targets
45
40
46
46
33
35
15
2$ 18 14 10
Meth, RT �lone such as cytomegalovirus, the poliovirus
0 - Meth, TMZ/ RT
receptor, and EGFRvlll. It is also possible
Fig. 1.38 MGMT promoter methylation (meth) and progression-free survival in glioblastoma patients randomized to that combining vaccine approaches with
treatment with radiotherapy (RT} alone or radiotherapy plus temozolomide (TMZ). Reprinted from Hegi ME et al. (982). the checkpoint inhibitors discussed be-
low could increase efficacy. The glioblas-
{350,1895). Inactivation of the p53 path- analyses encompassing the full genome, toma microenvironment shows profound
way leads to defects in apoptosis and epigenome, and transcriptome, or even alterations compared with the normal
cell cycle arrest. Mutations of the retino- the proteome and metabolome, possibly brain microenvironment. Blood-brain
blastoma protein pathway in glioblasto- at the single-cell level (1906). will be nec- barrier leakage, hypoxia, acidosis, accu-
mas result in failure of appropriate cell essary to identify driving changes that mulation of soluble mediators, and attrac-
cycle arrest. Point mutations of the RAS are accessible to targeted therapeutic tion of stromal cells from the peripheral
genes in glioblastoma are rare, but the approaches. circulation markedly alter the immuno-
RAS pathway is secondarily activated Recently, a population of glioma-initi- biology of gliomas (415} One compel-
through insulin-like growth factor recep- ating, potentially neural stem cell-like ling explanation is that a pool of particu-
tor, EGFR, and PDGF receptor signalling. glioma cells has been identified in glio- larly resistant and slow-proliferating cells
Downstream events such as silencing blastoma (122,780,2365) These cells called glioma-initiating cells, perhaps lo-
of the NF1 tumour suppressor gene can are highly tumorigenic in immunosup- cated in a perivascular or hypoxic niche,
also activate the RAS pathway, causing pressed mice, inducing intracranial tu- may act as a source for repopulation of
uncontrolled cellular proliferation. Simi- mours with a much smaller cellular inocu- the bulk tumour, thus complicating im-
larly, the Pl3K pathway can be activated lum than do non-stem cell-like glioma munotherapeutic approaches. Glioma-
by abnormal IGF1, epidermal growth fac- cells from glioblastomas. The intracranial initiating cells have been investigated
tor, or PDGF signalling, or downstream tumours induced by these stem cell-like with respect to their immunosuppressive
by abnormalities in the PTEN gene {891, glioma cells have the morphological hall- potential and as a potential target for im-
2432). These redundant signalling path- marks of glioblastoma. They respond to munotherapy, given that they have anti-
way abnormalities suggest that a sin- treatment with bevacizumab (a neutral- genic properties that are different from
gle, specific, small-molecule signalling- izing antibody to VEGF), and the same those of bulk tumour cells {423} Mo-
pathway inhibitor could only be effective humanized VEGF-neutralizing antibody lecular mechanisms of immunosuppres-
in treating glioblastoma if it targeted a has achieved a 60-65% response rate in sion involve transforming growth factor
downstream and driving factor. Deter- a phase 11 trial in recurrent glioblastomas beta, STAT3, PDL1 (which is expressed
mining such an effect may be difficult, (2671 }. One of the mechanisms of ra- by many cancer cells and binds to its
without orthotopic patient-derived mod- diation resistance, activation of the DNA receptor PD1 to suppress T-cell prolif-
els or clinical data available to develop checkpoint response, may exist preferen- eration and IL2 production (385}), and
tightly constructed controls. For example, tially in the stem cell-like glioma cell pop- CD276 {1469) In addition, CTLA4, an
the assumption that testing individual ulation (122}. BMP4 causes a significant immunoglobulin similar to the costimula-
glioblastoma biopsies for EGFRvlll and reduction of stem-like precursor cells of tory protein CD28 that is expressed pref-
PTEN (1634} could potentially enable the human glioblastoma and abolishes their erentially on helper T cells, transduces
identification of patients responsive to the tumour-initiating capacities in vivo. inhibitor signals upon engagement of
EGFR kinase inhibitors is complicated Given the marked vascularity of glio- the costimulatory receptors COBO and
by the fact that this signature character- blastomas, antiangiogenic agents such CD86, which are expressed on antigen-
izes a poor prognostic subgroup that as bevacizumab and cediranib are now presenting cells (2681). The kynurenine
currently lacks therapeutic implication widely used for the treatment of these pathway may also be involved in altering
{2614) This suggests that more intensive tumours. On scans, notable responses the immunobiology within gliomas; for

44 Diffuse gliomas
•*
1. Cross-Resistance 2. Tumor Heterogeneity

� p
�...... -- Ionizing Radiation

.v
� :


_ .... .,, Gefitinib
RTK

J-- y-irradiation/Gefitinib
I Y= EGFR Y= 6EGFR Y= PDGFR I
Tumor Cells Y= MET Y= IGFR1

+ a-sense Bcl-xl,

...
3. Cancer Stem Cell Resistance 4. Tumor Microenvironment

Fig. 1.39 Malignant glioma and therapeutic resistance. Glioblastomas are adept at evading inhibition of EGFR receptor function through several possible mechanisms. (1) Brain
tumour cells that are intractable to DNA damage-induced apoptosis may also tolerate apoptotic cues driven by TKl-mediated inhibition of EGFR. Combinatorial therapy using
inhibitors of anti-apoptotic activity may overcome this cross-resistance. (2) lntratumoural diversity within glioblastomas may drive resistance to single agent-based anti-EGFR
therapy due to: RTK co-activation, PTEN deletion/mutations, and tumour cell-tumour cell interactions via secreted molecules. PTEN* denotes mutation. (3) Efflux of EGFR TKls
and increased genetic stability may lead to the maintenance of cancer stem cell populations and tumour relapse. (4) Enhanced immunosuppression mediated by circulating growth
factors, cytokines, and suppressor T cells can antagonize the systemic immune responses generated by anti-EGFR immunotherapies. Additionally, circulating IL6 in the tumour
microenvironment can facilitate resistance intracellularly via activation of the JAK/STAT3/Bcl-xl pathway. Reprinted from Taylor TE et al. {2526A}.

example, tryptophan is catabolized by


key rate-limiting dioxygenases, chiefly in-
doleamine 2,3-dioxygenase, creating an
immunosuppressive milieu due to deple-
tion of tryptophan and accumulation of
immunosuppressive tryptophan catabo-
lites such as kynurenine {11 l.

Glioblastoma, IOH-wildtype 45
,

Giant cell glioblastoma Ohgaki H. often located subcortically in the temporal


Kleihues P. and parietal lobes. On CT and MRI, they
Plate K.H. can mimic a metastasis.
Nakazato Y.
Bigner D.D. Macroscopy
Due to its high connective tissue content,
giant cell glioblastoma may have the gross
appearance of a firm, well-circumscribed
Definition mass, which can be mistaken for a me-
A rare histological variant of IDH-wildtype tastasis or (when attached to the dura) a
glioblastoma, histologically characterized meningioma. Lesions less rich in connec-
by bizarre, multinucleated giant cells and tive tissue may have features more typical
an occasionally abundant reticulin network. of glioblastoma (1533)
Despite the high degree of anaplasia, gi-
ant cell glioblastoma is often more circum- Microscopy
scribed and has a somewhat better pro- Giant cell glioblastoma is histologically
gnosis than ordinary glioblastoma (1356, characterized by numerous multinucleated
1853). The genetic profile differs from that giant cells, small fusiform syncytial cells,
of IDH-wildtype glioblastoma in the high Fig. 1.41 Giant cell glioblastoma. The multinucleated and a reticulin network (1584). The giant
frequency of TP53 mutations (1932) and in giant cells are easily recognizable in the smear cells are often extremely bizarre, can be
AURKB expression (2533). whereas EGFR preparation {1956}. as large as 0.5 mm in diameter, and can
amplification is rare. contain anywhere from a few nuclei to> 20.
cell glioblastoma (n = 592) was 54.5 years Giant cells are often angulated, may con-
ICD-0 code 9441/3 (1853). The male-to-female ratio is 1.1-1.5:1 tain prominent nucleoli, and on occasion
(1356,1533,1853). contain cytoplasmic inclusions. Both pali-
Grading sading and large ischaemic necroses are
Giant cell glioblastoma corresponds histo- Localization observed. Atypical mitoses are frequent,
logically to WHO grade IV. The localization of giant cell glioblastoma is but the overall proliferation rate is similar
similar to that of IDH-wildtype glioblastoma to that of ordinary glioblastomas. A typical,
Epidemiology (1356,1853). (although variable) feature is the perivas-
Giant cell glioblastomas account for < 1% cular accumulation of tumour cells with the
of all glioblastomas (1853). but may be Clinical features formation of a pseudorosette-like pattern
more common in paediatric populations Giant cell glioblastomas develop de novo (1533). Occasionally, perivascular lympho-
(1356). Giant cell glioblastoma develops in after a short preoperative history and with- cyte cuffing is observed. Microvascular
younger patients than does ordinary glio- out clinical or radiological evidence of a proliferation is not common.
blastoma, with an age distribution covering less-malignant precursor lesion. The symp-
a wider range, including children (1226, toms are similar to those of IDH-wildtype lmmunophenotype
1356,1658,1931). In a study of 16 430 glio- glioblastoma. GFAP expression is consistently present,
blastomas in the SEER database of the but the level of expression is highly vari-
United States National Cancer Institute, Imaging able. TP53 mutation is present in > 80%
the mean age of patients with giant cell Giant cell glioblastomas are distinctive be- of all cases, and these tumours typically
glioblastoma (n = 171) was 51 years, sig- cause of their circumscription. They are show marked nuclear accumulation of p53
nificantly younger than that of patients with
glioblastoma (62 years) (1356). Similarly, in
a SEER-based study of 69 935 glioblasto-
mas, the mean age of patients with giant

Fig. 1.40 Giant cell glioblastoma. The cut surface shows


a multinodular lesion with necrosis and haemosiderin
deposits.
-------
Fig. 1.42 Multinucleated cells in giant cell glioblastoma. The number of nuclei ranges from a few to > 20.

46 Diffuse gliomas
protein (1233,19311. Neuronal markers are
virtually all negative, unlike in pleomorphic
xanthoastrocytoma (1597f.

Genetic profile
Giant cell glioblastomas do not carry IDH
mutations and are therefore considered
variants of IDH-wildtype glioblastoma.
They are further characterized by frequent
mutations in TP53 (occurring in 75-90%
of cases) and PTEN (in 33%), but typi-
cally lack EGFR amplification/overexpres-
sion and homozygous CDKN2A deletion
(1596,1658,1931,1932}.
These attributes indicate that giant cell
glioblastoma has a hybrid profile, shar-
ing with IDH-wildtype glioblastoma a
short clinical history, the absence of a
less-malignant precursor lesion, and fre-
quent PTEN mutations. Like !DH-mutant
glioblastoma, which typically develops
through progression from a lower-grade
astrocytoma, giant cell glioblastoma has a
high frequency of TP53 mutations (1932}.
The level of AURKB expression at the
mRNA and protein levels is significantly
higher in giant cell glioblastomas than in is somewhat better than that of ordinary with ordinary glioblastoma (3.4%) (1356}
ordinary glioblastomas, and ectopic over- glioblastoma (323,1060,1356,1820,1853, Similarly, a study of 67 509 glioblastomas
expression of aurora kinase B induces a 2347). A study of 16 430 glioblastomas in in the USA National Cancer Data Base
significant increase in the proportion of the SEER database found that the medi- found that patients with giant cell glio-
multinucleated giant cells in TP53-mu- an survival time of patients with giant cell blastoma (n = 592) had a median overall
tant U373-MG (but not TP53-wildtype glioblastoma was 11 months, significantly survival of 13.5 months, versus 9.8 and
U87-MG) malignant glioma cells (2533). longer than that of patients with ordinary 8.8 months for patients with ordinary glio-
glioblastoma (8 months) (1356). The over- blastoma and gliosarcoma, respectively
Prognosis and predictive factors all 5-year survival rate among patients {1853).
Most giant cell glioblastomas have a with giant cell glioblastoma was > 10%,
poor prognosis, but the clinical outcome significantly higher than that of patients

Table 1.04 Genetic profiles of histologically defined glioblastoma (GBM) variants; adapted from Oh JE et al. {1819)
PrimaryGBM Secondary GBM
Gliosarcoma Giant cell GBM
(IDH-wildtype) (IDH-mutant)
Age at GBM diagnosis 59 years 56 years 44 years
-------- 43 years
Male-to-female ratio 1.4 1.4 1.6 1.0
Length of clinical history 3.9 months 3.0 months 1.6 months 15.2 months
IDH1/2 mutation 0% 0% 5% 100%
PTEN mutation 24% 41% 33% 5%
ATRX expression loss 0% 0% 19% 100%
TERT mutation 72% 83% 25% 26%
TP53 mutation 23% 25% 84% 74%
Loss of 19q 4% 18% 42% 32%
EGFR amplification 42% 5% 6% 4%
Light blue, typical for IDH-wildtype GBMs; yellow, typical for IDH-mutant GBMs. Giant cell GBM shares characteristics with both GBM types.

Giant cell glioblastoma 47


,

Gliosarcoma Burger P.C. Spread


Giangaspero F. Like in ordinary glioblastoma, the infil-
Ohgaki H. trative growth of this variant is generally
Biernat W. restricted to the brain parenchyma. In-
vasion of the subarachnoid space is un-
common. Haematogenous spread with
extracranial metastases is rare, but has
been reported {148).
Definition in patients aged 40-60 years (mean age:
A variant of IDH-wildtype glioblastoma, 52 years). Rare cases occur in children Macroscopy
characterized by a biphasic tissue pat- {1228l. Males are more frequently affect- Due to its high connective tissue con-
tern with alternating areas displaying glial ed, with a male-to-female ratio of 1.8:1. tent, gliosarcoma has the gross appear-
and mesenchymal differentiation. ance of a firm, well-circumscribed mass,
Gliosarcoma principally affects adults. Localization which can be mistaken for a metastasis
The entity was originally defined as a Gliosarcomas are usually located in the or (when attached to the dura) a menin-
glioblastoma in which the sarcomatous cerebral hemispheres, involving the tem- gioma. Lesions less rich in connective
component was the consequence of poral, frontal, parietal, and occipital lobes tissue may have features more typical of
malignant transformation of proliferating in decreasing order of frequency. Rarely, glioblastoma.
tumour vessels {682). but there is cy- gliosarcoma occurs in the posterior fossa
togenetic and molecular evidence for a {1774,1793). lateral ventricles {2243). or Microscopy
monoclonal origin of both the glial and spinal cord {370l. Like standard glioblas- A mixture of gliomatous and sarcoma-
mesenchymal components. Although tomas, some gliosarcomas are multifocal tous tissues confers to gliosarcoma a
usually associated with classic (astro- {1881). striking biphasic tissue pattern. The gli-
cytic) glioblastoma, gliosarcomas can al portion is astrocytic and anaplastic,
also arise in ependymoma (ependymo- Clinical features mostly showing the typical features of a
sarcoma) {2153l and oligodendroglioma The clinical profile of gliosarcoma is the glioblastoma. Epithelial differentiation,
(oligosarcoma) {2152l. Gliosarcomas can same as that of IDH-wildtype glioblas- manifesting as carcinomatous features
present de nova or appear during the toma, with symptoms of short duration {1871} with gland-like or adenoid forma-
post-treatment phase of glioblastoma. that reflect the location of the tumour and tions and squamous metaplasia {1250,
The prognosis is poor. Occasional cases increased intracranial pressure. Gliosar- 1734}, occurs in the glial portions of
disseminate systemically and/or pen- comas can appear at the time of initial some cases. By definition, the sarcoma-
etrate the skull. presentation of glioblastoma, but many tous component shows signs of malig-
occur during the post-treatment course nant transformation (e.g. nuclear atypia,
ICD-0 code 9442/3 of the disease {939} mitotic activity, and necrosis) and often
demonstrates the pattern of a spindle
Grading Imaging cell sarcoma, with densely packed long
Gliosarcoma corresponds histologically In cases with a predominant sarcomatous bundles of spindle cells surrounded indi-
to WHO grade IV component, the tumour presents as a vidually by reticulin fibres. Occasionally,
well-demarcated hyperdense mass with this component has considerably more
Epidemiology homogeneous contrast enhancement, pleomorphism {779,1774l. A subset of
Gliosarcomas account for approximately and may mimic meningioma {2166,2466l. cases show additional lines of mesen-
2% of all glioblastomas {779). although In cases with a predominant gliomatous chymal differentiation, such as the forma-
higher frequencies (as high as 8%) have component, the radiological features are tion of cartilage {119). bone {1608). os-
also been reported {1713,2242l. The age similar to those of glioblastomas. teoid-chondroid tissue {520,973,2487).
distribution is similar to that of glioblasto- smooth and striated muscle {1262}, and
ma overall, with preferential manifestation even lipomatous features {755). Primitive

Fig. 1.44 Gliosarcoma. Spindle cells, often mitotically


active, are a typical feature in smear preparations.
Reprinted from Burger PC (312).

48 Diffuse gliomas
neuronal components occur rarely a sarcomatous phenotype (1179,1633) mutations, but infrequent EGFR ampli-
(2349). Distinction of the two components In a study using FISH, 2 of 3 gliosarco- fication {7,2095}, suggesting that they
is facilitated by combined histochemical mas showed identical numerical aberra- have a distinct profile, similar to that of
and immunohistochemical staining. Col- tions of chromosomes 10 and 17 in the IDH-wildtype glioblastoma (except for
lagen deposition in the mesenchymal glial and mesenchymal components, the infrequent EGFR amplification) Com-
part is well demonstrated by a trichrome whereas in the third case, trisomy X was parative genomic hybridization in 20
stain. Similarly, reticulin staining shows restricted to the chondrosarcomatous gliosarcomas found that several chromo-
abundant connective tissue fibres in the element (1913) Similar cytogenetic pat- somal imbalances were common: gains
mesenchymal (but not glial) component. terns were observed in both glial and on chromosomes 7 (present in 75% of
The demonstration of a clearly malignant mesenchymal components in another cases), X (20%), 9q (15%), and 20q (15%)
mesenchymal component negative for study, using FISH, comparative genomic and losses on chromosomes 10 (35%),
GFAP is important to distinguish true glio- hybridization, microsatellite allelic imbal- 9p (35%), and 13q (15%) l7I [http://www.
sarcomas from glioblastomas with florid ance analysis, and cytogenetic analysis progenetix.net/progenetix/194423]. Simi-
fibroblastic proliferation (desmoplasia) (232} These results suggested that both lar genetic alterations have been found in
elicited by meningeal invasion. components were derived from neoplas- the gliomatous and mesenchymal com-
tic glial cells. This explanation has been ponents, indicating a monoclonal origin.
/mmunophenotype further supported by the observation of Expression of SNA12, TWIST, MMP2, and
The reticulin-free glial component is posi- p53 immunoreactivity in both tumour MMP9 is characteristic of mesenchymal
tive for GFAP. The mesenchymal com- components (39). Proof of a monoclonal tumour areas, suggesting that the mech-
ponent is largely negative for GFAP, but origin has been demonstrated in 2 cases anisms involved in epithelial-mesenchy-
isolated positive spindle cells are com- of gliosarcoma in which the gliomatous mal transition in epithelial neoplasms
mon. Staining for R132H-mutant IDH1 and sarcomatous components each may also play roles in mesenchymal
is negative in almost all cases (1183) contained an identical TP53 mutation differentiation in gliosarcomas (17371.
lmmunopositivity for p53, if present, is {197). In addition, identical PTEN, TP53, Microarray-based comparative genomic
identified in both glial and mesenchymal and TERT mutations were detected in hybridization analysis in glial and mes-
components (197,1038). the gliomatous and sarcomatous tumour enchymal tumour areas also suggests
components of other gliosarcoma cases that the mesenchymal components may
Cell of origin (2095,26791. The monoclonality of the be derived from glial cells with additional
Originally, gliosarcoma was thought to two components of the gliosarcomas genetic alterations in a small proportion
be a collision tumour, with a separate was also confirmed by identification of of gliosarcomas {1736}. See Table 1 04
astrocytic component and independent COKN2A deletion and MDM2 and COK4 on p. 47.
development of the sarcomatous portion coamplification in both tumour areas
from the proliferating vessels. Several im- {20951. These studies strongly support Prognosis and predictive factors
munohistochemical studies seemed to the hypothesis that the sarcomatous ar- Large clinical trials have failed to reveal
support this assumption, by demonstrat- eas are a result of a phenotypic change any significant differences in outcome
ing immunoreactivity for von Willebrand in the glioblastoma cells, rather than an between gliosarcoma and classic glio-
factor (2276}, UEA-1 (2370}, and mono- indication of the coincidental develop- blastoma (7791. However, there have
histiocytic markers (885,1312} Another ment of two separate neoplasms. been multiple reports of gliosarcomas
hypothesis was that the sarcomatous with systemic metastases and even inva-
portion resulted from advanced glioma Genetic profile sion of the skull {148,1902,2302)
dedifferentiation with subsequent loss Gliosarcoma contains PTEN muta-
of GFAP expression and acquisition of tions, CDKN2A deletions, and TP53

Gliosarcoma 49
Epithelioid glioblsstoms Ellison D.W. astrocytoma has been identified by bi-
Kleinschmidt-DeMasters B.K. opsy as pre-existent or coexistent with
Park S.-H. the epithelioid tumour {285,1297,1735,
1795,1977) The fact that anaplastic pro-
gression of a pleomorphic xanthoastro-
cytoma can manifest as an epithelioid
glioblastoma raises the possibility that
the two entities may be related, an asso-
Definition ciation reinforced by their sharing a high
A high-grade diffuse astrocytic tumour frequency of BRAF V600E mutation {48,
variant with a dominant population of 2508).
closely packed epithelioid cells, some Approximately 50% of epithelioid glio-
rhabdoid cells, mitotic activity, microvas- blastomas harbour a BRAF V600E mu-
cular proliferation, and necrosis. tation, but it is unknown what genetic
Epithelioid glioblastomas occur predomi- mechanism(s) might be responsible for
nantly in young adults and children, are the emergence of epithelioid morphology
preferentially located in the cerebrum or di- in the remainder. One microarray-based
encephalon, and are aggressive tumours comparative genomic hybridization study
with short survival, particularly in children. of a coexistent diffuse astrocytoma and
Compared with other glioblastomas, more epithelioid glioblastoma identified three
epithelioid glioblastomas (-50%) contain a copy number alterations observed only
BRAFV600E mutation. in epithelioid tumour cells: a homozygous
Epithelioid glioblastoma is distinct from gli- deletion in LSAMP and heterozygous de-
omas or glioneuronal tumours with a poor- Fig. 1.47 Epithelioid glioblastoma. Complex abnormali- letions in TENM3 and LRP1B. This result
ties (including solid and cystic areas and focal
ly differentiated SMARCB1-deficient com- awaits independent validation, but sug-
enhancement) usually characterize the MRI.
ponent (which may contain rhabdoid cells gests that BRAF V600E mutation alone
and exhibit a polyimmunophenotype). It is may be permissive but not sufficient for
also distinct from glioblastoma with epi- ICD-0 code 9440/3 development of epithelioid phenotype.
thelial metaplasia, which exhibits glandu-
lar formations and squamous metaplasia. Grading Localization
The term "rhabdoid glioblastoma" should Epithelioid glioblastoma corresponds Epithelioid glioblastomas have been
be avoided. Tumours previously reported histologically to WHO grade IV. described mainly in the cerebral cortex
as such are either epithelioid glioblasto- and diencephalon. Temporal and frontal
mas without evidence of SMARCB1 or Epidemiology lobes are common sites, but any lobe
SMARCA4 alteration or rare glioblastomas Incidence data are not yet available for can be affected { 285, 331, 1297, 1699).
with a population of SMARCB1-deficient this uncommon variant, which has been Rare examples occur in the cerebellum,
cells, from which the epithelioid glioblasto- inconsistently defined in various studies. but none have been reported in the spi-
ma, with its different genetic profile, should nal cord {1302).
be distinguished. Epithelioid glioblastoma Etiology
may coexist with pleomorphic xanthoas- The etiology and cell of origin of epithe- Clinical features
trocytoma, but the relationship between lioid glioblastoma are unknown, but most The clinical manifestation of epithelioid
epithelioid glioblastoma and malignant cases arise de novo. In several cases of glioblastoma parallels that of other glio-
progression in pleomorphic xanthoastro- epithelioid glioblastomas with confirmed blastomas; most patients present with
cytoma requires further clarification. SMARCB1 expression, a low-grade symptoms and signs of raised intracranial

......:.-�--_,. • Jli,,,.'..-�..
Fig. 1.48 Epithelioid glioblastoma. A Foci of necrosis are found in most cases. B Tumours may contain cells with an epithelioid, rhabdoid, or gemistocytic morphology.

50 Diffuse gliomas
,. ..
s-
�- ...
• ,;·
"' .
;
.· . ..'•.. •· ,.
'
. ' '

.
'"' '
• ••

pressure, although a minority of patients membrane, eosinophilic cytoplasm, a express melan-A, desmin, myoglobin, or
manifest neurological deficit or epilepsy paucity of cytoplasmic processes, and a smooth muscle antigen. SMARCB1 is re-
(331,1297,1699). Studies rarely suggest laterally positioned nucleus. There is vari- tained throughout the tumour cell popula-
a precursor lesion, but there have been ation in how often cytoplasmic filamen- tion. In cases where it has been sought,
several reports of transformation from tous-like balls are described or sought expression of SMARCA4 has been dem-
pleomorphic xanthoastrocytoma or WHO by electron microscopy, but some rhab- onstrated (285). lmmunohistochemistry
grade II astrocytoma (1297,2508). doid cells are found in epithelioid glio- using the VE1 antibody that recognizes
blastoma. Less xanthomatous change V600E-mutant BRAF shows reactivity in
Imaging is seen in epithelioid glioblastoma than about 50% of epithelioid glioblastomas, a
On MRI, epithelioid glioblastoma char- in pleomorphic xanthoastrocytoma, al- result concordant with sequencing results
acteristically presents as a gadolinium- though exceptional cases have been {1298).
enhancing solid mass, occasionally with noted to contain giant cells {1298). lipidi-
cysts (285,1297,2508). Epithelioid glio- zation {2151). a desmoplastic response Genetic profile
blastomas are prone to haemorrhage and (2151). or cytoplasmic vacuoles (1795). A BRAF V600E mutation is detected in
often spread through the leptomeninges. Rosenthal fibres and eosinophilic granu- about half of all epithelioid glioblastomas
lar bodies are not features of epithelioid {285,1297). An H3F3A K27M mutation
Spread glioblastoma. By definition, squamous has been reported in a single epithe-
Epithelioid glioblastomas demonstrate nests, glandular formation, and adenoid lioid glioblastoma, but other H3F3A and
a tendency to spread throughout the features are absent (2151). Necrosis is HIST1H3B mutations have not been re-
neuraxis, showing leptomeningeal dis- present, but is usually of the zonal rather ported {285). Epithelioid glioblastomas
semination in as many as one third of all than palisading type. Some reports have also lack IOH1 and IDH2 mutations.
patients (976,1299,1699,2755}. One re- noted a relative paucity of microvascular Copy number alterations in genes in-
ported paediatric tumour spread to the proliferation, but others have found no volved in high-grade gliomas are oc-
scalp via a shunt device {285). substantial difference in the vasculature casionally present; EGFR amplification,
patterns of epithelioid glioblastoma and homozygous deletion of COKN2A, and
Macroscopy classic glioblastoma {285). loss of PTEN have been reported, but
Epithelioid glioblastomas are typically copy number alterations at the PDGFRA,
unifocal lesions, although at least one lmmunophenotype PTEN, and METloci are rare (285,1297)
case with multifocal distribution has Epithelioid glioblastomas show immunore-
been reported, and metastatic disease activity for vimentin and S100. They also Genetic susceptibility
may occur {788). Although no feature show immunoreactivity for GFAP, although Epithelioid glioblastomas have not been
is pathognomonic on gross examina- it is often patchy and in a few cases entire- reported in association with dysgenetic
tion, haemorrhage and necrosis may be ly absent from most areas of the tumour. or hereditary tumour syndromes.
prominent. A superficial location, even Some epithelioid glioblastomas express
with dural attachment, has occasionally epithelial markers, cytokeratins, and EMA Prognosis and predictive factors
been noted. Leptomeningeal spread is (285,2151). Because classic glioblasto- In both adult and paediatric patients, epi-
relatively common. Cyst formation oc- mas sometimes express cytokeratin AE1/ thelioid glioblastoma has a particularly poor
curs, but is not a common feature. AE3 or EMA {965,1818). an epithelial im- prognosis, even for a glioblastoma {285,
munophenotype should not be consid- 426,1299). It demonstrates early progres-
Microscopy ered diagnostic of the epithelioid variant sion and a median survival of 6.3 months
Epithelioid glioblastomas are dominated in isolation, without additional morphologi- (range: 0.6-82 months} in adults and
by a relatively uniform population of epi- cal evidence. Most authors have noted fo- 5.6 months (range: 1.5-9.7 months} in chil-
thelioid cells showing focal discohesion, cal immunoreactivity for synaptophysin dren, despite various adjuvant therapies.
scant intervening neuropil, a distinct cell or NFP. Epithelioid glioblastomas do not

Epithelioid glioblastoma 51
,

Glioblastoma, IDH-mutant Ohgaki H.


Kleihues P.
Reifenberger G.
Yan H.
von Deimling A. Weller M.
Louis D.N.

Definition I Glial progenitor cells : I Glial progenitor cells I


A high-grade glioma with predominantly
astrocytic differentiation; featuring nucle-
ar atypia, cellular pleomorphism (in most
Common precursor cells with IDH1!2 mutation
cases), mitotic activity, and typically a dif-

I I
fuse growth pattern, as well as microvas- TP53mutation (>65%) TERTmutation (-70%)
cular proliferation and/or necrosis; with a ATRXmutation (>65%) Loss 1 p/19q(>75%)
C/Cmutation (-40%)
mutation in either the IDH1 or IDH2 gene. FUBP1 mutation (-15%)
Clinical history
IDH-mutant glioblastomas account for
approximately 10% of all glioblastomas.
3-6 months I Diffuse astrocytoma I Oligodendroglioma
Glioblastomas that develop through
malignant progression from diffuse as-
trocytoma (WHO grade II) or anaplastic
TERTmutation (-70%)
!
Anaplastic Anaplastic

l
EGFRamplification (-35%)
astrocytoma (WHO grade Ill) are almost TP53mutation (-30%) astrocytoma oligodendroglioma
always associated with IDH mutation and PTENmutation (-25%)
LOH 10p (-50%) LOH 19q (-50%)
therefore carry the synonym "secondary LOH 10q (>60%)
LOH 10q (-70%)
glioblastoma. IDH-mutant". IDH-mutant TERTmutation (30%)
I
glioblastoma is morphologically indistin- Glioblastoma I
I Glioblastoma
guishable from IDH-wildtype glioblas- IDH-wildtype
I
I IDH-mutant
toma, except for a lesser extent of necro- I
I
I
sis. IDH-mutant glioblastomas manifest Neural, classical, mesenchymal, : Proneural transcriptional profile

�----------------------- �--------------------------------------------
in younger patients (with a mean patient proneural transcriptional profiles , Hypermethylation phenotype
age at diagnosis of 45 years), are pref- Fig. 1.50 Genetic pathways to IDH-wildtype and !DH-mutant glioblastoma. This chart is based on the hypothesis
erentially located in the frontal lobe. and that !DH-mutant glioblastomas share common glial progenitor cells not only with diffuse and anaplastic astrocytomas.
carry a significantly better prognosis than but also with oligodendrogliomas and anaplastic oligodendrogliomas. Adapted from Ohgaki Hand Kleihues P {1830}.
IDH-wildtype glioblastomas {1797,2810).

grading reflects the natural course of the glioblastoma had a histologically proven
Genetic hallmarks disease rather than response to therapy. prior low-grade glioma {605). When IDH1
The defining genetic hallmark is the Therefore. because most patients even- mutations were used as a genetic mark-
presence of IDH mutations {1895). tually succumb to high-grade disease. er. secondary glioblastomas accounted
which are associated with a hyper- IDH-mutant glioblastomas are designat- for 9% of all glioblastomas at the popula-
methylation phenotype {1810). These ed as WHO grade IV. tion level (1797) and for 6-13% of cases
mutations are the earliest detectable in hospital-based studies {118,1078,1417,
genetic alteration in precursor low- Synonym 2810}
grade diffuse astrocytoma. indicating Secondary glioblastoma. I DH-mutant
that these tumours are derived from Age distr;bution
common neural precursor cells that Epidemiology At the population level. secondary glio-
differ from those of IDH-wildtype glio- blastomas develop in patients signifi-
blastoma. Additional typical genetic Incidence cantly younger (mean: 45 years) than do
alterations are TP53 and ATRX muta- Until the discovery of IOH1 mutation as primary glioblastomas (mean: 62 years)
tions and loss of chromosome arm a molecular marker. the diagnosis of {1823,1826}. Correspondingly, the mean
10q {1822,1830}. secondary glioblastomas was based on age of patients with IOH1-mutant glio-
clinical observations (i.e. neuroimaging blastoma is 48 years. significantly youn-
ICD-0 code 9445/3 and/or histological evidence of a preced- ger than that of patients with glioblasto-
ing low-grade or anaplastic astrocytoma) mas that lack IDH1 mutations (61 years)
Grading (1823,1827). In a population-based study {1797). Several hospital-based studies
IDH-mutant glioblastoma corresponds in Switzerland. using clinical criteria and have also shown that patients with IDH-
histologically to WHO grade IV. histopathological evidence. only 5% of all mutant glioblastoma were significantly
The prognosis of IDH-mutant glioblasto- glioblastomas diagnosed were second- younger than those with IDH-wildtype
ma is considerably better than that of IDH- ary {1823,1826). Similarly, another study glioblastoma {214,1078,2810}.
wildtype glioblastoma; however. WHO showed that 19 of 392 cases (5%) of

52 Diffuse gliomas
SeX diSt(lbUtion Table 1.05 Key characteristics of IDH-wildtype and I DH-mutant glioblastoma in adults
one population-based study of !DH-mu- IDH-wildtype glioblastoma IDH-mutant glioblastoma References
tant glioblastoma found a male-to-female Primary glioblastoma, Secondary glioblastoma,
Synonym (1830}
ratio (1830) that is significantly lower than IDH-wildtype I DH-mutant
seen in patients with IDH-wildtype glio- Not identifiable; Diffuse astrocytoma
blastoma (1823). In a multicentre study, Precursor lesion (1827}
develops de novo Anaplastic astrocytoma
49 of 618 glioblastomas (7.9%) carried an
Proportion of glioblastomas -90% -10% (1797}
!OH1 mutation, and the male-to-female
ratio was 0.96:1, versus a ratio of 1.63:1 (214, 1078, 1797,
Median age at diagnosis -62 years -44 years
2103}
for IDH-wildtype glioblastomas (1417f.
Male-to-female ratio 1.42:1 1.05:1 (214,1417,1797}
Localization Mean length of clinical history 4 months 15 months (1797}
Whereas IDH-wildtype glioblastomas Median overall survival
show a widespread anatomical distribu- Surgery+ radiotherapy 9.9 months 24 months (1797}
tion, !DH-mutant glioblastomas have a Surgery + radiotherapy
15 months 31 months (2810}
striking predominance of frontal lobe in- + chemotherapy
volvement, in particular in the region sur- Location Supratentorial Preferentially frontal (1417}
rounding the rostral extension of the lat-
Necrosis Extensive Limited (1417}
eral ventricles (1417f. This is similar to the
preferential localization of WHO grade 11 TERT promoter mutations 72% 26% (1801, 1830}
!DH-mutant diffuse astrocytoma (2428) TP53 mutations 27% 81% (1797}
and !DH-mutant and 1p/19q-codeleted ATRX mutations Exceptional 71% (1519}
oligodendroglioma (1418,2871). support-
EGFR amplification 35% Exceptional (1797}
ing the hypothesis that these gliomas de-
velop from a distinct population of com- PTEN mutations 24% Exceptional (1797}
mon precursor cells {158,1830}

Clinical features glioblastoma, !DH-mutant glioblastoma which are hallmarks of IDH-wildtype glio-
presents more frequently with non-en- blastoma, are usually absent.
Clinical history hancing tumour components on MRI,
The mean length of the clinical history of with larger size at diagnosis, with lesser Microscopy
patients with clinically diagnosed second- extent of oedema, and with increased The histological features of I DH-mutant
ary glioblastoma was 16.8 months, sig- prevalence of cystic and diffuse compo- glioblastomas are similar to those of
nificantly longer than that of patients with nents {644,1417). IDH-wildtype glioblastomas. However,
primary glioblastoma (6.3 months) (1823, morphological studies have revealed
1826,1827) Correspondingly, patients Macroscopy two significant differences. Areas of is-
with secondary glioblastoma harbour- Like all glioblastomas, !DH-mutant glio- chaemic and/or palisading necrosis
ing an IOH1 mutation had a much longer blastomas diffusely infiltrate the brain have been observed in 50% of IDH-
mean clinical history (15.2 months) than parenchyma, without clear macroscopic mutant glioblastomas, significantly less
did patients with primary IDH-wildtype delineation. However, large yellowish ar- frequently than in IDH-wildtype glio-
glioblastomas (3.9 months) (1797). eas of central necrosis or haemorrhage, blastomas (90%) (1797l, whereas focal

v -
Symptoms
., �
100
Because !DH-mutant glioblastomas are /
90
preferentially located in the frontal lobe,
/
behavioural and neurocognitive changes
v,
80
/ v
are likely to predominate, although focal
v
QI
70
/
v,

neurological deficits (e.g. hemiparesis "'u 60


0
and aphasia) also frequently occur. Due
to the slow evolution from diffuse astrocy-
';f!. so
/ V
/
/ /DH-wildtype (n=641 J
QI /DH-mutant (n-Sl)
>
·,p Median age 44 years J Median aqe 62 years
toma or anaplastic astrocytoma, tumour- �::, 40

associated oedema is less extensive and E 30


,/
/
/
symptoms of intracranial pressure may ::,
/ /
v
u 20
develop less rapidly than in patients with /"
primary IDH-wildtype glioblastoma. 10

-- /

=
Imaging 10 20 30 40 so 60 70 80 90 100

Unlike in IDH-wildtype glioblastoma, Age at diagnosis :��:;i��:•


large areas of central necrosis are usu- Fig. 1.51 Cumulative age distribution of glioblastomas with and without IDH1 mutations. Combined data from
ally absent. Compared with IDH-wildtype Nobusawa S et al. (1797} and Cancer Genome Atlas Research Network (350}.

Glioblastoma, !DH-mutant 53
diagnosis in 385 of 407 cases {95%)
{1797}. In this regard, IDH mutations are
considered to be a defining diagnostic
molecular marker of glioblastomas de-
rived from I DH-mutant diffuse astrocy-
toma or IDH-mutant anaplastic astrocy-
toma that is more objective than clinical

"
and/or histopathological criteria.
5 years
Timing of /DH mutation
IDH mutations are an early event in glio-
magenesis and persist during progres-
sion to IDH-mutant glioblastoma. Analy-
sis of multiple biopsies from the same
patients revealed no cases in which the
IDH1 mutation occurred after the ac-
Fig. 1.52 These MRls show the typical progression of a frontotemporal /OHi-mutant diffuse astrocytoma (left) to an quisition of a TP53 mutation {2709). An
IDH1-mutant secondary glioblastoma (right) over a period of 5 years (1533). Note the absence of central necrosis.
exception is IDH1 mutations in patients
with Li-Fraumeni syndrome. caused by a
oligodendroglioma-like components are Cell of origin germline TP53 mutation. In this inherited
more common in IDH-mutant glioblasto- Despite their similar histological fea- tumour syndrome, the TP53 mutation is
mas than in IDH-wildtype glioblastomas tures, IDH-mutant glioblastomas and by definition the initial genetic alteration
(54% vs 20%) {1797). A larger propor- IDH-wildtype glioblastomas seem to be and significantly affects the subsequent
tion of tumour cells with oligodendroglial derived from different precursor cells. acquisition of the IDH1 mutation. The
morphology has also been reported in Secondary glioblastomas, astrocytomas. R132C (CGT---,TGT) mutation. which is
/OH1-mutant glioblastomas. in a large and oligodendrogliomas contain IDH uncommon in sporadic cases. was the
study of 618 cases {1417) mutations. share a preferential frontal lo- only IDH1 mutation found in diffuse as-
cation. and have been hypothesized to trocytomas. anaplastic astrocytomas,
lmmunophenotype originate from precursor cells located in and secondary glioblastomas carrying a
The presence of IDH1 R132H (the most (or migrating to) the frontal lobe. In con- TP53 germ line mutation {2710)
frequent /OH1 mutation in oligodendro- trast. IDH-wildtype glioblastomas have
gliomas. astrocytomas. and IDH-mutant more widespread locations. Additional Types of /DH mutation
glioblastomas) can be detected immuno- evidence supporting this hypothesis in- All reported IDH1 mutations are located
histochemically using an antibody to the cludes the observation that glioblasto- at the first or second base of codon 132
gene product. R132H-mutant IDH1 {357) mas that are IDH-mutant and those that {118,1895,2709). The most frequent is
Positivity in a glioblastoma is indicative of are IDH-wildtype develop in patients of the R132H (CGT--->CAT) mutation, found
an I DH-mutant glioblastoma. but negativ- different ages. have a different sex distri- in 83-91% of astrocytic and oligoden-
ity could be due to the presence of one bution. and have a significantly different droglial gliomas {118,2709,2810). Other
of the less common types of IDH1 mu- clinical outcome {1830). Their stem cells mutations are rare. including R132C
tation or an IDH2 mutation {953) Lack may also be different; in one study, the (CGT--->TGT; found in 3.6-4.6% of cases).
of immunoreactivity could also indicate relative content of CD133-positive cells R132G (in 0.6-3.8%), R132S (in 0.8-
the presence of a primary IDH-wildtype was significantly higher in primary glio- 2.5%). and R132L (in 0.5-4.4%) {118,
glioblastoma. blastomas than in secondary glioblasto- 2709,2810).
Mutations in the ATRX gene are typically mas, and CD133 expression was associ- The IDH2 gene encodes the only human
present with IDH1/2 mutations and TP53 ated with neurosphere formation only in protein homologous to IDH1 that uses
mutations in WHO grade II or Ill diffuse primary glioblastomas {158).
astrocytomas and IDH-mutant glioblas-
tomas {1160). Mutations in ATRX result Genetic profile
in lack of expression. which can be dem-
onstrated immunohistochemically {2750). !DH mutations
lmmunohistochemical overexpression of IDH mutations were first reported in 2008
p53 is frequent. reflecting the high fre- {1895). and the authors noted that "mu-
quency of TP53 mutations (present in the tations in IDH1 occurred in a large pro-
vast majority of cases). Overexpression portion of young patients and in most pa-
of EGFR is unusual; EGFR amplification tients with secondary glioblastomas and
is a hallmark of IDH-wildtype glioblas- were associated with an increase in over-
toma. GFAP expression is consistently all survival". In one study, the presence
Fig. 1.53 lmmunohistochemistry of a glioblastoma using
present. although the level of expression of IDH1 mutations as a genetic marker of an antibody to R132H-mutant IDH1. Note the strong
is variable. secondary but not primary glioblastoma cytoplasmic expression by tumour cells and the lack of
corresponded to the respective clinical expression in the vasculature.

54 Diffuse gliomas
NADP+ as an electron acceptor. IDH2 studies found that 3-6% of patients with Genetic alterations typical of IDH-
mutations are all located at residue R172, /OH1-mutant glioblastomas clinically di- wildtype glioblastoma include EGFR
which is the analogue of the R132 resi- agnosed as primary were 13-27 years amplification, PTEN mutation, TERT
due in JOH1 (2805l. It is located in the ac- younger than those with glioblasto- promoter mutation, and complete loss
tive site of the enzyme and forms hydro- mas without IDH1 mutations (118,1829, of chromosome 10 (89,752,1801,1823,
gen bonds with the isocitrate substrate. 2561,2810} The possibility exists that 1827). Alterations more common in
!DH2 mutations are rare in IDH-mutant these glioblastomas have rapidly pro- secondary IDH-mutant glioblastomas
glioblastoma and located at codon 172 gressed from precursor astrocytomas include TP53 mutations and 19q loss
(2810}, with the R172K mutation being that escaped clinical diagnosis and (1754,1823,1827}. However, until the iden-
the most frequent. were therefore misclassified as primary tification of IDH mutation as a molecular
glioblastomas. marker of secondary glioblastoma (118,
Secondary glioblastomas without /DH 1797, 2709, 2810}, the patterns of genetic
mutation A TRX mutations alterations, although different, did not en-
The few clinically diagnosed second- Mutations in the ATRX gene cause loss able unequivocal diagnosis of these two
ary glioblastomas that lack an IDH mu- of expression. They are typically present glioblastoma subtypes.
tation have infrequent TP53 mutations, with IDH and TP53 mutations in WHO
and patients have a shorter clinical his- grade II I DH-mutant diffuse astrocytoma, Expression profile
tory and a poor prognosis (1797). Most WHO grade Ill IDH-mutant anaplastic More than 90% of IDH-mutant glioblas-
secondary glioblastomas lacking IDH1 astrocytoma, and I DH-mutant glioblasto- tomas have a proneural expression sig-
mutations develop through progression mas (1160,1519). nature, and approximately 30% of glio-
from a WHO grade Ill anaplastic astro- blastomas with a proneural signature are
cytoma, whereas the majority of second- Other genetic alterations IDH-mutant (2645). These findings sug-
ary /OH1-mutant glioblastomas progress Genetic profiling of primary and second- gest that I DH-mutant glioblastomas are a
from a WHO grade II diffuse astrocytoma ary glioblastomas has shown that the fre- relatively homogeneous group of gliomas,
(1797). Therefore, some tumours diag- quency of TP53 mutations is significantly characterized by a proneural expression
nosed as anaplastic astrocytoma may higher in secondary glioblastomas (67%) pattern. IDH-mutant diffuse astrocytoma
actually have been primary glioblasto- than in primary glioblastomas (11%) and IDH-mutant and 1p/19q-codeleted
mas misdiagnosed due to a sampling (2705). EGFR overexpression prevails oligodendroglioma also have the typical
error. in glioblastomas that are IDH-wildtype proneural signature (496}, further sup-
but is rare in secondary glioblastomas porting the assumption that these neo-
Primary glioblastomas with /DH mutation (2705). In one study, only 1 of 49 glio- plasms share common neural progenitor
In a population-based study, only 14 of blastomas showed both TP53 mutation cells. In contrast, IDH-wildtype glioblas-
407 glioblastomas (3.4%) clinically diag- and EGFR overexpression, suggesting tomas are heterogeneous, with several
nosed as primary carried an IDH1 mu- that these alterations are mutually exclu- distinct expression profiles.
tation. The patients were approximately sive events that could define two differ-
10 years younger than patients with sec- ent genetic pathways in the evolution of Hypermethylation phenotype
ondary glioblastomas, but the genetic glioblastoma (2705). Subsequent studies IDH-mutant glioblastomas show con-
profiles of the two tumour groups were provided evidence that primary and sec- certed CpG island methylation at many
similar, including frequent TP53 muta- ondary glioblastomas develop through loci (1810}. A similar hypermethyla-
tions and absence of EGFR amplification distinct genetic pathways. tion phenotype has been observed in
(1797). Similarly, several hospital-based IDH-mutant diffuse astrocytomas (454)

100

.8 - 80

iv .6 60
.z
<'::,.

e .4
(/)
40
::, Secondary GBM IDH mutated
u
Mutated IDH1
.2 (median, 24.0 months, n=17) 20

Wild-type IDH1
a (median. 9.9 months, n=186)
10 20 30 40 S-0 60
A a 12 24 36 48 60 Months 8 Months
Fig. 1.54 A Cumulative survival rate of patients with glioblastoma treated with surgery plus radiotherapy. Note that patients carrying an IDH1 mutation had significantly longer overall
survival survival. Reprinted from Nobusawa Set al. {1797}. B In a more recent study, the median survival was 31 months for 14 patients with mutated IDH1 or IDH2, versus 15 months for
115 patients with wildtype IDH1 or IDH2. For patients with I DH-mutant glioblastomas, survival was calculated from the date of the secondary diagnosis. Reprinted from Yan H et al. (281 O}.

Glioblastoma, IDH-mutant 55
and oligodendrogliomas (454l, as well as neural precursor cells that already carry
in IDH-mutant acute myeloid leukaemia a germline TP53 mutation.
{706}. The introduction of mutant IDH1 Glioblastoma, NOS
into primary human astrocytes alters spe- Prognosis and predictive factors
cific histone markers and induces exten- In his pioneering work on glioblastomas, Definition
sive DNA hypermethylation, suggesting H-J Scherer (2265) wrote, in 1940: "From A high-grade glioma with predominantly
that the presence of an IOH1 mutation is a biological and clinical point of view, the astrocytic differentiation; featuring nucle-
sufficient to establish a hypermethylation secondary glioblastomas developing in ar atypia, cellular pleomorphism (in most
phenotype (2589). Moreover, IDH muta- astrocytomas must be distinguished from cases), mitotic activity, and typically a
tions disrupt chromosomal topology and 'primary' glioblastomas. They are prob- diffuse growth pattern, as well as micro-
thus allow aberrant chromosomal regula- ably responsible for most of the 'glioblas- vascular proliferation and/or necrosis; in
tory interactions that induce oncogene tomas of long clinical duration' mentioned which /OH mutation status has not been
expression, including glioma oncogenes in the literature." This early observation fully assessed.
such as PDGFRA (713A} has been repeatedly confirmed. In a Determination of IDH mutation status
1980-1994 population-based study, the is important for all glioblastomas; how-
Genetic susceptibility median overall survival of patients with ever, if full testing is not possible (see
Astrocytic gliomas, including diffuse as- clinically diagnosed secondary glioblas- Genetic parameters in Glioblastoma,
trocytomas. anaplastic astrocytomas, toma was 7.8 months, significantly longer IOH-wildtype, p 28), the diagnosis of
and secondary glioblastomas, are the than that of patients with primary glio- glioblastoma, NOS, can be made, pro-
most frequent brain tumours associated blastoma (4.7 months) (18231. Similarly, vided that the histological variants giant
with TP53 germline mutations in fami- the analysis of patients who were treated cell glioblastoma, gliosarcoma, and epi-
lies with Li-Fraumeni syndrome (1294, with surgery and radiotherapy showed thelioid glioblastoma can be ruled out.
1841) In patients from three families that the mean overall survival of patients
with Li-Fraumeni syndrome, IDH1 mu- with IDH-mutant glioblastomas was ICD-0 code 9440/3
tations were observed in 5 astrocytic 27.1 months, 2.4 times as long as that of
gliomas that developed in carriers of a patients with IDH-wildtype glioblastoma Grading
TP53 germline mutation. All 5 contained (11.3 months) (1797). In another study, Glioblastoma, NOS, and its variants (e.g.
the R132C (CGT->TGT) mutation (2710). patients with IDH-mutant glioblastomas giant cell glioblastoma that cannot be
which in sporadic astrocytic tumours ac- treated with radiotherapy/chemotherapy tested for IDH mutation status) corre-
counts for < 5% of all IDH1 mutations had an overall survival time of 31 months, spond histologically to WHO grade IV
overall (118,2709,2810). This remark- twice as long as that of patients with IDH-
ably selective occurrence suggests wildtype glioblastoma (28101
a preference for R132C mutations in

56 Diffuse gliomas
� \_____j___�

h�Hawk;nsC
-• DJffuse midline glioma, \ Ellison DW
J-13 K27M-mutant Sturm D.

....'·.. •. ..
Definition glioma (DIPG), respectively. Mitotic ac-
Other midline sites • • • ••
An infiltrative midline high-grade glioma
with predominantly astrocytic differentia-
tion and a K27M mutation in either H3F3A
tivity is present in most cases, but is not
necessary for diagnosis; microvascular
proliferation and necrosis may be seen.
Thalamus •:• :•

..
or HIST1 H3B/C. Tumour cells diffusely infiltrate adjacent Pons fC-: ·• •''
H3 K27M-mutant diffuse midline glioma and distant brain structures. The progno- Spinal cord • ( ••
predominates in children but can also be sis is poor, despite current therapies, with
0 10 20 30 40 50 60
seen in adults, with the most common lo- a 2-year survival rate ot « 10%.
cations being brain stem, thalamus, and Patient age (years)
spinal cord. Brain stem and pontine ex- ICD-0 code 9385/3 Fig. 1.56 Location of diffuse midline gliomas, H3 K27M-
amples were previously known as brain mutant. Pontine gliomas manifest primarily in children,
stem glioma and diffuse intrinsic pontine whereas spinal cord lesions predominantly affect adults.
Reprinted from Solomon DA et al. {2392A}.

Grading
H3 K27M-mutant diffuse midline glioma
corresponds to WHO grade IV.

Epidemiology
Incidence data on diffuse gliomas specif-
ically arising in midline structures are not
available, because large brain tumour
registries have not yet included these as
a distinct category. The median patient
age at diagnosis of diffuse midline glioma
is 5-11 years, with pontine tumours aris-
ing earlier on average (at -7 years) than
their thalamic counterparts (at -11 years).
There is no clear sex predilection {1229,
1358,1863,2700,2776l.

Localization
Diffuse midline gliomas typically arise
in the pons, thalamus, or spinal cord,
with occasional examples involving the
cerebellum.

Clinical features
Most patients with DIPG present with
brain stem dysfunction or cerebrospinal
fluid obstruction, typically developing
over a short period of time (1-2 months).
Classic clinical symptoms include the tri-
ad of multiple cranial neuropathies, long
tract signs, and ataxia {2700). With tha-
lamic gliomas, common initial symptoms
Fig. 1.55 H3 K27M-mutant diffuse midline glioma. A Sagittal T2-weighted MRI showing a diffusely infiltrating include signs of increased intracranial
pontine glioma expanding the pons with crowding of the neural structures at the craniocervical junction and tonsillar
pressure, motor weakness/hemiparesis,
herniation to the level of the C1 posterior arch. B Axial FLAIR MRI demonstrates a diffusely infiltrating pontine glioma
expanding the pons and encasing the basilar artery. C Axial FLAIR MRI shows an expansile left thalamic glioma and and gait disturbance {1358}.
associated obstructive hydrocephalus, with mild to moderate dilatation of the lateral ventricles, periventricular oedema,
and distortion of the third ventricle. D Expansile thalamic glioma showing heterogeneous enhancement on the post- Imaging
gadolinium coronal T1 sequence. On MRI, diffuse midline gliomas are

Diffuse rnidline glioma. H3 K27fv1-mutant 57


usually T1-hypointense and T2-hyperin- synaptophysin immunoreactivity can be
tense. Contrast enhancement, necrosis, focal, but chromogranin-A and NeuN are
and/or haemorrhage may be present. not typically expressed. An H3F3A K27M
DIPG typically presents as a large, ex- mutation can be detected by immune-
pansile, and often asymmetrical brain histochemistry using a mutation-specific
stem mass occupying more than two antibody. Nuclear p53 immunopositivity
thirds of the pons {2700). There may be may be present, suggesting an under-
an exophytic component encasing the lying TP53 mutation, which is present in
basilar artery or protruding into the fourth about 50% of cases. In about 10-15% of
ventricle. Infiltration into the cerebellar cases, an ATRX mutation leads to loss of
peduncles, the cerebellar hemispheres, Fig. 1.57 Axial section of H3 K27M-mutant diffuse nuclear ATRX expression.
the midbrain, and the medulla is fre- mid line glioma showing expansion of the pons with areas
quent. Contrast enhancement rarely in- of haemorrhage and yellowish discolouration suggesting Cell of origin
volves> 25% of the tumour volume {135). necrosis. The common (epi)genetic features of
diffuse gliomas arising in midline loca-
Spread areas of necrosis or haemorrhage show tions, especially the pons and the thala-
Two large autopsy-based studies have focal discolouration and softening. mus, suggest a distinct developmental
found that leptomeningeal dissemination origin (1175,2443). Examination of the
of DIPG occurs in about 40% of cases Microscopy spatiotemporal distribution of neural pre-
{304,2831 }. Diffuse tumour invasion of Diffuse midline gliomas infiltrate grey and cursor cells in the human brain stem in-
the brain stem is common among DIPGs, white matter structures. The tumour cells dicated a nestin- and OLIG2-expressing
with 25% spreading to involve the upper are generally small and monomorphic, neural precursor-like cell population in
cervical cord and thalamus as well (304, but can be large and pleomorphic. They the ventral pons, which has been pro-
362,2831 ). Some patients exhibit distal typically have an astrocytic morphology, posed as the possible cell of origin of
spread as far as the frontal or (rarely) although oligodendroglial morphology is diffuse pontine gliomas (1701) However,
the occipital lobes, creating some phe- also a recognized pattern. About 10% of the exact cell or cells of origin of diffuse
notypic overlap with gliomatosis cerebri. DIPGs lack mitotic figures, microvascu- midline gliomas remain unknown.
No large series of diffuse midline glio- lar proliferation, and necrosis, and thus
mas centred in the thalamus has been are histologically consistent with WHO Genetic profile
reported, but some cases of gliomatosis grade II. The remaining cases are high-
cerebri have been reported to show an grade, with 25% containing mitotic fig- Mutation spectrum
H3F3A K27M mutation, suggesting that ures and the remainder containing mitot- Sequencing studies have identified re-
the thalamic version has a similar tenden- ic figures as well as foci of necrosis and current heterozygous mutations at po-
cy for diffuse invasion. microvascular proliferation. However, for sition K27 in the histone coding genes
DIPG, grade does not predict the out- H3F3A, HIST1H3B, and HIST1H3C in
Macroscopy come in genetically classic cases (see high-grade gliomas from the pons (in
Diffuse infiltration of CNS parenchyma by Prognosis and predictive factors). -80% of cases), thalamus (in -50%),
tumour cells produces distortion and en- and spinal cord (in -60%) (305,721,
largement of anatomical structures. Sym- lmmunophenotype 2304,2521,2791,2792}. Within the brain,
metrical or asymmetrical fusiform en- Virtually all tumour cells express NCAM1. these mutations occur exclusively in dif-
largement of the pons is typical in cases S100, and OLIG2, but immunoreactivity fuse midline gliomas. K27M mutations
of diffuse pontine glioma. Tumours with for GFAP is variable in these neoplasms. affecting H3.3 (encoded by H3F3A) are
MAP2 expression is common, and about three times as prevalent as the

58 Diffuse gliomas
in TP53, PPM10, CHEK2, or ATM; occur- Table 1.06 Additional mutations in H3 K27M-mutant
ring in -70% of cases), and to a lesser diffuse midline glioma
extent the retinoblastoma protein path- TP53 mutation 50%
way (305,2521,2792). Activating muta- PDGFRA amplification 30%
tions or fusions targeting FGFR1 were CDK4/6 or CCND1-3 amplification 20%
specifically identified in a small propor-
tion of thalamic high-grade gliomas ACVR1 mutation 20%
(10%) {721}. In contrast, recurrent muta- PPM1 D mutation 15%
tions in ACVR1, the gene encoding the MYC/PVT1 amplification 15%
BMP receptor ACVR1, were detected in
ATRXmutation 15%
a subset (-20%) of DIPGs, and seem to
correlate with H3.1 mutations (305,2521, CDKN2A/B homozygous deletion <5%
2792).
same mutation in histone variant H3.1
(occurring in HIST1H3B or HIST1H3C). Structural variations Genetic susceptibility
The K27M substitution (lysine replaced High-level focal amplifications detected Rarely, patients with Li-Fraumeni syn-
by methionine at amino acid 27) results in in diffuse midline gliomas include ampli- drome or neurofibromatosis type 1 pre-
a decrease in H3K27me3, thought to be fication of PDGFRA (in as many as 50% sent with midline infiltrating gliomas, but
due to inhibition of PRC2 activity (1480) of DIPGs), MYC/MYCN (in as many as there is no known specific genetic sus-
In addition to the specific mutations in 35%), COK4/6 or CCND1-3 (in 20%), /02 ceptibility for these tumours.
histone variants H3.3 and H3.1, chro- (in 10%), and MET (in 7%), whereas ho-
matin regulation is further targeted by mozygous deletion of CDKN2A/B or loss Prognosis and predictive factors
additional non-recurrent mutations in of RB1 or NF1 is detected only very rarely For diffuse midline gliomas in general, the
a diverse range of chromatin readers (in < 5% of cases). Fusion events involv- finding of an H3 K27M mutation confers
and writers, such as members of the ing the tyrosine kinase receptor gene a worse prognosis than that of wildtype
mixed-lineage leukaemia (MLL), lysine- FGFR1 occur in thalamic diffuse gliomas, cases {304,1263). In diffuse midline glio-
specific demethylase, and chromodo- and a small proportion (4%) of pontine mas arising in the thalamus, high-grade
main helicase DNA-binding protein fami- gliomas have been found to carry neu- histology is associated with short overall
lies {2792}. rotrophin receptor (NTRK) fusion genes. survival regardless of histone gene sta-
Other mutations in canonical cancer Common broad chromosomal alterations tus. This does not seem to be the case
pathways frequently target the recep- include single copy gains of chromo- for tumours with classic clinical and radi-
tor tyrosine kinase I RAS I Pl3K pathway some 1q and chromosome 2. In addition, ological features arising in the pons, but
(e.g. mutations in PDGFRA, PIK3CA, there may be a subset of pontine gliomas < 10% of these patients survive beyond
PIK3R1, or PTEN; occurring in -50% of (as many as 20%) that harbour few copy 2 years.
cases), the p53 pathway (e.g. mutations number changes {251,305,2792).

Diffuse midline glioma, H3 K27M-mutant 59


;,'

Oligodendroglioma, IDH-mutant and Reifenberger G.


Collins V.P.
Cairncross J.G.
Yokoo H.
1 p/19q-codeleted Hartmann C.
Hawkins C
Yip S.
Louis D.N.
Kros J.M.

Definition Genetic classification of analysed with comprehensive molecu-


A diffusely infiltrating, slow-growing gli- oligodendroglial tumours lar testing or in which the test results
oma with IDH1 or IDH2 mutation and In contrast to the 2007 WHO classifica- were inconclusive or uninformative.
codeletion of chromosomal arms 1 p and tion, the current WHO classification of Rare tumours demonstrate classic oli-
19q. oligodendrogliomas requires demon- godendroglial histology but lack \DH
Histologically, \DH-mutant and 1p/19q- stration of IDH1 or IDH2 mutation, typi- mutation and 1p/19q codeletion on
codeleted oligodendroglioma is com- cally by immunohistochemistry using molecular testing, a situation most fre-
posed of tumour cells morphologically the mutation-specific antibody against quently encountered in paediatric oligo-
resembling oligodendrocytes, with iso- R132H-mutant IDH1 (followed by DNA dendrogliomas. Such tumours should
morphic rounded nuclei and an artefactu- genotyping when R132H-mutant IDH1 not be classified as oligodendroglioma,
ally swollen clear cytoplasm on routinely immunostaining is negative), as well as NOS, and must be further evaluated
processed paraffin sections. Microcalci- demonstration of 1p/19q codeletion by to exclude histological mimics, such
fications and a delicate branching cap- FISH, chromogenic in situ hybridiza- as dysembryoplastic neuroepithelial
illary network are typical. The presence tion, or molecular genetic testing. The tumour, clear cell ependymoma, neu-
of an astrocytic tumour component is WHO classification does not mandate rocytoma, and pilocytic astrocytoma.
compatible with the diagnosis when mo- the use of a particular method for mo- Once histological mimics are excluded,
lecular testing reveals the entity-defining lecular testing of these markers, but such tumours can be tentatively clas-
combination of IDH mutation and 1p/19q recommends that 1 p/19q assays be sified as oligodendroglioma lacking
codeletion. The vast majority of IDH- able to detect whole-arm chromosomal IDH mutation and 1p/19q codeletion
mutant and 1p/19q-codeleted oligoden- losses. Pathologists should have expe- (paediatric-type oligodendroglioma). In
drogliomas occur in adult patients, with rience with their method of choice and children, most of these tumours seem
preferential location in the cerebral hemi- be aware of potential methodological to belong to a group of paediatric low-
spheres (most frequently in the frontal and interpretationa\ pitfalls. grade diffuse gliomas that are geneti-
lobe) On the rare occasion that the entity- cally characterized by FGFR1, MYB, or
defining molecular markers (ie. IDH MYBL1 alterations (see 0/igodendrogli-
ICD-0 code 9450/3 mutation and 1p/19q codeletion) can- oma lacking !OH mutation and 1p/19q
not be fully determined and classic codeletion, p. 68).
Grading histological oligodendroglioma features Tumours demonstrating evidence of
IDH-mutant and 1p/19q-codeleted oligo- are present, classification of the tumour 1p/19q codeletion but lacking detect-
dendroglioma corresponds histologically as oligodendroglioma, NOS, is recom- able IDH1 or IDH2 mutations should be
to WHO grade II. mended (see 0/igodendroglioma, NOS, further evaluated to exclude the possi-
Oligodendroglial tumours constitute a p. 69). This diagnosis indicates that the bility of incomplete deletions on 1 p and/
continuous spectrum ranging from well- tumour is a histologically classic oligo- or 19q. Such alterations may be pres-
differentiated, slow-growing neoplasms dendroglioma, which will likely exhibit ent i.n subsets of IDH-wildtype (mostly
to frankly malignant tumours with rapid a clinical behaviour similar to that of an high-grade) gliomas, including glioblas-
growth. The WHO grading system tra- IDH-mutant and 1p/19q-codeleted oli- tomas, and have been associated with
ditionally distinguished two malignancy godendroglioma, but that could not be poor outcome.
grades for oligodendroglioma: grade II for
well-differentiated tumours and grade Ill
for anaplastic tumours. Several studies longer survival of patients with WHO data from a large combined Japanese
have reported WHO grading as an inde- grade II versus grade Ill oligodendro- and The Cancer Genome Atlas (TCGA)
pendent predictor of survival for patients glial tumours with concurrent IDH muta- cohort suggest a limited prognostic role
with oligodendroglial tumours {686, tion and TERT promoter mutation (with of histological grading in patients with
830,1446,1826). However, these studies a median survival of 205.5 months with WHO grade II and Ill oligodendroglial
did not consider IDH mutation status, grade II tumours versus 127.3 months tumours {2464) However, interpreta-
which is prognostically relevant. A more with grade Ill) {1268) In contrast, a ret- tion of these retrospective data requires
recent study confirmed WHO grade II as rospective analysis of 212 patients with caution, because of the potential bias
a favourable prognostic factor independ- I DH-mutant and 1 p/19q-codeleted oli- due to lack of inclusion of prognostically
ent of 1p/19q codeletion in 95 patients godendroglial tumours did not find WHO relevant clinical information (e.g. extent
with oligodendroglial tumours {2256) grade to be a significant predictor of of resection) and variable postoperative
Other authors have reported significantly overall survival {1836). Similarly, recent treatment of the patients. The current

60 Diffuse gliomas
Registry of the United States (CBTRUS), 225 gliomas (2164) Concurrent HHV6A
the adjusted annual incidence rate of oli- infection and diffuse leptomeningeal oli-
godendroglioma is estimated at 0.26 cas- godendrogliomatosis has been reported
es per 100 000 population {1863) The in a 2-year-old boy (2445). and there
incidence rates for anaplastic oligoden- have been reports of isolated cases of
droglioma and oligoastrocytic tumours oligodendroglioma arising in patients
are estimated at 0.11 and 0.21 cases per with immunodeficiency (including pa-
100 000 population, respectively. Oligo- tients with HIV infection) (499) and after

O w ro �
_ Anaplastic oligodendroglioma
� �
Ase at diagnosis
oo ro
dendroglial tumours account for 1.7% of organ transplantation (2271 l. Rare cases
_ Oli1odendroglioma
all primary brain tumours (oligodendro- of oligodendroglioma in association with
Fig. 1.60 Cumulative age distribution (both sexes) gliomas for 1.2% and anaplastic oligo- a demyelinating disease have also been
of 105 cases of !DH-mutant and 1p/19q-codeleted dendrogliomas for 0.5%) and for 5.9% reported (373). However, epidemiologi-
oligodendroglioma (mean patient age at diagnosis:
of all gliomas. Oligoastrocytic gliomas cal data do not indicate an increased
44.3 years) and 126 cases of I DH-mutant and 1 p/19q-
codeleted anaplastic oligodendroglioma (mean age: 45.7 account for 0.9% of all primary brain tu- incidence of gliomas in patients with au-
years). Combined data from the German Glioma Network mours and for 3.3% of all gliomas. toimmune disease (990).
and the University of Heidelberg.
Age and sex distnbution Localization
Epidemiological data based on histo- I DH-mutant and 1 p/19q-codeleted oligo-
WHO classification adheres to the prior logical classification indicate that most of dendrogliomas arise preferentially in the
two-tiered histological grading of oligo- these tumours arise in adults, with peak white matter and cortex of the cerebral
dendroglial tumours, with well-differen- incidence in patients aged 35-44 years hemispheres. The frontal lobe is the most
tiated tumours assigned a WHO grade {1826,1863). Oligodendrogliomas are common location (involved in > 50% of
of II and anaplastic tumours a WHO rare in children, accounting for only 0.8% all patients) followed in order of decreas-
grade of Ill. Future studies should inves- of all brain tumours in patients aged ing frequency by the temporal, parietal,
tigate whether the traditional histological < 15 years and 1.8% in adolescents and occipital lobes. Involvement of more
grading criteria should be modified and/ aged 15-19 years {1863). with paediatric than one cerebral lobe or bilateral tumour
or complemented by molecular marker oligodendrogliomas frequently lacking spread is not uncommon. Rare loca-
assessments, such as CDKN2A dele- IDH mutation and 1p/19q codeletion (see tions include the posterior fossa, basal
tion or TCF12 mutation, which has been 0/igodendroglioma lacking /OH muta- ganglia, and brain stem. Leptomenin-
linked to clinically aggressive behaviour tion and 1p/19q codeletion, p. 68). With geal spread is seen in only a minority of
in IDH-mutant and 1p/19q-codeleted oli- rare exceptions, children with 1 p/19q- patients {2163). There have been rare
godendroglioma (1407). codeleted oligodendroglioma are usually cases of primary leptomeningeal oligo-
older than 15 years at diagnosis (2157). dendrogliomas (1775) or oligodendroglial
Historical annotation Overall, men are affected more frequently gliomatosis cerebri {2489}. Primary oli-
The first description of an oligodendro- than women, with a male-to-female ratio godendrogliomas of the spinal cord are
glioma was published by Bailey and of 1.3:1 (1863). In the USA, oligodendro- rare, accounting for only 1.5% of all oligo-
Cushing (108) in 1926. This publication glioma is more common in Whites than dendrogliomas and 2% of all spinal cord
was followed in 1929 by the classic pa- in Blacks, with an incidence rate ratio of tumours (729). Rare cases of primary
per by Bailey and Bucy (107). "Oligo- 2.51 (1863). spinal intramedullary oligodendroglioma
dendrogliomas of the brain". The asso- and secondary meningeal dissemination
ciation between 1p/19q codeletion and Etiology have been reported, including a 1 p/19q-
oligodendroglial histology was reported Rare cases of oligodendroglial tumours codeleted tumour (898}. Isolated cases
in 1994 (2091) and 1995 (161,1359). fol- diagnosed after brain irradiation for oth- of oligodendroglioma arising in ovarian
lowed in 2008 (118) and 2009 {2810) by er reasons have been documented (49, teratomas have been reported, although
the first reports on IDH mutation. 600). Although oligodendrogliomas and there was no assessment of IDH muta-
oligoastrocytomas are among the most tion and 1 p/19q codeletion status (2592}.
Epidemiology frequent types of CNS tumours induced
Note that these and the following para- experimentally in rats by chemical car- Clinical features
graphs mostly refer to epidemiological cinogens such as ethylnitrosourea and Approximately two thirds of patients pres-
and clinical data based on histological tu- methylnitrosourea, there is no convincing ent with seizures (1680}. Other common
mour classification according to the pre- evidence of an etiological role for these presenting symptoms include headache
vious WHO classification. For the most substances in human gliomas. Similarly, and other signs of increased intracranial
part, this information can be considered the existence of a viral etiology of oligo- pressure, focal neurological deficits, and
to be similar for the newly defined entity dendroglioma is uncertain. Polyomavirus cognitive or mental changes {1446,1845).
of IDH-mutant and 1p/19q-codeleted oli- (SV40, BK virus, and JC virus) genome In older studies, durations of > 5 years
godendroglioma, as well as for oligoden- sequences and proteins have been de- between the onset of symptoms and
droglioma, NOS. tected in oligodendrogliomas (570) and a diagnosis were common, but modern
case of oligoastrocytoma {2099). Howev- neuroimaging has markedly reduced the
Incidence er, other authors found no frequent poly- time to diagnosis {1845).
According to the Central Brain Tumor omavirus sequences in a large series of

Oligodendroglioma, IDH-mutant and 1p/19q-codeleted 61


.,

Fig. 1.61 A Recurrent oligodendroglioma with bilateral, diffuse infiltration of the frontal and temporal lobes. B Small Fig. 1.62 Oligodendroglioma (OL) of the temporal lobe
oligodendroglioma of the medial basal ganglia, compressing the right lateral ventricle. Note the homogeneous cut with infiltration of the hippocampus (HC). Note the zone of
surface. calcification (arrowheads) at the periphery of the lesion.

Imaging structures, brain stem, cerebellum, and cellular tumours. Areas of increased eel-
On CT, /DH-mutant and 1p/19q-codelet- spinal cord {2489) lularity, often in the form of circumscribed
ed oligodendrogliomas usually present nodules, can occur in some otherwise
as hypodense or isodense well-demar- Macroscopy well-differentiated tumours, so wide sam-
cated mass lesions, typically located in Oligodendroglioma usually presents as a pling of resection specimens is required.
the cortex and subcortical white matter. relatively well-defined, soft, greyish-pink However, small biopsies sometimes show
Calcification is common but not diagnos- mass. The tumour is typically located in only scattered oligodendroglioma cells
tic. MRI shows a lesion that is T1-hypoin- the cortex and white matter, leading to infiltrating the brain parenchyma, which
tense and T2-hyperintense The lesion is blurring of the grey matter-white mat- are identifiable by their characteristic nu-
usually well demarcated and shows little ter boundary. Local invasion into the clei and (if the most common IDH muta-
perifocal oedema {2678). Some tumours overlying leptomeninges may be seen. tion is present) by immunostaining for
show heterogeneous features due to in- Calcification is frequent and may impart R132H-mutant IDH1 (see lmmunophe-
tratumoural haemorrhages and/or areas a gritty texture to the tumour. Occasion- notype). Classic oligodendroglioma cells
of cystic degeneration. Gadolinium en- ally, densely calcified areas may present have uniformly round nuclei that are slight-
hancement has been detected in < 20% as intratumoural stones. Zones of cystic ly larger than those of normal oligodendro-
of WHO grade II oligodendrogliomas but degeneration, as well as intratumoural cytes and show an increase in chromatin
in > 70% of WHO grade Ill anaplastic haemorrhages, are common. Rare cases density or a delicate salt-and-pepper
oligodendrogliomas {1260) Contrast with extensive mucoid degeneration look pattern similar to that of neuroendocrine
enhancement in low-grade oligodendro- gelatinous. tumours. A distinct nuclear membrane is
gliomas has been associated with less often apparent. In routinely formalin-fixed
favourable prognosis {2621). Demonstra- Microscopy and paraffin-embedded material, there is
tion of elevated 2-hydroxyglutarate lev- a tendency for the tumour cells to undergo
els by MR spectroscopy is a promising Histopathology degeneration by acute swelling, which re-
new means of non-invasive detection of Oligodendrogliomas are diffusely infil- sults in an enlarged rounded cell with a
!DH-mutant gliomas (including oligoden- trating gliomas of moderate cellularity well-defined cell membrane and clear
drogliomas) {449). Differences in certain that in classic cases are composed of cytoplasm around a central spherical nu-
features between 1 p/19q-codeleted and monomorphic cells with uniform round cleus. This creates the typical honeycomb
1 p/19q-intact low-grade gliomas have nuclei and variable perinuclear haloes on or fried-egg appearance, which although
been reported on MR spectroscopy {292, paraffin sections (a honeycomb or fried- artefactual is a helpful diagnostic feature
685,2678), but reliable discrimination by egg appearance). Additional features in- when present. However, this artefact is not
neuroimaging is not yet possible. clude microcalcifications, mucoid/cystic seen in smear preparations or frozen sec-
degeneration, and a dense network of tions, and may also be absent in rapidly
Spread delicate branching capillaries. Mitotic fixed tissue and in paraffin sections made
!DH-mutant and 1p/19q-codeleted oligo- activity is either absent or low. Nuclear from frozen material.
dendrogliomas characteristically extend atypia and an occasional mitosis are Some oligodendrogliomas contain tu-
into adjacent brain in a diffuse manner. compatible with the diagnosis of a WHO mour cells with the appearance of small
Like other diffuse gliomas, oligodendro- grade II tumour, but brisk mitotic activity, gemistocytes with a rounded belly of
glioma can also (although rarely) mani- prominent microvascular proliferation, eccentric cytoplasm that is positive for
fest at initial clinical presentation with a and spontaneous necrosis are indica- GFAP. These cells have been referred
gliomatosis cerebri pattern of extensive tors of anaplasia, corresponding to WHO to as minigemistocytes or microgemisto-
involvement of the CNS, with the affected grade Ill (see Anaplastic o/igodendrogli- cytes. Gliofibrillary oligodendrocytes are
area ranging from most of one cerebral oma, /DH-mutant and 1p/19q-codeleted, typical-looking oligodendroglioma cells
hemisphere (three lobes or more) to both p. 70). on routine stains but show a thin peri-
cerebral hemispheres with additional nuclear rim of positivity for GFAP {994).
involvement of the deep grey matter Cellular composition GFAP-negative mucocytes or even sig-
Oligodendrogliomas are moderately net ring cells are occasionally seen. Rare

62 Diffuse gliomas
• ..., • ' ..
.
··«'1
� .
-
�'\
.... , -'i' \
... -
Fig. 1.63 I DH-mutant and 1 p/19q-codeleted oligodendroglioma. A Typical honeycomb or fried-egg pattern of oligodendrogliomas: the tumour cells show a clear perinuclear halo
.
• ""

and a sharply delineated plasma membrane; although this feature is an artefact that occurs during tissue processing, it is a hallmark of oligodendroglioma. B Perinuclear clearing.
C Delicate "chicken-wire" network of branching capillaries. Note the moderate nuclear atypia and occasional microcalcification. D Conspicuous network of branching capillaries.

cases of oligodendroglioma consisting astrocytic morphology is also compatible Mineralization and other degenerative
largely of signet ring cells (called sig- with this diagnosis when molecular test- features
net ring cell oligodendroglioma) have ing confirms IDH mutation and 1p/19q A frequent histological feature is the pres-
been described 11373). and eosinophilic codeletion; in other words, diffuse glio- ence of microcalcifications (sometimes
granular cells are present in some oligo- mas with oligoastrocytoma histology or associated with blood vessels) within the
dendrogliomas 12498}. Rare cases with with ambiguous histological features tumour tissue proper or in the invaded
neurocytic or ganglioglioma-like differ- should be diagnosed as I DH-mutant and brain. Mineralization along blood vessels
entiation have also been reported 11939, 1 p/19q-codeleted oligodendroglioma typically takes the form of small, punctate
1948). The presence of these various when molecular testing reveals this en- calcifications, whereas microcalcifica-
cellular phenotypes does not preclude tity-defining genotype !349,2464,2731} tions in the brain (called calcospher-
an oligodendroglioma diagnosis if the Reactive astrocytes are typically scat- ites) tend to be larger, with an irregular
tumour is positive for IDH mutation and tered throughout oligodendrogliomas and sometimes laminated appearance.
1p/19q codeletion. The presence of tu- and may be particularly prominent at the However, this feature is not specific for
mour cells with fibrillar or gemistocytic tumour borders. oligodendroglial tumours, and due to

B OLIG2. C GFAP.

Oligodendroglioma, I DH-mutant and 1 p/19q-codeleted 63


.,

- .)-_ .
Fig. 1.65 I DH-mutant and 1 p/19q-codeleted oligodendroglioma. A Gliofibrillary oligodendrocytes, GFAP stain. B This oligodendroglioma shows an unusually large number of
minigemistocytes - oligodendroglioma cells with a small globular paranuclear area of strongly GFAP-positive cytoplasm.

generally incomplete tumour sampling, an antibody specific for R132H-mutant oligodendrogliomas !1336} GFAP and
is sometimes not found in the available IDH1 j360l. Positive R132H-mutant IDH1 vimentin immunostaining is often posi-
tissue sections even when clearly dem- staining greatly facilitates the immuno- tive in the astrocytic-appearing tumour
onstrated on CT. Areas characterized histochemical differential diagnosis of components of IDH-mutant and 1p/19q-
by extracellular mucin deposition and/ oligodendroglioma versus other clear codeleted oligodendrogliomas that his-
or microcyst formation are frequent. Rare cell tumours of the CNS and non-neo- tologically resemble oligoastrocytoma.
tumours are characterized by marked plastic and reactive lesions !356,357). Cytokeratins are absent, although cer-
desmoplasia !1156). However, a lack of R132H-mutant IDH1 tain antibody cocktails, such as AE1/
immunopositivity does not exclude oli- AE3, may give false-positive staining due
Vasculature godendroglioma, given the possibility of to cross-reactivity j1773l.
Oligodendrogliomas typically show a less common IDH1 and IDH2 mutations Several antigens are specifically ex-
dense network of branching capillaries that cannot be detected with the R132H- pressed by normal oligodendrocytes in
resembling the pattern of chicken wire. In mutant IDH1 antibody and instead re- vivo or in vitro, including myelin basic
some cases, the capillary stroma tends quire DNA sequence analysis. Unlike protein; proteolipid protein; myelin-asso-
to subdivide the tumour into lobules. most IDH-mutant diffuse astrocytomas, ciated glycoprotein; galactolipids, such
There is a tendency for intratumoural I DH-mutant and 1 p/19q-codeleted oli- as galactocerebroside and galactosul-
haemorrhages. godendrogliomas typically retain nuclear fatide; certain gangliosides; and several
expression of ATRX !1519,2105} In addi- enzymes, such as carbonic anhydrase C,
Growth pattern tion, I DH-mutant and 1 p/19q-codeleted CNP, glycerol-3-phosphate dehydroge-
Oligodendrogliomas grow diffusely in oligodendrogliomas usually lack wide- nase, and lactate dehydrogenase. How-
the cortex and white matter. Within the spread nuclear p53 staining, a finding ever, none of these antigens has demon-
cortex, tumour cells tend to form sec- consistent with the mutual exclusivity of strated significance as a diagnostically
ondary structures such as perineuronal TP53 mutation and 1p/19q deletion in useful marker for oligodendrogliomas.
satellitosis, perivascular aggregates, and IDH-mutant gliomas !349,2464). Some are not expressed in oligodendro-
subpial accumulations. Circumscribed Oligodendrogliomas are consistently irn- glioma cells (e.g. myelin basic protein
leptomeningeal infiltration may induce a munopositive for MAP2, S100 protein, !1746)), some are expressed only in a
desmoplastic reaction. A rare spongio- and LEU? !221,1746,2087). MAP2 often minority of cases (e.g. myelin-associated
blastic growth pattern consists of paral- reveals perinuclear cytoplasmic immu- glycoprotein !1746). galactocerebroside,
lel rows of tumour cells with somewhat nostaining without significant process proteolipid protein, and CNP !2457)), and
elongated nuclei forming rhythmic pali- labelling. However, all three markers some have expression that is not restrict-
sades. Occasionally, perivascular pseu- are also commonly positive in astrocytic ed to oligodendroglial tumour cells (e.g.
dorosettes are seen, although some of gliomas. Similarly, the oligodendrocyte carbonic anhydrase C !1747))
these are a result of perivascular neuropil lineage-associated transcription fac- Synaptophysin immunoreactivity of re-
formation within foci of neurocytic differ- tors OLIG1, OLIG2, and SOX10 are ex- sidual neuropil between the tumour cells
entiation !1948). These patterns are gen- pressed in oligodendrogliomas but also is frequently seen in oligodendroglial tu-
erally present only focally. in other gliomas !121,1500). GFAP is mours and should not be mistaken for
detectable in intermingled reactive as- evidence of neuronal or neurocytic dif-
lmmunophenotype trocytes but can also be found in neo- ferentiation. However, IDH-mutant and
To date, no single immunohistochemical plastic cells such as minigemistocytes 1 p/19q-codeleted oligodendroglioma
marker has been found that is specific for and gliofibrillary oligodendrocytes j994, may contain neoplastic cells that express
oligodendroglial tumour cells. The major- 2087). Vimentin is infrequently expressed synaptophysin and/or other neuronal
ity of oligodendrogliomas demonstrate in well-differentiated oligodendroglio- markers, such as NeuN and neurofila-
strong and uniform immunoreactivity with mas but more often found in anaplastic ments !1939,1948} lmmunostaining for

64 Diffuse gliomas
the proneural alpha-internexin protein is are common in !DH-mutant and 1p/19q- diastase-sensitive periodic acid-Schiff
frequent [6071 but cannot substitute as codeleted oligodendrogliomas but positivity, immunoreactivity for EMA and
a reliable surrogate marker for 1 p/19q exceptional in JOH-mutant diffuse as- desmoplakin, and lack of JOH mutation.
codeletion (625). Similarly, NOGO-A trocytomas. The considerable overlap Metastatic clear cell carcinomas dif-
positivity is typical of, but not exclusive between TERT promoter mutation and fer from oligodendrogliomas in that they
to, 1 p/19q-codeleted oligodendroglio- 1p/19q codeletion in JOH-mutant gliomas have sharp tumour borders, are immuno-
rnas [16001 suggests that TERT promoter mutation reactive for cytokeratins and EMA, and
may be useful as a surrogate marker for lack R132H-mutant IDH1 immunostain-
Proliferation 1p/19q codeletion. However. minor sub- ing or other IDH mutations.
Mitotic activity is low or absent in WHO sets of JOH-mutant and 1p/19q-codelet- In adult patients, the differential diagnosis
grade II oligodendrogliomas. Accord- ed oligodendrogliomas have been re- with pilocytic astrocytoma rarely poses a
ingly, the Ki-67 proliferation index is usu- ported to lack TERT promoter mutation, major problem, because foci of classic
ally < 5% (see Prognosis and predictive and some JOH-mutant but 1p/19q-intact pilocytic features are usually present in
factors). On average, the Ki-67 prolifera- astrocytomas may carry TERT promoter pilocytic astrocytomas, and JOH muta-
tion index is significantly lower in WHO mutations (89,1314,1408,2464). There- tion and 1 p/19q deletion are absent.
grade II oligodendrogliomas than in ana- fore. the general use of TERT promoter Neuroradiological features, such as mid-
plastic oligodendrogliomas. However. a sequencing instead of 1 p/19q codeletion line tumour location and mural nodule/
definitive diagnostic cut-off point cannot testing is not recommended. lmmuno- cyst formation, may also provide helpful
be established, due to marked variability histochemical features that may further ancillary information. Molecular demon-
in staining results between institutions. support the differential diagnosis include stration of BRAF fusion genes supports
Nevertheless, interobserver agreement frequent nuclear p53 immunostaining the diagnosis of pilocytic astrocytoma,
on Ki-67 proliferation index scoring as and Joss of nuclear ATRX expression in although BRAF gain and KIAA1549-
determined by MIB1 monoclonal anti- diffuse astrocytomas. BRAFfusions have also been reported in
body staining was good when six pathol- Other neoplastic lesions that can histo- some !DH-mutant and 1p/19q-codeleted
ogists independently reviewed the same logically mimic oligodendroglioma are oligodendroglial tumours (104,1282). In
set of MIB1-stained slides from 30 oligo- clear cell ependymoma, neurocytoma, children, the molecular distinction of oli-
dendrogliomas (20241. Minigemistocytes and dysembryoplastic neuroepithelial godendroglioma from pilocytic astrocy-
are reported to be mostly MIB1-negative tumour. These entities and oligodendro- toma is challenging because paediatric
and thus non-proliferative. whereas gliofi- gliomas share the feature of neoplastic oligodendrogliomas typically lack com-
brillary oligodendrocytes are more com- cells with uniform, round nuclei and clear bined IDH mutation and 1p/19q codele-
monly positive (1371). Other proliferation cytoplasm, collectively referred to as tion but occasionally demonstrate BRAF
markers, such as PCNA (2096). TOP2A oligodendroglial-like cells. In the routine fusion genes (see 0/igodendroglioma
(1344). and MCM2 (2736) have been re- diagnostic setting, evidence of an JOH lacking /OH mutation and 1p/19q codele-
ported to correlate with WHO grade and mutation, typically demonstrated by posi- tion, p. 68). This differential diagnosis
survival in patients with oligodendroglial tive R132H-mutant IDH1 immunostaining, must therefore rely primarily on histologi-
tumours, but do not provide clear advan- rules out all these differential diagnoses. cal and ancillary radiological features,
tages over MIB1 immunostaining in the In the absence of IDH mutation, immu- unless advanced molecular testing
routine setting. nostaining for neuronal markers, in par- methods based on large-scale methyla-
ticular diffuse synaptophysin and at least tion and/or mutation profiling that provide
Differential diagnosis focal NeuN, provides further evidence entity-specific methylation or mutation
The morphological spectrum of JOH-mu- for neurocytoma. Similarly, rare cases of profiles can be applied. The differential
tant and 1p/19q-codeleted oligodendro- liponeurocytoma are distinguished from diagnosis of diffuse leptomeningeal glio-
gliomas is broad, with some similarities oligodendroglioma by extensive positive neuronal tumour (p. 152) is facilitated by
to a variety of reactive and neoplastic le- staining for neuronal markers, the ab- the clinical presentation and the charac-
sions. Macrophage-rich processes such sence of JOH mutation, and the presence teristic molecular profile, with combined
as demyelinating diseases or cerebral in- of lipidized cells resembling adipocytes. KIAA1549-BRAFgene fusion and solitary
farcts should be readily distinguished by Clear cell ependymomas often show at 1p deletion (or 1p/19q codeletion) but ab-
immunostaining for macrophage mark- least focal perivascular pseudorosettes sence of JOH mutation (21561
ers and lack of IDH mutation. Reactive and dot-like or ring-shaped EMA im-
changes such as the increased numbers munoreactivity. Formation of specific Cell of origin
of oligodendrocytes sometimes seen in glioneuronal elements, absence of IDH Although the designation of CNS neo-
partial lobectomy specimens performed mutation and 1 p/19q codeletion, and (in plasms as oligodendroglial tumours
for intractable seizures can also be dis- a subset) eosinophilic granular bodies, could imply histogenesis from cells of the
tinguished by lack of JOH mutation. a CD34-positive cell population, and/or oligodendroglial lineage, the evidence
The differential diagnosis with diffuse BRAF V600E mutation, distinguish dys- supporting this assumption is circum-
astrocytoma relies on histological, immu- embryoplastic neuroepithelial tumour stantial, based solely on morphological
nohistochemical, and molecular features. from !DH-mutant and 1p/19q-codeleted similarities of the neoplastic cells in these
Most importantly, !DH-mutant diffuse oligodendroglioma (414). A rare differen- tumours to normal oligodendrocytes. It
astrocytomas lack 1 p/19q codeletion. tial diagnosis is clear cell meningioma, is also unknown whether human oligo-
In addition, TERT promoter mutations which can be distinguished by abundant dendrogliomas arise from neoplastic

Oligodendrogl1oma, JOH-mutant and 1p/19q-codeleted 65


.,

Fig. 1.66 I DH-mutant and 1 p/19q-codeleted oligodendroglioma. A,B lmmunohistochemistry showing the expression of R132H-mutant IDH1 protein. C Retained expression of ATRX
in tumour cell nuclei.

transformation of mature oligodendro- codon 172 mutations are present, with IDH-mutant and 1p/19q-codeleted oligo-
cytes, immature glial precursors, or the proportion of IDH2 mutations being dendrogliomas lack ATRX mutation but
neural stem cells. Experimental data in higher in oligodendroglial gliomas than virtually always carry activating muta-
transgenic mice indicate that gliomas in astrocytic gliomas {953). Combined tions in the TERT promoter region, lead-
with oligodendroglial histology may origi- whole-arm loss of 1 p and 19q is invaria- ing to increased expression of TERT {89,
nate from different cell types in the CNS, bly associated with IDH mutation, indicat- 1270,1314). In fact, TERTpromoter muta-
including neural stem cells, astrocytes, ing that detection of 1 p/19q codeletion in tion is strongly associated with 1 p/19q
and oligodendrocyte precursor cells the absence of IDH mutation should raise codeletion in IDH-mutant gliomas and
{2872). An oligodendroglioma-like phe- suspicion of incomplete/partial deletions, is an early event in oligodendroglioma
notype is commonly found in transgenic which have been detected in subsets of development {2464) However, TERT
brain tumours, despite a variety of target- IDH-wildtype anaplastic astrocytomas promoter mutations are also frequent in
ed cell types and oncogenic events {589, and glioblastomas, with associated poor IDH-wildtype glioblastomas {1270). Con-
2725). Some studies have suggested a outcome {2658). sequently, large-scale sequencing stud-
likely origin of oligodendrogliomas from ies have identified three major groups
NG2-positive and asymmetrical cell divi- Aberrant genes on 1p or 19q of cerebral gliomas, with distinct biolo-
sion-defective oligodendroglial progeni- Oligodendroglial tumours have frequent gies and clinical outcomes. These three
tor cells {1954,2449). However, oligo- mutations in the human homologue of groups are defined, respectively, by IDH
dendroglial precursor cells may give rise the Drosophila capicua gene (CIC) on mutation associated with 1p/19q codele-
to either oligodendroglial or astrocytic 19q13.2 {183,2825). with the majority of tion and TERT promoter mutation, IDH
gliomas, depending on the genes driv- IDH-mutant and 1p/19q-codeleted oli- mutation associated with TP53 and fre-
ing transformation {1508}, suggesting a godendrogliomas harbouring CIC muta- quent ATRX mutation, and IDH-wildtype
dominance of oncogenic signalling over tions {2219,2464,2825). A smaller subset status associated with TERT promoter
cell of origin in determining the glioma of these tumours also carries mutations mutation and glioblastoma-associated
phenotype. in the FUBP1 gene on 1p31.1. One study genomic aberrations {349,2464).
identified IDH mutation, 1p/19q codele-
Genetic profile tion, and TERTpromoter mutation as ear- Other genetic alterations
ly genetic changes in oligodendroglioma Mutations in NOTCH1, and less com-
Cytogenetics pathogenesis, whereas CIC mutations monly in other NOTCH pathway genes,
Cytogenetic studies of oligodendro- may appear later in tumour progression have been detected in approximately
glioma have revealed an unbalanced {2464). Other genes on 1p (e.g. CAMTA1, 15% of oligodendrogliomas {349,2464).
translocation between chromosomes 1 CHD5, CITED4, DFFB, DIRAS3, PROX1, Other less commonly mutated genes in-
and 19 that results in loss of the der(1; 19) ATRX, AJAP1, and TP73) and 19q (e.g. clude epigenetic regulator genes such
(p10;q10) chromosome, causing whole- EMP3, ARHGAP35, PEG3, and ZNF296) as SETD2 and other histone methyltrans-
arm deletions of 1p and 19q, and reten- have been reported to show aberrant ferase genes, as well as genes encoding
tion of the der[t(1;19)(q10;p10)] chromo- promoter methylation and/or reduced components of the SWI/SNF chromatin-
some {887,1151). expression in IDH-mutant and 1p/19q- remodelling complex {349,2464). Unlike
codeleted oligodendrogliomas {2126). in IDH-mutant astrocytic tumours, TP53
IDH mutation and 1p/19q codeletion Epigenetic silencing of the pH regulator mutation is usually absent and mutu-
The entity-defining alteration in oligo- gene SLC9A1 on 1p has been linked to ally exclusive with 1p/19q deletion {349,
dendrogliomas is concurrent mutation lower intracellular pH and attenuation of 2464}
of IDH1 or IDH2 and whole-arm deletion acid load recovery in oligodendroglioma
of 1p and 19q. The vast majority(> 90% cells, which may contribute to the distinc- Epigenetic and transcriptional changes
{953)) of IDH mutations in WHO grade II tive biology of I DH-mutant and 1 p/19q- I DH-mutant and 1 p/19q-codeleted oli-
oligodendrogliomas are the IDH1 R132H codeleted oligodendrogliomas {217). godendrogliomas are characterized by
mutation, which is readily detectable by widespread changes in DNA methylation,
immunohistochemistry {360). In < 10% TERT promoter mutations leading to concurrent hypermethylation
of cases, other IDH1 codon 132 or IDH2 Unlike IDH-mutant diffuse astrocytomas, of multiple CpG islands, a phenomenon

66 Diffuse gliomas
that is referred to as G-CIMP {1810). and/or maturation of oligodendroglial child with hereditary non-polyposis colo-
Mechanistically, this phenomenon has cells {2085). PDGFA and PDGFB, as well rectal cancer and oligodendroglioma
been linked with mutant IDH proteins as the corresponding receptors (PDG- (1642); and an adolescent with Lynch
producing 2-hydroxyg(�tarate,. _which FRA and PDGFRB) are commonly co- syndrome and anaplastic oligodendro-
functions as a competitive inhibitor of expressed in oligodendrogliomas {582} glioma (978). One child with retinoblas-
alpha-ketoglutarate-dependent dioxy- However, PDGFRA mutations are rare, toma syndrome and anaplastic oligoden-
genases including histone demethylases and elevated expression seems to be droglioma {16) and identical twins with
and the TET family of 5-methylcytosine independent of 1p/19q codeletion {955) Oiiier-type multiple enchondromatosis
hydroxylases {1540,2804). This in turn The functional role of PDGFB has been and oligodendroglioma have also been
leads to increased histone methylation demonstrated by the induction of oligo- documented {411}.
and G-CIMP {1810,2589). One study dendrogliomas in transgenic mice with
suggested that CIC mutations cooper- targeted overexpression of this growth Prognosis and predictive factors
ate with IDH mutations to further increase factor in neural stem or progenitor cells
2-hydroxyglutarate levels (443). In fact, {521,1508). VEGF and its receptors serve Prognosis
DNA methylation profiles in IDH-mutant as angiogenic factors in oligodendroglial WHO grade II IDH-mutant and 1p/19q-
and 1 p/19q-codeleted oligodendroglio- tumours, particularly in anaplastic oligo- codeleted oligodendrogliomas are typi-
mas differ from those in IDH-mutant but dendrogliomas {402,456). cally slow-growing tumours and are asso-
1p/19q-intact astrocytomas {1723,2464, ciated with relatively long overall survival.
2751). a distinction that has been referred Genetic susceptibility A population-based study from Switzer-
to as G-CIMP type A versus G-CIMP type Most oligodendrogliomas develop spo- land demonstrated a median survival
B (2464). and can be exploited for clas- radically, in the absence of an obvi- time of 11.6 years for oligodendroglioma
sification purposes (e.g. by 450k meth- ous familial clustering or a hereditary and 6.6 years for oligoastrocytoma (both
ylation bead array analysis {2751 )). As a predisposition syndrome. Large-scale defined according to histological criteria
consequence of G-CIMP, many different genotyping data indicate a strong asso- only), as well as 10-year survival rates of
genes may be epigenetically inactivated ciation between a low-frequency SNP at 51% and 49%, respectively (1826). The
in oligodendrogliomas, including genes 8q24.21 and increased risk of !DH-mu- CBTRUS has documented 5-year sur-
on 1p and 19q as well as genes on other tant oligodendroglioma and astrocytoma vival rates of 79.5% and 61.1%, as well
chromosomes, such as the tumour sup- {1152). Other genetic polymorphisms that as 10-year survival rates of 62.8% and
pressors COKN2A, COKN2B, RB1, and have been associated with increased oli- 46.9%, for oligodendroglioma and oli-
many others (2126). MGMT promoter hy- godendroglioma risk include the GSTT1 goastrocytic glioma, respectively (1863).
permethylation and reduced expression null genotype {1245) and SNPs in the However, survival estimates have varied
is also common {1733). At the mRNA lev- GLTSCR1 and ERCC2 genes {2814). markedly. Some single-institution studies
el, I DH-mutant and 1p/19q-codeleted oli- Germline mutations in shelterin com- have documented even longer median
godendrogliomas often show a proneural plex genes, including the POT1 gene, overall survival times (e.g. > 15 years
glioblastoma-like gene expression signa- have been linked to familial oligoden- (1845)) for patients with low-grade oli-
ture (608,2731}. droglioma (110). The largest series of godendroglioma, and others have docu-
non-syndromic familial glioma cases mented shorter median survivals (e.g.
Growth factors and receptors published to date comprised 841 pa- 3.5 years (567)). Most of the variability
About half of all WHO grade II oligoden- tients from 376 families with � 2 affected in these studies is likely due to differing
drogliomas and anaplastic oligodendro- family members each {2212) Within this diagnostic criteria, the absence of mo-
gliomas show strong expression of EGFR cohort, 59 patients (8%) were diagnosed lecular information on IDH mutation and
mRNA and protein in the absence of with WHO grade II oligodendroglioma, 1 p/19q codeletion, and differing treat-
EGFR gene amplification {2090). The si- 29 patients (3.9%) with WHO grade II ment approaches.
multaneous expression of the mRNAs for oligoastrocytoma, 31 patients (4.2%) with Oligodendrogliomas generally recur lo-
the pre-pro forms of EGFR and/or trans- WHO grade Ill anaplastic oligodendro- cally. Malignant progression of recur-
forming growth factor alpha indicates the glioma, and 12 patients (1.6%) with WHO rence is common, although it takes
possibility of auto-, juxta-, or paracrine grade Ill anaplastic oligoastrocytoma. longer on average than in diffuse astro-
growth stimulation via the EGFR system Isolated cases of familial oligodendro- cytomas (1121}. Rare cases of gliosar-
(6261 One study reported that high EGFR glioma with 1p/19q codeletion have also coma (so-called oligosarcoma) arising
expression was linked to shorter survival been reported (713,1858). from I DH-mutant and 1 p/19q-codeleted
in patients with 1 p/19q-codeleted oligo- Only a few patients with oligodendroglial oligodendroglioma have been reported
dendrogliomas of WHO grade II but long- tumours have been reported in families (1004,2152). Unusual cases of oligoden-
er survival in patients with WHO grade Ill with hereditary cancer predisposition droglioma, including 1 p/19q-codeleted
anaplastic oligodendrogliomas {1036). syndromes. These include one patient tumours, have been noted in which the
Several other growth factors (including manifesting BRCA 1 mutation with oligo- patients developed systemic metasta-
basic fibroblast growth factor, platelet- dendroglioma {1165); one patient with ses, usually at late stages of the disease
derived growth factors, transforming Turcot syndrome, germline PMS2 muta- {1148,1651}. It has been suggested that
growth factor beta, IGF1, and nerve tion, and two metachronous glioblas- oligodendroglial tumours with 1p/19q
growth factor) have been reported to be tomas showing histological features of codeletion may be more prone to extra-
involved in the regulation of proliferation oligodendroglial differentiation (2525); a neural metastasis despite their generally

Oligodendroglioma, !DH-mutant and 1p/19q-codeleted 67


favourable prognosis {1651}, but this hy- Oligodendroglioma lacking the other 50 cases showed histological
pothesis remains unproven. IDH mutation and 1p/19q codeletion features of other entities, such as pilo-
(paediatric-type oligodendroglioma) cytic astrocytoma, dysembryoplastic
Clinical factors A small subset of histologically classic neuroepithelial tumour, and oligoas-
Features that have been associated oligodendrogliomas are found to lack trocytoma {2157). Of the 50 confirmed
with more favourable outcome include IDH mutation and 1p/19q codeletion cases of oligodendroglioma with classic
younger patient age at operation, loca- on appropriate molecular testing. This histology, 38 tumours were low-grade
tion in the frontal lobe, presentation with group includes the majority of oligoden- and 12 tumours were anaplastic. All
seizures. high postoperative Karnofsky drogliomas in children and adolescents 50 cases were diffusely infiltrating glio-
score, lack of contrast enhancement {1361,2057,2157). In these cases, it is mas composed of uniform round cells
on neuroimaging, and macroscopically important to check carefully for and ex- with perinuclear haloes and formation
complete surgical removal (2621 ). One clude histological mimics that may con- of secondary structures (predominantly
study found that a greater extent of re- tain oligodendrocyte-like tumours cells, perineuronal satellitosis). Calcifications
section was associated with longer over- in particular dysembryoplastic neuro- and microcysts were also frequent.
all and progression-free survival, but did ectodermal tumour, extraventricular However, most tumours lacked /OH1
not prolong the time to malignant pro- neurocytoma, clear cell ependymoma, R132H and 1p/19q codeletion, and his-
gression (23861. and pilocytic astrocytoma (see Differ- tological progression was rare {2157).
ential diagnosis). However, differential High-throughput molecular profiling of
Histopathology diagnosis can be difficult, because in- paediatric low-grade diffuse gliomas
The histological features that have been dividual tumours can show overlapping revealed duplications of portions of the
linked to worse prognosis include ne- histological features of related tumours, FGFR1 gene or rearrangements of MYB
crosis, high mitotic activity, increased such as oligodendroglioma, angiocen- in > 50% of cases, including tumours
cellularity, nuclear atypia, cellular pleo- tric glioma, and dysembryoplastic with oligodendroglial or oligoastrocytic
morphism, and microvascular prolifera- neuroepithelial tumour {1257). More- histology {2855). Rearrangements in
tion (see Anap!astic o!igodendroglioma, over, molecular studies indicate that a the MYB-related MYBL 1 transcription
!DH-mutant and 1p/19q-codeleted, subset of paediatric oligodendroglio- factor gene have also been reported
p. 70). However, the prognostic signifi- mas carry the oncogenic BRAF fusion {2068). These findings indicate that the
cance of each of these histological fea- genes {1391) that are typically detected majority of diffuse gliomas in children,
tures requires re-evaluation in patients in pilocytic astrocytoma {1176). Thus, including tumours with oligodendroglial
with molecularly characterized !DH-mu- paediatric low-grade gliomas seem to or oligoastrocytic histology, are geneti-
tant and 1p/19q-codeleted tumours. The constitute an overlapping spectrum of cally and biologically distinct from their
presence of minigemistocytes and/or entities that can be difficult to distin- adult counterparts. However, additional
gliofibrillary oligodendrocytes does not guish by histological means alone. A studies are needed to comprehensively
seem to influence survival (1374). Similar- careful review of 100 tumours originally characterize the molecular profile of
ly, marked desmoplasia does not relate classified as oligodendrogliomas in these rare tumours and to clarify wheth-
to distinct outcome (1156) It is assumed children and adolescents confirmed the er they constitute a distinct entity.
that an astrocytic-looking tumour compo- diagnosis for only 50 tumours, whereas
nent within an I DH-mutant and 1 p/19q-
codeleted oligodendroglioma does not an independent prognostic value of the 1216), but others did not find longer
indicate shorter survival, but this remains Ki-67 proliferation index {604), whereas progression-free survival when patients
to be confirmed. more recent data from patients with ana- were not treated with upfront radiother-
plastic oligodendroglioma have shown a apy or chemotherapy (951 I. Similarly,
Proliferation prognostic impact of the index on univari- neither IDH mutation nor MGMT pro-
Several single-institution studies have ate but not multivariate analysis (2034) moter methylation were linked to longer
found that a higher Ki-67 prolifera- Among paediatric oligodendrogliomas, progression-free survival in patients with
tion index, typically > 3-5%, correlates the Ki-67 proliferation index is higher WHO grade II glioma treated by neuro-
with worse prognosis in patients with in WHO grade Ill tumours than in WHO surgical resection only (27,951). In con-
oligodendroglial tumours. One study, grade II tumours {2157), but a study of 20 trast, IDH mutation, 1p/19q codeletion,
of 32 patients with WHO grade 11 oligo- paediatric low-grade oligodendroglioma and MGMT promoter methylation were
dendrogliomas, found that a Ki-67 prolif- cases did not show a prognostic value found to be associated with better thera-
eration index of > 3% was indicative of for the Ki-67 proliferation index (259l peutic response and longer survival in
a worse prognosis (980). Another study, patients with low-grade glioma treated
of 89 patients with oligodendroglioma, Genetic alterations with adjuvant radiotherapy or chemo-
reported a 5-year survival rate of 83% The prognostic versus predictive role of therapy {951,1256} A study of 360 pa-
for patients whose oligodendrogliomas 1p and 19q loss in WHO grade II gliomas tients with WHO grade II diffuse glioma
had a Ki-67 proliferation index of < 5%, is a matter of debate. Some studies sug- demonstrated no prognostic role for IDH
versus a rate of only 24% with a Ki-67 gested that WHO grade II gliomas with mutation, whereas 1p/19q codeletion
proliferation index of > 5% (567). This deletion of 1 p or with 1 p/19q codeletion was associated with longer overall sur-
finding is similar to those of other studies are associated with longer survival in- vival and TP53 mutation with shorter
(493l In general, older studies reported dependent from adjuvant therapy {686, overall survival {1281 ). Thus, it is likely

68 Diffuse gliomas
that a prognostically favourable role of patients treated with radiotherapy
!DH mutation, 1p/19q losses, and MGMT plus PCV chemotherapy versus radio- Oligodendroglioma, NOS
promoter methylation in WHO grade II therapy alone (2338). However, recent
glioma is linked to higher sensitivity to cy- long-term follow-up data also showed a Definition
totoxic treatment rather than to generally major increase in overall survival after A diffusely infiltrating glioma with classic
more indolent behaviour independent radiotherapy plus PCV chemotherapy, oligodendrog/ial histology, in which mo-
of therapy. It remains to be investigated in particular for patients with 1 p/19q- lecular testing for combined /OH muta-
whether genetic or epigenetic alterations codeleted low-grade oligodendroglio- tion and 1p/19q codeletion could not be
in other genes can improve prognostic mas {2481,2612) Adjuvant chemother- completed or was inconclusive.
assessment within the group of patients apy with temozolomide may also be a The diagnosis of oligodendroglioma,
with !DH-mutant and 1p/19q-codeleted feasible therapeutic strategy for pa- NOS, is reserved for diffusely infiltrat-
WHO grade 11 oligodendrogliomas. tients with progressive low-grade oligo- ing WHO grade II gliomas with clas-
dendroglioma, with 1p/19q codeletion sic oligodendroglial histology but with-
Predictive factors suggested as a predictive marker for out confirmation of IDH mutation and
The optimal postoperative treatment of better response to temozolomide {1022, 1p/19q codeletion, due to limited tissue
patients with !DH-mutant and 1p/19q- 1256,1476). Another study reported that availability, low tumour-cell content, in-
codeleted WHO grade 11 oligodendro- IDH mutation predicts better response conclusive test results. or other circum-
gliomas is a matter of ongoing discus- to radiochemotherapy in patients with stances impeding molecular testing. In
sion After tumour resection, radiotherapy WHO grade II glioma (1835) Collec- general, molecular testing for IDH muta-
and chemotherapy are often deferred tively, these findings agree with data tion and 1 p/19q codeletion is important
until tumour progression because ther- from phase Ill trials in patients with ana- for WHO classification of oligodendro-
apy-associated neurotoxicity is a major plastic gliomas (WHO grade Ill) that in- glial tumours, implying that the diagnosis
concern in patients with expected long- dicated IDH mutation {346) and 1p/19q of oligodendroglioma, NOS, should be
term survival Patients with symptomatic codeletion as predictive markers for limited to a small minority of cases. lm-
residual and progressive tumours after long-term survival after combined treat- munohistochemical demonstration of
surgery usually receive upfront treatment ment with radiotherapy and PCV chem- IDH mutation (in particular IOH1 R132H)
with radiotherapy and/or chemotherapy. otherapy {344,2615). The presence of and nuclear positivity for ATRX support
The European Organisation for Research MSH6 mismatch repair gene mutations the diagnosis. However, unless success-
and Treatment of Cancer (EORTC) 22845 has been linked to temozolomide resist- fully tested for 1 p/19q codeletion, glio-
trial showed that adjuvant radiotherapy ance independent of MGMT promoter mas with oligodendroglial histology, JOH
prolonged progression-free but not over- methylation status {1779). One study re- mutation, and nuclear ATRX positivity still
all survival in patients with progressive ported that 1 p/19q codeletion predict- correspond to the diagnosis of oligoden-
WHO grade II gliomas (2613) The Ra- ed a lower risk of pseudoprogression in droglioma, NOS. lmmunohistochemical
diation Therapy Oncology Group (RTOG) patients with oligodendroglial tumours positivity for oligodendroglioma-associ-
9802 trial initially also showed only in- despite being associated with longer ated markers such as alpha-internexin
creased progression-free survival for survival (1504l. {607,625) and NOGO-A {1600). as well
as immunohistochemical demonstration
of loss of nuclear CIC or FUBP1 expres-
sion [146,401). are not sufficient to substi-
tute for 1p/19q codeletion testing.
Unlike with IDH-mutant and 1p/19q-
codeleted oligodendroglioma, the pres-
ence of a conspicuous astrocytic compo-
nent is not compatible with the diagnosis
of oligodendroglioma, NOS (see 0/igoas-
trocytoma, NOS, p. 75).

ICD-0 code 9450/3

Grading
Oligodendroglioma, NOS, corresponds
histologically to WHO grade II.

Oligodendroglioma, IDH-mutant and 1p/19q-codeleted 69


Anaplastic oligodendroglioma, Reifenberger G.
Collins VP
Cairncross J.G.
Yokoo H.
IDH-mutant and 1p/19q-codeleted Hartmann C.
Hawkins C.
Yip S.
Louis D.N.
Kras J.M.

Definition analyses that do not carefully control for Epidemiology


An /OH-mutant and 1p/19q-codeleted different treatments and other prognosti- Note that these and the following para-
oligodendroglioma with focal or diffuse cally relevant parameters. Histological graphs mostly refer to epidemiological
histological features of anaplasia (in par- features that have been linked to high- and clinical data based on histological
ticular, pathological microvascular prolif- grade malignancy of oligodendroglioma tumour classification according to the
eration and/or brisk mitotic activity). in previous studies are high cellularity, previous WHO classification. For the
In IDH-mutant and 1p/19q-codeleted marked cytological atypia, high mitotic most part, this information can be con-
anaplastic oligodendroglioma, necro- activity, pathological microvascular pro- sidered to be similar for the newly defined
sis (with or without palisading) may be liferation, and necrosis with or without entity IDH-mutant and 1p/19q-codeleted
present, but does not indicate progres- palisading. Anaplastic oligodendroglioma anaplastic oligodendroglioma, as well as
sion to glioblastoma. The presence of an usually shows several of these features. for anaplastic oligodendroglioma, NOS.
astrocytic tumour component is compat- However, the individual impact of each
ible with the diagnosis when molecular parameter is unclear, in particular be- Incidence
testing reveals combined IDH mutation cause most of the older studies were not According to a statistical report from
and 1p/19q codeletion. The vast major- restricted to patients with I DH-mutant and the Central Brain Tumor Registry of the
ity of IDH-mutant and 1p/19q-codeleted 1 p/19q-codeleted oligodendroglial tu- United States (CBTRUS), anaplastic oli-
anaplastic oligodendrogliomas manifest mours. Endothelial (microvascular) prolif- godendroglioma (as defined by histology
in adult patients. The tumours are pref- eration and brisk mitotic activity (defined only) has an estimated annual incidence
erentially located in the cerebral hemi- as ;:,: 6 mitoses per 10 high-power fields) rate of 0.11 cases per 100 000 popula-
spheres, with the frontal lobe being the have been suggested to be of particular tion and accounts for 0.5% of all primary
most common location. importance as indicators of anaplasia in brain tumours {1863). Approximately one
oligodendroglial tumours {830). Thus, third of all oligodendroglial tumours are
ICD-0 code 9451/3 the diagnosis of IDH-mutant and 1p/19q- anaplastic oligodendrogliomas {1863).
codeleted anaplastic oligodendroglioma
Grading should require at least the presence of Age and sex distribution
I DH-mutant and 1 p/19q-codeleted ana- conspicuous microvascular proliferation Anaplastic oligodendrogliomas manifest
plastic oligodendroglioma corresponds and/or brisk mitotic activity. Detection of preferentially in adults, with a median pa-
histologically to WHO grade Ill. a single mitosis in a resection specimen tient age at diagnosis of 49 years {1863).
There is clear evidence from prospective is not sufficient for classifying an oligo- Patients with anaplastic oligodendroglio-
clinical trials {344,2615,2740) and large dendroglial tumour as WHO grade Ill mas are approximately 6 years older on
cohort studies {2464,2731) that patients anaplastic oligodendroglioma. In border- average than patients with WHO grade II
with IDH-mutant and 1p/19q-codeleted line cases, immunostaining for MIB1 and oligodendrogliomas, but this difference
anaplastic oligodendroglial tumours have attention to clinical and neuroradiologi- appears to be much smaller when only
a significantly better prognosis than do pa- cal features, such as rapid symptomatic patients with IDH-mutant and 1p/19q-
tients with I DH-mutant but 1 p/19q-intact growth and contrast enhancement, may codeleted tumours are considered. Ana-
or IDH-wildtype anaplastic astrocytic gli- provide additional information for assess- plastic oligodendroglioma is very rare in
omas. In contrast, the prognostic value of ing the prognosis. children. In the CBTRUS, there were no
WHO grading within the group of patients
with IDH-mutant and 1p/19q-codeleted
oligodendroglial tumours is less clear
(see 0/igodendroglioma, /OH-mutant and
1p/19q-codeleted, p. 60) Two recent un-
controlled retrospective studies suggest-
ed a limited role of WHO grading in pre-
dicting patient outcome {1836,2464). a
finding in line with retrospective data ob-
tained from a large cohort of patients with
IDH-mutant astrocytic gliomas {2103).
However, treatment regimens often differ
for patients with low-grade versus high- Fig. 1.67 IDH-mutant and 1p/19q-codeleted anaplastic oligodendroglioma. T1-hypointense lesion with focal contrast
grade oligodendroglioma, so potential enhancement following gadolinium administration (+Gd). T2 (FLAIR) shows the extent of the lesion and FET-PET
bias cannot be excluded in retrospective demonstrates increased metabolic activity.

70 Diffuse gliomas
patients with anaplastic oligodendro- {2616). Seizures are also common but
glioma among the 12 103 children aged are less frequent than in patients with
o-14 years registered with neuroepithe- low-grade oligodendroglioma (933)
lial tumours, and only 32 patients with Anaplastic oligodendroglioma develops
anaplastic oligodendroglioma were doc- either de novo (typically with a short pre-
umented among the 3051 children regis- operative history) or by progression from
tered with neuroepithelial tumours in the a pre-existing WHO grade 11 oligoden-
15-19 years age group (1863). In an in- droglioma. The mean time to progression
stitutional cohort of 50 paediatric patients from WHO grade II oligodendroglioma to
with oligodendroglioma, 12 patients were WHO grade Ill anaplastic oligodendro-
diagnosed with anaplastic oligodendro- Fig. 1.68 Large, macroscopically well-delineated glioma has been estimated at approxi-
gliomas, including 4 patients with 1p/19q anaplastic oligodendroglioma of the left basal ganglia, mately 6-7 years (1446,1826).
codeletion (2157). compressing the adjacent lateral ventricle, with shift of
Overall, anaplastic oligodendroglioma midline structures towards the right hemisphere. The Imaging
shows a slight male predominance, with arrowhead points to a ventricular shunt. Anaplastic oligodendrogliomas can
a male-to-female ratio of 1.2:1 reported show heterogeneous patterns, due to the
among 1650 patients (1863). In the USA, a preference for the frontal lobe, followed variable presence of necrosis, cystic de-
anaplastic oligodendroglioma is more by the temporal lobe. However, the tu- generation, intratumoural haemorrhages,
common in Whites than in Blacks, with an mours can also originate at other sites and calcification. On CT and MRI, con-
incidence ratio of 2.4:1 (1863). within the CNS, and there have been rare trast enhancement is seen in most pa-
cases of spinal intramedullary anaplastic tients and can be either patchy or ho-
Etiology oligodendroglioma. mogeneous (2616). However, lack of
See Etiology (p. 61) in Oligodendroglio- contrast enhancement does not exclude
ma, JOH-mutant and 1p/19q-codeleted. Clinical features anaplastic oligodendroglioma. Ring en-
Patients with anaplastic oliqodendro- hancement is less common in IDH-mu-
Localization glioma commonly present with focal tant and 1p/19q-codeleted anaplastic
Anaplastic oligodendrogliomas and neurological deficits, signs of increased oligodendrogliomas than in malignant
WHO grade 11 oligodendrogliomas share intracranial pressure, or cognitive deficits

- - ,:·. . . ...� �.
Fig. 1.69 Anaplastic oligodendroglioma. A Typical image of a cellular glioma with honeycomb cells and mitotic activity (arrows). B Marked nuclear atypia and brisk mitotic activity.
C Focal necrosis with palisading tumour cells. D Marked microvascular proliferation.

Anaplastic oligodendroglioma, IDH-mutant and 1p/19q-codeleted 71


,

gliomas, in particular glioblastomas with- WHO grade Ill IDH-mutant and 1p/19q- Cell of origin
out these molecular markers (345,2616l. codeleted anaplastic oligodendroglioma See Cell of origin (p. 65) in 0/igodendrogli-
is appropriate. The presence of a con- oma, /OH-mutant and 1p/19q-code/eted
Macroscopy spicuous astrocytic component is also
The macroscopic features are similar to compatible with this diagnosis provid- Genetic profile
those of WHO grade II oligodendroglio- ing the tumour demonstrates IDH muta-
mas, except that anaplastic oligoden- tion and 1p/19q codeletion. However, in Cytogenetics
drogliomas may show areas of tumour cases that cannot be conclusively tested The hallmark cytogenetic alteration is
necrosis. for combined IDH mutation and 1p/19q combined whole-arm deletion of 1p and
codeletion, the presence of a conspicu- 19q, typically as a consequence of an
Microscopy ous astrocytic component is not compat- unbalanced translocation between chro-
Anaplastic oligodendrogliomas are cel- ible with anaplastic oligodendroglioma, mosomes 1 and 19 (887,1151l Concur-
lular, diffusely infiltrating gliomas that NOS (see Anaplastic oligodendroglio- rent polysomy of 1 p and 19q seems to be
may show considerable morphological ma, NOS, p. 74). Instead, classification more common in anaplastic oligodendro-
variation. The majority of the tumour cells as anaplastic oligoastrocytoma, NOS, gliomas than in low-grade oligodendro-
typically demonstrate features that are or glioblastoma is more appropriate, de- gliomas and has been associated with a
reminiscent of oligodendroglial cells, i.e. pending on the absence or presence of higher Ki-67 proliferation index and less
rounded hyperchromatic nuclei, perinu- necrosis. favourable clinical outcome (216,2748l
clear haloes, and few cellular processes. Molecular cytogenetic studies have iden-
Focal microcalcifications are often pres- lmmunophenotype tified various chromosomal abnormalities
ent. Mitotic activity is usually prominent, Anaplastic oligodendrogliomas have the other than 1 p/19q codeletion in anaplas-
with one study suggesting a cut-off point same immunoprofile as WHO grade II oli- tic oligodendrogliomas; however, each
of 6 mitoses per 10 high-power fields godendroglioma, except that proliferative was limited to only a small proportion of
(830l. Occasional anaplastic oligoden- activity, as most commonly determined tumours. These include gains on chro-
drogliomas with 1p/19q codeletion are by MIB1 immunostaining, is generally mosomes 7, 8q, 11q, 15q, and 20, as well
characterized by marked cellular pleo- higher (i.e MIB1-positive cells usually as losses on chromosomes 4q, 6, 9p,
morphism, with multinucleated giant cells account for > 5% of the tumour cells). 10q, 11, 13q, 18, and 22q (1084,1353,
(called the polymorphic variant of Zulch) However, a definitive MIB1 cut-off point 2577). Double-minute chromosomes, a
(1000l. Rare cases with sarcoma-like for distinction between WHO grade II and cytogenetic hallmark of gene amplifica-
areas (called oligosarcoma) have also 111 oligodendrogliomas has not been es- tion, are rare (2537l, corresponding to
been observed (1004,2152l Gliofibrillary tablished (see lmmunophenotype, p. 64,
oligodendrocytes and minigemistocytes in 0/igodendroglioma, /OH-mutant and
are frequent in some anaplastic oligo- 1 p/19q-codeleted). Table 1.07 Genetic profile of IDH-mutant and 1p/19q-
dendrogliomas (1374l. The characteristic codeleted anaplastic oligodendroglioma
vascular pattern of branching capillaries Differential diagnosis %of
Gene Change References
is often still recognizable, although path- The differential diagnosis includes a va- cases
ological microvascular proliferation is riety of other clear cell tumour entities IDH1/
Mutation 100% (349,2464)
usually prominent. Formation of second- that can be distinguished by distinct im- IDH2
ary structures, in particular perineuronal munohistochemical profiles and absence 1p/19q Codeletion 100% (349,2464)
satellitosis, is frequent in areas of cortical of combined IDH mutation and 1p/19q
Promoter
tumour infiltration. Anaplastic oligoden- codeletion (see Differential diagnosis, TERT > 95% (349,2464)
mutation
drogliomas may contain necrosis, includ- p. 65, in 0/igodendroglioma, /OH-mutant
ing palisading necrosis resembling that of and 1p/19q-codeleted) Distinction of Promoter
MGMT > 90% (1733,2731)
methylation
glioblastoma. However, as long as the tu- anaplastic oligodendroglioma from ma-
mour shows the IDH-mutant and 1p/19q- lignant small cell astrocytic tumours CIC Mutation -60% (349,2464)

codeleted genotype, classification as (including small cell glioblastoma) is of CDKN2AI


p14ARF LOH -40% (45A}
major importance, because malignant
small cell astrocytic tumours behave CDKN2AI Promoter
much more aggressively, often with a p14ARF 15-30% (2702A,2779}
methylation
clinical course typical of IDH-wildtype FUBP1 Mutation -30% (349,2464)
glioblastomas (1937l Unlike IDH-mutant
NOTCH1 Mutation 20-30% (349,2464)
and 1 p/19q-codeleted anaplastic oligo-
dendrogliomas, small cell astrocytomas PIK3CA Mutation 10-20% (349,2464)

and glioblastomas lack combined IDH PIK3R1 Mutation -9% (349,2464)


mutation and 1 p/19q codeletion but of- TCF12 Mutation 7.5% (1407)
ten demonstrate EGFR amplification and
AR/01A Mutation -7% (349,2464)
chromosome 10 losses (1183,1937l.

Fig. 1.70

Anaplastic oligodendroglioma. MIB1
CDKN2C
Homozygous
deletion or <5%
(1069,1993,
2464)
mutation
immunohistochemistry shows high proliferative activity.

72 Diffuse gliomas
Fig. 1.71 Anaplastic oligodendroglioma. A R132H-mutant IDH1 stains tumour cells but is negative in blood vessels. B Cortical infiltration zone with tumour cells expressing R132H-
mutant IDH1, including perineuronal satellitosis.

the low frequency of gene amplification anaplastic oligodendrogliomas due to ho- anaplastic oligodendroglioma {1863).
detected in molecular profiling studies mozygous deletion, mutation, or aberrant However, none of these data considered
(2464,2731 ). promoter methylation {666,1084,2779) In IDH mutation or 1p/19q codeletion sta-
isolated cases, homozygous deletion or tus, which have been strongly associated
Molecular genetics mutation of the CDKN2C gene at 1p32 with improved response to adjuvant radi-
As the entity name suggests, IDH muta- has been observed in tumours without otherapy/chemotherapy and longer sur-
tion and 1 p/19q codeletion are the defin- CDKN2A deletions {1069,1993). Losses vival {344,2615). A retrospective analysis
ing genetic aberrations of IDH-mutant of 10q, mutations of the PTEN tumour of 1013 adult patients with anaplastic
and 1 p/19q-codeleted anaplastic oligo- suppressor gene, and amplifications of oligodendroglial tumours found a median
dendroglioma. Promoter mutations in proto-oncogenes are infrequent in IDH- overall survival of 8.5 years in patients
TERT are also present in the vast ma- mutant and 1 p/19q-codeleted anaplastic with 1 p/19q-codeleted tumours, versus
jority of these tumours, as is the case oligodendrogl iomas { 666, 1083, 2246). 3.7 years in patients with 1p/19q-intact
for IDH-mutant and 1p/19q-codeleted Epigenetically, these tumours are char- tumours {1436). Long-term follow-up
low-grade oligodendroglioma (89,1314, acterized by G-CIMP type A {2464) and data from the Radiation Therapy Oncol-
1408,2464). Investigation of regionally often demonstrate a proneural glioblas- ogy Group (RTOG) 9402 and European
or spatially distinct tissue samples from toma-like expression profile. The MGMT Organisation for Research and Treatment
individual patients has indicated that IDH promoter is typically hypermethylated of Cancer (EORTC) 26951 trials indicate
mutation, 1p/19q codeletion, and TERT {1733). even higher median overall survival times
promoter mutation are early events in (> 10 years) for patients with I DH-mutant
oligodendroglioma development that are Genetic susceptibility and 1 p/19q-codeleted anaplastic oligo-
shared by neoplastic cells from different See Genetic susceptibility (p. 67) in dendrogliomas who were treated with
tumour areas as well as between primary 0/igodendroglioma, /OH-mutant and combined radiotherapy and PCV chemo-
and recurrent tumours {2464). As in low- 1 p/19q-codeleted. therapy {344,2615). Patients rarely devel-
grade oligodendroglioma, CIC mutation op cerebrospinal fluid spread or systemic
is frequent in anaplastic oligodendro- Prognosis and predictive factors metastases. Local tumour progression is
glioma, whereas FUBP1 mutation occurs the most common cause of death.
in fewer cases. Other (less commonly) Prognosis
mutated genes include NOTCH pathway Recent therapeutic advances have mark- Predictive factors
genes (in particular, NOTCH1), various edly improved survival times of patients Clinical factors that have been associ-
epigenetic regulator genes (e.g. SETD2), with anaplastic oligodendrogliomas. Be- ated with longer survival are younger
and Pl3K pathway genes (e.g. P/K3CA) fore the introduction of adjuvant treat- age at diagnosis, higher Karnofsky per-
{2464). Mutations in the oligodendroglial ment with combined radiochemotherapy, formance score, and greater extent of
lineage-associated transcription factor reported median survival times ranged resection (1095,2617,2740). Previous re-
gene TCF12 have been detected in a from < 1 year {567) to 3.9 years {2337). section for lower-grade tumour has also
small subset (7.5%) of anaplastic oligo- A population-based analysis from Swit- been linked to more favourable prognosis
dendrogliomas and are associated with zerland reported a median survival time (2621 ). Recursive partitioning analysis of
an aggressive tumour type {1407). Over- of 3.5 years for patients with anaplastic various clinical and histological param-
all, the average number of chromosomes oligodendroglioma, which was markedly eters together with the 1 p/19q codele-
involved in copy number abnormalities is shorter than the median of 11.6 years for tion status identified five distinct prog-
higher in anaplastic oligodendrogliomas patients with WHO grade 11 oligodendro- nostic classes, defined mainly by patient
than in low-grade oligodendrogliomas glioma {1826). The CBTRUS calculated age, tumour location, and 1 p/19q status
{1353,2160). The COKN2A and CDKN2B 5- and 10-year survival rates of 52.2% (1884). A study based on the EORTC
loci on 9p21 are altered in a subset of and 39.3%, respectively, for patients with 26951 trial found a strong prognostic

Anaplastic oligodendroglioma, IDH-mutant and 1p/19q-codeleted 73


Anaplastic oligodendroglioma,
Anaplastic oligodendroglioma chromosomal imbalances (mostly NOS
lacking IDH mutation and genomic deletions) (1759}.
1 p/19q codeletion In adult patients, anaplastic oligo- Definition
In paediatric patients, isolated cases of dendroglioma without combined IDH A diffusely infiltrating anaplastic glioma
tumours with classic oligodendroglial mutation and 1 p/19q codeletion is not with classic oligodendroglial histolo- ·
histology and histological features of considered a distinct tumour entity or gy, in which molecular testing for com-
anaplasia but without IDH mutation and variant in the WHO classification. In- bined JOH mutation and 1p/19q cooete-,
1 p/19q codeletion have been reported stead, such tumours should be carefully tion could not be completed or was
{2157LHowever, the prognostic role of evaluated for genetic changes associ- inconclusive.
anaplastic features in 'these rare paedi- ated with IDH-wildtype glioblastomas As with oligodendroglioma, NOS, im-
atric oJigodendroglial tumours remains (in particular, gains of chromosorne 7, munohistochemical demonstration of
unknown {2157}. The· genetic altera- losses of chromosome 10, and EGFR IDH mutation (in particular IOH1 R132H)
tions in these rare paediatric tumours gene amplification), to exclude, for ex- and nuclear positivity for ATRX supports
are distinct from those in their adult ample, small cell astrocytoma/glioblas- the diagnosis of an oligodendroglial tu-
counterparts and may involve diverse toma (see Differential diagnosis). mour but cannot substitute for 1 p/19q .
codeletion testing, because nuclear ATRX .
expression is retained in a subset of IDH- .·'i
impact of the Ki-67 proliferation index on RTOG 9402 trial, patients with 1p/19q- mutant but 1p/19q-intact anaplastic astro- i,f
univariate analysis but no independent codeleted anaplastic gliomas had medi- cytomas. Similarly, immunopositivity for
influence on multivariate analysis {2034}. an overall survival times of 14.7 years with alpha-internexin and loss of nuclear CIC
In addition to 1p/19q codeletion and IDH radiotherapy plus PCV chemotherapy or FUBP1 expression supports the diag-
mutation, G-CIMP status and MGMT pro- versus 7.3 years with radiotherapy alone. nosis of anaplastic oligodendroglioma, ·
moter methylation have been reported In the EORTC 26951 trial, median overall NOS, but does not suffice for diagnosis ·
to provide prognostically relevant infor- survival was not reached after> 10 years of I DH-mutant and 1 p/19q-codeleted
mation for patients with anaplastic glio- of follow-up in patients with 1 p/19q- anaplastic oligodendroglioma (see the.
mas {2618,2619,2620,2740}. Long-term codeleted anaplastic gliomas treated by box Anaplastic oligodendroglioma lack-
follow-up data from the RTOG 9402 and radiotherapy plus PCV chemotherapy ing JOH mutation and 1p/19q codeletion).
EORTC 26951 trials suggest not only a versus 9.3 years in patients treated with The histological features of anaplastic ,
prognostic but also a predictive role of radiotherapy alone. In both trials, pa- oligodendroglioma, NOS, largely corre-
1 p/19q codeletion for long-term survival tients with 1 p/19q-intact tumours had sig- spond to those described for !DH-mutant.
after aggressive multimodal treatment nificantly shorter overall survival in both and 1 p/19q-codeleted anaplastic oligo-
consisting of surgical resection followed treatment arms. These data indicate that dendroglioma. However, the presence of
by upfront combined radiotherapy and molecular testing of anaplastic gliomas is a conspicuous astrocytic component is
PCV chemotherapy {344,26151 In the important and can guide therapy 127301. not compatible with the diagnosis of ana- .
plastic oligodendroglioma, NOS. Areas of ·
necrosis, including palisading necrosis,
are compatible with the diagnosis of ana- ·
plastic oligodendroglioma, NOS, provided
the tumour shows the typical cytological -
characteristics and other histological hall-
marks of oligodendroglioma, such as the
branching capillary network and micro-
calcifications. However, the differential
diagnosis of glioblastoma (WHO grade IV),
in particular the small-cell variant, must
be thoroughly considered in such cases,
and is in fact more appropriate when IDH ·
mutations are absent and genetic features
of glioblastoma (such as gain of chro-
mosome 7, loss of chromosome 10, and
EGFR amplification) are detectable.

ICD-0 code

Grading
Anaplastic oligodendroglioma, NOS,
corresponds histologically to WHO
grade Ill.

74 Diffuse gliomas
Oligoastrocytoma, NOS Reifenberger G.
Collins V.P.
Cairncross J.G.
Yokoo H.
Hartmann C. Yip S.
Hawkins C. Louis D.N.
Kros J.M.

Definition subjected to molecular testing for IDH with immunohistochemistry for ATRX and
A diffusely infiltrating, slow-growing gli- mutation and 1p/19q codeletion. Tumours R132H-mutant IDH1, followed by testing
oma composed of a conspicuous mix- with combined IDH mutation and 1p/19q for 1p/19q codeletion, and then followed
ture of two distinct neoplastic cell types codeletion are classified as IDH-mutant by IDH sequencing of the tumours that
morphologically resembling tumour cells and 1p/19q-codeleted oligodendroglio- were negative for R132H-mutant IDH1
with either oligodendroglial or astro- ma, irrespective of a mixed or ambiguous (2105). With this approach, the vast ma-
cytic features, and in which molecular histology. Tumours with IDH mutation but jority of diffuse and anaplastic gliomas
testing could not be completed or was without 1 p/19q codeletion are classified can be assigned to one of three major
inconclusive. as IDH-mutant diffuse astrocytoma, also glioma classes: IDH-mutant diffuse as-
In oligoastrocytoma, NOS, tumour cells irrespective of mixed or ambiguous his- trocytomas, IDH-mutant and 1p/19q-
with oligodendroglial or astroglial fea- tology. lmmunohistochemical testing for codeleted oligodendrogliomas, or IDH-
tures can be either diffusely intermingled loss of nuclear ATRX expression may also wildtype gliomas (including IDH-wildtype
or separated into distinct, biphasic areas. provide diagnostically helpful informa- glioblastomas). Overall, the diagnosis of
The mixed or ambiguous cellular dif- tion. Loss of ATRX positivity in the tumour oligoastrocytoma, NOS, should therefore
ferentiation precludes histological clas- cell nuclei but retained nuclear expres- remain exceptional (i.e. restricted to dif-
sification as either diffuse astrocytoma, sion in non-neoplastic cells (e.g. vascular fuse gliomas in which histology does not
NOS, or oligodendroglioma, NOS. Oli- endothelia, reactive astrocytes, and acti- allow for unequivocal stratification into ei-
goastrocytoma, NOS, is an exceptional vated microglial cells) supports the diag- ther astrocytic or oligodendroglial lineage
diagnosis, because most diffuse glio- nosis of IDH-mutant diffuse astrocytoma tumours and that cannot be subjected to
mas with mixed or ambiguous histology (2105,2220). Similarly, strong nuclear appropriate molecular testing or in which
can be classified as either IDH-mutant immunopositivity for p53, typically as a molecular findings are inconclusive).
diffuse astrocytoma or IDH-mutant and consequence of TP53 mutation leading
1p/19q-codeleted oligodendroglioma, to nuclear accumulation of mutant p53 ICD-0 code 9382/3
based on molecular testing. Oligoastro- protein, is often associated with loss of
cytomas, NOS, usually manifest in adult nuclear ATRX expression and is virtually Grading
patients, with preferential localization in mutually exclusive with 1p/19q codeletion Oligoastrocytoma, NOS, corresponds
the cerebral hemispheres. (2220). Recent data support a diagnostic histologically to WHO grade II.
algorithm for diffuse and anaplastic glio-
Genetic classification of mas based on stepwise analyses starting
oligoastrocytomas
The WHO classification discourages the
diagnosis of tumours as oligoastrocy-
toma or mixed glioma. Molecular genetic
analyses have clearly shown that the vast
majority of tumours previously classified
as oligoastrocytomas have a genetic pro-
file typical of either diffuse astrocytoma
(i.e. IDH mutation combined with TP53
mutation and ATRX mutation I loss of nu-
clear ATRX expression) or oligodendro-
glioma (i e. IDH mutation combined with
1 p/19q codeletion and TERT promoter
mutation) {349,2220,2464). The histo-
logical criteria provided in previous WHO
classifications for oligoastrocytic gliomas
left room for considerable interobserver
variability {494,2611 ). Thus, diffuse glio-
mas, including those with mixed or am-
biguous histological features, should be

Oligoastrocytoma, NOS 75
..
initial diagnosis Anaplastic oligoastrocytoma,
0 OA A GBM GBMo GBMs gcGBM GS NOS
Definition
An oligoastrocytoma, NOS, with focal or
diffuse histological features of anaplasia,
including increased cellularity, nuclear
atypia, pleomorphism, and brisk mitotic
activity
Anaplastic oligoastrocytoma, NOS, is
an exceptional diagnosis, because most
high-grade gliomas with mixed or am-
biguous histology can be classified as
either I DH-mutant anaplastic astrocyto-
ma or I DH-mutant and 1p/19q-codeleted
anaplastic oligodendroglioma, based on
molecular testing. Moreover, if molecu-
lar testing of an anaplastic glioma with
mixed or ambiguous histology shows an
IDH-wildtype status, IDH-wildtype glio-
blastoma must be considered as a likely
differential diagnosis, because most
0 A-IDHmut A-IDHwt GBM GBM-IDHmut gcGBM GS IDH-wildtype anaplastic gliomas have
genetic profiles of glioblastoma and are
integrated diagnosis associated with correspondingly poor
Fig. 1.73 Changes from initial (purely histology-based) to integrated diagnosis in 405 adult patients with supratentorial prognosis. Anaplastic oligoastrocyto-
glioma. Width of bars indicates relative proportions of the initial tumour groups. A, astrocytoma; OA, oligoastrocytoma; mas, NOS, usually manifest in adult pa-
0, oligodendroglioma; GBM, glioblastoma; GBMo, glioblastoma with oligodendroglial component; GBMs, secondary tients, with preferential localization in the
glioblastoma; gcGBM, giant cell glioblastoma; GS, gliosarcoma. Note that all oligoastrocytomas were redistributed cerebral hemispheres.
to other diagnoses (!DH-mutant and 1p/19q-codeleted oligodendroglioma, !DH-mutant astrocytoma, IDH-wildtype
astrocytoma, or IDH-wildtype glioblastoma) after molecular marker analysis, including IDH mutation I G-CIMP, ATRX
loss of nuclear expression, 1 p/19q codeletion, and 7p gain / 1 Oq loss. Reprinted from Reuss DE et al. (2105). ICD-0 code 9382/3

Grading
Anaplastic oligoastrocytoma, NOS,
corresponds histologically to WHO
grade Ill.

Oligoastrocytoma, genetic alterations, including 1 p/19q requires detailed molecular and immuno-
dual-genotype codeletion, in the oligodendroglial and histochemical analyses to unequivocally
Dual-genotype oligoastrocytoma is not astrocytic tumour components of the demonstrate the two genetically distinct
considered to be a distinct entity or vast majority of oligoastrocytomas (1359, tumour cell subpopulations. Care must
variant in the WHO classification. The 2050). However, a few instances of oli- be taken to avoid misinterpretations (e.g.
designation refers to an IDH-mutant oli- goastrocytoma showing evidence of ge- due to artefactually negative nuclear im-
goastrocytoma of WHO grade II that is netically distinct oligodendroglial and munostaining for ATRX) and false-posi-
composed of a conspicuous mixture of astrocytic cell populations have been tive detection of 1p/19q-codeleted nuclei
astrocytic and oligodendroglial tumour reported (1067,2050,2754). It can be as- by FISH (e.g. due to incomplete chro-
cell populations demonstrating molecu- sumed that dual-genotype oligoastrocy- mosomal representation in transected
lar evidence of an astrocytic genotype tomas are monoclonal tumours that arise nuclei, aneuploidy/polyploidy, or partial
(i.e. IDH mutation combined with TP53 from an I DH-mutant cell of origin but later hybridization failure) (763). Overall, dual-
mutation I nuclear p53 accumulation, during tumorigenesis develop cytologi- genotype oligoastrocytoma seems to be
loss of nuclear ATRX expression, and cally distinct subpopulations of tumour very rare, but its true incidence is difficult
lack of 1p/19q codeletion) and an oligo- cells with an astrocytic genotype (i.e. to ascertain because tissue sampling is
dendroglial genotype (i.e IDH mutation TP53 and A TRX alteration but no 1 p/19q rarely comprehensive in diffuse gliomas
combined with 1p/19q codeletion, lack codeletion) or an oligodendroglial geno- and molecular analysis of DNA extracted
of TP53 mutation I nuclear p53 accu- type (i.e. 1p/19q codeletion but no TP53 from a bulk sample may not detect such
mulation, and retained nuclear ATRX ex- or ATRX alteration). Classification of du- molecular heterogeneity.
pression). Studies have revealed shared al-genotype oligoastrocytoma therefore

76 Diffuse gliomas
Genetic classification of anaplastic Anaplastic oligoastrocytoma, (i.e. IDH mutation combined with 1p/19q
oligoastrocytomas dual-genotype codeletion, lack of TP53 mutation I nu-
The WHO classification discourages Dual-genotype anaplastic oligoastrocy- clear p53 accumulation, and retained
the diagnosis of tumours as anaplastic toma is not considered to be a distinct nuclear ATRX expression). To date, only
oligoastrocytoma or anaplastic mixed entity or variant in the WHO classifica- isolated cases of tumours considered to
glioma. Molecular genetic analyses have tion. The designation refers to an IDH- be dual-genotype anaplastic oligoas-
not demonstrated entity-specific genetic mutant anaplastic oligoastrocytoma of trocytoma have been reported {1067,
or epigenetic profiles for these tumours. WHO grade Ill that demonstrates mo- 2754}. These tumours occurred in the
Instead. large-scale molecular profiling lecular evidence of genetically distinct cerebral hemispheres of adult patients.
studies have shown that the vast major- astrocytic and oligodendroglial tumour Utmost care must be taken to exclude
ity of anaplastic oligoastrocytomas have cell populations characterized by an technical shortcomings that may lead to
genetic alterations and DNA methylation astrocytic genotype (i.e. IDH mutation artefactual regional variation in immu-
profiles typical of either IDH-mutant ana- combined with TP53 mutation I nuclear nohistochemical staining for ATRX or in
plastic astrocytoma (with commonly as- p53 accumulation, loss of nuclear ATRX FISH analysis for 1p/19q codeletion.
sociated TP53 mutation and ATRX muta- expression, and lack of 1p/19q codele-
tion/ loss of nuclear ATRX expression) or tion) and an oligodendroglial genotype
!DH-mutant and 1p/19q-codeleted ana-
plastic oligodendrogliomas (with com-
monly associated TERT promoter muta- including those with a mixed or ambigu- astrocytomas. !DH-mutant and 1p/19q-
tions). A third, smaller subset of cases ous histological phenotype, should be codeleted anaplastic oligodendroglio-
correspond to IDH-wildtype anaplastic subjected to thorough molecular test- mas, or IDH-wildtype anaplastic glio-
gliomas, which commonly demonstrate ing, most notably for IDH mutation and mas (most of which are characterized by
hallmark genetic aberrations of glioblas- 1p/19q codeletion. As with oligoastrocy- glioblastoma-associated genetic profiles
toma - in particular, gain of chromosome toma. NOS, immunohistochemical test- and the associated poor prognosis).
7 and loss of chromosome 10 combined ing for loss of nuclear ATRX expression Overall, the diagnosis of anaplastic oli-
with TERT promoter mutation and gene and nuclear accumulation of p53 may goastrocytoma, NOS, should therefore
amplifications (such as EGFR amplifica- provide additional, diagnostically helpful remain exceptional, i.e. restricted to
tion) (349,2464,2731,2751 l. The criteria information {2105,2220l. This integrated anaplastic gliomas with astrocytic and
provided in previous WHO classifications approach enables a clear diagnostic oligodendroglial components that cannot
for anaplastic oligoastrocytoma left room stratification of the vast majority of ana- be subjected to appropriate molecular
for considerable interobserver variability plastic gliomas into one of the three testing or in which molecular findings are
{1370,2611}. Thus, anaplastic gliomas, major classes: !DH-mutant anaplastic inconclusive.

Oligoastrocytoma, NOS 77
CHAPTER 2

Other astrocytic tumours


Pilocytic astrocytoma
Pilomyxoid astrocytoma
Subependymal giant cell astrocytoma
Pleomorphic xanthoastrocytoma
Anaplastic pleomorphic xanthoastrocytoma
Pilocytic astrocytoma Collins V.P.
Tihan T.
Jones 0.
Giannini C.
VandenBerg S.R. Rodriguez F.
Burger PC. Figarella-Branger 0.
Hawkins C.

Definition 100 000 population, which declines sub- intraventricular and their site of origin dif-
An astrocytoma classically character- stantially trorn s 14 years to 15-19 years ficult to define.
ized by a biphasic pattern with variable (1863). Pilocytic astrocytoma is the most
proportions of compacted bipolar cells common glioma in children, without a Clinical features
with Rosenthal fibres and loose, textured significant sex predilection. It accounts Pilocytic astrocytomas produce focal
multipolar cells with microcysts and oc- for 33.2% of all gliomas in the 0-14 years neurological deficits or non-localizing
casional granular bodies. age group {1862) and 17.6% of all child- signs, such as macrocephaly, head-
Genetically, pilocytic astrocytomas are hood primary brain tumours {1863). ache, endocrinopathy, and increased
characterized by the presence of mu- Similarly, in a study of 1195 paediatric tu- intracranial pressure due to mass effect
tations in genes coding for proteins in- mours from a single institution, pilocytic or ventricular obstruction. Seizures are
volved in the MAPK pathway {1176,2855). astrocytoma was the single most com- uncommon because the lesions involve
The most frequent genetic change is tan- mon tumour (accounting for 18% of cas- the cerebral cortex infrequently {466,
dem duplication of 7q34 involving the es overall) in the cerebral compartment 725). Given their slow rate of growth,
BRAF gene resulting in oncogenic BRAF {2174). In adults, pilocytic astrocytoma pilocytic tumours' clinical presentation
fusion proteins. Pilocytic astrocytomas tends to appear about a decade earlier is generally that of a slowly evolving le-
are the most common glioma in children (mean patient age: 22 years) than does sion. Pilocytic astrocytomas of the op-
and adolescents, and affect males slight- WHO grade II diffuse astrocytoma {784), tic pathways often produce visual loss.
ly more often than females. They are but relatively few arise in patients aged Proptosis may be seen with intraorbital
preferentially located in the cerebellum > 50 years. examples. Early, radiologically detected
and cerebral midline structures (i.e. the lesions may not be associated with visual
optic pathways, hypothalamus, and brain Localization symptoms or ophthalmological deficits
stem) but can be encountered anywhere Pilocytic astrocytomas arise throughout (1054,1515}. Hypothalamic/pituitary dys-
along the neuraxis {1533) They are gen- the neuraxis; however, in the paediat- function, including obesity and diabetes
erally circumscribed and slow-growing, ric population, more tumours arise in insipidus, is often apparent in patients
and may be cystic. Pilocytic astrocytoma the infratentorial region. Preferred sites with large hypothalamic tumours (2159)
largely behaves as is typical of a WHO include the optic nerve (optic nerve Some hypothalamic/chiasmatic lesions
grade I tumour {1533), and patients can glioma) {1054), optic chiasm/hypothala- in young children have been associated
be cured by surgical resection. However, mus (2159), thalamus and basal ganglia with leptomeningeal seeding and a poor
complete resection may not be possible (1622), cerebral hemispheres {725,1994), outcome (1934}; see Pilomyxoid astro-
in some locations, such as the optic path- cerebellum (cerebellar astrocytoma) cytoma (p. 88), a variant of pilocytic as-
ways and the hypothalamus. Pilocytic (974), and brain stem (dorsal exophytic trocytoma. Pilocytic astrocytomas of the
astrocytomas, particularly those involv- brain stem glioma) {311,709,1995) Pilo- thalamus generally present with signs of
ing the optic pathways, are a hallmark of cytic astrocytomas of the spinal cord are cerebrospinal fluid obstruction or neu-
neurofibromatosis type 1 (NF1). less frequent but not uncommon (1677, rological deficits (such as hemiparesis)
Pilomyxoid astrocytoma is considered to 2186). Large hypothalamic, thalamic, and due to internal capsule compression.
be a variant of pilocytic astrocytoma. brain stem lesions may be predominantly Cerebellar pilocytic astrocytomas usu-
ally present during the first two decades

---
ICD-0 code 9421/1 100 of life, with clumsiness, worsening head-
ache, nausea, and vomiting. Brain stem
Grading
90

80
.,- examples most often cause hydroceph-
Pilocytic astrocytoma corresponds histo- 70
/ alus or signs of brain stem dysfunction.
logically to WHO grade I. ! 60
I Unlike diffuse astrocytoma of the pons,
-� so / which produces symmetrical so-called
Epidemiology �
] 40 ,/ pontine hypertrophy, pilocytic tumours of
Pilocytic astrocytoma accounts for 5.4% u 30
the brain stem are usually dorsal and ex-
/
of all gliomas (1826). According to a 20
ophytic into the cerebellopontine angle.
10
/
2007-2011 statistical report from the Cen- I Spinal cord examples produce non-spe-
tral Brain Tumor Registry of the United 10 20 30 40 so 60
cific signs of an expanding mass (1678,
States (CBTRUS), pilocytic astrocytoma Age at diagnosis 2074,2186}.
is most common during the first two dec- Fig. 2.01 Cumulative age distribution {both sexes) of
ades of life, with an average annual age- pilocytic astrocytomas, based on biopsies from 205 Imaging
adjusted incidence rate of 0.84 cases per patients treated at the University Hospital Zurich {1533). Most cerebellar and cerebral pilocytic

80 Other astrocytic tumours


astrocytomas are well demarcated,
with a round or oval shape and smooth
margins. Calcifications are occasion-
ally present. Heterogeneity as .a result
of cyst-like areas intermixed with gen-
erally intensely enhancing soft tissue
components is typical. Consistent with
the tumour's low biological activity, sur-
rounding vasogenic oedema (if present)
is much less extensive than in higher-
grade glial neoplasms. About two thirds
of cases manifest as a cyst-like mass
with an enhancing mural nodule; the
other third present either as a cyst-like
mass with a central non-enhancing zone
or as a predominantly solid mass with a
minimal or no cyst-like portion {474,1927).
In those with a cyst-like morphology, the
cyst wall may or may not enhance (165).
and cyst wall enhancement may or may
not indicate tumour involvement (165).
Occasional atypical imaging features of
these tumours include multiple cyst-like
masses and haemorrhage. Fig. 2.02 Most pilocytic astrocytomas are strongly and diffusely enhancing. A Left optic nerve. B Hypothalamic.
Most optic nerve gliomas are pilocytic as- C Basal ganglia. D Tectal. E Cerebellar. F Spinal. Some tumours are solid (A, B, D) and others are cystic with an
enhancing mural nodule (C, E, F).
trocytomas, and characteristically mani-
fest as a kinked or buckled enlargement
of the optic nerve. Tumours in this loca- Macroscopy Microscopy
tion often have different imaging appear- Most pilocytic astrocytomas are soft, This astrocytic tumour of low to moderate
ances depending on whether the patient grey, and relatively discrete. lntratu- cellularity often has a biphasic pattern,
has NF1. NF1-associated tumours more moural or paratumoural cyst formation is with varying proportions of compacted
commonly affect the optic nerve, rarely common, including with a mural tumour bipolar cells with Rosenthal fibres and
extend beyond the optic pathway, and nodule. In spinal cord examples, syrinx loose-textured multipolar cells with mi-
are usually not cyst-like, whereas those formation may extend over many seg- crocysts. The biphasic pattern is best
not associated with NF1 usually involve ments (1678). Chronic lesions may be seen in cerebellar tumours. However,
the optic chiasm, extend extraoptically, calcified or may stain with haemosiderin. pilocytic astrocytoma can exhibit a wide
and are frequently cyst-like {1342). Optic nerve tumours often circumfer- range of tissue patterns, sometimes sev-
entially involve the subarachnoid space eral within the same lesion. A variant of
Spread (2427). the compact, piloid pattern occurs when
Otherwise typical pilocytic astrocytomas
very occasionally seed the neuraxis, on
rare occasions even before the primary
tumour is detected {775,1934,1996). The
proliferation rate in such cases varies,
but is usually low {1681). Therefore, this
atypical behaviour of pilocytic astrocyto-
ma cannot be predicted. The hypothala-
mus is usually the primary site. Neuraxis
seeding does not necessarily indicate
future aggressive growth; both the pri-
mary lesion and the implants may grow
only slowly (775,1681,1996}. The implants
may be asymptomatic, and long-term
survival is possible, even without adju-
vant treatment {1996). The pilomyxoid
astrocytoma variant (see Pilomyxoid as-
trocytoma, p. 88), typically occurring in
Fig. 2.03 Cerebellar pilocytic astrocytoma. A Axial T1-weighted MRI shows a left cerebellar cyst-like mass with
the hypothalamic region, more often un- slightly hyperintense mural nodule along its lateral margin. B Axial T2-weighted MRI shows hyperintensity of the cyst-
dergoes craniospinal spread (2555). like portion of the mass with lower signal intensity of the mural nodule. C Postcontrast axial T1-weighted MRI shows
heterogeneous intense enhancement of the mural nodule.

Pilocytic astrocytoma 81
Fig. 2.04 Pilocytic astrocytoma. A Diffuse infiltration of the hippocampus and neighbouring structures. Note the focal formation of cysts. B Large, partially cystic pilocytic astrocytoma
of the cerebellum with a typical mural nodule {882}. C Large pilocytic astrocytoma extending into the basal cisterns.

the elongated cells are less compact but cytological variation, and basic cytologi- peripheral, more infiltrative areas. Cells
separated by mucin. In such cases, indi- cal patterns are often present in combi- closely resembling oligodendrocytes
vidual cell processes can be seen, and nation. Compact portions of the tumour are typically less frequent (480}, but
cell shape varies to include more fult- yield bipolar piloid cells; long, hair-like can constitute a dominant component in
bodied and pleomorphic, less obviously processes that often extend across a some cases, particularly in the cerebel-
pitoid cells. A distinctive lobular pattern full microscopic field; and Rosenthal fi- lum. These cells strongly express OLIG2
results when leptomeningea/ involvement bres. Nuclei are typically elongated and {481}, although other tissue patterns may
induces a desmoplastic reaction. At this cytologically bland. Due to their high express OLIG2 as welt. With the cells ar-
site, tissue texture varies but Rosenthal content of retractile eosinophilic fibrils, ranged in sheets or dispersed within pa-
fibres are usually abundant. Rare mi- these cells are strongly immunopositive renchyma, the overall appearance may
toses, hyperchromatic and pleomorphic for GFAP. Cells derived from microcystic resemble that of an otigodendroglioma,
nuclei, glomeruloid vascular prolifera- areas have round to oval, cytologically particularly in a limited sample. The find-
tion, infarct-like necrosis, and infiltration bland nuclei; a small cell body; and rela- ing of foci of classic pilocytic astrocytoma
of Jeptomeninges are compatible with the tively short, cobweb-like processes that usually enables the correct classification
diagnosis of pilocytic astrocytoma and are fibril-poor and only weakly positive of such lesions. Regimented palisades
are not signs of malignancy. for GFAP. This growth pattern may be as- (a so-called spongiob/astoma pattern)
Due to the heterogeneity of histological sociated with eosinophilic granular bod- are a striking feature in some pilocytic
features, smear preparations of pilo- ies. Cells indistinguishable from those astrocytomas.
cytic astrocytomas show considerable of diffuse astrocytoma may populate

Fig. 2.05 Typical histological features of pilocytic astrocytoma. A Densely fibrillary areas composed of bipolar cells with long thin processes and rich in Rosenthal fibres as seen on
intraoperative cytological smears and (B) on histological sections. C Eosinophilic granular bodies. Neoplastic cell nuclei are hyperchromatic. A rare mitotic figure is present (arrow).
D Loosely arranged microcystic areas. E Pilocytic astrocytoma is typically a highly vascular tumour and shows both glomeruloid vessels and (F) thick-walled hyalinized vessels. The
vascular changes correlate with the frequent presence of contrast enhancement on imaging.

82 Other astrocytic tumours



Fig. 2.06 Pilocytic astrocytoma. A Typical biphasic pattern with alternating compact and loose architectural patterns.
rounded nuclei and occasional microcystic changes.

Although many pilocytic astrocytomas extent, pre-existing neurons are some- a property best seen in smear prepara-
are benign, some show considerable hy- times trapped, and some lesions even tions. Rosenthal fibres are most common
perchromasia and pleomorphism. Rare demonstrate limited cytological atypia. in compact, piloid tissue. They appear
mitoses are present, but brisk mitotic ac- Such lesions should be distinguished bright blue on Luxol fast blue staining.
tivity, in particular diffuse brisk mitotic ac- from ganglion cell tumours. The pilo- Although helpful in diagnosis, their pres-
tivity, characterizes anaplastic change, myxoid variant (see Pilomyxoid astrocy- ence is not obligatory, and they are nei-
which has prognostic implications l2150l toma, p. 88) is characterized by uniform ther specific to pilocytic astrocytoma nor
(see p. 88). In occasional (often cerebel- bipolar cells in a myxoid background indicative of neoplasia. They are often
lar) tumours, a diffuse growth pattern and frequent perivascular arrangements. seen in ganglioglioma and are a com-
overshadows more typical compact Sometimes this pattern is associated mon finding in chronic reactive gliosis.
and microcystic features. In such cases, with more conventional pilocytic regions Densely fibrillar, paucicellular lesions
the presence of hyperchromatic nuclei or so-called intermediate tumours !1168). containing Rosenthal fibres are as likely
or mitotic figures can cause confusion Although astrocytomas in children are to be reactive gliosis as pilocytic astrocy-
with high-grade diffuse astrocytoma. usually classified as either the pilocytic toma. Ultrastructurally, Rosenthal fibres
Less worrisome are obvious degenera- or the diffuse type, many cases do not lie within astrocytic processes and con-
tive atypia with pleomorphism and nu- in fact fit clearly into either category on sist of amorphous, electron-dense ele-
clear-cytoplasmic pseudoinclusions, the basis of histology alone. Increasingly, ments surrounded by intermediate (glial)
frequently seen in long-standing lesions. molecular profiling plays an ancillary role filaments !588). Being composed of
Multiple nuclei within large or giant cells in this distinction. In some cases, small alpha-B-crystallin !862}, they lack GFAP
are localized circumferentially (called a biopsy size contributes to difficulties in immunoreactivity except in their fibril-rich
pennies-on-a-plate arrangement) !974). classification, especially in cases with a periphery.
Hyalinized and glomeruloid vessels are brain stem or spinal location.
prominent features of some cases. Any Eosinopht!ic granular bodies
necrosis is often infarct-like and non- Rosenthal fibres Eosinophilic granular bodies form globu-
palisading. Perivascular lymphocytes These tapered, rounded, or corkscrew- lar aggregates within astrocytic process-
may also occur. Because pilocytic astro- shaped, brightly eosinophilic, hyaline es. They are brightly eosinophilic in H&E
cytomas overrun normal tissue to some masses are intracytoplasmic in location, sections, periodic acid-Schiff-positive,

B OLIG2 expression is restricted to pseudo-oligodendroglial cells.

Pilocync astrocytoma 83
,

and immunoreactive for alpha-t-antichy- astrocytomas, it is not surprising that re- Growth pattern
motrypsin and alpha-1-antitrypsin {1236). gressive features are common. Markedly Typically, pilocytic astrocytomas are
Although eosinophilic granular bodies hyalinized, sometimes ectatic vessels macroscopically somewhat discrete.
may be present in pilocytic astrocytoma, are one such feature. When neoplastic Thus, when the anatomical site permits
they are more frequent in other glial or cells are scant, it can be difficult to distin- (e.g. in the cerebellum or cerebral hemi-
glioneuronal neoplasms, in particular guish these tumours from cavernous an- spheres), many can be removed in toto
ganglion cell tumours and pleomorphic giomas with accompanying piloid gliosis. {725,974,1994). Microscopically, howev-
xanthoastrocytomas. Evidence of previous haemorrhage (hae- er, many lesions are not well defined with
mosiderin) increases the resemblance. respect to surrounding brain. Lesions
Vasculature Presentation with acute haemorrhage is typically permeate parenchyma for dis-
Pilocytic astrocytomas are highly vascu- infrequent. Calcification, infarct-like ne- tances of millimetres to several centime-
lar, as evidenced by their contrast en- crosis, and lymphocytic infiltrates are tres, often entrapping normal neurons in
hancement {761). Although generally ob- additional regressive changes. Over- the process. However, pilocytic tumours
vious in H&E sections, this vascularity is all, calcification is an infrequent finding, are relatively solid compared with dif-
accentuated on immunostains for mark- and is only rarely present in optic nerve fuse gliomas, and do not aggressively
ers of basement membrane (e.g. colla- or hypothalamic/thalamic tumours or in overrun surrounding tissue. This prop-
gen IV and laminin) and endothelial cells superficially situated cerebral examples. erty, evidenced by at least partial lack of
(e.g. CD31 and CD34). This glomeruloid Cysts are a common feature of pilocytic axons on Bodian or Bielschowsky silver
vasculature may also line tumoural cyst astrocytoma, especially in the locations impregnations, and NFP immunostains,
walls and is occasionally seen at some specified above. Neovascularity often is of diagnostic value. Pilocytic astrocy-
distance from the lesion, but this should lines cyst walls, resulting in a narrow tomas of the optic nerve and chiasm dif-
not prompt tumour misclassification or band of intense contrast enhancement fer somewhat in their macroscopic and
overgrading. There may be structural at the circumference of some cysts. microscopic patterns of growth; they are
and biological differences from similar Dense piloid tissue with accompanying often less circumscribed and therefore
vessels occurring in glioblastomas, with Rosenthal fibres external to this vascular difficult to delineate either macroscopi-
the microvasculature of glioblastomas layer is sometimes seen. When this layer cally or microscopically. They share the
exhibiting a looser, more disorganized ar- is narrow and well demarcated from the same propensity for leptomeningeal in-
chitecture and differences in expression surrounding normal tissue, it can be as- volvement as seen in pilocytic tumours
of angiogenesis-related genes {1727) sumed to be reactive in nature. In other at other sites, but are somewhat more
instances, the glial zone is more promi- diffuse, especially within the optic nerve.
Regressive changes nent, less demarcated, and neoplastic. This is particularly evident when patholo-
Given the indolent nature and of- gists attempt to determine the extent of
ten slow clinical evolution of pilocytic a lesion by analysis of sequential nerve

Table 2.01 Genetic alterations affecting the MAPK pathway in pilocytic astrocytomas and their diagnostic utility; adapted from Collins VP et al. (482}
%of
Gene Change Diagnostic utility References
tumours
Tandem duplications resulting in KIAA1549-BRAF fusion proteins,
BRAFand Common in pilocytic astrocytomas, particularly cerebellar;
all having the BRAF kinase domain and with the BRAF N-terminal >70% (1176, 1178,2855}
KIAA1549 rare in other tumour forms
regulatory domain replaced by the N-terminal end of KIAA 1549
BRAFand Deletions or translocations resulting in BRAF fusion proteins, all
Occur in pilocytic astrocytomas; extremely rare in other
various other having the BRAF kinase domain and with the BRAF regulatory -5% (1176,2855}
entities
genes domain replaced by the N-terminal part of another gene
Occurs mainly in supratentorial pilocytic astrocytomas;
BRAF V600E mutation -5% also in gangliogliomas, pleomorphic xanthoastrocytomas, (1176,2855)
and dysembryoplastic neuroepithelial tumours
Typically germline; closely associated with optic pathway
NF1 Loss of wild type and retained inherited mutation -8% (1176,2855}
pilocytic astrocytoma
Found mainly in midline pilocytic astrocytomas; frequency
FGFR1 Mutation <5% (1176,2855}
not established in other entities
Rare in pilocytic astrocytoma; also observed in other low-
FGFR1 Fusions I internal tandem duplication <5% (1176,2855}
grade gliomas
Rare in pilocytic astrocytoma; frequency not established
NTRKfamily Fusions -2% (1176,2855)
in other entities
Single Rare in pilocytic astrocytoma; frequency not established
KRAS Mutation (1176,2855}
cases in other entities
Fusions with consequences similar to those of BRAF fusions, i.e.
Single Rare in pilocytic astrocytoma; frequency not established in
RAF1 the loss of the regulatory domain and its replacement by the N- (1176,2855}
cases other entities
terminal end of SGRAP3

84 Other astrocytic tumours


.J: ._...
margins. Microscopically, the lesion can
be followed to a point beyond which it
becomes less cellular, but there is no
clearly defined termination.
There has been considerable discus-
sion regarding the existence of a diffuse
variant of pilocytic astrocytoma, particu-
larly in the cerebellum {847,974,1882}
Although some such cases are simply
classic pilocytic tumours in which the in-
filtrative edge is somewhat broader than
expected or an artefact of plane of sec-
tion, there are also occasional distinctly
infiltrative lesions that mimic diffuse fibril-
lary astrocytoma, even in large speci-
mens. In two large studies, the outcomes
for children with so-called diffuse pilo-
cytic astrocytoma of the cerebellum were

p
SRGAP3:RAF1 1 'Yo
favourable, supporting the idea that such
tumours belong to the spectrum of pilo- 7% ( RAF1
cytic astrocytoma {974,1882). True infil-
trating diffuse astrocytomas account for KIAA1549:BRAF: 70 'Yo \

---
as many as 15% of all astrocytic tumours Other BRAF Fusion: 5 %
of the cerebellum, but most are high- 8RAFMutation: 5 % �
grade (i.e. WHO grade Ill or IV) {974).

/nftltration of the meninges
Involvement of the subarachnoid space
is common in pilocytic astrocytoma. It
Nucleus

(§iv
/
��\
is not indicative of aggressive or malig-
nant behaviour and does not portend
subarachnoid dissemination; rather, it is
a characteristic and even diagnostically
helpful feature. Leptomeningeal inva- � ,--,.
sion can occur at any tumour site, but is
particularly common in the cerebellum I Cytoplasm
and optic nerve. The leptomeningeal Fig. 2.08 Pilocytic astrocytoma. Illustration of the MAPK pathway components with the approximate incidence rates of
component may be reticulin-rich, more the various mutations found in a series of pilocytic astrocytomas. Adapted by permission from Jones DTW et al. {1176}.
commonly in the optic nerve than in the
cerebellum. Another typical pattern of ex- without necrosis {2150). Tumours with in a subset of these tumours, particu-
traparenchymal spread is extension into these features should not be designated larly those developing in the cerebellum
perivascular spaces. glioblastoma, because their prognosis is {2145}, as well as absence of the R132H-
not uniformly grim. In general, the pres- mutant IDH1 protein. In the future, com-
Anaplastic transformation (p1/ocytic astro- ence of necrosis in pilocytic astrocyto- prehensive molecular characterization
cytoma with anaplasia) mas with anaplasia is associated with a will facilitate a more objective distinction
As a group, pilocytic astrocytomas are better prognosis than in classic cohorts from morphologically similar high-grade
remarkable in that they maintain their of glioblastoma. The diagnosis of pilo- gliomas. In a recent study of 886 cases
WHO grade I {325) status over years and cytic astrocytoma with anaplasia may be of paediatric low-grade glioma, with long
even decades. The alterations that occur preferable. Many such tumours had pre- clinical follow-up, there were no patients
over time are typically in the direction of viously been irradiated {591,2565), and with the KIAA1549-BRAFfusion who un-
regressive change rather than anaplasia. therefore radiation therapy may be a fac- derwent anaplastic transformation and
One large study found the acquisition of tor promoting anaplastic change in some died of their disease {1683).
atypia (in particular increased cellularity, cases. However, pilocytic astrocytomas
nuclear abnormalities, and occasional with anaplasia (including cases that de- lmmunophenotype
mitoses) to be of no prognostic signifi- velop in the setting of NF1) also occur in Pilocytic astrocytomas are well-differen-
cance {2565). In a subsequent series, patients without prior irradiation {2150). tiated gliomas of the astrocytic lineage,
anaplasia in pilocytic astrocytoma was Grading criteria and nomenclature for and strongly express GFAP, 8100, and
found to occur primarily in the form of dif- these tumours are yet to be defined. OLIG2 {1865). Synaptophysin may dem-
fuse, brisk mitotic activity (usually > 4 mi- Molecular studies, although limited, onstrate partial or weak reactivity, which
toses per 10 high-power fields), with or have demonstrated BRAF duplications should not be interpreted as evidence for

Pilocytic astrocytoma 85
gene {123,575,1961l. This is a tandem
duplication resulting in a transforming fu-
sion gene between KIAA 1549 and BRAF
{1178l. The N-terminus of the KIAA1549
KIAA1549 protein replaces the N-terminal regula-
tory region of BRAF, retaining the BRAF
kinase domain, which is consequently
uncontrolled and constitutively activates
the MAPK pathway {724,1178,2360l. This
abnormality is found at all anatomical lo-
cations, but is most frequent in cerebel-
lar tumours and somewhat less common
at other sites. The gene fusions involve
nine different combinations of KIAA1549
KIAA1549 and BRAF exons, making identification
by RT-PCR or immunochemistry diffi-
cult, resulting in many centres accept-
ing the demonstration of a duplication at
7q34 (usually using FISH probes) as evi-
dence of the KIAA 1549-BRAF fusion. In
small numbers of cases, eight additional
gene partners for BRAF fusions have
also been identified (FAM131B, RNF130,
CLCN6, MKRN1, GNA 11, OKI, FXR1, and
Fig. 2.09 Pilocytic astrocytoma: the development of the KIM 1549-BRAFfusion gene. The upper black box represents
7q34. Both KIM 1549 and BRAF read towards the centromere (cent.). A fragment of approximately 2 Mb is duplicated MACF1), with fusions occurring by vari-
(involving parts of both genes) and inserted at the break point, producing tandem duplication and fusion between the 5' ous genetic rearrangements (including
end of KIM 1549 and the 3' end of the BRAF gene (which codes for the kinase domain). The fusion gene thus codes deletions and translocations) and all re-
for the BRAF kinase domain together with the N-terminal part of KIAA 1549, replacing the BRAF regulatory domain. sulting in loss of the N-terminal regulatory
The red and green dots show the location of FISH probes that can be used to identify the occurrence of the tandem region of the BRAF protein, with retention
duplication, as demonstrated in the lower part of the figure, showing interphase normal and tumour nuclei with the of the kinase domain {1176,2855l.
tandem duplication hybridized with such probes. The two interphase copies of chromosome 7 in the normal nucleus
each show a single red and a single green signal adjacent to each other. In contrast, the tumour cell nuclei show one
In addition to harbouring BRAF fusion
pair of normal chromosomal signals, but the other copy of chromosome 7 shows an additional (yellow) signal, due to the genes, essentially all pilocytic astrocyto-
fusion of the extra, now adjacent, red and green signals. Reprinted from Collins VP et al. {482). mas studied in sufficient detail have been
shown to have an alteration affecting some
a glioneuronal tumour. NFP typically out- not exclude other substitutions at posi- component of the MAPK pathway {1176,
lines a mainly solid tumour, highlighting tion 132 or mutations of the IDH2 protein. 2855l. These alterations include the well-
normal surrounding axons in compressed Because essentially all pilocytic astrocy- documented NF1 mutations, the hotspot
adjacent CNS parenchyma. Optic nerve tomas have activating genetic alterations BRAF mutation commonly known as the
tumours generally produce a fusiform in components of the MAPK pathway V600E mutation, KRAS mutations, recur-
expansion of the nerve and have axons {1176). phosphorylated MAPK irnrnu- rent aberrations affecting the FGFR1 and
present among tumour cells, as do oc- nostaining is a consistent feature. Label- the NTRK-family receptor kinase genes,
casional cerebellar examples. Rosenthal ling with immunohistochemical markers and very occasional RAF1 gene fusions
fibres are positive for alpha-B-crystallin, of mTOR pathway activation (e.g. phos- with SRGAP3, which occur in a similar
with GFAP staining limited to their periph- phorylated S6) is more variable {1076l. manner to the BRAF fusions: a tandem
ery. Unlike in diffuse astrocytomas, p53 lmmunohistochemistry for V600E-mutant duplication at 3p25, with the fusion pro-
protein staining is weak to absent, which BRAF is positive in a small subset lacking tein lacking the RAF regulatory domain
is consistent with the rarity of TP53 mu- KIAA1549-BRAFfusion and other MAPK but retaining the kinase domain with loss
tations in pilocytic astrocytoma. lmmu- pathway alterations. The Ki-67 prolifera- of kinase control. In cases with altera-
nohistochemistry for the R132H-mutant tion index is low in most cases of pilocytic tions of the NTRK genes, the alterations
IDH1 protein has been suggested to be astrocytoma, consistent with low prolifer- are also in the form of gene fusions, with
useful for the distinction of pilocytic as- ation rates {826l. Increases in the Ki-67 several different 5' partners that contain
trocytomas from diffuse gliomas, given proliferation index have been associated a dimerization domain. This is presumed
that reactivity is absent in essentially all with more aggressive behaviour in some to lead to constitutive dimerization of the
pilocytic astrocytomas {359l. However, studies but not others {258,2552l NTRK fusion proteins and activation of
paediatric diffuse astrocytic tumours the kinase {1176,2855l The changes are
may also lack IOH1 or IDH2 mutations. Genetic profile more varied in the case of FGFR1. They
In addition, reliable antibodies are cur- The most frequent abnormality in pilocyt- include hotspot point mutations (N546K
rently available for only the most com- ic astrocytomas overall (found in > 70% of and K656E), FGFR1-TACC1 fusions simi-
mon IDH1 amino acid substitutions at all cases) is an approximately 2 Mb dupli- lar to those seen in adult glioblastoma
position 132, so a negative result does cation of 7q34, encompassing the BRAF {2364). a novel internal duplication of the

86 Other astrocytic tumours


and has also been reported in dysem-
bryoplastic neuroepithelial tumours (414,
2280). This mutation can now be identi-
fied using a monoclonal antibody that
specifically recognizes the V600E-mu-
tant BRAF protein (355).
A summary of genetic aberrations report-
ed to date in pilocytic astrocytoma, as
well as their diagnostic utility, is provided
in Table 2.01 (p. 84).

Genetic susceptibility
Pilocytic astrocytomas are the principal
• •KIAA1549:BRAF CNS neoplasm associated with NF1,
which occurs in approximately 1 in 3000
• •Other RAF fusion births. Although NF1 is inherited in an au-
•BRAF mut
tosomal dominant manner, about half of
all cases are associated with a de novo
• •KRASmut mutation (see p. 294). The NF1 gene,
located on the long arm of chromosome
•FGFRl mut/fus/dup •NFl (germline)
17, encodes the neurofibromin protein,
•NTRK fusion •None identified which acts in the MAPK pathway as a
Fig. 2.10 Pilocytic astrocytoma. Pie charts summarizing the known frequencies of the various MAPK pathway alterations GAP for RAS, thereby facilitating the
in different anatomical locations of the brain (i.e. posterior Iossa, diencephalon, and cerebral hemispheres), calculated deactivation of RAS. Patients with NF1
from a total of 188 pilocytic astrocytoma cases {482, 1176,2855). Reprinted from Collins VP et al. (482). have only one wildtype NF1 gene copy.
Subsequent functional loss of this single
kinase domain of FGFR1, and an inter- 1176,1824,2855). Pilocytic astrocytomas wildtype copy results in the absence of
nal tandem duplication of FGFR1 {1176, with anaplastic change have yet to be the NF1-encoded GTPase and conse-
2855). A summary of the incidence of studied in any detail. However, in a recent quent overactivity of RAS and the MAPK
all MAPK pathway alterations identified study of 26 cases that had progressed to pathway. This so-called second hit in
to date in pilocytic astrocytomas of all high-grade glioma, none had a BRAFfu- pilocytic astrocytomas can occur as a
anatomical sites is shown in Table 2.01 sion (1683). result of point mutation, focal or broader
(p. 84). Chromosomal polysomies (in The presence of the KIAA1549-BRAF LOH, or DNA hypermethylation (909}.
particular of chromosomes 5, 6, 7, 11, fusion in an appropriate morphological Reports have suggested that germline
and 15) have been reported in tumours context and together with other findings mutations affecting the 5'-most third of
of teenagers and adults (1177,2237). supports the diagnosis of pilocytic astro- the NF1 gene may be associated with a
The incidence of the various MAPK path- cytoma. However, the absence of such greater chance of developing pilocytic
way alterations is not consistent across a fusion provides no diagnostic informa- astrocytomas of the optic pathway (235,
all anatomical locations {123,1113,1176, tion, because there are so many other 2332). although further study is needed.
2855) The KIAA1549-BRAF fusion is ways in which the MAPK pathway has Approximately 15% of individuals with
extremely common in the cerebellum been found to be activated in pilocytic NF1 develop pilocytic astrocytomas
(found in -90% of cases) but less com- astrocytomas. The KIAA1549-BRAF fu- (1481}. particularly of the optic nerve/
mon supratentorially (found in -50%). sion has also been reported in several pathways (optic pathway glioma), but
FGFR1 alterations are restricted mainly adult gliomas but rarely in other histolo- other anatomical sites (sometimes multi-
to midline structures, whereas the BRAF gies, with the exception of the rare diffuse ple) can also be affected. Conversely, as
V600E mutation and NTRK-family fusions leptomeningeal glioneuronal tumour (see many as one third of all patients with a
are more common in supratentorial tu- p. 152; formerly referred to as dissemi- pilocytic astrocytoma of the optic nerve
mours. Variation is also observed in the nated oligodendroglial-like leptomen- have NF1 (1304). These tumours typically
transcriptome and methylome of pilocytic ingeal neoplasm), in which the fusion is arise during the first decade of life; few
astrocytomas, with infratentorial and su- frequently found together with a solitary patients with NF1 develop an optic path-
pratentorial tumours distinguishable on 1p deletion (2156). Unfortunately, many way glioma after the age of 10 years.
the basis of their gene expression or of the non-KIAA1549-BRAF alterations Pilocytic astrocytomas are also associat-
DNA methylation signatures {1424,2335, are also found to be similarly aberrant in ed with Noonan syndrome (a neuro-car-
2527). The reason for this relationship the tumour types that are most frequently dio-facial-cutaneous syndrome), which is
between site I cell of origin and certain among the differential diagnosis of pi- caused by germline mutations of MAPK
molecular alterations is unclear. Unlike in locytic astrocytoma. For example, the pathway genes (i.e. PTPN11, SOS1,
diffuse astrocytomas, the average muta- BRAF V600E mutation occurs in a small KRAS, NRAS, RAF1, BRAF, SHOC2,
tion rate is low in pilocytic astrocytomas. subset of pilocytic astrocytomas, but is and CBL) (83,2135). The PTPN11 gene
TP53 mutations seem to play no role in a common alteration in gangliogliomas is mutated in about 50% of patients with
the development of these tumours {1098, and pleomorphic xanthoastrocytomas Noonan syndrome and is occasionally

Pilocytic astrocytoma 87
mutated (together with FGFR1) in spo- either recur or progress, eventually lead- a wide spectrum of morphologies and
radic pilocytic astrocytomas {1176). How- ing to death. However, this is usually after behaved in a more aggressive manner,
ever, the number of pilocytic astrocyto- a prolonged clinical course with multiple with decreased survival, than typical pi-
mas reported in patients with Noonan recurrences {725,974,1678,1996,2159). locytic astrocytomas. However, despite
syndrome is small {744,1743,2233,2300). Pilocytic astrocytomas of the optic nerve exhibiting morphological features of
in patients with NF1 {2148) seem to have anaplasia similar to those of diffuse as-
Prognosis and predictive factors a more indolent behaviour than that of trocytic tumours, they did not behave as
Pilocytic astrocytoma is typically a slow- their sporadic counterparts {1516). aggressively. Even so-called high-grade
growing, low-grade tumour with a favour- Histological malignancy in pilocytic as- pilocytic astrocytoma with anaplastic fea-
able prognosis. Overall survival rates at 5 trocytoma is rare; in one study of clas- tures and necrosis did not behave like a
and 10 years are> 95% after surgical in- sic pilocytic astrocytoma, the incidence glioblastoma. Pilocytic astrocytomas with
tervention alone {291,325,688,725,1606, of malignancy occurring spontaneously anaplastic features such as increased mi-
1826). Stability of WHO grade is typically was 0.9%, and the incidence of occur- totic count (i.e. > 4 mitoses per 10 high-
maintained for decades {149,325,1878). rence after radiation was 1.8% {2565). power fields) and necrosis behaved, in
The tumours may even spontaneously Descriptions of anaplastic features in pi- respective studies, more similarly to dif-
regress, although this is rare {897,1894, locytic astrocytomas come mainly from fuse low-grade or anaplastic astrocyto-
2421 ). Very occasional cases progress isolated case reports and small series mas than to anaplastic astrocytomas or
with more anaplastic features after a vari- {374,591,1251,2417,2682). in which ma- glioblastomas {2150).
able interval, most often after irradiation lignant histological features correlate less Pilomyxoid astrocytoma (see below), a
or chemotherapy treatment {1957,2145). reliably with prognosis than those seen recognized pilocytic astrocytoma vari-
There are few long-term studies docu- in patients with diffusely infiltrative astro- ant typically occurring in young children,
menting the ultimate outcome of patients cytomas. Malignant transformation is fre- almost exclusively in the hypothalam-
with pilocytic astrocytoma. quently reported in association with prior ic I third ventricular region, reportedly
Patient age and extent of resection are treatment {61,176,307,1296,1364,2303, has a higher frequency of local recur-
key prognostic factors {2431} Depend- 2312,2420,2565,2603) A retrospective rence and may undergo cerebrospinal
ing on the location and size of the tumour, study of 34 pilocytic astrocytomas with fluid seeding {1534,2555).
pilocytic astrocytoma may not be ame- spontaneous anaplastic histological fea-
nable to gross total resection and may tures found that these tumours exhibited

Pilomyxoid astrocytoma Burger P.C. Jones D. Clinical features


Tihan T. Giannini C. Pilomyxoid astrocytomas present with
Hawkins C. Rodriguez F. non-specific signs and symptoms that de-
VandenBerg S.R. Figarella-Branger D. pend on the anatomical site. Radiological
examination shows a well-circumscribed
mass with relatively distinct borders. The
Definition an increased likelihood of recurrence, it tumour is typically T1-hypointense and
A variant of pi!ocytic astrocytoma histo- is uncertain whether pilomyxoid astrocy- T2-hyperintense. Compared with pile-
logical/y characterized by an angiocen- toma truly corresponds histologically to cytic astrocytomas, pilomyxoid astrocy-
tric arrangement of monomorphous, bi- WHO grade II. Therefore, a definite grade tomas are more likely to have necrosis
polar tumour cells in a prominent myxoid assignment is not recommended at this and more likely to have signal intensity
background {2555). time. extending into adjacent structures. Cysts
Pilomyxoid astrocytoma is preferentially and calcifications are less common
located in the hypothalamic/chiasmatic Epidemiology {1322). Cerebrospinal fluid dissemination
region but shares other locations with The incidence of pilomyxoid astrocytoma can occur before presentation.
pilocytic astrocytoma (e.g. the thalamus, is unknown because this entity is typi-
temporal lobe, brain stem, and cerebel- cally grouped with pilocytic astrocytoma. Spread
lum) {1533). Pilomyxoid astrocytoma pre- Pilomyxoid astrocytoma accounts for a These tumours are usually confined to
dominantly affects infants and young small proportion of tumours historically the site of origin, but they may spread
children (with a median patient age of 10 classified as pilocytic astrocytoma. within cerebrospinal pathways.
months). It may grow more rapidly and
have a less favourable prognosis than Localization Macroscopy
pilocytic astrocytoma, due to local recur- The hypothalamic/chiasmatic region is I ntraoperative reports often describe a
rence and cerebrospinal spread {270). the most common location, although the solid gelatinous mass. The tumours may
tumour can also occur in the thalamus, infiltrate the parenchyma, and a clear
ICD-0 code 9425/3 cerebellum, brain stem, temporal lobe, surgical plane may not be identifiable.
and spinal cord {187,480,688)
Grading Microscopy
Although some reports have suggested Pilomyxoid astrocytoma is dominated by

88 Other astrocytic tumours


pilocytic astrocytoma do not warrant a
diagnosis of pilomyxoid astrocytoma.

lmmunophenotype
lmmunohistochemical staining demon-
strates strong and diffuse reactivity for
GFAP, S100 protein, and vimentin. Some
cases are positive for synaptophysin,
but staining for NFP or chromogranin-A
is typically negative. CD34 expression
has been reported in some cases in the
hypothalamic/chiasmatic region. Stain-
ing for V600E-mutant BRAF is negative
{480) In limited studies, the Ki-67 prolif- astrocytomas, although cases in patients
eration index was found to vary substan- with neurofibromatosis type 1 and pilo-
tially among pilomyxoid astrocytomas, myxoid astrocytoma have been reported
ranging from 2% to 20% {688,1326). (1261). A case occurring in the context of
There is considerable overlap of Ki-67 Noonan syndrome has been reported as
proliferation index values between pilo- well (1743).
Fig. 2.11 Pilomyxoid astrocytomas are usually large, myxoid and pilocytic astrocytomas.
contrast-enhancing masses that are most frequent in the Prognosis and predictive factors
hypothalamic and suprasellar areas.
Cell of origin In general, pilomyxoid astrocytomas are
The cell of origin for pilomyxoid astro- more aggressive and more prone to lo-
a markedly myxoid background, mono- cytoma is unclear. Some reports of pilo- cal recurrence and cerebrospinal spread
morphous bipolar cells, and a predomi- myxoid astrocytoma underscore its close than are pilocytic astrocytomas (480,
nantly angiocentric arrangement. The association with pilocytic astrocytoma, 688), but there is considerable varia-
tumour typically has a compact, non- suggesting a common astrocytic origin. tion in behaviour, and not all are more
infiltrative architecture, but some cases A suggested origin is from radial glia in aggressive than WHO grade I pilocytic
are infiltrative, trapping normal brain ele- proximity to the optic nerve {437,2527) astrocytoma. Given the complexity of
ments such as ganglion cells. The lesion clinical, anatomical, and pathological in-
is composed of small, monomorphous Genetic profile terrelationships, it is unclear whether the
bipolar cells whose processes often ra- Although very few pilomyxoid astrocyto- more aggressive behaviour of some pi-
diate from vessels, producing a form of mas have been included in large-scale lomyxoid astrocytomas is related to their
pseudorosette. As strictly defined, the le- genomic studies, the data obtained to intrinsic pathological features or to their
sion does not contain Rosenthal fibres or date suggest that these tumours are ge- unfavourable hypothalamic/chiasmatic
eosinophilic granular bodies, but mitotic netically similar to WHO grade I pilocytic location. Complete surgical excision,
figures may be present. Vascular prolif- astrocytomas (1034,2855), with some which is the most reliable prognostic
eration is present in some cases, often showing KIAA1549-BRAFfusion (480). factor for pilocytic astrocytoma, cannot
in the form of linear glomeruloid tufts as- usually be achieved in this anatomical
sociated with cystic degeneration. Rare Genetic susceptibility region.
examples are focally necrotic. Focal pilo- To date, no genetic susceptibility has
myxoid changes in an otherwise typical been established for most pilomyxoid

Pilornyxoid astrocytoma 89
Subependymal giant cell astrocytoma Lopes M.B.S.
Wiestler 0.0.
Stemmer-Rachamimov A.O.
Sharma M.C.
Vinters HV
Santosh V

Definition occur in infants, and several congenital dilated vessels are occasionally seen
A benign, slow-growing tumour com- cases diagnosed either at birth or by an- within the tumours. Like other brain neo-
posed of large ganglionic astrocytes, tenatal MRI have been reported {1070, plasms, SEGAs may show a high ratio
typically arising in the wall of the lateral 1630,1964,2067}. of choline to creatinine and a low ratio
ventricles. of N-acetylaspartate to creatinine on
Subependymal giant cell astrocytoma Localization proton MR spectroscopy, which seems
(SEGA) has a strong association with the SEGAs arise from the lateral walls of the to be a valuable tool for the early detec-
tuberous sclerosis syndrome (p. 306). lateral ventricles adjacent to the foramen tion of neoplastic transformation of sub-
of Monro. ependymal nodules {1979}. Leptomen-
ICD-0 code 9384/1 ingeal dissemination with drop metasta-
Clinical features ses has been described {2532}.
Grading Most patients present either with epilepsy
SEGA corresponds histologically to WHO or with symptoms of increased intracrani- Macroscopy
grade I. al pressure. Massive spontaneous haem- SEGAs are typically located in the walls
orrhage has been reported as an acute of the lateral ventricles over the basal
Epidemiology manifestation {2188). With the current ganglia. The tumours are sharply demar-
practice of early screening of patients cated multinodular lesions. Cysts of vari-
Incidence with tuberous sclerosis, many SEGAs are ous sizes are commonly seen. Areas of
SEGA is the most common CNS neo- diagnosed at an initial stage while still remote haemorrhage may be seen due
plasm in patients with tuberous scle- clinically asymptomatic {1378,2188}. to the high vascularity of the tumour. Ne-
rosis, but it is uncertain whether the tu- crosis is rare, but focal calcifications are
mour also occurs outside this setting {26, Imaging common.
1809,2188). The incidence rate of SEGA On CT, SEGAs present as solid, partially
among patients with confirmed tuberous calcified masses located in the walls of Microscopy
sclerosis is 5-15% {26,1809,2188). and the lateral ventricles, mostly near the fora- SEGAs are circumscribed, often calci-
the tumour is one of the major diagnostic men of Monro. lpsilateral or bilateral ven- fied tumours. They are composed mainly
criteria of tuberous sclerosis {1809). tricular enlargement may be apparent. of large, plump cells that resemble gemi-
On MRI, the tumours are usually hetero- stocytic astrocytes and are arranged in
Age and sex distribution geneous, isointense, or slightly hypoin- sweeping fascicles, sheets, and nests.
This tumour typically occurs during the tense on T1-weighted images, and hy- Clustering of tumour cells and perivascu-
first two decades of life and only infre- perintense on T2-weighted images, with lar palisading are common features. The
quently arises de novo after the age of marked contrast enhancement {1094). tumour cells show a wide spectrum of
20-25 years {1809). However, cases can Prominent signal voids that represent phenotypes. Typical appearances range

Fig. 2.14 Subependymal giant cell astrocytom extending Fig. 2.15 Subependymal giant cell astrocytoma (postcontrast axial T1-weighted MRI). A A right subependymal giant
into the left ventricle and causing hydrocephalus. cell astrocytoma near the foramen of Monro, with avid enhancement. B After 3 months of mTOR inhibitor treatment,
the tumour shows decreased size and enhancement.

90 Other astrocytic tumours


from polygonal cells with abundant, and uniform, intense immunoreactivity for
glassy cytoplasm (resembling gemisto- S100 protein (1526,2334,2832). Variable
cytic astrocytes) to smaller, spindle cells immunoreactivity for neuronal markers
within a variably fibrillated matrix. Giant and neuropeptides has been detected in
pyramidal-like cells with a ganglionic ap- SEGAs. The neuron-associated class Ill
pearance are common; these large cells beta-tubulin is more widespread in its dis-
often have an eccentric, vesicular nucle- tribution than any other neuronal epitope,
us with distinct nucleoli. Nuclear pseu- whereas neurofilament is more restricted
doinclusions can be seen in some cases. and mainly highlights cellular processes
considerable nuclear pleomorphism and and a few ganglionic cells (1526). Simi-
multinucleated cells are frequent. A rich larly, SEGA shows variable immunoex-
vascular stroma with frequent hyalinized pression for synaptophysin within the
vessels and infiltration of mast cells and ganglionic component {2334) Focal
lymphocytes (predominantly T lympho- immunoreactivity for NeuN {2832) and
cytes) is a consistent feature (2334) Pa- neuropeptides has also been detected
renchymal or vascular calcifications are (1526). Neural stem cell markers, includ-
frequent (883,1964,2334). and SEGAs ing nestin and SOX2 are also expressed
may demonstrate increased mitotic activ- in SEGAs (1964); however, unlike in cor-
ity; however, these features do not seem tical dysplasias, CD34 immunoreactivity
to denote an adverse clinical course. is not seen. These findings suggest cel-
Similarly, the occasional presence of en- lular lineages with a variable capacity for
dothelial proliferation and necrosis are A divergent phenotypes, including glial,
not indicative of anaplastic progression. neuronal, and neuroendocrine differen-
The rare examples of SEGA outside the tiation. Loss of either hamartin or tuberin
setting of tuberous sclerosis that recur immunoexpression is commonly seen in
have not been reported to show malig- SEGAs, and combined loss occurs in
nant transformation (2808). rare cases (1693).
The Ki-67 proliferation index as deter-
mined by MIB1 monoclonal antibody Cell of origin
staining is generally low (mean: 3.0%), The histogenesis of SEGA is unclear. The
further supporting the benign nature of histological and radiological features of
these neoplasms {913,2333). The TOP2A SEGAs and subependymal nodules are
labelling index is also low (mean: 2.9%) similar, and there is radiological evidence
(2333). Although extremely uncommon, Fig. 2.16 Subependymal giant cell astrocytoma. supporting the development of some
craniospinal dissemination has been re- A Coronal section of the left hemisphere of a patient with subependymal nodules into SEGAs over
tuberous sclerosis, showing a subependymal giant cell
ported in SEGA with an increased Ki-67 time, suggesting that these entities may
astrocytoma (arrowheads} and multiple cortical tubers
proliferation index but without malignant (arrows). B Multiple subependymal nodules on the walls constitute a continuum {241,468). Evi-
features (2532). of the lateral ventricles. dence of biallelic inactivation of the TSC1
Ultrastructural features of neuronal differ- or TSC2 genes and activation of the
entiation, including microtubules, occa- lmmunophenotype mTOR pathway in SEGAs supports the
sional dense-core granules, and (rarely) SEGA has been designated as an as- hypothesis that SEGAs arise as a con-
synapses may be detectable, and bun- trocytoma, but due to its usually mixed sequence of a two-hit mechanism {406).
dles of intermediate filaments are seen in glioneuronal phenotype, it has also been SEGAs demonstrate glial, neuronal, and
the cytoplasmic processes of the spindle called subependymal giant cell tumour mixed neuroglial features (morphologi-
astrocytic cells (302,1009,1188). {302,1526). The tumour cells demon- cal, immunohistochemical, and ultra-
strate variable immunoreactivity for GFAP structural), which suggests a neuroglial

.. � ' -
Fig. 2.17 Subependymal giant cell astrocytoma. A Cellular heterogeneity is typical for these tumours; elongated cells arranged within sweeping fascicles are admixed with large
cells. B Infiltration of inflammatory cells, including mature lymphocytes and mast cells, is a consistent feature. C Multifocal calcification within the tumour and/or blood vessels is
commonly seen.

Subependymal giant cell astrocytoma 91


.,

. -
Fig. 2.18 Subependymal giant cell astrocytoma. A Mixed population of polygonal to ganglionic-like cells. B Collection of large tumour cells, with perivascular pseudorosettes
reminiscent of ependymoma.

progenitor cell of origin with the capacity within the cell and form a complex {486, these two genes is 1:1 (2231 l. TSC1 or
to undergo differentiation along glial, neu- 1212, 1987). A mutation of either gene re- TSC2 mutations are identified in about
ronal, and neuroendocrine lines {1526, sults in disrupted function of the tuberin- 85% of patients with tuberous sclerosis.
2334). This hypothesis is supported by hamartin complex, with similar resulting The remaining 15% of cases may be
data from mouse models in which loss of disease phenotypes. mosaics or have a mutation in an unana-
Tsc1 or activation of the mTOR pathway Approximately 60% of patients with tu- lysed non-coding gene area. Mosaicism
in subventricular zone neural progenitor berous sclerosis have sporadic disease has been reported for TSC1 and TSC2
cells resulted in the formation of SEGA (i.e. with no family history), indicating a mutations in some parents of patients
and subependymal nodule-like lesions high rate of de novo mutations {2229A}. with sporadic cases and in patients with
in the lateral ventricles {1566,2862Al. In affected kindreds, the disease follows tuberous sclerosis (2229,2646). Alter-
an autosomal dominant pattern of inherit- natively, there may be a third, unknown
Genetic profile ance, with high penetrance but consider- locus, although to date there is no evi-
SEGA has a strong association with tu- able phenotypic variability (2354). dence to support this possibility (2231).
berous sclerosis, a genetic disease In sporadic tuberous sclerosis cases, Patients with tuberous sclerosis with no
caused by inactivating mutations in the mutations in TSC2 are 5 times more com- identified mutations have a milder pheno-
TSC1 gene at 9q or the TSC2 gene at mon than mutations in TSC1 (51,516, type than do patients with TSC1 or TSC2
16p. The proteins encoded by the TSC 1174}, whereas in families with multiple mutations (2231 l.
genes, tuberin and hamartin, interact members affected, the mutation ratio of Like in other circumscribed astrocytomas,

,i'
Fig. 2.19 Subependymal giant cell astrocytoma. A Tumours may express primitive neural markers, including nestin. B Diffuse nuclear expression of the primitive neural marker
SOX2. C The majority of tumour cells are immunoreactive for GFAP. D Class Ill beta-tubulin (also called TUJ1) is the most ubiquitous neuronal-associated marker in these tumours.
E Focal expression of NFP. F MIB1/Ki-67 immunohistochemistry showing a low proliferation rate.

92 Other astrocytic tumours


including . pilocytic astrocytoma and 1163,1255,1694). LOH at 16p and 21q symptomatic lesions tend to have greater
pleomorph1c xanthoastrocytomas, BRAF has been observed in two separate morbidity {551). Careful follow-up of re-
V600E mutations have been found in cases {2782). and partial loss of chro- sidual tumour is recommended because
sEGAs: in 6 of 14 cases in one series mosome 22q was observed in two ad- of the potential for late recurrences. Opti-
(1448) and in 1 of 3 cases in another ditional paediatric patients with tuberous mal outcome is associated with early de-
{2280}. regardless of whether the tumour sclerosis {560). tection and treatment. In individuals with
occurred in the setting of tuberous scle- tuberous sclerosis, surveillance by MRI
rosis. Investigations of SEGAs have also Genetic susceptibility every 1-3 years until the age of 25 years
demonstrated LOH in the TSC2 gene SEGA has a strong association with the is recommended {551,1378). Inhibition of
{992,1694). Lost or reduced tuberin. and inherited tuberous sclerosis syndrome mTOR1 with everolimus has been report-
hamartin expression has been described (see Tuberous sclerosis, p. 306). ed as a promising therapeutic approach
in SEGAs from patients with both TSC1 in patients with inoperable tuberous scle-
and TSC2 germline mutations, suggest- Prognosis and predictive factors rosis-associated SEGAs {732A.733}.
ing a two-hit tumour suppressor model SEGA has a good prognosis when gross
for the pathogenesis of SEGAs {992, total resection is achieved. Larger or

Subependymat giant cell astrocytoma 93


.,

Pleomorphic xanthoastrocytoma Giannini C.


Paulus W.
Louis D.N.
Liberski P.P.
Figarella-Branger D.
Capper D.

Definition
An astrocytic glioma with large pleomor-
phic and frequently multinucleated cells,
spindle and lipidized cells, a dense peri-
cellular reticulin network, and numerous
eosinophilic granular bodies.
Pleomorphic xanthoastrocytoma tumour
cells are neoplastic astrocytes, but there
is often neuronal differentiation {828,
1007,1254). Mitotic activity is low(< 5 mi-
toses per 10 high-power fields). BRAF
V600E mutation is common in pleomor-
phic xanthoastrocytoma, and its pres-
ence in the absence of an IDH mutation
strongly supports the diagnosis. Pleo-
morphic xanthoastrocytoma is rare (con- Fig. 2.21 Pleomorphic xanthoastrocytoma. T1-weighted MRI with contrast enhancement. A,B The neoplasms in the
stituting< 1% of all astrocytic neoplasms) temporal lobe present as superficial nodular enhancing cystic tumours. Note the scalloping of overlying bone (A).
and most commonly affects children and
young adults, with a median patient age
at diagnosis of 22 years {825). The tu- Epidemiology function is also possible. The rare TP53
mour has a typical superficial location in Pleomorphic xanthoastrocytoma is a rare mutations encountered do not suggest
the cerebral hemispheres, most frequent- brain tumour, accounting for < 1% of all particular carcinogenic insults (824,1238,
ly in the temporal lobe, with involvement primary brain tumours. The 2014 statisti- 1917).
of the adjacent leptomeninges and with cal report published by the Central Brain
cyst formation. Despite its alarming his- Tumor Registry of the United States (CB- Localization
tological appearance, pleomorphic xan- TRUS) lists pleomorphic xanthoastrocy- A superficial location, involving the lep-
thoastrocytoma has a relatively favour- toma among the "unique astrocytoma tomeninges and cerebrum (meningoce-
able prognosis compared with diffusely variants" and reports an annual inci- rebral) is characteristic of this neoplasm.
infiltrative astrocytoma, with 70.9% re- dence of 0.3 cases per 100 000 popula- Approximately 98% of cases occur su-
currence-free and 90.4% overall survival tion {1863}. pratentorially, most commonly in the tem-
rates at 5 years {1081}. Pleomorphic xanthoastrocytoma typical- poral lobe {825,1254). Cases involving
ly develops in children and young adults the cerebellum and spinal cord have also
ICD-0 code 9424/3 {825}, with mean and median patient be reported (836,1749}, and two cases of
ages of 25.9 and 22 years, respectively primary pleomorphic xanthoastrocytoma
Grading {1935}, but occurrence in older patients, of the retina in children have been report-
Pleomorphic xanthoastrocytoma corre- including patients in their seventh and ed (2847).

l
sponds histologically to WHO grade II. eighth decades of life, has also been re-
ported (1935). There is no sex bias. One Clinical features
study in the USA found these tumours to Due to the superficial cerebral location of
Males Females be more common in the Black population the lesion, many patients present with a
90-100
80-89 {1935). fairly long history of seizures. Cerebellar
70-79 and spinal cord cases have symptoms
60-69 Etiology that reflect these sites of involvement.
50-59 No specific etiologies have been im-
plicated in the genesis of pleomorphic Imaging
xanthoastrocytoma. The occasional Pleomorphic xanthoastrocytoma is usu-
association with cortical dysplasia or ally a supratentorial mass, peripherally lo-
with ganglion cell lesions suggests that cated and frequently cystic, involving cor-
60 50 40 30 20 10 0 0 10 20 30 40 50 60
their formation may be facilitated in tex and overlying leptomeninges. CT and
Number of cases Number of cases
malformative states {1409). Given re- MRI scans outline the tumour mass and/
Fig. 2.20 Age and sex distribution of patients with ports in patients with neurofibromatosis or its cyst. On CT, tumour appearance
pleomorphic xanthoastrocytoma. type 1 {2023}, a relation to defective NF1 is variable (hypodense, hyperdense, or

94 Other astrocytic tumours


,, '
" - • .... •'Y.. �
Fig. 2.22 Histological features of pleomorphicxanthoastrocytoma. A Leptomeningeal pleomorphic xanthoastrocytoma, sharply delineated from the underlying cerebral cortex.
B Granular bodies, intensely eosinophilic or pale, are an almost invariable finding. C Tumour cells showing nuclear and cytoplasmic pleomorphism and xanthomatous change.
D Mature ganglion cell and lymphocytic infiltrates; reprinted from Kros JM et al. {1375).

mixed), with strong, sometimes heteroge- Microscopy also frequent {825). The third histologi-
neous contrast enhancement {1857). Tu- The adjective "pleomorphic" refers to the cal hallmark of pleomorphic xanthoas-
mour cysts are hypodense. On MRI, the variable histological appearance of the trocytoma is the presence of reticulin
solid portion of the tumour is either hy- tumour, in which spindled cells are in- fibres, which are best seen using silver
pointense or isointense to grey matter on termingled with mononucleated or multi- impregnation. Reactive changes in the
T1-weighted images and shows a hyper- nucleated giant astrocytes, the nuclei of meninges are not the only source of re-
intense or mixed signal on T2-weighted which show great variation in size and ticulin fibres; individual tumour cells may
and FLAIR images, whereas the cystic staining. lntranuclear inclusions are fre- be surrounded by basement membranes
component is isointense to cerebrospinal quent {825). as are prominent nucleoli. that also stain positively for reticulin, and
fluid. Postcontrast enhancement is mod- In some cases, the neoplastic astrocytes these can be recognized ultrastructurally
erate or strong {1857). Perifocal oedema are closely packed, creating a so-called as pericellular basal laminae. By defini-
is usually not pronounced, due to the epithelioid pattern {1106). In other cases, tion, the mitotic count is < 5 mitoses per
slow growth of the tumour. sheets of fusiform cells are encountered. 10 high-power fields. Necrosis is rarely
The term "xanthoastrocytoma" refers to present in WHO grade II pleomorphic
Macroscopy the presence of large, often multinucleat- xanthoastrocytoma in the absence of
Pleomorphic xanthoastrocytomas are ed xanthomatous cells that have intracel- brisk mitotic activity, but necrosis in itself
usually superficial tumours extending to lular accumulation of lipids. This is usually is insufficient for a WHO grade Ill desig-
the leptomeninges. They are frequently in the form of droplets, which often oc- nation (see Anaplastic pleomorphic xan-
accompanied by a cyst, sometimes cupy much of the cell body, pushing cy- thoastrocytoma, p. 98).
forming a mural nodule within the cyst toplasmic organelles and glial filaments
wall. Features such as invasion of the to the periphery. This feature generally Differential diagnosis
dura {543). predominantly exophytic makes the astrocytic character easy to Because pleomorphic xanthoastrocyto-
growth {1688). multifocality {1629). and recognize by H&E or GFAP stains. Gran- ma often includes a diffusely infiltrating,
leptomeningeal dissemination {1905) are ular bodies (either intensely eosinophilic non-pleomorphic component, diffuse
exceptional. or pale) are a nearly invariable finding astrocytoma is a common differential
{825). Focal collections of small lympho- diagnosis. In addition to the presence of
cytes, occasionally with plasma cells, are histologically more typical pleomorphic

Pleomorphic xanthoastrocytoma 95
,

xanthoastrocytoma. The tumours are


predominantly diploid (1047,2701), occa-
sionally with polyploid populations (1047},
possibly due to subgroups of particularly
bizarre, multinucleated tumour cells.
Comparative genomic hybridization of 50
cases identified loss of chromosome 9 as
the hallmark alteration (present in 50% of
cases) {2713}. Homozygous 9p21.3 de-
letions involving the CDKN2AICDKN28
loci were identified in 6 of 10 cases (60%)
{2713}. Accordingly, loss of CDKN2A
protein expression was documented by
immunohistochemistry in 61 % of pleo-
xanthoastrocytoma components, the expressed in pleomorphic xanthoastro- morphic xanthoastrocytomas (1316}.
combination of BRAF V600E mutation cytoma cells (2083) BRAFV600E muta- BRAF point mutations occur in approxi-
and absence of an IDH mutation sup- tion, a common alteration in pleomorphic mately 50-78% of cases (584,601,1082,
ports the diagnosis of pleomorphic xanthoastrocytoma, can be detected by 1316,2277,2280). Most are of the V600E
xanthoastrocytoma. Uncommonly, gan- immunohistochemistry using a specific type, with only a single documented ex-
glioglioma can present with a glial com- antibody (1082,2280). The immunophe- ception (2280}. However, BRAF V600E
ponent resembling pleomorphic xan- notype with V600E-mutant BRAF expres- mutations are not specific to pleomor-
thoastrocytoma; rare cases of composite sion and loss of CDKN2A expression phic xanthoastrocytoma, and are also
tumours with features of pleomorphic (secondary to homozygous deletion of observed in other primary CNS tumours,
xanthoastrocytoma and ganglioglioma CDKN2A) is frequently observed in pleo- in particular ganglioglioma and pilocytic
in which the two neoplastic compo- morphic xanthoastrocytoma (1316) astrocytoma (414,599,1297,2277,2280}
nents coexist side by side with minimal The occurrence of BRAF mutations
intermingling have been reported {1339, Prol!feration seems to be unrelated to the presence of
1943). Pleomorphic xanthoastrocytoma In most pleomorphic xanthoastrocyto- histological features of anaplasia.
areas showing eosinophilic granular mas, mitotic figures are rare or absent, Two studies investigating a total of 7 non-
bodies and spindle-shaped cells may and the Ki-67 proliferation index is gener- BRAF-mutant pleomorphic xanthoastro-
be reminiscent of pilocytic astrocytoma, ally c 1% {825) cytomas identified 1 tumour harbouring
but areas with more typical histology, a TSC2 mutation, 1 with an NF1 mutation,
in the absence of BRAF translocations Cell of origin and 1 with an ETV6-NTRK3 fusion (184,
or other genetic aberrations leading to As originally proposed by Kepes, Rubin- 2855) In four series (one published only
MAPK activation, support the diagnosis stein, and Eng in 1979 {1254), it has been in abstract form) with a total of 123 tu-
of pleomorphic xanthoastrocytoma. Mes- postulated that pleomorphic xanthoas- mours, mutations in the TP53 gene were
enchymal tumours may also enter the dif- trocytoma originates from subpial astro- found in only 7 cases (6%) {824,1238,
ferential diagnosis, but this consideration cytes. This hypothesis would explain the 1917), and were unrelated to the pres-
is usually refuted by positivity for GFAP in superficial location of most cases, and ence of histological features of anaplasia.
unequivocal tumour (non-reactive) cells, is supported by ultrastructural features Amplifications of the EGFR, CDK4, and
although GFAP positivity may be focal or shared between subpial astrocytes and MDM2 genes were absent in a series of
even absent in small specimens. the neoplastic cells in pleomorphic xan- 62 tumours (1238). No IDH1 mutations
thoastrocytoma, in particular the pres- were detected in a series of 7 tumours
lmmunophenotype ence of a basal lamina surrounding indi- (by sequencing) (118} or in a series of
Although the essential nature of pleo- vidual cells. However, the expression of 60 cases (by immunohistochemistry for
morphic xanthoastrocytoma is clearly neuronal markers (828} and CD34 {1316, R132H-mutant IDH1) {1081}. These find-
and uniformly glial, with nearly invariable 2083} in many pleomorphic xanthoastro- ings molecularly distinguish pleomorphic
immunoreactivity for GFAP and S100 cytomas, as well as the occasional as- xanthoastrocytoma from diffusely infiltrat-
protein {825,828}, the tumours have a sociation with cortical dysplasia (1271), ing cerebral astrocytoma.
significant tendency to exhibit neuronal suggests a more complex histogenesis Although there are few data on the oc-
differentiation. Expression of neuronal and a possible origin from multipotent currence of BRAFfusions in pleomorphic
markers (including synaptophysin, neu- neuroectodermal precursor cells or from xanthoastrocytoma, there was no evi-
rofilament, class Ill beta-tubulin, and a pre-existing hamartomatous lesion. dence of 7q34 duplication, which is often
MAP2) has been reported with variable associated with BRAF fusion, in a series
frequency in tumours that have otherwise Genetic profile of 10 cases analysed by microarray-
typical histological features of pleomor- Complex karyotypes have been docu- based comparative genomic hybridiza-
phic xanthoastrocytoma {828,2013). In mented, with gains of chromosomes 3 tion {1961).
some cases, this biphenotypic glioneu- and 7 and alterations of the long arm of
ronal appearance has been confirmed ul- chromosome 1 (1495,2253,2254}, but Genetic susceptibility
trastructurally {1007). CD34 is frequently these are not specific to pleomorphic There are no distinct associations with

96 Other astrocytic tumours


hereditary tumour syndromes, with the
exception of several reports of pleo-
rnorphic xanthoastrocytoma m patients
with neurofibromatosis type 1 (14,1380,
1751,1870) Given the high frequency of
MAPK pathway alterations observed in
sporadic pleomorphic xanthoastrocy-
torna, an association with NF1 is not un-
expected. Familial clustering of pleomor-
phic xanthoastrocytoma has not been
documented. One case of a pleomorphic
xanthoastrocytoma with classic histologi-
cal and molecular features developing in
DiGeorge syndrome has been reported
(860)

Prognosis and predictive factors


Pleomorphic xanthoastrocytoma behaves
in a less malignant fashion than might be
suggested by its highly pleomorphic his-
tology 11254), but it has an only relatively
favourable prognosis. In a retrospective
series of 74 cases, 54 patients with WHO
grade II pleomorphic xanthoastrocytoma
had an average 5-year recurrence-free
survival rate of 70.9% and 5-year over-
all survival rate of 90.4% (1081 ). Extent of
resection was confirmed to be the most
significant predictive factor of recurrence (74.4%} (1081). The estimated 5-year grade Ill) pleomorphic xanthoastrocy-
1825,1081). Among patients whose tu- overall survival rate for these patients was toma. BRAFV600E mutation status is not
mours had low mitotic activity (i.e. < 5 mi- 89.4%, and was similar between children significantly different between paediatric
toses per 10 high-power fields), the esti- (92.9%} and adults (86.8%) {1081). and adult tumours {1081). It is unclear
mated recurrence-free survival rate was BRAF V600E mutation is more com- whether the presence of BRAF V600E
73.7%, and was similar between paedi- mon in WHO grade II pleomorphic xan- mutation has prognostic significance.
atric patients (73.5%) and adult patients thoastrocytoma than in anaplastic (WHO

Pleomorphic xanthoastrocytoma 97
Anaplastic pleomorphic Giannini C
Paulus W
xanthoastrocytoma Louis ON.
Liberski P.P
Figarella-Branger 0.
Capper 0.

Definition pleomorphic xanthoastrocytoma. In one xanthoastrocytoma, manifesting both at


A pleomorphic xanthoastrocytoma with series of 74 cases, anaplasia was pres- initial diagnosis and at recurrence, may
� 5 mitoses per 10 high-power fields. ent in 23 cases {31%) at first diagnosis, demonstrate less pleomorphism and a
Patients with anaplastic pleomorphic in 23% of paediatric cases. and in 37% of more diffusely infiltrative pattern than
xanthoastrocytoma have significantly adult cases {1081 ). typical classic WHO grade II pleomor-
worse survival than those whose tumours phic xanthoastrocytoma. Although his-
show < 5 mitoses per 10 high-power Localization tological patterns of anaplasia have not
fields 1825,1081}. Necrosis may be pres- Like WHO grade II pleomorphic xan- been formally studied in pleomorphic
ent, but its significance in the absence thoastrocytoma, anaplastic pleomor- xanthoastrocytoma, small-cell, fibrillary,
of increased mitotic activity is unknown phic xanthoastrocytoma is typically a and epithelioid/rhabdoid transformation
(1081). supratentorial tumour, with the temporal have been reported (1248).
The frequency of BRAF V600E mutation lobe being the most commonly involved Anaplastic pleomorphic xanthoastrocy-
is lower among anaplastic pleomorphic lobe. toma may be difficult to distinguish from·
xanthoastrocytomas than among WHO epithelioid glioblastoma (see p. 50) both
grade II pleomorphic xanthoastrocyto- Clinical features histologically and molecularly, because
mas. and the prognostic significance of The signs and symptoms of anaplastic both tumour types commonly exhibit
the mutation is unknown {1081,2290). pleomorphic xanthoastrocytoma are sim- BRAF V600E mutations {1297); in fact,
ilar to those of WHO grade II pleomorphic pleomorphic xanthoastrocytoma recur-
ICD-0 code 9424/3 xanthoastrocytoma. Seizures are the ring as epithelioid glioblastoma has also
most common presenting symptom. been described 12508). Anaplastic pleo-
Grading morphic xanthoastrocytomas lack the .
Anaplastic pleomorphic xanthoastrocy- Imaging cytological uniformity typical of epithe-
toma corresponds histologically to WHO Like WHO grade II pleomorphic xan- lioid glioblastomas and have eosinophilic
grade Ill. thoastrocytoma, anaplastic pleomorphic granular bodies, which are not seen in
xanthoastrocytoma is often a circum- epithelioid glioblastoma (1297,1299). In
Epidemiology scribed supratentorial mass, peripherally many anaplastic pleomorphic xanthoas-
No specific epidemiological data are located and frequently cystic, involving trocytomas, a lower-grade component
available regarding anaplastic pleomor- the cortex and overlying leptomeninges. with features typical of pleomorphic xan-
phic xanthoastrocytoma compared with thoastrocytoma is present at least focally
Microscopy (825). There have been rare reports of
Anaplasia in pleomorphic xanthoastro- a SMARCB1-deficient, atypical teratoid/
cytoma typically manifests as brisk mi- rhabdoid tumour-like lesion arising i ·
totic activity in a tumour that otherwise pleomorphic xanthoastrocytoma, associ
retains all the diagnostic histological ated with loss of SMARCB1 expression i
features of pleomorphic xanthoastrocy- the morphologically distinct high-grad
toma. High levels of mitotic activity may rhabdoid component (396,1157). Sue
be focal or diffuse. Necrosis is frequently clonal evolution may be associated wit
present, almost always in association a highly aggressive biology even whe
with brisk mitotic activity. Microvascular the original tumour is low-grade; the ter
proliferation is uncommon and usually "SMARCB 1-deficient anaplastic plea
associated with brisk mitotic activity and morphic xanthoastrocytoma" has bee
necrosis. Anaplasia may be present at suggested for such a lesion.
the time of first diagnosis or at the time
of recurrence, suggesting progression /mmunophenotype
from a lower-grade to an anaplastic pleo- The phenotypic profile of anaplasti
morphic xanthoastrocytoma. However, pleomorphic xanthoastrocytoma is sirni
some tumours that display features of lar to that of WHO grade 11 pleomorphi
anaplasia at first resection occasionally xanthoastrocytoma.
demonstrate features at the upper lim-
Fig. 2.25 Anaplastic pleomorphic xanthoastrocytoma.
T1-weighted MRI with contrast sequences demonstrates its of a WHO grade II tumour at the time Genetic profile
a large non-homogeneously enhancing tumour with of recurrence, likely reflecting tumour The genetic alterations specific to ana•
mass effect and moderate surrounding oedema. heterogeneity. Anaplastic pleomorphic plastic pleomorphic xanthoastrocytom

98 Other astrocytic tumours


- .
Fig. 2.26 Anaplastic pleomorphic xanthoastrocytoma. A The tumour shows classic features with pleomorphic and xanthomatous (arrow) cells and (B) brisk mitotic activity (arrows),
including atypical mitoses. C The tumour recurred 1 year later and showed remaining areas with pleomorphic morphology and (D) monomorphous areas with epithelioid and rhabdoid
cells with mitotic activity (arrows).

are unknown. Although some anaplas- Prognosis and predictive factors survival rate for patients with tumour ne-
tic xanthoastrocytomas may develop A consistent relationship between mitotic crosis was worse than that for those with-
through malignant progression from activity and outcome in pleomorphic xan- out (42.2% vs 90.2%, P = 0.0002). The
WHO grade 11 pleomorphic xanthoastro- thoastrocytoma has emerged, whereas dataset was insufficient to detect a differ-
cytoma, the sequence of underlying ge- the relationship between necrosis and ence in survival between patients whose
netic events has not yet been determined. outcome remains unclear !825,1081). tumours had � 5 mitoses per 10 high-
The frequency of BRAF V600E mutation One study found that a mitotic count of power fields and necrosis (n = 14; 7 pa-
is lower in anaplastic pleomorphic xan- � 5 mitoses per 10 high-power fields (with tients died and 6 cases recurred) and
thoastrocytoma (9 of 19 cases [47.4%] one high-power field covering 0.23 mm2) those whose tumours had � 5 mitoses
in one study) than in pleomorphic xan- can be used to establish the diagnosis. per 10 high-power fields but no necrosis
thoastrocytoma (30 of 40 cases [75.0%] In that study, significant mitotic activ- (n = 5; 2 patients died and 3 cases re-
in a separate study), and its prognostic ity (defined as � 5 mitoses per 10 high- curred). Of the 2 patients whose tumours
significance is unknown !1081,2290). power fields) and/or necrosis was en- had necrosis but not increased mitotic
BRAF V600E mutation status is not sig- countered in 31% of tumours at initial activity, one had no evidence of disease
nificantly different between paediatric presentation !1081). These features were after 10 years of follow-up; the other had
and adult cases !1081). found to be associated with shorter sur- only limited follow-up (1 month), but was
vival l1081 ). The 5-year overall survival still alive at that time. Between children
Genetic susceptibility rate for patients whose tumours showed and adults, there were no significant
No distinct associations between ana- � 5 mitoses per 10 high-power fields was differences in 5-year recurrence-free
plastic pleomorphic xanthoastrocytoma significantly worse than that for patients survival (67.9% vs 62.4%, P = 0.39) or
and hereditary tumour syndromes have whose tumours showed < 5 mitoses per 5-year overall survival (87.4% vs 76.3%,
been reported. 10 high-power fields (89.4% vs 55.6%, P = 0.83). The prognostic significance of
P = 0.0005) And the 5-year overall BRAFV600E mutation remains unknown.

Anaplastic pleomorphic xanthoastrocytoma 99


CHAPTER 3

Ependymal tumours
Subependymoma
Myxopapillary ependymoma
Ependymoma
Ependymoma, RELA fusion-positive
Anaplastic ependymoma
Subependymoma Mclendon R.
Schiffer D.
Rushing E.J.
Hirose T.
Rosenblum M.K. Santi M.
Wiestler O.D.

Definition
A slow-growing, exophytic, intraventric-
ular glial neoplasm characterized by
clusters of bland to mildly pleomorphic,
mitotically inactive cells embedded in an
abundant fibrillary matrix with frequent
microcystic change.
Subependymomas are often detected in-
cidentally, by neuroimaging or at autopsy,
and have a very favourable prognosis
11126,1211,1533,2794). Some tumours
have the admixed histological features of
both subependymoma and ependymoma.

ICD-0 code 9383/1

Grading
Subependymoma corresponds histologi-
cally to WHO grade I.

Epidemiology
The true incidence of subependymoma Fig. 3.02 A Subependymoma filling the right lateral ventricle, with displacement of the septum pellucidum to the
contralateral hemisphere. The tumour is sharply delineated and only focally attached to the ventricular wall; the
is difficult to determine, because these
cut surface is greyish-white with some small haemorrhages; note the old cystic infarct in the left corpus caudatum
tumours frequently remain asymptomatic (arrowhead). B Large subependymoma filling the left ventricle and a smaller one in the right ventricle. C Posterior Iossa
and are often found incidentally at autop- subependymoma in the caudal region of the fourth ventricle. Note the compression of the dorsal medulla (arrowheads).
sy. In two studies, subependymomas ac- D Third ventricle subependymoma (arrowheads).
counted for approximately 8% of ependy-
mal tumours 11398,2274), and in one of Localization with motor and sensory deficits accord-
the studies, they accounted for 0.51% of Subependymomas are distinguished by ing to the affected anatomical segment.
all CNS tumours resected at a single in- their intraventricular location, sharp de- Incidental detection of asymptomatic
stitution 11398). marcation, slow growth, and usually non- subependymomas at autopsy is common
Subependymomas develop in both sex- invasive behaviour. The most frequent {2201).
es and in all age groups, but occur most site is the fourth ventricle (accounting for
frequently in middle-aged and elderly 50-60% of cases), followed by the lateral Imaging
patients. The male-to-female ratio is ap- ventricles (accounting for 30-40%). Less Subependymomas present as sharply
proximately 2.3:1 12056,2201 ). common sites include the third ventricle demarcated nodular masses that are
and septum pellucidum. In rare cases, usually non-enhancing. Calcification and
tumours occur intraparenchymally in the foci of haemorrhage may be apparent.
cerebrum {1279). In the spinal cord, sub- I ntramedullary cases are typically ec-
ependymomas manifest as cervical and centric in location, rather than centrally
cervicothoracic intramedullary or (rarely) positioned as is typical of intraspinal
extramedullary masses 11130,2201) ependymomas. The lesions are hypoin-
tense to hyperintense on both T1- and T2-
Clinical features weighted MRI, with minimal to moderate
Subependymomas may become clinical- enhancement 12056,2201 ).
ly apparent through ventricular obstruc-
tion and increased intracranial pressure. Macroscopy
30 20 10 10 20 30
Spontaneous intratumoural haemorrhage These tumours present as firm nodules of
Number of cases Number of cases has been observed {31,369). Rare in- various sizes, bulging into the ventricular
Fig. 3.01 Age and sex distribution of subependymoma, traparenchymal tumours exhibit marked lumen. In most cases, the diameter does
based on 167 cases; data from the Central Brain Tumor oedema and are associated with sei- not exceed 1-2 cm. lntraventricular and
Registry of the United States (CBTRUS), 1995-2002. zures {2056). Spinal tumours manifest spinal subependymomas are generally

102 Ependymal tumours


Fig. 3.03 Subependymoma. A Subependymomas have a coarse fibrillar matrix and contain clusters of uniform nuclei. B Microcysts are common. C Cells are uniform, often
appearing as bare nuclei against the fibrillary matrix.

well demarcated. Large subependymo- lmmunophenotype a set of 41-year-old identical twins 11799).
mas of the fourth ventricle may cause lmmunoreactivity for GFAP is usually in a set of 22-year-old identical twins
brain stem compression. Rare cases present, although to various extents, and {464). and in a brother and sister aged
arising in the cerebellopontine angle immunoreactivity can also be found for 28 and 31 years, respectively 1430). In a
have been described in both adults and neural markers of low specificity, such family with several brain tumours, three
children 11048). as NCAM1 and neuron-specific enolase siblings developed subependymomas of
12832). Unlike in classic ependymomas, the fourth ventricle: two brothers (aged
Microscopy EMA is rarely expressed in subependy- 27 and 57 years) and a sister (aged
Subependymomas are characterized by momas. One study of potential therapeu- 49 years). In addition, a younger sibling
clusters of small uniform nuclei embedded tic targets reported that TOP2B, MDM2, (aged 13 years) had a pontine tumour
in a dense fibrillary matrix of glial cell pro- nucleolin, HIF1-alpha, and phosphoryl- of undetermined pathology and an el-
cesses with frequent occurrence of small ated STAT3 are frequently expressed in der brother (aged 57 years) developed
cysts, particularly in lesions originating in subependymomas {1329). a fourth ventricular ependymoma {1032).
the lateral ventricles. Tumour cell nuclei A prenatal, maldevelopmental origin of
appear isomorphic and resemble those of Cell of origin familial subependymomas has been
subependymal glia. In solid tumours, occa- Proposed cells of origin include sub- suggested 1464). but the occurrence in
sional pleomorphic nuclei may be encoun- ependymal glia 1101,1716). astrocytes a family with several brain tumours and
tered; however, nuclear variation is typical of the subependymal plate, ependymal in a father (aged 47 years) and son (aged
in multicystic tumours. Some subependy- cells {2205). and a mixture of astrocytes 22 years) is more suggestive of a genetic
momas exhibit low-level mitotic activity, and ependymal cells 1746,2257). susceptibility {1032,2209)
but this is exceptional. Calcifications and
haemorrhage can occur. Prominent tumour Genetic profile Prognosis and predictive factors
vasculature may rarely be accompanied by A recent study using DNA methylation Subependymomas have a good progno-
microvascular proliferation. Occasionally, profiles of ependymomas from all age sis 11126). To date, no recurrences after
cell processes are oriented around ves- groups and subependymomas from adult gross total resection have been reported
sels, forming ependymal pseudorosettes. patients to characterize the full range of {2257). Complete excision may not be fea-
In some cases, a subependymoma consti- disease identified nine molecular groups sible for all tumours arising from the floor
tutes the most superficial aspect of a clas- of ependymoma across three anatomical of the fourth ventricle; however, debulking
sic ependymoma or (more rarely) a tany- sites: the supratentorial compartment. alone usually yields an excellent progno-
cytic ependymoma !1279); such combined posterior fossa, and spinal compartment sis, given that these tumours grow slowly
tumours are classified as mixed ependymo- 11880). Groups dominated by subepen- and residual tumour may take decades to
ma-subependymoma and are graded on dymomas were found to occur in all three manifest as a symptomatic mass 1321 ). Al-
the basis of the ependymoma component of these anatomical locations. Posterior though recurrences are generally not ex-
12201 ). In one rare example, the subepen- Iossa and spinal subependymomas har- pected with subtotally resected tumours
dymomatous element predominated and boured chromosome 6 copy number al- 12028). rare cases have been reported,
seemed to signify a good overall prognosis terations, whereas supratentorial tumours and cerebrospinal fluid spread has been
12201) Examples of subependymoma with showed virtually none. The supratento- documented as well 12321,2677). Mitotic
melanin formation 12179). rhabdomyosarco- rial and posterior Iossa subependymoma activity is usually low or absent. Scattered
matous differentiation 12566). and sarcoma- groups showed excellent overall survival. mitoses and cellular pleomorphism are
tous transformation of vascular stromal ele- of no clinical significance 12020,2201 ).
ments 11532) have been reported. Genetic susceptibility Ki-67/MIB1 immunohistochemical studies
At the ultrastructural level, subependymo- Familial occurrence of subependymo- have found proliferation index values of
mas show cells with typical ependymal mas is rare, but well documented. All < 1%, compatible with the slow growth of
characteristics, including cilium formation published cases have been located in this entity, although one study of 2 cases
and microvilli, and sometimes with abun- the fourth ventricle. Reports have de- indicated that recurrence rate may corre-
dant intermediate filaments 1101,1716,2274) scribed the simultaneous manifestation late with proliferation index 11354).
of fourth ventricular subependymomas in

Subependymoma 103
Myxopapillary ependymoma Mclendon R.
Schiffer D.
Rosenblum M.K.
Wiestler OD.

Definition The average patient age at presentation


A glial tumour arising almost exclusive- is 36 years, with a range of 6-82 years.
ly in the region of the conus medullaris, In one study of 320 ependymomas of the
cauda equina, and filum terminale, and filum terminale, 83% were of the myxo-
histologically characterized by elongat- papillary type, with a male-to-female ratio
ed, fibrillary processes arranged in radial of 2.21 {395). The youngest patient was
patterns around vascularized, mucoid, an infant aged 3 weeks {459).
fibrovascular cores.
Myxopapillary ependymoma is a slow- Localization
growing variant of ependymoma. It typi- Myxopapillary ependymomas occur
cally occurs in young adults. The tumour almost exclusively in the region of the Fig. 3.05 This macroscopic photograph of a transected
generally has a favourable prognosis, conus medullaris, cauda equina, and fi- myxopapillary ependymoma shows its typical sausage
but can be difficult to resect completely. lum terminale. They may originate from appearance externally, with the mucinous, pinkish-white
When this is the case, residual tumour ependymal glia of the filum terminale to variegated appearance on cut surface.
may recur repeatedly, as the tumour be- involve the cauda equina, and only rarely
comes entangled with spinal nerves. invade nerve roots or erode sacral bone. exhibiting spinal metastatic dissemina-
Multifocal tumours have been described tion at presentation {674,2108).
ICD-0 code 9394/1 {1748). Myxopapillary ependymomas can
occasionally be observed at other loca- Macroscopy
Grading tions, such as the cervicothoracic spinal Myxopapillary ependymomas are lobu-
Histologically, myxopapillary ependymo- cord {2396). the fourth ventricle {1502). lated, soft, and grey or tan. They are of-
ma corresponds to WHO grade I. How- the lateral ventricles {2247). and the brain ten encapsulated. Gelatinous alterations,
ever, this variant may have a more ag- parenchyma {2698). Subcutaneous sac- cyst formation, and haemorrhage may be
gressive biological behaviour in children rococcygeal or presacral myxopapillary apparent.
and a poor outcome after incomplete ependymomas constitute a distinct sub-
resection. group. They are thought to originate from Microscopy
ectopic ependymal remnants {1089). In classic cases, cuboidal to elongated
Epidemiology lntrasacral variants can clinically mimic tumour cells are radially arranged in pap-
Myxopapillary variants account for chordoma. illary fashion around hyalinized fibrovas-
9-13% of all ependymomas {1398,2274). cular cores. Some examples show little
In the conus medullaris I cauda equina Clinical features or no papillary structuring and consist
region, myxopapillary ependymomas Myxopapillary ependymomas are typi- largely of confluent, polygonal tumour
are the most common intramedullary cally associated with back pain, often of cell sheets or fascicles of spindled cells.
neoplasm, with annual incidence rates of long duration. Alcian blue-positive myxoid material ac-
about 0.08 cases per 100 000 males and cumulates between tumour cells and
0.05 cases per 100 000 females {1863). Imaging blood vessels, also collecting in micro-
Myxopapillary ependymomas are typi- cysts (which help to identify largely solid,
cally sharply circumscribed and con- non-papillary examples). Rounded eo-
Males
85+
Females trast-enhancing. Extensive cystic change sinophilic structures (so-called balloons)
75-84
and haemorrhage may be seen. that are periodic acid-Schiff-positive
and exhibit spiculated reticulin staining
65-74
Spread are seen in some cases. Mitotic activity
55-64
In a recent review of 183 patients with and the Ki-67 proliferation index are low
45-54
myxopapillary ependymomas, distant {2019) Histological features of anaplasia
35-44
spinal metastases were found in 17 pa- are most exceptional {98).
20-34
tients (9.3%) and brain metastases in Ultrastructurally, the cells do not show
0-19
11 (6 0%). Factors associated with dis- polarity, but do show adherens junctions
60 40 20 20 40 60
tant treatment failure included young with cytoplasmic thickening and wide
Number of cases Number of cases
Fig. 3.04 Age and sex distribution of myxopapillary age, lack of initial adjuvant radiotherapy, spaces containing amorphous material
ependymoma, based on 311 cases; data from the Central and incomplete excision {2711). Paedi- or loose filaments {2076,2402}. Extracel-
Brain Tumor Registry of the United States (CBTRUS), atric patients are particularly prone to lular spaces, delineated by cells with
1995-2002. basal membranes, contain projected

104 Ependymal tumours


., "
Fig. 3.06 Myxopapillary ependymoma. A Tumour cells accumulate around vessels with mucoid degeneration. B Tumour cells interspersed between large vessels with mucoid
degeneration. C Elongated fibrillary processes extend through myxoid regions to reach a blood vessel. D Myxopapillary ependymoma of the cauda equina. Perivascular tumour
cells consistently express GFAP.

villi {2076). Few cilia, complex interdigi- myxopapillary ependymomas, whereas survival rate of 98.4% after total or par-
tations, and abundant basement mem- labelling for CAM5.2, CK5/6, CK?, CK20, tial resection {2770). Late recurrence
brane structures have been described or 34betaE12 has been reported as ab- and distant metastases can occur after
(2019). with the distinctive feature of sent or exceptional in this setting (1427, incomplete resections in both adults and
some examples being aggregations of 2641} children (35,674) Children have had a
microtubules within endoplasmic reticu- less predictable outcome in some stud-
lum complexes {1018,1019} The pres- Genetic profile ies, even with apparent gross total resec-
ence of adherens junctions and intra- A recent study using DNA methylation tion (2216,2426). In a series of 183 cases,
cytoplasmic lumina with microvilli was profiles to characterize the full range of treatment failure occurred in approxi-
recently confirmed (2686). disease identified nine molecular groups mately one third of the patients. Recur-
of ependymoma across three anatomical rence was mainly local and was more fre-
lmmunophenotype sites: the supratentorial compartment, quent in younger patients and those not
Diffuse immunoreactivity for GFAP dis- posterior fossa, and spinal compartment treated initially with adjuvant radiotherapy
tinguishes myxopapillary ependymomas (1880} Myxopapillary ependymomas (1388,2711). Gross total resection plays
from metastatic carcinomas, chordomas, were found in one molecular group from an important role in improving outcome,
myxoid chondrosarcomas, paraganglio- the spinal compartment. They were char- and adjuvant radiotherapy improves pro-
mas, and schwannomas (1427,2641}. acterized by polyploidy (in particular gression-free survival {1389,2578).
Labelling for S100 or vimentin is also gains across multiple chromosomes) and Although age seems to be the strongest
typical, and reactivity for CD99 and an excellent outcome. predictor of recurrence, expression of
NCAM1 is frequently seen (1427). lm- EGFR has also been cited as a potential
munoreactivity for the AE1/AE3 cytoker- Prognosis and predictive factors biomarker of recurrence {2647).
atin cocktail is a common feature of Prognosis is favourable, with a 5-year

Myxopapillary ependymoma 105


,

Ependymoma Ellison D.W.


Mclendon R.
Korshunov A.
Ng H.-K.
Wiestler O.D. Witt H.
Kros J.M. Hirose T.

Definition anaplastic ependymoma are consid- 0-14 years) to 4.5% (at 15-19 years) to
A circumscribed glioma composed of ered to correspond histologically to WHO 4.0% (at 20-34 years) {1863). In children
uniform small cells with round nuclei in grades II and 111, respectively. However, aged < 3 years, as many as 30% of all
a fibrillary matrix and characterized by no association between grade and bio- CNS tumours are ependymomas. In Can-
perivascular anucleate zones (pseu- logical behaviour or survival has been ada, a mean annual incidence of 4.6 cas-
dorosettes) with ependymal rosettes also definitively established {248,633,703}. es per 100 000 population was estimated
found in about one quarter of cases. Of the various studies of prognostic for ependymomas in infants (2046). In
Classic ependymoma generally has a variables and outcome in ependymoma, the USA, ependymoma is more common
low cell density and a low mitotic count. those that have not found an association in Whites than in African-Americans, with
It very rarely invades adjacent CNS pa- between grade (II vs 111) and progression- an incidence rate ratio of 1.67:1. No such
renchyma to any significant extent. Cilia free or overall survival outnumber those difference is found between Hispanic
and microvilli are seen on ultrastructural that have. The published ratios of grade 11 and non-Hispanic populations {1863).
examination. to grade Ill tumours in series of ependy- In the spinal cord, ependymomas are
Classic ependymomas are mainly in- momas vary widely, from 17:1 to 1:7, and the most common neuroepithelial neo-
tracranial tumours; they do occur in the the explanation for such inconsistent data plasms, accounting for 50-60% of all
spinal cord, but the myxopapillary vari- is multifactorial {633,856,2557). The inter- spinal gliomas in adults {2653). but they
ant is more common at this site. Classic pretation of most histopathological varia- are rare in children {164).
ependymomas occur in both adults and bles used in this classification for grading
children, although most posterior fossa purposes is subjective, and ependymo- Age and sex distr;bution
tumours present in childhood. Ependy- mas are morphologically heterogeneous. Ependymomas can develop in patients
momas have a variable clinical outcome, For example, the significance of grading of any age, with reported patient ages
which is primarily dependent on extent of on the basis of focal microvascular prolif- ranging from birth to 81 years [http://
surgical resection, the use of irradiation eration or focally increased mitotic counts www.cbtrus.org]. However, incidence is
as an adjuvant therapy, and molecular within large areas of bland architectural greatly dependent on histological variant,
group {857,1880). and cytological features is difficult to molecular group, and location. Posterior
Three distinct histopathological pheno- determine. There have been few studies fossa ependymomas are most common
types, which are classified as ependymo- of the prognostic significance of WHO among children, with a mean patient
ma variants (although without particular grade or individual pathological features age at presentation of 64 years {2295).
clinicopathological significance) can be across large trial cohorts in which proper and spinal tumours dominate a second
a prominent component of both classic multivariate analyses of prognostic vari- age peak at 30-40 years. Supratentorial
and anaplastic ependymoma: papillary ables can be performed, and there have ependymomas affect paediatric as well
ependymoma, clear cell ependymoma, been only three studies in which evalua- as adult patients. The overall male-to-
and tanycytic ependymoma. tion of histological variables was defined female ratio is 1.77:1, but this ratio varies
stringently and undertaken by several significantly across different anatomical
ICD-0 code 9391/3 observers {633,856,2557). Due to these sites and molecular groups (1171,1880,
limiting factors, grading is almost never 2046). In the USA, classic and anaplastic
Grading used for therapeutic stratification of pa- ependymoma have an approximate com-
Traditionally, classic ependymoma and tients with ependymoma. Given that spe- bined annual incidence of 0.29 cases in
cific genetic alterations and molecular males and 0.22 in females.
groups have recently been proposed as
/ .......
100

90
prognostic or predictive factors for these Localization
/.,..,
tumours (1560,1880,2767}. the practice Ependymomas may occur along the
80

70
v of histologically grading ependymoma ventricular system or spinal canal, in
./
'#. 60
/ may soon become obsolete altogether. the cerebral hemispheres, or at extra-
·� 50
,.v CNS sites. Overall, 60% of the tumours
140 ./ Epidemiology develop in the posterior fossa, 30% in
u 30

20
J the supratentorial compartment, and
10
/ Incidence 10% in the spinal canal [http://seer
/
In the USA, ependymomas account for cancer. gov/arc hive/csr/1975_2004].
Age at diagnosis 6.8% of all neuroepithelial neoplasms. The In adult patients, infratentorial and spi-
Fig. 3.07 Cumulative age distribution (both sexes) of incidence rate decreases with increasing nal ependymomas occur with almost
ependymoma, based on 298 cases (227 4). patient age at diagnosis, from 5.6% (at equal frequency, whereas infratentorial

106 Ependymal tumours


and lung. Myxopapillary ependymomas
occur in the subcutaneous tissue of the
sacrococcygeal area.

Clinical features
The clinical manifestations depend on
tumour localization. Ependymomas of
the posterior fossa can present with
signs and symptoms of hydrocephalus
and raised intracranial pressure, such
as headache, nausea, vomiting, and diz- Fig. 3.09 Ependymoma in a child, filling the entire
ziness. Involvement of cerebellar and lumen of the fourth ventricle. Note the compression and
brain stem structures may cause ataxia, displacement of the medulla.
visual disturbance, paresis, or cranial
nerve deficits. Patients with supratento- demonstrate histopathological overlap
rial ependymomas may show focal neu- with small cell glioblastoma !1463)
rological deficits or epilepsy, as well as
features of raised intracranial pressure. Microscopy
Enlargement of the head or separation Classic ependymoma is a well-delineat-
of the cranial sutures can be evident in ed glioma with monomorphic cells char-
young babies. Spinal ependymomas can acterized by a variable density and round
present with back pain and focal motor to oval nuclei with speckled nuclear chro-
and sensory deficits or paraparesis. matin. The key histological features are
perivascular anucleate zones (pseudoro-
Imaging settes) and (true) ependymal rosettes.
Gadolinium-enhanced MRI shows The pseudorosettes are composed of
well-circumscribed masses with vari- tumour cells radially arranged around
ous degrees of contrast enhancement. blood vessels, creating perivascular anu-
Ventricular obstruction or brain stem cleate zones of fine fibrillary processes.
displacement and hydrocephalus are The ependymal rosettes and tubular ca-
Fig. 3.08 A Fourth ventricle ependymoma. Note the common accompanying features. Su- nals are composed of bland cuboidal or
enlargement of the aqueduct and hydrocephalus of the pratentorial tumours often exhibit cystic columnar tumour cells arranged around
third ventricle. B Cervical ependymoma. Sagittal MRI components. lntratumoural haemor- a central lumen. Pseudorosettes can be
shows an ependymoma in the upper cervical spinal cord rhage and calcification are occasionally found in practically all ependymomas,
(left, arrowhead) with marked gadolinium enhancement
observed. Gross infiltration of adjacent whereas ependymal rosettes are present
(right), delineated on both sides by a typical cyst.
brain structures and oedema are very in only a minority.
rare. MRI is particularly useful for deter- Cell density can vary considerably in
ependymomas predominate in children mining the relationship with surrounding ependymoma, and a high nuclear-to-
!1385). In children aged < 3 years at structures, invasion along the cerebro- cytoplasmic ratio may not necessarily
presentation, 80% of ependymomas are spinal fluid pathway, and syrinx forma- be associated with brisk mitotic activity
in the posterior fossa !2046). Posterior tion. Cerebrospinal fluid spread is a key or other anaplastic features, particularly
fossa ependymomas are located in the factor for staging, prognostication, and in supratentorial vascular ependymoma,
fourth ventricle and sometimes involve treatment. which demonstrates a distinctive branch-
the cerebellopontine angle; in the fourth ing network of delicate capillary blood
ventricle, 60%, 30%, and 10% of the Macroscopy vessels and focal clear-cell change.
tumours originate in the floor, lateral as- Ependymomas are well-circumscribed Some posterior fossa ependymomas
pect, and roof, respectively !1087,2234) tumours usually arising in or near the contain nodules of high tumour cell
Supratentorial ependymomas arise from ventricular system. They are tan-coloured density, often with an increased mitotic
the lateral or third ventricles (in 60% of and are soft and spongy, occasionally count. This biphasic pattern can accom-
cases) or from the cerebral hemispheres, with gritty calcium deposits. Tumours pany a distinctive cerebriform folding of
without obvious connection to a ventri- arising in the caudal fourth ventricle often the tumour surface.
cle (in 40% of cases). In the spinal cord, flow through the foramina of Luschka and Other histological features include re-
cervical or cervicothoracic localization is Magendie to wrap around the brain stem's gions of myxoid degeneration, intratu-
common among classic ependymomas. cranial nerves and vessels, and have moural haemorrhage, dystrophic calci-
In contrast, the myxopapillary variant been termed "plastic ependymomas" fication, and (occasionally) metaplastic
predominantly affects the conus and !503). Rarely, ependymomas can occur cartilage or bone. Prominent hyalinization
cauda equina. Rare extra-CNS ependy- within the cerebral hemisphere, where of tumour vessels is sometimes found,
momas have been observed in the ova- they are well circumscribed !2356). Other especially in posterior Iossa and spinal
ries !1327). broad ligaments !159). pelvic rare examples, often recurrent tumours, ependymomas. Regions of geographi-
and abdominal cavities, mediastinum, infiltrate the cerebral parenchyma and cal necrosis may be observed in classic

Ependymoma 107
.,

ependymoma, but palisading necrosis located at the luminal surface, junctional other gliomas {1101,1865,2031l. Focal cy-
and microvascular proliferation are only complexes at the lateral surface, and tokeratin immunoreactivity can be seen in
focal features in this tumour, with its lack of a basement membrane at the some cases {2641l. Rarely, ependymo-
bland cytology and low mitotic count. internal surface. The cells may form mi- mas can express neuronal antigens {72,
The interface between tumour and CNS crorosettes into which microvilli and cilia 2036,2154l. L1CAM expression is evident
parenchyma is typically well demarcated, project. Junctional complexes (zonulae in supratentorial ependymomas with a
although evidence of brain tissue infiltra- adherentes) irregularly linked by zonu- C11orf95 rearrangement {1891l
tion may occasionally be encountered. lae occludentes or gap junctions, as well
Three distinct histopathological pheno- as cell processes filled with intermedi- Cell of origin
types, which are classified as ependymo- ate filaments, may also be encountered Stem cells isolated from ependymomas
ma variants (although without particular {858l. A basal lamina may be present at have a radial glia phenotype, suggesting
clinicopathological significance) can be the interface between tumour cells and that radial glia cells are the histogenetic
a prominent component of both classic vascularized stroma. source of these tumours {2526l. Further
and anaplastic ependymoma: papillary research has implicated distinct groups
ependymoma. clear cell ependymoma, lmmunophenotype of stem cells that are specific to anatomi-
and tanycytic ependymoma. lmmunoreactivity for GFAP is usually ob- cal site; cerebral neural stem cells and
Rare ependymomas have been reported served in pseudorosettes, but is more adult spinal neural stem cells are poten-
with lipomatous metaplasia, widespread variable in other elements of the tumour, tial cells of origin for cerebral and spinal
pleomorphic giant cells, extensive tu- such as rosettes and papillae. Ependy- ependymomas. respectively, and these
mour cell vacuolation, melanotic differen- momas typically express 8100 protein origins would explain the predominant
tiation. signet ring cells, and neuropil-like and vimentin {1285l EMA immunoreac- locations of these tumours in the differ-
islands. tivity can be found in most ependymo- ent age groups (1171,1891l. The impor-
mas. with expression along the luminal tance of anatomical site in ependymoma
Ultrastructure surface of some ependymal rosettes or biology is demonstrated by the molecu-
Ependymomas retain the characteristic manifesting as dot-like perinuclear or lar groups of the disease as defined by
ultrastructural properties of ependymal ring-like cytoplasmic structures {1241 l. methylome profiling, which is considered
cells, such as cilia with a 9 + 2 microtubu- OLIG2 expression is characteristically to reflect histogenesis (1880)
lar pattern, blepharoblasts and microvilli sparse in ependymomas compared with

108 Ependymal tumours


• '·, , '",,,. '
'.·�' :-. . '
• I •

'f
'
ii'�.... ,
.
,...
ii•,
' .. ,!.
, It •
,

' .. ,:· •.. .
• s. I �
, I.\
, •
...
. :
... Z
,,.._., 1)
I

< r"
'-7'
Fig. 3.11 Ependymoma. A GFAP immunoreactivity is largely restricted to perivascular tumour cells (1291). B EMA staining with dot-like cytoplasmic reactivity; immunohistochemical
detection of EMA has been used to identify intracytoplasmic microrosettes, a feature found in both typical and tanycytic ependymomas, but infrequently (if at all) in myxopapillary
ependymomas. C EMA immunoreactivity in ependymoma demonstrates round intracytoplasmic microrosettes.

Genetic profile ependymomas {1891). characterize the a standardized incidence ratio of 3.70
Molecular alterations are very common in other two supratentorial groups: ST-EPN- {1722,2570). However, ependymomas
ependymoma and comprise cytogenetic, RELA and ST-EPN-YAP1. The other two do not demonstrate mutations in APC
genetic, epigenetic, and transcriptomic posterior fossa groups (PF-EPN-A and {1849). In a Japanese family, two of four
changes. Ependymomas display a broad PF-EPN-B) match those previously called siblings developed a cervical spinal cord
range of cytogenetic aberrations, most group A and group B in some molecu- ependymoma and one had a schwan-
commonly gains of chromosomes 1q, 5, lar studies {1026,1560,2696,2767) The noma. Neurofibromatosis type 2 was
7, 9, 11, 18, and 20 and losses of chro- final two molecular groups for the spinal excluded, and genetic analysis revealed
mosomes 1p, 3, 6q, 6, 9p, 13q, 17, and cord (including the cauda equina) con- a common allelic loss at 22q11.2-qter in
22 (1267,1351). Supratentorial tumours tain myxopapillary ependymomas and two of the affected siblings. The authors
preferentially show loss of chromosome classic ependymomas, respectively, who studied this family suggested the ex-
9 (372,888,1159,1171,2767,2860); in par- and are termed SP-MPE and SP-EPN. istence of a tumour suppressor gene on
ticular, homozygous deletion of COKN2A In infants and young children, posterior chromosome 22 related to the genesis of
has been recurrently demonstrated in su- fossa ependymomas mainly fall into the familial ependymomas (2827).
pratentorial ependymomas (1639,2008, PF-EPN-A group, whereas PF-EPN-B
2526). Gain of chromosome 1q has been tumours occur mainly in adolescents Prognosis and predictive factors
reported as a reproducible prognostic and adults. Copy number alterations, The identification of clinicopathological
marker in several trial cohorts, being as- particularly gains and losses of whole variables of prognostic value in ependy-
sociated with poor outcome in posterior chromosomes and chromosome arms, momas is an important but challenging
fossa tumours (1266,1351,2767) Mono- characterize PF-EPN-B ependymomas, issue (2275). In particular, the clinical util-
somy 22 and deletions or translocations whereas PF-EPN-A ependymomas show ity of individual histopathological features
of chromosome 22q are particularly com- few copy number alterations. Two molec- or tumour grade remains highly contro-
mon in spinal cord tumours and tumours ular groups, ST-EPN-RELA and PF-EPN- versial {633,776). Gain of chromosome
associated with neurofibromatosis type 2 A, are associated with a particularly poor 1q has been reported to be a potential
(936) The NF2 gene is involved in epen- prognosis (1880,2696,2767) outcome indicator among tested molecu-
dymoma tumorigenesis, and NF2 muta- Posterior fossa ependymomas have a lar markers {372,857,1266,1351,1639).
tions occur frequently in spinal ependy- very low mutation rate and lack recurrent and outcome correlates of molecular
momas {207,621). somatic mutations on analysis by whole- groups may prove significant in the future
Using DNA methylation profiling, several genome sequencing methods (1560, ( 1560, 1880, 2696, 2767).
studies have provided support for the 1891). Supratentorial ependymomas are
existence of distinct molecular groups characterized by a recurrent structural Patient age and extent of resection
among ependymomas {1049,1424,1560, variant, the C11orf95-RELA fusion gene, Children with ependymoma fare worse
1880). These groups show strong re- which is a by-product of chromothrip- than adults. This difference may reflect
lationships to certain anatomical sites sis and occurs in 70% of paediatric su- the more frequent occurrence of paediat-
(see Table 3.01, p. 110). In a large cohort pratentorial ependymomas. ric tumours in the posterior fossa versus
(containing > 500 tumours), three groups the predominantly spinal location for adult
were identified in each of the three CNS Genetic susceptibility tumours. Children aged < 1 year have a
compartments (i.e. supratentorial, poste- Spinal ependymomas occur in neurofi- 5-year overall survival rate of 42.4% (791 ).
rior fossa, and spinal) {1880). Tumours bromatosis type 2, indicating a role of the With increasing age, the 5-year overall
with a subependymomatous morphology NF2 gene in these neoplasms. Other he- survival rate improves, to 55.3% among
were classified into separate spinal, pos- reditary forms of ependymoma are rarely 1-4-year-olds, 74.7% among 5-9-year-
terior fossa, and supratentorial groups, observed (309) Two patients with Tur- olds, and 76.2% among 10-14-year-olds.
called SP-SE, PF-SE, and ST-SE, re- co! syndrome and ependymomas have Extent of surgical resection is consistent-
spectively. Fusion genes involving either been reported, and parental colon can- ly reported to be a reliable indicator of
RELA or YAP1, as previously described cer is associated with an increased risk outcome; gross total resection is associ-
for a large proportion of supratentorial of ependymomas among offspring, with ated with significantly improved survival

Ependymoma 109
,

Table 3.01 Key characteristics of the nine molecular groups of ependymoma; based on data from a single study (1880)
Anatomical Genetic Dominant Age at Molecular groups of ependymoma
Group Outcome
location characteristic pathology presentation
Transcriptome and methylome profil-
Infancy to ing have established the relevance of
ST-EPN-RELA RELA fusion gene Classic/anaplastic Poor
adulthood anatomical site to ependymoma biol-
Supratentorial Infancy to ogy. In a recent study that will likely
ST-EPN-YAP1 YAP1 fusion gene Classic/anaplastic Good
childhood serve as the basis for the future mo-
ST-SE Balanced genome Subependymoma Adulthood Good lecular classification of the disease
{1880}, nine groups of ependymoma
PF-EPN-A Balanced genome Classic/anaplastic Infancy Poor
were described; three for each of the
Genome-wide Childhood to three major CNS anatomical compart-
Posterior Iossa PF-EPN-B Classic/anaplastic Good
polyploidy adulthood ments (i.e the supratentorial com-
PF-SE Balanced genome Subependymoma Adulthood Good partment, posterior fossa, and spinal
Childhood to compartment). The modal patient
SP-EPN NF2 mutation Classic/anaplastic Good age at presentation, clinical outcome,
adulthood
Spinal
and frequencies of histopathological
Genome-wide
SP-MPE Myxopapillary Adulthood Good variants and genetic alterations vary
polyploidy
across these groups.
SP-SE 6q deletion Subependymoma Adulthood Good

{238,1604,1644). In the Children's Oncol- occur. Metastatic disease is associated and grade 111 ependymomas is so unreli-
ogy Group (COG) ACNS0121 trial, re- with a poor prognosis. able {633).
section was an independent risk factor,
irrespective of pathological grade {776f. Histopathology Molecular groups
In another study, which included children One major and unresolved issue con- A molecular classification of the disease
aged < 3 years at presentation, a better cerns the accurate definition of anapla- will likely supersede attempts to use
5-year survival rate was achieved after sia, because an inconsistent relationship histopathological variables in the strati-
complete resection (43%) than after in- between pathological variables and out- fication of patients for adjuvant therapy.
complete resection (36%) {2046l. come has emerged over several dec- Nine molecular groups of ependymoma
ades of study {650,704,857,1643,2178, have recently been identified, three from
Tumour location 2778l. Of the features usually associated each principal anatomical compartment
Tumour site has been identified as an with anaplastic change in gliomas, only across the neuraxis {1880). There is a
important prognostic factor. Supratento- mitotic index, several other indices of strong association between two of these
rial ependymomas are associated with proliferation, and foci of poorly differenti- groups, ST-EPN-RELA and PF-EPN-A,
better survival rates than are posterior ated tumour cells seem to be consistently and a poor outcome (Table 3.01).
fossa neoplasms, especially in children associated with survival in ependymoma,
{649,2060}. Spinal ependymomas have and not in all studies {857,1345,1398,
a significantly better outcome than do 2275,2557l. As a result, very few clini-
intracranial tumours, although late re- cal trials use grade to stratify therapy,
currences (> 5 years after surgery) can because the distinction between grade II

110 Ependymal tumours


. •• • •� -
Fig. 3.13 Clear cell ependymoma, characterized by a
relatively high cell density without significant increase
in proliferation. Note the clear perinuclear cytoplasm
resembling tumour cells in an oligodendroglioma.

Papillary ependymoma Clear cell ependymoma Tanycytic ependymoma


Definition Definition Definition
A rare histological variant of ependy- A histological variant of ependymoma A histological variant of ependymoma
moma characterized by well-formed characterized by an oligodendrocyte-like characterized by arrangement of tumour
papillae. appearance, with perinuclear haloes due cells in fascicles of variable width and
to cytoplasmic clearing. cell density and by elongated cells with
ICD-0 code 9393/3 The clear cell ependymoma variant is spindle-shaped nuclei.
most frequently located in the supraten-
Microscopy torial compartment of young patients. ICD-0 code 9391/3
Ependymomas form linear, epithelial-like
surfaces along their cerebrospinal fluid ICD-0 code 9391/3 Microscopy
exposures. Some ependymomas have The tanycytic phenotype is most com-
a papillary architecture that results when Microscopy monly found in spinal cord ependymo-
exuberant growths occasionally arise in Clear cell ependymomas display an oli- mas and manifests as irregular fascicles
which finger-like projections are lined by godendroglial phenotype, with cytoplas- of elongated cells. The fascicles are of
a single layer of cuboidal tumour cells mic clearing that creates clear perinucle- variable width and cell density. Rosettes
with smooth contiguous surfaces and ar haloes. Most ependymomas with this are rarely seen in these ependymomas,
GFAP-immunopositive tumour cell pro- phenotype are supratentorial vascular and pseudorosettes can be subtle. Like
cesses; in contrast, choroid plexus pap- tumours in young patients {727,1673}, but in other ependymomas, the nuclei display
illomas and metastatic carcinomas form the clear-cell phenotype can occasion- speckled (salt-and-pepper) chromatin,
bumpy, hobnail cellular surfaces that ally be found in posterior fossa or spinal and anaplastic features are uncommon.
do not feature extensive GFAP reactiv- tumours. The term "tanycytic ependymoma" is
ity. Unlike the papillae in choroid plexus Clear cell ependymoma must be distin- used for this variant because its spindly,
tumours, the papillae in ependymomas guished from oligodendroglioma, cen- bipolar elements resemble tanycytes -
lack a basement membrane beneath the tral neurocytoma, clear cell (renal cell) the paraventricular cells with elongated
(neuro-)epithelial cells, which in ependy- carcinoma, and haemangioblastoma. cytoplasmic processes that extend to
momas send fibrillary processes down to Ependymal and perivascular rosettes, ependymal surfaces {711}. Because
a vascular core in the same architectural immunoreactivity for GFAP and EMA, ependymal rosettes are typically absent
arrangement as a pseudorosette. and ultrastructural studies can be helpful and pseudorosettes only vaguely deline-
in this differential diagnosis. Some data ated, these lesions may be mistaken for
suggest that clear cell ependymoma may astrocytomas, in particular pilocytic as-
follow a more aggressive course than trocytomas. However, their ultrastructural
other variants {727). The clear cell tumour characteristics are ependymal.
of the lateral ventricles once classified as
ependymoma of the foramen of Monro
{2876) is now recognized as central neu-
rocytoma in most instances (see Central
neurocytoma, p. 156).

Ependymoma 111
Ependymoma, RELA fusion-positive Ellison D.W.
Korshunov A.
WittH.

Definition not have a specified morphology l1891l


A supratentorial ependymoma character- They exhibit the standard range of archi-
ized by a RELA fusion gene. tectural and cytological features found in
The genetically defined RELA fusion- supratentorial ependymomas, but they
positive ependymoma accounts for often have a distinctive vascular pat-
approximately 70% of all childhood su- tern of branching capillaries or clear-cell
pratentorial tumours {1891 l and a lower change. Uncommon variants of ependy-
proportion of such ependymomas in moma (e.g. tanycytic ependymoma) do
adult patients {1880l. Ependymomas in not tend to be RELA fusion-positive.
the posterior fossa and spinal compart-
ments do not harbour this fusion gene. /mmunophenotype
RELA fusion-positive ependymomas RELA fusion-positive ependymomas
exhibit a range of histopathological fea- demonstrate the immunoreactivities
tures, with or without anaplasia. for GFAP and EMA described in other
ependymomas. Expression of L1CAM
ICD-0 code 9396/3 correlates well with the presence of a
RELA fusion in supratentorial ependymo-
Grading mas {1891l, but L1CAM can also be ex- Fig. 3.16 RELA fusion-positive ependymoma.
RELA fusion-positive ependymomas are pressed by other types of brain tumours. lnterphase FISH with break-apart probes around the
classified according to their histopatho- RELA gene. Overlapping probes (yellow) indicate an
logical features into WHO grade II or Genetic profile intact RELA gene, but probe separation (red/green)
grade Ill. No grade I ependymoma has The C11orf95-RELA fusion is the most occurs with rearrangement of the RELA gene.
been recorded as containing this genetic common structural variant found in
alteration. ependymomas {1880,1891,1974l 1t forms
in the context of chromothripsis, a shat- formalin-fixed, paraffin-embedded tis-
Microscopy tering and reassembly of the genome sue is interphase FISH with break-apart
RELA fusion-positive ependymomas do that rearranges genes and produces probes around both genes. Rearrange-
oncogenic gene products {2852l. RELA ment in the context of chromothripsis
fusion-positive ependymomas show splits the dual-colour signals in probe
constitutive activation of the NF-kappaB sets for C11orf95 and RELA {1891l.
pathway, the RELA-encoded transcrip-
tion factor p65 being a key effector in this Prognosis and predictive factors
pathway. Rarely, C11orf95 or RELA can The data available to date (which come
be fused with other genes as a result of from only a single study) suggest that
chromothripsis {1891 l. RELA fusion-positive ependymomas
The presence of a C11orf95-RELA fu- have the worst outcome of the three su-
sion gene can be detected by various pratentorial molecular groups {1880l.
methods, but a simple approach using
Fig. 3.15 RELA fusion-positive ependymoma. L 1 CAM
protein expression correlates well with the presence of a
RELA fusion gene.

112 Ependymal tumours


Anaplastic ependymoma Ellison D.W.
Mclendon R.
Korshunov A.
Ng H.-K.
Wiestler 0.0. WittH.
Kros J.M. Hirose T.

Definition
A circumscribed glioma composed of
uniform small cells with round nuclei in
a fibrillary matrix and characterized by
perivascular anucleate zones (pseudoro-
settes), ependymal rosettes in about one
quarter of cases, a high nuclear-to-cyto-
plasmic ratio, and a high mitotic count.
A diagnosis of anaplastic ependymo-
ma can be confidently made when an
ependymal tumour shows a high cell
Fig. 3.17 Sagittal, gadolinium-enhanced, T1-weighted Fig. 3.18 Anaplastic ependymoma of the lateral ventricle
density and elevated mitotic count along-
MRI of an anaplastic ependymoma of the fourth ventricle. in a 4-year-old boy, with extensive involvement of the
side widespread microvascular prolif- right frontal lobe.
eration and necrosis. Like the classic
tumour, anaplastic ependymoma rarely In a recent study that will likely serve as and biological behaviour or survival has
invades adjacent CNS parenchyma to the basis for the future molecular classifi- been definitively established !248,633,
any significant extent. Cilia and microvilli cation of the disease !1880). nine groups 703}. Of the various studies of prognostic
are seen on ultrastructural examination. of ependymoma were described (three variables and outcome in ependymoma,
Anaplastic ependymomas are mainly for each of the three major CNS ana- those that did not find an association be-
intracranial tumours; they are rare in the tomical compartments: the supratentorial tween grade (II vs Ill) and progression-
spinal cord. Anaplastic ependymomas compartment, posterior Iossa, and spinal free or overall survival outnumber those
occur in both adults and children, al- compartment). The modal patient age at that did. The published ratios of grade II
though most posterior fossa tumours presentation, clinical outcome, and fre- to grade 111 tumours in series of ependy-
present in childhood. Clear cell, papil- quencies of histopathological variants momas vary widely, from 17:1 to 1:7, and
lary, or tanycytic morphology can be a and genetic alterations vary across these the explanation for such inconsistent
feature of both classic and anaplastic groups (see Table 3.01, p. 110). data is multifactorial !633,856,2557)
ependymomas. Anaplastic ependymo- The interpretation of most histopathologi-
mas have a variable clinical outcome, ICD-0 code 9392/3 cal variables used in this classification
which is primarily dependent on extent of for grading purposes is subjective, and
surgical resection and molecular group Grading ependymomas are morphologically het-
!857,1880}. Traditionally, classic ependymoma and erogeneous. There have been few stud-
In defining molecular groups of epen- anaplastic ependymoma are consid- ies of the prognostic significance of WHO
dymoma, transcriptome or methylome ered to correspond histologically to grade or individual pathological features
profiling has established the relevance of WHO grades II and Ill, respectively. in large trial cohorts in which proper mul-
anatomical site to ependymoma biology. However, no association between grade tivariate analyses of prognostic variables

Anaplastic ependymoma 113


can be performed, and there have been
only three studies in which evaluation of
histological variables was defined strin-
gently and undertaken by several ob-
servers {633,856,2557). Due to these
limiting factors, grading is hardly ever
used for therapeutic stratification of pa-
tients with ependymoma. Given that spe-
cific genetic alterations and molecular
groups have recently been proposed as
prognostic or predictive factors for these
tumours (1560,1880,2767}. the practice
of histologically grading ependymoma
may soon become obsolete altogether.

Microscopy
Anaplastic ependymomas almost always
remain circumscribed masses, but can
rarely invade adjacent brain tissue in
the manner of diffuse glioma. Anaplastic
ependymomas generally have a high nu-
clear-to-cytoplasmic ratio. Occasionally,
the cell density is so high that anaplastic
ependymomas can be mistaken for em-
bryonal tumours. All anaplastic ependy-
momas demonstrate brisk mitotic activity,
and high mitotic counts have been asso-
ciated with a poor outcome in posterior
fossa tumours. Microvascular prolifera-
tion and palisading necrosis often ac-
company the mitotic activity, although
these features can also be focal in clas-
sic ependymomas with lower cell density
and few mitotic figures. Pseudorosettes
are a defining feature, but in some poorly
differentiated supratentorial anaplastic
ependymomas, they can be difficult to
find.

lmmunophenotype
Anaplastic ependymoma has the same
immunoprofile as classic ependymoma,
except that indices of tumour growth
fraction, such as the Ki-67 proliferation
index, are higher .

.
•,.

!
Fig. 3.19C Anaplastic ependymoma. High Ki-67 labelling
index.

114 Ependymal tumours


CHAPTER 4

Other gliomas
Chordoid glioma of the third ventricle
Angiocentric glioma
Astroblastoma
Chordoid glioma of the third ventricle Brat D.J.
Fuller G.N.

Definition chordoid gliomas arise in the region of


A slow-growing, non-invasive g/ial tumour the lamina terminalis in the ventral wall of
located in the third ventricle, histological- the third ventricle (1461,1904). In at least
ly characterized by clusters and cords some cases, radiological studies have
of epithelioid tumour cells expressing demonstrated an intraparenchymal hy-
GFAP, within a variably mucinous stroma pothalamic component (2001}
typically containing a lymphoplasmacytic
infiltrate. Clinical features
Chordoid gliomas of the third ventricle Chordoid gliomas occur in adults and to
occur in adults and have a favourable date have arisen only in the third ventricu-
prognosis, particularly in the setting of lar region. Most cases present with signs
gross total resection. and symptoms of obstructive hydroceph-
alus, including headache, nausea, vomit-
ICD-0 code 9444/1 ing, and ataxia (272,576). Other clinical
features include endocrine abnormalities
Grading reflecting hypothalamic compression Fig. 4.02 MRI of a chordoid glioma of the third ventricle
(e.g. hypothyroidism, amenorrhoea, and demonstrating a large, contrast-enhancing, sharply
Chordoid glioma of the third ventri-
delineated mass that fills the anterior third ventricle and
cle corresponds histologically to WHO diabetes insipidus), visual field distur- compresses adjacent structures.
grade II. bances due to compression/displace-
ment of the optic chiasm, and personality a fibrosing pattern with abundant fibrosis.
Epidemiology changes including psychiatric symptoms The fibrosing pattern tends to be more
These tumours are rare, with slightly more and memory abnormalities. common in older patients (196). Other
than 80 cases ever reported. Chordoid (rare) tissue patterns include papillary,
gliomas occur most frequently in adults, Imaging alveolar, and pseudoglandular patterns.
although patient age at presentation var- On neuroimaging, chordoid gliomas Individual tumour cells have abundant
ies widely (5-71 years) Most patients present as well-circumscribed ovoid eosinophilic cytoplasm. In some cases,
present between the ages of 35 and masses within the anterior third ventri- limited glial differentiation in the form of
60 years (mean: 46 years), and there is a cle. On MRI, they are T1-isointense to coarsely fibrillar processes can also be
2:1 female predominance (272,576). Pae- brain and show strong, homogeneous seen (272). Neoplastic nuclei are moder·
diatric examples are rare (379). contrast enhancement (2001). Mass ef- ate in size, ovoid, and relatively uniform.
fect is generally distributed symmetri- Mitoses are absent in most tumours; when
Localization cally and causes vasogenic oedema in present, they are rare (< 1 mitosis per
Chordoid gliomas occupy the anterior compressed adjacent CNS structures, 10 high-power fields) A stromal lympho-
portion of the third ventricle, with larger including the optic tracts, basal ganglia, plasmacytic infiltrate, often containing
tumours also filling the middle and poste- and internal capsules. Most tumours are numerous Russell bodies, is a consistent
rior aspects (2001 }. They generally arise continuous with the hypothalamus and finding. Consistent with their radiographi-
in the midline and displace normal struc- some appear to have an intrinsic anterior cal appearance, the tumours are archi-
tures in all directions as they enlarge. hypothalamic component, suggesting a tecturally solid and show little tendency
Neuroimaging findings, including reports potential site of origin (1461 }. to infiltrate surrounding brain structures.
of small, localized tumours, suggest that Reactive astrocytes, Rosenthal fibres,
Microscopy and often chronic inflammatory cells in·
100
Chordoid gliomas are solid neoplasms, eluding lymphocytes, plasma cells, and
90
80
most often composed of clusters and Russell bodies are seen in adjacent non·
70 cords of epithelioid tumour cells within a neoplastic tissue.
� 60
variably mucinous stroma that typically
I so
contains a lymphoplasmacytic infiltrate. Electron microscopy
1 40

a 30 Three less common histological patterns Parallels have been drawn with the ultra-
20

10 �-
have also been reported: a solid pattern structural morphology of ependymoma
0
., ., with sheets of polygonal epithelioid tu- (1904} and specialized ependymoma of
0 10 20 30 40 so )0

Age at diagnosis mour cells without mucinous stroma, a the subcommissural organ (389). The fea-
Fig. 4.01 Cumulative age distribution (both sexes) of fusiform pattern with groups of spindle- tures of chordoid glioma include interme-
chordoid glioma of the third ventricle, based on 43 cases. shaped cells among loose collagen, and diate filaments, intercellular lumina, apical

116 Other gliomas


Fig. 4.03 Chordoid glioma of the third ventricle. A Histologically, tumours are characterized by cohesive clusters of epithelioid cells with abundant pink cytoplasm and a bubbly,
bluish, mucin-rich stroma. B At higher magnification, nuclei are oval, moderate in size, and bland, and have dispersed chromatin. Mitotic activity and nuclear atypia are absent.
C In almost every instance, tumour cells also form solid arrangements of either nests or linear arrays. D,E Lymphoplasmacytic infiltrate is present in nearly all chordoid gliomas, and
Russell bodies can be identified (E, arrows). F The border between chordoid glioma and adjacent brain is well defined, with little evidence of tumour infiltration, often with chronic
inflammation (arrow) and Rosenthal fibres in the neighbouring brain.

microvilli, hemidesmosomes, and basal and the intensity of immunostaining vary are consistently negative 12089). The
lamina. Some have also been suggested depending on the antibody clone used proliferative potential of chordoid gliomas
to contain secretory granules {389}. 1196). Staining for vimentin and CD34 corresponds to that of other low-grade
is also strong, whereas immunoreactiv- gliomas. The Ki-67 proliferation index is
lmmunophenotype ity for 8100 protein, EMA, and cytokera- low, with values of 0-1.5% in one study
The most distinctive immunohistochemi- tin is variable. Epidermal growth factor 1272) and < 5% in other reports 12089).
cal feature of chordoid gliomas is their receptors and merlin are expressed, R132H-mutant IDH1 immunostaining is
strong, diffuse reactivity for GFAP {272, whereas nuclear accumulation of p53 is negative 1196)
2235). These tumours consistently ex- weak or absent. Neuronal and neuroen- lmmunohistochemistry can be useful in
press TTF1 in most nuclei, although the docrine markers (e.g. synaptophysin, the differential diagnosis of chordoid gli-
percentage of immunoreactive nuclei neurofilaments, and chromogranin-A) oma versus other chordoid neoplasms.

Chordord glioma of the third ventricle 117


demonstrated losses at 11q13 and 9p21
(1035) No EGFR amplifications, chromo-
some 7 gain, or TP53 mutations were
noted. Another study (which used PCR-
based techniques, DNA sequencing,
and comparative genomic hybridization)
detected no consistent chromosomal
imbalances or consistent alterations of
TP53, CDKN2A, EGFR, CDK4, or MDM2
(2089). Neither IDH mutations (i.e, IDHt
or /OH2mutations) nor BRAFV600E mu-
tations were found in a series of 16 chor-
doid gliomas (196).

Prognosis and predictive factors


Chordoid gliomas are slow-growing and
Fig. 4.04 Chordoid glioma of the third ventricle. Tumour cells with diffuse immunoreactivity for GFAP.
histologically low-grade. Gross total re-
section is the treatment of choice and
Chordoid meningiomas usually contain supplied by a report of abnormal cilia can result in long-term recurrence-free
small foci of classic meningioma with in a juxtanuclear location (1904). The survival (576). However, the tumours' lo-
whorl formation and psammoma bodies; presence of a cytological zonation pat- cation within the third ventricle and their
they are also immunopositive for EMA, tern and secretory vesicles indicated a attachment to hypothalamic and supra-
but negative for GFAP and CD34 (2235). specialized ependymal differentiation, as sellar structures often make complete re-
Chordomas strongly express cytokerat- might be expected of cells derived from a section impossible. Postoperative tumour
ins and brachyury, but lack immunoreac- circumventricular organ such as the lam- enlargement has been noted in half of
tivity for GFAP and CD34. ina terminalis. A recent study reported all patients who undergo subtotal resec-
strong expression of TTF1 in both chor- tion. Among the reported cases of chor-
Cell of origin doid gliomas and the organum vasculo- doid gliomas, approximately 20% of the
The ultrastructural demonstration of mi- sum of the lamina terminalis, suggesting patients died in the perioperative period
crovilli and hemidesmosome-like struc- an organum vasculosum origin (196). or from tumour regrowth (1394). Specific
tures in chordoid glioma supports an postoperative complications include dia-
ependymal histogenesis (389). Further Genetic profile betes insipidus, amnesia, and pulmonary
evidence of ependymal or specialized A study using microarray-based com- embolism (576).
ependymal differentiation has been parative genomic hybridization and FISH

118 Other gliomas


Angiocentric glioma Burger PC
Jouvet A.
Preusser M.
Rosenblum M.K.
Ellison D.W.

Definition
An epilepsy-associated, stable or
slow-growing cerebral tumour primarily
affecting children and young adults, his-
tologica!ly characterized by an angiocen-
tric pattern of growth, monomorphous
bipolar cells, and features of ependymal
differentiation.
Angiocentric glioma (also called mono-
morphous angiocentric glioma 12691)
and angiocentric neuroepithelial tumour Fig. 4.05 Angiocentric glioma. A T2-weighted, fluid-suppressed MRI demonstrates the neoplasm as a well-defined
11467)) has an uncertain relationship to hyperintense mass in close proximity to the cingulate gyrus of the right frontal lobe. Note the lesion's primarily cortical
other neoplasms exhibiting ependymal localization. B The bright lesion with little mass effect is based largely in the amygdala.
differentiation. Examples with this pattern
have been reported as cortical ependy- involve children. Males and females are Spread
moma 11465). Tumours harbouring both affected equally frequently. Angiocentric glioma has an infiltrative
angiocentric glioma and ependymoma appearance, locally trapping neurons
patterns have also been reported {26231. Localization and other pre-existing parenchymal ele-
The relationship between angiocentric A superficial, cerebrocortical location is ments. Extensions along parenchymal
glioma and classic ependymoma thus typical. vessels and the subpial zone are com-
remains to be defined. mon 12691}.
Clinical features
ICD-0 code 9431/1 Angiocentric gliomas are epilepto- Macroscopy
genic lesions, with chronic and intrac- Gross features have not been detailed.
Grading table partial epilepsy being particularly One temporal lobe example was de-
Angiocentric glioma corresponds histo- characteristic. scribed at surgery as producing hip-
logically to WHO grade I. pocampal enlargement with darkening
Imaging and induration of the amygdala. The grey
Synonym On MRI, the superficial, if not cortically matter-white matter boundary may be
Angiocentric neuroepithelial tumour (not based, lesion is well circumscribed, bright blurred.
recommended) on FLAIR images, and non-contrast-en-
hancing. A cortical band of T1-hyperinten- Microscopy
Epidemiology sity is present in some cases. A stalk-like A unifying feature is the structure of re-
Incidence figures are not yet available extension to the subjacent lateral ventricle markably monomorphic, bipolar spin-
for this uncommon lesion. Most cases is another variable feature {1337). dled cells oriented around cortical blood

Angiocentric glioma 119


Cell of origin
It has been suggested that these tumours
derive, via a maldevelopmental or neo-
plastic process, from the bipolar radial
glia that span the neuroepithelium during
embryogenesis, and that they may share
similar ependymoglial traits or be capa-
ble of generating ependymocytes {1467).

Genetic profile
Limited numbers of angiocentric gliomas
have been studied for genetic alterations.
However, all 4 cases analysed in two
genomic studies showed copy number
alterations or rearrangements at the MYB
locus on 6q23 {2068,2855). In the re-
arrangements, MYB was fused with OKI
or ESR1. Recently, MYB rearrangements
have been documented in all of 19 stud-
ied angiocentric gliomas, with MYB-OKI
\ fusions found in 6 of 7 cases in which the
Fig. 4.07 Angiocentric glioma. A Compact areas that resemble schwannoma. B Tumour cells in perivascular fusion partner could be identified {118A).
pseudorosettes express GFAP. C Longitudinally oriented GFAP-positive cells. D EMA-positive dot-like structures In another study, an analysis of genomic
corresponding to microlumina.
imbalances by chromosomal compara-
tive genomic hybridization revealed loss
vessels (of all calibres) in single- or mul- complex microvascular proliferation nor of chromosomal bands 6q24 to q25 as
tilayered sleeves that extend lengthwise necrosis is seen. Examples with multiple the only alteration in 1 of 8 cases. In 1
along vascular axes or as radial pseu- mitoses occur, and one such case oc- of 3 cases, a high-resolution screen by
dorosettes of ependymomatous appear- curred as a mitotically active, anaplastic microarray-based comparative genomic
ance. Cells with prominent cytoplasm and astrocytoma-like lesion {2691 }. hybridization identified a copy number
distinct cell borders give an epithelioid gain of two adjacent clones from chro-
appearance to some cases. In such cas- Electron microscopy mosomal band 11p11.2, containing the
es, perivascular formations may be simi- Evidence of ependymal differentiation PTPRJ gene {2035). Angiocentric glio-
lar to pseudorosettes in astroblastoma. includes tumoural microlumina filled with mas lack IDH1, IDH2, and BRAF V600
Tumour cells often aggregate beneath microvilli and delimited by elongated in- mutations {300,1780,2855).
the pia-arachnoid complex in horizon- termediate junctions {2691).
tal streams or in perpendicular, strikingly Genetic susceptibility
palisading arrays, and can diffusely col- /mmunophenotype Angiocentric gliomas have not been re-
onize the neuroparenchyma proper at Spindled and epithelioid tumour cells are ported in association with dysgenetic
variable density. The nuclei are slender, GFAP-reactive, do not label for neuronal syndromes or in familial forms.
with granular chromatin stippling. Some antigens (e.g. synaptophysin, chromogra-
examples have regions of solid growth nin-A, and NeuN), and frequently exhibit Prognosis and predictive factors
containing more conspicuously fibrillary ependymomatous features in their dot- These are typically indolent and radiolog-
elements in compact, miniature schwan- like, microlumen-type cytoplasmic label- ically stable tumours for which excision
noma-like nodules as well as rounded ling for EMA. Surface EMA expression alone is generally curative, but specific
epithelioid cells in nests and sheets inter- may also be seen in epithelioid perivas- prognosis and predictive factors have
rupted by irregular clefts or cavities. The cular and subpial formations. Neither not been determined. One subtotally ex-
epithelioid cells may contain paranuclear, aberrant p53 expression by tumour cells cised example recurred as an anaplastic
round, or oval eosinophilic densities with nor anomalous labelling patterns of in- and ultimately fatal lesion; the case ex-
an internal granular stippling. These cy- cluded neurons for synaptophysin, chro- hibited typical histological features at
toplasmic structures correspond to EMA- mogranin-A, and NeuN have been found. presentation but (unusually) affected an
immunoreactive microlumina (as seen in R132H-mutant IDH1 staining is negative. adult {2691). High-grade gliomas with an-
conventional ependymomas). Included The reported Ki-67 proliferation index in giocentric features have been reported,
neurons, which are interpreted either as primary neurosurgical material ranges but the relationship of these to the clas-
entrapped {2691} or as possibly intrinsic trom s 1% (in most reported cases) to 5%. sic lesion remains unclear {1541,1691).
to the lesion {1467). do not exhibit sig- One anaplastic recurrence exhibited el- Longer follow-up is needed to determine
nificant dysmorphism. Mitoses are inap- evation of the Ki-67 proliferation index to the relationship between level of mitotic
parent or rare in most cases, and neither 10% (up from 1% in the primary) {2691) activity and outcome {1490,1780).

120 Other gliomas


Astroblastoma Aldape K.D.
Rosenblum M.K.
Brat D.J.

Definition terized by the presence of focal or multi- was involved in 144 cases (81%) and the
A rare g!ial neoplasm composed of cells focal regions of high cellularity, anaplas- infratentorial in 33 cases (19%). The spi-
that are positive for GFAP and have tic nuclear features, increased mitotic nal cord is only rarely involved.
broad, non- or slightly tapering process- activity (> 5 mitoses per 10 high-power
es radiating towards central blood ves- fields}, microvascular proliferation, and Imaging
sels (astroblastic pseudorosettes) that necrosis with palisading. In most cases, On CT and MRI, astroblastomas present
often demonstrate sclerosis. these high-grade features are found fo- as well-demarcated, non-calcified, nod-
Astroblastoma mainly affects children, cally in the setting of a classic, better- ular or lobulated masses with frequent
adolescents, and young adults, and oc- differentiated astroblastoma. The Ki-67 cystic change and conspicuous contrast
curs nearly exclusively in the cerebral proliferation index in these malignant as- enhancement {2318j.
hemispheres. Neoplasms exhibiting troblastomas is typically> 10%.
foci of astroblastoma-type perivascular Macroscopy
structuring but having components of Epidemiology Astroblastoma is greyish pink or tan, and
otherwise conventional astrocytoma or These are unusual tumours, and uniform its consistency depends on the extent of
ependymoma should not be given this diagnostic criteria have not been applied; associated collagen deposition. Foci of
designation. therefore, definitive epidemiological data necrosis or haemorrhage do not neces-
are not available. However, astroblasto- sarily indicate anaplasia.
ICD-0 code 9430/3 mas seem to be most frequent in chil-
dren, adolescents, and young adults. The Microscopy
Grading tumours may show a predominance in fe- Intermediate filament-laden cell pro-
The biological behaviour of astroblas- males {2204j. One report on 116 cases cesses that form parallel or radial ar-
toma varies. In the absence of sufficient from the published literature suggested a rays terminating on vascular basement
clinicopathological data, it would be pre- substantial (70%) female predominance membranes, forming astroblastic pseu-
mature to establish WHO grade(s) at this {2447l, which is consistent with prior dorosettes, are commonly observed
time. However, the literature has catego- conclusions {236,2539}; however, an- {1066j. These structures are composed
rized these tumours as either well differ- other study, which compiled 239 cases of elongated tumour cells containing
entiated or malignant (anaplastic). In one from SEER data, found an approximately abundant eosinophilic cytoplasm, with a
series, well-differentiated tumours had equal distribution among males and fe- single, prominent process extending to
low mitotic activity (1 mitosis per 10 high- males {28}. a central blood vessel. Cross-sections
power fields) and an average Ki-67 pro- of pseudorosettes have a radiating ap-
liferation index of 3% {267). Regional Localization pearance, whereas longitudinal sections
(infarct-like} necrosis has been noted in Astroblastomas typically involve the appear ribbon-like. The presence of so-
30% of these tumours. Neither vascular cerebral hemispheres {28,1066,2204, called stout processes, which extend to
proliferation nor spontaneous necrosis 2447j. Among 177 intracranial cases central vessels, is critical to the defini-
with palisading has been seen. in the SEER data for which the site was tion of astroblastoma. These distinctive
Malignant astroblastomas are charac- known, the supratentorial compartment broad or columnar cellular processes are

...
..........,�
Fig. 4.08 Astroblastoma.

Astroblastoma 121
accepted. Bailey and Cushing thought
these tumours arose from embryonic
cells programmed to become astrocytes
{109). The presence of intermediate fila-
ments on ultrastructural examination and
a lack of evidence of neuronal or (in most
cases) ependymal differentiation, togeth-
er with positive staining for GFAP and
S100 protein, suggest that the tumour
may be derived from a cell most simi-
lar to an astrocyte. The tanycyte, a cell
with features intermediate between those
of astrocytes and ependymal cells, has
been suggested as a cell of origin for as-
troblastoma on the basis of ultrastructural
observations (1381,1387).

Genetic profile
DNA copy number aberrations identified
by comparative genomic hybridization
have been described for 7 cases (2671,
and the most common alterations iden-
tified in this small series included gains
of chromosomes 19 and 20q, which fre-
quently occurred together. Less common
were losses on chromosomes 10 and X,
generally positive for GFAP, whereas fi- examining ultrastructural features (1381, and a gain of 9q. Conventional cytoge-
brillarity is generally lacking in the tumour 1387} found cell body polarization with netic studies performed on 2 cases led to
stroma. Vascular hyalinization is a con- investing basement membranes, apical the same overall conclusion {265,11401.
spicuous feature of astroblastoma and cytoplasmic blebs capped by microvilli Although the number of cases studied to
ranges from focal and mild to extensive with purse-string pedicular constrictions, date is small, the findings are consistent
and severe. The combination of mild vas- and lamellar cytoplasmic interdigitations with the hypothesis that astroblastoma
cular hyalinization and radiating tumour (called pleatings). Zonula adherens-type is distinct from conventional glial neo-
cells produces a papillary architecture in junctions framing occasional microro- plasms. A recent case report on a single
a subset of tumours. In mildly hyalinized settes and rare cilia were also identified tumour noted that neither IOH1 nor IOH2
pseudorosettes, perivascular tumour in these cases. mutation was detected in the tumour
cells become separated from the central (745). and a report on a series of 9 cases
vessel by a greater distance and often /mmunophenotype of astroblastoma noted a lack of positive
appear cuboidal. At low magnification, Cytoplasmic immunoreactivity for vimen- immunostaining for R132H-mutant IDH1
more extensive vascular hyalinization tin, S100, and GFAP is characteristic, al- in all cases {95).
gives the impression of numerous pink though the extent of labelling (particularly
hyaline rings, with a paucity of interven- for GFAP) varies considerably (338,1984, Prognosis and predictive factors
ing tumour cells. The interface with adja- 2204). Cell membranes may label for In general, high-grade histology has
cent brain is well delineated and is char- EMA (338,1140}, typically as a focal phe- been found to be associated with recur-
acterized by a pushing or non-infiltrative nomenon. Less-consistent immunolabel- rence, progression, and worse prognosis
border. Focal infiltration of adjacent tis- ling is reported with CAM5.2 (1140,1381, (236,2539). although this association has
sues by tongues of neoplastic cells is 1984} and cytokeratin (1381 }. Reactivity recently been questioned {1135). In one
seen occasionally, but diffuse infiltration for neuron-specific enolase is variable study, only a single recurrence was noted
of surrounding parenchyma is not. The (338,1066,1984). and studied cases have in 14 informative cases treated by gross
reported Ki-67 proliferation index varies been negative for synaptophysin (1140}. total resection, at a mean follow-up of
from 1% to 18% (265,1140). A relationship In isolated examples, neuronal cadher- 24 months (265) An analysis of the litera-
between proliferation index and outcome in (1381} and cell adhesion molecules ture suggested that gross total resection
has not been established in the literature, (including CD44, NCAM1, GJB1, and resulted in a 5-year survival rate of 95%
although an elevated index tends to be GJB2) are positive (1381,1984). Staining (2447). and gross total resection of even
associated with high-grade histology. for OLIG2 can be present (745). high-grade astroblastoma may result in a
Compelling evidence of neuronal differ- favourable outcome (236). Data from the
entiation has not been reported, and no Cell of origin SEER registry indicate that infratentorial
ependymal features have been encoun- The histogenesis of astroblastoma is con- location may portend improved outcome.
tered in most studied cases. Two studies troversial, and the entity is not universally

122 Other gliomas


CHAPTER 5

Choroid plexus tumours


Choroid plexus papilloma
Atypical choroid plexus papilloma
Choroid plexus carcinoma
.,

Choroid plexus papilloma Paulus W.


Brandner S.
Hawkins C.
Tihan T.

Definition present in patients aged < 20 years,


A benign ventricular papillary neoplasm whereas fourth ventricle tumours are
derived from choroid plexus epithelium, evenly distributed across all age groups.
with very low or absent mitotic activity.
Histologically, choroid plexus papilloma Etiology
closely resembles the non-neoplastic Genomic analysis of choroid plexus pa-
choroid plexus, and most tumour cells pilloma suggests a role of genes involved
are positive for the potassium channel in the development and biology of plexus
KIR7.1. Patients are usually cured by epithelium (i.e OTX2 and TRPM3). It is
complete surgical resection. thought that their alteration may contri-
bute to the initial steps of choroid plexus
ICD-0 code 9390/0 oncogenesis {1136). Earlier reports of a
possible role of SV40 {1058} have not
been confirmed in more recent studies. Fig. 5.02 Choroid plexus papilloma. T1-weighted MRI
Grading
demonstrates a large multilobulated tumour within the left
Choroid plexus papilloma corresponds lateral ventricle.
histologically to WHO grade I. Localization
Choroid plexus papillomas are located
Epidemiology within the ventricular system where the been debated whether overproduction of
Although choroid plexus tumours consti- normal choroid plexus can be found. cerebrospinal fluid is a major contributing
tute 0.3-0.8% of all brain tumours overall, They are seen most often in the lateral factor to hydrocephalus {182}
they account for 2-4% of those that occur ventricles, followed by the fourth and
in children aged < 15 years, and for 10- third ventricles. They are rarely found Imaging
20% of those occurring in the first year of within the spinal cord or in ectopic loca- On CT and MRI, choroid plexus papil-
life. In the SEER database, choroid plex- tions {1916). Multifocal occurrence is ex- lomas usually present as isodense or
us tumours account for 0.77% of all brain ceptional {1959). A meta-analysis found hyperdense, T1-isointense, T2-hyperin-
tumours and for 14% of those occurring that the median patient age was 1.5 years tense, irregularly contrast-enhancing,
in the first year of life {351 }. The average for tumours in the lateral and third ven- well-delineated masses within the ven-
annual incidence is 0.3 cases per 1 mil- tricles, 22.5 years for those in the fourth tricles, but irregular tumour margins and
lion population {1196,2117,2777). Choroid ventricle, and 35.5 years for those in the disseminated disease may occur {895).
plexus papillomas (grade I) account for cerebellopontine angle {2777).
58.2% of the choroid plexus tumours in Spread
the SEER database. Congenital tumours Clinical features Even benign choroid plexus papilloma
and fetal tumours have been observed Choroid plexus tumours tend to block may seed cells into the cerebrospinal
in utero using ultrasound techniques. cerebrospinal fluid pathways. Accord- fluid; in rare cases, this can result in drop
The overall male-to-female ratio is 1.2:1; ingly, patients present with signs of hy- metastases in the surroundings of the
for lateral ventricle tumours, the ratio is drocephalus (in infants, increased cir- cauda equina {2440).
1 :1, and for fourth ventricle tumours, 3:2. cumference of the head), papilloedema,
About 80% of lateral ventricular tumours and raised intracranial pressure. It has

Lateral ventricle 41h ventricle


90-100
B0-,19

70-79
60-69

50-59
41>-49

30-39
20-29

• 10-19

�..11111111111�·
150 125 100 75 50 25 0
0-
9
������
0 25 50 75 100 125 150

Number of cases Number of cases


Fig. 5.01 Age versus localization of choroid plexus tumours, Fig. 5.03 A Macroscopic appearance of choroid plexus papilloma showing cauliflower-like appearance. B Choroid
based on a compilation of 264 published cases (2777). plexus papilloma arising in the posterior third ventricle producing partial obstruction with ventricle dilatation.

124 Choroid plexus tumours


Fig. 5.04 Choroid plexus papilloma. A Well-differentiated papillary pattern composed of a single layer of monomorphic tumour cells.
monolayer along vessels.

Macroscopy cobblestone-like surface. Rarely, choroid carcinomas, limiting its usefulness (41).
Choroid plexus papillomas are circum- plexus papillomas can acquire unusual Most choroid plexus tumours demon-
scribed cauliflower-like masses that may histological features, including oncocytic strate variably positive staining for S100,
adhere to the ventricular wall, but are change, mucinous degeneration, melani- possibly related to older patient age and
usually well delineated from brain paren- zation and tubular glandular architecture better prognosis (1916}. Membranous
chyma. Cysts and haemorrhages may of tumour cells, neuropil-like islands, and expression of the inward rectifier potas-
occur. lntraoperative observations in rare degeneration of connective tissue (e.g. sium channel KIR7.1 has been found in
atypical choroid plexus papillomas dem- xanthomatous change; angioma-like in- normal choroid plexus (in 34 of 35 sam-
onstrate a highly vascular tumour with crease of blood vessels; and bone, car- ples) and in choroid plexus papilloma (in
a propensity to bleed {24941, and this tilage, or adipose tissue formation) {87, 12 of 18 cases) but not in 100 cases of
feature is also observed in some typical 299,960). other primary brain tumours and cerebral
choroid plexus papillomas. metastases {958). Another study found
lmmunophenotype KIR7.1 in 30 of 30 choroid plexus tumours
Microscopy Nearly all choroid plexus tumours ex- and the glutamate transporter EAAT1 in
Delicate fibrovascular connective tissue press cytokeratins and vimentin {594, 32 of 35 cases, whereas these markers
fronds are covered by a single layer of 912). Most demonstrate positive staining were absent in 4 endolymphatic sac tu-
uniform cuboidal to columnar epithelial for CK7, and positivity for CK20 is less mours (2284). EAAT1 has also been de-
cells with round or oval, basally situat- common. In one study, none of the tu- scribed to differentiate between neoplas-
ed monomorphic nuclei. Mitotic activity mours were found to be CK20-positive tic and non-neoplastic choroid plexus,
is absent or very low (< 2 mitoses per when CK7-negative (1088). EMA is often because expression in the non-neoplas-
10 high-power fields) Brain invasion with negative or only weakly and focally posi- tic choroid plexus was seen in only 4% of
cell clusters or single cells, high cellu- tive (594,1610); strong positive staining cases (180).
larity, necrosis, nuclear pleomorphism, for EMA favours other neoplasms. Tran-
and focal blurring of the papillary pat- sthyretin is positive in normal choroid Genetic profile
tern are unusual, but can occur. Choroid plexus and in most choroid plexus tu- No large-scale sequencing studies of
plexus papilloma closely resembles non- mours (1916). but staining may be nega- choroid plexus papilloma have been pub-
neoplastic choroid plexus, but the cells tive or variable among choroid plexus lished to date. TP53 mutations are rare in
tend to be more crowded, elongated, or papillomas and choroid plexus carcino- choroid plexus papillomas (present in
stratified in comparison with the normal mas, and is also seen in some metastatic < 10% of cases) {2483}. Both classic cy-
togenetic and genome-wide array-based
approaches demonstrated hyperdiploidy
in choroid plexus papilloma (598,1648,
2123) In a series of 36 choroid plexus tu-
mours, MGMT promoter methylation was
found in all cases (961 ).

Genetic susceptibility
Choroid plexus papilloma is a major dia-
gnostic feature of Aicardi syndrome, a
genetic but sporadic condition presum-
A ably linked to the X chromosome and de-
Fig. 5.05 Choroid plexus papilloma. lmmunohistochemistry for the potassium channel KIR7.1. A Choroid plexus fined by the triad of total or partial agen-
tumour cells in cerebrospinal fluid. B Typical membranous labelling of the apical cell surface of tumour cells. esis of the corpus callosum, chorioretinal

Choroid plexus papilloma 125


lacunae, and infantile spasms {30). In Prognosis and predictive factors initial disease also had local recurrence
the setting of an X;17(q12;p13) transloca- Choroid plexus papilloma can be cured (1365). A meta-analysis of 566 choroid
tion, hypomelanosis of Ito has been as- by surgery alone, with a 5-year survival plexus tumours found that 1-year, 5-year,
sociated with the development of choroid rate as high as 100%. In a series of 41 pa- and 10-year projected survival rates,
plexus papilloma in several cases (2842). tients with choroid plexus papilloma, the respectively, were 90%, 81%, and 77%
Duplication of the short arm of chromo- 5-year rates of local control, control of in choroid plexus papilloma compared
some 9, a rare constitutional abnormality, relapse in the brain at a site distant from with only 71%, 41%, and 35% in choroid
was associated with pathologically con- the initial disease, and overall survival plexus carcinoma {2777). Choroid plexus
firmed hyperplasia of the choroid plexus were 84%, 92%, and 97%, respectively; papilloma in children aged < 36 months
in 1 of 2 cases, and with a choroid plexus the 5-year local control rate was better also had excellent prognosis after sur-
papilloma in another (1803}. after gross total resection than after sub- gery alone {1415). Malignant progression
total resection (100% vs 68%). Tumours of choroid plexus papilloma is rare, but
that relapsed at sites distant from the has been described {452,1147).

Atypical choroid plexus papilloma Paulus W.


Brandner S.
Hawkins C.
Tihan T.

Definition Localization demonstrate a highly vascular tumour


A choroid plexus papilloma that has in- Atypical choroid plexus papillomas arise with a propensity to bleed (2494), but
creased mitotic activity but does not fulfill in locations where normal choroid plexus this feature is also observed in choroid
the criteria for choroid plexus carcinoma. can be found. Whereas typical choroid plexus papillomas.
Atypical choroid plexus papillomas in plexus papillomas occur in the supraten-
children aged > 3 years and in adults torial and infratentorial regions with nearly Microscopy
are more likely to recur than their classic equal frequency, atypical choroid plexus Atypical choroid plexus papilloma is
counterparts. papillomas are more common within the defined as a choroid plexus papilloma
lateral ventricles {1146}. In a large series with increased mitotic activity. One study
ICD-0 code 9390/1 of patients with atypical choroid plexus found that a mitotic count of � 2 mitoses
papillomas, 83% of the tumours were lo- per 10 randomly selected high-power
Grading cated in the lateral ventricles, 13% in the fields (with 1 high-power field corre-
Atypical choroid plexus papilloma corre- third ventricle, and 3% in the fourth ven- sponding to 0.23 mm2) can be used to
sponds histologically to WHO grade II. tricle (2790). establish this diagnosis {1146) The same
study showed that one or two of the fol-
Epidemiology Clinical features lowing four features may also be present:
In the SEER database, choroid plexus Like choroid plexus papilloma, atypical increased cellularity, nuclear pleomor-
tumours account for 0.77% of all brain tu- choroid plexus papilloma blocks cere- phism, blurring of the papillary pattern
mours and for 14% of those occurring in brospinal fluid pathways, and patients (solid growth), and areas of necrosis;
the first year of life, with atypical choroid present with hydrocephalus, papilloede- however, these features are not required
plexus papilloma (grade II) accounting ma, and raised intracranial pressure. for a diagnosis of atypical choroid plexus
for 7.4% of choroid plexus tumours {351}. papilloma.
In the CPT-SIOP-2000 study, patients Imaging
with atypical choroid plexus papilloma No differences in MRI characteristics lmmunophenotype
were younger (with a median patient age have been reported between choroid Atypical choroid plexus papillomas carry
of 0.7 years) than were patients with cho- plexus papilloma and atypical choroid a higher risk of recurrence compared with
roid plexus papilloma or choroid plexus plexus papilloma {2790). typical choroid plexus papilloma, and irn-
carcinoma (both with median patient munohistochemical results also correlate
ages of 2.3 years) (2790}. Spread with recurrence. For example, negative
Atypical choroid plexus papilloma has S100 protein staining in > 50% of the tu-
Etiology been reported to present with metastasis mour cells has been associated with a
No differences have been established at diagnosis in 17% of cases {2790}. more aggressive clinical course {1916).
between the etiology of atypical choroid An inverse correlation has been found
plexus papilloma and choroid plexus Macroscopy between transthyretin staining intensity
papilloma. lntraoperative observations in rare and rate of local recurrence {1146}. In
atypical choroid plexus papillomas one study, positive CD44 staining was

126 Choroid plexus tumours


associated with atypical choroid plexus times as likely as those with < 2 mitoses
papilloma and choroid plexus carcino- per 10 high-power fields to recur after
ma, correlating with the infiltrative nature 5 years of follow-up (1146). The overall
of these tumours {2632). but this associa- and event-free survival rates for atypical
tion has not been validated. Otherwise, choroid plexus papillomas are intermedi-
the immunohistochemical profile of atypi- ate between those for choroid plexus pa-
cal choroid plexus papilloma, including pilloma and choroid plexus carcinoma;
positivity for KIR71.' is similar to that of the 5-year overall survival and event-free
choroid plexus papillorna {2215) survival rates for atypical choroid plexus
papilloma are 89% and 83%, respec-
Genetic profile Fig. 5.06 Atypical choroid plexus papilloma showing tively {2790). There is evidence that the
Atypical choroid plexus papillomas seem focal increase of proliferation (Ki-67 proliferation index). diagnosis of atypical choroid plexus pa-
to be more similar genetically to choroid pilloma is prognostically relevant in chil-
plexus papilloma than to choroid plexus Prognosis and predictive factors dren aged > 3 years and in adults, but
carcinoma {1648). Consistent with the In a series of 124 choroid plexus papil- not in children aged < 3 years, who may
entity's defining histological feature (i.e. lomas, multivariate analysis showed that have good prognosis even with choroid
increased mitotic activity), RNA expres- increased mitotic activity was the only plexus papillomas with high proliferation
sion profiling and gene set enrichment histological feature independently as- (2544).
analysis have revealed higher expression sociated with recurrence; tumours with
of cell cycle-related genes in atypical � 2 mitoses per 10 high-power fields,
choroid plexus papilloma than in choroid which constituted the definition of atypi-
plexus papilloma {1136). cal choroid plexus papilloma, were 4.9

Atypical choroid plexus papilloma 127


Choroid plexus carcinoma Paulus W.
Brandner S.
Hawkins C.
Tihan T

Definition Etiology
A frankly malignant epithelial neoplasm Most choroid plexus carcinomas occur
most commonly occurring in the lateral sporadically, but they can also occur in
ventricles of children, showing at least association with hereditary syndromes
four of the following five histological fea- such as Aicardi syndrome 12488) or
tures: frequent mitoses, increased cellu- (more frequently) Li-Fraumeni syndrome
lar density, nuclear pleomorphism, blur- (LFS) 11379) (seep. 310).
ring of the papillary pattern with poorly
structured sheets of tumour cells, and Localization
necrotic areas. The great majority of choroid plexus car-
Choroid plexus carcinoma frequently in- cinomas are located within and around Fig. 5.08 A large choroid plexus carcinoma in the lateral
vades neighbouring brain structures and the lateral ventricles 11415) ventricle with extensive invasion of brain tissue.
metastasizes via cerebrospinal fluid. Pre-
served nuclear expression of SMARCB1 Clinical features Microscopy
and SMARCA4 (i.e. no inactivation of the Like choroid plexus papilloma, choroid This tumour shows frank signs of ma-
SMARCB1 or SMARCA4 gene) in virtu- plexus carcinoma blocks cerebrospinal lignancy. One study defined tumours
ally all tumours helps in the differential fluid pathways and causes symptoms as frankly malignant if they showed at
diagnosis with atypical teratoid/rhabdoid related to hydrocephalus, such as in- least four of the following five histologi-
tumour. creased intracranial pressure, enlarged cal features: frequent mitoses (usually
head size, nausea, and vomiting 11821. > 5 mitoses per 10 high-power fields),
ICD-0 code 9390/3 increased cellular density, nuclear pleo-
Imaging morphism, blurring of the papillary pat-
Grading On MRI, choroid plexus carcinomas typi- tern with poorly structured sheets of
Choroid plexus carcinoma corresponds cally present as large intraventricular le- tumour cells, and necrotic areas 11146).
histologically to WHO grade Ill. sions with irregular enhancing margins, Diffuse brain invasion is common.
a heterogeneous signal on T2-weighted
Epidemiology and T1-weighted images, oedema in lmmunophenotype
In the SEER database, choroid plexus adjacent brain, hydrocephalus, and dis- Like choroid plexus papillomas, choroid
tumours account for 0.77% of all brain seminated tumour 11660) plexus carcinomas express cytokerat-
tumours and for 14% of those occurring ins, but they are less frequently positive
in the first year of life, with choroid plex- Spread for S100 protein and transthyretin. There
us carcinoma (grade Ill) accounting for Choroid plexus carcinoma presents with is usually no membranous positivity
34.4% of choroid plexus tumours 1351). metastases at diagnosis in 21% of cas- for EMA. Positivity for p53 protein has
About 80% of all choroid plexus carcino- es 12790). A 20-year longitudinal cohort been reported in choroid plexus carci-
mas occur in children. study of 31 patients with choroid plexus nomas that also harboured TP53 muta-
papillomas and 8 patients with choroid tion {24831. Choroid plexus carcinomas
plexus carcinomas showed that the risk have a higher proliferation index than do
of local recurrence or metastasis associ- choroid plexus papillomas and atypical
ated with choroid plexus carcinoma was choroid plexus papillomas. One study re-
20 times the risk associated with choroid ported a mean Ki-67 proliferation index
plexus papilloma 1182). of 1.9% (range: 0.2-6%) for choroid plex-
us papilloma, 13.8% (range: 7.3-60%) for
Macroscopy choroid plexus carcinoma, and < 0.1%
Choroid plexus carcinomas are highly for normal choroid plexus 12607). An-
vascular tumours with a propensity to other study found a mean Ki-67 prolifera-
bleed, but this feature can be seen with tion index of 4.5% (range: O 2-17.4%) for
all types of choroid plexus tumours. choroid plexus papilloma, 18.5% (range:
Choroid plexus carcinomas are invasive 4.1-29.7%) for choroid plexus carcinoma,
tumours that may appear solid, haemor- and 0% for normal choroid plexus {3631.
rhagic, and necrotic. Distinct membranous staining for the po-
Fig. 5.07 MRI of a choroid plexus carcinoma in the lateral tassium channel KIR?.1 is seen in about
ventricle of a 5-year-old child with a TP53 germline mutation. half of all choroid plexus carcinomas.

128 Choroid plexus tumours


Almost all choroid plexus carcinomas re- in virtually all choroid plexus carcinomas. recommended that any patient with a
tain nuclear positivity for SMARCB1 and Both classic cytogenetic and genome- choroid plexus carcinoma be tested for
SMARCA4. wide array-based approaches demon- a TP53 germline mutation, even in the
strate either hyper- or hypodiploidy in absence of a family history of LFS (864)
Genetic profile choroid plexus carcinomas {1648,2123, Choroid plexus carcinoma has also been
Choroid plexus carcinomas are char- 2843). TP53 mutations in choroid plexus described in rhabdoid tumour predispo-
acterized by complex chromosomal al- carcinoma are associated with increased sition syndrome, a familial cancer syn-
terations that are related to patient age genomic instability {2483l, in particular drome caused by germline mutation in the
(2198). No large-scale sequencing stud- hypodiploidy {1648). SMARCB1 gene (2327); however, these
ies of choroid plexus carcinoma have cases most likely constitute intraventricu-
been published to date. About 50% of Genetic susceptibility lar atypical teratoid/rhabdoid tumours
all choroid plexus carcinomas harbour About 40% of choroid plexus carcino- rather than choroid plexus carcinomas.
TP53 mutations. The combination of the mas occur in the setting of germline TP53
TP53-R72 variant and the MOM2SNP309 mutations I LFS {2483). Within the spec- Prognosis and predictive factors
polymorphism, which is associated with trum of tumours occurring in the setting The 3-year and 5-year progression-free
reduced TP53 activity, was observed of LFS, choroid plexus carcinomas are survival rates of choroid plexus carcinoma
in the majority (> 90%) of patients with among the less frequent manifestations. have been reported as 58% and 38%, re-
TP53-wildtype choroid plexus carcino- However, the link to this inherited tumour spectively, and the overall survival rates as
mas (2483l, implicating p53 dysfunction syndrome is so strong that it has been 83% and 62%. Deaths from disease be-
yond 5 years were also noted (2844). Only
extent of surgery had a significant impact
on survival for choroid plexus carcinoma.
The use of adjuvant radiation therapy in
patients with choroid plexus carcinoma
undergoing surgery was not found to be
associated with improved overall survival
(351} Some children with choroid plexus
carcinomas have been found to have
TP53 gene mutations in association with
LFS, and several recent studies have sug-
gested that the absence of TP53 muta-
tions as identified by immunohistochemi-
cal staining is associated with a more
favourable outcome compared with p53-
'• positive tumours {878,2483,2844). One
of these studies also suggested that the
prognosis of TP53-mutant tumours may
be improved with intensive chemotherapy
{2844). A more recent study demonstrat-
-· ed on multivariate analysis that choroid
plexus carcinomas with loss of chromo-
Fig. 5.10 Choroid plexus carcinoma. A Pleomorphism. B High mitotic activity. C Solid growth, high cellular density, some arm 12q were associated with a
and focal membranous staining for KIR7.1. D Infiltration of neighbouring brain tissue. lmmunochemistry for transthyretin significantly shorter survival than were tu-
(ITR). mours without this alteration {2198).

Choroid plexus carcinoma 129


J?
CHAPTER 6

Neuronal and mixed neuronal-glial tumours


Dysembryoplastic neuroepithelial tumour
Gangliocytoma
Ganglioglioma
Anaplastic ganglioglioma
Dysplastic cerebellar gangliocytoma
(Lhermitte-Duclos disease)
Desmoplastic infantile astrocytoma and ganglioglioma
Papillary glioneuronal tumour
Rosette-forming glioneuronal tumour
Diffuse leptomeningeal glioneuronal tumour
Central neurocytoma
Extraventricular neurocytoma
Cerebellar liponeurocytoma
Paraganglioma
Dysembryoplastic neuroepithelial Pietsch T.
Hawkins C.
tumour Varlet P.
BIOmcke I.
Hirose T.

Definition obtained from epilepsy surgery centres,


A benign glioneuronal neoplasm typical- depending on the histopathological dia-
ly located in the temporal lobe of children gnostic criteria used (2541). In a series
or young adults with early-onset epilepsy; of 1511 long-term epilepsy-associated
predominantly with a cortical location and tumours reviewed at the German Neuro-
a multinodular architecture; and with a pathological Reference Center for Epilep-
histological hallmark of a specific glioneu- sy Surgery, ONTs accounted for 17.8% of
ronal element characterized by columns the tumours in adults and 23.4% in chil-
made up of bundles of axons oriented dren (219). In another series, consisting
perpendicularly to the cortical surface. of all neuroepithelial tumours diagnosed
These columns are lined by oligoden- in a single institution since 1975, ONTs
drocyte-like cells embedded in a mu- accounted for 1.2% of the tumours diag-
coid matrix and interspersed with floating nosed in patients aged :.::; 20 years and
neurons. If only the specific glioneuronal 0.2% in patients aged > 20 years (2175).
element is observed, the simple form of
dysembryoplastic neuroepithelial tumour Age and sex distnbution
(ONT) is diagnosed. Complex variants Patient age at the onset of symptoms is
of ONT additionally contain glial tumour an important diagnostic criterion. In about
components, often with a nodular appear- 90% of cases, the first seizure occurs
ance. The presence of IOH mutation or before the age of 20 years (mean patient
1p/19q codeletion excludes the diagnosis age at seizure onset: 15 years), with sei-
of ONT. Long-term follow-up after epilep- zure onset reported in patients aged from
sy surgery shows an excellent outcome; 3 weeks (1854) to 38 years (2078). The
recurrence or progression is exceptional. mean patient age at surgery and histo-
pathological diagnosis is considerably
ICD-0 code 9413/0 older, with a mean age at epilepsy surgery
of 25.8 years. However, earlier detection
Grading of ONTs by MRI in children and young
ONT corresponds histologically to WHO adults with focal epilepsy, and access to
grade I. paediatric and adult epilepsy surgery pro-
grammes are important factors for achiev- Fig. 6.02 Oysembryoplastic neuroepithelial tumour
(ONT). A T2-weighted MRI of a tumour in the right
Epidemiology ing long-term seizure control (219,251,
temporal lobe with a pseudocystic appearance (arrow).
382,689,845,1144,1551,1745,1800) There B On MRI, a right mesiotemporal ONT is T2-hyperintense
Incidence is a slight predominance of ONT in male with a multicystic appearance (arrow).
The reported incidence rates of ONTs vary patients (accounting for 56.7% of cases).
widely (from 7% to 80%) in tumour series brain stem (749). Multifocal ONTs have
Localization also been reported, indicating that these
100�--
ONTs can be located in any part of the tumours can also be found in the region of
901---4-·-+-_,...--� c,,r--, supratentorial cortex, but show a predilec- the third ventricle, the basal ganglia, and
tion for the temporal lobe (348). preferen- the pons (1376,1468,2282,2739).
�1of----t--,r--+--/---r--+---+---+ tially involving the mesial structures (404,
.,,
0 60 1----+--I·
530,531, 532, 534, 537, 566, 1033, 1903, Clinical features
�so f----t-+---++--+---+---+--+---+
� 40 1-----,�-----t---+---4--+-----l- 2026). The second most frequent location Patients with supratentorial ONTs typi-
l 30 1---#-+-l-+- is the frontal lobe (348). In a meta-analysis cally present with drug-resistant focal epi-
u ,o ---+---+-- 4·---t--4 of 624 reported ONTs, 67.3% were located lepsy, with or without secondary seizure
in the temporal lobe, 16.3% in the frontal generalization, and with no neurological
w w � � � 60 ro lobe, and 16.4% in other locations (2543). deficit. However, a congenital neurologi-
Age at diagnosis
-Surgery - Onset
such as the caudate nucleus {394,896) or cal deficit may be present in a minority of
Fig. 6.01 Cumulative age distribution of dysembryoplastic lateral ventricles {1848). the septum pellu- cases {404,534,537,566,689,1800,1903.
neuroepithelial tumours, based on 224 cases from cidum {111,949). the trigonoseptal region 2026,2078,2177,2515). The duration of
the German Neuropathological Reference Center for (896). the midbrain and tectum {1399). the seizures prior to surgical interven-
Epilepsy Surgery, University of Erlangen, Germany. and the cerebellum {534,1382,2819) or tion varies from weeks to decades (with

132 Neuronal and mixed neuronal-glial tumours


vicinity of contrast-enhancing regions and
haemorrhages (348) On CT and MRI,
about 20% to one third of DNTs show con-
trast enhancement, often in multiple rings
rather than homogeneously (348,1800,
2412). Nodular or ring-shaped contrast
enhancement may occur in a previously
non-enhancing tumour (348,24121, and
increased lesion size, with or without peri-
tumoural oedema, may also be observed
Fig. 6.03 A postmortem specimen of a dysembryoplastic
on imaging follow-up. However, in DNTs, Fig. 6.04 Dysembryoplastic neuroepithelial tumour. A
neuroepithelial tumour with multinodular architecture in these changes are not signs of malignant low-power micrograph showing a nodular growth pattern.
the mesial temporal lobe (arrow). transformation but are usually due to is-
chaemic and/or haemorrhagic changes mucoid, alcianophilic matrix {1323). Scat-
a mean duration of 10.8 years), resulting (536,1155,1860). tered, interspersed stellate astrocytes
in variability in patient age at pathological positive for GFAP are associated with this
diagnosis (with a mean age at surgery of Macroscopy glioneuronal element. Constituent oligo-
25.8 years). DNTs vary in size from a few millimetres to dendrocyte-like, neuronal, and astrocytic
several centimetres (1903). In their typical cell populations can be relatively hetero-
Imaging location, they are often easily identifiable geneous from case to case and from area
Cortical topography and the absence of at the cortical surface and may show an to area within the same tumour. DNTs do
mass effect and of significant tumoural exophytic portion. However, leptomenin- not contain dysplastic ganglion cells such
oedema are important criteria for differ- geal dissemination is not a typical feature as those described in gangliogliomas (i.e.
entiating between DNTs and diffuse glio- of DNTs. The appearance of the tumour binucleated neurons, large neurons that
mas. DNTs usually encompass the thick- on cut sections may reflect the complex are either pyramidal or similar to resident
ness of the normal cortex. In a minority histoarchitecture of the lesion. The most cortical neurons, and unequivocal cluster-
of cases, the area of signal abnormality typical feature is the viscous consistency ing not otherwise explicable by anatomi-
also extends into the subcortical white of the glioneuronal component, which cal region). Depending on the extent of
matter (348,530,531,537,689,1395,1860, may be associated with multiple or single fluid extravasation, subtle variation from a
2412) On MRI, most DNTs present as firmer nodules. The affected cortex is of- columnar to an alveolar or a more com-
T2-hyperintense multiple or single pseu- ten expanded. pact structure may be observed (531 ).
docysts. Non-cystic tissue is hypointense Several histological forms of ONT have
or nearly isointense to grey matter on T1- Microscopy been described, but this subclassification
weighted images and hyperintense on The histopathological hallmarks are a has no clinical or therapeutic implications
T2-weighted and FLAIR images (348). In multinodular growth pattern and the so- (2543). Two histological forms of ONT (the
tumours that are located at the convex- called specific glioneuronal element, simple form and the complex form) have
ity, deformation of the overlying calvaria which is characterized by columns ori- been defined, but the criteria for further
is often seen on imaging, and this finding ented perpendicularly to the cortical sur- unspecific and diffuse forms are still un-
further supports the diagnosis of ONT { face, formed by bundles of axons lined by der debate.
530,531,537,1395,2078,2412,2515). Cal- small oligodendrocyte-like cells. Between
cifications are often seen on CT When these columns, neurons with normal cy- Simple form
present, they occur within more deeply tology (which may represent entrapped In this morphological variant, the tu-
located tumour portions, usually in the cortical neurons) appear to float in a mour consists of the unique glioneuronal

. .
Fig. 6.05 Dysembryoplastic neuroepithelial tumour. A Mucin-rich cortical nodule with columnar architecture. B The so-called specific glioneuronal element is characterized by
oligodendrocyte-like cells embedded in a mucoid matrix with interspersed fioating neurons.

Dysembryoplastic neuroepithelial tumour 133


,


!\ .
Fig. 6.06 Dysembryoplastic neuroepithelial tumour. A Glial nodule with piloid features can be seen in a tumour of the complex variant. B This glial nodule in a complex tumour
shows marked endothelial proliferation.

element. It may show a patchy pattern variants of ONT have been described. markers including S100 protein and the
1531), owing to the juxtaposition of foci of These non-specific histological forms ac- glial transcription factor OLIG2. They
tumour and of easily recognizable cortex. counted for 20-50% of the ONTs included can also express myelin-oligodendro-
Other tumour components are absent. in three studies 1537,1903,2610) Because cyte glycoprotein, indicating oligoden-
they lack the specific glioneuronal ele- droglial differentiation {2781), as well
Complex form ment, these variants of ONT are often in- as NOGO-A 11600). GFAP is absent in
In this variant, glial nodules, which give distinguishable from gangliogliomas or oligodendrocyte-like cells, whereas the
the tumour its characteristic multinodular other gliomas by conventional histology, scattered stellate astrocytes within the
architecture, are seen in association with particularly when the cortical topography specific glioneuronal element are positive
the specific glioneuronal element. The of the tumour is not apparent on non-rep- for GFAP. Floating neurons can be visual-
heterogeneous appearance of these tu- resentative samples. lmmunohistochem- ized by the nuclear NeuN epitope {2772,
mours is due to the presence of additional istry and molecular features can help to 2773). Variable expression of MAP2 can
histological components resembling as- exclude typical diffuse astrocytomas and be found in oligodendrocyte-like cells, but
trocytic or oligodendrocytic differentia- oligodendrogliomas of adulthood (see Ge- staining is typically faint, and the strong
tion. These constituent cell populations netic profile). Criteria for the differentiation perinuclear expression found in diffuse
can vary from case to case, as well as of such diffuse and non-specific variants oligodendrogliomas is not detectable
from area to area within the same tumour. of ONT from other long-term epilepsy-as- {224). The oncofetal antigen C034 has
The glial components seen in the complex sociated tumours such as gangliogliomas been described with variable incidences
form of ONTs have a highly variable ap- are not well established, and the concept in ONTs 1222,414,2543). ONT cells should
pearance. They may form typical nodules of additional histological variants of ONT not label with antibodies against mutant
or may show a relatively diffuse pattern. remains controversial. A novel classifica- IOH1 {356) or K27M-mutant H3.3 {150).
They may closely resemble conventional tion scheme for long-term epilepsy-asso- Varying proportions of ONTs have been
categories of gliomas or may show unusu- ciated tumours has been proposed that described to stain with antibodies against
al features. They often mimic pilocytic as- includes these variants as well as mixed V600E-mutant BRAF protein {414,2543).
trocytomas and may show nuclear atypia, tumours 1219). It awaits further molecu- The Ki-67 proliferation index of ONTs has
rare mitoses, or microvascular-like pro- lar-diagnostic and clinicopathological been reported to vary from 0% to 8% fo-
liferation and ischaemic necrosis. Their confirmation. cally 1530,531,537,1903,2026,2515).
microvascular network can vary from
meagre to extensive and may include glo- Cortical dysplasia Differential diagnosis
merulus-like formations. In these vessels, Focal cortical dysplasia is found in ONTs The histological diagnosis of ONT may
the endothelial cells may be hyperplastic {2543). It should be diagnosed only in be difficult, in particular with limited ma-
and mitotically active. Within the glial com- areas of cortical abnormalities without terial. En bloc resection during epilepsy
ponents, frankly hamartomatous (usually tumour cell infiltration and classified as surgery is therefore recommended 1219).
calcified) vessels are common 1531,537, focal cortical dysplasia Type lllb accord- The typical columnar architecture of the
2078). Malformative vessels can cause ing to the classification proposed by the specific glioneuronal element can be
haemorrhage 1536,689,1860,2412,2542). International League Against Epilepsy obscured when the samples are not ad-
(ILAE) 1225). equately oriented, and due to its semiliq-
Non-specific and diffuse forms uid consistency, this element can be lost
On the basis of their similar clinical pre- lmmunophenotype as a result of inadvertent surgical aspira-
sentation, cortical topography, neuro- The so-called specific glioneuronal el- tion and/or fragmentation during fixation.
radiological features, and stability on ement shows a consistent pattern in It is therefore important that the diagno-
long-term preoperative imaging follow- immunohistochemistry. The small oli- sis of ONT be considered in any case
up, non-specific and diffuse histological godendrocyte-like cells express glial in which all of the following features are

134 Neuronal and mixed neuronal-glial tumours


present: history of focal seizures (usually Genetic susceptibility
beginning before the age of 20 years), no DNTs occasionally occur in patients with
progressive neurological deficit, predom- neurofibromatosis type 1 or XYY syn-
inantly cortical topography of a supraten- drome (1218,1376,1468).
torial lesion, no mass effect (except if
related to a cyst), and no peritumoural Prognosis and predictive factors
oedema (530,532,537). DNTs are benign lesions, and their sta-
bility has been demonstrated in a study
oysembryop/astic neuroep,thelial tumour that included 53 patients for whom suc-
versus /ow-grade diffuse glioma cessive preoperative CT or MRI was avail-
oNTs lack mutations in IDH1 or IOH2, Fig. 6.07 Dysembryoplastic neuroepithelial tumour. able, with a mean duration of follow-up of
which are found in a large proportion of Floating neurons show immunoreactivity for synapto- 4.5 years (2412). However, variable histo-
diffuse astrocytomas and oligodendro- physin, whereas small oligodendrocyte-like cells are morphological features and diagnoses of
gliomas of adults (2810). However, in immunonegative. multiple variants of ONT overlapping with
the paediatric setting, such mutations other glioneuronal tumour entities make
are rare in diffuse oligodendrogliomas from DNTs is prognostically important. reliable analysis of prognostic and predic-
(2157) However, examples of composite/mixed tive factors difficult (219). Long-term clini-
Clinical and radiological criteria help ganglioglioma and ONT have been re- cal follow-up usually demonstrates no evi-
in distinguishing these benign tumours ported and were considered to constitute dence of recurrence, even in patients with
from diffuse gliomas. Diffusely infiltrat- a transitional form between the two tu- only partial surgical removal {530,531,
ing low-grade gliomas with a microcystic mours (229,404,1008,2021,2346,2543). 534,537,689, 1287, 1451, 1551,2078,2232,
matrix may mimic a so-called specific Such cases should be diagnosed as 2412). lschaemic or haemorrhagic chang-
glioneuronal element. Residual neurons mixed forms. In a recent proposal for a es may occur, with or without an increase
may appear as floating neurons over- neuropathology-based classification of in size of the lesion or peritumoural oede-
run by glioma cells. Oligodendroglioma long-term epilepsy-associated tumours, ma (536,689,1155,1860,2412). Reported
may exhibit a nodular pattern and can the term "composite neuroepithelial tu- risk factors for the development of recur-
induce secondary architectural changes mours" was proposed for such lesions rent seizures during long-term follow-up
of the cortex, which can make this tu- (219). after operation include a longer preopera-
mour difficult to distinguish from a focal tive history of seizures (94,991). the pres-
cortical dysplasia. lmmunohistochemis- Cell of origin ence of residual tumour (1800). and the
try for GFAP and MAP2 is very helpful, Several factors suggest that DNTs have a presence of cortical dysplasia adjacent to
because the oligodendrocyte-like cells dysontogenetic/malformative origin, such ONT (2224).
of the specific glioneuronal element lack as the young age at onset of symptoms Among the > 1000 DNTs that have been
GFAP (which is typically seen in diffuse and bone deformity adjacent to the tu- reported, malignant transformation is a
astrocytomas) and they also lack the mours (530,531,534,537). However, the rare event. MRI may reveal tumour recur-
strong ring-shaped cytoplasmic staining exact histogenesis of ONT remains un- rence, but histopathology often remains
for MAP2 observed in diffuse oligoden- known (219,2543). benign. Only 20 histologically proven
drogliomas (224). cases have exhibited clear in situ tumour
Genetic profile recurrence, and only 6 showed malignant
Dysembryop/astic neuroeptlhelial tumour Paediatric DNTs have been shown to transformation (413). Ray et al. (2077) re-
versus gang/iog/ioma have stable genomes (1874). whereas ported that the prognosis can be favour-
Gangliogliomas can also pose a chal- recurrent gains of whole chromosomes able after gross resection of the recur-
lenge for the differential diagnosis of 5 and 7 were found in approximately rent ONT. Anaplastic transformation has
DNTs, because (1) the dysplastic gan- 20% and 30% of cases, respectively, in occurred after radiation and/or chemo-
glion cells of gangliogliomas may not be a series of adult DNTs (2016). As in other therapy (2077,2203) Atypical clinical
present in small or non-representative glioneuronal tumours, recurrent BRAF presentation and ambiguous histopatho-
samples, (2) these tumours may show V600E mutations were identified in 30% logical diagnosis must also be taken into
a multinodular architecture, (3) small of DNTs (414,2015). These mutations account, and have interfered with reliable
gangliogliomas may show predominant were associated with activation of mTOR prediction of the biological behaviour of
cortical topography, and (4) the clinical signalling (2015) Neither TP53 muta- ONT from published case series (219,
presentation of gangliogliomas is often tions (751) nor IDH1/IDH2mutations have 2541). Patients with ONT recurrence may
similar to that of DNTs. However, gangli- been detected in DNTs (2810). Similarly, require closer follow-up. These biologi-
oglioma should be suspected when the codeletion of whole chromosome arms cal events further support the neoplastic
tumour shows perivascular lymphocytic 1p/19q has not been demonstrated in (rather than dysplastic or hamartomatous)
infiltration, a network of reticulin fibres, ONT (1874,2016). These markers of dif- nature of ONT.
and/or a large cystic component, or fuse gliomas were also found to be ab-
when the tumour has prominent immuno- sent in 2 cases of ONT of the septum pel-
reactivity for the class II epitope of CD34. lucid um {2800). H3F3A K27M mutations
Because gangliogliomas can undergo were not present in ONT (833).
malignant transformation, their distinction

Dysembryoplastic neuroepithelial tumour 135


Gangliocytoma Capper D.
Becker A.J.
Huse JT.
Rosenblum M.K.
Giannini C. Blumcke I.
Figarella-Branger D. Wiestler OD.

Definition
A rare, well-differentiated, slow-growing
neuroepithelial neoplasm composed of
irregular clusters of mostly mature neo-
plastic ganglion cells, often with dysplas-
tic features.
The stroma consists of non-neoplastic
glial elements. Transitional forms be-
tween gangliocytoma and ganglioglioma
exist, and the distinction of these two en-
tities is not always possible.

ICD-0 code 9492/0

Grading
Gangliocytoma corresponds histologi-
cally to WHO grade I.

Epidemiology
Gangliocytomas are rare tumours pre-
dominantly affecting children. The rela-
tive incidence reported in epilepsy surgi-
cal series ranges from 0% to 3.2% {2541l.

Localization
Like gangliogliomas, these tumours can
occur throughout the CNS. Dysplastic Fig. 6.08 Gangliocytoma. A Focal accumulation of multipolar, dysplastic neurons. B Clusters of mature neurons are
cerebellar gangliocytoma (Lhermitte- embedded in an otherwise normal brain matrix. C Stellate ramification of GFAP-positive astrocytes is compatible with
non-neoplastic nature in a gangliocytoma.
Duclos disease) is discussed in sepa-
rate sections of this volume (see pp. 142,
314). Gangliocytoma of the pituitary is re- may show a lower degree of differentia- Genetic profile
viewed in the volume WHO classification tion, but the presence of mitotically active Genetic data specifically addressing
of tumours of the endocrine organs. neuroblasts is not compatible with the gangliocytomas have not been reported.
diagnosis of gangliocytoma and should A close genetic relationship to gangli-
Clinical features prompt the differential diagnosis of CNS oglioma seems possible.
Gangliocytomas share the clinical fea- ganglioneuroblastoma (see p. 207). The
tures of gangliogliomas. stroma consists of non-neoplastic glial el- Genetic susceptibility
ements but may be difficult to distinguish Dysplastic cerebellar gangliocytoma
Imaging from a low-cellularity glial component of (Lhermitte-Duclos disease), which is as-
Radiological data specifically addressing a ganglioglioma. A network of reticulin sociated with Cowden syndrome, is cov-
gangliocytoma have not been reported. fibres may be present, particularly in a ered in separate sections of this volume
perivascular location. (see pp. 142, 314) No distinct genetic
Microscopy susceptibility factors have been reported
Gangliocytomas are composed of irregu- lmmunophenotype for classic gangliocytoma.
lar groups of large, multipolar neurons The neoplastic ganglion cells are typically
(often with dysplastic features). Binucle- positive (to various degrees) for synapto- Prognosis and predictive factors
ated neurons are commonly observed. physin, neurofilament, chromogranin-A, Gangliocytomas are benign tumours with
The density of dysplastic ganglion cells is and MAP2. The neuronal nuclear antigen favourable outcome. Specific prognos-
typically low; it may be close to the den- NeuN is often only weakly expressed, or tic or predictive factors have not been
sity of neurons in grey matter, but is high- may be negative. reported.
er in some cases. Some ganglionic cells

136 Neuronal and mixed neuronal-glial tumours


Multinodular and vacuolating with globose amphophilic cytoplasm It is not clear whether multinodular and
neuronal tumour of the cerebrum and large nuclei with vesicular chroma- vacuolating neuronal tumour is a neo-
Multinodular and vacuolating neuronal tin and distinctive nucleoli resembling plastic process or a hamartomatous/
tumour of the cerebrum is a recently small neurons. The cells do not display malformative process. Mutation testing
reported clinicopathological lesion with the multinucleation or conspicuous dys- has shown no BRAF V600 hotspot mu-
uncertain class assignment {1068). A to- morphism associated with neoplastic tations (7 cases tested) {228,759,1068)
tal of 14 cases have been reported; they ganglion cells {1068). The cells are ar- or IDH1/IOH2 mutations (3 cases test-
typically occurred in adults, predomi- ranged in nodules involving the deep ed) {228,759). The single point muta-
nantly in the temporal lobe, and were half of the cortex and/or subcortical tion MAP2K1 056P (c.167A->C) has
most frequently associated with seizures white matter, showing prominent intra- been reported in 1 case, and there was
or seizure equivalents {228,759,1068, cellular and stromal vacuolation. The no obvious evidence of copy number
1738). Multinodular and vacuolating cells express neuronal markers, includ- variation by microarray-based compara-
neuronal tumours can also present inci- ing synaptophysin and the neuronal tive genomic hybridization analysis in
dentally, without seizures {228) On T2- protein ELAV3/4, but not the neuronal 2 other cases {1068) The behaviour of
weighted and FLAIR MRI, this tumour is nuclear protein NeuN, chromogranin, or multinodular and vacuolating neuronal
characterized by hyperintensity and ab- the glial marker GFAP. They frequently tumour is benign, and gross total re-
sence of enhancement, often showing a express OLIG2. The lesions are fre- moval or substantial resection provides
subtle or conspicuous internal nodulari- quently associated with the presence disease control.
ty. Histologically, it is composed of small of ramified, CD34-positive neural ele-
to medium-sized neuroepithelial cells ments in neighbouring cerebral cortex.

. - ..__ ..._. .,,. ----r.


Fig. 6.09 Multinodular and vacuolating neuronal tumour of the cerebrum. A On MRI, the tumour presents as a superficial lesion with a high T2 signal (arrowhead) and (B) with a
somewhat nodular appearance on T1-weighted images. C Nodular pattern. Nissl stain. D Small neurons are neurofilament-positive.

Gangliocytoma 137
Ganglioglioma Becker A.J.
Wiestler OD.
Figarella-Branger D.
Blumcke I.
Capper D.

Definition
A well-differentiated, slow-growing glio-
neuronal neoplasm composed of dys-
plastic ganglion cells (i.e. large cells with
dysmorphic neuronal features, without
the architectural arrangement or cytolog-
ical characteristics of cortical neurons) in
combination with neoplastic glial cells.
Gangliogliomas preferentially present in
the temporal lobe of children or young
adults with early-onset focal epilepsy.
lntracortical cystic structures and a cir-
cumscribed area of cortical (and sub-
cortical) signal enhancement are charac-
teristic on FLAIR and T2-weighted MRI.
BRAFV600E mutation occurs in approxi- Fig. 6.11 Ganglioglioma. A Relatively small enhancing nodule and a large, associated septated cyst in a 15-year-
mately 25% of gangliogliomas, whereas old male. The cyst wall is not enhancing. B Coronal T2-weighted imaging shows a cortical/subcortical lesion with
IDH mutation or combined loss of chro- intracortical cysts, bony remodelling, and a hypointense portion suggesting calcification (arrow).
mosomal arms 1p and 19q exclude a di-
agnosis of a ganglioglioma. The progno- The available data indicate that ganglio-
sis is favourable, with a recurrence-free gliomas and gangliocytomas together
survival rate as high as 97% at 7.5 years. account for 0.4% of all CNS tumours and
1.3% of all brain tumours {1207,1552).
ICD-0 code 9505/1 There are no population-based epide-
miological data on gangliogliomas.
Grading Patient age at presentation ranges from
Most gangliogliomas correspond his- 2 months to 70 years. Data from several
tologically to WHO grade I. Some gan- large series with a total of 626 patients
gliogliomas with anaplastic features in indicate a mean/median patient age at
their glial component (i.e. anaplastic diagnosis of 8.5-25 years. The male-
Fig. 6.12 Ganglioglioma of the right temporal lobe also
gangliogliomas) are considered to cor- to-female ratio is 1.1-1.9:1 {1428,2027, involving the hippocampal formation.
respond histologically to WHO grade Ill 2541,2774). In a survey conducted by the
{1552,1570} (seep. 141) Criteria for WHO German Neuropathological Reference duration from 1 month to 50 years before
grade 11 have been discussed but not es- Center for Epilepsy Surgery of 212 chil- diagnosis, with a mean/median duration
tablished {1552,1570). dren with gangliogliomas, the mean pa- of 6-25 years {1428,2027,2774) For tu-
Epidemiology tient age at surgery was 9.9 years and mours involving the brain stem and spinal
48% of the patients were girls. cord, the mean durations of symptoms
100
�i---
90
80
v Localization
/ These tumours can occur throughout the Table 6.01 Localization of gangliogliomas
/v CNS, including in the cerebrum, brain Localization Tumours
/ stem, cerebellum, spinal cord, optic
v nerves, pituitary, and pineal gland. The
Temporal 509 84%
/ Frontal 28 5%
majority (> 70%) of gangliogliomas oc-
/ cur in the temporal lobe {226,1010,1428, Parietal 17 3%
10
/
2027,2774) Occipital 12 2%
I/
10 20 30 40 50 60 Multiple lobes 30 5%
Age at diagnosis Clinical features
The symptoms vary according to tu- Other 6 1%
Fig. 6.10 Cumulative age distribution of ganglioglioma
mour size and site. Tumours in the cer- Based on 602 surgical specimens submitted to the
at diagnosis, based on 602 cases from the German
Neuropathological Reference Center for Epilepsy ebrum are frequently associated with German Neuropathological Reference Center for
a history of focal seizures ranging in Epilepsy Surgery.
Surgery.

138 Neuronal and mixed neuronal-glial tumours


before diagnosis are 1. 25 and 1 A years,
respectively {1428l. Gangliogliomas have
been reported in 15-25% of patients un-
dergoing surgery for control of seizures
(2541l. They are the tumours most com-
monly associated with chronic temporal
lobe epilepsy {219l.

imaging
Classic imaging features include intra-
cortical cyst(s) and a circumscribed
area of cortical (and subcortical) signal
increase on FLAIR and T2-weighted im-
ages 12602). Contrast enhancement var-
ies in intensity from none to marked, and
may be solid, rim, or nodular. Tumour
, . . . .. ..,, -
Fig. 6.13 Ganglioglioma (GG) with sharp demarcation from the adjacent brain parenchyma (NCx) and infiltration into
calcification can be detected in 30% of subarachnoid space (arrow).
tumours. Scalloping of the calvaria may
be seen adjacent to superficially located
cerebral tumours. of ganglioglioma. Some additional his- has been used for the identification of
topathological features frequently iden- lesional ganglion cells in gangliogliomas
Macroscopy tified in gangliogliomas are dystrophic {1318l. lmmunostains for the oncofetal
Gangliogliomas are macroscopically calcification, either within the matrix or as marker CD34 can also be helpful. CD34
well-delineated solid or cystic lesions, neuronal/capillary incrustation; extensive is not present in neurons in adult brain,
usually with little mass effect. Calcifica- lymphoid infiltrates along perivascular but is consistently expressed in 70-80%
tion may be observed. Haemorrhage and spaces or within the tumour/brain paren- of gangliogliomas, especially variants
necrosis are rare. chyma; and a prominent capillary net- emerging from the temporal lobe. CD34-
work. In a few cases, the prominent capil- immunopositive neural cells are promi-
Microscopy lary network manifests as a malformative nent not only in confluent areas of the
The histopathological hallmark of gan- angiomatous component. tumour, but also in peritumoural satellite
gliogliomas is a combination of neuronal Some tumours also display a clear-cell lesions {222l. lmmunoreactivity for GFAP
and glial cell elements, which may exhibit morphology, which raises the differen- characterizes the cells that form the neo-
marked heterogeneity. The spectrum of tial diagnoses of oligodendroglioma and plastic glial component of ganglioglio-
ganglioglioma varies from tumours with dysembryoplastic neuroepithelial tumour. mas. Unlike in diffuse gliomas, MAP2
a predominantly neuronal phenotype to Ganglion cells may also be a component immunoreactivity is faint or absent in the
variants with a dominant glial popula- of extraventricular neurocytic tumours astrocytic component of gangliogliomas
tion. Some cases also contain cells of and papillary glioneuronal tumours. Fo- {224l. Ki-67/MIB1 labelling involves only
intermediate differentiation. Dysplastic cal cortical dysplasia is an uncommon the glial component, with the mean Ki-67
neurons in gangliogliomas may be char- finding in gangliogliomas {225l. It should proliferation index ranging from 1.1% to
acterized by clustering, a lack of cyto- be diagnosed only in areas of cortical ab- 2.7% {1010,1552,2027,2774l.
architectural organization, cytomegaly, normalities without tumour cell infiltration
perimembranous aggregation of Nissl and classified as focal cortical dysplasia Genetic profile
substance, or the presence of binucle- Type lllb according to the classification
ated forms (seen in < 50% of cases). proposed by the International League Cytogenetic alterations
The glial component of gangliogliomas Against Epilepsy (ILAE) {127,225). About 100 gangliogliomas have been
shows substantial variability, but con- studied cytogenetically {188,1029,2408,
stitutes the proliferative cell population lmmunophenotype 2674,2824}. Of these, chromosomal ab-
of the tumour. It may include cell types Antibodies to neuronal proteins such as normalities were found in about one third.
resembling fibrillary astrocytoma, oligo- MAP2, neurofilaments, chromogranin-A, Gain of chromosome 7 is the most com-
dendroglioma, or pilocytic astrocytoma. and synaptophysin demonstrate the neu- mon alteration, although the structural
Eosinophilic granular bodies are encoun- ronal component in gangliogliomas and and numerical abnormalities differ from
tered more often than Rosenthal fibres. A highlight its dysplastic nature. To date, case to case. Associations between cy-
fibrillary matrix is usually prominent and there is no specific marker to differenti- togenetic data and outcome have not
may contain microcystic cavities and/ ate dysplastic neurons from their normal been fully addressed, but the karyotype
or myxoid degeneration. Rarefaction of counterparts; however, chromogranin-A was abnormal in 3 cases with adverse
white matter is another common feature. expression is usually very weak or absent outcome {1141,1142,2674l. In one study,
Gangliogliomas may develop a reticulin in normal neurons, whereas diffuse and chromosomal imbalances were detected
fibre network apart from the vasculature. strong expression suggests a dysplas- in 5 of 5 gangliogliomas by comparative
Occasional mitoses and small foci of ne- tic neuron. For cases with BRAF V600E genomic hybridization {2824l.
crosis are compatible with the diagnosis mutation, a mutation-specific antibody

Ganglioglioma 139
Molecular genetics gangliogliomas; it is also observed in identified fusions of BRAF to KIAA1549,
A BRAF V600E mutation is the most other brain tumours, in particular pleo- FXR1, and MACF1 in 3 gangliogliomas
common genetic alteration in ganglio- morphic xanthoastrocytomas, pilocytic not harbouring a BRAF V600E mutation,
gliomas, occurring in 20-60% of inves- astrocytomas, and dysembryoplas- indicating alternative mechanisms for
tigated cases (414,601,2280). This broad tic neuroepithelial tumours (414,2280}. MAPK pathway activation in some cases
range of reported mutation frequency is V600E-mutant BRAF protein is localized (2855). The presence of a BRAF V600E
probably related to the sensitivity of the mainly to ganglion cells, but can also be mutation was associated with shorter
detection methods used, as well as the found in cells of intermediate differentia- recurrence-free survival in one series of
patient age range in the investigated se- tion, as well as in the glial compartment, paediatric gangliogliomas. The tumours
ries, given that BRAF point mutations are suggesting that ganglion and glial cells in this series were mainly extratemporal
particularly frequent in younger patients in ganglioglioma may derive from a com- and not related to long-term epilepsy
(1318}. BRAF V600E is not specific for mon precursor cell (1318). One study (518). Transcriptomic profiles indicate

"'
:r'i�·
Fig. 6.15 Ganglioglioma. A GFAP expression. B Synaptophysin expression. C Strong expression of V600E-mutant BRAF protein.

140 Neuronal and mixed neuronal-glial tumours


reduced expression of some neurode- reported in a child with neurofibromato- However, any correlation between histo-
velopmental genes, including LDB2 in sis type 2 (2252). Ganglioglioma has also logical anaplasia and clinical outcome is
neuronal components (93,675). Detec- been reported in two patients with Peutz- inconsistent {1207,1428,1552), and most
tion of an IDH1/2 mutation excludes the Jeghers syndrome (553,2101). studies lack molecular analysis to ex-
diagnosis of ganglioglioma or gangliocy- clude high-grade gliomas. In a series of
toma and strongly supports the diagno- Prognosis and predictive factors paediatric gangliogliomas, shorter recur-
sis of diffuse glioma (601,1037) Detec- Gangliogliomas are benign tumours, with rence-free survival was associated with
tion of TP53 or PTEN mutations or COK4 a 7.5-year recurrence-free survival rate of absence of oligodendroglial morphology,
or EGFR amplification does not support a 97% (1552). Good prognosis is associ- higher glial cell density, microvascular
diagnosis of ganglioglioma. ated with tumours in the temporal lobe, proliferation, and a prominent lympho-
complete surgical resection, and chronic plasmacytic inflammatory infiltrate {518),
Genetic susceptibility epilepsy. Anaplastic changes in the glial as well as absence of chronic epilepsy,
one case of ganglioglioma of the optic component and a high Ki-67 prolifera- extratemporal localization, a gemisto-
nerve was reported in a patient with neu- tion index and p53 labelling index may cytic cell component, and intratumoural
rofibromatosis type 1 (1657), and one indicate aggressive behaviour and less expression of CD34 {1570).
case of spinal cord ganglioglioma was favourable prognosis {1010,1207,2027}

Anaplastic ganglioglioma Becker A.J. Blurncke I.


Wiestler O.D. Capper D.
Figarella-Branger D.

Definition proliferation and necrosis (1010,1552, respective low-grade gangliogliomas,


A glioneuronal tumour composed of dys- 2027,2774). Few cases have been ob- as detected by microarray-based com-
plastic ganglion cells and an anaplastic served in which a malignant glioma arose parative genomic hybridization and inter-
glial component with elevated mitotic from the site of a previously resected phase FISH (1029).
activity. ganglioglioma (226). Exclusion of diffuse
glioma with entrapped pre-existing neu- Molecular genetics
ICD-0 code 9505/3 ronal cells is obligatory for the diagnosis In one study, CDKN2A deletion was
of anaplastic ganglioglioma and may be observed in 2 of 3 anaplastic gan-
Grading facilitated by additional genetic analysis gliogliomas {2660). In another study,
Anaplastic gangliogliomas corre- (1037). BRAF V600E mutation was found in
spond histologically to WHO grade Ill 3 of 6 cases (2280).
(1552,1570). Genetic profile
Prognosis and predictive factors
Microscopy Cytogenetics Two studies suggested that patients
In anaplastic gangliogliomas, malignant Analyses of 2 gangliogliomas and their with anaplastic gangliogliomas had sub-
changes almost invariably involve the anaplastic recurrences (WHO grade Ill) stantially reduced 5-year overall and
glial component and include increased have been reported. The losses of progression-free survival rates and in-
cellularity, pleomorphism, and increased CDKN2A/B and OMBT1 or gain/amplifi- creased recurrence rates {1552,1570).
numbers of mitotic figures. Other poten- cation of CDK4 found in the anaplastic Another study did not find histological
tial anaplastic features include vascular tumours were already present in the grade to be predictive of outcome (1428).
However, the low numbers of anaplastic
gangliogliomas included in these stud-
ies may limit the generalizability of these
cliniconeuropathological correlations. In-
triguingly, a paediatric series of ganglio-
gliomas reported favourable 5-year over-
all and event-free survival rates despite
anaplastic neuropathological features
{1227).

Anaplastic ganglioglioma 141


,

Dysplastic cerebellar gangliocytoma Eberhart C.G.


Wiestler 00.
(Lhermitte-Duclos disease) Eng C.

Definition patient age at onset, but most cases have


A rare, benign cerebellar mass composed been identified in adults. A review of the
of dysplastic ganglion cells that conform literature indicates that Lhermitte-Duclos
to the existing cortical architecture. disease can be diagnosed in patients as
In dysplastic cerebellar gangliocytoma young as 3 years old and as old as in the
(Lhermitte-Duclos disease), the enlarged eighth decade of life (641,1525,2864).
ganglion cells are predominantly located PTEN mutations have been identified
within the internal granule layer and thick- in virtually all cases of adult-onset Lher-
en the cerebellar folia. The mass is a major mitte-Duclos disease but not in child-
CNS manifestation of Cowden syndrome hood-onset cases (2864). suggesting that
(seep. 314), an autosomal dominant con- the biologies of the two are different.
dition that causes a variety of hamartomas The single most comprehensive clinical
and neoplasms. epidemiological study estimated the prev-
alence of Cowden syndrome to be 1 case
Historical annotation per 1 million population (2414). However,
Dysplastic cerebellar gangliocytoma was after identification of the gene for Cowden Fig. 6.18 MRI of dysplastic cerebellar gangliocytoma
syndrome (1499). a molecular-based es- (Lhermitte-Duclos disease).
first described in 1920 by Lhermitte and
Duclos (1482) and by Spiegel (2406). The timate of prevalence in the same popula-
disease has also been called "cerebellar tion was 1 case per 200 000 population present. Variable periods of preoperative
granule cell hypertrophy", "diffuse hypertro- (1764). Due to difficulties in recognizing symptoms have been reported, with a
phy of the cerebellar cortex", and "ganglio- this syndrome, prevalence figures are mean length of approximately 40 months
matosis of the cerebellum". Eventually, the likely to be underestimates. In one study (2654). Because cerebellar lesions may
association between Cowden syndrome of 211 patients with Cowden syndrome, develop before the appearance of other
and dysplastic cerebellar gangliocytoma 32% developed Lhermitte-Duclos dis- features of Cowden syndrome, patients
was recognized (641,1873,2654,2656). ease (2125). with Lhermitte-Duclos disease should
be monitored for the development of ad-
ICD-0 code 9493/0 Localization ditional tumours, including breast cancer
Dysplastic cerebellar gangliocytoma in females.
Grading (Lhermitte-Duclos disease) lesions are
It is not yet clear whether dysplastic cer- found in the cerebellar hemispheres. Imaging
ebellar gangliocytoma (Lhermitte-Duclos Neuroradiological studies demonstrate
disease) is neoplastic or hamartomatous. Clinical features distorted architecture in the affected cer-
If neoplastic, it corresponds histologically The most common clinical presentations ebellar hemisphere, with enlarged cer-
to WHO grade I. of Lhermitte-Duclos disease include dys- ebellar folia and cystic changes in some
metria; other cerebellar signs; and signs cases. MRI is particularly sensitive in
Epidemiology and symptoms of mass effect, obstructive depicting the enlarged folia, with alternat-
Due to the rarity of Lhermitte-Duclos hydrocephalus, and increased intracrani- ing T1-hypointense and T2-hyperintense
disease, there has not been a system- al pressure. Cranial nerve deficits, mac- tiger-striped striations (843, 1664,2718).

-
atic study to determine the distribution of rocephaly, and seizures are also often The lesions typically do not enhance
100

90
)
80

70
/
/
/
./
I
/
,_.,.
20

10
I/

Age at diagnosis

Fig. 6.17 Cumulative age distribution of Cowden Fig. 6.19 Dysplastic cerebellar gangliocytoma (Lhermitte-Duclos disease). A Macroscopy with delineated enlargement
syndrome (both sexes) in 75 cases. of cerebellar folia (arrowheads). B Low-power microscopy showing enlargement and distortion of cerebellar cortex.

142 Neuronal and mixed neuronal-glial tumours


-
Fig. 6.20 Dysplastic cerebellar gangliocytoma. A The internal granule layer of the cerebellum at the top of the image is filled with dysplastic ganglion cells. B The dysplastic ganglion
cells are strongly immunopositive for phosphorylated S6. C lmmunohistochemical stains for PTEN show loss of expression in the enlarged neurons, with preserved staining in vessels.

Advanced techniques (including FDG- lmmunophenotype have a germline mutation in PTEN, includ-
PET (972l, MR spectroscopy (679l, and The dysplastic neuronal cells are immu- ing intragenic mutations, promoter muta-
diffusion-weighted imaging (1686)) have nopositive for synaptophysin Antibod- tions, and large deletions/rearrangements
also been used to characterize Lher- ies specific to the Purkinje cell antigens {1499,1589,28651. Pilot data suggest
mitte-Duclos disease. Infiltrating medul- LEU4, PCP2, PCP4, and calbindin have that a subset of individuals with Cowden
loblastomas have been reported to mimic been found to label a minor subpopula- syndrome and Cowden syndrome-like
dysplastic cerebellar gangliocytoma on tion of large atypical ganglion cells, but not symptoms but without PTEN mutations
imaging (602,1686}. to react with the majority of the neuronal instead have germline variants of SOHB
elements, suggesting that only a small (on 1p35-36) or SOHO (on 11q23), both of
Spread proportion of neurons are derived from a which have been shown to affect the same
Dysplastic cerebellar gangliocytoma can Purkinje cell source (926,2351). lmmuno- downstream signalling pathways as PTEN
recur locally, but it does not spread to other histochemistry also demonstrates loss of (AKT and MAPK) {1782).
structures in the brain or outside of the CNS. PTEN protein expression in most dysplas-
tic cells and increased expression of phos- Genetic susceptibility
Macroscopy phorylated AKT and S6, reflecting aberrant Dysplastic cerebellar gangliocytoma
The affected cerebellum displays a dis- signalling that is predicted to result in in- (Lhermitte-Duclos disease) is a compo-
crete region of hypertrophy and a coarse creased cell size and lack of apoptosis (3, nent of Cowden syndrome (also called
gyral pattern that extends into deeper lay- 2864). Undetectable or very low prolifera- PTEN hamartoma syndrome). Cowden
ers. Dysplastic cerebellar gangliocytomas tive activity has been reported in the few syndrome is an autosomal dominant dis-
are usually confined to one hemisphere, cases analysed with proliferation markers order characterized by multiple hamar-
but they can occasionally be multifocal. (3,926). tomas involving tissues derived from all
three germ cell layers. The classic hamar-
Microscopy Cell of origin toma associated with Cowden syndrome
The dysplastic gangliocytoma of Lher- It remains unclear whether Lhermitte- is trichilemmoma. People with Cowden
mitte-Duclos disease causes diffuse en- Duclos disease is hamartomatous or syndrome also have a high risk of breast
largement of the molecular and internal neoplastic in nature. Malformative histo- and thyroid carcinomas, mucocutaneous
granular layers of the cerebellum, which pathological features, very low or absent lesions. non-malignant thyroid abnor-
are filled by ganglionic cells of various size proliferative activity, and the absence malities, fibrocystic disease of the breast,
(3f. An important diagnostic feature is the of progression support classification as gastrointestinal hamartomas, early-on-
relative preservation of the cerebellar ar- hamartoma. However, recurrent growth set uterine leiomyomas, macrocephaly,
chitecture; the folia are enlarged and dis- has occasionally been noted, and dys- and mental retardation. The syndrome is
torted but not obliterated. A layer of abnor- plastic gangliocytomas can develop in caused by germline mutations of the PTEN
mally myelinated axon bundles in parallel adult patients with previously normal MRI gene. A subset of individuals with Cowden
arrays is often observed in the outer mo- scans (3,926,1580}. It has been sug- syndrome and Cowden syndrome-like
lecular layer. Scattered cells morphologi- gested that the primary cell of origin is the symptoms but without germline PTEN mu-
cally consistent with granule neurons are cerebellar granule neuron (926}, and that tations have been found to harbour germ-
also sometimes found under the pia or in a combination of aberrant migration and line variants of SOHB or SOHO {245f.
the molecular layer. The resulting structure hypertrophy of granule cells is responsi-
of these dysmorphic cerebellar folia has ble for formation of the lesions (3f. Murine Prognosis and predictive factors
been referred to as inverted cerebellar cor- transgenic models based on localized Although several recurrent Lhermitte-Du-
tex. Purkinje cells are reduced in number PTEN loss support this hypothesis {1401). clos disease lesions have been reported,
or absent. Calcification and ectatic vessels no clear prognostic or predictive factors
are commonly present within the lesion. Genetic profile have emerged. However, patients should
Vacuoles are sometimes observed in the Approximately 85% of Cowden syndrome be followed for other potential manifesta-
molecular layer and white matter (3f. cases, as strictly defined by the Interna- tions of Cowden syndrome, particularly
tional Cowden Consortium (ICC) criteria, breast cancer in female patients.

Dysplastic cerebellar gangliocytoma (Lhermitte-Duclos disease) 143


Desmoplastic infantile astrocytoma and Brat D.J.
VandenBerg S.R.
ganglioglioma Figarella-Branger D.
Reuss D.E.

Definition
A benign glioneuronal tumour composed
of a prominent desmoplastic stroma with
a neuroepithelial population restricted
either to neoplastic astrocytes - desmo-
plastic infantile astrocytoma (DIA) - or to
astrocytes together with a variable mature
neuronal component - desmoplastic in-
fantile ganglioglioma (DIG) - sometimes
with aggregates of poorly differentiated
cells {1533}. Fig. 6.22 A Desmoplastic infantile astrocytoma. Coronal contrast-enhanced T1 -weighted MRI in a 10-month-old child
DIA and DIG typically occur in infants, with a large head, showing a mixed cystic and solid mass in the left cerebral hemisphere. The solid portion enhances
most often as a large and cystic lesion strongly, abutting and thickening the adjacent meninges. B Desmoplastic infantile ganglioglioma. Postcontrast axial
involving the superficial cerebral cortex MRI demonstrating a superficial enhancing component of a desmoplastic infantile ganglioglioma together with a large
septated cystic component that enlarges the skull and displaces the mid line.
and leptomeninges, often attached to
dura.
also stressed the presence of immature more than one lobe; preferentially the
ICD-0 code 9412/1 neuroepithelial cell aggregates. Because frontal and parietal, followed by the tem-
both lesions have similar clinical, neu- poral, and (least frequently) the occipital
Grading roimaging, and pathological features
DIA and DIG correspond histologically to (including a favourable prognosis), they Clinical features
WHO grade I. are now categorized together as DIA and The signs and symptoms are of short
DIG in the WHO classification. duration and include increasing head
Synonyms and historical annotation circumference, tense and bulging fonta-
DIA was first described in the literature Epidemiology nelles, lethargy, and the setting-sun sign.
by Taratuto et al. {2514). as superficial DIAs and DIGs are rare neoplasms of Occasionally, patients present with sei-
cerebral astrocytoma attached to dura early childhood. Their incidence can only zures, focal motor signs, or skull bossing
with desmoplastic stromal reaction. It be estimated from their frequency in insti- over the tumour.
was included in the first edition of the tutional series. One series of 6500 CNS
WHO classification in 1993 under the tumours from patients of all ages reported Imaging
term "desmoplastic cerebral astrocytoma 22 cases of DIG (0 3%) {2628l In a se- On CT, DIAs and DIGs present as large,
of infancy" {1290}. In 1987, VandenBerg ries of CNS intracranial tumours limited to hypodense cystic masses with a solid
et al. {2629} described desmoplastic children, 6 DIAs were found, accounting isodense or slightly hyperdense super-
supratentorial neuroepithelial tumours for 1.25% of all paediatric brain tumours ficial portion that extends to the overly-
of infancy with divergent differentiation {2514). In studies limited to brain tumours ing meninges and displays contrast en-
(DIG), occurring in the same clinical set- of infancy, DIA and DIG accounted for hancement. The cystic portion is usually
ting. Unlike DIA, DIG was described as 15.8% of the tumours {2875). located deep in the cerebrum, whereas
having a neuronal component with vari- The age range for 84 reported cases of the solid portion is peripheral. On MRI,
able differentiation, and this description DIA/DIG was 1-24 months, with a medi- T1-weighted images show a hypointense
an age of 6 months and a male-to-female cystic component with an isointense pe-
19+-
ratio of 1.5:1 {1533l. The large majority ripheral solid component that enhances
17-18. of infantile cases present within the first with gadolinium. On T2-weighted imag-
15-16 year of life. Several non-infantile cas- es, the cystic component is hyperintense
13-14-
es, with patient ages ranging from 5 to and the solid portion is heterogeneous.
11·12-
25 years, have more recently been re- Oedema is usually absent or modest
7-8- ported. There is a strong male predomi- {2571).
o-5·6, _
------------
nance in the non-infantile cases reported
3.4 ---------- to date {1928l. Macroscopy
10 15 20 25 30
These tumours are large (measuring as
Fig. 6.21 Age distribution (months, both sexes) of Localization much as 13 cm in diameter) and have
desmoplastic infantile astrocytoma and ganglioglioma, DIAs and DIGs invariably arise in the su- deep uniloculated or multiloculated cysts
based on 84 published cases. pratentorial region and commonly involve filled with clear or xanthochromic fluid.

144 Neuronal and mixed neuronal-glial tumours


C Desmoplastic

The solid superficial portion is primarily cell population in DIA. In DIG, neoplastic angiomatoid vessels, but microvascular
extracerebral, involving leptomeninges astrocytes are predominant, but a neo- proliferation is not evident 1539,1538).
and superficial cortex. It is commonly plastic neuronal component with variable
attached to the dura, firm or rubbery in differentiation is also present, most often Electron microscopy
consistency, and grey or white in colour. taking the form of ganglion cells 12628). Astrocytic tumour cells are characterized
There is no gross evidence of haemor- In addition to this desmoplastic lepto- by intermediate filaments that are typi-
rhage or necrosis. meningeal component, both DIA and cally arranged in bundles and by scat-
DIG contain a population of poorly differ- tered cisternae of rough endoplasmic re-
Microscopy entiated neuroepithelial cells with small, ticulum and mitochondria. An extensive
The diagnostic features are those of a round, deeply basophilic nuclei and mini- basal lamina surrounds individual tumour
slow-growing superficial neuroepithelial mal surrounding cytoplasm. This imma- cells. Fibroblasts containing granular en-
tumour composed of a dominant desmo- ture component, lacking desmoplasia, doplasmic reticulum and well-developed
plastic leptomeningeal component and may predominate in some areas. A cor- Golgi complexes are also apparent,
most often containing a variable poorly tical component devoid of desmoplasia particularly in collagen-rich areas 1539,
differentiated neuroepithelial component. may also be observed; this component 1538). The neuronal cells of DIG contain
The desmoplastic leptomeningeal com- is often multinodular, with some nodules dense-core secretory granules and may
ponent consists of a mixture of fibroblast- being microcystic. develop small processes containing neu-
like spindle-shaped cells, often with a There is a sharp demarcation between rofilaments. In neuronal cells, immuno-
wavy pattern with abundant connective the cortical surface and the desmo- electron microscopy has shown filamen-
tissue, intermixed with slightly pleomor- plastic tumour, although Virchow-Robin tous reactivity to neurofilament heavy
phic neoplastic neuroepithelial cells spaces in the underlying cortex are often polypeptide in cell bodies and processes
with eosinophilic cytoplasm arranged in filled with tumour cells. Calcifications are lacking a basal lamina.
fascicles or demonstrating storiform or common, but mononuclear inflammatory
whorled patterns 12514,2629} Reticulin infiltrates are usually lacking. Mitotic ac- lmmunophenotype
impregnations show a prominent reticu- tivity and necrosis are uncommon and In the desmoplastic leptomeningeal
lin-positive network surrounding almost are mostly restricted to the population component, fibroblast-like cells express
every cell and mimicking a mesenchymal of poorly differentiated neuroepithe- vimentin, most express GFAP, and a few
tumour. Astrocytes are the sole tumour lial cells 12628). Some tumours contain express SMA. Most neuroepithelial cells

Desrnoplasnc infantile astrocytoma and gangliogiloma 145


.,

,. {
.. � • I I -
'I
� JI

'\.
(1
• �

Fig. 6.24 A Desmoplastic infantile ganglioglioma. The poorly differentiated component is immunoreactive for the neuronal marker MAP2. B Desmoplastic infantile cerebral
astrocytoma. GFAP-expressing neoplastic astrocytes arranged in streams; the desmoplastic component remains unstained with (C) a marked desmoplastic component demonstrated
by reticulin staining.

also react with GFAP. Astrocytes largely and DIG {99,1538l. The absence of the identified the BRAF V600E mutation in
predominate in this component. Anti- genetic alterations typical of most dif- 2 of 18 DIAs/DIGs {808,1315l. The au-
bodies to collagen IV react in a reticulin- fuse astrocytomas suggests that diffuse thors of both studies concluded that this
like pattern around the tumour cells {99, astrocytomas are not related to these activating mutation was an uncommon
1538l. Expression of neuronal markers neoplasms. finding in DIA/DIG
(e.g. synaptophysin, neurofilament heavy
polypeptide, and class Ill beta-tubulin) Genetic profile Prognosis and predictive factors
is observed in neoplastic neuronal cells; Classic cytogenetic analysis has been Most studies indicate that gross total
mostly in ganglion cells, but also in cells carried out on only a limited number of resection results in long-term survival in
lacking overt neuronal differentiation on cases, in which either a normal karyo- cases of DIA and DIG. In one study of
routine stains {1918l. In the poorly dif- type or non-clonal abnormalities were 8 patients with DIA (median follow-up:
ferentiated neuroepithelial component, described {393l. A comparative genomic 15.1 years), 6 patients survived to the
cells react with GFAP {1776l and vimen- hybridization study of 3 cases of DIA and end of follow-up (the other 2 died peri-
tin, but also with neuronal markers and DIG did not reveal any consistent chromo- operatively) {2514l In another study, no
MAP2 {1918,2628l. Desmin expression somal gains or losses {1369l. One case deaths or evidence of tumour recurrence
may be encountered, but epithelial mark- of DIG showed a loss on 8p22-pter, and were observed among 14 patients with
ers (e.g. cytokeratins and EMA) are not one DIA showed a gain on 13q21. A more DIG (median follow-up: 8.7 years) {2628l
expressed. recent genome-wide DNA copy number Thus, surgery alone, with total removal,
The Ki-67 proliferation index ranges analysis of 4 DIAs and 10 DIGs found seems to provide local tumour control.
from < 0.5% to 5%, with the majority of that large chromosomal alterations were In cases of subtotal resection or biopsy,
reported values being < 2% {1369l. In rare, but demonstrated focal recurrent most tumours are stable or regrow only
the unusual DIAs and DIGs that show losses at 5q13.3, 21q22.11, and 10q21.3 slowly. There have been reports of radio-
histological anaplasia, mitotic activity is {832l. Frequent gains were seen at 7q31, logical regression of some tumours after
readily identified and a Ki-67 proliferation which includes the gene for hepatocyte subtotal resection {2500l. Dissemination
index as high as 45% has been reported growth factor receptor (MET), and less- of these tumours through the cerebrospi-
{547,1963l. frequent gains were noted at 4q12 (KOR, nal fluid has been reported, but should
KIT, and POGFRA) and 12q14.3 (MOM2). be considered a rare event {527,547).
Cell of origin No evidence of KIAA 1549-BRAF fusions Long-term tumour control can be
The exact cellular origins of DIAs and was noted in these cases. Unsupervised achieved by total surgical resection of DIA
DIGs have not been established. The clustering and principal component and DIG despite the presence of prim-
presence of primitive small-cell popula- analysis of the copy number alterations itive-appearing cellular aggregates with
tions that express both glial and neuronal in this group of DIAs and DIGs did not mitotic activity or foci of necrosis. Among
proteins might suggest that these are separate the tumours based on histologi- the DI Gs that show foci of frank anaplasia
progenitor cells to the better-differentiat- cal class, suggesting that they constitute (i.e. with high mitotic rate, microvascular
ed neuroepithelial components and sup- a single molecular genetic entity with a proliferation, and perinecrotic palisading
ports the hypothesis that DIAs and DIGs spectrum of histological features {832l. tumour cells), there have been reports of
are embryonal neoplasms programmed Early molecular studies of DIA did not un- long-term survival after gross total resec-
to progressive maturation. The special- cover any LOH on chromosomes 10 and tion {2451,2500l. Tumour progression
ized subpial astrocytes of the developing 17 or any TP53mutations {1538l. More re- has been reported in DIGs that could
brain have been suggested as a poten- cently, the mutational status of BRAFhas not be completely resected, especially in
tial cell of origin, given that they are su- been investigated in two relatively large those with a high proliferation index and
perficially localized and form a continu- series of DIAs and DIGs. In one study, anaplastic features. Histological features
ous, limiting basal lamina, similar to the BRAFV600E mutations were found in 1 of of glioblastoma have been described in
abundant basal lamina production of DIA 14 tumours. In the other, pyrosequencing the recurrence {547,1523,1963l.

146 Neuronal and mixed neuronal-glial tumours


papillary glioneuronal tumour Nakazato Y.
Figarella-Branger D.
Becker A.J.
Rosenblum M.K.
Komori T.
Park S.-H.

Definition 1998, but was previously described un- emotional affect have also been encoun-
A /ow-grade biphasic neoplasm with as- der other designations, including "pseu- tered. Haemorrhage as the initial pre-
trocytic and neuronal differentiation and dopapillary ganglioglioneurocytoma" sentation has been reported {303,1888l.
histopathological features including a and "pseudopapillary neurocytoma with Some cases are asymptomatic and dis-
pseudopapillary structure composed of glial differentiation" (1274l covered incidentally, even when the tu-
flat to cuboidal astrocytes that are pos- mour is large (as large as 6 cm in one
itive for GFAP lining hyalinized vessels, Epidemiology case) (572,586,1324}.
interpapillary collections of synapto-
physin-positive neurocytes, and occa- Incidence Imaging
sional ganglion cells; with low mitotic PGNTs are rare neoplasms, accounting Radiologically, a cystic component is
activity and infrequent necrosis or micro- for < 0.02% of intracranial tumours (22l. typical, but the tumours can be broadly
vascular proliferation. classified into four groups: cysts with
Papillary glioneuronal tumour (PGNT) is Age and sex distribution mural nodules, masses that are cystic
a rare glioneuronal tumour that preferen- The median patient age at diagnosis is only, mixed cystic and solid masses, and
tially occurs in the supratentorial com- 23 years (range: 4-75 years). A recent re- masses that are solid only (2285). The
partment in young adults, with no sex view of the literature showed that 35% of tumours are usually located in the white
preference. A circumscribed cystic or patients were aged < 18 years and 60% matter, frequently near the ventricle (65,
solid mass with contrast enhancement were aged < 26 years. No sex predilec- 2285} On MRI, the solid portion is isoin-
is characteristic on MRI. A novel translo- tion has been found (2285l. tense or hypointense on T1-weighted
cation, t(9;17)(q31 ;q24), which results in images and diffusion-weighted images,
an SLC44A1-PRKCA fusion oncogene, Localization and hyperintense on T2-weighted images
is present in a high proportion of cases. PGNTs are typically located in the cere- and FLAIR images (65,1439} Most of the
The prognosis is good. bral hemispheres, often in proximity to tumours (85%) have no (or only minimal)
the ventricles, and with a predilection for peritumoural oedema, even when they
ICD-0 code 9509/1 the temporal lobe (283,1324,2022l Oc- are large in volume {2285,2606}. About
casionally, they grow intraventricularly 10% of the reported cases showed overt
Grading (572,2011l. On MRI and CT, the tumours or occult signs of intratumoural haemor-
Most PGNTs correspond histologically present as demarcated, solid to cystic, rhage {169}. Calcification has also been
to WHO grade I and behave in a manner contrast-enhancing masses with little reported in some cases {2505,2757).
consistent with this grade (2285l, but a mass effect. A cystic or mural nodule ar-
minority of cases show atypical histologi- chitecture may be seen. Spread
cal features or late biological progression To date, leptomeningeal or cerebrospinal
(1102, 1139, 1731 l. Clinical features fluid seeding has not been reported.
The principal manifestations include Only one case has shown primary ex-
Synonyms and historical annotation headaches and seizures. Rarely, neu- traneural metastases, which involved
PGNT was established as a clinicopatho- rological deficits, such as disturbances bilateral pleura, pericardium, and the left
logical entity by Komori et al. (1324l in of vision, gait, sensation, cognition, and

Fig. 6.25 Papillary glioneuronal tumour. A T2-weighted MRI shows a solid and cystic mass in the right frontal lobe. B This mass is hypointense on T1-weighted MRI. C Peripheral
enhancement is noted in the solid portion on postcontrast T1-weighted MRI. D "C-methionine PET reveals that this mass has high uptake in the peripheral portion.

Papillary glioneuronal tumour 147


breast and occurred 4.5 years after initial The background can be fibrillary or mi- Electron microscopy
resection {250). crocystic. Neurocytes often resemble The limited ultrastructural studies on
oligodendrocyte-like cells {1324,1484, these tumours have identified three cell
Macroscopy 1731 ). lnterpapillary neuronal cells show types: astrocytic, neuronal, and poorly
PGNTs are usually composed of solid considerable variation in size and shape differentiated { 257, 1324, 1731). Astrocytes
and cystic elements {2285). Almost all {1324,2633). Vascular structures can be contain cytoplasmic bundles of interme-
cases are supratentorial. Most occur prominent and occasionally manifest diate filaments and are separated from
within the cerebral hemisphere (espe- as a mass of hyalinized material without vascular adventitia by a basal lamina.
cially the frontal or temporal lobe), but much intervening tumour {2583). Neurons are characterized by neuronal
some are intraventricular {1731). They are In addition to neuronal cells, minigemi- processes filled with parallel rnicrotu-
usually grey and friable and may be cal- stocytes with eccentrically located nu- bules, their terminations with clear vesi-
cified. Rare cases have presented with clei and eosinophilic cytoplasm are cles, and occasional synapses. Poorly
extensive haemorrhage, mimicking a occasionally observed in interpapillary differentiated cells are polygonal and
cavernoma {169). spaces {1102,2509). At the periphery of contain dense bodies and free ribo-
the lesion, scattered tumour cells are in- somes but no intermediate filaments in
Microscopy termingled with gliotic brain tissue, which the cytoplasm. Minigemistocytes with
contains Rosenthal fibres, eosinophilic numerous bundles of glial filaments have
Histopathology granular bodies, haemosiderin pigment, also been reported {1102).
PGNT is characterized by a prominent and microcalcifications {1324,2583)
pseudopapillary architecture, a single These glial elements lack both nuclear lmmunophenotype
or pseudostratified layer of flattened or atypia and mitotic activity. Microvascular The cytoplasm and processes of flat-
cuboidal glial cells with round nuclei and proliferation or necrosis is exceptional, tened to cuboidal cells covering hyalin-
scant cytoplasm around hyalinized blood even in cases with relatively increased ized vessels are immunostained by anti-
vessels, and intervening collections of proliferative activity {1484), although bodies to GFAP, S100 protein, and nestin
neurocytes and medium-sized gan- there have been a few reports of exam- {1324,1484,2509). In some cases, oligo-
glion cells with accompanying neuropil. ples with frank anaplasia {9,1102). dendrocyte-like cells that are positive for
OLIG2 and negative for GFAP surround

148 Neuronal and mixed neuronal-glial tumours


J
A Mitotic figures are rarely seen. B Ganglioid cells and neurocytes with fibrillary neuropil. C Minigemistocytes are occasionally observed

this layer (2509l. The minigemistocytes differentiation is presumed, with paraven- Prognosis and predictive factors
demonstrate intense immunoreactivity tricular examples possibly deriving from In most cases, gross total resection with-
for GFAP. The neuronal cells and neuropil the subependymal matrix (1324l. out adjuvant therapy results in long-term
stain with antibodies to synaptophysin, recurrence-free survival. Therefore, sur-
neuron-specific enolase, and class 111 Genetic profile I genetic susceptibility gical removal is the main prognostic fac-
beta-tubulin (1324,2509l. The majority of The translocation t(9;17)(q31;q24), result- tor. Anaplastic features (e.g. mitoses, mi-
the neuronal cells are positive for NeuN; ing in an SLC44A 1-PRKCA fusion onco- crovascular proliferation, necrosis, and a
however, NFP expression is mostly con- gene, is present in a high proportion of high proliferation rate) have been report-
fined to large ganglion cells, and chro- cases. The prognosis is good. ed in rare cases and were variably asso-
mogranin-A expression is absent (1324l. To date, no familial or syndrome- ciated with aggressive behaviour (1139,
associated cases of PGNTs have been 1772l. There have been no reported cas-
Pro!tferation reported. An initial series of 6 patients es that progressed or recurred after sur-
The Ki-67 proliferation index generally reported no 1p abnormalities in PGNTs gery when the Ki-67 proliferation index
does not exceed 1-2%, but cases dis- (2509l. Recently, an FGFR1 N546K mu- was « 5.0% {1888l. However,> 20 cases
playing elevated activity (ranging from tation was observed in a single PGNT have been reported to have atypical fea-
10% to> 50%) have been reported (250l. (800l. Another study described recurrent tures, with an elevated Ki-67 proliferation
SLC44A1-PRKCA fusions (278l To date, index (of> 5.0%) Of these tumours, 50%
Cell of origin comparative genomic hybridization has eventually progressed or recurred (9,22,
An origin from multipotent precur- not revealed an aberration profile char- 250,1102,1139,1439,1888,1976l.
sors capable of divergent glioneuronal acteristic of this tumour (670,1731l.

Papillary glioneuronal tumour 149


Rosette-forming glioneuronal tumour Hainfellner J.A.
Giangaspero F.
Rosenblum M.K.
Gessi M.
Preusser M.

Definition
A neoplasm composed of two distinct
histological components: one containing
uniform neurocytes forming rosettes and/
or perivascular pseudorosettes and the
other being astrocytic in nature and re-
sembling pilocytic astrocytoma.
Rosette-forming glioneuronal tumour is
slow-growing, preferentially affects young
adults, and occurs predominantly in the
fourth ventricle, but can also affect other
sites such as the pineal region, optic chi-
asm, spinal cord, and septum pellucidum.

ICD-0 code 9509/1


Fig. 6.29 Rosette-forming glioneuronal tumour. A T1-weighted MRI shows low intensity of the tumour mass and focal
Grading gadolinium enhancement. B T2-weighted MRI demonstrates a relatively hyperintense midline tumour occupying the
Rosette-forming glioneuronal tumour fourth ventricle and involving cerebellar vermis.
corresponds histologically to WHO
grade I, and its generally favourable Imaging and wall or floor of the fourth ventricle. An
postoperative course is consistent with On MRI, rosette-forming glioneuronal intraventricular component is typical, oc-
this grade. tumour presents as a relatively circum- casionally with aqueductal extension.
scribed, solid tumour showing T2-hyper-
Epidemiology intensity, T1-hypointensity, and focal/mul- Microscopy
Rosette-forming glioneuronal tumour is tifocal gadolinium enhancement. Rosette-forming glioneuronal tumours
known to be rare, but specific popula- are generally demarcated, but limited
tion-based incidence rates are not yet Spread infiltration may be seen. They are char-
available. Localization outside of the fourth ventricle acterized by a biphasic neurocytic and
(e.g. in the pineal region, optic chiasm, glial architecture !1119,1325,2033). The
Localization spinal cord, and septum pellucidum) has neurocytic component consists of a uni-
Rosette-forming glioneuronal tumours been described. In rare cases, dissemi- form population of neurocytes forming
arise in the midline, usually occupy the nation throughout the ventricular system neurocytic rosettes and/or perivascu-
fourth ventricle and/or aqueduct, and can occur l58,68,2285,2803l. lar pseudorosettes. Neurocytic rosettes
can extend to involve adjacent brain feature ring-shaped arrays of neurocytic
stem, cerebellar vermis, pineal gland, Macroscopy nuclei around delicate eosinophilic neu-
or thalamus. Secondary hydrocephalus Rosette-forming glioneuronal tumour ropil cores. Perivascular pseudorosettes
may be seen. Localization outside the most commonly involves the cerebellum feature delicate cell processes radiating
fourth ventricle (e.g. in the pineal region,
optic chiasm, spinal cord, and septum
pellucidum) has been described, and
in rare cases, dissemination throughout
the ventricular system can occur !58,68,
2285,2803l.

Clinical features
Patients most commonly present with
headache (a manifestation of obstructive
hydrocephalus) and/or ataxia. Cervical
pain is occasionally experienced. Rare
cases are asymptomatic and discovered
as incidental imaging findings. . ., \.
·� \.
Fig. 6.30 Rosette-forming glioneuronal tumour consists of two components: neurocytic (left) and astrocytic (right).

150 Neuronal and mixed neuronal-glial tumours


A ;I' .r
Fig. 6.31 Rosette-forming glioneuronal tumour. A Neurocytic rosette: a ring-like array of neurocytic tumour cell nuclei around an eosinophilic neuropil core. B Perivascular
pseudorosette with delicate cell processes radiating towards a capillary. C Synaptophysin immunoreactivity in the pericapillary area of a perivascular pseudorosette.

towards vessels. Both patterns, when form surface contacts with perikarya and Genetic profile
viewed longitudinally, may show a colum- other cytoplasmic processes. PIK3CA and FGFR1 mutations (in hotspot
nar arrangement. Neurocytic tumour cells codons Asn546 and Lys656) have been
have spherical nuclei with finely granular lmmunophenotype found in rosette-forming glioneuronal tu-
chromatin and inconspicuous nucleoli, lmmunoreactivity for synaptophysin is mours {340,629,804,2547!. Despite the
scant cytoplasm, and delicate cytoplas- present at the centres of neurocytic ro- histological similarity between rosette-
mic processes. These neurocytic struc- settes and in the neuropil of perivascular forming glioneuronal tumours and pilocyt-
tures may lie in a partly microcystic, pseudorosettes {1119,1325,2033!. Both ic astrocytomas, KIAA1549-BRAFfusions
mucinous matrix. The glial component the cytoplasm and processes of neuro- and BRAF V600E mutations have not
of rosette-forming glioneuronal tumour cytic tumour cells may express MAP2 been described in rosette-forming glio-
typically dominates and in most areas and neuron-specific enolase. In some neuronal tumour {629,803,807,1220}. and
resembles pilocytic astrocytoma. Astro- cases, NeuN positivity can be observed the tumour has not shown evidence of
cytic tumour cells are spindle to stellate in neurocytic tumour cells. GFAP and IDH1/2 mutations (629,2392,2547,2801 l
in shape, with elongated or oval nuclei 8100 immunoreactivity is present in the or 1p/19q codeletion (2694,2801!. Mass
and moderately dense chromatin. Cyto- glial component, but absent in rosettes spectrometry array mutation profiling with
plasmic processes often form a compact and pseudorosettes. The Ki-67 prolifera- a panel covering multiple hotspots for
to loosely textured fibrillary background. tion index is low: < 3% in reported cases. single-nucleotide variants did not reveal
In some areas, the glial component may mutations in AKT1, AKT2, AKT3, EGFR,
be microcystic, containing round to oval, Cell of origin GNAO, GNAS, KRAS, MET. NRAS, or RET
oligodendrocyte-like cells with occasion- Neuroimaging, histological findings, and (629l.
al perinuclear haloes. Rosenthal fibres, molecular evidence indicate that rosette-
eosinophilic granular bodies, microcal- forming glioneuronal tumour may arise Genetic susceptibility
cifications, and haemosiderin deposits from brain tissue surrounding the ven- One reported patient with rosette-form-
may be encountered. Overall, cellularity tricular system. For cases affecting the ing glioneuronal tumour had a type I Chi-
is low. Mitoses and necroses are absent. fourth ventricle, an origin from the sub- ari malformation {1325l. Rosette-forming
Vessels may be thin-walled and dilated or ependymal plate or the internal granule glioneuronal tumours have also been
hyalinized. Thrombosed vessels and glo- cell layer of cerebellum has been sug- described in patients with neurofibroma-
meruloid vasculature may also be seen. gested (1325,25471. tosis type 1 (22631. as well as Noonan
Ganglion cells are occasionally present, syndrome (1220l
but adjacent, perilesional cerebellar cor-
tex does not show dysplastic changes. Prognosis and predictive factors
H1047R The clinical outcome of these generally

!
Electron microscopy benign lesions is favourable in terms of
Astrocytic cells of the glial component survival, but disabling postoperative defi-
contain dense bundles of glial filaments. cits have been reported in approximately
Rosette-forming neurocytic cells are in- half of cases. In rare cases, tumour dis-
timately apposed and feature spherical semination and progression is possible
nuclei with delicate chromatin, cytoplasm {58,2285).
containing free ribosomes, scattered
profiles of rough endoplasmic reticulum,
prominent Golgi complexes, and occa-
sional mitochondria. Loosely arranged cy-
toplasmic processes form the centres of
rosettes and contain aligned microtubules
as well as occasional dense-core gran-
A C A T c A
Fig. 6.32 Rosette-forming glioneuronal tumour. Detection
ules. Presynaptic specializations may be of a missense mutation in exon 20 of PIK3CA by Sanger
seen, and mature synaptic terminals may sequencing.

Rosette-forming glioneuronal tumour 151


Diffuse leptomeningeal Reifenberger G.
Rodriguez F.
glioneuronal tumour Burger PC
Perry A.
Capper D.

Definition have been associated with more aggres-


A rare glioneuronal neoplasm charac- sive clinical behaviour (2149} Due to the
terized by predominant and widespread limited patient numbers and inadequate
leptomeningeal growth, an o/igoden- clinical follow-up reported to date, the
droglial-like cytology, evidence of neu- WHO classification does not yet assign a
ronal differentiation in a subset of cases, distinct WHO grade to this entity.
and a high rate of concurrent KIAA1549-
BRAF gene fusions and either solitary 1p Synonyms
deletion or 1p/19q codeletion in the ab- Disseminated oligodendroglioma-like
sence of /OH mutation. leptomeningeal neoplasm; primary
Diffuse leptomeningeal glioneuronal tu- leptomeningeal oligodendro-gliomatosis
mours preferentially occur in children
but have also been reported in young Historical annotation
adults. The characteristic widespread In 1942, Beck and Russell reported
leptomeningeal tumour growth is read- 4 cases of oligodendrogliomatosis of the
ily detectable as diffuse leptomeningeal cerebrospinal pathway that would corre- Fig. 6.33 Diffuse leptomeningeal glioneuronal tumour.
Numerous superficial hyperintense cystic-like lesions are
enhancement on MRI. An associated spond to diffuse leptomeningeal glioneu-
best appreciated on T2-weighted MRI.
parenchymal, typically intraspinal mass ronal tumours (1511.
is sometimes present. The oligoden-
droglial-like tumour cells show frequent Epidemiology Etiology
immunopositivity for OLIG2 and S100, Diffuse leptomeningeal glioneuronal tu- The etiology of diffuse leptomeningeal
whereas GFAP and synaptophysin ex- mours are rare, and data on incidence glioneuronal tumours is unknown. The
pression is more variable. Most tumours are not available. In the largest published vast majority of tumours develop spon-
are histologically low-grade lesions, but series, of 36 patients (2149). the median taneously, without evidence of genetic
a few demonstrate features of anaplasia. age at diagnosis was 5 years, with the predisposition or exposure to specific
Most tumours show slow clinical progres- patient ages ranging from 5 months to carcinogens. One patient with a Sp dele-
sion, with a more aggressive course oc- 46 years. Only 3 of the 36 patients were tion syndrome has been reported (21491.
casionally encountered. aged > 18 years, supporting the prefer-
ential manifestation in children also found Localization
Grading in other, smaller studies. Males are more These tumours preferentially involve the
The vast majority of diffuse leptomenin- commonly affected than females. Among spinal and intracranial leptomeninges. In
geal glioneuronal tumours present his- the 60 patients reported in seven inde- the intracranial compartment, leptomenin-
tologically as low-grade lesions. How- pendent studies, 38 patients were male geal growth is most commonly seen in the
ever, a subset of tumours show features (a male-to-female ratio of 1.7:1) (446, posterior fossa, around the brain stem, and
of anaplasia with increased mitotic and 786,1258,2030,2109,2149,2294} along the base of the brain. One or more
proliferative activity as well as glomeru- circumscribed, intraparenchymal, cystic
loid microvascular proliferation, which or solid tumour nodules may be seen, with

c
Fig. 6.34 Diffuse leptomeningeal glioneuronal tumour. A Diffuse leptomeningeal thickening (arrows) is a consistent feature on postmortem studies; parenchymal cysts may also be
encountered in some cases, usually in a superficial location. B Note the extensive intraventricular involvement, as well as the parenchymal cysts. C Extensive spinal leptomeningeal
involvement.

152 Neuronal and mixed neuronal-glial tumours


spinal intramedullary lesions being more Imaging demonstrate obstructive hydrocepha-
common than intracerebral masses. MRI shows widespread diffuse lepto- lus with associated periventricular
meningeal enhancement and thickening T2-hyperintensity.
Clinical features along the spinal cord, often extending
Patients often present with acute onset intracranially to the posterior fossa, brain Macroscopy
of signs and symptoms of increased stem, and basal brain. Small cystic or Postmortem investigations have confirmed
intracranial pressure due to obstructive nodular T2-hyperintense lesions along radiological findings and demonstrated
hydrocephalus, including headache, the subpial surface of the spinal cord widespread diffuse leptomeningeal tumour
nausea, and vomiting. Opisthotonos and or brain are frequent. Discrete intra- spread in the spinal and intracranial com-
signs of meningeal or cranial nerve dam- parenchymal lesions, most commonly partments {2149). Multifocal extension of
age may be present. Some patients show in the spinal cord, were found in 25 of tumour tissue along Virchow-Robin spac-
ataxia and signs of spinal cord compres- 31 patients (81 %) in the largest reported es and areas of limited brain invasion are
sion. Rare patients present with seizures. cohort {2149). Patients also commonly common. Tumour growth along peripheral

�· ...,.,;;.:
- .
. ...� . '
; ·"
C Diffuse

Diffuse leptomeningeal glioneuronal tumour 153


,

nerve roots and infiltration of basal cranial nests in the leptomeninges, with desmo- observed. Rosenthal fibres are usually
nerves and ganglia may also be seen. plastic and myxoid changes commonly absent. The intraparenchymal compo-
present. A storiform pattern may be ob- nent resembles a diffusely infiltrative
Microscopy served in desmoplastic areas. Mitotic glioma, mostly oligodendroglioma-like,
Cerebrospinal fluid examination demon- activity is usually low, and other histo- although astrocytic features occasionally
strates elevated protein levels, although logical features of anaplasia are absent predominate.
cytology is often negative {2149,2294!. in most cases. However, rare examples
Therefore, the diagnosis usually requires show histological evidence of anaplasia lmmunophenotype
a meningeal tumour biopsy. Histological- either at primary presentation or after The oligodendroglial-like tumour cells
ly, diffuse leptomeningeal glioneuronal tumour progression {2149l. Anaplastic typically express OLIG2, MAP2, and
tumours are low- to moderate-cellularity features include brisk mitotic activity (de- S100 protein {2030,2149!. GFAP immuno-
neoplasms composed of relatively mono- fined as � 4 mitoses per 10 high-power positivity in tumour cells is seen in < 50%
morphic oligodendroglial-like tumour fields) {2149l. Areas of necrosis are usu- of the cases and is often restricted to a
cells with uniform, medium-sized round ally absent on biopsies but have been minor proportion of neoplastic cells. Ex-
nuclei and inconspicuous nucleoli. Like detected in individual cases at autopsy pression of synaptophysin is detectable
in oligodendroglioma, artificial perinu- {2149l. A small subset of tumours contain in as many as two thirds of the tumours,
clear haloes and cytoplasmic swelling ganglion or ganglioid cells. Neuropil as- and is particularly common in those con-
are commonly seen in formalin-fixed, sociated with ganglion cells and neuro- taining neuropil aggregates and ganglion
paraffin-embedded tissue sections. The pil-like islands have also been observed. cells. lmmunostaining for NeuN, neurofil-
tumour cells grow diffusely or in small Rarely, eosinophilic granular bodies are aments, EMA, and R132H-mutant IDH1 is

1.2

0.8

0.4

0.0

-0.4

-0.8

-1.2

Fig. 6.38 Diffuse leptomeningeal glioneuronal tumour. DNA copy number profile determined by 450k methylation bead array analysis; note the deletion of 1 p and the circumscribed
gains on 5p and 7q in a case with KIAA 1549-BRAFfusion.

154 Neuronal and mixed neuronal-glial tumours


negative The Ki-67 proliferation index is Cell of origin 9 cases (2156). and immunohistochemis-
usually low, with a median value of 1.5% The cellular origin of diffuse leptomenin- try for R132H-mutant IDH1 was negative
reported in one series (2149). However, a geal glioneuronal tumours is unknown. in 14 cases (2030,2149) DNA sequenc-
subset of cases have an elevated Ki-67 The absence of obvious parenchymal ing revealed neither IDH1 nor /OH2muta-
proliferation index (2030,2149,2294), lesions in some patient suggests an ori- tions in 6 cases investigated (D. Capper,
_ with one study reporting less favourable gin from displaced neuroepithelial cells University of Heidelberg, personal com-
outcome associated with a proliferation within the meninges. However, an in- munication, September 2015).
" index of> 4% {2149). traparenchymal origin is also possible,
given that small intraparenchymal foci Genetic susceptibility
Differential diagnosis are frequently present in addition to the In a series of 36 cases, no evidence of
The differential diagnosis includes lep- diffuse leptomeningeal tumour spread. recognized genetic predisposing fea-
tomeningeal dissemination of primar- tures or other tumour syndromes was
ily intraparenchymal diffuse astrocytic or Genetic profile observed, although one patient had a
oligodendroglial gliomas. Pilocytic astro- The most frequent genetic alteration constitutional 5p deletion, one a coexist-
cytoma may also show leptomeningeal in diffuse leptomeningeal glioneuronal ing type I Chiari malformation, and one a
dissemination. The absence of morpho- tumours reported to date is KIAA 1549- factor V Leiden mutation (2149).
logical features of pilocytic differentiation, BRAF fusion, with 12 of 16 investigated
the lack of (or only focal) GFAP immuno- cases (75%) showing this alteration Prognosis and predictive factors
reactivity, the presence of synaptophy- (2156). Deletions of chromosomal arm Diffuse leptomeningeal glioneuronal tu-
sin-positive cells, and the characteristic 1p are also frequently observed in FISH mours may go through periods of stabil-
molecular profile of KIAA1549-BRAF analysis and were reported in 10 of ity or slow progression over many years,
fusion combined with isolated 1p dele- 17 tumours (59%) in one series (19, although often with considerable mor-
tion or 1p/19q codeletion (2156) suggest 253,786,2149,2156). In single cases with bidity (2149). In a retrospective series
that diffuse leptomeningeal glioneuronal SNP array data, complete 1p arm loss of 24 cases, with a median available fol-
tumour is a distinct entity. Pleomorphic was demonstrated (2149,2156). Codele- low-up of 5 years, 9 patients (38%) died
xanthoastrocytomas are readily distin- tions of 1p and 19q have been observed between 3 months and 21 years after
guished by their pleomorphic histology. at a lower frequency, present in 3 of 17 tu- diagnosis (median: 3 years) (2149). and 8
Diffuse leptomeningeal glioneuronal tu- mours (18%) in one series (2156). In one of the 24 patients lived for > 10 years after
mours also lack BRAF V600E mutations case with 1p/19q codeletion, a t(1p;19q) diagnosis (2149). Mitotic activity of any
(2156). a molecular marker found in most (q10;p10) translocation was demonstrat- degree (P = 0.045), a Ki-67 proliferation
pleomorphic xanthoastrocytomas and a ed by FISH (2185). BRAF V600E point index ot z 4% (P = 0.01). and glomeruloid
subset of gangliogliomas (2280). mutations were not observed in a series of microvascular proliferation (P = 0.009) at
the initial biopsy were each associated
with decreased overall survival (2149).

Diffuse leptomeningeal glioneuronal tumour 155


.,

Central neurocytoma Figarella-Branger D.


Soylernezoqlu F.
Burger P.C.
Park S.-H.
Honavar M.

Definition
An uncommon intraventricular neoplasm
composed of uniform round cells with a
neuronal immunophenotype and low pro-
liferation index.
Central neurocytoma is usually located in
the region of the foramen of Monro, pre-
dominantly affects young adults, and has
a favourable prognosis.

ICD-0 code 9506/1

Grading
Central neurocytoma corresponds histo-
logically to WHO grade 11. The tumours
are usually benign, but some recur, even Fig. 6.40 Central neurocytoma. A Large intraventricular tumour of the lateral ventricle with hypointensity on T1-
after total surgical removal. Because the weighted MRI. B Strong contrast enhancement on T1-weighted MRI after gadolinium injection.
prognostic values of anaplastic features
and the Ki-67 proliferation index are still The diagnosis of central neurocytoma equally affected, with a male-to-female
uncertain, it is not yet possible to attribute should be restricted to neoplasms locat- ratio of 1.02:1. Population-based inci-
two distinct histological grades (I and 11) ed within the intracerebral ventricles. Tu- dence rates for central neurocytoma are
to central neurocytoma variants. mours mimicking central neurocytomas not available. In large surgical series,
but occurring within the cerebral hemi- central neurocytomas account for 0.25-
Synonyms and historical annotation spheres (so called cerebral neurocyto- 0.5% of all intracranial tumours {969}.
The term "central neurocytoma" was mas) {1792} or the spinal cord {476,25191
coined by Hassoun et al. {967) to de- were later documented. Neoplasms that Localization
scribe a neuronal tumour with pathologi- arise within the CNS parenchyma and Central neurocytomas are typically lo-
cal features distinct from cerebral neu- share histological features with the more cated supratentorially in the lateral
roblastomas, occurring in young adults, common central neurocytomas but ex- ventricle(s) and/or the third ventricle. The
located in the third ventricle, and histo- hibit a wider morphological spectrum most common site is the anterior portion
logically mimicking oligodendroglioma. are now called extraventricular neurocy- of one of the lateral ventricles, followed
These tumours had been previously re- tomas {8161 (seep. 159). Some tumours by combined extension into the lateral
ported as ependymomas of the foramen have neurocytic tumour cells but are not and third ventricles, and by a bilateral in-
of Monro or intraventricular oligoden- classified as central or extraventricular traventricular location. Attachment to the
drogliomas. Central neurocytomas were neurocytomas; for example, neurocytic septum pellucidum seems to be a feature
then reported in other intraventricular differentiation has been reported in an of the tumour {1462,2047,2690). Isolated
locations, mainly in the lateral and third increasing number of tumours with dis- third and fourth ventricular occurrence is
rare {491,2638).

-
ventricles, but also the fourth ventricle. tinctive morphological features, some of
these tumours emerging as new entities,
100
such as cerebellar liponeurocytoma and Clinical features
90
80
.,...-- papillary glioneuronal tumour {13241 and Most patients present with symptoms of
70
/ variants {387). increased intracranial pressure, rather
/
! 60 than with a distinct neurological deficit.
I
·� so
I Epidemiology The clinical history is short (median: 1.7-
] 40
I In an analysis of> 1000 cases, the mean 3 months) {1462).
u 30
20
I patient age at clinical manifestation was
./ 28.5 years; 46% of patients were diag- Imaging
10

0
v nosed in the third decade of life and 70% On CT, the masses are usually isodense
0 10 20 30 40 50 60 70
Age at diagnosis
between the ages of 20 and 40 years. or slightly hyperdense. Calcifications
Fig. 6.39 Cumulative age distribution (both sexes) Patient ages reported in the literature and cystic changes may be seen {597).
of central neurocytoma (excluding extraventricular range from 8 days to 82 years, although On MRI, central neurocytomas are T1-
neurocytoma), based on 201 cases. paediatric cases are rare. Both sexes are isointense to brain and have a so-called

156 Neuronal and mixed neuronal-gllal tumours


may be observed, even in the same spec- prominent Golgi complex, and some cis-
imen. These include an oligodendroglio- ternae of rough endoplasmic reticulum,
ma-like honeycomb architecture, large often arranged in concentric lamellae.
fibrillary areas mimicking the irregular ro- Numerous thin and intermingled cell pro-
settes in pineocytomas, cells arranged in cesses containing microtubules, dense-
straight lines, and perivascular pseudo- core vesicles, and clear vesicles are al-
rosettes as observed in ependymomas. ways observed !388,969) Well-formed
The cells are isomorphous, with a round or abnormal synapses may be present,
or oval nucleus with finely speckled chro- but are not required for the diagnosis.
matin and variably present nucleoli. Cap-
Fig. 6.41 A large central neurocytoma filling both lateral illary blood vessels, usually arranged in lmmunophenotype
ventricles and extending into the third ventricle and the an arborizing pattern, give the tumours a Synaptophysin is the most suitable and
temporal horn of the left ventricle {1291 }. neuroendocrine appearance. Calcifica- reliable diagnostic marker, with immuno-
tions are seen in half of all cases, usually reactivity diffusely present in the tumour
soap-bubble multicystic appearance on distributed throughout the tumour. Rarer matrix, especially in fibrillary zones and
T2-weighted images. They often exhibit findings include Homer Wright rosettes perivascular nucleus-free cuffs !705,969).
FLAIR hyperintensity, with a well-defined and ganglioid cells !2134,26611. Mitoses Most cases are also immunoreactive for
margin. In all cases, heterogeneous en- are exceptional, and necrosis or haemor- NeuN, although the intensity and extent of
hancement after gadolinium injection is rhage is rare. the labelling vary !2399,2638) Other neu-
observed, and the tumour may show vas- In rare instances, anaplastic histological ronal epitopes (e.g. class Ill beta-tubulin
cular flow voids. Haemorrhage may be features (i.e. brisk mitotic activity, micro- and MAP2) are also usually expressed,
seen !597,1788,26901. An inverted alanine vascular proliferation, and necrosis) can whereas expression of chromogranin-A,
peak and a notable glycine peak on pro- occur in combination, and tumours with NFP, and alpha-internexin is lacking, ex-
ton MR spectroscopy are useful in the dif- these features are called atypical central cept in very rare cases showing ganglio-
ferential diagnosis of intraventricular neo- neurocytomas !969,2661,2818}. This term cytic differentiation. Although most stud-
plasms !597,15711. is also used for central neurocytomas with ies have found GFAP to be expressed
a Ki-67 proliferation index of :::: 2% or 3% only in entrapped reactive astrocytes,
Spread !2053,2478). even when there are no as- this antigen has been reported in tumour
Craniospinal dissemination is exception- sociated anaplastic histological features. cells by some authors !2478,2582,2661,
al l643,2567f. 2662). OLIG2 has also been reported in
Electron microscopy tumour cells in some cases !1500). This
Macroscopy Although it was electron microscopy that immunoprofile helps to distinguish cen-
Central neurocytoma is greyish and fri- allowed Hassoun et al. !9691 to discover tral neurocytoma from ependymoma and
able. Calcifications and haemorrhages the neuronal differentiation of cells form- pineocytoma; ependymoma is synapto-
can occur. ing central neurocytomas, electron mi- physin-negative and more diffusely posi-
croscopy is no longer required for the tive for GFAP than is central neurocytoma,
Microscopy diagnosis of central neurocytoma, due to and pineocytoma expresses NFPs and
Neurocytoma is a neuroepithelial tumour the tumour's characteristic clinicopatho- synaptophysin !1187,2638) Central neu-
composed of uniform round cells that logical features and immunohistochemi- rocytomas are not immunoreactive for the
show immunohistochemical and ultra- cal profile. When performed, electron R132-mutant IDH1 gene product !356).
structural features of neuronal differentia- microscopy shows regular round nuclei and p53 expression is usually lacking
tion. Additional features include fibrillary with finely dispersed chromatin and a !2638}
areas mimicking neuropil and a low prolif- small distinct nucleolus in a few cells.
eration rate. Various architectural patterns The cytoplasm contains mitochondria, a

Central neurocytoma 157


,

.......
Fig. 6.43 Central neurocytoma. A,B Round cells with nucleus-free areas of neuropil. C Synaptophysin is consistently expressed by the tumour cells, including in their processes
(1291}. D NeuN is diffusely expressed in all tumour nuclei. E GFAP is seen in only a few reactive astrocytes. F Ultrastructure showing numerous cell processes filled with
neurotubules and synaptic structures containing dense-core granules and clear vesicles.

Prol!feration including MYCN gain. A transcriptomic necessary after gross total resection,
The Ki-67 proliferation index is usually low study showed overexpression of genes many authors have recommended post-
(< 2%), but higher values can occur in atyp- involved in the WNT signalling pathway, operative radiotherapy after incomplete
ical central neurocytomas 12053,2478). calcium function, and maintenance of resection to prevent recurrence 11907).
neural progenitors 12637). These genes The prognostic relevance of atypical
Cell of origin may contribute to central neurocytoma histological features in central neurocy-
The cell of origin is still unknown. Because tumorigenesis. tomas and any consequent treatment
of the frequent involvement of the septum Central neurocytomas have not been re- strategy is more controversial; there have
pellucidum and the predominant neu- ported to exhibit 1p/19q codeletion, with been reports of central neurocytoma de-
ronal differentiation of the tumour, central the exception of a single case of atypical void of anaplastic histological features
neurocytoma was first thought to derive neurocytoma located in the insular cortex but associated with adverse outcome
from the nuclei of the septum pellucidum {1718). {2638).
{967). However, given the evidence for Shorter recurrence-free intervals have
both glial and neuronal differentiation in Prognosis and predictive factors been reported by some authors for cen-
some tumours, central neurocytoma may The clinical course of central neurocy- tral neurocytomas with a Ki-67 prolifera-
in fact derive from neuroglial precursor toma is usually benign, with the extent of tion index of > 2% or 3% {1561,2478).
cells with the potentiality of dual differ- resection being the most important prog- but this was not found by others 12638).
entiation. These tumours could originate nostic factor. Craniospinal dissemination Anaplastic histological features are not
from the subependymal plate of the lat- is exceptional {643,2567) In a meta- generally associated with poor progno-
eral ventricles {2662). An origin from analysis of 310 cases of central neuro- sis. However, a mitotic count of � 3 mi-
circumventricular organs has also been cytoma, the local control rates at 3 and toses per 10 high-power fields has been
proposed {1186). 5 years after gross total resection were found to be a predictor of higher risk of
95% and 85%, respectively, versus 55% recurrence {1462,2638). In one multicen-
Genetic profile and 45% after subtotal resection {2052). tre study of 71 patients, incomplete re-
Central neurocytomas contain numerous Although most clinical studies support section was predictive of poor outcome
DNA copy number alterations {1350). the assumption that radiotherapy is not {2638).

158 Neuronal and mixed neuronal-glial tumours


extraventricular neurocytoma Figarella-Branger D.
Soylemezoglu F.
Burger PC.
Park S.-H.
Honavar M.

Definition
A tumour composed of small uniform
cells that demonstrate neuronal differen-
tiation but are not /DH-mutant, and that
presents throughout the CNS, without
apparent association with the ventricular
system
Extraventricular neurocytoma is usually
well circumscribed and slow-growing,
and shares most histological features
with central neurocytoma Because Fig. 6.44 Extraventricular neurocytoma. A Hyperintense frontal mass on T2-weighted MRI. B Hypointense mass with
some tumours (including pilocytic as- no contrast enhancement is seen on T1-weighted MRI after gadolinium administration.
trocytomas, dysembryoplastic neuroepi-
thelial tumours, gangliogliomas, papillary literature ranges from 1 to 79 years, with extraventricular neurocytomas have
glioneuronal tumours [PGNTs], oligo- the median age in the fourth decade of been associated with seizures, head-
dendrogliomas with neurocytic features, life (938,2470). aches, visual disturbances, hemiparesis,
and diffuse leptomeningeal glioneuronal There does not seem to be a signifi- and cognitive disturbances; thalamic
tumours) show synaptophysin expres- cant relationship between sex and the and hypothalamic lesions with increased
sion, synaptophysin expression is not incidence of extraventricular neurocy- intracranial tension; and spinal lesions
sufficient to establish a diagnosis of ex- toma. The male-to-female ratio is about with motor, sensory, and sphincter dys-
traventricular neurocytoma. Strong and 1 :1, with a slight female predominance function (769,2470).
diffuse OLIG2 expression is not com- seen in some series and a slight male
patible with a diagnosis of extraventricu- predominance seen in others {269,769, Imaging
lar neurocytoma. Some entities that are 938). However, because most cases re- On MRI, extraventricular neurocytoma
genetically well defined (e.g. I DH-mutant ported before 2012 were not screened presents as a solitary, well-demarcated
and 1 p/19q-codeleted oligodendroglio- for IDH mutations, it is difficult to be cer- mass of non-specific signal intensity and
ma and PGNTs) should be excluded by tain of the diagnosis of extraventricular variable contrast enhancement, with a
appropriate genetic testing. However, the neurocytoma. cystic component in 58% of cases, mild
genetic characteristics of extraventricular perilesional oedema in 51.5%, and cal-
neurocytoma are not yet well defined. Etiology cification in 34% {269,1518,2822}. The
No specific etiology has been impli- solid component is predominantly isoin-
ICD-0 code 9506/1 cated in the genesis of extraventricular tense on T1-weighted images, but may
neurocytoma. be hypointense. On T2-weighted and
Grading FLAIR images, the signal is predomi-
Extraventricular neurocytomas corre- Localization nantly hyperintense.
spond histologically to WHO grade II. The cerebral hemispheres are the most
There have been suggestions for further common site for extraventricular neu- Spread
grading on the basis of mitotic rate, Ki-67 rocytoma (affected in 71% of cases). Extraventricular neurocytoma spread is
proliferation index, and the presence or The tumours most often affect the fron- particularly rare, although craniospinal
absence of vascular proliferation and/or tal lobes (in 30% of cases), followed by dissemination can occur as remote me-
necrosis {769). The term "atypical" has the spinal cord (in 14%). These tumours tastasis or along the surgical route (938}.
been used tor lesions with an elevated can occur in the thalamus, hypothalamic Cases of diffuse leptomeningeal neuroep-
mitotic rate and Ki-67 proliferation index region, cerebellum, and pons, with iso- ithelial tumour have been reported mainly
(1213). There is some evidence that each lated cases reported in cranial nerves, in children. These cases share with ex-
of these factors is associated with risk of the cauda equina, the sellar region, and traventricular neurocytoma some histolog-
recurrence, but definitive grading criteria even outside the craniospinal compart- ical and immunohistochemical features,
have not yet been established. ment {769,2470). including proliferation of a uniform popula-
tion of oligodendrocyte-like cells express-
Epidemiology Clinical features ing synaptophysin but not R132H-mutant
Extraventricular neurocytoma can pres- The clinical presentation varies accord- IDH1 {2294). These tumours represent a
ent in patients of any age. The patient ing to the location of the tumour and new entity (see Diffuse leptomeningeal
age at diagnosis of cases reported in the whether it exerts a mass effect. Cerebral glioneuronal tumour, p. 152).

Extraventricular neurocytoma 159


.
• ,• • • .
�-·
• • • • ••• •


Fig. 6.45 Extraventricular neurocytoma. A Spinal extraventricular neurocytoma composed of monomorphic cells that have round nuclei with fine nuclear chromatin.
B Focal ganglionic differentiation. C The cells, often with perinuclear haloes, have round nuclei with finely stippled chromatin and small nucleoli. D Occasional neoplastic cells
express GFAP. E Nuclear expression of NeuN. F Diffuse cytoplasmic immunoreactivity for synaptophysin is characteristic of extraventricular neurocytoma. The sparse neuropil
between tumour cells is also stained.

Macroscopy essential for the diagnosis of extraven- mandatory, to rule out diffuse glioma with
Some examples are well circumscribed tricular neurocytoma and is present in neurocytic differentiation 1356,2822} Be-
(sometimes with a cyst-mural nodule oligodendroglioma-like cells and larger cause 1 p/19q codeletion is a hallmark of
configuration), whereas others are more neurons. Clusters of neurosecretory oligodendroglioma, it is likely that pre-
infiltrative and therefore less discrete. granules are visualized by chromogranin- sumed extraventricular neurocytomas
A staining in some cases with ganglion exhibiting 1p/19q codeletion are in fact
Microscopy cells. In lesions with a glial component, oligodendrogliomas 12147) The differen-
A wide variety of histopathological ap- these cells may be positive for GFAP. tial diagnosis also includes ganglioglio-
pearances have been reported in this R132-mutant IDH1 protein is absent 121, ma, dysembryoplastic neuroepithelial
histologically heterogeneous lesion, 2822} Expression of OLIG2 has been tumour, and even PGNT, so it is recom-
which is usually more complex than the reported in some cases, but a more con- mended to test for BRAFV600E mutation
highly cellular, cytologically monomor- vincing example was immunonegative for and for SLC44A1-PRKCA fusion, a recur-
phous central neurocytoma 1269,816}. this marker. IDH and 1p/19q status were rent genetic alteration reported in PGNT
Tumours with the dense cellularity and not reported for these cases 11714,1832). 1278).
neuropil islands common in central neu- To date, neither high-throughput geno-
rocytoma are diagnostically straightfor- Cell of origin typing nor sequencing studies have
ward, but uncommon. More often, the There is no consensus on the putative been carried out. Microarray-based com-
tumours are less cellular and have an cell of origin. Extraventricular neurocy- parative genomic hybridization has been
oligodendroglioma-like appearance due toma may arise from mislocated neural performed in 2 cases, revealing distinct
to small, uniform round cells with arte- progenitor cells in the brain parenchyma profiles, with loss and gain of multiple
factually cleared cytoplasm embedded 1269} These neural progenitor cells dif- chromosomal loci 11732).
in a fibrillar matrix. Unlike in central neu- ferentiate into distinct cell lineages (neu-
rocytoma, ganglion cell differentiation is rocytic and astrocytic) in this particular Prognosis and predictive factors
common. Ganglioid cells - neurons inter- microenvironment 11876). Although extraventricular neurocytoma
mediate in size between ganglion cells is generally benign and has a low rate of
and neurocytes - are also frequent. Hya- Genetic profile recurrence, outcomes are known to vary
linized vessels and calcifications may be Although deletion of chromosome arms considerably. In one study, the presence
present. A glial astrocytic component is 1p and 19q (either in isolation or in com- of 1 p/19q codeletion was a poor prog-
uncommon, and can be difficult to distin- bination) has been found in extraven- nostic factor 12147). Gross total resection
guish from reactive gliosis. Cases with a tricular neurocytoma, neither IDH1/2 has been associated with a low rate of
convincing ganglion cell component can mutation nor MGMT methylation has recurrence 1269,769,1213}. Subtotally re-
be labelled ganglioneurocytoma. been reported 121,356,1206,1718,1732}. sected lesions are often stable, but recur-
Therefore, if extraventricular neurocyto- rence is possible 1269,769,1213}
lmmunophenotype ma is suspected on pathological exami-
lmmunoreactivity for synaptophysin is nation, investigation for IDH mutation is

160 Neuronal and mixed neuronal-glial tumours


cerebellar liponeurocytoma Kleihues P
Giangaspero F.
Chimelli L.
Ohgaki H.

Definition
A rare cerebellar neoplasm with ad-
vanced neuronal/neurocytic differentia-
tion and focal lipoma-like changes.
cerebellar liponeurocytoma affects
adults. has low proliferative activity, and
usually has a favourable prognosis. How-
ever, recurrence can occur and malig-
nant progression has been reported.

ICD-0 code 9506/1

Grading
Cerebellar liponeurocytoma corresponds
histologically to WHO grade II. Recur-
l
rences have been reported in almost Fig. 6.47 Cerebellar liponeurocytoma. A T1-weighted MRI (with gadolinium) of a recurrent liponeurocytoma, presenting
50% of cases. Recurrent tumours may as an irregular, strongly enhancing lesion in the right cerebellar hemisphere. Reprinted from Jenkinson MD et al. {1153).
display increased mitotic activity, an in- B Axial T1-weighted MRI after gadolinium administration shows areas of prominent hypointense signal within a well-
creased Ki-67 proliferation index, vascu- demarcated isodense tumour {53).
lar proliferation, and necrosis {817,1900,
2054). The time to clinical progression liponeurocytoma have been reported in Enhancement with gadolinium is usu-
ls often long (mean: 6.5 years). but in the English-language literature {1790}. ally heterogeneous, with areas of tumour
some cases relapse occurs within a few The mean patient age is 50 years (range: showing variable degrees of enhance-
months {1153}. 24-77 years), with peak incidence in the ment. On T2-weighted MRI, the tumour
third to sixth decades of life. There is no is slightly hyperintense to the adjacent
Synonyms significant sex predilection (1044,1908) brain, with focal areas of marked hyperin-
The terms "neurolipocytoma" {635}. "me- tensity. Associated oedema is minimal or
dullocytoma" [8171, "lipomatous glioneu- Localization absent {34). Fat-suppressed images may
rocytoma" (57}. and "lipidized mature Cerebellar liponeurocytoma most com- be helpful in supporting a preoperative
neuroectodermal tumour of the cerebel- monly involves the cerebellar hemi- diagnosis {53).
lum" (866} have been proposed. The spheres, but can also be located in the
term "cerebellar liponeurocytoma" is now paramedian region or vermis and extend Microscopy
widely accepted and is supported by ge- to the cerebellopontine angle or fourth Cerebellar liponeurocytoma is a very rare
netic analyses that indicate that this le- ventricle {1790). adult cerebellar neoplasm composed of
sion is a rare but distinct clinicopathologi- a uniform population of small neurocytic
cal entity [1044,1790,2400}. Clinical features cells arranged in sheets and lobules and
Headache and other symptoms and with regular round to oval nuclei, clear
Epidemiology signs of raised intracranial pressure (ei- cytoplasm, and poorly defined cell mem-
More than 40 cases of cerebellar ther from the lesion itself or due to ob- branes. The histological hallmark of this
structive hydrocephalus) are common entity is focal accumulation of lipid-laden
25 -�-----------,,--, presentations. Cerebellar signs, includ- cells that resemble adipocytes but con-
"'" ing ataxia and disturbed gait, are also stitute lipid accumulation in neuroepithe-
� 20
15 common (1867). lial tumour cells.
I� 15 Electron microscopy shows dense-core
e Imaging and clear vesicles, microtubule-contain-
�.!ll
o, 10
On CT, the tumour is variably isodense ing neurites, and (occasionally) synapse-
� or hypodense, with focal areas of marked like structures (766,817)
§ 5
o hypoattenuation corresponding to fat The growth fraction of the small-cell com-
w m m � m w ro w density (53,1790}. On T1-weighted MRI, ponent, as determined by the Ki-67 pro-
Age at diagnosis the tumour is isointense to hypointense, liferation index, is usually in the range of
Fig. 6.46 Age distribution of cerebellar liponeurocytoma, with patchy areas of hyperintensity corre- 1-3%, but can be as high as 6%, with a
. based on 25 published cases. sponding to regions of high lipid content. mean value of 2.5% (635,1200,2400}. In

Cerebellar liponeurocytoma 161


.,

.B . . .
,
Fig. 6.49 Cerebellar liponeurocytoma. A Small tumour cells and neoplastic cells with lipomatous differentiation express MAP2; similarly, liponeurocytomas also express synaptophysin.
B Expression of the astrocytic marker GFAP is observed in most cases, but only focally. C The Ki-67 proliferation index (as determined by nuclear MIB1 monoclonal antibody staining)
is usually low.

the adipose component, the Ki-67 prolif- 2054). Similarly, the lipidized component tent expression of neuronal markers, in-
eration index is even lower. Features of may be markedly reduced or even ab- cluding neuron-specific enolase, syn-
anaplasia such as nuclear atypia, necro- sent {2054) in recurrent lesions. aptophysin, and MAP2. Focal GFAP
sis, and microvascular proliferation are expression by tumour cells, which in-
typically absent in primary lesions, but /mmunophenotype dicates astrocytic differentiation, is ob-
may be found in recurrent tumours {817, lmmunohistochemically, there is consis- served in most cases {2400} One report

..
.
•t-'.
:• �- ..
... . �
Fig. 6.50 Cerebellar liponeurocytoma. Electron microscopy shows dense-core and clear vesicles, microtubule-containing neurites, and occasionally synapse-like structures.

162 Neuronal and mixed neuronal-glial tumours


genetic pathways !1044). BRAF and /DH
AAG G AIA1)-fTI T T G c T TG AG IG�GIC G TG T mutations were absent in a recently re-
90 100 50 ported case in which the recurrent lesion
showed anaplastic changes !2054)

Prognosis and predictive factors


The low proliferative activity is accom-
panied by a favourable clinical outcome.
Most patients with sufficient follow-up
survived > 5 years, largely irrespective of
� whether adjuvant radiotherapy was ad-
Codon 200 AA T --ACT Codon 272 GTG --GCG ministered. The longest known survival
Fig. 6.51 Cerebellar liponeurocytoma. Examples of TP53 missense mutations in codons 200 and 272 detected in is 18 years, in a patient whose treatment
cerebellar liponeurocytoma (1044). was limited to surgical excision. Howev-
er, recurrence and radioresistance have
mentioned immunoreactivity to desmin an origin of cerebellar liponeurocytoma been reported !1153} Of 21 patients with
and morphological features of incipient from cerebellar progenitors, which are follow-up data, 6 (29%) died between
myogenic differentiation !866). distinct from cerebellar granule progeni- 6 months and 2 years after surgical inter-
The growth fraction as determined by tors and aberrantly differentiate into adi- vention, 5 (24%) died 2-4 years after sur-
the Ki-67 proliferation index is usually in pocyte-like tumour cells !73). gical intervention, and 10 (48%) survived
the range of 1-3%, but can be as high for 5-16 years after surgical intervention.
as 10% in recurrent lesions !2054). In the Genetic profile The 5-year survival rate was 48%, and
adipose component, the Ki-67 prolifera- Genetic analysis of 20 cases showed the the mean overall survival was 5.8 years
tion index is lower. presence of TP53 missense mutations in !32,1044). However, 62% of the patients
20%. The absence of isochromosome developed a recurrence, after 1-12 years
Cell of origin 17q and the lack of PTCH, CTNNB1, and (mean: 6.5 years). In 3 patients, there was
A recent study demonstrated that the APC mutations suggest that liponeurocy- a second relapse 1-5 years later (mean:
transcription factor NGN1, but not ATOH1. tomas are not likely a variant of medul- 3 years). Recurrent tumours may show
is expressed in cerebellar liponeurocy- loblastoma. This assumption was further increased mitotic activity, increased pro-
toma (unlike in normal adult cerebellum) supported by gene expression profiles liferative activity, vascular proliferation,
and that adipocyte fatty acid-binding suggesting that this neoplasm is closer and necrosis !817,2054). although some
protein, typically found in adipocytes, is to neurocytoma than to the medullo- tumour recurrences lack these atypical
significantly overexpressed in cerebel- blastoma subgroups !1044). However, histopathological features !1153). Histo-
lar liponeurocytoma compared with both the presence of TP53 mutations, which pathological properties predicting recur-
normal adult cerebellum and human me- are absent in central neurocytomas, in- rence have not been identified.
dulloblastoma. These findings suggest dicates development through different

Cerebellar liponeurocytoma 163


Paraganglioma Brandner S.
Soffer D.
Stratakis CA
Yousry T.

Definition ratio: 1 .4-1.7:1) {2790). Jugulotympanic clinical features. Common presenting


A unique neuroendocrine neoplasm, paragangliomas are more common in symptoms include a history of low-back
usually encapsulated and benign, arising Caucasians, have a strong female predi- pain and sciatica. Less common manifes-
in specialized neural crest cells associ- lection, and occur mainly in the fifth and tations are numbness, paraparesis, and
ated with segmental or collateral auto- sixth decades of life {1111) In a series of sphincter symptoms. Fully developed
nomic ganglia (paraganglia), consisting 200 cases of paragangliomas, 9% were cauda equina syndrome is uncommon.
of uniform chief cells exhibiting neuronal located intraspinally {2790) Phaeochro- Signs of increased intracranial pressure
differentiation forming compact nests mocytomas and paragangliomas are and papilloedema are an unusual pre-
(Zellballen) surrounded by sustentacular rare tumours with a combined estimated sentation {15,120,328,794,2236) Endo-
cells and a delicate capillary network. annual clinical incidence of 3 cases per crinologically, functional paragangliomas
In the CNS, paragangliomas primarily 1 million population {147). of the cauda equina region are extremely
affect the cauda equina I filum terminale rare {794) They present with signs of
and jugulotympanic regions. Localization catecholamine hypersecretion, such as
Overall, paragangliomas of the cauda episodic or sustained hypertension, pal-
ICD-0 code 8693/1 equina region constitute 3.4-3.8% of all pitations, diaphoresis, and headache.
tumours affecting this location {2765, Another unusual presentation of cauda
Grading 2815). Other spinal levels are involved far equina paraganglioma is with subarach-
Paragangliomas of the filum termina- less often; 19 paragangliomas have been noid haemorrhage {1492). Cerebrospinal
le correspond histologically to WHO reported in the thoracic region, most of fluid protein levels are usually markedly
grade I. which were extradural with an intraverte- increased {2389,2395).
bral and paraspinal component {397,489, The reported paragangliomas of the tho-
Epidemiology 795,2363,2531) and 5 of which involved racic spine presented with signs of spinal
Paragangliomas of the CNS are uncom- the cervical region {211,422,1542,1831, cord compression or signs of catechola-
mon. Most present as spinal intradural 2456). lntracranial paragangliomas are mine hypersecretion {1143,2363) About
tumours in the region of the cauda equi- usually extensions of jugulotympanic 36% of all jugulotympanic paraganglio-
na I filum terminale. Almost 300 cases paragangliomas {1111 ). However, rare mas extend into the cranial cavity {1111).
have been reported since 1970, when examples of purely intracranial tumours These most often present with pulsatile
cauda equina region paraganglioma have been situated in the sellar region tinnitus and lower cranial nerve dysfunc-
was first described {1666) Many other {417,592,1742). cerebellopontine angle tion {1111).
cases have undoubtedly gone unre- {559,844). cerebellar parenchyma {1496,
ported. Cauda equina paragangliomas 2025,2226). and various locations in the Imaging
generally affect adults, with peak inci- forebrain { 638, 1646, 2097, 2828). MRI is the investigative procedure of
dence in the fourth to sixth decades of choice, although the findings are non-
life. Patient age ranges from 9 to 75 years Clinical features specific {15); the appearance of para-
(mean age: 46 years) {2811). with a slight Like other spinal tumours, cauda equina ganglioma is indistinguishable from that
male predominance (male-to-female paragangliomas exhibit no distinctive of schwannoma or ependymoma {1682).

Males Females
90-100

80-89
70-79

60-69
50-59

40-49

30-39
20-29
10-19

0-9
30 20 10 10 20 30 Fig. 6.53 A lntraoperative aspect of a spinal paraganglioma attached to the filum terminale. B Macroscopic aspect
Number of cases Number of cases of a spinal paraganglioma attached to a nerve root. A well-circumscribed, solid tumour, partly attached to a spinal root;
Fig. 6.52 Age and sex distribution of spinal formalin fixed.
paraganglioma, based on 71 published cases.

164 Neuronal and mixed neuronal-glial tumours


gross total removal {2438). Cerebrospinal
fluid seeding of spinal paragangliomas
has occasionally been documented
{487,2144,2438,25401 Although para-
gangliomas in general are considered
benign, about 10-20% of them have
metastatic potential {14371. In contrast,
metastasis outside the CNS (to the bone)
from cauda equina paragangliomas has
been reported only once {17121. As is
the case in paragangliomas in general,
there is no single histological parameter
that can predict malignant behaviour in
cauda equina paragangliomas {16551.
Numerous factors have been associated
with malignancy in paragangliomas in
general, including SDHB mutations, high
proliferation index, and large tumour size.
Tumour size cut-off points of 5-6 cm of
diameter and 80-150 g of weight have
Fig. 6.54 MRI of spinal paraganglioma: a T2-weighted image (left), a T1-weighted image (centre), and a T1-weighted been suggested to predict malignant be-
image with gadolinium contrast enhancement (right) show the encapsulated, well-demarcated tumour. haviour {16551. However, the only accept-
ed criterion for malignancy is the pres-
The tumour can be hypointense, isoin- A salt-and-pepper appearance, caused ence of distant metastasis {378,16551.
tense, or hyperintense on T1- and T2- by the hypervascular structure, can be
weighted images, and gadolinium en- seen on T2-weighted images. Cystic Macroscopy
hancement may be present or absent. changes can be caused by intratumoural Generally, paragangliomas are oval to
Typically, paraganglioma presents as haemorrhage. Plain X-rays are usually sausage-shaped, delicately encapsu-
a sharply circumscribed, occasionally non-informative, but rarely show erosion lated, soft, reddish-brown masses that
partly cystic mass that is hypointense or (scalloping) of vertebral laminae due to bleed freely. The 59 spinal tumours in-
isointense to spinal cord on T1-weighted chronic bone compression. cluded in one group of five series meas-
images, markedly contrast-enhancing, ured 10-112 mm in greatest dimension
and hyperintense on T2-weighted im- Spread {1609, 1682,2811 }. Capsular calcification
ages. The presence of a T2-hypointense The vast majority of cauda equina para- and cystic components may be found.
rim (the so-called cap sign, related to gangliomas are slow-growing and cur- The tumours occasionally penetrate dura
haemosiderin content) is considered di- able by total excision; with long-term fol- and invade bone. Most paragangliomas
agnostically helpful {1478,2811,28151. low-up, it is estimated that 4% recur after of the cauda equina are entirely intradural
and are attached either to the filum ter-
minale or (less often) to a caudal nerve
root {23951.

Microscopy
Paragangliomas are well-differentiated
tumours resembling normal paragan-
glia. They are composed of chief (type I)
cells arranged in nests or lobules (called
Zellballen) surrounded by an inconspicu-
ous single layer of sustentacular (type II)
cells. The Zellballen are also surrounded
by a delicate capillary network and a del-
icate supporting reticulin fibre network
that may undergo sclerosis. The uni-
form round or polygonal chief cells have
central, round to oval nuclei with finely
stippled chromatin and inconspicuous
nucleoli. Degenerative nuclear pleomor-
phism is typically mild. The cytoplasm is
usually eosinophilic and finely granular.
In some cases, it is amphophilic or clear.
Sustentacular cells are spindle-shaped;

Paraganglloma 165
encompassing the lobules, their long
processes are often so attenuated that
they are not visible on routine light mi-
croscopy and can be detected only on
immunostains for S100 protein. Approxi-
mately 25% of all cauda equina paragan-
gliomas are so-called gangliocytic para-
gangliomas, containing mature ganglion
cells and a Schwann cell component
!313). Ependymoma-like perivascular
formations are also common. Some tu-
mours show architectural features remi-
niscent of carcinoid tumours, including
angiomatous, adenomatous, and pseu-
dorosette patterns {2395). Tumours com-
posed predominantly of spindle {1712}
and melanin-containing cells (called mel-
anotic paragangliomas) {773,1712) have
also been described at this site, as has
oncocytic paraganglioma {1889) Foci of
haemorrhagic necrosis may occur, and
scattered mitotic figures can be seen.
Neither these features nor nuclear pleo-
morphism is of prognostic significance Gangliocytic paragangliomas containing syndromes caused by any SDH muta-
(23951. a variable mixture of epithelioid neuroen- tion. Although SDHB immunohistochem-
docrine cells, Schwann cell-like cells, istry has become part of the routine as-
Ultrastructure and scattered ganglion cells can show sessment of paragangliomas in many
The distinctive ultrastructural feature of cytokeratin positivity in the epithelioid centres (1655). it is probably of limited
chief cells is the presence of dense-core cells (894,23281. Expression of 5-HT value in cauda equina paragangliomas
(neurosecretory) granules measuring and various neuropeptides (e.g. soma- given the very low rate of SDH mutations
100-400 nm (mean: 140 nm). Sustentac- tostatin and met-enkephalin) has been (1420,1955).
ular cells are characterized by an elon- demonstrated in paragangliomas of the Sustentacular cells show inconsistent
gated nucleus with marginal chromatin, cauda equina region (1712,23951. Loss (sometimes only focal) S100 protein re-
increased cytoplasmic electron density, of SDHB expression is considered a sur- activity (3131 and usually show staining
relative abundance of intermediate fila- rogate marker for familial paraganglioma for GFAP as well. Chief cells may also
ments, and lack of dense-core granules
(647,23951.

/mmunophenotype
Consistent with their neuroendocrine
differentiation, the chief cells of para-
gangliomas are immunoreactive for the
commonly used neuroendocrine mark-
ers chromogranin-A and synaptophysin
{1303,1655,2395). The diagnosis of para-
ganglioma in other sites is confirmed by
positivity for tyrosine 3-monooxygenase,
the rate-limiting enzyme in catechola-
mine synthesis (1655). However, this test
is usually not required for cauda equina
paragangliomas, because it distinguish-
es paragangliomas from other neuroen-
docrine carcinomas, which are not con-
sidered in the differential diagnosis in
this location. Whereas paragangliomas
in other sites are cytokeratin-immunon-
egative, those arising from the cauda
equina can show positivity for cytokerat- �
ins, typically in the form of perinuclear Fig. 6.57 Paraganglioma immunophenotype. A Chromogranin. B Synaptophysin. C Spinal paragangliomas express
immunoreactivity {432,894, 1655,24381. cytokeratins. D S100 is expressed by sustentacular cells and occasional chief cells.

166 Neuronal and mixed neuronal-glial tumours


shOW variable 8100 immunoreactivity. Genetic susceptibility with recurrent spinal paraganglioma and
Recent guidelines suggest that prolif- It is estimated that as many as half of all a cerebellar metastasis {1605). System-
eration rate (mitotic activity and Ki-67 phaeochromocytomas/paragangliomas ic paragangliomas may be multifocal,
proliferation index) should be recorded in adults and > 80% of these tumours in but no association has been reported
tor paragangliomas as well as for other children are inherited {2139) between cauda equina paragangliomas
neuroendocrine tumours {1655}; how- To date, autosomal dominant germline and other spinal paragangliomas. Con-
ever, the value of proliferation markers in mutations of > 10 genes have been current cases of spinal paraganglioma
cauda equina paragangliomas has not described in association with these tu- with brain tumours (339,487). spinal epi-
been established. mours: VHL (associated with von Hippel- dural haemangioma {2735). syringomye-
Lindau disease); RET (associated with lia {2419). and intramedullary cysts {672)
Cell of origin multiple endocrine neoplasia type 2); have been reported, but these associa-
The histogenesis of cauda equina para- NF1 (associated with neurofibromatosis tions may be coincidental.
ganglioma is a matter of debate. Some type 1); genes coding for the subunits of Paraganglioma and gastrointestinal stro-
authors favour an origin from paragan- the succinate dehydrogenase enzyme - mal tumour are associated as charac-
glion cells associated with regional au- SOHO (associated with inherited para- teristics of Carney-Stratakis syndrome,
tonomic nerves and blood vessels, de- ganglioma-1 ), SOHA and SOHAF2 (as- a familial syndrome inherited in an au-
spite the fact that such cells have not sociated with paraganglioma-2), SOHC tosomal dominant manner {2436) The
been identified at this site {1513) Others (associated with paraganglioma-3), and disease is caused by mutations and/or
have suggested that peripheral neuro- SOHB (associated with paraganglio- deletions of SOHA, SOHB, SOHC, and
blasts normally present in the adult filum ma-4) - which forms part of mitochondri- SOHO, in > 90% of the described cases
terminale undergo paraganglionic dif- al complex II (778,840); and the tumour {1898}. Paragangliomas or phaeochro-
ferentiation {339,2204). Jugulotympanic suppressors TMEM127 (2049} and MAX mocytomas can also be found as part of
paragangliomas presumably arise from (4841. Multiple paragangliomas and/or an allelic condition called Carney triad.
microscopic paraganglia within the tem- phaeochromocytomas are often caused This sporadic syndrome is seen almost
poral bone (1412). Of interest, although by SOHO, SOHAF2, SOHB, SOHC, exclusively in females and may be due to
perhaps not relevant to histogenesis, are and SOHA mutations. whereas isolated epigenetic alterations of SOHC {930) or
reports of the coexistence of paragan- phaeochromocytomas are also associ- other defects of the SOHC chromosomal
glioma and myxopapillary ependymoma ated with TMEM127 and MAX mutations locus on 1q {1614}. Paragangliomas and/
in the cauda equina region {1243} and (778). An association between paragan or phaeochromocytomas can also be
of a biphasic tumour consisting primarily gliomas/phaeochromocytomas and fu- seen in association with renal cancer
of paraganglioma and to a lesser extent marase defects was recently reported (841). pituitary tumours (2798). and pos-
ependymoma (339}. {380). NF1 mutations seem to be more sibly thyroid tumours (1777).
frequent than previously thought {2726). In general, a single benign paragan-
Genetic profile Genes with roles in multiple neoplasia glioma may not be indicative of any ge-
The mutations found in paragangliomas syndromes, such as the VHL gene, may netic predisposition, whereas multiple
are described in Genetic susceptibility also be epigenetically modified (711; the paragangliomas or an association of a
The genetic and epigenetic profiles VHL gene is epigenetically inactivated in paraganglioma with another neoplasia
(i.e methylation, expression {930,1475). phaeochromocytomas and abdominal (e.q. phaeochromocytoma, gastrointes-
microRNA {540). and metabolomics paragangliomas {71). tinal stromal tumour, renal cancer, pitui-
(381,1090)) of phaeochromocytomas/ Spinal paragangliomas may be non- tary adenoma, or thyroid cancer) should
paragangliomas with succinate dehydro- familial in most cases, but a study of prompt investigation of a possible genet-
, genase defects differ dramatically from 22 spinal paragangliomas showed an ic syndrome underlying the presentation.
those with other genetic causes. SOHO germline mutation in one patient

Paraganglioma 167
CHAPTER 7

Tumours of the pineal region


Pineocytoma
Pineal parenchymal tumour of intermediate differentiation
Pineoblastoma
Papillary tumour of the pineal region
Pineocytoma Nakazato Y.
Jouvet A.
Vasiljevic A.

Definition affect adults, with a mean patient age


A well-differentiated pineal parenchymal of 42.8 years {91,1187,1674,2279,2809l.
neoplasm composed of uniform cells There is a female predominance, with a
forming large pineocytomatous rosettes male-to-female ratio of 0.6:1.
and/or of pleomorphic cells showing
gangliocytic differentiation. Localization
Pineocytoma is a rare neoplasm. It ac- Pineocytomas typically remain localized
counts for about 20% of all pineal pa- in the pineal area, where they compress
renchymal tumours and typically affects adjacent structures, including the cere-
adults, with a mean patient age at diag- bral aqueduct, brain stem, and cerebel-
nosis of 43 years. There is a female pre- lum. Protrusion into the posterior third
dominance, with a male-to-female ratio ventricle is common.
of 0.6:1. Other characteristics are exclu-
sive localization in the pineal region and Clinical features
a well-demarcated solid mass without Because of expansile growth in the pin-
infiltrative or disseminating growth. Spe- eal region, pineocytomas present with Fig. 7.02 Sagittal gadolinium-enhanced T1-weighted
MRI of a pineocytoma in the pineal region.
cific genetic alterations have not yet been signs and symptoms related to increased
identified. The prognosis is good after to- intracranial pressure due to aqueductal
tal surgical removal. obstruction, neuro-ophthalmological dys-
function (i.e. Parinaud syndrome), and
ICD-0 code 9361/1 brain stem or cerebellar dysfunction
{240,445,465,927,2279}. Common pres-
Grading entations include headache, papilloede-
Pineocytoma corresponds histologically ma, ataxia, impaired vision, nausea and
to WHO grade I. vomiting, impaired ambulation, loss of
upward gaze, dizziness, and tremor.
Epidemiology
Pineal region tumours account for < 1% Imaging
of all intracranial neoplasms, and ap- On CT, pineocytomas usually present as Fig. 7.03 Sagittal section of a large pineocytoma
extending into the third ventricle. Note the granular cut
proximately 27% of pineal region tumours globular, well-delineated masses < 3 cm
surface with occasional cysts.
are of pineal parenchymal origin {1332, in diameter. They appear hypodense and
2458l. Of these, pineocytomas account homogeneous, some harbouring either
for 17-30% (mean: 20%) {91,1187,2279, peripheral or central calcification {435l. locally but are not associated with cere-
2809l. Before the classification of pineal Occasional cystic changes may be seen brospinal fluid seeding {677l
parenchymal tumour of intermediate dif- and are usually not easily confused with
ferentiation as a distinct entity, as many typical pineal cysts {665l Most tumours Macroscopy
as 60% of pineal parenchymal tumours exhibit heterogeneous contrast enhance- Pineocytomas are well-circumscribed le-
were classified as pineocytomas {997, ment. lsodense to slightly hyperdense sions with a greyish-tan, homogeneous
1638l. Pineocytomas can occur in pa- appearance and homogeneous contrast or granular cut surface {240,997,2258l.
tients of any age, but most frequently enhancement on CT have also been Degenerative changes, including cyst
reported {1751). Accompanying hydro- formation and foci of haemorrhage, may
cephalus is a common feature {2279). On be present {1638).
80-89-

70-79---
MRI, the tumours tend to be hypointense
or isointense on T1-weighted images and Microscopy

·�·------------·
hyperintense on T2-weighted images,
50-59 ------------·
with strong, homogeneous contrast en- Histopathology
30-39 -------
hancement {1751}. The margins with sur- Pineocytoma is a well-differentiated, mo-
20-29 ---------- rounding structures are usually well de- derately cellular neoplasm composed
0-9-
fined, and are best analysed by MRI. of relatively small, uniform, mature cells
resembling pinealocytes. It grows pri-
Fig. 7.01 Age distribution of patients with pineocytoma, Spread marily in sheets, and often features large
based on 63 cases, both sexes. Strictly defined pineocytomas grow pineocytomatous rosettes composed of

170 Tumours of the pineal region


. ... - - ...
Fig. 7.04 A Typical pineocytoma. A sheet of tumour cells with scattered pineocytomatous rosettes. B Pineocytoma. Uniform tumour cells resembling pinealocytes.

abundant delicate tumour cell processes. is characterized by large ganglion cells annulate lamellae, cilia with a 9 + O mi-
Pineocytomatous rosettes are not seen and/or multinucleated giant cells with bi- crotubular pattern, microtubular sheaves,
in normal pineal gland. In pineocytoma, zarre nuclei (240,997,1384,1638,2279). fibrous bodies, vesicle-crowned rodlets,
poorly defined lobules may be seen, but The mitotic activity of this pattern is still heterogeneous cytoplasmic inclusion,
a conspicuous lobular architecture is in- low, despite the tumours' ominous nu- and membrane whorls, as well as mito-
stead a feature of normal pineal gland. clear appearance. The stroma of pineo- chondrial and centriolar clusters. Mem-
Most nuclei are round to oval, with incon- cytoma consists of a delicate network of brane-bound dense-core granules and
spicuous nucleoli and finely dispersed vascular channels lined by a single layer clear vesicles are present in the cyto-
chromatin. Cytoplasm is moderate in of endothelial cells and supported by plasm and cellular processes. The cellu-
quantity and homogeneously eosino- scant reticulin fibres. Microcalcifications lar processes show occasional synapse-
philic. Processes are conspicuous and are occasionally seen but usually corre- like junctions.
short. often ending in club-shaped ex- spond to calcifications of the remaining
pansions that are optimally demonstrated pineal gland. lmmunophenotype
by neurofilament immunostaining or sil- Pineocytoma cells usually show strong
ver impregnation. Mitotic figures are lack- Electron microscopy immunoreactivity for synaptophysin, neu-
ing (with < 1 mitosis per 10 high-power Ultrastructurally, pineocytomas are com- ron-specific enolase, and NFP. Variable
fields) in all but occasional large speci- posed of clear and various numbers of staining has also been reported for other
mens (1187). Pineocytomatous rosettes dark cells joined with zonulae adherentes neuronal markers, including class Ill be-
vary in number and size. Their anucleate (968,997,1185,1674). The cells extend ta-tubulin, microtubule-associated pro-
centres are composed of delicate, en- tapering processes that occasionally ter- tein tau, UCHL1, chromogranin-A, and
meshed cytoplasmic processes resem- minate in bulbous ends. Their cytoplasm the neurotransmitter 5-HT (477,1185,1187,
bling neuropil (240,1187,1811,2279). The is relatively abundant and contains well- 1384,1811,2809). Photosensory differen-
nuclei surrounding the periphery of the developed organelles. Pineocytoma tiation is associated with immunoreactiv-
rosette are not regimented. A pleomor- cells share numerous ultrastructural fea- ity for S-arrestin and rhodopsin (1638,
phic cytological variant is encountered tures with normal mammalian pinealo- 1811,1933). In pleomorphic variants, the
in some pineocytomas (701). This variant cytes, such as paired twisted filaments,

Fig. 7.05 Pleomorphic pineocytoma. A Pleomorphism and gangliocytic differentiation. B Mono- and multinucleated ganglion cells.

Pineocytoma 171
.,

Fig. 7.06 Pineocytoma. A Pineocytomatous rosettes show intense immunoreactivity for synaptophysin.
for NFP. C Pleomorphic cells often show immunoreactivity for NFP.

ganglioid cells usually express several pinealocytes predominate. To a vari- the RB1 gene and pineocytoma has not
neuronal markers, especially NFP. able extent, pineal parenchymal tumours been established. A microarray analysis
mimic the developmental stages of the of pineocytoma has shown high-level ex-
Pro/Jferation human pineal gland. In tissue culture, pression of genes coding for enzymes
In most cases, mitotic figures are very pineocytoma cells are also capable of related to melatonin synthesis (i.e. TPH1
rare or absent {702,1187,1217). The mean synthesizing 5-HT and melatonin (700). and ASMT) and genes involved in retinal
Ki-67 proliferation index is < 1% {91,702, The immunoexpression of CRX and phototransduction (i.e. OPN4, RGS16,
1217l, ASMT in pineocytoma is an additional and CRB3). These reactivities indicate
indication that these tumours are biologi- bidirectional neurosecretory and photo-
Cell of origin cally linked to pinealocytes {754,2241}. sensory differentiation {698).
The histogenesis of pineal parenchymal CRX is a transcription factor involved in
tumours is linked to the pinealocyte, a the development and differentiation of Genetic susceptibility
cell with photosensory and neuroendo- pineal cell lineage, and ASMT is a critical No syndromic associations or genetic
crine functions. The ontogeny of the hu- enzyme for the synthesis of melatonin (a susceptibilities have been demonstrated.
man pineal gland recapitulates the phy- hormone produced by the pineal gland). The occurrence of pineocytoma in sib-
logeny of the retina and the pineal organ lings was reported in one family {796).
(1675). During the late stages of intrauter- Genetic profile
ine life and the early postnatal period, the Conventional cytogenetic studies based Prognosis and predictive factors
human pineal gland consists primarily of on karyotypes are rare. Karyotype analy- The clinical course of pineocytomas is
cells arranged in rosettes similar to those sis of 3 pineocytomas demonstrated characterized by a long interval (4 years
of the developing retina. These feature a pseudodiploid or hypotriploid profile in one series) between the onset of symp-
abundant melanin pigment as well as with various numerical and structural ab- toms and surgery {240). No strictly clas-
cilia with a 9 + 0 microtubular pattern. By normalities, including loss of all or part sified pineocytomas have been shown
the age of 3 months, the number of pig- of chromosome 22, loss or partial dele- to metastasize {677,2278). The reported
mented cells gradually decreases so that tion of chromosome 11, loss of chromo- 5-year survival rate of patients with pine-
pigment becomes undetectable by his- some 14, and gain of chromosomes 5 ocytoma ranges from 86% to 91% (677.
tochemical methods {1675). As differen- and 19 {162,525,2061) However, no 2278). In one series, the 5-year event-
tiation progresses, cells that are strongly chromosomal gains or losses were found free survival rate was 100% (677). Extent
immunoreactive for neuron-specific eno- by comparative genomic hybridization of surgery is considered to be the major
lase accumulate. By the age of 1 year, analysis {2122). A relationship between prognostic factor for pineocytoma (465).

e ' .. IJ: \ • .
Fig. 7.07 Pineocytoma. A In a pineocytomatous rosette, tumour cells surround an eosinophilic fibrillated core. B Argyrophilic tumour cell processes end with club-shaped
expansions in the pineocytomatous rosettes (Bodian silver impregnation). C Ultrastructure of a pineocytomatous rosette, showing numerous cell processes filled with clear vesicles,
dense-core granules, and mitochondria.

172 Tumours of the pineal region


Pineal parenchymal tumour of Jouvet A.
Nakazato Y.
intermediate differentiation Vasiljevic A.

Definition anaplasia" (2586). and "pineoblastoma pineal gland into the posterior third ven-
A tumour of the pineal gland that is in- with lobules" (240). These terms have ob- tricle, with compression of the corpora
termediate in malignancy between pineo- scured the value of the designation, and quadrigemina and compromise of cere-
cytoma and pineoblastoma and is com- they are not recommended. brospinal fluid flow through the aqueduct.
posed of diffuse sheets or large lobules In earlier studies, no true PPTID group Compression of the superior colliculus by
of monomorphic round cells that appear was identified. However, mixed pineo- the expanding mass may cause ocular
more differentiated than those observed cytoma-pineoblastoma was sometimes movement abnormalities (i.e. Parinaud
in pineoblastomas. described as an intermediate tumour be- syndrome), including paralysis of upward
Pleomorphic cytology may be present. tween pineocytoma and pineoblastoma gaze, pupillary abnormalities (i.e. slightly
Pineal parenchymal tumours of interme- (1638,2279). By definition, this tumour is dilated pupils that react to accommoda-
diate differentiation (PPTIDs) occur main- composed of clearly delineated areas of tion but not to light), and nystagmus re-
ly in adults (mean patient age: 41 years), pineoblastoma admixed with well-demar- tractorius (69,195,757,2319). A rare case
and show variable biological and clini- cated areas of pineocytoma. Neoplastic of apoplectic haemorrhage of a PPTID
cal behaviour, from low-grade tumours cells lack the so-called intermediate mor- with sudden-onset symptoms has been
with frequent local and delayed recur- phology that is required for a diagnosis reported (2684).
rences to high-grade tumours with risk of of PPTID. Mixed pineocytoma-pineo-
craniospinal dissemination. Accordingly, blastoma should not be included in the Imaging
mitotic activity, Ki-67 proliferation index, PPTID group, but rather belongs in the On imaging, PPTIDs usually present as
and neuronal and neuroendocrine differ- pineoblastoma group (see Pineoblasto- bulky masses with local invasion. They
entiation are also variable, and reported ma, p. 176). are more rarely circumscribed. On CT, the
5-year overall survival rates range from tumours may show occasional peripheral
39% to 74% (677,1187). Epidemiology so-called exploded calcifications (1321).
PPTIDs account for approximately 45% On MRI, PPTIDs are heterogeneous and
ICD-0 code 9362/3 of all pineal parenchymal tumours, with mostly hypointense on T1-weighted im-
a range of 21-54% in most recent series ages and hyperintense on T2-weighted
Grading (91,1105,2809,2868). Earlier reported in- images. On both CT and MRI, postcon-
The biological behaviour of pineal paren- cidence rates of PPTID were even more trast enhancement is usually marked and
chymal tumour of intermediate differen- variable, from 0% to 59%, reflecting the heterogeneous (1105,1321).
tiation is variable and may correspond to general ignorance of this pineal paren-
WHO grades II or Ill, but definite histolog- chymal tumour, the frequent misdiag- Spread
ical grading criteria remain to be defined. nosis of the entity, and/or the inclusion PPTIDs have a potential for local recur-
of mixed pineocytoma-pineoblastomas rence and craniospinal dissemination
Synonyms and historical annotation and other unusual pineal parenchymal tu- (677,1105,2708,2835) Local recurrence
The category of PPTID was first clearly mours in this group (240,1187,1638,1811) occurs in approximately 22% of cases
introduced in 1993 by Schild et al. (1185, PPTIDs can occur in patients of any age, (677). Craniospinal dissemination may be
1674,2279). PPTIDs have been reported but most frequently affect adults, with observed at the time of diagnosis (in 10%
under various names, such as "malignant a mean patient age of 41 years (range: of cases) or may occur during the course
pineocytoma" (997), "pineocytoma with 1-83 years) (754,1105,1187,2809). There of the disease (in 15% of cases) (677).
is a slight female preponderance, with a
male-to-female ratio of 0.8:1. Macroscopy
80-89.
The gross appearance of PPTIDs is simi-
70-79-
Localization lar to that of pineocytomas. They are cir-

·�·
60-69 .
PPTIDs are localized in the pineal region. cumscribed, soft in texture, and lacking
S0.59 .

,0.39
... ..
Clinical features
gross evidence of necrosis. An irregular
tumour surface was observed by endos-
20-29 .
The clinical presentation is similar to that copy in a case with spinal metastasis
10-,, _ of other pineal parenchymal tumours. (1105).
o-s _ The main symptoms are headaches and
10 15 25 30
vomiting, related to increased intracranial Microscopy
Fig. 7.08 Age distribution of pineal parenchymal tumours pressure caused by obstructive hydro- PPTID may exhibit two architectural
of intermediate differentiation, based on 142 published cephalus. Hydrocephalus is the con- patterns: diffuse (neurocytoma- or oli-
cases, both sexes. sequence of tumoural extension of the godendroglioma-like) and/or lobulated

Pineal parenchymal tumour of intermediate differentiation 173


.,

I - • Jf_ ...._ .. - _.; w;;;;;:,'-'"'�-.a....�--


fig. 7.09 Pineal parenchymal tumour of intermediate differentiation. A Neurocytoma-like appearance in a pineal parenchymal tumour of intermediate differentiation. Tumour with
moderate cellularity and round nuclei harbouring salt-and-pepper chromatin; the fibrillary background is characterized by small pseudorosettes; larger pseudorosettes, as seen in
pineocytomas, are not observed. B Pseudolobulated pineal parenchymal tumour of intermediate differentiation. In this tumour, large fibrous vessels delineate poorly defined lobules
of neoplastic cells.

(with vessels delineating vague lobules) wide range of reported mitotic counts nuclear NeuN staining in PPTID (1217,
(1187). Transitional cases also exist, de- reflects the difficulty in making the 1487,2684). ASMT-positive cells are sig-
fined as tumours in which typical pineo- diagnosis (often in a small biopsy); there nificantly more numerous in PPTIDs than
cytomatous areas are associated with a were O mitoses per 10 high-power fields in pineoblastomas (754). In pleomorphic
diffuse or lobulated pattern more consist- in 54% of cases, 1-2 in 28%, 3-6 in 15%, variants, the ganglioid cells usually ex-
ent with PPTID. PPTIDs are characterized and rarely> 6 (1187). press several neuronal markers, espe-
by moderate to high cellularity. The neo- In the PPTID group, the mean Ki-67 pro- cially NFP (701,1384).
plastic cells usually harbour round nuclei liferation index is usually significantly dif-
showing mild to moderate atypia and a ferent from those of pineocytomas and Cell of origin
so-called salt-and-pepper chromatin. pineoblastomas, ranging from 3.5% to Pineal parenchymal tumours arise from
The cytoplasm of cells is more easily 16.1% !91,1105,2122,2835,2868). pinealocytes or their precursor cells, and
distinguishable than in pineoblastoma. the close kinship among pineocytoma,
A pleomorphic cytological variant may lmmunophenotype PPTID, and pineoblastoma is evidenced
be encountered in PPTIDs as well as in lmmunohistochemistry shows synapto- by several shared clinical, morphologi-
pineocytomas (701,1384). This variant is physin positivity (91,754,1105,1187). Vari- cal, and genetic features (698,1187,1674,
characterized by bizarre ganglioid cells able labelling is also seen with antibod- 2279). The immunoexpression of cone-
with single or multiple atypical nuclei and ies to NFP and chromogranin-A (1185, rod homeobox (CRX) and acetylseroto-
abundant cytoplasm. Mitotic activity is 1187,2586,2809). GFAP and S100 pro- nin methyltransferase (ASMT) proteins
low to moderate. tein are usually expressed in astrocytic in PPTID is an additional indication that
interstitial cells (91,1187). Ganglion cells PPTIDs are biologically linked to pinealo-
Proltferation in pleomorphic variants may also ex- cytes (754,2241). CRX is a transcription
PPTID is a potentially aggressive neo- press S100 protein (1187). Like in other factor involved in the development and
plasm. In a large published series, the pineal parenchymal tumours, there is no differentiation of pineal cell lineage, and

- ..
Fig. 7.10 Pineal parenchymal tumour of intermediate differentiation. A Diffuse pineal parenchymal tumour of intermediate differentiation. This tumour is composed of round cells with
a conspicuous cytoplasm and round to oval nuclei with delicate chromatin. B Pleomorphic cells in a low-grade pineal parenchymal tumour of intermediate differentiation.

174 Tumours of the pineal region


Fig. 7.11 Pineal parenchymal tumour of intermediate differentiation.
expression of chromogranin-A.

ASMT is a critical enzyme for the synthe- Prognosis and predictive factors spinal dissemination (28%) (677). The
sis of melatonin (a hormone produced by Compared with pineoblastomas, PPTIDs low-grade and high-grade prognostic
the pineal gland). are more likely to present with local- groups also showed a significantly differ-
ized disease, and they have a better ent mean Ki-67 proliferation index (5.2%
Genetic profile prognosis, with a median overall survival vs 11.2%) {702). Although an associa-
By comparative genomic hybridization, of 165 months (vs 77 months for pineo- tion has been found in some studies be-
frequent chromosomal changes have blastoma) and a median progression- tween NFP immunopositivity and a bet-
been identified in PPTIDs. An average of free survival of 93 months (vs 46 months ter prognosis, the use of this criterion to
3.3 gains and 2 losses has been report- for pineoblastoma) (1549). In one study, assess prognosis in pineal parenchymal
ed (2122). The most common chromo- prognosis was related to mitotic count tumours remains controversial {91,1187,
somal imbalances in PPTID are 4q gain, and to neuronal differentiation as as- 2835). In another study, PPTIDs in the
12q gain, and 22 loss. In one RT-PCR sessed by anti-NFP immunohistochem- low-risk group (defined by < 3 mitoses
analysis, the expression of four genes istry {677,1187). Low-grade PPTIDs were per 10 high-power fields and a Ki-67 pro-
(PRAME, C024, POU4F2, and HOXD13) defined as tumours showing < 6 mitoses liferation index of < 5%) had better over-
in high-grade PPTID was high, almost to per 10 high-power fields and expression all survival and progression-free survival
the levels seen in pineoblastoma, and in of NFP in numerous cells {1187). In this than did PPTIDs in the high-risk group
contrast to the low expression of these group, the 5-year overall survival rate (defined by � 3 mitoses per 10 high-
genes in pineocytoma and low-grade was 74%. Recurrences occurred in 26% power fields or a Ki-67 proliferation index
PPTID {698}. One analysis showed ex- of patients and most were mainly local of � 5%) {2835). The relevance of these
pression of EGFRvlll in a PPTID without and delayed {677). High-grade PPTIDs criteria (mitotic count, NFP immunoex-
concomitant EGFR gene amplification were defined as tumours showing < 6 pression, and Ki-67 proliferation index)
(1487). mitoses per 10 high-power fields but no requires confirmation by further studies;
or only rare expression of NFP, or show- consequently, there are currently no rec-
Genetic susceptibility ing � 6 mitoses per 10 high-power fields ommended criteria for grading PPTID
No syndromic associations or genetic and NFP expression in numerous cells (see Grading, p. 173). Transformation of
susceptibilities have been reported for {1187). In this group, the 5-year overall PPTID into pineoblastoma has been rare-
PPTID. survival rate was 39%. Risk of recur- ly reported (1050,1272).
rence was higher (56%), as was risk of

Pineal parenchymal tumour of intermediate differentiation 175


Pi neoblastoma Jouvet A.
Vasiljevic A.
Nakazato Y.
Tanaka S.

Definition are infrequent (435,1751 l Nearly all pa-


A poorly differentiated, highly cellular, tients show obstructive hydrocephalus
malignant embryonal neoplasm arising in {435,1751,2373) On T1-weighted MRI,
the pineal gland. the tumours are often hypointense to
Pineoblastoma usually occurs within the isointense, with heterogeneous contrast
first two decades of life (mean patient enhancement. They are isointense to
age: 17.8 years), with a predilection for mildly hyperintense on T2-weighted im-
children. It is histologically characterized ages (435,1751,2093,2553)
by the presence of patternless sheets
of small immature neuroepithelial cells Spread
with a high nuclear-to-cytoplasmic ratio, Pineoblastomas directly invade neigh-
hyperchromatic nuclei, and scant cyto- bouring brain structures (including the
plasm. Proliferation activity is high, with leptomeninges, third ventricle, and tee-
frequent mitoses and a Ki-67 proliferation tal plate) and tend to disseminate along
index of > 20%. SMARCB1 nuclear ex- cerebrospinal fluid pathways (240)
pression is retained, enabling distinction Fig. 7.13 Pineoblastoma. Axial contrast-enhanced T1- Craniospinal dissemination is observed
from atypical teratoid/rhabdoid tumour. weighted MRI in a 3-year-old child with large head shows in 25-33% of patients at initial diagnosis
severe obstructive hydrocephalus and a heterogeneously
lsochromosome 17q or amplification of (677,1457,2517,2651 l
enhancing pineal mass.
19q13.42 are usually not seen. Pineo-
blastomas tend to spread via cerebro- predominance, with a male-to-female ra- Macroscopy
spinal fluid pathways and often follow ag- tio of 0.71. Pineoblastomas are poorly demarcated,
gressive clinical courses. invasive masses of the pineal region.
Localization They are soft, friable, and pinkish grey
ICD-0 code 9362/3 Pineoblastomas are localized in the pin- {240,2258) Haemorrhage and/or ne-
eal region. crosis may be present, but calcification
Grading is rarely seen. Invasion of surrounding
Pineoblastoma corresponds histologi- Clinical features structures is a common finding.
cally to WHO grade IV The clinical presentation of pineoblas-
toma is similar to that of other tumours Microscopy
Epidemiology of the pineal region. The main symptoms Pineoblastomas resemble other so-called
Pineoblastomas are rare, accounting are related to elevated intracranial pres- small blue round cell or primitive neuro-
for approximately 35% of all pineal pa- sure (primarily from obstructive hydro- ectodermal tumours of the CNS and are
renchymal tumours (from 24% to 61% cephalus) and include headaches and composed of highly cellular, patternless
depending on the series) {91,1187,2279, vomiting {508,671,1457) Ocular symp- sheets of densely packed small cells.
2651,2809). They can occur at any age, toms and signs may also be observed, The cells feature somewhat irregular
but most present in the first two decades such as decreased visual acuity and Par- nuclear shapes, a high nuclear-to-cyto-
of life, with a distinct predilection for chil- inaud syndrome. The interval between plasmic ratio, hyperchromatic nuclei with
dren (mean patient age 17.8 years) {91, initial symptoms and surgery may be
1187,2279,2809). There is a slight female < 1 month (508).

Imaging
10-19 I On neuroimaging, pineoblastomas pres-
60-69 • ent as large, multilobulated masses in
50-59 - the pineal region and show frequent in-
40-49- vasion of surrounding structures, includ-
30-39-- ing the tectum, thalamus, and splenium
10-19
of the corpus callosum (435,1751,2093,
20·29 ----· _
2553). Small cystic/necrotic areas and
oedema may be observed {1343,2093,
0·9 ----------·
10 20 30 40 50 2553). On CT, pineoblastomas are usual- Fig. 7.14 Pineoblastoma. A large and haemorrhagic
Fig. 7.12 Age distribution of pineoblastoma, based on ly slightly hyperdense, with postcontrast tumour localized in the pineal region and invading the
113 published cases, both sexes. enhancement {435,1751) Calcifications third ventricle.

176 Tumours of the pineal region


B High cellularity with numerous mitotic figures.

an occasional small nucleolus, scant cy- Mixed pineocytoma-pineoblastoma pineoblastoma because of their pineal
toplasm, and indistinct cell borders. The Mixed tumours are somewhat contro- localization, primitive neuroectodermal
diffuse growth pattern is interrupted only versial neoplasms showing a biphasic tumour-like component, and highly ag-
by occasional rosettes. Pineocytomatous pattern with distinct alternating areas gressive clinical course. Historically,
rosettes are lacking, but Homer Wright resembling pineocytoma and pineoblas- pineal anlage tumours were named af-
and Flexner-Wintersteiner rosettes may toma. Most importantly, areas resembling ter their shared histological features with
be seen. Flexner-Wintersteiner rosettes pineocytoma must be distinguished from melanotic neuroectodermal tumour of
indicate retinoblastic differentiation, as overrun normal parenchyma {997,1638, infancy (or retinal anlage tumour, a be-
do highly distinctive but infrequently oc- 1811,2279} nign tumour typically located in the max-
curring fleurettes. Mitotic activity varies, illa with local aggressiveness). Despite
but is generally high, and necrosis is Pineal an/age tumours shared features with pineoblastomas,
common {997,1187,1674,2279}. Pineal anlage tumours are extremely rare pineal anlage tumours have a distinct
neoplasms of the pineal region. They are morphology. They are characterized by
often considered a peculiar variant of a combination of neuroectodermal and

Pineoblastoma 177
Fig. 7.17 Pineal anlage tumour. A Striated muscle cells. B Tubular structures composed of epithelioid cells containing melanin pigment. C Ganglion cells may be seen. D Area
resembling pineoblastoma with sheets of small blue round cells.

heterologous ectomesenchymal compo- abundant euchromatin as well as hetero- and includes reactivity for neuronal,
nents. The neuroepithelial component chromatin. The cytoplasm is scant and glial, and photoreceptor markers. Posi-
is characterized by pineoblastoma-like contains polyribosomes, few profiles of tivity for synaptophysin, neuron-specific
sheets or nests of small blue round cells, rough endoplasmic reticulum, and small enolase, NFP, class Ill beta-tubulin, and
neuronal ganglionic/glial differentiation, mitochondria, as well as occasional mi- chromogranin-A may also be seen, as
and/or melanin-containing epithelioid crotubules, intermediate filaments, and may S-arrestin staining {1187,1638,1811,
cells. The ectomesenchymal component lysosomes {1587,1674,1811) Dense-core 1933,2809) Reactivity for GFAP should
contains rhabdomyoblasts, striated mus- granules are rarely seen in the cell body prompt the exclusion of entrapped reac-
cle, and/or cartilaginous islands {29,177, {1587,1674). Cell processes, which are tive astrocytes. SMARCB1 is consistently
2288). Given these distinctive character- poorly formed and short, may contain mi- expressed in pineoblastomas {1669,
istics, it is likely that pineal anlage tumour crotubules as well as scant dense-core 2497).
constitutes a separate entity. granules {1587). Bulbous endings are
not seen {1674). Junctional complexes of Cell of origin
Proliferation zonula adherens and zonula occludens Pineoblastomas share morphological
The mean Ki-67 proliferation index in type may be present between cells and and immunohistochemical features with
pineoblastoma ranges from 23.5% to processes {1185,1587,1674,1811) Syn- cells of the developing human pineal
50.1% {91,702,754,2122) apses are absent {1811) Cilia with a gland and retina. Evidence of this ontoge-
9 + 0 microtubular pattern are occasion- netic concept includes the expression of
Electron microscopy ally seen {1587) Rarely, cells radially ar- ASMT, CRX, S-arrestin, and rhodopsin in
Characterized by a relative lack of sig- ranged around a small central lumen are pineal parenchymal tumours {754,1638,
nificant differentiation, the fine structure encountered {1811). 2241) and the occasional association
of pineoblastoma is similar to that of between bilateral retinoblastoma and
any poorly differentiated neuroectoder- /mmunophenotype pineoblastoma (a condition called trilat-
mal neoplasm. The cells are round to The immunophenotype of pineoblasto- eral retinoblastoma syndrome) {544). The
oval, with slightly irregular nuclei and mas is similar to that of pineocytomas occasional progression from lower-grade

178 Tumours of the pineal region


pineal parenchymal tumours to pineo- chromosomes 9, 13, and 16 (286,1668, Prognosis and predictive factors
blastomas also supports this concept 2122,2206} No aberrations of the TP53 Pineoblastoma is the most aggressive
(1050,1272} or CDKN1A genes have been detected of the pineal parenchymal tumours, as
(2584,2585}. Pineoblastomas are known evidenced by the occurrence of cranio-
Genetic profile to occur in patients with RB1 gene ab- spinal seeding and (rarely) extracranial
conventional cytogenetic studies in normalities, and the prognosis of such metastasis (677,997,1114,2279}. Overall
pineoblastomas have shown various cases is significantly worse than that of survival has been short; older studies
. numerical and structural abnormalities, sporadic cases (1991); however, the sta- reported median values ranging from
but non-random aberration has not been tus of RB1 in sporadic pineoblastomas 1.3 to 2.5 years (412,677,1638}. but re-
consistently described (286}. lsochro- is not clearly defined. In one microarray cent studies have reported improved
mosome i?q, a common chromosomal analysis of pineal parenchymal tumours, median overall survival times reaching
abnormality in medulloblastoma, has four genes (PRAME, CD24, POU4F2, and 4.1-8.7 years (671,1128}. Similarly, re-
been observed in a few karyotypes of HOXD13) were significantly upregulated ported 5-year overall survival rates vary
pineoblastomas, but was absent in mi- in pineoblastomas and high-grade pineal from 10% to 81%. Disseminated disease
croarray-based comparative genomic parenchymal tumours of intermediate dif- at the time of diagnosis (as determined
hybridization studies (1668,2122,2206}. ferentiation (698}. by cerebrospinal fluid examination and
On comparative genomic hybridiza- MRI of the spine) (412,671,2517}. young
tion analysis, the genomic imbalance in Genetic susceptibility patient age (610,1005,2517}. and par-
pineoblastomas is less than has been Pineoblastomas can occur in patients tial surgical resection (1128,1457,2517}
� observed in CNS primitive neuroectoder- with familial (bilateral) retinoblastoma, a are negative prognostic predictors. Ra-
i mal tumours, with an average of 5.6 chro- condition called trilateral retinoblastoma diotherapy treatment seems to positively
mosomal changes in one study and with syndrome (544}. and these tumours have affect prognosis (671,1457,2279}. The
the changes observed being unrelated also been reported in patients with famil- 5-year survival of patients with trilateral
to lower-grade tumours of the pineal re- ial adenomatous polyposis (772,1086}. retinoblastoma syndrome has significant-
gion (1668,2122}. No amplicon of the Pineoblastomas can also occur in pa- ly increased in the past decade (from 6%
J9qi3.42 region has been detected to tients with DICER1 germline mutations to 44%), probably due to better chemo-
date in pineal neoplasms diagnosed as (545,2210}; in these cases, the biallelic therapy regimens and earlier detection of
pineoblastomas. Conventional cytoge- inactivation of DICER1, mainly by allelic pineal disease (544f.
netic studies and comparative genomic loss of the wild-type allele, may play
hybridization analyses have shown fre- a role in pineoblastoma pathogenesis
quent structural alterations of chromo- (545,2210}.
some 1 and losses involving all or part of

Pineoblastoma 179
Papillary tumour of the pineal region Jouvet A.
Vasiljevic A.
Nakazato Y.
Paulus W.
Hasselblatt M.

Definition Clinical features


A neuroepithelial tumour localized in the Symptoms are non-specific, may be of
pineal region and characterized by a short duration, and include headache
combination of papillary and solid areas, due to obstructive hydrocephalus and
with epithelial-like cells and immunoreac- Parinaud syndrome {676).
tivity for cytokeratins (especially CK18).
Papillary tumour of the pineal region af- Imaging
fects children and adults (mean patient On neuroimaging, papillary tumours of
age: 35 years) and presents as a large, the pineal region present as well-circum-
well-circumscribed mass, often with T1- scribed heterogeneous masses com-
hyperintensity. The tumours are associ- posed of cystic and solid portions and
ated with frequent recurrence (occurring centred by the posterior commissure or
in 58% of cases by 5 years), but spinal the pineal region. Aqueductal obstruc-
dissemination is rare. Overall survival is tion with hydrocephalus is a frequent
73% at 5 years and 71.6% at 10 years. associated finding {65,2012,2114,2248}.
The tumours may demonstrate intrinsic Fig. 7.19 MRI of papillary tumour of the pineal region,
located in the posterior part of the third ventricle, showing
ICD-0 code 9395/3 T1-hyperintensity {392,408,2248} In the
contrast enhancement.
absence of calcification, haemorrhage,
Grading melanin, or fat on imaging, this T1-hy- eosinophilic columnar cells. In cellular ar-
The biological behaviour of papillary tu- perintensity may be related to secretory eas, cells with a somewhat clear or vacu-
mour of the pineal region is variable and material with high protein and glycopro- olated cytoplasm (and occasionally with
may correspond to WHO grades II or Ill, tein content {408). However, others have an eosinophilic periodic acid-Schiff-
but definite histological grading criteria found this feature to be absent {65,1280). positive cytoplasmic mass) may also be
remain to be defined. Postcontrast enhancement is usually seen. The nuclei are round to oval, with
heterogeneous. stippled chromatin; pleomorphic nuclei
Epidemiology may be present. The mitotic count ranges
Because these tumours are so rare, in- Spread from O to 13 mitoses per 10 high-power
cidence data are not available. The Papillary tumour of the pineal region is fields {984) Necrotic foci may be seen.
181 papillary tumours of the pineal re- characterized by frequent local recur- Vessels are hyalinized and often have a
gion reported in the literature include rence (with at least one relapse in 57% of pseudoangiomatous morphology, with
examples in both children and adults patients in one series). Spinal dissemina- multiple lumina {696). Microvascular
{697,699,872,957,984}. Reported patient tion is reported rarely (in 7% of cases in proliferation is usually absent. When the
ages range from 1 to 71 years, with a me- one series) {676,1280). pineal gland is present, there is a clear
dian of 35 years. No sex predilection has demarcation between the tumour and the
been shown, with a male-to-female ratio Macroscopy adjacent gland.
of 1.06:1. Papillary tumours of the pineal region
present as relatively large (20-54 mm) Pro!tferation
Localization {696). well-circumscribed tumours indis- Mitotic activity is moderate in most cases
By definition, papillary tumours of the pin- tinguishable grossly from pineocytoma. (872,1184). with a median of 2 mitoses
eal region arise in the pineal region. per 10 high-power fields reported in the
Microscopy largest published series {696). Increased
Papillary tumour of the pineal region is mitotic activity (:?: 3 mitoses per 10 high-
70-79 - an epithelial-looking tumour with papil- power fields) was observed in 48% of tu-
60-69 --- lary features and more densely cellular mours in one series {984) and in 33% of
50-59 ------- areas, often exhibiting ependymal-like tumours in another study {696). Marked
40-49 ----------- differentiation (true rosettes and tubes). mitotic activity (:?: 10 mitoses per 10 high-
30-39 ----------- Papillary tumour of the pineal region may power fields) was reported in only 9%
20-29 -----------
exhibit a prominent papillary architecture of papillary tumours of the pineal region
10-19 ----------
0-9 - or, conversely, a more solid morphology {696,985). In one series of 33 papillary
10 20 30 40 in which papillae are barely recognizable tumours of the pineal region, the Ki-67
Fig. 7.18 Age distribution of papillary tumours of the {696,984). In papillary areas, the vessels proliferation index ranged from 1.0%
pineal region, based on 181 published cases, both sexes. are covered by layers of large, pale to to 29.7% (median: 7.5%) {696). In two

180 Tumours of the pineal region


.
Fig. 7.20 Papillary tumour of the pineal region. A The vascular axes of neoplastic papillae often harbour multiple capillaries, resulting in a pseudoangiomatous appearance.
B In some tumours, bizarre pleomorphic cells are observed; this nuclear atypia is more dystrophic in nature than related to anaplasia. C Neoplastic cells detached from the papillary
vascularized core, leading to an apparent clear perivascular space. Note the extensive necrosis.

other series, increased proliferative activ- present in some cells (512,696,1184l. that papillary tumours of the pineal region
ity (defined as a Ki-67 proliferation index may originate from remnants of the spe-
of z 10%) was observed in 39% and 40% lmmunophenotype cialized ependymal cells of the subcom-
of cases, respectively (696,984l High The most distinctive immunohistochemi- missural organ (1184). Further evidence
proliferative activity has been linked to cal feature of papillary tumours of the for a putative origin from specialized
younger patient age (699l. pineal region is their reactivity for kerat- ependymocytes of the subcommissural
ins (KL1. AE1/AE3, CAMS 2, and CK18), organ comes from the high levels of ex-
Electron microscopy particularly in papillary structures. GFAP pression in papillary tumour of the pineal
On electron microscopy, papillary tu- expression is less common than in region of genes expressed in the sub-
mours of the pineal region show com- ependymomas. Papillary tumours of the commissural organ, including ZFHX4,
bined ependymal, secretory, and neu- pineal region also stain for vimentin, S100 SPOEF, RFX3, TTR, and CALCA (698,
roendocrine features. Papillary tumours protein, neuron-specific enolase, MAP2, 986l. Papillary tumours of the pineal re-
of the pineal region are usually com- NCAM1. and transthyretin {957,2344). gion have a claudin expression profile
posed of alternating clear and dark epi- Focal membrane or dot-like EMA staining similar to that of the subcommissural or-
thelioid cells adjoined at the apical region as encountered in ependymomas is rare gan in human fetuses (i.e. claudin-1 and
by well-formed intercellular junctional {957,1184,1383). NFP immunolabelling is -3 positivity and claudin-2 negativity) and
complexes. Apical poles of cells show never seen, whereas the neuroendocrine rats (i.e. claudin-3 positivity) (696,2474l.
numerous microvilli with occasional cilia. markers synaptophysin and chromogra-
The nuclei are oval, indented, or irregu- nin-A are sometimes weakly and focally Genetic profile
lar and are frequently found at one pole expressed (1184l. Most papillary tumours In two comparative genomic hybridiza-
of the cell. lnterdigitated ependymal-like of the pineal region are characterized by tion studies, recurrent chromosomal
processes are seen at the basal pole of the absence of staining for membranous imbalances included losses of chro-
some cells and are bordered by a base- KIR7.1, cytoplasmic stanniocalcin-1, cad- mosome 10 (in 7 of 8 cases) as well as
ment membrane. Zonation of organelles herin-1, and claudin-2, markers that are gains on chromosomes 4 (in 6 of 8 cas-
may be observed. The cytoplasm is usu- frequently present in choroid plexus tu- es) and 9 (in 7 of 8 cases) (902,957). On
ally rich in organelles, which include nu- mours (696,699,957). high-resolution copy number analysis,
merous clear and coated vesicles, mito- losses affecting chromosome 10 were
chondria, and rare dense-core vesicles. Cell of origin observed in all 5 cases examined; losses
Rough endoplasmic reticulum is abun- lmmunohistochemical findings (i.e. cy- of chromosomes 3, 14, and 22 and gains
dant, and dilated cisternae filled with a tokeratin positivity) and ultrastructural of whole chromosomes 8, 9, and 12 were
granular secretory product may be seen. demonstration of ependymal, secretory, observed in > 1 case. These findings
Perinuclear intermediate filaments are and neuroendocrine organelles suggest were confirmed in a recent study that

- . .' '!! I :...::.�..:.l'..i!�lt!f.1...;jlj!lil!Lf.:


Fig. 7.21 Papillary tumour of the pineal region. A CK18 is expressed in the epithelial-like neoplastic cells. lmmunoexpression may be diffuse (as in this example) or may predominate
in perivascular areas. B Expression of CK18 predominates in perivascular areas. C NCAM1 (also called CD56) is usually strongly expressed, whereas only faint and focal
immunopositivity is reported in choroid plexus tumours.

Papillary tumour of the pineal region 181


also showed distinct DNA methylation progression occurred in 72% of cases, 8-96 months) versus 68 months (range:
profiles differentiating papillary tumours and the 5-year estimates of overall and 66-70 months) for those whose tumours
of the pineal region from ependymomas progression-free survival were 73% and showed < 3 mitoses. Similarly, increased
(986}. Genetic alterations of PTEN (one 27%, respectively. Incomplete resec- proliferative activity was associated with
homozygous deletion and two exon 7 tion tended to be associated with de- shorter progression-free survival; pa-
point mutations) have been encoun- creased survival and with recurrence tients whose tumours had a Ki-67 prolif-
tered (872}. No expression of the V600E- {699). In an updated retrospective series eration index ot z 10% had a median pro-
mutant BRAF protein has been detected of 44 patients, only gross total resection gression-free survival time of 29 months
by immunohistochemistry in these tu- and younger patient age were associ- (range: 0-64 months) versus 67 months
mours (460}. ated with overall survival; radiotherapy (range: 44-90 months) for those whose
and chemotherapy had no significant tumours had a Ki-67 proliferation index
Genetic susceptibility impact {676). Another study, of 19 pa- of < 10%. The tumours of the 3 patients
No syndromic association or evidence tients, also found no significant effect who succumbed to disease all showed
of genetic susceptibility has been of clinical factors on overall survival or increased mitotic and proliferative activ-
documented. progression-free survival {984). In that ity {984). The usefulness of mitotic count
series, increased mitotic activity was or proliferation index in defining a more
Prognosis and predictive factors significantly associated with shorter aggressive subset of papillary tumours of
The clinical course of papillary tumours progression-free survival; patients whose the pineal region requires confirmation in
of the pineal region is often complicated tumours showed > 3 mitoses per 10 high- further studies. Recurrences might show
by local recurrences. In a retrospective power fields had a median progression- higher proliferative activity {902,1447}
multicentre study of 31 patients, tumour free survival time of 52 months (range:

182 Tumours of the pineal region


CHAPTER 8

Embryonal tumours
Medullob/astomas, genetically defined
Medulloblastoma, WNT-activated
Medulloblastoma, SHH-activated
Medulloblastoma, non-WNT/non-SHH
Medulloblastomas, histologically defined
Medulloblastoma, classic
Desmoplastic/nodular medulloblastoma
Medulloblastoma with extensive nodularity
Large cell I anaplastic medulloblastoma

Embryonal tumour with multilayered rosettes, C19MC-altered


Medulloepithelioma
CNS neuroblastoma
CNS ganglioneuroblastoma
CNS embryonal tumour, NOS
Atypical teratoid/rhabdoid tumour
.,

Medulloblastoma Ellison OW
Eberhart C.G.
Pietsch T.
Pfister S.

Introduction respective cell signalling pathways. The 2016 WHO classification of medulloblastomas
In this update of the WHO classifica- four principal groups emerged from clus- Medulloblastomas, genetically defined
tion, medulloblastomas are classified tering analyses following transcriptome,
Medulloblastoma, WNT-activated
according to molecular characteristics microRNA, and methylome profiling, and
in addition to histopathological features. there is excellent concordance across Medulloblastoma, SHH-activated and TP53-mutant
The molecular classification relates to these platforms for the assignment of Medulloblastoma, SHH-activated and
the clustering of medulloblastomas into individual tumours (142,1804} There are TPS3-wildtype
groups on the basis of transcriptome or also significant associations between the Medulloblastoma, non-WNT/non-SHH
methylome profiling and has been intro- four groups and specific genetic altera-
Medul/oblastoma, group 3
duced because of its increasing clinical tions and clinicopathological variables.
utility (1804). A histopathological classifi- In the updated WHO classification, WNT- Medul/oblastoma, group 4
cation has also been retained, due to its activated medulloblastomas (accounting Medulloblastomas, histological/y defined
clinical utility when molecular analysis is for -10% of cases) and SHH-activated Medulloblastoma, classic
limited or not feasible. medulloblastomas (-30% of cases) are
Desmoplastic/nodular medulloblastoma
Transcriptome profiling studies of medul- listed separately from non-WNT/non-
loblastomas indicate that these tumours SHH tumours, which comprise group 3 Medulloblastoma with extensive nodularity
can be separated into several distinct tumours (-20% of cases) and group 4 Large cell / anaplastic medulloblastoma
molecular clusters (2524), which by tumours (-40% of cases). Group 3 and Medul/oblastoma, NOS
consensus have been distilled into four group 4 medulloblastomas are listed as
principal groups: WNT-activated medul- provisional variants, because they are
loblastomas, SHH-activated medullo- not as well separated as WNT-activated Medulloblastoma has always been con-
blastomas, group 3 medulloblastomas, and SHH-activated medulloblastomas sidered to be an embryonal tumour of
and group 4 medulloblastomas. in molecular clustering analyses and by the cerebellum. However, WNT-acti-
Tumours in the WNT-activated and SHH- current clinical laboratory assays {448, vated medulloblastomas are thought
activated groups show activation of their 1335) to arise from cells in the dorsal brain
stem (831), although not all brain stem
Table 8.01 Medulloblastoma subtypes characterized by combined genetic and histological parameters embryonal tumours are WNT-activated
Genetic profile Histology Prognosis medu I loblastomas.
The established morphological variants of
Low-risk tumour; classic morphology found
Classic medulloblastoma (i.e. desmoplastic/nod-
in almost all WNT-activated tumours
Medulloblastoma, WNT-activated ular medulloblastoma, medulloblastoma
Large cell / anaplastic Tumour of uncertain clinicopathological with extensive nodularity, and large cell or
(very rare) significance
anaplastic medulloblastomas) have their
Classic Uncommon high-risk tumour own particular clinical associations (619,
High-risk tumour; prevalent in children 1603,1626,1627} Large cell and ana-
Medulloblastoma, SHH-activated, Large cell / anaplastic
TPS3-mutant
aged 7-17 years plastic medulloblastomas were listed as
Desmoplastic/nodular Tumour of uncertain clinicopathological separate variants in the previous version
(very rare) significance of the classification, but because nearly
Classic Standard-risk tumour all large cell tumours also demonstrate
an anaplastic component and both vari-
Tumour of uncertain clinicopathological
Large cell / anaplastic ants are associated with a poor outcome,
Medulloblastoma, SHH-activated, significance
they are commonly considered for clinical
TP53-wildtype Low-risk tumour in infants; prevalent in
Desmoplastic/nodular purposes as being in a single combined
infants and adults
category of large cell / anaplastic medul-
Extensive nodularity Low-risk tumour of infancy loblastoma (2208,2664). This association
Medulloblastoma, Classic Standard-risk tumour and its designation have been recog-
non-WNT/non-SHH, group 3 Large cell / anaplastic High-risk tumour
nized in the update of the classification.
The molecular and morphological variants
Standard-risk tumour; classic morphology
Classic of medulloblastoma listed in the new clas-
Medulloblastoma, found in almost all group 4 tumours
sification demonstrate particular relation-
non-WNT/non-SHH, group 4 Tumour of uncertain clinicopathological
Large cell/ anaplastic (rare) ships (631} All true desmoplastic/nodular
significance
medulloblastomas and medulloblastomas

184 Embryonal tumours


Table 8.02 Characteristics of genetically defined medulloblastomas
WNT· SHH-activated Non-WNT/non·SHH
activated TP53-wildtype TP53-mutant Group 3 Group 4
Infancy Infancy
predominant age(s) at
presentat_
io_n ,_
Childhood
_ Adulthood
Childhood
Childhood
I All age groups
----+
Male-to-female ratio 1:2 1:1 1:1 2:1 3:1
-
Predominant Classic
Classic Desmoplastic/nodular Large cell / anaplastic Classic
pathological variant(s) Large cell / anaplastic
1---

Frequent copy number


I PTCH1 deletion
MYCN amplification
MYC amplification MYCN amplification
Monosomy 6 GL/2 amplification
alterations 10q loss lsodicentric 17q lsodicentric 17 q
17p loss I
I
PTCH1 mutation
CTNNB1 mutation PVT1-MYC KOM6A
Frequent genetic SMO mutation (adults) TPS3 mutation
DDX3X mutation GF/1/GF/1 B structural GF/1/GF/1 B structural
alterations SUFU mutation (infants) variants
TP53 mutation variants
TERT promoter mutation
Genes with germline
mutation
APC
7i:J I TP53 �

I Cerebellar granule neuron cell precursors of the external


+--C-0133+/lineage-
- --
neural stem cells
Lower rhombic lip granule cell layer and cochlear nucleus
Proposed cell of origin Unknown
I
progenitor cells (Cerebellar granule neuron
(Neural stem cells of the subventricular zone)' cell precursors of the ex-
ternal granule cell layer)"
*Parentheses indicate a less likely origin.

with extensive nodularity align with the into an integrated diagnosis that brings its origins in cells of the dorsal brain stem
SHH-activated molecular group. Virtually together molecular group, histopatholog- that are derived from the lower rhombic
all WNT-activated tumours have classic ical variant, and specific genetic altera- lip. Medulloblastoma variants show a
morphology. Most large cell I anaplastic tion to enhance the level of diagnostic broad range of morphological features,
tumours belong either to the SHH-activat- precision. including neurocytic and ganglionic dif-
ed group or to group 3. lmmunohistochemical assays that work ferentiation, and distinct biological be-
on formalin-fixed paraffin-embedded tis- haviours. In making a diagnosis medul-
Integrated diagnosis sue and are readily available worldwide loblastoma, it is important to exclude
This updated classification is intended to can be used to discern some genetically histopathologically similar entities that
encourage an integrated approach to di- defined variants of medulloblastoma and arise in the posterior Iossa, such as high-
agnosis {1535). When molecular analysis genetic alterations with clinical utility grade small cell gliomas, embryonal
is feasible, combined data on both mo- {1240). However, the updated classifica- tumour with multilayered rosettes, and
lecular group and morphological variant tion does not make specific recommen- atypical teratoid/rhabdoid tumours.
provide optimal prognostic and predic- dations regarding the merits of the vari-
tive information. This approach is further ous methods for determining molecular Grading
enhanced when specific genetic data groups or genetic alterations. Irrespective of their histological or genetic
are integrated into the diagnosis, e.g. by characterization, medulloblastomas cor-
the inclusion of TP53 gene status in the Definition respond histologically to WHO grade IV
classification. An embryonal neuroepithelial tumour
SHH-activated medulloblastomas are arising in the cerebellum or dorsal brain 100
a heterogeneous group; a tumour with stem, presenting mainly in childhood
TP53 mutation and large cell I anaplastic and consisting of densely packed small ; 80

morphology has an abysmal progno- round undifferentiated cells with mild to � 701---1---t---t--t--t--t--

sis, in contrast to SHH-activated and moderate nuclear pleomorphism and a o 60 !----+--+-


TP53-wildtype medulloblastomas with high mitotic count. ! so
extensive nodularity, which have a good Medulloblastoma is the most common
£.
°S
40
30
clinical outcome if treated appropriately CNS embryonal tumour and the most E
:, 20
u
12870). common malignant tumour of childhood. 10

Some molecular genetic alterations cur- It is now classified into molecular (i.e.
10 20 30 40 50 60
rently used in the risk stratification of me- genetic) variants as well as morphologi- Age at diagnosis
dulloblastomas, such as MYC amplifica- cal variants, all with clinical utility. Most Fig. 8.01 Cumulative age distribution of medulloblastoma
tion, are not included in the classification, medulloblastomas arise in the cerebel- (both sexes), based on 831 cases (2008-2015). Data
but could nevertheless be incorporated lum, but the WNT-activated variant has from the Brain Tumor Reference Center, Bonn.

Medulloblastoma 185
Fig. 8.02 Medulloblastoma. A Sagittal section. The tumour occupies mostly the lower part of the cerebellum. B Typical gross postmortem appearance of a medulloblastoma in the
cerebellar midline, occupying the cerebellar vermis. C Diffuse CSF seeding by a medulloblastoma into the basal cisterns and meninges.

B ....
'i'!' .
Fig. 8.03 A Numerous medulloblastoma metastases of various sizes on the falx cerebri and the inner surface of the Fig. 8.04 Infiltration by a cerebellar medulloblastoma of
dura mater covering the left cerebral hemisphere. Some smaller dural metastases are present on the contralateral side. the subarachnoid space. Note the clusters of tumour cells
B Multiple nodules in the cauda equina of the spinal cord representing CSF drop metastases of a medulloblastoma. in the molecular layer, particularly in the subpial region.

Epidemiology in incidence at 3 and 7 years of age


{2138). Of all patients with medulloblas-
Medulloblastoma, NOS Incidence toma, 77% are aged < 19 years {673).
Medulloblastoma is the most common The tumour has an overall male-to-fe-
The diagnosis of medulloblastoma, NOS, CNS embryonal tumour of childhood. male ratio of 1. 7: 1. Among patients aged
is appropriate when an embryonal neural Of all paediatric brain tumours, medullo- > 3 years, the male-to-female ratio is 2:1,
tumour is located in the fourth ventricle or blastoma is second in frequency only to but the incidence rates among boys and
cerebellum and the nature of biopsied tis- pilocytic astrocytoma, and accounts for girls aqed s 3 years are equal {511,899).
sue prevents classification of the tumour 25% of all intracranial neoplasms {673). The various molecular groups and histo-
into one of the genetically or histologi- The annual overall incidence of medullo- pathological variants of medulloblastoma
cally defined categories of medulloblas- blastoma is 1.8 cases per 1 million popu- have different age distributions {631,
toma. This situation usually arises when lation, whereas the annual childhood in- 1334,2524)
there is uncertainty about a tumour's cidence is 6 cases per 1 million children;
architectural and cytological features as these incidence rates have not changed Localization
a result of insufficient tissue sampling or over time {1896l As is the case with other Medulloblastomas grow into the fourth
the presence of tissue artefacts. For the high-grade brain tumours, the incidence ventricle or are located in the cerebel-
diagnosis of medulloblastoma, NOS, it is of medulloblastoma differs across ethnic- lar parenchyma {213). Some cerebel-
important to exclude histopathologically ities. In the USA, overall annual incidence lar tumours can be laterally located in a
similar entities, such as high-grade small is highest among White non-Hispanics hemisphere.
cell gliomas, embryonal tumour with mul- (2.2 cases per 1 million population), fol-
tilayered rosettes, and atypical teratoid/ lowed by Hispanics (21 per 1 million) Clinical features
rhabdoid tumours. and African Americans (1.5 per 1 mil- Medulloblastomas growing in the fourth
lion) [http://www.cbtrus.org/2011-NPCR- ventricle cause increased intracranial
ICD-0 code 9470/3 SEER/WEB-0407-Report-3-3-2011.pdf]. pressure by exerting mass effect and
As many as a quarter of all medulloblas- blocking cerebrospinal fluid pathways.
tomas occur in adults, but < 1% of adult Therefore, most patients present with a
intracranial tumours are medulloblasto- short history of raised intracranial pres·
mas {1645). sure: headaches that have increased in
frequency and severity, frequent nausea
Age and sex distnbution upon waking, and bouts of vomiting. Cer-
The median patient age at diagnosis of ebellar ataxia is common. Symptoms and
medulloblastoma is 9 years, with peaks signs relating to compression of cranial

186 Embryonal tumours


nerves or long tracts passing through the
brain stem are uncommon.

Spread
Like other embryonal tumours, medul-
loblastoma has a propensity to spread
through cerebrospinal fluid pathways to
seed the neuraxis with metastatic tumour
deposits. Rarely, it spreads to organ
systems outside the CNS, particularly
to bones and the lymphatic system. Re-
ports of metastasis to the peritoneum im-
plicate ventriculoperitoneal shunts.

Macroscopy
Most medulloblastomas arise in the re-
gion of the cerebellar vermis, as pink
or grey often friable masses that fill the
fourth ventricle. Medulloblastomas lo-
cated in the cerebellar hemispheres tend
to be firm and more circumscribed, and
generally correspond to the desmoplas- Fig. 8.05 Medulloblastoma with myogenic or melanotic differentiation. A Striated muscle fibres. B Anti-fast myosin
tic/nodular variant with SHH pathway immunostaining of highly differentiated, striated myogenic cells. C Biphasic pattern of small undifferentiated embryonal
activation. Small foci of necrosis can be cells and large rhabdomyoblasts immunostaining for myoglobin. D Melanotic cells commonly appear as tubular
epithelial structures, which are immunopositive for HMB45 and cytokeratins.
grossly evident, but extensive necrosis is
rare. In disseminated medulloblastoma,
discrete tumour nodules are often found setting of a classic or large cell I anaplas- express no neural antigens, the expres-
in the craniospinal leptomeninges or tic tumour and are no longer considered sion of markers of neuronal differen-
cerebrospinal fluid pathways. distinct histopathological variants. These tiation is common. lmmunoreactivity for
are the rare (accounting for < 1% of synaptophysin, class Ill beta-tubulin, or
Microscopy cases) medulloblastoma with myogenic NeuN is demonstrated at least focally in
Several morphological variants of medul- differentiation (previously called medullo- most medulloblastomas. Homer Wright
loblastoma are recognized, alongside myoblastoma) and medulloblastoma rosettes and nodules of neurocytic dif-
the classic tumour: desmoplastic/nodular with melanotic differentiation (previously ferentiation are immunopositive for these
medulloblastoma, medulloblastoma with called melanocytic medulloblastoma). markers. In contrast, expression of NFPs
extensive nodularity, and large cell I ana- Although these tumours are very rare, it is rare. GFAP-immunopositive cells are
plastic medulloblastoma. Their specific is not uncommon for their phenotypes to often found among the undifferentiated
microscopic architectural and cytological occur together. embryonal cells of a medulloblastoma;
features are described in the correspond- In addition to a conventional embryonal however, they generally show the typical
ing sections of this volume. A dominant element, medulloblastoma with myogen- spider-like appearance of reactive as-
population of undifferentiated cells with a ic differentiation contains a variable num- trocytes and tend to be more abundant
high nuclear-to-cytoplasmic ratio and mi- ber and distribution of spindle-shaped near blood vessels. These cells are usu-
totic figures is a common feature, justify- rhabdomyoblastic cells and sometimes ally considered to be entrapped astro-
ing the designation "embryonal", but it is large cells with abundant eosinophilic cytes, although the observation of similar
important to consider other entities in the cytoplasm {2211,2383). Occasionally, cells in extracerebral metastatic deposits
differential diagnosis. High-grade small elongated differentiated strap cells, with raises the possibility that at least some
cell gliomas and some ependymomas the cross-striations of skeletal muscle, are well-differentiated neoplastic astro-
have an embryonal-like cytology, and are evident. cytes. Cells showing GFAP immunore-
elements of the embryonal tumour with Medulloblastoma with melanotic differen- activity and the cytological features of
multilayered rosettes or atypical teratoid/ tiation contains a small number of mel- bona fide neoplasia can be observed
rhabdoid tumour can be identical to me- anin-producing cells, which sometimes in approximately 10% of medulloblasto-
dulloblastoma. Any of these entities can form clumps {730,1965} These may ap- mas. In medulloblastoma with myogenic
be confused with medulloblastoma, es- pear entirely undifferentiated (like other differentiation, cells demonstrating myo-
pecially in small biopsies, so determining embryonal cells) or have an epithelioid genic differentiation are immunopositive
a tumour's immunophenotype or genetic phenotype. Epithelioid melanin-produc- for desmin or myogenin, but not alpha-
profile is an important part of working ing cells may form tubules, papillae, or SMA. In medulloblastomas with mel-
through the differential diagnosis. cell clusters. anotic differentiation. melanin-producing
Two distinctive morphological variants of cells express HMB45 or melan-A, and
medulloblastoma are described in this lmmunophenotype the clumps of epithelioid cells associated
section, because they may occur in the Although a few medulloblastomas with focal melanin production generally

Medulloblastoma 187
show immunoreactivity for cytokeratins. Genetic susceptibility syndrome l249A). and Nijmegen break-
Nuclear SMARC81 and SMARCA4 ex- Medulloblastomas occur in the setting age syndrome l1058A).
pression is retained in all medulloblasto- of several inherited cancer syndromes: Genetic susceptibility to medulloblas-
ma variants; the loss of expression of one naevoid basal cell carcinoma syndrome toma has been documented in rnonozv,
of these SWI/SNF complex proteins in the (also called Gorlin syndrome; seep. 319), gotic twins !434). siblings, and relatives
context of an embryonal tumour is char- Li-Fraumeni syndrome; see p. 310), mis- !1065,2666). Association with other brain
acteristic of atypical teratoid/rhabdoid match repair cancer syndrome (Turcot tumours !673) and Wilms tumour !1846,
tumour. syndrome, p. 317), Rubinstein-Taybi 2062) has also been reported.

Medulloblastomas, genetically defined

Medulloblastoma, Ellison D.W. Pietsch T.


WNT-activated Giangaspero F. Wiestler O.D.
Eberhart C.G. Pfister S.
Haapasalo H.

Definition immunohistochemistry or gene expres-


An embryonal tumour of the cerebellum I sion profiling demonstrate monosomy 6
fourth ventricle composed of small uni- and/or harbour a CTNNB1 mutation.
form cells with round or oval nuclei that WNT-activated medulloblastoma is
demonstrate activation of the WNT sig- thought to originate from the dorsal brain
nalling pathway. stem to fill the fourth ventricle. WNT-acti-
Nearly all WNT-activated medulloblasto- vated tumours account for approximately
mas are classic tumours. WNT-activated 10% of all medulloblastomas. Most cas-
tumours with an anaplastic morphol- es present in children aged between 7
ogy have been reported, but are very and 14 years, but they can also occur in
rare. The WNT pathway activation that young adults. This variant has an excel- Fig. 8.06 WNT-activated medulloblastomas are usually
is characteristic of this medulloblastoma lent prognosis with standard therapeutic centred on the foramen of Luschka, but tumours
can be demonstrated by the accumu- approaches. Besides CTNNB1, genes frequently spread along the lateral wall of the fourth
lation of beta-catenin immunoreactiv- that are recurrently mutated in WNT- ventricle and appear intraventricular.
ity in tumour cell nuclei, but an optimal activated medulloblastomas include
evaluation combines this method with TP53, SMARCA4, and ODX3X. Grading
detection of monosomy 6 or CTNNB1 WNT-activated medulloblastoma corre-
mutation; approximately 85% of WNT- ICD-0 code 9475/3 sponds histologically to WHO grade IV.
activated medulloblastomas defined by However, this genetically defined variant

J,, - ,A •
Fig. 8.07 A Non-WNT medulloblastoma often shows beta-eaten in immunoreactivity restricted to the plasma membrane and cytoplasm. B WNT-activated medulloblastoma. Nuclear
immunoreactivity for beta-catenin indicates activation of the WNT pathway. C WNT-activated medulloblastoma. lmmunoreactivity for beta-catenin manifests as groups of positive
nuclei in some WNT-activated medulloblastomas.

188 Embryonal tumours


has an excellent prognosis with standard been reported, but are very rare (631 ). that approximately 90% of WNT-activat-
therapeutic approaches. Desmoplastic/nodular medulloblastomas ed medulloblastomas contained somatic
do not occur in this group. mutations in exon 3 of CTNNB1 (1804).
Epidemiology Other recurrently mutated genes in WNT-
WNT-activated tumours account for lmmunophenotype activated medulloblastomas include
about 10% of all medulloblastomas (634, WNT-activated medulloblastomas have DDX3X (in 50% of cases), SMARCA4 (in
777,1972). They typically occur in older the same neural protein irnrnunopro- 26.3%), KMT20 (in 12.5%), and TP53 (in
children and also account for a signifi- file as other classic medulloblastomas. 12 5%) In addition to CTNNB1 mutation,
cant proportion (-15%) of adult medul- Their growth fraction as estimated by monosomy 6 has long been established
loblastomas, but hardly ever occur in the Ki-67 proliferation index is also the as a hallmark genetic aberration in WNT-
infants. same. Nearly all medulloblastomas show activated medulloblastomas, occurring in
some cytoplasmic immunoreactivity for approximately 85% of cases (472,1334).
.. Localization beta-catenin, but WNT-activated tumours
, Some reports indicate that WNT-activated show nuclear beta-catenin immunoreac- Genetic susceptibility
medulloblastomas are all located in the tivity in most cells, although staining can There is a rare association between APC
cerebellar midline, with or without close be patchy in about one quarter of cases germline mutation and WNT-activated
contact to the brain stem (831,2534). and (630,631). medulloblastoma {937,1057).
one report suggests that these tumours
are more precisely localized to the cer- Cell of origin Prognosis and predictive factors
ebellar peduncle or cerebellopontine an- WNT-activated medulloblastomas are The prognosis of patients with WNT-
gle (1936). thought to arise from cells in the dorsal activated medulloblastoma is excellent;
brain stem that originate from the lower with current surgical approaches and ad-
Imaging rhombic lip {831). The DNA methylation juvant therapy regimens, overall survival
MRI of WNT-activated medulloblastomas fingerprint of these tumours, which might is close to 100% {634,2549). Unlike in
shows tumours located in the cerebel- be the best evidence for cell of origin in SHH-activated medulloblastomas, TP53
lar midline I cerebellopontine angle, with human tissues, indicates that WNT-acti- mutations in WNT-activated medulloblas-
many in close contact to the brain stem vated medulloblastoma has a profile dis- tomas (which are all somatic and most
(831). tinct from those of other medulloblastoma commonly heterozygous) do not confer
subgroups (1049,1972,2345) a worse prognosis (2870). Predictive bio-
Microscopy markers have not yet been established
Nearly all WNT-activated medulloblas- Genetic profile within the WNT-activated molecular
tomas have a classic morphology; ana- A recent meta-analysis of all large next- group.
plastic WNT-activated tumours have generation sequencing datasets showed

Medulloblastoma, WNT-activated 189


Medulloblastoma, Eberhart C.G. Pietsch T. be age-dependent. A third study found
SHH-activated Giangaspero F. Wiestler 0.0. that in older children and young adults,
Ellison D.W. Pfister S. SHH-activated medulloblastomas grow
Haapasalo H. predominantly in the rostral cerebellar
hemispheres, whereas in infants they
more frequently involve the vermis
Medulloblastoma, a classic or large cell I anaplastic mor- !2716). Specific data on the localization
SHH-activated andTP53-mutant phology, particularly in older children. of SHH-activated and TP53-mutant or
Patients with SHH-activated and TP53-wildtype medulloblastoma are not
Definition TP53-wildtype medulloblastomas are yet available.
An embryonal tumour of the cerebellum generally children aged < 4 years, ado-
with evidence of SHH pathway activa- lescents, or young adults. In addition Imaging
tion and either germline or somatic TP53 to genetic changes activating SHH sig- On CT and MRI, medulloblastomas pres-
mutation. nalling, mutations in DDX3X or KMT20 ent as solid, intensely contrast-enhanc-
In large series of tumours, SHH-activated and amplification of MYCN or MYCL ing masses. SHH-activated medulloblas-
medulloblastomas tend to have similar are sometimes seen, as are deletions of tomas are most often identified in the
transcriptome, methylome, and micro- chromosomal arms 9q, 10q, and 14q. lateral hemispheres, but can also involve
RNA profiles. SHH pathway activation in Clinical outcomes in patients with SHH- midline structures !831,2534} Oedema
TP53-mutant tumours is associated with activated tumours are variable. was relatively common in one imaging
amplification of GL/2, MYCN, or SHH. series that included 12 desmoplastic/
Mutations in PTCH1, SUFU, and SMO are ICD-0 code 9471/3 nodular medulloblastomas and 9 me-
generally absent. Large cell I anaplastic dulloblastomas with extensive nodular-
morphology and chromosome 17p loss Grading ity !743). A nodular, so-called grape-like
are also common in SHH-activated and Like all medulloblastomas, SHH-activat- pattern on MRI often characterizes me-
TP53-mutant tumours. Patterns of chro- ed and TP53-mutant medulloblastoma dulloblastoma with extensive nodularity
mosome shattering known as chromoth- and SHH-activated and TP53-wildtype because of the tumour's distinctive and
ripsis are often present. medulloblastoma correspond histologi- diffuse nodular architecture !820,1744).
SHH-activated tumours account for ap- cally to WHO grade IV. Medulloblastomas involving the periph-
proximately 30% of all medulloblastomas eral cerebellar hemispheres in adults oc-
and originate from rhombic lip-derived Epidemiology casionally present as extra-axial lesions
cerebellar granule neuron precursors, SEER data from 1973-2007 suggest me- resembling meningiomas or acoustic
the proliferation of which is dependent on dulloblastoma incidence rates of 6.0 cas- nerve schwannomas !154).
SHH signalling activity. SHH-activated es per 1 million children aged 1-9 years
and TP53-mutant medulloblastomas are and 0.6 cases per 1 million adults aged Spread
rare and generally found in children aged > 19 years (2382). SHH-activated medul- Medulloblastomas have the potential
4-17 years. Clinical outcomes in patients loblastomas in general show a bimodal to invade locally, metastasize through
with SHH-activated and TP53-mutant tu- age distribution, being most common in the cerebrospinal fluid, or (more rarely)
mours are very poor. infants and young adults, with a male- spread outside the CNS. Overall, SHH-
to-female ratio of approximately 1.5:1 activated medulloblastomas are less fre-
ICD-0 code 9476/3 !1804). In contrast, SHH-activated and quently metastatic than group 3 tumours,
TP53-mutant tumours are generally but spread within the neuraxis is often
found in children aged 4-17 years (1333}. a presenting feature of SHH-activated
Medulloblastoma, In one study that included 133 SHH-ac- and TP53-mutant medulloblastoma.
SHH-activated andTP53-wildtype tivated medulloblastomas, 28 patients The molecular groups of medulloblas-
(21%) had a TP53 mutation, and the me- toma, including SHH-activated tumours,
Definition dian age of these patients was approxi- have been shown to remain stable in
An embryonal tumour of the cerebellum mately 15 years !2870).
with molecular evidence of SHH pathway
activation and an intact TP53 locus. Localization
SHH pathway activation in TP53-wildtype SHH-activated medulloblastomas were
tumours can be associated with germline proposed in one report to involve main-
or somatic mutations in the negative reg- ly the lateral cerebellum, a finding re-
ulators PTCH1 or SUFU, as well as acti- lated to their origin from granule neuron
vating somatic mutations in SMOor (rare- precursors (831 }. A subsequent study
ly) amplification of GL/2. Desmoplastic/ that included 17 SHH-activated medul-
nodular medulloblastomas and medul- loblastomas found that although 9 of
loblastomas with extensive nodularity are those tumours were hemispheric, the
always included in the SHH-activated other 8 were centred in, or significantly Fig. 8.08 FLAIR MRI of SHH-activated medulloblastoma.
group, but tumours with a hedgehog sig- involved, the vermis !2534). The locali- These tumours often originate from the cerebellar
nalling pathway signature can also have zation of SHH-activated tumours may hemisphere.

190 Embryonal tumours


comparisons of primary and metastatic
lesions (2692l. However, TP53 mutation
can sometimes be seen in a local or dis-
tant relapse even when it is not present in
the primary medulloblastoma (1003).

Macroscopy
some SHH-activated medulloblasto-
mas tend to be firm and more circum-
scribed than other tumours, reflecting .
intratumoural desmoplasia. Small foci of •
Fig. 8.09 SHH-activated and TPSJ.mutant medulloblastoma. A Marked anaplasia and mitotic activity, consistent with
necrosis can be grossly evident, but ex- large cell/ anaplastic medulloblastoma. B lmmunoreactivity for p53, reflecting the presence of a TP53 mutation.
tensive necrosis is rare in SHH-activated
tumours. Like other medulloblastomas, SHH-ac- desmoplastic/nodular medulloblastomas
tivated medulloblastomas can express (2296l. PTC1 is an inhibitor of hedgehog
Microscopy MAP2 and synaptophysin. In SHH-acti- signalling and is particularly important in
oesmoplastic/nodular and medulloblas- vated medulloblastomas with features of cerebellar development. The pathway li-
toma with extensive nodularity variants medulloblastoma with extensive nodular- gand SHH is secreted by Purkinje cells
of medulloblastoma are always included ity, strong NeuN nuclear labelling can and is a major mitogen for cerebellar gran-
in the SHH-activated group, but this mo- be seen in large islands with advanced ule cell progenitors in the external germi-
lecular group can also have a classic or neurocytic differentiation. Pathological nal layer (2715l. Activation of the pathway
large cell I anaplastic morphology. In one p53 accumulation can be detected in a occurs when the SHH ligand binds to
study, diffuse anaplasia was seen in 66% small proportion of SHH-activated me- PTC1, releasing PTC1 from SMO inhibition
of all SHH-activated and TP53-mutant dulloblastomas, frequently in association and activating GLI transcription factors in
medulloblastomas, but in less than 10% with signs of cytological anaplasia. This the primary cilia (280l. The SHH pathway
of TP53-wildtype tumours (2870) is correlated with somatic TP53 muta- can thus be aberrantly activated by loss
tion (2482}, and can also be linked to of PTC1 function or increased activity of
/mmunophenotype germline TP53 mutations (Li-Fraumeni SHH, SMO, or GLI factors.
Gene expression and methylation profil- syndrome) {2075l. Early array-based expression studies
ing remain the gold standard for defining identified active SHH signalling in a sub-
molecular groups of medulloblastoma. Cell of origin set of medulloblastomas, which were often
However, SHH-activated medulloblas- SHH-activated medulloblastomas are desmoplastic/nodular or medulloblas-
tomas express a signature of activated thought to derive from ATOH1-positive tomas with extensive nodularity (2000,
hedgehog signalling, and several pro- cerebellar granule neuron precursors 2549l Larger mRNA expression profiling
teins have been found to be useful as (2310,2817l. It has also been suggested experiments confirmed the existence of
surrogate markers for this activity, includ- that a proportion of SHH-activated me- this group, and it was adopted as one of
ing GAB1 (631}, TNFRSF16 (332,1402}, dulloblastomas could arise from granule four principal molecular groups by an in-
and SFRP1 (1805l. One study defined a neuron precursors of the cochlear nuclei, ternational consensus panel (2524}. Analy-
diagnostic immunohistochemical method a derivative of the auditory lower rhombic ses of genome-wide methylation profiles
that can distinguish between WNT-ac- lip of the brain stem {884l. Approximately have also supported the existence of a
tivated, SHH-activated, and non-WNT/ half of all patients with SHH-activated distinct SHH-activated group, and can be
non-SHH tumours using formalin-fixed and TP53-mutant medulloblastoma have performed in formalin-fixed tissue (1049l.
paraffin-embedded material (631l. GAB1 TP53 mutations in the germline (2870). Smaller sets of SHH-associated genes
and YAP1 are the immunohistochemi- This suggests that this mutation may play measured using gene counting technol-
cal markers indicating SHH activation. a key role in early transformation. ogy or quantitative RT-PCR can also be
The anti-GAB1 antibody labelled only used to define this molecular group for the
tumours with an SHH-activated profile Genetic profile purpose of clinical classification (1808,
or PTCH1 mutation, whereas the anti- Mutations and other genetic alterations 2355l
YAP1 antibody labelled tumour cells in activating hedgehog signalling are the Structural variations or mutations in DNA
both WNT-activated and SHH-activated main molecular drivers of SHH-activated are often distinct across the various me-
medulloblastomas, but not non-WNT/ medulloblastoma, with alterations involv- dulloblastoma molecular groups. High-lev-
non-SHH medulloblastomas. Non-des- ing known pathway genes in 116 (87%) el amplifications associated with the SHH-
moplastic SHH-activated medulloblas- of 133 SHH-activated tumours in a recent activated group include loci containing
tomas generally show widespread and study (1333). PTCH1, the principal gene MYCL, GL/2, PPM10, YAP1, and MOM4
strong immunoreactivities for GAB1 and underlying naevoid basal cell carcinoma (1807l. The MYCN locus is often ampli-
YAP1, whereas desmoplastic/nodular tu- syndrome, which predisposes patients to fied in both SHH-activated and group 4
mours display stronger staining for these developing basal cell carcinoma and me- medulloblastomas. Many of these altered
proteins within internodular regions {631, dulloblastoma, was mapped to 9q22 by loci have known links to the SHH pathway.
1670l. linkage analysis. LOH in this region has MYCN expression is directly regulated by
also been demonstrated in many sporadic GLI transcription factors in both granule

Medulloblastoma. SHH-activated 191


cell precursors and tumours {1839). YAP1 mutation or chromothripsis should be tumours, they were significantly associ-
is also a known target and important ef- tested for germline alterations, and this ated with a poor prognosis only in SHH-
fector of hedgehog signalling in neoplastic high-risk group of tumours is considered activated tumours (2870).
and non-neoplastic cerebellar progenitors a distinct variant. Mutations in TP53 are Patient outcome with SHH-activated and
(690). Homozygous deletions at the PTEN most commonly found in the DNA bind- TP53-wildtype medulloblastomas is varied
and PTCH1 loci have also been found pref- ing regions encoded by exons 4 through 8 and shows a significant association with
erentially or exclusively in SHH-activated {2870). Approximately half of all SHH-ac- pathological features. Medulloblastomas
tumours compared with other molecular tivated and TP53-mutant medulloblasto- with extensive nodularity in infants are not-
groups (1807). mas have been shown to have germline ed for a good prognosis, and most desrno.
Somatic mutations are relatively rare in rather than somatic alterations. Although plastic/nodular tumours in this young age
medulloblastoma compared with other tu- some WNT-activated medulloblastomas group also have a relatively good outcome
mours, with a median of 12 non-silent and have mutations in TP53, these changes (2208). The situation for SHH-activated
4 silent mutations reported in one study of have so far been somatic {2870) and TP53-wildtype medulloblastomas with
coding regions across 92 primary medul- Another inherited syndrome associated a classic or large cell I anaplastic morphol-
loblastoma/normal sample pairs (2042). with SHH-activated medulloblastoma is ogy is less clear, though these appear to
In this and a similar study (2142). muta- naevoid basal cell carcinoma syndrome have a worse outcome than the two types
tions in PTCH1, SUFU, and other genes (also called Gorlin syndrome). It is charac- of desmoplastic medulloblastoma, at least
associated with the hedgehog signalling terized by multiple basal cell carcinomas in infancy.
pathway were found exclusively in SHH- of the skin, odontogenic jaw keratocysts, Molecular factors predicting response to
activated tumours. However, in some medulloblastoma, and developmental ab- therapies targeting SHH are beginning to
SHH-activated tumours, no clear genetic normalities. Most naevoid basal cell car- emerge. Small-molecule inhibitors that act
explanation for SHH pathway activation cinoma syndrome cases are due to het- on the SMO receptor have been shown to
can be determined. erozygous germline mutations in PTCH1, be effective in some medulloblastomas
The most common somatic point muta- with mutations identified in 97 of 171 pa- with hedgehog activation, but not in other
tions are in the TERT promoter, resulting in tients (56%) in one study {2376). However, molecular groups (2143,2355). However,
increased telomerase activity. An analysis mutations in PTCH1 and TP53 are thought SHH-activated medulloblastoma can also
of 466 medulloblastomas revealed TERT to be mutually exclusive {1333) The age be resistant to SMO inhibitors, due to acti-
mutations in 21% overall, with the highest distributions of these groups also differ; vation of the pathway downstream of phar-
frequency - 83% (55 of 66 cases) - identi- medulloblastomas with germline PTCH1 macological blockade. Such downstream
fied among the adult cases of the SHH- or SUFU mutations present in infants and activation can be present at diagnosis or
activated group, in which they were linked children aged < 4 years. whereas those can develop as a therapeutic resistance
to good outcomes {2098). Point mutations with a germline TP53 mutation occur in mechanism. It has been suggested that
in TP53, particularly in SHH-activated rath- older children. the genetic mechanism of pathway activa-
er than WNT-activated medulloblastomas, tion is linked to the likelihood of response
are associated with chromothripsis, in Prognosis and predictive factors to SMO inhibition (1333,2143) Adults with
which chromosomes shatter and acquire SHH-activated and TP53-mutant medullo- SHH-activated medulloblastoma are more
multiple rearrangements simultaneously in blastomas are associated with a very poor likely to harbour activating alterations in
a single catastrophic event (2075). Overall, outcome In one study, the 5-year overall PTCH1 or SMO resulting in tumours sen-
the frequency of specific genetic chang- survival of patients with an SHH-activated sitive to SMO inhibitors, whereas SHH-
es in SHH-activated medulloblastomas medulloblastoma was 76% for those with a activated medulloblastomas from infants
seems to be somewhat different in infants, TP53-wildtype tumour and 41% for those and children (including SHH-activated
children, and adults {1333). with a TP53-mutant tumour {2870). Known and TP53-mutant tumours) often contain
clinical high-risk factors, such as metastat- downstream alterations in SUFU, GL/2,
Genetic susceptibility ic disease, are also associated with TP53 and MYCN that are refractory to these
Inherited point mutations in TP53 in Li- mutation within the SHH-activated group. pharmacological agents. It has been sug-
Fraumeni syndrome can result in medul- It is becoming increasingly clear that some gested that DNA-damaging alkylating
loblastoma, and it has been shown that genetic prognostic markers must be in- agents and radiation should be avoided
these medulloblastomas belong to the terpreted within the context of molecular whenever possible when treating patients
SHH-activated group and are prone to group. For example, although TP53 muta- with a germ line TP53 mutation (2075)
chromothripsis (2075). Patients whose tions were found in 16% of WNT-activat-
SHH-activated tumours harbour TP53 ed tumours and 21% of SHH-activated

192 Embryonal tumours


Medulloblastoma, Ellison OW. a variable extent, and tumour cells are
non-WNT/non-SHH Eberhart C.G. rarely immunopositive for GFAP. With a
Pfister S. panel of three antibodies (to beta-catenin,
GAB1, and YAP1), non-WNT/non-SHH tu-
mours show cytoplasmic (but not nuclear)
beta-catenin immunoreactivity, and the tu-
mour cells are immunonegative for GAB1
Definition diagnosis, a presentation that is a par- and YAP1 (631).
An embryonal tumour of the cerebellum ticular feature of group 3 medulloblasto-
consisting of poorly differentiated cells mas in infancy (1334,1804). Genetic profile
and excluded from the WNT-activated and Overexpression of MYC is a cardinal fea-
SHH-activated groups by molecular testing. Microscopy ture of group 3 medulloblastomas, and
Non-WNT/non-SHH medulloblastomas Most non-WNT/non-SHH medulloblasto- MYC amplification (often accompanied
are either group 3 or group 4 medulloblas- mas have a classic morphology. These by MYC-PVT1 fusion (1807)) is relatively
tomas. These are classic or large cell I tumours occasionally exhibit areas of ro- common among group 3 medulloblasto-
anaplastic tumours that cluster into two sette formation or a palisading pattern of mas. However, MYC amplification is found
groups in terms of transcriptome, methy- tumour cell nuclei. Nodule formation can mainly in infant disease, and occurs in
lome, and microRNA profiles as analysed occur in the absence of desmoplasia in < 25% of group 3 tumours overall (632,
across large series of medulloblastomas. non-WNT/non-SHH medulloblastomas. 1426). Other recurrently mutated or focally
Distinct methylome profiles probably re- A reticulin preparation demonstrates no amplified genes include SMARCA4 (al-
flect a different histogenesis of the four strands of collagen around or between tered in 10.5% of cases), OTX2 (in 7.7%),
medulloblastoma groups, although tu- the nodules, which otherwise show neu- CTDNEP1 (in 4.6%), LRP1B (in 4.6%), and
mours from groups 3 and 4 are more simi- rocytic differentiation and a reduced KMT20 (in 4%) (1804). Two recurrent on-
lar to each other than to WNT-activated growth fraction, in a similar manner to cogenes in group 3 medulloblastomas
or SHH-activated medulloblastomas (see nodules associated with desmoplasia in are the homologues GFl1 and GFl1B,
Table 8.01, p. 184). desmoplastic/nodular medulloblastoma which are activated through a mechanism
The group 3 transcriptome profile is char- or medulloblastoma with extensive nodu- called enhancer hijacking (1806). By far
acterized by relatively high expression of larity. Large cell I anaplastic tumours in the most common cytogenetic aberrations
MYC, and MYC amplification is overrep- the non-WNT/non-SHH molecular group in medulloblastoma (occurring in -80%
resented in this molecular group. Group 4 generally belong to group 3. of group 4 tumours) involve copy number
tumours are characterized by recurrent al- alterations on chromosome 17: 17p dele-
terations in KDM6A and SNCAIP, as well lmmunophenotype tion, 17q gain, or a combination of these in
as in other genes. Non-WNT/non-SHH tu- The neural marker immunohistochemical the form of an isodicentric 17q (631,1334,
mours account for approximately 60% of profile of non-WNT/non-SHH medullo- 1804). The most frequently mutated or fo-
all medulloblastomas and typically have blastoma is the same as those of classic cally amplified genes in group 4 tumours
classic histopathological features. Most or large cell I anaplastic tumours in the are KOM6A (altered in 13% of cases), the
non-WNT/non-SHH tumours present in WNT-activated or SHH-activated groups. locus around SNCAIP (in 10.4%), MYCN
childhood; they are relatively uncommon The tumours express synaptophysin to (in 6.3%), KMT2C (in 5.3%), COK6 (in
in infants and adults. 4 7%), and ZMYM3 (in 3.7%) (1804). Acti-
vated GFI oncogenes have also been ob-
ICD-0 code 9477/3 served in a subset of group 3/4 tumours
that do not cluster reliably into one or other
Epidemiology group (1806).
Group 3 medulloblastomas account for
approximately 20% of all cases, and for Prognosis and predictive factors
a higher proportion of cases (-45%) in MYC amplification has long been estab-
infants. Group 3 medulloblastoma is ex- lished as a genetic alteration associated
ceedingly rare in adults {1334). Group 4 with poor outcome in patients with medullo-
medulloblastomas are the largest mo- blastoma (620,632,2268). This observation
lecular group, accounting for about 40% is reflected in the relatively poor outcome
of all tumours. Peak incidence occurs in of group 3 medulloblastomas, but MYC
patients aged 5-15 years, with lower inci- amplification has prognostic significance
dence in infants and adults (1807) even among group 3 tumours (2345). Met-
astatic disease at the time of presentation,
Spread Fig. 8.10 MYC amplification. Nuclei show multiple
which is associated with poor outcome,
Metastatic disease is present in about clumped MYC signals indicative of double minutes seems to be the most robust prognostic
40% of group 3 tumours at the time of (green). The red signals from centromeric probes indicate marker among group 4 tumours (2345).
chromosome 8 copy number.

Medulloblastoma, non-WNT/non-SHH 193


Medulloblastomas, histologically defined tumours, but these are never associ-
ated with internodular desmoplasia or
perinodular collagen when examined in
a reticulin preparation. Additionally, these
Medulloblastoma, classic Ellison D.W. Pietsch T. non-desmoplastic nodular medulloblas-
Eberhart C.G. Wiestler 0.0. tomas are non-WNT/non-SHH tumours,
Giangaspero F. Pfister S. unlike desmoplastic/nodular tumours,
Haapasalo H. which belong to the SHH-activated
group.

Definition Epidemiology lmmunophenotype


An embryonal neuroepithelial tumour The classic medulloblastoma is more fre- Classic medulloblastomas express vari-
arising in the cerebellum or dorsal brain quent than its variants in childhood, but is ous non-specific neural markers, such
stem, consisting of densely packed small less common than desmoplastic/nodular as NCAM1, MAP2, and neuron-specific
round undifferentiated cells with mild to medulloblastoma in infants and adults. enolase. Most cases are immunopositive
moderate nuclear pleomorphism and a for synaptophysin and NeuN, but these
high mitotic count. Microscopy neuronal markers may also be absent.
Classic medulloblastomas lack sig- Classic medulloblastomas are the arche- lmmunoreactivity for NFPs is very rare.
nificant intratumoural desmoplasia, the typal CNS small blue round cell tumour. Cells showing GFAP expression and an
marked nuclear pleomorphism of the They consist of a syncytial arrangement embryonal morphology can be observed
anaplastic variant, and the cytological of densely packed undifferentiated em- in as many as 10% of medulloblastomas
features of the large cell variant. Classic bryonal cells. Mitotic figures and ap- {314). When present, these cells are infre-
medulloblastomas account for 72% of all optotic bodies are found among the tu- quent and tend to be scattered through-
medulloblastomas. They occur through- mour cells. lntratumoural desmoplasia is out the tumour, which is unlike the pattern
out the patient age range of medulloblas- lacking, but pericellular desmoplasia is of GFAP immunoreactivity in small-cell
toma, from infancy to adulthood, but pre- induced where tumour cells invade the astrocytic tumours.
dominantly in childhood, and are found leptomeninges. Homer Wright rosettes Nuclear SMARCB1 and SMARCA4 ex-
in all four molecular medulloblastoma are found in some classic (and large cell I pression is retained in all medulloblas-
groups. anaplastic) medulloblastomas. toma types; the loss of expression of one
Occasionally, nodules of neurocytic dif- of these SWI/SNF complex proteins is
ICD-0 code 9470/3 ferentiation and reduced cell prolifera- diagnostic of atypical teratoid/rhabdoid
tion are evident in some areas of classic tumour.

•L --· � ·•· • - -
Fig. 8.11 Histopathological features of the classic medulloblastoma. A Typical syncytial arrangement of undifferentiated tumour cells. B Area with Homer Wright (neuroblastic)
rosettes. C Arrangement of tumour cells in parallel rows (spongioblastic pattern).

-.a .,
B Focal GFAP staining of tumour cells. C Clusters of medulloblastoma cells expressing retinal S-antigen.

194 Embryonal tumours


Desmoplastic/nodular Pietsch T. Wiestler O.D. all cases !1627,2207} In one retrospec-
medulloblastoma Ellison OW. Pfister S. tive cohort of adult patients, desmoplas-
Haapasalo H. Eberhart C.G. tic/nodular medulloblastoma constituted
Giangaspero F. 21% of all medulloblastomas (1347).

Localization
Definition Gorlin syndrome). Desmoplastic/nodular Unlike most classic (non-WNT) medul-
An embryonal neural tumour arising in medulloblastoma displays pathological loblastomas, which are restricted to the
the cerebellum and characterized by activation of the SHH pathway, which is midline, desmoplastic/nodular medul-
nodular, reticulin-free zones and interven- caused by mutations in genes that en- loblastoma may arise in the cerebellar
ing densely packed, poorly differentiated code components of the pathway, such hemispheres and in the vermis. Most me-
cells that produce an intercellular net- as PTCH1, SMO, and SUFU. Genetic dulloblastomas occurring in the cerebel-
work of reticulin-positive collagen fibres. and histological features of classic me- lar hemispheres are of the desmoplastic/
Desmoplastic/nodular medulloblastoma dulloblastoma, such as isochromosome nodular type !332)
is characterized by specific clinical, ge- 17q and neuroblastic rosettes, are ab-
netic, and biological features. It occurs in sent. Desmoplastic/nodular medulloblas- Imaging
the cerebellar hemispheres and the mid- toma overlaps histologically with MBEN, On MRI, desmoplastic/nodular medul-
line and has a bimodal patient age dis- which contains large irregular reticulin- loblastomas present as solid, frequently
tribution, with a relatively high incidence free regions of neurocytic differentiation contrast-enhancing masses. Tumours
in young children and adolescents, as between narrow desmoplastic strands involving the peripheral cerebellar hemi-
well as among adults. In early child- of proliferating embryonal cells. Desmo- spheres in adults occasionally present as
hood, it is associated with naevoid basal plastic/nodular medulloblastoma is asso- extra-axial lesions.
cell carcinoma syndrome (also called ciated with a more favourable outcome in
young children than are non-desmoplas- Spread
. tic variants of medulloblastoma. Tumours can relapse locally, metastasize
' I
• 'oeveloplng via cerebrospinal fluid pathways, and
I
·I EGL��· ICD-0 code 9471/3 in rare cases spread to extra-CNS sites
I'---... Rhombic
.• Lip Grading
100

.,, ........
,/ -
.
90
Like all medulloblastomas, desmoplastic/ 80
nodular medulloblastoma corresponds � 70
./
./
histologically to WHO grade IV. 0
-;,.
60
,.v
� 50
"'� 40 ,, /
Epidemiology I 30
I
Desmoplastic/nodular medulloblastomas u
20

are estimated to account for 20% of all 10


I
/
medulloblastomas (1972). In children 10 20 30 40 50 60 70
aged < 3 years, desmoplastic/nodular Age at diagnosis

Fig. 8.14 The developing human posterior Iossa. EGL, medulloblastoma accounts for 47-57% of Fig. 8.15 Cumulative age distribution of 180 cases (both
external granule layer; VZ, ventricular zone. sexes). Data from the Brain Tumor Reference Center, Bonn.

Desmoplastic/nodular medulloblastoma 195


Fig. 8.16 Desmoplastic/nodular medulloblastoma. A T1-weighted, (B) T2-weighted contrast-enhanced MRI of tumours in the cerebellar hemisphere. C T1-weighted, contrast-
enhanced MRI of a tumour in the vermis.

such as the skeletal system. At diagno- Microscopy hyperchromatic and moderately pleo-
sis, metastatic disease is found less fre- Desmoplastic/nodular medulloblastoma morphic nuclei, which produce a dense
quently with desmoplastic/nodular me- is characterized by nodular, reticulin- intercellular reticulin fibre network (818,
dulloblastomas than with other variants. free zones (so-called pale islands) sur- 1234). In rare cases, this defining pat-
rounded by densely packed, undiffer- tern is not present throughout the en-
entiated, highly proliferative cells with tire tumour and there is instead a more

,2r...;1=a
4. ,·i�.
,=,.;....,....,., ,,,L:
Fig. 8.17 Desmoplastic/nodular medulloblastoma. A Pale nodular areas surrounded by densely packed hyperchromatic cells. B Reticulin silver impregnation showing the reticulin-
free pale islands. C MIB1 monoclonal antibody staining shows that the proliferative activity predominates in the highly cellular, intermodal areas. D Neuronal differentiation, shown
by immunoreactivity for neuron-specific enolase, occurs mainly in the pale islands.

196 Embryonal tumours


syncytial arrangement of non-desmo-
plastic embryonal cells present in a few
areas. The nodules contain tumour cells
with features of variable neurocytic matu-
ration embedded in a neuropil-like fibril-
tary matrix. The level of mitotic activity in
the nodules is lower than in the internod-
ular areas. Neuroblastic rosettes are not
found in desmoplastic/nodular medul-
loblastoma. Tumours with small nodules ,,���di(.'g-It..
.:. . ., .�li..
f �
'11,..-,;,:l'a'.l,i-·i::t.,,.:I<.J-...:..i!WII!
'"'

can easily be overlooked if no reticulin Fig. 8.18 Desmoplastic/nodular medulloblastoma. A The nodules represent zones of neuronal maturation and show
staining is performed. Medulloblastomas intense immunoreactivity for synaptophysin. B SHH activation can be visualized by immunohistochemistry with
that show only an increased amount of antibodies against SHH targets, in this case, with antibodies against TNFRSF16 (also called p75-NGFR) (332,1401),
reticulin fibres (without a nodular pat- which is strongly expressed in the synaptophysin-negative internodular areas.
tern) or that show a focal nodular pattern
without desmoplasia are not classified as
desmoplastic/nodular medulloblastoma on SHH (produced by Purkinje cells) as a syndrome are mainly desmoplastic vari-
11627); the two characteristic features mitogen 12715}. ants (i.e. desmoplastic/nodular medul-
must occur together for a diagnosis of loblastoma or medulloblastoma with
desmoplastic/nodular medulloblastoma. Genetic profile extensive nodularity) {67). It has been
Oesmoplastic/nodular medulloblastoma shown that the risk of medulloblastoma
Jmmunophenotype displays pathological activation of the in PTCH1-related naevoid basal cell car-
The nodules in desmoplastic/nodular SHH pathway, which is often caused by cinoma syndrome is approximately 2%,
medulloblastoma show variable expres- mutations in genes encoding members and that the risk is 20 times the value in
sion of neuronal markers, including syn- of the pathway, including PTCH1, SMO, SUFU-related naevoid basal cell carci-
aptophysin and NeuN. Nodules with and SUFU 11422,1973,2523). In a recent noma syndrome 12376). In children with
very strong NeuN expression, which is analysis using next-generation sequenc- SUFU-related naevoid basal cell carci-
an indicator of advanced neurocytic dif- ing, 85% of desmoplastic/nodular medul- noma syndrome, brain MRI surveillance
ferentiation, are typical of medulloblas- loblastomas carried genetic alterations is highly recommended 12376). Germline
toma with extensive nodularity, but can in PTCH1, SUFU, SMO, SHH, GL/2, or mutations of SUFU have also been de-
also occur in the desmoplastic/nodular MYCN. This study showed a predomi- scribed in patients with desmoplastic
variant. The Ki-67 proliferation index is nance of SUFU and PTCH1 mutations medulloblastomas that do not fulfil the
much higher in internodular areas than in young children, whereas PTCH1 and diagnostic criteria for naevoid basal cell
in nodules 11627). Activation of the SHH SMO mutations were more common in carcinoma syndrome 1294). The families
pathway can be inferred by immunohis- adults {1333) Rare mutations in other of infants with desmoplastic medulloblas-
tochemistry for specific targets, such as genes (e.g. LOB1) have also been de- tomas should be offered genetic coun-
GAB1 and TNFRSF16 1631,1402). These scribed. Recurrent ODX3X mutations, as selling because of the high frequency of
markers are expressed predominantly well as TERT promoter mutations, have germline alterations and naevoid basal
in internodular areas. GFAP expression been identified. Allelic losses of regions cell carcinoma syndrome 1787).
can be found specifically in tumour cells on chromosomes 9q and 10q are found
in a subset of cases 1332). Widespread in some desmoplastic/nodular medullo- Prognosis and predictive factors
and strong nuclear accumulation of p53, blastomas {2296}, whereas isochromo- In most cases, desmoplastic/nodular
suggesting a TP53 mutation, can be de- some 17q, which is a marker of midline medulloblastoma in early childhood has
tected in rare desmoplastic/nodular me- classic (non-WNT) and large cell I ana- an excellent outcome with surgery and
dulloblastomas, frequently in association plastic medulloblastomas, is absent from chemotherapy alone {2207). In a meta-
with signs of cytological anaplasia. This desmoplastic/nodular medulloblastoma analysis of prognostic factors in infant
finding can accompany either somatic 11334). medulloblastoma, progression-free or
or germline TP53 alteration (Li-Fraumeni overall survival at 8 years was signifi-
syndrome) 12075,2482). Genetic susceptibility cantly better for desmoplastic variants
Naevoid basal cell carcinoma syndrome than for other medulloblastomas {2208).
Cell of origin (also called Gorlin syndrome) is caused No survival difference between desmo-
Desmoplastic/nodular medulloblastomas mainly by heterozygous germline muta- plastic/nodular medulloblastoma and
are derived from granule cell progenitor tions in PTCH1 and rarely by germline classic medulloblastoma was found in a
cells forming the external granule cell mutations in SUFU or PTCH2 12376). European multicentre trial involving older
layer during cerebellar development Medulloblastomas occurring in the con- children with standard-risk medulloblas-
1332} These progenitors are dependent text of naevoid basal cell carcinoma toma 11431).

Oesmoplastic/nodular medulloblastoma 197


Medulloblastoma with Giangaspero F. Pietsch T. Imaging
extensive nodularity Ellison D.W. Wiestler O.D. On MRI, medulloblastoma with extensive
Eberhart C.G. Pfister S. nodularity presents as a very large multi-
Haapasalo H. nodular lesion with enhancing grape-like
structures involving the vermis and some-
times the cerebellar hemispheres {2821).
Rare cases have a peculiar gyriform pre-
Definition sentation, in which the cerebellar folia are
100
An embryonal tumour of the cerebellum �� well-delineated and enlarged, with con-
characterized by many large reticulin-free
nodules of neurocytic cells against a neu- � 80
90
v: trast enhancement {25,787). Downward
herniation of the cerebellar tonsils and
ropil-like matrix and by narrow internodu-
QJ

� 70 / effacement of the cisternal spaces of the


lar strands of poorly differentiated tumour 0
u
60 / posterior fossa can be observed.
'#. J
cells in a desmop/astic matrix. � so
In the medulloblastoma with extensive
:,::;
� 40
/ Spread
nodularity (MBEN), the internodular retic-
::,

5 30
/ Medulloblastoma with extensive nodu-
ulin-rich component with embryonal cells
u
20
I larity can relapse locally or (rarely) can
is a minor element. This variant occurs 10 metastasize via cerebrospinal fluid path-
predominantly in infants and is associ- 0 ways. However, such cases seem to re-
ated with a favourable outcome with cur- <1 <2 <3 <4 <5 <6 spond well to subsequent treatment and
Age at diagnosis (years)
rent treatment regimens. have a favourable prognosis (820,2208).
MBEN is closely related to desmoplastic/ Fig. 8.19 Cumulative age distribution of 24 cases (both
nodular medulloblastoma, with which it sexes). Data from the Brain Tumor Reference Center, Bonn. Microscopy
overlaps histopathologically and geneti- Medulloblastoma with extensive nodular-
cally; both variants are SHH-activated tu- < 3 years (in whom desmoplastic/nodular ity differs from the related desmoplastic/
mours. MBEN is associated with naevoid medulloblastomas account for as many nodular variant in that is has an expanded
basal cell carcinoma syndrome (also as 50% of cases {1627,2207)), medullo- lobular architecture due to the fact that
called Gorlin syndrome). blastoma with extensive nodularity has the reticulin-free zones become unusu-
been reported to account for 20% of all ally enlarged and rich in neuropil-like tis-
ICD-0 code 9471/3 cases (787). sue. These zones contain a population of
small cells with round nuclei, which show
Grading Localization neurocytic differentiation and exhibit a
Like all medulloblastomas, MBEN corre- More than 80% of medulloblastomas streaming pattern. Mitotic activity is low
sponds histologically to WHO grade IV. with extensive nodularity are located in or absent in these neurocytic areas. The
the vermis (787). This localization con- internodular component is markedly re-
Epidemiology trasts with that of desmoplastic/nodular duced in some areas (820,1627,2459).
In large series, medulloblastomas with medulloblastoma, which more frequently After radiotherapy and/or chemother-
extensive nodularity account for 3.2- involves the cerebellar hemispheres. apy, medulloblastomas with extensive
4.2% of all medulloblastoma variants nodularity occasionally undergo further
overall {619,1972). but in children aged

Fig. 8.20 Medulloblastoma with extensive nodularity. A Multinodular and gyriform pattern. B In a 1-month-old girl, the gadolinium-enhanced sagittal T1-weighted MRI shows a huge
lesion involving both cerebellar hemispheres and the vermis. The lesion has a multinodular and gyriform pattern of enhancement. C Note the downward herniation of the tumour
through the foramen magnum (arrow) and the marked effacement of the cisternal spaces of the posterior Iossa. There is also supratentorial hydrocephalus and macrocrania.

198 Embryonal tumours


maturation into tumours dominated by members of the SHH pathway. Most cas- SUFU-related naevoid basal cell carcino-
ganglion cells {419,538). es harbour a SUFU mutation {294). How- ma syndrome, neuroimaging surveillance
ever, a recent study of 4 medulloblasto- is recommended {2376). Families with
lmmunophenotype mas with extensive nodularity found a children that present with MBEN should
Like in desmoplastic/nodular medullo- PTCH1 mutation in 2 of the tumours and be offered genetic counselling because of
blastomas, the neuropil-like tissue and an SUFU and SMO mutation in one each the high frequency of naevoid basal cell
the differentiated neurocytic cells within of the other 2 tumours {1333}. carcinoma syndrome {787,2376).
nodules are strongly immunoreactive for
synaptophysin and NeuN and the Ki-67 Genetic susceptibility Prognosis and predictive factors
proliferation index is much higher in in- In the majority of cases, naevoid basal cell Medulloblastoma with extensive nodular-
ternodular areas {1627). Activation of the carcinoma syndrome is caused by germ- ity has an excellent outcome in the ma-
SHH pathway can be demonstrated by line mutations of PTCH1. In a few cases, jority of cases {787,1603,2208). In an in-
immunohistochemistry for specific targets germline mutations instead occur in SUFU ternational meta-analysis of survival and
such as GAB1 {631} or TNFRSF16 {1402) {2376} or PTCH2 (667,747) Naevoid basal prognostic factors in infant medulloblas-
cell carcinoma syndrome is diagnosed in torna. the progression-free and overall
Cell of origin 5.8% of all patients with medulloblastoma, survival rates of 21 cases of medulloblas-
Medulloblastoma with extensive nodu- but in 22.7% of patients with a desmo- toma with extensive nodularity at 8 years
larity, like most SHH-activated medul- plastic/nodular tumour variant and 41% were 86% and 95%, respectively {2208}.
loblastomas, seems to be derived from of patients with medulloblastoma with Metastatic disease at presentation did
ATOH1-positive cerebellar granule neu- extensive nodularity. The risk of medullo- not affect the favourable prognosis, sug-
ron precursors {2310,2817). blastoma in PTCH1-related naevoid basal gesting that a diagnosis of medulloblas-
cell carcinoma syndrome is approximately toma with extensive nodularity confers
Genetic profile 2%, and the risk is 20 times the value in a better outcome regardless of adverse
Medulloblastoma with extensive nodular- SUFU-related naevoid basal cell carcino- clinical features {2208}
ity carries mutations in genes encoding ma syndrome {294,1333). In children with

Medulloblastoma with extensive nodularity 199


Large cell I anaplastic Ellison OW Pietsch T. the anaplastic variant {821,1626}; its cells
medulloblastoma Giangaspero F. Wiestler O.D. are large and monomorphic with promi-
Eberhart CG. Pfister S. nent nucleoli, but it shows the high rate of
Haapasalo H. cell turnover seen in anaplastic tumours.
Nearly all large cell medulloblastomas
contain regions with an anaplastic phe-
notype; the pure form is rare.
Definition cell I anaplastic (LC/A) variant {822,1671,
An embryonal neural tumour of the cer- 2664). This variant accounts for approxi- Genetic profile
ebellum or dorsal brain stem character- mately 10% of all medulloblastomas and Large cell I anaplastic medulloblastomas
ized by undifferentiated cells with marked occurs across the patient age range of are found mainly among group 3 or SHH-
nuclear pleomorphism, prominent nucle- the medulloblastoma. activated medulloblastomas. Among
oli, cell wrapping, and high mitotic and group 3 medulloblastomas, which over-
apoptotic counts. ICD-0 code 9474/3 express MYC, large cell I anaplastic
Large cell I anaplastic (LC/A) medullo- morphology (in particular the large cell
blastoma demonstrates severe anapla- Grading phenotype) is associated with MYC am-
sia or a large cell phenotype across the Like all medulloblastomas, LC/A medul- plification {620). SHH-activated LC/A
majority of the tumour. 'Severe anaplasia' loblastoma corresponds histologically to tumours are particularly associated with
combines marked nuclear pleomorphism WHO grade IV. GL/2 and MYCN amplification, TP53 mu-
with abundant mitotic activity and apo- tations (which are often germline), and
ptosis {619,1626}. The large cell pheno- Epidemiology specific patterns of chromosome shat-
type manifests uniform round nuclei with Large cell I anaplastic medulloblastomas tering called chromothripsis {1333,2075).
prominent nucleoli. lntratumoural des- are found across all molecular groups of
moplasia is not a feature of this variant, the disease, accounting for about 10% of Prognosis and predictive factors
but desmoplasia can be evident when all tumours {619,1626). They can occur At the time the tumours were first de-
tumour cells overrun the leptomeninges. in patients of any age. Considered sepa- scribed, large cell and anaplastic medul-
By definition, classic morphology, with rately, anaplastic medulloblastomas are loblastomas were strongly suspected to
only mild to moderate nuclear pleomor- about 10 times as prevalent as large cell behave more aggressively than classic
phism, cannot be present across a ma- medulloblastomas {630). They are most tumours, often presenting with metastatic
jority of the tumour. frequent among group 3 and SHH-acti- disease {288,821 }. In retrospective stud-
Anaplastic and large cell medulloblasto- vated medulloblastomas; LC/A tumours ies of patients in trial cohorts, large cell I
mas were initially described as separate hardly ever occur in the WNT-activated anaplastic morphology has been shown
variants, but the pure large cell tumour group. to be an independent prognostic indica-
is extremely rare; most large cell me- tor of outcome {619,632}; in current trials,
dulloblastomas also contain regions of Microscopy LC/A tumours are regarded as high-risk
anaplasia {288,619,821,1626}. Both mor- Anaplasia as a feature of an embryonal tumours warranting intensified adjuvant
phological variants are associated with a tumour was first proposed for medul- therapy. The 5-year progression-free sur-
poor outcome with standard therapies, loblastomas with marked nuclear pleo- vival rate for LC/A medulloblastomas is
as well as with amplification of MYC or morphism accompanied by particularly 30-40% {632,2192). although SHH-acti-
MYCN, although these associations are high mitotic and apoptotic counts {288, vated LC/A tumours with a TP53 mutation
not strong and depend on the tumour's 619). Nuclear moulding and cell wrap- and group 3 tumours with MYC amplifica-
molecular group. For clinical purposes, ping are additional features. Large cell tion can behave even more aggressively
the two variants are regarded as equiva- medulloblastoma lacks the variability in {2192,2870).
lent, so are now combined as the large cell size and shape that characterizes

� .,. . �
Fig. 8.22 Large cell/ anaplastic medulloblastoma. A,B Increased nuclear size, pleomorphism, and prominent nucleoli. Tumour "cell wrapping" is also evident (B).

200 Embryonal tumours


Embryonal tumour with multilayered Korshunov A.
Mclendon R.
Sarkar C.
Ng H.-K.
rosettes, C19MC-altered Judkins A.R.
Pfister S.
Huang A.
Kool M.
Eberhart C.G. Wesseling P.
Fuller G.N.

Definition varied terminology previously applied to


An aggressive CNS embryonal tumour these tumours, their rarity, and the fact
with multilayered rosettes and alterations that some lack signature microscopic
(including amplification and fusions) in features and must be distinguished ge-
the C19MC locus at 19q13.42 netically from other CNS embryonal
C19MC-altered embryonal tumours with lesions. Initially, most ETMRs were de-
multilayered rosettes (ETMRs) can develop scribed as single cases, and the largest
in the cerebrum, brain stem, or cerebellum cumulative series includes approximate-
and span a broad histological spectrum. ly 100 samples. With few exceptions,
As well as multilayered rosettes, many ETMRs affect children aged < 4 years,
tumours also contain both primitive em- the vast majority of cases occurring dur-
bryonal regions and differentiated areas ing the first 2 years of life. There is an
with broad swaths of neoplastic neuropil. almost equal distribution between males Fig. 8.23 Well-circumscribed cortical ETMR.
Most paediatric CNS embryonal tumours and females, with a male-to-female ratio
previously classified as embryonal tumour of 1.1:1 1801,1349,2403). histopathological features with intracrani-
with abundant neuropil and true rosettes, al ETMR but disclose striking molecular
ependymoblastoma, and medulloepithelio- Localization diversity and consequently deserve a
ma are included in this group 11349,2403). ETMRs develop in both the supratento- separate nosological designation 11129,
However, any CNS embryonal tumour with rial and infratentorial compartments. The 1345A).
C19MC amplification or fusion qualifies for most common site is the cerebral hemi-
this designation, including those without sphere (affected in 70% of cases), with Clinical features
distinctive histopathological features. frequent involvement of the frontal and The most common clinical manifestations
parietotemporal regions 1801,1349} Oc- are symptoms and signs of increased in-
ICD-0 code 9478/3 casionally, these tumours can be very tracranial pressure (i.e. headache, vom-
large, involving multiple lobes and even iting, nausea, and visual disturbances).
Grading both cerebral hemispheres. An infraten- Focal neurological signs (i.e. ataxia or
Like other CNS embryonal tumours, torial location is less frequent, with either weakness) are more common in older
C19MC-altered ETMR corresponds his- cerebellum or brain stem affected in 30% children and in cases with infratentorial
tologically to WHO grade IV of cases. Tumour protrusion into the ce- location.
rebellopontine cistern may be observed.
Epidemiology Some extracranial tumours (e.g. intraocu- Imaging
The true incidence of C19MC-altered lar medulloepithelioma and sacrococ- CT and MRI usually show contrast-
ETMR is difficult to determine due to the cygeal ependymoblastoma) share some enhancing large tumour masses,

.--·-.
Fig. 8.24 Embryonal tumour with abundant neuropil and true rosettes. Biphasic histological pattern: areas of small Fig. 8.25 Multi layered rosette: a key diagnostic feature of
embryonal cells with multilayered rosettes and neuropil-like areas with neoplastic neurocytic cells. embryonal tumours with multilayered rosettes.

Embryonal tumour with multilayered rosettes, C19MC-altered 201


..,:-t ·-·
Fig. 8.27 ETMR, with morphology of embryonal tumour with abundant neuropil and true rosettes. A Synaptophysin expression within neuropil areas. B Vimentin expression.
C Intense LIN28A immunoreactivity.

sometimes containing cysts or calcifica- debris. The cells facing the lumen have a structures in the otherwise paucicellular
tion. The radiological differential diagno- defined apical surface with a prominent neuropil-like areas, and rarely neoplas-
sis of these lesions includes other CNS internal limiting membrane in some ro- tic ganglion cells can be found between
embryonal tumours, desmoplastic in- settes. The nuclei of the rosette-forming the cells composing the layers of the
fantile ganglioglioma and supratentorial cells tend to be pushed away from the rosettes.
anaplastic ependymoma. lumen towards the outer cell border. In
most tumours, a defined outer mem- Ependymoblastoma
Spread brane around rosettes is lacking. There This pattern of ETMR features sheets
The tumour may be locally and widely are three histological patterns found in and clusters of poorly differentiated cells
infiltrative. Widespread leptomeningeal ETMR , C91 MC-altered. On the basis of incorporating numerous multilayered
dissemination, extracranial invasive their molecular commonality, these are rosettes, but typically lacks a neuropil-
growth in the soft tissues, and extracra- now considered to constitute either vari- like matrix and ganglion cell elements.
nial metastases have all been reported ous points along a morphological spec- Rosettes are intermixed with small to
{1300,1349,2771). trum or diverse differentiation within a medium-sized embryonal cells that have
single tumour entity, rather than distinct a high nuclear-to-cytoplasmic ratio and
Macroscopy nosological categories {1191,1349). variably developed fibrillary processes.
ETMR is usually greyish pink, and well
circumscribed, with areas of necrosis Embryona/ tumour wlfh abundant Medu//oeplfhelioma
and haemorrhage and minute calcifica- neuropi/ and true rosettes This pattern of ETMR typically presents
tions. Some tumours are cystic. Wide- This pattern of ETMR shows a biphasic as a distinct cerebral mass in young
spread leptomeningeal dissemination architecture featuring dense clusters of children. It is characterized by papillary,
and extraneural metastases are frequent small cells with round or polygonal nu- tubular, and trabecular arrangements of
in the terminal stage of disease. clei, scanty cytoplasm, and indistinct cell neoplastic pseudostratified epithelium
bodies, as well as large, paucicellular, with an external (periodic acid-Schiff-
Microscopy fibrillar/neuropil-like areas, infrequently positive and collagen IV-positive) limit-
Rosettes are a frequent and characteris- containing neoplastic neurocytic and ing membrane, resembling the primitive
tic histopathological feature of C19MC- ganglion cells (618,801). In some cases, neural tube. On the luminal surface of
altered embryonal tumours. They are the neuropil has a fascicular quality. Hy- these tubules, cilia and blepharoblasts
multilayered and mitotically active struc- percellular areas contain numerous mi- are absent. Mitotic figures are abundant
tures consisting of pseudostratified neu- toses and apoptotic bodies. In the aggre- and tend to be located near the lumi-
roepithelium with a central, round, or gates of small cells, multilayered rosettes nal surface. In zones away from tubular
slit-like lumen. The lumen of the rosette are often present. In some cases, these and papillary structures, there are large
is either empty or filled with eosinophilic rosettes are observed as highly cellular sheets of poorly differentiated cells with

202 Embryonal tumours


hyperchromatic nuclei and a high nucle-
ar-to-cytoplasmic ratio. Clusters of multi-
layered rosettes may be seen here. Tu-
mour cells range from embryonal cells to
mature neurons and astrocytes. Rare tu-
mours display mesenchymal differentia-
tion or contain melanin pigment {36,301).
During tumour progression (local or dis-
tant recurrence), ETMRs with abundant
neuropil tend to show loss of neuropil-
like foci and may exhibit either extended
clusters of embryonal cells with multilay-
ered rosettes or prominent papillary and
tubular structures {1346,1349). In a few
embryonal tumours with abundant neuro-
pil and rosettes, post-treatment neuronal
Fig. 8.28 ETMR, ependymoblastoma morphology. Nests of poorly differentiated tumour cells and numerous
and glial/astrocytic maturation resem- multilayered rosettes.
bling a low-grade glioneuronal tumour
has been reported (78,611,1413}. Other
tumours show complete loss of key his-
topathological patterns of ETMR during
progression and instead resemble other
embryonal neoplasms (2771)

Electron microscopy
The tumour cells that make up the embry-
onal areas and rosettes have a compact
arrangement. They have large nuclei and
scanty cytoplasm with only a few orga-
nelles (1429,1997,2576}. The cells form-
ing rosettes are joined with junctional
complexes and show abortive cilia and a
few basal bodies at an apical site. On the
luminal side, there is an amorphous sur-
face coating but no true membrane. Tu- Fig. 8.29 ETMR, medulloepithelioma morphology. Papillary-like and tubular structures resembling the primitive neural
bular and papillary structures in the me- tube.
dulloepithelioma pattern show extensive
lateral junctions (zonulae adherentes)
and a basal lamina on the outer sur-
face, consisting of a distinct continuous
basement membrane. The neuropil-like
tumour component shows long cellular
processes containing numerous microtu-
bules as well as neurosecretory granules
in ganglion-like tumour cells.

lmmunophenotype
The primitive neuroepithelial component
of ETMR is intensely immunoreactive for
nestin and vimentin (618,801}. Multilay-
ered rosettes and tubular structures are
also positive for these markers and fre-
quently demonstrate an expression gra- Fig. 8.30 ETMR, medulloepithelioma morphology. Coexistence of tubular structures, trabecular structures, and
dient, with basal labelling greater than lu- multilayered rosettes.
minal labelling. The small-cell areas and
true rosettes may also show focal expres- strongly for synaptophysin, NFPs, and strong and diffuse nuclear immunoreac-
sion of cytokeratins, EMA, and CD99, but NeuN. lmmunoreactivity for GFAP high- tivity for INl1 throughout all components.
are usually negative for neuronal and glial lights scattered cells resembling reac- The Ki-67 proliferation index ranges from
markers. In contrast, the neuropil-like ar- tive astrocytes, but may also be present 20% to 80%, with the immunolabelling
eas (including neoplastic neurons) stain in some embryonal cells. ETMRs show highlighting cellular areas; rosettes and

Embryonal tumour with multilayered rosettes, C19MC-altered 203


tubular structures are also proliferative. FISH analysis applying a target probe to
Recently, the LIN28A protein has been the C19MC locus is a very helpful tool for
suggested as an immunohistochemical the diagnosis of ETMR in routine neuro-
diagnostic marker for ETMR (1348,1349, pathological practice {1346,1349,1796,
1967,2403l. LIN28A is a protein that binds 1798}. In a series of 97 histologically typi-
small RNAs, and it is known to be a nega- cal ETMRs, FISH revealed the prototypic
tive regulator of the let-7 family of microR- C19MC amplification in 96% of samples,
NAs, which may act as tumour suppressor irrespective of the histological type {1349}.
microRNAs. LIN28A is a conserved cyto- All supratentorial ETMRs demonstrated
plasmic protein, but may be transported to amplification of C19MC, whereas a small
the nucleus, where it seems to regulate the subset of infratentorial tumours disclosed
Fig. 8.31 C19MC-altered embryonal tumour with
translation and stability of mRNA. Strong only gain of 19q. Another study evaluated
multilayered rosettes. Amplification of the C19MC locus
and diffuse LIN28A cytoplasmic immu- at 19q13.42. the specificity of C19MC amplification in a
nostaining is found in ETMRs irrespective series of > 400 paediatric brain tumours
of their morphology (1349,2403l. LIN28A Genetic profile and found that the amplification was re-
immunoreactivity is most prominent and Various high-resolution cytogenetic tech- stricted to CNS embryonal tumours with
intense in multilayered rosettes and poorly niques (e.g. microarray-based compara- increased expression of LIN28A (2403l.
differentiated small-cell areas, as well as tive genomic hybridization, SNP arrays, Gene expression and methylation profiling
in the papillary and tubular structures of and methylation arrays) disclose recur- reveals a common molecular signature for
the medulloepithelioma pattern, whereas rent copy number aberrations in ETMRs: ETMR, regardless of morphological pat-
only single collections of positive cells are gains of chromosomes 2, 7q, and 11q tern or tumour location (1349,2403} Un-
observed within the neuropil-like tumour and loss of 6q l384,1349,2115,2403l. supervised clustering analysis showed
areas. However, LIN28A expression is These methods also reveal a focal high- that the methylation and gene expression
not specific for ETMR; it is also found in level amplicon at 19q13.42, spanning a profiles generated for ETMRs are clearly
some gliomas, atypical teratoid/rhabdoid 0.89 Mb region (1346,1349,1491,1798, distinct from other paediatric brain tu-
tumours, germ cell tumours, teratomas, 1962,2403} This amplicon is identified in mours, confirming their nosological status
and non-CNS neoplasms (2403,2723l the majority of ETMRs and seems to be a as a single CNS tumour entity (1349}.
Nevertheless, this immunohistochemical specific and sensitive diagnostic marker
marker can be very useful in support of a for these tumours. This unique amplicon Prognosis and predictive factors
diagnosis of ETMR. covers a cluster of microRNAs (a poly- ETMRs demonstrate rapid growth and
cistron) named C19MC, which is clearly are associated with an aggressive clini-
Cell of origin upregulated in these tumours {1491, cal course, with reported survival times
A shared molecular signature between 1962l This genetic aberration has not typically averaging 12 months after
the histological types of ETMR suggests been detected in other paediatric brain combination therapies {801,1349,2403l
that they may share a common origin, tumours (1346,1491}. A recent study also Many patients experience local tumour
such as a primitive cell population in the demonstrated complex rearrangements regrowth, and a smaller number de-
subventricular zone, with further evolu- at 19q13.42, as well as fusion of C19MC velop widespread tumour dissemination
tion into a wider range of morphological to the TTYH1 gene (1295l. As a result, the and systemic metastases. Gross total
appearances and mimics (1349}. promoter of TTYH1 drives expression of tumour resection and radiation may pro-
C19MC microRNAs. vide some benefit {10,50,1578,1762} Pa-
tient survival does not differ significantly
between the three histological types of
ETMR {1349}. Recurrent chromosomal
aberrations are also not associated with
clinical outcome. In extremely rare cases
with a long-term survival, post-treatment
neuronal differentiation has been pro-
posed as a favourable indicator of out-
come (78,611,1413l.

i � � i � i i i� n� i i
Fig. 8.32 Cytogenetic profile (methylation array) prototypic for ETMR: amplification of C19MC (arrow) and gain of
chromosome 2.

204 Embryonal tumours


················································ and span a broad histological spectrum.
However. many have the morphological
Embryonal tumour with features of tumours previously classified
multilayered rosettes, NOS as ependymoblastoma or embryonal tu-
mour with abundant neuropil and true ro-
Definition settes. These tumours are now regarded
An aggressive CNS embryonal tumour as patterns of ETMR. Medulloepithelio-
with multilayered rosettes, in which copy mas in which copy number at the 19q13
number at the 19q13 C19MC locus ei- C19MC locus shows no alteration or is
ther shows no alteration or has not been not tested are considered separately,
tested because they are considered genetically
Embryonal tumours with multilayered ro- distinct (see p. 207).
settes (ETMRs), NOS, can develop in the
cerebrum. brain stem, and cerebellum ICD-0 code 9478/3

Embryona! tumour with multilayered rosettes, C19MC-altered 205


Other CNS Mclendon R.
Judkins A.R.
Ng H.-K.
Huang A.
areas. Between 50% and 70% of all CNS
embryonal tumours contain calcifica-
embryonal tumours Eberhart CG.
Fuller G.N.
Kool M.
Pfister S.
tion. Oedema surrounding parenchymal
masses is not usually extensive. MRI ap-
Sarkar C. pearances can vary, depending on the
site of origin. The tumours are T1-hypoin-
tense relative to cortical grey matter. They
look similar on T2-weighted imaging,
Definition Epidemiology but cystic or necrotic areas are hyper-
A group of rare, poorly differentiated em- CNS embryonal neoplasms account for intense. There is contrast enhancement
bryonal neoplasms of neuroectodermal approximately 1% of brain tumours over- with gadolinium on T1-weighted imaging.
origin that lack the specific histopatho- all, but constitute 13% of neoplasms aris- Regions of haemorrhage (if present) are
logical features or molecular alterations ing in children aged 0-14 years {1863l. In T2-hypointense.
that define other CNS tumours. one study, they constituted about 3-5%
of all paediatric brain tumours (i.e. those Spread
Currently, the medulloblastoma, the em- in patients aqed s 18 years) {1967l. Metastatic dissemination is evident in
bryonal tumour with multilayered rosettes Shifting diagnostic criteria and nomen- 25-35% of CNS embryonal tumours at
(ETMR) characterized by genetic altera- clature make precise analysis of CNS presentation, mostly in the subarachnoid
tions at the C19MC locus on 19q.13.42, embryonal tumour epidemiology difficult, space, including the spinal canal. There-
and the atypical teratoid/rhabdoid tu- but at least one study has found an in- fore, diagnostic lumbar puncture for cytol-
mour (AT/RT) characterized by loss of crease in the incidence of medulloblas- ogy, as well as spinal MRI, is mandatory
SMARCB1 (INl1) or SMARCA4 (BRG1) tomas and other embryonal tumours over for all patients {1045). Extraneural me-
expression, represent genetically de- the past two decades {1896l. The Central tastases to the bone, liver, and cervical
fined embryonal tumours of the CNS. Brain Tumor Registry of the United States lymph nodes have been reported {167!.
There remain several CNS embryonal (CBTRUS) reports an overall average an-
tumours for which no genetic data have nual age-adjusted incidence rate in child- Macroscopy
yet facilitated a molecular classification. hood of 0.12 cases per 100 000 popula- CNS embryonal tumours are found most
Histologically, one resembles ETMR and tion, with a median patient age at onset commonly in the cerebrum, but can also
has been included alongside that tumour of 3.5 years and no significant sex pre- occur in the brain stem, spinal cord, or
as ETMR, NOS. The other resembles AT/ dominance {1862l. Although most CNS suprasellar region {805l. Those in the su-
RT and is designated CNS embryonal tu- embryonal tumours occur in infancy and prasellar region tend to be smaller than
mour with rhabdoid features. Others are childhood, some cases do arise in adults those in the cerebrum. The tumours are
included here: medulloepithelioma, CNS {157,806,1273,1968!. However, the inci- typically well-circumscribed pink mass-
neuroblastoma, CNS ganglioneuroblas- dence of adult CNS embryonal tumours es, and demarcation between tumour
toma, and CNS embryonal tumour, NOS. is difficult to determine because of their and brain ranges from indistinct to clear
rarity and lack of signature biomarkers. cut. The tumours are most often solid, but
The introduction of ETMR, C19MC-al- can contain cystic areas as well as areas
tered into this updated classification has Localization of haemorrhage and necrosis. They are
complicated the presentation of epidemi- CNS embryonal tumours are typically lo- soft unless they contain a prominent des-
ological and clinical data for embryonal cated in the cerebral hemispheres, with moplastic component, in which case they
tumours that are not defined genetically, rare examples occurring in the brain are firm and often have a tan colour.
because it is not yet clear whether such stem {741l and spinal cord {2643).
data for C19MC-altered ETMR differ sig- Cell of origin
nificantly from the broader category of Imaging The histogenesis of CNS embryonal tu-
CNS embryonal tumours. On CT, CNS embryonal tumours have mours other than medulloblastoma has
Data based on the broad category of similar appearances, regardless of site. been controversial for many years, and
CNS embryonal tumours and probably They are isodense to hyperdense, and the only point on which consensus has
applicable to medulloepithelioma, CNS density increases after injection of con- been achieved is that these poorly dif-
neuroblastoma, CNS ganglioneuroblas- trast material. They can present as solid ferentiated tumours arise from primitive
toma, and CNS embryonal tumour, NOS, masses or may contain cystic or necrotic neuroepithelial cells {1289,2171}.
are presented in this overarching section
of the chapter ahead of the definitions Table 8.03 Biomarkers useful in the characterization of small cell, embryonal-appearing tumours
and specific information on microscopy Associated tumour
Biomarker
and immunophenotype for each tumour.
C19MC amplification or LIN28Aexpression Embryonal tumour with multilayered rosettes
Grading SMARCB1 loss or SMARCA4 loss Atypical teratoidlrhabdoid tumour
All CNS embryonal tumours correspond H3 K27 and G34 mutations Paediatric-type glioblastomas
histologically to WHO grade IV
C11orf95-RELA fusion gene or L 1CAM expression Supratentorial ependymoma
IDH1 or IDH2 mutation Adult-type diffuse gliomas

206 Embryonal tumours


prognosis and predictive factors
overall, CNS embryonal tumours show
aggressive clinical behaviour and have
a very poor prognosis, with multiple local
relapses and widespread leptomeningeal
dissemination. Overall survival at 5 years
in paediatric patients with CNS embryonal
tumours is reported to be 29-57% (43,
440,478,590,1173,2082,2558}, which is
poorer than the survival rate for children
with medulloblastoma (809,2558) Factors
associated with poor prognosis in paedi-
atric patients include young age, meta-
static disease, and incomplete resection
(440,478,806,810, 1173,1474,2082,2558}.

················································
Medulloepithelioma
immunolabelling is uniformly high among generally reactive astrocytes. Groups of
Definition embryonal cells. LIN28A is expressed by neurocytic cells express synaptophysin
A CNS embryonal tumour with a promi- medulloepitheliomas that lack C19MC or NeuN. Ki-67 immunolabelling is high
nent pseudostratified neuroepithelium amplification (2403l in embryonal cells, but lower elsewhere.
that resembles the embryonic neural
tube in addition to poorly differentiated
neuroepithelial cells.
Even though medulloepithelioma is a CNS neuroblastoma CNS ganglioneuroblastoma
very rare tumour, a significant propor-
tion of those analysed genetically have Definition Definition
not shown C19MC alterations (2403). A CNS embryonal tumour characterized A CNS embryonal tumour characterized
Although a diagnostic genetic signature by poorly differentiated neuroepithelial by poorly differentiated neuroepithelial
has yet to be defined for such tumours, cells, groups of neurocytic cells, and a cells and groups of neurocytic and gan-
they are grouped as a tumour entity, dis- variable neuropil-rich stroma. glion cells.
tinct from ETMR. This exceedingly rare tumour shows a This rare tumour shows varying degrees
distinctive pattern of differentiation, not of neuronal differentiation, but dystrophic
ICD-0 code 9501/3 unlike some peripheral neuroblastomas. ganglion cells are a prominent element.

Microscopy ICD-0 code 9500/3 ICD-0 code 9490/3


Medulloepithelioma consists of sheets of
embryonal cells interspersed to a vari- Microscopy Microscopy
able extent by tubular and trabecular ar- Zones of neurocytic differentiation are Varying degrees of neuronal differentia-
rangements of a neoplastic pseudostrati- found among sheets of densely packed tion characterize this tumour, which also
fied neuroepithelium, which appears primitive embryonal cells. Neurocytic dif- contains sheets of primitive embryonal
similar to embryonic neural tube. This ferentiation manifests as cells with slightly cells. Neurocytic and ganglion cells, the
has a periodic acid-Schiff-positive ex- larger nuclei and variably distinct cyto- latter occasionally binucleated, are usu-
ternal limiting membrane, and its luminal plasm set against a faintly fibrillar matrix at ally present as small groups, rather than
surface lacks cilia and blepharoblasts. lower density than the embryonal cells. Ar- dispersed diffusely among the embryo-
Neural differentiation may occasionally chitectural features include Homer Wright nal cells. Dispersed cells with a neuronal
be found in the form of cells with a dys- rosettes, palisading patterns of cells, and morphology and appearing to form a
trophic neuronal or astrocytic morphol- regions of necrosis with granular calcifica- pattern are more likely to be entrapped
ogy. Mitotic figures are readily found. tion. Exceptionally, a Schwannian stroma neurons. Mitotic figures and apoptotic
may be present. bodies are readily found among embryo-
lmmunophenotype nal cells. Architectural features include
Embryonal cells in medulloepithelioma lmmunophenotype Homer Wright rosettes, palisading pat-
rarely show immunoreactivity for neu- The embryonal cells may be immunoneg- terns of cells, and regions of necrosis
ronal markers, such as synaptophysin ative for neural markers, such as synap- with granular calcification.
and NFPs, and GFAP-positive tumour tophysin or GFAP, but some might show
cells are exceptional. The neuroepithe- weak expression of synaptophysin. Very lmmunophenotype
lium may show patchy expression of cy- rarely, GFAP expression is found in a few Embryonal cells may be immunon-
tokeratins and, less often, EMA. Ki-67 tumour cells, but GFAP-positive cells are egative for neural markers, such as

Other CNS embryonal tumours 207


.... :' - .
Fig. 8.34 CNS ganglioneuroblastoma. A Nodule of admixed neurocytic and ganglion cells among embryonal cells. B Embryonal cells next to large cells with variable neuronal
differentiation embedded in a fibrillary stroma.

synaptophysin or GFAP, but some usually used. Embryonal tumours such as me- cells with round to oval nuclei and a high
show weak expression of synaptophysin. dulloepitheliomas, ependymoblastomas, nuclear-to-cytoplasmic ratio. Commonly
Groups of neurocytic and ganglion cells and embryonal tumours with abundant encountered features include frequent
express synaptophysin, NFPs, MAP2, neuropil and true rosettes, which exhibit mitoses and apoptotic bodies. More
and NeuN. Ki-67 immunolabelling is high alterations at the 19q13 C19MC locus and tightly packed regions with angular or
in embryonal cells, but lower elsewhere. were previously included within the broad moulded cells may also be identified,
CNS PNET designation, are now consid- and Homer Wright rosettes can be found.
ered to be a distinct genetically defined Necrosis and vascular endothelial prolif-
entity, called embryonal tumour with eration may also be seen. Calcification
CNS embryonal tumour, NOS multi layered rosettes (ETMR), C19MC- is relatively common within degenerative
altered. Molecular genetic advances regions. A fibrous stroma is occasionally
Definition now permit the improved classification present and can vary from a delicate lob-
A rare, poorly differentiated embryonal of other CNS embryonal tumours as ular framework to dense fibrous cords.
neoplasm of neuroectodermal origin that well; for example, the diagnosis of atypi- Some variants of CNS embryonal tumour
lacks the specific histopathological fea- cal teratoid/rhabdoid tumour is made by display distinctive architectural or cyto-
tures or molecular alterations that define demonstrating mutation of SMARCB1 logical features. The latter often mani-
other CNS tumours. or SMARCA4 or loss of expression of fests as neuronal differentiation.
SMARCB1 (INl1) or SMARCA4 (BRG1).
ICD-0 code 9473/3 Other high-grade undifferentiated neural lmmunophenotype
neoplasms that are composed of small CNS embryonal tumour can show vari-
CNS embryonal tumours, NOS, are uniform cells and enter the differential di- able expression of divergent neuroepithe-
characterized by poorly differentiated agnosis of CNS embryonal tumour often lial markers, including proteins associated
neuroepithelial cells with a variable ca- have their own molecular signatures (see with both glial differentiation (GFAP) and
pacity for divergent differentiation along Table 8 03, p. 206). neuronal differentiation (synaptophysin,
neuronal, astrocytic, myogenic, or mel- NFP, and NeuN) {875). Expression of these
anocytic lines. CNS embryonal tumours Malignant gliomas with primitive neuronal markers is typically present in a thin rim
can have histological features overlap- components, though rare, constitute of cytoplasm, although NeuN is nuclear.
ping those of other brain tumours, many a separate entity and were initially de- Antibodies that recognize GFAP may also
of which have been reclassified from this scribed in adults {1099,1625,1946,2343) label reactive astrocytes. However, poorly
group through the identification of unique This entity is considered a variant of glio- differentiated regions of the neoplasm
molecular biomarkers. blastoma rather than a CNS embryonal often predominate, in which no immuno-
The current definition of CNS embryonal tumour (see p. 33). histochemical signs of neural differentia-
tumour, NOS, is more circumscribed than tion are apparent. The Ki-67 proliferation
in prior WHO classifications, in which Microscopy index is typically variable, but a very high
the umbrella designation "CNS primitive CNS embryonal tumours are poorly dif- growth fraction, often with > 50% of immu-
neuroectodermal tumour (PNET)" was ferentiated neoplasms composed of nopositive tumour cells, is usual.

208 Embryonal tumours


Atypical teratoid/rhabdoid tumour Judkins A.Fl.
Eberhart C.G.
Wesseling P.
Hasselblatt M.

Definition account for :::>: 10% of all CNS tumours in


A malignant CNS embryonal tumour infants {192).
composed predominantly of poorly dif-
ferentiated elements and frequently in- Age and sex distnbution
cluding rhabdoid cells, with inactivation AT/RT is a paediatric tumour. It most often
of SMARCB1 (INl1) or (extremely rarely} presents in patients aged < 3 years and
SMARCA4 (BRG1). rarely in children aged > 6 years. There
Atypical teratoid/rhabdoid tumours (AT/ is a male predominance, with a male-to-
RTs) occur most frequently in young chil- female ratio of 1.6-2:1 {1002,2530). AT/
dren. Neoplastic cells demonstrate his- RT rarely occurs in adults {2063).
tological and immunohistochemical evi-
dence of polyphenotypic differentiation Localization
along neuroectodermal, epithelial, and In two large series of paediatric cases,
mesenchymal lines. Diagnosis of AT/RT the ratio of supratentorial to infratentorial
requires demonstration of inactivation of tumours was 4:3 {1002,2530) Supraten-
SMARCB1 or, if intact, SMARCA4 genes, torial tumours are often located in the
either by routine immunohistochemical cerebral hemispheres and less frequent- Fig. 8.36 AT/RT Axial T1-weighted contrast-enhanced
staining for their proteins or by other ap- ly in the ventricular system, suprasellar image demonstrates a heterogeneously enhancing left
cerebellar mass.
propriate means. Tumours with this mor- region, or pineal gland. lnfratentorial tu-
phology but lacking this molecular genet- mours can be located in the cerebellar
ic confirmation should be classified as hemispheres, cerebellopontine angle,
CNS embryonal tumours with rhabdoid and brain stem, and are relatively preva-
features (p, 212). lent in the first 2 years of life. Infrequently,
AT/RT arises in the spinal cord. Seeding
ICD-0 code 9508/3 of AT/RT via the cerebrospinal fluid path-
ways is common and is found in as many
Grading as one quarter of all patients at presenta-
AT/RT corresponds histologically to WHO tion {1002}. lnfratentorial localization is
grade IV very rare in adult patients diagnosed with
AT/RT {646).
Epidemiology Fig. 8.37 AT/RT with multiple haemorrhages, arising in
In several large series, AT/RTs account- Clinical features the right cerebellopontine angle.
ed for 1-2% of all paediatric brain tu- The clinical presentation is variable, de-
mours {2116,2783). AT/RT is very rare in pending on the age of the patient and the
adult patients {2063). However, due to location and size of the tumour. Infants, variably contrast-enhancing, and lepto-
the preponderance of cases in children in particular, present with non-specific meningeal dissemination can be seen in
aged < 3 years, AT/RTs are estimated to signs of lethargy, vomiting, and/or failure as many as a quarter of cases at presen-
to thrive. More specific problems include tation {1659)
head tilt and cranial nerve palsy, most
Males 12
commonly sixth and seventh nerve pare- Macroscopy
11 sis. Headache and hemiplegia are more These tumours (and their deposits along
10
commonly reported in children aged the cerebrospinal fluid pathways) gen-
> 3 years. erally have a gross appearance similar
to that of medulloblastoma and other
Imaging CNS embryonal tumours. They tend to
CT and MRI findings are similar to those be soft, pinkish-red, and often appear
in patients with other embryonal tumours. to be demarcated from adjacent paren-
AT/RTs are isodense to hyperintense on chyma. They typically contain necrotic
10 15 20
FLAIR images and show restricted diffu- foci and may be haemorrhagic. Those
20 15 10
Number of cases Number of cases sion. Cystic and/or necrotic regions are with significant amounts of mesenchy-
Fig. 8.35 Age and sex distribution of atypical teratoid/ apparent as zones of heterogeneous mal tissue may be firm and tan-white
rhabdoid tumour, based on 73 cases (1002,2530). signal intensity. Almost all tumours are in some regions. Tumours arising in the

Atypical teratoid/rhabdord tumour 209


.,

l -
Fig. 8.38 Atypical teratoid/rhabdoid tumour. A Rhabdoid cells with vesicular chromatin, prominent nucleoli, and eosinophilic globular cytoplasmic inclusions. B Tumour cells with
abundant, pale eosinophilic neoplasm.

cerebellopontine angle wrap themselves perikaryon {202,916). A frequently en- necrosis and haemorrhage are common-
around cranial nerves and vessels and countered artefact in these cells is cyto- ly encountered in these tumours.
invade brain stem and cerebellum to plasmic vacuolation. Rhabdoid cells may
various extents. Rarely, AT/RT can show be arranged in nests or sheets and often lmmunophenotype
bony involvement {2697). have a jumbled appearance. However. AT/RTs demonstrate a broad spectrum
these cells are the exclusive or predomi- of immunohistochemical reactivities that
Microscopy nant histopathological finding in only a align with their histological diversity
AT/RTs are heterogeneous lesions that minority of cases. However. the rhabdoid cells character-
can often be difficult to recognize solely Most tumours contain variable compo- istically demonstrate expression of EMA,
on the basis of histopathological criteria nents with primitive neuroectodermal, SMA. and vimentin. lmmunoreactivities
{324,2172). The most striking feature in mesenchymal, and epithelial features. A for GFAP. NFP. synaptophysin. and cy-
many cases is a population of cells with small-cell embryonal component is the tokeratins are also commonly observed.
classic rhabdoid features. eccentrically most commonly encountered, present in In contrast, germ cell markers and mark-
located nuclei containing vesicular chro- two thirds of all tumours. Mesenchymal ers of skeletal muscle differentiation are
matin, prominent eosinophilic nucleoli. differentiation is less common and typi- not typically expressed. lmmunohisto-
abundant cytoplasm with an obvious eo- cally presents as areas with spindle cell chemical staining for expression of the
sinophilic globular cytoplasmic inclusion. features and a basophilic or mucopoly- SMARCB1 protein (INl1) has been shown
and well-defined cell borders. The cells saccharide-rich background. Epithelial to be a sensitive and specific test for the
typically fall along a spectrum ranging differentiation is the least common histo- diagnosis of AT/RT. In normal tissue and
from those with a classic rhabdoid phe- pathological feature. It can take the form most neoplasms. SMARCB1 is a consti-
notype to cells with less striking nuclear of papillary structures. adenomatous ar- tutively expressed nuclear protein; in AT/
atypia and large amounts of pale eosino- eas. or poorly differentiated ribbons and RT, there is loss of nuclear expression of
philic cytoplasm. The cytoplasm of these cords. A myxoid matrix occurs uncom- SMARCB1 {1192). Paediatric CNS em-
cells has a finely granular. homogene- monly; in cases where this is the predomi- bryonal tumours without rhabdoid fea-
ous character or may contain a poorly nant histopathological pattern. distinction tures. but with loss of SMARCB1 expres-
defined, dense. pink body resembling from choroid plexus carcinoma can be sion in tumour cells. qualify as AT/RTs
an inclusion. Ultrastructurally, rhabdoid challenging. Mitotic figures are usually as well {918}. Rare SMARCB1-deficient
cells typically contain whorled bundles of abundant. Broad areas of geographical non-rhabdoid tumours forming cribriform
intermediate filaments filling much of the strands, trabeculae. and well-defined

•' ,_· -

Fig. 8.39 Atypical teratoid/rhabdoid tumour. A Gland-like component. B Spindle cell morphology. C Mucopolysaccharide-rich background.

210 Embryonal tumours


urfaces are called cribriform neuroepi-
�helial tumours {964). Cribriform neuroep-
'thelial tumour is most likely an epithelioid
�ariant of AT/RT but is characterized by
a relatively favourable prognosis {92,612,
964). Some authors have reported inac-
tivation of SMARCB1 in choroid plexus
carcinomas, but others believe that such
tumours have histopathological features
justifying a diagnosis of AT/RT, and that
classic choroid plexus carcinomas do not
lose SMARCB1 expression (802,1190}.
Rarely, tumours with clinical and morpho-
logical features of AT/RT and retained
sMARCB1 expression are encountered.
Loss of nuclear expression of SMARCA4
(BRG1) is rarely seen in such cases
(959). AT/RTs in children have marked
proliferative activity; often having a Ki-67
proliferation index of > 50% {1016) Lim-
ited data are available on adult patients, •� • •
but in some cases the proliferation index Fig. 8.40 Atypical teratoid/rhabdoid tumour. A Strong expression of vimentin. B Membranous and cytoplasmic
is significantly lower {1550). expression of EMA. C Expression of GFAP (brown) and NFP (red). D Loss of expression of SMARCB1 (INl1) in nuclei
of tumour cells, with retained expression in intratumoural blood vessels.
Cell of origin
The histogenesis of rhabdoid tumours is expression {192). Homozygous deletions diagnosed in a newborn and his mother,
unknown. Neural, epithelial, and mesen- of the SMARCB1 locus are detected in respectively, providing a link between AT/
chymal markers can all be expressed, 20-24% of cases (192,2530}. In other RT and small cell carcinoma of the ovary
and given these tumours' association cases, one SMARCB1 allele is mutated of hypercalcaemic type (728)
with young children, it has been sug- and the second allele is lost by deletion The specific functions of SMARCB1 and
gested that they derive from pluripotent or mitotic recombination. Rare tumours SMARCA4 and their role in malignant
fetal cells (247,1864). Meningeal, neural demonstrate two coding sequence muta- transformation are not entirely clear, but
crest, and germ cell origins have also tions. Nonsense and frameshift mutations loss of SMARCB1 seems to result in
been proposed ( 451, 1887, 2172}. AT/RTs that are predicted to lead to truncations widespread but specific deregulation of
arising in the setting of ganglioglioma, of the protein are identified in the major- genes and pathways associated with cell
or other low-grade CNS lesions, suggest ity of these cases (192). Localization of cycle, differentiation, and cell survival
the possibility of progression from other mutations within the SMARCB1 gene (1458,2579). Cell cycle regulatory genes
tumour types {55,1157). seems to vary somewhat between rhab- that are overexpressed in AT/RT include
doid tumours arising at various sites in CCND1 and AURKA {2858). Loss of
Genetic profile the body, and exons 5 and 9 contain hot- SMARCB1 leads to transcriptional activa-
AT/RTs can occur sporadically or as spots in CNS AT/RT. Because expression tion of EZH2, as well as repression and
part of a rhabdoid tumour predisposition of SMARCB1 is sometimes decreased increased H3K27me3 of polycomb gene
syndrome (192). Mutation or loss of the in AT/RT in the absence of genetic al- targets as part of the broader SWI/SNF
SMARCB1 locus at 22q11.2 is the ge- terations, its promoter was analysed in modulation of the polycomb complex to
netic hallmark of this tumour {194,2649). 24 cases, using bisulfite sequencing and maintain the epigenome {52,2761} The
Whole-exome sequencing demonstrates methylation-sensitive PCR, but no evi- Hippo signalling pathway is involved in
that the genomes of AT/RTs are remark- dence of hypermethylation was detected the detrimental effects of SMARCB1 de-
ably simple; they have an extremely low (2853). No relationship between the type ficiency, and its main effector, YAP1, is
rate of mutations, with loss of SMARCB1 of SMARCB1 alteration and outcome overexpressed in AT/RT {1145).
being the primary recurrent event. The could be established.
SMARCB1 protein is a component of the Very infrequently, tumours are encoun- Genetic susceptibility
mammalian SWI/SNF complex, which tered that have features of AT/RT and in- In familial cases of rhabdoid tumours, i.e.
functions in an ATP-dependent manner tact SMARCB1 protein expression; these in rhabdoid tumour predisposition syn-
to alter chromatin structure (2137). Loss may instead have mutation and inactiva- drome 1 (involving the SMARCB1 gene)
of SMARCB1 expression at the protein tion of SMARCA4, another component and rhabdoid tumour predisposition syn-
level is seen in almost all AT/RTs, and of the SWI/SNF complex {2293} These drome 2 (involving the SMARCA4 gene),
most of the tumours have detectable tumours are associated with very young unaffected adult carriers and gonadal
deletions or mutations of SMARCB1; the patient age and a poor prognosis {962). mosaicism have been identified {962,
other cases exhibit loss of SMARCB1 In one family with a germline SMARCA4 1133,2327). Because the risk of germline
function due to reduced RNA or protein mutation, AT/RT and ovarian cancer were mutations has been reported to be z 33%

Atypical teratoid/rhabdoid tumour 211


in SMARCB1-deficient AT/RT and may among children on standard-dose ther- ················································
be substantially higher in SMARCA4- apy 11414). In contrast, the Children's CNS embryonal tumour with
deficient tumours 1962}, it is important to Cancer Group study CCG-9921, with rhabdoid features
perform molecular genetic studies in all standard-dose chemotherapy alone, re-
newly diagnosed cases l192,962f. ported a 5-year event-free survival rate of Definition
14% ± 7% and an overall survival rate of A highly malignant CNS embryonal tu-
Prognosis and predictive factors 29% ± 9% 1809). mour composed predominantly of poor-
Although overall the prognosis of AT/RTs ly differentiated elements and including
is poor, emerging data from a series of The results of global genomic and tran- rhabdoid cells, either with expression of
clinical trials have demonstrated that not scriptional analysis of AT/RT suggest that SMARCB1 (INl1) and SMARCA4 (BRG1)
all AT/RTs share the same uniformly dis- these tumours may constitute at least two or in which SMARCB1 and SMARCA4
mal prognosis. Retrospective analysis of different molecular classes, with distinct status cannot be confirmed.
children with AT/RTs who were enrolled outcomes. One study suggested that In CNS embryonal tumour with rhabdoid
at the German HIT trial centre between tumours with enrichment of neurogenic features, neoplastic cells demonstrate
1988 and 2004 demonstrated a 3-year or forebrain markers are associated with histological and immunohistochemical
overall survival rate of 22% and an event- supratentorial location, more favourable evidence of polyphenotypic differen-
free survival rate of 13%, and also identi- response to therapy, and better long- tiation along neuroectodermal, epithelial,
fied a subset of patients (14%) who were term survival, whereas AT/RT with mes- and mesenchymal lines.
long-term event-free survivors 12665). In enchymal lineage markers are more fre- Thus, the pathological features of these
a large prospective trial incorporating quently infratentorial and associated with tumours are the same as those for AT/
chemotherapy in an intensive multimodal worse long-term survival 12569). It may RTs. Because of the extreme rarity of
treatment approach that included radia- ultimately be possible to risk-stratify AT/ CNS embryonal tumour- with rhabdoid
tion, a 2-year progression-free survival RTs on the basis of molecular group, lo- features, no data are available to deter-
rate of 53% ± 13% and an overall survival cation, extent of resection, and disease mine whether its epidemiological or clini-
rate of 70% ± 10% were found 1433). Fur- stage. However, even for the favourable cal characteristics are significantly differ-
ther demonstrating the overall efficacy of subgroup this remains an aggressive ent from those of AT/RTs
high-dose chemotherapy with radiation, disease, with 5-year progression-free
a retrospective review of the Canadian and overall survival rates of 60% and re- ICD-0 code 9508/3
Brain Tumour Consortium data found, currence occurring in about one third of
that children who received high-dose patients. Grading
chemotherapy, and in some cases ra- CNS embryonal tumour with rhabdoid
diation, had a 2-year overall survival rate features corresponds histologically to
of 60% ± 12.6% versus 21.7% ± 8.5% WHO grade IV

212 Embryonal tumours


CHAPTER 9

Tumours of the cranial and paraspinal nerves


Schwannoma
Melanotic schwannoma
Neurofibroma
Perineurioma
Hybrid nerve sheath tumours
Malignant peripheral nerve sheath tumour
.,

Schwannoma Antonescu C.R.


Louis D.N.
Hunter S.
Perry A.
Reuss D.E.
Stemmer-Rachamimov A.O.

Definition
A benign, typically encapsulated nerve
sheath tumour composed entirely of
well-differentiated Schwann cells, with
loss of merlin {the NF2 gene product) ex-
pression in conventional forms.
Schwannomas are solitary and sporadic
in the vast majority of cases, can affect
patients of any age, and follow a benign
clinical course. Multiple schwannomas
are associated with neurofibromatosis
type 2 (NF2) and schwannomatosis.

ICD-0 code 9560/0

Grading Fig. 9.02 A Vestibular schwannoma. Postcontrast T1-weighted MRI showing the typical ice-cream-cone shape of a
Conventional, non-melanotic schwanno- vestibular schwannoma, with the cone representing the portion within the internal auditory canal and the scoop of ice
mas and their variants correspond histo- cream the portion in the cerebellopontine angle. B Paraspinal schwannoma. Postcontrast T1-weighted MRI showing
logically to WHO grade I. both intraspinal extramedullary and extraspinal components, with a point of constriction at the nerve exit.

Synonyms
Neurilemoma; neurinoma

Epidemiology
Schwannomas account for 8% of all intra-
cranial tumours, 85% of cerebellopontine
angle tumours, and 29% of spinal nerve
root tumours {2204). Approximately 90%
of cases are solitary and sporadic and
4% arise in the setting of NF2. Of the 5%
of schwannomas that are multiple but not
associated with NF2 {375). some may be
associated with schwannomatosis {1558).
Patients of any age can be affected, Fig. 9.03 Schwannoma. A External surface showing the parent nerve of origin and a variably translucent capsule.
B On cut surface, schwannomas often show a glistening to mucoid appearance, variable cystic degeneration, and
but paediatric cases are rare. The peak
yellow spots reflecting collections of xanthomatous macrophages.
incidence is in the fourth to sixth dec-
ades of life. Most studies show no sex
predilection: but some series have shown show a strong predilection for the eighth
a female predominance among intracra- cranial nerve in the cerebellopontine an-
nial tumours {526,1613,2204). Cerebral gle, particularly in NF2 {1893}. They arise
90-100
intraparenchymal schwannomas are as- at the transition zone between central
80-89

70-79
sociated with a younger patient age and and peripheral myelination and affect the
60-69
a male predominance {375). Schwan- vestibular division. The adjacent coch-
50-59 nomas of spinal cord parenchyma are lear division is almost never the site of
4
30-39
1).4

20-29
9

1111 too rare for their epidemiology to be as-


sessed {996).
origin. This characteristic location, which
is not shared by neurofibromas or malig-
nant peripheral nerve sheath tumours,
- 10-19
0-9
Localization results in diagnostically helpful enlarge-
80 60 40 20 20 40 60 80 The vast majority of schwannomas occur ment of the internal auditory meatus on
Number of cases Number of cases outside the CNS. Peripheral nerves in the neuroimaging. lntralabyrinthine schwan-
Fig. 9.01 Age and sex distribution of schwannomas, based skin and subcutaneous tissue are most nomas are uncommon {1763}. lntraspinal
on 582 patients treated at the University Hospital Zurich. often affected. lntracranial schwannomas schwannomas show a strong predilection

214 Tumours of the cranial and paraspinal nerves


of the tumour typically shows light-tan
glistening tissue interrupted by bright yel-
low patches, with or without cysts and
haemorrhage. Infarct-like necrosis related
to degenerative vascular changes may
be evident in sizable tumours, such as the
giant lumbosacral, retroperitoneal, and
pelvic schwannomas that often erode ad-
jacent vertebral bodies.

Fig. 9.04 Vestibular schwannoma. A Large vestibular schwannoma compressing neighbouring cerebellar structures Microscopy
and the brain stem. Note the pressure-induced cyst formation in the cerebellar white matter. B Schwannoma Conventional schwannoma is composed
(arrowhead) originating from the left vestibulocochlear nerve (VCN). entirely of neoplastic Schwann cells, oth-
er than inflammatory cells, which may be
tor sensory nerve roots; motor and auto- hallmark of NF2. Pain is the most com- focally numerous. Two basic architectural
nomic nerves are affected far less often. mon presentation for schwannomas in patterns, in varying proportions, are typi-
occasional CNS schwannomas are not patients with schwannomatosis. cally present: areas of compact, elongat-
associated with a recognizable nerve; ed cells with occasional nuclear palisad-
these include approximately 70 reported Imaging ing (Antoni A pattern) and less cellular,
cases of spinal intramedullary schwan- MRI shows a well-circumscribed, some- loosely textured cells with indistinct pro-
nomas and 40 cases of cerebral paren- times cystic and often heterogene- cesses and variable lipidization (Antoni B
chymal or intraventricular schwannomas ously enhancing mass {375). Vestibular pattern). A retiform pattern is uncommon-
!375,528,1278,2722!. Dural examples schwannomas often display a classic ly seen. The Schwann cells that make up
are rare (77l. Peripheral nerve schwan- ice-cream-cone sign, with the tapered the tumour have moderate quantities of
nomas, unlike neurofibromas, tend to be intraosseous cone exiting the internal eosinophilic cytoplasm without discern-
attached to nerve trunks, most often in- auditory canal and expanding out to a ible cell borders. Antoni A tissue features
volving the head and neck region or flex- rounded cerebellopontine angle mass. normochromic spindle-shaped or round
or surfaces of the extremities. Visceral Masses in paraspinal and head and neck nuclei approximately the size of those of
schwannomas are rare, as are osseous sites may be associated with bone ero- smooth muscle cells, but tapered instead
examples (1662,2038}. sion, which is sometimes evident on plain of blunt-ended. In Antoni B tissue, the
X-ray. Paraspinal examples may also tumour cells have smaller, often round to
Clinical features show a dumbbell shape, with a point of ovoid nuclei. The Antoni A pattern con-
Peripherally situated schwannomas may constriction at the neural exit foramen. sists of densely packed spindled tumour
present as incidental (asymptomatic) cells arranged in fascicles running in dif-
paraspinal tumours, as spinal nerve tu- Macroscopy ferent directions. The tumour nuclei may
mours with radicular pain and signs of Most schwannomas are globoid masses show a tendency to align in alternating
nerve root I spinal cord compression, or measuring < 10 cm. With the exception parallel rows, forming nuclear palisades.
as eighth cranial nerve tumours with re- of rare examples arising at intraparenchy- When marked, nuclear palisades are re-
lated symptoms (including hearing loss, mal CNS sites, viscera, skin, and bone, ferred to as Verocay bodies. All schwan-
tinnitus, and occasional vertigo). Mo- they are usually encapsulated. In periph- noma cells show a pericellular reticulin
tor symptoms are uncommon because eral tumours, a nerve of origin is identified pattern corresponding to surface base-
schwannomas favour sensory nerve in less than half of cases, often draped ment membranes. In Antoni B areas,
roots. Bilateral vestibular tumours are the over the tumour capsule. The cut surface the tumour cells are loosely arranged.

Schwannoma 215
s:

B Diffuse s100

Collections of lipid-laden cells may be Ancient schwannoma composed exclusively or predominantly


present within either Antoni A or An- Nuclear pleomorphism, including bi- of Antoni A tissue and devoid of well-
toni B tissue. Schwannoma vasculature zarre forms with cytoplasmic-nuclear formed Verocay bodies !2786). The most
is typically thick-walled and hyalinized inclusions, and the occasional mitotic common location of cellular schwan-
Dilated blood vessels surrounded by figure may be seen, but should not be noma is at paravertebral sites in the pel-
haemorrhage are common. In the setting misinterpreted as indicating malignancy. vis, retroperitoneum, and mediastinum
of NF2, vestibular schwannomas may Analogous to other schwannomas, there !2786}. Cranial nerves, especially the fifth
show a predominance of Antoni A tissue, is typically diffuse S100 and collagen IV and eighth, may be affected !376). The
whorl formation, and a lobular grape-like positivity, extensive SOX10 expression, clinical presentation of cellular schwan-
growth pattern on low-power examina- and a low Ki-67 proliferation index in the noma is similar to that of conventional
tion !2387). Molecular data suggest that enlarged atypical cells. schwannoma, but the histological fea-
these are polyclonal and likely constitute tures of hypercellularity, fascicular cell
the confluence of multiple small schwan- growth, occasional nuclear hyperchro-
nomas !578). Malignant transformation, Cellularschwannoma masia and atypia, and low to medium
less often microscopic than extensive mitotic activity (usually < 4 mitoses per
and transcapsular !1628,2789}. rarely ICD-0 code 9560/0 10 high-power fields, but occasionally
occurs in conventional schwannomas. as many as � 10 mitoses per 10 high-
The cellular schwannoma variant is de- power fields) may lead to a misdiagno-
fined as a hypercellular schwannoma sis of malignancy (malignant peripheral

216 Tumours of the cranial and paraspinal nerves


nerve sheath tumour) (1924). In one se- The tumour has a rare association with NF2 gene as a tumour suppressor inte-
ries. cellular schwannomas were found NF2 (but not with NF1) and has also been gral to the formation of sporadic schwan-
to differ from malignant peripheral nerve noted to occur in patients with schwanno- nomas {1117,2314). The NF2 gene and
sheath tumours in that the schwannomas matosis (1108). Cranial and spinal nerves the merlin protein (also called schwan-
had 8chwannian whorls, a peritumoural are usually spared. nomin) that it encodes are discussed in
capsule, subcapsular lymphocytes, detail in the chapter on neurofibroma-
macrophage-rich infiltrates, and an ab- Electron microscopy tosis type 2. Inactivating mutations of
sence of fascicles, as well as strong, Ultrastructural features are diagnostic. the NF2 gene have been detected in
widespread expression of 8100, 80X10, The cells have convoluted, thin cytoplas- approximately 60% of all schwanno-
neurofibromin, and CDKN2A (p16), with mic processes that are nearly devoid of mas {204,1116,1117,2190,2575). These
a Ki-67 proliferation index < 20% in most pinocytotic vesicles but are lined by a genetic events are predominantly small
examples (1924). Cellular schwanno- continuous basal lamina. 8tromal long- frameshift mutations that result in truncat-
mas are benign. Although recurrences spaced collagen (called Luse bodies) ed protein products (1536). Although not
are seen, notably in intracranial, spinal, is a common finding in conventional described for exons 16 or 17, mutations
and sacral examples (376). no cellular schwannoma but less so in the cellular occur throughout the coding sequence
schwannoma is known to have metasta- variant. of the gene and at intronic sites. In most
sized or to have followed a clinically ma- cases, such mutations are accompanied
lignant, fatal course. Only two examples lmmunophenotype by loss of the remaining wildtype allele
of cellular schwannoma, one associated The tumour cells strongly and diffusely on chromosome 22q. Other cases dem-
with NF2, have been reported to have un- express 8100 protein {2724); often ex- onstrate loss of chromosome 22q in the
dergone malignant transformation (82). press 80X10, LEU?, and calretinin {707, absence of detectable NF2 gene muta-
1924); and may focally express GFAP tions. Nevertheless, loss of merlin ex-
{1635). All schwannoma cells have sur- pression, shown by western blotting or
Plexfformschwannoma face basal lamina, so membrane staining immunohistochemistry, appears to be a
for collagen IV and laminin is extensive universal finding in schwannomas, re-
ICD-0 code 9560/0 and most commonly pericellular. Low- gardless of their mutation or allelic status
level p53 protein immunoreactivity may {1014,1074,2222). This suggests that ab-
The plexiform schwannoma variant is be seen, particularly in cellular schwan- rogation of merlin function is an essential
defined as a schwannoma growing in a nomas {376). NFP-positive axons are step in schwannoma tumorigenesis. Loss
plexiform or multinodular manner and can generally absent, but small numbers may of chromosome 22 has also been noted
be of either conventional or cellular type be encountered in schwannomas, par- in cellular schwannoma (1522). Other ge-
(20,2787) Presumably involving multiple ticularly in tumours associated with NF2 netic changes are rare in schwannomas,
nerve fascicles or a nerve plexus, the vast or schwannomatosis (2714). A mosaic although small numbers of cases with
majority arise in skin or subcutaneous tis- pattern of 8MARCB1 (INl1) expression loss of chromosome 1p, gain of 9q34,
sue of an extremity, the head and neck, is seen in 93% of tumours from patients and gain of 17q have been reported
or the trunk, with deep-seated examples with familial schwannomatosis, 55% of (1471,2699).
also documented {20). These tumours tumours from patients with sporadic
have been described both in childhood schwannomatosis, 83% of NF2-associat- Genetic susceptibility
and at birth {2788). Despite an often rap- ed tumours, and only 5% of solitary, spo- Although most schwannomas are spo-
id growth, hypercellularity, and increased radic schwannomas {1909). radic in occurrence, multiple schwan-
mitotic activity, the behaviour is that of a nomas may occur in the setting of two
benign tumour, prone to local recurrence Genetic profile tumour syndromes. Bilateral vestibular
but with no metastatic potential {2788). Extensive analyses have implicated the schwannomas are pathognomonic of

8chwannoma 217
deposition. Similarly, melanotic schwanno-
mas feature true melanosomes and less.
uniform envelopment of individual cells by
basal lamina on electron microscopy.
The peak age incidence of melanotic
schwannoma is a decade younger than
that of conventional schwannoma. Mel-
anotic schwannomas occur in both non-
psammomatous (720) and psammoma-
tous {368,624) varieties. The vast majority
of non-psammomatous tumours affect
spinal nerves and paraspinal ganglia,

. .,
whereas psammomatous lesions also in-
volve autonomic nerves of viscera, such

-
...,."· as the intestinal tract and heart. Cranial
nerves may also be affected. Distinguish-
Fig. 9.10 Melanotic schwannoma with clusters of plump, spindled, and epithelioid, heavily pigmented tumour cells.
ing between the two varieties of melanot-
ic schwannoma is important, because
NF2, whereas multiple, mostly non-ves- Prognosis and predictive factors about 50% of patients with psammoma-
tibular schwannomas in the absence of Schwannomas are benign, slow-growing tous tumours have Carney complex, an
other NF2 features are characteristic of tumours that infrequently recur and only autosomal dominant disorder {367) char-
schwannomatosis (see Schwannomato- very rarely undergo malignant change acterized by lentiginous facial pigmen-
sis, p. 301). Patients with schwannoma- {2789). Recurrences are more common tation, cardiac myxoma, and endocrine
tosis present with multiple, often painful (occurring in 30-40% of cases) for cel- hyperactivity. Endocrine hyperactivity
schwannomas, which in some cases lular schwannomas of the intracranial, includes Cushing syndrome associated
are segmental in distribution. Germline spinal, and sacral regions {376) and for with adrenal hyperplasia and acromegaly
SMARCB1 mutations at 22q11.23 have plexiform schwannoma {2788). due to pituitary adenoma {368). Slightly
been found in half of all familial and more than 10% of all melanotic schwan-
< 10% of all sporadic schwannomatosis nomas follow a malignant course {367).
cases {1064,2325,2381). Somatic NF2
inactivation has been shown in tumours, Melanotic schwannoma ICD-0 code 9560/1
but germline NF2 mutations are absent
(1115,1557). Germline loss-of-function Definition Genetic susceptibility
mutations in LZTR1 predispose individu- A rare, circumscribed but unencapsu- Allelic loss of the PRKAR1A region on 17q
als to an autosomal dominant inherited lated, grossly pigmented tumour com- has been reported in tumours from pa-
disorder of multiple schwannomas, and posed of cells with the ultrastructure and tients with Carney complex, but has not
are identified in approximately 80% of immunophenotype of Schwann cells but been documented in non-psammoma-
22q-related schwannomatosis cases that contain melanosomes and are reac- tous melanotic schwannomas {2435).
that lack mutations in SMARCB1 (1980). tive for melanocytic markers. Psammomatous melanotic schwannoma
The presence of LZTR1 mutations also In melanotic schwannoma, cytological is a component of Carney complex, in
confers an increased risk of vestibular atypia (including hyperchromasia and which patients have loss-of-function
schwannoma, constituting further over- macronucleoli) is common. Unlike in germline mutations of the PRKAR1A
lap with NF2 {2377). conventional schwannomas, collagen IV gene on chromosome 17q, encoding the
and laminin usually envelop cell nests cAMP-dependent protein kinase type 1-
rather than showing extensive pericellular alpha regulatory subunit {2435)

.'\/�)���:;}-· �
t • !J'.1i�iiffi�t i '·
Fig. 9.11 Melanotic schwannoma. A Psammomatous calcification is a diagnostically useful feature of psammomatous melanotic schwannomas. B This dual stain reveals diffuse
immunoreactivity for melan-A (in red} and a low Ki-67 proliferation index, with positive nuclei (in brown). The low proliferation index helps distinguish this lesion from melanoma.
C Psammomatous melanotic schwannoma. The extensive pericellular collagen-IV expression in this case supports its Schwannian nature, given that melanocytic tumours are not
associated with basement membrane deposition.

218 Tumours of the cranial and paraspinal nerves


Neurofibroma Perry A.
von Deimling A.
Louis D.N.
Hunter S.
Reuss D.E.
Antonescu C.R.

Definition
A benign, well-demarcated, intraneural
or diffusely infiltrative extraneural nerve
sheath tumour consisting of neoplastic,
well-differentiated Schwann cells inter-
mixed with non-neoplastic elements in-
cluding perineurial-like cells, fibroblasts,
mast cells, a variably myxoid to collage-
nous matrix, and residual axons or gan-
glion cells.
Multiple and plexiform neurofibromas are Fig. 9.12 Total spine MRI in a patient with neurofibromatosis type 1 with extensive bilateral paraspinal disease burden.
typically associated with neurofibromato- Neurofibromas involve nearly every nerve root; also note the thoracic spine curvature defect.
sis type 1 (NF1), whereas sporadic neu-
rofibromas are common, mostly cutane-
ous tumours that can affect patients of Clinical features nerve are affected. On cut surface, they
any age and any area of the body. Rarely painful, neurofibroma presents as are firm, glistening, and greyish tan.
a mass. Deeper tumours, including par-
ICD-0 code 9540/0 aspinal forms, present with motor and Microscopy
sensory deficits attributable to the nerve Neurofibromas are composed in large
Grading of origin. The presence of multiple neu- part of neoplastic Schwann cells with thin,
Neurofibroma corresponds histologically rofibromas is the hallmark of NF1, in as- curved to elongated nuclei and scant cy-
to WHO grade I. sociation with many other characteristic toplasm, as well as fibroblasts in a matrix
manifestations (see Neurofibromatosis of collagen fibres and Alcian blue-posi-
Epidemiology type 1, p. 294). tive myxoid material. These cells have
Neurofibromas are common and occur considerably smaller nuclei than those
either as sporadic solitary nodules un- Macroscopy of schwannomas. The cell processes
related to any apparent syndrome or (far Cutaneous neurofibromas are nodular to are thin and often not visible on routine
less frequently) as solitary, multiple, or polypoid and circumscribed, or are dif- light microscopy. Residual axons are
numerous lesions in individuals with NF1. fuse, and involve skin and subcutaneous often present within neurofibromas, and
Patients of any race, age, or sex can be tissue. Neurofibromas confined to nerves can be highlighted with neurofilament
affected. are fusiform and (in all but their proximal immunohistochemistry or Bodian silver
and distal margins) well circumscribed. impregnations. Large diffuse neurofi-
Localization Plexiform neurofibromas consist either of bromas often contain highly character-
Neurofibroma presents most commonly multinodular tangles (resembling a bag istic tactile-like structures (specifically
as a cutaneous nodule (localized cuta- of worms), when involving multiple trunks pseudo-Meissner corpuscles) and may
neous neurofibroma), less often as a cir- of a neural plexus, or of rope-like le- also contain melanotic cells. Stromal col-
cumscribed mass in a peripheral nerve sions, when multiple fascicles of a large, lagen formation varies greatly in abun-
(localized intraneural neurofibroma), or non-branching nerve such as the sciatic dance and sometimes takes the form of
as a plexiform mass within a major nerve dense, retractile bundles with a so-called
trunk or plexus. Least frequent is diffuse shredded-carrot appearance. lntraneural
but localized involvement of skin and neurofibromas often remain confined to
subcutaneous tissue (diffuse cutaneous the nerve, encompassed by its thickened
neurofibroma) or extensive to massive in- epineurium. In contrast, tumours arising
volvement of soft tissue of a body area in small cutaneous nerves commonly
(localized gigantism and elephantiasis spread diffusely into surrounding der-
neuromatosa). Neurofibromas rarely in- mis and soft tissues. Unlike in schwan-
volve spinal roots sporadically, but com- nomas, blood vessels in neurofibromas
monly do so in patients with NF1, in which generally lack hyalinization, and although
multiple bilateral tumours are often asso- neurofibromas sometimes resemble the
ciated with scoliosis and risks of malig- Antoni B regions of a schwannoma, they
nant transformation (1778}; in contrast, Fig. 9.13 Neurofibroma of a spinal root, with a firm generally lack Antoni A-like regions and
they almost never involve cranial nerves. consistency and homogeneous cut surface. Verocay bodies.

Neurofibroma 219

Fig. 9.14 Plexiform neurofibroma. A Multinodular pattern from involvement of multiple fascicles. Note the associated diffuse neurofibroma in the background, with pseudo-Meissner
corpuscles (upper left). B EMA immunostaining highlights the perineurium surrounding multiple involved fascicles, whereas the tumour cells are mostly negative.

Ancient neurofibroma Plexiform neurofibroms positive, but the proportion of reactive


This pattern is defined by degenerative Plexiform neurofibroma is a variant de- cells is smaller than in schwannoma.
nuclear atypia alone (analogous to an- fined by involvement of multiple fascicles, A similar pattern is seen with nuclear
cient schwannoma) and should be dis- which are expanded by tumour cells and SOX10 positivity {1222). Expression of
tinguished from atypical neurofibroma, collagen, but commonly demonstrate re- basement membrane markers is more
given the lack of any associated clinical sidual, bundled nerve fibres at their cen- variable than in schwannoma. Unlike per-
relevance {2146}; this is similar to ancient tres. Rare neurofibromas are thought to ineuriomas, neurofibromas contain only
schwannoma, which has degenerative exhibit limited perineurial differentiation limited numbers of EMA-positive cells,
atypia but lacks any other features of (2845) or form a hybrid neurofibroma/ with reactivity most apparent in residual
malignancy. perineurioma (2146). perineurium. Scattered cells, presumably
the perineurial-like cells seen ultrastruc-
Atypical neurofibroms ICD-0 code 9550/0 turally also show GLUT1 {1012) or claudin
Atypical neurofibroma is a variant de- positivity (2044). Neurofilament staining
fined by worrisome features such as Electron microscopy reveals entrapped axons in intraneural
high cellularity, scattered mitotic figures, Electron microscopy shows a mixture of and plexiform neurofibromas, whereas
monomorphic cytology, and/or fascicular cell types, the two most diagnostically KIT staining highlights recruited mast
growth in addition to cytological atypia, important being the Schwann cell and cells and a subset of stromal cells may
may show premalignant features (see the perineurial-like cell {648). The peri- stain for CD34. Like in other nerve sheath
Genetic profile) and is notoriously difficult neurial-like cell features long, very thin tumours, GFAP expression may be seen.
to distinguish from low-grade malignant cell processes, pinocytotic vesicles, and
peripheral nerve sheath tumour (MPNST) interrupted basement membrane. Fibro- Genetic profile
{178). blasts and mast cells are least frequent. Given their mixed cellular composition, it
has been difficult to determine whether
ICD-0 code 9540/0 lmmunophenotype neurofibromas are monoclonal. Notably,
Staining for 8100 protein is invariably allelic loss of the NF1 gene region of

B In pseudo-Meissner

220 Tumours of the cranial and paraspinal nerves


17q seems to be confined to the S100-
positive Schwann cells in neurofibro-
mas (1947). suggesting that they are the
clonal neoplastic element. This is further
supported by in vitro experiments show-
ing biallelic inactivation of the NF1 gene
in cultured Schwann cells from neurofi-
bromas (1564}, confirming the two-hit hy-
pothesis for the genesis of these lesions,
at least those arising from patients with
NF1. However, some data also suggest
that S1 GO-negative perineurial-like cells
are neoplastic, and that neurofibromas
arise from dedifferentiated myelinating
Schwann cells in a process similar to effects of NF1 loss include activation of Genetic susceptibility
wound healing in peripheral nerve (2112). the RAS/MAPK and AKT/mTOR path- The occurrence of multiple and plexiform
The situation in sporadic tumours has ways. Less is known about cutaneous neurofibromas is a hallmark of NF1 (see
yet to be fully elucidated, but the mor- neurofibromas, although evidence sug- Neurofibromatosis type 1, p. 294). Neu-
phological similarity between sporadic gests a similar mechanism but with a rofibromas are exceptionally uncommon
and inherited neurofibromas, as well as different cell of origin (e.g. dermal skin- in NF2 and schwannomatosis.
the clear involvement of the NF1 gene in derived precursors) and different hotspot
sporadic MPNSTs, suggests that NF1 al- mutations {1442,2545). Prognosis and predictive factors
terations are also involved in the genesis Additional chromosomal losses are not Plexiform neurofibromas and neurofibro-
of sporadic neurofibromas, and this has common in neurofibromas, but have mas of major nerves are considered po-
in fact been documented in rare cases been noted on 19p, 19q, and 22q in tential precursors of M PNST. Malignant
(156,2434). Neurofibromas do not nor- NF1-associated neurofibromas and on transformation occurs in 5-10% of large
mally develop in mouse models with in- 19q and 22q in sporadic neurofibromas plexiform tumours, but is a rare event in
activation of NF1 in Schwann cells, unless (1320). CDKN2A/CDKN2B losses are diffuse cutaneous and massive soft tis-
the mouse itself is also NF1 haploinsuffi- generally considered a sign of malignant sue neurofibromas. A patient with a siza-
cient, analogous to human patients with transformation, and these losses are also ble plexiform neurofibroma is highly likely
NF1; evidence suggests that haploinsuf- commonly found in tumours diagnosed to have NF1 and should be investigated
ficient mast cells within the tumour mi- as atypical neurofibromas in patients with for other evidence of the disorder. Similar
croenvironment stimulate NF1-deficient NF1, suggesting that such tumours may to low-grade MPNST, an atypical neurofi-
Schwann cells to grow and form neurofi- be premalignant lesions (155). broma that extends to a surgical margin
bromas (2869} or that additional signal- has a low risk of subsequent recurrence,
ling pathways of nerve injury are required but essentially no associated mortality
for tumour formation. The downstream (179).

Neurofibroma 221
Perineurioma Antonescu C.R.
Perry A.
Reuss D.E.

Definition ratio of 2:1), and present with non-specif-


A tumour composed entirely of neoplas- ic mass effects. Both the intraneural and
tic perineurial cells. soft tissue variants of perineurioma are
lntraneural perineuriomas are benign rare, accounting for approximately 1% of
and consist of proliferating perineurial nerve sheath and soft tissue neoplasms,
cells within the endoneurium, forming respectively. More than 50 cases of intra-
characteristic pseudo-onion bulbs. Soft neural perineurioma have been reported
tissue perineuriomas are typically not as- to date, including cranial nerve exam-
sociated with nerve, are variably whorled, ples. More than 100 cases of soft tissue
and are usually benign. Malignant soft perineurioma have been described l695,
tissue perineurioma is a rare variant of 827,879,1042,2070}, including an intra-
malignant peripheral nerve sheath tu- osseous example surrounding a cranial
mour displaying perineurial differen- nerve 1134).
tiation. lntraneural perineurioma, long
mistakenly considered a form of hyper- Localization have been reported 1756,1011,1221,2176,
trophic neuropathy, is now recognized as lntraneural perineuriomas primarily affect 2460) and are apparently unassociated
a neoplasm 1637). peripheral nerves of the extremities; cra- with neurofibromatosis type 1 11011)
nial nerve lesions are rare 159,515,1483).
ICD-0 code 9571/0 One example was reportedly associated Macroscopy
with Beckwith-Wiedemann syndrome lntraneural perineurioma produces a
Grading 1424). Soft tissue perineuriomas are lo- segmental, tubular, several-fold enlarge-
lntraneural perineuriomas correspond cated in the deep soft tissue and are ment of the affected nerve. Individual
histologically to WHO grade I. Soft tissue grossly unassociated with nerve. Visceral nerve fascicles appear coarse and pale.
perineuriomas range from benign (corre- involvement is rare 11041,2608). One ex- Most lesions are < 10 cm long, but one
sponding histologically to WHO grade I) ample involving the CNS arose within a 40-cm-long sciatic nerve example has
to variably malignant (corresponding his- lateral ventricle l829} been reported l637) Although multiple
tologically to WHO grades 11-111). fascicles are often involved, a bag-of-
Clinical features worms plexiform growth is not seen. In-
Epidemiology In intraneural perineuriomas, progressive volvement of two neighbouring spinal
lntraneural perineuriomas typically pres- muscle weakness (with or without obvi- nerves has been reported l637}. Soft
ent in adolescence or early adulthood ous atrophy) is more frequent than are tissue perineuriomas are solitary, gener-
and show no sex predilection. Soft tissue sensory disturbances. Malignant exam- ally small (1.5-7 cm), and well circum-
perineuriomas occur in adults, predomi- ples of soft tissue perineuriomas prone scribed but unencapsulated. Rarely, the
nantly females (with a female-to-male to recurrence and occasional metastasis tumours can be multinodular l82}. On cut

222 Tumours of the cranial and paraspinal nerves


surface, they are firm and greyish white
to infrequently focally myxoid. Malignant
soft tissue perineuriomas are usually not
associated with a nerve and may feature
invasive growth and variable necrosis.

Microscopy
lntraneural perineurioma consists of
neoplastic perineurial cells proliferat-
ing throughout the endoneurium. These
cells form concentric layers around ax-
ons, causing enlargement of fascicles
and forming characteristic pseudo-
onion bulbs. This distinctive architectural
feature is best seen on cross-section,
wherein fascicles vary in cellularity. The
proliferation of cytologically normal-look-
ing perineurial cells largely takes place
within endoneurium, but perineurium is
often affected as well. Particularly large
whorls can envelop numerous nerve fi-
bres. Occasionally, perineurial cells en-
closing one or several axons contribute
to an adjacent onion bulb as well. Thus,
pseudo-onion bulbs anastomose, form-
ing a complex endoneurial network. Even
within a single fascicle, cell density and
the complexity of the lesion can vary. Mi-
totic activity is rare. In early lesions, ax-
onal density and myelination may be al-
most normal, whereas in fully developed Necrosis is typically lacking. The sclerot- thin cytoplasmic processes bearing nu-
lesions, when most fibres are surrounded ic variant, characterized by an abundant merous pinocytotic vesicles and are lined
by perineurial cells and therefore widely collagenous stroma, has been described by patchy surface basement membrane.
separated, myelin is often scant or absent occurring mainly in the fingers of young Stromal collagen may be abundant.
on Luxol fast blue staining. At late stages, male patients {695). This variant features Soft tissue perineuriomas typically con-
only Schwann cells without accompany- only crude whorl formation, occasionally sist of spindle-shaped cells with long,
ing axons may remain at the centre of the centred on a minute nerve. A reticular exceedingly thin cytoplasmic processes
perineurial whorls. Hyalinization may be variant that occurs at a variety of anatom- embedded in an abundant collagenous
prominent. ical sites and primarily affects adults fea- stroma. Cytoplasm is scant and contains
Soft tissue perineuriomas are composed tures a lace-like or reticular growth pat- sparse profiles of rough endoplasmic
of spindled, wavy cells with remarkably tern composed of anastomosing cords of reticulum, occasional mitochondria, and
thin cytoplasmic processes arranged in fusiform cells {879). Malignant soft tissue a few randomly distributed intermediate
lamellae and embedded in collagen fi- perineuriomas (i.e. perineurial malignant filaments. The processes exhibit numer-
bres. Crude whorls or storiform arrange- peripheral nerve sheath tumours) are ous pinocytotic vesicles and a patchy
ments are commonly seen. Aggregates uncommon and characterized by hyper- lining of basement membrane. lntercellu-
of collagen fibres are often encircled by cellularity, hyperchromasia, and often lar tight junctions are relatively frequent.
long, remarkably narrow tumour cell pro- brisk (but variable) mitotic activity (WHO One example featuring ribosome-lamella
cesses. Nuclei are elongate with tapered grade II), necrosis usually being a feature complexes has been reported {580). Ma-
ends and are often curved or wrinkled. of WHO grade Ill tumours. Progressive lignant soft tissue perineuriomas show
Nucleoli are inconspicuous. Granu- malignant change of WHO grade II to similar ultrastructural features {1011,
lar cells are a very uncommon feature grade Ill lesions may be seen, but trans- 2176,2460}, only a subset being poorly
of perineurioma {585). Mitoses vary in formation of benign soft tissue perineu- differentiated {1011).
number. In the largest published series riomas to malignant examples has not
{1042}, the mitotic count ranged from Oto been documented. lmmunophenotype
13 (mean: 1) mitoses per 30 high-power Like normal perineurial cells, all intraneu-
fields, with 65% of the tumours showing Electron microscopy ral perineuriomas are immunoreactive for
none. Degenerative atypia (i.e. nuclear lntraneural perineuriomas feature myeli- vimentin. The pattern of EMA staining is
pleomorphism, hyperchromasia, and cy- nated nerve fibres circumferentially sur- membranous, as are those of collagen IV
toplasmic-nuclear inclusions) is seen pri- rounded by ultrastructurally normal-look- and laminin. Axons at the centre of pseu-
marily in long-standing tumours {1042) ing perineurial cells. The cells have long, do-onion bulbs and residual Schwann

Perineurioma 223
cells stain for NFP and 8100 protein, re- which often have a low mitotic index, studies of malignant soft tissue perineu-
spectively. Staining for p53 protein has malignant soft tissue perineuriomas are riomas have been reported.
also been reported (637). Soft tissue more proliferative, with mitotic counts of
perineurioma features the same basic 1-85 mitoses (median: 16) per 10 high- Prognosis and predictive factors
immunophenotype. Claudin-1 (719,2070) power fields in the largest reported series lntraneural perineuriomas are benign.
and GLUT1 (1012} are also diagnostically {1011 }. Long-term follow-up indicates that they
useful markers. Unlike various other soft do not have a tendency to recur or me-
tissue tumours, perineuriomas generally Genetic profile tastasize. Biopsy alone is sufficient for
lack reactivity for CD34, MUC4, and in Both intraneural and soft tissue perineu- diagnosis. Conventional soft tissue peri.
particular, 8100 protein. Malignant soft riomas feature the same cytogenetic ab- neuriomas are usually amenable to gross
tissue perineuriomas usually show at normality: monosomy of chromosome 22 total removal. Recurrences are very in-
least some EMA staining and lack 8100 (637,827). Loss of chromosome 13, an frequent, even in cases with histological
protein reactivity. abnormality found in several soft tissue atypia, and none have been reported
tumours, has also been described in soft to metastasize. Neither sclerotic nor re-
Proltferation tissue perineurioma {1717). Loss of chro- ticular tumours are prone to recurrence
lntraneural perineuriomas, despite a pau- mosome 10 and a small chromosome {695,879). Malignant perineuriomas are
city of mitoses, may show a Ki-67 prolif- 22q deletion involving NF2 have also far less prone to metastasize {756,1011,
eration index of 5-15% (637) In contrast been reported {281,2311} No genetic 1221) than are conventional malignant
to benign soft tissue perineuriomas. peripheral nerve sheath tumours (1011].

Hybrid nerve sheath tumours Antonescu C.R.


Stemmer-Rachamirnov A.O.
Perry A.

Definition 1040). They are rarely associated with eosinophilic cytoplasm with indistinct
Benign peripheral nerve sheath tumours cranial or spinal nerves. Most tumours cell borders, arranged in a storiforrn,
(PNSTs) with combined features of more showing biphasic schwannoma and re- whorled, and/or lamellar architecture
than one conventional type (i.e. neurofi- ticular perineurioma have been reported (1040). The tumours may exhibit myx-
broma, schwannoma, and perineurioma). on the digits {1661 }. oid stromal changes (seen in half of all
Two of the more common types of hybrid cases) and often display degenerative
nerve sheath tumours are schwannoma/ Clinical features cytological atypia similar to the ancient
perineurioma, which typically occurs The clinical features of peripheral nerve changes seen in schwannoma.
sporadically, and neurofibroma/schwan- sheath hybrid tumours are similar to or Hybrid neurofibromas/schwannomas are
noma, which is typically associated with indistinguishable from those of other be- tumours in which two distinct compo-
schwannomatosis, neurofibromatosis nign PNSTs and largely depend on the nents are recognized: a schwannoma-
type 1 (NF1) or neurofibromatosis type 2 site of origin. Hybrid nerve sheath tu- like component with nodular Schwann
(NF2). Rare cases of neurofibroma/peri- mours (including spinal examples) may cell proliferation (which may contain
neurioma have also been described, cause neurological deficit or pain when Verocay bodies) and a neurofibroma-like
usually associated with NF1. they involve a large peripheral nerve. component with a mixed cellular popula-
tion, myxoid change, and collagen {681,
Etiology Macroscopy 945). Plexiform architecture is common.
Hybrid schwannoma/perineurioma oc- The gross appearance and radiological The schwannoma-like component is
curs sporadically {1040}, whereas neu- findings of hybrid tumours are indistin- mainly composed of cellular Antoni A
rofibroma/schwannoma can occur in guishable from those of other PNSTs (i e areas, often containing Verocay bodies
the setting of either schwannomatosis or schwannomas and neurofibromas). with Schwann cells demonstrating nu-
neurofibromatosis {945). Similarly, hybrid clear palisading. In contrast, the neurofi-
neurofibroma/perineurioma tumours are Microscopy broma-like component may demonstrate
more commonly reported in association Hybrid schwannoma/perineurioma tu- abundant fibroblasts, collagen, and
with NF1 (1091,1199). mours show predominantly Schwannian myxoid changes, with Schwannian cells
cytomorphology but have a perineurio- having a distinctive elongated and wavy
Localization ma-like architecture. These tumours are appearance.
Hybrid nerve sheath tumours show a usually well circumscribed but unencap- The few examples of hybrid neurofibroma/
wide anatomical distribution and often sulated, and composed of spindle cells perineurioma that have been described
involve the dermis and subcutis (681, with plump, tapering nuclei and palely consisted of plexiform neurofibromas

224 Tumours of the cranial and paraspinal nerves


with considerable areas of perineuri- The most helpful stains for the work-up of lesions within the spectrum of PNSTs in
omatous differentiation, in patients with a neurofibroma/schwannoma are those NF1 l17).
NF1 11199) In these lesions, biphasic that highlight the presence of a mono-
(Schwannian and perineuriomatous) dif- morphic Schwann cell population in the Genetic susceptibility
ferentiation was apparent mainly on im- schwannoma component (i.e. S100 and More than half of all patients with hybrid
munohistochemistry, with rare cases in SOX10) or a polymorphic cell population PNSTs have multiple PNSTs, suggesting
which the neurofibromatous and perineu- in the neurofibroma component, includ- a tumour syndrome. Hybrid neurofibro-
riomatous areas were recognizable on ing Schwann cells (S100 and SOX10), ma/schwannoma is a common tumour
routine H&E stains 11199). perineurial cells (EMA and GLUT1), and type in schwannomatosis, occurring in
fibroblasts. Entrapped axons may be 71% of patients. There is also a striking
lmmunophenotype seen in neurofibromatosis-associated association with neurofibromatosis l945)
Schwannoma/perineurioma hybrid tu- schwannomas, but the presence of en- where this hybrid lesion is more common
mours show dual differentiation by im- trapped large bundles of axons is more in NF2 (occurring in 26% of cases) than
munohistochemistry, with the Schwann- common in neurofibromas l681). in NF1 (occurring in 9% of cases). Within
ian cells (plump-spindled) being positive In neurofibroma/perineurioma, the bipha- patients with schwannomatosis, 61% of
for S100 protein whereas the perineurial sic Schwannian and perineuriomatous the developed tumours had the appear-
cells (slender-spindled) show variable differentiation is apparent by immunohis- ance of schwannoma-like nodules within
immunoreactivity for EMA, claudin-1, and tochemistry, with the perineuriomatous a neurofibroma-like tumour, correspond-
GLUT1 12816). Using double staining for areas staining positively for EMA, GLUT1, ing to hybrid neurofibroma/schwannoma
EMA and S100 protein, parallel layers of and claudin-1 and negatively for S100 1945). The presence of hybrid morphol-
alternating S100-positive and EMA-posi- protein 11199). In fact, intraneural perineu- ogy and/or mosaic SMARCB1 (INl1)
tive cells can be seen, with no coexpres- rial proliferations have been documented expression on immunohistochemistry
sion of antigens by the same cells 11040). by screening neurofibromatous lesions suggests that a schwannoma may be
On the basis of these dual-labelling re- and normal nerves in patients with NF1 associated with a form of neurofibroma-
sults, most tumours were found to be using a battery of perineurial markers, tosis, in particular NF2 and schwanno-
composed of about two thirds Schwann supporting the existence of both pure matosis 11909,1990) Similarly, hybrid
cells and one third perineurial cells. and hybrid perineuriomatous neurofibroma/perineurioma occurs most-
ly in association with NF1 11199)

Hybrid nerve sheath tumours 225


.,

Malignant peripheral nerve sheath Reuss D.E.


Louis D.N.
Perry A.
Hirose T.
tumour Hunter S. Antonescu C.R.

Definition dilection. Childhood and adolescent cas-


A malignant tumour with evidence of es account for 10-20% of MPNSTs {66). 80-89+

Schwann cell or perineurial cell differen- 70-79

tiation, commonly arising in a peripheral Etiology 60-69

nerve or in extraneural soft tissue. About 50% of all MPNSTs are associated 50-59

- ::�: !!!!!!!
Malignant peripheral nerve sheath tu- with NF1, in which setting they typically
mours (MPNSTs) primarily affect young arise from deep-seated plexiform neu-
to middle-aged adults, but also affect rofibromas or large intraneural neurofi- - 20-29-
10-19
adolescents; about 50% of MPNSTs are bromas. About 40% of MPNSTs arise in 0-9
associated with neurofibromatosis type patients without known predisposition 150 100 50 50 100 150

1 (NF1), in which they often arise from a {606,1055). and 10% of MPNSTs are Number of cases Number of cases

pre-existing plexiform or intraneural neu- associated with previous radiation ther- Fig. 9.22 Age and sex distribution of MPNST, based
rofibroma and affect younger patients. apy {1416). Only rare examples develop on 1711 histologically confirmed cases. Data from the
In contrast, most sporadic cases arise from conventional schwannoma {1628). Surveillance, Epidemiology, and End Results Program
(SEER), National Cancer Institute, Washington DC.
from large peripheral nerves without an ganglioneuroblastoma/ganglioneuroma
associated benign precursor. Most MP- {2113). or phaeochromocytoma {2223).
NSTs show combined inactivation of NF1, Imaging
CDKN2A, and PRC2 component genes. Localization Imaging findings correspond to those
Large and medium-sized nerves are dis- of soft tissue sarcoma. Inhomogeneous
ICD-0 code 9540/3 tinctly more prone to involvement than contrast enhancement and irregularity of
are small nerves. The most commonly contour (a reflection of invasion) are com-
Grading involved sites include the buttock and monly seen. FOG-PET is a sensitive tool
Clinically validated and reproducible thigh, brachia! plexus and upper arm, for the detection of MPNSTs in patients
grading systems for MPNST are gener- and paraspinal region. The sciatic nerve with NF1 {441,574).
ally lacking. One approach to MPNST is most frequently affected. Cranial nerve
grading is to divide the tumours into low- MPNSTs are rare, and more commonly Spread
grade (-15% of cases) and high-grade arise from schwannomas than do MP- MPNSTs often infiltrate adjacent soft tis-
(-85% of cases) {2146). albeit without NSTs located elsewhere {2260). Primary sues and may spread along intraneu-
robust and validated criteria. Low-grade intraparenchymal MPNST is rare {129). ral and haematogenous routes. About
MPNSTs are well-differentiated tumours 20-25% of patients develop metastases,
most often arising in transition from Clinical features most commonly to the lungs {2874).
neurofibroma. An increased mitotic rate The most common presentation in the
is often seen but is not required for the extremities is a progressively enlarging Macroscopy
diagnosis {1166). Conventional mono- mass, with or without neurological symp- The gross appearance of MPNST varies
morphous spindle cell MPNSTs, highly toms. Spinal tumours often present with greatly. Because a significant proportion
pleomorphic MPNSTs, and MPNSTs with radicular pain {2439). of these tumours arise in neurofibroma,
divergent differentiation (e.g. malignant
triton tumour; glandular MPNST; and os-
teosarcomatous, chondrosarcomatous,
and angiosarcomatous differentiation)
are all considered high-grade.

Epidemiology
MPNSTs are uncommon, accounting for
� 5% of all malignant soft tissue tumours
{1479). MPNSTs primarily affect adults
in the third to the sixth decades of life.
The mean age of patients with MPNSTs
associated with NF1 is approximately Fig. 9.23 Malignant peripheral nerve sheath tumour (MPNST). A Resected MPNST with its parent nerve on the left.
a decade younger (28-36 years) than Note the skeletal muscle that forms part of the surgical margin, indicating invasion into the surrounding soft tissues.
that of patients with sporadic cases (40- B On cut surface, this tumour shows the classic variegated appearance, with fleshy soft cellular regions alternating with
44 years) {606,1055).There is no sex pre- yellow foci of necrosis.

226 Tumours of the cranial and paraspinal nerves


some as focal transformations, the pro- tightly packed spindle cells with variable Malignant peripheral nerve sheath
cess may be minimally apparent on gross quantities of eosinophilic cytoplasm. Nu- tumour with divergent differentiation
examination. In contrast, larger, typically clei are typically elongated and wavy
high-grade tumours originating in or un- and (unlike those of smooth muscle) have Synonyms
associated with a nerve produce either tapered ends. The tumours show either Malignant triton tumour; glandular malig-
fusiform, expansile masses or globular, alternating loose and densely cellular ar- nant peripheral nerve sheath tumour
entirely unencapsulated soft tissue tu- eas or a diffuse. growth pattern. Perivas-
mours. Both types infiltrate surrounding cular hypercellularity and tumour aggre- A variety of mesenchymal tissues such
structures. The vast majority of tumours gates appearing to herniate into vascular as cartilage, bone, skeletal muscle,
are > 5 cm, and examples > 10 cm are lumina are also common {1924). Unusual smooth muscle, and angiosarcoma-like
common. Their consistency ranges from growth patterns may be seen, includ- areas can be present in MPNSTs. MP-
soft to hard, and the cut surface is typi- ing haemangiopericytoma-like areas or NSTs showing rhabdomyosarcomatous
cally cream-coloured or grey. Foci of rarely, nuclear palisading. MPNSTs grow differentiation are called malignant triton
necrosis and haemorrhage are common within nerve fascicles but commonly in- tumours. Nearly 60% of patients with ma-
and may be extensive. vade adjacent soft tissues. A pseudo- lignant triton tumour have NF1. Glandular
capsule of variable thickness is often MPNST is a variant containing glandular
Microscopy present. Three quarters of these tumours epithelium that resembles that of intes-
MPNSTs vary greatly in appearance. have geographical necrosis and mi- tine. Neuroendocrine differentiation is fre-
Many exhibit a herringbone (fibrosarco- totic activity, often showing > 4 mitoses quently seen, whereas squamous epithe-
ma-like) or interwoven fasciculated pat- per high-power field (high-grade). lium is far less often encountered. Three
tern of cell growth. Both patterns feature quarters of the patients have NF1 {2785)

Malignant peripheral nerve sheath tumour 227


The common association with NF1 and a MPNST with perineurial differentiation conventional MPN8Ts, but unlike MP-
spindle-cell background indistinguisha- (malignant perineurioma) N8Ts, they carry an SS18-SSX2 or SS18-
ble from that of ordinary MPN8T suggest SSX1 fusion gene {2259).
a close relationship between high-grade ICD-0 code 9540/3
MPN8T with divergent differentiation and /mmunophenotype
conventional high-grade MPN8T Rare MPN8Ts show histological and Only 50-70% of MPN8Ts exhibit 8100
ultrastructural features of perineurial dif- protein staining. Reactivity is grade-re-
The following two variants are likely to ferentiation. Like benign perineuriomas, lated. In high-grade tumours reactivity is
be different not only in their histological these tumours are EMA-positive and either patchy or found only in individual
characteristics but also in their genetics, 8100-negative, but show hypercellular- cells, whereas in low-grade examples
lack of syndromic association, and/or bi- ity, nuclear atypia, and increased mitotic it may be extensive (2724). However, in
ological behaviour, and should therefore activity. Perineurial MPN8Ts have the po- epithelioid MPN8T, diffuse 8100 protein
be strictly distinguished from convention- tential to metastasize, but appear to be expression is common and 8MARCB1
al MPN8T: epithelioid MPN8T and peri- less aggressive than conventional MP- (INl1) is lost in 50% of cases (718). Lack
neurial MPN8T (malignant perineurioma). N8Ts (1011,2176). of immunostaining for HMB45 and mel-
an-A, taken together with origin from
Differential diagnosis of MPNSTs a peripheral nerve or a benign nerve
Epithelioid malignant peripheral The distinction of MPN8Ts from other sheath tumour may help distinguish
nerve sheath tumour high-grade. sarcomas relies mainly on epithelioid MPN8T from malignant mela-
demonstration of tumour origin from ei- noma. lmmunostaining for p53 protein is
ICD-0 code 9540/3 ther a peripheral nerve or a benign pre- positive in 75% of all MPN8Ts, in contrast
cursor, or on immunohistochemical or to the infrequent staining in neurofibro-
Less than 5% of MPN8Ts are either genetic features. Until proven otherwise, mas {931), although cellular schwanno-
partially or purely epithelioid (718,1162, malignant spindle cell tumours in patients mas can also be positive (1924). Most
1435). This variant shows no association with NF1 should be considered to be MP- MPN8Ts are also negative for CDKN2A
with NF1 and can arise from malignant N8Ts. One particular entity to consider (p16), in contrast to the consistent but of-
transformation of a schwannoma (1628, in the differential diagnosis of MPN8T ten patchy expression in cellular schwan-
2789}. Both superficial (above the fascia) is synovial sarcoma of nerve. 8ynovial nomas {1785,1924). However, some atyp-
and deep-seated examples have been sarcomas are common soft tissue sar- ical neurofibromas also show CDKN2A
reported. The risks of recurrence, metas- comas but can also occur as distinct (p16) loss (2862). EGFR is expressed in
tasis, and disease-related death seem to rare primary tumours of nerve. These tu- about one third of MPN8Ts but is absent
be lower than those associated with con- mours show considerable morphological in cellular schwannomas. Diffuse loss of
ventional MPN8T (1162) and immunohistochemical overlap with 80X10 occurs in 75% of MPN8Ts, but

228 Tumours of the cranial and paraspinal nerves


sox10 is retained in cellular schwan-
nomas 11924). Full-length neurofibromin
is absent in about 90% of MPNSTs as-
sociated with NF1 and 43% of sporadic
MPNSTs. Diffuse loss of neurofibromin is
not observed in synovial sarcoma, soli-
tary fibrous tumour, dedifferentiated lipo-
sarcoma, myxoid liposarcoma, cellular
schwannoma, or low-grade fibromyxoid
sarcoma, but may occur in myxofibro-
sarcoma, pleomorphic liposarcoma, and
undifferentiated pleomorphic sarcoma
11924,2102). MPNSTs with divergent dif-
ferentiation show expression of related
differentiation markers (e.g. desmin in
malignant triton tumour or keratin, car-
cinoembryonic antigen, and neuroen-
docrine markers in glandular MPNST).
In most MPNSTs, the Ki-67 proliferation
index is> 20% 11924).
Loss of H3K27me3 expression was re- �
cently shown to be a highly specific Fig. 9.28 Perineurial malignant peripheral nerve sheath tumour. A Storiform pattern and focal necrosis (upper right).
marker for MPNST, although only mod- B A whorling pattern. C Patchy immunoreactivity for EMA and (D) the presence of long thin processes, pinocytotic
estly more sensitive than S100 protein vesicles, and patchy basement membrane support perineurial differentiation.
and SOX10, being documented in >90%
of radiation-associated MPNST and in pathogenesis of most MPNSTs, a hypoth- tumours. Although only a small number
>60% of NF1-related MPNST l2038A, esis that is also supported by functional of cases have been analysed to date, ep-
2254A). The H3K27me3 loss of expres- data. NF1 encodes for the important ithelioid MPNSTs do not have the same
sion is more variable within the sporadic RasGAP neurofibromin, and its inactiva- genetic profile as conventional MPNSTs,
MPNST, ranging from 49-95% among the tion increases the levels of active RAS. suggesting that they may constitute a
2 recent studies, most likely related to the Loss of function of PRC2 through dele- distinct entity 11460). Molecular data on
different criteria applied in diagnosis. tion/mutation of SUZ12 or EEO leads to perineurial MPNST are lacking.
decreased levels of H3K27me3 and in-
Cell of origin creased levels of H3K27ac, thereby am- Genetic susceptibility
Mouse models of peripheral nerve sheath plifying the transcription of RAS target Approximately half of all MPNSTs mani-
tumours associated with NF1 suggest genes {548). COKN2A encodes for the fest in patients with NF1 (see Neurofibro-
that MPNSTs do not directly derive from important cell cycle regulators p16 and matosis type 1, p. 294). This association
neural crest stem cells, but instead from p14ARF, and deletion of the COKN2A is particularly strong for malignant triton
more differentiated Schwann cells 11182, locus enables evasion from hyperac- tumour and glandular MPNST. Patients
2793,2859) Embryonic Schwann cell tive RAS-induced senescence, promot- with NF1 and plexiform neurofibromas
precursors have been identified (using ing sustained proliferation {2324,2336). have the highest risk of developing
cell-lineage tracing) as cells of origin in a In addition, genetic alterations in TP53 MPNST 12588}.
mouse model of plexiform neurofibroma are found in about 42% of all MPNSTs,
1429). further supporting tumour progression Prognosis and predictive factors
11460). MPNSTs typically have complex MPNSTs (except those with perineurial
Genetic profile numerical and structural karyotypic ab- differentiation) are highly aggressive tu-
MPNST associated with NF1, sporadic normalities. Common abnormalities in- mours with a poor prognosis. In a large
MPNST, and radiation-induced MPNST clude gains of chromosomes 2, 7p, Sq, retrospective study, truncal location, tu-
share highly recurrent genetic inactiva- 14, and 17q and losses of chromosomes mour size � 5 cm, local recurrence, and
tion in NF1, COKN2A, and the PRC2 com- 9p, 11q, 13q, 17p, and 1811362). Gains at high-grade designation had adverse
ponents SUZ12 and EEO 11460,2857). 16p or losses from 10q or Xq have been impacts on disease-specific survival
Biallelic inactivation of NF1 is present in reported as negative prognostic factors 12439} The study also reported a trend
benign neurofibromas, but mutations in 1274). Gene amplifications that occur towards decreased survival in patients
additional genes are rarely found. How- in a subset of tumours include ITGB4, with cases associated with NF1 com-
ever, atypical neurofibromas, which are POGFRA, BIRC5, CCNE2, EGFR, HGF, pared with sporadic cases. Gains at 16p,
presumed to be MPNST precursors, of- MET, TERT, and COK4, with COK4 am- losses from 10q or Xq, and COK4 ampli-
ten show additional deletions in COKN2A plifications being an independent pre- fications have been reported as negative
1155). These genetic data suggest that the dictor of poor survival {1579,2834). No prognostic factors {274,2834).
combined inactivation of NF1, COKN2A, cytogenetic differences have been noted
and PRC2 components is critical for the between sporadic and NF1-associated

Malignant peripheral nerve sheath tumour 229


CHAPTER10
Meningiomas
Meningioma
Meningothelial meningioma
Fibrous meningioma
Transitional meningioma
Psammomatous meningioma
Angiomatous meningioma
Microcystic meningioma
Secretory meningioma
Lymphoplasmacyte-rich meningioma
Metaplastic meningioma
Chordoid meningioma
Clear cell meningioma
Atypical meningioma
Papillary meningioma
Rhabdoid meningioma
Anaplastic (malignant) meningioma
Meningioma Perry A.
Louis ON
Rushing E.J.
Mawrin C.
Budka H. Claus E.B.
von Deimling A. Loeffler J.
Sahm F. Sadetzki S.

Definition tumours overall, although tumours of the Etiology


A group of mostly benign, slow-growing meninges account for just 2.8% of all Ionizing radiation is the only established
neoplasms that most likely derive from paediatric primary brain tumours {596) environmental risk factor for meningioma,
the meningothelial cells of the arachnoid More than 90% of all meningiomas are with higher risk among people who were
layer. solitary. About 20-25% and 1-6% of exposed in childhood than as adults. At
There are three major groups of meningi- meningiomas are WHO grades II and 111, high dose levels, data exist for patients'
omas, which differ in grade and biologi- respectively { 1837, 1951, 2448]. treated with therapeutic radiation to the
cal behaviour (see Table 10.01) head {189,1001,2213]. Evidence also ex-
Age and sex distnbution ists for lower dose levels {470,1607,1920}. ·
ICD-0 code 9530/0 The median age of patients with meningi- Two studies of imaging technologies (e.g.
oma is 65 years, with risk increasing with CT) that use diagnostic levels of radiation.
Grading age {596]. Age-adjusted incidence rates higher than those used for dental or plain·
Most meningiomas correspond histologi- vary significantly by sex. Females are at X-rays reported links with subsequent
cally to WHO grade I (benign). Certain greater risk than males, with annual inci- brain tumours (glioma and meningioma)
histological subtypes or meningiomas, dence rates of 10. 5 cases per 100 000 fe- {1607,1920). Researchers from the Tinea ·
with specific combinations of morpholog- males and 4.8 cases per 100 000 males Capitis Cohort Study have studied genet.::·
ical parameters, are associated with less {596]. This difference is greatest prior ic predisposition for radiation-assoclaterf
favourable clinical outcomes and cor- to menopause, with the highest female- meningioma and have found strong sup- •
respond histologically to WHO grades 11 to-male ratio (3 15:1) in the 35-44 years port for genetic susceptibility to the de- .
and Ill (Table 10.01) age group {2746). Grade II and Ill le- velopment of meningioma after exposure
sions occur at higher rates in males. In- to ionizing radiation {717).
Epidemiology cidence also varies significantly by race, An association between hormones and·
with reported annual incidence rates per meningioma risk is suggested by sev-s
Incidence 100 000 population of 9.1, 7.4, and 4.8 eral findings, including the increased·
The lifetime risk of developing meningi- in Blacks, Whites, and Asians I Pacific incidence of the disease in women ver-·
oma is approximately 1%. It is the most Islanders, respectively, in the USA {596). sus men; the presence of progesterone •
frequently reported brain tumour in the receptors in most meningiomas; and re-
USA. accounting for 36% of all brain ports of modestly increased risk assocl-c
ated with the use of endogenous/exog-
enous hormones, body mass index, and
current smoking, as well as decreased
risk associated with breastfeeding for
2". 6 months {471]. A recent large case-��
control study found that among female
subjects, members of the case group:
were more likely than members of the ·•
control group to report hormonally relat- ·
ed conditions, including uterine fibroids
(OR 1.2, 95% Cl: 1.0-1.5), endometrio- � -
sis (OR 1.5, 95% Cl: 1.5-2.1), and breast
cancer (OR: 14, 95% Cl 0.8-2.3) {469}. ,,
Evidence of a possible interaction be- ;
tween smoking and sex was also report-.·
ed {716}. Attempts to link specific chemi-
cals, dietary factors, and occupations, as '
well as head trauma and mobile phone
use, with meningiomas have provided
inconclusive findings. However, allergic
0-19 20-34 35-44 45.54 55-64 65-74 75-84 85+ conditions (e.g. asthma and eczema)
Fig. 10.01 Age- and sex-specific incidence rates (per 100 000 population) of meningioma in the USA (2002-2006). have been associated with reduced risk
The left scale refers to the bar graph, the right scale to the female-to-male incidence ratio, which is indicated for each of meningioma {2747}.
age group by a diamond {27 46).

232 Meningiomas
Table 10.01 Meningioma variants grouped by WHO grade and biological behaviour
Meningiomas with low risk of recurrence and aggressive behaviour:
ICD-Ocode
Meningothelial meningioma WHO grade I 9531/0
fibrous (fibroblastic) meningioma WHO grade I 9532/0
Transitional (mixed) meningioma WHO grade I 9537/0
Psammomatous meningioma WHO grade I 9533/0
Angiomatous meningioma WHO grade I 9534/0
Microcystic meningioma WHO grade I 9530/0
Secretory meningioma WHO grade I 9530/0
Lymphoplasmacyte-rich meningioma WHO grade I 9530/0
Metaplastic meningioma WHO grade I 9530/0
Meningiomas with greater likelihood of recurrence and aggressive behaviour:
Chordoid meningioma WHO grade II 9538/1
Clear cell meningioma WHO grade II 9538/1
Atypical meningioma WHO grade II 9539/1
Papillary meningioma WHO grade Ill 9538/3
Rhabdoid meningioma WHO grade Ill 9538/3
Anaplastic meningioma WHO grade Ill 9530/3
Meningiomas of any subtype with high proliferation index

Localization dural masses. Calcification is common,


The vast majority of meningiomas arise and is best seen on CT. A characteris- Fig. 10.02 Meningioma. A T1-weighted, post-constrast
in intracranial, intraspinal, or orbital lo- tic feature is the so-called dural tail sur- coronal MRI showing a meningioma of the cerebral
cations. lntraventricular and epidural ex- rounding the dural perimeter of the mass. convexity with a prominent dural 'tail'. B Postcontrast
amples are uncommon. Rare examples This familiar imaging sign corresponds T1-weighted MRI of a meningioma with homogeneous
have been reported outside the neural to reactive fibrovascular tissue and does contrast enhancement. The adjacent cortex appears
to mould around the tumour. The trailing contrast into
axis (e.g. in the lung). Within the cranial not necessarily predict foci of dural in-
adjacent dura is referred to as the dural tail sign and
cavity, common sites include the cere- volvement by tumour. Peritumoural cere- corroborates the impression of an extra-axial mass.
bral convexities (with tumours often locat- bral oedema is occasionally prominent,
ed parasagittally, in association with the in particular with certain histological vari-
falx and venous sinus), olfactory grooves, ants and high-grade examples (1856).
sphenoid ridges, para-/suprasellar re- Cyst formation may occur within or at
gions, optic nerve sheath, petrous ridg- the periphery of a meningioma. Neuro-
es, tentorium, and posterior fossa. Most imaging features are not entirely specific
spinal meningiomas occur in the thoracic for identifying meningiomas, predicting
region. Atypical and anaplastic menin- tumour behaviour, or excluding other
giomas most commonly affect the con- diagnoses.
vexities and other non-skull base sites
(1214). Metastases of malignant menin- Spread
giomas most often involve lung, pleura, Even benign meningiomas commonly
bone, or liver. invade adjacent anatomical structures
(especially dura), although the rate and
Clinical features extent of local spread are often greater
Meningiomas are generally slow-grow- in the more aggressive subtypes. Thus,
ing and produce neurological signs and depending on location and grade, some
symptoms due to compression of adja- meningiomas produce considerable pa-
cent structures; the specific deficits de- tient morbidity and mortality. Extracra-
pend on tumour location. Headache and nial metastases are extremely rare, oc-
seizures are common (but non-specific) curring in about 1 in 1000 meningiomas
presentations. and most often in association with WHO
grade 111 tumours. The rare metastases of Fig. 10.03 Large meningioma originating from the
Imaging histologically benign meningiomas typi- olfactory groove. Note the smooth, slightly lobulated
On MR I, meningiomas typically present as cally occur after surgery, but can arise surface. MCA, middle cerebral artery; ON, optic nerve;
isodense, uniformly contrast-enhancing de novo as well. PG, pituitary gland.

Meningioma 233
Fig. 10.04 Macroscopy of meningioma. A Meningioma of the left parasagittal region of the parietal lobe. The tumour compresses the cerebral cortex, but does not infiltrate.
B Large lateral meningioma compressing the left frontal cortex. Note the attachment to the dura mater and the sharp delineation from brain structures. In this location, meningiomas
are often fiat rather than round. C Meningioma of the medial sphenoid wing encasing the carotid artery. D Large meningioma of the lateral ventricles and the third ventricle causing
a hydrocephalus. E Large meningioma originating from the clivus. Note the compression of the brain stem with residual haemorrhage. The basilar artery (arrowhead) is entrapped
by the tumour but not occluded. F Spinal meningioma compressing the spinal cord.

Macroscopy attachment. Invasion of dura or dural si- sections and Table 10.01). The criteria
Most meningiomas are rubbery or firm, nuses is fairly common. Occasional men- used to diagnose atypical and anaplastic
well-demarcated, sometimes tabulated, ingiomas invade into the adjacent skull, meningiomas are applied independent of
rounded masses that feature broad dural where they may induce characteristic specific meningioma subtype.
hyperostosis, which is highly indicative of
bone invasion. Meningiomas may attach /mmunophenotype
to or encase cerebral arteries, but only The vast majority of meningiomas stain
rarely infiltrate arterial walls. They may for EMA, although this immunoreactivity
also infiltrate the skin and extracranial is less consistent in atypical and malig-
compartments, such as the orbit. Adja- nant lesions. Vimentin positivity is found
cent brain is often compressed but rarely in all meningiomas, but is relatively non-
frankly invaded. In certain sites, particu- specific. Somatostatin receptor 2A is ex-
larly along the sphenoid wing, menin- pressed strongly and diffusely in almost
giomas may grow as a flat, carpet-like all cases (including anaplastic menin-
mass, a pattern called en plaque menin- giomas), but can also be encountered
gioma. Some meningiomas appear gritty in neuroendocrine neoplasms (1641).
on gross inspection, implying the pres- S100 protein positivity is most common
ence of numerous psammoma bodies. in fibrous meningiomas, but is not usually
Bone formation is far less common. Atyp- diffuse, as it is in schwannomas. Other
ical and anaplastic meningiomas tend to potentially useful immunohistochemical
be larger and often feature necrosis. markers in selected cases include Ki-67
and progesterone receptor (see Progno-
Microscopy sis and predictive factors).
Meningiomas exhibit a wide range of his- Diagnostic ultrastructural features of
tological appearances. Of the subtypes meningiomas include abundance of
in the WHO classification, the most com- intermediate filaments (vimentin), com-
mon are meningothelial, fibrous, and plex interdigitating cellular processes
transitional meningiomas. Most of the (particularly in meningothelial variants),
Fig. 10.05 Dura mater showing multiple meningiomas
subtypes behave in a benign fashion, but and desmosomal intercellular junctions.
that differ greatly in size. They are located unilaterally,
without extending beyond the falx cerebri (FC) to the four distinct variants, which are catego- These cell surface specializations, as
contralateral side. Multiple dural meningiomas have been rized as WHO grade II and Ill, are more well as intermediate filaments, are few
shown to be of clonal origin and probably result from likely to recur and to follow a more ag- in fibrous meningiomas, the cells be-
spread through the dura (2413). gressive clinical course (see the following ing separated by collagen. Secretory

234 Meningiomas
rneningiomas feature single or multiple sites, create stop codons, or result in rare alterations {1615,2291 ). Additional
epithelial-like lumina within single cells. frameshifts occurring mainly in the 5'- altered genes have been identified us-
These cell surfaces show short apical most two thirds of the gene (1536) The ing genomic and targeted sequencing
rnicrovilli and surround electron-dense common, predictable effect of these mu- strategies in a subset of NF2-wildtype
secretions (1393). Microcystic meningi- tations is a truncated and presumably meningiomas (262,467,2217) These
omas feature long cytoplasmic process- non-functional merlin protein (also called alterations preferentially affect WHO
es enclosing intercellular electron-lucent schwannomin). The frequency of NF2 grade I meningiomas at the skull base. A
matrix, with cells joined by desmosomes. mutations varies among meningioma hotspot mutation in the AKT1 gene (AKT1
variants. Fibroblastic and transitional E17K) is found in about 13% of meningi-
Proliferation meningiomas, which are preferentially omas, mostly of meningothelial or transi-
In general, cellular proliferation increases located at the convexity, often carry NF2 tional subtype. Another mutation affects
in proportion to grade. The mitotic index mutations {954,1367,2727). In contrast, the TRAF7 gene (mutated in 8-24% of
and Ki-67 proliferation index correlate meningothelial, secretory, and microcyst- cases), almost always occurring in com-
approximately with volume growth rate. ic meningiomas located at the skull base bination with KLF4 mutations, which are
Studies have suggested that meningi- only rarely harbour NF2 mutations, but also found in 93-100% of secretory men-
omas with an index of > 4% have an in- are driven by other genetic alterations ingiomas {2104). Mutations in the SMO
creased risk of recurrence similar to that (see Other genes). In line with this, most gene are found in 4-5% of WHO grade I
of atypical meningioma, whereas those non-NF2 meningioma families develop meningiomas, mostly those located in
with an index of > 20% are associated meningothelial tumours (1536]. Further- the medial anterior skull base. Addition-
with death rates analogous to those as- more, reduced expression of merlin has ally, SMARCE1 mutations have recently
sociated with anaplastic meningioma been observed in various histopathologi- been identified in clear cell meningiomas
(1953). However, significant differences cal variants of meningiomas, but seems to (2375).
in technique and interpretation make be rare in meningothelial tumours (1455). Deletions of the 9p21 region, including
it difficult to establish definitive cut-off In atypical and anaplastic meningiomas, the CDKN2A gene, are particularly com-
points that would translate accurately NF2 mutations occur in approximately mon in anaplastic meningiomas and are
from one laboratory to another. 70% of cases, matching the frequency associated with shortened survival (246,
of NF2 mutations in benign fibroblastic 1938). Mouse models have also shown
Cell of origin and transitional meningiomas. This in- that adding CDKN2A/B loss to NF2 loss
Meningiomas are thought to be derived dicates that NF2 inactivation is an early enhances the rate of formation of men-
from meningothelial (arachnoid) cells. tumorigenic event, a theory supported by ingiomas, including high-grade forms
findings in mouse models (1204). Mouse (1960} Additionally, higher-grade men-
Genetic profile models have also indicated an origin ingiomas escape senescence due to
The current model of meningioma genet- from prostaglandin-0 synthase-positive overexpression of telomerase, which in
ics in WHO grades 1-111 is summarized in meningeal precursor cells with inactivat- a subset of cases occurs due to TERT
Fig. 10.06. ed NF2 (1205). NF2-driven meningiomas promoter mutations {876). The poten-
Meningiomas were among the first solid with malignant progression show more tial prognostic value of this marker was
tumours recognized as having cytoge- genetic instability than do NF2-intact shown in a study of 252 meningiomas re-
netic alterations, the most consistent meningiomas {877). In radiation-induced vealing hotspot C228T and C250T muta-
change being monosomy 22 {2846} In meningiomas, the frequencies of NF2 tions in 1.7%, 5.7%, and 20.0% of WHO
general, karyotypic abnormalities are mutations and loss of chromosome 22 grade I. 11, and Ill meningiomas respec-
more extensive in atypical and anaplastic are lower, whereas structural abnormali- tively, with median progression free sur-
meningiomas (16151. Other cytogenetic ties of chromosome 1 p are more com- vival times of 10.1 versus 179 months in
changes associated with meningioma in- mon, suggesting a different molecular mutant versus wildtype tumours {2220A].
clude deletion of chromosome 1p (which pathogenesis (1164,1501). Other molecular alterations associated
is associated with poor outcome) {1132) with high tumour grade and aggressive
and losses of chromosomes 6q, 9p, 10, Other genes clinical behaviour include losses of the
14q, and 18q (which occur in higher- The close association of NF2 mutations NDRG2 gene on 14q11.2 and the MEG3
grade tumours) (2846). Chromosomal in meningiomas with allelic loss on chro- gene on 14q32, gains of the RPS6KB1
gains reported in higher-grade meningi- mosome 22 and mouse modelling of gene and other loci on the 17q23 am-
omas include gains of chromosomes 1q, meningioma development clearly sug- plicon, loss of various NF2 homologues
9q, 12q, 15q, 17q, and 20q. gest that NF2 is the major meningioma within the erythrocyte membrane protein
tumour suppressor gene on that chromo- band 4.1 family (e.g. the EPB41L3 gene
TheNF2gene some (2727). However, deletion studies on chromosome 18p11.3), and loss of
Mutations in the NF2 gene are detected of chromosome 22 have also detected the protein 4.18 binding partner, CADM1
in most meningiomas associated with losses and translocations of genetic ma- (116,333,343,1548, 16151.
neurofibromatosis type 2 (NF2) and as terial outside the NF2 region, raising the
many as 60% of sporadic meningiomas possibility that other tumour-associated Clonality of solttary, recurrent, and
[1466,2727). In most cases, such muta- genes are located there. Candidate multiple meningiomas
tions are small insertions or deletions or genes include LARGE, MN1, AP1B1, and Studies of X chromosome inactivation us-
are nonsense mutations that affect splice SMARCB1, although these show only ing Southern blot analysis indicate that

Meningioma 235
controls to report a first-degree family
Arachnoidal cap cell (PGDS•)
history of meningioma (469). In the Inter-
t phone Study, the largest study of genetic
polymorphisms and meningioma risk,
Gradel - Grade II investigators found a significant associa-
tion with meningioma for 12 SNPs drawn
NF2 from DNA repair genes (181). The group
SMARCB1 SMARCE1") reported a novel and biologically intrigu-
I!!0 f
� KLF4 ing association between meningioma risk
1il c:0 TRAF7 and three variants in BRIP1 (17q22), the
:;
:E .s SMO h TERT promoter gene encoding the FACJ protein, which
interacts with BRCA 1. A genome-wide
AKT1
association study identified a single sus-
ceptibility locus at 10p12.31 (MLLT10)
Loss:22q
{593). MLLT10 is implicated in various
Loss: 1p / 6q / 10 / 14q / 18q leukaemias and activates the WNT path-
Gain: 1q / 9q / 12q / 15q / 17q / 20q way. A rare association with naevoid ba-
sal cell carcinoma syndrome {also called
Loss: 9p
Gorlin syndrome) has also been suggest-
TSLC1 t PDGFR t EGFR t ed, based on germline SUFU or PTCH1
mutations (1265,2202). The relationship
PRt 11MP3 t NDRG2 t MEG3 t between meningioma and other inher-
ited tumour syndromes, such as Werner
VEGF t IGF t WNT t ---
syndrome and Cowden syndrome, is less
CDKN2A/ 28 t defined.
hTERT t ====
Prognosis and predictive factors
Fig. 10.06 Genetic model of meningioma tumorigenesis and progression; figure adapted from Karajannis MA and
Zagzag D (eds): Molecular Pathology of Nervous System Tumors. Chapter 17. Springer 2015. Mutations are labelled The major prognostic questions regard-
in grey, with light grey indicating mutations occurring in meningiomas without NF2 alterations. Cytogenetic aberrations ing WHO grade II and Ill meningiomas in-
are labelled in blue, and gene expression changes are labelled in green. Bar length indicates the relative frequency volve estimates of recurrence and overall
of an alteration within the given tumour grade. *SMARCE1 mutations have been found nearly exclusively in clear survival, respectively.
cell meningiomas. EGFR, epidermal growth factor receptor; IGF, insulin-like growth factor; PDGFR, platelet-derived
growth factor receptor; PGDS', prostaglandin 02 synthase-positive precursor cells in murine meningioma models; PR, Clinical factors
progesterone receptor.
In most cases, meningiomas can be re-
moved entirely, as assessed by operative
meningiomas are monoclonal tumours through dural spread, a hypothesis also or neuroradiological criteria. In one se-
{1118). although PCR-based assays have supported by the common findings of ries, 20% of gross totally resected benign
suggested that a small proportion could peritumoural implants in 10% of menin- meningiomas recurred within 20 years
be polyclonal {2867). Nevertheless, both giomas {244) and of small meningothe- {1197). The major clinical factor in recur-
the Southern blot data (1118} and the lial nests in grossly unremarkable du- rence is extent of resection, which is influ-
finding that the overwhelming majority ral strips from the convexities of patients enced by tumour site, extent of invasion,
of meningiomas with NF2 mutations only with meningiomas {244). Nevertheless, it attachment to vital intracranial structures,
have a single mutation {2727) indicate is possible that some cases with multiple and the skill of the surgeon. Other clinical
that meningiomas are clonal. Similarly, all meningiomas constitute genetic mosa- factors, such as young patient age and
recurrent meningiomas have been found ics, with segmental, dural constitutional male sex are less powerful predictors of
to be clonal with respect to the primary NF2 mutations. Germline mutations in recurrence; both are partially explained
tumour {2663). The clonality of multiple the SMARCB1 gene can also give rise by the increased frequency of high-grade
meningiomas has also been analysed to multiple schwannomas and meningi- meningiomas in such patients.
using studies of X chromosome inac- omas {2622).
tivation and by mutation analysis of the HhwpaMohgyandgradmg
NF2 gene in multiple tumours from the Genetic susceptibility Some histological variants of meningi-
same patient {1433,2413} In these stud- Although the vast majority of meningi- oma are more likely to recur. However,
ies, the majority of lesions from patients omas are sporadic, rare examples oc- overall, WHO grade is the most useful
with ;:: 3 meningiomas were shown either cur as part of tumour predisposition morphological predictor of recurrence
to have the same copy of the X chromo- syndromes, with NF2 being by far the {Table 10.01). Benign meningiomas
some inactivated or to carry the same most common. However, family history have recurrence rates of about 7-25%,
NF2 mutation. These data provide strong studies have also suggested a role for whereas atypical meninqiornas recur in
evidence for a clonal origin of multiple inherited susceptibility beyond NF2, in- 29-52% of cases, and anaplastic men-
meningiomas in most patients. They cluding one study in which patients with ingiomas at rates of 50-94%. Even within
also suggest that multiple lesions arise meningioma were 4.4 times as likely as the benign meningiomas however, the

236 Meningiomas
presence of some atypical features (but Progesterone receptor status because a significant subset of histologi-
not enough for WHO grade II designa- Progesterone receptor expression is cally and clinically benign meningiomas
tion) increases the risk of subsequent inversely associated with meningioma also lack the receptor, the significance of
progression/recurrence over those with grade. Its absence negatively impacts this finding in the absence of other prog-
no atypical features at all l1581A) Malig- disease-free intervals in some series, nostic features should not be overstated.
nant histological features are associated but in most, it is not independent of other
with shorter survival times: 2-5 years de- known prognostic factors, such as grade
pending largely on the extent of resection !1940,21811. Almost all WHO grade Ill
pOA,483A,1951). meningiomas are receptor negative, but

Meningioma variants Perry A. Sahm F. Grading


Louis D.N Mawrin C. Meningothelial meningioma corresponds
Budka H. Rushing E.J. histologically to WHO grade I.
van Deimling A.

Fibrous meningioma
Meningothelial meningioma lobules should not be confused with the
sheeting or loss of architectural pattern Definition
Definition seen in atypical meningioma. This sub- A variant of meningioma that consists of
A classic and common variant of me- type is encountered most often in the an- spindled cells forming parallel, storiform,
ningioma, with medium-sized epithelioid terior skull base and is less likely to be and interlacing bundles in a collagen-rich
tumour cells forming lobules, some of driven by NF2 mutations !942,1367). matrix.
which are partly demarcated by thin col- Because the tumour cells so closely re- In fibrous meningioma, whorl formation
lagenous septa. semble those of the normal arachnoid and psammoma bodies are infrequent.
Like normal arachnoid cap cells, the tu- cap, reactive meningothelial hyperplasia Nuclear features characteristic of menin-
mour cells of meningothelial meningioma occasionally resembles this meningi- gothelial meningioma are often found fo-
are largely uniform, with oval nuclei with oma variant. The most florid examples of cally. The tumour cells form fascicles with
delicate chromatin and variable nuclear meningothelial hyperplasia are typically various amounts of intercellular collagen,
holes (i.e empty-looking clear spaces) found adjacent to optic nerve gliomas, which are striking in some cases. These
and nuclear pseudoinclusions (i.e. cyto- other tumour types, or haemorrhage; fascicles may raise the differential diag-
plasmic invaginations). Eosinophilic cyto- meningothelial hyperplasia is also com- nosis of solitary fibrous tumour I haeman-
plasm is abundant, and the delicate, intri- monly found in the setting of chronic giopericytoma (SFT/HPC), but only SFT/
cately interwoven tumour cell processes renal disease, advanced patient age, HPC expresses nuclear STAT6 !2308).
seen ultrastructurally cannot be dis- arachnoiditis ossificans, spontaneous Rare fibrous meningiomas also include
cerned on light microscopy, explaining intracranial haemorrhage, and occasion- nuclear palisades resembling Verocay
the outdated synonym "syncytial menin- ally in patients with diffuse dural thicken- bodies, a diagnostic pitfall exacerbated
gioma". Whorls and psammoma bodies ing and contrast enhancement on neuro- by frequent S100 expression in this sub-
are infrequent in meningothelial men- imaging !1945). type, albeit not as diffusely staining as
ingioma; when present, they tend to be most schwannomas. Like other subtypes,
less formed than in transitional, fibrous, ICD-0 code 9531/0 most fibrous meningiomas express EMA.
or psammomatous subtypes. Larger

I I
Fig. 10.07 Meningothe/ial meningioma with lobular Fig. 10.08 Fibrous meningioma. A A distinctive feature of this variant is the development of abundant reticulin and
growth pattern, syncytium-like appearance due to poorly collagen fibres between the individual cells. B Most fibrous meningiomas express EMA.
defined cell borders, scattered clear nuclear holes, and
occasional intranuclear pseudoinclusions (arrows).

Meningioma variants 237


NF2 mutations and convexity locations (usually of the transitional type) contain-
are common. ing a predominance of psammoma bod-
ies over tumour cells.
ICD-0 code 9532/0 In psammomatous meningioma, the
psammoma bodies often become con-
Grading fluent, forming irregular calcified masses
Fibrous meningioma corresponds histo- and occasionally bone. In some tumours,
logically to WHO grade I. the tumour cells are almost completely
replaced by psammoma bodies, and in-
tervening meningothelial cells are hard to
Transitional meningioma find. Psammomatous meningiomas char-
acteristically occur in the thoracic spinal
Definition region of middle-aged to elderly women,
A common variant of meningioma that and the majority of these tumours behave
contains meningothelial and fibrous pat- in an indolent fashion.
terns as well as transitional features. Fig. 10.11 Angiomatous meningioma on FLAIR MRI,
In transitional meningioma, lobular and ICD-0 code 9533/0 showing marked peritumoural brain oedema.
fascicular foci appear side by side with
conspicuous tight whorls and psammoma Grading inhibin alpha and brachyury expression
bodies. NF2 mutations and origin from the Psammomatous meningioma corre- {133} rather than meningothelial markers
convexity are common {942,1367}. sponds histologically to WHO grade I. such as EMA and somatostatin recep-
tor 2A. The designation "angiomatous"
ICD-0 code 9537/0 should not be equated with the obsolete
Angiomatous meningioma term "angioblastic meningioma" (see
Grading Solitary fibrous tumour I haemangioperi-
Transitional meningioma corresponds Definition cytoma, p. 249). Angiomatous meningi-
histologically to WHO grade I. A variant of meningioma that features nu- omas do not exhibit aggressive behav-
merous blood vessels, which often consti- iour, although adjacent cerebral oedema
tute a greater proportion of the tumour mass may be out of proportion to tumour size
Psammomatous meningioma than do the intermixed meningioma cells. {1856}. Unlike most meningiomas, which
Angiomatous meningioma is also known have a normal diploid or monosomy 22
Definition as vascular meningioma. The tumour karyotype, angiomatous meningiomas
A designation applied to meningiomas cells may be hard to recognize as menin- are often aneuploid and commonly have
gothelial, with cytological features often polysomies, particularly for chromo-
overlapping with those of the microcystic somes 5, 13, and 20 {2}.
meningioma. The vascular channels are
small to medium-sized, thin- or thick- ICD-0 code 9534/0
walled, and variably hyalinized. Moderate
to marked degenerative nuclear atypia is Grading
common, but the vast majority of these Angiomatous meningioma corresponds
tumours are histologically and clinically histologically to WHO grade I.
benign {963} The differential diagnosis
includes vascular malformations and
haemangioblastoma, although the stro-
Fig. 10.09 Transitional meningioma with prominent whorl mal cells of haemangioblastoma feature
formation.

-- ::.ti
Fig.10.10 Psammomatous meningioma. A CT showing bone-like density of a psammomatous meningioma (arrowhead) involving the cervicomedullary junction. B Almost complete
replacement of the meningioma by psammomatous calcifications (postdecalcification specimen). C EMA immunostaining reveals meningioma cells between psammoma bodies.

238 Meningiomas
B Tumour cells showing

Fig.10.13 Microcystic meningioma on T2-weighted MRI, Fig. 10.14 Microcystic meningioma. A Cobweb-like background with numerous delicate processes. B Thin processes
with macrocysts and adjacent brain oedema. are often evident on EMA immunostaining.

Microcystic meningioma ICD-0 code 9530/0 These pseudopsammoma bodies show


immunoreactivity for carcinoembryonic
Definition Grading antigen and a variety of other epithe-
A variant of meningioma characterized Microcystic meningioma corresponds lial and secretory markers, and the sur-
by cells with thin, elongated processes histologically to WHO grade I. rounding tumour cells are positive for
encompassing microcysts and creating a both carcinoembryonic antigen and cy-
cobweb-like background. tokeratin. Secretory meningiomas may
Occasionally, grossly or radiologically Secretory meningioma be associated with elevated blood levels
discernible macrocysts are also evident of carcinoembryonic antigen that drop
in microcystic meningioma. Degenera- Definition with resection and rise with recurrence
tive nuclear atypia is common, but mi- A variant of meningioma characterized by (1531}. Mast cells may be numerous
crocystic meningiomas are typically the presence of focal epithelial differen- and there is often peritumoural oedema
benign. Like in the angiomatous variant tiation in the form of intracellular lumina (2560}. Genetically, secretory meningi-
with which microcystic meningioma is of- containing periodic acid-Schiff-positive omas are characterized by the combina-
ten intermixed, accompanying cerebral eosinophilic secretions called pseu- tion of KLF4 K4090 and TRAF7 muta-
oedema is common (1875}. dopsammoma bodies. tions (2104}.

Fig. 10.15 Small secretory meningioma (T) on T2- B Evidence of


weighted MRI, showing extensive peritumoural brain
oedema.

Meningioma variants 239


Fig. 10.17 Lymphoplasmacyte-rich meningioma. Signal
heterogeneity on FLAIR MRI likely corresponds to
pockets of inflammation.

ICD-0 code 9530/0

Grading
Secretory meningioma corresponds his-
tologically to WHO grade I.

Lymphoplasmacyte-rich
meningioma
Definition
A rare variant of meningioma that features
extensive chronic inflammatory infiltrates,
often overshadowing the inconspicuous
meningothelial component.
Some previously reported cases of lym-
phoplasmacyte-rich meningioma likely
constitute inflammatory processes with
associated meningothelial hyperpla-
sia, although cases behaving similarly
to conventional meningioma have also Fig. 10.20 Chordoid meningioma. A On cut surface, the mucoid matrix is grossly evident. B Eosinophilic tumour
cells in a mucous-rich matrix. C Trabeculae of eosinophilic and vacuolated epithelioid cells associated with a basophilic
been described {1421} Neuroimaging mucin-rich stroma. D Vimentin immunohistochemistry highlights the ribbon-like architecture.
features vary widely, with frequent peri-
tumoural oedema and occasional multi-
focality or diffuse carpet-like meningeal often predominate and plasma cells are These alterations have no known clinical
involvement resembling pachymeningi- not always conspicuous, the alternative significance, and many probably do not
tis. Systemic haematological abnormali- term "inflammation-rich meningioma" has constitute true metaplasia (e.g. lipid ac-
ties, including hyperglobulinaemia and also been proposed (1421l. cumulation rather than true lipomatous
iron-refractory anaemia have also been metaplasia {483}). Clinical correlation is
reported {2866l. Because macrophages ICD-0 code 9530/0 occasionally needed to distinguish os-
sified meningiomas from meningiomas
. .. Grading exhibiting bone invasion.
Lymphoplasmacyte-rich meningioma
corresponds histologically to WHO ICD-0 code 9530/0
grade I.
Grading
Metaplastic meningioma corresponds
Metaplastic meningioma histologically to WHO grade I.

Definition
�;������ t
A variant of meningioma with striking fo- Chordoid meningioma
Fig. 10.19 Lipoma-like metaplastic meningioma.
cal or widespread mesenchymal compo-
Positivity for EMA in lipoma-like cells suggests fat nents including osseous, cartilaginous, Definition
accumulation in meningioma cells rather than true lipomatous, myxoid, and xanthomatous A rare variant of meningioma that histo·
adipocyte metaplasia. tissue, either singly or in combinations. logically resembles chordoma, featuring

240 Meningiomas
pattemles:bm�e�
chitecture and round to polygonal cells
with clear, glycogen-rich cytoplasm and
prominent blocky perivascular and inter-
stitial collagen.
The perivascular and interstitial collagen
occasionally coalesces into large acel-
lular zones of collagen or forms brightly
eosinophilic amianthoid-like collagen. It
shows prominent periodic acid-Schiff-
positive and diastase-sensitive cytoplas-
mic glycogen. Whorl formation is vague
at most and psammoma bodies are in-
conspicuous. Clear cell meningioma has
a proclivity for the cerebellopontine angle
and spine, especially the cauda equina
region. It also tends to affect younger
patients, including children and young
adults. Clear cell meningiomas are asso-
ciated with more aggressive behaviour,
including frequent recurrence and occa-
sional cerebrospinal fluid seeding (2873).
Familial examples have been reported in
association with SMARCE1 mutations
(2379). The more aggressive behaviour
' 1_ -- • • �...:..•
of these tumours, corresponding to WHO
Fig. 10.21 Clear cell meningioma. A Sheets of rounded clear cells and perivascular interstitial collagenization. grade 11, has been most clearly demon-
B Abundant periodic acid-Schiff-positive intracytoplasmic glycogen. C lmmunoreactivity for EMA. strated when the clear-cell pattern is pre-
dominant and well developed.
cords or trabeculae of eosinophilic, often of these tumours, corresponding to WHO
vacuolated cells set in an abundant mu- grade II, has been most clearly demon- ICD-0 code 9538/1
coid matrix. strated when the chordoid pattern is pre-
In chordoid meningioma, chordoid areas dominant and well developed. Grading
are often interspersed with more typical Clear cell meningioma corresponds his-
meningioma tissue; pure examples are ICD-0 code 9538/1 tologically to WHO grade II.
uncommon. Chronic inflammatory infil-
trates are often patchy when present, Grading
but may be prominent. Chordoid men- Chordoid meningioma corresponds his- Atypical meningioma
ingiomas are typically large, supraten- tologically to WHO grade II.
torial tumours. They have a very high Definition
rate of recurrence after subtotal resec- A meningioma of intermediate grade be-
tion (501 }. Infrequently, patients have Clear cell meningioma tween benign and malignant forms, with
associated haematological conditions, increased mitotic activity, brain invasion
such as anaemia or Castleman disease Definition on histology, or at least three of the follow-
{1249). The more aggressive behaviour A rare variant of meningioma with a ing features: increased cellularity, small

Fig. 10.22 Brain-invasive meningioma. A Tongue-like protrusions into adjacent brain parenchyma. B Leptomeningeal meningioma in a child, with extensive perivascular spread
along Virchow-Robin spaces and hyalinization mimicking meningioangiomatosis. This unusual pattern of spread should not be misinterpreted as true brain invasion. C Entrapped
GFAP-positive islands of gliotic brain parenchyma at periphery of the tumour.

Meningioma variants 241


underlying parenchyma. without an inter-
vening layer of leptomeninges. This caus-
es reactive astrocytosis. with entrapped
islands of GFAP-positive parenchyrna
at the periphery of the tumour. Exten-
sion along perivascular Virchow-Robin
spaces does not constitute brain invasion
because the pia is not breached in this
form of spread; perivascular spread and
hyalinization can mimic meningioangio-
Fig. 10.23 Atypical meningioma. A Atypical meningioma is most reliably identified by increased mitotic activity
matosis. but does not constitute true brain
(arrows). Note the absence of nuclear atypia in this example. B The micronecrosis seen in this image is considered invasion. Such examples are most com-
spontaneous in that it was not iatrogenically induced (e.g. by embolization). monly encountered in children {819,19441
Brain invasion can occur in meningiomas
that otherwise appear benign. atypical. or
anaplastic (malignant). The presence of
brain invasion is associated with a higher
likelihood of recurrence. Because histo-
logically benign and histologically atypi-
cal brain-invasive meningiomas both have
recurrence and mortality rates similar to
those of atypical meningiomas as defined
using other criteria {19521, brain invasion
is a criterion for atypical meningioma.
Fig. 10.24 Papillary meningioma. A Postcontrast T1-weighted MRI. Occasional papillary meningiomas feature
a caulifiower-like imaging appearance. B Nucleus-free perivascular zone resembling the pseudorosette of an ICD-0 code 9539/1
ependymoma; the additional presence of mitotic figures is evident on the right.
Grading
cells with a high nuclear-to-cytoplasmic because it results more commonly from Atypical meningioma corresponds histo-
ratio, prominent nucleoli, sheeting (i.e. degenerative changes. Clinical risk fac- logically to WHO grade II.
uninterrupted patternless or sheet-like tors for atypical meningioma include
growth), and foci of spontaneous (i.e. not male sex. non-skull base location, and
iatrogenica!ly induced) necrosis. prior surgery {1214f. Atypical meningi- Papillary meningioma
In one series. increased mitotic activity omas have been associated with high re-
was defined as 2'. 4 mitoses per 10 high- currence rates, even after gross total re- Definition
power (40x magnification. 0.16 mm2) section {23l Bone involvement has been A rare variant of meningioma defined by the
fields {1952f. An alternative grading ap- associated with increased recurrence presence of a perivascu/ar pseudopapillary
proach simply combines hypercellularity rates in the setting of atypical meningi- pattern constituting most of the tumour.
with a mitotic count of 2'. 5 mitoses per oma {771f. This pseudopapillary architecture is
10 high-power fields {1569}. Despite the Invasion of the brain by meningioma is characterized by loss of cohesion. with
tumour's name. nuclear atypia itself is not characterized by irregular, tongue-like clinging of tumour cells to blood ves-
useful in grading atypical meningioma. protrusions of tumour cells infiltrating sels and a perivascular nucleus-free

�·· i
Fig. 10.25 Papillary meningioma. A At low magnification, the pseudopapillary pattern is evident, with loss of cellular cohesion away from central vascular cores. B This meningioma
combines a papillary growth pattern with rhabdoid cytology, including globular paranuclear inclusions (inset).

242 Meningiomas
Fig. 10.26 Rhabdoid meningioma. A Eccentrically placed vesicular nuclei, prominent nucleoli, and eosinophilic globular/fibrillar paranuclear inclusions. B Vimentin-positive
paranuclear inclusions. C Rhabdoid meningioma cell showing a paranuclear whorled bundle of intermediate filaments with entrapped organelles.

zone resembling the pseudorosettes of demonstrated when the papillary pattern prominent eosinophilic paranuclear in-
ependymoma. This feature frequently in- is predominant and well developed. clusions, appearing either as discernible
creases in extent with recurrences, and whorled fibrils or compact and waxy.
other high-grade histological features are ICD-0 code 9538/3 These rhabdoid cells resemble those de-
almost always found. Papillary meningi- scribed in other tumours, including atypi-
omas tend to occur in young patients, in- Grading cal teratoid/rhabdoid tumour of the brain;
cluding children {1545). An invasive ten- Papillary meningioma corresponds histo- however, unlike in atypical teratoid/rhab-
dency, including brain invasion, has been logically to WHO grade Ill {1545). doid tumour, SMARCB1 expression is
noted in 75% of cases, recurrence in retained in rhabdoid meningioma {1941).
55%, metastasis (mostly to lung) in 20%, Rhabdoid cells may become increas-
and death of disease in about half {1368, Rhabdoid meningioma ingly evident with tumour recurrences.
1901). Some meningiomas combine a Most rhabdoid meningiomas have a high
papillary architecture with rhabdoid cy- Definition proliferation index and other histological
tology {2796). The more aggressive be- An uncommon variant of meningioma features of malignancy. Some combine
haviour of these tumours, corresponding that consists primarily of rhabdoid cells: a papillary architecture with rhabdoid
to WHO grade 111, has been most clearly plump cells with eccentric nuclei, open cytology (see Papillary meningioma,
chromatin, a prominent nucleolus, and p. 242). When the rhabdoid features are

Fig. 10.27 Anaplastic (malignant) meningioma (common, albeit non-specific macroscopic features). A Note the soft, gelatinous consistency on cut surface, as well as the large yellow
zone of necrosis on the right. B The superior sagittal sinus is occluded by tumour. C The inner surface of the skull is moth-eaten due to extensive bone invasion by an adjacent
meningioma.

Fig. 10.28 Bilateral parasagittal anaplastic (malignant) Fig. 10.29


meningioma. Irregular borders and highly invasive like anaplastic (malignant) meningioma.
growth pattern on postcontrast T1-weighted MRI. cytology.

Meningioma variants 243


, :�-,.
Fig. 10.30 Anaplastic (malignant) meningioma. A Carcinoma-like anaplastic meningioma. Sheet-like growth with large epithelioid cells, abundant cytoplasm, and prominent nucleoli.
B More than 20 mitoses per 10 high-power fields. Eight mitotic figures can be seen in this single high-power field. C Chondrosarcoma-like focus.

fully developed and combined with other ICD-0 code 9538/3 account for 1-3% of meningiomas over-
malignant features, these meningiomas all. In addition to high mitotic counts,
often have an aggressive clinical course Grading most also show extensive necrosis and
consistent with WHO grade Ill (1252, Rhabdoid meningioma corresponds his- a Ki-67 proliferation index > 20%. Rare
1950). However, some meningiomas tologically to WHO grade Ill. cases show true epithelial or mesenchy-
show rhabdoid features only focally and/ mal metaplasia {1910). Confirmation of
or lack other histological features of ma- the meningothelial origin of cases with
lignancy; such cases are less aggres- Anap/astic (malignant) diffuse anaplasia often requires either a
sive as a group (2639A). Therefore, it is meningioma history of meningioma at the same site
suggested that they be graded as nor- or immunohistochemical, ultrastructural,
mally, but with the added descriptor of Definition and/or genetic support.
"with rhabdoid features" and a comment A meningioma that exhibits overtly malig- Anaplastic meningiomas are often fa-
that closer follow-up may be warranted nant cytology (resembling that of carcino- tal, with average survival times ranging
(2639A). Rare forms appear rhabdoid on ma, melanoma, or high-grade sarcoma) from < 2 years to > 5 years, depending
histology, but ultrastructurally show inter- and/or markedly elevated mitotic activity. greatly on the extent of resection (1951,
digitating processes rather than the typi- In one study, markedly elevated mitotic 2448). Clinical risk factors for anaplastic
cal paranuclear aggregates of intermedi- activity was defined as � 20 mitoses per meningioma include a non-skull base or-
ate filaments (823). 10 high-power (0.16 mm2) fields (1951). igin, male sex, and prior surgery (1214).
Anaplastic (malignant) meningiomas Because malignant progression in

244 Meningiomas
rneningiomas, as in gliomas, is a contin- Other morphological variants encountered secondarily in a variety
uum of increasing anaplasia, determining of meningiomas is that of meningothe-
the cut-off point between atypical and Due to the wide morphological spec- lial rosettes, which are composed mostly
anaplastic meningioma is sometimes trum that can be encountered in men- of cell processes and collagen, with or
challenging. ingiomas, rare examples are difficult without a central gland-like lumen (1520).
to classify as any of the well-accepted Most tumours once called pigmented
ICD-0 code 9530/3 variants. These include meningiomas meningiomas are now known to be mel-
with oncocytic, mucinous, sclerosing, anocytomas. However, the recruitment of
Grading whorling-sclerosing. G FAP-expressing, melanocytes from the adjacent meninges
Anaplastic (malignant) meningioma corre- and granulofilamentous inclusion-bear- into the substance of a true meningioma
sponds histologically to WHO grade Ill. ing features {46,173,781,917,1039,2168). accounts for dark pigmentation in rare
However, there is currently insufficient cases (1768).
evidence that these tumours constitute
distinct variants. Another rare pattern

meningothelial rosettes.

..._ .. � .:
Fig. 10.33 Meningioma with oncocytic features. A Marked degenerative nuclear atypia. B EMA expression. C lmmunostain for antimitochondrial antigen confirms the oncocytic
changes.

Meningioma variants 245


v'

·i
CHAPTER 11

Mesenchymal, non-meningothelial tumours


Solitary fibrous tumour I haemangiopericytoma
Haemangioblastoma
Haemangioma
Epithelioid haemangioendothelioma
Angiosarcoma
Kaposi sarcoma
Ewing sarcoma I peripheral PNET
Lipoma
Angiolipoma
Hibernoma
Li posarcoma
Desmoid-type fibromatosis
Myofibroblastoma
Inflammatory myofibroblastic tumour
6 Benign fibrous histiocytoma
Fibrosarcoma
Undifferentiated pleomorphic sarcoma I
malignant fibrous histiocytoma
Leiomyoma
Leiomyosarcoma
Rhabdomyoma
Hhabdornyosarcorna
Chondroma
Chondrosarcoma
Osteoma
Osteochondroma
Osteosarcoma
Mesenchymal, non-meningothelial Antonescu C.R.
Paulus W.
Bouvier C.
Figarella-Branger D.
tumours Perry A.
Rushing E.J.
von Deimling A.
Wesseling P.
Hainfellner J.A.

Definition and the other benign mesenchymal tu- terminale and also occur at the thoracic
Benign and malignant mesenchymal tu- mours are even rarer. In two series, of level. lntraventricular and tuber cinereurn
mours originating in the CNS, with ter- 19 and 17 cases, sarcomas accounted lipomas are rare (233). Occasional CNS
minology and histological features cor- tor « 0.1-0.2% of the intracranial tumours lipomas have a fibrous connection with
responding to their soft tissue and bone (1838,1919}. The higher values that have surrounding soft or subcutaneous tis-
counterparts. been reported in the past are a reflection sue. Osteolipomas have predilection for
Mesenchymal tumours arise more com- of overdiagnosis related to historical clas- the suprasellar/interpeduncular regions
monly in the meninges than in the CNS sification schemes. The most common {233,2367} Most spinal lipomas (in par-
parenchyma or choroid plexus. In prin- tumour types include fibrosarcoma and ticular angiolipomas) arise in the epidural
ciple, any mesenchymal tumour may undifferentiated pleomorphic sarcoma I space.
arise within or secondarily impact on the malignant fibrous histiocytoma (MFH)
nervous system, but the primary mesen- (1838,1919}. Clinical features
chymal CNS tumours are very rare. They Mesenchymal tumours can occur in pa- The clinical symptoms and signs are
can occur in patients of any age and tients of any age. Rhabdomyosarcoma variable and non-specific, and depend
arise more commonly in supratentorial occurs preferentially in children, whereas largely on tumour location. Spontaneous
than in infratentorial or spinal locations. undifferentiated pleomorphic sarcoma I regression is rare {1390). Whereas the
The clinical symptoms and neuroradio- MFH and chondrosarcoma usually mani- neuroradiological appearance of most
logical appearance of most tumours are fest in adults. As a whole, sarcomas show mesenchymal tumours is non-specific,
non-specific. no obvious sex predilection. the neuroimaging characteristics of li-
The histological features of mesenchy- poma are virtually diagnostic; on MRI,
mal tumours affecting the CNS are similar Etiology T1-weighted images show the high sig-
to those of the corresponding extracra- lntracranial fibrosarcoma, undifferenti- nal intensity of fat, which then disappears
nial soft tissue and bone tumours (7141. ated pleomorphic sarcoma I MFH, chon- with fat-suppression techniques. Speck-
Solitary fibrous tumour I haemangioperi- drosarcoma, and osteosarcoma can oc- led calcifications are typical of chondroid
cytoma (by far the most common mesen- cur several years after irradiation (453, and osseous tumours.
chymal, non-meningothelial neoplasm) 1791}. Isolated cases of intracranial and
and peripheral nerve sheath tumours (the spinal fibrosarcoma, undifferentiated Spread
most common neoplasms of cranial and pleomorphic sarcoma I MFH, and angio- Primary meningeal sarcomatosis is a dif-
paraspinal nerves) are described sepa- sarcoma have also been related to previ- fuse leptomeningeal sarcoma lacking
rately. Antiquated nosological terms, ous trauma or surgery (1020}, an etiology circumscribed masses (2598}. Although
such as "spindle cell sarcoma", "pleo- that may be more common to desmoid- this entity is strictly defined as a non-
morphic sarcoma," and "myxosarcoma" type fibromatosis (1684}. EBV is associ- meningothelial mesenchymal tumour,
have been replaced by designations ated with the development of intracranial most published cases have not been
indicating more specific differentiation smooth muscle tumours in immunocom- diagnosed according to modern nomen-
or updated terminology (714}. The non- promised patients (287). clature. Re-examination of published
specific diagnostic term "meningeal sar- cases using immunohistochemistry has
coma" is also to be avoided, because it Localization revealed that most cases actually con-
has been used in the past to denote both Tumours arising in meninges are more stitute carcinoma, lymphoma, glioma, or
sarcomatoid anaplastic meningiomas common than those originating within embryonal tumour.
and various types of sarcomas. CNS parenchyma or in choroid plexus.
Most mesenchymal tumours are su- Macroscopy
Grading pratentorial, but rhabdomyosarcomas The macroscopic appearance of mesen-
Mesenchymal, non-meningothelial tu- are more often infratentorial. Chondrosar- chymal tumours depends entirely on their
mours range from benign neoplasms comas involving the CNS arise most often differentiation and is similar to that of the
(corresponding histologically to WHO in the skull base. Among benign mesen- corresponding extracraniospinal soft tis-
grade I) to highly malignant sarcomas chymal lesions, intracranial lipomas have sue tumours. Lipomas are bright yellow,
(corresponding histologically to WHO a characteristic location and typically oc- lobulated lesions. Epidural lipomas are
grade IV). cur at midline sites, such as the anterior delicately encapsulated and discrete,
corpus callosum, quadrigeminal plate, whereas intradural examples are often
Epidemiology suprasellar and hypothalamic regions, intimately attached to leptomeninges
The various forms of lipoma account and auditory canal. Spinal cord exam- and CNS parenchyma. Chondromas are
for only 0.4% of all intracranial tumours, ples involve the conus medullaris-filum demarcated, bosselated, greyish-white,

248 Mesenchymal. non-meningothelial tumours


and variably translucent, and typically of meningeal-based Ewing sarcoma I 2 years. Systemic metastasis of intracra-
form large, dural-based masses indent- peripheral primitive neuroectodermal nial sarcoma is relatively common. Nev-
ing brain parenchyma. Meningeal sarco- tumour have shown the typical EWSR1- ertheless, primary CNS sarcomas are
mas are firm in texture and tend to invade type rearrangements found in bone and less aggressive than glioblastomas; in a
adjacent brain. lntracerebral sarcomas soft tissue counterparts (565,1617). series of 18 cases, the estimated 5-year
appear well delineated, but parenchy- survival rates for high-grade and low-
mal invasion is a feature. The cut surface Genetic susceptibility grade primary CNS sarcomas were 28%
of sarcomas is typically firm and fleshy; Several associations with inherited dis- and 83%, respectively (1838}.
high-grade lesions often show necrosis ease have been noted. lntracranial carti-
and haemorrhage. laginous tumours may be associated with
Maffucci syndrome and Oiiier disease; Solitary fibrous tumour I
Cell of origin lipomas with encephalocraniocutaneous haemangiopericytoma
Mesenchymal tumours affecting the CNS lipomatosis; and osteosarcoma with Pa-
are thought to arise from craniospinal get disease. Giannini C.
meninges, vasculature, and surrounding Rushing E.J.
osseous structures. Given that cranial Prognosis and predictive factors Hainfellner JA
and intracranial mesenchymal structures Whereas most benign mesenchymal tu- Bouvier C.
(e.g. bone, cartilage, and muscle) are mours can be completely resected and Figarella-Branger D.
in part derived from the neuroectoderm have a favourable prognosis, primary in- von Deimling A.
(ectomesenchyme), development of the tracranial sarcomas are aggressive and Wesseling P.
corresponding sarcoma types could also associated with a poor outcome. Local Antonescu C.R.
constitute reversion to a more primitive recurrence and/or distant leptomeningeal
stage of differentiation. Lipomas aris- seeding are typical. For example, despite Definition
ing within the CNS are often associated aggressive radiation and chemotherapy, A mesenchymal tumour of fibroblas-
with developmental anomalies, and may almost all reported cases of CNS rhab- tic type, often showing a rich branch-
be congenital malformations rather than domyosarcoma have been fatal within ing vascular pattern, encompassing a
true neoplasms, in particular those with histological spectrum of tumours pre-
partial or complete agenesis of the cor- viously classified separately as men-
pus callosum and spinal dysraphism with ingeal solitary fibrous tumour and
tethered cord (218,998,2823) lntracrani- haemangiopericytoma.
al rhabdomyosarcoma may also be as- Most meningeal solitary fibrous tu-
sociated with malformations of the CNS mours I haemangiopericytomas harbour
(999). a genomic inversion at the 12q13 locus,
20
fusing the NAB2 and STAT6 genes (444,
10 ;-----t-"--'-:::.a-"'!�--i-
Genetic profile 2141}, which leads to STAT6 nuclear
The molecular genetic alterations of expression that can be detected by im-
Age at diagnosis
intracranial sarcomas may be similar to munohistochemistry. Detection of STAT6
Fig. 11.01 Cumulative age distribution (both sexes)
those of corresponding extracranial soft of haemangiopericytoma phenotype (186 cases) and nuclear expression or NAB2-STAT6 fu-
tissue tumours (79}, but few data are solitary fibrous tumour phenotype (157 cases). Data sion is highly recommended to confirm
available to date. However, examples derived from Fargen KM et al. (669). the diagnosis.

Fig. 11.02 Solitary fibrous tumour I haemangiopericytoma. A The falcine tumour in a 51-year-old man shows postgadolinium enhancement to a variable extent. B The tumour of the
posterior Iossa I cerebellopontine angle in a 55-year-old man shows postgadolinium enhancement. C Tumour of the upper thoracic spine in a 45-year-old man (a T1 mass, 1.6 cm in
length) shows postgadolinium enhancement and a dural tail, mimicking meningioma.

Solitary fibrous tumour I baernanoropericytorna 249


Fig. 11.03 Solitary fibrous tumour phenotype. A Patternless architecture characteristic of this phenotype. B The tumour is composed of cells with bland ovoid to spindle-shaped
nuclei and scant eosinophilic cytoplasm. C Stromal and perivascular hyaline collagen deposition and (D) keloidal or amianthoid-like collagen are common.

A negative result should prompt consid- by high cellularity and a delicate, rich with the solitary fibrous tumour pheno-
eration of alternative diagnoses (see Dif- network of reticulin fibres typically invest- type is generally benign, provided gross
ferential diagnosis). When STAT6 immu- ing individual cells. Thin-walled branch- total resection can be achieved, whereas
nohistochemistry or NAB2-STAT6 fusion ing haemangiopericytoma-like (staghorn) tumours with the haemangiopericytoma
testing are not or cannot be performed, vessels are a feature shared by both phenotype have a high rate of recur-
this should be indicated in the report. phenotypes. rence (> 75% at 10 years) and may de-
The histological spectrum encompasses Solitary fibrous tumour I haemangioperi- velop extracranial metastases, especially
two main morphological variants: the cytoma (SFT/HPC) is rare (accounting in bones, lungs, and liver (in -20% of
solitary fibrous tumour phenotype char- for < 1% of all primary CNS tumours) cases).
acterized by a patternless architecture or and most commonly affects adults (in
short fascicular pattern, with alternating the fourth to sixth decade of life) The tu- ICD-0 codes
hypocellular and hypercellular areas with mours are typically dural-based and of- Solitary fibrous tumour I
thick bands of collagen, and the haeman- ten supratentorial, with about 10% being haemang iopericytoma
giopericytoma phenotype characterized spinal. The clinical behaviour of tumours Grade 1 8815/0
Grade 2 8815/1
Grade 3 8815/3

Grading
Outside the CNS (e.g. in the soft tissue,
pleura, and other visceral sites), the term
"haemangiopericytoma" has long been
subsumed into the designation "solitary
fibrous tumour", with unified criteria for
malignancy. These criteria include hy-

.c;: -
·S.- ��,;f_'.'ifiill����i�
Fig. 11.04 Solitary fibrous tumour. A Strong and diffuse positivity for CD34 is typical of this phenotype.
percellularity and necrosis in addition to
elevated mitotic count (which remains
the best indicator of poor outcome) 1714,
localization of STAT6 protein detected by immunohistochemistry. 861l. A mitotic count of > 4 mitoses per

250 Mesenchymal, non-meningothelial tumours


ltuiiti'l•��;'.�-�ll!'i,�i! ,.'i,' :: .:-.-�:.
t���r:t.�mf,:;�� :··:\.""\..,:. _·_...ic:=-__........_...,._._
Fig. 11.05 The haemangiopericytoma phenotype. A Diffuse high cellularity, with thin-walled, branching vessels. B Closely apposed cells with round to ovoid nuclei arranged in a
haphazard pattern, with limited intervening stroma. C Numerous mitoses. D Focal necrosis is typically present.

10 high-power fields is required for the One study proposed a three/four-tiered solitary fibrous tumours I haemangioperi-
designation of solitary fibrous tumour grading scheme (grades I, Ila, llb, and cytomas has significant therapeutic im-
as malignant, irrespective of solitary fi- Ill) after combining the solitary fibrous plications, the current consensus is that it
brous tumour or haemangiopericytoma tumour and haemangiopericytoma des- is still premature to consider replacing the
histopathological phenotype {714}. In the ignations and applying common (but current three-tiered with another grading
CNS, the classification and grading of slightly different) criteria based on hy- system. Larger studies are needed to de-
these tumours has been somewhat dif- percellularity, necrosis, and mitotic count termine whether the malignancy defini-
ferent, resulting in a three-tiered system, (::; 5 vs > 5 mitoses per 10 high-power tion (i.e. > 4 mitoses per 10 high-power
with grade I tumours considered benign fields). In that study, mitotic count was fields), which is applied to non-CNS sites
and typically treated by surgical resec- found to be the only independent prog- irrespective of solitary fibrous tumour or
tion alone, and grade II and Ill tumours nostic factor for progression-free and haemangiopericytoma histopathologi-
considered malignant and treated with overall survival in multivariate analysis cal phenotype, is relevant to meningeal
adjuvant therapy, typically radiotherapy. {256). Because the distinction between locations.
A hypocellular, collagenized tumour with grade I versus grades II and Ill meningeal
a classic solitary fibrous tumour pheno-
type is considered to correspond histo-
logically to grade I, whereas more dense-
ly cellular tumours mostly corresponding
to the haemangiopericytoma phenotype
are considered malignant. Tumours with
a haemangiopericytoma phenotype are
subclassified as grade 11 or grade 111
(anaplastic) depending on mitotic count
(< 5 vs � 5 mitoses per 10 high-power
fields) {1533,1637). Patients diagnosed
with grade II or Ill haemangiopericytoma
benefit from adjuvant radiotherapy (813, Fig. 11.06 Haemangiopericytoma. A Only focal positivity for CD34 is present in this malignant tumour. B Nuclear
814). localization of STAT6 protein is detected by immunohistochemistry.

Solitary fibrous tumour I haemangiopericytoma 251


gland {2854). and sellar region {1189)

Clinical features
In most cases, the symptoms and signs
are consistent with their localization
accompanied by mass effect, with in�
creased intracranial pressure due to
tumour size {859,1637} Massive intra-
cranial haemorrhage {1602} and hypo-
glycaemia from tumours that release
insulin-like growth factor {2391 l are rare
complications.

Fig. 11.07 lntradural extramedullary solitary fibrous tumour I haemangiopericytoma. A T1-weighted MRI. lsointense Imaging
tumour (15 mm x 15 mm x 15 mm) located at T10-T11. B T2-weighted MRI. C Postcontrast T1-weighted MRI. The Plain CT images show solitary, irregularly
lesion is homogeneously and diffusely enhanced after gadolinium injection.
contoured masses without calcifications
or hyperostosis of the adjacent skull.
On MRI, the tumours are isointense on
T1-weighted images, with high or mixed
intensity on T2-weighted images, along
with variable contrast enhancement. Du-
ral contrast enhancement at the periph-
ery of the lesion (dural tail) and flow voids
may be observed {2693} Arteriography
reveals a hypervascular mass with promi-
nent draining veins.

Macroscopy
Solitary fibrous tumours I haemangioperi-
cytomas are usually well-circumscribed,
firm white to reddish-brown masses, de-
pending on the degree of collagenous
stroma and cellularity. Occasionally, they
show infiltrative growth or lack dural at-
tachment {365,377,1656} Variable myxoid
or haemorrhagic changes may be present.

Microscopy
Solitary fibrous tumour I haemangioperi-
hypercellular areas. B Highly cellular area. cytoma shows two main distinct histo-
expressed in all tumour nuclei. logical phenotypes, although cases with
intermediate/hybrid morphology are also
Epidemiology Age and sex distnbution seen. The classic solitary fibrous tumour
The true incidence and prevalence of Peak incidence occurs in the fourth to phenotype shows a patternless archi-
this entity are difficult to ascertain, due fifth decade of life, and males are affect- tecture characterized by a combination
to inconsistent nomenclature. In the 2014 ed slightly rnore frequently than females of hypocellular and hypercellular areas
statistical report published by the Central {522,1637,2269} Primary CNS solitary separated by thick bands of hyalinized,
Brain Tumor Registry of the United States fibrous tumours I haemangiopericyto- sometimes keloidal or amianthoid-like
(CBTRUS), because of their rarity, soli- mas have been reported in the paediatric collagen and thin-walled branching
tary fibrous tumour I haemangiopericyto- population, although they are exceed- haemangiopericytoma-like (stag horn)
mas are grouped with other mesenchy- ingly rare {1623,2550) vessels. The tumour cells have a mono-
mal tumours of the meninges, which as morphic ovoid to spindle-shaped cy-
a group have an average annual age-ad- Localization tomorphology, with scant eosinophilic
justed incidence rate of 0.08 cases per Most solitary fibrous tumours I haeman- cytoplasm. The nuclei are round or
100 000 population {1863). Data from giopericytomas are dural-based (often oval, with moderately dense chromatin
large series suggest that solitary fibrous supratentorial), and about 10% are spi- and inconspicuous nucleoli lacking the
tumour I haemangiopericytomas consti- nal. Skull base, parasagittal, and falcine pseudoinclusions characteristic of men-
tute < 1% of all CNS tumours {522,859, locations are especially common {1637, ingiomas. Mitoses are generally scarce,
1637,2269). 2269). Uncommon locations include the rarely exceeding 3 per 10 high-power
cerebellopontine angle {2516}. pineal fields {365,714) In contrast, the classic

252 Mesenchymal, non-meningothelial tumours


haemangiopericytoma phenotype is A
characterized by high cellularity, with
closely apposed ovoid cells arranged in
STATS protein
a haphazard pattern with limited interven- N-ter PIO DNA bind. dom. SH2 TAD
ing stroma (1637). Mitotic activity and ne-
crosis are often present, but are variable.
A rich network of reticulin fibres typically NAB2 protein
invests individual or small groups of tu-
N-ter
mour cells. Invasion of brain parenchyma NCD1 --- NCD2
or engulfment of vessels or nerves may
be noted in either pattern (1656). Calci- NAB2-STAT6fusion protein
fications, including psammoma bodies,
are not seen. Rarely, a variably prominent N-ter NCD1 ---... NCD2 SH2 TAD
adipocytic component may be present.

Differential diagnosis
The differential diagnosis includes both B
meningothelial and soft tissue neo-
plasms. Fibrous meningioma is a close
other tumors HPC/SFT
mimic of solitary fibrous tumour (365}, but
typically expresses EMA and is negative
for CD34 and nuclear STAT6 expression.

)
Dural-based Ewing sarcoma I peripheral
primitive neuroectodermal tumour shares
the hypercellularity and CD99 positivity of
haemangiopericytoma, but lacks nuclear
STAT6 staining and is characterized by
EW5R1 gene rearrangement in the great
majority of cases (565). Both primary NAB2 wt, STAT6 wt NAB2-STAT6 fusion
and metastatic monophasic synovial sar-
comas can simulate solitary fibrous tu- Fig. 11.09 Solitary fibrous tumour I haemangiopericytoma (HPC/SFT). A The break point in STAT6 varies, but is
mour I haemangiopericytoma. lmmuno- within or N-terminal to the SH2 domain. The break point in NAB2 also varies, but is usually C-terminal to the nuclear
localization sequence (N). B Preservation of the NAB2 nuclear localization signal results in nuclear localization of the
reactivity for EMA and TLE1 and/or FISH fusion protein {2308). All-a. dom., all-alpha domain; DNA bind. dom., DNA-binding domain; NCD1, NAB-conserved
analysis for the presence of 5518 gene domain 1; NCD2, NAB-conserved domain 2; N-ter, N-terminus; PIO, protein interaction domain; SH2, SRC homology
rearrangement support this diagnosis domain; TAD, transcriptional activator domain; wt, wildtype.
(1507). Mesenchymal chondrosarcoma,
a rare malignant tumour with a bimorphic
pattern, is composed of sheets and nests The individual tumour cells are sur- proximity ligation assay (1317,2308). Other
of poorly differentiated small round cells rounded by electron-dense, extracellular markers, such as desmin, SMA, cytokera-
interrupted by islands of well-differentiat- basement membrane-like material - the tin, EMA, and progesterone receptor, may
ed hyaline cartilage and a branching vas- ultrastructural equivalent of the reticulin be rarely encountered as a focal finding
culature. Because the cartilage islands network visible on light microscopy. True (1656,1949,2066,27641. ALDH1A1 gene
can be extremely focal, this entity may desmosomes and gap junctions (as seen overexpression, which was initially detect-
be mistaken for malignant solitary fibrous in meningiomas) are absent (524). ed by microarray analysis, can be demon-
tumour I haemangiopericytoma if insuffi- strated by immunohistochemistry showing
ciently sampled (714). Malignant periph- lmmunophenotype ALDH1 positivity in 84% of cases, com-
eral nerve sheath tumour rarely occurs Solitary fibrous tumours I haemangioperi- pared to only 1% of meningiomas (2551.
in the meninges and may resemble the cytomas are typically diffusely positive for
haemangiopericytoma phenotype. How- vimentin and CD34 (1949}, although loss of Proliferation
ever, malignant peripheral nerve sheath CD34 expression is common in malignant The median Ki-67 proliferation index
tumour is usually negative for CD34 and tumours. The NAB2-5TAT6 fusion leads to was 10% (range: 0.6-36%) in a series of
STAT6 and may show focal expression of nuclear relocalization of STAT6 protein and 31 haemangiopericytomas (2039} An-
8100 protein and SOX10. can be detected with very high specific- other study reported a median Ki-67 pro-
ity and sensitivity by immunohistochemis- liferation index of 5% in a series of 29 sol-
Electron microscopy try (1317,1866,23081. Meningiomas show itary fibrous tumours and 10% in a series
Solitary fibrous tumours I haemangio- faint nuclear and/or cytoplasmic positiv- of 43 cellular solitary fibrous tumours I
pericytomas are composed of closely ity, but no strong isolated nuclear STAT6 haemangiopericytomas (256)
apposed elongated cells with short pro- immunostaining (2308). The NAB2-5TAT6
cesses that contain small bundles of in- fusion can also be detected at the protein Cell of origin
tracytoplasmic intermediate filaments. level with high specificity and sensitivity by The histogenesis of CNS solitary fibrous

Solitary fibrous tumour I haemangiopericytoma 253


,

Fig. 11.1 O Haemangioblastoma. A MRI of a cerebellar haemangioblastoma. The tumour is multicystic and enhancing. B MRI shows a cerebellar haemangioblastoma with an
enhancing mural nodule. Note the compressed fourth ventricle in front of the tumour. C MRI of a cervical spinal cord haemangioblastoma. There is an associated large cyst around
the enhancing mural nodule (arrow).

tumour/haemangiopericytoma remains a is traditionally considered to be benign, considered to be grade I and the hae-
matter of debate. Their fibroblastic nature provided that gross total resection can mangiopericytoma phenotype grade II or
and the presence of a common NAB2- be achieved and atypical histological 111) may need to be adapted to improve
STAT6 gene fusion (444,2141,2308) are features are absent (365). The prognostic the prognostic significance.
strong arguments for grouping CNS soli- significance of focal (rather than general-
tary fibrous tumour and haemangioperi- ized) atypia in these tumours and of inva-
cytoma together under the term "solitary sion in bone and dural sinuses is unclear Haemangioblastoma
fibrous tumour I haemangiopericytoma". (209,669}. In contrast, even after gross to-
tal resection, tumours with the haemangi- Plate K.H.
Genetic profile opericytoma phenotype have a high rate Aldape K.D.
The central (and to date specific) genetic of recurrence (> 75% in patients followed Vortmeyer A.O.
abnormality in solitary fibrous tumour I for> 10 years). In addition, about 20% of Zagzag D.
haemangiopericytoma is a genomic in- patients with tumours of the haemangio- Neumann H.P.H.
version at the 12q13 locus, fusing the pericytoma phenotype develop extracra-
NAB2 and STAT6 genes in a common nial metastases, especially to bone, lung, Definition
direction of transcription (444,2141). This and liver (209,815,903,2673}. Gross total A tumour histologically characterized by
gene fusion is present in most cases, re- resection favourably affects recurrence neoplastic stromal cells and abundant
gardless of histological grade or anatom- and survival, and patients benefit from small vessels.
ical location (meningeal, pleural, or soft adjuvant radiotherapy (813,814,815,903}. Haemangioblastoma is a benign slow-
tissue). The recent discovery of NAB2- As discussed previously, the historical growing tumour of adults, typically oc-
STAT6 fusion in the majority of solitary three-tiered grading system (in which curring in the brain stem, cerebellum.
fibrous tumours I haemangiopericytomas the solitary fibrous tumour phenotype is and spinal cord. Haemangioblastomas
has provided strong evidence for a mor-
phological continuum. The presence of
the NAB2-STAT6 fusion protein results in
nuclear localization of STAT6 and strong
nuclear positivity for STAT6 by immuno-
histochemistry (2308}.
In contrast, the rare sinonasal haemangi-
opericytoma does not exhibit the NAB2-
STAT6 fusion, but instead harbours CTN-
NB1 mutations (18,929).

Genetic susceptibility
There is no evidence of familial cluster-
ing of meningeal solitary fibrous tumour I
haemangiopericytoma.

Prognosis and predictive factors B


The clinical behaviour of tumours with Fig. 11.11 Haemangioblastoma. A,B The tumour is multicystic (white arrow) and well demarcated from the surrounding
the solitary fibrous tumour phenotype cerebellum (black arrows).

254 Mesenchymal, non-meningothelial tumours


Fig. 11.12 Haemangioblastoma. Cerebral angiogram Fig. 11.13 Cerebellar haemangioblastoma. A CT image. B CT angiography shows the tumour receiving vascularization
demonstrates two tumours; the larger one is in the right from the left superior cerebellar artery.
cerebellar hemisphere (black arrow) and the smaller one
is in the left cerebellar hemisphere (white arrow); both Localization Imaging
are hypervascular. Haemangioblastomas can occur in any Haemangioblastomas have a charac-
part of the nervous system. Sporadic teristic radiographical appearance on
tumours occur predominantly in the cer- both CT and MRI, as well as a distinct
occur in sporadic forms (accounting for ebellum, usually in the hemispheres (in prolonged vascular stain on cerebral
-70% of cases) and in association with -80% of cases). Haemangioblastomas angiography {112). MRI is the preferred
the inherited von Hippel-Lindau disease associated with VHL are often multiple imaging modality, but in rare cases an-
(VHL) (accounting for -30% of cases). (in 65% of patients) and affect the brain giography better detects occult vascu-
The VHL tumour suppressor gene is in- stem, spinal cord, and nerve roots in ad- lar nodules that may not be apparent on
activated both in VHL-associated cases dition to the cerebellum. Supratentorial standard imaging. MRI studies typically
and in most sporadic cases. and peripheral nervous system lesions show a gadolinium-enhancing mass, with
are rare. an associated cyst in approximately 75%
ICD-0 code 9161/1 of cases. The solid component is usually
Clinical features peripherally located within the cerebel-
Grading Symptoms of intracranial haemangioblas- lar hemisphere. Flow voids may be seen
Haemangioblastoma corresponds histo- toma generally arise from impaired cere- within the nodule due to enlarged feed-
logically to WHO grade I. brospinal fluid flow due to a cyst or solid ing/draining vessels. Evidence of calcifi-
tumour mass, resulting in an increase of cation is usually absent on imaging. Spi-
Epidemiology intracranial pressure and hydrocephalus. nal cord haemangioblastomas are often
Haemangioblastomas are uncommon tu- Cerebellar deficits such as dysmetria and associated with a syrinx.
mours that occur as sporadic lesions and ataxia can also occur. Spinal tumours
in familial forms associated with VHL. Ac- become symptomatic due to local com- Macroscopy
curate incidence rates are not available. pression resulting in symptoms such as Most haemangioblastomas (60%) pres-
Haemangioblastomas usually occur in pain, hypaesthesia, and incontinence. ent as well-circumscribed, partly cystic,
adults. The average patient age at pre- Haemangioblastomas produce erythro- highly vascularized lesions; about 40%
sentation of VHL-associated tumours is poietin, which causes secondary poly- are completely solid. Occasionally, the
approximately 20 years younger than that cythaemia in about 5% of patients {853). tumour is yellow due to rich lipid content.
of sporadic tumours {1567). The male-to- Haemorrhage is a rare complication of The classic appearance is that of a cyst
female ratio is approximately 1 :1. CNS haemangioblastoma {639,855). with a solid vascular nodule that abuts a
pial surface {2683).

Haemangioblastoma 255
. -�·..,....
,, ,... ...

·� .. ·
..-
,-
(
surrounding neural tissues rarely occurs .
� Mitotic figures are rare. Stromal cells ac-
count for only 10-20% of the cells and
constitute the neoplastic component of
the tumour. Their nuclei can vary in size
with occasional atypical and hyperchro�
matic nuclei. However, haemangioblasto-
ma's most characteristic and distinguish-
ing morphological feature is numerous
lipid-containing vacuoles, resulting in the
typical clear-cell morphology of haeman-
gioblastoma, which may resemble meta-
static renal cell carcinoma (RCC). The
fact that patients with VHL are also prone
to RCC adds to the complexity of this
differential diagnosis. Cases of tumour-
to-tumour metastasis (RCC metastatic
to haemangioblastoma) have also been
reported in this setting 1934}

lmmunophenotype
The stromal and capillary endothelial cells
differ significantly in their expression pat-
terns. Stromal cells lack endothelial cell
markers, such as von Willebrand factor
and CD34, and do not express endotheli-
Microscopy Numerous thin-walled vessels are ap- um-associated adhesion molecules such
Haemangioblastomas are character- parent and are readily outlined by a reti- as CD31 1336,2769). Unlike endothe-
ized by two main components: (1) stro- culin stain. In accordance with the highly lial cells, stromal cells variably express
mal cells that are characteristically large vascular nature of haemangioblastoma, neuron-specific enolase, NCAM1, 8100,
and vacuolated but can show consider· intratumoural haemorrhage may occur. and ezrin 1336,337,1100) Vimentin is the
able cytological variation and (2) abun- In adjacent reactive tissues, particularly major intermediate filament expressed by
dant vascular cells. Cellular and reticular in cyst and syrinx walls, astrocytic glio- stromal cells. Stromal cells also express
variants of haemangioblastoma are dis- sis and Rosenthal fibres are frequently a variety of other proteins, including
tinguished by the abundance and mor- observed. The tumour edge is gener- CXCR4 11498,2839}. aquaporin-1 l1524l,
phology of the stromal cell component. ally well demarcated, and infiltration into brachyury, several carbonic anhydrase
isozymes 12040). and EGFR 1335). but
they do not usually express GFAP 12769).
Table 11.01 Common immunohistochemical profiles of haemangioblastoma and renal cell carcinoma Both HIF1A 12841) and HIF2A 1712} have
Molecule Haemangioblastoma Renal cell carcinoma been detected in stromal cells, and they
Aquaporin-1 + {428,2720) +/- {2720) may drive the expression of VEGF, which
is also abundant in stromal cells 11363}.
Brachyury + {133) - (133)
The corresponding receptors VEGFR1
CD10 - {1999,2720); +/- {2132) + {1999,2132,2720) and VEGFR2 12768). are expressed on
AE1/AE3 - {577,2720) + {577,2720) endothelial cells, suggesting a paracrine
CAM5.2 - {1999) + {1999) mode of angiogenesis activation 1971,
2437). The endothelial cells of haeman-
02-40 +/- {2132); + {2193) +!- {2132); - {2193)
gioblastomas also express receptors for
EMA +/- {2720) + {2720) other angiogenic growth factors, includ-
- {366,577,1021,1999}; ing platelet-derived growth factors !335)
lnhibin alpha + {366,577,1021,1999,2132,2720}
+/- {2132,2720) lmmunohistochemistry is useful for distin-
LEU? +!- {1061); + {1999) - {1061); +/- {1999) guishing haemangioblastoma from RCC.
NCAM1 (also called CD56) + {1999) - {1999) RCCs are positive for epithelial markers
such as EMA, whereas haemangioblas-
Neuron-specific enolase +/- {1061) +!- {1061)
tomas are negative. Additional potentially
PAX2 - {366,2132) + {366,2132) useful markers include 02-40 12193} and
PAX8 - {366) + {366) inhibin alpha 11021). which are both posi-
tive in haemangioblastoma but generally
Renal cell carcinoma marker - {1093,2132) + {1093); +/- {2132)
negative in RCC. CD10 staining shows
8100 +/- {1061) +/- {1061)
the opposite results 11193).

256 Mesenchymal, non-meningothelial tumours


choroid plexus cells {215). neuroendo- {873) The VHL tumour suppressor pro-
crine cells {153). fibrohistiocytic cells tein controls cell cycle regulation and, in
{1767). cells of neuroectodermal deriva- a prolyl hydroxylase-dependent manner,
tion {12). and heterogeneous cell popu- the degradation of HIF1A and HIF2A.
lations {2512). More recent study re- As a consequence, loss of function of
ports have noted that the stromal cells VHL leads to upregulation of hypoxia-re-
of haemangioblastoma express proteins sponsive genes such as those encoding
common to embryonal haemangioblast VEGF and erythropoietin, despite the ab-
progenitor cells {854). One such protein, sence of tumour hypoxia. This condition
SCL, has a distribution of expression in is called pseudohypoxia {680)
the developing nervous system that is
similar to the topographical distribution Genetic susceptibility
of haemangioblastoma tumours in pa- Haemangioblastoma is a frequent mani-
Cell of origin tients, suggesting that stromal cells may festation of VHL (occurring in 60% of
Stromal cells constitute the neoplastic be haemangioblasts or haemangioblast patients) {850). Genetic counselling and
cell type in haemangioblastoma {2437, progenitor cells {854,2352,2669} molecular genetic screening for germline
2670l and are surrounded by abundant mutations in the VHL gene are therefore
non-neoplastic cells, including endothe- Genetic profile essential for patients with haemangio-
lial cells, pericytes, and lymphocytes. Biallelic inactivation of the VHL gene is blastoma {850l
This intercellular genomic heterogeneity a frequent occurrence in familial hae-
suggests that the majority of nucleated mangioblastomas, but is not common Prognosis and predictive factors
cells in the tumour mass do not arise in sporadic tumours. However, studies The prognosis of sporadic CNS haeman-
from a common ancestral clone, support- on sporadic tumours (including somatic gioblastoma is excellent if surgical re-
ing the hypothesis that the tumour mass mutation analyses, assessments of al- section can be performed successfully,
histology is a consequence of reactive lelic loss, deep-coverage DNA sequenc- which is typically possible. Permanent
angiogenesis resulting from paracrine ing, and hypermethylation studies) have neurological deficits are rare {490} and
signalling mediated by the VHL tumour found loss or inactivation of the VHL gene can be avoided when CNS haemangio-
suppressor protein-deficient HIF1A- in as many as 78% of cases {851,1456, blastomas are diagnosed and treated
expressing stromal cells {2329,2437). 2329). suggesting that loss of function early {852). Patients with sporadic tu-
The exact origin of these stromal cells of VHL is a central event in haemangio- mours have a better outcome than those
is still controversial, but on the basis of blastoma formation. No other gene has with tumours associated with VHL, be-
immunohistochemical expression data, been found to be significantly mutated cause patients with VHL tend to develop
various cell types have been suggested, {2329l. The VHL tumour suppressor pro- multiple lesions {1687)
including glial cells {56). endothelial cells tein forms a ternary complex (called the
{1194). arachnoid cells {1695). embryonic VCB complex) with TCEB1 and TCEB2

Haemangioblastoma 257
Other Antonescu C.R. Bouvier C. Epithelioid
Paulus W. Figarella-Branger D. haemangioendothelioma
mesenchymal Perry A.
Rushing E.J.
von Deimling A.
Wesseling P. Definition
tumours Hainfellner JA A low-grade malignant vascular neo-
plasm characterized by the presence of
epithelioid endothelial cells, arranged in
Haemangioma cords and single cells embedded in a
distinctive chondromyxoid or hyalinized
Definition stroma.
A benign vascular neoplasm greatly vary- Epithelioid haemangioendothelioma is
ing in size. Most haemangiomas affecting rarely located in the skull base, dura, or
the CNS are primary lesions of bone that brain parenchyma {106,1802l. Its cells
impinge on the CNS secondarily. Dural (1, contain relatively abundant eosinophilic
1244} and parenchymal (2361} haeman- cytoplasm, which may be vacuolated.
giomas are less common. In general, the nuclei are round or occa-
Histologically, haemangiomas have pre- sionally indented or vesicular, and show
dominantly capillary-type growth and oc- only minor atypia. Mitoses and limited
cur mostly in the paediatrics age group. Fig. 11.17 Capillary haemangioma. Reddish-brown necrosis may be seen. Small intracyto-
Infantile haemangiomas are consistently sponge-like appearance of an intraosseous capillary plasmic lumina (blister cells) are seen,
positive for GLUT1, a marker that is typi- haemangioma on cut surface of a skull resection but well-formed vascular channels are
cally used to distinguish haemangioma specimen. typically absent. lmmunohistochemical
from arteriovenous malformation in ex- studies (e.g. for CD31 and ERG) confirm
tracranial locations (409l. However, in fibrotic walls. Cerebral cavernous mal- these tumours' endothelial nature. Ap-
a recent study, both cerebral cavern- formations can be inherited as an auto- proximately 90% of epithelioid haeman-
ous malformations and cerebral arterio- somal dominant disorder (accounting for gioendotheliomas harbour the recurrent
venous malformations showed endotheli- 20% of cases), but most are sporadic. t(1 ;3)(p36;q25) translocation, which re-
al immunoexpression of GLUT1 and thus Biallelic somatic and germline mutations sults in a WWTR1-CAMTA1 fusion (651,
seemed to differ from most other arterio- in one of the cerebral cavernous malfor- 2510l. A smaller subset of epithelioid
venous malformations {1632l. mation genes have been implicated as a haemangioendotheliomas have a t(x;11)
two-hit mechanism of inherited cerebral
Ma/formative vascular lesions cavernous malformation pathogenesis
Most vascular lesions of the CNS are l33l. Another tumefactive vascular lesion
malformative in nature, including cerebral that can occur in the brain or meninges
cavernous malformation (also called cav- is intravascular papillary endothelial hy-
ernous angioma or cavernoma), arterio- perplasia, a papillary proliferation of en-
venous malformation, venous angioma, dothelium associated with thrombosis
and capillary telangiectasia. Cerebral (1366l.
cavernous malformations typically occur
in the brain, but the spinal cord and the ICD-0 code 9120/0
eye may also be involved. Histologically,
cerebral cavernous malformations are Fig. 11.18 Epithelioid haemangioendothelioma.
composed of an admixture of capillary- lntracytoplasmic lumina and a basophilic stroma are
prominent features of this example.
type and large saccular vessels with

258 Mesenchymal, non-meningothelial tumours


(p11 ;q22) translocation, which results in a Kaposi sarcoma A wide patient age range has been re-
YAP1-TFE3 fusion {80l ported, although peak incidence occurs
Definition in the second decade of life. The histol-
ICD-0 code 9133/3 A malignant neoplasm characterized by ogy, immunophenotype, and biology are
spindle-shaped cells forming slit-like essentially identical to those of tumours
blood vessels, which is only rarely en- encountered in bone or soft tissue {714l.
Angiosarcoma countered as a parenchymal or meninge- The tumour is composed of sheets of
al tumour, typically in the setting of HIV small, round, primitive-appearing cells
Definition type 1 infection or AIDS {115,334). with scant clear cytoplasm and uniform
A high-grade malignant neoplasm with In this setting, it is often difficult to deter- nuclei with fine chromatin and smooth
evidence of endothelial differentiation. mine whether a Kaposi sarcoma lesion nuclear contours. Homer Wright rosettes
The rare examples of angiosarcoma that is primary or metastatic. The tumour is are occasionally seen, but are usually not
originate in brain or meninges {1636l almost always immunopositive for HHV8 prominent. Most tumours stain at least
vary in differentiation from patently vas- {2140l. focally with synaptophysin and neuron-
cular tumours with anastomosing vascu- specific enolase, whereas cytokeratin
lar channels lined by mitotically active, ICD-0 code 9140/3 is only focally seen (in as many as 20%
cytologically atypical endothelial cells of cases). CD99 shows strong and dif-
to poorly differentiated, often epithelioid fuse membranous immunoreactivity in
solid lesions, in which immunoreactivity Ewing sarcoma I peripheral the vast majority. CD99 can also be ex-
for vascular markers (e.g. CD31, CD34, primitive neuroectodermal pressed (although this expression is usu-
ERG, and FLl1) is required for definitive ally more patchy and cytoplasmic) in oth-
tumour
diagnosis. Occasional cytokeratin reac- er tumour types, including solitary fibrous
tivity complicates the distinction of poorly Definition tumour I haemangiopericytoma and (less
differentiated angiosarcoma from meta- A small round blue cell tumour of neuro- commonly), medulloblastoma or other
static carcinoma {2131l. No genetic ab- ectodermal origin that involves the CNS embryonal CNS neoplasms. Therefore,
normalities have been described for CNS either as a primary dural neoplasm {565, most Ewing sarcoma I peripheral primi-
angiosarcomas. 1617, 1696,2358} or by direct extension tive neuroectodermal tumours must be
from contiguous bone or soft tissue (e.g. confirmed at the molecular level either by
ICD-0 code 9120/3 skull, vertebra, or paraspinal soft tissue). RT-PCR for the presence of an EWSR1-
Radiologically, primary CNS Ewing sar- FL/1 or EWSR1-ERG fusion transcript or
coma I peripheral primitive neuroecto- by FISH for EWSR1 gene rearrangement
dermal tumour can mimic meningioma. {279l. Alternative gene fusions in the

B Diffuse membrane immunoreactivity for CD99. C FISH results positive for

Angiosarcoma 259
site, may incorporate intradural portions Liposarcoma
of cranial nerve roots and their ganglia.
Many also feature striated muscle or oth- Definition
er mesenchymal tissues. Some of these A malignant tumour composed entirely or
lesions have been referred to as choristo- partly of neoplastic adipocytes.
mas. It has even been suggested that lntracranial liposarcoma is extremely
intracranial lipomas containing various rare. An example associated with sub-
other tissue types represent a transition dural haematoma has been described
between lipoma and teratoma {2572). {461}, and an example of gliosarcoma
Whether these various lesions are neo- with a liposarcomatous element has
Fig. 11.22 Postcontrast T1-weighted MRI of a
plasms or malformative overgrowths is been reported {242}.
dural-based Ewing sarcoma I peripheral primitive yet to be determined.
neuroectodermal tumour mimicking meningioma. ICD-0 code 8850/3
Epidural lipomatosis
Ewing sarcoma-like family of tumours Epidural lipomatosis is a rare lesion that
have also been described, including consists of diffuse hypertrophy of spinal Desmoid-type fibromatosis
CIC-DUX4 {1104) and the BCOR-CCNB3 epidural adipose tissue. As such, it is not
intrachromosomal inversion {1971). a neoplasm but a metabolic response, Definition
often to chronic administration of steroids A locally infiltrative but cytologically be-
ICD-0 code 9364/3 {919). nign lesion composed of uniform my-
ofibroblastic-type cells in an abundant
collagenous stroma, arranged in inter-
Lipoma Angiolipoma secting fascicles {1684}.
Desmoid-type fibromatosis must be dis-
Definition Definition tinguished from cranial fasciitis of child-
A benign lesion that microscopically re- A lipoma variant with prominent vascular- hood, a process histologically related to
sembles normal adipose tissue {306} ity; by definition, the vessels are of cap- nodular fasciitis, featuring rapid growth
lntracranial lipomas are believed to be illary type and are generally most prom- within the deep scalp, lacking an intra-
congenital malformations rather than true inent in the periphery, often containing dural component, and having no ma-
neoplasms, resulting from abnormal dif- dispersed fibrin thrombi. lignant potential {1438). Cranial infantile
ferentiation of the meninx primitiva (the The proportions of adipose cells and myofibromatosis also enters into the dif-
undifferentiated mesenchyme) (1203). vasculature in angiolipoma vary {1982, ferential diagnosis but often displays a
2367}. With time, interstitial fibrosis may biphasic pattern, with alternating hae-
ICD-0 code 8850/0 ensue. Angiolipomas may be overdiag- mangiopericytoma-like areas and myoid
nosed because ordinary haemangiomas components {2479}.
Microscopy are often accompanied by fat; however,
ordinary haemangiomas have a more ICD-0 code 8821/1
Lipoma heterogeneous spectrum of vascular
Most lipomas show lobulation at low mag- channels, including thick-walled venous-
nification. Lipomas have an inconspicu- type vascular channels and cavernous Myofibroblastoma
ous capillary network. Patchy fibrosis is spaces.
a common feature, and calcification and Definition
myxoid change are occasionally seen. ICD-0 code 8861/0 A benign mesenchymal neoplasm com-
Osteolipomas are exceedingly rare and posed of spindle-shaped cells with fea-
may show zonation, with central adipose tures of myofibroblasts embedded in a
tissue and peripheral bone {2367). Hibernoma stroma that contains coarse bands of hy-
alinized collagen and conspicuous mast
Complex lipomatous lesions Definition cells, admixed with a variable amount of
Complex lipomatous lesions vary con- A very rare lipoma variant within the CNS, adipose tissue {714}
siderably in terms of their histological composed of uniform granular or multi- Myofibroblastomas of the CNS are simi-
composition. For example, lumbosacral vacuolated cells with small, centrally lo- lar to their mammary-type counterparts,
lipomas (leptomyelolipomas) that occur cated nuclei, resembling brown fat {442}. being part of a larger spectrum of CD34-
in the context of tethered spinal cord syn- positive lesions (including spindle cell
drome are likely malformative and may ICD-0 code 8880/0 lipoma and cellular angiofibroma) that
consist of subcutaneous and intradural show common 13q14 losses by FISH or
components linked by a fibrolipomatous loss of RB immunoexpression {420,742}.
stalk that may attach to the dorsum of the Myofibroblastomas are also strongly pos-
cord or to the filum terminale {998). Lipo- itive for desmin {2348}.
mas of the cerebellopontine angle {201},
an uncommonly affected off-midline ICD-0 code 8825/0

260 Mesenchymal, non-meningothelial tumours


Scattered giant cells and/or inflammatory
cells are commonly seen. Many tumours
that had an intraparenchymal component
and were initially described as fibrous
xanthoma were subsequently shown to
be GFAP-positive and reclassified as
pleomorphic xanthoastrocytoma (1254)

ICD-0 code 8830/0

Fibrosarcoma
Definition
A rare sarcoma type showing monomor-
phic spindle cells arranged in intersect-
ing fascicles (a so-called herringbone
pattern).
Fibrosarcomas are markedly cellular, ex-
hibit brisk mitotic activity, and often fea-
ture necrosis (789). Similar histological
features are seen in adult-type fibrosar-
coma; congenital/infantile fibrosarcoma;
and fibrosarcomatous transformation in
the setting of a solitary fibrous tumour I
haemangiopericytoma or dermatofibro-
Fig. 11.23 Fibrosarcoma. A This hypercellular radiation-induced neoplasm shows features of fibrosarcoma with sarcoma protuberans. Sclerosing epithe-
intersecting fascicles of spindled cells; note the entrapped island of gliotic brain tissue. B lmmunostain for GFAP lioid fibrosarcoma affecting the CNS has
highlights entrapped islands of gliotic brain, whereas tumour cells are negative. C Increased mitotic activity. also been reported (205)

Inflammatory myofibroblastic inflammatory myofibroblastic tumour, in- ICD-0 code 8810/3


tumour cluding ROS1, PDGFRB. and RET (81,
1539} Most inflammatory myofibroblastic
Definition tumours have favourable outcomes after Undifferentiated pleomorphic
A distinctive neoplastic proliferation. usu- gross total resection. sarcoma I malignant fibrous
ally composed of bland myofibroblas-
histiocytoma
tic-type cells intimately associated with a ICD-0 code 8825/1
variable lymphoplasmacytic infiltrate and Definition
arranged in loose fascicles within an oe- The differential diagnosis of inflammatory A malignant neoplasm composed of
dematous stroma. myofibroblastic tumour includes hypertro- spindled, plump, and p/eomorphic giant
The term "inflammatory myofibroblas- phic intracranial pachymeningitis. a pseu- cells that can be arranged in a storiform
tic tumour" was once considered to be dotumoural lesion that entails progressive or fascicular pattern.
synonymous with inflammatory pseudo- dural thickening due to pachymeningeal Most cases of undifferentiated pleomor-
tumour or plasma cell granuloma and fibrosis and chronic inflammation. These phic sarcoma I malignant fibrous histio-
to be a variant of inflammatory fibrosar- lesions are often associated with autoim- cytoma are overtly malignant. featuring
coma. Inflammatory myofibroblastic tu- mune disorders (-2507). and some have
mour of the CNS is rare and can occur recently been reclassified as lgG4-relat-
in patients of any age. The radiological ed disease (1509}.
characteristics of these tumours are of-
ten similar to those of meningiomas. All
three patterns reported in inflammatory Benign fibrous histiocytoma
myofibroblastic tumours of soft tissue
(myxoid-nodular fasciitis-like, fibroma- Definition
tosis-like, and scar-like) have also been A lesion composed of a mixture of spin-
observed in the CNS. Approximately dled (fibroblast-like) and plump (tustio-
50% of all inflammatory myofibroblastic cyte-like) cells arranged in a storiform
tumours harbour ALK gene rearrange- pattern.
Fig. 11.24 Undifferentiated pleomorphic sarcoma.
ment and overexpress ALK (81}. Gene Benign fibrous histiocytoma (also called This highly pleomorphic dural-based sarcoma shows
fusions involving other kinases have also fibrous xanthoma or fibroxanthoma) immunoreactivity for vimentin, but no expression of more
been implicated in the pathogenesis of may involve dura or cranial bone (2604). specific lineage markers.

Inflammatory myofibroblastic tumour 261


-,

numerous mitoses as well as necrosis. the paraspinal region or epidural space) exceptionally rare. Most tumours consist
Only isolated cases of the inflammatory (1595,2851) Parenchymal examples primarily of undifferentiated small cells,
variant of this entity have been reported are rare (623). An association with EBV whereas strap cells with cross striations
to involve brain {1598). and immunosuppression has been es- are only occasionally observed. lmmu-
tablished (2851}, but leiomyosarcomas nostaining for desmin and myogenin
ICD-0 code 8802/3 arising in these settings are less clearly usually confirms the diagnosis. Rhab-
malignant and may respond to immune domyosarcomas must be differentiated
reconstitution. Most leiomyosarcomas from other brain tumours that occasion-
Leiomyoma diffusely express desmin and SMA. ally show skeletal muscle differentiation,
such as medullomyoblastomas, gliosar-
Definition ICD-0 code 8890/3 comas, MPNSTs, germ cell tumours, and
A benign smooth muscle tumour that can even rare meningiomas {1120). Malignant
typically be readily recognized by its pat- ectomesenchymoma, a mixed tumour
tern of intersecting fascicles, being com- Rhabdomyoma composed of ganglion cells or neuro-
posed of eosinophilic spindle cells with blasts and one or more mesenchymal
blunt-ended nuclei {1506} Definition elements (usually rhabdomyosarcoma)
As a rule, leiomyomas lack mitotic activ- A benign lesion consisting of mature stri- may also occur in the brain (1919)
ity and cytological atypia Occasional ated muscle.
examples feature nuclear palisading and One reported case of rhabdomyoma as- ICD-0 code 8900/3
should not be mistaken for schwannoma. sociated with a cranial nerve also fea-
Diffuse leptomeningeal leiomyoma (1195) tured a minor adipose tissue component
and an angioleiomyomatous variant (2630). However, most reported intra- Chondroma
{1410} have been described. EBV- and neural examples are in fact neuromuscu-
AIDS-associated cases have also been lar choristoma (also called benign triton Definition
reported (1225). tumour). This is an important diagnostic A benign, well-circumscribed neoplasm
distinction given the high risk of postop- composed of low-cellularity hyaline
ICD-0 code 8890/0 erative fibromatosis associated with neu- cartilage.
romuscular choristoma (979). CNS rhab- Isolated cases of intracranial chon-
domyomas must also be distinguished droma (usually dural-based) have been
Leiomyosarcoma from skeletal muscle heterotopia, most of reported (500,1397). Outside the CNS,
which occur within prepontine leptome- chondromas often develop in the skull
Definition ninges (710). and only secondarily displace dura and
A malignant neoplasm with predominant- brain. Malignant transformation of a large
ly smooth muscle differentiation. ICD-0 code 8900/0
The morphological variants of leiomyo-
sarcoma include epithelioid leiomyosar-
coma, myxoid leiomyosarcoma, granular Rhabdomyosarcoma
cell leiomyosarcoma, and inflammatory
leiomyosarcoma (715} Definition
lntracranial leiomyosarcomas (555, A malignant neoplasm with predominant-
1537), like their soft tissue counterparts, ly skeletal muscle differentiation.
retain the intersecting fascicles at 90° Whether meningeal or parenchymal,
angles and eosinophilic cytoplasm, but nearly all primary CNS rhabdomyo-
show marked nuclear pleomorphism, sarcomas are of the embryonal type
increased mitotic activity, and necro- {2069,2513}; alveolar rhabdomyosar-
sis. Most intracranial leiomyosarcomas coma (1259} and a rhabdomyosarcoma- Fig. 11.26 Dural chondroma, a meningeal tumour
arise in or adjacent to the dura (e.g. in tous element in gliosarcoma (2465) are composed of mature hyaline cartilage.

262 Mesenchymal, non-rneninqothelral tumours


cNS chondroma to chondrosarcoma has
been documented over a long clinical
course (1689).

.co-o code 9220/0

Chondrosarcoma
Definition
A malignant mesenchymal tumour with Fig. 11.27 Chondrosarcoma. A MRI of a low-grade chondrosarcoma arising from the vertebral column. Note the
cartilaginous differentiation. lobulated contours and invasion into adjacent soft tissues. B Meningeal low-grade chondrosarcoma showing glistening
Most chondrosarcomas arise de novo, nodules of hyaline cartilage with focal softening and liquefaction.
but some develop in a pre-existing be-
nign cartilaginous lesion.

ICD-0 code 9220/3

Microscopy

Convenvonalchondrosarcorna
lntracranial, extraosseous chondrosar-
comas of the classic type are rare {407,
1352,1855,1919). The same is true for
extraskeletal myxoid chondrosarcoma,
which has been reported to arise within
the brain (416) as well as in the leptome-
ninges of the brain. Chondrosarcomas
arising in the skull base, in particular in
the midline, should be distinguished from
(chondroid) chordoma. Unlike chordo-
mas, chondrosarcomas are non-reactive
for keratin, EMA (1685), and brachyury
(2672).

IV!esenchyrnalchondrosarcorna
Mesenchymal chondrosarcoma also
arises more often in bones of the skull
or spine than within dura or brain paren-
chyma (2200,2262,2820). Some tumours
consist primarily of the small-cell com-
ponent, punctuated by scant islands of
atypical hyaline cartilage, whereas in oth-
ers the chondroid element predominates.
The histological pattern of the mesenchy-
mal component resembles either a Ewing
sarcoma-like small blue cell malignancy
or a solitary fibrous tumour I haemangio-
pericytoma, due to its distinctive stag-
horn vascular network. The transition be-
tween the chondroid and mesenchymal
component is typically abrupt; however,
a more gradual transition can also occur,
and this pattern is more challenging to
distinguish from small cell osteosarco-
mas. When the cartilage is not obvious,
SOX9 immunostaining may be useful,
because it is positive in the undifferenti-
ated mesenchymal component {668). Fig. 11.29 Dural osteoma. A Postcontrast T1-weighted MRI reveals a non-enhancing extra-axial mass. B Bone-
In difficult cases, the diagnosis can be window CT reveals an osseous mass involving the dura and inner table of the skull.

Chondrosarcoma 263
.,

Osteochondroma is characterized by the


presence of a cartilaginous cap and a fi-
brous perichondrium that extends to the
periosteum of the bone.

ICD-0 code 9210/0

Osteosarcoma
A
Fig. 11.30 Dural osteoma. A Bosselated dural-based osseous tumour. B On microscopy, gliotic brain parenchyma is
Definition
seen adjacent to the dense bone of the dural osteoma. A malignant bone-forming mesenchymal
tumour.
confirmed by the presence of NCOA2 from asymptomatic dural calcification, Osteosarcoma predominantly affects
gene rearrangements (2688l. ossification related to metabolic disease adolescents and young adults. The pre-
or trauma, and rare examples of astro- ferred sites are the skull and the spine
cytoma and gliosarcoma with osseous and more rarely the meninges and the
Osteoma differentiation. brain (144,352,2214,2227,2326l Bone
matrix or osteoid deposition by the pro-
Definition ICD-0 code 9180/0 liferating tumour cells is required for the
A benign bone-forming tumour with limit- diagnosis. Osteosarcomatous elements
ed growth potential. may exceptionally be encountered as
Isolated cases of intracranial osteoma Osteochondroma components of germ cell tumour and
(usually dural-based) have been reported gliosarcoma (132}
(427l. Outside the CNS, osteomas often Definition
develop in the skull and only secondarily A benign cartilaginous neoplasm, which ICD-0 code 9180/3
displace dura and brain. Histologically, may be pedunculated or sessile, arising
they correspond to similar tumours aris- on the surface of the bone.
ing in bone and must be distinguished

264 Mesenchymal, non-meningothelial tumours


CHAPTER12
Melanocytic tumours
Meningeal melanocytosis
Meningeal melanomatosis
Meningeal melanocytoma
Meningeal melanoma
Melanocytic tumours Brat D.J.
Perry A.
Wesseling P.
Bastian B.C.

Definition
Primary melanocytic neoplasms of the
CNS are diffuse or localized tumours that
presumably arise from leptomeningeal
melanocytes.
Benign lesions that are diffuse without
forming macroscopic masses are termed
melanocytosis, whereas malignant dif-
fuse or multifocal lesions are termed
melanomatosis. Benign or intermediate- A
grade tumoural lesions are termed mel- Fig. 12.01 A Meningeal melanocytosis involving the subarachnoid space of the cerebral hemispheres. B Meningeal
anocytomas. Malignant lesions that are melanomatosis infiltrating the meninges around the brain stem and cerebellum.
discrete tumours are termed melanomas.
lmmunophenotype Genetic susceptibility
Microscopy Most tumours react with the anti-melano- Little is known regarding the inherited
Diagnosis of this family of neoplasms somal antibodies HMB45, melan-A, and susceptibility of these neoplasms, if any.
hinges on the recognition of tumour cells microphthalmia-associated transcrip- Both the cutaneous naevi and the CNS I
that have melanocytic differentiation. tion factor (MITF). They also express melanocytic neoplasms that arise in the
Most melanocytic neoplasms display S100 protein. Staining for vimentin and setting of neurocutaneous melanosis
melanin pigment finely distributed within neuron-specific enolase are variable. are strongly associated with activating
the tumour cytoplasm and coarsely dis- Among the discrete melanocytic tumours somatic mutations of NRAS, most often
tributed within the tumour stroma and (melanocytoma and melanoma), expres- involving codon 61 (1921,2225). BRAF
the cytoplasm of tumoural macrophages sion of collagen IV and reticulin is lack- V600E mutations have also been identi-
(melanophages). Rare melanocytomas ing around individual tumour cells, but is fied in the cutaneous naevi of patients
and occasional primary melanomas do present around blood vessels and larger with neurocutaneous melanosis (albeit
not demonstrate melanin pigment; diag- tumoural nests (1406). There is only rare less frequently), but mutations have not
nosis then relies more heavily on immu- expression of GFAP, NFPs, cytokeratins, yet been described in CNS melanocytic
nohistochemistry and mutation profile. or EMA. The Ki-67 proliferation index is neoplasms in this setting. Although the
Identification of melanocytic lesions usu- typically < 1-2% in melanocytomas and NRAS mutations are not inherited, they
ally requires histopathological examina- averages about 8% in primary melano- are thought to occur early in embryogen-
tion, but the diagnosis of melanocytosis mas (266}. esis, prior to the migration of melanocyte
and melanomatosis has occasionally precursors to their locations in the skin
been made by cerebrospinal fluid cytol- Cell of origin and leptomeninges (1405). In patients
ogy (2106}. Melanocytic neoplasms of the nervous with neurocutaneous melanosis, the mul-
system and its coverings are thought to tiple congenital melanocytic naevi and
Differential diagnosis arise from leptomeningeal melanocytes associated leptomeningeal melanocytic
Primary CNS melanocytic neoplasms that are derived from the neural crest lesions contain the same NRAS muta-
must be distinguished from other mel- (1405). These melanocytes may differ tion, whereas the normal skin and blood
anotic tumours that involve the CNS, in- developmentally from melanocytes in cells are not mutated, suggesting that a
cluding metastatic melanoma and other epithelia (e.g. the epidermis and mucosal single postzygotic mutation of NRAS is
primary tumours undergoing melaniza- membrane) and seem to be more closely responsible for the multiple cutaneous
tion, as can occur in melanotic schwan- related to melanocytes in the uveal tract and leptomeningeal melanocytic lesions
noma, medulloblastoma, paraganglioma, (139). In the normal CNS, melanocytes in this phacomatosis (1286f.
and various gliomas (266,14051. There is are preferentially localized at the base
little evidence supporting the existence of the brain, around the ventral medulla
of a true melanotic meningioma, although oblongata, and along the upper cervical
rare melanocytic colonization of meningi- spinal cord. Melanocytic neoplasms as-
omas has been documented (1768). Mel- sociated with neurocutaneous melanosis
anotic neuroectodermal tumour of infan- likely derive from melanocyte precursor
cy (retinal anlage tumour) has also been cells that reach the CNS after acquiring
reported at intracranial locations (1219). somatic mutations, mostly of NRAS.

266 Melanocytic tumours


B Highly pleomorphic melanin-laden cells

Meningeal melanocytosis and decade, with an equal sex distribution radiological evidence of CNS involve-
meningeal melanomatosis and no racial predisposition {1201). ment has been reported in as many as
23% of asymptomatic children with giant
Localization congenital naevi {726). As part of a com-
Meningealme/anocytosis Melanocytosis and melanomatosis in- pletely distinct clinical entity, melanocy-
volve the supratentorial leptomeninges, tosis may also be associated with con-
Definition the infratentorial leptomeninges, and may genital naevus of Ota {117).
Meningeal melanocytosis is a diffuse or involve the superficial brain parenchyma
multifocal proliferation of cytologically by extending into perivascular Virchow- Imaging
bland melanocytic cells that arises from Robin spaces. They generally involve CT and MRI of melanocytosis and mela-
the leptomeninges and involves the sub- large expanses of the subarachnoid nomatosis show diffuse thickening and
arachnoid space. space with focal or multifocal nodularity enhancement of the leptomeninges,
Meningeal melanocytosis cells can occasionally seen. The sites of highest often with focal or multifocal nodularity
spread into the perivascular spaces with- frequency include the cerebellum, pons, {1981). Depending on melanin content,
out frank invasion of the brain {1533}. medulla, and temporal lobes. diffuse and circumscribed melanocytic
tumours of the CNS may have a char-
ICD-0 code 8728/0 Clinical features acteristic appearance on MRI due to
Neurological symptoms associated with the paramagnetic properties of melanin,
Meningeal melenometosis melanocytosis or melanomatosis arise resulting in an isodense or hyperintense
secondarily to either hydrocephalus or signal on T1-weighted images and a hy-
Definition local effects on the CNS parenchyma. pointense signal on T2-weighted images
A primary CNS melanoma that arises Neuropsychiatric symptoms, bowel and {2372).
from leptomeningeal melanocytes and bladder dysfunction, and sensory and
displays a diffuse pattern of spread motor disturbances are common. Once Macroscopy
throughout the subarachnoid space and malignant transformation occurs, symp- Diffuse melanocytic neoplasms present
Virchow-Robin spaces, often with CNS toms progress rapidly, with increasing as dense black replacement of the suba-
invasion. intracranial pressure resulting in irritabil- rachnoid space or as dusky clouding of
ity, vomiting, lethargy, and seizures. the meninges.
ICD-0 code 8728/3 Neurocutaneous ·melanosis is a combi-
nation of melanocytosis or melanoma- Microscopy
Epidemiology tosis with giant or numerous congenital The pathological proliferation of lepto-
Diffuse meningeal melanocytic neo- melanocytic naevi of the skin, usually in- meningeal melanocytes and their pro-
plasms are rare, and population-based volving the trunk or head and neck, and duction of melanin account for the main
incidence is not available {1201}. Menin- with various other malformative lesions, microscopic findings in these diseases
geal melanocytosis and melanomatosis such as Dandy-Walker syndrome, sy- {583). Tumour cells diffusely involving
are strongly linked with neurocutaneous ringomyelia, and lipomas {563,564, the leptomeninges assume a variety of
melanosis, a rare phacomatosis that is 1574) Approximately 25% of patients shapes, including spindled, round, oval,
typically associated with giant congenital with meningeal melanocytosis have sig- and cuboidal. In melanocytosis, indivi-
naevi and presents before the age of 2 nificant concomitant cutaneous lesions. dual cells are cytologically bland and ac-
years. In one series of 39 such cases, the Conversely, about 10-15% of patients cumulate within the subarachnoid space
age at presentation for CNS manifesta- with large congenital melanocytic naevi and Virchow-Robin spaces without dem-
tions ranged from stillbirth to the second of the skin develop clinical symptoms re- onstrating overt CNS invasion {2372).
lated to CNS melanocytosis {564), and Unequivocal CNS parenchymal invasion

Meningeal melanocytosis and meningeal melanomatosis 267


Fig. 12.03 MRI features of meningeal melanocytoma. A T1-weighted axial images (pre-contrast) reveal a hyperintense, well-circumscribed mass in the midline of the cerebellum
arising from the dura. B On T2-weighted images, the mass is hypointense. C Following the administration of contrast agent, the melanocytoma shows homogeneous enhancement.

should not be seen in melanocytosis; if


it is identified, the tumour must be con-
sidered melanomatosis. Similarly, the
presence of mitotic activity, severe cyto-
logical atypia, or necrosis also warrants a
diagnosis of melanomatosis.

Prognosis and predictive factors


Diffuse melanocytic neoplasms of the
leptomeninges generally carry a poor
prognosis even in the absence of histo-
logical malignancy {2106}.

Meningeal me/anocytoma
Definition
A well-differentiated, solid, and non-intll-
trative melanocytic neoplasm that arises
from leptomeningea/ melanocytes.
Meningeal melanocytoma is charac-
terized by the presence of epithelioid,
fusiform, polyhedral, or spindled mel-
anocytes, without evidence of anaplasia,
necrosis, or elevated mitotic activity.

ICD-0 code 8728/1

Epidemiology
Melanocytomas account for 0.06-0.1%
of brain tumours, and the annual inci-
dence has been estimated at 1 case per
Fig. 12.04 Meningeal melanocytoma. A Loose or tight nests of low-grade, pigmented spindle cells with intervening
10 million population (1149) Melanocy-
stroma containing higher levels of melanin pigment. Note the vague, loosely formed whorls and fascicles.
B Accumulation of large, melanin-containing macrophages (melanophages) is seen between tumour cells with a more tomas can occur in patients of any age
spindled phenotype. The nuclei of tumour cells are bean-shaped and have micronucleoli. Melanin within the cytoplasm (range: 9-73 years), but are most Ire·
of melanophages is typical in larger aggregates. C Melanin-containing macrophages (melanophages). quent in the fifth decade of life (mean

268 Melanocytic tumours


age 45-50 years). There is a slight fe- features, such as those of melanocy-
male predominance, with a female-to- toma, but showing CNS invasion or el-
male ratio of 1.5:1 {266,2687). evated mitotic activity, should be classi-
fied as intermediate-grade melanocytic
Localization neoplasms {266)
Most melanocytomas arise in the ex-
tramedullary, intradural compartment at Electron microscopy
the cervical and thoracic spine. They can The cells of melanocytoma lack junctions
be dural-based or associated with nerve and contain melanosomes at various
roots or spinal foramina {266,849). Less stages of development. Unlike in schwan-
frequently, they arise from the leptome- nomas, a well-formed pericellular basal Fig. 12.05 Spinal meningeal melanoma with diffuse
ninges in the posterior fossa or supraten- lamina is lacking, but groups of melano- infiltration of the leptomeninges and focal infiltration of
torial compartments. The trigeminal cave cytoma cells may be ensheathed {38} the medulla.
is a site with a peculiar predilection for Unlike in meningioma. no desmosomes
primary melanocytic neoplasms, and tu- or interdigitating cytoplasmic processes
mours at this site are associated with an are present. showing a slight predilection for the spi-
ipsilateral naevus of Ota {117,1970} nal cord and posterior Iossa {760}.
Genetic profile
Clinical features Hotspot mutations of GNAQ or GNA 11, Clinical features
Melanocytomas present with symptoms most often involving codon 209, are fre- Like melanocytomas, melanomas pres-
related to compression of the spinal cord, quent in melanocytoma, similar to those ent with focal neurological signs depend-
cerebellum, or cerebrum by an extra- present in uveal melanoma and blue nae- ing on location {266).
axial mass, with focal neurological signs vus {1313,1404,1405,1724). Cytogenetic
depending on location {266}. losses involving chromosome 3 and the Imaging
long arm of chromosome 6 have been re- CNS structures adjacent to a melanoma
Imaging ported for melanocytoma as well. are often T2-hyperintense, indicating
Melanocytomas and melanomas gener- vasogenic oedema generated in re-
ally show homogeneous enhancement Prognosis and predictive factors sponse to rapid tumour growth.
on postcontrast images. Tumours with Melanocytoma lacks anaplastic features,
abundant melanin will show a charac- but a few undergo local recurrences; Macroscopy
teristic pattern of T1 hyperintensity (pre- intermediate-grade melanocytic tumours Malignant melanomas are typically
contrast) and T2 hypointensity. typically invade the CNS, although too solitary, with extra-axial location. Pig-
few have been reported to reliably pre- mentation may vary from black to red-
Macroscopy dict the biology of these tumours {266). dish brown, blue, or macroscopically
Melanocytoma are solitary mass lesions, Rare examples of malignant transforma- non-pigmented.
generally extra-axial, that may be black, tion of a melanocytoma have been re-
reddish brown, blue, or macroscopically ported {2182l Microscopy
non-pigmented. Primary meningeal melanoma is histo-
logically similar to melanomas arising
Microscopy AAeningealrnelanorna in other sites. Anaplastic spindled or
Melanocytomas are solitary low-grade epithelioid cells arranged in loose nests,
tumours with no invasion of surrounding Definition fascicles, or sheets display variable cyto-
structures {266,1405,2687). Slightly spin- A primary malignant neoplasm of the plasmic melanin {266,760). Some mela-
dled or oval tumour cells containing vari- CNS that arises from leptomeningeal nomas contain large cells with bizarre
able melanin often form tight nests with a melanocytes, presents as a solitary mass nuclei, numerous typical and atypical
superficial resemblance to the whorls of lesion, and displays aggressive growth mitotic figures, significant pleomorphism,
meningioma. Heavily pigmented tumour properties. and large nucleoli; others are densely
cells and tumoural macrophages are es- cellular and less pleomorphic, usually
pecially seen at the periphery of nests. ICD-0 code 8720/3 consisting of tightly packed spindle cells
Other melanocytoma variants demon- with a high nuclear-to-cytoplasmic ratio.
strate storiform, vasocentric, and sheet- Epidemiology Melanomas are more pleomorphic, aria-
like arrangements. Only rare amelanotic Primary meningeal melanomas have plastic, and mitotically active, and have
melanocytomas have been described. been reported in patients aged 15- a higher cell density, than melanocytoma
The nuclei are oval or bean-shaped, oc- 71 years (mean: 43 years). An annual in- and often demonstrate unequivocal tis-
casionally showing grooves, with small cidence of 0.5 cases per 10 million popu- sue invasion or coagulative necrosis.
eosinophilic nucleoli. Cytological atypia lation has been reported{988l. Meningeal melanomatosis may arise
and mitoses are generally absent (on from diffuse spreading of a primary ma-
average, < 1 mitosis per 10 high-power Localization lignant meningeal melanoma through the
fields). It has been suggested that mel- Meningeal melanomas are dural-based subarachnoid space.
anocytic tumours with bland cytological and occur throughout the neuraxis,

Meningeal melanoma 269


tumours. Mutations in genes typically
seen in cutaneous or acral melanomas
such the TERT promoter, NRAS, BRAF
and KIT, are rare in primary CNS melano�
cytic neoplasms of adults, and when en-
countered raise suspicion of a metasta-
sis. However, the uncertainty involved in
establishing the primary versus metastat-
ic nature of a melanoma may confound
interpretation of molecular studies.
Genomic alterations may be helpful in
distinguishing between different types of
melanotic tumours of the CNS; in particu-
lar, DNA methylation patterns have been
shown to identify melanotic neoplasms
in a manner concordant with mutation
spectrum and histological classes (498,
13'13,'1406).

Prognosis and predictive factors


Fig. 12.06 Thoracic meningeal melanoma. The epithelioid and spindled tumour cells have ample, eosinophilic Malignant melanoma is a highly aggres-
cytoplasm and a large vesicular nucleus with prominent nucleolus, and show marked mitotic activity (arrows). Dispersed sive and radioresistant tumour with poor
melanophages are present between the tumour cells; in this tumour, a GNA 11 mutation was identified. prognosis, and it can metastasize to re-
mote organs. Nevertheless, the progno-
Genetic profile Primary CNS melanomas also harbour sis of the primary meningeal melanoma
In the case of childhood CNS melanomas GNAQ or GNA11 mutations, albeit at a seems to be better than that of meta-
and neurocutaneous melanosis, there is lower frequency than do melanocyto- static examples, particularly if localized
a strong link to mutations in NRAS, es- mas. These tumours appear to progress and complete resection is possible (739).
pecially codon 61 {192'1}. Less frequently, to melanoma analogous to uveal mela- Considering the close relationship to
BRAF V600E mutations have been de- noma, in which GNAQ or GNA11 muta- uveal melanoma, BAPi loss is likely an
scribed in the congenital melanocytic tions arise early, and are followed by in- adverse prognostic markers in those pri-
naevi of patients with neurocutaneous activation of BAP1 or mutation of SF381 mary CNS melanomas that are not asso-
melanosis (2225). or EIF1AX as they evolve into malignant ciated with congenital naevi.

270 Melanocytic tumours


CHAPTER13
Lymphomas
Diffuse large 8-cell lymphoma of the CNS
Immunodeficiency-associated CNS lymphomas
lntravascular large 8-cell lymphoma
Low-grade 8-cell lymphomas
T-cell and NK{T-cell lymphomas
Anaplastic large cell lymphoma
MALT lymphoma of the dura
Lymphomas Deckert M.
Paulus W.
Kluin P.M.
Ferry JA

Diffuse large 8-cell lymphoma polyomaviruses SV40 and BK virus (1702, fluid (CSF) analysis is limited (1340)
of the CNS 1725)) do not play a role. Meningeal dissemination is diagnosed
in 15.7% of cases (12.2% by CSF cyto-
Definition Localization morphology, 10.5% by PCR, and 4.1%
A diffuse large B-ce/1 /ymphoma (OLBCL) About 60% of all PCNSLs involve the su- by MRI) (1341). Pleocytosis is found in
confined to the CNS at presentation. pratentorial space, including the frontal 35-60% of PCNSL cases and correlates
Excluded from this entity are lymphomas lobe (in 15% of cases), temporal lobe (in with meningeal dissemination {1341). Cell
arising in the dura, intravascular large B- 8%), parietal lobe (in 7%), and occipital counts may even be normal. The CSF
eel! lymphoma, and lymphomas of T-cell lobe (in 3%), basal ganglia and periven- harbours neoplastic cells in a minority
or NK-cell lineage (which may also pres- tricular brain parenchyma (in 10%), and of patients with leptomeningeal involve-
ent in the CNS), as well as lymphomas corpus callosum (in 5%). The posterior ment, and their detection may require
with systemic involvement or with sec- fossa and spinal cord are less frequently repeated lumbar puncture. The combi-
ondary involvement of the CNS. Immuno- affected (in 13% and 1% of cases, re- nation of cytological and immunohisto-
deficiency-associated primary CNS lym- spectively) {562). A single tumour is en- chemical analysis with multiparameter
phomas are discussed separately. countered in 60-70% of patients, with flow cytometry may enhance detection
the remainder presenting with multiple of CSF lymphoma cells (2299). PCR
ICD-0 code 9680/3 tumours (562). The leptomeninges may analysis of the CDR3 region of the IGH
be involved, but exclusive meningeal gene region followed by sequencing of
Epidemiology manifestation is unusual. Ocular manifes- the PCR products may identify a clonal
Primary CNS lymphomas (PCNSLs) ac- tation (i.e. in the vitreous, retina, or op- B-cell population in the CSF, but does
count for 2.4-3% of all brain tumours tic nerve) occurs in 20% of patients and not enable lymphoma classification. El-
and 4-6% of all extranodal lympho- may antedate intracranial disease (1123). evated CSF levels of miR-21, miR-19,
mas {2286). The overall annual inci- Extraneural dissemination is very rare. In and miR-92a have been reported to dif-
dence rate of PCNSL is 0.47 cases per cases with systemic spread, PCNSL has ferentiate PCNSL from inflammatory and
100 000 population {2652). In the past a propensity to home to the testis, an- other CNS disorders {124); however, the
two decades, an increased incidence other immunoprivileged organ (237,1124). diagnostic usefulness of this parame-
has been reported in patients aged > 60 ter and its potential as a marker during
years (2652). PCNSL can affect patients Clinical features follow-up (125) are yet to be definitively
of any age, with a peak incidence dur- Patients present with cognitive dysfunc- established.
ing the fifth to seventh decade of life. The tion, psychomotor slowing, and focal
median patient age is 56 years, and the neurological symptoms more frequently Macroscopy
male-to-female ratio is 3:2. than with headache, seizures, and cra- As observed on postmortem examina-
nial nerve palsies. Blurred vision and eye tion, PCNSLs occur as single or multiple
Etiology floaters are symptoms of ocular involve- masses in the brain parenchyma, most
In immunocompetent individuals, etio- ment {140,1340,1926). frequently in the cerebral hemispheres.
logical factors are unknown. Viruses (e.g. Often, they are deep-seated and ad-
EBV, HHV6 (1915), HHV8 {1704), and the Imaging jacent to the ventricular system. The
MRI is the -rnost sensitive technique to tumours can be firm, friable, granular,
detect PCNSL, which is hypointense on haemorrhagic, and greyish tan or yellow,
T1-weighted images, isointense to hy- with central necrosis. Or they can be vir-
Males
90-100
perintense on T2-weighted images, and tually indistinguishable from the adjacent
BO<l9
densely enhancing on postcontrast im- neuropil. Demarcation from surrounding
70-79
60-<;9
ages. Peritumoural oedema is relatively parenchyma is variable. Some tumours
50-59 limited and is less severe than in malig- appear well delineated, like metastases.
4().,49 nant gliomas and metastases (1340). When diffuse borders and architectural
30-39 Meningeal involvement may present as effacement are present, the lesions re-
20-29
hyperintense enhancement {1403). With semble gliomas. Like malignant gliomas,
10-19
steroid therapy, lesions may vanish within these tumours may diffusely infiltrate
0-9

40 30 20 10 10 20 30 40
hours (562). large areas of the hemispheres without
% of cases % of cases forming any distinct mass, a manifesta-
Fig. 13.01 Age and sex distribution of primary CNS Spread tion which has been referred to as 'lym-
lymphomas in immunocompetent patients. The diagnostic value of cerebrospinal phomatosis cerebri'. However, this term

272 Lymphomas
pattern is frequent. Infiltration of cerebral 2128l. BCL2 expression is common; 82%
blood vessels causes fragmentation of PCNSLs have a BCL2-high, MYC-high
of the argyrophilic fibre network. From phenotype {296l. Mitotic activity is brisk;
these perivascular cuffs, tumour cells in- accordingly, the Ki-67 proliferation index
vade the neural parenchyma, either with usually exceeds 70% or even 90% {296l.
a well-delineated invasion front with small Apoptotic cells may be frequent. With the
clusters or with single tumour cells dif- exception of isolated cases, there is no
fusely infiltrating the tissue, which shows evidence of EBV infection {1705l, and the
a prominent astrocytic and microglial presence of EBV should prompt evalua-
activation and harbours reactive inflam- tion for underlying immunodeficiency.
Fig. 13.02 Primary CNS lymphoma. Postcontrast
matory infiltrates consisting of mature
T1-weighted MRI showing multifocal disease with T and B cells. Morphologically, PCNSLs Genetic profile
homogeneous contrast enhancement and relatively little consist of large atypical cells with large PCNSLs are mature B-cell lymphomas.
surrounding oedema. Periventricular disease is evident round, oval, irregular, or pleomorphic nu- The tumour cells correspond to late ger-
in both the frontoparietal region and the brain stem. clei and distinct nucleoli, corresponding minal centre exit B cells with blocked
to centroblasts or immunoblasts. Some terminal B-cell differentiation. Thus, they
does not define a distinct disease entity, cases show a relatively monomorphic carry rearranged and somatically mutat-
so it should not replace the specific diag- cell population, with intermingled mac- ed immunoglobulin (IG) genes with evi-
nosis (i.e. DLBCL of the CNS). Meningeal rophages mimicking the appearance of dence of ongoing somatic hypermutation
involvement may resemble meningitis or Burkitt lymphoma. {1705,1925,2548l. Consistent with the on-
meningioma, or can even be inconspicu- going germinal centre programme, they
ous macroscopically. lmmunophenotype show persistent BCL6 activity {296). The
The tumour cells are mature B cells with process of somatic hypermutation is not
Microscopy a PAX5-positive, CD19-positive, CD20- confined to its physiological targets (IG
positive, CD22-positive, CD79a-positive and BCL6 genes}, but extends to other
Stereotactic biopsy phenotype. lgM and lgD, but not lgG, genes that have been implicated in tum-
Stereotactic biopsy is the gold standard are expressed on the surface {1707l, origenesis, including BCL2, MYC, PIM1,
for establishing the diagnosis and classi- with either kappa or lambda light chain PAX5, RHOH, KLHL14, OSBPL10, and
fication of CNS lymphoma. It is important restriction. Most express BCL6 (60-80%) SUS02{297,1709,2639l. These data indi-
to withhold corticosteroids before biopsy, and MUM1/IRF4 (90%), whereas plasma cate that aberrant somatic hypermutation
because they induce rapid tumour wan- cell markers (e.g. CD38 and CD138) are has a major impact on the pathogenesis
ing. Corticosteroids have been shown to usually negative. Less than 10% of all of PCNSL. The fixed lgM/lgD phenotype
prevent diagnosis in as many as 50% of PCNSLs express CD10 {561l CD10 ex- of the tumour cells is in part due to mis-
cases {298l. pression is more frequent in systemic carried IG class switch rearrangements
DLBCL; therefore, CD10 positivity in a during which the Smu region is deleted
Histopathology CNS lymphoma with DLBCL characteris- {1707). PRDM1 mutations also contribute
PCNSLs are highly cellular, diffusely tics should prompt a thorough investiga- to impaired IG class switch recombina-
growing, patternless tumours. Centrally, tion for systemic DLBCL that might have tion {502l.
large areas of geographical necrosis metastasized to the CNS. HLA-A/B/C Translocations affect the IG genes (in
are common, and may harbour viable and HLA-DR are variably expressed, with 38% of cases) and BCL6 (in 17-47%)
perivascular lymphoma islands. At the approximately 50% of PCNSLs having recurrently, whereas MYC translocations
periphery, an angiocentric infiltration lost HLA class I and/or II expression {237, are rare and translocations of the BCL2

Diffuse large B-cell lymphoma of the CNS 273


DNA hypermethylation of OAPK1 (in 84%
of cases), CDKN2A (in 75%), MGMT (in
52%), and RFC (in 30%) may be of po-
tential therapeutic relevance {457,475,
692,2309).

Genetic susceptibility
In immunocompetent individuals, genetic
predispositions to PCNSL have not been
described. About 8% of patients with
PCNSL have had a prior extracranial tu-
mour {2100), most of which arose in the
haematopoietic system. In patients with
PCNSL with preceding extraneural lym-
phoma, comparative molecular analyses
of primary and secondary lymphomas
may confirm or exclude a common clonal
origin of these tumours, distinguishing
CNS relapse from an unrelated second-
ary cerebral lymphoma. However, be-
•- ...
Fig. 13.05 Primary CNS lymphoma. A Diffuse large 8-cell lymphoma. B Tumour cells express the pan-8-cell marker
cause these analyses are not routinely
performed, information about a possi-
CD20. C Expression of the BCL6 protein by the tumour cells. D Strong nuclear expression of the MUM1 protein by the ble association is usually lacking. In in-
majority of tumour cells of a primary CNS lymphoma. dividual patients, associations between
PCNSL and other tumours (e.g. car-
gene are absent {296,341,1710). FISH and of PCNSLs have lost expression of HLA cinoma, meningioma, and glioma) or
genome-wide SNP analyses have shown class I and II gene products, respectively hereditary tumour syndromes (e.g. neu-
recurrent gains of genetic material, most {237). rofibromatosis type 1) are likely to be
frequently affecting 18q21.33-q23 (in Several important pathways (i e. the B- coincidental.
43% of cases), including the BCL2 and eel! receptor, the toll-like receptor, and Folate and methionine metabolism has
MALT1 genes; chromosome 12 (in 26%); the NF-kappaB pathway) are frequently been proposed to be relevant to PCNSL
and 10q23.21 (in 21%) {2309). Losses of activated due to genetic alterations af- susceptibility. The G allele of the meth-
genetic material most frequently involve fecting the genes CD79B (in 20% of yltetrahydrofolate homocysteine S-meth-
6q21 (in 52% of cases), 6p21 (in 37%), cases), INPP50 (in 25%), CBL (in 4%), yltransferase c.2756A-+G (D919G) mis-
8q12.1-q12.2 (in 32%), and 10q23.21 BLNK (in 4%), CARD11 (in 16%), MALT1 sense polymorphism was found to be
{2309). Heterozygous or homozygous (in 43%), BCL2 (in 43%), and MYDBB (in underrepresented among patients with
Joss or partial uniparental disomies of > 50%), which may foster proliferation PCNSL, suggesting a protective function
chromosomal region 6p21.32 affect 73% and prevent apoptosis {865,1357,1703, of this allele {1512).
of PCNSLs; this region harbours the HLA 1706,1708,2309)
class II-encoding genes HLA-ORB, Epigenetic changes may also contribute Prognosis and predictive factors
HLA-DOA, and HLA-008 {1181,2127, to PCNSL pathogenesis, including epi- PCNSL has a considerably worse out-
2309). Correspondingly, 55% and 46% genetic silencing by DNA methylation. come than does systemic DLBCL. Older

arr[hg19] 18q 12-3q23(42,514,448-78,007, 784}x4


11 21 11 2 • 12 2 21 I 2 32 21.33 22 3 23

MALTl BCL2
, .e ..------------,------------------------�-------------,
�.5

�.o
,.,

·1.0

·:?.O

.c.s

.... ..... ..... "'"' .....


,,..,
-).0+---�--�--�-'---�--�--�--�--�----r---�----r-----r-----r-----r-----r---'
..... . .. ..... '°"' ..... .....
Fig. 13.06 Primary CNS lymphoma. Microarray-based comparative genomic hybridization demonstrating copy number and allelic profile of a sample with a gain in 18q. The log2
ratio is displayed from the p arm to the q arm of the chromosome, showing a gain of two copies of the same allele in 18q12.3qter (shaded region: log2 ratio = 1; four allele states).

274 Tumours of the haematopoietic system


�C9�
patient age (> 65 years) is a major nega-
tive prognostic factor and is associated
Pathogenesis of PCNSL
with reduced survival and an increased
DNA methylation gain of genetic material
risk of neurotoxicity {4,1340).
High-dose methotrexate-based poly- 18 21
chemotherapy is currently the treatment ��=:2A} .... Loss�f / '" q .... ac�i�;�on
MGMT expression loss of genetic material
of choice {1340). The inclusion of whole-
brain irradiation may improve outcome, � SµDeletion .... NoCSR
but bears the risk of neurotoxicity result- SHM /ASHM
CDKN2A .... proliferation
ing in severe cognitive, motor, and au-
tonomic dysfunction, particularly in el- 6p21 (HLA) .... 'Immune
derly patients {4). The missense variant escape
Tc2c.776C--+G mutation of transcobala- point mutation
min C is associated with shorter survival (not SHM/ASHM)

and neurotoxicity {1511). Most protocols SHIP


BCR signaling
report a median progression-free sur- romoter substitution CD79B
... NF-KB
CBL
vival of about 12 months and an overall activation

....
BLNK
survival of approximately 3 years. In a NF-KB
CARD11

....
subgroup of elderly patients with PCNSL GAPD, BCL6 activation
with methylated MGMT, temozolomide HSP90a
MyDBB
NF-KB
monotherapy appeared to be therapeuti- IGH, IGL, ... activation
cally effective {1400). PRDM1 ...Block of B cell
The presence of reactive perivascu- differentiation
lar CD3 T-cell infiltrates on biopsy has Fig. 13.07 Pathogenesis of primary CNS lymphoma (PCNSL). Alterations of specific pathways contribute to the
been associated with improved survival lymphomagenesis of PCNSL. ASHM, aberrant somatic hypermutation; BCR, B-cell receptor; CSR, class-switch
{2004). LM02 protein expression by the recombination; SHM, somatic hypermutation.
tumour cells has been associated with
prolonged overall survival {1528). BCL6
expression has been suggested as a
prognostic marker in several studies,
although conflicting conclusions as to
whether it is a favourable or unfavourable
marker have been reported {1700,2037,
2071,2393). In one study, del(6)(q22)
was associated with inferior overall sur-
vival {341).

Comcoia-ma/aetea lymphoma A
Because the tumour cells are highly sus- Fig. 13.08 A Large, necrotizing diffuse large 8-cell lymphoma of the right hemisphere, extending via the corpus
callosum into the white matter of the left hemisphere. The patient was an HIV-1-infected infant. B Primary malignant
ceptible to steroid-induced apoptosis,
CNS lymphomas of the basal ganglia. Note the additional foci in the left insular region (arrowheads).
PCNSL may vanish rapidly on MRI and
within biopsies following corticosteroid
administration. Microscopically, neoplas- Immunodeficiency-associated responsible for the significant increase
tic B cells may be present in only small CNS lymphomas in the incidence of CNS lymphoma in the
numbers or may even be absent. Sam- 1980s. Generally, immunosuppression-
ples may show non-specific inflammato- Inherited or acquired immunodeficiency associated CNS lymphoma is EBV-re-
ry and reactive changes and/or necrosis; predisposes patients to CNS lymphoma. lated. Thus, the lymphoma cells express
foamy macrophages are frequent {298, Immunodeficiency syndromes include EBNA1-6, LMP1, EBER1, and EBER2
561). In some cases, PCR analysis of the ataxia-telangiectasia, Wiskott-Aldrich {1310).
CDR3 region of the IGH gene may reveal syndrome, and lgA deficiency. Autoim-
a monoclonal B-cell population. Howev- mune disorders (e.g. systemic lupus AIDS-related diffuse large 8-cell
er, pseudoclonality due to very low num- erythematosus and Sjogren syndrome), lymphoma
bers of B cells poses a problem. iatrogenic immunosuppression (either Within this category, AIDS-related diffuse
for the purpose of organ transplantation large B-cell lymphoma (DLBCL) of the
Sentinel lesions or due to treatment with drugs such as CNS, EBV-positive DLBC, NOS , lympho-
In rare cases, PCNSL has been reported methotrexate, azathioprine, or mycophe- matoid granulomatosis, and monomor-
to be preceded (by as long as 2 years) nolate for a wide variety of other diseas- phic or polymorphic post-transplant lym-
by demyelinating and inflammatory le- es), and immune system senescence are phoproliferative disorders may primarily
sions similar to multiple sclerosis {45, associated with an increased risk of CNS manifest within the CNS.
1071,1386). lymphoma. HIV infection was largely

Immunodeficiency-associated CNS lymphomas 275


.,

AIDS-related DLBCL of the CNS shares mimicking those of cerebral infarction or usually do well {100,1503,1886,2005
the morphological features of primary subacute encephalopathy. 2270,2368,2593) '
CNS lymphoma in immunocompetent Macroscopy reveals infarcts (acute and/ Microscopic examination reveals a
patients, with the exception of a more fre- or old), necrosis, and/or haemorrhage, dense, diffuse, or perivascular infiltrate
quent multifocal presentation, EBV asso- although abnormalities may be incon- predominantly of small lymphocytes'
ciation, and a tendency to contain more spicuous. Microscopically, large atypical with variable numbers of plasma cell�
and larger areas of necrosis. These ar- B cells can be seen occluding cerebral admixed. lmmunophenotyping shows a
eas of necrosis may simulate necrotizing blood vessels. Lack of CD29 and ICAM1 predominance of CD20-positive B cells
cerebral toxoplasmosis, which may occur (CD54) expression is thought to underlie (usually CDS-negative and CD10-
concomitantly {2425). HIV-associated the tumour cells' inability to migrate trans- negative), sometimes with a component
primary CNS lymphoma has become rar- vascularly {2003). of monotypic plasma cells, with a low pro-
er with the introduction of HAART therapy liferation index. These lymphomas, when
{2652). ICD-0 code 9712/3 subclassified, have been designated as
small lymphocytic lymphoma (positive
EBV+ diffuse large 8-csll lymphoma, for CD5 and CD23) {100). lymphoplas-
NOS Miscellaneous rare lymphomas macytic lymphoma {1125,1503,2368).
EBV-positive DLBCL, NOS, may affect in the CNS extranodal MALT lymphoma {1125,1503,
the CNS in elderly patients with no known 2368). and follicular lymphoma {1125)
immunodeficiency. Lymphomagenesis is Other than diffuse large B-cell lymphoma
attributed to the immunosenescence that of the CNS (i.e. primary CNS lymphoma), r-oeu and NK/T-cs/1 lymphomas
can develop with advancing age {1131). lymphomas manifesting primarily in the Primary T-cell lymphoma of the CNS is
CNS are rare {1503}. They include low- very rare, accounting for approximately
Lymphomatoid granulomatosis grade B-cell and T-cell lymphomas, very 2% of all primary lymphomas in the CNS
This affects the brain in 26% of cases rare Burkitt lymphoma {1503). and high- {693,2341). These lymphomas appear to
{1911 ). The lesions are characterized grade T-cell and NK/T-cell lymphomas be more common in Asia than elsewhere
by angiocentric and angiodestructive {893,1503,1529,1817,2017). Low-grade {450) and mainly affect young to middle-
lymphoid infiltrates. The proportion of lymphomas account for approximately aged adults {1503). Reported cases are
EBV-expressing CD20-positive, CD30- 3% of all CNS lymphomas; the majority often not subclassified except that they
positive or -negative, CD15-negative are of B-cell lineage {1125,1503). Con- are generally reported to be peripheral
large neoplastic B cells contributing to vincing cases of Hodgkin lymphoma T-cell lymphomas. They occur as single
these infiltrates is variable and may be primary to the CNS are vanishingly rare or multiple tumours in the cerebral hemi-
small. They are admixed with CD4 and {799) The differential diagnosis includes spheres (in 64% of cases), basal ganglia
CDS T lymphocytes. The infiltrates in- secondary CNS involvement by a sys- (11%), corpus callosum (13%), brain stem
vade blood vessel walls and may induce temic lymphoma and a chronic inflamma- (9%), cerebellum (7%), meninges (2%),
infarct-like necrosis of tumour and/or tory process, and likely also EBV-positive and spinal cord (4%) Microscopically,
brain tissue. DLBCL, NOS. Meticulous staging to ex- malignant T cells express CD45 and T-
The CNS may be the only part of the body clude a systemic lymphoma is essential. cell antigens (CD2, CD3, CD4 or CDS,
affected by post-transplant lymphoprolif- For low-grade lymphomas, careful path- CD5, and CD7), although loss of T-cell
erative disorder, although CNS involve- ological evaluation to confirm a neoplas- antigens may occur. Some cases are
ment is not frequent. For more details on tic process is critical. positive for the cytotoxic granule proteins
morphology and grading, see the WHO granzyme B and perforin. Molecular ge-
classification of tumours of haematopoi- Low-grads 8-csll lymphomas netic demonstration of T-cell monoclonal-
etic and lymphoid tissues {2473). Low-grade B-cell lymphomas of the CNS ity can be helpful in distinguishing T-cell
almost exclusively affect adults. Patients lymphoma from T-cell-rich large B-cell
ICD-0 code 9766/1 present with seizures, visual defects, fo- lymphoma and inflammation. Their histo-
cal neurological findings, and/or memory logical and immunophenotypic features
impairment {100,1125,1886,2368,2593) may vary, but low-grade appearance is
lntravascu/ar large 8-cell There is no association with immunode- relatively common {450,1503,2341). The
lymphoma ficiency, but one patient with MALT lym- prognosis appears to be similar to or
phoma had a long history of white matter perhaps slightly better than that of dif-
Definition disease with some features of multiple fuse large B-cell lymphoma. Anaplastic
A distinctive lymphoma characterized by sclerosis {2270). and another patient with large cell lymphoma arising in the CNS
exclusively intravascular growth. MALT lymphoma had Chlamydophila has distinctive features and is discussed
Except for the solely cutaneous cases psittaci infection {2005) Treatment has separately.
CNS involvement occurs in > 75-85% varied widely and has included com- EBV-positive nasal-type extranodal
of cases {174). The brain is nearly always plete or partial resection, steroids, radia- NK/T-cell lymphoma primary to the CNS
involved, and spinal cord involvement tion, chemotherapy, and combinations mainly affects young to middle-aged
is less common. The hallmark intravas- of these. The behaviour is indolent com- adult men, has pathological features
cular growth leads to clinical symptoms pared with that of usual primary CNS dif- similar to those seen in other sites, and
fuse large B-cell lymphoma, and patients

276 Lymphomas
is associated with a very poor prognosis
(893,1817,2017l

Anaplastic large cell lymphoma

Primary CNS anaplastic large cell


1ymphoma, ALK-pos!live
Primary CNS anaplastic large cell lym-
phoma (ALCL) is rare. Of all cases of
ALCL, < 1% are primary in the CNS
(2756l Patients are children and young
adults, with reported patient ages rang-
ing from 23 months to 31 years and with
a male preponderance {770,797,1650,
1890,2657l There are no known risk fac-
tors. Patients present with headache, sei-
Fig. 13.09 MALT lymphoma of the dura. Vaguely nodular and diffuse proliferation of lymphoid cells in the frontal dura.
zures, nausea, fever, or a combination of
these (174,1890l They are often initially
thought to have an infection rather than a involvement has been reported {2450l. often mimicking a meningioma {1107,
neoplasm {693,770,1890l. Patients usu- The pathological findings are similar to 1209,1392,2587,2642}. Lymphomas are
ally have one or more intracerebral mass- those of ALK-positive anaplastic large typically localized at presentation {1107,
es, often accompanied by involvement cell lymphoma, except that ALK is not 1392,2587,2642!; in a small minority of
of the leptomeninges and occasionally expressed. The prognosis appears to be cases, extradural disease is identified
even the dura or skull (1890,2199,2756l. worse than that of ALK-positive anaplas- {2642l. Treatment has varied, but nearly
Rarely, lymphoma is confined to the lep- tic large cell lymphoma {797l all patients achieve complete remission
tomeninges {1650,1890l and typically remain well on follow-up
This is an aggressive lymphoma, but with ICD-0 code 9702/3 ( 1107, 1208,1209,1392,1464,2587,2642}.
prompt diagnosis and administration of The histological and immunophenotypic
appropriate chemotherapy, complete re- findings are similar to those of MALT lym-
mission and long-term survival can often Extranodal marginal zone phomas in other sites. The lymphomas
be achieved (770,797,1890,2756l lymphoma of mucosa- are composed of small lymphocytes,
Most cases of ALCL are of the com- marginal zone cells with slightly irregular
associated lymphoid tissue
mon type (1890}. but primary CNS ALCL and pale cytoplasm, few large cells, and,
of the lymphohistiocytic variant (2756l (MALT lymphoma) of the dura frequently, many plasma cells, some-
and combined lymphohistiocytic and Lymphomas primary in the dura mater times with remnants of reactive follicles
small-cell variants (693l have also been are much less frequent than those pri- {1107,1208,1392,2587,2642l and occa-
reported. Biopsy shows large atypical mary in the brain {1107). By far the most sionally with amyloid {1464,2587). Dural
cells, including hallmark cells, with a common dural lymphoma is MALT lym- lymphoma may invade adjacent brain,
variable admixture of reactive cells. The phoma {1392,1464). Exceedingly rare with a tendency to involve Virchow-Robin
tumour cells are positive for CD30, ALK, cases of MALT lymphoma arising in the spaces (1107,1209,2642l The neoplas-
and usually EMA, and show variable ex- brain have also been reported {2270, tic cells are CDS-negative and CD10-
pression of T-lineage antigens {770,1890, 2717). Dural MALT lymphoma affects negative B cells, often with a component
2657l. The underlying genetic abnormali- adults, and women are affected more of- of monotypic plasma cells, indicating
ty is a translocation of the NPM1 and ALK ten than men, with a male-to-female ratio plasmacytic differentiation {1209,2642).
genes {1890l. of approximately 1 :5 {1107,2587,2642). The monotypic plasma cells are lgG4-
Most MALT lymphomas arise in the cra- positive in some cases (6 of 19 cases
ICD-0 code 9714/3 nial dura; patients present with symp- in one series), but evidence of systemic
toms that include headache, seizures, lgG4-related disease is absent (2642}.
Primary CNS anaplastic large cell dizziness, focal neurological defects, Trisomies, most often of chromosome 3,
lymphoma, ALK-negative and visual changes {63,1107,1208,1209, are occasionally detected {2587,2642l.
Primary CNS ALK-negative anaplastic 1392,2642} MALT lymphoma arising in MALT lymphoma-associated trans loca-
large cell lymphoma is a rare neoplasm the dura over the spinal cord is much less tions are rare {186,2642).
that affects adults of both sexes. Pa- common {2642l and may be associated
tients present with one or more intra- with spinal cord compression {1209). Ra- ICD-0 code 9699/3
parenchymal lesions, which are usually diographical evaluation reveals a mass
supratentorial {797l. Diffuse white matter or plaque-like thickening of the dura,

MALT lymphoma of the dura 277


CHAPTER14
Histiocytic tumours
Langerhans cell histiocytosis
Erdheim-Chester disease
Rosai-Dorfman disease
Juvenile xanthogranuloma
Histiocytic sarcoma
Histiocytic tumours Paulus W
Perry A.
Sahm F.

Langerhans cell histiocytosis cases, and cerebral atrophy occurs in


8% {2018}. Rare intraparenchymal CNS
Definition masses have also been described.
A clonal proliferation of Langerhans-type
cells that express CD1a, langerin Clinical features
(CD207), and 5100 protein. The most common neurological sign of
Langerhans cell histiocytosis most fre- Langerhans cell histiocytosis is diabe-
quently affects children. The CNS can tes insipidus (occurring in 25% of cases
be affected via direct invasion from overall and 50% of cases of multisystemic
craniofacial bone and skull base or from disease), with about 60% of patients also
meninges, via extra-axial masses of the showing signs of hypothalamic dysfunc- Fig. 14.01 Langerhans cell histiocytosis. X-ray showing
hypothalamic-pituitary region, or in a tion (e.q obesity, hypopituitarism, and bone lucency at site of disease.
leukoencephalopathy-like pattern, and growth retardation). The clinical features
primary intraparenchymal CNS masses of Langerhans cell histiocytosis-associ- eccentric, ovoid, and reniform or convo-
may also occur. This entity was previous- ated neurodegenerative lesions range luted, with linear grooves and inconspic-
ly referred to as eosinophilic granuloma from asymptomatic imaging abnormali- uous nucleoli. There is abundant pale to
and/or histiocytosis X. ties to tremors, gait disturbances, dysar- eosinophilic cytoplasm, and Touton gi-
thria, dysphagia, motor spasticity, ataxia, ant cells may be seen. Copious collagen
ICD-0 code 9751/3 behavioural disturbances, learning dif- deposition is common. In the neurode-
ficulties, global cognitive deficits, and/or generative lesions of the cerebellum and
Epidemiology psychiatric disease {889). brain stem, there are often no Langer-
Most cases of Langerhans cell histiocy- hans cells, but marked inflammation ac-
tosis occur in childhood, with an annual Imaging companies severe neuronal and axonal
incidence of 0.5 cases per 100 000 indi- Histiocytic lesions of the skull present loss (889). Eosinophils may aggregate
viduals aged < 15 years. as patchy T2-hyperintense lesions, and undergo necrosis, producing granu-
eventually resulting in a punched-out or lomas or abscesses.
Etiology geographical appearance of the bone
The etiology of Langerhans cell histiocy- {2848). lmmunophenotype
tosis is largely unknown. In most patients, Within the CNS parenchyma, Langer- Neoplastic Langerhans cells consistently
there is either mild or no underlying de- hans cell histiocytosis presents as hy- express CD1a, S100 protein, vimen-
fect in immunological integrity. Neverthe- perintense lesions with non-specific tin, and usually langerin (CD207) {438f.
less, an abnormal immune response is enhancement on T2-weighted images. CD68 and HLA-DR content is low and
thought to play a potentially important eti- Involvement of the pituitary region may generally paranuclear. Expression of PT-
ological role; for example, data suggest be associated with loss of the posterior PRC and lysozyme is low. The Ki-67 pro-
defective interactions between T cells pituitary bright spot in T1-weighted im- liferation index is highly variable and can
and macrophages. ages and thickening of the pituitary stalk reach 50%.
in contrast-enhanced images (513,28481.
Localization Genetic profile
Imaging studies of Langerhans cell his- Macroscopy Langerhans cell histiocytosis lesions
tiocytosis have shown that the most com- lntracranial Langerhans cell histiocyto- carry BRAF V600E mutations in 38-
mon presentation of CNS involvement is sis lesions are often yellow or white and 58% of cases (103,2218,2249). Of the
as lesions of the craniofacial bone and range from discrete dural-based nodules BRAF-wildtype Langerhans cell histio-
skull base (seen in 56% of cases), with or to granular parenchymal infiltrates. CNS cytosis cases, 33-50% harbour MAP2K1
without soft-tissue extension. lntracranial, lesions may be well delineated or poorly mutations {290,398)
extra-axial masses are also common, defined.
particularly in the hypothalamic-pituitary Genetic susceptibility
region (seen in 50% of all cases), me- Microscopy The occurrence of multifocal Langerhans
ninges (in 29%), and choroid plexus (in Langerhans cell histiocytosis infiltrates cell histiocytosis in monozygotic twins,
6%). A leukoencephalopathy-like pattern, are composed of Langerhans cells, mac- sometimes with simultaneous onset of
with or without dentate nucleus or basal rophages, lymphocytes, plasma cells, disease, has been repeatedly reported
ganglia neurodegeneration, is seen in and variable eosinophils. The nuclei of and suggests genetic susceptibility in
36% of all Langerhans cell histiocytosis Langerhans cells are typically slightly at least some cases (1562}. It has also

280 Histiocytic tumours


Clinical features
Patients with Erdheim-Chester disease
most commonly present with cerebel-
lar signs (41%), pyramidal syndromes
(45%), and/or diabetes insipidus (47%).
Seizures, headaches, neuropsychiatric/
cognitive impairments, sensory deficits,
cranial neuropathies, and asymptomatic
imaging lesions have also been reported
(1411 ).

Imaging
Extra-axial Erdheim-Chester disease
may resemble meningioma on imaging
(2848). Erdheim-Chester disease lesions
have been reported to demonstrate de-
layed gadolinium enhancement (2554)

Macroscopy
Erdheim-Chester disease manifests as
widespread infiltrative parenchymal le-
Fig. 14.02 Langerhans cell histiocytosis. A Mixed infiltrate composed of histiocytes, lymphocytes, eosinophils, sions (44%), dural thickening I a menin-
and multinucleated cells. B The nuclei of Langerhans cells are typically slightly eccentric, ovoid, and reniform or gioma-like mass (37%), or a combination
convoluted, with linear grooves and inconspicuous nucleoli. C CD1a expression by neoplastic Langerhans cells. (19%) (1411)
D Electron microscopy showing characteristic rod-shaped structures (Birbeck granules).

Microscopy
been suggested that interferon gamma by atypical organ involvement and an Erdheim-Chester disease is composed
and IL4 polymorphisms affect suscep- aggressive clinical course (1051); this of lipid-laden histiocytes with small nu-
tibility to Langerhans cell histiocytosis form is very rare, and the frequency of clei, Touton-type multinucleated giant
and might be responsible for some of the CNS involvement is unknown. Neurode- cells, scant lymphocytes, rare eosino-
clinical variation (541). generative Langerhans cell histiocytosis phils, and variable fibrosis or gliosis.
seems to be progressive, with neurocog- The histiocytic tumour cells can show
Prognosis and predictive factors nitive symptoms present in about 25% of a broad morphological spectrum, with
The overall survival rates of patients with patients after 6 years (889). round or elongated, foamy or eosinophil-
Langerhans cell histiocytosis at 5, 15, ic cytoplasm.
and 20 years are 88%, 88%, and 77%,
respectively, with an event-free survival Erdheirn--(;hesterdisease lmmunophenotype
rate of only 30% at 15 years (2758). Uni- The neoplastic histiocytes are positive for
focal Langerhans cell histiocytosis may Definition CD68 and negative for CD1a and S100
spontaneously recover or requires only Erdheim-Chester disease manifesting in protein.
minimal treatment (e.g. surgical resec- the brain or the meninges.
tion), whereas multisystemic disease Erdheim-Chester disease of the CNS Genetic profile
with organ dysfunction may require corresponds histologically and immuno- Erdheim-Chester disease carries BRAF
systemic chemotherapy. The mortality histochemically to its counterparts occur- V600E mutations in at least 50% of cases
rate for multisystemic Langerhans cell ring elsewhere. Systemic lesions may be and NRAS and PIK3CA mutations in 17%
histiocytosis with organ dysfunction is present or absent. and 12% of cases, respectively. Expres-
20%. Late sequelae are seen in 64% of sion of mutant BRAF protein has been
all patients with Langerhans cell histiocy- ICD-0 code 9750/1 reported to be limited to histiocytes (946).
tosis, including skeletal defects (in 42%),
diabetes insipidus (in 25%), growth fail- Epidemiology Prognosis and predictive factors
ure (in 20%), hearing loss (in 16%), and The mean patient age is 53 years (383). For tumours with BRAFV600E mutation,
other CNS dysfunction (in 14%) (2758). therapy with vemurafenib may lead to
No prognostic significance of histopatho- Localization complete clinical remission of CNS le-
logical features such as cytological atyp- Erdheim-Chester disease may involve sions and systemic disease (655,947).
ia and mitotic activity was found in most the brain (preferentially the cerebellum
studies. However, malignant Langerhans and brain stem), spinal cord, cerebello-
cell histiocytosis does exist, character- pontine angle, choroid plexus, pituitary,
ized morphologically by malignant-ap- meninges, and orbit (1411).
pearing Langerhans cells and clinically

Erdheim-Chester disease 281


Rossi-Dorfman disease weight loss are absent in 70% of patients,
and 52% have no associated systemic
Definition disease !2045).
Rosai-Oorfman disease manifesting in
the brain or the meninges. Imaging
Rosai-Dorfman disease of the CNS Rosai-Dorfman disease of the CNS of-
corresponds histologically and immuno- ten resembles meningioma on imaging.
histochemically to its counterparts occur- However, a lower T2 signal in Rosai-
ring elsewhere. Dorfman disease may help to differenti-
ate it from meningioma !1330).
Epidemiology
The mean patient age is 21 years !1568) Macroscopy
Rosai-Dorfman disease of the CNS is
Localization typically a firm, vaguely lobulated, yellow
Rosai-Dorfman disease of the CNS to greyish-white dural mass.
forms solitary or multiple dural masses, Fig. 14.03 MRI of a case of dural-based Rosai-Dorfman
especially in the cerebral convexity, cra- disease mimicking a meningioma. Microscopy
nial base, and cavernous sinuses, as well Rosai-Dorfman disease presents as a
as parasagittal, suprasellar and petro- vomiting, dizziness, epilepsy, fever, ma- multinodular mass composed of a mixed
clival regions !2551 }. Parenchymal or in- laise, weight loss, night sweats, and/ inflammatory infiltrate including large
trasellar lesions may also occur. or specific localized symptoms !2551} pale histiocytes, numerous lymphocytes
Patients with sellar lesions present with and plasma cells, and variable fibrosis.
Clinical features signs of hypopituitarism and diabetes Emperipolesis with histiocytic engulfment
Patients with Rosai-Dorfman disease insipidus. The classic systemic signs of of intact lymphocytes, plasma cells, neu-
present with headache, nausea and cervical lymphadenopathy, fever, and trophils, and occasionally eosinophils is
typical, but may be inconspicuous on
H&E staining. However, emperipolesis
is not pathognomonic for Rosai-Dorf-
man disease and may occasionally
be encountered in other neoplastic or
non-neoplastic histiocytes and even in
astrocytes.

lmmunophenotype
The neoplastic histiocytes are posi-
tive for CD11c, CD68, L1 antigen (clone
MAC387), and S100 protein; variably
positive for lysozyme; and negative for
CD1a and langerin.

Genetic profile
No recurrent genetic aberrations have
been identified in Rosai-Dorfman dis-
ease to date. Clonality studies have dem-
onstrated polyclonality of the infiltrates
in two cases of Rosai-Dorfman disease
!1912).

Juvenile xanthogranuloma
Definition
Juvenile xanthogranuloma arising in the
brain or the meninges, either with or with-
out cutaneous lesions.
Juvenile xanthogranuloma of the CNS
,I>
corresponds histologically and irn-
Fig. 14.04 Rosai-Dorfman disease. A Multinodular mass composed of a mixed inflammatory infiltrate, including munohistochemically to its cutaneous
large pale histiocytes, numerous lymphocytes, and plasma cells. B Emperipolesis with histiocytic engulfment of counterpart.
intact lymphocytes, plasma cells, neutrophils, and eosinophils. C CD45 expression by phagocytosed haematopoietic
cells.
282 Histiocytrc tumours
Epidemiology
The mean patient age is 22 months
(1134).

Localization
Juvenile xanthogranuloma of the CNS
localizes to the brain (53%), intradural ex-
tramedullary spine (13%), or nerve roots
(15%), with meningeal involvement also
being common (568).

Clinical features

.
The signs and symptoms of juvenile
xanthogranuloma depend on the sites
of involvement, but often include sei-
zures, diabetes insipidus, and visual �
Fig. 14.05 Juvenile xanthogranuloma. Note numerous histiocytic cells and two large multinucleated Touton cells.
disturbances.

Macroscopy
Juvenile xanthogranuloma lesions are of-
ten received as fragmented, soft, yellow
to tan-pink biopsy specimens.

Microscopy
Juvenile xanthogranuloma is composed
of rounded to spindled, variably vacuolat-

-
ed histiocytes, scattered Touton and for-
eign body-type giant cells, lymphocytes,
and occasional eosinophils (1411) Fig. 14.06 Histiocytic sarcoma. A Large, highly pleomorphic histiocytic tumour cells with irregular nuclei and prominent
nucleoli. B The lineage-specific marker CD163 was diagnostic in this high-grade neoplasm.
lmmunophenotype
The neoplastic histiocytes of juvenile Localization lmmunophenotype
xanthogranuloma are CD1a-negative, Reported cases of histiocytic sarcoma in The tumour cells are typically positive for
CD11c-positive, CD68-positive, fac- the CNS have involved the brain paren- histiocytic markers (e.g. CD68, CD163,
tor Xllla-positive, negative or positive chyma, meninges, and cavernous sinus lysozyme, CD11c, and CD14), variably
for MAC387, lysozyme-negative, and {421). positive for CD34, and negative for my-
81 GO-negative. eloid antigens, dendritic antigens, CD30,
Clinical features ALK, and other lymphoid markers, as
Patients with histiocytic sarcoma present well as for glial, epithelial, and melano-
Histiocytic sarcoma with variable initial signs and symptoms, cytic antigens. The tumour cells are
which tend to progress rapidly due to dis- negative for the follicular dendritic cell
Definition seminated disease {421 ). antigens CD23 and CD35. However, fol-
A rare, aggressive, malignant neoplasm licular dendritic cell sarcoma expressing
with the histological and immunopheno- Macroscopy these antigens may primarily arise in the
typic characteristics of mature histiocytes. Histiocytic sarcomas are destructive, brain and must be differentiated from his-
Most cases of histiocytic sarcoma occur soft, fleshy whit€ masses with occasional tiocytic sarcoma (966).
in adults. yellow necrotic foci.
Genetic profile
ICD-0 code 9755/3 Microscopy Histiocytic sarcoma is not defined by a
Histiocytic sarcoma is characterized by certain genetic profile, although a sin-
Epidemiology highly cellular, non-cohesive infiltrates gle case with BRAF V600E mutation has
The mean reported patient age in case of large, moderately pleomorphic, mitoti- been described (1085). They usually
series is 52 years (1043). cally active histiocytes with abundant eo- lack IGH or T-cell receptor gene clonal-
sinophilic cytoplasm, variably indented ity (497). Cases with evidence of clonal
Etiology to irregular nuclei, and often prominent rearrangement are attributed to transdif-
Isolated cases of radiation-associated nucleoli. Occasional multinucleated or ferentiation from neoplastic T- or B-cell
histiocytic sarcoma of the CNS have spindled forms are also common, as is precursor lesions (2493).
been reported {399,2795). background reactive inflammation (421).

Histiocytic sarcoma 283


CHAPTER15
Germ cell tumours
Germinoma
Embryonal carcinoma
Yolk sac tumour
Choriocarcinoma
Teratoma
Mixed germ cell tumour
Germ cell tumours Rosenblum M.K.
Nakazato Y.
lchimura K.
Leuschner I.
Matsutani M. Huse J.T.

Definition Peak incidence occurs in patients aged most common site) (1025,2251,2278).
In the CNS, the morphological, immuno- 10-14 years, and a clear majority of cas- Suprasellar examples originate in the
phenotypic, and (in some respects) ge- es of all histological types involve males neurohypophyseal I infundibular stalk.
netic homo/agues of gonadal and other (210,485,1017,2251}. Analysis of a regis- lntraventricular, diffuse periventricular,
extraneuraxial germ cell neoplasms. try containing 1463 Japanese patients thalamostriate, cerebral hemispheric,
The major germ cell tumour types are found that 70% were aged 10-24 years cerebellar, bulbar, intramedullary, and
germinoma, teratoma. yolk sac tumour, and 73% were male (485). Congenital intrasellar variants can be encountered,
embryonal carcinoma, and choriocarci- examples (typically teratomas) are well as can congenital holocranial examples
noma. Neoplasms harbouring multiple recognized, but only 2.9% of patients in (usually teratomas). Germinomas prevail
types are called mixed germ cell tumours. this series were aged < 5 years, and only in the suprasellar compartment and ba-
Otherwise pure germinomas containing 6.2% were aged > 35 years. The great sal ganglionic I thalamic regions, and
syncytiotrophoblastic giant cells are rec- majority of pineal region cases affect non-germinomatous subtypes predomi-
ognized as a distinct variant. Teratomas boys, whereas an excess of suprasellar nate at other sites. Multifocal CNS germ
are subclassified as mature, immature, or lesions occur in girls. In the Japanese cell tumours usually involve the pineal re-
exhibiting malignant transformation. registry cited above, 89% of teratomas. gion and suprasellar compartment, either
78% of germinomas, and 75% of other simultaneously or sequentially. Bilateral
Epidemiology germ cell tumour types arose in males. basal ganglionic and thalamic lesions
CNS germ cell tumours principally affect Pure germinomas outnumber other types. are also well recognized.
children and adolescents and seem to be followed by mixed lesions and teratomas.
more prevalent in eastern Asia than in the In a series of 153 histologically verified Clinical features
Europe and the USA. These tumours ac- cases (1612). 41.1% were germinomas The clinical manifestations and their du-
count for 2-3% of all primary intracranial (including examples with syncytiotropho- ration vary with histological type and
neoplasms and for 8-15% of paediatric blastic elements. which accounted for location. Germinomas are generally as-
examples in series from Japan; Taiwan, 5.2% of all cases). 32% were mixed germ sociated with a more protracted symp-
China; and the Republic of Korea (485A, cell tumours, 19.6% were teratomas (of tomatic interval than are other types. Le-
1017,1192A,1612,2452l, but for only 0.3- which 63.3% were mature, 23.3% were sions in the pineal region compress and
0.6% of primary intracranial tumours and immature, and 13.3% exhibited malig- obstruct the cerebral aqueduct, resulting
3-4% of those affecting children in se- nant elements), 3.3% were embryonal in progressive hydrocephalus with intra-
ries in Europe and North America (210, carcinomas, 2% were yolk sac tumours, cranial hypertension. These lesions are
506A, 558, 596, 1025, 1154, 1862A, 1863}. and 2% were choriocarcinomas. How- also prone to compressing and invading
The highest reported incidence statis- ever, the relative incidence rates of the the tectal plate, producing a paralysis of
tics come from Japan, where a recent specific tumour types vary according to upwards gaze and convergence called
survey of Kumamoto Prefecture revealed location. Parinaud syndrome. Neurohypophyseal/
an annual age-adjusted incidence rate suprasellar germ cell tumours impinge
of 045 cases per 100 000 population Etiology on the optic chiasm, causing visual field
aged < 15 years. more than double the CNS germ cell tumours' predilection to defects, and often disrupt the hypo-
rates in the USA and Germany (1573). occur in peripubertal patients, their local- thalamohypophyseal axis, precipitating
ization in diencephalic centres regulating diabetes insipidus and manifestations of
Males
90-100
Females gonadal activity, and their increased inci- pituitary failure such as delayed growth
80-89 dence in Klinefelter syndrome have been and sexual maturation. The secretion by
70-79 regarded as evidence that elevated cir- neoplastic syncytiotrophoblasts of hCG,
60-69
culating gonadotropin levels play a role in a stimulant of testosterone production.
50-59
their pathogenesis. The association with can cause precocious puberty (isosex-
40-49

30-39
Klinefelter syndrome could also reflect ual pseudoprecocity) in boys. The ad-
20-29 X chromosome overdosage, a common ditional expression of cytochrome P450
10-19 genetic feature of these neoplasms. aromatase. which catalyses the conver-
0-9 sion of C19 steroids to estrogen, may
600 400 200 200 400 600
Localization explain the rare instances of precocious
Number of cases Number of cases
Fig. 15.01 Age and sex distribution of CNS germ cell
About 80% of CNS germ cell tumours puberty in girls with hCG-producing tu-
tumours, based on 1463 cases; data from a report arise along a midline axis extending from mours (1814) In this setting, hCG may
published by the Brain Tumor Registry of Japan (1969- the pineal gland {their most common site) also have some intrinsic follicle stimulat-
1996). to the suprasellar compartment {their next ing hormone-like activity (2415).

286 Germ cell tumours


imaging Alternative hypotheses instead implicate CDK I retinoblastoma protein I E2F path-
_Germ cell tumours other than teratomas native stem cells of embryonic (i.e pluri- way {2536).
-tend to present as solid and contrast- potent) or neural type 11850,25021. These Upregulated KIT/RAS signalling, which
enhancing on CT and MRI, with germi- formulations require the selective genetic is a feature of gonadal and mediasti-
nomas often enhancing more homoge- programming of such precursors along nal seminomas, is also evident in most
neously than other subtypes 1748,14971. the germ cell differentiation pathway, as intracranial germinomas; most manifest
Tumour tissue is usually hypointense/ well as their neoplastic transformation. mutually exclusive activating KIT or RAS
· isointense on Ti-weighted images and Supporting the neural stem cell hypoth- family member mutations that are associ-
isointense/hyperintense on T2-weighted esis is evidence that such cells share ated with severe chromosomal instability
images Thalamic and basal ganglia ger- hypomethylation of the imprinted SNRPN !758,2689). These alterations are only
minomas may be more prone to calcifica- gene with primordial germ cell elements infrequently found in other germ cell tu-
tion and cystic change than those in the and intracranial germ cell tumours (as mour types. Inactivating mutation of CBL,
pineal or suprasellar regions and can ex- well as with gonadal and other extraneu- which encodes a negative KIT regulator,
hibit little Ti-weighted signal abnormality raxial germ cell tumours) 114591. Experi- has also been described in CNS germi-
and only poorly defined T2-hyperinten- mental data suggest that overexpression noma (2689} Less prevalent abnormali-
t sity, with minimal or no enhancement. of OCT4 in such cells induces teratoma ties manifested by a variety of intracranial
lntratumoural cysts. calcified regions, formation 125021. Some authors consid- germ cell tumour types include inactivat-
and components with the low-signal- er the characteristically pure teratomas ing mutations of BCORL 1 (a tumour sup-
attenuation characteristics of fat sug- of the spinal cord to be bona fide neo- pressor gene) and activating AKT/mTOR
gest teratoma. whereas haemorrhage is plasms of germ cell origin 1371. but oth- lesions (2689). Epigenetic alterations
commonly associated with tumours com- ers contend that they are in fact complex shared by gonadal and some intracranial
posed (at least in part) of choriocarcino- malformations 113191. germ cell tumours include hypomethyla-
ma. Neuroendoscopic evaluation can re- tion of the imprinted SNRPN gene {1459)
veal tumour spread along or beneath the Genetic profile and of the JGF2/H19 imprinting control re-
ependymal lining of a ventricle that is not At the genetic level, pure intracranial gion [2359). Finally, as a pertinent nega-
evident on MRI 12732} Congenital germ teratomas presenting as congenital or in- tive. amplification of the chromosome 19
cell neoplasms (typically teratomas) can fantile growths differ fundamentally from microRNA cluster (C19MC) has not been
be detected in utero by ultrasonography CNS germ cell tumours arising after early identified in immature teratoma, despite
or fast MRI 11583} childhood. Whereas pure intracranial the frequent occurrence of neural tube-
teratomas presenting as congenital or like rosettes in this tumour type [1798).
Cell of origin infantile growths resemble teratomas of
Although the histological, immunophe- the infant testis in their typically diploid Genetic susceptibility
notypic. and genetic attributes shared status and general chromosomal integ- CNS germ cell tumours typically oc-
by gonadal and neuraxial germ cell rity (2119). CNS germ cell tumours aris- cur sporadically. An increased risk of
tumours are compatible with the tradi- ing after early childhood, irrespective of intracranial (and mediastinal) germ cell
tional assumption that neuraxial germ their histological composition, share with tumorigenesis is associated with Kline-
cell tumours derive from primordial germ their testicular counterparts in young felter syndrome, which is characterized
cells that either aberrantly migrate to or men characteristically aneuploid pro- by a 47 XXY genotype {1202). This as-
purposefully target developing CNS, the files, complex chromosomal anomalies, sociation may reflect an increased dos-
issue of histogenesis remains controver- and overlapping patterns of net genetic age of an X chromosome-associated
sial. Studies of the human CNS, includ- imbalance (1833,2121,2292,2453,2536} gene, given that CNS (and other) germ
ing immunohistochemical analysis of These imbalances are primarily gains of cell tumours commonly exhibit additional
fetal pineal glands with antibodies to the 12p, 8q, tq, 2p, 7q, 10q, and X. as well X chromosomes, as well as the potenti-
primordial germ cell marker PLAP, have as losses of t lq, 13, Sq, 10q, and 18q. ating effects of chronically elevated gon-
never shown it to harbour primitive germ Whether 12p gain and isochromosome adotropin levels. Down syndrome, which
cell elements 1684). However, it has been 12p formation, which are especially char- is associated with an increased risk of
argued that germ cells might differentiate acteristic of testicular and mediastinal testicular germ cell tumorigenesis, may
into divergent forms upon entering the germ cell tumours, also occur at relatively also be complicated by intracranial germ
CNS. Specifically, an enigmatic popula- high frequencies in the CNS setting has cell neoplasia (436,956) CNS germ cell
tion of skeletal muscle-like cells native been debated, as has the prevalence of tumours have also been described in
to the developing pineal gland has been X duplication in this locale. The weight the setting of neurofibromatosis type 1
suggested to descend from primitive ger- of data suggests that fewer CNS germ [2784} in siblings (84,2675). in a par-
minal elements migrating during neuro- cell tumours exhibit isochromosome 12p ent and child (84). and in the fetus (an
embryogenesis 12173). Cited in support (1833,2121,2292,2453,2536}. Regions intracranial teratoma) of a woman with
of this seemingly far-fetched idea is the of particular gain have been reported independent ovarian teratoma (2009).
fact that striated muscle-type cells of un- to encompass CCND2 and PRDM14, a Rarely, patients with CNS germ cell tu-
known function also populate the thymus, regulator of primordial germ cell specifi- mours have been reported to develop
another organ ostensibly devoid of germ cation, and losses of the RB1 locus have second gonadal or mediastinal germ cell
cells and yet a favoured site of extrago- been suggested to implicate the cyclin I neoplasms (956,1109,2707). One such
nadal germ cell tumorigenesis. patient had Down syndrome {956). It has

Overview 287
pure germinoma {2495). The most viru- germ cell tumour, and it can also occur
lent CNS germ cell tumours are yolk sac as a component of a mixed germ cell tu-
tumours, embryonal carcinomas, chorio- mour, in combination with other germ cell
carcinomas, and mixed lesions in which tumours. Germinoma is extremely sensi-
these types are prominent. In contrast, tive to radiotherapy, and cure rates are
immature teratomas and mixed tumours high.
dominated by teratoma or germinoma
with only minor high-grade, non-germino- ICD-0 code 9064/3
matous components seem to occupy an
intermediate position in terms of biologi- Macroscopy
cal behaviour {1611,2251). Survival rates Germinomas are composed of soft and
as high as 60-70% have been achieved friable tan-white tissue. They are gener-
through treatment of these more aggres- ally solid, but may exhibit focal cystic
sive tumours with combined chemo- change. Haemorrhage and necrosis are
therapy and irradiation {1665) Local re- rare.
Fig. 15.02 MRI of a solid, contrast-enhancing germinoma currence and cerebrospinal fluid-borne
of the pineal region, with a smaller cerebrospinal fluid- dissemination are the usual patterns of Microscopy
borne metastasis in the suprachiasmatic cistern. progression, but abdominal contamina- Germinomas contain large, undifferenti-
tion via ventriculoperitoneal shunts and ated cells that resemble primordial ger-
haematogenous spread (principally to minal elements. These are arranged in
lung and bone) can also occur. sheets, lobules, or (in examples mani-
festing stromal desmoplasia) regimented
cords and trabeculae. The cytological
Germinoma features include round, vesicular, and
centrally positioned nuclei; prominent
Definition nucleoli; discrete cell membranes; and
A malignant germ cell tumour histologi- relatively abundant cytoplasm, which is
cally characterized by the presence of often clear due to glycogen accumula-
large primordial germ cells with promi- tion. Mitotic activity is apparent and may
Fig. 15.03 Germinoma of the suprasellar region in a nent nucleoli and variable cytoplasmic be conspicuous, but necrosis is uncom-
7-year-old girl. clearing. mon. Delicate fibrovascular septa (varia-
Nearly all germinomas contain a substan- bly infiltrated by small lymphocytes) are a
been suggested that germline variants of tial population of reactive lymphoid cells. typical feature; some germinomas show
a chromatin-modifying gene, JMJ01C, Germinoma is the most common CNS a lymphoplasmacellular reaction so florid
may be associated with an increased risk
of intracranial germ cell tumours in Japa-
nese {2689).

Prognosis and predictive factors


The factor that bears most heavily on
outcome is histological subtype {1025,
1611,1612,2278). Mature teratomas are
potentially curable by surgical excision.
Pure germinomas are remarkably radio-
sensitive, with long-term survival rates of
> 90% after craniospinal irradiation alone
{1665). The addition of chemotherapy to
treatment regimens may provide compa-
rable germinoma control at lower radia-
tion doses and field volumes {1210,1449,
1611,1665,2251). Whether germinomas
harbouring syncytiotrophoblastic cells
or associated with elevated beta-hCG
levels carry an increased risk of recur-
rence and require intensified therapy
has been controversial {1665). A recent
study demonstrating beta-hCG mRNA
expression across CNS germ cell tumour
types did not find that mRNA levels cor-
related with recurrence in the setting of

288 Germ cell tumours


Embryonal carcinoma
Definition
An aggressive non-germinomatous ma-
lignant germ cell tumour characterized
by large epithelioid cells resembling
those of the embryonic germ disc.
Other common features of embryonal
carcinoma are geographical necrosis, a
high mitotic count, and pseudoglandular
or pseudopapillary structures. Embryonal
carcinoma can also occur as a compo-
nent of a mixed germ cell tumour, in com-
bination with other germ cell tumours.

ICD-0 code 9070/3

Macroscopy
Embryonal carcinomas are solid lesions
composed of friable greyish-white tissue
. """ that may exhibit focal haemorrhage and
Fig. 15.05 Germinoma. A Membranous and Golgi region immunolabelling for KIT. B lmmunoreactivity for OCT4. necrosis.
C Cytoplasmic and membranous reactivity for PLAP. D Expression of KIT protein in tumour cells.
Microscopy
that it obscures the neoplastic elements. Their biological significance has been Embryonal carcinomas are composed
Germinomas that provoke an intense controversial {1665), but the presence of of large cells that proliferate in nests and
granulomatous response can resemble such cells should not prompt a diagnosis sheets, form abortive papillae, or line
sarcoidosis or tuberculosis (1331). of choriocarcinoma. gland-like spaces. Tumour cells excep-
The reactive lymphoid elements within tionally form so-called embryoid bodies
/mmunophenotype germinomas are usually dominated by replete with germ discs and miniature
Consistent cell membrane and Golgi T cells, including both CD4-expressing amniotic cavities. Other typical histo-
region immunoreactivity for KIT and helper/inducer and CDS-expressing logical features include macronucleoli,
membranous 02-40 labelling distin- cytotoxic/suppressor elements (2250, abundant clear to violet-hued cytoplasm,
guish germinomas from solid variants 2719), but CD20-labelling B cells and a high mitotic count, and zones of coagu-
of embryonal carcinoma and yolk sac CD138-labelling plasma cells may be lative necrosis.
tumour (1080). Less consistent (and conspicuous, and constitute evidence
non-specific) is cytoplasmic or membra- of humoral immune responses to tumour lmmunophenotype
nous PLAP expression {210,1017,2196). {2759). Cytoplasmic immunoreactivity for CD30,
Germinomas regularly display immuno- although potentially shared by the epi-
reactivity for the RNA-binding LIN28A thelial and mesenchymal components
protein !353) and nuclear expression of of teratomas, distinguishes embryonal
the transcription factors NANOG {2239), carcinomas from other germ cell tumours
OCT4 (1080), ESRG (2695), UTF1 (1885), (1080) Uniformly and strongly reactive
and SALL4 (1631), but are typically non- for cytokeratins and often positive for
reactive for CD30 and alpha-fetoprotein PLAP (210,684,1017,2196), these tu-
(210,684,1017,2196). A minority display mours also display labelling for LIN28A
cytoplasmic labelling by the CAM5.2 (353) and nuclear expression of OCT4
and AE1/AE3 cytokeratin antibodies, (1080), ESRG (2695), UTF1 (1885),
a phenomenon which may, along with SALL4 {1080), and SOX2 (2240). KIT
ultrastructural evidence of intercellular expression may be seen and is gener-
junction and lumen formation {1672), in- ally focal and non-membranous {1080),
dicate a capacity to differentiate along but embryonal carcinomas are typically
epithelial lines or towards embryonal negative for alpha-fetoprotein, beta-hCG,
carcinoma, but one that is without dem- and human placental lactogen {210,684,
onstrated clinical importance. Otherwise 1017,2196)
pure germinomas may harbour syncytio-
trophoblastic elements that express be-
ta-hCG and human placental lactogen.

Embryonal carcinoma 289


Choriocarcinoma
Definition
An aggressive non-germinomatous ma-
lignant germ cell tumour composed
of syncytiotrophoblasts, cytotropho-
blasts, and occasionally intermediate
trophoblasts.
Necrosis and haemorrhage are often
present in choriocarcinoma. There is
usually a marked elevation of hCG in the
blood or cerebrospinal fluid. Choriocarci-
noma can also occur as a component of
a mixed germ cell tumour, in combination
with other germ cell tumours.

ICD-0 code 9100/3

• ,0 Macroscopy
• I � �,· � Choriocarcinomas are solid, typically
s'. H1-
Y-. ;?- "' ... V . _...._-�""'
.• _ c. haemorrhagic, and often extensively
Fig. 15.07 Yolk sac tumour. A Typical sinusoidal growth pattern. B Schiller-Duval body and numerous mitoses. necrotic.
C Reticular growth pattern with numerous hyaline globules. D Alpha-fetoprotein immunolabelling.
Microscopy
Yolk sac tumour projections to form papillae called Schil- Histological diagnosis requires the pres-
ler-Duval bodies. Yolk sac tumours can ence of both cytotrophoblastic elements
Definition also contain eccentrically constricted and syncytiotrophoblastic giant cells.
An aggressive non-germinomatous ma- cysts delimited by flattened epithelial The giant cells typically contain multiple
lignant germ cell tumour composed of elements (termed 'polyvesicular vitelline hyperchromatic or vesicular nuclei, often
primitive germ cells arranged in various pattern'), enteric-type glands with goblet clustered in a knot-like fashion, within
patterns, which can recapitulate the yolk cells, and foci of hepatocellular differenti- a large expanse of basophilic or vio-
sac, allantois, and extra-embryonic mes- ation (termed 'hepatoid variant'). Brightly laceous cytoplasm. Neoplastic syncytio-
enchyme and produce alpha-fetoprotein. eosinophilic, periodic acid-Schiff-posi- trophoblast surrounds or partially drapes
Yolk sac tumour can also occur as a tive, and diastase-resistant hyaline glob- cytotrophoblastic components, which
component of a mixed germ cell tu- ules are characteristic, although variable; consist of cohesive masses of large
mour, in combination with other germ cell they may occupy the cytoplasm of epi- mononucleated cells with vesicular nu-
tumours. thelial cells or lie in extracellular spaces. clei and clear or acidophilic cytoplasm.
Mitotic activity varies considerably, and Ectatic vascular channels, blood lakes,
ICD-0 code 9071/3 necrosis is uncommon. and extensive haemorrhagic necrosis are
characteristic.
Macroscopy lmmunophenotype
Yolk sac tumours are typically solid and Cytoplasmic immunoreactivity of epi- lmmunophenotype
greyish tan. They are usually friable or thelial elements for alpha-fetoprotein, al- Syncytiotrophoblasts are characterized
(due to extensive myxoid change) gelati- though potentially shared by the enteric by diffuse cytoplasmic immunoreactivity
nous in consistency. Focal haemorrhage glandular components of teratomas, dis- for beta-hCG and human placental lac-
may be apparent. tinguishes -yolk sac tumours from other togen {210,684,1017,2196l. Cytokeratin
germ cell neoplasms {210,684,1017,
Microscopy 2196l. Hyaline globules are also reactive.
This neoplasm is composed of primitive- Epithelial components consistently label
looking epithelial cells (which putatively for cytokeratins, are frequently positive
differentiate towards yolk sac endoderm) for glypican-3 {1631), and may also be
set in a loose, variably cellular, and often positive for PLAP. Yolk sac tumours ex-
myxoid matrix resembling extraembry- hibit labelling for LIN28A {353l and nu-
onic mesoblast. The epithelial elements clear expression of SALL4 {1631l OCT4
may form solid sheets, but are more com- expression is exceptional. KIT reactivity
monly arranged in a loose network of ir- is also rare; when present, it is typically
regular tissue spaces (termed 'reticular focal, cytoplasmic rather than membra-
pattern') or around anastomosing sinu- nous, and without Golgi area accentua-
soidal channels as a cuboidal epithelium, tion {1080l. Human placental lactogen
in some cases draped over fibrovascular and beta-hCG are not expressed.

290 Germ cell tumours


Mature teretoms

ICD-0 code 9080/0

Microscopy
Mature teratomas consist entirely of fully
differentiated, adult-type tissue elements
that exhibit little or no mitotic activity. Ec-
todermal components commonly include
epidermis and skin appendages, cen-
Fig. 15.09 Sagittal T1-weighted MRI of a teratoma in the
tral nervous tissue, and choroid plexus. Fig. 15.10 Large immature teratoma of the cerebellum
pineal region, occupying the dorsal aspect of the third Smooth and striated muscle, bone, car- in a 4-week-old infant, with characteristic cysts and
ventricle. tilage, and adipose tissue are typical chondroid nodules.
mesodermal components. Glands, often
cystically dilated and lined by respiratory
labelling is also demonstrable, with some or enteric-type epithelia, are the usual encountered are rhabdomyosarcomas
choriocarcinomas also expressing PLAP, endodermal components, but hepatic and undifferentiated sarcomas (210,
but KIT and OCT4 labelling are not seen and pancreatic tissue may also be en- 1612,2196), followed by enteric-type
(1080). countered. Gut- and bronchus-like struc- adenocarcinomas (737, 1275). squa-
tures replete with muscular coats or car- mous carcinomas (1612). and primitive
tilaginous rings, respectively, as well as neuroectodermal tumours (2595). Eryth-
Teratoma mucosa may also be formed. roleukaemia (987) and leiomyosarcoma
(2369) have also been reported to arise
Definition Immature teratoma in this setting, as has a carcinoid tumour
A germ cell tumour composed of somatic associated with an intradural spinal tera-
tissues derived from two or three of the ICD-0 code 9080/3 toma {1096). The pathogenesis of an in-
germ layers (i.e. the ectoderm, endo- trasellar tumour containing elements of
derm, and mesoderm). Microscopy germinoma and Burkitt-like B-cell lym-
Teratomas can be further subclassified Immature teratomas consist of incom- phoma is unclear (2609). Yolk sac tumour
as mature teratomas, which are com- pletely differentiated elements resem- components (rather than teratomatous
posed exclusively of mature, adult-type bling fetal tissues. When admixed with components) have been speculated to
tissues (e.g. mature skin, skin appendag- mature tissues, the presence of any im- be the progenitors of select enteric-type
es, adipose tissue, neural tissue, smooth mature teratoma component mandates adenocarcinomas originating in intracra-
muscle, cartilage, bone, minor salivary classification of the tumour as immature nial germ cell tumours (737).
glands, respiratory epithelium, and gas- teratoma, even if the incompletely differ-
trointestinal epithelium) and immature entiated elements constitute only a small
teratomas, which contain immature, em- part of the neoplastic process. Common Mixed germ cell tumour
bryonic, or fetal tissues either exclusively features are compact and mitotically ac-
or in addition to mature tissues. Rare tive stroma reminiscent of embryonic ICD-0 code 9085/3
teratomas contain a component result- mesenchyme, as well as primitive neuro-
ing from the malignant transformation of ectodermal elements that may form neu- Macroscopy
a somatic tissue, usually a carcinoma or roepithelial multilayered rosettes with a The appearance of a mixed germ cell tu-
sarcoma, but embryonal tumours with the central lumen or canalicular arrays that mour reflects the macroscopic features
features of a primitive neuroectodermal resemble developing neural tube. Clefts of the constituent germ cell tumour com-
tumour can also arise in this setting in the lined by melanotic neuroepithelium are ponents, as have been described for the
CNS, a fact prompting careful evaluation often seen; these result from abortive pure forms.
in certain clinicopathological settings, differentiation of the retinal pigment
such as a pineal tumour of childhood. epithelium. Microscopy
Pathologists should specify the type of Any combination of germ cell tumour var-
secondary cancer present and avoid iants can be encountered. Pathologists
the non-specific designation "malignant Teratoma with malignant reporting such lesions must specify the
teratoma". transformation subtypes present and state the relative
proportions of each.
ICD-0 code 9080/1 ICD-0 code 9084/3
lmmunophenotype
Microscopy Individual components have the same
Teratoma-containing intracranial germ antigenic profiles described for the pure
cell tumours can include a variety of so- forms of these tumour variants.
matic-type cancers; the most commonly

Teratoma 291
CHAPTER16
Familial tumour syndromes
Neurofibromatosis type 1
Neurofibromatosis type 2
Schwannomatosis
Von Hippel-Lindau disease
Tuberous sclerosis
Li-Fraumeni syndrome
Cowden syndrome
Turcot syndrome
Naevoid basal cell carcinoma syndrome
Rhabdoid tumour predisposition syndrome
Neurofibromatosis type 1 Reuss D.E.
von Deimling A.
Perry A.

Definition Neurofibromas
An autosomal dominant disorder char- Among the major subtypes of neurofibro-
acterized by neurofibromas, multiple ma, the dermal and plexiform variants are
cate-au-lait spots, axillary and inguinal characteristic of NF1. Deep-seated local-
freckling, optic gliomas, osseous lesions ized intraneural neurofibromas arise less
and iris hamartomas (Lisch nodules). Pa- commonly, and may cause neurological
tients with neurofibromatosis type 1 (NF1) symptoms. Plexiform neurofibromas pro-
have an increased risk for malignant pe- duce diffuse enlargement of major nerve
ripheral nerve sheath tumour (MPNST), trunks and their branches, sometimes
gastrointestinal stromal tumour, rhabdo- yielding a rope-like mass, and are almost
myosarcoma, juvenile chronic myeloid pathognomonic of NF1. Plexiform neu-
leukaemia, duodenal carcinoids, C-cell rofibromas may develop during the first
hyperplasia I medullary thyroid carcino- 1-2 years of life, as single subcutaneous Fig. 16.01 Pilocytic astrocytoma of the optic nerve
(optic nerve glioma; arrowhead} in a patient with
mas, other carcinomas, and phaeochro- swellings with poorly defined margins.
neurofibromatosis type 1.
mocytoma. The disorder is caused by They may also cause severe disfigure-
mutations of the NF1 gene on chromo- ment later in life, affecting large areas
some 17q11.2. of the body. If these tumours arise in the
head or neck region, they can impair vital
OMIM number (1624) 162200 functions. Plexiform neurofibromas have
a lifetime risk of malignant progression to
Incidence/epidemiology MPNST of about 10% (1006}.
The birth frequency of NF1 has been esti-
mated to be about 1 case per 3000 births Malignant peripheral nerve sheath
(658,2680). tumours
The MPNSTs that arise in patients with
Sites of involvement NF1 usually occur at a younger age, may
Multiple sites and organ systems may be be multiple, and may include divergent
involved. The most commonly involved differentiations with rhabdomyoblastic
are the central and peripheral nervous elements (malignant triton tumour) or
system, the skin, the eyes, and the bones glandular elements (glandular MPNST)
(see Table 16 01). MPNSTs reduce life expectancy signifi-
cantly (663}.

Gliomas Fig. 16.02 Macroscopic preparation of a bilateral optic


Table 16.01 National Institutes of Health {NIH) diagnostic Most gliomas in patients with NF1 are nerve glioma in a patient with neurofibromatosis type 1.
criteria for neurofibromatosis type 1 (NF1) {739A}
pilocytic astrocytomas within the optic
The presence of l!: 2 of the following features is nerve. Bilateral growth is characteristic of
diagnostic:
NF1. Optic nerve gliomas in patients with
Six or more cate-au-Iait macules more than 5 mm NF1 may remain static for many years,
in greatest diameter in prepubertal individuals and some may regress (1516}. Other glio-
and more than 15 mm in greatest diameter in
mas observed at an increased frequency
postpubertal individuals
in NF1 include diffuse astrocytomas and
Two or more neurofibromas of any type or one glioblastomas {908,1470).
plexiform neurofibroma
Freckling in the axillary or inguinal regions Other CNS mamfestations
Opticglioma The following features are more frequent
Two or more Lisch nodules (iris hamartomas}
in NF1: macrocephaly (2476}, learning
disabilities and attention deficit hyper- Fig. 16.03 Bilateral optic nerve glioma in a patient
A distinctive osseous lesion such as sphenoid activity disorder (1075). epilepsy (1859).
dysplasia or tibial pseudarthrosis with neurofibromatosis type 1. Note the enlargement
aqueductal stenosis, hydrocephalus of the compartments of the optic nerves and collar-like
A first-degree relative (parent, sib, or offspring) with (587}, and symmetrical axonal neuropa- extension into the subarachnoid space. Masson stain.
NF1 as defined by the above criteria thy (691}.

294 Familial tumour syndromes


Extraneural mamfestations Table 16.02 Manifestations of neurofibromatosis type 1 can be found in two major isoforms, of
Abnormalities ofpigmentation Tumours 2818 amino acids (type 1) and 2839 ami-
cafe-au-lait spots, freckling, and Lisch Neurofibromas no acids (type 2), respectively. Neurofi-
nodules all involve alterations of melano- Dermal neurofibroma bromin has a predicted molecular weight
cytes. Cate-au-lait spots are often the Localized intraneural neurofibroma of 320 kDa but runs in western blots at
first manifestation of NF1 in the newborn. Plexiform neurofibroma 220-250 kDa, most likely due to protein
Their number and size increase during in- Gliomas folding in denaturing gels {906). Although
fancy, but may remain stable or even de- Pilocytic astrocytoma, especially in the optic neurofibromin is expressed almost ubiq-
crease in adulthood. Histopathologically, pathway (optic glioma) uitously in most mammalian tissues, the
Diffuse astrocytoma
the ratio of melanocytes to keratinocytes highest levels have been found in the
Anaplastic astrocytoma
is higher in the unaffected skin of patients Glioblastoma
CNS and peripheral nervous system and
with NF1, and this is more marked in the in the adrenal gland {911).
Sarcomas and stromal tumours
cafe-au-lait spots {738). Melanocytes in
Malignant peripheral nerve sheath tumour (including
cafe-au-lait spots have been shown to malignant triton tumour) Gene function
harbour somatic NF1 mutations in ad- Rhabdomyosarcoma Neurofibromin harbours a small central
dition to the germline mutation {552). Gastrointestinal stromal tumour GAP-related domain, and thus belongs
Axillary and/or inguinal freckling occurs Neuroendocrine/neuroectodermal tumours
to the group of mammalian RAS-GAPs.
in the vast majority of patients with NF1 Phaeochromocytoma GAPs strongly accelerate the intrinsic
{2476). The histopathological features of Carcinoid tumour GTPase activity of RAS, thereby promot-
these freckles are indistinguishable from Medullary thyroid carcinoma ing the conversion of active GTP-bound
those of cate-au-lait spots. Lisch nodules C-cell hyperplasia RAS to the inactive GDP-bound form
are small, elevated, pigmented hamarto- Haematopoietic tumours {234). In vitro, neurofibromin acts as the
mas on the surface of the iris. The pres- Juvenile chronic myeloid leukaemia GAP for the classic RAS proteins (HRAS,
ence of Lisch nodules is a particularly Juvenile xanthogranuloma NRAS, and KRAS) and related subfam-
useful diagnostic criterion, because they Other features ily members (RRAS, RRAS2, and MRAS
occur in nearly all adults with NF1 {1544) Osseous lesions
{1821).
Scoliosis Of the several potential effector path-
Osseous and vascular lesions Short stature ways of active RAS proteins, the best-
In NF1, the orbits are often affected by Macrocephaly studied and probably most important in
sphenoid wing dysplasia. In addition, Pseudarthrosis the context of NF1 are the MAPK and the
spinal deformities often result in severe Sphenoid wing dysplasia RAS/Pl3K/AKT/mTOR pathways {910).
scoliosis, which may require surgical Eyes Both pathways have numerous and often
intervention. Thinning, bending, and Lisch nodules synergistic effects in tumour cells, regu-
pseudarthrosis may affect the long bones Nervous system lating proliferation, differentiation, migra-
(predominantly the tibia). Osteopenia/ Learning disabilities and attention deficit tion, apoptosis, and angiogenesis {1620).
osteoporosis and short stature may also hyperactivity disorder There is also some evidence of growth
be a component of NF1 {628,683). Fibro- Epilepsy regulatory functions outside of the neu-
Peripheral neuropathy
muscular dysplasia of the renal and other rofibromin GAP-related domain. Mice
Hydrocephalus (aqueductal stenosis)
arteries, cerebral aneurysm, and stenosis with a constitutional homozygous Nf1
of the internal carotid, or cerebral arteries Vascular lesions knockout (Nf1-/-) die in utero at day 13.5
Fibromuscular dysplasia/hyperplasia of renal artery
have also been reported {740,1815). of embryogenesis, due to abnormal car-
and other arteries
diac development {261,1110). Isolated
Tumours Skin reconstitution of the GAP-related domain
Cafe-au-lait spots
Patients with NF1 have increased risks of in these Nf1-/- mice rescues cardiovas-
Freckling (axillary and/or inguinal)
developing rhabdomyosarcomas, juve- cular development, but is insufficient to
nile chronic myeloid leukaemia, juvenile inhibit overgrowth of neural crest-de-
xanthogranulomas, gastrointestinal stro- rived tissues, leading to perinatal lethal-
mal tumours, duodenal carcinoids, C-cell OMG. There are 12 non-processed NF1 ity {1103).Neurofibromin also controls
hyperplasia I medullary thyroid carcino- pseudogenes localized on eight chromo- adenylyl cyclase activity and intracellular
mas, other carcinomas, and phaeochro- somes. None of these pseudogenes ex- cAMP levels {529,2568) The mechanism
mocytomas {2879). tends beyond exon 29. for neurofibromin-mediated cAMP regu-
lation remains unclear, but both RAS-de-
Gene structure Gene expression pendent and RAS-independent models
The NF1 locus is on chromosome 17q11.2 The NF1 transcript is approximately have been reported {289,940).
{2315) The NF1 gene is large, contain- 13 kb long and includes three alterna-
ing 59 exons and spanning about 350 kb tively spliced isoforms (exons 9a, 23a, Genotype/phenotype
{2194). One of the two extensive intrans, and 48a), which are variably expressed Genotype-phenotype correlations are
27b, includes coding sequences for three depending on tissue type and differentia- complicated by the unusually high de-
embedded genes that are transcribed in tion {2194). The product of the gene, neu- gree of variable expressivity within fami-
a reverse direction: EV/2A, EV/28, and rofibromin, is a cytoplasmic protein that lies with NF1. The correlation between

Neurofibromatosis type 1 295


clinical manifestations and the degree of 30
relatedness of patients suggests a role D Meet NIH NF1
for modifying non-allelic genes 12477) criteria (100%)
Only two clear genotype-phenotype cor-
relations have been established to date.
About 5-10% of patients have the NF1
25
• Cate-au-lait SPots
(100%)
microdeletion syndrome, caused by un- 20 D Inguinal/axillary
equal homologous recombination of NF1 freckling (95%)
(/)
repeats resulting in the loss of approxi- .....
ro D 2 neurofibromas or
mately 1.5 Mb of DNA on 17q, including Q) 15
the entire NF1 gene and 13 surrounding >- 1 plexiform (85%)
genes {549). Patients with this syndrome 2 or more lisch
tend to have a more severe phenotype, 10
nodules (85%)
including facial dysmorphism, mental
retardation, developmental delay, in-
5 • Optic pathway
glioma (15%)


creased burden of neurofibromas, and
increased risk of MPNST development MPNST{10%)
{550). suggesting involvement of addi- 0
tional genes 11899). SUZ12 is one of the
codeleted genes in patients with NF1 NF1 Manifestation
microdeletion syndrome, and loss of this Fig. 16.04 Mean ages of onset for common clinical manifestations in patients with neurofibromatosis type 1 (NF1 ). The
gene has been shown to play an impor- estimated frequencies for each manifestation within the patient population are given in parentheses. MPNST, malignant
tant role in MPNST development 1548). peripheral nerve sheath tumour; NIH, National Institutes of Health.
A specific 3 bp in-frame deletion in exon
17 of the NF1 gene (c.2970-2972 de-
lAAT) is associated with the absence of as plexiform neurofibromas or optic path- have a sporadic, de novo NF1 mutation.
cutaneous neurofibromas and clinically way gliomas. For each offspring of a person with NF1,
obvious plexiform tumours 12600}. the chance of inheriting the pathogenic
There are several additional reports sug- Spinal neuroflbromatosis NF1 gene is 50%. The disease pen-
gesting potential less-established geno- Spinal neurofibromatosis is a condition etrance is 100%. Prenatal and preim-
type-phenotype correlations. NF1 muta- in which multiple bilateral spinal neurofi- plantation mutation testing are available.
tions in patients with optic pathway glioma bromas occur, but there are few or no Genetic testing can be diagnostic in
appear to cluster at the 5' end of the gene cutaneous manifestations of NF1. Mis- children who fulfil some but not all of the
encompassing exons 1-15 {235,2332}. sense mutations are found more often National Institutes of Health (NIH) criteria,
The heterozygous c.5425C->T missense in patients with spinal neurofibromatosis and may be helpful in certain cases for
variant (p.Arg1809Cys) has been associ- than in those with a classic NF1 pheno- distinguishing NF1 phenotypes from oth-
ated with a mild phenotype with multiple type, but no clear genotype-phenotype er conditions, such as Legius syndrome.
cate-au-lait spots and skinfold freckling association has been established 12197). Mutation screening of the NF1 gene is
only {1978). NF1 splice-site mutations Both familial and sporadic cases occur difficult due to its large size, the pres-
have been putatively associated with an 1326,1897,1985,2601 ). ence of pseudogenes, and the diversity
increased tendency to develop MPNSTs of mutations 11652). More than 1000 dif-
and gliomas 154). A 1 A Mb microduplica- Differential diagnosis ferent mutations have been reported at
tion encompassing the NF1 gene is re- NF1 belongs to a heterogeneous group [https://grenada.lumc.nl/LOVD2/mende-
ported not to be associated with a typical of developmental syndromes known as lian_genes/home.php?select_db=NF1]
NF1 phenotype, but with learning difficul- RASopathies {2073). All RASopathies are and [http://www.hgmd.org]. Using com-
ties and dysmorphic features 11698) caused by germline mutations in genes prehensive screening techniques that
encoding for members or regulators of may include various strategies such as
Mosaicism the RAS/MAPK pathway. RASopathies long-range RT-PCR, protein truncation
The occurrence of a segmental or region- also show some phenotypic overlap; for testing, cDNA sequencing, FISH, next-
al form of NF1, caused by somatic mo- example, NF1 without neurofibromas and generation sequencing, and/or multiplex
saicism at the NF1 gene locus, further ex- Legius syndrome (caused by SPRE01 ligation-dependent probe amplification,
tends the range of variability {1242,1517) mutation) may be clinically indistinguish- as many as 95% of mutations may be
Patients with this form of NF1 often have able 1275). detected in individuals fulfilling the NIH
only pigmentary manifestations within the criteria for NF1 11601,1652). More than
affected limb or region; however, some Genetic counselling 80% of mutations are predicted to either
patients develop classic tumours, such NF1 is inherited in an autosomal domi- encode a truncated protein or result in no
nant manner, but about half of all patients protein production.

296 Familial tumour syndromes


Neurofibromatosis type 2 Stemmer-Rachamimov A.O.
Wiestler OD.
Louis D.N.

Definition
An autosomal dominant disorder char-
acterized by neoplastic and dysplastic
lesions that primarily affect the nervous
system, with bilateral vestibular schwan-
nomas as a diagnostic hallmark. Other
manifestations include schwannomas of
other cranial nerves, spinal and periph-
eral nerves, and the skin; intracranial
and spinal meningiomas; gliomas, in
particular spinal ependymomas; and a
variety of non-tumoural and dysplastic/
developmental lesions, including menin- Fig. 16.05 T1-weighted, contrast-enhanced MRI from a patient with neurofibromatosis type 2. A Bilateral vestibular
gioangiomatosis, glial hamartomas, ocu- schwannomas (arrowheads), the diagnostic hallmark of neurofibromatosis type 2. B Multiple meningiomas presenting
lar abnormalities (e.g. posterior subcap- as contrast-enhanced masses.
sular cataracts, retinal hamartomas, and
epiretinal membranes), and neuropa- Diagnostic criteria Confirmatory testing for NF2 mutations
thies. NF2 is caused by mutations of the The diagnosis of NF2 is based on clini- may be helpful when a patient does not
NF2 gene on chromosome 22q12. cal features and may be challenging be- meet the clinical criteria for a definite di-
cause of the wide variability of symptoms agnosis but the phenotype is suggestive.
OMIM number (1624l 101000 and time of onset. Particularly difficult to
diagnose are genetic mosaics (account- Nervous system neoplasms
Incidence/epidemiology ing for 30% of sporadic cases), in which
The disorder affects between 1 in 25 000 segmental involvement or milder disease Schwannomas
and 1 in 40 000 individuals (662). About may occur (1307l, and paediatric cases Schwannomas associated with NF2
half of all cases are sporadic, occurring in which the full manifestation of the dis- are WHO grade I tumours composed
in individuals with no family history of NF2 ease has not yet developed. The distinc- of neoplastic Schwann cells, but differ-
and caused by newly acquired germline tion from other forms of neurofibromatosis ing from sporadic schwannomas in sev-
mutations. In the past, the considerable (neurofibromatosis type 1 and schwanno- eral ways. NF2 schwannomas present
variability of the clinical manifestations matosis) is difficult in some cases. There in younger patients (in the third decade
of NF2 resulted in underdiagnosis of the is clinical phenotypic overlap between of life) than do sporadic tumours (in the
syndrome. NF2 mosaic, early NF2, and schwanno- sixth decade), and many patients with
matosis; some cases that fulfil the clinical NF2 develop the diagnostic hallmark of
diagnostic criteria for schwannomatosis the disease, bilateral vestibular schwan-
have later proven to be NF2 (1990l. nomas, by their fourth decade of life
Table 16.03 National Institutes of Health (NIH) I
Manchester criteria for neurofibromatosis type 2 (NF2) The original clinical diagnostic criteria {659,1599l. NF2 vestibular schwanno-
(655A} for NF2 were established at the National mas may entrap seventh cranial nerve
The presence of one or more of the following Institutes of Health (NIH) Consensus De- fibres {1198l and have higher proliferative
features is diagnostic: velopment Conference on Neurofibroma- activity {75L although these features do
tosis in 1987 (1758l. Several revisions of
Bilateral vestibular schwannomas
these criteria have since been proposed:
A first-degree relative with NF2 AND unilateral the NIH 1991 criteria, the Manchester
vestibular schwannoma OR any two of: meningioma,
criteria (see Table 16.03), the National
schwannoma, glioma, neurofibroma, posterior
subcapsular lenticular opacities*
Neurofibromatosis Foundation (NNFF)
criteria, and the Baser criteria. Each of
Unilateral vestibular schwannoma AND any two of:
these revisions expanded the original cri-
meningioma, schwannoma, glioma, neurofibroma,
posterior subcapsular lenticular opacities*
teria, aiming to also identify patients with
multiple NF2 features who do not present
Multiple meningiomas AND unilateral vestibular
with bilateral vestibular schwannomas
schwannoma OR any two of: schwannoma, glioma,
neurofibroma, cataract*
and have no family history of NF2 (137,
659,905l. Fig. 16.06 Bilateral vestibular schwannomas, diagnostic
*Any two of::: two individual tumours or cataracts.
for neurofibromatosis type 2.

Neurofibromatosis type 2 297


.,

not necessarily connote more aggressive


behaviour. In addition to the vestibular
division of the eighth cranial nerve, other
sensory nerves may be affected, includ-
ing the fifth cranial nerve and spinal dor-
sal roots. However, motor nerves such
as the twelfth cranial nerve may also be
involved {659,1536). Cutaneous schwan-
nomas are common and may be plexi-
form {659,1599).
NF2 schwannomas may have a multilob-
ular (cluster-of-grapes) appearance on
both gross and microscopic examination
{2738). and multiple schwannomatous
tumourlets may develop along individual
nerves, particularly on spinal roots {1536,
2424). A mosaic pattern of immunostain-
ing for SMARCB1 expression (indicating
patchy loss) has been reported in most
syndrome-associated schwannomas, in-
cluding both NF2 and schwannomatosis
{1909).
,
Meningiomas Fig. 16.08 Cerebral microhamartomas in a patient with neurofibromatosis type 2. A These lesions are scattered
Multiple meningiomas are the second throughout the cortex and (B) show strong immunoreactivity for 8100.
hallmark of NF2 and occur in half of all
patients with the disorder (1536). NF2-
associated meningiomas occur earlier in
life than sporadic meningiomas, and may
be the presenting feature of the disorder,
especially in the paediatric population
(656,659,1599). Although most NF2-as-
sociated meningiomas are WHO grade I
tumours, several studies have suggested

Fig. 16.09 A Multiple schwannomas of spinal roots. B Schwannomas in neurofibromatosis type 2 often show a distinct
nodular pattern.

that NF2-associated meningiomas have masses {2158,2204) Diffuse and pilocyt-


a higher mitotic index than sporadic men- ic astrocytomas have been reported in
ingiomas (74,1942). All major subtypes of NF2, but many probably constitute misdi-
meningioma can occur in patients with agnosed tanycytic ependymomas (923).
NF2, but the most common subtype is
fibroblastic (74,1536). NF2-associated Neuroftbromas
rneninqiornas can occur throughout the Cutaneous neurofibromas have been re-
cranial and spinal meninges, and may af- ported in NF2. However, on histological
fect sites such as the cerebral ventricles. review, many such neurofibromas prove
to be schwannomas, including plexiform
Gliomas schwannomas misdiagnosed as plexi-
Approximately 80% of gliomas in patients form neurofibromas.
with NF2 are spinal intramedullary or
cauda equina tumours, with an additional Other nervous system lesions
10% of gliomas occurring in the medulla
(2158). Ependymomas account for most Schwannosis is a proliferation of
of the histologically diagnosed gliomas Schwann cells, sometimes with entan-
in NF2, and for almost all spinal gliomas gled axons. but without frank tumour
(2158,2204) In most cases, NF2 spinal formation. In patients with NF2, schwan-
Fig. 16.07 Numerous schwannomas of the cauda ependymomas are multiple, intramed- nosis is often found in the spinal dorsal
equina in a patient with neurofibromatosis type 2. ullary, slow-growing, asymptomatic root entry zones (sometimes associated

298 Familial tumour syndromes


with a schwannoma of the dorsal root) or glial hamartias suggests the possibil- Gene function). lsoform 2, encoded by
in the perivascular spaces of the central ity that haploinsufficiency during devel- exons 1-16, exists only in an unfolded
spinal cord, where the nodules look more opment underlies these malformations state {901,28021
like small traumatic neuromas 12195, 124221
2204). Less robust but otherwise identi- Gene mutations
cal schwannosis has been reported in lntracranial calcifications are frequently Numerous germline and somatic NF2
reactive conditions. noted in neuroimaging studies of patients mutations have been detected, support-
with NF2. The most common locations ing the hypothesis that NF2 functions as
Meningioangiomatosis is a cortical lesion are the cerebral and cerebellar cortices, a tumour suppressor gene {900,1536)
characterized by a plaque-like prolifera- periventricular areas, and choroid plexus. Germline NF2 mutations differ somewhat
tion of meningothelial and fibroblast-like from the somatic mutations identified in
cells surrounding small vessels. It occurs Peripheral neuropathies not related to tu- sporadic schwannomas and meningi-
both sporadically and in NF2. Menin- mour mass are increasingly recognized ornas. The most frequent germline muta-
gioangiomatosis is usually a single, in- as a common feature of NF2 1500,1536}. tions are point mutations that alter splice
tracortical lesion, although multifocal ex- Mononeuropathies may be the present- junctions or create new stop codons
amples occur as well, as do non-cortical ing symptom in children 16561, whereas {252, 1536,1556,1647, 2190,2221,2575).
lesions {2195,22041 Meningioangioma- progressive polyneuropathies are more Germline mutations are found in all parts
tosis may be predominantly vascular (re- common in adults. Sural nerve biopsies of the gene (with the exception of the al-
sembling a vascular malformation) or pre- from patients with NF2 suggest that NF2 ternatively spliced exons), but they occur
dominantly meningothelial, sometimes neuropathies are mostly axonal and may preferentially in exons 1-8 {16471 One
with an associated meningioma. Spo- be secondary to focal nerve compres- possible hotspot for mutations is posi-
radic meningioangiomatosis is a single sion by tumourlets or onion-bulb-like tion 169 in exon 2, in which a C-->T tran-
lesion that usually occurs in young adults Schwann cell or perineurial cell prolif- sition at a CpG dinucleotide results in a
or children, who present with seizures or erations without associated axons {2405, stop at codon 57 {252,1647); other CpG
persistent headaches. In contrast, NF2- 25461 dinucleotides are also common targets
associated meningioangiomatosis may for C-->T transitions {2221 I
be multifocal and is often asymptomatic Extraneural manifestations can also oc-
and diagnosed only at autopsy {2423). cur. Posterior lens opacities are common Gene expression
and highly characteristic of NF2. A variety The NF2 gene is expressed in most
Glial hamartias (also called microham- of retinal abnormalities (including hamar- normal human tissues, including brain
artomas) of the cerebral cortex are cir- tomas, tufts, and dysplasias) may also be {2190,2575).
cumscribed clusters of cells with medium found {4031 Ocular abnormalities may
to large atypical nuclei. These lesions be helpful in the diagnosis of paediatric Gene function
are scattered throughout the cortex and patients. Skin lesions other than cutane- The predicted protein product shows
basal ganglia and show strong immuno- ous nerve sheath tumours, primarily ca- a strong similarity with the highly con-
reactivity for S100, but are only focally fe-au-lait spots, have been reported. served protein 4.1 family of cytoskeleton-
positive for GFAP. Glial hamartias are associated proteins, which includes pro-
common in and pathognomonic of NF2 Gene structure tein 4.1, talin, moesin, ezrin, radixin, and
{2195,2752). and are not associated with The NF2gene {2190,2575) spans 110 kb protein tyrosine phosphatases. The simi-
mental retardation or astrocytomas. The and consists of 17 exons. NF2 mRNA larity of the NF2-encoded protein to the
hamartias are usually intracortical, with a transcripts encode at least two major ERM proteins (moesin, ezrin, and radixin)
predilection for the molecular and deep- protein forms generated by alternative resulted in the name "merlin" {2575); the
er cortical layers, but have also been splicing at the C-terminus. lsoform 1, en- alternative name "schwannomin" has
observed in the basal ganglia, thalamus, coded by exons 1-15 and 17, has intra- also been suggested {21901 Members of
cerebellum, and spinal cord 127521 The molecular interactions similar to the ERM the protein 4.1 family link the cell mem-
fact that merlin expression is retained in proteins - ezrin, radixin, and moesin (see brane to the actin cytoskeleton. These

q��.·�
i;..-....:.::,•'-"-U't."'·i·t���· � �. .
Fig. 16.10 A Meningioangiomatosis associated with neurofibromatosis type 2. An intracortical lesion composed of a perivascular proliferation of cells (predominantly meningothelial)
in Virchow-Robin spaces. B Diffuse cortical meningioangiomatosis associated with neurofibromatosis type 2. Trichrome stain. C Multiple Schwann cell tumourlets arising in the
cauda equina of a patient with neurofibromatosis type 2 (Luxol fast blue, H&E).

Neurofibromatosis type 2 299


WNT/beta-catenin pathways. Many mer-
Extracellular matrix lin binding partners have been identified
including integrins and tyrosine recepto�
kinases (1288,1726). Recent data sug-
Growth factor receptor gest that merlin also suppresses signal-
ling at the nucleus, where it suppresses
the E3 ubiquitin ligase IL17RB (1493).

Genotype/phenotype
The clinical course in patients with NF2
varies widely between and (to a lesser
Inactive merlin extent) within families (659,1599). Some
I I Rae/Pak families feature early onset with diverse
p p NHERF tumours and high tumour load (Wishart
type), whereas others present later, with
only vestibular schwannomas (Gardner

.
type). An effect of maternal inheritance
on severity has been noted, as have fami-
lies with genetic anticipation. All families
with NF2 show linkage of the disease to
Cytoskeleton Cell proliferation chromosome 22 (1757), implying a single
responsible gene. Correlations of geno-
Fig. 16.11 In its active {hypophosphorylated) state, merlin suppresses cell proliferation and motility by inhibiting
type with phenotype have therefore been
the transmission of growth signals from the extracellular environment to the Rae/PAK signalling system. Inactivated
{phosphorylated) merlin dissociates from its protein scaffold, thus disinhibiting Rae/PAK signalling as well as cell used to attempt to predict clinical course
proliferation and motility. on the basis of the type of the underlying
NF2 mutation. Nonsense and frameshift
mutations are often associated with a
proteins consist of a globular N-terminal molecule, which is inhibited by phospho- more severe phenotype, whereas mis-
FERM domain, an alpha-helical domain rylation of the C-terminus on serine resi- sense mutations, large deletions, and
containing a praline-rich region, and a dues (2339). Although the precise mech- somatic mosaicism have been associ-
C-terminal domain. The N-terminal do- anism of tumour suppression by merlin ated with milder disease (138,664,1308,
main interacts with cell membrane pro- is still unknown, the structural similarity 1647). Phenotypic variability is observed
teins such as CD44, CD43, ICAM1, and of merlin to the ERM proteins suggests in splice-site mutations, with more severe
ICAM2; the C-terminal domain contains that merlin provides regulated linkage phenotypes observed in mutations up-
the actin-binding site. Merlin lacks the between membrane-associated proteins stream from exon 7 (1306).
actin-binding site in the C-terminus but and the actin cytoskeleton; the tumour
may have an alternative actin-binding suppressor activity is thought to be ex- Genetic counselling
site (2802}. The ERM proteins and mer- erted by regulation of signal transmission The risk of transmission to offspring is
lin may be self-regulated by head-to-tail from the extracellular environment to the 50%. Prenatal diagnosis by mutation
intramolecular associations that result in cell (1619) and activation of downstream analysis and testing of children of pa-
folded and unfolded states. The folded pathways including the MAPK, FAK/SRC, tients with NF2 is possible when the mu-
state of merlin is the functionally active Pl3K/AKT, Rac/PAK/JNK, mTORC1, and tation is known.

300 Familial tumour syndromes


Schwan nomatosis Stemmer-Rachamimov A.O.
Hulsebos T.J.M.
Wesseling P.

Table 16.04 The current proposed clinical and molecular


Definition mosaic immunohistochemical staining
diagnostic criteria for schwannomatosis {1990)
A usually sporadic and sometimes au- for SMARCB1 protein (i.e. an intimate
Molecular criteria for definite schwannomatosis:
tosomal dominant disorder character- mixture of positive and negative tumour
ized by multiple schwannomas (spinal, Two or more schwannomas or meningiomas cell nuclei), but with considerable inter-
cutaneous, and cranial) and multiple (pathology proven) AND genetic studies of at least and intratumoural heterogeneity (with
two tumours with LOH for chromosome 22 and two
meningiomas (cranial and spinal), asso- < 10% to> 50% immunonegative nuclei).
different NF2 mutations OR
ciated with inactivation of the NF2 gene Mosaic SMARCB1 staining is also often
in tumours but not in the germline, and One schwannoma or meningioma (pathology present in schwannomas of patients with
proven) AND SMARCB1 germline mutation
caused by mutations in SMARCB1 on sporadic schwannomatosis and NF2,
22q or LZTR1 on 22q. Clinical criteria for definite schwannomatosis: corroborating an interaction between
For a diagnosis of schwannomatosis, it Two or more schwannomas (non-dermal, one NF2 and SMARCB1 in the pathogenesis
is important to exclude the other forms pathology proven) AND no bilateral vestibular of these tumours {1064,1909). In contrast,
of neurofibromatosis by confirming the schwannomas (by thin-slice MRI) OR sporadic schwannomas rarely show mo-
absence of vestibular schwannomas on One schwannoma or meningioma (pathology saic SMARCB1 staining.
MRI and the absence of other manifesta- proven) AND first-degree relative affected by
tions of neurofibromatosis type 2 (NF2) or schwannomatosis Meningiomas
neurofibromatosis type 1. Clinical criteria for possible schwannomatosis: Various studies have shown that
SMARCB1 germline mutations also pre-
Two or more schwannomas (no pathology) OR
OMIM number {1624l 162091 dispose individuals to the development
Severe chronic pain associated with a schwannoma of multiple meningiomas, with preferen-
Synonyms tial location of cranial meningiomas at the
The terms used to describe this disorder schwannomas may develop only later in falx cerebri {102,455,2622). The reported
in the past include "neurilemmomatosis", the course of the disease {661 }. There proportion of schwannomatosis patients
"multiple schwannomas", and "multiple can be overlap between the clinical fea- who develop a meningioma is 5% {2375).
neurilemmomas". tures of early NF2 or NF2 mosaics and Occasionally, patients present with multi-
schwannomatosis {1990l. ple meningiomas.
Incidence/epidemiology
In several reports, schwannomatosis was Nervous system neoplasms Extraneural manifestations
found to be almost as common as NF2, Extraneural manifestations associated
with an estimated annual incidence of Schwannomas with schwannomatosis are rare, unlike
1 case per 40 000-80 000 population Patients with schwannomatosis typically those associated with neurofibromato-
{76,2322l Familial schwannomatosis have multiple schwannomas. These tu- sis types 1 and 2. One study reported
accounts for only 10-15% of all cases mours may develop in spinal roots, crani-
{1558,2322l. al nerves, skin, and (occasionally) unilat-
erally in the vestibular nerve. Cutaneous
Diagnostic criteria schwannomas may be plexiform. The
Reports of patients with multiple non- tumours have a segmental distribution
vestibular schwannomas date back to in about 30% ef patients with schwan-
1984 {2350l, but it was long debated nomatosis {1555,1649,2377,2378}. Se-
whether the condition constitutes a vere pain associated with the tumours
form of attenuated NF2 or a separate is characteristic of the disease. This is a
entity. Standardized clinical diagnostic distinguishing feature from NF2, in which
criteria for schwannomatosis were first pain is rare and neurological deficits and
developed by a panel of experts in a polyneuropathy are common {1649). His-
consensus meeting in 2005 {1555l and tologically, schwannomatosis tumours
later revised in 2012 to include molecu- may display prominent myxoid stroma
lar diagnosis {1990l. The exclusion of and an intraneural growth pattern, and
NF2 by clinical criteria and by imaging of are sometimes misdiagnosed as neu-
the vestibular nerves is essential for the rofibromas or malignant peripheral nerve
diagnosis of schwannomatosis. The dis- sheath tumours {1649). Fig. 16.12 Coronal MRI (short T1 inversion recovery
tinction may be particularly challenging Many schwannomas of patients sequence) showing multiple, bright, discrete tumours in
in paediatric patients, because vestibular with familial schwannomatosis show a patient with schwannomatosis.

Schwannomatosis 301
,

protein, resulting in GO/G1 cell cycle ar-


rest {185,2648).
According to the tumour suppressor gene
model, both copies of the SMARCB1
gene are inactivated in these tumours. In
schwannomas of patients with schwan-
nomatosis with SMARCB1 germline mu-
tation in addition to loss of the second
SMARCB1 allele, there is also inactivation
of both copies (by mutation and deletion)
of the NF2 gene, located 6 Mb distal to
SMARCB1 on chromosome 22 {260,921,
2325). The deletion of SMARCB1 and
NF2 is a consequence of the loss of one
copy of chromosome 22 {922).
Based on these observations, a four-hit,
Fig. 16.13 Schwannomas in schwannomatosis are often myxoid, with an appearance that mimics that of neurofibromas.
three-step model of tumorigenesis in
schwannomatosis is proposed: (inher-
a uterine leiomyoma in a patient with to be elucidated, and other families af- ited) SMARCB1 germline mutation oc-
schwannomatosis; the tumour had a fected by schwannomatosis with C006 curs (hit 1), followed by loss of the other
molecular profile similar to that of the involvement have not yet been reported. chromosome 22 with the wildtype copy
schwannomas, as well as the corre- Germline mutations of the NF2 gene of SMARCB1 and one copy of NF2(hits 2
sponding mosaic staining for SMARCB1 have been excluded in schwannomato- and 3), followed by a somatic mutation of
protein, indicating that the SMARCB1 sis {1115,1237,1557). but the presence of the remaining copy of the NF2 gene (hit
defect in schwannomatosis may occa- other, somatically acquired mutations in 4). This model, with somatic mutations of
sionally contribute to the oncogenesis of the NF2 gene is characteristic in schwan- the NF2 gene as the last step, explains
extraneural neoplasms {1062}. nomatosis-associated schwannomas. the observation of different NF2 muta-
One third of all patients with sporadic tions in multiple schwannomas in a sin-
Inheritance and genetic heterogeneity schwannomatosis have segmental dis- gle patient with schwannomatosis {1990).
The great majority of schwannomatosis tribution of their tumours, suggesting so- This four-hit model of genetic events also
cases are sporadic, with only 15% of matic mosaicism for the causative gene, occurs in meningiomas in patients with
patients having a positive family history. but this has not yet been demonstrated SMARCB1 germline mutation {102,455,
In the familial form, the disease displays for SMARCB1 or LZTR1. 2622).
an autosomal dominant pattern of inherit- In schwannomatosis, most germline mu-
ance, with incomplete penetrance {1557) The SMARCB 1 gene tations in SMARCB1 are non-truncating
In 2007, the SMARCB1 gene on chromo- missense mutations, splice-site muta-
some arm 22q was identified as a famil- Gene structure and expression tions, or in-frame deletions, which are
ial schwannomatosis-predisposing gene The SMARCB1 gene is located in chro- predicted to result in the synthesis of
{1064). In subsequent studies, the gene mosome region 22q11.23 and contains an altered SMARCB1 protein with modi-
proved to be involved in about 50% of nine exons spanning 50 kb of genomic fied activity {2380} The mosaic staining
familial cases, but in � 10% of sporadic DNA {2649). Alternative splicing of exon 2 pattern seen in many schwannomatosis-
cases {260,921,2191,2380}. results in two transcripts and two proteins associated schwannomas suggests the
In 2014, the LZTR1 gene, also on 22q, with lengths of 385 and 376 amino acid absence of SMARCB1 protein in part of
was identified as a second causative residues, respectively. The so-called the tumour cells {1909). Truncating non-
gene in schwannomatosis {1980). In SNF5 homology domain in the second sense and frameshift mutations, also re-
patients with schwannomatosis without half of the protein harbours highly con- ported in patients with schwannomatosis,
SMARCB1 germline mutations, LZTR1 served structural motifs through which generate a premature termination codon
mutations were found in about 40% of fa- SMARCB1 interacts with other proteins and are predicted to result in the ab-
milial and 25% of sporadic cases {1073, {2430). The SMARCB1 protein is a core sence of SMARCB1 protein expression.
1877, 2377) The fact that most schwan- subunit of mammalian SWI/SNF chroma- For the truncating mutations in exon 1 of
nomatosis cases cannot be explained by tin remodelling complexes, which regu- SMARCB1, it was recently demonstrated
the involvement of SMARCB1 or LZTR1 late the expression of many genes by us- that translational reinitiation at a down-
suggests the existence of additional ing ATP for sliding the nucleosomes along stream AUG codon occurs, resulting in
causative genes {1073). the DNA helix, facilitating or repressing the synthesis of an N-terminally truncated
Recently, a germline missense mutation transcription {2760). The SMARCB1 pro- SMARCB1 protein {1063}. Other mecha-
was identified in the C006 gene on chro- tein functions as a tumour suppressor nisms, such as alternative splicing, may
mosome arm 14q, which segregated with via repression of CCND1 gene expres- operate to overcome the deleterious ef-
the disease in a large family affected by sion, induction of the COKN2A gene, and fect of truncating mutations in the other
schwannomatosis {2856). However, the hypophosphorylation of retinoblastoma exons of SMARCB1.
oncogenic effect of the mutation has yet

302 Familial tumour syndromes


SMARCB 1 mutations in rhabdo1d Germline Somatic
tumours
SMARCB1 germline mutations may also
predispose individuals to the develop- I Schwannoma 1 I
ment of rhabdoid tumours (very aggres-
sive tumours of childhood). a disorder /Hit2
called rhabdoid tumour predisposition
Loss 22 m NF2 mutation m
syndrome 1 (p. 321). In rhabdoid tu- Hit 1
mours, the two copies of the SMARCB1
gene are inactivated by a truncating mu-
\
\
+ ..
ml

tation and deletion of the wildtype gene, Hit 3 Hit 4


SMARCBl m +
resulting in total loss of SMARCB1 ex-
pression in tumour cells. This is in con- NF2 + +
trast to the presence of non-truncating I Schwannoma 2 i
SMARCB1 mutations and the mosaic
SMARCB1 expression in the schwanno-
mas of patients with schwannomatosis. m
Loss 22 m NF2 mutation
Because children with a rhabdoid tumour
usually die before the age of 3 years. + m2
familial inheritance of the predisposition
is extremely rare, and most cases are
sporadic (2327,2522). However, 35% of Fig. 16.14 The four-hit mechanism for the formation of tumours in schwannomatosis.
patients with sporadic rhabdoid tumour
carry a germline SMARCB1 alteration as
the first hit {249,616). A few families have are also found in the schwannomas of antibody demonstrates absent or re-
been reported in which the affected in- patients with schwannomatosis with a duced expression of the protein, consist-
dividuals inherited a SMARCB1 mutation germline LZTR1 mutation, suggesting ent with its function as a tumour suppres-
and developed schwannomatosis or a that the four-hit, three-step model of tu- sor {1877). Germline LZTR1 mutations
rhabdoid tumour (371,616,24721 Howev- morigenesis also applies to these tu- (but no germline NF2 mutations) were
er, the schwannomas in these families (as mours (1073,1877,1980,2377}. However, also found in 3 of 39 patients with a unilat-
well as the rhabdoid tumours) displayed unlike in SMARCB1-associated schwan- eral vestibular schwannoma and at least
total loss of SMARCB1 protein expres- nomas (in which LOH for chromosome one other schwannoma. suggesting that
sion {371,24721. 22 occurs by loss of chromosome). in unilateral vestibular schwannoma may be
LZTR1-associated schwannomas, mitot- present in schwannomatosis, especially
The LZTR1 gene ic recombination has been found in 30% in cases with a LZTR1 germline mutation
of cases (2377). (23771.
Gene structure and expression
The LZTR1 gene is situated proximal Gene mutations Genetic counselling
to SMARCB1 in chromosome region The reported LZTR1 germline mutations The risk of transmission to offspring is
22q11.21 and contains 21 exons. span- in schwannomatosis include non-trun- assumed to be 50% for patients carry-
ning 17 kb of genomic DNA (1980). The cating (missense and splice-site) as well ing a germline mutation of SMARCB1
wildtype gene codes for a protein with as truncating (nonsense and frameshift) or LZTR1 and in familial cases in which
a length of 840 amino acid residues. mutations and are found along the entire the germline is unidentified. The risk of
LZTR1 may function as a substrate adap- coding sequence of the gene, affecting transmission to the offspring of patients
tor in cullin-3 ubiquitin ligase complexes, the functionally important domains of the with sporadic cases with no SMARCB1
binding to cullin-3 and to substrates tar- LZTR1 protein (1073,1877,1980,2377}. or LZTR1 mutation is unknown {1990}.
geted for ubiquitination (735). Somatical- lmmunostaining of LZTR1-associated
ly acquired NF2 mutations and deletions schwannomas with an LZTR1-specific

Schwannomatosis 303
Von Hippel-Lindau disease Plate K.H.
Vortmeyer A.O.
Zagzag D.
Neumann H.P.H.
Aldape K.D.

Definition Table 16.06 Sites of involvement in von Hippel-Lindau patient age of 25 years), and thus offer
An autosomal dominant disorder charac- disease the possibility of an early diagnosis
terized by the development of clear cell Non·
Organ/
renal cell carcinoma (RCC), capillary hae- Tumours neoplastic CNS
tissue
mangioblastoma of the CNS and retina, lesions CNS haemangioblastomas develop
phaeochromocytoma, and pancreatic CNS Haemangioblastoma mainly in young adults (at a mean patient
and inner ear tumours. Von Hippel-Lin- Eye (retina) Haemangioblastoma age of 29 years). They are predominantly
dau disease (VHL) is caused by germline located in the cerebellum, followed by
Clear cell renal cell
mutations of the VHL tumour suppressor Kidney
carcinoma
Cysts the brain stem and spinal cord. Approxi-
gene, located on chromosome 3p25-26. mately 25% of all cases are associated
The von Hippel-Lindau disease tumour Adrenal with VHL.
Phaeochromocytoma
gland
suppressor protein (VHL protein) plays a
key role in cellular oxygen sensing. Neuroendocrine islet Adrenal gland
Pancreas Cysts
cell tumours Phaeochromocytomas may constitute a
OMIM number (1624l 193300 Inner ear
Endolymphatic sac major clinical challenge, particularly in
tumour families affected by VHL with predisposi-
Historical annotation Papillary tion to the development of these tumours.
Epididymis
Lindau described capillary haemangio- cystadenoma They are often associated with pancre-
blastoma and noted its association with atic cysts.
retinal vascular tumours (previously de-
scribed by von Hippel) and tumours of Sites of involvement Other extrarenal mamfestations
the visceral organs, including the kidney. Renal lesions in carriers of VHL germline Other extrarenal manifestations include
mutations are either cysts or clear cell neuroendocrine tumours, endolymphatic
Incidence/epidemiology RCCs. They are typically multifocal and sac tumours of the inner ear, and epididy-
VHL is estimated to have an annual in- bilateral. The mean patient age at mani- mal and broad ligament cystadenomas.
cidence rate of 1 affected individual per festation is 37 years (vs 61 years for spo-
36 000-45 500 population. radic clear cell RCC), with a patient age Gene structure
at onset of 16-67 years. There is a 70% The VHL tumour suppressor gene is lo-
Diagnostic criteria chance of developing clear cell RCC by cated at chromosome 3p25-26. It has
The clinical diagnosis of VHL is based the age of 70 years. Metastatic RCC is three exons and a coding sequence of
on the presence of capillary haemangio- the leading cause of death from VHL. 639 nucleotides. Germline mutations of
blastoma in the CNS or retina and the The median life expectancy of patients the VHL gene are spread over the three
presence of one of the typical VHL-asso- with VHL is 49 years. exons. Missense mutations are most
ciated extraneural tumours or a pertinent common, but nonsense mutations, mi-
family history. Germline VHL mutations Eye crodeletions/insertions, splice-site muta-
can virtually always be identified in VHL. Retinal haemangioblastomas manifest tions, and large deletions also occur. In
earlier than kidney cancer (at a mean accordance with the function of VHL as
a tumour suppressor gene, VHL gene

Table 16.05 Key characteristics of sporadic


haemangioblastoma and haemangioblastoma associated
with von Hippel-Lindau disease (VHL)
VHL·
Criterion Sporadic
associated
Female 41% 56%
Patient age 44 years (7-82) 23 years (7-64)
lntracranial 79% 73%
Spinal 11% 75%
Multiple 5% 65%
Fig. 16.15 Von Hippel-Lindau disease. A Bilateral adrenal phaeochromocytoma and (B) multiple pancreatic
neuroendocrine tumours.

304 Familial tumour syndromes


cellular responses to hypoxia) for ubiq-
uitination and proteasomal degradation.
The beta-domain of the VHL protein inter-
acts with HIF1A. Binding of the hydroxy-
lated subunit of the VHL protein causes
polyubiquitination and thereby targets
HIF1A for proteasome degradation. Un-
der hypoxic conditions or in the absence
of functional VHL, HIF1A accumulates
and activates the transcription of hypox-
ia-inducible genes, including VEGF-A, Fig. 16.17 Endolymphatic sac tumour. Papillary fronds
POGFB, TGFA, and EPO. Constitutive and colloid secretions.
overexpression of VEGF explains the ex-
traordinary capillary component of VHL-
Fig. 16.16 Retinal angioma in van Hippel-Lindau associated neoplasms. VEGF has been Invasion
disease. targeted as a novel therapeutic approach Only wildtype (not tumour-derived) VHL
using neutralizing anti-VEGF antibody. protein binds to fibronectin. As a result,
mutations are also common in sporadic Induction of EPO is responsible for the VHL-/- RCC cells show a defective as-
haemangioblastomas and RCCs. occasional paraneoplastic erythrocytosis sembly of an extracellular fibronectin
in patients with kidney cancer and CNS matrix. Through down-regulation of the
Gene expression haemangioblastoma. cellular response to hepatocyte growth
The VHL gene is expressed in a variety factor I scatter factor and reduced levels
of human tissues, in particular epithelial Cell cycle ext! of TIMP2, VHL protein-deficient tumour
skin cells; the gastrointestinal, respira- Recent studies in RCC cell lines suggest cells exhibit a significantly higher capac-
tory, and urogenital tracts; and endocrine that the VHL protein is involved in the ity for invasion.
and exocrine organs. In the CNS, immu- control of cell cycle exit, i.e. the transition
noreactivity for the VHL protein is promi- from the G2 phase into the quiescent GO Genotype/phenotype
nent in neurons, including Purkinje cells phase, possibly by preventing accumu- Germline mutations of the VHL gene are
of the cerebellum {1526A}. lation of the cyclin-dependent kinase in- spread over the three exons. Missense
hibitor CDKN1B. mutations are most common, but non-
Gene function sense mutations, microdeletions/inser-
The VHL tumour suppressor gene was tions, splice-site mutations, and large
identified in 1993. Mutational inactiva-
tion of the VHL gene in affected family
members is responsible for their genetic
susceptibility to tumour development at
various organ sites, but the mechanisms
by which the inactivation or loss of the
suppressor gene product (the VHL pro- Eloogio B
tein) causes neoplastic transformation
are only partly understood {873).

Transcription elongation factor B binding


One signalling pathway points to a role
of the VHL protein in protein degrada-
tion and angiogenesis. The alpha domain
of the VHL protein forms a complex with
TCEB2, TCEB1, cullin-2, and RBX1 that
has ubiquitin ligase activity, thereby tar-
geting cellular proteins for ubiquitination

••
and proteasome-mediated degradation.
The domain of the VHL gene involved in


- l t ••
the binding to transcription elongation fac-
tor B (also called elongin) is frequently mu-
tated in neoplasms associated with VHL. VEGF
Fig. 16.18 VHL is a classic tumour suppressor gene. The VHL gene product (pVHL) has many different functions. The
HIF1 beta domain forms a complex with elongin and other proteins that regulate the function of hypoxia-inducible factors,
The VHL protein plays a key role in cel- including hypoxia-inducible factor protein (HIF) and VEGF. Under normoxic conditions, HIF degrades. Under hypoxic
lular oxygen sensing, by targeting hy- conditions, HIF accumulates. If pVHL is inactivated, there is no degradation of HIF, leading to an accumulation of VEGF,
poxia-inducible factors (which mediate which explains why tumours associated with van Hippel-Lindau disease are highly vascularized.

Von Hippel-Lindau disease 305


deletions also occur. The spectrum of and phaeochromocytomas, and is main- Genetic counselling
clinical manifestations of VHL reflects the ly caused by missense mutations. Patients with VHL germline mutations
type of germline mutation. Type 2C is characterized by frequent require ongoing medical-genetic coun-
Type 1 is characterized by frequent hae- phaeochromocytomas but absence of selling. Analyses for germline mutations
mangioblastomas and RCCs but rare or haemangioblastomas and RCCs. It is of the VHL gene are recommended for
absent phaeochromocytomas, and is caused by VHL missense mutations, but every patient with retinal or CNS has.
typically caused by deletions, trunca- unlike the other types, shows no evidence mangioblastoma, particularly for younger
tions, and missense mutations. of hypoxia-inducible factor dysregulation. patients and those with multiple lesions,
Type 2A carries a high risk of develop- In accordance with the function of VHL in order to promptly detect tumours as-
ing haemangioblastomas and phaeo- as a tumour suppressor gene, VHL gene sociated with VHL. Periodic screening of
chromocytomas, but rarely RCCs, and is mutations are common in sporadic hae- patients with VHL is mandatory, begin-
caused by missense mutations. mangioblastomas (occurring in as many ning with retinoscopy at 5 years of age
Type 28 is characterized by a high fre- as 78% of cases) and are ubiquitous in and by MRI of the CNS and abdomen at
quency of haemangioblastomas, RCCs, clear cell RCCs. 10 years of age.

Tuberous sclerosis Lopes M.B.S.


Wiestler O.D.
Stemmer-Rachamimov A.O.
Sharma M.C.
Santosh V.
Vinters H.V.

Definition Diagnostic criteria revised in 2012 at the International Tu-


A group of autosomal dominant disor- The diagnosis of tuberous sclerosis is berous Sclerosis Complex Consensus
ders characterized by hamartomas and based primarily on clinical features and Conference 11809} These criteria are es-
benign neoplastic lesions that affect the may be challenging due to the consid- pecially important because genetic test-
CNS and various non-neural tissues. erable variability in phenotype, patient ing is performed in relatively few centres,
Major CNS manifestations of tuberous age at symptom onset, and penetrance and therefore may not be accessible to
sclerosis include cortical hamartomas among mutation carriers. The diagnos- many clinicians. Clinical manifestations
(tubers), subcortical glioneuronal hamar- tic criteria for tuberous sclerosis were are categorized as either major or mi-
tomas, subependymal glial nodules, and Table 16.07 Clinical diagnostic criteria for tuberous
nor features. The diagnostic categories,
subependymal giant cell astrocytomas sclerosis; adapted from Northrup H et al. (1809) which are based on the number of major/
(SEGAs). Major extraneural manifesta- minor manifestations present in a given
Major features
tions include cutaneous angiofibromas ;:: 3 hypomelanotic macules ;:: 5 mm in diameter individual, define disease likelihood as
(so-called adenoma sebaceum), peau ;:: 3 angiofibromas or fibrous cephalic plaque being definite or possible 11809) (see Ta-
chagrin, subungual fibromas, cardiac ;:: 2 ungual fibromas ble 16.07). Most patients have manifesta-
rhabdomyomas, intestinal polyps, viscer- Shagreen patch tions of tuberous sclerosis before the age
al cysts, pulmonary lymphangioleiomy- Multiple retinal hamartomas of 10 years, although some cases may
Cortical dysplasias (including tubers and cerebral
omatosis, and renal angiomyolipomas. manifest much later in life 126} Confirma-
white matter radial migration lines)
Tuberous sclerosis is caused by a muta- Subependymal nodules
tory testing for TSC1 or TSC2 mutations
tion of TSC1 on 9q or TSC2on 16p. Subependyrnal giant cell astrocytoma may be helpful when a patient does not
Cardiac rhabdomyoma meet the clinical criteria for a definite
OMIM numbers 11624} Lymphangioleiomyomatosis diagnosis but the phenotype is compel-
Tuberous sclerosis 1 191100 ;:: 2 angiomyolipomas ling. However, the TSC genes are large
Tuberous sclerosis 2 613254 Minor features and complex, the genetic abnormalities
Confetti skin lesions vary substantially (from point mutations to
Incidence/epidemiology ;:: 4 dental enamel pits deletions), and the testing is not widely
The variability of the clinical manifesta- ;:: 2 intraoral fibromas available. Prenatal diagnosis by mutation
Retinal achromic patch
tions of tuberous sclerosis previously analysis is possible when the mutation in
Multiple renal cysts
led to underdiagnosis. Recent data indi- Non-renal hamartomas
other family members is known.
cate that the disorder affects as many as
Definitive diagnosis: 2 major features or 1 major
25 000-40 000 individuals in the USA and Sites of involvement
feature with ;:: 2 minor features
about 1-2 million individuals worldwide,
with an estimated prevalence of 1 case Possible diagnosis: 1 major feature or z 2 minor Clinical features
features Tuberous sclerosis tends to shorten
per 6000-10 000 live births 11809).

306 Familial tumour syndromes


lifespan (as compared with lifespan in a Table 16.08 Major manifestations of tuberous sclerosis tubers, white matter heterotopia, and
Caucasian control population), but often Manifestation Frequency subependymal hamartomatous nodules
only slightly. The most common causes CNS (i.e. candle guttering or dripping identi-
of death in the second decade of life are Cortical tuber 90-100% fied on neuroimaging studies). Cortical
brain tumours and status epilepticus, Subependymal nodule 90-100% tubers in tuberous sclerosis may be de-
followed by renal abnormalities {1809}. White matter hamartoma and white
90-100% tected by CT or MRI {2342); structural ab-
In patients aged > 40 years, mortality matter heterotopia normalities representative of tubers may
Subependymal giant cell
is most commonly associated with re- 6-16% be co-registered with metabolic brain
astrocytoma
nal abnormalities (i.e. cystic disease or studies, for example, using FOG-PET.
neoplasm) or an unusual proliferative Skin These combined investigations, together
Facial angiofibroma (adenoma
lung condition, lymphangioleiomyomato- 80-90% with intraoperative electrocorticography
sebaceum)
sis. Cardiac rhabdomyomas are often a can identify which of many tubers are
Hypomelanotic macule 80-90%
presenting feature of tuberous sclerosis Shagreen patch 20-40% most likely to be epileptogenic in a given
in newborns and infants aged < 2 years, Forehead plaque 20-30% individual, facilitating tuberectomy as a
and more than half of all individuals found Peri- and subungual fibroma 20-30% reasonable surgical approach to treat-
to have cardiac rhabdomyomas have Eye
ing intractable seizures in patients with
tuberous sclerosis {2133}. Cutaneous Retinal hamartoma 50% tuberous sclerosis. These malformative
manifestations include hypomelanotic Retinal giant cell astrocytoma 20-30% lesions have a strong association with
nodules, facial angiofibromas, and sha- Hypopigmented iris spot 10-20% the development of epilepsy, especially
green patches. Ungual (or subungual) Kidney infantile spasms and generalized tonic-
fibromas often only develop in childhood. Multiple, bilateral angiomyolipoma 50% clonic seizures. They also resemble spo-
Renal angiomyolipomas develop in as Renal cell carcinoma 1.2% radic malformations of cortex not associ-
many as 80% of people with tuberous Polycystic kidney disease 2-3% ated with tuberous sclerosis, classified as
sclerosis by the age of 10 years. Renal Isolated renal cyst 10-20% cortical dysplasia Type llb according to
cysts are present in as many as 20% of Heart the classification proposed by the Inter-
affected individuals, but polycystic kid- Cardiac rhabdomyoma 50% national League Against Epilepsy (ILAE)
ney disease only occurs in 3-5%. Lym- Digestive system {225). Microscopically, they consist of gi-
phangioleiomyomatosis is a condition Microhamartomatous rectal polyp 70-80% ant cells (like those seen in SEGA) and
of unknown pathogenesis that severely Liver hamartoma 40-50% dysmorphic neurons, disrupted cortical
impairs lung function and may be fatal; Hepatic cyst 24% lamination, gliosis, calcification of blood
Adenomatous polyp of the
it is present in as many as 40% of adult Rare vessel walls and/or parenchyma, and my-
duodenum and small intestine
women with tuberous sclerosis. All the elin loss. The surrounding cortex usually
phenotypic features of tuberous sclerosis Lung demonstrates a normal cytoarchitecture
can also occur sporadically in individuals Lymphangioleiomyomatosis 1-2.3% on cursory examination, although this
Pulmonary cyst 40%
without the genetic condition {2133). For conclusion is being questioned based on
Micronodular pulmonary hyper-
example, about 50% of patients with lym- Rare more detailed immunohistochemical and
plasia of type II pneumocytes
phangioleiomyomatosis do not have tu- morphometric investigations {1072,1582).
Other
berous sclerosis; sporadic angiomyolipo- Dysmorphic neurons and giant cells may
Gingival fibroma 50-70%
mas can occur but are typically solitary, Pitting of dental enamel 30% be seen in all cortical layers and the un-
whereas tuberous sclerosis-associated Bone cyst 40% derlying white matter. The dysmorphic
angiomyolipomas are often multiple or bi- Arterial aneurysm (intracranial neurons show altered radial orientation
Rare
lateral. The proteins tuberin and hamartin arteries, aorta, and axillary artery) in the cortex, aberrant dendritic arbori-
(products of the TSC2 and TSC1 genes, zation, and accumulation of perikaryal
respectively; see Other CNS manifesta- fibrils. The perikaryal fibrils can be high-
tions) are identifiable by immunohisto- In people with tuberous sclerosis, these lighted using silver impregnation tech-
chemistry and western blotting in many manifestations are linked to the structural niques, which show many neurons with
organs and tissues throughout the body changes that involve cortex and subcor- neurofibrillary tangle-like morphology.
{1170} tical white matter, usually as tubers (see Another frequently observed element in
Neurological symptoms are among the Other CNS manifestations), although me- tubers and adjacent brain (cortex and
most frequently observed and serious ticulous autopsy studies on small num- white matter) is the characteristic so-
(sometimes life-threatening) manifesta- bers of patients have also suggested called balloon cell. Balloon cells resem-
tions of tuberous sclerosis {505,2538}. that there may be more subtle degrees of ble gemistocytic astrocytes in that they
The most common initial signs of tuber- cortical and white matter disorganization have eosinophilic glassy cytoplasm and
ous sclerosis are intractable epilepsy {1582). may be clustered in small groups, but un-
including infantile spasms (in 80-90% like gemistocytes, they often show promi-
of cases), cognitive impairment (in 50%), Subependymal giant cell astrocytoma nently nucleolated nuclei {505,1009,
autism spectrum disorder (in as many as Seep. 90. 1072). A spectrum of cellular elements
40%), and neurobehavioural disorders with features of both neurons and astro-
(in � 60%) {863,2538}; these presenta- Other CNS mamfestations cytes may be noted in the brains of peo-
tions may have any of several etiologies. CNS lesions include cerebral cortical ple with tuberous sclerosis. Although the

Tuberous sclerosis 307


Fig. 16.19 Unusual cells within and adjacent to tuberous sclerosis cortical tubers. Panel A (inset, black arrow) shows a cell with eosinophilic cytoplasm and unusual dendritic
arborization; a nearby cell (white arrow) shows neuronal morphology, with a nucleolated nucleus and amphophilic cytoplasm lacking obvious Nissl substance. Panel B shows a
dysmorphic enlarged neuron (arrows). Panels C and D show balloon-like cells with eosinophilic cytoplasm; the cell in C is binucleated, whereas the cell in D shows a cytoplasmic
vacuole (arrow). All panels are from H&E-stained sections.

neurons express neuronal-associated proteins were developed. lmmunostain- cortical disorganization, and (cortical) gi-
proteins. they display cytoarchitectural ing a given tuber with antihamartin or ant cells with organellar dysfunction with-
features of immature or poorly differen- antituberin antibodies does not provide in the brains of affected animals {874)
tiated neurons, such as reduced axonal evidence of which mutation is present in This phenotype could be rescued by
projections and spine density (1009, a given subject, and therefore is not of postnatal administration of sirolimus (also
1072}. Giant cells in cortical tubers show great diagnostic value. Both proteins are known as raparnycin), which resulted in
a cellular and molecular heterogeneity widely expressed throughout the CNS of abrogation of both seizures and prema-
similar to that seen in SEGA. and immu- the normal developing brain (1169,2655) ture death.
nohistochemical markers characteristic Many approaches have been taken to
of glial and neuronal phenotypes sug- studying tubers, especially surgically re- Extraneura/ mamfestations
gest a mixed glioneuronal origin of these sected lesions, because DNA, mRNA, The extraneural manifestations of tu-
cells. Many giant cells in tubers express and proteins are better preserved within berous sclerosis and the frequencies
nestin mRNA and protein (506} Some gi- them than in autopsy specimens, and au- at which they occur are summarized in
ant cells demonstrate immunoreactivity topsies of patients with tuberous sclerosis Table 16.08.
for GFAP (1009}. but others with an iden- are rare. Cell biology approaches have
tical morphological phenotype express also been taken to examining the biology Molecular genetics
neuronal markers. including gap junction of hamartin and tuberin and how they Tuberous sclerosis is caused by inac-
beta-2 protein and gap junction beta-1 may mediate cell adhesion through the tivating mutations in one of two genes:
protein (also called connexins 26 and ERM proteins (ezrin, radixin, and moes- TSC1 at 9q or TSC2 at 16p. The proteins
32). neurofilaments. class Ill beta-tubulin, in) and the GTPase Rho (1423). Deep encoded by the TSC genes. tuberin and
MAP2, and alpha-internexin (506,1009}. sequencing of TSC1, TSC2, and KRAS hamartin, interact within the cell and form
However, formation of well-defined syn- demonstrates that small second-hit mu- a complex (486,1212,1987} Mutation of
apses between giant cells and adjacent tations in these genes are rare events either gene results in disrupted function
neurons is not a consistent finding. Cor- within tubers (2048). Insulin signalling of the tuberin-hamartin complex, result-
tical hamartomas morphologically indis- pathways •(normally impacted through ing in similar disease phenotypes. In
tinguishable from tubers may occur in inhibition by both tuberin and hamartin) sporadic tuberous sclerosis cases. mu-
chronic focal epilepsies without clinical show subtle but definite differences in tations are 5 times as common in TSC2
or genetic evidence of an underlying tuberous sclerosis tubers versus foci of as in TSC1 (51,516,1174}. whereas in
tuberous sclerosis condition (223,225, severe (i.e. ILAE Type llb) cortical dys- families with multiple affected members
2305). The pathogenesis of these spo- plasia {1690}. Electrophysiological ap- the mutation ratio of the two genes is
radic lesions is unclear. Subependymal proaches have also shown differences in 1:1 (2231). TSC1 or TSC2 mutations are
hamartomas are elevated, often calcified neurophysiological and synaptic abnor- identified in about 85% of patients with
nodules. They are composed of cells in- malities in surgically resected brain tis- tuberous sclerosis. The remaining 15% of
distinguishable from those found in cor- sue samples from patients with tuberous cases may be mosaics or have a muta-
tical tubers, but are smaller in size than sclerosis versus patients with severe cor- tion in an unanalysed non-coding gene
cortical tubers. tical dysplasia (390). A mouse model of area. Mosaicism has been reported tor
Soon after the TSC2 and TSC1 genes tuberous sclerosis, in which mosaic Tsc1 TSC1 and TSC2 mutations in some par-
were first cloned in the 1990s, probes for loss was induced in neural progenitor ents of patients with sporadic cases and
the gene transcripts and the translated cells, showed megalencephaly, marked in patients with tuberous sclerosis (2229,

308 Familial tumour syndromes


2646). Alternatively, there may be a third, of the 180 kOa protein product tuberin the two-hit hypothesis for tuber formation
unknown locus, although to date there bears significant homology with the cata- (683A,2862A).
is no evidence to support this possibility lytic domain of RAP1GAP, a member of
(2231 ). Patients with tuberous sclerosis the RAS family. Signalling pathways involving tuberin
with no mutations identified have milder and hamartin
phenotype than do patients with TSC1 or Gene mutations The tuberin-hamartin complex is a sig-
TSC2 mutations (2231) The mutational spectrum of TSC2 is wid- nalling node that integrates growth fac-
er than that of TSC1; it includes large de- tor and stress signals from the upstream
The TSC1 gene letions and missense mutations, and Jess P/3K/AKT pathway and transmits signals
The TSC1 gene maps to chromosome frequently, splice junction mutations { 51, downstream to coordinate multiple cel-
9q34 (486) and contains 23 exons (2625). 516,1174). Exons 16, 33, and 40 have the lular processes, including cell prolifera-
21 of which carry coding information. highest number of mutations. Large dele- tion and cell size (486,1212,1987). The
tions in the TSC2 gene may extend into complex negatively regulates the mTOR
Gene expression the adjacent PKD1 gene. with a resulting pathway (96,783,2528). Disruption of
The TSC1-encoded protein, harnar- phenotype of tuberous sclerosis and pol- the tuberin-hamartin complex causes
tin, has a molecular weight of 130 kOa. ycystic kidney disease (231,2626). upregulation of the mTOR pathway and
Hamartin is strongly expressed in brain, Multiple studies of genotype-phenotype increases proliferation and cell growth
kidney, and heart, all of which are tissues correlations have demonstrated that through two effector molecules: 4E-BP1
frequently affected in tuberous sclerosis TSC2 mutations are associated with a and S6K1 {96,2528). The understand-
(1986). Its pattern of expression overlaps more severe phenotype overall: earlier ing of the basic mechanism of mTOR
with that of tuberin, the product of the seizure onset, higher number of tubers, pathway activation in tuberous sclerosis
TSC2gene. and lower cognition index. However, lesions has led to the use of mTOR in-
within that spectrum, TSC2missense mu- hibitors in the treatment of manifestations
Gene mutations tations are associated with milder pheno- of tuberous sclerosis. Several tuberous
Mutation analysis of large cohorts (418, types (97,2231}. sclerosis-associated tumours, renal an-
2626) showed that the most common Like in other tumour suppressor gene giomyolipomas, SEGAs, and lymphangi-
mutations in the TSC1 gene are small syndromes, somatic inactivation of the oleiomyomas show significant size re-
deletions and nonsense mutations (each wildtype allele (i.e, LOH for the TSC1 or duction in response to treatment with
accounting for -30% of all mutations in TSC2 locus) has been reported in kid- mTOR inhibitors, and regrow when treat-
the gene). Virtually all mutations result in ney and cardiac lesions associated with ment is stopped. The effects of mTOR
a truncated gene product, and more than tuberous sclerosis, as well as in SEGAs inhibitors are currently being evaluated
half of the changes affect exons 15 and (406). However, there is conflicting evi- for the clinical management of epilepsy
17 {2626). dence of whether a so-called second hit and other neurological manifestations of
is required for cortical tuber formation, tuberous sclerosis {509).
The TSC2 gene raising the possibility that some lesions
The TSC2 gene maps to chromosome in tuberous sclerosis may be due to hap- Inheritance and genetic heterogeneity
16p13.3 {1212) and contains 40 exons. loinsufficiency (1787,2762). Furthermore, Most tuberous sclerosis cases (-60%)
there is no evidence of inactivation of are sporadic, with no family history, indi-
Gene expression TSC1 or TSC2 in focal cortical dyspla- cating a high rate of de novo mutations
TSC2 encodes a large transcript of sias, which are histologically very similar (2229A). In affected kindreds, the dis-
5.5 kb, which shows widespread expres- to tuberous sclerosis tubers (2775). In ease follows an autosomal dominant pat-
sion in many tissues, including the brain one recent study, loss of TSC1 in perive- tern of inheritance, with high penetrance
and other organs affected in tuberous ntricular zone neuronal stem cells was but considerable phenotypic variability
sclerosis. Alternatively spliced mRNAs sufficient to cause aberrant migration (2354).
have been reported (2799). A portion and giant cell phenotype, supportive of

Tuberous sclerosis 309


Li-Fraumeni syndrome Olivier M.
Kleihues P.
Ohgaki H.

Table 16.09 Frequency of tumour manifestation in various organ/tissue sites in TP53 germline mutation carriers
Definition
An autosomal dominant disorder char- % of all tumours
Organ/tissue Typical histological types in TP53 germ line
acterized by multiple primary neoplasms
mutation carriers
in children and young adults, with a
predominance of soft tissue sarcomas, Breast Carcinoma 31%
osteosarcomas, breast cancer, brain tu- Soft tissue Soft tissue sarcoma 14%
mours, and adrenocortical carcinoma. CNS Astrocytoma, glioblastoma, medulloblastoma, choroid plexus tumour 13%
Li-Fraumeni syndrome (LFS) is most
Adrenal gland Adrenal cortical carcinoma 12%
commonly caused by a germline muta-
tion in the TP53 tumour suppressor gene Bone Osteosarcoma 9%
on chromosome 17p13 (736,1575,2634).
diagnosis of LFL are sarcoma at any age carcinoma) at any age, plus one first- or
OMIM number (1624) 151623 in the proband, plus any two of the follow- second-degree relative in the same line-
ing tumours within the family (including age with any cancer diagnosed at an age
Incidence/epidemiology within a single individual): breast cancer ot « 60 years (208}.
TP53 germline mutations have been es- at < 50 years, brain tumour, leukae-
timated to occur at a rate of about 1 in mia, adrenocortical tumour, melanoma, The Chompret cotene
5000 to 1 in 20 000 births, and to account prostate cancer, pancreatic cancer at The 2009 version of the Chompret criteria
for as many as 17% of all familial cancer < 60 years, or sarcoma at any age (1841}. for the diagnosis of LFL are (1) a tumour
cases (864,1883} Genetic and pedigree belonging to the LFS tumour spectrum
information on 767 families containing The LFL -8 defimtion (i.e. soft tissue sarcoma, osteosarcoma,
carriers of a TP53 germline mutation is The criteria of the LFL-B definition (the premenopausal breast cancer, brain tu-
available in the International Agency for definition by Birch) for the diagnosis of mour, adrenocortical carcinoma, leukae-
Research on Cancer (IARC) TP53 Muta- LFL are any childhood cancer or sar- mia, or lung adenocarcinoma in situ) at
tion Database [http://p53.iarc.fr]. coma, brain tumour, or adrenocortical < 46 years in the proband and at least
carcinoma at < 45 years in the proband, one first- or second-degree relative ei-
Diagnostic criteria plus one first- or second-degree relative ther with an LFS tumour (other than
with a cancer typically associated with breast cancer if the proband is affected
Classic Li-Fraumeni syndrome clinical LFS (i.e. sarcoma, breast cancer, brain by breast cancer) at < 56 years or with
craene tumour, leukaemia, or adrenocortical multiple tumours; or (2) multiple tumours
The classic LFS clinical criteria used to
identify an affected individual in a fam-
ily affected by LFS are (1) occurrence of BREAST (30.6%) 34yrs
sarcoma before the age of 45 years, (2) SOFT TISSUES (13.8%) ----····22yrs
at least one first-degree relative with any
tumour before the age of 45 years, and BRAIN (13%) ••••• 18yrs

(3) a second- or first-degree relative with ADRENAL GLAND (11.7%) -7yrs


cancer before the age of 45 years or a
BONES (8.6%)
sarcoma at any age (1486).
HEMATOP. SYSTEM (2.7) • 24yrs
Cr1teria for diagnosis of Li-Fraumeni-ltke SKIN (2.5%) I 45 yrs
syndrome
LUNG (2.5%) .44 yrs
Three main sets of criteria for the diagno-
sis of Li-Fraumeni-like syndrome (LFL), COLON (2.1%) I 39 yrs
a variant of LFS, have been proposed to OVARY (1.9%) 40 yrs
better identify TP53 germline mutation
carriers: the LFL-E2 definition, the LFL-B STOMACH (1.4%) I 39 yrs Females •Males

definition, and the Chompret criteria Others(9.3%) - 34yrs

-30 20 70 120 170 220 270 320 370 420


The LFL -E2 defimtion
The criteria of the LFL-E2 definition Tumourcounts
(the second definition by Eeles) for the Fig. 16.20 Target organs for tumorigenesis in 1350 patients carrying a TP53 germline mutation.

310 Familial tumour syndromes


80 (except multiple breast cancers) in the
proband, the first of which occurs at
70 ADRENAL GLAND
< 46 years, and with at least two belong-
60
ing to the LFS spectrum; or (3) adreno-
50 cortical carcinoma or choroid plexus
% 4-0 carcinoma in the proband, regardless of
30 family history (2559)
20
10
Sites of involvement
Breast cancer, soft tissue sarcomas,
0
CNS tumours, adrenal tumours, and
35 bone tumours are the most frequent man-
30 SOFT TISSUE ifestations of LFS, accounting for about
80% of all tumours in patients carrying
25
a TP53 germline mutation. The sporadic
% 20 counterparts of these tumours also show
a high frequency of TP53 mutations,
15
suggesting that in these neoplasms,
10
TP53 mutations are capable of initiating
5 the process of malignant transformation
0 (1294). In general, tumours associated
with a TP53 germline mutation develop
35
earlier than their sporadic counterparts,
30 BRAIN but there are marked organ-specific dif-
25 ferences. Adrenocortical carcinoma as-
sociated with a TP53 germline mutation
20
% develops almost exclusively in children,
15 in contrast to sporadic adrenocortical
10 carcinoma, which has a broad age dis-
tribution with peak incidence in patients
5
aged> 40 years (136).
0
50 Nervous system neoplasms
BONE In the 944 individuals carrying a TP53
4-0 germline mutation who were included in
the IARC TP53 Database as of Novem-
30 ber 2013, a total of 1485 tumours were
% 20
reported; 192 (13%) of which were locat-
ed in the nervous system. The male-to-
female ratio of patients with brain tumours
10
associated with TP53 germline mutation
is 1.5:1 [http://p53.iarc.fr].
0
As with sporadic brain tumours, the age
30 of patients with nervous system neo-
25
BREAST plasms associated with TP53 germline
mutations shows a bimodal distribution.
20 The first incidence peak is in children
% 15
(mainly medulloblastomas and related
primitive neuroectodermal tumours, cho-
10 roid plexus tumours, and ependymo-
mas), and the second is in the third and
5
fourth decades of life (mainly astrocytic
0 brain tumours).
0-5 6-10 11-15 16-20 21-25 26-30 31-35 36-40 41-45 >45
Gene structure
Age at onset (years)
The TP53 gene on chromosome 17p13
Fig. 16.21 The age distribution of tumours in carriers of a TP53 germline mutation shows remarkable differences.
has 11 exons spanning 20 kb. Exon 1 is
Adrenocortical carcinomas occur almost exclusively in children aged s 5 years. Soft tissue tumours (sarcomas) and
brain tumours show an incidence peak in young children, whereas the remainder of the cases show a wide age non-coding, and exons 5 to 8 are highly
distribution. Bone tumours have an incidence peak in the second decade of life, and breast carcinomas occur in young conserved among vertebrates.
adults. Reprinted from Olivier M et al. (1294).

Li-Fraumeni syndrome 311


Distribution of TP53 germline mutations
>45 •Gliana Most germline mutations of the TP53
• PNET/Medulloblastoma gene are spread over exons 5-8. with
41-45 major hotspots at codons 133, 175, 245
Choroid plexus tumors
........ 248, and 273 or 337. Missense mutation�
(/) 36-40 • Others
roQ) are most common. but nonsense muta-

-
• Unclas. tions, deletions/insertions. and splice-
>- 31-35
(/) site mutations also occur. Mutations ob-
'cij 26-30
0 served at these codons are missense
c
0) mutations that result in mutant proteins
ro 21-25
'5 with complete loss of function, dominant
ro
Q)
16-20 negative phenotypes, and oncogenic
activities.
0) 11-15
-c Some codons (e.g. Cys176 and Arg249)
6-10 that are commonly somatically mutated
in sporadic tumours have never been
0-5 reported as germline mutations (18411.
Residue 176 (Cys) is involved in the co-
0 10 20 30 40 50 60 ordination of a zinc atom that forms a
Tumor counts bridge between domain 1 and domain 3,
Fig. 16.22 Patient age at diagnosis and histological diagnosis of 173 brain tumours in carriers of a TP53 germline and is crucial in stabilizing the architec-
mutation [http://p53.iarc.fr/SelectedStatistics.aspx]. PNET, primitive neuroectodermal tumour; Unclas., unclassified. ture of the whole DNA-binding domain.
Residue 249 (Arg) makes essential con-
tacts with several residues of the scaffold
through hydrogen bridges {4471. Two
residues (codons 133 and 337) are hot-
Family 1 Family2
spots for TP53 germline mutations, but
TP53 germ line mutation TP53 germline mutation
these are less frequent in sporadic can-

62r••
(codon 248, CGG·> TGG, Arg-> Trp) (codon 248, CGG·> TGG, Arg·> Trp)

cers. A mutation at codon 133 (M133T)


has been found in families with clustering
of early-onset breast cancers (3-6 cases
per family, with a mean patient age at on-
D set of 34 years) [http://p53.iarc.fr]. and a
40-50
BT
36 LGA 47LGA 29 AA mutation at codon 337 (R337H) has been
nd R132C R132C
37 GBM Normal brain frequently found in Brazilian children af-
nd Wt
fected by adrenocortical carcinomas
{2111 I and Brazilian families affected by
LFL {51.
Family 3
4ACCWt TP53 germline mutation Types of TP53 mutations
6CPP Wt
WBCWI
(codon 236 deletion) The proportion of G:C->A:T transitions
O.BCPC WBC at CpG sites is higher in TP53 germline
Wt WI
WBC mutations than in somatic mutations,
Wt
but the proportions of G:C->A:T transi-
tions at non-CpG sites and of G:C->T:A
transversions are lower. G:C->A:T transi-
tions at CpG sites are considered to be
endogenous (e.g. resulting from deami-
nation of 5-methylcytosine. which occurs
spontaneously in almost all cell types
nd but is usually corrected by DNA repair
Fig. 16.23 IDH1 mutations in three Li-Fraumeni families carrying a TP53 germline mutation. The IDH1 codon 132 mechanisms). The difference observed
status is indicated in blue letters. Wt, /OH1-wildtype; nd, not determined. DNA sequencing revealed identical IDH1 may thus be explained by the fact that
R132C mutations in astrocytomas. All mutations were heterozygous. Round symbols, females; square symbols, males; non-CpG G:C->A:T and G:C->T:A mu-
black gender symbol, carrier of TP53 germline mutation; white gender symbol, TPS3-wildtype; grey gender symbol, no tations are associated with exogenous
DNA available. Numbers below symbols indicate age at diagnosis or age at death (slash through symbol). All mutations carcinogen exposure, whereas germline
were R132C mutations (CGT->TGT), which in sporadic astrocytomas account for< 5% of IDH1 mutations.
mutations seem to result mainly from en-
AA, anaplastic astrocytoma; LGA, low-grade diffuse astrocytoma; GBM. glioblastoma multiforme; ACC. adrenocortical
carcinoma; CPP, choroid plexus papilloma; CPC, choroid plexus carcinoma; S, schwannoma; BT, brain tumour (no dogenous processes {1842,1843).
histological verification); WBC, white blood cells. No detailed medical history was available for family members without
alphabetical letters. Reprinted from Watanabe T et al. (2710).

312 Familial tumour syndromes


and cell type. TP53 thus influences a
wide range of biological and physiologi-
cal processes (925). The functions of p53
rely mainly on its transcriptional activity,
but it can also act via interactions with
various proteins. The roles of other pro-
tein isoforms in these activities remain
largely unknown (1581).
In most human cancers, TP53 is inacti-
vated through gene mutations that con-
fer loss of the tumour suppressor role.
p53-mutant proteins differ from each
other in the extent to which they have lost
suppressor function and in their capac-
ity to inhibit wildtype p53 in a dominant
negative manner (1232,1958). In addi-
tion, some p53 mutants seem to exert an
oncogenic activity of their own, but the
molecular basis of this gain-of-function
Fig. 16.24 Hotspot codon positions associated with adrenal gland carcinoma and brain tumours. A three-dimensional
phenotype is still unclear (212). The func-
view of the central DNA-binding domain of the p53 protein in complex with DNA. Structural groups of residues are
shown in different colours: group 1 residues, which correspond to L2 and L3 loops (binding in the minor groove of the
tional characteristics of each p53-mutant
DNA helix) are red; group 2 residues, which correspond to L 1 loop and S2-S2'-H2 motifs (binding in the major groove protein may depend, at least in part, on
of the DNA helix) are yellow; and group 3 residues, which correspond to the non-DNA-binding loops, the beta-sheet the degree of structural perturbation that
skeleton, or the oligomerization domain, are cyan. The codon positions of the mutations associated with adrenal gland the mutation imposes on the protein.
carcinoma and brain tumours are indicated in violet and blue, respectively (1841).
Genotype/phenotype
Multiple germline mutations progression, DNA integrity, and the sur- Among 139 families with at least one case
Recently, a family affected by LFS, with vival of cells exposed to DNA-damaging of brain tumour, the mean number of
a TP53 germline mutation (codon 236 agents and non-genotoxic stimuli such CNS tumours per family was 1.55. Sev-
deletion) and multiple nervous system as hypoxia. DNA damage or hypoxia in- eral reported families showed a remarka-
tumours, was found to have additional duces a transient nuclear accumulation ble clustering of brain tumours (155,1283,
germline mutations. Missense muta- and activation of the p53 protein, with 271 OJ. This raises the question of whether
tions in the MSH4 DNA repair gene transcriptional activation of target genes some mutations carry an organ-specific
(c.2480T-->A; p.1827N) were detected that are responsible for the induction of or cell-specific risk.
in 3 patients with gliomas (2 anaplastic cell cycle arrest or apoptosis (1311,1477). An analysis of the IARC TP53 Database
astrocytomas, 2 glioblastomas). Another [http://p53.iarc.fr] of germline mutations
2 family members, who developed pe- Gene function showed that brain tumours were more
ripheral schwannomas without a TP53 The p53 protein is a multifunctional tran- likely to be associated with missense
germline mutation, also carried the MSH4 scription factor involved in several path- mutations located in the DNA-binding
germline mutation, as well as a germline ways. Its best-characterized functions surface of p53 protein that make contact
mutation of the LATS1 gene (c 286C-->T; are in the control of cell cycle progres- with the minor groove of DNA {1841). The
p.R96W), a downstream mediator of the sion, DNA integrity, and the survival of type of mutation was also associated
NF2 gene {1283). cells exposed to DNA-damaging agents. with the patient age at onset of brain
However, accumulating evidence indi- tumours; truncating mutations were as-
Gene expression cates that p53 also regulates other im- sociated with early-onset brain tumours
The TP53 tumour suppressor gene en- portant processes, such as cell oxidative {1841 }. Familial clustering may also be
codes a 2.8 kb transcript encoding a metabolism, the cellular response to nu- due to gene-environment interactions;
393 amino acid protein that is widely trient deprivation, fertility, and the division for example, exposure of families to simi-
expressed at low levels. This protein and renewal of stem cells. The extent and lar environmental carcinogens or lifestyle
is a multifunctional transcription fac- consequences of the biological response factors has been suggested in stomach
tor involved in the control of cell cycle elicited by p53 vary according to stress and breast cancer (1294).

Li-Fraumeni syndrome 313


.,

Cowden syndrome Eberhart C.G.


Wiestler OD.
Eng C.

Definition Table 16.10 International Cowden Consortium diagnostic criteria for Cowden syndrome
An autosomal dominant disorder charac- operational diagnostic criteria on the basis of the published literature
terized by multiple hamartomas involving Pathognomonic criteria and their own clinical experience {640,
tissues derived from all three germ cell Adult Lhermitte-Duclos disease (LDD) 2849). Trichilemmomas and papilloma-
layers and a high risk of breast, epithe- Mucocutaneous lesions tous papules are particularly important
lial thyroid, endometrial, renal, and colon T richilemmomas (facial) to recognize. Cowden syndrome usually
cancers. Facial trichilemmomas are high- Acral keratoses presents within the third decade of life.
ly characteristic of Cowden syndrome, Papillomatous papules It has variable and broad expression
which is caused mainly by germline mu- Mucosal lesions and an age-related penetrance. By the
tations in PTEN. Adult-onset dysplastic Major criteria third decade of life, 99% of affected in-
cerebellar gangliocytoma (Lhermitte- Breast cancer dividuals have developed mucocutane-
Duclos disease) is also considered to Thyroid cancer ( especially follicular) ous stigmata, although any of the other
be pathognomonic {245) Recently, other Macrocephaly (> 97th percentile) features could already be present. The
Cowden syndrome predisposition genes Endometrial carcinoma most commonly reported manifestations
have also been identified: the SDH Minor criteria are mucocutaneous lesions, thyroid ab-
genes, PIK3CA, and KLLN. Other thyroid lesions (e.g. goitre or nodule) normalities, fibrocystic disease and car-
Mental retardation cinoma of the breast, gastrointestinal
OMIM number {1624) 158350 Hamartomatous intestinal polyps hamartomas, multiple early-onset uterine
Li po mas leiomyomas, macrocephaly (specifically,
Incidence/epidemiology Fibrocystic breast disease megalencephaly), and mental retardation
Before the identification of PTEN, the in- Fibromas {941,1525,2414,2849}.
Genitourinary tumours (e.g. uterine fibroids, renal cell
cidence of Cowden syndrome was esti-
carcinoma) or malformations
mated to be 1 case per 1 million popu- Dysplastic cerebellar gangliocytoma
lation {2414} After this gene for Cowden Requirements for diagnosis (Lhermitte-Duclos disease)
syndrome had been identified {1499). a Mucocutaneous lesions if: This unusual tumour of the CNS is closely
molecular-based estimate of prevalence - Six or more facial papules (of which three or associated with Cowden syndrome { 641,
in the same population was 1 case per more must be trichilemmoma), or 1533,1873) Adult-onset Lhermitte-Du-
- Cutaneous facial papules and oral mucosal
200 000 population {1764). Due to dif- clos disease, even in the absence of
papillomatosis, or
ficulties in recognizing this syndrome, - Oral mucosal papillomatosis and acral other features or family history, is highly
prevalence figures are likely to be under- keratoses, or predictive of a germline mutation in PTEN
estimates. One recent study estimated - Six or more palmoplantar keratoses {2864}, and Lhermitte-Duclos disease
de novo PTEN mutation frequency to be - Two or more major criteria met (one must be is now considered pathognomonic for
about 11% at minimum and 48% at maxi- macrocephaly or LDD) Cowden syndrome. Other malignancies
mum {1653). One major criteria and three minor criteria and benign tumours have also been
Four minor criteria
reported in patients or families with
Diagnostic criteria Requirements for diagnosis in individuals with a Cowden syndrome. Some authors be-
The National Comprehensive Cancer family member with Cowden syndrome lieve that endometrial carcinoma could
Network has established a set of opera- A pathognom'bnic criterion also be a component tumour of Cowden
tional clinical diagnostic criteria for iden- Any one major criterion with or without minor criteria syndrome. It remains to be determined
tifying individuals with possible Cowden Two minor criteria whether other tumours (e.g. sarcomas,
History of Bannayan-Riley-Ruvalcaba syndrome
syndrome [http://www.nccn.org]. A re- lymphomas, leukaemias, and meningi-
cent prospective study subsequently omas) are true components of the syn-
led to the development of the Cleveland adult-onset Lhermitte-Duclos disease drome. For details, see Oysplastic cere-
Clinic score - a semiquantitative scoring was revised from a major diagnostic cri- bellar gangliocytoma (Lhermitte-Ouclos
system that has shown greater adequacy terion to a pathognomonic criterion and disease), p. 142.
than the National Comprehensive Can- given the highest weight (of 10) in the
cer Network criteria [http://www.lerner. PTEN Cleveland Clinic scoring system. Intestinal hamartomatous polyps
ccf.org/gmi/ccscore] {2503) In response Because of the variable and broad ex- In a small but systematic study of 9 well-
to the results of a study that found PTEN pression of Cowden syndrome and the documented cases of Cowden syndrome
mutations in 15 of 18 unselected patients lack of uniform diagnostic criteria prior to (7 of which had a germline mutation in
with a pathological diagnosis of dys- 1996, the International Cowden Consor- PTEN), all 9 patients had hamartoma-
plastic cerebellar gangliocytoma {2864}, tium (ICC) {1765) compiled operational tous polyps {2712). Several varieties of

314 Familial tumour syndromes


hamartomatous polyps are seen in this are approximately 11 % (among women), identified as an important downstream
syndrome, including hamartomas most and 1 % (among both sexes), respec- response to PTEN dysfunction or defi-
similar to juvenile polyps composed of tively. Breast cancer has also been rarely ciency (see Gene expression), mTOR in-
a mixture of connective tissues normally observed in men with Cowden syndrome hibition was shown to be effective in vitro
present in the mucosa (principally smooth {1589}. In women with Cowden syndrome, and in animal models {24071 The mTOR
muscle in continuity with the muscularis estimates of the lifetime risk of breast inhibitor sirolimus (also known as rapam-
mucosae), lipomatous and ganglioneu- cancer range from 25% to 50% {640,941, ycin) was shown to be effective in a child
romatous lesions, and lymphoid hyper- 1525,2414,2849}. The mean patient age with Proteus syndrome who also carried
plasia {364,2712}. These polyps are found at diagnosis is probably about 10 years a germline mutation in PTEN {1594f. An
in the stomach. duodenum, small bowel, younger than for breast cancer occurring open-label phase II trial (NCl-08-C-0151)
and colon. Those in the colon and rectum in the general population {1525,24141. of sirolimus for the treatment of human
usually measure 3-10 mm in diameter, Although Rachel Cowden died of breast Cowden syndrome and other syndromes
but can reach 2 cm or more. Some of the cancer at the age of 31 years {293,1521} characterized by germline PTEN muta-
polyps are no more than tags of mucosa, and the youngest reported patient age tions was recently completed, but the
s, but others have a more definite structure. at diagnosis of breast cancer is 14 years results have not yet been published
Examples containing adipose tissue have {2414}, the great majority of breast can- [https://clinicaltrials.gov/ct2/show/study/
been described. The mucosal glands cers are diagnosed in patients aged NCT00971789].
within the lesion are normal or elongated > 30-35 years (range 14-65 years)
and irregularly formed, but the overlying {1525f. The predominant histology is Chromosomal location and mode of
epithelium is normal and includes goblet ductal adenocarcinoma. Most Cowden transmission
cells and columnar cells {364f. The pres- syndrome breast carcinomas occur in Cowden syndrome is an autosomal
ence of some ganglion tissue is not unu- the context of ductal carcinoma in situ, dominant disorder, with age-related
sual in the juvenile-like polyps. Lesions in atypical ductal hyperplasia, adenosis, penetrance and variable expression
which autonomic nerves are predominant and sclerosis {2298f. 1641,1786,2504}. The major Cowden
(resulting in a ganglioneuroma-like ap- syndrome susceptibility gene, PTEN, is
pearance) have also been described, but Thyroid cancer located on 10q23.3 {1489,1499,1765}.
seem to be exceptional {1434}. The vast The lifetime risk of epithelial thyroid can- Other predisposition genes include the
majority of Cowden syndrome hamar- cer can be as high as 10% in patients SDH genes, PIK3CA, AKT1, and KLLN
tomatous polyps are asymptomatic, al- with Cowden syndrome. Because of {168,1781,1782,1852}.
though adenomatous polyps and colon the small number of cases, it is unclear
cancers have been observed in young whether the average patient age at onset Gene structure
patients with this condition l2850f. The in this setting is truly younger than that PTEN, on 10q23, consists of nine exons
association of gastrointestinal malignan- in the general population. Histologically, spanning 120-150 kb of genomic dis-
cy with Cowden syndrome is unknown, thyroid cancer is predominantly follicular tance and encodes a 1.2 kb transcript
but appears to be likely. In a study of carcinoma, although papillary histology and a 403 amino acid lipid dual-specific-
9 individuals with Cowden syndrome, gly- has also been rarely observed 1941,1576, ity phosphatase (dephosphorylating both
cogenic acanthosis of the oesophagus 2414,2849} Medullary thyroid carcinoma protein and lipid substrates). which is
was found in 6 of 7 individuals with PTEN has not been observed in patients with homologous to the focal adhesion mole-
mutation 12712). It is likely that many Cowden syndrome. cules tensin and auxilin {1489,1589,2418}.
more patients with Cowden syndrome will The amino acid sequence that is homolo-
be identified in the future with ongoing Skin tumours gous to tensin and auxilin is encoded by
screening for colon cancer, which should The most important benign tumours in exons 1-6. A classic phosphatase core
enable a more precise characterization of Cowden syndrome are trichilemmomas motif is encoded within exon 5, which
the phenotypic gastrointestinal features and papillomatous papules of the skin. is the largest exon, constituting 20% of
of this disease and the possible risk of Benign tumours and disorders of the the coding region 11485,1489,2418}. A
gastrointestinal cancer. In an unselected breast and thyroid are the next most longer isoform of PTENhas also been de-
series of 4 children with juvenile polyposis common, and probably constitute true scribed, which apparently interacts with
of infancy, this deletion also involved BM- component features of the syndrome. Fi- the mitochondrion. but its clinical impact
PR1A, upstream of PTEN. Subsequently, broadenomas and fibrocystic disease of is still unclear {2043}.
germline deletion involving both PTEN the breast are common signs of Cowden
and BMPR1A was shown to characterize syndrome, as are follicular adenomas Gene expression
at least a subset of juvenile polyposis of and multinodular goitre of the thyroid. PTEN is virtually ubiquitously expressed
infancy {571 l {2418}. Detailed studies of expression
Prognosis and predictive factors in human development have not been
Breast cancer There have been no systematic studies performed, and only a single study has
The two most commonly occurring can- to indicate whether the prognosis for pa- examined PTEN expression during hu-
cers in Cowden syndrome are carci- tients with Cowden syndrome who have man embryogenesis using a monoclonal
nomas of the breast and thyroid 12414, cancer is different from that of their coun- antibody against the terminal 100 amino
2849f. In the general population, the life- terparts with sporadic cancer. acids of PTEN {842}. The study revealed
time risks of breast and thyroid cancers When activated mTOR signalling was high levels of expression of PTEN protein

Cowden syndrome 315


in the skin, thyroid, and CNS - organs P1(4.5)P approximately 60% of the patients car-
that are affected by the component ne- ried a germline mutation in PTEN {1592}.
oplasias of Cowden syndrome. It also Pl(3,4,5)P Of the 27 familial cases studied, 11 were
revealed prominent expression in the classified as exhibiting true overlap of
developing autonomic nervous system Cowden syndrome and Bannayan-Ri-
and gastrointestinal tract. Early embry- AKT ley-Ruvalcaba syndrome, and 10 of those
onic death in Pten-1- mice also implies 11 had a PTEN mutation. Another 10%
a crucial role for PTEN in early develop- of patients with Bannayan-Riley-Ruval-
ment {581,1992,2463). PTEN is a tumour caba syndrome were subsequently found
mTOR to harbour larger germline deletions of
suppressor and a dual-specificity lipid
phosphatase that plays multiple roles in Migration PTEN {2865). The overlapping mutation
Survival�/
the cell cycle, apoptosis, cell polarity, spectrum, existence of true-overlap fa-
cell migration, and even genomic sta- milial cases, and genotype-phenotype
Angiogenesis Proliferation
bility {1729,2340,2849). The major sub- associations suggest that the presence
strate of PTEN is Pl P 3, which is part of the Fig. 16.25 Schematic representation of the Pl3K/AKT/ of germline PTEN mutation is associated
mTOR signalling pathway. When PTEN is downregulated,
Pl3K pathway (517,767,1488,1563,2411}. with cancer, and strongly suggest that
AKT is upregulated, leading to upregulation of mTOR.
When PTEN is ample and functional, Reprinted from Blumenthal GM and Dennis PA {220). Cowden syndrome and Bannayan-Riley-
PIP3 is converted to PIP2, which results Ruvalcaba syndrome are allelic and part
in hypophosphorylated AKT, a known only looked at the nine exons of PTEN; of a single spectrum at the molecular lev-
cell-survival factor. Hypophosphorylated presumably, further mutations would el. The aggregate term "PTEN hamartoma
AKT is apoptotic. When PTEN is in the have been identified in the promoter or tumour syndrome" was first proposed in
cytoplasm, it predominantly signals via in SDHBISDHD. A single-centre study 1999 {1592) and has since become even
its lipid phosphatase activity down the involving 37 unrelated families affected more apt, now that germline PTEN muta-
Pl3K/AKT pathways (1676). In contrast, by Cowden syndrome (as strictly defined tions have been identified in autism spec-
when PTEN is in the nucleus, it predomi- by the ICC criteria) found a mutation fre- trum disorder with macrocephaly, Proteus
nantly signals via protein phosphatase quency of 80% {1589). Exploratory geno- syndrome, and VATERL association (the
activity down the cyclin-01/MAPK path- type-phenotype analyses showed that co-occurrence of several birth defects)
way, eliciting G1 arrest at least in breast the presence of a germline mutation was with macrocephaly (330,2081,2863). In
and glioma cells {767,768,1488,1676) It associated with a familial risk of devel- one case, the identification of a germline
is also thought that PTEN can dephos- oping malignant breast disease (1589). intragenic PTEN mutation in a patient
phorylate FAK and inhibit integrin and Additionally, missense mutations and/or thought to have juvenile polyposis (1844)
MAPK signalling {892,2501}. mutations of the phosphatase core motif was subsequently considered to exclude
seem to be associated with a surrogate that specific clinical diagnosis; the finding
Genotype/phenotype for disease severity (multiorgan involve- instead suggests a molecular designation
ment). A small study of 13 families with of PTEN hamartoma tumour syndrome
Gene mutations 8 PTEN mutation-positive members did {642,1052,1053,1396,1593,1851 ). This
Approximately 85% of Cowden syn- not show any genotype-phenotype as- conclusion has been further supported by
drome cases, as strictly defined by the sociations (1764}. but this may be due to the identification of germline PTEN muta-
ICC criteria, have a germline mutation the small sample size. tions/deletions in individuals with juvenile
in PTEN. including intragenic mutations. polyps, and of large deletions involving
promoter mutations, and large deletions/ Bannayan-R,!ey-Ruvalcaba syndrome both PTEN and BMPR1A in juvenile poly-
rearrangements {1499,1589,2865). If the Bannayan-Riley-Ruvalcaba syndrome, posis of infancy {571,2469). An important
diagnostic criteria are relaxed, this muta- which is characterized by macrocepha- finding of the polyp-ascertainment study
tion frequency drops to 10-50% {1554, ly, lipomatosis, haemangiomatosis. and was that the reasons for referral listed in
1766,2581). A formal study that ascer- speckled penis, was previously thought the original pathology reports were often
tained 64 unrelated Cowden syndrome- to be clinically distinct, but is now con- incorrect, suggesting that re-review of all
like cases found a mutation frequency sidered likely to be allelic to Cowden polyp histologies by gastrointestinal pa-
of 2% if the criteria were not met, even syndrome (1591). In a combined cohort thologists based in major academic medi-
if the diagnosis was made short of only of 16 sporadic and 27 familial cases of cal centres is a vital step for determining
one criterion (1590). However, this study Bannayan-Riley-Ruvalcaba syndrome, correct genetic etiology (2469).

316 Familial tumour syndromes


Turcot syndrome Cavenee W.K.
Hawkins C.
Leung SY
Van Meir E.G.
Burger PC. Tabori U.

Definition testing for mismatch repair cancer syn- Extraneural mamfestations


When the association of brain tumours drome {2763}. Detection of germline bi- More than 90% of patients present with
with gastrointestinal polyps and cancers allelic mutation in one of the four main cafe-au-lait macules and other dermato-
was originally described, it was called mismatch repair genes is required for the logical abnormalities {113}. These must
Turcot syndrome {2590}. Now, these as- diagnosis of mismatch repair cancer syn- be distinguished from neurofibromatosis
sociations are considered to constitute drome, but the abundance of variants of type 1-related skin lesions. Haemato-
two very distinct cancer syndromes with unknown significance and the technical logical malignancies, predominantly T-
distinct inheritance and cancer spec- problems with sequencing PMS2, which cell lymphoma, occur mostly in the first
trums {1886A}. has multiple pseudogenes, has led to decade, in as many as 30% of patients,
Early-onset colon cancer and gliomas the development of several functional whereas gastrointestinal polyposis and
have also been reported in Li-Fraumeni assays that can aid in rapid detection cancers are present in virtually all pa-
syndrome (see p. 310). of mismatch repair deficiency in urgent tients by the second decade of life. Other
cases. Unlike in Lynch syndrome, micro- cancers (e.g. urinary tract cancers and
satellite instability is not a reliable test for sarcomas) have also been reported {113,
Brain tumour-po/yposis mismatch repair cancer syndrome. Most 2763).
syndrome 1 (BTP1) I Mismatch mutations cause loss of gene expression;
correspondingly, immunohistochemical Gene structure
repair cancer syndrome
staining demonstrates loss of expression The genetic defect underlying mismatch
An autosomal dominant cancer syndrome of the protein encoded by the gene in repair cancer syndrome is the inability to
with reduced penetrance, caused by bi- both tumour and normal tissue in > 90% recognize and repair DNA mismatches
allelic mutations in one of four mismatch of cases {113). Cell-based assays on nor- during replication. Of the components of
repair genes (MLH1, PMS2, MSH2, and mal fibroblasts and lymphoblasts can de- the mismatch repair apparatus, the hu-
MSH6) and hence also called mismatch tect microsatellite instability, resistance to man genes causing mismatch repair can-
repair cancer syndrome. several compounds {230), and failure to cer syndrome are MLH1 at chromosome
Unlike heterozygous carriers (i.e. patients repair G-T mismatches {2353). 3p21.3, MSH2 at 2p16, MSH3 at 5q11-
with Lynch syndrome 1), who develop q13, MSH6 at 2p16, PMS1 at 2q32, and
mostly colon and genitourinary cancers Sites of involvement PMS2 at 7p22. Recognition and repair
as adults, individuals with mismatch re- of base-pair mismatches in human DNA
pair cancer syndrome develop multiple Nervous system neoplasms is mediated by heterodimers of MSH2
brain tumours and other malignancies Brain tumours (most commonly malignant and MSH6, which form a sliding clamp
during childhood {615). Importantly, fam- gliomas) occur in the first two decades of on DNA. The C-terminus of PMS2 inter-
ily history is often uninformative for these life and account for 25-40% of all mis- acts with MLH1, and this complex binds
and Lynch-related cancers. match repair cancer syndrome cancers to MSH2/MSH6 heterodimers to form a
{113,2763}. Many of these brain tumours functional strand-specific mismatch rec-
OMIM number {1624) 276300 have prominent nuclear pleomorphism ognition complex {2366). Cells that are
and multinucleation reminiscent of pleo- deficient in any of the above genes are
Incidence/epidemiology morphic xanthoastrocytoma or giant cell defective in repair of mismatched bases
More than 200 cases of mismatch repair glioblastoma {6.52). Recognition of these and insertions/deletions of single nucleo-
cancer syndrome have been reported features may prompt immunohistochemi- tides, resulting in high mutation rates and
{113,2763} However, this syndrome is cal testing for loss of the mismatch repair microsatellite instability. Unlike in het-
underdiagnosed and highly prevalent in proteins. Oligodendrogliomas, pleomor- erozygous carriers (in whom microsatel-
South Asian and Middle Eastern coun- phic astrocytomas, and other low-grade lite instability is observed in all cancers),
tries, where consanguinity is high. gliomas have also been reported. Medul- cancers originating in patients with bial-
loblastoma and primitive neuroectoder- lelic mismatch repair cancer syndrome
Diagnostic criteria mal tumour have also been described often lack microsatellite instability and
The combination of cafe-au-lait macules; and can include some glial features. Mo- are characterized instead by extremely
consanguinity; and specific brain, haema- lecularly, these cancers have a unique high rates of single-nucleotide mutations
tological, and gastrointestinal cancers, in ultra-hypermutation phenotype, which {113,2353).
particular during childhood, should raise distinguishes them from other childhood
suspicion for mismatch repair cancer tumours {2353). Genotype/phenotype
syndrome. Recently, a scoring system In BTP1, the genotype/phenotype is dif-
was developed for proceeding to genetic ficult to ascertain due to the rarity of the

Turco! syndrome 317


syndrome. Whereas in Lynch syndrome Brain tumour-polyposis as additional cancers, such as osteo-
germline mutations in MLH1 and MSH2 syndrome 2 (BTP2) I Familial mas (in 50-90% of cases}, aggressive
are the most prevalent, in BTP1 muta- fibromatosis (in 10-15%), thyroid cancers
adenomatous polyposis
tions in PMS2 and MSH6 predominate (in 2-3%), and hepatoblastoma (in 1%)
and germline mutations in MSH2 are An autosomal dominant cancer syndrome 11472).
rarely observed. Heterozygous carriers caused by heterozygous mutations in the
are usually unaffected. tumour suppressor gene APC. Gene structure
In one study, the median patient age at The prominent cancers associated with BTP2 results from germline heterozygous
occurrence of glioblastoma in BTP1 I BTP2 are tumours of the gastrointestinal mutations in the tumour suppressor gene
mismatch repair cancer syndrome was tract. The main brain tumour reported in APC 11879). APC is located on chromo-
found to be 18 years (whereas the peak association with BTP2 is medulloblas- some 5q21 and is a major tumour sup-
incidence in the general population oc- toma (rather than gliomas, which are as- pressor in the WNT pathway. Activation
curs in patients aged 40-70 years) sociated with BTP1). of the pathway, most commonly through
{2624). These patients had an average alterations in beta-catenin, is observed in
survival of > 27 months, which is sub- OMIM number 11624} 276300 10-15% of all medulloblastomas /2524).
stantially longer than that of patients with but the association between WNT activa-
sporadic cases (12 month}. Many of the Incidence/epidemiology tion and BTP2-associated medulloblas-
long-term survivors belong to the group BTP2 is responsible for approximately toma is not clear.
of patients with biallelic germline PMS2 1% of all colon cancers. However, brain
mutation, some of whom were still alive tumours, specifically medulloblastoma, Genotype/phenotype
> 10 years after treatment of their gliomas are rare in BTP2, accounting for < 1% of In BTP2, the median patient age for oc-
1554,937,2574). all malignancies in this patient population currence of medulloblastomas was
{1472,2429). 15 years, which matches the patient
Genetic counselling age for the WNT-activated subtype of
Patients with mismatch repair cancer Diagnostic criteria sporadic medulloblastoma, although
syndrome and their family members may The hallmark of BTP2 is the emergence whether these tumours carry the same
benefit from genetic counselling, be- of hundreds to thousands of colonic pol- favourable prognosis as WNT-activated
cause surveillance protocols exist and yps at a young age {2636). medulloblastoma is unclear. Although
early detection may result in increased the numbers are small, in families affect-
survival for both biallelic and heterozy- Sites of involvement ed by familial adenomatous polyposis,
gous carriers (614,2635). The inherent the appearance of medulloblastoma at a
resistance of mismatch repair-deficient Nervous system neoplasms young age in patients with no evidence
cells to several common chemothera- Medulloblastoma is the only brain tumour of polyps may indicate a poor prognosis
pies, including temozolomide, should clearly associated with BTP2. However, 12624).
be considered in the management of although the risk of developing medul-
gliomas in the setting of mismatch repair loblastoma is 90 times as high among Genetic counselling
cancer syndrome. In contrast, the ultra- individuals with BTP2 as in the general BTP2 is a well-characterized syndrome
hypermutation phenotype of mismatch population, this tumour is still rarely ob- with clinical and molecular diagnostic
repair cancer syndrome-related cancers served in BTP2, accounting for < 1% of criteria /2636}. Surveillance protocols ex-
12353) can be exploited by potential ther- all malignancies in this patient population ist and preventive colectomy is required
apies such as immune checkpoint block- {1472) in most patients. Due to the rarity of brain
ade {1441). tumours in BTP2, surveillance does not
Extraneural mamfestations include brain MRI, and no specific thera-
Individuals with BTP2 are at high risk of pies are recommended for BTP2-related
developing. colorectal cancers as well medulloblastoma {1472,2636}.

318 Familial tumour syndromes


Naevoid basal cell carcinoma syndrome Eberhart C.G.
Cavenee W.K.
Pietsch T.

Definition fused ribs, and first-degree relatives with naevoid basal cell carcinoma syndrome
An autosomal dominant disease associ- the syndrome {1284,2331). The minor seem to be exclusively of the desmoplas-
ated with developmental disorders and criteria include medulloblastoma (mainly tic/nodular variants (see Desmoplastic/
predisposition to benign and malignant of the desmoplastic/nodular subtypes nodular medulloblastoma, p. 195, and
tumours, including basal cell carcinomas {67)), ovarian fibroma, macrocephaly, Medulloblastoma with extensive nodu-
of the skin, odontogenic keratocysts, pal- congenital facial abnormalities (e.g. cleft larity, p. 198) {67,787,2296,2376}. It has
mar and plantar dyskeratotic pits, intrac- lip or palate, frontal bossing, and hyper- therefore been proposed that desmo-
ranial calcifications, macrocephaly, and telorism), skeletal abnormalities (e.g. digit plastic medulloblastomas in children
medulloblastomas of the desmoplastic/ syndactyly), and radiological bone ab- aged < 2 years serve as a major criterion
nodular subtypes; caused by germline normalities (e.g. bridging of the sella tur- for the diagnosis of naevoid basal cell
mutations of genes encoding members cica) {67,1284). The diagnosis of naevoid carcinoma syndrome {67,787). The prog-
of the hedgehog signalling pathway, in- basal cell carcinoma syndrome is made nosis of naevoid basal cell carcinoma
cluding the PTCH1 gene on 9q22, its when two or more major or one major and syndrome-associated medulloblastomas
homologue PTCH2 on 1 p34, and the two or more minor criteria are present seems to be better than that of sporadic
SUFU gene on 10q24. {67). The clinical features manifest at dif- cases, and it has been suggested that
ferent points in life. Macrocephaly and rib radiation therapy protocols be adjusted
OMIM number {1624) 109400 anomalies can be detected at birth, and in patients aged < 5 years with naevoid
medulloblastoma typically develops with- basal cell carcinoma syndrome, to pre-
Synonyms in the first 3 years of life. Jaw cysts do not vent the formation of secondary tumours
Naevoid basal cell carcinoma syndrome become evident before the age of about {67,2416).
is also known as Gorlin syndrome, Gor- 10 years, and basal cell carcinomas can
lin-Goltz syndrome, basal cell naevus be detected 10 years later {746A}. Sever-
syndrome, and fifth phacomatosis. al other tumour types have also been re-
ported in individual patients with naevoid
Incidence/epidemiology basal cell carcinoma syndrome, includ-
A prevalence of 1 case per 57 000 pop- ing meningioma, melanoma, chronic
ulation has been reported {657). About lymphocytic leukaemia, non-Hodgkin
1-2% of patients with medulloblastoma lymphoma, ovarian dermoid cyst, and
carry a PTCH1 germline mutation {657). breast and lung carcinoma. However, the
Of 131 children with medulloblastomas, statistical significance of the association
6% had germline SUFU mutations {294) between these neoplasms and naevoid
About 2% of patients with naevoid basal basal cell carcinoma syndrome has yet
cell carcinoma syndrome with germline to be demonstrated (2331}. Radiation
PTCH1 mutations develop medulloblas- treatment of patients with naevoid basal
toma, and the risk is as much as 20% cell carcinoma syndrome, for example,
higher in patients with germline SUFU craniospinal irradiation for the treatment
mutations {2376). Naevoid basal cell of cerebellar medulloblastoma, induces
carcinoma syndrome caused by PTCH2 multiple basal cell carcinomas of the skin
mutations is rare; only two families have as well as various other tumour types
been described {667,747). within the radiation field {405,1813,2416}.

Diagnostic criteria Sites of involvement


Fig. 16.26 The SHH pathway. In normal development,
The most common manifestations of hedgehog signalling is activated by interaction of a
naevoid basal cell carcinoma syndrome Naevo1d basal cell carcinoma secreted hedgehog ligand (Hh) with the multipass
are multiple basal cell carcinomas, as syndrome-associated medulloblastoma transmembrane receptor PTCH. Ligand binding relieves
well as odontogenic keratocysts of the In a recent review of 33 reported me- the repressive effects of PTCH on SMO and permits the
jaw. In one study, basal cell carcinomas dulloblastoma cases associated with activation and nuclear translocation of GLI transcription
and odontogenic keratocysts were found naevoid basal cell carcinoma syndrome, factors. GLI activation is also promoted by FU, and
suppressed by SU(FU). COS2 proteins are thought to
together in > 90% of affected individuals all but one tumour had developed in chil-
serve as a scaffold for these interactions. Once in the
by the age of 40 years {660). Other ma- dren aged < 5 years, and 22 cases (66%) nucleus, GLI factors induce the transcription of various
jor criteria include calcification of the falx had presented in patients aged < 2 years pathway targets, including feedback loops involving
cerebri, palmar and plantar pits, bifid or {67). Medulloblastomas associated with PTCH1 and GL/1.

Naevoid basal cell carcinoma syndrome 319


Other CNS mamfestations
There is no statistically proven evidence
of an increased risk of other CNS neo-
plasms in naevoid basal cell carcinoma
syndrome. Nevertheless, several in-
stances of meningioma arising in pa-
tients with naevoid basal cell carcinoma
syndrome have been reported (40,2331l.
Various malformative changes of the
brain and skull, including calcification of
the falx cerebri and/or tentorium cerebelli
at a young age, dysgenesis of the cor-
pus callosum, congenital hydrocephalus,
and macrocephaly, may occur in affect-
ed family members.

Gene structure
Naevoid basal cell carcinoma syndrome
results from inactivating germline mu-
tations in the human homologue of the
Drosophila segment polarity patched Fig. 16.27 Germline PTCH mutations in 132 patients with naevoid basal cell carcinoma syndrome. Green triangles
gene (PTCH1) on 9q22 (924,1172). its represent nonsense mutations; open circles, splice mutations; purple circles, familial missense mutations; black
homologue PTCH2on 1p34 (667,747l, or triangles, de novo missense mutations; and blue squares, germ line conserved missense mutations. The thick black line
SUFU on 10q (2376l. The detection rate indicates the location of the sterol-sensing domain. Adapted from Lindstrom E et al. (1510).
of specific mutations has increased sig-
nificantly (with as many as 93% of cases (44,2433}. In the absence of ligand, PTC1 found positive in recent years), due to
found positive in recent years), due to the inhibits the activity of SMO (44,2433}. the development of improved methods
development of improved methods of de- Hedgehog signalling takes place in the of detection (746A} The mutations are
tection (746Al. primary cilium (96Al. Binding of hedge- distributed over the entire PTCH1 coding
The PTCH1 gene spans approximately hog proteins to PTC1 can relieve this region, with no mutational hotspots, and
50 kb of genomic DNA and contains inhibition of SMO, allowing its transloca- there seems to be no clear genotype-
2'. 23 exons (924, 1172). with alternative tion to the tip of the primary cilium, which phenotype correlation {2745). Missense
usage of five different first exons (1740l. results in the activation and translocation mutations cluster in a highly conserved
The PTCH2 gene on chromosome 1 p34 of GLI transcription factors into the cell region (the sterol-sensing domain), and
spans approximately 15 kb of genomic nucleus and transcription of a set of spe- particularly in transmembrane domain 4.
DNA and contains 22 coding exons cific target genes controlling the survival, Somatic PTCH1 mutations have been
(2384). The SUFU gene is a human hom- differentiation, and proliferation of pro- demonstrated in various sporadic human
ologue of the Drosophila suppressor of genitor cells. In vertebrates, this pathway tumours (for review, see (1510)), includ-
fused (Sufu) gene. It maps to 10q24 and is critically involved in the development of ing basal cell carcinoma (774,924,1172L
contains 12 exons (888A,1320Al. various tissues and organ systems, such trichoepithelioma (2668). oesophageal
as limbs, gonads, bone, and the CNS squamous cell carcinoma (1565L inva-
Gene expression (868,1092) Germline mutations in the sive transitional cell carcinoma of the
Tissue-specific expression patterns of SHH and PTCH genes have been found bladder (1621). and medulloblastoma
various PTC1 isoforms occur through ex- to cause holoprosencephaly {163,1678A, (1510,1973,2055,2667}. Like germline
tensive splicing events, including mRNA 2161}. PTCH2 encodes a homologue of mutations, the vast majority of mutations
species encoding dominant negative PTCH1, whereas SUFU is located down- detected in sporadic tumours result in
forms of PTC1 (1739,1740,2591}. stream in the hedgehog pathway. SUFU truncations at the protein level. There is
has been found to directly interact with no obvious clustering of mutation sites.
Gene function GLI proteins and is a negative regulator In a study of 68 sporadic medulloblasto-
The PTCH1 gene codes for a 12-trans- of hedgehog signalling {2433Al mas, PTCH1 mutations were exclusively
membrane protein (PTC1) expressed on detected in the desmoplastic variant.
many progenitor cell types. PTC1 func- Genotype/phenotype and not in the 57 tumours with clas-
tions as a receptor for members of the To date, numerous different PTCH1 ger- sic morphology (1973). Consistent with
secreted hedgehog protein family of mline mutations associated with naevoid these data, LOH analyses of sporadic
signalling molecules {1585,2433}. In hu- basal cell carcinoma syndrome have medulloblastomas have demonstrated
mans, this family consists of three mem- been reported {746A,1510}. although frequent allelic loss at 9q22.3-q31 in
bers: SHH, IHH, and DHH. The PTCH1 mutations are not detected in all cases desmoplastic medulloblastomas (in as
gene product has homology to bacterial {1588l. However, the detection rate of many as 50% of cases), but not in classic
transporter proteins (2491} and controls specific mutations has increased sig- medulloblastomas (42,2296}. These data
another transmembrane protein, SMO nificantly (with as many as 93% of cases are consistent with the observation that

320 Familial tumour syndromes


most medulloblastomas associated with {667,747). The SUFU mutations found to Genetic counselling
naevoid basal cell carcinoma syndrome be associated with naevoid basal cell The condition follows an autosomal
are of the desmoplastic/nodular variant carcinoma syndrome are missense mu- dominant pattern of inheritance, with full
types, indicating a strong association tations, splice site mutations, or deletions penetrance but variable clinical pheno-
between desmoplastic phenotype and {1905A,2376}. In 6% of patients with types. The rate of new PTCH1 mutations
pathological hedgehog pathway activa- medulloblastoma, germline SUFU muta- has not been precisely determined. It has
tion {67). PTCH2, a PTCH1 homologue tions were identified, including missense, been estimated that a high proportion
located at chromosome band 1 p34, has nonsense, and splice site mutations lead- (14-81%) of cases are the result of new
also been found to contain somatic muta- ing to a frameshift. Large duplications mutations {660,869,2331,2744). In 4 of
tions in isolated cases of medulloblasto- and deletions were also found, although 6 cases of naevoid basal cell carcinoma
ma and basal cell carcinoma {2384). This these patients do not fulfil the criteria of syndrome with a germline SUFU muta-
gene has been found to have caused naevoid basal cell carcinoma syndrome tion, the mutation was inherited from an
naevoid basal cell carcinoma syndrome (294,2523). Several somatic SUFU muta- unaffected, healthy parent. In the other 2
in two families; one mutation was a trun- tions have also been identified in some cases, the mutation was new {2376).
cating frameshift mutation and the other hedgehog-activated medulloblastomas
was a missense mutation encoding a pro- {1333)
tein that could not inhibit cell proliferation

Rhabdoid tumour predisposition Wesseling P


Biegel JA
syndrome Eberhart C.G.
Judkins A.R.

Definition Incidence/epidemiology childhood, but are occasionally found in


A disorder characterized by a markedly Germline SMARCB1 mutations are es- adults (2063)
increased risk of developing malignant timated to occur in more than one third Other CNS tumours that have been re-
rhabdoid tumours (MRTs), generally due of all patients with atypical teratoid/ ported to be associated with RTPS in-
to constitutional loss or inactivation of rhabdoid tumours (AT/RTs) (i.e. the CNS clude choroid plexus carcinoma {802).
one allele of the SMARCB1 gene - rhab- representative of MRTs) {192,616). Given medulloblastoma, and supratentorial
doid tumour predisposition syndrome this risk, it is important to investigate the primitive neuroectodermal tumour (2327).
1 (RTPS1) - or (extremely rarely) of the SMARCB1 status in all newly diagnosed However, because the histopathologi-
SMARCA4 gene - rhabdoid tumour pre- cases by molecular genetic analysis. cal distinction of these tumours from AT/
disposition syndrome 2 (RTPS2). Individuals with a germline mutation are RTs can be challenging, and because
SMARCB1 has also been identified as a more likely to present with a tumour in the the rhabdoid component may be missed
predisposing gene in familial schwanno- first year of life. due to sampling effects, the occurrence
matosis, and germline mutations in both of such tumours in the context of RTPS is
SMARCB1 and SMARCA4 contribute to Diagnostic criteria controversial {918,1190,1192,25991.
Coffin-Siris syndrome (a rare congenital Demonstration of a germline SMARCB1 SMARCB1 has also been identified as a
malformation syndrome characterized by or SMARCA4 mutation in a patient with gene predisposing individuals to familial
developmental delay or intellectual dis- MRT is sufficient for the diagnosis of schwannomatosis. This gene seems to
ability, coarse facial appearance, feed- RTPS1 or RTPS2, respectively {2293, be involved in about 50% of familial cas-
ing difficulties, frequent infections, and 2409) Children with multiple MRTs or es (but � 10% of sporadic cases) (260,
hypoplasia/aplasia of the fifth fingernails with affected siblings or other relatives 921,2191). SMARCB1 germline mutations
and fifth distal phalanges). are almost certain to be affected by are also reported to predispose individu-
RTPS themselves. als to the development of multiple menin-
OMIM numbers (1624) giomas, with preferential location of the
RTPS1 609322 Sites of involvement cranial meningiomas at the falx cerebri
RTPS2 613325 {102,455,2622].
Nervous system neoplasms A spectrum of CNS tumours, includ-
Synonyms Individuals with RTPS1 often present with ing meningiomas, gliomas, melanomas,
Rhabdoid tumour predisposition syn- AT/RT (p. 209). Patients with germline and carcinomas, may show rhabdoid
drome (RTPS) is also known as rhabdoid mutations or deletions of SMARCB1 may features. Generally, such so-called com-
predisposition syndrome and familial develop isolated AT/RTs or an AT/RT with posite rhabdoid tumours retain nuclear
posterior Iossa brain tumour syndrome of a synchronous renal or extrarenal MRT SMARCB1 staining (1941). strongly sug-
infancy. {1454) AT/RTs generally occur in early gesting that they do not contain the same

Rhabdoid tumour predisposition syndrome 321


genetic alterations as classic MRT and Gene structure and expression lytic subunit of the SWI/SNF complex
are therefore unlikely to be part of RTPS. The SMARCB1 gene was the first subu- was the second member of this comple;
nit of the SWI/SNF complex found to be reported to be involved in a cancer pre-
Extraneural mamfestations mutated in cancer. This gene is located disposition syndrome {193,2293l. More
By far the most frequent extra-CNS lo- in chromosome region 22q11.23 and recently, other genes encoding SW!/
cation of MRT is the kidney. Bilateral re- contains nine exons spanning 50 kb of SNF subunits have also been identified
nal MRTs are almost always associated genomic ONA 12649}. Alternative splic- as recurrently mutated in cancer. Collec-
with a germline SMARCB1 mutation, but ing of exon 2 results in two transcripts tively, 20% of all human cancers contain
infants with an isolated MRT may carry and two proteins with lengths of 385 and a SWI/SNF mutation, and because most
germline mutations as well. Occasionally, 376 amino acid residues, respectively. of these tumours are not classic MRTs,
MRTs have been reported to originate in The so-called SNF5 homology domain it can be expected that the definition of
the head and neck region, paraspinal in the second half of the protein har- RTPS will need adjustment in the near fu-
soft tissues, heart, mediastinum, and bours highly conserved structural motifs ture {1276l.
liver {2737). In a child surviving a tho- through which SMARCB1 interacts with
racic MRT, a conventional chondrosar- other proteins l2430l. The SMARCB1 Gene mutations
coma developed in the mandibula. The protein is a core subunit of mammalian The types of SMARCB1 mutations ob-
chondrosarcoma showed deletion of one SWI/SNF chromatin remodelling com- served in sporadic MRTs are similar to
SMARCB1 allele and a premature stop plexes. which regulate the expression the spectrum of germline mutations re-
codon in the remaining allele {723l. In of many genes by using ATP for sliding ported to date. However, single base
a patient with schwannomatosis, a leio- the nucleosomes along the ONA helix, deletions in exon 9 occur most often in·
myoma of the cervix uteri was reported facilitating or repressing transcription AT/RTs in patients without detectable
to display the genetic features that are l2760l The SMARCB1 protein functions germline alterations 1192,616}. The sec-
characteristic of germline SMARCB1 as a tumour suppressor via repression of ond inactivating event is most frequently
mutation-associated tumours 11062). CCN01 gene expression, induction of the a deletion of the wildtype allele, often
SMARCB1 mutations may occasion- CDKN2A gene, and hypophosphoryla- due to monosomy 22. In familial cases
ally underlie the oncogenesis of other tion of retinoblastoma protein, resulting in of RTPS, unaffected adult carriers have
neoplasms, such as the proximal type of GO/G1 cell cycle arrest {185,2136l Loss been identified. Alternatively, new muta-
epithelioid sarcoma l1697l, but to date, of SMARCB1 leads to transcriptional ac- tions can occur during oogenesis/sper-
these sarcomas have not been described tivation of EZH2 and to repression and matogenesis (gonadal mosaicism), or
in association with RTPS. increased H3K27me3 of polycomb gene postzygotically during the early stages·
In a family with a SMARCA4 germline targets as part of the broader SWI/SNF of embryogenesis {616,962,1133,2327}.
mutation, AT/RT and ovarian cancer were modulation of the polycomb complex to Compared with germline SMARCB1 mu-
diagnosed in a newborn and his mother, maintain the epigenome. The Hippo sig- tations in patients with rhabdoid tumours,
respectively, providing a link between AT/ nalling pathway is involved in the detri- schwannomatosis mutations are signifi-
RT and small cell carcinoma of the ovary mental effects of SMARCB1 deficiency, cantly more likely to occur at either end
of hypercalcaemic type l2766l After a and its main effector (YAP1) is overex- of the gene and to be non-truncating
subsequent study of three families with pressed in AT/RT 11145,2761). According mutations l2380l. One study reported a
small cell carcinoma of the ovary of hy- to the tumour suppressor gene model, family in which the affected individuals
percalcaemic type revealed deleterious both copies of the SMARCB1 gene are inherited a SMARCB1 mutation and de-
SMARCA4 mutations in all cases. it was inactivated in these tumours. veloped schwannomatosis or rhabdoid
suggested that these tumours should be The SMARCA4 gene, located on chro- tumour 12472).
renamed "MRT of the ovary" {728l. mosome 19p13.2 and encoding a cata-

322 Familial tumour syndromes


CHAPTER17
Tumours of the sellar region
Craniopharyngioma
Granular cell tumour of the sellar region
Pituicytoma
Spindle cell oncocytoma
,

Craniopharyngioma Buslei R.
Rushing E.J.
Paulus W.
Burger P.C.
Giangaspero F. Santagata S

Adamantinomatous Papillary
Definition 90-100 such as the sphenoid sinus {1338) and
A histological/y benign, partly cystic epi- 80-89
cerebellopontine angle {1277) have also
70-79
thelial tumour of the sellar region presum- been reported. Craniopharyngiomas,
60-69
ably derived from embryonic remnants 50-59
mainly the papillary variant, are also
of the Rathke pouch epithelium, with two 40-49 found in the third ventricle {507).
clinicopathological variants (adamantino- 30-39

matous and papillary) that have distinct 20-29 Clinical features


phenotypes and characteristic mutations. 10-19
The clinical features are non-specific
0-9
Adamantinomatous craniopharyngiomas 10
and include visual deficits (observed in
50 40 30 20 10 0 0 20 30 40 50
show CTNNB1 mutations and aberrant Number of cases Number of cases 62-84% of patients; more frequently in
nuclear expression of beta-catenin in as Fig. 17 .01 Age distribution of adamantinomatous and adults than in children) and endocrine
many as 95% of cases. Papillary crani- papillary craniopharyngioma, based on 224 cases. deficiencies (observed in 52-87% of pa-
opharyngiomas show BRAF V600E mu- tients; more frequently in children) {13).
tations in 81-95% of cases, which can incidence of 3.8 cases per 1 million chil- The endocrine disturbances observed
be detected by immunohistochemistry. dren {1572). They are the most common include deficiencies of growth hormone
Craniopharyngiomas may be infiltra- non-neuroepithelial intracerebral neo- (occurring in 75% of cases), luteinizing
tive and therefore clinically difficult to plasm in children, accounting for 5-11% hormone I follicle-stimulating hormone (in
manage. of intracranial tumours in this age group 40%), adrenocorticotropic hormone (in
Xanthogranuloma of the sellar region is {846,2174). 25%), and thyroid-stimulating hormone
a related but distinct clinicopathological (in 25%). Diabetes insipidus is noted in
entity. Age and sex distnbution as many as 17% of children and 30% of
Adamantinomatous craniopharyngioma adults. Cognitive impairment and per-
ICD-0 code 9350/1 has a bimodal age distribution {1784, sonality changes are observed in about
2836). with incidence peaks in children half of all patients {13). Obesity and hy-
Grading aged 5-15 years and adults aged 45- perphagia (signs of hypothalamic dys-
Craniopharyngiomas correspond histo- 60 years. Rare neonatal and fetal cas- function) have been described {1730,
logically to WHO grade I. es have been reported {431). Papillary 2362). although the occurrence of severe
craniopharyngiomas occur almost exclu- obesity can be reduced with resections
Epidemiology sively in adults, at a mean patient age of that spare the hypothalamus {636). Signs
40-55 years {507). Craniopharyngiomas of increased intracranial pressure are fre-
Incidence show no obvious sex predilection. quent, especially in cases with compres-
Craniopharyngiomas constitute 1.2- sion or invasion of the third ventricle.
4.6% of all intracranial tumours, account- Localization
ing for 0.5-2.5 new cases per 1 mil- The most common site for both subtypes Imaging
lion population per year {310). They are is the suprasellar cistern, with a minor in- Adamantinomatous craniopharyngiomas
more frequent in Japan, with an annual trasellar component. Unusual locations present as lobulated, multicystic masses

Fig. 17.02 Adamantinomatous craniopharyngioma. A Sagittal postcontrast T1-weighted MRI shows a large, Fig. 17.03 Papillary craniopharyngioma. This sagittal
enhancing, partially cystic suprasellar and sellar mass, characteristic of adamantinomatous craniopharyngioma. postcontrast T1-weighted MRI shows an enhancing
B Large adamantinomatous craniopharyngioma extending into the third ventricle. Postmortem X-ray showed extensive cystic mass involving the anterior third ventricle; the solid
calcification, which is typical for this craniopharyngioma variant. component shows papillary fronds.

324 Tumours of the sellar region


throughout the tumour epithelial cells
{2307). Papillary craniopharyngiomas
show more robust membranous expres-
sion of claudin-1 than do either their ada-
mantinomatous counterparts or Rathke
cleft cysts {2410).

Proliferation
Ki-67 immunoreactivity is concentrated
along the peripherally palisading cells in
the adamantinomatous type, and is more
randomly distributed in papillary lesions
{613,2058). The reported Ki-67 prolifera-
tion index varies considerably from case
to case, and is higher than might be ex-
pected given the relative indolence of
the neoplasms {613,2058). No consistent
Fig. 17.04 Adamantinomatous craniopharyngioma Fig. 17.05 Adamantinomatous craniopharyngioma. relationship between proliferation index
extending towards the cerebral peduncles; note the so- Fresh tumour material showing an uneven surface with and recurrence has been established.
called machine-oil appearance of the dorsal portion and small calcifications and fiakes of wet keratin (white
calcifications. deposits). Ultrastructure
Electron microscopy is seldom needed,
CT shows contrast enhancement of the Spread given the relatively typical features in
solid portions and of the cyst capsule, as Dissemination in the subarachnoid space most cases. In addition to glycogen and
well as typical calcifications. On MRI, the or implantation along the surgical track or the usual organelles, the constituent epi-
cystic areas are T1-hyperintense, where- path of needle aspiration is a rare compli- thelial cells contain tonofilaments joined
as the solid components and the mural cation {190,1450,1473,2165,2287). by desmosomes. Fenestrated capillary
nodules are T1-isointense, with a slightly endothelium, amorphous ground ma-
heterogeneous quality. On enhanced lmmunophenotype trix, and collagen fibrils characterize the
MRI images, the cystic portion is isoin- The tumour cells immunolabel with an- connective tissue strorna. Mineral pre-
tense with ring enhancement, whereas tibodies against pancytokeratin, CK5/6, cipitates appear to arise in membrane-
the solid components are hyperintense CK?, CK14, CK17, CK19, EMA, claudin-1 bound vesicles {24).
{2183). Adamantinomatous tumours may and beta-catenin. Only the adamantino-
superficially infiltrate neighbouring brain matous variant shows aberrant nuclear Differential diagnosis
and adhere to adjacent blood vessels accumulation of beta-catenin, especially Xanthogranulomas of the sellar region
and nerves. in the whorl-like cell clusters along the are histologically composed of cholester-
Papillary craniopharyngiomas are typi- tumour margin and in finger-like tumour ol clefts, macrophages (xanthoma cells),
cally non-calcified, solid lesions with a protrusions {1028). Papillary craniophar- multinucleated giant cells, chronic inflam-
more uniform appearance on CT and yngiomas harbour BRAF V600E muta- mation, necrotic debris, and haemosid-
MRI {507,2245). tions that can be detected by immuno- erin deposits {1914). Xanthogranuloma
histochemistry, with uniform staining of the sellar region is considered to be a

� . -
Fig. 17.06 Adamantinomatous craniopharyngioma. The distinctive epithelium features loose microcystic areas known
as stellate reticulum, whorls, basal palisading, and anucleate nests of pale, squamous ghost cells known as wet keratin.

Cranropharynqiorna 325
.
,����-r-. . -.-ea.���••-.��
...

., ,, , • ·-
•• , • . .. ..

- ". •••
•• •
.1• •

> : •
!·� •
...
Fig. 17.08 Adamantinomatous craniopharyngioma. A Finger-like tumour protrusions in the surrounding brain tissue. B Cell groups of an adamantinomatous craniopharyngioma in
the surrounding brain tissue, in which a distinct piloid gliosis with abundant Rosenthal fibres is evident.

Fig. 17.09 Adamantinomatous craniopharyngioma. A lmmunostaining for CK5/6 highlights foci of squamous differentiation, including foci of wet keratin. B Only focal immunoreactivity
for claudin-1. C Finger-like protrusions in the surrounding brain tissue harbouring cell clusters with aberrant nuclear accumulation of beta-catenin.

reactive lesion, most often to remnants of Rathke cleft cysts, beta-catenin local- craniopharyngiomas and Rathke cleft
Rathke cleft cyst (64). Foci of squamous izes to the cell membrane, whereas the cysts, as well as reports of unique con-
or cuboidal epithelium as well as small nuclear accumulation described in ada- genital craniopharyngiomas with amelo-
tubules may be encountered, whereas mantinomatous craniopharyngiomas is blastic, tooth bud, and adenohypophy-
typical areas of adamantinomatous epi- typically absent (1028}. seal primordia components (60,1721,
thelium are usually absent or amount to 2317,2807). Additional evidence is the
< 10% of the tissue (1914}. Epithelial cells Cell of origin report of a tumour arising from a Rathke
in xanthogranulomas do not exhibit nu- Several observations, including cytoker- cleft cyst that contained cells that were
clear accumulation of beta-catenin (329). atin expression profiles, indicate that transitional between squamous, mucus-
Epidermoid and Rathke cleft cysts craniopharyngiomas arise from neoplas- producing, and anterior pituitary lobe se-
are sometimes raised in the differen- tic transformation of ectodermal-derived cretory cells (1246}
tial diagnosis, especially in small tissue epithelial cell remnants of Rathke pouch The hypothesis that craniopharyngiomas
fragments. Epidermoid cysts are distin- and the craniopharyngeal duct. Epithelial are of neuroendocrine lineage is support-
guished by the presence of a uniloculat- cell rests have been reported to occur ed by the finding that scattered tumour
ed cavity lined by squamous epithelium between the roof of the pharynx and the cells can (rarely) express pituitary hor-
and filled with flaky, dry keratin. Rathke floor of the t�ird ventricle, most frequently mones {2475). chromogranin-A (2807).
cleft cysts enter into the differential diag- along the anterior infundibulum and the and hCG (2486}. The finding that crani-
nosis in particular when they show exten- anterior superior surface of the adenohy- opharyngiomas share stem cell markers
sive squamous metaplasia (2307). More pophysis - sites of the previous Rathke (e.g. SOX2, OCT4, KLF4, and SOX9) with
commonly, the cyst wall is lined by simple pouch and involuted duct that links these the normal pituitary gland further sup-
columnar or cuboidal epithelium, which structures. Metaplasia of cells derived ports a common origin {70,785).
often is ciliated, with mucinous goblet from the tooth primordia determine the
cells. A respiratory-type epithelium may adamantinomatous subtype, whereas Genetic profile
occasionally be present, accompanied metaplastic changes in cells derived The WNT signalling pathway is strongly
by a xanthogranulomatous reaction af- from buccal mucosa primordia give implicated in the pathogenesis of ada-
ter rupture of the cyst wall (932,2573, rise to the squamous papillary variant mantinomatous craniopharyngiomas.
2837). Unlike papillary craniopharyngio- (20141. Further support for the origin of Genetic analyses have confirmed that as
mas, Rathke cleft cysts lack BRAFV600E craniopharyngiomas from Rathke pouch many as 95% of the tumours show muta-
mutations, although cross-reactive stain- epithelium is the occasional occurrence tions in exon 3 of the beta-catenin gene
ing of cilia can be seen {1180,2307). In of mixed tumours with characteristics of (CTNNB1) (263,329,1231,2316}. These

326 Tumours of the sellar region


mutations within the degradation target- is significantly higher {1922,2806}; how- Macroscopy
ing box of beta-catenin lead to activation ever, there is a trend towards less radical Adamantinomatous craniopharyngioma
of the WNT signalling pathway, indicated extirpation in order to avoid hypothalamic typically presents as a lobulated solid
by aberrant cytoplasmic and nuclear ac- injury {1730}. mass, but on closer inspection often
cumulation of beta-catenin protein and Radiotherapy is widely used in incom- demonstrates a spongy quality as a re-
respective target gene activation {1077l. pletely resected tumours. Histological sult of a variable cystic component. On
Mouse models confirm the tumour-initiat- evidence of brain invasion, which is more sectioning, the yellowish-white cysts
ing strength of these alterations {790l. frequently documented in the adamanti- may contain dark greenish-brown liq-
In contrast, papillary craniopharyngio- nomatous type than in the papillary type, uid resembling machinery oil. The gross
mas harbour the BRAF V600E mutation does not correlate with a higher recur- appearance also reflects secondary
in nearly all cases {263,1432,2307l, The rence rate in cases with gross surgical changes such as fibrosis, calcifications,
apparent mutual exclusivity of CTNNB1 resection {2721 l. Some authors have ossification, and the presence of choles-
and BRAF V600E mutations in both documented a better prognosis for the terol-rich deposits.
craniopharyngioma variants demon- papillary variant {13,2520}. whereas oth-
strates that these histological categories ers found no significant differences {507, Microscopy
can be defined by their underlying mo- 2721 }. Malignant transformation of crani- Adamantinomatous craniopharyngiomas
lecular genetics. opharyngioma to squamous carcinoma can be recognized by the presence of
Comparative genomic hybridization stud- after multiple recurrences and irradiation well-differentiated epithelium disposed
ies on two large series of craniopharyn- is exceptional {2155,2388}. in cords, lobules, nodular whorls, and ir-
giomas have failed to show significant regular trabeculae bordered by palisad-
recurrent chromosomal imbalances in ei- ing columnar epithelium. These islands
ther adamantinomatous or papillary vari- Adamantinomatous of densely packed cells merge with
ants {2120,2829l. In contrast, a compara- craniopharyngioma loosely knit epithelium known as stellate
tive genomic hybridization-based study reticulum. Pale nodules of wet keratin
of nine adamantinomatous craniophar- Definition constituting anucleate ghost-like rem-
yngiomas revealed at least one genomic A craniopharyngioma characterized by nants of squamous cells may be found
alteration in 67% of cases {2129}. Simi- a distinctive epithelium that forms stel- in both the compact and looser areas.
larly, cytogenetic analysis of two cases late reticulum, wet keratin, and basal Cystic cavities containing cell debris and
revealed multiple chromosomal abnor- palisades, showing CTNNB1 mutations fibrosis are lined by flattened epithelium.
malities on chromosomes 2 and 12 {871, and aberrant nuclear expression of beta- Lymphocytic infiltrates are frequent and
1224l. catenin in as many as 95% of cases. giant cell-rich granulomatous inflam-
mation may be associated with choles-
Prognosis and predictive factors ICD-0 code 9351/1 terol clefts, although this is more typical
In several large series, at 10 years of of xanthogranuloma. Piloid gliosis with
follow-up, 60-93% of patients were re- Epidemiology abundant Rosenthal fibres is often seen
currence free and 64-96% were alive Adamantinomatous craniopharyngioma in the surrounding brain and should not
{507,2065,2806}. The most significant has a bimodal age distribution {1784, be mistaken for pilocytic astrocytoma.
factor associated with recurrence is the 2836}. with incidence peaks in children Rare examples of malignant transforma-
extent of surgical resection {1922,2721, aged 5-15 years and adults aged 45- tion, especially after multiple recurrences
2806}. with lesions > 5 cm in diameter 60 years. Rare neonatal and fetal cases and radiotherapy, have been reported
carrying a markedly worse prognosis in have been reported {431}. {1097,2155,2388l. but are extremely
an earlier series {2806l. After incomplete rare. Therefore, other diagnoses (e.g.
surgical resection, the recurrence rate

Craniopharyngioma 327
Fig. 17.11 Papillary craniopharyngioma. A lmmunostaining for p63 confirms that the great majority of this tumour is composed of squamous epithelium. B Small component of
surface respiratory epithelium is highlighted with CK? staining; this feature overlaps with the lining of Rathke cleft cyst, which can also show squamous metaplasia, mimicking papillary
craniopharyngioma. C Robust immunoreactivity for claudin-1. D Physiological expression of beta-catenin at the tumour cell membranes.

sinonasal carcinoma) should be carefully of cases), which can be detected by cholesterol-rich machinery oil-like fluid
excluded. immunohistochemistry. or calcifications. The surface of papil-
lary craniopharyngioma, like that of other
ICD-0 code 9352/1 papillary tumours, may appear corrugat-
Papillary craniopharyngioma ed or cauliflower-like.
Epidemiology
Definition Papillary craniopharyngiomas occur al- Microscopy
A papillary, mostly supratentorial or third most exclusively in adults, with a mean The essential features of papillary crani-
ventricular craniopharyngioma charac- patient age of 40-55 years {507). and opharyngioma include compact, mono-
terized by fibrovascular cores lined by show no obvious sex predilection. morphic sheets of well-differentiated
non-keratinizing squamous epithelium. squamous epithelium without surface
Papillary craniopharyngiomas occur al- Macroscopy keratinization. This variant typically lacks
most exclusively in adults and frequently Papillary • craniopharyngiomas are calcifications, picket fence-like pali-
show BRAFV600E mutations (in 81-95% solid or rarely cystic tumours, without sades, whorl-like cell nodules, and wet
keratin. T cells, macrophages, and neu-
trophils are scattered throughout the
fibrovascular cores and tumour epithe-
lium. Rudimentary papillae may surround
the fibrovascular cores. Rarely, ciliated
epithelium and periodic acid-Schiff-
positive goblet cells are encountered.

Fig. 17.12 Papillary craniopharyngioma. A Typical non-keratinizing squamous epithelium and focal lymphocytic
infiltrate. B V600E-mutant BRAF is consistently expressed.

328 Tumours of the sellar region


Granular cell tumour of the Fuller G.N.
Brat D.J.
sellar region Wesseling P.
Roncaroli F.

Definition
A circumscribed tumour that is composed
of large epithelioid to spindled cells with
distinctively granular, eosinophilic cyto-
plasm (due to an abundance of intracyto-
plasmic lysosomes) and that arises from
the neurohypophysis or infundibulum.
Granular cell tumour of the sellar region
generally exhibits slow progression and a
benign clinical course. Like pituicytomas
and spindle cell oncocytomas, granular
cell tumours show nuclear expression
of TTF1, suggesting that these three tu-
mours may constitute a spectrum of a
single nosological entity.
Fig. 17.14 Granular cell tumour of the sellar region. Postcontrast T1-weighted MRI. The sagittal plane (A) and coronal
ICD-0 code 9582/0 plane (B) show prominent contrast enhancement. Note the characteristic sellar/suprasellar anatomical location.

Grading Asymptomatic microscopic clusters of Other presenting signs and symptoms


Granular cell tumours correspond histo- granular cells, called granular cell tumou- include panhypopituitarism, galactor-
logically to WHO grade I. rettes 12330) or tumourlets 11547). are rhoea, amenorrhoea, decreased libido,
more common than larger, symptomatic and neuropsychological changes. Dia-
Synonyms tumours, and have been documented at betes insipidus has been reported but
Synonyms previously applied to granular incidence rates as high as 17% in post- is relatively uncommon 1479). Symptoms
cell tumour include: Abrikossoff tumour, mortem series 11547,2330,2564} usually develop slowly over a period of
choristoma, granular cell myoblastoma, years, although acute presentation with
and granular cell neuroma. Localization sudden-onset diplopia, confusion, head-
Granular cell tumours arise along the ache, and vomiting can occur 1479).
Epidemiology anatomical distribution of the neurohypo- There are no disease-specific signs or
Symptomatic granular cell tumours are physis, including the posterior pituitary symptoms that reliably distinguish gran-
relatively rare and typically present in and pituitary stalk I infundibulum. The ular cell tumours from other suprasellar
adulthood, with only exceptionally rare tumours exhibit a preference for the pi- mass lesions. Several cases have been
childhood cases 1166). There is a clear tuitary stalk and thus most frequently found in association with pituitary adeno-
female predominance, with a female-ta- arise in the suprasellar region, but may ma 1131,479,1527,2564}.
male ratio of > 2:1. Peak incidence oc- also arise from the posterior pituitary and
curs in the sixth decade of life in men present as an intrasellar mass; some ex- Imaging
and in the fifth decade of life in women. amples occupy both the intrasellar and MRI typically shows a well-circumscribed
suprasellar compartments, mimicking suprasellar mass that most commonly
pituitary macroadenoma. Granular cell displays homogeneous or heteroge-
Females tumours with morphological and immu- neous contrast enhancement. Tumour
90-100

80-89 nophenotypic features identical to those size most often ranges from 1.5 to 6.0 cm
70-79 of neurohypophyseal tumours have rarely 1479). Calcification is unusual and thus
60-69
been reported in other anatomical loca- helps to distinguish granular cell tumours
50-59
40-49
tions within the CNS, including the spi- from craniopharyngiomas. Similarly, a
30-39 nal meninges 11586). cranial meninges lack of a dural attachment (dural tail) and
20.29 12631). and third ventricle 12597). the anatomical location centred on the
10-19 pituitary stalk help to distinguish granular
0-9
10
Clinical features cell tumours from most regional meningi-
15 10 15
Number of cases Number of cases
The most common presenting symptom omas. In addition to suprasellar presen-
Fig. 17.13 Age and sex distribution of neurohypophyseal of granular cell tumour of the sellar re- tations, intrasellar and intrasellar/supra-
granular cell tumours, based on 66 symptomatic cases gion is visual field deficit secondary to sellar presentations are also recognized.
published in the literature; with a 1 :2.3 male-to-female ratio. compression of the optic chiasm 1479). Although there are no pathognomonic

Granular cell tumour of the sellar region 329


imaging features, cases in which the architecture is typically nodular; sheets membranous debris. A few other orga-
tumour can be clearly seen to be sepa- and/or spindled/fascicular patterns can nelles and intracytoplasmic filaments
rated from the pituitary by the inferior end also be seen, occasionally in a whorling may be observed, but neurosecretory
of the pituitary stalk are suggestive of pattern. Periodic acid-Schiff staining granules are absent (1521.
granular cell tumour {86). Nevertheless, of cytoplasmic granules is resistant to
due to the relative rarity of the tumour, diastase digestion. Small foci of foamy lmmunophenotype
the diagnosis is rarely anticipated prior to cells may be observed. The tumour cell Granular cell tumours are variably posi-
surgical resection, which is also the case nuclei are small, with inconspicuous nu- tive for CD68 (by KP1 staining), S100
for suprasellar pituicytomas. cleoli and evenly distributed chromatin. protein, alpha-1-antitrypsin, alpha-1-anti-
Perivascular lymphocytic aggregates chymotrypsin, and cathepsin B, and are
Macroscopy are common. Mitotic activity is usually negative for NFPs, cytokeratins, chro-
The tumours are usually lobulated and inconspicuous, and proliferative activ- mogranin-A, synaptophysin, desmin,
well circumscribed, with a soft but rub- ity is usually very low. Some lesions are SMA, and the pituitary hormones. Most
bery consistency firmer than that of pitui- characterized by nuclear pleomorphism, tumours are negative for GFAP, although
tary adenoma. The cut surface is typically prominent nucleoli, multinucleated cells, variable immunoreactivity has been not-
grey to yellow. Necrosis, cystic degener- and increased mitotic activity (with as ed in a subset of granular cell tumours.
ation, and haemorrhage are uncommon. many as 5 mitoses per 10 high-power Granular cell tumours show nuclear
The tumour may infiltrate surrounding fields and a Ki-67 proliferation index of staining for TTF1 (1452).
structures, such as the optic chiasm and 7%). Such tumours have been referred to
cavernous sinus; these features may pre- as atypical granular cell tumours by some Cell of origin
vent gross total surgical resection. authors, although the clinical and biologi- The finding that granular cell tumours
cal significance is uncertain {1230,2659). strongly express the nuclear transcrip-
Microscopy By electron microscopy, the cytoplasm tion factor TTF1, like pituicytes of the de-
Granular cell tumours consist of densely of the granular tumour cells is filled with veloping and mature neurohypophysis,
packed polygonal cells with abundant phagolysosomes containing unevenly suggests a pituicyte derivation (1452,
granular eosinophilic cytoplasm. The distributed electron-dense material and 1654). Both pituicytomas and spindle

330 Tumours of the sellar region


cell oncocytomas of the hypophysis neurohypophysis (1654,2499) Granular R132H-mutant IDH1, V600E-mutant
also express nuclear TTF1, suggesting cell tumours occasionally occur in the BRAF, or KIAA1549-BRAFfusion (1654).
a histogenesis from pituicytes for these CNS outside the pituitary gland (e.g. in
tumours as well. It has been speculated the meninges, cerebral hemispheres, Prognosis and predictive factors
that granular cell tumours and spindle third ventricle, or cranial nerves); these Most granular cell tumours are clinically
cell oncocytomas constitute pituicytomas tumours may be derived from glial cells, benign, with slow progression and lack of
that are composed of tumour cells with Schwann cells, or macrophages {479, invasive growth. Surgical removal is the
lysosome-rich and mitochondrion-rich 2118,2631 }. preferred therapy for larger tumours, but
cytoplasm, respectively, giving rise to the firm and vascular nature of pituitary
distinct morphologies (1301,1452,1654}. Genetic profile granular cell tumours, sometimes com-
The derivation of three morphologically A unique genetic signature for granu- bined with the lack of an obvious dissec-
distinct tumours from pituicytes might lar cell tumour has not been defined. tion plane from the adjacent brain, may
be explained by the existence of multi- The limited number of cases that have hamper gross total resection.
ple subtypes of pituicytes in the normal been tested to date have not shown

Granular cell tumour of the sellar region 331


Pituicytoma Brat D.J.
Wesseling P.
Fuller G.N.
Roncaroli F.

Definition ages of 40 and 60 years. The male-to-


A circumscribed and generally solid female ratio is 1.5:1 {504}.
low-grade glial neoplasm that originates
in the neurohypophysis or infundibulum Localization
and is composed of bipolar spindled Pituicytomas arise along the distribution
cells arranged in a fascicular or storiform of the neurohypophysis, including the in-
pattern. fundibulum I pituitary stalk and posterior
Like spindle cell oncocytomas and gran- pituitary. Accordingly, they may be locat-
ular cell tumours of the sellar region, ed in the sella, in the suprasellar region,
pituicytomas show nuclear expression or in both the intrasellar and suprasellar
of TTF1, suggesting that these three tu- compartments. Of these possibilities,
mours may constitute a spectrum of a purely intrasellar localization is the least
single nosological entity. common {504}.
Fig. 17.18 Pituicytoma showing solid, circumscribed
growth and diffuse contrast enhancement on T1-
ICD-0 code 9432/1 Clinical features weighted MRI.
The most common presenting signs and
Grading symptoms of pituicytoma are similar to
Pituicytomas correspond histologically to those of other slowly expansive non- Imaging
WHO grade I. hormonally active tumours of the sellar/ Pituicytomas are typically solid and cir-
suprasellar region that compress the op- cumscribed mass lesions of the sellar
Synonyms and historical annotation tic chiasm, infundibulum, and/or pituitary and suprasellar spaces. Most often,
In the past, the term "pituicytoma" was gland. These include visual disturbance; they are isointense to grey matter on T1-
used loosely for several histologically headache; and features of hypopituita- weighted images, hyperintense on T2-
distinct tumours of the sellar and supra- rism such as fatigue, amenorrhoea, de- weighted images, and uniformly contrast-
sellar region (e.g. granular cell tumours creased libido, and mildly elevated se- enhancing {504). Occasional tumours
and pilocytic astrocytomas). Since the rum prolactin (the stalk effect) {504,687, show heterogeneous contrast enhance-
publication of the more restricted defini- 2877}. Rarely, asymptomatic cases are ment, and rare examples have a cystic
tion of pituicytoma by Brat et al. {273} in found only at autopsy. component {273}.
2000 and its inclusion in the 2007 WHO
classification, the term has instead been
reserved for low-grade glial neoplasms
that originate in the neurohypophysis
or infundibulum and are histologically
distinct {270,273}. Synonyms that were
used in the past for pituicytoma include
"posterior pituitary astrocytoma" and "in-
fundibuloma".

Epidemiology
Pituicytomas are rare. Since 2000, ap-
proximately 70 examples have been
described in the literature, often in case
reports and small series {504,687,2877}.
The largest series reported to date con-
tains 9 tumours pooled from the consulta-
tion cases of two large institutions {273}.
Nearly all reported pituicytomas have
occurred in adult patients, with an aver-
age patient age at diagnosis of 50 years
(range: 7-83 years) {504}. Nearly two
thirds of all patients present between the
Fig. 17.19 Pituicytoma. Elongate and plump tumours cells arranged in a fascicular pattern.

332 Tumours of the sellar region


Fig. 17.20 Pituicytoma. A Patchy staining for GFAP. B Diffuse staining for S100 protein.

Macroscopy bodies (axonal dilatations for neuropep- supports a pituicyte derivation {1452,
Pituicytomas are solid and circumscribed tide storage in the neurohypophysis). 1654). Both granular cell tumours of the
masses that can measure up to several sellar region and spindle cell oncocy-
centimetres and have a firm, rubbery tex- lmmunophenotype tomas also express nuclear TTF1, sug-
ture. Only rarely has a cystic component Pituicytomas are low-grade gliomas that gesting a histogenesis from pituicytes for
been reported {273,2596l. Radiographi- are positive by immunohistochemistry these tumours as well. It has been specu-
cal studies may give the impression of a for vimentin and S100 protein and show lated that these granular cell tumours and
smoothly contoured tumour, but pituicy- nuclear staining for TTF1 {273,1452,1654, spindle cell oncocytomas constitute pi-
tomas can be firmly adherent to adjacent 1966l. GFAP staining is highly variable, tuicytomas that are composed of tumour
structures in the suprasellar space. ranging from faint and focal to moderate cells with lysosome-rich and mitochon-
and patchy, and is only rarely strongly drion-rich cytoplasm, respectively, giving
Microscopy positive. Strong, diffuse staining is more rise to distinct morphologies {1452,1654l.
Pituicytomas have a solid, compact ar- typical for S100 protein and vimentin. The derivation of three morphologically
chitecture and consist almost entirely of BCL2 staining is variable, but can be distinct tumours from pituicytes might
elongate, bipolar spindle cells arranged intense {1654,1966}. Stains for cytokerat- be explained by the existence of multiple
in a fascicular or storiform pattern {273, ins are negative, and those for EMA may subtypes of pituicytes in the normal neu-
1452,1654,1966l Although the tumours show a patchy pattern that is cytoplas- rohypophysis {1654,2499).
can show stubborn adherence to adja- mic rather than membranous. Pituicyto-
cent structures in the suprasellar space, mas do not demonstrate immunostaining Genetic profile
an infiltrative pattern is generally not seen for pituitary hormones or for neuronal or A genetic signature for pituicytoma has
under the microscope. Individual tumour neuroendocrine markers such as syn- not been defined. The limited number of
cells contain abundant eosinophilic cyto- aptophysin and chromogranin. NFP cases that have been tested to date have
plasm, and cell shapes range from short immunoreactivity is limited to axons in not shown R132H-mutant IDH1, V600E-
and plump to elongate and angulated. peritumoural neurohypophyseal tissue mutant BRAF, or KIAA 1549-BRAF fusion
Cell borders are readily apparent, es- and is not present within the tumour. The {1654). Microarray-based comparative
pecially on cross sections of fascicles. proliferation of these tumours is low as genomic hybridization analysis of a sin-
In strictly defined pituicytomas, there is measured by immunoreactivity for MIB1, gle case demonstrated losses of chro-
not a significant amount of cytoplasmic with the reported Ki-67 proliferation index mosome arms 1p, 14q, and 22q, with
granularity or vacuolization, and periodic ranging from 0.5% to 2.0% {273,1355). gains on 5p {1966).
acid-Schiff staining shows only minimal
reaction. Similarly, substantial oncocytic Cell of origin Prognosis and predictive factors
change is not a recognized feature of Pituicytomas presumptively arise from Pituicytomas are slowly enlarging, local-
pituicytoma. The nuclei are moderately specialized gli�I cells of the neurohy- ized tumours for which the current treat-
sized and oval to elongate, with only mild pophysis, called pituicytes {1150,1514, ment is surgical resection {687,2877) Lo-
irregularity of nuclear borders. Mitotic 2261 }. Such an origin accounts for the cal adherence of pituicytomas to regional
figures are rare. Reticulin fibres show a anatomical distribution of pituicytomas structures may preclude complete resec-
perivascular distribution, and intercellu- and is consistent with the tumour's mor- tion, and residual disease may gradually
lar reticulin is sparse. Importantly for the phological and immunophenotypic char- regrow over a period of several years.
differential diagnosis with pilocytic as- acteristics. The finding that pituicytomas There has been no correlation of prolif-
trocytoma and normal neurohypophysis, strongly express the nuclear transcrip- eration with clinical outcome. No instanc-
pituicytomas show no Rosenthal fibres, tion factor TTF1, like pituicytes of the de- es of malignant transformation or distant
eosinophilic granular bodies, or Herring veloping and mature neurohypophysis, metastasis have been reported to date.

Pituicytoma 333
.,

Spindle cell oncocytoma Lopes M.B.S.


Fuller G.N.
Roncaroli F.
Wesseling P

Definition
A spindled to epithelioid, oncocytic,
non-neuroendocrine neoplasm of the pi-
tuitary gland.
Spindle cell oncocytomas manifest in
adults and tend to follow a benign clinical
course. Like pituicytomas and granular
cell tumours of the sellar region, spindle
cell oncocytomas show nuclear expres-
sion of TTF1, suggesting that these three
tumours may constitute a spectrum of a
single nosological entity.

ICD-0 code 8290/0

Grading Fig. 17.21 Spindle cell oncocytoma. A Coronal T2-weighted MRI demonstrates a very large multilobulated sellar
Spindle cell oncocytoma corresponds mass with osseous remodelling and suprasellar extension; the mass laterally displaces the internal carotid arteries
histologically to WHO grade I. (arrowheads) and exerts mass effect on the right aspect of the optic chiasm (asterisk). B Coronal postcontrast
volumetric interpolated brain examination sequence demonstrates mildly heterogeneous enhancement with a small
Synonyms and historical annotation central non-enhancing component.
In the initial report of the entity in 2002,
Roncaroli et al. {2169) described spin- Epidemiology Localization
dle cell oncocytoma as spindle cell on- Spindle cell oncocytoma is a rare tumour, Spindle cell oncocytoma is a pituitary
cocytoma of the adenohypophysis. On but its true incidence is difficult to deter- tumour that may have a mixed intrasellar
the basis of the similar ultrastructural and mine. In the experience of one institution, and suprasellar location. Clinical mani-
immunohistochemical features of these spindle cell oncocytomas accounted for festation with a purely intrasellar tumour
neoplasms, derivation from folliculos- 0.4% of all sellar tumours {2169). About is relatively rare {504). Extension into the
tellate cells of the anterior pituitary was 25 cases have been reported in the litera- cavernous sinus {243,519} and invasion
suspected {2169}. However, more recent ture {1719). Based on the limited number of the sellar floor have been reported
data on the shared TTF1 immunoreactiv- of cases reported, spindle cell oncocyto- {1305).
ity of non-neoplastic pituicytes, pituicyto- ma is assumed to be a tumour of adults,
mas, granular cell tumours, and spindle with reported patient ages of 24-76 years Clinical features
cell oncocytomas may suggest a similar and a mean age of 56 years. The distri- The clinical presentation and neuroimag-
origin of these tumours and a shared link bution is equal between the sexes {1719) ing characteristics of spindle cell onco-
to the posterior pituitary {1452,1654). cytoma are indistinguishable from those

334 Tumours of the sellar region


- A
Fig. 17.23 Spindle cell oncocytoma. A Clear-cell appearance of the tumour cells arranged within a nested pattern. B Some examples have pleomorphic nuclei. C Oncocytic
changes may be variable within a given tumour.

of a non-functioning pituitary adenoma. can be heterogeneous {504). Calcifica- described {753). A clear margin or a
Visual disturbances have been found to tion may be seen on CT imaging {239}. pseudocapsule, as can be seen in some
be the most common presenting symp- Recurrent intratumoural bleeding has pituitary adenomas, is usually absent.
tom, followed by pituitary hypofunction been reported, leading to an erroneous
and (less frequently) headache, nausea, preoperative diagnosis of craniophar- Microscopy
and vomiting {504,171,2169}. Decreased yngioma {239}. The rich tumoural blood Spindle cell oncocytomas are typically
libido, sexual dysfunction, and oligomen- supply can be seen on MR angiography; composed of interlacing fascicles and
orrhoea or amenorrhoea secondary to pi- one study reported predominant feeding poorly defined lobules of spindle to epi-
tuitary stalk effect, with mild hyperprolac- from the bilateral internal carotid arteries thelioid cells with eosinophilic and vari-
tinaemia, have been documented {1719}. {753). ably oncocytic cytoplasm. Oncocytic
None of the patients reported to date had changes can be focal or widespread.
diabetes insipidus at onset {504). One Macroscopy The spectrum of morphological features
patient with aggressive spindle cell once- On gross inspection, spindle cell onco- that can be seen in spindle cell oncocy-
cytoma and involvement of the skull base cytoma is often indistinguishable from pi- toma is broad. Formations of whorls, fo-
presented with epistaxis {1305). tuitary adenoma. Most are large tumours, cal stromal myxoid changes {2189). clear
with an average craniocaudal dimension cells, osteoclastic-like giant cells, and
Imaging of 2.5-3 cm and a maximum reported follicle-like structures {2605,2830) have
The neuroimaging features of spindle size of 6.5 cm {519). They vary from soft, all been reported in individual cases,
cell oncocytoma are similar to those of creamy, and easily resectable lesions to in addition to the more typical features.
pituitary adenoma, but spindle cell once- firm tumours that adhere to surrounding Mild to moderate nuclear atypia and even
cytomas often avidly enhance following structures, and infrequently show de- marked pleomorphism may be seen. Fo-
administration of paramagnetic contrast, struction of the sellar floor {519,1305). ln- cal infiltrates of mature lymphocytes are
because of their greater vascular sup- tratumoural haemorrhage can occur, and seen in many lesions. The mitotic count
ply relative to adenomas. Enhancement severe intraoperative bleeding has been is typically low, often with an average

Spindle cell oncocytoma 335


.,

Fig. 17.26 Spindle cell oncocytoma. At the ultrastructural level, the tumour cells show numerous mitochondria
(oncocytic change) as well as several cell-cell junctions, mainly short desmosomes.

of < 1 mitotic figure per 10 high-power case (2605). Galectin-3 and annexin A1 to distinct morphologies (1452,1654).
fields. Recurrent lesions may or may not are also commonly expressed; although The derivation of three morphologically
show increased mitotic activity (1305, these markers are non-specific, they ini- distinct tumours from pituicytes might
1719). tially suggested a link to the folliculostel- be explained by the existence of multiple
late cell (2169}. subtypes of pituicytes in the normal neu-
Electron microscopy rohypophysis (1654,2499)
Ultrastructural examination is useful in Pro!tferation
the diagnosis of spindle cell oncocytoma The Ki-67 proliferation index is usually Genetic profile
(1305,2169}. Neoplastic cells appear low, with a reported range of 1-8% and In a study that included 7 spindle cell
spindled or polygonal in configuration an average value of 3%. One recurrent oncocytomas, none of the neoplasms
and are often filled with mitochondria, example had a Ki-67 proliferation index showed positive immunostaining for
which may appear abnormal. The tu- of 20%, but no data were available on the R132H-mutant IDH1 or any evidence of
mour cells lack secretory granules, a proliferation rate of the primary tumour BRAFV600E mutation or KIAA-BRAFfu-
key distinction from pituitary adenomas. (1305) sion {1654).
Well-formed desmosomes and interme-
diate-type junctions are encountered. Cell of origin Prognosis and predictive factors
lntracytoplasmic lumina with microvillous The histogenesis of spindle cell onco- Most spindle cell oncocytomas follow
projections have been reported (2605). cytoma remains unresolved. Initially, a a benign clinical course, but about one
derivation from folliculostellate cells of the third of reported cases have recurred, af-
lmmunophenotype adenohypophysis was postulated on the ter 3-15 years (1719} In only a small num-
Unlike pituitary adenomas, spindle cell basis of the immunoprofile (in particular ber of cases with recurrence did the orig-
oncocytomas lack immunoreactivity for galectin-3 and annexin A1 expression) inal neoplasm show an increased Ki-67
neuroendocrine markers and pituitary and ultrastructural features {2169). How- proliferation index (ranging from 10% to
hormones. Typically, spindle cell onco- ever, the finding that not only pituicyto- 20%) or marked cytological atypia (243,
cytomas express vimentin, 8100 protein, mas, but also granular cell tumours of the 1719}. Additional follow-up is needed to
BCL2, and EMA, and show staining with sellar region and spindle cell oncocyto- definitively determine the prognostic sig-
the antimitochondrial antibody MU213- mas express the nuclear transcription nificance of such features. Incomplete
UC clone 131-1, as well as nuclear factor TTF1, like pituicytes of the devel- resection is a risk factor for recurrence.
staining for TTF1 (1305,1452,1654,2167, oping and mature neurohypophysis, sug- Moderate tumour volume and absence
2169). EMA expression can vary from gests a pituicyte derivation {1452,1654). of invasion into surrounding structures
weak and focal to diffuse. The tumours It has been speculated that granular cell facilitate complete resection, whereas
are only focally positive for GFAP. CD44 tumours of the sellar region and spindle hypervascularity may hamper the proce-
and nestin have been reported in a sin- cell oncocytomas constitute variants of dure (1816). Recurrent tumours can fol-
gle case, suggesting a possible neuronal pituicytoma in which the tumour cells dis- low a more aggressive course, with an
precursor component {47}, and alpha- play lysosome-rich and mitochondrion- increased Ki-67 proliferation index and
crystallin B chain was seen in another rich cytoplasm, respectively, giving rise necrosis (1305).

336 Tumours of the sellar region


Metastatic tumours of the CNS Wesseling P.
von Deimling A.
Rosenblum M.K.
Mittelbronn M.
Aldape KO. Tanaka S.
Preusser M.

Definition Males Females


Tumours that originate outside the CNS
and spread via the haematogenous route
to the CNS or (less frequently) directly in-
vade the CNS from adjacent anatomical Primary
structures. tumours

Epidemiology

Incidence
Due to underdiagnosis and inaccurate
reporting, the incidence rates for brain
metastases reported in the literature Brain
probably underestimate the true inci- metastases
dence {731,2484). In a large, population-
based study in Sweden, the incidence
of patients admitted to hospital with
brain metastases doubled to 14 cases
Fig. 18.01 Relative frequencies of primary tumours and of brain metastases derived thereof {354,2032}. Tumours with
per 100 000 population between 1987 a high propensity to metastasize to the brain are lung cancer, breast cancer, renal cell carcinoma, and melanoma. In
and 2006. More efficient control of dis- this series of brain metastases, about 14% of cases in males and 8% of cases in females were diagnosed as carcinoma
ease spread outside the CNS and the of unknown primary (CUP). Based on the histology of archival tissue samples (874 cases collected in 1990-2011 at the
use of more advanced neuroimaging Institute of Neurology/Neuropathology in Vienna); metastases for which surgery was not performed are not represented.
techniques may have contributed to this The relative frequency of brain metastases may differ substantially in other regions of the world.
increase {2371 ). Autopsy studies have
reported that CNS metastases occur in at an age of 50-59 years; and with breast a predilection for brain involvement (e.g.
about 25% of patients who die of can- cancer at an age of 20-39 years {2484). lung cancer), and the introduction of new
cer {792). Leptomeningeal metastases The incidence of CNS metastases may therapeutic agents that prolong life and
occur in 4-15% of patients with solid tu- be increasing, in part due to increased are relatively efficacious overall but inef-
mours {400,2490} and dural metastases detection with improved imaging, an in- fective at preventing or treating metastat-
in 8-9% of patients with advanced can- crease in the incidence of tumours with ic disease within the CNS {1868,2484).
cer {14191 Spinal epidural metastases
are found in 5-10% and are much more
frequent than spinal leptomeningeal or
intramedullary metastases {1728).

Age and sex distnbution


CNS metastases are the most common
CNS neoplasms in adults, but metasta-
ses account for only about 2% of all pae-
diatric CNS tumours. As many as 30% of
adults and 6-10% of children with cancer
develop brain metastases. The relative
proportions of various primary tumours
are different between the two sexes, but
sex has no significant independent effect
on the occurrence of CNS metastasis for
most tumour types {130,731,2749). The
incidence of brain metastases has been
reported to be highest among patients di-
agnosed with primary lung cancer at an Fig. 18.02 Metastasis of an adenocarcinoma in the right frontal lobe. A Gadolinium-enhanced T1-weighted MRI
age of 40-49 years; with primary mela- showing a contrast-enhancing tumour surrounded by a large hypointense area corresponding to perifocal oedema.
noma, renal cancer, or colorectal cancer B Both the tumour and perifocal oedema show bright T2-hyperintensity.

338 Metastatic tumours


Fig. 18.03 Patterns of CNS metastases. A lntraparenchymal metastasis of a lung carcinoma extending into the left hemisphere via the splenium of the corpus callosum. B Cerebellar,
intraparenchymal metastasis of a ductal carcinoma of the pancreas extending to the leptomeningeal compartment. C Multiple dural metastases of a carcinoma of the gastric cardia.

Origin of CNS metastases hemispheres (particularly in arterial bor- melanoma, and haematopoietic tumours
The most common source of brain me- der zones and at the junction of cerebral (2490). Spinal epidural metastases are
tastasis in adults is lung cancer (espe- cortex and white matter), 15% in the cer- more common in cancer of the prostate,
cially adenocarcinoma and small cell ebellum, and 5% in the brain stem. Fewer breast, lung, and kidney, non-Hodgkin
carcinoma), followed by breast cancer, than half of all brain metastases are sin- lymphoma, and multiple myeloma. ln-
melanoma, renal cell carcinoma, and gle (i.e. the only metastatic lesion in the tramedullary spinal cord metastases are
colorectal cancer (354,1783,2032). Pros- brain), and very few are solitary (i.e the more common in small cell lung carci-
tate, breast, and lung cancer are the most only metastasis in the body) {792,1923) noma {1728).
common origins of spinal epidural metas- Other intracranial sites include the dura
tasis, followed by non-Hodgkin lympho- and leptomeninges; in these sites, ex- Clinical features
ma, multiple myeloma, and renal cancer tension from or to other compartments
(1728). Tumours and their molecular sub- is more common {1419,1760). The vast Symptoms and signs
types vary in their propensity to metas- majority of metastases affecting the spi- The neurological signs and symptoms
tasize to the CNS (130,510,2297). In as nal cord expand from the vertebral body of intracranial metastases are generally
many as 10% of patients with brain me- or paravertebral tissues into the epidural caused by increased intracranial pres-
tastases, no primary tumour is found at space {1728). Occasionally, metastatic sure or local effect of the tumour on the
presentation (2032). In children, the most CNS tumours seed along the walls of the adjacent brain tissue. The signs and
common sources of CNS metastases are ventricles or are located in the pituitary symptoms may progress gradually and
leukaemias and lymphomas, followed gland or choroid plexus. Rarely, tumour- include headache, altered mental sta-
by non-haematopoietic CNS neoplasms to-tumour metastasis occurs, with lung tus, paresis, ataxia, visual changes, nau-
such as germ cell tumours, osteosarco- and breast cancer being the most com- sea, and sensory disturbances. Some
ma, neuroblastoma, Ewing sarcoma, and mon donor tumours and meningioma the patients present acutely with seizure,
rhabdomyosarcoma (510,2749). Occa- most common recipient {1711). Of par- infarct, or haemorrhage {1453). The in-
sionally, primary neoplasms in the head ticular diagnostic challenge is metastasis terval between diagnosis of the primary
and neck region extend intracranially by of renal cell carcinoma to haemangio- tumour and the CNS metastasis is gener-
direct invasion (per continuitatem), some- blastoma in the setting of von Hippel-Lin- ally < 1 year for lung carcinoma, but can
times along cranial nerves, and present dau disease {62,1138). Dural metastases be multiple years for breast cancer and
as intracranial tumours {2323). are relatively common in cancer of the melanoma {2297). Many patients with
prostate, breast, and lung, and in hae- leptomeningeal metastasis have multi-
Localization matological malignancies {1419,1760). ple, varied neurological symptoms and
Approximately 80% of all brain me- Leptomeningeal metastases are more signs at presentation, including head-
tastases are located in the cerebral common in lu11g and breast cancer, ache, mental alteration, ataxia, cranial

t -·
Fig. 18.04 Patterns of CNS metastasis. A Extensive CSF spread of small cell lung carcinoma cells along the walls of both lateral ventricles and the third ventricle. B lntraventricular
metastasis of a lung adenocarcinoma infiltrating the choroid plexus with (C) TTF1 staining of tumour cell nuclei.

Metastatic tumours of the CNS 339


B
Fig. 18.05 Patterns of CNS metastasis. A Leptomeningeal metastasis of a non-Hodgkin lymphoma.

nerve dysfunction, and radiculopathy. and peritumoural oedema. Metastases demonstrate a pushing margin and/or in-
Cytological examination reveals malig- of adenocarcinomas may contain collec- vasion via Virchow-Robin spaces rather
nant cells in the initial cerebrospinal fluid tions of mucoid material. Haemorrhage is than a single-cell infiltration pattern (es-
sample in about 50% of such patients; relatively frequent in metastases of cho- pecially in focal areas) and can therefore
this proportion may increase to � 80% riocarcinoma, melanoma, and clear cell occasionally be confused for metastatic
when cerebrospinal fluid sampling is re- renal cell carcinoma. Melanoma metasta- tumours on a morphological basis.
peated and adequate volumes (� 10 ml) ses with abundant melanin pigment have
are available for cytological analysis a brown to black colour. Leptomeningeal lmmunophenotype
{400,1453). Spinal metastases generally metastasis may produce diffuse opacifi- The immunohistochemical characteristics
result in compression of the spinal cord cation of the membranes or present as of secondary CNS tumours are generally
or nerve roots and may cause back pain, multiple nodules {1923). Dural metas- similar to those of the tumours from which
weakness of the extremities, sensory tases can grow as localized plaques or they originate. lmmunohistochemical
disturbances, and incontinence over the nodules and as diffuse lesions. Primary analysis is often very helpful for
course of hours, days, or weeks {1453). neoplasms in the head and neck region distinguishing primary from secondary
that extend intracranially by direct inva- CNS tumours and for assessment of the
Imaging sion generally cause significant destruc- exact nature and origin of the metastatic
On MRI, intraparenchymal metastases tion of the skull bones. However, in some neoplasm (particularly in cases with an
are generally circumscribed and show cases, the skull is penetrated by rela- unknown primary tumour) {149,1923).
mild T1-hypointensity, T2-hyperintensi- tively subtle, perivascular or perineural
ty, and diffuse or ring-like contrast en- invasion, without major bone destruction Proltferation
hancement with a surrounding zone of {2323). Metastatic CNS tumours show variable
parenchymal oedema. Haemorrhagic and often marked mitotic activity. The
metastases and metastatic melanomas Microscopy proliferation index may be significantly
containing melanin pigment may dem- The histological and immunohisto- higher than in the primary neoplasm
onstrate hyperintensity on non-contrast chemical features of secondary CNS tu- {171).
MRI or CT {2833). In patients with lepto- mours are as diverse as those of the pri-
meningeal metastasis, MRI can show fo- mary tumours from which they arise. Most Pathogenesis
cal or diffuse leptomeningeal thickening brain metastases are fairly well demarcat- Before they present as haematogenous
and contrast enhancement (sometimes ed, with variable perivascular growth (so- metastases in the CNS, tumour cells must
with dispersed tumour nodules in the called vascular cooption) in the adjacent successfully complete a series of steps:
subarachnoid space). Enhancement and CNS tissue {172). On occasion, small cell escape from the primary tumour, entry
enlargement of the cranial nerves and carcinomas and lymphomas may show into and survival in the blood stream, ar-
communicating hydrocephalus may also more diffuse infiltration (pseudoglioma- rest and extravasation in the CNS, and
be found {848,1728). MRI can depict du- tous growth) in the adjacent brain pa- survival and growth in the CNS micro-
ral metastases as nodular masses or du- renchyma {145,1770). Tumour necrosis environment {1264,2032). The molecu-
ral thickening along the bone structures, may be extensive, leaving recognizable lar basis of CNS spread in the various
whereas metastases in vertebral bodies tumour tissue only at the periphery of the tumour types is poorly elucidated and
are visualized as discrete, confluent or lesion and around blood vessels {1923). requires further study. Secondary CNS
diffuse areas of low signal intensity. CT In leptomeningeal metastasis, the tumour tumours may also develop by direct ex-
scan may be useful for detection of bone cells are dispersed in the subarachnoid tension from primary tumours in adjacent
involvement {1419). and Virchow-Robin spaces and may in- anatomical structures (e.g. paranasal si-
vade the adjacent CNS parenchyma and nuses and bone) {2323). Such tumours
Macroscopy nerve roots {2490} Although the pattern are not formally considered metastases,
Metastases in the brain and spinal cord of infiltration is often helpful in distin- because they remain in continuity with
parenchyma often form grossly circum- guishing a metastatic tumour from a pri- the primary neoplasm. Once in contact
scribed and rounded, greyish-white or mary CNS tumour (e.g. a diffuse glioma), with the cerebrospinal fluid-containing
tan masses with variable central necrosis occasional cases of glioblastoma can compartments, cells of those tumours

340 Metastatic tumours


Table 18.01 lmmunohistochemical profiles of metastatic carcinomas and melanoma; adapted from Pekmezci Mand Perry A{1923}
NCAM1
CKS/6 (CDS&) CK7 CK20 TTF1 NapA GCDFP15 CDX2 RCCm PSA EMA PAXS Vim Melan·A

Squamous cell carcinoma + + +


Lung small cell carcinoma + + + +
Lung adenocarcinoma + + + +/- +
Breast adenocarcinoma +/- + + +
Colorectal adenocarcinoma + + +
Stomach adenocarcinoma + + + +
Renal cell carcinoma + + + +
Prostate adenocarcinoma + +
Urothelial carcinoma + + + +
Melanoma + +
-: usually negative; +/-: positive in a significant subset; +: usually positive.
Abbreviations: NapA, napsin-A; PSA, prostate-specific antigen; RCCm, renal cell carcinoma marker; Vim, vimentin.
Note: This table lists the most common patterns of expression, but many exceptions exist. The list in this table is not complete; other markers may also be useful, such as
HMB45 and microphthalmia-associated transcription factor (MITF) for melanocytic tumours. If the primary versus metastatic nature of the CNS neoplasm is uncertain, other
immunohistochemical markers are useful, such as GFAP and OLIG2 for gliomas; inhibin and 02-40 for haemangioblastomas; and GFAP, transthyretin, and KIR7.1 for choroid
plexus tumours.

may disseminate (seed) throughout the expression in breast cancer, BRAF mu- prognostic scores taking these param-
CNS. tation in melanoma, RAS mutation in eters into account have been described,
colorectal cancer, and ERBB2 amplifica- but require validation in independent
Molecular genetics and tion in gastro-oesophageal cancer {170). and prospective studies {1328,2404).
predictive factors For some markers, there are significant Other factors of prognostic significance
A wide range of tumour types cause discordance rates between primary tu- include the specific tumour type and the
brain metastases, and some molecularly mours and brain metastases, which may molecular drivers involved (e.g. ERBB2 in
defined tumour subtypes have higher influence the necessity of performing breast cancer) {1505). Neuroradiological
propensity to metastasize to the CNS biomarker analyses on brain metastasis parameters such as peritumoural brain
(e.g. ERBB2-positive and triple-negative tissue samples for therapy planning {170, oedema may also provide prognostic
breast cancer) {1505). Systemic therapy 1024). information {2401 ). In more recent stud-
with novel targeted agents is increasingly ies, the reported improvement in overall
being used for patients with CNS metas- Prognostic factors survival of patients with CNS metastases
tases. Biomarker tests to be considered The main established prognostic fac- may be attributable to improvements in
for such therapies include EGFR muta- tors for patients with brain metastases focal and systemic therapies in combina-
tion and ALK rearrangement in non-small are patient age, Karnofsky performance tion with earlier detection of such metas-
cell lung cancer, ERBB2 amplification status, number of brain metastases, and tases {1783).
and estrogen and progesterone receptor status of extracranial disease. Several

Metastatic tumours of the CNS 341


Contributors

Dr Kenneth D. ALDAPE* Dr Mitchel S. BERGER* Dr Corinne BOUVIER


Ontario Cancer Institute Department of Neurological Surgery Service d'Anatomie Pathologique et
Brain Tumor Biology Program University of California, San Francisco de Neuropathologie
Princess Margaret Cancer Centre 505 Parnassus Avenue, Room M786 H6pital de la Timone 2
Toronto Medical Discovery Tower San Francisco CA 94143-0112 265 rue Saint-Pierre
101 College Street, Room 14-601 USA 13385 Marseille Cedex 05
Toronto ON MSG 1 L7 Tel. +1415353 3933; +1415353 2637 FRANCE
CANADA Fax +1415353 3910 Tel. +33 4 13 42 90 13
Tel. +1416634 8793 bergerm@neurosurg.ucsf.edu Fax +33 4 13 42 90 42
kadalpe@gmail.com corinne. labit@ap-hm.fr

Dr Cristina R. ANTONESCU* Dr Jaclyn A. BIEGEL Dr Michael BRADA *


Department of Pathology Pathology and Laboratory Medicine Department of Molecular and
Memorial Sloan Kettering Cancer Center Children's Hospital Los Angeles and Clinical Cancer Medicine
1275 York Avenue Keck School of Medicine of USC University of Liverpool
New York NY 10021 4650 Sunset Boulevard, Mail Stop #173 Clatterbridge Cancer Centre
USA Los Angeles CA 90027 Bebington, Wirral CH63 4JY
Tel. + 1 212 639 5905 USA UNITED KINGDOM
Fax +1212717 3203 Tel. +1323361 8674 Tel. +44 15 148 277 93
antonesc@mskcc.org Fax + 1 323 644 8580 Fax +44 15 148 276 21
jbiegel@chla.usc.edu michael.brada@liverpool.ac.uk

Dr Jill BARNHOLTZ-SLOAN Dr Wojciech BIERNAT Dr Sebastian BRANDNER


Case Comprehensive Cancer Center Department of Pathomorphology Division of Neuropathology
Case Western Reserve University Medical University of Gdansk UCL Institute of Neurology and National
2-526 Wolstein Research Building ulica Smoluchowskiego 17 Hospital for Neurology and Neurosurgery
2103 Cornell Road 80-214 Gdansk Mailbox 126, Queen Square
Cleveland OH 44106-7295 POLAND London WC 1 N 3BG
USA Tel. +48 58 349 3750 UNITED KINGDOM
Tel. +1216368 1506 Fax +48 58 349 3750 Tel. +44 20 344 844 35
Fax + 1 216 368 2606 biernat@gumed.edu.pl Fax +44 20 344 844 86
jill.barnholtz-sloan@case.edu s.brandner@ucl.ac.uk

Dr Boris C. BASTIAN Dr Darell D. SIGNER Dr Daniel J. BRAT*


Department of Pathology and Dermatology Department of Pathology Department of Pathology and
University of California Duke University Medical Center Laboratory Medicine
San Francisco Helen Diller Box 3156, Research Drive, 177 MSRB Emory University Hospital
Family Comprehensive Cancer Center Durham NC 27710 H-176, 1364 Clifton Road NE
1450 Third Street, Box 3116 USA Atlanta GA 30322
San Francisco CA 94158-9001 Tel.+ 1 919 684 5018; + 1 919 684 6790 USA
USA Fax + 1 919 684 6458 Tel. +1 404 7121266
Tel. + 1 415 502 0267 darell.bigner@duke.edu Fax+14047120148
boris.bastian@ucsf.edu dbrat@emory.edu

Dr Albert J. BECKER Dr Ingmar BLOMCKE Dr Herbert BUDKA


Department of Neuropathology University of Erlangen Institute of Neuropathology
University of Bonn Medical Center Institute of Neuropathology University Hospital Zurich
Sigmund-Freud-Strasse 25 Schwabachanlage 6 Schmelzbergstrasse 12
53105 Bonn 91054 Erlangen 8091 Zurich
GERMANY GERMANY SWITZERLAND
Tel. +49 228 287 11352 bluemcke@uk-erlangen.de Tel. +41 44 255 25 02
Fax +49 228 287 14331 Fax +41 44 255 44 02
albert_becker@uni-bonn.de herbert.budka@usz.ch

*Indicates participation in the Working Group Meeting on the WHO Classification of Tumours of the Central Nervous System that was held in
Heidelberg, Germany, 21-24 June 2015.
# Indicates disclosure of interests.

342 Contributors
Dr Peter C. BURGER* Dr Elizabeth B. CLAUS Dr Judith A. FERRY
Department of Pathology Department of Biostatistics/ Department of Pathology
Johns Hopkins Medical Institutions Epidemiology/Neurosurgery Massachusetts General Hospital
Patl1ology Building, Zayed Tower Yale University School of Medicine 55 Fruit Street
Room 2101, 1800 Orleans Street 60 College Street, Box 208034 Boston MA 02114
Baltimore MD 21231 New Haven CT 06520-8034 USA
USA USA Tel. + 1 617 726 4826
Tel. +14109558378 Tel. + 1 203 785 2838 Fax +1617726 7474
Fax +1410614 9310 Fax + 1 203 785 6912 jferry@mgh.harvard.edu
pburger@jhmi.edu elizabeth.claus@yale.edu

Dr Rolf BUSLEI Dr V. Peter COLLINS* Dr Dominique FIGARELLA-BRANGER*


University of Erlangen Department of Pathology Laboratoire d'anatomie
Institute of Neuropathology University of Cambridge pathologique-neuropathologique
Schwabachanlage 6 Tennis Court Road H6pital de la Timone
91054 Erlangen Cambridge CB2 1 OP 264 rue Saint-Pierre
GERMANY UNITED KINGDOM 13385 Marseille Cedex 05
Tel. +49 9131 852 60 31 Tel. +44 1223 336 072; +44 1223 217 164 FRANCE
Fax +49 9131 852 60 33 Fax +44 1223 216 980 Tel. +33 4 13 42 90 38
rolf.buslei@uk-erlangen.de vpc20@cam.ac.uk Fax +33 4 91 38 44 11
domin ique. f igarella-branger@univ-amu.fr

Dr J. Gregory CAIRNCROSS Dr Martina DECKERT Dr Gregory N. FULLER*


Department of Clinical Neurosciences Department of Neuropathology Department of Pathology
University of Calgary and University of Cologne University of Texas
Foothills Medical Centre Kerpener Strasse 62 MD Anderson Cancer Center
HRIC 2AA-19 3280 Hospital Drive NW 50931 Cologne 1515 Holcombe Boulevard, Unit 085
Calgary AB T2N 4Z6 GERMANY Houston TX 77030
CANADA Tel. +49 221 478 52 65 USA
Tel. +1403210 3934 Fax +49 221 478 72 37 Tel. + 1 713 792 2042
Fax +1 403 210 8135 martina.deckert@uni-koeln.de Fax+ 1 713 792 3696
jgcairnx@ucalgary.ca gfuller@mdanderson.org

Dr David CAPPER*# Dr Charles G. EBERHART* Dr Marco GESSI


Institute of Pathology Department of Pathology Department of Neuropathology
Department of Neuropathology Johns Hopkins University University of Bonn Medical Center
Heidelberg University 720 Rutland Avenue, Ross Building 558 Sigmund-Freud-Strasse 25
Im Neuenheimer Feld 224 Baltimore MD 21205 53105 Bonn
69120 Heidelberg USA GERMANY
GERMANY Tel. + 1 4 10 502 5185 Tel. +49 228 287 166 42
Tel. +49 6221 56 37 254 Fax + 1 410 959 9777 Fax +49 228 287 143 31
Fax +49 6221 56 45 66 ceberha@jhmi.edu marco.gessi@ukb.uni-bonn.de
david.capper@med.uni-heidelberg.de mgessimd@yahoo.com

Dr Webster K. CAVENEE* Dr David W. ELLISON* Dr Felice GIANGASPERO*


Cellular and Molecular Medicine East Department of Pathology Department of Radiological, Oncological and
University of California, San Diego St. Jude Children's Research Hospital Anatomopathological Sciences
9500 Gilman Drive, #0660, Room 3080 262 Danny Thomas Place, MS 250 Policlinico Umberto I, Sapienza University
La Jolla, San Diego CA 92093-0660 Room C-5001 Viale Regina Elena. 324
USA Memphis TN 38105-3678 00161 Rome
Tel. + 1 858 534 7805 USA ITALY
Fax + 1 858 534 7750 Tel. + 1 901 595 5438 Tel. +39 06 49 73 710; +39 06 44 46 86 06
wcavenee@ucsd.edu Fax + 1 901 595 3JOO Fax +39 06 48 97 91 75
david.ellison@stjude.org felice.giangaspero@uniroma1.it

Dr Leila CHIMELLI Dr Charis ENG Dr Caterina GIANNINI*


Department of Pathology Genomic Medicine Institute Anatomic Pathology
University Hospital CFF - UFRJ Cleveland Clinic Lerner Research Institute Mayo Clinic College of Medicine
Avenida Epitacio Pessoa 4720/501 9500 Euclid Avenue, NE-50 200 First Street SW
Rio de Janeiro 22471-003 Cleveland OH 44195 Rochester MN 55905
BRAZIL USA USA
Tel. +55 21 99604 4880; +55 21 2226 9998 Tel.+ 1 216 444 3440 Tel. +15075381181
Fax +55 21 3207 6548 Fax+ 1 216 636 0655 Fax + 1 507 284 1599
leila.chimelli@gmail.com engc@ccf.org giannini.caterina@mayo.edu

Contributors 343
Dr Hannu HMPASALO Dr Takanori HIROSE* Dr Koichi ICHIMURA#
Department of Pathology Department of Pathology Division of Brain Tumor
University of Tampere I Finlab Laboratories Kobe University Hospital Translational Research
POB66 7-5-2 Kusunoki-cho. Chuo-ku National Cancer Center Research Institute
SF-335101 Tampere Hyogo Prefecture 5-1-1 Tsukiji, Chuo-ku
FINLAND Kobe City 650-0017 Tokyo 104-0045
Tel. +358 3 247 65 60 JAPAN JAPAN
Fax +358 3 247 55 03 thirose@hp.pref.hyogo.jp Tel. +81 3 3547 52 01 ext. 28 26
hannu.haapasalo@pshp.fi Fax +81 3 3542 25 30
kichimur@ncc.go.jp

Dr Johannes A. HAINFELLNER# Dr Mrinalini HONAVAR Dr David JONES#


Institute of Neurology Department of Anatomical Pathology Division of Pediatric Neurooncology
Medical University of Vienna Pedro Hispano Hospital German Cancer Research Center
Wahrinqer Gurtel 18-20 Rua de Alfredo Cunha 365 Im Neuenheimer Feld 280
1090 Vienna Matosinhos 69120 Heidelberg
AUSTRIA PORTUGAL GERMANY
Tel. +43 1 404 00 55 000 minalhonavar@gmail.com Tel. +49 6221 42 45 94
Fax +43 1 404 00 55 110 mrinalini.honavar@ulsm.min-saude.pt Fax +49 6221 42 46 39
johannes. hainfel lner@medun iwien. ac. at david.jones@dkfz.de

Dr Christian HARTMANN# Dr Annie HUANG Dr Anne JOUVET*


Department of Neuropathology Department of Paediatric Oncology Centre de Pathologie et de
Institute of Pathology Hospital for Sick Children Neuropathologie Est
Hannover Medical School 555 University Avenue Groupement Hospitalier Est
Carl-Neubert-Strasse 1 Toronto ON MSG 1X8 59 boulevard Pinel
30625 Hannover CANADA 69677 Bron Cedex
GERMANY Tel. +14168138221 FRANCE
Tel. +49 511 532 52 37 Fax +1416813 5327 Tel. +33 4 72 35 76 34
Fax +49 511 532 18 512 annie.huang@sickkids.ca Fax +33 4 72 35 70 67
hartmann.christian@mh-hannover.de anne. jouvet@chu-lyon.fr

Dr Martin HASSELBLATI Dr Theo J.M. HULSEBOS Dr Alexander R. JUDKINS


Institute of Neuropathology Department of Genome Analysis Pathology and Laboratory Medicine
University of Munster Academic Medical Center Children's Hospital Los Angeles and
Pottkamp 2 Meibergdreef 9 Keck School of Medicine of USC
48149 Muenster 1105 AZ Amsterdam 4650 Sunset Boulevard, Mail Stop #43
GERMANY THE NETHERLANDS Los Angeles CA 90027
Tel. +49 251 83 569 69 Tel. +31 20 566 30 24 USA
Fax +49 251 83 569 71 Fax +31 20 566 93 12 Tel. +1323361 4516
martin. hasselblatt@ukmuenster.de I. j hulsebos@amc uva.nl Fax + 1 323 361 8005
ajudkins@chlausc.edu

Dr Cynthia HAWKINS* Dr Stephen HUNTER Dr Paul KLEIHUES*


Division of Pathology, Department of Department of Pathology Faculty of Medicine
Paediatric Laboratory Medicine Emory University School of Medicine University of Zurich
Hospital for Sick Children 1364 Clifton Road NE Pestalozzistrasse 5
555 University Avenue Atlanta GA 30322 8032 Zurich
Toronto ON MSG 1X8 USA SWITZERLAND
CANADA Tel. +1404 7124278 Tel. +41 44 362 21 10
Tel. + 1 416 813 5938 Fax +1404 7120714 kleihues@pathol.uzh.ch
Fax +1416813 5974 stephenhunterwernory.orq
cynthia.hawkins@sickkids.ca

Dr Monika HEGI# Dr Jason T. HUSE Dr Bette K. KLEINSCHMIDT-DeMASTERS


Department of Clinical Neurosciences Department of Pathology Department of Neuropathology
University of Lausanne Hospital Memorial Sloan Kettering Cancer Center University of Colorado
Chemin des Boveresses 1275 York Avenue Anschutz Medical Campus
1066 Epalinges New York NY 10065 12605 East 16th Avenue, Room 3017
SWITZERLAND USA Aurora CO 80045
Tel. +41213142582 Tel. + 1 646 888 2642 USA
Fax+41213142587 Fax + 1 646 422 0856 bk.demasters@ucdenver.edu
monika.hegi@chuv.ch husej@mskcc.org

344 Contributors
Dr Philip M. KLUIN Dr lvo LEUSCHNER Dr Christian MAWRIN
Department of Pathology and Medical Biology Kiel Pediatric Tumor Registry Department of Neuropathology
University of Groningen Department of Pediatric Pathology Otto-von-Guericke University
Hanzeplein 1 University of Kiel Leipziger Strasse 44
9713 GZ Groningen Arnold-Heller-Strasse 3, House 14 39120 Magdeburg
THE NETHERLANDS 24105 Kiel GERMANY
Tel. +31 50 361 46 84 GERMANY christian.mawrin@med.ovgu.de
p.m.kluin@umcg.nl Tel. +49 431 597 34 44
Fax +49 431 597 34 86
ileuschner@path.uni-kiel.de

Dr Takashi KOMORI* Dr Pawel P. LIBERSKI Dr Roger McLENDON*#


Department of Laboratory Medicine and Department of Molecular Pathology and Department of Pathology
Pathology, Neuropathology Neuropathology Duke University Medical Center
Tokyo Metropolitan Neurological Hospital Medical University of Lodz DUMC Box 3712
2-6-1 Musashidai, Fuchu ulica Kosciuszki 4 Durham NC 27710
Tokyo 183-0042 90-419 Lodz USA
JAPAN POLAND Tel. +1919684 6940
Tel. +81 42 323 51 10 Fax +48 42 679 14 77 Fax +1919681 7634
Fax +81 42 322 62 19 ppliber@csk.am.lodz.pl mclen001@mc.duke.edu
komori-tk@igakuken.or.jp

Dr Marcel KOOL Dr Jay LOEFFLER Dr Michel MITTELBRONN


Division of Pediatric Neurooncology Department of Radiation Oncology Institute of Neurology (Edinger Institute)
German Cancer Research Center Massachusetts General Hospital Goethe University Frankfurt
Im Neuenheimer Feld 280 Clark Center for Radiation Oncology Heinrich-Hoffmann-Strasse 7
69120 Heidelberg 100 Blossom Street 60528 Frankfurt am Main
GERMANY Boston MA 02114-2606 GERMANY
Tel. +49 6221 42 46 36 USA Tel. +49 69 6301 841 69
Fax +49 6221 42 46 39 Tel. +16177241548 Fax +49 69 9301 841 50
m.kool@dkfz.de Fax + 1 617 724 8334 michel.mittelbronn@kgu.de
jloeffler@partners.org

Dr Andrey KORSHUNOV Dr M. Beatriz S. LOPES Dr Yoichi NAKAZATO


Clinical Cooperation Unit Neuropathology Department of Pathology (Neuropathology) Department of Pathology
German Cancer Research Center University of Virginia Health System Hidaka Hospital
Im Neuenheimer Feld 280 Box 800214 - HSC 886 Nakaomachi, Takasaki
69120 Heidelberg Charlottesville VA 22908-0214 Gunma 370-0001
GERMANY USA JAPAN
Tel. +49 6221 56 41 45 Tel. + 1 434 924 9175 Tel. +81 27 362 62 01
a. korshunov@dkfz-heidelberg.de Fax + 1 434 924 9177 Fax +81 27 362 89 01
msl2e@virginia.edu nakazato_yoichi@gunma-u.ac.jp

Dr Johan M. KROS* Dr David N. LOUIS* Dr Hartmut P.H. NEUMANN


Division of Pathology/Neuropathology James Homer Wright Pathology Laboratories Department of Nephrology and Hypertension
Erasmus Medical Center Massachusetts General Hospital Albert Ludwigs University of Freiburg
Dr. Molewaterplein 50 55 Fruit Street, Warren 225 Hugstetterstrasse 55
3015 GE Rotterdam Boston MA 02114 79106 Freiburg
THE NETHERLANDS USA GERMANY
Tel. +31 61 884 57 51 Tel. + 1 617 726 2966 Tel. +49 761 270 35 78
Fax +31 10 408 79 05 Fax +1 617 726 7533 Fax +49 761 270 37 78
j.m.kros@erasmusmc.nl dlouis@mgh.harvard.edu hartmut.neumann@uniklinik-freiburg.de

Dr Suet Yi LEUNG# Dr Masao MATSUTANI Dr Ho-Keung NG*


Department of Pathology Department of Neuro-Oncology/Neurosurgery Anatomical and Cellular Pathology
Li Ka Shing Faculty of Medicine Saitama Medical University International Prince of Wales Hospital
University of Hong Kong Queen Mary Hospital Medical Center Chinese University of Hong Kong
Pokfulam Road Yamane 1397, Hidaka-shi Sha tin
Hong Kong SAR Saitama 350-1298 Hong Kong SAR
CHINA JAPAN CHINA
Tel. +852 225 54 401 Tel. +8149276 15 51 Tel. +852 2632 33 37
Fax +852 287 25 197 Fax +81 492 76 15 51 Fax +852 2637 62 7 4
suetyi@hku.hk matutani@saitama-med.ac.jp hkng@cuhk.edu.hk

Contributors 345
Dr Hiroko OHGAKI* Dr Stefan PFISTER*# Dr Fausto RODRIGUEZ
Section of Molecular Pathology Division of Pediatric Neurooncology and Department of Pathology
International Agency for Research on Cancer Pediatrics Clinic Ill Division of Neuropathology
150 Cours Albert Thomas German Cancer Research Center Johns Hopkins Hospital
69372 Lyon Cedex 08 Im Neuenheimer Feld 280 and 430 Sheikh Zayed Tower, Room M2101
FRANCE 69120 Heidelberg 1800 Orleans Street
Tel. +33 4 72 73 85 34 GERMANY Baltimore MD 21231
Fax +33 4 72 73 86 98 Tel. +49 6221 42 46 18 USA
ohgakih@iarc.fr Fax +49 6221 42 46 39 Tel. +1443287 6646
stefan. pf ister@med. uni-heidelberg. de Fax +1410614 9310
frodrig4@jhmi.edu
Dr Magali OLIVIER Dr Torsten PIETSCH*# Dr Frederico RONCAROLI
Molecular Mechanisms and Department of Neuropathology Department of Pathology
Biomarkers Group University of Bonn Medical Center Salford Royal Foundation Hospital
International Agency for Research on Cancer Sigmund-Freud-Strasse 25 University of Manchester
150 Cours Albert Thomas 53105 Bonn Stott Lane
69372 Lyon Cedex 08 GERMANY Salford, Manchester M68HD
FRANCE Tel. +49 228 287 166 06 UNITED KINGDOM
Tel. +33 4 72 73 86 69 Fax +49 228 287 143 31 Tel: +44 161 206 5013
Fax +33 4 72 73 83 45 t. pietsch@uni-bonn.de federico.roncaroli@manchester.ac.uk
olivierm@iarc.fr

Dr Anne OSBORN# Dr Karl H. PLATE Dr Marc K. ROSENBLUM


Department of Radiology Institute of Neurology (Edinger Institute) Department of Pathology
University of Utah Goethe University Medical School Memorial Sloan Kettering Cancer Center
30 N 1900 E, #1A71 NeuroScienceCenter 1275 York Avenue
Salt Lake City UT 84132 Heinrich-Hoffmann-Strasse 7 New York NY 10021
USA 60528 Frankfurt am Main USA
Tel. + 1 801 581 7553 GERMANY Tel. + 1 212 639 3844
anne .osborn@hsc. utah. edu Tel. +49 69 6301 60 42 Fax +1212717 3203
Fax +49 69 6301 84 150 rosenbl 1@mskcc.org
karl-heinz. plate@kg u. de

Dr Sung-Hye PARK Dr Matthias PREUSSER Dr Brian ROUS*


Department of Pathology Department of Internal Medicine I and National Cancer Registration Service -
Seoul National University College of Medicine Comprehensive Cancer Center Vienna Eastern Office
Seoul National University Hospital Medical University of Vienna Victoria House, Capital Park
101 Daehak-ro, Jongno-gu Wii.hringer GOrtel 18-20 Fulbourn, Cambridge CB21 5XB
Seoul 110-744 1097 Vienna UNITED KINGDOM
REPUBLIC OF KOREA AUSTRIA Tel. +44 122 321 3625
Tel. +82 2 2072 30 90 Tel. +43 1 40400 44 570 Fax +44 122 321 3571
Fax +82 2 7 435 530 Fax +43 1 40400 60 880 brian.rous@phe.gov.uk
shparknp@snu.ac.kr matthias. preusser@meduniwien.ac. at

Dr Werner PAULUS* Dr Guido REIFENBERGER*# Dr Elisabeth J. RUSHING#


Institute of Neuropathology Institute of Neuropathology Institute of Neuropathology
University Hospital Munster University Hospital DOsseldorf University Hospital Zurich
Pottkamp 2 Moorenstrasse 5, Building 14.79, Floor Ill Schmelzbergstrasse 12
48149 Muenster 40225 DOsseldorf 8091 Zurich
GERMANY GERMANY SWITZERLAND
Tel. +49 251 83 569 66 Tel. +492118118660 Tel. +41 44 255 43 81
Fax +49 251 83 569 71 Fax +49 211 8117804 elisabethjane.rushing@usz.ch
werner.paulus@uni-muenster.de reifenberger@med.uni-duesseldorf.de

Dr Arie PERRY* Dr David E. REUSS Dr Siegal SADETZKI


Department of Pathology Institute of Pathology Cancer and Radiation Epidemiology Unit
Division of Neuropathology Department of Neuropathology Gertner Institute
University of California, San Francisco Heidelberg University Chaim Sheba Medical Center
505 Parnassus Avenue, M551, Box 0102 Im Neuenheimer Feld 224 5262000 Tel Hashomer
San Francisco CA 94143 69120 Heidelberg & Sackler Faculty of Medicine
USA GERMANY Tel Aviv University, Tel Aviv
Tel. + 1 415 476 5236 Tel. +49 6221 56 37 885 ISRAEL
Fax +1415476 7963 david.reuss@med.uni-heidelberg.de Tel. +972 3 530 32 62
arie.perry@ucsf.edu Fax +972 3 534 83 60
siegals@gertner.health.gov.il

346 Contributors
Dr Felix SAHM# Dr M.C. SHARMA Dr Dominik STURM
Department of Neuropathology Department of Pathology German Cancer Research Center and
Heidelberg University and Clinical All India Institute of Medical Sciences Heidelberg University Medical Center for
Cooperation Unit Neuropathology Ansari Nagar Children and Adolescents
German Cancer Research Center New Delhi 110029 Im Neuenheimer Feld 580
Im Neuenheimer Feld 224 INDIA 69120 Heidelberg
69120 Heidelberg Tel. +911165933 71 GERMANY
GERMANY Fax +91 11 686 26 63 Tel. +49 6221 42 46 76
Tel. +49 6221 56 378 86 sharmamehar@yahoo.co.in Fax +49 6221 42 46 39
felix.sahm@med.uni-heidelberg.de d.sturm@dkfz.de

Dr Sandro SANTAGATA# Dr Dov SOFFER Dr Mario L. SUV A


Department of Pathology Department of Pathology James Homer Wright Pathology Laboratories
Division of Neuropathology Tel Aviv Sourasky Medical Center Massachusetts General Hospital
Brigham and Women's Hospital. Harvard 6 Weizman Street 149 13th Street, Office 6010
Medical School, Harvard Institute of Medicine 64239 Tel Aviv Charlestown MA 02129
HIM-921, 77 Avenue Louis Pasteur ISRAEL USA
Boston MA 02115 Tel. +972 3 694 75 72 Tel. +1617726 5695
USA Fax +972 3 697 46 48 Fax +1617724 1813
Tel. + 1 617 525 5686 soffer@cc.huji.ac.il suva. mario@mgh. harvard. edu
ssantagata@partners.org

Dr Mariarita SANTI Dr Figen SOYLEMEZOGLU Dr Uri T ABORI


Division of Neuropathology Department of Pathology Paediatric Haematology/Oncology
Children's Hospital of Philadelphia Hacettepe University Hospital for Sick Children
34th Street and Civic Center Boulevard Tip Fakultesi, Patoloji Anabilim Dali 555 University Avenue
Philadelphia PA 19104 06100 Ankara Toronto ON M5G 1X8
USA TURKEY CANADA
Tel. +1215590 3184 Tel. +90 312 2419951 Tel. +1416813 8221
santim@chop.edu Fax +90 312 305 26 21 Fax +1416813 5327
figensoylemezoglu@gmail.com uri.tabori@sickkids.ca

Dr Vani SANTOSH Dr Anat 0. STEMMER-RACHAMIMOV Dr Shinya TANAKA


Department of Neuropathology Molecular Neuro-Oncology Laboratory Department of Cancer Pathology
National Institute of Mental Health and Massachusetts General Hospital Hokkaido University
Neuroscience CNY6. Building 149, 149 13th Street Graduate School of Medicine
Hosur Road Charlestown MA 02129 N15, W7
Bangalore 560029 USA Sapporo 060-8638
INDIA Tel.+ 1 617 726 5510 JAPAN
van i. santosh@gmai I. com Fax +1 617 726 5079 Tel. +81 11 706 50 52
astemmerrachamimov@partners.org Fax +81 11 706 59 02
tanaka@med.hokudai.ac.jp

Dr Chitra SARKAR Dr Constantine A. STRATAKIS Dr Tarik TIHAN


Department of Pathology Section on Endocrinology and Genetics Department of Pathology
All India Institute of Medical Sciences Eunice Kennedy Shriver National Institute of University of California, San Francisco Helen
Ansari Nagar Child Health and Human Development Diller Family Comprehensive Cancer Center
New Delhi 110029 Building 10, CRC, Room 1-3330 505 Parnassus Avenue, Moffitt
INDIA (East Laboratories). MSC 1103 San Francisco CA 94143-0511
Tel. +91 11 265 93 371 Bethesda MD 20892, USA USA
Fax +91 11 265 88 663 Tel. + 1 301 402 1998 Tel. +1415476 5236
sarkar .chitra@gmai I. com Fax +1 30140205.74 Fax +1415476 7963
stratakc@exchange.nih.gov tihan@itsa.ucsf.edu

Dr Davide SCHIFFER Dr Roger STUPP Dr Scott R. VANDENBERG


Policlinico di Monza Foundation Department of Oncology Department of Pathology
University of Turin University Hospital Zurich University of California
Via Cherasco 15, Corso Massimo D'Azeglio 51 Harnistrasse 100 San Diego School of Medicine
10126 Torino TO 8091 Zurich 9500 Gilman Drive, La Jolla
ITALY SWITZERLAND San Diego CA 92093
Tel. +39 011 696 44 79 Tel. +41 44 255 9779 USA
Fax +39 016 136 91 09 Fax +41 44 255 9778 Tel. + 1 858 534 0455
davide .schiffer@unito.it roger.stupp@usz.ch srvandenberg@mail. ucsd .edu

Contributors 347
Dr Erwin G. VAN MEIR Dr Alexander 0. VORTMEYER Dr Hai YAN#
Winship Cancer Institute Department of Pathology Molecular Oncogenomics Laboratory
Emory University School of Medicine Yale University School of Medicine Duke University
1365-C Clifton Road NE, Suite C 5078 310 Cedar Street, LH416 1998 MSRB, DUMC 3156
Atlanta GA 30322 New Haven CT 06520 Durham NC 27710
USA USA USA
Tel. + 1 404 778 5563 Tel. +1 203 785 6843 Tel. +1919668 7850
Fax+ 1 404 778 5550 Fax + 1 203 785 6899 hai.yan@duke.edu
evanmei@emory.edu alexander.vortmeyer@yale.edu

Dr Pascale VARLET# Dr Michael WELLER# Dr Stephen YIP


Laboratoire d'anatomie pathologique Department of Neurology Pathology and Laboratory Medicine
Centre hospitalier Sainte-Anne University Hospital Zurich University of British Columbia
1 rue Cabanis Frauenklinikstrasse 26 Vancouver General Hospital
75674 Paris Cedex 14 8091 Zurich 855 West 12th Avenue
FRANCE SWITZERLAND Vancouver BC V5Z 1 M9
Tel. +33 1 45 65 86 56 Tel. +41 44 255 55 00 CANADA
Fax +33 1 45 65 87 28 Fax +41 44 255 45 07 Tel. + 1 604 875 4111
p.varlet@ch-sainte-anne.fr michael.weller@usz.ch Fax + 1 604 875 4 797
stephen. yi p@vch.ca

Dr Alexandre VASILJEVIC Dr Pieter WESSELING* Dr Hideaki YOKOO


Centre de Pathologie et de Department of Pathology Department of Human Pathology
Neuropathologie Est VU University Medical Center Amsterdam and Gunma University
Groupement Hospitalier Est Radboud University Medical Center Nijmegen 3-39-22 Showa
59 boulevard Pinel Box 9101 Maebashi 371-8511
69677 Bron Cedex 6500 HB Nijmegen JAPAN
FRANCE THE NETHERLANDS Tel. +81 27 220 71 11
Tel. +33 4 72 12 96 04 Tel. +31 24 361 43 23 Fax +81 27 220 79 78
Fax +33 4 72 35 70 67 Fax +31 24 366 87 50 hyokoo@gunma-u.ac.jp
alexandre.vasiljevic@chu-lyon.fr pieter.wesseling@radboudumc.nl

Dr Roel G.W. VERHAAK Dr Wolfgang WICK*# Dr Tarek YOUSRY


Department of Genomic Medicine Neurology Clinic Division of Neuroradiology and Neurophysics
Department of Bioinformatics and Heidelberg University Hospital UCL Institute of Neurology
Computational Biology, University of Texas Im Neuenheimer Feld 400 Lysholm Department of Neuroradiology,
MD Anderson Cancer Center 69120 Heidelberg NHNN, University College London Hospitals
1400 Pressler Street, Unit #141 O GERMANY Queen Square
Houston TX 77030 Tel. +49 6221 56 70 75 London WC 1 N 3BG
USA Fax +49 6221 56 75 54 UNITED KINGDOM
Tel. + 1 713 563 2293 wolfgang. wick@med. u ni-hei de I berg. de t. yousry@ucl.ac. uk
Fax+ 1 713 563 4242
rverhaak@mdanderson.org
Dr Harry V. VINTERS# Dr Otmar D. WIESTLER* Dr David ZAGZAG
UCLA Pathology & Laboratory Medicine German Cancer Research Center Department of Pathology and Neurosurgery
David Geffen School of Medicine at UCLA Im Neuenheimer Feld 280 NYU Medical Center and School of Medicine
Box 951732, 18-1708 NPI 69120 Heidelberg 550 First Avenue
Los Angeles CA 90095-1732 GERMANY New York NY 10016
USA Tel. +49 6221 42 26 53 USA
Tel. +1310825 6191 Fax +49 6221 42 21 62 Tel.+ 1 212 263 2262
hvinters@mednet.ucla.edu o.wiestler@dkfz.de Fax +1212263 7916
dz4@nyu.edu

Dr Andreas VON DEIMLING* Dr Hendrik WITT


Institute of Pathology, Division of Department of Pediatric Oncology
Neuropathology and Clinical Cooperation Unit Heidelberg University
Neuropathology, Heidelberg University and Division of Pediatric Neurooncology
German Cancer Research Center German Cancer Research Center
Im Neuenheimer Feld 224 Im Neuenheimer Feld 430
69120 Heidelberg 69120 Heidelberg
GERMANY GERMANY
Tel. +49 6221 56 46 50 Tel. +49 6221 56 357 02
andreas.vondeimling@med.uni-heidelberg.de Fax +49 6221 56 455 5
h.witt@dkfz.de

348 Contributors
Declaration of interest statements

Dr Capper reports receiving income from two Dr Mclendon reports being part of a pending Dr Varlet reports receiving travel support
patents held by the German Cancer Research patent application, with Duke University, for a from Hoffmann-La Roche and Boehringer
Center (DKFZ): one for an antibody to detect technique to identify and measure FUBP1 and lngelheim. Dr Varlet reports that a close rela-
R132H-mutant IOH1 in fixed glioma samples, CIC in brain tumours. tive is employed by Novartis France.
licensed to Dianova, and one for an antibody
to detect V600E-mutant BRAF in formalin-fixed Dr Osborn reports having been a founder and Dr Vinters reports that the UCLA Department
paraffin-embedded tumour samples, licensed shareholder of Amirsys and Amirsys Publish- of Pathology and Laboratory Medicine con-
to Ventana Medical Systems (a member of the ing. Dr Osborn reports receiving personal ducts contract work for Neuroindex funded
Roche Group). consultancy fees from Elsevier. Or Osborn re- by an NIH grant. Or Vinters reports receiving
ports having received personal speaker's fees royalties from Mosby Elsevier. Or Vinters re-
Dr Hainfellner reports that the Austrian Brain from Mallinckrodt. ports holding shares in 3M Company; Gen-
Tumour Registry at the Institute of Neurology, eral Electric; Teva Pharmaceutical Indus-
Medical University of Vienna, receives re- Dr Pfister reports receiving non-financial re- tries; Pfizer: GlaxoSmithKline; and Becton,
search support from Roche. search support from lllumina. Or Pfister re- Dickinson and Company.
ports having a patent pending for a brain tu-
Dr Hartmann reports having received per- mour classification based on DNA methylation Dr Weller reports receiving personal con-
sonal consultancy fees from Apogenix. Or fingerprinting. sultancy fees from MagForce AG, lsar-
Hartmann reports receiving income from a na Therapeutics, Celldex Therapeutics,
patent held by OKFZ for an antibody to detect Dr Pietsch reports having received travel sup- lmmunoCellular Therapeutics, Roche, and
R132H-mutant IDHi, licensed to Oianova port from Affymetrix. Dr Pietsch reports having Merck Serono. Or Weller reports receiving per-
received personal speaker's fees from Roche. sonal speaker's fees from lsarna Therapeutics,
Or Hegi reports that her department at MSD, Roche, and Merck Serono. Dr Weller
the Lausanne University Hospital has re- Dr Reifenberger reports having received re- reports that the University Hospital Zurich
ceived non-financial research support from search funding from Roche and Merck and and University of Zurich receive research
MOxHealth. honoraria for advisory boards or lectures from support from Roche, Merck Serano, Bayer,
Amgen, Roche and Celldex. lsarna Therapeutics, Piqur Therapeutics, and
Dr lchimura reports receiving research sup- Novocure.
port from SRL. Dr lchimura reports receiving Dr Rushing reports having received research
travel support from MSD K.K. (a subsidiary funding from Novartis. Dr Wick reports holding patents related to
of Merck & Co.). Dr lchimura reports receiv- CD95L as a diagnostic marker, IOH1 immuno-
ing personal speaker's fees from Eisai Co., Dr Sahm reports having a patent pending for a therapy, and IOH1 antibody.
Astellas Pharma, Otsuka Pharmaceutical, method for detecting the presence of antigens
Sanofi K.K., Daiichi Sankyo, Chugai Pharma- in situ. Dr Yan reports being the chief security officer
ceutical, and Teijin Pharma. of Genentron Health and having substantial
Dr Santagata reports havinq cofounded shareholding in the company. Or Yan reports
Dr Jones reports receiving non-financial re- Bayesian Diagnostics. Dr Santagata reports receiving personal consultancy fees from
search support from lllumina. Or Jones reports having received personal consultancy fees Sanofi and Blueprint Medicines and reports re-
having a patent pending for a brain tumour from Bayesian Diagnostics. Dr Santagata re- ceiving royalties for I DH-targeted therapy and
classification based on ONA methylation fin- ports that a close relative receives royalties diagnosis from Agios and Personal Genome
gerprinting. from BioReference Laboratories for intellectual Diagnostics. Or Yan reports having received
property rights for a targeted genotyping plat- royalties from Blueprint Medicines. Dr Yan re-
Dr Leung reports that the Department of form for cancer diagnostics. ports having a patent pending for /OH 1 and
Pathology at the University of Hong Kong TEAT mutations in gliomas. Dr Yan reports that
has a collaborative research agreement with his laboratory at the Duke University Medical
Merck, Pfizer, and Servier. Center received research support from Gilead
Sciences and Sanofi.

Declaration of interest statements 349


IARC/WHO Committee for the International Classification of
Diseases for Oncology (ICD-0)

Dr Freddie BRAY Dr Robert JAKOB Dr Hiroko OHGAKI


Section of Cancer Surveillance Data Standards and Informatics Section of Molecular Pathology
International Agency for Research on Cancer Information, Evidence and Research International Agency for Research on Cancer
150 Cours Albert Thomas World Health Organization (WHO) 150 Cours Albert Thomas
69372 Lyon Cedex 08 20 Avenue Appia 69372 Lyon Cedex 08
FRANCE 1211 Geneva 27 FRANCE
Tel. +33 4 72 73 84 53 SWITZERLAND Tel. +33 4 72 73 85 34
Fax +33 4 72 73 86 96 Tel. +4122791 58 77 Fax +33 4 72 73 86 98
brayf@iarc.fr Fax +41 22 791 48 94 ohgakih@iarc.fr
jakobr@who.int

Dr David W. ELLISON Dr Paul KLEI HUES Dr Marion PINEROS


Department of Pathology Faculty of Medicine Section of Cancer Surveillance
St. Jude Children's Research Hospital University of Zurich International Agency for Research on Cancer
262 Danny Thomas Place, MS 250 Pestalozzistrasse 5 150 Cours Albert Thomas
Room C-5001 8032 Zurich 69372 Lyon Cedex 08
Memphis TN 38105-3678 SWITZERLAND FRANCE
USA Tel. +41 44 362 21 10 Tel. +33 4 72 73 84 18
Tel.+ 1 901 595 5438 kleihues@pathol.uzh.ch Fax +33 4 72 73 80 22
Fax +1 9015953100 pinerosm@iarc.fr
david.ellison@stjude.org

Mrs April FRITZ Dr David N. LOUIS Dr Brian ROUS


A. Fritz and Associates, LLC James Homer Wright Pathology Laboratories National Cancer Registration Service -
21361 Crestview Road Massachusetts General Hospital Eastern Office
Reno NV 89521 55 Fruit Street, Warren 225 Victoria House. Capital Park
USA Boston MA 02114 Fulbourn, Cambridge CB21 5XB
Tel. + 1 775 636 7243 USA UNITED KINGDOM
Fax + 1 888 891 3012 Tel. + 1 617 726 2966 Tel. +44 122 321 3625
april@afritz.org Fax + 1 617 726 7533 Fax +44 122 321 3571
dlouis@mgh.harvard.edu brian. rous@phe.gov. uk

Dr Leslie H. SOBIN
Frederick National Laboratory for
Cancer Research
The Cancer Human Biobank
National Cancer Institute
6110 Executive Boulevard, Suite 250
Rockville MD 20852, USA
Tel. + 1 301 443 7947
Fax + 1 301 402 9325
leslie.sobin@nih.gov

350 ICD-0 Committee


Sources of figures and tables

Sources of figures 1.33A-D Reprinted from Rong Y, neuropathology. PB Medical


Durden DL, Van Meir EG, Publishing: Baltimore.
1 01 Louis ON et al., 'Pseudopalisading' 1.45A,B Kleihues P
1.02 From N Engl J Med, Yan H. necrosis in glioblastoma: a 1.46A,B Kleihues P
Parsons OW, Jin G, et al., IDH1 familiar morphologic feature 1.46C Nakazato Y
and IDH2 mutations in gliomas, that links vascular pathology, 1.47 Kleinschmidt-DeMasters BK
360, 765-73. Copyright© hypoxia. and angiogenesis, J 1.48A,B Kleinschmidt-DeMasters BK
2009 Massachusetts Medical Neuropathol Exp Neurol. 2006, 1.49A,B Ellison OW
Society. Reprinted with 65, 6, 529-39, by permission 1.49C Kleinschmidt-DeMasters BK
permission from Massachusetts of Oxford University Press I 1.50 Adapted from Ohgaki H,
Medical Society. the American Association of Kleihues P (2013). The
1.03 Reuss DE Neuropathologists, Inc. definition of primary and
1.04A,B Kleihues P 1.34 © 2005 Mica Duran, secondary glioblastoma. Clin
1 05A Nakazato Y http://www.micaduran.com. Cancer Res. 15;19:764-72.
1 05B Rushing EJ info@micaduran.com. 1.51 IARC; based on combined data
1 06 Perry A Adapted from Rong Y, from Nobusawa S et al. [ 17971
1 07 Kros JM Durden DL. Van Meir EG, and The Cancer Genome Atlas
1 08A,B Nakazato Y et al., 'Pseudopalisading' Research Network {350)
1.09A,B Fuller GN necrosis in glioblastoma: a 1.52 Kleihues P
1.10A Nobusawa S familiar morphologic feature 1.53 CapperD
Department of Human that links vascular pathology, 1.54A Reprinted from Nobusawa
Pathology, Gunma University hypoxia, and angiogenesis, J S, Watanabe T, Kleihues P,
Graduate School of Medicine, Neuropathol Exp Neurol. 2006, et al. (2009). IDH1 mutations
Gunma, Japan 65, 6, 529-39, by permission as molecular signature and
1.108 Kleihues P of Oxford University Press I predictive factor of secondary
1.10C Ohgaki H the American Association of glioblastomas. Clin Cancer
1.11A-C Kleihues P Neuropathologists, Inc. Res. 15:6002-7.
1.12 Burger PC 1.35 Kleihues P 1.548 From N Engl J Med, Yan H,
1.13 Rushing EJ 136A Kleihues P Parsons OW, Jin G, et al., IDH1
1.14A Burger PC 1.36B Iwasaki Y (deceased) and IDH2 mutations in gliomas.
1.148 Perry A 1.37 Adapted from Suva ML. 360. 765-73. Copyright©
1.15A Ohgaki H Riggi N, Bernstein BE (2013) 2009 Massachusetts Medical
1.15B Perry A Epigenetic reprogramming Society. Reprinted with
1.15C Kleihues P in cancer. Science. permission from Massachusetts
1.16 From N Engl J Med, Cancer 339(6127) 1567-70. Reprinted Medical Society.
Genome Atlas Research with permission from AAAS. 1.55A-D Laughlin S
Network, Brat DJ, Verhaak 1.38 From N Engl J Med. Hegi ME, Department of Diagnostic
RG, et al., Comprehensive, Diserens AC, Gorlia T, et al., Imaging
integrative genomic analysis MGMT gene silencing and Hospital for Sick Children
of diffuse lower-grade gliomas. benefit from temozolomide Toronto (ON), Canada
372, 2481-98. Copyright© in glioblastoma, 352, 1.56 Reprinted from Solomon DA,
2015 Massachusetts Medical 997-1003. Copyright © 2005 Wood MD, Tihan T, et al.
Society. Reprinted with Massachusetts Medical (2015). Diffuse midline
permission from Massachusetts Society. Reprinted with gliomas with histone H3-K27M
Medical Society. permission from Massachusetts mutation: a series of 47 cases
117 IARC; based on data from Medical Society. assessing the spectrum of
Nobusawa Set al. [17971 1.39 Reprinted from Taylor TE, morphologic variation and
1.18A-C Perry A Furnari FB, Cavenee WK associated genetic alterations.
1.19 Burger PC (2012). Targeting EGFR for Brain Pathol. doi: 10.1111/
1.20A,B Kleihues P treatment of glioblastoma: bpa.12336. [Epub ahead of
1.21A-D Kleihues P molecular basis to overcome print]© 2015 International
1.22 Burger PC resistance. Curr Cancer Drug Society of Neuropathology;
1.23 Kleihues P Targets. 12(3):197-209. with permission of John Wiley
1.24A,B Louis ON Reprinted by permission of & Sons.
1.25A-C Perry A Eureka Science Ltd.© 2012 1.57 Hawkins C
1.26A-C Perry A Bentham Science Publishers. 1.58A Ellison OW
1.27A-D Perry A 1.40 Nakazato Y 1.588 Hawkins C
1.28A Nakazato Y 1.41 Burger PC 1.59 Perry A
1.28B Reifenberger G 1.42 Nakazato Y 1.60 IARC, based on combined
1.29A,B Perry A 1.43A-C Nakazato Y data from the German Glioma
1.29C lwasakiY(deceased) 1.430 Kleihues P Network (Reifenberger G) and
1.30A,B Perry A 1.44 Reprinted from Burger the University of Heidelberg
1.31 Louis ON PC (2009). Smears and (Capper D, von Deimling A)
1.32 Perry A frozen sections in surgical 1.61A,B Kleihues P
1

Sources of figures 351


,>'

1.62 Kleihues P 2.15A,B Ornan DA 3.19C Nakazato Y


1.63A,C Nakazato Y Department of Radiology and 3.20A,B Ellison OW
1.638 Yip S Medical Imaging
1.630 Nobusawa S University of Virginia School of 4.01 Brat DJ
Department of Human Medicine, Charlottesville (VA). 4.02 Brat DJ
Pathology, Gunma University USA 4.03A-F Brat DJ
Graduate School of Medicine. 2.16A Vonsattel J-P 4.04 Brat DJ
Gunma. Japan Columbia University 4.05A Tihan T
1.64A Reifenberger G New York (NY), USA 4.058 Burger PC
1 648,C Nakazato Y 2.168 Paulus W 4.06A Figarella-Branger D
1.65A,B Nakazato Y 2.17A,B Santosh V 4.068 Fuller GN
1.66A,C Yip S 217C Lopes MBS 4.07A,C,D Burger PC
1.668 Nakazato Y 2.18A Lopes MBS 4.078 Figarella-Branger D
1.67 Reifenberger G 2.188 Perry A 4.08A,B Brat DJ
1.68 Kleihues P 2.19A,B Sharma MC 4.09A-C Brat DJ
1.69A,D Reifenberger G 2.19C-F Lopes MBS 4.090 Rushing EJ
1.698 Nakazato Y 2.20 Giannini C
1.69C VandenBerg SR 2.21A,B Giannini C 5.01 Paulus W
1.70 Reifenberger G 2.22A-C Giannini C 5.02 Figarella-Branger D
1.71A,B Reifenberger G 2.220 Reprinted from Hum Pathol, 5.03A Rosenblum MK
1.72 Kleihues P 22(11), Kros JM, Vecht CJ, 5.038 Rorke-Adams LB
1.73 Reprinted from Acta Stefanko SZ, The pleomorphic 5.04A Paulus W
Neuropathol, ATRX and IDH1- xanthoastrocytoma and its 5.048 Figarella-Branger D
R 132H immunohistochemistry differential diagnosis: a study of 5.05A,B Paulus W
with subsequent copy number five cases. 1128-35, Copyright 5.06 Fuller GN
analysis and IDH sequencing (1991), with permission from 5.07 Vital A
as a basis for an "integrated" Elsevier. Service de Pathologie
diagnostic approach 2.23A,B Giannini C Centre Hospitalier Universitaire
for adult astrocytoma, 2.24A-D Giannini C de Bordeaux
oligodendroglioma and 2.25 Giannini C Bordeaux. France
glioblastoma. 129, 2015. 2.26A-D Giannini C 5.08 Louis ON
133-46, Reuss DE, Sahm F, 5.09A Brandner S
Schrimpf D. et al.,© Springer- 3.01 Mclendon R 5.098 Nakazato Y
Verlag Berlin Heidelberg 2014; 3.02A-D Kleihues P 5.10A,C Paulus W
With permission of Springer. 3.03A Rushing EJ 5.108 Fuller GN
3.038,C Ellison OW 5.100 Kleihues P
2.01 Kleihues P 3.04 Mclendon R
2.02A-F Giannini C 3.05 Mclendon R 6.01 Pietsch T
2.03A-C Koeller K 3.06A,C Mclendon R 6.02A BIOmcke I
Department of Radiology 3.068 Santi M 6.028 Hattingen E
Mayo Clinic 3.060 Wiestler OD Institute of Neuropathology
Rochester (MN). USA 3.07 Schiffer D University of Bonn Medical
2.04A,B Kleihues P 3.08A Santi M Center, Bonn, Germany
2.04C Paulus W 3.088 Westphal MM 6.03 BIOmcke I
2.05A-F Giannini C Department of Neurosurgery 6.04 BIOmcke I
2.06A Perry A University Cancer Center 6.05A,B Perry A
2.068 Figarella-Branger D Hamburg 6.06A,B Varlet P
2.07A,B Figarella-Branger D Hamburg, Germany 6.07 Varlet P
2.08 Adapted by permission from 3.09 Kleihues P 6.08A Kleihues P
Macmillan Publishers Ltd: Nat 3.10A Louis ON 6.088.C BIOmcke I
Genet. Jones DTW, Hutter 3.108,C Perry A 6.09A,B Giannini C
B, Jager N, et al. (2013). 3.100 Kleihues P 6.09C,D BIOmcke I
Recurrent somatic alterations of 3.11A Kleihues P 6.10 BIOmcke I
FGFR1 and NTRK2 in pilocytic 3.118,C Mclendon R 6.11A Osborn A
astrocytoma. 45(8) 927-32, 3.12A Perry A 6.118 Urbach H
copyright 2013; Reprinted 3.128 .Nakazato Y Department of Neuroradiology
from Collins VP, Jones DTW. 3.12C Kleihues P University Medical Center
Giannini C (2015). Pilocytic 3.13 Nakazato Y Freiburg, Freiburg im Breisgau,
astrocytoma: pathology, 3.14A Ellison OW Germany
molecular mechanisms and 3.148 Rosenblum MK 6.12 Kleihues P
markers. Acta Neuropathol. 3.15 Ellison OW 6.13 BIOmcke I
129(6)775-88. 3.16 Ellison OW 6 14A,C BIOmcke I
2.09 Reprinted from Collins VP, 3.17 Westphal MM 6.148,D Varlet P
Jones DTW, Giannini C Department of Neurosurgery 6.15A Kleihues P
(2015). Pilocytic astrocytoma: University Cancer Center 6.158 Perry A
pathology, molecular Hamburg 6.15C Capper D
mechanisms and markers. Acta Hamburg, Germany 6.16A,B Perry A
Neuropathol. 129(6):775-88. 3.18 Jellinger K 6.17 Eberhart CG
2.10 See 2.09 Institute of Clinical 6.18 Allen SH
2.11 Tihan T Neurobiology Carolina Radiology Associates,
2.12 Tihan T Vienna. Austria Mcleod Regional Medical
2.13A,B Tihan T 3.19A Rosenblum MK Center
2.14 Giannini C 3.198 Schiffer D Florence (SC), USA

352 Sources of figures


6.19A Reifenberger G 6.53A Zrinzo L 8.10 Ellison OW
6198 Nakazato Y National Hospital for Neurology 8.11A,B Giangaspero F
6.20A-C Eberhart CG and Neurosurgery 8.11C Kleihues P
621 Brat DJ London, United Kingdom 8.12A,B Kleihues P
6.22A Osborn A 6.538 Jaunmuktane Z 8.12C Rorke-Adams LB
6.228 Taratuto AL Department of Neuropathology 8.13A,B Ellison OW
6.23A Taratuto AL UCL Institute of Neurology 8.14 Eberhart CG
6.238,C Nakazato Y London, United Kingdom 8.15 Pietsch T
6.230 Brat DJ 6.54 Yousry T 8.16A,B Warmuth-Metz M
6.24A Brat DJ 6.55 Yousry T Department of Neuroradiology
6.248 Taratuto AL 6.56 Brandner S University Hospital WOrzburg
6.24C Rorke-Adams LB 6.57A-C Yousry T Wurzburg, Germany
6.25A-D Park S-H 6.570 Perry A 8.16C Perry A
6.26A Figarella-Branger D 8.17A,C,D Pietsch T
6.268 Nakazato Y 7.01 Jouvet A 8.178 Perry A
6.27A-D Nakazato Y 7 02 Sasajima T 8.18A Giangaspero F
6.28A-C Nakazato Y Department of Neurosurgery 8.188 Pietsch T
6.29A,B Hainfellner JA Akita University Graduate 8.19 Pietsch T
6.30 Hainfellner JA School of Medicine. Hondo, 8.20A,B Doz F
6.31A-C Hainfellner JA Akita, Japan Department of Paediatric
6.32 Gessi M 7.03 Nakazato Y Oncology, lnstitut Curie
6.33 Rodriguez F 7.04A Vasiljevic A Paris, France
6.34A Krawitz S 7.048 Nakazato Y 8.20C Garre ML
Department of Pathology 7.05A.B Nakazato Y Neuroncology Unit
University of Manitoba 706A Vasiljevic A lstituto Giannina Gaslini
Winnipeg (MB), Canada 7.068,C Nakazato Y Genoa, Italy
6348,C Perry A 7.07A-C Nakazato Y 8.21A,B Giangaspero F
6.35A Perry A 7 08 Vasiljevic A 8.21C,D Ellison OW
6.358-0 Rodriguez F 7.09A JouvetA 8.22A,B Ellison OW
6.36A-C Perry A 7 098 Fevre Montange M 8.23 Eberhart CG
6.37A-C Perry A 7.10A Fevre Montange M 8.24 Korshunov A
6.38 CapperD 7.108 Vasiljevic A 8.25 Korshunov A
6.39 Soylernezoqlu F 7.11A Fevre Montange M 8.26A-C Korshunov A
6.40A,B Figarella-Branger D 7.118 Vasiljevic A 8.27A-C Korshunov A
6.41 Soffer D 7.11C Nakazato Y 8.28 Korshunov A
6.42A Vasiljevic A 7.12 Vasiljevic A 8.29 Korshunov A
6.428 Kleihues P 7.13 Osborn A 8.30 Korshunov A
6.43A Kleihues P 7.14 Taratuto AL 8.31 Korshunov A
6.438,E,F Figarella-Branger D 7.15A Vasiljevic A 8.32 Korshunov A
6.43C Burger PC 7.158 Rosenblum MK 8.33 Ellison OW
6.430 Vasiljevic A 7.15C Ellison OW 8.34A,B Perry A
6.44A,B Honavar M 7.150 Kros JM 8.35 Judkins AR
1 6.45A Furtado A 7.16A-C Vasiljevic A 8.36 Tamrazi B
Anatomic Pathology Service 7.17A-D Fevre Montange M Department of Radiology
Vila Nova de Gaia-Espinho 7.18 Vasiljevic A Children's Hospital Los Angeles
Medical Center 7.19 Figarella-Branger D Los Angeles (CA), USA
Vila Nova de Gaia, Portugal 7.20A-C Vasiljevic A 8.37 Rorke-Adams LB
6.458-F Honavar M 7.21A-C Vasiljevic A 8.38A Judkins AR
6.46 Ohgaki H 8.388 Wesseling P
6.47A Reprinted from Jenkinson 8.01 Pietsch T 8.39A-C Judkins AR
MD, Bosma JJ, Du Plessis 8.02A-C Kleihues P 8.40A Wesseling P
D, et al. (2003). Cerebellar 8.03A,B Kleihues P 8.408-D Judkins AR
liponeurocytoma with 8.04 Kleihues P 8.41A,B Ellison OW
an unusually aggressive 8.05A Nakazato Y 8.41C Hasselblatt M
clinical course: case report. 8.058 Kalimo H
Neurosurgery. 53(6): 1425-7; 8.05C Wiestler OD 9.01 Kleihues P
discussion 1428. 8.050 Garcia-Bragado F 9.02A,B Perry A
6.478 Kollias S J. Servicio Anatomia Patol6gica 9.03A,B Perry A
Department of Neuroradiology Complejo Hospitalario de 9.04A,B Kleihues P
University Hospital Zurich Navarra 9.05A,B Perry A
Zurich, Switzerland Pamplona, Spain 9 06A-C Perry A
6.48A Ohgaki H 806 Palay Z 9.07 Perry A
6.488 Kleihues P Department of Diagnostic 9.08A-D Perry A
6.49A Giangaspero F Imaging, St. Jude Children's 9.09A Stemmer-Rachamimov AO
6.498,C Ohgaki H Research Hospital 9098 Perry A
6.50 Cenacchi G Memphis (TN). USA 9.10 Woodruff JM (deceased)
Department of Biomedical and 807A-C Ellison OW 9.11A-C Perry A
Neuromotor Sciences 808 Palay Z 9.12 Perry A
University of Bologna Department of Diagnostic 9.13 Budka H
Bologna, Italy Imaging, St. Jude Children's 9.14A,B Perry A
6.51 Ohgaki H Research Hospital 9.15A,B Perry A
6.52 Soffer D Memphis (TN). USA 9.16A,B Perry A
8.09A,B Hawkins C 9.17A.B Perry A

Sources of figures 353


9.18 Perry A 11.15C,D Perry A 16.05A,B Plate KH
9.19A-C Perry A 11.16 Plate KH 16.06 Louis DN
9.20A-C Folpe A 11.17 Perry A 16.07 Wiestler OD
Department of Laboratory 11.18 Perry A 16.08A,B Wiestler OD
Medicine and Pathology 11.19A,B Perry A 1609A Wiestler OD
Mayo Clinic 11.20A-C Perry A 16.098 Louis DN
Rochester (MN), USA 11.21A-C Perry A 16.10A,B Louis DN
9.21A-C Antonescu CR 11.22 Perry A 16.10C Wiestler OD
9.22 Scheithauer BW (deceased) 11.23A-C Perry A 16.11 Stemmer-Rachamimov AO
9.23A,B Perry A 11.24 Perry A 16.12 Salamipour H
9.24A,B Reuss DE 11.25A-C Perry A Newton-Wellesley Radiology
9.24C-F Perry A 11.26 Perry A Associates, Newton-Wellesley
9.25A,B Perry A 11.27A,B Perry A Hospital, Newton (MA), USA
9.25C Woodruff JM (deceased) 11.28A-D Perry A 16.13 Stemmer-Rachamimov AO
9.26A,C Reuss DE 11.29A,B Perry A 16.14 Hulsebos T JM
9.268 Woodruff JM (deceased) 11.30A,B Perry A 16.15A,B Neumann HPH
9.27A-C Perry A 16.16 Neumann HPH
9.28A-D Hirose T 12.01A,B Nakazato Y 16.17 Perry A
12 02A,B Rorke-Adams LB 16.18 Moch H
1001 Claus EB 12.03A-C Wesseling P Department of Pathology
10.02A Osborn A 12.04A-C Brat DJ University Hospital Zurich
10028 Perry A 12 05 Louis DN Zurich, Switzerland
10.03 Kleihues P 12.06 Wesseling P 16.19A-D Vinters HV
1004A-C,E Kleihues P 16.20 Olivier M
10.04D,F Budka H 13.01 Paulus W 16.21 Reprinted from Olivier M,
10.05 Kleihues P 13.02 Perry A Goldgar DE, Sodha N, et
1006 Mawrin C 1303 Haltia MJ al. (2003). Li-Fraumeni and
10.07 Perry A 13.04 Nakazato Y related syndromes: correlation
10.08A Santi M 13.05A Nakazato Y between tumor type, family
10.088 Perry A 13.05B-D Deckert M structure, and TP53 genotype.
10.09 Perry A 1306 Deckert M Cancer Res. 15;63:6643-50.
10.10A-C Perry A 13.07 Deckert M 16.22 Olivier M
10.11 Perry A 13.08A,B Kleihues P 16.23 Reprinted from Acta
10.12A-C Perry A 13.09 Deckert M Neuropathol. Selective
10.13 Perry A acquisition of IDH1 R132C
1014A,B Perry A 14.01 Perry A mutations in astrocytomas
10.15 Perry A 14.02A Peiffer J (deceased) associated with Li-Fraumeni
1016A,B Perry A 14 028-D Perry A syndrome, 117, 2009, 653-6,
1017 Perry A 14.03 Perry A Watanabe T, Vital A, Nobusawa
10.18A,B Perry A 1404A-C Perry A S, et al.,© Springer-Verlag
10.19 Perry A 1405 Perry A 2009; With permission of
1020A,C,D Perry A 14.06A,B Perry A Springer.
10208 Nakazato Y 16.24 Olivier M
10.21A-C Perry A 15.01 Matsutani M 16.25 Reprinted by permission from
10.22A-C Perry A 1502 Rosenblum MK Macmillan Publishers Ltd: Eur
10.23A,B Perry A 15.03 Nakazato Y J Hum Genet Blumenthal GM,
10.24A,B Perry A 15 04A,C Rosenblum MK Dennis PA PTEN hamartoma
10.25A,B Perry A 15 04B,D Nakazato Y tumor syndromes. 16: 1289-
10.26A-C Perry A 1505A Rosenblum MK 300. Copyright 2008.
10.27A-C Perry A 15 05B-D Nakazato Y 16.26 Eberhart CG
10.28 Perry A 15.06 Rosenblum MK 16.27 Adapted from Lindstrom E,
10.29A,B Perry A 15.07A-D Rosenblum MK Shimokawa T, Toftqard R, et
10.30A-C Perry A 15.08 Rosenblum MK al. (2006). PTCH mutations:
10.31A-F Perry A 15.09 Westphal MM distribution and analyses. Hum
1032A-C Perry A Department of Neurosurgery Muta!. 27(3):215-9. ©2006
10.33A-C Perry A University Cancer Center WILEY-LISS, INC.
Hamburg
11 01 Giannini C Hamburg, Germany 17 01 Paulus W
11.02A-C Giannini C 15.10 Olvera-Rabiela JE (deceased), 17 02A Perry A
11 03A-D Giannini C Rosenblum MK 17.028 Kleihues P
11.04A,B Giannini C Department of Pathology 17 03 Perry A
11.05A-D Giannini C Memorial Sloan-Kettering 17.04 Kleihues P
11.06A,B Giannini C Cancer Center, New York (NY), 17.05 Buslei R
11.07A-C Bouvier C USA 17.06 Perry A
11.08A-D Bouvier C 17.07 Paulus W
11.09 von Deimling A 16.01 Kros JM 17.08A,B Buslei R
11.10A-C Zagzag D 1602 Nelson JS 1709A Perry A
11.11A,B Zagzag D Department of Pathology 17.098,C Buslei R
11.12 Zagzag D Louisiana State University 17.10A,B Perry A
11.13A,B Zagzag D Medical Center 17.11A-C Perry A
11.14A Nakazato Y New Orleans (LA), USA 17.110 Buslei R
11.148 Perry A 16.03 Burger PC 17.12A,B Santagata S
11.15A,B Zagzag D 16.04 Perry A 17.13 Fuller GN

354 Sources of figures


17 14A,B Fuller GN 1 04 Adapted from Oh JE, Ohta
17 15A,B Fuller GN T, Nonoguchi N, et al.
17 16A,B Fuller GN (2015). Genetic alterations
17 17A,B Fuller GN in gliosarcoma and giant cell
17.18 Brat DJ glioblastoma. Brain Pathol.
17 19 Brat DJ PM ID: 26443480.
17 20A.B Brat DJ 1.05 Ohgaki H, Kleihues P, van
17.20C Lopes MBS Deimling A, Louis ON,
1721A,B Ornan DA Reifenberger G, Yan H, Weller M
Department of Radiology and 1.06 Hawkins C, Ellison OW, Sturm D
Medical Imaging 1.07 Kleihues P, Reifenberger G
University of Virginia
Charlottesville (VA), USA 2.01 Adapted from Collins VP,
17 22A,B Roncaroli F Jones DTW, Giannini C
17 23A,C Lopes MBS (2015). Pilocytic astrocytoma
17.238 Roncaroli F pathology, molecular
17.24A Lopes MBS mechanisms and markers. Acta
17.248,C,E,F Roncaroli F Neuropathol. 129(6):775-88.
17.240 Perry A
17.25 Lopes MBS 3.01 Ellison OW
17.26 Roncaroli F
6.01 Blurncke I
18.01 Wesseling P
18 02A,B Westphal MM 8.01 Ellison OW
Department of Neurosurgery 8.02 Ellison OW
University Cancer Center 8.03 Mclendon R
Hamburg
Hamburg, Germany 10.01 Louis ON, Perry A
18.03A-C Kleihues P
1804A Wesseling P 11.01 Plate KH, Aldape KO,
18.048 Kleihues P Vortmeyer AO, Zagzag D,
1804C Wesseling P Neumann HPH
18.05A Wesseling P
18.058 Wesseling P 16.01 Kleihues P
18.05C Wesseling P 16.02 van Deimling A
16.03 Kleihues P
Sources of figures on front cover 16.04 Stemmer-Rachamimov AO,
Hulsebos TJM, Wesseling P
Top left Kleihues P 16.05 Plate KH, Vortmeyer AO,
Top centre Perry A Zagzag D, Neumann HPH,
Top right Wiestler OD Aldape KO
Middle left Perry A 16.06 Plate KH, Vortmeyer AO,
Middle centre Perry A Zagzag D, Neumann HPH,
Middle right Rorke-Adams LB Aldape KO
Bottom left Adapted by permission 16.07 Adapted from Pediatr Neural,
from Macmillan Publishers 49(4), Northrup H, Krueger
Ltd: Nat Genet. Jones DA, International Tuberous
DTW, Hutter B. Jager N, Sclerosis Complex Consensus
et al. (2013). Recurrent Group, Tuberous sclerosis
somatic alterations of FGFR1 complex diagnostic criteria
and NTRK2 in pilocytic update: recommendations of
astrocytoma. 45(8):927-32, the 2012 International Tuberous
copyright 2013; Reprinted Sclerosis Complex Consensus
from Collins VP, Jones DTW, Conference, 243-54, Copyright
Giannini C (2015). Pilocytic 2013, with permission from
astrocytorna pathology, Elsevier.
molecular mechanisms and 16.08 Sharma MC
markers. Acta Neuropathol. 16.09 Olivier M; based on data from
129(6):775-88. the IARC TP53 Database (R17,
Bottom centre Perry A November 2013)
Bottom right Adapted from Suva 16.10 Eng C
ML, Riggi N, Bernstein
BE (2013). Epigenetic 18.01 Adapted from Pekmezci M,
reprogramming in cancer. Perry A (2013). Neuropathology
Science. 339(6127):1567-70. of brain metastases. Surg
Reprinted with permission Neural Int. 4(Suppl 4):S245-55.
from AAAS.

Sources of tables

1.01 Kleihues P
1.02 Ellison OW
1.03 Cavenee WK

Sources of tables 355


References

1. Abe M, Tabuchi K, Tanaka S, Hodozuka A, PMID:22271522 Dev. 27(3):164-71. PMID:15737696 44. Alcedo J, Noll M (1997). Hedgehog and its
Kunishio K, Kubo N, et al. (2004 ). Capillary 16. Aerts I, Pacquement H, Doz F, Mosseri V, 30A. Aizer AA, Bi WL, Kandola MS, Lee EQ, patched-smoothened receptor complex: a novel
hemangioma of the central nervous system. J Desjardins L, Sastre X, et al. (2004). Outcome Nayak L, Rinne ML, et al. (2015). Extent of signalling mechanism at the cell surface. Biol
Neurosurg. 101 (1 ):73-81. PMID: 15255254 of second malignancies after retinoblastoma: a resection and overall survival for patients with Chem. 378(7):583-90. PMID:9278137
2. Abedalthagafi MS, Merrill PH, Bi WL, Jo- retrospective analysis of 25 patients treated at atypical and malignant meningioma. Cancer. 45. Alderson L: Fetell MR, Sisti M, Hochberg F,
nes RT, Listewnik ML, Ramkissoon SH, et the lnstitut Curie. Eur J Cancer. 40(10):1522-9. 121 (24):4376-81. PMID:26308667 Cohen M, Louis DN (1996). Sentinel lesions of
al. (2014). Angiomatous meningiomas have PMID:15196536 31. Akamatsu Y, Utsunomiya A, Suzuki S, Endo primary CNS lymphoma. J Neural Neurosurg
a distinct genetic profile with multiple chro- 17. Agaimy A (2014). Microscopic intraneural T, Suzuki I, Nishimura S, et al. (2010). Sub· Psychiatry. 60(1):102-5. PMID:8558135
mosomal polysomies including polysomy of perineurial cell proliferations in patients with ependymoma in the lateral ventricle manifes- 45A. Alentorn A, Dehais C, Ducray F, Carpen-
chromosome 5. Oncotarget. 5(21):10596-606. neurofibromatosis type 1. Ann Diagn Pathol. ting as intraventricular hemorrhage. Neural Med tier C, Mokhtari K, Figarella-Branger D, et al.
PMID:25347344 18(2):95-8. PMID:24461704 Chir (Tokyo). 50(11):1020-3. PMID:21123990 (2015). Allelic loss of 9p21.3 is a prognostic
3. Abel TW, Baker SJ, Fraser MM, Tihan T, 18. Agaimy A, Barthelrnefs S, Geddert H, Boltze 32. Aker FV, Ozkara S, Eren P, Peker 0, Ar- factor in 1p/19q codeleted anaplastic gliomas.
Nelson JS, Yachnis AT, et al. (2005). Lhermit- C, Moskalev EA, Koch M, et al. (2014). Pheno- rnaqan S, Hakan T (2005). Cerebellar lipon- Neurology. 85(15):1325-31. PMID:26385879
ts-Duclos disease: a report of 31 cases with typical and molecular distinctness of sinonasal eurocytoma/lipidized medulloblastoma. J Neu- 46. Alexander RT, Mclendon RE, Cummings
immunohistochemical analysis of the PTEN/ haemangiopericytoma compared to solitary rooncol. 71(1):53-9. PMID:15719276 T J (2004). Meningioma with eosinophilic granu-
AKT/mTOR pathway. J Neuropathol Exp Neu- fibrous tumour of the sinonasal tract. Histopa- 33. Akers AL, Johnson E, Steinberg GK, lar inclusions. Clin Neuropathol. 23(6):292-7.
rol. 64(4):341-9. PMID:15835270 thology. 65(5):667-73. PMID:24807787 Zabramski JM, Marchuk DA (2009). Biallelic PMID:15584214
4. Abrey LE, DeAngelis LM, Yahalom J (1998). 19. Agamanolis DP, Katsetos CD, Klonk CJ, somatic and germline mutations in cerebral ca· 47. Alexandrescu S, Brown RE, Tandon N
Long-term survival in primary CNS lymphoma. J Bartkowski HM, Ganapathy S, Staugaitis SM, et vernous malformations (CCMs): evidence for a Bhattacharjee MB (2012). Neuron precurso;
Clin Oncol. 16(3):859-63. PMID:9508166 al. (2012). An unusual form of superficially dis· two-hit mechanism of CCM pathogenesis. Hum features of spindle cell oncocytoma of adenohy-
5. Achatz Ml, Olivier M, Le Calvez F, Mar- seminated glioma in children: report of 3 cases. Mol Genet. 18(5):919-30. PMID:19088123 pophysis. Ann Clin Lab Sci. 42(2):123-9.
tel-Planche G, Lopes A, Rossi BM, et al. (2007). J Child Neural. 27(6):727-33. PMID:22596013 34. Akhaddar A, Zrara I, Gazzaz M, El Maust· PMI0:22585606
The TP53 mutation, R337H, is associated with 20. Agaram NP, Prakash S, Antonescu CR archid B, Benomar S, Boucetta M (2003). 48. Alexandrescu S, Korshunov A, Lai SH
l.i-Fraumeni and Li-Fraumeni-like syndromes (2005). Deep-seated plexiform schwannoma: a Cerebellar liponeurocytoma (lipomatous me- Oabiri S, Patil S, Li R, et al. (2015). Epithelioid
in Brazilian families. Cancer Lett. 245(1-2):96- pathologic study of 16 cases and comparative dulloblastoma). J Neuroradiol. 30(2):121-6. Glioblastomas and Anaplastic Epithelioid Ple-
102. PMID:16494995 analysis with the superficial variety. Am J Surg PMID:12717299 omorphic Xanthoastrocytomas-Same Entity or
6. Acker T, Plate KH (2004). Hypoxia and hy- Pathol. 29(8):1042-8. PMID:16006798 35. Akyurek S, Chang EL, Yu TK, Little D, First Cousins? Brain Pathol. PMID:26238627
poxia inducible factors (HIF) as important regu- 21. Agarwal S, Sharma MC, Sarkar C, Suri V, Allen PK, Mccutcheon I, et al. (2006). Spinal 49. Alexiou GA, Moschovi M, Georgoulis G,
lators of tumor physiology. Cancer Treat Res. Jain A, Sharma MS, et al. (2011 ). Extraventri· myxopapillary ependymoma outcomes in pati- Neroutsou R, Stefanaki K, Sfakianos G, et al.
117:219-48. PMID:15015563 cular neurocytomas: a morphological and his· ents treated with surgery and radiotherapy at (2010). Anaplastic oligodendrogliomas after
7. Actor B, Cobbers JM, BOschges R, Wolter togenetic consideration. A study of six cases. M.D. Anderson Cancer Center. J Neurooncol. treatment of acute lymphoblastic leukemia in
M, Knobbe CB, Lichter P, et al. (2002). Com· Pathology. 43(4):327-34. PMID:21532524 80(2):177-83. PMID:16648988 children: report of 2 cases. J Neurosurg Pediatr.
prehensive analysis of genomic alterations in 22. Agarwal S, Sharma MC, Singh G, Suri V, 36. Al-Hussaini M, Abuirmeileh N, Swaidan M, 5(2):179-83. PMI0:20121367
gliosarcoma and its two tissue components. Sarkar C, Garg A, et al. (2012). Papillary gli· AI-Jumaily U, Rajjal H, Musharbash A, et al. 50. Alexiou GA, Stefanaki K, Vartholomatos
Genes Chromosomes Cancer. 34(4):416-27. oneuronal tumor-a rare entity: report of four (2011). Embryonal tumor with abundant neuro- G, Sfakianos G, Prodromou N, Moschovl M
PMID:12112531 cases and brief review of literature. Childs Nerv pil and true rosettes: a report of three cases of (2013). Embryonal tumor with abundant neuro-
8. Adair JE, Johnston SK, Mrugala MM, Beard Sys!. 28(11 ):1897-904. PMID:22868530 a rare tumor, with an unusual case showing rh· pil and true rosettes: a systematic literature re-
BC, Guyman LA, Baldock AL, et al. (2014). 23. Aghi MK, Carter BS, Cosgrove GR, Oje· abdomyoblastic and melanocytic differentiation. view and report of 2 new cases. J Child Neural.
Gene therapy enhances chemotherapy toleran- mann RG, Amin·Hanjani S, Martuza RL, et al. Neuropathology. 31 (6):620-5. PMID:22103481 28(12):1709-15. PMI0:23334078
ce and efficacy in glioblastoma patients. J Clin (2009). Long-term recurrence rates of atypical 37. al-Sarraj ST, Parmar D, Dean AF, Phook- 51. Ali JB, Sepp T, Ward S, Green AJ, Yates JR
Invest. 124(9):4082-92. PMID:25105369 meningiomas after gross total resection with un G, Bridges LR (1998). Clinicopathological (1998). Mutations in the TSC1 gene account for
9. Adam C, Polivka M, Carpentier A, George or without postoperative adjuvant radiation. study of seven cases of spinal cord teratoma: a minority of patients with tuberous sclerosis. J
B, Gray F (2007). Papillary glioneuronal tumor: Neurosurgery. 64(1):56-60, discussion 60. a possible germ cell origin. Histopathology. Med Genet. 35(12):969-72. PMt0:9863590
not always a benign tumor? Clin Neuropathol. PMID:19145156 32(1):51-6. PMID:9522216 52. Alimova I, Birks DK, Harris PS, Knipstein
26(3):119-24. PMID:19157003 24. Agozzino L, Ferraraccio F, Accardo M, 38. Alameda F, Lloreta J, Galit6 E, Roquer J, JA, Venkataraman S, Marquez VE, et al.
10. Adamek D, Sofowora KD, Cwiklinska M, Esposito S, Agozzino M, Cuccurullo L (2006). Serrano S (1998). Meningeal melanocytoma: (2013). Inhibition of EZH2 suppresses self-re-
Herman-Sucharska I, Kwiatkowski S (2013). Morphological and ultrastructural findings of a case report and literature review. Ultrastruct newal and induces radiation sensitivity in atypi-
Embryonal tumor with abundant neuropil and prognostic impact in craniopharyngiomas. UI· Pathol. 22(4):349-56. PMID:9805360 cal rhabdoid teratoid tumor cells. Neuro Oneal.
true rosettes: an autopsy case-based update trastruct Pathol. 30(3):143-50. PMID:16825115 39. Albrecht S, Connelly JH, Bruner JM (1993). 15(2):149-60. PMI0:23190500
and review of the literature. Childs Nerv Syst. 25. Agrawal D, Singha! A, Hendson G, Durity Distribution of p53 protein expression in glios- 53. Alkadhi H, Keller M, Brandner S, Yonekawa
29(5):849-54. PMID:23358909 FA (2007). Gyriform differentiation in me- arcomas: an immunohistochemical study. Acta Y, Kollias SS (2001). Neuroimaging of cerebel-
11. Adams S, Teo C, McDonald KL, Zinger dulloblastoma - a radiological predictor of Neuropathol. 85(2):222-6. PMID:8382897 lar liponeurocytoma. Case report. J Neurosurg.
A, Bustamante S, Lim CK, et al. (2014). In· histology. Pediatr Neurosurg. 43(2):142-5. 40. Albrecht S, Goodman JC, Rajagopolan S, 95(2):324-31. PMID:11780904
volvement of the kynurenine pathway in PMID:17337929 Levy M, Cech DA, Cooley LD (1994). Malignant 54. Alkindy A, Chuzhanova N, Kini U, Cooper
human glioma pathophysiology. PLoS One. 26. Ahlsen G, Gillberg IC, Lindblom R, Gill- meningioma in Gorlin's syndrome: cytogenetic ON, Upadhyaya M (2012). Genotype-pheno-
9(11):e112945. PMID:25415278 berg C (1994). Tuberous sclerosis in Western and p53 gene analysis. Case report. J Neuro- type associations in neurofibromatosis type 1
12. Adams SA, Hilton DA (2002). Recurrent Sweden. A population study of cases with early surg. 81(3):466-71. PMID:8057157 (NF1): an increased risk of tumor complications
haemangioblastoma with glial differentiation. childhood onset. Arch Neurol. 51(1):76-81. 41. Albrecht S, Rouah E, Becker LE, Bruner J in patients with NF1 splice-site mutations? Hum
Neuropathol Appl Neurobiol. 28(2):142-6. PMID:8274113 (1991). Transthyretin immunoreactivity in cha· Genomics. 6:12. PMI0:23244495
PMID:11972801 27. Ahmadi R, Stockhammer F, Becker N, Hoh- raid plexus neoplasms and brain metastases. 55. Allen JC, Judkins AR, Rosenblum MK,
13. Adamson TE, Wiestler OD, Kleihues P, len K, Misch M, Christians A, et al. (2012). No Mod Pathol. 4(5):610-4. PMID:1758873 Biegel JA (2006). Atypical teratoid/rhabdoid
Ya§argil MG (1990). Correlation of clinical and prognostic value of IDH1 mutations in a series 42. Albrecht S, von Deimling A, Pietsch T, Gi· tumor evolving from an optic pathway ganglio-
pathological features in surgically treated era· of 100 WHO grade II astrocytomas. J Neuroon- angaspero F, Brandner S, Kleihues P, et al. glioma: case study. Neuro Oneal. 8(1):79-82.
niopharyngiomas. J Neurosurg. 73(1):12-7. col. 109(1):15-22. PMID:22528790 (1994). Microsatellite analysis of loss of hetero· PMID:16443951
PMID:2352012 28. Ahmed KA.Allen PK, Mahajan A, Brown PD, zygosity on chromosomes 9q, 11p and 17p in 56. Alles JU, Bossie! K, Schachenmayr W
14. Adeleye AO, Okolo CA, Akang EE, Adesina Ghia AJ (2014). Astroblastomas: a Surveillance, medulloblastomas. Neuropathol Appl Neuro- (1986). Hemangioblastoma of the cerebellUm-
AM (2012). Cerebral pleomorphic xanthoast- Epidemiology, and End Results (SEER)·based biol. 20(1):74-81. PMID:8208343 an immunocytochemical study. Clin Neuropa-
rocytoma associated with NF1: an updated patterns of care analysis. World Neurosurg. 43. Albright AL, Wisoff JH, Zeltzer P, Boyett J, thol. 5(6):238-41. PMID:3815934
review with a rare atypical case from Africa. 82(1·2):e291-7. PMID:24141003 Rorke LB, Stanley P, et al. (1995). Prognostic 57. Alleyne CH Jr, Hunter S, Olson JJ, Barrow
Neurosurg Rev. 35(3):313-9, discussion 319. 29. Ahuja A, Sharma MC, Suri V, Sarkar C, factors in children with supratentorial (non· OL (1998). Lipomatous glioneurocytoma of
PMID:22020543 Sharma BS, Garg A (2011). Pineal anlage tu- pineal) primitive neuroectoderrnal tumors. A the posterior fossa with divergent differentiati-
15. Adriani KS, Stenvers DJ, lmanse JG (2012). mour - a rare entity with divergent histology. J neurosurgical perspective from the Children's on: case report. Neurosurgery. 42(3):639-43.
Pearls & oy-sters: Lumbar paraganglioma: can Clin Neurosci. 18(6):811-3. PMID:21435885 Cancer Group. Pediatr Neurosurg. 22(1):1-7. PMID:9526999
you see it in the eyes? Neurology. 78(4):e27-8. 30. Aicardi J (2005). Aicardi syndrome. Brain PMID:7888387 58. Allinson KS, O'Oonovan OG, Jena R, Cross

356 References
JJ, Santarius TS (2015). Rosette-forming glio- Tatagiba M, Thomas S, Brandis A, et al. (1997). Leptomeningeal dissemination of malignant MGMT methylation analysis of glioblasloma
neuronal tumor with dissemination throughout Proliferation potential and histological features gliomas. Incidence, diagnosis and outco- on the lnfinium methylalion BeadChip identifies
the ventricular system: a case report. Clin Neu- in neurofibromatosis 2-associated and spora- me. Acta Neurochir (Wien). 126(2-4):84-92. two distinct CpG regions associated with gene
ropathol. 34(2):64-9. PMID:25373141 dic meningiomas. J Neurosurg. 87(4):610-4. PMID:8042560 silencing and outcome, yielding a prediction
59. Almefty R, Webber BL, Arnautovic Kl (2006). PMID:9322850 91. Arivazhagan A, Anandh B, Sanlosh V, model for comparisons across datasets, tumor
lntraneural perineurioma of the third cranial ner- 75. Antinheimo J, Haapasalo H, Seppala M, Chandramouli BA (2008). Pineal parenchy- grades, and GIMP-status. Acta Neuropathol.
ve: occurrence and identification. Case report. J Sainio M, Carpen 0, Jaaskelainen J (1995). mal tumors-utility of immunohistochemical 124(4):547-60. PMID:22810491
Neurosurg 104(5):824-7. PMID:16703891 Proliferative potential of sporadic and neuro- markers in prognostication. Clin Neuropathol. 106. Baehring JM, Dickey PS, Bannykh SI
60. Alomari AK, Kelley BJ, Damisah E, Marks fibromatosis 2-associated schwannomas as 27(5):325--33. PMID:18808064 (2004 ). Epithelioid hemangioendolhelioma
A, Hui P, Diluna M, et al. (2015). Cranio- studied by MIB-1 (Ki-67) and PCNA labeling. 92. Arnold MA, Stallings-Archer K, Marlin E, of the suprasellar area: a case report and re-
pharyngioma arising in a Rathke's cleft cyst: J Neuropathol Exp Neurol. 54(6):776-82. Grondin R, Olshefski R, Biegel JA, et al. (2013). view of the literature. Arch Pathol Lab Med.
case report. J Neurosurg Pediatr. 15(3):250-4. PMID: 7595650 Cribriform neuroepithelial tumor arising in the 128(11):1289-93. PMID:15504067
PMID:25555112 76. Antinheimo J, Sankila R, Carpen 0, Puk- lateral ventricle. Pediatr Dev Pathol. 16(4):301- 107. Bailey P, Bucy PC (1929). Oligodendroglio-
61. Alpers CE, Davis RL, Wilson CB (1982). kala E, Sainio M, Jaaskelainen J (2000). Po- 7. PMID:23495723 mas of the brain. J Path Bae!. 32:735-51.
Persistence and late malignant transforma- pulation-based analysis of sporadic and type 93. Aronica E, Boer K, Becker A, Redeker S, 108. Bailey P, Cushing H (1926). A classification
tion of childhood cerebellar astrocytoma. 2 neurofibromatosis-associated meningiomas Spliet WG, van Rijen PC, et al. (2008). Gene of tumors of the glioma group on a histogenelic
Case report. J Neurosurg. 57(4):548-51. and schwannomas. Neurology. 54(1):71-6. expression profile analysis of epilepsy-associa- basis with a correlation study of prognosis. Phi-
PMID:7108605 PMID:10636128 ted gangliogliomas. Neuroscience. 151(1):272- ladelphia: Lippincott.
62. Alanoz MA, Santaguida C, Guiel MC, Del 77. Anton T, Guttierez J, Rock J (2006). Ten- 92. PMID:180937 40 109. Bailey P, Cushing H (1926). Classification
Maestro RF (2005). Spinal hemangioblastoma torial schwannoma: a case report and review 94. Aronica E, Leenstra S, van Veelen CW, of the Tumors of the Glioma Group. Philadelph-
containing metastatic renal cell carcinoma in of the literature. J Neurooncol. 76(3):307-11. van Rijen PC, Hulsebos T J, Tersmette AC, et ia: Lippincott.
von Hippel-Lindau disease. Case report and PMID:16200344 al. (2001 ). Glioneuronal tumors and medically 110. Bainbridge MN, Armstrong GN, Gramal-
review of the literature. J Neurosurg Spine. 78. Antonelli M, Korshunov A, Mastronuzzi A, intractable epilepsy: a clinical study with long- ges MM, Bertuch AA, Jhangiani SN, Dodda-
3(6):495--500. PMID:16381215 Diomedi Camassei F. Carai A, Colafati GS, term follow-up of seizure outcome after surgery. paneni H, et al.; Gliogene Consortium (2015).
63. Altundag MK, Ozisik Y, Valcin S, Akyol F, et al. (2015). Long-term survival in a case of Epilepsy Res. 43(3):179-91. PMID 11248530 Germline mutations in shelterin complex genes
Uner A (2000). Primary low grade B-cell lym- ETANTR with histological features of neuron- 94A. Arvela E, Soderstrom M, Korhonen M, are associated with familial glioma. J Natl Can-
phoma of the dura in an immunocompetent al maturation after therapy. Virchows Arch. Halmesrnaki K, Alback A, Lepantalo M, et al. cer Inst. 107(1):384. PMID:25482530
patient. J Exp Clin Cancer Res. 19(2):249-51. 466(5):603-7. PMID:25697539 (2010). Finnvasc score and modified Prevent 111. Baisden BL, Brat DJ, Melhem ER, Rosen-
PMID:10965827 79. Antonescu CR (2006). The role of genetic Ill score predict long-term outcome after inf- blum MK, King AP, Burger PC (2001 ). Dysem-
64. Amano K, Kubo 0, Komori T, Tanaka M, Ka- testing in soft tissue sarcoma. Histopathology. rainguinal surgical and endovascular revas- bryoplastic neuroepithelial tumor-like neoplasm
wamata T, Hori T, et al. (2013). Clinicopatholo- 48(1):13-21. PMID:16359533 cularization for critical limb ischemia.J Vase of the septum pellucidum: a lesion often misdi-
gical features of sellar region xanthogranuloma: 80. Antonescu CR, Le Loarer F, Mosquera JM, Surg52(5):1218-25. PMID:20709482 agnosed as glioma: report of 10 cases. Am J
correlation with Rathke's cleft cyst. Brain Tumor Sboner A, Zhang L, Chen CL, et al. (2013). 95. Asha U, Mahadevan A, Sathiyabama D, Ra- Surg Pathol. 25(4):494-9. PMID:11257624
Pathol. 30(4):233-41. PMID:23322180 Novel YAP1-TFE3 fusion defines a distinct vindra T, Sagar BK, Bhat DI, et al. (2015). Lack 112. Baker KB, Moran CJ, Wippold FJ 2nd,
65. Amemiya S, Shibahara J, Aoki S, Takao H, subset of epithelioid hemangioendothelioma. of IDH1 mutation in astroblastomas suggests Smimiotopoulos JG, Rodriguez FJ, Meyers SP,
Ohtomo K (2008). Recently established entities Genes Chromosomes Cancer. 52(8):775-84. putative origin from ependymoglial cells? Neu- et al. (2000). MR imaging of spinal hemangiob-
of central nervous system tumors: review of PMID:23737213 ropathology. 35(4):303-11. PMID:25786545 lasloma. AJR Am J Roentgenol. 174(2):377-82.
radiological findings. J Comput Assist Tomogr. 81. Antonescu CR, Suurmeijer AJ, Zhang L, 96. Aslrinidis A, Henske EP (2005). Tuberous PMID:10658709
32(2):279-85. PMID:18379318 Sung YS, Jungbluth AA, Travis WO, et al. sclerosis complex: linking growth and energy 113. Bakry D, Aronson M, Durno C, Rimawi
66. Amirian ES, Goodman JC, New P, Scheurer (2015). Molecular characterization of inflam- signaling pathways with human disease. Onco- H, Farah R, Alharbi OK, et al. (2014). Genetic
ME (2014). Pediatric and adult malignant peri- matory myofibroblastic tumors with frequent gene. 24(50):7475-81. PMID 16288294 and clinical determinants of constitutional mis-
pheral nerve sheath tumors: an analysis of data ALK and ROS 1 gene fusions and rare no- 96A. Athar M, Li C, Kim AL, Spiegelman VS, match repair deficiency syndrome: report from
from the surveillance, epidemiology, and end vel RET rearrangement. Am J Surg Pathol. Bickers DR (2014). Sonic hedgehog signaling the constitutional mismatch repair deficien-
results program. J Neurooncol. 116(3):609-16. 39(7):957-67. PMID:25723109 in basal cell nevus syndrome. Cancer Res. cy consortium. Eur J Cancer. 50(5):987-96.
PMID:24390465 82. Antonescu CR, Woodruff JM (2006). Pri- 74(18):4967-75. PMID:25172843 PMID:24440087
67. Amlashi SF, Riffaud L, Brassier G, Morandi mary Tumors and Cranial, Spinal and Periphe- 97. Au KS, Williams AT, Roach ES, Batchelor 114. Bakshi R, Shaikh ZA, Kamran S, Kinkel PR
X (2003). Nevoid basal cell carcinoma syndro- ral Nerves. In: Mclendon RE, Rosenblum MK, L, Sparagana SP, Delgado MR, et al. (2007). (1999). MRI findings in 32 consecutive lipomas
me: relation with desmoplastic medulloblasto- Bigner DD, editors. Russel and Rubinstein's Pa- Genotype/phenotype correlation in 325 indi- using conventional and advanced sequences. J
ma in infancy. A population-based study and thology of the Nervous System. Hoder Arnold; viduals referred for a diagnosis of tuberous Neuroimaging. 9(3):134-40. PMID:10436754
review of the literature. Cancer. 98(3):618-24. pp. 787-835. sclerosis complex in the United Slates. Genet 115. Baldini F, Baiocchini A, Schinina V, Agra-
PMID:12879481 83. Aoki Y, Niihori T, Narumi Y, Kure S, Mat- Med. 9(2):88-100. PMID:17304050 Ii C, Giancola ML, Alba L, et al. (2013). Brain
68. Anan M, Inoue R, Ishii K, Abe T, Fujiki M, subara Y (2008). The RAS/MAPK syndromes: 98. Awaya H, Kaneko M, Amatya VJ, Takes- localization of Kaposi's sarcoma in a patient tre-
Kobayashi H, et al. (2009). A rosette-forming novel roles of the RAS pathway in human ge- hima Y, Oka S, lnai K (2003). Myxopapillary ated by combination antiretroviral therapy. BMC
glioneuronal tumor of the spinal cord: the first netic disorders. Hum Muta!. 29(8):992-1006. ependymoma with anaplastic features. Pathol Infect Dis. 13:600. PMID:24359263
case of a rosette-forming glioneuronal tumor PMID:18470943 Int. 53(10):700-3. PMID:14516321 116. Balik V, Srovnal J, Sulla I, Kalila 0, Folla-
originating from the spinal cord. Hum Pathol. 84.Aoyama I, Kondo A, Ogawa H, lkai Y (1994). 99. Aydin F, Ghatak NR, Salvant J, Muizelaar nova T, Vaverka M, et al. (2013). MEG3: a novel
40(6):898-901. PMID: 19269010 Germinoma in siblings: case reports. Surg Neu- P (1993). Desmoplastic cerebral astrocytoma of long noncoding potentially tumour-suppressing
69. Anan M, Ishii K, Nakamura T, Yamashita M, ral. 41(4):313-7. PMID:8165502 infancy. A case report with immunohistochemi- RNA in meningiomas. J Neurooncol. 112(1):1-
Katayama S, Sainoo M, et al. (2006). Postope- 85. Appin CL, Gao J, Chisolm C, Torian M, Alexis cal, ultrastructural and proliferation studies. Acta 8. PMID:23307326
rative adjuvant treatment for pineal parenchy- D, Vincentelli C, et al. (2013). Glioblastoma with Neuropathol. 86(6):666-70. PMID:7906073 117. Balmaceda CM, Fe tell MR, O'Brien JL,
mal tumour of intermediate differentiation. J Clin oligodendroglioma component (GBM-0): mole- 100. Aziz M, Chaurasia JK, Khan R, Afroz N Housepian EH (1993). Nevus of Ota and lep-
Neurosci. 13(9):965--8. PMID:16904896 cular genetic and clinical characteristics. Brain (2014). Primary low-grade diffuse small lympho- lomeningeal melanocytic lesions. Neurology.
70. Andoniadou CL, Gaston-Massuet C, Reddy Pathol. 23(4):454-61. PMID:23289977 cylic lymphoma of the central nervous system. 43(2):381-6. PMID:8437707
R, Schneider RP, Blasco MA, Le Tissier P, et al. 86. Aquilina K, Kamel M, Kalimuthu SG, Marks BMJ Case Rep. 2014:2014. PMID:24729110 118. Balss J, Meyer J, Mueller W, Korshunov
(2012). Identification of novel pathways invol- JC, Keohane C (2006). Granular cell tumour of 101. Azzarelli B, Rekate HL, Roessmann A, Hartmann C, van Deimling A (2008). Ana-
ved in the pathogenesis of human adamantino- the neurohypophysis: a rare sellar tumour with U (1977). Subependymoma: a case report lysis of the IDH1 codon 132 mutation in brain
matous craniopharyngioma. Acta Neuropathol. specific radiological and operative features. Br J with ultrastructural study. Acta Neuropalhol. tumors. Acta Neuropathol. 116(6):597-602.
124(2):259-71. PMID:22349813 Neurosurg. 20(1):51-4. PMID:16698612 40(3):279-82. PMID:203160 PMID:18985363
71. Andreasson A, Kiss NB, Caramuta S, Su- 87. Aquilina K, Nanra JS, Allcutt DA, Farrell M 102. Bacci C, Sestini R, Provenzano A, Pagani- 118A. Bandopadhayay P, Ramkissoon LA, Jain
laiman L, Svahn F, Backdahl M, et al. (2013). (2005). Choroid plexus adenoma: case report ni I, Mancini I, Porfirio B, et al. (2010). Schwan- P, Berglhold G, Wala J, Zeid R, et al. (2016).
The VHL gene is epigenetically inactivated and review of the literature. Childs Nerv Sys!. nomatosis associated with multiple meningi- MYB-QKI rearrangements in angiocenlric glioma
in pheochromocytomas and abdominal pa- 21(5):410-5. PMID:15565450 omas due to a familial SMAR CB 1 mutation. drive tumorigenicity through a tripartite mecha-
ragangliomas. Epigenetics. 8(12):1347-54. 88. Arai H, lkota H, Sugawara K, Nobusawa Neurogenetics. 11(1 ):73-80. PMID:19582488 nism. Nat Genet. 48(3) 273-82. PMID:26829751
PMID:24149047 S, Hirata J, Nakazato Y (2012). Nestin expres- 103. Badalian-Very G, Vergilto JA, Degar BA, 119. Banerjee AK, Sharma BS, Kak VK, Ghalak
72. Andreiuolo F, Puget S, Peyre M, Dan- sion in brain tumors: its utility for pathological MacConaill LE, Brandner B, Calicchio ML, el al. NR (1989). Gliosarcoma with cartilage formati-
tas-Barbosa C, Boddaert N, Philippe C, et al. diagnosis and correlation with the prognosis (2010). Recurrent BRAF mutations in Langer- on. Cancer. 63(3):518-23. PMID:2643455
(2010). Neuronal differentiation distinguishes of high-grade gliomas. Brain Tumor Pathol. hans cell histiocytosis. Blood. 116(11):1919-23. 120. Bannykh S, Strugar J, Baehring J (2005).
supratentorial and infratentorial childhood 29(3):160-7. PMID:22350668 PMID:20519626 Paraganglioma of the lumbar spinal canal. J
ependymomas. Neuro Oneel. 12(11):1126-34. 89. Arita H, Narita Y, Fukushima S, Tateishi K, 104. Badiali M, Gleize V, Paris S, Moi L, Elhoua- Neurooncol. 75(2):119. PMID:16283440
PMID:20615923 Matsushita Y, Yoshida A, et al. (2013). Upregu- dani S, Arcelia A, et al. (2012). KIAA 1549-BRAF 121. Bannykh SI, Stolt CC, Kim J, Perry A,
73. Anghileri E, Eoli M, Paterra R, Ferroti P, Pol- lating mutations in the TERT promoter com- fusions and IDH mutations can coexist in diffu- Wegner M (2006). Oligodendroglial-specific
lo B, Cuccarini V, et al. (2012). FABP4 is a can- monly occur in adult malignant gliomas and are se gliomas of adults. Brain Pathol. 22(6):841-7. transcriptional factor SOX10 is ubiquitously
didate marker of cerebellar liponeurocytomas. strongly associated with total 1p19q loss. Acta PMID:22591444 expressed in human gliomas. J Neurooncol.
J Neurooncol. 108(3):513-9. PMID:22476608 Neuropalhol. 126(2):267-76. PMID:23764841 105. Bady P, Sciuscio D, Diserens AC, Bloch 76(2): 115--27. PMID:16205963
74. Antinheimo J, Haapasalo H, Haltia M, 90. Arita N, Taneda M, Hayakawa T (1994). J, van den Bent MJ, Marosi C, el al. (2012). 122. Bao S, Wu Q, Mclendon RE, Hao Y, Shi Q,

References 357
Hjelmeland AB, et al. (2006). Glioma stem cells 139. Bastian BC (2014). The molecular pa- Brain Pathol. 20(2):441-50. PMID:19725831 Invasion patterns in brain metastases of so-
promote radioresistance by preferential acti- thology of melanoma: an integrated taxonomy 158. Beier D, Hau P, Proescholdt M, Lohmei- lid cancers. Neuro Oneal. 15(12):1664-72
vation of the DNA damage response. Nature. of melanocytic neoplasia. Annu Rev Pathol. er A, Wischhusen J, Oefner PJ, et al. (2007). PMID:24084410 .
444(7120):756-60. PMID 17051156 9:239-71. PMID:24460190 CD133(+) and CD133(-) glioblastoma-derived 173. Berho M, Suster S (1994). Mucinous-
123. Bar EE, Lin A, Tihan T. Burger PC, Eber- 140. Batchelor T, Loeffler JS (2006). Primary cancer stem cells show differential growth cha- meningioma. Report of an unusual variant '01
hart CG (2008). Frequent gains at chromosome CNS lymphoma. J Clin Oneal. 24(8):1281-8. racteristics and molecular profiles. Cancer Res. meningioma that may mimic metastatic mu--
7q34 involving BRAF in pilocytic astrocytoma. PMID:16525183 67(9):4010-5. PMID:17483311 cin-producing carcinoma. Am J Surg Pathel
J Neuropathol Exp Neural. 67(9):878-87. 141. Batchelor Tl, Reardon DA, de Groot JF, 159. Bell DA, Woodruff JM, Scully RE (1984). 18(1):100-6. PMID:8279622 ..
PMID:18716556 Wick W, Weller M (2014). Antiangiogenic the- Ependymoma of the broad ligament. A report 174. Be_ristain X, Azzarelli B (2002). Ttce-
124. Baraniskin A, Kuhnhenn J, Schlegel U, rapy for glioblastoma: current status and future of two cases. Am J Surg Pathol. 8(3):203-9. neuroloqical masquerade of inlravascula(
Chan A, Deckert M, Gold R, et al. (2011 ). Identi- prospects. Clin Cancer Res. 20(22):5612-9. PMID:6703196 lymphomatosis. Arch Neural. 59(3):439-43.
fication of microRNAs in the cerebrospinal fluid PMID:25398844 159A. Bell RJ, Rube HT, Kreig A, Mancini A, PMID:11890850
as marker for primary diffuse large B-cell lym- 142. Batara NV, Sturm D, Jones DT, Kool M, Fouse SD, Nagarajan RP, et al. (2015). Can- 175. Berkman RA, Clark WC, Saxena A, Ro-
phoma of the central nervous system. Blood. Pfister SM, Northcott PA (2014). Transitioning cer. The transcription factor GABP selectively bertson JT, Oldfield EH, Ali IU (1992). Clonal
117(11):3140-6. PMID:21200023 from genotypes lo epigenotypes: why the time binds and activates the mutant TERT promo- composition of glioblastoma multiforme. J "leu:
125. Baraniskin A, Kuhnhenn J, Schlegel U, has come for medulloblastoma epigenomics. ter in cancer. Science. 348(6238):1036-9. rosurg. 77(3):432-7. PMID:1324297
Schmiegel W, Hahn S, Schroers R (2012). Neuroscience. 264:171-85. PMID:23876321 PMID:25977370 176. Bernell WR, Kepes JJ, Seitz EP (1972).
MicroRNAs in cerebrospinal fluid as biomarker 143. Balzdorf U, Malamud N (1963). The pro- 160. Bellail AC, Hunter SB, Brat DJ, Tan C, Van Late malignant recurrence of childhood cers,
for disease course monitoring in primary cen- blem of multicentric gliomas. J Neurosurg. Meir EG (2004). Microregional extracellular ma- bellar astrocytoma. Report of two cases .. J Neu-
tral nervous system lymphoma. J Neurooncol. 20:122-36. PMID:14192080 trix heterogeneity in brain modulates glioma cell rosurg. 37(4):470-4. PMID:5070873
109(2):239-44. PMID:22729947 144. Bauman GS, Wara WM, Ciricillo SF, Da- invasion. Int J Biochem Cell Biol. 36(6):1046- 177. Berns S, Pearl G (2006). Review of pineal
126. Barker FG 2nd, Davis RL, Chang SM, vis RL, Zager S, Edwards MS (1997). Primary 69. PMID:15094120 anlage tumor with divergent histology. Arch Pa- _
Prados MD (1996). Necrosis as a prognostic intracerebral osteosarcoma: a case report. J 161. Bello MJ, Leone PE, Vaquero J, de Cam- thol Lab Med. 130(8):1233-5. PMID 16879032°
factor in glioblastoma multiforme. Cancer. Neurooncol. 32(3):209-13. PMID:9049882 pos JM, Kusak ME, Sarasa JL, et al. (1995). 178. Bernthal NM, Jones KB, Monument MJ
77(6):1161-6. PMID:8635139 145. Baumert BG, Rutten I, Dehing-Oberije C, Allelic loss at 1p and 19q frequently occurs in Liu T, Viskochil D, Randall RL (2013). LosLi�
127. Barkovich AJ, Guerrini R, Kuzniecky RI, Twijnstra A, Dirx MJ, Debougnoux-Huppertz association and may represent early oncogenic translation: ambiguity in nerve sheath tumor
Jackson GD, Dobyns WB (2012). A develop- RM, el al. (2006). A pathology-based substra- events in oligodendroglial tumors. Int J Cancer. nomenclature and its resultant treatment.effect.
mental and genetic classification for malfor- te for target definition in radiosurgery of brain 64(3):207-10. PMID:7622310 Cancers (Basel). 5(2):519-28. PMID:24216989.,
mations of cortical development: update 2012. metastases. Int J Radial Oneal Biol Phys. 162. Bello MJ, Rey JA, de Campos JM, Ku- 179. Bernthal NM, Putnam A, Jones KB, Vi�-
Brain. 135(Pt 5):1348-69. PMID:22427329 66(1):187-94. PMID:16814946 sak ME (1993). Chromosomal abnormalities kochil D, Randall RL (2014) The effect of
128. Barnard RO, Geddes JF (1987). The inci- 146. Baumgarten P, Harter PN, Tiinjes M, Cap- in a pineocytoma. Cancer Genet Cytogenel. surgical margins on outcomes for low grade
dence of multifocal cerebral gliomas. A histolo- per D, Blank AE, Sahm F, el al. (2014). Loss of 71(2):185-6. PMID:8281527 MPNSTs and atypical neurofibroma. J Surg On-
gic study of large hemisphere sections. Cancer. FUBP1 expression in gliomas predicts FUBP1 163. Belloni E, Muenke M, Roessler E, Traver- eal. 110(7):813-6. PMID:25111615
60(7):1519-31. PMID:3113716 mutation and is associated with oligodendroglial so G, Siegel-Bartelt J, Frumkin A, et al. (1996). 180. Beschorner R, Pantazis G, Jeibmann
129. Barnard ZR, Agarwalla PK, Jeyaretna differentiation, IDH1 mutation and 1p/19q loss Identification of Sonic hedgehog as a candidate A, Boy J, Meyermann R, Mittelbronn M. et al.. ..
DS, Farrell CJ, Gerstner ER, Tian D, et al. of heterozygosity. Neuropathol Appl Neurobiol. gene responsible for holoprosencephaly. Nat (2009). Expression of EAAT-1 distinguishes
(2011 ). Sporadic primary malignant intrace- 40(2):205-16. PMID:24117 486 Genet. 14(3):353-6. PMID:8896571 choroid plexus tumors from normal and reactive
rebral nerve sheath tumors: case report and 147. Baysal BE (2002). Hereditary paragangli- 164. Benesch M, Frappaz D, Massimino M choroid plexus epithelium. Acta Neuropathol.
literature review. J Neurooncol. 104(2):605-10. oma targets diverse paraganglia. J Med Genet. (2012). Spinal cord ependymomas in chil- 117(6):667-75. PMID:19283393 .
PMID:21327709 39(9):617-22. PMID:12205103 dren and adolescents. Childs Nerv Sysl. 181. Bethke L, Murray A, Webb E, Schoe
130. Barnholtz-Sloan JS, Sloan AE, Davis FG, 148. Beaumont TL, Kupsky WJ, Barger GR, 28(12):2017-28. PMID:22961356 M, Muir K, McKinney P, et al. (2008). Co
Vigneau FD, Lai P, Sawaya RE (2004). Inciden- Sloan AE (2007). Gliosarcoma with multiple ex- 165. Beni-Adani L, Gomori M, Spektor S, hensive analysis of DNA repair gene variants
ce proportions of brain metastases in patients tracranial metastases: case report and review Constantini S (2000). Cyst wall enhancement and risk of meningioma. J Natl Cancer lnsr·
diagnosed (1973 to 2001) in the Metropolitan of the literature. J Neurooncol. 83(1):39-46. in pilocytic astrocytoma: neoplastic or reactive 100(4)270-6. PMID:18270339
Detroit Cancer Surveillance System. J Clin On- PMID:17171442 phenomena. Pediatr Neurosurg. 32(5):234-9. 182. Bettegowda C, Adogwa 0, Mehta V,
eal. 22(14):2865-72. PMID:15254054 149. Becher MW, Abel TW, Thompson RC, PMID:10965269 Chaichana KL, Weingart J, Carson BS, et ,iL_
131. Barrande G, Kujas M, Gancel A, Turpin Weaver KD, Davis LE (2006). lmmunohisto- 166. Benites Filho PR, Sakamoto D, Machuca (2012). Treatment of choroid plexus tumors: a
G, Brucker! E, Kuhn JM, et al. (1995). [Gra- chemical analysis of metastatic neoplasms of TN, Serapiao MJ, Ditzel L, Bleggi Torres LF 20-year single institutional experience. J Neu-
nular cell tumors. Rare tumors of lhe neu- the central nervous system. J Neuropathol Exp (2005). Granular cell tumor of the neurohy- rosurg Pediatr. 10(5):398-405. PMID:22938081 ..
rohypophysis]. Presse Med. 24(30):1376-80. Neural. 65(10):935-44. PMID:17021398 pophysis: report of a case with unusual age 183. Bettegowda C, Agrawal N, Jiao Y, Sausen·-
PMID:8545314 150. Bechel D, Gielen GG, Korshunov A, Pfister presentation. Virchows Arch. 447(3):649-52. M, Wood LD, Hruban RH, et al. (2011). Muta-
132. Barresi V, Cerasoli S, Morigi F, Cremonini SM, Rousso C, Faury D, et al. (2014). Specific PMID:16133355 tions in CIC and FUBP1 contribute to human au:
AM, Volpini M, Tuccari G (2006). Gliosarcoma detection of methionine 27 mutation in histo- 167. Bennett JP Jr, Rubinstein LJ (1984). The godendroglioma. Science. 333(6048):145:}-5;-
with features of osteoblastic osteosarcoma: a ne 3 variants (H3K27M) in fixed tissue from biological behavior of primary cerebral neurob- PMID:21817013 -
review. Arch Pathol Lab Med. 130(8):1208-11. high-grade astrocytomas. Acta Neuropalhol. lastoma: a reappraisal of the clinical course in 184. Bettegowda C, Agrawal N, Jiao Y, Wan!! ...
PMID:16879025 128(5):733-41. PMID:25200321 a series of 70 cases. Ann Neural. 16(1):21-7. Y, Wood LD, Rodriguez FJ, et al. (2013). Ex<r
133. Barresi V, Vitarelli E, Branca G, Antonelli 151. Beck DJK, Russell DS (1942). Oliqo- PMID:6431897 mic sequencing of four rare central nervous, ..
M, Giangaspero F, Barresi G (2012). Expressi- dendrogliomatosis of the cerebrospinal pa- 168. Bennett KL, Mester J, Eng C (2010). system tumor types. Oncotarget. 4(4):572-83,_ -
on of brachyury in hemangioblastoma: potential thway. Brain. 352-72. Germline epigenetic regulation of KILLIN in PMID:23592488
use in differential diagnosis. Am J Surg Pathol. 152. Becker DH, Wilson CB (1981). Sympto- Cowden and Cowden-like syndrome. JAMA. 185. Betz BL, Strobeck MW, Reisman DN,
36(7):1052-7. PMID:22446946 matic parasellar granular cell tumors. Neurosur- 304(24):2724-31. PMID:21177507 Knudsen ES, Weissman BE (2002). Re-expres>
134. Barrett AW, Hopper C, Landon G (2002). gery. 8(2):173-80. PMID:6259552 169. Benzagmout M, Karachi C, Mokhtari K, sion of hSNF5/INl1/BAF47 in pediatric tumor ..
Intra-osseous soft tissue perineurioma of the in- 153. Becker I, Paulus W, Roggendorf W Capelle L (2013). Hemorrhagic papillary glio- cells leads to G1 arrest associated wilh inducti-
ferior alveolar nerve. Oral Oneal. 38(8):793-6. (1989). Histogenesis of stromal cells in cere- neuronal tumor mimicking cavernoma: two case on of p16ink4a and activation of RB. Oncogene'.:,.
PMID:12570059 bellar hemangioblastomas. An immunohisto- reports. Clin Neural Neurosurg. 115(2):200-3. 21(34):5193-203. PMID:12149641 .
135. Bartels U, Hawkins C, Vezina G, Kun L, chemical study. Am J Pathol. 134(2):271-5. PMID:22717600 186. Bhagavathi S, Greiner TC, Kazmi SA, Fu:-.. ..
Souweidane M, Bouffet E (2011). Proceedings PMID:2916647 170. Berghoff AS, Bartsch R, Wohrer A, Streu- K, Sanger WG, Chan WC (2008). Extranodal - se
of the diffuse intrinsic pontine glioma (DIPG) 154. Becker RL, Becker AD, Sobel DF (1995). bel B, Birner P, Kras JM, el al. (2014). Predictive marginal zone lymphoma of the dura mater with
Toronto Think Tank: advancing basic and trans- Adult medulloblastoma: review of 13 cases with molecular markers in metastases to the central lgH/MALT1 lranslocation and review of litera- ·_
lational research and cooperation in DIPG. J emphasis on MRI. Neuroradiology. 37(2):104- nervous system: recent advances and future lure. J Hematop. 1(2):131-7. PMID:19669212 t
Neurooncol. 105(1):119-25. PMID:21901544 8. PMID:7760992 avenues. Acta Neuropathol. 128(6):879-91. 187. Bhargava D, Sinha P, Chumas P, Al-Tami-_.'· ..
136. Barzilay JI, Pazianos AG (1989). Ad- 155. Beert E, Brems H, Daniels B, De We- PMID:25287912 mi Y, Shivane A, Chakrabarty A, et al. (2013):�·
renocortical carcinoma. Ural Clin North Am. ver I, Van Calenbergh F, Schoenaers J, el al. 171. Berghoff AS, llhan-Mutlu A, Dinhof C, Occurrence and distribution of pilomyxoid·�-
16(3):457-68. PMID:2665272 (2011 ). Atypical neurofibromas in neurofibro- Magerle M, Hackl M, Widhalm G, et al. (2015). astrocytoma. Br J Neurosurg. 27(4):413-8:··,
137. Baser ME, Friedman JM, Joe H, Shen- matosis type 1 are premalignant tumors. Ge- Differential role of angiogenesis and tumour PMID:23281683 •
ton A, Wallace AJ, Ramsden RT, et al. (2011). nes Chromosomes Cancer. 50(12):1021-32. cell proliferation in brain metastases according 188. Bhattacharjee MB, Anmstrong DD, Vogel
Empirical development of improved diagnostic PMID:21987 445 to primary tumour type: analysis of 639 cases. H, Cooley LD (1997). Cytogenetic analysis oh
criteria for neurofibromatosis 2. Genet Med. 156. Beert E, Brems H, Renard M, Ferreira Neuropathol Appl Neurobiol. 41(2):e41-55. 120 primary pediatric brain tumors and literature �
13(6):576-81. PMID:21451418 JF, Melotte C, Thoelen R, et al. (2012). Bialle- PMID:25256708 review. Cancer Genet Cytogenet. 97(1):39-53.}
138. Baser ME, Kuramoto L, Joe H, Friedman lic inactivation of NF1 in a sporadic plexiform 171A. Berghoff AS, Preusser M (2014). BRAF PMID:9242217 .",
JM, Wallace AJ, Gillespie JE, et al. (2004). Ge- neurofibroma. Genes Chromosomes Cancer. alterations in brain tumours: molecular patho- 189. Bhatti P, Veiga LH, Ronckers CM, Sigurd> -; ..
notype-phenotype correlations for nervous sys- 51(9):852-7. PMID:22585738 logy and therapeutic opportunities. Curr Opin son AJ, Stovall M, Smith SA, et al. (2010). Risk :-�

..
tem tumors in neurofibromatosis 2: a populati- 157. Behdad A, Perry A (2010). Central nervous Neural. 27(6):689-96. PMID:25268071 of second primary thyroid cancer after radiolhe- ...
on-based study. Am J Hum Genet. 75(2):231-9. system primitive neuroectodermal tumors: a cli- 172. Berghoff AS, Rajky 0, Winkler F, Bartsch rapy for a childhood cancer in a large cohort ..
PMID:15190457 nicopathologic and genetic study of 33 cases. R, Furtner J, Hainfellner JA, et al. (2013). study: an update from the childhood canceri;.'

358 References
survivor study. Radial Res. 174(6):741-52. Res. 68(19):7838-45. PMID:18829539 223. Blumcke I, Lobach M, Wolf HK, Wiestler 238. Boop FA (2011 ). Repeat surgery for re-
PMID:21128798 207. Birch BD, Johnson JP, Parsa A, Desai OD (1999). Evidence for developmental pre- sidual ependymoma. J Neurosurg Pediatr.
190. Bianco AdeM, Madeira LV, Rosemberg S, RD, Yoon JT, Lycette CA, et al. (1996). Fre- cursor lesions in epilepsy-associated glioneu- 8(3):244-5, discussion 245. PMID:21882913
Shibata MK (2006). Cortical seeding of a cra- quent type 2 neurofibromatosis gene transcript ronal tumors. Microsc Res Tech. 46(1 ):53-8. 239. Borges MT, Lillehei KO, Kleinschmidt-De-
niopharyngioma after craniotomy: Case report. mutations in sporadic inlramedullary spinal cord PMID:10402272 Masters BK (2011). Spindle cell oncocytoma
Surg Neural. 66(4):437-40, discussion 440. ependymomas. Neurosurgery. 39(1):135-40. 224. Blumcke I, Muller S, Buslei R, Riederer with late recurrence and unique neuroimaging
PMID:17015135 PMID:8805149 BM, Wiestler OD (2004). Microtubule-asso- characteristics due to recurrent subclinical in-
191. Biegel JA (1999). Cytogenetics and mole- 208. Birch JM, Blair V, Kelsey AM, Evans DG, ciated protein-2 immunoreactivity: a useful tratumoral bleeding. J Neurooncol. 101(1):145-
cular genetics of childhood brain tumors. Neuro Harris M, Tricker KJ, et al. (1998). Cancer phe- tool in the differential diagnosis of low-grade 54. PMID:20495848
Oneal. 1(2):139-51. PMID:11550309 notype correlates with constitutional TP53 ge- neuroepithelial tumors. Acta Neuropathol. 240. BoritA, Blackwood W, MairWG (1980). The
192. Biegel JA (2006). Molecular genetics of notype in families with the Li-Fraumeni syndro- 108(2):89-96. PMID:15146346 separation of pineocytoma from pineoblastoma.
atypical teratoid/rhabdoid tumor. Neurosurg me. Oncogene. 17(9):1061-8. PMID:9764816 225. Blumcke I, Thom M, Aronica E, Armstrong Cancer. 45(6):1408-18. PMID:6986979
Focus 20(1):E11. PMID:16459991 209. Bisceglia M, Galliani C, Giannatempo G, DD, Vinters HV, Palmini A, et al. (2011). The 241. Borkowska J, Schwartz RA, Kotulska K,
193. Biegel JA, Busse TM, Weissman BE Lauriola W, Bianco M, D'angelo V, et al. (2011 ). clinicopathologic spectrum of focal cortical dys- Jozwiak S (2011). Tuberous sclerosis complex:
(2014). SWI/SNF chromatin remodeling com- Solitary fibrous tumor of the central nervous plasias: a consensus classification proposed by tumors and tumorigenesis. Int J Derrnatol.
plexes and cancer. Am J Med Genet C Semin system: a 15-year literature survey of 220 ca- an ad hoc Task Force of the ILAE Diagnostic 50(1):13-20. PMID:21182496
Med Genet. 166C(3):350--<J6. PMID:25169151 ses (August 1996-July 2011). Adv Anal Pathol. Methods Commission. Epilepsia. 52(1):158-74. 242. Borota OC, Scheie D, Bjerkhagen B, Ja-
194. Biegel JA, Zhou JY. Rorke LB, Stenstrom 18(5):356-92. PMID:21841406 PMID:21219302 cobsen EA, Skullerud K (2006). Gliosarcoma
C, Wainwright LM, Fogelgren B (1999). Germ- 210. Bjornsson J, Scheithauer BW, Okazaki H, 226. Blumcke I, Wiestler OD (2002). Ganglio- with liposarcomatous component, bone infiltra-
line and acquired mutations of INl1 in atypical Leech RW (1985). lntracranial germ cell tumors: gliomas: an intriguing tumor entity associated tion and exlracranial growth. Clin Neuropathol.
teratoid and rhabdoid tumors. Cancer Res. pathobiological and immunohistochemical as- with focal epilepsies. J Neuropathol Exp Neural. 25(4):200-3. PMID:16866302
59(1):74-9. PMID:9892189 pects of 70 cases. J Neuropathol Exp Neural. 61(7):575-84. PMID:12125736 243. Borota OC, Scheithauer BW, Fougner
195. Bielle F, Navarro S, Bertrand A, Cornu 44(1):32-46. PMID:4038412 227. Bodey B, Bodey B Jr, Siegel SE (1995). SL, Hald JK, Ramm-Pettersen J, Bollers-
P, Mazeron JJ, Jouvet A, et al. (2014). Late 211. Blades DA, Hardy RW, Cohen M (1991). lmmunophenotypic characterization of infiltra- lev J (2009). Spindle cell oncocytoma of the
dural relapse of a resected and irradiated Cervical paraganglioma with subsequent in- ting polynuclear and mononuclear cells in child- adenohypophysis: report of a case with marked
pineal parenchymal tumor of intermediate dif- lracranial and inlraspinal metastases. Case re- hood brain tumors. Mod Pathol. 8(3):333-8. cellular atypia and recurrence despite adju-
ferentiation. Clin Neuropathol. 33(6):424-7. port. J Neurosurg. 75(2):320-3. PMID:2072174 PMID:7617661 vant treatment. Clin Neuropathol. 28(2):91-5.
PMID:24887399 212. Blandino G, Levine AJ, Oren M (1999). 228. Bodi I, Curran 0, Selway R, Elwes R, PMID:19353839
196. Bielle F, Villa C, Giry M, Bergemer-Fou- Mutant p53 gain of function: differential ef- Burrone J, Laxton R, et al. (2014). Two ca- 244. Borovich B, Doran Y (1986). Recurrence
quet AM, Polivka M, Vasiljevic A, et al.; RENOP fects of different p53 mutants on resistance ses of multinodular and vacuolating neuron- of intracranial meningiomas: the role played by
(2015). Chordoid gliomas of the third ventricle of cultured cells to chemotherapy. Oncogene. al tumour. Acta Neuropathol Commun. 2:7. regional multicentricity. J Neurosurg. 64(1):58-
share TIF-1 expression with organum vasculo- 18(2):477-85. PMID:9927204 PMID:24444358 63. PMID:3941351
sum of the lamina terminalis. Am J Surg Pathol. 213. Blaser SI, Harwood-Nash DC (1996). 229. Bodi I, Selway R, Bannister P, Doey L, 245. Bosman FT, Carneiro F, Hruban RH, The-
39(7):948-56. PMID:25786084 Neuroradiology of pediatric posterior Iossa Mullatli N, Elwes R, et al. (2012). Diffuse form ise ND, editors. (2010). WHO Classification of
197. Biernat W, Aguzzi A, Sure U, Grant JW, medulloblastoma. J Neurooncol. 29(1):23-34. of dysembryoplastic neuroepithelial tumour: Tumours of the Digestive System. 4th ed. Lyon:
Kleihues P, Hegi ME (1995). Identical muta- PMID:8817413 the histological and immunohistochemical fea- International Agency for Research on Cancer.
tions of the p53 tumor suppressor gene in the 214. Bleeker FE, Alai NA, Lamba S, Jonker A, tures of a dislinct entity showing transition to 246. Bostrom J, Meyer-Puttlitz B, Wolter M,
gliomatous and the sarcomatous components Rijkeboer D, Bosch KS, el al. (2010). The prog- dysembryoplastic neuroepithelial tumour and Blaschke B, Weber RG, Lichter P, et al. (2001).
of gliosarcomas suggest a common origin nostic IDH1( R132) mutation is associated with ganglioglioma. Neuropathol Appl Neurobiol. Alterations of the tumor suppressor genes
from gliat cells. J Neuropalhol Exp Neural. reduced NADP+-dependent IDH activity in gli- 38(5):411-25. PMID:21988102 CDKN2A (p16(1NK4a)), p14(ARF), CDKN2B
54(5):651-6. PMID:7666053 oblastoma. Acta Neuropathol. 119(4):487-94. 230. Bodo S, Colas C, Buhard 0, Collura A, (p15(1NK4b)), and CDKN2C (p18(1NK4c)) in
198. Biernat W, Huang H, Yokoo H, Kleihues PMID:20127344 Tina\ J, Lavoine N, el al.; European Consor- atypical and anaplastic meningiomas. Am J Pa-
P, Ohgaki H (2004 ). Predominant expression 215. Bleistein M, Geiger K. Franz K, Stoldt P, tium "Care for CMMRD" (2015). Diagnosis of thol. 159(2):661-9. PMID:11485924
of mutant EGFR (EGFRvlll) is rare in primary Schlote W (2000). Transthyretin and transferrin Constitutional Mismatch Repair-Deficiency 247. Bouffard JP, Sandberg GD, Golden JA,
glioblastomas. Brain Pathol. 14(2):131-6. in hemangioblastoma slromal cells. Pathol Res Syndrome Based on Microsatellite Instabili- Rorke LB (2004). Double immunolabeling
PMID:15193025 Pract. 196(10):675-81. PMID:11087054 ty and Lymphocyte Tolerance to Methylaling of central nervous system atypical teratoid/
199. Biernat W, Kleihues P, Yonekawa Y, Oh- 216. Blesa D, Mollejo M, Ruano Y, de Lope AR, Agents. Gastroenterology. 149(4):1017-1029. rhabdoid tumors. Mod Pathol. 17(6):679-83.
gaki H (1997). Amplification and overexpres- Fiario C, Ribalta T, el al. (2009). Novel genomic e3. PMID:26116798 PMID:15105808
sion of MDM2 in primary (de nova) glioblasto- alterations and mechanisms associated with 231. Boehm D, Bacher J, Neumann HP (2007). 248. Boutte\ E, Perilongo G, Canete A, Mas-
mas. J Neuropathol Exp Neural. 56(2):180-5. tumor progression in oligodendroglioma and Gross genomic rearrangement involving the simino M (1998). lntracranial ependymomas
PMID:9034372 mixed oligoastrocytoma. J Neuropathol Exp TSC2-PKD1 contiguous deletion syndro- in children: a critical review of prognostic
200. Biernat W, Tohma Y, Yonekawa Y, Klei- Neural. 68(3):27 4-85. PMID:19225409 me: characterization of the deletion event by factors and a plea for cooperation. Med Pedi-
hues P, Ohgaki H (1997). Alterations of cell 216A. Blitshteyn S, Crook JE, Jaeckle KA quantitative polymerase chain reaction dele- atr Oneal. 30(6):319-29, discussion 329-31.
cycle regulatory genes in primary (de nova) and (2008). Is there an association between menin- tion assay. Am J Kidney Dis. 49(1):e11-21. PMID:9589080
secondary glioblastomas. Acta Neuropathol. gioma and hormone replacement therapy? J PMID:17185137 249. Bourdeau\ F, Lequin D, Bruqieres L, Reyn-
94(4):303-9. PMID:9341929 Clin Oneal. 26(2):279-82. PMID:18182668 232. Baerman RH, Andert K, Herath J, Borell aud S, Dufour C, Doz F, et al. (2011). Frequent
201. Bigelow DC, Eisen MD, Smith PG, Yousem 217. Blough MD, Al-Najjar M, Chesnelong C, T, Johnson N, Schaeffer-Klein J, et al. (1996). hSNF5/INl1 germline mutations in patients with
DM, Levine RS, Jackler RK, et al. (1998). lipo- Binding CE, Rogers AD, Luchman HA, el al. The glial and mesenchymal elements of glio- rhabdoid tumor. Clin Cancer Res. 17(1):31-8.
mas of the internal auditory canal and cerebel- (2012). DNA hypermelhylation and 1p Loss si- sarcomas share similar genetic alterations. PMID:21208904
lopontine angle. Laryngoscope. 108(10):1459- lence NHE-1 in oligodendroglioma. Ann Neural. J Neuropathol Exp Neural. 55(9):973-81. 249A. Bourdeau\ F, Miquel C, Richer W, Grill
69. PMID:9778284 71(6):845-9. PMID:22718548 PMID:8800093 J, Zerah M, Grison C, et al. (2014). Rubin-
202. Biggs PJ, Garen PD, Powers JM, Garvin 218. Blount JP, Elton S (2001). Spinal lipomas. 233. Bognar L, Balint K, Bardoczy z (2002). stein- Taybi syndrome predisposing to non-WNT,
AJ (1987). Malignant rhabdoid tumor of the cen- Neurosurg Focus. 10(1):e3. PMID:16749755 Symptomatic osteolipoma of the tuber cinere- non-SHH, group 3 medulloblastoma. Pediatr
tral nervous system. Hum Pathol. 18(4):332-7. 219. Blumcke I, Aronica E, Urbach H, Alexopou- um. Case report. J Neurosurg. 96(2):361-3. Blood Cancer. 61(2):383-6. PMID:24115570
PMID:3030922 los A, Gonzalez-Martinez JA (2014 ). A neuropa- PMID:11838812 250. Bourekas EC, Bell SD, Ladwig NR, Gand-
203. Bigner SH, Mclendon RE, Fuchs H, thology-based approach lo epilepsy surgery 234. Boguski MS, McCormick F (1993). Pro- he AR, Shilo K, McGregor JM, el al. (2014).
McKeever PE, Friedman HS (1997). Chromo- in brain tumors and proposal for a new termi- teins regulating Ras and its relatives. Nature. Anaplastic papillary glioneuronal tumor with
somal characteristics of childhood brain tu- nology use for long-term epilepsy-associated 366(6456):643-54. PMID:8259209 extraneural metastases. J Neuropathol Exp
mors. Cancer Genet Cytogenet. 97(2):125-34. brain tumors. Acta Neuropathol. 128(1):39-54. 235. Bolcekova A, Nemethova M, Zatkova Neural. 73(5):474-6. PMID:24709681
PMID:9283596 PMID:24858213 A, Hlinkova K, Pozgayova S, Hlavata A, et al. 251. Bourgeois M, Sainte-Rose C, Lellouch-Tu-
204. Bijlsma EK, Merel P, Bosch DA, Wesler- 220. Blumenthal GM, Dennis PA (2008). PTEN (2013). Clustering of mutations in the 5' tertile biana A, Malucci C, Brunelle F, Maixner W,
veld A, Delattre 0, Thomas G, et al. (1994). hamartoma tumor syndromes. Eur J Hum Ge- of the NF1 gene in Slovakia patients with optic et al. (1999). Surgery of epilepsy associated
Analysis of mutations in the SCH gene in net. 16(11):1289-300. PMID:18781191 pathway glioma. Neoplasma. 60(6):655-65. with focal lesions in childhood. J Neurosurg.
schwannomas. Genes Chromosomes Cancer. 221. Blumcke I, Becker AJ, Normann S, Hans V, PMID:23906300 90(5):833-42. PMID:10223448
11(1):7-14. PMID:7529050 Riederer BM, Krajewski S, et al. (2001). Dislincl 236. Bonnin JM, Rubinstein LJ (1989). Astrob- 252. Bourn D, Carter SA, Mason S, Gareth D,
205. Bilsky MH, Schefler AC, Sandberg DI, Dun- expression pattern of microtubule-associated lastomas: a pathological study of 23 tumors, Evans R, Strachan T (1994). Germline muta-
kel IJ, Rosenblum MK (2000). Sclerosing epit- protein-2 in human oligodendrogliomas and with a postoperative follow-up in 13 patients. tions in the neurofibromatosis type 2 tumour
helioid fibrosarcomas involving the neuraxis: re- glial precursor cells. J Neuropathol Exp Neural. Neurosurgery. 25(1):6-13. PMID:2755581 suppressor gene. Hum Mol Genet. 3(5):813-6.
port of three cases. Neurosurgery. 47(4):956-9, 60(10):984-93. PMID:11589429 237. Booman M, Douwes J, Glas AM, Rie- PMID:8081368
discussion 959-60. PMID:11014436 222. Blumcke I, Giencke K, Wardelmann E, mersma SA, Jordanova ES, Kok K, et al. 253. Bourne TD, Mandell JW, Matsumoto JA,
206. Binning MJ, Niazi T, Pedone CA, Lal B, Beyenburg S, Kral T, Sarioglu N, et al. (1999). (2006). Mechanisms and effects of loss of Jane JA Jr, Lopes MB (2006). Primary dissemi-
Eberhart CG, Kim KJ, el al. (2008). Hepatocyte The CD34 epitope is expressed in neopla- human leukocyte antigen class II expression nated leptomeningeal oligodendroglioma with
growth factor and sonic Hedgehog expression stic and malforrnative lesions associated with in immune-privileged site-associated B-cell 1p deletion. Case report. J Neurosurg. 105(6)
in cerebellar neural progenitor cells coslimulate chronic, focal epilepsies. Acta Neuropathol. lymphoma. Clin Cancer Res. 12(9):2698-705. Suppl:465-9. PMID:17184079
medulloblastoma initiation and growth. Cancer 97(5):481-90. PMID:10334485 PMID:16675561 254. Bourne TD, Schiff D (2010). Update on

References 359
molecular findings, management and outco- outcome. Am J Surg Pathol. 25(10):1252-60. MM, Proko E, Kocak M, et al. (2007). Clinical Degl'innocenti DR, Moncini D, Spacca B, Giorv
me in low-grade gliomas. Nat Rev Neurol. PMID:11688459 and molecular characteristics of malignant dano F, et al. (2_013)_. Ang1ocentric glioma: cli-
6(12):695-701. PMID:21045797 270. Brat DJ, Scheithauer BW, Fuller GN, Tihan transformation of low-grade glioma in children. nical, morphological, 1mmunoh1stochemicaJ anc(
255. Bouvier C, Bertucci F, Metellus P, Finetti T (2007). Newly codified glial neoplasms of the J Clin Oncol. 25(6):682-9. PMID:17308273 molecular features in three pediatric cases. Clin. _
P, Maues de Paula A, Forest F, et al. (2013). 2007 WHO Classification of Tumours of the 285. Broniscer A, Tatevossian RG, Sabin ND, Neuropathol. 32(2):107-13. PMID23073165
ALDH1 is an immunohistochemical diagnostic Central Nervous System: angiocentric glioma, Klimo P Jr, Dallon J, Lee R, et al. (2014). Clini- 301. Buccoliero AM, Castiglione F, Rossi De _
marker for solitary fibrous tumours and hae- pilomyxoid astrocytoma and pituicytoma. Brain cal, radiological, histological and molecular cha- gl'lnnocenti D, Franchi A, Paglierani M, Sanzl
mangiopericytomas of the meninges emerging Pathol. 17(3):319-24. PMID:17598825 racteristics of paediatric epithelioid glioblasto- M, et al. (2010). Embryonal tumor with abun-
from gene profiling study. Acta Neuropathol 271. Brat DJ, Scheithauer BW, Medina-Flores ma. Neuropathol Appl Neurobiol. 40(3):327-36. dant neuropil and true rosettes: morphologi-
Commun. 1:10. PMID:24252471 R, Rosenblum MK, Burger PC (2002). Infiltra- PMID24127995 cal, immunohistochemical, ultrastructural and
256. Bouvier C, Metellus P, de Paula AM, Vasil- tive astrocytomas with granular cell features 286. Brown AE, Leibundgut K, Niggli FK, Betts molecular study of a case showing features o(
jevic A, Jouvet A, Guyotat J, et al. (2012). Soli- (granular cell astrocytomas): a study of histo- DR (2006). Cytogenetics of pineoblastoma: four medullo_epithelioma and areas of mesenchymal
tary fibrous tumors and hemangiopericytomas pathologic features, grading, and outcome. Am new cases and a literature review. Cancer Ge- and epithelial differenliation. Neuropathology. _
of the meninges: overlapping pathological fea- J Surg Pathol. 26(6):750-7. PMID:12023579 net Cytogenet. 170(2):175-9. PMID:17011992 30(1):84-91. PMID:19563506 '
tures and common prognostic factors suggest 272. Brat DJ, Scheithauer BW, Staugaitis SM, 287. Brown HG, Burger PC, Olivi A, Sills AK, 302. Buccoliero AM, Franchi A, Castigli1r
the same spectrum of tumors. Brain Pathol. Cortez SC, Brecher K, Burger PC (1998). Third Barditch-Crovo PA, Lee RR (1999). lntracranial ne F, Gheri CF, Mussa F, Giordano F, et al.- =
22(4):511-21. PMID:22082190 ventricular chordoid glioma: a distinct clinico- leiomyosarcoma in a patient with AIDS. Neuro- (2009). Subependymal giant cell astrocytoma ·
257. Bouvier-Labit C, Daniel L, Dufour H, Grisoli pathologic entity. J Neuropathol Exp Neurol. radiology. 41(1):35-9. PMID:9987766 (SEGA): Is it an astrocytorna? Morphological
F, Figarella-Branger D (2000). Papillary glioneu- 57(3):283--90 PMID:9600220 288. Brown HG, Kepner JL, Perlman EJ, Fried- immunohistochemical and ultrastructural study:
ronal tumour: clinicopathological and biochemi- 273. Brat DJ, Scheithauer BW, Staugaitis SM, man HS, Strother DR, Duffner PK, et al. (2000). Neuropathology. 29(1):25-30. PMID:18564101'
cal study of one case with 7 -year follow up. Acta Holtzman RN, Morgello S, Burger PC (2000). "Large cell/anaplastic" medulloblastomas: a 303. Buccoliero AM, Giordano F, Mussa F
Neuropathol. 99(3):321--6. PMID:10663977 Pituicytoma: a distinctive low-grade glioma Pediatric Oncology Group Study. J Neuropathol Taddei A. Genitori L, Taddei GL (2006). Pa: -
258. Bowers DC, Gargan L, Kapur P, Reisch JS, of the neurohypophysis. Am J Surg Pathol. Exp Neurol. 59(10):857-65. PMID:11079775 pillary glioneuronal tumor radiologically mimF •.
Mulne AF, Shapiro KN, et al. (2003). Study of 24(3):362--8. PMID:10716149 289. Brown JA, Gianino SM, Gutmann DH eking a cavernous hemangioma with hemorr-
the MIB-1 labeling index as a predictor of tumor 273A. Brede! M, Scholtens DM, Yadav AK, (2010). Defective cAMP generation under- hagic onset. Neuropathology. 26(3):206--11.
progression in pilocytic astrocytomas in children Alvarez AA, Renfrow JJ, Chandler JP, et al. lies the sensitivity of CNS neurons to neu- PMID:16771176
and adolescents. J Clin Oncol. 21(15):2968--73. (2011). NFKBIA deletion in glioblastomas. N rofibromatosis-1 heterozygosity. J Neurosci. 304. Buczkowicz P, Bartels U, Bouffet E, Be-
PMID:12885817 Engl J Med. 364(7):627-37. PMID:21175304 30(16):5579--89. PMID:20410111 cher 0, Hawkins C (2014). Histopathologica(
259. Bowers DC, Mulne AF, Weprin B, Bruce 27 4. Brekke HR, Ribeiro FR, Kolberg M, 290. Brown NA, Furtado LV, Betz BL, Kiel MJ, spectrum of paediatric diffuse intrinsic ponti-· ·
DA, Shapiro K, Margraf LR (2002). Progno- Agesen TH, Lind GE, Eknaes M, et al. (2010). Weigelin HC, Lim MS, et al. (2014). High preva- ne glioma: diagnostic and therapeutic impli,
stic factors in children and adolescents with Genomic changes in chromosomes 10, 16, and lence of somatic MAP2K 1 mutations in BRAF cations. Acta Neuropathol. 128(4 ):573-81.
low-grade oligodendrogliomas. Pediatr Neuro- X in malignant peripheral nerve sheath tumors V600E-negative Langerhans cell histiocytosis. PMID:25047029
surg. 37(2):57-63. PMID:12145513 identify a high-risk patient group. J Clin Oncol. Blood. 124(10):1655-8. PMID:24982505 305. Buczkowicz P, Hoeman C, Rakopoulos ·
260. Boyd C, Smith MJ, Kluwe L. Balogh A, 28(9):1573-82. PMID:20159821 291. Brown PD, Buckner JC, O'Fallon JR, lturria P, Pajovic S, Letourneau L, Dzamba M, et al, •
Maccollin M, Plotkin SR (2008). Alterations 275. Brems H, Chmara M, Sahbatou M, Denay- NL, Brown CA, O'Neill BP, et al.; North Central (2014). Genomic analysis of diffuse intrinsic �
in the SMARCB1 (INl1) tumor suppressor er E, Taniguchi K, Kato R, et al. (2007). Germ- Cancer Treatment Group; Mayo Clinic (2004). pontine gliomas identifies three molecular sub-:""
gene in familial schwannomatosis. Clin Genet. line loss-of-function mutations in SPRED1 cau- Adult patients with supratentorial pilocytic ast- groups and recurrent activating ACVR1 muta-
74(4):358-66. PMID:18647326 se a neurofibromatosis 1-like phenotype. Nat rocytomas: a prospective multicenter clinical tri- tions. Nat Genet. 46(5):451-6. PMID:24705254-
261. Brannan Cl, Perkins AS, Vogel KS, Ratner Genet. 39(9):1120-6. PMID:17704776 al. Int J Radial Oncol Biol Phys. 58(4):1153-60. 306. Budka H (1974). lntracranial lipomatous %

N, Nordlund ML, Reid SW, et al. (1994). Targe- 276. Brennan C (2011 ). Genomic profiles of gli- PMID:15001258 hamartomas (intracranial "lipomas"). A study·;
ted disruption of the neurofibromatosis type-1 oma. Curr Neurol Neurosci Rep. 11(3):291-7. 292. Brown R, Zlatescu M, Sijben A, Roldan of 13 cases including combinations with me-
gene leads to developmental abnormalities in PMID:21465149 G, Easaw J, Forsyth P, et al. (2008). The use dulloblastoma, colloid and epidermoid cysts,
heart and various neural crest-derived tissues. 277. Brennan CW, Verhaak RG, McKenna A, of magnetic resonance imaging to noninvasi- angiomatosis and other malformations. Acta·
Genes Dev. 8(9):1019-29. PMID:7926784 Campos B, Noushmehr H, Salama SR, et al.; vely detect genetic signatures in oligodendro- Neuropathol. 28(3):205-22. PMID:4611131
262. Brastianos PK, Horowitz PM, Santagata S, TCGA Research Network (2013). The somatic glioma. Clin Cancer Res. 14(8):2357-62. 307. Budka H (1975). Partially resected and
Jones RT, McKenna A, Getz G, et al. (2013). genomic landscape of glioblastoma. Cell. PMID:18413825 irradiated cerebellar astrocytoma of childhood: _
Genomic sequencing of meningiomas identifies 155(2):462-77. PMID:24120142 293. Brownstein MH, Wolf M, Bikowski JB malignant evolution after 28 years. Acta Neu-
oncogenic SMO and AKT1 mutations. Nat Ge- 278. Bridge JA, Liu XQ, Sumegi J, Nelson M, (1978). Cowden's disease: a cutaneous mar- rochir (Wien). 32(1-2):139-46. PMID:1163315. •
net. 45(3):285-9. PMID:23334667 Reyes C, Bruch LA, et al. (2013). Identification ker of breast cancer. Cancer. 41 (6):2393--8. 308. Budka H, Chimelli L (1994). Lipomatous
263. Brastianos PK, Taylor-Weiner A, Manley of a novel, recurrent SLC44A 1-PRKCA fusion PMID:657103 medulloblastoma in adults: a new tumor type
PE, Jones RT, Dias-Santagata D, Thorner in papillary glioneuronal tumor. Brain Pathol. 294. Bruqieres L, Remenieras A, Pierron G, with possible favorable prognosis. Hum Pathol.,
AR, et al. (2014). Exome sequencing identifies 23(2):121--8. PMJD:22725730 Varlet P, Forget S, Byrde V, et al. (2012). High 25(7):730-1. PMID8026834
BRAF mutations in papillary craniopharyngio- 279. Bridge RS, Rajaram V, Dehner LP, Pfei- frequency of germline SUFU mutations in chil- 309. Bukhtoiarov AP, Zil'berberg LB, Levitski?
mas. Nat Genet. 46(2):161-5. PMID:24413733 fer JD, Perry A (2006). Molecular diagnosis of dren with desmoplasticlnodular medulloblasto- VA (1975). [Evacuanon hospitals in Pyatigorsk =,
264. Brat DJ, Castellano-Sanchez AA, Hunter Ewing sarcoma/primitive neuroectodenmal tu- ma younger than 3 years of age. J Clin Oncol. during the Great Patriotic War] . Zdravookhr
SB, Pecot M, Cohen C, Hammond EH, et al. mor in routinely processed tissue: a comparison 30(17):2087-93. PMID 22508808 Ross Fed : 22-3. PMID:131445
(2004 ). Pseudopalisades in glioblastoma are of two FISH strategies and RT-PCR in malig- 296. Brunn A, Nagel I, Montesinos-Rongen M, 310. Bunin GR, Surawicz TS, Witman PA, Pres-·-
hypoxic, express extracellular matrix protea- nant round cell tumors. Mod Pathol. 19(1):1-8. Klapper W, Valer I, Paulus W, et al. (2013). ton-Martin S, Davis F, Bruner JM (1998). The
ses, and are formed by an actively migrating PMID:16258512 Frequent triple-hit expression of MYC, BCL2, descriptive epidemiology of craniopharyngio- .'
cell population. Cancer Res. 64(3):920-7. 280. Briscoe J, Therond PP (2013). The me- and BCL6 in primary lymphoma of the central ma. J Neurosurg. 89(4):547-51. PMID:9761047"··
PMID:14871821 chanisms of Hedgehog signalling and its roles nervous system and absence of a favorable 311. Burger PC (1996). Pathology of brain stem·;
265. Brat DJ, Cohen KJ, Sanders JM, Feuer- in development and disease. Nat Rev Mol Cell MYC(low)BCL2 (low) subgroup may underlie astrocytomas. Pediatr Neurosurg. 24(1):35-40. ·
stein BG, Burger PC (1999). Clinicopathologic Biol. 14(7):416--29. PMID:23719536 the inferior prognosis as compared to systemic PMID:8817613
features of astroblastoma. J Neuropathol Exp 281. Brock JE, Perez-Atayde AR, Kozakewich diffuse large B cell lymphomas. Acta Neuropa- 312. Burger PC (2009). Smears and frozen sec-
Neurol. 58:509. HP, Richkind KE, Fletcher JA, ;Jargas SO thol. 126(4):603--5. PMID:24061549 tions in surgigal neuropathology. PB Medical • � •
266. Brat DJ, Giannini C, Scheithauer BW, Bur- (2005). Cytogenetic aberrations in perineurio- 297. Bruno A, Boisselier B, Labreche K, Marie Publishing: Baltimore. .•
ger PC (1999). Primary melanocytic neoplasms ma: variation with subtype. Am J Surg Pathol. Y, Polivka M, Jouvet A, et al. (2014). Mutatio- 313. Burger PC (2012). Paraganglioma of the,:"
of the central nervous systems. Am J Surg Pa- 29(9):1164-9. PMID:16096405 nal analysis of primary central nervous sys- filum terminale. In: Burger PC, Scheithauer BW,
thol. 23(7):745-54. PMID:10403296 282. Brodbelt A, Greenberg D, Winters T. Wil- tem lymphoma. Oncotarget. 5(13):5065-75. Kleinschmidt-DeMaster BK, Ersen A, Rodriguez
267. Brat DJ, Hirose Y, Cohen KJ, Feuer- liams M, Vernon S, Collins VP; (UK) National PMID:24970810 FJ, Tihan T, et al., editors. Diagnostic Pathofo- ._,
stein BG, Burger PC (2000). Astroblastoma: Cancer Information Network Brain Tumour 298. Bruck W, Brunn A, Klapper W, Kuhlmann gy, Neuropathology. Salt Lake City: Amysys. -
clinicopathologic features and chromosomal Group (2015). Glioblastoma in England: T, Metz I, Paulus W, et al.; Netzwerk Lymphome 314. Burger PC, Grahmann FC, Bliestle A,
abnormalities defined by comparative geno- 2007-2011. Eur J Cancer. 51(4):533-42. und Lymphomatoide Uisionen des Nervensys- Kleihues P (1987). Differentiation in the medul·
mic hybridization. Brain Pathol. 10(3):342-52. PMID:25661102 tems (2013). [Differential diagnosis of lymphoid loblastoma. A histological and immunohistoche-
PMID:10885653 282A. Broderick DK, Di C, Parrett T J, Samuels infiltrates in the central nervous system: experi- mica! study. Acta Neuropathol. 73(2):115-23.
268. Brat DJ, Ryken TC, Kalkanis SN, Olson JJ; YR, Cummins JM, Mclendon RE, et al. (2004). ence of the Network Lymphomas and Lympho- PMID:3604579
AANS/CNS Joint Guidelines Committee (2014). Mutations of PIK3CA in anaplastic oligodendro- matoid Lesions in the Nervous System]. Patho- 315. Burger PC, Green SB (1987). Patient age,
The role of neuropathology in the management gliomas, high-grade astrocytomas, and me- loge. 34(3):186-97. PMID:23471726 histologic features, and length of survival in
of progressive glioblastoma a systematic dulloblastomas. Cancer Res. 64(15):5048--50. 299. Buccoliero AM, Bacci S, Mennonna P, patients with glioblastoma multiforme. Cancer.
review and evidence-based clinical practice PMID:15289301 Taddei GL (2004). Pathologic quiz case: in- 59(9):1617-25. PMID:3030531
guideline. J Neurooncol. 118(3):461-78. 283. Broholm H, Madsen FF, Wagner AA, Lau- fratentorial tumor in a middle-aged woman. 316. Burger PC, Heinz ER, Shibata T, Kleihues
PMID:24733643 rsen H (2002). Papillary glioneuronal tumor-a Oncocytic variant of choroid plexus papillo- P (1988). Topographic anatomy and CT correla-
269. Brat DJ, Scheithauer BW, Eberhart new tumor entity. Clin Neuropathol. 21(1):1-4. ma. Arch Pathol Lab Med. 128(12):1448--50. tions in the untreated glioblastoma multiforme.
CG, Burger PC (2001). Extraventricular neu- PMID:11846038 PMID 15578895 J Neurosurg. 68(5):698-704. PMID:2833587
rocytomas: pathologic features and clinical 284. Broniscer A, Baker SJ, West AN, Fraser 300. Buccoliero AM, Castiglione F, 317. Burger PC, Kleihues P (1989). Cytologic

360 References
composition of the untreated glioblastoma with 59(3):229-40. PMID:10744061 348. Campos AR, Clusmann H, van Lehe M, PMID:24929912
implications for evaluation of needle biopsies. 333. Buschges R, lchimura K, Weber RG, Rei- Niehusmann P, Becker AJ, Schramm J, et al. 363. Carlotti CG Jr, Salhia B, Weitzman S,
Cancer 63(10):2014-23. PMID:2539242 fenberger G, Collins VP (2002). Allelic gain and (2009). Simple and complex dysembryoplastic Greenberg M, Dirks PB, Mason W, et al. (2002).
318. Burger PC, Pearl DK, Aldape K, Yates AJ, amplification on the long arm of chromosome neuroepithelial tumors (ONT) variants: clinical Evaluation of proliferative index and cell cycle
Scheithauer BW, Passe SM, et al. (2001 ). Small 17 in anaplastic meningiomas. Brain Pathol. profile, MRI, and histopathology. Neuroradiolo- protein expression in choroid plexus tumors
cell architecture-a histological equivalent of 12(2):145-53. PMID:11958368 gy. 51(7):433-43. PMID:19242688 in children. Acta Neuropathol. 103(1):1-10.
EGFR amplification in glioblastoma multiforme? 334. Buttner A, Marquart KH, Mehraein P, Weis 349. Brat DJ, Verhaak RG, Aldape KO, Yung PMID:11837741
J Neuropathol Exp Neural. 60(11):1099-104. S (1997). Kaposi's sarcoma in the cerebel- WK, Salama SR, Cooper LA, et al.; Cancer 364. Carlson GJ, Nivatvongs S, Snover DC
PMID:11706939 lum of a patient with AIDS. Clin Neuropathol. Genome Atlas Research Network (2015). Com- (1984). Colorectal polyps in Cowden's disease
319. Burger PC, Scheithauer B (2008).Tumors 16(4):185-9. PMID:9266142 prehensive, Integrative Genomic Analysis of (multiple hamartoma syndrome). Am J Surg Pa-
of the Central Nervous System. AFIP Atlas of 335. Bohling T, Hatva E, Kujala M, Claes- Diffuse Lower-Grade Gliomas. N Engl J Med. thol. 8(10):763--70. PMID:6496844
Tumour Pathology, Series 4, Fascicle 7 Was- son-Welsh L, Alitalo K, Haltia M (1996). Ex- 372(26):2481-98. PMID:26061751 365. Carneiro SS, Scheithauer BW, Nascimen-
hington DC: American Registry of Pathology. pression of growth factors and growth factor 350. Cancer Genome Atlas Research Network to AG, Hirose T, Davis DH (1996). Solitary fi-
320. Burger PC, Scheithauer BW (1994). Tu- receptors in capillary hemangioblastoma. (2008). Comprehensive genomic characteriz- brous tumor of the meninges: a lesion distinct
mors of the Central Nervous System. Washing- J Neuropathol Exp Neural. 55(5):522-7. ation defines human glioblastoma genes and from fibrous meningioma. A clinicopathologic
ton: Armed Forces Institute of Pathology. PMID:8627342 core pathways. Nature. 455(7216):1061-8. and immunohistochemical study. Am J Clin Pa-
321. Burger PC, Scheithauer BW, Vogel FS 336. Bohling T, Maenpaa A, Timonen T, Van- PMID:18772890 thol. 106(2):217-24 PMID:8712177
(2002). Surgical Pathology of the Nervous tunen L, Paetau A, Haltia M (1996). Different 351. Cannon DM, Mahindra P, Gondi V, Kruser 366. Carney EM, Banerjee P, Ellis CL, Alba-
System and Its Coverings. 4th ed. London: expression of adhesion molecules on stromal TJ, Kozak KR (2015). Choroid plexus tumor dine R, Sharma R, Chaux AM, et al. (2011).
Churchill Livingston. cells and endothelial cells of capillary heman- epidemiology and outcomes: implications for PAX2(-)/PAX8(-)rinhibin A(+) immunoprofile in
322. Burger PC, Vogel FS, Green SB, Strike gioblastoma. Acta Neuropathol. 92(5):461-6. surgical and radiotherapeutic management. J hemangioblastoma: A helpful combination in
TA (1985). Glioblastoma multiforme and ana- PMID:8922057 Neurooncol. 121(1):151-7. PMtD:25270349 the differential diagnosis with metastatic clear
plastic astrocytoma. Pathologic criteria and 337. Bohling T, Turunen o, Jaaskelanen J, 352. Cannon TC, Bane BL, Kistler D, Schoen- cell renal cell carcinoma to the central ner-
prognostic implications. Cancer. 56(5): 1106-11. Carpen 0, Sainio M, Wahlstrom T, et al. (1996). hals GW, Hahn M, Leech RW, et al. (1998). vous system. Am J Surg Pathol. 35(2):262-7.
PMID:2990664 Ezrin expression in stromal cells of capillary he- Primary intracerebellar osteosarcoma arising PMID:21263247
323. Burger PC, Vollmer RT (1980). Histologic mangioblastoma. An immunohistochemical sur- within an epidermoid cyst. Arch Pathol Lab 367. Carney JA (1990). Psammomatous me-
factors of prognostic significance in the gliob- vey of brain tumors. Am J Pathol. 148(2):367- Med. 122(8):737-9. PMID9701337 lanotic schwannoma. A distinctive, heritable tu-
lastoma multiforme. Cancer. 46(5):1179-86. 73. PMID:8579099 353. Cao D, Liu A, Wang F, Allan RW, Mei K, mor with special associations, including cardiac
PMID:6260329 338. Cabello A, Madero S, Castresana A, Di- Peng Y, et al. (2011 ). RNA-binding protein myxoma and the Cushing syndrome. Am J Surg
324. Burger PC, Yu IT, Tihan T, Friedman HS, az-Lobato R (1991). Astroblastoma: electron LIN28 is a marker for primary extragonadal Pathol. 14(3):206-22. PMID:2305928
Strother DR, Kepner JL, et al. (1998). Atypical microscopy and immunohistochemical fin- germ cell tumors: an immunohistochemical 368. Carney JA, Gordon H, Carpenter PC,
teratoid/rhabdoid tumor of the central nervous dings: case report. Surg Neurol. 35(2):116-21. study of 131 cases. Mod Pathol. 24(2):288-96. Shenoy BV, Go VL (1985). The complex of
system: a highly malignant tumor of infancy and PMID:1990478 PMID:21057460 myxomas, spotty pigmentation, and endocrine
childhood frequently mistaken for medullob- 339. Caccamo DV, Ho KL, Garcia JH (1992). 354. Capper D, Berghoff AS, Magerle M, overactivity. Medicine (Baltimore). 64(4):270-
lastoma: a Pediatric Oncology Group study. Am Cauda equina tumor with ependymal and llhan A, Wohrer A, Hackl M, et al. (2012). lm- 83. PMID:4010501
J Surg Pathol. 22(9):1083-92. PMID:9737241 paraganglionic differentiation. Hum Pathol. munohistochemical testing of BRAF V600E 369. Carrasco R, Pascual JM, Navas M, Fra-
325. Burkhard C, Di Paire PL, Schuler D, Schu- 23(7):835-8. PMID:1612583 status in 1, 120 tumor tissue samples of pati- ga J, Manzanares-Soler R, Sola RG (2010).
ler G, Ya$argil MG, Yonekawa Y, et al. (2003). 340. Cachia D, Prado MP, Theeler B, Hamilton ents with brain metastases. Acta Neuropathol. Spontaneous acute hemorrhage within a sub-
A population-based study of the incidence J, McCutcheon I, Fuller GN (2014). Synchro- 123(2):223-33. PMID:22012135 ependymoma of the lateral ventricle: successful
and survival rates in patients with pilocytic nous rosette-forming glioneuronal tumor and 355. Capper D, Preusser M, Habel A, Sahm emergent surgical removal through a frontal
astrocytoma. J Neurosurg. 98(6):1170-4. diffuse astrocytoma with molecular charac- F, Ackermann U, Schindler G, et al. (2011). transcortical approach. Neurocirugia (Astur).
PMID:12816259 terization: a case report. Clin Neuropathol. Assessment of BRAF V600E mutation status 21(6):478-83. PMID:21165545
326. Burkitt Wright EM, Sach E, Sharif S, Quar- 33(6):407-11. PMID:24986181 by immunohistochemistry with a mutation-spe- 370. Carstens PH, Johnson GS, Jelsma LF
rell 0, Carroll T, Whitehouse RW, et al. (2013). 341. Cady FM, O'Neill BP, Law ME, Decker cific monoclonal antibody. Acta Neuropathol. (1995). Spinal gliosarcoma: a light, immunohis-
Can the diagnosis of NF1 be excluded clini- PA, Kurtz OM, Giannini C, et al. (2008). Del(6) 122(1):11-9. PMID:21638088 tochemical and ultrastructural study. Ann Clin
cally? A lack of pigmentary findings in families (q22) and BCL6 rearrangements in primary 356. Capper D, Reuss D, Schittenhelm J, Hart- Lab Sci. 25(3):241-6. PMID:7605106
with spinal neurofibromatosis demonstrates a CNS lymphoma are indicators of an aggressive mann C, Bremer J, Sahm F, et al. (2011). Muta- 371. Carter JM, O'Hara C, Dundas G, Gilchrist
limitation of clinical diagnosis. J Med Genet. clinical course. J Clin Oneal. 26(29):4814--9. tion-specific IDH1 antibody differentiates oll- D, Collins MS, Eaton K, et al. (2012). Epithe-
50(9):606-13. PMID:23812910 PMID:18645192 godendrogliomas and oligoastrocytomas from lioid malignant peripheral nerve sheath tumor
327. Burnett ME, White EC, Sih S, van Haken 342. Cahill DP, Levine KK, Betensky RA, Codd other brain tumors with oligodendroglioma-like arising in a schwannoma, in a patient with
MS, Cogen PH (1997). Chromosome arm 17p PJ, Romany CA, Reavie LB, et al. (2007). Loss morphology. Acta Neuropathol. 121(2):241-52. "neuroblastoma-like" schwannomatosis and a
deletion analysis reveals molecular genetic he- of the mismatch repair protein MSH6 in human PMID:21069360 novel germline SMARCB1 mutation. Am J Surg
terogeneity in supratentorial and infratentorial glioblastomas is associated with tumor progres- 357. Capper D, Sahm F, Hartmann C, Meyer- Pathol. 36(1):154-60. PMID:22082606
primitive neuroectodermal tumors of the cent- sion during temozolomide treatment. Clin Can- mann R, van Deimling A, Schittenhelm J (2010). 372. Carter M, Nicholson J, Ross F, Crolla J,
ral nervous system. Cancer Genet Cytogenet. cer Res. 13(7):2038-45. PMID:17404084 Application of mutant IDH1 antibody to differen- Allibone R, Balaji V, et al. (2002). Genetic ab-
97(1):25-31. PMID:9242214 343. Cai DX, James CD, Scheithauer BW, tiate diffuse glioma from nonneoplastic central normalities detected in ependymomas by com-
328. Bush K, Bateman DE (2014 ). Papilloede- Couch FJ, Perry A (2001). PS6K amplificati- nervous system lesions and therapy-induced parative genomic hybridisation. Br J Cancer.
ma secondary to a spinal paraganglioma. Pract on characterizes a small subset of anaplastic changes. Am J Surg Pathol. 34(8):1199-204. 86(6):929-39. PMID:11953826
Neural. 14(3):179-81. PMID:23918468 meningiomas. Am J Clin Pathol. 115(2):213--8. PMID:20661018 373. Carvalho AT, Linhares P, Castro L, Sa MJ
329. Buslei R, Nolde M, Hofmann B, Meissner PMID:11211609 358. Capper D, Simon M, Langhans CD, Okun (2014). Multiple sclerosis and oligodendroglio-
S, Eyupoglu IY, Siebzehnnibl F, et al. (2005). 344. Cairncross G, Wang M, Shaw E, Jen- JG, Tonn JC, Weller M, et al.; German Glioma ma: an exceptional association. Case Rep Neu-
Common mutations of beta-catenin in adaman- kins R, Brachman D, Buckner J, et al. (2013). Network (2012). 2-Hydroxyglutarate concen- ral Med. 2014:546817. PMID:25180114
tinomatous craniopharyngiomas but not in other Phase Ill trial of chemoradiotherapy for ana- tration in serum from patients with gliomas 374. Casadei GP, Arrigoni GL, D'Angelo V, Biz-
tumours originating from the sellar region. Acta plastic oligodendroglioma: long-term results does not correlate with IDH1/2 mutation sta- zozero L (1990). Late malignant recurrence of
Neuropathol. 109(6):589-97. PMID: 15891929 of RTOG 9402. J Clin Oneal. 31 (3):337 �3. tus or tumor size. Int J Cancer. 131(3):766-8. childhood cerebellar astrocytoma. Clin Neuro-
330. Butler MG, Dasouki MJ, Zhou XP, Talebi- PMID:23071247 PMID:21913188 pathol. 9(6):295-8. PMID:2286021
zadeh Z, Brown M, Takahashi TN, et al. (2005). 345. Cairncross JG, Ueki K, Zlatescu MC, Lis- 359. Capper D, Weissert S, Balss J, Habel 375. Casadei GP, Komori T, Scheithauer BW,
Subset of individuals with autism spectrum le DK, Finkelstein OM, Hammond RR, et al. A, Meyer J, Jager D, et al. (2010). Characte- Miller GM, Parisi JE, Kelly PJ (1993). lntracra-
disorders and extreme macrocephaly associ- (1998). Specific genetic predictors of chemo- rization of R132H mutation-specific IDH1 an- nial parenchymal schwannoma. A clinicopatho-
ated with germline PTEN tumour suppressor therapeutic response and survival in patients tibody binding in brain tumors. Brain Pathol. logical and neuroimaging study of nine cases. J
gene mutations. J Med Genet. 42(4):318-21. with anaplastic oligodendrogliomas. J Natl Can- 20(1):245-54. PMID:19903171 Neurosurg. 79(2):217-22. PMID:8331403
PMID:15805158 cer Inst. 90(19):1473--9. PMID:9776413 360. Capper D, Zentgraf H, Balss J, Hartmann 376. Casadei GP, Scheithauer BW, Hirose T,
331. Byeon SJ, Cho HJ, Baek HW, Park CK, 346. Cairncross JG, Wang M, Jenkins RB, C, van Deimling A (2009). Monoclonal antibody Manfrini M, Van Hautan C, Wood MB (1995).
Choi SH, Kim SH, et al. (2014). Rhabdoid Shaw EG, Giannini C, Brachman DG, et al. specific for IDH1 R132H mutation. Acta Neuro- Cellular schwannoma. A clinicopathologic,
glioblastoma is distinguishable from classi- (2014). Benefit from procarbazine, lomustine, pathol. 118(5):599-601. PMID:19798509 DNA now cytometric, and proliferation marker
cal glioblastoma by cytogenetics and mole- and vincristine in oligodendroglial tumors is 361. Cardoso C, Lutz Y, Mignon C, Compe E, study of 70 patients. Cancer. 75(5):1109-19.
cular genetics. Hum Pathol. 45(3):611-20. associated with mutation of IDH. J Clin Oncol. Depetris D, Mattei MG, et al. (2000). ATR-X PMID:7850709
PMID:24457079 32(8):783--90. PMID:24516018 mutations cause impaired nuclear location and 377. Cassarino OS, Auerbach A, Rushing EJ
332. Buhren J, Christoph AH, Buslei R, Albrecht 347. Camelo-Piragua S, Jansen M, Ganguly A, altered DNA binding properties of the XNP/ (2003). Widely invasive solitary fibrous tumor
S, Wiestler OD, Pietsch T (2000). Expression of Kim JC, Cosper AK, Dias-Santagata D, et al. ATR-X protein. J Med Genet. 37(10):746-51. of the sphenoid sinus, cavernous sinus, and
the neurotrophin receptor p75NTR in medullob- (2011 ). A sensitive and specific diagnostic panel PMID:11015451 pituitary Iossa. Ann Diagn Pathol. 7(3):169-73.
lastomas is correlated with distinct histological to distinguish diffuse astrocytoma from astrocy- 362. Caretti V, Bugiani M, Frere! M, Schellen PMID:12808569
and clinical features: evidence for a medul- tosis: chromosome 7 gain with mutant isocitrate P, Jansen M, van Vuurden D, et al. (2014). 378. Casso! CA, Winer D, Liu W, Guo M, Ezzat
loblastoma subtype derived from the external dehydrogenase 1 and p53. J Neuropathol Exp Subventricular spread of diffuse intrinsic pon- S, Asa SL (2014). Tyrosine kinase receptors as
granule cell layer. J Neuropathol Exp Neurol. Neurol. 70(2):110-5. PMID:21343879 tine glioma. Acta Neuropathol. 128(4):605-7. molecular targets in pheochromocytomas and

References 361
paragangliomas. Mod Pathol. 27(8):1050-62. ependymomas of the cauda equina. A review 411. Chang S, Prados MD (1994). Identical hemangioblastomas and its correlation with
PMID:24390213 of the clinical characteristics]. Minerva Chir. twins with Ollier's disease and intracranial gli- cyst formation. J Neurooncol. 80(3):219-25.
379. Castellano-Sanchez AA, Schemankewitz 52(5):629-33. PMID:9297152 omas: case report. Neurosurgery. 34(5):903-6, PMID:17077939
E, Mazewski C, Brat DJ (2001 ). Pediatric chor- 396. Chacko G, Chacko AG, Dunham CP, Jud- discussion 906. PMID:8052390 429. Chen Z, Liu C, Patel AJ, Liao CP, Wang
doid glioma with chondroid metaplasia. Pediatr kins AR, Biegel JA, Perry A (2007). Atypical te- 412. Chang SM, Lillis-Hearne PK, Lar- Y, Le LQ (2014). Cells of origin in the embryo-
Dev Pathol. 4(6):564-7. PMID:11826363 ratoid/rhabdoid tumor arising in the setting of a son DA, Wara WM, Bollen AW, Prados MD nic nerve roots for NF1-associated plexiforrn
380. Castro-Vega LJ, Buffet A, De Cubas AA, pleomorphic xanthoastrocytoma. J Neurooncol. (1995). Pineobtastoma in adults. Neuro- neurofibroma. Cancer Cell. 26(5):695-706.
Casc6n A, Menara M, Khalifa E, et al. (2014). 84(2):217-22. PMID:17431546 surgery. 37(3):383-90, discussion 390-1. PMID:25446898
Germline mutations in FH confer predisposition 397. Chacon-Quesada T, Rodriguez GJ, Maud PMID:7501100 430. Cheng TM, Coffey RJ, Gelber BR, Scheit-
to malignant pheochromocytomas and para- A, Ramos-Duran L, Torabi A, Fitzgerald T, et al. 413. Chao L, Tao XB, Jun YK, Xia HH, Wan WK, hauer BW (1993). Simultaneous presentation
gangliomas. Hum Mal Genet. 23(9):2440-6. (2015). Trans-arterial Onyx Embolization of a Tao QS (2013). Recurrence and histological of symptomatic subependymomas in sib-
PMID:24334767 Functional Thoracic Paraganglioma. Neuroin- evolution of dysembryoplastic neuroepithelial lings: case reports and review. Neurosurgery.
381. Castro-Vega LJ, Letouze E, Burnichon N, tervention. 10(1):34-8. PMID:25763296 tumor: A case report and review of the literature. 33(1):145-50. PMID:8355833
Buffet A, Disderot PH, Khalifa E, et al. (2015). 398. Chakraborty R, Hampton OA, Shen X, Oneal Lett. 6(4):907-14. PMID:24137435 431. Chentli F, Belhimer F, Kessaci F, Man-
Multi-omics analysis defines core genomic alte- Simko SJ, Shih A, Abhyankar H, et al. (2014). 414. Chappe C, Padovani L, Scavarda D, souri B (2012). Congenital craniopharyngi-
rations in pheochromocytomas and paragangli- Mutually exclusive recurrent somatic mutations Forest F, Nanni-Metellus I, Loundou A, et al. oma: a case report and literature review. J
omas. Nat Commun. 6:6044. PMID:25625332 in MAP2K1 and BRAF support a central role for (2013). Dysembryoplastic neuroepithelial tu- Pediatr Endocrinol Metab. 25{11-12):1181-3.
382. Cataltepe 0, Turanli G, Yalnizoglu D, ERK activation in LCH pathogenesis. Blood. mors share with pleomorphic xanthoastrocyto- PMID:23329768
Topcu M, Akalan N (2005). Surgical manage- 124(19):3007-15. PMID:25202140 mas and gangliogliomas BRAF(V600E) mutati- 432. Chetty R (1999). Cytokeratin expression
ment of temporal lobe tumor-related epilepsy 399. Chalasani S, Hennick MR, Hocking WG, on and expression. Brain Pathol. 23(5):574-83. in cauda equina paragangliomas. Am J Surg
in children. J Neurosurg. 102(3) Suppl:280-7. Shaw GR, Lawler B (2013). Unusual presen- PMID:23442159 Pathol. 23(4):491. PMID:10199484
PMID:15881751 tation of a rare cancer: histiocytic sarcoma in 415. Charles NA, Holland EC, Gilbertson R, 433. Chi SN, Zimmerman MA, Yao X, Cohen KJ
383. Cavalli G, Guglielmi B, Berti A, Cam- the brain 16 years after treatment for acute lym- Glass R, Kettenmann H (2012). The brain tu- Burger P, Biegel JA, et al. (2009). Intensive mu1'.
pochiaro C, Sabbadini MG, Dagna L (2013). phoblastic leukemia. Clin Med Res. 11(1):31-5. mor microenvironment. Glia. 60(3):502-14. timodality treatment for children with newly dia-
The multifaceted clinical presentations and PMID:22997353 PMID:22379614 gnosed CNS atypical teratoid rhabdoid tumor. J
manifestations of Erdheirn-Chester disease: 400. Chamberlain M, Soffietti R, Raizer J, Ruda 416. Chaskis C, Michotte A, Goossens A, Clin Oneal. 27(3):385-9. PMID:19064966
comprehensive review of the literature and of R, Brandsma D, Boogerd W, et al. (2014). Stadnik T, Koerts G, D'Haens J (2002). Pri- 434. Chidambaram B, Santhosh V, Shankar
10 new cases. Ann Rheum Dis. 72(10):1691-5. Leptomeningeal metastasis: a Response As- mary intracerebral myxoid chondrosarcoma. SK (1998). Identical twins with medulloblasto-
PMID:23396641 sessment in Neuro-Oncology critical review Case illustration. J Neurosurg. 97(1):228. ma occurring in infancy. Childs Nerv Syst.
384. Ceccom J, Bourdeau! F, Loukh N, Rigau of endpoints and response criteria of publis- PMID:12134922 14(9):421-5. PMID:9808250
V, Milin S, Takin R, et al. (2014). Embryonal tu- hed randomized clinical trials. Neuro Oneal. 417. Chaudhry NS, Ahmad F, Blieden C, Mor- 435. Chiechi MV, Smirniotopoulos JG, Mena H
mor with multilayered rosettes: diagnostic tools 16(9): 1176-85. PMID:24867803 cos JJ (2013). Suprasellar and sellar paragan- (1995). Pineal parenchymal tumors: CT and MR
update and review of the literature. Clin Neuro- 401. Chan AK, Pang JC, Chung NY, Li KK, glioma presenting as a nonfunctioning pituitary features. J Comput Assist Tomogr. 19(4):509-
pathol. 33(1 ):15-22. PMID:23863344 Poon WS, Chan OT, et al. (2014). Loss of macroadenoma. J Clin Neurosci. 20(11):1615- 17. PMID:7622675
385. Ceeraz S, Nowak EC, Noelle RJ (2013). CIC and FUBP1 expressions are potential 8. PMID:23876285 436. Chik K, Li C, Shing MM, Leung T, Yuen PM
87 family checkpoint regulators in immune markers of shorter time to recurrence in oligo- 418. Cheadle JP, Reeve MP, Sampson JR, (1999). lntracranial germ cell tumors in children
regulation and disease. Trends lmmunol. dendroglial tumors. Mod Pathol. 27(3):332-42. Kwiatkowski DJ (2000). Molecular genetic with and without Down syndrome. J Pediatr
34(11):556-63. PMID:23954143 PMID:240307 48 advances in tuberous sclerosis. Hum Genet. Hematol Oneal. 21(2):149-51. PMID:10206462
387. Cenacchi G, Giangaspero F (2004). Emer- 402. Chan AS, Leung SY, Wong MP, Yuen ST, 107(2):97-114. PMID:11030407 437. Chikai K, OhnishiA, Kato T, Ikeda J, Sawa-
ging tumor entities and variants of CNS neo- Cheung N, Fan YW, etal. (1998). Expression of 419. Chelliah D, Mensah Sarfo-Poku C, Stea mura Y, Iwasaki Y, et al. (2004). Clinico-patholo-
plasms. J Neuropathol Exp Neurol. 63(3):185- vascular endothelial growth factor and its recep- BO, Gardetto J, Zumwalt J (2010). Medullob- gical features of pilomyxoid astrocytoma of the
92. PMID:15055442 tors in the anaplastic progression of astrocyto- lastoma with extensive nodularity undergoing optic pathway. Acta Neuropathol. 108(2):109-
388. Cenacchi G, Giangaspero F, Cerasoli S, ma, oligodendroglioma, and ependymoma. Am post-therapeutic maturation to a gangliocytoma: 14. PMID:15168135
Manetta V, Martinelli GN (1996). Ultrastructural J Surg Pathol. 22(7):816-26. PMID:9669344 a case report and literature review. Pediatr Neu- 438. Chikwava K, Jaffe R (2004). Langerin
characterization of oligodendroglial-like cells in 403. Chan CC, Koch CA, Kaiser-Kupfer Ml, rosurg. 46(5):381-4. PMID:21389751 (CD207) staining in normal pediatric tissues,
central nervous system tumors. Ultrastruct Pa- Parry DM, Gutmann DH, Zhuang Z, et al. 420. Chen BJ, Marino-Enriquez A, Fletcher reactive lymph nodes, and childhood histiocytic
thol. 20(6):537-47. PMID:8940761 (2002). Loss of heterozygosity for the NF2 gene CD, Hornick JL (2012). Loss of retinoblastoma disorders. Pediatr Dev Pathol. 7(6):607-14.
389. Cenacchi G, Roncaroli F, Cerasoli S, Fi- in retinal and optic nerve lesions of patients with protein expression in spindle cell/pleomorphic PMID:15630529
carra G, Merli GA, Giangaspero F (2001). Chor- neurofibromatosis 2. J Pathol. 198(1):14-20. lipomas and cytogenetically related tumors: 439. Chino! OL, Wick W, Mason W, Henriksson
doid glioma of the third ventricle: an ultrastruc- PMID:12210058 an immunohistochemical study with diagnostic R, Saran F, Nishikawa R, et al. (2014). Beva-
tural study of three cases with a histogenetic 404. Chan CH, Bittar RG, Davis GA, Kalnins implications. Am J Surg Pathol. 36(8):1119-28. cizumab plus radiotherapy-temozolomide for
hypothesis. Am J Surg Pathol. 25(3):401-5. RM, Fabinyi GC (2006). Long-term seizure PMID:22790852 newly diagnosed glioblastoma. N Engl J Med.
PMID:11224612 outcome following surgery for dysembryoplastic 421. Chen CJ, Williams EA, McAneney TE, 370(8):709-22. PMID:24552318
390. Cepeda C, Andre VM, Yamazaki I, Haupt- neuroepithelial tumor. J Neurosurg. 104(1):62- Williams BJ, Mandell JW, Shaffrey ME (2015). 440. Chintagumpala M, Hassall T, Palmer S, As-
man JS, Chen JY, Vinters HV, et al. (2010). 9. PMID:16509148 Histiocytic sarcoma of the cavernous sinus: hley D, Wallace D, Kasow K, et al. (2009). A pi-
Comparative study of cellular and synaptic 405. Chan GL, Little JB (1983). Cultured diploid case report and literature review. Brain Tumor lot study of risk-adapted radiotherapy and che-
abnormalities in brain tissue samples from pe- fibroblasts from patients with the nevoid basal Pathol. 32(1):66-71. PMID:24807104 motherapy in patients with supratentorial PNET.
diatric tuberous sclerosis complex and cortical cell carcinoma syndrome are hypersensitive 422. Chen G, Luo Z, Liu T, Yang H (2011). Fun- Neuro Oneal. 11(1):33-40. PMID:18796696
dysplasia type II. Epilepsia. 51 Suppl 3:160-5. to killing by ionizing radiation. Am J Pathol. ctioning paraganglioma of lhe cervical spine. 441. Chirindel A, Chaudhry M, Blakeley JO,
PMID:20618424 111(1):50-5. PMID:6837723 Orthopedics. 34{10):e700-2. PMID:21956072 Wahl R (2015). 18F-FDG PET/CT qualitative
391. Cerami E, Gao J, Dogrusoz U, Gross 406. Chan JA, Zhang H, Roberts PS, Jozwiak 423. Chen J, McKay RM, Parada LF (2012). and quantitative evaluation in neurofibromat-
BE, Sumer SO, Aksoy BA, et al. (2012). The S, Wieslawa G, Lewin-Kowalik J, et al. (2004). Malignant glioma: lessons from genomics, osis type 1 patients for detection of malignant
cBio cancer genomics portal: an open plat- Pathogenesis of tuberous sclerosis subependy- mouse models, and stem cells. Cell. 149(1):36- transformation: comparison of early to de-
form for exploring multidimensional cancer mal giant cell astrocytomas: biallelic inactivation 47. PMID:22464322 layed imaging with and without liver activity
genomics data. Cancer Discov. 2(5):401-4. of TSC1 or TSC2 leads to mTOR·activation. J 424. Chen L, Li Y, Lin JH (2005). lntraneural normalization. J Nucl Med. 56(3):379-85.
PMID:22588877 Neuropathol Exp Neural. 63(12):1236-42. perineurioma in a child with Beckwith-Wiede- PMID:25655626
392. Cerase A, Vallone IM, Di Pietro G, Oliveri PMID:15624760 mann syndrome. J Pediatr Surg. 40{2):E12-4. 442. Chitoku S, Kawai S, Watabe Y, Nishitani
G, Miracco C, Venturi C (2009). Neuroradiologi- 407. Chandler JP, Yashar P, Laskin WB, Russell PMID:15750909 M, Fujimoto K, Otsuka H, et al. (1998). lntra-
cal follow-up of the growth of papillary tumor of EJ (2004). lntracranial chondrosarcoma: a case 425. Chen L, Voronovich Z, Clark K, Hands I, dural spinal hibernoma: case report. Surg
the pineal region: a case report. J Neurooncol. report and review of the literature. J Neuroon- Mannas J, Walsh M, et al. (2014). Predicting Neurol. 49(5):509-12, discussion 512-3.
95(3):433-5. PMID:19517065 col. 68(1):33-9. PMID:15174519 the likelihood of an isocitrate dehydrogenase 1 PMID:9586928
393. Cerda-Nicolas M, Lopez-Gines C, Gil-Ben- 408. Chang AH, Fuller GN, Debnam JM, or 2 mutation in diagnoses of infiltrative glioma. 443. Chittaranjan S, Chan S, Yang C, Yang KC,
se R, Donat J, Fernandez-Delgado R, Pellin A, Karis JP, Coons SW, Ross JS, et al. (2008). Neuro Oneal. 16(11):1478-83. PMID:24860178 Chen V, Moradian A, et al. (2014). Mutations
et al. (2006). Desmoplastic infantile gangliogli- MR imaging of papillary tumor of the pineal 426. Chen SC, Lin OS, Lee CC, Hung SC, Chen in CIC and IDH1 cooperatively regulate 2-hy-
oma. Morphological, immunohistochemical and region. AJNR Am J Neuroradiol. 29(1):187-9. YW, Hsu SP, et al. (2013). Rhabdoid glioblasto- droxyglutarate levels and cell clonogenicity.
genetic features. Histopathology. 48(5):617-21. PMID:17925365 ma: a recently recognized subtype of glioblasto- Oncotarget. 5(17):7960-79. PMID:25277207
PMID:16623795 409. Chang MW (2003). Updated classifi- ma. Acta Neurochir (Wien). 155(8):1443-8, 444. Chmielecki J, Crago AM, Rosenberg M,
394. Cervera-Pierot P, Varlet P, Chodkiewicz cation of hemangiomas and other vascular discussion 1448. PMID:23812963 O'Connor R, Walker SR, Ambrogio L, et al.
JP, Daumas-Duport C (1997). Dysembryopla- anomalies. Lymphat Res Biol. 1(4):259-65. 427. Chen SM, Chuang CC, Toh CH, Jung SM, (2013). Whole-exome sequencing identifies
stic neuroepithelial tumors located in the cau- PMID:15624554 Lui TN (2013). Solitary intracranial osteoma a recurrent NAB2-STAT6 fusion in solitary
date nucleus area: report of four cases. Neu- 410. Chang Q, Pang JC, Li KK, Poon WS, Zhou with attachment to the falx: a case report. World fibrous tumors. Nat Genet. 45(2):131-2.
rosurgery. 40(5):1065-9, discussion 1069-70. L, Ng HK (2005). Promoter hypermethylation J Surg Oneal. 11 :221. PMID:24010982 PMID:23313954
PMID:9149266 profile of RASSF1A, FHIT, and sFRP1 in in- 428. Chen Y, Tachibana 0, Oda M, Xu R, 445. Cho BK, Wang KC, Nam DH, Kim DG,
395. Cervoni L, Celli P, Caruso R, Gagliardi tracranial primitive neuroectodermal tumors. Hamada J, Yamashita J, et al. (2006). In- Jung HW, Kim HJ, et al. (1998). Pineal tumors:
FM, Cantore GP (1997). (Neurinomas and Hum Pathol. 36(12):1265-72. PMID:16311119 creased expression of aquaporin 1 in human experience with 48 cases over 10 years. Childs

362 References
Nerv Syst. 14(1-2):53-8. PMID:9548342 463. Claes A, Idema AJ, Wesseling P (2007). of 11 cases and literature review. Mayo Clin 494. Coons SW, Johnson PC, Scheithauer BW,
446. Cho HJ, Myung JK, Kim H, Park CK, Kim Diffuse glioma growth: a guerilla war. Acta Neu- Proc. 78(5):567-73. PMID: 127 44543 Yates AJ, Pearl DK (1997). Improving diagno-
SK. Chung CK, et al. (2015). Primary diffuse ropathol. 114(5):443-58. PMID:17805551 480. Colin C, Padovani L, Cheppe C, Mercurio stic accuracy and interobserver concordance
leptomeningeal glioneuronal tumors. Brain Tu- 464. Clarenbach P, Kleihues P, Metzel E, Dich- S, Scavarda D, Loundou A, et al. (2013). Out- in the classification and grading of primary gli-
mor Pathol. 32(1 ):49-55. PMID:24770606 gans J (1979). Simultaneous clinical manifesta- come analysis of childhood pilocytic astrocyto- omas. Cancer. 79(7):1381-93. PMID:9083161
441. Cho Y, Gorina S, Jeffrey PD, Pavletich NP tion of subependymoma of the fourth ventricle mas: a retrospective study of 148 cases at a 495. Cooper ERA (1935). The relation of oli-
(1994). Crystal structure of a p53 tumor sup- in identical twins. Case report. J Neurosurg. single institution. Neuropathol Appl Neurobiol. godendrocytes and astrocytes in cerebral tu-
pressor-DNA complex: understanding lurnori- 50(5):655-9. PMID:571009 39(6):693-705. PMID:23278243 mours. J Path Bact. 41 :259-66.
genic mutations. Science. 265(5170):346-55. 465. ClarkAJ, Sughrue ME, Ivan ME.Aranda D, 481. Colin C, Virard I, Baeza N, Tchoghandjian 496. Cooper LA, Gutman DA, Long Q, John-
PMID:8023157 Rutkowski MJ, Kane AJ, et al. (2010). Factors A, Fernandez C, Bouvier C, et al. (2007). Rele- son BA, Cholleti SR, Kure T, et al. (2010). The
448. Cho Y J, Tsherniak A, Tamayo P, Santag- influencing overall survival rates for patients vance of combinatorial profiles of intermediate proneural molecular signature is enriched in
ata S, Ligon A, Greulich H, et al. (2011). Inte- with pineocytoma. J Neurooncol. 100(2):255- filaments and transcription factors for glioma oligodendrogliomas and predicts improved
grative genomic analysis of medulloblastoma 60. PMID:20461445 histogenesis. Neuropathol Appl Neurobiol. survival among diffuse gliomas. PLoS One.
identifies a molecular subgroup that drives poor 466. Clark GB, Henry JM, McKeever PE 33(4):431-9. PMID:17442061 5(9):e12548. PMID:20838435
clinical outcome. J Clin Oneal. 29(11):1424-30. (1985). Cerebral pilocytic astrocytoma. Cancer. 482. Collins VP, Jones DT, Giannini C (2015). 497. Gopie-Bergman C, Wotherspoon AC, Nor-
PMID:21098324 56(5):1128-33. PMID:4016701 Pilocytic astrocytoma: pathology, molecular ton AJ, Diss TC, Isaacson PG (1998). True his-
449. Choi C, Ganji SK, DeBerardinis RJ, Ha- 467. Clark VE, Erson-Omay EZ, Serin A, Yin mechanisms and markers. Acta Neuropathol. tiocytic lymphoma: a morphologic, immunohis-
tanpaa KJ, Rakheja D, Kovacs Z, et al. (2012). J, Cotney J, Ozduman K, et al. (2013). Geno- 129(6):775-88. PMID:25792358 tochemical, and molecular genetic study of
2-hydroxyglutarate detection by magnetic mic analysis of non-NF2 meningiomas reveals 483. Colnat-Coulbois S, Kremer S, Weinbreck 13 cases. Am J Surg Pathol. 22(11):1386-92.
resonance spectroscopy in I DH-mutated pa- mutations in TRAF7, KLF4, AKT1, and SMO. N, Pinelli C, Auque J (2008). Lipomatous PMID:9808131
tients with gliomas. Nat Med. 18(4):624-9. Science. 339(6123):1077-80. PMID:23348505 meningioma: report of 2 cases and review of 498. Cornejo KM, Hutchinson L, Cesar EF,
PMID:22281806 468. Clarke MJ, Foy AB, Wetjen N, Raffel C the literature. Surg Neural. 69(4)398-402, di- Smith T, Tomaszewicz K, Dresser K, et al.
450. Choi JS, Nam DH, Ko YH, Seo JW, Choi (2006). Imaging characteristics and growth of scussion 402. PMID:17825370 (2013). Is it a primary or metastatic melanocytic
YL, Suh YL, et al. (2003). Primary central ner- subependymal giant cell astrocytomas. Neuro- 483A. Combs SE, Schulz-Ertner D, Debus J, neoplasm of the central nervous system?:
vous system lymphoma in Korea: comparison surg Focus. 20(1):E5. PMID:16459995 van Deimling A, Hartmann C (2011). Improved A molecular based approach. Pathol Int.
of B- and T-cell lymphomas. Am J Surg Pathol. 468A. Claus EB, Black PM, Bondy ML, Calvo- correlation of the neuropathologic classification 63(11):559-64. PMID:24274719
27(7):919-28. PMID 12826884 coressi L, Schildkraut JM, Wiemels JL, et al. according to adapted world health organization 499. Corti ME, Yampolsky C, Metta H, Valerga
451. Chou SM, Anderson JS (1991). Primary (2007). Exogenous hormone use and meningi- classification and outcome after radiotherapy in M, Sevlever G, Capizzano A (2004). Oligo-
CNS malignant rhabdoid tumor (MRT): report of oma risk: what do we tell our patients? Cancer. patients with atypical and anaplastic meningio- dendroglioma in a patient with AIDS: case re-
two cases and review of literature. Clin Neuro- 110(3):471-6. PMID:17580362 mas. lntJ Radial Oneel Biol Phys. 81(5):1415- port and review of the literature. Rev Inst Med
pathol. 10(1):1-10. PMID:2015720 469. Claus EB, Calvocoressi L, Bondy ML, 21. PMID:20932661 Trap Sao Paulo. 46(4):195-7. PMID:15361970
452. Chow E, Jenkins JJ, Burger PC, Re- SchildkrautJM, WiemelsJL, Wrensch M (2011). 484. Camino-Mendez I, Gracia-Aznsrez FJ, 500. Cesar M, lplikcioglu AC, Bek S, Gokduman
ardon DA, Langston JW, Sanford RA, et al. Family and personal medical history and risk Schiavi F, Landa I, Leandro-Garcia LJ, Let6n CA (2005). lntracranial falcine and convexity
(1999). Malignant evolution of choroid plexus of meningioma. J Neurosurg. 115(6):1072-7. R, et al. (2011 ). Exome sequencing identi- chondromas: two case reports. Br J Neurosurg.
papilloma. Pediatr Neurosurg. 31(3):127-30. PMID:21780859 fies MAX mutations as a cause of hereditary 19(3):241-3. PMID:16455525
PMID:10708353 470. Claus EB, Calvocoressi L, Bondy ML, pheochromocytoma. Nat Genet. 43(7):663-7. 501. Couce ME, Aker FV, Scheithauer BW
453. Chowdhary A. Spence AM, Sales L, Rosto- Schildkraut JM, Wiemels JL, Wrensch M PMID:21685915 (2000). Chordoid meningioma: a clinicopa-
mily RC, Rockhill JK, Silbergeld DL (2012). (2012). Dental x-rays and risk of meningioma. 485. Committee of Brain Tumor Registry of Ja- thologic study of 42 cases. Am J Surg Pathol.
Radiation associated tumors following thera- Cancer. 118(18):4530-7. PMID:22492363 pan (2003). Report of Brain Tumor Registry of 24(7):899-905. PMID:10895812
peutic cranial radiation. Surg Neural Int. 3:48. 471. Claus EB, Calvocoressi L, Bondy ML, Japan (1969-1996). Neural Med Chir (Tokyo). 502. Courts C, Montesinos-Rongen M, Brunn A,
PMID:22629485 Wrensch M, Wiemels JL, Schildkraut JM (2013). 43 Suppl:i-vii, 1-111. PMID:14705327 Bug S, Siemer D, Hans V, et al. (2008). Recur-
454. Christensen BC, Smith AA, Zheng S, Exogenous hormone use, reproductive factors, 485A. Committee of Brain Tumor Registry of rent inactivation of the PRDM1 gene in primary
Koestler DC, Houseman EA, Marsit CJ, et al. and risk of intracranial meningioma in females. Japan (2014). Report of Brain Tumor Registry central nervous system lymphoma. J Neuropa-
(2011). DNA methylation, isocitrate dehydro- J Neurosurg. 118(3):649-56. PMID:23101448 of Japan (2001-2004), Vol. 13. Neural Med Chir thol Exp Neural. 67(7):720-7. PMID:18596541
genase mutation, and survival in glioma. J Natl 472. Clifford SC, Lusher ME, Lindsey JC, (Tokyo) 54:1-102. 503. Courville CB, Broussalian SL (1961). Pla-
Cancer Inst. 103(2):143-53. PMID:21163902 Langdon JA, Gilbertson RJ, Straughton D, et 486. Connor JM, Pirrit LA, Yates JR, Fryer AE, stic ependymomas of the lateral recess. Report
455. Christiaans I, Kenter SB, Brink HG, van Os al. (2006). Wnt!Wingless pathway activation Ferguson-Smith MA (1987). Linkage of the tu- of eight verified cases. J Neurosurg. 18:792-9.
TA, Baas F, van den Munckhof P, et al. (2011). and chromosome 6 loss characterize a distinct berous sclerosis locus to a DNA polymorphism PMID:13881784
Germline SMARCB1 mutation and somatic NF2 molecular sub-group of medu\loblastomas as- detected by v-abl. J Med Genet. 24(9):544-6. 504. Covington MF, Chin SS, Osborn AG (2011).
mutations in familial multiple meningiomas. J sociated with a favorable prognosis. Cell Cycle. PMID:2889832 Pituicytoma, spindle cell oncocytoma, and gra-
Med Genet. 48(2):93-7. PMID:20930055 5(22):2666-70. PMID 17172831 487. Constantini S, Soffer D, Siegel T, Shalit MN nular cell tumor: clarification and meta-analysis
456. Christov C, Adle-Biassette H, Le Guerinel 473. Clynes D, Higgs DR, Gibbons RJ (2013). (1989). Paraganglioma of the thoracic spinal of the world literature since 1893. AJNR Am J
C, Natchev S, Gherardi RK (1998). lmmunohis- The chromatin remodeller ATRX: a repeat of- cord with cerebrospinal fluid metastasis. Spine Neuroradiol. 32(11 ):2067-72. PMID:21960498
tochemical detection of vascular endothelial fender in human disease. Trends Biochem Sci. (Phila Pa 1976). 14(6):643-5. PMID:2749382 505. Grine PB, Mehta R, Vinters HV (2010).
growth factor (VEGF} in the vasculature of oli- 38(9):461-6. PMID:23916100 488. Conte D, Huh M, Goodall E, Delorme M, Pathogenesis of TSC in the brain. In: Tuberous
godendrogliomas. Neuropathol Appl Neurobiol. 474. Coakley KJ, Huston J 3rd, Scheithauer Parks RJ, Picketts DJ (2012). Loss of Atrx sen- sclerosis complex. Genes, clinical features, and
24(1):29-35. PMID:9549726 BW, Forbes G, Kelly PJ (1995). Pilocytic ast- sitizes cells to DNA damaging agents through therapeutics. Kwiatkowski DJ, Whittemore VH,
457. Chu LC, Eberhart CG, Grossman SA, Her- rocytomas: well-demarcated magnetic reso- p53-mediated death pathways. PLoS One. Thiele EA, eds. Wiley-Blackwell: Weinheim,
man JG (2006). Epigenetic silencing of multiple nance appearance despite frequent infiltration 7(12}:e52167. PMID:23284920 Germany, pp. 161-185.
genes in primary CNS lymphoma. Int J Cancer. histologically. Mayo Clin Proc. 70(8):747-51. 489. Conti P, Mouchaty H, Spacca B, Bucco- 506. Grine PB, Trojanowski JO, Dichter MA,
119(10):2487-91. PMID:16858686 PMID:7630212 liero AM, Conti R (2006). Thoracic extradural Eberwine J (1996). Embryonic neuronal mar-
458. Ciardiello F, Tortora G (2001). A novel 475. Cobbers JM, Wolter M, Reifenberger J, paragangliomas: a case report and review kers in tuberous sclerosis: single-cell mole-
approach in the treatment of cancer: targeting Ring GU, Jessen F, An HX, et al. (1998). Fre- of the literature. Spinal Cord. 44(2):120-5. cular pathology. Proc Natl Acad Sci U S A.
the epidermal growth factor receptor. Clin Can- quent inactivation of CDKN2A and rare mutati- PMID:16130022 93(24):14152-7. PMID:8943076
cer Res. 7(10):2958-70. PMID:11595683 on of TP53 in PCNSL. Brain Pathol. 8(2j:263- 490. Conway JE, Chou D, Clatterbuck RE, 506A. Crocetti E, Trama A, Stiller C, Caldarella
459. Cimino PJ, Agarwal A, Dehner LP (2014). 76. PMID:9546285 Brem H, Long DM, Rigamonti D (2001). He- A, Soffietti R, Jaal J, et al. (2012). Epidemiology
Myxopapillary ependymoma in children: a study 476. Coca S, Moreno M, Martos JA, Rodriguez mangioblastomas of the central nervous system of glial and non-glial brain tumours in Europe.
of 11 cases and a comparison with the adult ex- J, Barcena A, Vaquero J (1994). Neurocytoma in von Hippel-Lindau syndrome and sporadic di- Eur J Cancer. 48(10):1532-42. PMID:22227039
perience. Pediatr Blood Cancer. 61(11):1969- of spinal cord. Acta Neuropathol. 87(5):537-40. sease. Neurosurgery. 48(1):55-62, discussion 507. Crotty TB, Scheithauer BW, Young WF Jr,
71. PMID:25066546 PMID:8059608 62-3. PMID:11152361 Davis DH, Shaw EG, Miller GM, et al. (1995).
460. Cimino PJ, Gonzalez-Cuyar LF, Perry A, 477. Coca S, Vaquero J, Escandon J, Moreno 491. Cook DJ, Christie SD, Macaulay RJ, Papillary craniopharyngioma: a clinicopa-
Dahiya S (2015). Lack of BRAF-V600E Muta- M, Peralba J, RodriguezJ (1992). lmmunohisto- Rheaume DE, Holness RO (2004). Fourth ven- thological study of 48 cases. J Neurosurg.
tion in Papillary Tumor of the Pineal Region. chemical characterizalion of pineocytomas. Clin tricular neurocytoma: case report and review of 83(2):206-14. PMID:7616262
Neurosurgery. 77(4):621-8. PMID:26125673 Neuropathol. 11(6):298-303 PMID:1473313 lhe literature. Can J Neural Sci. 31(4):558-64. 508. Cuccia V, Rodriguez F, Palma F, Zuccaro
461. Cinalli G, Zerah M, Carteret M, Doz F, Vi- 478. Cohen BH, Zeltzer PM, Boyett JM, Geyer PMID:15595267 G (2006). Pinealoblastomas in children. Childs
nikoff L, Lellouch-Tubiana A, et al. (1997). Sub- JR, Allen JC, Finlay JL, et al. (1995). Prognostic 492. Coons SW, Johnson PC (1993). Regional Nerv Syst. 22(6):577-85. PMID:16555075
dural sarcoma associated with chronic subdural factors and treatment results for supratentorial heterogeneity in the proliferative activity of hu- 509. Curatolo P, Moavero R (2012). mTOR
hematoma. Report of two cases and review primitive neuroectodermal tumors in children man gliomas as measured by the Ki-67 labeling Inhibitors in Tuberous Sclerosis Com-
of the literature. J Neurosurg. 86(3):553-7. using radiation and chemotherapy: a Childrens index. J Neuropathol Exp Neural. 52(6):609-18. plex. Curr Neurophanmacol. 10(4):404-15.
PMID:9046316 Cancer Group randomized trial. J Clin Oneel. PMID:8229080 PMID:23730262
462. Ciriello G, Miller ML, Aksoy BA, Senba- 13(7):1687-96. PMID:7602359 493. Coons SW, Johnson PC, Pearl DK 510. Curless RG, Toledano SR, Ragheb J, Cle-
baoglu Y, Schultz N, Sander C (2013). Emer- 479. Cohen-Gadol AA, Pichelmann MA, Link (1997). The prognostic significance of Ki-67 veland WW, Falcone S (2002). Hematogenous
ging landscape of oncogenic signatures across MJ, Scheithauer BW, Krecke KN, Young WF Jr, labeling indices for oligodendrogliomas. Neu- brain metastasis in children. Pediatr Neural.
human cancers. Nat Genet. 45(10):1127-33. et al. (2003). Granular cell tumor of the sellar rosurgery. 41 (4):878-84, discussion 884-5. 26(3):219-21. PMID:11955930
PMID:24071851 and suprasellar region: clinicopathologic study PMID:9316050 511. Curran EK, Sainani KL, Le GM, Propp

References 363
JM, Fisher PG (2009). Gender affects survival 114(4)681-8. PMID:15064624 humans and mice. Neuro Oncol. 12(3):233-42. Zakrzewski K, Liberski PP (1999). Cytogenetic
for medulloblastoma only in older children and 527. Darwish B, Arbuckle S, Kellie S, Besser PMID:20167811 changes in two cases of subependymal ·gi-
adults: a study from the Surveillance Epidemio- M, Chaseling R (2007). Desmoplastic infantile 543. De Jesus 0, Rifkinson N (1995). Pleomor- ant-cell astrocytoma. Cancer Genet Cytogenet.
logy and End Results Registry. Pediatr Blood ganglioglioma/astrocytoma with cerebrospinal phic xanthoastrocytoma with invasion of the 109(1):29-33. PMID:9973956
Cancer. 52(1):60-4. PMID:19006250 metastasis. J Clin Neurosci. 14(5):498-501. tentorium and falx. Surg Neurol. 43(1):77-9. 561. Deckert M, Brunn A, Montesinos-Rongen
511A. Custer B, Longstreth WT, Phillips LE, PMID:17386372 PMID 7701430 M, Terreni MR, Ponzoni M (2014). Primary lym.
Koepsell TD, Van Belle G (2006). Hormonal ex- 528. Darwish BS, Balakrishnan V, Maitra R 544. de Jong MC, Kors WA, de Graaf P, phoma of the central nervous system-a diag.
posures and the risk of intracranial meningioma (2002). lntramedullary ancient schwannoma Castelijns JA, Kivela T, Moll AC (2014). Trila- nostic challenge. Hematol Oncol. 32(2):57-67.
in women: a population-based case-control stu- of the cervical spinal cord: case report and re- teral retinoblastoma: a systematic review and PMID:23949943
dy. BMC Cancer. 6:152. PMID:16759391 view of literature. J Clin Neurosci. 9(3):321-3. meta-analysis. Lancet Oncol. 15(10):1157-67. 562. Deckert M, Engert A, Bruck W, Ferreri AJ
512. Cykowski MD, Wartchow EP, Mierau GW, PMID 12093146 PMID:25126964 Finke J, lllerhaus G, et al. (2011). Modern con'.
Stolzenberg ED, Gumerlock MK, Fung KM 529. Dasgupta B, Dugan LL, Gutmann DH 545. de Kock L, Sabbaghian N, Druker H, We- cepts in the biology, diagnosis, differential diag.'Y ·
(2012). Papillary tumor of the pineal region: ul- (2003). The neurofibromatosis 1 gene product ber E, Hamel N, Miller S, et al. (2014). Germ- nosis and treatment of primary central nervous
trastructural study of a case. Ultrastruct Pathol. neurofibromin regulates pituitary adenylate line and somatic DICER1 mutations in pineob- system lymphoma. Leukemia. 25(12):1797-
36(1 ):68-77. PMID:22292738 cyclase-activating polypeptide-mediated signa- lastoma. Acta Neuropathol. 128(4):583-95. 807. PMID:21818113
513. D'Ambrosio N, Soohoo S, Warshall C, ling in astrocytes. J Neurosci. 23(26):8949-54. PMID:25022261 563. DeDavid M, Orlow SJ, Provost N, Mar-
Johnson A, Karimi S (2008). Craniofacial and PMID:14523097 546. de la Monte SM, Horowitz SA (1989). He· ghoob AA, Rao BK, Huang CL, et al. (1997). A
intracranial manifestations of langerhans cell 530. Daumas Duport C (1995). Patterns of tu- mangioblastomas: clinical and histopathological study of large congenital melanocytic nevi and
histiocytosis: report of findings in 100 pati- mor growth and problems associated with histo- factors correlated with recurrence. Neurosur- associated malignant melanomas: review of ca- .
ents. AJR Am J Roentgenol. 191 (2):589-97. logical typing of low-grade gliomas. In: Apuzzo gery. 25(5):695-8. PMID2586723 ses in the New York University Registry and th;
PMID18647937 LJ, editor. Benign Cerebral Gliomas. Park Rid- 547. De Munnynck K, Van Gool S, Van Calen- world literature. J Am A cad Dermatol. 36(3 Pt •.
515. da Cruz Perez DE, Arnanaias de Aguiar FC ge: AANS; pp. 125-47. bergh F, Demaerel P, Uyttebroeck A, Buyse G, 1):409-16. PMID:9091472 .
Jr, Leon JE, Graner E, Paes de Almeida 0, Var- 531. Daumas-Duport C (1993). Dysembryo- et al. (2002). Desmoplastic infantile ganglioglio· 564. DeDavid M, Orlow SJ, Provost N, Mar-
gas PA (2006). lntraneural perineurioma of the plastic neuroepithelial tumours. Brain Pathol. ma: a potentially malignant tumor? Am J Surg ghoob AA, Rao BK, Wasti 0, et al. (1996).
tongue: a case report. J Oral Maxillofac Surg. 3(3):283-95. PMID:8293188 Pathol. 26(11):1515-22. PMID:12409729 Neurocutaneous melanosis: clinical features
64(7):1140-2. PMID:16781350 532. Daumas-Duport C (1995). Dysembryopla- 548. De Raedt T, Beert E, Pasmant E, Luscan of large congenital melanocytic nevi in pa:
516. Dabora SL, Jozwiak S, Franz ON, Roberts stic neuroepithelial tumours in epilepsy surgery. A, Brems H, Ortonne N, et al. (2014). PRC2 tients with manifest central nervous system
PS, Nieto A, Chung J, et al. (2001). Mutational In: Guerrini R, editor. Dysplasia of Cerebral loss amplifies Ras-driven transcription and melanosis. J Am Acad Dermatol. 35(4):529-38.
analysis in a cohort of 224 tuberous sclero- Cortex and Epilepsy. New York: Raven Press; confers sensitivity to BRD4-based therapies. PMID:8859278
sis patients indicates increased severity of pp. 125-47. Nature. 514(7521):247-51. PMID:25119042 565. Dedeurwaerdere F, Giannini C, Sciot R
TSC2, compared with TSC1, disease in mul- 533. Daumas-Duport C, Scheithauer B, O'Fal- 549. De Raedt T, Brems H, Lopez-Correa C, Rubin BP, Perilongo G, Borghi L, et al. (2002)'.
tiple organs. Am J Hum Genet. 68(1):64-80. lon J, Kelly P (1988). Grading of astrocytomas. Vermeesch JR, Marynen P, Legius E (2004). Primary peripheral PNET /Ewing's sarcoma
PMID:11112665 A simple and reproducible method. Cancer. Genomic organization and evolution of the NF1 of the dura: a clinicopathologic entity distinct
517. Dahia PL, Aguiar RC, Alberta J, Kum JB, 62(10):2152-65. PMID:3179928 microdeletion region Genomics. 84(2):346-60. from central PNET. Mod Pathol. 15(6):673-8. ·
Caron S, Sill H, et al. (1999). PTEN is inversely 534. Daumas-Duport C, Scheithauer BW, Cho- PMID:15233998 PMID:12065782 ..
correlated with the cell survival factor AkUPKB dkiewicz JP, Laws ER Jr, Vedrenne C (1988). 550. De Raedt T, Brems H, Wolkenstein P, 566. Degen R, Ebner A, Lahl R, Leonhardt
and is inactivated via multiple mechanismsin Dysembryoplastic neuroepithelial tumor: a Vidaud D, Pilotti S, Perrone F, et al. (2003). S, Pannek HW, Tuxhorn I (2002). Various fin.
haematological malignancies. Hum Mol Genet. surgically curable tumor of young patients Elevated risk for MPNST in NF1 microdeletion dings in surgically treated epilepsy patients
8(2):185-93. PMID:9931326 with intractable partial seizures. Report of thir- patients. Am J Hum Genet. 72(5):1288-92. with dysembryoplastic neuroepithelial tumors
518. Dahiya S, Haydon DH, Alvarado D, Gur- ty-nine cases. Neurosurgery. 23(5):545-56. PMID:12660952 in comparison with those of patients wilh other
nett CA, Gutmann DH, Leonard JR (2013). PMID:3143922 551. de Ribaupierre S, Dorfmuller G, Bulteau C, low-grade brain tumors and other neuronal
BRAF(V600E) mutation is a negative prognosti- 535. Daumas-Duport C, Scheithauer BW, Kelly Fohlen M, Pinard JM, Chiron C, et al. (2007). migration disorders. Epilepsia. 43(11):1379-84.
cator in pediatric ganglioglioma. Acta Neuropa- PJ (1987). A histologic and cytologic method for Subependymal giant-cell astrocytomas in pedi- PMID12423388
thol. 125(6):901-10. PMID:23609006 the spatial definition of gliomas. Mayo Clin Proc. atric tuberous sclerosis disease: when should 567. Dehghani F, Schachenmayr W, Laun
519. Dahiya S, Sarkar C, Hedley-Whyte ET, 62(6):435-49. PMID:2437 411 we operate? Neurosurgery. 60(1):83-9, discus- A, Korf HW (1998). Prognostic implication of .
Sharma MC, Zervas NT, Sridhar E, et al. (2005). 536. Daumas-Duport C, Varlet P (2003). sion 89-90. PMID:17228255 histopathological, immunohistochemical and 't_-
Spindle cell oncocytoma of the adenohypophy- IDysembryoplastic neuroepithelial tumors]. 552. De Schepper S, Maertens 0, Callens T, clinical features of oligodendrogliomas: a study •
sis: report of two cases. Acta Neuropathol. Rev Neurol (Paris). 159(6-7 Pt 1):622-36. Naeyaert JM, Lambert J, Messiaen L (2008). of 89 cases. Acta Neuropathol. 95(5):493-504. :-
110(1):97-9. PMID:15973544 PMID:12910070 Somatic mutation analysis in NF1 cafe au PMID:9600596
520. Dahlback HS, Gorunova L, Micci F, Scheie 537. Daumas-Duport C, Varlet P, Bacha S, lait spots reveals two NF1 hits in the me- 568. Deisch JK, Patel R, Koral K, Cope- Yo·
D, Brandal P, Meling TR, et al. (2011). Molecu- Beuvon F, Cervera-Pierot P, Chodkiewicz JP lanocytes. J Invest Dermatol. 128(4):1050-3. koyama SD (2013). Juvenile xanthogranulomasc·
lar cytogenetic analysis of a gliosarcoma with (1999). Dysembryoplastic neuroepithelial tu- PMID:17914445 of the nervous system: A report of two cases
osseous metaplasia. Cytogenet Genome Res. mors: nonspecific histological forms - a study 553. De Tommasi A, l.uzzi S, D'Urso Pl, De and review of the literature. Neuropathology. ·
134(2):88-95. PMID:21555877 of 40 cases. J Neurooncol. 41 (3):267-80. Tommasi C, Resta N, Ciappetta P (2008). 33(1 ):39-46. PMID:22640164
521. Dai C, Celestino JC, Okada Y, Louis ON, PMID:10359147 Molecular genetic analysis in a case of gan- 569. Del Bigio MR, Deck JH (1993). Rosenthal
Fuller GN, Holland EC (2001). PDGF autocrine 538. de Chadarevian JP, Montes JL, O'Gorman glioglioma: identification of a new mutation. fibers producing a granular cell appearance in
stimulation dedifferentiates cultured astrocytes AM, Freeman CR (1987). Maturation of cere- Neurosurgery. 63(5):976-80, discussion 980. a glioblastoma. Acta Neuropathol. 86(1):100-4.
and induces oligodendrogliomas and oligoast- bellar neuroblastoma into ganglioneuroma with PMID:19005389 PMID:8396834
rocytomas from neural progenitors and ast- melanosis. A histologic, immunocytochemical, 554. De Vos M, Hayward BE, Charlton R, Taylor 570. Del Valle L, Enam S, Lara C, Ortiz-Hidalgo
rocytes in vivo. Genes Dev. 15(15):1913-25. and ultrastructural study. Cancer. 59(1):69-76. GR, Glaser AW, Picton S, et al. (2006). PMS2 C, Katsetos CD, Khalili K (2002). Detection of
PMID:11485986 PMID:3539310 mutations in childhood cancer. J Natl Cancer JC polyomavirus DNA sequences and cellu·
522. Damodaran 0, Robbins P, Knuckey N, 539. de Chadarevian JP, Pattisapu JV, Faerber Inst. 98(5):358-61. PMID:16507833 lar localization of T-antigen and aqnoprolein-
Bynevelt M, Wong G, Lee G (2014). Primary EN (1990). Desmoplastic cerebral astrocytoma 555. de Vries J, Scheremet R, Altmannsber- in oligodendrogliomas. Clin Cancer Res.
intracranial haemangiopericytoma: comparison of infancy. Light microscopy, immunocytoche- ger M, Michilli R, Lindemann A, Hinkelbein W 8(11):3332-40. PMID:12429619
of survival outcomes and metastatic potential in mistry, and ultrastructure. Cancer. 66(1):173-9. (1994). Primary leiomyosarcoma of the spinal 571. Delnatte C, Sanlaville D, Mougenot JF, ·
WHO grade II and Ill variants. J Clin Neurosci. PMID:2354404 leptomeninges. J Neurooncol. 18(1):25-31. Vermeesch JR, Houdayer C, Blois MC, et al.
21(8):1310-4. PMID:24726230 540. de Cubas AA, Leandro-Garcia LJ, Schi- PMID:8057131 (2006). Contiguous gene deletion within cbro-
523. Dang L, White OW, Gross S, Bennett avi F, Mancikova V, Cornino-Mendez I, lngla- 556. de Vries NA, Hulsman D, Akhtar W, de mosome arm 1 Oq is associated with juvenile .
BO, Bittinger MA, Driggers EM, et al. (2009). da-Perez L, et al. (2013). Integrative analysis Jong J, Miles DC, Blom M, et al. (2015). Prolon- polyposis of infancy, reflecting cooperation bet· ·
Cancer-associated IDH1 mutations produce of miRNA and mRNA expression profiles in ged Ezh2 Depletion in Glioblastoma Causes a ween the BMPR1A and PTEN tumor-suppres-
2-hydroxyglutarate. Nature. 462(727 4 ): 739-44. pheochromocytoma and paraganglioma identi- Robust Switch in Cell Fate Resulting in Tumor sor genes. Am J Hum Genet. 78(6):1066-74. ··
PMID:19935646 fies genotype-specific markers and potentially Progression. Cell Rep. PMID:25600873 PMID:16685657
524. Dardick I, Hammar SP, Scheithauer BW regulated pathways. Endocr Reial Cancer. 557. DeAngelis LM (2001). Brain tumors. N 572. Demetriades AK, Al Hyassat S, AI-Sarraj
(1989). Ultrastructural spectrum of hemangi- 20(4):477-93. PMID:23660872 Engl J Med. 344(2):114-23. PMID:11150363 S, Bhangoo RS, Ashkan K (2013). Papillary
opericytoma: a comparative study of fetal, adult, 541. De Filippi P, Badulli C, Cuccia M, De 558. Dearnaley DP, A'Hern RP, Whittaker S, glioneuronal tumour: a review of the literature
and neoplastic pericytes. Ultrastruct Pathol. Silvestri A, Dametto E, Pasi A, et al. (2006). Bloom HJ (1990). Pineal and CNS germ cell with two illustrative cases. Br J Neurosurg.
13(2-3):111-54 PMID:2734855 Specific polymorphisms of cytokine genes tumors: Royal Marsden Hospital experience 27(3):401-4. PMID:23173837
525. Dario A, Cerati M, Taborelli M, Finzi G, are associated with different risks to develop 1962-1987. Int J Radial Oncol Biol Phys. 573. Demuth T, Berens ME (2004). Mole· .
Pozzi M, Dorizzi A (2000). Cytogenetic and ul- single-system or multi-system childhood Lan- 18(4):773-81. PMID:2323968 cular mechanisms of glioma cell migration
trastructural study of a pineocytoma case report. gerhans cell histiocytosis. Br J Haematol. 559. Deb P, Sharma MC, Gaikwad S, Gupta A, and invasion. J Neurooncol. 70(2):217-28.
J Neurooncol. 48(2):131-4. PMID:11083076 132(6):784-7. PMID:16487180 Mehta VS, Sarkar C (2005). Cerebellopontine PMID:15674479
526. Darrouzet V, Martel J, Enee V, Bebear JP, 542. de Groot JF, Fuller G, Kumar AJ, Piao Y, angle paraganglioma - report of a case and 57 4. Derlin T, Tornquist K, Munster S, Apo-
Guerin J (2004). Vestibular schwannoma sur- Eterovic K, Ji Y, et al. (2010). Tumor invasion review of literature. J Neurooncol. 74(1):65-9. stolova I, Hagel C, Friedrich RE, et al. (2013).
gery outcomes: our multidisciplinary experien- after treatment of glioblastoma with bevacizu- PMID:16078110 Comparative effectiveness of 18F-FDG PET/ ..
ce in 400 cases over 17 years. Laryngoscope. mab: radiographic and pathologic correlation in 560. Debiec-Rychter M, Jesionek-Kupnicka D, CT versus whole-body MRI for detection of ··

364 References
malignant peripheral nerve sheath tumors Pathol. 22(6):869-70. PMID:2305087 4 611. Dunham C, Suga E, Tobias V, Wills E, PMID:2009534
in neurofibromatosis type 1. Clin Nucl Med. 593. Dobbins SE, Broderick P, Melin B, Feych- Perry A (2007). Embryonal tumor with abundant 627. Ekstrand AJ, Sugawa N, James CD,
38(1):e19-25. PMID:23242059 ting M, Johansen C, Andersson U, et al. (2011). neuropil and true rosettes (ETANTR): report of a Collins VP (1992). Amplified and rearran-
575. Oeshmukh H, Yu J, Shaik J, MacDonald Common variation at 10p12.31 near MLLT10 case with prominent neurocytic differentiation. J ged epidermal growth factor receptor genes
TJ, Perry A, Payton JE, et al. (2011). Identifi- influences rneningiorna risk. Nat Genet. Neurooncol. 84(1):91-8. PMID:17332950 in human glioblastornas reveal deletions of
cation of transcriptional regulatory networks 43(9):825-7. PMID:21804547 612. Dunham C, Yip S (2013). Cribriforrn neu- sequences encoding portions of the N- and/
specific to pilocytic astrocytoma. BMC Med 594. Doglioni C, Dell'Orto P, Coggi G, luzzo- roepithelial tumor or atypical teratoid/rhabdoid or C-terminal tails. Proc Natl Acad Sci U S A.
Genomics. 4:57. PMID:21745356 lino P, Bonternpini L, Viale G (1987). Choroid tumor? J Neurosurg Pediatr. 11(4):486-8. 89(10):4309-13. PMID:1584765
576. Desouza RM, Bodi I, Thomas N, Marsh H, plexus tumors. An irnrnunocytochernical study PMID:23394355 628. Elefteriou F, Kolanczyk M, Schindeler
Crocker M (2010). Chordoid glioma: ten years with particular reference to the coexpression 613. Duo D, Gasverde S, Benech F, Zenga F, A, Viskochil DH, Hock JM, Schorry EK, et al.
of a low-grade tumor with high morbidity. Skull of intermediate filament proteins. Arn J Pathol. Giordana MT (2003). MIB-1 immunoreactivity (2009). Skeletal abnormalities in neurofibro-
Base 20(2):12!>--38. PMID:20808539 127(3):519-29. PMID:2438940 in craniopharyngiornas: a clinico-pathological rnatosis type 1 : approaches to therapeutic op-
577. Dessauvagie BF, Wong G, Robbins PD 595. Dohrmann GJ, Farwell JR, Flannery JT analysis. Clin Neuropathol. 22(5):229-34. tions. Am J Med Genet A. 149A(10):2327-38.
(2015). Renal cell carcinoma to haemangiob- (1976). Glioblastorna multiforme in children. J PMID:14531547 PMID:19764036
lastoma metastasis: a rare manifestation of Neurosurg. 44(4):442-8. PMID:176331 614. Duma CA, Aronson M, Tabori U, Malkin 629. Ellezarn B, Theeler BJ, Luthra R, Adesina
Von Hippel-Lindau syndrome. J Clin Neurosci. 596. Dolecek TA, Propp JM, Stroup NE, Kruch- 0, Gallinger S, Chan HS (2012). Oncologic AM, Aldape KO, Gilbert MR (2012). Recurrent
22(1):215-8. PMID:25088480 ko C (2012). CBTRUS statistical report: primary surveillance for subjects with biallelic mismatch PIK3CA mutations in rosette-forming glioneu-
578. Dewan R, Pemov A, Kim HJ, Morgan brain and central nervous system tumors diag- repair gene mutations: 10 year follow-up of a ronal tumor. Acta Neuropathol. 123(2):285-7.
KL, Vasquez RA, Chittiboina P, et al. (2015). nosed in the United States in 200!>---2009. Neu- kindred. Pediatr Blood Cancer. 59(4):652-6. PMID:21997360
Evidence of polyclonality in neurofibromatosis ro Oncol. 14 Suppl 5:v1-49. PMID:23095881 PMID:22180144 630. Ellison OW (2010). Childhood rnedullob-
type 2-associated multilobulated vestibular 597. Donoho D, Zada G (2015). Imaging of 615. Durno CA, Sherman PM, Aronson M, Mal- lastorna: novel approaches to the classification
schwannomas. Neuro Oneal. 17(4):566-73. central neurocytornas. Neurosurg Clin N Arn. kin D, Hawkins C, Sakry D, et al.; International of a heterogeneous disease. Acta Neuropathol.
PMID:25452392 26(1):11-9. PMID:25432179 BMMRD Consortium (2015). Phenotypic and 120(3):30!>--16. PMID:20652577
580. Dhimes P, Martinez-Gonzalez MA, Cara- 598. Donovan MJ, Yunis EJ, DeGirolarni U, genotypic characterisation of biallelic mismatch 631. Ellison OW, Dalton J, Kocak M, Nichol-
bias E, Perez-Espejo G (1996). Ultrastructural Fletcher JA, Schofield DE (1994). Chromoso- repair deficiency (BMMR-0) syndrome. Eur J son SL, Fraga C, Neale G, et al. (2011 ). Me-
study of a perineurioma with ribosome-lamella me aberrations in choroid plexus papillornas. Cancer. 51(8):977-83. PMID:25883011 dulloblastoma: clinicopathological correlates
complexes. Ultrastruct Pathol. 20(2):167-72. Genes Chromosomes Cancer. 11(4):267-70. 616. Eaton KW, Tooke LS, Wainwright LM, of SHH, WNT, and non-SHH/WNT molecular
PMID:8882362 PMID:7533531 Judkins AR, Biegel JA (2011). Spectrum of subgroups. Acta Neuropathol. 121(3):381-96.
581. Di Cristofano A, Pesce B, Cordon-Cardo 599. Donson AM, Kleinschrnidt-DeMasters BK, SMARCB1/INl1 mutations in familial and spo- PMID:21267586
C, Pandolfi PP (1998). Pten is essential for em- Aisner DL, Bemis LT, Birks DK, Levy JM, et radic rhabdoid tumors. Pediatr Blood Cancer. 632. Ellison OW, Kocak M, Dalton J, Megahed
bryonic development and tumour suppression. al. (2014). Pediatric brainstem gangliogliornas 56(1):7-15. PMID:21108436 H, Lusher ME, Ryan SL, et al. (2011). Definition
Nat Genet. 19(4):348-55. PMID:9697695 show BRAF(V600E) mutation in a high percen- 617. Eberhart CG (2007). In search of the rne- of disease-risk stratification groups in childhood
582. Di Rocco F, Carroll RS, Zhang J, Black tage of cases. Brain Pathol. 24(2):173-83. dulloblast: neural stern cells and embryonal medulloblastorna using combined clinical, pa-
PM (1998). Platelet-derived growth factor PMID:24238153 brain tumors. Neurosurg Clin N Am 18: 59-69, thologic, and molecular variables. J Clin Oneal.
and its receptor expression in human oligo- 600. Doskaliyev A, Yamasaki F, Kenjo M, Sh- viii-ix. PMID:17244554 29(11):1400--7. PMID:20921458
dendrogliomas. Neurosurgery. 42(2):341-6. restha P, Saito T, Hanaya R, et al. (2008). 618. Eberhart CG, Brat DJ, Cohen KJ, Bur- 633. Ellison OW, Kocak M, Figarella-Branger D,
PMID:9482185 Secondary anaplastic oligodendrogliorna after ger PC (2000). Pediatric neuroblastic brain Felice G, Catherine G, Pietsch T, et al. (2011).
583. Di Rocco F, Sabatino G, Koutzoglou M, cranial irradiation: a case report. J Neurooncol. tumors containing abundant neuropil and true Histopathological grading of pediatric ependy-
Battaglia D, Caldarelli M, Tamburrini G (2004). 88(3):299-303. PMID:18373067 rosettes. Pedialr Dev Pathol. 3(4):346-52. rnorna: reproducibility and clinical relevance in
Neurocutaneous melanosis. Childs Nerv Syst. 601. Dougherty MJ, Santi M, Brose MS, Ma C, PMID:10890250 European trial cohorts. J Negat Results Bio-
20(1):23--8. PMID 14576958 Resnick AC, Sievert AJ, et al. (2010). Activating 619. Eberhart CG, Kepner JL, Goldthwaite PT, med. 10:7. PMID:21627842
584. Dias-Santagata D, Lam Q, Vernovsky mutations in BRAF characterize a spectrum Kun LE, Duffner PK, Friedman HS, et al. (2002). 634. Ellison OW, Onilude OE, Lindsey JC, Lus-
K. Vena N, Lennerz JK, Borger DR, et al. of pediatric low-grade gliornas. Neuro Oneal. Histopathologic grading of rnedulloblastornas: her ME, Weston CL, Taylor RE, et al.; United
(2011 ). BRAF V600E mutations are common 12(7):621-30. PMID:20156809 a Pediatric Oncology Group study. Cancer. Kingdom Children's Cancer Study Group Brain
in pleomorphic xanthoastrocytoma: diagno- 602. Douglas-Akinwande AC, Payner TD, 94(2):552-60. PMID:11900240 Tumour Committee (2005). beta-Catenin status
stic and therapeutic implications. PLoS One. Hattab EM (2009). Medulloblastoma mimi- 620. Eberhart CG, Kratz J, Wang Y, Summers predicts a favorable outcome in childhood me-
6(3):e17948. PMID:21479234 cking Lhermitte-Duclos disease on MRI and K, Stearns D, Cohen K, et al. (2004). Histopa- dulloblastorna: the United Kingdom Children's
585. Diaz-Flores L, Alvarez-Arguelles H, Madrid CT. Clin Neural Neurosurg. 111(6):536-9. thological and molecular prognostic markers Cancer Study Group Brain Tumour Committee.
JF, Varela H, Gonzalez MP, Gutierrez R (1997). PMID:19233547 in rnedulloblastorna: c-myc, N-rnyc, TrkC, J Clin Oneal. 23(31):7951-7. PMID:16258095
Perineurial cell tumor (perineurioma) with 603. Oraqojevic S, Mehraein P, Bock HJ (1973). and anaplasia. J Neuropathol Exp Neural. 635. Ellison OW, Zygrnunt SC, Weller RO
granular cells. J Cutan Pathol. 24(9):57!>--9. [Lipoma of the temporal region with cortical 63(5):441-9. PMID:15198123 (1993). Neurocytorna/liporna (neurolipocytorna)
PMID:9404856 malformation and epilepsy. Clinicopathological 621. Ebert C, von Haken M, Meyer-Puttlitz of the cerebellum. Neuropathol Appl Neurobiol.
586. Dim DC, Lingarnfelter DC, Taboada EM, study (author's transl)). Arch Psychiatr Nervenkr B, Wiestler OD, Reifenberger G, Pietsch T, 19(1):9!>--8. PMID:8474606
Fiorella RM (2006). Papillary glioneuronal tu- (1970). 217(4):335-42. PMID:4203636 et al. (1999). Molecular genetic analysis of 636. Elowe-Gruau E, Beltrand J, Brauner R,
mor: a case report and review of the literature. 604. Dratrnan MB (1979). Mechanism of thy- ependyrnal tumors. NF2 mutations and chro- Pinto G, Samara-Boustani D, Thalassinos C, et
Hurn Pathol. 37(7):914-8. PMID 16784993 roxine action. N Engl J Med. 300(23):1336. mosome 22q loss occur preferentially in intra- al. (2013). Childhood craniopharyngiorna: hypo-
587. Dincsr A, Yener U, bzek MM (2011). Hy- PMID:220535 rnedullary spinal ependymomas. Am J Pathol. thalamus-sparing surgery decreases the risk of
drocephalus in patients with neurofibrornatosis 605. Dropcho EJ, Soong SJ (1996). The pro- 155(2):627-32. PMID:10433955 obesity. J Clin Endocrinol Metab. 98(6):2376-
type 1: MR imaging findings and the outcome gnostic impact of prior low grade histology in 622. Eckel-Passow JE, Lachance DH, Mo- 82. PMID:23633208
of endoscopic third ventriculostomy. AJNR Am patients with anaplastic gliomas: a case-control linaro AM, Walsh KM, Decker PA, Sicotte 637. Emory TS, Scheithauer BW, Hirose T,
J Neuroradiol. 32(4):643-6. PMID:21330395 study. Neurology. 47(3):684-90. PMID:8797465 H, et al. (2015). Gliorna Groups Based on Wood M, Onofrio BM, Jenkins RB (1995). Intra-
588. Dinda AK, Sarkar C, Roy S (1990). 606. Ducatman BS, Scheithauer BW, Piepgras 1p/19q, IDH, and TERT Promoter Mutations neural perineurioma. A clonal neoplasm associ-
Rosenthal fibres: an imrnunohistochernical, DG, Reiman HM, llstrup OM (1986). Malignant in Tumors. N Engl J Med. 372(26):2499-508. ated with abnormalities of chromosome 22. Am
ultrastructural and immunoelectron micros- peripheral nerve sheath tumors. A clinicopatho- PMID:26061753 J Clin Pathol. 103(6):696-704. PMID:7785653
copic study. Acta Neuropathol. 79(4):456-60. logic study of 120 cases. Cancer. 57(10)'.2006- 623. Eckhardt BP, Brandner S, Zollikofer CL, 638. En-Nafaa I, Latib R, Fikri M, Cisse A, El
PMID:2339597 21. PMID:3082508 Wentz KU (2004). Primary cerebral leiornyos- Kettani N, Charni I, et al. (2010). [Frontoparietal
589. Ding H, Shannon P, Lau N, Wu X, Roncari 607. Ducray F, Crinere E, ldbaih A, Mokhtari K, arcorna in a child. Pediatr Radial. 34(6):495-8. paragangliorna: a case report]. J Radial. 91(12
L, Baldwin RL, et al. (2003). Oligodendroglio- Marie Y, Paris S, et al. (2009). alpha-lnternexin PMID:15057493 Pt 1):1318-9. PMID:21242920
rnas result from the expression of an activated expression identifies 1p19q codeleted gliornas. 624. Edwards A, Bermudez C, Piwonka G, Berr 639. Ene Cl, Morton RP, Ferreira M Jr, Sekhar
mutant epidermal growth factor receptor in a Neurology. 72(2):156-61. PMID:19139367 ML, Zamorano J, Larrain E, et al. (2002). Car- LN, Kirn LJ (2015). Spontaneous Hemorrhage
RAS transgenic mouse astrocytorna model. 608. Ducray F, ldbaih A, de Rsynies A, Bieche ney's syndrome: complex myxornas. Report of from Central Nervous System Hernangiob-
Cancer Res. 63(5):1106-13. PMID 12615729 I, Thillet J, Mokhtari K, et al. (2008). Anaplastic four cases and review of the literature. Cardio- lastomas. World Neurosurg. 83(6):1180.e13-7.
590. Dirks PB, Harris L, Hoffman HJ, oligodendrogliomas with 1p19q codeletion have vasc Surg. 10(3):264-75. PMID:12044436 PMID:25727302
Humphreys RP, Drake JM, Rutka JT (1996). a proneural gene expression profile. Mol Can- 625. Eigenbrod S, Roeber S, Thon N, Giese 640. Eng C (1997). Cowden Syndrome. J Genet
Supratentorial primitive neuroecloderrnal tu- cer. 7:41. PMID:18492260 A, Krieger A, Grasbon-Frodl E, et al. (2011). Couns. 6(2):181-92. PMID:26142096
mors in children. J Neurooncol. 29(1):7!>--84. 609. Duffell 0, Farber L, Chou S, Hartmann JF, a-lnternexin in the diagnosis of oligodendro- 641. Eng C, Murday V, Seal S, Mohammed
PMID:8817418 Nelson E (1963). Electron microscopic observa- glial tumors and association with 1p/19q sta- S, Hodgson SV, Chaudary MA, et al. (1994).
591. Dirks PB, Jay V, Becker LE, Drake JM, tions on astrocytomas. Am J Pathol. 43:539---45. tus. J Neuropathol Exp Neurol. 70(11):970--8. Cowden syndrome and Lherrnitte-Duclos di-
Humphreys RP, Hoffman HJ, et al. (1994). De- PMID:14068386 PMID:22002423 sease in a family: a single genetic syndrome
velopment of anaplastic changes in low-grade 610. Duffner PK, Horowitz ME, Krischer JP, Bur- 626. Ekstrand AJ, James CD, Cavenee WK, with pleiotropy? J Med Genet. 31(6):458--1,1.
astrocytomas of childhood. Neurosurgery. ger PC, Cohen ME, Sanford RA, et al. (1999). Seliger B, Pettersson RF, Collins VP (1991). PMID:8071972
34(1):68-78 PMID:8121571 The treatment of malignant brain tumors in in- Genes for epidermal growth factor receptor, 642. Eng C, Peacocke M (1998). PTEN and
592. do Nascimento A, Maranha LA, Correda- fants and very young children: an update of the transforming growth factor alpha, and epider- inherited harnartorna-cancer syndromes. Nat
to RA, Araujo JC, Bleggi-Torres LF (2012). 33 Pediatric Oncology Group experience. Neuro mal growth factor and their expression in human Genet. 19(3):223. PMID:9662392
year-old woman with a large sellar tumor. Brain Oneal. 1(2):152-61. PMID:11554387 gliornas in vivo. Cancer Res. 51(8):2164-72. 643. Eng DY, DeMonte F, Ginsberg L, FullerGN,

References 365
,/

Jaeckle K (1997). Craniospinal dissemination of medulloblastoma. Br J Cancer. 64(5):959-61. Mohamed FB, Chen CY, Stein H (1997). Para· Oligodendroglial tumors: refinement of candida-
central neurocytoma. Report of two cases. J PMID:1931625 ganglioma of the cauda equina with associated te regions on chromosome arm 1 p and correla.
Neurosurg 86(3):547-52. PMID:9046315 658. Evans DG, Howard E, Giblin C, Clancy intramedullary cyst: MR findings. AJNR Am J tion of 1 p/19q status with survival. Brain Pathol.
644. Eoli M, Menghi F, Bruzzone MG, De Simo· T, Spencer H, Huson SM, et al. (2010). Birth Neuroradiol. 18(8):1588-90. PMID:9296205 14(2):121-30. PMID:15193024
ne T, Valletta L, Pollo B, et al. (2007). Methyla· incidence and prevalence of tumor-prone 673. Farwell JR, Dohrmann GJ, Flannery JT 687. Feng M, Carmichael JD, Bonert V, Ban-
tion of 06-methylguanine DNA methyltrans· syndromes: estimates from a UK family ge· (1984). Medulloblastoma in childhood: an epi- nykh S, Mamelak AN (2014). Surgical ma-
ferase and loss of heterozygosity on 19q and/ netic register service. Am J Med Genet A. demiological study. J Neurosurg. 61(4):657-64. nagement of pituicytomas: case series and
or 17 p are overlapping features of secondary 152A(2):327-32. PMID:20082463 PMID:6470775 comprehensive literature review. Pituitary.
glioblastomas with prolonged survival. Clin 659. Evans DG, Huson SM, Donnai D, Neary 67 4. Fassett DR, Pingree J, Kestle JR (2005). 17(5):399-413. PMID:24037647
Cancer Res. 13(9):2606-13. PMID:17473190 W, Blair V, Newton V, et al. (1992). A clinical The high incidence of tumor dissemination in 688. Fernandez C, Figarella-Branger D, Girard
645. Erdem·Eraslan L, Gravendeel LA, de Rooi study of type 2 neurofibromatosis. Q J Med. myxopapillary ependymoma in pediatric pa· N, Bouvier-Labit C, Gouvernet J, Paz Paredes
J, Eilers PH, ldbaih A, Spliet WG, et al. (2013). 84(304):603-18. PMID:1484939 tients. Report of five cases and review of the A, et al. (2003). Pilocytic astrocytomas in child-
Intrinsic molecular subtypes of glioma are prog- 660. Evans DG, Ladusans EJ, Rimmer S, Bur- literature. J Neurosurg. 102(1) Suppl:59-64. ren: prognostic factors-a retrospective study of
nostic and predict benefit from adjuvant procar- nell LO, Thakker N, Farndon PA (1993). Com· PMID:16206735 80 cases. Neurosurgery. 53(3):544--53, discus-
bazine, lomustine, and vincristine chemothera- plications of the naevoid basal cell carcinoma 675. Fassunke J, Majores M, Tresch A, Niehus- sion 554-5. PMID:12943571
py in combination with other prognostic factors syndrome: results of a population based study. mann P, Grote A, Schoch S, et al. (2008). Array 689. Fernandez C, Girard N, Paz Paredes A
in anaplastic oligodendroglial brain tumors: a J Med Genet. 30(6):460-4. PMID:8326488 analysis of epilepsy-associated gangliogliomas Bouvier-Labit C, Lena G, Figarella-Branger o
report from EORTC study 26951. J Clin Oneal. 661. Evans DG, Mason S, Huson SM, Ponder reveals expression patterns related to aberrant (2003). The usefulness of MR imaging in the
31 (3):328-36. PMID:23269986 M, Harding AE, Strachan T (1997). Spinal and development of neuronal precursors. Brain. diagnosis of dysembryoplastic neuroepithelial
646. Erickson ML, Johnson R, Bannykh SI, cutaneous schwannomatosis is a variant form 131(Pt 11):3034-50. PMID:18819986 tumor in children: a study of 14 cases. AJNR Am
de Lotbiniere A, Kim JH (2005). Malignant of type 2 neurofibromatosis: a clinical and mo- 676. Fauchon F, Hasselblatt M, Jouvet A, J Neuroradiol. 24(5):829-34. PMID:12748079
rhabdoid tumor in a pregnant adult female: lecular study. J Neural Neurosurg Psychiatry. Champier J, Popovic M, Kirollos R, etal. (2013). 690. Fernandez-L A, Northcott PA, Dallon J
literature review of central nervous system 62(4):361-6. PMID:9120449 Role of surgery, radiotherapy and chemothera- Fraga C, Ellison D, Angers S, et al. (2009)'.
rhabdoid tumors. J Neurooncol. 74(3):311-9. 662. Evans DG, Moran A, King A, Saeed S, py in papillary tumors of the pineal region: a YAP1 is amplified and up-regulated in hedge.
PMID:16132523 Gurusinghe N, Ramsden R (2005). Incidence multicenter study. J Neurooncol. 112(2):223- hog-associated medulloblastomas and medi-
647. Erlandson RA (1994). Paragangliomas. of vestibular schwannoma and neurofibro- 31. PMID:23314823 ates Sonic hedgehog-driven neural precursor
Diagnostic Transmission Electron Microscopy matosis 2 in the North West of England over 677. Fauchon F, Jouvet A, Paquis P, Saint-Pier· proliferation. Genes Dev. 23(23):2729-41.
of Tumors. Diagnostic Transmission Electron a 10-year period: higher incidence than pre· re G, Mottolese C, Ben Hassel M, et al. (2000). PMID:19952108
Microscopy ofTumors. New York: Raven Press; viously thought. Otol Neurotol. 26(1 ):93-7. Parenchymal pineal tumors: a clinicopathologi· 691. Ferner RE, Hughes RA, Hall SM
pp. 615-22. PMID:15699726 cal study of 76 cases. Int J Radial Oncol Biol Upadhyaya M, Johnson MR (2004). Neuro'.
648. Erlandson RA, Woodruff JM (1982). Peri- 663. Evans DG, O'Hara C, Wilding A, Ingham Phys. 46(4):959-68. PMID:10705018 fibromatous neuropathy in neurofibromato-
pheral nerve sheath tumors: an electron micros· SL, Howard E, Dawson J, et al. (2011). Morta· 677A. Faulkner C, Ellis HP, Shaw A, Penman sis 1 (NF1). J Med Genet. 41(11):837-41.
copic study of 43 cases. Cancer. 49(2):273-87. lity in neurofibromatosis 1: in North West Eng- C, Palmer A, Wragg C, et al. (2015). BRAF PMID:15520408
PMID:7053827 land: an assessment of actuarial survival in a Fusion Analysis in Pilocytic Astrocytomas: 692. Ferreri AJ, Dell'Oro S, Capello D, Ponzoni
649. Ernestus RI, Schroder R, StOtzer H, Klug region of the UK since 1989. Eur J Hum Genet. KIAA1549·BRAF 15-9 Fusions Are More Fre- M, luzzolino P, Rossi D, et al. (2004). Aberrant
N (1996). Prognostic relevance of localization 19(11):1187-91. PMID:21694737 quent in the Midline Than Within the Cerebel· methylation in the promoter region of the re·
and grading in intracranial ependymomas of 664. Evans DG, Wallace AJ, Wu CL, Trueman lum. J Neuropathol Exp Neural. 74(9):867-72. duced folate carrier gene is a potential mecha-
childhood. Childs Nerv Syst. 12(9):522-6. L, Ramsden RT, Strachan T (1998). Somatic PMID:26222501 nism of resistance to methotrexate in primary
PMID:8906366 mosaicism: a common cause of classic disea- 678. Faulkner C, Palmer A, Williams H, Wragg central nervous system lymphomas. Br J Hae-
650. Ernestus RI, Schroder R, Stiitzer H, Klug se in tumor-prone syndromes? Lessons from C, Haynes HR, White P, et al. (2014). EGFR matol. 126(5):657-64. PMID:15327516
N (1997). The clinical and prognostic relevance type 2 neurofibromatosis. Am J Hum Genet. and EGFRvlll analysis in glioblastoma as 693. Ferreri AJ, Reni M, Pasini F, Calderoni A,
of grading in intracranial ependymomas. Br J 63(3):727-36. PMID:9718334 therapeutic biomarkers. Br J Neurosurg. 1-7. Tirelli U, Pivnik A, et al. (2002). A multicenter
Neurosurg. 11(5):421�. PMID:9474274 665. Fakhran S, Escott EJ (2008). Pineocytoma PMID:25141189 study of treatment of primary CNS lymphoma.
651. Errani C, Zhang L, Sung YS, Hajdu M, mimicking a pineal cyst on imaging: true diag· 679. Fauria-Robinson C, Nguyen J, Palacios Neurology. 58(10):1513-20. PMID:12034789
Singer S, Maki RG, et al. (2011 ). A novel nostic dilemma or a case of incomplete irna- E, Castillo-Jorge S (2014). Dysplastic cere- 694. Ferry JA, Harris NL, Picker LJ, Weinberg
WWTR1-CAMTA1 gene fusion is a consistent ging? AJNR Am J Neuroradiol. 29(1):159-63. bellar gangliocytoma lhermitte-duclos disease OS, Rosales RK, Tapia J, et al. (1988). Intra·
abnormality in epithelioid hemangioendothelio· PMID:17925371 imaging and magnetic resonance spectres- vascular lymphomatosis (malignant angioen·
ma of different anatomic sites. Genes Chroma· 666. Fallon KB, Palmer CA, Roth KA, Nabors copy. J La State Med Soc. 166(5):193-6. dotheliomatosis). A B-cell neoplasm expres-
somes Cancer. 50(8):644-53. PMID:21584898 LB, Wang W, Carpenter M, et al. (2004). Pro· PMID:25369219 sing surface homing receptors. Mod Pathol.
652. Erson-Omay EZ, <;:aglayan AO, Schultz gnostic value of 1p, 19q, 9p, 10q, and EG· 680. Favier J, Gimenez-Roqueplo AP (2010). 1(6):444-52. PMID:3065781
N, Weinhold N, Omay SB, bzduman K, et FR-FISH analyses in recurrent oligodendroglio· Pheochromocytomas: the (pseudo)-hypoxia 695. Fetsch JF, Miettinen M (1997). Sclerosing
al. (2015). Somatic POLE mutations cause mas. J Neuropathol Exp Neural. 63(4):314-22. hypothesis. Best Pract Res Clin Endocrinol Me· perineurioma: a clinicopathologic study of 19
an ultramutated giant cell high-grade glioma PMID:15099021 tab. 24(6):957-68. PMID:21115164 cases of a distinctive soft tissue lesion with a
subtype with better prognosis. Neuro Oncol. 667. Fan Z, Li J, Du J, Zhang H, Shen Y, Wang 681. Feany MB, Anthony DC, Fletcher CD predilection for the fingers and palms of young
17(10):1356-64. PMID:25740784 CY, et al. (2008). A missense mutation in (1998). Nerve sheath tumours with hybrid fea· adults. Am J Surg Pathol. 21(12):1433-42.
653. Esteller M, Garcia-Foncillas J, Andion E, PTCH2 underlies dominantly inherited NBCCS lures of neurofibroma and schwannoma: a con· PMID:9414186
Goodman SN, Hidalgo OF, Vanaclocha V, et in a Chinese family. J Med Genet. 45(5):303-8. ceptual challenge. Histopathology. 32(5):405- 696. Fevre Montange M, Vasiljevic A, Bergemer
al. (2000). Inactivation of the DNA-repair gene PMID:18285427 10. PMID:9639114 Fouquet AM, Bernier M. Champier J, Chretien
MGMT and the clinical response of gliomas to 668. Fanburg-Smith JC, Auerbach A, Marwaha 682. Feigin IH, Gross SW (1955). Sarcoma ari- F, et al. (2012). Histopathologic and ultrastruc-
alkylating agents. N Engl J Med. 343(19):1350- JS, Wang Z, Rushing EJ (2010). Reappraisal sing in glioblastoma of the brain. Am J Pathol. tural features and claudin expression in papil-
4. PMID:11070098 of mesenchymal chondrosarcoma: novel mar· 31(4):633-53. PMID:14388124 lary tumors of the pineal region: a multicenter
654. Esteller M, Hamilton SR, Burger PC, phologic observations of the hyaline cartilage 683. Feldman OS, Jordan C, Fonseca L (2010). analysis. Am J Surg Pathol. 36(6):916-28.
Baylin SB, Herman JG (1999). Inactivation of and endochondral ossification and beta-ca· Orthopaedic manifestations of neurofibromato- PMID:22588068
the DNA repair gene 06-methylguanine-DNA tenin, Sox9, and osteocalcin immunostaining sis type 1. J Am Acad Orthop Surg. 18(6):346- 697. Fevre Montange M, VasiljevicA, Champier
methyltransferase by promoter hypermethylati- of 22 cases. Hum Pathol. 41 (5):653-62. 57. PMID:20511440 J, Jouvet A (2015). Papillary tumor of the pineal
on is a common event in primary human neopla- PMID:20138330 683A. Feliciano OM, Quon JL, Su T, Taylor region: Histopathological characterization and
sia. Cancer Res. 59(4):793-7. PMID:10029064 669. Fargen KM, Opalach KJ, Wakefield D, MM, Bordey A (2012). Postnatal neurogenesis review of the literature. Neurochirurgie. 61(2-
655. Euskirchen P, Haroche J, Emile JF, Bu· Jacob RP, Yachnis AT, Lister JR (2011). The generates heterotopias, olfactory micronodu- 3):138-42 PMID:24556386
chert R, Vandersee S, Meisel A (2015). Com· central nervous system solitary fibrous tumor: les and cortical infiltration following single-cell 698. Fevre-Montanqe M, Champier J, Szalhrna-
plete remission of critical neurohistiocytosis by a review of clinical, imaging and pathologic fin· Tsc1 deletion. Hum Mal Genet. 21(4):799-810. ri A, Wierinckx A, Mottolese C, Guyotat J, et al.
vemurafenib. Neural Neuroimmunol Neuroin- dings among all reported cases from 1996 to PMID:22068588 (2006). Microarray analysis reveals differential
flamm. 2(2):e78. PMID:25745636 2010. Clin Neural Neurosurg. 113(9):703-10. 684. Felix I, Becker LE (1990-1991). lntracrani- gene expression patterns in tumors of the pine·
655A. Evans DG. Neurofibromatosis 2. 1998 PMID:21872387 al germ cell tumors in children: an immunohis- al region. J Neuropathol Exp Neural. 65(7):675-
(Updated 2011 Aug 18]. In: Pagon RA, Adam 670. Faria C, Miguens J, Antunes JL, Barroso tochemical and electron microscopic study. Pe- 84. PMID:16825954
MP, Ardinger HH, et al., editors. GeneReviews® C, Pimentel J, Martins MdoC, el al. (2008). diatr Neurosurg. 16(3):156-62. PMID:1966857 699. Fevre-Montange M, Hasselblatt M, Fi·
(Internet). Seattle (WA): University of Washing- Genetic alterations in a papillary glioneuron· 685. Fellah S, Caudal D, De Paula AM, Do- garella-Branger D, Chauveinc L, Champier J,
ton, Seattle; 1993-2016. Available from: http:// al tumor. J Neurosurg Pediatr. 1(1):99-102. ry-Lautrec P, Figarella-Branger D, Chino! 0, et Saint-Pierre G, el al. (2006). Prognosis and
www.ncbi.nlm.nih.gov/books/NBK1201/ PMID:18352813 al. (2013). Multimodal MR imaging (diffusion, histopathologic features in papillary tumors of
656. Evans DG, Birch JM, Ramsden RT 671. Farnia B, Allen PK, Brown PD, Khatua perfusion, and spectroscopy): is it possible to the pineal region: a retrospective multicenter
(1999). Paediatric presentation of type 2 neu- S, Levine NB, Li J, et al. (2014). Clinical out· distinguish oligodendroglial tumor grade and study of 31 cases. J Neuropathol Exp Neural.
rofibromatosis. Arch Dis Child. 81(6):496-9. comes and patterns of failure in pineoblasto- 1 p/19q codeletion in the pretherapeutic diag· 65(10):1004--11. PMID:17021405
PMID: 10569966 ma: a 30-year, single-institution retrospective nosis? AJNR Am J Neuroradiol. 34(7):1326-33. 700. Fevre-Montanqe M, Jouvet A, Privat K,
657. Evans DG, Farndon PA, Burnell LO, Gat- review. World Neurosurg. 82(6):1232-41. PMID:23221948 Korf HW, Champier J, Reboul A, et al. (1998).
tamaneni HR, Birch JM (1991). The incidence PMID:25045788 686. Felsberg J, Erkwoh A, Sabel MC, Kirsch lmmunohistochemical, ultrastructural, bioche-
of Gorlin syndrome in 173 consecutive cases of 672. Faro SH, Turtz AR, Koenigsberg RA, L, Fimmers R, Blaschke B, et al. (2004). mical and in vitro studies of a pineocytoma.

366 References
Acta Neuropathol. 95(5):532-9. PMID:9600600 B, Sadetzki S (2011). Possible interaction bet- resolved through single-nucleus sequencing. 745. Fu YJ, Taniguchi Y, Takeuchi S, Shiga A,
701. Fevre-Montanqe M, Szathmari A, Cham- ween ionizing radiation, smoking, and gender Cancer Discov. 4(8):956-71. PMID:24893890 Okamoto K, Hirata J, et al. (2013). Cerebral
pier J, Mokhtari K, Chretien F, Coulon A, et al. in the causation of meningioma. Neuro Oncol. 732A. Franz ON, Belousova E, Sparagana S, astroblastoma in an adult: an immunohistoche-
(2008). Pineocytoma and pineal parenchymal 13(3):345-52. PMID:21339193 Bebin EM, Frost M, Kuperman R, et al. (2013). mical, ultrastructural and genetic study. Neuro-
tumors of intermediate differentiation presen- 717. Flint-Richter P, Sadetzki S (2007). Ge- Efficacy and safety of everolimus for sub- pathology. 33(3):312-9. PMID:22994361
ting cytologic pleomorphism: a multicenter stu- netic predisposition for the development of ependymal giant cell astrocytomas associated 746. Fu YS, Chen AT, Kay S, Young H (1974).
dy. Brain Pathol. 18(3):354-9. PMID:18371183 radiation-associated meningioma: an epide- with tuberous sclerosis complex (EXIST-1 ): Is subependymoma (subependymal glomerate
702. Fevre-Montanqe M, Vasiljevic A, Frappaz miological study. Lancet Oncol. 8(5):403-10. a multicentre, randomised, placebo-control- astrocytoma) an astrocytoma or ependymo-
D, Champier J, Szathmari A, Aubriot Lorton MH, PMID:17466897 led phase 3 trial. Lancet. 381(9861):125-32. ma? A comparative ultrastructural and tissue
et al. (2012). Utility of Ki67 immunostaining in 718. Folpe AL (2014). Selected topics in the PMID:23158522 culture study. Cancer. 34(6):1992-2008.
the grading of pineal parenchymal tumours: a pathology of epithelioid soft tissue tumors. Mod 733. Franz ON, Belousova E, Sparagana S, PMID:4434329
multicentre study. Neuropathol Appl Neurobiol. Pathol. 27 Suppl 1:564-79. PMID24384854 Bebin EM, Frost M, Kuperman R, et al. (2014). 746A. Fujii K, Miyashita T (2014). Gorlin syn-
38(1):87-94. PMID:21696422 719. Folpe AL, Billings SD. McKenney JK, Everolimus for subependymal giant cell ast- drome (nevoid basal cell carcinoma syndro-
703. Figarella-Branger D, Civatte M, Bou- Walsh SV, Nusrat A, Weiss SW (2002). Ex- rocytoma in patients with tuberous sclerosis me): update and literature review. Pediatr Int.
vier-Labit C, Gouvernet J, Gambarelli D, Gentet pression of claudin-1, a recently described tight complex: 2-year open-label extension of the 56(5):667-74. PMID:25131638
JC, et al. (2000). Prognostic factors in intracra- junction-associated protein, distinguishes soft randomised EXIST-1 study. Lancet Oneal. 747. Fujii K, Ohashi H, Suzuki M, Hatsuse H,
nial ependymomas in children. J Neurosurg. tissue perineurioma from potential mimics. Am 15(13):1513-20. PMID:25456370 Shiohama T, Uchikawa H, et al. (2013). Fra-
93(4):605-13. PMID:11014538 J Surg Pathol. 26(12):1620-6. PMID:12459629 734. Frappaz D, Ricci AC, Kohler R, Bret P, meshift mutation in the PTCH2 gene can cause
704. Figarella-Branger D, Gambarelli D, Dolio 720. Font RL, Truong LO (1984). Melanotic Mottolese C (1999). Diffuse brain stem tumor in nevoid basal cell carcinoma syndrome. Fam
C, Devictor B, Perez-Castillo AM, Genitori L, et schwannoma of soft tissues. Electron-micros- an adolescent with multiple enchondromatosis Cancer 12(4):611-4. PMID23479190
al. (1991). lnfratentorial ependymomas of child- copic observations and review of literature. Am (Ollier's disease). Childs Nerv Syst. 15(5):222- 748. Fujimaki T, Matsutani M, Funada N, Kiri-
hood. Correlation between histological features, J Surg Pathol. 8(2):129-38. PMID:6703189 5. PMID 10392492 no T, Takakura K, Nakamura 0, et al. (1994).
immunohistological phenotype, silver nucleolar 721. Fontebasso AM, Papillon-Cavanagh S, 735. Frattini V, Trifonov V, Chan JM, Castano CT and MRI features of intracranial germ
organizer region staining values and post-ope- Schwartzentruber J, Nikbakht H, Gerges N, Fi- A, Lia M, Abate F, et al. (2013). The integra- cell tumors. J Neurooncol. 19(3):217-26.
rative survival in 16 cases. Acta Neuropathol. set PO, et al. (2014). Recurrent somatic muta- ted landscape of driver genomic alterations PMID:7807172
82(3):208-16. PMID:1718129 tions in ACVR1 in pediatric midline high-gra- in glioblastoma. Nat Genet. 45(10):1141-9. 749. Fujimoto K, Ohnishi H, Tsujimoto M,
705. Figarella-Branger D, Pellissier JF, Dau- de astrocytoma. Nat Genet. 46(5):462-6. PMID:23917401 Hoshida T, Nakazato Y (2000). Dysembryo-
mas-Duport C, Delisle MB, Pasquier B, Parent PMID:24705250 736. Frebourg T, Barbier N, Yan YX, Garber JE, plastic neuroepithelial tumor of the cerebellum
M, et al. (1992). Cenlral neurocytomas. Critical 722. Fontebasso AM, Schwartzentruber J, Dreyfus M, Fraumeni J Jr, et al. (1995). Germ- and brainstem. Case report. J Neurosurg.
evaluation of a small-cell neuronal tumor. Am J Khuong-Quang DA, Liu XY, Sturm D, Korshu- line p53 mutations in 15 families with Li-Frau- 93(3):487-9. PMID:10969950
Surg Pathol. 16(2):97-109. PMID:1370756 nov A, et al. (2013). Mutations in SETD2 and meni syndrome. Am J Hum Genet. 56(3):608- 750. Fujisawa H, Kurrer M, Reis RM, Yonekawa
706. Figueroa ME, Abdel-Wahab 0, Lu C, Ward genes affecting histone H3K36 methylation 15. PMID:7887414 Y, Kleihues P, Ohgaki H (1999). Acquisition of
PS, Patel J, Shih A, et al. (2010). Leukemic target hemispheric high-grade gliomas. Acta 737. Freilich RJ, Thompson SJ, Walker RW, the glioblastoma phenotype during astrocytoma
IDH1 and IDH2 mutations result in a hyper- Neuropathol. 125(5):659-69. PMID23417712 Rosenblum MK (1995). Adenocarcinomat- progression is associated with loss of heterozy-
methylation phenotype, disrupt TET2 function, 723. Forest F, David A, Arrufat S, Pierron G, ous transformation of intracranial germ cell gosity on 1 Oq25-qter. Am J Pathol. 155(2):387-
and impair hematopoietic differentiation. Can- Ranchere-Vince D, Stephan JL, et al. (2012). tumors. Am J Surg Pathol. 19(5):537-44. 94. PMID:10433932
cer Cell. 18(6):553-67. PMID:21130701 Conventional chondrosarcoma in a survivor PMID:7726363 751. Fujisawa H, Marukawa K, Hasegawa M,
707. Fine SW, McClain SA, Li M (2004). lm- of rhabdoid tumor: enlarging the spectrum of 738. Frenk E, Marazzi A (1984). Neurofibroma- Tohma Y, Hayashi Y, Uchiyama N, et al. (2002).
munohistochemical staining for calretinin is tumors associated with SMAR CB 1 germline tosis of van Recklinghausen: a quantitative stu- Genetic differences between neurocytoma
useful for differentiating schwannomas from mutations. Am J Surg Pathol. 36(12):1892-6. dy of the epidermal keratinocyte and melanocy- and dysembryoplastic neuroepithelial tumor
neurofibromas. Am J Clin Pathol. 122(4):552-9. PMID:23154773 te populations. J Invest Dermatol. 83(1 ):23-5. and oligodendroglial tumors. J Neurosurg.
PMID:15487453 724. Forshew T, Tatevossian RG, Lawson PMID:6203987 97(6):1350-5. PMID:12507133
708. Fischer I, Gagner JP, Law M, Newcomb AR, Ma J, Neale G, Ogunkolade BW, et al. 739. Freudenstein D, Wagner A, Bornemann 752. Fujisawa H, Reis RM, Nakamura M, Co-
EW, Zagzag D (2005). Angiogenesis in gliomas: (2009). Activation of the ERK/MAPK pathway: A, Ernemann U, Bauer T, Duffner F (2004). Pri- lella S, Yonekawa Y, Kleihues P, et al. (2000).
biology and molecular pathophysiology. Brain a signature genetic defect in posterior fossa pi- mary melanocytic lesions of the CNS: report of Loss of heterozygosity on chromosome 10 is
Pathol. 15(4):297-310. PMID:16389942 locytic astrocytomas. J Pathol. 218(2):172-81. five cases. Zentralbl Neurochir. 65(3):146-53. more extensive in primary (de nova) than in
709. Fisher PG, Breiter SN, Carson BS, Wha- PMID:19373855 PMID:15306980 secondary glioblastomas. Lab Invest. 80(1):65-
ram MD, Williams JA, Weingart JD, et al. 725. Forsyth PA, Shaw EG, Scheithauer BW, 739A. Friedman JM. Neurofibromatosis 1. 1998 72. PMID:10653004
(2000). A clinicopathologic reappraisal of brain O'Fallon JR, Layton DD Jr, Katzmann JA [Updated 2014 Sep 4). In: Pagon RA, Adam MP, 753. Fujisawa H, Tohma Y, Muramatsu N, Kida
stem tumor classification. Identification of pi- (1993). Supratentorial pilocytic astrocytomas. A Ardinger HH, et al., editors. GeneReviews® [In- S, Kaizaki Y, Tamamura H (2012). Spindle cell
locystic astrocyloma and fibrillary astrocytoma clinicopathologic, prognostic, and flow cytomet- ternet]. Seattle (WA): University of Washington, oncocytoma of the adenohypophysis with mar-
as distinct entities. Cancer. 89(7):1569-76. ric study of 51 patients. Cancer. 72(4):1335-42. Seattle; 1993-2016. Available from: http://www. ked hypervascularity. Case report. Neurol Med
PMID:11013373 PMID:8339223 ncbi.nlm.nih.gov/books/NBK 1109/ Chir (Tokyo). 52(8):594-8. PMID:22976144
710. Fix SE, Nelson J, Schochet SS Jr (1989). 726. Foster RD, Williams ML, Barkovich AJ, 740. Friedman JM, Arbiser J, Epstein JA, 754. Fukuda T, Akiyama N, lkegami M, Takahas-
Focal leptomeningeal rhabdomyomatosis of Hoffman WY, Mathes SJ, Frieden IJ (2001). Gutmann DH, Huot SJ, Lin AE, et al. (2002). hi H, Sasaki A, Oka H, et al. (2010). Expression
the posterior Iossa. Arch Pathol Lab Med. Giant congenital melanocytic nevi: the signifi- Cardiovascular disease in neurofibromatosis 1: of hydroxyindole-0-methyltransferase enzyme
113(8):872-3. PMID:2757487 cance of neurocutaneous melanosis in neuro- report of the NF1 Cardiovascular Task Force. in the human central nervous system and in
711. Flament-Durand J, Brion JP (1985). Tany- logically asymptomatic children. Plast Reconstr Genet Med. 4(3):105-11. PMID:12180143 pineal parenchymal cell tumors. J Neuropathol
cytes: morphology and functions: a review. Int Surg. 107(4):933-41. PMID:11252085 741. Friedrich C, Warmuth-Metz M, van Bueren Exp Neural. 69(5):498-510. PMID:20418777
Rev Cytol. 96:121-55. PMID:2416706 727. Fouladi M, Helton K, Dalton J, Gilger E, AO, NowakJ, Bison B, van Hoff K, et al. (2015). 755. Fukuda T, Yasumichi K, Suzuki T (2008).
712. Flamme I, Krieg M, Plate KH (1998). Gajjar A, Merchant T, et al. (2003). Clear cell Primitive neuroectodermal tumors of the brains- lmmunohistochemistry of gliosarcoma with
Up-regulation of vascular endothelial growth ependymoma: a clinicopathologic and ra- tem in children treated according to the HIT liposarcomatous differentiation. Pathol Int.
factor in stromal cells of hemangioblastomas diographic analysis of 10 patients. Cancer. trials: clinical findings of a rare disease. J Neu- 58(6):396-401. PMID:18477220
is correlated with up-regulation of the tran- 98(10):2232-44. PMID:14601094 rosurg Pediatr. 15(3):227-35. PMID:25555122 756. Fukunaga M (2001 ). Unusual malignant
scription factor HRF/HIF-2alpha. Am J Pathol. 728. Foulkes WO, Clarke BA, Hasselblatt 742. Fritchie KJ, Carver P, Sun Y, Batiouchko perineurioma of soft tissue. Virchows Arch.
153(1):25-9. PMID:9665461 M, Majewski J, Albrecht S, McCluggage WG G, Billings SD, Rubin BP, et al. (2012). Solitary 439(2):212-4. PMID:11561764
713. Flannery T, Cawley D, Zulfiger A, Alderazi (2014). No small surprise - small cell carcinoma fibrous tumor: is there a molecular relationship 757. Fukuoka K, Sasaki A, Yanagisawa T, Su-
Y, Heffernan J, Brett F, et al. (2008). Familial of the ovary, hypercalcaemic type, is a malig- with cellular angiofibroma, spindle cell lipoma, zuki T, Wakiya K, Adachi J, et al. (2012). Pineal
occurrence of oligodendroglial tumours. Br J nant rhabdoid tumour. J Pathol. 233(3):209-14. and mammary-type myofibroblastoma? Am J parenchymal tumor of intermediate differenti-
Neurosurg. 22(3):436-8. PMID:18568735 PMID:24752781 Clin Pathol. 137(6):963-70. PMID:22586056 ation with marked elevation of MIB-1 labeling
713A. Flavahan WA, Drier Y, Liau BB, Gillespie 729. Fountas KN, Karampelas I, Nikolakakos 742A. Fritz A, Percy C, Jack A, Shanmugarat- index. Brain Tumor Pathol. 29(4):229-34.
SM, Venteicher AS, Stemmer-Rachamimov AO, LG, Troup EC, Robinson JS (2005). Primary nam K, Sobin L, Parkin OM, et al. (2000). Inter- PMID:22362162
et al. (2016). Insulator dysfunction and oncoge- spinal cord oligodendroglioma: case report national Classification of Diseases for Oncology 758. Fukushima S, Otsuka A, Suzuki T, Yana-
ne activation in IDH mutant gliomas. Nature. and review of the literature. Childs Nerv Syst. (ICD-0). Third edition. World Health Organiza- gisawa T, Mishima K, Mukasa A, et al.; ln-
529(7584):110-4. PMID:26700815 21(2):171-5. PMID:15138790 tion: Geneva. tracranial Germ Cell Tumor Genome Analysis
714. Fletcher COM, Bridge JA, Hogendoorn 730. Fowler M, Simpson DA (1962). A malig- 743. Fruehwald-Pallamar J, Puchner SB, Consortium (iGCT Consortium) (2014). Mutual-
PCW, Mertens F (2013). WHO Classification nant melanin-forming tumour of the cerebellum. Rossi A, Garre ML, Gama A, Koelblinger C, ly exclusive mutations of KIT and RAS are as-
of Tumours of Soft Tissue and Bone. 4th ed. J Pathol Bacteriol. 84:307-11. PMID:13958991 et al. (2011). Magnetic resonance imaging sociated with KIT mRNA expression and chro-
Lyon: IARC. 731. Fox BO, Cheung VJ, Patel AJ, Suki D, spectrum of medulloblastoma. Neuroradiology. mosomal instability in primary intracranial pure
715. Fletcher COM, Unni KK, Mertens F, edi- Rao G (2011 ). Epidemiology of metastatic brain 53(6) 387-96. PMID:21279509 germinomas. Acta Neuropathol. 127(6):911-25.
tors. (2002). World Health Organization Classi- tumors. Neurosurg Clin N Am. 22(1):1-6, v. 744. Fryssira H, Leventopoulos G, Psoni S, PMID:24452629
fication of Tumours. Pathology and Genetics of PMID:21109143 Kitsiou-T zeli S, Stavrianeas N, Kanavakis 759. Fukushima S, Yoshida A, Narita Y, Arita
Tumours of Soft Tissue and Bone. 3rd ed. Lyon: 732. Francis JM, Zhang CZ, Maire CL, Jung E (2008). Tumor development in three pati- H, Ohno M, Miyakita Y, et al. (2015). Multino-
IARC Press. J, Manzo VE, Adalsteinsson VA, et al. (2014). ents with Noonan syndrome. Eur J Pediatr. dular and vacuolating neuronal tumor of the
716. Flint-Richter P, Mandelzweig L, Oberman EGFR variant heterogeneity in glioblastoma 167(9):1025-31. PMID:18057963 cerebrum. Brain Tumor Pathol. 32(2):131-6.

References 367
PMID:25146549 776. Gajjar A, Packer RJ, Foreman NK, Cohen 15(1):35-47. PMID:24314616 809. Geyer JR, Sposto R, Jennings M, Boyett
760. Fuld AD, Speck ME, Harris BT, Simmons K, Haas-Kogan D, Merchant TE; COG Brain 792. Gavrilovic IT, Posner JB (2005). Brain me- JM, Axtell RA, Breiger D, et al.; Children's Can-
NE, Corless CL, Tsongalis GJ, et al. (2011 ). Tumor Committee (2013). Children's Oncology tastases: epidemiology and pathophysiology. J cer Group (2005). Multiagent chemotherapy
Primary melanoma of the spinal cord: a case Group's 2013 blueprint for research: central Neurooncol. 75(1):5-14. PMID:16215811 and deferred radiotherapy m infants with mali-
report, molecular footprint, and review of the nervous system tumors. Pediatr Blood Cancer. 793. Geddes JF, Thom M, Robinson SF, Re- gnant brain tumors: a report from the Children's
literature. J Clin Oneal. 29(17):e499-502. 60(6):1022-6. PMID:23255213 vesz T (1996). Granular cell change in ast- Cancer Group. J Clin Oneal. 23(30):7621-31.
PMID:21444862 777. Gajjar A, Pfister SM, Taylor MD, Gilbert- rocytic tumors. Am J Surg Pathol. 20(1):55-63. PMID:16234523
761. Fulham MJ, Melisi JW, Nishimiya J, Dwyer son RJ (2014). Molecular insights into pediatric PMID:8540609 810. Geyer JR, Zeltzer PM, Boyett JM, Rorke
AJ, Di Chiro G (1993). Neuroimaging of juvenile brain tumors have the potential to transform 794. Gelabert-Gonzalez M (2005). Paragangli- LB, Stanley P, Albright AL, et al. (1994). Sur-
pilocytic astrocytomas: an enigma. Radiology. therapy. Clin Cancer Res. 20(22):5630-40. omas of the lumbar region. Report of two cases vival of infants with primitive neuroectodermal
189(1):221-5. PMID:8372197 PMID:25398846 and review of the literature. J Neurosurg Spine. tumors or malignant ependymomas of the CNS
762. Fuller C, Fouladi M, Gajjar A, Dalton J, 778. Galan SR, Kann PH (2013). Genetics and 2(3):354-65. PMID:15796363 treated with eight drugs in 1 day: a report from
Sanford RA, Helton KJ (2006). Chromosome molecular pathogenesis of pheochromocyto- 795. Gempt J, Baldawa SS, Weirich G, Del- the Childrens Cancer Group. J Clin Oneal.
17 abnormalities in pediatric neuroblastic tumor ma and paraganglioma. Clin Endocrinol (Oxf), bridge C, Hempel M, Lohse P, et al. (2013). 12(8):1607-15. PMID:8040673
with abundant neuropil and true rosettes. Am J 78(2):165-75. PMID:23061808 Recurrent multiple spinal paragangliomas as 811. Gherardi R, Baudrimont M, Nguyen JP,
Clin Pathol. 126(2):277-83. PMID:16891204 779. Galanis E, Buckner JC, Dinapoli RP, a manifestation of a metastatic composite pa- Gaston A, Cesaro P, Degos JD, et al. (1986).
763. Fuller CE, Perry A (2002). Fluorescence Scheithauer BW, Jenkins RB, Wang CH, et al. raganglioma-ganglioneuroblastoma. Acta Neu- Monstrocellular heavily lipidized malignant
in situ hybridization (FISH) in diagnostic and (1998). Clinical outcome of gliosarcoma compa- rochir (Wien). 155(7):1241-2. PMID:23532344 glioma. Acta Neuropathol. 69(1-2):28-32.
investigative neuropathology. Brain Pathol. red with glioblastoma multiforme: North Central 796. Gem pt J, Ringel F, Oexle K, Delbridge PMID:3515829
12(1):67-86. PMID:11770903 Cancer Treatment Group results. J Neurosurg. C, F6rschler A, Schlegel J, et al. (2012). Fa- 812. Gheyi V, Hui FK, Doppenberg EM, Todd W
764. Fuller GN, Signer SH (1992). Ampli- 89(3):425-30. PMID:9724117 milial pineocytoma. Acta Neurochir (Wien). Broaddus WC (2004). Glioblastoma multiform�
fied cellular oncogenes in neoplasms of the 780. Galli R, Binda E, Orfanelli U, Cipelletti B, 154(8):1413-6. PMID:22699425 causing calvarial destruction: an unusual ma-
human central nervous system. Mutat Res. Gritti A, De Vitis S, et al. (2004). Isolation and 797. George DH, Scheithauer BW, Aker FV, nifestation revisited. AJNR Am J Neuroradiol.
276(3):299-306. PMID: 137 4522 characterization of tumorigenic, stem-like neu- Kurtin PJ, Burger PC, Cameselle-Teijeiro J, et 25(9):1533-7. PMID:15502132
765. Fuller GN, Hess KR, Rhee CH, Yung WK, ral precursors from human glioblastoma. Can- al. (2003). Primary anaplastic large cell lympho- 813. GhiaAJ,Allen PK, Mahajan A, Penas-Pra-
Sawaya RA, Bruner JM, et al. (2002). Molecular cer Res. 64(19):7011-21. PMID:15466194 ma or the central nervous system: prognostic do M, Mccutcheon IE, Brown PD (2013). ln-
classification of human diffuse gliomas by mul- 781. Gallina P, Buccoliero AM, Mariotti F, effect or ALK-1 expression. Am J Surg Pathol. tracranial hemangiopericytoma and the role of
tidimensional scaling analysis of gene expres- Mennonna P, Di Lorenzo N (2006). Oncocytic 27(4):487-93. PMID:12657933 radiation therapy: a population based analysis.
sion profiles parallels morphology-based clas- meningiomas: Cases with benign histopatholo- 798. Gerson SL (2004). MGMT: its role in can- Neurosurgery. 72(2):203-9. PMID:23149953
sification, correlates with survival, and reveals gical features and a favorable clinical course. J cer aetiology and cancer therapeutics. Nat Rev 814. Ghia AJ, Chang EL, Allen PK, Mahajan A,
clinically-relevant novel glioma subsets. Brain Neurosurg. 105(5):736-8. PMID:17121136 Cancer. 4(4):296-307. PMID:15057289 Penas-Prado M, Mccutcheon IE, et al. (2013).
Pathol. 12(1):108-16. PMID:11771519 782. Gao J, Aksoy BA, Dogrusoz U, Dresdner 799. Gerstner ER, Abrey LE, Schiff D, Ferre- lntracranial hemangiopericytoma: patterns of
766. Fung KM, Fang W, Norton RE, Torres N, G, Gross B, Sumer SO, et al. (2013). Integra- ri AJ, Lister A, Montoto S, et al. (2008). CNS failure and the role of radiation therapy. Neu-
Chu A, Langford LA (2003). Cerebellar central tive analysis of complex cancer genomics and Hodgkin lymphoma. Blood. 112(5):1658-61. rosurgery. 73(4):624-30, discussion 630-1.
liponeurocytoma. Ultrastruct Pathol. 27(2):109- clinical profiles using the cBioPortal. Sci Signal. PMID:18591379 PMID:23839520
14. PMID:12746202 6(269):pl1. PMID:23550210 800. Gessi M, Abdel Moneim Y, Hammes J, 815. Ghose A, Guha G, Kundu R, Tew J, Chau-
767. Furnari FB, Huang HJ, Cavenee WK 783. Gao X, Zhang Y, Arrazola P, Hino 0, Ko- Waha A, Pietsch T (2014). FGFR1 N546K dhary R (2014). CNS Hemangiopericytoma: A
(1998). The phosphoinositol phosphatase bayashi T, Yeung RS, et al. (2002). Tse tumour mutation in a case of papillary glioneuronal tu- Systematic Review of 523 Patients. Am J Clin
activity of PTEN mediates a serum-sensitive suppressor proteins antagonize amino-acid- mor (PGNT). Acta Neuropathol. 127(6):935-6. Oneal. PMID:25350465
G 1 growth arresl in glioma cells. Cancer Res. TOR signalling. Nat Cell Biol. 4(9):699-704. PMID:24777483 816. Giangaspero F, Cenacchi G, Losi L,
58(22):5002-8. PMID:9823298 PMID:12172555 801. Gessi M, Giangaspero F, Lauriola L, Gar- Cerasoli S, Bisceglia M, Burger PC (1997).
768. Furnari FB, Lin H, Huang HS, Cavenee 784. Garcia OM, Fulling KH (1985). Juvenile pi- diman M, Scheithauer BW, Halliday W, et al. Extraventricular neoplasms with neurocyto-
WK (1997). Growth suppression of glioma cells locytic astrocytoma of the cerebrum in adults. A (2009). Embryonal tumors with abundant neu- ma features. A clinicopathological study of
by PTEN requires a functional phosphatase distinctive neoplasm with favorable prognosis. J ropil and true rosettes: a distinctive CNS primi- 11 cases. Am J Surg Pathol. 21(2):206-12.
catalytic domain. Proc Natl Acad Sci U S A Neurosurg. 63(3):382-6. PMID:4020465 tive neuroectodermal tumor. Am J Surg Pathol. PMID:9042288
94(23):12479-84. PMID:9356475 785. Garcia-Lavandeira M, Saez C, Diaz-Rodri- 33(2):211-7. PMID:18987548 817. Giangaspero F, Cenacchi G, Roncaroli
769. Furtado A, Arantes M, Silva R, Romao H, guez E, Perez-Romero S, Senra A, Dieguez C, 802. Gessi M, Giangaspero F, Pietsch T F, Rigobello L, Manetta V, Gambacorta M, et
Resende M, Honavar M (2010). Comprehensi- et al. (2012). Craniopharyngiomas express em- (2003). Atypical teratoid/rhabdoid tumors and al. (1996). Medullocytoma (lipidized medul-
ve review of extraventricular neurocytoma with bryonic stem cell markers (SOX2, OCT4, KLF4, choroid plexus tumors: when genetics "sur- loblastoma). A cerebellar neoplasm of adults
report of two cases, and comparison with cen- and SOX9) as pituitary stem cells but do not prise" pathology. Brain Pathol. 13(3):409-14. with favorable prognosis. Am J Surg Pathol.
tral neurocytoma. Clin Neuropathol. 29(3): 134- coexpress RET/GFRA3 receptors. J Clin En- PMID:12946029 20(6):656-64. PMID:8651344
40. PMID:20423686 docrinol Metab. 97(1):E80-7. PMID:22031517 803. Gessi M, Lambert SR, Lauriola L, Waha 818. Giangaspero F, Chieco P, Ceccarelli C,
770. Furuya K, Takanashi S, Ogawa A, Ta- 786. Gardiman MP, Fassan M, Orvieto E, A, Collins VP, Pietsch T (2012). Absence of Lisignoli G, Pozzuoli R, Gambacorta M, et al.
kahashi Y, Nakagomi T (2014). High-dose D'Avella D, Denaro L, Calderone M, et al. KIM 1549-BRAF fusion in rosette-forming glio- (1991). "Desmoplastic" versus "classic" me-
metholrexate monotherapy followed by radia- (2010). Diffuse leptomeningeal glioneuronal tu- neuronal tumors of the fourth ventricle (RGNT). dulloblastoma: comparison of DNA content,
tion for CD30-positive, anaplastic lymphoma mors: a new entity? Brain Pathol. 20(2):361-6. J Neurooncol. 110(1):21-5. PMID:22814862 histopathology and differentiation. Virchows
kinase-1-positive anaplastic large-cell lympho- PMID:19486008 804. Gessi M, Moneim YA, Hammes J, Go- Arch A Pathol Anal Histopalhol. 418(3):207-14.
ma in the brain of a child. J Neurosurg Pediatr. 787. Garre ML, Cama A, Bagnasco F, Marana schzik T, Scholz M, Denkhaus D, et al. (2014). PMID:1900966
14(3):311-5 PMID:25014324 G, Giangaspero F, Brisigotti M, et al. (2009). FGFR1 mutations in Rosette-forming glioneu- 819. Giangaspero F, Guiducci A, Lenz FA,
771. Gabeau-Lacet D, Aghi M, Betensky RA, Medulloblastoma variants: age-dependent ronal tumors of the fourth ventricle. J Neuropa- Mastronardi L, Burger PC (1999). Meningioma
Barker FG, Loeffler JS, Louis ON (2009). Bone occurrence and relation to Gorlin syndrome-a thol Exp Neural. 73(6):580-4. PMID:24806303 with meningioangiomatosis: a condition mimi-
involvement predicts poor outcome in atypical new clinical perspective. Clin Cancer Res. 805. Gessi M, Setty P, Bisceglia M, zur Muehlen cking invasive meningiomas in children and
meningioma. J Neurosurg. 111(3):464-71. 15(7):2463-71. PMID:19276247 A, Lauriola L, Wah a A, et al. (2011 ). Supraten- young adults: report of two cases and review
PMID:19267533 788. Gasco J, Franklin B, Fuller GN, Salin- torial primitive neuroectodermal tumors of the of the literature. Am J Surg Pathol. 23(8):872-5.
772. Gadish T, Tulchinsky H, Deutsch AA, Ra- as P, Prabhu S (2009). Multifocal•epithelioid central nervous system in adults: molecular and PMID:10435554
bau M (2005). Pinealoblastoma in a patient glioblastoma mimicking cerebral metastasis: histopathologic analysis of 12 cases. Am J Surg 820. Giangaspero F, Perilongo G, Fondelli MP,
with familial adenomatous polyposis: variant case report. Neurocirugia (Astur). 20(6):550-4. Pathol. 35(4):573-82. PMID:21378543 Brisigotti M, Carollo C, Burnelli R, et al. (1999).
of T urcot syndrome type 2? Report of a case PMID:19967320 806. Gessi M, van Bueren A, Treszl A, zur Medulloblastoma with extensive nodularity: a
and review of the literature. Dis Colon Rectum. 789. Gaspar LE, Mackenzie IR, Gilbert JJ, Miihlen A, Hartmann W, Wanmuth-Metz M, et variant with favorable prognosis. J Neurosurg.
48(12):2343-6. PMID:16400511 Kaufmann JC, Fisher BF, Macdonald DR, et al. al. (2014). MYCN amplification predicts poor 91(6):971-7. PMID:10584843
773. Gaffney EF, Doorly T, Dinn JJ (1986). Ag- (1993). Primary cerebral fibrosarcomas. Clini- outcome for patients with supratentorial pri- 821. Giangaspero F, Rigobello L, Badiali M,
gressive oncocytic neuroendocrine tumour ('on- copathologic study and review of the literature. mitive neuroectodermal tumors of the central Loda M, Andreini L, Basso G, et al. (1992). Lar-
cocytic paraganglioma') of the cauda equina. Cancer. 72(11):3277-81. PMID:8242554 nervous system. Neuro Oncol. 16(7):924-32. ge-cell medulloblastomas. A distinct variant with
Histopathology 10(3):311-9. PMID:2422107 790. Gaston-Massuet C, Andoniadou CL, Si- PMID:24470553 highly aggressive behavior. Am J Surg Pathol.
77 4. Gailani MR, Stahle-Backdahl M, Leff ell gnore M, Jayakody SA, Charolidi N, Kyeyune 807. Gessi M, Waha A, Setty P, Waha A, 16(7):687-93. PMID:1530108
DJ, Glynn M, Zaphiropoulos PG, Pressman R, et al. (2011). Increased Wingless (Wnt) si- Pietsch T (2011). Analysis of KIM1549-BRAF 822. Giangaspero F, Wellek S, Masuoka J, Ges-
C, et al. (1996). The role of the human homo- gnaling in pituitary progenitor/stem cells gives fusion status in a case of rosette-forming glio- si M, Kleihues P, Ohgaki H (2006). Stratification
logue of Drosophila patched in sporadic basal rise to pituitary tumors in mice and humans. neuronal tumor of the fourth ventricle (RGNT). of medulloblastoma on the basis of histopatho-
cell carcinomas. Nat Genet. 14(1 ):78-81. Proc Natl Acad Sci U S A 108(28):11482-7. Neuropathology. 31 (6):654-7. PMID:21518014 logical grading. Acta Neuropathol. 112(1 ):5-12.
PMID:8782823 PMID:21636786 808. Gessi M, Zur Miihlen A, Hammes J, Waha PMID:16685513
775. Gajjar A, Bhargava R, Jenkins JJ, Heide- 791. Gatta G, Botta L, Rossi S, Aareleid T, A, Denkhaus D, Pietsch T (2013). Genome-wi- 823. Giannini C, Fratkin JD, Wyatt-Ashmead
man R, Sanford RA, Langston JW, et al. (1995). Bielska-Lasota M, Clavel J, et al.; EUROCARE de DNA copy number analysis of desmoplastic J, Aleff PC (2014). Rhabdoid-like meningioma
Low-grade astrocytoma with neuraxis dissemi- Working Group (2014). Childhood cancer survi- infantile astrocytomas and desmoplastic infan- with inclusions consisting of accumulations of
nation at diagnosis. J Neurosurg. 83(1):67-71. val in Europe 1999-2007: results of EUROCA- tile gangfiogliomas. J Neuropathol Exp Neurol. complex interdigitating cell processes rather
PMID:7782852 RE-5-a population-based study. Lancet Oncol. 72(9):807-15. PMID:23965740 than intermediate filaments. Acta Neuropalhol.

368 References
127(6):937-9. PMID:24691783 M, Tan PH, Agaimy A, et al. (2014). Succinate A comparative study. J Neuropathol Exp Neural. Chan J, Dreyfuss JM, Park PJ, et al. (2010).
824. Giannini C, Hebrink D, Scheithauer BW, dehydrogenase (SDH)-deficient renal carci- 31(1):54-71. PMID:5060130 Genomic profiling reveals alternative genetic
Dei Tos AP, James CD (2001). Analysis of p53 noma: a morphologically distinct entity: a clini- 859. Goellner JR, Laws ER Jr, Soule EH, Oka- pathways of meningioma malignant progression
mulation and expression in pleomorphic xan- copathologic series of 36 tumors from 27 pa- zaki H (1978). Hemangiopericytoma of the dependent on the underlying NF2 status. Clin
thoastrocytoma. Neurogenetics. 3(3):159-62. tients. Am J Surg Pathol. 38(12):1588-602. meninges. Mayo Clinic experience. Am J Clin Cancer Res. 16(16):4155-64. PMID:20682713
PMID 11523567 PMID:25025441 Pathol. 70(3):375-80. PMID:707404 878. Gozali AE, Britt B, Shane L, Gonzalez I,
825. Giannini C, Scheithauer BW, Burger PC, 842. Gimm 0, Attie-Bitach T, Lees JA, Vekem- 860. Goerig M (1990). [Comments on the paper Gilles F, McComb JG, et al. (2012). Choroid
Brat DJ, Wollan PC, Lach B, et al. (1999). ans M, Eng C (2000). Expression of the PTEN by J.-P. Jantzen et al. An active charcoal filter plexus tumors; management, outcome, and
Pleomorphic xanthoastrocytoma: what do we tumour suppressor protein during human de- for removing volatile anestheticsj. Anaesthesist. association with the Li-Fraumeni syndrome:
really know about it? Cancer. 85(9):2033-45. velopment. Hum Mol Genet. 9(11):1633-9. 39(12):637-9. PMID:2073047 the Children's Hospital Los Angeles (CHLA)
PMID:10223246 PMID:10861290 861. Gold JS, Antonescu CR, Hajdu C, Ferrone experience, 1991-2010. Pediatr Blood Cancer.
826. Giannini C, Scheithauer BW, Burger PC, 843. Giorgianni A, Pellegrino C, De Benedictis CR, Hussain M, Lewis JJ, et al. (2002). Clinico- 58(6):905-9. PMID:21990040
Christensen MR, Wollan PC, Sebo T J, et al. A, Mercuri A, Baruzzi F, Minolta R, et al. (2013). pathologic correlates of solitary fibrous tumors. 879. Graadt van Roggen JF, McMenamin ME,
(1999). Cellular proliferation in pilocytic and dif- Lhermitte-Duclos disease. A case report. Neu- Cancer. 94(4):1057-68. PMID:11920476 Belchis DA, Nielsen GP, Rosenberg AE, Flet-
fuse astrocytomas. J Neuropathol Exp Neural. roradiol J. 26(6):655-60. PMID:24355184 862. Goldman JE, Corbin E (1991). Rosenthal cher CD (2001 ). Reticular perineurioma: a dis-
58(1):46-53. PMID:10068313 844. Gire J, Deveze A, Garcia S, Menelli C, fibers contain ubiquitinated alpha 8-crystallin. tinctive variant of soft tissue perineurioma. Am
827. Giannini C, Scheithauer BW, Jenkins RB, Curto CL, Tardivet L, et al. (2008). [Paragan- Am J Pathol. 139(4):933-8. PMID:1656764 J Surg Pathol. 25(4):485-93. PMID:11257623
Erlandson RA, Perry A, Borell TJ, et al. (1997). glioma of the cerebellopontine angle: report of 863. Gomez MR (1991). Phenotypes of the 880. Grabb PA, Albright AL, Pang D (1992). Dis-
Soft-tissue perineurioma. Evidence for an ab- two casesj. Rev Laryngol Otol Rhino! (Bord). tuberous sclerosis complex with a revision of semination of supratentorial malignant gliomas
normality of chromosome 22, criteria for diag- 129(3):213-6. PMID:19694167 diagnostic criteria. Ann NY Acad Sci. 615:1-7. via the cerebrospinal fluid in children. Neurosur-
nosis, and review of the literature. Am J Surg 845. Giuliani M, Galassi E, Zucchelli M, Volpi PMID:2039135 gery 30(1 ):64-71. PMID:1738457
Pathol. 21 (2):164-73. PMID:9042282 L (2005). Seizure outcome of lesionectomy in 864. Gonzalez KO, Noltner KA, Buzin CH, Gu 882. Graham DI, Lantos PL, editors. (2002).
828. Giannini C, Scheithauer BW, Lopes MB, glioneuronal tumors associated with epilepsy D, Wen-Fong CY, Nguyen VQ, et al. (2009). Greenfield's Neuropathology. 7th ed. London:
Hirose T, Kras JM, VandenBerg SR (2002). in children. J Neurosurg. 102(3) Suppl:288-93. Beyond Li Fraumeni Syndrome: clinical charac- Arnold.
lmmunophenotype of pleomorphic xanthoast- PMID:15881752 leristics of families with p53 germline mutations. 883. Grajkowska W, Kotulska K, Jurkiewicz E,
rocytoma. Am J Surg Pathol. 26(4):479-85. 846. Gjerris F, Agerlin N, 80rgesen SE, Buhl L, J Clin Oneal. 27(8):1250-6. PMID:19204208 Roszkowski M, Daszkiewicz P, Jozwiak S, et
PMID:11914626 Haase J, Klinken L, et al. (1998). Epidemiology 865. Gonzalez-Aguilar A, ldbaih A, Boisselier al. (2011 ). Subependymal giant cell astrocyto-
829. Giannini C, Scheithauer BW, Steinberg J, and prognosis in children treated for intracranial B, Habbita N, Rossetto M, Laurenge A, et al. mas wilh atypical histological features mimi-
Cosgrove T J (1998). lntraventricular perineurio- tumours in Denmark 1960-1984. Childs Nerv (2012). Recurrent mutations of MYD88 and cking malignant gliomas. Folia Neuropathol.
ma: case report. Neurosurgery. 43(6):1478-81, Syst. 14(7):302-11. PMID:9726580 TBL 1XR1 in primary central nervous system 49(1):39-46. PMID:21455842
discussion 1481-2. PMID:9848865 847. Gjerris F, Klinken L (1978). Long-term lymphomas. Clin Cancer Res. 18(19):5203-11. 884. Gramme! D, Warmuth-Metz M, van Bueren
830. Giannini C, Scheithauer BW, Weaver AL, prognosis in children with benign cerebellar PMID:22837180 AO, Kool M, Pietsch T, Kretzschmar HA, et al.
Burger PC, Kras JM, Mork S, et al. (2001 ). astrocytoma. J Neurosurg. 49(2):179-84. 866. Gonzalez-Campora R, Weller RO (1998). (2012). Sonic hedgehog-associated medullob-
Oligodendrogliomas: reproducibility and pro- PMID:671072 Lipidized mature neuroectodermal tumour lastoma arising from the cochlear nuclei of the
gnostic value of histologic diagnosis and gra- 848. Gteissner B, Chamberlain MC (2006). of the cerebellum with myoid differentiation. brainstem. Acta Neuropathol. 123(4):601-14.
ding. J Neuropathol Exp Neural. 60(3):248-62. Neoplastic meningitis. Lancet Neural. 5(5):443- Neuropathol Appl Neurobiol. 24(5):397-402. PMID:22349907
PMID:11245209 52. PMID:16632315 PMID:9821171 885. Grant JW, Steart PV, Aguui A, Jones
831. Gibson P, Tong Y, Robinson G, Thompson 849. Glick R, Baker C, Husain S, Hays A, 867. Goodrich LV, Milenkovic L, Higgins KM, DB, Gallagher PJ (1989). Gliosarcoma: an
MC, Currie OS, Eden C, et al. (2010). Subty- Hibshoosh H (1997). Primary melanocytomas Scott MP (1997). Altered neural cell fates and immunohistochemical study. Acta Neuropathol.
pes of medulloblastoma have distinct develop- of the spinal cord: a report of seven cases. Clin medulloblastoma in mouse patched mutants. 79(3):305-9. PMID:2609937
mental origins. Nature. 468(7327):1095-9. Neuropathol. 16(3):127-32. PMID:9197936 Science. 277(5329):1109-13. PMID:9262482 886. Gravendeel LA, Kouwenhoven MC, Ge-
PMID:21150899 850. Gliisker S, Bender BU, Apel TW, Natt E, 868. Goodrich LV, Scott MP (1998). Hedgehog vaert 0, de Rooi JJ, Stubbs AP, Duijm JE, et
832. Gielen GH, Gessi M, Buttarelli FR, Baldi van Velthoven V, Scheremet R, et al. (1999). and patched in neural development and disea- al. (2009). Intrinsic gene expression profiles
C, Hammes J, zur Muehlen A, et al. (2015). Ge- The impact of molecular genetic analysis of the se. Neuron. 21(6):1243-57. PMID:9883719 of gliomas are a better predictor of survival
netic Analysis of Diffuse High-Grade Astrocyto- VHL gene in patients with haemangioblastomas 869. Gorlin RJ (1987). Nevoid basal-cell than histology. Cancer Res. 69(23):9065-72.
mas in Infancy Defines a Novel Molecular Enti- of the central nervous system. J Neural Neuro- carcinoma syndrome. Medicine (Baltimore). PMID:19920198
ty. Brain Pathol. 25(4):409-17 PMID:25231549 surg Psychiatry. 67(6):758-62. PMID:10567493 66(2):98-113. PMID:3547011 887. Griffin CA, Burger P, Morsberger L, Yo-
833. Gielen GH, Gessi M, Hammes J, Kramm 851. Gliisker S, Bender BU, Apel TW, van 870. Gorovets D, Kannan K, Shen R, Kastenhu- nescu R, Swierczynski S, Weingart JD, et al.
CM, Waha A, Pietsch T (2013). H3F3A K27M Velthoven V, Mulligan LM, Zentner J, et al. ber ER, lslamdoust N, Campos C, et al. (2012). (2006). Identification of der(1;19)(q10;p10) in
mutation in pediatric CNS tumors: a marker for (2001 ). Reconsideration of biallelic inactivation IDH mutation and neuroglial developmental five oligodendrogliomas suggests mechanism
diffuse high-grade astrocytomas. Am J Clin Pa- of the VHL tumour suppressor gene in heman- features define clinically distinct subclasses of of concurrent 1 p and 19q loss. J Neuropathol
thol. 139(3):345-9. PMID:23429371 gioblastomas of the central nervous system. lower grade diffuse astrocytic glioma. Clin Can- Exp Neural. 65(10):988-94. PMID:17021403
834. Giese A, Bjerkvig R, Berens ME, Westphal J Neural Neurosurg Psychiatry. 70(5):644-8. cer Res. 18(9):2490-501. PMID:22415316 888. Grill J, Avet-Loiseau H, Lellouch-Tubiana
M (2003). Cost of migration: invasion of mali- PMID:11309459 871. Gorski GK, McMorrow LE, Donaldson A, sevenet N, Terrier-Lacombe MJ, Venuat
gnant gliomas and implications for treatment. 852. Gliisker S, Berlis A, Pagenstecher A, Vou- MH, Freed M (1992). Multiple chromosomal AM, et al. (2002). Comparative genomic hybri-
J Clin Oneal. 21(8):1624-36. PMID 12697889 gioukas VI, Van Velthoven V (2005). Characte- abnormalities in a case of craniopharyngio- dization detects specific cytogenetic abnorma-
835. Giese A, Loo MA, Tran N, Haskett D, rization of hemangioblastomas of spinal nerves. ma. Cancer Genet Cytogenet. 60(2):212-3. lities in pediatric ependymomas and choroid
Coons SW, Berens ME (1996). Dichotomy of Neurosurgery. 56(3):503-9, discussion 503-9. PMID:1606570 plexus papillomas. Cancer Genet Cytogenet.
astrocytoma migration and proliferation. Int J PMID:15730575 872. Goschzik T, Gessi M, Denkhaus D, Pielsch 136(2):121-5. PMID:12237235
Cancer. 67(2):275-82. PMID:8760599 853. Gliisker S, Kruger MT, Klingler JH, Wlodar- T (2014). PTEN mutations and activation of the 888A. Grimm T, Teglund S, Tackels D, Sangior-
836. Gil-Gouveia R, Cristino N, Farias JP, ski M, Klompen J, Schatlo B, et al. (2013). He- Pl3K/AktlmTOR signaling pathway in papillary gi E, Gurrieri F, Schwartz C, et al. (2001 ). Geno-
Trindade A, Ruivo NS, Pimentel J (2004). Ple- mangioblastomas and neurogenic polyglobulia. tumors of the pineal region. J Neuropathol Exp mic organization and embryonic expression of
omorphic xanthoastrocytoma of the cerebel- Neurosurgery. 72(6):930-5, discussion 935. Neural. 73(8):747-51. PMID:25003235 Suppressor of Fused, a candidate gene for the
lum: illustrated review. Acta Neurochir (Wien). PMID:23407287 873. Gossage L, Eisen T, Maher ER (2015). split-hand/split-foot malformation type 3. FESS
146(11 ):1241-4. PMID:15455217 854. Gliisker S, Li J, Xia JB, Okamoto H; Zeng VHL, the story of a tumour suppressor gene. Lett. 505(1):13-7. PMID:11557033
837. Gilbert MR, Dignam JJ, Armstrong TS, W, Lanser RR, et al. (2006). Hemangioblasto- Nat Rev Cancer. 15(1):55-64. PMID:25533676 889. Grois N, Fahrner B, Arceci RJ, Henter JI,
Wefel JS, Blumenthal OT, Vogelbaum MA, et al. mas share protein expression with embryonal 874. Goto J, Talas OM, Klein P, Qin W, Che- McClain K, Lassmann H, Nanduri V, Prosch
(2014). A randomized trial of bevacizumab for hemangioblast progenitor cell. Cancer Res. kaluk YI, Anderl S, et al. (2011). Regulable H, Prayer D, (2010). Central nervous system
newly diagnosed glioblastoma. N Engl J Med. 66(8):4167-72. PMID:16618738 neural progenitor-specific Tsc1 loss yields giant disease in Langerhans cell histiocytosis J
370(8):699-708. PMID:24552317 855. Gliisker S, Van Velthoven V (2005). Risk of cells with organellar dysfunction in a model of Pediatr 156: 873-81, 881.e1. PMID:20434166
838. Gilbert MR, Wang M, Aldape KO, Stupp R, hemorrhage in hemangioblastomas of the cen- tuberous sclerosis complex. Proc Natl Acad Sci 890. Grosse G, Lindner G, Matthies HJ (1976).
Hegi ME, Jaeckle KA, et al. (2013). Dose-dense lral nervous system. Neurosurgery. 57(1 ):71-6, US A. 108(45):E1070-9. PMID:22025691 [Effect of orotic acid on the in vitro cultured ner-
temozolomide for newly diagnosed glioblasto- discussion 71-6. PMID:15987542 875. Gould VE, Rorke LB, Jansson OS, Mo- ve tissuej. Z Mikrosk Anal Forsch. 90(3):499-
ma: a randomized phase Ill clinical trial. J Clin 856. Godfraind C (2009). Classification and lenaar WM, Trojanowski JQ, Lee VM, et al. 506. PMID:1088565
Oneal. 31(32):4085-91. PMID:24101040 controversies in pathology of ependymo- (1990). Primitive neuroectodermal tumors of 891. Filbin MG, Dabral SK, Pazyra-Murphy MF,
839. Gilbertson RJ, Ellison OW (2008). The mas. Childs Nerv Syst. 25(10):1185-93. lhe central nervous system express neuroen- Ramkissoon S, Kung AL, Pak E, et al. (2013).
origins of medulloblastoma subtypes. Annu Rev PMID:19212775 docrine markers and may express all classes of Coordinate activation of Shh and Pl3K signa-
Pathol. 3:341-65. PMID:18039127 857. Godfraind C, Kaczmarska JM, Kocak intermediate filaments. Hum Pathol. 21(3):245- ling in PTEN-deficient glioblastoma: new thera-
840. Gill AJ, Benn DE, Chou A, Clarkson A, M, Dalton J, Wright KO, Sanford RA, et al. 52. PMID:2155868 peutic opportunities. Nat Med. 19(11):1518-23.
MuljonoA, Meyer-Rochow GY, et al. (2010). lrn- (2012). Distinct disease-risk groups in pedia- 876. Goutagny S, Nault JC, Mallet M, Henin D, PMID:24076665
munohistochemistry for SDHB triages genetic tric supratentorial and posterior Iossa epen- Rossi JZ, Kalamarides M (2014). High inciden- 892. Gu J, Tamura M, Yamada KM (1998).
testing of SDHB, SDHC, and SOHO in parag- dymomas. Acta Neuropathol. 124(2):247-57. ce of activating TERT promoter mutations in Tumor suppressor PTEN inhibits integrin- and
anglioma-pheochromocytoma syndromes. Hum PMID:22526017 meningiomas undergoing malignant progressi- growth factor-mediated mitogen-activated pro-
Pathol. 41 (6):805-14. PMID:20236688 858. Goebel HH, Cravioto H (1972). Ultrastruc- on. Brain Pathol. 24(2):184-9. PMID:24261697 tein (MAP) kinase signaling pathways. J Cell
841. Gill AJ, Hes 0, Papathomas T, Sedivcova ture of human and experimental ependymomas. 877. Goutagny S, Yang HW, Zucman-Rossi J, Biol. 143(5):1375-83. PMID:9832564

References 369
893. Guan H, Huang Y, Wen W, Xu M, Zan astrocytoma. Genome Res. 23(3):431-9. 925. Hainaut P, Olivier M, Wiman K (2013).p53 G, Piotrowski A, Hellstrom AR: Mantripragada
Q, Zhang Z (2011 ). Primary central nervous PMID:23222849 in the Clinics. New York: Springer. K, et al. (2007). Comprehensive genetic and
system extranodal NK/T-cell lymphoma, nasal 910. Gutmann DH, Parada LF, Silva AJ, Ratner 926. Hair LS, Symmans F, Powers JM, Carmel epigenetic analysis of sporadic meningioma for
type: case report and review of the literature. N (2012). Neurofibromatosis type 1: modeling P (1992). lmmunohistochemistry and prolifera- macro-mutations on 22q and micro-mutations.
J Neurooncol. 103(2):387-91. PMID:20845062 CNS dysfunction. J Neurosci. 32(41):14087- tive activity in Lhermitte-Duclos disease. Acta within the NF2 locus. BMC Genomics. 8:16.
894. Gucer H, Mete O (2014). Endobronchial 93. PMID:23055477 Neuropalhol. 84(5):570-3. PMID:1462769 PMID 17222329 ···
gangliocytic paraganglioma: not all keratin-po- 911. Gutmann DH, Wood DL, Collins FS 927. Hajnsek S, Paladino J, Gadze ZP, Nan- 943. Hao C, Beguinot F, Condorelli G, Trencia
sitive endobronchial neuroendocrine neoplas- (1991). Identification of the neurofibromatosis kovic S, Mrak G, Lupret V (2013). Clinical A, Van Meir EG, Yong VW, et al. (2001). lnduc-
ms are pulmonary carcinoids. Endocr Pathol. type 1 gene product. Proc Natl Acad Sci U SA. and neurophysiological changes in patients tion and intracellular regulation of tumor necro,
25(3):356-8. PMID:23912549 88(21):9658-62. PMID:1946382 with pineal region expansions. Coll Antropol. sis factor-related apoptosis-inducing ligand
895. Guermazi A, De Kerviler E, Zagdanski 912. Gyure KA, Morrison AL (2000). Cytokeratin 37(1):35-40. PMID:23697248 (TRAIL) mediated apotosis in human malig-
AM, Frija J (2000). Diagnostic imaging of cho- 7 and 20 expression in choroid plexus tumors: 928. Haliloglu G, Jobard F, Oguz KK, Anlar B, nant glioma cells. Cancer Res. 61(3):1162-70.
roid plexus disease. Clin Radial. 55(7):503-16. utility in differentiating these neoplasms from Akalan N, Coskun T, et al. (2008). L-2-hydroxy- PMID:11221847
PMID:10924373 metastatic carcinomas. Mod Pathol. 13(6):638- glularic aciduria and brain tumors in children 944. Hardee ME, Zagzag D (2012). Mechanis-
896. Guesmi H, Houtteville JP, Courtheoux P, 43. PMID:10874668 with mutations in the L2HGDH gene: neuroima- ms of glioma-associated neovascularization
Derion JM, Chapon F (1999). IDysembryopla- 913. Gyure KA, Prayson RA (1997). Subepen- ging findings. Neuropediatrics. 39(2):119-22. Am J Pathol. 181(4):1126-41. PMID:22858156.
stic neuroepithelial tumors. Report of 8 cases dymal giant cell astrocytoma: a clinicopatholo- PMID:18671189 945. Harder A, Wesemann M, Hagel C, Schit-
including two with unusual localization]. Neuro- gic study with HMB45 and MIB-1 immunohis- 929. Haller F, Bieg M, Moskalev EA, Barthel- tenhelm J, Fischer S, Tatagiba M, el al. (2012).
chirurgie. 45(3):190-200. PMID:10567958 tochemical analysis. Mod Pathol. 10(4):313-7. meB S, Geddert H, Boltze C, et al. (2015). Hybrid neurofibroma/schwannoma is over-
897. Gunny RS, Hayward RD, Phipps KP, Har- PMID:9110292 Recurrent mutations within the amino-terminal represented among schwannomalosis and
ding BN, Saunders DE (2005). Spontaneous 914. Gotze S, Wolter M, Reifenberger G, Muller region of !3-catenin are probable key molecular neurofibromatosis patients. Am J Surg Pathol...
regression of residual low-grade cerebellar pi- 0, Sievers S (2010). Frequent promoter hyper- driver events in sinonasal hemangiopericytoma. 36(5):702-9. PMID:22446939
locytic astrocytomas in children. Pediatr Radial. methylation of Wnt pathway inhibitor genes in Am J Pathol. 185(2):563-71. PMID:25482924 946. Haroche J, Charlotte F, Arnaud L, von
35(11 ):1086-91. PMID 16047140 malignant astrocytic gliomas. Int J Cancer. 930. Haller F, Moskalev EA, Faucz FR, Bart- Deimling A, Helias-Rodzewcz Z, Hervier B et
898. Guppy KH, Akins PT, Moes GS, Prados 126(11):2584-93. PMID:19847810 helrnels S, Wiemann S, Bieg M, et al. (2014). al. (2012). High prevalence of BRAF V600E
MD (2009). Spinal cord oligodendroglioma with 915. Haapasalo H, Isola J, Sallinen P, Kalima Aberrant DNA hypermethylation of SDHC: a mutations in Erdheim-Chester disease but nor"
1 p and 19q deletions presenting with cerebral H, Helin H, Ranta/a I (1993). Aberrant p53 ex- novel mechanism of tumor development in Car- in other non-Langerhans cell histiocytoses.
oligodendrogliomatosis. J Neurosurg Spine. pression in astrocytic neoplasms of the brain: ney triad Endocr Reial Cancer. 21(4):567-77. Blood. 120(13):2700-3. PMID:22879539
10(6):557-63. PMID:19558288 association with proliferation. Am J Pathol. PMID:24859990 947. Haroche J, Cohen-Aubart F, Emile JF
899. Gurney JG, Smith MA, Bunin GR CNS and 142(5):1347-51. PMID:7684193 931. Halling KC, Scheithauer BW, Halling AC, Maksud P, Drier A, Toledano D, et al. (2015i'
miscellaneous intracranial and intraspinal neo- 916. Haas JE, Palmer NF, Weinberg AG, Beck- Nascimento AG, Ziesmer SC, Roche PC, et Reproducible and sustained efficacy of tar-
plasms. In: Ries LAG, Smith MA, Gurney JG, with JB (1981). Ultrastructure of malignant rh- al. (1996). p53 expression in neurofibroma geted therapy with vemurafenib in patients
Linet M, Tamra T, Young JL, Bunin GR, editors abdoid tumor of the kidney. A distinctive renal and malignant peripheral nerve sheath tumor. with BRAF(V600E)-mutated Erdheim-Ches-
(1995).Cancer incidence and survival among tumor of children. Hum Pathol. 12(7):646-57. An immunohistochemical study of sporadic ter disease. J Clin Oneal. 33(5):411-8.
children and adolescents United States SEER PMID 7275104 and NF1-associated tumors. Am J Clin Palhol. PMID:25422482
Program 1975-1995. NIH Pub no. 99-4649 917. Haberler C, Jarius C, Lang S, Rossler K, 106(3):282-8. PMID:8816583 948. Harris CP, Townsend JJ, Brockmeyer
Bethesda, MD: National Cancer Institute. Gruber A, Hainfellner JA, et al. (2002). Fibrous 932. Hama S, Arita K, Nishisaka T, Fukuha- DL, Heilbrun MP (1991). Cerebral granular
900. Gusella JF, Ramesh V, MacCollin M, Ja- meningeal tumours with extensive non-cal- ra T, Tominaga A, Sugiyama K, et al. (2002). cell tumor occurring with glioblastoma multi-
coby LB (1996). Neurofibromatosis 2: loss of cifying collagenous whorls and glial fibrillary Changes in the epithelium of Rathke cleft cyst forme: case report. Surg Neural. 36(3):202-6.
merlin's protective spell. Curr Opin Genet Dev. acidic protein expression: the whorling-sclero- associated with inflammation. J Neurosurg. PMID: 1652163
6(1):87-92. PMID:8791482 sing variant of meningioma. Neuropathol Appl 96(2):209-16. PMID:11838792 949. Harter DH, Omeis I, Forman S, Braun A
901. Gusella JF, Ramesh V, MacCollin M, Ja- Neurobiol. 28(1):42-7. PMID:11849562 933. Hamasaki T, Yamada K, Kuratsu J (2013). (2006). Endoscopic resection of an inlraventri-
coby LB (1999). Merlin: the neurofibromatosis 918. Haberler C, Laggner U, Slave I, Czech Seizures as a presenting symptom in neurosur- cular dysembryoplastic neuroepithelial tumor
2 tumor suppressor. Biochim Biophys Acta. T, Ambros IM, Ambros PF, et al. (2006). lm- gical patients: a retrospective single-institution of the septum pellucidum. Pediatr Neurosurg.
1423(2):M29-36. PMID:10214350 munohistochemical analysis of INl1 protein in analysis. Clin Neural Neurosurg. 115(11):2336- 42(2):105-7.PM/016465080
902. Gutenberg A, Brandis A, Hong B, Guna- malignant pediatric CNS tumors: Lack of INl1 40. PMID:24011499 950. Hartmann C, Hentschel B, Simon M, West-
wan B, Enders C, Schaefer IM, et al. (2011). in atypical teratoid/rhabdoid tumors and in a 934. Hamazaki S, Nakashima H, Matsumoto phal M, Schackert G, Tonn JC, et al.; German·
Common molecular cytogenetic pathway in pa- fraction of primitive neuroectodermal tumors K, Taguchi K, Okada S (2001). Metastasis of Glioma Network (2013). Long-term survival in
pillary tumors of the pineal region (PTPR). Brain without rhabdoid phenotype. Am J Surg Pathol. renal cell carcinoma to central nervous system primary glioblastoma with versus without iso-
Pathol. 21(6):672-7. PMID:21470326 30(11):1462-8. PMID:17063089 hemangioblastoma in two patients with van Hip- citrate dehydrogenase mutations. Clin Cancer
903. Guthrie BL, Ebersold MJ, Scheithauer BW, 919. Haddad SF, Hitchon PW, Godersky JC pel-Lindau disease. Pathol Int. 51(12):948-53. Res. 19(18):5146-57. PMID:23918605
Shaw EG (1989). Meningeal hemangiopericyto- (1991). Idiopathic and glucocorticoid-induced PMID:11844068 951. Hartmann C, Hentschel B, Tatagiba
ma: histopathological features, treatment, and spinal epidural lipomatosis. J Neurosurg. 935. Hamilton JD, Rapp M, Schneiderhan T, M, Schramm J, Schnell 0, Seidel C, et al.;
long-term follow-up of 44 cases. Neurosurgery. 74(1)38-42. PMID:1984504 Sabel M, Hayman A, Scherer A, et al. (2014). German Glioma Network (2011 ). Molecular
25(4):514-22. PMID:2797389 920. Haddad SF, Moore SA, Schelper RL, Glioblastoma multiforme metastasis outside the markers in low-grade gliomas: predictive or
904. Gutman DA, Cooper LA, Hwang SN, Hol- Goeken JA (1992). Vascular smooth muscle CNS: three case reports and possible mecha- prognostic? Clin Cancer Res. 17(13):4588-99.
der CA, Gao J, Aurora TD, et al. (2013). MR hyperplasia underlies the formation of glomeru- nisms of escape. J Clin Oneal. 32(22):e80-4. PMID:21558404
imaging predictors of molecular profile and sur- loid vascular structures of glioblastoma multifor- PMID:24567 434 952. Hartmann C, Hentschel B, Wick W, Capper
vival: multi-institutional study of the TCGA gli- me. J Neuropathol Exp Neural. 51(5):488-92. 936. Hamilton RL, Pollack IF (1997). The mo- D, Felsberg J, Simon M, et al. (2010). Patients
oblastoma data set. Radiology. 267(2):560-9. PMID:1381413 lecular biology of ependymomas. Brain Palhol. with IDH1 wild type anaplastic astrocytomas
PMID:23392431 921. Hadfield KO, Newman WG, Bowers NL, 7(2):807-22. PMID:9161731 exhibit worse prognosis than IDH1-mutated
905. Gutmann DH, Aylsworth A, Carey JC, Korf Wallace A, Bolger C, Colley A, et al. (2008). 937. Hamilton SR, Liu B, Parsons RE, Papad- glioblastomas, and IDH1 mutation status ac-
B, Marks J, Pyeritz RE, et al. (1997). The diag- Molecular characterisation of SMARCB1 and opoulos N, Jen J, Powell SM, et al. (1995). The counts for the unfavorable prognostic effect
nostic evaluation and multidisciplinary manage- NF2 in familial and sporadic schwannomatosis. molecular basis of T urcot's syndrome. N Engl J of higher age: implications for classification of
ment of neurofibromatosis 1 and neurofibromat- J Med Genet. 45(6):332-9. PMID:18285426 Med. 332(13):839-47. PMID:7661930 gliomas. Acta Neuropathol. 120(6):707-18.
osis 2. JAMA. 278(1):51-7. PMID:9207339 922. Hadfield KO, Smith MJ, Urquhart JE, 938. Han L, Niu H, Wang J, Wan F, Shu K, Ke PMID:21088844
906. Gutmann DH, Collins FS (1993). The Wallace AJ, Bowers NL, King AT, et al. (2010). C, et al. (2013). Extraventricular neurocytoma in 953. Hartmann C, Meyer J, Balss J, Capper
neurofibromatosis type 1 gene and its protein Rates of loss of heterozygosity and mitotic re- pediatric populations: A case report and review D, Mueller W, Christians A, et al. (2009). Type
product, neurofibromin. Neuron. 10(3):335-43. combination in NF2 schwannomas, sporadic of the literature. Oneal Lett. 6(5):1397-405. and frequency of IDH1 and IDH2 mutations
PMID:8461130 vestibular schwannomas and schwannomato- PMID:24179531 are related to astrocytic and oligodendroglial
907. Gutmann DH, Donahoe J, Perry A, Lemke sis schwannomas. Oncogene. 29(47):6216-21. 939. Han SJ, Yang I, Otero JJ, Ahn BJ, Tihan T, differentiation and age: a study of 1,010 diffu-
N, Gorse K, Kittiniyom K, et al. (2000). Loss of PMID:20729918 McDermott MW, et al. (2010). Secondary glios- se gliomas. Acta Neuropathol. 118(4):469-74.
DAL-1, a protein 4.1-related tumor suppressor, 923. Hagel C, Stemmer-Rachamimov AO, Bor- arcoma after diagnosis of glioblastoma: clinical PMID:19554337
is an important early event in the pathogenesis nemann A, Schuhmann M, Nagel C, Huson S, experience with 30 consecutive patients. J Neu- 954. Hartmann C, Siebems J, Gehlhaar C, Si-
of meningiomas. Hum Mal Genet. 9(10):1495- et al. (2012). Clinical presentation, immunohis- rosurg. 112(5):990-6. PMID:19817543 mon M, Paulus W, von Deimling A (2006). NF2
500. PMID:10888600 tochemistry and electron microscopy indicate 940. Hannan F, Ho I, Tong JJ, Zhu Y, Nurnberg mutations in secretory and other rare variants
908. Gutmann DH, James CD, Poyhonen M, neurofibromatosis type 2-associated gliomas P, Zhong Y (2006). Effect of neurofibromatosis of meningiomas. Brain Pathol. 16(1):15-9.
Louis ON, Ferner R, Guha A, et al. (2003). to be spinal ependymomas. Neuropathology. type I mutations on a novel pathway for ade- PMID:16612978
Molecular analysis of astrocytomas presenting 32(6):611-6. PMID:22394059 nylyl cyclase activation requiring neurofibromin 955. Hartmann C, Xu X, Bartels G, Holtkamp N,
after age 10 in individuals with NF1. Neurology. 924. Hahn H, Wicking C, Zaphiropoulous PG, and Ras. Hum Mal Genet. 15(7):1087-98. Gonzales IA, Tallen G, et al. (2004). Pdgfr-alpha
61(10):1397-400. PMID:14638962 Gailani MR, Shanley S, Chidambaram A, et PMID:16513807 in 1 p/19q LOH oligodendrogliomas. Int J Can-
909. Gutmann DH, Mclellan MD, Hussain I, al. (1996). Mutations of the human homolog 941. Hanssen AM, Fryns JP (1995). Cow- cer. 112(6):1081-2. PMID:15386438
Wallis JW, Fulton LL, Fulton RS, et al. (2013). of Drosophila patched in the nevoid basal cell den syndrome. J Med Genet. 32(2):117-9. 956. Hashimoto T, Sasagawa I, lshigooka M,
Somatic neurofibromatosis type 1 (NF1) inac- carcinoma syndrome. Cell. 85(6):841-51. PMID:7760320 Kubota Y, Nakada T, Fujita T, et al. (1995).
tivation characterizes NF1-associated pilocytic PMID:8681379 942. Hansson CM, Buckley PG, Grigelioniene Down's syndrome associated with intracranial

370 References
germinoma and testicular embryonal carcino- growth factors and receptors in capillary he- extragonadal germ cell tumor. Cancer Genet progressive, therapeutically targetable disease.
ma. Ural Int. 55(2):120-2. PMID:8533196 mangioblastomas and hemangiopericytomas. Cytogenet. 57(1):41-6. PMID:1756483 Cancer Cell. 27(1):72-84. PMID:25533335
957. Hasselblatt M, Blurncke I, Jeibmann A, Am J Pathol. 148(3):76:,...75. PMID:8774132 988. Helseth A, Helseth E, Unsgaard G (1989). 1004. Hiniker A, Hagenkord JM, Powers MP,
Rickert CH, Jouvet A, van de Nes JA, et al. 972. Hayasaka K, Nihashi T, Takebayashi Primary meningeal melanoma. Acta Oneal. Aghi MK, Prados MD, Perry A (2013). Gliosar-
(2006). lmmunohistochemical profile and chro- S, Bundoh M (2008). FOG PET in Lhermit- 28(1):103-4. PMID:2706127 coma arising from an oligodendroglioma (oli-
mosomal imbalances in papillary tumours of te-Duclos disease. Clin Nucl Med. 33(1):52-4. 989. Hemminki K, Kyyronen P, Vaittinen P gosarcoma). Clin Neuropathol. 32(3):165-70.
the pineal region. Neuropathol Appl Neurobiol. PMID:18097262 (1999). Parental age as a risk factor of child· PMID:23254140
32(3):278-83. PMID:16640646 973. Hayashi K, Ohara N, Jeon HJ, Akagi S, Ta- hood leukemia and brain cancer in offspring. 1005. Hinkes BG, von Hoff K, Deinlein F, War-
958. Hasselblatt M, Bohm C, Tatenhorst L, kahashi K, Akagi T, et al. (1993). Gliosarcoma Epidemiology. 10(3):271-5. PMID:10230837 muth-Metz M, Soerensen N, Timmermann B, et
Dinh V, Newrzella D, Keyvani K, et al. (2006). with features of chondroblastic osteosarcoma. 990. Hemminki K, Liu X, Forsti A, Ji J, Sundquist al. (2007). Childhood pineoblastoma: experien-
Identification of novel diagnostic markers for Cancer. 72(3):850-5. PMID:8334639 J, Sundquist K (2013). Subsequent brain tu- ces from the prospective multicenter trials HIT·
choroid plexus tumors: a microarray-based 974. Hayostek CJ, Shaw EG, Scheithauer B, mors in patients wilh autoimmune disease. SKK87, HIT-SKK92 and HIT91. J Neurooncol.
approach. Am J Surg Palhol. 30(1):66-74. O'Fallon JR, Weiland TL, Schomberg PJ, et al. Neuro Oneal. 15(9):1142-50. PMID:23757294 81(2):217-23. PMID:16941074
PMID:16330944 (1993). Astrocytomas of the cerebellum. A com- 991. Hennessy MJ, Elwes RD, Rabe-Hesketh 1006. Hirbe AC, Gutmann DH (2014). Neuro-
959. Hasselblatt M, Gesk S, Oyen F, Rossi parative clinicopathologic study of pilocytic and S, Binnie CD, Pelkey CE (2001). Prognostic fibromatosis type 1: a multidisciplinary appro-
S, Viscardi E, Giangaspero F, et al. (2011). diffuse astrocytomas. Cancer. 72(3):856-69. factors in the surgical treatment of medically ach to care. Lancet Neural. 13(8):834-43.
Nonsense mutation and inactivation of SMAR- PMID:8334640 intractable epilepsy associated with mesial tern- PMID:25030515
CA4 (BRG1) in an atypical teratoid/rhabdoid 975. He J, Mokhtari K, Sanson M, Marie Y, Ku- poral sclerosis. Acta Neural Scand. 103(6):344-- 1007. Hirose T, Giannini C, Scheithauer BW
tumor showing retained SMARCB1 (INl1) jas M, Huguet S, et al. (2001). Glioblastomas 50. PMID:11421846 (2001 ). Ultrastructural features of pleomorphic
expression. Am J Surg Pathol. 35(6):933-5. with an oligodendroglial component: a patholo- 992. Henske EP, Wessner LL, Golden J, Scheit- xanthoastrocytoma: a comparative study with
PMID:21566516 gical and molecular study. J Neuropathol Exp hauer BW, Vortmeyer AO, Zhuang Z, et al. glioblastoma multiforrne. Ultrastruct Pathol.
960. Hasselblatt M, Jeibmann A, Guerry M, Neural. 60(9):863-71. PMID:11556543 { 1997). Loss of tuberin in both subependymal 25(6):469-78. PMID:11783911
Senner V, Paulus W, Mclendon RE (2008). 976. He MX, Wang JJ (2011). Rhabdoid gli- giant cell astrocytomas and angiomyolipomas 1008. Hirose T, Scheithauer BW (1998). Mixed
Choroid plexus papilloma with nsuropil-li- oblastoma: case report and literature review. supports a two-hit model for the pathogenesis dysembryoplastic neuroepithelial tumor and
ke islands. Am J Surg Pathol. 32(1):162-6. Neuropathology. 31 (4):421-6. PMID:21092062 of tuberous sclerosis tumors. Am J Pathol. ganglioglioma. Acta Neuropathol. 95(6):649-
PMID:18162784 977. Heaphy CM, de Wilde RF, Jiao Y, Klein AP, 151(6) 1639-47. PMID:9403714 54. PMID:9650758
961. Hasselblatt M, Muhlisch J, Wrede 8, Kai- Edil BH, Shi C, et al. (2011). Altered lelomeres 993. Henson JW, Schnitker BL, Correa KM, van 1009. Hirose T, Scheithauer BW, Lopes MB,
linger B, Jeibmann A, Peters 0, et al. (2009). in tumors wilh ATRX and DAXX mutations. Deimling A, Fassbender F. Xu HJ, et al. (1994). Gerber HA, Altermatt HJ, Hukee MJ, et al.
Aberrant MGMT (06-methylguanine-DNA Science. 333(6041):425. PMID:21719641 The retinoblastoma gene is involved in malig- (1995). Tuber and subependymal giant cell as-
methyltransferase) promoter methylation 978. Heath JA, Ng J, Beshay V, Coleman nant progression of astrocytomas. Ann Neural. trocytoma associated with tuberous sclerosis:
in choroid plexus tumors. J Neurooncol. L, Lo P, Amor DJ (2013). Anaplastic oligo- 36(5):714-21. PMID:7979217 an immunohistochemical, ultrastructural, and
91(2)151-5. PMID:18795231 dendroglioma in an adolescent with Lynch 994. Herpers MJ, Budka H (1984). Glial fibrillary immunoelectron and microscopic study. Acta
962. Hasselblatt M, Nagel I, Oyen F, Bartelheim syndrome. Pediatr Blood Cancer. 60(6):E1:,...5_ acidic protein (GFAP) in oligodendroglial tu- Neuropathol. 90(4):387-99. PMID:8546029
K, Russell RB, Schuller U, et al. (2014). SMAR- PMID:23255519 mors: gliofibrillary oligodendroglioma and Iran· 1010. Hirose T, Scheithauer BW, Lopes MB,
CA4-mutated atypical teratoid/rhabdoid tumors 979. Hebert-Blouin MN, Scheithauer BW, Amra- silional oligoastrocytoma as subtypes of oligo- Gerber HA, Altermatt HJ, VandenBerg SR
are associated with inherited germline altera- mi KK, Durham SR, Spinner RJ (2012). Fibro- dendroglioma. Acta Neuropathol. 64(4):265-72. (1997). Ganglioglioma: an ultrastructural
tions and poor prognosis. Acla Neuropathol. matosis: a potential sequela of neuromuscular PMID:6391068 and immunohistochemical study. Cancer.
128(3):453-6. PMID:25060813 choristoma. J Neurosurg. 116(2):399-408. 995. Herpers MJ, Ramaekers FC, Aldeweireldt 79(5):989-1003. PMID:9041162
963. Hasselblatt M, Nolte KW, Paulus W PMID:21819193 J, Moesker 0, Slooff J (1986). Co-expression 1011. Hirose T, Scheithauer BW, Sano T
(2004). Angiomatous meningioma: a clinicopa- 980. Heegaard S, Sommer HM, Broholm H, of glial fibrillary acidic protein- and virnen- (1998). Perineurial malignant peripheral nerve
lhologic study of 38 cases. Am J Surg Pathol. Broendstrup O (1995). Proliferating cell nuc- tin-type intermediate filaments in human ast- sheath tumor (MPNST): a clinicopathologic, im-
28(3):390-3. PMID:15104303 lear antigen and Ki-67 immunohistochemistry rocytomas. Acta Neuropathol. 70(3-4):333-9. munohistochemical, and ultraslructural study of
964. Hasselblatt M, Oyen F, Gesk S, Kordes U, of oligodendrogliomas with special referen- PMID:3020864 seven cases. Am J Surg Pathol. 22(11):1368-
Wrede B, Bergmann M, et al. (2009). Cribriform ce to prognosis. Cancer. 76(10):1809-13. 996. Herregodts P, Vloeberghs M, Schmedding 78. PMID:9808129
neuroepithelial tumor (CRINET): a nonrhabdoid PMID:8625052 E, Goossens A, Stadnik T, D'Haens J (1991). 1012. Hirose T, Tani T, Shimada T, lshizawa K,
ventricular lumor with IN11 loss and relatively 981. Heesters M, Molenaar W, Go GK (2003). Solilary dorsal intramedullary schwannoma. Shimada S, Sano T (2003). lmmunohistochemi-
favorable prognosis. J Neuropathol Exp Neural. Radiotherapy in supratentorial gliomas. A study Case report. J Neurosurg. 74(5):816-20. cal demonstration of EMA/Glut1 -positive peri-
68(12):1249-55. PMID:19915490 of 821 cases. Strahlenther Onkol. 179(9):606- PMID:2013780 neurial cells and CD34-positive fibroblastic cells
965. Hasselblatt M, Paulus W (2003). Sensiti- 14. PMID:14628126 997. Herrick MK, Rubinstein LJ (1979). The in peripheral nerve sheath tumors. Mod Pathol.
vity and specificity of epithelial membrane an- 982. Hegi ME, Diserens AC, Gorlia T, Hamou cytological differentiating potential of pineal 16(4):293-8. PMID:12692193
tigen staining patterns in ependymomas. Acta MF, de Tribolet N, Weller M, et al. (2005). parenchymal neoplasms (true pinealomas). A 1013. Hiroyuki M, Ogino J, Takahashi A, Hase-
Neuropathol. 106(4):385--8 PMID:12898159 MGMT gene silencing and benefit from te- clinicopathological study of 28 tumours . Brain gawa T, Wakabayashi T (2015). Rhabdoid gli·
965A. Hasselblatt M, Riesmeier 8, Lechtape mozolomide in glioblastoma. N Engl J Med. 102: 289-320. PMID:88244 oblastoma: an aggressive variaty of astrocytic
B, Brentrup A, Slummer W, Albert FK, et al. 352(10):997-1003. PMID:15758010 998. Hertzler DA 2nd, DePowell JJ, Stevenson tumor. Nagoya J Med Sci. 77(1-2):321--8.
(2011). BRAF-KIM1549 fusion transcripts are 983. Hegi ME, Janzer RC, Lambiv WL, Gorlia T, CB, Mangano FT (2010). Tethered cord syn- PMID:25797998
less frequent in pilocytic astrocytomas diag- Kouwenhoven MC, Hartmann C, et al.; Europe- drome: a review of the literature from embryo· 1014. Hitotsumatsu T, lwaki T, Kitamoto T, Mi-
nosed in adults. Neuropathol Appl Neurobiol. an Organisation tor Research and Treatment of logy to adult presentation. Neurosurg Focus. zoguchi M, Suzuki SO, Hamada Y, et al. (1997).
37(7):803-6. PMID21696415 Cancer Brain Tumour and Radiation Oncology 29(1):E1. PMID:20593997 Expression of neurofibromatosis 2 protein in
966. Hasselblatt M, Sepehrnia A, von Falken- Groups; National Cancer Institute of Canada 999. Herva R, Serio W, Laitinen J, Becker LE human brain tumors: an immunohistochemi-
hausen M, Paulus W (2003). lnlracranial follicu- Clinical Trials Group (2012). Presence of an (1996). lntraventricular rhabdomyosarcoma cal study. Acta Neuropathol. 93(3):225--32.
lar dendritic cell sarcoma. Case report. J Neuro- oligodendroglioma-like component in newly after resection of hyperplastic choroid plexus. PMID:9083553
surg. 99(6):1089-90. PMID:14705740 diagnosed glioblastoma identifies a pathoge- Acta Neuropathol. 92(2):213-6. PMID:8841669 1015. Hjalmars U, Kulldorff M, Wahlqvist Y,
967. Hassoun J, Gambarelli D, Grisoli F, Pellet netically heterogeneous subgroup and lacks 1000. Hewer E, Beck J, Murek M, Kappeler A, Lannering B (1999). Increased incidence ra-
W, Salamon G, Pellissier JF, et al. (1982). Cent- prognostic value: central pathology review of Vassella E, Vajtai I (2014). Polymorphous oligo- tes but no space-time clustering of childhood
ral neurocytoma. An electron-microscopic study lhe EORTC_26981/NCIC_CE.3 trial. Acta·Neu- dendroglioma of Zulch revisited: a genetically astrocytoma in Sweden, 1973-1992: a popu-
of two cases. Acta Neuropalhol. 56(2):151-6. ropathol. 123(6):841-52. PMID:22249618 heterogeneous group of anaplastic gliomas lation-based study of pediatric brain tumors.
PMID:7064664 984. Heim S, Beschorner R, Mittelbronn M, including tumors of bona fide oligodendroglial Cancer. 85(9):2077-90. PMID:10223251
968. Hassoun J, Gambarelli D, Peragut JC, Keyvani K, Riemenschneider MJ, Vajtai I, et differentiation. Neuropathology. 34(4):323-32. 1016. Ho OM, Hsu CY, Wong TT, Ting LT, Ch·
Toga M (1983). Specific ultrastructural mar- al. (2014). Increased mitotic and proliferative PMID:24444336 iang H (2000). Atypical teratoidlrhabdoid tumor
kers of human pinealomas. A study of tour activity are associated with worse prognosis in 1001. Hijiya N, Hudson MM, Lensing S, Za- of the central nervous system: a comparative
cases. Acta Neuropathol. 62(1-2):31-40. papillary tumors of the pineal region. Am J Surg cher M, Onciu M, Behm FG, et al. (2007). study with primitive neuroectodermal tumor/me-
PMID:6318505 Pathol. 38(1):106-10. PMID:24121176 Cumulative incidence of secondary neoplasms dulloblastoma. Acta Neuropathol. 99(5):482-8.
969. Hassoun J, Soylemezoqlu F, Gambarelli 985. Heim S, Caras R, Ganslandt 0, Kufeld M, as a first event after childhood acute lympho- PMID:10805090
D, Figarella-Branger D, van Ammon K, Kleihues Bhimcke I, Paulus W, et al. (2013). Papillary tu- blastic leukemia. JAMA. 297(11):1207-15. 1017. Ho OM, Liu HC (1992). Primary intracra-
P (1993). Central neurocytoma: a synopsis of mor of the pineal region with anaplastic small PMID:17374815 nial germ cell tumor. Pathologic study of 51 pa-
clinical and histological features. Brain Pathol. cell component. J Neurooncol. 115(1):127-30. 1002. Hilden JM, Meerbaum S, Burger P, Finlay tients. Cancer. 70(6):1577-84. PMID:1325276
3(3):297-306. PMID:8293189 PMID:23817812 J, Janss A, Scheithauer BW, et al. (2004). Cen- 1018. Ho KL (1990). lntercellular septate-like
970. Hatton BA, Villavicencio EH, Tsuchiya KO, 986. Heim S, Sill M, Jones OT, Vasiljevic A, tral nervous system atypical teratoid/rhabdoid junction of neoplastic cells in myxopapillary
Pritchard JI, Ditzler S, Pullar B, et al. (2008). Jouvet A, Fevre-Montanqe M, et al. (2015). tumor: results of therapy in children enrolled ependymoma of the filum terminale. Acta Neu-
The Smo/Smo model: hedgehog-induced me- Papillary tumor of the pineal region: A distinct in a registry. J Clin Oneal. 22(14):2877-84. ropathol. 79(4):432-7. PMID:2339595
dulloblastoma with 90% incidence and lepto- molecular entity. Brain Pathol. PMID:26113311 PMID:15254056 1019. Ho KL (1990). Microtubular aggregates
meningeal spread. Cancer Res. 68(6):1768-76. 987. Heimdal K, Evensen SA, Fossa SD, 1003. Hill RM, Kuijper S, Lindsey JC, Pe· within rough endoplasmic reticulum in myxopa-
PMID:18339857 Hirscberg H, Langholm R, Br0gger A, et al. trie K, Schwalbe EC, Barker K, et al. (2015). pillary ependymoma of the filum terminale. Arch
971. Hatva E, Bohling T, Jiiiiskeliiinen J, Per- (1991). Karyotyping of a hematologic neoplasia Combined MYC and P53 defects emerge at Pathol Lab Med. 114(9):956-60. PMID:2390011
sico MG, Haltia M, Alitalo K (1996). Vascular developing shortly after treatment for cerebral medulloblastoma relapse and define rapidly 1020. Ho YS, Wei CH, Tsai MD, Wai YY (1992).

References 371
lntracerebral malignant fibrous histiocytoma: 1036. Horbinski C, Hobbs J, Cieply K, Dacie S, Mutations in the SMAD4/DPC4 gene in juve- 1069. Husemann K, Wolter M, Buschges R
case report and review of the literature. Neuro- Hamilton RL (2011 ). EGFR expression strati- nile polyposis. Science. 280(5366):1086-8. Bostrom J, Sabel M .. Reifenberger G {1999)'.
surgery. 31(3):567-71. PMID:1328926 fies oligodendroglioma behavior. Am J Pathol. PMID:9582123 ldenufication of two distinct deleted regions on
1021. Hoang MP, Amirkhan RH (2003). lnhibin 179(4):1638-44. PMID:21839716 1054. Hoyt WF, Baghdassarian SA (1969). the short arm of chromosome 1 and rare muta-
alpha distinguishes hemangioblastoma from 1037. Horbinski C, Kofler J, Yeaney G, Came- Optic glioma of childhood. Natural history and tion of the CDKN2C gene from 1 p32 in oligo.
clear cell renal cell carcinoma. Am J Surg Pa- lo-Piragua S, Venneti S, Louis DN, et al. (2011). rationale for conservative management. Br J dendroglial tumors. J Neuropathol Exp Neural.
thol. 27(8):1152-6. PMID:12883249 lsocitrate dehydrogenase 1 analysis differenti- Ophthalmol. 53(12):793-8. PMID:5386369 58(10):1041-50. PMID:10515227
1022. Hoang-Xuan K, Capefle L, Kujas M, ates gangliogliomas from infiltrative gliomas. 1055. Hruban RH, Shiu MH, Senie RT, Woo- 1070. Hussain N, Curran A, Pilling D, Mallu-
Taillibert S, Duffau H, Lejeune J, et al. (2004). Brain Pathol. 21(5):564-74. PMID:21314850 druff JM (1990). Malignant peripheral nerve ci CL, Ladusans EJ, Alfirevic Z, et al. (2006).
Temozolomide as initial treatment for adults 1038. Horiguchi H, Hirose T, Kannuki S, Na- sheath tumors of the buttock and lower extremi- Congenital subependymal giant cell astrocyto-
with low-grade oligodendrogliomas or oligoast- gahiro S, Sano T (1998). Gliosarcoma: an ty. A study of 43 cases. Cancer. 66(6):1253-65. ma diagnosed on fetal MRI. Arch Dis. Child.
rocytomas and correlation with chromosome immunohistochemical, ultrastructural and flu- PMID:2119249 91(6):520. PMID:16714726
1p deletions. J Clin Oneal. 22(15):3133-8. orescence in situ hybridization study. Pathol Int. 1056. Hsu DW, Louis DN, Efird JT, Hed- 1071. Husseini L, Saleh A, Reifenberger G
PMID:15284265 48(8):595-602. PMID:9736406 ley-Whyte ET (1997). Use of MIB-1 (Ki-67) Hartung HP, Kieseier BC (2012). lnllammatory
1023. Hobbs J, Nikiforova MN, Fardo DW, Bor- 1039. Horiguchi H, Hirose T, Sano T, Nagahiro immunoreactivity in differentiating grade fl and demyehnaling brain lesions heralding primary
toluzzi S, Cieply K, Hamilton RL, et al. (2012). S, Seki K, Fujimoto N, et al. (2000). Meningi- grade Ill gliomas. J Neuropathol Exp Neural. CNS lymphoma. Can J Neurol Sci. 39(1):6-10.
Paradoxical relationship between the degree oma with granulofilamentous inclusions. Ultra- 56(8):857-65. PMID:9258255 PMID:22384490
of EGFR amplification and outcome in gliob- struct Pathol. 24(4):267-71. PMID:11013967 1057. Huang H, Mahler-Araujo BM, Sankila 1072. Huttenlocher PR, Heydemann PT (1984).
lastomas. Am J Surg Pathol. 36(8):1186-93. 1040. Hornick JL, Bundock EA, Fletcher CD A, Chimelli L, Yonekawa Y, Kleihues P, et al. Fine structure of cortical tubers in tuberous sc-
PMID 22472960 (2009). Hybrid schwannoma/perineurioma: (2000). APC mutations in sporadic medul- lerosis: a Golgi study. Ann Neural. 16(5):595-
1024. Hoefnagel LD, van der Groep P, van de clinicopathologic analysis of 42 distinctive be- loblastomas. Am J Pathol. 156(2):433-7. 602. PMID:6508241
Vijver MJ, Boers JE, Wesseling P, Wesseling nign nerve sheath tumors. Am J Surg Pathol. PMID:10666372 1073. Hutter S, Piro RM, Reuss DE, Hovestad(
J, et al.; Dutch Distant Breast Cancer Metas- 33(10):1554-61. PMID:19623031 1058. Huang H, Reis R, Yonekawa Y, Lopes V, Sahm F, Farschtschi S, et al. (2014). Whole
tases Consortium (2013). Discordance in ERa, 1041. Hornick JL, Fletcher CD (2005). Intes- JM, Kleihues P, Ohgaki H (1999). Identificati- exome sequencing reveals that the majority of
PR and HER2 receptor status across different tinal perineuriomas: clinicopathologic defini- on in human brain tumors of DNA sequences schwannomatosis cases remain unexplained
distant breast cancer metastases within the tion of a new anatomic subset in a series of specific for SV40 large T antigen. Brain Pathol. after excluding SMARCB1 and LZTR1 germline
same patient. Ann Oneal. 24(12):3017-23. 10 cases. Am J Surg Pathol. 29(7):859-65. 9(1):33-42. PMID:9989448 variants. Acta Neuropathol. 128(3):449-52.
PMID:24114857 PMID:15958849 1058A. Huang J, Grotzer MA, Watanabe T, PMID:25008767
1025. Hoffman HJ, Otsubo H, Hendrick EB, 1042. Hornick JL, Fletcher CD (2005). Soft tis- Hewer E, Pietsch T, Rutkowski S, et al. (2008). 1074. Huynh DP, Mautner V, Baser ME, Stav-
Humphreys RP, Drake JM, Becker LE, et al. sue perineurioma: clinicopathologic analysis of Mutations in the Nijmegen breakage syndrome rou D, Pulst SM (1997). lmmunohislochemicaJ
(1991). lntracranial germ-cell tumors in child- 81 cases including those with atypical histolo- gene in medulloblastomas. Clin Cancer Res. detection of schwannomin and neurofibromin
ren. J Neurosurg. 74(4):545-51. PMID:1848284 gic features. Am J Surg Pathol. 29(7):845-58. 14(13):4053-8. PMID:18593981 in vestibular schwannomas, ependymomas
1026. Hoffman LM, Donson AM, Nakachi I, PMID:15958848 1059. Huang JG, Kavar B, Smith PD (2007). and meningiomas. J Neuropathol Exp Neural:
Griesinger AM, Birks DK, Amani V, et al. (2014). 1043. Hornick JL, Jaffe ES, Fletcher CD (2004) lntradural extramedullary spinal spread of oli- 56(4):382-90. PMID:9100669
Molecular sub-group-specific immunopheno- Extranodal histiocytic sarcoma: clinicopatho- goastrocytoma. J Clin Neurosci. 14(9):879-82. 1075. Hyman SL, Arthur Shores E, North KN
typic changes are associated with outcome in logic analysis of 14 cases of a rare epithelioid PMID:17582770 (2006). Learning disabilities in children with
recurrent posterior Iossa ependymoma. Acta malignancy. Am J Surg Pathol. 28(9):1133-44. 1060. Huang MC, Kubo 0, Tajika Y, Takakura neurofibromatosis type 1: subtypes, cognitive
Neuropathol. 127(5):731-45. PMID:24240813 PMID:15316312 K (1996). A clinico-immunohistochemical stu- profile, and attention-deficit-hyperactivity dis-
1027. Hofman S, Heeg M, Klein JP, Krikke AP 1044. Horstmann S, Perry A, Reifenberger G, dy of giant cell glioblastoma. Noshuyo Byori. order. Dev Med Child Neurol. 48(12):973-7.
(1998). Simultaneous occurrence of a supra- Giangaspero F, Huang H, Hara A, et al. (2004). 13(1):11-6. PMID:8916121 PMID:17109785
and an infratentorial glioma in a patient with Genetic and expression profiles of cerebellar 1061. Hufnagel T J, Kim JH, True LD, Manueli- 1076. Hutt-Cabezas M, Karajannis MA, Zagzag
Ollier's disease: more evidence for non-me- liponeurocytomas. Brain Pathol. 14(3):281-9. dis EE (1989). lmmunohistochemistry of capil- D, Shah S, Horkayne-Szakaly I, Rushing EJ,
sodermal tumor predisposition in multiple en- PMID:15446583 lary hemangioblastoma. lmmunoperoxidase-la- el al. (2013). Activation of mTORC1/mTORC2
chondromatosis. Skeletal Radial. 27(12):688- 1045. Horten BC, Rubinstein LJ (1976). Pri- beled antibody staining resolves the differential signaling in pediatric low-grade glioma and
91. PMID:9921931 mary cerebral neuroblastoma. A clinicopatho- diagnosis with metastatic renal cell carcinoma, pilocytic astrocytoma reveals m TOR as a the-
1028. Hofmann BM, Kreutzer J, Saeger W, logical study of 35 cases. Brain. 99(4):735-56. but does not explain the histogenesis of the rapeutic target. Neuro Oneal. 15(12):1604-14.
Buchfelder M, Blurncke I, Fahlbusch R, et al. PMID:1030655 capillary hemangioblastoma. Am J Surg Pathol. PMID:24203892
(2006). Nuclear beta-catenin accumulation as 1046. Hoshino T, Wilson BC, Ellis WG (1975). 13(3):207-16. PMID:2465700 1077. HolskenA, Kreutzer J, Hofmann BM, Hans
reliable marker for the differentiation between Gemislocytic astrocytes in gliomas. An auto- 1062. Hulsebos TJ, Kenter S, Siebers-Renell V, Oppel F, Buchfelder M, et al. (2009). Target
cystic craniopharyngiomas and ralhke cleft radiographic study. J Neuropathol Exp Neural. U, Hans V, Wesseling P, Flucke U (2014). SM- gene activation of the Wnt signaling pathway
cysts: a clinico-pathologic approach. Am J Surg 34(3):263-81. PMID:167133 ARCB1 involvement in the development of lei- in nuclear beta-catenin accumulating cells of
Pathol. 30(12):1595-603. PMID:17122517 1047. Hosokawa Y, Tsuchihashi Y, Okabe H, omyoma in a patient with schwannomatosis. Am adamantinomatous craniopharyngiomas. Brain
1029. Hoischen A, Ehrler M, Fassunke J, Simon Toyama M, Namura K, Kuga M, et al. (1991). J Surg Pathol. 38(3):421-5. PMID:24525513 Pathol. 19(3):357-64. PMID:18540944
M, Baudis M, Landwehr C, et al. (2008). Com- Pleomorphic xanthoastrocyloma. Ultrastructu- 1063. Hulsebos T J, Kenter S, Verhagen WI, 1078. lchimura K, Pearson DM, Kocialkowski S,
prehensive characterization of genomic aber- ral, immunohistochemical, and DNA cytofluo- Baas F, Flucke U, Wesseling P (2014). Prema- Backlund LM, Chan R, Jones DT, et al. (2009).
rations in gangliogliomas by CGH, array-ba- rometric study of a case. Cancer. 68(4):853-9. ture termination of SMARCB1 translation may IDH1 mutations are present in the majority of
sed CGH and interphase FISH. Brain Pathol. PMID:1855184 be followed by reinitiation in schwannomato- common adult gliomas but rare in primary
18(3):326-37. PMID:18371186 1048. Hou Z, Wu Z, Zhang J, Zhang L, nan sis-associated schwannomas, but results in glioblastomas. Neuro Oncol. 11(4):341-7.
1030. Holash J, Maisonpierre PC, Compton R, Liu B, et al. (2013). Clinical features and absence of SMARCB1 expression in rhabdoid PMID:19435942
D, Boland P, Alexander CR, Zagzag D, et al. management of intracranial subependymo- tumors. Acta Neuropathol. 128(3):439-48. 1079. lchimura K, Schmidt EE, Miyakawa A,
(1999). Vessel cooption, regression, and growth mas in children. J Clin Neurosci. 20(1):84-8. PMID:24740647 Goike HM, Collins VP (1998). Distinct patterns
in tumors mediated by angiopoietins and VEGF. PMID:23117139 1064. Hulsebos T J, Plomp AS, Walterman RA, of deletion on 10p and 10q suggest involvement
Science. 284(5422):1994-8. PMID:10373119 1049. Hovestadt V, Remke M, Kool M, Pietsch Robanus-Maandag EC, Baas F, Wesseling P of multiple tumor suppressor genes in the de-
1031. Homma T, Fukushima T, Vaccarella S, T, Northcott PA, Fischer R, et al. (2013). Robust (2007). Germline mutation of INl1/SMARCB1 velopment of astrocytic gliomas of different ma-
Yonekawa Y, Di Paire PL, Franceschi S, et al. molecular subgrouping and copy-number pro- in familial schwannomatosis. Am J Hum Genet. lignancy grades. Genes Chromosomes Cancer.
(2006). Correlation among pathology, geno- filing of medulloblastoma from small amounts 80(4):805-10. PMID:17357086 22(1):9-15. PMID:9591629
type, and patient outcomes in glioblastoma. of archival tumour material using high-density 1065. Hung KL, Wu CM, Huang JS, How SW 1080. lczkowski KA, Butler SL, Shanks JH,
J Neuropathol Exp Neural. 65(9):846-54. DNA methylation arrays. Acta Neuropalhol. (1990). Familial medulloblastoma in siblings: Hossain D, Schall A, Meiers I, et al. (2008).
PMID:16957578 125(6):913-6. PMID:23670100 report in one family and review of the literature. Trials of new germ cell immunohistochemi·
1032. Honan WP, Anderson M, Carey MP, Wil- 1050. Howard BM, Hofstetter C, Wagner PL, Surg Neural. 33(5):341-6. PMID:2184531 cal stains in 93 extragonadal and metastatic
liams B (1987). Familial subependymomas. Br J Muskin ET, Lavi E, Boockvar JA (2009). Trans- 1066. Husain AN, Leestma JE (1986). Cerebral germ cell tumors. Hum Pathol. 39(2):275-81.
Neurosurg. 1(3):317-21. PMID:3268127 formation of a low-grade pineal parenchymal tu- astroblastoma: immunohistochemical and ultra- PMID:18045648
1033. Honavar M, Janeta I (1994). 73 cases mour to secondary pineoblastoma. Neuropalhol structural features. Case report. J Neurosurg. 1081. Ida CM, Rodriguez FJ, Burger PC, Ca- (
of dysembryoplastic neuroepilhelial tumour: Appl Neurobiol. 35(2):214-7. PMID:19284482 64(4)657-61. PMID:3950749 ron AA, Jenkins SM, Spears GM, et al. (2015).
lhe range of histological appearances. Brain 1051. Howard JE, Dwivedi RC, Masterson L, 1067. Huse JT, Diamond EL, Wang L, Rosen- Pleomorphic Xanthoastrocytoma: Natural His-
Pathol. 4:428. Jani P (2015). Langerhans cell sarcoma: a sys- blum MK (2015). Mixed glioma with molecular tory and Long- Term Follow-Up. Brain Pathol.
1034. Horbinski C (2013). To BRAF or not tematic review. Cancer Treat Rev. 41(4):320- features of composite oligodendroglioma and 25(5):575-86 PMID:25318587
to BRAF: is that even a question anymo- 31. PMID:25805533 astrocytoma: a true "oligoastrocytoma"? Acta 1082. Ida CM, Vrana JA, Rodriguez FJ, Jen-
re? J Neuropathol Exp Neural. 72(1):2-7. 1052. Howe JR, Ringold JC, Summers RW, Neuropathol. 129(1):151-3. PMID:25359109 toft ME, Caron AA, Jenkins SM, et al. (2013).
PMID:23242278 Mitros FA, Nishimura DY, Stone EM (1998). 1068. Huse JT, Edgar M, Halliday J, Mikolaenko lmmunohistochemistry is highly sensitive and
1035. Horbinski C, Dacie S, Mclendon RE, Cie- A gene for familial juvenile polyposis maps I, Lavi E, Rosenblum MK (2013). Multinodular specific for detection of BRAF V600E mutation
ply K, Datto M, Brat DJ, et al. (2009). Chordoid to chromosome 18q21.1. Am J Hum Genet. and vacuolating neuronal tumors of the ce- in pleomorphic xanthoastrocytoma. Acta Neuro-
glioma: a case report and molecular characte- 62(5):1129-36. PMID:9545410 rebrum: 10 cases of a distinctive seizure-as- pathol Commun. 1:20. PMID:24252190
rization of five cases. Brain Pathol. 19(3):439- 1053. Howe JR, Roth S, Ringold JC, Summers sociated lesion. Brain Pathol. 23(5):515-24. 1083. ldbaih A, Crioiere E, Marie Y, Rousseau
48. PMID:18652591 RW, Jarvinen HJ, Sistonen P, et al. (1998). PMID:23324039 A, Mokhtari K, Kujas M, et al. (2008). Gene

372 References
amplification is a poor prognostic factor in Absence of p53 gene mutations in a tumor 1115. Jacoby LB, Jones D, Davis K, Kronn large B-cell lymphoma of !he elderly: a clini-
anaplastic o/igodendrogliomas. Neuro Oneal. panel representative of pilocytic astrocytoma D, Short MP, Gusella J, et al. (1997). Mole- copathologic study of five cases. Brain Tumor
10(4):540-7. PM/0:18544654 diversity using a p53 functional assay. In! J cular analysis of the NF2 tumor-suppressor Palhol. 31(4):265-73. PMID:24399201
1084. /dbaih A, Ducray F, Dehais C, Courdy Cancer. 76(6):797-800. PM/0:9626343 gene in schwannomatosis. Am J Hum Genet. 1132. Jansen M, Mahapatra G, Betensky
C, Carpentier C, de Bernard S, et al.; POLA 1099. /shizawa K, Kan-nuki S, Kumagai H, Ko- 61(6):1293-302. PMID:9399891 RA, Keohane C, Louis ON (2012). Gain of
Network (2012). SNP array analysis reveals mori T, Hirose T (2002). Lipomatous primitive 1116. Jacoby LB, MacCollin M, Barone R, chromosome arm 1q in atypical meningioma
novel genomic abnormalities including copy neuroectodermal tumor with a glioblastoma Ramesh V, Gusella JF (1996). Frequency and correlates with shorter progression-free survi-
neutral loss of heterozygosity in anaplastic component: a case report. Acta Neuropathol. distribution of NF2 mutations in schwannomas. val. Neuropathol Appl Neurobiol. 38(2):213-9.
oligodendrogliomas. PLoS One. 7(10):e45950. 103(2):193-8. PMID 11810187 Genes Chromosomes Cancer. 17(1 ):45-55. PMID:21988727
PMID:23071531 1100. lshizawa K, Komori T, Hirose T (2005). PM/0:8889506 1133. Janson K, Nedzi LA, David 0, Schorin M,
1085. ldbaih A, Mokhtari K, Emile JF, Galanaud Stromal cells in hemangioblastoma: neuroecto- 1117. Jacoby LB, MacCollin M, Louis ON, Walsh JW. Bhaltacharjee M, et al. (2006). Pre-
D, Be/aid H, de Bernard S, et al. (2014). Dra- dermal differentiation and morphological simila- Mohney T, Rubio MP, Pulaski K, el al. (1994). disposition to atypical teratoid/rhabdoid tumor
matic response of a BRAF V600E-mutated pri- rities to ependymoma. Pathol Int. 55(7):377-85. Exon scanning for mutation of the NF2 gene in due to an inherited IN/1 mutation. Pediatr Blood
mary CNS histiocytic sarcoma to vemurafenib. PMID:15982211 schwannomas. Hum Mo/ Genet. 3(3):413-9. Cancer. 47(3):279-84. PMI0:16261613
Neurology. 83(16):1478-80. PMID:25209580 1101. lshizawa K, Komori T, Shimada S, Hirose PMID:8012353 1134. Janssen 0, Harms D (2005). Juvenile
1086. Ikeda J, Sawamura Y, van Meir EG T (2008). Olig2 and CD99 are useful negative 1118. Jacoby LB, Pulaski K, Rouleau GA, xanthogranuloma in childhood and adolescen-
(1998). Pineoblastoma presenting in familial markers for the diagnosis of brain tumors. Clin Martuza RL (1990). Clonal analysis of human ce: a clinicopathologic study of 129 patients
adenomatous polyposis (FAP): random asso- Neuropathol. 27(3):118-28. PMID:18552083 meningiomas and schwannomas. Cancer Res. from the kiel pediatric tumor registry. Am J Surg
ciation, FAP variant or Turco! syndrome? Br J 1102. lshizawa T, Komori T, Shibahara J, lshiza- 50(21):6783-B. PMID:2208143 Pathol. 29(1):21-8. PM/0:15613853
Neurosurg. 12(6):576-8. PM/0:10070471 wa K, Adachi J, Nishikawa R, et al. (2006). Pa- 1119. Jacques TS, Eldridge C, Pate/A, Saleem 1135. Janz C, Buhl R (2014). Astroblastoma:
1087. lkezaki K, Matsushima T, Inoue T, Yokoya- pillary glioneuronal tumor with minigemistocytic NM, Powell M, Kitchen ND, el al. (2006). Mixed report of two cases with unexpected clinical be-
ma N, Kaneko Y, Fukui M (1993). Correlation of components and increased proliferative activity. glioneuronal tumour of the fourth ventricle with havior and review of the literature. Clin Neurol
microanatomical localization with postoperative Hum Pathol. 37(5):627-30. PMID:16647962 prominent rosette formation. Neuropathol Appl Neurosurg. 125:114-24. PMID:25108699
survival in posterior Iossa ependymomas. Neu- 1103. lsmat FA, Xu J, Lu MM, Epstein JA Neurobiol. 32(2):217-20. PMID:16599951 1136. Japp AS, Gessi M, Messinq-Jimqer M,
rosurgery. 32(1 ):38-44. PMID:8421555 (2006). The neurofibromin GAP-related do· 1120. Jacques TS, Valentine A, Bradford R, Denkhaus 0, Zur MQhlen A, Wolff JE, el al.
1088. lkota H, Tanaka Y, Yokoo H, Nakazato main rescues endothelial but no! neural Mclaughlin JE (2004). December 2003: a (2015). High-resolution genomic analysis does
Y (2011 ). Clinicopathologica/ and immunohis- crest development in Nf1 mice. J Clin Invest. 70-year-old woman with a recurrent meningeal not qualify atypical plexus papilloma as a se-
lochemical study of 20 choroid plexus tumors: 116(9):2378-84. PM/0:16906226 mass. Recurrent meningioma with rhabdomyo- parate entity among choroid plexus tumors.
their histological diversity and the expression of 1104. Italiano A, Sung YS, Zhang L, Singer S, sarcomatous differentiation. Brain Palhol. J Neuropathol Exp Neural. 74(2):110-20.
markers useful for differentiation from rnetas- Maki RG, Coindre JM, et al. (2012). High pre- 14(2):229-30. PM/0:15193039 PMID:25575132
latic cancer. Brain Tumor Pathol. 28(3):215-21. valence of CIC fusion with double-homeobox 1121. Jaeckle KA, Decker PA, Ballman KV, 1137. Jaros E, Perry RH, Adam L, Kelly PJ,
PM/0:21394517 (DUX4) transcription factors in EWSR1-negati· Flynn PJ, Giannini C, Scheithauer BW, et al. Crawford PJ, Kalbag RM, et al. (1992). Prog-
1089. llhan /, Berberoglu S, Kutluay L, Maden ve undifferentiated small blue round cell sarco- (2011). Transformation of low grade glioma nostic implications of p53 protein, epidermal
HA (1998). Subcutaneous sacrococcygeal mas. Genes Chromosomes Cancer. 51 (3):207- and correlation with outcome: an NCCTG da- growth factor receptor, and Ki-67 labelling in
myxopapillary ependymoma. Med Pedialr On- 18. PM/0:22072439 !abase analysis. J Neurooncol. 104(1):253-9. brain tumours. Br J Cancer. 66(2):373-85.
eal. 30(2):81-4. PMID:9403014 1105. Ito T, Kanno H, Sato K, Oikawa M, Ozaki PMID:21153680 PMID:1503912
1090. lmperiale A, Moussallieh FM, Roche P, Y, Nakamura H, et al. (2014). Clinicopathologic 1122. Jaffe ES, Harris NL, Vardiman JW, Cam- 1138. Jarrell ST, Vortmeyer AO, Linehan WM,
Battini S, Cicek AE, Sebag F, et al. (2015). Me- study of pineal parenchymal tumors of interme- po E, Arber DA, editors. (2010). Hernatopatho- Oldfield EH, Lanser RR (2006). Metastases
labolome profiling by HRMAS NMR spectrosco- diate differentiation. World Neurosurg. 81(5· logy. 1st Philadelphia. Saunders/ Elsevier. to hemangioblastomas in von Hippel-Lindau
py of pheochromocytomas and paragangliomas 6):783-9. PMID:23396072 1123. Jahnke K, Korfel A, Komm J, Bechrakis disease. J Neurosurg. 105(2):256-63.
detects SDH deficiency: clinical and pathophy- 1106. lwaki T, Fukui M, Kondo A, Matsushima NE, Stein H, Thiel E, el al. (2006). lntraocular PMID:17219831
siological implications. Neoplasia. 17(1 ):55-65. T, Takeshita I (1987). Epithelial properties of lymphoma 2000-2005: results of a retrospecti- 1139. Javahery RJ, Davidson L, Fangusaro
PMID:25622899 pleomorphic xanthoastrocytomas determined ve multicentre trial. Graef es Arch Clin Exp Opht- J, Finlay JL, Gonzalez-Gomez I, McComb JG
1091. lnatomi Y, Ito T, Nagae K, Yamada Y, in ultrastructural and immunohistochemical halmol. 244(6):663-9. PMID:16228920 (2009). Aggressive variant of a papillary glio-
Kiyomatsu M, Nakano-Nakamura M, et al. studies. Acta Neuropathol. 74(2):142-50. 1124. Jahnke K, Thiel E, Marius P, Herrlinger neuronal tumor. Report of 2 cases. J Neurosurg
(2014). Hybrid perineurioma-neurofibroma in PMID:3673505 U, Weller M, Fischer L, el al.; German Primary Pediatr. 3(1):46-52. PMID:19119904
a patient with neurofibromatosis type 1, cli- 1107. Iwamoto FM, DeAngelis LM, Abrey LE Central Nervous System Lymphoma Study 1140. Jay V, Edwards V, Squire J, Rutka
nically mimicking malignant peripheral nerve (2006). Primary dural lymphomas: a clinico- Group (2006). Relapse of primary central J (1993). Astroblastoma: report of a case
sheath tumor. Eur J Dermalol. 24(3):412-3. pathologic study of treatment and outcome nervous system lymphoma: clinical features, with ultrastructural, cell kinetic, and cytoge-
PMID:24751814 in eight patients. Neurology. 66(11):1763-5. outcome and prognostic factors. J Neurooncol. netic analysis. Pediatr Pathol. 13(3):323-32.
1092. Ingham PW (1998). The patched gene in PMID:16769960 80(2):159-65. PMID:16699873 PMID:8516227
development and cancer. Curr Opin Genet Dev. 1108. Iwashita T, Enjoji M (1987). Plexiform 1125. Jahnke K, Thiel E, Schilling A, Herrlin- 1141. Jay V, Squire J, Becker LE, Humphreys
8(1):88-94. PMID:9529611 neurilemmoma: a clinicopathological and im- ger U, Weller M, Coupland SE, et al. (2005). R (1994). Malignant transformation in a gan-
1093. Ingold B, Wild PJ, Nocito A, Amin MB, munohistochemical analysis of 23 tumours from Low-grade primary central nervous system glioglioma with anaplastic neuronal and ast-
Storz M, Heppner FL, et al. (2008). Renal cell 20 patients. Virchows Arch A Pathol Anal Histo- lymphoma in immunocompetent patients. Br J rocytic components. Report of a case with flow
carcinoma marker reliably discriminates central pathol. 411(4):305-9. PMID:3114942 Haemalol. 128(5):616-24. PMID:15725082 cytometric and cytogenetic analysis. Cancer.
nervous system haemangioblasloma from brain 1109. Iwata H, Mori Y, Takagi H, Shirahashi K, 1126. Jain A, Amin AG, Jain P, Burger P, Jal- 73(11):2862-8. PMID:8194028
metastases of renal cell carcinoma. Histopalho- Shinoda J, Shimokawa K, el al. (2004). Medi· lo GI, Lim M, et al. (2012). Subependymoma: 1142. Jay V, Squire J, Blaser S, Hoffman HJ,
logy. 52(6):674-81. PMID:18393979 astinal growing teratoma syndrome after cispla- clinical features and surgical outcomes. Neural Hwang P (1997). lnlracranial and spinal metas-
1094. Inoue Y, Nemoto Y, Murata R, Tashiro T, tin-based chemotherapy and radiotherapy for Res. 34(7):677-84. PMID22747714 tases from a ganglioglioma with unusual cyto-
Shakudo M, Kohno K, et al. (1998). CT and MR intracranial germinoma. J Thorac Cardiovasc 1127. Jain RK, Carmeliet P (2012). SnapShol: genetic abnormalities in a patient with complex
imaging of cerebral tuberous sclerosis. Brain Surg. 127(1):291-3. PMID:14752454 Tumor angiogenesis. Cell. 149(6):1408-1408. partial seizures. Childs Nerv Syst. 13(10):550-
Dev. 20(4):209-21. PM/0:9661965 1110. Jacks T, Shih TS, Schmitt EM, Bronson e1. PMID:22682256 5. PMID:9403205
1095. Cairncross G, Berkey B, Shaw E, Jenkins RT, Bernards A, Weinberg RA (1994). Tumour 1128. Jakacki RI, Burger PC, Kocak M, Bo- 1143. Jeffs GJ, Lee GY, Wong GT (2003). Fun-
R, Scheilhauer B, Brachman 0, el al.; Interg- predisposition in mice heterozygous for a tar- yett JM, Goldwein J, Mehta M, et al. (2015). ctioning paraganglioma of the thoracic spine:
roup Radiation Therapy Oncology Group Trial geted mutation in Nf1. Nat Genet. 7(3):353-61. Outcome and prognostic factors for children case report. Neurosurgery. 53(4):992-4, dis-
9402 (2006). Phase Ill trial of chemotherapy PMID:7920653 with supratentorial primitive neuroectodermal cussion 994-5. PMID 14519233
plus radiotherapy compared with radiotherapy 1111. Jackson CG (2001). Glomus tympanicum tumors treated with carboplatin during radio- 1144. Jehi L, Yardi R, Chagin K, Tassi L, Russo
alone for pure and mixed anaplastic oligo- and glomus jugulare tumors . Otolaryngol Clin therapy: a report from the Children's Oncology GL, Worrell G, et al. (2015). Development and
dendroglioma: Intergroup Radiation Therapy North Am 34: 941-70, vii. PMID:11557448 Group. Pedialr Blood Cancer. 62(5):776-83. validation of nomograms to provide individuali-
Oncology Group Trial 9402. J Clin Oncol. 1112. Jackson M, Hassiotou F, Nowak A (2015). PMID:25704363 sed predictions of seizure outcomes after epi-
24(18):2707-14. PMID:16782910 Glioblasloma stem-like cells: at the root of tu- 1129. Jakobiec FA, Kool M, Stagner AM, Pfister lepsy surgery: a retrospective analysis. Lancet
1096. Ironside JW, Jefferson AA, Royds JA, mor recurrence and a therapeutic target. Car- SM, Eagle RC, Proia AD, el al. (2015). lntrao- Neural. 14(3):283-90. PMID:25638640
Taylor CB, Timperley WR (1984). Carcinoid cinogenesis. 36(2):177-85. PMID:25504149 cular Medulloepitheliomas and Embryonal Tu- 1145. Jeibmann A, Eikmeier K, Linge A, Kool
tumour arising in a recurrent intradural spi- 1113. Jacob K, Albrecht S, So/lier C, Faury 0, mors With Multilayered Rosettes of the Brain: M, Koos B, Schulz J, et al. (2014). Identi-
nal teratoma. Neuropathol Appl Neurobiol. Sader E, Montpetit A, et al. (2009). Duplication Comparative Roles of LIN28A and C19MC. fication of genes involved in the biology of
10(6)479-89. PMID:6084821 of 7q34 is specific to juvenile pilocytic aslrocyto- Am J Ophthalmol. 159(6):1065-1074.e1. atypical teraloid/rhabdoid tumours using Dro-
1097. Ishida M, Hotta M, Tsukamura A, Taga mas and a hallmark of cerebellar and optic pa- PMID:25748578 sophila melanogaster. Nat Commun. 5:4005.
T, Kato H, Ohta S, et al. (2010). Malignant thway tumours. Br J Cancer. 101(4):722-33. 1130. Jallo GI, Zagzag D, Epstein F (1996). PMID:24892285
transformation in craniopharyngioma after PMID:19603027 lntramedullary subependymoma of the 1146. Jeibmann A, Hasse/blatt M, Gerss J,
radiation therapy: a case report and review 1114. Jacobs JJ, Rosenberg AE (1989). Ex- spinal cord. Neurosurgery. 38(2):251-7. Wrede B, Egensperger R, Beschorner R, et
of the literature. Clin Neuropathol. 29(1 ):2-8. tracranial skeletal metastasis from a pinea- PMID:8869051 al. (2006). Prognostic implications of atypical
PMID:20040326 loblastoma. A case report and review of the 1131. Jamal SE, Li S, Bajaj R, Wang Z, Kenyon histologic features in choroid plexus papilloma.
1098. Ishii N, Sawamura Y, Tada M, Daub OM, literature. Clin Orthop Relat Res. (247):256-60. L, Glass J, et al. (2014). Primary central ner- J Neuropathol Exp Neural. 65(11):1069-73.
Janzer RC, Meagher-Villemure M, et al. (1998). PMID:2676297 vous system Epstein-Barr virus-positive diffuse PMID:17086103

References 373
1147. JeibmannA, Wrede B, Peters 0, Wolff JE, M, Schramm J, Wiestler OD, et al. (2001 ). al. (2015). Cross-reactivity of the BRAF VE1 an- leiomyomatosis). Zentralbl Pathol. 140(2):195-
Paulus W, Hasselblatt M (2007). Malignant pro- Comparative analysis of the NF2, TP53, PTEN, tibody with epitopes in axonemal dyneins leads 200. PMID:7947627
gression in choroid plexus papillomas. J Neu- KRAS, NRAS and HRAS genes in sporadic and to staining of cilia. Mod Pathol. 28(4):596-606. 1196. Janisch W, Staneczek W (1989). [Pri-
rosurg. 107(3) Suppl:199-202. PMID:17918524 radiation-induced human meningiomas. Int J PMID:25412847 mary tumors of the choroid plexus. Frequency,
1148. Jellinger K (2009). Metastatic oligo- Cancer. 94(2):218-21. PMID:11668501 1181. Jordanova ES, Riemersma SA, Philip- localization and age]. Zentralbl Allg Pathol.
dendrogliomas: a review of the literature and 1165. Johannsson 0, Ostenmeyer EA, Hakans- pa K, Giphart-Gassler M, Schuuring E, Kluin 135(3):235-40. PMID:2773602
case report. Acta Neurochir (Wien). 151(8):987. son S, Friedman LS, Johansson U, Sellberg G, PM (2002). Hemizygous deletions in the HLA 1197. Jaaskelainen J (1986). Seemingly com-
PMID:19424658 et al. (1996). Founding BRCA1 mutations in he- region account for loss of heterozygosity in plete removal of histologically benign intracra-
1149. Jellinger K, Bock F, Brenner H (1988). reditary breast and ovarian cancer in southern the majority of diffuse large B-cell lymphomas nial meningioma: late recurrence rate and
Meningeal melanocytoma. Report of a case and Sweden. Am J Hum Genet. 58(3):441-50. of the testis and the central nervous system. factors predicting recurrence in 657 patients. A
review of the literature. Acta Neurochir (Wien). PMID:8644702 Genes Chromosomes Cancer. 35(1 ):38-48. multivariate analysis. Surg Neural. 26(5):461-9.
94(1-2):78-87. PMID:3051898 1166. Goldblum JR, Weiss SW, Folpe AL PMID:12203788 PMID:3764651
1150. Jenevein EP (1964). A neurohypophyseal (2013). Enzinger and Weiss's Soft Tissue Tu- 1182. Joseph NM, Mosher JT, Buchstal- 1198. Jaaskelainen J, Paetau A, Pyykkii t,
tumor originating from pituicytes. Am J Clin Pa- mors. 6th ed. Philadelphia: Elsevier Saunders. ler J, Snider P, McKeever PE, Lim M, et al. Blomstedt G, Paiva T, Troupp H (1994). Interfa-
thol. 41:522-6. PMID:14165459 1167. Johnson BE, Mazor T, Hong C, Barnes (2008}. The loss of Nf1 transiently promotes ce between the facial nerve and large acoustic
1151. Jenkins RB, Blair H, Ballman KV, Giannini M, Aihara K, Mclean CY, et al. (2014). Mutatio- self-renewal but not tumorigenesis by neural neurinomas. lmmunohistochemical study of the
C, Arusell RM, Law M, et al. (2006). A 1(1;19) nal analysis reveals the origin and therapy-dri- crest stem cells. Cancer Cell. 13(2):129-40. cleavage plane in NF2 and non-NF2 cases. J
(q10;p10) mediates the combined deletions of ven evolution of recurrent glioma. Science. PMID:18242513 Neurosurg. 80(3):541-7. PMID:8113868
1 p and 19q and predicts a better prognosis of 343(6167):189-93. PMID:24336570 1183. Joseph NM, Phillips J, Dahiya S, M Fe- 1199. Kacerovska D, Michal M, Kuroda N
patients with oligodendroglioma. Cancer Res. 1168. Johnson MW, Eberhart CG, Perry A, licella M, Tihan T, Brat DJ, et al. (2013). Diag- Tanaka A, Sima R, Oenisjuk N, et al. (2013)'.
66(20):9852-61. PMID:17047046 Tihan T, Cohen KJ, Rosenblum MK, et al. nostic implications of IDH1-R132H and OLIG2 Hybrid peripheral nerve sheath tumors, inclu-
1152. Jenkins RB, Xiao Y, Sicotte H, Decker (2010). Spectrum of pilomyxoid astrocytomas: expression patterns in rare and challenging gli- ding a malignant variant in type 1 neurofibro-
PA, Kollmeyer TM, Hansen HM, et al. (2012). intermediate pilomyxoid tumors. Am J Surg Pa- oblastoma variants. Mod Pathol. 26(3):315-26. matosis. Am J Dermatopathol. 35(6):641-9.
A low-frequency variant at 8q24.21 is strongly thol. 34(12):1783-91. PMID:21107083 PMID:23041832 PMID:23676318
associated with risk of oligodendroglial tumors 1169. Johnson MW, Emelin JK, Park SH, Vin- 1184. Jouvet A, Fauchon F, liberski P, 1200. Kachhara R, Bhattacharya RN, Nair S,
and astrocytomas with IDH1 or IDH2 mutation. ters HV (1999). Co-localization of TSC1 and Saint-Pierre G, Didier-Bazes M, Heitzmann Radhakrishnan VV (2003). liponeurocytoma
Nat Genet. 44(10):1122-5. PMID:22922872 TSC2 gene products in tubers of patients with A, et al. (2003). Papillary tumor of the pine- of the cerebellum-a case report. Neural India.
1153. Jenkinson MD, Bosma JJ, Du Plessis D, tuberous sclerosis. Brain Pathol. 9(1):45-54. al region. Am J Surg Pathol. 27(4):505-12. 51(2):274-6. PMID:14571027
Ohgaki H, Kleihues P, Warnke P, et al. (2003). PMID:9989450 PMID:12657936 1201. Kadonaga JN, Frieden IJ (1991). Neu-
Cerebellar liponeurocytoma with an unusually 1170. Johnson MW, Kerfoot C, Bushnell T, Li 1185. Jouvet A, Fevre-Montanqe M, Besancon rocutaneous melanosis: definition and review
aggressive clinical course: case report. Neu- M, Vinters HV (2001 ). Hamartin and tuberin R, Derrington E, Saint-Pierre G, Belin MF, et al. of the literature. J Am Acad Denmatol. 24(5 Pt
rosurgery. 53(6):1425-7, discussion 1428. expression in human tissues. Mod Pathol. (1994). Structural and ultrastructural characteri- 1):747-55. PMID:1869648
PMID:14633310 14(3):202-10. PMID:11266527 stics of human pineal gland, and pineal paren- 1202. Kaido T, Sasaoka Y, Hashimoto H, Taira
1154. Jennings MT, Gelman R, Hochberg F 1171. Johnson RA, Wright KD, Poppleton H, chymal tumors. Acta Neuropathol. 88(4):334- K (2003). De nova germinoma in the brain in
(1985). lntracranial germ-cell tumors: natu- Mohankumar KM, Finkelstein D, Pounds SB, 48. PMID:7839826 association with Klinefelter's syndrome: case
ral history and pathogenesis. J Neurosurg. et al. (2010). Cross-species genomics matches 1186. Jouvet A, Lellouch-Tubiana A, Boddaert report and review of the literature. Surg Neural.
63(2):155-67. PMID:2991485 driver mutations and cell compartments to mo- N, Zerah M, Champier J, Fevre-Montanqs M 60(6):553-8, discussion 559. PMID:14670679
1155. Jensen RL, Caamano E, Jensen EM, del ependymoma. Nature. 466(7306):632-6. (2005). Fourth ventricle neurocytoma with li- 1203. Kakita A, lnenaga C, Kameyama S,
Couldwell WT (2006). Development of contrast PMID:20639864 pomatous and ependymal differentiation. Acta Masuda H, Ueno T, Honma J, et al. (2005).
enhancement after long-tenm observation of a 1172. Johnson Rl, Rothman Al, Xie J, Neuropathol. 109(3):346-51. PMID:15627205 Cerebral lipoma and the undertying cortex of
dysembryoplastic neuroepithelial tumor. J Neu- Goodrich LV, Bare JW, Bonifas JM, et al. 1187. Jouvet A, Saint-Pierre G, Fauchon F, the temporal lobe: pathological features asso-
rooncol. 78(1):59-62. PMID:16314940 (1996). Human homolog of patched, a candi- Privat K, Bouffet E, Ruchoux MM, et al. (2000). ciated with the malformation. Acta Neuropathol.
1156. Jentoft M, Giannini C, Rossi S, Mota date gene for the basal cell nevus syndrome. Pineal parenchymal tumors: a correlation of 109(3):339-45. PMID: 15622498
R, Jenkins RB, Rodriguez FJ (2011). Oligo- Science. 272(5268):166!>-71. PMID:8658145 histological features with prognosis in 66 cases. 1204. Kalamarides M, Niwa-Kawakita M, Leblo-
dendroglial tumors with marked desmoplasia: 1173. Johnston Dl, Keene Dl, Lafay-Cousin Brain Pathol. 10(1):49-60. PMID:10668895 is H, Abramowski V, Perricaudet M, Janin A, et
clinicopathologic and molecular features of l, Steinbok P, Sung l, Carrel AS, et al. (2008). 1188. Jozwiak J, Jozwiak S, Skopinski P al. (2002). Nf2 gene inactivation in arachnoidal
7 cases. Am J Surg Pathol. 35(6):845-52. Supratentorial primitive neuroectodermal tu- (2005). lmmunohistochemical and microscopic cells is rate-limiting for meningioma develop-
PMID:21552114 mors: a Canadian pediatric brain tumor con- studies on giant cells in tuberous sclerosis. His- ment in the mouse. Genes Dev. 16(9): 1060-5.
1157. Jeong JY, Suh YL, Hong SW (2014). Aty- sortium report. J Neurooncol. 86(1):101-8. tot Histopathol. 20(4):1321-6. PMID:16136513 PMID:12000789
pical teratoid/rhabdoid tumor arising in pleomor- PMID:17619825 1189. Juco J, Horvath E, Smyth H, Rotondo F, 1205. Kalamarides M, Stemmer-Rachamimov
phic xanthoastrocytoma: a case report. Neuro- 1174. Jones AC, Shyamsundar MM, Thomas Kovacs K (2007). Hemangiopericytoma of the AO, Niwa-Kawakita M, Chareyre F, Taranchon
pathology. 34(4):398-405. PMID:25268025 MW, Maynard J, ldziaszczyk S, Tomkins S, et sella mimicking pituitary adenoma: case report E, Han ZY, et al. (2011). Identification of a
1159. Jeuken JW, Sprenger SH, Gilhuis J, al. (1999). Comprehensive mutation analysis of and review of the literature. Clin Neuropathol. progenitor cell of origin capable of generating
Teepen Hl, GrotenhuisAJ, Wesseling P (2002). TSC1 and TSC2-and phenotypic correlations in 26(6):288-93. PMID:18232595 diverse meningioma histological subtypes. On-
Correlation between localization, age, and chro- 150 families with tuberous sclerosis. Am J Hum 1190. Judkins AR, Burger PC, Hamilton Rl, cogene. 30(20):2333-44. PMID:21242963
mosomal imbalances in ependymal tumours as Genet. 64(5):1305-15. PMID:10205261 Kleinschmidt-DeMasters B, Perry A, Pomeroy 1206. Kaloshi G, Alikaj V, RrojiA, Vreto G, Petre-
detected by CGH. J Pathol. 197(2):238-44. 1175. Jones C, Baker SJ (2014). Unique SL, et al. (2005). INl1 protein expression dis- la M (2013). Visual and auditory hallucinations
PMID:12015749 genetic and epigenetic mechanisms driving tinguishes atypical teratoid/rhabdoid tumor from revealing cerebellar extraventricular neurocyto-
1159A. Jhawar BS, Fuchs CS, Colditz GA, paediatric diffuse high-grade glioma. Nat Rev choroid plexus carcinoma. J Neuropathol Exp ma: uncommon presentation for uncommon
Stampfer MJ (2003). Sex steroid hormone ex- Cancer. 14(10):14. PMID:25230881 Neural. 64(5):391-7. PMID15892296 tumor in uncommon location. Gen Hosp Psychi-
posures and risk for meningioma. J Neurosurg. 1176. Jones DT, Hutter B, Jager N, Korshunov 1191. Judkins AR, Ellison DW (2010). Epen- atry. 35(6):680.e1-3. PMID:24199787
99(5):841>-53. PMID:14609164 A, Kool M, Warnatz HJ, et al.; International dymoblastoma: dear, damned, distracting dia- 1207. Kalyan-Raman UP, Olivero WC (1987).
1160. Jiao Y, Killela PJ, Reitman ZJ, Rasheed Cancer Genome Consortium PedBrain Tumor gnosis, farewel!I'. Brain Pathol. 20(1):133-9. Ganglioglioma: a correlative clinicopathological
AB, Heaphy CM, de Wilde RF, et al. (2012). Fre- Project (2013). Recurrent somatic alterations PMID:19120373 and radiological study of ten surgically treated
quent ATRX, CIC, FUBP1 and IDH1 mutations of FGFR1 and NTRK2 in pilocytic asttocytoma. 1192. Judkins AR, Mauger J, Ht A, Rorke cases with follow-up. Neurosurgery. 20(3):428-
refine the classification of malignant gliomas. Nat Genet. 45(8):927-32. PMID:23817572 LB, Biegel JA (2004). lmmunohistochemi- 33. PMID:3574619
Oncotarget. 3(7):709-22. PMID:22869205 1177. Jones DT, lchimura K, Liu L, Pearson cal analysis of hSNF5/INl1 in pediatric CNS 1208. Kambham N, Chang Y, Matsushima AY
1161. Jimsheleishvili S, Alshareef AT, Papadi- DM, Plant K, Collins VP (2006). Genomic ana- neoplasms. Am J Surg Pathol. 28(5):644-50. (1998). Primary low-grade B-cell lymphoma of
mitriou K, Bregy A, Shah AH, Graham RM, et lysis of pilocytic astrocytomas at 0.97 Mb re- PMID:15105654 mucosa-associated lymphoid tissue (MALT)
al. (2014). Extracranial glioblastoma in trans- solution shows an increasing tendency toward 1192A. Jung KW, Ha J, lee SH, Won Y J, Yoo H arising in dura. Clin Neuropathol. 17(6):311-7.
plant recipients. J Cancer Res Clin Oneal. chromosomal copy number change with age. (2013). An updated nationwide epidemiology of PMID:9832258
140(5):801-7. PMID:24595597 J Neuropathol Exp Neural. 65(11):1049-58. primary brain tumors in republic of Korea. Brain 1209. Kamoshima Y, Sawamura Y, Sugiyama
1162. Jo VY, Fletcher CD (2015). Epithelioid PMID:17086101 Tumor Res Treat. 1(1):16-23. PMID:24904884 T, Yamaguchi S, Houkin K, Kubota K (2011}.
malignant peripheral nerve sheath tumor: clini- 1178. Jones DT, Kocialkowski S, Liu l, Pe- 1193. Jung SM, Kuo rr (2005). lmmunoreacti- Primary central nervous system mucosa-as-
copathologic analysis of 63 cases. Am J Surg arson DM, Backlund LM, lchimura K, et al. vity of CD10 and inhibin alpha in differentiating sociated lymphoid tissue lymphoma-case re-
Pathol. 39(5):673-82. PMID:25602794 (2008). Tandem duplication producing a novel hemangioblastoma of central nervous system port. Neurol Med Chir (Tokyo). 51(7):527-30.
1163. Jozwiak S, Kwiatkowski D, Kotulska K, oncogenic BRAF fusion gene defines the ma- from metastatic clear cell renal cell carcinoma. PMID:21785250
Larysz-Brysz M, Lewin-Kowalik J, Grajkowska jority of pilocytic astrocytomas. Cancer Res. Mod Pathol. 18(6):788-94. PMID:15578072 1210. Kanamori M, Kumabe T, Saito R, Yamas-
W, et al. (2004). Tuberin and hamartin expressi- 68(21):8673-7. PMID:18974108 1194. Jurco S 3rd, Nadji M, Harvey DG, Parker hita Y, Sonoda Y, Ariga H, et al. (2009). Optimal
on is reduced in the majority of subependymal 1179. Jones H, Steart PV, Weller RO (1991). JC Jr, Font RL, Morales AR (1982). Hemangi- treatment strategy for intracranial germ cell tu-
giant cell astrocytomas in tuberous sclerosis Spindle-cell glioblastoma or gliosarcoma? oblastomas: histogenesis of the stromal cell mors: a single institution analysis. J Neurosurg
complex consistent with a two-hit model of Neuropathol Appl Neurobiol. 17(3):177-87. studied by immunocytochemistry. Hum Pathol. Pediatr. 4(6):506-14. PMID:19951035
pathogenesis. J Child Neural. 19(2):102-6. PMID:1653908 13(1):13-8. PMID:6176519 1211. Kandenwein JA, Bostroem A, Feuss
PMID:15072102 1180. Jones RT, Abedalthagafi MS, Brahman- 1195. Janisch W, Janda J, link M, Pietsch T, Simon M (2011). Surgical ma-
1164. Joachim T, Ram Z, Rappaport ZH, Simon dam M, Greenfield EA, Hoang MP, Louis DN, et (1994). [Primary diffuse leptomeningeal nagement of intracranial subependymomas.

37 4 References
Acta Neurochir (Wien). 153(7):1469-75. Kramm CM, Wolff JE (2009). Anaplastic gang- 1243. Keith J, Lownie S, Ang LC (2006). 1260. Khalid L, Carone M, Dumrongpisutikul N,
PMID:21499782 lioglioma in children. J Neurooncol. 92(2):157- Co-existence of paraganglioma and myxopa- lntrapiromkul J, Bonekamp 0, Barker PB, et al.
1212. Kandt RS, Haines JL, Smith M, Northrup 63. PMID: 19043777 pillary ependymoma of the cauda equina. Acta (2012). Imaging characteristics of oligodendro-
H, Gardner RJ, Short MP, et al. (1992). Lin- 1228. Karremann M, Rausche U, Fleischhack Neuropathol. 111(6):617-8. PMI0:16718356 gliomas that predict grade. AJNR Am J Neuro-
kage of an important gene locus for tuberous G, Nathrath M, Pietsch T, Kramm CM, et al. 1244. Kelleher T, Aquilina K, Keohane C, radiol. 33(5):852-7. PMID:22268087
sclerosis to a chromosome 16 marker for poly- (2010). Clinical and epidemiological characte- O'Sullivan MG (2005). lntramedullary capillary 1261. Khanani MF, Hawkins C, Shroff M,
cystic kidney disease. Nat Genet. 2(1 ):37-41. ristics of pediatric gliosarcomas. J Neurooncol. haemangioma. Br J Neurosurg. 19(4):345-8. Dirks P, Capra M, Burger PC, et al. (2006).
PMID:1303246 97(2):257-65. PMID:19806321 PMID:16455542 Pilomyxoid astrocytoma in a patient with neuro-
1213. Kane AJ, Sughrue ME, Rutkowski MJ, 1229. Karremann M, Rausche U, Roth D, Kuhn 1245. Kelsey KT, Wrensch M, Zuo ZF, Miike fibromatosis. Pediatr Blood Cancer. 46(3):377-
Aranda D, Mills SA, Lehil M, et al. (2012). Atypia A, Pietsch T, Gielen GH, et al. (2013). Cere- R, Wieneke JK (1997). A population-based 80. PMID:15800886
predicting prognosis for intracranial extraventri- bellar location may predict an unfavourable case-control study of the CYP206 and GSTT1 1262. Khanna M, Siraj F, Chopra P, Bhalla S,
cular neurocytomas. J Neurosurg. 116(2):349- prognosis in paediatric high-grade glioma. Br J polymorphisms and malignant brain tumors. Roy S (2011). Gliosarcoma with prominent
54. PMID:22054208 Cancer. 109(4):844-51. PMID:23868007 Pharmacogenetics. 7(6):463-8. PMID:9429231 smooth muscle component (gliomyosarcoma):
1214. Kane AJ, Sughrue ME, Rutkowski MJ, 1230. Kasashima S, Oda Y, Nozaki J, Shirasaki 1246. Kepes JJ (1978). Transitional cell tumor a report of 10 cases. Indian J Pathol Microbiol.
Shangari G, Fang S, McDermott MW, et al. M, Nakanishi I (2000). A case of atypical gra- of the pituitary gland developing from a Rathke's 54(1):51-4. PMID:21393877
(2011 ). Anatomic location is a risk factor for nular cell tumor of the neurohypophysis. Pathol cleft cyst. Cancer. 41(1):337-43. PMID:626939 1263. Khuong-Quang DA, Buczkowicz P, Ra-
atypical and malignant meningiomas. Cancer. Int. 50(7):568-73. PMID:10886742 1247. Kepes JJ (1987). Astrocytomas: old and kopoulos P, Liu XY, Fontebasso AM, Bouffet
117(6):1272-8. PMID:21381014 1231. Kato K, Nakatani Y, Kanno H, lnayama newly recognized variants, their spectrum of E, et al. (2012). K27M mutation in histone
1215. Kannan K, Inagaki A, Silber J, Gorovets Y, ljiri R, Nagahara N, et al. (2004). Possible morphology and antigen expression. Can J H3.3 defines clinically and biologically distinct
D, Zhang J, Kastenhuber ER, et al. (2012). linkage between specific histological structures Neurol Sci. 14(2):109-21. PMID:3607613 subgroups of pediatric diffuse intrinsic pontine
Whole-exome sequencing identifies ATRX and aberrant reactivation of the Wnt pathway in 1248. Kepes JJ (1993). Pleomorphic xanthoast- gliomas. Acta Neuropathol. 124(3):439-47.
mutation as a key molecular determinant in lo- adamantinomatous craniopharyngioma. J Pa- rocytoma: the birth of a diagnosis and a con- PMID:22661320
wer-grade glioma. Oncotarget. 3(10):1194-203. thol. 203(3):814-21. PMID:15221941 cept. Brain Pathol. 3(3):269-74. PMID:8293186 1264. Kienast Y, von Baumgarten L, Fuhrmann
PMID23104868 1232. Kato S, Han SY, Liu W, Otsuka K, Shiba- 1249. Kepes JJ, Chen WY, Connors MH, Vo- M, Klinker! WE, Goldbrunner R, Herms J, et al.
1216. Kanner AA, Staugaitis SM, Castilla EA, ta H, Kanamaru R, et al. (2003). ·understanding gel FS (1988). "Chordoid" meningeal tumors (2010). Real-time imaging reveals the single
Chernova 0, Prayson RA, Vogelbaum MA, the function-structure and function-mutation in young individuals with peritumoral lympho- steps of brain metastasis formation. Nat Med.
et al. (2006). The impact of genotype on out- relationships of p53 tumor suppressor protein plasmacellular infiltrates causing systemic 16(1):116-22. PMID:20023634
come in oligodendroglioma: validation of the by high-resolution missense mutation analysis. manifestations of the Castleman syndrome. A 1265. Kijima C, Miyashita T, Suzuki M, Oka
loss of chromosome arm 1 p as an important Proc Natl Acad Sci U S A. 100(14):8424-9. report of seven cases. Cancer. 62(2):391-406. H, Fujii K (2012). Two cases of nevoid basal
factor in clinical decision making. J Neurosurg. PMID:12826609 PMID:3383139 cell carcinoma syndrome associated with
104(4):542-50. PMID:16619658 1233. Katoh M, Aida T. Sugimoto S, Suwamura 1250. Kepes JJ, Fulling KH, Garcia JH (1982). meningioma caused by a PTCH1 or SUFU
1217. Kanno H, Nishihara H, Oikawa M, Ozaki Y, Abe H, lsu T, et al. (1995). lmmunohistoche- The clinical significance of "adenoid" forma- germline mutation. Fam Cancer. 11(4):565-70.
Y, Murata J, Sawamura Y, et al. (2012). Expres- mical analysis of giant cell glioblastoma. Pathol tions of neoplastic astrocytes, imitating me- PMID:22829011
sion of 06-methylguanine DNA methyltransfera- Int. 45(4):275-82. PMID:7550996 tastatic carcinoma, in gliosarcomas. A review 1266. Kilday JP, Mitra B, Domerg C, Ward J,
se (MGMT) and immunohistochemical analysis 1234. Katsetos CD, Herman MM. Frankfurter A, of five cases. Clin Neuropathol. 1(4):139-50. Andreiuolo F, Osteso-lbanez T, et al. (2012).
of 12 pineal parenchymal tumors. Neuropatho- Gass P, Collins VP, Walker CC, et al. (1989). PMID:6188569 Copy number gain of 1 q25 predicts poor pro-
logy. 32(6):647-53. PMID:22458700 Cerebellar desmoplastic medulloblastomas. A 1251. Kepes JJ, Lewis RC, Vergara GG (1980). gression-free survival for pediatric intracranial
1218. Kannuki S, Bando K, Soga T, Matsumoto further immunohistochemical characterization Cerebellar astrocytoma invading the muscula- ependymomas and enables patient risk strati-
K, Hirose T (1996). [A case report of dysem- of the reticulin-free pale islands. Arch Pathol ture and soft tissues of the neck. Case report. J fication: a prospective European clinical trial co-
bryoplastic neuroepithelial tumor associated Lab Med. 113(9):1019-29 PMID:2505732 Neurosurg. 52(3):414-8. PMID:7359199 hort analysis on behalf of the Children's Cancer
with neurofibromatosis type 1 ). No Shinkei 1235. Katsetos CD, Herman MM, Krishna L, 1252. Kepes JJ, Moral LA, Wilkinson SB, Leukaemia Group (CCLG), Societe Francaise
Geka. 24(2):183-8. PMID:8849480 Vender JR, Vinores SA, Agamanolis DP, et al. Abdullah A, Llena JF (1998). Rhabdoid trans- d'Oncologie Pediatrique (SFOP), and Internati-
1219. Kapadia SB, Frisman OM, Hitchcock (1995). Calbindin-028k in subsets of medul- formation of tumor cells in meningiomas: a onal Society for Pediatric Oncology (SIOP). Clin
CL, Ellis GL, Popek EJ (1993). Melanotic neu- loblastomas and in the human medutloblasto- histologic indication of increased proliferative Cancer Res. 18(7):2001-11. PMID:22338015
roectodermal tumor of infancy. Clinicopatho- ma cell line 0283 Med. Arch Pathol Lab Med. activity: report of four cases. Am J Surg Pathol. 1267. Kilday JP, Rahman R, Dyer S, Ridley L,
logical, immunohistochemical, and flow cyto- 119(8):734-43. PMID:7646332 22(2):231-8. PMID:9500225 Lowe J, Coyle B, et al. (2009). Pediatric epen-
metric study. Am J Surg Pathol. 17(6):566-73. 1236. Katsetos CD, Krishna L, Friedberg E, 1253. Kepes JJ, Rubinstein LJ (1981). Mali- dymoma: biological perspectives. Mal Cancer
PMID:8392815 Reidy J, Karkavelas G, Savory J (1994). Lobar gnant gliomas with heavily lipidized (foamy) Res. 7(6):765-86. PMID:19531565
1220. Karafin M, Jallo GI, Ayars M, Eberhart pilocytic astrocytomas of the cerebral hemi- tumor cells: a report of three cases with im- 1268. Killela PJ, Pirozzi CJ, Healy P, Reitman
CG, Rodriguez F J (2011 ). Rosette forming spheres: II. Pathobiology-morphogenesis of munoperoxidase study. Cancer. 47(10):2451-9. ZJ, Lipp E, Rasheed BA, et al. (2014). Muta-
glioneuronal tumor in association with Noonan the eosinophilic granular bodies. Clin Neuropa- PMI0:7023643 tions in IDH1, IDH2, and in the TERT promo-
syndrome: pathobiological implications. Clin thol. 13(6):306-14. PMID:7851045 1254. Kepes JJ, Rubinstein LJ, Eng LF (1979). ter define clinically distinct subgroups of adult
Neuropathol. 30(6):297-300. PMID:22011734 1237. Kaufman DL, Heinrich BS, Willett C, Perry Pleomorphic xanthoastrocytoma: a distinctive malignant gliomas. Oncotarget. 5(6):1515-25.
1221. Karaki S, Mochida J, Lee YH, Nishimura A, Finseth F, Sobel RA, et al. (2003). Somatic meningocerebral glioma of young subjects with PMID:24722048
K, Tsutsumi Y (1999). Low-grade malignant pe- instability of the NF2 gene in schwannomatosis. relatively favorable prognosis. A study of 12 1269. Killela PJ, Pirozzi CJ, Reitman ZJ, Jones
rineurioma of the paravertebral column, trans- Arch Neurol. 60(9):1317-20. PMID:12975302 cases. Cancer. 44(5):1839-52. PMID:498051 S, Rasheed BA, Lipp E, et al. (2014). The ge-
forming into a high-grade malignancy. Pathol 1238. Kaulich K, Blaschke B, Numann A, von 1255. Kerfoot C, Wienecke R, Menchine M, netic landscape of anaplastic astrocytoma. On-
Int. 49(9):820-5. PMID:10504555 Deimling A, Wiestler OD, Weber RG, et al. Emelin J, Maize JC Jr, Welsh CT, et al. (1996). cotarget. 5(6):1452-7. PMID:24140581
1222. Karamchandani JR, Nielsen TO, van de (2002). Genetic alterations commonly found in Localization of tuberous sclerosis 2 mRNA and 1270. Killela PJ, Reitman ZJ, Jiao Y, Bettegow-
Rijn M, West RB (2012). Sox10 and S100 in diffusely infiltrating cerebral gliomas are rare its protein product tuberin in normal human da C,Agrawal N, Diaz LA Jr, etal. (2013). TERT
the diagnosis of soft-tissue neoplasms. Appl or absent in pleomorphic xanthoastrocytomas. brain and in cerebral lesions of patients with promoter mutations occur frequently in gliomas
lmmunohistochem Mol Morphol. 20(5):445-50. J Neuropathol Exp Neurol. 61(12):1092-9. tuberous sclerosis. Brain Pathol. 6(4):367-75. and a subset of tumors derived from cells with
PMID:22495377 PMID:12484572 PMID:8944308 low rates of self-renewal. Proc Natl Acad Sci U
1223. Karamitopoulou E, Perentes E, Diaman- 1239. Kaur B, Khwaja FW, Severson EA, Ma- 1256. Kesari S, Schiff 0, Drappatz J, LaFrankie SA. 110(15):6021-6. PMID:23530248
tis I, Maraziotis T (1994). Ki-67 immunoreacti- theny SL, Brat OJ, Van Meir EG (2005). Hypo- 0, Doherty L, Macklin EA, et al. (2009). Phase 1271. Kim B, Chung CK, Myung JK, Park SH
vity in human central nervous system tumors: xia and the hypoxia-inducible-factor pathway in II study of protracted daily temozolomide for (2009). Pleomorphic xanthoastrocytoma asso-
a study with MIB 1 monoclonal antibody on ar- glioma growth and angiogenesis. Neuro Oneal. low-grade gliomas in adults. Clin Cancer Res. ciated with long-standing Taylor-type IIB-focal
chival material. Acta Neuropathol. 87(1):47-54. 7(2):134-53. PMID:15831232 15(1):330-7. PMID:19118062 cortical dysplasia in an adult. Pathol Res Pract.
PMID:7511316 1240. Kaur K, Kakkar A, Kumar A, Mallick S, 1257. Keser H, Barnes M, Moes G, Lee HS, 205(2):113--7. PMI0:18657915
1224. Karnes PS, Tran TN, Cui MY, Raffel C, Julka PK, Gupta 0, et al. (2015). Integrating Tihan T (2014). Well-differentiated pediatric 1272. Kim BS, Kim DK, Park SH (2009). Pineal
Gilles FH, Barranger JA, et al. (1992). Cytoge- molecular subclassification of medulloblasto- glial neoplasms with features of oligodendrogli- parenchymal tumor of intermediate differentiati-
netic analysis of 39 pediatric central nervous mas into routine clinical practice: A simplified oma, angiocentric glioma and dysembryoplastic on showing malignant progression at relapse.
system tumors. Cancer Genet Cytogenet. approach. Brain Pathol. PMID:26222673 neuroepithelial tumors: a morphological diagno- Neuropathology. 29(5):602-8. PMID:19170892
59(1):12-9. PMID:1313329 1241. Kawano N, Yasui Y, Utsuki S, Oka H, Fujii stic challenge. Turk Patoloji Derg. 30(1):23-9. 1273. Kim DG, Lee DY, Paek SH, Chi JG, Choe
1225. Karpinski NC, Yaghmai R, Barba 0, K, Yamashina S (2004 ). Light microscopic de- PMID:24448703 G, Jung HW (2002). Supratentorial primitive
Hansen LA (1999). Case of the month: March monstration of the microlumen of ependymoma: 1258. Kessler BA, Boekhout C, Jaikumar S, neuroectodermal tumors in adults. J Neuroon-
1999-A 26 year old HIV positive male with a study of the usefulness of antigen retrieval Hipps J, Lee YZ (2015). Disseminated oligo- col. 60(1):43-52. PMI0:12416545
dura based masses. Brain Pathol. 9(3):609-10. for epithelial membrane antigen (EMA) immu- dendroglial-like leptomeningeal tumor with ana- 1274. Kim DH, Suh YL (1997). Pseudopapillary
PMID:10416997 nostaining. Brain Tumor Pathol. 21(1):17-21. plastic progression and presumed extraneural neurocytoma of temporal lobe with glial diffe-
1226. Karremann M, Butenhoff S, Rausche U, PMID:15696964 disease: case report. Clin Imaging. 39(2):300- rentiation. Acta Neuropathol. 94(2):187-91.
Pietsch T, Wolff JE, Kramm CM (2009). Pedi- 1242. Kehrer-Sawatzki H, Cooper ON (2008). 4. PMID:25518979 PMID:9255395
atric giant cell glioblastoma: New insights into Mosaicism in sporadic neurofibromatosis type 1259. Khalatbari MR, Hamidi M, Moharamzad Y 1275. Kim ES, Kwon MJ, Song JH, Kim DH,
a rare tumor entity. Neuro Oncol. 11(3):323--9. 1: variations on a theme common to other (2013). Primary alveolar rhabdomyosarcoma of Park HR (2015). Adenocarcinoma arising
PMID:19050301 hereditary cancer syndromes? J Med Genet. the brain with long-term survival. J Neurooncol. from intracranial recurrent mature teratoma
1227. Karremann M, Pietsch T. Janssen G, 45(10):622-31. PMID:18511569 115(1):131-3. PMID:23857335 and featuring mutated KRAS and wild-type

References 375
J'

BRAF genes. Neuropathology. 35(1):44-9. Tumours. Pathology and Genetics of Tumours in sporadic neurofibromatosis 2 patients. Hum PMID:23530928
PMID:25039399 of the Nervous System. 3rd ed. Lyon: IARC Mel Genet. 7(13):2051-5. PMID:9817921 1324. Komori T, Scheithauer SW, Anthony DC,
1276. Kim KH, Roberts CW (2014). Mechanis- Press. 1309. Knobbe CB, Trampe-Kieslich A, Rei· Rosenblum MK, Mclendon RE, Scott RM, et
ms by which SMAR CB 1 loss drives rhabdoid 1292. Kleihues P, Kiessling M, Janzer RC fenberger G (2005). Genetic alteration and al. (1998). Papillary glioneuronal tumor: a new
tumor growth. Cancer Genet. 207(9):365-72. (1987). Morphological markers in neuro-oncolo- expression of the phosphoinositol-3-kinase/ variant of mixed neuronal-glial neoplasm. Am J
PMID:24853101 gy. Curr Top Pathol. 77:307-38. PMID:2827963 Akt pathway genes PIK3CA and PIKE in human Surg Pathol. 22(10):1171-83. PMID:9777979
1277. Kim MS, Kim YS, Lee HK, Lee GJ, Choi 1293. Kleihues P, Louis ON, Scheithauer glioblastomas. Neuropathol Appl Neurobiol. 1325. Komori T, Scheithauer BW, Hirose T
CY, Lee CH (2014). Primary intracranial ectopic SW, Rorke LB, Reifenberger G, Burger PC, 31(5):486-90. PMID:16150119 (2002). A rosette-forming glioneuronal tumor of
craniopharyngioma in a patient with probable et al. (2002). The WHO classification of tu- 1310. Knowles OM (1999). Immunodeficien- the fourth ventricle: infratentorial form of dysem-
Gardner's syndrome. J Neurosurg. 120(2):337- mors of the nervous system. J Neuropathol cy-associated lymphoproliferative disorders. bryoplastic neuroepithelial tumor? Am J Surg
41. PMID:24266539 Exp Neural. 61(3):215-25, discussion 226-9. Mod Pathol. 12(2):200-17. PMID:10071343 Pathol. 26(5):582-91. PMID:11979088 .
1278. Kim SD, Nakagawa H, Mizuno J, Inoue PMID:11895036 1311. Ko LJ, Prives C (1996). p53: puzzle 1326. Komotar RJ, Burger PC, Carson BS
T (2005). Thoracic subpial intramedullary 1294. Kleihues P, Schauble B, zur Hausen A, and paradigm. Genes Dev. 10(9):1054-72. Brem H, Olivi A,_ Goldthwaite PT, et al. (2004)'.
schwannoma involving a ventral nerve root: Esteve J, Ohgaki H (1997). Tumors associa- PMID:8654922 Pilocytic and pilornyxoid hypothalamic/chias-
a case report and review of the literature. ted with p53 germline mutations: a synopsis 1312. Kechi N, Budka H (1987). Contribution matic astrocytomas. Neurosurgery. 54(1):72-9,
Surg Neural. 63(4):389-93, discussion 393. of 91 families. Am J Pathol. 150(1):1-13. of histiocytic cells to sarcomatous develop- discussion 79-80. PMID:14683543
PMID:15808734 PMID:9006316 ment of the gliosarcoma. An immunohistoche- 1327. Komuro Y, Mikami M, Sakaiya N, Ku-
1279. Kim Y, Lee SY, Yi KS, Cha SH, Gang MH, 1295. Kleinman CL, Gerges N, Papillon-Ca- mical study. Acta Neuropathol. 73(2):124-30. rahashi T, Komiyama S, Tei C, et al. (2001).
Cho BS, et al. (2014). lnfratentorial and intrapa- vanagh S, Sin-Chan P, Pramatarova A, Quang PMID:3111162 Tumor imprint cytology of ovarian ependymo-
renchymal subependymoma in the cerebellum: DA, et al. (2014). Fusion of TTYH1 with the 1313. Koelsche C, Hovestadt V, Jones OT, Cap- ma. A case report. Cancer. 92(12):3165-9.
case report. Korean J Radial. 15(1):151-5. C19MC microRNA cluster drives expression per D, Sturm D, Sahm F, et al. (2015). Melanotic PMID:11753996
PMID:24497806 of a brain-specific DNMT38 isoform in the tumors of the nervous system are characterized 1328. Kondziolka D, Parry PV, Lunsford LO
1280. Kim YH, Kim JW, Park CK, Kim DG, Sohn embryonal brain tumor ETMR. Nat Genet. by distinct mutational, chromosomal and epi- Kano H, Flickinger JC, Rakfal S, et al. (2014)'.
CH, Chang KH, et al. (2010). Papillary tumor of 46(1):39-44. PMID:24316981 genomic profiles. Brain Pathol. 25(2):202-8. The accuracy of predicting survival in individual
pineal region presenting with leptomeninge- 1296. Kleinman GM, Schoene WC, Walshe PMID:25399693 patients with cancer. J Neurosurg. 120(1):24-
al seeding. Neuropathology. 30(6):654-60. TM 3rd, Richardson EP Jr (1978). Malignant 1314. Koelsche C, Sahm F, Capper D, Reuss D, 30. PMID:24160479
PMID:20374498 transformation in benign cerebellar astrocyto- Sturm D, Jones OT, et al. (2013). Distribution of 1329. Kong LY, Wei J, Haider AS, Liebelt BO
1281. Kim YH, Nobusawa S, Mittelbronn ma. Case report. J Neurosurg. 49(1):111-8. TERT promoter mutations in pediatric and adult Ling X, Conrad CA, et al. (2014). Therapeuti�
M, Paulus W, Brokinkel B, Keyvani K, et al. PMID:660255 tumors of the nervous system. Acta Neuropa- targets in subependymoma. J Neuroimmunol.
(2010). Molecular classification of low-grade 1297. Kleinschmidt-DeMasters BK. Aisner thol. 126(6):907-15. PMID:24154961 277(1-2):168-75. PMID:25465288
diffuse gliomas. Am J Pathol. 177(6):2708-14. DL, Birks DK, Foreman NK (2013). Epithelioid 1315. Koelsche C, Sahm F, Paulus W, Mittel- 1330. Konishi E, lbayashi N, Yamamoto S,
PMID:21075857 GB Ms show a high percentage of BRAF V600E bronn M, Giangaspero F, Antonelli M, et al. Scheithauer BW (2003). Isolated intracranial
1282. Kim YH, Nonoguchi N, Paulus W, Bro- mutation. Am J Surg Pathol. 37(5):685-98. (2014). BRAF V600E expression and distri- Rosai-Dorfman disease (sinus histiocytosis with
kinkel B, Keyvani K, Sure U, et al. (2012). Fre- PMID23552385 bution in desmoplastic infantile astrocytoma/ massive lymphadenopathy). AJNR Am J Neuro-
quent BRAF gain in low-grade diffuse gliomas 1298. Kleinschmidt-DeMasters BK, Aisner DL, ganglioglioma. Neuropathol Appl Neurobiol. radiol. 24(3):515-8. PMID:12637307
with 1p/19q loss. Brain Pathol. 22(6):834-40. Foreman NK (2015). BRAF VE1 immunore- 40(3):337-44. PMID:23822828 1331. Konno S, Oka H, Utsuki S, Kondou K,
PMID:22568401 activity patterns in epithelioid glioblastomas 1316. Koelsche C, Sahm F, Wohrer A, Jeib· Tanaka S, Fujii K, et al. (2002). Germinoma with
1283. Kim YH, Ohta T, Oh JE, Le Calvez-Kelm positive for BRAF V600E mutation. Am J Surg mann A, Schittenhelm J, Kohlhof P, et al. a granulomatous reaction. Problems of differen-
F, McKay J, Voegele C, et al. (2014). TP53, Pathol. 39(4):528-40. PMID:25581727 (2014). BRAF-mutated pleomorphic xanthoast- tial diagnosis. Clin Neuropathol. 21(6):248-51.
MSH4, and LATS1 germline mutations in a fa- 1299. Kleinschmidt-DeMasters BK, Alassiri rocytoma is associated with temporal location, PMID: 12489672
mily with clustering of nervous system tumors. AH, Birks DK, Newell KL, Moore W, Lillehei KO reticulin fiber deposition and CD34 expression. 1332. Konovalov AN, Pitskhelauri DI (2003).
Am J Pathol. 184(9):2374-81. PMID:25041856 (2010). Epithelioid versus rhabdoid glioblasto- Brain Pathol. 24(3):221-9. PMID:24345274 Principles of treatment of the pineal re-
1284. Kimonis VE, Goldstein AM, Pastakia B, mas are distinguished by monosomy 22 and 1317. Koelsche C, Schweizer L, Renner M, gion tumors. Surg Neural. 59(4):250-68.
Yang ML, Kase R, DiGiovanna JJ, et al. (1997). immunohistochemical expression of INl-1 but Warth A, Jones OT, Sahm F, et al. (2014). PMID:127 48006
Clinical manifestations in 105 persons with ne- not claudin 6. Am J Surg Pathol. 34(3):341-54. Nuclear relocation of STAT6 reliably predicts 1333. Kool M. Jones OT, Jager N, Northcott PA,
void basal cell carcinoma syndrome. Am J Med PMID:20118769 NAB2-STAT6 fusion for the diagnosis of solitary Pugh T J, Hovestadt V, et al.; ICGC PedBrain
Genet. 69(3):299-308. PMID 9096761 1300. Kleinschmidt-DeMasters BK, Boylan A, fibrous tumour. Histopathology. 65(5):613-22. Tumor Project (2014). Genome sequencing of
1285. Kimura T, Budka H, Soler-Federsppiel S Capocelli K, Boyer PJ, Foreman NK (2011). PMID:24702701 SHH medulloblastoma predicts genotype-rela·
(1986). An immunocytochemical comparison of Multinodular leptomeningeal metastases from 1318. Koelsche C, Wohrer A. Jeibmann A, ted response to smoothened inhibition. Cancer
the glia,associated proteins glial fibrillary acidic ETANTR contain both small blue cell and ma- Schittenhelm J, Schindler G, Preusser M, et al. Cell. 25(3):393-405. PMID:24651015
protein (GFAP) and S-100 protein (S100P) in turing neuropil elements. Acta Neuropathol. (2013). Mutant BRAF V600E protein in ganglia· 1334. Kool M, Korshunov A, Remke M, Jones
human brain tumors. Clin Neuropathol. 5(1):21- 122(6):783-5 PMID:22033877 glioma is predominantly expressed by neuronal OT, Schlanstein M, Northcott PA, et al. (2012).
7. PMID:3512139 1301. Kleinschmidt-DeMasters BK, Lopes MB tumor cells. Acta Neuropathol. 125(6):891-900. Molecular subgroups of medulloblastoma:
1286. Kinsler VA, Thomas AC, Ishida M, Bui- (2013). Update on hypophysitis and TTF-1 PMID:23435618 an international meta-analysis of transcripto-
strode NW, Loughlin S, Hing S, et al. (2013). expressing sellar region masses. Brain Pathol. 1319. Koen JL, Mclendon RE, George TM me, genetic aberrations, and clinical data of
Multiple congenital melanocytic nevi and neu- 23(5):495-514. PMID:23701182 (1998). lntradural spinal teratoma: evidence for WNT, SHH. Group 3, and Group 4 medullob-
rocutaneous melanosis are caused by postzy- 1302. Kleinschmidt-DeMasters BK, Meltesen a dysembryogenic origin. Report of four cases. lastomas. Acta Neuropathol. 123(4):473-84.
gotic mutations in codon 61 of NRAS. J Invest L, McGavran L, Lillehei KO (2006). Characte- J Neurosurg. 89(5):844-51. PMID:9817426 PMID:22358457
Dermatol. 133(9):2229-36. PMID:23392294 rization of glioblastomas in young adults. Brain 1320. Koga T, Iwasaki H, lshiguro M, Matsuzaki 1335. Kool M, Koster J, Bunt J, Hassell NE, La·
1287. Kirkpatrick PJ, Honavar M, Janota Pathol. 16(4):273-86. PMID:17107596 A, Kikuchi M (2002). Losses in chromosomes keman A, van Sluis P, et al. (2008). Integrated
I, Polkey CE (1993). Control of temporal 1303. Kliewer KE, Cochran AJ (1989). A review 17, 19, and 22q in neurofibromatosis type 1 genomics identifies five medulloblastoma sub-
lobe epilepsy following en bloc resection of of the histology, ultrastructure, immunohisto- and sporadic neurofibromas: a comparative types with distinct genetic profiles, pathway sig·
low-grade tumors. J Neurosurg. 78(1):19-25. logy, and molecular biology of extra-adren- genomic hybridization analysis. Cancer Genet natures and clinicopathological features. PLoS
PMID:8416237 al paragangliomas. Arch Pathol Lab Med. Cytogenet. 136(2):113-20. PMID: 12237234 One. 3(8):e3088. PMID:18769486
1288. Kissil JL, Wilker EW, Johnson KC, Eck- 113(11):1209-18. PMID:2684087 1320A. Kogerman P. Grimm T, Kogerman 1336. Koperek 0, Gelpi E, Birner P, Haberler
man MS, Yaffe MB, Jacks T (2003). Merlin, the 1304. Klintworth GK, Garner A, editors. (2008). L, Krause D. Unden AB, Sandstedt B. et al. C, Budka H. Hainfellner JA (2004). Value and
product of the Nf2 tumor suppressor gene. is an Garner and Klintworth's Pathobiology of ocular (1999). Mammalian suppressor-of-fused modu- limits of immunohistochemistry in differential di·
inhibitor of the p21-activated kinase, Pak1. Mol disease. 3rd Boca Raton. CRC Press. lates nuclear-cytoplasmic shuttling of Gli-1. Nat agnosis of clear cell primary brain tumors. Acta
Cell. 12(4):841-9. PMID:14580336 1305. Kloub 0, Perry A, Tu PH, Lipper M, Cell Biol. 1(5):312-9. PMID:10559945 Neuropathol. 108(1):24-30. PMID:15108012
1288A. Kita D, Yonekawa Y, Weller M, Ohgaki H Lopes MB (2005). Spindle cell oncocytoma 1321. Komakula S, Warmuth-Metz M, Hilden- 1337. Koral K, Koral KM, Sklar F (2012). An·
(2007). PIK3CA alterations in primary (de nova) of the adenohypophysis: report of two recur- brand P, Loevner L, Hewlett R, Salzman K, et giocentric glioma in a 4-year-old boy: imaging
and secondary glioblastomas. Acta Neuropa- rent cases. Am J Surg Pathol. 29(2):247-53. al. (2011). Pineal parenchymal tumor of inter- characteristics and review of the literature. Clin
thol. 113(3):295-302. PMID:17235514 PMID:15644783 mediate differentiation: imaging spectrum of Imaging. 36(1):61-4. PMID:22226445
1289. Kleihues P, Burger PC, Scheithauer 1306. Kluwe L, MacCollin M, Tatagiba M, Tho- an unusual tumor in 11 cases. Neuroradiology. 1338. Koral K, Weprin B, Rollins NK (2006).
SW (1993). The new WHO classification of mas S, Hazim W, Haase W, et al. (1998). Phe- 53(8):577-84. PMID:21080159 Sphenoid sinus craniopharyngioma simu-
brain tumours. Brain Pathol. 3(3):255-68. notypic variability associated with 14 splice-site 1322. Komakula ST, Fenton LZ, Klein- lating mucocele. Acta Radial. 4 7(5):494-6.
PMID:8293185 mutations in the NF2 gene. Am J Med Genet. schmidt-DeMasters BK, Foreman NK (2007). PMID:16796313
1290. Kleihues P, Burger PC, Scheithauer BW, 77(3):228-33. PMID:9605590 Pilomyxoid astrocytoma: neuroimaging with 1339. Kordek R, Biernat W, Sapieja W, Alwasiak
editors. (1993). Histological Typing of Tumours 1307. Kluwe L, Mautner V, Heinrich B, Dezube clinicopathologic correlates in 4 cases followed J, Liberski PP (1995). Pleomorphic xanthoast-
of the Central Nervous System. World Health R, Jacoby LB, Friedrich RE, et al. (2003). Mole- over time. J Pediatr Hematol Oneel. 29(7):465- rocytoma with a gangliomatous component: an
Organization International Histological Classifi- cular study of frequency of mosaicism in neuro- 70. PMID:17609624 immunohistochemical and ultrastructural study.
cation of Tumours. 2nd ed. Berlin, Heidelberg: fibromatosis 2 patients with bilateral vestibular 1323. Komori T, Arai N (2013). Dysembryo- Acta Neuropathol. 89(2):194-7. PMID:7732793
Springer Verlag. schwannomas. J Med Genet. 40(2):109-14. plastic neuroepithelial tumor, a pure glial 1340. Korfel A, Schlegel U (2013). Diagnosis
1291. Kleihues P, Cavenee WK, editors. (2000). PMID:12566519 tumor? lmmunohistochemical and morpheme- and treatment of primary CNS lymphoma. Nat
World Health Organization Classification of 1308. Kluwe L, Mautner VF (1998). Mosaicism tric studies. Neuropathology. 33(4):459-68. Rev Neural. 9(6):317-27. PMID:23670107

376 References
1341. Korfel A, Weller M, Marius P, Roth P, correlated with increased Ki-67 labeling index. PMID:17549014 1387. Kujas M, Faillot T, Lalam T, Roncier B,
Klasen HA, Roeth A, et al. (2012). Prognostic Neurol India. 57(2):191-3. PMID:19439853 1371. Kros JM, Hop WC, Godschalk JJ, Krish- Catala M, Poirier J (2000). Astroblastomas re-
,mpact of meningeal dissemination in primary 1355. Kowalski RJ, Prayson RA, Mayberg nadath KK (1996). Prognostic value of the proli- visited. Report of two cases with immunocyto-
CNS lymphoma (PCNSL): experience from the MR (2004). Piluicyloma. Ann Diagn Pathol. feration-related antigen Ki-67 in oligodendrogli- chemical and electron microscopic study.
G-PCNSL-SG1 trial. Ann Oncol. 23(9):2374- 8(5):290-4. PMID:15494936 omas. Cancer. 78(5):1107-13. PMID:8780550 Histogenetic considerations. Neuropathol Appl
80. PMID:22396446 1356. Kozak KR, Moody JS (2009). Giant cell 1372. Kros JM, Schouten WC, Janssen PJ, Neurobiol. 26(3):295-8. PMID:10886687
1342. Kornreich L, Blaser S, Schwarz M, glioblastoma: a glioblastoma subtype with dis- van der Kwast TH (1996). Proliferation of ge- 1388. Kukreja S, Ambekar S, Sharma M, Sin
Shuper A, Vishne TH, Cohen IJ, et al. (2001). tinct epidemiology and superior prognosis. Neu- mistocytic cells and glial fibrillary acidic protein AH, Nanda A (2015). Outcome predictors in the
Optic pathway glioma: correlation of imaging ro Oncol. 11(6):833-41. PMID:19332771 (GFAP)-positive oligodendroglial cells in glio- management of spinal myxopapillary ependy-
findings with the presence of neurofibromato- 1357. Kraan W, Horlings HM, van Keimpema M, mas: a MIB-1/GFAP double labeling study. Acta moma: an integrative survival analysis. World
sis. AJNR Am J Neuroradiol. 22(10):1963-9. Schilder-Toi EJ, Oud ME, Scheepstra C, et al. Neuropathol. 91(1):99-103. PMID:8773153 Neurosurg. 83(5):852-9 PMID:25108296
PMID:11733333 (2013). High prevalence of oncogenic MYD88 1373. Kros JM, van den Brink WA, van Loon- 1389. Kukreja S, Ambekar S, Sin AH, Nanda A
1343. Korogi Y, Takahashi M, Ushio Y (2001). and CD79B mutations in diffuse large B-cell van Luyt JJ, Stefanko SZ (1997). Signet-ring (2014). Cumulative survival analysis of patients
MRI of pineal region tumors. J Neurooncol. lymphomas presenting at immune-privileged cell oligodendroglioma-report of two cases and with spinal myxopapillary ependymomas in the
54(3):251-61. PMID:11767291 sites. Blood Cancer J. 3:e139. PMID:24013661 discussion of lhe differential diagnosis. Acta first 2 decades of life. J Neurosurg Pediatr.
1344. Korshunov A, Golanov A (2001 ). The 1358. Kramm CM, Butenhoff S, Rausche U, Neuropathol. 93(6):638-43. PMID:9194905 13(4):400-7. PMID:24527863
prognostic significance of DNA topoisomera- Warmuth-Metz M, Kortmann RD, Pietsch T, el 1374. Kros JM, Van Eden CG, Stefanko SZ, 1390. Kulkarni AV, Pierre-Kahn A, Zerah M
se II-alpha (Ki-S1), p21/Cip-1, and p27/Kip-1 al. (2011 ). Thalamic high-grade gliomas in chil- Waayer-Van Batenburg M, van der Kwast (2004). Spontaneous regression of congenital
protein immunoexpression in oligodendrogli- dren: a distinct clinical subset? Neuro Oncol. TH (1990). Prognostic implications of glial spinal lipomas of the conus medullaris. Report
omas. Arch Pathol Lab Med. 125(7):892-8. 13(6):680-9. PMID:21636712 fibrillary acidic protein containing cell types in of two cases. J Neurosurg. 101(2) Suppl:226-7.
PMID:11419973 1359. Kraus JA, Koopmann J, Kaskel P, Maintz oligodendrogliomas. Cancer. 66(6):1204-12. PMID:15835113
1345. Korshunov A, Golanov A, Sycheva R, D, Brandner S, Schramm J, et al. (1995). Sha- PMID:2205356 1391. Kumar A, Pathak P, Purkait S, Faruq
T1mirgaz V (2004). The histologic grade is a red allelic losses on chromosomes 1 p and 19q 1375. Kros JM, Vecht CJ, Stefanko SZ (1991). M, Jha P, Mallick S, et al. (2015). Oncogenic
main prognostic factor for patients with intracra- suggest a common origin of oligodendroglioma The pleomorphic xanthoastrocytoma and its dif- KIAA1549-BRAF fusion with activation of
nial ependymomas treated in the microneuro- and oligoastrocytoma. J Neuropathol Exp Neu- ferential diagnosis: a study of five cases. Hum the MAPK/ERK pathway in pediatric oligo-
surgical era: an analysis of 258 patients. Can- rol. 54(1):91-5. PMID:7815084 Pathol. 22(11):1128-35. PMID:1743696 dendrogliomas. Cancer Genet. 208(3):91-5.
cer. 100(6) 1230-7. PMID:15022291 1360. Kraus JA, Lamszus K, Glesmann N, Beck 1376. Krossnes BK, Wester K, Moen G, M0rk PMID:25794445
1345A. Korshunov A, Jakobiec FA, Eberhart M, Wolter M, Sabel M, et al. (2001). Molecular SJ (2005). Multifocal dysembryoplastic neu- 1392. Kumar S, Kumar D, Kaldjian EP, Bau-
CG, Hovestadt V, Capper D, Jones DT, et al. genetic alterations in glioblastomas with oli- roepithelial tumour in a male with the XYY syn- serman S, Raffeld M, Jaffe ES (1997). Primary
(2015). Comparative integrated molecular godendroglial component. Acta Neuropathol. drome. Neuropathol Appl Neurobiol. 31(5):556- low-grade B-cell lymphoma of the dura: a muco-
analysis of intraocular medulloepitheliomas 101(4):311-20. PMID:11355302 60. PMID:16150126 sa associated lymphoid tissue-type lymphoma.
and central nervous system embryonal tumors 1361. Kreiger PA, Okada Y, Simon S, Rorke 1377. Krouwer HG, Davis RL, Silver P, Prados Am J Surg Pathol. 21(1):81-7. PMID8990144
with multilayered rosettes confirms that they LB, Louis DN, Golden JA (2005). Losses of M (1991). Gemistocytic astrocytomas: a 1393. Kuratsu J, Matsukado Y, Sonoda H
are distinct nosologic entities. Neuropathology. chromosomes 1 p and 19q are rare in pedi- reappraisal. J Neurosurg. 74(3):399-406. (1983). Pseudopsammoma bodies in meningo-
35(6):538-44. PMID:26183384 atric oligodendrogliomas. Acta Neuropalhol. PMID:1993905 theliomatous meningioma. A histochemical and
1346. Korshunov A, Remke M, Gessi M, Ryz- 109(4):387-92. PMID:15739101 1378. Krueger DA, Northrup H; International ultrastructural study. Acta Neurochir (Wien).
hova M, Hielscher T, Witt H, et al. (2010). Focal 1362. Kresse SH, Skarn M, Ohnstad HO, Nam- Tuberous Sclerosis Complex Consensus Group 68(1-2):55-62. PMID6858731
genomic amplification al 19q13.42 comprises les HM, Bjerkehagen B, Myklebost 0, et al. (2013). Tuberous sclerosis complex surveillan- 1394. Kurian KM, Summers DM, Statham PF,
a powerful diagnostic marker for embryonal (2008). DNA copy number changes in high-gra- ce and management: recommendations of the Smith C, Bell JE, Ironside JW (2005). Third ven-
tumors with ependymoblaslic rosettes. Acta de malignant peripheral nerve sheath tumors by 2012 International Tuberous Sclerosis Com- tricular chordoid glioma: clinicopathological stu-
Neuropathol. 120(2):253-60. PMID:20407781 array CGH. Mol Cancer. 7:48. PMID:18522746 plex Consensus Conference. Pediatr Neurol. dy of two cases with evidence for a poor clinical
1347. Korshunov A, Remke M, Werft W, Ben- 1363. Krieg M, Marti HH, Plate KH (1998). 49(4 ):255-65. PMID:24053983 outcome despite low grade histological featu-
ner A, Ryzhova M, Witt H, et al. (2010). Adult Coexpression of erythropoietin and vascular 1379. Krutilkova V, Trkova M, Fleitz J, Gregor V, res. Neuropathol Appl Neurobiol. 31(4):354-61.
and pediatric medulloblastomas are geneti- endothelial growth factor in nervous system Novotna K, Krepelova A, et al. (2005). Identifi- PMID:16008819
cally distinct and require different algorithms tumors associated with von Hippel-lindau tu- cation of five new families strengthens the link 1395. Kuroiwa T, Bergey GK, Rothman Ml,
for molecular risk stratification. J Clin Oncol. mor suppressor gene loss of function. Blood. between childhood choroid plexus carcinoma Zoarski GH, Wolf A, Zagardo MT, et al. (1995).
28(18):3054-60. PMID:20479417 92(9):3388-93 PMID:9787178 and germline TP53 mutations. Eur J Cancer. Radiologic appearance of the dysembryoplastic
1348. Korshunov A, Ryzhova M, Jones DT, 1364. Krieger MD, Gonzalez-Gomez I, Levy 41(11):1597-603. PMID15925506 neuroepithelial tumor. Radiology. 197(1 ):233-8.
Northcott PA, van Sluis P, Volckmann R, et al. ML, McComb JG (1997). Recurrence patterns 1380. Kubo 0, Sasahara A, Tajika Y, Kawamura PMID:7568829
(2012). LIN28A immunoreactivity is a potent and anaplastic change in a long-term study H, Kawabatake H, Takakura K (1996). Pleomor- 1396. Kurose K, Araki T, Matsunaka T, Takada
diagnostic marker of embryonal tumor with mul- of pilocytic astrocytomas. Pediatr Neurosurg. phic xanthoastrocytoma with neurofibromatosis Y, Emi M (1999). Variant manifestation of Cow-
tilayered rosettes (ETMR). Acta Neuropathol. 27(1):1-11. PMID:9486830 type 1: case report. Noshuyo Byori. 13(1):79- den disease in Japan: hamartomatous polypo-
124(6):875-81. PMID:23161096 1365. Krishnan S, Brown PD, Scheithauer BW, 83. PMID:8916131 sis of the digestive tract with mutation of the
1349. Korshunov A, Sturm D, Ryzhova M, Ho- Ebersold MJ, Hammack JE, Buckner JC (2004). 1381. Kubota T, Salo K, Arishima H, Takeuchi PTEN gene. Am J Hum Genet. 64(1):308-10.
vesladt V, Gessi M, Jones DT, et al. (2014). Choroid plexus papillomas: a single instituti- H, Kitai R, Nakagawa T (2006). Astroblastoma: PMID:9915974
Embryonal tumor with abundant neuropil and onal experience. J Neurooncol. 68(1):49-55. immunohistochemical and ultrastructural study 1397. Kurt E, Beute GN, Sluzewski M, van Rooij
true rosettes (ETANTR), ependymoblastoma, PMID:15174521 of distinctive epithelial and probable tanycytic WJ, Teepen JL (1996). Giant chondroma of the
and medulloepilhelioma share molecular si- 1366. Kristof RA, Van Roost D, Wolf HK, Schr- differentiation. Neuropathology. 26(1):72-81. falx. Case report and review of the literature. J
milarity and comprise a single clinicopatholo- amm J (1997). lnlravascular papillary endothe- PMID:16521483 Neurosurg. 85(6):1161-4. PMID:8929512
gical entity. Acta Neuropathol. 128(2):279-89. lial hyperplasia of the sellar region. Report of 1382. Kuchelmeister K, Demirel T, Scblorer 1398. Kurt E, Zheng PP, Hop WC, van der Wei-
PMID:24337497 three cases and review of the literature. J Neu- E, Bergmann M, Gullotta F (1995). Dysem- den M, Bol M, van den Bent MJ, et al. (2006).
1350. Korshunov A, Sycheva R, Golanov A rosurg. 86(3):558-63. PMID:9046317 bryoplastic neuroepithelial tumour of the ce- Identification of relevant prognostic histopatho-
(2007). Recurrent cylogenetic aberrations 1367. Kros J, de Greve K, van Tilborg A, Hop W, rebellum. Acta Neuropathol. 89(4):385-90. logic features in 69 intracranial ependymomas,
in central neurocytomas and their biological Pieterman H, Avezaat C, et al. (2001 ). NF2 sta- PMID:7610772 excluding myxopapillary ependymomas and
relevance. Acla Neuropathol. 113(3):303-12. tus of meningiomas is associated with Jumour 1383. Kuchelmeister K, Huqens-Penzel M, subependymomas. Cancer. 106(2):388-95.
PMID:17123091 localization and histology. J Pathol. 194(3):367- Jodicke A, Schachenmayr W (2006). Papillary PMID:16342252
1351. Korshunov A, Witt H, Hielscher T, Benner 72. PMID:11439370 tumour of the pineal region: histodiagnoslic 1399. Kurtkaya-Yapicier 0, Elmaci I, Boran B,
A, Remke M, Ryzhova M, et al. (2010). Molecu- 1368. Kros JM, Cella F, Bakker SL, Paz Y Geu- considerations. Neuropalhol Appl Neurobiol. Kili� T, Sav A, Pamir MN (2002). Dysembryo-
lar staging of inlracranial ependymoma in chil- ze D, Egeler RM, Egeler RM (2000). Papillary 32(2):203-8. PMID:16599948 plastic neuroepithelial tumor of the midbrain
dren and adults. J Clin Oncol. 28(19):3182-90. meningioma with pleural metastasis: case re- 1384. Kuchelmeister K, von Borcke IM, Klein H, teclum: a case report. Brain Tumor Pathol.
PMID:20516456 port and literature review. Acta Neurol Scand. Bergmann M, Gullotta F (1994). Pleomorphic 19(2):97-100. PMID:12622140
1352. Korten AG, ter Berg HJ, Spincemaille GH, 102(3):200-2. PMID:10987382 pineocytoma with extensive neuronal differen- 1400. Kurzwelly D, Glas M, Roth P, Weimann E,
van der Laan RT, Van de Wei AM (1998). ln- 1369. Kros JM, Delwel EJ, de Jong TH, Tanghe tiation: report of two cases. Acta Neuropathol. Lohner H, Waha A, et al. (2010). Primary CNS
lracranial chondrosarcoma: review of the litera- HL, van Run PR, Vissers K, et al. (2002). Des- 88(5):448-53. PMID:7847074 lymphoma in lhe elderly: temozolomide therapy
ture and report of 15 cases. J Neurol Neurosurg moplaslic infantile astrocytoma and ganglioglio- 1385. Kudo H, Oi S, Tamaki N, Nishida Y, Mat- and MGMT status. J Neurooncol. 97(3):389-92.
Psychiatry. 65(1):88-92. PMID:9667567 ma: a search for genomic characteristics. Acta sumoto S (1990). Ependymoma diagnosed PMID:19841864
1353. Koschny R, Holland H, Koschny T, Neuropathol. 104(2):144-8. PMID:12111357 in the first year of life in Japan in collaborati- 1401. Kwon CH, Zhu X, Zhang J, Knoop LL,
Vitzthum HE (2006). Comparative geno- 1370. Kros JM, Gorlia T, Kouwenhoven MC, on with the International Society for Pediatric Tharp R, Smeyne RJ, et al. (2001 ). Pten
mic hybridization pattern of non-anaplastic Zheng PP, Collins VP, Figarella-Branger D, et Neurosurgery. Childs Nerv Syst. 6(7):375-8. regulates neuronal soma size: a mouse mo-
and anaplastic oligodendrogliomas-a me- al. (2007). Panel review of anaplastic oligo- PMID:1669244 del of Lhermitle-Duclos disease. Nat Genet.
ta-analysis. Pathol Res Pract. 202(1):23-30. dendroglioma from European Organization For 1386. Kuhlmann T, Lassmann H, Bruck W 29(4):404-11. PMID:11726927
PMID:16356658 Research and Treatment of Cancer Trial 26951: (2008). Diagnosis of inflammatory demye- 1402. Kuchler J, Hartmann W, Waha A, Koch
1354. Koutourousiou M, Georgakoulias N, Kon- assessment of consensus in diagnosis, intluen- lination in biopsy specimens: a practical A, Endl E, Wurst P, el al. (2011). p75(NTR) in-
togeorgos G, Seretis A (2009). Subependymo- ce of 1 p/19q loss, and correlations with outco- approach. Acta Neuropathol. 115(3):275-87. duces apoptosis in medulloblastoma cells. lnl J
mas of the lateral ventricle: tumor recurrence me. J Neuropathol Exp Neurol. 66(6):545-51. PMID:18175128 Cancer. 128(8):1804-12. PMID:20549701

References 377
1403. Kuker w. Nagele T, Korte! A. Heck! S, malignant peripheral nerve sheath tumors. Ann CTN NB 1 -mutated adamantinomatous cranio- 1451. Lee DY, Chung CK, Hwang YS, Choe
Thiel E, Bamberg M, et al. (2005). Primary cen- Surg Oncol. 20(1):66-72. PMID:22878618 pharyngioma. Acta Neuropathol. 127(6):927-9. G, Chi JG, Kim HJ, et al. (2000). Dysembryo-
tral nervous system lymphomas (PCNSL): MRI 1417. Lai A, Kharbanda S, Pope WB, Tran A, PMID:24715106 plastic neuroepithelial tumor: radiological fin-
features at presentation in 100 patients. J Neu- Solis OE, Peale F, et al. (2011). Evidence for 1433. Larson JJ, Tew JM Jr, Simon M, Menon dings (including PET, SPECT, and MRS) and
rooncol. 72(2):169-77. PMID:15925998 sequenced molecular evolution of IDH1 mutant AG (1995). Evidence for clonal spread in the surgical strategy. J Neurooncol. 47(2):167-74.
1403A. Kusters-Vandevelde HV, Creytens D, glioblastoma from a distinct cell of origin. J Clin development of multiple meningiomas. J Neu- PMID:10982159
Grunsven AC, Jeunink M, Winnepenninckx V, Oncol. 29(34):4482-90. PMID:22025148 rosurg. 83(4):705-9. PMID:7674021 1452. Lee EB, Tihan T, Scheithauer BW, Zhang
Groenen PJ, et al. (2016). SF3B1 and EIF1AX 1418. Laigle-Donadey F, Martin-Duverneuil 1434. Lashner BA, Riddell RH, Winans CS PJ, Gonatas NK (2009). Thyroid transcription
mutations occur in primary leptomeningeal me- N, Lejeune J, Criniere E, Capelle L, Duffau (1986). Ganglioneuromatosis of the colon factor 1 expression in sellar tumors: a histo-
lanocytic neoplasms; yet another similarity to H, et al. (2004). Correlations between mole- and extensive glycogenic acanthosis in Cow- genetic marker? J Neuropathol Exp Neurol.
uveal melanomas. Acta Neuropathol Commun. cular profile and radiologic pattern in oligo- den's disease. Dig Dis Sci. 31(2):213-6. 68(5):482...£. PMID:19525896
4(1):5. PMID:26769193 dendroglial tumors. Neurology. 63(12):2360-2. PMID:3943449 1453. Lee EQ (2015). Nervous system me-
1404. Kusters-Vandevelde HV, Klaasen A, Kiis- PMID:15623700 1435. Laskin WB, Weiss SW, Bratthauer GL tastases from systemic cancer. Continuum
ters B, Groenen PJ, van Engen-van Grunsven 1419. Laigle-Donadey F, Taillibert S, Mokhta- (1991). Epithelioid variant of malignant periphe- (Minneap Minn). 21 2 Neuro-oncology:415-28.
IA, van Dijk MR, et al. (2010). Activating muta- ri K, Hildebrand J, Delattre JY (2005). Dural ral nerve sheath tumor (malignant epithelioid PMID:25837904
tions of the GNAQ gene: a frequent event in pri- metastases. J Neurooncol. 75(1):57-61. schwannoma). Am J Surg Pathol. 15(12):1136- 1454. Lee HY, Yoon CS, Sevenet N, Rajalingam
mary melanocytic neoplasms of the central ner- PMID:16215816 45. PMID:1746681 V, Delattre 0, Walford NQ (2002). Rhabdoid
vous system. Acta Neuropathol. 119(3):317-23. 1420. Lakings DR, Gehrke CW, Waalkes TP 1436. Lassman AB. Iwamoto FM, Cloughesy TF, tumor of the kidney is a component of the rh-
PMID:19936769 (1976). Determination of trimethylsilyl methyla- Aldape KD, Rivera AL, Eichler AF, et al. (2011). abdoid predisposition syndrome. Pediatr Dev
1405. Kiisters-Vandevelde HV, Kiisters B, van ted nucleic acid bases in urine by gas-liquid International retrospective study of over 1000 Pathol. 5(4):395-9. PMID:12016529
Engen-van Grunsven AC, Groenen PJ, Wesse- chromatography . J Chromatogr 116: 69-81. adults wilh anaplastic oligodendroglial tumors. 1455. Lee JH, Sundaram V, Stein DJ, Kinney
ling P, Blokx WA (2015). Primary melanocytic PMID:1411 Neuro Oneal. 13(6):649-59. PMID:21636710 SE, Stacey DW. Golubic M (1997). Reduced
tumors of the central nervous system: a review 1421. Lal A, Dahiya S, Gonzales M, Hiniker 1437. Lau D, La Marca F, Camelo-Piragua S, expression of schwannominlmerlin in hu-
with focus on molecular aspects. Brain Pathol. A, Prayson R, Kleinschmidt-DeMasters BK, Park P (2013). Metastatic paraganglioma of man sporadic meningiomas. Neurosurgery.
25(2):209-26. PMID:25534128 et al. (2014). lgG4 overexpression is rare in the spine: case report and review of the lite- 40(3):578...£7. PMID:9055299
1406. Kiisters-Vandevelde HV, van Engen-van meningiomas with a prominent inflammatory rature. Clin Neurol Neurosurg. 115(9):1571-4. 1456. Lee JY, Dong SM, Park WS, Yoo NJ, Kim
Grunsven IA, Kiisters B, van Dijk MR, Groe- component: a review of 16 cases. Brain Pathol. PMID:23398849 CS, Jang JJ, et al. (1998). Loss of heterozygo-
nen PJ, Wesseling P, et al. (2010). Improved 24(4):352-9. PMID:24467316 1438. Lauer DH, Enzinger FM (1980). Cranial sity and somatic mutations of the VHL tumor
discrimination of melanotic schwannoma from 1422. Lam CW, Xie J, To KF, Ng HK, Lee KC, fasciitis of childhood. Cancer. 45(2):401-6. suppressor gene in sporadic cerebellar he-
melanocytic lesions by combined morphological Yuen NW, et al. (1999). A frequent activa- PMID:7351023 mangioblastomas. Cancer Res. 58(3):504-8.
and GNAQ mutational analysis. Acta Neuropa- ted smoothened mutation in sporadic basal 1439. Lavrnic S, Macvanski M, Ristic-Balos D, PMID:9458097
thol. 120(6):755-64. PMID:20865267 cell carcinomas. Oncogene. 18(3):833-6. Gavrilov M, Damjanovic D, Gavrilovic S, et al. 1457. Lee JY, Wakabayashi T, Yoshida J
1407. Labreche K, Simeonova I, Kamoun A, PMID:9989836 (2012). Papillary glioneuronal tumor: unexplo- (2005). Management and survival of pineob-
Gleize V, Chubb D, tetouze E, et al.; POLA 1423. Lamb RF, Roy C, Diefenbach T J. Vinters red entity. J Neurol Surg A Cent Eur Neurosurg. lastoma: an analysis of 34 adults from the
Network (2015). TCF12 is mutated in anapla- HV, Johnson MW, Jay DG, et al. (2000). The 73(4):224-9. PMID:21842459 brain tumor registry of Japan. Neural Med
stic oligodendroglioma. Nat Commun. 6:7207. TSC1 tumour suppressor hamartin regula- 1440. Laws ER Jr, Goldberg WJ, Bernstein Chir (Tokyo). 45(3):132-41, discussion 141-2.
PMID:26068201 tes cell adhesion through ERM proteins and JJ (1993). Migration of human malignant ast- PMID:15782004
1408. Labussiere M, Di Stefano AL. Gleize the GTPase Rho. Nat Cell Biol. 2(5):281-7. rocytoma cells in the mammalian brain: Sche- 1458. Lee S, Cimica V, Ramachandra N, Zag-
V, Boisselier B, Giry M, Mangesius S, et al. PMID:10806479 rer revisited. Int J Dev Neurosci. 11(5):691-7. zag D, Kalpana GV (2011). Aurora A is a repres-
(2014). TERT promoter mutations in gliomas. 1424. Lambert SR, Witt H. Hovestadt V, Zuck- PMID:8116480 sed effector target of the chromatin remodeling
genetic associations and clinico-pathological nick M, Kool M, Pearson DM, et al. (2013). 1441. Le DT, Uram JN, Wang H, Bartlett BR, protein INl1/hSNF5 required for rhabdoid tu-
correlations. Br J Cancer. 111(10):2024-32. Differential expression and methylation of brain Kemberling H, Eyring AD, et al. (2015). PD-1 mor cell survival. Cancer Res. 71(9):3225-35.
PMID:25314060 developmental genes define location-specific Blockade in Tumors with Mismatch-Repair PMID:21521802
1409. Lach B, Duggal N, DaSilva VF, Benoit subsets of pilocytic astrocytoma. Acta Neuropa- Deficiency. N Engl J Med. 372(26):2509-20. 1459. Lee SH, Appleby V, Jeyapalan JN, Palmer
BG (1996). Association of pleomorphic xan- thol. 126(2):291-301. PMID:23660940 PMID:26028255 RD, Nicholson JC, Sottile V, et al. (2011). Varia-
thoastrocytoma with cortical dysplasia and neu- 1425. Lambiv WL, Vassallo I, Delorenzi M, Shay 1442. Le LQ, Shipman T, Burns DK, Parada ble methylation of the imprinted gene, SNRPN,
ronal tumors. A report of three cases. Cancer. T, Diserens AC, Misra A, et al. (2011). The Wnt LF (2009). Cell of origin and microenviron- supports a relationship between intracranial
78(12):2551-63. PMID:8952564 inhibitory factor 1 (WIF1) is targeted in gliob- ment contribution for NF1-associated dermal germ cell tumours and neural stem cells. J Neu-
1410. Lach B, Duncan E, Rippstein P, Benoit lastoma and has a tumor suppressing function neurofibromas. Cell Stem Cell. 4(5):453-63. rooncol. 101 (3):41 S--28. PMID:20582452
BG (1994). Primary intracranial pleomorphic potentially by induction of senescence. Neuro PMID:19427294 1460. Lee W, Teckie S, Wiesner T, Ran L, Prieto
angioleiomyoma-a new morphologic vari- Oneal. 13(7):736-47. PMID:21642372 1443. Le Mercier M, Hastir D, Moles Lopez X, Granada CN, Lin M, et al. (2014). PRC2 is recur-
ant. An immunohistochemical and electron 1426. Lamont JM, McManamy CS, Pearson De Neve N, Maris C, TrepantAL, etal. (2012).A rently inactivated through EED or SUZ12 loss in
microscopic study. Cancer. 74(7):1915-20. AD, Clifford SC, Ellison DW (2004). Combined simplified approach for the molecular classifica- malignant peripheral nerve sheath tumors. Nat
PMID:8082097 histopathological and molecular cytogenetic tion of glioblastomas. PLoS One. 7(9):e45475. Genet. 46(11):1227-32. PMID:25240281
1411. Lachenal F, Cotton F, Desmurs-Clavel stratification of medulloblastoma patients. Clin PMID:23029035 1461. Leeds NE, Lang FF, Ribalta T, Sawaya
H, Haroche J, Taillia H, Magy N. et al. (2006). Cancer Res. 10(16):5482-93. PMID:15328187 1445. LeBoit PE, Burg G, Weedon D, Sarasin R, Fuller GN (2006). Origin of chordoid glioma
Neurological manifestations and neuroradiolo- 1427. Lamzabi I, Arvanitis LD, Reddy VB, A, editors. (2005). World Health Organization of the third ventricle. Arch Pathol Lab Med.
gical presentation of Erdheim-Chester disea- Bitterman P, Gattuso P (2013). lmmunophe- Classification of Tumours. Pathology and Ge- 130(4):460-4. PMID:16594739
se: report of 6 cases and systematic review notype of myxopapillary ependymomas. Appl netics of Skin Tumours. 3rd ed. Lyon: IARC 1462. Leenstra JL, Rodriguez FJ, Frechette
of the literature. J Neurol. 253(10):1267-77. lmmunohistochem Mol Morphol. 21 (6):485-9. Press.1157. CM, Giannini C, Stafford SL, Pollock BE, et al.
PMID:17063320 PMID:23455181 1446. Lebrun C, Fontaine D, Ramaioli A, Van- (2007). Central neurocytoma: management re-
1412. Lack EE (1994). Paragangliomas. In: 1428. Lang FF, Epstein FJ, Ransohoff J, Allen denbos F, Chanalet S, Lonjon M, et al.; Nice commendations based on a 35-year experien-
Sternberg SS, editor. Diagnostic Surgical Pa- JC, Wisoff J, Abbott IR, et al. (1993). Central Brain Tumor Study Group (2004). Long-term ce. Int J Radial Oneal Biol Phys. 67(4):1145-54.
thology. New York: Raven Press; pp. 55S--621. nervous system gangliogliomas. Part 2: Cli- outcome of oligodendrogliomas. Neurology. PMID:17187939
1413. Lafay-Cousin L, Hader W, Wei XC, nical outcome. J Neurosurg. 79(1>):867-73. 62(10):1783-7. PMID:15159478 1463. Lehman NL (2008). Patterns of brain in-
Nordal R, Strother D, Hawkins C, et al. (2014). PMID 8246055 1447. Lechapt-Zalcman E, Chapon F, Guillamo filtration and secondary structure formation in
Post-chemotherapy maturation in supratentorial 1429. Langford LA (1986). The ultrastructure JS, Khouri S, Menegalli-Boggelli D, Loussouarn supratentorial ependymal tumors. J Neuropa-
primitive neuroectodermal tumors. Brain Pathol. of the ependymoblastoma. Acta Neuropathol. D, et al. (2011). Long-term clinicopathological thol Exp Neurol. 67(9):900-10. PMID:18716554
24(2):166-72. PMID:24033491 71(1-2):136-41. PMID:3776467 observations on a papillary tumour of the pineal 1464. Lehman NL, Horoupian DS, Warnke
1414. Lafay-Cousin L, Hawkins C, Carrel AS, 1430. Langford LA, Camel MH (1987). Pali- region. Neuropathol Appl Neurobiol. 37(4):431- RA, Sundram UN, Peterson K, Harsh GR 4th
Johnston D, Zelcer S, Wilson B, et al. (2012). sading pattern in cerebral neuroblastoma mi- 5. PMID:20942871 (2002). Dural marginal zone lymphoma with
Central nervous system atypical teratoid rhab- micking the primitive polar spongioblastoma. 1448. Lee D, Cho YH, Kang SY, Yoon N, Sung massive amyloid deposition: rare low-grade
doid tumours: the Canadian Paediatric Brain An ultrastructural study. Acta Neuropathol. CO, Suh YL (2015). BRAF V600E mutations primary central nervous system B-cell lympho-
Tumour Consortium experience. Eur J Cancer. 73(2):153-9. PMID:3604582 are frequent in dysembryoplastic neuroepit- ma. Case report. J Neurosurg. 96(2):368-72,
48(3):353-9. PMID:22023887 1431. Lannering B, Rutkowski S, Doz F, Pizer helial tumors and subependymal giant cell PMID:11838814
1415. Lafay-Cousin L, Keene D. Carrel AS, B, Gustafsson G, Navajas A, et al. (2012). Hy- astrocytomas. J Surg Oneal. 111(3):359-64. 1465. Lehman NL, Jorden MA, Huhn SL,
Fryer C, Brossard J, Crooks B, et al. (2011). perfractionated versus conventional radiothera- PMID:25346165 Barnes PD, Nelson GB, Fisher PG, et al.
Choroid plexus tumors in children less than 36 py followed by chemotherapy in standard-risk 1449. Lee D, Suh YL (2010). Histologically con- (2003). Cortical ependymoma. A case report
months: the Canadian Pediatric Brain Tumor medulloblastoma: results from the randomized firmed intracranial germ cell tumors; an analysis and review. Pediatr Neurosurg. 39(1 ):50-4.
Consortium (CPBTC) experience. Childs Nerv multicenter HIT-SIOP PNET 4 trial. J Clin Oncol. of 62 patients in a single institute. Virchows PMID:12784079
Syst. 27(2):25S--64. PMID:20809071 30(26):3187-93. PMID22851561 Arch. 457(3):347-57. PMID:20652714 1466. Lekanne Deprez RH, Bianchi AB, Groen
1416. Lafemina J, Qin LX, Moraco NH, Anto- 1432. Larkin SJ, Preda V, Karavitaki N, Gross- 1450. Lee DK, Jung HW, Kim DG, Paek SH, NA, Seizinger BR, Hagemeijer A, van Drunen
nescu CR, Fields RC, Crago AM, et al. (2013). man A, Ansorge O (2014). BRAF V600E Gwak HS, Choe G (2001). Postoperative spinal E, et al. (1994). Frequent NF2 gene tran-
Oncologic outcomes of sporadic, neurofibro- mutations are characteristic for papillary seeding of craniopharyngioma. Case report. J script mutations in sporadic meningiomas and
matosis-associated, and radiation-induced craniopharyngioma and may coexist with Neurosurg. 94(4):617-20. PMID:11302661 vestibular schwannomas. Am J Hum Genet.

378 References
54(6):1022-9. PMID:7911002 156(4):627-39. PMID:24553727 Kleinschmidt-DeMasters BK (2008). Radiati- Hou GQ, et al. (2013). MR imaging of cereb-
1467. Lellouch-Tubiana A, Boddaert N, Bour- 1485. Li DM, Sun H (1997). TEP1, encoded by on-induced meningiomas: clinical, cytogenetic, ral extraventricular neurocytoma: a report of 9
geois M, Fohlen M, Jouvet A, Delalande 0, et a candidate tumor suppressor locus, is a no- and microarray features. Acta Neuropathol. cases. AJNR Am J Neuroradiol. 34(3):541-6.
al. (2005). Angiocentric neuroepithelial tumor vel protein tyrosine phosphatase regulated by 116(3):289-301. PMID:18604545 PMID:23042917
(ANET): a new epilepsy-related clinicopatholo- transforming growth factor beta. Cancer Res. 1502. Lim SC, Jang SJ (2006). Myxopapillary 1519. Liu XY, Gerges N, Korshunov A, Sabha
gical entity with distinctive MRI. Brain Pathol. 57(11):2124-9. PMID:9187108 ependymoma of the fourth ventricle. Clin Neural N, Khuong-Quang DA, Fontebasso AM, et al.
15(4):281-6. PMID:16389940 1486. Li FP, Fraumeni JF Jr, Mulvihill JJ, Blatt- Neurosurg. 108(2):211-4. PMID:16412846 (2012). Frequent ATRX mutations and loss of
1468. Lellouch-Tubiana A, Bourgeois M, Veke- ner WA, Dreyfus MG, Tucker MA, et al. (1988).A 1503. Lim T, Kim SJ, Kim K, Lee JI, Lim H, Lee expression in adult diffuse astrocytic tumors
mans M, Robain O (1995). Dysembryoplastic cancer family syndrome in twenty-four kindreds. DJ, et al. (2011). Primary CNS lymphoma other carrying IDH111DH2 and TP53 mutations. Acta
neuroepithelial tumors in two children with Cancer Res. 48(18):5358-62. PMID:3409256 than DLBCL: a descriptive analysis of clinical Neuropathol. 124(5):615-25. PMID:22886134
neurofibromatosis type 1. Acta Neuropathol. 1487. Li G, Mitra S, Karamchandani J, Edwards features and treatment outcomes. Ann He- 1520. Liverman C, Mafra M, Chuang SS, Shi-
90(3):319-22 PMID:8525807 MS, Wong AJ (2010). Pineal parenchymal matol. 90(12):1391-8. PMID:21479535 vane A, Chakrabarty A, Highley R, et al. (2015).
1469. Lemke D, Pfenning PN, Sahm F, Klein tumor of intermediate differentiation: clinico- 1504. Lin AL, Liu J, Evans J, Leuthardt EC, A clinicopalhologic study of 11 rosette-forming
AC, Kempf T, Warnken U, et al. (2012). Costi- pathological report and analysis of epidermal Rich KM, Dacey RG, et al. (2014). Codeletions meningiomas: a rare and potentially confu-
mulatory protein 41gB7H3 drives the malignant growth factor receptor variant Ill expression. at 1 p and 19q predict a lower risk of pseudo- sing pattern. Acta Neuropathol. 130(2):311-3.
phenotype of glioblastoma by mediating immu- Neurosurgery. 66(5):963-8, discussion 968. progression in oligodendrogliomas and mixed PMID:26106026
ne escape and invasiveness. Clin Cancer Res. PMID:20404701 oligoastrocytomas. Neuro Oneal. 16(1 ):123-30. 1521. Lloyd KM 2nd, Dennis M (1963). Cow-
18(1):105-17. PMID:22080438 1488. Li J, Simpson L, Takahashi M, Miliare- PMID:24285548 den's disease. A possible new symptom com-
1470. Leonard JR, Perry A, Rubin JB, King AA, sis C, Myers MP, Tonks N, et al. (1998). The 1505. Lin NU, Amiri-Kordestani L, Palmieri plex with multiple system involvement. Ann
Chicoine MR, Gutmann DH (2006). The role of PTENIMMAC1 tumor suppressor induces cell 0, Liewehr DJ, Steeg PS (2013). CNS me- Intern Med. 58:136-42. PMID:13931122
surgical biopsy in the diagnosis of glioma in in- death that is rescued by the AKTlprotein kina- tastases in breast cancer: old challenge, new 1522. Ladding P, Kindblom LG, Angervall L,
dividuals with neurofibromatosis-1. Neurology. se B oncogene. Cancer Res. 58(24):5667-72. frontiers. Clin Cancer Res. 19(23):6404-18. Stenman G (1990). Cellular schwannoma. A
67(8):1509-12. PMID: 17060590 PMID:9865719 PMID:24298071 clinicopathologic study of 29 cases. Virchows
1471. Leone PE, Bello MJ, Mendiola M, Kusak 1489. Li J, Yen C, Liaw D, Podsypanina K, Bose 1506. Lin SL, Wang JS, Huang CS, Tseng Arch A Pathol Anal Histopathol. 416(3):237-48.
ME, De Campos JM, Vaquero J, et al. (1998). S, Wang SI, et al. (1997). PTEN, a putative HH (1996). Primary intracerebral leiomyo- PMID:2105560
Allelic status of 1p, 14q, and 22q and NF2 gene protein tyrosine phosphatase gene mutated ma: a case with eosinophilic inclusions of 1523. Loh JK, Lieu AS, Chai CY, Howng SL
mutations in sporadic schwannomas. Int J Mol in human brain, breast, and prostate cancer. actin filaments. Histopathology. 28(4):365-9. (2011). Malignant transformation of a desmo-
Med. 1(5):889-92. PMID:9852312 Science. 275(5308):1943-7. PMID:9072974 PMID:873234 7 plastic infanlile ganglioglioma. Pediatr Neural.
1472. Leoz ML, Carballal S, Moreira L, Ocana T, 1490. Li JY, Langford LA, Adesina A, Bodhired- 1507. Lin Y J, Yang QX, nan XY, Li B, Li Z 45(2):135-7. PMID:21763958
Balaguer F (2015). The genetic basis of familial dy SR, Wang M, Fuller GN (2012). The high (2013). Unusual primary intracranial dural-ba- 1524. Longatti P, Basaldella L, Orvieto E, Dei
adenomatous polyposis and its implications for mitotic count detected by phospho-histone H3 sed poorly differentiated synovial sarcoma with Tos AP, Martinuzzi A (2006). Aquaporin 1 ex-
clinical practice and risk management. Appl Clin immunostain does not alter the benign behavi- t(X: 18)(p11: q11). Neuropathology. 33(1):75- pression in cystic hemangioblastomas. Neuros-
Genet. 8:95-107. PMID:25931827 or of angiocentric glioma. Brain Tumor Pathol. 82. PMID:22537253 ci Lett. 392(3):178-80. PMID:16300893
1473. Lermen 0, Frank S, Hassler W 29(1):68-72. PMID:21892765 1508. Lindberg N, Jiang Y, Xie Y, Bolouri H, 1525. Longy M, Lacombe D (1996). Cowden
(2010). Postoperative spinal recurren- 1491. Li M, Lee KF, Lu Y, Clarke I, Shih D, Eber- Kastemar M, Olofsson T, et al. (2014). On- disease. Report of a family and review. Ann
ce of craniopharyngioma. Acta Neurochir hart C, et al. (2009). Frequent amplification of cogenic signaling is dominant to cell of origin Genet. 39(1):35-42. PMID:9297442
(Wien). 152(2):309-11, discussion 311. a chr19q13.41 microRNA polycistron in aggres- and dictates astrocytic or oligodendroglial 1526. Lopes MB, Altermatt HJ, Scheithauer
PMID:19838829 sive primitive neuroectodermal brain tumors. tumor development from oligodendrocyte pre- BW, Shepherd CW, VandenBerg SR (1996).
1474. Lester RA, Brown LC, Eckel LJ, Foote RT, Cancer Cell. 16(6):533-46. PMID:19962671 cursor cells. J Neurosci. 34(44):14644-51. lmmunohistochemical characterization of sub-
NageswaraRao AA, Buckner JC, et al. (2014). 1492. Li P, James SL, Evans N, Davies AM, PMID:25355217 ependymal giant cell astrocytomas. Acta Neu-
Clinical outcomes of children and adults with Herron B, Sumathi VP (2007). Paraganglio- 1509. Lindstrom KM, Cousar JB, Lopes MB ropathol. 91(4):368-75. PMID:8928613
central nervous system primitive neuroecto- ma of the cauda equina with subarachnoid (2010). lgG4-related meningeal disease: clini- 1526A. Los M, Jansen GH, Kaelin WG, Lips
dermal tumor. J Neurooncol. 120(2):371-9. haemorrhage. Clin Radial. 62(3):277-80. co-pathological features and proposal for dia- CJ, Blijham GH, Voest EE (1996). Expressi-
PMID:25115737 PMID:17293223 gnostic criteria. Acta Neuropathol. 120(6):765- on pattern of the von Hippel-Lindau protein
1475. tetouze E, Martinelli C, Lorio! C, Burni- 1493. Li W, Cooper J, Zhou L, Yang C, Erd- 76. PMID:20844883 in human tissues. Lab Invest. 75(2):231-8.
chon N, Abermil N, Ottolenghi C, et al. (2013). jument-Bromage H, Zagzag D, et al. (2014). 1510. Lindstrom E, Shimokawa T, Toftgard PMID:8765323
SDH mutations establish a hypermethylator Merlin/NF2 loss-driven tumorigenesis linked to R, Zaphiropoulos PG (2006). PTCH muta- 1527. Losa M, Saeger W, Martini P, Pandolfi
phenotype in paraganglioma. Cancer Cell. CRL4(DCAF1 )-mediated inhibition of the hippo tions: distribution and analyses. Hum Muta!. C, Terreni MR, Taccagni G, et al. (2000). Acro-
23(6):739-52. PMID:23707781 pathway kinases Lats1 and 2 in the nucleus. 27(3):215-9. PMID:16419085 megaly associated with a granular cell tumor of
1476. Levin N, Lavon I, Zelikovitsh B, Fuchs Cancer Cell. 26(1):48-60. PMID:25026211 1511. Linnebank M, Moskau S, Kowall A, the neurohypophysis: a clinical and histological
D, Bokstein F, Fellig Y, et al. (2006). Progres- 1494. Li W, Graeber MB (2012). The molecular Semmler A, Bangard C, Vogt-Schaden M, et study. Case report. J Neurosurg. 93(1):121-6.
sive low-grade oligodendrogliomas: response profile of microglia under the influence of glioma. al. (2012). Association of transcobalamin c. PMID:10883914
to temozolomide and correlation between Neuro Oneal. 14(8):958-78. PMID:22573310 776C>G with overall survival in patients with 1528. Lassos C, Bayraktar S, Weinzierl E, You-
genetic profile and 06-methylguanine DNA 1495. Li YS, Ramsay DA, Fan YS, Armstrong primary central nervous system lymphoma. Br J nes SF, Hosein PJ, Tibshirani RJ, et al. (2014).
methyltransferase protein expression. Cancer. RF, Del Maestro RF (1995). Cytogenetic evi- Cancer. 107(11):1840-3. PMID:23099805 LM02 and BCL6 are associated with improved
106(8):1759-65. PMID:16541434 dence that a tumor suppressor gene in the long 1512. Linnebank M, Schmidt S, K61sch H, survival in primary central nervous system
1477. Levine AJ (1997). p53, the cellular gate- arm of chromosome 1 contributes to glioma Linnebank A, Heun R, Schmidt-Wolf IG, et al. lymphoma. Br J Haematol. 165(5):640-8.
keeper for growth and division. Cell. 88(3):323- growth. Cancer Genet Cytogenet. 84(1):46-50. (2004 ). The methionine synthase polymorphism PMID:24571259
31. PMID:9039259 PMID:7497442 D919G alters susceptibility to primary cent- 1529. Lotan I, Khlebtovsky A, lnbar E, Strenov
1478. Levy RA (1993). Paraganglioma of the 1496. Li ZJ, Lan XL, Hao FY, Yao WC, Wang ral nervous system lymphoma. Br J Cancer. J, Djaldetti R, Steiner I (2012). Primary brain
filum terminale: MR findings. AJR Am J Roent- MY, Chen XO, et al. (2014). Primary cerebel- 90(10):1969-71. PMID:15138479 T-cell lymphoma in an HTLV-1 serologically
genol. 160(4):851-2. PMID:8456679 lar paraganglioma: a pediatric case report 1513. Lipper S, Decker RE (1984). Parag- positive male. J Neural Sci. 314(1-2):163-5.
1479. Lewis JJ, Brennan MF (1996). Soft tis- and review of the literature. Pediatr Neural. anglioma of the cauda equina. A histologic, PMID:22118868
sue sarcomas. Curr Probl Surg. 33(10):817-72. 50(4):303-6. PMID:24485927 immunohistochemical, and ultrastructural stu- 1530. Louis DN (1994). The p53 gene and pro-
PMID 8885853 1497. Liang L, Korogi Y, Sugahara T, lkilshirna dy and review of the literature. Surg Neural. tein in human brain tumors. J Neuropathol Exp
1480. Lewis PW, Muller MM, Koletsky MS, I, Shigematsu Y, Okuda T, et al. (2002). MRI of 22(4):415-20. PMID:6474349 Neural. 53(1):11-21. PMID:8301315
Cordero F, Lin S, Banaszynski LA, et al. (2013). intracranial germ-cell lumours. Neuroradiology. 1514. Liss L, Kahn EA (1958). Pituicytoma, 1531. Louis DN, Hamilton AJ, Sobel RA, Oje-
Inhibition of PRC2 activity by a gain-of-function 44(5):382-8. PMID:12012121 tumor of the sella turcica; a clinicopatho- mann RG (1991). Pseudopsammomatous
H3 mutation found in pediatric glioblastoma. 1498. Liang X, Shen D, Huang Y, Yin C, Boja- logical study. J Neurosurg. 15(5):481-8. meningioma with elevated serum carcinoem-
Science. 340(6134):857-61. PMID:23539183 nowski CM, Zhuang Z, et al. (2007). Molecular PMID:13576191 bryonic antigen: a true secretory meningio-
1481. Lewis RA, Gerson LP, Axelson KA, pathology and CXCR4 expression in surgically 1515. Listernick R, Charrow J, Greenwald M, ma. Case report. J Neurosurg. 74(1):129-32.
Riccardi VM, Whitford RP (1984). von Reck- excised retinal hemangioblastomas associated Mets M (1994). Natural history of optic pathway PMID:1984492
linghausen neurofibromatosis. II. Incidence of with von Hippel-Lindau disease. Ophthalmolo- tumors in children with neurofibromatosis type 1532. Louis ON, Hedley-Whyte ET, Martuza RL
optic gliomata. Ophthalmology. 91 (8):929-35. gy. 114(1):147-56. PMID:17070589 1: a longitudinal study. J Pediatr. 125(1):63-6. (1990). Sarcomatous proliferation of the vascu-
PMID:6436764 1499. Liaw D, Marsh DJ, Li J, Dahia PL, Wang PMID:8021787 lature in a subependymoma: a follow-up study
1482. Lhermitte J, Duclos P (1920). Sur un gan- SI, Zheng Z, et al. (1997). Germline mutations 1516. Listernick R, Ferner RE, Liu GT, Gut- of sarcomatous dedifferentiation. Acta Neuro-
glioneurome diffus du coertex du cervelet. Bull of the PTEN gene in Cowden disease, an inhe- mann DH (2007). Optic pathway gliomas in pathol. 80(5):573-4. PMID:2251917
Assoc Fr Etud Cancer. 9:99-107. rited breast and thyroid cancer syndrome. Nat neurofibromatosis-1: controversies and re- 1533. Louis DN, Ohgaki H, Wiestler OD, Cave-
1483. Li D, Schauble B, Moll C, Fisch U (1996). Genet. 16(1):64-7. PMID:9140396 commendations. Ann Neural. 61(3):189-98. nee WK, editors. (2007). WHO Classification of
lntratemporal facial nerve perineurioma. Laryn- 1500. Ligon KL, Alberta JA, Kho AT, Weiss J, PMID 17387725 Tumours of the Central Nervous System. 4th
goscope. 106(3 Pt 1):328-33. PMID:8614198 Kwaan MR, Nutt CL, et al. (2004). The oligo- 1517. Listernick R, Mancini AJ, Charrow J ed. Lyon: International Agency for Research
1484. Li D, Wang JM, Li GL, Hao SY, Yang dendroglial lineage marker OLIG2 is universally (2003). Segmental neurofibromatosis in child- on Cancer.
Y, Wu Z, et al. (2014). Clinical, radiological, expressed in diffuse gliomas. J Neuropathol hood. Am J Med Genet A. 121A(2):132-5. 1534. Louis ON, Ohgaki H, WiesUer OD, Ca-
and pathological features of 16 papillary gli- Exp Neural. 63(5):499-509. PMID:15198128 PMID:12910491 venee WK, Burger PC, Jouvet A, et al. (2007).
oneuronal tumors. Acta Neurochir (Wien). 1501. Lillehei KO, Donson AM, 1518. Liu K, Wen G, Lv XF, Deng YJ, Deng YJ, The 2007 WHO classification of tumours of

References 379
the central nervous system. Acta Neuropathol. H, Elger CE, Wiesller OD, et al. (2003). The and renal cell carcinoma. J Med Genet. complex. Acta Neuropathol. 123(5):685-93.
114(2):97-109. PMID:17618441 spectrum of long-term epilepsy-associated 27(5):311-4. PMID:2352258 PMID:22327361
1535. Louis DN, Perry A, Burger P, Ellison DW, tumors: long-term seizure and tumor outco- 1568. Mahzoni P, Zavareh MH, Bagheri M, 1583. Marden FA, Wippold FJ 2nd, Perry A
Reifenberger G, von Deimling A, et al.; Inter- me and neurosurgical aspects. Epilepsia. Hani N, Moqtader B (2012). lntracranial RO- (2003). Fast magnetic resonance imaging
national Society Of Neuropathology-Haarlem 44(6):822-30 PMID:12790896 SAi-DORFMAN Disease. J Res Med Sci. in steady-state precession (true FISP) in the
(2014). International Society Of Neuropatholo- 1552. Luyken C, BIOmcke I, Fimmers R, Urbach 17(3) 304-7. PMID:23267385 prenatal diagnosis of a congenital brain tera-
gy-Haarlem consensus guidelines for nervous H, Wiesller OD, Schramm J (2004). Supratento- 1569. Maier H, Ofner D, Hittmair A, Kitz K, Bu- toma. J Comput Assist Tomogr. 27(3):427-30.
system tumor classification and grading. Brain rial gangliogliomas: histopathologic grading and dka H (1992). Classic, atypical, and anaplastic PMID:12794611
Pathol. 24(5):429-35. PMID:24990071 tumor recurrence in 184 patients with a median meningioma: three histopathological subtypes 1584. Margetts JC, Kalyan-Raman UP
1536. Louis DN, Ramesh V, Gusella JF (1995). follow-up of 8 years. Cancer. 101(1):146-55. of clinical relevance. J Neurosurg. 77(4):616- (1989). Giant-celled glioblastoma of brain. A
Neuropathology and molecular genetics of PMID:15222000 23. PMID:1527622 clinico-pathological and radiological study of
neurofibromatosis 2 and related tumors. Brain 1553. LObbe J, von Ammon K, Watanabe K, 1570. Majores M, von Lehe M, Fassunke J, ten cases (including immunohistochemistry
Pathol. 5(2):163-72. PMID:7670657 Hegi ME, Kleihues P (1995). Familial brain tu- Schramm J, Becker AJ, Simon M (2008). Tu- and ultrastructure). Cancer. 63(3):524-31.
1537. Louis DN, Richardson EP Jr, Dickersin mour syndrome associated with a p53 germline mor recurrence and malignant progression PMID:2912529
GR, Petrucci DA, Rosenberg AE, Ojemann deletion of codon 236. Brain Pathol. 5(1 ):15-23. of gangliogliomas. Cancer. 113(12):3355-63. 1585. Marigo V, Davey RA, Zuo Y, Cunningham
RG (1989). Primary inlracranial leiomyosarco- PMID:7767 487 PMID:18988291 JM, Tabin CJ (1996). Biochemical evidence
ma. Case report. J Neurosurg. 71(2):279-82. 1554. Lynch ED, Ostenmeyer EA, Lee MK, 1571. Maj6s C, Aguilera C, Cos M, Camins A, that patched is the Hedgehog receptor. Nature.
PMID:27 46352 Arena JF, Ji H, Dann J, et al. (1997). Inherited Candiota AP, Delgado-Gofii T, et al. (2009). In 384(6605):176-9. PMID:8906794
1538. Louis DN, von Deimling A, Dickersin GR, mutations in PTEN that are associated with vivo proton magnetic resonance spectroscopy 1586. Markesbery WR, Duffy PE, Cowen D
Dooling EC, Seizinger BR (1992). Desmoplastic breast cancer, cowden disease, and juvenile of intraventricular tumours of the brain. Eur Ra- (1973). Granular cell tumors of the central
cerebral astrocytomas of infancy: a histopatho- polyposis. Am J Hum Genet. 61(6):1254-60. diol. 19(8):2049-59. PMID:19277673 nervous system. J Neuropathol Exp Neurol.
logic, immunohistochemical, ultrastructural, PMID:9399897 1572. Makino K, Nakamura H, Yano S, Kuratsu 32(1)92-109. PMID:4346659
and molecular genetic study. Hum Pathol. 1555. MacCollin M, Chiocca EA, Evans J; Kumamoto Brain Tumor Group (2010). Po- 1587. Markesbery WR, Haugh RM, Young AB
23(12):1402-9. PMID:1468778 DG, Friedman JM, Horvitz R, Jaramillo D, pulation-based epidemiological study of primary (1981). Ultrastructure of pineal parenchymal
1539. Lovly CM, Gupta A, Lipson D, Otto G, el al. (2005). Diagnostic criteria for schwan- intracranial tumors in childhood. Childs Nerv neoplasms. Acta Neuropathol. 55(2):143-9.
Brennan T, Chung CT, et al. (2014). Inflam- nomatosis. Neurology. 64(11 ):1838-45. Syst. 26(8):1029-34. PMID:20349186 PMID:7315200
matory myofibroblastic tumors harbor multiple PMID:15955931 1573. Makino K, Nakamura H, Yano S, Kuratsu 1588. Marsh A, Wicking C, Wainwright B,
potentially actionable kinase fusions. Cancer 1556. MacCollin M, Ramesh V, Jacoby LB, J; Kumamoto Brain Tumor Research Group Chenevix-Trench G (2005). DHPLC analysis
Discov. 4(8):889-95. PMID:24875859 Louis DN, Rubio MP, Pulaski K, et al. (1994). (2013). Incidence of primary central nervous of patients with Nevoid Basal Cell Carcinoma
1540. Lu C, Ward PS, Kapoor GS, Rohle D, Mutational analysis of patients with neurofibro- system germ cell tumors in childhood: a regio- Syndrome reveals novel PTCH missense muta-
Turcan S, Abdel-Wahab 0, et al. (2012). IDH matosis 2. Am J Hum Genet. 55(2):314-20. nal survey in Kumamoto prefecture in southern tions in the sterol-sensing domain. Hum Mutat.
mutation impairs histone demethylation and PMID:7913580 Japan. Pediatr Neurosurg. 49(3):155-8. 26(3):283. PMID:16088933
results in a block to cell differentiation. Nature. 1557. MacCollin M, Willett C, Heinrich B, Ja- PMID:24751890 1589. Marsh DJ, Coulon V, Lunetta KL, Roc-
483(7390):474-8. PMID:22343901 coby LB, Acierno JS Jr, Perry A, et al. (2003). 1574. Makkar HS, Frieden IJ (2002). Conge- ca-Serra P, Dahia PL, Zheng Z, et al. (1998).
1541. Lu JQ, Patel S, Wilson BA, Pugh J, Meh- Familial schwannomatosis: exclusion of the nital melanocytic nevi: an update for the pe- Mutation spectrum and genotype-phenotype
ta V (2013). Malignant glioma with angiocentric NF2 locus as the genmline event. Neurology. diatrician. Curr Opin Pediatr. 14(4):397-403. analyses in Cowden disease and Bannay-
features. J Neurosurg Pediatr. 11 (3):350-5. 60(12):1968-74. PMID:12821741 PMID:12130901 an-Zonana syndrome, two hamartoma syndro-
PMID:23240849 1558. MacCollin M, Woodfin W, Kronn D, Short 1575. Malkin D, Li FP, Strong LC, Fraumeni JF mes with germline PTEN mutation. Hum Mol
1542. Lu L, Dai Z, Zhong Y, Lv G (2013). MP (1996). Schwannomatosis: a clinical and Jr, Nelson CE, Kim DH, et al. (1990). Germ line Genet. 7(3):507-15. PMID:9467011
Cervical intradural paraganglioma presen- pathologic study. Neurology. 46(4):1072-9. p53 mutations in a familial syndrome of breast 1590. Marsh DJ, Dahia PL, Caron S, Kum
ting as progressive cervicodynia: case report PMID:8780094 cancer, sarcomas, and other neoplasms. Scien- JB, Frayling IM, Tomlinson IP, et al. (1998).
and literature review. Clin Neurol Neurosurg. 1559. Machein MR, Plate KH (2004). Role of ce. 250(4985):1233-8. PMID:1978757 Germline PTEN mutations in Cowden syndro-
115(3):359-61. PMID:2272177 4 VEGF in developmental angiogenesis and in 1576. Mallory SB (1995). Cowden syndrome me-like families. J Med Genet. 35(11):881-5.
1543. Lu-Emerson C, Duda DG, Emblem KE, tumor angiogenesis in the brain. Cancer Treat (multiple hamartoma syndrome). Dermatol Clin. PMID9832031
Taylor JW, Gerstner ER, Loeffler JS, et al. Res. 117:191-218. PMID:15015562 13(1):27-31. PMID:7712647 1591. Marsh DJ, Dahia PL, Zheng Z, Liaw D,
(2015). Lessons from anti-vascular endotheli- 1560. Mack SC, Witt H, Piro RM, Gu L, Zuy- 1577. Malrnstrorn A, Grnnberg BH, Marosi C, Parsons R, Gorlin RJ, et al. (1997). Germlioe
al growth factor and anti-vascular endothelial derduyn S, Stutz AM, el al. (2014). Epigenomic Stupp R, Frappaz D, Schultz H, et al.; Nordic mutations in PTEN are present in Bannay-
growth factor receptor trials in patients with alterations define lethal CIMP-positive ependy- Clinical Brain Tumour Study Group (NCBTSG) an-Zonana syndrome. Nat Genet. 16(4):333-4.
glioblastoma. J Clin Oncol. 33(10):1197-213. momas of infancy. Nature. 506(7489):445-50. (2012). Temozolomide versus standard 6-week PMID:9241266
PMID:25713439 PMID:24553142 radiotherapy versus hypofractionated radiothe- 1592. Marsh DJ, Kum JB, Lunetta KL, Bennett
1544. Lubs ML, Bauer MS, Formas ME, Djo- 1561. Mackenzie IR (1999). Central neurocyto- rapy in patients older than 60 years with gliob- MJ, Gorlin RJ, Ahmed SF, et al. (1999). PTEN
kic B (1991). Lisch nodules in neurofibroma- ma: histologic atypia, proliferation potential, lastoma: the Nordic randomised, phase 3 trial. mutation spectrum and genotype-phenotype
tosis type 1. N Engl J Med. 324(18):1264-6. and clinical outcome. Cancer. 85(7):1606-10. Lancet Oneal. 13(9):916-26. PMID:22877848 correlations in Bannayan-Riley-Ruvalcaba
PMID:1901624 PMID:10193953 1578. Manjila S, Ray A, Hu Y, Cai DX, Cohen syndrome suggest a single entity with Cowden
1545. Ludwin SK, Rubinstein LJ, Russell DS 1562. Mader I, Stock KW, Radue EW, Stein- ML, Cohen AR (2011). Embryonal tumors with syndrome. Hum Mol Genet 8(8):1461-72.
(1975). Papillary meningioma: a malignant va- brich W (1996). Langerhans cell histiocytosis in abundant neuropil and true rosettes: 2 illustrati- PMID 10400993
riant of meningioma. Cancer. 36(4):1363-73. monocygote twins: case reports. Neuroradiolo- ve cases and a review of the literature. Neuro- 1593. Marsh DJ, Roth S, Lunetta KL, Hemminki
PMID:1175134 gy. 38(2): 163-5. PMID:8692432 surg Focus. 30(1):E2. PMID:21194275 A, Dahia PL, Sistonen P, et al. (1997). Exclusi-
1546. Lun M, Lok E, Gautam S, Wu E, Wong 1563. Maehama T, Dixon JE (1998). The tumor 1579. Mantripragada KK, Spurlock G, Kluwe on of PTEN and 10q22-24 as the susceptibility
ET (2011 ). The natural history of extracranial suppressor, PTEN/MMAC1, dephosphoryla- L, Chuzhanova N, Ferner RE, Frayling IM, et locus for juvenile polyposis syndrome. Cancer
metastasis from glioblastoma multiforme. J les the lipid second messenger, phospha- al. (2008). High-resolution DNA copy number Res. 57(22):5017-21. PMID:9371495
Neurooncol. 105(2):261-73. PMID:21512826 tidylinositol 3,4,5-trisphosphate. J Biol Chem. profiling of malignant peripheral nerve sheath 1594. Marsh DJ, Trahair TN, Martin JL, Chee
1547. Luse SA, Kernohan JW (1955). Granu- 273(22):13375-8. PMID:9593664 tumors using targeted microarray-based com- WY, Walker J, Kirk EP, et al. (2008). Rapamy-
lar-cell tumors of the stalk and posterior lobe 1564. Maertens 0, Brems H, Vandesompele parative genomic hybridization. Clin Cancer cin treatment for a child with germline PTEN
of the pituitary gland. Cancer. 8(3):616-22. J, De Raedt T, Heyns I, Rosenbaum T, et al. Res. 14(4):1015-24. PMID:18281533 mutation. Nat Clin Pract Oneel. 5(6):357-61.
PMID:14379151 (2006). Comprehensive NF1 screening on cul- 1580. Marano SR, Johnson PC, Spetzler RF PMID:18431376
1548. Lusis EA, Watson MA, Chicoine MR, tured Schwann cells from neurofibromas. Hum (1988). Recurrent Lhermitte-Duclos disease in 1595. Marshman LA, Pollock JR, King A, Cha-
Lyman M, Roerig P, Reifenberger G, el al. Mutat. 27(10):1030-40. PMID:16941471 a child. Case report. J Neurosurg. 69(4):599- wda SJ (2005). Primary extradural epithelioid
(2005). Integrative genomic analysis identifies 1565. Maesawa C, Tamura G, lwaya T, Ogasa- 603. PMID:3418394 leiomyosarcoma of the cervical spine: case
NDRG2 as a candidate tumor suppressor gene wara S, Ishida K, Sato N, et al. (1998). Muta- 1581. Marcel V, Dichtel-Danjoy ML, Sagne C, report and literature review. Neurosurgery.
frequently inactivated in clinically aggressive tions in the human homologue of the Droso- Hafsi H, Ma D, Ortiz-Cuaran S, et al. (2011). 57(2):E372, discussion E372. PMID:16094142
meningioma Cancer Res. 65(16):7121-6. phila patched gene in esophageal squamous Biological functions of p53 isoforms through 1596. Martinez R, Roggendorf W, Baretton G,
PMID 16103061 cell carcinoma. Genes Chromosomes Cancer. evolution: lessons from animal and cellular Klein R, Toed! G, Lichter P, et al. (2007). Cyto-
1549. Lutterbach J, Fauchon F, Schild SE, Ch- 21(3):276-9. PMID:9523206 models. Cell Death Differ. 18(12):1815-24. genetic and molecular genetic analyses of giant
ang SM, Pagenstecher A, Volk B, et al. (2002). 1566. Magri L, Cambiaghi M, Cominelli M, Al- PMID:21941372 cell glioblastoma multiforme reveal distinct pro-
Malignant pineal parenchymal tumors in adult faro-Cervello C, Cursi M, Pala M, et al. (2011). 1581A. Marciscano AE, Stemmer-Rachami- files in giant cell and non-giant cell subpopula-
patients: patterns of care and prognostic fac- Sustained activation of m TOR pathway in mov AO, Niemierko A, Larvie M, Curry WT, tions. Cancer Genet Cytogenet. 175(1):26-34.
tors. Neurosurgery. 51(1):44-55, discussion embryonic neural stem cells leads to develop- Barker FG 2nd, et al. (2016). Benign menin- PMID:17498554
55-6. PMID:12182434 ment of tuberous sclerosis complex-associ- giomas (WHO Grade I) with atypical histolo- 1597. Martinez-Diaz H, Kleinschmidt-DeMas-
1550. Lutterbach J, Liegibel J, Koch D, Madlin- ated lesions. Cell Stem Cell. 9(5):447-62. gical features: correlation of histopathological ters BK, Powell SZ, Yachnis AT (2003). Giant
ger A, Frommhold H, Pagenstecher A (2001). PMID:22056141 features with clinical outcomes. J Neurosurg. cell glioblastoma and pleomorphic xanthoast-
Atypical teratoid/rhabdoid tumors in adult pati- 1567. Maher ER, Yates JR, Ferguson-Smith 124(1):106-14. PMID:26274991 rocytoma show different immunohistochemical
ents: case report and review of the literature. J MA (1990). Statistical analysis of the two stage 1582. Marcotte L, Aronica E, Baybis M, Crino PB profiles for neuronal antigens and p53 but share
Neurooncol. 52(1):49-56. PMID:11451202 mutation model in von Hippel-lindau disease, (2012). Cytoarchitectural alterations are wides- reactivity for class Ill beta-tubulin. Arch Pathol
1551. Luyken C, BIOmcke I, Fimmers R, Urbach and in sporadic cerebellar haemangioblastoma pread in cerebral cortex in tuberous sclerosis Lab Med. 127(9):1187-91. PMID:12946225

380 References
1598. Martinez-Salazar A, Su pier M, Rojiani AM gastrointestinal stromal tumors. J Clin Endocri- Z. Abel TW, et al. (2009). Diagnostic utility of (1995). Screening for germ-line mutations in
(1997). Primary intracerebral malignant fibrous nol Metab. 92(8):2938-43. PMID:17535989 SALL4 in primary germ cell tumors of the cen- the NF2 gene. Genes Chromosomes Cancer.
histiocytoma: immunohistochemical findings 1615. Mawrin C, Perry A (2010). Pathologi- tral nervous system: a study of 77 cases. Mod 12(2):117-27. PMID:7535084
and etiopathogenetic considerations. Mod Pa- cal classification and molecular genetics of Pathol. 22(12):1628-36. PMID:19820689 1648. Merino DM, Shlien A, Villani A, Pien-
thol. 10(2):149-54. PMID:9127321 meningiomas. J Neurooncol. 99(3):379-91. 1632. Meijer-Jorna LB, Aronica E, van der Loos kowska M, Mack S, Ramaswamy V, et al.
1599. Martuza RL, Eldridge R (1988). Neu- PMID:20809251 CM, Troost D, van der Wal AC (2012). Conge- (2015). Molecular characterization of choroid
rofibromatosis 2 (bilateral acoustic neurofi- 1616. Mayer-Proschel M, Kalyani AJ, Mujtaba nital vascular malformations-cerebral lesions plexus tumors reveals novel clinically relevant
bromatosis). N Engl J Med. 318(11):684-8. T. Rao MS (1997). Isolation of lineage-restric- differ from extracranial lesions by their immu- subgroups. Clin Cancer Res. 21(1):184-92.
PMID:3125435 ted neuronal precursors from mullipotent neu- ne expression of the glucose transporter pro- PMID:25336695
1600. Marucci G, Di Oto E, Farnedi A, Panzac- roepithelial stem cells. Neuron. 19(4):773-85. tein GLUT1. Clin Neuropathol. 31(3):135-41. 1649. Merker VL, Esparza S, Smith MJ,
chi R, Ligorio C, Foschini MP (2012). Nogo-A: a PMID:9354325 PMID:22551917 Stemmer-Rachamimov A, Plotkin SR (2012).
useful marker for the diagnosis of oligodendro- 1617. Mazur MA, Gururangan S, Bridge JA, 1633. Meis JM, Ho KL, Nelson JS (1990). Clinical features of schwannomatosis: a re-
glioma and for identifying 1p19q codeletion. Cummings T J, Mukundan S, Fuchs H, et al. Gliosarcoma: a histologic and immunohisto- trospective analysis of 87 patients. Oncologist.
Hum Pathol. 43(3):374-80. PMID:21835431 (2005). lntracranial Ewing sarcoma. Pediatr chemical reaffirmation. Mod Pathol. 3(1):19-24. 17(10):1317-22. PMID:22927469
1601. Maruoka R, Takenouchi T, Torii C, Shimi- Blood Cancer. 45(6):850-6. PMID:15929128 PMID:2155418 1650. Merlin E, Chabrier S, Verkarre V, Cra-
zu A, Misu K, Higasa K, et al. (2014). The use 1618. McCabe MG, lchimura K, Liu L, Plant 1634. Mellinghoff IK, Wang MY, Vivanco I, mer E, Delabesse E, Stephan JL (2008).
of next-generation sequencing in molecular dia- K, Backlund LM, Pearson OM, et al. (2006). Haas-Kogan DA, Zhu S, Dia EQ, et al. (2005). Primary leptomeningeal ALK+ lymphoma in
gnosis of neurofibromatosis type 1: a validation High-resolution array-based comparative ge- Molecular determinants of the response of gli- a 13-year-old child. J Pediatr Hematol Oneal.
study. Genet Test Mal Biomarkers. 18(11):722- nomic hybridization of medulloblastomas and oblastomas to EGFR kinase inhibitors. N Engl J 30(12):963-7. PMID:19131793
35. PMID:25325900 supratentorial primitive neuroectodermal tu- Med. 353(19):2012-24. PMID:16282176 1651. Merrell R, Nabors LB, Perry A, Palmer
1602. Maruya J, Seki Y, Morita K, Nishimaki K, mors. J Neuropathol Exp Neural. 65(6):549-61. 1635. Memoli VA, Brown EF, Gould VE (1984). CA (2006). 1 p/19q chromosome deletions in
Minakawa T (2006). Meningeal hemangiope- PMID:16783165 Glial fibrillary acidic protein (GFAP) immuno- metastatic oligodendroglioma. J Neurooncol.
ricytoma manifesting as massive intracranial 1619. McClatchey Al, Giovannini M (2005). reactivity in peripheral nerve sheath tumors. 80(2):203---7. PMID:16710746
hemorrhage-two case reports. Neural Med Chir Membrane organization and tumorigenesis-the Ultrastruct Pathol. 7(4):269-75. PMI0:6543600 1652. Messiaen LM, Callens T, Mortier G,
(Tokyo). 46(2):92-7. PMID:16498220 NF2 tumor suppressor, Merlin. Genes Dev. 1636. Mena H, Ribas JL, Enzinger FM, Parisi Beysen 0, Vandenbroucke I, Van Roy N, et
1603. Massimino M, Antonelli M, Gandola 19(19):2265-77. PMID:16204178 JE (1991 ). Primary angiosarcoma of the central al. (2000). Exhaustive mutation analysis of the
L, Miceli R, Pollo B, Biassoni V, et al. (2013). 1620. McCubrey JA, Steelman LS, Abrams SL, nervous system. Study of eight cases and re- NF1 gene allows identification of 95% of muta-
Histological variants of medulloblastoma are Lee JT, Chang F, Bertrand FE, et al. (2006). Ro- view of the literature. J Neurosurg. 75(1 ):73-6. tions and reveals a high frequency of unusual
the most powerful clinical prognostic indi- les of the RAF/MEK/ERK and Pl3K/PTEN/AKT PMID:2045922 splicing defects. Hum Mutat. 15(6):541-55.
cators. Pediatr Blood Cancer. 60(2):210-6. pathways in malignant transformation and drug 1637. Mena H, Ribas JL, Pezeshkpour GH, PMID:10862084
PMID:22693015 resistance. Adv Enzyme Regul. 46:249-79. Cowan DN, Parisi JE (1991). Hemangiope- 1653. Mester J, Eng C (2012). Estimate of
1604. Massimino M, Solera CL, Garre ML, Bi- PMID:16854453 ricytoma of the central nervous system: a de nova mutation frequency in probands with
assoni V, Gama A, Genitori L, et al. (2011 ). Se- 1621. McGarvey TW, Maruta Y, Tomaszews- review of 94 cases. Hum Pathol. 22(1):84-91. PTEN hamartoma tumor syndrome. Genet
cond-look surgery for ependymoma: the Italian ki JE, Linnenbach AJ, Malkowicz SB (1998). PMID:1985083 Med. 14(9):819-22. PMID:22595938
experience. J Neurosurg Pediatr. 8(3):246-50. PTCH gene mutations in invasive transitional 1638. Mena H, Rushing EJ, Ribas JL, Delahunt 1654. Mete 0, Lopes MB, Asa SL(2013). Spind-
PMID:21882914 cell carcinoma of the bladder. Oncogene. B, McCarthy WF (1995). Tumors of pineal pa- le cell oncocytomas and granular cell tumors of
1605. Masuoka J, Brandner S, Paulus W, 17(9):1167-72. PMID:9764827 renchymal cells: a correlation of histological the pituitary are variants of pituicytoma. Am J
Soffer D, Vital A, Chimelli L, et al. (2001). 1622. McGirr SJ, Kelly PJ, Scheithauer BW features, including nucleolar organizer re- Surg Pathol. 37(11):1694-9. PMID:23887161
Germline SDHD mutation in paraganglioma (1987). Stereotactic resection of juvenile pi- gions, with survival in 35 cases. Hum Pathol. 1655. Mete 0, Tischler AS, de Krijger R, McNi-
of the spinal cord. Oncogene. 20(36):5084-6. locytic astrocytomas of the thalamus and 26(1):20-30. PMtD:7821912 col AM, Eisenhofer G, Pacak K, et al. (2014).
PMID:11526495 basal ganglia. Neurosurgery. 20(3):447-52. 1639. Mendrzyk F, Korshunov A, Benner A, Protocol for the examination of specimens
1606. Mathew RK, O'Kane R, Parslow R, Stiller PMID:3553982 Toedt G, Pfister S, Radlwimmer B, et al. (2006). from patients with pheochromocytomas and
C, Kenny T, Picton S, et al. (2014). Comparison 1623. McHugh BJ, Baranoski JF, Malhotra A, Identification of gains on 1 q and epidermal extra-adrenal paragangliomas. Arch Pathol Lab
of survival between the UK and US after sur- Vortmeyer AO, Sze G, Duncan CC (2014). ln- growth factor receptor overexpression as Med. 138(2):182-8. PMID:24476517
gery for most common pediatric CNS tumors. tracranial infantile hemangiopericytoma. J Neu- independent prognostic markers in intracra- 1656. Metellus P, Bouvier C, Guyotat J, Fuentes
Neuro Oneel. 16(8):1137-45. PMID:24799454 rosurg Pediatr. 14(2):149-54. PMID:24905842 nial ependymoma. Clin Cancer Res. 12(7 Pt S, Jouvet A, Vasiljevic A, et al. (2007). Solitary
1607. Mathews JD, Forsythe AV, Brady Z, But- 1624. McKusick VA: Online Mendelian Inheri- 1):2070-9. PMID:16609018 fibrous tumors of the central nervous system:
ler MW, Goergen SK, Byrnes GB, et al. (2013). tance in Man !www.omim.org] 1640. Mendrzyk F, Radlwimmer B, Joos S, Ko- clinicopathological and therapeutic considera-
Cancer risk in 680,000 people exposed to 1625. Mclendon RE, Bentley RC, Parisi JE, kocinski F, Benner A, Stange DE, et al. (2005). tions of 18 cases. Neurosurgery. 60(4):715-22,
computed tomography scans in childhood or Tien RD, Harrison JC, Tarbell NJ, et al. (1997). Genomic and protein expression profiling discussion 722. PMID:17415209
adolescence: data linkage study of 11 million Malignant supratentorial glial-neuronal neo- identifies CDK6 as novel independent progno- 1657. Meyer P, Eberle MM, Probst A, Tolnay
Australians. BMJ. 346:12360. PMID:23694687 plasms: report of two cases and review of the stic marker in medulloblastoma. J Clin Oneal. M (2000). !Ganglioglioma of optic nerve in
1608. Mathews T. Moossy J (1974). Gliomas literature. Arch Pathol Lab Med. 121 (5):485-92. 23(34):8853-62. PMID:16314645 neurofibromatosis type 1. Case report and re-
containing bone and cartilage. J Neuropathol PMID:9167602 1641. Menke JR, Raleigh DR, Gown AM, Tho- view of the literature]. Klin Monbt Augenheilkd.
Exp Neural. 33(3):456-71. PMID:4365915 1626. McManamy CS, Lamont JM, Taylor RE, mas S, Perry A, Tihan T (2015). Somatostatin 217(1):55-8. PMID:10949818
1609. Mathen B, Carpentier A, Clemenceau Cole M, Pearson AD, Clifford SC, et al.: Uni- receptor 2a is a more sensitive diagnostic 1658. Meyer-Puttlitz B, Hayashi Y, Waha A,
S, Boch AL, Bitar A, Mokhtari K, et al. (2012). ted Kingdom Children's Cancer Study Group marker of meningioma than epithelial membra- Rollbrocker B, Bostrom J, Wiestler OD, et al.
!Paraganglioma of the cauda equina region: (2003). Morphophenotypic variation predicts ne antigen. Acta Neuropathol. 130(3):441-3. (1997). Molecular genetic analysis of giant
Report of six cases and review of the literature]. clinical behavior in childhood non-desmoplastic PMID:26195322 cell glioblastomas. Am J Pathol. 151(3):853-7.
Neurochirurgie. 58(6):341-5. PMID:22770767 medulloblastomas. J Neuropathol Exp Neural. 1642. Menke FH, Kaspers GL, Meijer GA, PMID:9284834
1610. Matsushima T, Inoue T, Takeshita I, Fukui 62(6):627-32. PMID:12834107 Claes K, van Hagen JM, Gille JJ (2004). A ho- 1659. Meyers SP, Khademian ZP, Biegel JA,
M, lwaki T. Kitamoto T (1988). Choroid plexus 1627. McManamy CS, Pears J, Weston CL, mozygous MSH6 mutation in a child with cafe- Chuang SH, Korones DN, Zimmerman RA
papillomas: an immunohistochemical study Hanzely Z, Ironside JW, Taylor RE, et al.; Clini- au-lait spots, oligodendroglioma and rectal can- (2006). Primary intracranial atypical teratoid/
with particular reference to the coexpression cal Brain Tumour Group (2007). Nodule forma- cer. Fam Cancer. 3(2):123-7. PMID:15340263 rhabdoid tumors of infancy and childhood:
of prealbumin. Neurosurgery. 23(3):384-9. tion and desmoplasia in medulloblastomas-de- 1643. Merchant TE, Jenkins JJ, Burger PC, MRI features and patient outcomes. AJNR Am
PMID:3226520 fining the nodular/desmoplastic variant and its Sanford RA, Sherwood SH, Jones-Wallace D, J Neuroradiol. 27(5):962-71. PMID:16687525
1611. Matsutani M; Japanese Pediatric Brain biological behavior. Brain Pathol. 17(2):151-64. et al. (2002). Influence of tumor grade on time to 1660. Meyers SP, Khademian ZP, Chuang SH,
Tumor Study Group (2001 ). Combined chemo- PMID:17388946 progression after irradiation for localized epen- Pollack IF, Korones ON, Zimmerman RA(2004).
therapy and radiation therapy for CNS germ cell 1628. McMenamin ME, Fletcher CD (2001). dymoma in children. Int J Radial Oneal Biol Choroid plexus carcinomas in children: MRI
tumors-the Japanese experience. J Neuroon- Expanding the spectrum of malignant change Phys. 53(1):52-7. PMID:12007941 features and patient outcomes. Neuroradiology.
col. 54(3):311-6. PMID:11767296 in schwannomas: epithelioid malignant change, 1644. Merchant TE, Li C, Xiong X, Kun LE, 46(9):770-80. PMID:15309348
1612. Matsutani M, Sano K, Takakura K, Fuji- epithelioid malignant peripheral nerve sheath Boop FA, Sanford RA (2009). Conformal ra- 1661. Michal M, Kazakov DV, Belousova I, Bi-
maki T, Nakamura 0, Funata N, et al. (1997). tumor, and epithelioid angiosarcoma: a study diotherapy after surgery for paediatric epen- sceglia M, Zamecnik M, Mukensnabl P (2004).
Primary intracranial germ cell tumors: a clinical of 17 cases. Am J Surg Pathol. 25(1):13---25. dymoma: a prospective study. Lancet Oneal. A benign neoplasm with histopathological featu-
analysis of 153 histologically verified cases. J PMID:11145248 10(3):258-66. PMID:19274783 res of both schwannoma and retiform perineu-
Neurosurg. 86(3):446-55. PMID:9046301 1629. McNatt SA, Gonzalez-Gomez I, Nelson 1645. Merchant TE, Pollack IF, Loeffler JS rioma (benign schwannoma-perineurioma): a
1613. Matthies C, Samii M (1997). Management MD, McComb JG (2005). Synchronous rnul- (2010). Brain tumors across the age spectrum: report of six cases of a distinctive soft tissue tu-
of 1000 vestibular schwannomas (acoustic neu- ticentric pleomorphic xanthoastrocytoma: case biology, therapy, and late effects. Semin Radial mor with a predilection for the fingers. Virchows
romas): clinical presentation. Neurosurgery. report. Neurosurgery. 57(1):E191, discussion Oneal. 20(1):58-66. PMID:19959032 Arch. 445(4):347-53. PMID:15322875
40(1):1-9, discussion 9-10. PMID:8971818 E191. PMID:15987556 1646. Mercuri S, Gazzeri R, Galarza M, Espo- 1662. Miettinen M, Shekitka KM, Sobin LH
1614. Matyakhina L, Bei TA, McWhinney SR, 1630. Medhkour A, Traul D, Husain M (2002). sito S, Giordano M (2005). Primary meningeal (2001 ). Schwannomas in the colon and rec-
Pasini B, Cameron S, Gunawan B, et al. (2007). Neonatal subependymal giant cell ast- pheochromocytoma: case report. J Neurooncol. tum: a clinicopathologic and immunohistoche-
Genetics of carney triad: recurrent losses at rocytoma. Pediatr Neurosurg. 36(5):271-4. 73(2):169-72. PMID:15981108 mical study of 20 cases. Am J Surg Pathol.
chromosome 1 but lack of germline mutations PMID:12053047 1647. Merel P, Hoang-Xuan K, Sanson M, 25(7):846-55. PMID:11420455
in genes associated with paragangliomas and 1631. Mei K, Liu A, Allan RW, Wang P, Lane Bijlsma E, Rouleau G, Laurent-Puig P, et al. 1663. Migheli A, Cavalla P, Marino S, Schiffer

References 381
D (1994). A study of apoptosis in normal and 1679. Ming JE, Kaupas ME, Roessler E. Brun- with special reference to its cytogenesis. Neurol R, Gesk S, Martin-Subero JI, Schaller C, Van
pathologic nervous tissue after in situ end-labe- ner HG, Nance WE, Stratton RF, el al. (1998). Med Chir (Tokyo). 33(7):420-4. PMID:7692317 Roost D, et al. (2002). lnterphase cytogenetic
ling of DNA strand breaks. J Neuropathol Exp Mutations of PATCHED in holoprosencephaly. 1696. Mobley BC, Roulston D, Shah GV, Bi- analysis of lymphoma-associated chromoso�
Neurol. 53(6):606-16. PMID:7525880 Am J Hum Genet. 63:A140-140. jwaard KE, McKeever PE (2006). Peripheral mal breakpoints in primary diffuse large B-cell
1664. Milbouw G, Born JD, Martin D, Collignon 1680. Mirsattari SM, Chong JJ, Hammond RR, primitive neuroectodermal tumor/Ewing's lymphomas of the central nervous system.
J, Hans P, Reznik M, et al. (1988). Clinical and Megyesi JF, Macdonald DR, Lee DH, et al. sarcoma of the craniospinal vault: case re- J Neuropathol Exp Neurol. 61(10):926-33.
radiological aspects of dysplastic gangliocyto- (2011 ). Do epileptic seizures predict outcome in ports and review. Hum Pathol. 37(7):845-53. PMID:12387458
ma (Lhermitte-Duclos disease): a report of two patients with oligodendroglioma? Epilepsy Res. PMID:16784984 1711. Moody P, Murtagh K, Piduru S, Brem s
cases with review of the literature. Neurosur- 94(1-2):39-44. PMID:21315558 1697. Modena P, Lualdi E, Facchinetti F, Galli L, Murtagh R, Rojiani AM (2012). Tumor-to-tumo;
gery. 22(1 Pt 1):124-8. PMID:3278250 1681. Mishima K, Nakamura M, Nakamura Teixeira MR, Pilotti S, et al. (2005). SMARCB1/ metastasis: pathology and neuroimaging consi-
1665. Millard NE, Dunkel IJ (2014). Advances H, Nakamura 0, Funata N, Shitara N (1992). INl1 tumor suppressor gene is frequently inac- derations. Int J Clin Exp Pathol. 5(4):367-73.
in the management of central nervous system Leptomeningeal dissemination of cerebellar pi- tivated in epithelioid sarcomas. Cancer Res. PMID:22670183
germ cell tumors. Curr Oncol Rep. 16(7):393. locytic astrocytoma. Case report. J Neurosurg. 65(10):4012-9. PMID:15899790 1712. Moran CA, Rush W, Mena H (1997).
PMID:24838613 77(5):788-91. PMID:1403124 1698. Moles KJ, Gowans GC, Gedela S, Bev- Primary spinal paragangliomas: a clinicopa-
1666. Miller CA, Torack RM (1970). Secretory 1682. Mishra T, Goel NA, Goel AH (2014). Pri- ersdorf D, Yu A, Seaver LH, et al. (2012). NF1 thological and immunohistochemical study
ependymoma of the filum terrninale. Acta Neu- mary paraganglioma of the spine: A clinicopa- microduplications: identification of seven nonre- of 30 cases. Histopathology. 31(2):167-73.
ropathol. 15(3):240-50. PMID:4193811 thological study of eight cases. J Craniovertebr lated individuals provides further characterizati- PMID:9279569 .
1667. Miller CR, Dunham GP, Scheithauer BW, Junction Spine. 5(1):20-4. PMID:25013343 on of the phenotype. Genet Med. 14(5):508-14. 1713. Morantz RA, Feigin I, Ransohoff J 3rd
Perry A (2006). Significance of necrosis in gra- 1683. Mistry M, Zhukova N, Merico D, Rako- PMID:22241097 (1976). Clinical and pathological study of 24
ding of oligodendroglial neoplasms: a clinicopa- poulos P, Krishnatry R, Shago M, et al. (2015). 1699. Momota H, Iwami K, Fujii M, Motomura cases of gliosarcoma. J Neurosurg. 45(4):398-
thologic and genetic study of newly diagnosed BRAF mutation and CDKN2A deletion defi- K, Natsume A, Ogino J, et al. (2011 ). Rhabdoid 408. PMID 956876
high-grade gliomas. J Clin Oncol. 24(34):5419- ne a clinically distinct subgroup of childhood glioblastoma in a child: case report and litera- 1714. Moriguchi S, Yamashita A, Marutsuka
26. PMID:17135643 secondary high-grade glioma. J Clin Oncol. ture review. Brain Tumor Pathol. 28(1):65-70. K, Yoneyama T, Nakano S, Wakisaka S, et al.
1668. Miller S, Rogers HA, Lyon P, Rand V, 33(9):1015-22. PMID:25667294 PMID:21213124 (2006). Atypical extraventricular neurocytoma.
Adamowicz-Brice M, Clifford SC, et al. (2011). 1684. Mitchell A, Scheithauer BW, Ebersold MJ, 1700. Momota H, Narita Y, Maeshima AM, Miya- Pathol Int. 56(1):25-9. PMID:16398676
Genome-wide molecular characterization of Forbes GS (1991). lntracranial fibromatosis. kita Y, Shinomiya A, Maruyama T, et al. (2010). 1715. Moskowitz SI, Jin T, Prayson RA (2006).
central nervous system primitive neuroectoder- Neurosurgery. 29(1 ):123-6. PMID:1870673 Prognostic value of immunohistochemical profi- Role of MIB1 in predicting survival in patients
mal tumor and pineoblastoma. Neuro Oncol. 1685. Mitchell A, Scheithauer BW, Unni KK, le and response to high-dose methotrexate the- with glioblastomas. J Neurooncol. 76(2):193-
13(8):866-79. PMID:21798848 Forsyth PJ, Wold LE, McGivney DJ (1993). rapy in primary CNS lymphoma. J Neurooncol. 200. PMID:16234986
1669. Miller S, Ward JH, Rogers HA, Lowe Chordoma and chondroid neoplasms of the 98(3):341-8. PMID:20012911 1716. Moss TH (1984). Observalions on
J, Grundy RG (2013). Loss of INl1 protein spheno-occiput. An immunohistochemical 1701. Monje M, Mitra SS, Freret ME, Raveh TB, the nature of subependymoma: an electron
expression defines a subgroup of aggressive study of 41 cases with prognostic and noso- Kim J, Masek M, et al. (2011). Hedgehog-res- microscopic study. Neuropathol Appl Neurobiol.
central nervous system primitive neuroecto- logic implications. Cancer. 72(10):2943-9. ponsive candidate cell of origin for diffuse intrin- 10(1):63-75. PMID:6738805
dermal tumors. Brain Pathol. 23(1):19-27. PMID:7693324 sic pontine glioma. Proc Natl Acad Sci U S A. 1717. Mott RT, Goodman BK, Burchette JL,
PMID:22672440 1686. Mittal P, Gupta K, Saggar K, Kaur S 108(11):4453-8. PMID:21368213 Cummings T J (2005). Loss of chromosome 13
1670. Min HS, Lee JY, Kim SK, Park SH (2013). (2009). Adult medulloblastoma mimicking 1702. Montesinos-Rongen M, Besleaga R, in a case of soft tissue perineurioma. Clin Neu-
Genetic grouping of medulloblastomas by re- Lhermitte-Duclos disease: can diffusion weigh- Heinsohn S, Siebert R, Kabisch H, Wiestler OD, ropathol. 24(2):69-76. PMID:15803806
presentative markers in pathologic diagnosis. ted imaging help? Neurol India. 57(2):203-5. et al. (2004). Absence of simian virus 40 DNA 1718. Mrak RE, Yasargil MG, Mohapatra G, Ea-
Transl Oncol. 6(3):265-72. PMID:23730405 PMID:19439857 sequences in primary central nervous system rel J Jr, Louis DN (2004). Atypical extraventri-
1671. Min HS, Lee YJ, Park K, Cho BK, Park 1687. Miyagami M, Katayama Y, Nakamura lymphoma in HIV-negative patients. Virchows cular neurocytoma with oligodendroglioma-like
SH (2006). Medulloblastoma: hislopathologic S (2000). Clinicopathological study of vas- Arch. 444(5):436-8. PMID:15042369 spread and an unusual pattern of chromosome
and molecular markers of anaplasia and biolo- cular endothelial growth factor (VEGF). p53, 1703. Montesinos-Rongen M, Godlewska 1p and 19q loss. Hum Pathol. 35(9):1156-9.
gic behavior. Acta Neuropathol. 112(1):H-20. and proliferative potential in familial von Hip- E, Brunn A, Wiestler OD, Siebert R, Deckert PMID:15343519
PMID:16691420 pel-lindau disease and sporadic hemangiob- M (2011 ). Activating L265P mutations of the 1719. Mu Q, Yu J, Ou L, Hu X, Gao H, Liu P,
1672. Min KW, Scheithauer BW (1990). Pi- lastomas. Brain Tumor Pathol. 17(3):111-20. MYD88 gene are common in primary central et al. (2015). Spindle cell oncocytoma of the
neal germinomas and testicular seminoma: a PMID:11310918 nervous system lymphoma. Acta Neuropathol. adenohypophysis: two case reports and a re-
comparative ultrastructural study with special 1688. Miyagi Y, Suzuki SO, lwaki T, Shima F, ls- 122(6):791-2. PMID:22020631 view of the literature. Mol Med Rep. 12(1):871-
references to early carcinomatous transfor- hido K, Araki T, et al. (2001 ). Pleomorphic xan- 1704. Montesinos-Rongen M, Hans VH, Eis-Hii- 6. PMID:25777996
mation. Ultrastruct Pathol. 14(6):483-96. thoastrocytoma with predominantly exophytic binger AM, Prinz M, Schaller C, Van Roost D, 1720. Mueller W, Mizoguchi M, Silen E,
PMID:2281547 growth: case report. Surg Neurol. 56(5):330-2. et al. (2001 ). Human herpes virus-8 is not as- D'Amore K, Nutt CL, Louis DN (2005). Muta-
1673. Min KW, Scheithauer BW (1997). Clear PMID:11750009 sociated with primary central nervous system tions of the PIK3CA gene are rare in human
cell ependymoma: a mimic of oligodendroglio- 1689. Miyamori T. Mizukoshi H, Yamano K, lymphoma in HIV-negative patients. Acta Neu- glioblastoma. Acta Neuropathol. 109(6):654-5.
ma: clinicopathologic and ultrastructural con- Takayanagi N, Sugino M, Hayase H, et al. ropathol. 102(5):489-95. PMID:11699563 PMID:15924252 ,,ii;
siderations. Am J Surg Pathol. 21 (7):820-6. (1990). lntracranial chondrosarcoma-case 1705. Montesinos-Rongen M, Kiippers R, 1721. Muller C, Adroos N, Lockhat Z, SlavikT,
PMID:9236838 report. Neurol Med Chir (Tokyo). 30(4):263-7. Schluter D, Spieker T, Van Roost D, Schaller Kruger H (2011 ). Toothy craniopharyngioma:
1674. Min KW, Scheilhauer BW, Bauserman SC PMID:1696697 C, et al. (1999). Primary central nervous system a literature review and case report of cranio-
(1994). Pineal parenchymal tumors: an ultra- 1690. Miyata H, Chiang AC, Vinters HV (2004). lymphomas are derived from germinal-center B pharyngioma with extensive odontogenic diffe-
structural study with prognostic implications. Ul- Insulin signaling pathways in cortical dysplasia cells and show a preferential usage of the V4- rentiation and tooth formation. Childs Nerv Syst.
trastruct Pathol. 18(1-2):69-85. PMID:8191649 and TSC-tubers: tissue microarray analysis. 34 gene segment. Am J Pathol. 155(6):2077- 27(2):323-6. PMID:20922394
1675. Min KW, Seo IS, Song J (1987). Post- Ann Neurol. 56(4):510-9. PMID:15455398 86. PMID:10595937 1722. Mullins KJ, Rubio A, Myers SP, Korones
natal evolution of the human pineal gland. 1691. Miyata H, Ryufuku M, Kubota Y, Ochiai 1706. Montesinos-Rongen M, Schmitz R, Brunn DN, Pilcher WH (1998). Malignant ependymo-
An immunohistochemical study. Lab Invest. T, Niimura K, Hori T (2012). Adult-onset angio- A, Gesk S, Richter J, Hong K, et al. (2010). mas in a patient with Turcot's syndrome: case
57(6):724-8. PMID:3695415 centric glioma of epithelioid cell-predominant Mutations of CARD11 but notTNFAIP3 may ac- report and management guidelines. Surg Neu-
1676. Minaguchi T, Waite KA, Eng C (2006). type of the mesial temporal lobe suggestive tivate the NF-kappaB pathway in primary CNS rol. 49(3):290-4. PMID:9508117
Nuclear localization of PTEN is regulated by of a rare but distinct clinicopathological subset lymphoma. Acta Neuropathol. 120(4):529-35. 1723. Mur P, Mollejo M, Ruano Y, de Lope AR,
Ca(2+) through a tyrosil phosphorylation-inde- within a spectrum of angiocentric cortical epen- PMID:20544211 Fiaiio C, Garcia JF, et al. (2013). Codelelion of
pendent conformational modification in major dymal tumors. Neuropathology. 32(5):479-91. 1707. Montesinos-Rongen M, Schmitz R, 1 p and 19q determines distinct gene methyla-
vault protein. Cancer Res. 66(24):11677-82. PMID:22151480 Courts C, Stenzel W, Bechtel D, Niedobitek G, tion and expression profiles in !DH-mutated
PMID:17178862 1692. Mizoguchi M, Nutt CL, Mohapatra G, et al. (2005). Absence of immunoglobulin class oligodendroglial tumors. Acta Neuropathol.
1677. Minehan KJ, Brown PD, Scheithauer Louis DN (2004 ). Genetic alterations of phos- switch in primary lymphomas of the central 126(2):277-89. PMID:23689617
BW, Krauss WE, Wright MP (2009). Progno- phoinositide 3-kinase subunit genes in human nervous system. Am J Pathol. 166(6):1773-9. 1724. Murali R, Wiesner T, Rosenblum MK.
sis and treatment of spinal cord astrocytoma. glioblastomas. Brain Pathol. 14(4):372-7. PMID:15920162 Bastian BC (2012). GNAQ and GNA11 muta-
Int J Radial Oncol Biol Phys. 73(3):727-33. PMID:15605984 1708. Montesinos-Rongen M, Schafer E, Sie- tions in melanocylomas of the central nervous
PMID:18687533 1693. Mizuguchi M, Ikeda K, Takashima S bert R, Deckert M (2012). Genes regulating system. Acta Neuropathol. 123(3):457-9.
1678. Minehan KJ, Shaw EG, Scheithauer (2000). Simultaneous loss of hamartin and the B cell receptor pathway are recurrently PMID:22307269
BW, Davis DL, Onofrio BM (1995). Spinal tuberin from the cerebrum, kidney and heart mutated in primary central nervous system 1725. Murray JM, Morgello S (2004). Polyo-
cord astrocyloma: pathological and treatment with tuberous sclerosis. Acta Neuropathol. lymphoma. Acta Neuropathol. 124(6):905-6. maviruses and primary central nervous sys-
considerations. J Neurosurg. 83(4):590-5. 99(5):503-10. PMID:10805093 PMID:23138649 tem lymphomas. Neurology. 63(7):1299-301.
PMID:7674006 1694. Mizuguchi M, Kato M, Yamanouchi H, 1709. Montesinos-Rongen M, Van Roost D, PMID:15477558
1678A. Ming JE, Kaupas ME, Roessler E, Ikeda K, Takashima S (1996). Loss of tuberin Schaller C, Wiestler OD, Deckert M (2004). Pri- 1726. Murthy A, Gonzalez-Agosti C, Cordero E,
Brunner HG, Golabi M, Tekin M, et al. (2002). from cerebral tissues with tuberous sclerosis mary diffuse large B-cell lymphomas of the cen- Pinney D, Candia C, Solomon F, el al. (1998).
Mutations in PATCHED-1, the receptor for and astrocytoma. Ann Neurol. 40(6):941-4. tral nervous system are targeted by aberrant NHE-RF, a regulatory cofactor for Na(+)-H+
SONIC HEDGEHOG, are associated with ho- PMID:9007104 somatic hyperrnutation. Blood. 103(5):1869-75. exchange, is a common interactor for mer- :!
loprosencephaly. Hum Genet. 110(4):297-301. 1695. Mizuno J, Iwata K, Takei Y (1993). lm- PMID:14592832 lin and ERM (MERM) proteins. J Biol Chem.
PMID:11941477 munohistochemical study of hemangioblastoma 1710. Montesinos-Rongen M, Ziihlke-Jenisch 273(3):1273-6. PMID:9430655

382 References
1727. Mustafa D, Swagemakers S, French P, paraganglioma: a case report and review of Pleomorphic xanthoastrocytoma in two siblings imaging. J Neurooncol. 121(1):209-15.
Luider TM, van der Spek P, Kremer A, et al. the literature. Neuroradiology. 47(10):753-7. with neurofibromatosis type 1 (NF-1). Clin Neu- PMID:25293439
(2013). Structural and expression differences PMID:16047139 ropathol. 31(1):54-6. PMID:22192706 1779. Nguyen SA, Stechishin OD, Luchman
between the vasculature of pilocytic astrocyto- 1743. Nair S, Fort JA, Yachnis AT, Williams CA 1762. Neelima R, Easwer HV, Kapilamoorthy HA, Lun XO, Senger DL, Robbins SM, et
mas and glioblastomas. J Neuropathol Exp (2015). Optic nerve pilomyxoid astrocytoma in TR, Hingwala DR, Radhakrishnan VV (2012). al. (2014). Novel MSH6 mutations in treat-
Neurol. 72(12):1171-81. PMID:24226271 a patient with Noonan syndrome. Pediatr Blood Embryonal tumor with multilayered rosettes: ment-na"ive glioblastoma and anaplastic oli-
1728. Mut M, Schiff D, Shaffrey ME (2005). Cancer. 62(6):1084-6. PMID:25585602 Two case reports with a review of the literature. godendroglioma contribute to temozolomide
Metastasis to nervous system: spinal epidural 1744. Naitoh Y, Sasajima T, Kinouchi H, Mika- Neurol India. 60(1 ):96-9. PMID:22406791 resistance independently of MGMT promoter
and intramedullary metastases. J Neurooncol. wa S, Mizoi K (2002). Medulloblastoma with 1763. Neff BA, Willcox TO Jr, Sataloff RT methylation. Clin Cancer Res. 20(18):4894-
75(1):43-56. PMID:16215815 extensive nodularity: single photon emission (2003). lntralabyrinthine schwannomas. Otol 903. PMID 25078279
1729. Myers MP, Stolarov JP, Eng C, Li J, Wang CT study with iodine-123 metaiodobenzylgua- Neurotol. 24(2):299-307. PMID 12621348 1780. Ni HC, Chen SY, Chen L, Lu DH, Fu Y J,
SI, Wigler MH, et al. (1997). P-TEN, the tumor nidine. AJNR Am J Neuroradiol. 23(9):1564-7. 1764. Nelen MR, Kremer H, Konings IB, Schou- Piao YS (2015). Angiocentric glioma: a report of
suppressor from human chromosome 10q23, is PMID:12372749 te F, van Essen AJ, Koch R, et al. (1999). No- nine new cases, including four with atypical his-
a dual-specificity phosphatase. Proc Natl Acad 1745. Najm I, Jehi L, Palmini A, Gonzalez-Mar- vel PTEN mutalions in patients with Cowden tological features. Neuropathol Appl Neurobiol.
Sci USA. 94(17):9052-7. PMID:9256433 tinez J. Paglioli E, Bingaman W (2013). Tem- disease: absence of clear genotype-phenotype 41(3):333-46. PMID:24861831
1730. Miiller HL, Gebhardt U, Teske C, Faldum poral patterns and mechanisms of epilepsy correlations. Eur J Hum Genet. 7(3):267-73. 1781. Ni Y, He X, Chen J, Moline J, Mester J,
A, Zwiener I, Wanmuth-Metz M, et al.: Study surgery failure. Epilepsia. 54(5):772-82. PMID:10234502 Orloff MS, et al. (2012). Germline SDHx vari-
Committee of KRANIOPHARYNGEOM 2000 PMID:23586531 1765. Nelen MR, Padberg GW, Peeters EA, Lin ants modify breast and thyroid cancer risks in
(2011). Post-operative hypothalamic lesions 1746. Nakagawa Y, Perentes E, Rubinstein LJ AY, van den Helm B, Frants RR, et al. (1996). Cowden and Cowden-like syndrome via FAD/
and obesity in childhood craniopharyngioma: (1986). lmmunohistochemical characterization Localization of the gene for Cowden disease to NAO-dependant destabilization of p53. Hum
results of the multinational prospective trial of oligodendrogliomas: an analysis of multi- chromosome 10q22-23. Nat Genet. 13(1):114- Mol Genet. 21(2):300-10. PMID:21979946
KRANIOPHARYNGEOM 2000 after 3-year ple markers. Acta Neuropathol. 72(1):15-22. 6. PMID:8673088 1782. Ni Y, Zbuk KM, Sadler T, Patocs A,
follow-up. Eur J Endocrinol. 165(1):17-24. PMID:2435103 1766. Nelen MR, van Staveren WC, Pee- Lobo G, Edelman E, et al. (2008). Germline
PMID:21490122 1747. Nakagawa Y, Perentes E, Rubinstein ters EA, Hassel MB, Gorlin RJ, Hamm H, et mutations and variants in the succinate dehy-
1731. Myung JK, Byeon SJ, Kim B, Suh J. Kim LJ (1987). Non-specificity of anti-carbonic al. (1997). Germline mutations in the PTEN/ drogenase genes in Cowden and Cowden-like
SK, Park CK, et al. (2011). Papillary glioneu- anhydrase C antibody as a marker in human MMAC1 gene in patients wilh Cowden disease. syndromes. Am J Hum Genet. 83(2):261-8.
ronal tumors: a review of cliniccpathologic and neurooncology. J Neuropathol Exp Neurol. Hum Mol Genet. 6(8):1383-7. PMID:9259288 PMID:18678321
molecular genetic studies. Am J Surg Pathol. 46(4):451-60. PMID:3110380 1767. Nemes Z (1992). Fibrohistiocytic diffe- 1783. Nieder C, Spanne 0, Mehta MP, Grosu
35(12):1794-805. PMID:22020040 1748. Nakama S, Higashi T, Kimura A, Yama- rentiation in capillary hemangioblastoma. Hum AL, Geinilz H (2011 ). Presentation, patterns of
1732. Myung JK, Cho HJ, Park CK, Chung muro K, Kikkawa I, Hoshino Y (2005). Double Pathol. 23(7):805-10. PMID:1351864 care, and survival in patients with brain metas-
CK, Choi SH, Kim SK, et al. (2013). Clinico- myxopapillary ependymoma of the cauda equi- 1768. Nestor SL, Perry A, Kurtkaya 0, Abell-Al- tases: what has changed in the last 20 years?
pathological and genetic characteristics of ex- na. J Orthop Sci. 10(5):543-5. PMID:16193371 eff P, Rosemblat AM, Burger PC, et al. (2003). Cancer 117(11 ):2505-12. PMID:24048799
travenlricular neurocytomas. Neuropathology. 1749. Nakamura M, Chiba K, Matsumoto M, Melanocytic colonization of a meningothelial 1784. Nielsen EH, Feldt-Rasmussen U, Pouls-
33(2):111-21. PMID:22672632 Ikeda E, Toyama Y (2006). Pleomorphic xan- meningioma: histopalhological and ultra- gaard L, Kristensen LO, Astrup J, Jerqensen
1733. M611emann M, Wolter M, Felsberg J, thoastrocytoma of the spinal cord. Case report. structural findings with immunohistochemical JO, et al. (2011 ). Incidence of craniopharyn-
Collins VP, Reifenberger G (2005). Frequent J Neurosurg Spine. 5(1):72-5. PMID:16850961 and genetic correlation: case report. Neu- gioma in Denmark (n = 189) and estimated
promoter hypermethylation and low expression 1751. Nakamura M, Saeki N, lwadate Y, Suna- rosurgery. 53(1):211-4, discussion 214-5. world incidence of craniopharyngioma in child-
of the MGMT gene in oligodendroglial tumors. mi K, Osato K, Yamaura A (2000). Neurora- PMID:12823892 ren and adults. J Neuroonccl. 104(3):755-63.
Int J Cancer. 113(3):379-85. PMID:15455350 diological characteristics of pineocytoma and 1769. Neumann HP, Eggert HR, Weigel K, Frie· PMID:21336771
1734. Merk SJ, Rubinstein LJ, Kepes JJ, Pe- pineoblastoma. Neuroradiology. 42(7):509-14. dburg H, Wiestler OD, Schollmeyer P (1989). 1785. Nielsen GP, Stemmer-Rachamimov AO,
rentes E, Uphoff OF (1988). Patterns of epithe- PMID:10952183 Hemangioblastomas of the central nervous lno Y, Moller MB, Rosenberg AE, Louis ON
lial metaplasia in malignant gliomas. II. Squa- 1752. Nakamura M, Watanabe T, Klangby U, system. A 10-year study with special reference (1999). Malignant transformation of neurofi-
mous differentiation of epithelial-like fonmations Asker C, Wiman K, Yonekawa Y, et al. (2001). lo von Hippel-Lindau syndrome. J Neurosurg. bromas in neurofibromatosis 1 is associated
in gliosarcomas and glioblastomas. J Neuropa- p14ARF deletion and methylation in genetic 70(1):24-30. PMID:2909683 with CDKN2A/p16 inactivation. Am J Pathol.
thol Exp Neurol. 47(2):101-18. PMID:3339369 pathways to glioblastomas. Brain Pathol. 1770. Neves S, Maza! PR, Wanschitz J, Rud- 155(6):1879-84. PMID:10595918
1735. Nagai S, Kurimoto M, lshizawa S, 11(2):159-68. PMID:11303791 nay AC, Drlicek M, Czech T, et al. (2001 ). Pseu- 1786. Nieuwenhuis MH, Kets CM, Murphy-Ryan
Hayashi N, Hamada H, Kamiyama H, et al. 1753. Nakamura M, Watanabe T, Yonekawa dogliomatous growth pattern of anaplastic small M, Yntema HG, Evans DG, Colas C, et al.
(2009). A rare astrocytic tumor with rhabdoid Y, Kleihues P, Ohgaki H (2001 ). Promoter cell carcinomas metastatic to the brain. Clin (2014). Cancer risk and genotype-phenotype
features. Brain Tumor Pathol. 26(1):19-24. melhylation of the DNA repair gene MGMT Neuropathol. 20(1 ):38-42. PMID: 11220694 correlations in PTEN hamartoma tumor syndro-
PMID:19408093 in astrocytomas is frequently associated with 1771. Newcomb EW, Madonia WJ, Pisharody me. Fam Cancer. 13(1):57-63. PMID:23934601
1736. Nagaishi M, Kim YH, Mittelbronn M, G:C -> A:T mutations of the TP53 tumor sup- S, Lang FF, Koslow M, Miller DC (1993). Acor- 1787. Niida Y, Stemmer-Rachamimov AO, Lo-
Giangaspero F, Paulus W, Brokinkel B, et al. pressor gene. Carcinogenesis. 22(10):1715-9. relative study of p53 protein alteration and p53 grip M, Tapon D, Perez R, Kwiatkowski DJ, et
(2012). Amplification of the STOML3, FREM2, PMID:11577014 gene mutation in glioblastoma multiforme. Brain al. (2001). Survey of somatic mutations in tu-
and LHFP genes is associated with mesenchy- 1754. Nakamura M, Yang F, Fujisawa H, Pathol. 3(3):229-35. PMI0:8293182 berous sclerosis complex (TSC) hamartomas
mal differentiation in gliosarcoma. Am J Pathol. Yonekawa Y, Kleihues P, Ohgaki H (2000). 1772. Newton HB, Dalton J, Ray-Chaudhury A, suggests different genetic mechanisms for pa-
180(5):1816-23. PMID:22538188 Loss of heterozygosity on chromosome 19 in Gahbauer R, McGregor J (2008). Aggressive thogenesis of TSC lesions. Am J Hum Genet.
1737. Nagaishi M, Paulus W, Brokinkel B, Vital secondary glioblastomas. J Neuropathol Exp papillary glioneuronal tumor: case report and 69(3):493-503. PMID:11468687
A, Tanaka Y, Nakazato Y, et al. (2012). Tran- Neurol. 59(6):539-43. PMI0:10850866 literature review. Clin Neuropathol. 27(5):317- 1788. Niiro T, Tokimura H, Hanaya R, Hirano
scriptional factors for epilhelial-mesenchymal 1755. Nakamura M, Yonekawa Y, Kleihues P, 24. PMID:18808063 H, Fukukura Y, Sugiyma K, et al. (2012). MRI
transition are associated with mesenchymal Ohgaki H (2001 ). Promoter hypenmethylation 1773. Ng HK, Lo ST (1989). Cytokeralin im- findings in patients with central neurocytomas
differentiation in gliosarcoma. Brain Pathol. of the RB1 gene in glioblastomas. Lab Invest. munoreactivity in gliomas. Histopathology. with special reference to differential diagnosis
22(5):670-6. PMID:22288519 81(1):77-82. PMID:11204276 14(4):359-68. PMID:2472343 from other ventricular tumours near the fora-
1738. Nagaishi M, Yokoo H, Nobusawa S, Fujii 1756. Nakano I (2015). Stem cell signature in 1774. Ng HK, Poon WS (1990). Gliosarcoma of men of Monro. J Clin Neurosci. 19(5):681-6.
Y, Sugiura Y, Suzuki R, et al. (2015). Localized glioblastoma: therapeutic development for a the posterior fossa with features of a malignant PMID:22410173
overexpression of alpha-internexin within no- moving target. J Neurosurg. 122(2):324-30. fibrous histiocytoma. Cancer. 65(5):1161-6. 1789. Nishikawa R, Furnari FB, Lin H, Arap W,
dules in multinodular and vacuolating neuronal PMID:25397368 PMID:2154322 Berger MS, Cavenee WK, et al. (1995). Loss
tumors. Neuropathology. PMID:26073706 1757. Narod SA, Parry OM, Parboosingh J, Le- 1775. Ng HK, Poon WS (1999). Diffuse lepto- of P161NK4 expression is frequent in high
1739. Nagao K, Togawa N, Fujii K, Uchikawa noir GM, Ruttledge M, Fischer G, et al. (1992). meningeal gliomatosis with oligodendroglioma. grade gliomas. Cancer Res. 55(9):1941-5.
H, Kohno Y, Yamada M, et al. (2005). Detecting Neurofibromatosis type 2 appears to be a ge- Pathology. 31(1):59-63. PMID:10212927 PMID:7728764
tissue-specific alternative splicing and disea- netically homogeneous disease. Am J Hum 1776. Ng TH, Fung CF, Ma LT (1990). The pa- 1790. Nishimoto T, Kaya B (2012). Cerebel-
se-associated aberrant splicing of the PTCH Genet. 51 (3):486-96. PMID:1496982 thological spectrum of desmoplastic infantile lar liponeurocytoma. Arch Pathol Lab Med.
gene with exon junction microarrays. Hum Mol 1758. National Institutes of Health Consensus gangliogliomas. Histopathology. 16(3):235-41. 136(8):965-9. PMID:228497 4 7
Genet. 14(22):3379-88. PMID:16203740 Development Conference (1988). Neurofibro- PMID:2332209 1791. Nishio S, Morioka T, lnamura T, Takes-
1740. Nagao K, Toyoda M, Takeuchi-Inoue K, matosis. Conference statement. Arch Neurol. 1777. Ngeow J, Mester J, Rybicki LA. Ni Y, Milas hita I, Fukui M, Sasaki M, et al. (1998). Radi-
Fujii K, Yamada M, Miyashita T (2005). Identifi- 45(5):575-8. PMtD:3128965 M, Eng C (2011 ). Incidence and clinical charac- ation-induced brain tumours: potential late
cation and characterization of multiple isofonms 1759. Nauen D, Haley L, Lin MT, Perry A, Gian- teristics of thyroid cancer in prospective series complications of radiation therapy for brain tu-
of a murine and human tumor suppressor, nini C, Burger PC, et al. (2015). Molecular Ana- of individuals with Cowden and Cowden-like mours. Acta Neurochir (Wien). 140(8):763-70.
patched, having distinct first exons. Genomics. lysis of Pediatric Oligodendrogliomas Highlights syndrome characterized by germline PTEN, PMID:9810442
85(4):462-71. PMID:15780749 Genetic Differences with Adult Counterparts SDH, or KLLN alteralions. J Clin Endocrinol 1792. Nishio S, Takeshita I, Kaneko Y, Fukui M
1741. Nagashima T, Hoshino T, Cho KG (1987). and Other Pediatric Gliomas. Brain Pathol. Metab. 96(12):E2063-71. PMID:21956414 (1992). Cerebral neurocytoma. A new subset of
Proliferative potential of vascular components PMID:26206478 1778. Nguyen R, Dombi E, Akshintala S, Bald· benign neuronal tumors of the cerebrum. Can-
in human glioblastoma multiforme. Acta Neuro- 1760. Nayak L, Abrey LE, Iwamoto FM win A, Widemann BC (2015). Characterization cer. 70(2):529-37. PMID:1617603
pathol. 73(3):301-5. PMID:3039783 (2009). lntracranial dural metastases. Cancer. of spinal findings in children and adults with 1793. Nitta H, Hayase H, Moriyama Y, Ya-
1742. Naggara 0, Varlet P, Page P, Op- 115(9):1947-53. PMID:19241421 neurofibromatosis type 1 enrolled in a natu- mashima T, Yamashita J (1993). Gliosarcoma
penheim C, Meder JF (2005). Suprasellar 1761. Neal MT, Ellis TL, Stanton CA (2012). ral history study using magnetic resonance of the posterior cranial Iossa: MRI findings.

References 383
Neuroradiology. 35(4):279-80. PMID:8492894 49(4):243-54. PMID:24053982 oligodendroglial gliomas. J Neuropathol Exp hypotheses on the etiology of cancer. IARC Sci
1794. Noble M, Wren D, Wolswijk G (1992). The 1810. Noushmehr H, Weisenberger DJ, Diefes Neurol. 64(6):479-89. PMID:15977639 Publ. 157:247-70. PMID:15055300
0-2A(adult) progenitor cell: a glial stem cell of K, Phillips HS, Pujara K, Berman BP, et al.; Can- 1827. Ohgaki H, Kleihues P (2007). Genetic pa- 1844. Olschwang S, Serova-Sinilnikova OM
the adult central nervous system. Semin Cell cer Genome Atlas Research Network (2010). thways lo primary and secondary glioblasloma. Lenoir GM, Thomas G (1998). PTEN germ-lin�
Biol. 3(6):413-22. PMID:1489973 Identification of a CpG island methylator pheno- Am J Pathol. 170(5):1445-53. PMID:17456751 mutations in juvenile polyposis coli. Nat Genet.
1795. Nobusawa S, Hirato J, Kurihara H, Oga- type that defines a distinct subgroup of glioma. 1828. Ohgaki H, Kleihues P (2009). Genetic 18(1):12-4. PMID9425889
wa A, Okura N, Nagaishi M, et al. (2014). ln- Cancer Cell. 17(5):510-22. PMID:20399149 alterations and signaling pathways in the evo- 1845. Olson JD, Riedel E, DeAngelis LM
tratumoral heterogeneity of genomic imbalance 1811. Numoto RT (1994). Pineal parenchy- lution of gliomas. Cancer Sci. 100(12):2235-41. (2000). Long-term outcome of low-grade oli-
in a case of epithelioid glioblastoma with BRAF mal tumors: cell differentiation and prognosis. PMID:19737147 godendroglioma and mixed glioma. Neurology:
V600E mutation. Brain Palhol. 24(3):239-46. J Cancer Res Clin Oncol. 120(11):683-90. 1829. Ohgaki H, Kleihues P (2011). Genetic 54(7):1442-8. PMID:10751254
PMID:24354918 PMID:7525594 profile of astrocylic and oligodendroglial gli- 1846. Olson JM, Breslow NE, Barce J (1993),
1796. Nobusawa S, Orimo K, Horiguchi K, /kola 1812. Nutt CL, Mani DR, Betensky RA, Ta- omas. Brain Tumor Pathol. 28(3):177-83. Cancer in twins of Wilms tumor patients. Am J
H, Yokoo H, Hirato J, el al. (2014). Embryonal mayo P, Cairncross JG, Ladd C, et al. (2003). PMID:21442241 Med Genet. 47(1):91-4. PMID:8396323
tumor with abundant neuropil and true rosettes Gene expression-based classification of ma- 1830. Ohgaki H, Kleihues P (2013). The definiti- 1847. Onda K, Davis RL, Wilson CB, Hoshino T
with only one structure suggestive of an epen- lignant gliomas correlates better with survival on of primary and secondary glioblastoma. Clin (1994). Regional differences in bromodeoxyu-
dymoblastic rosette. Pathol Int. 64(9):472-7. than histological classification. Cancer Res. Cancer Res. 19(4):764-72. PMID:23209033 ridine uptake, expression of Ki-67 protein, and
PMID:25186165 63(7):1602-7. PMID:12670911 1831. Ojha BK, Sharma MC, Rastogi M, nucleolar organizer region counts in glioblasto-
1797. Nobusawa S, Watanabe T, Kleihues P, 1813. O'Malley S, Weitman D, Olding M, Sek- Chandra A, Husain M, Husain N (2007). Dum- ma multiforme. Acta Neuropalhol. 87(6):586--
Ohgaki H (2009). IDH1 mutations as molecular har L (1997). Multiple neoplasms following bbell-shaped paraganglioma of the cervical 93. PMID:8091951
signature and predictive factor of secondary gli- craniospinal irradiation for medulloblastoma in spine in a child. Pediatr Neurosurg. 43(1):60-4. 1848. Onguru 0, Deveci S, Sirin S, Timurkay-
oblaslomas. Clin Cancer Res. 15(19):6002-7. a patient with nevoid basal cell carcinoma syn- PMID:17190992 nak E, Gunhan O (2003). Dysembryoplastic
PMID19755387 drome. Case report. J Neurosurg. 86(2):286-8. 1832. Okada M, Yano H, Hirose Y, Nakayama neuroepithelial tumor in the left lateral vent-
1798. Nobusawa S, Yokoo H, Hiralo J, KakitaA, PMID:9010431 N, Ohe N, Shinoda J, el al. (2011). Olig2 is use- ricle. Minim Invasive Neurosurg. 46(5):306--9.
Takahashi H, Sugino T, el al. (2012). Analysis 1814. O'Marcaigh AS, Ledger GA, Roche PC, ful in the differential diagnosis of oligodendrogli- PMID:14628248
of chromosome 19q13.42 amplification in em- Parisi JE, Zimmerman D (1995). Aromatase omas and extraventricular neurocylomas. Brain 1849. Onilude OE, Lusher ME, Lindsey JC,
bryonal brain tumors with ependymoblastic mul- expression in human germinomas with possib- Tumor Pathol. 28(2):157-61. PMID:21312066 Pearson AD, Ellison DW, Clifford SC (2006).
tilayered rosettes. Brain Pathol. 22(5):689-97. le biological effects. J Clin Endocrinol Metab. 1833. Okada Y, Nishikawa R, Matsutani M, Lou- APC and CTNNB1 mutations are rare in spo-
PMID:22324795 80(12):3763-6. PMID:8530631 is DN (2002). Hypomethylaled X chromosome radic ependymomas. Cancer Genet Cytogenet:
1799. Noell S, Beschorner R, Bisdas S, Beyer 1815. Oderich GS, Sullivan TM, Bower TC, gain and rare isochromosome 12p in diverse in- 168(2) 158-61. PMID 16843107
U, Weber RG, Fallier-Becker P, et al. (2014). Si- Gloviczki P, Miller DV, Babovic-Vuksanovic D, tracranial germ cell tumors. J Neuropathol Exp 1850. Oosterhuis JW, Stoop H, Honecker F,
multaneous subependymomas in monozygotic et al. (2007). Vascular abnormalities in patients Neurol. 61(6):531-8. PMID:12071636 Looijenga LH (2007). Why human extraqo-
female twins: further evidence for a common with neurofibromatosis syndrome type I: clinical 1834. Okamoto Y, Di Patre PL, Burkhard C, nadal germ cell tumours occur in the midline
genetic or developmental disorder background. spectrum, management, and results. J Vase Horstmann S, Jourde B, Fahey M, et al. (2004). of the body: old concepts, new perspectives.
J Neurosurg. 121(3):570-5. PMID:24655099 Surg. 46(3):475-84. PMID:17681709 Population-based study on incidence, survival Int J Androl. 30(4):256-63, discussion 263-4.
1800. Nolan MA, Sakuta R, Chuang N, Otsubo 1816. Ogiwara H, Dubner S, Shafizadeh S, rates, and genetic alterations of low-grade diffu- PMID:17705807
H, Rutka JT, Snead QC 3rd, et al. (2004). Raizer J, Chandler JP (2011 ). Spindle cell on- se astrocylomas and oligodendrogliomas. Acta 1851. Orloff MS, Eng C (2008). Genetic an_d
Dysembryoplastic neuroepithelial tumors cocytoma of the pituitary and pituicytoma: Two Neuropathol. 108(1):49-56. PMID:15118874 phenotypic heterogeneity in the PTEN hamarto-
in childhood: long-term outcome and prog- tumors mimicking pituitary adenoma. Surg Neu- 1835. Okita Y, Narita Y, Miyakita Y, Ohno M, ma tumour syndrome. Oncogene. 27(41):5387-
nostic features. Neurology. 62(12):2270-6. rol Int. 2:116. PMID:21886889 Matsushita Y, Fukushima S, et al. (2012). 97. PMID:18794875
PMID:15210893 1817. Ogura R, Aoki H, Natsumeda M, Shi- IDH1/2 mutation is a prognostic marker for sur- 1852. Orloff MS, He X, Peterson C, Chen F,
1801. Nonoguchi N, Ohta T, Oh JE, Kim YH, mizu H, Kobayashi T, Saito T, et al. (2013). vival and predicts response to chemotherapy Chen JL, Mester JL, et al. (2013). Germline
Kleihues P, Ohgaki H (2013). TERT promoter Epstein-Barr virus-associated primary cen- for grade II gliomas concomitantly treated with PIK3CA and AKT1 mutations in Cowden and
mutations in primary and secondary gliob- tral nervous system cytotoxic T-cell lym- radiation therapy. lnl J Oncol. 41(4):1325-36. Cowden-like syndromes. Am J Hum Genet.
lastomas. Acta Neuropathol. 126(6):931-7. phoma. Neuropathology. 33(4):436-41. PMID:22825915 92(1):76--80. PMID:23246288
PMID:23955565 PMID:23279449 1836. Olar A, Wani KM, Alfaro-Munoz KD, He- 1853. Ortega A, Nuno M, Walia S, Mukherjee D,
1802. Nora FE, Scheithauer BW (1996). Pri- 1818. Oh D, Prayson RA (1999). Evaluation athcock LE, van Thuijl HF, Gilbert MR, et al. Black KL, Patil CG (2014). Treatment and survi-
mary epithelioid hemangioendothelioma of of epithelial and keratin markers in gliob- (2015). IDH mutation status and role of WHO val of patients harboring histological variants of
the brain. Am J Surg Pathol. 20(6):707-14. lastoma multiforme: an immunohistochemical grade and mitotic index in overall survival in glioblastoma. J Clin Neurosci. 21(10):1709-13.
PMID:8651350 study. Arch Pathol Lab Med. 123(10):917-20. grade 11-111 diffuse gliomas. Acta Neuropathol. PMID:24980627
1803. Norman MG, Harrison KJ, Poskitt KJ, Ka- PMID:10506444 129(4):585-96. PMID:25701198 1854. Ortega-Aznar A, Romero-Vidal FJ, de
lousek DK (1995). Duplication of 9P and hyper- 1819. Oh JE, Ohta T, Nonoguchi N, Salomi K, 1837. Olar A, Wani KM, Sulman EP, Mansouri la Torre J, Castellvi J, Nogues P (2001). Neo-
plasia of the choroid plexus: a pathologic, radio- Capper D, Pierscianek D, et al. (2015). Genetic A, Zadeh G, Wilson CD, et al. (2015). Mitotic natal tumors of the CNS: a report of 9 cases
logic, and molecular cytogenetics study. Pediatr alterations in gliosarcoma and giant cell gliob- Index is an Independent Predictor of Recurren- and a review. Clin Neuropathol. 20(5):181-9.
Pathol Lab Med. 15(1):109-20. PMID:8736601 lastoma. Brain Pathol. PMID:26443480 ce-Free Survival in Meningioma. Brain Pathol. PMID:11594502
1804. Northcott PA, Jones DT, Kool M, Ro- 1820. Oh T, Rutkowski MJ, Safaee M, Sun 25(3):266-75 PMID:25040885 1855. Oruckaptan HH, Berker M, Soylemezoglu
binson GW, Gilbertson RJ, Cho Y J, et al. MZ, Sayegh ET, Bloch 0, et al. (2014). Sur- 1838. Oliveira AM, Scheilhauer BW, Salomao F, Ozcan OE (2001). Parafalcine chondrosar-
(2012). Medulloblastomics: the end of the vival outcomes of giant cell glioblastoma: DR, Parisi JE, Burger PC, Nascimento AG coma: an unusual localization for a classical
beginning. Nat Rev Cancer. 12(12):818-34. institutional experience in the management of (2002). Primary sarcomas of the brain and spi- variant. Case report and review of the literature.
PMID:23175120 20 patients. J Clin Neurosci. 21(12):2129-34. nal cord: a study of 18 cases. Am J Surg Pathol. Surg Neurol. 55(3):174-9. PMID:11311919
1805. Northcott PA, Korshunov A, Witt H, Hiel- PMID:25037316 26(8):1056--63. PMID:12170093 1856. Osawa T, Tosaka M, Nagaishi M, Yos-
scher T, Eberhart CG, Mack S, et al. (2011). 1821. Ohba Y, Mochizuki N, Yamashita S, Chan 1839. Oliver TG, Grasfeder LL, Carroll AL, himoto Y (2013). Factors affecting peritumoral
Medulloblastoma comprises four distinct mo- AM, Schrader JW, Hattori S, et al. (2000). Re- Kaiser C, Gillingham CL, Lin SM, et al. (2003). brain edema in meningioma: special histologi-
lecular variants. J Clin Oncol. 29(11):1408-14 gulatory proteins of R-Ras, TC21/R-Ras2, and Transcriptional profiling of the Sonic hedgehog cal subtypes with prominently extensive edema.
PMID:20823417 M-Ras/R-Ras3. J Biol Chem. 275(26):20020-6. response: a critical role for N-myc in proliferati- J Neurooncol. 111(1):49-57. PMID:23104516
1806. Northcott PA, Lee C, ZichnerT, Stutz AM, PMID:10777492 on of neuronal precursors. Proc Natl Acad Sci U 1857. Osborn AG, et al. (2004). Diagnostic Ima-
Erkek S, Kawauchi D, et al. (2014). Enhancer 1822. Ohgaki H, Burger P, Kleihues P (2014). SA. 100(12):7331-6. PMID:12777630 ging Brain. first Salt Lake City. Utah: Amirsys.
hijacking activates GFl1 family oncogenes in Definition of primary and secondary glioblasto- 1840. Oliver TG, Read TA, Kessler JD, Meh- 1858. Osorio JA, Hervey-Jumper SL, Walsh
medulloblastoma. Nature. 511(7510):428-34. ma-response. Clin Cancer Res. 20(7):2013. meti A, Wells JF, Huynh TT, el al. (2005). Loss KM, Clarke JL, Butowski NA, Prados MD, et
PMID:2504304 7 PMID:24557936 of patched and disruption of granule cell de- al. (2015). Familial gliomas: cases in two pairs
1807. Northcott PA, Shih DJ, Peacock J, Gar- 1823. Ohgaki H, Dessen P, Jourde B, Horst- velopment in a pre-neoplastic stage of medul- of brothers. J Neurooncol. 121(1):135-40.
zia L, Morrissy AS, Zichner T, et al. (2012). mann S, Nishikawa T, Di Patre PL, et al. (2004). loblastoma. Development. 132(10):2425-39. PMID:25208478
Subgroup-specific structural variation across Genetic pathways to glioblastoma: a populati- PMID:15843415 1859. Ostendorf AP, Gutmann DH, Weisenberg
1,000 medulloblastoma genomes. Nature. on-based study. Cancer Res. 64(19):6892-9. 1841. Olivier M, Goldgar DE, Sodha N, Oh- JL (2013). Epilepsy in individuals with neurofi-
488(7409):49-56. PMID:22832581 PMID:15466178 gaki H, Kleihues P, Hainaut P, el al. (2003). bromatosis type 1. Epilepsia. 54(10):1810-4.
1808. Northcott PA, Shih DJ, Remke M, Cho Y J, 1824. Ohgaki H, Eibl RH, Schwab M, Rei- Li-Fraumeni and related syndromes: correla- PMID:24032542
Kool M, Hawkins C, el al. (2012). Rapid, reliab- chel MB, Mariani L, Gehring M, et al. (1993). tion between tumor type, family structure, and 1860. Ostertun B, Wolf HK, Campos MG, Matus
le, and reproducible molecular sub-grouping of Mutations of the p53 tumor suppressor gene in TP53 genotype. Cancer Res. 63(20):6643-50. C, Solymosi L, Elger CE, et al. (1996). Dysem-
clinical medulloblastoma samples. Acta Neuro- neoplasms of the human nervous system. Mol PMID:14583457 bryoplaslic neuroepithelial tumors: MR and CT
pathol. 123(4):615-26. PMID:22057785 Carcinog. 8(2):74-80. PMID:8397797 1842. Olivier M, Hollstein M, Hainaut P (2010). evaluation. AJNR Am J Neuroradiol. 17(3):419-
1809. Northrup H, Krueger DA; International 1825. Ohgaki H, Kleihues P (2005). Epidemio- TP53 mutations in human cancers: origins, con- 30. PMID:8881234
Tuberous Sclerosis Complex Consensus Group logy and etiology of gliomas. Acta Neuropathol. sequences, and clinical use. Cold Spring Harb 1861. Ostrom QT, Bauchet L, Davis FG, De-
(2013). Tuberous sclerosis complex diagno- 109(1):93-108. PMID:15685439 Perspect Biol. 2(1):a001008. PMID:20182602 ltour I, Fisher JL, Langer CE, et al. (2014). The
stic criteria update: recommendations of the 1826. Ohgaki H, Kleihues P (2005). Popu- 1843. Olivier M, Hussain SP, Caron de Fro- epidemiology of glioma in adults: a "state of the
2012 !international Tuberous Sclerosis Com- lation-based studies on incidence, survival mentel C, Hainaut P, Harris CC (2004). TP53 science" review. Neuro Oncol. 16(7):896--913.
plex Consensus Conference. Pediatr Neurol. rates, and genetic alterations in astrocytic and mutation spectra and load: a tool for generating PMID:24842956

384 References
1862. Ostrom QT, de Blank PM, Kruchko C, J, Benelli M, Barbetti L, et al. (2015). Expanding 63 affected individuals and clinical evidence for treatment strategies for central neurocytoma.
Petersen CM, Liao P, Finlay JL, et al. (2015). the mutational spectrum of LZTR1 in schwan- heterogeneity. Am J Med Genet. 52(4):450-61. J Clin Neurosci. 20(9):1193-9. PMID:23810386
Alex's Lemonade Stand Foundation Infant and nomatosis. Eur J Hum Genet. 23(7):963-8 PMID:7747758 1908. Patel N, Fallah A, Provias J, Jha NK
Childhood Primary Brain and Central Nervous PMID:25335493 1894. Parsa CF, Hoyt CS, Lesser RL, Wein- (2009). Cerebellar liponeurocytoma. Can J
System Tumors Diagnosed in the United States 1878. Pagni CA, Giordana MT, Canavero S stein JM, Strother CM, Muci-Mendoza R, et al. Surg. 52(4):E117-9 PMID:19680499
in 2007-2011. Neuro Oneel. 16 Suppl 10:x1- (1991). Benign recurrence of a pilocytic cere- (2001 ). Spontaneous regression of optic glio- 1909. Patil S, Perry A, Maccollin M, Dong S,
36. PMID:25542864 bellar astrocytoma 36 years after radical remo- mas: thirteen cases documented by serial neu- Betensky RA, Yeh TH, et al. (2008). lmmunohis-
1862A. Ostrom QT, Gittleman H, Farah P, val: case report. Neurosurgery. 28(4):606-9. roimaging. Arch Ophthalmol. 119(4):516-29. tochemical analysis supports a role for IN11/SM-
Ondracek A, Chen Y, Wolinsky Y, et al. (2013). PMID:2034360 PMID:11296017 ARCB1 in hereditary forms of schwannomas,
CBTRUS statistical report: Primary brain and 1879. Pagon RA, Adam MP, Ardinger HH, et 1895. Parsons DW, Jones S, Zhang X, Lin JC, but not in solitary, sporadic schwannomas.
central nervous system tumors diagnosed in al. (1993-2015).GeneReviews® [Internet). Leary RJ, Angenendt P, et al. (2008). An inte- Brain Pathol. 18(4):517-9 PMID:18422762
the United States in 2006-2010. Neuro Oneel. [Updated 2014 Mar 27). Seattle: University of grated genomic analysis of human glioblasto- 1910. Patil S, Scheithauer BW, Strom RG, Maf-
15 Suppl 2:ii1-56. PMID:24137015 Washington. ma multiforme. Science. 321(5897):1807-12. ra M, Chicoine MR, Perry A (2011). Malignant
1863. Ostrom QT, Gittleman H, Liao P, Rouse 1880. Pajtler KW, Witt H, Sill M, Jones DT, Ho- PMID:18772396 meningiomas with epithelial (adenocarcino-
C, Chen Y, Dowling J, et al. (2014). CBTRUS vestadt V, Kratochwil F, et al. (2015). Molecular 1896. Partap S, Curran EK, Propp JM, Le ma-like) metaplasia: a study of 3 cases. Neuro-
statistical report: primary brain and central ner- Classification of Ependymal Tumors across All GM, Sainani KL, Fisher PG (2009). Medullob- surgery. 69(4):884-92. PMID:21558975
vous system tumors diagnosed in the United CNS Compartments, Histopathological Grades, lastoma incidence has not changed over time: 1911. Patsalides AD, Atac G, Hedge U, Janik
States in 2007-2011. Neuro Oneel. 16 Suppl and Age Groups. Cancer Cell. 27(5):728-43. a CBTRUS study. J Pediatr Hematol Oneel. J, Grant N, Jaffe ES, et al. (2005). Lymphoma-
4:iv1-63. PMID:25304271 PMID:25965575 31(12):970-1. PMID:19887963 toid granulomatosis: abnormalities of the brain
1864. Ota S, Crabbe DC, Tran TN, Triche TJ, 1881. Pakos EE, Goussia AC, Zina VP, Pitouli 1897. Pascual-Castroviejo I, Pascual-Pascual at MR imaging. Radiology. 237(1 ):265-73.
Shimada H (1993). Malignant rhabdoid tumor. EJ, Tsekeris PG (2005). Multi-focal gliosarco- SI, Viano J, Velazquez-Fragua R, Lopez-Gu· PMID:16100084
A study with two established cell lines. Cancer. ma: a case report and review of the literature. J tierrez JC (2012). Bilateral spinal neurofibromas 1912. Paulli M, Bergamaschi G, Tenon L, Viglio
71(9):2862-72. PMID:8385567 Neurooncol. 74(3):301-4. PMID:16086111 in patients with neurofibromatosis 1. Brain Dev. A, Rosso R, Facchetti F, et al. (1995). Evidence
1865. Otero JJ, Rowitch D, Vandenberg S 1882. Palma L, Russo A, Celli P (1984). Pro- 34(7) 563--9. PMID:21999966 for a polyclonal nature of the cell infiltrate in si-
(2011). OLIG2 is differentially expressed in pe- gnosis of the so-called "diffuse" cerebellar 1898. Pasini B, McWhinney SR, Bei T, Ma- nus histiocytosis with massive lymphadenopa-
diatric astrocytic and in ependymal neoplasms. astrocytoma. Neurosurgery. 15(3):315-7. tyakhina L, Stergiopoulos S, Muchow M, et thy (Rosai-Dorfman disease). Br J Haematol.
J Neurooncol. 104(2):423--38. PMID:21193945 PMID:6483145 al. (2008). Clinical and molecular genetics of 91 (2):415-8. PMID:854 7085
1866. Ouladan S, Trautmann M, Orouji E, Hart- 1883. Palmero El, Achatz Ml, Ashton-Prolla patients with the Carney-Stratakis syndrome 1913. Paulus W, Bayas A, Ott G, Roggendorf W
mann W, Huss S, Buttner R, et al. (2015). Dif- P, Olivier M, Hainaut P (2010). Tumor protein and germline mutations of the genes coding for (1994). lnterphase cytogenetics of glioblastoma
ferential diagnosis of solitary fibrous tumors: A 53 mutations and inherited cancer: beyond the succinate dehydrogenase subunits SDHB, and gliosarcoma. Acta Neuropathol. 88(5):420-
study of 454 soft tissue tumors indicating the di- Li-Fraumeni syndrome. Curr Opin Oneel. SDHC, and SDHD. Eur J Hum Genet. 16(1):79- 5. PMID:7847070
agnostic value of nuclear STAT6 relocation and 22(1):64-9. PMID:19952748 88. PMID 17667967 1914. Paulus W, Honegger J, Keyvani K, Fahl-
ALDH1 expression combined with in situ pro- 1884. Panageas KS, Reiner AS, Iwamoto 1899. Pasmant E, Masliah-Planchon J, Levy busch R (1999). Xanthogranuloma of the sellar
ximity ligation assay. Int J Oneel. 46(6):2595- FM, Cloughesy TF, Aldape KD, Rivera AL, et P, Laurendeau I, Ortonne N, Parfait B, et al. region: a clinicopathological entity different from
605. PMID:25901508 al. (2014). Recursive partitioning analysis of (2011 ). Identification of genes potentially in- adamantinomatous craniopharyngioma. Acta
1867. Owler BK, Makeham JM, Shingde M, prognostic variables in newly diagnosed ana- volved in the increased risk of malignancy in Neuropathol. 97(4):377-82. PMID:10208277
Besser M (2005). Cerebellar liponeurocytoma. plastic oligodendroglial tumors. Neuro Oneel. NF1-microdeleted patients. Mot Med. 17(1- 1915. Paulus W, Jellinger K, Hallas C, Ott G,
J Clin Neurosci. 12(3):326-9. PMID:15851097 16(11):1541-6. PMID:24997140 2):79-87. PMID:20844836 Muller-Hermelink HK (1993). Human herpesvi-
1868. Owonikoko TK, Arbiser J, Zelnak A, Shu 1885. Pantazis G, Harter PN, Capper D, Kohl- 1900. Pasquale G, Maria BA, Vania P, Gas- rus-6 and Epstein-Barr virus genome in primary
HK, Shim H, Robin AM, et al. (2014). Current hof P, Mittelbronn M, Schittenhelm J (2014). tone P, Nicola DL (2009). Cerebellar lipon- cerebral lymphomas. Neurology. 43(8):1591-3.
approaches to the treatment of metastatic brain The embryonic stem cell factor UTF1 serves eurocytoma: an updated follow-up of a case PMID:8394522
tumours. Nat Rev Clin Oneel. 11(4):203--22. as a reliable diagnostic marker for germinomas. presenting histopathological and clinically ag- 1916. Paulus W, Janisch W (1990). Clinicopa-
PMID:24569448 Pathology. 46(3):225-9. PMID:24614704 gressive features. Neural India. 57(2):194-6. thologic correlations in epithelial choroid plexus
1869. Ozawa T, Brennan CW, Wang L, Squatrito 1886. Papanicolau-Sengos A, Wang-Rodriguez PMID:19439854 neoplasms: a study of 52 cases. Acta Neuropa-
M, Sasayama T, Nakada M, et al. (2010). PDG- J, Wang HY, Lee RR, Wong A, Hansen LA, et al. 1901. Pasquier B, Gasnier F, Pasquier D, Ked- thol. 80(6):635-41. PMID:1703384
FRA gene rearrangements are frequent genetic (2012). Rare case of a primary non-dural cent- dari E, Morens A, Couderc P (1986). Papillary 1917. Paulus W, Lisle DK, Tonn JC, Wolf HK,
events in PDGFRA-amplified glioblastomas. ral nervous system low grade B-celt lymphoma meningioma. Clinicopathologic study of seven Roggendorf W, Reeves SA, et al. (1996). Mo-
Genes Dev. 24(19):2205-18. PMID:20889717 and literature review. Int J Clin Exp Pathol. cases and review of the literature. Cancer. lecular genetic alterations in pleomorphic xan-
1870. Ozek MM, Sav A, Pamir MN, Ozer AF, 5(1):89-95. PMID:22295152 58(2):299-305. PMID:3719522 thoastrocytoma. Acta Neuropathol. 91(3):293--
Ozek E, Erzen C (1993). Pleomorphic xan- 1886A. Paraf F, Jothy S, Van Meir EG (1997). 1902. Pasquier B, Pasquier D, N'Golet A, Panh 7. PMID:8834542
thoastrocytoma associated with von Reckling- Brain tumor-polyposis syndrome: two ge- MH, Couderc P (1980). Extraneural metastases 1918. Paulus W, Schlote W, Perentes E, Jacobi
hausen neurofibromatosis. Childs Nerv Syst. netic diseases? J Clin Oneel. 15(7):2744-58. of astrocytomas and glioblastomas: clinicopa- G, Warmuth-Metz M, Roggendorf W (1992).
9(1):39-42. PMID:8481944 PMID:9215849 thological study of two cases and review of li- Desmoplastic supratentorial neuroepithelial
1871. Ozolek JA, Finkelstein SD, Couce ME 1887. Parham DM, Weeks DA, Beckwith JB terature. Cancer. 45(1):112-25. PMID:6985826 tumours of infancy. Histopathology. 21(1):43-9.
(2004). Gliosarcoma with epithelial differen- (1994). The clinicopathologic spectrum of puta- 1903. Pasquier B, Peoc'H M, Fabre-Bocquen- PMID:1634201
tiation: immunohistochemical and molecular tive extrarenal rhabdoid tumors. An analysis tin B, Bensaadi L, Pasquier D, Hoffmann D, et 1919. Paulus W, Slowik F, Jellinger K (1991).
characterization. A case report and review of 42 cases studied with immunohistochemis- al. (2002). Surgical pathology of drug-resistant Primary intracranial sarcomas: histopatho-
of the literature. Mod Pathol. 17(6):739-45. try or electron microscopy. Am J Surg Pathol. partial epilepsy. A 10-year-experience with a logical features of 19 cases. Histopathology.
PMID:15148503 18(10):1010-29. PMID:8092393 series of 327 consecutive resections. Epileptic 18(5):395-402. PMID:1715839
1872. Ozoren N, EI-Deiry WS (2003). Cell sur- 1888. Park CK, Phi JH, Park SH (2015). Glial Disord. 4(2):99-119. PMID:12105073 1920. Pearce MS, Salotti JA, Little MP,
face Death Receptor signaling in normal and tumors with neuronal differentiation. Neurosurg 1904. Pasquier B, Peoc'h M, Morrison AL, Gay McHugh K, Lee C, Kim KP, et al. (2012). Ra-
cancer cells. Semin Cancer Biol. 13(2):135-47. Clin N Am. 26(1):117-38. PMID:25432191 E, Pasquier D, Grand S, et al. (2002). Chordoid diation exposure from CT scans in childhood
PMID:12654257 1889. Park DH, Park YK, Oh JI, Kwon TH, glioma of the third ventricle: a report of two and subsequent risk of leukaemia and brain
1873. Padberg GW, Schol JD, Vielvoye GJ, Chung HS, Cho HD, et al. (2002). Oncocytic new cases, with further evidence supporting tumours: a retrospective cohort study. Lancet.
Bots GT, de Beer FC (1991). Lhermitte-Duclos paraganglioma of the cauda equina in il child. an ependymal differentiation, and review of the 380(9840):499-505. PMID:22681860
disease and Cowden disease: a single Case report and review of the literature. Pediatr literature. Am J Surg Pathol. 26(10):1330-42. 1921. Pedersen M, Kusters-Vandevelde HV,
phakomatosis. Ann Neural. 29(5):517-23. Neurosurg. 36(5):260-5. PMID:12053045 PMID:12360048 Vires A, Greenen PJ, Sanchez-Laorden B,
PMID:1859181 1890. Park JS, Park H, Park S, Kim SJ, Seel 1905. Passone E, Pizzolitto S, D'Agostini S, Gilhuis JH, et al. (2013). Primary melanoma
1874. Padovani L, Colin C, Fernandez C, HJ, Ko YH (2013). Primary central nervous sys- Skrap M, Gardiman MP, Nocerino A, et al. of the CNS in children is driven by congenital
Maues de Paula A, Mercurio S, Scavarda D, tem ALK positive anaplastic large cell lympho- (2006). Non-anaplastic pleomorphic xanthoast- expression of oncogenic NRAS in melanocytes.
et al. (2012). Search for distinctive markers in ma with predominantly leptomeningeal invol- rocytoma with neuroradiological evidences of Cancer Discov. 3(4):458-69. PMID:23303902
DNT and cortical grade II glioma in children: vement in an adult. Yonsei Med J. 54(3):791-6. leptomeningeal dissemination. Childs Nerv 1922. Pekmezci M, Louie J, Gupta N, Bloomer
same clinicopathological and molecular enti- PMID:23549832 Syst. 22(6):614-8. PMID:16369851 MM, Tihan T (2010). Clinicopathological cha-
ties? Curr Top Med Chem. 12(15):1683--92. 1891. Parker M, Mohankumar KM, Punchi- 1905A. Pastorino L, Ghiorzo P, Nasti S, Battis· racteristics of adamantinomatous and papillary
PMID:22978341 hewa C, Weinlich R, Dalton JD, Li Y, et al. tuzzi L, Cusano R, Marzocchi C, et al. (2009). craniopharyngiomas: University of California,
1875. Paek SH, Kim SH, Chang KH, Park CK, (2014). C11orf95-RELA fusions drive oncoge- Identification of a SUFU germline mutation in a San Francisco experience 1985-2005. Neu-
Kim JE, Kim DG, et al. (2005). Microcystic nic NF-xB signalling in ependymoma. Nature. family with Gorlin syndrome. Am J Med Genet rosurgery. 67(5):1341-9, discussion 1349.
meningiomas: radiological characteristics of 16 506(7489):451-5. PMID:24553141 A. 149A(7):1539-43. PMID:19533801 PMID:20871436
cases. Acta Neurochir (Wien). 147(9):965-72, 1892. Parkin DM, Whelan SL, Ferlay J, Teppo 1906. Patel AP, Tirosh I, Trombetta JJ, Shalek 1923. Pekmezci M, Perry A (2013). Neuropa-
discussion 972. PMID:16028111 L, Thomas DB (2003). Cancer Incidence in Five AK, Gillespie SM, Wakimoto H, et al. (2014). thology of brain metastases. Surg Neural Int. 4
1876. Paek SH, Shin HY, Kim JW, Park SH, Son Continents. IARC Scientific Publications. Volu- Single-cell RNA-seq highlights intratumoral Suppl 4:8245-55. PMID:23717796
JH, Kim DG (2010). Primary culture of central me 155. Lyon: IARC Press. heterogeneity in primary glioblastoma. Science. 1924. Pekmezci M, Reuss DE, Hirbe AC, Da-
neurocytoma: a case report. J Korean Med Sci. 1893. Parry DM, Eldridge R, Kaiser-Kupfer Ml, 344(6190):1396-401. PMID:24925914 hiya S, Gutmann DH, van Deimling A, et al.
25(5):798-803. PMID:20436722 Bouzas EA, Pikus A, Patronas N (1994). Neuro- 1907. Patel DM, Schmidt RF, Liu JK (2013). (2015). Morphologic and immunohistochemical
1877. Paganini I, Chang VY, Capone GL, Vitte fibromatosis 2 (NF2): clinical characteristics of Update on the diagnosis, pathogenesis, and features of malignant peripheral nerve sheath

References 385
tumors and cellular schwannomas. Mod Pathol. rhabdoid meningiomas. Mod Pathol. 18(7):951- in patients with neurofibromatosis type I. J histopathological, and molecular characteri-
28(2): 187-200. PMID:25189642 8. PMID:15761491 Pediatr Hematol Oneal. 33(5):e198-201. stics of medulloblastomas in the prospective
1925. Pels H, Montesinos-Rongen M, Schal- 1942. Perry A, Giannini C, Raghavan R, Scheit- PMID:21572348 HIT2000 multicenter clinical trial cohort. Acta
ler C, Schlegel U, Schmidt-Wolf IG, Wiestler hauer BW, Banerjee R, Margraf L, et al. (2001). 1958. Petitjean A, Mathe E, Kato S, lshioka C, Neuropathol. 128(1):137-49. PMID:24791927
OD, et al. (2005). VH gene analysis of primary Aggressive phenotypic and genotypic features Tavtigian SV, Hainaut P, et al. (2007). Impact 1973. Pietsch T, Waha A, Koch A, Kraus J, Alb-
CNS lymphomas. J Neurol Sci. 228(2):143--7. in pediatric and NF2-associated meningio- of mutant p53 functional properties on TP53 recht S, Tonn J, et al. (1997). Medulloblastomas
PMID:15694195 mas: a clinicopathologic study of 53 cases. J mutation patterns and tumor phenotype: les- of the desmoplastic variant carry mutations of
1926. Pels H, Schlegel U (2006). Primary Neuropathol Exp Neurol. 60(10):994-1003. sons from recent developments in the IARC the human homologue of Drosophila patched.
central nervous system lymphoma. Curr Treat PMID:11589430 TP53 database. Hum Mutat. 28(6):622-9. Cancer Res. 57(11):2085-8. PMID:9187099
Options Neurol. 8(4):346-57. PMID:16942677 1943. Perry A, Giannini C, Scheithauer BW, PMID:17311302 1974. Pietsch T, Wohlers I, Goschzik T, Dre-
1927. Pencalet P, Maixner W, Sainte-Rose C, Rojiani AM, Yachnis AT, Seo IS, et al. (1997). 1959. Peyre M, Bah A, Kalamarides M (2012). schmann V, Denkhaus D, Dorner E, et al.
Lellouch-Tubiana A, Cinalli G, Zerah M, et al. Composite pleomorphic xanthoastrocytoma Multifocal choroid plexus papillomas: case (2014). Supratentorial ependymomas of child-
(1999). Benign cerebellar astrocytomas in child- and ganglioglioma: report of four cases and report. Acta Neurochir (Wien). 154(2):295-9. hood carry C11orf95-RELA fusions leading to
ren. J Neurosurg. 90(2):265-73. PMID:9950497 review of the literature. Am J Surg Pathol. PMID:21953479 pathological activation of the NF-KB signaling
1928. Per H, Kontas 0, Kumandas S, Kurtsoy A 21(7):763-71. PMID:9236832 1960. Peyre M, Stemmer-Rachamimov A, pathway. Acta Neuropathol. 127(4):609-11.
(2009). A report of a desmoplastic non-infantile 1944. Perry A, Kurtkaya-Yapicier 0, Scheit- Clermont-Taranchon E, Quentin S, EI-Taraya PMID:24562983
ganglioglioma in a 6-year-old boy with review hauer BW, Robinson S, Prayson RA, Klein- N, Walczak C, et al. (2013). Meningioma pro- 1975. Pignatli F, van den Bent M, Curran D, De-
of the literature. Neurosurg Rev. 32(3):369-74, schmidt-DeMasters BK, et al. (2005). Insights gression in mice triggered by Nf2 and Cdkn2ab bruyne C, Sylvester R, Therasse P, et al.; Euro-
discussion 374. PMID:19280238 into meningioangiomatosis with and without inactivation. Oncogene. 32(36):4264-72. pean Organization for Research and Treatment
1929. Peraud A, Ansari H, Bise K, Reulen HJ meningioma: a clinicopathologic and genetic PMID:23045274 of Cancer Brain Tumor Cooperative Group;
(1998). Clinical outcome of supratentorial ast- series of 24 cases with review of the literature. 1961. Pfister S, Janzarik WG, Remke M, Ernst European Organization for Research and Tre-
rocytoma WHO grade II. Acta Neurochir (Wien). Brain Pathol. 15(1):55-65. PMID:15779237 A, Werft W, Becker N, et al. (2008). BRAF gene atment of Cancer Radiotherapy Cooperative
140(12):1213-22. PMID:9932120 1945. Perry A, Lusis EA, Gutmann DH (2005). duplication constitutes a mechanism of MAPK Group (2002). Prognostic factors for survival in
1930. Peraud A, Kreth FW, Wiestler OD, Klei- Meningothelial hyperplasia: a detailed clinico- pathway activation in low-grade astrocytomas. adult patients with cerebral low-grade glioma.
hues P, Reulen HJ (2002). Prognostic impact of pathologic, immunohistochemical and genetic J Clin Invest. 118(5):1739-49. PMID:18398503 J Clin Oneal. 20(8):2076-84. PMID:11956268
TP53 mutations and P53 protein overexpres- study of 11 cases. Brain Pathol. 15(2):109-15. 1962. Pfister S, Remke M, Castaldi M, Bai AH, 1976. Pimentel J, Barroso C, Miguens J, Firmo
sion in supratentorial WHO grade II astrocyto- PMID:15912882 Muckenthaler MU, Kulozik A, et al. (2009). C, Antunes JL (2009). Papillary glioneuronal tu-
mas and oligoastrocytomas. Clin Cancer Res. 1946. Perry A, Miller CR, Gujrati M, Scheithau- Novel genomic amplification targeting the mor-prognostic value of the extension of surgi-
8(5):1117-24. PMID:12006527 er BW, Zambrano SC, Jost SC, et al. (2009). microRNA cluster at 19q13.42 in a pediatric cal resection. Clin Neuropathol. 28(4):287-94.
1931. Peraud A, Watanabe K, Plate KH, Yone- Malignant gliomas with primitive neuroectoder- embryonal tumor with abundant neuropil and PMID:19642508
kawa Y, Kleihues P. Ohgaki H (1997). p53 muta- mal tumor-like components: a clinicopathologic true rosettes. Acta Neuropathol. 117(4):457-64. 1977. Pimentel J, Silva R, Pimentel T (2003).
tions versus EGF receptor expression in giant and genetic study of 53 cases. Brain Pathol. PMID:19057917 Primary malignant rhabdoid tumors of the cen-
cell glioblastomas. J Neuropathol Exp Neurol. 19(1):81-90. PMID:18452568 1963. Phi JH, Koh EJ, Kim SK, Park SH, Cho tral nervous system: considerations about two
56(11):123641. PMID:9370234 1947. Perry A, Roth KA, Banerjee R, Fuller BK, Wang KC (2011). Desmoplastic infantile as- cases of adulthood presentation. J Neurooncol.
1932. Peraud A, Watanabe K, Schwechheimer CE, Gutmann DH (2001 ). NF1 deletions in trocytoma: recurrence with malignant transfor- 61(2):121-6. PMID:12622450
K, Yonekawa Y, Kleihues P, Ohgaki H (1999). S-100 protein-positive and negative cells of mation into glioblastoma: a case report. Childs 1978. Pinna V, Lanari V, Daniele P, Consoli
Genetic profile of the giant cell glioblastoma. sporadic and neurofibromatosis 1 (NF1)-asso- Nerv Syst. 27(12):2177-81. PMID:21947035 F, Agolini E, Margiotti K, et al. (2015). p.Ar-
Lab Invest. 79(2):123-9. PMID:10068201 ciated plexiform neurofibromas and malignant 1964. Phi JH, Park SH, Chae JH, Hong KH, g1809Cys substitution in neurofibromin is asso-
1933. Perentes E, Rubinstein LJ, Herman MM, peripheral nerve sheath tumors. Am J Pathol. Park SS, Kang JH, et al. (2008). Congenital ciated with a distinctive NF1 phenotype without
Donoso LA (1986). S-antigen immunoreactivity 159(1):57-61. PMID:11438454 subependymal giant cell astrocytoma: clinical neurofibromas. Eur J Hum Genet. 23(8):1068-
in human pineal glands and pineal parenchymal 1948. Perry A, Scheithauer BW, Macaulay considerations and expression of radial glial 71. PMID:25370043
tumors. A monoclonal antibody study. Acta Neu- RJ, Raffel C, Roth KA, Kras JM (2002). Oligo- cell markers in giant cells. Childs Nerv Syst. 1979. Pinto Gama HP, da Rocha AJ, Braga FT,
ropathol. 71 (3-4 ):224-7. PMID:3541480 dendrogliomas with neurocytic differentiation. A 24(12):1499-503. PMID:18629509 da Silva CJ, Maia AC Jr, de Campos Meirelles
1934. Perilongo G, Carollo C, Salviati L, Murgia report of 4 cases with diagnostic and histoge- 1965. Phillips CL, Miles L, Jones BV, Sutton RG, et al. (2006). Comparative analysis of MR
A, Pillon M, Basso G, et al. (1997). Diencephalic netic implications. J Neuropathol Exp Neural. M, Crone K, Fouladi M (2011 ). Medulloblasto- sequences to detect structural brain lesions in
syndrome and disseminated juvenile pilocytic 61(11):947-55. PMID:12430711 ma with melanotic differentiation: case report tuberous sclerosis. Pediatr Radiol. 36(2):119-
astrocytomas of the hypothalamic-optic chiasm 1949. Perry A, Scheithauer BW, Nascimento and review of the literature. J Neurooncol. 25. PMID:16283285
region. Cancer. 80(1 ):142-6. PMID:9210720 AG (1997). The immunophenotypic spectrum 103(3):759-64. PMID:20953660 1980. Piotrowski A, Xie J, Liu YF, Poplawski
1935. Perkins SM, Mitra N, Fei W, Shinohara of meningeal hemangiopericytoma: a com- 1966. Phillips JJ, Misra A, Feuerstein BG, AB, Gomes AR, Madanecki P, et al. (2014).
ET (2012). Patterns of care and outcomes of parison with fibrous meningioma and solitary Kunwar S, Tihan T (2010). Pituicytoma: cha- Gerrnline loss-of-function mutations in LZTR1
patients with pleomorphic xanthoaslrocytoma: fibrous tumor of meninges. Am J Surg Pathol. racterization of a unique neoplasm by histolo- predispose to an inherited disorder of multi-
a SEER analysis. J Neurooncol. 110(1):99-104. 21(11):1354-60. PMID:9351573 gy, immunohistochemistry, ultrastructure, and ple schwannomas. Nat Genet. 46(2):182-7.
PMID:22843450 1950. Perry A, Scheithauer BW, Stafford SL, array-based comparative genomic hybridiz- PMID:24362817
1936. Perreault S, Ramaswamy V, Achrol AS, Abell-Aleff PC, Meyer FB (1998). "Rhabdoid" ation. Arch Pathol Lab Med. 134(7):1063-9. 1981. Pirini MG, Mascalchi M, Salvi F, Tassinari
Chao K, Liu TT, Shih D, et al. (2014). MRI sur- meningioma: an aggressive variant. Am J Surg PMID:20586639 CA, Zanella L, Bacchini P, et al. (2003). Primary
rogates for molecular subgroups of medullob- Pathol. 22(12):1482-90. PMID:9850174 1967. Picard D, Miller S, Hawkins CE, Bouffet diffuse meningeal melanomatosis: radiolo-
lastoma. AJNR Am J Neuroradiol. 35(7):1263- 1951. Perry A, Scheithauer BW, Stafford SL, E, Rogers HA, Chan TS, et al. (2012). Markers gic-pathologic correlation. AJNR Am J Neurora-
9. PMID:24831600 Lohse CM, Wollan PC (1999). "Malignancy" in of survival and metastatic potential in childhood diol. 24(1):115-8. PMID:12533338
1937. Perry A, Aldape KO, George DH, Bur- meningiomas: a clinicopathologic study of 116 CNS primitive neuro-ectodermal brain tumours: 1982. Pirotte B, Krischek 8, Levivier M, Bolyn
ger PC (2004). Small cell astrocytoma: an patients, with grading implications. Cancer. an integrative genomic analysis. Lancet Oncol. S, Brucher JM, Brotchi J (1998). Diagnostic
aggressive variant that is clinicopathologically 85(9) 2046-56. PMID:10223247 13(8):838-48. PMID:22691720 and microsurgical presentation of intracra-
and genetically distinct from anaplastic oli- 1952. Perry A, Stafford SL, Scheithauer BW, 1968. Pickuth D, Leutloff U (1996). Computed nial angiolipomas. Case report and review
godendroglioma. Cancer. 101(10):2318-26. Suman VJ, Lohse CM (1997). Meningioma gra- tomography and magnetic resonance ima- of the literature. J Neurosurg. 88(1):129-32.
PMID:15470710 ding: an analysis of histologic parameters. Am ging findings in primitive neuroectodermal PMID:9420085
1938. Perry A, Banerjee R, Lohse CM, Klein- J Surg Pathol. 21(12):1455-65. PMLD:9414189 tumours in adults. Br J Radiol. 69(817):1-5. 1983. Pitche P, Ategbo S, Gbadoe A, Bassu-
schmidt-DeMasters BK, Scheithauer BW 1953. Perry A, Stafford SL, Scheithauer BW. PMID:8785615 ka-Parent A, Mouzou B, TchangaY-Walla K
(2002). A role for chromosome 9p21 deletions in Suman VJ, Lohse CM (1998). The prognostic 1969. Pierallini A, Bonamini M, Pantano P, Pal- (1997). [Bullous toxidermatosis and HIV infecti-
the malignant progression of meningiomas and significance of MIB-1, p53, and DNA flow cyto- meggiani F, Raguso M, Osti MF, et al. (1998). on in hospital environment in Lome (Togo)J. Bull
the prognosis of anaplastic meningiomas. Brain metry in completely resected primary meningi- Radiological assessment of necrosis in gliob- Soc Pathol Exot. 90(3):186-8. PMID:9410257
Pathol. 12(2):183--90. PMID:11958372 omas. Cancer. 82(11):2262-9. PMID:9610708 lastoma: variability and prognostic value. Neu- 1984. Pizer BL, Moss T, Oakhill A, Webb D,
1939. Perry A, Burton SS, Fuller GN, Robinson 1954. Persson Al, Petritsch C, Swartling FJ, lts- roradiology. 40(3):150-3. PMID:9561517 Coakham HB (1995). Congenital astroblasto-
CA, Palmer CA, Resch L, et al. (2010). Oligo- ara M, Sim FJ, Auvergne R, et al. (2010). Non- 1970. Piercecchi-Marti MD. Mohamed H, ma: an immunohistochemical study. Case re-
dendroglial neoplasms with ganglioglioma-like stem cell origin for oligodendroglioma. Cancer Liprandi A, Gambarelli D, Grisoli F, Pellissier port. J Neurosurg. 83(3):550-5. PMID:7545227
maturation: a diagnostic pitlall. Acta Neuropa- Cell. 18(6):669-82. PMID:21156288 JF (2002). lntracranial meningeal melanocyto- 1985. Pizzuti A, Bottillo I, Inzana F, Lanari V,
thol. 120(2):237-52. PMID:20464403 1955. Persu A, Hamoir M, Gregoire V, Garin ma associated with ipsilateral nevus of Ota. Buttarelli F, Torrente I, et al. (2011). Familial spi-
1940. Perry A, Cai DX, Scheithauer BW, Swan- P, Duvivier E, Reychler H, et al. (2008). High Case report. J Neurosurg. 96(3):619-23. nal neurofibromatosis due to a multiexonic NF1
son PE, Lohse CM, Newsham IF, et al. (2000). prevalence of SDHB mutations in head and PMID:11883852 gene deletion. Neurogenetics. 12(3):233-40.
Merlin, DAL-1, and progesterone receptor ex- neck paraganglioma in Belgium. J Hypertens. 1971. Pierron G, Tirode F, Lucchesi C, Reynaud PMID:21365283
pression in clinicopathologic subsets of menin- 26(7):1395-401. PMID:18551016 S, Ballet S, Cohen-Goga S, et al. (2012). A new 1986. Plank TL, Logginidou H, Klein-Szanto A,
gioma: a correlative immunohistochemical 1956. Burger PC (2009). Smears and Frozen subtype of bone sarcoma defined by BCOR- Henske EP (1999). The expression of hamar-
study of 175 cases. J Neuropathol Exp Neurol. Sections in Surgical Neuropathology. Baltimore CCNB3 gene fusion. Nat Genet. 44(4):461-6. tin, the product of the TSC1 gene, in normal
59(10):872-9. PMID:11079777 (MD): PB Medical Publishing. PMID:22387997 human tissues and in TSC1- and TSC2-linked
1941. Perry A, Fuller CE, Judkins AR, Dehner 1957. Peters KB, Cummings T J, Gururangan 1972. Pietsch T. Schmidt R. Remke M, angiomyolipomas. Mod Pathol. 12(5):539-45.
LP, Biegel JA (2005). INl1 expression is re- S (2011). Transformation of juvenile pilocytic Korshunov A, Hovestadt V, Jones DT, et al. PMID:10349994
tained in composite rhabdoid tumors, including astrocytoma to anaplastic pilocytic astrocytoma (2014). Prognostic significance of clinical, 1987. Plank TL, Yeung RS, Henske EP (1998).

386 References
Hamartin, the product of the tuberous sclerosis C, Tinguely M, Jouvet A, Ferreri AJ, et al.; Inter- Pathol. 3(1):11-8. PMID:9990108 2038. Prevot S, Bienvenu L, Vaillant JC, de
1 (TSC1) gene, interacts with tuberin and ap- national Extranodal Lymphoma Study Group 2021. Prayson RA (1999). Composite ganglio- Saint-Maur PP (1999). Benign schwannoma
pears to be localized to cytoplasmic vesicles. (2007). Reactive perivascular T-cell infiltrate glioma and dysembryoplastic neuroepithelial of the digestive tract: a clinicopathologic and
Cancer Res. 58(21):4766--70. PMID:9809973 predicts survival in primary central nervous tumor. Arch Pathol Lab Med. 123(3):247-50. immunohistochemical study of five cases, in-
1988. Plate KH, Breier G, Weich HA, Risau W system B-cell lymphomas. Br J Haematol. PMID:10086515 cluding a case of esophageal tumor. Am J Surg
(1992). Vascular endothelial growth factor is a 138(3):316--23. PMID:17555470 2022. Prayson RA (2000). Papillary glioneuron- Pathol. 23(4):431-6. PMID:10199472
potential tumour angiogenesis factor in human 2005. Ponzoni M, Bonetti F, Poliani PL, Vermi al tumor. Arch Pathol Lab Med. 124(12):1820- 2038A. Prieto-Granada CN, Wiesner T, Mes-
gliomas in vivo. Nature. 359(6398):845-8. W, Bottelli C, Dolcetti R, et al. (2011). Cen- 3. PMID:11100065 sina JL, Jungbluth AA, Chi P, Antonescu CR
PMID:1279432 tral nervous system marginal zone B-cell 2023. Prayson RA (2012). Pleomorphic xan- (2015). Loss of H3K27me3 Expression Is a
1989. Plate KH, Scholz A, Dumont DJ (2012). lymphoma associated with Chlamydophila thoastrocytoma arising in neurofibromato- Highly Sensitive Marker for Sporadic and Ra-
Tumor angiogenesis and anti-angiogenic thera- psittaci infection. Hum Pathol. 42(5):738-42. sis Type 1. Clin Neuropathol. 31(3):152-4. diation-induced MPNST. Am J Surg Pathol.
py in malignant gliomas revisited. Acta Neuro- PMID:21239044 PMID:22551920 IEpub ahead of print] PMID:26645727
pathol. 124(6):763-75. PMID:23143192 2006. Ponzoni M, Ferreri AJ, Campo E, Fac- 2024. Prayson RA, Castilla EA, Hembury TA, 2039. Probst-Cousin S, Bergmann M, Schroder
1990. Plotkin SR, Blakeley JO, Evans DG, Ha- chetti F, Mazzucchelli L, Yoshino T, et al. Liu W, Noga CM, Prok AL (2003). lnterobser- R, Kuchelmeister K, Schmid KW, Ernestus RJ,
nemann CO, Hulsebos T J, Hunter-Schaedle (2007). Definition, diagnosis, and management ver variability in determining MIB-1 labeling in- et al. (1996). Ki-67 and biological behaviour in
K, et al. (2013). Update from the 2011 Inter- of intravascular large B-cell lymphoma: propo- dices in oligodendrogliomas. Ann Diagn Pathol. meningeal haemangiopericytomas. Histopatho-
national Schwannomatosis Workshop: From sals and perspectives from an international con- 7(1):9-13. PMID:12616468 logy. 29(1):57-61. PMID:8818695
genetics to diagnostic criteria. Am J Med Genet sensus meeting. J Clin Oneal. 25(21):3168-73. 2025. Prayson RA, Chahlavi A, Luciano M 2040. Proescholdt MA, Mayer C, Kubitza
A. 161A(3):405-16. PMID:23401320 PMID: 17577023 (2004). Cerebellar paraganglioma. Ann Diagn M, Schubert T, Liao SY, Stanbridge EJ, et al.
1991. Plowman PN, Pizer B, Kingston JE 2007. Popova SN, Bergqvist M, Dimberg A, Pathol. 8(4):219-23. PMID:15290673 (2005). Expression of hypoxia-inducible carbo-
(2004). Pineal parenchymal tumours: II. On Edqvist PH, Ekman S, Hesselager G, et al. 2026. Prayson RA, Estes ML (1992). Dysem- nic anhydrases in brain tumors. Neuro Oneal.
the aggressive behaviour of pineoblastoma in (2014). Subtyping of gliomas of various WHO bryoplastic neuroepithelial tumor. Am J Clin 7(4):465-75. PMID:16212811
patients with an inherited mutation of the RB1 grades by the application of immunohisto- Pathol. 97(3):398-401. PMID:1543164 2041. Pruchon E, Chauveinc L, Sabatier L,
gene. Clin Oneal (R Coll Radial). 16(4):244-7. chemistry. Histopathology. 64(3):365-79. 2027. Prayson RA, Khajavi K, Comair YG Dutrillaux AM, Ricoul M, Delattre JY, et al.
PMID 15214647 PMID:24410805 (1995). Cortical architectural abnormalities (1994). A cytogenetic study of 19 recurrent gli-
1992. Podsypanina K, Ellenson LH, Nemes A, 2008. Poppleton H, Gilbertson RJ (2007). Stem and MIB1 immunoreactivity in gangliogliomas: omas. Cancer Genet Cytogenet. 76(2):85-92.
Gu J, Tamura M, Yamada KM, et al. (1999). cells of ependymoma. Br J Cancer. 96(1):6-10. a study of 60 patients with intracranial tumors. PMID:7923073
Mutation of Pten/Mmac1 in mice causes neo- PMID:17179988 J Neuropathol Exp Neurol. 54(4):513-20. 2042. Pugh T J, Weeraratne SD, Archer TC, Po-
plasia in multiple organ systems. Proc Natl 2009. Poremba C, Dockhorn-Dworniczak PMID:7541447 meranz Krummel DA, Auclair D, Bochicchio J, et
Acad Sci US A. 96(4):1563-8. PMID:9990064 B, Merritt V, Li CY, Heidi G, Tauber PF, et al. 2028. Prayson RA, Suh JH (1999). Subepen- al. (2012). Medulloblastoma exome sequencing
1993. Pohl U, Cairncross JG, Louis ON (1999). (1993). Immature teratomas of different origin dymomas: clinicopathologic study of 14 tumors, uncovers subtype-specific somatic mutations.
Homozygous deletions of the CDKN2C/ carried by a pregnant mother and her fetus. including comparative MIB-1 immunohisto- Nature. 488(7 409):106--10. PMID:22820256
p181NK4C gene on the short arm of chromo- Diagn Mal Pathol. 2(2):131-6. PMID:8269278 chemical analysis with other ependymal neo- 2043. Pulido R, Baker SJ, Barata JT, Carracedo
some 1 in anaplastic oligodendrogliomas. Brain 2010. Portela A, Esteller M (2010). Epigenetic plasms. Arch Pathol Lab Med. 123(4):306--9. A, Cid VJ, Chin-Sang ID, et al. (2014). A uni-
Pathol. 9(4):639-43. PMID:10517502 modifications and human disease. Nat Biotech- PMID:10320142 fied nomenclature and amino acid numbering
1994. Pollack IF, Claassen D, al-Shboul Q, Ja- nol. 28(10):1057-68. PMID:20944598 2029. Preston DL, Ron E, Yonehara S, Kobuke for human PTEN. Sci Signal. 7(332):pe15.
nosky JE, Deutsch M (1995). Low-grade glio- 2011. Portela-Oliveira E, Torres US, Lancellotti T, Fujii H, Kishikawa M, et al. (2002). Tumors of PMID:24985344
mas of the cerebral hemispheres in children: an CL, Souza AS, Ferraz-Filho JR (2014). Solid the nervous system and pituitary gland associa- 2044. Pummi KP, Aho HJ, Laato MK, Pelton en
analysis of 71 cases. J Neurosurg. 82(4):536-- intraventricular papillary glioneuronal tumor: ted with atomic bomb radiation exposure. J Natl JT, Peltonen SA (2006). Tight junction proteins
47. PMID:7897512 magnetic resonance imaging findings with Cancer Inst. 94(20):1555-63. PMID:12381708 and perineurial cells in neurofibromas. J Histo-
1995. Pollack IF, Hoffman HJ, Humphreys RP, histopathological correlation. Pediatr Neurol. 2030. Preuss M, Christiansen H, Merkenschla- chem Cytochem. 54(1):53-61. PMID:16087703
Becker L (1993). The long-term outcome after 50(2):199-200. PMID:24314675 ger A, Hirsch FW, Kiess W, MOiier W, et al. 2045. Purav P, Ganapathy K, Mallikarjuna VS,
surgical treatment of dorsally exophytic brain- 2012. Poulgrain K, Gurgo R, Winter C, Ong (2015). Disseminated oligodendroglial cell-like Annapurneswari S, Kalyanaraman S, Reginald
stem gliomas. J Neurosurg. 78(6):859-63. B, Lau Q (2011). Papillary tumour of the pi- leptomeningeal tumors: preliminary diagnostic J, et al. (2005). Rosai-Dorfman disease of
PMID:8487066 neal region. J Clin Neurosci. 18(8):1007-17. and therapeutic results for a novel tumor entity. the central nervous system. J Clin Neurosci.
1996. Pollack IF, Hurtt M, Pang D, Albright AL PMID:21658955 J Neurooncol. 12(6):656-9. PMID:16099162
(1994). Dissemination of low grade intracranial 2013. Powell SZ, YachnisAT, Rorke LB, Rojiani 2031. Preusser M, Budka H. Rossler K, Hain- 2046. Purdy E, Johnston DL, Bartels U, Fryer
astrocytomas in children. Cancer. 73(11):2869- AM, Eskin TA (1996). Divergent differentiation fellner JA (2007). OLIG2 is a useful immunohis- C, Carrel AS, Crooks B, et al. (2014). Ependy-
78. PMID:8194029 in pleomorphic xanthoastrocytoma. Evidence tochemical marker in differential diagnosis of moma in children under the age of 3 years: a re-
1997. PoltakA, Friede RL(1977). Fine structure for a neuronal element and possible relations- clear cell primary CNS neoplasms. Histopatho- port from the Canadian Pediatric Brain Tumour
of medulloepithelioma. J Neuropathol Exp Neu- hip to ganglion cell tumors. Am J Surg Pathol. logy. 50(3):365-70. PMID:17257132 Consortium. J Neurooncol. 117(2):359-64.
ral. 36(4):712-25 PMID:886367 20(1):80-5. PMID:8540612 2032. Preusser M, Capper D, llhan-Mutlu A, PMID:24532240
1998. Pollock E, Ford-Jones EL, Corey M, 2014. Prabhu VC, Brown HG (2005). The pa- Berghoff AS, Birner P, Bartsch R, et al. (2012). 2047. Qian H, Lin S, Zhang M, Cao Y (2012).
Barker G, Mindorff CM, Gold R, et al. (1991 ). thogenesis of craniopharyngiomas. Childs Nerv Brain metastases: pathobiology and emer- Surgical management of intraventricular central
Use of the Pediatric Risk of Mortality score to Syst. 21(8-9):622-7. PMID:15965669 ging targeted therapies. Acta Neuropathol. neurocytoma: 92 cases. Acta Neurochir (Wien).
predict nosocomial infection in a pediatric in- 2015. Prabowo AS, Iyer AM, Veersema TJ, 123(2):205-22. PMID:22212630 154(11) 1951-60. PMID22941394
tensive care unit. Grit Care Med. 19(2):160-5. Anink JJ, Schouten-van Meeteren AY, Spliet 2033. Preusser M, Dietrich W, Czech T, Pray- 2048. Qin W, Chan JA, Vinters HV, Mathern
PMID:1989753 WG, et al. (2014). BRAF V600E mutation is er D, Budka H. Hainfellner JA (2003). Roset- GW, Franz ON, Taillon BE, et al. (2010). Ana-
1999. Polydorides AD, Rosenblum MK, Edgar associated with m TOR signaling activation in te-forming glioneuronal tumor of the fourth lysis of TSC cortical tubers by deep sequencing
MA (2007). Metastatic renal cell carcinoma to glioneuronal tumors. Brain Pathol. 24(1):52-66. ventricle. Acta Neuropathol. 106(5):506--8. of TSC1, TSC2 and KRAS demonstrates that
hemangioblastoma in von Hippel-lindau di- PMID:23941441 PMID:12915951 small second-hit mutations in these genes are
sease. Arch Pathol Lab Med. 131(4):641-5. 2016. Prabowo AS, van Thuijl HF, Scheinin I, 2034. Preusser M, Hoeftberger R, Woehrer rare events. Brain Pathol. 20(6):1096-105.
PMID:17425399 Sie D, van Essen HF, Iyer AM, et al. (2015). A, Gelpi E, Kouwenhoven M, Kras JM, et PMID:20633017
2000. Pomeroy SL, Tamayo P, Gaasenbeek Landscape of chromosomal copy number aber- al. (2012). Prognostic value of Ki67 index in 2049. Qin Y, Yao L, King EE, Buddavarapu K,
M, Sturla LM, Angelo M, Mclaughlin ME, et rations in gangliogliomas and dysembryoplastic anaplastic oligodendroglial tumours-a trans- Lenci RE, Chocron ES, et al. (2010). Germline
al. (2002). Prediction of central nervous sys- neuroepithelial tumours. Neuropathol Appl Neu- lational study of the European Organization mutations in TMEM127 confer susceptibility to
tem embryonal tumour outcome based on robiol. 41 (6):743-55. PMID:25764012 for Research and Treatment of Cancer Brain pheochromocytoma. Nat Genet. 42(3):229-33.
gene expression. Nature. 415(6870):436-42. 2017. Prajapati HJ, Vincentelli C, Hwang SN, Tumor Group. Histopathology. 60(6):885-94. PMID:20154675
PMID:11807556 Voloschin A, Crocker I, Dehkharghani S (2014). PMID:22335622 2050. Ou M, Olofsson T, Sigurdardottir S, You
2001. Pamper MG, Passe TJ, Burger PC, Primary CNS natural killer/T-cell lymphoma of 2035. Preusser M, Hoischen A, Novak K, Czech C, Kalima H, Nisfer M, et al. (2007). Genetically
Scheithauer BW, Brat DJ (2001 ). Chordoid gli- the nasal type presenting in a woman: case re- T, Prayer D, Hainfellner JA, et al. (2007). Angio- distinct astrocytic and oligodendroglial compo-
oma: a neoplasm unique to the hypothalamus port and review of the literature. J Clin Oneel. centric glioma: report of clinico-palhologic and nents in oligoastrocytomas. Acta Neuropathol.
and anterior third ventricle. AJNR Am J Neuro- 32(8):e26--9. PMID:24419127 genetic findings in 8 cases. Am J Surg Pathol. 113(2):129-36. PMID 17031656
radiol. 22(3):464-9. PMID:11237967 2018. Prayer D, Grois N, Prosch H, Gadner H, 31(11):1709-18. PMID:18059228 2051. Quillien V, Lavenu A, Karayan-Tapon
2002. Pompili A, Calvosa F, Caroli F, Mastroste- Barkovich AJ (2004). MR imaging presentation 2036. Preusser M, Laggner U, Haberler C, L, Carpentier C, Labussiere M, Lesimple T,
fano R, Occhipinti E, Raus L, et al. (1993). The of intracranial disease associated with Langer- Heinz! H, Budka H, Hainfellner JA (2006). Com- et al. (2012). Comparative assessment of 5
lransdural extension of gliomas. J Neurooncol. hans cell histiocytosis. AJNR Am J Neuroradiol. parative analysis of NeuN immunoreactivity in methods (methylation-specific polymerase
15(1):67-74. PMID:8455064 25(5):880-91. PMID:15140741 primary brain tumours: conclusions for rational chain reaction, Methylight, pyrosequencing,
2003. Ponzoni M, Arrigoni G, Gould VE, Del 2019. Prayson RA (1997). Myxopapillary epen- use in diagnostic histopathology. Histopatholo- methylation-sensitive high-resolution mel-
Curto B, Maggioni M, Scapinello A, et al. dymomas: a clinicopathologic study of 14 cases gy. 48(4):438-44. PMID:16487366 ting, and immunohistochemistry) to analyze
(2000). Lack of CD 29 (beta1 integrin) and CD including MIB· 1 and p53 immunoreactivity. Mod 2037. Preusser M, Woehrer A, Koperek 0, 06-methylguanine-DNA-methyltranferase in
54 (ICAM-1) adhesion molecules in intravascu- Pathol. 10(4):304-10. PMID:9110291 Rottenfusser A, Dieckmann K, Gatterbauer B, a series of 100 glioblastoma patients. Cancer.
tar lymphomatosis. Hum Pathol. 31(2):220-6. 2020. Prayson RA (1999). Clinicopathologic et al. (2010). Primary central nervous system 118(17):4201-11. PMID:22294349
PMID:10685637 study of 61 patients with ependymoma inclu- lymphoma: a clinicopathological study of 75 ca- 2052. Rades D, Fehlauer F, Lamszus K,
2004. Ponzoni M, BergerF, Chassagne-Clement ding MIB-1 immunohistochemistry. Ann Diagn ses. Pathology. 42(6):547-52. PMID:20854073 Schild SE, Hagel C, Westphal M, et al. (2005).

References 387
Well-differentiated neurocytoma: what is 110(20):8188-93. PMID:23633565 PMID:8504413 2101. Resta N, Lauriola L, Puca A, Susca FC
the best available treatment? Neuro Oneel. 2069. Raney RB, Ater JL, Herman-Liu A, Leeds 2085. Reifenberger G, Louis DN (2003). Oligo- Albanese A: Sabatino G, et al. (2006). Gang'.
7(1):77-83. PMID:15701284 NE, Cleary KR, Womer RB, et al. (1994). Pri- dendroglioma: toward molecular definitions in hoghoma ansmg m a Peutz-Jeghers patient: a
2053. Rades D, Schild SE, Fehlauer F (2004). mary intraspinal soft-tissue sarcoma in child- diagnostic neuro-oncology. J Neuropathol Exp case report with molecular implications. Acta
Prognostic value of the MIB-1 labeling index for hood: report of two cases with a review of the Neurol. 62(2):111-26. PMID:12578221 Neuropathol. 112(1):106-11. PMID:16733653
central neurocytomas. Neurology. 62(6):987-9. literature. Med Pediatr Oneel. 23(4):359-64. 2086. Reifenberger G, Reifenberger J, lchimura 2102. Reuss DE, Habel A, Hagenlocher c
PMID:15037708 PMID:8058008 K, Meltzer PS, Collins VP (1994). Amplification Mucha J, Ackermann U, Tessmer C, et ai
2054. Radke J, Gehlhaar C, Lenze D, Capper 2070. Rankine AJ, Filion PR, Platten MA, of multiple genes from chromosomal region (2014). Neurofibromin specific antibody diffe-
D, Bock A, Heppner FL, et al. (2015). The evo- Spagnolo DV (2004 ). Perineurioma: a clinico- 12q13-14 in human malignant gliomas: preli- rentiates malignant peripheral nerve sheath
lution of the anaplastic cerebellar liponeurocyto- pathological study of eight cases. Pathology. minary mapping of the amplicons shows prefe- tumors (MPNST) from other spindle cell neo-
ma: case report and review of the literature. Clin 36(4):309-15. PMID:15370128 rential involvement of CDK4, SAS, and MDM2. plasms. Acta Neuropathol. 127(4):565-72.
Neuropathol. 34(1):19-25. PMID:25250652 2071. Raoux D, Duband S, Forest F, Trombert Cancer Res. 54(16):4299-303. PMID:8044775 PMID:24464231
2055. Raffel C, Jenkins RB, Frederick L, Hebrink B, Chambonniere ML, Dumollard JM, et al. 2087. Reifenberger G, Szymas J, Wechsler W 2103. Reuss DE, Mamatjan Y, Schrimpf D,
D, Alderete B, Fults DW, et al. (1997). Sporadic (2010). Primary central nervous system lym- (1987). Differential expression of glial- and neu- Capper D, Hovestadt V, Kratz A, et al. (2015).
medulloblastomas contain PTCH mutations. phoma: immunohistochemical profile and prog- ronal-associated antigens in human tumors of IDH mutant diffuse and anaplastic astrocyto-
Cancer Res. 57(5):842-5. PMID:9041183 nostic significance. Neuropathology. 30(3):232- the central and peripheral nervous system. Acta mas have similar age at presentation and
2056. Ragel BT, Osborn AG, Whang K, Town- 40. PMID:19925562 Neuropathol. 74(2):105-23. PMID:3314309 little difference in survival: a grading problem
send JJ, Jensen RL, Couldwell WT (2006). 2072. Rasheed BK, Mclendon RE, Friedman 2088. Reifenberger G, Weber RG, Riehmer V, for WHO. Acta Neuropathol. 129(6):867-73.
Subependymomas: an analysis of clinical and HS, Friedman AH, Fuchs HE, Bigner DD, et Kaulich K, Willscher E, Wirth H, et al.; German PMID:25962792
imaging features. Neurosurgery. 58(5):881-90, al. (1995). Chromosome 10 deletion map- Glioma Network (2014). Molecular characteriza- 2104. Reuss DE, Piro RM, Jones DT, Simon
discussion 881-90. PMID:16639322 ping in human gliomas: a common deletion tion of long-term survivors of glioblastoma using M, Ketter R, Kool M, et al. (2013). Secretory
2057. Raghavan R, Balani J, Perry A, Margraf region in 10q25. Oncogene. 10(11):2243-6. genome- and transcriptome-wide profiling. Int J meningiomas are defined by combined KLF4
L, Veno MB, Cai DX, et al. (2003). Pediatric PMID:7784070 Cancer. 135(8):1822-31. PMID:24615357 K409Q and TRAF7 mutations. Acta Neuropa-
oligodendrogliomas: a study of molecular al- 2073. Rauen KA (2013). The RASopathies. 2089. Reifenberger G, Weber T, Weber RG, thol. 125(3):351-8. PMID:23404370
terations on 1 p and 19q using fluorescence in Annu Rev Genomics Hum Genet. 14:355-69. Wolter M, Brandis A, Kuchelmeister K, et al. 2105. Reuss DE, Sahm F, Schrimpf D, Wiestler
situ hybridization. J Neuropathol Exp Neurol. PMID:23875798 (1999). Chordoid glioma of the third ventricle: B, Capper D, Koelsche C, et al. (2015). ATRX
62(5):530-7. PMID:12769192 207 4. Rauhut F, Reinhardt V, Budach V, Wie- immunohistochemical and molecular genetic and IDH1-R132H immunohistochemistry with
2058. Raghavan R, Dickey WT Jr, Margraf LR, demayer H, Nau HE (1989). lntramedullary pi- characterization of a novel tumor entity. Brain subsequent copy number analysis and IDH
White CL 3rd, Coimbra C, Hynan LS, et al. locytic astrocytomas-a clinical and morphologi- Pathol. 9(4):617-26. PMID:10517500 sequencing as a basis for an "integrated" dia-
(2000). Proliferative activity in craniopharyngi- cal study after combined surgical and photon or 2090. Reifenberger J, Reifenberger G, lchimura gnostic approach for adult astrocytoma, oligo-
omas: clinicopathological correlations in adults neutron therapy. Neurosurg Rev. 12(4):309-13. K, Schmidt EE, Wechsler W, Collins VP (1996). dendroglioma and glioblastoma. Acta Neuropa-
and children. Surg Neurol. 54(3):241-7, discus- PMID:2594208 Epidermal growth factor receptor expressi- thol. 129(1):133-46. PMID:25427834
sion 248. PMID:11118571 2075. Rausch T, Jones DT, Zapatka M, Stutz on in oligodendroglial tumors. Am J Pathol. 2106. Reyes-Mugica M, Chou P, Byrd S, Ray
2059. Raghavan R, Steart PV, Weller RO AM, Zichner T, Weischenfeldt J, et al. (2012). 149(1):29-35. PMID:8686753 V, Castelli M, Gattuso P, et al. (1993). Nevome-
(1990). Cell proliferation patterns in the diag- Genome sequencing of pediatric medullob- 2091. Reifenberger J, Reifenberger G, Liu L, lanocytic proliferations in the central nervous
nosis of astrocytomas, anaplastic astrocytomas lastoma links catastrophic DNA rearrangements James CD, Wechsler W, Collins VP (1994). system of children. Cancer. 72(7):2277-85.
and glioblastoma multiforme: a Ki-67 study. with TP53 mutations. Cell. 148(1-2):59-71. Molecular genetic analysis of oligodendroglial PMID:8374887
Neuropathol Appl Neurobiol. 16(2):123-33. PMID:22265402 tumors shows preferential allelic deletions on 2108. Rezai AR, Woo HH, Lee M, Cohen H,
PMID:2161084 2076. Rawlinson DG, Herman MM, Rubinstein 19q and 1p. Am J Pathol. 145(5):1175-90. Zagzag D, Epstein FJ (1996). Disseminated
2060. Raghunathan A, Wani K, Armstrong TS, LJ (1973). The fine structure of a myxopapillary PMID:7977648 ependymomas of the central nervous system. J
Vera-Bolanos E, Fouladi M, Gilbertson R, et al.; ependymoma of the filum terminale. Acta Neu- 2092. Reifenberger J, Ring GU, Gies U, Cob- Neurosurg. 85(4):618-24. PMID:8814165
Collaborative Ependymoma Research Network ropathol. 25(1):1-13. PMID:4727729 bers L, Oberstrass J, An HX, et al. (1996). Ana- 2109. Rhiew RB, Manjila S, Lozen A, Guthikon-
(2013). Histological predictors of outcome in 2077. Ray WZ, Blackburn SL, Casavilca-Zam- lysis of p53 mutation and epidermal growth fac- da M, Sood S, Kupsky WJ (2010). l.eptornenin-
ependymoma are dependent on anatomic site brano S, Barrionuevo C, Orrego JE, Heinicke tor receptor amplification in recurrent gliomas geal dissemination of a pediatric neoplasm with
within the central nervous system. Brain Pathol. H, et al. (2009). Clinicopathologic features of re- with malignant progression. J Neuropathol Exp 1p19q deletion showing mixed immunohis-
23(5):584-94. PMID:23452038 current dysembryoplastic neuroepithelial tumor Neurol. 55(7):822-31. PMID:8965097 tochemical features of an oligodendrogliilma
2061. Rainho CA, Rogatto SR, de Moraes and rare malignant transformation: a report of 5 2093. Reis F, Faria AV, Zanardi VA, Menezes and neurocytoma. Acta Neurochir (Wien).
LC, Barbieri-Neto J (1992). Cytogenetic study cases and review of the literature. J Neuroon- JR, Cendes F, Queiroz LS (2006). Neuroi- 152(8):1425-9. PMID:20446099
of a pineocytoma. Cancer Genet Cytogenet. col. 94(2):283-92. PMID:19267228 maging in pineal tumors. J Neuroimaging. 2110. Rhodes RH, Cole M, Takaoka Y, Roess-
64(2):127-32. PMID:1486561 2078. Raymond AA, Halpin SF, Alsanjari N, 16(1):52-8. PMID:16483277 mann U, Cotes EE, Simon J (1994). Intra-
2062. Rainov NG, l.ubbe J, Renshaw J, Prit- Cook MJ, Kitchen ND, Fish DR, et al. (1994). 2094. Reis RM, Hara A, Kleihues P, Ohgaki ventricular cerebral neuroblastoma. Analysis
chard-Jones K, Luthy AR. Aguzzi A (1995). Dysembryoplastic neuroepithelial tumor. Fea- H (2001 ). Genetic evidence of the neoplastic of subtypes and comparison with hemisphe-
Association of Wilms' tumor with primary brain tures in 16 patients. Brain. 117(Pt 3):461-75. nature of gemistocytes in astrocytomas. Acta ric neuroblastoma. Arch Pathol Lab Med.
tumor in siblings. J Neuropathol Exp Neurol. PMID:8032857 Neuropathol. 102(5):422-5. PMID:11699553 118(9):897-911. PMID:8080360
54(2):214-23. PMID:7876889 2079. Raza SM, Lang FF, Aggarwal BB, Fuller 2095. Reis RM, Konu-Lebleblicioglu D. Lopes 2111. Ribeiro RC, Sandrini F, Figueiredo B,
2063. Raisanen J, Biegel JA, Hatanpaa KJ, GN, Wildrick DM, Sawaya R (2002). Necrosis JM, Kleihues P, Ohgaki H (2000). Genetic pro- Zambelli GP, Michalkiewicz E, Lafferty AR, et
Judkins A, White CL, Perry A (2005). Chromo- and glioblastoma: a friend or a foe? A review file of gliosarcomas. Am J Pathol. 156(2):425- al. (2001 ). An inherited p53 mutation that con-
some 22q deletions in atypical teratoid/rhab- and a hypothesis. Neurosurgery. 51(1):2-12, 32. PMID:10666371 tributes in a tissue-specific manner to pediatric
doid tumors in adults. Brain Pathol. 15(1):23-8. discussion 12-3. PMID:12182418 2096. Reis-Filho JS, Faoro LN, Carrilho C, adrenal cortical carcinoma. Pree Natl A cad Sci
PMID:15779233 2080. Read TA, Fogarty MP, Markant SL, Bleggi-Torres LF, Schmitt FC (2000). Evaluati- US A. 98(16):9330-5. PMID:11481490
2064. Raja Al, Yeaney GA, Jakacki RI, Ha- Mclendon RE, Wei Z, Ellison DW, et al. on of cell proliferation, epidermal growth factor 2112. Ribeiro S, Napoli I, White IJ, Parrinello
milton RL, Pollack IF (2008). Extraventricular (2009). Identification of CD15 as a marker for receptor, and bcl-2 immunoexpression as prog- S, Flanagan AM, Suter U, et al. (2013). Injury
neurocytoma in neurofibromatosis Type 1: tumor-propagating cells in a mouse model of nostic factors for patients with World Health Or- signals cooperate with Nf1 loss to relieve
case report. J Neurosurg Pediatr. 2(1):63-7. medulloblastoma. Cancer Cell. 15(2):135-47. ganization grade 2 oligodendroglioma. Cancer. the tumor-suppressive environment of adult
PMID:18590398 PMID:19185848 88(4):862-9. PMID:10679656 peripheral nerve. Cell Rep. 5(1):126-36.
2065. Rajan B, Ashley S, Gorman C, Jose CC, 2081. Reardon W, Zhou XP, Eng C (2001). A 2097. Reithmeier T, Gumprecht H, Stolzle A, PMID:24075988
Horwich A, Bloom HJ, et al. (1993). Cranio- novel germline mutation of the PTEN gene in Lumenta CB (2000). lntracerebral paragang- 2113. Ricci A Jr, Parham DM, Woodruff JM,
pharyngioma-a long-term results following limi- a patient with macrocephaly, ventricular dilata- lioma. Acta Neurochir (Wien). 142(9):1063-6. Callihan T, Green A, Erlandson RA (1984).
ted surgery and radiotherapy. Radiother Oneel. tion, and features of VATER association. J Med PMID:11086818 Malignant peripheral nerve sheath tumors ari-
26(1):1-10. PMID:8438080 Genet. 38(12):820-3. PMID:11748304 2098. Remke M, Ramaswamy V, Peacock J, sing from ganglioneuromas. Am J Surg Pathol.
2066. Rajaram V, Brat DJ, Perry A (2004). 2082. Reddy AT. Janss AJ, Phillips PC, Weiss Shih DJ, Koelsche C, Northcott PA, et al. (2013). 8(1):19-29. PMID:6696163
Anaplastic meningioma versus meningeal he- HL, Packer RJ (2000). Outcome for children TERT promoter mutations are highly recurrent 2114. Rickard KA, Parker JR, Vitaz TW, Plaga
mangiopericytoma: immunohistochemical and with supratentorial primitive neuroectoder- in SHH subgroup medulloblastoma. Acta Neu- AR, Wagner S, Parker JC Jr (2011). Papillary
genetic markers. Hum Pathol. 35(11):1413-8. mal tumors treated with surgery, radiation, ropathol. 126(6):917-29. PMID:24174164 tumor of the pineal region: two case studies
PMID:15668900 and chemotherapy. Cancer. 88(9):2189-93. 2099. Rencic A, Gordon J, Otte J, Curtis M, and a review of the literature. Ann Clin Lab Sci.
2067. Raju GP, Urion DK, Sahin M (2007). PMID:10813733 Kovatich A, Zellick P, et al. (1996). Detection 41(2):174-81. PMID:21844577
Neonatal subependymal giant cell astrocytoma: 2083. Reifenberger G, Kaulich K, Wiestler OD, of JC virus DNA sequence and expression of 2115. Rickert CH, Hasselblatt M (2006). Cyto-
new case and review of literature. Pediatr Neu- Blurncke I (2003). Expression of the CD34 an- the viral oncoprotein, tumor antigen, in brain of genetic features of ependymoblastomas. Ada
rol. 36(2):128-31. PMID:17275668 tigen in pleomorphic xanthoastrocytomas. Acta immunocompetent patient with oligoastrocyto- Neuropathol. 111(6):559-62. PMID:16718352
2068. Ramkissoon LA, Horowitz PM, Craig JM, Neuropathol. 105(4):358-64. PMID:12624789 ma. Pree Natl Acad Sci US A. 93(14):7352-7. 2116. Rickert CH, Paulus W (2001). Epidemio-
Ramkissoon SH, Rich BE, Schumacher SE, et 2084. Reifenberger G, Liu L, lchimura K, PMID:8692997 logy of central nervous system tumors in child-
al. (2013). Genomic analysis of diffuse pediatric Schmidt EE, Collins VP (1993). Amplification 2100. Reni M, Ferreri AJ, Zeidan MC, Villa E hood and adolescence based on the new WHO
low-grade gliomas identifies recurrent oncoge- and overexpression of the MDM2 gene in a (1997). Primary brain lymphomas in patients classification. Childs Nerv Syst. 17(9):503-11.
nic truncating rearrangements in the transcrip- subset of human malignant gliomas without with a prior or concomitant malignancy. J Neu- PMID:11585322
tion factor MYBL 1. Pree Natl Acad Sci U S A. p53 mutations. Cancer Res. 53(12):2736-9. rooncol. 32(2):135-42. PMID:9120542 2117. Rickert CH, Paulus W (2001). Tumors

388 References
of the choroid plexus. Microsc Res Tech. Central neurocytoma. A clinicopathological, C, Bryant SC, Jenkins RB (2008). Epithelial and radiologic and histopathologic features. Neuro-
52(1):104-11. PMID:11135453 immunohistochemica/ and u/lrastructural study pseudoepithelial differentiation in glioblastoma radiol J. 26(6):639-48. PMID:24355182
2118. Rickert CH, Paulus W (2002). Genetic of 7 cases. Pathol Res Pract. 191(2):100-11. and gliosarcoma: a comparative morpho- 2167. Romero-Rojas AE, Melo-Uribe MA, Ba-
characterisation of granular cell tumours. Acta PMID:7567679 logic and molecular genetic study. Cancer. rajas-Solano PA, Chinchilla-Olaya SI, Escobar
Neuropathol. 103(4):309-12. PMID:11904749 2135. Roberts AE, Allanson JE, Tartaglia M, 113(10):2779-89. PMID:18816605 LI, Hernandez-Walteros DM (2011). Spindle
2119. Rickert CH, Paulus W (2002). No chro- Gelb BD (2013). Noonan syndrome. Lancet. 2152. Rodriguez FJ, Scheithauer BW, Jenkins cell oncocytoma of the adenohypophysis. Brain
mosomal imbalances detected by comparative 381 (9863):333-42. PMID:23312968 R, Burger PC, Rudzinskiy P, Vlodavsky E, et Tumor Pathol. 28(4):359-64. PMID:21833579
genomic hybridisation in a case of fetal imma- 2136. Roberts CW, Biegel JA (2009). The al. (2007). Gliosarcoma arising in oligodendro- 2168. Roncaroli F, Riccioni L, Cerati M, Ca-
ture teratoma. Childs Nerv Syst. 18(11):639-43. role of SMARCB1/IN11 in development of rh- glial tumors ("oligosarcoma"): a clinicopatho- pella C, Calbucci F, Trevisan C, et al. (1997).
PMID 12420126 abdoid tumor. Cancer Biol Ther. 8(5):412-6. logic study. Am J Surg Pathol. 31(3):351-62. Oncocytic meningioma. Am J Surg Pathol.
2120. Rickert CH, Paulus W (2004). Compa- PMID:19305156 PMID:17325476 21(4):375-82. PMID:9130983
ralive genomic hybridization in central and 2137. Roberts CW, Orkin SH (2004). The SW// 2153. Rodriguez FJ, Scheithauer BW, Perry 2169. Roncaroli F, Scheithauer BW, Cenacchi
peripheral nervous system tumors of childhood SNF complex-chromatin and cancer. Nat Rev A, Oliveira AM, Jenkins RB, Oviedo A, et al. G, Horvath E, Kovacs K, Lloyd RV, et al. (2002).
and adolescence. J Neuropathol Exp Neurol. Cancer. 4(2):133-42. PMID:14964309 (2008). Ependymal tumors with sarcomatous 'Spindle cell oncocytoma' of the adenohypophy-
63(5):399-417. PMID:15198120 2138. Roberts RO, Lynch CF, Jones MP, Hart change ("ependymosarcoma"): a clinicopatho- sis: a tumor of folliculostellate cells? Am J Surg
2121. Rickert CH, Simon R, Bergmann M, MN (1991). Medulloblasloma: a population-ba- logic and molecular cytogenetic study. Am J Pathol. 26(8):1048-55. PMID:12170092
Dockhorn-Dworniczak B, Paulus W (2000). sed study of 532 cases. J Neuropalhol Exp Surg Pathol. 32(5):699-709. PMID:18347506 2170. Rong Y, Durden DL, Van Meir EG, Brat
Comparative genomic hybridization in pineal Neurol. 50(2):134-44. PMID:2010773 2154. Rodriguez FJ, Scheithauer BW, Robbins DJ (2006). 'Pseudopalisading' necrosis in gli-
germ cell tumors. J Neuropathol Exp Neurol. 2139. Robertson RP (2011).Translational En- PD, Burger PC, Hessler RB, Perry A, et al. oblastoma: a familiar morphologic feature that
59(9):815-21. PM/0:11005262 docrinology & Metabolism. Neoplasia Update (2007). Ependymomas with neuronal differenti- links vascular pathology, hypoxia, and angioge-
2122. Rickert CH, Simon R, Bergmann M, Chevy Chase, Maryland 20815: The Endocrine ation: a morphologic and immunohistochemical nesis. J Neuropathol Exp Neural. 65(6):529-39.
Dockhorn-Dworniczak B, Paulus W (2001). Society. spectrum. Acta Neuropathol. 113(3):313-24. PMID:16783163
Comparative genomic hybridization in pineal 2140. Robin YM, Guillou L, Michels JJ, Coindre PMID:17061076 2171. Rorke LB (1983). The cerebellar me-
parenchymal tumors. Genes Chromosomes JM (2004). Human herpesvirus 8 immunos- 2155. Rodriguez FJ, Scheithauer BW, Tsuno- dulloblastoma and its relationship to primitive
Cancer. 30(1):99-104. PMID:11107183 taining: a sensitive and specific method for da S, Kovacs K, Vidal S, Piepgras DG (2007). neuroeclodermal tumors. J Neuropathol Exp
2123. Rickert CH, Wiest/er OD, Paulus W diagnosing Kaposi sarcoma in paraffin-embed- The spectrum of malignancy in craniopharyn- Neural. 42(1):1-15. PMID6296325
(2002). Chromosomal imbalances in choroid ded sections. Am JC/in Pathol. 121(3):330-4. gioma. Am J Surg Pathol. 31(7):1020-8. 2172. Rorke LB, Packer RJ, Biegel JA (1996).
plexus tumors. Am J Pathol. 160(3):1105-13. PMID:15023036 PMID:17592268 Central nervous system atypical leratoid/
PMID:11891207 2141. Robinson DR, Wu YM, Kalyana-Sunda- 2156. Rodriguez FJ, Schniederjan MJ, Nico- rhabdoid tumors of infancy and childhood: de-
2124. Rickman OS, Bobek MP, Misek DE, Kuick ram S, Cao X, Lonigro RJ, Sung YS, et al. laides T, Tihan T, Burger PC, Perry A (2015). finition of an entity. J Neurosurg. 85(1):56-65.
R, Blaivas M, Kurnit DM, et al. (2001 ). Distincti- (2013). Identification of recurrent NAB2-STAT6 High rate of concurrent BRAF-KIM 1549 gene PMID:8683283
ve molecular profiles of high-grade and /ow-gra- gene fusions in solitary fibrous tumor by inte- fusion and 1 p deletion in disseminated oligo- 2173. Rosai J, Parkash V, Reuter V (1994). In-
de gliomas based on oligonucleotide microar- grative sequencing. Nal Genet. 45(2):180-5. dendroglioma-like leptomeningeal neoplasms vited Review: The Origin of Mediastinal Germ
ray analysis. Cancer Res. 61(18):6885-91. PMID:23313952 (DOLN). Acta Neuropathol. 129(4):609-10. Cell Tumors. Int J Surg Pathol. 2:73-8.
PMID:11559565 2142. Robinson G, Parker M, Kranenburg TA, PMID:257207 45 2174. Rosemberg S, Fujiwara D (2005). Epi-
2125. Riegert-Johnson DL, Gleeson FC, Ro- Lu C, Chen X, Ding L, et al. (2012). Novel muta- 2157. Rodriguez FJ, Tihan T, Lin D, McDonald demiology of pediatric tumors of lhe nervous
berts M, Tholen K, Youngborg L, Bullock M, tions target distinct subgroups of medulloblasto- W, Nigro J, Feuerstein B, et al. (2014). Clinico- system according to the WHO 2000 classifi-
et al. (2010). Cancer and Lhermitte-Duclos ma. Nature. 488(7409):43-8. PMID:22722829 pathologic features of pediatric oligodendroglio- cation: a report of 1, 195 cases from a single
disease are common in Cowden syndrome 2143. Robinson GW, Orr BA, Wu G, Gur- mas: a series of 50 patients. Am J Surg Pathol. institution. Childs Nerv Syst. 21(11):940-4.
patients. Hered Cancer Clin Pract. 8(1):6. urangan S, Lin T, Qaddoumi I, et al. (2015). 38(8):1058-70. PMID:24805856 PMID:16044344
PMID:20565722 Vismodegib Exerts Targeted Efficacy Against 2158. Rodriguez HA, Berthrong M (1966). Mul- 2175. Rosemberg S. Vieira GS (1998). [Dysem-
2126. Riemenschneider MJ, Reifenberger G Recurrent Sonic Hedgehog-Subgroup Medul- tiple primary intracranial tumors in von Reck- bryoplastic neuroepithelial tumor. An epidemio-
(2009). Molecular neuropathology of gliomas. loblastoma: Resulls From Phase II Pediatric linghausen's neurofibromalosis. Arch Neurol. logical study from a single institution]. Arq Neu-
lnl J Mal Sci. 10(1):184-212. PMID:19333441 Brain Tumor Consortium Studies PBTC-025B 14(5):467-75. PMID:4957904 ropsiquiatr. 56(2):232-6. PMID:9698733
2127. Riemersma SA, Jordanova ES, Schop and PBTC-032. J Clin Oneal. 33(24):2646-54. 2159. Rodriguez LA, Edwards MS, Levin VA 2176. Rosenberg AS, Langee CL, Stevens
RF, Philippa K, Looijenga LH, Schuuring E, el PMID:26169613 (1990). Management of hypothalamic gliomas GL, Morgan MB (2002). Malignant peripheral
al. (2000). Extensive genetic alterations of the 2144. Roche PH, Figarella-Branger D, Regis in children: an analysis of 33 cases. Neu- nerve sheath tumor with perineurial differentia-
HLA region, including homozygous deletions J, Peragut JC (1996). Cauda equina parag- rosurgery. 26(2):242-6, discussion 246-7. tion: "malignant perineurioma". J Gulan Pathol.
of HLA class II genes in B-cell lymphomas anglioma with subsequent intracranial and PMID:2308672 29(6):362-7. PMID:12135468
arising in immune-privileged sites. Blood. intraspinal metastases. Acta Neurochir (Wien). 2160. Roerig P, Nessling M, Radlwimmer 2177. Rosenberg DS, Demarquay G, Jouvet
96(10):3569-77. PMID:11071656 138(4):475-9. PMID:8738400 B, Joos S, Wrobel G, Schwaenen C, et al. A, Le Bars D, Slreichenberger N, Sindou M,
2128. Riemersma SA, Oudejans JJ, Vonk MJ, 2145. Rodriguez EF, Scheithauer BW, Giannini (2005). Molecular classification of human glio- et al. (2005). [11C]-Methionine PET: dysem-
Dreef EJ, Prins FA, Jansen PM, et al. (2005). C, Rynearson A, Gen L, Hoesley B, etal. (2011). mas using matrix-based comparative genomic bryoplastic neuroepithelial tumours compared
High numbers of tumour-infiltrating activated Pl3KIAKT pathway alterations are associated hybridization. lnl J Cancer. 117(1):95-103. with other epileptogenic brain neoplasms. J
cytotoxic T lymphocytes, and frequent loss with clinically aggressive and hislologically ana- PMID:15880582 Neural Neurosurg Psychiatry. 76(12):1686-92.
of HLA class I and II expression, are featu- plastic subsets of pilocytic astrocyloma. Acta 2161. Roessler E, Belloni E, Gaudenz K, Jay P, PMID:16291894
res of aggressive B cell lymphomas of lhe Neuropathol. 121(3):407-20. PMID:21113787 Berta P, Scherer SW, et al. (1996). Mutations 2178. Rosenblum MK (1998). Ependymal
brain and testis. J Pathol. 206(3):328-36. 2146. Rodriguez FJ, Folpe AL, Giannini C, in the human Sonic Hedgehog gene cause tumors: A review of their diagnostic surgical
PM/0: 15887291 Perry A (2012). Pathology of peripheral nerve holoprosencephaly. Nal Genet. 14(3):357-60. pathology. Pedialr Neurosurg. 28(3):160-5.
2129. Rienstein S, Adams EF, Pilzer D, Gol- sheath tumors: diagnostic overview and update PMID:8896572 PMID:9705595
dring AA, Goldman B, Friedman E (2003). on selected diagnostic problems. Acta Neuro- 2162. Roggendorf W, Strupp S, Paulus W 2179. Rosenblum MK, Erlandson RA, Aleksic
Comparative genomic hybridization analysis of pathol. 123(3):295-319. PMID:22327363 (1996). Distribution and characterization SN, Budzilovich GN (1990). Melanotic ependy-
craniopharyngiomas. J Neurosurg. 98(1):162- 2147. Rodriguez FJ, Mola RA, Scheithauer of microglia/macrophages in human brain moma and subependymoma. Am J Surg Pathol.
4. PMID:12546365 BW, Giannini C, Blair H, New KC, et al. (2009). tumors. Acta Neuropathol. 92(3):288-93. 14(8):729-36. PMID:2378394
2130. Ringertz N (1950). Grading of gliomas. lnlerphase cytogenetics for 1p19q and �1;19) PMID:8870831 2180. Rosenblum MK, Erlandson RA, Budzi-
Acta Pathol Microbial Scand. 27(1):51-64. (q10;p10) may distinguish prognostically rele- 2163. Roldan G, Chan J, Eliasziw M, Cairn- lovich GN (1991). The lipid-rich epithelioid gli-
PMID:15406242 vant subgroups in extraventricular neurocyto- cross JG, Forsyth PA (2011). Leptomeningeal oblastoma. Am J Surg Pathol. 15(10):925-34.
2131. Ritter JH, Mills SE, Nappi 0, Wick MR ma. Brain Pathol. 19(4):623-9. PMID:18710393 disease in oligodendroglial tumors: a popula- PMID:1718177
(1995). Angiosarcoma-like neoplasms of 2148. Rodriguez FJ, Perry A, Gutmann DH, tion-based study. J Neuroonco/. 104(3):811-5. 2181. Roser F, Nakamura M, Bellinzona M, Ro-
epithelial organs: true endothelial tumors or O'Neill BP, Leonard J, Bryant S, et al. (2008). PMID:21373968 sahl SK, Ostertag H, Samii M (2004). The pro-
variants of carcinoma? Semin Diagn Pathol. Gliomas in neurofibromatosis type 1: a clinico- 2164. Rollison DE, Utaipat U, Ryschkewitsch C, gnostic value of progesterone receptor status
12(3):270-82. PMID:8545593 pathologic study of 100 patients. J Neuropathol Hou J, Goldthwaite P, Daniel R, el al. (2005). in meningiomas. J Clin Palhol. 57(10):1033-7.
2132. Rivera AL, Takei H, Zhai J, Shen SS, Ro Exp Neural. 67(3)240-9. PMID:18344915 Investigation of human brain tumors for the PMID:15452155
JY, Powell SZ (2010). Useful immunohistoche- 2149. Rodriguez FJ, Perry A, Rosenblum MK, presence of polyomavirus genome sequences 2182. Roser F, Nakamura M, Brandis A, Hans
mical markers in differentiating hemangiob- Krawitz S, Cohen KJ, Lin D, et al. (2012). Dis- by lwo independent laboratories. Int J Cancer. V, Vorkapic P, Samii M (2004 ). Transition from
lastoma versus metastatic renal cell carcinoma. seminated oligodendroglia/-like leplomeningeal 113(5):769-74. PMID:15499616 meningeal melanocytoma lo primary cere-
Neuropathology. 30(6):580-5. PMID:20374497 tumor of childhood: a distinctive clinicopatho- 2165. Romani R, Niemela M, Celik 0, lsarakul bral melanoma. Case report. J Neurosurg.
2133. Roach ES, Sparagana SP (2010). Diag- logic entity. Acta Neuropalhol. 124(5):627-41. P, Paetau A, Hernesniemi J (2010). Ectopic re- 101(3):528-31. PMID:15352613
nostic criteria for tuberous sclerosis complex. PMID:22941225 currence of craniopharyngioma along the surgi- 2183. Rossi A, Gama A, Gonsales A, Gandolfo
In: Tuberous sclerosis complex. Genes, clinical 2150. Rodriguez FJ, Scheithauer BW, Burger cal route: case report and literature review. Acta C, Garre ML, Milanaccio C, et al. (2006). Neu-
features, and therapeutics. Kwiatkowski DJ, PC, Jenkins S, Giannini C (2010). Anaplasia Neurochir (Wien). 152(2):297-302, discussion roimaging of pediatric craniopharyngiomas: a
Whittemore VH, Thiele EA, eds. Wiley-Black- in pilocytic astrocytoma predicts aggressive 302. PMID:19499168 pictorial essay. J Pediatr Endocrinol Metab. 19
well: Weinheim, Germany, pp. 21-25. behavior. Am J Surg Pathol. 34(2):147-60. 2166. Romero-Rojas AE, Diaz-Perez JA, Ari- Suppl 1 :299-319. PMID:16700305
2134. Robbins P, Segal A, Narula S, Sto- PMID:20061938 za-Serrano LM, Amaro D, Lozano-Castillo 2184. Rossi ML, Jones NR, Candy E, Nicoll
kes B, Lee M, Thomas W, et al. (1995). 2151. Rodriguez FJ, Scheithauer BW, Giannini A (2013). Primary gliosarcoma of the brain: JA, Compton JS, Hughes JT, et al. (1989). The

References 389
mononuclear cell infiltrate compared with survi- clinicopathologic and flow cytometric study of 13(4 ):393-9. PMID:24506340 (2005). Mutational analysis of the TSC1 and
val in high-grade astrocytomas. Acta Neuropa- 13 cases presenting in the central nervous sys- 2217. Sahm F, Bissel J, Koelsche C, Schweizer TSC2 genes in a diagnostic setting: genoty-
thol. 78(2):189-93. PMID:2750489 tem. Cancer. 77(9):1884-91. PMID:8646689 L, Capper D, Reuss D, et al. (2013). AKT1E17K pe-phenotype correlations and comparison of
2185. Rossi S, Rodriguez FJ, Mota RA, Dei 2201. Rushing EJ, Cooper PB, Quezada M, Be- mutations cluster with meningothelial and tran- diagnostic DNA techniques in Tuberous Sclero-
Tos AP, Di Paola F, Bendini M, et al. (2009). gnami M, Crespo A, Smirniotopoulos JG, et al. sitional meningiomas and can be detected by sis Complex. Eur J Hum Genet. 13(6):731-41.
Primary leptomeningeal oligodendroglioma (2007). Subependymoma revisited: clinicopa- SFRP1 immunohistochemistry. Acta Neuropa- PMID:15798777
with documented progression to anaplasia and thological evaluation of 83 cases. J Neurooncol. thol. 126(5):757-62. PMID:24096618 2232. Sandberg DI, Ragheb J, Dunoyer C, Bha-
t(1 ;19)(q10;p10) in a child. Acta Neuropathol. 85(3):297-305. PMID:17569000 2218. Sahm F, Capper D, Preusser M, Meyer J, tia S, Olavarria G, Morrison G (2005). Surgical
118(4):575-7. PMID:19562354 2202. Rushing EJ, Olsen C, Mena H, Rueda Stenzinger A, Lasitschka F, et al. (2012). BRAF- outcomes and seizure control rates after resec-.
2186. Rossitch E Jr, Zeidman SM, Burger PC, ME, Lee YS, Keating RF, et al. (2005). Central V600E mutant protein is expressed in cells of lion of dysembryoplastic neuroepithelial tumors.
Cumes JT, Harsh C, Anscher M, et al. (1990). nervous system meningiomas in the first two variable maturation in Langerhans cell histiocy- Neurosurg Focus. 18 6A:E5. PMID:16048291
Clinical and pathological analysis of spinal decades of life: a clinicopathological analysis of tosis. Blood. 120(12):e28-34. PMID:22859608 2233. Sanford RA, Bowman R, Tomita T, De
cord astrocytomas in children. Neurosurgery. 87 patients. J Neurosurg. 103(6) Suppl:489-95. 2219. Sahm F, Koelsche C, Meyer J, Pusch S, Leon G, Palka P (1999). A 16-year-old male
27(2):193-6. PMID:2385335 PMID:16383246 Lindenberg K, Mueller W, et al. (2012). CIC and with Noonan's syndrome develops progressive
2187. Rostomily RC, Bermingham-McDonogh 2203. Rushing EJ, Thompson LD, Mena H FUBP1 mutations in oligodendrogliomas, oli- scoliosis and deteriorating gait. Pediatr Neuro-
0, Berger MS, Tapscott SJ, Reh TA, Olson (2003). Malignant transformation of a dysem- goastrocytomas and astrocytomas. Acta Neuro- surg. 30(1):47-52. PMID:10202309
JM (1997). Expression of neurogenic basic bryoplastic neuroepithelial tumor after radi- pathol. 123(6):853-60. PMID:22588899 2234. Sanford RA, Gajjar A (1997). Epen-
helix-loop-helix genes in primitive neuroecto- ation and chemotherapy. Ann Diagn Pathol. 2220. Sahm F, Reuss D, Koelsche C, Capper D, dymomas. Clin Neurosurg. 44:559-70.
dermal tumors. Cancer Res. 57(16):3526-31. 7(4):240-4. PMID:12913847 Schittenhelm J, Heim S, et al. (2014). Farewell PMID:10080027
PMID:9270024 2204. Russell DS, Rubinstein LJ (1989). Patho- to oligoastrocytoma: in situ molecular genetics 2235. Sangoi AR, Dulai MS, Beck AH, Brat
2188. Roth J, Roach ES, Bartels U, J6iwiak logy of Tumours of the Nervous System. Lon- favor classification as either oligodendroglioma DJ, Vogel H (2009). Distinguishing chordoid
S, Koenig MK, Weiner HL, et al. (2013). Sub- don: Edward Arnold. or astrocytoma. Acta Neuropathol. 128(4):551- meningiomas from their histologic mimics: an
ependymal giant cell astrocytoma: diagnosis, 2205. Russell, DS and Rubinstein, LJ ().Pa- 9. PMID:25143301 immunohistochemical evaluation. Am J Surq
screening, and treatment. Recommendations thology of tumours of the nervous system. 5th 2220A. Sahm F, Schrimpf D, Olar A, Koelsche Pathol. 33(5):669-81. PMID:19194275
from the International Tuberous Sclerosis London: Edward Arnold. C, Reuss D, Bissel J, et al. (2015). TERT 2236. Sankhla S, Khan GM (2004). Cauda
Complex Consensus Conference 2012. Pediatr 2206. Russo C, Pellarin M, Tingby 0, Bollen Promoter Mutations and Risk of Recurrence equina paraganglioma presenting with in-
Neural. 49(6):439-44. PMID:24138953 AW, Lamborn KR, Mahapatra G, et al. (1999). in Meningioma. J Natl Cancer Inst. 108(5). tracranial hypertension: case report and review
2189. Rotman JA, Kucharczyk W, Zadeh G, Comparative genomic hybridization in patients PMID:26668184 of the literature. Neural India. 52(2):243-4.
Kiehl TR, Al-Ahmadi H (2014). Spindle cell with supratentorial and infratentorial primitive 2221. Sainz J, Figueroa K, Baser ME, Mautner PMID 15269482
oncocytoma of the adenohypophysis: a case neuroectodermal tumors. Cancer. 86(2):331-9. VF, Pulst SM (1995). High frequency of nonsen- 2237. Sanoudou D, Tingby 0, Ferguson-Smith
report illustrating its natural history with 8-year PMID:10421270 se mutations in the NF2 gene caused by C to T MA, Collins VP, Coleman N (2000). Analysis of
observation and a review of the literature. Clin 2207. Rutkowski S, Bode U, Deinlein F, Ot- transitions in five CGA codons. Hum Mol Genet. pilocytic astrocytoma by comparative genomic
Imaging. 38(4):499-504. PMID:24721021 tensmeier H, Warmuth-Metz M, Soerensen 4(1):137-9. PMID:7711726 hybridization. Br J Cancer. 82(6):1218-22.
2190. Rouleau GA, Merel P, Lutchman M, San- N, et al. (2005). Treatment of early childhood 2222. Sainz J, Huynh DP, Figueroa K, Ragge PMID:10735509
son M, Zucman J, Marineau C, et al. (1993). medulloblastoma by postoperative chemothe- NK, Baser ME, Pulst SM (1994). Mutations of 2238. Sanson M, Marie Y, Paris S, ldbaih A,
Alteration in a new gene encoding a putative rapy alone. N Engl J Med. 352(10):978-86. the neurofibromatosis type 2 gene and lack of Laffaire J, Ducray F, et al. (2009). lsocitrate
membrane-organizing protein causes neuro-fi- PMID:15758008 the gene product in vestibular schwannomas. dehydrogenase 1 codon 132 mutation is an im-
bromatosis type 2. Nature. 363(6429):515-21. 2208. Rutkowski S, van Hoff K, Emser A, Hum Mol Genet. 3(6):885-91. PMID:7951231 portant prognostic biomarker in gliomas. J Clin
PMID:8379998 Zwiener I, Pietsch T, Figarella-Branger D, et al. 2223. Sakaguchi N, Sano K, Ito M, Baba T, Oneal. 27(25):4150-4. PMID:19636000
2191. Rousseau G, Noguchi T, Bourdon V, (2010). Survival and prognostic factors of early Fukuzawa M, Hotchi M (1996). A case of van 2239. Santagata S, Hornick JL, Ligon KL
Sobol H, Olschwang S (2011). SMARCB1/ childhood medulloblastoma: an international Recklinghausen's disease with bilateral pheo- (2006). Comparative analysis of germ cell
INl1 germline mutations contribute to 10% of meta-analysis. J Clin Oneal. 28(33):4961-8. chromocytoma-malignant peripheral nerve she- transcription factors in CNS germinoma reveals
sporadic schwannomatosis. BMC Neural. 11 :9. PMID:20940197 ath tumors of the adrenal and gastrointestinal diagnostic utility of NANOG. Am J Surg Pathol.
PMID:21255467 2209. Ryken TC, Robinson RA, VanGilder autonomic nerve tumors. Am J Surg Pathol. 30(12):1613-8. PMID:17122519
2192. Roussel MF, Robinson GW (2013). Role JC (1994). Familial occurrence of subepen- 20(7):889-97. PMID8669538 2240. Santagata S, Ligon KL, Hornick JL
of MYC in Medulloblastoma. Cold Spring Harb dymoma. Report of two cases. J Neurosurg. 2224. Sakuta R, Otsubo H, Nolan MA, Weiss (2007). Embryonic stem cell transcription fac-
Perspect Med. 3(11):3. PMID:24186490 80(6):1108-11. PMID:8189269 SK, Hawkins C, Rutka JT, et al. (2005). Recur- tor signatures in the diagnosis of primary and
2193. Roy S, Chu A, Trojanowski JQ, Zhang PJ 2210. Sabbaghian N, Hamel N, Srivastava rent intractable seizures in children with cortical metastatic germ cell tumors. Am J Surg Pathol.
(2005). D2-40, a novel monoclonal antibody A, Albrecht S, Priest JR, Foulkes WD (2012). dysplasia adjacent to dysembryoplastic neu- 31(6):836-45. PMID:17527070
against the M2A antigen as a marker to distin- Germline DICER1 mutation and associated loss roepithelial tumor. J Child Neural. 20(4):377- 2241. Santagata S, Maire CL, ldbaih A, Geffers
guish hemangioblastomas from renal cell car- of heterozygosity in a pineoblastoma. J Med 84. PMID 15921242 L, Correll M, Holton K, et al. (2009). CRX is a di-
cinomas. Acta Neuropathol. 109(5):497-502. Genet. 49(7):417-9. PMID:22717647 2225. Salgado CM, Basu D, Nikiforova M, agnostic marker of retinal and pineal lineage tu-
PMID:15864611 2211. Sachdeva MU, Vankalakunti M, Rangan Bauer BS, Johnson D, Rundell V, et al. (2015). mors. PLoS One. 4(11):e7932. PMID:19936203
2194. Rubin JB, Gutmann DH (2005). Neurofi- A, Radotra BD, Chhabra R, Vasishta RK (2008). BRAF mutations are also associated with neu- 2242. Sarkar C, Sharma MC, Sudha K, Gaik-
bromatosis type 1 - a model for nervous system The role of immunohistochemistry in medul- rocutaneous melanocytosis and large/giant wad S, Varma A (1997). A clinico-pathological
tumour formation? Nat Rev Cancer. 5(7):557- lomyoblastoma-a case series highlighting congenital melanocytic nevi. Pediatr Dev Pa- study of 29 cases of gliosarcoma with special
64. PMID:16069817 divergent differentiation. Diagn Pathol. 3:18. thol. 18(1):1-9. PMID:25490715 reference to two unique variants. Indian J Med
2195. Rubinstein LJ (1986). The malformative PMID:18439235 2226. Salunke PS, Gupta K, Srinivasa R, Sura Res. 106:229-35. PMID:9378529
central nervous system lesions in the central 2212. Sadetzki S, Bruchim R, Oberman B, S (2011). Functional? Paraganglioma of the ce- 2243. Sarkar H, K S, Ghosh S (2013). Pure int-
and peripheral forms of neurofibromatosis. A Armstrong GN, Lau CC, Claus EB, et al.; Glio- rebellum. Acta Neurochir (Wien). 153(7):1527- raventricular origin of gliosarcoma - a rare entity.
neuropathological study of 22 cases. Ann N Y gene Consortium (2013). Description of selec- 8. PMID:21491190 Turk Neurosurg. 23(3):392-4. PMID:23756982
Acad Sci. 486:14-29. PMID:3105387 ted characteristics of familial glioma patients 2227. Salvati M, Ciappetta P, Raco A (1993). 2244. Sarkaria JN, Kitange GJ, James CD,
2196. Rueda-Pedraza ME, Heifetz SA, Sester- - results from the Gliogene Consortium. Eur J Osteosarcomas of the skull. Clinical re- Plummer R, Calvert H, Weller M, et al. (2008).
henn IA, Clark GB (1987). Primary intracranial Cancer. 49(6):1335-45. PMID:23290425 marks on 19 cases. Cancer. 71(7):2210-6. Mechanisms of chemoresistance to alkylating
germ cell tumors in the first two decades of life. 2213. Sadetzki S, Chetrit A, Freedman L, PMID:8453540 agents in malignant glioma. Clin Cancer Res.
A clinical, light-microscopic, and immunohisto- Stovall M, Madan B, Novikov I (2005). Long- 2228. Salvati M, Oppido PA, Artizzu S, Fioren- 14(10):2900-8. PMID:18483356
chemical analysis of 54 cases. Perspect Pediatr term follow-up for brain tumor development za F, Puzzilli F, Orlando ER (1991). Multicen- 2245. Sartoretti-Schefer S, Wichmann W,
Pathol. 10:160-207. PMID:3588245 after childhood exposure to ionizing radiation tric gliomas. Report of seven cases. Tumori. Aguzzi A, Valavanis A (1997). MR differentia-
2197. Ruggieri M, Polizzi A, SpaliceA, Salpietro for tinea capitis. Radial Res. 163(4):424-32. 77(6):518-22. PMID:1666470 tion of adamantinous and squamous-papillary
V, Caltabiano R, D'Orazi V, et al. (2015). The PMID:15799699 2229. Sampson JR, Maheshwar MM, Aspin- craniopharyngiomas. AJNR Am J Neuroradiol.
natural history of spinal neurofibromatosis: a 2214. Saesue P, Chankaew E, Chawalparit 0, wall R, Thompson P, Cheadle JP, Ravine D, 18(1):77-87. PMID:9010523
critical review of clinical and genetic features. Na Ayudhya NS, Muangsomboon S, Sangruchi et al. (1997). Renal cystic disease in tuberous 2246. Sasaki H, Zlatescu MC, Betensky RA,
Clin Genet. 87(5):401-10 PMID:25211147 T (2004 ). Primary extraskeletal osteosarcoma sclerosis: role of the polycystic kidney disea- Ina Y, Cairncross JG, Louis DN (2001 ). PTEN is
2198. Ruland V, Hartung S, Kordes U, Wolff JE, in the pineal region. Case report. J Neurosurg. se 1 gene. Am J Hum Genet. 61(4):843-51. a target of chromosome 1 Oq loss in anaplastic
Paulus W, Hasselblatt M (2014). Choroid ple- 101(6):1061-4 PMID:15597771 PMID:9382094 oligodendrogliomas and PTEN alterations are
xus carcinomas are characterized by complex 2215. Safaee M, Oh MC, Bloch 0, Sun MZ, 2229A. Sampson JR, Scahill SJ, Stephenson associated with poor prognosis. Am J Pathol.
chromosomal alterations related to patient age Kaur G, Auguste Kl, et al. (2013). Choroid ple- JB, Mann L, Connor JM (1989). Genetic aspects 159(1):359-67. PMID:11438483
and prognosis. Genes Chromosomes Cancer. xus papillomas: advances in molecular biology of tuberous sclerosis in the west of Scotland. J 2247. Sato H, Ohmura K, Mizushima M, Ito J,
53(5):373-80. PMID:24478045 and understanding of tumorigenesis. Neuro On- Med Genet. 26(1):28-31 PMID:2918523 Kuyama H (1983). Myxopapillary ependymoma
2199. Rupani A, Modi C, Desai S, Rege J eal. 15(3):255-67. PMID:23172371 2230. Sanai N, Alvarez-Buylla A, Berger of the lateral ventricle. A study on the mecha-
(2005). Primary anaplastic large cell lympho- 2216. Safaee M, Oh MC, Mummaneni PV, MS (2005). Neural stem cells and the origin nism of its stromal myxoid change. Acta Pathol
ma of central nervous system-a case report. Weinstein PR, Ames CP, Chou D, et al. (2014). of gliomas. N Engl J Med. 353(8):811-22. Jpn. 33(5):1017-25. PMID:6359815
J Postgrad Med. 51(4):326-7. PMID:16388180 Surgical outcomes in spinal cord ependymo- PMID:16120861 2248. Sato TS, Kirby PA, Buatti JM, Moritani T
2200. Rushing EJ,Armonda RA.Ansari Q, Mena mas and the importance of extent of resection in 2231. Sancak 0, Nellis! M, Goedbloed M, (2009). Papillary tumor of the pineal region: re-
H (1996). Mesenchymal chondrosarcoma: a children and young adults. J Neurosurg Pediatr. Elfferich P, Wouters C, Maat-Kievit A, et al. port of a rapidly progressive tumor with possible

390 References
multicentric origin. Pediatr Radiol. 39(2):188- 40:159-98. Neuropathol. 29(5):323-9. PMID:20860896 pathological features of breast disease in Cow-
90. PMID:19037636 2266. Scherer HJ (1938). Structural develop- 2284. Schittenhelm J, Roser F, Tatagiba M, Be- den's syndrome: an underrecognized syndrome
2249. Satoh T, Smith A, Sarde A, Lu HC, Mian ment in gliomas. Am J Cancer. 34:333-51. schorner R (2012). Diagnostic value of EAAT-1 with an increased risk of breast cancer. Hum
S, Trouillet C, et al. (2012). B-RAF mutant al- 2267. Scherer HJ (1940). The forms of growth and Kir7 .1 for distinguishing endolymphatic sac Pathol. 29(1):47-53. PMID:9445133
leles associated with Langerhans cell histiocy- in gliomas and their practical significance. tumors from choroid plexus tumors. Am J Clin 2299. Schroers R, Baraniskin A, Heute C, Vor-
tosis, a granulomatous pediatric disease. PLoS Brain. 63:1-35. Pathol. 138(1):85-9. PMID:22706862 gerd M, Brunn A, Kuhnhenn J, et al. (2010).
One. 7(4):e33891. PMID:22506009 2268. Scheurlen WG, Schwabe GC, Joos S, 2285. Schlamann A, von Bueren AO, Hagel C, Diagnosis of leptomeningeal disease in diffuse
2250. Sawamura Y, Hamou MF, Kuppner MC, Mollenhauer J, Sorensen N, Kuhl J (1998). Mo- Zwiener I, Seidel C, Kortmann RD, et al. (2014). large B-cell lymphomas of the central nervous
de Tribolet N (1989). lmmunohistochemical and lecular analysis of childhood primitive neuroec- An individual patient data meta-analysis on system by flow cytometry and cytopathology.
in vitro functional analysis of pineal-germino- todermal tumors defines markers associated characteristics and outcome of patients with pa- Eur J Haematol. 85(6):520-8. PMID:20727005
ma infiltrating lymphocytes: report of a case. with poor outcome. J Clin Oncol. 16(7):2478- pillary glioneuronal tumor, rosette glioneuronal 2300. Schuettpelz LG, McDonald S, Whitesell
Neurosurgery. 25(3):454-7, discussion 457-8. 85. PMID:9667267 tumor with neuropil-like islands and rosette for- K, Desruisseau DM, Grange DK, Gurnett CA,
PMID:2671789 2269. Schiariti M, Goetz P, EI-Maghraby H, ming glioneuronal tumor of the fourth ventricle. et al. (2009). Pilocytic astrocytoma in a child
2251. Sawamura Y, Ikeda J, Shirato H, Tada Tailor J, Kitchen N (2011). Hemangiopericyto- PLoS One. 9(7):e101211. PMID:24991807 with Noonan syndrome. Pediatr Blood Cancer.
M, Abe H (1998). Germ cell tumours of the ma: long-term outcome revisited. Clinical article. 2286. Schlegel U (2009). Primary CNS lym- 53(6):1147-9. PMID:19621452
central nervous system: treatment considera- J Neurosurg. 114(3):747-55. PMID:20672899 phoma. Ther Adv Neurol Disord. 2(2):93-104. 2301. Schumacher T, Bunse L, Pusch S,
tion based on 111 cases and their long-term 2270. Schiefer Al, Vastagh I, Molnar MJ, Be- PMID:21180644 Sahm F, Wiestler B, Quandt J, et al. (2014).
clinical outcomes. Eur J Cancer. 34(1):104-10. reczki D, Varallyay G, Deak B, et al. (2012). Ex- 2287. Schmalisch K, Beschorner R, Psaras T. A vaccine targeting mutant IDH1 induces an-
PMID 9624246 tranodal marginal zone lymphoma of the CNS Honegger J (2010). Postoperative intracranial titumour immunity. Nature. 512(7514):324-7.
2252. Sawin PD, Theodore N, Rekate HL arising after a long-standing history of atypical seeding of craniopharyngiomas-report of three PMID:25043048
(1999). Spinal cord ganglioglioma in a child with white matter disease. Leuk Res. 36(7):e155-7. cases and review of the literature. Acta Neu- 2302. Schuss P, Ulrich CT. Harter PN, Tews
neurofibromatosis type 2. Case report and lite- PMID:22520340 rochir (Wien). 152(2):313-9, discussion 319. DS, Seifert V, Franz K (2011). Gliosarcoma with
rature review. J Neurosurg. 90(2) Suppl:231-3. 2271. Schiff D, O'Neill B, Wijdicks E, Antin JH, PMID:19859655 bone infiltration and extracranial growth: case
PMID:10199253 Wen PY (2001 ). Gliomas arising in organ trans- 2288. Schmidbauer M, Budka H, Pilz P (1989). report and review of literature. J Neurooncol.
2253. Sawyer JR, Roloson GJ, Chadduck WM, plant recipients: an unrecognized complication Neuroepithelial and ectomesenchymal diffe- 103(3):765-70. PMID:20957407
Boop FA (1991). Cytogenetic findings in a ple- of transplantation? Neurology. 57(8):1486-8. rentiation in a primitive pineal tumor ("pineal 2303. Schwartz AM, Ghatak NR (1990). Mali-
omorphic xanthoastrocytoma. Cancer Genet PMID:11673595 anlage tumor"). Clin Neuropathol. 8(1):7-10. gnant transformation of benign cerebellar ast-
Cytogenet. 55(2):225-30. PMID:1933824 2272. Schiffer D, Cavalla P, Migheli A, Chio A, PMID:2650944 rocytoma. Cancer. 65(2):333-6. PMID:2403835
2254. Sawyer JR, Thomas EL, Roloson GJ, Giordana MT, Marino S, et al. (1995). Apopto- 2289. Schmidt MC, Antweiler S, Urban N, Mu- 2304. Schwartzentruber J, Korshunov A, Liu
Chadduck WM, Boop FA (1992). Telomeric sis and cell proliferation in human neuroepit- eller W, Kuklik A, Meyer-Puttlitz B, et al. (2002). XY, Jones DT. Pfaff E, Jacob K, et al. (2012).
associations evolving to ring chromosomes helial tumors. Neurosci Lett. 195(2):81-4. Impact of genotype and morphology on the Driver mutations in histone H3.3 and chromatin
in a recurrent pleomorphic xanthoastrocyto- PMID:7478273 prognosis of glioblastoma. J Neuropathol Exp remodelling genes in paediatric glioblastoma.
ma. Cancer Genet Cytogenet. 60(2):152-7. 2273. Schiffer D, Chio A, Giordana MT. Leone Neurol. 61(4):321-8. PMID:11939587 Nature. 482(7384):226-31. PMID:22286061
PMID:1606558 M, Soffietti R (1988). Prognostic value of his- 2290. Schmidt Y, Kleinschmidt-DeMasters BK, 2305. Schwartzkroin PA, Wenzel HJ (2012).
2254A. Schaefer IM, Fletcher CD, Hornick JL tologic factors in adult cerebral astrocytoma. Aisner DL, Lillehei KO, Damek D (2013). Ana- Are developmental dysplastic lesions epi-
(2016). Loss of H3K27 trimethylation distingu- Cancer. 61(7):1386-93. PMID:3345492 plastic PXA in adults: case series with clinicopa- leptogenic? Epilepsia. 53 Suppl 1 :35-44.
ishes malignant peripheral nerve sheath tumors 2274. Schiffer D, Chio A, Giordana MT, Migheli thologic and molecular features. J Neurooncol. PMID:22612807
from histologic mimics. Mod Pathol.29(1):4-13. A, Palma L, Pollo B, et al. (1991). Histologic 111(1):59-69. PMID:23096133 2306. Schwechheimer K, Huang S, Cavenee
PMID:26585554 prognostic factors in ependymoma. Childs Nerv 2291. Schmitz U, Mueller W, Weber M, Se- WK (1995). EGFR gene amplification-rearran-
2256. Scheie D, Meling TR, Cvancarova M, Syst. 7(4):177-82. PMID:1933913 venet N, Delattre 0, von Deimling A (2001). gement in human glioblastomas. Int J Cancer.
Skullerud K, M0rk S, Lote K, et al. (2011). Pro- 2275. Schiffer D, Giordana MT (1998). Pro- INl1 mutations in meningiomas at a potential 62(2):145-8. PMID:7622287
gnostic variables in oligodendroglial tumors: a gnosis of ependymoma. Childs Nerv Syst. hotspot in exon 9. Br J Cancer. 84(2):199-201. 2307. Schweizer L, Capper D, Holsken A, Fahl-
single-institution study of 95 cases. Neuro On- 14(8)357-61. PMID:9753400 PMID:11161377 busch R, Flitsch J, Buchfelder M, et al. (2015).
eel. 13(11):1225-33. PMID:21856683 2276. Schiffer D, Giordana MT, Mauro A, Mig- 2292. Schneider DT, Zahn S, Sievers S, Ale- BRAF V600E analysis for the differentiation
2257. Scheithauer BW (1978). Symptomatic heli A (1984). GFAP, F VIII/RAg, laminin, and fi- mazkour K, Reifenberger G, Wiestler OD, et of papillary craniopharyngiomas and Rath-
subependymoma. Report of 21 cases with re- bronectin in gliosarcomas: an immunohistoche- al. (2006). Molecular genetic analysis of central ke's cleft cysts. Neuropathol Appl Neurobiol.
view of the literature. J Neurosurg. 49(5):689- mical study. Acta Neuropathol. 63(2):108-16. nervous system germ cell tumors with com- 41(6):733-42 PMID:25442675
96. PMID:712391 PMID:6428154 parative genomic hybridization. Mod Pathol. 2308. Schweizer L, Koelsche C, Sahm F, Piro
2258. Scheithauer BW (1999). Pathobiology of 2277. Schiffman JD, Hodgson JG, VandenBerg 19(6):864-73. PMID:16607373 RM, Capper D, Reuss DE, et al. (2013). Menin-
the pineal gland with emphasis on parenchy- SR, Flaherty P, Polley MY, Yu M, et al. (2010). 2293. Schneppenheim R, Fruhwald MC, Gesk geal hemangiopericytoma and solitary fibrous
mal tumors. Brain Tumor Pathol. 16(1):1-9. Oncogenic BRAF mutation with CDKN2A S, Hasselblatt M, Jeibmann A, Kordes U, et tumors carry the NAB2-STAT6 fusion and can
PMID:10532417 inactivation is characteristic of a subset of pe- al. (2010). Germline nonsense mutation and be diagnosed by nuclear expression of STAT6
2259. Scheithauer BW, Amrami KK, Folpe AL, diatric malignant astrocytomas. Cancer Res. somatic inactivation of SMARCA4/BRG 1 in protein. Acta Neuropathol. 125(5):651-8.
Silva Al, Edgar MA, Woodruff JM, et al. (2011). 70(2):512-9. PMID:20068183 a family with rhabdoid tumor predisposition PMID:23575898
Synovial sarcoma of nerve. Hum Pathol. 2278. Schild SE, Scheithauer BW, Haddock syndrome. Am J Hum Genet. 86(2):279-84. 2309. Schwindt H, Valer I, Kreuz M, Monte-
42(4):568-77. PMID:21295819 MG, Wong WW, Lyons MK, Marks LB, et al. PMID:20137775 sinos-Rongen M, Brunn A, Richter J, et al.
2260. Scheithauer BW, Erdogan S, Rodriguez (1996). Histologically confirmed pineal tumors 2294. Schniederjan MJ, Alghamdi S, Castel- (2009). Chromosomal imbalances and partial
FJ, Burger PC, Woodruff JM, Kros JM, et al. and other germ cell tumors of the brain. Cancer. lano-Sanchez A, Mazewski C, Brahma B, uniparental disomies in primary central nervous
(2009). Malignant peripheral nerve sheath 78(12):2564-71. PMID:8952565 Brat DJ, et al. (2013). Diffuse leptomeningeal system lymphoma. Leukemia. 23(10):1875-84.
tumors of cranial nerves and intracranial cont- 2279. Schild SE, Scheithauer BW, Schomberg neuroepithelial tumor: 9 pediatric cases with PMID:19494841
ents: a clinicopathologic study of 17 cases. Am PJ, Hook CC, Kelly PJ, Frick L, et al. (1993). Pi- chromosome 1p/19q deletion status and IDH1 2310. Schuller U, Heine VM, Mao J, Kho AT,
J Surg Pathol. 33(3):325-38. PMID:19065105 neal parenchymal tumors. Clinical, pathologic, (R132H) immunohistochemistry. Am J Surg Pa- Dillon AK, Han YG, et al. (2008). Acquisition of
2261. Scheithauer BW, Horvath E, Kovacs and therapeutic aspects. Cancer. 72(3):870-80. thol. 37(5):763-71. PMID:23588371 granule neuron precursor identity is a critical
K (1992). Ultrastructure of the neurohypo- PMID:8334641 2294A. Schoemaker MJ, Swerdlow AJ, determinant of progenitor cell competence to
physis. Microsc Res Tech. 20(2):177-86. 2280. Schindler G, Capper D, Meyer J, Janzarik Hepworth SJ, van Tongeren M, Muir KR, form Shh-induced medulloblastoma. Cancer
PMID:1547358 W, Omran H, Herold-Mende C, et al. (2011). McKinney PA (2007). History of allergic disea- Cell. 14(2):123-34. PMID:18691547
2262. Scheithauer BW, Rubinstein LJ (1978). Analysis of BRAF V600E mutation in 1,320 se and risk of meningioma. Am J Epidemiol. 2311. Sciot R, Dal Cin P, Hagemeijer A, De
Meningeal mesenchymal chondrosarcoma: nervous system tumors reveals high mutation 165(5):477-85. PMID:17182979 Smet L, Van Damme B, Van den Berghe H
report of 8 cases with review of the literature. frequencies in pleomorphic xanthoastrocytoma, 2295. Schoenberg BS, Schoenberg DG, Chris- (1999). Cutaneous sclerosing perineurioma
Cancer. 42(6):2744-52. PMID:365318 ganglioglioma and extra-cerebellar pilocytic as- tine BW, Gomez MR (1976). The epidemiology with cryptic NF2 gene deletion. Am J Surg Pa-
2263. Scheithauer BW, Silva Al, Ketterling trocytoma. Acta Neuropathol. 121(3):397-405. of primary intracranial neoplasms of childhood. thol. 23(7):849-53. PMID:10403310
RP, Pula JH, Lininger JF, Krinock MJ (2009). PMID:21274720 A population study. Mayo Clin Proc. 51(1):51-6. 2312. Scott RM, Ballantine HT Jr (1973). Cere-
Rosette-forming glioneuronal tumor: report 2281. Schittenhelm J, Mittelbronn M, Nguyen PMID:1249998 bellar astrocytoma: malignant recurrence after
of a chiasmal-optic nerve example in neuro- TD, Meyermann R, Beschorner R (2008). WT1 2296. Schofield D, West DC, Anthony DC, prolonged postoperative survival. Case report.
fibromatosis type 1: special pathology report. expression distinguishes astrocytic tumor cells Marshal R, Sklar J (1995). Correlation of loss J Neurosurg. 39(6):777-9. PMID:4759666
Neurosurgery. 64(4):E771-2, discussion E772. from normal and reactive astrocytes. Brain Pa- of heterozygosity at chromosome 9q with his- 2313. Seiz M, Tuettenberg J, Meyer J, Essig M,
PMID:193498062264 thol. 18(3):344-53. PMID:18371184 tological subtype in medulloblastomas. Am J Schmieder K, Mawrin C, et al. (2010). Detection
2264. Schenk PW, Van Es S, Kesbeke F, 2282. Schittenhelm J, Mittelbronn M, Wolff M, Pathol. 146(2):472-80. PMID:7856756 of IDH1 mutations in gliomatosis cerebri, but
Snaar-Jagalska BE (1991). Involvement of Truebenbach J, Will BE, Meyermann R, et al. 2297. Schouten LJ, Rutten J, Huveneers HA, only in tumors with additional solid component:
cyclic AMP cell surface receptors and G-pro- (2007). Multifocal dysembryoplastic neuroepit- Twijnstra A (2002). Incidence of brain metasta- evidence for molecular subtypes. Acta Neuro-
teins in signal transduction during slug migra- helial tumor with signs of atypia after regrowth. ses in a cohort of patients with carcinoma of the pathol. 120(2):261-7. PMID:20514489
tion of Dictyostelium discoideum. Dev Biol. Neuropathology. 27(4):383-9. PMID:17899694 breast, colon, kidney, and lung and melanoma. 2314. Seizinger BR, Martuza RL, Gusella JF
145(1):110-8. PMID:1850366 2283. Schittenhelm J, Psaras T (2010). Gli- Cancer. 94(10):2698-705. PMID:12173339 (1986). Loss of genes on chromosome 22 in
2265. Scherer H-J (1940). Cerebral astrocyto- oblastoma with granular cell astrocytoma fea- 2298. Schrager CA, Schneider D, Gruener AC, tumorigenesis of human acoustic neuroma. Na-
mas and their derivatives. Am J Cancer. tures: a case report and literature review. Clin Tsou HC, Peacocke M (1998). Clinical and ture. 322(6080):644-7. PMID:3092103

References 391
2315. Seizinger BR, Rouleau GA, Ozelius LJ, Majounie E, Shenton A, Baser M, et al. (2011). multiforme. J Neurosurg. 101(2):219-26. (2012). Catecholamine-secreting paraganglio-
Lane AH, Faryniarz AG, Chao MV, et al. (1987). A molecular analysis of individuals with neuro- PMID:15309911 ma of the thoracic spinal column: report of an
Genetic linkage of von Recklinghausen neurofi- fibromatosis type 1 (NF1) and optic pathway 2348. Shinojima N, Ohta K, Yano S, Nakamura unusual case and review of the literature. Neu-
bromatosis to the nerve growth factor receptor gliomas (OPGs), and an assessment of ge- H, Kochi M, lshimaru Y, et al. (2002). Myofibrob- rosurgery. 70(4):E1049-52, discussion E1052.
gene. Cell. 49(5):589-94. PMID:2884037 notype-phenotype correlations. J Med Genet. lastoma in the suprasellar region. Case report. PMID:21788916
2316. Sekine S, Shibata T, Kokubu A, Morishita 48(4):256-60. PMID:21278392 J Neurosurg. 97(5):1203-7. PMID:12450045 2364. Singh D, Chan JM, Zoppoli P, Niola F
Y, Noguchi M, Nakanishi Y, et al. (2002). Crani- 2333. Sharma M, Ralte A, Arora R, Santosh V, 2349. Shintaku M, Yoneda H, Hirata J, Nagaishi Sullivan R, Castano A, et al. (2012). Transfor'.
opharyngiomas of adamantinomatous type har- Shankar SK, Sarkar C (2004). Subependymal M, Okabe H (2013). Gliosarcoma with ependy- ming fusions of FGFR and TACC genes in hu-
bor beta-catenin gene mutations. Am J Pathol. giant cell astrocytoma: a clinicopathological mal and PNET-like differentiation. Clin Neuro- man glioblastoma. Science. 337(6099):1231-5.
161(6):1997-2001. PMID:12466115 study of 23 cases with special emphasis on pathol. 32(6):508-14. PMID 23863343 PMID22837387
2317. Sekine S, Takata T, Shibata T. Mori M, proliferative markers and expression of p53 2350. Shishiba T. Niimura M, Ohtsuka F, Tsuru 2365. Singh SK, Hawkins C, Clarke ID, Squire
Morishita Y, Noguchi M, et al. (2004). Expres- and retinoblastoma gene proteins. Pathology. N (1984). Multiple cutaneous neurilemmomas JA, Bayani J, Hide T, et al. (2004). Identification
sion of enamel proteins and LEF1 in adaman- 36(2):139-44. PMID:15203749 as a skin manifestation of neurilemmomato- of human brain tumour initiating cells. Nature.
tinomatous craniopharyngioma: evidence for its 2334. Sharma MC, Ralte AM, Gaekwad S, sis. J Am Acad Dermatol. 10(5 Pt 1):744-54. 432(7015):396-401. PMID:15549107
odontogenic epithelial differentiation. Histopa- Santosh V, Shankar SK, Sarkar C (2004). PMID:6427303 2366. Sinicrope FA, Sargent DJ (2012). Mo-
thology. 45(6):573-9. PMID:15569047 Subependymal giant cell astrocytoma-a clini- 2351. Shiurba RA, Buffinger NS, Spencer EM, lecular pathways: microsatellite instability in
2318. Sener RN (2002). Astroblastoma: dif- copathological study of 23 cases with special Urich H {1991). Basic fibroblast growth factor colorectal cancer: prognostic, predictive, and
fusion MRI, and proton MR spectroscopy. emphasis on histogenesis. Pathol Oneal Res. and somatomedin C in human medulloepithe- therapeutic implications. Clin Cancer Res.
Comput Med Imaging Graph. 26(3):187-91. 10(4):219-24. PMID:15619643 lioma. Cancer. 68(4):798-808. PMID:1855180 18(6):1506-12. PMID:22302899
PMID:11918982 2335. Sharma MK, Mansur DB, Reifenberger G, 2352. Shively SB, Falke EA, Li J, Tran MG, 2367. Sinson G, Gennarelli TA, Wells GB
2319. Senft C, Raabe A. Hattingen E. Sommer- Perry A, Leonard JR, Aldape KD, et al. (2007). Thompson ER, Maxwell PH, et al. (2011). De- (1998). Suprasellar osteolipoma: case report.
lad D, Seifert V, Franz K (2008). Pineal paren- Distinct genetic signatures among pilocytic as- velopmentally arrested structures preceding Surg Neural. 50(5):457-60. PMID:9842872
chymal tumor of intermediate differentiation: trocytomas relate to their brain region origin. cerebellar tumors in van Hippel-Lindau disease. 2368. Skardelly M, Pantazis G, Bisdas S,
diagnostic pitfalls and discussion of treatment Cancer Res. 67(3):890-900. PMID:17283119 Mod Pathol. 24(8):1023-30. PMID:21499240 Feigl GC, Schuhmann MU, Tatagiba MS, et
options of a rare tumor entity. Neurosurg Rev. 2336. Sharpless NE, Bardeesy N, Lee KH, Car- 2353. Shlien A, Campbell BB, de Borja R, al. (2013). Primary cerebral low-grade B-cell
31(2):231-6. PMID 18266015 rasco D, Castrillon DH, AguirreAJ, et al. (2001). Alexandrov LB, Merica D, Wedge D, et al.; lymphoma, monoclonal immunoglobulin depo-
2320. SenGupta SK, Webb S, Cooke RA, Loss of p161nk4a with retention of p19Arf Biallelic Mismatch Repair Deficiency Consorti- sition disease, cerebral light chain deposition
Igo JD (1992). Breast filariasis diagnosed by predisposes mice to tumorigenesis. Nature. um (2015). Combined hereditary and somatic disease and "aggregoma": an update on clas-
needle aspiration cytology. Diagn Cytopathol. 413(6851):86-91. PMID:11544531 mutations of replication error repair genes result sification and diagnosis. BMC Neural. 13:107.
8(4):392-3. PMID:1638940 2337. Shaw EG, Scheithauer BW, O'Fallon JR, in rapid onset of ullra-hypermutated cancers. PMID:23947787
2321. Seal HJ, Hwang SK, Choi YL, Chi JG, Tazelaar HD, Davis DH (1992). Oligodendro- Nat Genet. 47(3):257-62. PMID:25642631 2369. Skullerud K, Stenwig AE, Brandtzaeg P,
Jung HW (2003). A case of recurrent sub- gliomas: the Mayo Clinic experience. J Neuro- 2354. Short MP, Richardson EP Jr, Haines JL, Nesland JM, Kerty E, Langmoen I, et al. (1995).
ependymoma with subependymal seeding: surg. 76(3):428-34. PMID:1738022 Kwiatkowski DJ (1995). Clinical, neuropatho- lntracranial primary leiomyosarcoma arising in
case report. J Neurooncol. 62(3):315-20. 2338. Shaw EG, Wang M, Coons SW, Brach- logical and genetic aspects of the tuberous a teratoma of the pineal area. Clin Neuropathol.
PMID: 12777084 man DG, Buckner JC, Stelzer KJ, et al. (2012). sclerosis complex. Brain Pathol. 5(2):173-9. 14(4):245-8. PMID:8521631
2322. Seppala MT, Sainio MA, Haltia MJ, Randomized trial of radiation therapy plus pro- PMID:7670658 2370. Slowik F, Jellinger K, Gasz6 L, Fischer J
Kinnunen JJ, Setala KH, Jaaskelainen JE carbazine, lomustine, and vincristine chemothe- 2355. Shau Y, Robinson DM, Amakye DD, {1985). Gliosarcomas: histological, immunohis-
(1998). Multiple schwannomas: schwannomat- rapy for supratentorial adult low-grade glioma: Rose KL, Cho YJ, Ligon KL, et al. (2015). A tochemical, ultrastructural, and tissue culture
osis or neurofibromatosis type 2? J Neurosurg. initial results of RTOG 9802. J Clin Oneal. five-gene hedgehog signature developed as a studies. Acta Neuropathol. 67(3-4):201-10.
89(1):36-41. PMID:9647170 30(25):3065-70. PMID:22851558 patient preselection tool for hedgehog inhibitor PMID:4050334
2323. Serracino HS, Kleinschmidt-Demasters 2339. Shaw RJ, Paez JG, Curto M, Yaktine therapy in medulloblastoma. Clin Cancer Res. 2371. Smedby KE, Brandt L, Backlund ML,
BK (2013). Skull invaders: when surgical pa- A, Pruitt WM, Saotome I, et al. (2001). The 21(3):585-93. PMID:25473003 Blomqvist P (2009). Brain metastases admis-
thology and neuropathology worlds collide. Nf2 tumor suppressor, merlin, functions in 2356. Shuangshoti S, Rushing EJ, Mena H. sions in Sweden between 1987 and 2006. Br
J Neuropathol Exp Neural. 72(7):600-13. Rae-dependent signaling. Dev Cell. 1(1):63-72. Olsen C, Sandberg GD (2005). Supratentori- J Cancer. 101(11):1919-24. PMID:19826419
PMID:23771219 PMID:11703924 al extraventricular ependymal neoplasms: a 2372. Smith AB, Rushing EJ, Smirniotopoulos
2324. Serrano M, Lin AW, McCurrach ME, 2340. Shen WH, Balajee AS, Wang J, Wu H, clinicopathologic study of 32 patients. Cancer. JG (2009). Pigmented lesions of the cen-
Beach D, Lowe SW (1997). Oncogenic ras pro- Eng C, Pandolfi PP, et al. (2007). Essential role 103(12):2598-605. PMID:15861411 tral nervous system: radiologic-pathologic
vokes premature cell senescence associated for nuclear PTEN in maintaining chromosomal 2357. Shweiki D, ltin A, Soffer D, Keshet E correlation. Radiographies. 29(5):1503-24.
with accumulation of p53 and p161NK4a. Cell. integrity. Cell. 128(1):157-70. PMID:17218262 (1992). Vascular endothelial growth factor PMID:19755608
88(5):593-602. PMID:9054499 2341. Shenkier TN, Blay JY, O'Neill BP, Poortm- induced by hypoxia may mediate hypoxia-initi- 2373. Smith AB, Rushing EJ, Smirniotopoulos
2325. Sestini R, Bacci C, Provenzano A, Ge- ans P, Thiel E, Jahnke K, et al. (2005). Primary ated angiogenesis. Nature. 359(6398):843-5. JG (2010). From the archives of the AFIP: le-
nuardi M, Papi L (2008). Evidence of a four-hit CNS lymphoma of T-cell origin: a descriptive PMID:1279431 sions of the pineal region: radiologic-pathologic
mechanism involving SMARCB1 and NF2 in analysis from the international primary CNS 2358. Siami-Namini K, Shuey-Drake R, Wilson correlation. Radiographies. 30(7):2001-20.
schwannomatosis-associated schwannomas. lymphoma collaborative group. J Clin Oneal. D, Francel P, Perry A, Fung KM (2005). A 15-ye- PMID:21057132
Hum Mutat. 29(2):227-31. PMID:18072270 23(10):2233-9. PMID:15800313 ar-old female with progressive myelopathy. 2374. Smith JS, Tachibana I, Passe SM, Hunt-
2326. Setzer M, Lang J, Turowski B, Marquardt 2342. Shepherd CW, Houser OW, Gomez MR Brain Pathol. 15(3):265-7. PMID:16196395 ley BK, Borell TJ, llurria N, et al. (2001). PTEN
G (2002). Primary meningeal osteosarco- (1995). MR findings in tuberous sclerosis com- 2359. Sievers S, Alemazkour K, Zahn S, Perl- mutation, EGFR amplification, and outcome
ma: case report and review of the literature. plex and correlation with seizure development man EJ, Gillis AJ, Looijenga LH, et al. (2005). in patients with anaplastic astrocytoma and
Neurosurgery. 51(2):488-92, discussion 492. and mental impairment. AJNR Am J Neurora- IGF2/H19 imprinting analysis of human germ glioblastoma mulliforme. J Natl Cancer Inst.
PMID:12182789 diol. 16(1):149-55. PMID:7900584 cell tumors (GCTs) using the methylation-sen- 93(16):1246-56. PMID:11504770
2327. Sevenet N, Sheridan E, Amram D, 2343. Shibahara J, Fukayama M (2005). Se- sitive single-nucleotide primer extension me- 2375. Smith MJ (2015). Germline and somatic
Schneider P, Handgretinger R, Delattre O condary glioblastoma with advanced neuronal thod reflects the origin of GCTs in different mutations in meningiomas. Cancer Genet.
(1999). Constitutional mutations of the hSNF5/ immunophenotype. Virchows Arch. 447(3):665- stages of primordial germ cell development. 208(4):107-14. PMID:25857641
INl1 gene predispose toa variety of cancers.Am 8. PMID:15968544 Genes Chromosomes Cancer. 44(3):256-64. 2376. Smith MJ, Beetz C, Williams SG, Bhas-
J Hum Genet. 65(5):1342-8. PMID:10521299 2344. Shibahara J, Toda T. Morita l\, Mori H, PMID:16001432 kar SS, O'Sullivan J, Anderson B, et al. (2014).
2328. Shankar GM, Chen L, Kim AH, Ross GL, Aoki S, Fukayama M (2004). Papillary neu- 2360. Sievert AJ, Jackson EM, Gai X, Hakonar- Germline mutations in SUFU cause Gorlin syn-
Folkerth RD, Friedlander RM (2010). Composi- roepithelial tumor of the pineal region. A case son H, Judkins AR, Resnick AC, et al. (2009). drome-associated childhood medulloblastoma
te ganglioneuroma-paraganglioma of the filum report. Acta Neuropathol. 108(4):337-40. Duplication of 7q34 in pediatric low-grade and redefine the risk associated with PTCH 1
terminale. J Neurosurg Spine. 12(6):709-13. PMID:15221340 astrocytomas detected by high-density sing- mutations. J Clin Oneal. 32(36):4155-61.
PMID:20515359 2345. Shih DJ, Northcott PA, Remke M, Korshu- le-nucleotide polymorphism-based genotype PMID:25403219
2329. Shankar GM, Taylor-Weiner A, Lelic N, nov A, Ramaswamy V, Kool M, et al. (2014). arrays results in a novel BRAF fusion gene. 2377. Smith MJ, Isidor B, Beetz C, Williams SG,
Jones RT, Kim JC, Francis JM, et al. (2014). Cytogenetic prognostication within medullob- Brain Pathol. 19(3):449-58. PMID: 190167 43 Bhaskar SS. Richer W, et al. (2015). Mutations
Sporadic hemangioblastomas are characteri- lastoma subgroups. J Clin Oneal. 32(9):886-96. 2361. Simon SL, Moonis G, Judkins AR, Scobie in LZTR1 add to the complex heterogeneity of
zed by cryptic VHL inactivation. Acta Neuropa- PMID:24493713 J, Burnett MG, Riina HA, et al. (2005). lntracra- schwannomatosis. Neurology. 84(2):141-7.
thol Commun. 2:167. PMID:25589003 2346. Shimbo Y, Takahashi H, Hayano M, Ku- nial capillary hemangioma: case report and re- PMID:25480913
2330. Shanklin WM (1953). The origin, histolo- magai T, Kameyama S (1997). Temporal lobe view of the literature. Surg Neural. 64(2):154-9. 2378. Smith MJ, Kulkarni A, Rustad C, Bowers
gy and senescence of tumorettes in the human lesion demonstrating features of dysembryopla- PMID:16051010 NL, Wallace AJ, Holder SE, et al. (2012). Vesti-
neurohypophysis. Acta Anal (Basel). 18(1):1- stic neuroepithelial tumor and ganglioglioma: a 2362. Simoneau-Roy J, O'Gorman C, Pencharz bular schwannomas occur in schwannomatosis
20. PMID:13064969 transitional form? Clin Neuropathol. 16(2):65-8. P, Adeli K, Daneman D, Hamilton J (2010). In- and should not be considered an exclusion cri-
2331. Shanley S, Ratcliffe J, Hockey A, Haan PMID:9101106 sulin sensitivity and secretion in children and terion for clinical diagnosis. Am J Med Genet A.
E, Oley C, Ravine D, et al. (1994). Nevoid basal 2347. Shinojima N, Kochi M, Hamada J, Naka- adolescents with hypothalamic obesity follo- 158A(1):215-9. PMID:22105938
cell carcinoma syndrome: review of 118 affec- mura H, Yano S, Makino K, et al. (2004). The wing treatment for craniopharyngioma. Clin En- 2379. Smith MJ, Wallace AJ, Bennett C, Has-
ted individuals. Am J Med Genet. 50(3):282-90. influence of sex and the presence of giant docrinol (OxD. 72(3):364-70. PMID:19486023 selblatt M, Elert-Dobkowska E, Evans LT, et
PMID:8042673 cells on postoperative long-term survival in 2363. Simpson LN, Hughes BO, Karikari 10, al. (2014). Germline SMARCE1 mutations
2332. Sharif S, Upadhyaya M, Ferner R, adult patients with supratentorial glioblastoma Mehta Al, Hodges TR, Cummings T J, et al. predispose to both spinal and cranial clear

392 References
cell meningiomas. J Pathol. 234(4):436-40. than primary glioblastoma. Int J Clin Exp Pathol. Neuro Oneal. 16(2):256-64. PMID 24305709 Pediatr. 9(6):646-53. PMID:22656257
PMID:25143307 4(7):651-60. PMID:22076165 2411. Stambolic V, Suzuki A, de la Pompa JL, 2427. Stern J, Jakobiec FA, Housepian EM
2380. Smith MJ, Wallace AJ, Bowers NL, Ea- 2395. Sonneland PR, Scheithauer BW, Brothers GM, Mirtsos C, Sasaki T, et al. (1998). (1980). The architecture of optic nerve gliomas
ton H, Evans DG (2014). SMARCB1 mutations LeChago J, Crawford BG, Onofrio BM (1986). Negative regulation of PKB/Akt-dependent cell with and without neurofibromatosis. Arch Opht-
in schwannomatosis and genotype correla- Paraganglioma of the cauda equina region. survival by the tumor suppressor PTEN. Cell. halmol. 98(3):505-11. PMID:6767467
tions with rhabdoid tumors. Cancer Genet. Clinicopathologic study of 31 cases with special 95(1):29-39. PMID:9778245 2428. Stockhammer F, Misch M, Helms HJ,
207(9):373-8. PMID:24933152 reference to immunocytology and ultrastruc- 2412. Stanescu Casson R, Varlet P, Beuvon F, Lengler U, Prall F, von Deimling A, et al. (2012).
2381. Smith MJ, Wallace AJ, Bowers NL, ture. Cancer. 58(8):1720-35. PMID:2875784 Daumas Duport C, Devaux B, Chassoux F, et IDH1/2 mutations in WHO grade II astrocyto-
Rustad CF, Woods CG, Leschziner GD, et al. 2396. Sonneland PR, Scheithauer BW, Onof- al. (2001 ). Dysembryoplastic neuroepithelial tu- mas associated with localization and seizure
(2012). Frequency of SMARCB1 mutations in rio BM (1985). Myxopapillary ependymoma. mors: CT, MR findings and imaging follow-up: a as the initial symptom. Seizure. 21(3):194-7.
familial and sporadic schwannomatosis. Neuro- A clinicopathologic and immunocytochemical study of 53 cases. J Neuroradiol. 28(4):230-40. PMID:22217666
genetics. 13(2):141-5. PMID:22434358 study of 77 cases. Cancer. 56(4):883-93. PMID:11924137 2429. Stoffel EM, Mangu PB, Gruber SB, Ha-
2382. Small NR, Drummond KJ (2012). The in- PMID:4016681 2413. Stangl AP, Wellenreuther R, Lenartz D, milton SR, Kalady MF, Lau MW, et al.; American
cidence of medulloblastomas and primitive neu- 2397. Sonoda Y, Yoshimoto T, Sekiya T (1995). Kraus JA, Menon AG, Schramm J, et al. (1997). Society of Clinical Oncology; European Society
rectodermal tumours in adults and children. J Homozygous deletion of the MTS1/p16 and Clonality of multiple meningiomas. J Neurosurg. of Clinical Oncology (2015). Hereditary colo-
Clin Neurosci. 19(11):1541-4. PMID:22981874 MTS2/p15 genes and amplification of the CDK4 86(5):853-8. PMID:9126902 rectal cancer syndromes: American Society of
2383. Smolle E, AI-Qubati S, Stefanits H, Ha- gene in glioma. Oncogene. 11(10):2145-9. 2414. Starink TM, van der Veen JP, Arwert F, de Clinical Oncology Clinical Practice Guideline
berler C, Kleinert R, Haybaeck J (2012). Me- PMID:7478535 Waal LP, de Lange GG, Gille JJ, et al. (1986). endorsement of the familial risk-colorectal
dullomyoblastoma: a case report and literature 2398. Sorensen AG, Emblem KE, Polaskova The Cowden syndrome: a clinical and genetic cancer: European Society for Medical Oncolo-
review of a rare tumor entity. Anticancer Res. P, Jennings D, Kim H, Ancukiewicz M, et al. study in 21 patients. Clin Genet. 29(3):222-33. gy Clinical Practice Guidelines. J Clin Oneal.
32(11):4939-44. PMID:23155263 (2012). Increased survival of glioblastoma pa- PMID:3698331 33(2):209-17. PMID25452455
2384. Smyth I, Narang MA, Evans T, Hei- tients who respond to antiangiogenic therapy 2415. Starzyk J, Starzyk B, Bartnik-Mikula 2430. Stojanova A, Penn LZ (2009). The role of
mann C, Nakamura Y, Chenevix-Trench G, with elevated blood perlusion. Cancer Res. A, Urbanowicz W, Dziatkowiak H (2001 ). Go- INl1/hSNF5 in gene regulation and cancer. Bio-
el al. (1999). Isolation and characterization of 72(2):402-7. PMID:22127927 nadolropin releasing hormone-independent chem Cell Biol. 87(1):163-77. PMID:19234532
human patched 2 (PTCH2), a putative tumour 2399. Soylemezoglu F, Onder S, Tezel GG, precocious puberty in a 5 year-old girl with 2431. Stokland T, Liu JF, Ironside JW, Ellison
suppressor gene inbasal cell carcinoma and Berker M (2003). Neuronal nuclear antigen suprasellar germ cell tumor secreting beta-hCG DW, Taylor R, Robinson KJ, et al. (2010). A
medulloblastoma on chromosome 1 p32. Hum (NeuN): a new tool in the diagnosis of central and alpha-fetoprotein. J Pediatr Endocrinol Me- multivariate analysis of factors determining
Mol Genet. 8(2):291-7. PMID:9931336 neurocytoma. Pathol Res Pract. 199(7):463-8. tab. 14(6):789-96. PMID 11453531 tumor progression in childhood low-grade gli-
2385. Snuderl M, Fazlollahi L, Le LP, Nitta M, PMID:14521262 2416. Stavrou T, Bromley CM, Nicholson HS, oma: a population-based cohort study (CCLG
Zhelyazkova BH, Davidson CJ, et al. (2011). 2400. Soylemezoglu F, Soffer D, Onol B, Byrne J, Packer RJ, Goldstein AM, et al. (2001). CNS9702). Neuro Oneal. 12(12):1257-68.
Mosaic amplification of multiple receptor tyrosi- Schwechheimer K, Kleihues P (1996). Lipo- Prognostic factors and secondary malignancies PMID:20861086
ne kinase genes in glioblastoma. Cancer Cell. matous medulloblastoma in adults. A distinct in childhood medulloblastoma. J Pediatr He- 2432. Stammel JM, Kimmelman AC, Ying H,
20(6):810-7. PMID:22137795 clinicopathological entity. Am J Surg Pathol. matol Oneal. 23(7):431-6. PMID:11878577 Nabioullin R, Ponugoti AH, Wiedemeyer R, et
2386. Snyder LA, Wolf AB, Oppenlander ME, 20(4):413-8. PMID:8604807 2417. Stuer C, Vilz B, Majores M, Becker A, al. (2007). Coactivation of receptor tyrosine
Bina R, Wilson JR, Ashby L, et al. (2014). The 2401. Spanberger T, Berghoff AS, Dinhof C, Schramm J, Simon M (2007). Frequent recur- kinases affects the response of tumor cells to
impact of extent of resection on malignant llhan-Mutlu A, Magerle M, Hutterer M, et al. rence and progression in pilocytic astrocyto- targeted therapies. Science. 318(5848):287-
transformation of pure oligodendrogliomas. J (2013). Extent of peritumoral brain edema cor- ma in adults. Cancer. 110(12):2799-808. 90. PMID:17872411
Neurosurg. 120(2):309-14. PMID:24313617 relates with prognosis, tumoral growth pattern, PMID:17973253 2433. Stone DM, Hynes M, Armanini M, Swan-
2387. Sobel RA (1993). Vestibular (acoustic) HIF1a expression and angiogenic activity in 2418. Steck PA, Pershouse MA, Jasser SA, son TA, Gu Q, Johnson RL, et al. (1996). The
schwannomas: histologic features in neurofibro- patients with single brain metastases. Clin Exp Yung WK, Lin H, Ligon AH, et al. (1997). ldenti- tumour-suppressor gene patched encodes a
matosis 2 and in unilateral cases. J Neuropathol Metastasis. 30(4):357-68. PMID:23076770 fication of a candidate tumour suppressor gene, candidate receptor for Sonic hedgehog. Nature.
Exp Neural. 52(2):106-13. PMID:8440992 2402. Specht CS, Smith TW, DeGirolami U, MMAC1, at chromosome 10q23.3 that is muta- 384(6605):129-34. PMID:8906787
2388. Sofela AA, Hettige S, Curran 0, Bassi Price JM (1986). Myxopapillary ependymo- ted in multiple advanced cancers. Nat Genet. 2433A. Stone DM, Murone M, Luoh S, Ye W,
S (2014). Malignant transformation in cranio- ma of the filum terminale. A light and electron 15(4):356-62. PMID:9090379 Armanini MP, Gurney A, et al. (1999). Charac-
pharyngiomas. Neurosurgery. 75(3):306-14, microscopic study. Cancer. 58(2):310-7. 2419. Steel TR, Botterill P, Sheehy JP (1994). terization of the human suppressor of fused, a
discussion 314. PMID:24978859 PMID:3521831 Paraganglioma of the cauda equina with asso- negative regulator of the zinc-finger transcrip-
2389. Soffer D, Pittaluga S, Caine Y, Feinsod 2403. Spence T, Sin-Chan P, Picard D, Barszc- ciated syringomyelia: case report. Surg Neural. tion factor Gli. J Cell Sci. 112(Pt 23):4437-48.
M (1983). Paraganglioma of cauda equina. A zyk M, Hoss K, Lu M, et al. (2014). CNS-PNETs 42(6):489-93. PMID:7825103 PMID:10564661
report of a case and review of the literature. with C19MC amplification and/or LIN28 expres- 2420. Steinberg GK, Shuer LM, Conley FK, 2434. Storlazzi CT, Von Steyern FV, Domans-
Cancer. 51(10):1907-10. PMID:6831356 sion comprise a distinct histogenetic diagno- Hanbery JW (1985). Evolution and outcome in ki HA, Mandahl N, Mertens F (2005). Biallelic
2390. Soffietti R, Baumert BG, Bello L, von stic and therapeutic entity. Acta Neuropathol. malignant astroglial neoplasms of the cerebel- somatic inactivation of the NF1 gene through
Deimling A, Duffau H, Frenay M, et al.; Europe- 128(2):291-303. PMID:24839957 lum. J Neurosurg. 62(1):9-17. PMID:3964859 chromosomal translocations in a sporadic
an Federation of Neurological Societies (2010). 2404. Sperduto PW, Chao ST, Sneed PK, Luo 2421. Steinbok P, Poskitt K, Hendson G (2006). neurofibroma. Int J Cancer. 117(6):1055-7.
Guidelines on management of low-grade glio- X, Suh J, Roberge D, et al. (2010). Diagno- Spontaneous regression of cerebellar ast- PMID:15986446
mas: report of an EFNS-EANO Task Force. Eur sis-specific prognostic factors, indexes, and rocytoma after subtotal resection. Childs Nerv 2435. Stratakis CA (2002). Mutations of the
J Neural. 17(9):1124-33. PMID:20718851 treatment outcomes for patients with newly di- Syst. 22(6):572-6. PMID:16552566 gene encoding the protein kinase A type I-al-
2391. Sohda T, Yun K (1996). Insulin-like agnosed brain metastases: a multi-institutional 2422. Stemmer-Rachamimov AO, Gonza- pha regulatory subunit (PRKAR1A) in patients
growth factor II expression in primary menin- analysis of 4,259 patients. Int J Radial Oneal lez-Agosti C, Xu L, Burwick JA, Beauchamp R, with the "complex of spotty skin pigmentation,
geal hemangiopericytoma and its metastasis to Biol Phys. 77(3):655-61 PMID:19942357 Pinney D, et al. (1997). Expression of NF2-en- myxomas, endocrine overactivity, and schwan-
the liver accompanied by hypoglycemia. Hum 2405. Sperleld AD, Hein C, Schroder JM, Lu- coded merlin and related ERM family proteins in nomas" (Camey complex). Ann N Y Acad Sci.
Pathol. 27(8) 858-61. PMID:8760024 dolph AC, Hanemann CO (2002). Occurrence the human central nervous system. J Neuropa- 968:3-21. PMID:12119264
2392. Solis OE, Mehta RI, Lai A, Mehta RI, and characterization of peripheral nerve in- thol Exp Neural. 56(6):735-42. PMID:9184664 2436. Stratakis CA, Carney JA (2009). The tri-
Farchoukh LO, Green RM, et al. (2011). Roset- volvement in neurofibromatosis type 2. Brain. 2423. Stemmer-Rachamimov AO, Horgan MA, ad of paragangliomas, gastric stromal tumours
te-forming glioneuronal tumor: a pineal region 125(Pt 5):996-1004. PMID:11960890 Taratuto AL, Munoz DG, Smith TW, Frosch and pulmonary chondromas (Carney triad), and
case with IDH1 and IDH2 mutation analyses 2406. Spiegel E (1920). Hyperplasie des Klein- MP, et al. (1997). Meningioangiomatosis is as- the dyad of paragangliomas and gastric stromal
and literature review of 43 cases. J Neurooncol. hirns. Beitr Pathol Anal. 67:539-48. sociated with neurofibromatosis 2 but not with sarcomas (Carney-Stratakis syndrome): mole-
102(3):477-84. PMID:20872044 2407. Squarize CH, Castilho RM, Gutkind JS somatic alterations of the NF2 gene. J Neuro- cular genetics and clinical implications. J Intern
2392A. Solomon DA, Wood MD, Tihan T, Bollen (2008). Chemoprevention and treatment of ex- palhol Exp Neural. 56(5):485-9. PMID:9143261 Med. 266(1):43-52. PMID:19522824
AW, Gupta N, Phillips JJ, et al. (2015). Diffuse perimental Cowden's disease by m TOR inhibi- 2424. Stemmer-Rachamimov AO, lno Y, Lim 2437. Stratmann R, Krieg M, Haas R, Plate
Midline Gliomas with Histone H3-K27M Mutati- tion with rapamycin. Cancer Res. 68(17):7066- ZY, Jacoby LB, MacCollin M, Gusella JF, et al. KH (1997). Putative control of angiogenesis in
on: A Series of 4 7 Cases Assessing lhe Spec- 72. PMID:18757421 (1998). Loss of the NF2 gene and merlin occur hemangioblastomas by the von Hippel-lindau
trum of Morphologic Variation and Associated 2408. Squire JA, Arab S, Marrano P, Bayani by the tumorlet stage of schwannoma develop- tumor suppressor gene. J Neuropathol Exp
Genetic Alterations. Brain Pathol. doi: 10.1111/ J, Karaskova J, Taylor M, et al. (2001). Mo- ment in neurofibromatosis 2. J Neuropathol Exp Neural. 56(11):1242-52. PMID:9370235
bpa.12336. [Epub ahead of print] lecular cytogenetic analysis of glial tumors Neural. 57(12):1164-7. PMID:9862639 2438. Strommer KN, Brandner S, Sarioglu AC,
2393. Song MK, Chung JS, Joo YD, Lee SM, using spectral karyotyping and comparative 2425. Stenzel W, Pels H, Staib P, lmpekoven Sure U, Yonekawa Y (1995). Symptomatic ce-
Oh SY, Shin DH, et al. (2011). Clinical importan- genomic hybridization. Mol Diagn. 6(2):93-108. P, Bektas N, Deckert M (2004 ). Concomitant rebellar metastasis and late local recurrence of
ce of Bcl-6-positive non-deep-site involvement PMID:11468694 manifestation of primary CNS lymphoma and a cauda equina paraganglioma. Case report. J
in non-HIV-related primary central nervous sys- 2409. Sredni ST, Tomita T (2015). Rhabdoid Toxoplasma encephalitis in a patient with AIDS. Neurosurg. 83(1):166-9. PMID:7782837
tem diffuse large B-cell lymphoma. J Neuroon- tumor predisposition syndrome. Pediatr Dev J Neural. 251(6):764-6. PMID:15311360 2439. Stucky CC, Johnson KN, Gray RJ, Pockaj
col. 104(3):825-31. PMID:21380743 Pathol. 18(1):49-58. PMID:25494491 2426. Stephen JH, Sievert AJ, Madsen PJ, Ju- BA, Ocal IT, Rose PS, et al. (2012). Malignant
2394. Song X, Andrew Allen R, Terence Dunn 2410. Stache C, Holsken A, Fahlbusch R, dkins AR, Resnick AC, Storm PB, et al. (2012). peripheral nerve sheath tumors (MPNST):
S, Fung KM, Farmer P, Gandhi S, et al. (2011). Flitsch J, Schlaffer SM, Buchfelder M, et al. Spinal cord ependymomas and myxopapillary the Mayo Clinic experience. Ann Surg Oneal.
Glioblastoma with PNET-like components has a (2014). Tight junction protein claudin-1 is dif- ependymomas in the first 2 decades of life: a 19(3):878-85. PMID:21861229
higher frequency of isocitrate dehydrogenase 1 ferentially expressed in craniopharyngioma clinicopathological and immunohistochemi- 2440. Stuivenvolt M, Mandi E, Verheul J,
(IDH1) mutation and likely a better prognosis subtypes and indicates invasive tumor growth. cal characterization of 19 cases. J Neurosurg Fleischeuer R, Tijssen CC (2012). Atypical

References 393
transformation in sacral drop metastasis from tumors: frequency and association with clinico- N Am. 26(1):99-104. PMID:25432188 S, Shigematsu H (1987). Mixed glioblastoma
posterior Iossa choroid plexus papilloma. BMJ pathologic features. Hum Pathol. 41 (2):232-ll. 2471. Swensen AR, Bushhouse SA (1998). and sarcoma with osteoid-chondral tissue. Clin
Case Rep. 2012:2012. PMID:22922909 PMID:19801160 Childhood cancer incidence and trends in Min- Neuropathol. 6(4):160-3. PMID:3115659
2441. Stupp R, Hegi ME, Gorlia T, Erridge SC, 2454. Sullivan JP, Nahed BV, Madden MW, Oli- nesota, 1988-1994. Minn Med. 81(12):27'-32. 2488. Taggard DA, Menezes AH (2000). Three
Perry J, Hong YK, et al.; European Organisa- veira SM, Springer S, Bhere D, et al. (2014). PMID:9866372 choroid plexus papillomas in a patient with Ai-
tion for Research and Treatment of Cancer Brain tumor cells in circulation are enriched for 2472. Swensen JJ, Keyser J, Coffin CM, Biegel cardi syndrome. A case report. Pediatr Neuro-
(EORTC); Canadian Brain Tumor Consortium; mesenchymal gene expression. Cancer Discov. JA, Viskochil DH, Williams MS (2009). Famili- surg. 33(4):219-23. PMID:11124640
CENTRIC study team (2014). Cilengitide com- 4(11):1299-309. PMID:25139148 al occurrence of schwannomas and malignant 2489. Taillibert S, Chodkiewicz C, Laigle-Do-
bined with standard treatment for patients with 2455. Sulman EP, Guerrero M, Aldape K rhabdoid tumour associated with a duplication nadey F, Napolitano M, Cartalat-Carel S, San-
newly diagnosed glioblastoma with methyla- (2009). Beyond grade: molecular pathology in SMARCB1. J Med Genet. 46(1):68-72. son M (2006). Gliomatosis cerebri: a review
ted MGMT promoter (CENTRIC EORTC of malignant gliomas. Semin Radial Oneal. PMID:19124645 of 296 cases from the ANOCEF database
26071-22072 study): a multicentre, randomi- 19(3):142-9. PMID:19464628 2473. Swerdlow SH, Campo E, Harris NL, Jaffe and the literature. J Neurooncol. 76(2):201-5.
sed, open-label, phase 3 trial. Lancet Oneal. 2456. Sundgren P, Annertz M, Englund E, ES, Pileri SA, Stein H, et al., editors. (2008). PMID:16200347
15(10):1100-8. PMID:25163906 Strombled LG, Hallas S (1999). Paragang- WHO Classification of Tumours of Haemat- 2490. Taillibert S, Laigle-Donadey F, Chodkie-
2442. Stupp R, Hegi ME, Mason WP, van den liomas of the spinal canal. Neuroradiology. opoietic and Lymphoid Tissues. 4th ed. Lyon: wicz C, Sanson M, Hoang-Xuan K, Delattre JY
Bent MJ, Taphoorn MJ, Janzer RC, et al.; Eu- 41(10):788-94. PMID:10552032 International Agency for Research on Cancer. (2005). Leptomeningeal metastases from solid
ropean Organisation for Research and Treat- 2457. Sung CC, Collins R, Li J, Pearl DK, 2474. Szathmari A, Champier J, Ghersi-Egea malignancy: a review. J Neurooncol. 75(1):85--
ment of Cancer Brain Tumour and Radiation Coons SW, Scheithauer BW, et al. (1996). JF, Jouvet A, Watrin C, Wierinckx A, et al. 99. PMID:16215819
Oncology Groups; National Cancer Institute of Glycolipids and myelin proteins in human oli- (2013). Molecular characterization of circum- 2491. Taipale J, Cooper MK, Maiti T, Beachy
Canada Clinical Trials Group (2009). Effects of godendrogliomas. Glycoconj J. 13(3):433-43. ventricular organs and third ventricle ependy- PA (2002). Patched acts catalytically to sup-
radiotherapy with concomitant and adjuvant te- PMID:8781974 ma in the rat: potential markers for periventri- press the activity of Smoothened. Nature.
mozolomide versus radiotherapy alone on sur- 2458. Surawicz TS, McCarthy BJ, Kupelian cular tumors. Neuropathology. 33(1 ):17-29. 418(6900):892-7. PMID:12192414
vival in glioblastoma in a randomised phase Ill V, Jukich PJ, Bruner JM, Davis FG (1999). PMID:22537279 2492. Takahashi E, Kajimoto K. Fukatsu T,
study: 5-year analysis of the EORTC-NCIC trial. Descriptive epidemiology of primary brain and 2475. Szeifert GT, Pasztor E (1993). Could era- Yoshida M, Eimoto T, Nakamura S (2005). lnt-
Lancet Oneel. 10(5):459-66. PMID 19269895 CNS tumors: results from the Central Brain Tu- niopharyngiomas produce pituitary hormones? ravascular large T-cell lymphoma: a case report
2443. Sturm D, Bender S, Jones OT, Lichter P, mor Registry of the United States, 1990-1994. Neural Res. 15(1):68-9. PMID:8098858 of CD30-positive and ALK-negative anaplastic
Grill J, Becher 0, et al. (2014). Paediatric and Neuro Oneal. 1(1):14-25. PMID:11554386 2476. Szudek J, Birch P, Riccardi VM, Evans type with cytotoxic molecule expression. Vir-
adult glioblastoma: multiform (epi)genomic cul- 2459. Suresh TN, Santosh V, Yasha TC, An- DG, Friedman JM (2000). Associations of clini- chows Arch. 447(6):1000-6. PMID:16189700
prits emerge. Nat Rev Cancer. 14(2):92-107. andh B, Mohanty A, lndiradevi B, et al. (2004). cal features in neurofibromatosis 1 (NF1 ). Ge- 2493. Takahashi E, Nakamura S (2013). His-
PMID:24457416 Medulloblastoma with extensive nodularity: net Epidemiol. 19(4):429-39. PMID:11108651 tiocytic sarcoma : an updated literature review
2444. Sturm D, Witt H, Hovestadt V, Khuong- a variant occurring in the very young-clinico- 2477. Szudek J, Joe H, Friedman JM (2002). based on the 2008 WHO classification. J Clin
Quang DA, Jones OT, Konermann C, et al. pathological and immunohistochemical study Analysis of intrafamilial phenotypic variation in Exp Hematop. 53(1):1-8. PMID:23801128
(2012). Hotspot mutations in H3F3A and IDH 1 of four cases. Childs Nerv Syst. 20(1):55-60. neurofibromatosis 1 (NF1 ). Genet Epidemiol. 2494. Takahashi M, Yamamoto J, Aoyama
define distinct epigenetic and biological subg- PMID:14657995 23(2):150-64. PMID:12214308 Y, Soejima Y, Akiba D, Nishizawa S (2009).
roups of glioblastoma. Cancer Cell. 22(4):425-- 2460. Suster D, Plaza JA, Shen R (2005). 2478. S6ylemezoglu F, Scheithauer BW, Esteve Efficacy of multi-staged surgery and adjuvant
37. PMID:23079654 Low-grade malignant perineurioma (perineurial J, Kleihues P (1997). Atypical central neurocyto- chemotherapy for successful treatment of aty-
2445. St6dberg T, Deniz Y, Esteitie N, Jacobs- sarcoma) of soft tissue: a potential diagnostic ma. J Neuropathol Exp Neural. 56(5):551-6. pical choroid plexus papilloma in an infant: case
son B, Mousavi-Jazi M, Dahl H, et al. (2002). A pitfall on fine needle aspiration. Ann Diagn Pa- PMID:9143268 report. Neural Med Chir (Tokyo). 49(10):484-7.
case of diffuse leptomeningeal oligodendroglio- lhol. 9(4):197-201. PMID:16084452 2479. S6ylemezoglu F, Tezel GG, Koybasoqlu PMID:19855149
matosis associated with HHV-6 variant A. Neu- 2461. Suva ML, Rheinbay E, Gillespie SM, Patel F, Er U, Akalan N (2001 ). Cranial infantile rnyo- 2495. Takami H, Fukushima S, Fukuoka K,
ropediatrics. 33(5):266-70. PMID 12536370 AP, Wakimoto H, Rabkin SD, et al. (2014). Re- fibromatosis: report of three cases. Childs Nerv Suzuki T, Yanagisawa T, Matsushita Y, et al.
2446. Sugawa N, Ekstrand AJ, James CD, constructing and reprogramming the tumor-pro- Syst. 17(9):524-7. PMID:11585325 (2015). Human chorionic gonadotropin is
Collins VP (1990). Identical splicing of aberrant pagating potential of glioblastoma stem-like 2480. Taal W, Oosterkamp HM, Walenkamp expressed virtually in all intracranial germ
epidermal growth factor receptor transcripts cells. Cell. 157(3):580-94. PMID:24726434 AM, Dubbink HJ, Beerepoot LV, Hanse MC, cell tumors. J Neurooncol. 124(1):23-32.
from amplified rearranged genes in human 2462. Suva ML, Riggi N, Bernstein BE (2013). et al. (2014). Single-agent bevacizumab or lo- PMID:25994796
glioblastomas. Proc Natl Acad Sci U S A. Epigenetic reprogramming in cancer. Science. mustine versus a combination of bevacizumab 2496. Takami H, Yoshida A, Fukushima S, Ari-
87(21):8602-6. PMID:2236070 339(6127):1567-70. PMID:23539597 plus lomustine in patients with recurrent gliob- ta H, Matsushita Y, Nakamura T, et al. (2015).
2447. Sughrue ME, Choi J, Rutkowski MJ, 2463. Suzuki A, de la Pompa JL, Stambolic lastoma (BELOB trial): a randomised control- Revisiting TP53 Mutations and lmmunohis-
Aranda D, Kane AJ, Barani IJ, et al. (2011). V, Elia AJ, Sasaki T, del Barcc Barrantes I, et led phase 2 trial. Lancet Oneal. 15(9):943-53. tochemistry-A Comparative Study in 157
Clinical features and post-surgical outccme of al. (1998). High cancer susceptibility and em- PMID:25035291 Diffuse Gliomas. Brain Pathol. 25(3):256-65
patients with astroblastoma. J Clin Neurosci. bryonic lethality associated with mutation of 2481. Taal W, van der Rijt CC, Dinjens WN, Sil- PMID:25040820
18(6):750-4. PMID:21507653 the PTEN tumor suppressor gene in mice. Curr levis Smitt PA, Wertenbroek AA, Bromberg JE, 2497. Takei H, Adesina AM, Mehta V, Powell
2448. Sughrue ME, Sanai N, Shangari G, Parsa Biol. 8(21):1169-78. PMID:9799734 et al. (2015). Treatment of large low-grade oli- SZ, Langford LA (2010). Atypical teratoid/rhab-
AT, Berger MS, McDermott MW (2010). Outco- 2464. Suzuki H, Aoki K, Chiba K, Sato Y, Shi- godendroglial tumors with upfront procarbazine, doid tumor of the pineal region in an adult. J
me and survival following primary and repeat ozawa Y, Shiraishi Y, et al. (2015). Mutational lomustine, and vincristine chemotherapy with Neurosurg. 113(2):374-9. PMID:19911885
surgery for World Health Organization Grade landscape and clonal architecture in grade long follow-up: a retrospective cohort study with 2498. Takei Y, Mirra SS, Miles ML (1976). Eo-
Ill meningiomas. J Neurosurg. 113(2):202-9. II and Ill gliomas. Nat Genet. 47(5):458-68. growth kinetics. J Neurooncol. 121 (2):365--72. sinophilic granular ceels in oligodendrogliomas.
PMID:20225922 PMID:25848751 PMID:25344884 An ultrastructural study. Cancer. 38(5):1968-
2449. Sugiarto S. Persson Al, Munoz EG, 2465. Svajdler M Jr, Rychly B, Gajdos M, Pa- 2482. Tabori U, Baskin B, Shago M, Alon N, 76. PMID:991110
Waldhuber M, Lamagna C, Ander N, et al. taky F, Frohlichova L, Perry A (2012). Gliosar- Taylor MD. Ray PN, et al. (2010). Universal 2499. Takei Y, Seyama S, Pearl GS, Tindall
(2011). Asymmetry-defective oligodendrocyte coma with alveolar rhabdomyosarcoma-like poor survival in children with medulloblastoma GT (1980). Ultrastructural study of the human
progenitors are glioma precursors. Cancer Cell. component: report of a case with a hitherto harboring somatic TP53 mutations. J Clin On- neurohypophysis. II. Cellular elements of neural
20(3):328-40. PMID:21907924 undescribed sarcomatous component. Cesk eal. 28(8):1345--50. PMID:20142599 parenchyma, the pituicytes. Cell Tissue Res.
2450. Sugino T, Mikami T, Akiyama Y, Wa- Patel. 48(4):210-4. PMID:23121030 2483. Tabori U, Shlien A, Baskin B, Levitt S, 205(2):273-87. PMID:7188885
nibuchi M, Hasegawa T, Mikuni N (2013). 2466. Swaidan MY, Hussaini M, Sultan I, Man- Ray P, Alon N, et al. (2010). TP53 alterations 2500. Takeshima H, Kawahara Y, Hirano H,
Primary central nervous system anaplastic sour A (2012). Radiological findings in gliosar- determine clinical subgroups and survival of pa- Obara S, Niiro M, Kuratsu J (2003). Postope-
large-cell lymphoma mimicking lymphomato- coma. A single institution experience. Neurora- tients with choroid plexus tumors. J Clin Oneal. rative regression of desmoplastic infantile
sis cerebri. Brain Tumor Pathol. 30(1):61-5. diol J. 25(2):173-80. PMID:24028910 28(12):1995--2001. PMID:20308654 gangliogliomas: report of two cases. Neu-
PMID:22426596 2467. Swanson KR, Bridge C, Murray JD, Al- 2484. Tabouret E, Chino! 0, Metellus P, Tai- rosurgery. 53(4):979-83, discussion 983-4.
2451. Sugiyama K, Arita K, Shima T, Nakao- vord EC Jr (2003). Virtual and real brain tumors: let A, Viens P. Goncalves A (2012). Recent PMID:14519230
ka M, Matsuoka T, Taniguchi E, et al. (2002). using mathematical modeling to quantify glioma trends in epidemiology of brain metastases: 2501. Tamura M, Gu J, Matsumoto K, Aota
Good clinical course in infants with desmo- growth and invasion. J Neural Sci. 216(1):1-10. an overview. Anticancer Res. 32(11):4655--62. S, Parsons R, Yamada KM (1998). Inhibition
plastic cerebral neuroepithelial tumor treated PMID:14607296 PMID:23155227 of cell migration, spreading, and focal adhe-
by surgery alone. J Neurooncol. 59(1):63-9. 2468. Swartling FJ, Savov V, Persson Al, Chen 2485. Tachibana 0, Nakazawa H. Lampe J, sions by tumor suppressor PTEN. Science.
PMID:12222839 J, Hackett CS, Northcott PA, et al. (2012). Dis- Watanabe K, Kleihues P, Ohgaki H (1995). Ex- 280(5369):1614-7. PMID:9616126
2452. Suh YL, Koo H, Kim TS, Chi JG, Park SH, tinct neural stem cell populations give rise to pression of Fas/AP0-1 during the progression 2502. Tan C, Scotting P J (2013). Stem cell
Khang SK, et al.; Neuropathology Study Group disparate brain tumors in response to N-MYC. of astrocytomas. Cancer Res. 55(23):5528-30. research points the way to the cell of origin
of the Korean Society of Pathologists (2002). Cancer Cell. 21(5):601-13. PMID:22624711 PMID:7585627 for intracranial germ cell tumours. J Pathol.
Tumors of the central nervous system in Korea: 2469. Sweet K, Willis J, Zhou XP, Gallione C, 2486. Tachibana 0, Yamashima T, Yamashita J, 229(1):4-11. PMID:22926997
a multicenter study of 3221 cases. J Neuroon- Sawada T, Alhopuro P, et al. (2005). Molecu- Takabatake Y (1994). lmmunohistochemical ex- 2503. Tan MH, Mester J, Peterson C, Yang Y,
ccl. 56(3):251-9. PMID:12061732 lar classification of patients with unexplained pression of human chorionic gonadotropin and Chen JL, Rybicki LA, et al. (2011 ). A clinical sco-
2453. Sukov WR, Cheville JC, Giannini C, hamartomatous and hyperplastic polyposis. P-glycoprotein in human pituitary glands and ring system for selection of patients for PTEN
Carlson AW, Shearer BM, Sinnwell JP, et al. JAMA. 294(19):2465-73. PMID:16287957 craniopharyngiomas. J Neurosurg. 80(1 ):79- mutation testing is proposed on the basis of a
(2010). lsochromosome 12p and polysomy 12 2470. Sweiss FB, Lee M, Sherman JH (2015). 84. PMID:7903692 prospective study of 3042 probands. Am J Hum
in primary central nervous system germ cell Extraventricular neurocytomas. Neurosurg Clin 2487. Tada T, Katsuyama T, Aoki T, Kobayashi Genet. 88(1):42-56. PMID:21194675

394 References
2504. Tan MH, Mester JL, Ngeow J, Rybicki LA, Central neurocytomas of the cervical spi- J, Chow WY, et al. (2013). lmplicalions of review of 46 cases. J Neurooncol. 115(1):53-9.
Orloff MS, Eng C (2012). Lifetime cancer risks nal cord. Report of lwo cases. J Neurosurg. tumor location on subtypes of medulloblasto- PMID:23824534
in individuals with germline PTEN mutations. 81(2):288-93. PMID:8027814 ma. Pediatr Blood Cancer. 60(9):1408-10. 2551. Tian Y, Wang J, Ge Jz, Ma Z, Ge M
Clin Cancer Res. 18(2):400--7. PMID:22252256 2520. Tavangar SM, Larijani B, Mahta A, PMID:23512859 (2015). lntracranial Rosai-Dorfman disease mi-
2505. Tan W, Huang W, Xiong J, Pan J, Geng Hosseini SM, Mehrazine M. Bandarian F 2535. Terada T (2015). Expression of cytoke- micking multiple meningiomas in a child: a case
D, Jun Z (2014). Neuroradiological features (2004 ). Craniopharyngioma: a clinicopatho· ratins in glioblasloma multiforme. Pathol Oneal report and review of the literature. Childs Nerv
of papillary glioneuronal tumor: a study of 8 logical study of 141 cases. Endocr Pathol. Res. 21(3):817-9. PMID:25633990 Syst. 31(2):317-23. PMJD:25183389
cases. J Comput Assist Tomogr. 38(5):634-8. 15(4):339-44. PMID:15681858 2536. Terashima K, Yu A, Chow WY, Hsu WC. 2552. Tibbetts KM, Emnett RJ, Gao F, Perry A,
PMID:24879457 2521. Taylor KR, Mackay A, Truffaux N, But- Chen P, Wong S, et al. (2014). Genome-wide Gutmann DH, Leonard JR (2009). Histopatho-
2506. Tanaka K, Waga S, ltho H, Shimizu DM, terfield YS, Morozova 0, Philippe C, el al. analysis of DNA copy number alterations and Jogic predictors of pilocytic astrocytoma event-
Namiki H (1989). Superficial location of malig- (2014). Recurrent activating ACVR1 mutations loss of heterozygosity in intracranial germ cell free survival. Acta Neuropathol. 117(6):657-65.
nant glioma with heavily Jipidized (foamy) tumor in diffuse intrinsic ponline glioma. Nat Genet. tumors. Pediatr Blood Cancer. 61(4):593-600. PMID:19271226
cells: a case report. J Neurooncol. 7(3):293-7. 46(5):457-61. PMID:24705252 PMID:24249158 2553. Tien RD, Barkovich AJ, Edwards MS
PMID:2795123 2522. Taylor MD, Gokgoz N, Andrulis IL, 2537. Thiel G, Losanowa T, Kintzel D, Nisch G, (1990). MR imaging of pineal tumors. AJR Am
2507. Tanaka M, Suda M, Ishikawa Y, Fujitake Mainprize TG, Drake JM, Rutka JT (2000). Martin H, Vorpahl K, et al. (1992). Karyolypes J Roentgenol. 155(1):143-51. PMID:2162137
J, Fujii H, Tatsuoka Y (1996). Idiopathic hyper- Familial poslerior fossa brain tumors of in- in 90 human gliomas. Cancer Genet Cytogenet. 2554. Tien RD, Brasch RC, Jackson DE, Dil-
trophic cranial pachymeningitis associated with fancy secondary to germline mutation of the 58(2):109-20. PMID:1551072 lon WP (1989). Cerebral Erdheim-Chester
hydrocephalus and myocarditis: remarkable hSNF5 gene. Am J Hum Genet. 66(4):1403-6. 2538. Thiele EA (2010). Managing and un- disease: persistent enhancement with Gd-DT-
steriod-induced remission of hypertrophic dura PMID:10739763 derstanding epilepsy in tuberous sclero- PA on MR images. Radiology. 172(3):791-2.
mater. Neurology. 46(2):554-6. PMJD:8614532 2523. Taylor MD, Liu L, Raffel C, Hui CC, Main- sis complex. Epilepsia. 51 Suppl 1 :90-1. PMID:2772189
2508. Tanaka S, Nakada M, Nobusawa S, Su- prize TG, Zhang X, et al. (2002). Mutations in PMID:20331728 2555. Tihan T, Fisher PG, Kepner JL, Godfraind
zuki SO, Sabit H, Miyashita K, et al. (2014). SUFU predispose to medulloblastoma. Nat Ge- 2539. Thiessen B, Finlay J, Kulkarni R, Rosen- C, McComb RD, Goldthwaite PT, et al. (1999).
Epithelioid glioblastoma arising from pleomor- net. 31(3):306-10. PMID:12068298 blum MK (1998). Astroblastoma: does histo- Pediatric astrocytomas with monomorphous
phic xanthoastrocytoma with the BRAF V600E 2524. Taylor MD, Northcott PA, Korshunov A, logy predict biologic behavior? J Neurooncol. pilomyxoid features and a less favorable outco-
mulalion. Brain Tumor Pathol. 31(3):172-6. Remke M, Cho YJ, Clifford SC, et al. (2012). 40(1):59-65. PMID:9874187 me. J Neuropathol Exp Neural. 58(10):1061-8.
PMID:24894018 Molecular subgroups of medulloblastoma: 2540. Thines L, Lejeune JP, Ruchoux MM, PMID:10515229
2509. Tanaka Y, Yokoo H, Komori T. Makita Y, the current consensus. Acta Neuropathol. Assaker R (2006). Management of delayed 2556. Tihan T, Vohra P, Berger MS. Keles GE
lshizawa T, Hirose T, el al. (2005). A distinct 123(4):465-72. PMID:22134537 intracranial and intraspinal metastases of (2006). Definition and diagnostic implications
pattern of Olig2-positive cellular distribution in 2525. Taylor MD, Perry J, Zlatescu MC. intradural spinal paragangliomas. Acta Neu- of gemistocytic aslrocytomas: a pathological
papillary glioneuronal tumors: a manifestation Slemmer-Rachamimov AO, Ang LC, Ina Y, et rochir (Wien). 148(1):63-6, discussion 66. perspective. J Neurooncol. 76(2):175-83.
of the oligodendroglial phenotype? Acta Neuro- al. (1999). The hPMS2 exon 5 mutation and PMID:16283104 PMID:16132490
pathol. 110(1):3s-47. PMID:15906048 malignant glioma. Case report. J Neurosurg. 2541. Thom M, Blurncke I, Aronica E (2012). 2557. Tihan T, Zhou T. Holmes E, Burger PC,
2510. Tanas MR, Sboner A, Oliveira AM, Er- 90(5):946-50. PMID:10223463 Long-term epilepsy-associated tumors. Brain Ozuysal S, Rushing EJ (2008). The progno-
ickson-Johnson MR, Hespell J, Hanwright PJ, 2526. Taylor MD, Poppleton H, Fuller C, Su X. Pathol. 22(3):350-79. PMID:22497610 stic value of histological grading of posterior
et al. (2011). Identification of a disease-defi- Liu Y, Jensen P, et al. (2005). Radial glia cells 2542. Thom M, Gomez-Anson B, Revesz T, fossa ependymomas in children: a Children's
ning gene fusion in epithelioid hemangioen- are candidate stem cells of ependymoma. Can- Harkness W, O'Brien CJ, Kett-White R, et al. Oncology Group study and a review of pro-
dolhelioma. Sci Transl Med. 3(98):98ra82. cer Cell. 8(4):323-35. PMID:16226707 (1999). Spontaneous intralesional haemorrha- gnostic factors. Mod Pathol. 21(2):165-77.
PMID:21885404 2526A. Taylor TE, Furnari FB, Cavenee WK ge in dysembryoplastic neuroepithelial tumours: PMID:18084249
2510A. Tanboon J, Williams EA, Louis DN (2012). Targeting EGFR for treatment of gliob- a series of five cases. J Neural Neurosurg Psy- 2558. Timmermann B, Kortmann RD, Kuhl
(2015). The Diagnostic Use of lmmunohisloche- lasloma: molecular basis to overcome resistan- chiatry. 67(1):97-101. PMID:10369831 J, Rutkowski S, Meisner C, Pietsch T, et al.
mical Surrogates for Signature Molecular Ge- ce. Curr Cancer Drug Targets. 12(3):197-209. 2543. Thom M, Toma A, An S, Martinian L, (2006). Role of radiotherapy in supratenlorial
netic Alterations in Gliomas. J Neuropathol Exp PMID:22268382 Hadjivassiliou G, Ratilal B, et al. (2011). One primitive neuroectodermal tumor in young child-
Neural. [Epub ahead of print] PMID:26671986 2527. Tchoghandjian A, Fernandez C, Colin C, hundred and one dysembryoplastic neuroepit- ren: results of the German HIT-SKK87 and HIT-
2511. Tandon A, Schiff D (2014). Therapeutic El Ayachi I, Voutsinos-Porche B, Fina F, et al. helial tumors: an adult epilepsy series with SKK92 trials. J Clin Oneal. 24(10):1554-60.
decision making in palienls wilh newly diagno- (2009). Pilocytic astrocytoma of the optic pa- immunohistochemical, molecular genetic, and PMID:16575007
sed low grade glioma. Curr Treat Options On- thway: a tumour deriving from radial glia cells clinical correlations and a review of the litera- 2559. Tinat J, Bougeard G, Baert-Desur-
eal. 15(4):529-38. PMID:25139406 with a specific gene signature. Brain. 132(Pt ture. J Neuropathol Exp Neural. 70(10):859-78. mont S, Vasseur S, Martin C, Bouvignies E,
2512. Tanimura A, Nakamura Y, Hachisuka 6):1523-35. PMJD:19336457 PMID:21937911 et al. (2009). 2009 version of the Chompret
H, Tanimura Y, Fukumura A (1984). Heman- 2528. Tee AR, Fingar DC, Manning BD, Kwi- 2544. Thomas C, Ruland V, Kordes U, Hartung criteria for Li Fraumeni syndrome. J Clin
gioblastoma of lhe central nervous system: atkowski DJ, Cantley LC, Blenis J (2002). S, Capper D, Pietsch T, et al. (2015). Pediatric Oneal. 27(26):e108-9, author reply e110.
nature of the stromal cells as studied by the Tuberous sclerosis complex-1 and -2 gene atypical choroid plexus papilloma reconsidered: PMID:19652052
immunoperoxidase technique. Hum Pathol. products function together to inhibit mamma- increased mitotic activity is prognostic only in 2560. Tirakotai W, Menne! HD, Celik I, Hellwig
15(9):866-9. PMID:6432675 lian target of rapamycin (mTOR)-mediated older children. Acta Neuropathol. 129(6):925-7. D, Bertalanffy H, Riegel T (2006). Secretory
2513. Taratuto AL, Molina HA, Diez B. Zucca- downstream signaling. Proc Natl Acad Sci U S PMID:25935663 meningioma: immunohistochemical findings
ro G, Monges J (1985). Primary rhabdomyo- A. 99(21):13571-6. PMID:12271141 2545. Thomas L, Spurlock G, Eudall C, Thomas and evaluation of mast cell infiltration. Neuro-
sarcoma of brain and cerebellum. Report of 2529. Tehrani M. Friedman TM. Olson JJ, Brat NS, Mort M, Hamby SE, et al. (2012). Exploring surg Rev. 29(1):41-8. PMID:16010579
four cases in infants: an immunohistochemi- DJ (2008). lntravascular Jhrombosis in central the somatic NF1 mutational spectrum associa- 2561. Toedt G, Barbus S, Wolter M, Felsberg
cal study. Acta Neuropathol. 66(2):98-104. nervous system malignancies: a potential role ted wilh NF1 cutaneous neurofibromas. Eur J J, Tews B, Blond F, et al. (2011). Molecular
PMJD:4013672 in astrocytoma progression to glioblastoma. Hum Genet. 20(4):411-9. PMID:22108604 signatures classify astrocytic gliomas by IDH1
2514. Taraluto AL, Monges J, Lylyk P, Leiguar- Brain Pathol. 18(2):164-71. PMID:18093251 2546. Thomas PK, King RH, Chiang TR, mutation status. lntJ Cancer. 128(5):1095-103.
da R (1984). Superficial cerebral aslrocytoma 2530. Tekautz TM, Fuller CE, Blaney S, Foula- Scaravilli F, Sharma AK, Downie AW (1990). PMID:20473936
attached to dura. Report of six cases in infants. di M, Broniscer A, Merchant TE, et al. (2005). Neurofibromatous neuropathy. Muscle Nerve. 2562. Tohma Y, Gratas C, Biernat W, Peraud
Cancer. 54(11):2505-12. PMID:6498740 Atypical teratoid/rhabdoid tumors (ATRT): 13(2):93-101. PMID:2156160 A, Fukuda M, Yonekawa Y, et al. (1998). PTEN
2515. TaratutoAL, Pamala H, Sevlever G, Gallo improved survival in children 3 years of age 2547. Thommen F, Hewer E, Schafer SC, (MMAC1) mutations are frequent in primary
G, Monges J (1995). Dysembryoplastic neu- and older with radiation therapy and high-dose Vassella E, Kappeler A, Vajtai I (2013). Rosel- glioblastomas (de nova) but not in secondary
roepithelial tumor: morphological, immunocyto- alkylator-based chemotherapy. J Clin Oneal. le-forming glioneuronal tumor of the cerebellum glioblastomas. J Neuropathol Exp Neural.
chemical, and deoxyribonucleic acid analyses 23(7): 1491-9. PMID:15735125 in statu nascendi: an incidentally detected di- 57(7):684-9. PMID:9690672
in a pediatric series. Neurosurgery. 36(3):474- 2531. Telera S, Carosi M, Cerasoli V, Facciolo minutive example indicates derivation from the 2563. Tohma Y, Gratas C, Van Meir EG, Des-
81. PMJD:7753346 F, Occhipinti E, Vidiri A, et al. (2006). Hemotho- internal granule cell layer. Clin Neuropathol. baillets I, Tenan M, Tachibana 0, et al. (1998).
2516. Tashjian VS, Khanlou N, Vinters HV, rax presenting as a primitive thoracic paragan- 32(5):370-6. PMID:23547894 Necrogenesis and Fas/AP0-1 (CD95) ex-
Canalis RF, Becker DP (2009). Hemangiope- glioma. Case illustration. J Neurosurg Spine. 2548. Thompsett AR, Ellison DW, Stevenson pression in primary (de nova) and secondary
ricytoma of lhe cerebelloponline angle: a case 4(6):515. PMJD:16776367 FK, Zhu D (1999). V(H) gene sequences from glioblastomas. J Neuropathol Exp Neural.
report and review of the literature. Surg Neural. 2532. Telfeian AE, Judkins A, Younkin D, Pol- primary central nervous system lymphomas in- 57(3):239-45. PMID:9600216
72(3):290-5. PMID:18786704 lock AN, Crino P (2004). Subependymal giant dicate derivation from highly mutated germinal 2564. Tomita T, Gates E (1999). Pituitary
2517. Tate M, Sughrue ME, Rutkowski MJ, cell astrocytoma with cranial and spinal me- center B cells with ongoing mutational activity. adenomas and granular cell tumors. Inci-
Kane AJ, Aranda D, McClinlon L, el al. (2012). tastases in a patient with tuberous sclerosis. Blood. 94(5):1738-46. PMID:10477699 dence, cell type, and location of tumor in 100
The long-term poslsurgical prognosis of pati- Case report. J Neurosurg. 100(5) Suppl Pedi- 2549. Thompson MC, Fuller C, Hogg TL, Dalton pituitary glands at autopsy. Am J Clin Pathol.
ents with pineoblasloma. Cancer. 118(1):173- atrics:498-500. PMID:15287462 J, Finkelstein D, Lau CC, et al. (2006). Geno- 111(6):817-25. PMID:10361519
9. PMJD:21717450 2533. Temme A, Geiger KD, Wiedemuth R, mics identifies medulloblastoma subgroups that 2565. Tomlinson FH, Scheithauer BW, Hayes-
2518. Tatevossian RG, Tang B, Dalton J, Fors- Conseur K. Pietsch T, Felsberg J, et al. (2010). are enriched for specific genetic alterations. J tek CJ, Parisi JE, Meyer FB, Shaw EG, et al.
hewT, Lawson AR. Ma J, et al. (2010). MYB up- Giant cell glioblastoma is associated with al- Clin Oneal. 24(12):1924-31. PMID:16567768 (1994). The significance of atypia and hislo-
regulation and genetic aberrations in a subset tered aurora b expression and concomitant 2550. nan R, Hao S, Hou Z, Bian L, Zhang Jogic malignancy in pilocytic astrocytoma of
of pediatric low-grade gliomas. Acta Neuropa- p53 mutation. J Neuropathol Exp Neural. Y, Wu W, et al. (2013). Clinical characteristics the cerebellum: a clinicopathologic and flow
lhol. 120(6):731-43. PMID:21046410 69(6):632-42. PMID:20467329 and prognostic analysis of recurrent hemangi- cytometric study. J Child Neural. 9(3):301-10.
2519. Taller SB, Borges LF, Louis DN (1994). 2534. Teo WY, Shen J, Su JM, Yu A, Wang opericytoma in the central nervous system: a PMID:7930411

References 395
2566. Tomlinson FH, Scheithauer BW, Kelly of two cases. Acta Neuropathol. 91(6):5B---7. B Cell Pathol Incl Mol Pathol. 53(1 ):52---7. and the UK Medical Research Council (2005).
PJ, Forbes GS (1991). Subependymoma with PMID:8781655 PMID:2885972 Long-term efficacy of early versus delayed
rhabdomyosarcomatous differentiation: report 2583. Tsukayama C, Arakawa Y (2002). A pa- 2598. Uluc K, Arsava EM, Ozkan B, Gila A, radiotherapy for low-grade astrocytoma and
of a case and literature review. Neurosurgery. pillary glioneuronal tumor arising in an elderly Zorlu F, Tan E (2004). Primary leptomeningeal oligodendroglioma in adults: the EORTC 22845
28(5):761---8. PMID:1876259 woman: a case report. Brain Tumor Pathol. sarcomatosis; a pathology proven case with randomised trial. Lancet. 366(9490):985---90.
2567. Tamura N, Hirano H, Watanabe 0, Wat- 19(1 ):35---9. PMID 12455887 challenging MRI and clinical findings. J Neu- PMID:16168780
arai J, Itch Y, Mineura K, et al. (1997). Central 2584. Tsumanuma I, Sato M, Okazaki H, rooncol. 66(3):307---12. PMID:15015662 2614. van den Bent MJ, Brandes AA, Rampling
neurocytoma with clinically malignant beha- Tanaka R, Washiyama K, Kawasaki T, et al. 2599. Uno K, Takita J, Yokomori K, Tanaka Y, R, Kouwenhoven MC, Kras JM, Carpentier
vior. AJNR Am J Neuroradiol. 18(6):1175-8. (1995). The analysis of p53 tumor suppressor Ohta S, Shimada H, et al. (2002). Aberrations AF, et al. (2009). Randomized phase II trial of
PMID:9194446 gene in pineal parenchymal tumors. Noshuyo of the hSNF5/INl1 gene are restricted to ma- erlotinib versus temozolomide or carmustine
2568. Tong J, Hannan F, Zhu Y, Bernards A, Byori. 12(1):39-43. PMID:7795728 lignant rhabdoid tumors or atypical teratoid/ in recurrent glioblastoma: EORTC brain tumor
Zhong Y (2002). Neurofibromin regulates G 2585. Tsumanuma I, Tanaka R, Abe S, Ka- rhabdoid tumors in pediatric solid tumors. group study 26034. J Clin Oneel. 27(8):1268---
protein-stimulated adenylyl cyclase activity. Nat wasaki T, Washiyama K, Kumanishi T (1997). Genes Chromosomes Cancer. 34(1):33-41. 74. PMID:19204207
Neurosci. 5(2):95---6. PMID:11788835 Infrequent mutation of Waf1/p21 gene, a CDK PMID:11921280 2615. van den Bent MJ, Brandes AA, Taphoorn
2569. Torchia J, Picard D, Lafay-Cousin L, Ha- inhibitor gene, in brain tumors. Neural Med 2600. Upadhyaya M, Huson SM, Davies M, MJ, Kras JM, Kouwenhoven MC, Delattre JY,
wkins CE, Kim SK, Letourneau L, et al. (2015). Chir (Tokyo). 37(2):150---6, discussion 156---7. Thomas N, Chuzhanova N, Giovannini S, et et al. (2013). Adjuvant procarbazine, lomus-
Molecular subgroups of atypical teratoid rhab- PMID:9059037 al. (2007). An absence of cutaneous neurofi- tine, and vincristine chemotherapy in newly
doid tumours in children: an integrated genomic 2586. Tsumanuma I, Tanaka R, Washiyama bromas associated with a 3-bp inframe deleti- diagnosed anaplastic oligodendroglioma: long-
and clinicopathological analysis. Lancet Oneel. K (1999). Clinicopathological study of pineal on in exon 17 of the NF1 gene (c.2970---2972 term follow-up of EORTC brain tumor group
16(5):569---82. PMID:25882982 parenchymal tumors: correlation between his· delAAT): evidence of a clinically significant NF1 study 26951. J Clin Oneel. 31(3):344---50.
2570. Torres CF, Korones DN, Pilcher W (1997). topathological features, proliferative potential, genotype-phenotype correlation. Am J Hum Ge- PMID:23071237
Multiple ependymomas in a patient with Tur- and prognosis. Brain Tumor Pathol. 16(2):61---8. net. 80(1):140--51. PMID:17160901 2616. van den Bent MJ, Bromberg JE (2012).
cot's syndrome. Med Pediatr Oneel. 28(1):59--- PMID:10746962 2601. Upadhyaya M, Spurtock G, Kluwe L, Anaplastic oligodendroglial tumors. Handb Clin
61. PMID:8950338 2587. Tu PH, Giannini C, Judkins AR, Schwalb Chuzhanova N, Bennett E, Thomas N, et al. Neural. 105:467---84. PMID:22230513
2571. Trehan G, Bruge H, Vinchon M, Khalil JM, Burack R, O'Neill BP, et al. (2005). Clini- (2009). The spectrum of somatic and germ- 2617. van den Bent MJ, Carpentier AF, Brandes
C, Ruchoux MM, Dhellemmes P, et al. (2004). copathologic and genetic profile of intracranial line NF1 mutations in NF1 patients with spinal AA, Sanson M, Taphoom MJ, Bernsen HJ, et
MR imaging in the diagnosis of desmoplastic marginal zone lymphoma: a primary low-grade neurofibromas. Neurogenetics. 10(3):251---63. al. (2006). Adjuvant procarbazine, lomustine,
infantile tumor: retrospective study of six ca- CNS lymphoma that mimics meningioma. J Clin PMID:19221814 and vincristine improves progression-free sur-
ses. AJNR Am J Neuroradiol. 25(6):1028---33. Oneel. 23(24):5718---27. PMID:16009945 2602. Urbach H (2012). High-field magnetic vival but not overall survival in newly diagnosed
PMID:15205142 2588. Tucker T, Wolkenstein P, Revuz J, Zeller resonance imaging for epilepsy. Neuroima- anaplastic oligodendrogliomas and oligoast-
2572. Tresser N, Parveen T, Roessmann U J, Friedman JM (2005). Association between ging Clin N Am. 22(2):173---89, ix-x. [ix-x.] rocytomas: a randomized European Organi-
(1993). lntracranial lipomas with teratomatous benign and malignant peripheral nerve she· PMID:22548927 sation for Research and Treatment of Cancer
elements.Arch Pathol Lab Med. 117(9):918---20. ath tumors in NF1. Neurology. 65(2):205---11. 2603. Ushio Y, Arita N, Yoshimine T, Ikeda T, phase Ill trial. J Clin Oneel. 24(18):2715---22.
PMID:8368905 PMID:16043787 Mogami H (1987). Malignant recurrence of PMID:16782911
2573. Trifanescu R, Ansorge 0, Wass JA, 2589. Turcan S, Rohle D, Goenka A, Walsh LA, childhood cerebellar astrocytoma: case report. 2618. van den Bent MJ, Dubbink HJ, Marie Y,
Grossman AB, Karavitaki N (2012). Rathke's Fang F, Yilmaz E, et al. (2012). IDH1 mutation is Neurosurgery. 21(2):251---5. PMID:3309714 Brandes AA, Taphoom MJ, Wesseling P, et al.
cleft cysts. Clin Endocrinol (Oxf). 76(2):151---60. sufficient to establish the glioma hyperrnethyla- 2604. Usul H, Kuzeyli K, Cakir E, Caylan R, (2010). IDH1 and IDH2 mutations are progno-
PMID:21951110 tor phenotype. Nature. 483(7390):479-83. Sayin OC, Peksoylu B, et al. (2005). Giant stic but not predictive for outcome in anaplastic
2574. Trimbath JD, Petersen GM, Erdman PMID:22343889 cranial extradural primary fibroxanthoma: oligodendroglial tumors: a report of the Europe-
SH, Ferre M, Luce MC, Giardiello FM (2001). 2590. Turcot J, Despres JP, St Pierre F (1959). a case report. Surg Neural. 63(3):281-4. an Organization for Research and Treatment of
Cafe-au-lait spots and early onset colorectal Malignant tumors of the central nervous system PMID:15734528 Cancer Brain Tumor Group. Clin Cancer Res.
neoplasia: a variant of HNPCC? Fam Cancer. associated with familial polyposis of the colon: 2605. Vajtai I, Beck J, Kappeler A, Hewer E 16(5):1597---604. PMID:20160062
1(2) 101---5. PMID:14574005 report of two cases. Dis Colon Rectum. 2:465--- (2011 ). Spindle cell oncocytoma of the pituitary 2619. van den Bent MJ, Dubbink HJ, Sanson
2575. Trofatter JA, MacCollin MM, Rutter JL, 8. PMID:13839882 gland with follicle-like component: organotypic M, van der Lee-Haartoo CR, Hegi M, Jeuken
Murrell JR, Duyao MP, Parry DM, et al. (1993). A 2591. Uchikawa H, Toyoda M, Nagao K, differentiation to support its origin from follicu- JW, et al. (2009). MGMT promoter methylation
novel moesin-, ezrin-, radixin-like gene is a can- Miyauchi H, Nishikawa R, Fujii K, et al. (2006). lo-steilate cells. Acta Neuropathol. 122(2):253--- is prognostic but not predictive for outcome
didate for the neurofibromatosis 2 tumor sup- Brain· and heart-specific Patched-1 containing 8. PMID:21590491 to adjuvant PCV chemotherapy in anaplastic
pressor. Cell. 72(5):791---800. PMID:8453669 exon 12b is a dominant negative isoform 2606. Vajtai I, Kappeler A, Lukes A, Arnold M, oligodendroglial tumors: a report from EORTC
2576. Troost D, Jansen GH, Dingemans KP and is expressed in medulloblastomas. Bio· Luthy AR, Leibundgut K (2006). Papillary glio- Brain Tumor Group Study 26951. J Clin Oneel.
(1990). Cerebral medulloepithelioma-electron chem Biophys Res Commun. 349(1):277-83. neuronal tumor. Pathol Res Pract. 202(2):107--- 27(35):5881---6. PMID:19901104
microscopy and immunohistochemistry. Acta PMID:16934747 12. PMID:16413693 2620. van den Bent MJ, Gravendeel LA, Gorlia
Neuropathol. 80(1):103---7. PMID:2360414 2592. Ud Din N, Memon A, Aftab K, Ahmad Z, 2607. Vajtai I, Varga Z, Aguui A (1996). MIB- T, Kras JM, Lapre L, Wesseling P, et al. (2011 ).
2577. Trost D, Ehrler M, Fimmers R, Felsberg J, Ahmed R, Hassan S (2012). Oligodendrogli· 1 immunoreactivity reveals different labelling A hypermethylated phenotype is a better predic-
Sabel MC, Kirsch L, et al. (2007). Identification oma arising in the glial component of ovarian in low-grade and in malignant epithelial neo- tor of survival than MGMT methylation in ana-
of genomic aberrations associated with shorter teratomas: a series of six cases and review plasms of the choroid plexus. Histopathology. plastic oligodendroglial brain tumors: a report
overall survival in patients with oligodendro- of literature. J Clin Pathol. 65(7):631-4. 29(2):147---51. PMID:8872148 from EORTC study 26951. Clin Cancer Res.
glial tumors. Int J Cancer. 120(11):236!,--76. PMID:22496515 2608. Val-Bernal JF, Hernando M, Garijo 17(22):714S--55. PMID:21914791
PMID 17285580 2593. Ueba T, Okawa M. Abe H, Inoue T, Iaka- MF, Villa P (1997). Renal perineurioma in 2621. Van den Bent MJ, Reni M, Gatta G, Vecht
2578. Tsai CJ, Wang Y, Allen PK, Mahajan A, no K, Hayashi H, et al. (2013). Central nervous childhood. Gen Diagn Pathol. 143(1):75---81. C (2008). Oligodendroglioma. Grit Rev Oneel
McCutcheon IE, Rao G, et al. (2014). Outco- system marginal zone B-cell lymphoma of mu- PMID:9269912 Hematol. 66(3):262---72. PMID:18272388
mes after surgery and radiotherapy for spinal cosa-associated lymphoid tissue type involving 2609. Valdez R, McKeever P, Finn WG, Ge- 2622. van den Munckhof P, Christiaans I, Ken-
myxopapillary ependymoma: update of the the brain and spinal cord parenchyma. Neuro- barski S, Schnitzer B (2002). Composite germ ter SB, Baas F, Hulsebos TJ (2012). Germline
MD Anderson Cancer Center experience. Neu- pathology. 33(3):306---11. PMID:22994302 cell tumor and B-cell non-Hodgkin's lympho- SMARCB1 mutation predisposes to multiple
rosurgery. 75(3):205---14, discussion 213-4. 2594. Ueki K, Ono Y, Henson JW, Efird JT, von ma arising in the sella turcica. Hum Pathol. meningiomas and schwannomas with pre-
PMID:24818785 Deimling A, Louis DN (1996). CDKN2/p16 or 33(10):1044--7. PMID:12395379 ferential location of cranial meningiomas at
2579. Tsikitis M, Zhang Z, Edelman W, Zagzag RB alterations occur in the majority of gliob- 2610. Valenti MP, Froelich S, Armspach JP, the falx cerebri. Neurogenetics. 13(1):1---7.
D, Kalpana GV (2005). Genetic ablation of Cy- lastomas and are inversely correlated. Cancer Chenard MP, Dietemann JL, Kerhli P, et al. PMID:22038540
clin 01 abrogates genesis of rhabdoid tumors Res. 56(1):150---3. PMID:8548755 (2002). Contribution of SISCOM imaging in the 2623. Van Gompel JJ, Koeller KK, Meyer FB,
resulting from lni1 loss. Pree Natl Acad Sci U S 2595. Ulbright TM, Hattab EM, Zhang S, Ehr· presurgical evaluation of temporal lobe epilepsy Marsh WR, Burger PC, Roncaroli F, et al.
A. 102(34):12129---34. PMID:16099835 lich Y, Foster RS, Einhorn LH, et al. (2010). related to dysembryoplastic neuroepithelial tu- (2011). Cortical ependymoma: an unusual epi-
2580. Tso CL, Freije WA, Day A, Chen Z, Primitive neuroectodermal tumors in patients mors. Epilepsia. 43(3):270---6. PMID:11906512 leptogenic lesion. J Neurosurg. 114(4):1187---
Merriman B, Perlina A, et al. (2006). Distin- with testicular germ cell tumors usually re- 2611. van den Bent MJ (2010). lnterobserver 94. PMID:21235315
ct transcription profiles of primary and se- semble pediatric-type central nervous system variation of the histopathological diagnosis 2624. Van Meir EG (1998). "Turcot's syndro-
condary glioblastoma subgroups. Cancer Res. embryonal neoplasms and lack chromosome in clinical trials on glioma: a clinician's per- me": phenotype of brain tumors, survival and
66(1):159---67. PMID:16397228 22 rearrangements. Mod Pathol. 23(7):972-80. spective. Acta Neuropathol. 120(3):297---304. mode of inheritance. Int J Cancer. 75(1 ):162-4.
2581. Tsou HG, Teng DH, Ping XL, Brancolini V, PMID:20348883 PMID:20644945 PMID:9426707
Davis T, Hu R, et al. (1997). The role of MMAC1 2596. Ulm AJ, Yachnis AT, Brat DJ, Rhoton 2612. van den Bent MJ (2014). Practice ch- 2625. van Slegtenhorst M, de Hoogt R, Harm-
mutations in early-onset breast cancer: causa- AL Jr (2004). Pituicytoma: report of two ca- anging mature results of RTOG study 9802: ans C, Nellist M, Janssen B, Verhoef S, et al.
tive in association with Cowden syndrome and ses and clues regarding histogenesis. Neu- another positive PCV trial makes adjuvant (1997). Identification of the tuberous sclerosis
excluded in BRCA 1-negative cases. Am J Hum rosurgery. 54(3):753---7, discussion 757---8. chemotherapy part of standard of care in gene TSC1 on chromosome 9q34. Science.
Genet. 61(5):1036-43. PMID9345101 PMID:15028154 low-grade glioma. Neuro Oneel. 16(12):1570-4. 277(5327):805-8. PMID:9242607
2582. Tsuchida T, Matsumoto M, Shirayama 2597. Ulrich J, Heitz PU, Fischer T, Obrist E, PMID:25355680 2626. van Slegtenhorst M, Verhoef S, Tempel·
Y, lmahori T, Kasai H, Kawamoto K (1996). Gullolta F (1987). Granular cell tumors: eviden- 2613. van den Bent MJ, Afra D, de Witte 0, Ben aars A, Bakker L, Wang Q, Wessels M, et al.
Neuronal and glial characteristics of central ce for heterogeneous tumor cell differentiation. Hassel M, Schraub S, Hoang-Xuan K, et al.; (1999). Mutational spectrum of the TSC1 gene
neurocytoma: electron microscopical analysis An immunocytochemical study. Virchows Arch EORTC Radiotherapy and Brain Tumor Groups in a cohort of 225 tuberous sclerosis complex

396 References
patients: no evidence for genotype-pheno- 2641. Vege KO, Giannini C, Scheithauer BW 2658. Vogazianou AP, Chan R, Backlund LM, Visakorpi T, Kallio M, Jaaskelainen J (1996).
type correlation. J Med Genet. 36(4):285--9. (2000). The immunophenotype of ependymo- Pearson OM, Liu L, Langford CF, et al. (2010). Outcome of 31 inlracranial haemangiopericyto-
PMID:10227394 mas. Appl lmmunohistochem Mal Morphol. Distinct patterns of 1 p and 19q alterations iden- mas: poor predictive value of cell proliferation
2627. van Thuijl HF, Mazor T, Johnson BE, 8(1):25-31. PMID:10937045 tify subtypes of human gliomas that have dif- indices. Acta Neurochir (Wien). 138(12):1399-
Fouse SD, Aihara K, Hong C, et al. (2015). 2642. Venkataraman G, Rizzo KA, Chavez JJ, ferent prognoses. Neuro Oneal. 12(7):664-78. 408. PMID:9030346
Evolution of DNA repair defects during malig- Streubel B, Raffeld M, Jaffe ES, et al. (2011). PMID:20164239 2674. Wacker MR, Cogen PH, Etzell JE, Dane-
nant progression of low-grade gliomas after Marginal zone lymphomas involving meningeal 2659. Vogelgesang S, Junge MH, Pahnke J, shvar L, Davis RL, Prados MD (1992). Diffuse
temozolomide treatment. Acta Neuropathol. dura: possible link to lgG4-related diseases. Gaab MR, Warzok RW (2002). August 2001: leptomeningeal involvement by a ganglioglioma
129(4):597---{,07. PMID:25724300 Mod Pathol. 24(3):355-66. PMID:21102421 Sellar/suprasellar mass in a 59-year--0ld in a child. Case report. J Neurosurg. 77(2):302-
2628. VandenBerg SR (1993). Desmoplastic in- 2643. Venkataraman S, Pandian C, Kumar woman. Brain Palhol. 12(1):135--6, 139. 6. PMID:1625019
fantile ganglioglioma and desmoplastic cerebral SA (2013). Primary spinal primitive neuroecto- PMID:11770897 2675. Wakai S, Segawa H, Kitahara S, Asano
astrocytoma of infancy. Brain Pathol. 3(3):275- dermal tumour - a case report. Ann Neurosci. 2660. van Deimling A, Fimmers R, Schmidt T, Sano K, Ogihara R, et al. (1980). Teratoma in
81. PMID:8293187 20(2):80-2. PMID:25206019 MC, Bender B, Fassbender F, Nagel J, et al. the pineal region in two brothers. Case reports.
2629. VandenBerg SR, May EE, Rubinstein LJ, 2644. Venneti S, Santi M, Felicella MM, Yarilin (2000). Comprehensive allelotype and genetic J Neurosurg. 53(2):239-43. PMID:7431063
Herman MM, Perentes E, Vinores SA, et al. D, Phillips JJ, Sullivan LM, et al. (2014). A sen- anaysis of 466 human nervous system tumors. 2676. Wakamatsu T, Matsuo T, Kawano S,
(1987). Desmoplastic supratenlorial neuroepit- sitive and specific histopalhologic prognostic J Neuropathol Exp Neural. 59(6):544-58. Teramoto S, Matsumura H (1971). Glioblasto-
helial tumors of infancy with divergent differen- marker for H3F3A K27M mutant pediatric gli- PMID:10850867 ma with extracranial metastasis through vent-
tiation potential ("desmoplastic infantile ganglio- oblastomas. Acta Neuropathol. 128(5):743-53. 2661. van Deimling A, Janzer R, Kleihues P, riculopleural shunt. Case report. J Neurosurg.
gliomas"). Report on 11 cases of a distinctive PMID:25200322 Wiestler OD (1990). Patterns of differentiation 34(5):697-701. PMID:4326303
embryonal tumor with favorable prognosis. J 2645. Verflaak RG, Hoadley KA, Purdom E, in central neurocytoma. An immunohistochemi- 2677. Waldron JS, Tihan T (2003). Epide-
Neurosurg. 66(1):58-71. PMID:3097276 Wang V, Qi Y, Wilkerson MD, et al., Cancer cal study of eleven biopsies. Acta Neuropathol. miology and pathology of intraventricular
2630. Vandewalle G, Brucher JM, Michotte A Genome Atlas Research Network (2010). In- 79(5):473-9. PMID:2109481 tumors. Neurosurg Clin N Am. 14(4):469-82.
(1995). lntracranial facial nerve rhabdomyo- tegrated genomic analysis identifies clinically 2662. van Deimling A, Kleihues P, Saremas- PMID:15024796
ma. Case report. J Neurosurg. 83(5):919-22. relevant subtypes of glioblastoma charac- lani P, Yasargil MG, Spoerri 0, Sudhof TC, et 2678. Walker C, Baborie A, Crooks D, Wilkins
PMID:7472566 terized by abnormalities in PDGFRA, IDH1, al. (1991 ). Histogenesis and differentiation S, Jenkinson MD (2011 ). Biology, genetics
2631. Vang R, Heck K, Fuller GN, Medeiros EGFR, and NF1. Cancer Cell. 17(1):98-110. potential of central neurocytomas. Lab Invest. and imaging of glial cell tumours. Br J Radial.
LJ (2000). Granular cell tumor of intracrani- PMID:20129251 64(4):585-91. PMID:1901927 84(Spec No 2):890-106. PMID:22433833
al meninges. Clin Neuropathol. 19(1):41-4. 2646. Verhoef S, Bakker L, Tempelaars AM, 2663. van Deimling A, Larson J, Wellenreuther 2679. Walker C, Joyce KA, Thompson-Hehir J,
PMID:10774952 Hesseling-Janssen AL, Mazurczak T, Jozwiak R, Stangl AP, van Velthoven V, Warnick R, et al. Davies MP, Gibbs FE, Halliwell N, et al. (2001).
2632. Varga Z, Vajtai I, Aguzzi A (1996). The S, et al. (1999). High rate of mosaicism in tu- (1999). Clonal origin of recurrent meningiomas. Characterisation of molecular alterations in
standard isoform of CD44 is preferentially berous sclerosis complex. Am J Hum Genet. Brain Pathol. 9(4):645-50. PMID:10517503 microdissected archival gliomas. Acta Neuropa-
expressed in atypical papillomas and carcino- 64(6):1632-7. PMID:10330349 2664. van Hoff K, Hartmann W, van Bueren AO, thol. 101(4):321-33. PMID:11355303
mas of the choroid plexus. Pathol Res Pract. 2647. Verma A, Zhou H, Chin S, Bruggers C, Gerber NU, Grotzer MA, Pietsch T, et al. (2010). 2680. Walker L, Thompson D, Easton D, Pon-
192(12):1225-31. PMID:9182293 Kestle J, Khalua S (2012). EGFR as a predictor Large cell/anaplastic medulloblastoma: outco- der B, Ponder M, Frayling I, et al. (2006). A
2633. Varikatt W, Dexter M, Mahajan H, Mura- of relapse in myxopapillary ependymoma. Pedi- me according to myc status, histopathological, prospective study of neurofibromatosis type
li R, Ng T (2009). Usefulness of smears in atr Blood Cancer. 59(4):746-8. PMID:22190537 and clinical risk factors. Pediatr Blood Cancer. 1 cancer incidence in the UK. Br J Cancer.
intra-operative diagnosis of newly described 2648. Versteege I, Medjkane S, Rouillard D, 54(3):369-76. PMID:19908297 95(2):233-8. PMID:16786042
entities of CNS. Neuropathology. 29(6):641-8. Delattre O (2002). A key role of the hSNF5/ 2665. von Hoff K, Hinkes B, Dannenmann-Stern 2681. Walker LS, Sansom OM (2011). The
PMID:19563508 IN11 tumour suppressor in the control of the E, van Bueren AO, Warmuth-Metz M, Soeren- emerging role of CTLA4 as a cell-extrinsic re-
2634. Varley JM, McGown G, Thorncroft M, G1-S transition of the cell cycle. Oncogene. sen N, et al. (2011). Frequency, risk-factors gulator of T cell responses. Nat Rev lmmunol.
Santibanez-Kore! MF, Kelsey AM, Tricker KJ, 21(42):6403-12 PMID:12226744 and survival of children with atypical teratoid 11(12):852-63. PMID:22116087
el al. (1997). Germ-line mutations of TP53 2649. Versteege I, Sevenet N, Lange J, Rous- rhabdoid tumors (AT/RT) of the CNS diagnosed 2682. Wallner KE, Gonzales MF, Edwards
in Li-Fraumeni families: an extended study seau-Merck MF, Ambros P, Handgretinger R, between 1988 and 2004, and registered to the MS, Wara WM, Sheline GE (1988). Treatment
of 39 families. Cancer Res. 57(15):3245--52. et al. (1998). Truncating mutations of hSNF5/ German HIT database. Pediatr Blood Cancer. results of juvenile pilocytic astrocytoma. J Neu-
PMID:9242456 IN11 in aggressive paediatric cancer. Nature. 57(6):978-85. PMID:21796761 rosurg. 69(2):171-6. PMID:3392563
2635. Vasen HF, Ghorbanoghli Z, Bourdeau! F, 394(6689):203...{J. PMID:9671307 2666. van Koch CS, Gulati M, Aldape K, Berger 2683. Wanebo JE, Lanser RR, Glenn GM, Old-
Cabaret 0, Caron 0, Duval A, et al.; EU-Con- 2650. Vescovi AL, Galli R, Reynolds BA (2006). MS (2002). Familial medulloblastoma: case re- field EH (2003). The natural history of heman-
sortium Care for CMMR-D (C4CMMR-D) Brain tumour stem cells. Nat Rev Cancer. port of one family and review of the literature. gioblaslomas of the central nervous system in
(2014). Guidelines for surveillance of individu- 6(6):425-36. PMID:16723989 Neurosurgery. 51(1):227-33, discussion 233. patients with von Hippel-lindau disease. J Neu-
als with constitutional mismatch repair-deficien- 2651. Villa S, Miller RC, Krengli M, Abusaris H, PMID:12182422 rosurg. 98(1):82-94. PMID:12546356
cy proposed by the European Consortium "Care Baumert BG, Servagi-Vernat S, et al. (2012). 2667. Vorechovsky I, Tingby 0, Hartman M, 2684. Wang CC, Turner J, Steel T (2013).
for CMMR-D" (C4CMMR-D). J Med Genet. Primary pineal tumors: outcome and prognostic Stromberg B, Nister M, Collins VP, et al. (1997). Spontaneous pineal apoplexy in a pineal pa-
51 (5):283-93. PMID:24556086 factors-a study from the Rare Cancer Network Somatic mutations in the human homologue renchymal tumor of intermediate differentiation.
2636. Vasen HF, Tomlinson I, Castells A (2015). (RCN). Clin Transl Oneal. 14(11):827-34. of Drosophila patched in primitive neuroec- Cancer Biol Med. 10(1):43-6. PMID:23691444
Clinical management of hereditary colorectal PMID:22914906 todermal tumours. Oncogene. 15(3):361...{J. 2685. Wang F, Travins J, DeLaBarre B, Pen-
cancer syndromes. Nat Rev Gastroenterol He- 2652. Villano JL, Koshy M, Shaikh H, Dolecek PMID:9233770 ard-Lacronique V, Schalm S, Hansen E, et al.
patol. 12(2):88-97. PMID:25582351 TA, McCarthy BJ (2011). Age, gender, and racial 2668. vorecnovsky I, Unden AB, Sandstedt (2013). Targeted inhibition of mutant IDH2 in
2637. Vasiljevic A, Champier J, Figarella-Bran- differences in incidence and survival in primary B, Toftgard R, Stahle-Backdahl M (1997). Tri- leukemia cells induces cellular differentiation.
ger D, Wierinckx A, Jouvet A, Fevre-Montan- CNS lymphoma. Br J Cancer. 105(9):1414-8. choepitheliomas contain somatic mutations in Science. 340(6132):622-6. PMID:23558173
ge M (2013). Molecular characterization of PMID:21915121 the overexpressed PTCH gene: support for a 2686. Wang H, Zhang S, Rehman SK, Zhang
central neurocytomas: potential markers for 2653. Villano JL, Parker CK, Dolecek TA gatekeeper mechanism in skin tumorigenesis. Z, Li W, Makki MS, et al. (2014). Clinicopatho-
tumor typing and progression. Neuropathology. (2013). Descriptive epidemiology of ependy- Cancer Res. 57(21 ):4677-81. PMID:9354420 logical features of myxopapillary ependymoma.
33(2):149-61. PMID:22816789 mal tumours in the United States. Br J Cancer. 2669. Vortmeyer AO, Frank S, Jeong SY, Yuan J Clin Neurosci. 21(4):569-73. PMID:24332590
2638. Vasiljevic A, Francois P, Loundou A, Fe- 108(11):2367-71. PMID:23660944 K, lkejiri B, Lee YS, et al. (2003). Developmen- 2687. Wang H, Zhang S, Wu C, Zhang Z,
vre-Montange M, Jouvet A, Roche PH, et al. 2654. Vinchon M, Blond S, Lejeune JP, Krivosik tal arrest of angioblastic lineage initiates tumo- Qin T (2013). Melanocytomas of the central
(2012). Prognostic factors in central neurocyto- I, Fossati P, Assaker R, et al. (1994). Associa- rigenesis in von Hippel-lindau disease. Cancer nervous system: a clinicopathological and mo-
mas: a multicenter study of 71 cases. Am J Surg tion of Lhermitte-Duclos and Cowden disease: Res. 63(21):7051-5. PMID:14612494 lecular study. Eur J Clin Invest. 43(8):809-15.
Pathol. 36(2):220-7. PMID:22251941 report of a new case and review of the literature. 2670. Vortmeyer AO, Gnarra JR, Emmert-Buck PMID:23683178
2639. Valer I, Montesinos-Rongen M, Schles- J Neural Neurosurg Psychiatry. 57(6):699-704. MR, Katz D, Linehan WM, Oldfield EH, et al. 2688. Wang L, Motoi T, Khanin R, Olshen A,
ner M, Haake A, Purschke F, Sprute R, et al. PMID:8006650 (1997). van Hippel-lindau gene deletion detec- Mertens F, Bridge J, et al. (2012). Identificati-
(2015). The mutational pattern of primary lym- 2655. Vinters HV, Park SH, Johnson MW, Mi- ted in the stromal cell component of a cerebellar on of a novel, recurrent HEY1 -NCOA2 fusion
phoma of the central nervous system deter- schel PS, Catania M, Kerfoot C (1999). Cortical hemangioblastoma associated with von Hip- in mesenchymal chondrosarcoma based on a
mined by whole-exome sequencing. Leukemia. dysplasia, genetic abnormalities and neuro- pel-lindau disease. Hum Pathol. 28(5):540-3. genome-wide screen of exon-level expression
29(3):677-85. PMID:25189415 cutaneous syndromes. Dev Neurosci. 21 (3- PMID:9158701 data. Genes Chromosomes Cancer. 51(2):127-
2639A. Vaubel RA, Chen SG, Raleigh DR, 5):248-59. PMID:10575248 2671. Vredenburgh JJ, Desjardins A, Herndon 39. PMID:22034177
Link MJ, Chicoine MR, Barani I, et al. (2016). 2656. Vital A, Vital C, Martin-Negrier ML, JE 2nd, Dowell JM, Reardon DA, Quinn JA, et 2689. Wang L, Yamaguchi S, Burstein MD, Te-
Meningiomas With Rhabdoid Features Lacking McGrogan G, Bioulac P, Trojani M, et al. (1994). al. (2007). Phase II trial of bevacizumab and rashima K, Chang K, Ng HK, etal. (2014). Novel
Other Histologic Features of Malignancy: A Lhermitte-Duclos type cerebellum hamarto- irinotecan in recurrent malignant glioma. Clin somatic and germline mutations in intracranial
Study of 44 Cases and Review of the Litera- ma and Cowden disease. Clin Neuropalhol. Cancer Res. 13(4):1253-9. PMID:17317837 germ cell tumours. Nature. 511(7508):241-5.
ture. J Neuropathol Exp Neural. 75(1):44-52. 13(4):229-31. PMID:7955671 2672. Vujovic S, Henderson S, Presneau N, PMID:24896186
PMID:26705409 2657. Vivekanandan S, Dickinson P, Bessell E, Odell E, Jacques TS, Tirabosco R, et al. (2006). 2690. Wang M, Jia D, Shen J, Zhang J, Li G
2640. Vecht CJ, Kerkhof M, Duran-Pena A O'Connor S (2011). An unusual case of primary Brachyury, a crucial regulator of notochordal (2013). Clinical and imaging features of central
(2014). Seizure prognosis in brain tumors: new anaplastic large cell central nervous system development, is a novel biomarker for chordo- neurocytomas. J Clin Neurosci. 20(5):679-85.
insights and evidence-based management. On- lymphoma: an 8-year success story. BMJ Case mas. J Pathol. 209(2):157---{,5_ PMID:16538613 PMID:23522930
cologist. 19(7):751-9. PMID:24899645 Rep. 2011 :2011. PMID:22707580 2673. Vuorinen V, Sallinen P, Haapasalo H, 2691. Wang M, Tihan T, Rojiani AM, Bodhireddy

References 397
SR, Prayson RA, lacuone JJ, et al. (2005). Brain Pathol. 6(3):217-23, discussion 23-4. 2721. Weiner HL, Wisoff JH, Rosenberg ME, PMID:7745429
Monomorphous angiocentric glioma: a dis- PMID:8864278 Kupersmith MJ, Cohen H, Zagzag D, et al. 2735. Wester DJ, Falcone S, Green BA, Camp
tinctive epileptogenic neoplasm with features 2706. Watanabe K, Tachibana 0, Yonekawa Y, (1994). Craniopharyngiomas: a clinicopa- A, Quencer RM (1993). Paraganglioma of the fi.
of infiltrating astrocytoma and ependymoma. Kleihues P, Ohgaki H (1997). Role of gemis- thological analysis of factors predictive of !um: MR appearance. J Comput Assist Tomogr.
J Neuropathol Exp Neural. 64(10):875-81. tocytes in astrocytoma progression. Lab Invest. recurrence and functional outcome. Neuro- 17(6):967-9. PMID:8227586
PMID:16215459 76(2):277-84. PMID:9042164 surgery. 35(6):1001-10, discussion 1010-1. 2736. Wharton SB, Chan KK, Anderson JR
2692. Wang X, Dubuc AM, Ramaswamy 2707. Watanabe T, Makiyama Y, Nishimoto H, PMID:7885544 Stoeber K, Williams GH (2001 ). Replicativ�
V, Mack S, Gendoo OM, Remke M, et al. Matsumoto M, Kikuchi A, Tsubokawa T (1995). 2722. Weiner HL, Zagzag D, Babu R, Wein· Mcm2 protein as a novel proliferation marker in
(2015). Medulloblastoma subgroups remain Metachronous ovarian dysgerminoma after a reb HJ, Ransohoff J (1993). Schwannoma of oligodendrogliomas and its relationship to Ki67
stable across primary and metastatic com- suprasellar germ-cell tumor treated by radiation the fourth ventricle presenting with hemifacial labelling index, histological grade and progno-
partments. Acta Neuropathol. 129(3):449-57. therapy. Case report. J Neurosurg. 83(1):149- spasm. A report of two cases. J Neurooncol. sis. Neuropathol Appl Neurobiol. 27(4):305-13.
PMID:25689980 53. PMID:7782834 15(1):37-43. PMID:8455061 PMID:11532161
2693. Wang XQ, Zhou Q, Li ST, Liao CL, Zhang 2708. Watanabe T, Mizowaki T, Arakawa Y, lizu- 2723. Weingart MF, Roth JJ, Hutt-Cabezas 2737. White FV, Dehner LP, Belchis DA, Conard
H, Zhang BY (2013). Solitary fibrous tumors of ka Y, Ogura K, Sakanaka K, et al. (2014). Pineal M, Busse TM, Kaur H, Price A, et al. (2015). K, Davis MM, Stocker JT, et al. (1999). Conge-
the central nervous system: clinical features parenchymal tumor of intermediate differentiati- Disrupting LIN28 in atypical teratoid rhabdoid nital disseminated malignant rhabdoid tumor: a
and imaging findings in 22 patients. J Comput on: Treatment outcomes of five cases. Mal Clin tumors reveals the importance of the mitogen distinct clinicopathologic entity demonstrating
Assist Tomogr. 37(5):658-65. PMID:24045237 Oneal. 2(2):197-202. PMID:24649332 activated protein kinase pathway as a the- abnormalities of chromosome 22q11. Am J
2694. Wang Y, Xiong J, Chu SG, Liu Y, Cheng 2709. Watanabe T, Nobusawa S, Kleihues P, rapeutic target. Oncotarget. 6(5):3165-77. Surg Pathol. 23(3):249-56. PMID:10078913
HX, Wang YF, et al. (2009). Rosette-forming Ohgaki H (2009). IDH1 mutations are early PMID:25638158 2738. White W, Shiu MH, Rosenblum MK,
glioneuronal tumor: report of an unusual case events in the development of astrocytomas and 2724. Weiss SW, Langloss JM, Enzinger FM Erlandson RA, Woodruff JM (1990). Cellular
with intraventricular dissemination. Acta Neuro- oligodendrogliomas. Am J Pathol. 174(4):1149- (1983). Value of S-100 protein in the diagnosis schwannoma. A clinicopathologic study of 57
pathol. 118(6):813-9. PMID:19585134 53. PMID:19246647 of soft tissue tumors with particular reference patients and 58 tumors. Cancer. 66(6):1266-
2695. Wanggou S, Jiang X, Li Q, Zhang L, Liu 2710. Watanabe T, Wal A, Nobusawa S, Klei- to benign and malignant Schwann cell tumors. 75. PMID:2400975
D, Li G, et al. (2012). HESRG: a novel biomar- hues P, Ohgaki H (2009). Selective acquisition Lab Invest. 49(3):299-308. PMID:6310227 2739. Whittle IR, Dow GR, Lammie GA, Ward-
ker for intracranial germinoma and embryo- of IDH1 R132C mutations in astrocytomas as- 2725. Weiss WA, Burns MJ, Hackett C, Alda- law J (1999). Dsyembryoplastic neuroepithe-
nal carcinoma. J Neurooncol. 106(2):251-9. sociated with Li-Fraumeni syndrome. Acta Neu- pe K, Hill JR, Kuriyama H, et al. (2003). Ge- lial tumour with discrete bilateral multifocality:
PMID:21861197 ropathol. 117(6):653-6. PMID 19340432 netic determinants of malignancy in a mouse further evidence for a germinal origin. Br J Neu-
2696. Wani K, Armstrong TS, Vera-Bolanos E, 2711. Weber DC, Wang Y, Miller R, Villa S, model for oligodendroglioma. Cancer Res. rosurg. 13(5):508-11. PMID:10627786
Raghunathan A, Ellison D, Gilbertson R, et al.; Zaucha R, Pica A, et al. (2015). Long-term 63(7):1589-95. PMID:12670909 2740. Wick W, Hartmann C, Engel C, Stoffels
Collaborative Ependymoma Research Network outcome of patients with spinal myxopapil- 2726. Welander J, Larsson C, Backdahl M, Felsberg J, Stockhammer F, et al. (2009).
(2012). A prognostic gene expression signature lary ependymoma: treatment results from the M, Hareni N, Sivlsr T, Brauckhoff M, et al. NOA-04 randomized phase Ill trial of sequen-
in infratentorial ependymoma. Acta Neuropa- MD Anderson Cancer Center and institutions (2012). Integrative genomics reveals frequent tial radiochemotherapy of anaplastic glioma
thol. 123(5):727-38. PMID:22322993 from the Rare Cancer Network. Neuro Oneal. somatic NF1 mutations in sporadic pheochro- with procarbazine, lomustine, and vincristine or
2697. Warmuth-Metz M, Bison B, Gerber NU, 17(4):588-95 PMID:25301811 mocytomas. Hum Mal Genet. 21(26):5406-16. temozolomide. J Clin Oneal. 27(35):5874-80.
Pietsch T, Hasselblatt M, Fruhwald MC (2013). 2712. Weber HC, Marsh DJ, Lubensky IA, Lin PMJD:23010473 PMID:19901110
Bone involvement in atypical teratoid/rhabdoid AY, Eng C (1998). Germline PTEN/MMAC1/ 2727. Wellenreuther R, Kraus JA, Lenartz D, 2741. Wick W, Meisner C, Hentschel B, Plat-
tumors of the CNS. AJNR Am J Neuroradiol. TEP1 mutations and association with gast- Menon AG, Schramm J, Louis ON, et al. (1995). ten M, Schilling A, Wiestler B, et al. (2013).
34(10):2039-42. PMID:23681355 rointestinal manifestations in Cowden disease. Analysis of the neurofibromatosis 2 gene re- Prognostic or predictive value of MGMT pro-
2698. Warnick RE, Raisanen J, Adornato Gastroenterology. 114S:G2902. veals molecular variants of meningioma. Am J moter methylation in gliomas depends on
BT, Prados MD, Davis RL, Larson DA, et al. 2713. Weber RG, Hoischen A, Ehrler M, Zip- Pathol. 146(4):827-32. PMID:7717450 IDH1 mutation. Neurology. 81(17):1515-22.
(1993). lntracranial myxopapillary ependymo- per P, Kaulich K, Blaschke B, et al. (2007). 2728. Weller M, Felsberg J, Hartmann C, Ber- PMID:24068788
ma: case report. J Neurooncol. 15(3):251-6. Frequent loss of chromosome 9, homozygous ger H, Steinbach JP, Schramm J, et al. (2009). 27 42. Wick W, Platten M, Meisner C, Felsberg
PMID:8360710 CDKN2A/p14(ARF)/CDKN2B deletion and low Molecular predictors of progression-free and J, Tabatabai G, Simon M, et al.; NOA-08 Stu-
2699. Warren C, James LA, Ramsden RT, Wal- TSC1 mRNA expression in pleomorphic xan- overall survival in patients with newly diagnosed dy Group of Neuro-oncology Working Group
lace A, Baser ME, Varley JM, et al. (2003). Iden- thoastrocytomas. Oncogene. 26(7):1088-97. glioblastoma: a prospective translational study (NOA) of German Cancer Society (2012). Te-
tification of recurrent regions of chromosome PMID:16909113 of the German Glioma Network. J Clin Oneal. mozolomide chemotherapy alone versus radio·
loss and gain in vestibular schwannomas using 2714. Wechsler J, Lantieri L, Zeller J, Voisin 27(34):5743-50. PMID:19805672 therapy alone for malignant astrocytoma in the
comparative genomic hybridisation. J Med Ge- MC, Martin-Garcia N, Wolkenstein P (2003). 2729. Weller M, Tabatabai G, Kastner B, Fels- elderly: the NOA-08 randomised, phase 3 trial.
net. 40(11):802-6. PMID:14627667 Aberrant axon neurofilaments in schwannomas berg J, Steinbach JP, Wick A, et al.; DIRECTOR Lancet Oneal. 13(7):707-15. PMID:22578793
2700. Warren KE (2012). Diffuse intrinsic pon- associated with phacomatoses. Virchows Arch. Study Group (2015). MGMT promoter methyla- 27 43. Wick W, Weller M, van den Bent M, San-
tine glioma: poised for progress. Front Oneal. 443(6):768-73. PMID:14508685 tion is a strong prognostic biomarker for benefit son M, Weiler M, von Deimling A, et al. (2014).
2:205. PMID:23293772 2715. Wechsler-Reya RJ, Scott MP (1999). from dose-intensified temozolomide rechallen- MGMT testing-the challenges for biornar-
2701. Wasdahl DA, Scheithauer BW, Andrews Control of neuronal precursor proliferation in ge in progressive glioblastoma: the DIREC- ker-based glioma treatment. Nat Rev Neural.
BT, Jeffrey RA Jr (1994). Cerebellar pleomor- the cerebellum by Sonic Hedgehog. Neuron. TOR trial. Clin Cancer Res. 21(9):2057-64. 10(7):372-85. PMID:24912512
phic xanthoastrocytoma: case report. Neu- 22(1):103-14. PMID:10027293 PMID:25655102 2744. Wicking C, Gillies S, Smyth I, Shanley
rosurgery. 35(5):947-50, discussion 950-1. 2716. Wefers AK, Warmuth-Metz M, Pbschl 2730. Weller M, van den Bent M, Hopkins K, S, Fowles L, Ratcliffe J, et al. (1997). De novo
PMID:7838347 J, von Bueren AO, Monoranu CM, Seelos K, Tonn JC, Stupp R, Falini A, et al.; European mutations of the Patched gene in nevoid basal
2702. Watanabe K, Ogata N, von Ammon K, et al. (2014). Subgroup-specific localization of Association for Neuro-Oncology (EANO) Task cell carcinoma syndrome help to define the cli-
Yonekawa Y, Nagai M, Ohgaki H, et al. (1996). human medulloblastoma based on pre-ope- Force on Malignant Glioma (2014). EANO gui- nical phenotype.Am J Med Genet. 73(3):304-7.
lmmunohistochemical assessments of p53 pro- rative MRI. Acta Neuropathol. 127(6):931-3. deline for the diagnosis and treatment of ana- PMID:9415689
tein accumulation and tumor growth fraction du- PMID:24699697 plastic gliomas and glioblastoma. Lancet Oneal. 2745. Wicking C, Shanley S, Smyth I, Gillies S,
ring the progression of astrocytomas. In: Brain 2717. Wei D, Rich P, Bridges L, Martin AJ, Chau 15(9):e395-403. PMID:25079102 Negus K, Graham S, et al. (1997). Most germ·
Tumour Research and Therapy. Nagai I, Bodi I, et al. (2013). Rare case of cerebral 2731. Weller M, Weber RG, Willscher E, Rieh· line mutations in the nevoid basal cell carcino-
2702A. Watanabe T, Nakamura M, Yonekawa MALToma presenting with stroke-like symp- mer V, Hentschel B, Kreuz M, et al. (2015). Mo- ma syndrome lead to a premature termination
Y, Kleihues P, Ohgaki H (2001 ). Promoter hy- toms and seizures. BMJ Case Rep'.2013:2013. lecular classification of diffuse cerebral WHO of the PATCHED protein, and no genotype-phe·
permethylation and homozygous deletion of the PMID:23608841 grade 11/111 gliomas using genome- and tran- notype correlations are evident. Am J Hum Ge-
p14ARF and p161NK4a genes in oligodendro- 2718. Wei G, Zhang W, Li Q, Kang X, Zhao H, scriptome-wide profiling improves stratification net. 60(1):21-6. PMID:8981943
gliomas. Acta Neuropathol. 101(3):185-9. Liu X, et al. (2014). Magnetic resonance charac- of prognostically distinct patient groups. Acta 2746. Wiemels J, Wrensch M, Claus EB (2010).
PMID:11307615 teristics of adult-onset Lhermitte-Duclos disea- Neuropathol. 129(5):679-93. PMID:25783747 Epidemiology and etiology of meningioma. J
2703. Watanabe K, Peraud A, Gratas C, Wa- se: An indicator for active cancer surveillance? 2732. Wellons JC 3rd, Reddy AT, Tubbs RS, Neurooncol. 99(3):307-14. PMID:20821343
kai S, Kleihues P, Ohgaki H (1998). p53 and Mol Clin Oneal. 2(3):415-20. PMID:24772310 Abdullatif H, Oakes WJ, Blount JP, et al. (2004). 2747. Wiemels JL, Wrensch M, Sison JD, Zhou
PTEN gene mutations in gemistocytic ast- 2719. Wei YQ, Hang ZB, Liu KF (1992). In situ Neuroendoscopic findings in patients with in· M, Bondy M, Calvocoressi L, et al. (2011). Re-
rocytomas. Acta Neuropathol. 95(6):559-64. observation of inflammatory cell-tumor cell in- tracranial germinomas correlating with diabetes duced allergy and immunoglobulin E among
PMJD:96507 46 teraction in human seminomas (germinomas): insipidus. J Neurosurg. 100(5) Suppl Pediat- adults with intracranial meningioma cornpa-
2704. Watanabe K, Sato K, Biernat W, Tachi- light, electron microscopic, and immunohis- rics:430-6. PMID:15287450 red to controls. Int J Cancer. 129(8):1932-9.
bana 0, van Ammon K, Ogata N, et al. (1997). tochemical study. Hum Pathol. 23(4):421-8. 2733. Wen PY, Kesari S (2008). Malignant glio- PMID:21520030
Incidence and timing of p53 mutations during PMID:1563744 mas in adults. N Engl J Med. 359(5):492-507. 2748. Wiens AL, Cheng L, Bertsch EC, John-
astrocytoma progression in patients with mul- 2720. Weinbreck N, Marie B, Bressenot A, PMID:18669428 son KA, Zhang S, Hattab EM (2012). Polysomy
tiple biopsies. Clin Cancer Res. 3(4):523-30. Montagne K, Joud A, Baumann C, et al. (2008). 2734. Wesseling P, Schlingemann RO, Riet- of chromosomes 1 and/or 19 is common and
PMID:9815715 lmmunohistochemical markers to distinguish veld FJ, Link M, Burger PC, Ruiter DJ (1995). associated with less favorable clinical outco-
2705. Watanabe K, Tachibana 0, Sala K, between hemangioblastoma and metastatic Early and extensive contribution of pericytes/ me in oligodendrogliomas: fluorescent in situ
Yonekawa Y, Kleihues P, Ohgaki H (1996). clear-cell renal cell carcinoma in the brain: vascular smooth muscle cells to microvascu- hybridization analysis of 84 consecutive ca-
Overexpression of the EGF receptor and p53 utility of aquaporin1 combined with cytokeratin lar proliferation in glioblastoma multiforme: an ses. J Neuropathol Exp Neural. 71(7):618-24.
mutations are mutually exclusive in the evolu- AE1/AE3 immunostaining. Am J Surg Pathol. immuno-light and immune-electron microscopic PMID:22710961
tion of primary and secondary glioblastomas. 32(7):1051-9. PMID:18496143 study. J Neuropathol Exp Neural. 54(3):304-10. 2749. Wiens AL, Hattab EM (2014). The

398 References
pathological spectrum of solid CNS metastases 0, Colas C, Entz-Werle N, et al.; EU-Consor- ependymoma. Am J Surg Pathol. 28(7):914-20. Witte D, Meijer D, et al. (2008). Plexiform and
in the pediatric population. J Neurosurg Pediatr. tium Care for CMMRD (C4CMMRD) (2014). PMID:15223962 dermal neurofibromas and pigmentation are
14(2):129-35. PMID:24926970 Diagnostic criteria for constitutional mismatch 2779. Wolter M, Reifenberger J, Blaschke caused by Nf1 loss in desert hedgehog-ex-
2750. Wiestler B, Capper D, Holland-Letz T, repair deficiency syndrome: suggestions of 8, lchimura K, Schmidt EE, Collins VP, et al. pressing cells. Cancer Cell. 13(2):105-16.
Korshunov A, van Deimling A, Pfister SM, et the European consortium 'care for CMMRD' (2001). Oligodendroglial tumors frequently PMID:18242511
al. (2013). ATRX loss refines the classification (C4CMMRD). J Med Genet. 51 (6):355-65. demonstrate hypermethylation of the CDKN2A 2794. Wu L, Yang T, Deng X, Yang C, Zhao
of anaplastic gliomas and identifies a subgroup PMID:24737826 (MTS1, p161NK4a), p14ARF, and CDKN2B L, Fang J, et al. (2014). Surgical outcomes in
of IDH mutant astrocytic tumors with better 2764. Winek RR, Scheithauer SW, Wick MR (MTS2, p151NK4b) tumor suppressor genes. spinal cord subependymomas: an institutional
prognosis. Acta Neuropathol. 126(3):443-51. (1989). Meningioma, meningeal hemangi- J Neuropathol Exp Neural. 60(12):1170-80. experience. J Neurooncol. 116(1):99-106.
PMID:23904111 opericytoma (angioblastic meningioma), pe- PMID:11764089 PMID:24062139
2751. Wiestler B, Capper D, Sill M, Jones DT, ripheral hemangiopericytoma, and acoustic 2780. Wong AJ, Signer SH, Signer DD, Kinz- 2795. Wu W, Tanrivermis Sayit A, Vinters HV,
Hovestadt V, Sturm D, el al. (2014). Integrated schwannoma. A comparative immunohistoche- ler KW, Hamilton SR, Vogelslein B (1987). Pope W, Mirsadraei L, Said J (2013). Primary
DNA methylalion and copy-number profiling mical study. Am J Surg Pathol. 13(4):251-61. Increased expression of the epidermal growth central nervous system histiocytic sarcoma pre-
identify three clinically and biologically relevant PMID:2648875 faclor receptor gene in malignant gliomas is senting as a postradiation sarcoma: case report
groups of anaplastic glioma. Acta Neuropalhol. 2765. Wippold F J 2nd, Smimiotopoulos JG, invariably associaled with gene amplification. and literature review. Hum Pathol. 44(6):1177-
128(4):561-71. PMID:25008768 Pilgram TK (1997). Lesions of the cauda equi- Proc Natl Acad Sci U SA. 84(19):6899-903. 83. PMID:23356953
2752. Wiestler OD, van Siebenlhal K, Schmitt na: a clinical and palhology review from the Ar- PMID:3477813 2796. Wu YT, Ho JT, Lin YJ, Lin JW (2011).
HP, Feiden W, Kleihues P (1989). Dislribution med Forces Institute of Pathology. Clin Neural 2781. Wong K, Gyure KA, Prayson RA, Mor- Rhabdoid papillary meningioma: a clinicopa-
and immunoreactivity of cerebral micro-ha- Neurosurg. 99(4):229-34. PMID:9491294 rison AL, Le TQ, Armstrong RC (1999). Dysem- thologic case series study. Neuropathology.
martomas in bilateral acoustic neurofibromat- 2766. Witkowski L, Lalonde E, Zhang J, Alb- bryoplastic neuroepithelial tumor: in situ hybri- 31 (6):599-605. PMID:21382093
osis (neurofibromatosis 2). Acta Neuropathol. recht S, Hamel N, Cavallone L, et al. (2013). Fa- dization of proteolipid protein (PLP) messenger 2797. W6hrer A, Waldh6r T, Heinzl H, Hackl
79(2):137-43 PMID:2596263 milial rhabdoid tumour 'avant la lettre'-from pa- ribonucleic acid (mRNA). J Neuropathol Exp M, Feichtinger J, Gruber-M6senbacher U, et
2752A. Wigertz A, Lonn S, Hall P, Auvinen A, thology review to exome sequencing and back Neural. 58:542-542. al. (2009). The Austrian Brain Tumour Regis-
Christensen HC, Johansen C, et al. (2008). Re- again. J Pathol. 231(1):35-43 PMID:23775540 2782. Wong KK, Tsang YT, Chang YM, Su J, try: a cooperative way to establish a populati-
productive factors and risk of meningioma and 2767. Witt H, Mack SC, Ryzhova M, Bender S, Di Francesco AM, Meco D, et al. (2006). Ge- on-based brain tumour registry. J Neurooncol.
glioma. Cancer Epidemiol Biomarkers Prev. Sill M, lsserlin R, el al. (2011). Delineation of nome-wide allelic imbalance analysis of pedi- 95(3):401-11. PMID:19562257
17(10):2663-70. PMID:18843008 two clinically and molecularly distinct subgroups atric gliomas by single nucleotide polymorphic 2798. Xekouki P, Stratakis CA (2012). Suc-
27528. Wigertz A, Lonn S, Schwartzbaum J, of posterior fossa ependymoma. Cancer Cell. allele array. Cancer Res. 66(23):11172-8. cinate dehydrogenase (SDHx) mulations in
Hall P, AuvinenA, Christensen HC, et al. (2007). 20(2):143-57. PMID:21840481 PMID:17145861 pituitary tumors: could this be a new role for
Allergic condilions and brain tumor risk. Am J 2768. Wizigmann-Voos S, Breier G, Risau W, 2783. Wong TT, Ho DM, Chang KP, Yen SH, mitochondrial complex II and/or Krebs cycle
Epidemiol. 166(8):941-50. PMID:17646205 Plate KH (1995). Up-regulation of vascular en- Guo WY, Chang FC, et al. (2005). Primary pe- defects? Endocr Reial Cancer. 19(6):C33-40.
2753. Wikslrand CJ, Reisl CJ, Archer GE, dothelial growth factor and its receptors in van diatric brain tumors: statistics of Taipei VGH, PMID:22889736
Zalulsky MR, Signer DD (1998). The class Ill Hippel-lindau disease-associated and sporadic Taiwan (1975-2004). Cancer. 104(10):2156- 2799. Xiao GH, Jin F, Yeung RS (1995). Identi-
variant of the epidermal growth factor recep- hemangioblastomas. Cancer Res. 55(6):1358- 67. PMID:16220552 fication of tuberous sclerosis 2 messenger RNA
tor (EGFRvlll): characlerization and ulilization 64. PMID:7533661 2784. Wong TT, Ho DM, Chang TK, Yang DD, splice variants that are conserved and differen-
as an immunotherapeulic large!. J Neurovirol. 2769. Wizigmann-Voos S, Plate KH (1996). Pa- Lee LS (1995). Familial neurofibromatosis 1 tially expressed in rat and human tissues. Cell
4(2):14!>-58. PMID:9584952 thology, genetics and cell biology of hemangi- with germinoma involving the basal ganglion Growth Differ. 6(9):1185-91. PMID:8519695
2754. Wilcox P, Li CC, Lee M, Shivalingam B, oblastomas. Histol Histopathol. 11(4):1049-61. and thalamus. Childs Nerv Syst. 11(8):456-8. 2800. Xiong J, Liu Y, Chu SG, Chen H, Chen
Brennan J, Suter CM, et al. (2015). Oligoast- PMID:8930647 PMID:7585682 HX, Mao Y, et al. (2012). Dysembryoplastic neu-
rocytomas: throwing the baby out with the 2770. Woehrer A, Hackl M, Waldhor T, Weis S, 2785. Woodruff JM, Christensen WN (1993). roepithelial tumor-like neoplasm of the septum
bathwater? Acta Neuropathol. 129(1):147-9. Pichler J, Olschowski A, et al.; Austrian Brain Glandular peripheral nerve sheath tumors. Can- pellucidum: review of 2 cases with chromosome
PMID:25304041 Tumour Registry (2014). Relative survival of cer. 72(12)361!>-28. PMID:8252477 1p/19q and IDH1 analysis. Clin Neuropathol.
2755. Willard N, Kleinschmidl-DeMasters BK patients with non-malignant central nervous 2786. Woodruff JM, Godwin TA, Erlandson RA, 31(1):31-8. PMID:22192702
(2015). Massive dissemination of adult gliob- system tumours: a descriptive study by the Susin M, Martini N (1981). Cellular schwanno- 2801. Xiong J, Liu Y, Chu SG, Chen H, Chen
laslomas. Clin Neuropathol. 34(6):330-42. Auslrian Brain Tumour Registry. Br J Cancer. ma: a variely of schwannoma sometimes mis- HX, Mao Y, et al. (2012). Rosette-forming gli-
PMID:26308254 110(2):286-96. PMID:24253501 taken for a malignant tumor. Am J Surg Pathol. oneuronal lumor of lhe septum pellucidum with
27 56. Williams D, Mori T, Reiter A, Woessman 2771. WoehrerA, Slave I, PeyrlA, Czech T, Dor- 5(8):733-44. PMID:7337161 extension to the supratentorial ventricles: rare
W, Rosolen A, Wrobel G, et al.; European Inter- fer C, Prayer D, et al. (2011). Embryonal tumor 2787. Woodruff JM, Marshall ML, Godwin TA, case with genetic analysis. Neuropathology.
group for Childhood Non-Hodgkin Lymphoma, with abundant neuropil and true rosettes (ET- Funkhouser JW, Thompson NJ, Erlandson RA 32(3):301-5. PMID:22017246
lhe Japanese Pediatric Leukemia/Lymphoma ANTR) with loss of morphological but retained (1983). Plexiform (multinodular) schwannoma. 2802. Xu HM, Gutmann DH (1998). Merlin diffe-
Study Group (2013). Central nervous system genetic key features during progression. Acta A tumor simulating the plexiform neurofibroma. rentially associates with the microtubule and ac-
involvement in anaplastic large cell lymphoma Neuropathol. 122(6):787-90. PMID:22057788 Am J Surg Pathol. 7(7):691-7. PMID:6638259 tin cytoskeleton. J Neurosci Res. 51(3):403-15.
in childhood: results from a multicentre Europe- 2772. Wolf HK, Buslei R, Blurncke I, Wiest- 2788. Woodruff JM, Scheithauer BW, Kurt- PMID:9486775
an and Japanese study. Pediatr Blood Cancer. ler OD, Pietsch T (1997). Neural antigens in kaya-Yapicier 0, Raffel C, Amr SS, LaQuaglia 2803. Xu J, Yang Y, Liu Y, Wei M, Ren J, Chang
60(10):E118-21. PMID:23720354 oligodendrogliomas and dysembryoplastic MP, et al. (2003). Congenital and childhood Y, et al. (2012). Rosette-forming glioneuronal
2757. Williams SR, Joos BW, Parker JC, Parker neuroepilhelial tumors. Acta Neuropathol. plexiform (multinodular) cellular schwannoma: tumor in lhe pineal gland and the third ventricle:
JR (2008). Papillary glioneuronal tumor: a case 94(5):436-43. PMID:9386775 a troublesome mimic of malignant periphe- a case with radiological and clinical implica-
report and review of the literature. Ann Clin Lab 2773. Wolf HK. Buslei R, Schmidl-Kaslner R, ral nerve sheath tumor. Am J Surg Pathol. tions. Quant Imaging Med Surg. 2(3):227-31.
Sci. 38(3):287-92. PMID:18715860 Schmidl-Kaslner PK, Pietsch T, Wiestler OD, et 27(10):1321-9. PMID:14508393 PMID:23256084
2758. Willis B, Ablin A, Weinberg V, Zager S, al. (1996). NeuN: a useful neuronal marker for 2789. Woodruff JM, Selig AM, Crowley K, Al· 2804. Xu W, Yang H, Liu Y, Yang Y, Wang P, Kim
Wara WM, Matthay KK (1996). Disease course diagnostic histopathology. J Histochem Cyto- len PW (1994). Schwannoma (neurilemoma) SH, el al. (2011). Oncometabolile 2-hydroxyg-
and late sequelae of Langerhans' cell histio- chem. 44(10):1167-71. PMID:8813082 with malignant transformation. A rare, distincti- lutarate is a competitive inhibitor of a-kelog-
cytosis: 25-year experience at the University 2774. Wolf HK, Muller MB, Spanle M, Zentner ve peripheral nerve tumor. Am J Surg Pathol. lutarate-dependent dioxygenases. Cancer Cell.
of California, San Francisco. J Clin Oneal. J, Schramm J, Wiestler OD (1994). Ganglio- 18(9):882-95. PMID:8067509 19(1):17-30. PMID:21251613
14(7):2073-82. PMID:8683239 glioma: a detailed histopathological a�d im- 2790. Wrede 8, Hasselblatt M, Peters 0, Thall 2805. Xu X, Zhao J, Xu Z, Peng 8, Huang Q, Ar-
2759. Willis SN, Mallozzi SS, Rodig SJ, Cronk munohistochemical analysis of 61 cases. Acta PF, Kulluk T, Moghrabi A, et al. (2009). Atypical nold E, et al. (2004). Structures of human cyto-
KM, McArdel SL, Caron T, et al. (2009). The Neuropathol. 88(2):166-73. PMID:7985497 choroid plexus papilloma: clinical experience solic NADP-dependent isocitrate dehydrogena-
microenvironment of germ cell tumors harbors 2775. Wolf HK, Normann S, Green AJ, van Ba- in the CPT-SIOP-2000 study. J Neurooncol. se reveal a novel self-regulatory mechanism
a prominent antigen-driven humoral response. kel I, Blumcke I, Pietsch T, et al. (1997). Tube- 95(3):383-92. PMID:19543851 of activity. J Biol Chem. 279(32):33946-57.
J lmmunol. 182(5):3310-7. PMID:19234230 rous sclerosis-like lesions in epileptogenic hu- 2791. Wu G, Broniscer A, McEachron TA, Lu C, PMID:15173171
2760. Wilson BG, Roberts CW (2011). SWI/ man neocortex lack allelic loss at the TSC1 and Paugh BS, Becksfort J, et al.; St. Jude Child- 2806. Ya�argil MG, Curcic M, Kis M, Siegent-
SNF nucleosome remodellers and cancer. Nat TSC2 regions. Acta Neuropathol. 93(1):93-6. ren's Research Hospital-Washington Univer- haler G, Teddy PJ, Roth P (1990). Total remo-
Rev Cancer. 11(7):481-92. PMID:21654818 PMID:9006662 sity Pediatric Cancer Genome Project (2012). val of craniopharyngiomas. Approaches and
2761. Wilson BG, Wang X, Shen X, McKenna 2776. Wolff 8, Ng A, Roth D, Parthey K, War- Somatic histone H3 alterations in pediatric dif- long-term results in 144 patients. J Neurosurg.
ES, Lemieux ME, Cho Y J, et al. (2010). Epige- muth-Metz M, Eyrich M, et al. (2012). Pediatric fuse intrinsic pontine gliomas and non-brains- 73(1):3-11. PMID:2352020
netic antagonism between polycomb and SWI/ high grade glioma of the spinal cord: results tem glioblastomas. Nat Genet. 44(3):251-3. 2807. Yamada H, Haratake J, Narasaki T, Oda
SNF complexes during oncogenic transformati- of lhe HIT-GBM database. J Neurooncol. PMID:22286216 T (1995). Embryonal craniopharyngioma. Case
on. Cancer Cell. 18(4):316-28. PMID:20951942 107(1):139-46. PMID:21964697 2792. Wu G, Diaz AK, Paugh BS, Rankin SL, report of the morphogenesis of a craniopharyn-
2762. Wilson C, Bonnet C, Guy C, ldziaszczyk 2777. Wolff JE, Sajedi M, Brant R, Coppes MJ, Ju 8, Li Y, et al.; St. Jude Children's Rese- gioma. Cancer. 75(12):2971-7. PMID:7773950
S, Colley J, Humphreys V, et al. (2006). Tsc1 Egeler RM (2002). Choroid plexus tumours. Br arch Hospital-Washington University Pediatric 2808. Yamamoto K, Yamada K, Nakahara T,
haploinsufficiency without mammalian target J Cancer. 87(10):1086-91. PMID:12402146 Cancer Genome Project (2014). The genomic Ishihara A, Takaki S, Kochi M, et al. (2002).
of rapamycin activation is sufficient for renal 2778. Wolfsberger S, Fischer I, H6ftberger R, landscape of diffuse intrinsic pontine glioma Rapid regrowth of solitary subependymal giant
cyst formation in Tsc1 +/. mice. Cancer Res. Birner P, Slave I, Dieckmann K, et al. (2004). and pediatric non-brainstem high-grade glioma. cell astrocytoma-case report. Neural Med Chir
66(16):7934-8. PMID:16912167 Ki-67 immunolabeling index is an accurate pre- Nat Genet. 46(5):444-50. PMID:24705251 (Tokyo). 42(5):224-7. PMID:12064158
2763. Wimmer K, Kratz CP, Vasen HF, Caron dictor of outcome in patients with intracranial 2793. Wu J, Williams JP, Rizvi TA, Kordich JJ, 2809. Yamane Y, Mena H, Nakazato Y (2002).

References 399
lmmunohistochemical characterization of pine- 1 p/19q loss distinguish oligodendrogliomas PMID:16024619 Research Hospital-Washington University
al parenchymal tumors using novel monoclonal from other cancers. J Pathol. 226(1):7-16. 2840. Zagzag D, Lukyanov Y, Lan L, Ali MA, Pediatric Cancer Genome Project (2013).
antibodies to the pineal body. Neuropathology. PMID:22072542 Esencay M, Mendez 0, et al. (2006). Hypo- Whole-genome sequencing identifies genetic
22(2):66-76. PMID:12075938 2826. Yip S, Miao J, Cahill DP, lafrateAJ, Aldape xia-inducible factor 1 and VEGF upregulate alterations in pediatric low-grade gliomas. Nat
2810. Yan H, Parsons DW, Jin G, Mclendon K, Nutt CL, et al. (2009). MSH6 mutations arise CXCR4 in glioblastoma: implications for angio- Genet. 45(6):602-12. PMID:23583981
R, Rasheed BA, Yuan W, et al. (2009). IDH1 in glioblastomas during temozolomide therapy genesis and glioma cell invasion. Lab Invest. 2856. Zhang K, Lin JW, Wang J, Wu X, Gao H
and IDH2 mutations in gliomas. N Engl J Med. and mediate temozolomide resistance. Clin 86(12):1221-32. PMID:17075581 Hsieh YC, et al. (2014). A germline missens�
360(8):765-73. PMID:19228619 Cancer Res. 15(14):4622-9. PMID:19584161 2841. Zagzag D, Zhong H, Scalzitti JM, Laug- mutation in COQ6 is associated with suscepti-
2811. Yang C, Li G, Fang J, Wu L, Yang T, 2827. Yokota T, Tachizawa T, Fukino K, Tera- hner E, Simons JW, Semenza GL (2000). Ex- bility to familial schwannomatosis. Genet Med.
Deng X, et al. (2015). Clinical characteristics moto A, Kouno J, Matsumoto K, et al. (2003). pression of hypoxia-inducible factor 1 alpha in 16(10):787-92. PMID:24763291
and surgical outcomes of primary spinal pa- A family with spinal anaplastic ependymoma: brain tumors: association with angiogenesis, in- 2857. Zhang M, Wang Y, Jones S, Sausen M,
ragangliomas. J Neurooncol. 122(3):539-47. evidence of loss of chromosome 22q in tumor. J vasion, and progression. Cancer. 88(11):2606- McMahon K, Sharma R, et al. (2014). Somatic
PMID:25720695 Hum Genet. 48(11):598-602. PMID:14566482 18. PMID:10861440 mutations of SUZ12 in malignant peripheral
2812. Yang HJ, Kim JE, Paek SH, Chi JG, 2828. Yoo JH, Rivera A, Naeini RM, Yedururi 2842. Zajac V, Kirchhoff T, Levy ER, Horsley nerve sheath tumors. Nat Genet. 46(11):1170-
Jung HW, Kim DG (2003). The significance of S, Bayindir P, Megahead H, et al. (2008). Me- SW, Miller A, Steichen-Gersdorf E, et al. (1997). 2. PMID:25305755
gemistocytes in astrocytoma. Acta Neurochir lanotic paraganglioma arising in the temporal Characterisation of X;17(q12;p13) translocation 2858. Zhang ZK, Davies KP, Allen J, Zhu L,
(Wien). 145(12):1097-103, discussion 1103. horn following Langerhans cell histiocytosis. breakpoints in a female patient with hypomela- Pestell RG, Zagzag D, et al. (2002). Cell cycle
PMID:14663567 Pediatr Radial. 38(5):571-4. PMID:18196230 nosis of Ito and choroid plexus papilloma. Eur J arrest and repression of cyclin D1 transcription
2813. Yang I, Tihan T, Han SJ, Wrensch MR, 2829. Yoshimoto M, de Toledo SR, da Silva NS, Hum Genet. 5(2):61-8. PMID:9195154 by INl1/hSNF5. Mel Cell Biol. 22(16):5975-88.
Wieneke J, Sughrue ME, et al. (2010). CDS+ Bayani J, Bertoui AP, Stavale JN, et al. (2004). 2843. Zakrzewska M, Wojcik I, Zakrzewski K, PMID:12138206
T-cell infiltrate in newly diagnosed glioblasto- Comparative genomic hybridization analysis of Polis L, Grajkowska W, Roszkowski M, et al. 2859. Zheng H, Chang L, Patel N, Yang J, Lowe
ma is associated with long-term survival. J Clin pediatric adamantinomatous craniopharyngio- (2005). Mutational analysis of hSNF5/INl1 and L, Burns DK, et al. (2008). Induction of abnor-
Neurosci. 17(11):1381-5. PMID:20727764 mas and a review of the literature. J Neurosurg. TP53 genes in choroid plexus carcinomas. mal proliferation by nonmyetinating schwann
2814. Yang P, Kollmeyer TM, Buckner K, 101(1) Suppl:85-90. PMID:16206977 Cancer Genet Cytogenet. 156(2):179-82. cells triggers neurofibroma formation. Cancer
Bamlet W, Ballman KV, Jenkins RB (2005). 2830. Yoshimoto T, Takahashi-Fujigasaki J, ln- PMID:15642401 Cell. 13(2):117-28. PMID:18242512
Polymorphisms in GLTSCR1 and ERCC2 oshita N. Fukuhara N, Nishioka H, Yamada S 2844. Zaky W, Dhall G, Khatua S, Brown RJ, 2860. Zheng PP, Pang JC, Hui AB, Ng HK
are associated with the development of oli- (2015). TTF-1-positive oncocytic sellar tumor Ginn KF, Gardner SL, et al. (2015). Choroid (2000). Comparative genomic hybridization
godendrogliomas. Cancer. 103(11):2363-72. with follicle formation/ependymal differentia- plexus carcinoma in children: the Head Start detects losses of chromosomes 22 and 16 as
PMID 15834925 tion: non-adenomatous tumor capable of two experience. Pediatr Blood Cancer. 62(5):784-9. the most common recurrent genetic alterations
2815. Yang SY, Jin Y J, Park SH, Jahng TA, Kim different interpretations as a pituicytoma or a PMID:25662896 in primary ependymomas. Cancer Genet Cyto-
HJ, Chung CK (2005). Paragangliomas in the spindle cell oncocytoma. Brain Tumor Pathol. 2845. Zamecnik M, Michal M (2001 ). Perineuri- genet. 122(1):18-25. PMID:11104027
cauda equina region: clinicopathoradiologic fin- 32(3):221-7. PMID:25893822 al cell differentiation in neurofibromas. Report of 2861. Zhong H, De MarzoAM, Laughner E, Lim
dings in four cases. J Neurooncol. 72(1 ):49-55. 2831. Yoshimura J, Onda K, Tanaka R, Ta- eight cases including a case with composite pe- M, Hilton DA, Zagzag D, et al. (1999). Overex-
PMID:15803375 kahashi H (2003). Clinicopathological study rineurioma-neurofibroma features. Pathol Res pression of hypoxia-inducible factor 1 alpha in
2816. Yang X, Zeng Y, Wang J (2013). Hybrid of diffuse type brainstem gliomas: analysis of Pract. 197(8):537-44. PMID:11518046 common human cancers and their metastases.
schwannoma/perineurioma: report of 10 Chi- 40 autopsy cases. Neural Med Chir (Tokyo). 2846. Zang KD (2001 ). Meningioma: a cyto- Cancer Res. 59(22):5830-5. PMID:10582706
nese cases supporting a distinctive entity. Int J 43(8):375-82, discussion 382. PMID:12968803 genetic model of a oomplex benign human 2862. Zhou H, Coffin CM, Perkins SL, Tripp
Surg Pathol. 21(1):22-8. PMID:22832113 2832. You H, Kim YI, Im SY, Suh-Kim H, Paek tumor, including data on 394 karyotyped ca- SR, Liew M, Viskochil DH (2003). Malignant
2817. Yang ZJ, Ellis T, Markant SL, Read SH, Park SH, et al. (2005). lmmunohistochemi- ses. Cytogenet Cell Genet. 93(3-4):207-20. peripheral nerve sheath tumor: a comparison
TA, Kessler JD, Bourboulas M, et al. (2008). cal study of central neurocytoma, subependy- PMID:11528114 of grade, immunophenotype, and cell cycle/
Medulloblastoma can be initiated by deletion moma, and subependymal giant cell astrocyto- 2847. Zarate JO, Sampaolesi R (1999). Pleo- growth activation marker expression in sporadic
of Patched in lineage-restricted progenitors ma. J Neurooncol. 74(1):1-8. PMID:16078101 morphic xanthoastrocytoma of the retina. Am J and neurofibromatosis 1-related lesions. Am J
or stem cells. Cancer Cell. 14(2):135-45. 2833. Young RJ, Sills AK, Brem S, Knopp EA Surg Pathol. 23(1 ):79-81. PMID:9888706 Surg Pathol. 27(10):1337-45. PMID:14508395
PMID:18691548 (2005). Neuroimaging of metastatic brain disea- 2848. Zaveri J, La Q, Yarmish G, Neuman J 2862A. Zhou J, Shrikhande G, Xu J, McKay
2818. Yasargil MG, van Ammon K, von Deim- se. Neurosurgery. 57(5) Suppl:S10-23, S1-4. (2014). More than just Langerhans cell his- RM, Burns DK, Johnson JE, Parada LF (2011).
ling A, Valavanis A, Wichmann W, Wiestler OD PMID:16237282 tiocytosis: a radiologic review of histiocytic Tsc1 mutant neural stem/progenitor cells exhibit
(1992). Central neurocytoma: histopathological 2834. Yu J, Deshmukh H, Payton JE, Dunham disorders. Radiographies. 34(7):2008-24. migration deficits and give rise to subependy-
variants and therapeutic approaches. J Neuro- C, Scheithauer BW, Tihan T, et al. (2011). Ar- PMID:25384298 mal lesions in the lateral ventricle. Genes Dev.
surg. 76(1):32-7. PMID:1727166 ray-based comparative genomic hybridization 2848A. Zawlik I, Kita D, Vaccarella S, Mittel- 25(15):1595-600. PMID:21828270
2819. Yasha TC, Mohanty A, Radhesh S, identifies CDK4 and FOXM1 alterations as bronn M, Franceschi S, Ohgaki H (2009). Com- 2863. Zhou X, Hampel H, Thiele H, Gorlin
Santosh V, Das S, Shankar SK (1998). ln- independent predictors of survival in malignant mon polymorphisms in the MDM2 and TP53 RJ, Hennekam RC, Parisi M, et al. (2001).
fratentorial dysembryoplastic neuroepithelial peripheral nerve sheath tumor. Clin Cancer genes and the relationship between TP53 muta- Association of germline mutation in the PTEN
tumor (DNT) associated with Arnold-Chiari Res. 17(7):1924-34. PMID:21325289 tions and patient outcomes in glioblastomas. tumour suppressor gene and Proteus and Pro-
malformation. Clin Neuropathol. 17(6):305-10. 2835. Yu T, Sun X, Wang J, Ren X, Lin N, Lin Brain Pathol. 19(2):188-94. PMID:18462472 teus-like syndromes. Lancet. 358(9277):210-1.
PMID:9832257 S (2015). Twenty-seven cases of pineal paren- 2849. Zbuk KM, Eng C (2007). Cancer pheno- PMID:11476841
2820. Yassa M, Bahary JP, Bourguoin P, Belair chymal tumours of intermediate differentiation: mics: RET and PTEN as illustrative models. Nat 2864. Zhou XP, Marsh DJ, Morrison CD, Chau-
M. Berthelet F, Bouthillier A (2005). lntra-pa- mitotic count, Ki-67 labelling index and extent of Rev Cancer. 7(1):35-45. PMID:17167516 dhury AR, Maxwell M, Reifenberger G, et al.
renchymal mesenchymal chondrosarcoma resection predict prognosis. J Neural Neurosurg 2850. Zbuk KM, Eng C (2007). Hamartomatous (2003). Germline inactivation of PTEN and dys-
of the cerebellum: case report and review of Psychiatry. PMID:25911570 polyposis syndromes. Nat Clin Pract Gastroen- regulation of the phosphoinositol-3-kinase/Akt
the literature. J Neurooncol. 74(3):329-31. 2836. Zacharia BE, Bruce SS, Goldstein H, terol Hepatol. 4(9):492-502. PMID:17768394 pathway cause human Lhermitte-Duclos disea-
PMID:16187026 Malone HR, Neugut Al, Bruce JN (2012). Inci- 2851. Zevallos-Giampietri EA, Yaiies HH, se in adults. Am J Hum Genet. 73(5):1191-8.
2821. Yeh-Nayre LA, Malicki DM, Vinocur DN, dence, treatment and survival of patients with Orrego Puelles J, Barrionuevo C (2004). Pri- PMID:14566704
Crawford JR (2012). Medulloblastoma with craniopharyngioma in the surveillance, epide- mary meningeal Epstein-Barr virus-related 2865. Zhou XP, Waite KA, Pilarski R, Hampel
excessive nodularity: radiographic features miology and end results program. Neuro Oneal. leiomyosarcoma in a man infected with human H, Fernandez MJ, Bos C, et al. (2003). Germ-
and pathologic correlate. Case Rep Radiol. 14(8):1070-8. PMID:22735773 immunodeficiency virus: review of literature, line PTEN promoter mutations and deletions
2012:310359. PMID:23133782 2837. Zada G, Lin N, Ojerholm E, Ramkisso- emphasizing the differential diagnosis and pa- in Cowden/Bannayan-Riley-Ruvalcaba syn-
2822. Yi KS, Sohn CH, Yun TJ, Choi SH, Kim on S, Laws ER (2010). Craniopharyngioma thogenesis. Appl lmmunohistochem Mol Mor- drome result in aberrant PTEN protein and
JH, Han MH, et al. (2012). MR imaging findings and other cystic epithelial lesions of the sellar phol. 12(4):387-91. PMID:15536343 dysregulation of the phosphoinositol-3-kinase/
of extraventricular neurocytoma: a series of region: a review of clinical, imaging, and his- 2852. Zhang CZ, Leibowitz ML, Pellman D Akt pathway. Am J Hum Genet. 73(2):404-11.
ten patients confirmed by immunohistoche- topathological relationships. Neurosurg Focus. (2013). Chromothripsis and beyond: rapid ge- PMID:12844284
mistry of IDH1 gene mutation. Acta Neurochir 28(4):E4. PMID:20367361 nome evolution from complex chromosomal 2866. Zhu HD, Xie Q, Gong Y, Mao Y, Zhong P,
(Wien). 154(11):1973-9, discussion 1980. 2838. Zagzag D, Esencay M, Mendez 0, Yee rearrangements. Genes Dev. 27(23):2513-30. Hang FP, et al. (2013). Lymphoplasmacyte-rich
PMID:22945896 H, Smirnova I, Huang Y, et al. (2008). Hypoxia- PMID:24298051 meningioma: our experience with 19 cases and
2823. Yildiz H, Hakyemez B, Koroglu M, Ye- and vascular endothelial growth factor-induced 2853. Zhang F, Tan L, Wainwright LM, Barto- a systematic literature review. Int J Clin Exp
sildag A, Baykal B (2006). lntracranial lipomas: stromal cell-derived factor-1alpha/CXCR4 lomei MS, Biegel JA (2002). No evidence for Med. 6(7):504-15. PMID:23936588
importance of localization. Neuroradiology. expression in glioblastomas: one plausible ex- hypermethylation of the hSNF5/INl1 promoter 2867. Zhu J, Frosch MP, Busque L, Beggs AH,
48(1):1-7. PMID:16237548 planation of Scherer's structures. Am J Pathol. in pediatric rhabdoid tumors. Genes Chromoso- Dashner K, Gilliland DG, et al. (1995). Analy-
2824. Yin XL, Hui AB, Pang JC, Poon WS, Ng 173(2):545-60. PMID:18599607 mes Cancer. 34(4):398-405. PMID:12112529 sis of meningiomas by methylation- and tran-
HK (2002). Genome-wide survey for chromo- 2839. Zagzag D, Krishnamachary B, Yee H, 2854. Zhang J, Cheng H, Qiao Q, Zhang JS, scription-based clonality assays. Cancer Res.
somal imbalances in ganglioglioma using com- Okuyama H, Chiriboga L, Ali MA, et al. (2005). Wang YM, Fu X, et al. (2010). Malignant solitary 55(17):3865-72. PMID:7641206
parative genomic hybridization. Cancer Genet Strom al cell-derived factor-1 alpha and CXCR4 fibrous tumor arising from the pineal region: 2868. Zhu L, Ren G, Li K, Liang ZH, Tang WJ,
Cytogenet. 134(1):71-6. PMID:11996800 expression in hemangioblastoma and clear case study and literature review. Neuropatho- Ji YM, et al. (2011 ). Pineal parenchymal tu-
2825. Yip S, Butterfield YS, Morozova 0, Chit- cell-renal cell carcinoma: von Hippel-Lindau logy. 30(3):294-8. PMID:19845865 mours: minimum apparent diffusion coefficient
taranjan S, Blough MD, An J, et al. (2012). loss-of-function induces expression of a ligand 2855. Zhang J, Wu G, Miller CP, Tatevossian in prediction of tumour grading. J Int Med Res.
Concurrent CIC mutations, IDH mutations, and and its receptor. Cancer Res. 65(14):6178-88. RG, Dalton JD, Tang B,etal.; St. Jude Children's 39(4):1456-63. PMID:21986148

400 References
2869. Zhu Y, Ghosh P, Charnay P, Burns
DK, Parada LF (2002). Neurofibromas in
NF1: Schwann cell origin and role of tumor
environment. Science. 296(5569):920-2.
PMID:11988578
2870. Zhukova N, Ramaswamy V, Remke M,
Pfaff E, Shih DJ, Martin DC, et al. (2013). Sub-
group-specific prognostic implications of TP53
mutation in medulloblastoma. J Clin Oncol.
31(23):2927-35. PMID:23835706
2871. Zlatescu MC, TehraniYazdi A, Sasaki
H, Megyesi JF, Betensky RA, Louis ON, et
al. (2001). Tumor location and growth pattern
correlate with genetic signature in oligodendro-
glial neoplasms. Cancer Res. 61(18):6713-5.
PMID 11559541
2872. Zong H, Parada LF, Baker SJ (2015). Cell
of origin for malignant gliomas and its implica-
tion in therapeutic development. Cold Spring
Harb Perspect Biol. 7(5) PMID:25635044
2873. Zorludemir S, Scheithauer BW, Hirose
T, Van Houten C, Miller G, Meyer FB (1995).
Clear cell meningioma. A clinicopathologic
study of a potentially aggressive variant of
meningioma.Am J Surg Pathol. 19(5):493-505.
PMID: 7726360
2874. Zou C, Smith KO, Liu J, Lahat G, Myers
S, Wang WL, et al. (2009). Clinical, pathologi-
cal, and molecular variables predictive of mali-
gnant peripheral nerve sheath tumor outcome.
Ann Surg. 249(6):1014-22. PMID:19474676
2875. Zuccaro G, TaratutoAL, Monges J (1986).
lntracranial neoplasms during the first year of
life. Surg Neurol. 26(1):29-36. PMID:3715697
2876. Zulch KJ (1957). Brain Tumours.
Their Biology and Pathology. New York:
Springer-Verlag.
2877. Zygourakis CC, Rolston JD, Lee HS, Par-
low C, Kunwar S,Aghi MK (2015). Pituicytomas
and spindle cell oncocytomas: modern case se-
ries from the University of California, San Fran-
cisco. Pituitary. 18(1):150-8. PMID:24823438
2878. Ziilch KJ, editor (1979). Histological ty-
ping of tumours of the central nervous system.
1st Edition Geneva: World Health Organization.
2879. Zoller ME, Rembeck B, Oden A, Samu-
elsson M, Angervall L (1997). Malignant and be-
nign tumors in patients with neurofibromatosis
type 1 in a defined Swedish population. Cancer.
79(11 ):2125-31. PMID:9179058

References 401
Subject index

1p34 319-321 Alpha-1-antitrypsin 38, 84, 330 Basal cell naevus syndrome 319
1p35-36 143 Alpha-B-crystallin 83 B-cell receptor 274, 275
2-hydroxyglutarate 21, 22, 26, 62, 67 Alpha-fetoprotein 289, 290 BCL2 273,274,333,336
2p16 43, 317 Alpha-internexin 65, 69, 74, 157, 308 BCL6 273-275
2q32 317 Alpha-ketoglutarate-dependent Bcl-xl 45
3p21.3 43, 317 dioxygenases 67 BCOR-CCNB3 intrachromosomal inversion
3p25 86,304 Anaplastic astrocytoma, IDH-mutant 18, 21, 260
4E-BP1 309 24-27, 54,55, 76, 77 BCORL1 287
4q12 146 Anaplastic astrocytoma, IDH-wildtype 8, 10, Benign fibrous histiocytoma 261
5-HT 166, 171, 172 23, 25. 27,30 Beta-catenin 37, 188, 189, 193, 300, 318,
5-methylcytosine hydroxylase 67 Anaplastic astrocytoma, NOS 27 324-328
5q11-q13 317 Ana plastic ependymoma 106, 108, 113, 202 Beta-hCG 288-290
5q13.3 146 Anaplastic ganglioglioma 138, 141 BIRC5 229
6p21 274 Anaplastic large cell lymphoma 276, 277 BK virus 61, 272
6q21 274 Anaplastic (malignant) meningioma 244 BLNK 274
7p22 43,317 Anaplastic oligoastrocytoma. dual-genotype BMPR1A 315, 316
7q31 146 77 BNIP 37
7q34 80.86,96 Anaplastic oligoastrocytoma, NOS 76 Brachyury 256
8p22-pter 146 Anaplastic oligodendroglioma 33, 34, 61, 67, BRAF mutations 16, 86, 96, 140, 341
8q12.1-q12.2 274 68, 70-74, 76 Brain tumour-polyposis syndrome 1 I
9p21.3 96 Anaplastic oligodendroglioma, I DH-mutant Mismatch repair cancer syndrome 317
9q22 191,319,320 and 1p/19q-codeleted 34, 61, 70, 72-74, Brain tumour-polyposis syndrome 2 I Familial
10q21.3 146 76 adenomatous polyposis 318
10q23.21 274 Anaplastic oligodendroglioma lacking IDH BRCA1 67, 236
10q24 319,320 mutation and 1 p/19q codeletion 7 4 BRG1 206, 208, 209, 211, 212
11q23 143 Anaplastic oligodendroglioma, NOS 72, 74 BRIP1 236
12q14.3 146 Anaplastic pleomorphic xanthoastrocytoma BTP1 See Brain tumour-polyposis
13q21 146 98, 99 syndrome 1 I Mismatch repair cancer
16p13.3 309 Ancient neurofibroma 220 syndrome
17q11.2 294, 295 Angiocentric glioma 119, 120 BTP2 See Brain tumour-polyposis
18q21.33-q23 27 4 Angiocentric neuroepithelial tumour 119 syndrome 2 I Familial adenomatous
19p13.2 322 Angiolipoma 260 polyposis
21q22.11 146 Angiomatous meningioma 233, 238, 239 Butterfly glioma 30
22q11.23 218, 302, 322 Angiopoietin 37
34betaE12 105 Angiosarcoma 259 c
Annexin A 1 336 C11orf95 108, 109, 112, 206
A Antoni A pattern 215 C19 286
Abrikossott tumour 329 Antoni B pattern 215 C228T TERT mutation 235
ACKR3 37 AP181 235 C250T TERT mutation 235
Actin 38, 299, 300 APC 109, 163, 185, 189, 318 CADM1 235
ACVR1 59 Aquaporin-1 256 Cate-au-lait spots/macules 294-296, 299,
Adamantinomatous craniopharyngioma 324, ARHGAP35 66 317
327 ASMT 172, 174, 175, 178 CALCA 181
Adenoid glioblastoma 35 Astroblastoma 121, 122 CAM5.2 105, 122, 181, 256, 289
AE1/AE3 38, 51, 64, 105, 181,256, 289 Ataxia-telangiectasia 275 cAMP 218, 295
AIDS-related diffuse large B-cell lymphoma ATM 59 CAMTA1 66, 258
275 ATOH1 163, 191, 199 Carbonic anhydrase 37, 64, 256
AKT 31, 39,43, 143, 221, 287,295,300, 309, AT/RT See Atypical teratoid/rhabdoid tumour CARD11 274
316 Atypical choroid plexus papilloma 126, 127 CBL 87, 274, 287
AKT1 151,235,315 Atypical meningioma 241 CBTRUS See Central Brain Tumor Registry of
AKT1 E17K mutation 235 Atypical neurofibroma 220, 221 the United States
AKT2 151 Atypical teratoid/rhabdoid tumour 206, CCDC26 42
AKT3 151 209-212,321,322 CCND1 59, 211, 302, 322
AKT/mTOR pathway 43, 221, 287, 295, 316 AURKA 211 CCND2 21, 287
ALCL See Primary CNS anaplastic large cell AURKB 46, 47 CCNE2 229
lymphoma, ALK-positive CD1a 280-283
ALDH1 253 8 CD2 276
ALDH1A1 253 Bannayan-Riley-Ruvalcaba syndrome 314, CD3 240, 275, 276
ALK 261,277,283,341 316 CD4 38, 276, 289
Alpha-1-antichymotrypsin 38, 84, 330 BAP1 270 CD5 276, 277

402 Subject index


CD? 276 CIC-DUX4 intrachromosomal inversion 260 Diffuse leptomeningeal leiomyoma 262
CD8 38, 276, 289 CITED4 66 Diffuse leptomeningeal
CD10 256,273, 276, 277 CK5/6 105, 325, 326, 341 oligodendrogliomatosis 61
CD11c 282, 283 CK? 105, 125.244,325,328,341 Diffuse midline glioma, H3 K27M-mutant 57,
CD14 283 CK14 325 58
CD15 276 CK17 325 DIG See Desmoplastic infantile ganglioglioma
CD19 273 CK18 180, 181 DiGeorge syndrome 97
CD20 273, 274, 276, 289 CK19 325 DIPG See Diffuse intrinsic pontine glioma
CD22 273 CK20 105, 125, 341 DIRAS3 66
CD23 276, 283 Class Ill beta-tubulin 91, 96, 146, 149, 157, Disseminated oligodendroglioma-like
CD24 175, 179 171, 178, 187, 308 leptomeningeal neoplasm 152
CD28 44 Claudin-1 224 DLBCL See Diffuse large B-cell lymphoma
CD29 276 CLCN6 86 DMBT1 141
CD30 276, 277, 283, 289 Clear cell ependymoma 60, 65, 68, 106, 108, DNA hypermethylation 56, 87, 274
CD31 37,84,256,258,259 111 DNT See Dysembryoplastic neuroepithelial
CD35 283 Clear cell meningioma 233, 241 tumour
CD38 273 Clear cell renal cell carcinoma 304 Dorsal exophytic brain stem glioma 80
CD43 300 CNS embryonal tumour, NOS 208 Down syndrome 287
CD44 122, 126,300,336 CNS embryonal tumour with rhabdoid Drosophila capicua gene 66
CD45 276, 282 features 212 Dysembryoplastic neuroepithelial tumour 60,
CD54 276 CNS ganglioneuroblastoma 136, 206, 207, 65, 68, 84,87, 132, 133, 139, 140, 159,
CD56 181, 256, 341 208 160
CD68 35,240,280-283,330 CNS neuroblastoma 206, 207 Dysplastic cerebellar gangliocytoma
CD79a 273 Coffin-Siris syndrome 321 (Lhermitte-Duclos disease) 136, 142,
CD79B 274 Collagen IV 84, 146, 202, 216-218, 223, 266 143,314
CD80 44 Conventional chondrosarcoma 263 Dysplastic gangliocytoma 143
CD86 44 COQ6 302
CD99 105,203,253,259 Corticoid-mitigated lymphoma 275 E
CD133 54, 185 Cowden syndrome 136, 142, 143, 236, EAAT1 125
CD138 273, 289 314-316 EBV 11, 248, 262, 272,273, 275, 276
CD163 35, 283 Craniopharyngioma 324 EBV+ diffuse large B-cell lymphoma, NOS
CD207 280 CRB3 172 276
CD276 44 CRX 172, 174, 178 EED 229
CDC42 31 CTDNEP1 193 EGFRvlll 38, 39, 44, 175
CDK4 40, 49, 59, 96, 118, 141, 229 CTNNB1 163, 185, 188, 189, 254, 324, 326, EIF1AX 270
CDK6 193 327 Embryonal carcinoma 289
CDKN1A 179 Cullin-2 305 Embryonal tumour with abundant neuropil
CDKN1B 305 CXCR4 37, 256 and true rosettes 201, 202
CDKN2A I CDK4 / retinoblastoma protein Cys176 312 Embryonal tumour with multilayered rosettes,
pathway 40 C19MC-altered 201
CDKN2B 28, 40, 42, 67, 73, 96, 221 D Embryonal tumour with multilayered rosettes,
CDKN2C 72, 73 02-40 256, 289, 341 NOS 205
CDR3 272, 275 DAPK1 274 EMP3 66
CDX2 341 DDX3X 185, 188-190, 197 Encephalocraniocutaneous lipomatosis 249
Cellular schwannoma 216 der(1 ;19)(p10;q10} 66 Endolymphatic sac tumour 304, 305
Central Brain Tumor Registry of the United der[t(1; 19)(q10;p10)] 66 EORTC See European Organisation for
States 61, 70, 80, 94, 102, 104, 206, 252 Desmin 51, 163, 187, 228, 229, 253, 260, Research and Treatment of Cancer
Central neurocytoma 111, 156-160 262, 330 Eosinophilic granular bodies 82, 83, 139
Cerebellar astrocytoma 80 Desmoid-type fibromatosis 260 EPB41L3 235
Cerebellar liponeurocytoma 156, 161-163 Desmoplakin 65 Ependymal rosettes 108
CHD5 66 Desmoplastic infantile astrocytoma 144 Ependymoblastoma 202
CHEK2 59 Desmoplastic infantile ganglioglioma 144, Ependymoma 106-109, 111, 112,298
Chiari malformation 155 202 Ependymoma, RELA fusion-positive 112
Chompret criteria 310 Desmoplastic/nodular medulloblastoma 184, Epidermal growth factor 31, 44
Chondroma 262 195-197,319 Epithelioid glioblastoma 50, 51
Chondrosarcoma 21, 244, 248, 253, 263, DFFB 66 Epithelioid haemangioendothelioma 258
322 DIA See Desmoplastic infantile astrocytoma Epithelioid malignant peripheral nerve sheath
Chordoid glioma of the third ventricle DICER1 germline mutation 179 tumour 228
116-118 EPO 305
Chordoid meningioma 233, 240, 241 Diffuse astrocytoma, IDH-mutant 18-25, ERBB2 341
Choriocarcinoma 290 53-55,64-66, 75 ERCC2 67
Choristoma 262, 329 Diffuse astrocytoma, IDH-wildtype 23 Erdheim-Chester disease 281
Choroid plexus carcinoma 126-128, 129, Diffuse astrocytoma, NOS 23 ERG 258, 259
210, 311, 312, 321 Diffuse intrinsic pontine glioma 57 Erythropoietin 37, 255, 257
Choroid plexus papilloma 124 Diffuse large B-cell lymphoma of the CNS ESR1 120
Chromatin-related genes 40 272 ESRG 289
Chromothripsis 109, 112, 190, 192, 200 Diffuse leptomeningeal glioneuronal tumour ETMR See Embryonal tumour with
CIC 66, 67,69, 72-74, 260 152, 154, 155, 159 multilayered rosettes

Subject index 403


ETV6-NTRK3 fusion 96 Glial hamartias 299 Homer \Nright rosettes 33, 157, 177, 187,
European Organisation for Research and Glioblastoma, IDH-mutant 18, 21-25, 29, 194,207, 208, 259
Treatment of Cancer 21, 69, 73 32-34,38,39, 47,52-56 HOXD13 175, 179
Everolimus 93 Glioblastoma, IDH-wildtype 21, 25, 27, 28, HRAS 295
EVl2A 295 29, 31, 32, 34,36,38-41,43, 46-49, Human placental lactogen 289, 290
EV12B 295 52-56, 76 Hybrid nerve sheath tumours 224
Ewing sarcoma/ peripheral primitive Glioblastoma, NOS 28, 56 Hypermethylation phenotype 55
neuroectodermal tumour 249, 253, 259, Glioblastoma with a primitive neuronal
260 component 32, 33 I
E\NSR1 249, 253, 259 Glioblastoma with epithelial metaplasia 35 ICAM1 276, 300
Extranodal marginal zone lymphoma of Gliomatosis cerebri growth pattern 23, 27, 30 ICAM2 300
mucosa-associated lymphoid tissue Gliosarcoma 35, 36, 38. 47, 48, 49, 56, 67, IDH1 R132H mutation 20, 23, 27, 34, 42, 54,
(MALT lymphoma) of the dura 277 76, 260, 262, 264 66, 68,69, 74
Extraventricular neurocytoma 159 GLTSCR1 67 lgA deficiency 275
EZH2 40,41,211,322 GLUT1 220,224,225,258 lgD 273
Ezrin 256, 299, 308 Glycerol-3-phosphate dehydrogenase 64 IGF1 44, 67
GNA 11 86, 269, 270 IGF2/H19 287
F GNAQ 151, 269, 270 lgG 273
Factor V Leiden mutation 155 GNAS 151 lgG4-related disease 261
Factor Xllla 283 Gorlin-Goltz syndrome 319 IGH 272, 275, 283
FAK 31,300.316 Gorlin syndrome 188, 192, 195, 197, 198, lgM 273
FAK/SRC pathway 300 236,319 IL2 44
FAM131B 86 Granular cell astrocytoma 35 IL8 37, 38
Familial adenomatous polyposis 179, 318 Granular cell glioblastoma 34, 35 IL8R 37
Familial posterior fossa brain tumour Granular cell myoblastoma 329 IL 17RB 300
syndrome of infancy 321 Granular cell neuroma 329 Immature teratoma 291
FGFR1 23, 39,59,60.68, 84, 86-88, 149, Granular cell tumour of the sellar region 329 Immunodeficiency-associated CNS
151 GSTI1 67 lymphomas 275
FGFR1-TACC1 fusion 39, 86 Inflammatory myofibroblastic tumour 261
FGFR3-TACC3 fusion 39 H lnfundibuloma 332
Fibrillary astrocytoma 18, 85, 139 H3.3 G34R mutation 42 lnhibin alpha 256
Fibroblast growth factor 31, 67 H3F3A 40-42,51, 57,58, 135 INl1 177, 203,206, 208-212, 217, 225, 228
Fibrosarcoma 261 H3K4 41 INPP50 274
Fibrous meningioma 237. 238, 253 H3K27me3 40,59,211,229,322 Intestinal hamartomatous polyps 314
Fibrous xanthoma 261 H3 K27M-mutant diffuse midline glioma 57, lntracranial calcifications 299
Fibroxanthoma 261 58 lntravascular large B-cell lymphoma 276
Fifth phacomatosis 319 H3 K27M mutation 23, 28, 38, 59 ITGB4 229
Flexner-\Nintersteiner rosettes 177 H3K36me3 41
FL11 259 Haemangioblastoma 254-256, 304 J
Follicular lymphoma 276 Haemangioma 258 JAK 45
FUBP1 66,69, 72-74 Haemangiopericytoma See Solitary fibrous JC virus 61
FXR1 86, 140 tumour / haemangiopericytoma JMJD1C 288
HDAC2 41 Juvenile xanthogranuloma 282, 283, 295
G HDAC9 41
G34 mutations 40-42, 206 Hepatocyte growth factor 31, 37, 146, 305 K
GAB1 191, 193, 197, 199 Hereditary non-polyposis colorectal cancer K27 mutation 40, 41, 58, 206
GABP 40 67 K27M-mutant 28, 38, 59, 134
Galactocerebroside 64 HGF 229 Kaposi sarcoma 259
Galectin-3 336 HHV6 272 KDM4D 41
Gangliocytoma 136 HHV6A 61 KDM5A/B/C 41
Ganglioglioma 84, 87, 133-136, 138, HHV8 259, 272 KDM6A 41, 185, 193
139-141, 155, 159 Hibernoma 260 KDM6A/B 41
GAP 87, 295 HIF1 31,103,305 KOR 146
GCDFP15 341 HIF1A 36-38, 256, 257,305 KIAA1549-BRAFfusion 84-87, 89, 154, 155.
G-CIMP 21, 41,67, 73, 74, 76 HIF2A 256, 257 331,333,336
Gemistocytic astrocytoma 21-23 High-grade astrocytoma 24 KIR7.1 124, 125, 127-129. 181, 341
Gemistocytic astrocytoma, IDH-mutant 22 HIST1H3B 40, 42,51,57-59 KIT 146, 220. 270, 287,289-291
Germinoma 288, 289 HIST1H3B/C 40, 57 KL 1 181
GFl1 185, 193 HIST1H3B K27M mutation 42 KLF4 235,239,326
GF11B 185, 193 HIST1H3C 58, 59 KLHL 14 273
Giant cell glioblastoma 35, 43, 46, 47, 56, Histiocytic sarcoma 283 KLLN 314,315
76,317 HIV 61,259, 275, 276 KMT2B 41
GJB1 122 HLA-A/B/C 273 KMT2C 41, 193
GJB2 122 HLA-DOA 274 KMT2D 41. 189, 190, 193
Glandular malignant peripheral nerve sheath HLA-DOB 274 KRAS 23,84,86,87, 151,295,308
tumour 227 HLA-DR 273, 280 Kynurenine 44, 45
GLI 191, 319, 320 HLA-DRB 274
GLl2 185, 190-192, 197, 200 HMB45 187, 228, 266, 341

404 Subject index


L MAP2K 1 137, 280 MRAS 295
L 1 antigen 282 Mature teratoma 291 MSH2 43, 317, 318
L1CAM 108, 112, 206 MAX 167 MSH3 43, 317
L2HGDH 43 MBEN See Medulloblastoma with extensive MSH4 313
L-2-hydroxyglutaric aciduria 42 nodularity MSH6 43, 69, 317, 318
Lactate dehydrogenase 37, 64 MDM2 39, 40, 49, 96, 103, 118, 129. 146 mTOR1 93
Laminin 84, 217, 218, 223 MDM4 39, 191 mTORC1 300
Langerhans cell histiocytosis 280, 281 Medulloblastoma. classic 184, 194 MUC4 224
Langerin 280, 282 Medulloblastoma, group 3 184, 193 Multifocal glioblastoma 31
LARGE 235 Medulloblastoma, group 4 184, 193 Multinodular and vacuolating neuronal tumour
Large cell I anaplastic medulloblastoma 184, Medulloblastoma, non-WNT/non-SHH 184, of the cerebrum 137
200 193 Multiple endocrine neoplasia type 2 167
LATS1 313 Medulloblastoma, NOS 184, 186 Multiple germline mutations 313
LDB1 197 Medulloblastoma. SHH-activated and Multiple neurilemmomas 301
LDB2 141 TP53-mutant 184, 190 MUM1/IRF4 273
Leiomyoma 262 Medulloblastoma, SHH-activated and MYB 23, 60, 68, 120
Leiomyosarcoma 262 TP53-wildtype 184, 190, 191 MYC 21,34,59, 185, 193,200,273
Leptomyelolipomas 260 Medulloblastoma with extensive nodularity MYCL 190, 191
LEU4 143 184, 198, 199,319 MYCN 34,59, 158, 185, 190-193, 197,200
LEU? 64,217,256 Medulloblastoma, WNT-activated 184.188, MYC-PVT1 fusion 193
LFL-8 310 189,318 MYD88 274
LFL-E2 310 Medullocytoma 161 Myelin-associated glycoprotein 64
LFS See Li-Fraumeni syndrome Medulloepithelioma 201, 202-204, 206, 207 Myelin basic protein 64
Lhermitte-Duclos disease 136, 142, 143, 314 MEG3 235 Myofibroblastoma 260
Li-Fraumeni syndrome 21, 42, 43, 54, 56, 59, Melan-A 51, 187, 228, 266, 341 Myogenin 187, 262
128, 188, 191, 192, 197, 310, 317 Melanotic neuroectodermal tumour of infancy Myxopapillary ependymoma 104, 105, 109
LIN28A 202,204, 206,207, 289, 290 (retinal anlage tumour) 266 167
Lipidized glioblastoma 35 Melanotic schwannoma 218
Lipidized mature neuroectodermal tumour of Meningeal melanocytoma 268 N
the cerebellum 161 Meningeal melanocytosis 266, 267 N546K mutation 86, 149
Lipoma 240, 260 Meningeal melanoma 269 NAB2 249,250,253,254
Lipomatous glioneurocytoma 161 Meningeal melanomatosis 266, 267, 269 NAB2-STAT6 fusion 249, 250, 253, 254
Liposarcoma 260 Meningioangiomatosis 299 NADP+ 55
Lisch nodules 294, 295 Meningioma 232 Naevoid basal cell carcinoma syndrome 188,
LM02 275 Meningioma with oncocytic features 245 191, 192, 195, 197-199, 236, 319-321
Low-grade 8-cell lymphomas 276 Meningothelial meningioma 233, 237 NANOG 289
LRP18 50, 193 Merlin 117, 214, 217, 235, 299, 300 Napsin-A 341
LSAMP 50 Mesenchymal chondrosarcoma 253, 263 NCAM1 58, 103, 105, 122, 181, 194, 256,
Lupus erythematosus 275 MET 39, 51, 59, 146, 151, 229 341
Lymphomatoid granulomatosis 276 Metaplastic meningioma 233, 240 NCOA2 264
Lymphomatosis cerebri 272 Metastatic tumours of the CNS 338 NDRG2 235
Lymphoplasmacyte-rich meningioma 240 MGMT promoter methylation 21, 28, 41-44, Nerve growth factor 67
Lymphoplasmacytic lymphoma 276 68, 69, 74, 125 Nestin 38
Lynch syndrome 67, 317, 318 Microcystic meningioma 233, 239 Neurilemmomatosis 301
Lys656 mutation 151 Microtubule-associated protein tau 171 Neurilemoma 214
Lysozyme 280, 282, 283 Microvascular proliferation 17, 32, 36, 37, 46, Neurinoma 214
LZTR1 218,301-303 98, 114, 148, 157, 180 Neuroendocrine islet cell tumour 304
Minigemistocytes 23, 62, 64, 68, 72, 148, 149 Neurofibroma 219, 221, 294, 295, 298
M Mismatch repair cancer syndrome 188, 317, Neurofibromatosis type 1 42, 43, 59, 80, 89,
MAC387 282, 283 318 94, 97, 135, 141, 151, 167, 219, 222, 224,
MACF1 86, 140 MITF 266, 341 226, 274, 287,294-297,301,317
Maffucci syndrome 249 Mixed germ cell tumour 291 Neurofibromatosis type 2 109, 297, 301
Malignant ectomesenchymoma 262 Mixed pineocytoma-pineoblastoma 173, 177 Neurolipocytoma 161
Malignant fibrous histiocytoma See MKRN1 86 Neuromuscular choristoma 262
Undifferentiated pleomorphic sarcoma I MLH1 43, 317, 318 Neuron-specific enolase 256
malignant fibrous histiocytoma MLL 41, 59 Nf1-/- 295
Malignant perineurioma See MPNST with MLLT10 236 NF1 See Neurofibromatosis type 1
perineurial differentiation MMP2 31, 49 NF2 See Neurofibromatosis type 2
Malignant peripheral nerve sheath tumour MMP9 31, 49 NF-kappaB 112, 27 4
226, 228, 294 MN1 235 NFKB 1 A deletion 39
Malignant peripheral nerve sheath tumour Moesin 299, 308 NGN1 163
with divergent differentiation 227 Monomorphous angiocentric glioma 119 Nijmegen breakage syndrome 188
Malignant pineocytoma 173 Monosomy 22 42, 238 NOGO-A 65, 69, 134
Malignant triton tumour 227 Mosaicism 92, 296, 308 Non-polyposis colorectal carcinoma 42
MALT1 274 MPNST See Malignant peripheral nerve Non-WNT/non-SHH medulloblastoma 193
MALT lymphoma See Extranodal marginal sheath tumour Noonan syndrome 87-89, 151
zone lymphoma of mucosa-associated MPNST with perineurial differentiation NOTCH1 66, 72, 73
lymphoid tissue of the dura (malignant perineurioma) 228 NPM1 277

Subject index 405


NAAS 87, 151,266,270,281,295 Peripheral primitive neuroectodermal tumour Protein 4.1 299
NTRK 42,59,84,86,87 See Ewing sarcoma I peripheral primitive PSA 341
neuroectodermal tumour Psammomatous meningioma 233, 238
0 Peutz-Jeghers syndrome 141 Pseudopalisading 38
OCT4 287, 289-291, 326 PF-EPN-A 109, 110 Pseudopapillary ganglioglioneurocytoma 147
OLIG1 64 PF-EPN-8 109, 110 Pseudopapillary neurocytoma with glial
Oligoastrocytoma, dual-genotype 76 PGNT See Papillary glioneuronal tumour differentiation 147
Oligoastrocytoma, NOS 72, 69, 75-77 Phaeochromocytoma 167, 295, 304, 306 Pseudorosette 46, 89, 111, 122, 151, 166,
Oligodendroglial gliomatosis cerebri 61 PHLDB1 42 242
Oligodendroglioma, IDH-mutant and 1p/19q- Pl3K 28,39, 44,59, 73, 295,300,309,316 PTC1 191, 320
codeleted 18, 20, 53, 55, 60-70, 75. 76, Pl3K/AKT pathway 295, 300, 309, 316 PTCH 163,319,320
159 PIK3CA 39, 43, 59, 72. 73, 151, 281, 314, PTCH1 185, 190-192, 195, 197, 199, 236,
Oligodendroglioma lacking IDH mutation and 315 319-321
1 p/19q cod el et ion 68 PIK3R1 39. 43, 59, 72 PTCH2 197, 199, 319-321
Oligodendroglioma, NOS 60, 69 Pilocytic astrocytoma 16, 25, 40, 41, 60, 65, PTHR1 43
Oiiier-type multiple enchondromatosis 21, 68, 80-89, 93, 96, 111, 134, 139, 140, PTPN11 87
42, 67, 249 150, 151, 159, 186, 294,298,332,333, PTPRC 280
OMG 295 327 PTPRD 39
OPN4 172 Pilomyxoid astrocytoma 80, 81, 83, 88, 89 PTPRJ 120
Optic nerve glioma 80, 294 PIM1 273 PVT1-MYC fusion 185
OSBPL 10 273 Pineal anlage tumour 177
Osteochondroma 264 Pineal parenchymal tumour of intermediate Q
Osteolipoma 248, 260 differentiation 173 OKI 86, 120
Osteoma 264 Pineoblastoma 176, 178, 179
Osteosarcoma 264, 310 Pineoblastoma with lobules 173 R
OTX2 124, 193 Pineocytoma 170 R132C IDH1 mutation 54, 56, 312
Pineocytomatous rosettes 170-172 R132G IDH1 mutation 54
p Pineocytoma with anaplasia 173 R172K IDH2 mutation 55
p14ARF 40, 72,229 PIP2 316 R337H TP53 mutation 312
p53/MDM2/p14ARF pathway 40 PIP3 316 Rae 31,300
p65 112 Pituicytoma 332, 333, 336 Rac/PAK/JNK pathway 300
P450 286 PKD1 309 Radixin 299, 308
Paediatric diffuse astrocytoma 23 PLAP 287, 289-291 RAF1 84, 86, 87
Paediatric high-grade diffuse astrocytic Platelet-derived growth factors 67, 256 RasGAP 229
tumours 42 Pleomorphic xanthoastrocytoma 84, 87, 93, RASopathies 296
Paediatric-type oligodendroglioma See 94,96,98, 140, 155, 261 RB1 39, 40, 43, 59, 67, 172, 179, 287
Oligodendroglioma lacking IDH mutation Plexiform neurofibroma 220, 295 RBX1 305
and 1 p/19q codeletion Plexiform schwannoma 217 RCC See Clear cell renal cell carcinoma
Paget disease 249 PMS1 317 RELA fusion gene 109, 110, 112, 206
Papillary craniopharyngioma 324, 325, 327, PMS2 43, 67, 317, 318 RET 151, 167, 261
328 Polycythaemia 255 Retinoblastoma protein 40, 42-44, 59, 287,
Papillary cystadenoma 304 Polyomavirus 61 302,322
Papillary ependymoma 106, 108, 110, 111 Polyvesicular vitelline pattern 290 Retinoblastoma syndrome 67, 178, 179
Papillary glioneuronal tumour 147-149, 159, Posterior lens opacities 299 RFC 274
160 Posterior pituitary astrocytoma 332 RFX3 181
Papillary meningioma 233, 242, 243 POT1 67 RGS16 172
Papillary tumour of the pineal region 180, POU4F2 175, 179 Rhabdoid meningioma 233, 243, 244
181 PPM1D 59, 191 Rhabdoid tumour predisposition syndrome
Paraganglioma 164-167. 266 PPTID See Pineal parenchymal tumour of 129, 211,303,321
PAX2 256 intermediate differentiation Rhabdomyoma 262
PAX5 273 PRAME 175, 179 Rhabdomyosarcoma 248, 262, 295
PAX8 256, 341 PRC2 40,41,59,226,229 RhoA 31
PCNSL See Primary CNS lymphoma PRDM1 273' Rhodopsin 171, 178
PCP2 143 PRDM14 287 RHOH 273
PCP4 143 PRDX1 66 RNF130 86, 276
PCV chemotherapy 69, 73, 74 Primary CNS anaplastic large cell lymphoma, ROS1 261
PD1 44 ALK-negative 277 Rosai-Dorfman disease 282
PDGF 37, 44 Primary CNS anaplastic large cell lymphoma, Rosenthal fibres 51, 80-84, 86, 89, 116, 117,
PDGFA 67 ALK-positive 277 139, 148, 151, 154, 256,326, 327,333
PDGFB 67, 305 Primary CNS lymphomas 272 Rosette-forming glioneuronal tumour 150,
PDGFRA 21,39,42,51,56,59,67, 146,229 Primary glioblastoma, IDH-wildtype 28, 30, 151
PDGFRB 37, 67, 261 40, 41,53,54,56 Rosette-forming meningioma 245
POL 1 44 Primary leptomeningeal oligodendroglioma RPS6KB1 235
PEG3 66 61 RRAS 295
Perineurioma 220, 222-224, 228 Primary leptomeningeal oligodendro- RRAS2 295
Peripheral neuropathies 299 gliomatosis 152 RTEL 1 42
PRKCA 147, 149, 160 RTPS See Rhabdoid tumour predisposition
Progesterone receptor 253 syndrome

406 Subject index


Rubinstein-Taybi syndrome 188 SOX10 64, 216, 217,220, 221, 225, 228, TPH1 172
229, 253 TRAF7 235, 239
s SPDEF 181 Transforming growth factor beta 37, 44, 67
S6K1 309 SP-EPN 109, 110 Transitional meningioma 238
SALL4 289,290 Spindle cell oncocytoma 334-336 Triton tumour 226-229, 262, 294, 295
S-arrestin 171, 178 SP-MPE 109, 110 TRPM3 124
Schiller-Duval bodies 290 Spongioblastoma pattern 82 TSC1 91-93, 306-309
Schwannoma 109, 120, 164, 214-220, SPRED1 296 TSC2 91-93, 96,306-309
224-226, 228, 229, 262, 266, 297-299, SP-SE 109, 110 TTF1 117, 118,329-334,336, 339,341
301-303, 312 SRGAP3 86 TTR 129, 181
Schwannomatosis 214, 215, 217, 218, 221, SS18 228, 253 TTYH1 204
224, 225, 297,298,301-303, 321,322 STAT3 44, 45, 103 Tuberous sclerosis 90-93, 306-309
Schwannomin 217, 235, 299 STAT6 237,249-254 Turco! syndrome 42, 67, 109, 188, 317
Schwannosis 298 ST-EPN-RELA 109, 110 TWIST 49
Sclerosing epithelioid fibrosarcoma 261 ST-EPN-YAP1 109, 110
SDHA 167 Subependymal giant cell astrocytoma 90-93, u
SDHAF2 167 306-308 U373-MG 47
SOHB 143, 165-167, 316 Subependymoma 102, 103 UCHL 1 171
SOHC 167 Succinate dehydrogenase 167 UEA-1 49
SOHO 143, 167, 316 SUFU 185, 190. 192, 195, 197, 199, 236, Ultra-hypermutation phenotype 317, 318
Secondary glioblastoma, IOH-mutant 22, 40, 319-321 Undifferentiated pleomorphic sarcoma I
41,43,52-56, 76 SUS02 273 malignant fibrous histiocytoma 261
Secondary structures 30, 36, 64, 68, 72 SUZ12 229, 296 uPA 31
Secretory meningioma 233, 239, 240 SV40 61, 124,272 uPAR 31
SEGA See Subependymal giant cell SWI/SNF chromatin-remodelling complex 66, UTF1 289
astrocytoma 188, 194, 211, 302, 322
SET02 41,42,66, 73 Symmetrical axonal neuropathy 294 v
SF3B 1 mutation 270 Syncytiotrophoblastic cells 288 V600E-mutant BRAF protein 87, 134, 140,
SFRP1 191 Syncytiotrophoblastic giant cells 286, 290 182
SFT/HPC See Solitary fibrous tumour I VEGF 31,37,38,44, 67, 256,257,305
haemangiopericytoma T VEGFA 37
SHOC2 87 t(9;17)(q31;q24) 147, 149 VEGFR1 256
Sjogren syndrome 275 Talin 299 VEGFR2 37, 256
SLC9A1 66 Tanycytic ependymoma 103, 106, 108, 111, VHL See Von Hippel-Lindau disease
SLC44A1 147, 149, 160 112 Von Hippel-Lindau disease 167, 255, 304,
SLC44A1-PRKCA fusion 147, 149, 160 Tc2c.776C--,G mutation 275 305, 339
SMA 37, 145, 187, 210, 253, 262, 330 TCEB1 257, 305 Von Willebrand factor 49, 256
Small cell astrocy1oma 74 TCEB2 257, 305
Small cell glioblastoma 33, 72, 107 T-cell and NK/T-cell lymphomas 276 w
Small lymphocytic lymphoma 276 T-cell antigens 276 Werner syndrome 236
SMARCB 1-deficient anaplastic pleomorphic TCF12 61, 72, 73 Wilms tumour 188
xanthoastrocytoma 98 TENM3 50 Wiskott-Aldrich syndrome 275
SMARCE1 235, 236, 241 Teratoma 291 WNT/beta-catenin signalling 37, 300
SMO 185, 190-192, 195, 197, 199, 235, 319, Teratoma with malignant transformation 291 WNT signalling pathway 158, 188, 326, 327
320 TGFA 305 WT1 38
SNAl2 49 TIMP2 305 WWTR1-CAMTA1 fusion 258
SNCAIP 193 TLE1 253
SNF5 302, 322 TMEM127 167 y
SNP 67, 155,204,274 TNFRSF6 38 YAP1 109, 110, 191-193, 211, 259, 322
SNRPN 287 TNFRSF10B 38 Yolk sac tumour 290, 291
Solitary fibrous tumour I TNFRSF16 191, 197, 199
haemangiopericytoma 237, 249, 261 TNFSF6 38 z
SOS1 87 TNFSF10 38 ZFHX4 181
SOX2 91,92, 289,326 Toll-like receptor 274 ZMYM3 193
SOX9 263, 326 Touton cells 283 ZNF296 66
TP53-R72 variant 129

Subject index 407


List of abbreviations

AIDS Acquired immunodeficiency syndrome


ATP Adenosine triphosphate
cAMP Cyclic adenosine monophosphate
cDNA Complementary deoxyribonucleic acid
Cl Confidence interval
CNS Central nervous system
CT Computed tomography
DNA Deoxyribonucleic acid
EBV Epstein-Barr virus
EGFR Epidermal growth factor receptor
EMA Epithelial membrane antigen
FOG-PET 18F-fluorodeoxyglucose positron emission tomography
FISH Fluorescence in situ hybridization
G-CIMP Glioma CpG island methylator phenotype
GDP Guanosine diphosphate
GFAP Glial fibrillary acidic protein
GTP Guanosine-5'-triphosphate
H&E Haematoxylin and eosin
HAART Highly active antiretroviral therapy
HHV6 Human herpesvirus 6
HHV8 Human herpesvirus 8
HIV Human immunodeficiency virus
ICD-0 International Classification of Diseases for Oncology
LOH Loss of heterozygosity
MIM number Mendelian Inheritance in Man number
MR angiography Magnetic resonance angiography
MR spectroscopy Magnetic resonance spectroscopy
MRI Magnetic resonance imaging
mRNA Messenger ribonucleic acid
NFP Neurofilament protein
NOS Not otherwise specified
OR Odds ratio
PCR Polymerase chain reaction
PCV Procarbazine, lomustine, and vincristine
RNA Ribonucleic acid
RT-PCR Reverse transcriptase polymerase chain reaction
SEER Surveillance, Epidemiology, and End Results
SNP Sngle nucleotide polymorphism
SV40 Simian virus 40
TNM Tumour, node, metastasis

408 List of abbreviations

You might also like