You are on page 1of 490

©International Agency for Rase rch on Cane r 2021

All rights reserved

Suggested citation
WHO Classification of Tumours Ed1lorral Board. Central nervous system tumours.
Lyon (Franc ): International Agency for Research on Cancer, 2021.
(WHO classification of tumours serres, 5th ed., vol. 6)
https.//publications.iarc. fr/601.

Sales, rights, and permissions


Print copies are distributed by WHO Press, World Health Organization, 20 Avenue Appia, 1211 Geneva 27, Switzerland
Tel · +41 22 791 3264; Fax: +41 22 791 4857; emai l: bookorders@who.int; website: https://whobluebooks.rarc fr

To purchase IARC publications in electronic format, see the IARC Publications website (https://publications.iarc.fr).

Requests for permission to reproduce or translate !ARC publications - whether for sale or for non-commercial distribution - should be submi ed
through the IARC Publications website (https://publicatlons.iarc.fr/Rights-And-Permissions).

Third-party materials
If you wish to reuse material from thrs work that Is attributed to a thrrd party, such as figures , tables, or boxes, rt rs your responsibil ity to deterrnne
whether permission rs needed for that reuse and to obtain permission from the copyright holder. See Sources, pages 493-499. The nsk of claim 3
resulting from infringement of any third-party-owned component in the work rests solely with the user.

General disclaimers
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 WHO or contributing agencies concerning the legal status of any country, territory, crty, or area, or of its authorities , or concerning the
del1m1tation of its frontiers or boundaries. Dotted and dashed lines on maps represent approximate border lines for which there may not yet be full
agreement.

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

All reasonable precautions have been taken by WHO to verify the information contained in this publication. However, the published material is
being distributed without warranty of any kind, either expressed or implied. The responsibility for the interpretation and use of the material hes w1
the reader. In no event shall WHO or contributing agencies be liable for damages arising from its use.

First print run (7500 copies)

Updated corrigenda can be found at https://publicatlons.iarc.fr

IARC Library Cataloguing-in-Publication Data


Names: WHO Classification of Tumours Editorial Board .
Title: Central nervous system tumours I edited by WHO Classification of Tumours Editorial Board.
Description: Fifth edition. I Lyon : International Agency for Research on Cancer, 2021 I Series: World Health Organization classification o tum ur ·
I Includes bibliographical references and index.
Identifiers: JSBN 9789283245087 (pbk.) j ISBN 9789283245094 (ebook)
Sub1ects· MESH Central Nervous System Neoplasms.
Class1flcatlon. NLM WL 15
This volume was produced in collaboration with

The Consortium to Inform Molecular and Practical Approaches to


CNS Tumor Taxonomy (clMPACT)

The WHO classification of central nervous system tumours presented in this book reflects
the views of the WHO Classification of Tumours Editorial Board that convened
via video conference 24-26 August 2020.
The WHO Classification of Tumours
Editorial Board

Expert members: Central nervous system tumours


Brat, Daniel J. Pfister, Stefan M.
Northwestern University Feinberg School of Medicine Hopp Children's Cancer Center Heidelberg (K1TZ) German Cancer
Chicago Research Center (DKFZ), and Heidelberg University Hospital
Heidelberg
Ellison, David W.
St. Jude Children's Research Hospital Reifenberger, Guido
Memphis Heinrich Heine University DOsseldorf
Dusseldorf
Figarella-Branger, Dominique
Assistance Publique des H6pitaux de Marseille Soffletti, Riccardo
Marseille University of Turin and City of Health and Science Hospital, Turin
Turin
Hawkins, Cynthia E.
Hospital for Sick Children von De1mling, Andreas
Toronto Heidelberg University and German Cancer Research Center (DKFZ)
Heidelberg
Louis , David N.
Massachusetts General Hospital and Harvard Medical School Wessel1ng, Pieter
Boston Princess Maxima Center for Pediatric Oncology, Utrecht, and
Amsterdam University Medical CentersNUmc, Amsterdam
Ng, Ho-Keung
Prince of Wales Hospital, Shat1n
Hong Kong SAR

Perry, Arie
University of California, San Francisco
San Francisco

For the complete list of all contributors and their affil1at1ons, see pages 483-488.
The WHO Classification of Tumours
Editorial Board (continued)

Standing members
came1ro. Fatima Oliva, Esther
1patimup/13S Massachusetts General Hospital
Porto Boston

Chan. John K.C Rous, Brian


Queen Elizabeth Hospital Public Health England
Kowloon, Hong Kong SAR Fulbourn, Cambridge

Cheung, Annie Nga-Yin Singh, Rajendra


University of Hong Kong Northwell Health
Hong Kong SAR Lake Success

Cree, Ian A. (Editorial Board Chair) Soares, Fernando Augusto


International Agency for Research on Cancer Rede D'Or Hospitals
Lyon Sao Paulo

Field, Andrew S. Srigley, John R.


St Vincent's Hospital Sydney Trillium Health Partners
Darlinghurst Mississauga

Gill, Anthony J. Tan, Puay Hoon


Royal North Shore Hospital Singapore General Hospital
St Leonards Singapore

Lakhani, Sunil R. Thompson, Lester D.R.


University of Queensland and Pathology Queensland Head and Neck Pathology Consultations
Royal Brisbane and Women's Hospital Woodland Hills
Herston
Tsao, Ming Sound
Lax, Sigurd F. University Health Network
General Hospital Graz II, Medical University of Graz Toronto
Graz
Tsuzuki, Toyonori
Lazar, Alexander J. Aichi Medical University Hospital
University of Texas MD Anderson Cancer Center Nagakute
Houston
Washington, Mary K.
Moch, Helger Vanderbilt University Medical Center
University of Zurich and University Hospital Zurich Nashville
Zurich

Ochiai, Atsushi
National Cancer Center
Kashiwa

For the complete list of all contributors and their affiliations, see pages 483-488.
WHO Classification of Tumours
C ntr I N rv us System Tumours

1/\1 H' l d1lu1s la11A Croo


Di lnn1 Lol<u lio!ty
Lnurn A. N. Pe fmoen
VAierie I\ Wl1ito

I p1c1 m1nlogy Ari ana Znaor

Asiedua Asan te

Assis t nts Ann e-Sophie Hameau


Laura Brispot

Technica l Editing Jessica Cox


Julia Slone-Murphy

Principal Information Assistant Alberto Machado


Inform ation Assistant Catarina Marques

Layout Meaghan Fortune

Radiology Advisor Sona A. Pungavkar, Global Hospitals, Mumbai, India

Printed by Maestro Gestlon D'Edition


69100 Vi!leurbanne, France
Printed in Italy

Publisher International Agency for Research on Cancer (!ARC)


150 Cours Albert Thomas
69372 Lyon Cedex 08, France
Contents

List of abbreviations xi Ependymal tumours


Introdu ction l') (j
Foreword xii Supratentorial ependymoma 6
Supratentorial ependymoma, ZFTA fu s1on- pos1t1ve 164
ICD-0 topographical coding Supratentorial ependymoma, YAP I fusion - pos1t1ve 167
Poste rior fossa ependymoma 169
ICD-0 morphological coding 1 Posterior fossa group A (PFA) ependymoma 172
CNS tumours 2 Posterior fossa group B (PFB) ependymoma 175
Spinal ependymoma 177
Introduction to CNS tumours 7 Spinal ependymoma, M YCN-am plified 180
Myxopapillary ependymoma 183
2 Gliomas, glioneuronal tumours, and neuronal tumours 15 Subependymoma 186
Introducti on 16
Adult-type diffuse gliomas 3 Choroid plexus tumours 189
Astrocytoma, !DH-mutant 19 Choroid plexus papilloma 190
Ol igodendroglioma , IDH-mutant and 1p/19q- Atypical choroid plexus papilloma 193
codeleted 28 Choroid plexus carcinoma 195
Glioblastoma, IDH-wildtype 39
Paed iatric-type diffuse low-grade gliomas 4 Embryonal tumours 199
Diffuse astrocytoma, MYB- or MYBL 1-altered 56 Medulloblastoma
Angiocentric glioma 59 Introduction 200
Polymorphous low-grade neuroepithelial tumour of Medulloblastomas, molecularly defined
the young 62 Medulloblastoma , WNT-activated 203
Diffuse low-grade glioma, MAPK pathway- altered 65 Medulloblastoma , SHH-activated and TP53-wildtype 205
Paediatric-type diffuse high-grade gliomas Medulloblastoma , SHH-activated and TP53-mutant 208
Diffuse midline glioma, H3 K27-altered 69 Medulloblastoma, non-WNT/non-SHH 211
Diffuse hemispheric glioma, H3 G34-mutant 74 Medulloblastomas, histologically defined
Diffuse paediatric-type high-grade glioma, Medulloblastoma, histologically defined 213
H3-wildtype and IDH-wildtype 77 Other CNS embryonal tumours
Infant-type hemispheric glioma 81 Introduction 220
Circumscribed astrocytic gliomas Atypical teratoid/rhabdoid tumour 221
Pilocytic astrocytoma 83 Crlbriform neuroepithelial tumour 226
High-grade astrocytoma with piloid features 90 Embryonal tumour with multilayered rosettes 228
Pleomorph ic xanthoastrocytoma 94 CNS neuroblastoma, FOXR2-activated 232
Subependymal giant cell astrocytoma 100 CNS tumour with BCOR internal tandem duplication 235
Chordoid glioma 104 CNS embryonal tumour NEC/NOS 238
Astroblastoma , MN1-altered 107
Gl ioneuronal and neuronal tumours 5 Pineal tumours 241
Ganglioglioma 111 Introduction 242
Gangliocytoma 116 Pineocytoma 243
Desmoplastic infantile ganglioglioma I Pineal parenchymal tumour of intermediate
desmoplastic infantile astrocytoma 119 differentiation 246
Dysembryoplastic neuroepithelial tumour 123 Pineoblastoma 249
Diffuse glioneuronal tumour with Papillary tumour of the pineal region 253
oligodendroglioma-like features and nuclear Desmoplastic myxoid tumour of the pineal reg ion .
clusters 127 SMARCB1 -mutant 256
Papillary glioneuronal tumour 130
Rosette-forming glioneuronal tumour 133 6 Cranial and paraspinal nerve tumours 259
Myxoid glioneuronal tumour 136 introduction 260
Diffuse leptomeningeal glioneuronal tumour 139 Schwannoma 261
Mult1nodular and vacuolating neuronal tumour 143 Neurofibroma 265
Dysplastic cerebellar gangliocytoma (Lhermitte- Perineurioma 269
Duclos d isease) 146 Hybrid nerve sheath tumours 271
Central neurocytoma 149 Malignant melanotic nerve sheath tumour _73
Extraventri cular neurocytoma 153 Malignant peripheral nerve sheath tumour 275
Cerebellar liponeurocytoma 156 Cauda equina neuroendocrine tumou r (previously
paraganglioma) 279
7 Meningioma 283 11 Germ cell tumours 381
8 Mesenchymal, non-meningothelial tumours involving the 12 Tumours of the sellar region 391
CNS Introduction 392
299
Introduction 300 Adamant1nomatous craniopharyng1oma 393
Soft tissue tumours Papillary craniopharyng1oma 397
Fibroblastic and myof1broblastic tumours Pituicytoma, granular cell tumour of the sellar
Solitary fibrous tumour 301 region , and spindle cell oncocytoma 401
Vascular tumours Pituitary adenoma I pituitary neuroendocnne tumour 406
Haemangiomas and vascular malformations 306 Pituitary blastoma 415
Haemangioblastoma 310
Skeletal muscle tumours 13 Metastases to the CNS 417
Rhabdomyosarcoma 314 Metastases to the brain and spinal cord parenchyma 418
Tumours of uncertain differentiation Metastases to the meninges 421
lntracranial mesenchymal tumour, FET::CREB
fusion-positive 317 14 Genetic tumour syndromes involving the CNS 423
CIC-rearranged sarcoma 320 Introduction 424
Primary intracranial sarcoma, O/CER1-mutant 323 Neurofibromatosis type 1 426
Ewing sarcoma 326 Neurofibromatosis type 2 429
Chandra-osseous tumours Schwannomatosis 434
Chondrogenic tumours Von Hippel-Lindau syndrome 437
Mesenchymal chondrosarcoma 330 Tuberous sclerosis 441
Chondrosarcoma 332 Ll-Fraumeni syndrome 446
Notochordal tumours Cowden syndrome 449
Chordoma 335 Constitutional mismatch repair deficiency syndrome 452
Famil ial adenomatous polyposis 1 456
9 Melanocytic tumours 339 Naevoid basal cell carcinoma syndrome 458
Introduction 340 Rhabdoid tumour predisposition syndrome 460
Diffuse meningeal melanocytic neoplasms Carney complex 462
Melanocytosis and melanomatosis 341 DICER1 syndrome 464
Circumscribed meningeal melanocytic neoplasms Familial paraganglioma syndromes 467
Melanocytoma and melanoma 344 Melanoma-astrocytoma syndrome 471
Familial retinoblastoma 473
10 Haematolymphoid tumours involving the CNS 349 BAP1 tumour predisposition syndrome 475
Introduction 350 Fanconi anaemia 478
Lymphomas ELP1-medulloblastoma syndrome 481
CNS lymphomas
Primary diffuse large 8-cell lymphoma of the CNS 351 Contributors 483
Immunodeficiency-associated CNS lymphomas 356
Lymphomatoid granulomatosis 358 Declaration of interests 489
lntravascular large B-cell lymphoma 360
Miscellaneous rare lymphomas in the CNS IARC/WHO Committee for ICD-0 491
MALT lymphoma of the dura 362
Other low-grade 8-cell lymphomas of the CNS 364 Sources 493
Anaplastic large cell lymphoma (ALK+/ALK-) 366
T-cell and NK/T-cell lymphomas 368 References 501
Histiocytic tumours
Erdheim-Chester disease 370 Subject index 557
Rosa1- Dorfman disease 372
Juvenile xanthogranuloma 374 Previous volumes in the series 568
Langerhans cell histiocytosis 376
Histlocytic sarcoma 379
List of abbreviations

AIDS acquired 1mmunodef ic1ency syndrome ITD internal tandem duplication


AMT PET a-[ 11 C]methyl -L- tryptophan positron emission M:F ratio male-to-female ratio
tomography MALT mucosa-associated lymphoid tissue
ATP adenos1ne lriphosphate MALT lymphoma extranodal marginal zone lymphoma of mucosa-
bp base pair(s) associated lymphoid tissue
cAMP cyc lic adenosine monophosphate MIM number Mendelian Inheritance 1n Man number
CBTRUS Central Brain Tumor Registry of the United States MITF melanogenes1s-assoc1ated transcription factor
clMPACT Consortium to Inform Molecular and Practical MRI magnetic resonance imaging
Approaches to CNS Tumor Taxonomy mRNA messenger ribonucleic acid
CMV cytomegalovirus N:C ratio nuclear-to-cytoplasmic ratio
CNS central nervous system NCCN National Comprehensive Cancer Network
COG Children 's Oncology Group NEC not elsewhere classified
CSF cerebrospinal fluid NOS not otherwise specified
CT computed tomography NSE neuron-specific enolase
DNA deoxyribonucleic acid OS overall survival
EBV Epstein-Barr vi ru s PAS periodic acid-Schiff
ER estrogen receptor PCR polymerase chain reaction
EU-AHAB European Rh abdoi d Reg istry PCV procarbazine , lomustine. and v1ncristine
FDG PET 18F-fluorodeoxyglucose positron emission PET posi tron emission tomog raphy
tomography PET-CT pos itron em ission tomography-computed
FET PET 18F-f luoroethyltyrosine positron emission tomography
tomography PFA posterior fossa group A
FISH fluorescence in situ hybridization PFB posterior fossa group B
FLAIR fluid-attenuated inversion recovery PFS progression-free su rvival
FNA fine-needle aspiration PR progesterone receptor
G-CIMP glioma CpG island methylator phenotype RNA ribonucle ic aci d
H&E haematoxylin and eosin RT-PCR reverse transcriptase polymerase chain reac tion
HBV hepatitis B virus SEER Program Surveillance, Epidem iology, and End Results
HGVS Human Genome Variation Society Program
HHV6 human herpesvirus 6 SNP single-nucleotide polymorph ism
HIV human immunodeficiency virus SSTR PET somatostatin receptor pos itron em ission
HPF high-power field(s) tomography
HPV human papillomavirus TAM tumour-associated macrophage
HTLV human T-lymphotropic virus TKO tyrosine kinase domain
IARC International Agency for Research on Cancer TNM tumour, node, metastasis
ICD-11 International Classification of Diseases, 11th Treg cell T regulatory cell
revision tRNA transfer ribonucleic ac id
ICD-0 International Classification of Diseases for t-SNE I-distributed stochastic neighbour embedding
Oncology VATERNACTERL vertebral defects, anal atresia, cardiac
ICD-0-3 International Classification of Diseases for association malformations, tracheo-oesophageal fistula with
Oncology , 3rd edition oesophageal atresia , rad ial or renal dysplasia, and
lg immunoglobulln limb abnormal ities

Li st of abbreviations xi
Foreword
The WHO Classification of Tumours, published as a series of books (also known as the WHO Blue Books) and now as a website
(https://tumourclassification.iarc.who.int), is an essential tool for standardizing diagnostic practice worldwide. It also serves as a
vehicle for the translation of cancer research into practice. The diagnostic criteria and standards that make up the classif1cat1on
are underpinned by evidence evaluated and debated by experts in the field . About 200 authors and editors participate 1n the
production of each book, and they give their time freely to this task . I am very grateful for their help; it is a remarkable team effort
This sixth volume of the fifth edition of the WHO Blue Books has , like the preceding five , been led by the WHO Class1ficat1on of
Tumours Ed itorial Board, composed of standing and expert members . The standing members , who have been nominated by
pathology organizations, are the equivalent of the series editors of previous editions. The expert members for each volume, equiv-
alent to the volume editors of previous editions , are selected on the basis of informed bibliometric analysis and advice from the
standing members . The diagnostic process is increasingly multidisciplinary, and we are delighted that several radiology and clinical
experts have joined us to address specific needs.
The most conspicuous change to the format of the books in the fifth edition is that tumour types common to multiple systems
are dealt with together - so there are separate chapters on mesenchymal (non-meningothelial) tumours. melanocytic tumours ,
haematolymphoid tumours, and germ cell tumours. There is also a chapter on genetic tumour syndromes. Genetic disorders are
of increasing importance to diagnosis in individual patients, and the study of these disorders has undoubtedly informed our under-
standing of tumour biology and behaviour over the past decade.
We have attempted to take a more systematic approach to the multifaceted nature of tumour classification ; each tumour type
is described on the basis of its localization , clinical features , epidemiology, etiology, pathogenesis , histopathology, diagnostic
molecular pathology, staging, and prognosis and prediction . We have also included information on macroscopic appearance and
cytology, as well as essential and desirable diagnostic criteria. This standardized , modular approach makes it easier for the books
to be accessible online, but it also enables us to call attention to areas in which there is little information , and where serious gaps 1n
our knowledge remain to be addressed .
The organ ization of the WHO Blue Books content now follows the normal progression from benign to malignant - a break with the
fourth edition , but one we hope will be welcome.
The volumes are still organized by anatomical site (digestive system , breast, soft tissue and bone, etc.), and each tumour type is
listed within a hierarchical taxonomic classification that follows the format below, which helps to structure the books in a systematic
manner:

Site: e.g. CNS embryonal tumours


Category: e.g. medulloblastoma
Family (class): e.g. molecularly defined medulloblastomas
Type: e.g. SHH-activated and TP53-wildtype medulloblastoma
Subtype: e.g. provisional molecular subgroup SHH-1

The issue of whether a given tumour type represents a distinct entity rather than a subtype continues to exercise pathologists, and
it is the topic of many publications in the literature. We continue to deal with this issue on a case-by-case basis, but we believe there
are inherent rules that can be applied . For example, tumours in which multiple histological patterns contain shared truncal mutations
are clearly of the same type , despite the differences in their appearance. Equally, genetic heterogeneity within the same tumour
type may have implications for treatment. A small shift in terminology in the fifth edition is that the term "variant" in reference to a
specific kind of tumour has been wholly superseded by "subtype", in an effort to more clearly differentiate this meaning from that of
"variant" in reference to a genetic alteration .
Another important change in this edition of the WHO Classification of Tumours series is the conversion of mitotic count from the
traditional denominator of 10 HPF to a defined area expressed in mm 2 . This serves to standardize the true area over which mitoses
are enumerated, because different microscopes have high-power fields of different sizes . This change will also be helpful for any-
one reporting using digital systems .

We are continually working to improve the consistency and standards within the classification . In addition to having moved to the
International System of Units (SI) for all mitotic counts, we have standardized genomic nomenclature by using Human Genome
Variation Society (HGVS) notation . For CNS tumours, mutation of histone genes is of particular importance, and we have Incor-
porated the latest nomenclature to ensure that histone sequence variants have an unambiguous description in the classification
We have also further standardized our use of units of length, adopting the convention used by the International Collaboration on

xii Foreword
Cd•ll l I Ht'l ' 1 irl111q (111tp //ww·.:v f( u (;)I 1rH orq) w rj the lJK Royal Co.!eg8 of PrJ hol0g1sts (hit JS //N'N'N rq:iath !Jrg/) ~;rJ tl'"•::jt th~ SIZ8
l 'f tt111 1 ll'" 1<.. tk'W q1\ ~11 n ' l lu 1vnly 1n m1lltmP.tres (rPm) rattler tt ar ce~t1 metr~s (cm) fh1" 15 dearer. 1n ()r ir 118w AnrJ ::i 10 d> thA Y.;8
l'' dt'1 11nr1I r'P1nts a ommon sou 1c.e of mPd1c::ii errors .
l he \'VHL l~itH? Books are much appres1ated by pathologists and 0 increasing irPpor ancE' to pract1troners of other r,lrn•r:.81 d•-;
L1~111n1?s involved in cnncer management. as well as to researchers The ed tonal board and I certainly hope that the s~riP.s N1ll
\ t•nt111uc 10 meet the need tor standards 1n d1agno~is and to fac1 1tate the trarisla· on of d1agnost1c research into practice Norldw 1 d~
11 is part1l ularly 1111portant that cancers continue to be class•'ied ard d iagnosed according to he same standards 1nternat1onall; so
that pcit1ents can benefit from mullicentre clinical trials, as Nell as from the results 0 local trials conduced on different cont1nen s

< I /}

~~-
Or Ian A Cree
--
Head WHO Classification of Tumou rs Programme
International Agency fo r Researc h on Cancer
November 2021

For e~ o rd ' '''


ICD-0 topographical coding of central nervous system
tumours

The IC0 -0 topography cod · s ror the main anatomica l sites covered In thi s volume are as foll ows (9901 :

C70 Meninges
C70.0 Cere bral mening es
C70 1 Spinal meninges
C70 9 Meninges. NOS

C71 Brain
C71 .0 Cerebrum
C71 1 Frontal lobe
C71 .2 Temporal lobe
C71 .3 Parietal lobe
C71.4 Occipital lobe
C71 .5 Ventricle, NOS
C71 .6 Cerebellum, NOS
C71.7 Brain stem
C71 .8 Overlap ping lesion of brain
C71.9 Brain , NOS

C72 Spinal cord, cranial nerves, and other parts of central nervous system
C72 .0 Spinal cord
C72.1 Cauda equina
C72.2 Olfactory nerve
C72 .3 Optic nerve
C72.4 Acoustic nerve
C72 .5 Cranial nerve, NOS
C72.8 Overlapping lesion of brain and central nervous system
C72 .9 Nervous system , NOS

C75 Other endocrine glands and related structures


C75 .1 Pituitary gland
C75 .2 Craniopharyngeal duct
C75 .3 Pineal gland

ICD-0 morphological coding: Introduction


The ICD-0 coding system uses a topography (T) code and a morphology (M) code together, but these are presented in separate
lists for ease of use. Behaviour is coded /0 for benign tumours; /1 for unspecified, borderline, or uncertain behaviour; /2 for carci-
noma in situ and grade Ill intraepithelial neoplasia; /3 for malignant tumours, primary site; and /6 for malignant tumours, metastatic
site. Behaviour code /6 is not generally used by cancer registries . For various reasons , the ICD-0 morphology terms may not
always be identical to the entity names used in the WHO classification , but they should be sufficiently similar to avoid confusion .
The designation "NOS" ("not otherwise specified ") is provided to make coding possible when subtypes exist but exact classification
may not be possible in small biopsies or certain other scenarios . Therefore, it is usual to have "NOS" even when a more specific
alternative term is listed in ICD-0.

ICD-0 coding of central nervous system tumours


ICD-0 coding of central nervous system tumours
ICD-0-3 .2 ICD -0 label (subtypes are indicated 1n grey text, with the label indented);
Please note that the WHO classification of tumour types is more readily reflected in the table of contents

Gllomas, glioneuronal tumours, and neuronal tumours


Adult-type diffuse gliomas
Astrocytoma, !DH-mutant
9400/3 Astrocytoma, !DH-mutant, grade 2
9401,'3 Astrocytoma. !DH-mutant. grade 3
9445/3 Aslrocytoma IDH-mutant, grade 4
Oligodendroglioma, !DH-mutant and 1p/19q-codeleted
9450/3 Oiigodendrogl1oma, !DH-mutant and 1p/19q-codeleted, grade 2
9451 '3 Ol1godendrogltoma, JOH -mutant and 1p/19q-codeleted, grade 3
9440/3 Glioblastoma, IDH-wildtype

Paediatric-type diffuse low-grade gliomas


9421/1 Diffuse astrocytoma , MYB- or MYBL1-alteredt
9431/1 Angiocentric glioma
9413/0 Polymorphous low-grade neuroepithelial tumour of the youngt
9421/1 Diffuse low-grade glioma, MAPK pathway-alteredt

Paediatric-type diffuse high-grade gliomas


9385/3 Diffuse midline glioma, 1-13 K27-alteredt
9385/3 Diffuse hemispheric glioma, H3 G34-mutantt
9385/3 Diffuse paediatric-type high-grade glioma, H3-wildtype and IDH-wildtypet
9385/3 Infant-type hemispheric gliomat

Circumscribed astrocytic gliomas


9421/1 Pilocytic astrocytoma
9421/3* High-grade astrocytoma with piloid features
9424/3 Pleomorphic xanthoastrocytoma
9384/1 Subependymal giant cell astrocytoma
9444/1 Chordoid glioma
9430/3 Astroblastoma, MN1-alteredt

Glioneuronal and neuronal tumours


9505/1 Ganglioglioma
9492/0 Gangliocytoma
9412/1 Desmoplastic infantile ganglioglioma
9412/1 Desmoplastic infantile astrocytoma
9413/0 Dysembryoplastic neuroepithelial tumour
n/a Diffuse glioneuronal tumour with oligodendroglioma-like features and nuclear clusters (provisional entity)
9509/1 Papillary glloneuronal tumour
9509/1 Rosette-forming glioneuronal tumour
9509/1 Myxoid glioneuronal tumourt
9509/3 ~ Diffuse leptomeningeal glioneuronaJ tumour
9509/0* Multinodular and vacuolating neuronal tumour
9493/0 Dysplastic cerebellar gangliocytoma (Lhermitte-Duclos disease)
9506/1 Central neurocytoma
9506/1 Extraventricular neurocytoma
9506/1 Cerebellar llponeurocytoma

Ependymal tumours
9391/3 Supratentorial ependymoma, NOSt
9396/3 Supratentorial ependymoma, ZFTA fusion-posltive 1
9396/3 Supratentorial ependymoma, YAP1 fusion-positivet
9391/3 Posterior fossa ependymoma, NOS 1
9396/3 Posterior fossa group A (PFA) ependymomat
9396/3 Posterior fossa group B (PFB) ependymomat
9391/3 Spinal ependymoma, NOS 1
9396/3 Spinal ependymoma, MYCN-ampl1fied t
9394/1 Myxopapillary ependymoma
9383/1 Subependymoma

~ ICD-0 coding of central nervous system tumours


ICD-0-3 2 ICD -0 label (subtypes a1e ind icated 1n grey text, with the lab8l 1ndented);
Please note tt1at tho WHO c lassif1cat1 on of tumour types is more readi ly reflected 1n the table of contents

Choroid plexus tumours


9390/0 Ct1oro1d plexus papilloma
9390/1 Atypical choroid plexus pa pil loma
9390/3 Choroid plexus carc inoma

Embryonal tumours
Medulloblastomas. molecularly defined
9475/3 Medulloblastoma , WNT-activated
9471/3 Medulloblastoma, SHH-activated and TP53-wildtype
9476/3 Med ulloblastoma, SHH-activated and TP53-mutan t
9477/3 Medu lloblastoma, non-WNT/non-S HH

Medulloblastomas, histologically defined


9470/3 Medul lob lastoma, histological ly defined
9..171 3 Desrnoplast1c nodular medulloblastoma
9.::- ~ '3 Medulloblastoma with extensive nodularity
9~7..1 / 3 Large ce!I medulloblastoma
9!..7.1/3 Anaptast1c medullobtastoma

Other CNS embryonal tumours


9508/3 Atypical teratoid/rhabdoid tumour
n/a Cribritorm neuroepithel ial tumour (provisional entity)
9478/3 Embryonal tumour with multilayered rosettes
9500/3 CNS neuroblastoma, FOXR2-activatedt
9500/3 CN S tum our with BCOR internal tandem duplicationt
9473/3 CNS embryonal tumour, NEC/NOS

Pineal tumours
9361 / 1 Pineocytoma
9362/3 Pineal parenchymal tumour of intermediate differentiation
9362/3 Pineoblastoma
9395/3 Papillary tumour of the pineal region
n/a Desmop lastic myxoid tumour of the pineal region, SMARCB 1-mutant (provisional entity)

Cranial and paraspinal nerve tumours


9560/0 Schwannoma
9540/0 Neurofi broma
9550/0 Plex1form neurof1broma
9571 /0 Perineu rioma
9563/0 Hybrid nerve sheath tumour
9540/3 Malignant melanotic nerve sheath tumour
9540/3 Malignant peripheral nerve sheath tumour
8693/3 Cauda equ ina neuroendocrine tumour (previously paraganglioma)

Meningioma
9530/0 Meni ng ioma

ICD-0 codin g of cen tral nervous system tumours 3


ICD -0-3 2 ICD 0 lnbel (sut>typ0s dro indir .ilf~cl in qrny tu;<t, with the label 1rnJm1ted),
Pleast> 11ote thnt the WHO d1s' ;1f1cdt1on of lurnr1111 typos 1•., more madily reflected in the table of contents

Mesenchymal, non-meningothellal tumours involving the CNS


F1broblastic and myollbroblastic tumours
8815/1 Solitary fibrous tumour

Vascular tumours
9121/0 Cavernous haemangioma
9131/0 Capillary haemang1oma
9123/0 Arteriovenous malformation
9161/1 Haemang ioblastoma

Skeletal muscle tumours


8910/3 Embryonal rhabdomyosarcoma
8920/3 Alveolar rhabdomyosa rcoma
8901 /3 Rhabdomyosarcoma, pleomorphic-type
8912/3 Spindle cell rhabdomyosarcoma

Tumours of uncertain differentiation


n/a lnlracranial mesenchymal tumour, FET::CREB fusion-positive (provisional entity)
9367/3 CIC-rearranged sarcoma
9480/3 Primary intracranial sarcoma, DICER1-mutantt
9364/3 Ewing sarcoma

Chondrogenic tumours
9240/3 Mesenchymal chondrosarcoma
9220/3 Chondrosarcoma
9243/3 Dedifferentiated chondrosarcoma

Notochordal tumours
9370/3 Chordoma

Melanocytic tumours
Diffuse meningeal melanocytic neoplasms
8728/0 Meningeal melanocytosis
8728/3 Meningeal melanomatosis

Circumscribed meningeal melanocytic neoplasms


8728/1 Meningeal melanocytoma
8720/3 Meningeal melanoma

Haematolymphoid tumours involving the CNS


CNS lymphomas
9680/3 Primary diffuse large 8-cell lymphoma of the CNS
9766/1 Lymphomatoid granulomatosis
9766/1 Lymphomatoid granulomatosis, grade 1
9766/1 Lymphomatoid granulomatosis, grade 2
9766/3 Lymphomatoid granulomatosis, grade 3
9712/3 lntravascular large 8-cell lymphoma

Miscellaneous rare lymphomas in the CNS


9699/3 MALT lymphoma of the dura
9671/3 Lymphoplasmacytic lymphoma
9690/3 Follicular lymphoma
9714/3 Anaplastic large cell lymphoma (ALK+/ALK-)
9702/3 T-cell lymphoma
9719/3 NK/T-cell lymphoma

Hisdocytic tumours
9749/3 Erdheim- Chester disease
9749/3 Rosai - Dorfman disease1
9749/1 Juvenile xanthogranulomat
9751/1 Langerhans cell histiocytosis
9755/3 Histiocytic sarcoma

4 ICD -0 coding of central nervous system tumours


ICD-0-3.'2 ICD 0 label (subtypes are Indicated in grey text, with the label indented),
Please note that lhe WHO class1f1cation of tumou r types 1s more readily reflected in the table of contents

Germ cell tumours


9080/0 Mature teratoma
9080/3 Immature teratoma
9084/3 Teratoma with somatic-type malignancy
9064/3 Germ1noma
9070/3 Embryonal carcinoma
9071/3 Yolk sac tumour
9100/3 Chonocarc1noma
9085/3 Mixed germ cell tumour

Tumours of the sellar region


9351/1 Adamantinomatous c ranlopharyngioma
9352/1 Papillary craniopharyng ioma
9432/1 P1tu1cytoma
9582/0 Granul ar cell tumour of the sellar reg ion
8290/0 Spindle cell oncocytoma
8272/3 Pituitary adenoma I p ituitary neuroendocrine tumour (PitNET)l
8273/3 Pituitary blastoma

These morphology codes are from the International Classification of Diseases for Oncology , third edition, second revision (ICD-0-3.2)
114141 . Behaviour is coded /0 for benign tumours ; /1 for unspecified , borderline, or uncertain behaviour; /2 for carcinoma in situ and grade
Ill intraepithelial neoplasia; /3 for malignant tumours, primary site; and /6 for malignant tumours , metastatic site. Behaviour code /6 is not
generally used by cancer registries.
This classification is modified from the previous WHO classification, taking into account changes in our understanding of these lesions.
n/a, not available (provisional entity) .
· Codes marked with an asterisk were approved by the !ARC/WHO Committee for ICD-0 at its meeting in May 2021.
1 Labels marked with a dagger have undergone a change in terminology of a previous code.

ICD-0 coding of central nervous system tumours 5


-15% 8-

~a~ ~ aaa ~ aaa ~ ~

c c LCA, eta IC Clas c > LCA eta c >L c

'
-75% -55% -45% -85%

14+
7+
-60% -8~0 -85°

7+
I 7q
-7 0
Introduction to CNS tu111ours LOU IS ON
Ellison OW
Perry A
Wesselin g P

Th e fifth -edition Centra l nervous system tumours volu me of the In this fifth -edition volume, for consistency with the other fifth-
WHO Classification of Tumours series (also known as the WH O edition WHO Blue Books, the term "type" is used instead of "entity··.
Blue Books) is based on the revised fourth-edition vo lume pub- and "subtype" is used instead of "variant ". Only types are listed in
lished in 2016 , on the many developments in the fie ld that fo l- the classification, with the subtypes listed in the Subtype(s) sub-
lowed the 2016 classification, and on the recommendations of sections and then described in the Histopathology or Diagnostic
the Consortium to Inform Molecular and Practical Approaches molecular pathology subsections of individual sections. As a result
to CNS Tumor Taxonomy (c lMPACT) 11933 ,1932,1935, 838 , of this change and because grading 1s being applied within types
356 ,1946,1934 ,1944,357,836). The fifth-edition c lassi fi cation (see below), meningioma is treated as a single type with only one
is presented in the tab le of contents of th is volume . The fifth entry in the classification, with its many histological subtypes and
edition moves further in advancing th e role of molecular diag- different grades described within that entry.
nostic s in CNS tumour c lassification but rem ai ns rooted in other
established approaches to tu mour characte rization, including CNS tumour nomenclature
histology and immunohistochemistry. The increasing impact of For CNS tumou r nomenclature , the fifth edition fo llows the
molecul ar app roaches has necessitated a seri es of changes , recommendations of the clMPACT-Utrecht meeting to make
which are summarized in this introdu ctory chapter. nomenclature more consistent and simple j1944J . In the past,
some tumour names included anatomical site modifiers (e.g.
CNS tumour taxonomy "chordoid glioma of the third ventricle") whereas others did not,
CNS tumour classification has long been based on histological despite occurring in specific locations (e.g . "medulloblastoma") ,
findings supported by ancillary tissue-based tests (e.g. immu- and some included genetic modifiers (e .g . "glioblastoma, IDH-
nohlstochemical , ultrastructural). The 2016 classification intro- wildtype") whereas others did not, despite having specific
duced molecular markers as key aspects of classification for a genotypes (e.g. "atypical teratoid/rhabdoid tumour"). Names
relatively small set of enti ties. Given the large increase in knowl- have therefore been simplified as much as poss ible, and on ly
edge of the molecu lar bas is of these tumours , the current fifth location , age, or genetic modifiers with remaining c linical util-
edition refers to numerous molecular changes that are impor- ity have been used (e.g . "extraventricular neurocytoma" vs
tant for the most ac curate class ification of CNS neoplasms. A "central neurocytoma"). Importantly, for tumours with features
summary of the most salient molecular alterations for each CNS that are highly characteristic (e.g . the stereotypical location of
tumou r type is listed in Table 1.01 . chordoid gliomas in the third ventricle) , these specific features
As the mol ecular underpinnings of brain and spinal cord are included in the tumour definitions and descriptions even if
tumours have been further elucidated, challenges have arisen they are not part of the tumour name . Moreover, tumour names
in how to organize the classification of these lesions . Some sometimes ref lect characteristic features that are not found in al l
tumour types are readily and consistently characterized by such lesions . For example, some myxopapillary ependymomas
molecular features; some are only sometimes able to be clas- are not myxoid , some are not papillary, and some are neither
sified by molecular parameters; and others are rarely or never myxoid nor papillary in substantial ways ; and xanthomatous
diagnosed using such approaches . The resulting nosological change is fou nd in a relatively small percentage of pleomorph ic
organ ization is therefore also mixed . For some tumour families , xanthoastrocytomas. Nonetheless , such names represent char-
the fifth edition has grouped tumours according to the genetic acteristic, if not universal , features . The terms may also ref lect
c hanges that effect a complete diagnosis (e.g . IDH and H3 sta- historical associations that have become embedded in common
tus) ; for some , by looser, oncogenic associations (e .g . MA PK usage; for instance. although a medulloblast has not been iden-
pathway alterations); for others , by histological and histogenetic tified in developmental studies, the term "medulloblastoma" is
si mi larities even though molecular signatures vary (e.g . see neo- deeply ing rained in tumour terminology, and chang ing the name
plasms discussed in Chapter 2: Gliomas, glioneuronal tumours, could be quite disruptive to clinical care , scientific experiments
and neuronal tumours) ; and for many (e.g . medullob lastoma), that rely on prior data , and epidemiological studies. Lastly, with
by using molecular features to define new subtypes . This hybrid the change to grading within tumour types (see below}, modi-
taxonomy represents the current state of the field , maturing from fier terms like "anaplastic" are not routinely included ; familiar
what was possible for the 2016 WHO classification but prob- names like "anaplastic astrocytoma" and "anaplastic ol igoden -
ably only an intermediate stage on the way to an even more droglioma" do not, therefore , appear in this classification .
molecul ar future class ification . Examples where the transitional
state of the c urrent classification is especially apparent include Gene and protein nomenclature for CNS tumour
the famil y of paediatric -type diffuse low-grade gliomas (p . 56), classification
1n which some entities have been lumped and others split , with The fifth edition of the WHO Classification of Tumours foll ows
such consensus decis ion s being based on the state of the field the Human Genome Organisation (HUGO) Gene Nomenclature
at the time of final editorial di sc ussi ons. Committee (HGNC) system for gene symbols and gene names
(https //Vvww genenames orgl) 1390), the Human Genome Vari-
ation Society (HGVS) recommendations for sequence variants
(https //varnomen .hgvs org/) (7351. and the International System
A sequence alteration relative to a transcript reference
seq uence is reported using the prefix "c." for the coding DNA
sequence, followed by the nucleotide number and the nucleo-
l
for Human Cytogenetic Nomenclature (ISCN) 2020 reporting tide change . The predicted protein sequence change then fol -
gu1del1nes for chromosomal alterations !2053). Gene symbols lows the prefix "p.", specifying the reference amino acid, the
are presented in italics but proteins and gene groups (e .g . the amino acid number, and the variant amino acid resulting from
family of IDH genes) are not italicized the mutation For example, the most common BRAF variant 1s

Table 1.01 The _most common diagnostic molecular alterations in major primary CNS tumo urs (continued on the next page)

Tumour type Characteristically altered genes I molecular profile..


Adult-type diffuse gllomas
Astrocytoma. !DH-mutant IDH1 , IDH2
Ol1 godendrogl1 oma, IDH-mutant and 1p/19q-codeleted IDH1, IDH2, 1p/19q
Glioblastoma. IDH-wildtype IDH-wildtype, chromosomes 7 and 10, TERT, EGFR, others
Paediatric-type diffuse low-grade gliomas
Diffuse astrocytoma, MYB- or MYBL 1-altered MYB, MYBL1
Angiocentric ghoma MYB
Polymorphous low-grade neuroepithelial tumour of the young BRAF, FGFR genes
Diffuse low-grade glloma, MAPK pathway-altered MAPK pathway genes
Paediatric-type diffuse high-grade gliomas
Diffuse m1dilne glioma, H3 K27-altered H3 p.K28 (K27) , EGFR, EZHIP
Drtfuse hemispheric glioma, H3 G34-;'nutant H3 p.G35 (G34)
Diffuse paediatric-type high-grade glioma, H3-wildtype and IDH-wildtype IDH-wildtype, H3-wildtype, methylome
Infant-type hemispheric glioma RTK genes
Circumscribed astrocytlc gliomas
Pilocytic astrocytoma KIAA1549::BRAF, BRAF
High-grade astrocytoma with piloid features Methylome
Pleomorphic xanthoastrocytoma BRAF, CDKN2A
Subependymal giant cell astrocytoma TSC1 , TSC2
Chordoid glioma PRKCA
Astroblastoma, MN1-altered MN1 , BEND2
Glioneuronal and neuronal tumours
Ganglion cell tumours BRAF
Dysembryoplastic neuroepithelial tumour FGFR1
Diffuse glioneuronal tumour with oligodendroglioma-like features and nuclear clusters Methylome
Papillary glioneuronal tumour PRKCA
Rosette-forming glioneuronal tumour FGFR1, PIK3CA, NF1
Myxo1d glioneuronal tumour PDGFRA

Diffuse leptomeningeal glioneuronal tumour KIAA 1549:: BRAF, 1p, methylome


Multinodular and vacuolating neuronal tumour MAPK pathway genes
Dysplastic cerebellar gangliocytoma (Lhermitte-Duclos disease) PTEN

Extraventricular neurocytoma FGFR genes (FGFR1 :: TACC1) , IDH-wildtype


Ependymal tumours
Supratenlorial ependymomas ZFTA (C11orf95), YAP1

Posterior Iossa ependymomas PFA molecular profile, PFB molecular profile


Spinal ependymomas NF2, MYCN
- - - -- -- -
C19MC . chromosome 19 microRNA cluster; PFA, poste~lor Iossa group A; PFB, posterior Iossa group B; RTK, receptor tyrosine kinase; SHH , sonic hedgehog.
•Details of molecular alterations are found in the respective sections.

ln troduct 1on 10 CNS tumou1s 9


Table 1.01 The most common diagnostic molecular alterations in major primary CNS tumours (continued)
-- - - - - - - ----------~--
Tumour type Characteristlcally altered genes I molecular profiles•
Embryonal tumours
Medulloblastoma, WNT-activated WNT pathway genes
Medulloblastoma. SHH-activated SHH pathway genes, TP53
Medulloblastoma. non-WNT/non-SHH Methylome
Atypical teratoid/rhabdo1d tumour SMARCB1 , SMARCA4
Embryonal tumour with multilayered rosettes C19MC, DICER1
CNS neuroblastoma, FOXR2-activated FOXR2
CNS tumour with BCOR internal tandem duplication BCOR
Desmoplastic myxoid tumour of the pineal region, SMARCB1-mutanl SMARCB1
Mening1oma NF2, AKT1, TRAF7, SMO, PIK3CA; and in subtypes KLF4, SMARCE1 , BAP1
Solitary fibrous tumour NAB2'. :STAT6
Meningeal melanocytic tumours NRAS (diffuse); GNAO, GNA 11, PLCB4, CYSLTR2 (circumscribed)
Tumours of the seltar region
Adamantinomatous craniopharyngioma CTNNB1
Papillary craniopharyngioma BRAF
C19MC. chromosome 19 micro RNA cluster; PFA, posterior fossa group A; PFB, posterior Iossa group B; RTK, receptor tyrosine kinase; SHH, sonic hedgehog.
•Details of molecular alterations are found in the respective sections.

BRAF:c.1799T>A p.Val600Glu (or BRAF:c.1 799T>A p.V600E grading have changed for the fifth edition : neoplasms are
if single-letter amino acid codes are preferred , as have been graded within tumour types (rather than across different types),
used throughout this volume). Notably, however, this example and Arabic numerals are used (rather than Roman numerals).
assumes that a particular BRAFtranscript reference sequence Nonetheless, because CNS tumour grading still differs from
accession and version have previously been defined (e.g. other tumour grading , the fifth edition endorses the use of the
NM_004333.5). term "CNS WHO grade" when assigning grade.
For some genes, such as those in the H3 histone group,
which are sometimes altered in CNS tumours, there is the Grading within types
potential for contusion with amino acid numbering. Histone As mentioned, CNS tumours have traditionally had a grade
amino acid positions are typically described in the context of assigned to each entity, and grades were applied across dif-
the protein sequence lacking the initiating methionine, resulting ferent entities (1943}. For example , in recent prior editions of
in a single amino acid difference in numbering compared with the WHO classification , if a tumour had been classified as an
the predicted sequence derived from the corresponding gene anaplastic astrocytoma, it was automatically assigned a CNS
transcript. Therefore, the description of histone sequence alter- WHO grade of 111 ; there was no option to grade an anaplastic
ations in many cancers has, to date, differed from the HGVS astrocytoma as grade I, 11 , or IV. Notably, anaplastic (malignant)
numbering by omitting the first amino acid . Next-generation meningiomas were also assigned a CNS WHO grade of 111. Even
sequencing reports, however, follow HGVS guidelines. The though tumours like meningiomas and astrocytomas are bio-
coexistence of these two nomenclatures may lead to confusion logically unrelated, grade Ill tumours in these different catego-
tor pathologists, oncologists, and researchers . To address this ries had somewhat similar survival times. But these were only
issue, the fifth edition uses the legacy protein numbering sys- roughly similar, with the behaviour of an anaplastic astrocytoma
tem in parentheses after the protein-level variant description, for often being quite different from that of an anaplastic (malig-
example "H3-3A:c .103G>A p.G35R (G34R)" or "H3-3A:c .83A>T nant) meningioma. This approach thus correlated grade to an
p.K28M (K27M)". In these examples, as noted above, prior defi- idealized clinical- biological behaviour: at one end of the spec-
nition of the accession and version of the reference transcript trum, grade I tumours were curable if they could be surgically
is required . removed ; at the other, grade IV tumours were highly malignant,
leading to death in a relatively short period of time.
CNS tumour grading This entity-specific and clinical approach to CNS tumour grad·
CNS tumour grading has for many decades differed from the ing was different from the approach used for other (non-CNS)
grading of other (non-CNS) neoplasms, since brain and spinal tumour types (1943) . Most tumours in other organ systems are
cord tumours have had grades applied across different entities graded within tumour types; for example, a malignant peripheral
11943). As discussed below, the fifth edition has moved CNS nerve sheath tumour can be grade 1, 2, or 3. In the 2016 WHO
tumour grading closer to how grading is done for non-CNS neo- classification of CNS tumours , solitary fibrous tumour / haeman-
plasms, but 1t has retained some key aspects of traditional CNS giopericytoma was graded in the latter manner, using a single
tumour grading because of how embedded such grading is in name but with the option of three grades . In this fifth-edition
neu ro -oncology practice. Two specific aspects of CNS tumour volume, the shift to within-type grading has been extended to all
rategones This chan~~ was made for a few major reasons : (1) in editorial d1scuss1ons for the fifth edition, it was argued that
to provide more tlex1b11ity 1n using grade relative to the tumour grades should not be assigned 1f designation of a grade could
type. (2) to emphasize blolog1cal similarities within tumour types confuse clinical care . For instance, WNT-activated medulloblas-
(rather than to approximate clinical behaviour), and (3) to con- toma is an embryonal tumour with an aggressive behaviour if
torm with WHO grading of non-CNS tumour types . left untreated, but it is responsive to current therapeutic regi-
mens such that nearly all patients have long-term survival Des-
c~ 1 ;.cl1/';;1-.,1/oo -1 1 or3c.'1ng ignating this tumour as grade 4 (equivalent to many untreatable
Nonetheless. because CNS tumour grading has for decades paediatric brain tumours with a dismal outcome) could give an
been linked to overall expected clinical-biological behaviours incorrect impression of the prognosis when therapeutic options
(see above) , the fifth-edition WHO c lassification of CNS tumours are discussed in the clinic. Likewise, designating this tumour as
has generally retained the ranges of g rades used for tumour g rade 1, as one might do with neoplasms having a similar prog-
types in prior editions . In this context, for exampl e, !DH -mutant nosis on the basis of surgery alone, could give a false sense
astrocytomas extend from grade 2 to grade 4, and meningiomas that the tumour is biologically benign . Examples of how this
from grade 1 to grade 3; in other wo rd s, at least at the present might apply in other settings are given in Box 1.01 .
time. there 1s neither a grade 1 IOI-I -mutant astrocytoma nor a
grade 4 meningioma . Moreover, given that tumours are graded Combmed h1stolog1cal and molecular grading
on the basis of thei r expected natural history, highly mali gnant CNS tumour grading has traditionally been based exclusively
tumours tor which there are treatments that may greatly modu- on histological parameters , but many molec ular markers can
late their behaviour (e.g . medulloblastoma, germinoma) are still now provide powerful prognostic information . For this reason,
assigned a grade 4 designation in the fifth edition . molecular parameters have now been added for grading and
The above approach to grading is a compromise, since estimating prognosis in multi ple tumou r types. Examples in the
the original underlying prognostic correlations were based on fifth edition include COKN2A and/or COKN28 homozygous
natural history, at a time when few effective the rapies we re deletion in IDH-mutant astrocytomas , as well as TERT pro-
available. Today, however, estimating natural history is nearly moter mutation , EGFR amplification, and +7/-10 copy-number
impossi ble, since practically all patients receive therapies changes in IDH-wildtype glioblastoma. In these situations. a
that can affect overall survival (3346} . In the current context molecular parameter often overrides histolog ical fin dings in
of modern therap ies that markedly affect patient survival, the assigning a grade. Specific instances are discussed with the
necessity of grading every tumou r type is questionable. In fact, relevant tumour types.

Box1.01 Layered report structure, with two examples


Information to be Included:
Tumour site
Integrated diagnosis (combined tissue-based histological and molecular diagnosis)
Histopathological classification
CNS WHO grade
Molecular information (listed)

Example A
This layered report illustrates {1) use of site In the diagnosis, (2) use of a histological diagnosis that does not designate "anaplasia" but the report still assigns a grade, and
(3) use of the NOS designation (in this instance because the case cannot be worked up adequately at a molecular level).

Cerebrum:
Integrated diagnosis: Supratentorial ependymoma NOS
Histopathologlcal classification: Ependymoma
CNS WHO grade: 3
Molecular Information: Derivatives extracted from FFPE tissue were of insufficient quality for sequencing, and insufficient tissue remained for FISH studies.

Examples
This layered report illustrates (1) a tumour type with a subtype, (2) lack of a definite grade, and (3) that the integrated diagnosis does not necessarily include the histological
designation.
Cerebrum:
Integrated diagnosis: Diffuse low-grade glloma, MAPK pathway-altered
Subtype: diffuse low-grade gtioma, FGFR1 TKD-<!uptlcated
Hlstopathologlcal classification: Oligodendrogtioma
CNS WHO grade: Not assigned
Molecular Information: Duplication of the FGFR1TKO (next-generation sequencing)

FF PE. formal1n-f1xed, paraffin-embedded ; TKO, tyrosine kinase domain.

tnt rucluct 1on iu CN tufllours 11


--... .

Table 1.02 Approximate number of fields per 1 mm 2 based on the field diameter and volume , Roman numerals have been used only 1n d iscussion~
its corresponding area referring to studies that used the older designations).
-------.
Fleld diameter (mm) Approximate number of
Reid area (mm2)
fields per 1 mmz NOS and NEC diagnoses
0.40
As detailed elsewhere (1946,1934}. the designations "not oth.
0.126 8
erwise specified (NOS)" and "not elsewhere classified (N EC)'
0.41 0.132 8 allow the ready separation of standard , well-charactenzec
0.42 0.138 7 WHO diagnoses from those diagnoses that result from either (1)
0.43 0.1 45
a lack of necessary diagnostic (e.g. molecular) information {)'
7
(2) non-diagnostic (i .e. for a WHO diagnosis) or negative results
0.44 0.152 7 The "NOS" designation indicates that the diagnostic lnforma.
0.45 0.159 6 tion (histological or molecular) necessary to assign a specific
0.46 0.166 6
WHO diagnosis is not available, alerting the oncologist that a
full molecular workup has not been undertaken or was not sue.
0.47 0.173 6 cessful. In contrast, the "NEC" designation indicates that the
0.48 0.181 6 necessary diagnostic testing has been successfully performed
0.49 0.188 5 but that the results do not readily allow for a WHO diagnosis -
for example, if there is a mismatch between clinical , histological,
0.50 0.196 5
immunohistochemlcal , and/or genetic features . NEC diagno.
0.51 0.204 5 ses are similar to what pathologists have termed "descriptive
0.52 0.212 5 diagnoses", in which the pathologist uses a non-WHO term
to describe the tumour. In this regard, the "NEC " designation
0.53 0.221 5
alerts the oncologist that the tumour does not conform to a
0.54 0.229 4 standard WHO diagnosis, despite the case having received an
0.55 0.237 4 adequate pathologlcal workup . NOS and NEC diagnoses are
0.56 0.246 4 facilitated by the use ot layered integrated reports (see below
and Box 1.01 , p . 11 ).
0.57 0.255 4
0.58 0.264 4 Novel diagnostic technologies
0.59 0.273 4 Over the past century, many novel technologies have impacted
tumour classification, including light microscopy, tissue stains,
0.60 0.283 4
electron microscopy, immunohistochemistry, molecular genet-
0.61 0.292 3 ics , and most recently a variety of broad molecular profiling
0.62 0.302 3 approaches . Each new method burst onto the scene prom1s1ng
to revolutionize classification , and each then eventually found
0.63 0.312 3
a specific niche alongside the existing methods , rather than
0.64 0.322 3 replacing them . In the past couple of decades, nucleic acid-
0.65 0.332 3 based approaches (e .g. DNA and RNA sequencing , ONA FISH,
RNA expression profiling) have clearly shown their abilities to
0.66 0.342 3
contribute to tumour diagnosis and classification , as evidenced
0.67 0.352 3 by the changes in the revised fourth-edition WHO classlfica·
0.68 0.363 3 tion of CNS tumours (2016) and in this fifth-edition volume. The
0.374 3 global availability of such technologies was already increasing
0.69
as the 2016 volume was being prepared (62,1121. and the pas!
few years have witnessed a further expansion of their use. as
Arabic versus Roman numerals well as the emergence of skilful ways to adapt histological to
Traditionally, CNS WHO tumour grades were given as Roman molecular classification recommendations {2808,2994). This
numerals . However, a danger of using Roman numerals in a fifth -edition WHO Blue Book thus incorporates more molecular
within -type grading system is that a "II " and a "Ill " or a "Ill " approaches for the classification of CNS tumours .
and a "IV" can quite easily be mistaken for one another, and Over the past decade, methylome profiling - the use of arrays
an uncaught typographical error could have clinical conse- to determine ONA methylation patterns across the genome - haS
quences (this was less likely when each tumour type had a also emerged as a powerful approach to CNS tumour classifica-
dilferent name. e.g . when "anaplastic" was present in addi- tion [460,463 ,1458}. Most CNS tumour types and subtypes can
tion to "grade Ill ") . Moreover, the fifth -edition WHO Blue Books be reliably identified by their methylome profile , although cave-
emphasize a more uniform approach to tumour classification ats remain in that the optimal methodolog1cal approaches for
and grading across organ systems. and they favour the use of methylome profiling have not yet been determined , regulator}'
Arabic numerals . Given these considerations, particularly to issues have yet to be resolved , and the technology is currently
decrease the possibility of errors. all CNS WHO grades have not widely available [1944f Copy-number profiles can also be
been changed to Arabic numerals in the fifth edition (in this derived from methylation or other data (e.g. 1p/19q codeletiOfl
+7/-10 signature , amplifications, homozygous deletions, and

12 lril rnrJu1 i1t)11 I{) C NS 1u 1 1CJurs


Indications of fu sion event s). At this time, methylome profi ling Box1.02 Newly recognized tumour types
can be recommended as an effective method for brain and
Diffuse astrocytoma, MYB- or MYBL 1-altered
spinal cord tumour classification when used alongside stan-
dard technologies , including histology. Indeed , most tu mour Polymorphous low-grade neuroepitheJ1al tumour of I.he young
types and subtypes can also be reliably identified by other Diffuse low-grade glioma, MAPK pathway-altered
techni.ques (e . ~ . by a combination of histology and a defi ning Diffuse hemispheric glloma, H3 G34--mutant
genetic alteration) However, methylome profili ng may be the Diffuse paed1atnc-type high-grade glioma, H3-wildtype and IOH-wtldtype
most effective way to characterize some diagnostically chal- Infant-type hemispheric glioma
lenging neoplasms, and it may be the only way (currently) to High-grade astrocytoma with ptloid features
identify some rare tumour types and subtypes. The method also
Diffuse gfioneuronal tumour with oligodendroglioma-like features and nuclear
has utility when small biopsy samples are limiting for standard clusters (provisional entity)
technologies Methylome profiling may also be used as a surro-
Myxoid glioneuronaJ tumour
gate marker tor genetic events. for instance when a methylome
Multinodular and vacuolating neuronal tumour
signature is characteristic of an IDH-wildtype glioblastoma in
Supratentorial ependymoma, YAP1tusio~sitive
the absence of IDH mutation testing; but methylome profiling
cannot serve as a surrogate when the demonstration of spe- Posterior fossa group A ependymoma
cific mutations prior to patient treatment is required for targeted Posterior Iossa group B ependymoma
therapies or cl inical trial participation . Moreover, in the interpre- Spinal ependymoma, MYCN-amplified
tation of methylome profiling results , careful attention must be Cribriform neuroepithehal tumour (provisional entity)
paid to the common calibrated score threshold. As discussed CNS neuroblastoma, FOXR2-aclivated
In detail elsewhere {463). thresholds may be set at 0.84 or 0.90 : CNS tumour with BCOR internal tandem duplication
pathologists should be wary about endorsing suggested diag-
Desmoplastic myxoid tumour of the pineal region, SMARCBt-mutant
noses with scores < 0.84 and should probably discard recom-
lntracranial mesenchymal tumour, FET::CREB fusion-positive (provisional entity)
mendations if scores are < 0.50 . As with other diagnostic tests,
the patholog ist must take into account histological features CIC-rearranged sarcoma
(e .g. tumour cell amount and purity) in interpreting results . For Primary intracraniaf sarcoma, DICER1-mutant
the fifth-ed ition WHO classification of CNS tumours, therefore, Pituitary blastoma
it is assumed that most tumour types have a distinct methyla-
tion signature {460). and this is not specified in each definition;
however, information about diagnostic methylation profiling is in counts across specimens . In practice. most quant1f1cation
included in the Definition and Essential and desirable diagnos- of proliferation (mitotic counting . Ki-67/MIB1 labelling) has pri-
tic criteria subsections of the tumour types for which such infor- oritized hotspots (e .g. in meningiomas). but rt can be useful to
mation can provide more critical guidance for diagnosis. include background proli'feration esti mates 1n such assessments
as well (e.g . focally elevated labelling in a diffuse glioma). Digital
Quantification of mitotic counts assessments based on defined-area fields will facilitate this.
The fifth-edition WHO Blue Books aim to standardize mitotic
counting across tumour types . In this CNS volume, therefore , Integrated and layered diagnoses
where possible , mitotic counts have been expressed as the Because of the growing importance of molecular information in
number of mitoses per a defined area in mm 2 , as well as per CNS tumour classification , diagnoses and diagnostic reports
the commonly used denominator of 10 HPF. Given that individ- need to combine different data types into a single integrated
ual microscopes have high-power fields of different sizes, this diagnosis. Such integrated diagnoses form the backbone of
change should standardize the true area over which mitoses the fifth-edition WHO classification of CNS tumours. Even
are enumerated . The approximate number of fields per 1 mm 2 diagnostic terms that do not incorporate a molecular term may
based on field diameter and corresponding area is presented in require a molecular characteristic for diagnosis (e.g. atypical
Table 1.02 . As an example, the discussion on diagnosis of atypi- teratoid/rhabdoid tumour) . Thus. to display the full amount of
cal meningiomas specifies "~ 2.5 mitoses per mm 2 (4 mitoses diagnostic information available, the use of layered (or tiered)
per 10 consecutive 0.16 mm 2 fields)". Therefore. a diagnosis of diagnostic reports is strongly encouraged , as endorsed by the
atypical meningioma based on mitotic count requires the find- International Society of Neuropathology - Haarlem consensus
ing of~ 4 mitoses within a 1.6 mm 2 area. If, for instance, one's guidelines (1939} and the International Collaboration on Cancer
ocular or digital field dimensions are> 0.16 mm 2 , then the con- Reporting (1945} . Such reports feature an integrated diagnosis
version could come out to only 7 or 8 consecutive high-power at the top, followed by layers that display histological , molecular,
fields rather than 10. and other key types of information (see Box 1.01 , p. 11 ).
It is recognized , however, that such a conversion introduces For some tumour types in this fifth-edition volume. the listed
a number of practical challenges. including that mitotic count- diagnostic terms are general ones (e.g. "diffuse high-grade pae-
ing has been reported in many older studies without specifically diatric-type glioma, 1-13-wildtype", wd1ffuse low-grade glioma ,
quantifying high-power field size . In the future. ideally, cut-off MAPK pathway- altered "), and mix-and-match approaches.
values using measured high-power fields should be established using a matrix of histological and molecular features . are
via new and/or repeated studies . And as pathologists adopt necessary to arrive at a specific integrated diagnosis. These
digital approaches to counting mitoses , the use of defined approaches are described for each of these tumour groups and
field sizes will prove helpful, such as for assessing variability are similar to how the 2016 volume classified medulloblastomas

In roduct1on ro CNS tumours 13


Box 1.03 Tumour types with revised nomenclature or revised placement of CNS tumours . In addition , changes have been made to the
Astrocytoma, IDH-mutant nomenclature of some entities , both to clarify molecular altera.
tions and to follow the nomenclature guidelines 1n clMPACT
Diffuse midllne glioma, H3 K27-altered
update 6 !1944) (see Box 1.03). Other nomenclature changes
Chordoid glioma
were made to standardize type names with those in other WHO
Astroblastoma, MN1-altered Blue Books, such as for peripheral nerve and other soft tissue
Supratentorlal ependymoma. ZFTA fusion-positive tumours .
Embryonal tumour with multilayered rosettes Some proposed tumour types were discussed and provision-
Malignant melanotlc nerve sheath tumour ally accepted as types because they appeared to be distinct ,
Solitary fibrous tumour clinicopathological entities . but additional published studies are
Mesenchymal chondrosarcoma (formerly a subtype) needed for full acceptance; these provisional entities are desig-
Adamantlnomatous craniopharyngioma (formerly a subtype) nated as such in Box 1.02 (p . 13). Others were also discussed,
but it was decided that the published literature left too many
Papillary craniopharyngioma (formerly a subtype)
unanswered questions about the nature of the proposed type
Pituicytoma, granular cell tumour of the sellar region, and spindle cell oncocytoma
for it to be added as a provisional entity at this time An exam-
(now grouped rather than separate)
ple of this is neuroepithelial tumour, PATZ1 fusion-positive , few
Pituitary adenoma I pituitary neuroendocrine tumour
cases of which are described in the literature !2928 ,3037.410).
Although unpublished data suggest that these lesions have
(1939} and to what clMPACT update 4 recommended for pae- distinct molecular alterations, there is marked heterogeneity in ..
diatric low-grade diffuse gliomas (838} : an integrated diagnosis their histopathological appearances and clinical courses , and
optimally comb ines a term from a histologically defined list of therefore more published data are needed to evaluate whether
tumours and a term from a genetically defined list of tumours neuroepithelial tumour, PATZ1 fusion-positive , is truly a distinct
(e.g. see Example B in Box 1.01 , p. 11). Even though each list tumour type .
may contain many items, some combinations are more common Prior editions of the WHO classi'fication of CNS tumours
than others, such that the resulting number of routinely used included more comprehensive lists of "non-brain" tumours . But
integrated diagnoses is typically manageable. because some of these neoplasms are only rarely encountered
In this fifth-edition volume, the essential and desirable diag- in the CNS and their diagnostic information is more comprehen-
nostic criteria for each tumour type are presented in systemati- sively covered In other WHO Blue Books , this fifth-ed ition CNS
cally structured text boxes. in the hope that such a format will volume lists fewer tumours in some sections , such as those on
make it easier for the user to evaluate whether key diagnostic mesenchymal , non-meningothelial tumours and haematolym-
criteria are present and to discern what comb inations of such phoid tumours . The reader is referred to the correspond ing
criteria are sufficient for diagnosis. The essential diagnostic cri- WHO Blue Books for information on those types .
teria are considered must-have features , but there may be dif-
ferent combinations that can confirm a diagnosis (i .e. not all cri- Conclusion
teria are necessarily required for the diagnosis to be rendered) . All classifications are imperfect representations , reflecting the
For the tumour types for which th is is the case, the user should state of understanding in a field at a particular time as well as
pay close attention to the "OR" designations under the "Essen- how that information is interpreted by a handful of experts . The
tial " heading within the Essential and desirable diagnosUc cri- fifth-edition Central nervous system volume of the WHO Classifi-
teria boxes . The desirable diagnostic criteria are nice-to-have cation of Tumours series , like its predecessors , should therefore
features (i .e. they support a diagnosis but are not mandatory). be seen as a work in progress , as a stage in evolution . We have
attempted to introduce new knowledge into the classification
Newly recogn ized entities and revised nomenclature as carefully but progressively as possible, by introducing newly
The major changes to the classification are discussed in the recognized entities, phasing out ostensibly obsolete tumour
introduction sections relating to the families of tumours (e .g. see types , and adjusting the taxonomic structure. It is hoped that
Gliomas, glioneuronal tumours, and neuronal tumours: lntroduc- such changes and their explanations provide practical guid-
Uon) and are only briefly mentioned here. ance to pathologists and specialists in neuro-oncology around
A number of newly recognized types (see Box 1.02, p. 13) the world and that such progress benefits the patients who are
have been accepted into the fifth-edition WHO classification affected by CNS tumours .

14 Introduct ion 10 CNS tumours


Louis ON Perry A
Gliomas , glioneuronal tumours, and Brat DJ Pfister SM
Ellison OW Reifenberger G
neuronal tumours : Introduction Figarella-Branger 0 van Deimlrng A
Hawkins CE

groups . they are compared in a series of t-d istribu ed stoc


Gliomas, glioneuronal tumours , and neuronal tumours are the
neighbour embedding (t-SNE) plots based on DNA me hyl
most common and most varied tumours affecting the paren-
data: Fig . 2.01 shows an overall plot of gliomas . ghoneur
chyma of the CNS . This fifth edition of the WHO classiflcation
tumours . and neuronal tumours, and the rest of the figures
of CNS tumours divides them into six different groups: (1) adult-
this introduction show individual plots of adult-type diffuse
type diffuse gliomas (the brain tumours that of ten constitute the
mas, paediatric-type diffuse low-grade gliomas, paediatrrc-typS
bulk of adult neuro-oncology practice, e.g. glioblastoma , IDH -
diffuse high-grade gliomas, circumscribed astrocytic g lio~
wildtype) , (2) paediatric-type diffuse low-grade gliomas (typi-
and glioneuronal and neuronal tumours.
cally associated with a favourable outcome), (3) paediatric-type
A number of newly recognized types have been added
diffuse high-grade gliomas (generally aggressive tumours),
the classification of the gliomas as well as the glioneuronaJ
(4) circumscribed astrocytic gliomas ("circumscribed" referring
neuronal tumours: diffuse astrocytoma, MYB- or MYBL 1-alterea
to their more contained growth pattern , as opposed to the inher-
polymorphous low-grade neuroepithelial tumour of the y •
ently "diffuse" tumours in groups 1, 2, and 3), (5) glioneuronal
diffuse low-grade glioma, MAPK pathway-altered; diffuse
and neuronal tumours (a diverse group of tumours. commonly
spheric glioma, H3 G34-mutant; diffuse paediatric-type h. .
featuring neuronal differentiation), and (6) ependymal tumours
(ependymomas; now classified by site as well as histological grade glioma, H3-wildtype and IDH-wildtype; infant-type he
and molecular features) . Notably, choroid plexus tumours, with spheric glioma; high-grade astrocytoma with piloid features
their marked epithelial characteristics, are now separated from diffuse glioneuronal tumour with oligodendroglioma-like fea res
the category of gliomas, glioneuronal tumours, and neuronal and nuclear clusters (provisional); myxoid glioneuronal tumo
tumours . To illustrate some biological relationships among these and multinodular and vacuolating neuronal tumour.

··~
.. .
·~~6!
••
• • • • i: I Ol>godendrogl~~a. IOH-mut~r·~~nd_Jp! 19Q-axie~-
-}:~'41
, ..
10

.•. ...
••
~ ~

..:".
.
• •. #e f AstrocytorN": 1c5i=kTiutanl

• ·:iA'
--~~•• • ~qlof!la IDH·muta~t -~<1e) J

·10

·20 -
·20 - 10 10

Fig. 2.01 Gliomas, glioneuronal tumours, and neuronal tumours: molecular groups. Fl~. 2:02 Adult-type . diffuse gliomas: molecular groups. Unsupervised, notrl'
~ns_upervised , non-linear \-distributed stochastic neighbour embedding {t·SNE) pro- t-d1~tnbuted stochastic neighbour embedding (t-SNE) projection of methyla00n arrar
1ection of methylation array profiles from 2632 tumours. Samples were selected from pro~lles from 343 tumours. Samples were selected from a large database of > so ro>
a l~~e database _of> 50 OO?.br~in tumour datasets to serve as reference profiles for ~ram tumour datasets t? se~e ~s reference profiles tor training a supervised classrfCa-
training a s~perv1~ed class1flca_11.on '.11odel based on strict criteria: all these samples t~on model based on stnct cntena: all these samples showed a high cahbrated c~
showe? a h19~ calibrated class1f1cat1on score {> 0.9) when applying the brain tumour tion score (> 0.9) when applying the brain tumour classifier available at https:/twww.rro-
class1fl~r available ~t https:l!www.molecularneuropathology.org. CN, central neuro- le~ularneurof atholo~y.org . "A~tr.ocytoma, IDH-mutant" and "Astrocytoma. IDH-mutant
cytoma, DGONC, diffuse ghoneuronal tumour with oligodendroglioma-llke features (h_1gh-gra?el .comprise two distinct methylatlon groups that were strongly associated
and nuclear clusters; DLGNT, diffuse leptomeningeal glioneuronal tumour· RGNT with surv~val m several different patient cohorts {2916}. Further molecular heterogenelt)'
rosette-forming glioneuronal tumour. ' ' of t~e "Glioblastoma, IDH-wildtype" group can also be assessed by methylation analysis.
which ha~ revea!ed ~e~eral stable molecular subgroups (not shown here) that appear to
be associated with d1stmct prognosis and/or response to therapy {3427}.

16 Gl1omas , glioneuronal tumou rs, and neurona l tumou rs


L / _E;!!" ~r '111 " 11r J ~I V2l '1
1
"'"ri

10
~
.. ....
. .-: :
..
.. ..
/

.3 - 10

.•••.c ••

-6
f' · .r !-~'"' -1 ,\-~rud•' ri1

---
t•ro1::2_1!h" l1al !I rnour of lh-;_: you-;;-q
- -
. ..
-20
. ' .. , ..•
. :. ~?.~.:-:
• • • 9:' ••
·.. i: -

-10 10

Fig. 2.03 Paediatric-type diffuse low-grade gliomas: molecular groups. Unsuper- Fig. 2.04 Paediatric-type diffuse high-grade gllomas: molecular groups. Unsuper-
vised, non-linear I-distributed stochastic neighbour embedding (t-SNE) projection of vised, non-linear !-distributed stochastic neighbour embedding (t-SNE) projection of
methylation array profiles from 66 tumours. Samples were selected from a large da- methylation array profiles from 218 tumours. Samples were selected from a large da-
tabase of > 50 000 brain tumour datasets to serve as reference proriles for training a tabase of > 50 000 brain tumour datasets to serve as reference profiles for training a
supervised classification model based on strict criteria: all these samples showed a supervised classification model based on strict criteria: all these samples showed a
high calibrated classification score (> 0.9) when applying the brain tumour classifier high calibrated classification score (> 0.9) when applying the brain tumour classifier
available at https:/lwww.molecularneuropathology.org. available at https://www.molecularneuropathology.org.

The ependymomas have been reclassified ln two ways: (1) by major types; in addition, their nomenclature and grading have
anatomical site and (2) by the addition of genetic or epigenetic been changed .
types: supratentorial ependymoma, YAP1 fusion-positive;
posterior fossa group A (PFA) ependymoma; posterior fossa Division of diffuse gliomas into adult-type and
group B (PFB) ependymoma; and spinal ependymoma, MYCN- paediatric-type
amplified . The approach to classifying ependymal tumours by The fifth edition recognizes the clinical and molecular distinc-
anatomical site is discussed in Ependymal tumours: Introduc- tions between diffuse gliomas that occur primarily in adults
tion (p. 159). (termed "adult-type") and those that occur primarily in ch ildren
For some of these entities (most notably for diffuse paediatric- (termed "paediatric-type"). Note the use of the word "primarily"
type high-grade glioma, H3-wildtype and IDH-wildtype, and for here; paediatric-type tumours may sometimes occur in adults,
diffuse low-grade glioma, MAPK pathway-altered), histological particularly younger adults, and adult-type tumours may (more
appearance and defined molecular features must be combined rarely) occur in children . Nonetheless, the division of the clas-
to arrive at an integrated diagnosis. Such data are most effec- sification into adult-type and paediatric-type diffuse gliomas is
tively displayed as layered diagnoses. These approaches are hoped to be a major step forward in clearly separating these
discussed for the relevant types and subtypes. clinically and biologically distinct groups of tumours . The need
There have also been some nomenclature changes to exist- to do so has been considered for a long time, but the elucida-
ing entities . For example, the diffuse midline glioma is now des- tion of molecular differences has now made this possible. It is
ignated as '' H3 K27-altered" rather than "H3 K27M-mutant" in specifically hoped that this distinction will enable better care of
order to recognize the various manners in which the pathogenic children with brain tumours .
pathway can be altered in these tumours. Astroblastoma has
been specified as "MN1-altered" to improve diagnostic focus Simplification of the classification of common adult-type
for this entity. For other tumour types, changes in nomenclature diffuse gliomas
relating to the inclusion of genetic and anatomical site modifiers In the 2016 WHO classification of CNS tumours , the common
have followed the recommendations of the Consortium to Inform diffuse gliomas of adults were divided into 15 entities , largely
Molecular and Practical Approaches to CNS Tumor Taxonomy because different grades were assigned to different entities
(clMPACT) update 6 11944). (e.g. anaplastic oligodendroglioma was considered a different
As discussed in more detail below, three sets of major type from oligodendroglioma) and because NOS designations
changes have affected classification and grading of the diffuse were assigned to distinct entities (e.g. diffuse astrocytoma
gliomas. The paediatric-type diffuse gliomas have been sepa- NOS). In contrast, this fifth edition includes only three types:
rated from the adult-type diffuse gliomas. And for the adult-type astrocytoma, IDH-mutant; oligodendroglioma , !DH-mutant and
diffuse gliomas , their classification has been simplified to three 1p/19q-codeleted ; and glioblastoma, IDH-wildtype.

Gl1omas , glioneuronal tumours , and neuronal tumours 17


This focusing of the classification has resulted f_rom (1) more 1
''II . ..
• ·~· • • P1 1ncv!lc. astrr1rytorn1
ecumenical use of the designations "not otherwise specifieo I
•t • ....
• l , ••• (NOS)" and "not elsewhere classified (NEC)", as discussed ,

.'.. ... ..'·.


10
""<.. the general introduction to this volume and in clMPACT update

. ·.~t ... ...........


.
... , .......v.·......•..-. ,,,. .
;. ...
11946); (2) recogn ition of the value of molecular d iagnostics to
assign older entities (e.g. ol igoastrocytoma or IDH-wildtype
..:-:~.··
\
... ·... ~..
diffuse astrocytoma) to more objectively defined types; ano
.........
·'~· ..
. ....
. ·.. · ..~~·.-c:.::
.. ..
. ... . .
: . ... .... .
(3) use of grades within types 1357,1944) rather than requiring
.·.... . each grade to have a different name (see Chapter 1: Introduc-

..·:::-:·...:.. . . .
.·::*:. tion to CNS tumours) .

.. Nomenclature and grading of common adult-type diffuse


astrocytic g/iomas
Pl on1orp l1 1r ~an:~"
°'-"°''~m~'. ; In the 2016 classification, IDH-mutant diffuse astrocytic tumours
· 10
:.• . were assigned to three different tumour types (diffuse astrocy-
toma , anaplastic astrocytoma, and glioblastoma) depending on
.. . 't.
.•...:•. ..·...
... histological parameters. In the current classification , however
·: fl:.. all !DH -mutant diffuse astrocytic tumours are considered a
· ~.,.---~~~~~~~~ :::::..._~-
. Hig~ilrade astrocyt~a-with p1to1~eatu~es
···~~-~
• :~.
. single type (astrocytoma, !DH-mutant) and are then graded as
CNS WHO grade 2, 3, or 4 (see Chapter 1: Introduction to CNS
·20

-20
tumours) . Moreover, grading is no longer entirely histological.
20
because the finding of CDKN2A and/or COKN2B homozygous
Rg.2.05 Circumscribed astrocytic gliomas: molecular groups. Unsupervised, non- deletion results in a CNS WHO grade of 4 even In the absence
linear t-distributed stochastic neighbour embedding (t-SNE) projection of methylation of microvascular proliferation or necrosis.
array profiles from 420 tumours. Samples were selected from a large database of For IDH-wildtype diffuse astrocytic tumours in adults, a num- W
> 50 000 brain tumour datasets to serve as reference profiles for training a supervised
ber of papers have reported that the presence of at least one of ,
classification model based on strict criteria: all these samples showed a high cali-
brated classification score (> 0.9) when applying the brain tumour classifier available three genetic parameters ( TERT promoter mutation , EGFR gene f
at https://www.molecularneuropathology.org. amplification, and the combination of whole chromosome 7 •
gain and whole chromosome 10 loss [+7/-10]) appears to be
sufficient to assign the highest CNS WHO grade (CNS WHO •
D ~ ,., ..:~·c ~ f~n~f'Ol1oqlioma I desmoplashc infant1 'e a:;trocytoma , grade 4) {356 ,3168). The fifth edition therefore incorporates

\.. ··.···
...... these three genetic parameters as criteria for the diagnosis
of glioblastoma. IDH-wildtype. As a result , IDH-wildtype glio-
Cer r;,1 r e u rocyt ....,,-:3
- -- ~.,
I
Roserte-form ing ghoneuronal tumour ; • blastoma can be diagnosed in the setting of an IDH-wildtype
•• • t
:··t.
. ·.f*':
....
•y •
_§J~n~I~~I•. ;••
....... '...
..: . .·.:.,..... . ..
l
I ~~.o!.Q_g!1one u rona l tumour
diffuse astrocytic glioma if there is microvascular proliferation,
necrosis, TERT promoter mutation , EGFR gene amplification, or
+7/-10 chromosome copy-number alteration .
... .. .: ':\-;: .
.·:·.. -~.··..·.
- --- ~1 -_.····.··=
Dysembryopl.:>st: c ~euroep~thel1a_l_.!umo ur , ••

. Extravuntncular neurocytom_a ,..- •


.
·=·· ., .
! : .•. ..

' ·

~··
.5

1Papillary g~oneurona l tumour I

1 Diffuse toptomentngeal9tioneu~nat tumoi,ir I


.
· 10
y;·.
DGONC I •: ..,,
.JO -1 0 10
· 20

Flg.2.08 Glioneuronal and neuronal tumours: molecular groups. Unsupervised, non-


linear !-distributed stochastic neighbour embedding (t-SNE) projection of methylatlon
array profiles from 297 tumours. Samples were selected from a large database of
> 50 000 brain tumour datasets to serve as reference profiles for training a supervised
classification model based on strict criteria: all these samples showed a high cali-
brated classification score (> 0.9) when applying the brain tumour classifier available at
https://www.molecularneuropathology.org. DGONC, diffuse glioneuronal tumour with
oligodendroglioma·like features and nuclear clusters.

18 GIHirnss gl1CJr1 ~ urorial tu 1 1 ou r ~ . ar 1J 118uronal tu mou r


Astrocytoma , !DH-mutant Brat DJ
Cahill DP
Komor1 T
Reuss DE
Cimino PJ Suva ML
Huse JT van Deimling A
Kleinschmidt- Weller M
DeMasters BK

Definition
Astrocytoma , IDH -mutant , 1s a diffusely infiltrating /OH1- or /OH2-
mutant glioma with frequent ATRX and/or TP53 mutation and
absence of 1p/19q codeletion (CNS WHO grade 2, 3, or 4).

ICD-0 coding
9400/3 Astrocytoma, !DH-mutant, grade 2
9401 /3 Astrocytoma, !DH-mutant, grade 3
9445/3 Astrocytoma, !DH-mutant, grade 4

ICD-11 coding
2AOO .OY & XH6PH6 Other specified gliomas of brain & Astro-
cytoma, NOS
2AOO .OY & XH2HK4 Other specified gliomas of brain & Diffuse
astrocytoma, !DH-mutant

Related terminology
Not recommended: diffuse astrocytoma , !DH-mutant; anaplas-
tic astrocytoma, !DH-mutant; glioblastoma, !DH-mutant; low-
grade astrocytoma; lower-grade astrocytoma; high-grade
astrocytoma; infi ltrating astrocytoma; diffuse glioma.

Subtype(s)
Astrocytoma, !DH-mutant, CNS WHO grade 2; astrocytoma ,
!DH -mutant, CNS WHO grade 3; astrocytoma, !DH-mutant,
CNS WHO grade 4

Localization
Astrocytomas with IDH1 or IDH2 mutation can be located 1n any
region of the CNS , includ ing the brainstem and spinal cord, but
they most commonly develop in the supratentorial compartment Fig. 2.07 Astrocytoma, IDH-mutant. CNS WHO grade 2. This unsuspected tumour
and are usually centred near or within the frontal lobes j 1789, was identified at autopsy in a man in his thirties who had last been known to be alive
3040,473). This localization is similar to that of !DH-mutant and 2 days prior; he was found dead at home. A Note the exophyt1c right parieto-occipital
1p/19q-codeleted ollgodendroglioma /1790,3617}. A geneti- mass lesion that blurs cortical anatomical features and Is associated with cerebral
oedema. Herniation was identified at autopsy and histological examination proved
cally distinct form of IDH-mutant astrocytoma has recently been
IDH-mutant status. B Coronal sectioning of the brain (after brief formalin fixation) re-
described in the infratentorial compartment. vealed a non-necrotic, Ill-defined gelatinous tumour in the right parieto-occipital lobe,
with mass effect. Note the ventricular compression and blumng of the grey matter-
Clinical features white matter junction produced by the tumour compared with the normal left side of
Development of signs and symptoms Is rarely abrupt unless the the brain.
diagnosis Is revealed by neuroimaging after onset of epileptic
seizures. A small subset of tumours are diagnosed incidentally difficulties. changes in sensory or motor function , or changes in
when neuroimaging is performed after trauma or for headache vision, may be pre-exisung . With frontal lobe tumours. changes in
13417}. Neurocognitive function in patients with !DH-mutant behaviour or personality may be the initial clinical feature and may
astrocytomas is relatively preserved, compared with that in have been present for months or even years before diagnosis.
patients with similar-sized IDH-wlldtype tumours 13403); this
may be because of slower growth, which allows for compensa- Imaging
tory neuroplasticity. Among !DH -mutant astrocytomas, higher- Neurolmaging findings of !DH-mutant astrocytomas can vary
grade tumours are assumed to be associated with shorter based on location , extent of disease, and tumour grade. On
clinical history, but this has not been confirmed In contemporary CT, !DH-mutant astrocytoma, CNS WHO grade 2, is noted as
studies. a poorly defined, homogeneous, low-density mass without
Seizures are a common presenting sign; however, subtle contrast enhancement. Calcification and cystic change may
neurological abnormaJities, such as speech or language be present. Midline deviation , extensive oedema, contrast

Gl1 omas , gl1 oneuronal tumours and n uronal tumours 19


in their thirties or forties (median age : 38 years) with CNS VVHo
grade 2 or 3 tumours 1452,26551: C.NS WHO grade 2 and J
astrocytomas have similar age d1stribut1ons 12655], where
patients with CNS WHO grade 4 tumours t~nd to be sligh ,
older 12655). IDH mutations are uncommon in paediatric glio..
mas ; when present , they are usually seen in adolescents 12s3 1
917) . !DH -mutant astrocytomas are uncommon in patients agec
> 55 years (203,2687) . There is a slight male predominance
among CNS WHO grade 2 and 3 !DH-mutant astrocytomas
(452) . One study of CNS WHO grade 4 tumours fou.nd that their
M:F ratio is significantly lower than that of IDH-w1ldtype gho...
blastomas (2313) . Recent work indicates that most CNS WHO
grade 4 IDH -mutant astrocytom~s occur de novo, rather thar
with a history of a lower-grade gl1oma {1714} .

Etiology
Genetic susceptibility
Most !DH-mutant astrocytomas develop sporadically, in the
absence of a familial or hereditary predisposition syndrome
Genome-wide association studies indicate an association
between a low-frequency SNP at 8q24 .21 and increased ns
of IDH-mutant gliomas, including oligodendroglioma and astro-
cytoma (1466}. Increased !DH-mutant astrocytoma risk is also
associated with variants at 8q24.21 (the CCOC26 locus), as well
as with variants at the PHLOB1 , AKT3, IDH1, and 02HGOH loci
(2065,2907,823}. Rare genetic syndromes predispose to IDH·
mutant astrocytoma. For example, it is the brain tumour most
frequently associated with Li-Fraumeni syndrome, which is
Fig.2.08 Astrocytoma, IDH-mutant. A CNS WHO grade 3 tumour. Postcontrast T1-
we1ghted MRI demonstrating an infiltrative mass involving the left frontal and parietal characterized by germline TP53 mutations {1648,2320} (see Li-
lobes that is T1-hypointense and without contrast enhancement, a pattern typical of Fraumeni syndrome, p. 446). In patients from three families with
grade 2 and 3 IDH-mutant astrocytomas. B CNS WHO grade 4 tumour. Postcontrast Li-Fraumeni syndrome, IDH1 mutations were observed in five
T1-weighted MRI showing a mass centred in the right frontoparietal region demonstrat- astrocytomas that developed in members with a TP53 germ-
ing rim enhancement surrounding central necrosis with adjacent T1 hypointensity, typi- line mutation. All five contained the IDH1 :c .394C>T p.R132C
cal of a grade 4 IDH-mutant astrocytoma. C CNS WHO grade 3 tumour. T2-weighted
mutation (3398}, which in sporadic astrocytic tumours accounts
MRI demonstrating a homogeneously hyperintense mass centred in the left frontal lobe
and involving white matter and cortex, with mass effect leading to midline shift. The for < 5% of all IDH1 mutations {186 ,3397,3514}. This selective
corresponding FLAIR image (D) shows heterogeneous signal across the lesion, and occurrence suggests a preference for IDH1 p.R132C mutations
this T2-FLAIR mismatch is characteristic of IDH-mutant astrocytoma. D CNS WHO in neural precursor cells that already carry a germline TP53
grade 3 tumour from the same patient shown in panel C. FLAIR MRI demonstrating a mutation . !DH-mutant gliomas (oligodendrogliomas and astro-
mass lesion centred in the left frontal lobe involving the white matter and cortex with cytomas) have also been diagnosed in patients with inherited
heterogeneous signal intensity and demonstrating distinct regions of hyperintensity and Oiiier disease, which predisposes to multiple enchondromatosis
hypointensity. The corresponding T2-weighted image (C) shows homogeneous signal
intensity, and this T2-FLAIR mismatch is characteristic of IDH-mutant astrocytoma.
and chondrosarcoma {980,1332,3123,320). !DH-mutant astro-
cytomas have not been associated with other diffuse glioma
predisposition syndromes including neurofibromatosis type 1
enhancement, and central hypodensity due to necrosis become (6751, POT1 germline mutation {176 ,27471. or melanoma-astro-
evident at higher grades. MRI typically shows T1 hypodensity cytoma syndrome (507).
and T2 hyperintensity, with enlargement and distortion of involved /OH1-mutant astrocytomas in children and young adults are
areas. T2 hyperintensity is often paired with relative hypointen- enriched for germline mutations in mismatch repair genes 1766J.
sity on FLAIR sequences (known as the T2-FLAIR mismatch For these patients, immunohistochemical staining for mismatch
sign) , a finding highly suggestive of CNS WHO grade 2 and repair proteins is usually an effective screening tool {1781
grade 3 !DH-mutant astrocytomas (2419) . Gadolinium enhance- Importantly, the management of these tumours may be different
ment is uncommon in CNS WHO grade 2 !DH-mutant astrocyto- from that of other /OH1-mutant gliomas [3222) .
mas (3277,3602,1518), but it is present at increasing frequency
in CNS WHO grade 3 and grade 4 tumours. A pattern of rim Other etiological factors
enhancement around central necrosis is most common in CNS Diffuse gliomas can arise after therapeutic radiation for another
WHO grade 4 tumours. More extensive peritumoural oedema is CNS malignancy, but these tumours lack IDH mutations {1928}
noted in higher-grade lesions. Although gliomas can be induced experimentally in rats with
chemical carcinogens such as ethylnitrosourea and methylnitro·
Epidemiology sourea, there is no convincing evidence that these substances
Precise population-based data on the incidence of !DH -mutant have an etiological role in human gliomas. Similarly, although
astrocytomas are not available. The majority of patients present polyomavirus (SV40, BK virus , and JC virus) genome sequences

20 Gl1omas glioneuronai tumours , and neuronal tumours


and proteins have been reported in gl!omas , they were only with IDH-wildtype gl1oblastomas . !DH-mutant gltomas show
rarely found 1n a more recent large series of 225 gllomas !2712] . a much stronger predilection for 1nvolv1ng frontal locauons ,
indicating that they may originate from precursor cells located
Pathogenesis In (or m1grat1ng to) the frontal lobe Moreover, the preferential
Cell of origin frontal lobe localization may be a consequence of the frontally
Identif ication of the cell(s) of origin of !DH -mutant astrocytoma restncted expression of the glutamate dehydrogenase gene
remains an area of active investigation (see also Oligodendro- GLU02, which encodes GDH2 , a hom1no1d-spec1f1c enzyme
g/10ma , !OH-mutant and 1p/19q-codeleted, p. 28) . Morphology that facilitates glu amate turnover in human forebra1n 1540,
and single-cell RNA-sequencing analysis of human tumours 33541 . GDH2 activity may partially compensate for IDH muta-
supports the notion that !DH-mutant astrocytomas are com- tion-induced metabolic alterations, thereby fac ilitating the
posed of a mixture of malignant cell types that recapitulate early stages of !DH -mutant astrocytoma grow h. Together,
astrocytic and oligodendroglial lineages , as well as neural these observations support the hypothesis that !DH-mutant
precursor-like cells 13309}. Experimental transformation of astrocytic gl1omas develop from a distinct population of pre-
immortalized human astrocytes with IOH1 p .R132H repro - cursor cells, although this has not yet been studied carefully
grammes their cellular lineage and favours the emergence of a across grades (2313 ,3309) .
neural precursor state 13239}. Experiments in transgenic mice
indicate that astrocytomas may originate from different CNS Genetic profile
cell types. including neural precursor cells , oligodendrocyte !DH mutations were first reported in 2008 (24051 and are a defin-
precursor cells , and astrocytes 13619). Neural and ollgoden- ing feature for IDH-mutant astrocytoma . CNS WHO grades 2, 3,
drocyte precursor cells may give rise to either ollgodendroglial and 4. IDH mutations are an early event in gliomagenes1s and
or astrocyt1c phenotypes in gliomas, depending on the genes persist during tumour progression in most cases. Analysis of
driving transformation !1915,1901 ,1685}. This suggests an serial biopsies from the same patients have not yet uncovered
interplay between oncogenic events and the cell(s) of origin, cases 1n which an /OH1 mutation occurred after the acqu1s1-
and indicates that both are critical in determining the result- tion of a TP53 mutation (3397) . An exception 1s IOH·t mutations
ing glioma phenotype. !DH-mutant gliomas and IDH-wildtype in patients with Li-Fraumeni syndrome. in which the germline
gl1oblastomas may derive from different precursor cells . The TP53 mutation is the initial genetic alteration and influences the
differing patient ages. sex distribution , and clinical outcome subsequent acquisition of the !OH1 mutation (see Li-Fraumeni
suggest that !DH-mutant and IDH-wildtype gliomas have dis- syndrome, p . 446) (3398}. Reported !OH1 mutations are usually
tinct cellular pathogenetic mechanisms 123131. Compared located at the first or second base of codon 132 !186,2405,

Gl1omCl.s . gliunciuronal tun 1ours . o.nJ neuror.a1 tum-Jurs 21


Fig. 2.1 0 Astrocytoma, IDH-mutant, CNS WHO grade 3. A lmmunohistochemistry for GFAP shows strong and diffuse immunoreactivity in the tumour cell cytoplasm and gfial
processes in this !DH-mutant astrocytoma with gemistocytic differentiation. B lmmunohistochemistry for IDH1 p.R132H demonstrates strong cytoplasmic reactivity in all neoplas·
tic cells, indicating an IDH1p.R132H mutation. C lmmunohistochemistry for p53 shows strong nuclear staining in a large percentage of neoplastic cells, which correlateswell wilti
TP53 mutation in the setting of IDH-mutant astrocytoma. D lmmunohistochemistry for ATRX reveals loss of nuclear staining in neoplastic cells, but retention of nucJear staining
1n non-neoplastic endothelial cells and glia, indicating ATRX loss or mutation in this JOH-mutant astrocytoma.

3397 ). The most frequent is the IDH1 :c .395G>A p.R132H muta-


tion, found in 83-91% of !DH-mutant gl iomas (186,3397,3514) .
{1948,3240) and to favour the emergence or maintenance ol 1
a stem cell- like state prone to self-renewal and tumorigenesis
Other mutations are rare, including IDH1:c .394C>T p.R132C {3239). IDH mutations may also promote glioma formation by
(found in 3.6-4 .6% of cases), p.R132G (in 0.6- 3.8%), p.R132S disrupting the binding of the methylation -sensitive insulator
(in 0.8-2.5%) , and p.R132L (in 0.5-4.4%) (186,3397,3514) . protein CTCF, thus affecting chromosomal topology and allow- l
The IDH2 gene encodes the only human protein homologous ing aberrant chromosomal reg ulatory interactions that induce '
to IDH1 that also uses NADP+ as an electron acceptor. IDH2 oncogene expression {945). MGMT promoter methylation is 1

mutations in gl iomas are located at resi due p.R172, with the also commonly observed in IDH-mutant gliomas [2204,32401.
p.R172K mutation being the most frequent (3 502,351 4). IDH2 MGMT encodes a DNA repair protein {598) that removes pro-
p.R172 is the analogue of the p.R132 residue in IDH1 , and it is mutagenic alkyl groups from the 06 position of guanine in DNA
located in the enzyme's active site, forming hydrogen bonds thereby blunting the treatment effects of some alkylating agents
with the isocitrate substrate. /OH2 mutations are much less fre- {860,1062) (see also Glioblastoma, IDH-wildtype, p. 39). How-
quent than IDH1 mutations in IDH-mutant astrocytoma. ever, the predictive ro le of MGMT promoter methylation may be
Glioma-associated IDH1 and IDH2 mutations impart a gain- limited to tumours that additionally exhibit loss of one copy of
of-function phenotype to the respective metabolic enzymes chromosome 10, where MGMT is located (e.g . IDH-wildtype
IDH1 and IDH2, which then overproduce the oncometabolite glioblastoma).
2-hydroxyglutarate (674) . The physiological consequences of !DH -mutant astrocytomas also harbour class-defining loss-
2-hydroxyglutarate overproduction are widespread and include of-function mutations in TP53 and ATRX [1474,1547,452}. ATRX l
profound effects on cellular epigenomic states and gene regu- encodes an essential chromatin-binding protein, and i.ts del~ I
lation 1591 ,931) . The introduction of mutant /OH1 into primary ciency has been associated with epigenomic dysregulat1on an
· · dice
human astrocytes alters specific histone markers and induces telomere dysfunction [611} . In particular, ATRX mutations in L 1
. · k as alter-
extensive DNA hypermethylation (termed the "glioma-associ- an abnormal telomere maintenance mechanism nown I
t t . ns anC1 •
10
ated CpG island methylator phenotype [G-CIMP)"), suggesting native lengthening of telomeres 1·1276). ATRX mu a n
that the presence of an IDH1 mutation is sufficient to estab- alternative lengthening of telomeres are mutually exclusive wdit ,
· h nco e:i
lish a hypermethylation phenotype 13240). Widespread hyper- activating promoter mutations of the TERT gene, wh1c e , '
ter muta
methylation in gene promoter regions is thought to silence the the catalytic component of telomerase. TERT promo 1
expression of several important cellular differentiation factors
. h
tions are rare in IDH -mutant asrrocytomas. butt ay ar
e pres 9r.1

22 Gliomas ' gl1or1euro11eil turriuur~ ' (-j(l(j r 1eurnn.JI tu 1) IOll(!:i


t
Fig. 2.11 Astrocytoma, IDH-mutant, CNS WHO grade 4 with giant cell features. A Giant cell tumours are often IDH-wildtype, but this example was IDH-mutant. Next-generation
sequencing revealed blallelic MLH1 inactivation, as well as hypermutation, including mutations of the IDH1, TP53, and ATRX genes. B This tumour with a mismatch repair
defect showed GFAP immunoreactivity. C Although most giant cell glioblastomas are IDH-wlldtype, this one was !DH-mutant, as evidenced by the immunoreactivity for IDH1
p.R132H. D This giant cell tumour with a mismatch repair defect showed diffuse p53 positivity, consistent with TP53 mutation.

in the vast majority of !DH-mutant oligodendrogliomas and IDH- Macroscopic appearance


wildtype glioblastomas {452,824,1614}. !DH-mutant astrocytomas of low histological grade are expan-
ATRX deficiency has also been associated with general- sile and blur the grey matter-white matter junction. They enlarge
ized genomic instability, which can induce p53-dependent and distort invaded anatomical structures and may show large
cell death in some contexts {628}. Therefore, TP53 mutations or small cysts. Extensive microcyst formation occasionally pro-
in !DH-mutant astrocytomas may enable tumour cell survival in duces a gelatinous appearance, or a single large cyst filled
the setting of ATRX loss. The genomic instability of !DH-mutant with clear fluid . Large, grossly detectable calcifications are not
astrocytomas is reflected in characteristic DNA copy-number present, but diffuse grittiness Is occasionally noted . Higher-
abnormal ities, which include low-level amplification events grade examples may show similar features, but large coales-
involving the oncogenes MYC and CCND2 in mutually exclusive cent zones of yellowish discolouration due to necrosis and/or
subsets (452). Copy-number events typically associated with haemorrhage may also be present.
IDH-wildtype glioblastoma, such as EGFR amplification as well
as PTEN mutation or deletion, are rarely encountered, empha- Hlstopathology
sizing the biological differences between !DH-mutant and IDH- !DH-mutant astrocytomas range from well-differentiated , low-
wildtype astrocytomas {452,824,1145}. cell-density, and slow-growing tumours (CNS WHO grade 2)
to highly anaplastlc, hypercellular, and rapidly progressive
Genetic alterations associated with tumour progression tumours (CNS WHO grade 4).
Multiple retrospective studies indicate that homozygous dele- CNS WHO grade 2 tumours are composed of well-differen-
tion of CDKN2A and/or CDKN28 Is associated with shorter sur- tiated fibrillary glial cells that diffusely infiltrate the CNS paren-
vival in patients with IDH-mutant astrocytomas, corresponding chyma individually, usually without cellular cohesion, generating
to CNS WHO grade 4 behaviour (see also Prognosis and pre- a loosely structured, often microcystic matrix 13303,1398). Cel-
diction, below) 1357). Alterations in other genes encoding mem- lularity is mildly to moderately increased compared with that of
bers of the RB1 pathway, including COK4 amplification and RB1 normal brain , and mild nuclear atypia is characteristic . Tumour
mutation or homozygous deletion, may also be associated with cell density and cellular morphology may vary, the latter with
accelerated growth (357) . POGFRA amplification {357), MET respect to cell size, cytoplasmic abundance, and prominence
alterations 11875,1362), MYCN amplifications, and mutations in of cellular processes . Histological recognition of neoplastic
PIK3R1 and PIK3CA have been associated with shorter survival astrocytes depends mainly on nuclear characteristics . Com-
and may play a role in tumour progression 1357). pared with those in normal astrocytes, the nuclei in !DH-mutant

Gl1ornas, glioneuronal tumou rs, and neu ron al 1un1ours 23


Flg.2.12 Astrocytoma, IDH-mutant, CNS WHO grade 4 with giant cell features. A This tumour with a mismatch repair defect showed loss of ATRX expression by 1m-
munohistochemistry. a MLH1 expression was lost in tumour nuclei but retained in endothelial cells and other non-neoplastic cells In this tumour with a mismatch repa11
defect. c In addition to loss of MLH1 in tumour cells, secondary PMS2 loss was detected by immunohistochemlstry, as expected, given that these two proteins norm
form heterodimers.

astrocytomas are enlarged , and they display irregular nuclear dispersion. Multinucleated tumour cells and abnormal mitoses
contours , an uneven chromatin pattern , and hyperchromasia. may be seen. By definition , microvascular proliferation (multilay-
Overall , monomorphic nuclei and rounded nuclear contours ered endothelia within vessels) and necrosis are absent.
may be seen , occasionally showing morphological overlap with CNS WHO grade 4 tumours must manifest necrosis and/or
oligodendrogl ial tumours. Nucleoli are typically indistinct and microvascular proliferation in addition to the features of CNS
are most often not visible. Unlike in cells undergoing reactive WHO grade 3 lesions, but the designation of CNS WHO grade 4
astrocytosis, cellular processes in JOH-mutant astrocytomas JOH -mutant astrocytoma is also warranted if the tumour shows
usually vary from one tumour cell to the next. Mitotic activity is homozygous deletion of COKN2A and/or CDKN28, even in the
absent or uncommon in CNS WHO grade 2 tumours; a single absence of necrosis or microvascular proliferation (see Grading
mitosis within a resection specimen is compatible with a CNS and Diagnostic molecular pathology) (357). For more informa-
WHO grade 2 designation [357}. tion about the histopathology of microvascular proliferation , see
The principal feature distinguishing CNS WHO grade 3 astro- the Histopathology subsection (p. 45) in Glioblastoma. IDH-
cytomas from CNS WHO grade 2 astrocytomas is increased wildtype.
mitotic activity and histological anaplasia (see Grading , below). The term "glioblastoma" is no longer applied to CNS WHO
However, the threshold for a CNS WHO grade 3 designation has grade 4 JOH-mutant astrocytoma. Morpholog ically, however.
not been established in JOH-mutant astrocytoma cohorts. One the histology of individual cells of CNS WHO grade 4 IOH-
mitotic figure may be sufficient for assigning grade 3 within a mutant astrocytoma has considerable overlap with that of
very small biopsy, whereas more mitoses are required in larger IOH-wildtype glioblastoma, and distinguishing between them
resection specimens 1357). Grade 3 tumours also often display requires testing for JOH mutations. Nevertheless, some features
increased cell density and greater nuclear atypia, including differ. Areas of ischaemic zonal and/or palisading necrosis have
variation in nuclear size and shape, chromatin coarseness, and been observed in 50% of CNS WHO grade 4 IOH-mutant astro-
cytomas, considerably less frequently than in IOH-wildtype
glioblastoma, where they are found in as many as 90% of cases
(2269). Focal oligodendroglioma-like components are more
common in CNS WHO grade 4 IOH-mutant astrocytoma than in
IOH-wildtype glioblastoma 12269,1789).
Gemistocytic differentiation can be noted focally, regionally,
or nearly uniformly in all grades of IOH-mutant astrocytoma.
However, the gemistocytic tissue pattern is not specific to IOH-
mutant astrocytomas and can be noted in IOH-wildtype gliomas
as well. To be considered a major tissue pattern , gemistocytes
should account for (approximately) > 20% of all tumour cells -
a somewhat arbitrary, but useful , criterion 13394,1744,3198).
Gemisto?ytes are characterized by plump, glassy, eosinophilic
cell bodies and stout, randomly oriented processes that form
a coars~ fibrillary network. Nuclei are typically eccentric, with
small, distinct nucleoli and densely clumped chromatin . Perivas-
cular l~mpho?yte cuffing is frequent (417}. Th is tissue pattern is
J ~ .. - ~ associated with a focal gain of chromosome 12p encompassing
Flg. 2.13 Astrocytoma , !DH-mutant, CNS WHO grade 4 with gllosarcoma features. CCND2 l2780) . No definite associations with clinical behaviour
Although most ghosarcomas are IDH-wildtype, this example was IDH-mutant, as evi- are known .
denced by 1mmunoreactlvity tor IDH1 p.R132H protein.

24 Gltornas . glio11eurorn-.il lu1 nours . , rid neuronal tun 1ours


Proliferation
In older studies. when JOH statu s was not 1nco rporo ted Into
the diagnosis. as1rocy tomas with ~ 2 mitoses w1t111n the enti re
specimen were associated with shorter survival than those
Table2.01 Grading criteria for astrocytoma, IDH·mutant

Grade Criteria
----~---

• A diffusely infiltrative astrocytic gliorna with an IDH1


,,
"
w1tl~ < 2 mitoses (633.1091 ,6841. but several studies of patients or IDH2 mutation that is well differentiated and lacks
h1stolog1cal features of anaplasia
with JOH -mutant astrocytoma have not corroborated these
CNS WHO grade 2
thresholds fo r mitotic activity (3539,23 18,809) . However, oth- • Mitotic activity is not detected or very low
ers have demonstrated that traditional grading schemes based • Microvascular proliferation. necrosis, and homozygous
on mitoses can stratlf y risk among patients with grade 2 and deletions of CDKN2A and/or CDKN2B are absent
grade 3 JOH -mutant astrocytomas 1597,291 6.3522) . To date, no
• A dttfusely infiltrative astrocytic glioma with an IDH1 or
studies have established an alternative mitotic count th reshold IDH2 mutation that exhibits focal or dispersed anapla·
that more reliably stratifies risk among histological CNS WHO sia and displays significant mitotic activity
CNS WHO grade 3
grade 2 and grade 3 !DH -mutant astrocytomas .
• Microvascular proliferation, necrosis, and homozygous
Similarly. studies of proliferation (e.g. based on the Ki- 67 deletions of CDKN2A and/or CDKN2B are absent
Index) have not identifie d criteria that unequ ivoca lly stratify
risk among patients with !DH -mutant astrocytomas (809) . The • A diffusely infiltrative astrocytic glioma with an IDH1 or
IDH2 mutation that exhibits microvascular proliferation
growth fraction as determined by the Ki- 67 pro Iiferati on index is CNS WHO grade 4
or necrosis or homozygous deletion of CDKN2A and/or
usually < 4% for CNS WHO grade 2 !DH -mutant astrocytomas. CDKN2B, or any combination of these features
In CN S WHO grad e 3 tumours, the Ki-67 proliferation index is
usually 1n the range of 4- 10%, but it can overlap with values for
CNS WHO grad e 2 tu mou rs at one end of the range and CNS astrocytosis. Because most !DH -mutant diffuse astrocytomas
WHO grade 4 tumours at the other \632 ,1457,1557,2601) . Ki-67 have IOH1 p.R132H mutations, the fi nding of positive immuno-
proliferation index values In CNS WHO grade 4 !DH-mutant staining for IDH1 p.R132H excludes reactive astrocytosis {445)
astrocytomas vary cons iderably and do not appear to be asso- Prior freezing of tissue for intraoperative diagnosis, however,
ciated wi th survival {362) . may abrogate IDH1 p.R132H immunostaining and can distort
nuclear cytological features .
Grading JOH-mutant astrocytoma has a wide differential diagnosis
Astrocy toma . !DH-mutant, can be designated as CNS WHO with other diffusely growing brain tumours . The primary con -
grade 2, 3, or 4, according to the criteria in Table 2.01 [357) . siderations are oligodendroglioma, IDH-mutant and 1p/ 19q-
codeleted; glioblastoma, IDH-wildtype; diffuse mid line glloma,
lmmunophenotype H3 K27- altered ; diffuse hemispheric glioma, H3 G34- mutant;
Individual tumour cells of !DH-mutant astrocytoma reliably diffuse astrocytoma, MYB- or MYBL 1-altered; diffuse low-grade
express GFAP, although to varying degrees {1647). Gemisto- glioma, MAPK pathway-altered ; and high-grade astrocytoma
cytic tumour cells are typically strongly and uniformly positive with piloid features .
for GFAP {3395) . OLIG2 is a transcription factor that shows
strong nuclear immunoreactivity in most forms of diffuse glio- Cytology
mas , including IDH-mutant astrocytomas . One study concluded The use of cytological preparations at the time of intraoperative
that OLIG2 expression was lower in high-grade !DH-mutant consultation can add value to frozen section evaluafon , because
astrocytomas than in other !DH-mutant giiomas (1892,2824}. certain cellular features (e.g. coarse chromatin , nuclear contour
Vimentin is often positive in tumour cells, with a labelling pattern Irregularity, fibrillary processes) may be enhanced or accentu-
similar to that of GFAP {1296}. lmmunohistochemical assays ated . Individual cells show enlarged , angulated , and hyperchro-
used as surrogates for genetic alterations (e.g. IDH1 p.R132H, matic nuclei and may display elongated eosinophillc cellular
p53 , ATRX) are discussed below, in the Diagnostic molecular processes, which often form a loose background of fibrillarity
pathology subsection. that is helpful in defining glial differentiation . With increasing
grade, greater neoplastic cytological pleomorphism and more
Differential diagnosis mitotic figures can be found. In CNS WHO grade 4 !DH-mutant
For CNS WHO grade 2 tumours, the main differential diagnos- astrocytomas, necrotic debris and/or vessels with endothelial
tic considerations are normal brain and reactive astrocytosis. proliferation may also be present In cytological preparations .
The cytoplasm of normal human astrocytes, in contrast to that
of !DH-mutant astrocytoma cells , is not distinct from the back- Diagnostic molecular pathology
ground neuropil. Their nuclei are small and regular, displaying Many of the signature molecular characteristics of !DH-mutant
delicate and uniform chromatin patterns. Reactive astrocytes astrocytoma can be demonstrated immunohistochemlcatly.
are defined by enlarged nuclei (often with a single prominent A routine immunoh1stochemical panel for the initial diagnostic
nucleolus) and clearly distinguished cytoplasm . Reactive gemi- workup of diffuse gliomas in adults involves IDH1 p.R132H ,
stocytes are a specific type of astrocytes that are character- p53, and ATRX {3131,3303).
ized by abundant eosinophillc cytoplasm , eccentric nuclei, lmmunohistochemical staining for IDH1 p.R132H is highly
and prominent glial processes. Reactive astrocytes are often sensitive and specific for the /DH1 p.R132H mutation , and
uniformly distributed throughout tissue and often show simi- immunopositivity is strong evidence of !DH-mutant glioma
lar nuclear and cytoplasmic features , unlike neoplastic cells. (465,351}. IDH1 p.R132H immunohistochemistry has become
Microcystic change strongly favours neoplasia over reactive a critical initial test for the classification of gliomas and also

Gllomas , glioneuronal tumours , and neuronal tumours 25


Fig. 2.14 Astrocytoma, IDH-mutant, CNS WHO grade 3. A Cytopathological smear preparations highlight the large round eosinophilic cell bodies and delicate glial proce
of astrocytoma cells with gemistocytic differentiation. B Smear preparation shows IDH-mutant astrocytoma cells with enlarged, hyperchromatic, and irregular nuclei wit 3
background of extensive fibrillarity created by the elongated fibrillar processes emanating from tumour cell bodies.

helps to distinguish true neoplasia from reactive gliosis (445 , supports the diagnosis of !DH-mutant astrocytoma but is noi
464}. The p.R132H variant accounts for approximately 90% of a surrogate for IDH assessment because loss of nuclear ATRX
all IDH mutations in supratentorial astrocytomas. Of note, more is also found in H3-altered diffuse gliomas and occasionally 1
rarely occurring primary infratentorial !DH-mutant astrocytomas IDH-wildtype gliomas. In add ition, although the rate of ATRX
show a distinctively different spectrum of IDH mutations, and loss is > 90% in supratentorial !DH-mutant astrocytomas, the
about 80% are of the non-p.R132H type \187) . Gene sequenc- rate in infratentorial IDH-mutant astrocytomas is only about 50%
ing analysis of IDH1 codon 132 and IDH2 codon 172 is recom- {187} . The combination of mutations in IOH1 or IDH2 and ATRX
mended in the event of a negative or indeterminate result with in a diffuse glioma (including by immunohistochemistry) is sutti-
the IDH1 p.R132H immunohistochemical stain , in order to rule cient for the diagnosis of IDH-mutant astrocytoma, obviating the
out the possibility of a non-p.R132H IDH mutation [452,3514}. need for 1p/19q testing in order to exclude oligodendroglioma
Given the low frequency of IDH1 and IDH2 mutations in CNS Rare cases of dual-genotype !DH-mutant gliomas have been
WHO grade 4 gliomas arising in patients aged > 55 years , described; distinct regions within these tumours have oligoden-
sequencing analysis need not follow a negative IDH1 p.R132H droglioma morphology and 1p/19q codeletion , while other regions
immunostain in this patient population [746,2687,1245}. have astrocytic morphology, ATRX loss, and TP53 mutations
In the setting of an IDH-mutant glioma , the detection of strong {'1382,2587). A more recent publication documented two dual·
and diffuse p53 immunopositivity can be used as a surrogate genotype !DH-mutant gllomas that displayed uniform tumour
for TP53 mutations and in support of the diagnosis of IDH- morphology throughout, as well as ATRX loss, TP53 mutations,
mutant astrocytoma. TP53 mutation often leads to reduced and 1p/19q codeletion in all tumour regions tested 13588). These
degradation of the protein , and to its nuclear accumulation; cases, although rare, indicate that the defining molecular altera-
however, not all TP53 mutations manifest as strong nuclear tions of IDH-mutant astrocytomas and oligodendroglioma are not
immunoreactivity, and nonsense mutations In particular can absolutely mutually exclusive. The precise classification for these
sometimes be associated with a complete absence of staining . dual-genotype !DH-mutant gliomas has not been established
Strong nuclear p53 immunohistochemical positivity In > 10% However, a layered diagnostic approach that includes morpho·
of tumour nuclei correlates well with TP53 mutations in the set- logical findings and molecular alterations, combined with an
ting of an JOH-mutant glioma, but it is best evaluated in the "NEC" designation, may be appropriate {1946 ,1939).
context of morphology and other immunohistochemistry in the As discussed above, a molecular marker that is strongly asso-
diagnostic panel , such as ATRX {3131,1108 ,3112). Most IDH- ciated with unfavourable prognosis in IDH-mutant astrocytoma is
mutant astrocytomas show even more widespread (> 50%) p53 homozygous deletion of CDKN2A and/or CDKN2B {29161. This
expression 13112). has prompted grading of !DH-mutant astrocytoma with homozy·
Inactivating ATRX alterations commonly co-occur with TP53 gous CDKN2A and/or COKN2B deletion as CNS WHO grade 4,
mutations in JOH-mutant astrocytomas {821 ,279 ,1405). These irrespective of other morphologicaJ signs of high-grade malig·
often result in a truncated protein and abrogated protein expres- nancy such as necrosis or microvascular proliferation l357J.
sion , leading to loss of nuclear ATRX immunoreactivity 12657). Methylation profiling readily identifies JOH -mutant astrocy·
ATRX immunopositivity in the nuclei of endothelial cells and toma because of the profound influence of IDH mutations on
neurons serves as an internal positive control {313'11 because the methylome. Accordingly, the presence of an /OH1 or !DH
ATRX protein is prone to rapid degradation in tissue with even mutation can be reliably inferred by this method , although the
minimal hypoxic damage, so areas of tissue showing nuclear specific amino acid exchange impacting either /OH1 or IDH2
ATRX immunopositivity in these cells should be assessed . cannot be determined . Methylat1on profiling can also be used
Loss of nuclear ATRX expression in neoplastic cells strongly to distinguish between subgroups of !DH-mutant astrocytomas.

26 G l1 0 111as , gl 1oneurona l tumou rs, and neuronal turn o ws


Box 2.01 01agnost1c crltena for astrocyt.oma, !OH-mutant
including those of low and high grade in the supratentonal com -
partment as well as those in the 1nfratentorial regions [460,187), Essential:
see also Fig 2 02 (p. ·16). Copy-number profiles calculated from A diffusely infiltrating ghoma
methylome data can also be useful for determining COKN2A
AND
and/or CDKN2B and lp/19q codeletion status IDH1codon132 or /DH2codon 172 m1ssense mutabon

Essential and desirable diagnostic criteria AND


Loss of nuclear ATAX expression or ATRX mutation
See Box 2 01
OR
Staging Exdus1on of combrned whole-arm dele~ons of 1p and 19q
Nol clinically re levant
Desirable:
Prognosis and prediction TP53 mutation or strong nuclear expression of p53 in > 10% of tumour cells
Clinical prognostic factors Melhylat1on profile of astrocytoma, !OH-mutant
Studies specifically addressing !DH -mutant astrocytomas have Astrocytic differentiation by morphology
confirmed the assoc iation of younger age with longer survival
(2916 ,3522). Similarly, the extent of resection and the presence
of postoperative residual tumour have been shown to correlate grade 4 (123 .2916), with expected median OS of about 3 years
with overall survival (OS) 11514,3442 ,234). (3346} The 1nclus1on of COKN2A and/or CDKN28 homozygous
deletion as a cnterion 1s based on evidence that the occurrence
Proliferation of these deletions in !DH-mutant astrocytomas ot h1stolog1cal
Proliferative activity quantified by mitotic count remains a grad- grade 2 or 3 is associated with worse outcome, corresponding
ing criterion for !DH-mutant astrocytomas, yet several studies of to CNS WHO grade 4 . Some studies have further concluded
IDH-mutant cohorts have not shown significant nsk stratification that homozygous deletion of COKN2A and/or COKN28 is asso-
(3539,2318.809) . Other studies have demonstrated that tradi- ciated with worse outcome among patients with histolog1cally
tional grading schemes based on mitotic activity can stratify defined CNS WHO grade 4 !DH-mutant astrocytomas !"1714,
nsk among patients with CNS WHO grade 2 and 3 !DH-mutant 2916).
astrocytomas (597,2916,3522). In one study of histological In addition to CDKN2A and/or COKN28 homozygous dele-
grade 2 and 3 diffuse gllomas, investigators were not able to tion, several other molecular alterations 1n !DH-mutant astrocy-
identify a Ki-67 threshold that could reliably stratify risk among toma have been associated with malignant progression . Some
!DH-mutant astrocytomas {809). of these occur concomitantly in higher-grade tumours , compli-
cating the assessment of the prognostic impact of individual
Hfstopathological and genetic factors alterations. For example, in some reports , a significantly worse
The histopatholog1cal factors relevant for grading (mitotic activ- prognosis was associated with homozygous deletion of RB1
ity, microvascular proliferation, and necrosis) are relevant for (2916) or amplification of COK4 l597,3522,1 22L whereas others
prognosis (see Proliferation and Grading, above). Patients with did not corroborate these associations {596,123} . DNA meth-
!OH-mutant CNS WHO grade 2 astrocytomas have a median ylation profiling has identified two grade-related methylation
OS of > 10 years (123,2916) . An !DH-mutant astrocytoma groups that are associated with differences in OS 1460,2916}.
that contains considerable mitotic activity and histological Amplif1catlon of PDGFRA was associated with worse progno-
anaplas1a yet lacks microvascular proliferation, necrosis, and sis in several studies (2498,2916,3522). Other copy-number
COKN2A and/or COKN2B homozygous deletion currently fits alterations with a reported negative association with prognosis
into the designation of CNS WHO grade 3 !DH-mutant astro- include MET amplification (1875). MYCN ampl ification [2916}.
cytoma , and patients with such tumours have typical median and chromosome 14q loss 13387,596 ,597). One study found
OS in the range of 5-10 years 13346). that activating point mutations In PIK3R1 were associated with
IDH-mutant astrocytomas with microvascular proliferation, shorter OS in univariate analysis (122) . Evidence suggests that
necrosis, or CDKN2A and/or CDKN2B homozygous deletion a higher global copy-number variation load also correlates with
(or any combination of these features) correspond to CNS WHO worse prognosis (122,2122 ,2123.2741}.
Reifenberger G Snuderl ~'1
Oligodendroglioma , !DH-mutant Ca1rncross JG Suva ML
F1garella-Branger D Van Den Beni M J
and 1p/19q-codeleted Hartmann C Yip S
Kros JM Yokoo H
Louis ON

Definition Subtype(s)
Oligodendrogl1oma , !DH-mutant and 1p/19q-codeleted, is a dif- Oligodendroglioma , JOH -mut ant and 1p/19q-codeleted , err;
fusely infiltrating glioma with IDH1 or /OH2mutation and codele- WHO grade 2; oligodendroglioma , JOH -mutant and 1p/19ri.
tion of chromosome arms 1p and 19q (CNS WHO grade 2 or 3). codeleted, CNS WHO grade 3

ICD-0 cod ing Localization


9450/3 Oligodendroglioma , JOH-mutant and 1p/19q-codeleted, Among 5542 histologically defined oligodendrogl iomas regis-
grade 2 tered in the Central Brain Tumor Registry of the United States
9451 /3 Oligodendroglioma, JOH-mutant and 1p/19q-codeleted, (CBTRUS) database , 59% were located in the frontal lobe, w,,
grade 3 In the temporal lobe , 10% in the parietal lobe, and 1% 1n the
occipital lobe {498). Among 470 genetically defined CNS WHG
ICD-11 coding grade 3 oligodendrogl iomas of the French national POLA ne _
2AOO .OY & XH7K31 Other specified gliomas of brain & Oligo- work , 62% were frontal tumours , 16% were temporal , 15% were
dendroglioma, JOH -mutant and 1p/19q-codeleted parietal , and 6% were occipital (2542) . Other studies have alsc
shown a clear predilection for the frontal lobes in !DH-mutant
Related terminology and 1p/19q-codeleted oligodendrogliomas {2970 ,1143.34421
No recommended: anaplastic oligodendroglioma, !DH-mutant Less common locations include the posterior fo ssa. basa
and 1p/19q-codeleted. ganglia , and brainstem . Exceptional cases of !DH-mutant and
1p/19q-codeleted oligodendroglioma show widespread 1ntrac-
erebral dissemination corresponding to a gliomatos1s cerebr 1

pattern {1298) . Leptomeningeal spread is occasionally seer


in patients with JOH -mutant and 1p/19q-codeleted oligoden -
droglioma, in particular at recurrence {106). Primary leptome-
ningeal manifestation of IDH-mutant and 1p/19q-codeleted
oligodendroglioma has also been reported {183) . Rare cases
of intramedullary spinal oligodendroglioma are on record
but data on genotype are usually lacking {970 ,1264}. Rarely
patients may present with multifocal tumours {1036) . Individual
cases of morphologically defined oligodendrogliomas (not
genetically characterized) that developed from ovarian terato-
mas have been reported {3247).

Clinical features
Seizures are the presenting symptom in approximately two thirds
of patients with !DH-mutant and 1p/19q-codeleted oligodendro-
gl1oma (3589,401 ). Additional common initial symptoms include
headache, other signs of increased intracranial pressure, focal
neurological deficits , and cognitive changes . These signs and
symptoms are nonspecific and depend on the tumour's location
and speed of growth . With advanced imaging becoming more
widely available for symptom screening , incidental diagnosis 1s
more frequently reported , accounting for 10% of cases in one
study j3442J .

Imaging
Fig. 2.15 Ol1godendroglioma, IDH-mutant and 1p/19q-codeleted, CNS WHO grade JOH-mutant and 1p/19q-codeleted oligodendrogliomas usu·
2 A predominantly left frontal low-grade oligodendrogl1oma A There is involvement ally appear on CT as hypodense or isodense mass lesions that
of the corpus callosum and mass ettect, CT without intravenous contrast shows the
are typically located in the cortex and subcortical white matter
presence of a calc1f1cation in a hypodense region. B Involvement of the corpus cal-
losum and mass effect on T2-we1ghted MRI. C The tumour produces a hyperintense (2971 ). Calcifications are commonly seen , but they are not d1ag·
lesion on FLAIR MRI , with cortical involvement, diffuse borders, and signal hetero- nost1c ; some tumours show intratumoural haemorrhages and/
geneity D There 1s absence of contrast enhancement after Intravenous gadolinium or areas of cystic degeneration {2971 ). MRI typically shows a
administration on T1-we1ghted MRI. T1 -hypointense and T2-hyperintense mass with indistinct tumour
margins . Signal 1ntensit1es on T"l -we1ghted and T2-weighted tumours include a considerable subset of gltomas without IDH
MRI are often heterogeneous Gadolinium contrast enhance- mutation and 1p/'19q codeletion (928 ,3100,794) .
ment can be detected in < 20% of CNS WHO grade 2 oligo-
dendrogliomas, but it is present in > 70% of CNS WHO grade 3 Incidence
ol1godendrogltomas , where it is associated with microvascular The reported incidence rate (cases per 100 000 person-years) of
proliferation and less favourable prognosis [1600,3061 ,1221 , h1stologlcally diagnosed ollgodendrogliomas ranges from 0.10 in
2739) . IDH-mutant and 1p/19q-codeleted ollgodendrogliomas the Republic of Korea (1826) to 0.50 in France 1677]; rn the USA.
showed higher microvascularity (higher rCBV) and higher vas- the incidence rate is 0.23 /2344}. For histologically diagnosed
cular heterogeneity than IDH-mutant diffuse astrocytomas of CNS WHO grade 3 oligodendrogliomas, the incidence rate is 0.06
corresponding grade 11810). Magnetic resonance spectros- in the Republic of Korea 11826). 0.11 in the USA 12344). and 0.39
copy and rad1omics can identify differences in certain features in France 16771 Thus, 0.9% of all brain tumours in the USA are
between 1p/19q-codeleted and 1p/19q-intact low-grade di f- CNS WHO grade 2 oligodendrogliomas and 0.4% are CNS WHO
luse gliomas. but these techniques have limited sensitivity and grade 3 oligodendrogliomas (2344). Approximately one third of
spec1f1c1ty (-80% in validation series) and cannot yet replace all oligodendroglial tumours correspond to CNS WHO grade 3
molecular diagnostics 1387,903,3356,3279). Demonstration [2344). A decrease 1n the incidence of oligodendrogliomas from
of elevated 2-hydroxyglutarate levels by magnetic resonance 2000 to 2013 has been reported, a finding probably related to
spectroscopy is a new means of non-invasively detecting IDH- changes in diagnostic criteria over time [15) .
mutant gliomas (Including oligodendrogliomas), but it remains
technically challenging (580) . PET imaging may allow the dis- Age and sex distribution
tinction between CNS WHO grade 2 and 3 !DH-mutant gliomas, Oligodendrogliomas manifest preferentially in adults, with a
but reported series tend to be small and unvalidated 12299). median age at diagnosis of 43 years reported in the population-
based CBTRUS dataset for patients with histologically defined
Spread CNS WHO grade 2 oligodendroglioma and 50 years for those
IDH -mutant and 1p/19q-codeleted oligodendrogliomas char- with CNS WHO grade 3 oligodendroglioma {2344) . The median
acteristically extend into adjacent brain in a diffuse manner. ages were comparable for patients with IDH-mutant and 1p/19q-
Like other diffuse gliomas, they occasionally have a gliomatosis codeleted oligodendrogliomas: 41 years for patients with CNS
cerebri pattern 11298). In late-stage disease especially, distant WHO grade 2 tumours and 47 years for patients with CNS WHO
leptomeningeal spread may occur in some patients {106}. Rare grade 3 tumours {1246}. Overall, histolog1cally defined CNS
cases of extracranial metastases of oligodendrogliomas, mostly WHO grade 3 oligodendroglioma shows a slight male predomi-
CNS WHO grade 3, have been reported (2082,2945,420). At nance, with an M:F ratio of 1.2:1 reported among 5476 patients
times , patients with progressive tumours without treatment {2344}. CNS WHO grade 3 oligodendroglloma is more com-
options may show slow clinical deterioration despite the pres- mon In White populations than in Black populations, with an
ence of large enhancing lesions. incidence ratio of 2.3:1 (2344). Oligodendrogliomas are rare in
children, and few data are available on IDH-mutant and 1p/19q-
Epidemiology codeleted ollgodendrogliomas in this population . In one study,
The following paragraphs mostly refer to epidemiological data 3 (14%) of 22 tumours with the typical morphological character-
based on histological tumour classification, because popula- istics of oligodendroglioma demonstrated IDH1 p.R132H muta-
tion-based data on molecularly defined oligodendrogliomas are tion and 1p/19q codeletion (2703) . These 3 patients were aged
not yet available. Thus , the available information must be inter- 16-19 years . indicating that IDH-mutant and 1p/19q-codeleted
preted with caution as histologically defined oligodendroglial oligodendrogliomas are rare in children .

Flg.2.16 Oligodendrogl1oma , IDH -mutant and 1p/19q-codeleted, CNS WHO grade 3. Neurolmaging teatures . T1-hypointense lesion with focal contrast enhancement after
gadolinium administration (+Gd). T2-FLAIR shows the extent of the lesion, and FET PET demonstrates increased metabolic activity

Gl1u111JS 0110'1Purl nal tun1ours and 'l '.:lu ro11al tumours 29


Other etiological factors
The potential role of vi ral infections in the etiology of IDH -mut<lII'
• Cycling
and 1p/19q-codeleted oli godendroglioma has been debat ,
Several studies have reported the detection of CMV in gliorr. ~
inc luding oli godendrogliomas {270 ,2831 l. However, other st1/J.
ies have concl uded that CMV is not present in gliomas 1222
Similarly, there have been contrad ictory fin dings reported f·:r '
members of the polyomavi rus fam ily (BK vi rus , J C vi rus. SV4(
{726 ,2651 ,2712}. Whole-genome and RN A sequencing , wr.1C"
provided increased sensitivity and specificity for detecting w~
Astro-llke Oligo-like
genomes and transcripts , revealed on ly a low-percentage assr,
Lineage score ciation between HPV and/or HBV and low-grade gliomas includ-
ing oligodendrogliomas 13054}. It was also determined t ;,•
Fig. 2.17 Oligodendroglioma, IDH-mutant and 1p/19q-codeleted. Single-cell RNA- previous find ings of CMV in gliomas were probably a resul r,
sequencing analysis of human oligodendroglioma. Inferred developmental hierarchy laboratory contam ination . Dysregulation of the immune syster
based on genome-wide expression programmes, showing that oligodendroglioma including immunodeficiency due to HIV infection , posttransplar·
cells can recapitulate stem-like states or more differentiated states along the ollgoden-
immunosuppression therapy, or demyelinati ng disease, ha~
droglial or astrocytic lineages. Cycling cells (marked in red) are enriched in stem-like
states (3080,3202). been associated with rare cases of oligodendroglioma 1640
3041,2835). However, epidemiological data do not indicate ar
increased incidence of gliomas in patients with autoimmuns
Etiology disease {1288). Rat models have shown that nitrosoureas (e g
Genetic susceptibility ethylnitrosourea and methylnitrosourea) are chem ical carc1nc-
The eti ology of IDH-mutant and 1p/19q-codeleted oligoden- gens that may induce CNS tumours , including gliomas with ar
drogl ioma is unclear. Most tumours develop sporadically, in oligodendroglial phenotype {283). However, cancer studies ,.,
the absence of documented familial clustering or a hereditary humans are not available for these compounds.
cancer pred isposition syndrome . However, both familial and
sporadic gliomas frequently display shared genomic land- Pathogenesis
scapes , and common core pathways might be targeted by Cell of origin
both germline and somatic alterations (1434}. Earlier studies The cell (or cells) of origin of !DH-mutant and 1p/19q-codeletec
identified SNPs in the BICRA (GLTSCR1) and ERCC2 genes oligodendroglioma remains unknown . Morphology and single-
as well as the GSTT1 null genotype with increased risk of oli- cell RNA-sequencing analysis of human tumours supports
godendroglioma (3521 ,1587}. Germline mutations of POT1, a the notion that oligodendrogliomas are composed of a mix·
shelter in complex gene, have been associated with familial ture of malignant cell types that recapitulate oligodendroghal
oligodendroglioma (176} . Cases of familial oligodendroglioma and astrocytic lineages, as well as neural precursor-like cells
with 1p/19q codeletion have been reported (944,2339}. Given {3202}. Experimental transformation of immortalized human ghai
that pathological production of 2-hydroxyglutarate, resulting cells with IDH1 p.R132H reprogrammes their cellular lineage
from somatic mutations in IDH1 or IDH2, is found in all oligo- and favours the emergence of a neural precursor state {32391
dendrogliomas and IDH-mutant astrocytomas , it is of interest Experiments in transgenic mice indicate that gliomas with oh-
that variants (particularly rs5839764) in or near the 02HGDH godendroglial histology may originate from different cell types
gene , wh ich codes for D-2-hydroxyglutarate dehydrogenase, in the CNS, including neural precursor cells, astrocytes, anc
showed genome-wide association with IDH-mutant gliomas oligodendroglial precursor cells {3619) . An oligodendroglioma-
{823} . The same study identified rs111976262, located near like phenotype is commonly found in transgenic brain tumours.
the FAM20C gene, as showing genome-wide association with despite such tumours showing a variety of targeted cell types
IDH-mutant, TERT promoter-mutant, and 1p/19q-codeleted and oncogenic events {758,3412). Studies have suggested
oligodendrogliomas . that oligodendrogliomas probably originate from oligodendro-
1
Gliomas have been reported in specific hereditary cancer glial precursor cells {2477,3065). Oligodendroglial precursor
syndromes including germline BRCA 1 mutations, constitutional cells have also been suggested as the cell of origin in other
mismatch repair deficiency syndrome, Lynch syndrome (also classes of gliomas and may give rise to either oligodendroglial
known as hereditary non-polyposis colorectal cancer), and or astrocytic phenotypes in gliomas, depending on the genes
hereditary retinoblastoma , yet oligodendrogliomas are uncom- driving transformation {1915 ,1901). Thus, the interplay betweeo
mon {1484 ,3154 ,2071 ,1277,22} . Patients with the enchondro- oncogenic events and the cell(s) of origin plays a critical role 1n
matosis syndromes Oiiier disease and Maffucci syndrome, determining the resulting glioma phenotype.
which are associated with somatic (or postzygotic) IDH mosai-
cism , present with multiple benign cartilaginous tumours (95}. Genetic profile
A retrospective cohort study showed that those patients may The entity-defining alterations in oligodendrogliomas are m1·
develop gliomas with an anatomical presentation and a grad- sense mutations affecting IDH1 codon 132 or IDH2 codon 172
ing distribution similar to those of gliomas in non-syndromic combined with whole-arm deletions of 1p and 19q. More than
patients , but they are typically younger and more often have 90% of IDH mutations in oligodendrogliomas correspond
multicentric lesions (320] . However, none of the gliomas in this the canonical IDH1 p.R132H mutation ; the remaining tumours
enchondromatosis cohort harboured 1p/19q codeletion . carry non-canonical mutations , with a higher proportion ol

30 Gl1omas. ~J l i oneuronal tumourt,, and 11 urona l tumours


.
• ••

• ••

··'.-
•••


..
•' . .
• •
• ,,

•• .
••
\
A .. • • •, •' ' .. • • ,'
Flg.2.18 Oligodendroglioma, !DH-mutant and 1p/19q-codeleted, CNS WHO grade 2. A Oligodendrogliomas often infiltrate the cortex. and individual tumour cells congregate
around neuronal cell bodies. B Oligodendroghoma cells are sometimes embedded within a light-blue mucinous matrix.

IOH2 mutations in oligodendrogliomas than in astrocytomas oligodendrogliomas \261,3537) , with large-scale sequencing
(see also Astrocytoma, /OH-mutant, p. 19) {1246.452 ,824 , studies reporting CIC mutations in as many as 70% of oligo-
126). The 1p/19q codeletion has been cytogenetically linked dendrogliomas (452 ,824}. CIC is a constitutive transcriptional
to an unbalanced translocation between chromosomes 1 and repressor of genes essential in development, cellular growth ,
19 that results in loss of the der(1 ;19)(p10;q10) chromosome, and metabolism that is relieved by receptor tyrosine kinase sig-
causing whole-arm deletions of 1p and 19q, and retention of nalling (154,3471,38) , and it has been associated with various
the der[t(1 ;19)(q10;p10)] chromosome (1165,1465} . Incomplete/ features of neoplastic behaviour (2940 ,2317) . CIC mutations in
partial deletions on either chromosome arm are not compatible oligodendrogliomas are hemizygous and include almost equal
with the diagnosis of !DH-mutant and 1p/19q-codeleted oligo- proportions of nonsense or truncating mutations and recurrent
dendroglioma, but they have been detected in a proportion of missense mutations . The latter are preferentially found in the
IDH-wildtype glioblastomas {3339} . HMG-box DNA-binding domain in exon 5 and the C1 motif in
The vast majority of IDH-mutant and 1p/19q-codeleted oli- exon 20. They appear to be unique to oligodendroglioma and
godendrogliomas carry TERT promoter hotspot mutations (134, not present in other CIC-mutant tumour types 11823,2317,
1614,1678). However, !DH-mutant and 1p/19q-codeleted oligo- 2948) . This suggests phenotypic uniqueness of these mis-
dendrogliomas arising in teenagers often lack TERT promoter sense CIC mutations in oligodendrogliomas . and that these
mutation \1833) . When present, TERT promoter mutation is mutations act cooperatively with IDH mutations to contribute to
assumed to be an early (i .e. clonal) event in oligodendroglioma the pathological upregulatlon of 2-hydroxyglutarate production
development \3082,881), which remains stable during tumour {574) and activation of tile MAPK signalling pathway {1823.960) .
progression and at recurrence {44). Mechanistically, the TERT Spatial and temporal profiling of oligodendrogliomas . which
promoter mutations generate de nova ETS transcription factor have a low mutation burden, has also confirmed the presence
binding sites \1343} , which results in transcriptional upregula- of clones bearing unique CIC mutations. suggesting the pres-
t1on of TERT expression , thereby driving telomere stabilization , ence of selective pressures to escape normal CIC regulatory
cellular immortalization, and proliferation 11214 ). control {3082,3202,208}. CIC truncating mutations most likely
Mutations of CIC (the human orthologue of the Drosophila disrupt protein-protein interaction with binding partners includ-
melanogaster capicua gene), located in chromosome band ing ATXN ·1L, which appears to result in reciprocal phenotypic
19q13.2, are also frequent 1n IDH-mutant and 1p/19q-codeleted alterations (3366,3469,3471 }.

Gl1 ornas . g l1 one urona1 tumuu rs , and neL..ronal tumours 31


Approximately 20- 30% of !DH-mutant and 1p/19q-codeleted survival 1886,64). In line with these findings , homozygr,.
oligodendrogliomas harbour somatic mutations of FUBP 1. CDKN2A deletion was indicative of short survival in a prospe;
located at chromosome 1p31 .1, a region with consiste nt loss tive cohort study of patients with CNS WHO grade 3 IDH-muta"
of heterozygos1ty in these tumours {261 .2779). FU BP1 is a tran- an d 1p/19-codeleted oligodendroglioma (1231. Other alterat 1G,,·
scriptional regulator essential for normal stem cell self-renewal associated with tumou r progression and/or shorter surwr
{2589,1390). It has recently been identified as a pleiotro pic include PIK3CA mutation 13140,380). TCF12 mutation 1177Bc:
regulator of alternative splicing of tumour suppressor genes and genetic aberrations causing increased MYC s1gnallir;
and oncogenes {842) . The combined loss of CIC and FUBP1 11 537) . Whereas IDH mutation . 1p/19q codeletion . and TEFi.
protein expression . as a surrogate marker of CIC and FUBP1 promoter muta tion are clonal alterations 1n oligodendrogliom- ~
nonsense or truncating mutations . has been associated wi th mutations in CIC, FUBP1, TCF1 2, and other genes may be su
a shorter time to recurrence in patients with 1p/1 9q -c odeleted clonal and thu s associ ated with tumour progression (3082,88i
oligodendroglioma {508) .
Approximately 15% of oligodendrog liomas carry mutations In Epigenetic changes
NOTCH1, and less commonly in other NOTC H pathway genes IDH-mutant and 1p/19q-codeleted oli godendrogliomas sho:,
{452.3082) . NOTCH1 mutation was linked to shorter survival in concurrent hypermethylation of multip le CpG islands, ccr
one study (122}. Other less common ly mutated genes include responding to the glioma CpG Island methylator pheno yP~
epigenetic regulator genes such as SETD2 (and other histone (G-CIMP)l2287 }. Th is phenomenon has been closely linked ,
methyltransferase genes), PIK3CA, an d genes encod ing com - IDH mutation causing increased levels of 2-hydroxyglutara:e
ponents of the SWl/S NF chromatin remodelling complex {452 , which functions as a competitive inhibitor of a-ketoglutarate -
3082} . dependent dioxygenases . includ ing histone demethylasi::
and the TET family of 5-methylcytosine hydroxylases (194~
Genetic alterations associated with tumour progression 3501 }. This in turn leads to increased histone methylation ar.~
The number of broad c opy-nu mber aberrations increases from G-CIMP {2287,32401 . DNA methylation profiles of IDH-mutar·
CNS WHO grade 2 to CNS WHO grade 3 oligodendrogliomas and 1p/19q-codeleted oligodendrogliomas differ from th ose ~ ·
{122). Deletions on 9p involving the CDKN2A and/or CDKN2B IDH-mutant but 1p/19q-intact astrocytomas , and they cant·
locus have been associated with CNS WHO grade 3 (2640, used for diagnostic purposes {460J . G-CIMP may correlate wit~
2106) . contrast enhancement on MRI {2658 ,64}, and shorter epigenetic silencing of multiple genes in oligodendrogliomas
including genes on 1p and 19q, as well as genes on other chro- Mineralization and other degenerative features
mosomes , such as the tumour suppressors COKN2A , CDKN2B, Microcalcifications are frequent , found with in the tumour itself or
and RB1 (2673) . MGMT promoter methylation is detectabl e in in the invaded brain . Calcifications we re recorded in 71 (45%)
the majority of oligodendrogl1omas 12141). At the mRNA level, of 157 CNS WHO grade 3 !DH-mutant and 1p/19q-codeleted
IDH-mutant and 1p/ 19q-codeleted oligodendrogliomas typi- oligodendrogliomas , {928}. Mineralization along blood ves-
cally show a proneural glioblastoma- like gene-expression sig- sels typically takes the form of small , punctate calcifications ,
nature {798,3418). whereas microcalcificatlons in the brain (called calcospherites)
ten d to be larger, with an irre gular and sometimes laminated
Macroscopic appearance appearance. However, this featu re is not specific for oligoden -
Oligodendroglioma typically appears macrosco pically as a droglioma, and because of incom plete tumour sampling, it is
relatively well-defined , soft, greyish -pink mass located in the sometimes not found histologically even when clea rly demon-
cortex and white matter. with blurrin g of the grey matter- wh ite strated on CT. Areas characterized by extracellular mucin depo-
matter boundary. Local invasion into the overlying leptomenin- sition and/or microcyst forma tion are frequ ent. Rare tumours are
ges may be seen . Calcification is frequent and may impart a characterized by marked desmoplasia {1470}.
gritty texture . Occasionally, densely calcified areas may occur
as lntratumoural stones. Zones of cystic degeneration , as well Vasculature
as intratumoural haemorrhages , are common . Rare cases with Oligodendrogliomas typ ically show a dense network of branch-
extensive mucoid degeneration look gelatinous. Areas of necro- ing capillaries resemb ling chicken wire . In some cases , the cap-
sis may be discerni ble in CNS WHO grade 3 tumours. illary stroma tends to subd ivide the tumour into lobules. In CNS
WHO grade 3 tumours , focal or dispersed pathological micro-
Hlstopathology vascular proliferation is frequent. Oligodendrogli omas have a
Cellular composition tendency to develop intratumoural haemorrhages.
Classic oligodendroglioma cells have uniformly round nuclei that
are slightly larger than those of normal oligodendrocytes and Growth pattern
show an increase in chromatin density or a del icate salt-and- Oligodendrogliomas grow diffusely in the cortex and wh ite mat-
pepper pattern . A disti nct nuclear membrane is often apparent. ter; however, some tumours feature distinct nodules of higher
In formalin-fixed , paraffin-embedded tissue, tumour cells often cellularity against a background of diffuse infiltration . Occa-
appear as rounded cells with well-defined cell membranes and sional tumours show a gliomatosis cerebri- like pattern involving
clear cytoplasm around the central spherical nucleus. This cre- more than two cerebral lobes {1298} . Within the cortex, tumour
ates the typical honeycomb or fried-egg appearance, which, cells often form secondary structures such as perineuronal sat-
although artefactual, is a helpful diagnostic feature. This artefact ellitosis, perivascular aggregates, and subpial accumulations .
is not seen in smear preparations or frozen sections, and it may Circumscribed leptomeningeal infiltration may Induce a desmo-
also be absent in rapidly fixed tissue and in formalin-fixed , par- plastic reaction . Oligodendrogliomas can have a rare spongio-
affin-embedded sections made from frozen material. Reactive blastic growth pattern consisting of parallel rows of tumour cells
astrocytes are scattered throughout oligodendrogliomas and with somewhat elongated nuclei forming rhythm ic palisades.
are particularly prominent at the tumour borders . Oligodendro- Occasionally, perivascular pseudorosettes are seen , although
gliomas may contain tumour cells that look like small gemisto- some of these are a result of perivascular neuropil formation
cytes with a rounded belly of eccentric cytoplasm that is positive within foci of neurocytic differentiation {2471 }. These patterns
for GFAP, which are termed "minigemistocytes" or "microgemi- are generally present only focally.
stocytes". Gliofibrillary oligodendrocytes are typical-looking
oligodendroglioma cells with a thin perinuclear rim of positivity Proliferation
for GFAP (1295). Gliofibrillary oligodendrocytes and minigemis- Mitotic activity Is low or absent in CNS WHO grade 2 oligoden-
tocytes are more commonly seen in CNS WHO grade 3 tumours . drogliomas, but it is usually prominent in CNS WHO grade 3
GFAP-negative mucocytes or even signet-ring cells are occa- tumours. Accordingly, the Ki-67 (MIB1) proliferation Index is
sionally present, with individual cases reported to consist largely usually low (< 5%) in CNS WHO grade 2 oligodendrogliomas
of signet- ring cells [1743). Eosinophilic granular cells occur in and elevated in CNS WHO grade 3 oligodendrogliomas, being
some ol1godendrogl iomas {3114}. Rare cases with neurocytic or generally > 10% in the large French national POLA cohort of
ganglioglloma-like differentiation have also been reported {2466, CNS WHO grade 3 tumours \928 ,929,2546). However, a defini-
2471) . Occasional CNS WHO grade 3 oligodendrogllomas fea- tive Ki-67 (MIB1) cut-off value has not been established due
ture multinucleated giant cells {1301), and rare cases contain to marked variability in staining results between institutions
sarcomatous areas {1310,2699}. The presence of these various and non-uniform counting approaches . One study reported a
cellular phenotypes does not preclude an ollgodendroglioma Ki-67 Index of ~ 15% as an independent marker of shorter sur-
diagnosis if the tumour is !DH-mutant and 1p/19q-codeleted. vival in patients with !DH-mutant and 1p/19q-codeleted CNS
Tumour cells with fibrillary or gemistocytlc astrocytic morphol- WHO grade 3 oligodendrogliomas (2546}. A mitotic count of
ogy are also compati ble with this diagnosis when IDH mutation ~ 5 mitoses/mm was also associated with shorter survival but
2

and 1p/19q codeletion are present. Thus, irrespective of oligo- only on univariate, not multivariate, analysis \2546}. Another
dendroglial, oligoastrocytic, astrocytic , or ambiguous features study showed that mitotic count was not associated with out-
on histology, detection of combined IDH mutation and 1p/19q come in patients with !DH-mutant and 1p/19q-codeleted oli-
codelet1on indicates an !DH -mutant and 1p/19q-codeleted oli- godendrogliomas 12318}. Other proliferation markers , such as
godendroglioma 1452,3082,3418 }. PCNA 12647). TOP2A 11717], MCM2 (3422). and MCM6 (2546},

Gliomas , glloneuronal tumours , and neuronal tumours 33


~--~-- .J
m no ::itoche 1 1cal features of IDH-mutant and 1p/19q-codeleted oligodendroglioma. A MAP2. B OLIG2. C GFAP. D Gliofibrillary oligodendrocytes (GFAP s

also correlate with CNS WHO grade and/or survival but do not palisading . CNS WHO grade 3 oligodendrogliomas usually
proJ1de clear advantages over Ki-67 (MI B1 ). show several of these features . However, the individual impact f
of each feature is unclear, in particular because most prognos-
Grading tic studies have not previously been confined to IDH-mutarn ;
Ol1 gooendrogl1omas comprise a continuous spect rum of and 1p/19q-codeleted tumours. Microvascular proliferation and •
Jmo rs ranging from well-differentiated , slow-growing neo-
piasrns to fr ankly malignant tumours with rapid growth . In prior
brisk mitotic activity, defined as ~ 2.5 mitoses/mm 2 (equating t
to ~ 6 mitoses/ 10 HPF of 0.55 mm in diameter and 0.24 ml112
ed. ions of rhe WHO c lassification of CN S tumours, two grades in area) have been reported as indicators of short survival in a
-11ere distin guished: oligodendroglioma, CNS WHO grade 2, study of histologically defined oligodendrogliomas {1096}. Other ~
a d oligodendroglioma, CNS WHO grade 3. CNS WHO grade studies of 1p/19q-codeleted CNS WHO grade 3 oligodendro-
retained prognostic significance in patients with !DH-mutant gliomas suggested that microvascular proliferation and micro-
and 1p/19q-codeleted oligodendrogliomas {597}, but the cri te- vascular proliferation with necrosis are linked to shorter survival
ria for distinction between grades were not well defined . Histo- than is elevated mitotic activity of ~ 2.5 mitoses/mm 2 (equating
logical features that have been linked to higher grade are high to ~ 6 mitoses/ 10 HPF of 0.55 mm in diameter and 0.24 mnr
cellulanty, marked cytological atypia, brisk mitotic activity, path- in area) without microvascular proliferation and necrosis (928,
ological microvascular proliferation, and necrosis with or without 929). However, data defining a clear cut-off point for a mitotic
co..,11· :~a~ o sr ,...,gJ S"'es C S · 0 g ade 2 f o CNS WHO
graae 3 cl DH-rriutarit ard 10 9o-ccde e ed o goaend g o-
mas are . . ta a1able e\.e ,reless. de'e on o ra e mi oses 1
a esec: n spec r'T'>en s '10t suf c e!"l or d agno_ g C S HO
graoe 3 IOY -rr1.. an· and 1o 9q-code e•ed o g e droghoma
In boroer ,..,e ~ases pro 11 'e at o ar' ers h e -67 ( IB 1) and
at·er on :c c1 'l•ca and 11eu oraa olog.cal ea ures (e.g rapid
s rnp.o,.,..,at c Q'O'. -n and co rasi enha cement) may provide
elo u1 aco t o-.a1 1n.orrnai.on Of""'l zygous dele"on 1nvolv1ng
the car< -24 a :: or CDK.N2B locus IS OU d m a s all subse
(< ~oc-o) o' C S .~HO grade 3 oltgodendrog1iomas but not tn
CNS · HO grade 2 ol godendrog' omas. and 1 has bee Jinked
to red ~ea s0 1
al 1ndepe den of rr11crovascular prolifera ion
wi· or •, o · ecros1s {123}. T erefore. CDKN2A homozy-
gous oe'et er. ay serve as a molecular ma er of C S WHO
graoe 3 " IDH- u a ·a d p/19q-codele ed ohgodendroglio-
mas A1thoug assess ent o his marker may not be routinely Fig. 2.23 Ohgodendroghoma. IDH-mutant and 1
req u1rea 1n UIT'O s t at ca his olog1cally be unequivocally lntraoperative smear section of an olioodendrc~>m a
ass1g-iec ,o el ner C S WHO grade 2 or CNS WHO grade 3, lary bacJi.ground aSSOClation with deJteate va
test1rg o COKN2A homozygous deletion may be he Ipf ul, for
exampie 1 urio r samples Wlt borderline histological features been associated with 1p/ 19q codeletion in IOH-mutant ghomas
(i.e. nen present. a COKN2A homozygous dele ion indicates a (933.904 }. but it cannot subst1 u e for 1p/ 19Q tes 1ng (2440}.
CNS HO graae 3 tumour)
Differential diagnosis
Im unoohenotype IDH-mutant and 1p/1 9q-codeleted oligodendroghomas may
Most 0!1gooendrogliomas demonslra e immunoreac iv1ty with h1stologically mimic various other lesions. Macrophage-nc
the an· b ay against IDH1 p .R132H {465J. which fac1£itates the lesions such as those c haracteris c of demyelinating diseases
d r erenual diagnosis versus other clear cell tumours as well or resulting from cerebral infarction are readily distinguished
as non-neoplastic and reactive lesions (461.462}. !OH-mutant by immunostaining for macrophage markers and lack of IDH
and p/1 9q-codeleted oligodendrogliomas retam nuclear mutation. The relative increase of oligodendrocytes sometimes
expression of ATRX {1 916 .2657 1and typically lack widespread seen in partial lobeciomy specimens performed for intractable
nuclear p53 starn1ng . consistent with the near exclusivity of seizures also lack !DH mutation. IDH-mutani astrocytomas lack
ATRX a d TP53 mutation versus 1p/ 19q codeletion in IDH- 1p/19q codeletion and show frequent nuclear p53 1mmuno-
mu.an ghomas {452.3082}. Ohgodendrogliomas are immu- stainrng and loss of nuclear ATRX . In fact. loss of nuclear ATRX
noposi 1ve for MAP2. S100 . and C057 (LEU7) (298,2198 , is sufficient to diagnose an IDH-mutant astrocytoma without
2637 }: however, these markers are also positive in astrocytic additional testing for 1p/ 19q codeletion 11 935}. TERT promoter
gliomas Similarly. the oligodendrocyte lineage transcription mutations are common in !DH-mutant and 1p/19q-codeleted
factors OLIG1 . OLIG2, and SOX10 are expressed in oligoden- oligodendrog liomas. although rare cases have been reported
drogl1 ornas but also in astrocyt1c gliomas {1 92,18921. GFAP is to lack TERT promoter mutation. and some IDH-mutant but
de ectable 1n interming led reactive astrocytes but may also 1p/1 9q-intact astrocytomas may carry TEAT promoter muta-
s1ain neoplastic ce ll s such as minigem1stocytes and gliofi- tions {134,1678,1780.3082}. Other morphological mimics,
brillary ol1 godendrocytes (1295,26371. Antigens expressed like neurocytoma, liponeurocytoma, and dysembryoplastic
by normal oli godendrocytes. including myelin basic protein neuroepithelial tumour, can be ruled out by their lack of IDH
(MBP) . myelin proieol1 p1d protein (PLP), myelin-associated gly- mutation. Ependymomas containing clear cells differ from oligo-
coprotein (MAG), galactolipids (e.g . galactocerebroside and dendrogliomas by their perivascular pseudorosettes and dot-
galac osu lfat1de), certain gangliosides. and several enzymes like or ring-shaped EMA immunoreactivity. as well as a lack of
(e.g . CAii [carbonic anhydrase C], CNP. glycerol-3-phosphate IOH mutation and frequent ZFTA (C11orf95) fusions. Clear cell
dehydrogenase. and LOH) are not diagnostically useful mark- meningioma can be distinguished by EMA and desmoplakin
ers for oligodendrogliomas {2198,3073,2199}. Synaptophysin positivity, IDH-wildtype status. and loss of nuclear SMARCE1
immunoreact1v1ty of residual neuropil between the tumour {3148}. Metastatic clear cell carcinomas differ from ohgoden-
cells 1s frequent and should not be mistaken for neuronal or drogliomas by their sharp tumour borders. cytokeratin and
neurocytic d1fferennat1on . However, oligodendrogliomas may EMA positivity, and lack of IDH mutation. Pilocytic astrocyto-
also contain neoplastic cells that express synaptophysin and/ mas with oligodendroglial features are IOH-wildtype and carry
or NeuN and neurofilaments (2466,24711. lmmunostaining for MAPK pathway gene alteratmns, in particular FGFR1 alterations
a-internex1n protein 1s frequent (7971 (e.g. in one study it was {2932}. However. rare cases of IDH-mutant and 1p/ 19q-code-
found in 88 5% of IDH-mutant and 1p/19q-codeleted CNS leted oligodendrogliomas with KIAA 1549::BRAF fusions have
WHO grade 3 oli godendrogllomas {928}). but it cannot be con- been reported (171,1628}. In children , diffuse gliomas with MYB.
sidered a surrogate marker for 1p/19q codeletion 1829}. Simi- MYBL1, FGFR1 , or BRAF alterations may have h1stolog 1cal fea-
larly, NOGO-A positivit y is common but not exclusive (2026}. tures of oligodendroglioma or oligoastrocytoma but are b1olog1-
Reduced nuclear expression of H3 p .K28me3 (K27me3) has cally distinct tumours {838}. The differential diagnosis of diffuse

Gltornas glioneuronal tumours ar d . e0ror3 '._.r"1Cur - 35


IDH -mutant astrocytoma or IDH -mutant and 1p/19q-codeleted
oligodendroglioma (452,2781,3082) . Thus, diffuse gliomas with
mixed or ambiguous histological features should be evaluated
for IDH mutation and loss of nuclear ATRX expression, as Well
as tor 1p/19q codeletion when nuclear ATRX is retained 11935).
Rare cases of dual-genotype oligoastrocytomas have been
reported . These are characterized by two distinct !DH-mutant
tumour cell populations: one showing astrocytoma-associated
alterations, such as ATRX loss and TP53 mutation, and the
other showing oligodendroglioma-associated 1p/19q codele-
tion 12587,204 ,1382,3444). The WHO Classification of Tumours
does not consider these tumours to be a distinct type or sub-
type of IDH-mutant diffuse glioma, but they may be tentatively
Fig. 2.24 Oligodendroglioma, IDH-mutant and 1p/19q-codeleted. CNS WHO grade 2.. classified as dual-genotype oligoastrocytoma NEC /1946).
FISH demonstrates 1p/19q codeletion (without polysomy). A The 1p signal is red and
the 1q signal is green. B The 19q signal is red and the 19p signal is green. Cytology
In cytological preparations , oligodendroglial tumour cells show
leptomeningeal glioneuronal tumour is facilitated by the clinical uniform round nuclei and well-delineated cytoplasm , with only a
presentation and the combination of MAPK pathway gene alter- modest degree of glial fibrillarity. The perinuclear haloes that are
ation (in particular K/AA1549::BRAFfusion) and 1p deletion (or typical of histological preparations are not appreciated in smear
1p/19q codeletion) but absence of IDH mutation {2702). Malig- specimens . In CNS WHO grade 3 oligodendrogliomas , intensely
nant small cell astrocytic tumours , including IDH-wildtype glio- eosinophilic cytoplasmic granules are occasionally noted . Reac-
blastomas and H3 G34-mutant diffuse hemispheric gliomas , tive astrocytes harbouring eosinophilic cytoplasm with multipolar
must be separated from highly cellular oligodendrogliomas by processes may also be present. In some cases , microcalcifica-
their IDH-wildtype status and specific alterations , including tions and prominent vasculature can be appreciated .
frequent EGFR amplification and chromosome 10 loss !1500,
2464), or mutations leading to H3 p.G35 (G34) variants . Diagnostic molecular pathology
Oligodendrogliomas are molecularly defined by IDH1 or IOH2
Oligodendroglioma NOS, oligoastrocytoma NOS, mutations and 1p/19q codeletion . Nearly all tumours have a
and oligoastrocytoma NEC TERT promoter mutation , lack ATRX mutation, and show pre-
Rare cases of diffuse gliomas with classic oligodendroglioma served nuclear ATRX expression . TP53 mutations are uncom-
histology in which molecular testing for combined IDH muta- mon {452,1999,1). Diagnosis of oligodendrogliomas requires
tion and 1p/19q codeletion failed (e.g . because of limited tissue demonstration of IDH mutation by IDH1 p.R132H immunohis-
availability, low tumour cell content, inconclusive test results , or tochemistry and/or sequencing of the IDH1 or IDH2 gene, as
other circumstances impeding molecular testing) or could not well as demonstration of 1p/19q codeletion by FISH, chromo-
be completed can be histologically classified as oligodendro- genic in situ hybridization, or molecular genetic testing . In the
glioma NOS {1946) and designated as CNS WHO grade 2 or absence of IDH1 p. R132H-positive immunohistochemistry,
3 depending on the presence or absence of histological fea- sequencing for less common mutations in IDH1(codon132) and
tures of anaplasia. lmmunohistochemical demonstration of IDH IDH2 (codon 172) should be performed . No particular method
mutation and retained nuclear positivity for ATRX may support for molecular testing for 1p/19q codeletion is required , but it is
the oligodendroglioma diagnosis. However. unless successfully recommended that 1p/19q assays be able to detect whole-arm
tested for 1p/19q codeletion , such tumours cannot be classi- chromosomal losses. Only complete losses of both chromosome
fied as oligodendroglioma, IDH-mutant and 1p/19q-codeleted. arms are diagnostic for oligodendroglioma, because partial or
lmmunohistochemical positivity for oligodendroglioma-asso- isolated loss of 1p or 19q may be present In some IDH-wildtype
ciated markers such as a-internexin {797,829) and NOGO-A glioblastomas and !DH-mutant astrocytomas . FISH probes in
{2026), reduced nuclear H3 p.K28me3 (K27me3) immunostain- commonly deleted regions or loss of only a few PCR probes in
ing {933 ,904), and immunohistochemical loss of nuclear CIC or loss-ot-heterozygosity analysis may not reflect whole-arm losses
FUBP1 expression {221,508) are not sufficient to substitute for and thus may lead to false positive results {1341). In addition , any
1p/19q codeletion testing . copy-number analysis requires sufficient tumour cell content.
The diagnosis of oligoastrocytoma NOS is reserved for dif- preferably > 30% , to avoid false negative results when assess-
fuse gliomas that are composed of a conspicuous mixture of ing 1p/19q codeletion . lmmunohistochemical detection of IDH1
two distinct neoplastic cell types morphologically resembling p.R132H expression and preserved nuclear ATRX expression,
tumour cells with either oligodendroglial or astrocytic features, without demonstration of 1p/19q codeletion, is not sufficient to
and in which testing for IDH mutation , nuclear ATRX expres- diagnose an !DH -mutant and 1p/19q-codeleted oligodendro-
sion , and 1p/19q codeletion failed or cou ld not be completed . glioma, even with classic histology. In !DH-mutant gliomas with
In such cases , tumour cells with oligodendroglial or astroglial preserved nuclear ATRX expression by immunohistochemistry,
features can be either diffusely intermingled or separated into 1p/19q analysis remains critical tor accurate molecular diagno-
distinct, biphasic areas. However, the diagnosis of oligoas- sis . Most !DH -mutant and 1p/19q-codeleted oligodendroglio-
trocytoma is discouraged because molecular analyses have mas carry TERT promoter mutations {134) ; however, detection
shown that these tumours carry a genetic prof lie typical of either of a TERT promoter mutation in an !OH -mutant glloma is not

36 Gl101ridS glir_111Luror1fil umuurs, ancJ r1eurona l tumours


sufficient for an oligodendroglioma diagnosis, because rare inclusion of gliomas without IDH mutation and 1p/19q codeletion .
cases are TERT-wildtype, including tumours In teenage patients Retrospective molecular stratification of older series confirmed
j1833) . TERTpromoter mutations are also observed in a subset that only subsets (30- 80%) of tumours corresponded to IDH-
of 1p/19q-intact !DH-mutant astrocytomas {2442,2277) . DNA mutant and 1p/19q-codeleted ollgodendrogliomas {928 ,3100 ,
methylation array analysis reveals a diagnostic molecular profile 794] . Overall , !DH-mutant and 1p/19q-codeleted oligodendro-
by combining the detection of an oligodendroglloma-associated gllomas are associated with favourable response to therapy and
methylation signature and 1p/19q codeletion {460,463). Copy- median survival times of> 10 years . For example , patients with
number analysis by FISH also provides information on polysomy CNS WHO grade 3 !DH-mutant and 1p/19q-codeleted oligo-
1 of 1q and 19p, which has been detected in subsets of 1p/19q- dendroglioma who participated in prospective clinical trials and
codeleted oligodendrogliomas of CNS WHO grades 2 and 3 and were treated with a combination of radiotherapy and procar-
is associated with shorter survival \537,2974,3436}. Homozy- bazine , lomustine, and vlncristine (PCV) chemotherapy showed
gous deletion of CDKN2A has been detected in a small propor- a median survival of ~ 14 years (3276). Oligodendrogliomas
, tion of CNS WHO grade 3 !DH-mutant and 1p/19q-codeleted generally recur locally but may show leptomeningeal spread.
1 oligodendrogliomas , but not in those of CNS WHO grade 2, and Malignant progression at recurrence is common, although it
it was reported to be an independent marker of shorter survival usually takes longer in oligodendroglioma than in !DH-mutant
1123}. astrocytoma {1440) .
Tumours that cannot be fully analysed for IDH mutation and
1p/19q codeletion but demonstrate classic histological features Clinical factors
of oligodendroglioma are classified as oligodendroglioma NOS Clinical factors associated with more favourable outcome
{1946) . Th is indicates that the tumour is a histologically classic include younger patient age at diagnosis, frontal lobe loca-
oligodendroglioma that will probably exhibit clinical behaviour tion, presentation with seizures, high postoperative Karnofsky
similar to that of an !DH-mutant and 1p/19q-codeleted oligo- score, and macroscopically complete surgical removal (3275}.
1 dendroglioma, but that it could not be molecularly analysed or Many of these factors, including age, are confirmed in studies
that its test results were inconclusive or uninformative {1946). on molecularly defined oligodendroglioma, but limited follow-up
Tumours that demonstrate oligodendroglial histology but lack remains an issue (2546,597) .
IDH mutation and 1p/19q codeletion should not be classified
as oligodendroglioma NOS but must be further evaluated to Imaging
exclude histological mimics, such as dysembryoplastic neu- The presence of contrast enhancement on imaging is indicative
roepitheiial tumour, clear cell ependymoma, neurocytoma, of worse outcome in IDH-mutant CNS WHO grade 2 and 3 glio-
polymorphous low-grade neuroepithelial tumour of the young, mas, Including oligodendrogliomas (1221,3061). An increased
and pilocytic astrocytoma, as well as molecularly distinct dif- growth rate on follow-up MRI has been associated with histo-
fuse gliomas that are characterized by BRAF, FGFR1, MYB, or logical features of anaplasia, including microvascular prolifera-
MYBL1alterations1838). tion and higher mitotic count, with contrast enhancement on
neuroimaglng, and with shorter progression-free survival (PFS)
1 Essential and desirable diagnostic criteria (2739).
See Box 2.02.
Surgery
Staging Greater extent of resection has been associated with longer
Not clinically relevant overall survival (OS) and PFS in patients with CNS WHO
grade 2 oligodendroglloma, but it did not prolong the time to
Prognosis and prediction malignant progression [2977). Studies using volumetric tumour
Survival data for histologically diagnosed tumours In older stud- assessment show that extensive resections are associated with
ies and population-based registries are confounded by the improved outcomes. However. leaving some tumour tissue
behind appears to have less impact on the survival of patients
with oligodendroglioma than on that of patients with !DH-mutant
Box2.02 Diagnostic criteria for oligodendroglioma, !DH-mutant and 1p/19q-codeleted
astrocytoma (3442,2418,759}. perhaps because of the higher
Essential: sensitivity of ollgodendrogliomas to radiotherapy and chemo-
A diffusely infiltrating glioma therapy.
ANO
IDH1 codon 132 or IDH2 oodon 172 mlssense mutatlon1 Histological features
AND Histological features that have been linked to worse progno-
sis include necrosis, high mitotic activity, increased cellularity,
Combined whole-arm deletions of 1p and 19q
nuclear atypia, cellular pleomorphism, and microvascular prolif-
Desirable: eration. However, the prognostic significance of each of these
DNA methylome profile of oligodendmglioma, IDH-mutant and 1p/19q-codeleted requires re-evaluation in patients with molecularly character-
Retained nuclear expression of ATAX ized tumours. In !DH-mutant and 1p/19q-codeleted CNS WHO
TERT promoter mutation grade 3 oligodendrogliomas, high mitotic count(~ 2.5 mitoses/
'IDH mutatJon analysis may not be required when DNA methylome profiling is mm 2 , equating to ~ 6 mitoses/10 HPF of 0.55 mm in diameter
performed and unequivocally assigns the tumour to the methylatlon class oligodendro- and 0.24 mm 2 in area) was linked to shorter PFS and OS in both
1
ghoma, IDH-mutant and 1p/19q-codeleted. univariate and multivariate analyses 1929). The presence of

Gl1omas , g l1or1 -"Uronal tumours, nd neuro11al tumours 37


Progression Free Survival correlated with shorter OS in univariate and multivariate ana1y_
ses (2546) . The MCM6 and Ki -67 indices also correlated With
OS in 30 patients with CNS WHO grade 2 oligodendrogliomas
2
(2546) . High mitotic count (? 2.5 mitoses/mm • equating to
? 6 mitoses/10 HPF of 0.55 mm in diameter and 0.24 mm2 iri
area) was associated with an increased growth rate on follow-
up MRI and shorter PFS in patients with CNS WHO grade 2 and
3 ollgodendrogliomas (2739).

Genetic alterations
Currently available evidence from retrospective studies sug-
gests that the presence of 1q and 19p co-polysomy detected
by FISH concurrent with 1p/19q codeletion is associated with
earlier recurrence and shorter survival (537,2974,3436) . Allelic
10 1a 20 2a
losses on 9p have been detected in about one third of CNS
0 N T~~tment
WHO grade 2 IDH-mutant and 1p/19q-codeleted oligoden-
32l7 H t 1 - RT drogliomas, but they were not associated with shorter survival
27 Q 29 21 I -RllPCV
{3441 J. Other studies reported that allelic losses of 9p21.3
Fig. 2.25 Oligodendroglioma, !DH-mutant and 1p/19q-codeleted. Progression-free (the CDl<N2A gene locus) were linked to shorter survival in
survival (PFS) of 80 patients in the prospective randomized European Organisation for
patients with CNS WHO grade 3 oligodendroglioma (886 ,64).
Research and Treatment of Cancer (EORTC) 26951 study on adjuvant procarbazine,
lomustine, and vincristine (PCV} chemotherapy after 59.4 Gy radiotherapy (RT); the
Homozygous deletion involving the COKN2A gene locus Is not
20-year PFS rate for the patients who received RT plus PCV chemotherapy was 31.3%, observed in CNS WHO grade 2 oligodendrogliomas (3441,
versus 10.8% for the patients who received only RT. 123), but It is found in a small subset of CNS WHO grade 3
oligodendrogliomas , in which it has been associated with poor
microvascular proliferation and/or necrosis was of prognostic sig- outcome (123). Other alterations that have been linked to less
nificance in cases lacking CDKN2A homozygous deletion (123). favourable outcome of patients with CNS WHO grade 3 oli-
godendroglioma include P/K3CA mutation (3140,380), TCF12
CNS WHO grading mutation (1778), and increased MYC signalling (1537). PTEN
Older studies reported CNS WHO grade as an independent mutation has been associated with shorter survival of patients
predictor of survival for patients with oligodendroglial tumours with CNS WHO grade 2 oligodendroglioma (3441}. Higher
(905 ,1096,1824,2312). However, these studies antedate the tumour mutation burden was found to predict shorter survival
molecular criteria for oligodendroglioma . In one study of with !DH-mutant gliomas including oligodendrogliomas (73).
patients with gliomas with concurrent IDH mutation and TERT CIC mutation has been reported as a marker of poor prognosis
promoter mutation , patients with grade 2 tumours had longer (1120). but this finding was not confirmed in other series (794,
survival times than those with grade 3 tumours (median OS: 3441} . No impact on outcome was observed for COK4 amplifi-
205 .5 months vs 127.3 months, respectively) {1613}. A recent cation or RB1 homozygous deletion (123}.
multicentre study observed a median OS of 188 months in
patients with grade 2 oligodendrogliomas versus 119 months in Treatment
patients with grade 3 tumours {974}. This difference remained The optimal postoperative treatment of patients with IDH-
significant in a multivariate analysis. A study of 176 patients with mutant and 1p/19q-codeleted oligodendrogliomas of CNS
IDH-mutant and 1p/19q-codeleted oligodendrogliomas (CNS WHO grade 2 is a matter of ongoing discussion . After tumour
WHO grades 2 and 3) also revealed shorter OS for patients with resection , radiotherapy and chemotherapy are often deferred
CNS WHO grade 3 tumours {597}. In contrast , a retrospective until tumour progression because therapy-associated neurotox-
analysis of 212 patients with !DH-mutant and 1p/19q-codeleted icity is a major concern (3417). Patients with symptomatic and
oligodendrogliomas did not detect CNS WHO grade as a sig- progressive tumours , with CNS WHO grade 3 tumours, or with
nificant predictor of OS (2318) . Similarly, data from a combined large residual tumours after surgery usually receive immedi-
cohort from Japan and The Cancer Genome Atlas (TCGA) sug- ate further treatment with radiotherapy and/or chemotherapy
gested that grading had a limited prognostic role (3082}. The (3417}. The European Organisation for Research and Treatment
interpretation of these retrospective studies requires caution of Cancer (EORTC) 22845 trial showed that adjuvant radiother-
because other prognostically relevant factors, such as extent of apy prolonged PFS but not OS in patients with progressive CNS
resection, were not considered, and patients received variable WHO grade 2 gliomas {3272). Long-term follow-up data from
postoperative treatments. randomized trials showed a major increase in OS after radio-
therapy plus PCV chemotherapy in patients with CNS WHO
Proliferation grade 3 oligodendrogliomas (402,3273,438) . Adjuvant chemo·
A study of 220 patients with !DH-mutant and 1p/19q-codeleted therapy with temozolomide or PCV may also be a feasible thera-
CNS WHO grade 3 oligodendroglioma revealed that label- peutic strategy for patients with progressive CNS WHO grade 2
ling index values of ~ 50% for MCM6 and ~ 15% for Ki-67 oligodendroglioma {1322,1598, 1859,3095) .

38 Gli rJmas . gl1one urorial tumours , and neurona l tumours


Glioblastoma, IDH-wildtype Louis ON
Aldape KO
Perry A
Reifenberger G
Capper O Sarkar C
Giannini C Soffietti R
Horbinsk1 CM Suva ML
Ng HK WickW

Definition
Glioblastoma , IOH -wi ldtype, is a diffuse, astrocytic glloma that
is IDH -wi ldtype and H3-wildtype and has one or more of the
following histological or genetic features: microvascular prolif-
eration . necrosis, TERT promoter mutation, EGFR gene amplifi-
cation , +7/-10 chromosome copy-number changes (CNS WHO
grade 4).

ICD-0 coding
9440/3 Glioblastoma , IOH-wildtype

ICD-11 coding
2AOO.OO & XH5571 Glioblastoma of brain & Glioblastoma, IOH-
wildtype
Flg.2.26 Glioblastoma, IDH-wildtype. A Glioblastoma, IDH-wildtype, with sarco-
Related terminology matous component (gliosarcoma). On this postcontrast Tl-weighted MRI of a patient
Not recommended: glioblastoma multiforme. with prior history of IDH-wlldtype glioblastoma, a solid-appearing component filled the
prior resection cavity and was found to be a newly developed sarcomatous component
in the recurrent tumour. B Epithehoid glioblastoma. IDH-wildtype. Postcontrast T1 -
Subtype(s) weighted MRI showing a solid-appearing enhancing mass.
Giant cell glioblastoma; gliosarcoma; epithelioid glioblastoma
(e.g. in tumours that meet genetic definitions of IOH-wildtype
Localization glioblastoma but lack histological microvascular prol iferation
Glioblastoma, IOH-wildtype, is most often centred in the sub- and necrosis), tumours show only modest or patchy contrast
cortical white matter and deeper grey matter of the cerebral enhancement or may lack central necrosis . They may extend
hemispheres , affecting all cerebral lobes {2344} . In many into adjacent lobes. into the opposite hemisphere through the
cases , tumour infiltration extends into the adjacent cortex and corpus callosum, and down into the brainstem . In the settin g
through the corpus callosum into the contralateral hemisphere. of a ring-enhancing mass, biopsies showing high-grade astro-
Glioblastoma , IOH-wildtype, also affects the brainstem, cer- cytoma but without frank histological features of glioblastoma
ebellum. and spinal cord ; however, in midline locations, other should be suspected to have been inadequately sampled .
diffuse gliomas should also be considered (e .g . diffuse midline The morphological subtypes of glioblastoma cannot be distin-
glioma , H3 K27-altered) . guished on MRI , although there are some differences: giant cell
glioblastomas may be more circumscribed and located subcor-
Clinical features tically (2298); gliosarcomas may appear as well-demarcated
Symptoms depend largely on tumour location, manifesting as lesions with a higher risk of cortical involvement and abutting
focal neurological deficits (e.g. hemiparesis, aphasia, visual dura {3086,27 14,975,3536}; and epithelioid glioblastomas often
field defects) and/or seizures (in as many as 50% of patients). consist of a well-circumscribed enhancing mass, rarely with a
Symptoms of elevated intracranial pressure, such as headache, cystic component /384 ,1370,3587). Radiomic approaches have
nausea, and vomiting, may coexist. Behavioural and neurocog- been developed to predict gene expression profiles of newly
nitive changes are common, especially in elderly patients. Neu- diagnosed glioblastomas based on computational analysis of
rological symptoms are usually progressive, but in a minority of conventional and advanced MRI images /1978 ,2209,586,752).
patients , acute onset may occur due to an intracranial haemor- thus increasing diagnostic and prognostic capabilities ( 141 ,
rhage . The time from symptom onset to diagnosis is < 3 months 3558,1608) .
in as many as 68% of patients and < 6 months in as many as
84% /2311) . In general, patients with histological subtypes such Spread
as giant cell glioblastoma /2298), gliosarcoma /975). and eplthe- A subset (ranging from 0.5% to 35% in different studies) of glio-
lioid glioblastoma /2143,1651,428,3587) present similarly. blastomas occur with multiple lesions, termed "multifocal" or
"rnulticentric " glioblastomas {2922,2420.3187,2426,1187,777} .
Imag ing Mult1focal glioblastomas demonstrate contiguous pathways of
Glloblastomas are irregularly shaped , often with a ring-enhanc- spread between foci, whereas multicentric glioblastomas are
ing component around a darker central area of necrosis, and widely separated . On careful histological analysis, only 2.4% of
surrounding oedema of varying amounts. Less commonly glioblastomas are truly multiple independent tumours {216 ,252.

G l1orn s. glloneuron ..: I tumours and neuronal tu111ours 39


Fig. 2.27 Glioblastoma, IDH-wildtype. Axial (A) and coronal (B) images of ring-enhancing tumour on postcontrast T1-weighted MRI. C FLAIR MRI shows the extent of vasogenic
oedema .

2759) . Occasional cases of multifocal epithelioid glioblastoma one member of a family or are inherited as part of genetic
as well as gliosarcoma have been reported [1046,1688,1758 , tumour syndromes [2342). The latter include Lynch syndrome,
2375). The exact pathogenetic mechanisms of multifocality are constitutional mismatch repair deficiency syndrome, Li-Frau-
unknown , although recent studies have suggested that these meni syndrome, and neurofibromatosis type 1. Genome-wide
tumours have frequent EGFR alterations with co-occurrence of association studies identified genomic variants in TERT, EGFR,
PTEN and TERTpromoter mutations {777,12). CCDC26, CDKN2B, PHLOB1, TP53, and RTEL1 associated
Despite the infiltrative growth of glioblastoma and its ability with an increased risk of glioma (2342) ; others showed that cer-
to seed the cerebrospinal fluid (e .g . along ventricular surfaces tain SNPs were associated with increased risk for gliomas, and
or via drop metastases) , extension into the dura mater, venous that these SNPs were different from those in patients with other
sinuses, and bone is uncommon {136 ,1159,1081,2534}. Extra- brain tumours {2065,1634,2346).
cranial metastasis is rare, occurring in only 0.4-0 .5% of cases, The incidence of glioblastoma seems to be increasing, which
mostly at the time of recurrence, with the most common sites suggests that environmental factors have a role in its develop-
being bones, lymph nodes, liver, and lungs {479,658}. Metas- ment {2496}. but although many environmental factors have
tasis has also been documented in association with interstitial been studied as potential causes, investigations have been
therapies and ventricular shunts {1224,1958,3355). Metastases inconclusive or negative for most, Including non-ionizing radia-
have also occurred in epithelioid glioblastoma and gliosarcoma tion (e.g . from mobile phones) and occupational exposures
{384 ,3253} . [3623,2346) . The only validated risk factor is ionizing radiation
to the head and neck [3480,2346). For example, patients who
Epidemiology received treatment for acute lymphoblastlc leukaemia were
Glioblastoma is the most frequent malignant brain tumour in more prone to developing glioblastoma {2795 ,2893,956), and
adults, accounting tor approximately 15% of all intracranial neo- there is an increased risk of gliomas among survivors of atomic
plasms and 45 - 50% of all primary malignant brain tumours . bomb irradiation, but there is no increased risk associated
It can manifest in patients of any age but preferentially affects with diagnostic irradiation [2555). A decreased risk has been
older adults, with peak incidence in patients aged 55-85 years. observed among individuals with a history of allergies or atopic
In children , it accounts tor approximately 3% of all CNS tumours diseases /2342).
{2344) . The M:F ratio for glioblastoma is 1.60:1 in the USA {2345)
and 1.28:1 in Switzerland (2310) . Pathogenesis
Annual age-adjusted incidence rates for glioblastoma have Cell of origin
increased in recent years to 3- 6 cases per 100 000 people, as Mouse modelling experiments suggest that a range of primary
documented in reports from the USA, Canada, England, and CNS cell types can be transformed into malignant cells that
Austral ia {2344,3358 ,2495,765). The increase cannot be fully recapitulate features of glioblastoma. These include oligoden-
accounted for by improvements in diagnostic techniques and drocyte precursor cells (1915), neural precursor cells [1333),
life style changes, and environmental factors might be responsi- astrocytes /1333), and neurons (9841. with the susceptibility
ble {2495,692,107,3330 ,1236,3342). Glioblastoma may be less to transformation declining with lineage restriction {60). Deep
common in Asian and African countries , which may be attribut- genetic sequencing studies of human glioblastomas suggest
able to d ifferences in age distribution and partly to under-ascer- that a neural precursor in the subventricular zone is a likely cell
tainment {747,2003) . of origin {1837) . This interpretation is supported by the coinci-
dent anatomical position of neural precursor cells in the sub·
Etiology ventricular zone and by the identification of stem cell- like cells
The etiology of most glioblastomas remains unknown {2346) . directly from glloblastomas {2943,2504,1111 }. Yet the question
A very small proportion of glioblastomas occur in more than of whether stem cell- like cells in glioblastoma are the result

40 ( ,1 1orr 1; s. qlioi 1~ _Jrumi. I tun 1our::,, ;.-md neur or al tun 1o urs


1
of the tran sformation of a neural precursor in the CNS or are
generated by ded ifferentiation of a lineage-res tricted ce ll type
remains unresolved . Single-cell RNA-sequencing analysis of
human tumours supports that glioblastoma is composed of
a mi xture of cell states that recapi tulate neurodevelopmental
traiectories (neural progenitor- like, oligoden drocytic progeni-
tor-like . astrocyte-like states) and are infl uenced by interactions
with immune cells (mesenchymal-like state). Genetic events
driving glioblastoma may skew the cellular lineages; in addition ,
the potential for cellular plasticity re presents an obstacle in the
search for the cell(s) of origin (2226).

Invasion, secondary structures, and metastasis


Infiltrative spread is a defin ing feature of all diffuse gllomas , but
glioblastoma is particularly notorious for its invasion of neigh-
bouring brain structures concurrent with rearrangement of the
extracellu lar matrix (4131. Infiltration occurs most readily along
white matter tracts , including the internal capsule, forn lx, ante-
rior commissure, and optic radiation , but it can also involve cor-
tical and deep grey matter structures. When infiltration extends
(AC-like)
through the corpus callosum , with subsequent growth in the Flg.2.28 Glioblastoma, IDH-wildtype. Single-cell RNA sequencing of glioblastoma
contralateral hemisphere, the result can be a bilateral , sym- identifies four malignant cellular states and their intermediates. Two-dimensional rep-
metrical lesion (butter-fly glioma) . resentation of cellular states, in which each dot represents a malignant cell and each
Other infiltrative patterns (including perineuronal satellito- coloured quadrant corresponds to one cellular state. The exact position and colour in-
s1s, perivascular aggregation , and subpial spread) give rise to tensity of each dot reflects the cell's relative score. AC, astrocyte; MES, mesenchymal;
secondary structures (3569). Infiltrative cells are located both NPC, neural progenitor cell; OPC. ollgodendrocytic progenitor cell.
inside and outside the contrast-enhancing rim of a glioblas-
toma and generally create a gradient of decreasing cell den- (see below). Many growth factors expressed in gl ioblastoma
sity with increasing distance from the tumour centre. Individual also stimulate migration by activating correspond ing recep tor
infiltrating tumour ce lls can be histologically identified several tyrosine klnases and downstream mediators that more direc tly
centimetres from the tumour epicentre, both in regions that promote migration . In EGFR-amplified glioblastomas, cells
are T2-hyperintense on MRI and in regions that appear unin- with amplification are enriched at the infiltrating edges , sug-
volved . These infiltrating cells are the most likely source of local gesting that this alteration has a role in peripheral expansion
recurrence after initial therapy, because they escape surgical {2975) . The overall mass migration of glioblastoma is rad ially
resection , do not receive the highest dose of radiotherapy, and outwards , away from central necrosis and associated severe
involve regions with an intact blood-brain barrier (which dimin- hypoxia, with migration rates substantially greater than th ose
ishes chemotherapeutic bioavailability) {1101) . In cases where of gliomas lacking necrosis (2720,3088). Hypoxia promotes
the dominant mass of glioblastoma is effectively controlled by invasion through the activation of HIF1 and other hypoxia-
therapy, distant invasion of brainstem structures is recognized inducible transcription factors /1574 ,3571 ). and glioma stem-
as a common cause of death {787}. Interestingly, a pattern of like cells respond to hypoxia by increasing HIF1-dependent
increased Infiltration has been observed in a subset of patients expression of alarm1n receptors /2386) and add itional factors
with glioblastoma treated with antiangiogenic therapies , pre- associated with cell motility /358 ,2720}. Activation of a pro-
sumably due to vascular normalization /702}. Like other diffuse migration transcriptional programme seems to be associated
gliomas , gl loblastoma can manifest at initial clinical presenta- with a decrease in proliferation , wh ich may have therapeutic
tion with a gliomatosis cerebri pattern of extensive involvement consequences {1396 ,1101 ,1102).
of the CNS, including multiple cerebral lobes, as well as addi- Although seeding of the cerebrospinal flu id can occu r rn the
tional involvement of deep grey matter structures , brainstem, setting of glioblastoma, systemic metastasis is uncommon, and
cerebellum , and spinal cord . the pathogenesis of glioblastoma metastasis remain s largely
tv1ec hanisms that promote invasive properties of glioblas- unknown /2626,2394) . The rarity of metastasis may be attri but-
toma cells include those involved in cell motility, cell-matrix able to immune mechanisms or inhospitable environments that
and cel l-cell interactions , and remodelling of the extracel- suppress metastatic implantation and growth , as well as to the
lular matrix, as well as microenvironmental influences 1235, relatively short survival of patients with glioblastoma (479,658,
734). Tumour cells produce migration -enhancing extracellular 1476,822). Nonetheless , circulating tumour cells have been
matrix components and secrete proteolytic enzymes that per- identified in the blood of some patients with gl iobl astoma, and
mit invasion. Gl iomas also express a variety of integrin recep- these cells express genes associated with stern ness and mes-
tors that mediate interaction s with molecules in the extracel - enchymal differentiation [3284 ,30691.
lular space and lead to alterations of the cellular cytoskeleton
and activation of intracellular signall ing networks such as Proliferation and apoptosis
the AKT, mTOR , and MAPK pathways . Tumour-associated Tumour cell proliferation is a hallmark of gl ioblastoma In most
macrophages (TAM s) are also involved in glioma invasion cases , mitoses are readily visible, but there is often intratumoural

Gl1om as , gl1 oneuronal tumours, and 11eurunal lt 1mours 41


Necrosis
70% In addition to microvascular proliferation , the other diagnostic
60% histological feature of glioblastoma is necrosis. There are sev-
50% eral postulated mechanisms by which necrosis develops in glio-
40% blastoma. One is that rapidly growing blood vessels within the
30% tumour have poorly formed luminal surfaces, which are throm-
20%
bogenic . This is exacerbated by glioblastoma cells secreting
10%
pro-coagulation molecules, such as tissue factor (TF), into the
0%
Aie (yem) <35 35-39 4()-44 45-49 50-54 55-59 60-64 65-69 70-74 75-79 80. circulation (3256). Thrombosis leads to infarction of surround-
(n=ll) (n=l6) (n=29) (n=61) (n=72) (n=9n (n=l09) (n=87) (n=SS) (n=57) (n=23)
ing tissues, creating a microenvironment that is acidic, low in
- 1q32.l (MDM4) - 4ql2 (PDGFRA) - 7pll.2 (EGF R) 12q14 .l (COK4) - 12ql5 (MDM2) oxygen, and low in glucose. Nearby glioblastoma cells migrate
away from such a hostile microenvironment, often creating pali-
Fig. 2.29 Glioblastoma, IDH-wildtype. Rate of recurrent amplifications in 647 molecu- sades of HIF1 a-expressing , less-proliferative tumour cells (3572,
larly confirmed cases of glioblastoma, IDH-wildtype (methylatlon groups RTK1 , RTK2,
358,3160,2720,775). The relationship between thrombosis and
and MES) plotted against age at diagnosis. Amplification rates in patients diagnosed
after the age of 40 years show little variation. Tumours of the methylation groups RTK1 , necrosis Is much stronger in IDH-wildtype glioblastoma than in
RTK2, and MES are rare in patients aged < 35 years and possibly do not occur in pa- !DH-mutant CNS WHO grade 4 astrocytomas. In the latter, TF
tients aged < 25 years. Data from {460} combined with institutional cases from Charite expression is greatly reduced, and when necrosis is present, it
Berlin and UZ Leuven. MES, mesenchymal; RTK, receptor tyrosine kinase. is usually less extensive than that in IDH-wildtype glioblastoma
and often lacks associated microthrombi (3256,1789}.
heterogeneity in the density of mitotic figures . Similarly, Ki-67
proliferation index values can vary greatly from region to region Inflammation
within a glioblastoma, sometimes ranging from about 5% to well Inflammatory infiltrates vary among glioblastomas: the major-
over 50% . Like other cancers , glioblastomas most commonly ity (~80%) are myeloid cells, with lymphocytes constituting a
overcome RB1-regulated cell-cycle control via molecular events smaller proportion (1659) . Myeloid cells are predominantly
like CDKN2A and/or CDKN2B deletion , CDK4 amplification , monocyte-derived macrophages and microglia - collectively
and RB1 inactivation (370). Glioblastoma proliferation is also referred to as tumour-associated macrophages (TAMs) -
directly stimulated by neuronal firing via neuron-glioma syn- whereas neutrophils are less abundant (1659). CD4+ T cells
apses (3307). Similar to what has been observed in glial inva- are the most abundant lymphocytes, followed by CD8+ T cells,
sion , proliferating glioblastoma cells recapitulate mechanisms NK cells, and B cells {1659).
found in normal dividing neural/glial progenitor cells (2226}. TAMs are highly plastic cells that are attracted to and
Along with mitoses, a common finding in most high-grade reprogrammed by glioma tumour cells via the secretion
CNS tumours (including gl ioblastoma) is tumour cell apopto- of chemokines and other soluble factors to promote inva-
sis. Apoptosis is a form of programmed cell death and can be sion, angiogenesis , and immunosuppression (1200}. When
caused by a number of factors, including a hostile microenviron- recruited to the glioma milieu , TAMs are reprogrammed to
ment (e.g. near necrotic areas) and a dysregulated cell cycle. promote glioma homeostasis and progression using mecha-
However. glioblastoma cells suppress apoptosis primarily via nisms that involve toll-like receptors and matrix metalloprotein-
specific genetic alterations, such as inactivating TP53 muta- ases. Functionally, TAMs in glioblastoma are thought to exert
tions , homozygous CDKN2A (p14ARF) deletion, and/or MDM2 predominantly immunosuppressive effects (2578), including
or MOM4 amplification {370). As a result, although apoptotic recruitment of CD4+, FOXP3+ T regulatory (Treg) cells and
cells can be found scattered throughout glioblastomas , they myeloid-derived suppressor cells (518,1100,2349) . Although
are outnumbered by proliferative cells and have no prognostic cytotoxic CDS+ T cells are usually thought to be hypoactive
value {2110 ,2836} . through immune checkpoint signalling, mediated by recep-
tors like PD1 (3478), brisk cytotoxic T-cell infiltration has been
Microvascular proliferation associated with longer survival {3519} . Treg cells constitute
One of the major histological features of glioblastoma is rapid about 10% of all intratumoural T cells in glioblastoma, and they
blood vessel growth, called microvascular proliferation, which are not associated with prognosis (3181,1284) .
manifests as multilayered small-calibre blood vessels . In some
glioblastomas, endothelial and smooth muscle cell I pericyte Cytogenetics and numerical chromosome alterations
overgrowth is so prominent that the vessels acquire a glomeru - Whole chromosome 7 gain (trisomy 7) and whole chromo-
loid shape, and can even show mitoses {1211,2194,3421) . Micro- some 10 loss (monosomy 10) are the most frequent numeri-
vascular proliferation is triggered by a number of mechanisms, cal chromosome alterations in glioblastoma and commonly
including perinecrotic hypoxia, which leads to HIF1a-mediated occur in combination (+7/-10) {3036); less common are gains
VEGF expression , thereby stimulating new blood vessel growth restricted to 7q and/or losses restricted to 1Oq . The most com-
{361 ,599) . Despite the prominent role of microvascular prolif- mon gene amplification involves the EGFR locus at 7p11. 2 (see
eration in glioblastoma biology, the antl-VEGF monoclonal anti- below) {3036). The sensitivity and specificity for the diagnosis of
body bevacizumab does not extend the overall survival (OS) of IDH-wildtype glioblastoma were reported, respectively, as 59%
patients with glioblastoma 11104}. and 98% for +7/-10, and as 36% and 100% for EGFR ampli~
fication {3036) . Other frequent numerical chromosome altera·
lions in 101-1 -wildtype glioblastomas are losses on 9p (including
homozygous deletion of the CDKN2A and/or CDKN28 locus at

42 (1l1rJ 11 1as gl 1CJ11e •H<Jr1a l t11111 n• w :. ancJ n<.; u 1o r1 a l turn<1LHS


9p21), 13q. 22q . and the sex chromosomes , as well as gains of p14ARF- MOM2- MDM4- p53 pathway
chromosomes 19 and 20 \370 ,318) Gene am p lifications often The p53 pathway is c entral to the induction of DNA repair, cell-
manifest cytogenet1cally as extrachromosomal double-minu te cycle arrest, an d apoptosis . Genetic dysregulation of thi s path -
chromosomes and can be detected in as many as 50% of glio- w ay occurs in nearly 90% of glioblastomas 1451,370). MDM2
blastoma karyotypes 1281). and MDM4 are both inhi bitors of p53, and they are activated
by amplification in about 15% of glioblastomas 1370 ). MDM2 is
Epidermal growth factor receptor itself inhibited by p14ARF, which is an altern ate read ing frame
The receptor tyrosine kinase (RTK) EGFR (HER1 ) is frequently protein encoded by the COKN2A locus. This locus is deleted
altered in IDH -wild type g liob lastoma. Overall , about 60% in about 60% of glioblastomas, resul ti ng in inactivation of Ule
of tumours show evidence of EGFR ampl ification , mutation , p53 pathway and activation of the RB1 cell -cycle pathway (see
rearrangement, or altered splicing \370 ). The most frequent of below) 1451 ,370) . TP53 itself is mutated or deleted in 20-25%
these alterations is EGFR amplification 11008). wh ich occurs in of IDH-wiidtype glioblastomas 13564,370}. Genetic alterations
about 40 % of al l IDH-wildtype glioblastomas 1370,3036) and in affecting COKN2A (p14ARF) , MDM2 or MOM4, and TP53 are
as many as 60% of g liob lastomas in the DNA methylation group mostly exclusive of one another 1370) .
RTK2, but only in about 25% of RTK1 and mesenchymal glio-
blastomas 13060 ,3036). In the majority of cases , EGFR amplifi- COK4/6-COKN2A/B-RB1 cell-cycle pathway
cations are associated with a second EGFR alteration, such as CDK4 and CDK6 catalyse the phosphorylation of RB1 and
extracellular domain mutations or in-frame intragenic deletions thereby the release of E2F transcription factors , wh ich then
encod ing eiiher EGFRv/11 or other alternative transcripts 1370, induce the expression of genes involved in the progression from
2868 ,3063). EGFR gene fusions are discussed below. G1 to S phase of the cell cycle . p16 (encoded by CDKN2A) is an
inhibitor of CDK4. Approximately 80% of glioblastomas demon-
Pl3K-AKT-mTOR pathway strate one or more genetic alterations of the CDK4/6-RB1 cell-
The P13K pathway is important for regulating cell growth . cycie pathway {451 ,370}. Most frequent are COKN2A deletions
Signalling is activated by RTKs and/or RAS and inhibited by that typically also involve nearby COKN2B (-60%) , amplifica-
PTEN . In IDH-wildtype glioblastoma, alterations of RTK genes , tions of CDK4 or COK6(-15%), and mutations/deletions of RB1
Pl3K pathway genes, and PTEN are found in about 90% of (-8%) {2259,2636 ,451,370). CDKN2A deletion and RB1 altera-
cases{370,451 }. Amplifications, truncations , and fusions of tions are mutually exclusive 13249,451), whereas CDKN2A dele-
RTK genes are particularly frequent , typically involving EGFR tion and CDK4 amplification may occasionally coexist {2259,
(-60% , see above), PDGFRA (10-15%), MET(2-5%) , or FGFR3 451 ,370}. In rare cases, the cell-cycle pathway is alternatively
(-3%) 12942,3315 ,370/. Mutations of NF1 that activate the Pl3K activated by deletion of COKN2C or amplifications of COK6 or
pathway via reduced RAS inhibition are present in approxi- CCND2 {451,370] .
mately 10% of cases {370). PTEN, which encodes the central
inhibitor of the pathway, shows mutation/deletion in about 40% Gene fusions
of glioblastomas , whereas mutations of PIK3CA, PIK3R1, or Gene fusions in IDH-wildtype glioblastoma are mostly confined
other Pl3K pathway genes are observed in 25-30% 1370}. RTK to members of the RTK family and recurrently involve EGFR
alterations may co-occur with P13K or PTEN alterations in about (6-13%); FGFR3 (-3%); MET (1-4%); or NTRK1 , NTRK2, or
40% of cases , whereas P13K mutations and PTEN mutations/ NTRK3 (1-2%) (see Table 2.02). EGFR fusions typically occur
deletions appear to be mutually exclusive /370} . as part of complex rearrangements at chromosome band
7p11 .2 {370). Fusion partners are mostly neighbouring genes of

Table2.02 Gene fusions with estimated frequencies of> 1% in IDH-wlldtype glioblastoma


Gene Frequent tu.Ion partner(•) Estimated frequency Comment
Co-occurs with EGFR amplification and often results
EGFR SEPTIN14, various others -!H3% in EGFR C-termlnal truncation {981 ,370.1031 ,3192)

Recurrently caused by chromosome 4p16.3 tandem


FGFR3 TACC3 -3% duplication, associated with amplification of CDK4 {2942,2400,370,193,751,907,752}
(22-44%) and MDM2 (W-25%)

More typical for !DH-mutant high-grade astrocytoma


MET PTPRZ1 , various others - 1-4% and diffuse. paediatric-type high-grade glioma, H3- {193,907,1362)
wild!ype and IDH-wildtype
NTRK1, Fusions also observed in CNS WHO grade 1-3
NTRK2i Various - t-2% gllomas; NTRK3tuslon very rare {981 ,3605,907,3192}
NTRK3

Gl1ornas , glioneu ronal tumour s nd neuronal turno u1s 43


to shorter survival and a low rate of response to PD1 immune
Active chromatin Repressive chromatin
checkpoint inhi bition in glioblastoma [3222}.

Epigenetics, chromatin, and promoter methylation


The interplay between epigenetic regulation and glioblastoma
tumorlgenesis has several modalities . Epigenetic modifiers can
be bona f Ide oncogenes or tumour suppressors affected by
gain- or loss-of-function genetic alterations, resulting in the dis-

,a_, •"" '. ,


0 " "'""'' 0 H3CM>• 1l
• ..,.,... I
ruption of epigenetic regulatory processes by affecting histone
modifications, DNA methylation , and chromatin remodelling
(3079}. Nearly half of 291 IDH-wildtype glioblastomas profiled
Flg.2.30 Glioblastoma, IDH-wildtype. DNA methylatlon, hlstone modifications, and
by whole-exome sequencing harboured one or more non.
numerous chromatin regulators determine the global structure of chromatin. Active
chromatin (left) is globally accessible for transcriptional regulation. Repressive chro- synonymous mutations affecting chromatin organization [370).
matin (right) sequesters portions of the genome, is enriched for characteristic histone Even in the absence of direct genetic alterations, epigenetic
modifications, and is refractory to regulatory activity. DNMTs, DNA methyltransferas- modifiers can modulate gene expression and impact glioma-
es; HDMs, histone demethylases; HMTs, histone methyltransferases. relevant processes {3058,3078).
A key function of chromatin regulation is to maintain inac-
EGFR (e .g. SEPTIN14, PSPH, SEC61G, SDK1), and the fusions tive portions of the genome in repressive structures . Canoni-
frequently cause truncation of the EGFR C-terminal autophos- cal repressive states include heterochromatin marked by H3
phorylation domain [981,370,3192). which has been shown to p.K28me3 (K27me3), a mark deposited by PRC2 and its cata-
be transforming (579) . EGFRfusion co-occurs with EGFRampli- lytic subunit, EZH2. EZH2 is overexpressed in IDH -wildtype
fication {981). FGFR3:: TACC3 fusions and EGFR ampli·fications glioblastoma and various other cancer types, presumably con-
are mutually exclusive, and tumours with FGFR3::TACC3 fusions tributing to the silencing of key tumour suppressor genes {714).
have higher rates of COK4 and MDM2 amplification {751,2942, Loss of function of EZH2 can also promote cancer in a con-
752,907) . The FGFR3:: TACC3 fusion is recurrently caused text-dependent manner. Although loss-of-function mutations of
by a tandem duplication on 4p16.3 [2400), which may allow EZH2 in IDH-wildtype glioblastoma are rare(< 1%), inhibition of
screening by copy-number analysis {370). The initially identi- EZH2 enzymatic activity occurs through mutation of H3 genes
fied FGFR1 :: TACC1 fusion [2942) was not observed in numer- in diffuse midline gliomas, resulting in p.K28M (K27M)-mutant
ous follow-up studies and is therefore no longer considered protein (370,2867,3481). Mutations in ATRX, which encodes
typical of glioblastoma {2400,370,751 ,193). MET fusions are a chromatin remodeller that deposits H3.3 in pericentromeric
more common in high-grade !DH-mutant astrocytomas {1362} and subtelomeric regions, were observed in 13 (4.5%) of 291
and diffuse, paediatric-type high-grade glioma, H3-wildtype IDH-wildtype glioblastomas (370}, in contrast to 60-70% of
and IDH-wildtype (1415}, but they may also occur in adult-type !DH-mutant gliomas and about 30% of paediatric high-grade
IDH -wildtype glioblastomas {907,1362,193}. NTRK1, NTRK2, gliomas {1474,2867,3482).
and NTRK3 fusions occur infrequently {981,3605,907,3192). Within chromatin , actively transcribed gene bodies are
PDGFRA fusions have been described but are exceedingly marked by H3 p. K37me3 (K36me3), a mark deposited by the
rare 165,370,907,2358}, whereas truncating deletion of exons 8 methyltransferase SETD2. Mutations in SETD2 occur in only
and 9 of PDGFRA recurrently occurs in POGFRA-amplified about 2% of IDH-wildtype glioblastomas and are more com-
tumours [2358} . Other potentially relevant fusions have been mon in paediatric and IDH-mutant gliomas {370,958) . Enhanc-
reported (e.g. PTPRZ1 ::ETV1, KLHU::BRAF, CEP85L::ROS1 , ers and promoters are marked by histone acetylation and H3
CCDC127: :TERD but require further validation {2038,3192). p.K5me (K4me). The methylatlon mark is catalysed by com-
plexes that contain MLL homologues. Missense mutations in
Tumour mutation burden KMT28, KMT2C, and KMT20 have been detected in rare cases
Tumour mutation burden has been proposed as a potential (2-3%) of IDH-wildtype glioblastoma . Histone deacetylases and
biomarker for estimating the abundance of neoantigens, which a range of histone demethylases are also infrequently mutated
may have relevance for immunotherapy. In one study of IDH- in IDH-wildtype glioblastoma, broadly affecting chromatin activ-
wildtype glioblastoma , a high mutation burden (defined as ity. Both histone and DNA demethylases are inhibited by IDH
> 20 mutations/1 .4 Mb) was identified in 5 (2.7%) of 182 cases mutations [3058).
and was associated with mutation or loss of immunohistocheml- Epigenetic gene silencing by DNA methylation is another
cal expression of one or more mismatch repair markers (MLH1, common mechanism of inactivating genes [2543 ,1151,1796).
MSH2 , MSH6 , PMS2) [1323}. Recurrent glioblastoma shows The MGMT gene encodes a DNA repair protein (598) and Is
higher rates of mismatch repair deficiency (-10%), mostly transcriptionally silenced by promoter methylation in approxi-
due to acquired MSH6 loss, and this is associated with a dra- mately 40 - 50% of IDH -wi ldtype glioblastomas (370,861,"1279.
matic increase of mutation burden 13372,1411}. A link between 2588) . MGMT specifically removes promutagenlc alkyl groups
alkylating agent / temozolomide treatment and occurrence of from the 06 position of guanine in DNA , thereby blunting the
MSH6 mutation has been demonstrated (1380,435,451 ,3538). treatment effects of alkylating agents (860,1062). MGMT pro-
Furthermore , POLE-m utated glioblastomas have a particularly moter methylation is present in 40- 50% of glioblastomas (370,
high tumour mutation burden and may demonstrate a better 861,1279,2588) and is predictive of benefit from therapy with
response to immunotherapy than do other glioblastomas 11323, alkylating agents such as temozolomlde (see below) (1279,
1483 ,8581 A high tumour mutation burden has been linked 1998,3430). A higher frequency of MGMT promoter methylation

44 r j l1u r 1 1(-1'-, ql 1<Jr 1uurur 1~1I rur nuurs , al 1cl 11 euruna l turnou rs
(> 75%) is associated with tumours that have the glioma CpG
island methylator phenotype (G-CJMP), which is characteristic
of JOH -mutant gliomas [172,370 ,2313} . Distinct DNA methyla-
tlon subclasses of glioblastoma have been suggested, partly
correlated to tumour genotypes and potentially to developmen-
tal origins !370,3060 ,460) .

Macroscopic appearance
Glioblastomas are often large at presentation and can occupy
much of a lobe. They are usually unilateral, but they can cross
the corpus callosum and be bilateral (a butterfly lesion). Most
hemispheric glioblastomas are clearly intraparenchymal and
centred in the white matter. Infrequently, they are superficial
and contact the leptomeninges and dura, sometimes mimicking
a metastasis or meningioma. Cortical infiltration may produce A
a thickened tan cortex overlying a necrotic zone in the wh ite
matter.
Glioblastomas are poorly delineated ; the cut surface Is varia-
ble in colour, with peripheral greyish to pink masses and central
areas of yellowish necrosis . In some areas, necrotic tissue may
also border adjacent brain structures without an intermediate
zone of macroscopically detectable tumour. Central necrosis
can occupy as much as 80% of the total tumour. Glioblastomas
are often stippled with red and brown foci of recent and remote
haemorrhage. Extensive haemorrhages can occur and cause
stroke-like symptoms, sometimes as the first sign of the tumour.
Macroscopic cysts, when present, contain a turbid fluid of liq-
uefied necrotic tumour tissue, in contrast to the well-delineated
cysts present in lower-grade diffuse astrocytomas .
Epithelioid glioblastomas are typically single lesions , although B
at least one multifocal example has been reported , and meta- Ag. 2.31 Glioblastoma, IDH-wildtype. A Glioblastoma with bilateral, symmetrical in-
static disease may occur (1046); leptomeningeal spread is also vasion of the corpus callosum and adjacent white matter of the cerebral hemispheres
relatively common . (butterfly glioblastoma). B Large glioblastoma of the left frontal lobe with typical
coloration: whitish-grey tumour tissue in the periphery, yellow areas of necrosis, and
Histopathology extensive haemorrhage. Note extension through the corpus callosum into the nght
hemisphere.
Glioblastoma, IDH-wildtype, is typically a diffusely infiltrating ,
highly cellular glioma composed of astrocytic, usually poorly
differentiated tumour cells that show nuclear atypia and often tumour periphery. The circumferential region of high cellularity
marked pleomorphism. Mitotic activity Is readily identifiable in and abnormal vessels corresponds to the contrast-enhancing
most cases and is often brisk. Microvascular proliferation and ring seen radiologically and is therefore an appropriate target
necrosis, with or without perinecrotic palisading , are charac- for biopsy. Microvascular proliferation is seen throughout the
teristic diagnostic features. Jn an JOH- and H3-wildtype diffuse lesion but is usually most marked around necrotic foci and in
glioma, at least one of these features (i.e. microvascular prolifer- the peripheral zone of infiltration.
ation or necrosis) is sufficient for the diagnosis of glioblastoma.
In specimens from treated patients, therapy-induced necrosis, Cellular heterogeneity and glioblastoma patterns
in particular radionecrosis , must be distinguished from innate Few human neoplasms are as morphologically heterogeneous
tumour necrosis. as glioblastoma. Poorly differentiated, fusiform, round , or pleo-
As the outdated term "glioblastoma multiforme" suggests, the rnorphic cells may prevail, but better-differentiated neoplas-
h1stopathology of this tumour is highly variable, which some- tic astrocytes are often discernible, at least focally \414) . The
times makes histopathological diagnosis difficult on specimens transition between areas that still have recognizable astrocytic
obtained by stereotactic needle biopsy (414} . Some lesions differentiation and highly anaplastic (small, round , primitive-
show a high degree of cellular and nuclear polymorphism, appearing) cells may be either continuous or abrupt. In gemis-
with numerous multinucleated giant cells; others are markedly tocytic lesions , anaplastic tumour cells may be diffusely mixed
cellular but relatively monomorphic. The astrocytic nature of with differentiated gemistocytes. An abrupt change in morphol ~
the neoplasms is easily identifiable (at least focally) in some ogy may reflect the emergence of a distinct tumour clone due
tumours, but it may be difficult to recognize in poorly differenti- to subclonal molecular diversification during tumour evolution
ated lesions (see Primitive neuronal cells and glioblastoma with a pnmitive
The distribution of histological features within a glioblastoma neuronal component. below) {997}.
is variable, but large necrotic areas usually occupy the tumour Cellular pleomorphism includes the formation of small , undif-
centre , whereas viable tumour cells tend to be found in the ferentiated , spindled, lipidized . granular, epithelioid , and/or

Gl1 om3S , gli oneuron I tu mours. di 1cl neurl1n.JI tur lours 45


giant cells. In some tumours, these patterns can dominate, for gemistocytic astrocytes (> 20% of tumour cells) (see Astrocy-
example in areas of bipolar, fusiform cells that form intersecting toma, /OH-mutant, p. 19).
bundles and fascicles resembling a spindle cell sarcoma. The
accumulation of epithelioid tumour cells with well-delineated Giant cells and giant cell glioblastoma
plasma membranes and a lack of cell processes, such as In Large, multinucleated tumour cells may be present in glio-
epithelioid glioblastomas (see below), may mimic metastatic blastoma and occur with a spectrum of increasing size and
carcinoma or melanoma. pleomorphism . Multinucleated giant cells are not found in all
Some glioblastomas have well-recognized patterns that are glioblastomas nor are they associated with a more aggressive
characterized by a predominance of a particular cell type. clinical course 1412}. The designation of a glioblastoma as a
These morphologies are discussed in the following subsec- giant cell glioblastoma - a longstanding and established histo-
tions , along with the corresponding histological subtypes and pathological subtype of glioblastoma - should be reserved for
patterns that can be established if a particular cellular morphol- those tumours in which bizarre, multinucleated giant cells are
ogy predominates . a dominant histopathological component. Glioblastomas aris-
ing from constitutional mismatch repair deficiency often exhibit
Gemistocytes and gemistocytic astrocytic neoplasms severe nuclear atypia and multinucleation (1616) .
Gemistocytes are cells with copious, glassy, non-fibrillary Giant cell glioblastomas are rare, accounting for < i% of all
cytoplasm that displaces the dark, angulated nucleus to the glioblastomas 12333,284). although they may be more common
periphery of the cell . Processes radiate from the cytoplasm but in paediatric populations (1561,1731,2098,2454). The M:F ratio
are stubby, not elongated . GFAP staining is generally positive . is 1.1-1 .5:1 11731,2333}. Giant cell glioblastomas typically
Perivascular lymphocytes frequently populate gemistocytic develop de nova after a short preoperative history and without
regions , but they are inconspicuous in other regions in the clinical or radiological evidence of a precursor lesion. They are
same neoplasm. Gemistocytes may be present in IDH-wildtype often located subcortically in the temporal and parietal lobes,
glioblastoma as well as 1n IDH -mutant astrocytoma; the term and they may be distinctive because of their circumscription.
"gemistocyt1c astrocytoma" describes a typically !DH -mutant which on imaging and intraoperatively can mimic a metastasis.
astrocytoma that 1s characterized by a large proportion of Giant cell glioblastomas are frequently rich in reticulin, are tirrn
and well circumscribed , and may be mistaken tor a metastasis
·- - ~
Fig. 2.33 Giant cell glioblastoma. A Numerous bizarre multinucleated giant cells. B GFAP immunoreactivity. C,D Giant cell glioblastoma in a patient with constitutional mis-
match repair deficiency syndrome (CMMRD). C Next-generation sequencing of this tumour and matched blood showed a biallelic germline mutation of PMS2, consistent with
a diagnosis of CMMRD. D This immunostain for PMS2 shows a lack of staining in both tumour cells and non-neoplastic cells, suggestive of CMMRD, which was subsequently
confirmed by next-generation sequencing. Note that CMMRD-associated glioblastomas mostly occur in young children and epigenetically align with the paediatric-type diffuse
high-grade gliomas (see Constitutional mismatch repair deficiency syndrome, p. 452).

or even a meningioma (when attached to the dura). They are DNA mismatch repair genes (205) . lmmunohistochemistry may
characterized histologically by numerous multinucleated giant show loss of one or a pair of mismatch repair proteins in giant
cells, in a background of small often fusiform cells {2012}. The cell glioblastomas , which is associated with a mutation in the
giant cells are often extremely bizarre; they can be as large as expected mismatch repair gene {205} .
0.5 mm in diameter and contain anywhere from a few to > 20 The prognosis of giant cell glioblastoma is poor, but the clini-
nuclei. Mitoses are frequent and can be seen both in giant cells cal outcome may be slightly better than that of ordinary glio-
and in the smaller tumour cells. A typical although variable fea- blastoma {418,1368,1731 ,2307,2333,2915) ; for example, in two
ture is the perivascular accumulation of tumour cells with the studies, median survival times of patients with giant cell glio-
formation of a pseudorosette-like pattern {1937}. Occasionally, blastoma were longer (11 and 13.5 months) than in standard
perivascular lymphocyte cuffing is observed . Palisading necro- glioblastoma (8 and 9.8 months) {1731,2333}.
sis or large ischaemic necrotic zones may be present, whereas
microvascular proliferation is not common . Giant cell glioblas- Mesenchymal metaplasia and gliosarcoma
toma shows consistent GFAP expression , although the level of In general , "metaplasia" refers to the phenomenon whereby a
expression is variable. OLIG2 expression is often found, either differentiated cell acquires morpholog ical features typical of
diffusely or focally, and more commonly in small tumour cells another type of differentiated cell. The term Is also used to
than in giant cells {1500}. designate aberrant differentiation in neoplasms. Metaplastic
The giant cell phenotype typically reflects a state of genomic changes in glioblastoma may be mesenchymal or epithelial
instability, often with superimposed TP53 mutations and/or mis- (see next subsection) . Mesenchymal metaplasia may corre-
match repair defects. Although most giant cell glioblastomas spond to differentiation along various lineages, with spindled
are IOH - and H3-wildtype, !OH-mutant gllomas and H3-mutant cells resembling fibroblast-like differentiation being most com-
gliornas may show giani cell features 12235). Genetically, giant mon . Patterns resembling osseous , chondroid , adipocytic, or
cell glioblastoma does not seem to represent a distinct tumour myogenic differentiation are rare . Sarcomatous metaplasia is
entity, but it stratifies into different genotypes. encountered most often within the setting of an IOH-wildtype
Especially in young patients , numerous multinucleated giant glioblastoma, but metaplasia can also be seen rarely 1n IOH -
cells in glioblastomas may point to an underlying defect in ONA mutant astrocytomas . H3-mutant gliomas , !DH-mutant and
repair due to inherited or acquired mutations in POLE {858) or 1p/19q -codeleted oligodendrogliomas (oligosarcoma) 12699).

Gl1ornas, gl1oneurona1 tumours . and r1euro1nl rumours 47


.. .I

-.. ,.,'Ji''
1
~

'· ~,
-

r;.
'

-
C ,. .,. D· \.. ~ :i...~ ."".·: ::. .,~~
Fig.2.34 Gliosarcoma. A Gliosarcoma with osteosarcoma-like foci. B Alternating areas of reticulin-poor glioma and reticulin·rich sarcoma. C GFAP highlights the glioma
components. D The glioma regions are positive for OUG2.

and ependymomas (ependymosarcoma) {2700). It may be activity, microvascular proliferation , and/or necrosis. The sarco-
encountered either de nova at presentation or at the time matous component often demonstrates the pattern of a spin-
of recurrence . The designation of gliosarcoma, also a long- dle cell sarcoma, with densely packed long bundles of spin-
standing and established histopathological subtype of glio- dle cells surrounded individually by reticulin fibres . The glial
blastoma , should be reserved for tumours showing prominent component, typically seen as reticulin-free nests or islands of
mesenchymal differentiation, characterized by a biphasic pat- fibrillary ~r gen:iistocytic astrocytoma cells, is positive for glial
tern with alternating areas displaying glial and mesenchymal markers, 1nclud1ng GFAP and OLIG2, which are negative or only
differentiation. focally _positive in the sarcomatous component (2304,1500).
Gliosarcomas are rare , accounting for approximately 2% of Occasionally, the sarcomatous component shows considerable
glioblastomas (1023,1730,1369). Their age distribution is similar pleomorphlsm (2240}. A subset of cases show additional lines
to that of glioblastoma overall, with preferential manifestation in of mesenchymal differentiation, such as the formation of carti-
patients aged 40-60 years (mean age: 52 years). Rare cases lage (190}, bone {2036}, osteoid-chondroid tissue {669,1271 ,
occur in children (1563), and the M:F ratio Is 1.4- 1.8:1 (1227}. 3102). smooth and striated muscle {1603 ,3084) , and even lipo-
Gliosarcoma typically occurs de nova with symptoms of short matous features {1001). Primitive neuronal components occur
duration that ref le ct the location of the tumour and increased rarely (1554,3529) . The gliosarcoma subtype of IOH -wildtype
intracranial pressure. Gliosarcoma can also. arise seco~dar­ glioblastoma is negative for IDH1 p.R132H and does not show
ily after conventional adjuvant treatment of high-grade gl1oma IDH1 or IDH2 mutations .
(1226) . It usually occurs supratentorially, involving the temporal , The sarcomat~us areas of gliosarcoma are thought to result
frontal , parietal , and occipital lobes (in descending _order of fre- from a phenotyp1c change In the glioblastoma cells (rather than
quency). Posterior fossa (2240,2263}, lateral ventricles (2811 J. from the coincidental development of two separate neoplasms,
and spinal cord 1475) are rare locations, and some tumours are termed a "collision tumour ", as originally hypothesized), and
multifocal 12375). . they may reflect the clonal evolution of a tumour. This hypoth-
Because of its high connective-tissue content, g_llosarcoma esis Is supported by studies that have demonstrated common
has the gross appearance of a firm , well-circumscribed mass, molecular abnormalities between the glial and sarcomatous
whi ch can be mistaken for a metastasis or (when attached to components of the tumour, including gain of chromosome 7 and
the dura) a meningioma . Histologically, it is characteri~ed by loss of chromosome 10 {312) and identical mutations in TP53,
a mixture of gl1omatous and sarcomatous tissues .. which ~y PTEN, and TERT (280,2646,3357). as well as CDKN2A dele·
def init1on show high-grade malignant features including m1tot1c tion and MOM2 and CDK4 co-amplification (2646} . Results frorn
microarray-based comparative genomic hybridization analysis Epithelioid g/ioblastoma
in the glial and mesenchymal tumour areas also su ggest that Epithelioid glioblastoma is a histological subtype of glioblas-
the mesenchymal components may be derived from glial cells toma defined by a mostly sharply demarcated. loosely cohesive
with additional genetic alterations in a subset of gliosarcomas agg regate of large epithelioid to rhabdoid cells with abundant
!21901. cytoplasm. large vesicular nuclei, and prominent macronucle-
Gliosarcomas have a genetic profile that is similar, but not oli. sometimes mimicking metastatic carcinoma or melanoma.
identical , to that of IDH-wildtype glloblastoma: PTEN mutati ons, Recent studies suggest that epithelfoid features are most com-
CDKN2A deletions, and TP53 mutations, but infrequent EGFR mon in three distinct molecular subclasses: {1) a prognostically
amplification !17,2646) . Chromosomal imbalances are com - more favourable tumour of children and young adults that over-
mon, with frequent gains on chromosome 7 (up to 75%), and laps greatl y with pleomorphic xanthoastrocytoma genetically
losses of chromosome 9 (mostly correlating to COKN2A loss) (BRAF p.V600E mutation and homozygous COKN2A deletions)
and chromosome 10 (up to 72%) !17,1 947}. At the protein level , and epigenetically (DNA methylation profile); (2) a poor-prog-
expression of SNA12, TWIST, MMP2, an d MMP9 is characteris- nosis tumour of older adults that has features of conventional
tic of mesenchymal tumour areas, suggesting that the mecha- IDH -wildtype glioblastoma (albeit with more frequent BRAF
nisms involved in epithelial-mesenchymaJ transition in epithel ial p.V600E mutations); and (3) an intermediate-prognosis tumour
neoplasms may also pertain to mesenchymal differentiation in with features of the RTK1 -type paediatric high-grade glioma,
gliosarcomas !2191}. frequently associated with PDGFRA amplification and ch romo-
The prognosis of patients with gliosarcoma is poor, with OS thripsis (1716}. Rare examples of H3 K27- altered diffuse midline
being similar to that of patients with histologically classic IDH- gl ioma also show eplthelioid features {2988}, and other glioma
wildtype glioblastoma (1 023). There have been multiple reports types may occur with this pattern as well.
of gliosarcomas with spinal and systemic metastases and even Epithelioid glioblastomas are dominated by a re latively uni-
invasion of the skull (149,2841 ,227,2863,108). form population of epithelioid cells showing focal loss of cohe-
sion , scant intervening neuropil, a distinct cell membrane, abun-
Epithelial metaplasia dant eosinophilic cytoplasm, and eccentric or centrally located
Epithelial metaplasia in glioblastoma is rare and may comprise nuclei. At least focal rhabdoid cytology is seen in most tumours.
areas of squ amous or adenomatous differentiation . Tumour Exceptional cases have contained giant cells (1652}. lipidiza-
cells can display features of squamous epithelial cells, including tion !2698}. a desmoplastic response {2698}. or cytop lasmic
epithelial whorls with keratin pearls and immunohistochemlcal vacuoles {2266}. Epithelioid glioblastoma may show areas with
expression of squamous cell-associated markers like CK5/6 pleomorphic xanthoastrocytoma-like histology, although this
!416,2164 ,2698). Other cases may contain foci with glandular appearance does not reliably associate with the pleomorphic
and ribbon -like epithelioid structures that mimic metastatic xanthoastrocytoma-like molecular group (1716}.
adenocarcinoma !2728). Some glioblastomas may contain Rosenthal fibres and eosinophilic granular bodies are uncom-
so many of these structures that they are referred to as either mon. Necrosis is often present, but it is usually zonal rather than
adenoid glioblastoma (when they retain their glial immunophe- palisading . Some reports have noted a relative paucity of micro-
notype) or glioblastoma with epithel ial metaplasia (when they vascular proliferation , but others found no substantial difference
show a true epithelial immu nophenotype) !2698}. SmaJI cells from classic glioblastoma in vascular patterns !384}.
with more marked epithelial features and more cohesiveness Epithelioid glioblastomas show immunoreactlvity for GFAP,
are less common {1591}. Adenoid features and true epithelial although it is often patchy (and in a few cases , entirely absent);
metaplasia are sli ghtly more common in gliosarcoma than in therefore, OLIG2 positivity may be helpful for establish ing
ordinary glioblastoma !1591 ,2164 ,2698} . glial lineage !71). Some tumours are focally immunoreactive

# -

Flg.2,35 Adenoid features in ghoblastoma, IDH-wildtype. A Adenocarcinoma·like cytology with anaplaslic ep1thelio1d cells arranged in nests and rows. I Despite the carc1no-
rna-like appearance, the glial h1stogenesis of this glloblastoma Is supported by strong nuclear expression of OLIG2.

Gliornas . gl1oneuronal tumours . amJ neuro11al tumours 49


- . .. ,. -.
..... .. ·-~
Ftg.2.36 Epithelioid glioblastoma A In contrast to most glioblastomas, the epithelioid pattern Is often associated with relatively sharp demarcation from adjacent brain. a Tu-
mour cells are loosely cohesive and composed of large epithelioid to rhabdoid cells, often resembling metastatic melanoma or carcinoma. C Some examples show only limited
GFAP expression. D A second glial marker (OLIG2) was positive in this case, helping to establish the diagnosis. E lmmunoreactivity for BRAF p.V600E-mutant protein.

for EMA and cytokeratin cocktails (which is probably due to However, more recent studies suggest that like lower-grade
cross-reactivity with GFAP) (384,2698}, but CAM5 .2 immuno- tumours with both oligodendroglial and astrocytic-appearlng
reactivity is typically negative. Most authors have noted focal components (oligoastrocytomas), glioblastomas with oligoden-
immunoreactivity for synaptophysin or neurofilaments. Expres- droglial components are molecularly heterogeneous. Since
sion of 8100 and BRAF p.V600E may be mistaken for evidence 2016, the WHO classification has not considered glioblastoma
of metastatic melanoma, but epithelioid glioblastomas do not with an oligodendroglioma component to be a distinct diag-
express specific melanocytic markers such as HMB45 and nostic entity; instead, such tumours genetically correspond
melan-A. SMARCB1 expression and (in cases where it has to (1) IDH-wildtype glioblastoma (in particular the small cell
been sought) SMARCA4 expression are retained {384). lmmu- pattern , given the morphological overlap with oligodendroglial
nohistochemical staining for BRAF p.V600E is seen in roughly cells), (2) IDH -mutant diffuse astrocytoma (CNS WHO grade 3
half of all cases of epithelioid glioblastoma, but it is most com- or 4), or (3) IDH-mutant and 1p/19q-codeleted CNS WHO
mon in the pleomorphic xanthoastrocytoma-li ke molecular grade 3 oligodendroglioma (1312}.
group and least common in the paediatric RTK1 molecular
group !1652,1716). Small cells and small cell glioblastoma
Some IDH-wildtype glioblastomas feature a predominance of
Oligodendrocyte-lfke cells cells with highly mon.omorphi.c, small , round to slightly elon-
Occasional glioblastomas contain oligodendrocyte-like clear gated , ~yperchromat1c ~ucle1 and minimal discernible cyto-
cells with round nuclei that mimic oligodendroglioma, some- plasm , little .nu.clea.r atyp1a , and (often) brisk mitotic activity. In
times includ ing a chicken wire-like capillary network and the zone of 1nf1ltrat1on, tumour cells can be difficult to identify,
microcalcifications. Oligodendroglioma-like foci may be focal given their small size and bland cytology. GFAP immunore-
or diffuse , although individual thresholds for identi fying oli- activity variably highlights delicate processes, and the Ki-67
godendroglial features vary greatly. Notably, FGFR3: :TACC3 proliferation index is typically high . Because of their nuclear
fusion- positive glioblastomas often show this pattern 1276). regularity, clear haloes , microcalcifications , and chicken wire-
Two large studies of malignant gliomas in the pre-IDH era like microvasculature, these tumours may resemble anaplastic
suggested that necrosis was associated with a significantly oligodendrogliomas (2464) . But unlike oligodendrogliomas,
worse prognosis in the setting of anaplastic glioma with both small cell g~ioblastomas a~~ u~iformly IDH-wildtype, fre-
oli godendrogl ial and astrocytic components 12113,3274). quently showing EGFR ~mplif1cat1on (in -70% of cases) and
Such tumours were also previously classified as glioblastomas chromosome 10 losses (in > 95%) . IDH mutations are absent
with an oligodendroglial component and reported to have a (1500,2464 ,3117), as is 1p/19q codeletion (3117} . The clinical
better prognosis than classic glioblastoma 11273,1337,17341. behaviour of the small cell pattern is similar to that of other

50 ( Jl1 1J11 1as r:.J l 1 r; r •~u 1 0 1 1r-i l tumo urs and neu ron al tum ours
gl ioblastomas , with an OS time of < 12 months in two series Primitive neuronal cells and glioblastoma with a primitive
(2464 ,3117) . In one of these series , about a third of the tumours neuronal component
appeared as non-enhancing or minimally enhancing masses Rare glioblastomas may occur with one or more solid-looking
with no evidence of microvascular proliferation or necrosis primitive nodules showing immature cells with variable neu-
on histology \2464) . However, follow-up imaging 2-3 months ronal differentiation (2470). The primitive foci are often sharply
later often showed ring enhancement, and survival times were demarcated from the adjacent glioma, and they display mark-
shorter for these patients (median : 6 months), consistent with edly increased cellularity, with high N:C ratios and mitotic-kar-
such situations being early presentations of glioblastoma yorrhectic index values . More variable features include Homer
(2464 }. Wright rosettes, cell wrapping , and anaplastic cytology similar
to that of medulloblastoma or other CNS embryonal neoplasms. forms between granular cells and neoplastic astrocytes can
Additional features include immunoreactivity for neuronal be identified in some cases, but in others it is difficult to iden.
markers such as synaptophysin , reduction or loss of GFAP tity any conventional astrocytoma component. Although larger
expression, and a markedly elevated Ki -67 proliferation index and more coarsely granular, the tumour cells may resemble
compared with that in adjacent areas of glioma. Survival time macrophages . Especially in the context of perivascular chronic
and genetic background are similar to those of glioblastoma in inflammation , the tumour cells may be misinterpreted as a mac.
general 12470). However, these tumours were reported to show rophage-rich lesion such as demyelinating disease. Given their
a high rate (30- 40%) of cerebrospinal fluid dissemination and lysosomal content, granular tumour cells are sometimes immu.
increased frequency (-40%) of MYCN or M YC gene amp li- noreactive for macrophage markers such CD68, but not for
fication . Spread to the lungs has also been reported (3 11 9) . lineage-specific markers such as CD163 . Some cells may have
MYC amplifications are found only in the primitive-appearing peri pheral lmmunopositivity for GFAP, or the cells may be corn-
nodules , and it is likely that such alterations drive the primitive- pletely GFAP-negative (364,1051). When granular cell change is
appearing clonal transformation at least in part, given that a ex tensive, the tumou rs have been termed "g ranular ce ll astro-
similar phenotype has been observed in Mycn-drive n murine cy toma" or "granular ce ll glioblastoma". These lesions have a
forebrain tumours (3089) . A primitive neuronal com ponent has distin ct histological appearance and are typically characterized
also been reported in !DH-mutant high-grade astrocytic glio- by aggressive glioblastoma-li ke cli nical behaviou r {364}, even
mas {1500 ,2991 ,3495) and to a lesser extent in H3 G34- mutant when the histology otherwi se suggests a lower-grad e designa-
diffuse hemispheric gliomas, which were previously mistaken tion . A review of 59 reported patients found median survival
for supratentorial primitive neuroectodermal tumours in roughly times of 11 months for patients with CNS WHO grade 2 granu-
halt of all cases , even though they do not always show immuno- lar cell astrocytoma and 9 months for patients with CNS WHO
reactivity tor neuronal markers {1 713). Similarly, H3 K27- altered grade 3- 4 tumours (2843) . Another recent study of 39 patients
diffuse midline gliomas may al so show primitive foc i resembling (including patients with tumours histologically corresponding
an embryonal neoplasm (2988). to CNS WHO grades 2, 3, and 4) showed a mean OS of only
11 .3 months; notably, survival did not correlate significantly with
Granular cells and granular cell astrocytoma/glioblastoma CNS WHO grade, extent of granular cell change, sex, or Ki-67
Large cells with a granular, PAS-positive cytoplasm may be (MIB1) index (3337) . That study did not find IDH mutations, but
scattered within IDH-wil dtype glioblastomas . In rare instances , it identified TERT promoter mutations and +7/-10 copy-number
they dominate and create an appearance similar to that of changes in the majority of tumours , consistent with IDH -wildtype
granular cell tumou rs in other parts of the body. Transitional glioblastoma in general {3337).
Lipidized cells and heavily fipidized gfiobfastoma
Cells with foamy cytoplasm are anoth er feature occ asionally
observed in glioblastoma. Th e rare lesions in wh ich they pre-
dominate have been designated malignant gllomas with heavily
lipidized (foamy) tumour ce lls {1 589 ,1594 ,2728,3 129). The
lipidized cells may be grossly enlarged {1080), and lobules of
juxtaposed fully lipidized adlpocyte -like ce lls can simulate adi -
pose tissue . Pleomorph ic xa nthoastrocytoma should be consi d-
ered in the differential diagnosis of such lesion s.

/mmunophenotype
By definition , IDH -wi ldtype glioblastomas lack immunostain-
ing for IDH1 p.R132H and do not demonstrate positivity with
mutation-speci fic antibod ies against H3 p.K28M (K27M),
H3 .3 p.G35R (G34R), or H3 .3 p.G35V (G34V) . Nuclear
immunostaining for ATRX is retained in the vast majority of
tumo urs , and widespread nuclear positivity for p53 is seen
in approximately 25- 30% of tumours . Nuclear p53 positivity
is particul arl y frequent in the giant cell glioblastoma subtype . Flg.2.40 Glioblastoma, IDH-wildtype. This intraoperative smear preparation shows
elongated to irregular hyperchromatic nuclei, with thin eosinophilic cytoplasmic pro-
Gliob lastomas often express GFAP, but the degree of reactiv-
cesses.
ity differs markedly between cases; for example, gemistocytic
areas are frequently strongly positive , whereas primitive cel-
lular components are often negative. S100 expression is also cellular extensions. Multinucleation can be seen and is promi-
common. OLIG2 is a highly specific glioma marker and may be nent in some cases , as are mitotic figures .
of diagnostic utility, being strongly positive more commonly in
astrocytomas and oligodendrogliomas than in ependymomas Diagnostic molecular pathology
and non-gl ial tumours {1421 ,2348 ,334 ,3225); tumours with low IDH-wildtype glioblastomas lack mutations in IDH1 codon 132
nuclear expression of OLIG2 (< 30%) after adjuvant treatments and IOH2 codon 172, and they do not carry H3 p.K28 (K27 )
may have a shorter time to recurrence and be associated or H3 p.G35 (G34) mutations. Absence of immunoreactivity
with shorter survival (334) . Cytokeratin positivity may primar- for IDH1 p.R132H Is sufficient (i .e. without further sequenc-
ily indicate cross-reactivity with GFAP; lmmunostaining with ing) to diagnose IDH-wildtype glioblastoma in a patient aged
the keratin antibody cocktail AE1/AE3 is most often positive, in ~ 55 years at diagnosis who has a histologically classic glio-
contrast to the lack of positivity detected for most other kera- blastoma not located in midline structures and no history of a
tins 13165). However, glioblastomas with epithelial metaplasia pre-existing lower-grade glioma {1940} . This practical approach
may show expression of epithelial markers including cytoker- is possible because the probability of a non-canonical IDH
atins in the epithelial component. Sarcomatous components mutation is< 1% in glioblastomas from patients aged~ 55 years
in gliosarcoma typically lack expression of glial markers but (539} . In patients aged < 55 years, or in patients with a his-
react positively for vimentin . Rare cases may show expression tory of lower-grade glioma and/or whose tumours show immu-
of markers indicating differentiation along myogenic or other nohistochemical loss of nuclear ATRX expression , negative
mesenchymal lineages (see Gliosarcoma, above). Cancer IDH1 p.R132H immunostaining should be followed by DNA
stem cell biomarkers such as CD133, CD44, SOX2, OCT4, and sequencing for less common IDH1 or IDH2 mutations. When
nestin may be found in glloblastomas {1260,132,54) but are of no IDH mutations are detected by sequencing , such tumours
limited significance In diagnostic work . Notably, intratumoural are classified as glioblastoma, IDH-wildtype. However, tumours
heterogeneity for immunohistochemical positivity is common in located in midline structures should additionally be evaluated
glioblastomas, with differential expression of markers such as for H3 p.K28M (K27M) mutation to exclude diffuse midline
nestin, MAP2, and GFAP within different regions of the same glioma, H3 K27-altered . In hemispheric tumours , particularly in
tumour (249) . Expression of EGFR is frequent in IDH-wildtype younger patients, H3 G34-mutant diffuse hemispheric gllomas
gl ioblastoma and particularly strong in tumours with EGFR should be excluded by immunohistochemistry for H3.3 p.G35R
amplificaiion , approximately half of which additionally show (G34R) or H3.3 p.G35V (G34V) mutation or by H3-3A (H3F3A)
immunopositivity for EGFRvlll (906) . sequencing .
Frequent and diagnostically relevant molecular alterations in
Cytology IDH-wildtype glioblastomas include TERT promoter mutations,
lntraoperative smear preparations of glioblastomas are valuable EGFR gene amplification , and a +7/-10 genotype (3036). The
and complement the findings from histological sections. Cyto- presence of at least one of these aberrations in an IDH- and
logical preparations usually demonstrate marked hypercellular- H3-wildtype diffuse glioma allows for the diagnosis of IDH-
ity and nuclear pleomorphism , along with a discernible fibrillary wildtype glioblastoma even in the absence of microvascular
background that is useful for establishing glial differentiation. proliferation and/or necrosis {356 ,1944).
Eosinoph1 lic cytoplasm and processes vary, from naked nuclei In addition , demonstration of a DNA methylation profile of
lacking visi ble cytoplasm to gemistocytes showing elongated IDH-wildtype glioblastoma with a significant calibrated score is

Gl1ornas . giloneuronal tumours. ::im.i neuronal tumou1 s 53


sufficient for the diagnosis 1460). DNA methylation profiles may Box2.03 Diagnostic criteria for glioblastoma, IDH-wildtype
further stratify molecular subgroups, with the RTK1 , RTK2/clas- Essential:
sic , and mesenchymal subgroups being most common in adult An IDH-wildtype, H3-wildtype, diffuse astrocytic glioma
patients 13060,460). In this age group, the clinical relevance of AND
methylation-based subgroups is still limited . In contrast, high-
One or more of the following :
grade gliomas of children and adolescents may demonstrate
• Microvascular proliferation
less common DNA methylation profiles that have been linked
to significantly longer survival 11721 ,1723). DNA methylation • Necrosis
profiling can also facilitate the diagnosis of challenging cases • TERT promoter mutation
by helping to distinguish glioblastoma from histologically similar • EGFR gene amplification
entities [1559 ,1458,463). • +71-10 chromosome copy-number alterations
BRAF p.V600E mutation is rare in IDH -wildtype glioblastoma
Desirable:
{28421 but is detectable in as many as 50% of glioblastomas
DNA methylation profile of glloblastoma, IDH-wildtype
with epithelioid histology [1651} : BRAFp.V600E is found in 79%
of pleomorphic xanthoastrocytoma- like tumours and 35% of
adult-type IDH-wildtype glioblastomas, but 0% of paediatric Staging
RTK1 tumours [384,1651 ,1716). TP53 mutations are detect- Not clinically relevant
able in about a quarter of all IDH-wildtype glioblastomas but
are found in > 80% of giant cell glioblastomas {2454}, which Prognosis and prediction
less commonly carry EGFR amplification and TERT promoter Most glioblastoma patients die within 15-18 months after ther-
mutations (2304}. EGFR amplification appears to be frequent apy with chemoradiation . The 5-year survival rate has been
in small cell gl ioblastomas [415 ,2464}, whereas MYC or MYCN reported as 6.8% in the USA between 2012 and 2016 [2344)
amplification has been linked to primitive neuronal components and as 10% in clinical trials with somewhat more favourable
(2470} . However, none of these alterations is specific or suf- patient selection (3057). Younger age(< 50 years) , high perfor-
fici ent for the respective morphological subtypes or patterns . mance status , and complete tumour resection are associated
Gliosarcomas rarely demonstrate EGFR amplification but oth- with longer survival , as is MGMT promoter methylation [3057
erwise lack disti nguishing genetic alterations (17,2304) . Novel 2638). Patients with IDH-wildtype glioblastomas have shorter
predictive biomarkers for molecularly targeted therapies in survival times than patients with CNS WHO grade 4 IDH-mutani
subsets of glioblastoma patients are under evaluation ; these astrocytomas with similar histological features [1244} .
include high tumour mutation burden , BRAF p.V600E mutation , Some individuals with glioblastomas benefit from current
NTRK or FGFR gene fam ily fusions , and MET amplification or treatments, including maximal safe surgery, radiation , alkylating
fusions (1821) . chemotherapy, and bevacizumab (where approved), as well as
MGMT promoter methylation status is commonly determined experimental and immunological interventions. However, there
in IDH-wildtype glioblastomas because it provides clinically is marked heterogeneity in treatment response , which probably
relevant information on response to chemotherapy and survival reflects the biological heterogeneity of the disease. For exam-
of patients treated with temozolomide (1279,3431) or temozolo- ple, more extensive surgery may be limited by the location of
mide plus lomustine (CCNU) [1299}. In elderly patients , MGMT the tumour in an eloquent area and by the often diffusely infiltra-
promoter methylation status may guide decisions on chemo- tive growth pattern [3409,3459) . And for most treatments, the
therapy or radiotherapy (2634,3430,1998). basic molecular mechanisms for primary or acquired resistance
Gene expression patterns can be used to distinguish glio- are unknown or incompletely understood . Biological properties
blastoma from pilocytic astrocytoma 12671}, other malignant such as hypoxia, necrosis , DNA repair capacity, specific muta-
astrocytomas {1009), and ollgodendroglioma [1009). as well as tions (e.g. in PTEN) , and volume of the disease may influence
JOH-mutant glioma from IDH-wildtype glioma, across grades radiation response (3427,3439 ,3430,2476 ,1963), but most such
and histology 1370,1145,2287,3060}. Unsupervised analysis of observations lack clinical validation .
expression profiles can be used to cluster gliomas Into groups
that correlate with histology and grade {1162,2293) and may be Age
a better predictor of patient outcome [854) . A commonly used Clinical trials have shown that younger patients(< 50 years) with
glioblastoma gene expression classifier defines proneural , clas- glioblastoma have longer survival times [3057,573,1104,1105).
sic, and mesenchymal subtypes , which correlate in part with with the age effect persisting through all age groups in a lin-
mutations in TP53, mutation/amplification in POGFRA or EGFR, ear manner 12309). In addition , coexisting medical and social
and deletion/mutation in NF1 13315,3381), but single-cell RNA conditions probably contribute to the poorer life expectancy of
sequencing showed that individual cells characteristic of differ- elderly patients (> 65 years) with glioblastoma . There are no
ent subtypes can be found within the same glloblastom.a and validated age cut-off points for clinical decisions that are based
that such expression subtypes may be related to malignant on the distinction between fit and frail patients; however. fit older
cell states reminiscent of neurodevelopmental cell types 12416, patients may benefit from chemoradiation 12476), and for frail
2226) . To date , gene expression profiles have not gained clear patients , MGMT promoter methylation status can influence the
significance in clinical diagnostics . choice between chemotherapy and radiotherapy alone /2046.
3430,3427).
Essential and desirable diagnostic criteria
See Box 2.03
Histopathology 17801, with IDH-wildtype and TERTpromoter-w1ldtype glroblas-
Histological features do not confer significant prognostic tomas tending to occur in younger patients and having more
information in IDH-wildtype glioblastoma, although necrosis frequent P13K pathway mutations (3448) . EGFR amplification
has been associated with shorter survival (199,412 ,1337). In
some studies . giant cell glioblastoma has been noted to have
and overexpression have been suggested as poor prognostic
factors in glioblastomas (1870,2961 ,1779], especially in highly
tf
a somewhat better prognosis than other types of glioblastoma amplified tumours (2175). but a consistent relationship between
(1731 .2333 ,2908) ; one study showed poorer prognosis for EGFR amplification (or EGFRvll I status) and survival has not
gliosarcoma than other gl ioblastomas 12960), whereas another been found in other studies (2309,377,138,1325.906). Allelic
showed no survival difference for gliosarcomas 1975), and most loss of 1Oq was associated with shorter survival in one report
studies of epithelioid glioblastoma have shown a poor progno- (2309); however, PTEN mutation did not correlate with progno-
sis (384 ,543 ,1653). Tumours with a primitive neuronal compo- sis in other studies (2309,2850 ,2961 ,3415,408).
nent, particularly those with MYC or MYCN amplification, may Molecularly targeted therapies for patients with glioblastoma
have a greater tendency for cerebrospinal fluid spread (2470). have been tested, but they have not yet yielded major suc-
cesses (1821) . Patients with BRAF p.V600E mutations may
Biomarkers respond to BRAF inhibitors (494,1544,34731. and there are
MGMT promoter methylation is an independent prognostic preliminary reports of patients with NTRK fusion-driven high-
marker for longer OS in glioblastoma (3415) and a strong predic- grade glioma responding to NTRK inhibitors (3615 ,1027). Earlier
tive marker for response to alkylating and methylating chemo- attempts to target EGFR or EGFRvlll did not prove effective, but
therapy (1279,3416.3429,3430,3094) . More than 90% of longer- they have resulted in lessons learned . For example, EGFRvlll
term surviving patients with glioblastoma have MGMT promoter may be lost in a substantial proportion of glioblastomas at recur-
methylation 19269895) versus only about 30% of the general rence irrespective of treatment, thus mandating target assess-
patient population with glioblastoma {3056). MGMT promoter ment at the time of Intended treatment (3414) . Targets should
methylation also correlates with longer progression-free survival be prospectively validated, so efforts need to be directed to the
and OS in elderly patients treated with temozolomide (3430, parallel development of accurate, reproduc ible, and feasible
1998). Limitations for the general im plementation of MGMT pro- tests (3428,67) . And, as mentioned . a general challenge 1n the
moter methylation as a standardized test include variability of development of molecularly targeted therapies is intratumoural
methods, determination of cut-off values, and reproducibility of and intertumoural heterogeneity {2416,25791. including plastic-
test results (3431). In add ition, there are groups of patients who ity over time.
seemingly benefit from treatment despite having a non-methyl- Tumour mutation burden serves as a biomarker in some solid
ated MGMT promoter 11279,2476) . Therefore, in some settings , cancers treated with checkpoint inhibitors , but it has not been
MGMT promoter methylation testing has been restricted to trials shown to be useful in patients with glioblastoma (2797,3222) .
that require stratification or inclusion of patients according to However, the hypermutant phenotype may be predictive specif-
the test result. ically for checkpoint inhibitor response in glioblastomas arising
TERT promoter mutation has been associated with more from genetically induced hypermutation (335) rather than the
aggressive behaviour in IDH-wildtype glioblastoma (3022, more common treatment-induced higher mutation load (32221 .

Gltomus . glior1euronLil tun ou rs. and neuronal tumours 55


Diffuse astrocytoma, Hawkins CE
Blumcke I
Jones DTW
Najm I
MYB- or MYBL 1-altered Capper D
Ellison OW
Rosenblum MK

Definition
Diffuse astrocy toma. MYB- or M YBU-a ltered, is a diffusely
infiltrative astroglial neoplasm composed of monomorphic cel ls
with genetic alterations in MYB or MYBL 1 (CNS WHO grade 1).

ICD-0 coding
9421/1 Diffuse astrocytoma, MYB- or MYBU-altered

ICD-11 coding
2AOO .OY & XH6PH6 Other specified gliomas of brain & Astro-
cytoma, NOS
Flg.2.41 Diffuse astrocytoma, MYB- or MYBL1-altered . MRI of a MYBL1-altered
diffuse astrocytoma located in the Inferior frontal gyrus in a 39-year-old patient with
Related terminology epilepsy since the age of 3 years. The tumour (white arrow) Is T1 -hypointense (lei~
Not recommended: isomorphic astrocytoma variant; isomor- T2-hyperintense (middle), and FLAIR-hyperintense (right).
phic diffuse glioma.
Localization
Subtype(s) Diffuse astrocytoma, MYB- or MYBU-altered , is most com·
Diffuse astrocytoma, M YB-altered ; diffuse astrocytoma, MYBL 1- manly a cerebral tumour with cortical and subc ortical com-
altered ponents. In 40 reported cases , the tumour was centred in the
temporal (42.5%), frontal (27.5%), occipital (20%) , and parietal

. . ... . . .·. . ·
,"". . _. . - ... . .. ' !:·.
... ..
' : '"
,. ,·,. l •
.. ·: . •. ··~
~
·. \

•'
.. .. '. '
,.. I o •

,, ....
.
:- : . ..
•'
,.
·. ...
..
·t . :
. ....

. ..'· .
: • I I

~.
~
:. "
f • )': •••
' I
. ' .
r •
t •.., .. .• •

.. . . "·
, .. \ ..
:. :
(10%) lobes 12953,304) . Rare brainstem cases have also been likely to be < 0.5% . Paediatric series each described< 10 cases
reported (2768] . (3142,3597,2616,2584,188]. with no clear sex predilection . The
largest series to date includes data on 20 patients with a MYB or
Clinical features MYBL1 alteration 13404!, with a median age of 29 years (range:
Patients typically present with drug-resistant epileptic seizures. 4-50 years) and a male preponderance (M :F ratio: 3·1).
which often have been present since childhood 12857,302 ,
3404). Therefore, this neoplasm belongs within the broad cat- Etiology
egory of long-term epilepsy- associated tumours 12953,3404 , Unknown
302] . In one series, 81% of patients with MYB- or MYBL1-altered
diffuse astrocytoma developed epilepsy during childhood , Pathogenesis
and the median age at onset of epilepsy was 1O years (range: MYB and Its closely related family member MYBL1 are tran-
1-35 years) (3404) . However, only 23% of patients had a resec- scriptional transactivators that are important for cell prolifera-
tion as children, with the median age at surgery being 29 years tion and are downregulated with cellular dif'ferentiation 1367]. In
and the median time to operation after epilepsy onset being gliomas and other cancers , the genes encoding these proteins
15 years. undergo structural rearrangements that result in truncation of
the C-terminal negative-regulatory domains of the proteins ,
Imaging which , In many cases, leads to their overexpression 1838]_
Diffuse astrocytoma, MYB- or MYBL 1-altered, is typically These truncated proteins are oncogenic {2616/.
hypointense on T1 , shows mixed signal or hyperintensity on
T2-FLAIR , is non-enhancing , and does not show restricted dif- Macroscopic appearance
fusion 1566,297,3404) . Tumours are mostly well defined , but These tumours are typically unencapsulated , soft to friable .
they may show diffuse growth patterns, at least focally j566, grey-white masses .
3404). Large cysts are occasionally observed 13404).
H istopathology
Epidemiology A proliferation of relatively monomorphic glial cells with bland .
Diffuse astrocytoma, MYB- or MYBL 1-altered, Is a rare tumour. round to ovoid or spindled nuclei , diffusely disposed in a fibril-
In a population-based series of paediatric low-grade gliomas, lar matrix or permeating neuropll , is characteristic {2584,566 ,
tumours with MYB or MYBL 1 alterations accounted for about 2% of 3404} . Frequently, tumour cells barely raise the normal cell
cases [2768), with the overall incidence among all brain tumours density of infiltrated parenchyma and may therefore be difficult

G l1ornas gl1oneu ro11 I lu rnours . d tlli nt'ur011.J.l lu111ours 57


Box 2.04 Diagnostic criteria for diffuse astrocytoma , MYB- or MYBL1-altered MYBL 1-altered diffuse astrocytoma from adult-type IDH-mutan
Essential: or IDH -wi ldtype di ffuse astrocytic gliomas , given the ir distinc1
Diffuse astrocytoma without histological features of anaplasia biological behaviour.
AND
No mutations in IDH or H3 genes
Cytology
Not clinically relevant
AND
Structural variant of MYB or MYBL 1 Diagnostic molecular pathology
OR Sequencing demonstrates a structural variation that results 1r
DNA methylation profile aligned with diffuse astrocytoma, MYB- or MYBL f- a fusion between MYB or MYBL 1 and a partner gene (838!
altered The most frequently reported partner genes are PCDHGA1
Desirable: MMP16, and MAML2 [3597,2584 ,3404 ,566} . MYB rarely part-
Absence of OLIG2 and MAP2 expression ners with OKI in this tumour ; a MYB::OKI fusion is typically
found in angiocentric glioma (566} . Diffuse astrocytoma, MYB-
or M YBU-altered, is IDH- and H3-wildtype. Although sequenc-
to recogn ize as neoplastic [566 ,3404}. A vague angiocentric ing determ ines the natu re of the gene fusion , other methods
polarity may be regionally evident, partic ularly in the MYB- (e.g. interphase FISH) can also demonstrate a rearrangemenl
altered subtype. Entrapped neurons often attest to the infiltra- of MYB or MYBL 1.
tive nature of these astrocytomas . Mitotic activity is typically
absent or low, and microvascular pro liferation and necrosis are Essential and desirable diagnostic criteria
not seen . Occasionally, c ases wi th more pleomorphic nuclei See Box 2.04.
and sl ightly elevated proliferati on are observed , especially in
paed iatric patients. Staging
Not applicable
lmmunohistochemistry
lmmunoreactivity for GFAP and low Ki-67 labell ing index values Prognosis and prediction
are typical. One stu dy reported that expression of MAP2 is lim- Available outcome data are limited but suggest a benign clini-
ited to residual neurons and neuronal processes and that tumour cal behaviour. In a series of 11 paed iatric patients, 9 had stable
cells are negative for OLIG2 and CD34 {3404}. IDH1 p.R132H is disease or no evidence of disease after a median fo llow-up o
negative , and nuclear ATRX expression is retained (3404}. 12 years (566} ; 1 patient requ ired surgery for recurrent disease
after 17 months, and another patient d ied of the disease. In a
Differential diagnosis second paediatric series of 16 patients with MYB- or MYBLl-
Some areas in ang iocentric gl iomas have the same archi- altered gliomas, all were alive after a median fo llow-up ol
tectural an d cytolog ical features that characterize MYB- or 6.2 years (2768}; 4 patients who did not receive an initial gross
MYBL1-altered diffuse astrocytoma , and these two tumour total resection required surgery for recurrence . In a series o
types are considered to have overlapping morphology. A his- 18 patients (predominantly adults) with a med ian follow-up o!
togenetic relationshi p between these tumour types is re inforced 2.5 years , only 1 patient required surgery for recurrence after
at the genetic level; practically all angiocentric gliomas have 3 years , and none died {3404}. Of patients with epil epsy, about
a rearrang ement of MYB, most commonly associated with a 90% became seizure-free after resection and the remainder
MYB::OKI fusion . It is more important to distinguish MYB- or had a reduction in seizure frequency {3404}.

1 1
..J
r
d I 11
-:..i
IC .). .J
<•i ii r1 r 1H~ ;rrH ii:i l lun 1nurs . ancJ 11 urm1c:il 1urnour!::l
Angiocentric glioma Ellison OW
Jones OTW
Ligon KL
Preusser WM
Rosenblum MK

Definition
Angiocentric glioma is a diffuse glioma composed mainly of
thin , cytologically bland , bipolar cells aggregating at least partly
in perivascular spaces . Almost all angiocentric gliomas have
a MYB :: OKI gene fusion , and the remainder generally have
another MYB alteration (CNS WHO grade 1).

ICD-0 coding
9431 /1 Ang iocentric glioma

ICD-11 coding
2AOO.OY & XH41C5 Other specified gliomas of brain & Angi -
ocentric glioma

Related terminology
Not recommended: angiocentric neuroepithelial tumour, mono-
morphous angiocentric glioma. Fig.2.44 Angiocentric glioma. T2-weighted MRI showing a mostly cortical parietal
lesion on the right side, with minimal mass effect.
Subtype(s)
None surrounding the tumour on T1-weighted MRI is present in some
cases. A stalk-like extension to the adjacent lateral ventricle and
Localization dystrophic calcification are other variable features (1705 ,103}.
Angiocentric gliomas are typically located in the cerebral cor-
tex , but the brainstem is an increasingly recognized site for this Epidemiology
tumour {646 ,3399 ,510,678 ,566). Population-based epidemiological data are not yet available for
this uncommon tumour. Most cases occur in children and young
Clinical features adults , with a median age of 13 years (range: 2-79 years) at pre-
Patients with angiocentric gliomas typically present with chronic sentation . No clear sex predilection is yet apparent, although
and intractable partial epilepsy. Headache and visual impair- slightly more male patients than female patients with angiocen-
ment are other commonly reported symptoms {2953] . tric glioma are described in the literature {103] .

Imaging Etiology
In the cerebral cortex, angiocentric gliomas are commonly The vast majority of ang iocentric gliomas are sporadic and have
located in the temporal or frontal lobes. Brainstem examples not been associated with any specific risk factors . Only single
have also been reported {566]. On MRI , these tumours are often cases have been reported in association with neurofibromato-
well-circumsc ribed , non- contrast-enhancing, and hyperintense sis type 1 and Koolen-de Vries syndrome 12184,1792}, so it is
on T2-weighted and FLAIR images. A rim-like hyperintensity unclear whether these are merely coincidental.

Fig. 2.45 Ang1ocentric glioma. A Elongated cells with bipolar cytoplasmic processes are found in most angiocentric gliomas, particularly in solid areas of the tumour. I Note
both the pronounced angiocentric pattern and the entrapped neurons. C Entrapped neurons are present among tumour cells that show a syncytial pattern.

G l1 omas. gl1on u1ona l lum 0urs . Gncl neu1 onell tU1nours 59


.. ··· !'lllr"l~..-

· ·~-"'· 1· - ~~E
Fig. 2.46 Anglocentrlc glioma. A The perivascular tumour cells are GFAP-immunoreactive. B Dot-like or ring-like immunoreactivlty for EMA is variably present across regions
of most angiocentric gliomas. C Unlike in most low-grade astrocytomas, the tumour cells are predominantly OLIG2-negative, although there are many entrapped immunoreactive
ollgodendrocytes. D NFP staining highlights numerous entrapped axons, consistent with an infiltrative growth pattern .

Pathogenesis produce induration 13377}, or they may be soft and gelatinous


Practically all angiocentric gliomas harbour rearrangements of {2887). Cystic changes may occur and solid components have
the MYB transcription factor gene at the 6q23.3 locus. MYB is been described as tan-grey in colour {2247).
most commonly fused with OKI, in association with deletion of
the intervening region . More rarely, MYB is fused with ESR1, Histopathology
PCOHGA1 , or other genes {3597,2616,2584,2768}. or it is Typically, angiocentric gliomas have cytologically uniform.
amplified /3142) . Angiocentric gliomas typically lack any other bipolar spindle cells with slender nuclei and granular chromatin
oncogenic mutations or copy-number alterations 1396,3597, oriented around blood vessels of every calibre; these cells are
2247], although two cases have been reported to have both a radially arrayed in a rosette-like pattern or form monolayered or
MYB:: OKI fusion and a BRAFp.V600E mutation 12584). In DNA multilayered sleeves lengthwise along vascular axes. Spindle
methylation profiling studies, angiocentric gliomas cluster with cell elements can permeate the parenchyma extensively and
or close to other tumours that contain MYB alterations, such as at variable density, fashion compact nodules of schwannoma-
paediatric diffuse astrocytomas and diffuse astrocytoma, MYB- like appearance, be disposed in tight and intersecting fascicles.
or MYBU-altered /2584 ,3404 ,566). and aggregate beneath the pial surface in horizontal streams or
MYB :: OKI is oncogenic and has been proposed to drive in a perpendicular and palisading alignment. Myxoid and micro-
tumorigenesis through simultaneous deletion of a negative- cystic alterations may be conspicuous. Entrapped neurons
regulatory domain in MYB, functional deletion .of the tum?ur are not conspicuously dysmorphic 13377). Some angiocentnc
suppressor OKI, and enhancer translocation forcing expr~ss~on gliomas harbour epithelioid elements that may contain rounded
of the constitutively active MYB::OKI allele {188) . MYB act1vat1on paranuclear structures with an internal granular stippling {2134).
appears to drive MAPK signalling , in common with .almost a.II which correspond to EMA-immunoreactive microlumina of th0
paediatric low-grade gliomas; one study showed higher ac~1- type seen In ependymomas and demonstrated in angiocentric
vation of the MAPK pathway in MYB-altered tumours than 1n gliomas at the ultrastructural level {3377). Although histolog1·
other low-grade gliomas, including those with a BRAF p.V600E cally typical examples with apparent mitotic activity occur (2132.
1873), mitoses are sparse in most cases, and neither mlcrovas-
mutation 12768).
cular proliferation nor necrosis is seen . Increased prolifer t1ve
activity and other anaplastic features have been reported rarely.
Macroscopic appearance .
Ang iocentric gliomas expand involved structures , with blur- but the clin ical significance of such findings is uncertain pa73.
ring of the cortical grey matter- white matter junction. They may 2051 .1883).
/mmunophenotype Box2.05 Diagnostic criteria for angiocentric glioma
On immunohistochemlcal study, GFAP expression is the rule ,
Essential:
and a near-constant finding is at least regional EMA immu-
Glloma with diffuse growth architecture and a focal angiocentric pattern
' nolabelling of the cytoplasm in a dot-like or ring-like (microlu-
men) pattern !3377,1852,2247) . Membranous EMA expression AND
characterizes some epithelioid cells in perivascular and subpial Monornorphic spindled cells with irnmunophenotypic and/or ultrastructural evidence
.. locations . Tumour cells do not label for neuronal markers and of astrocytic and ependymal differentiation
are, in contrast to most other low-grade astrocytomas, predomi- Desirable:
nantly OLIG2-negative. Ki-67 immunolabelling Index values are Lack of anaplastic features
generally < 5% (frequently < 1%), but a labelling index of 10% Alteration of MYB
was found in an otherwise typical lesion that had not recurred DNA methylation profile aligned with diffuse glioma, MYB- or MYBL 1-altered
6 years after surgery (1873).

Cytology
Not clin ically relevant with a relatively high frequency of MYB alterations is paediatric
diffuse astrocytoma, MYB- or MYBU-altered .
Diagnostic molecular pathology
Practically all angiocentric gliomas show rearrangements and/ EssentiaJ and desirable diagnostic criteria
or copy-number alterations , including deletion or amplifica- See Box 2.05.
tion (3142}. at the MYB locus on 6q23 .3 (2616,2584}. Most
rearrangements involve fusions between the MYB and OKI Staging
genes !566). In data taken from several studies , a MYB altera- Not applicable
tion was found in 73 (99%) of 74 angiocentric gliomas (3142,
2584,188,510 ,678 ,566) . Where appropriate analyses could be Prognosis and prediction
undertaken, a MYB:: OK/fusion was demonstrated in 41 (87%) o'f Angiocentric gliomas usually have an indolent behaviour and
47 tumours from these series . Rarely, MYB has been shown to are radiologically stable (2887,566). In most cases , gross total
fuse with several other genes, such as PCOHGA 1 (566}. Angi- resection can be achieved and is curative. Postoperative com-
ocentric gliomas lack mutations in TP53, ATRX, IDH1 , IDH2, plications and tumour recurrence are uncommon (103}. There
and histone H3 genes (2603,2584,566}. Another CNS tumour are no known prognostic or predictive factors .

Gliomas , glioneuronal tumours , ,. nd neuronal tumours 61


Rosenblum MK
Polymorphous low-grade neuroepithelial Bl umcke I
Ellison OW
tumour of the young Huse JT

Definition the temporal lobes , mostly on the right side and with frequenl
Polymorphous low-grade neuroep ithelial tumour of the young involvement of medial/posteroinferior structures (1384,1485,
(PLNTY ) is an indolent cerebral neoplasm characterized by a 549). Other reported locations include the frontal , parietal , and
strong association with seizures in young individuals , diffuse occipital lobes as well as the third ventricular region (1485).
growth patterns , frequent presence of ol igodendroglioma-
like components , calc ification , CD34 immunoreactivity, and Clinical features
MAPK pathway- activating genetic abnormalities (CNS WHO PLNTYs typically cause seizures and are associated in many
grade 1). cases with refractory epilepsy (particularly partial complex epi-
lepsy), but they can also cause headache or dizziness {1384,
ICD-0 coding 288,2679 ,1193,549). On neuroimaging , PLNTYs often have cys-
9413/0 Polymorphous low-grade neuroepithellal tumour of the tic, as well as solid , components , and they are often densely
young calcified on CT (1384 ,1485,549). PLNTYs are FLAIR-hypenn-
tense on MRI , often displaying signal heterogeneity, with cal-
ICD-11 coding cified regions appearing T1/T2-hypointense, and non-calcified
2A00 .2Y Other specified tumours of neuroepithelial tissue of components appearing T2-hyperintense with variable T1 signal
brain intensity (1384,.1485 ,549} . Patchy or nodular contrast enhance-
ment is observed in a minority of cases, but there is no substan-
Related terminology tial oedema or mass effect.
Not recommended: diffuse glioneuronal tumour (74}; diffuse or
nonspecific form of dysembryoplastic neuroepithelial tumour; Epidemiology
massively calcified low-grade glioma (1189,1302) . Population-based incidence data are unavailable. PLNTYs have
been reported In patients ranging in age from 4 to 57 years
Polymorphous low-grade neuroepithelial tumours of the young [1384,2679,549,1485), with most occurring in the second and
have also been described under the generic designation of third decades of life (median age at diagnosis: 16 years). There
"long-term epilepsy-associated tumour" (3177,297} . is no clear sex predilection .

Subtype(s) Etiology
None Factors predisposing to the development of PLNTYs are
unknown. An isolated example has been associated with germ-
Localization line ATM mutation [3075) .
PLNTYs are cerebral tumours that usually have cortical and
subcortical components. Approximately 80% have involved Pathogenesis
Somatic MAPK pathway-activating genetic events (particu-
larly BRAF mutations and FGFR fusions) clearly play a role in
the development of PLNTYs, with the tumours' aberrant CD34
expression possibly reflecting an origin from developmentally
dysregulated neural precursors (300,301,1384}. The specific
mechanisms by which these genetic alterations contribute to
the pathogenesis of PLNTYs are not clear.

Macroscopic appearance
PLNTYs have been described as unencapsulated, soft to frl·
able, grey-white masses that are indistinctly demarcated fro(l'l
normal brain (2679,549) .

Hlstopathology
PLNTYs exhibit both infiltrative and compact growth patterns.
Usually present and often dominant are oligodendroglioma-1iK8
components , which range from elements having uniformly small
and round nuclei with perinuclear haloes to populations exhibit·
Flg.2.47 Polymorphous low grade neuro0p1tliellal tumour ol Ille young . FLAIR -~1y­
ing obvious variation in nuclear size and shape with wrinkled or
perintr::1t&e solid cumponent 5 . cy 511c changer. , and tlt e absence of mass effect are
grooved nuclear membranes ancJ intranuclear pseudomclus1ons
sc:i:n 1r1 ll11s MRI

f:,;.; ( , r1, r, 1 , , I , ,1,, , 11 1 .1 , !I 11JI111 ,, JI',. H 11 J li t t JI 1 II r, ti l t 11111 11 Jr ',


11384.2679 ,1485,549) . PLNTYs may also contain elements of labelling index values are generally low (~ 1- 2%). but higher
astrocyt1c or morphologically ambiguous appearance, the for- values (up to 5%) have been reported !1384)
mer including fibrillary, spindled , and pleomorphic forms . Some
examples display subtle perivascular pseudorosettes . Calcifi- Differential diagnosis
cations are present in the large majority and may be coarse PLNTYs may be histological!y indistinguishable from oligoden-
and confluent. Generally absent are foci of necrosis, micro- drogliomas or diffuse astrocytomas, but the latter are gener-
vascular proliferation, gemistocytic elements, myxoid micro- ally C0 34-negative (whereas PLNTYs are CD34-positive) and
cysts . neurocytic rosettes, and Rosenthal fibres. Eosinoph illc PLNTYs do not manifest IDH mutations or 1p/19q codeletion
granular bodies are only occasionally present [3075) . Patently like the other tu mour types, and they display MAPK pathway-
neoplastic neuronal compo nents are typ ically not in evidence, act ivating genetic abnormalities typically foreign to the adult-
although modestly dysmorphic neurons in small numbers may type Infi ltrating gliomas (see Diagnostic molecular pathology).
be encountered . The nosolog ical position of neoplasms oth - An ol igodendrog lioma-like entity closely resembling PLNTY
erwise qualifying as PLNTYs but having nodular ganglion cell in morphology, immunophenotype, clinical presentation, and
aggregates is unclear. PLNTYs characteristically manifest little, demonstration of BRAF p.V600E mutations, but manifesting
if any. mitotic activity. However, one study described an exam - chromosomal instability, has been described and could rep-
ple with increased mitoti c activity and illustrated an unusually resent a PLNTY subtype (1003}. Also resembling PLNTYs at
pleomorphic case with atypical mitoses (1485) . t11e morphological level, potentially expressing C034 and (like
select PLNTYs) exhibiting FGFR3::TACC3 gene fusions (see
/mmunophenotype Diagnostic molecular pathology), is a group of aggressive
On immunohistochem ical assessment, PLNTYs exhibit expres- gliomas that principally arise in middle-aged and older adults,
sion of GFAP (th is may be reg ional or generalized), OLIG2, and generally harbour high-grade components that would quality
CD34 11384,2679,1485,549) ; the last may be patchy, or diffuse as anaplastic astrocytoma or gl ioblastoma , and display copy-
and inte nse. CD34 labelling is displayed both by tumou r cells and number abnormalities and other genetic al terations associated
by ramified neural elements in the associated cerebral cortex. with IDH-wildtype glioblastomas {276).
There is retained tumour cell expression of ATRX but no label-
ling of IDH1 p.R132H . PLNTYs are typically negative for EMA, Cytology
synaptophysin, chromogranin , NeuN , and HuC/HuD. lmmu no- PLNTYs cannot be identified on cytological grounds alone.
reactivity for BRAF p.V600E may be encountered , reflecting In smear preparations, tumour cells may appear as rounded
the presence of these mutations in a subset of PLNTYs. Ki-67 nuclei devoid of cytoplasm , or they may exhibit irregular nuclear


Fig. 2.48 Polymorphous low-grade neuroepllhelial tumour of the young. A Oligodendrogljoma-like features include small round nuclei, perinuclear haloes, and delicate branch-
ing capillaries. B Mild nuclear pleomorphism and membrane irregularities are common. C Astroglial elements here exhibit spindling and more conspicuous atypia without mitotic
activity. D Vascular mineralization was evident in this example. E A diffuse growth pattern and conspicuous calcification are seen.
contours and cytoplasmic processes . The latter may anchor Box2.06 Diagnostic criteria for polymorphous low-grade neuroepithelial tumour of the
tumour cells to capillaries in a pseudorosette fashion {1384}. young
Essential:
Diagnostic molecular pathology
Diffuse growth pattern (at least regionally)
PLNTYs are consistently associated with MAPK pathway-acti-
AND
vating abnormalities, which must be demonstrated for confident
diagnosis. These specifically include BRAF p.V600E mutations Oligodendroglioma-like components (although these may be minor)
[1384,288 ,1485,1193,3075}, as well as fusions involving FGFR2 AND
or FGFR3 {1384,2679 ,1485,3075 ,549}. One such fusion, Few (if any) mitotic figures
FGFR2:: CTNNA3, has yet to be reported in any other CNS AND
neoplasm [180}, whereas others, including FGFR2:: SHTN1 Regional CD34 expression by tumour cells and by ramified neural cells in associated
(KIAA1598) , FGFR2:: 1NA , and FGFR3:: TACC3, are also cerebral cortex
encountered in various other entities. One FGFR3::TACC3- AND
fused PLNTY also manifested low-level FGFR3 amplification, IDH-wlldtype status
but whether these changes involved the same allele was not AND
clear [549) . A partial duplication of NTRK2 was detected in a
Unequivocal expression of BRAF p.V600E on immunohistochemical assessment
single case {1485}. PLNTYs do not harbour IDH or ATRXmuta-
OR
tions, do not exhibit 1p/19q codeletion , and have a distinct
DNA methylation profile most closely aligned to that of gangli- Molecular diagnostic evidence of BRAF p. V600E mutations, FGFR2 or FGFR3
fusions, or potentially other MAPK pathway-<lriving genetic abnormalities
oglioma {1384) .
Desirable:
Essential and desirable diagnostic criteria Conspicuous calcification (characteristic, although not constant)
See Box 2.06 . Absence of 1p/19q codeletion

Staging
Not applicable as progressive at 60 months after complete resection occurred
in the setting of germline ATM mutation and displayed unusually
Prognosis and prediction complex copy-number abnormalities {3075}, whereas an exam·
A series providing extended follow-up data suggests that pie displaying FGFR3: :TACC3fusion coupled with uncharacter·
PLNTYs generally behave in CNS WHO grade 1 fashion and istic somatic mutations involving TP53, ATRX, PTEN, and TEK
are amenable to control by excision {1384). At postopera- (as well as RB1 mutation in a recurrence) underwent malignant
tive intervals of 12- 89 months (mean: 47 months), only 1 of transformation to glioblastoma-like histology {182}. Additional
9 patients had evidence of possible local recurrence after gross case identification and follow-up are required to determine the
total resection . Tumour removal effected relief from seizures or long-term risk of recurrence and biological progression associ·
reduced seizure frequency in most cases. One PLNTY reported ated with these neoplasms.

·1·
Diffuse low-grade glioma, Jacques TS
Capper D
MAPK pathway- altered Giannini C
Orr BA
Tabori U

Definition Clinical features


Diffuse low-grade glioma, MAPK pathway-altered, is a low- Symptoms and signs depend on location . Epilepsy, sometimes
grade glioma with diffuse astrocytic or oligodendroglial mor- longstanding, is common . Location-specific neurological defi-
phology that generally occurs in childhood and is characterized cits generally combine with more nonspecific manifestations
by a pathogenic alteration in a gene that codes for a MAPK of raised intracranial pressure. More detailed clinical data for
pathway protein . Typically, these tumours have an Internal tan- these molecularly defined tumours are currently unavailable.
dem duplication (ITD) or mutation in the tyrosine kinase domain
(TKO) of FGFR1 , or a BRAF p.V600E mutation. The tumour is Imaging
IDH-wildtype and H3-wildtype and does not have a homozy- On neuroimaging, diffuse low-grade gliomas may have a more
gous deletion of CDKN2A. diffuse picture with T2-FLAIR than do pilocytic astrocytomas,
but both morphologies often appear as heterogeneously
ICD-0 coding enhancing masses with cystic elements .
9421/1 Diffuse low-grade glioma, MAPK pathway-altered
Epidemiology
ICD-11 coding There are no epidemiological studies of this specific tumour
2AOO .OY Other specified gliomas of brain type. However, it Is clearly rare . In a series of 843 patients aged
< 19 years with low-grade gliomas and without neurofibroma-
Related terminology tosis type 1 (NF1), 5.9% of tumours had the morphology of a
Not recommended: diffuse astrocytoma; diffuse astrocytoma, diffuse astrocytoma and 3.0% had that of an oligodendroglioma
IDH-wildtype; oligodendroglioma; paediatric-type oligoden- {2768}. Reports of this tumour have, by definition, focused on
droglioma. tumours affecting children .

Subtype(s) Etiology
Diffuse low-grade glioma, FGFR1 tyrosine kinase domain- There are no definite causative factors . However, there are
duplicated; diffuse low-grade glioma, FGFR1-mutant; diffuse descriptions of diffuse low-grade astrocytomas in patients with
low-grade glioma, BRAF p.V600E-mutant NF1 !2704}, raising the possibility that NF1-associated gliomas
may include tumours within the spectrum of MAPK pathway-
Localization altered diffuse low-grade glioma.
MAPK pathway-altered diffuse low-grade gliomas are described
throughout the craniospinal axis , particularly the cerebral hemi- Pathogenesis
spheres . Among paediatric low-grade gliomas, specific altera- By definition, these tumours harbour an abnormality in the
tions are more common in specific locations {2768), raising the MAPK pathway, are IDH-wildtype and H3-wildtype, and do not
possibility that there may be regional differences in subtypes. show homozygous deletion of CDKN2A 1838}. In the context

Fin 2 so D'ff . MAPK 1 It ed Diffuse low-grade glioma In a 1-year-old girl. MRI shows a mass within the medial aspect of the right temporal lobe,
•· • 1 use 1ow-grade gl1oma pat iway- a er T · h d
with low Tl signal (A). high T2 signa'I (BJ, and heterogeneous enhancement with a cystic element on postcontrast 1-we1g te imaging (C).

Cl1 u 1 11 c1 ~ . CJl1ur1euro11 <J I t u 111 ,11 w ~ . :11 1d r1e ~ 11,11 1 ~ i l tu1n u r ~_; 65
I -

Flg.2.51 Diffuse low-grade glioma, MAPK pathway-altered. A low-grade diffuse glioma ot moderate cellularity (A) entrapping normal neurons (B) and showing subpial aggrega-
tion (C) typical of diffuse gliomas. The tumour cells have a bland appearance and are mildly atypical (B,D).

of paediatric diffuse low-grade gliomas, the MAPK alteration is Diffuse /ow-grade glioma, FGFR1 TKO-duplicated
most likely to be a BRAF p.V600E mutation or an alteration of or FGFR1-mutant
FGFR1 (duplication of the TKO, one or sometimes two slngle- Diffuse low-grade gliomas with FGFR1 alterations typically have
nucleotide variants, or a fusion gene) (3597,2584,2768}. the morphological features of an oligodendroglioma [25841.
Molecular alterations appear to associate with morphology; although astrocytomas can also have this genetic profile. A
for example. alterations of FGFR1 are frequent in oligoden- nodular architecture is occasionally present, and there is some
drogliomas but less so in diffuse astrocytomas (2584,2768}. overlap with the morphology of dysembryoplastlc neuroeplthe·
However, aside from being diffuse low-grade gliomas , these llal tumour, which shares these genetic alterations (2584,2768).
MAPK pathway-altered tumours are not defined by a distinctive Mitotic activity is rare or absent, and there is no microvascular
morphology or by a single DNA methylation or gene expression proliferation or necrosis. These tumours are diffusely immu·
profile. Jn future , it is likely that a comprehensive analysis of this nopositive for OLIG2 and show variable expression of GFAP.
rare group of tumours will resolve their status and clinicopatho- lmmunoreactivity for CD34 is typically restricted to a few cells.
unlike the widespread expression evident in PLNTY.
logical associations.
The differential diagnosis for diffuse low-grade glioma with
FGFR1 alterations includes dysembryoplastic neuroepitheiial
Macroscopic appearance . . tumour, PLNTY, and adult-type !DH-mutant oligodendroglioma.
Specific macroscopic appearances have not been described in
Dysembryoplastic neuroepithelial tumours can only be distin·
the literature. However, these tumours are likely to have features
guished by the presence of their defining specific glloneuronal
similar to those of other (I DH-mutant) diffuse low-grade gllomas .
element and , in complex cases , glial nodules. PLNTYs have a
distinctive cytology and show widespread strong expression
Histopathology . . of CD34. Adult-type oligodendroglioma is distinguished by the
MAPK pathway- altered diffuse low-grade glloma~ typ1call~ presence of an IDH mutation combined with 1p/19q codeletion.
have a bland appearance, being composed of mildly atypi-
cal glial cells at relatively low density and entrapped ~ormal Diffuse /ow-grade glioma, BRAF p.V600E- mutant
parenchyma . The pattern of infiltration is not as extensi.ve .as Diffuse low-grade gliomas with a BRAF p.V600E mutation are
. mas · it Is more in line
seen in some IDH-mutant low-gra d.e g I10 . · . . and composed of well-differentiated glial cells with bland , ovoid
with the degree of infiltration found 1n ang1ocentnc gliomas to spindle-shaped nuclei and fine fibrillary processes. Neither
polymorphous low-grade neuroepitheli.al tumour .of the Y~~~ Rosenthal fibres nor eosinophilic granular bodies are typicallY
(PLNTY) ' yet exceeds the peripheral dincorporation

of no .
s such as p1 1o-
found . Subpial aggregation of tumour cells , a typical seconctarY
cells sometimes found in other low-gra e 9 1oma · structure of diffuse gliomas , may be present. Mitotic activity is
cyt1c astrocytoma and pleomorphic xanthoastrocytoma .
s
y 11 ur it:L H G I 1al 1urnour. · 31 1cJ 11eu ronal tumour~
66
B .... I • • ,I
~ .
Rg. 2.52 Dlff use low-grade glioma, MAPK pathway-altered. Temporal lobe tumour In a 7-year-old patient. A Mildly atypical neoplastic gllal cells with an astrocytic or oligoden-
drocytic morphology are diffusely distributed In cerebral cortex and underlying white matter. Nodule formation is rare. An FGFR1 tyrosine kinase domain (TKO) duplication was
detected. B A flbrlllary matrix is found In some regions.

~ ­
Rg.2.53 Diffuse low-grade glioma, MAPK pathway-altered. Neoplastic astrocytic glioma cells show immunopositlvity for GFAP (A), OLIG2 (B), and the mutant protein BRAF
p.V600E (C).

rare or absent, and there is no microvascular proliferation or Diagnostic molecular pathology


necrosis. Tumour cells are typically immunoposltive for OLIG2 The diagnosis requires evidence of a MAPK pathway alteration
and GFAP, which highlights the fine cell processes . lmmunore- In the absence of an IDH or 1-13 p.K28M (K27M) mutation and
act1vity for BRAF p.V600E illustrates the BRAF mutation. without homozygous deletion of CDKN2A. The most commonly
The differential diagnosis of diffuse low-grade glioma with a observed genetic alterations in these tumours occur in BRAF
BRAF p.V600E mutation includes pilocytic astrocytoma, gan- (generally a p.V600E mutation) and in FGFR1 (TKO duplication,
glioglioma, and pleomorphic xanthoastrocytoma. In addition, one or sometimes two single-nucleotide variants, or a fusion
it is important to exclude IDH-mutant and H3 K27-altered dif- gene) (3597,2584,2768). Further rare alterations are seen in a
fuse midllne gliomas, which can have histopathological features variety of other MAPK genes, including FGFR2, NTRK1 , NTRK2,
identical to those of a BRAF p.V600E-mutant diffuse low-grade NTRK3, MAP2K1, and MET (2768). Although there is emerging
glioma. evidence that they may also result in MAPK pathway activation
Despite its diffuse nature, this tumour type appears to have {2768), diffuse astrocytomas with MYB or MYBL 1 alterations are
a better outcome than do CNS WHO grade 2 IDH-mutant dif- listed separately under the family of paediatric-type diffuse low-
fuse gliomas (see Prognosis and prediction, below), and a CNS grade gllomas.
WHO grade has yet to be assigned . DNA methylation profiling has not demonstrated a single cluster
for MAPK pathway-altered diffuse low-grade gliomas Instead,
Cytology some data suggest that they cluster with other tumour types that
The intraoperative cytological features of MAPK pathway- have shared genetic alterations l2584,460). Therefore, care is
altered diffuse low-grade gliomas have not been described, but required to exclude other tumour types with distinct morphologi-
these tumours are likely to show features of a low-grade astro- cal feaiures, and DNA methylation analysis can be helpful when
cytoma or oligodendrogl1oma . histopathological examination is difficult or inconclusive (463).

Gl1 un 1' s gl1oneuronal tumours dnd neur . n::il turnolirs 67


Box2.07 Diagnostic criteria for diffuse low-grade glioma. MAPK pathway-altered Prognosis and prediction
Essential: It Is currently uncertain to what extent MAPK-altered diffuse
Diffuse glioma with absent or minimal mitotic activity and neither microvascular
low-grade gliomas, as a group, will resolve into distinct tumour
proliferati on nor necrosis types, and the factors that best predict their outcome wm
AND probably evolve as the molecular nosology of this concept is
resolved . Therefore, some caution is warranted when making
Genetic alteration in the MAPK pathway
clinical and prognostic correlations .
AND
Although these are low-grade tumours. outcome will depend
IDH-wildtype and H3-wildtype on location, morphology, and molecular alteration . IDH-wildtype
AND diffuse low-grade gliomas in children rarely undergo anaplastic
Absence of homozygous deletion of CDKN2A progression. However, some investigators have reported that a
Desirable: broad range of paediatric low-grade gliomas and glioneuronaJ
Onset in childhood, adolescence, or early adulthood tumours with a BRAF p.V600E mutation and COKN2A deletion
have a higher rate of progression and tendency for late transfor-
Absence of morphological features or DNA methylation profile suggestive of an
alternative tumour type in which FGFR or BRAF abnormalities occur mation 12127,1806).
The availability of novel targeted therapies for MAPK path-
way-altered gliomas may change the prognostic and pre-
Essential and desirable diagnostic criteria dictive value of the above alterations . Specifically, BRAF
See Box 2.07. p.V600E-mutant diffuse gliomas respond favourably to BRAF
inhibitors (1239). Diffuse gliomas with FGFR1 alterations and
Staging non-canonical BRAF mutations may also respond to MEK
Not clinically relevant inhibitors.

68 Gl1orna gltor 1euronal tumours , and neuronal tumou rs


Diffuse midline glioma, H3 K27- altered Varlet P
Baker SJ
Leske H
Orr BA
Ellison OW Solomon DA
Jabado N Suva ML
Jones C Warren KE
Jones DTW

Definition involvement in 40% of cases, as well as diffuse spread to involve


Diffuse midline glioma, H3 K27-altered, is an infi ltrative mid- the thalamus, cervical cord, and even the frontal lobe 1403).
line glioma with loss of H3 p.K28me3 (K27me3) and usually
either an H3 c .83A>T p.K28M (K27M) substitution in one of the Clinical features
histone H3 isoforms, aberrant overexpression of EZH IP, or an Most patients with DIPG present with a short medical history
EGFR mutation (CNS WHO grade 4) . (< 2 months), with the classic triad : cranial nerve palsy (82%),
long tract signs such as pyramidal tract impairment (51%), and
ICD-0 coding ataxia (62%) (1329). Common initial symptoms of thalamic
9385/3 Diffuse midline glioma , H3 K27-al tered DMG s include intracranial hypertension and motor or sensory
deficit (3026}.
ICD-11 coding
2AOO .OY & XH7692 Other specified gliomas of brain & Diffuse Imaging
midline glioma, H3 K27 M-m utant On MRI , DIPGs c lassically have their epicentre in the pons and
typ ically involve > 50% of its surface , often asymmetrically, with
Related terminology frequent encasement of th e basilar artery [3024}. There may be
Acceptable: diffuse intrinsi c pontine glioma. an exophytic component and/or infiltration into the midbrain ,
the cerebellar peduncles , and the cerebellar hemispheres .
Subtype(s) Thalam ic tumours may be un il ateral or bilateral, the latter being
Diffuse mi dl ine gli oma, H3 .3 K27-mutant; diffuse midline more frequent in the EGFR-mutant subtype {383}. The lesions
glioma, H3.1 or H3.2 K27- mutant; diffuse midline glioma, show T1 hypointensity or isointensity and T2 hyperintensity, but
H3-wildtype wi th EZHIP overexpression ; diffuse midline glioma, FLAIR sequences and contrast enhancement patterns can vary
EGFR-mutant considerably, with many tumours not being contrast-enhancing
(1083 ,2145) .
Localization
Paediatric diffuse midl ine gliomas (DMGs) are preferentially Epidemiology
located in the brainstem or pons (the latter making it a diffuse Epidemiological data remain scarce for this rec ently described
intrinsic pontine glioma [DIPG]), or they are bithalamlc , whereas entity. The incidence of DIPG is esti mated to be 0.54 cases per
DMG s in adolescents and adults predominantly arise unilater- 1 million person-years overall , and 2.32 cases per 1 million per-
ally in the thalamus , or in the spinal cord {2738 ,2101}. Localiza- son-years in people aged ~ 20 years , with no sex predilection
tion in other mid li ne sites , such as the pineal region , hypothala- {1329}. DIPG represents 10- 15% of all paed iatric brain tumours
mus , and cerebellum , is exceptional {1103,2101 ,2988}. Large and 75% of all paediatric brainstem tumours . Thalamic DMGs
autopsy-based studies of DI PG have described leptomeningeal are rarer, representing 1-5% of paediatric brain tumours (25%

Fig. 2.54 Diffuse midline gllomas, H3 K27-altered, in classic midline locations. A A ~l al MRI demonstrates a T2-bright lesion expanding the pons and encasing the basilar
artery. B MRI showing heterogeneous enhancemen t of a pontine lesion on a postgadolinium T1 sequence. C Axi al MRI shows a left unlthalam1c T2-bright lesion with associated
obstructive hydrocephalus. There is no peritumoural oedema. D Sagittal postgadolinium Tl sequence reveals an intramedullary tumour with heterogeneous annular enhance-
men1 .

Gl1ornas 0l10 11eLJl\Jl1cll tur11uurs ,\Ill! l1t'Llllll 1,11 iurn()~llS 69


of thalamic tumours) {2766) . Spinal DMGs represent about 40% acts as an endogenous H3 p.K28M (K27M) mimic {1448). The
of spinal astrncytomas in paediatric and adult series {502,1070) . marked decrease in H3 p.K28me3 (K27me3) is associated with
EGFR-mutant DMGs most often occur during childhood , with gene expression changes resulting from the release of the poised
a median patient age of 7-8 years {383,2145). In the paediat- state of bivalent promoters 11804,2937). In some studies, changes
ric population, the occurrence of H3.3 p.K28M (K27M)-mutant in the transcriptome are relatively modest {636,1010,1247).
DMGs and H3-wildtype DMGs with EZH/Poverexpression peaks
at about 7-8 years , whereas H3.1 or H3.2 p.K28M (K27M)-mutant Cell of origin and effects on differentiation
DMGs occur in younger patients (median patient age: -5 years) As PRC2 and its deposition of H3 p.K28me3 (K27me3) play
{1329,1976,483) . critical roles in maintaining cell fate , H3 p.K28M (K27M) can
enhance self-renewal and disrupt differentiation, with differing
Etiology effects in different progenitor populations {1804,2193,1010]
There is no known specific genetic susceptibility for DMG but, Some studies indicate that the cell of origin, which could be
exceptionally, DMGs may occur in the setting of a cancer pre- neural stem cells, adopts an oligodendrocyte precursor cell-
disposition syndrome such as Li-Fraumeni syndrome or mis- like transcriptome (932) .
match repair deficiency.
Cooperating mutations
Pathogenesis Somatic heterozygous H3 p.K28 (K27) mutations are invariably
Loss of H3 p.K28me3 (K27me3) maintained during the disease , and they are generally found
Despite representing only a small proportion (3-17%) of the total to be associated with collaborating mutations, which may also
cellular H3 pool , H3 p.K28 (K27) mutations occurring in either represent subclonal populations {2258,1327,3332] . These target
canonical (H3.1 or H3.2) or non-canonical (H3 .3) sequence canonical cancer-associated pathways have subtype-specific
variants result in widespread loss of H3 p.K28me3 (K27me3) enrichment and include p53 (TP53, PPM10, ATM), predorn1·
on the wildtype histone H3 11862,237,514,3308!. The inhibitory nantly in H3.3 p.K28M (K27M)-mutant and EGFR-mutant cases.
interaction between H3 p.K28M (K27M) - and, rarely, H3 p.K281 and P13K or MAPK (PIK3CA, PIK3R1, PTEN), largely in the H3.1
(K271) - and EZH2 (the methyltransferase catalytic subunit of or H3 .2 p.K28M (K27M) -mutant subtype. Co-occurrence ol
PRC2) probably drives this dominant negative effect 11862,2.37, H3 .3 p.K28M (K27M) and BRAF p.V600E mutations may be
1247,1519,1827,3014,3481). In H3-wildtype cases , PRC2 1nh1b1- observed 1404,957,3482,3150). Gain-of-function mutation and
t1on is tl1ought to be mediated by overexpresslon of EZHIP, which genetic amplification of growth factor receptors involved in

70 GlicJff1 t.b gl1on!:' 11runal tun 1uu rs . t..llld 11t:'urOndl tu1 11u ur s
.,• .

.
'1

..,,
-'~ ·
Fig. 2.56 Diffuse midline glioma, H3 K27-altered. A ponUne glioma showing infiltrative tumour cells associated with vascular proliferation (A), loss of H3 p.K28me3 (K27me3)
immunostaining in tumour cells with retention In endothelial cells (B), negative H3 p.K28M (K27M) lmmunoreactivity (C), and tumour cells with Intense nuclear staining for EZHIP
(internal negative controls represented by endothelial cells and residual neurons) (D).

brain development are common in p.K28M (K27M)-mutant sub- EGFR-mutant subtype, which is typically GFAP-positive but
types , and they are found in PDGFRA (hindbrain , diencephalon, may lack OLIG2 and SOX10. Neurofilament and synaptophysin
telencephalon), FGFR1 (diencephalon), and ACVR1 (hindbrain) . stains highlight the infiltrated neuropil in the background . but
they are negative in the tumour cells .
Macroscopic appearance The combination of H3 p.K28M (K27M) and H3 p.K28me3
Diffuse infiltration of the parenchyma by neoplastic cells and (K27me3) antibodies Is highly effective as a diagnostic aid .
related oedema causes enlargement and distortion of anatomi- Positive H3 p.K28M (K27M) nuclear staining (for the H3 K27-
cal structures, as well as softening and discolouration of tissues altered subtypes) in combination with the loss of nuclear H3
with haemorrhagic or necrotic zones (403}. p .K28me3 (K27me3) immunoreactivity enables the detection
of single tumour cells in infiltrating zones (1372,2988 ,3308).
Histopathology Although no H3 p.K28M (K27M) staining is observed in DMGs
DMGs diffusely infiltrate the CNS parenchyma, usually without with H3 p.K281 (K271) mutation or EZHIP overexpression , these
particular perivascular or perineuronal tropism. Most cells are cases can be recognized by the loss of nuclear H3 p.K28me3
small and monomorphic, but they can be polymorphous, show- (K27me3) immunostaining and should be further evaluated by
ing astrocytic, piloid, oligodendroglial, giant cell, undifferenti- molecular analyses (2384) . In addition , EZHIP (CXorf67) anti-
ated , or epithelioid cytology {2988}. Even though mitotic figures bodies are available that highlight EZHIP overexpression (483) ,
are frequent and microvascular proliferation and/or necrosis which Is usually absent in H3 p.K28M (K27M)-altered DMGs.
may be observed , these features are not required for diagnosis About 50% of cases show nuclear accumulation of p53, sug-
and they are not independent predictors of survival (404,484, gesting an underlying TP53 mutation, and 15% of cases show a
1935,403). Rosenthal fibres and eosinophilic granular bodies loss of nuclear ATRX expression (404,484).
are not typically encountered .
In EGFR-mutant DMGs , mitotic activity is often present, but Differential diagnosis
necrosis or microvascular proliferation Is rare. DMGs are con- Brainstem tumours constitute a heterogeneous group rang-
sidered CNS WHO grade 4, irrespective of the presence of ing from low-grade primary CNS tumours (e.g. pllocytic astro-
microvascular proliferation or necrosis. cytoma, ganglioglioma, and MYB- or MYBL 1-altered diffuse
astrocytoma) [25561 to high-grade tumours comprising H3
lmmunophenotype K27-altered DMG as well as H3-wildtype tumours (e .g. the
DMGs typically express OLIG2, MAP2, and S100, whereas diffuse paediatric-type high-grade glioma MYCN subtype.
1mmunoreactivity for GFAP is variable apart from in the atypical teratoid/rhabdoid tumour, and embryonal tumour with

Gl1omas, gl1oneuronal tumours , and neuronal tumours 71


•· •
••••
\
.. ...., .: ..
.... '.. ' . '•
.
.
• I _,
. ....
• •


..,
•.._ :-· E ~ -~
Fig. 2.57 Diffuse midline glioma, EGFR-mutant. A Axial FLAIR MRI of a bithalamic EGFR-mutant glioma in a child. B,C Histology of a bithalamic glioma harbouring a small
in-frame insertion in exon 20 of the EGFR gene, demonstrating a diffuse astrocytlc glioma. D lmmunohlstochemlstry for H3 p.K28me3 (K27me3) showing loss in the majority of
tumour cell nuclei in this bithalamic glloma with EGFR mutation that Is H3-wildtype. E lmmunohistochemistry for H3 p.K28M (K27M)-mutant protein is negative in the majority of
bithalamic gliomas with EGFR mutation.

multilayered rosettes) /3145 ,404] . This reinforces the role of EGFR alleles , and frequent KRAS p.G12R mutations , often with
biopsy in the accurate diagnosis of brainstem tumours. Various accompanying BRAFmutation or fusion {567} .
midline circumscribed glial or glioneuronal tumours , includ-
ing pilocytic astrocytomas /2556}, subependymomas /3528}, Cytology
and gangliogliomas {2368], have been described as having Not clinically relevant
the same H3 p.K28 (K27) mutations; although the H3 p.K28M
(K27M) mutation probably imparts a poorer prognosis in these Diagnostic molecular pathology
entiiies, they should not be given a diagnosis of H3 K27-altered Distinct patte~ns of biological features (methylation profiling.
DMG . In posterior fossa group A (PFA) ependymomas, H3 gene expression , co-segregating mutations) and clin ical cor-
p.K28 (K27) mutations are extremely rare {2556,2765}, but loss relations (age, location , outcome) suggest the presence of four
of H3 p.K28me3 (K27me3) or EZHIP overexpression occurs subtypes of DMGs that are defined by the driving oncohistone
/2384 ,1448); these tumou rs can be distinguished from DMGs alteration (483,485}: (1) H3.3 p.K28M (K27M)-mutant, (2) H3.1
on a morphological basis. or 3.2 p.K28M (K27M)-mutant, (3) H3-wildtype with EZHIP
Exceedingly rare, non-midline, cortical or hemispheric diffuse o~er~~pressi.on, an~ (4) ~GFR-mutant (see Pathogenesis). No
gliomas with H3 p.K28M (K27M) mutation have been described s1gnif1cant h1stolog1cal differences have been noted between
11976,1977,1927}. To date, the biology and prognosis for such these subtypes.
tumours remain unknown, and the current recommendation is For . the H3 K27- mutant subtypes, a somatic heterozygous
to report them as "diffuse hemispheric glioma with H3 p.K28M mutation In one of the genes encoding histone H3 variants leads
(K27M) mutation not elsewhere classified (NEC)". to the amino acid substitution of lysine (K) to methionine (M) or.
EGFR-mutant DMG must be distinguished from pilocytic rarely, isoleucine (I) at position 27 (as measured from the start ol
astrocytoma of the tectum (tectal glioma), which is an exophytic t~e processed H3, i.e. after t.he cleavage of the initiating methio·
tumour emanating from the tectum of the mldbrain into the nine) 1484). However, genomic sequencing reports may list these
posterior third ventricle . Tectal gliomas may occasionally show mutations as H3 p.K28M (K27M) or H3 p.K281 (K271). It is ex ep-
T2-FLAIR hypenntensity extending into the bilateral tha!ar:ni , tional for H3 p.K28M (K27M) or H3 p.K281 (K27 1) mutations to
mimicking bithalamic glioma . Tectal gliomas can ~e d1s_t1n - co-occur with mutations in IDH1 or IDH2 or with H3.3 p.G35R
guished from EGFR-mutant DMG on the basis of h1stolog1cal (G34R) or H3.3 p . G~5V (G34V) mutations . Similarly, COKN:!A
features and molecular signatures , with tectal gliomas typically and/or CDKN2B deletions. TERT promoter mutations, and MGMT
displaying intact H3 p.K28me3 (K27me3) staining , w1ldtype promoter methylat1on represent rare events in DMGs 11976,19771·

72 Gl1ur11<:.1~ 9liur1t;urlJl1dl tu111<HJIS 11r1U 11L'u1u11 c. tl t l11 1 1uur~


fGFR-mutant DMG is genetically defined by abnormalities Box2.08 Diagnostic criteria for diffuse midline glioma, H3 K27-altered
in the EGFR oncog ene on chromosome band 7p11.2 . Most Essential:
tumours harbour small in-frame insertions/du plications within
A diffuse glioma
exon 20, which encodes the intrace llular tyrosine ki nase domain
(TKO) , whereas others harbour missense mutations in exons AND
encoding parts of the extracell ular domain , most commonly Loss of H3 p.K28me3 {K27me3) (immunohistochemistry)
p.A289T or p.A289V. Rare instances of EGFR gene amplifica- AND
tion occurring in the absence of an identifiable mutation have Midiine location
also been reported 12145,2933]. AND
H3-wildtype DMGs with EZHIP overexpression represent the Presence of an H3 p.K28M (K27M) or p.K281{1<271) mutation (for H3 K27-
rarest subtype {483) . In addition to immunohistochemical analy- mutant subtypes)
ses , EZHIP overexpression can be measured by molecular OR
analyses , such as RN A expression microarrays. Presence of a pathogenic mutation or amplification of EGFR (for the EGFR-
mutant subtype)
Essential and desirable diagnostic criteria OR
See Box 2.08 .
Overexpression of EZHIP (for the H3-wildtype with EZHIP overexpression
subtype)
Staging
OR
Becau se of the diffusely infiltrative nature of this tumour, and the
Methylation profile of one of the subtypes of diffuse midline glioma
potential of leptomeningeal spread, imaging of the whole CNS
is recommended. Desirable:
Results from molecular analyses that enable discrimination of the H3.1 or H3.2
Prognosis and prediction p.K28 (K27)-mutant subtype from the H3.3 p.K28 (K27)-mutant subtype
Independently of the location , the prognosis of DMG is poor, with
a 2-year survival rate of < 10% {1976}. Surgical options are lim- TP53 mutations have been shown to be associated with radiore-
ited due to tumour location. Patients with DMGs harbouring either sistance (3419}. According to one study, the median survival for
an H3.1 or H3.2 p.K28M (K27M) mutation or showing EZHIP children with bithalamic gliomas with confirmed EGFR mutation
overexpression have slightly longer overall survival (16 months) was 10-14 months, with most children succumbing to their dis-
than do patients with DMGs that bear an H3.3 p.K28M (K27M) ease within 2 years (2145) . Historical outcomes have also been
mutation (11 months), respectively 1483,485,1329}. Age (< 3 or poor for children with either radiographically or histologically
> 10 years), longer symptom latency (> 24 weeks), and systemic diagnosed bithalamic gliomas (3546,761 ,2086,1177,2596,1206,
therapy at diagnosis are predictors of longer survival (1329}. 2607,1447,2382,2479,3026,2894,383,2264}.

Gl iornas . giioneuroria l tumours . and neuron:.:il tum ours 73


Diffuse hemispheric glioma , Korshunov A Rodriguez FJ
Solomon DA
Capper D
H3 G34-mutant Jones DTW Sturm D
Leske H Warren KE
Orr BA Weller M

Definition
Diffuse hemispheric glioma, H3 G34-mutant, is an infiltrative
glioma involving the cerebral hemispheres, with missense muta-
tion of the H3-3A gene that results in one of the following sub-
stitutions of the histone H3 protein: c.103G>A p.G35R (G34R),
c.103G>C p.G35R (G34R), or c .104G>T p.G35V (G34V) (CNS
WHO grade 4) .

ICD-0 coding
9385/3 Diffuse hemispheric glioma, H3 G34-mutant

ICD-11 coding
2AOO.OY Other specified gliomas of brain

Related terminology
Fig. 2.58 Diffuse hemispheric glioma, H3 G34-mutant. Palisading necrosis.
Not recommended: paediatric glioblastoma, H3.3 G34-mutant.

Subtype(s) temporal lobe, with occasional multifocal manifestation, includ-


None ing leptomeningeal dissemination. Necrosis, cystic changes,
haemorrhage, and calcifications can be observed (3320}.
Localization
Diffuse hemispheric glioma , H3 G34-mutant, arises in the cere- Epidemiology
bral hemispheres. Occasional spread to midline structures and H3 G34-mutant diffuse hemispheric gliomas typically affect
leptomeningeal dissemination have been observed (1713,1976}. adolescents and young adults (median age:15-19 years).
Some studies have indicated that these tumours account for
Clinical features approximately 16% of hemispheric paediatric high-grade
Patients develop clinical symptoms according to the anatomi- gliomas (1565,1976), whereas others , including unpublished
cal structures involved , including seizures and motor or sensory prospective studies, have indicated a lower incidence [2867}
deficits. There is a male predominance, with an M:F ratio of 1.4:1 (1713,
1976}.
Imaging . .
MRI characteristics of H3 G34-mutant diffuse hemispheric Etiology
glioma are similar to those of other high-grade non-midline There is no known specific genetic susceptibility for H3 G34-
giiomas . MRI typically reveals a contrast-e~hancing '.umour mutant diffuse hemispheric glioma. No risk factors have been
with mass effect in cortical areas , often involving the parietal or reported to date.
~
·~

Pathogenesis positivity, loss of ATRX expression, and nuclear accumulation


An acquired missense mutation in the H3-3A (H3F3A) gene, of p53 in the majority of tumour cells. A further notable feature
resulting in substitution at position p.G35 (G34) on the tail of the is OLIG2 negativity. GFAP expression is variable and can be
histone variant H3 .3, plays a key oncogenic role in the patho- negative, especially in cases with more primitive embryonal-like
genesis of this entity. Several studies indicate that replace- morphology {3060,1713) . The Ki-67 proliferation index is usu-
ment of the amino acid glycine (G) with arginine (R) or valine ally high. Mutation-specific antibodies against H3.3 p.G35R
(V) leads to steric hindrance and blocks the capacity of SETD2 (G34R)- and p.G35V (G34V)-mutant proteins are available
13523,3599,891 J and KDM2A 1554) to bind to the mutant his- {1856AJ. However, false negative immunoreactivity in H3 G34-
tone H3.3 tail. Impaired binding of these H3 p.K37 (K36) meth- mutant cases has been described {1097}.
ylation-modulating enzymes results in diminished levels of H3
p.K37me2 (K36me2) and H3 p.K37me3 (p.K36me3) on the Gra.dlng
mutant histone H3.3 tail. H3 G34-mutant diffuse hemispheric glioma corresponds to
Studies on H3.3 p.G35V (G34)-mutant cells demonstrate that CNS WHO grade 4, regardless of the presence or absence of
differential binding of H3 p.K37me3 (K36me3) induces a tran- necrosis or microvascular proliferation {1713).
scriptional reprogramming, recapitulating that of the develop-
ing forebrain, and causes prominent upregulation of the proto- Cytology
oncogene MYCN 1289). Not clinically relevant

Macroscopic appearance Diagnostic molecular pathology


Diffuse infiltration of the parenchyma generates enlargement The molecularly defining diagnostic criterion is a missense
and distortion of brain structures, as well as softening and dis- mutation replacing glycine (G) with arginine (R) (in > 94%
colouration with haemorrhagic and/or necrotic zones . of cases) or valine (V) (< 6%) at p.G35 (G34) of the histone
variant H3 .3, encoded by the H3-3A (H3F3A) gene: c.103G>A
Histopathology p .G35R (G34R), c.1 03G>C p.G35R (G34R), or c.104G>T
H3 G34-mutant diffuse hemispheric glioma typically has a p.G35V (G34V) 11857}. So far, the p.G35R (G34R) or p.G35V
glioblastoma-like pattern, characterized by a highly cellular, (G34V) substitution in diffuse hemispheric gliomas has been
infiltrative astrocytic tumour with brisk mitotic activity. Addi- exclusively identified in H3-3A and not in other histone genes
tionally, microvascular proliferation and/or necrosis are usually {1713,1976]. About 90% of tumours bear TP53 mutations and
seen , although the presence of these features is not required f~r approximately 95% have alterations in the ATRX gene 117131.
the diagnosis. In some cases, multinucleated and pleomorph1c The H3.3 p.G35 (G34) mutations in diffuse hemispheric glio-
cells can be observed . An alternative pattern resembles the mas are mutually exclusive with IDH1 or IOH2 and H3 p.K28M
morphology of CNS embryonal tumours, where tun:our cell~ are (K27M) or p.K281 (K271) mutations l3060}. DNA methylation
rather small and monomorphlc, with hyperchromat1c nuclei and patterns and gene expression signatures can be used to dif-
scant cytoplasm . Here, Homer Wright rosettes are occasio~­ ferentiate H3 G34-mutant diffuse hemispheric gliomas from
ally present, whereas microvascular proliferation and necrosis other entities. Although H3 G34-mutant diffuse hemispheric
are less prominent 11713] . Ganglion cell differentiation has also gliomas show widespread DNA hypomethylation, MGMT is
been reported in rare cases 1111]. often methylated 13060.1976,33621. So far, no differences
between p.G35R (G34R) and p.G35V (G34V) mutations have
been reported .
lmrnunophenotype
The typical immunohistochemical pattern of H3 . ~~4-mutant
diffu se hemispheric glioma comprises MAP2 pos1t1v1ty, FOXG1

Clturn~1~; . i;Jl 1011euror1ul t ur 1 1ou r ~ . <lr1d neui1..1ric:1I 1u111ou1s 75


Box2.09 Diagnostic criteria for diffuse hemispheric glioma, H3 G34-mutant Essential and desirable diagnostic criteria
Essential: See Box 2.09.
Cellular, Infiltrative glloma with mitotic activity
AND Staging
In addition to cerebral imaging , radiological investigation of the
H3.3 p.G35R (G34R) or p.G35V (G34V) mutation (H3-3A [H3F3A] c.103G>A,
c.103G>C, or c.104G>T)
spine should be performed .
AND
Prognosis and prediction
Hemispheric location
The prognosis for patients with H3 G34-mutant diffuse hem 1•
AND (for unresolved lesions) spheric glioma is poor, with a median progression-free survival
Methylatlon profile of diffuse hemispheric glioma, H3 G34--fnutant of 9 months and a median overall survival of 18-22 months /17 13.
Desirable: 1976). Patients experience mainly local recurrences , althoug
OLIG2 immunonegativity leptomeningeal dissemination can occur. MGMT methylatio
Loss of ATRX expression and the absence of oncogene amplifications (such as of PDG-
FRA, EGFR, COK4, MDM2, CDK6, CCND2, MYC, MYCN) may
Diffuse p53 immunopositivity
be associated with longer overall survival /1713}.

76 (.;liCifllo [ , q l1ur1 Uf Ulli.i l lu fllO LJr t> , ~ncJ I H ~ Uf(Jl1.1 I !lJ ill OLJ r ~;
Diffuse paediatric-type high-grade glioma, Ca pper D
Jones DTW
H3-wildtype and IDH-wildtype Tabori U
Varl et P

Definition
Diffuse paediatric-type high-grade glioma (pHGG) , H3-wildtype
and IDH -wildtype , is a diffuse glioma with histological features
of malignancy, typi cally occurring in children , adolescents , or
young adults , which is wildtype tor histone 1-13, IDH1 , and IDH2
(CNS WHO grade 4) .

ICD-0 coding
9385/3 Diffuse paediatric-type high-grade glioma, H3-wl ldtype
and IDH -wildtype

ICD-11 coding
2AOO .OY Other specified gliomas of brain

Related terminology Flg.2.61 Dilluse paediatric-type high-grade glioma, H3-w1ldtype and IDH-wildtype,
Not recommended: paediatric-type glioblastoma , H3-wildtype ; MYCN subtype. Axial T1 -weighted MRI demonstrating a well-circumscribed lesion in
paediatric glioblastoma, H3-wildtype; methylatlon class glio- the left insular/temporal region with relatively homogeneous contrast enhancement
blastoma midline (MC GBM MID) ; diffuse high-grade glioma and central cystic changes.
in childhood, H3-wildtype, group C (pHGG WT-C) ; methyla-
tion class glioblastoma RTK3 (MC GBM RTK 111); diffuse high- WT-B) ; methylation class glioblastoma MYCN (MC GBM
grade glioma in childhood, H3-wildtype, group B (pHGG MYCN).

'•
>"
c) .. ~
( -
.r

,.. ,,,
r.
.
c _, .. .I
,. • ..,_. ·-....- - , . ,.

Flg.2.62 Diffuse paediatric-type high-grade glioma, H3-wildtype and IDH-wildtype, MYCN subtype. A The tumour is highly ~ellular, reminiscent of embryonal morphology,
and it shows areas of sharp demarcation from the brain tissue. B In addition to having gltal areas, the tumour may show a prominent embryonal appearance. C GFAP may be
expressed only in scattered cells . o Neuronal markers (here, neurofilament) may also be expressed.

G liomas , Lll1011eu ronal rurnou1 s . ancl neu1011al tumours 77


Clinical features
Like with other high-grade gliomas . patients develop clin1c;1
symptoms according to the anatomical structures involved
Symptoms can include seizures and motor or sensory deficits

Imaging
The MRI characte ristics of pHGGs, H3 -wildtype and IDH-
wil dtype, are similar to those of other high-grade gliomas
MRI typically reveals a contrast-enhancing tumour with mass
effect. pHGG MYCN tumours may be better circumscribed
with only slight perilesi onal oedema and homogeneous contra5t
enh ancement (3146 ,3145). Imag ing c harac teristics for the other
subtypes have not been reported .

Epidemiology
Epidem iological data for pHGG , H3-wildtype and IDH-wildtype
. do not yet exist. In a cl inical trial of non-brainstem paediatnc
Fig. 2.63 Diffuse paediatric-type high-grade glioma, H3-wildtype and IDH-wildtype, high-grade gl iomas in chil dren aged 3- 18 years , 32 (-40%) of
RTK1 subtype, in Lynch syndrome. This glial tumour of a 10-year-old child shows nu- 74 tumours with DNA methylation data corresponded to pHGG
merous pleomorphic giant cells. Genetic analysis revealed a germline MSH2 mutation H3-wildtype and IDH -wildtype (1977}. The median age of
and a high tumour mutation burden. Methylation profiling indicated the RTK1 subtype. patients with pHGG , H3-wildtype and IDH-wildtype, is currently
lmmunohistochemistry showed MSH2/MSH6 loss in tumour cells, and retention In nor- unclear because published series have focused on paediatric
mal cells. compatible with Lynch syndrome. cases (1976 ,1723) and the occurrence in the adult population
may therefore be underestimated .

Etiology
Gliomas arising after therapeutic rad iation , or, as described
above, In the context of germline mismatch repair de'ficiency.
typically harbour molecular characteristics compatible with
pHGG , H3-wildtype and IDH-wildtype (1928) and are predomi-
nantly of the pHGG RTK1 molecular subtype.
Gliomas arising in the context of constitutional mismatch
repair deficiency syndrome (CMMRD), Lynch syndrome, or
Li- Fraumeni syndrome should be recogn ized as distinct from
spontaneously arising diffuse paediatric-type high-grade glio-
mas . For more details , see Chapter 14: Genetic tumour syn-
dromes involving the CNS.

Pathogenesis
It is currently unclear why tumours arising subsequent to cranial
irradiation and those occurring in the context of CMMRD and
Lynch syndrome predominantly belong to the pHGG RTK1 sub·
Fig. 2.64 Radiation-induced diffuse paediatric-type high-grade glioma, H3-wildtype
and IDH-wildtype, RTK1 subtype. In areas, the tumour may show extensive myxoid
type. It is likely that there are similarities in the cellular origins
of these tumours and those of sporadically arising pHGG RTK1
change
tumours .
pHGGs, H3-wildtype and IDH -wildtype, harbour somatic
Subtype(s)
Diffuse paediatric-type high-grade glioma RTK2 ; diffuse pae - alterations of know~ oncogenic drivers that are expected to
diatric-type high-grade glioma RTK1 ; diffuse paediatric -type play a. c e n~ral role 1~ pathogenesis. Frequently observed are
alteration s 1n TP53 (1n 30-50% of cases), in genes encoding
high-grade glioma MYC N
members of the RAS/MAPK and Pl3K pathways, in MYCN, and/
or in /02 1404,3059, 172 1,1976).
Localization
pHGGs. H3-wildtype and IDH -wil dtype, have been reported to
arise throughout the supratentorial brain , brains_tem, a~ d cer-
Macroscopic appearance
ebellum 11723) . The molecular subtypes have. slightly different o.iff u s~ infiltration of_the parenchyma causes enlargement and
d1 sto rt1 0 ~ of the brain structums, as well as softening and dis·
site predilections · pHGG RTK2 tumou rs mostly involve su~ raten­
colouration, with haemorrhagic/necrotic zones.
torial structures (in 96% of cases), pHGG RTK1 tumours 1nvo~ve
the supratentorial brain (in 82% of cases) an d lnfratentonal/
Histopathology
brainstem sites (in 18% of cases), and pHGG M YC ~ tumou rs
~li s t opath o l ogy typ ically .shows features of either a glioblastorna·
involve the supratentorial brain (in 86% of cases) and 1nfratento-
li ke mali gnant tumour (with rrntot1c activity, vascular proliferatiofl.
rial/bra1nstem structures (in 14% of cases) I1723).

78 •
I
12
! MYCN

08 ;!
o~
"
00

~-. :. . '. I ..
•• • •. I •..!
. '

--0."

f

I
!.
d
--0 8

- 1.2
I

Flg.2.65 Diffuse paediatric-type high-grade glioma, H3-wildtype and IDH-wlldtype, MYCN subtype. Copy-number profile calculated from DNA methylation data, showing a
MYCN amplification among other changes. Note that the adult-type +71-10 chromosomal pattern is not present.

and necrosis) or a primitive , undifferentiated morphology. Areas are more likely to be adult-type epithelioid glioblastomas 11976,
of glial differentiation and primitive differentiation can often be 1721). The chromosomal pattern of +7/-10 and/or EGFRv/l/muta-
found in the same specimen . In some cases, vascu lar prolifera- tions typical of adult-type glioblastoma are typically absent in
tion and necrosis may be absent. pHGG, H3-wildtype and IDH-wildtype (3036). Other differentials
For the pHGG MYCN subtype, a biphasic pattern with areas to consider include CNS embryonal tumours {3059) and , for pos-
of diffuse infiltration and highly cellular circumscribed nodules terior fossa tumours, medulloblastomas. Medulloblastomas are
has been described (3146). Tumours are often composed of typically negative for OLIG2, whereas most pHGGs, H3-wildtype
large cells with distinct nucleoli and may show a mix of spindle- and IDH-wildtype, express this marker. CNS embryonal tumours
shaped and epithelioid cells (3146}. should be considered less likely in older children and adults than
in young children, and molecular alterations compatible with this
lmmunophenotype diagnosis should be clearly demonstrated in these cases. In the
Data on the immunophenotype of pHGG, H3-wildtype and postirradiation setting, the possibility of a pHGG versus relapse
IDH-wildtype, have not been specifically reported. Most of an embryonal tumour should be considered 12493).
reported cases were identified molecularly among series of
paediatric glioblastomas or high-grade gliomas, and an immu- Cytology
nophenotype compatible with such a diagnosis can therefore Not relevant
be expected , including at least focal positivity for GFAP and/
or OLIG2 11976,1721). However, tumours of the pHGG MYCN Diagnostic molecular pathology
molecular subtype may be largely negative for glial markers and Initial testing should exclude alterations in histone H3 and in
instead express neuronal markers . All cases should have pre- IDH1 or IDH2. Alterations frequently encountered in these
served expression of H3 p.K28me3 (K27me3) (3146). tumours include POGFRA amplification or mutation , TP53
mutation, NF1 alterations, EGFR amplification or mutation , or
Differential diagnosis MYCN amplification. DNA methylation profiling is used for the
Depending on the age of the patient and the location of the identification of pHGG, H3-wildtype and IDH-wildtype, and it
tumour, a large number of differential diagnoses should be con- may be useful in guiding additional molecular testing 11723,
sidered . In particular, for infants, infant-type hemisphe~ic. glion:a 19~6,1 ·9~7}. Three molecular subgroups may be recognized by
and desmoplastic infantile ganglioglioma I desmoplast1c infantile their d1st1nct DNA methylation profiles or enrichment for molecu-
astrocytoma should be excluded; these typically harbour altera- lar alterations : pHGG RTK1, pHGG RTK2, and pHGG MYCN .
tions of BRAF or fusions involving NTRK genes, ROS1, ALK, or pHGG RTK1 is enriched for PDGFRA amplifications (- 33% of
MET H3 p.K28me3 (K27me3) is retained in pHGGs, H~-~ildty~e cases), pHGG RTK2 is enriched for EGFR amplifications (- 50%
and IDH -wildtype, distinguishing them from diffuse m1dllne gllo- of cases) and TERTpromoter mutations (-64% of cases) (17231.
mas. Tumours with glioblastoma morphology but the DNA m~th­ and pHGG MYCN is enriched for MYCNamplifications (-50% of
ylation profile of pleomorphic xanthoastrocytoma or a comb1~a­ cases) 1460,1723}. Tumours associated with CMMRD or Lynch
tion of BRAF mutation and COKN2A and/or COKN2B deletion syndrome are typically of the pHGG RTK1 subtype.

G lior 11 as . g li on urona l 1umours. un rieu run.:.il tumours 79


Essential and desirable diagnostic criteria Box2.10 Diagnostic criteria for diffuse paediatric-type high-grade glioma (pHGG), H.:
See Box 2.10. w1ldtype and IDH-wildtype
Essen tis/:
Staging A diffuse glioma with mitotic activity occurring in a child or young adult
Not relevant AND
Absence of mutations in IDH1 or IDH2
Prognosis and prediction
pHGGs, H3-wildtype and IDH-wildtype , are aggressive AND
tumours, and they are considered CNS WHO grade 4. Data Absence of mutations in H3 genes
from one multi-institutional series indicate a 2-year survival rate AND
of 23 .5% and a median overall survival (OS) of 17.2 months Methylation profile aligned with pHGG RTK1, pHGG RTK2, or pHGG MYCN
11976). Outcome was shown to be worst with pHGG MYC N OR
(median OS: 14 months), Intermediate with pHGG RTK1 Key molecular features: PDGFRA alteration, EGFR alteration, or MYCN
(median OS: 21 months), and better with pHGG RTK2 (median amplification
OS: 44 months) 11723). Unlike for adult high-grade gliomas, the
findi ng of vascular endothelial proliferation and/or necrosis is Desirable:
not prognostic 13292). Few data exist on potentially targetable
Microvascular proliferation
alterations in pHGG. H3-wildtype and IDH-wildtype; recurrent
Necrosis, typically palisading
alterations of PDGFRA and EGFR may be the most frequent of
such alterations, but their predictive ability is yet to be dem- H3 p.K28me3 (K27me3) retained
onstrated . Hypermutant tumours, most frequently arising in the
context of CMMRD, may respond to immunotherapy {1977) (see
Constitutional mismatch repair deficiency syndrome, p. 452).

80 Gl 1omas glioneuronal tumours and neuronal tumou rs


Infant-type hemispheric glioma Jacques TS
Bandopadhayay P
Jon es C
Tabori U
Varle P

Definition Epidemiology
Infant-type hemispheric glioma is a cerebral hemispheric, high- AU reported cases have occurred early in childhood . mostly 1n
grade cellular astrocytoma that arises in early childhood, typi- the first year of life. In one cohort, the median age at presenta-
cally with receptor tyrosine kinase (RTK) fus ions including those tion was 2.8 months (range: 0-12 months) (1179).
in the NTRK family or in ROS1 , ALK, or MET
Etiology
ICD-0 coding Unknown
9385/3 Infant-type hemispheric glioma
Pathogenesis
ICD-11 coding Structural genomic variants, often driven by focal intragernc
2AOO.OY Other specified gl iomas of brain DNA copy-number changes, result in the acquisition of fu sion
genes involving numerous 5' partners and the receptor tyrosine
Related terminology kinases NTRK1 , NTRK2, NTRK3, ALK. ROS1. or MET at the 3'
None end . These may be either interchromosomal or 1ntrachromo-
somal and may resu lt from small interstitial deletions or amplifi-
Subtype(s) cations. They cause the aberrant expression of an active kinase
Infant-type hemispheric glioma, NTRK-altered; infant-type hem- domain , driving tumorigenesis via signalling through canonical
ispheric glioma, ROS1-altered ; infant-type hemispheric glioma, Pl3K and/or MAPK pathways. There are generally no other
ALK-altered ; infant-type hemispheric glioma, MET-altered genetic alterations, rendering the tumours particularly sensitive
to targeted inhibition of the relevant RTK 11179.6031.
Localization
These gliomas appear in the supratentorial compartment, usu- Macroscopic appearance
ally as large masses 129,616,2322,3264,2238,603,1 179) . There There are few macroscopic pathology descriptions , but these
is frequently superficial involvement that includes the leptome- tumours are large, and some have a cystic component and
ninges 1603). a solid portion . Necrosis or haemorrhage can occur (29,616,
2322,3264,2238,603,1179).
Clinical features
The presentation is usually acute. During infancy, children with Histopathology
infant-type hemispheric glioma may present with nonspecific The histological descriptions derive from two large studies
signs and symptoms ranging from agitation to lethargy. Head (1179,603) and a few case reports (29,616.2322,3264 ,2238) .
circumference may be large. Some tumours can be diagnosed The original diagnoses were often glioblastomas or other
antenatally. high-grade gliomas (84%) (1179,603 ,29,616,3615) but also
included anaplastic gangliogllomas, desmoplastic infantile
ganglioglioma/astrocytoma , ependymomas , and CNS primitive Box2.11 Diagnostic criteria for infant-type hemispheric glioma
neuroectodermal tumours {603 ,2322] .
Essential:
The tumours are frequently cellular, are well demarcated from
Cellular astrocytoma
the adjacent brain parenchyma , and involve the leptomenin-
ges {603). Astrocytic , often spindle-shaped , cells with mild to AND
moderate pleomorphism are arranged in fascicles or uniform Presentation in early childhood
sheets . There is frequently palisading necrosis, mitotic activ- AND
ity, and microvascular proliferation . More rarely, a gemistocytic Cerebral hemispheric location
morphology is seen . Some tumours (including some with ALK AND
fu sions) may be more heterogeneous, with ependymal differen- Presence of a typical receptor tyrosine kinase abnormality (e.g. fusion in an
tiation {603 ,2322] or a biphasic appearance with low-grade and NTRK family gene or in ROS1, MET1, or ALK)
high-grade components or occasional ganglion cells (603 ,1179, OR
3264,2238). Th is tumour type is not currently graded . Methylation profile aligned with infant-type hemispheric glioma

lmmunophenotype
The glial component shows immunoreactivity for GFAP but Staging
does not usually express neuronal markers (2322,3264,3615) . Occasional cases show leptomeningeal dissemination (2322]
ALK immunostaining can be found in at least some tumours so craniospinal imaging is prudent. However, a formal staging
with ALK fusions 11179,2322} . NTRK immunostaining is not system is not available.
helpful because of the high level of NTRK expression in normal
brain . Prognosis and prediction
Prospective outcome data for this new entity are lacking. His·
Differential diagnosis torically, high-grade gliomas in infants have been recognized
The differential diagnosis includes other high-grade gliomas , to have better outcomes than those in older ch ildren (801)
desmoplastic infantile ganglioglioma/astrocytoma, gangli- In this context, it is notable that most descriptions of infantile
oglioma, and ependymomas in infants. hemispheric gliomas report a higher survival rate than those ol
typical high-grade gliomas (29,616 ,2322,3264,2238,603,11791.
The total number of clinically annotated cases with in each
Cytology . .
lntraoperative cytology is not well described in genetically molecular subtype remains relatively small , but individual driver
defi ned cases , but it is likely to mimic the features seen on par- events may be associated with distinct cl inical outcomes. In one
study, patients with ALK-rearranged tumours appeared to have
affin histology.
a better 5-year overall survival rate than patients with tumours
that har?oured .ROS1 alterations (53 .8% vs 25% , respectively).
Diagnostic molecular pathology
RTK fusions can be therapeutically targeted and are present and patients with NTRK fusion- positive tumours had an inter·
in 60- 80% of cases . Therefore, where possible, in infants, me?iate p'.ognosls (5-year overall survival rate: 42.9%) (1179)
Patients ~1th ALK-rearra.nged tumours with low-grade histology
routine testi ng for such fusions should be consid~red {1 :79 ,
had survival rates superior to those of patients with ALK-altered
603), both to establish the diagnosis and to provide options
high-grade gliomas . However, prospective studies across larger
for therapy. . cohorts are needed to confirm these initial observations. Finall
These tumours form a distinct subgroup by methylat1~n array
the prese~ce of RTK-activating fusions offers an opportunity tor
profiling regardless of formal tumour grading or RTK fus1.on typ~
therapy with small-molecule inhibitors. Responses to specific
{603) . This can also be used to establish the diagnosis but it
inhibitors ha~e been re.ported .{603 ,786 ,3615} and may chanQ
may not be sufficient to enable targeted therapy.
the prognosis of infantile hemispheric gl l oma~ . Further studies
are needed to evaluate the efficacy of these inhibitors and tha
Essential and desirable diagnostic criteria overall survival rates for each subtype.
See Box 2.11.

82 r-1i<,Ul[J S glio r18urur1al tu1 nou rs, <rnd neuronal lumours


Pilocytic astrocytoma Tihan T
Figarella-Branger 0
Jones DTW
Pfister SM
Giannini C Rodriguez FJ
Gupta K Tabori U
Hawkins CE Varlet P
Jacques TS

Definition
Pilocytic astrocytoma is an astrocytic neoplasm with variable
proportions of bipolar hair-like (pilocytic) cells, compact and
loose or myxoid regions, Rosenthal fibres, and eosinophilic
granular bodies. Pilocytic astrocytoma is associated with MAPK
pathway gene alterations (most often KIAA1549: :BRAF gene
fusions) (CNS WHO grade 1).

ICD-0 coding
9421 /1 Pilocytic astrocytoma

ICD-11 coding
2AOO.OY & XH29Q5 Other specified gliomas of brain & Pilo-
Flg.2.68 Pilocytic astrocytoma. Typical MRI appearance of pilocytic astrocytoma in
myxoid astrocytoma the posterior Iossa of a 5-year-old boy, showing a large cystic lesion with an enhancing
mural nodule. The tumour has BRAF kinase domain duplication with KIAA1549::BRAF
Related terminology gene fusion . A T2-weighted image. B Contrast-enhanced T1 -we1ghted image.
Not recommended (obsolete): juvenile pilocytic astrocytoma.
be asymptomatic , and long-term survival is possible, even
Subtype(s) without adjuvant treatment l2532J .
Pilomyxoid astrocytoma; pilocytic astrocytoma with histological
features of anaplasia Imaging
Pilocytic astrocytomas have a wide spectrum of imaging fea-
Localization tures , but about two thirds appear as a well-circumscribed
Pilocytic astrocytomas can arise throughout the neuraxis, but cystic lesion with an enhancing mural nodule on MRI. The
they are most common in the cerebellum, especially in children remainder often appear either as a cyst-like mass with a central
1207,621 l. Other preferred sites are the optic nerve, midline loca- non-enhancing zone or as a predomin antly solid mass 1612,
tions (brainstem, optic chiasm I hypothalamus, basal ganglia), 2451] . The cyst wall enhancement is variable, and enhancement
and spinal cord . Tumours in the cerebral hemispheres are rare does not necessarily indicate tumour involvement. Calcifica-
in children , but in adults they occur here with equal frequency tion may be present. Pilocytic astrocytomas are often contrast
as in the cerebellum (621}. enhancing, with the solid tumour component typically isoin-
tense to hypointense on T1 imaging and hyperintense on T2.
Clinical features Imaging characteristics are often not specific enough to allow a
Presenting signs and symptoms, which are usually due to diagnosis without biopsy. Imaging characteristics of optic nerve
mass effect or ventricular obstruction, include macrocephaly, tumours vary, with neurofibromatosis type 1 (NF1)-associated
headache. endocrinopathy, and evidence of increased intra- tumours rarely extending beyond the optic pathway and often
cranial pressure . The slow growth sometimes leads to diag- appearing solid, and non-NF1 counterparts involving the optic
nostic delay because symptoms are subtle. Focal neurological chiasm, extending beyond the optic pathway, and frequently
signs relate to tumour location . Optic pathway tumours often being cystic {1711}.
produce visual loss 11360,1911). Brainstem pilocytic astrocy-
tomas most often cause hydrocephalus or signs of brainstem Epidemiology
dysfunction . Patients with thalamic and other supratentorial Pilocytic astrocytoma accounts for 5% of all primary brain
tumours present with focal motor deficiencies or movement tumours \2344] . It is most common during the first two decades
disorders, whereas spinal cord lesions are associated with of life, with an average annual age-adjusted incidence rate of
back pain , paresis, and kyphoscoliosis . Hypothalamic/pitui- 0.91 cases per 100 000 population 12344}. Pilocytic astrocy-
tary dysfunction . including obesity and diabetes insipidus. is toma accounts for 17.6% of all childhood primary brain tumours
often apparent in patients with large hypothalamic tumour_ s and is the most common glioma in children . The incidence rate
12706). Especially in infants, midline tumours can be associ- is highest In young children and decreases with advancing age
ated with emaciation and failure to thrive (diencephalic syn- 12344]. Pilocytic astrocytoma is rare in older adults 12344}.
drome) , with a poor clinical outcome 12125,2459). Primary
dissemination at diagnosis may also be more common in this Etiology
age group 12125,2459) . However, neuraxis seeding does not Although most cases are sporadic [31961. pilocytic astrocy-
necessarily indicate aggressive growth 12532]. Seeding may tomas are also the principal CNS tumour type in a group of

Gl1omas , gl1oneuronal rumours . and neuronal tumou 1s 83


neurodevelopmental diseases with germline mutations in MAPK inci dence of MAPK pathway gene alterations identified to date
pathway genes, including NF1 , Noonan syndrome, and enceph - Is summarized in Table 2.03 . Polysomies (In particular of chro.
alocraniocutaneous lipomatosis. NF1 (see Neurofibromatosis mosomes 5, 6, 7, 11 , and 15) are reportedly more common ·
type 1, p. 426) is caused by NF1 germline mutations , whereas tumou rs of teenagers and adults {1494}.
Noonan syndrome is most frequently caused by mutations in The incidence of the various gene alterations varies with ana.
PTPN11 or RAFt (2859). and encephalocraniocutaneous lipo- tomica l location {1493 ,3597}, and distinct anatomical subse
matosis by FGFR1 germline mutations {240,3263) . of tumours may be distinguished on the basis of gene expres.
sion and DNA methylation profiles {1795 ,2897,3157,4601. The
Pathogenesis KIAA 1549::8RAF fusion is common in cerebellar tumours, b
Pilocytic astrocytomas are associated with genetic abnormali- less common supratentorially. FGFR1 alterati ons are wide~
ties in genes encoding members of the MAPK pathway. The distributed , whereas BRAF p.V600E mutations are more com-
most frequent abnormality (found in -60% of all cases) is a mon in supratentorial tumours. lnfratentorial and supratentorial
duplication/rearrangement of approximately 2 Mb at 7q34, tumours may also be distinguishable on the basis of their gene
encompassing the BRAF gene {194,742,2491 ,2768} and result- expression or DNA methylation signatures (1795,2897,315n
ing in gene fusions involving various combinations of KIAA 1549 The average mutation burden is low.
and BRAF exons, which makes it difficult to com prehenslvely
detect all possible fusions by RT-PCR. Macroscopic appearance
Essentially all pilocytic astrocytomas studied genomically Most pilocytic astrocytomas are soft, grey, and relatively dis-
had a genetic alteration affecting the MAPK pathway {1493, crete. lntratumoural or paratumoural cyst formation, including
3597,1605). These alterations include NF1 mutations, wh ich are mural tumour nodules , is common. Chronic lesions may be cal-
mostly germli ne mutations in patients with NF1 {1201 }; hotspot cified . Spinal tumours may be associated with syrinx formation
BRAF p.V600E mutations; BRAF fusions with partners other {2121} . Optic nerve tumours often circumferentially infiltrate the
than KIAA 1549; BRAF insertions; KRAS mutations; FGFR1 optic sheath {326}.
mutations or fusions; very occasional NTRK family receptor
tyrosine kinase fusions; and RAF1 gene fusions, usually with Histopathology
SRGAP3 but exceptionally wi th other partners {3532,2424} . Pilocytic astrocytomas have low to moderate cellularity. Neo-
NTRK genes fuse with several different 5' partners that contain plastic cells range widely in their morphology and include vary-
a dimerization domain. Th is is presumed to lead to constitutive ing proportions of piloid and oligodendrocyte-like cells . Nucle'
dimerization of the NTRK fu sion proteins and activation of the are round to elongate. Multinucleated cells with horseshoe·
kinase (1493,3597). shaped nuclear clusters (pennies-on-a-plate pattern) are often
The FGFR1 alterations seen in pilocytic astrocytomas overlap seen. In some cases, hyperchromasia and pleomorphism are
with those seen in other paediatric low-grade glial and glioneu- obvious but mitotic figu res are rare (1091} . Rare cases have
ronal tumours . They include hotspot point mutations (p.N546K brisk mitotic activity, which may imply aggressive behaviollf
and p.K656E) {32261. FGFR1 :: TACC1 fusions, and an internal (2697}. Rosenthal fibres and eosinophil ic granular bodies are
tandem duplication of the kinase domain of FGFR1 {1493). The common , but they vary in prominence. Myxoid background

Table2.03 Molecular alterations and their prevalence in pilocytic astrocytomas


%oftumours Prevalence
Genetic atteraflon
-----
Common, particularly cerebellar; rare in other tumour forms except diffuse leptomeningeal
KJAA 1549: :BRAF fusion >60% {1496,2936}
glioneuronal tumour

Other BRAF fusJons <5% Occasional; very rare in other entities {1287}

BRAF mutations - 5-10% occasional, mainly supratentorial; also in many other glial and glioneuronal tumours {2842,14951
(especially p.V600E)
- 10-15% Typically germline; common with optic pathway tumours {1198,1201)
NF1 mutation
< 5% Found mainly in midline tumours; also observed In other low-grade gliomas {1493,3226,29321
FGFR1 mutation
FGFR11uslons < 5% FGFR1 ::TACC1 fusion more common; also observed in other low-grade gliomas {1493,2932}
{etpeelaJly FGFR1:: TACC1)
- 2% Rare; also observed in other glial/glioneuronal tumours {1493,2424}
NTRK family fusions
Very rare Jn pilocytic astrocytoma; extremely rare In other entities (2898,1454}
KRA6 mutation Single cases
Very rare in pilocytic astrocytoma; extremely rare in other entities (with the possible exception
Single cases (1495,962,3532)
RAF1fuslon of pleomorphic xanthoastrocytoma)
Very rare in pllocytic astrocytoma; more common in infantile hemispheric gUoma; extremely
Single cases {2:663,2768}
ROS1fu on1 rare in other entities
Very rare in pilocytlc astrocytoma; frequency In other entities not determined (probably
Other alterations Single cases {2768}
extremely rare)
(of MET, RET, etc.)

84
Fig. 2.69 Pilocytic astrocytoma. A Typical biphaslc appearance of pilocytic astrocytoma. Compact areas that often harbour Rosenthal fibres alternate with loose and somewhat
myxoid regions. B Rosenthal fibres and eosinophilic granular bodies. Typically, Rosenthal fibres are more abundant than eosinophillc granular bodies in pilocytic astrocytomas,
but both can be observed, as in this case. C Hyaline vessels and Rosenthal fibres. In most pllocytic astrocytomas, vascular structures within the tumour demonstrate brisk
hyalinization. Numerous Rosenthal fibres are also observed. D Tumour with oligodendrocyte-like cells. Even though this tumour resembles oligodendroglioma cytologically, the
presence of eosinophilic granular bodies at higher magnification provides a useful diagnostic clue for pilocytic astrocytoma. E Pilocytic astrocytomas often harbour large cells
with multiple nuclei (the pennies-on-a-plate appearance). F Infiltrative edge. In many pllocytic astrocytomas, infiltrative-appearing regions can be recognized. In small biopsies,
these areas may evoke the impression of a diffuse astrocytoma.

with microcystic c hange is common , as are degenerative lmmunophenotype


changes , including c alcifications, hyal inized vessels , and lmmunohistochemistry demonstrates strong diffuse positivity for
haemorrhages [621} . Various histolog ical patterns can be seen GFAP, S100, and OLIG2. Many cases are positive for synapto-
in pilocytic astrocytomas: (1) a biphasic pattern , in which com- physin but negative for NFP, NeuN, and chromogranin . CD34 is
pact areas rich in bipolar cells and Rosenthal fibres alternate usually negative, although expression has been reported in the
with loose and microcystic reg ions rich in oligodendrocyte-like hypothalamic/chiasmatic reg ion (523). IDH1 p. R132H expres-
cells; (2) a predominantly compact , pilold pattern , with abun- sion is absent and the H3 p.K28M (K27M) stain is negative, with
dant Rosenthal fibres; and (3) a more dispersed pattern , rich in rare exceptions . Most tumours show strong and diffuse SOX10
oligodendrocyte-like cells , mimicking oligodendroglioma. The and p16 staining , with SOX10 and OLIG2 positivity helping to
biphasic pattern is common in cerebellar tumours . The com- distinguish pilocytic astrocytoma from ependymoma (1655) .
pact pattern is often seen in adults, and the oligodendrocyte- The Ki-67 index is usually low, with only focal increases.
like pattern may be associated with FGFR1 alterations ( ~ 768) .
Occasionally, typical pilocytic astrocytomas have foc i that Subtypes
resemble pilomyxoid astrocytoma; these are called intermedi- Pilomyxoid astrocytoma
ate tumours . Rare examples also demonstrate the regimented Pilomyxoid astrocytoma (3197) shares many features with clas-
palisaded (spongioblastoma) pattern . Pilocytic astrocytomas sic pilocytic astrocytoma but differs in some important clin-
show highly vascular areas with thin glomerular ca pil laries icopathological respects (1694 ,910). It is a tumour of infancy
often arranged in a linear fashion and associated with cystic occurring in the hypothalamic/chiasmatic region, has a higher
structures , or they have thick-walled, hyalinized vesse ls and rate of recurrence and a poorer outcome than classic pilocytic
regressive changes. Glomeruloid microvascular proliferations astrocytoma, and shows a propensity for cerebrospinal dis-
line the cyst wall and should not prompt a higher grade desig- semination (1694,1471). Pilomyxoid astrocytoma is defined as
nation . Infarct-like necrosis can occur, but palisading necrosis a tumour with monomorphic piloid cytology, a diffusely myxoid
is exceptional. Leptomenlngeal involvement can occur in any background, and increased cellularity compared with that of
location, sometimes with extensive desmoplastic reaction . c las.sic pil~cytic astrocytoma (3197}. It also has a prominent
Some tumours may mimic diffuse astrocytomas on hlstopathol- ang1ocentn? arrangement of tumour cells and typically lacks
ogy because of a surprising degree of infiltration, despite often Rosenthal f 1bres and eosinophilic granular bodies. On neuroim-
appearing solid rad iologically. Entrapped neurons can also be agl~g, pilomyxoid astrocytomas appear similar to classic pilo-
mistaken for a neuronal component (e.g. ganglioglloma). cyt1c astrocytomas , but they are more often solid and uniformly

Gliom s, glioneuronal tumours . nnd neuronal tumours 85


Fig. 2.70 Pilomyxoid astrocytoma. Tumour in a ?-month-old girl. There was spinal and brainstem dissemination at diagnosis. She had three recurrences over the following 4yeari
while being treated with trametinib with partial response. The tumour has BRAF kinase domain duplication with KIAA 1549::BRAF gene fusion . Contrast-enhanced MRI. AC01:r
nal. B Axial. C Sagittal.

enhancing {79,2188) . Some pilomyxoid astrocytomas develop (mean age: 32 years ; range : 3-75 years) , mostly involved the
into a classic pilocytic astrocytoma on recurrence . Rare hybrid posterior fossa , and showed heterogeneous genetic features
pilomyxoid/pil ocytic astrocytomas have been reported, but their with alterations typical of pilocytic astrocytoma and other glio-
biological behaviour is poorly defined 11487}. Molecular studies mas {26931, including BRAF duplications (30%) , germline or
identify MAPK pathway gene alterations similar to those in clas- somatic NF1 mutations (33%) , loss of nuclear ATRX expres-
sic pilocytic astrocytoma, but differences have been reported sion (57%), and an alternative-lengthening-of-telomeres phe-
11471 ,1656,1756}. Further studies are needed to elucidate the notype (69%). Features associated with worse overall surviva1
precise molecular profile of pilomyxoid astrocytomas 1460}. included necrosis , subtotal resection , alternative lengthening
of telomeres, and ATRX loss (P < 0 .05) . The overlap between
P1locytic astrocytoma with histological features of anaplasia pilocytic astrocytoma with histological anaplasia and the rare
Pil ocytic astrocytomas are remarkable in that they maintain midline pilocytic astrocytoma with double mutant FGFA1.
their CNS WHO grade 1 1421} status over decades. The terms BRAF, or NF1 and H3 p.K28M (K27M) remains to be deter·
"anaplastic pilocytic astrocytoma" and "pilocytlc astrocytoma mined , although both have been associated with aggressive
with histological anaplasia" have been proposed for tumours behaviour {2643,2693,1493} .
with morphological features of pilocytic astrocytoma but show- In a cohort of predominantly adult patients across > 20 lnsu-
ing brisk mitotic activity with or without necrosis {2697). Ana- tutions worldwide, who were considered to have histological~
plastic changes may be present at initial diagnosis or at recur- defined anaplastic pilocytic astrocytoma , 81% of the tumours
rence . In a study of 36 pilocytic astrocytomas with anaplasia harboured a distinct methylome signature, referred to as "DNA
defined histologically, they predominantly occurred in adults methylation class anaplastic astrocytoma with piloid features'

A B
1
Flg. 2,71 Opuc patliway glioma (ptlocytic astrocytoma). A Cross -section reveals.an expanded optic ne.rve with subarachno1d extension of the tumour. The latter is a comrno'
feature of ptlocyt1c astrocyloma. B A GFAP stain highlights the regions of tumour involvement 1n this optic pathway glloma.
k'
, ~

Fig. 2.73 Pilomyxoid astrocytoma. A Loose, mucin-rich arrangement of piloid cells, some of which form angiocentric structures resembling perivascular pseudo rosettes. B Pi-
lomyxoid astrocytoma In a 10-month-old boy, located in the hypothalamic region. The typical angiocentric and markedly myxoid pattern is seen throughout the tumour. The tumour
has BRAF kinase domain duplication with KIAA 1549::BRAF gene fusion.

{2643). These tumours are now considered a distinct type, Differential diagnosis
designated as "high-grade astrocytoma with piloid features". A relevant differential diagnosis in the presence of microvas-
A similar DNA methylation profile was found in 36% of histo- cular proliferation and/or necrosis is a high-grade astrocytic
logically defined cerebellar glloblastomas (2644). Although glioma including glioblastoma. Solid growth pattern , low mitotic
this methylation class is enriched for neoplasms pathologically activity, and bipolar cells, in addition to molecular features, help
diagnosed as pilocytic astrocytoma with histological anaplasia, resolve this differential diagnosis. Rosenthal fibre-rich piloid
the two categories do not overlap completely and their relation- gliosis may also mimic pilocytic astrocytoma, but it lacks the
ship remains to be elucidated . loose component. In the midline, an H3 K27-altered diffuse
H1stolog1cal and molecular diagnostic criteria established for midline glioma must be excluded, although rare pilocytic astro-
pilocyt1c astrocytoma with anaplasia in adults may not be appli- cytomas acquire an H3 p.K28M (K27M) mutation in addition to
cable to children . In one study of 31 paediatric patients (aged their MAPK gene alteration . Other differential diagnoses include
< 16 years), on multivariate analysis, only young age(< 6 years) ganglloglioma, dysembryoplastic neuroepithelial tumour,
and partial resection were associated with decreased progres- pleomorphic xanthoastrocytoma. rosette-forming glioneuronal
sion-free survival 11040). Necrosis and high mitotic activity tumour, ependymoma (especially tanycytic), and diffuse lep-
were not significantly associated with survival. Nuclear ATRX tomeningeal glioneuronal tumour. Some tumours of this last
expression was preserved in all tumours. and only one tumour type may be virtually indistinguishable from pilocytic astrocy-
matched the methylation class of high-grade astrocytoma with toma on routine histology.
plio1d features, with an additional case showing homozygous A note of caution is warranted in regard to anaplastic trans-
deletion of COKN2A and/or CDKN28. formation after radiation therapy, especially with a long interval

Gl1omas. gl!oneuronal tumours , dnci llHL1ro11::-11 tu1 nuurs 87


and wi hout the presence of a piloc ic c pone : so
hese tumours may instead be a seco d pn ary. i.e. ad· •·
induced igh-grade g lioma I 441}.

Cytology
lntraopera 1ve smears of pilocytic astrocyto a are c arac•er-
ized by cells with long . fine. hair-Ii e processes. T e cells s -
ally show uniform, rou d o spindled nuclei 1 h mini al a pia.
Rosenthal fibres . eos1nophilic granular bodies. and gl e lo·d
vessels can be present. In some cases. ere ma ' be oege -
era i e atyp1a, with multmucleated cells.

Diagnostic molecular pathology


The most frequent genebc al eratio in pilocy ic astroc -
toma 1s a chromosome 7q34 rearrange ent resul ng 1n a
KIAA 1549::BRAF tandem dupllca io and s1on { 496.2491 ,
962} In small numbers of cases. al emate BRAF f s1ons a e
also been 1dent1fied . occurring by anous genetic rearrange-
ments (including deletions and transloca ions) b all resul ing
1n loss of the N-term1nal regulator region o the BAAF protein.
wnh retention of the kmase domain { 493.3597 287.32 1}. Other
MAPK pathway gene arerat1ons aJso occ r (see Table 2.03,
p. 84) 11493,3597 )
The presence of the KIAA 1549-BRAF us on ( r other MAP J\/
gene alterations} supports the diagnosis of p1locync astrocy- gl
torna 1n an appropriate morphological conte However. t e

88 J, t d ~- C' ' i ... ' , . ~ ' - (


Essential and desirable diagnostic criteria
see Box 2 12 Box2.12 Olognos t1c criteria for classic pilocyllc astrocytoma and pilomyxoid astro·
cytoma
Staging Claulc pllocytlc 111trocytomo
For inf ratentonal and spinal tumours , p1locyt1c c strocytom st g- Essential:
1ng involves cerebral and spinal MRI. wh r as for suprat ntonc: 1 Classic histological featuros or p locytlo trocytoma, such as blphaslc compact
turnours spinal MRI is only perform d 1f ther Is ev1d nc of 1ntra- and loo e growth patterns, ptloid cytology, and low proliferattve actMty, wth or
cran1al d1ssem1nat1on Cerebrosp1nal llu1d c tology is only done if without Aosenttial fibres and/or eoS1nophUlc granular bodies
rad1olog1cal 1nd1ngs 1nd1cate disseminated disease I1122) OR
A low-grade pilold estrooytio neoplasm wilh a sohlafy MAPK alteration, 84.ICh as
Prognosis and prediction KlAA 1549::BRAFfusion
In ost large series. p1loc t1 . astrocyt mas re associated
Pllomyxold astrocytom
,..,1 a fa\ourab1e o erall surv1 al even after multiple progr s-
s 0'1S :'\hen con1pletel ' resected. iloc tic a~troc tomas r rely Esuntla/:
recur {-6 7 2~301 Because of the fa ourable o er II utcome, A monomarph c, loose, myxold neoplasm with pUold cytology and prominent angJ,
rad at1on-s('anng a proact1es are commonly re omm nded ocentnc pattern, ofton without Rosenthal rlbres and eosinophillc granular bodies
117~91 Ana because the ma1or1t of tumours harbour alter lions OR
1n ~A.PK pat \•.a genes . he r ma fa urabt r sp nd to t r- An astrocytic neoplasm with pilomyxold features and a solitaty MAPK alteratlon,
geied l\1EK 1n 1b. 1un. T e o ·eratl I ng-term utcome of sucl1 such as KIAA1549::8RAFfuson
appr a hes 1s . et t be deterfT'l n d In cases of ggr s 1ve
turn 1.1r bera\ 1 r and resistance to chemot rap , a biopsy likely to act aggressively 13197 1. The prognostic signi ficance of
rra · 'e cor.s•dere t all for molecular charactenzalton A pilocytic astrocytoma with histological anaplas1a needs to be
cau .onar) sta•e ..... ~n, is arranted p1lom 01 as r yloma is determined .

Gl11..J t rld~ 9 11unl"u run.il 1 Jl"•J'J' . r; j 1,r•ur< 1ul lun'uurs 89


Capper D
High-grade astrocytoma with Jones DTW
Rodriguez FJ
piloid features Varlet P

Definition
High-grade astrocytoma with piloid features (HGAP) is an astro-
cytoma showing a distinct DNA methylat1on profile , often with
high-grade p1lo1d and/or glioblastoma-like histological features .
Alterations of MAPK pathway genes are often combined with
homozygous deletion involvin g the CDKN2A and/or COKN2B
locus , and/or ATRX mutation or loss of nuclear ATRX expres-
sion .

ICD-0 coding
9421 /3 High-grade astrocytoma with piloid features

ICD-11 coding
2AOO .OY & XH6PH6 Other specified gliomas of brain & Astro- flg. 2.76 High-grade astrocytoma with piloid features. A T2-weighted MRI
cytoma. NOS hyperintense lesion without sharp tumour margins 1n the dorsal pons and nght
peduncle. B FET PET indicates highly Increased amino acid metaboLism in the
Related terminology
Not recommended: anaplastic astrocytoma with piloid features ; Etiology
anaplastic pilocytic astrocytoma. Risk factors
In most patients , HGAP occurs de nova . In the remairn
Subtype(s) patients, the tumours may develop from a pre-existing IOwef·
None grade astrocytic tumour, including pilocytic astrocytoma. In one
series, the precursor lesion dated back > 10 years in 4 (18%) a
Localization 22 cases (2643) . A prior history of cerebral Irradiation is uncom.
HGAP may occur throughout the entire CNS . Most frequently, mon, reported in 4 (5%) of 83 patients (2643}. and no defini•e
tumours originate in the posterior fossa, where they typically etiological role for irradiation has been established.
affect the cerebellum (In 74% of cases) . They can also be
localized in the supratentorial (17%) and spinal (7%) regions Genetic factors
12643). Rare instances of HGAP have been reported in patients wit
neurofibromatosis type 1 (NF1) 12643). Associations with other
Clinical features tumour predisposition syndromes have not been reported .
Clinical signs and symptoms depend largely on tumour location .
Clinical features distinct from those of other types of gliomas in Pathogenesis
the same locations have not been reported . On imaging, some Molecular data imply that three pathways are centrally involved
tumours may appear as a ring-enhancing mass , mimicking the in the pathogenesis of HGAP: the MAPK pathway is frequen~v
radiological appearance of glioblastoma. activated by mutations : the retinoblastoma tumour suppressor
protein cell-cycle pathway is frequently deregulated by COKN2A
Epidemiology and/or COKN2B inactivation or occasionally by COK4 amph·
Comprehensive epldemiologlcal data are not available, but fication ; and telomere maintenance is frequently activated by
several case series suggest that HGAPs are rare . In non- pop- ATRX alterations and, rarely, TERT promoter mutations 126431
ulation-based case series, HGAPs (identified mostly in adults) In about half of all cases of HGAP, all three pathways are altereo
have been estimated to account for about 1-3% of brain tumours simultaneously; in the remaining cases . only one or two (or. ver y
{2573 ,1458,460); however, the potential of selection bias in rarely, none) of these alterations are detectable {26431.
such series may mean this estimate is too high . In a population- The temporal order of these alterations is not known , but rare
based study of 306 paediatric brain tumours in the United King- tumours developing in patients with NF1 may indicate that MAPK
dom , not a single HGAP was identified 12505). Furthermore, pathway gene alterations are an initiating genetic event. For a
in a paediatric study of 31 anaplastic pilocytic astrocytomas, subset of HGAPs, lower-grade precursor lesions such as pilo·
only 1 tumour (3%) was molecularly confirmed as HGAP (1040) . cytic astrocytomas have been reported , but a molecular worl<uP
HGAP thus appears to be very rare in the paediatric popula- of these potential precursors has not been performed [2643).
tion . In the combined data from three studies, the median age HGAPs typically have numerous chromosomal alteration
of reported patients was 40 years (range: 4- 88 years) , and the with more than three structuraJ aberrations found in 88% of cases
M:F ratio was balanced (1 :1) 12643,1458,10401. (26431 . Besides fre quent homozygous deletion of COKN2A and!

90 Gl 1orr1as . glioneurona l tumours, and neuro1 1al tun1ours


Ag. 2.77 High-grade astrocytoma with piloid features. This tumour type has a wide range of morphologies. A About 40% of cases have a mainly pilo1d morphology, sometimes
with numerous Rosenthal fibres. B About 45% of cases show a mainly glioblastoma-like morphology. C About 10% of cases have features reminiscent of pleomorphic xanthoas-
trocy1oma. D Tumours may rarely resemble ep1thelioid glioblastoma.

or CDKN2B, other chromosomal alterations that are recurrently Areas of solid tumour growth are frequent, but invasive growth
seen and might play pathogenetic roles include partial gains of into the adjacent parenchyma may also be observed 12643}.
chromosome arms 12q and 17q (in -30% of cases each), losses
of 1p and Sp (in -20% of cases each), and partial losses of chro- lmmunophenotype
mosomes 14 and 19q (in -20% of cases each) {2643}. A suggestive immunohistochemical marker observed in about
40% of HGAPs is loss of nuclear ATRX expression in the tumour
Macroscopic appearance cells. with non-neoplastic cell nuclei remaining ATRX-positive
Macroscopic or imaging features specific fo r HGAP have not /2643) . 101-11 p.R132H immunohistochemistry is negative. Very
been reported . Some tumours may have areas of central necro- rare tumours with expression of the H3 p .K28M (K27M)-mutant
sis mimicking glloblastoma. protein have been reported , but the definitive classification of
these tumours has yet to be establ ished 12643).
Histopathology
The histological features of HGAP vary considerably and are not Differential diagnosis
sufficiently distinct to diagnose this glioma type without additional Because of the broad and diagnostically ambiguous spectrum
molecular testing. In general , tumours appear as moderately of histological features, a wide range of gliomas represent rel-
cell-dense and moderately pleomorphic astrocytic gliomas. The evant differential diagnoses, including IDH-wildtype gl1oblas-
growth pattern may resemble that of glioblastoma or pleomorphic toma, pleomorphic xanthoastrocytoma (CNS WHO grade 2 or
xanthoastrocytoma, or the tissue may be enriched for thin , hair- 3), and pilocytic astrocytoma (especially tumours with histologi-
like (piloid) cytoplasmic processes (hence the name "high-grade cal features of anaplas1a) .
astrocytoma with piloid features"). In about a third of cases, The first study of HGAP demonstrated that in a predominantly
eosinoph1lic granular bodies or Rosenthal fibres are observed . adult population , about 80% of tumours histolog1cally con-
Almost 90% show alterations of vasculature, either in the form of sidered to be anaplastic pllocytic astrocytomas represented
hypertrophy and/or multilayering, or as glomeruloid proliferation . HGAP upon DNA methylation analysis 12643). In contrast, in
One third of the tumours show necrosis (with or without palisad- a purely paediatric population of anaplastic pilocytlc astrocy-
ing), and about 80% have~ 0.42 mitoses/mm 2 (equating to~ 1 toma, only ·1 (3%) of 31 tumours was molecularly confirmed
rnltosis/10 HPF of 0.55 mm in diameter and 0.24 mm 2 in area). as HGAP {"1040}. It was further shown that about one third of

·I
~

Fig. 2.78 Infiltration patterns of high-grade astrocytoma with piloid features. A Sparse infiltration of cerebellar white matter. Note the single pleomorphic cells. B Diffuse
infiltration of the tumour into the granular layer of the cerebellar cortex. C Diffuse Infiltration of the tumour into the pons, with residual pigmented neurons of the locus coeru-
leus. D ATRX immunohistochemistry of the same case as in C, with nuclear ATRX loss in tumour cells but retained staining in pre-existent cells.

histologically defined cerebellar glioblastomas molecularly rep- molecular class is included in widely used machine learning-
resented HGAP {2644}. based classifiers {460}. A combination of histology with certain
genetic markers may be suggestive of the diagnosis but may
Cytology not clearly distinguish these tumours from other gliomas such
Not clinically relevant as pleomorphic xanthoastrocytoma or glioblastoma.
A variety of MAPK pathway gene alterations have been
Diagnostic molecular pathology reported {1040,2643}, the most frequent being NF1 alterations,
Currently, DNA methylation profiling is the only method for KIAA1549::BRAFfusions, and FGFR1 mutations (Table 2.04). In
definitively establishing a diagnosis of HGAP {2643}. The one tumour, an NF1 alteration was combined with an FGFR1

12

0.8

OA

00

-o.e
COKN2A / B

-12

Flg. 2.79 High-grade astrocytoma with p1loid features. The DNA copy-number profile of this. case shows, among other changes, several chromosomal alterations that recur·
rently occur in this tumour type: homozygous deletion of CDKN2A and/or CDKN2B (observed in close to 80%), partial gain of 12q and 17q (each in _30 %), and 1p loss (in -2~~

92 G liomas , gl1oneuronal tumours . and neuronal tumours


Box2.13 Diagnostic criteria for high-grade astrocytoma with pilo1d features

P;iocy<"""'~
Essential:
An astrocytic glioma
.. AND
~ A DNA methylatlon profile of high-grade astrocytoma with piloid features

PXA. . . 4l:. Olff"" Desirable:

__.
l• o<omoolog"I
G lloblas t o m ~, IOH-wildtype ..- - ,. glioneuronal tumour MAPK pathway gene alteration
Homozygous deletion or mutation of CDKN2A and/or CDKN2B,
or amplification of CDK4
Mutation of AmX or loss of nuclear ATRX expression

t
HGAP Anaplastic histological features

observed in some cases (2643 }. ATRXmutations and/or loss of


Diffuse mldline glioma,
H3 K27M -mutant
ATRX expression was observed in 33 (45%) of cases. In rare
Diffu se hemispheric glloma,, . Instances without ATRX alteration , TEAT promoter mutations
H3 G34-mutant
were detected (2 [3%) of 74, both p.C228T mutations) {2643) .
In 2 (3%) of the 74 tumours, H3 p.K28M (K27M) mutations were
Flg.2.80 High-grade astrocytoma with piloid features (HGAP) is defined by a specific identified, but the definitive classification of these tumours has
DNA methylation profile. \-distributed stochastic neighbour embedding (t-SNE) of DNA
yet to be established (2643) .
methylation data of HGAP and several relevant differential diagnoses. HGAP forms a
distinct molecular group. Data combined from {460) and {2643). MES, mesenchymal;
PXA, pleomorphic xanthoastrocytoma; RTK, receptor tyrosine kinase. Essential and desirable diagnostic criteria
See Box 2.13.
Table2.04 Frequency of MAPK alterations detected to date in high-grade astrocytoma
with piloid features Staging
Not established
iype of MAPK alteration Frequency
NF1 mutation1 19% Prognosis and prediction
NF1 heterozygous deletion- 12% Prognostic data on patients diagnosed with HGAP are, to date,
only available from a single retrospective study {2643 }. In this
K/AA 1549.:BRAFfusion 20%
study, the 5-year overall survival rate of patients diagnosed with
8RAFp.V600E mutation 1% HGAP was approximately 50% . Overall survival was shorter
FGFR1 p.K656E/N or p.N546D/K mutation 17% than that of patients with conventional pilocytic astrocytoma
(CNS WHO grade 1) and !DH-mutant astrocytoma (CNS WHO
FGFRt::TACC1 fusion 2%
grade 3), longer than that of patients with IDH-wildtype glioblas-
KRAS mutation 3% toma, and approximately comparable to that of patients with
Three cases had both an NF1 mutation and an NF1 heterozygous deletion.
1
!DH-mutant astrocytoma (CNS WHO grade 4) {2643}. Associa-
Data from Reinhardt et al. {2643) . tions of prognosis and histological features were not identified ,
and fatal outcomes were also seen in patients whose tumours
mutation , but MAPK pathway gene mutations otherwise occur lacked necrosis (8 of 28 patients died within 2 years of diagno-
in a mutually exclusive fashion (2643}. The rate of occurrence sis) or lacked mitoses (3 of 10 patients died with in 2 years). A
of BRAF p.V600E is notably low, and the overall frequencies of methylated MGMT promoter was reported in 46% of HGAPs,
reported MAPK pathway gene alterations are remarkably differ- without a statistical association with patient outcome; however,
ent from those of pleomorphic xanthoastrocytoma and pilocytic information on treatment of the patients by alkylating agent
astrocytoma. chemotherapy was not available {2643). More data are requ ired
In one study of 74 cases, homozygous deletion (or, very for assignment of a definitive CNS WHO grade, but current data
rarely, mutation) of CDKN2A and/or CDKN28 occurred In about suggest a clinical behaviour roughly correspond ing to CNS
80% of tumours {2643}. Alternatively, CDK4 amplifications were WHO grade 3.

Gl1om<:1s. ~lioneuronal tun1ours . and neuronal tumou rs 93

--
Giann ini C Jones DTW
Pleomorphic xanthoastrocytoma Capper D Louis ON
Figarella-Branger D Paulus W
Jacques TS Tabori U

Definition Age Distribution


Pleomorphic xanthoastrocytoma (PXA) is an astrocytoma with 25

large pleomorphic (frequently multinucleated) cells , spindle


cells, and lipidized cells, often with numerous eosinophilic
-VI
c:
Q)
:;:
20

granular bodies and reticulin deposition , and characteristically [ 15


with BRAF p.V600E mutation (or other MAPK pathway gene .....0
alterations) and homozygous COKN2A and/or CDKN28 dele- ~ 10
Q)
tion (CNS WHO grade 2 or 3). .Q
E
:J
ICD-0 coding z
9424/3 Pleomorphic xanthoastrocytoma 4 s 7
Decade
ICD-11 coding
2AOO.OY & XH99U2 Other specified gliomas of brain & Pl eo- •F•M
morphic xanthoastrocytoma F1g. 2.81 Pleomorphic xanthoastrocytoma. Age and sex distribution in a recent series
of 67 patients {3299}.
Related terminology
Not recommended: pleomorphic xanthoastrocytoma with ana- Clinical features
plastic features; anaplastic pleomorphic xanthoastrocytoma Many patients present with a long history of seizures. Cerebellar
(for CNS WHO grade 3). and spinal cord tumou rs have symptoms reflecting these sites
of involvement. In cases with deep localization (e.g. brainstem)
Subtype(s) and/or wider infiltration , gross total resection cannot be achieved.
None
Imaging
Localization On imaging , PXA is usually peripherally located and freque nt~
A superficial location involving the leptomeninges and cerebrum cystic , involving the cerebral cortex and overlying leptomenin-
is typical. The majority of tumours (98%) occu r supratentorially, ges. On CT, the tumour appearance is variable (hypodense,
most often in the temporal lobe {1400}. PXAs involving the cer- hyperdense, or mixed), with strong , sometimes heterogeneous,
ebellum and spinal cord have been reported {1106,2202}. as contrast enhancement {2337). The tumour cysts are hypodense.
well as 2 childhood cases in the reti na (3582}. On MRI , the solid portion of the tumour is either hypointense

Flg. 2.82 Pleomorphic xanthoastrocytoma. A T1 -we1ghted postconlrast MRI of a CNS WHO grad: 2 tumour fo_rming a right tempo~al superficial enhancing nodule with a smal:
cystic component and scalloping of the bone. B T1 -welghted postcontrast MRI of a CNS WHO grade 2 tumou1showing a superf1c1al enhancing mural nodule and a large cys
causing moderate m1dline shift. c T1 -weighted postcontrast MRI of a CNS WH O grade 3 tumour forming a large, heterogeneously enhancing tumour wi th moderate surrounding
oedema mass effect.

94 (.,l1<JrT1ri:-:i g\1oneuru11al 1un 1ours and n uronal tumours


I
.
.

Fig. 2.83 Pleomorphic xanthoastrocytoma, classic histology. A Pleomorphlc xanthoastrocytoma Is a cellular tumour with marked cellular pleomorphism that is apparent even
at low power. B Perlvascular lymphocytic cuffing is common. C Tumour cells show marked cytoplasmic and nuclear pleomorphlsm with frequent intensely eosinophilic granular
bodies (arrow). D There are frequent pale granular bodies (arrow) and cytoplasmic vacuolation with xanthomatous cells.

or isointense to grey matter on T1-weighted images and shows {2344) . It occurs equally in male and female patients an d typi-
a hyperintense or mixed signal on T2-weighted and FLAIR cally develops in children and young adults {1092}. Mean age
images , whereas the cystic component is isointense to cerebro- at diagnosis is 26 .3 years (med ian : 20.5 years) 12460). How-
spinal fluid. Postcontrast enhancement is moderate or strong ever, older patients (up to the eighth decade of li fe) may be
(2337). Adjacent oedema is usually not pronounced . affected {2460). There are few data on the relative preval ence
of CNS WHO grade 2 versus CNS WHO grade 3 tumours,
Epidemiology but in one series, anaplasia was present in 31 % of cases at
PXA accounts for < 0.3% of primary CNS tumours, with an first diagnosis (in 23% of paediatric patients and 37% of adu lt
annual incidence of < 0.7 cases per 100 000 population patients) 11400).

J
Fig. 2.84 Pleomorphlc xanthoastrocytoma, spindle cell histology. Some tumours have a predominant cellular and spindle appearance with relative monomorph1sm (A}, or with
only rare eosinophll1c granular bodies (B), or with large pleomorphic cells (C).

G l1om s . g l1oneu ro ric I tumours aric1 neur on.JI tumourn 95


Pathogenesis
It has been proposed that PXA originates from subp1al astrr
cytes 11595). This hypothesis would explain the superficial loc~
tlon of most tumours, and it is supported by the fact that subo;:
astrocytes and tumour cells in PXA have the same ultrast~u;.
tural features . -
PXA typically carries alterations in genes encoding member·
of the MAPK pathway (most frequently BRAF p.V600E muta~or­
combined with homozygous deletion of the tumour suppreSSo
genes CDKN2A and/or COKN28 at 9p21.3 [2499 ,3621 ,309S
3299,2842,1680,3300,1361). PXA may carry TERT prornot~
mutations or (less frequently) amplifications, and these TER-
alterations may be more common in tumours with anapla.
sia (1678 ,1962,2499,3299). TP53 mutations are rare in PXAs I
(1090,1573,2430,2499). Alterations in other genes (includ1('<j ]
SMARCB1, BCOR, BCORL1, ARID1A, ATRX, PTEN, FANCA
FANCD2, FANG/, FANCM, PRKDC, NOTCH2, NOTCHJ
NOTCH4, and BCL6 12499,3299,3300)) have been descnbea
but their pathogenetlc significance is uncertain .

Macroscopic appearance
PXAs are sometimes yellow (from lipidization), partially cystic
superficial cortical masses , although their gross appearance
may be nonspecific. They may extend into the adjacent lep-
tomeninges .

Histopathology
PXA typically demonstrates a mostly solid , non-infiltrauve
growth pattern , although microscopic invasion at the peripher1
is common . Tumours are composed of a mixture of spindled
epithelioid , pleomorphic , and multinucleated astrocytes that are ~
sometimes tilled with lipid droplets (xanthomatous cells). Intra·
nuclear pseudoinclusions, prominent nucleoli , and lymphocyrc
infiltration are frequent (1092) . Granular bod ies, both pale anc
brightly eosinophilic, are characteristic (1092}. Reticulin fibres
surrounding individual tumour cells are mainly encounter.ad J
in leptomeningeal areas . Most PXAs have low mitotic act1v1~ ~
Anapiasia manifests as brisk mitotic activity, either focally or dil·
fusely, and occurs at first diagnosis or at recurrence . Necros1s •s
frequent as well , whereas microvascular proliferation is uncorr.·
mon {1092}. CNS WHO grade 3 PXAs may demonstrate less
pleomorphism and a more diffusely infiltrative pattern than th0·r
grade 2 counterparts . Among the common cytological patterns
of anaplasia, small cell, fibrillary, and ep ithelioid/rhabdoid sub-
Flg.2.85 Pleomorphlc xanthoastrocytoma, CNS WHO grade 3. A This tumour types have been reported [3300) .
shows classic features, witll pleomorph1c and xanthomatous cells (inset) and brisk
mitotic activity (arrows). The tumour recurred 1 year later and showed remaining areas Grading
with pleomorphic morphology (B), transitioning to monomorphlc areas with epithelioid CNS WHO grade 2 is assigned to tumours with < 2.5 mitos0-
and rhabdoid morphology (C).
mm2 (equating to < 5 mitoses/10 HPF of 0.23 mm 2 1n area ano
0.54 mm in diameter). Tumours showing 2 2.5 m1toses/fT1fTl
Etiology (equating to 2 5 mitoses/10 HPF of 0.23 mm 2 in area and 0.54rnrr
No specific etiology is known . PXA may be encountered in in diameter) are CNS WHO grade 3 (1400,3300) . Necrosis IJ
patients with neurofibromatosis type 1 12560,20,1747,2224, almost always seen in tumours with high mitotic activity, a~
2359), which 1s In keeping with the high frequency of MAPK path - its significance in isolation, if any, is indeterminate at presen·,
. 'e,'< c
way gene alterations in PXA . Rare cases have been reported In CNS WHO grade 3 tumours, a mean Ki-67 labelling 1na r
15% has been reported (2499). whereas it is generally< 1 ~
0 1
In DiGeorge syndrome {2177), familial melanoma-astrocytoma
syndrome (with CDKN2A inactivation) 1507). Down syndrome CNS WHO grade 2 tumours (1092) . CNS WHO grade 3 t~ ·anv rnoU'5
12484), and Sturge - Weber syndrome 11612). may occur de novo or at recurrence of a PXA that was inill
CNS WHO grade 2 (33001 .

96 Gl1 o rn as. gl 1or1r· 1r.J 11 a l t u1 11 u 11r ~,, c.lf 'ci 11c ur u1 1;t1 ru 11 111' Jr s
/mmunophenotype
PXAs typically express GFAP and 8100 /1092,1094,1314). 8100
Is often diffusely positive . whereas at least focal positivity for
GFAP is common . Most tumours are positive for CD34 12635)
and focally express neuronal markers (synaptophysin, neuro-
tilament, class Ill P-tubulin , and MAP2) 11094,2548). although
the positive cells do not resemble neurons. BRAF p.V600E
expression (VE1 antibody) is present in 60-80% of PXAs 11401,
2842.1680,3300,2499) . Focal 8MARCB1 (INl1) loss has been
reported in rare cases that transformed into malignant neo-
plasms resembling atypical teratoid/rhabdoid tumours (3300,
2268) .

Differential diagnosis
The most frequent differential diagnosis is ganglioglioma, which
can have a glial component resembling PXA . Rare cases of
composite tumours have also been reported 11706,2468}. Both
PXA and ganglioglioma exhibit accumulation of eosinophilic
granular bodies, lymphocytic infiltration, CD34 expression , and
BRAF p.V600E; however, true ganglion cells are usually absent
in PXA . Given this overlap, the diagnosis of ganglioglioma
should be regarded with caution in cases with homozygous
deletion of CDKN2A and/or CDKN28.
PXA should be distinguished from giant cell glioblastoma, with
which it shares the features of gross circumscription , reticulin
deposition , marked pleomorphism , multinucleated giant cells,
and lymphocytic infiltration. However, the immunophenotype -
in particular p53 and neuronal antigen expression {2023) - and
the molecular profile are markedly dif'ferent (see Glioblastoma,
IDH-wildtype, p. 39).
In cases showing a dominant population of epithelioid cells
and frank anaplasia, especially in the absence of a history of
a CNS WHO grade 2 PXA, the main differential diagnosis is
epithelioid glioblastoma. because these tumours often carry
a BRAF p.V600E mutation . Approximately 60% of epithelioid
glioblastomas (the PXA-like epithelioid glioblastoma subset)
were shown to cluster by methylation profiling with canonical
PXAs (1716) . These tumours frequently carried a BRAFp.V600E
mutation (79%), COKN2A homozygous deletion (61%), and
TERT promoter mutations (30%); they lacked oncogene ampli-
fications and showed a low frequency of 1Oq loss. Although
such tumours have a more favourable outcome than typical
IDH -wildtype glioblastomas, it is unclear whether they are truly
equivalent to PXA and will have similar outcomes.
Flg.2.86 Pleomorphic xanthoastrocytoma, CNS WHO grade 3. Additional patterns of
Cytology anaplasla include monomorphic small cells (A) showing brisk mitoses (Inset, arrows),
lntraoperative smears show a variable population of pleomor- flbrlllary morphology showing brisk mitoses (B, arrows), and a pseudopapillary pattern
(C) showing brisk mitoses (Inset).
phic and spindled neoplastic cells with tibrillary processes
11475) . Large, bizarre cells with binucleatlon or trinucleation are
common. whereas cells with cytoplasmic microvacuoles con- immunohistochem1stry 11231). Tumours without BRAF p.V600E
sistent with lipid ized astrocytes are rare . mutation can harbour a wide variety of alternative MAPK path-
way gene alterations , mostly affecting BRAF (non-p.V600E
Diagnostic molecular pathology mutations, non - KIAA 1549::BRAF fusions) . NTRK1 , NTRK2.
MAPK pathway gene alterations NTRK3, RAF1, and NF1, and possibly additional genes. The
Essentially all PXAs harbour genetic alterations in a MAPK path- frequency of BRAF mutation is unrelated to CNS WHO grade or
way gene causing aberrant activation ot this pathway. By far elevated mitotic activity \2842,33001 .
the most frequent alteration is BRAF p.V600E (accounting tor
-60% of cases In previous series 12842,7561 and as many as CDKN2A and/or CDKN2B homozygous deletion
80% in combined data from two recent studies 12499,32991) . As many as 94% of PXAs harbour alterations of CDKN2A and/
In most cases , this missense mutation is detectable using or CDKN2B, in most cases 1n the form of homozygous deletion

Gl 1omds . Jlion ·uro1 tdl turnour~ and neurona l tumour 97


Fig. 2.87 Pleomorphic xanthoastrocytoma. A Reticulin deposition is present in approximately 60% of cases. B GFAP expression In large pleomorphic and spindle cells. C s :~·
Is typically diffusely positive. D Neuronal markers are often expressed, in particular synaptophysin, shown here in a large pleomorphic and vacuolated tumour cell. ECW
positivity 1s frequent but variable.

is highly suggestive o'f a diagnosis of PXA. although other rar~


examples of gangliogliomas, epithelioid glioblastomas, arr
high-grade astrocytomas with piloid features have also be€f
reported to have this molecular constellation (2444,1716,220C
2643).

TERT alterations
TERT promoter mutation and (less frequently) TERT ampl1fica
tion have been identified in PXA In varying proportions: they are
more common in anaplastic tumours than in other PXAs (167
1962,2499,3299).

DNA methylation profiling


A DNA methylation profile for PXA has been reported (46 I
which may be particularly useful in tumours with amb1guot. 5
morphology, but it is largely confirmatory In those with cl, ·
sic histology (463,3299). Tumours with a methylation profile ··
PXA (and harbouring combined BRAF p.V600E mutation ar..:
Fig. 2.88 Pleomorphic xanthoastrocytoma. BRAF p.V600E mutation can be detected CDKN2A and/or CDKN28 deletion) can be identified in substar
by 1mmunostaining. tial numbers among histological series of paediatric glioblast~
mas 11721,4601. epithelioid glioblastomas (1716,71) , and astrv·
(2499,3299) . In one study, 18 of 19 tumours showed homozy- blastomas 1317,1848,3474). as well as in occasional embry ,,
gous COKN2A and/or COKN2B deletions , and the remaining tumours !3059), gangliogliomas (460). and atypical teratOJ",
tumour showed loss of protein expression , indicating that inac- rhabdoid tumours {460l . Although it has been suggested tr.a,
tivation ot CDKN2A and/or COKN28 may be even more preva- the morphological spectrum of molecularly defined PXA coL. t
lent than previously thought , and possibly a defining molecular therefore be substantially wider than previously thought. furtll,1.
alteration of PXA {2499) The combination of BRAF p.V600E studies are required before a conclusion can be drawn regar.
mutation and COKN2A and/or COKN28 homozygous deletion ing the def inltive classification of such cases (463) .

98 Gl101neis . g1 10 11 uron.-ll tur11CJIJl'S, ;::rill 11L:urc; 11 :.1 11umou rs


Essential and desirable diagnostic criteria Box2.14 Diagnostic criteria for pleomorphic xanthoastrocytoma
see Box 2.14.
Essential:
An astrocytoma with pleomorphic tumour cells, including large multinucleated cells,
Staging spindle cells, xanthomatous (lipldized) cells. and eosinophillc granular bodies
In contrast to most other types of circumscribed gliomas, PXAs
tend to disseminate during tumour progression . MRI of the spine Desirable:
is recommended at the time of clinical progression . Retlculin deposition
BRAF mutation or other MAPK pathway gene alterations, combined with
Prognosis and prediction homozygous deletion of CDKN2A and/or CDKN2B
PXA behaves in a less malignant fashion than might be sug- A DNA methylome profile of pleomorphic xanthoastrocytoma
gested by its highly pleomorphic histology (1595), but It fre-
quently recurs and is associated with decreased survival
compared with other CNS WHO grade 1 or grade 2 gliomas in
children and young adults , in particular pilocytic astrocytoma.
Furthermore , malignant progression is more common In PXA
than in other RAS/MAPK-driven CNS WHO grade 1 or grade 2
gliomas [1400}. Upon progression , survival is markedly reduced,
even with currently available therapy {2023 ,2127,2768).
Extent of resection is the most significant prognostic factor
associated with recurrence {1092 ,1400). A consistent relation-
ship has emerged between mitotic activity and outcome, and
on th is basis , tumours are divided into CNS WHO grades 2
and 3 [1092 ,1400]. In a retrospective series of 74 patients with
PXA , the 5-year recurrence-free survival rates were 70.9% for ...
patients with grade 2 tumours and 48.9% for those with grade 3 ... J

tumours (P = 0.092). The 5-year overall survival rate of patients --. --;-- . - -. - .- - - -:- .. --:--- . -. ---~ -.--~ . .-:-:--.--=----··
with grade 2 tumours was also significantly higher than that of Flg. 2.89 Pleomorphic xanthoastrocytoma. Chromosomal microarray. Typical copy-
patients with grade 3 tumours (90 .4% vs 57.1%, P = 0.0003). number profile, demonstrating copy-neutral loss of heterozygosity of chromosome 9
Tumour necrosis was significantly associated with lower 5-year with homozygous deletion of CDKN2A and/or CDKN28. Additional whole-chromo-
some gains were present, including gains of chromosomes 4, 14, and 21 (three copies)
overall survival rates (42 .2% when present vs 90.2% when
and chromosome 7 (four copies).
absent, P = 0.0002). But the dataset was too small to detect
a difference in survival between patients whose tumours had
high mitotic counts and necrosis versus those with only necrosis homozygous deletion [1004). TERT promoter alterations may
(1400) . be linked with a more aggressive phenotype and have been
The prognostic significance of CNS WHO grading of PXA proposed as a marker of anaplastic transformation {2499,1353,
has recently been confirmed in two large independent stud- 3299).
ies (1004,3299). When cases that clustered with "methylation Because CNS WHO grade 2 PXA tends to recur, dissemi-
cluster PXA" by DNA methylation profiling (460) were stratified nate, and progress to higher-grade PXA , early intervention with
by tumour grade, the prognostic value of grade was still sig- complete surgical resection is critical and may be followed by
nificant (3299). MAPK pathway gene aberrations , In particular a watch-and-wall strategy after gross total resection {3417).
BRAF p.V600E , as well as homozygous deletion of CDKN2A Patients with CNS WHO grade 3 PXA should be managed with
and/or CDKN28, are central to the underlying genetics of PXA additional therapy (adults probably with postoperative radio-
but are not associated with tumour grade or prognosis {3300, therapy) {3417). Targeted therapies are important to consider
3299). Response to targeted BRAF p.V600E therapy, however, for patients, especially when gross total removal cannot be
is not hampered by the presence of COKN2A and/or CDKN2B achieved, even while their tumours are still lower-grade.

Gliomas , glloneuronal tumours and neuron.:il tumours 99


Lopes MBS
Subependymal giant cell astrocytoma Cotter JA
Rodriguez FJ
Santosh V
Sharma MC
Stemmer-Rachamimov AO

Definition
Subependymal giant cell astrocytoma (SEGA) is a periventricu-
lar tumour composed partly of large ganglion-like astrocytes,
and strongly associated with tuberous sclerosis (TS) (CNS
WHO grade 1).

ICD-0 coding
9384/1 Subependymal giant cell astrocytoma

ICD-11 coding
2AOO.OY & XH1 L48 Other specified gliomas of brain & Sub-
ependymal giant cell astrocytoma
Flg. 2.90 Subependymal giant cell astrocytoma, postcontrast axial T1-weighted
Related terminology MRI. A A right subependymal giant cell astrocytoma near the foramen of Monro, with
Not recommended: subependymal giant cell tumour. avid enhancement. B After 3 months of treatment with an mTOR Inhibitor, the tumour
shows decreased size and enhancement.
Subtype(s)
None Spread
Leptomeningeal dissemination with drop metastases is rare,
Localization having been described only in two cases {3162,34).
SEGAs typically arise from the subependymal tissue of the
lateral ventricles adjacent to the foramen of Monro. Rare loca- Epidemiology
tions include the third ventricle {1925 ,2896) and the retina Incidence
{2378). SEGA is the most common CNS neoplasm in patients with TS
(37,2286,2731). The incidence rate of SEGA among patients
Clinical features with TS is 5-15% {37,2286,2731}, and the tumour is one of the
Most patients present with signs and symptoms of increased major diagnostic criteria of TS 12286). The calculated overall
intracranial pressure. Tumour growth at the foramen of Monro incidence of SEGAs in the US Surveillance, Epidemiology, and
can block cerebrospinal fluid circulation, leading to obstructive End Results Program (SEER) 18 database is 0.027 cases per
hydrocephalus {1127). Massive spontaneous haemorrhage may 100 000 person-years (2245}. It is uncertain whether the tumour
be an acute manifestation {2731). With the current practice of also occurs outside the setting of TS or if it harbours currently
early screening of patients with TS , SEGAs may be diagnosed undetectable TSC gene alterations \2286) .
while still clinically asymptomatic {1745,2731}. Growth of sub-
ependymal nodule(s) (SENs) into a SEGA is usually a grad- Age and sex distribution
ual process, which occurs at the highest rate in the first two This tumour typically occurs during the first two decades
decades ot life {1127}. Marked growth In< 12 months has rarely of life and only infrequently arises de nova after the age of
been reported {2189}. 20-25 years {2286). SEGA can occur in infants and several
congenital cases diagnosed at birth or by anten~tal MRI have
Imaging been reported {1385 ,2061 ,2494 ,2608).
On CT, SEGAs appear as solid, partially calcified masses
located In the walls ot the lateral ventricles, mostly near the tora- Etiology
men of Monro. lpsilateral or bilateral ventricular enlargement may SEGA has a strong association with inherited TS (see Tuberous
be apparent. On MRI, the tumours are usu~lly het~rogeneous, sclerosis, p. 441 ).
isointense, or slightly hypointense on T1-we1ghted images, and
hyperintense on T2-weighted images, with marked contr.ast Pathogenesis
enhancement (1413) . Prominent signal voids, represent~ng Evidence of bialleli~ inactivation of the TSCt or TSC2 gene sup·
dilated vessels , are occasionally seen . SEGAs may show a high ports the .hypothes1~ that SEGAs arise as a consequence of a
choline-to-creatinine ratio and a low ratio of N-acetylaspartat~ to second-hit mec~an1sm (512}. Activation of the mTOR pathwa
creat1nine on proton magnetic resonance spectroscopy, wh1~h has been shown 1n SEGAs, and clinical trials have shown reduc·
seems to be a valuable tool for the early detection of neoplastic t1ons 1n tumour volumes using mTOR inhibitors 19781.
transformation of SEN to SEGA 12517].
Cell of origin Examples of SEGAs in the absence of TS have been reported ,
SEGAs demonstrate glial, neuronal , and mixed neuroglial fea- but these tumours may harbour currently undetectable TSC
tures (morphological, immunohistochemical, an d ultrastruc- gene alterations (e .g. low-level somatic mosa1c1sm or large
tural), suggesting a cell of origin with the capaci ty to underg o deletions) , or they may have other mechanisms of inactivation
differentiation along glial, neuronal, an d neuroen docrine lines (226,3 191 .
(2896 ,1925). This hypothesis has been recently supported by BRAF p.V600E mutations were found in rare SEGAs 1n two
data from mouse models in whic h loss of Tsc 1 or ac tivation of case series (1828 ,2842}, including two patients with "definite"
the mTOR pathway in su bventric ular zo ne neu ral pro genitor TS by clinical criteria . However, BRAF p.V600E mutations were
cells resulted in the formation of SEGA- and SEN -li ke lesions in not identi fied in a recent larger series of 58 SEGAs (319] . DNA
the lateral ve ntric le (1984,2529). SEGA and SEN also have si mi- methylation-based classification studies support SEGA as a
lar histological and radiolog ica l features; the main distinction is distinct tumou r entity (460) .
based on size (SEGAs are:::>: 5 mm , SENs < 5 mm) and growth
over time (which occurs only in SEGA). Radiolog ical evidence Macroscopic appearance
supports the transition of some (5 - 15%) SENs into SEGAs over SEGAs are sharply demarcated , multinodular, solid tumours
ti me, suggesting that these tumours represent a continuum arising from the wall of the lateral ventricle , close to the fora-
(1127,604,327). Several studies showed nuclear expression of men of Monro. Less frequently, they arise in the third ventricle
thyroid tran scription factor 1 (TTF1 , also known as NKX2-1) in (1925 ,2896). Morphologically similar neoplasms may develop
SEGAs . Given that TTF1 expression is transiently present in the inside the eye in association with the retina in patients with TS ,
medial gangl ionic eminence in the fetal brain, this suggests a and outside the ventricles sporadically or in patients with TS or
derivation of SEGAs from a regional progenitor cell (1229,1303). neurofibromatosis type 1 (2378}. The tumou rs show zones of
calcification , often with cystic change and foci of haemorrhage
Genetic profile
SEGAs have a strong association with TS and typically show evi- Histopathology
dence of biallel ic inactivation of TSC1 (15%) or TSC2 (56%), with Histologically, SEGAs are ci rcumscribed , moderately cellular
the second hit frequently observed as deletion or loss of hetero- tumours composed of a wide spectrum of glial phenotypes.
zygosity (1290,2137,319). Lost or reduced tuberln and hamartin Typical appearances range from polygonal cells with abundant.
expression has been described in SEGAs from patients with glassy cytoplasm to smaller spindle cell s and gem1stocyte-
either TSC1 or TSC2germline mutations {1290,1507,i596,2137}. like cells arranged in sweeping fascicles . sheets , or nests with

Fig. 2.91 Subependymal giant cell astrocytoma. A Large cells with voluminous cytoplasm and well-delineated borders may be present. B Elongated cytoplasmic profiles and
a more ganglioid appearance may be a feature .

.- .
flg.2.92 Subependymal giant cell astrocytoma . The CNS WHO grade 1 designation is not changed by mitotic activity (A), by the rare presence of m1crovascular proliferation
-
(BJ. or by necro sis (even if palisading) (C).

GltO nl dS , <J ll011t'!l ll 0r di tu r t •\ ) Ll f S ·in d r, •u1u•1,1! tumc.urs 101


Fig. 2.93 Subependymal giant cell astrocytoma. A GFAP expression is more variable than S100, and absent in individual cells, but it is usually present at least focally. I Tu
mours uniformly express S100. C Class Ill P-tubulin (as recognized by TUJ1) is the most ubiquitous neuronal marker In these tumours. D Markers of neuronal differentiation
including synaptophysln, are frequently positive In these tumours.

Intervening fibrillary septa. Giant pyramidal-like cells with a gan- phenotype it has also been termed "subependymal giant CBI
glionic-like appearance (without Niss! substance) are common; tumour" {1925 ,398). Tumour cells demonstrate variable immu·
these large cells have often eccentric, vesicular nuclei with dis- noreactivity for GFAP and a uniform and intense immunore-
tinct nucleoli. Nuclear pseudoinclusions can be seen in some activity for 8100 (1925,3550,2896). Variable immunoreactiv1ty
cases . Considerable nuclear pleomorphism and multinucleated for neuronal markers and neuropeptides has been detected
cells are frequent. Clustering of tumour cells and a perivascular Neuron-associated class Ill P-tubulin appears more widespreao
rosette-like pattern resembling that of ependymoma are com- in its distribution than any other neuronal epitope, whereas neu·
mon features . A rich vascular stroma with frequent hyalinized rofilament is more restricted and mainly highlights cellular pro·
vessels and infiltration by mast cells and lymphocytes, predomi- cesses and a few ganglionic cells (1925). 8EGAs are variably
nantly T lymphocytes , is a constant feature {2896). Parenchymal lmmunoreactive for synaptophysin (2896). NeuN (3550), ano
or vascular calcifications are frequently seen (2896,1161,2494). neuropeptides {1925) . Neural stem cell markers including nest1n
The presence of mitoses, vascular proliferation , or necrosis and 80X2 are also expressed in SEGAs {2494). but unlike 1n
does not indicate anaplastic progression (3509) . cortical dysplasias, CD34 immunoreactivity is not seen. Loss
of either hamartin or tuberin immunoexpression alone is corn·
Proliferation manly seen in 8EGAs, and rarely a combined loss may be
The proliferation index as measured by Ki-67 (MIB1) immu- present {2136) . In addition , 8EGAs show strong immunoreact1~·
nostaining is generally low (mean: 3.0%), providing further ity for phosphorylated 86, consistent with mTOR pathway act1•
support for the benign nature of these neoplasms (1205, vation (512) . SEGAs show nuclear immunoreactivity for TTF1. a
2895} . The topoisomerase II labelling Index is also reportedly feature shared by other tumours arising from ventral forebraln
low (mean : 2.9%) {2895) . Although extremely uncommon, structures {1303,1658 ,277) . This helps differentiate 8EGA fron1
craniospinal dissemination has been reported in 8EGAs with its close morphological mimics [1229). thus widening the spec·
increased Ki-67 (MIB1) index values but without other malig- trum of TTF1-positive CNS tumours.
nant features {3162) .
Ultrastructure
lmmunophenotype Ultrastructural features of neuronal differentiation, including
8EGA has been designated as a distinctive, well-circumscribed microtubules, occasional dense-core granules, and (rarely)
astrocytoma, but because of its usually mixed glioneuronal synapses , may be detectable; bundles of intermediate filaments

102 Gl 1ornas . gliom::uronal tumo urs . and neuronal tumours


are seen 111 the cytoplasmic processes of the spindled astro- Box2.15 Diagnostic critena for subependymal giant cell astrocytoma
cyt1c ce lls (1 3 17. 150 6 .3 981
Essentis/:
CharacteristJc histological features, with multiple gllal phenoty~es in~fuding
Cytology polygonal cells, gem1stocyte-like cells, spindle cells, and ganghornc-ltke cells
Cytological preparations of SEGAs show the diverse cellu-
AND
lar elements that constitute the tumours , including elongated
spindle-shaped and strap cells with long , thick cell processes lmmunoreactlVity for glial markers (GFAP, S100)
to more pleomorph1c b1nucleated or multinucleated cells . The AND
combination of cytological features with clinical and radiological Vanable expression of neuronal markers (class 111 JHubulin, neurofilament.
findings can be diagnostic of SEGAs in intraoperat1ve consulta- synaptophysin, NeuN)
tions 11449.2 2131 . Desirable:
Nuclear immunoexpression of thyroid transcription factor ·1 (TTF1)
Diagnostic molecular pathology
Lost or reduced expression of tuberin and hamartin
Molecular analyses are usually not needed to establish the
lmmunoexpress1on of phosphorylated S6
diagnosis of SEGA In histologically ambiguous cases, DNA
methylome profiling and analyses for TSC 1 or TSC2 alterations DNA methylome profile of subependymal giant cell astrocytoma
may help to establish the diagnosis. History of tuberous sclerosis
TSC1 or TSC2 mutation
Essential and desirable diagnostic criteria
See Box 215 . lesions tend to have greater morb1d1ty 1713). Careful follow-up
of residual tumour is recommended because of the potential
Staging for late recurrences. Optimal outcome is associated with early
Not clin ically relevant detection and treatment. In individuals with TS, surveillance
by MRI every 1-3 years until the age of 25 years 1s recom-
Prognosis and prediction mended {713,1745) . Inhibition of mTOR with everolimus has
Patients with SEGAs have a favourable prognosis when gross been reported to result in significant reduction of tumour size
total resection of the tumour is achieved. Larger or symptomatic and control of SEGA progression {976 ,977.978) .

Gl1omas, gl1oneurona l tumours and neuronal turnours 103


Chordoid glioma Fuller GN
Brat DJ
Kleinschmidt-DeMasters BK
Sanson M
Solomon DA

Definition
Chordoid glioma is a well-circumscribed gllal neoplasm that
arises in the anterior third ventricle, is histologically charac-
terized by clusters and cords of GFAP-expressing epithelioid
cells , and exhibits a recurrent p.0463H mlssense mutation in
the PRKCA gene (CNS WHO grade 2).

ICD-0 coding
9444/1 Chordoid glioma

ICD-11 coding
2AOO.OY & XH9HV1 Other specified gliomas of brain & Chor-
doid glioma

Related terminology Flg.2.94 Chordoid glioma. A Axial MRI from a tumour in a 67-year-old man shows
Not recommended: chordoid glioma of the third ventricle. the typical imaging features of chordoid glioma, including sharp circumscription, large
size, contrast enhancement, and compression of nearby structures. B Sagittal MRI
from the same patient shows the tumour located in the anterior third ventricle. Nole
Subtype(s)
the lack of Involvement of the pituitary gland, lateral ventricle, and corpus callosum.
None
approximately 45 years , although age at presentation varies
Localization widely (5-71 years). A female predominance has been noted
~hordoid gliomas have a stereotypical location in the anterior (M:F ratio: 1:2) {366,743,104}.
portion of the third ventricle, with larger tumours filling the mid-
dle and posterior aspects {2533}. They arise in the midiine and Etiology
displace normal structures as they enlarge. Neuroimaging find- No risk factors or inherited genetic susceptibility have been
ings suggest an origin in the region of the lamina terminalis in reported .
the ventral wall of the third ventricle {1844,2412) .
Pathogenesis
Clinical features Two independent studies identified a novel missense mutation
Presenting signs and symptoms typically reflect obstructive affecting codon 463 of the PRKCA gene as the molecular hallmark
hydrocephalus, with headache, nausea, vomiting, and ataxia alteration {1139,2726). PRKCA encodes the catalytic a-subunit of
1366,743). Other features may include endocrine abnormalities PKC, which functions In intracellular signalling downstream of
reflecting hypothalamic compression (hypothyroidism, amen- multiple transmembrane receptors. Although PRKCA is occa-
orrhoea, diabetes insipidus); visual field disturbances due to sionally mutated in other cancers, this specific p.D463H muta-
compression of the optic chiasm; and personality changes, tion has not been re~orted in other human tumour types to date.
psychiatric symptoms, or memory abnormalities . The mutation .re~ults 1n th~ su~stitution of histidine for aspartate at
codon 463 w1th1n the active site of the kinase domain, where the
Imaging side chain of aspartate normally functions as the proton acceptor
Chordoid gliomas are well-circumscribed ovoid or multilobu- du:ing the A:P hydr~lysis rea_ ction . The precise oncogenic mech-
lated masses within the anterior third ventricle. MRI shows T1 anisms of this mutation remain to be elucidated, but the mutation
isointensity to brain and strong homogeneous enhancement m_ay modify substrate speci'ficity or catalytic activity (1139,2726).
{2533) . Mass effect is distributed symmetrically, with vasogenic High levels of phosphorylated E~K have been found, suggesting
oedema in compressed adjacent CNS structures, including the that the PRKCA p.D463H mutation may function at least in part 1

optic tracts , basal ganglia, and internal capsules. Most tumours by activating the MAPK signalling pathway {1139 } .

are continuous with the hypothalamus; some appear to have an


Intrinsic anterior hypothalamic component, suggesting a poten- Cell of origin
tial site of origin 11844). On the basis of anatomical location, consistent immunoreactiv-
i!Y for thyroid transcription factor 1 (TIF1 ), and ependymoma·
Epidemiology like ult'.astructural feat~r~s, chordoid gliomas are hypothesized
Chordoid gliomas account for< 0.1% of primary brain tumours. to originate from specialized tanycytic ependymal cells of the
They most frequently occur in adults, with a median age of organum vasculosum of the lamina terminalis {497,24 12,277).

104 Gl1ornas , gl1oneuronal tumours , and neuronal tumours


'1

~ "-
FIg. 2.95 Chordo1d glioma. A The demarcation between the tumour and adjacent brain tissue is often sharp; individual cell infiltration is not seen. The adjacent brain tissue
in this example shows chronic inflammation and numerous Rosenthal fibres. a Typical histological appearance of a chordoid glioma composed of cords and clusters of epi-
thelloid tumour cells in a myxo1d stroma, resembllng the notochordal tumour chordoma. C This chordoid glioma is composed of plump epithelioid cells in a prominent myxoid
stroma. D This chordoid glloma is composed of bipolar spindled cells In a prominent myxold stroma. E The lymphoplasmacytic infiltrates in these tumours can have plasma
cells containing eos1nophilic small globular Russell bodies. F Chordoid glioma demonstrating a dense rim of lymphoplasmacytic Inflammation at the periphery of the tumour.

Macroscopic appearance often containing numerous Russell bodies . is a common find-


Chordoi d gliomas are well demarcated , often multilobulated , ing . Consistent with the imaging appearance. th ere is little
and typical ly soft , grey, and gelatinous. tendency for brain infiltration . Reactive astrocytes, Rosenthal
fibres , and chronic inflammatory cel ls may be seen 1n adjacent
Histopathology non-neoplastic tissue.
Chordoid gliomas are sol id neoplasms , most often composed of
clusters and cords of epithelioid cells within a variably mucinous lmmunophenotype
stroma. Th ree less common histological patterns have been Chordoid gllomas show strong , diffuse expression of GFAP
reported : a solid pattern with sheets of polygonal epithelioid {366 ,2800] and consistently express the transcription factor
cells without appreciable mucinous stroma, a fusiform pattern TTF1 (NKX2-1). The percentage and intensity of nuclear TIF1
with groups of spindle-shaped cells among loose collagen , and staining vary depending on the antibody c lone used [277). with
a fibrosing pattern with abundant collagenization . The fibrosing rare examples showing minimal to no expression . Expression of
pattern tends to be more common in older patients (277] . Individ- vimentin and CD34 is strong . Expression of 8100 and EMA is
ual tumour cells have abundant eoslnophilic cytoplasm . Nuclei variable. Neuronal and neuroendocrine markers (synaptophy-
are moderate 1n size, ovoid , and relatively uniform . Mitoses are sin, neurofilament, chromogranin A) are consistently negative
usually rare or absent. A stromal lymphoplasmacytic infiltrate, {2639]. The Ki-67 proliferation index is usually < 2% {366 1.

.·- .
,, ..... . .. ... . .
..-.·. ..
~' '
-:.

'

.- , ,..-..., . --·_
,

.
~

• '• "" ~_,

.~
' . . .c .. , #
.....
• ..
-..
.;.
.~ -
' •
~ -~
... ..

Fig. 2.96 Chordold glioma. A All tumours show diffuse cytoplasmic 1mmunoreactlvity for GFAP, as shown here. Note the lymphoplasmacytic infiltrates that are GFAP-immu-
noneg~t ive , shown in the lower right of this image. B Strong diffuse cytoplasmic immunost~inin~ for CD34 is typically seen in chordoid glioma. c Nuclear positivity for thyroid
transcription factor t (TTF1 , a homeobox transcription factor encoded by the gene NKX2-1) 1s typical of chordold gliomas.

Gl1 ornas . glloneur nal tumours . and neuronal tumo urs 105
Differential diagnosis Box2.16 Diagnostic criteria for chordold glioma
The principal tumour types in the differential diagnosis are Essential:
other chordo1d neoplasms, including chordoma and chordoid A glial neoplasm with chordoid features located In the anterior third ventricle
meningioma
Desirable:
Chordomas can be differentiated based on their consistent
expression of brachyury and cytokeratins, with lack of GFAP Nuclear thyroid transcription factor 1 (TTF1) immunopositivity
and CD34 expression . PRKCA p.D463H mutation or DNA methylation profile aligning with chordoid glioma
Chordoid meningiomas usually display small foci of whorl for-
mation and psammoma bodies , and they are immunopositive
for EMA and SSTR2A , but negative for GFAP and CD34 (2800). pathogenic alterations in genes characteristic of other brain
as well as being negative for TTF1 (1740) . tumour entities (e.g . !OH1, !DH2, H3-3A. H3C2 [HIST1H3B]
The differential diagnosis also includes epithelioid haeman- FGFR1, BRAF, NF1, CDKN2A, TP53) . A distinct epigenetic sig-
gioendothelioma, which is composed of cords of cells , some- nature of chordoid glioma has also been identified, and DNA
times with a myxoid/mucinous background . Although both methylation profiling represents an ancillary method for diag-
tumours share immunoreactivity for CD34, epithelioid haeman- nostic confirmation [463).
gioendothelioma is additionally positive for CD31, VEGF, and
factor VIII, but negative for GFAP {3077) . Essential and desirable diagnostic criteria
See Box 2.16.
Cytology
Not clinically relevant Staging
Not clinically relevant
Diagnostic molecular pathology
The p.D463H missense mutation in the PRKCA gene is nearly Prognosis and prediction
ubiquitous in chordoid glioma, having been found in 28 of Factors impacting morbidity, mortality, and recurrence have not
29 tumours studied to date {1139,2726) . This mutation has not been clearly elucidated. The treatment of chordoid glioma 1s
been identified in any other human tumour, although the PRKCA based on maximal tumour resection while avoiding complica-
gene is involved in fusions in papillary glioneuronal tumou r tions such as diabetes insipidus and other endocrine dysfunc-
11354). Therefore, the PRKCA p.D463H mutation is a diagnos- tions {104) . Radiotherapy may be considered in patients with
tic hallmark. Chordoid gliomas have lacked accompanying su btotal resection , but the benefit is not well established.
Orr BA
Astroblastoma, MN1-altered Brat DJ
Aldape KO Rosenblum MK
ldbaih A Solomon DA
KoolM Sturm D

Definition
Astroblastoma , MN1-altered, is a circumscribed glial neoplasm
with MN1 alteration that is composed of round. cuboidal, or
columnar cells with variable pseudopapillary or perivascular
growth . perivascular anucleate zones, and vascular and peri-
cellular hyalinization .

ICD-0 coding
9430/3 Astrobl astoma, MN1-altered

ICD-11 coding
2A00.4 & XH1 DC5 Astroblastoma of the brain & Astroblastoma

Related terminology Flg.2.97 Astroblastoma, MNt-altered. Postcontrast, T1 -we1ghted MRI (A) and T2-
Not recommended: CNS high-grade neuroepithelial tumour weighted MRI (B) show a large, well-demarcated, contrast-enhancing, solid and cystic
with MN1 alteration . right temporoparietal mass.

Subtype(s) specific mechanism by which MN1 fusions drive tumour devel-


None opment remains unknown.

Localization Macroscopic appearance


Astroblastoma , MN1-altered , occurs predominantly in the cer- The gross appearance of MN1-altered astroblastoma has not
ebral hemispheres. most often in the frontal and parietal lobes, been described . Histologically defined astroblastomas are grey-
but also in occipital and temporal regions !546] . lntraventricular, ish-pink or tan and have well-demarcated borders with adjacent
brainstem , and spinal cord examples have been documented brain. Foci of necrosis or haemorrhage may be present.
{3512,2914,546) .
Histopathology
Clinical features The histological hallmark of MN1-altered astroblastoma is the
Presenting symptoms include headache, seizures , paralysis , astroblastic pseudorosette , a perivascular structuring of neo-
nausea, and vom iting {1315 ,2103) . plastic cells that appear radially arrayed , often forming papil-
lary or pseudopapillary formations in cross-section , and regi-
Imaging mented in ribbon-like/trabecular alignment in tangential views
On MRI , MN1-altered astroblastomas are well-demarcated , (360 ,3059,3474 ,1315,1 848 ,2103}. In the classic form , tumour
solid or cystic masses that are isointense or hypointense on T1 cells are anchored to centrally positioned blood vessels by
imaging and hyperintense on T2 imagin g, with heterogeneous eosinophilic cytoplasmic processes that are well defined , stout,
contrast enhancement and perilesional oedema {1315,2103] . or only slightly tapered . These lend inverted columnar or low
cuboidal profiles to neoplastic elements . Round cell elements
Epidemiology may also be found , which may mimic a primitive or embryo-
Patient ages range from 3 months to 40 years at clinical pre- nal neoplasm in hypercellular regions . Rhabdoid cytology can
sentation (median: 15 years) [546 ,3512] . These tumours have a also be encountered (2914). Astroblastic pseudorosettes may
striking female predominance, with women accounting for 39 of only focally emerge against a background of sheet-like tumour
41 reported cases included in a recent meta-analysis 1546}. growth . Spindled cells forming fascicles may be encountered. A
typical (though inconstant) and especially prominent feature of
Etiology MN1-altered astroblastomas in some studies {18481 is vascular
Acquired fusions involving the MN1 gene play a key pathogenic and stromal sclerosis, which may be limited or extensive. with
role in this tumour type . There is no known specific genetic sus- broad regions of hyallnization containing only remnant tumour
ceptibility for MN1-altered astroblastoma. cell cords . Circumscription is the rule , with pushing borders
or only limited CNS invasion being characteristic. Permeative
Pathogenesis growth in the manner of diffuse gliomas is not a feature . Mitotic
Elevated expression of MN1 fusion partners BEND2 and CXXCS activity is highly variable and some cases exhibit necrosis and
suggests an activating, gain-of-function event (3059). but the mlcrovascular proliferation . Histological features for grading

Gliomas . glioneuronal tumours , and neuronal tumours 107


Fig. 2.98 Astroblastoma, MN1-altered. A The histological hallmark is the astroblastic rosette, characterized by radially oriented tumour cells with elongate, stout processes and
distally located nuclei extending to a central vascular structure. B A ribbon-like or trabecular pattern is noted when elongated tumour cells oriented to central blood vessels are
seen in cross-section. C This tumour included focal rhabdoid cytological features. D Perivascular and pericellular hyalinization is typical but varies considerably in its severify
and extent. E Note the sharp demarcation from adjacent brain parenchyma.

have not been clearly defined and a definitive CNS WHO grade astroblastomas , as have cell body polarization with investing
has not been established for MN1-altered astroblastoma. basement membranes, lamellar cytoplasmic interdigitations
(pleating), and apical cytoplasmic blebs with purse-string con-
Electron microscopy strictions and capping microvilli {1754,1749).
An ultrastructural study of an MN1-altered astroblastoma
revealed intercellular lumina containing microvilli and framed by lmmunophenotype
elongated , zonula adherens-type cytoplasmic junctions {2914). Cytoplasmic immunoreactivity for GFAP is characteristic.
Such features have also been noted in histologically defined although the extent varies considerably. The large majority
of MN1-altered astroblastomas display at least focal nuclear
OLIG2 expression (3474 ,1315,1848,2103,3147} . Cytoplas-
mic EMA labelling is regularly seen but varies in its distribu-
tion as diffuse, membranous , dot-like, or ring-like (3474,1315,
2103 ,3147) . lmmunoreactivity for podoplanin (02-40) is typical
(1315) . The MN1-altered astroblastomas studied to date were
not immunoreactive for L1CAM (a surrogate marker of ZFTA
[C11orf95] fusion - positive ependymomas) (3474,1315) . A broad
range of Ki -67 labelling index values has been communicated
(360 ,1315,1848,2103) .

Differential diagnosis
The histological features noted in MN1-altered astroblastoma
are not entity-specific and may be displayed focally or exten-
sively by other tumours that are proved, on molecular diag -
nostic assessment, to represent ZFTA (C11orf95) fusion-posi-
tive ependymomas , BRAF-mutant epithelioid glioblastomas,
BRAF-mutant pleomorphic xanthoastrocytomas, embryonal
neoplasms , IDH-wildtype glioblastomas , or other gliomas
(3474 ,1848,317,546,3147,3059) . ZFTA (C11orf95) fusion and
BRAF mutation are mutually exclusive with MN1 alterations
and do not support a diagnosis of MN1-altered astroblastoma .
Although a small subset of MN1-altered astroblastomas may
harbour genetic alterations typical of IDH-wildtype glioblastoma
(e.g. EGFR amplification, or homozygous deletion involving the
CDKN2A and/or CDKN28 locus), IDH -wildtype glioblastomas
are characterized by an infiltrative pattern of growth, distinc-
tive cellular morphology, additional genetic alterations (e.g.
TERT promoter mutations, gain of chromosome 7, loss of chro-
mosome 10), and an absence of MN1 alterations. A subset of
histologically defined astroblastomas do not share a molecu-
lar signature with currently established molecular CNS tumour
types {3474) .

Cytology
The intraoperative smear/squash preparation cytology of MN1-
altered astroblastomas has not been described.

Diagnostic molecular pathology


Astroblastoma, MN1-altered, ls characterized by structural
rearrangements of the MN1 gene at chromosome band 22q12.1.
MN1 fusions most often occur in-frame with BEND2 at chromo-
some band Xp22.13, but other partners (including CXXC5) have
also been described {3059,3474,2103,1315,1848). MN1 altera-
tions can be detected by a variety of methodologies, includ-
ing FISH , RT-PCR , RNA sequencing, and next-generation
DNA sequencin g (3059,3474,1315,1848,317). Although MN1
fusion is often the solitary pathogenic alteration identified, a
subset of MN1-altered astroblastomas harbour accompanying
CDKN2A homozygous deletion (3474 ,1848}. Recurrent chro-
mosomal copy-nu mber changes in MN1-altered astroblastoma
include monosomy 16 and partial losses of 22q and X, probably
reflecting the chromosomal rearrangement process leading to
MN1 ::BEND2 fusion (3059 ,3474 ,1315,1848}. Individual exam-
ples of astroblastoma-like tumours harbouring EWSR1 :: BEND2
fusion instead of MN1 :: BEND2 in the spinal cord have been occur in as many as 70% of histologically defined astroblas-
reported (3512) . However, the biological nature and clinical tomas , and > 60% of MN1 fusion-positive CNS tumours show
outcomes of these rare cases have not been elucidated to astroblastoma morphology (3059,3474,2103,1315 ,1848,546).
date , and the designation "not elsewhere classified (N~C)" MN1-altered astroblastomas display a distinct DNA methylation
is recommended for such tumours at present. MN1 fusions pattern that rel iably d istinguishes them from other tumour types

Gl1omas, glioneuronal tumours , and neuronal tumours 109


with astroblastomatous rosettes (3059,3474 ,1315,460.18481 Box2.17 Diagnostic criteria for astroblastoma. MNt-altered
(see Differential diagnosis. above) . Essential:
A glial neoplasm with astroblastJc perivascular pseudorosettes
Essential and desirable diagnostic criteria
AND
See Box 2.17.
MN1 alteration
Staging AND (for unre olved lesions)
Not clinically relevant ONA methylatJon profile of astroblastoma, MN1·altered
Desirable:
Prognosis and prediction GFAP immunoreactlvily
Among histologically defined astroblastomas. high-grade his-
EMA lmmunoreactlvity
tology has been found to be associated with recurrence. tumour
progression, and worse prognosis !321 .31741 . Outcome data
for patients with MN1-altered astroblastoma are limited. and the
relationship between specific clinical. histological. or molecu- sur ival rates at 5 years and 1O years are close to 90% and
lar features and outcome has not been established. In the fe 50%. respectively, conservative management may be war-
cohorts with molecularly confirmed cases. MN1-altered astro- ranted (1848.5461 . Radiotherapy and chemotherapy seem ben-
blastomas are characterized by frequent local recurrence but eficial when surgery is not feasible (411,2103,2078). Aside from
good overall survival !3059,3474 .18481. Maximal safe surgery surgical resection, no additional prognostic factors have been
is associated with longer survival {3147}. Considering that the identified (31471 .

110 Gllomas . glioneuronal rumours , and neuronal tumou rs


Ganglioglioma Solomon DA
BIOmcke I
Capper D
Gupta K
Varlet P

Definition Table2.05 Localization of gangliogliomas


Ganglioglioma is a well -differentiated, slow-growing glioneu- Aelalvt
LocalJzatJon Total cases
ronal neoplasm composed of a co mbination of neoplastic frecp!ncy
ganglion and glial cells , which is molecularly characterized by
Temporal 604 77%
genetic alterations that cause MAPK pathway activation (CNS
WHO grade 1). Frontal 58 7%

Parietal 24 3%
ICD-0 coding 22 3%
Occipital
9505/1 Ganglioglioma
Multiple lobes 63 8%
ICD-11 coding Other sites 15 2%
2A00 .21 & XH5FJ3 Mixed neuronal -glial tumours & Gangli-
Data from 786 surgical specimens submitted to the German Neuropathology Reference
oglioma, NOS Center for Epilepsy Surgery.

Related terminology
None tumours involving the brainstem and spinal cord , the mean dura-
tion of symptoms before diagnosis is 1.25 years and 1.4 years,
Subtype(s) respectively {1801). Gangliogliomas have been reported in
None 10- 25% of patients undergoing surgery for control of seizures
(3177,304}. They are the tumours most commonly associated
Localization with chronic temporal lobe epilepsy {297,304}.
These tumours can occur throughout the CNS, Including in
the cerebrum , brainstem , cerebellum , spinal cord , and optic Imaging
nerves, as wel l as within the ventricular system, although the The neuroimaging appearance of gangliogliomas is variable,
majority(> 70%) occur in the temporal lobes (Table 2. 05) {2562, but they often display a mix of solid and cystic components. The
3467,1801 ,1316,306}. classic imaging features describe a well-del ineated, T1 -hypoin-
tense, T2-hyperintense cyst, with an enhancing nodule. How-
Clinical features ever, the contrast enhancement pattern is variable. Scalloping
The symptoms vary according to tumour size and site. Tumours of the calvaria may be seen in cortically based tumours. Calci-
in the cerebrum are frequently associated with a history of focal fications may be detected . No imaging characteristics (cystic
seizures , wh ich ranges in duration from 1 month to 50 years component, tumour size, contrast enhancement) have been
before d iagnosis (typically 5-10 years) (2562,3467,1801 }. For shown to be significantly associated with morphological fea-
tures or tumour genotype {2444}.

100
Epidemiology
90 A popu lation-based study calculated a yearly incidence rate of
IO --~ ganglioglioma of 0 .186 cases per 100 000 population world-
-1 wid e, without significant ethnicity proclivity {677}. Ganglioglio-
70
i I mas have been reported in patients ranging from o to 70 years
5 60
'5 of age, most occurring in the first and second decades of life
I
~

i
50

- I
-1 (median age at diagnosis: 12 years) {1802,304). In a single-cen-
tre study, ganglioglioma was more prevalent in male patients
:I 40 i-
E (59.8%) than In female patients (40.2%), and there was a similar
a 30
ratio in the European Epilepsy Brain Bank cohort {799,304}.
20

10
0 LL-~~~~~~~~~~~~~~...__~~

0 10 '° 40
50
l 70
Etiology
The vast majority of gangliogliomas are sporadic tumours.
However, a small subset (< 2%) arise in the setting of neurofi-
Age 8t d)agnosil bromatosis type 1 due to germline mutations or deletions in the
Flg. 2.101 Ganglioglioma Cumulative patient age distribution of ganglioglioma at di- NF1 tumour suppressor gene (2695,11 ·14.2444}. No known risk
agnosis, based on 887 cases from the German Neuropathology Reference Center for factors or environmental exposures have been linked with gan-
Epilepsy Surgery. glloglioma .

Gl1omas , glioneuronal tumou rs. ancJ neur n.JI tumour::; 111


Fig. 2.102 Ganglioglioma. A solid and cystic neoplasm in the temporal lobe of the brain. A Axial FLAIR MRI. B Coronal FLAIR MRI. C Axial T2-weighted MRI.

Pathogenesis other oncogenic mutations in the BRAF gene are often present,
Gangliogliomas result from genetic aberrations causing activa- including recurrent small in-frame insertions at codon p.R506
tion of the MAPK signalling pathway that drives cell prolifera- in approximately 10% of cases (2444). BRAF gene fusions are
tion . Most common are p.V600E hotspot mutations in the BRAF recurrently present in gangliogliomas lacking BRAF mutations,
gene, which lead to the substitution of glutamic acid for valine at most commonly with KIAA 1549 as the fusion partner in spinal
amino acid 600 within the P-loop of the serine/threonine kinase cord tumours, and with other fusion partners in tumours within
domain that causes constitutive activation. BRAF p.V600E the cerebral hemispheres {2444). Gangliogliomas with wildtype
mutation has been found in gangliogliomas at frequencies rang- BRAF alleles instead display a diverse array of other genetic
ing from approximately 10% to 60% depending on the study alterations that similarly cause activation of the MAPK pathway,
and anatomical site, with the highest frequencies in cortical which include RAF1 gene fusions, activating KRAS mutations.
tumours and the lowest in spinal cord tumours {783,2842,1682, and inactivating NF1 mutations or deletions {2444}.
523,3597,666,2616,780,1190,2584,1066,2368,548,3045,2444, The pathogenesis of ganglioglioma was addressed in a trans-
2768). In gangliogliomas lacking p.V600E hotspot mutations, genic mouse model engineered to express Brat p.V600E muta-
tions {1687}. When the mutation was successfully integrated into
neuronal cell progenies , 90% of the mice showed spontaneous
tonic-clonic seizures, which could be prevented with the small-
molecule RAF inhibitor vemurafenib . The tumorigenic proper-
ties were mostly due to Brat p.V600E integration into the glial
cell lineage. These studies experimentally confirmed the long-
term observation that tumorigenesis in ganglioglioma is related
to the glial component, whereas the epileptogenic phenotype
associates with the neuronal component {306}.

Macroscopic appearance
Gangliogliomas are macroscopically well-delineated solid or
cystic lesions, usually with little mass effect. Calcification may
be observed . Haemorrhage and necrosis are rare .

Histopathology
Gangliogliomas are biphasic tumours composed of a variable
admixture of neuro~al and glial elements, which may exhibit
marked hete:ogene1ty. The two components may be intermixed
or geographically separate. The neuronal element is composed
of dysmorphic ganglion cells that may demonstrate abnormal
clu~tering, a lack of cytoar~hitectural organization, cytomegaly,
penmembranous aggregation of Nissl substance or binucle-
ate.d forms .(seen in < 5?% ~f cases) . The glial 'component
which constitutes the prol1ferat1ve cell population of the tumour,
may resemble fibrillary astrocytoma, oligodendroglioma, or
pilocytic astrocytoma (1190,1066,2444} . Eosinophilic granular
bodies are encountered more of ten than Rosenthal fibres (346 7.
B 24~4} . ~!though gangliogliomas are generally well demarcated
Fig. 2.103 Gangl1oglioma. A Ganglioglioma of the right medial temporal lobe. Coronal
. r1orna cen tred in the on 1mag1ng, they often demonstrate an infiltrative growth pattern
postmortem section. B Surgical resection specimen of a gang I1og
cerebral hemisphere, demonstrating a discrete mass lesion with a cystic component. microscopically (1190,297,296,299,2953) . Extension into the
. .-. .;

.
I
. ~

~ • j

, •
: ~. ... .
•. ·Ji. •
~.

.......
4"' ·
~ l ·.1, : •
41: .. .., r!l... . . . ~-.
. ~ • '. ... .. • •
, .,, .
JI' ~ •
.J ....... ~
• 4 1 " .· •
.. ' .~ -~ ' • •

,' .,.... ?... •


... . ~ •• ~. . :I ~ • ' ·- / ·:--·:'!-
•.. •. ,
,.,
-
:
I
• . . <" ./
·~
,. ; _. .
. "'··

. ., "· H .. ~ .: , ~ ,.. -:-


,., ..,.,
\

G. ; "" · • •• ' •• J - l. . ...

Flg.2.104 Ganglioglioma. A Gangliogliomas are glioneuronal neoplasms composed of an admixture of neoplastic ganglion and glial cells. B The neoplastic ganglion cells in
this ganglioglioma show abnormal clustering and cytomegaly. C The neoplastic ganglion cells in this example demonstrate frequent binucleated forms. D The neoplastic glial
cells in this ganglioglioma feature astrocytic morphology. E Eosinophillc granular bodies are a common finding In gangliogliomas. F Despite a well-circumscribed appearance
on radiological imaging, gangliogliomas often microscopically permeate into adjacent cerebral cortex. G This ganglioglioma shows dystrophic calcification that is encrusting
capillaries and neuronal cell bodies. H Gangliogliomas frequently demonstrate perivascular lymphoplasmacytic infiltrates.

subarachnoid space is common . Gangliogliomas may uncom- and usually limited to the glial component {2562 ,1316). The
monly develop a reticulin fibre network apart from the vascula- VE1 antibody that recognizes BRAF p.V600E-mutant protein
ture (which is a feature more typical of pleomorphic xanthoas- can be used with appropriate controls to identify the subset
trocytoma). Mitotic activity is typically low or absent. Additional of gangliogliomas that are positive for this genetic alteration
histopathological features frequently include dystrophic calcifi- (1682). However, the immunostaining intensity levels are typi-
cation , either within the matrix or as neuronal/capillary incrusta- cally lower than those observed in BRAF-mutant melanomas
tion ; extensive lymphoid infiltrates along perivascular spaces; and other tumours, and in some gangliogl iomas the positivity
and a prominent capillary network {306). is most prominent in or exclusively limited to the ganglion cell
Focal cortical dysplasia (FCD) arising in association with component (1682) .
ganglioglioma is a frequently reported finding , but it remains
controversial whether this actually represents infiltration of gan- Grading
glioglioma or non-neoplastic dysplasia in most cases {305). Ganglioglioma is a CNS WHO grade 1 neoplasm. However,
It should be diagnosed only in areas of cortical abnormalities gangliogliomas with anaplasla in the glial component (termed
without tumour cell infiltration and classified as FCD type 3b "anaplastic ganglioglioma"), with features including conspicu-
according to the classification proposed by the International ous mitotic activity, high Ki-67 proliferation index, necrosis, and
League Against Epilepsy (ILAE) {305,200) . microvascular proliferation, have been reported both at initial
presentation and at time of recurrence {2562 ,1460,1801 ,1959,
lmmunophenotype 1562,1955,1066,3578,3167}. However, most of these prior stud-
Neuronal markers such as MAP2, neurofilament, chromogra- ies lacked molecular analysis to exclude other high-grade gli-
nin A, and synaptophysin highlight the neuronal component oma subtypes. Further studies are needed to confirm the exist-
in gangliogliomas {303). However, to date, there is no specific ence of anaplastic ganglioglioma and establish its diagnostic
marker to unequivocally differentiate neoplastic neurons from criteria.
their normal counterparts . Chromogranin A expression Is usu-
all y very weak or absent in normal neurons, whereas diffuse Differential diagnosis
and strong expression suggests a neoplastic neuron. Addi- The diagnosis of ganglioglioma requires the histological iden-
tionally, neoplastic neurons typically have low or absent NeuN tification of a neuronal component, but the near-normal mor-
expression , in contrast to normal cortical neurons. lmmuno- phology of the neuronal component in some cases remains a
histochemistry tor GFAP and OLIG2 highlights the neoplastic challenging issue because there is no specific marker protein
glial cell component 11316). As many as 80% of gangliogliomas 'for differentiating the neoplastic ganglion cells from native neu-
contain ram ified cells either within the tumour or in the adjacent rons . The diagnosis of ganglioglioma should be considered
cortex that express the oncofetal epitope CD34. which is not in all cases of low-grade neuroepithelial tumou rs associated
normally expressed outside of vascular endothelial cells in the with focal seizures that are difficult to classify, especially when
mature brain 1300,306) . Ki -67 labelling is typically low(< 5%) located In the temporal lobe.

Gl1om s, glioneuronal tumours , . nd rieurond.I tumours 113


:'> r. ' ! r'
"'., A"-:
Fig. ~.105 Ganglioglioma. A Syn.aptophysln immunostaining highlights the neoplastic ganglion cells in gangliogliomas. B The glial component of gangliogliomas demonstrates
labelling for GFAP. C Many ganghogliomas contain CD34-positive ramified or stellate cells either within the tumour or In the adjacent cerebral cortex. D This ganglioglioma
demonstrates diffuse strong expression of BRAF p.V600E-mutant protein.

The differential diagnosis for ganglioglioma can include FCO, of C034 immunoreactivity, and pathogenic FGFR1 alteration
diffuse gliomas, and other glioneuronal tumours, specifically all provide support for a diagnosis of ONT over ganglioglioma
dysembryoplastic neuroepithelial tumour (ONT), polymorphous (299). Abundant calcifications, diffuse strong C034 staining of
low-grade neuroepithelial tumour of the young (PLNTY), and tumour cells, and FGFR2 or FGFR3 fusions all provide support
multinodular and vacuolating neuronal tumour. The differentia- for a diagnosis of PLNTY over ganglioglioma {1384) . Multinodu-
tion from FCD rests principally on the presence of a convinc- lar growth pattern, stromal and neuronal vacuolation . OUG2
ing neoplastic glial component in ganglioglioma; in challenging positivity of neoplastic neuronal cells, and MAP2K1 mutation
cases, the identification of BRAF p.V600E mutation or other all provide support for a diagnosis of multinodular and vacu-
MAPK pathway alterations provides support for a diagnosis of olatin~ neuron~I tumo~r ?ver ganglioglioma {1383,2443). Lastly
ganglioglioma, because FCD is genetically characterized by ganglion cell d1fferent1at1on has been reported in a wide spec·
alterations in the Pl3K/AKT/mTOR pathway and an absence trum of CNS tumour entities (e.g . central and extraventricular
of MAPK alterations {305,2016}. As discussed above, identify- neurocytoma, diffuse leptomenlngeal glioneuronal tumour.
ing the neoplastic neuronal component of ganglioglioma c.an papillary g.lioneuronal tumour, pleomorphic xanthoastrocytoma,
be challenging , and diffuse gliomas with entrapped native paragangl1oma of the cauda equina, H3 G34-mutant diffuse
neurons can enter the differential diagnosis in such cases . hemispheric glioma, CNS neuroblastoma), which require dif·
Reduced or absent NeuN expression in the neuronal cells, ferentiation from ganglioglioma. The presence of xanthomatous
eosinophilic granular bodies , and CD34-positi~e r~mlfi~d cells tumour cells, intercellular basement membrane deposition (as
can all provide support for a diagnosis of gangl1ogl1oma instead highlighted by reticulin staining), and focal CDKN2A homozy·
of diffuse glioma . Add itionally, gangliogliomas h~rbour BRAF gous/biallelic deletion all provide support for a diagnosis or
p.V600E mutation or other MAPK pathway alterations and la~k pleomorphic xanthoastrocytoma over ganglioglioma.
the IDH mutation that characterizes IDH-mutant adult-type. dif-
Cytology
fuse gliomas and the MYB or MYBL 1 fusion t~at characterizes
Not clinically relevant
angiocentric glioma and paediatric-type diffuse low-~rade
gliomas 11340,3404}. In low-grade glioneuro.nal tu~our~ with ~n
Diagnostic molecular pathology
oligodendrocyte -like glial component, the ~1fferent1al diagnosis
should include both ONT and PLNTY. Mult1nodular growth pat- For ~atients with g~nglioglioma with a typical clinical and his-
tological presentation, molecular testing may not be critical.
tern , the presence of a specific glioneuronal element, absence

<111d n -uro11a l tumours


114
Box2.1 8 Diagnostic criteria for ganglioglloma tumour should prompt consideration of this alternative diagno-
Essential: sis . /OH mutation (either /OH1 p.R132 or /DH2 p.R172), MYB or
Intra-axial low-grade glioneuronal tumour
AND
MYBL1 fusion , and PRKCA fusion are not compatible with the
diagnosis of ganglioglioma. Lastly, genome-wide DNA methyla-
tion profiling has established a distinct epigenetic signature of
~
'8
£'
Combination of neoplastic ganglion and glial cells
ganglioglloma that may aid in the classification of diagnostically
AND (for unresolved lesions)
challenging tumours (3045,460) . However, the low tumour cell
BRAFp.V600E mutation or other MAPK pathway alteration content in ganglioglioma often limits the applicability of DNA
OR methylation profiling (1004) .
Methylation profile of ganglioglioma
Desirable: Essential and desirable diagnostic criteria
Absence of !DH mutation See Box 2.18.

Staging
However, for cases with diagnostic uncertainty, the initial molec- Not clin ically relevant
ular workup should focus on BRAF p.V600E mutation testing
either by sequencing or by immunohistochemistry using the Prognosis and prediction
mutant-specific antibody VE1 (1682}. Many gangliogliomas have Prog nostic factors are difficult to define because of the com-
low tumour cell content, so the selection of areas for molecular plexity of the clinical picture, anatomical location , and molecu-
testing should be made cautiously, and sensitive sequencing lar interrelationships in retrospective cohorts over the past few
methods capable of detecting variants at low allele frequen- decades , as well as the small numbers of patients . However,
cies should be employed . In gangliogliomas negative for BRAF ganglioglloma is generally a low-grade indolent tumour with
p.V600E mutation, other BRAF alterations including mutations excellent prognosis in combined paediatric and adult cohorts
and fusions are commonly found , as well as various other (15-year overall survival rate: 83-94%) (1959,624,3560). The
genetic alterations causing activation of the MAPK signalling best prognostic indicator, including for better seizure outcome,
pathway, such as RAF1 fusion , KRAS mutation , and NF1 muta- is complete surgical resection (1959,624 ,3560}. The incidence
tion or deletion (2444). Most commonly, gangliogliomas harbour of tumour progression is quite variable depending on the series
a solitary pathogenic alteration causing activation of the MAPK (range: 16-35%) (3560 ,3577}, but no consistent correlations
pathway. However, rare gangliogliomas have been reported between histological features , imaging data, and clinical out-
with dual BRAF p.V600E mutation and CDKN2A homozygous come have been found (1959,523,666 ,2444}. In a recent large
deletion, which may potentially represent an adverse prognos- series, BRAFp.V600E mutation conferred poor outcome relative
tic factor in gangliogl ioma (2444). However, this genetic pattern to other genetic alterations (such as BRAF fusion in paediatric
is far more common in pleomorphic xanthoastrocytoma and low-grade glioma) when considered as a group {2768); how-
should prompt consideration of this alternative diagnosis (2499, ever, the prognostic value of BRAF p.V600E mutation versus
3299). A small number of ganglioglioma-like tumours centred other alterations specifically in ganglioglioma was not clearly
in midline structures of the CNS have been reported with dual defined in that study. Prospective randomized studies testing
BRAF p.V600E and H3 p.K28M (K27M) mutations (2368 ,1654, small-molecule inhibitors of RAF and MEK will be important to
2768} ; however, the exact natu re of these tumours is unknown at validate the potential clinical benefit suggested by initial case
present . A small subset of histologically defined gangliogliomas reports {2753 ,725 ,35 ,2015 ,3221 ,1041}. The rare gangliogliomas
have been reported with FGFR1 or FGFR2 mutations or fusions with dual BRAF p.V600E mutation and CDKN2A homozygous
!2444) . However, FGFR1 alterations are more characteristic of deletion may potentially have an increased risk of recurrence
other glioneuronal tumour entities (e.g. ONT, rosette-forming compared with the vast majority of gangliogliomas without this
gl ioneuronal tumour, and extraventricular neurocytoma), and accompanying CDKN2A homozygous deletion (1806,2444}.
the finding of FGFR1 alteration in a low-grade neuro~pitheli~I The ganglioglioma-like tumours centred in midline structures of
tumour should prompt consideration of these alternative enti - the CNS with co-occurring BRAF p.V600E mutations and H3
ties . Additionally, FGFR2 fusions are characteristic of PLNTY, p .K28M (K27M) mutation have been associated with poor out-
and the finding of FGFR2 fusion in a low-grade glioneuronal comes (2368 ,1654,2768}.

Gliornc1b , yl1 oneuror1al rumours , :ind neuronal tumours 115


Gang Iiocytoma Giannini C
Blumcke I
Hawkins CE
Huse JT
Rosenblum MK

Definition Etiology
Gangliocytoma is a neuroepithelial neoplasm composed of No distinct genetic susceptibility factors have been reported
irregular clusters of mostly mature neoplastic ganglion cells, for classic gangliocytoma. Dysplastic cerebellar gangliocy-
often with dysplastic features (CNS WHO grade 1). toma, which is associated with Cowden syndrome, is covered
in a separate section - see Oysplastic cerebellar gangliocytoma
ICD-0 coding (Lhermitte-Duclos disease) (p. 146).
9492/0 Gangliocytoma
Pathogenesis
ICD-11 coding Genetic data specifically addressing gangiiocytomas have not
2A00.21 & XH6KA6 Mixed neuronal-glial tumours & Ganglio- been reported. A close genetic relationship with ganglioglioma
cytoma seems possible.

Related terminology Macroscopic appearance


None Ganglion cell neoplasms are generally well-circumscribed,
grey, solid or cystic lesions.
Subtype(s)
None Histopathology
Gangliocytomas are composed of large, patently neuronal cells
Localization that lie singly or irregularly clustered in a matrix that may be indis-
Like gangliogliomas, these tumours can occur throughout the tinguishable from normal neuropil or may be more coarsely fibnl-
CNS. In a large series of seizure-associated tumours, > 80% lar and vacuolated. Binucleation and cytoplasmic ballooning or
of gangliocytomas were located in the temporal lobe {304}. vacuolization are common. Microcalcifications can be present
Dysplastic cerebellar gangliocytoma is discussed in a separate The discrete micronodular substructure of the multinodular and
section - see Oysplastic cerebellar gangliocytoma (Lhermitte- vacuolating neuronal tumour is not seen. Glial elements are
Duclos disease) (p. 146). Gangliocytoma of the pituitary is only sparsely represented and free of atypia. Because a vari-
reviewed in the volume on tumours of endocrine organs {1919). ety of CNS tumours can exhibit gangliocytomatous maturation
(including _neuro~lastomas , medulloblastomas, and embryonal
Clinical features tumours with mult1layered rosettes), the presence of proliferative
Gangliocytomas have th e same clinical features as ganglioglio- small cell components or neuroblasts must be excluded. Mitotic
mas. activity is absent and Ki-67 labelling is negligible.

Epidemiology . lmmunophenotype
Gangliocytomas are rare tumours that predominantly affect c~1l­ Neoplastic ganglion cells exhibit variable immunoreactivity
dren . The relative incidence reported in epilepsy surgery series for synaptophysin , chromogranin , NFPs, and MAP2. NeuN
ranges from < 1% to 3 .2% {3177,304) .

A -
Flg. 2.106 Gangl1ocytoma. Right frontal lobe. Rela~i~ely c!rcumscribed lesion involving cortex and superficial white matter. A T2-hyperintense on MRI. 8 Tl-isointense. C C~
trast enhancement 1s present after gadolinium administration.

116 Clio111d'::. (Jil r 11 l-: Ufl)f 1dl IUllilJIJf~ n11d flt !l ll<Jilli l ll/f[l()tl!S
i
Fig.2.107 Gangliocytoma. A Clusters of large ganglion cells In a matrix indistinguishable from normal neuropil. Microcalcifications are present. B Large ganglion cells with
cytoplasmic ballooning; one is binucleated.

expression may be diminished or lost. GFAP labelling is the underlying white matter. Balloon cells are highlighted by
restricted to reactive astroglia. vimentin and aB-crystallin . Although FCD type 2b is associ-
ated with a thickening of the cortex and blurring of the grey
Grading matter-white matter junction macroscopically, it lacks the
Gangliocytoma corresponds histologically to CNS WHO mass-like circumscribed appearance of gangliocytoma. In the
grade 1. suprasellar region, gangliocytoma should be distinguished
from sporadic hypothalamic hamartoma (also referred to as
Differential diagnosis "hamartoblastoma"), a congenital mass-like lesion that is
Among non-neoplastic neuronal lesions, focal cortical dys- characteristically located in the floor of the hypothalamus,
plasia (FCD) , primarily type 2, should be considered in the involving the tuber cinereum and the mammillary bodies pos-
differential diagnosis of gangliocytoma. FCD type 2a is char- teriorly and extending inferiorly into the interpeduncular cis-
acterized by the disruption of cortical lamination with dysmor- tern . Hypothalamic hamartoma consists of collections of small
phic neurons that have enlarged cell bodies and prominent to medium-sized neurons typically arranged in nodules and
accumulation of Niss! substance and that lack anatomical ori- separated by hypocellular neuropil resembling normal CNS
entation . The dysmorphic neurons in FCD show cytoplasmic neuropil . The glial/neuronal composition varies widely, with
accumulation of neurofilaments (SMl32) and , unlike in gan- some lesions comprising predominantly neurons, and others
gliocytoma, typically retain NeuN expression. In FCD type 2b, predominantly glial cells (both astrocytes and oligodendro-
in addition to dysmorphic neurons, balloon cells with ample cytes) {634) . Hypothalamic hamartoma is the pathognomonic
eosinophilic and glassy cytoplasm, at times multinucleated , manifestation of Pallister-Hall syndrome, a rare autosomal
are present, particularly in the deep layers of the cortex and dominant malformative disorder that includes (in addition to

A
flg.2.108 Gangliocytoma. A Neoplastic ganglion cells are positive for chromogranin. B Neurofilament, like synaptophysin. is typically immunopositive in ganglion cells as well
as (diffusely) in background neuropil.

Cil1or11C1s , ylloncuro11ci1 tu1111JLrr s dl1ti neuronal tu 11io ur s 117


hypothalarrnc harnartoma) polydactyly and imper orate anus Box2.19 D1agnost1c criteria for gangltocytoma
as well as abnormalities of e pituitary. larynx. and genitouri-
nary tract {8051 GL/3 germltne muta ions have been associ- A tumefaclJve leSJOn with presence of irregular groups of large. mature ganglioo
ated with Pallister-Hall syndrome {7331 Mutations 1 GL/3 and
AND
a variety of other genes often related to the SHH pathway have
been reported 1n a subset of sporadic hypothalamic hamarto-
ng normal neuropl. somellm s more coarsely fibnllar or vacuolated
mas 11308). Mixed gangltocytoma-adenoma also kno n as
"pituitary adenoma with gangllocyt1c d1fferent1at1on· or -pitui- typical and bin aled gan hon c lls
tary adenoma-neuronal chonstoma·. typically 1n Ives the eytt)J)la:smic oa11<:ioo1111 or vacuolization
sella turc1ca and suprasellar region without connection 1th
the hypothalamus In add1t1on to 1s ganghocyt1c componen .
mixed gangliocytoma-adenoma 1s charac enzed b t e pres- nd acuolatmg n uronal tumour. frequently harbour alterations
ence of an adenomatous component. mos of en a so inc s 1tu nts of the RAS/MAPK signalling pathway.
troph adenoma man ifest1ng as acromegal or a m1 ed u ur
producing prolactm and growth ho one I 926) I and d sir bl diagnostic criteria

Cytology
A characteristic profile 1n smear/squas
been described II rele ant

Diagnostic molecular pathology and pr d c on


To date. no signature mo ecutar abnorma~1t1es ~nc:nacv·1onldS r benign tumours with a favou rable out-
ascribed to ganghocytoma specif ca•t a •fic pro nost1c or predictive factors have not been
1cally related ent1 1es mcludt g ga g og

8 s ~ rr•::L g -
cri:::~'
0 '"' - 1 • '""'' u
·i c;j _,I
' _;rS and neu'cna1 .u!TOUf S
Oesmoplastic infantile gang lioglioma / Figarella-Branger D
Gessi M
desmoplastic infantile astrocytoma Reuss DE
Solomon DA
Varlet P

Definition Clinical features


Desmoplastic infantile ganglioglioma (DIG) and desmoplas- The most frequent clinical signs are increased head circumfer-
tic infantile astrocytoma (DIA) are benign glioneuronal or glial ence, bulg ing of the fontanelles , lethargy, and the sunset sign
tumours . occurring predominantly in the cerebral hemispheres (3136 ,3287).
of infants, that are driven by MAPK pathway activation and com-
posed of a mixed astrocytic and neuronal component (DIG) or Imaging
an astrocytic component only (DIA) embedded in an extensive The radiological aspects of DIG and DIA are similar: both appear
desmoplastic stroma, often containing foci of undifferentiated as a large, superficially located , solid and cystic tumour that is
emb ryonal-like tumour cells (CNS WHO grade 1). enhancing after contrast administration. On CT, the solid part of the
DIG/DIA is isodense or slightly hyperdense, with strong enhance-
ICD-0 coding ment after iodine contrast administration; the cystic component
9412/1 Desmoplastic infantile ganglioglioma is frequently hypodense or isodense [170 ,271). On MRI , the cys-
9412/1 Desmoplastic infantile astrocytoma tic part is unilocular or multicystic, hypointense on T1-weighted
sequences, and hyperintense on T2-weighted sequences ,
ICD-11 coding whereas the solid component is frequently duraJ based and
2A00.21 & XH6TQ7 Mixed neuronal -glial tumours & Desmo- appears hypointense on both T1- and T2-weighted sequences,
plastic infantil e ganglioglioma with contrast enhancement after gadolinium injection (271).
2A00 .21 & XH7M44 Mixed neuronal-glial tumours & Desmo-
plastic infantile astrocytoma Epidemiology
DIG/DIAs are rare primary brain tumours, but their true inci-
Related terminology dence is not well defined, because they are usually included in
Not recommended: superficial cerebral astrocytoma attached the large group of glioneuronal tumours. DIG/DIAs account for
to the dura. 0.4% of brain tumours, with an M:F ratio of 1.8:1 [677,3288} . They
represent 1.25% of intracranial tumours in children [3136) and
Subtype(s) 1.3-15.8% of infantile brain tumours 13622,1542). The vast major-
None ity of cases occur before the age of 24 months. Rare non-infantile
cases have been reported , but these were in the pre-genetic era
Localization and might represent misclassified tumour types [2453).
DIG/ DIAs typically arise in the cerebral hemispheres, involving
the superficial cortex and leptomen in ges, often with attachment Etiology
to the dura. Rare cases have been reported in other locations No risk factors have been reported for DIG/DIA, and no inher-
(spinal , posterior fossa, intraventricular, suprasellar), but exten- ited genetic susceptibility or association with known tumour
sive genetic analysis of such cases has not been performed predisposition syndromes has been established . DIG/ DIA is
12221 ,2210 ,3364) . genetically driven by somatic alterations causing activation of
the MAPK signalling pathway, most commonly via mutation or

Fig. 2.109 Desmoplastic Infantile ganglioglioma in the right hemisphere of an .18-month-old girl. Axial (A) and cor?.nal (8) T2-welghted MRI shows multiple cystic components
with cerebrospinal fluid- like signal, and a peripheral solid component that 1s 1so1ntense lo grey matter. C Suscept1 b1l1ty- we1gh~ed MRI shows no blooming effect (pseudoenlarge-
ment of the lesion). 0 Diffusion-weighted MAI demonstrates a solid hyperintense portion, possibly due to desmoplastlc reaction E Contrast-enhanced Tl -weighted MAI shows
intense homogeneous enhancement of the solid component and no enhancement of the cyst walls. F Colour cerebral blood volume map (dynamic susceptibility contrast perfu·
s1on) shows relative hyperperfusion of the solid enhancing component compared with the contralateral while matter.

Gl1on1a::. . £Jl1or1eu1 ondl umotir.,, · 11 ci neuiun...11 [uniou 1s 119


Fig. 2.110 Desmoplastic infantile ganglioglioma. A The tumour is superficial, with sharp demarcation from the cortex. B The tumour shows a marked desmoplastic component
evidenced by reticulin staining, whereas the adjacent cortex is negative.

fusion involving the BRAFor RAF1 genes {1679,1163,525,3364, component consists of a mixture of ·fibroblast-like, spindle-
295}. Most DIG/DIAs demonstrate a flat copy-number profile, shaped cells intermixed with a collagen matrix. Reticulln-rich
and recurrent cytogenetic alterations have not been identified basal lamina classically surrounds almost every cell (3136,
(1742,1077,3364}. 3288}. In DIA, the neuroepithelial component comprises an
astrocytic population only; in DIG, a neoplastic neuronal com-
Pathogenesis ponent with ganglionic differentiation is also observed {32881.
Although the exact cell of origin remains uncertain, DIG/DIA has The neoplastic astrocytes are arranged in fascicles or dem-
been postulated to arise from a population of specialized sub- onstrate storiform or whorled patterns. In addition , DIG/DIAs
pial astrocytes in the developing brain {1942,164}. However, the often contain foci of primitive, embryonal-like tumour cells. This
presence of undifferentiated foci of embryonal-like tumour cells immature component, lacking desmoplasia, may predominate
in most cases raises the possibility of an early progenitor cell in some areas.
that gives rise to a tumour with varying degrees of progressive There is a sharp demarcation between the cortical surface
maturation . DIG/DIA is genetically driven by the activation of the and the desmoplastic tumour, although Virchow-Robln spaces
MAPK signalling pathway {1679,1163 ,525 ,3364,295}. are often filled with tumour cells . Calcifications are common.
but perivascular mononuclear inflammatory infiltrates and xan-
Macroscopic appearance thomatous cells are usually absent. Necrosis is uncommon
DIG/DIAs typical ly contain large uniloculated or multiloculated and is typically restricted to the foci of primitive, embryonal-like
cysts filled with clear and colourless or xanthochromic fluid . The cells . Glomeruloid microvascular proliferation is usually absent
solid superficial portion is primarily extracerebral , involving the {271 ]. In most cases, mitotic figures are limited to the foci of
leptomeninges and superficial cortex. It Is commonly attached embryonal-like tumour cells and do not exceed 0.8 mitose
to the dura, is firm or rubbery in consistency, and is grey or white mm 2 (equating to < 2 mitoses/10 HPF of 0.55 mm in diameter
in colour. There is typically no gross evidence of haemorrhage and 0.24 mm 2 in area) in the desmoplastic component.
or necrosis (3136,3288}.
lmmunohistochemistry
Histopathology . The glial component strongly expresses GFAP, whereas
DIG/DIAs are biphasic tumours composed of a prominent des- neuronal markers (synaptophysin, neurofilament, NeuN) are
moplastic leptomeningeal stroma and a variable proportion of observed in neoplastic ganglion cells as well as in cells lack-
neuroeplthelial component. The desmoplastic leptomeningeal ing obvious neuronal differentiation [2431 ,525}. The Ki-67

120 Gl101nas , gl1un•.:::LJJl)rla l turnour~ . anu 11euror1a l (Ul l lOl l fS


Flg.2.112 Desmoplastic infantile ganglioglloma. A Neoplastic cells are arranged in streams in the desmoplastlc component. B Some cells express synaptophysin. C A few
cells are GFAP-positive. D The Ki-67 labelllng index is very low in desmoplastic areas.

proliferation index within the desmoplastic component ranges


from < 0.5% to 5% , with the majority of reported values being
< 2% 11742,525}. However, the Ki-67 index can be significantly
elevated (as high as 20%) in the foci of embryonal-like tumour
cells {1077}. lmmunohistochemistry using the VE1 clone is use-
ful for detecting DIG/DIAs that harbour BRAF p.V600E muta-
tion , but this mutation-specific antibody does not recognize
the p.V600D mutation or any of the other BRAF mutations or
rearrangements found in DIG/DIAs without p.V600E mutation
(525 ,1679,1163}.

Differential diagnosis
The major differential diagnoses for DIG/DIA are ganglloglioma,
pleomorphic xanthoastrocytoma, and the newly defined tumour
type infant-type hemispheric glioma. Like DIG/DIA, gangli-
oglioma and pleomorphic xanthoastrocytoma are typically Flg.2.113 Desmoplastic Infantile astrocytoma. Dittuse BRAF p.V600E immunoex-
presslon.
solid and cystic tumours with frequent BRAF mutations, but
ganglioglioma and pleomorphic xanthoastrocytoma are typi-
cally much smaller and occur in older children, in contrast to the Diagnostic molecular pathology
very large size and the infantile onset of DIG/DIA. Infant-type DIG/DIAs are IDH - and histone H3-wildtype tumours character-
hemispheric glioma shares the characteristics of infantile onset ized by genetic alterations causing activation of the MAPK sig-
and hemispheric location with DIG/DIA , but infant-type hemi- nalling pathway, most commonly via mutation or fusion involving
spheric gliomas usually lack desmoplasia and demonstrate a~ BRAF or RAF1 {1679,1163,525 ,3364,295 ,27681 . BRAF muta-
infiltrative growth pattern (unlike DIG/DIAs), and they are geneti- tions can include p.V600E and other variants at the same codon ,
cally characterized by fusions involving receptor tyrosine kinase such as p.V600D, in addition to variants at other locations or
genes (ALK, ROS1, MET, NTRK1 , NTRK2, and/or NTRK3) (460, fusions involving partners other than KIAA 1549. These BRAF or
463 ,33641 . RAFt. mutatio~s or fusions are typically present as the sole path -
ogenic alteration identified, and DIG/DIAs lack the COKN2A
Cytology and(or CDKN2B homozygous deletion that commonly accom -
l\Jot clinica lly relevant panies BRAF alterations in pleomorphic xanthoastrocytoma

Uliomas , glioneuror1al tumours , dlld ri urun~il tumouis 121


{3364) . Although individual cases of histologically defined DI G/ Box 2.20 Essential and desirable diagnostic criteria for desmoplastic infantileastrocy.
DIAs harbouring ALK or NTRK fusions have been reported toma (DIA) and desmoplastic infantile ganglioglioma (DIG)
13364,295}, most hemispheric gliomas occurring in infants that Essential:
harbour ALK, ROS1, MET, or NTRK fusions have been shown Biphasic morphology with a dominant desmoplastlc leptomeningeal component
to epigenetically cluster with infant-type hemispheric glioma admixed with a neuroepithelial component containing astrocytic cells only (DIA) or
(1179,603) . Whether there are true DIG/DIAs that also harbour containing astrocytes and neuronal cells (DIG)
fusions in receptor tyrosine kinase genes (ALK, ROS1 , M ET, AND (for unresolved lesions)
NTRK1, NTRK2, NTRK3, or FGFR1) remains to be established. Methylation profile of DIG/DIA
DNA methylation profiling has revealed that DI G/DIAs have an
OR
epigenetic signature distinct from all other primary CNS tumour
BRAF or RAFI mutation or fusion, occurring in the absence of homozygous
entities characterized to date, including pleomorphic xanthoas-
deletion of CDKN2A and/or CDKN2B
trocytoma and the aforementioned infant-type hemispheric
glioma {460,463,3364) . Desirable:
Tumour with a cystic component and a solid portion, with leptomenlngeal involve-
Essential and desirable diagnostic criteria ment, usually attached to the dura
See Box 2.20. Infantile onset (typically at < 24 months)

Staging consideration , especially for patients with progression of the


Not clinically relevant residual tumour and/or leptomen ingeal dissemination (3364,
3288}. Small-molecule tyrosine kinase inhibitors targeting
Prognosis and prediction the MAPK signalling pathway are also a therapeutic option in
The prognosis of DIG/DIA is excellent when total surg ical cases demonstrating BRAF mutations {294}. Although some
removal is achieved , with no relapse in most cases with fol low- DIG /DIAs demonstrate foci of frank anaplasia (e.g. high mitotic
up ranging from 5 to 15 years {31 36 ,3287,271 ). Long-term out- count, palisad ing necrosis), there is no clear relationship
come is better with hemispheric location than with suprasellar between anaplasia and clinical outcomes (711 ,3224,24921
location 12221 J, and leptomeningeal dissem ination and/or mul- Rare cases of recurrence accompanied by malignant transfor-
tifocal disease, although rare, are most frequently associated mation of DIG/DIA occurring as late as 8-10 years after the
with suprasellar location (2210]. In cases of subtotal resection first resection have been reported , accompanied by acquired
or biopsy only, careful follow-up is mandatory to mon itor for TP53, ATRX, or BCORL1 mutations in individual cases (1920,
regrowth of the residual tumour, which typ ically remains sta- 2492,3364,2554) .
ble or grows slowly over years {271, 295), but regression has Despite the malignant transformation observed histologically,
also been documented {311 6). In cases of subtotal resection , some patients remained al ive for 2 3 years after the second sur-
adjuvant chemotherapy and/or radiotherapy is a therapeutic gery (1920,3364) .

122 G li omas , y l1orieuronal tun 1r;u r s . w 1d neuronal lumours


Dysembryoplastic neuroepithelial tumour Pietsch T
Ellison OW
Hirose T
Jacques TS
Schuller U
Varlet P

Definition
Dysembryoplasti~ neuroepithelial tumour (ONT) is a glioneu-
ronal neoplasm 1n the cerebral cortex of children or young
adults, characterized by the occurrence of a pathognomonic
glioneuronal element that may be associated with glial nodules
and activating mutations of FGFR1 (CNS WHO grade 1).

ICD-0 coding
9413/0 Oysembryoplastic neuroepithelial tumour

ICD-11 coding
2A00 .21 & XHOH76 Mixed neuronal-gllal tumours & Oysem-
bryoplastic neuroepithelial tumour

Related terminology
None
Flg.2.114 Dysembryoplastic neuroepithelial tumour. A Coronal T2-weighted MRI of
a temporal tumour. B Coronal FLAIR MRI of a temporal tumour.
Subtype(s)
None
Epidemiology
Localization Incidence
ONTs can be located in any part of the cerebral cortex , but they The estimated incidence of ONT is 0.03 cases per 100 000 per-
show a predilection for the temporal lobe (67.3% of cases, pref- son-years. An analysis of SEER data from 2004-2013 found that
erentially involving mesial structures) {449,3180) and the frontal the incidence of ONT was lower in the Black, American Indian /
lobe (16 .3% of cases); the remaining cases (16.4%) are located Alaskan Native , and Asian or Pacific Islander populations than
in other regions 13180). in White people {2242). In a large epilepsy surgery series, ONTs
accounted for 5.9% of the cases {304}.
Clinical features
Patients with ONTs typically present with drug-resistant focal Age and sex distribution
epilepsy with an onset in childhood, adolescence, or early In about 90% of patients with ONT, the first seizure occurs
adulthood . before the age of 20 years, with reported ages at seizure onset
ranging from 1 week to 30 years (2628 ,2242} . There is a slight
Imaging predominance of ONT in male patients (accounting for approxi-
DNTs usually encompass the thickness of the normal cortex. mately 55% of cases in a large series) {304,2680) .
The main distinctive characteristics for differentiating ONT from
diffuse gliomas are a Jobulated architecture, sharply de'fined Etiology
margin, and absence of mass effect with no significant peritu- Most ONTs are sporadic and caused by FGFR1 alterations
moural oedema. ONTs appear as cystic or multicystlc lesions, {2584, 2680, 934, 3597, 2040, 2767, 1828, 3075 l. although accu-
hypointense or nearly isointense to grey matter on T1-weighted mulating data suggest that they may also occur in the setting
MRI , and hyperintense on both T2-weighted and FLAIR MRI of RASopathies (a group of neurodevelopment diseases with
{449) . Marked high signal intensity within the mass (soap- germline mutations in the RAS signalling pathway), such as neu-
bubble appearance) with intracystic septa, thin FLAIR hyper- rofibromatosis type 1 {1853,198) or Noonan syndrome [2926,
intensity surrounding the tumour (rim sign) {2403). triangular 2059). or in families with an FGFR1 germllne mutation {26801.
cortical based , and remodelling of the adjacent inner table of
bone (indicating a slow-growing lesion) are common findings Pathogenesis
11423,74). Calcifications and haemorrhage are rare 1663). About Recent comprehensive genomic analyses revealed FGFR 1altera-
one third of complex ONTs exhibit enhancement after gadolin - tions in approximately 40-80% of DNTs, with BRAFp.V600E muta-
ium administration, with a nodular pattern , peripheral rim-like tion reported in as many as 50% of DNTs in some studies 12584,
enhancement, or both {663 ,74) . 2680,934,3597,523,2550,2040,2767,1828,3075). POGFRA and
NF1 mutations were also reported in a few cases 12584,30751.
These mutations are typically mutually exclusive [934 ,1528). The
FGFR genetic alterations induce autophosphorylation of FGFR1

Gl1omas , g lioneuronal tumours , and neuronal rumours 123


Histopathology .
The histopathological hallmarks are the mu lt1nod ular intracriri
cal growth pattern and the pathognomoni.c speci fic glioneurl)r,1
element, i.e. the presence of columns orien ted perpen dicular;
to the cortical surface , formed by bundles of axons lined r1~
small oligodendroglia-like cel ls. Between these colum ns, ne ,.
rans with normal cytology appear to fl oat in a muco1d matr,,
(floating neurons) (685). Oyspl astic gang lion ic cells are absen:
lntrano dular glial com ponents can be hi gh ly heterogeneou)
with oligodendrogl ia-like, neu ronal , or piloid or stellate astrocytu::
cell s. The ol igodendroglia-like component generally predorn•
nates (299,3045) . Mitoses are rare . High cellularity, necr 0s,3
sign ificant perivascu lar lymphoid infiltrates, and eosinophilic
granular bod ies are absent. The microvascular network is alsc
heterogeneous, varying from meagre to extens ive , and it may
include glomerular formations as seen in pilocytic astrocy c-
mas . Focal cortical dysplasia type 3b has been described ,n
association with ONTs (683 ,3180) but remains controversial. ,1
may be diagnosed only if areas of focal cortical dysplasia type ·
without tumour cell infiltration can be identified (305}.

Recognized histopathologica/ patterns


Several histological forms of ONT, without clinical or therapeutic
implications , have been described (687).
The simple form consists of the unique glioneuronal element
The complex form consists of the specific glioneuronal ele-
ment in combination with glial nodules, and it resembles other
gliomas such as pilocytic astrocytomas or oligodendrogliomas
{569,3180,299,685). Within the glial components , calcified ves-
sels are common and can cause haemorrhage {3178).
The diffuse/nonspecific form, which lacks the specific gho-
neuronal element {687,3180}, remains controversial; numerous
terms have been used for it in the past, including "diffuse 011-
godendroglial tumour" (2584). "ONT-like tumour" (2680), "glio-
neuronal tumour NOS" (3045). "paediatric oligodendroglioma
(2703), and "diffuse glioneuronal tumour " (296,3180,74). The
variability of its histomorphological appearance is related to a
low interobserver diagnostic concordance (299). Recent data
have shown that clear phenotype-genotype correlations are
difficult to establish, even for experienced neuropatholog1sts
using advanced molecular techniques (3045).

lmmunohistochemistry
The small oligodendroglia-like cells express glial markers includ-
ing S100, the glial transcription factor OLIG2, and POGFRA
but not GFAP. MAP2 expression is typically faint in these cells
the ~trong perinuclear expression found in oligodendrogl10-
mas 1s not detectable (303). The floating normal neurons can
be depicted by immunostaining for NeuN , but they are usuall
negative for chromogranin A. BRAF p.V600E-mutant protein
and then upregulate the MAPK and P13K pathways {2680,2767, and C034 have been described with variable incidences 1n
1960). FGFR1 activity is associated with Inhibition of oligodendro- ONT {523 ,3180 ,299). but they seem more characteristic of gan·
glial precursor differentiation (3611 ). gliogliomas (3045 ,299) . ONT cells do not label with antibodies
against IOH1 p.R132H {461 ).
Macroscopic appearance
On the cut su rface , ONTs are usually poorly defined and located Differential diagnosis
mainly in the expanded grey matter and subcortical white ~at­ Sampling artefacts may make the diagnosis challeng ing . Clus-
ter. ONTs vary 1n size from a few millimetres to several centime - ters of abnormal neurons not otherwise explicable by and-
tres 124 11 l and contain mucoid substances, solid areas , and tomical locali~ ~tion may be focal and detectable only in sonitl
smal l cysts in va rying proporti ons (685,1338) . cases . In add1t1on , the architectural heterogeneity of ONT (wh1d1
..• ./ . •
• •


• •
••
.
·' • •
•. ' .
• •
. ..-. . : •..
•• •

•• . ••

# ..

generates inherent sampl ing bias) and the semiliquid consist- or mitotic activity, set against a variably mucinous or fibrillary
ency of the specific glioneuronal element, which can be lost matrix. However, in more complex DNTs , there may be overlap
during the neurosurgical procedure, pose further challenges . with other glial tumours .
ONTs do not contain dysplastic ganglion cells such as those
described in gangliogliomas (i .e. binucleated neurons and large Diagnostic molecular pathology
neuron clusters not otherwise explicable by anatomical region) Among FGFR1 alterations , intragenic duplication (internal tan-
(299] . Ganglioglioma should be suspected when the tumour dem duplication [ITD]) of the tyrosine kinase domain (TKO) of
shows perivascular lymphocytic infiltration, a desmoplastic FGFR1 is the most prevalent aberration (accounting for -40-
component, eosinophilic granular bodies, a large cystic compo- 60% of cases) (2584,2680,934,2040}. followed by missense
nent, or a prominent component of C034-positive satellite ce lls . mutations at mutation hotspots in FGFR1 (2680,934,2584,
The strong perinuclear expression of MAP2 found in diffuse oli- 3075] . FGFR1 mutations were identified in both famil ial and spo-
godendrogliomas is not detectable in the oligodendroglia-like radic cases, with double or multiple mutations often present on
cells of ONT {303}. the same allele (in cis) (2680,2584). FGFR1 ::TACC1 fusion and
Myxoid glioneuronal tumour, formerly known as ONT of the complete duplication of FGFR1 have also been reported (2680,
septum pellucidum (177,1072], has been separated from con- 2584). FGFR1 alterations are characteristic of ONT (although
ventional ONT because it harbours a specific molecular altera- not specific for it), and they are considered to be the main
tion (PDGFRA p.K385 mutation) not found in conventional ONT, molecular driver of this tumour {2680,934,2040). In particular,
and it has the potential for ventricular dissemination {1954,569] . TKO duplication is known to be relatively specific to ONT (934,
The spectrum of low-grade epilepsy-associated brain 2040,2584).
tumours is widening rapidly with new descriptions of various BRAF p.V600E mutation has been reported in as many as
rare histomolecular tumour types; therefore, the differential 50% of DNTs in some studies. The wide range in incidence
diagnosis requires a combined histological , neuroradiological, of the various alterations probably stems from differences in
and molecular diagnostic approach. Multinodular and vacu- the morphological criteria used to make the diagnosis of ONT
olating neuronal tumours have a multinodular architecture but a across the various studies; of note, several studies have shown
predominant neuronal (i .e. gangliocytoma-like) component with marked variation in the reported frequencies of DNTs, as well
vacuolating cells, mainly in the subcortical white matter {2443}. as marked interobserver variability in making this diagnosis
MAP2K1 mutation or (more rarely) BRAF mutations are found (299,296). In contrast to FGFR1 alterations, several studies
instead of the FGFR1 alterations typical of ONT (2443,1383} . failed to identify BRAF mutations in ONTs containing the spe-
Polymorphous low-grade neuroepithelial tumour of the young. is cific glioneuronal element (2680,2040}. Therefore, alternative
a morphologically variable entity and is mainly ollgodendrogl1a- diagnoses, including ganglioglioma or MAPK pathway-altered
like similar to ONT but with a more infiltrative growth pattern, diffuse low-grade glioma, should be carefully considered in the
cal~ifications and intense C034 immunopositivity of tumour presence of a BRAF p.V600E mutation .
cells . They h~rbour either BRAF p.V600E ~utatlons or fusion In support of this, DNA methylation or transcriptional pro-
events involving FGFR2 or FGFR3 {1384). Diffuse astrocytoma, filing identifies different molecular groups of epilepsy-asso-
MYB- or MYBL 1-altered , is a more monomorphic tumour, where ciated tumours (3046,299]. and although these groups cor-
the regular astrocytic cells are scattered in a fine bub?IY neu- relate only partially with histological patterns, they separate
rop il. Unlike in angiocentric gliomas with MYB:: QKI fusion tran- epilepsy-associated tumours into those with FGFR1 muta-
scripts , angiocentric patterns are absent or focal (3404) . tions and those with BRAF mutations, with the former being
enriched for morphologically defined DNTs. These data sug-
Cytology . . gest that, according to molecular criteria, DNTs have a distinct
Smear (squash) cytological findings typical!~ reveal a spr~ad methylation and transcriptional profile , and most carry FGFR1
of uniiorm rounded cells, without substantial pleomorph1sm mutations.

Gliomas, glioneuronal tumours, and neuronal tumours 125


Box2.21 Diagnostic criteria for dysembryoplastic neuroepithelial tumour Essential and desirable diagnostic criteria
Essential: See Box 2.21 .
Cortical glioneuronal tumour
AND Staging
Presence of the specific glioneuronal element Not clinically relevant
AND (for unresolved lesions)
Prognosis and prediction
FGFR1 gene alteration (FGFR1 internal tandem dupJicabon [ITD], fusion, DNTs are considered benign lesions {3016}. lschaemic or
missense mutation}
haemorrhagic changes may occur with or without an increase
OR
in size of the lesion or peritumoural oedema 1686,91 t,1469
Methylation profile of dysembryoplastlc neuroeplthelial tumour 2341 ,3016}. Risk factors for the development of recurrent sei-
Desirable: zures include a longer preoperative history of seizures /140
Early-onset focal epilepsy 12891. the presence of residual tumour /2273}, and the pres-
ence of cortical dysplasia adjacent to ONT 12789).
MRI may reveal tumour recurrence, but histopathology often
remains benign . Malignant transformation has been docu.
Regarding copy-number aberrations, gains at chromo- mented in rare cases with 12756,2627} or without {2756,2627,
somes 5-7 and loss of chromosome 22 have been reported in 3180,522,1280,2039} radiation and/or chemotherapy.
a few cases 12551 ,26801.

126 Gltomas, gltoneuronal tumours. and neuronal tumours


.
Diffuse glioneuronal tumour Sahm F
Haberler C
with oligodendroglioma-like features Pfister SM
Schuller U
and nuclear clusters

Definition Localization
Diffuse glioneuronal tumour with ol igodendroglioma-like fea- All cases reported so far were supratentorial. Of 21 cases with
tures and nuclear clusters (OGONC) is a provisional tumour data available, 11 were located in the temporal lobe , 4 in the
type proposed as a neuroepithel ial tumour characterized by parieto-occipital region , 5 in the frontal lobe , and 1 in a lateral
variably differentiated cells frequently showing perinuclear ventricle (737,2506} .
haloes , scattered multinucleated cells , and nuclear clusters,
with a distinct ONA methylation profile and frequent monosomy Clinical features
of chromosome 14. There are currently no known tumour-specific symptoms .
Patients present with symptoms characteristic for the tumour
ICD-0 cod ing location within the brain .
None
Epidemiology
ICD-11 cod ing Th e majority of DGONC cases occur in paediatric patients ,
2AOO 21 Mixed neuronal-glial tumours with a median age of 9 years in the 23 cases tor which the age
of initial diag nosis is known . However, the age range is wide
Related terminology (1 patient was aged 75 years) . There is an equal sex distribution
None (13 male pati ents and 11 fem ale patients reported) (737,2506).

Subtype(s) Etiology
None Unknow n

..
Flg.2.117 Diffuse glioneuronal tumour with oligodendroglioma-like features and nuclear clusters. A Monomorphic clear cell morphology, resembling oligodendroglio-
ma. B Clear cell histology can resemble oligodendroglioma. C Several nuclear clusters (some 1nd1cated by arrows). D Tumour with clear cell morphology and several ring- or
C-shaped nuclear clusters E Tumour with oligodendroglioma·like perinuclear haloes.

Gl10111as , g l1uneurnr1tll tu1riour~. and neuronal tumours 127


ONA methylatlon cluaterlng (n • 191)
type or subtype of a CNS embryonal tumour. Larger series a1e
needed for a definite classification {2506) .
.....
OBM_POOPRA Macroscopic appearance
O_IOH '
CJIM.,.K27 ~ Not reported
I

OLONT, MC-2 Histopathology


. . ~ ' ·. DGONCs were described by Capper et al. in 2018, identified
C: NC:YT
l'~CORT
by the detection of a global DNA methylation profile that is dis-
OLONT, MC-1
tinct from that of all brain tumour entities previously analysed
1 0 - LCJG_MYI , ; .,...
,. CNS NB-FOXR2
(460) . Subsequently, histological features were investigated in a
supervised fashion on 13 samples of this cluster, for which his-
ONlfT
tology slides were available. The histological characterization in
C:NI HO~IT .aCOR
..... . " ' . l!VNCYT the literature may therefore not cover the entire spectrum [7371
Another study reported 4 samples from 3 additional patients
-- -- .~ . . ...
CHI H T.jilN1
;' . .. (2506) .
' The tumours show a variation in differentiation and cellularity
·" . I
I OOONC
..
• •• .) : ranging from moderate to high in some cases. They are com·
CNI l!FT .CIC \ \ ..··, I

posed of small to medium-sized cells with scant cytoplasm, and


I I I l I
they commonly show perinuclear haloes like in oligodendro·
- 10 -5 0 5 10 glioma. In addition to small round nuclei , larger irregular nuclei
t1n11 are present. the latter showing marked nuclear pleomorphism
Flg. 2.11 Diffuse glloneuronal tumour with oligodendroglioma·like features and nu· in some cases. A characteristic feature is the presence of scat-
cl r clusters (OGONC). DNA methylation profiling. DGONC samples (n = 31) were tered multinucleated cells (pennies on a plate) and nuclear clus·
compa1 d with 160 well-characterized reference samples representing CNS tumours ters composed of large pleomorphic nuclei. Mitotic frequency is
ol d fin d histological and/or molecular subgroups, confirming the distinct nature of variable, ranging from single mitotic figures in well-differentiated
th DGONC nt1ly. Reference cohorts: central neurocytoma (CNCYT); CNS high-grade tumours to brisk mitotic activity in undifferentiated tumours.
n uro pith II I tumour with BCOR alteration (CNS HGNET-BCOR); CNS Ewing sar-
Microcalcifications and larger confluent calcifications are com·
com I mlly tumour with CIC alteration (CNS EFT-CIC); CNS high-grade neuroepithe-
11 I tumour with MNt Iler t1on (CNS HGNET·MN1 ); CNS neuroblastoma with FOXR2 manly encountered . The tumours infiltrate diffusely into the brain.
ctlv hon (CNS NB-FOXR2): diffuse leptomeningeal glloneuronal tumour, methylation
cl s 1 nd cl ss 2 (DLGNT. MC-1 and MC-2): dysembryoplastic neuroeplthelial tu- lmmunohistochemistry
mour (DNET); t tncular neurocytoma (EVNCYT); H3 K27M-mutant glioblas- The tumour cells are diffusely OLIG2-positive in most cases,
tom (GSM_K27); H IOH-wildtype glioblastoma RTK1 group (GBM_PDGFRA); low- and GFAP-negative. Synaptophysin reveals a neuropil back·
gr d gllom . MYB· or M BL1 ltered (LGG_MYB); !DH-mutant, 1pJ19q-codeleted ground. NeuN and MAP2 are expressed in a small proportion
ollgod ndroghom (Q_IDH); hemispheric pilocytic astrocytoma (PA_CORT). of cells {737) .

Cytology
Insufficient data available

Diagnostic molecular pathology .


DGONCs sho~ a d.istinct DNA methylation profile. In t-distri.b·
uted stochastic neighbour embedding (t-SNE) analysis with
t~e reference samples of Capper et al. (460). DGONCs torrn a
distinct cluster close to FOXR2-activated CNS neuroblastorna

dfld r' turor al tJno JfS


Monosomy of chromosome 14 has been reported in 30 of Box2.22 Diagnostic criteria for diffuse glioneuronal tumour with oligodendroglioma-
31 investigated cases . No other specific recurrent alteration is like features and nuclear clusters (DGONC)
known 1737.2506) .
Essential:
Methylation profile of DGONC
Essential and desirable diagnostic criteria
See Box 2.22. AND
Nuclear clusters of small to medium-sized cells exhibiting oligodendroglioma-like
Staging morphology
Not clinically relevant AND
Strong expression of both OLIG2 and synaptophysin
Prognosis and prediction AND
Outcome data are available for 26 patients. The 5-year pro- Absence of widespread GFAP expression
gression-free survival rate was 81%, and the 5-year overall
Desirable:
survival rate was 89% {737,2506). Treatment data are available
Monosomy of chromosome 14
for 3 patients , all of whom received radiochemotherapy {2506).
The patients' initial diagnoses varied widely, from low-grade to Caveat: DNA methylation profiling is so far the only method to clearly identify_DGONC.
high-grade, so some may have received aggressive therapy. It If DNA methylation profiling is not available, morphological features may provide an
approximation.
is therefore unclear to what extent treatment may have impacted
the reported outcome data.

Gl1omas, glioneuronal tumou rs, and neuronal tumours 129


Pap illary glioneuronal tumour Varlet P
Komori T
Park SH
Rosenblum MK

Definition
Papillary glioneuronal tumour (PGNT) is a glioneuronal tumour
exhibiting a biphasic pattern with variable representation of pseu -
dopapillary glial structures and interpapillary neuronal compo -
nents, and with PRKCA gene fusion (mainly SLC44A 1::PRKCA
fusion) (CNS WHO grade 1).

ICD-0 coding
9509/1 Papillary glioneuronal tumour

ICD-11 coding
2A00.21 & XH3XU4 Mixed neuronal-glial tumours & Papillary
glioneuronal tumour

Related terminology
Flg. 2.120 Papillary glioneuronal tumour. A Contrast-enhanced T1-weighted image
Not recommended: pseudopapillary ganglioneurocytoma; reveals peripheral enhancement in the cyst. B T2-weighted image shows a cystic
pseudopapillary neurocytoma with glial differentiation . mass pushing the anterior horn of the lateral ventricle.

Subtype(s) tumours have no or only minimal peritumoural oedema, even


None when large (1867,3127) .

Localization Epidemiology
PGNTs are supratentorial, most frequently affecting the tempo- PGNT is rare and the precise incidence remains to be deter-
ral lobe (28%), and they are often in close proximity to the lateral mined . It is a tumour of young adults, with a median patient
ventricles (28%) {1354) . age at diagnosis of 16 years (range: 6-54 years) (1354). No sex
predilection was found in a cohort of patients with PGNT with
Clinical features PRKCA fusion (1354).
Principal manifestations include headaches and seizures.
Haemorrhagic presentation has been reported {399,243) . Etiology
Unknown
Imaging
On MRI, the tumour is well demarcated , solid, and cystic, with Pathogenesis
a contrast-enhancing portion and little mass effect. The solid The hallmark of PGNT is PRKCA gene fusion, mainly
portion is isointense or hypointense on T1 -weighted images SLC44A 1::PRKCA fusion ; the only alternative PRKCA fusion
and hyperintense on T2-weighted images or FLAIR . Most of the partner that has been reported is NOTCH1 {1354,2369,3731

.. .; -
Flg.2.121 Papillary glioneuronaJ tumour. A Biphasic appearance with pse~dopapillary giial structures and interpapillary neuronal component. I lnterpapillary componeril
comprises neurocytes and ganglioid cells. C Hyalinized vessels can be prominent.
Rg. 2.122 Papillary glioneuronal tumour. A GFAP-positive cells in pseudopapillary structures. B lnterpapillary cells show synaptophysin immunoreactivity.

The canonical SLC44A 1::PRKCA fusion results from a reciprocal


translocation t(9 ;17)(q31;q24), with consequent generation of a
constitutively expressed oncoprotein {373) . PRKCA encodes
protein kinase C alpha (PKCA), which is a member of the fam-
ily of calcium- and phospholipid-dependent serine/threonine
kinases that are involved in the MAPK signalling pathway.

Macroscopic appearance
PGNTs are well-delin eated , solid , and cystic lesions . Calcifica-
tion and haemorrhage may be observed .

Histopathology
PGNT is characterized by a distinctive biphasic pattern , con-
sisting of (1) a glial pseudopapillary architecture and (2) an Fig.2.123 Papillary glioneuronal tumour. lnterphase FISH using a fusion probe for
interpapillary component, heterogeneously distributed , with SLC44A1 (green) and PRKCA (orange), showing SLC44A1 ::PRKCA fusion signals (ar-
rows).
considerable variation in size between cases. In a series of
14 molecularly confirmed PGNTs , the pseudopapillary struc-
tures were constant except in 1 case {1354}. The cuboidal index generally does not exceed 2%, but elevated activity (rang-
tumour cells cover hyalinized blood vessels in a single or pseu- ing from 10% to > 50%) has been reported in non-molecularly
dostratified layer. The glial cells have round nuclei and scant confirmed cases (340,18).
cytoplasm. Cellularity varies from case to case . Monomorphic
neurocytes or medium-sized neurons are distributed in a neu- Differential diagnosis
ropil background 11692}. Ganglion cells are not frequent (seen PGNT diagnosis is challenging . In a series of 28 histologically
in 3 of the 14 cases). microcalcifications were present in 5 of diagnosed PGNTs (1354), 17 (60%) clustered with an alterna-
14 cases 11354}, and eosinophilic granular bodies are rare tive methylation class (mainly dysembryoplastic neuroepithelial
11692) . Occasional mitoses may be seen, but microvascular tumour, pilocytic astrocytoma , or pleomorphic xanthoastro-
proli1eration and necrosis are absent 11354}. A few reports have cytoma) . This may reflect the morphological heterogeneity
described anaplastic features in PGNT, but those cases were of PGNT: the neuronal component may be limited, making it
not molecularly confirmed {1459,18). challenging to differentiate from astroblastoma, ependymoma,
pilocytic astrocytoma, or pleomorphic xanthoastrocytoma; con-
lmmunophenotype versely, the neuronal component can be prominent with only
The 1mmunophenotype is usually biphenotypic: glial in pseu- focal papillary architecture (mimicking extraventricular neurocy-
dopaptllary structures and neuronal in interpapillary areas. The toma or ganglioglioma). Moreover, perivascular tropism is not
cuboidal glial cells draping vessels are positive for GFAP and specific to PGNT and is shared by various glial or glioneuronal
S100 . In some cases, oligodendrocyte-like cells that express tumours such as pilomyxoid astrocytoma, astroblastoma,
OLIG2 but are GFAP-negative may be seen 13130). The neu- e~en?ymoma, dysembryoplastic neuroepithelial tumour. gan-
ronal cells and f ibrillary background (neuropil) express neuronal gl1ogl1oma, and angiocentric glioma .
marker s such as synaptophysin and NeuN . However, neurofila-
rnent is mostly confined to ganglioid cells, and chromogranin A Cytology
is not widely expressed 11692). Extravascular CD34 expression Not clinically relevant
may be observed but only focally 123691. The Ki -67 proliferation

lion 1<1''"' · olloneu


:::; ro na
· I tun ·1ours, and neuronal tumours 131
Box2.23 Diagnostic criteria for papillary glioneuronal tumour formalin-fixed, paraffin-embedded sections {2369), but RNA
Essential: sequencing can also be used 1460}. No PRKCA recurrent muta.
Biphasic histological and immunophenotypic pattern with pseudopapillary glial lining
tion point has been reported in PGNT.
and interpapillary neuronal components PGNTs exhibit a highly characteristic and diagnostic meth-
ylation profile 11354}. Copy-number variation analysis in th(:
AND
PGNT methylation group reveals flat profiles or only focai
PRKCA gene fusion (mostly SLC44A 1::PRKCA)
gains in a region of 17q that includes PRKCA (in 50% of case 51
AND (for unresolved lesions)
{1354} .
Methylation profile of papillary glioneuronal tumour
Desirable: Essential and desirable diagnostic criteria
Well-delineated, solid and cystic tumour See Box 2.23 .
Caveat: Diagnosis should be heavily weighted towards molecular findings because
morphological analyses frequently result in mistyping {1354}. Staging
Not clinically relevant

Diagnostic molecular pathology Prognosis and prediction


PRKCA gene fusion (mainly SLC44A1 :: PRKCA) is the hallmark PGNTs correspond to CNS WHO grade 1, and gross total
of PGNT {1354,2369,373}. To date, SLC44A1 ::PRKCA fusion resection constitutes the main prognostic factor. One molecu-
has not been observed in other brain tumour types except one larly confirmed case has been described as recurrent (1354!
neurocytoma {1354). Two alternative fusions , PRKCA::FAT1 Morphologically diagnosed cases with a Ki-67 proliferation
and PRKCA::FAM91A 1, have been described in the diffuse index > 20% or anaplastic features have been reported , wrt
glioma MYB- or MYBL 1-altered methylation cluster {1354} . The tumoural progression or dissemination {39,1459). Such features
fusion gene can be detected by simple interphase FISH on have not been described in PRKCA-fused cases .

132 Gl 1urr1C:JS , ~l1oneuronal turn o tJrs , and ne uronal tumou rs


Rosette-forming glioneuronal tumour Hainfellner JA
Jacques TS
Jones DTW
Rosenblum MK
Sievers P

Definition Clinical features


Rosette-form ing glioneuronal tumour (RGNT) is a glioneuronal Patients most commonly present with headache and diminished
tumour composed of two distinct histological components: one vision I papilloedema and/or ataxia, vertigo, diplopia, and dys-
containing uniform neurocytes forming rosettes and/or perivas- arthria. Cervical pain is occasionally experienced . Rare cases
cular pseudorosettes, and the other being glial in nature , with are asymptomatic and discovered as incidental imaging find-
piloid and oligodendroglia-like cells , resembling pilocytic astro- ings.
cytoma. These tumours are characterized by FGFR1 mutation
with frequent co-occurrence of a PIK3CA and/or NF1 mutation Imaging
(CNS WHO grade 1). On CT, RGNTs appear hypodense . On MRI , RGNTs appear
as relatively circumscribed, solid, cystic-solid , or multicystic
ICD-0 coding tumours showing hypointensity on T1-weighted images and
9509/1 Rosette-forming glioneuronal tumour diffusion-weighted imaging, hyperintensity on T2-weighted
images, and focal/multifocal and rim gadolinium enhancement.
ICD-11 coding Contrast enhancement may fluctuate and spontaneously disap-
2A00 .21 & XH2JU8 Mixed neuronal-glial tumours & Rosette- pear. Haemorrhage and peripheral heterogeneous enhance-
forming glioneuronal tumour ment is common {2060,1030,3128).

Related terminology Epidemiology


None RGNTs are slow-growing; they preferentially affect young
adults, adolescents, and children ; and they are rare . Specific
Subtype(s) population-based incidence rates are not available.
None
Etiology
Localization Genetic susceptibility
RGNTs arise in the midline. They usually occupy the fourth RGNT has been described in patients with neurofibromatosis
ventricle and/or aqueduct and can involve adjacent brainstem , type 1 {91,1588,2925) or Noonan syndrome {1555,1896) .
cerebellar vermis , quadrigeminal plate, pineal gland, or thala-
mus. Localization outside the fourth ventricle (e.g. in the pin- Signalling pathways
eal region , spinal cord , optic chiasm , septum pellucidum , and The MAPK and Pl3K signalling pathways appear to synergis-
diencephalic region) has been described {105,3497,50). In rare tically interact in the formation of RGNTs , and these tumours
cases , dissemination throughout the ventricular system can seem to be driven by constitutive activation of FGFR signalling
occur 13386 ,81}. together with frequent PIK3CA mutation {1896,2929).

Fig. 2.124 Rosette-lorming glioneuronal tumour A Axial T1 -weighted MRI wit1·1out contrast showing a cerebellar m1dline mass with solid and cystic components. 8 Axial T2·
we1gh1ed MRI with cerebrospinal fluid suppression demonstrating the heterogeneous s.lgnal of the partially solid , partially cystic rn1dltne mass. Note that the content of the cystic
components is not isointense to cerebrospinal fluid . c Sag1ttal T2-weighted MRI showing compression of the fourth ventricle due to the cerebellar m1dline mass. As an effect of
the obstructive hydrocephalus, the floor of the third ventricle is herniated into the suprasellar cistern .

U l1u 111<J'._, , ul 1<111 d ur<H1dl lur11ull 1'.> . drill rh ~ u r uri ~ d 11111 1utJ r' S 133
Pathogenesis have spherical nuclei with fine ly granular chromatin and incori .
Cell of origin spicuous nucleoli, scant cyto plasm , and delicate cytoplasmic
Neuroimaging, histological findings , and molecular evidence processes . These neurocytic structures may lie in a partly
indicate that RGNT may arise from brain tissue surrounding the microcystic , mucinou s matrix . The glial component of RG NT
ventricular system . For cases affecting the fourth ventricle, an typically dominates and in most areas resembles pilocyt1c
origin from the subependymal plate or the internal granule cell astrocytoma . Astrocytic tumour cel ls are spindle to stellate 1r
layer of cerebellum has been suggested (1693,3188}. shape, with elongated or oval nuclei and moderately dense
chromatin . Cytoplasmic processe s often form a compact to
Genetic profile loosely textured fibrillary background . In some areas , the glial
Epigenetically, RGNTs display a distinct DNA methylation pro- component may be microcystic , containing round to oval
fil e (2929}. At the genomic level , FGFR1 hotspot mutations are ol igodendrocyte-llke cells with perinuclear haloes . Rosenthal
typical, with co-occurrence of PIK3CA mutations in the majority fibres , eosinophil ic granular bod ies , microcalcifications, and
of cases and additional loss-of-function mutation in NF1 in a haemosiderin deposits may be encountered . Overall , cellular-
subset of cases (833,3188,1074,1643,2929). ity is low and necrosis is absent. Vessels may be thin-walled
and dilated or hyalinized . Thrombosed ve ssels and glomeru-
Macroscopic appearance loid vasculature may also be seen . Ganglion cells are occa-
RGNTs are soft, gelatinous , general ly well-demarcated tumours sionally present, but adjacent perilesional cerebellar cortex
(1693). does not show dysplastic changes .

Histopathology Proliferation
RGNTs are generally demarcated , but limited infiltration may The Ki-67 proliferation index is low(< 3% in rep orted cases) and
be seen . They are characterized by a biphasic neurocytic mitoses are usually absent.
and glial architecture (1693 ,2569 ,1438}. The neurocytic com -
ponent consists of a un iform population of neurocytes form- lmmunophenotype
in g neurocytic rosettes and/or perivascular pseudorosettes . lmmunoreactivity for synaptophysin is present at the centres of
Neu rocytic rosettes feature ring-shaped arrays of neurocytic neurocytic rosettes and in the neuropil of perivascular pseudor-
nuclei around del icate eosinophil ic neuropil cores . Perivascu- osettes (1693 ,2569,1438). Both the cytoplasm and processes
lar pseu dorosettes feature delicate cell processes rad iating of neurocytic tumour cells may express MAP2. In some cases.
toward s vessels . Both patterns , when viewed longitudinally, NeuN positivity can be observed in neurocytic tumour cells.
may show a co lumnar arrangement. Neurocytic tumour cells Tumour cells show nuclear expression of OLIG2. GFAP and

134 Gliomas. gl1oneuronal tumours. and neuronal tumours


s1 00 immunoreactivity is present in the gllal component but Box 2.24 Diagnostic criteria for rosette-forming glioneuronal tumour
absent in rose ttes an d pseudorosettes .

~
Essential:
Biphaslc histomorphology with a neurocytlc component and a glial component
Electron microscopy
Astrocytic cells of the glial component contain dense bundles AND

"
of glial filaments . Rosette-forming neurocytic cells are intimately Uniform neurocytes forming rosettes and/or perlvascular pseudorosettes associated
with synaptophysin expression
apposed and feature spherical nuclei with delicate chromatin ,
cytoplasm containing free ribosomes, scattered profiles of AND (for unresolved lesions)
rough endoplasmic reticu lum , prominen t Golgi complexes, and Small biopsies showing only one tumour component (neurocytic or gllal) and a
methylatlon profile of rosette-forming glioneuronal tumour
occasional mitochondria . Loosely arran ged cytoplasmic pro-
cesses form the centres of ro settes and contain aligned micro- Desirable:
tubu les as well as occasional dense-core granules . Presynaptic FGFR1 mutation with co-occurring PIK3CA and/or NF1 mutation
specializations may be seen , and mature synaptic terminals
may form surface contacts with perikarya and oth er cytoplas-
mic processes (1693) .
mutation (2929 ,19531. Accompanying NF1 mutation can also
Differential diagnosis occur, but this does not provide diagnostic specificity for RG NT
The main differential diagnosis is pilocytic astrocytoma. Di'ffuse because it can also be encountered 1n dysembryoplast1c
leptomeningeal gl ioneuronal tumour may also be a differential neuroe pithelial tumour or pilocytic astrocytoma (2929,1953).
diagnosis . Because of the high number of misdiagnoses , mo lecular testi ng
is highly recommended to confirm RGNT diagnosis (2929 }.
Cytology
In smear preparations , neurocytic cells feature round nuclei Essential and desirable diagnostic criteria
with granular chromatin, inconspicuous nucleoli, and scant See Box 2.24 .
.c
cytoplasm . Cytolog ical atypia Is absent. Delicate , elongated
processes are seen in the background . Pilold astrocytes with Staging
~
'

elongated nuclei and coarse bipolar processes may be evident. Not cl inically relevant
Clustering of tumour cells may be present (1693).
Prognosis and prediction
Diagnostic molecular pathology The cli nical outcome is favourable in terms of survival, but
~ . RGNTs are defined by a distinct DNA methylation profile . They d isab ling postoperative deficits have been reported in approxi-
are also characterized by FGFR1 hotspot mutation (FGFR1 mately half of th e cases. In rare cases. tumour dissemination,
p.N546 or p.K656) in combination with either PIK3CA or PIK3R1 recurrence, or progression has been described (3191 ,2846 ,81).

Gl1 omas . glioneuronnl tu rno 1rs , dlld neuro1 ,.:i.1 tum\lur~, 135
Myxoid glioneuronal tumour Solomon DA
BIOmcke I
Gessi M
Hawkins CE
Jones OTW

Definition
Myxoid glioneuronal tumou r is a low-grade glioneuronal tumour
typically arising in the septal nuclei , septum pellucidum , cor-
pus callosum , or periventricular white matter. The tumour is
characterized by a proliferation of oligodendrocyte -like tumour
cells embedded in a prominent myxoid stroma, of ten including
admixed floating neurons , neurocylic rosettes , and/or perivas-
cular neuropil. There is a recu rrent dinucleotide mutation at
codon p.K385 in the POGFRA gene (CNS WHO grade 1).

ICD-0 coding
9509/1 Myxoid glioneuronal tumour
Fig.2.127 Myxoid glioneuronal tumour. This tumour with characteristic POGFRA
ICD-11 coding p.K385 mutation is centred in the septa! nuclei at the base of the septum pelluc1durn
and is hyperintense on sagittal (A) and axial (B) T2-FLAIR MRI. There 1s also d1ssern1·
2A00.21 Mixed neuronal-glial tumours
nated disease throughout the ventricular system , with hyperintense nodules along the
ependymal surface of the lateral, third, and fourth ventricles (seen in A).
Related terminology
Not recommended: dysembryoplastic neuroepithelial tumour- Clinical features
like neoplasm of the septu m pellucidum /177}; septal dysem- Presentin g symptoms in patients with myxoid glioneuronaJ
bryoplastic neuroepithel ial tumour /569] . tumour are variable, but they most commonly include head-
aches, emesis, seizures, and behavioural disturbance /569].
Subtype(s)
None Imaging
On imaging , myxoid glioneuronal tumours are well-circum-
Localization scribed lesions that are T1-hypointense and T2-hyperintense,
Myxoid glioneuronal tumours are most commonly centred in without contrast enhancement or restricted diffusion. On sus-
the septal nuclei and septum pellucidum , but examples have ceptibility-weighted imaging , they occasionally demonstrate
also been reported in the genu of the corpus callosum and the artefact suggestive of blood products from prior intratumoural
periventricular white matter of the lateral ventricles (1954) . haemorrhage_Tumours centred in the septal nuclei and septum
Th is newly recognized tumour type probably encompasses pellucidum are often associated with obstructive hydrocephalus.
a large subset of those neoplasms previously reported as dys- whereas tumours centred in the corpus callosum and penven-
embryoplastic neuroepithelial tumour and rosette-forming gllo- tricular white matter have not been associated with hydrocepha-
neuronal tumour centred within the deep periventricular white lus. A subset of patients have disseminated disease throughout
matter, lateral ventricles , and septum peliucidum 11184,1243, the ventricular system at time of initial presentation 1569,1954)_
2785,489,3493,3492,1072}.

.·.· .
'
...•
~

•·'

t . 'I• : . • • - - •
Flg.2.128 Myxoid glioneuronal tumour. These tumours are histologically characterized by a prolif~ration of oligodendroc~t.e - like cells in a myxold stroma with a delicate capillary
network, occasionally containing floating neurons, neurocytic rosettes, and/or perlvascular neurop1L A Low-power magrnhcat1on. 8 High-power magnification.

136 Gl1omas . glioneuronal tumours . and neuronal tumours


Epidemiology
Myxoid glioneuronal tumours are rare primary brain tumours ,
with < 100 cases reported to date. They have an approximately
equal sex distribution and predom inantly occur in chi ldren and
young adults, with a peak incidence in the second and third
H istopathology
Myxoid glioneuronal tumour is a mostly circumscribed , non-
infiltrative glioneuronal neoplasm that is h1stolog1cally charac-
terized by a proliferation of oligodendrocyte-like tumour cells
embedded in a prominent myxoid stroma . Some examples
I
~

decades of life 1569,1954). contain admixed floating neurons and perivascular neuropli
resembling dysembryoplastic neuroep1thelial tumour, whereas
Etiology others contain neurocytic rosettes and perivascular neuro-
No risk factors or inherited genetic susceptibility have been pil resembling rosette-forming glioneuronal tumour. Myxo1d
reported to date. glioneuronal tumours lack the multinodularity with patterned
mucin-rich nodules that is characteristic of dysembryoplastic
Pathogenesis neuroepithelial tumours of the cerebral cortex . Rosenthal fibres ,
Myxoid glioneuronal tumours are genetically characterized by eosinophilic granular bodies, and microcaJcifications typical of
a recurrent dinucleotide mutation in the PDGFRA gene, which other low-grade neuroepithelial tumour entities are not com-
encodes platelet-derived growth factor receptor alpha (PDG - monly observed in myxoid glioneuronal tumours . Mitotic activity
FRA), a transmembrane receptor tyrosine kinase . The char- is typically very low or absent (569,1954).
acteristic mutation results in substitution of lysine with either
leucine or isoleucine at codon 385 (p.K385L or p.K3851) in lmmunophenotype
the vast majority of cases 12987,569,1954). These p.K385L By immunohistochemistry, myxoid glioneuronal tumours are
or p.K3851 mutations have been somatic (tumour-specific) in characterized by diffuse strong positivity for OLIG2, SOX10 ,
all examined cases to date, and they typically occur in the GFAP, and MAP2 in the oligodendrocyte-like tumour cells .
absence of accompanying PDGFRA gene amplification . Synaptophysin staining is typically absent in the oligodendro-
A single case has been described harbouring a POGFRA cyte-like cells , but it is found in the floating neurons , neurocytic
p.E362delinsEW mutation instead of the canonical dinucleo- rosettes , and perivascular neuropil . CD34 staining is typically
tide mutation at codon p.K385 1569). The PDGFRA mutation limited to the endothelial cells of the delicate capillary network.
is typically the solitary pathogenic alteration identified , with The Ki-67 labelling index is uniformly low(< 5%) (569,1954).
an absence of other accompanying genetic drivers In all
cases studied to date. Most cases have had balanced diploid Cytology
genomes without recurrent cytogenetic alterations. Although On intraoperative cytological preparation , these tumours are
the functional impact of this specific p.K385L or p.K3851 muta- characterized by ol igodendrocy te -like cells in a prominent
tion in POGFRA has not been studied to date, other mutations myxoid background . Occasional neurons or neurocytic rosettes
or intragenic deletions within the extracellular domain of the can be seen .
POGFRA gene , which have been recurrently found in high-
grade gliomas , have been shown to cause constitutive acti- Diagnostic molecular pathology
vation of the Intracellular tyrosine kinase domain (TKO) and Myxoid glioneuronal tumours are genetically characterized by
downstream activation of the Pl3K and MAPK signalling path- a dinucleotide mutation in the PDGFRA gene that results in
ways (2358,2425) . an amino acid substitution, either p.K385L or p.K3851 , within
the extracellular ligand-binding domain of the protein (2987,
Macroscopic appearance 569,1954}. Less commonly, other mutations in the extracellular
Myxoid glioneuronal tumours are often soft, gelatinous , grey domain of PDGFRA occur (569). Most cases have had balanced
lesions.

Domains Position
lg-like C2-type 1 24 - 113
• lg-like C2-type 2 117-201
• lg-like C2-type 3 202 - 306
• lg-like C2-type 4 319-410
~ p.K3851 (n=2) lg-like C2-type 5 414 -517
....
.... p.K385L (n=6)


Transmembrane domain
Protein tyrosine kinase
529 - 549
539 - 954

lg-like C2 lg-like C2 lg-like C2 lg-like C2 PDGFRA

I I I I I I I I I I I I I
2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 : exons

Flg.2.129 Myxoid glioneuronal tumour. Diagram of the PDGFRA protein annotated with the location of the characteristic p.K385L or p.K3851 d1nucleotide mutation lound 1n
8 cases of myxo1d glloneuronal tumour (1954).

Gliornas . gl 1on eur o11al tumour s . :md 11eur iJ rk\I tL JJ t.('L. r '; 137'
diploid genomes without recurrent cytogenetic alterations . Box2.25 Diagnostic criteria for myxoid glioneuronal tumour
Myxoid glioneuronal tumours lack genetic alterations that are Essential:
characteristic of other glioneuronal tumours and pilocytic astro- · prominent myxoid stroma
Oligodendrocyte-like tumour cells embedd ed 1n a
cytoma (e.g . BRAF, FGFR1 , Pll<3CA, PIK3R1, NF1 , PTPN11, or
AND .
MAP2K1 alterations) . Genome-wide DNA methylation profiling . callosum or periventricular
Location In septa! nuclei, septum pelluc1dum, corpus •
has revealed that myxold glioneuronal tumours harbour a dis-
white matter
tinct epigenetic signature that is closely related to that of dysem-
bryoplastic neuroepithelial tumour of the cerebral cortex (2987, Desirable:
PDGFRA p.K385UI dinucleotide mutation or (less commonly) other mutations in the
569 ,565) .
extracellular domain of PDGFRA
Essential and desirable diagnostic criteria Methylatlon profile of myxoid glioneuronal tumour
See Box 2.25 . Note: Desirable diagnostic criteria can be essential for unresolved cases.

Staging
Not clinically relevant ventricular system after subtotal resection . but they c ontinue to
be associated with indolent behaviour. High-grade transforma-
Prognosis and prediction tion of myxoid glioneuronal tumours has not been described
Myxoid glioneuronal tumours are indolent, slow-growing to date. Clinical experience for this rare tumour type remains
tumours associated with favourable long-term outcomes in the limited , but the outcomes reported to date are favourab le and
absence of radiotherapy and chemotherapy \569 ,1954}. A sub- equivalent to those of CNS WHO grade 1 tumour types such as
set of tumours can recur locally or disseminate throughout the pilocytic astrocytoma and rosette-forming glioneuronal tumour

138 Gl1omas . glloneuronal tumours . and neuronal tumours


Diffuse leptomeningeal glioneuronal tumour Perry A
CapperD
Ellison OW
Jones DTW
Reifenberger G

Definition
Diffuse leptomeningeal glioneuronal tumour (DLGNT) is a
glioneuronal neoplasm that commonly involves the Jeptomenin-
ges diffusely, is composed of oligodendrocyte-like cells , and
is molecularly characterized by chromosome arm 1p deletion
and a mitogen-activated protein kinase (MAPK) pathway gene
alteration, most commonly KIAA1549::BRAFfusion .

ICD-0 coding
9509/3 Diffuse leptomeningeal glioneuronal tumour

ICD-11 coding
2A00.21 Mixed neuronal-glial tumours

Related terminology
Not recommended: disseminated oligodendroglioma-like Jep-
tomeningeal neoplasm; primary leptomeningeal oligodendro-
gllomatosis.

Subtype(s)
Diffuse leptomeningeal glioneuronal tumour with 1q gain; dif-
fuse leptomeningeal glioneuronal tumour, methylation class 1
(DLGNT-MC-1); diffuse leptomeningeal glioneuronal tumour,
methylation class 2 (OLGNT-MC-2)

Localization
These tumours preferentially involve the spinal and intracranial
leptomeninges, although there are rare parenchymal examples
without a leptomeningeal component, most often located In the
spinal cord but occasionally also in the cerebral hemispheres
(568,1 545 ,125). In the intracranial compartment. leptomenin-
geal growth is most commonly seen in the posterior fossa,
around the brainstem , and along the base of the brain . One or Fig. 2.130 Diffuse leptomeningeal glioneuronal tumour. A Note the extensive 1ntra-
more circumscribed, intraparenchymal, cystic or solid tumour ventricular involvement, as well as the parenchymal cysts. B Extensive spinal lep-
nodules may be seen , with spinal lntramedullary lesions being tomeningeal involvement.
more common than intracerebral masses 12696).
Discrete intraparenchymal lesions , most commonly in the spi-
Clinical features nal cord, were found in 25 (81%) of 31 patients in the largest
Patients often present with acute onset of signs and symptoms reported cohort [2696) . Patients also commonly demonstrate
of increased lntracranial pressure due to obstructive hydro- obstructive hydrocephalus with associated periventricular T2
cephalus , including headache, nausea, and vomiting . Opis- hyperintens1ty.
thotonos and signs of spinal or cranial nerve damage may be
present. Some patients show ataxia and signs of spinal cord Epidemiology
compression . Rarely, patients present with epilepsy. DLGNTs are rare , and data on incidence are not available, but
these tumours mostly affect paediatric patients . In the largest
Imaging published series (36 patients) {2696). the median age at diag-
In most cases, MRI shows widespread diffuse leptomenin- nosis was 5 years (range: 5 months to 46 years). Another study
geal enhancement and thickening along the spinal cord, often found that the median age for DLGNT-Mc -·1 (5 years; ran ge
extending intracranially to the posterior fossa, bra1nstem, and 2- 23 years) was lower than that for OLGNT-MC -2 (14 years,
basal cisterns . Small cystic or nodular T2-hyperintense lesions range : 5- 47 years) (736) . The M·F ratio is roughly 1 fr 1 12659,
along the subp1al surface of the spinal cord or brain are frequent. 2696,2854,7361

Glromas . gl1oneuronal tumou 1s ,rn cJ nou ror1,JI tLWl L lHS 139


Etiology . .
The etiology of DLGNT is unknown . The vast maior.1ty of tu.mours
deve lop spontaneously, without evidence of gen~t1c pred1spos1-
tion or exposu re to specific carcinogens . In a series of 36 cases
no evidence of recognized genetic predisposing feature s Gr
other tumour syndromes was observed , although 1 ~atient had
a constitutional 5p deletion, 1 patient had a coex1s~1ng type 1
Chiari malformation , and 1 patient had a factor V Leiden muta-
tion 12696).

Pathogenesis
Cell of origin
The cellular origin of DLGNTs is unknown. The absence of obvi-
ous parenchymal lesions in some patients suggests an origin
from displaced neuroepithel ial cells within the meninges. How-
ever, an intraparenchymal origin is also possible , g iven that
small intraparenchymal foci are frequentl y present in addition
to the diffuse leptomeningeal tumour spread . Given the partial
overlap in genetic features with oligodendroglioma and pilocyt1c
astrocytoma, an ori gin from a precursor cell just upstream of
th is li neage segregation has also been specu lated {736) .

Genetic profile
A frequent genetic alteration in DLGNTs reported to date 1s
KIAA 1549::BRAF fusion, found in 41 (72%) of 57 investigated
cases from th e combined data of four independent studies
{2702). Less common MAPK alterations include other BRAF
alterations (including BRAF p.V600E mutations); NTRKI.
NTRK2, or NTRK3 gene fusions; FGFR1 mutation; and RAFI
rearrangements 1768,814,736 ,125}. Deletions of ch romosome
arm 1p are also frequently observed in FISH analys is and were
reported in 10 (59%) of 17 tumours in one series , and in 100% of
tumours identified by DNA methylation profil ing 124,342.1038.
2696 ,2702,736) . In cases with SNP array data or copy-number
profiling from DNA methylation data, complete 1p arm loss was
demonstrated 12696,2702,736). Codeletion of 1p and 19q was
observed at a frequency of 18% (3 of 17 tumours) in one series
12702) and in 10 (33%) of 30 tumours identified by DN A meth-
ylation profiling . In one case with 1p/1 9q codeletion , a t(1p ;19q)
(q10;p10) translocation was demonstrated by FISH {2730} No
mutations in IDH1 or IDH2 have been reported to date.
With!n two distinct DNA methyl ation subclasses , 1q gain was
found 1n all 13 cases of DLGNT-MC-2 and in 6 of 17 cases or
DLGNT-MC-1 in one study (63 % of all DLGNTs) {7361 . Another
g~oup similarly f?und 1q gain in 14 (56%) of 25 DLGNTs 15641.
Single cases with H3 p.K28M (K27M) mutation have been
reported , but their relationship to H3 K27- altered diffuse midline
glioma remains open {81 4.2208).

Macroscopic appearance
DLGNT is generally char~cterized by a highly mucoid cut
surface, whether present 1n the parenchyma, intraventricular
spaces, or leptomeningeal . spaces. Leptomeningeal tumour
fre q ~e ntly exte~ds along V1rchow- Robin spaces. sometimes
forming expansive cystic lesions. Obstructive hydrocephalus 15
"" , common .
Fig. 2.131 Diffuse leptomenlngeal glioneuronal tumour. A Marked expansion and
hyp~rcellularity of the leptomeninges B Extension along the perivascular Virchow-
Robm space Is evident within the adjacent brain parenchyma of this tumour. C Neu-
Histopathology
ronal differentiation Is evident in the form of neurocytic rosettes and perivascular pseu- The . often made on meningeal b 1opsy or "1
. diagnosis is most
dorosettes. D Neuronal diflerentlation is evident in the form of ganglion cells. biopsy taken from discrete intraparenchymal lesions . DLGN T::;

140 Gl1ornas , g11oneu1onc1I tumours arid neuronal tumours


;. ' ,. ..
• I •
, •'
r
I
I

•/
~
...
,! ..
. , . ... "'..
, . . .- . . "
..·. ' ,
~ .,

.'.

. .. '· • • .B
Fig. 2.132 Diffuse leptomeningeal glioneuronal tumour. A Tumour cells show extensive nuclear immunoreactivity tor OLIG2. B The tumour cells are GFAP-negative.

are low- to moderate-cellularity neoplasms composed of rela- reporting less favourable outcome associated with a prolifera-
I
tively monomorphic oligodendrocyte-like tumour cells with uni- tion index of > 4% (2696). Brisk mitotic activity was defined as
form , medium-sized round nuclei and inconspicuous nucleoli. ;;:: 1.7 mitoses/mm 2 (equating to;;:: 4 mitoses/10 HPF of 0.55 mm
Like in oligodendrogliomas, clear perinuclear haloes are some- in diameter and 0.24 mm 2 in area) in one study [2696) .
times seen in DLGNTs as an artefact in formalin-fixed, paraffin-
embedded tissue sections. The tumour cells grow diffusely or lmmunophenotype
in small nests in the leptomeninges, with desmoplastic and The oligodendroglial-like tumour cells typically express OLIG2 ,
myxoid changes commonly present. A storiform pattern may be MAP2, and S100 {2566}. GFAP immunoreactivity in tumour cells
observed in desmoplastic areas. Histological features of ana- is seen In < 50% of cases and is often restricted to a minor
plasia are rare and include increased cytological atypia, brisk proportion of neoplastic cells. Expression of synaptophysin is
mitotic activity, microvascular proliferation, and/or palisading detectable in as many as two thirds of the tumours. and it is
necrosis at primary presentation or after tumour progression particularly common in those containing neuropil aggregates
l2696); one example also featured a spongioblastic pattern with and ganglion cells. NeuN , neurofilament, and chromogranin
rhythmic nuclear palisades [3513}. A small subset of tumours stains are usually positive in only those tumours with more overt
contain overt neuronal differentiation, in the form of neurocytic neuronal features on routine histopathology. EMA and IDH1
rosettes , delicate perivascular pseudorosettes, neuropil-like p.R132H stains are negative.
islands, and/or ganglion cells. Rarely, eosinophilic granular
bodies are observed . Rosenthal fibres are usually absent. The Differential diagnosis
intraparenchymal component may resemble a dysembryoplas- The main differential diagnoses are intraparenchymal astrocytic
tic neuroepithelial tumour or a diffusely infiltrative glioma, mostly or oligodendroglial gliomas with leptomeningeal dissemination;
oligodendroglioma, although astrocytic features occasionally for instance, pilocytic astrocytoma may also show leptomenin-
predominate. geal dissemination. An absence of (or only focal) GFAP immu-
noreactivity, the presence of synaptophysin-positive cells , and
Grading the characteristic molecular profile of KIAA 1549::BRAF fusion
Histologically, the vast majority of DLGNTs are well-differenti- with isolated 1p deletion or 1p/19q codeletion in the absence
ated low-grade lesions. Nevertheless, a subset of tumours may of IDH mutation (2702,736) distinguish DLGNT from pilocytic
show histological features of anaplasia or molecular alterations astrocytoma, dysembryoplastic neuroepithelial tumour, oligo-
associated with shorter survival. The data for assigning distinct dendroglloma, ganglioglioma, and diffuse astrocytoma. Pleo-
grades to this tumour type and its subtypes are still limited. morphic xanthoastrocytomas can typically be distinguished
Nevertheless, the clinical courses reported to date have been by their pleomorphlc histology, often with associated BRAF
roughly similar to those of CNS WHO grade 2 entities for cases p.V600E mutation and CDKN2A and/or CDKN2B homozygous
of conventional DLGNT and DLGNT-MC-1, and to those of CNS deletion (2702,3300).
WHO grade 3 entities for tumours with anaplastic features, 1q
gain, and/or the DLGNT-MC-2 profile . Cytology
Cerebrospinal fluid examination demonstrates elevated protein
Prollferation levels, although cytology is often negative 12696,2854) . There-
Mitotic activity is sparse, and the Ki-67 proliferation index is usu- fore , the diagnosis usually requires a biopsy. lntraoperative
ally low, with a median value of 1.5% reported in one series smear from a parenchymal lesion often resembles oligodendro-
l2696J. However, some cases have an elevated Ki-67 prolif- glioma, whereas a meningeal biopsy is often less productive
eration index as evidence of anaplasia (25661. with one study due to increased fibrous tissue.
Fig. 2.133 Diffuse leptomeningeal glioneuronal tumour. FISH studies using BRAF (red) Fig.2.134 Diffuse leptomeningeal glioneuronal tumour with 1q gain. FISH studies ;s.
and KIAA1549 (green) probes show increased copy numbers and yellow fusion signals. ing chromosome 1p (orange) and 1q (green) probes show simultaneous 1p loss 0r 11
1 signal) and 1q gain (> 2 signals) in most tumour nuclei.

Box2.26 Diagnostic criteria for diffuse leptomeningeal glioneuronal tumour frequent MAPK alterations shou ld be explored For prognos·
Essential: tic purposes , chromosome 1q statu s should also be tested
Oligodendroglioma-like morphology Alternatively, DNA methylation profilin g can be performed '.or
DLGNT class assignment, as well as for chromosome 1p and
AND
1q copy-number alterations .
OLIG2 and synaptophysin immunoreactlvity
AND Essential and desirable diagnostic criteria
Chromosome arm 1p deletion See Box 2.26 .
AND
MAPK pathway alteration (mostly KIAA 1549::BRAFtusion) Staging
AND (for unresolved lesions) Not clinically relevant
Methylation profile of diffuse leptomeningeal glioneuronal tumour
Prognosis and prediction
Desirable:
DLGNTs may go through periods of stability or slow progres-
Childhood onset sion over many years , although often wi th considerable morbid-
Leptomeningeal dissemination ity 12696). In a retrospective series of 24 cases with a med1ar
Caveat: This tumour type shows molecular overlap with pilocytic astrocytoma available follow-up of 5 years , 9 patients (38%) died between
(KIAA 1549::BRAFfusion) and oligodendroglioma (1p/19q codeletion). All diffuse 3 months and 21 years after diagnosis (median : 3 years) 126961
leptomeningeal glioneuronal tumours are wildtype in IDH1 and IDH2. and 8 of the 24 patients lived for > 10 years after diagnosis
12696). Mitotic activity, a Ki-67 proli feration index of> 4% arc
microvascular proliferation at the initial biopsy were -each sig-
Diagnostic molecular pathology nificantly associated with decreased overall survival 126961
For cases with typical clinical and histological features , molecu- One study showed estimated 5-year overall survival rates or
lar testing may be less critical . However, for any cases with diag- 100% and 43% in the DLGNT-MC -1 and DLGNT-MC-2 sub-
nostic uncertainty, the initial molecular workup should include classes, respectively 1736). Another study showed substan-
chromosome 1p status and KIAA1549:: BRAFfusion testing . If tially decreased progress ion -free and overall survival times ror
1p deletion is not detected , the diagnosis of DLGNT is unlikely. patients whose DLGNT harboured 1q gain versus those without
If KIAA1549::BRAFfusion is not detected , the possibility of less this gain (564) .

142 Gl1ornas . y l torit~urona l turnuurs , and neuronal turnour::;


Multinodular and vacuolating Rosenblum MK
Giangaspero F

I
neuronal tumour Giannini C
Huse JT
Komori T
Pekmezci M

Definition
The multinodular and vacuolating neuronal tumour (MVNT) is
composed of monomorphic neuronal elements distributed in
LI
I ]4
discrete and coalescent nodules, with vacuolar changes in • Male (n=28)
tumour cells and their matrix (CNS WHO grade 1). ~ 1, J2
• Female (n =19)

I
11

F~ I ~
ICD-0 coding
9509/0 Multinodular and vacuolating neuronal tumour
a; I

~ : -. - --=
ICD-11 coding
2A00.21 Mixed neuronal-glial tumours

Related terminology
Not recommended: diffuse gangliocytoma. Patient age (in decades)

Subtype(s)
None Flg.2.135 Multinodular and vacuolating neuronal tumour. Distribution of cases by
patient age at diagnosis.

Localization nodules in deep cortex and superficial white matter, typically


MVNTs typically involve the deep cortical ribbon and superficial without associated mass effects, oedema, restricted diffusion ,
white matter, predominantly of the temporal lobes (75-80%), or contrast enhancement {1383,3179,2443 ,581}. Lesion stability
followed by the frontal lobes (10-15%), and then the parietal on surveillance imaging is the rule .
and occipital lobes {1383, 3179,2443 ,581) . A case involving the
basal gangl ia has been reported {3245). Epidemiology
Population -based incidence data are not available . However,
Clinical features pathologically confirmed cases have been encountered mainly
Seizures , particularly of complex partial type with or without in adults (age range: 5-7 1 years; median: 42 years) , with few
secondary general ization , are the most common manifestation paediatric examples recorded ; the M:F ratio is 1.5:1 {1383,3179,
(~ 60% of cases) , followed by headache (10-15%), episodic 2443,581}.
confusion, and dizziness . MVNTs may be incidentally discov-
ered {307,2918) . Etiology
Factors predisposing individuals to the development of MVNTs
Imaging have not been identified . An isolated lesion having the typical
Highly characteristic on MRI , although not always apparent, is neuroradiological features of MVNT, but not biopsied, has been
the clustering of discrete or coalescent T2-FLAIR-hyperintense reported in a patient with Klinefelter syndrome 12147).

Fig. 2.136 Multinodular and vacuolating neuronal tumour A Confluent and nodular FLAIR hyper intensity, involvement of cortex and supert1cial white matter. and absence ol
mass effect are characteristic on MRI. B MRI demonstrating FLAIR -hyperintense, clustered nodular lesions in the deep cortex and superficial subcort1cal white matter. C Axial
MRI demonstrating clustered , T2-hypenntense nodular lesions 1n the superficial subcort1cal wt11te matter.
. . ... ~
Fig. 2.137 Multinodular and vacuolating neuronal tumour. A Discrete tumour nodules with vacuolar changes are apparent.
size populate this nodule with vacuolar matrix changes.
e Mature-appearing neurons of intermediate to large

Pathogenesis RAF/MAPK signalling have been consistently identified , with


The clonal genetic abnormalities characterizing MVNTs indi- MAP2K1 and BRAFmutations (other than BRAFp.V600E) being
cate that these are neoplasms rather than malformations , but recurrently identified {2443}.
the precise role of MAPK pathway activation in this setting is
not known . An origin from developmentally dysregulated , par- Macroscopic appearance
tially arrested neuronal or glioneuronal precursors destined for MVNTs are characterized by multiple discrete or coalescent
deep cortical layers could account for the characteristic locali- grey nodules involving the deep cortex, grey matter-wh ite mat-
zation {31 79) . Somatic gene abnormalities that activate RAS/ ter junction, and subcortical white matter {1383,307).

·•

,. . .. •·
Fig. 2.138 Multinodular and vacuolating neuronal tumour. A Neoplastic neuronal elements manifest nuclear OLIG2 immunoreactivity. B Neoplastic neurons exhibit nuclear and
,_

cytoplasmic HuC/HuD immunoreactiv1ty. C The neurons of this example are synaptophysin-immunoreactive (but most only weakly). D Ramified, CD34-immunoreactive neural
elements are present in this example.

144 Gl1omas . gl1or 1euror1al turn our ~ . and neuronal tur 11ours
Box2.27 Diagnostic criteria for multinodular and vacuolating neuronal tumour
Essential:
Multinodularity
AND
lmmunophenotype
Tumour cells consistently express OLIG2, HuC/HuO, non-
phosphorylated 200 -kDa NFP, and doublecortin on immunohis-
tochemistry, and they often express MAP2 and synaptophysin
(although weakly, with much less labelling of the nodular matrix
I
~

Neuronal cytological features or tumour cell lmmunoreactivity for synaptophysln


HuCIHuD, or non-phosphorylated 200-kDa NFP ' than the cortex) [1383 ,2192,3179,581) . Strong matrix labelling
AND for a-internexin is present (2192,3179), but tumour cells do not
express phosphorylated NFPs or chromogranin, and they are
Absence of mitotic activity
negative or only weakly reactive for NeuN . Thus , they appear
AND to have an incom pletely matured neuronal immunophenotype.
Tumour cell I matrix vacuolation (but may be minimal) CD34 expression by ramified neural elements in associated
Desirable: cortex is common . GFAP labelling is restricted to reactive astro-
lmmunoreactivity for OLIG2 and intemexin A cytes .
Absence of NeuN or chromogranin expression
MAPK pathway-activating abnormalities Cytology
The identification of MVNTs requ ires examination of tissue sec-
tions. A characteristic profile in smear or squash preparations
has not been described .
Histopathology
MVNTs are typically composed of pale, hypomyelinated nod- Diagnostic molecular pathology
ules containing monomorph ic neuronal elements that are most MVNTs harbour MAPK pathway- activating abnormalities that
frequ ently of intermed iate to large size (but generally not achiev- most commonly involve small indels and hotspot mutations in
ing ganglion cell proportions) with round and vesicular nuclei , MAP2K1, followed by pathogenic mutations in BRAF (excluding
distinct nuc leoli, and amphophilic to eosinophilic cytoplasm the common p.V600E mutations, wh ich have not been docu-
{1383, 307, 2192,3179}. These lie in a delicate fibrillar matrix that mented in this tumour to date) and FGFR2 fusions {2443,581 ).
usually exhi bits conspicuous vacuolar change, with tumour
cells often displaying cytoplasmic/pericellular vacuolization as Essential and desirable diagnostic criteria
well. A more diffuse distribution of neurons producing a band- See Box 2.27.
like or gyriform expansion of the cortex or hippocampus may
be evident, and some examples comprise small neuronal ele- Staging
ments. Tumour ce lls are haphazardly distributed within nodules Not clinically relevant
and some may align along capillary vessels. In select cases,
small, oli godendrocyte-like cells are intermingled or clustered . Prognosis and prediction
Conspicuously dysmorphic and multinucleated neuronal forms MVNTs are benign . Disease progression or recurrence have
are typically absent, as are Rosenthal fibres , eosinophilic not been described after gross total excision, with subtotally
granular bod ies, myxoid microcysts and microcalcifications, resected tumours remaining stable (1383,3179,581}. The non-
althou gh some tumours with MVNT-type and gangliogliomatous progressive nature of lesions exhibiting the characteristic neu-
components have been described {3179,2443) . Mitotic figures , roradiological features of MVNTs has prompted the suggestion
vascular proliferation, and necrosis have not been identified. that these can be safely followed without recourse to biopsy
MVNTs can be associated with reg ional cortical disorganization {2292 ,88) .
{1383) and hippocampal sclerosis (442) .

G li mias. glion uronal tumours , ancJ neu1 onnl tu1 :1uu1 '-i 14 5
Dysplastic cerebellar gangliocytoma Hawkins CE
Blumcke I
(Lhermitte- Duclos disease) Eberhart CG
Eng CE
Park SH

Definition
Dysplastic cerebellar gangl iocytoma (Lhermitte- Duclos dis-
ease) is a cerebellar mass composed of dysplastic ganglion
cells that conform to the existing cortical architecture and
thicken the cerebellar fol ia (CNS WHO grade 1).

ICD-0 coding
9493/0 Dysplastic cerebellar gangliocytoma (Lherm itte-Duclos
disease)

ICD-11 coding
2A00 .21 & XH6KOO Mixed neuronal-glial tumours & Dysplastic
gangliocytoma of cerebellum (Lhermitte-Duclos)

Related terminology
Not recommended: cerebellar granule cell hypertrophy; diffuse
hypertrophy of the cerebellar cortex ; gangliomatosis of the
cerebellum .

Subtype(s)
None

Localization
The tumour develops in the cerebellum , usually unil aterally
(without preference for side) {1240,2758). Rarely, bilateral
tumours have been reported (1602,328).

Clinical features Fig.2.139 Dysplastic cerebellar gangliocytoma. A T2-weighted MRI of a 37-year-cic


Dysplastic cerebellar gangliocytoma, or Lhermitte-Duclos dis- woman shows a mass of 56 mm in diameter with the typical tiger-stripe pattern of aiter·
ease, was first described in 1920 (1863 ,3012). Patients with dys- nating Inner hyperintense and outer hypointense layers in the left cerebellum. B D:·
plastic cerebellar gangliocytoma most commonly present with fusion-weighted MRI reveals hyperintensity within the T2-hyperintense area of the 1,-
dysmetria or other cerebellar signs, and/or signs and symptoms mour. C,D Precontrast (C) and postcontrast (0) T1-weighted MRI shows the vascuia:r
pattern of enhancement between the folla .
of obstructive hydrocephalus and increased intracran ial pres-
sure. Cranial nerve deficits, macrocephaly, and seizures are
also often present. Variable periods of preoperative symptoms
have been reported , with a mean duration of approximately in adults . However, patients as young as 3 years and as 0 10
40 months {3331). as in the eighth decade of life have been reported (849,1924
3609) . PTENmutations have been identified in virtually all cases
Imaging of adult-onset dysplastic cerebellar gangl iocytoma but nor 10
Neuroradiological studies demonstrate distorted architecture childhood-onset cases (3609), suggesting that the two differ 1r
in the affected cerebellar hemisphere, with enlarged cerebellar their biology.
folia and cystic changes in some cases . MRI is particularly sen- Dysplastic cerebellar gangliocytoma is a component 01
sitive in depicting the enlarged folia, with alternating T1-hypoin- Cowden syndrome. The single most comprehensive clinica1
tense and T2-hyperintense tiger-stripe striations {1112,2111 , epidemiological study estimated the prevalence of Cowden
3408). The lesions typically do not enhance. Infiltrating medul- syndrome to be 1 case per 1 million person-years (3019 1 Hm'
loblastomas may mimic dysplastic cerebellar gangliocytoma on ever, after the identification of the gene for Cowd en syndromd
imaging 1784,2129) . 11889). a molecular-based estimate of prevalen c e in the same
population was 1 case per 200 000 population 12227). Becall::>t:
Epidemiology of difficulties in recognizing this syndrome , preval ence f1gurt' -
Because of the rarity of dysplastic cerebellar gangliocytoma, are likely to be underestimates. In one study of 2 11 patients w1tr
there has not been a systematic study to determine the distribu - Cowden syndrome, 32% developed dysplastic cerebellar g.:u -
tion of patient age at onset , but most cases have been identified gliocytoma 12672).

146 Gl1ornas, gl1oneurona l tumour..; , and neuronal tumours


I
'

Etiology and the absence of progression support classification as hamar-


I
Dysplastic cerebe llar gangl iocytoma is a component of toma. However, recurrent growth has occasionally been noted ,
Cowden syndrome, an autosomal dominant disorder charac- and dysplastic gangliocytomas can develop in ad ult patien ts
terized by multiple hamartomas involving tissues derived from with previously normal MRI findings {11 ,1218,2006). It has been
all th ree germ ce ll layers {3125 ). Approximately 85% of patients suggested that the primary cell of origin Is the cerebel lar gran -
with Cowden syndrome have a germline mutation in PTEN (also ule neuron \1218), and that a combination of aberrant migration
called "PTEN hamartoma syndrome"), including intragenic and hypertrophy of granule cells is responsible for formation of
mutati ons , promoter mutations, and large deletions/rearrange- the lesions {11 ). Murine transgenic models with localized PTEN
ments {1889,2018 ,3610 }. Patients without PTEN mutations may loss support this hypothesis \1776) .
have germ line variants of SOHB or SOHO, both of wh ich have
been shown to affect the same downstream signalling path- Macroscopic appearance
ways as PTEN \2250 }. Other alterations identified as predispos- The affected cerebellum displays a discrete reg ion of hypertro-
ing to non-PTEN mutation-positive Cowden and Cowden-like phy and a coarse gyral pattern that extends into deeper laye rs.
syndromes include SEC23B, USF3, KLLN, and WWP1 variants
{3533). WWP1 is an E3-ubiquitin ligase, and gain-of-function Histopathology
germline mutations increase ubiqu itination and degradation of Dysplastic cerebellar gangliocytoma causes diffuse enlarge-
PTEN, mimicking germl ine PTEN mutations {1843}. ment of the molecular and internal granular layers of the cer-
ebellum , which are filled by ganglionic cells of various sizes \ 11).
Pathogenesis An important diagnostic feature is the relative preservation of
It remains unclear wh ether dysplastic cerebellar gangliocytoma the cerebellar architecture; the folia are enlarged and distorted
is hamartomatous or neoplastic in nature. Malformative histo- but not obliterated . A layer of abnormally myelinated axon bun-
pathological fe atures , very low or absent proliferative activity, dles in parallel arrays is often observed in the outer molecular

,_ • .... . ). ~... '


n t , ~ -

fig. 2.141 Dysplastic cerebellar gangliocytoma (Lr1errn1tte-Duclos disease) A NeuN Hnmunoh1stochem1stry reveals the abnormal arrangement ol dysplast1c ganglion cel ls,
which are most densely present in the cerebellar molecular layer. B Some of the dysplast1c ganglion cells are Hnmunoreact1ve for phosphorylated mTOR.

Cl1or nas g l1or ur on al tumuur ~ . dr 1d , lt'Ur u 11, 11 tun·~·· :1 .. 1-F


eox 2.28 Diagnostic criteria for dysplastic cerebellar gangliocytoma (Lhermitte- Du- that only a small pro portion of neurons are derived frr)rr :::
clos disease) Purki nje cell source 11218,2919). lmmunohistochem1str f 81·,r,
Essential: demonstrate s loss of PTEN protein expression in most dysol;;-,
Gangllocytic lesion enlarging cerebellar folia tic cells and increased expression of phosphorylated AKT 3r~
AND 86 , reflecting aberrant sig nalling that is predicted to resul! -
Densely packed ganglionic cells of various sizes increased cell size and lack of apoptosis (11 ,3609) Undeti:;r,;.
able or very low proliferative activity has been reported in tre;
AND
few cases analysed with proliferation markers (11 ,1218}
Matrix resembling normal neuropil, sometimes more coarsely fibrillar or vacuolated
Desirable: Cytology
PTEN mutation/deletion or loss of expression Not clinically relevant
Abnormal myelination and vacuolization in the outer molecular layer
Calcification and ectatic vessels Diagnostic molecular pathology
See Pathogenesis, above.

layer. Scattered cells morpholog ically consistent with granule Essential and desirable diagnostic criteria
neurons are sometimes found under the pia or in the molecular See Box 2.28 .
layer. The resul ting structure of these dysmorphic cerebellar
foli a has been referred to as inverted cerebellar cortex. Purkinje Staging
cells are reduced in number or absent. Calcification and ectatic Not clinically relevant
vessels are commonly present with in the lesion . Vacuoles are
someti mes observed in the molecular layer and white matter Prognosis and prediction
111}. Although several recurrent dysplastic cerebe llar gangliocyto-
mas have been reported , most patients are c ured by surgery.
lmmunophenotype and no clear prognostic or predictive factors have emerged
The dysplastic neuronal cells are immunopositive for synapto- Because cerebellar lesion s may develop before the appear-
physin. Antibodies specific to the Purkinje cell anti gens CD3 ance of other features of Cowden syndrome. patients with
(LEU4), PCP2 , PCP4, and calbindin have been found to label dysplastic cerebellar gangliocytoma should be mon itored for
a minor subpopulation of large atypical gan glion cells , but not the deve lopment of add itional malignant and benig n tumours.
to react with the majority of the neuronal elements , suggesti ng including breast and thyroid cancers .

148 Gl1omas. glioneuronal tumou rs, and neuronal tumours


.J
Centi,al neurocytoma Park SH
Giangaspero F
Honavar M
Sievers P

Definition followed by combined extension into the lateral and third ven-
Central neurocytoma is an intraventricular neuroepithel ial tricles , and then by a bilateral intraventncular location . Central
tumour composed of un iform round cells with a neuronal immu- neurocytoma is usually attached to the septum pelluc1dum near
nophenotype and low proliferation index (CNS WHO grade 2). the foramen of Monro (1845 ,2585 ,3376}

ICD-0 coding Clinical features


9506/1 Central neurocytoma Most patients present with symptoms of increased intracranial
pressure, rather than with a distinct neurological deficit The
ICD-1 1 coding clinical history is short (median : 1.7-3 months) (1845} .
2A00 .3 & XHOC11 Central neurocytoma of brain & Central neu-
rocytoma Imaging
On CT, the masses are usually mixed solid and cystic (isodense/
Related terminology hyperdense). Calcifications may be seen (778} . On MRI , central
None neurocytomas are T1-isointense to the brain and have a soap-
bubble (multicystic) appearance on T2-weighted images . They
Subtype(s) often exhibit FLAIR hyperintensity, with a well-defined margin .
None In all cases , heterogeneous enhancemen t after gadolinium
injection is observed , and the tumour may show vascular flow
Localization voids. Haemorrhage may be seen (778 ,2257,3376) . An inverted
Central neurocytomas are typically located supratentorially in alanine peak and a notable glycine peak on proton magnetic
the lateral ventricle(s) and/or the third ventricle. The most com- resonance spectroscopy are useful in the differential diagnosis
mon site is the anterior portion of one of the lateral ventric les . of intraventricular neoplasms 1778,1988).

Fig. 2.142 Central neurocytoma. A T2-we1ghted MRI shows a mass of 52 rnrn 111diameter111 the left lateral ventri cle, mult1focal cysti c portions can be seen B T1-weighted MRI
reveals a soap bubble-like multicystic appearance. The tumour is attached to the septum pelluc1dum C The solid portions of the mass lesion reveal heterogeneou s enhance ·
rnent. D Diffusion-weighted MRI demonstrates diffusion rest11cl1on in the sol1dporMns ot the tumour, suggestive of high cellular1ty. E This mass has high ce rebral blood volume
suggestive of hypervascularity. f CT reveals multifocal hyperattenuat1ng calc1f1cat1ons w1tll111 the tumour.

Fig. 2.143 Central neurocytoma. A Typical central neurocytoma shows a sheet of monotonous cells with round nuclei and salt-and-pepper chromatin patterns. B Some t1 m~s
cytoplasmic clearing is prominent. C Dilated and hyalinized blood vessels are sometimes prominent D Numerous psammomatous calcifications may be present.

Spread Genetic profile


Craniospinal dissemination is exceptional and may occur in The exact molecular features of th is tumour are not known c
atypical central neurocytomas {851 ,3213 ,3017,2222) . date. Recurrent chromosomal alterations or mutations are
not observed, and most cases show a flat d isomy profile or
Epidemiology copy-number analysis 1463). There was one report of central
In an analysis of> 100 cases, the mean patient age at clinical neurocytomas having numerous DNA copy- number alterations
manifestation was 28.5 years; 46% of patients were diagnosed (1726). including MYCN gain . Another transcriptomic study
in the third decade of life and 70% were diagnosed between the showed overexpression of genes involved in the WNT signalling
ages of 20 and 40 years . Patient ages reported in the literature pathway, calcium function , and maintenance of neural progeni-
range from 8 days to 82 years, although paediatric cases are tors {3295) . Central neurocytomas have not been reported to
rare . Both sexes are equally affected , with an M:F ratio of 1.02:1. exhibit 1p/19q codeletion .
Population-based incidence rates for central neurocytoma are
not available. In large surgical series, central neurocytomas Methylation profile
account for 0 .25-0 .5% of all intracranial tumours (1262}. The methylation class of central neurocytomas exclusively corT'-
prises tumours with the histological diagnosi s of central neuro-
Etiology cytoma . A distinction between central neurocytoma and atypi-
Unknown cal central neurocytoma is currently not possible on the basis 01
methylation clustering (463}.
Pathogenesis
Cell of origin Macroscopic appearance
The cellular origin of central neurocytoma is unknown . Evidence Grossly, central neurocytoma is greyish and friable . Calcifica-
of both glial and neuronal differentiation in some tumours sug- tions and haemorrhage can be present.
gests an origin from neuroglial precursor cells with the potential-
ity of dual differentiation {1419,2365 ,1695). Central neurocytoma Hlstopathology
could originate from the subependymal plate of the lateral ven- Central neurocytoma i.s a neuroepith elial tumour composed or
tricles 13345). However, an origin from circumventricular organs uniform round cells w.1th a round ed nucleus with finely speck-
has also been proposed 11504). led chromatin and variably presen t nucleoli . Additional featur~s
include ,fibnllary areas min:icking neu ropil , an oligodendrc
gl1oma-l1ke. honeycomb architecture, large fibnllary areas rn 1111·
1ck1ng the irregular rosettes in pineocytomas, cells arrangeu 1r

150 Gl1omas , gl1on uronal tumours . and neuronal tumou rs


I

Fig. 2.144 Central neurocytoma. lmmunohistochemically, the tumour cell nuclei in this particular case are robustly positive for NeuN (A) and for transcription factor 1 (TTF1 ) (B).
The cytoplasm of the tumour cells is positive for synaptophysin (C) and for L1CAM (D).

straight lines, and perivascular pseudorosettes as observed index for predicting prognosis is still under debate [1408.429 ,
in ependymomas. Capillary blood vessels, usually arranged 2592,2998}.
1n an arborizing pattern, give the tumours a neuroendocrine
appearance. Calcifications are seen in half of all cases , usu- Electron microscopy
ally distributed throughout the tumour. Occasionally, lipomatous Electron microscopy shows regular round nuclei with finely
changes can be observed {1054,3500,3498). Rarer findings dispersed chromatin and a small , distinct nucleolus in a few
include Homer Wright rosettes and ganglioid cells {2684,3344) . cells . The cytoplasm contains mitochondria, a prominent Golgi
Some cases show increased vascularity with substantial intra- complex , and some cisternae of rough endoplasmic reticulum,
tumou ral haemorrhage (5-10% of cases) , and early organizing often arranged in concentric lamellae. Numerous intermingled
haematoma can be present, which results in heterogeneity on cell processes containing microtubules and dense-core or
T2-weighted images {3379) . In rare instances, anaplastic histo- clear vesicles are always observed {496,1262). Well-formed or
logical features (i .e. brisk mitotic activity, microvascular prolif- abnormal synapses may be present, but they are not required
eration, and necrosis) can occur in combination, and tumours for the diagnosis.
with these features are called atypical central neurocytomas
(1 262,3344,3531 ). lmmunophenotype
Synaptophysin expression is the most suitable and reliable
Grading diagnostic marker, with immunoreactivity diffusely present in
Central neurocytoma corresponds histologically to CNS WHO the tumour matrix, especially in fibrillary zones and perivascular
grade 2. Tumours usually show a favourable behaviour, but nucleus-free cuffs {930,1262). Most cases are also immunore-
some recur, even after total surgical removal. Moreover, several active for NeuN , although the intensity and extent of the label-
studies have shown increased aggressiveness in cases of atyp- ling vary {2997,3296) . Thyroid transcription factor 1 (TTF1 ; clone
ical features and/or a Ki-67 (MIB1) proliferation index > 2-3% SPT24) may also be positive in the tumour cell nuclei [1740) .
{1408,429,2592,2998). Other neuronal epitopes (e.g. class Ill p-tubulin and MAP2) are
usually expressed. In addition, UCAM can be positive in the
Proliferation tumour cel ls. In contrast, expression of chromogranin A, NFP,
Mitoses are exceptional, and the Ki -67 (MIB1) proliferation and a-internexin is absent. except in sporadic cases showing
index is usually low(< 2%) . However, cases with a Ki-67 (MIB1) gangliocytic differentiation . Although most studies have found
proliferation index as high as approximately 10% have been GFAP to be expressed only in entrapped reactive astrocytes .
reported . The Ki -67 (MIB1) proliferation index is considered a this antigen has been reported in tumour cells by some autho1 s
powerful prognostic marker, but the optimal threshold of thi s {2998 ,3233 ,3344,3345). OLIG2 is usually positive in occasional

Gl1orm:is, glioneuront:.11 tumours , ancJ neuronal tu11 1nu 1 ~ 151


Box2.29 Diagnostic criteria for central neurocytoma Essential and desirable diagnostic criteria
See Box 2.29.
Essential:
lntraventricular localization Staging
AND Not clin ically relevant
Oligodendroglioma-like monomorphic cells
AND Prognosis and prediction
Synaptophysin expression The clinical course of central neurocytoma is usually favourac1e
with the 5-year and 10-year overall survival rates estimated to ce
AND (for unresolved lesions)
96% and 82%, respectively 11408}. However, malignant bena.-
Methylation profile of central neurocytoma
iour with craniospinal dissemination has been reported 1 85 ~
Desirable: 3213 ,2222,3017). The extent of resection is an important prcg-
Young adult patient nostic factor. Specifically, a pooled analysis of > 400 pat1er: :~
In most cases, no sign of malignancy has demonstrated that gross total resection is superior to subr'.>
tal resection 12591). Nevertheless, recent literature has not tm,r-::
statistically significant differences in overall survival betwet"
cells in central neurocytoma (1892). However, a strong and dif- gross total and subtotal resection 11408,429}. Although grcss
fuse OLIG2 immunoreactivity would favour a diagnosis of intra- total resection surgery should always be attempted. this proc2-
ventricular oligodendroglioma {3489}. dure is not often feasible , because of the distinct location of trc
intraventricular area and vital neural structures in the pem21'-
Cytology tricular region {429}. It is now well established that a higher K:-c-
lntraoperative squash preparation shows a monotonous sheet ~MIB1) labelling index is associated with more aggressive beha\·
of round cells with rounded nuclei and salt-and-pepper chroma- 1our. A recent study reported a 2-year progression-free sur. 11,:;
tin without cell aggregation or clustering , resembling the smear rate of 48% for cases with a Ki-67 (MIB1) labelling index> 4°c . ~
pattern of pituitary adenoma 12899). contrast to 90% for those with a value s 4% 11408). However rr2
optimal threshold of this index is stil l a matter of debate l ...ic6
Diagnostic molecular pathology 429,2592,2998). Regardless of the threshold there is ev1cienCt
See Genetic profile under Pathogenesis, above. that adjuvant therapy should be considered for tumours ~''~("
higher Ki-67 (MIB1) index values (1408} .

152 GliomC:ts, gl1oneuronal tumours, and neuronal tumours


Extraventricular neurocytoma Sievers P
Giangaspero F

I
Honavar M
Park SH
Soffietti R

Definition Localization
Extraventricular neurocytoma is a usually well-circumscribed Extraventricular neurocytomas can arise in almost any locati on
neuronal neoplasm that arises throughout the CNS outside in the CNS without contact with the ventricular system . The most
the ventricular system , with histopathological characteristics frequently documented locations are the cerebral hemisphere s
resembling those of central neurocytoma but demonstrat- and cerebellum (3205 ,363 ,2935) . However, there are also
ing a much wider morphological spectrum, and with frequent reports of these tumours arising in the spinal cord , thalamus ,
FGFR1::TACC1 fusions (CNS WHO grade 2). hypothalamic region, and pons, with single cases reported in
cranial nerves, the cauda equina, and even in the sellar region
ICD-0 coding {1015,3091 J, although these cases were reported in the pre-
9506/1 Extraventricular neurocytoma genetics era.

ICD-11 coding Clinical features


2A00 .3 & XH2HS1 Central neurocytoma of brain & Extraven- The clinical manifestation of extraventricular neurocytoma varies
tricular neurocytoma according to the multiple locations described for these tumours
and whether the tumour exerts a mass effect. Patients may
Related terminology present with headache, seizures, visual disturbances , hemi-
None paresis , and cognitive disturbances , but they can also display
motor, sensory, and sphincter dysfunction, as well as epilepsy
Subtype(s) (363,1015,3091 ,3499,3590}.
None

l.111nm;1s , Jlioneur nal lurnour~; . :1rili ri1.: tiiu 11 .i1 tt1rn111.r 15:1
I ' _.;

fig.2.147 Extraventricular neurocytoma. A Diffuse cytoplasmic immunoreactivity for synaptophysin is characteristic of extraventricular neurocytoma. B Variable proportions
of GFAP-positive cells may be present in some tumours.

Imaging Macroscopic appearance


Extraventricu lar neurocytomas exhibit a wide and variable imag- Extraventricular neurocytomas are usually we ll c ircumscribed
ing spectrum. They usually appear as large, well-demarcated, sometimes with a cyst-mural nodule configuration , but they can
solitary, cystic-sol id masses that are frequently associated with occasionally be infiltrative.
moderate peritumoural oedema and calcifications {363 ,1473,
27 13). lntratumoural haemorrhage may be present. On MRI , the Histopathology
solid portion is isointense to hypointense on T1-weighted images Although some histopathological characteristics resemble cen-
and predominantly hyperintense on T2-weighted images. In tral neurocytoma, a much wider morphological spec trum has
most cases, MRI shows heterogeneous contrast enhancement been described in extraventricular neurocytomas. Histolog1-
11473,2713). cally, extraventricular neurocytomas are usually mode rate- to
high-cellularity neoplasms often composed of relatively mono-
Epidemiology morphic oligodendrogl ia-like tumour cells with small , uniform
Extraventricular neurocytomas are rare, with an annual incidence nuclei and sparse cytoplasm, frequently with clear perinuclear
rate in the USA of approximately 0.01 cases per 100 000 popu- haloes. Tumour cells are often arranged in sheets or cell clus-
lation (3205). They generally affect patients of any age, with ters with neuropil islands; less frequent patterns include neuro-
peak incidence in the third to fourth decades of life. There is no cytic rosettes or ribbons . A subset of tumours contain ganglion
significant sex predilection (3205, 2935 ,363) . or ganglioid cells . Microcalcifications and hyal inized vessels
may be present (1084,363,2935). A rare case w ith li pomatous
Etiology changes has been reported {436) . Mitotic activity is usually low
The etiology of extraventricular neurocytoma is unknown . No risk More recently, molecular investigations of extraventric ular neu-
factors or inherited genetic susceptibility have been reported to rocytomas revealed a high rate of histolog ically misinterpretea
date. cases and underlined the wide range of morpholog ical hetero-
geneity in this entity 12935).
Pathogenesis
Cell of origin Grading
The cellular origin of extraventricular neurocytoma is unknown . The majority of extraventricular neurocytomas manifest histo-
The fact that extraventricular neurocytomas form a distinct logically as low-grade tumours and correspo nd to CNS WHO
molecular group suggests that these tumours may arise from a grade 2. However, a subset of tumours (lackin g genetic analy-
specific precursor cell population {2935). ses) have been reported to show histopathological features
of atypia or anaplasia, with increased mitotic and proliferative
Genetic profile activity as well as microvascular prol iferatio n and/or necrosis,
The most frequent genetic alterations are FGFR1 ::TACC1 suggestive of a more aggressive clinical behaviour [ 1540,3563.
fusions. which were found in 60% of cases in one large series of 10151.
tumours identified by DNA methylation profiling . Less common
alterations include other FGFR gene fusions 12935). Neither lmmunophenotype
IDH gene mutations nor MGMT promoter methylation has been Diffu~e .cytoplasmic immunoreactivity for synaptophys1n is char-
reported to date {2187] . acteristic of extraventricular neurocytoma and is present 1n the
t~mour cells and neuropil islands [363 ,2 187,2935) . Chromogra
n1n A and NeuN labelling in some cases confirms neuronal d1r
ferentiation, but it might be very focal. In nearly half of the cast's.

154 Gliornas. gl1oneuronal tumou rs, a11d neuronal tumou rs


immunopositivity for GFAP has been reported 1363). Expres- Box 2.30 Diagnostic criteria for extraventricular neurocytoma
sion of OLIG2 has been observed in a subset of extraventricular
Essential:

I
neurocytoma, but it is more typically negative in the neoplastic
Extraventricular neurocytic neoplasm without IDH alteration
cells 12315,2187}. lmmunohistochemistry for IDH1 p.R132H Is
negative {26,3535,461). The Ki-67 proliferation index is usually AND
low (1-3%), but it can be elevated in a subset of cases {1540, Synaptophysin expression
363,2935). AND (for unresolved lesions)
Methylation profile of extraventricular neurocytoma
Differential diagnosis
Desirable:
Differential diagnostic considerations usually include oligoden-
FGFR1 alteration (mostly FGFR1 :: TACC1 fusion)
droglioma and other glioneuronal and neuronal tumours . The
diffuse cytoplasmic immunoreactivity for synaptophysin in the Caveat: Diagnosis should be heavily weighted towards molecular findings because
morphological analyses frequently result in mistyping {2935}.
absence of IDH mutation and 1p/19q codeletion distinguishes
extraventricular neurocytoma from oligodendroglioma. Central
neurocytomas can typically be distinguished by their intraven- additional molecular analyses are strongly advised for a precise
tricular localization . Because of a wide overlap with various diagnosis of extraventricular neurocytoma.
glioneuronal entities , additional molecular analyses are highly
rec ommended in cases with diagnostic uncertainty. Essential and desirable diagnostic criteria
See Box 2.30.
Cytology
Not clinically relevant Staging
Not clinically relevant
Diagnostic molecular pathology
Genome-wide DNA methylation profiling has provided evidence Prognosis and prediction
for a specific epigenetic signature of extraventricular neurocy- Extraventricular neurocytoma is generally a low-grade tumour
toma that is clearly distinct from that of other CNS tumours; it with a usually favourable prognosis {2935} . Gross total resection
has also indicated that many cases diagnosed histologically has been associated with a low rate of recurrence and good
(without additional molecular analysis) may represent misdi- seizure control {1540,363 ,1015). However, reports on the clinical
agnoses . FGFR1 :: TACC1 fusion is a highly frequent event in courses and outcomes of these tumours vary considerably, not
molecularly defined extraventricular neurocytoma , in addition least because of a wide overlap with other entities and possible
to a small number of other FGFR alterations {2935}. Therefore, misdiagnosis in some cases .

Gliomas, glioneuronal tumours, and neuronal tumours 155


Cerebellar liponeurocytoma Glangaspero F
Ch imell i L
Park SH
Sievers P

Definition Clinical features


Cerebellar liponeurocytoma is a cerebellar neoplasm with Headache and other symptoms and si g ns of raised 1ntrar,r';
advanced neuronal or neurocytic differentiation. variable glial nial pressure (either from the lesion itself or due to obstruct1.•;
differentiation , and focal lipoma-like changes (CNS WHO hydrocephalus) are common presentat ions Cerebellar s1q1v,
grade 2). 1nclud1ng ataxia and disturbed gait. are also c ommon 123521

ICD-0 coding Imaging


9506/1 Cerebellar liponeurocytoma On CT, the tumour is variably isodense or hypodense . N•:r
focal areas of marked hypoattenuation correspondi ng to h,g~
ICD-11 coding fat density (77,2260 ,1054). On T1 -weighted MRI , the tumour -;
2A00.3 & XH2GBO Central neurocytoma of brain & Cerebellar isointense to hypo1ntense, with patchy areas of hyperinteris 1,
liponeurocytoma corresponding to regions of high lipid content. Enhancerrir:;-·
with gadolinium is usually heterogeneous , with areas of turno1,·
Related terminology showing variable degrees of enhancement . On T2-we1ghte.:
Not recommended: lipomatous me dulloblastoma; lipidized an d FLAIR imaging, the tumour 1s hyperintense to the adj ace ~·
medulloblastoma; neurolipocytoma; medullocytoma; lipo- brain (105 4). Associated oedema is minimal or absent (48) F'.l'-
matous glioneurocytoma; li pidized mature neuroectoderma l suppressed images may be helpful in supporting a preope ~­
tumour of the cere bellum . tive diagnosis (77).

Subtype(s) Epidemiology
None More than 60 cases of cerebellar li poneurocytoma have been
reported in the English-languag e literature (2260 .1054,1606
Localization 738) . The mean patien t age is 50 years (range: 24-77 years1
Cerebellar liponeurocytoma most commonly Involves the cer- with peak incidence in the third to sixth decades of life . There 1s
ebellar hemispheres, but it can also be located in the paramed- no significant sex predilection (1346,2417} . Familial pred1spos:-
ian reg ion or verm is and extend to the cerebellopontine angle tion has been reported [2513 ,3466).
or fourth ventricle 12260,1054,1606,570}. Recent reports (1054,
3500,3498,436) have described a series of cases of liponeu- Etiology
roc ytoma-like tumours occurring in the cerebellum and in Unknown
supratentorial locations , causing confusion between bona fide
cerebellar liponeurocytoma and central or extraventricular neu- Pathogenesis
rocytoma with llpomatous changes . There are also reports of Cell of origin
multifocal tumours: a principal lesion with a satellite lesion in One study demonstrated that the transcr iption factor NGN ·1. bu
the opposite hemisphere (2447,1606), multiple bilateral lesions not ATO H1, is expressed in cerebe ll ar li poneurocytoma (unlike
/2875 ,2947). and even a case associated with leptomeningeal in normal adult cerebellum) and that ad ipocyte fatty acic-
spinal cord nodules at presentation (1699) . binding protein , typically found in adip ocytes , is s1gnificant1'1

flg. 2,148 Cerebellar liponeurocytoma. A r 2-weighted MRI depicts the mass as well circumscribed and mildly heterogeneous,_wlth small cystic areas medially 8 Inher ent•)
bright signal on T1-weighted pre-contrast images corresponding to lipid w1th1n the t.umour. C Postgadolin1um T1 -we1ghted imaging wuh fat saturation depicts mild diffuse ;:1;
hancement of the mass, while the bright signal in fat-nch areas 1s lost with fat saturation.

156 Gl1omas . glioneu ronal tumours . and neurona l tumou rs


Fig. 2.149 Cerebellar liponeurocytoma. A Classic histology showing neurocytes and lipidized tumour cells. B Intense immunoreactivity of neurocytes for synaptophy-
sin. C Uponeurocytomas also express NeuN. D GFAP-positive neoplastic cells can be present.

overexpressed in cerebellar liponeurocytoma compared with H istopathology


both normal adult cerebellum and human medulloblastoma. Cerebellar liponeurocytoma is composed of a uniform popula-
These findings suggest an origin of cerebellar liponeurocytoma tion of small neurocytic cells arranged in sheets and lobules
from cerebellar progenitors, which are distinct from cerebellar and with regular round to oval nuclei, clear cytoplasm , and
granule progenitors and aberrantly differentiate into adipocyte- poorly defined cell membranes . The histological hallmark of this
like tumour cells {115). entity is focal accumulation of lipid-laden cells that resemble
adipocytes but constitute lipid accumulation in neuroepithelial
Genetic profile tumour cells . Features of anaplasia, such as nuclear atypia,
Genetic analyses indicate that this lesion is a rare but dis- necrosis, and microvascular proliferation , are typically absent
tinct clinicopathological entity {1346,2260,2999,1085,463). in primary lesions, but they may be found in recurrent tumours
Genome-wide DNA methylation analysis of cerebellar liponeu- {1085 ,2594) . The lipidized component may be markedly
rocytomas has shown that they are molecularly distinct and reduced or even absent (2594} in recurrent lesions. However,
characterized by recurrent focal losses of chromosomes 14 and some tumour recurrences lack these atypical histopathological
2p 1463). These findings are supported by gene expression features {1467} .
data indicating that cerebellar liponeurocytomas have profiles
more similar to those of central neurocytoma than to those of Grading
medulloblastoma, and that they lack isochromosome 17q and Cerebellar liponeurocytoma corresponds histologically to CNS
mutations of PTCH1, CTNNB1, and APC {1346), which are seen WHO grade 2.
in a subset of medulloblastomas. TP53 missense mutations
were reported in 4 of 20 cases, which is a higher frequency than Proliferation
in medulloblastoma, and this is not an alteration typically seen The growth fraction as determined by the Ki-67 (MIB1) prolifera-
in central neurocytoma {1346) . Although single reports suggest tion index is usually in the range of 1-4%, but it can be as high
a possible inheritable predisposition (2513 ,3466}, no specific as 10% in cases of tumour recurrence (1054) .
genetic susceptibility is known . Cerebellar liponeurocytoma dis-
played a unique epigenetic signature (460) . Electron microscopy
Electron microscopy shows dense-core and clear vesi-
Macroscopic appearance cles, microtubule-containing neurites, and (occasJOnally)
Not reported

G l1ornas, gl1oneuronal t uniuur~;, ctiitl flt'Llf011dl tu111 l 'u1 s 157


, 2

oe

~--- -- -- - - - - --------__,...-.- ----


' ;

-O B

-12

~ ~ ~ ~ ~ ~ ~ ~ ~ ~ ~ ~n g ~
Ag.2.150 Cerebellar liponeurocytoma. Copy-number analysis showing focal losses of chromosomes 14p and 2p.

synapse-like structures associated with non-membrane-bound Box2.31 Diagnostic criteria for cerebellar liponeurocytoma
lipid vacuoles of variable size 11529,1011,1085). Essential:
Cerebellar localization
lmmunophenotype AND
lmmunohistochemically, there is consistent expression of neu-
Ollgodendroglioma-like monomorphic cells associated with focal llpoma-like changes
ronal markers, including synaptophysin, NeuN , and MAP2.
AND
Focal GFAP expression by tumour cells , which indicates astro-
cytic differentiation , is observed in most cases [1054 ,2999). Synaptophysln expression
One report mentioned immunoreactivity for desmin and mor- AND (for unresolved lesions)
phological features of incipient myogenic differentiation 11137). Methylation profile of cerebellar liponeurocytoma
Desirable:
Cytology Adult patient
Not clinically relevant
Focal GFAP immunoreactivity
Absence of histological features of malignancy
Diagnostic molecular pathology
See Genetic profile under Pathogenesis, above .

Essential and desirable diagnostic criteria progression-free survival was 10 years (mean: 8 .6 years)
See Box 2.31 . regardless of treatment {1504 ,77,433,47,1346,1895 ,2594 ,3500 ,
570) . Review of the literature showed that the tumour recur-
Staging rence rate in patients treated by complete tumour resection
Not clinically relevant (with or without adjuvant radiotherapy) was 15% , whereas the
recurrence rate in patients treated by incomplete tumour resec-
Prognosis and prediction tion (with or without radiotherapy) was 42% 11054}. Recurrent
This tumour has a favourable prognosis. Most patients with suf- tumours may show increased mitotic activity (with an increased
ficient follow-up survived for> 5 years, and gross total resection Ki-67 [MIB1] proliferation index as high as 30%), vascular pro-
and postoperative radiotherapy yield survival benefit {47,1467). liferation, and necrosis (1085 ,2594), although some tumour
On meta-analysis, the postoperative 5-year survival rate was recurrences lack these atypical histopatholog ical features
71 .3%, the 5-year progression-free survival rate was 60 .8% , (1467,1504}. All reported recurren ces were confined to the pos-
the mean overall survival was 16.3 years , and the median terior fossa [1433 ,2352,570} .

158 Gl1omas , gl1oneuronal tumours . and neuronal tumou rs


Ependymal tumours: Introduction Ellison DW

Where possible, ependymal tumours should now be classified but mutations in EZHIP and the histone H3 family resulting in H3
I
according to a comb ination of histopathological and molecular p.K28M (K27M), wh ich are of uncertain clinicopathological. sig-
features and anatomical site (836} . DNA methylation profiling ni"ficance, are found at frequencies of 9% and 4% , respectively
distinguishes types of ependymal tumours at different levels of (2373). Two of the molecular groups of spinal ependymomas are
the neuraxis , dividing them into molecular groups across the dominated by tumours with either a classic or a myxopapillary

I
three main anatomical compartments of the CNS: the supraten - morphology (2374). There is also an aggressive type of spinal
torial region, poste rior fossa , and spine (2374} . This pathobio- ependymoma characterized by high-grade histopathological
logical heterogeneity is now central to their classification . features and MYCN amplification (1079}.
One molecular group at each anatomical site consists almost The new WHO classification of ependymal tumours lists two
entirely of tumours with the morphological features of subepend - molecularly defined types of supratentorial ependymoma (with
ymoma (2374) . Of the two remain ing supratentorial molecular ZFTA or YAP1 fusion), two molecularly defin ed types of poste-
groups, one is dominated by ependymomas with fusion genes rior fossa ependymoma (PFA and PFB), and a spinal tumour
involving ZFTA (formerly known as C11orf95) , and the other con- defined by the presence of MYCN amplification . Also listed
tains tumours with a high frequency of fusion genes involving are ependymomas defined by anatomical location but not by
YAP1 (11 3). Methylation profiling divides the majority of poste- molecular alteration . These designations can be used either
rior fossa ependymomas into two main groups, posterior fossa when molecular analysis reveals a different molecular altera-
group A (PFA) and group B (PFB), which are also distinguished by tion to one that defines ependymomas at a particular site (in
global levels of H3 p.K28me3 (K27me3) (2374,2384). Posterior such cases , the term "not elsewhere classified [NEC]" is used)
fossa ependymomas generally lack recurrent mutations (2401 }, or when molecular analysis fails or is unfeasible (in which case

Molecular
Age Sex CNS WHO grade featun1a Outcome

~-
It d'd'd'9
---~
0
~- Atff
20
~ d d'9 2 13 ZFD!a.io...
CDK1QA~-
:I
(/)

At d' 99 9 2/3 YAP1 "'"*"-

I~[ "
£--
d'd'd'9
I 8*tad-

0
I Posterior Iossa e nd moma Group B
PriA At dd'9 2/ 3
EZHIP"""**>o1
H3 pJ<28M (K27M} -
Clv. lql/MJ e
ttlt d' 'l' 2 J3 .......
C"'-11e/

"" d' 'f'


Po!. tu11or fo ~ ..t ~ ubopendy 1 n>rna Cllf.llqdllliOol>

-'*
[ Supratentortal subependymoma ]

--
SP-EP d' d' 9 2/3
.,.. ·1: 1ij
.5
NF2-

"
2 ~

····~ ~
d'

- ~o ,
;I.. SuprJt...nlona l epcndymomJ YAP1

Supral1>ntonal ependymorna . ZFTA (C11orf95) fus1on-posillve 1


fu~.,n-pu~111v c ttlt d' 9 '1l'lt::#I
l.Qlr.111 e
-30 30 60

Rg.2.151 Ependymal tumours: molecular groups. Unsupervised, non-linear !-distribut- Fig. 2.152 Ependymal tumours: molecular groups. Group colours parallel those used
ed stochastic neighbour embedding (t-SNE) projection of methylation array profiles from in the I-distributed stochastJc neighbour embedding (t-SNE) plot. Outcome colour
ependymal tumours. Note that the posterior Iossa group A (PFA) ependymal tumours split code: green, good; yellow, intermediate; red, poor. Chr., chromosome; PFA, poste-
into two subgroups, which might reflect site of origin {2373). Samples were selected from rior fo~sa group A ependymoma; PFB, posterior Iossa group B ependymoma; PF-SE,
alarge database of> 50 000 brain tumour datasets to serve as reference profiles for train- posterior Iossa subependymoma; SP-EP, spinal ependymoma; SP-MP, myxopapil-
ing a supervised classification model based on strict criteria: all these samples showed a lary ependymoma; SP-MYCN, spinal ependymoma, MYCN-amplified; SP-SE, spinal
high calibrated classification score(> 0.9) for ependymoma methylation classes when ap- subependymoma; ST-SE, supratentorial subependymoma; ST-YAP1 , supratentorial
plying the brain tumour classifier available at https://www.molecularneuropathology.org. ependymoma . YAP1 ~usion-positive; ST-ZFTA. supratentorial ependymoma, ZFTA
In a t-SNE projection like this, occasional samples of one subclass may fall closer to (C11orf95) fus1on- pos1tive.
a different subclass because of the stochastic nature of the t-SNE algorithm and the
gradual rather than strict boundaries between the subclasses.

Gllomas, glioneuronal tumours . and neuronal tumours 159


"not otherwise specified (NOS]" is used). Myxopapillary epen- with supratentorial ependymoma remains established prau1r;':
dymoma and subependymoma remain listed as tumour types , (3311 ). The updated classification allows only a h1stoloq1c;:i11 1
like in the previous edition of the classification ; currently, molec- defined diagnosis of "ependymoma" to be made at any of th~
ular classification does not provide added clinlcopathological three anatomical sites; "anaplastic ependymoma" is no longer
utility tor these two tumours (836} . listed 1836}. However, as for other tumours in this edition of th"!
Afford ing no cl inicopathological utility, the papillary, clear cell , classification , a pathologist can still ch oose to assign either
and tanycytic morphological patterns are no longer listed as CNS WHO grade 2 or CNS WHO grade 3 to an ependymoma
subtypes of ependymoma; rather, they are included as patterns according to its histopathological features . In an integrated
in the histopathological description of the classic tumour. Long - diagnosis, CNS WHO grade can be presented in a tier adjacent
standing controversy surrounds the clinicopathological utility to one providing a histopathological diagnosis and another with
of grading ependymal tumours 1840}, although use of CNS molecular alterations and the methodologies used to determini;
WHO grade in the therapeutic stratification of adult patients them (1939}.

160 Gltomas , glioneuror1al tumours, and neuronal tumours


Supratentorial ependymoma Pajtler KW
Aldape KO
Pietsch T
Ruda R

I
Gilbertson RJ Taylor MD
Korshunov A Venneti S

Definition
Supratentorial ependymoma is a circumscribed supratentorial
gltoma that focally demonstrates pseudorosettes or ependymal
rosettes and comprises uniform small cells with round nuclei
embedded in a fibrillary matrix. The diagnosis of supratento-

I
rial ependymoma should be used either when genetic analy-
sis has not detected a pathogenic fusion gene involving ZFTA
(C11orf95) or YAP1 (not elsewhere classified; NEC) or when
such analysis has been unsuccessful or is not feasible (not oth-
erwise specified ; NOS) (CNS WHO grade 2 or 3).

ICD-0 coding
9391/3 Supratentorial ependymoma, NOS
Flg.2.153 Supratentorial ependymoma. A T2-weighted axial MRI showing a large
ICD-11 coding circumscribed mass In the cerebral hemisphere. B T1 -we1ghted gadolinium-enhanced
2AOO.OY & XH1511 Other specified gliomas of brain & Ependy- axial MRI in an adult, showing an intraventricular nodular lesion with heterogeneous
moma, NOS contrast enhancement and solid and cystic components.

Related terminology reflect the increasing frequency of spinal ependymomas with


None age. The overall M:F ratio for supratentorial ependymomas is
1.32:1 {2347}.
Subtype(s)
None Etiology
Radial glia are implicated in the histogenesis of supratentorial
Localization ependymoma {3155). Genetic susceptibility for supratentorial
Supratentorial ependymomas are localized to the cerebral hem- ependymoma specifically has not been reported .
ispheres and may or may not have an obvious connection to the
ventricular system . They occur more frequently in the frontal or Pathogenesis
parietal lobe than in the temporal or occipital lobe {2818,3378). The precise pathogenic mechanisms leading to fusion-negative
supratentorial ependymomas have yet to be elucidated {1002).
Clinical features
Common clinical manifestations include focal neurological defi- Macroscopic appearance
cits or seizures . Headache, nausea, and vomiting may occur Supratentorial ependymomas generally have a tan colour, are
due to raised intracranial pressure and hydrocephalus. Enlarge- soft or spongy, and can exhibit a gritty texture if calcified .
ment of the head or separation of the cranial sutures can be
evident in infants. The interval between initial symptoms and Histopathology
diagnosis is generally$ 6 months {16). The histopathological features of supratentorial ependymo-
mas are quite heterogeneous . Most have a relatively abrupt
Imaging interface with adjacent CNS parenchyma . Diffuse infiltration
On MRI, supratentorial ependymomas generally appear as of adjacent normal tissue is rare and tends to be seen after
masses with irregular contrast enhancement. They often dem- multiple recurrences . Perivascular anucleate zones (pseudor-
onstrate cysts or calcification and, more rarely, haemorrhage osettes) are generally present, but they can be subtle in some
or necrosis . Diffusion-weighted MRI frequently shows reduced examples. True ependymal rosettes are found in only a minor-
signal due to hypercellularity {728). ity of cases . Hyalinization of blood vessels and calcification
are frequent . A vascular pattern seen in supratentorial epen-
Epidemiology dymomas, but not other ependymal tumours, manifests as a
Supratentorial ependymomas of all types affect children and branching network of capillary blood vessels (926) . A clear
adults , and they account for approximately one third of intracra- ce ll phenotype is found more often in supratentorial ependy-
nial ependymomas !3311,2073). The proportion of ependymo- momas than in tumours in the posterior fossa or spinal cord .
rnas arising in the supratentorial compartment decreases with Tumour cell s have round or oval nuclei , which generally show
age: 41% in chi ldren, 27% in adolescents, 12% in young adults, speckled chromatin . Ultrastructurally, ependymal features
and 11% in adults aged > 45 years 1844/; the declining numbers

Gl1um<.s, glloneuron I tumou rs, ·:inl1 l1 , UIL)ll~ll u 1 n,1ur·; 161


Ag.2.154 Supratentorial ependymoma. A A branching network of small blood vessels is a frequent architectural feature of supratentorial ependymomas. Perivascular anuc'e-
ate zones can be subtle. B Ependymal rosettes are present in a minority of cases. C Clear cell change is a frequent cytological feature of supraten torial ependymomas. Nuclear
pleomorphism is mild in most cases, and the number of mitotic figures can be variable. D Nuclear pleomorphism is mild in most cases, but it can be marked in some cases.

(including intracytoplasmic villi , cilia , and complex intercellular Differential diagnosis


zipper-like junctions) are present {3087,1079}. The differential diagnosis of fusion-negative supratentona
ependymomas includes histopatholog ical mimics such as
Grading tumours with BCOR internal tandem dupl ication (ITD) and astrc-
The current classification allows only a histologically defined blastomas . These entities should be excluded by appropriate
diagnosis of "ependymoma" to be made at any of the three immunohistological and molecular analyses {2370,1002).
anatomical sites; "anaplastic ependymoma" is no longer listed
1836). However, supratentorial ependymomas can be assigned Cytology
CNS WHO grade 2 or 3, ideally in the context of an integrated Cytological preparations generally show uniform cells w11h
diagnosis {1939} . High-grade features in ependymomas round nuclei and sparse delicate cytoplasmic processes
include brisk mitotic activity and microvascular proliferation . Nuclear pleomorphism is generally mild, but it is exaggerated 1r
These are considered to have more prognostic impact than the rare giant cell phenotype (2376 ,1874).
other histopathological features , such as nuclear pleomor-
phism or tumour necrosis (1123} . Supratentorial ependymomas Diagnostic molecular pathology
with plentiful high-grade features are considered CNS WHO Molecular analysis of supratentorial ependymomas should be
grade 3, with the caveat that the histopathological grading of directed towards determining ZFTA (C11orf95) and YAP1 starus
ependymomas does not consistently relate to overall outcome and excluding tumours with similar histopathological features
(840,698}. such as BCOR-altered tumours and MN1-altered astroblasro-
mas . A variety of diagnostic tests can be used to detect the pres-
lmmunophenotype ence or absence of ZFTA (C11orf95) or YAP1 rearrangements
Most supratentorial ependymomas show immunoreactivity for including interphase FISH , RT-PCR -based sequencing meth-
8100 or GFAP, which is accentuated around blood vessels and ods, and next-generation sequencing (including transcripronie
perivascular pseudorosettes. Characteristic immunoreactivity sequencing) [2511 ,2374 ,2401 ,2370). In 17-30% of supratentc-
for EMA , with a paranuclear dot-like pattern and along luminal rial ependymomas , ZFTA (C11orf95) and YAP1 fusion genas
surfaces of true ependymal rosettes , can be found in many, but cannot be detected {1002,2288); a diagnosis of supratentoriJ1
not all , ependymomas . Expression of OLIG2 is usually present ependymoma can be used in this context . The detection ot J
in only a small percentage of tumour cells (1579,2348 ,1633}. genetic alteration not involving ZFTA (C 71orf95) or YAP 1 shoulu

162 Gliornas, glioneuronal tumours . and neuronal tumours


I

i. .
Fig.2.155 Supratentorial ependymoma. A,B By immunohistochemistry, GFAP is variably expressed In supratentorial ependymomas. C OLIG2 is usually present in sparse
tumour cells. D lmmunoreactivity for EMA is present in a variable number of tumour cells and usually manifests as paranuclear dots or ring structures.

prompt the use of the suffix "NEC". An inability to perform the Box2.32 Diagnostic criteria for supratentorial ependymoma NEC and supratentorial
ependymoma NOS
appropriate analysis prompts the addition of "NOS" {1946}.
Essential:
Essential and desirable diagnostic criteria Supratentorial tumour with morphological and immunohistochemical features of
See Box 2.32. ependymoma
AND
Staging For NEC: the detected genetic alteration is not a fusion gene involving either
Not clinically relevant ZFTA (C11orf95) or YAP1
OR
Prognosis and prediction For NOS: genetic analysis was unsuccessful or unfeasible
Most outcome data for ependymomas are derived from ret-
rospective studies in an era before molecular classification .
Among adults , supratentorial location is associated with a children and in adults, and second -look surgery for resection of
poorer outcome than infratentorial location (3311 }. The clinico- residual tumour is increasingly advocated . In addition to neuro-
pathological utility of grading for ependymal tumours remains surgical intervention, postoperative radiotherapy is considered
controversial {840}, although the use of CNS WHO grade in the the standard of care, in the absence of metastases . for lower-
therapeutic stratification of adult patients with supratentorial ing the risk of local recurrence (2074 ,2745}. The vast majority
ependymoma remains established practice (3311} . Infiltration of of tumour relapses are due to a lack of local control , and the
adjacent CNS parenchyma by ependymoma has been reported number of late failures is substantial , especially in adults. Cer-
as an adverse prognostic indicator (1123} . Complete surgical ebrospinal fluid spread develops in as many as 15% of patients .
resection is the best predictor of long-term survival both in more often in CNS WHO grade 3 tumours (2744).

Gl1olllas , glioneurorial tumours. and rH' LJr or ul tuil lL•u r ,~ 163


Pietsch T
Supratentorial ependymoma, Pajtler KW
Aldape KD Ruda R
ZFTA fusion- positive Gilbertson RJ
Korshunov A
Taylor MD
Venneti S

Definition
Supratentorial ependymoma, ZFTA fusion - positive, is a circum-
scribed supratentorial glioma with a ZFTA (formerly C11orf95)
fusion gene , focally demonstrating pseudorosettes or ependy-
mal rosettes and comprising uniform small cells with round
nuclei embedded in a fibrillary matrix. In most of these supraten-
torial ependymomas , ZFTA is fused with RELA.

ICD-0 coding
9396/3 Supratentorial ependymoma, ZFTA fusion-positive

ICD-11 coding
2AOO.OY & XH1511 Other specified gliomas of brain & Ependy-
moma, NOS
Fig. 2.156 Supratentorial ependymoma, ZFTA fusion-positive . A T1-weigh ted :jJ, ~.
Related terminology MRI with gadolinium, showing a well -demarcated left frontal lesion with heterogeneous
None contrast enhancement and areas of necrosis. B Coronal FLAIR MRI showing sG~ ·
spicuous surrounding oedema.
Subtype(s)
None Imaging
On neuroimaging , intratumoural haemorrhage , cysts , and peritt. . -
Localization moural oedema are common . High diffusion-weighted 1mag1ng
Most ZFTA fusion-positive cerebral ependymomas arise in the signals with concomitant low signals in apparent diffusion coef-
frontal or parietal lobe {2288,1893). Less common sites are ficient or T2-weighted images suggest d iffusion restriction Mos:
the thalamus or the region of the hypothalamus I third ventri- tumours show strong , but often inhomogeneous, enhancemer:
cle. lntracranial extra-axial ZFTA fusion-positive supratentorial in their solid components after intravenous gadolinium injection
ependymomas have been reported (1992,2288). {2326,1625,2288,1028).

Clinical features Epidemiology


Clinical symptoms and signs include focal neurological deficits ZFTA fusion-positive tumours account for the majority oi
or seizures, as well as features of raised intracranial pressure. supratentorial ependymomas and may occur both in ch il dren

.
Flg.2.157 Supratentorial ependymoma, ZFTA fusion-positive. Tumour cells with round nuclei set in a fibrillary matrix. Branching capillary blood vessels (Al or branching bl 'Cu
vessels and dystrophic calcification (I ) can be seen.

164 C3110mas . gliorit::uronal tumours . and neuronal tumours


I
.... --- ..
Ag. 2.158 Supratentorial ependymoma, ZFTA fusion-positive. A Universal cytoplasmic expression of L1CAM. B Nuclear immunoreactivity for p65 (encoded by RELA).

and in adults . The percentage of supratentorial ependymomas


wi th a ZFTA fusion varies between retrospective studies: 20-58%
Macroscopic appearance
Generally, supratentorial ependymomas with a ZFTA fusion are
I
in adults, 66-84% in children 12511,2374,3462,2370,3258). sharply demarcated tumours of soft consistency. Dystrophic
calcification and necrotic areas are common findings .
Etiology
Evidence from mouse modelling and cross-species genom- Histopathology
ics strongly suggests that ZFTA fusion-positive supratentorial Supratentorial ependymomas with a ZFTA fusion are demar-
ependymomas arise from radial glia 13155,2632,2357). Genetic cated from adjacent brain and composed of cells character-
susceptibility in association with this molecular entity has not ized mainly by round uniform nuclei with speckled chromatin
been reported . and poorly defined fibrillary cytoplasm . Pseudorosettes are
not prominent in most cases , and true ependymal rosettes are
Pathogenesis rare . These tumours often have a network of branching capillary
Fusion of the ZFTA gene with partner genes, mainly RELA , is blood vessels and a clear cell phenotype.
believed to be the principal oncogenic driver of the disease.
Rearrangements containing ZFTA have been demonstrated to lmmunophenotype
result from chromothriptic events on chromosome 11 {2401). A The immunophenotype of ZFTA fusion-positive ependymomas
pathological activation of NF-KB signalling has been demon- is similar to that of other ependymomas . Ependymomas with a
strated in supratentorial ependymomas with a ZFTA::RELA fusion ZFTA::RELA fusion show nuclear accumulation of p65 protein
12511 ,2401 ). Homozygous deletions of CDKN2A indicate a dis- (encoded by RELA) and universal cytoplasmic expression of
ru ption of cell-cycle control in a subset of these tumours 11512). L1CAM 12401 ,2511}. lmmunoreactivity fo r p65 has been found

~l
·,.
0

... 0..

U:•
z
L

Flg.2.159 SupratentoriaJ ependymoma, ZFTA fu sion- positive. Copy-number profile derived from lllumina 450K array data, showing chrornothnpsis on chromosome 11.

Glior 11as , glioneuron I tumuu1s, ~1n ci 118U I l ) f k11 tlJ11luu 1::; 165
to have a slightly higher specificity for this molecularly de'fined Box2.33 Diagnostic criteria for supratentorial ependymoma. ZFTA fusion-positive
ependymoma than does L1CAM expression (926) . Essential:
Supratentorial tumour with morphological and immunohistochemical features of
Grading ependymoma
zFTA-fused supratentorial ependymomas show varying
AND
degrees of anaplasia and have been regarded as CNS WHO
Gene fusion involving ZFTA (C11 orf95)
grade 2 or 3 on this basis . Such information should be included
in an integrated diagnosis [1939). Desirable:
DNA methylation profile aligned with supratentorial ependymoma, ZFTA fusion-
positive
Cytology
Cytological preparations generally show uniform cells with lmmunoreactlvity for p65 (RELA) or L1CAM
round nuclei and sparse delicate cytoplasmic processes .
Nuclear pleomorphism is generally mild, but it is exaggerated in Staging
the rare giant cell phenotype (2376,1874). Not clinically relevant

Diagnostic molecular pathology Prognosis and prediction .


Diagnostic tests for detecting ZFTA fusions include several Available clinical outcome data on molecularly definea
sequencing methods , interphase FISH , and molecular inversion supratentorial ependymomas suggest that ZFTA fus ion-posi-
profiling (2401,2511 ,2374,2370,1002,2236,1512) . An RT- PCR tive tumours have the poorest outcome. However, outcome data
method targeting the different types of ZFTA:: RELA fusion has derived from retrospective studies show great variance , and
been proposed for detecting the most frequent fusio ns (2374 , validation of those findings needs to be undertaken in prospec-
1067,1992) . DNA methylation- based classification can be help- tive therapeutic trials (2374 ,926 ,1893,3462,2370 ,1002,2073.
ful , complementing assays directed towards identification of the 3258) . Homozygous deletion of COKN2A and/or COKN28 has
fusion . been identified as an independent predictor of poor outcome
(overall survival) in a series of ependymomas with ZFTA: :RELA
Essential and desirable diagnostic criteria fusions (1512) .
See Box 2.33 .

166 Gilomas. gl1oneuronal tumours . and neuronal tumours


Pietsch T
Supratentorial ependymoma, Pajtl er KW
Aldape KO Ruda R

I
YAP1 fus ion- positive Gilbertson RJ
Korshunov A
Taylor MD
Venneti S
'

Definition
Supratentorial ependymoma, YAP1 fusion-positive, is a circum-
scribed supratentorial glioma with a YAP1 fusion gene, focally
demonstrating pseudorosettes or ependymal rosettes and
comprising uniform small cells with round nuclei embedded in a
f1brillary matrix. In most of these supratentorial ependymomas,
YAP1 is fused with MAMLD1.

ICD-0 coding
9396/3 Supratentorial ependymoma, YAP1 fusion-positive

ICD-11 coding
2AOO.OY & XH151 1 Other specified gliomas of brain & Ependy-
moma, NOS
Fig. 2.160 Supratentorial ependymoma, YAP1 fusion-positive. A T2-weighted coro-
nal MRI showing a large cystic tumour. B Ependymoma with YAP1: :MAMLD1 fusion.
Related terminology T1 -weighted axial MRI showing a large cystic tumour with contrast enhancement of
None solid tumour parts.

Subtype(s) Imaging
None Neuroimaging shows that tumours have sharp margins and
prominent cystic components . They are mostly isointense on
Localization T1- and T2-weighted images. Contrast enhancement of solid
Most YA P1 fusion-positive tumours are located within or adja- tumour components is heterogeneous . Peritumoural oedema is
cent to the lateral ventricle. variable {113) .

Clinical features Epidemiology


YAP1 fusion-positive supratentorial ependymomas are often YAP1 fusion-positive ependymomas are uncommon and
large by the time of presentation . Clinical features include symp- appear to be restricted to young ch il dren . In paediatric cohorts ,
toms and signs of raised intracranial pressure, as well as focal they account for 6-7.4% of supratentorial ependymomas . The
neurological deficits or seizures . M:F ratio is 0.3:1 {2374 ,3462,113 ,2370 ,3258}.

Gl1ornas , gli neuron I tumour s. a nd n t: uru11Ltl tu 11.ou r ~, 167


Box 2.34 Diagnostic criteria for supratentorlal ependymoma, YAP1 fusion-positive
Etiology
Data suggest that YAP1 fusion - positive ependymomas derive Essential:
from PAX6 -positive radial glial neural stem cells [2372 ,825). Supratentorial tumour with morphological and 1mmunohistochemlcal features of
ependymoma
Pathogenesis AND
Genomic fusions of the YAP1 gene with MAMLD1 or other
Gene fusion involving YAP1
partner genes appear to be the principal oncogenic driver
of the disease. Functional genomic analyses suggest. that Desirable:
YAP1 .:MAMLD1 fusions function as an oncogenic driver through DNA methylation profile aligned with supratentorial ependymoma. YAP1 fusiort-
the recruitment of nuclear factor I (NFI) and TEA domain (TEAD) positive
family members (2372,825}. No immunoreactivity for p65 (RELA) or L1CAM
PAS-positive eosinophilic granular bodies
Macroscopic appearance
Ependymomas with a YAP1 fusion have macroscopic appear-
ances similar to those of other supratentorial ependymomas . Cytology .
They are circumscribed and focally haemorrhagic , with a soft Cytological preparations generally show unifo_rm cells "'' 1-
consistency. round nuclei and sparse delicate cytoplasmic processes
Nuclear pleomorphism is generally mild , but it is exaggerated rr
Histopathology the rare giant cell phenotype (2376 ,1874).
Like other ependymal tumours, YAP1-fused supratentorial
ependymomas are demarcated from adjacent brain . They are Diagnostic molecular pathology
composed of relatively uniform cells with small to medium- Molecular testing for YAP1 fusions includes several sequencing
sized round or angulated nuclei. Ependymal rosettes are strategies and interphase FISH (1992,1002,2236) . DNA meth-
present in some tumours . Clear cell , papillary, or tanycytic ylation-based class ification can complement tests d1rectea
phenotypes have not been recorded . Mitotic activity is highly towards identification of the fusion .
variable. In most cases, the fibrillary matrix contains PAS-
positive eosinophilic granular bodies {113) . Frequent findings Essential and desirable diagnostic criteria
are vascular endothelial prol iferation , dystrophic calcification , See Box 2.34 .
and necrosis.
Staging
lmmunophenotype Not clinically relevant
Supratentorial ependymomas with a YAP1 fusion show wide-
spread and strong immunoreactivity for EMA (113}. There is no Prognosis and prediction
expression of L1CAM , and tumour cell nuclei are negative for Although often large at presentation and predominantly occur-
p65 (RELA}. ring in young children , ependymomas with a YAP1 fusion carr11
a prognosis in retrospectively stud ied cohorts that appears to
Grading be favourable when compared with that of other supratentona1
YAP1-fused supratentorial ependymomas show variable ependymal tumour types (113 ,2370 ,3258} . Molecular markers
degrees of anaplasia, and such information should be included or clinical characteristics further defining prognos is in these
in an integrated diagnosis (1939}. tumours are currently unknown .

168 Gl1omas glioneuronnl tumours , and neuronal tumours


Posterior fossa ependymoma Venneti S
Aldape KO

I
Pajtler KW
Pietsch T
Ramaswamy V
Taylor MD

Definition
Posterior fossa ependymoma is a circumscribed glioma in
the posterior fossa, focally demonstrating pseudorosettes or
ependymal rosettes and comprising uniform small cells with
round nuclei embedded in a fibrillary matrix. The diagnosis of

I
posterior fossa ependymoma should be used when molecular
analysis either cannot assign a molecular group (not elsewhere
classified ; NEC) or is not feasible (not otherwise specified; NOS)
(CNS WHO grade 2 or 3).

ICD-0 coding
9391 /3 Posterior fossa ependymoma, NOS

ICD-11 coding
2AOO.OY & XH1511 Other specified gliomas of brain & Ependy-
moma, NOS
Fig.2.162 Posterior Iossa ependymoma. MRI shows a tumour in the fourth ven tricle
Related terminology (arrows). Note that the aqueduct of Sylvius and the th ird ventricle are enlarged.
None

Subtype(s)
None

Localization
Posterior fossa ependymomas mainly arise in the region of the
fourth ventricle , including the floor, lateral aspect (cerebellar
peduncles), and roof. They can also occur in the cerebellopon-
tine angle {3463}.

Clinical features
Common clinical presentations relate to mass effect on sur-
rounding posterior fossa structures and include secondary
hydrocephalus. Clinical presentations vary by age and are often
nonspecific (e.g. headache, vomiting, and lethargy). Babies can
present with a rapidly growing head circumference. Flg.2.163 Posterior Iossa ependymoma. Ependymoma in the fourth ventricle (ar-
rows) displacing adjacent posterior Iossa structures.
Imaging
MRI usually demonstrates a homogeneous mass filling the neoplasms in children and adolescents (birth to 19 years) are
fourth ventricle. Haemorrhages and punctate calcifications may ependymomas {2344). In the USA , a higher incidence of epen-
be observed {2541 ). The presence of intratumoural cysts and dymomas is reported in White people , includ ing child ren wi th
necrosis can result in variable enhancement on gadolinium eastern European ancestry, than in the Black and Hispanic
injection. MRI can show lateral extension of the tumour via the populations {2054 ,2344,3591 ).
foram ina of Luschka and extension through the foramen of
Magendie into the cisterna magna {2541} . Etiology
The etiology is unknown . Associations with specific gene tic
Epidemiology susceptibilities have not been reported .
Posterior fossa ependymomas of all types can develop at any
age . However, they are most frequent in children , with a median Pathogenesis
age at presentation of 6 years . They are slightly more frequent Acros~ all type~ of posterior fossa ependymoma , copy-number
in male patients (52- 62%) (2054,2073 ,1043,2612,2032,3463) . alterations leading to altered gene expression are hypothesized
Accord ing to the Central Brain Tumor Regi stry of the United to pla~ an .essen.tial role in pathog enesis , as are epigenetic
States (CBTRUS), approximately 8% of all neuroepithelial alterations, 1nclud1ng aberrant DNA methylation patterns, EZHIP

Gl1omas , gl1oneuronal tumours, and 11 euru rul tumours 169


overexpression , and loss of H3 p .K28me3 (K 27me3) (1975 , cytoplasmi c clearing can be present, but this is more comrnor
1974,2373. 490) . in supratentorial ependymomas . Tumour cells with elongated
nuclei fo cally arrang ed in fascicles represent the tanycytic Pai.
Macroscopic appearance tern .
Posterior fossa ependymomas are usually circumscribed
tumours arising in the four th ve ntric le. They appear tan -col- Grading
oured and are soft or spongy, with a gritty consistency if calci- Posterior fos sa ependymomas can be assigned CNS WHrJ
fied . Tumour ce lls can grow through the foramina of Luschka grade 2 or 3, ideally in the context of an integrated diagnosi·
to envelop the lower cranial nerves and the posterior inferior (1939) . High-grade fe atu re s in ependymomas include bris~
cerebellar artery. mitotic activity and microvascular proliferation . These arc.
considered to have more prognostic impact than other histr:,:
Histopathology pathological features , such as nuclear pleomorph1sm or turno.Jr
Generally, posterior fossa epen dymomas are ci rcum scribed necrosis (1123) . However, efforts to risk-stratify cases on tr~
tumours composed of uniform smal l ce ll s with indistinct cyto- basis of histopatholog ical grading criteria have yielded incor.
plasmic borders and round nuclei. Nodules of tumour cell s, in sistent results (840,1123,2032 ,2073) .
which the cell densi ty is higher than in surrounding syncy tial
areas , are common . Perivascular pseudorosettes are almost lmmunophenotype
always present an d are characterized by tumour cells arranged Most posterior fossa ependymomas show immunoreact1VI',
in a radial fashion around blood vessels to create an interven- for S10 0 or GFAP, which is accentuated around blood ·1es-
ing anucleate zone. True ependymal rosettes are composed of sel s in perivascular pseudorosettes . EMA expression can 0 ~
columnar or cuboidal cells surrounding a central lumen . They seen in most ependymomas as a paranuclear dot-l ike patterr
are observed in a minority of cases . Regions of nuclear pleo- or ring -like structu res. However, this is not an entirely spec1f1c
morphism , increased mitotic activity in nodules , microcystic finding (1256 ,1579}. OLI G2 expression is usually absent. anc
change, calcification , and hyalinization of blood vessels can som e tumours ca n show focal immunoreactivity for cytokeratins
be observed . Rarely, cartilaginous or osseous metaplasia is (including CK7 and CK20) (1421 ,2348 ,2567,3302) .
present.
Some posterior fossa ependymomas can have a focal papil- Ultrastructure
lary or pseudopapillary architecture, including finger-like pro- Ependymomas demonstrate characteristic ultrastructural fea-
jections lined by a single layer of cuboidal cells or papillae in tures , including cili a (9 + 2 microtubular pattern), junct1ona1
wh ich a central blood vessel is covered by layers of tumour complexes on lateral surfaces of cells , and microvilli on lum1r:a
cells . Clear cell change mimicking oligodendroglioma-like surfaces (1124) .

170 Gl1urr1cis . gl1onc~u1onetl turnour ~_; <.Hld neuronal tumours


,,
ka

Fig. 2.165 Posterior Iossa ependymoma. A lmmunoreactivity for GFAP is typically present in many (but not all) tumour cells. Note accentuation of staining in perivascular pseu-
dorosettes. B lmmunoreactivity for EMA typically shows a paranuclear dot-like pattern and occurs along the luminal surface of true ependymal rosettes. C lmmunoreact1vity ior
NFP highlights axons in surrounding brain parenchyma and demonstrates the pushing border of the tumour. D At the ultrastructural level, the cells of an ependymoma typically
show cilia with a 9 + 2 microtubular pattern and microvilli at luminal surfaces.

Box2.35 Diagnostic criteria tor posterior Iossa ependymoma Absence of immunoreactivity for H3 p.K28me3 (K27me3) 1n the
Essential: nuclei of tumour cells is a surrogate marker for PFA ependy-
Posterior fossa tumour with morphological and immunohistochemical features of moma (2384), but classification using DNA methylation profiling
ependymoma is considered the standard method , because nuclear expres-
AND sion of H3 p.K28me3 (K27me3) is present in both PFB tumours
Absence of morphological features of subependymoma
and subependymomas . If appropriate molecular testing was
successfully performed but did not assign a molecular group,
AND (for NOS lesions)
a diagnosis of posterior fossa ependymoma can be used with
Molecular group evaluation was indeterminate, generated no result, or was not the suffix "NEC " [1946) . An inability to perform the appropriate
feasible
analysis prompts the addition of "NOS ".

Cytology Essential and desirable diagnostic criteria


Cytological preparations generally show uniform cells with See Box 2.35.
round nuclei and sparse delicate cytoplasmic processes .
Nuclear pleomorphism is generally mild , but it is exaggerated Staging
in the rare giant cell phenotype {2376) . Tumour cells can form Not clinically relevant
clusters and palisades around vascular structures, reflecting
the arrangement of perivascular pseudorosettes . Prognosis and prediction
There is no robust relationship between histological grade and
Diagnostic molecular pathology prognosis for posterior fossa ependymomas {840.11231. How-
If feasible, posterior fossa ependymomas should be assigned ever, the extent of surgical resection and the status of chromo-
to a molecular group (PFA. PFB , or subependymoma) 12374). some 1q are consistent outcome indicarors 1478,1611 .1123 .26121

Gliomas . gl1oneuronal tumou1s. l.lncl n •ur '- · 1d 1 t., l ._.r-; 17 1


Posterior fossa group A (PFA) Venneti S
Aldape KO
Pietsch T
Ramaswamy v
ependymoma Korshunov A
Pajtler KW
Taylor MD

Definition 100%
Posterior fossa group A (PFA) ependymoma is a circumscribed
posterior fossa glioma aligned with the PFA molecular group of VI
QJ 80%
VI
ependymomas, demonstrating pseudorosettes or ependymal ro
u
rosettes and comprising uniform small cells with round nuclei 0 60%
QJ
embedded in a fibrillary matrix . An ependymoma can be clas- C"I
sified as PFA by identifying a loss of nuclear H3 p.K28me3 ~
c 40%
QJ
(K27me3) expression in tumour cells or by DNA methylation u
Qj
profiling. a..
20%

ICD-0 coding
0%
9396/3 Posterior fossa group A (PFA) ependymoma 0-4 5-9 10-17 18+
Age group (years)
ICD-11 coding - EPN-PFA - EPN-PFB
2AOO.OY & XH1511 Other specified gliomas of brain & Ependy-
Fig.2.166 Posterior Iossa ependymoma. PFA ependymoma (EPN-PFA), sometimes
moma, NOS
referred to as "infantile posterior Iossa ependymoma", predominantly occurs in infan s
and children. PFB ependymoma (EPN-PFB) occurs mainly in older children and young
Related terminology adults.
None
Pathogenesis
Subtype(s) Cell of origin
None PFA ependymomas are thought to arise from an undifferentiated
glial stem or progenitor cell in the developing hindbrain [33381
Localization
Studies correlating neuroimaging with molecular group have Molecular profile
suggested that PFA ependymomas more frequently arise from PFA ependymomas exhibit characteristic DNA methylation pat-
the roof or lateral aspect of the fourth ventricle than from its floor terns, including hypermethylation of CpG islands and global
{3463,2373). DNA hypomethylation [1975,2374,223}. PFA ependymomas
show a global reduction of the repress ive histone mark H3
Clinical features p.K28me3 (K27me3) , which impacts several pathways , includ-
The clinical features of PFA ependymomas are similar to those ing neuroglial differentiation and cell-cycle regulation /223.
described for posterior fossa ependymomas in general. 2373}, and is caused by overexpression of EZHIP {2373) . EZHIP
phenotypically mimics the oncohistone H3 p .K28M (K27M ) oy
Epidemiology binding to the H3 p.K28 (K27) methyltransferase EZH2 and
PFA ependymomas predominantly occur in infants and chil- inhibiting the function of PRC2 [1448 ,2597,2520,1374}. Although
dren , with a median age of 3 years {3463 ,2373 ,2374,2612) . most PFA ependymomas do not carry recurrent genetic muta-
The proportion of posterior fossa ependymomas classified tions , about 9% exhibit mutations in EZHIP In addition , about
as PFA aligns with age: > 95% of posterior fossa ependymo- 4% harbour H3 p.K28M (K27M) mutations, which are mutually
mas in children aged < 6 years are PFA tumours, and PFB exclusive with EZHIP mutations (260 ,1065,2373,2765). A study
ependymomas are rare in this age group; the proportion of that examined 675 PFA ependymomas by DNA methylat 101
posterior fossa ependymomas classified as PFA decreases to profiling identified 2 molecular subgroups and g molecular
45-50% in adolescents and 5-11% in adults {3463 ,2373 ,2374, subtypes of PFA ependymomas {2373) . However the clinico-
2612) . PFA ependymomas are slightly more prevalent in male pathological implications of these findings have y~t to be fully
patients (59-62%) than in female patients {3463 ,2373 ,2374, evaluated .
2612) .
Macroscopic appearance
Etiology The macroscopic features of PFA ependymomas are s ·1ar ro
~!~hough the exact etiology of PFA ependymomas is unknown , those described for posterior fossa ependymorn as in g~~~ral
it is hypothesized that aberrant epigenetic alterations may be
central drivers. Associations with specific genetic susceptibili- Hlstopathology
ties have not been reported . PFA ependymomas show the histopathological feat ur s
described for posterior fossa ependymomas in gener'-u

172 Gl1ornas glioneuror1al turnnurs, and neuronal tumour s


High -grade features , including prominent mitotic activity and Grading
microvascular proliferation , were observed in 64% of PFA PFA ependymomas show varying degrees of anaplas1a and have
ependymomas in one study (2373] . Clear cell, papillary, and been regarded as Cl\IS WHO grade 2 or 3 on this basis Such
tanycytic patterns can be focally present (2236] . information should be included in an integrated diagnosis 11939]

Jmmunophenotype Cytology
PFA ependymomas exhibit a reduction in H3 p.K28me3 Cytological preparations generally show uniform cells with
(K27me3), which can be readily assessed by immunohisto- round nuclei and sparse delicate cytoplasmic processes
chemistry (2384,223,2373). Retained H3 p.K28me3 (K27me3) Nuclear pleomorphism is generally mild, but it is exaggerated
immunoreactlvity in endothelial cells can be used as an inter- in the rare giant cell phenotype (2376) . Tumour cells can form
nal control for the method. In most PFA ependymomas, tumour clusters and palisades around vascular structures, reflecting
cells show a global reduction in H3 p.K28me3 (K27me3) the arrangement of perivascular pseudorosettes.
expression , but variability in the proportion of immunonegative
cells can be encountered. A cut-off value of 80% immunoposi- Diagnostic molecular pathology
tive cells has been proposed , above which an epen dymoma is Demonstration of loss of H3 p.K28me3 (K27me3) by immuno-
more likely to fal l into the PFB mol ecular group {2384,223,1002, histochemistry, or assignment to the PFA molecular group by
1228,3487,3580). DNA methylation profiling , is required for a diagnosis of PFA

Flg.2.167 Posterior Iossa group A (PFA) ependymoma. A Nodules of high cell density are common in PFA epend ymomas. B Subtle pseudorosette formation and a high cell
density characterize some PFA ependymomas.

--
-~~> -
,
-
Flg.2.168 Posterior Iossa group A (PFA) ependymoma. A In most examples of this tumour type, immunoreactivity tor H3 p K28me3 (K27me3) 1n tumour cells 1s lost. B lm -
munoreactivity for H3 p.K28me3 (K27me3) can sometimes be retained in a variable proportion of tumour cells.

Gltt)n k l S, glioneur Jnal turn our::, .Jr 1u n e u1 (i1 ,,J! :u1 n,;u1s 173
Box2.36 Diagnostic criteria for posterior Iossa group A (PFA) ependymoma
ependymoma . Because of its prognostic significance across all
posterior fossa ependymomas \1123\, gain of chromosome 1q Essential:
is often assessed in PFA ependymomas, even though molecu- Posterior fossa tumour with morphological and immunohistochemical features of
lar subtypes of PFA ependymoma with or without 1q gain can ependymoma
be associated with a poor outcome (2373) . AND
Global reduction of H3 p.K28me3 (K27me3) in tumour cell nuclei
Essential and desirable diagnostic criteria
OR
See Box 2.36.
DNA methylation profile aligned with PFA ependymoma

Staging Desirable:
Not clinically relevant Stable genome on genome-wide copy-number analysis

Prognosis and prediction


Extent of surgical resection is associated with outcome \1123) .
PFA ependymomas have a poor prognosis compared with molecular subtypes of PFA ependymoma, those with or w1thou:
that of PFB ependymomas {2374) . Gain of chromosome 1q is 1q gain can have an equally poor outcome \2373 ). The prog-
a reproducible adverse prognostic indicator across all poste- nostic significance of an H3 p.K28me3 (K27me3) mu tation 1n a
rior fossa ependymomas {478,1611,1123) . However, among small proportion of PFA ependymomas is unknown .

174 Gliomas . ylioi ieuronal tumou rs. and neuronal tu rnours


Posterior fossa group B (PFB) Venneti S
Aldape KO

I
ependymoma Pajtler KW
Pietsch T
Ramaswamy V .
Taylor MD

Definition
Posterior fossa group B (PFB) ependymoma is a circumscribed
posterior fossa glioma aligned with the PFB molecular group of
ependymomas, demonstrating pseudorosettes or ependymal
rosettes and comprising uniform small cells with round nuclei
embedded in a fibrillary matrix . An ependymoma can be clas-
sified as PFB by DNA methylation profiling . Retention of nuclear
H3 p.K28me3 (K27me3) expression is observed , but it is not
specific for PFB ependymomas .

ICD-0 coding
9396/3 Posterior fossa group B (PFB) ependymoma

ICD-11 coding
2AOO.OY & XH1511 Other specified gliomas of brain & Ependy-
moma , NOS

Related terminology
Fig. 2.169 Posterior Iossa group B (PFB) ependymoma . T1 -welghted MRI shows a
None
large tumour in the fourth ventricle of a 37-year-old man.

Subtype(s) Epidemiology
None PFB ependymomas occur in adults and are more common in
adolescents than in children and infants /3463 ,2374 ,2612 ,2373 .
Localization 490,1513). The median age at presentation is 30 years (range:
PFB ependymomas can occur anywhere in the region of the 1-72 years). The relative frequency of the PFB molecular group
fourth ventricle and its exit foramina, but they are thought to among ependymomas is closely related to age: 90% 1n adults,
arise more frequently from the floor of the fourth ventricle than 20-50% in adolescents, and < 5% in infants and children aged
from the roof or lateral recesses [3463}. < 5 years /3463,2374,2612,490). PFB ependymomas are slightly
more prevalent in female patients (55-59%) [3463 ,2374,2612,
Clinical features 490}. Among the five molecular subgroups of PFB ependymoma
Clinical manifestations are similar to those observed in posterior {490) , PFB-1 , PFB-2, and PFB-3 tumours tend to occur in patients
fossa ependymomas in general. aged 25-30 years . PFB-4 tumours arise in a younger age group

B Expres -

Gllomas . gl1oneuronal tum our s . . _md nel i1 1.. r1 \I t1:·1 1L11 · 175
eox2.37 Diagnostic criteria for posterior Iossa group B (PFB) ependymoma immunohi stochemistry (2384,223,2373}_.. Rare ependyrnorr.~­
Essential: with a DNA methylation profile that class1f1e~ the~ as PFB 5hr,,
Posterior Iossa tumour with morphological and immunohlstochemical fea tu res of redu ced H3 p.K28me3 (K27me3), but the s1gnif1cance of thr:;·_,
ependymoma findings is unclear /2384 ,1002,2737) .
AND PFB ependymomas show varying degrees of anaplas13 ~r ·,
ONA methylation profile aligned with PFB ependymoma have been regarded as CNS WHO grade .2 or 3 on thi s h'1', .
Such information should be included in an integrated d1aqrry_ .
Desirable:
{1939) .
Chromosomal instability and aneuploidy on genome-wide copy-number analysis
Retained nuclear expression of H3 p.K28me3 (K27me3) Cytology .
Cytological preparation s generally show unifo.rm cells ,,,-,
round nuclei and sparse delicate cytoplasmic proces~H
(median age: 15 years) , whereas PFB -5 tumours occur in older Nuclear pleomorphism is generally mild, but it is exagger8 t~.::
individuals (median age: 40 years). PFB-2 and PFB -4 tumours in the rare giant cell phenotype \2376) . Tumour cells can frr
are more common in male patients , whereas PFB -3 and PFB -5 clusters and palisades around vascular structures , reflecti· ~
tumours are more common in female patients \490} . the arrangement of perivascular pseudorosettes .

Etiology Diagnostic molecular pathology


The etiology of PFB ependymoma remains to be elucidated . Demon stration of H3 p.K28me3 (K27me3) retention by 1 m rri~­
No association with specific genetic susceptibilities has been nohistochemistry or assignment to the PFB molecular gro•.. ;:
reported . by DNA methylation profiling is required for a diagnosis of PF~
ependymoma. Nuclear expression of H3 p .K28me3 (K27m~
Pathogenesis is retained in PFB ependymomas , but this finding is not specrt c.
The pathogenesis of PFB ependymomas is unclear, but it is PFB ependymomas exhibit widespread cytogenetic abnorm2 -
thought to be driven by epigenetic changes and copy-number ties , with many chromosomal aberrations {1975,2374,223 .49S
alterations that together produce aberrant gene expression . the most common of which include loss of 22q , monosomy;;
and trisomy 18 (in 50- 60% of cases) .
Macroscopic appearance
The macroscopic features of PFB ependymomas are similar to Essential and desirable diagnostic criteria
those described for posterior fossa ependymomas in general. See Box 2.37.

Histopathology Staging
PFB ependymomas show the histopathological features Not clinically relevant
described for posterior fossa ependymomas in general. High-
grade features , including prominent mitotic activity and microvas- Prognosis and prediction
cular proliferation, were observed in 41% of PFB ependymomas Incomplete surgical resection and loss of 13q were assoc 1 at~
in a study of 51 patients {2374). with a poor prognosis in a cohort of 212 PFB ependymomas..
Practically all PFB ependymomas exhibit retention of H3 Gain of 1q did not show a relationsh ip with overall prognosis r
p.K28me3 (K27me3), which can be readily assessed by these tumours \490) .

176 Gl1omas, glioneuronal tumours, and neuronal tumours


Spinal ependymoma Pietsch T
Aldape KO

I
Korshunov A
Pajtler KW
Taylor MD
.
Venneti S

Definition
Spinal ependymoma is a demarcated spinal glioma demon-
strating pseudorosett~s or ependymal rosettes and comprising
uniform small ~ells with round nuclei embedded in a fibrillary
matrix and , typically, a low level of mitotic activity. By definition,
the tumour lacks features of myxopapillary ependymoma or
subependymoma . When testing is feasible , MYCN amplification
is absent.

ICD-0 coding
9391/3 Spinal ependymoma, NOS

ICD-11 coding
2AOO.OZ & XH1511 Other and unspecified neoplasms of brain
or central nervous system & Ependymoma, NOS

Related terminology
None

Subtype(s) Fig.2.171 Spinal ependymoma, CNS WHO grade 2.. Sagittal T1-weighted MRI of a
spinal ependymoma showing gadolinium contrast enhancement.
None

Localization Etiology
Spinal ependymomas occur along the spinal canal and are Various studies have shown that 18-53% O"f patients with neu-
intramedullary tumours {1674) . A cervical or cervicothoracic roti bromatosis type 2 develop spinal ependymomas, but that
localization is common, in contrast to myxopapillary ependy- clinical symptoms related to these are evident in < 20% of cases
momas, which nearly always arise in the lumbar region (1674). (153 ,2525,652}. Spinal ependymomas develop more frequently
in patients with neurofibromatosis type 2 with germline non-
Clinical features sense and frameshift mutations in the NF2 gene than in those
Spinal ependymomas do not have clinical features specific with other types of NF2mutation (153}. A single Japanese family
enough to differentiate them from other intramedullary spinal with 2 of 4 siblings affected by cervical spinal ependymomas
cord tumours. Patients often present with back pain and a mye- has been described . Neurofibromatosis type 2 was excluded
lopathy (motor and sensory deficits related to dysfunction of the in this family, another tumour suppressor gene on chromo-
spinal cord). some 22q being considered causal (3540} .
Spinal ependymomas show frequent chromosomal altera-
Imaging tions, the most common being chromosome 22 loss, which
On MRI examination, spinal ependymomas are intramedullary occurs in the majority of cases (2374) . Sporadic spinal ependy-
tumours. They are contrast-enhancing and mostly hypointense momas frequently have a somatic NF2 mutation {820) .
on T1-weighted images and hyperintense on T2-weighted
images. They often display cystic changes, haemorrhage, Pathogenesis
necrosis , and/or calcification (1674). Approximately 60% of Spinal ependymomas are hypothesized to originate from radial
ependymomas are associated with an intramedullary cyst glia-like stem or progenitor cells [1490,3155) . Experimental Nf2
(syringomyelia) rostral or caudal to the tumour {1674) . inactivation in mice resulted in increased growth and reduced
apoptosis of embryonal spinal cord neural progenitor cells , sug -
Epidemiology gesting that NF2 activation has an important role in the patho-
Ependymal tumours represent 20.6% of primary spinal tumours genesis of spinal ependymomas (1035).
in children and adolescents and 17.6% of those in adults aged
~ 20 years, according to a statistical report from the Central Macroscopic appearance
Brain Tumor Registry of the United States (CBTRUS) {2344) . Spinal ependymomas are generally circumscribed tumours.
Across various studies, the median age at diagnosis of patients They appear soft and are mostly grey-white in colour. They can
with spinal ependymoma ranges from 25 to 45 years . The show cystic changes, calcification , and signs of haemorrhage.
reported M:F ratio ranges from 1:1.3 to 2.16:1 1238 J.

Gl1omas, glioneuronal tumows , a.ncl llt}urun:i l 1un 1ours H7


Grading
Although anaplast1c ependymoma has been removed frorn tr'!
classi fication , a pathologist can still choose to assign r;1tr ~r
CN S WH O grade 2 or grade 3 to an ependymoma , accorrJinq ·,,
its his topathological featu res 18361 Most spinal ependymr;rr;j·,
are CNS WH O grade 2; CNS WHO grade 3 tumours are rare.:
1852,4951. CN S WH O grade 3 spinal ependymomas show r:,r," .
spicuous mi totic activity, usually in the context of a high r,i; ,
density, and they tend to invade ad1acent spinal cord structur~c.
Where possible, such tumou rs should be d1stingu1shed frr;rr
MYCN-ampl ified spinal ependymoma and H3 K27-altered d1f
fuse midline glioma.

lmmunophenotype
lmmunoreactivity for GFAP, S100 . and vimentin is characteristic
as is focal dot-like or ring-like intracy toplasmic immunoreactl'1-
ity for EMA. In contrast to astrocytic spinal neoplasms, spinal
ependymomas are largely negative for OLI G2. They do not
,,. express SOX10 , wh ich is found in schwannoma , pilocytic astro-
Flg.2.172 Spinal ependymoma. Tumour showing characteristic perivascular anucle-
ate areas (pseudorosettes).
cytoma , and most diffuse gliomas.

Electron microscopy
Histopathology Ultrastructurally, ependymal features includ in g intracytoplas-
The classic form of spinal ependymoma is composed of iso- mic villi , cilia, and complex intercellular zipper-like junctions are
morphic glial cells with round to oval nuclei and indistinct cyto- present 13087,1079).
plasmic membranes . The cells are embedded in a fibrillary gl ial
matrix and have a moderate to high cell density. A characteris- Cytology
tic feature is the anucleate perivascular zone (pseudorosette) ; Cytological preparations generally show uniform cell s with round
tumour cells are radially arranged around a blood vessel, with nuclei and sparse delicate cytoplasmic processes . Nuclear
fibrillary processes creating the perivascular anucleate zone . pleomorphism is generally mild but can be increased in some
True ependymal rosettes with a central lumen or ependymal cases. Tumour cells can form clusters and pali sades around
tubules are present in only a minority of cases . Mitotic activ- vascular structures, reflecting the arrangement of perivascular
ity in the classic form is usually low. The rare tanycytic pattern pseudorosettes.
with prominent spindle-shaped cells and bipolar processes ,
often in the absence of pseudorosettes, is overrepresented in Diagnostic molecular pathology
spinal ependymomas and must be distinguished from pilocytic Spinal ependymomas with a typical morphology are easily rec-
astrocytoma and schwannoma. Ependymomas can show cal- ognized. They are also readily distinguished from myxopapillary
cification , haemorrhage, necrosis, cystic change, metaplastic ependymomas, subependymomas , and MYCN-ampl ified spi-
cartilage, and bone and myxoid degeneration . nal ependymoma by their DNA methylation profile 12374 ,30871

178 Giiomas , gilon uronal tumours , and neurona l tumou rs


Occasionally. a spinal ependymoma with a classic morphology Box 2.38 01;i9nost1c criteri;i tor sp1n1tl ~pendymom;i
e hibits the DNA methylat1on profile ot myxopapillary epenrly -
Es ntlsl:
moma 13462} The prognostic significance of a rnyxopap1llary
DNA methylation profile in the face of an ostensibly discordant Spinal rumour with morphologrcal and immunohistoci1em1cal features of
ependymoma
morphological d1agnos1s rerna1ns to be clarified Frequent loss
of chromosome 22q and mutations of NF2 are characteristic of AND
spinal ependymomas 12374,1674} By def init1on . MYCN amplifi- Absence of morphological features of myxopap11lary ependymoma or
cation is absent subependymoma
Desirable:
Essential and desirable diagnostic criteria DNA methylat1on profile aligned with spinal ependymoma
See Bo 2 38 . Loss of chromosome 22q
No MYCN amplification
Staging
Not clinically relevant
reflecting a large number of late relapses (1131 ). Extent of resec-
Prognosis and prediction tion is a prognostic factor in most studies . patients with gross
Spinal ependymomas are associated with a favourable outcome total resection having favourable progression-free survival
1n children and adults , with progression-free and overall survival (2306,330) . From the limited data available, it can be concluded
rates of 70-90% and 90-100%. respectively, over 5-10 years that the prognosis of CNS WHO grade 3 spinal ependymoma 1s
1238). However, progression-free survival declines over time , unfavourable (238) .

Gl1omas . gl1oneuronal tumours a11l.i neurc,1 i__1I tu. 11,_,l 11 -.., 179
Pietsch T
Spinal ependymoma , MYCN-amp lified Gia nnini C
Ald ape KO Ramaswamy V
Ko rshunov A Taylo r MD
Paj tler KW Venneti S

Definition
Spinal ependymoma, MYCN-amplified, is a well -demarcated
12
spinal glioma demonstrating pseudorosettes or ependymal Female
il
c:
rosettes and comprising un iform , densely packed small cells Q)
10
• M ale
with round nuclei embedded in a fibrillary matri x. Practically :.;:;
ro a
all tumours display microvascular proliferation, necrosis , and a ....a.0 6
high mitotic count. By definition , MYCN amplification is demon- ....
Q)
strated in tumour cells . .0 4
E
:::J
ICD-0 coding z
9396/3 Spinal ependymoma , MYCN-ampli fied
3 • 5
Decade
ICD-11 coding
2AOO .OZ & XH1511 Other and unspecified neoplasms of brain Flg.2.174 Spinal ependymoma, MYCN-amplified. Age and sex distribution of patients
or central nervous system & Ependymoma, NOS with spinal ependymoma, MYCN-ampllfied (all published cases (2825,3087,1079,259511

Related terminology Clinical features


None Presenting symptoms depend on tumour location, but they tyoi-
cally incl ude neck or back pain and progressive numbness and
Subtype(s) weakness in th e extremities .
None
Epidemiology
Localization MYCN-amplified spinal ependymoma is rare , with only
Tumours are localized to the spinal cord , primarily to the cervi- 27 reported cases (17 in women and 10 in men; M:F ratio
cal or thoracic levels (in 78% of cases) and less frequently to 1:1 .7). The med ian age at presentation was 31 years (range
lumbar levels (in 7% of cases) [2825 ,3087,1079,2595} . 12-56 years) (2825 ,3087,1079,2595}.
Primary tumours may be intramedullary (sometimes with an
exophytic component extending into the spinal canal (3087}) Etiology
or mostly extramedullary (1079,2595} . They are generally large No specific etiology has been identified. However, multiple
and involve multiple spinal segments. Leptomeningeal dissemi- schwannomas were reported in 1 patient, raising th e diag-
nation is frequent at diagnosis , or it occurs at some point during nostic possibility of neurofibromatosis type 2 (2825 ). No other
the course of the disease (2825,3087,1079,2595}.

Fig. 2.175 Spinal ependymorna, MYCN-amplified. A Tumour with a focal pseudopapillary architecture and densely packed cells. B High cell density, mitotic activity, and vagu~
pseudorosette formation are present.

180 Gl1omas . gl1oneuronal tumours , and neuronal tumours


patient has sh?wn any signs ~f neurofibromatosis type 2, and Two of four tumours with CNS WHO grade 2 h1stopalhol091r,al
no NF2 mutation was found 1n the 4 analysed cases {3087, features at presentation progressed to CNS WHO grndA 3 ;:1t
1079).
recurrence (2825 ,3087,1079).
Pathogenesis
lmmunophenotype
MYCN, a member of the MYC family of proto-oncogenes , On immunohistochemistry, tumour cells express GFAP and
encodes a transcription factor that regulates the expression show a focal cytoplasmic dot-like pattern of EMA expression
of genes involved in cell growth (2361 . How MYCN amplifica- {3087,1079) . MYCN protein expression can be detected by
tion contributes specifically to ependymoma development Is immunohistochemistry (1079,2595) . Cells are 1mmunonegative
unknown . for OLIG2 (3087). Nuclear immunoreactivity for H3 p.K28me3
(K27me3) was variably present in the 4 tumours from one study
Macroscopic appearance (3087). but was retained in all tumours from another (1079) .
The macroscopic features of this specific tumour type have not
been described , but they are unlikely to differ markedly from Grading
those reported for other circumscribed high-grade gliomas . Although nearly all MYCN-amplified spinal ependymomas
show high-grade histopathological features and have a poor
Histopathology prognosis, this molecularly defined ependymal tumour has yet
MYCN-amplified spinal ependymomas display perivascular to be assigned a CNS WHO grade (836) .
anucleate zones (pseudorosettes) an d can have a papil lary or
pseudopapillary architecture {3087,1079,2595). Most have high- Cytology
grade histopathological features, such as a high N:C ratio, plen- Tumour cells are generally small and have round hyperchro-
tiful mitotic activity, microvascular proliferation, and necrosis . matic nuclei and scant cytoplasm .


l

- •'

... .

.
, ')111, .;
., :•,#, •. .• .
. ,~ '. . .
":· ·, 41 ... . . . t

c .... . .:. .. .
fig. 2.176 Spinal ependymoma, MYCN-arnplified. A Extensive nuclear expr~ssion of MYCN is detected by 1mmunoh1stochem1stry. B lmmunoh1stochemistry shows many
tumour cells expressing GFAP, which is typically present in perivascular rad1~t1ng processes. C Nuclear 1mmunoreact1v1ty for H3 p.K28me3 (K27me3) can be completely lost,
partially lost (as shown here), or retained. D Electron microscopy. lntercellular 1unct1ons end at a m1crolumen filled with m1crovlil1 , typical of ependymal d1fferent1at1on. Cilia (black-
and-white arrows) and basal bodies (black arrows) are also present.

Gl1orT1as. gl1oneuronal tuni our:., d i 1(1 11('ltr l "1 11 tL.11 •1 " · - 181
Diagnostic molecular pathology
High-level MYCN amplification is present and remains stab1~J :::·
relapse (2825 ,3087,"1079,2595) . Additional chromosomal r:r,r;;
number alterations occur with variable frequency and 1ncl1JrJ'-
loss of chromosome 10 (in 32% of cases) and focal lrw,.:.~
on chromosome 11q (in 26% of cases) . Demonstration r,f ,..,~
p.K28me3 (K27me3) loss by immunohistochem1stry ri:i01Jir~­
assessment of histone H3 genes for genetic alterat1r;r-
because MYCN amplification can be found in diffuse rn1r.i11'~­
gliomas with H3 p.K28M (K27M) mutation 1404) ·
M YCN-amplified spinal ependymoma has a Ol'JA methylatr;-
profile distinct from that of other ependymal tumour types ~­
.. .. .. .. .. .. • • .. i--· well as from that of neuroblastoma and MYCN-ampl1f1ed paw~,-
Fig. 2.177 Spinal ependymoma, MYCN-amplified Chromos m . · · atric-type glioblastoma /1079 ,2595) .
amplification of 2p24.3-p24.2, including MYCN. . o al m1croarray showing

Essential and desirable diagnostic criteria


See Box 2.39.
Box2.39 Diagnostic criteria tor spinal ependymoma, MYCN-amplified
Essential: Staging
Spinal tumour with morphological and lmmunohistochemical features of Not cl inically relevant
ependymoma
AND Prognosis and prediction
MYCN-ampl ified spinal ependymoma is an aggressive turno1... :
MYCN amplification
associated with poor progression -free and overall survival corr-
Desirable: pared with that of other spinal ependymomas. Early metastasis
D~A methyla.tion profile aligned with spinal ependymoma, MYCN-amplified and dissemination throughout the neura xis are frequent. A·
High-grade h1stopathological features patients with reported follow-up data have relapsed deso1te
aggressive treatment /2825 ,3087,1079,2595) .

182 Gl1omas, glioneuronal tumours , and neuronal tumou rs


Myxopapillary ependymoma Rosenblum MK
Korshunov A

I
Pa1tler KW
Pietsch T
Taylor MD .
Venneti S

Definition
Myxopaplllary ependymoma is a glial neoplasm characterized
by the radial arrangement of spindled or epithelioid tumour cells
around blood vessels with perivascular myxoid change and
microcyst formation (CNS WHO grade 2).

ICD-0 coding
9394/1 Myxopapillary ependymoma

ICD-11 coding
2AOO .OY & XH15U1 Other specified gliomas of brain & Myxo-
papillary ependymoma

Related terminology
None

Subtype(s)
None

Localization
Myxopapillary ependymomas arise almost exclusively in , and
are the most common tumours of, the conus medullaris and
filum terminale, accounting for 83% of 320 filum terminale epen-
dymomas in one study (500) . Multifocality has been described Fig.2.178 Myxopapillary ependymoma. The well-circumscribed, contrast-enhancing
primary tumour in the Ilium is associated with a drop metastasis in the low thecal sac.
(2201 ), as have examples originating in the cervicothoracic spi-
nal cord (2993), lateral ventricle (3390), fourth ventricle (504),
and brain (2609). Tumours outside the CNS are also recog- SEER Program study (of cases in patients aged s; 21 years: USA,
nized ; these are most often sacrococcygeal (mimicking chor- 1973-2012, n = 122), the median patient age was 16 years, and
domas) or presacral in position , with rare examples described 63% of cases occurred in male patients [1956) .
in the uterine adnexa, ischioanal fossa , mediastinum, and lung
{3559). A conus I filum terminale primary must be excluded Etiology
when a myxopapillary ependymoma occurs at higher levels of Unknown
the neuraxis (84) .
Pathogenesis
Clinical features The pathogenesis of myxopapillary ependymomas is unknown.
Lower back pain , often chronic , is an almost constant manifes- A variety of recurring chromosomal copy-number abnormalities
tation of myxopapillary ependymomas , and it can be accompa- have been described in these tumours, but no consistent struc-
nied by sciatica , sensorimotor deficits indicative of myelopathy, tural variants or other driving mutations (3462,2708). Upregu-
impotence, or urinary and faecal incontinence. Urgent neuro- lation of key enzymes associated with the Warburg metabolic
surgical intervention may be required to restore lower extrem - phenotype, including HK2, PKM2 , and POK, has been demon-
ity function. Neuroimaging typically reveals an ovoid , sharply strated (1973).
delimited, and contrast-enhancing mass . Cerebrospinal fluid -
borne spread , particularly seeding of the distal thecal sac, may Macroscopic appearance
be evident at presentation . Often encapsulated , myxopapillary ependymomas are soft and
pink to tan -grey, may be grossly gelatinous, and can manifest
Epidemiology cystic changes and haemorrhage.
Incidence rates of 0.6- 1.0 cases per 1 million person-years have
been reported from the USA and Europe, with an M:F ratio of Histopathology
1.4-2:1 (2345 ,3400,214) . Myxopapillary ependymomas occur at Prototypical is the radial arrangement of cuboidal to elongated
all ages but most commonly affect adults; peak case rates were tumour cells around hyalinized fibrovascular cores in papillary
found in patients aged 25-29 years and 45-59 years in one SEER fashion , with accumulation of basophilic, myxoid material around
Program analysis (USA, 2004-2012, n = 773) (214) . In another blood vessels and in microcysts . Myxoid material, highlighted

Gl1omas , glioneuronal tu mour s, el l\ i net I!\ i11,1l u1n1L~1 ir~; 183


! - 1
Flg.2.179 Myxopapillary ependyr:ioma. A Epltheliold tumour cells are arranged around fibrovascular cores and microcysts containing myxoid materiaJ. B Tapering tumourr~
processes are oriented towards a f1brovascular core with collaring myxoid matrix.

lmmunophenotype
Diffuse immunoreactivity for GFAP distinguishes myxopapillar,
ependymomas from metastatic carcinomas , paragang l1om3s
schwannomas, chordomas , and myxoid chondrosarcom a~
/1799 ,3302) . lmmunolabell ing for S100 is also typical , and reac-
tivity for CD99 and CD56 is frequent {1799}. Tumour cell nuc i;· 1

are not immunoreactive for OLIG2 , and dot-like cytoplasmic


EMA labelling is typically absent. Myxopapillary ependymomas
are often labelled by the AE1/AE3 pancytokeratin cockta il, bur
they are generally negative for CAM5 .2, CK5/6 , CK?, and CK2S
{1799,3302} .

Cytology
lntraoperative squash and smear preparations of classic myxo-
papillary ependymomas show epithelioid to spindled cellu-
lar profiles , papillary structuring of tumour cells around blooa
vessels with perivascular myxoid change , and tumour cells
Fig.2.180 Myxopapillary ependymoma. Tumour cells are diffusely and strongly
arranged around myxoid microcysts . These features are diag·
GFAP-immunoreactive. nostic in the appropriate clinical setting. Such features may alsc
be evident in fine-needle aspiration material , which may oe
assessed for confirmatory GFAP expression (41 }.
by PAS and Alcian blue positivity, is useful in the identification of
examples manifesting little, if any, papillary structure and com- Diagnostic molecular pathology
posed instead of epithelioid cells In confluent sheets . Tumour Myxopapillary ependymomas with a classic morphology are
cell spindling and fascicular growth are common , and subpop- easily recognized , but these tumours also have a unique ONA
ulations of pleomorphic tumour giant cells can be seen in other- methylation profile (2374,3462 ,2236}. However, tumours w1tr
wise typical cases {3585) . Eosinophilic , PAS-positive spherules the histopathological features of classic ependymoma, part1cu·
(balloons) that exhibit spiculated reticulin staining are an occa- larly lumbosacral lesions with tanycytic or papillary patterns
sional featu re . Common secondary alterations include fibrosis, may also cluster with myxopapillary ependymomas /3462
haemorrhage, and haemosiderin deposition. Typical myxopap- 2236) . This reflects the fact that myxopapillary ependymomas
illary ependymomas show, at most, only low-level mitotic activ- can exhibit little myxoid change, form pseudorosettes of the
ity, and the Ki-67 labelling index usually does not exceed 2- 3% usual ependymal type, and manifest spindle cell (tanycytld
/2559) . Exceptional examples termed "anaplastic myxopaplllary features . The prognostic significance of a myxopapillary epen-
ependymomas" manifest regional hypercellularity and reduced dymoma methylation profile in the context of uncharacterisuc
mucin in association with at. least two of the following features: histopathological features remains to be clarified . Recurrent
~ 2 mitoses/mm 2 , Ki-67 labelling index ~ 10%, microvascular gains of chromosome 16 and losses of chromosome 10 ha~e
proliferation, and spontaneous necrosis {1835). been documented [3462) .

184 Gl1omas , glloneuronal tumours , and neuronal tumours


Box 2.40 Diagnostic criteria for myxopapillary ependymoma
Essential:
.
I
Glioma with papillary structures and perlvascular myxoid change or at least focal
myxold microcysts
.
AND
lmmunoreactivity for GFAP
AND (for unresolved lesions)
DNA methylation profile aligned with myxopapillary ependymoma
Desirable:
Papillary arrangements of tumour cells around vascularized fibromyxoid cores
Location in the filum terminale or conus medullaris

Essential and desirable diagnostic criteria


See Box 2.40.

Staging
Because myxopapillary ependymomas may exhibit leptome- Fig. 2.181 Myxopapillary ependymoma. Papillary structure, perlvascular myxoid
ningeal dissemination , some groups have recommended that change, and spindled tumour cells oriented towards fibromyxoid cores are evident 1n
craniospinal MRI and cerebrospinal fluid cytology should be this smear preparation.
performed after initial surgery and diagnosis (2745}.
which may be evident at diagnosis in 2:: 50% of patients (189,
Prognosis and prediction 3). Tumours arising in the conus have a poorer prognosis than
Spinal myxopapillary ependymomas are associated with a cauda equina examples because the former adhere densely
relatively favourable prognosis in children and adults , with to the spinal cord and are less amenable to resection . Rad io-
10-year overall survival rates > 90% (3400,189,214,2481, therapy improves progression-free survival (3400) . Cytological
3). Many patients, however, live with persistent disease and atypia and modest mitotic activity do not appear to influence
require repeated operations and adjuvant therapy, because outcome (2993). Tumours with anaplastic histology may carry
myxopapillary ependymomas often resist complete removal an increased risk of aggressive behaviour (1835). Spinal myxo-
owing to locally advanced growth and/or cerebrospinal fluid- papillary ependymomas rarely metastasize to extraneural sites ,
borne seeding of the thecal sac or more rostral neuraxis. Pae- but metastasis frequently complicates the course of sacrococ-
diatric patients are at heightened risk of such dissemination, cygeal tumours (3559}.

,1

Gl1omas, glioneuron I tumours , ancl neuron I tum uL1r s 185


Subependymoma Rosenblum MK
Korshunov A
Pajtler KW
Pietsch T
Taylor MD
Venneti S

Definition
Sube.pendymo~1a is a glioma characterized by the clustering
of uniform to mildly pleomorphic tumour ce ll nuclei in an abun-
dant fibrillary matrix prone to microcystic cha nge (C NS WH O
grade 1).

ICD-0 coding
9383/1 Subependymoma

ICD-11 coding
2AOO.OY & XH8FZ9 Other specif ied gl iomas of brain & Sub-
ependymoma

Related terminology
Not recommended: su bependymal glomerate astrocytoma {346).
Flg. 2.1 82 Subependymoma. A sharply circu mscribed, intraventricular mass with 'c:
Subtype(s) of wispy contrast enhancement is demonstrated in this T1-weighted MRI .
None
Etiology
Localization Predisposing factors await furt her definition . Familial casss
The most frequent sites of origin are the fourth ventricle (in including examples in monozygotic twins , are well documentec
50-60% of cases) and lateral ventricles (30-35%), followed but rare {2271). These include examp les associated wit~
distantly by the third ventricle and spinal cord, where sube- trichorhinophalangeal syndrome type 1 and germline TRPS '
pendymomas preferentially arise as eccentric masses in cervi- mutation; a subset of sporadic subependymomas also ar-
cothoracic segments (2755,269,3557,3293) . Cerebral, cerebel- bour TRPS1 mutations (937) . Isolated cases have also beer
lar, bulbar, and cerebellopontine angle examples have been described in patients with hered itary ani ridia and PAX6 mutc-
reported {1626,269} . tion {1982), as well as Noonan syndrome with germline PTPw·
mutation {323). Patients with cran iopharyn giomas have beer
Clinical features reported to develop rare th ird-ventricular subependymomas
Subependymomas are often asymptomatic and discovered [516}. Losses of chromosomes 19 and 6, th e latter restricted tc
only incidentally on neuroimaging for unrelated reasons or at infratentorial tumours, appear to play a ro le in many sporadic
autopsy. Symptomatic intracranial examples are typically asso- cases {3462) .
ciated with manifestations of ventricular obstruction and intra-
cranial hypertension, occasionally showing evidence of intra- Pathogenesis
tumoural/intraventricular haemorrhage. Sensorimotor deficits How the chromosomal or genetic abnormalities displayed m
indicative of myelopathy characterize intramedullary lesions. subependymomas contribute to tumour development is cur-
rently unknown .
Imaging
Most subependymomas are sharply demarcated , hypoin- Macroscopic appearance
tense or isointense on Tl-weighted MRI, and hyperintense on Subependymomas are firm , grey, and generally circumscribe-a.
T2-weighted MRI ; some exhibit calcification, cystic change, and intracranlal examples typically bulge into ventricles in an exo-
foci of contrast enhancement {2755,269). phytic fashion . Cystic changes , calc ificat ion, and focal haemor-
rhage (unusual) may be apparent.
Epidemiology .
Because subependymomas are often clinically silent, reli.ab le Histopathology
incidence figures are lacking . A SEER Program analys1~ ~f Typical Is the clustering of small , euch romatic . and round to ova1
466 intracranial cases (USA , 2004-2013) found an overall 1n~1- nuclei (resembling those of subependymal glia) in a voluminous
dence of 0.055 cases per 100 000 person-years, an M:F ratio matrix of fibrillary cytoplasmic processes . Microcystic change.:
of approximately 2.5:1, and peak incidence in adults age? are common , particularly in lateral ve ntricular subependymo-
40- 84 years (2243) . Subependymomas account for ~pprox1- mas , as are calcifications. Nuclear pl eomorphism and prol1 -
mately 8% of ependymal tumours and < 1% of intracranial neo- erative microvascular abnormalities may be encountered . w1 ri
exceptional cases exhibiting low-level mitotic activity and eve1'
plasms 12837,1765).

186 Gl1ornas, glioneu ronal tumours. and neuronal tumours


I
FJg.2.183 Subepen~ymoma. A Tumour cell nuclei are small, uniform, and without mitotic activity. B Tumour cell nuclei cluster in a dense fibrlllary matrix. ~ Microcystic change
in the fibrillary matrix is shown.

non-palisading necrosis (2755}. Just as classic ependymomas Box 2.41 Diagnostic criteria for subependymoma
can focally exhibit subependymoma-type histology, so may Essential:
subepen dymomas focally manifest perivascular pseudoro- Circumscribed glioma with clustering of tumour cell nuclei within expansive, focally
settes. Subependymoma-predominant neoplasms with nodules microcystlc fibrillary matrix
of classic ependymoma (termed "mixed ependymoma-sub- AND
ependymoma") are well recognized and mentioned below (see
Lack of conspicuous nuclear atypia
Prognosis and prediction). Otherwise typical examples may also
AND
harbour elements of fibrillary astroglial or (rarely) gemistocytic
morphology. Sclerotic and ectatic blood vessels, haemorrhage, Absent or minimal mitotic activity
and haemosiderin deposits are common . Odd ities include mel- AND (for unresolved lesions)
anotic pigmentation (2727) and sarcomatous change (3212, DNA methylation profile aligned with subependymoma
2700} .
Recurrent copy-number abnormalities are chromosome 19
/mmunophenotype loss and partial chromosome 6 loss (infratentorial cases)
Subependymomas manifest diffuse GFAP immunoreactivlty (3462) . TRPS1 mutations have been documented (937). Rare
and can display focal dot-l ike EMA expression , but they do brainstem gliomas exhibiting subependymoma-type histology
so less frequently than ependymomas (2755 ,3528}. Some are are H3 p.K28M (K27M)-mutant (3528).
reported to express OLIG2 or synaptophysin (269), but this
appears to be exceptional (3528) . SOX10 labelling , if present, is Essential and desirable diagnostic criteria
limited (1655) . Also reported is the expression of HIF1a, TOP2B , See Box 2.41 .
MDM2, nucleolin, and phosphorylated STAT3 (1697), as well as
aquaporin-1 and aquaporin-4 (2272} . Subependymomas retain Staging
ATRX expression, do not express the mutant IDH1 p.R132H Not clinically relevant
or BRAF p.V600E gene products , and (except for rare bulbar
lesions) are negative for H3 p.K28M (K27M) , but they retain H3 Prognosis and prediction
p.K28me3 (K27me 3) expression (3528). An excellent prognosis is associated with subependymomas
(2755 ,269,2243,3557,3293). Postsurg ical recurrence is rare ,
Cytology even after subtotal resection , and only exceptional instances of
The relatively uniform round or oval nuclear profiles , nuclear subependymal seeding or anaplastic progression have been
clustering , and fibrillary matrix of subependymomas are appar- reported {2880 ,269) . Cytolog ical pleomorph ism, occasional
ent in smear and squash preparations, which may also demon- mitoses, and necrosis have not proved prognostically significant
strate myxoid and microcystic changes (3209) . (2558,2755). A Ki-67 labelling index > 1% has characterized
some subependymomas exhibiting recurrence (1729 ,3206) or
Diagnostic molecular pathology dramatic interval growth on surveillance (2261 ). The traditional
Molecular analyses have shown subependymomas in the grading of mixed ependymoma-subependymoma according to
supratentorial, posterior fossa , and spinal anatomical compart- the histology of their ependymoma components is based on a
ments to have distinct DNA methylation profiles (2374 ,3462, historical series in which such lesions behaved more aggres-
2236) . However, although tumours at each site with the hlsto- sively than pure subependymomas (28271, but more recent
pathological features of subependymoma cluster together in analyses have not repl icated this observation (2755 ,269) .
these analyses and are not placed in other molecular groups, Assessments of chromosome 19 status and DNA methylation
some tumours el iciting the morphological diagnosis of classic profiling may prove useful in the risk stratification of patients with
ependymoma may also cluster with typica l subependymomas mixed or morphologically ambiguous lesions \3462) . The occur-
{3462,2236) . The prognostic significance of a subependymoma rence of H3 p.K28M (K27M) mutation in bra1nstem gliomas
DNA methylation profile in the face of an ostensibly discordant exhibiting subependymoma histology has not been associated
morphological diagnosis remains to be clarified . with rapidly fatal progression (3528) .

Gl1omas , glioneuronal turnour s unll rie~111..11ul tu111l~Ur':> 187


Choroid plexus papilloma Pietsch T
Hasselbl att M
Malkin 0
Paulu s W

Definition Imaging
Choroid plexus papilloma is an intraventricular papillary neo- On CT an d MRI , choroid plexus papillomas usually pre: w
plasm derived from choroid plexus epithelium , with very low or as isodense or hyperdense . T1-isointen se, T2-hyperintenst;
absent mitotic activity (CNS WHO grade 1). irregul arly con trast-enhancing , wel l-del ineated masses w1 t1- r
th e ventri cles , but irregu lar tumou r margins and d1 sserninat<:ir;
ICD-0 coding disease may occur (1178!.
9390/0 Choroid plexus papilloma
Spread
ICD-11 coding Even benign choroid plexus papill oma may seed cells into trs
2A00 .22 & XHORF9 Choroid plexus papilloma & Choroid plexus CSF; in rare cases , this can result in drop metastase s su rrouriJ-
papilloma , NOS ing the cauda equina (3055) .

Related terminology Epidemiology


None Although choroid plexus tumours con stitute 0.3-0 .8% of a"
brain tumours overall , they account for 2- 4% of those that occur
Subtype(s) in children aged < 15 years , and for 10- 20% of those occurrirg
None in the first year of life (455) . The average annual incidence s
0.3 cases per 1 million population {1452,2667,346 8) . Choro!d
Localization plexus papillomas account for 58 .2% of the c horoid plexus
Choroid plexus papillomas are located within the ventricular tumours in the SEER database. The M:F ratio is 1.2'1. About
system where normal choroid plexus can be found . They occur 80% of lateral ventricular tumours are found in patients agea
most often in the lateral ventricles, followed by the fourth and < 20 years , whereas fourth ventricle tumours are evenly distrib-
third ventricles. Rare cases have been described within the spi- uted across all age groups (3468) .
nal cord or in ectopic locations (2428) . Multifocal occurrence is
exceptional (2486}. Etiology
Environmental risk factors for the development of choro10
Clinical features plexus papilloma have not been confirmed . Earlier reports of a
By blocking cerebrospinal fluid (CSF) pathways, choroid plexus possible role of SV40 {1364) have not been conf irmed 1n more
papillomas tend to cause hydrocephalus and increased intra- recent studies. Genomic analysis of choroid plexus pap illoma
cranial pressure. It has been debated whether overproduction suggests a role for genes involved in the development and biol-
of CSF is a major contributing factor to hydrocephalus {259) . ogy of plexus epithelium (e.g. OTX2 and TRPM3). It is thought
that their alteration may contribute to the in itial steps of cho roid
plexus papilloma oncogenesis {1455).

Flg.3.01 Choroid plexus papilloma. A Sagittal, contrast-enhanced , T1 :weighted MRI shows a strongly enhancing , cauliflower-like mass in the th ird ven tricle 01 a 22_1110 ntn
old girl a Sagit1al , contrast-enhanced . Tl -weighted MRI shows a choroid plexus papllloma in the fourth ventricle of a 38-year-old man. c lntraoperative endoscopic view 01 .l
choroid plexus papilloma .

190 Clioro1d f; le;.us lumours


I

Fig.3.02 Choroid plexus papilloma. A Papillary pattern with a single layer of monomorphic cuboidal cells. B lmmunohistochemistry for the potassium channel Kir?.1 shows
typical membranous labelling of the apical surface of tumour cells. C lmmunohistochemlstry for S100. D lmmunohistochemistry for transthyretin (prealbumin).

Genetic susceptibility papillomas 12669}. contains genes that control the proliferation
Choroid plexus papilloma occurs in Aicardi syndrome, a disor- of choroid plexus progenitor cells {1012}. TP53 mutations are
der with lethality in males and presumably X-linked dominant rare in choroid plexus papillomas (present in < 10% of cases)
inheritance, which is defined by the triad of agenesis of the cor- 13097}. Epigenetic profiling identified three distinct methylation
pus callosum , chorioretinal lacunae, and infantile spasms (43}. groups; cluster analysis showed separation of most choroid
In the setting of an X;17(q12;p13) translocation, hypomelanosis plexus papillomas from choroid plexus carcinomas 13184,2507).
of Ito has been associated with the development of choroid
plexus papilloma in several cases (3574}. Gains of the short arm Macroscopic appearance
of chromosome 9, a rare constitutional abnormality, were shown Choroid plexus papillomas are circumscribed, cauliflower-like
to be associated with hyperplasia of the choroid plexus and with intraventricular masses. Cysts and haemorrhage may occur.
choroid plexus papilloma (2278 ,1012}.
Histopathology
Pathogenesis The well-developed papillary pattern is composed of fibro-
Choroid plexus papillomas are believed to derive from monocili- vascular fronds that are covered by a single layer of uniform
ated progenitors of plexus epithelium located in the roof plate, cuboidal to columnar epithelial cells with round or oval , mono-
and they show activation of the sonic hedgehog and Notch sig- morphic nuclei. Mitotic activity is absent or very low: < 1 mito-
nalling pathways known to play a crucial role in the proliferation sis/mm2 (equating to < 2 mitoses/10 HPF of 0.23 mm 2) (1463,
of plexus epithelial precursor cells {1876} . Notch signalling sup- 2927). Brain invasion with cell clusters or single cells, high cel-
presses multiciliate differentiation of progenitor cells and may lularity, necrosis , nuclear pleomorphism , and focal blurring of
allow sonic hedgehog-mediated proliferative signals via the the papillary pattern may occasionally occur. Cells tend to be
primary cilium in plexus papilloma cells {1876}. more crowded and nuclei more variable than in non-neoplastic
Both classic cytogenetic and genome-wide array-based choroid plexus. Choroid plexus papillomas can acquire unusual
approaches demonstrated hyperdiploidy with whole-chromo- histological features, including oncocytic change, mucinous
some gains in choroid plexus papilloma {779,2669,2079,1455). degeneration, melanization , tubular/glandular architecture
The pathogenetic impact of these chromosomal gains is not (adenoma), neuropil-like islands, and degeneration of connec-
understood. Because constitutional trisomy or tetrasomy of chro- tive tissue (e .g. xanthomatous change; angioma-like increase of
mosome 9p is linked to choroid plexus hyperplasia, it is specu- blood vessels; and bone, cartilage , or adipose tissue formation)
lated that this region, showing gains in 50% of sporadic plexus 12563,395 ,1253,2002).

Cho r01 d p lc>xu~ luin~iur ~ 191


not been described in other primary brain tumours or cerebr 81
metastases {1250,582}. The glutamate .transporter EAAT 1 -,
expressed in most choroid plexus papillo~as , wh~re as 1t 1,-,
absent in endolymphatic sac tumours and in > 95 Yo of nr;r
neoplastic choroid plexus specimens (2844,258) . Transthyret1r
(prealbumin) is positive in normal choroid plexus . but sta1 n1rr~
may be negative or variable among choroid plexu s pap1 llorrw,
and it is also seen in some metastatic carcinomas 1 2428 .~CJ
The Ki-67 proliferation index is usually < 5% and often < 1"~
(3244,3479).

Cytology
In CSF samples and cytological imprints, clusters of choro11
plexus papilloma cells show epithelioid morphology with isom0r-
fig. 3.03 Choroid plexus papilloma cells in cerebrospinal fluid. lmmunohistochemis-
try for the potassium channel Kir7.1.
phic round nuclei and moderately developed cytoplasms 116411

Box3.01 Diagnostic criteria for choroid plexus papilloma Diagnostic molecular pathology
Choroid plexus papillomas are easily recognized by their his-
Essential: tology. Genome -wide chromosomal copy-number analysis can
Demonstration of choroid plexus differentiation by histopathologlcal and demonstrate characteristic hyperploidy {1455). Choroid plexus
immunophenotypic features papillomas also show typical epigenetic signatures [460 ).
AND
Absent or low mitotic activity Essential and desirable diagnostic criteria
AND See Box 3 .01 .
lntraventricular or cerebellopontine angle location
Staging
Not relevant
/mmunophenotype
More than 90% of choroid plexus tumours are positive for cyto- Prognosis and prediction
keratins (usually CK7-positive and CK20-negative), vimentin , Prognosis is excellent, especially upon gross total resection
and 8100 (1204,2428). GFAP and EMA may be expressed , but In a series of 41 patients with choroid plexus papilloma, the
staining is often weak or focal (772,2207). Membranous staining 5-year overall survival rate was 97% (1738). Sim ilar results were
(mainly) of the apical border for the inward-rectifier potassium obtained in another series {1463) . Choroid plexus papillomas
channel Kir7.1 is typical for non-neoplastic choroid plexus epi- in children aged < 36 months also have an excellent prognosis

• thelium and is retained in > 80% of choroid plexus papillomas


and about 50% of choroid plexus carcinomas , whereas it has
after surgery alone (1786) . Malignant progression of choroid
plexus papilloma is rare (589,1464).

192 Ct"10ro1d plexus tumou rs


Atypical choroid plexus papilloma Pietsch T
Hasselblatt M
Malkin D
Paulus W

Definition Imaging
Atypical choroid plexus papil loma is a choroid plexus papilloma No differences in MRI characteristics have been reported
that has increased mitotic activity but does not fulfil the criteria between choroid plexus papilloma and atypical choroid plexus
for choroid plexus carcinoma (CNS WHO grade 2). papilloma (3479] .

ICD-0 coding Spread


9390/1 Atypical choroid plexus papilloma Atyp ica l choroid plexus papilloma has been reported with
metastasis at diagnosis in 17% of cases (3479}.
ICD-11 coding
2A00.22 & XH3Y57 Choroid plexus papilloma & Atypical cho- Epidemiology
roid plexus papilloma In two paediatric studies of choroid plexus tumours, patients
with atypical choroid plexus papilloma were on average
Related terminology younger (median age: 8 months in one study, 0.84 years in
None the other) than those with choroid plexus papilloma (median
age: 35 months in one, 2.18 years in the other) (3183 ,2927)).
Subtype(s) Although both studies included patients of up to 18 years of
None age, the oldest patients with atypical choroid plexus papilloma
were 10- and 11-year-olds.
Localization
Atypical choroid plexus papillomas arise in locations where nor- Etiology
mal choroid plexus can be found . In contrast to choroid plexus No differences have been established between the etiology of
papillomas (CNS WHO grade 1), which occur in the supratento- atypical choroid plexus papilloma and that of choroid plexus
rial and infratentorial regions with nearly equal frequency, atypi- papilloma.
cal choroid plexus papillomas are more common in the lateral
ventricles {1463,3479). Pathogenesis
Genetic profile
Clinical features Atypical choroid plexus papillomas are genetically highly similar
Like choroid plexus papillomas, atypical choroid plexu s papil- to choroid plexus papilloma but different from choroid plexus car-
lomas tend to block cerebrospinal fluid pathways, and patients cinoma (2079}. Consistent with the entity's defining histological
present with hydrocephalus and raised intracranial pressure. feature (i.e. increased mitotic activity), a higher RNA expression

Fig. 3.04 Atypical choroid plexus papilloma. A Increased mitotic activity in an otherwise well-differentiated papillary tumour. 8 Increased proliferative activity (Ki-67 immuno-
histochernistry).
. . . I horoid plexus papilloma
of cell cycle- related genes was found in atypi cal choroid plexus Box3.02 Diagnostic criteria for atypica c
papilloma than in choroid plexus papilloma 11455).
Essential:
Macroscopic appearance lntraventricular or cerebellopontine angle location
lntraoperative observations in atypical choroid plexus papi llo- AND · I d·
rnas demonstr~te_ a highly va~cular tumour with a propensity to Demonstration of choroid plexus differentiation by histopatholog1ca an immu-
bleed 13107). similar to choroid plexus papillomas . nophenotyplc fea tu res
AND 2
.
Histopathology Demonstration of<! 1 mitosls/mm 2 in a minimum of 2·3 mm (equating to
Atypical choroid plexus papilloma is a choroid plexus papilloma <! 2 mitoses/1 0 HPF of 0.23 mm2)
with increased mitotic activity. A mitotic count of 2: 1 mitosis/mm2 AND
(equating to 2: 2 mitoses/10 HPF of 0.23 mm 2) has been used to Absence of criteria qualifying for the diagnosis of choroid plexus carcinoma
establish this diagnosis 11463,2927) . One or two of the following
four features may also be present : increased ce llularity, nuclear Desirable:
pleomorphism , blur~ing of the papillary pattern (solid growth), . of hyperp 101'dYbYge name-wide chromosomal
In select cases: demonstration
and areas of necrosis; however, these features are not requ ired copy-number analysis
for a diagnosis of atypical choroid plexus papilloma.

tmmunophenotype Staging
The expression pattern corresponds to that of cho roid pl exus Not relevant
papill oma . Prognostic correlates have been described for vari -
ous antigens, such as S100, transthyretin, and CD44, but these Prognosis and prediction .
markers are not helpful in grading choroid plexus tumours [n The 5-year overal l su rvival and event-free survival rates_for atyp-
individual cases. The median Ki-67 in dex is 9.1% 13479]. ical choroid plexus papillomas (89% and 83%, respective ly) are
intermediate between those for choroid plexus papil loma ana
Cytology choroid plexus carcinoma 13479] . In a series of _1~4 atypica,
Cytological features are similar to those of choroid plexus papil - choroid plexus papill omas, increased mitotic act1v1ty was the
lornas 11641]. only histological feature independently associated with recur-
rence . Tumours with 2: 1 mitosis/mm 2 in a minimum of 2.3 mrr.-
Diagnostic molecular pathology (equating to 2: 2 mitoses/ 10 HPF), which constituted the defi-
Atypical choroid plexus pap illomas are recognized by their nition of atypical choroid plexus pap illoma, were 4 .9 times as
histology. Genome-wide chromosomal copy-number analysis likely to recur after 5 years of fo llow-up as were those with lower
can demonstrate ch aracteristic hyperploidy {1455}. which may mitotic counts 11463). Children ag ed< 3 years harbouring atypi-
be helpful in th e diagnostic differentiation from choroid plexus cal choroid plexus papil loma seem to have a good prognosis
carcinomas. {3183]. In older patients , cho roi d plexus papilloma is more likely
to recur; there is evidence that the diagnosis of atypical cho-
Essential and desirable diagnostic criteria roid plexus papilloma is prognostically re levant in children agea
See Box 3.02 . > 3 years and adults.

194 Choroid plexus tumours


Choroid plexus carcinoma Pietsch T
Hasselblatt M
Malkin D
Paulus W

Definition
Choroid plexus carcinoma (CPC) Is a malignant epithelial neo-
plasm of the choroid plexus that shows at least four of the fol -
lowing five histological features: frequent mitoses , increased
cellular density, nuclear pleomorphism, blurring of the papil -
lary pattern with poorly structured sheets of tumour cells , and
necrotic areas (CNS WHO grade 3).
I
.
.

ICD-0 coding
9390/3 Choroid plexus carcinoma

ICD-11 coding
2AOO & XH3M77 Primary neoplasms of brain & Choroid plexus
carcinoma

Related terminology Fig. 3.05 Choroid plexus carcinoma. A This T1 -weighted coronal MRI of a 5-year-old
None girl shows a contrast-enhancing tumour related to the lateral ventricle. B Axial T2-
weighted MRI of a 5-year-old girl.
Subtype(s)
None

Localization
Most CPCs are located in the lateral ventricles {1786) .

Clinical features
CPCs tend to block cerebrospinal fluid pathways and cause
symptoms related to hydrocephalus, such as increased intra-
cranial pressure, increased head size, nausea, and vomiting
{259).

Imaging
On MRI , CPCs typically appear as large intraventricular lesions
with irregular enhancing margins , a heterogeneous signal on
T2- and T1-weighted images, oedema in adjacent brain, hydro-
cephalus , and disseminated tumour {2100).

Epidemiology Flg.3.06 Choroid plexus carcinoma. A large choroid plexus carcinoma in the lateral
ventricle with extensive invasion of brain tissue.
In the SEER database, CPCs accounted for 34.4% of choroid
plexus tumours (455}. About 80% of all CPCs occur in children .
known to play a crucial role in the prolife ration of these cells
Etiology 11876). About 50% of CPCs carry TP53 mutations. In > 90<Jo of
Most CPCs occur sporadically, but about 40% occur in the con- TP53-wildtype CPCs , the combination of the TP53 p R72 vari-
text of Li-Fraumeni syndrome with germline TP53 pathogenic ant and the MDM2 SNP309 polymorphism , wh ich is associated
sequence variants 11746,3097}. It is recommended that any with reduced TP53 activity, was observed (30971, implicating
patient with a CPC, and their family, be offered genetic counsel- p53 dysfunction in virtually all CPCs . TP53 mutatio ns in CPC
ling and testing for TP53 germline mutations (1134,336) . CPC are associated with increased genomic instability (30 97), with
has also been described in Aicardi syndrome 13103). aneuploidy demonstrable by both classic cytog enet1c and
genome-wide array-based approaches (2079 ,2669,3575,2748 ,
Pathogenesis 1455). These complex chromosomal alterations are related
CPCs are believed to derive from monociliated progenitors of to patient age (27481. with ch ildhood CPC s showing marked
plexus epithelium located in the roof plate, and they show acti- hypodiploidy (1455) . TAF12, NFYC, an d RA0 54L oncogenes ,
vation of the sonic hedgehog and Notch signalling pathways within chromosomal gains at 1p35.3-p32, cooperate in diseRse

,.
Q ..
•'I
I

' ••
,,

~ .

••••
~ .


""
C.J
/·· .,
,,
....•
, • .. •
.
I


.:

• •

•• "
Rg.3.07 Choroid plexus carcinoma. A Increased cellularity, blurring of the papillary pattern, and necrosis. B Increased cellularity, nuclear pleomorphism , and mitotic ac!N-
ity. C lmmunohistochemistry for transthyretin highlights infiltration of surrounding brain tissue. D Ki-67 immunohlstochemlcal staining shows high proliferative activity.

initiation and progression and suggest potential therapeutic tar- Histopathology


gets !321 4}. CPCs demonstrate frank signs of malignancy, defined as
showing at least four of the following five histolog ical features
Macroscopic appearance increased cellular density; nuclear pleomorphism ; blurring of the
CPCs are highly vascular tumours that may appear solid, haem- papillary pattern with poorly structured sheets of tumour cells.
orrhagic, or necrotic, and that show invasive growth . necrotic areas; and frequent mitoses , usually > 2.5 mitoses/mm:

. . .. .. ~
, ...
~· .,_;,
wr · , • •
_..._. t , - ' !r,J- • .....
._.... -"
_. •
flg.3.08 Choroid plexus carcinoma. A lmmunohistochemistry for cytokeratln is positive. B lmmunohistochemisty for transthyretin (prealbumin). c lmmunohistochemis1ry
demonstrates a lack of expression of epithelial membrane antigen (EMA).

196 Cho roid plexus tumours


(equating to > 5 mitoses/10 HPF of 0.23 mm 2 ) 11463,2927). Dif-
fuse brain invasion is common . ' .
tmmunophenotype •
11 Like choroid plexus papillomas , CPCs express cytokeratins, but
i)
they are less frequently positive for S100 and transthyretin . There
is usually no membranous positivity for EMA . Distinct membra-

I
nous staining for the potassium channel Kir7.1 is seen in about
50% of CPCs. Nuclear accumulation of p53 has been reported in
CPCs that harboured TP53 mutation {3097) . CPCs retain nuclear '
positivity for SMARCB1 and SMARCA4. The median Kl-67 index
is reported as 20.3% (range: 7.8-42.5%) (3479).

Cytology
I
'I On touch preparation, CPC cells show high nuclear variance
and a high N:C ratio.

Diagnostic molecular pathology


CPCs can usually be identified by histological and immunophe-
notypic analysis. Genome-wide chromosomal copy-number Fig. 3.09 Choroid plexus carcinoma. Touch preparation shows epithelioid cells with
analysis shows complex chromosomal alterations and charac- pleomorphism.
teristic hypoploidy (1455], which may be helpful in the diagnos-
tic differentiation from atypical choroid plexus papillomas . TP53
mutations can be found in about 50% of cases by sequenc- Box3.03 Diagnostic criteria for choroid plexus carcinoma
ing . Methylome analysis has revealed three clinically distinct Essential:
subgroups of choroid plexus tumours, with all CPCs clustering Demonstration of choroid plexus differentiation by histopathological and immu-
within the same subgroup, together with prognostically unfa- nophenotypic features
vourable grade 1 and 2 choroid plexus tumours (3184]. AND
Presence of at least four of the following five histological features:
Essential and desirable diagnostic criteria
• Increased cellular density
See Box 3.03 .
• Nuclear pleomorphlsm
• Blurring of the papillary pattern with poorly structured sheets of tumour cells
1 Staging • Necrotic areas
Patients with CPC present with metastasis at diagnosis in 21% • Frequent mitoses, usually > 2.5 mitoses/mm 2 in a minimum of 2.3 mm2
of cases {3479], so investigation for the presence of metastases (equating to> 5 mitoses/1OHPF of 0.23 mm2)
is recommended . AND
lntraventricular location
Prognosis and prediction
The 5-year progression-free and overall survival rates in patients Desirable:
with CPC have been reported as 38% and 62% , respectively TP53 mutation analysis
(3576}. The extent of surgery has a significant impact on sur- Methylatlon profile of choroid plexus carcinoma
vival (455}. Several studies have suggested that the presence of In select cases: demonstration of hypoploidy by genome-wide chromosomal copy-
TP53 mutations, as identified by immunohistochemical staining , number analysis
is associated with a less favourable outcome (1157,3097,3576}.

Ch oro1li pl xus 1u1 ~l)L 1 r s 197


Ellison OW
Medulloblastoma: Introduction Taylor MD

M ed ullob_l a st~m as . display considerable biolog ical heteroge- Medulloblastomas occur in the setting of several inherited
neity. which 1s evi dent across the d iverse types of molecu- cancer syndromes 13392}. Germline mutati ons can occur 1n
larly defin ed medullob lastomas li sted in this classification and ELP1 {3393 }, SUFU and PTCH1 (naevoid bas~I ce ll carcinoma
among the morpholog ical patterns shown by these tumours . syndrome / Gorlin syndrome) {1381}, TP53 (L1 - Frau~en1 syn.
drome) 12331), APC (fam ilial adenomatous polypos1s) 116161,
Medulloblastoma as a unitary disease CREBBP (Rubinstein- Taybi syndrome) 1339}, NBN (NBSJ )
Medulloblastoma can arise at all ages but most commonly (Nijmegen breakage syndrome) 11366}, PALB2, and BRCA2,
occurs in childhood. It is the second most common CNS among others 13392,3234). .
malignant tumour in childhood , after high-grade glioma, and it Medulloblastomas grow into the fourth ventricle or are located
accounts for approximately 20% of all intracranial neoplasms in the cerebellar parenchyma [293) . Some cerebellar tumours
in this age group {1604,2344}. The annual overall incidence of can be laterally located in a hemisphere, and almost all of these
medulloblastoma is 1.8 cases per 1 million population , whereas belong to the sonic hedgehog (SHH)-activated molecu lar group
the annual chil dhood incidence is 6 cases per 1 million. These {3164) . Wingless/INT1 (WNT)-activated medulloblastomas are
rates have not changed over time [2406) . thought to arise from cells in the dorsal brainstem 11099,14721.
The median patient age at d iagnosis of medulloblastoma is although not all brainstem embryonal tumours are WNT-acti-
9 years, with peaks in inc idence at 3 and 7 years of age 12686} . vated medulloblastomas .
As many as one quarter of all medulloblastomas occur in adults , All types of medulloblastomas are considered to be embry-
but < 1% of adult intracranial tumours are medulloblastomas onal tumours and CNS WHO grade 4, even though some
{2075}. The tumour has an overall M:F ratio of 1.7:1. molecular groups and subgroups of medulloblastoma, such as
As with other high-grade brain tumours , the incidence of WNT-activated tumours, show a very good response to current
medu lloblastoma differs across ethnic groups . In the USA, therapeutic regimens and almost all of these patients can be
overall annual incidence is highest among White non-Hispanic cured . Small, poorly differentiated cells with a high N:C ratio
people (2.2 cases per 1 million population) , followed by among and high levels of mitotic activity and apoptosis dominate the
Hispanic people (2.1 per 1 mill ion) and African-American peo- histopathology. However, architectural and cytological diver-
ple (1 .5 per 1 million) {2344} . sity can manifest as nodule formation , neurocytic or ganglion

T1ble4.01 Characteristics of medulloblastoma molecular groups


Medulloblutoma molecular group
-------
SHff.lctlvltld SHH-actfvated Non-WNT/non-sHH Non·WNT/non-SHH
WN1Ctlvlted group3 group4
1P53-Wlldtype T'P53-nlJtlnt
--~~~~~~~~~~~~~~~---

Subgroups SHH-1 to SHH-4 SHH-3 Group 3/4 subgroups 1-8


RelatJVJ frequency 10% 20% 10% 25% 35%
Predominant
Childhood Infancy/adulthood Childhood Infancy/childhood All age groups
age group
M:Fratio 1:2 1:1 3:1 2:1 3:1
Predominant morphology Classic Desmoplastlc/nodular Large cell / anaplastic Classic Classic
MYC, MYCN MYCN, OTX2
Freq11ent COP¥-number MYCN amplification;
PTCH1 deletion; amplification; 1q, amplification; CDK6
•Iterations Monosomy 6 GL/2 amplification;
10q loss 7 gain; 1Oq, 16q loss; amplification; 7 gain; B.
17p loss
isodicentrlc 17q 11 loss; isodicen!Jic t 7q

GF/1, GF/18 activation ; GF/1 , GF/18 activation;


Frequent genetic PTCH1, SMO, SUFU, TP53, DDX3X, PRDM6 activation;
CTNNB1 , DDX3X SMARCA4, K8T804,
alterations ELP1 , DDX3X, KMT2D, U1 snRNA, TERT KDM6A, ZMYM3,
mutation CTDNEP1, KMT2D
U1 snRNA mutation mutation KMT2C. KMT20,
mutation
K8TBD4 mutation
Ge.neswlth
germline mutation APC PTCH1. SUFU, ELP1 TP53 Rare BRCA2, PALB2 Rare BRCA2, ~LB2
-
SHH, sonic hedgehog; snRNA, small nuclear RNA.

200 Embryonal tumours


cell differentiation , or even myogenic and/or melanotic differ-
entiation 1971 ,2057,2770 ,2497,2973) . Such varied morphologi-
ca l fe atures can be seen across medulloblastoma molecular C:HH~ ~: •

~ ~· :ww,..
groups.

Molecular heterogeneity
Medulloblastomas should now be classified according to a
combination of molecular and histopathological features . Their
$HH f "•
---.,.1':.71
~~"" ..
.
• . •·'..F. •• .
\\:
·. ).... ..')!. . • .
'
• •

molecular classification reflects biological heterogeneity that


can be demonstrated by the clustering of medulloblastomas
into groups using transcriptome or DNA methylation profiling [ non-~Tinon-Sl-fH _1_
12280). Initially, consensus that was built upon several datasets non-"'°"'Tl non-SHH 2
established four principal molecular groups: WNT-activated , r ~ '
·~,- - -c _1

SHH-activated , group 3 , and group 4 13153). Tumours in the


WNT and SHH groups show activation of their respective cell
signalling pathways . WNT and SHH medulloblastomas were
included in the 2016 WHO classi"fication of CNS tumours, and
SHH tumours were divided on the basis of TP53 status (TP53- -so
mutant and TP53-wildtype tumours having very different ciin-
icopathological and biological characteristics). Non-WNT/non-
SHH medulloblastomas comprise group 3 and group 4 tumours
- 100
(see Table 4.01) .
These groups are represented in the current classification ;
however, new subgroups have emerged at a more granular level , Fig. 4.01 Medulloblastoma molecular groups and subgroups. Unsupervised,
within the four principal molecular groups, having been discov- non-linear !-distributed stochastic neighbour embedding (t-SNE) projection of
ered through the analysis of large numbers of tumours 12865, methylation array profiles of 1089 medulloblastoma samples. Samples were se-
491 ,2279 ,2900,1757,1355). These new subgroups are intro- lected from a large database of > 50 000 brain tumour datasets to serve as refer-
duced in the sections on molecularly defined medulloblastomas ence profiles for training a supervised classification model based on strict criteria:
that follow. There are four subgroups of SHH medulloblastoma all these samples showed a high calibrated classification score (> 0.9) for medul-
loblastoma methylation class when applying the brain tumour classifier available at
and eight subgroups of non-WNT/non-SHH medulloblastoma
https://www.molecularneuropathology.org. In a t-SNE projection like this, occasional
12865,491 ,2900 ,1757,1355). Like the four principal molecular samples of one subclass may fall closer to a different subclass because of the sto-
groups of medulloblastoma, some of these subgroups are asso- chastic nature of the t-SNE algorithm and the gradual, rather than strict, boundaries
ciated with clinicopathological and genetic features that provide between the subclasses.

Group

Subgroup

Related SHH-I, SHH-beta, SHH-II, SHH-gamma,


SHH-alpha, SHH-child SHlttlel1a, SHH-adult
terminology SHH-Infant SHH-Infant
71'53-wlldtype I TP53-rn.dldad
Frequency 15-20% 15-20% 20-25% 10-15% 30-35%

...t...t..tt
f'
L
) { { I(:tttf
d-,, , r·
''~ 1\_
:
I\ ( - :
I

/I
l
,\

I '~ I' - I
,. I'

Sex d'~ d'd' ~~~ d' ~ ! d'd'd'd' ~ d' d' d' d' ~~~

Histology Oesmoplaslic > Classic DesmoplastidM8EN > Classic Classic > LCA : LCA > Classic Desmoplastic > Classic

heterogeneous prognosis good pmgnosls good plO!JlOSls ; poor prognosis tntennedlale prognosis

\/
K
PTCHl,SUFU PTCH1, SUFU
... \\ i
I ~ !~
PTCHl, EL.P1, : TP53, DDX3X.
DDX3X, : UI snANA.
10q-
14q-
~1

UI snRNA, TERT. PTCH1,


~
~ 10q-
14<t-

mutatk>ntdeletlon mutalion/dolelioo ~ : TERT11VJlalk>n DDX3X. SMO, CREBBP,


SMO, KMT2D muta.Uon SMO mulaJlon : MYCN, GU2 GSE1, FBXWl roolallon
PPM1D
a.~lficallon ~liftalllon

Flg.4.02 SHH-activated medulloblastoma subgroups. Demographic, clinical, and molecular features of the four molecular subgroups of SHH-activated medulloblastoma. LCA,
large cell I anaplastic; MBEN, rnedulloblastorna with extensive nodularity; SHH , sonic hedgehog.
Group

Sul>groop

8-10% 15-20%
Frequency 3-5% 10-15% 10-15% 8-10% 8-10%

Age
t ~tttf ..ti j ii j ittf ttHk
&&& ~ &&& ~
Sex cM& CflCfl &&& Cfl &&& Cfl && ~ ~ M ~ &J

Classic> LCA Classic Class le


Hlstology Classic LCA, Classic Classic > LCA Classic Classic

f t
' '· x '
Me!astasls

- 85% - 75%

,.x
5-yearOS - 75% - 55% - 45% - 85% - 60% - 80%

Cytogenetlcs
x 1q+ 117q

'·x 117q
10q-
16q- 'X 117q

16q-
117q
8-
11-
7+ I

I
Ji
117q

,a-
X,,
balanced

MYCamplilication
PRDM6 actlvafon
GF/1/GFNB acti\'alion GF/1/GF/18 activation PRDM6 actJvatlon
MYC, MYCN MYC. MYON KBTBD4 mutaUon
Unknown KDM6A, ZMYM3,
OTX2 ampllftcatlon KBTBD4, SMARC\4, ampllflcaUon ampllflcallon M YCN ampllllcatlon KM T2C mutation
CTDNEP1, KMTW
mutatlon

Flg.4.03 Non-WNT/non-SHH medulloblastoma subgroups. Demographic, clinical, and molecular features of the eight molecular subgroups of group 3/4 medulloblastomas. LCA,
large cell I anaplastic; OS, overall survival; SHH, sonic hedgehog.

clinical utility, either being of diagnostic or prognostic value or aligned to a particular molecular group - such as PTCH1 (SHH-
having implications for therapy. One example is the delineation activated group), CTNNB1 (WNT-activated group), and MYC
of two SHH subgroups, SHH-1 and SHH-2, both dominated (non-WNT/non-SHH subg roup 3) - are found to be altered at a
by medulloblastomas from young children {2691 ,1355,1303A}. relatively high frequency when sought in large tumour cohorts.
Cases in these subgroups have statistically significantly differ- Although other genetic alterations might be recorded at low fre-
ent outcomes, and recent clinical trial data suggest that spe- quency, they often converg e on key biolog ical pathways, such
cific chemotherapeutic regimens can help those patients with as histone modification {2282) .
tumours in the poorer prognosis subgroup (SHH-1) {2186 ,2690,
1303A). Integrated diagnosis
The histopathological classification of medulloblastomas A classification listing mol ecularly defi ned medulloblastomas
listed in the 2016 WHO classification of CNS tumours , compris- while also recognizing morp hological patterns with clinicopatho-
ing four morphological types (classic, desmoplastic/nodular, logical utility is intended to encourage an integrated approach
medulloblastoma with extensive nodularity, and large cell I to diagnosis {1939) . A combination of molecular analysis (e.g.
anaplastic), has now been combined into one section that DNA methylation profiling) and morpholog ical interpretation pro-
describes the morphological variation as patterns of a single vides optimal prognostic and pred ictive information . Integrating
tumour type: medulloblastoma, histologically defined . The mor- Information on genetic alterations further enhances the level ol
phological patterns have their own specific clinical associations diagnostic precision , for example by allowing SHH-activated
1818,2056,2057,2031 ). and molecularly defined medulloblas- medulloblastomas to be divided into tumours with wildtype or
tomas demonstrate specific associations with the morphologi- mutant TP53. Other genetic alterations currently used in the
cal pattern s. All true desmoplastic/nodular medulloblastomas risk stratification of medulloblastomas but not included in the
and medulloblastomas with extensive nodularity al ign with the classification , such as MYC am plification , could also be placed
SHH molecular group {837). and most are in the SHH-1 and into an integrated diagnosis to enhance precision . An integrated
SHH-2 subg roups (1355}. Nearly all WNT tumours have classic approach to diagnosis , with commentary, also offers an oppor-
morphology, and most large cell / anaplastic tumours belong tunity to list the methodolog ies used to provide molecular results
either to the SHH-3 subgroup or to the non-WNT/non-SHH (i.e. and to highlight th e clinical significance of germline mutations
group 3/4) subgroup 2 {1757). when a medullobl astoma arises in the setting of a hereditary
Recent studies have described the detailed genomic land- tumour syndrom e, such as naevoid basal ce ll carcinoma syn-
scape of medulloblastoma {2279,2283). Some driver genes drome (Gorlin syndrome) or Li- Fraum eni syndrome.

202 Embryonal tumours


Medulloblastoma, WNT-activated Ellison OW
Clifford SC
Kaur K
Korshunov A
Northcott PA
Taylor MD

Definition
Imaging h
Medulloblastoma, WNT-~ctivated, is an embryonal tumour aris- Neuroimaging of WNT-activated medulloblastomas s o.w s
ing from th~ dorsal bra1nstem demonstrating activation of the tumours located in the cerebellar midline or cerebellopon tine
WNT signalling pathway. angle , with many in close con tact with the brainstem !1099 f.
WNT-activated medulloblastomas have a relatively porous
ICD-0 coding blood- brain barrier when compared with other types of medul-
9475/3 Medulloblastoma, WNT-activated loblastoma and therefore enhan ce very brig htly 125001 .

ICD-1 1 coding Spread


2A00.10 & XHOZP6 Medulloblastoma of brain & Medulloblas- It is rare tor a patient with a WNT-activated medul lobl astoma to
toma, WNT-activated . NOS present with leptomeningeal metastases .

Related term inology Ep idemiology


None WNT-activated tumours account for about 10% of all medul-
loblastomas (841 ,1022,25091. They typically occur in children
Subtype(s) aged between 7 and 14 years , and they also account for
None 15- 20% of adult medulloblastomas 13472}. Slightly more female
than male patients have this type of medullo? l ~stoma . WNT-
Localization activated medulloblastomas hardly ever occur 1n infants (1757}.
WNT-activated medulloblastomas are located either around the
toramen of Luschka, appearing to arise from the brainstem or Etiology
cerebellum. or in the cerebellar midline, generally contiguous The vast majority of WNT-activated medulloblastomas are spo-
with the brainstem 11099,3164) . Extension towards the cerebel- radic and little is known about their etiology. A rare subset of
lopontine angle or cerebellar peduncle is noted in a clinically WNT~ activated medulloblastomas are diagnosed within the set-
significant number of cases (2463,2415,6201. ting of constitutional mismatch repair deficiency s~ndro.n:ie , in
individuals with germ line APC mutations and a pred1spos1t1on to
Clinical features colon cancer 13392,3076}.
Most patients present with symptoms and signs of raised intra-
cranial pressure from non-communicating hydrocephalus due Pathogenesis
to occlusion of the fourth ventricle by the primary tumour. WNT-activated medulloblastomas transcriptionally resemble
normal progenitor cells from the lower rhombic lip-derived
mossy fibre neuron lineage, consistent with an extracerebellar
(dorsal brainstem) origin 11099,14721. The DNA methylation pro-
file and transcriptional signatures of these tumours , which might
be the best evidence for the cell of origin in human tissues .
indicate that WNT-activated medulloblastoma has a profile dis-
tinct from those of tumours in other medulloblastoma molecular
groups (1356 ,2509,491 ,2279} .

Genetic profile
Large next-generation sequencing studies have confirmed that
86-89% of WNT-activated medulloblastomas harbour somatic
mutations in exon 3 of CTNNB1 (2284,2279 ,33921 . Among
WNT-activated medulloblastomas lacking somatic CTNNB1
mutations , most arise in children carrying pathogenic germ-
line APC mutations (33921. Other genes with somatic mutations
in WNT-activated medulloblastomas include those encodi ng
subunits of the SWl/SNF nucleosome-remodell ing complex
(SMARCA4 , AR/01A, AR/02; in 33% of cases) , ODX3X (in 36 %),
CSNK2B (in 14%) , TP53 (in 14%), KMT20 (in 14%), and PIK3CA
Flg. 4.04 Medulloblastoma, WNT-activated MRI of WNT-activated medulloblastoma
(in 11 %) [22791 . Cytogenetically, monosomy 6 occurring on the
arising in the cerebellopontine angle. background of an otherwise diploid genome is a characteristic

Embryonal tumou rs 203


genetic feature of . WNT-activated medulloblastomas and is
Box4.01 Diagnostic criteria for medulloblastoma, WNT-activated
observed 1n approximately 83% of cases (609,1703,2279).
Essential:
Macroscopic appearance A medulloblastoma
Medulloblastomas appear as friable pink masses . At s AND
· h . . urgery,
1ntratumoura 1 aemorrhage 1s particularly associated with WNT- WNT pathway activation
activated medulloblastomas {2500).
OR
Histopathology A DNA methylation profile aligned with medulloblastoma, WNT-activated

Nearly all WNT-activated medulloblastomas have a classic


morphology. Anaplastic WNT-activated tumours have been
commonly diploid for chromosome 6 and arise in older children
reported but are rare {837,1575). Desmoplastic/nodular medul- and young adults.
loblastomas do not occur in this group.
Essential and desirable diagnostic criteria
lmmunophenotype
See Box 4.01 .
Activation of the WNT pathway can be demonstrated by univer-
sal or patchy J3-catenin immunoreactivity in tumour cell nuclei Staging
{837,1576). Medulloblastomas in other molecular groups show Clinical staging procedures include MRI examinations of the
cytoplasmic expression of J3-catenin . CNS with contrast agent. This is complemented by lumbar
puncture postoperative cerebrospinal fluid cytology. The post-
Grading operative staging system developed by Chang and others in
WNT-activated medulloblastomas are assigned CNS WHO 1969 {519), which defines the following degrees of metastatic
grade 4. spread, is still being used :

Cytology MO No evidence of subarachnoid or haematogenous


Evaluation of the presence of tumour cells in cerebrospinal fluid metastasis
is required for staging. M1 Microscopic tumour cells found in the cerebrospinal fluid
M2 Gross nodular seeding demonstrated in the cerebellar/
Diagnostic molecular pathology cerebral subarachnoid space or in the third or lateral
CTNNB1 exon 3 mutations and monosomy 6 on the background ventricles
of a diploid genome are present in > 80% of WNT-activated M3 Gross nodular seeding in the spinal subarachnoid space
medulloblastomas {609}. These alterations have been used to M4 Metastasis outside the cerebrospinal axis
identify WNT-activated medulloblastomas, but DNA methyla-
tion profiling is considered the standard method for determin- Prognosis and prediction
ing medulloblastoma group or subgroup status {1356,2866}. The prognosis of children with WNT-activated medulloblastoma
Array-based DNA methylation analysis, array- and sequenc- is excellent despite the CNS WHO grade; with current surgical
ing-based RNA expression analysis, NanoString analysis, and approaches and adjuvant therapeutic regimens , the overall sur-
minimal methylation classifier assays have all been used for vival rate is close to 100% {841 ,2283) The excellent outcome
assignment of molecular group {2864,1715). In addition, immu- is expected for tumours with germline APC mutations, as well
nohistochemistry can be used to discriminate between WNT, as those with CTNNB1 mutations {3076) . Adult patients with
sonic hedgehog (SHH), and non-WNT/non-SHH medulloblas- WNT-activated medulloblastoma do not have such a favourable
tomas (837) . outcome (2649,2913 ,1148). Unlike TP53 mutations in SHH-
Two molecular subgroups of WNT-actlvated medulloblastoma activated medulloblastomas, TP53 mutations in WNT-activated
have been suggested: WNT-a and WNT-P {491) . WNT-a tumours medulloblastomas. which are all somatic and most commonly
show monosomy 6 and arise in children. WNT-p tumours are heterozygous , do not confer a poor prognosis (3614) .

204 [1 ri b r 1011r.i l tu r nours


Medulloblastoma, SHH -activated Ellison OW
Clifford SC
and TP53-wildtype KaurK
Korshunov A
Northcott PA
Taylor MD

Definition
to the pia mater. Mechanisms of metastasis for SHH-activated
Medulloblastoma. SHH-activated and TPSJ . d . medulloblastoma are unclear, with spread to the leptomeninges
b t -w1 type , 1s .an
1

em ryon~1 umour of the cerebellum demonstrating activatlon through the cerebrospinal fluid (CSF) or via a haematogenous
of. the s~rnc hedgehog (SHH) signalling pathway in combination route with return to the leptomeninges [1044). The molecular
with a w1ldtype TP53 gene (CNS WHO grade 4) . groups of medulloblastoma . including SHH-acyvated tumours.
have been shown to remain stable in comparisons of primary
ICD-0 coding and metastatic lesions {3382) . Whereas non-WNT/non-SHH
9471/3 Medulloblastoma. SHH-activated and TP53-wildtype medulloblastomas occur almost exclusively with distant CNS
metastases at the time of recurrence , a large proportion of SHH
ICD-11 coding medulloblastomas demonstrate isolated recurrences in the
2A00.10 & XH9M38 Medulloblastoma of brain & Medulloblas- tumour bed (2614,1309A).
toma, SHH-activated and TP53-wildtype
Epidemiology
Related terminology SEER data from 1973-2007 show annual medulloblastoma inci-
None dence rates of 6 cases per 1 million children aged 1-9 years
and 0.6 cases per 1 million adults aged > 19 years (2972).
Subtype(s) SHH-activated medulloblastomas in general show a bimodal
SHH-activated medulloblastomas comprise four provisional age distribution , being most common in infants and adults, with
molecular subgroups (SHH-1, SHH-2, SHH-3 , SHH-4), which an M:F ratio of approximately 1.5:1 (2280,1757).
can be demonstrated by DNA methylation or transcriptome pro-
filing (see Fig . 4.02, p. 201). Etiology
There are several hereditary tumour syndromes that predispose
Localization to the development of SHH-activated medulloblastoma (3392).
SHH-activated medulloblastomas arise in the cerebellar hemi- The canonical inherited syndrome associated with SHH-acti-
sphere or vermis and can sometimes involve both structures. vated and TP53-wildtype medulloblastoma is naevoid basal
However, localization of this tumour type is related to age. cell carcinoma syndrome (Gorlin syndrome). Medulloblastomas
Tumours in infants frequently involve the vermis , whereas hemi- in the setting of naevoid basal cell carcinoma syndrome are
spheric tumours are relatively infrequent in this age group . In always classified in the SHH molecular group, and most are
older chil dren and young adults , SHH -activated medulloblas- due to inactivating germline mutations in PTCH1 , the gene that
tomas arise mainly in the cerebellar hemispheres {1099,2463 , encodes the receptor for the SHH protein . Naevoid basal cell
3164,3405 ,3601) . carcinoma syndrome due to a SUFU or PTCH2 mutation is rare .
Germline SUFU mutations are largely restricted to infants, who
Clinical features exhibit developmental anomalies and predisposition to addi-
Most patients present with symptoms and signs of raised intra- tional malignancies. Germline mutations in ELP1 , which is close
cranial pressure from non-communicating hydrocephalus due to PTCH1 on chromosome 9q, have also been reported in SHH -
to occlusion of the fourth ventricle by the primary tumour. activated medulloblastoma !3393). Heterozygous germ line
mutations in GPR161 are exclusively associated with SHH-acti-
Imaging vated medulloblastoma and account for approximately 5% of
Neuroimaging shows SHH-activated medulloblastomas as subtype 1 tumours [232). The frequency of germline mutations
solid , intensely contrast-enhancing masses . Oedema was rela- in patients with SHH-activated medulloblastoma is estimated to
tively common in one imaging series that included 12 desmo- be ?: 40% (3393) .
plastic/nodular medulloblastomas and 9 medulloblastomas with
extensive nodularity (MBENs) {991). A grape-like pattern on MRI Pathogenesis
generally characterizes MBEN {1088,2197). Rarely, medullo- Cell of origin
blastomas involving the lateral cerebellum occur as extra-axial SHH-activated medulloblastomas are thought to derive from an
masses resembling meningiomas or acoustic nerve schwanno- ATOH1-positive cell in the external granule cell lineage of the
mas {230) . cerebellum {2860,3526) . These cells are unusual among neu-
rons in that they continue to divide after birth (3402). The m1to-
Spread gen that ~rimarily drives the expansion of the external granule
Medulloblastomas have the potential to invade locally, metasta- ~ell layer is the SHH protein, and many of the mutational events
size to the leptomeninges , or (rarely) spread outside the CNS . in this type of medulloblastoma lead to constitutive SHH activa-
Most metastases are found on the surface of the CNS, attached tion (1702) .

Embryonal tumou rs 205


Genetic profile latter is rare (837,3153). However, rec~rrence of desmoplast1c1
Germline or somatic mutations in SHH signalling pathway nodular SH H-ac tivated and TP53-w1ldtype tumours can he..
genes are cha racteri stic of most SHH -ac tivated medulloblas- associated with transformation to a focal anaplastic morph1;1:
tomas and cause SHH pathway activation . They include muta- ogy [1718) .
tions in PTCH1 (-4 0% of tumours) and SMO (- 10%), the protein
products of which form the receptor for the SHH protein , and lmmunophenotype
in SUFU (-10%), a cytoplasmic mediator of GLI transcription A panel of immunohistochemical markers can be used to icJs 11
factors . Amplifications of GL/1 or GL/2 (- 10%) and other down - tify SHH -activated tumours among medulloblastomas (but nr,·
stream SHH target genes (MYCN, MYCL , and YAP1 ; < 10%) other embryonal tumours) {837,15761 . SHH-activated turr.our-:
have also been found 12279). Alterations involving known SHH express GAB1 . Both SHH-activated and WNT-activated rned•).
pathway genes were reported in 116 (87%) of 133 SHH -acti - loblastomas express YAP1 , but SHH-activated medulloblas-
vated tumours in one study {1702) . tomas do not show nuclear immunoreactivity for P-caten1r,
Other genes commonly mutated in SHH -activated medullo- Typically, weak to moderate nuclear immunoreactivity for p53 ·s
blastoma , but not directly involved in the SH H signalling path- present in a few scattered tumour cells when TP53 1s wildtype
way, include DDX3X (-20%) , KMT20 (10-15%), and CREBBP
(-10%) {2279). Non-cod ing somatic TERT promote r alterations, Cytology
which affect telomere maintenance, are freq uent among non- Evaluation of the presence of tumour cells in CSF is required
infant SHH medulloblastomas. Most adult tum ours (> 80%) for staging .
harbour TERT promoter mutati on s, compared with about 15%
and 20% of ~umo u rs in infants and children , respectively. Diagnostic molecular pathology
Hypermethylat1 on of the TERT promoter is seen in most child- DNA methylation profiling is considered the gold-standard
hood SHH -activated medulloblastomas without TERT promoter method for determining medulloblastoma group or subgrouo
mutation, and both TEAT-mutant and TERT-wildtype SHH- status {1356,2866) . Array-based DNA methylation analy-
activated medulloblastomas are associated with elevated TERT sis, array- and sequencing-based RNA expression analysis
exp ression {2650,1903). Mutations in the U1 spliceosomal small NanoString analysis, and minimal methylation classifier assays
nuclear RNA (snRNA) are found in about 15% of SHH -activated have all been used for assignment of molecular group (2864.
medu lloblastomas {3083) . Like TERT alterations, U1 mutations 1715). In addition , immunohistochemistry can be used to dis·
are found in almost all SHH medulloblastomas in adults and a criminate between WNT-activated, SHH-activated , and non-
subset of SHH medulloblastomas in adolescents , but rarely in WNT/non-S HH medulloblastomas {837}.
children or infants {3393} . Common copy-number variations TP53 sequencing allows SHH-activated medulloblastomas to
in SHH -activated medulloblastoma include losses of chromo- be classified as wildtype or mutant, and analysis of SHH path-
some 9q and 10q, which harbour the PTCH1 (9q22) and SUFU way genes (PTCH1, SMO, SUFU) provides further diagnostic
(10q24) tumour suppressor gene loci , respectively {2284} . information . TP53 mutation and MYCN amplification (and large
cell / anaplastic morphology) are important for therapeutic
Molecular subgroups stratification , as these markers are associated with a poor prog-
Four provisional molecular subgroups of SHH -activated medul- nosis among SHH-activated medulloblastomas (2865 ,22791
loblastoma can be demonstrated by DNA methylation or tran- Given the high incidence of germline predisposition among
scriptome profiling (see Fig . 4.02, p. 201) {2865,491 ,2691, patients with SHH -activated medulloblastoma, germline analy-
30 83 ,3393 ,1303A}. Two occur mainly in infants: one (SHH-1) sis of PTCH 1, SUFU, TP53 , ELP1 , and GPR161 is recommended
is enriched with somatic and germline SUFU mutations and {3392).
chromosome 2 gain, and the other (SHH-2) is characterized by
9q loss and extensive nodular morphology. The other two sub- Essential and desirable diagnostic criteria
groups arise in older patients: one (SHH-3) is associated with See Box 4.02.
TP53 and ELP1 mutations {3393}, and the other (SHH -4) occurs
mainly in ad ults and is associated with near-universal U1 and Staging
TERT mutation s and frequent somatic PTCH1 or SMO altera- Clinical staging procedures include MRI examinations of the
tions 12650,1903,1702,973,3393). Consensus on molecular CNS with contrast agent. This is complemented by lumbar
subgroup nomenclature and defining features has not yet been puncture postoperative CSF cytology. The postoperative stag-
achieved through an international cooperative meta-analysis of ing system developed by Chang and others in 1969 !5191.
SHH subgroups as it has for non-WNT/non -SHH medullobl as- whic h defines the following degrees of metastatic spread , is still
tomas {2900) . being used :

Macroscopic appearance MO No evidence of subarachnoid or haematogenous


SHH-activated and TP53-wildtype med ullobl astomas ten d to be metastasis
firm (reflecting intratumoural desmoplasia) and ci rcumscribed. M1 Microscopic tumour cells foun d in the CSF
M2 Gross nodular seeding demonstrated in the cerebellar/
Histopathology cerebral subarachnoid space or in the third or lateral
Most SHH-activated and TP53-wildtype medullob lastomas ventricles
have a desmoplastic/nodular morphology or are MBENs {837, M3 Gross nodular seeding in the spinal subarachnoid space
1722). Others are classic or large cell I anaplastic , although the M4 Metastasis outside the cerebrospinal axis

206 Ernt;ryo11al tu1 11r_1urs


. . . f d lloblastoma, SHH-activated and TP53-wildtype
Prognosis and prediction Box 4.02 01agnost1c cntena or me u
The prognosis for all SHH -activated medulloblastomas is in ter- Essential:
mediate . between those for WNT-activated and group 3 medul-
A medulloblastoma
loblastomas . However, prognosis is highly variable among
patients with SHH tumours and is associated with the specific AND
clinicopathological and molecular features . Wildtype TP53 gene
In infants, desmoplastic/nodular tumours and MBENs are AND
typically SHH -activated and TP53-wildtype (2865 ,1702]. Such SHH pathway activation
tumours are associated with favourable outcomes, even when
OR fil aJ'gned with SHH-activated medulloblastoma
radiation -sparing treatment protocols are used at presentation A DNA methylatlon pro 1e '
(2763 ,2764}. Among infants with SHH-activated medulloblasto-
mas in one clinical trial {2691 J. SHH-1 tumours were associated

I
with a worse progression-free survival than SHH -2 tumours , } In the absence of these high-
86 3614
which are enriched for MBEN pathology; however, results from ~nd adolescents. 12 ~· this ~ e group have better outcomes
other trials differ or are equivocal (1783 ,2186} , probably reflect- nsk features . patients
1

5 11 8
J
}. Adult SHH -activated medul-
ing differences in cohort composition and treatment strategies (> 80% survival rate) (218~ ~I favourable prognosi s, although
loblastomas ha~e a reda ~ ·~ally controlled trial cohor ts in this
1
(e.g. different schedules of intrathecal methotrexate).
Metastatic disease and MYCN amplification are indepen- molecularly def 1ned an c ini
dently associated with a poor prognosis in non-infant children patient group are rare \973] .

Ernbryonal tumours 207


Ellison DW
Medulloblastoma, SHH-activated Clifford SC
KaurK
and TP53-mutant Korshunov I
Northwtt PA
Taylor MD

Definition often a presenting feature of SHH-activated and TPS1 rnut8r t


Medulloblastoma , SHH-activated and TP53-mu tant, is an medullobla stornas I2615, 2865).
embryonal tumour of the c r bellum demons tratin g ac tivati on
of the sonic t1edgehog (SHH) signa lling pathway in combination Epidemiology
with a mutant TP53 gene (CNS WHO grade 4). SEER data from 1973-2007 show annual medullobl;=_1s-
toma incidence rates of 6 cases per 1 million children aged
ICD-0 coding 1- 9 years and 0 .6 cases per 1 million adults aged > 19 ye::ws
9476/3 Medulloblastoma, SHH-ac tivated and TP53-mu tan t 12972 ). SHH -activated medulloblastomas in general '>how 8
bimodal age distribution, being most common in infants and
ICD-11 coding young adults , with an M:F ratio of approximately 1.5:1 [22801
2A00.10 & XH·1SH4 Medulloblastoma of brain & Medulloblas- In contrast, SHH-activated and TP53-mutant tumours are
toma, SHH-activated and TP53-mutant generally found in children aged 4- 17 years 11702). In one
study that included 133 SHH -ac tivated medulloblastomas
Related terminology 28 patients (21%) had a TP53 mutation , and the median age
None of these patients at presentation was approximately 15 years
(3614) .
Subtype(s)
SHH-activated medulloblastomas comprise four provisional Etiology
molecular subgroups (SHH-1, SHH-2, SHH -3, SHH -4), which There are several hereditary tumour syndromes that predis-
can be demonstrated by DNA methylation or transcriptome pro- pose to the development of SHH-activated medulloblastoma
filing (see Fig. 4.02, p. 201). 13392). Germline TP53 point mutations (Li-Fraumeni syn-
drome) predispose to medulloblastoma , and these tumours
Localization belong to the SHH -activated group (2622) . More than half of
Insufficient data exist to derive confi dent conclusions about the all SHH -acti vated and TP53-mutant medulloblastomas have
localization of TP53-mutant, SHH-activated medulloblastomas . germline rather than somatic TP53 alterations . Mutations 1n
However, TP53-mutant tumours tend to occur in children aged TP53 are most commonly found in the DNA-binding reg ions
5-14 years, and most medulloblastomas arising within this age encoded by exons 4 through 8 (3614) . Although some WNT-
range are found in the cerebellar hemispheres 12463,3164, activated medulloblastomas have mutations in TP53 , these
3405). have so far been somatic , and TP53 mutations do not por-
tend a negative prognosis in WNT-activated medulloblastoma
Clinical features (3614) .
Most patients present with symptoms and signs of raised intra-
cranial pressure from non-communicati ng hydrocephalus due Pathogenesis
to occlusion of the fourth ventricle by the primary tumour. TP53 mutations are reported in 10-15% of SHH -activatea
medulloblastomas, and more than half of these are germl1ne.
Imaging MYCN amplification is observed in 5-10% of SHH-activated
On CT and MRI, medulloblastomas appear as solid, intensely medulloblastomas (1702 ,2865 ,2279,3614). TP53 mutations
contrast-enhancing masses. SHH-activated medulloblastomas and MYCN amplifications occur as part of a constellation of
are most often identified in the lateral hemispheres but can also associated features alongside GL/2 amplification and chromo-
involve midline structures. TP53-mutant tumours are more likely thriptic rearrangements {1702,2865,2279,2622) . Isolated chro-
to be midline /1099,3164). mosome 17p deletion and loss of heterozygosity at the mutant
TP53 locus are characteristic of SHH-activated TP53-mutant
Spread tumours .
Medulloblastomas have the potential to invade locally, metasta- SHH-3 subgroup medulloblastomas characterized by TP53
size to the leptomeninges, or (rarely) spread outside the CNS . mutation, MYCN amplification , and/or large cell / anaplast1c
Most metastases are found on the surface of the CNS, attached morphology are reported not to have ELP1 mutations [33931
to the pia mater. Mechanisms of metastasis for SHH-activated TP53-mutant tumours do not form a discrete molecular sub-
medulloblastoma are unclear, with spread to the leptomeninges group upon c lass discovery analysis using genome-wide
through the cerebrospinal fluid (CSF) or via a haematogenous expression and DNA methylation- based techniques I1702,491
route with return to the leptomeninges 11044). SHH-activated 2865).
rriedulloblastomas are less frequently metastatic at presentation See also Medulloblastoma, SHH-activated and TP53-wildt)pr?
than molecular group 3 tumours , but leptomeningeal spread is (p 205).
~ "' -
I
Fl~·~·06 .s.HH -acti~ated a~d TP53-mutant medulloblastoma. Marked anaplasia and Fig. 4.~7 SHH-activated and TP53-mutant medulloblastoma. Widespr~ad strong nu-
m1tot1c act1v1ty, consistent with large cell / anaplastic medulloblastoma. clear immunoreactivity for p53, reflecting the presence of a TP53 mutation.

Macroscopic appearance Box4.03 Diagnostic criteria for medulloblastoma, SHH-activated and TP53-mutant
In general , medulloblastomas appear as friable pink masses . Essential:
No data exist to suggest that tumours of this specific type have
A medulloblastoma
any characteristic macroscopic feature .
AND
Histopathology Mutant TP53 gene
Diffuse anaplasia accompanied by a substantial large-cell AND
phenotype occurs in approximately 70% of SHH-activated and SHH pathway activation
TP53-mutant medulloblastomas. Other tumours are generally OR
desmoplastic/nodular with focal anaplasia {2613}. A DNA methylation profile aligned with SHH-activated medulloblastoma

lmmunophenotype
A panel of immunohistochemical markers can be used to iden- Mutation analysis of mutated SHH pathway genes (PTCH1 ,
tify SHH-activated tumours among medulloblastomas (but not SUFU, SMO) provides further diagnostic markers tor SHH-
other embryonal tumours) {837,1576}. SHH-activated tumours activated tumours .
express GAB1. Both SHH-activated and WNT-activated medul- For identification of a TP53-mutant and/or MYCN-amplified
loblastomas express YAP1 , but SHH-activated medulloblasto- SHH-activated medulloblastoma, assessment of TP53 muta-
mas do not show nuclear immunoreactivity for p-catenin . The tion and MYCN amplification status are essential. Large cell I
presence of a TP53 mutation is suggested by widespread anaplastic morphology and chromothriptic rearrangeme nts
strong immunoreactivity for p53 protein in tumour cell nuclei . are also associated with this tumour type and provide useful
supplementary assessments (2865,2279) . Given the associa-
Cytology tion of SHH -activated TP53-mutant medulloblastoma with Li-
Evaluation of the presence of tumour cells in CSF is required Fraumeni syndrome, and the high overall incidence of germ-
for staging. line predisposition within SHH-activated medulloblastoma ,
mutation analysis of tumour and blood samples for PTCH1 .
Diagnostic molecular pathology SUFU, TP53, ELP1 , and GPR161 and genetic counsell ing are
SHH-activated medulloblastomas comprise four provisional recommended for all patients with SHH -activated medullo-
molecular subgroups (SHH-1, SHH-2, SHH -3, SHH-4), which blastoma (3392) .
can be demonstrated by DNA methylation or transcriptome pro - MYCN amplification is also associated with group 4 non-
fil ing (see Fig . 4.02 , p. 201). SHH-activated and TP53-mutant WNT/non-SHH medulloblastoma, and TP53 mutation with WNT-
medulloblastomas almost always belong to subgroup SHH-3 activated medulloblastoma . However, neither alteration is asso-
\2865,49 1,2691 ,3083,3393) . ciated with a poor outcome when they arise in these specific
Groups and subgroups of medul loblastoma may be identi- contexts (3614,2279,2865 ,2913,1148) .
fied using DNA methylation profiling or gene expression pro-
filing and associated minimal classifier assay s (2866,1356, Essential and desirable diagnostic criteria
1715,2864). as well as by immunohistochemistry (837,2 117). See Box 4.03 .

Embryon~il tumours 209


Staging Prognosis and prediction
Clinical staging procedures include MRI examinations of the In non-infant children and adolescents with SHH -acti ialc.n
CNS with contrast agent. This is complemented by lumbar medulloblastoma, TP53 mutation. and MYCN ampht1catian a~;.
puncture postoperative CSF cytology. The postoperative stag- eii
associated with each other and with a very poor oute-0me 117
ing system developed by Chang and others in 1969 \519). 3614,2865,1148}. Among SHH group tumours. both altera 'r>n-
which defines the following degrees of metastatic spread, is still are consistently associated with poor outcomes in un 1 /<jr ~
being used: ate survival analyses and are independent predictors of W.1
outcomes when considered together in multivariate a naly~h".
MO No evidence of subarachnoid or haematogenous 13614,2913,2865,1148}.
metastasis
M1 Microscopic tumour cells found in the CSF
M2 Gross nodular seeding demonstrated in the cerebellar/
cerebral subarachnoid space or in the third or lateral
ventricles
M3 Gross nodular seeding in the spinal subarachnoid space
M4 Metastasis outside the cerebrospinal axis
Medulloblastoma, non-WNT/non -S HH Ellison OW
Clifford SC
Kaur K
Korshunov A
Northcott PA
Taylor MD

Definition
infants. Group 3 medulloblastomas are exceed ingly rare in
Medulloblastoma, no ~ -WNT/non-SHH , is an embryonal tumour adults 11703.491}. Group 4 medulloblastomas are the largest
of the ~er~bellum without a molecular signature associated molecular group, accounting for about 40% of all medulloblas-
with act1vat1on of the WNT or sonic hedgehog (SHH) signalling tomas. Peak Incidence occurs in patients aged 5-15 years , with
pathway. Non-WNT/non-SHH medulloblastomas are classi fied lower incidence in infants and adults {1703,491 ).
as group 3 or group 4 tumours and comprise eight molecular
subgroups, demonstrated by DNA methylation profiling .
Etiology
Very little is known about the molecular etiology of group 3
ICD-0 coding and group 4 medulloblastomas; generally, they are not asso-
9477/3 Medulloblastoma, non -WNT/non-SHH ciated with known hereditary tumour syndromes . Rare cases
of group 3 or group 4 medulloblastoma have bee~ re~orted in
ICD-11 coding Ind ividuals with a germ line CR EB BP mutation (Rub1nste1n-Tayb1
2A00.10 & XH8705 Medulloblastoma of brain & Medulloblas- syndrome) {689). Germline mutations of the DNA repair genes
toma , non-WNT/non-SHH PALB2 and BRCA2 have also been identified in non-WNT/non-
SHH medulloblastoma 12206).
Related terminology
None Pathogenesis
Cross-species sing le-cell transcriptomic studies have discerned
Subtype(s) putative cellular origins of grou p 4 medulloblastoma, including
Non-WNT/non-SHH medulloblastomas comprise eight molecu- upper rhombic lip- derived glutamatergic neurons from cerebel-
lar subgroups (group 3/4 subgroups 1-8), which can be dem- lar nuclei and unipolar brush cells 13338,1357). The pathogenesis
onstrated by DNA methylation profiling analysis of non-WNT/ of group 3 tumours remains less cl ear. Primitive nestin-positive
non-SHH group 3 and group 4 medulloblastomas (2865,491 , cerebellar stem or progenitor c ells are implicated by single-cell
2279,2900). transcriptomics {3338), and variou s neural stem or progenitor
cell populations demonstrate vu lnerability to transformation in
Localization mouse tumour modelling studies /1580,3 134,2437}.
Non-WNT/non-SHH medulloblastomas arise exclusively in the
cerebellum (usually in the midline), and almost always in its infe- Genetics
rior portion . Overexpression of MYC is a common feature of group 3 medul-
loblastomas , and MYC amplification, often accompanied by
Clinical features PVT1 :: MYC fusion {2284), occ urs in 17% of group 3 tumours
Most patients present with symptoms and signs of raised intra- 1839,2279). Other recurrently mutated or focally amplified
cranial pressure from non-communicating hydrocephalus due genes include SMARCA4 (mutated in 9% of cases) , CTDNEP1
to occlusion of the fourth ventricle by the primary tumour. (mutated in 5%), KMT20 (mutated in 5%), MYCN (amplified in
5%), and OTX2 (ampl ified in 3%) {2279). Two oncogenes in
Spread medulloblastomas from groups 3 an d 4 are the homologues
Mechanisms of metastasis for medulloblastoma are unclear, GF/1 and GF/18, which are aberrantly overexpressed through
with spread to the leptomeninges through the cerebrospinal a mechanism called enhancer hijacking in 15% and 12% of
fluid (CSF) or via a haematogenous route with return to the group 3 and group 4 tumours , respectively {2281 ,2279) . The
leptomeninges {1044). Patients with non-WNT/non-SHH medul- most common cytogenetic aberrations in medulloblastoma
loblastomas present almost universally with distant CNS metas- (occurring in 55- 58% of group 3 and 80-85% of group 4
tases at the ti me of recurrence /2614 ,1309A}. Metastatic dis- tumours) involve ch romosome 17 copy-number alterations:
ease is present at diagnosis in about 40% of group 3 tumours 17p deletion , 17q gain, or a combination of these in the form of
in infants and affects > 50% of patients with non-WNT/non-SHH an isodi centric 17q (837,1703,2280,2279) .
(grou p 3/4) subgroups 2- 511703 ,2280) . An isolated local recur- The most frequently mutated or focally amplified genes in
rence of a group 3 or group 4 medulloblastoma should be con - group 3 and 4 tumours are KDM6A (mutated 1n 7% of cases) ,
sidered a rad iation -induced neoplasm until proved otherwise by OTX2 (amplifi ed in 6%), ZMYM3 (mutated in 6%) , KMT2 C
biopsy. (mutated in 6%), KBT804 (mutated in 6%), MYCN (amplified
in 6%), ZIC 1 (mutated in 4%), CDK6 (amplified in 4%) , KMT20
Epidemiology (mutated in 3%), and TBR1 (mutated in 3%) \2279) . Enhancer
Group 3 tumours account for approximately 25% of all medul- hijacking of the SNCAJP gene locus leading to aberrant overex-
loblastomas, and for a hi gher proportion of cases (-40%) in pression of PRDM6 is specific to group 4 medulloblastorna an d
is seen in abou~ 17% of tumours (2284 ,2279). O le1erlous hel"e- Box4.04 Diagnostic criteria for medulloblastoma, non-WNT/non-SHH
rozyg~us germ line mutations .in BRCA2 and PALB2 are present
1~ 1-2.Yo of patients'. subs tantiated by tumour-associated muta- Essential:
tion signatures typical of homologous recombination e · A medulloblastoma
deficiency 13392). ~edullobla~tomas from both group ~ ~~~ AND
group 4 show recu.1rent som~t.1c ~enetic events that converge No WNT or SHH pathway activation
on the posttranslat1onal mod1f1cat1ons of histones, particular! OR
H3 p.K28 (K27) and H3 p.K5 (K4) /2689,795 ,1497). y A DNA methylation profile aligned with group 3 or group 4 medulloblastoma

Macroscopic appearance
Medul·l~blastomas a~pear. as friable pink masses , occasion- frequent in subgroup 2 . Metastatic disease at presentation 15
ally wit macroscopic foci of necrosis . At surgery, non -WNT/ relatively frequent in subgroups 2-5. A relatively poor outcornP,
non-SHH medulloblastomas show brainstem Invasion more is associated with tumours in subgroups 2 and 3.
often than do other .types of medulloblastomas [2463) . Group 3
tumours are more likely
. to contain macrocysts and are us ua II y Essential and desirable diagnostic criteria
sma II er at presentation than group 4 tumours . {3601,3581 ,681). See Box 4.04 .

Histopathology Staging
Most non-WNT/non-SHH medulloblastomas have a classic Clinical stag ing procedures include MRI examinations of the
morphology. Such tumours occasionally exhibit areas of Homer CNS with contrast agent. This is complemented by lumbar
Wright (neuroblastic~ rosette formation, or a palisading pattern puncture postoperative CSF cytology. The postoperative stag-
of tumour cell nuclei or even nodule formation , in the absence ing system developed by Chang and others in 1969 !519].
of desmoplasia (which has been termed "biphasic classic " mor- which defines the following degrees of metastatic spread , is still
phology) {20571. Large cell I anaplastic tumours can belong to being used :
either grou~ 3 or group 4 . However, they are present at a higher
frequency 1n group 3 {837,676} and are relatively enriched in MO No evidence of subarachnoid or haematogenous
group 3/4 subgroup 2 tumours (1355). Very rarely, desmoplas- metastasis
tic/nodular medulloblastomas have been assigned to the non- M1 Microscopic tumour cells found in the CSF
WNT/non-SHH group {2865} . M2 Gross nodular seeding demonstrated in the cerebellar/
cerebral subarachnoid space or in the third or lateral
lmmunophenotype ventricles
A panel of immunohistochemical markers can be used to iden- M3 Gross nodular seeding in the spinal subarachnoid space
tify non-WNT/non-SHH tumours among medulloblastomas {837, M4 Metastasis outside the cerebrospinal axis
1576). Unlike WNT and SHH medulloblastomas, non-WNT/non-
SHH tumours do not express YAP1. They do not express GABI Prognosis and prediction
and show no nuclear immunoreactivity for p-catenin. MYC amplification has long been established as a genetrc
alteration associated with poor outcome in patients with medul-
Cytology loblastoma {2S32 ,S19,S39J . This observation is reflected in the
Evaluation of the presence of tumour cells in CSF is required relatively poor outcomes ascribed to group 3 medulloblastomas
for staging . overall , but MYC ampl ification, isodicentric 17q, and metastatic
disease at diagnosis all have prognostic significance among
Diagnostic molecular pathology group 3 tumours {2913 ,2S65J. Metastatic disease at the time of
Analysis of DNA methylation profiles, either alone or in com- presentation , which is associated with poor outcome, is currently
bination with transcriptomic data, has identified molecularly the most robust prognostic marker among group 4 tumours
heterogeneous subgroups among group 3 and group 4 medul- (2913). High-risk DNA methylation patterns are also associated
loblastomas with distinct clinical and genetic associations with a poor prognosis {2865). In contrast , chromosome 7 gain.
{2279,2865,491 ). A large meta-analysis of 1501 medulloblasto- chromosome Sloss, chromosome 11 loss, and chromosome 17
mas studied by DNA methylation profiling supports the exist- gain have been implicated as markers of favourable outcome
ence of eight robust group 3 or group 4 subgroups , designated among group 4 medulloblastomas in retrospective clinical stud-
group 3/4 subgroups 1-8 {2900) (see Fig . 4.03, p. 202). Sub- ies {2913 ,2865,114SJ. The DNA methylation subgroups of non-
groups 2, 3, and 4 consist exclusively of group 3 medulloblas- WNT/non -S HH tumours exhibit disparate outcomes, with sub-
tomas, whereas subgroups 6, 7, and 8 predominantly comprise groups 2 and 3 exhibiting particularly poor outcomes \29001
group 4 medulloblastomas. Subgroups 1 and 5 are intermediate Favourable-risk cytogenetic aberrations (i .e. chromosome 7
subgroups, exhibiting molecular and cellular attributes charac - gain , chromosome Sloss, and chromosome 11 loss) are associ-
teristic of both group 3 and group 4 medulloblastomas (2279 , ated with subgroups 6 and 7, whereas poor-prognosis tumours.
with isochromosome 17q and otherwise quiet genomes . are
2900,1357) . Most non-WNT/non-S HH medulloblastomas have a
commonly associated with subgroup S I114S,2900I .
classic morphology, but large cell I anaplastic tumours are more

212 Err1txyor1al turr1<Jur::,


Medulloblastoma , histologically defined Korshunov A
Elli son OW
Giangaspero F
Orr BA
Pi etsch T
Taylor MD

Definition
ICD -1 1 coding
Medulloblastoma is an embryonal neuroepithelial tumour aris- 2A00.10 & XHORY1 Medulloblastoma of brain & Classic medul-
ing 1n the posterior fossa . h1~tologically characterized by small , loblastoma
poorly differentiated cells with a high N:C ratio and high levels 2A00.10 & XH7PN5 Medulloblastoma of brain & Oesrnoplast1c
of m1tot1c activi ty and apoptosis .
nodular medulloblastoma
2A00 .10 & XH6JN6 Medulloblastoma of brain & Medulloblas-
ICD-0 coding toma with extensive nodularity
9470/3 Medulloblastoma, histologically defined 2A00 .10 & XHOH95 Medulloblastoma of brain & Anaplast1c
9471 /3 Desmoplastic nodular medulloblastoma medulloblastoma
9471 /3 Medulloblastoma with extensive nodularity
9474/3 Large cell medulloblastoma Related terminology
9474/3 Anaplastic medulloblastoma Not recommended: cerebellar neuroblastoma .

f ig. 4.08 Desmoplasticmodular medulloblastoma. T1-weighted (A). and T2-weigh ted (B). contrast-enhanced MRI of tumours in the cerebellar hemisphere. C Tl-weighted,
contrast-enhanced MRI of a tumour 1n the verm1s.

- ...-..L:•::;,.;
fig. 4.09 Medulloblastoma with extensive nodularity. A In a 1-month-old girl , gadolinium -enhanced, sag1ttal. 11 we 1ghted MRI sl1ows a l1uge lesion 1nvolv1ng both cerebellar
hemispheres and the verm1s. The lesion has a mult1nodular and gynform pattern of enhancement There is also supratentoriJI hydrocephalus and mac1ocran1a B Mult1nodular
and gynform pailern . c Note the downward herniation of the tumour through the foram en magnum (arrow! ai1d the marlo.ed effacement of the c1sternal spaces of the posterior
fossa There 1s also supratentonal hydrocephalus and macrocran1a .

' 1 )r" 1 1 ,r •c' 213


medulloblastomas occurring in the cerebellar hern1sph8rF;', ;_ir".: r,i
the D/N type, especially in adults [3153 ,3164,2463 ,3405 3>Ir ;
Medulloblastomas with extensive nodularity IMBEr Jc,! ~re.
located in the vermis, with involvement of both hem1sph1=H<::~:
This localizat1on contrasts with that of D/l'J medullobl8st 0 r~a
which more frequently involves the cerebellar hemisphere-;
(1099,3164,3405 ,2463,3601 I.
Large cell / anaplastic (~C/~) medulloblastomas are typ1r::a1 11
located in the cerebellar m1dl1ne and involve the fourth ventricle
cavi ty an d adjacent brainstem and cerebellar structures LC/A
medulloblastomas with SH H activation can show lateral locali-
zation wi th extrace rebellar extension r1089,3405.36011

Clinical features
Most patients present with symptoms and signs of raised 1ntra-
cranial pressure from non-communicating hydrocephalus due
to occlusion of th e four th ventricle by the primary tumour

Imaging
On MR I, classic medulloblastomas generally appear as hyper-
intense, homogeneou s. contrast-en hancing masses with m1d-
line localization . Some medulloblastomas (frequently non-WNT/
non-SHH group 4 medull oblastomas) enhance inhomogene-
ously. WNT-activated medull oblastomas are typically located
in the cerebellar midline I cerebell opontine angle , with many in
close contact with the brainstem \31 64,2463,3405) .
D/N medulloblastomas appear as soli d , frequently contrast-
enhancing masses. Tumours originatin g peripherally 1n a cer-
ebellar hemisphere in adults occasionally occur as extra-axial
lesions \2463,3405 ,36011 . Infants frequentl y present with a very
superf icial lesion of the lateral cerebellar hemisphere, which 1s
den sely enhancing and nearly pathognomonic for SHH-acti-
vate d med ulloblastoma.
MBEN s ap pear as very large multi nodular lesions with an
enhanci ng bunch-of-grapes structure involving the verm1s and
sometimes the adjacent cerebellar hemis pheres \1088,21971 .
Rare cases have a peculiar gyriform appearance , 1n which

.
the cerebellar folia are well delineated and enlarged , with con-
trast enhancement \32) . Downward hernia tion of the cerebellar
tonsi ls and effacement of the ci sternal spaces of the posterior

.~ ~.l'-~
Flg.4.10 Histopathologlcal features of classic medulloblastoma. A Typical syncytial
fossa can be observed .
LC/A med ulloblastomas appear as heterogeneously contrast-
arrangement of undifferentiated tumour cells. B Area with Homer Wright {neuroblas- enhanci ng masses with foc i suggestive of necrosis and peritu-
tic) rosettes. c Arrangement of tumour cells in parallel rows {spongioblastic pattern). moural oedema {1089 ,3405 ,3601 }.

Subtype(s) Spread
Classic medulloblastoma; desmoplastic/nodular medulloblas- At diagnosis , classic medullobl astoma has disseminated to the
toma ; medulloblastoma wi th extensi ve nodularity ; large cell I leptomeningeal compartm ent of the CNS in as many as 40% of
anaplastic medulloblastoma c lassic medu lloblastomas [3 153,1703).
Leptomeningeal metastases are found 1n 20% of D/N medul-
lobl astomas at diagnosis. Most recurrences of D/N medullo-
Localization
Classic medulloblastomas are typica lly located in the cerebe l- blastoma are found loc ally in the tumour bed, in the posterior
lar midl ine , involving the fou rth ve ntric le cavi ty, with or without fossa , whereas metastatic spread to the leptomeninges or sys-
close contac t with the brainstem . Some classic (WN T- or sonic temically is less comm on among patients with these tumours
hedg ehog [SHH] -activated) medulloblastomas are localized (3 153 ,1703].
laterally, involving the cerebellar pedunc le and hemisphere MBEN can relap se locally or (rarely) metastasize via cer-
(3 153,3 164 ,2463 ,340 5, 3601 ]. ebrospinal fluid (CSF) pathways However, such cases seem
Oesmoplast1 c/nodular (0/N) medulloblastomas may arise to re spond well to subsequent treatm ent and have a favourable
both 1n the ce rebellar hemi sphere and in the vermis. Most prognosis [2614] .

'J 1 A i , , I ,, "\ I , .d t i 11 , 1r 1 1 ir <


I
.

At diagnosis, metastatic disease is found in as many as LC/A medulloblastomas can occur in patients of any age and
60-70% of patients with LC/A medulloblastomas . Tumours account for about 10% of all tumou rs. Consi dered separately.
recur frequently and metastasize via CSF pathways (1089, anaplastic medulloblastomas are about 10 times as prevalent
3405,3601}. as large cell medulloblastomas . They are most frequent among
medulloblastomas in the non-WNT/no n-SHH (group 3) and
Epidemiology SHH-activated , TP53-mutant groups, bu t very rare in the WNT-
Classic medulloblastomas account for 70-80% of all medul- activated group (839,3153 ,1703,1355}.
loblastomas (818 ,819 ,834) . They can occur at any age , from
infancy to adulthood , but predominantly arise in childhood Etiology
(60 - 70% of cases), and they are found in all four genetically Medulloblastomas occurring in the context of naevoid basal cell
defined medulloblastoma types but predominantly in WNT- carcinoma syndrome are mainly desmoplastic subtypes (D/N
act ivated and non -WNT/non -SHH medulloblastomas (1720 , medulloblastoma or MBEN). Th is syndrome is diagnosed in
3153 ,1703). 5.8% of all patients with medu llob lastoma , in contrast to 22.7%
D/N medulloblastomas are estimated to account for 20% of of patients with a D/N medull oblastoma and 41 % of patients with
all medulloblastomas . In ch il dren aged < 3 years , D/N medul - an MBEN {1702 ,5 ,3392,1722,17 18,1719,2510) . Conversely, the
loblastomas account for 40- 60% of all cases . In adult patients , risk of medulloblastoma is approximately 2% in PTCH1-related
D/N medulloblastomas c onstitute 20- 40 % of all histological naevoid basal cell ca rcinoma syndrome and 20 times higher
subtypes j1720,315 3,1703J in SUFU-related naevoid basal cell carcinoma syndrome [3152 ,
In large series , MBENs acc ou nt for 3.2-4.2% of all medul - 100 ,1042,391 ,2963,1 1821. Recurrent germline alterations in
loblastoma subtypes overall , but in chi ldren aged < 3 years (in EL P1 or GPR161 also pred ispose to medulloblastomas 1n this
whom D/ N medullobl astomas account for as many as 50% of (SHH -activated) group (3392,232,3393) . Because of the fre-
c ases), MBENs have been reported to acco unt fo r 20% of all quency of predisposing germline mutations in this patient popu -
cases {1088,1703,1722,1719). Both D/N medull ob lastoma and lation, genetic counselling is indicated for children and their
MBEN belong to th e SHH -activated molec ul ar medu ll oblastoma families diagnosed with D/N medulloblastoma or MBEN {10421.
type.
In rare cases, classic medulloblastomas are diagnosed D/N medulloblastomas tend to be firm and circumscribed
within the setting of constitutional mismatch repair deficiency reflecting intratumoural desmoplasia.
syndrome or Rubinstein-Taybi syndrome, or in individuals with MBENs tend to be firm , grape-like, and well circumscribed
germ line APC, BRCA2, or PALB2 mutations. LC/A medul\oblastomas appear as friable grey1sh-p1rK
The vast majority of LC/A medulloblastomas are sporadic, masses, occasionally with macroscopic foci of necrosis. At sur-
and little is known about their etiology. SHH-activated , TP53- gery, LC/A medulloblastomas often show cerebellar and bra1n-
mutant medulloblastomas are often diagnosed within the set- stem invasion {2463) .
ting of Li-Fraumeni syndrome.
Histopathology
Pathogenesis There are four established morphological subtypes of medu1-
See also the sections on Medulloblastomas, molecularly defined loblastoma. Each of these histologically defined subtypes has
(p. 203). particular clinical and molecular associations (818,2056.2057
D/N medulloblastomas are derived from granule cell progeni- 2031 ). Arch itectural and cytological diversity can manifest 1ot
tor cells forming the external granule cell layer during cerebellar only as nodule formation but also as neurocytic or ganglion ce11
development (3402,2860) . These progen itors are dependent on differentiation . Rarely, any histological subtype of medulloblas-
SHH (produced by Purkinje cells) as a mitogen . Recently, single- toma may show myogenic and/or melanotic differentiation. the
cell RNA sequencing revealed that these tumours contain cells terms "medullomyoblastoma" and "melanocyt1c medulloblas-
resembling different stages of granule cell precursor develop- toma'', respectively, have been used to describe these patrerns
ment (granule cell progenitor-like cells) {1355} . D/N medulloblas- {971,2057,2770 ,2497,2973) .
tomas in adults contain a higher proportion of undifferentiated
granule cell progenitor- like cells than do tumours in infants. Classic medulloblastoma
Like D/N medulloblastomas, MBENs are believed to derive Classic medulloblastomas are the archetypal CNS small blue
from cerebellar precursor cells of the granule cell lineage {3402, round cell tumour. They consist of densely packed , poorly dif-
2860) . ferentiated embryonal cells with hyperchromatic nuclei of vari-
Non-WNT/non-SHH group 3 LC/A medulloblastomas prob- ous shapes. Mitotic activity is increased and apoptot1c bodies
ably arise from a stem cell-like population in the early develop- can be found . lntratumoural desmoplasia is absent, but a des-
ing cerebellum {3338) . moplastic reaction can be induced where tumour cells invade
the leptomeninges . Homer Wrig ht rosettes are found in sorne
Macroscopic appearance classic medulloblastomas. Occasionally. nodules of neuro-
Classic medulloblastomas appear as friable pink masses , cytic differentiation and reduced cell proliferation are locallv
occasional ly with macroscopic foci of necrosis 12463). present in classic tumours . but these are never assoc1atad will'

216 !-rribry(Jr1 al tur nours

-
I
.

internodular desmoplasia or perinodular collagen when exam- index are much higher in the internodular areas than in the nod-
ined in a reticulin preparation. Such non-desmoplastic nodular ules.
medulloblastomas correspond to non-WNT/non-SHH tumours , Focal frank anaplasia can be seen occasionally within the
unlike the typical D/N medulloblastomas, which belong to the internodular areas , especially in SHH -activated and TP53-
SHH-activated type. WNT-activated classic medulloblastomas mutant tumours . Medulloblastomas that show only an increased
often show intense vascularization and blood-brain barrier dis- amount of reticulin (without a nodular pattern) or that show a focal
ruption {3153,1703). nodular patiern but without complete perinodular encircling by
reticul in are not classified as D/N medulloblastoma (see Clas-
Desmoplastic/nodular (DIN) medulloblastoma sic medu!loblastoma, above); the two characteristic features
DIN medulloblastoma is characterized by a bicompartmental must occur together for a diagnosis of D/N medulloblastoma
arrangement of nodular, reticulin-free zones (pale islands) sur- However, posttreatment progression-associated anaplasia with
rounded by densely packed, poorly differentiated , highly prolif- loss of the key diagnostic features has been described in D/N
erative cells with hyperchromatic and moderately pleomorphic medulloblastomas, especially in patients with germline PTCH1
nuclei , which produce an extensive network of intercellular retic- aberrations {171 8).
ulin fibres {818,2057,1086,837,2031). In rare cases , this defining
pattern is not present throughout the entire sample, with some Medulloblastoma with extensive nodularity (MBEN)
areas instead having a more syncytial arrangement of non-des- MBEN differs trom the related D/N subtype in that 1t has an
moplastic embryonal cells. The nodules contain tumour cells expanded lobular architecture due to the ret1culin-free zones
with features of variable neurocytic maturation embedded in a being substantially larger and richer in neuropil-like matrix
neuropil-like fibrillary matrix. Homer Wright rosettes are gen- {1088,1722,1719). These zones contain a population of small
erally not found in D/N medulloblastoma. Tumours with small cel ls with round nuclei, which show various degrees of neuro-
nodules can easily be overlooked if no reticulin staining is per- cytic differentiation and can exhibit a streaming pattern . The
formed . The level of mitotic activity and the Ki -67 proliferati on internodular component can vary from one area to another and

EmbryunJI tu not r~ 21 7
-
Rg. 4.15 Large cell / anaplastic medulloblastoma. A Increased nuclear size, pleomorphism, and prominent nucleoli. B Tumour cell wrapping is also ev ident.

appea r markedly reduced in some places. Like in D/N medul- typical of MBEN , but they can also occu r in D/N medulloblas-
loblastomas, mitotic activity and Ki -67 proliferation index is toma . GFAP expression can be frequently found in both com-
low or absent in the neurocytic areas and much higher in the ponents, most often in the internodular cells . D/N medulloblas-
internodular areas. After radiotherapy and/or chemotherapy, tomas express GAB1 , YAP1 , and the low-affinity nerve growth
MBENs occasionally undergo further maturation into tumours factor receptor p75-NGFR (particularly in internodular areas)
dominated by ganglion cells (533 ,700) . whereas OTX2 immunohistochemistry is consistently negative
(2508).
Large cell I anaplastic (LC/A) medulloblastoma In MBENs, the neuropil-like tissue matrix and the differenti-
Most LC/A medulloblastomas show a combination of large cell ated neurocytic cel ls with in nodules are strongly immunoreac-
and anaplastic features . Anaplasia as a feature of embryonal tive for synaptophysin. These latter cells are also strongly (and
tumours was first proposed for medulloblastomas with marked less variably than in D/N medulloblastomas) immunoreact1ve
nuclear pleomorphism accompanied by particularly high mitotic for NeuN {1722 ,1719). Like D/N med ulloblastomas, MBENs are
and apoptotic counts (818 ,1089). Nuclear moulding , cytoplas- negative for OTX2 and positive for GAB1 and p75-NGFR in
mic pseudoinclusions, and cell wrapping are typical features . internodular areas (1722,1719).
The large cell phenotype manifests more uniform round nuclei
with prominent nucleoli, lacking the variability in cell size and Differential diagnosis
shape that characterizes the anaplastic phenotype; its cells Embryonal tumours with multilayered rosettes and atypical tera-
are relatively large and monomorphic but show the high rate of toid/rhabdoid tumours may show histomorpholog ical overlap
turnover seen in anaplastic tumours. with medulloblastomas . Unlike med ulloblastomas, embryonal
tumours with multilayered rosettes are typically LIN28A-immu-
lmmunophenotype noreactive. Nuclear SMARCB1 and SMAR CA4 expression 1s
Classic medulloblastomas express various nonspecific neural retained in all medulloblastoma types; the loss of expression of
markers, such as CD56 (NCAM1), MAP2, and NSE. Most cases one of these SWl/SNF complex protei ns is diagnostic of atypical
are 1mmunopositive for synaptophysin and NeuN , but these neu- teratoid/rhabdoid tumours .
ronal markers may also be absent. lmmunoreactivity for NFPs
is very rare. Embryonal tumour cells showing GFAP expression Cytology
can be observed in rare cases . Some classic medulloblasto- Evaluation of CSF cytology is required for stag ing . In CSF sam-
mas express the transcription factor OTX2, with the exception of ples and touch preparations , small clusters of poorly differenti-
classic medulloblastomas with SHH activation (2508) . ated cells with mostly round hyperchromatic nuclei and scant
In D/N medulloblastomas , activation of the SHH pathway can cytoplasm can be seen [3113) . In samples of D/N medulloblas-
be inferred by immunohistochemistry for specific targets, such toma, neurocytic differentiation - characterized by smaller cell
as GAB1 and p75-NGFR (834,3153,837,1703). These markers size and round nuclei - may be observed , whereas samp les of
are predominantly expressed in internodular areas. The tran- LC/A medulloblastoma may reveal small cl usters of poorly dif-
scription factor OTX2 is negative, unlike in non-SHH medullo- ferentiated cells with large, atypical nuclei and scant cy toplasm,
blastomas (2508) . Widespread and strong nuclear accumula- nuclear moulding and wrapp ing ; and vi sib le nucleoli (31 13).
tion of p53 , suggesting a TP53 mutation, can be detected in
rare D/N medulloblastomas, frequently in association with signs Diagnostic molecular pathology
of cytologi cal anaplasia. This finding can accompany either See the sections on Medulloblastomas, molecularly defined
somatic or germline TP53 alteration (Li-Fraumeni syndrome) (p. 203).
(3096 ,36 14 ,1718]. The nodules in D/N medulloblastoma show In most cases , the molecular type of medulloblastoma can be
variable expression of neuronal markers, including synapto- identified by immunohistochemi stry, particularly for p-catenin,
phys1n and NeuN . Nodules with very strong NeuN expression, GAB1 , YAP1 , OTX2, p75 -NG FR , an d p53 (see lmmunopheno-
which 1s an indicator of advanced neurocytic differentiation , are type, above).

2 18 ~ ri 1t;r I' J11al h.Jn 1CJu rs


Essential and desirable diagnostic criteria Box4.05 Diagnostic criteria for classir:: medulloblastoma . h1stotog1r:ally defined
: e Box 4 05 .
Essential:
Staging A medulloblastoma
hnical staging procedures include MRI examinations of the AND
CNS with contrast agent. This is complemented by lumbar Absence of histological features qualifying for the diagnosis of desmoplastic/nodu-
puncture postoperative CSF cytology. The postoperative stag- lar medulloblastoma or medulloblastoma with extensive nodularity
ing. system developed by Chang and others in 1969 15191 AND
wh.1ch defines the following degrees of metastatic spread is still Absence of predominant areas with severe cytological anaplas1a and/or large cell
being used: ' cytology
AND
MO No evidence of subarachnoid or haematogenous Retained expression of SMARCB1 (INl1)
metastasis
M1 Microscopic tumour cells found in the CSF
M2 Gross nodular seeding demonstrated in the cerebellar/ to show an unfavourable clinical course 12057,839,2764 ,2031 ,
cerebral subarachnoid space or in the third or lateral 2691 j. Metastatic disease at presentation .did n~t affect the
ventricles favourable prognosis, suggesting that a d1agnos1s of MBEN
M3 Gross no?ular s.eeding in the spinal subarachnoid space confers a better outcome regardless of adverse clinical features.
M4 Metastasis outside the cerebrospinal axis However, germ line PTCH1 or SUFU alterations accompanied by
cancer-associated transcriptome signatures have recently been
Prognosis and prediction identified as molecular hallmarks of MBEN progression (1722,
See also the sections on Medulloblastomas, molecularly defined 1719).
(p. 203). At the time the tumours were first described , LC/A medul-
In most cases , D/N medulloblastoma in early childhood has loblastomas were strongly suspected to behave more aggres-
an excellent outcome with surgery and chemotherapy alone . In sively than other histological subtypes , with metastatic disease
a meta-analysis of prognostic factors in infant medulloblastoma often evident at presentation . In retrospective studies of patients
progression-free survival and overall survival were significant!~ in trial cohorts , LC/A morphology has been shown to be an
better for desmoplastic subtypes than for other medulloblasto- in dependent prognostic indicator of outcome . In contemporary
mas {2057,839,2764,2031 ,2691} . However, no difference in sur- trials, LC/A histology is regarded as a high-risk pattern war-
vival between D/N medulloblastoma and classic medulloblas- ranting intensified adjuvant therapy (819 ,2913 ,839,2832). The
toma was found in a European multicentre trial involving older 5-year progression-free survival rate for LC/A medulloblasto-
children with standard-risk medulloblastoma 12763). mas is 30-40% , although more aggressive behav iour can be
MBENs are typically associated with good to excellent out- shown by SHH-activated LC/A tumours with a TP53 mutation
comes, reaching an overall survival rate of almost 100% in rep- and by non-WNT/non-SHH group 3 tumours with MYC amplifi-
resentative series, although some cases have been reported cation (1702,3614 ,2865,1148).

bi 1bryuria1 lL nows
1 219
Wesseling P
Other CNS embryonal tumours : Pfister SM

Introduction

CNS embryonal tumours encompass a heterogeneous group


of malignant tumours that are composed o'f immature cells
resembling neural progenitors . They are mainly, but not exclu-
sively, a disease of childhood . Among CNS embryonal tumours ,
medulloblastomas are relatively common ; other CNS embryo-
nal tumours are rare. In contrast to medulloblastomas, which
by definition originate from the cerebellum or dorsal brainstem , 20
other CNS embryonal tumours may arise across the neuraxis.
The overarching term "CNS primitive neuroectodermal tumour"
previously encompassed many of the embryonal tumours
described in the following sections. However, this term is now
obsolete, thanks to an increased understanding of the hetero- oI
geneity and biology of these tumours and the emergence of a
classification based on molecular characteristics (3059 ,1944). ATRT-SHH I
I ATRT-::-MY~
The tumours discussed in the following sections include
. .·
- \
-- -~
atypical teratoid/rhabdoid tumour (AT/RT), embryonal tumour
with multilayered rosettes (ETMR), FOXR2-activated CNS - 20
?.: ,. :;'!-&.•
·)~·~·;;.
neuroblastoma, and CNS tumour with BCOR internal tandem "'"'
duplication (ITD). Whereas AT/RT and ETMR were included in
previous editions of this classification, FOXR2-activated CNS
neuroblastoma and CNS tumour with BCOR ITD are new. In -25 25 50

addition, cribriform neuroepithelial tumour has been introduced


Fig. 4.16 Other CNS embryonal tumour molecular groups. Unsupervised, non·linear
as a provisional entity within this category, and CNS embryo-
I-distributed stochastic neighbour embedding (t-SNE) projection of methylation array
nal tumour is included for embryonal tumours that defy a more profiles from non-medulloblastoma CNS embryonal tumours. Samples were selected
specific diagnosis. The designation "not elsewhere classified from a large database of > 50 000 brain tumour datasets to serve as reference profiles
(NEC)" can be added to a CNS embryonal tumour diagnosis in for training a supervised classification model based on strict criteria: all these samples
cases where adequate testing does not reveal signature molec- showed a high calibrated classification score (> 0.9) for tumours listed in the category
ular aberrations, and "not otherwise specified (NOS)" is used of "other CNS embryonal tumours" when applying the brain tumour classifier available
for cases where molecular analysis could not be (successfully) at https://www.molecularneuropathology.org. ATRT, atypical teratoid/rhabdoid tumou1.
performed (1946,1934).
Assessment of molecular characteristics is now considered Among the syndromes associated with non-medulloblastoma
the standard of care for CNS embryonal tumours and is exempli- CNS embryonal tumours , the risk of rhabdoid tumour predis-
fied by the evaluation of medulloblastoma molecular group and position syndrome 1 in a patient diagnosed with a SMARCB 1-
genetic profile (3153 ,835). For other CNS embryonal tumours , deticient tumour is reported to be between 26 % and 41 %, and
a diagnostic approach should seek the following: (1) biallelic the risk of rhabdoid tumour predisposition syndrome 2 in a
inactivation of the SMARCB1 (or rarely SMARCA4) gene in AT/ patient with a SMARCA4-deticient tumour appears to be much
RT (1840,1252,1481,2853), (2) C19MC alteration or O/CER1 higher (2885 ,1453,273,816 ,992,1254). Almost all patients with
mutation in ETMR {1650,1797), (3) FOXR2 activation In CNS a DICER1 -mutant ETMR carry a pathogen ic DICER1 germl1ne
neuroblastoma, and (4) a heterozygous ITD in BCOR exon 15 alteration .
in CNS tumour with BCOR ITD (3059,460) . lmmunohistochem - Like previous editions of the WHO classification of CNS
istry serves as a helpful surrogate for analysis of the actual tumours , this fifth edition should be re garded as a work rn
molecular alterations in some of these tumours , most notably progress. For example, we have included CNS tumours wrth
AT/RT. DNA methylation profiling is another useful alternative BCOR ITD under the heading Other CNS embryonal tumours.
diagnostic approach to this family of tumours . An integrated however, these tumours are not defin itively neural, and exon 15
diagnosis using tiers of histological and molecular information BCOR ITDs have been reported in several sarcomas. There 1s
in the pathology report is helpful for transparent and effective currently no consensus as to wheth er these tumours should be
communication of relevant tumour characteristics (1939 ,1945). considered neuroepithelial or mesenct1ymal neoplasms, and
Patients presenting with CNS embryonal tumours may have our perception of the nosology of such tumour types may netld
an inherited cancer syndrome and require genetic counselling . to change as new findings emerg e.

220 Embryonal turnours


Atypical teratoid/rhabdoid tumour Haberler C
Hasselblatt M
Huang A
Judkins AR
KoolM
Wesseling P

Definition upregulation of gen e profil es that have contributed to their


Aty~ical teratoid/rhabdoid tumour (AT/RT) is a high-grade nomenclature.
malignancy composed of poorly differentiated cells and a vari - These three sets of tumou rs can be reg arded as subtypes of
able number of rhabdoid cells, with the potential to differenti- AT/RT: AT/RT-SHH , AT/RT-TYR , and AT/RT-MYC .

I
ate along neuroepithelial , epithelial , and mesenchymal lines.
Genetically, these tumours are characterized by biallelic inac- Localization
tivation of SMARCB1 (also known as hSNFS, /N/1 , or BAF47) AT/RTs occur throughout the neuraxis . Supratentorial tumours , .
or rarely (in < 5% of cases) of SMARCA4 (BRG1) (CNS WHO which are more common with increasing age 12343). are often
grade 4). located In the cerebral hem ispheres and less frequently in the I

ventricular system, suprasellar region , or pin eal gland. lnfraten-


ICD-0 coding torial tumours can arise in the cerebellar hemisph eres, cerebel-
9508/3 Atypical teratoid/rhabdoid tumour lopontine angle, and brainstem . Spinal cord loc alization i~ rare
{1785,992}. Rare AT/RTs affecting adults tend to occur 1n the
ICD-11 coding cerebral hemispheres and sellar reg ion !515}.
2A00.1Y & XH7Z04 Other specified embryonal tumours of brain
& Atypical teratoid/rhabdoid tumour Clinical features
The clinical presentation is variable , depend ing on th e age of
Related terminology the patient and on the location and size of the tumour. Infants,
None in particular, present with nonspecific signs of leth argy, vomit-
ing, and/or failure to thrive . More spec ific signs inc lude head
Subtype(s) tilt and cranial nerve palsy, most common ly sixth an d seventh
DNA methylation profiling and gene expression profiling dem- nerve paresis . Headache and hemiplegia are more commonly
onstrate three molecular groups of AT/RT, each demonstrating reported in children aged > 3 years .

Molecular
AT/RT-TYR AT/RT-SHH AT/RT-MVC
subtype

Age and
~ 1~
~=
~
·
r:r .
~~» 1· ~ ~~·iI ~
:
I

' I
CT•

~ 0 -
I

- - -·- - -
er l'

at Ol --~----
sex distribution -~~ l s ~ ·~3 1 s ~ ~~ l t a
Age (months) Age (months) ~ Age (months)
An l-(0-1yUIS) 0 Toddlefs(2-5y~) OldercNl-(>3~)
_w
__.. _ec1_1an:ge;12 mo<1ths 6§23 Median.- 20 months Uecbn -U:' 77 montha
51% : 43% 55% =.= 45% 52% : 48%
M : F M • F Ms F

Supraten1Dfi!ll SupratenlOOa( Supraten tonal

Localization 25%~75% 65% ~ 3501 10 5 0 % § J 38%


~lnfratentDrial lntratenlorial lnfralenlonal
12% Spinal
Deregulated BMP signalling Neurogenesis
Melanogenesis SHH signalling MYC
pathways at the (GL/2, BOC, PTCHD2)
(TYR, TYRP, MITF)
gene expression Mesenchymal genes NOTCH signalling HOX duslElf genes
level (07X2, PDGFRB, BMP4) (ASCL.1, CBL , HES1)
MYCN
Overall
methylation
status
Hypermethylation Hypennethylation Hypomethylation
Predominant type Frequent point
Long-stretched deletions Homozygous focal
ofSMARCB1 or chromosome 22 mutations of SMARCB1 loss of SMA RCB 1
alteration

Flg.4.17 Atypical teratoid/rhabdoid tumour (AT/RT) consensus subtypes. Summary of demographic and molecular features of AT/RT subtypes.

Embryonal tumours 221


Etiology
Familial cases arise in the setting of rhabcJ 01d tumour prqrJ,,,
position syndrome 1 (SMARCB1 gene) or 2 (SMARCA1 rJ 8 ,.~ 1
12885,1 453 ,1254). The ri sk of germline mutations 1s r8pr;rt~rJ
to be between 26% and 4·1% 1n SMARCB1-defic1ent t1m1r; 1w
1273,816,992) and may be substantially higher 1n SMAACAa
defi cien t tumours 11254). De nova germl1ne mutations r.a,1 ~.
been described 1816,338) , and they accounted for two thirrJs
of germiine mutations in one study 1816] . Unaffected 8.di;!:
carriers /1 453,101) and gonadal mosaicism 12885) have beqr
report ed .

Pathogenesis
Mutation or loss of the SMA RCB 1 locus at 22q11.2 1s a genetic
hallmark of this tum our /33 18,275) . Whole-genome and whole-
Fig.4.18 Atypica l teratoid/rhabdoid tumour. A Axial T1 -weighted MRI. B Axial T2-
exome sequencing demonstrate remarkably simple genomes
we1ghted MRI demonstrating tumour heterogeneity.
and a mean mutation rate of 0.19 mutations/Mb , with loss of
SMARCB1 being the primary recurrent alteration (> 95% of
cases) /1840,1252,1481). SMARCB1 is a component of the mam-
malian SWl/SNF complex, which re models chromatin . affecting
transcriptional regulation and mediating cell differentiation and
lineage specification /3383 ,92 ,2130). Inactivation of SMARCB1 is
caused by structural variants (partial or complete deletion , copy-
neutral loss of heterozygosity, exon duplication, gene fusion. or
chromosomal inversion) and mutations (insertion/deletion . point
mutation, or frameshift mutation) [273,1053,1481,3215).
Rare tumours(< 5% of AT/RTs) with histopathological features
of AT/RT but retained SMARCB1 protein expression harbour
bialielic inactivation and no expression of the SMARCA4 protein.
another SWl/SN F complex component [2853). These tumours
are associated with very young age and poor prognosis 112541
The specific functions of SMARCB1 and SM ARCA4 , and th eir
roles in malignant transformation , are still not enti rely clear. Loss
of SMARCB1 disturbs the balance between activating SWl/SNF
complex members and the repress ive polycom b complex PRC2
at promoter and enhancer regions (1522 ,3383 ). Analyses of
Flg.4.19 Atypical teratoid/rhabdoid tumour. Tumour with multiple haemorrhagic ar- chromatin states show a complex interplay and divergent roles
eas, arising in the right cerebellopontine angle. for SWl/SNF and polycomb that results in repre ssion of neuronal
differentiation and tumour suppressor genes as well as activa-
Imaging tion of cell-cycle regulatory genes and oncogenes [1522.8561
MRI findings for AT/RTs are similar to those for other embryonal Alterations in SWl/SNF BAF and pBAF subun it com plexes have
tumours . Almost all tumours are variably contrast-enhancing been shown to contribute to the characteristic multilineage dif-
and show isointense or hyperintense signal intensity on FLAIR ferentiation , immune microenvironment, and potential prognosis
images and restricted diffusion [2099) . Differences in con- of these tumours (2385) .
trast enhancement, peripheral tumour cysts , and peritumoural Transcriptome and DNA methylation profiling separate AT,'
oedema have been described across the molecular groups RTs into three molecu lar groups wi th different methylation ana
12289) . transcriptional signatures [3216 ,1481,3215). which by consen·
sus have been designated as AT/RT-TYR, AT/RT-SHH , and AT
Spread RT-MYC [595 ,1320). These gro ups show differences in patient
Seeding of AT/RT via the cerebrospinal fluid (CSF) pathways is age, localization, and SMARCB 1 / chromosome 22 alteration
common and found in approximately one third of all patients at patterns.
presentation [1785 ,992) . AT/RT-S HH tumours (-4 4% of AT/RTs) overexpress proteins
in the SHH and Notch signall ing pathways and genes involvea
Epidemiology in axonal guidance or neuronal development. Localization is
In a US study using data from the Central Brain Tumor Registry most commonly infratentorial (-67%) and otherwise supratento·
of th e United States (CBTRUS), AT/RTs accounted for 1.6% of ri al. Median patient age is 20 months . Compound heterozygous
al l paed iatric CNS tumours and for 10.1% of CNS tumours in SMARCB1 point mu tations are frequently present in this groui:i
child ren aged < 1 year, with an M:F ratio of 1.2:1 [2343) . The 113201.
maiority of pati ents are aged < 2 years , with 33% aged s; 1 year AT/RT-TYR tumours (-34%) demonstrate upregulation ot prLi-
at diagnosis [964,23 43). Occurrenc e in adults is rare [515) . teins in th e melanosomal pathway, includi ng tyros1nase. µrote1115
I

in the BMP pathway; and development-related transcription fac- may be firm and tan-white in some regions . Tumours arising
tors, including OTX2. Localization is predominantly infratento- in the cerebellopontine ang le wrap themselves around cranial
rial, and patients with AT/RT-TYR tumours have the youngest nerves and vessels and invade brainstem and cerebellum to
age at presentation (median age: -12 months). SMARCB1 is various extents. Areas of haemorrhage and necrosis may be
inactivated mostly by mutation in one allele and whole or partial observed .
chromosome 22 loss removing the second allele {1320}.
AT/RT-MYC tumours (-22%) are characterized by expression Histopathology
of the MYC oncogene and HOXC cluster genes . Localization AT/RTs are heterogeneous tumours that can be difficult to rec-
is more commonly supratentorial than infratentorial. Rare spinal ognize solely on the basis of histopatholog1cal findings /419.
AT/RTs are generally AT/RT-MYC , and sellar AT/RTs in adults 2721 ). Characteristically, a population of rhabdoid cells and
also belong to this group {1480). Patients with AT/RT-MYC variable components with primitive neuroectodermal mes-
tumours are significantly older (median age: -27 months) than enchymal , and epithelial features are present Rhabdo1d cells
patients with AT/RT-SHH or AT/RT-TYR /1320} . fall along a spectrum from small cells with scant cytoplasm to
The histogenesis of rhabdoid tumours is unknown . They also large, typical rhabdoid cells with eccentrically located nuclei
occur outside the CNS (in kidneys and soft tissues) . Recent and extensive homogeneously eosinoph ilic cytoplasm Occa-
studies propose a cell of origin for AT/RT-SHH among neural sionally, intracytoplasmic globular eosinoph11ic 1nclus1ons are
progenitors and for AT/RT-TYR and AT/RT-MYC from cells out- present . Nuclei are round and contain vesicular chromatin and
side the neuroectoderm {1472) . Single AT/RTs arising in the set- prominent eosinophilic nucleoli Binucleated elemenrs may be
ting of low-grade glial/glioneuronal tumours, high-grade glioma, found . Cell borders are generally well defined . A frequently
and ependymoma suggest the possibility of progression from encountered artefact is cytoplasmic vacuolation .
other tumour types {80 ,2267,347,257 /. Rhabdoid cells are the exclusive or predominant histopatho-
logical finding in only a minority of cases and may be very rare or
Macroscopic appearance even completely lacking 1n some cases 11208). Small embryonal
The macroscopic appearance of AT/RTs is similar to that of other (medullobl.a stoma-like) cells can be present . rarely alongside
CNS embryonal tumours . AT/RTs tend to be soft and pink-red, H.omer Wnght or Flexner- Winterste1ner rosettes Mesenchymal
and they often appea r to be demarcated from adjacent paren- d1fferent1at1on typically demonstrates a spindle cell morphol-
chyma . Those with substantial amounts of mesenchymal tissue ogy, with cells e1tt1er being dispersed 1n a pale or basophilic
0 ,., :
Flg.4.21 Atypical teratoid/rhabdoid tumour. A Loss of nuclear immunoreactivity for SMARCB1 in tumour cells, with retained expression in endothelial cells. B Rare cases
manifest loss of SMARCA4 (BRG1) immunoreactivity, as illustrated here, but retain expression of SMARCB1 (INl1). C Patchy expression of EMA. D SMA positivity.

mucopolysaccharide-rich matrix or having a compact arrange- have been reported 11254,2029). Rare SMARC81-defic1ent
ment reminiscent of fibrosarcoma. Epithelial differentiation is the non-rhabdoid tumours forming cribriform strands, trabeculae
least common histopathological feature . It can take the form of and well-defined surfaces are recognized as cribriform neu-
papillary structures, adenomatous areas, or poorly differenti- roepithelial tumours {1255).
ated ribbons and cords. Mitotic figures are usually abundant.
Extensive geographical necrosis and haemorrhage are com- Cytology
monly encountered . Most AT/RTs are dominated by embryonal cells with hyperchro-
matic round or oval nuclei and minimal cytoplasm . Rhabdo1d
lmmunophenotype cells are slightly larger than embryonal cells and have an eccen-
AT/RTs demonstrate a broad spectrum of immunoreactivities . trically located nucleus and brightly eosinophilic cytoplasm
Rhabdoid cells characteristically demonstrate expression of Evaluation of CSF cytology is required for staging .
EMA. SMA, and vimentin. lmmunoreactivity for GFAP, NFP,
synaptophysin , and cytokeratins is also commonly observed . Diagnostic molecular pathology
Germ cell markers and markers of skeletal muscle differen- Due to the high risk of germline mutations in the setting of ATi
tiation are not typically expressed . Nuclear loss of SMARCB1 RT, particularly with very young children , molecular analyses
(INl1) protein expression is a highly sensitive marker for the of SMARCB1 or SMARCA4 should be performed and generic
diagnosis of AT/RT. Expression in non-neoplastic nuclei (e.g. counselling and germline analysis recommended . SMARCB 1
within vascular endothelial cells) serves as an internal posi- alterations comprise biallelic structural variations , a structural
tive control {1511) . CNS embryonal tumours without rhabdoid variation combined with a mutation , or compound heterozygous
features but with loss of nuclear SMARCB1 expression qualify mutations (1053,1320).
as AT/RTs {1208) . The three AT/RT subtypes can be identified as d1st1nct
SMARCB1 protein loss may also occur in poorly differentiated molecular groups using gene expression or DNA methylat1on
chordomas and , with a mosaic expression pattern, in schwan - profiling (1481 ,1856). ASCL1 and tyrosinase immunoreact1v1t1es
nomatosis-associated schwannomas {1258,1378). Tumours sus- are potential surrogate markers for AT/RT-SHH and AT/RT-TYR.
pected on morphological grounds of being AT/RT but showing respectively [3216.12591. pending validation studies .
retained SMARCB1 expression should be examined for loss of
nuclear SMARCA4 protein 11251). However, single AT/RTs with Essential and desirable diagnostic criteria
biallelic SMARCA4 inactivation but retained protein expression See Box 4.06.

224 E_1ntry(Jrial tumours


Staging . ..."
Clinical . staging procedures include MRI examinations of the
CNS with contrast agent. This Is complemented by lumbar

puncture postoperative CSF cytology. The postoperative stag - •
ing. syste~ developed . by Chang and others in 1969 (519),
wh.1ch defines the following degrees of metastatic spread , is still
being used :

MO No evidence of subarachnoid or haematogenous


metastasis
M1 Microscopic tumour cells found in the CSF
M2 Gross nodular seeding demonstrated in the cerebellar/

I
cerebral subarachnoid space or in the third or lateral
ventricles
M3 Gross no?ular s.eeding in the spinal subarachnoid space .
M4 Metastasis outside the cerebrospinal axis Fig. 4,;2 Atypical terat~id/rhabdoid tumour. LeptomeningeaJ .tumour spread. Cer-
ebrospinal fluid cytology shows large tumour cells ~ith rhab~o.1d features (extensive
Prognosis and prediction cytoplasm with eccentric nuclei). May-GrOnwald-G1emsa staining.
Overall , the prognosis of patients with AT/RT is poor. However,
data from retrospective studies and clinical trials have shown Box4.06 Diagnostic criteria for atypical teratoid/rhabdoid tumour
that AT/RTs do not always have a dismal outcome. In the Chil-
Essential:
dren's Oncology Group (COG) ACNS0333 trial , a regimen
A CNS embryonal tumour with a polyimmunophenotype
of high-dose chemotherapy with stem cell rescue and radio -
therapy was associated with a 4-year event-free survival rate of AND
37% and an overall survival rate of 43% (2630). A retrospective Loss of nuclear SMARCB1 or SMARCA4 expression in tumour cells
study of children enrolled in the German HIT trial demonstrated OR (for unresolved lesions)
a 3-year overall survival rate of 22% and an event-free survival A DNA methylation profile aligned with atypical teratoid/rhabdoid tumour
rate of 13%, but also identified a subset of patients (14%) who
were long-term event-free survivors {3347). In a small prospec- Desirable:
tive trial incorporating intensive multimodal treatment, including Rhabdoid cells
chemotherapy and irradiation, a 2-year progression-free sur- SMARCB1 or SMARCA4 aJteration
vival rate of 53% ± 13% (standard error) and a projected overall
survival rate of 70% ± 10% (standard error) were found (563}.
Similarly, a retrospective Canadian Brain Tumour Consortium analysed the prognostic impact of molecular group. In the EU-
study reported that high-dose chemotherapy, in some cases RHAB cohort, a non-AT/RT-TYR profile was identified as an inde-
without radiation , resulted in a 2-year overall survival rate of pendent negative prognostic marker. In the COG trial. the 4-year
60% ± 12.6% (standard error) (1785}. The European Rhabdoid survival rate was higher in patients with AT/RT-SHH tumours.
Registry (EU-RHAB) protocol , using an anthracycline-based Thus, although the impact of molecular group on patient outcome
induction and either radiotherapy or high-dose chemotherapy, awaits further validation , it may ultimately be possible to stratify
demonstrated 5-year overall and event-free survival rates of patients with AT/RT on the basis of molecular group, age, tumour
34.7% and 30.5% , respectively (992). site, and extent of resection . Significant immune cell infiltration
The different epigenomic landscapes of AT/RT subtypes could has been reported in AT/RT-MYC and AT/RT-TYR tumours (595,
be associated with distinct therapeutic vulnerabilities (3215,1328, 1856). suggesting that immune checkpoi nt inhibition 1s a poten-
2130}. Both the COG ACNS0333 trial and the EU -RHAB study tial therapeutic strategy for these tumours.

Ernbryon :1I tun ,r.J LJrs 225


Cribriform neuroepithelial tumour Hasselblatt M
CapperD
Jones DTW
Pietsch T

Definition
Cribriform neuroepithelial tumour (CR IN ET) is provisionally
defined as a non-rhabdoid neuroectodermal tumour charac-
terized by cribriform strands and ribbons , and showing loss of
nuclear SMARCB1 expression.

ICD-0 coding
None

ICD-11 coding
2A00.2Y Other specified tumours of neuroepithelial tissue of
brain

Related terminology
None
•-
Flg.4.23 Crlbriform neuroepithelial tumour. A highly cellular tumour composed of
Subtype(s) relatively small cells arranged in cribrlform strands, and trabeculae of varying thick·
None ness, forming well-defined surfaces.

Localization Etiology
CRINET is located in the vicinity of the fourth , third , or lateral SMARCB1 germline alterations (including famil ial cases with
ventricles {1255 ,2399,139,1073). unaffected adult carriers) have been reported {1395,1482).

Clinical features Pathogenesis


The clinical features of CRINET are nonspecific and related to Losses on chromosome 22q affecting the SMARCB1 region
tumour location . are the only recurrent chromosomal alteration {1073,1482) so
far described . SMARCB1 point mutations , as well as deletions
Epidemiology of exons 7 and 8 {1073) or duplication of exon 6 {1395). may
The median age of 10 children harbouring CRINET was represent the second hit.
20 months (range: 10-129 months). The M:F ratio was 1.5:1 On unsupervised clustering analysis of DNA methylation
(1482). profiles, CRINET grouped within the atypical teratoid/rhabdo1d

226 Embryonal tumours


tumour (AT/RT) molecular subtype AT/RT-TYR 114821. The Box4.07 Diagnostic criteria for cribriform neuroepithelial tumour rCRINETJ
observati on that exp.resslon of tyrosinase is present not only in
Essential:
AT/RT-TYR but also in CR INET suggests similarities also at the Highly cellular tumour characterized by the presence of cribriform strands and
protein expression level 11259 ,1482\.
ribbons
AND
Macroscopic appearance
Loss of nuclear SMARCB 1 protein expression of tumour cells
Insufficient data have been reported on the macroscopic
appearance of CRINET specifically.
Desirable:
Distinct expression of EMA highlighting cell surfaces
Hlstopathology
CRINET i~ a_highly cellular tumour charac terized by the pres- caveat: The distinction of CRIN ET from the AT/RT-TYR subgroup ls not fully established.
ence of cnbnform strands and ribbons . In more compact areas
small lumina and true rosettes are also present. but rhabdoid
tumour cells showi ng eosinophilic cytoplasms an d eccentric Cytology
nuclei with prominent nucleoli are not encountered. Not cl inically relevant

Immunophenotype Diagnostic molecular pathology . .


CR INETs should be assessed for SM ARCB1 def1c1ency.
Loss of nuclear SMARCB1 protein expression in tumour cells
is a feature shared with AT/RT (1073,1255\. There is distinct
expression of EMA {1073,1255). and there is frequent expres- Essential and desirable diagnostic criteria
sion of tyrosinase (1482}. MAP2, and synaptophysin as well as See Box 4.07.
vimentin. Expression of 8100 , GFAP, and cytokeratins is vari-
able !1073,1255\. Many CRINETs had initially been diagnosed Staging
as choroid plexus carcinoma {1482\, but staining for choroid None validated
plexus marker Kir7.1 is generally absent {1255}.
Prognosis and prediction .
Many patients with CRIN ET respond well to th erapy and experi-
Proliferation
ence long-term survival. In a retrospective series , the overall
Ki-67/MIB1 proliferation index values between 5% and 35%
survival of 10 patients with CRIN ET was found to be sig nificantly
have been reported {1073 ,1482}. In the largest series reported
longer than that of 27 patients with AT/ RT-TYR (14 82}.
to date, the median Ki -67/MIB1 proliferation index was 29%
(range: 15- 35%) {1482}.

t: 1nbryona1 lu111ours 227


Embryonal tumour with Korshunov A
Fuller Gf\l
Kool M
Sturm D
multilayered rosettes Ha.berl er C
Huang A
von Hoff K
Wesseling p

Definition Related terminology


A CNS embryonal tumour with multilayered rosettes (ETMR) Is Not recommended: embryonal tumour with abundant ne1;ropil
an embryonal neoplasm conforming to one of three morpho- and tru e rosettes (ETANTR) ; embryonal tumour with abundant
logical patterns - embryonal tumour with abundant neuropil and neuropil and ependymoblastic rosettes (ETAf\JER): epencJ;-
true rosettes, ependymoblastoma, or medulloepithelioma - and moblastoma; medulloepithelioma .
typically having a C19MC alteration or (rarely) a OICER1 muta-
tion (CNS WHO grade 4). Subtype(s)
Embryonal tumour with multilayered rosettes , C19MC -altered
ICD- 0 coding embryonal tumour with multilayered rosettes , DICER1-mutated
9478/3 Embryonal tumour with multilayered rosettes
Localization
ICD-11 coding Most ETMRs are intracranial ; examples in the spinal cord
2A00.1Y & XHOKZ2 Other specified embryonal tumours of brain are rare . The most common site is the cerebral hemisphere.
& Embryonal tumour with multilayered rosettes, NOS although 45% arise in non -hemispheric locations . Occasionally
2A00.1Y & XH51C5 Other specified embryonal tumours of brain ETMRs are very large, involving multiple cerebral lobes and
& Embryonal tumours with multilayered rosettes with C19 MC even both hemispheres . Posterior fossa ETMRs occur 1n bot
alteration cerebellum and brainstem , occasionally involving the cerebel-
lopontine cistern (3005 ,2290 ,1798).
01

o.o~,-

~ • p i H
i ~ ~ ~ ~ ~~~§uliti o
flt.4.26 Embryonal tumour with multilayered rosettes. Panchromosomal copy-number alterations shown by DNA methylation array. Amplification and rearrangement of the
C19MC locus on chromosome 19q and gain of chromosome 2 are prominent.

Clinical features Single-cell RNA sequencing data have id~ntified clusters of


ETM.Rs commonly produce symptoms and signs of raised intra- stem cell-like, oligodendrocyte precursor-like , and astrocyte-
c~arnal pressure (e.g . headache, nausea, vomiting, and visual like tumour cell populations (1797) .
d1sturban~es) . A few patients present with epilepsy due to a
small cortical tumour. Focal neurological signs are more com- Genetic profile
mon in older ch ildren and in patients with infratentorial tumours. Approximately 90% of ETMRs harbour specific structural
alterations of a microRNA cluster on chromosome 19q13.42
Imaging (C19MC), including focal high-level amplification , fusion to
The neuroimaging characteristics of ETMR vary; for example, TTYH1 , and other rare rearrangements (e.g. fusion to MY098 or
large cerebral tumours frequently enhance, whereas smaller, MIRLET7BHG), all of which produce strong upregulation of this
cortical tumours may be non-enhancing. Tumours may contain microRNA cluster as a driving event (1650,1797}.
cysts or calcification (2290}. Radiologically, brainstem ETMRs Rare ETMRs (5%) have a DICER1 mutation , and almost all of
may mimic diffuse midline glioma, although they tend to be these are in the setting of a O/CER1 genetic tumour syndrome
more circumscribed . (705}. Typically, the germline mutation is associated with a sec-
ond mutation in the (hotspot) RNase lllb domain that is important
Spread for microRNA processing {1798}. Other ETMRs without a C19MC
Most tumours (75%) are localized at presentation, with the alteration or OICER1 mutation may be driven by amplification of
remainder showing a range of tumour dissemination (stage the miR-17-92 microRNA cluster on chromosome 13 (1798).
M2-M4). Rarely, ETMRs with extracranial invasive growth and Despite ETMR subtypes having different mechanisms
extradural metastases have been reported (2886} . for deregulating microRNA processing , microRNA expres-
sion profiles are nevertheless very sim ilar across ETMRs. In
Epidemiology
The true incidence of ETMR is difficult to determine because of
its rarity. In addition, before its defining genetic alteration was
discovered, multiple diagnostic terms were used for its varied
"•
• .,
• ·>
.. •
·.
·" ...
morphological features. With few exceptions, ETMRs affect chil-
dren aged < 4 years, the vast majority of cases occurring during
the first 2 years of life. In larger cohorts, the incidence seems to
•..
be equally balanced between male and female patients (1798) .
~ ...
.. •
Etiology
No genetic susceptibility for patients with C19MC-altered
ETMRs has been reported. In contrast, almost all patients with
• \ .
DICER1-mutant ETMRs carry a pathogenic OJCER1 germline
alteration, which should prompt genetic testing and counselling
(1797) . No other risk factors have been reported to date.
\ ...
"\ .,

--· "
Pathogenesis
Cell of origin • .
Distinct histological patterns of ETMR share a molecular signa- •
Fig. 4.27 Embryonal tumour with multilayered rosettes. lnterphase FISH. Ampl1f1ca-
ture , which suggests that they have a common histogenesis. t1on (green signals) of the C19MC locus at 19q13.42.
,JI .... ~.

' B ~~- :~-·~-·: .. :~.-..· . .. , -• ., ..


Rg. 4.28_Embryonal tumour with multilayered rosettes (ETMR). A Tumour cells are strongly immunopositive for L\N28A. B Many cells are immunoposlt1ve for v1mentin c Tr~
neuropil-like matrix of ETMRs with an abundant neuropll and true rosettes pattern is synaptophysin-immunopositlve.

a recent study, R loop-associated chromosomal instability Ependymoblastoma


was suggested as a key factor in ETMR pathogenesis !1797). This pattern of ETMR features sheets of embryonal cell s incor-
Other re currently mutated genes, such as CTNNB1 (10%) and porating numerous multil ayered rosettes , but it typicall y lacks a
0
TP53 (7 Yo) , are likely to play a role during ETMR development. neuropil-like matrix and gan glion cell element
~elapsed ETMRs can acquire additional copy-number aberra-
tions (loss .of 6q , gains of 1q or 17q) and increased polyploidy Medulloepithe/ioma
accor:ipan1ed .by chromosomal instability, and they show a This pattern of ETMR is characterized by papillary, tubular or
large increase 1n somatic single-nucleotide variants !1797). trabecular arrangements of neoplastic pseudostratified neu-
roepithel ium with an external (PAS-positive) limiting membrane
Macroscopic appearance resembling the primitive neural tub e. On the lum1nal surface of
ETMR is a grey to pink, well-circumscribed tumour, with areas these tubules, cilia and blepharoplasts are absent. In zones
of necrosis and haemorrhage and , sometimes , minute calcifi- away from tubular and papillary structures, there are large
cations . Some tumours are cystic. Widespread leptomeningeal sheets of embryonal cells. Rare tumours display epithelial
dissemination and extradural metastases are frequent in the myeloid, osteoid , myoid, or other mesenchymal different1at1on.
terminal stage of disease. or they contain melanin pigment !75 ,397).
All morpholog ical patterns of ETMR show abundant m1tot1c
Histopathology figures and apoptotic bodies , indicating a high rate of cell turno-
By definition , rosettes are a characteristic histological fea- ver. The Ki-67 proliferation index ranges from 20% to 80% . After
ture of ETMRs. They are multilayered structures consisting therapy, some ETMRs show neuronal and glial different1at1on.
of embryonal cells in a pseudostratified neuroepithelium with resembl ing a low-grade glioneuronal tum ou r /806 ,1784.1191
a central round or slit-like lumen. The cells facing the lumen During progression , other tumours show comp lete loss of rec-
have a defined apical surface, with a prominent internal limiting ognizable ETMR patterns and instead resemble other embryo-
membrane. The nuclei of the rosette-forming cells tend to be nal neoplasms (3465) .
located away from the lumen towards the outer cell border. In
most tumours , a defined outer membrane around the rosettes is /mmunophenotype
absent. Mitotic figures are commonly observed in the embryo- Most of the embryonal component of ETMR (the roset tes
nal cells of rosettes. ETMRs have three histological patterns: and tubular structures) is intensely immunoreactive for nest1n
embryonal tumour with abundant neuropil and true rosettes, and vimentin (817,1068) . Embryonal cells and rosettes may
ependymoblastoma, and medulloepithelioma. On the basis of also show focal expression of cytokeratins (particularly 1n ihe
their molecular commonality, all three are now considered to medulloepithelloma pattern), EMA. and CD99 , but they are
constitute various points along a morphological spectrum of usually negative for neuronal and glial markers . In contrast, the
diverse differentiation within a single tumour entity, rather than neuropil-like matri x is strongly immunopositive for synaptophy-
distinct nosological tumour types /1510 ,1725]. sin, NFP, and NeuN . lmmunoreactivity for GFAP highlights sca1-
tered cells resembl ing reactive astrocytes , but it may also be
Embryonal tumour with abundant neuropil and true rosettes present in a few embryonal cells . ETMRs show strong and dif-
This pattern of ETMR shows a biphasic architecture featuring fuse nuclear immunoreactivity for SMARCB1 (INl1) throughout
densely packed , small embryonal cells with round or polygonal all components . Strong and diffuse cytoplasmic immunoreac-
nuclei and scant cytoplasm, as well as large, neuropil-like areas tivity for LIN28A is found in ETMRs irrespective of their morpho-
with sparse neoplastic neurocytic and ganglion cells /817,1068) . logical pattern (1725 ,30051. However, LIN 28A expression also
In some cases , the neuropil has a fascicular quality. Multilayered occurs in some gliomas, atypical teratoid/rhabdo1d tumours.
rosettes are often present among the embryonal cells , although germ cell tumours , teratom as, and some non-CNS neoplasms
in some cases they are observed in the otherwise paucicellular [3005,3411) .
neuropil -like areas .

230 ErntA yo11ul I umours


Dlfferent1al diagnosis Box 4.o8 Diagnostic cnteria for embryonal urnour with rnult1layered rosettes IE-MR 1
lntraocular medulloepithel iorna and sacrococcygea l ependy-
moblastorna share some histopathological features with CNS Essentfal:
A CNS embryonal tumour with the morphological and immunoh1s!ochemteal
ETMR bu t harbou r striking molecular differences and conse -
features of one of the three ETMR morphological patterns·
quently deserve a separate nosologic al desi gnation .
• Embryonal tumour with abundant neuropil and true rosettes
Cytology • Ependymoblastoma
Evaluation ?f tumour cell presence in cerebrosp1nal fluid cytol- • Medulloeplthelioma
ogy 1s required for stag ing . ANO
Genetic alteration defining one of the two ETMA molecular subtypes·
Diagnostic molecular pathology
• C19MC alteration
A limited range of recurrent molecular alterations is displayed
• D/CER1 mutation

I
by ETMRs. To date, structural variants of the C19MC microRNA
ANO (for unresolved lesions)
cluster at 19q13.42 have been found on ly in ETMRs , occurring
A DNA methylat1on profile aligned with ETMR
In a~proximately 90% of cases . These are usually foca l ampl i-
fications , but fusions can also occur, generally with TTYH1 .
C19M C _ alterations c an be detected by array-based copy- num-
ber prof1lrn g or interphase FI SH. It should be borne in mind that postoperative stag in g system developed by Chang and othe'.s
the ch arac~eristic morphological features of ETM R, including in 1969 1519). wh ich defines the following degrees of metastatic
rosettes , mig ht not always be present in tissue submitted for his- spread , is still be ing used:
topatholog ic al exam inati on. The com bination of LIN28A immu -
noreactivi ty and C19MC alterations by interphase FISH can be MO No evidence of su barachnoid or haematogenous
helpful in this situation . However. although C19MC alterations metastasis
are specific for ETMR , immunoreactivity for LIN 28A is not. M1 Microscopic tumour cells found in the _ cerebrosp1nal fl uid
About half of the ETMRs without a C19MC alteration harbour M2 Gross nodular seed ing demonstrated 1n the cerebellar/
DICER1 mutations. These are generally compound heterozy- cerebral subarac hnoid space or in the th ird or lateral
gous mutations , combining one somatic mutation (usually in ventricles
exon 24 or 25) and a second mutation in the patient's germline. M3 Gross nodular seed ing in th e spinal subarachnoid space
Germline testing for a DICER1 mutation should be undertaken M4 Metastasis outs ide the c erebros pinal axis
in patients with an ETMR that lacks a C19MC alteration . Not all
high-grade neuroepithel ial tumours with a OICER1 mutation are Prognosis and prediction
ETMRs; a DICER1 mutation can be found in other embryonal ETMRs demonstrate rap id growth and are associated with an
tumours and gliomas. Rare ETMRs without a C19MC alteration aggressive clin ical course , w ith repo rted survival times aver-
or DICER1 mutation should be classified as ETMR not else- ag ing 12 months after intensive com bination therapies 11068,
where classified (NEC). 1060,3004,1725,1352). Few prog nostic indicators have been
rel iably identified . Gross total rese ction , radiotherapy, and high-
Essential and desirable diagnostic criteria dose chemotherapy probably prolon g overall su rvival 11456}.
See Box 4.08 . Metastatic disease at presentatio n and brainstem tumours have
been significantly associated wi th an adverse outcome. Patient
Staging survival does not differ significantly between the three morpho-
Cl inical stag ing procedures include MRI examinations of log ical patterns of ETMR . From extremely rare cases with long-
the CNS with contrast agent. This is complemented by lum- term survival , posttreatment neuronal differentiation has been
bar puncture postoperative cerebrospinal flu id cytology. The proposed as a favourable indic ator of outcome {806 ,1784,119)

Ernbryoncll tu rnour::. 23 1
Wesseling P Korshunov A.
CNS neuroblastoma , FOXR2-activated Haberler C Sturm D
Huang A von Hoff K
KoolM

ICD-11 coding
2A00.1Y & XH85ZO Other specified embryonal tumours of bra1r
& Neuroblastoma, NOS

Related terminology
None

Subtype(s)
None

Localization
FOXR2-activated CNS neuroblastoma is typically located 1n the
cerebral hemisphere, with intraventricular location observed 1n
occasional cases [3059,2502}.

Ag.4.29 CNS neuroblastoma, FOXR2-activated. A Axial T2-weighted image show- Clinical features
ing a large, partially cystic mass involving the right parietal lobe (left). B Postcontrast Clinical data on FOXR2-activated CNS neuroblastoma, as diag-
T1-weighted image showing thickening and enhancement of overlying meninges as
nosed by current molecular criteria, are limited . Leptomeningeal
well as inhomogeneous enhancement of solid tumour components.
metastasis develops in some cases \3059,2502) .

Definition Imaging
CNS neuroblastoma, FOXR2-activated, is an embryonal neo- FOXR2-activated CNS neuroblastoma usually appears as a
plasm exhibiting varying degrees of neuroblastic and/or demarcated mass in a cerebral hemisphere. There may be a
neuronal differentiation, including foci of ganglion cells and prominent cystic component. The solid component may show
neuropil-rich stroma . It is characterized by activation of the moderate and heterogeneous enhancement (3059,2502,1016.
transcription factor FOXR2 by structural rearrangements (CNS 1450,1336}.
WHO grade 4).
Epidemiology
ICD-0 coding FOXR2-activated CNS neuroblastoma is a rare, recently
9500/3 CNS neuroblastoma, FOXR2-activated described tumour, and epidemiological data are incomplete.

B Some tumours show focal ganglion cell lonnatJon

232 Ernb ryona l tumours


However, th ese tumours usually occur in childhood , with a Macroscopic appearance
slight female preponderance {3059). FOXR2-activated CNS Insufficient data have been reported on the macroscopic
neuroblastoma may represent approximately 10% of tumours appearance of FOXR2-activated CNS neuroblastoma spe-
previously c lassified as primitive neuroectodermal tumour of the cifically. However, CNS embryonal tumours are typically well-
CNS {3059,1389). circumscribed pink masses . They are soft, unless they contain
a prominent desmoplastic component, in which case they are
Etiology firm and often have a tan colour.
No risk factors have been reported to date.
Histopathology
Pathogenesis In many microscopic reg ions, FOXR2-activated CNS neuro-
The exact cellular origin of CNS neuroblastomas rem ains blastoma consists of sheets of poorly differentiated cells with a
unknown . The most fre quent genetic alterations in tumours hi gh N:C ratio; round , oval , or angulated hyperchromatic nuclei ;
classified morphologically as CNS neu roblastoma are com - and abundant mitotic activity. Infiltration of adjacent CNS paren-
plex interchromosomal and intrachromosomai rearran gements chyma is variable. Areas of necrosis are common . Palisades of
converging on the transcription factor gene FOXR2. producing embryonal cell s and Homer Wright (neuroblastic) rosettes may
a fusion between the entire FOXR2 gene an d different gene be evident. Ce ll s showin g neurocytic differentiation and mature
partners {3059). The elevated FOXR2 expression levels in CNS ganglion cells may be present, often in clusters (known as gan-
neuroblastomas, compared with its expression in other CNS glioneuroblastomas).
tumour types and non-neoplastic brain tissue , are suggestive
of FOXR2 activation facilitated by promoters of active genes lmmunophenotype
(possibly through enhancer hijacking) [3059) . Although FOXR2 Most cells in FOXR2-activated CNS neuroblastomas strongly
has been demonstrated to play a causative role in the formation expres~ OLIG 2. lmmunoreactivity for synaptophysin can be
of CNS embryonal tumours, the exact underlying mechanisms fou nd 1~ son:e embryonal cells, but its expression 1s accen -
have yet to be elucidated !2528}. tuated in reg ions of neurocytic or ganglion cell differentiation
[1336 ). lmmunoreactivities for GFAP and vimentin are generally
absent, although .staining for the former can pick out reactive
astrocy tes. The Ki-67 proliferation index is high .

Embry )nal t1jrnours 233


Box4.09 Diagnostic criteria for CNS neuroblastoma, FOXR2-actlvated includes FOXR2-activated CNS neuroblas~omas is d1st1n<:,i
therefore, DNA methylation profil ing can facilitate the diagnosis
Essential:
of these tumours l3059,460l .
An embryonal tumour with foci of neuroblastlc or neuronal differentiation
AND Essential and desirable diagnostic criteria
Activation of FOXR2 by structural rearrangement and gene fusion See Box 4.09 .
OR (for unresolved lesions)
A DNA methylation profile aligned with CNS neuroblasloma, FOXR2-activated Stag ing .
Cl inical stagi ng procedures include MRI exam1nat1ons of ·ri~
CNS with contrast agent. This is complemented by lurnb::ir
Cytology puncture postoperative cerebrospinal fluid cytology The post-
Evaluation of cerebrospinal fluid for the presence of tumour operative stag ing system developed by Chang and others r
cells is required for staging . 1969 1519), wh ich defin es the following degrees of metastat1r:
spread , is still being used:
Diagnostic molecular pathology
Structural rearrangements of FOXR2 are frequent and usually MO No evidence of su barachnoid or haematogenous
complex , requiring next-generation sequencing methods for metastasis
detection . Furthermore, alterations affecting the FOXR2 locus M1 Microscopic tumou r cells found in the cerebrospinal fluid
on chromosome Xp11 .21 may be visible by copy-number M2 Gross nodular seedin g demonstrated in the cerebellar/
analysis. Gain of chromosome 1q is present in most tumours cerebral subarach noid space or in the third or lateral
13059). ventricles
Most CNS embryonal tumours that are not classified as M3 Gross nodular seedi ng in the spinal subarachnoid space
medulloblastoma, embryonal tumour with multilayered rosettes , M4 Metastasis outside the cerebrospinal axis
atypical teratoid/rhabdoid tumour, or pineoblastoma are
FOXR2-activated CNS neuroblastomas . However, high FOXR2 Prognosis and prediction
expression also occurs in a subset of high-grade gliomas and Limited information is avai lable on the prognosis of FOXR2-
(rarely) in medulloblastoma. The DNA methylation cluster that activated CNS neuroblastoma.

234 f rnbryonal tu mours


CNS tumour with We sseling P
Haberl er C
Korshunov A
Solomon DA
BCOR internal tandem duplication Huan g A
Kool M
Sturm D
von Hoff K

Definition
CNS tumour with BCOR internal tandem duplication (ITD) is a
malignant CNS tumour characterized by a predominantly solid
growth pattern , uniform oval or spindle-shaped cells with round

I
to oval nuclei, a dense capillary network, focal pseudorosette
formation , and an ITD in exon 15 of the BCOR gene .
.
tCD-0 coding
9500/3 CNS tumour with BCOR internal tandem duplication

JCD-11 coding
2A00.1Y Other specified embryonal tumours of brain

Related terminology Flg.4.32 CNS tumour with BCOR internal tandem duplication . A Axial MRI/FLAI R
Not recommended: CNS high-grade neuroepithelial tumour image of a tumour in the basal ganglia. B Coronal T2-weighted MRI of a tumour lo-
with BCOR internal tandem duplication . cated in the cerebellar hemisphere.

Subtype(s) Clinical features


None The clinical presentation is based on the location of the tumour
and encompasses symptoms of raised intracranial pressure
Localization (e.g. headache, vomiting, nausea, vi sual distu rb ances), focal
CNS tumour with BCOR ITD most commonly occurs in a cere- neurological deficits , and seizures .
bral or cerebellar hemisphere. Occurrence in the basal ganglia,
cerebellopontine angle, brainstem, or spinal cord has rarely Imaging
been described {3059,124,3547,918 ,710}. On MRI , these tumours are typically wel l demarc ated . They often
have a central cystic component and variable, inhomogeneous

Fn1bryor1al t1H11uur~~ 235


contrast enhancement. Tumours can be very large at presen - that probably contribute to pathogenesis in som e cases include
tation , involving multiple lobes and both cerebral hemispheres inactivating mutations in other genes , such as EP300
[918,368}. Location adjacent to the dura has been described SMARCA 2, STAG2, an d BCORL1 [918 ). BCOR expression 1s
for most of the neuroradiologically annotated cases , although higher in th is tu mour type than in most other CNS tumours, and
definite dural infiltration has not been observed (918}. activation of th e WNT signalling pathway is freq uently observed
(3059).
Spread The exact natu re of CNS tumours with BCOR ITD is not clear
So far, no patient with metastatic disease at presentation has The dupl icated region in exon 15 of BCOR is identical to that
been described. At relapse , leptomeningeal metastases, as of BCOR ITDs in c lear cell sarcomas of th e kidney, undiffer-
well as metastases to the bone and inoculation metastases entiated round cell sarcomas in infants , and primitive myxo1d
along the neurosurgical access route , have been observed mesenchymal tum our of infancy (3250 ,1553,2807). There 1s
1124,2392,1638,918}. currently no consensus as to whether CNS tumours should be
considered mesenchymal or neuroepithel ial neoplasms.
Epidemiology
Limited d ata are available, but the med ian age at presentation Macroscopic appearance
of reported pati ents is 3.5 years (range: 0-22 years), with a bal- Insufficient data have been published on this tumour 's macro·
anced M:F ratio {918). scopic appearance .

Etiology Histopathology
There is no known specifi c risk factor or genetic susceptibility CNS tumours with BCOR ITD are generally demarcated at the
for CNS tumours with BCOR ITD (918} . interface with adjacent CNS parenchyma, although infiltra!Jon
of the brain may sometimes occur [3547,918). Histolog1cal
Pathogenesis features can be variable (1 24,3547,9 18,1808). Tumours are
A somatic heterozygous ITD in the BCOR g ene plays a key generall y composed of un iform oval or spindle-shaped cells,
oncogenic role in the pathogenesis of this tu mour type [3 059). with round or oval nuc le i showing a delicate chromatin pattern
The majority of tumours harbou r a BCOR ITD as the solitary The cytoplasm is weakly stained with eosin . Most cases dem·
pathogenic alteration . Although it is very likely that the ITD in onstrate dispersed glioma- like fibrillary processes. but a com·
BCOR produces an activating , gain-of-function event, the spe- pact fascicular pattern can occur. A characteristic feature is tha
c ific mechan ism by which this recurrent alteration drives tu mour formation of ependymoma-like perivascular pseudorosettes A
development remains unknown . Ad ditional genetic alterations myxoid or microcysti c matrix is often encountered A branching

236 Embryona l tumou rs


capillary network is often present . Glomeruloid microvascular aox 4•10 Diagnostic critena for CNS tuwour NJth BCOR 1merria11ar&:.!rr 1'. .(I. ...a• -:,r
proliferation is not a typical feature . Necrosis, which is often pali-
sading . is commonly observed . Mitoses are frequently encoun-
tered , including foci with brisk mitotic activity. The differential
diagnosis includes glioma, ependymoma, and other embryonal
tumours.

lmmunophenotype An internal tandem dup6catio tn exon 15 BCOR


In the immunohistochemical assessment of CNS tumours with AND (for unresotved Jons)
............... .,_·m BCOR
BCOR ITD, expression of vimentin and CD56 is universal. A few A DNA me1hytalion pro afig ed . CNS
tumour cells can be immunopositive for OLIG2, GFAP, or S100 tandemdu ·
1918}, but t~eir widespread expression, as found in gliomas, is
absent. Penvascular pseudorosettes are GFAP-negative. unlike
in ependymomas . Variable expression of NeuN is detected in Essential and desirable diagnostic criteria
a few tumours 11662,124,3547). Neuronal markers are gener- See Box 4.10.
ally not expressed . Widespread strong nuclear expression of
SCOR is a sensitive marker, but it is not specific because it
may also occur in other tumours, such as solitary ·fibrous tumour
1124,3547,1209,918) . The Ki -67 labelling index is elevated and
ranges between 15% and 60% 1918,124).

Grading
lnsutficient data currently exist to assign a CNS WHO grade to
this tumour.
MO No evidence of s barac oid r aer-a:~ge,.... ~S
Cytology
Evaluation of cerebrospinal fluid for the presence of tumour M1
cells is required for staging. M2 ... :,..., 0 ;:e':::~~ 2-'

Diagnostic molecular pathology ventricles


The defining diagnostic finding is a heterozygous ITD in exon 15 M3 Gross nod la
of the BCOR gene. This BCOR ITD appears to be mutually e elu- M4 etastasis
sive with BCOR or BCORL 1 fusions and hotspot mutations in H3
or IDH genes (as found in infiltrating gliomas), ZFTA (C11orf95)
or YAP1 tusions (as found in supratentorial ependymomas , or
alterations in FOXR2 or MN1 (as found in other neuroepithelial
tumours) {3059.3218) . DNA methylation and gene e pression
profiles can be reliably used to differentiate CNS tumours with
BCOR ITD from other CNS tumours {3059,460}.
Wes sel1nq P VorshunrJ / I•
CNS en1bryonal tumour NEC/NOS Haberler C S urrn D
Hu;:rng A mn H r;ff V
Kool M

Definition
CNS embryonal tumour NEC/ NOS Is a tumour arising in the
~NS with ~mbryonal morphology and immunophenotype and
either lacking an alteration that would classify it as one of the
molecularly defined CNS embryonal tumours (not elsewhere
classified ; NEC) or not susceptible to further analysis (not other-
wise specified. NOS) .

ICD-0 coding
9473/3 CNS embryona l tumour, NEC/NOS

ICD-11 coding
2A00.1Y & XH8SH6 Other specified embryonal tumours of brain
& CNS embryonal tumour, NOS

Related terminology
Not recommended: primitive neuroectodermal tumour.

Subtype{s)
None

Localization Flg.4.35 CNS embryonal tumour. Postcon tras t. T1-we1ghted MRI showing a iarge
partly solid, partly cystic tumour in the left fron tal lobe of an infant. This 1s :~e
CNS embryonal tumours occur throughout the neuraxis , but same patient of whom the histology and copy-numbe r vari ation profile are shown
most are supratentorial. in Fig. 4.36.

Clinical features
Patients with CNS embryonal tumours generally present acutely cystic or necrotic areas . Most tumours show contrast enhance-
with symptoms and signs of raised intracranial pressure, epilepsy, ment and restricted diffusion (1450) .
or focal neurological deficit. Metastatic dissemination is evident
in 25- 35% of CNS embryonal tumours at presentation (1347). Epidemiology
Shifting diagnostic criteria and nomencl ature complicate analy-
Imaging sis of the epidemiology of the spectrum of CNS embryona1
Appearances on MRI can vary, depending on the site of origin. tumours. Although most of these tumou rs occur in infancy ana
CNS embryonal tumours often appear solid but may contain childhood , some are diagnosed in ad ults (16 18,233,10761.

: .. .. :: :': . i '!: j ,:
I. t. .. ,. - ! ;,.,)

B
Fig. 4.36 CNS embryonal tumour. A CNS embryonal tumour (from the same patient shown in. Fig..4.35) consis.ting of densely packed, small, poorly d1fferent1ated cells wit~ 3
high mitotic count. RNA sequencing revealed a BRD4::CREBBP fusion gene. Methylat1on prof1hng did not result 1n a match using the brain tumour class1f1er available at http~.
www.molecularneuropatholog y.org. B The copy-number variation profile of the tumour in panel A .(derived from genome-wide methylome analysis) did not reveal large chroino·
somal gains or losses. The tumour was classified as CNS embryonal tumour not elsewhere class1f1ed (NEC).

238 l 11i!ir /G l1 ctl l l J1 r1 u J1:,


Etiology Box4.11 Diagnostic criteria for CNS embryonal tumour NEC/NOS
No risk factors have been reported to date.
Essential:
Pathogenesis An embryonal tumour originating in the CNS
By definition.' CNS ~mbryon~I tumou:s NEC/NOS are distinct AND
from those with spec1f1c genetic alteration s; molecular drivers for Absence of criteria qualifying for the diagnosis of a more specific type of embryo-
this heterogeneous group of tumours remain to be elucidated . nal CNS tumour

Macroscopic appearance Desirable:


CNS embryonal tumours are often circumscribed and sol id, but Focal expression of neuronal markers and absence of glial markers
they can contain cystic areas , haemorrhage, and necrosis.

Hfstopathology
CNS embryonal tumours are characterized by sheets of densely
ol f exlc luslon, pehndtingh a
ar a terat1ons t a· c arac enze
grtea~er uthn1~serhsetatenrdoginegneoof uthsegmrooulepcuo~ , •...
~acked , immature cells that have a high N:C ratio and round , tumours . Examples with rare gene fusions are emerging , such
oval. or angulated hyperchromatic nuclei. Foci of neurocytic or as tumours wi th a BR04:: CREBBP fusion or ganglioneuroblas-
ganglion cell differentiation can be present. Mitotic activity is tomas with a M Y05A: :NTRK3 fusion (1425) . However, when
typically high . Infiltration of adjacent CNS parenchyma is vari- molecular analysis of CNS embryonal tumours fails to detect an
able. Areas of necrosis and haemorrhage may be present. The alteration that allows specific classification or is unsuccessful ,
heterogeneous nature of these high-grade tumours and their the designations "not elsewhere classified (NEC)" or :·not other-
variable behaviours do not readily allow distinction between wise specified (NOS)", respectively, should be applied (1946,
CNS WHO grades 3 and 4. 1934].

/mmunophenotype Essential and desirable diagnostic criteria


The neoplastic cells in CNS embryonal tumours are variably See Box 4.11 .
immunopositive for synaptophysin and OLIG2. They demon-
strate a high Ki-67 proliferation index. GFAP and vimentin are Staging
generally absent, although staining for the former can pick out Clinical staging procedures include MRI examinations of the
reactive astrocytes. CNS with contrast agent. This is complemented by lumbar
puncture postoperative cerebrospinal fluid cytology. The post-
Differential diagnosis operative staging system developed by Chang and others in
The differential diagnosis of CNS embryonal tumours encom- 1969 (519}, wh ich defines the following degrees of metastatic
passes many poorly differentiated embryonal tumours and spread , is still being used :
high-grade gliomas with specific molecular alterations. Just
a few examples are FOXR2-activated CNS neuroblastoma, MO No evidence of subarachnoid or haematogenous
atypical teratoid/rhabdoid tumour, and H3 G34-mutant diffuse metastasis
hemispheric glioma, all of which can have similar morphologi- M1 Microscopic tumour ce ll s found in the cerebrospinal fluid
cal features , especially in limited biopsies . Ideally, the diagnosis M2 Gross nodular seed ing demonstrated in the cerebellar/
of CNS embryonal tumour should be made by excluding other cerebral subarachnoid space or in the third or lateral
morphologically similar but molecularly distinct tumour types . ventricles
M3 Gross nodular seed ing in the spinal subarachnoid space
Cytology M4 Metastasis outside th e cerebrospinal axis
Evaluation of cerebrospinal fluid for the presence of tumour
cells is required for staging. Prognosis and prediction
As we understand more abou t the heterogeneity of tumours
Diagnostic molecular pathology with an embryonal morphology, historical data on the outcome
CNS embryonal tumour NEC/NOS shares a morphology and of patients with these tumours (which have included types of
immunophenotype with many other poorly differentiated poorly differentiated high-grade glioma) become less repre-
embryonal tumours and high-grade gliomas and is a diagnosis sentative of sp ecific tumour types (3059,1389).

Ernbryor1dl ru •nuu r:: , 239


Hasselblatt M
Pineal tumours: Introduction

Pineal region tumours encompass a heterogeneous group of Molecular profiling may also provide important prognostic
relatively rare neoplasms attecting different age groups . Pin- information. An example is the segregation of pineoblastoma into
eal parenchymal tumours include pineocytoma (CNS WHO four subtypes showing distinct molecular and clinical features
grade 1), pineal parenchymal tumour of intermediate differenti- (1) pineoblastoma, mi RNA processing-altered_1, arises in chil-
ation (PPTID ; CNS WHO grade 2-3) , and pineoblastoma (CNS dren and is characterized by mutations of DICER1, DROSHA , or
WHO grade 4). Papillary tumour of the pineal region (CNS OGCRB, as well as by intermediate outcome; (2) p1neoblastoma.
WHO grade 2-3), a neuroepithelial tumour thought to origi- mi RNA processing-altered_2 , mainly occurs in older children and
nate from specialized ependymal cells of the subcommissural is also characterized by OICER1, OROSHA, or DGCRB mutations,
organ , had already been included in previous editions of the but in this subgroup, outcome is excellent ; (3) pineoblastoma.
WHO classification of CNS tumours {1502). but desmoplastic MYC/FOXR2-activated , arises in infants and is characterized by
myxoid tumour, SMARCB1-mutant, a rare SMARCB1-mutant MYC activation and FOXR2 overexpression, as well as by a gen-
tumour lacking histopathological signs of malignancy (3185). erally poor prognosis; and (4) pineoblastoma, RB1-altered , arises
is new to this edition and has not yet had a CNS WHO grade in infants and shows similarities with retinoblastoma , with frequent
assigned . metastatic spread and a very poor prognosis {2490 ,1865,19131.
Wh ile histological grading criteria for PPTID; papillary The main clinical features of the pineoblastoma subtypes
tumour of the pineal region ; and desmoplastic myxoid tumour, and PPTID, as well as their genetic alterations and associated
SMARCB1-mutant, remain to be defined , molecular studies play hereditary cancer predisposition syndromes , are summarized
a more important role in the diagnosis of pineal region tumours . in Fig . 5.01. The distinct subtypes are illustrated in the !-distrib-
This is exemplified by the demonstration of KBTBD4 in-frame uted stochastic neighbour embedding (t-SNE) representation of
insertions, which is now a desirable criterion for the diagnosis pineal tumour DN A methylation profiles in Fig . 5.02.
of PPTID {1834).

"
"'

"'t. ~t. ~Tt
Gender
-'
Aqa (median. yr)

)..11 ,.,,
8.5

1:1 .6
,.._

11 .6

1.6:1
~
~,t
1.3

3.3:1
-- ~t 2.1

1:1
tT•33,0

1:1.3
20 J
: PB-RB_1 I
- -----

-,.
(M:F)

10

,.,_
Cencer DICER1 DICER1 litrodtory
pt9d1opolltion synaomo lynd'OIM t1Vnoblalloma

RBI KBT804


Genomic/ DICER! DICER1 FOXR2
LPnprrtarylumour 011110 p111~a1 m~1011 A.I

-
lnlnta1pl0mle DROSHA DROSHA WW.J:p'-ion Ktlal domlln 0
profile DGCRB ln..•ri
................. io..a'"""'dorl
MYC ml!f.171V2
~ Oesmoptastro myxokl lumour. SMARCS 1-mulanl
.J-
CytoglMOCI
7• 12• 17•
=<: =<: =<: ~
lltloncod
=<: , •.k.. / k f 111

/· ~
14- t eq- 1-.
- 10
\.
.Jt. Prn~.~·11 $
_ ;~-C.bht=~
t •rri.1

,~oa1 e 1":' l Papliiiiry lumuur OltM pln&.11 ro~lun fl


, 67.6'11 , fOO'll , 206'11 , 2U 'll , 86.1'11
Ult • 1.,. • -40 - 20 IQ

Fig. 5.01 Pineoblastoma subtypes and pineal parenchymal tumour of intermediate Fig. 5.02 Pineal tumours. t-distribuled stochastic neighbour embedding (t-SNEI
differentiation (PPTID). Main clinical features, genetic alterations, and association representation of DNA methylation profiles. PB-m1RNA 1, pineoblastoma. m1RNA
with hereditary cancer predisposition syndromes. OS, overall survival; PB-miRNAl , process1ng-altered_1; PB-miRNA2. pineoblasto111a, miR NA process1ng-altered_2;
pineoblastoma, m1RNA processing-altered_1; PB-mlRNA2, pineoblastoma, rniRNA PB-MYC/FOXR2, pineoblastorna, MYC/FOXR2-act1vated, PB ·RB1 . prneoblastonia.
processing-altered_2; PB-MYC/FOXR2, pineoblastoma, MYC/FOXR2-activated· RBI-altered; PPTID, pineal parenchymal tumour o! 1nte1med1ate d11lerent1arion
PB-RB1 , pineoblastoma, RBI-altered. '

242 P111e3I tumours


Pineocytoma Hasselblatt M
Huang A
Jones OTW
Orr BA
Snuderl M
Vasiljevic A

Definition
Pineocytoma is a well -differentiated pineal parenchymal neo-
plasm composed of (1) un iform c ells form ing large pineocy-
tomatous rosettes and/or (2) pleomorphic cells showing gan-
gliocytic differentiation (CNS WHO grade 1).

ICD-0 coding
9361 /1 Pineocytoma

ICD-11 coding
2A00.20 & XH1 K94 Tumours of the pineal gland or pineal region
& Pineocytoma

Related terminology
None

Subtype(s)
I
None

Localization Fig. 5.03 Plneocytoma. Sagittal , gadolinium-enhanced , T1-weighted MRI showing a


Pineocytomas typically remain localized in the pineal area. well-delineated tumo ur with strong contrast enhancement.
Compression of adjacent structures and protrusion into the pos-
terior third ventricle are common . hyperintense on T2-weighted images , with strong , homoge-
neous contrast en hancement (2203}. They can usually be easily
Clinical features distinguished from pineal cysts (885 ).
Patients with pineocytomas present with signs and symptoms
related to increased intracranial pressure due to aqueductal Spread
obstruction, neuro-ophthalmological dysfunction (Parinaud Pineocytomas g row locally and are not associated with cerebro-
syndrome), and brainstem or cerebellar dysfunction {325 ,578 , sp inal fluid seed ing {896) .
600,1219 ,2839).
Epidemiology
Imaging Pineal region tumours are rare and account for < 1% of all 1ntra-
On CT, pin eocytomas are usually globular, well -delineated cran ial neopl asms; approximately 27% of pineal region tumours
masses that appear hypodense and homogeneous, some are of pi neal parenchymal origin (1700 ,3074). and approxi -
harbouring calcific ation (571). On MRI , the tumours tend to mately 25% of these are pineocytom as (137,1505 ,2839 ,35101.
be hypointense or isointe nse on T1 -weighted images and Pineocytomas can occur at any age. but they most frequently

.
Fig. 5.04 Pineocytoma A Large fibrillary pineocyromatous rosettes are a character1s11c feature. B Pineocytomatous rosettes are large irregula . h·1· f b ·11 -
· 1 1 d h r eos1nop 11c 1 ri ary areas
surrounded by pinealocyte -like neoplastic cells. c Pleomorph1c pineocytoma. Mu tinuc eate p1eornorp 1c cell

P111eal tumours 243


· ----
Fig. 5.05 Pineocytoma. A lmmunoreactivity for synaptophysin is diffuse and highlights the plneocytomatous rosettes. B NFP immunoexpression is especially strong
pineocytomatous rosettes. C Pleomorphic pineocytoma. Pleomorphic cells often show immunoreactivity for NFP.
in the

affect adults, with a median patient age of 44 years (range : optimally demonstrated by neurof ilament immunostaining or
1.1-85 years) 1137,1505,2119.2839 ,3510) . There is a female silver impregnation . Pineocytomatous rosettes vary in number
predominance , with an M:F ratio of 0.6:1 . and size. Their anucleate centres are composed of delicate,
enmeshed cytoplasmic processes resembling neuropil 1325,
Etiology 1505,2291,2839). The nuclei surrounding the periphery of the
There are no syndromic associations or genetic susceptibili- rosette are not regimented .
ties reported. Occurrence of pineocytoma in siblings has been A pleomorphic cytological pattern is encountered in some
reported in one family 11056). pineocytomas (924). This pattern is characterized by large gan-
glion cells and/or multinucleated giant cells with bizarre nuclei
Pathogenesis 11751 ,2067,2839}. The stroma of pineocytoma consists of a
Cefl of origin delicate network of vascular channels lined with a single layer of
The histogenesis of pineal parenchymal tumours has been endothelial cells and supported by scant reticulin fibres. Micro-
linked to the pinealocyte. a cell with photosensory and neuroen- calcifications are occasionally seen but usually correspond to
docrine functions . Microarray analysis of pineocytoma showed calcifications of the remaining pineal gland .
high-level expression of genes coding for enzymes related to
melatonin synthesis and genes involved in retinal phototrans- Proliferation
duction (922). Mitotic figures are rare or absent {1505,1548}, even in pleomor-
phic cases {924}. The mean Ki-67 proliferation index is < 1%
Genetic profile {137,925,1548}.
No chromosomal gains or losses were found by comparative
genomic hybridization {2668). On DNA methylation profiling, Electron microscopy
cases diagnosed as pineocytoma grouped within a distinct Ultrastructurally, pineocytomas are composed of clear cells and
subgroup, which was in close proximity to normal pineal gland various numbers of dark cells joined with zonulae adherentes
tissue {2490). {1261,1297,1503,2119) . The cells extend tapering processes
that occasionally terminate in bulbous ends. Their cytoplasm
Macroscopic appearance is relatively abundant and contains well-developed organelles.
Pineocytomas are well -circumscribed lesions with a greyish- Pineocytoma cells share numerous ultrastructural features with
tan. homogeneous or granular cut surface (325,1297,2826) . normal mammalian pinealocytes, such as paired twisted fila-
Degenerative changes, including cyst formation and foci of ments, annulate lamellae , cilia with a 9 + O microtubular pattern .
haemorrhage, may occur (2067). microtubular sheaves, fibrous bodies, vesicle-crowned rodlets .
heterogeneous cytoplasmic inclusions, and membrane whorls.
Histopathology as well as mitochondrial and centriolar clusters . Membrane-
Pineocytoma 1s a well-differentiated , moderately cellular neo- bound dense-core granules and clear vesicles are present in
plasm composed of relatively small , uniform, mature cells the cytoplasm and cellular processes. The cellular processes
resembling pinealocytes. It grows primarily in sheets , and it show occasional synapse-like junctions.
often features large pineocytomatous rosettes composed of
aoundant delicate tumour cell processes. Pineocytomatous lmmunophenotype
rosettes are nor seen in the normal pineal gland. In pineocy- Pineocytomas usually show strong immunoreactivity for syn -
toma. poorly defined lobules may be seen , but a conspicu- aptophysin , NSE, and NFP. Variable staining has also been
OJS ;ob0lar arcr111ecture 1s instead a feature of normal pineal reported for other neuronal markers , including class Ill p-tubulin ,
g'ard t ~ost nuclei are round to oval, with inconspicuous the microtubule-associated protein tau, chromogranin A, and
nuc1eo! and 11nely dispersed chromatin Cytoplasm is moder-
0
the neurotransmitter serotonin (5-HT) (615 ,1503,1505,1751 .
ate in quanrny and homogeneously eosinophil1c. Processes are 2291,3510) . In pleomorphic cases, the gangliocytic cells usually
'.:C: "isp ·:.;o_, s o7ren er·d·ng 1n club-shaped expansions that are express multiple neuronal markers. especially NFP. Expression
Box 5.01 Diagnostic criteria tor p1neocytoma
o~ CRX , _a transcri~tion factor involved in the development and
d1fferent1ation of pineal cell lineage, Is an additional indication Essential:
that these tumours are biologically linked to pinealocytes f2806 Demonstration of pineal parenchymal differentiation by h~stopattiolog1cal and im-
650] . I • munophenotypic features (e.g. positivitY for synaptophysin)

AND
Differential diagnosis Absence of criteria quaflfying for the diagnosis of pineal parenchymal tumour of
The wall. of a pineal cys t may masquerade as a pineocytoma intermediate differentiation or pineoblastoma
especi~lly when the pineal parenchyma has lost its normal lobu~ AND
~ar architecture and is distorted. The distinction may be diffi cu lt
low proIiferativeJmitotlc activity
m small specimens. However, normal pineal parenchyma does
not show pin~oc~torr:ato.us rosettes . lmmunohistochemistry AND
may ~e helpful 1n h1ghllght1ng the typical layered architecture of Pineal region location
the pineal cyst wall: an inner GFAP-positive piloid gliotic layer
and an o~ter synaptophysin/NFP-positive pineal parenchymal
Essential and desirable diagnostic criteria
layer. Unllke pineal parenchymal tumour of intermediate dif-
ferentiation, pineocytoma does not show KBTBD4 alterations See Box 5.01 .
{ 2~~0 , 1834} . The pleomorphic pattern of pineocytoma may be
m1s1nterpreted as ganglloglioma. However, pleomorphic plneo- Staging
cytoma does not show a neoplastic glial component, tumoural Not relevant
CD34 expression, or BRAFp.V600E mutation .
Prognosis and prediction
The clinical course of pineocytomas is characterized by a
Cytology long interval between onset of symptoms and surgery [325) .
Limited clinical relevance The reported 5-year survival rate of patients with pineocytoma
ranges from 86% to 91% [896 ,2838) . In one series . the 5-year
Diagnostic molecular pathology event-free survival rate was 100% (896} . Extent of surgical
Pineocytomas do not show any recurrent genetic alterations but resection is considered to be the major prognostic factor (600) .
do have a distinct DNA methylation profile.

Pine I tumo 245


Pineal parenchymal tumour Hasselblatt M
Huang A
of intermediate differentiation Jones DTW
Orr BA
Snuderl M
Vasiljevic A

Definition
Pineal parenchymal tumour of intermediate differentiation
(PPTID) is a tumour of the pineal parenchyma that is intermedi-
ate in malignan.c y between pineocytoma and pineoblastoma ,
composed of diffuse sheets or large lobules of monomorphic
round cells that appear more differentiated than those observed
in pineoblastoma (CNS WHO grade 2 or 3) .

ICD-0 coding
9362/3 Pineal parenchymal tumour of intermediate differentia-
tion

ICD-11 coding
2A00 ._ 20 & XH1S48 Tumours of the pineal gland or pineal region
& Pineal parenchymal tumour of intermediate differentiation

Related terminology
Not recommended: malignant pineocytoma; pineocytoma with
anaplasia ; pineoblastoma with lobules .

Subtype(s) Fig.5.06 Pineal parenchymal tumour of intermediate different1at1on. A tumour located


None in the pineal region shows mild and heterogeneous contrast enhancement with inva-
sion of the tectal plate and compression of the aqueduct of Sylvius, mesencephalon.
pons, and cerebellar vermis (T1 gadolinium).
Localization
PPTIDs occur in the p ineal region. Genetic profile
Smal l in-frame insertions of KBTB04 are recurrent and charac-
Clinical features teristic {1834) . PPTIDs have relatively flat copy-number profiles .
The clinical presentation is similar to that of other tumours of the with some cases showing broad gains or losses [2490,2668!
pineal parenchyma (see under Pineocytoma , p . 243). On DNA methylation profiling , PPTID as a distinct molecular
subgroup can be separated into two further subtypes: PPTID-A
Imaging and PPTID-B 12490). The prognostic significance of these
PPTIDs usually appear larger and more heterogeneous than molecular subgroups requ ires further evaluation .
pineocytomas , and they often demonstrate local invasion
{1690). Macroscopic appearance
The macroscopic appearance is similar to that of pineocytomas
Spread
PPTIDs have the potential for local recurrence and craniospinal Histopathology
dissemination {896, 1427, 3396 ,3554) . PPTID may exhibit two architectural patterns: diffuse (neurocy-
toma- or oligodendroglioma-l ike) and/or lobulated (with vessels
Epidemiology delineating vague lobules) (1505l . PPTID is a potentially aggres-
PPTIDs account for approximately 45% of all pineal paren - sive neoplasm and is characterized by moderate to high cellu-
chymal tumours (137,1427,3510,3612). Median patient age is larity. The neoplastic cells usually harbour round nuclei showing
33 years (range: 3.5-64 years) . There is a slight female prepon - mild to moderate atypia and salt-and-pepper chromatin . The
derance (M :F ratio : 0 .8 :1) {999,1427,1505,3510). cytop lasm of cells in PPTID is more easily distinguishable than
in pineoblastoma.
Etiology
No syndromic associations or genetic susceptibilities have Grading
been reported . The biological behaviour of PPTIDs 1s variable Although the
majority correspond to CNS WHO grade 2, more aggressive
Pathogenesis cases may correspond to grade 3, but definite histological
Cell of origin grading criteria remain to be defined .
The histogenesis of PPTID has been linked to the pinealocyte.

246 l-'1n<.:~a1 turnuur~,


.•

'
c • •,
-
Ag.5.07 Pineal parenchymal tumour of intermediate differentiation. A Tumour cells are round and relatively monomorphic. with a conspicuous cytoplasm. Nuclei are round to
oval with a finely dispersed chromatin and a small nucleolus. e A few neoplastic cells show cytoplasmic immunoexpresslon of NFP. C In this example. the Ki-67 proliferation
index is about 15%. D Diffuse nuclear lmmunoexpression of CRX, consistent with pineal differentiation.

Proliferation Oifferential diagnosis


Mitouc activity is low to moderate (1505) . The Kl-67 proliferation Histological ly, PPTID may resemble central neurocytoma. Both
index ranges from 3.5% to 16.1% (137,1427,2668 ,3554 ,3612) . have a sh eet-like architecture, monomorphic round cells , and
diff use synaptophysin immunopositivity. However. PPTID does
lmmunophenotype not express NeuN . Checking the imaging is critical . because
PPTIDs usually stain positively for synaptophysin (137,999,1427, a tumour in the pineal region resembling neurocytoma is most
1505). Labelling for NFP and chromogranin A is variable (1503 , like ly a primary pineal parenchymal tumour
1505,3235 ,3510) . PPTIDs typ ically show diffuse(> 50% of cells) The histology of PPTID may also be confused with that of oli-
nuclear expression of CRX , a transcription factor involved in the godendrogl ioma. PPTID does not express OLIG2 or show the
differentiation of retinal and pineal lineages (650). typ ical molecular alterations of oligodendroglioma , such as IDH
mutati on s and 1p/19q codeletlon .

Flg.5.08 KBTB04 -mu tant pineal parenchymal tumour of intermediate differentiation. A Diffuse sheets of monomorphrc round cells . B Strong and diffuse synaptophysrn rm-
rnunoposltrvit y.
~ox5.02 Diagnostic criteria for pineal parenchymal tumour of Intermediate differen- Essential and desirable diagnostic criteria
tiation
See Box 5.02 .
Euentlal:
Demonstration of pineal parenchymaJ differentiation by hlstopathologlcal and lm- Staging
munophenotyplc teatures (e.g. positivity for synaptophysln) No staging system has been defined .
AND
lncreast!d prollferative/mltotlo activity Prognosis and prediction . .
In 29 studies including 127 patients with PPTID, m~d1an ov A3 I
0 1
AND
survival was 14 years , and the 5-year overall survival ra e wa'")
Absence of criteria qualifying for the diagnosis of pineoblastoma
84% 11996}. Median progression-free survival was 5 2 years
AND
and the 5-year progression-free survival rat~ was 52~/0 (19961 .
Pineal region location recurrences often involved spinal/leptomeningeal dissemina-
AND (for unresolved cases) tion {1996}. . .
DNA methylation profiling In one study, low-grade PPTIDs were defined as tumours w1tn
a low mitotic count and expression of NFP in numerous cells
Desirable: (1505}. In patients with low-grade PPTID , the_5-ye~r overall. sur-
Molecular demonstration of KBTBD4 in-frame insertions vival rate was 74% . Recurrences occurred 1n 26 Yo of patients
and were mainly local and delayed (896}. High-grade PPTIDs
were defined as tumours with a low mitotic count but no or only
~ixed pineocytom~- pineoblastoma, composed of clearly rare expression of NFP, or with a high mitotic count and NFP
delineated areas of pineoblastoma admixed with well-demar- expression in numerous cells (1505). In patients with high-grade
cated areas of pineocytoma {2067,2839}, should not be diag- PPTID, the 5-year overall survival rate was 39% , and the risks of
nosed as PPTIO (see Pineoblastoma , p. 249) . recurrence (56%) and spinal dissem ination (28%) were higher
than in patients with low-grade PPTID (896} . Low- and high-
Cytology grade prognostic groups also showed different mean Ki-67
Insufficient data available proliferation index values (5 .2% vs 11 .2%) 1925}.
In another study, patients with low-grade PPTIDs (with a Ki-67
Diagnostic molecular pathology proliferation index of < 5%) had better overall survival and pro-
Although a morphological diagnosis is still acceptable in the gression-free survival than did those with high-grade PPTIDs
absence of molecular data, evidence of a KBTBD4 alteration (with a Ki -67 proliferation index of~ 5%) {3554}.
is considered highly desirable for the diagnosis of PPTID. The The prognostic role of mitotic count, NFP immunopositi vity.
finding of this alteration in what appears to be another pineal and Ki-67 prol iferation index requires confirmation by further
parenchymal tumour type histologically, or conversely the studies, and the prognostic role of molecular subgrouping
absence of this alteration in a tumour otherwise resembling remains to be determined; consequently, there are currently no
PPTIO, should prompt careful consideration as to whether an recommended grading criteria for PPTID.
alternative diagnosis may be more suitable.

248 Pin al iur 11rJLW..i


Pineoblastoma Hasselblatt M
Huang A
Jones DTW
Orr BA
Snuderl M
Vasiljevic A

Definition
Pineoblastoma is a poorly differentiated cellu lar embryonal neo-
plasm arising in the pineal parenchyma (CNS WHO grade 4).

ICD-0 coding
9362/3 Pineoblastoma

ICD-11 coding
2A00.20 & XH 1ZH1 Tumours of the pineal gland or pineal region
& Pineoblastoma

Related terminology
None

Subtype(s) Fig. 5.09 Pineoblastoma. A This patient has obstruc tive triventricular hy?rocephal_us
Pineoblastoma , mi RNA processing-altered_1; pineoblastoma, (T2-weighted MR I). B A tumour located in the pineal region appears mildly hypo1n-
miRNA processing-altered_2; pineoblastoma , RB1-altered (pin- tense on T2-weighted MRI.
eal retJnoblastoma) ; pineoblastoma, MYC/FOXR2-activated
enhancement. On T2-we ighted images they are typically isoin -
Localization tense to mildly hyperintense (571 ,2203,2 645 ,3195).
Pineoblastomas are found in the pineal reg ion .
Spread
Clinical features Craniospinal dissemination is observed in 25 - 33% of patients
The clin ical presentation is similar to that of other tumours of the (896,1838 ,3139 ,3325) .
pineal parenchyma (see under Pineocytoma , p. 243).
Epidemiology
Imaging Pin eoblastomas account for approximately 35% of all pineal
Pineoblastomas are often detected as large tumours, frequently paren chymal tumours {137,1505,2839 ,3325 ,3510} . They can
showing invasion of surrounding structures and resulting in arise at any age but mostly occur in children . The median
obstructive hydrocephalus {571 ,2203 ,2958,2645,3195). On CT, patient age reported in a recent consensus paper {1913Al
pineoblastomas are usually slightly hyperdense with postcon- was 6 years (range: 0-41 .5 years) , in contrast to 33 years for
trast enhancement {571 ,2203). On T1 -weighted MRI , tumours pineal parenchymal tumour of intermediate differentiation . The
are often hypointense to isointense, with heterogeneous contrast median patient ag es for the molecu larly defined subtypes of

Table5.01 Clinical and molecular features of pineoblastoma subtypes


Plneoblastoma subtype

R81-altered
mlRNA processing-altered_1 mlRNA processlng-altered_.2 MYC/FOXR2-activated
(plneaJ retlnoblastoma)
Median patient age 8.5 years 11.6 years 2.1 years 1.3 years
Cancer predisposition DICER1 syndrome DICER1 syndrome Hereditary retinoblastoma None
Copy-number alterations; Copy-number alterations;
mutually exclusive mutations mutually exclusive mutations MYC amplification and ac!Jva-
Molecular features RB1 alterations tion ; chromosome 16q losses;
targeting DICER1, DROSHA, targeting DJCER1 , DROSHA,
or DGCRB or DGCRB FOXR2 overexpression

Median OS 10.4 years Not reached 2.8 years 1.2 years


5-year OS rate 68% 100% 29% 23%
5-year PFS rate 54% 93% 27% 13%
PFS, progression-free s-; ival ; OS, overall su~ival.

Pineal tumou rs 249


Genetic profile
C;t0genet1c, S'Jrj8S r31e S'"'"Jll,-,. r r'/8 ::;:·8': '",''_ ,_. ,.,..,,_
rnosome 1 3 r.d 0ss8S r r) 1 ,. j ~ "''"/,..,.._,'/I' 8'"., 1 • ,.~ ::;: - ',
1385 2115 .2668 27fj' I (S,t:;':; •re 0 ::igr~:: C rr~ 9'.: J ~r ,--:,,:;;•,.,- '°:~,
subsect10 be G:t)

Macroscopic appearance
P1neoblas ornas ars sr}~ ''au e ~,..'J ~ - / .....,,... '"J'S/ ~2~ ;..:::;,:
Haemorrhage and/o r.8cr0s s n a J cs cr838,.. •

Histopathology
Pineoblastomas esernole (/ S ' e~c,r J'Y'8. • I'"'j_.'; -:.' -.:;
CNS and are composea o~ Q" /cs Jar :::.3:·e--- 8:: -:..,.. ss·: -:.
densely packed sma ll ce ll s T e ce s 'ea:_1re ':G""'S , -- :::· S~ --
0

lar nuclear shapes. a high JC ra o n:1oers"r0...,...a· s - Js s ·. ·-


an occasional small nucleolus. scan c;~o0 asr. . a,..c .... :; -:- -·::
cel l borders. The diffuse gro..vth oa err 5 ~err .;~·is-: :- ,._, 1
occasional rosettes. Pineoc1toma ous ose:~ss ~-s -3:::::::- ·
but Homer Wright and F l e xn e r-Winters~e er r-:>ss·-<::: ~ :::. ,1 :s
seen . Flexner-Winterstein er rosettes 11101ca e re· .... cc ~s· i:. : ·-
ferentiation , as do highly dist1nc ·ve b 1n'rea:..er: 'J sscJ"--;_:
fleurettes Necrosis 1s common and m1 o ·c ac- / :; s ;;s-s - ~ •
high !1297,1505,211 9,2839 1. The mean Ki-67 pro :e . .a· s- ... ::::1
in pineoblastoma ranges from 23 5% o 50. ~o r· 3 32:. ::::~
2668) .

Mixed pineocytoma-pineoblastoma
Mixed pineocytoma-pineoblastoma is a some11 ar ccr:·:::.s·-
sial neoplasm showing a b1phasic paitern 1i n d 1 s~. rc: a :e . . -:=:-
ing areas resembling pineoblastoma and pineoc ;tora :... ·e:::.s
resembling pineocytoma must be distinguished 7' 0'T :: .2:" _ ....
normal parenchyma !1297,2067,229 1,2839 1.

Pineal an/age tumours


Pineal anlage tumours are extremely rare neoplasrs ~ ·.-e
pineal region . They are often considered a sub.ype o: c .... ~c­
blastoma because of their pineal region locahzaiioo: a ;:; -2:::-
blastoma-like tumour component, and an aggre ss1 e c r cc.
Flg. 5.10 Pineoblastoma. A This is a highly cellular tumour composed of small cells course. Historically, pineal an lage tumours ere narlec c.;~e­
with a high N:C ratio; atypical, angular, hyperchromatic nuclei; and frequent mitoses the histological features they share with melanot1c neurcec:c-
and apoptotic bodies . B These tumours frequently show areas of necrosis. C There dermal tumour of infancy (or reti nal anlage tumour a oer gr
1s diffuse expression of synaptophysin with paranuclear dot-like reactivity. tumour typically located in the maxilla) Pineal anlage tV"'Cu'S
are characterized by a combination of neuroectoaerrrai a"'c
pineoblastoma are shown in Table 5.01 (p. 249). There is a slight heterologous ectomesenchymal components. The neuroeo :~e­
female predominance, with an M:F ratio of 0.7:1. lial component is characterized by pineoblasroma-l1ke sree:s
or nests of small round blue cell s, neuronal gangllonic1g a :: 1L
Etiology ferentiation , and/or melanin-containing epithello1d ce1ls Tne
Genetic susceptibility ectomesenchymal compo nent contains rhabdomyob iasts st· -
P1neoblastomas can occur 1n patients with familial (bilateral) ated muscle, and/or cartilag inous islands !42 253 28491 T c
ret1nobla stoma - a condition called trilateral retinoblastoma cases diagnosed as pineal anlage tumour were class1f ea b1,
(703) - and th ere is a strong association with OICER1 germline DNA methylation profiling in the "p1neoblastoma MYC. FO R2-
rnu 1a11ons [706 .2769) . Cases in patients with familial adenoma- activated " group [186 5. 191 3AI and another shoved mo ecu·a'
to us polypos 1s have also been reported !1020 ,1404). features of an infantile cerebellar tumour resembling erior 1cra
tumour with mul t1layered rosettes . wnh DICER 1 mutation 13251:?
lmmunophenotype
Pineoblastomas stain positively for synaptophysin and NSE.
Staining for NFP and chromogranin A is less frequent than in
pinsocytomas {1505 ,2067,3510). SMARCB1 staining is consist-
ently retained {2116).

Differential diagnosis
The histopathology of pineoblastomas is not distinctive, as
they are composed of sheets of poorly differentiated embryo-
nal neoplastic cells. Confirming the location of the tumour in
the pineal region is thus a first critical step in ruling out other
non-pineal embryonal tumours, especially medulloblastomas .
Nuclear expression of SMARCB1 (INl1) and SMARCA4 (BRG1)
is retained in pineoblastomas, allowing their distinction from
atypical teratoid/rhabdoid tumour. Pineoblastomas are typi -
cally negative for LIN28A , a marker of embryonal tumour with
multilayered rosettes. Pineoblastomas typically show diffuse
(> 50% of cells) nuclear expression of CRX, a transcription fac-
tor involved in the differentiation of retinal and pineal lineages
{650) . Unlike pineal parenchymal tumours of intermediate dif-
ferentiation, pineoblastomas do not show KBTB04 alterations
{2490,1834).

Cytology
Cerebrospinal fluid cytology is used in staging . Cerebrospinal
fluid dissemination is characterized by clusters ot poorly dif-
ferentiated cells with round , hyperchromatic nuclei and scant
cytoplasm {3325).

D iagnostic molecular pathology


Copy-number alterations and/or mutually exclusive mutations
targeting DICER1, OROSHA , or OGCRB have been described
{706 ,2976 ,1865,2490,1913). DNA methylation profiling segre-
gates pineoblastoma into four molecular subgroups showing
d istinct genetic and clinical features 11865,2490,1913}:
Plneoblastoma, miRNA processing-altered_1 arises in chil -
dren and is characterized by copy-number alterations and/or
mutually exclusive mutations targeting O/CER1, DROSHA, or
Flg.5.11 Pineal anlage tumour. A Tubular structures composed of epithelioid cells
DGCRB, which cause aberrant microRNA processing 11865, containing melanin pigment. B Striated muscle cells.
2490,1913). . .
Pineoblastoma miRNA processing-altered_2 arises mainly
in somewhat olde~ children and is also characterized by copy-
Box5.03 Diagnostic criteria for pineoblastoma

number alterations and/or mutually exclusive mutations target- Essential:


ing OICER1, OROSHA , or OGCRB, which cause aberrant micro- Histopathological features of an embryonal tumour
RNA processing {1865 ,2490 ,1913). . . AND
Pineoblastoma, RB1-altered arises in Infants, show~ s1m1- High proliferative/mitotic activity
larities with retinoblastoma, and includes cases of trilateral
AND
retinoblastoma as well as sporadic pineal tumours with RB1
Pineal region location
alterations 11865,2490} . . . .
Pineoblastoma, MYC/FOXR2-activated arises in infa~ts and
Desirable:
exhibits recurrent focal gains or amplifications affecting the
MYC region as well as chromosome 16q losses 11865,24~~) . Retained nuclear SMARCB1 (INl1) staining
MYC activation and overexpression of FOXR2 are characteristic DNA methylation profile of pineoblastoma subtype
features (2490). .
Recently, WNT-activated embryonal tumours of the pineal
region have been report ed . Whether these tumours represent a Essential and desirable diagnostic criteria
novel pineoblastoma subgroup or ectopic WNT medulloblasto- See Box 5.03 .
mas remains uncertain !1914] .

?1neal rumours 251


Staging seeding and (rarely) extracranial metas t~ sis (896 .1297 111.-1")
Clinical staging procedures include MRI examinations of the 2839 ). Overa ll survival is short; older studies reported m8d18r
CNS with contrast agent. This is complemented by cerebrospl - · fro m 1.3 to 2 ·5 years 1521 ,896.2067) . b. ut rer,t:Jrt
va Iues ranging
nal flu id cytology at the time of diagnosis. The postoperative studies have reported improved medi_ an. overall survival t1rne s
staging system developed by Chang and others in 1969 1519) reach ing 4.1-8.7 years {893 ,1451). Similarly, .reported 5-year
(included in the Toronto staging system , endorsed by the Union overall surviva l rates vary from 10% to 81 % . D1ssem1 nated dis-
for International Cancer Control [UICC)), which defines the fol - ease at the time of diagnosis (as determined by cerebrosp1na1
lowing degrees of metastatic spread , is still being used: fluid examination an d MRI of the spine) {521,893,31391. young
patient age [801 ,1311 ,3139). and parti~I surgi_cal resection
MO No evidence of gross subarachnoid or haematogenous 11451 ,1838,3139) are negative prognostic . predictors. Radio-
metastasis therapy seems to positively affect prognosis !893 ,1838,2839)
M1 Microscopic tumour cells found in the cerebrospinal fluid The 5-year survival of patients with trilateral ret1noblastoma syn-
M2 Gross nodular seeding demonstrated in the cerebellar/ drome has increased over the past few decades , probably due
cerebral subarachnoid space or in the third or lateral to better chemotherapy re gimens and earlier detection of pineal
ventricles
disease (703). . .
M3 Gross nodular seeding in the spinal subarachnoid space The prognostic value of molecu lar subgrouping 1s high , as
M4 Metastasis outside the cerebrospinal axis shown in Table 5.01 (p . 249) [186 5,2490 ,1913}.
The recently described WNT-activated embryonal tumours of
Prognosis and prediction the pineal region have a good prognosis . At median follow-up
Pineoblastoma is the most aggressive of the pineal parenchy- of 3 years (range: 0.8- 12.8 years), all patients were alive without
mal tumours , as evidenced by the occurrence of craniospinal disease (n = 5) or with stable res idual disease (n = 2) (1914)

252 Pineal tumou rs


Papillary tumour of the pineal region Hasselblatt M
Huang A
Jones DTW
Orr BA
Snuderl M
Vasiljevic A

Definition
Spread
Papillary tumour ~f the pineal region (PTPR) is a neuroepithelial PTPRs are characterized by frequent local recurrence . Spinal
tumour ~hara~ten~ed. by a combination of papillary and solid dissemination may occur (895 ,1627).
areas, with ep1theilal-l1ke cells and immunoreactivity for cytoker-
atins (CNS WHO grade 2 or 3).
Epidemiology
There are no incidence data for these rare tumours . PTPRs
ICD-0 coding occur in children and adults . Reported patient ages range from
9395/3 Papillary tumour of the pineal reg ion 1 to 71 years, with a median of 35 years . There is no sex predi-
lection (921 ,923 ,1146,1249,1281 ).
ICD-11 coding
2A00 . 2~ & XH3904 Tumours of the pineal gland or pineal region Etiology
& Papillary tumour of the pineal region Unknown

Related terminology Pathogenesis


None Cell of origin
lmmunohistochemical findings and ultrastructural demonstra-
Subtype(s) tion of ependymal , secretory, and neuroendocrine organelles
None suggest that PTPR may originate from remnants of the special-
ized ependymal cells of the subcommissural organ (1502) . Fur-
Localization ther evidence for a putative origin from specialized ependymo-
PTPRs are located in the pineal region . cytes of the subcommissural org an comes from high levels of
expression of genes expressed in the subcommissural organ ,
Clinical features including SPOEF, ZFHX4, RFX3, TTR, and CALCA {922,1283).
The clinical features of PTPRs are similar to those of other pineal
parenchymal tumours (see under Pineocytoma , p. 243). Genetic profile
Recurrent chromosomal imbalances include losses from chro-
Imaging mosome 10 as well as gains on chromosomes 4 and 9 {1196,
PTPRs appear as well-circumscribed heterogeneous masses 1249,1283). Genetic alterations of PTEN have been reported
composed of cystic and solid portions. Aqueductal obstruction (1146).
with hydrocephalus is a frequent associated finding (97,2547, On DNA methylation profiling, PTPR as a d1st1nct molecular
2664,2816). Intrinsic T1 hyperintensity {499,517,2816) may be subgroup can be separated into two further subtypes: PTPR-A
rel ated to secretory material (517}. but this association remains and PTPR-B {1283,2490). The prognostic significance of these
controversial (97,1627). Postcontrast enhancement is heteroge- molecular subgroups needs further evaluation.
neous.

.
Flg.5.12 Papillary tumour ol !he p11 1eo.1 reg1d11 A 'J. 1g1ttal. T1 weighted MRI showing a heterogeneous !uniou1 in the p111eal reg1011 B Sag1ttJI con tras t enhanced, T1·we1ghted
MRI showing a heterogen eously enli a ic nq tu ,CJ •ll ,1 1!ht p1r1ea1region . C Sag1lta l. T2 wo1yhtcd rv!HI st1ow1110 ::.l) llH:l -:ysr1c are,1s ind tu1110ur or rt1e p1118al r891on

Pine I tumours 253


'ti!'
Fig. 5.13 Papillary tumour of the pineal region . A Papillary structures, which may alternate with diffuse solid areas. B Neoplastic cells, detached from the papillary vasculanzea
core, create an apparent clear perivascular space. Note the extensive necrosis in this case. C Tumour architecture is characterized by a variable number of papillary struc-
tures. D Papillae are covered by large, columnar, epithelial-like cells.

Macroscopic appearance cases may correspond to grade 3 , but definite histological


PTPRs appear as well-circumscri bed tumours, grossly indistin- grading criteria remain to be defined .
guishable from pineocytomas .
Proliferation
Histopathology Mitotic activity is moderate in most cases (1146,1502,920,1282\.
PTPR is an epithelial-looking tumour with papillary features and In one series , the Ki-67 proliferation index ranged from 1 0%
more densely cellular areas , often exhibiting ependymal-like dif- to 29 .7% (median : 7.5%), and increased proliferative activity
ferentiation (true rosettes and tubes) . PTPR may exhibit a promi- (defined as a Ki-67 proliferation index of ~ 10%) was observed
nent papillary architecture or, conversely, a more solid mor- in 39% of cases (920) ; in another series, Ki-67 ~ 10% was
phology in which papillae are barely recognizable (920 ,1281 ). observed in 40% of cases (1281}. High proliferative activity has
In papillary areas, the vessels are covered by layers of large, been linked to younger patient age (923) .
pale to eosinophilic, columnar cells . In cel lular areas, cells with
a somewhat clear or vacuolated cytoplasm (and occasionally lmmunophenotype
with an eosinophilic PAS-positive cytoplasmic mass) may also Th e most distinctive immunohistochemical feature of PTPRs 1s
be seen. The nuclei are round to oval, with stippled chromatin; their reactivity for cytokeratins (especially CK18), particularly in
pleornorphic nuclei may be present . Necrotic foci may be seen . papillary structures. SPDEF is frequently expressed and has
Vessels are hyal1nized and ofte n have a pseudoangiomatous been proposed as a diagnostic marker {1283). PTPRs also stain
morphology, with multiple lumina j920) . There is clear demarca- for vimentin, S100, NSE , MAP2, CD56, and transthyretin t1249.
tion tJetween the tumour and the adjace nt pineal gland . 2909). GFAP expression is less common than in ependymomas,
and focal membrane or dot-like EMA staining as encountered
Grading in epe ndymomas is rare (1249 ,1502,"1750). Neurofilament immu-
Tr1e b1rJkJg1cal behaviour ot PTPRs is variable . Although the nolabe lling is never seen, whereas the neuroendocrine markers
ma1or1ty cr.Jfre~)por1d to C f'-IS WHO grade 2, more aggressive synaptophysin and chromog ranin A are sometimes weakly and
focally expressed I1502} Most PTPRs are negative for Kir71,
E-cadherin (cadherin 1), and claud in-2 i e k
· h · · · mar ers frequently
present 1n c orold plexus tumours 1920 ,923 .12491.

Differential diagnosis
Because of its eplthelioid cytology and pap·illa h'
· . . ry arc 1tecture
PTPR may m1m1c pineal metastasis of adenoca · .'
· · · · h. . rc1noma . In this
s1tuat1on, c 11nica 1 1story and imaging workup f th .
o e patient
shou Id b e care f ully assessed and immunohistoch · 1
· f d d' em1ca stud-
ies per orme accor ingly . (e·g · thyroid transcr·1pt'ion factor 1
[TT'.1] , G ~TA3) . Choroid. plexus papilloma differs from PTPR
by its straightforward epithel . ial morphology and c onsp1cuous ·
basement men:brane . Unlike PTPR , choroid plexus tumours
show freq u~nt 1mmunoex.pression of E-cadherin and are typi-
cally C D56 -~~munon~gat1~e or only weakly positive 1927]. Mem-
branous staining for K1r7.1 1s frequent in choroid plexus tum
but rare in. PTP~ .11249]. In contrast to PTPRs, ependymi~~~
usually stain pos1t1vely for GFAP and lack CK18 expression Th
diagnosis of a pineal parenchymal tumour may be consid.ere~
i ~ small specimens ?r solid for~s of PTPR. The strong expres-
sion of synaptophys1n and CRX 1n pineal parenchymal tumours
allows their distinction from PTPR {650].

Cytology
Limited cl inical relevance
Flg.5.14 Papillary tumour of the pineal region. lmmunoexpress1on for CK18 typically
predominates in perivascular areas.

BoxS.04 Diagnostic criteri a for papillary tumour of the pineal region

Essential:
Papillary growth pattern with epithelial-like cells
I
AND
Diagnostic molecular pathology
DNA methylation profiling clearl y distinguishes PTPR from Characteristic lmmunohistochemical staining pattern (e.g. positivity for
cytokeratins, SPDEF, CD56)
ependymomas and pineal paren chymal tumours {1283).
AND

Essential and desirable diagnostic criteria Pineal region location


See Box 5.04. AND (for unresolved cases)
Confirmatory DNA methylatlon profiling
Staging
Not rel evant
factors on overall or progression-free survival [1281 ). In that
Prognosis and prediction series, increased mitotic activity was significantly associated
The clinical course of PTPR is o~en com plicated by local recur- with shorter progression-free survival. Increased proliferative
rences . In a retrospective multicentre study of 31 patients, activity was also associated with shorter progression-free sur-
tumour progression occurred in 72% of cases, and the esti- vival: patients whose tumours had a Ki-67 proliferation index of
mated 5-year overall and progression-free survival rates were ~ 10% had a median progression-free survival time of 29 months
73% and 27% . respectively 1923). Leptomen ingeal seeding (range: 0-64 months) , versus 67 months (range: 44-90 months)
through the cerebrospinal fluid has been rarely reported 1895). for those whose tumours had a Ki-67 proliferation index of
Incomplete resection tended to be associated with decreased < 10%. The tumours of the 3 patients who succumbed to dis-
survival and with recurrence 1923). In an updated retrospec- ease all showed increased mitotic and proliferative activity
tive series of 44 patients, only gross total resection and younger I1281}. The usefulness of mitotic count or proliferation index in
patient age were associated with overall su rvival ; radiothera py def in Ing a more aggressive subset of PTPRs requires conf 1rma-
and chemotherapy had no si gnificant impact (895). An~t.her tion in further studies . Recurrences may show higher prolifera-
study, of 19 patients , also found no significant effect of clinical tive activity (1196 ,1825).
Hasselblatt M
Desmoplastic myxoid tumour Huang A
Jones DTW
of the pineal region , SMARCB 1-mutant Orr BA
Snuderl M
Vasiljevic A

Definition Pathogenesis
Desmoplastic myxoid tumour (DMT) of the pineal region , Cell of origin
SMARCB1-mutant, is a tumour showing desmoplasia and myx- Unknown
oid changes but lacking histopathological signs of malignancy,
with alterations of the SMARCB1 region on ch romosome 22q11 . Genetic profile .
Apart from alterations affecting the SMARCB1 region on 22q1
ICD-0 coding chromosomal alterations are rare and non-recurrent 13185J.
None On DNA methylation profiling, DMT, SMARCB1-mutant. forms
a distinct group located in close proximity to one of the molecu-
ICD-11 coding lar subgroups of atypical teratoid/rhabdoid tumour (AT/RT-MYC)
2A00.20 Tumours of the pineal gland or pineal reg ion and to poorly differentiated chordoma /3185}.

Related terminology Macroscopic appearance


None Insufficient data available

Subtype{s) Histopathology
None DMT, SMARCB1-mutant, is characterized by an admixture of
variably dense cords of small to med ium-sized oval to spin-
Localization dled and epithelioid cells embedded in a heavily collagenized
All 7 cases reported to date were localized in the pineal region . matrix. Tumour cells are dispersed to a variable extent within a
loose pale basophilic myxoid matrix. Fascicular and whorling
Clinical features growth patterns may be encountered , and irregu larly shaped
The clinical features are similar to those of pineal parenchymal and elongated blood vessels with marked fibrosis are frequent.
tumours (see under Pineocytoma, p. 243). Scattered rhabdoid tumour cells are rare. Mitotic activity 1s
exceptional (< 1 mitosis/mm 2 ) (3185J
Epidemiology
The median age of 4 female and 3 male patients was 40 years Grading
(range: 15-61 years) {3185}. The biolog ical behaviour of DMT, SMARCB1-mutant, seems
to be less aggressive than that of atypical teratoid/rhabdoid
Etiology tumour /3185 }, but grad ing remains to be defined .
Unknown

256 P1r1e<.tl iur 110 . w ;


Proliferation Box 5.05 Diagnostic criteria for desmoplastic myxo1d tumour of the pineal region.
The Ki-67 (MIB1~ proliferation index is low (median: 3%), and SMARCB1-mutant
onl~ one tumour in a young patient showed higher proliferative
act1v1ty (15%) (3185}. Essential:
Desmoplasia and myxoid changes
lmmunophenotype AND
Tumour ~ells show loss of nuclear SMARCB1 (IN11 ) expression . Lack of histopathological signs of malignancy
E press1on of CD34 and EMA is often present 13185). AND
Loss of tumoural SMARCB1 expression
Cytology AND (for unresolved cases)
Insuffic ient data available Confirmatory DNA methylatlon profiling

Diagnostic molecular pathology


Tumours should be assessed for SMARCB1 deficiency (3185).

Essential and desirable diagnostic criteria


See Box 5.05 .

Staging
Not relevant

Prognosis and prediction


Gross total resection was achieved in 4 of 7 patients , and 3 of
7 patients did not show evidence of residual disease on postop-
erative MRI {3185). There was no evidence of metastatic disease
in any patient. After a median observation time of 48 months , ·'
3 patients were alive with stable disease, 1 patient had expe-
rienced tumour progression , and 3 patients had died from the

Fig.5.16 Desmoplastic myxoid tumour of the pineal region, SMARCB1-mutant. Note
disease {31 85). Prognostic markers have not yet been reported . loss of nuclear SMARCB1 staining in the tumour cells but not the non-neoplastic cells.

Pineal tumours 257


Perry A
Cranial and paraspinal nerve tumours:
Introduction

Nerve sheath tumours are common throughout the craniospinal of Tumours , it has similarly been appreciated that melanotic
axis and may be encountered either sporadically or as part of schwannoma is a highly distinct and frequently aggressive
a wide variety of tumour predisposition syndromes . including tumour type with unique genetic underpinnings that distinguish
neurofibromatosis type 1 and type 2, schwannomatosis, and it from all other nerve sheath tumours , including schwannomas
Carney complex. Whereas most are thought to arise from clas- Therefore, and in keeping with the changes in the soft tissue
sic peripheral nervous system elements (such as Schwann cells classification scheme, its name has been changed to "malignant
and, less commonly, perineurial cells), the paragangliomas melanotic nerve sheath tumour". The majority of both sporadic
involve specialized neuroendocrine cells of the sympathetic and Carney complex-associated malignant melanotic nerve
and parasympathetic nervous system. Therefore, cauda equina sheath tumours have an inactivated PRKAR1A gene, so immu-
neuroendocrine tumours (previously termed "CNS paragangli- nohistochemical loss of its protein product serves as a useful
omas") are now discussed within this chapter, rather than with diagnostic surrogate . The current chapter also updates the
neuronal and mixed neuronal-glial tumours as done previously. genetic advances in malignant peripheral nerve sheath tumour.
We now know that a wide variety of pathogenic germline altera- including useful biomarkers recently translated into molecular
tions predispose to familial paragangliomas , although those diagnostic tools. A growing body of evidence now suggests
arising in the filum terminale I cauda equina region appear to that epithelioid malignant peripheral nerve sheath tumour is also
be the exception to that rule, with recent data suggesting that genetically unique. Therefore , it could potentially be separated
they are biologically distinct from paragangliomas elsewhere out in future iterations, but for now it continues to be treated as a
in the body. Since the last edition of the WHO Classification subtype of malignant peripheral nerve sheath tumour.

260 Cran1:=il and parasp1nal nerve t.Jmours


Schwannoma Stemmer-Racham1mov AO
Jo VY
Reuss DE
Rodriguez FJ

Definition
Schwannoma is a ben ign nerve sheath tumour compo d
. I I . se
entire y or near Y entirely of differentiated neoplastic Schwann
cells (CNS WHO grade 1).

ICD-0 coding
9560/0 Schwannoma

ICD-11 coding
2A02.3 & XH98Z3 Benign neoplasm of cranial nerves &
Schwannoma

Related terminology
Not recommended: neurilemmoma.
Fig. 6.01 Schwannoma. A T1-weighted MRI showing the characteristic dumbbell
shape of a spinal root schwannoma. B Cut surface showing a globo1d encapsulated
Subtype(s) tan-grey tumour with areas of xanthic change (yellow) and attached nerve.
Ancient schwannoma; cellular schwannoma; plexiform schwan-
noma; epithelioid schwannoma; microcystic/reticular schwan-
noma Clinical features
Schwannomas are slow-growing tumours , often appearing as
Localization asymptomatic masses or incidental findings on imaging stud-
Common sites of origin are peripheral nerves in the skin and ies. Spinal schwannomas may elicit sensory or, less frequently,
subcutaneous tissues of the head and neck, or along the f lexor motor symptoms . Vestibular schwannomas often come to clini-
surfaces of the extremities. Spinal intradural extramedullary cal attention with hearing loss and vertigo . Painful schwanno-
examples are also common and form dumbbell tumours when mas may be associated with schwannomatosis .
growing through neural foramina. Multiple paraspinal schwan-
nomas are common in neurofibromatosis type 2 (NF2). Another Epidemiology
frequent location is the vestibular division of the eighth cranial More than 90% of schwannomas are solitary and sporadic .
nerve, and bilateral involvement is a definitional criterion for NF2 Schwannomas affect people of all ages , but the peak incidence
1862/. Spinal intramedullary and CNS sites are rare 1480). as is in the fourth to sixth decades of life. There is no known sex or
are cases involving viscera (such as the gastrointestinal tract) race predisposition .
or bone {3343/.

Nuclear palisades, known as Verocay bodies , are a typical


Etiology nodular growth pattern . Patients may also develop multiple
The etiology of most sporadic schwannomas is unknown , meningiomas (which are associated with increased morb1d1ty
although there is an established increased incidence associ- and mortality) and spinal epe ndymomas {8621 . NF2 is inherited
ated with prior irrad iation {2716 ,2565) . The re is an association in an autosomal dominant manner, with 50% of cases repre-
with NF2 or schwannomatosis in some cases (see Pathogen- senting new or sporad ic mutations .
esis, below, as well as Neurofibromatosis type 1, p . 426; Neu- Schwannomatosis is characterized by the presence of multi-
rofibromatosis type 2 , p . 429; and Schwannomatosis, p . 434). ple schwannomas, mo stly (but not invariably) in the absence of
vestibular nerve involvement. Cranial and cutaneous nerves are
Pathogenesis infrequently affected . Tumours are often associated with pain
A causal relationsh ip exists between schwannoma tumorigene- Germline mutation s of either the SMARCB1 or the LZTR1 tumour
sis and loss of expression of merlin (also called NF2 or schwan- suppressor gene are fo und in 69- 86 % of patients wi th familia l
nomin), the growth inhibitory protein product of the NF2 tumour schwannomatosis [251 8,2966 ,2366) and 40% of patients with
suppressor gene located at 22q12.2 [3031 ). NF2-inactivating sporadic schwannomatosis (1583) . The tumorigenesis appears
mutations have been detected in approximately 50- 75% of spo- to be complex , given that these tumours arise from a four-hit
radic cases 11 437,1807,2305 ,1268). Underlying genetic events mechanism that invol ves two gen es: a germl ine SMA RCB1 or
are predominantly frameshift and nonsense mutations, w ith loss LZTR1 mutation is fo llowed by a somatic NF2 mutation on the
of the remaining wil dtype allele on c hromosome 22. Other com- same ch rom osome 22, along with a deletion of the entire other
mon mutations involve the LATS 1, LATS2, AR/01A , AR/018, and chromosome 22, leading to biall elic inacti vation of both tumour
DDR1 genes, whereas a recurrent in-frame SH3 PXD2A: :HTRA 1 suppressor genes simultaneou sly. Not surprisingly, therefore,
fusion is foun d in roughly 10% of cases {2305,31). the somatic NF2 mutation often differs among schwannomas
Multiple schwannomas are a feature of NF2 and schwanno - from any one patient, as well as between the schwannomas of
matosis, both of which can also occur in mosaic or segmental different fami ly members .
forms {292 ,1583).
NF2-associated schwannomas common ly occur before the Macroscopic appearance
age of 30 years , whereas tumours in schwannomatosis usually Schwannomas are mai nly solitary and globoid , wi th a smooth
manifest later. Bilateral vestibular schwannoma is a hallmark of surface. Most measure < 100 mm 1n greatest dimension . bu r
NF2 , often showing multifocal nerve involvement and a distinc t giant schwannomas (which are mostly encountered 1n me

262 Cranial a.nc..l p;.:ira sp1r1al r1erv lumuu rs


lumbosacral region) are larger. Fewer than half of all schwanno- •
mas have an evident attached nerve, which is most often small '
and draped over the tumour capsule. Except for those arising
in CNS parenchyma, skin , viscera , and bone, the tumours are
bl
, 1 jj• •

usual!~ en?aps_ ulated ._ Sectioned tumours reveal firm , light-
tan, glistening tissue, interrupted by white/yellow areas and/or
I ':J , ,,
• I
, f 41 '
I I

patches of haemorrhage .

Histopathology
I,
' .~·
Conventional schwannoma is usually an encapsulated spin-
I
I
I
' •.
' f I
dle cell tumour composed nearly entirely of well-differentiated
Schwann cells. Schwannomas have a broad morphological
I
range. The large majority are biphasic tumours with compact
areas (Antoni A tissue) showing occasional nuclear palisad- •
' (
ing (Verocay bodies), alternating with loosely arranged foci
(Antoni B tissue). Cells of Antoni A tissue have modest eosino-
' • • ·, #' • ' '1
philic cytoplasm; no discernible cell borders; and normochro-
! .t I . . a-• • "
Flg. 6.04 Syndrome-associated schwannoma. Mosaic pattern of SMARCB1 (INl1 )
mic, elongated, tapered nuclei. Cytoplasmic nuclear inclu- immunostaining, suggestive of a schwannoma arising In neurofibromatosls type 2 or
sions, nuclear pleomorphism, and mitotic figures may be seen . schwannomatosis.
Palisading (Verocay bodies) takes the form of parallel rows of
Schwann cell nuclei separated by their aligned cell processes. schwannomas, albeit most often at their peri phery (2212] . EMA
Antoni B tissue commonly contains a cobweb-like network of highlights perineurial cells in the capsule , if present.
tumour processes with collections of lipid-laden histiocytes and Ancient schwannoma: Th is subtype d iffers from conven-

I
thick-walled, hyalinized blood vessels . Lymphoid aggregates tional schwannoma only by the presence of scattered atypical
are often present in a subcapsular distribution or at the periph- to bizarre-appearing nuclei, a feature that is often considered '
ery in unencapsulated tumours. A minority of schwannomas degenerative. Such cases may show ex tensive hyalinization or
deviate from the description above. Eighth cranial nerve and central ischaemic changes .
intestinal schwannomas predominantly show Antoni A tissue. Cellular schwannoma: This subtype is c omposed exclusively
The most extreme deviation is seen in the morphological sub- or predominantly of Antoni A tissue and is devoi d of Verocay
types (see below). bodies. The tumours most commonly involve large nerves and
Diffuse staining for S100 in cell nuclei and cytoplasm , which nerve plexuses at paravertebral sites and in the mediastinum ,
is more prominent in Antoni A areas than in Antoni B areas, is retroperitoneum , and pelvis [3475 ,3425 ,2441 ]. Cranial nerves
found in all tumours and subtypes . Similarly, SOX10 immunore- are rarely affected {857}. In add ition to the c ells being closely
activity is usually extensive {2276,1556}. Expression of GFAP is packed , they are often hyperchromatic and mitotically active
less frequent and more variable. Retroperitoneal and mediastinal Small areas of necrosis may be seen . These features may
lesions are commonly positive for keratin AE1/AE3 due to cross- raise suspicion for malignant peripheral nerve sheath tumour
reactivity with GFAP {890} . In contrast to the lattice-like stain- (MPNST); however, the presence of conventional feature s of
ing pattern in neurofibromas, CD34 is commonly positive only schwannoma (encapsulation , subcapsular lymphocytes , hya-
in subcapsular areas , although a small subset of cases show linized blood vessels , and Schwannian whorls) aid in this di s-
more extensive positivity. Staining for NFP is helpful in identi- tinction . Cellular schwannoma shows Ki-67 labelling hotspots
fying entrapped intratumoural axons, found in many sporadic (rather than diffuse inc re ases) , often with an index of< 20% (bu t

Flg.6.05 Epithelioid schwannoma A MululolJular growth pattern at low rnagnificat1on B Loss ot SMARCB1 expression which occ . , t
· ur s 1n 40010 o cases.

C rJri ·11 I ·
< 'c. anL p a r asp1 na l nerve turnour :::i 263
index values 20% do not exclude the diagnosis), and p16 and Box&.01 Diagnostic criteria for schwannoma
H3 p . ~28me3 (K27me3) positivity are retained {2441) .
Plexrfor~ schwannoma: Tumours of this subtype, which can Essential:
be conventional or cellular. often arise in skin or subcut·ane OLIS Histopathology of schwannoma, such as Antoni A or Antoni 8 areas
. . .
tissue. growing as thinly encapsulated plexiform or multinodular AND
tumours {244,9.47) . Less frequently, these tumours can occur in Ex1ensive S1 00 or SOX10 expression
t~1e deep .so'.t tissues .{25) ._ The tumours come to clinical atten-
tion early 1~ life._ often 1n childhood and even at birth {3476). with Desirable:
some predilect10~ for the trunk and the head and neck region. Verocay bodies
Most are spora?1c, _ b ut s_ o me occur in patients with NF2 or Subcapsular lymphocytes
schwannomatos1s . 81phas1c plexiform schwannomas a
·1 .d ·t· bl re more Hyalinized blood vessels
rea d 1y 1 ent1 1a e path?logically than are cellular examples.
Lack of a lattice-like pattern of CD34 staining
The tumours generally differ from conventional schwannoma in
Loss of SMARCB1 (INl1) expression (epithelioid schwannoma), or a mosaic
that t~ey ~a~k a well-formed capsule an d thick-walled vessels.
pattern of SMARCB1 (INl1 ) expression (syndrome-associated schwannoma)
_Ep1thelro1d ~chwannoma: Most epithelioid schwannomas
anse_as .sporadic _tumours, although some may be multiple and/
or arise in .the setting of schwannom atosis {12421479} . . Tum ours schwannoma, and the tumour cells are negative for more spe-
sh~w multliobulat~d .growth .of epith elioid cells arranged singly cific melanocytic markers, such as HMB45 11000); therefore.
or in nests, set w1th1n a vari ably myxoid or hyalinized stroma. these should not be equated with the more aggressive , Carney
Tumour cells have amphophilic to eosinophilic cytoplasm and complex / PRKAR1A- associated malignant melanotic nerve
uniform , round nuclei with small or inconspicuous nucleoli sheath tumour (previously termed "melanotic schwannoma")
occasionally with pseudoinc lusions . Some tumours may sho~
conventional areas of spind led morphology, Anton i A or Antoni s Cytology
tissue , and hyalinized vessels. Loss of SMARCB1 (INl1) expres- Asp irate smears of schwan noma typically yield cohesive syn-
sion is observed in approxi mately 40% of cases , associated cytial fragments of spindle cells 1532}. Within the fragmen ts,
with SMARCB1 inactivation {1479,2820}. Some examples show variably wavy and bent tumour cell nuclei with tapered edges
increased cytological atypia, and rare cases undergo malignant and fibrillary cytoplasm are seen. Nuclear pleomorphism or
transformation to epithelioid MPNST {1478}. degenerative atypia and intranuclear inclusions may be seen .
Microcystic/reticular schwannoma: This is the rarest sub- Schwannomas may be difficult to distinguish from other spindle
type of schwannoma, and th ese tumours seem to preferentially cell neoplasms on cytolog ical preparation alone, and their diag-
arise in visceral sites, most commonly in the gastrointestinal nosis requires correlation with core biopsy and/or immunoh1sto-
tract \1891) . In visceral sites, lesions are often unencapsulated. chemical staining {40}.
Microscopically, tumours are c haracterized by a microcyst-rich
network of interconnected bland spind le cells with eosinophilic Diagnostic molecular pathology
cytoplasm , associated with a myxoid , fibrillary, and/or hyalinized Loss of chromosome 22q and/or mutation of NF2 in schwan-
collagenous stroma. Antoni A tis sue is frequent , and tumours nomas are frequent but nonspec ific molecular alterations.
show strong and diffuse expression of S100. However, conven- Schwannomas exhibit a distinct DNA methylation pattern {460.
tional features of hyalinized blood vessels, foamy histiocytes, 2709,1676,31}.
and Verocay bodies are generally absent.
Other patterns: Although most syndrome-associated Essential and desirable diagnostic criteria
schwannomas are not histologically distinguishable from their See Box 6.01.
sporadic counterparts , several clinicopathological clues may
indicate a setting of NF2 or schwannomatosis: young patient Staging
age, multiple tumours , extensive longitudinal involvement of a Not clinically relevant
nerve , a discontinuous or multinodular growth pattern, and a
mosaic SMARCB1 immunostaining pattern (an admixture of Prognosis and prediction
positive and negative nuclei) {2421,443) . Some schwannomas Schwannomas are benign and do not usually recur if treated
feature predominantly small blue round cells , and may or may by gross total resection . Cellular and plexiform examples are
not have structures resembling Homer Wright rosettes or giant least amenable to total removal and sometimes can only be
rosettes, which surrou nd collagen fibres resembling those of debulked . Malignant transformation of conventional schwan-
low-grade fibromyxoi d sarcoma; these cases are often referred noma is exceptionally rare; in the small number of cases
to as neuroblastoma-like, although they lack increased prolif- reported to date, it has most often taken the form of epithelioid
erative activity and show a typical schwannoma immunoprofile MPNST {3477,2058 ,476) . Less common examples feature foci
\1728 ). Another rare pitfal l is a schwannoma wi th neuromelanin- of conventional MPNST, primitive neuroectodermal cells , rhab-
like pigment accumulation that is positive on Fontana- Masson domyosarcoma, and/or angiosarcoma (3477,3223,2058 ,1766.
staining. Nevertheless , the histology is otherwise typical of 476,27 ).

264 Crc.ir11Ct.I anli f.JdfaSD:r1al r1e1ve tu rrl(Jlil'


Neurofibroma Rodriguez FJ
Reuss DE
Stemrner-Racham1rnov AO

Definition
Clinical features
Neur?fi.broma is a benign peripheral nerve sheath tumour Cutaneous neurofibromas are usually asymptomatic and most
cons1st1ng of mature neoplastic Schwann cells intermixed with commonly occur as a mass . They are soft, mo?ile lesions with-
non-neoplastic cell types. All subtypes are considered CNS out a particular anatomical predilection . Patients with deep
WHO g~ad~ 1, ~xcept atypical neurofibromatous neoplasm of tumours often present with motor or sensory symptoms in the
uncertain b1olog1cal potential (ANNUBP), which is not assigned distribution of the affected nerve. Least commonly, the tumour
a grade. appears as a plaque-like, cutaneous and subcutaneous mass ,
mainly in the head and neck region, or as massive soft tissue
ICD-0 coding enlargement of a body region such as the shoulder or pelvic
9540/0 Neurofibroma girdle in patients with NF1 . The presence of multiple neuroflbro-
9550/0 Plexiform neurofibroma mas, a plexlform neurofibroma , or a massive soft tissue neurof 1-
broma should raise suspicion of underlying NF1 .
ICD-11 coding
2A02.3 & XH87J5 Benign neoplasm of cranial nerves & Neu- Epidemiology
rofibroma , NOS Neurofibromas are the most common peripheral nerve sheath

I
tumours , and the majority are sporadic solitary lesions . Less
Related terminology often , they are multiple in individuals with NF1 . Plexiform tumours
None are often congenital (2450}, whereas the localized cutaneous
and localized intraneural neurofibromas in NF1 begin to appear
Subtype(s) at about 5- 1Oyears of age. All demographic groups are affected
Cellular neurofibroma; atypical neurofibroma I atypical neurofi- and there is no sex predilection .
bromatous neoplasm of uncertain biological potential; plexiform
neurofibroma; diffuse neurofibroma; nodular neurofibroma; Etiology
massive soft tissue neurofibroma Unknown

Localization Pathogenesis
The most common site is the skin , with predominant dermal Conventional neurofibromas (including subtypes)
involvement. Less often involved are more-deeply situated A biallellc genetic inactivation of the tumour suppressor gene
medium-sized nerves, a nerve plexus, or a major nerve trunk. NF1 in a Schwann cell subpopulation is generally the only
Tumours may also arise from spinal nerve roots . Bilateral recurrent somatic event detectable {846,2450). Complete loss
involvement of multiple spinal roots is typical of neurofibroma- of function of the NF1 gene product , neurofibromin (NF1), is
tosis type 1 (NF1). Involvement of dorsal root ganglia may be considered a prerequisite for tumour development. Neurofi -
present. Cranial nerve examples are exceptional. b romin (NF1) is a negative regulator of RAS signalling and

'
Flg.6.06 Neurotibroma . Tota l spine MRI rn a patient w1t11r 1euro l1br ornato s1 ~ typt: 1 w1!11 uxte11s1ve bilateral µa rasp11 1al disease burden Neurolibrornas rnvolv~ nedrly every rierve
root ; also note the thoracrc spine curvalure detecr lsc.ol1os1s)

r r,:ri .. •1 ..H' ! p 1• 1-, , l1fldl nerv run '"'ur" 2u~


Fig.6.08 Neurofibroma. A Increased cellularlty (cellular neurofibroma) may be present usually as a focal finding In large, longstanding neurofibromas, but it lacks prognostc
significance in the absence of other atypical features. B Concentric arrangements (whorls) of Schwann cells around axons, resembling onion bulbs.

acts as a RAS-GAP {2938) . The best-characterized signalling Atypical neurofibroma I atypical neurofibromatous
pathways active in the context of NF1 tumorigenesis are the neoplasm of uncertain biological potential
RAS/RAF/MEK/ERK and Pl3K/AKT/mTOR pathways , which Histological features of atypical neurofibroma (AN )/AN NUBP
play important roles in cell growth, survival , (de)differentiation, described in the setting of NF1 are strong ly associated with
and migration (1923) . However, inacti vation of Nf1 alone was deletions of the CDKN2A and/or CDKN28 locus encod ing cell-
insufficient for neurofibroma development in several mouse cycle regulators p16 (p161NK4a) , p14ARF (both encoded by
models , because a contribution of the microenvironment is CDKN2A) , and/or p15 (p151NK4b; encoded by CDKN28) [231,
also requ ired . There is increasing evidence that inflammatory 2709,2109,474,2449) . One study showed an association of het-
si gnals mediated by various components of the microenviron- erozygous COKN2A and/or COKN28 deletion with cytolog1ca'
ment (e .g . mast cells , macrophages , lymphocytes , and den- atypia alone, and of homozygous COKN2A and/or COKN28
dritic ce lls), as well as Schwann cell interactions with axons, deletion with AN/ANNUBP histology, in different parts of the
are important for tumour development {2552,948 ,1888,1887) . same tumour (474}. Another study showed additional heterozy-
NF1 haploinsufficiency of the microenvironment and nerve gous loss of SMARCA2 in a portion of AN/ANNUBP, either as
injury may promote tumorigenesis (1887,2660). Dermal and part of a larger deletion together with COKN2A and/or COKN28
plexiform neurofibromas exhi bit distinct DNA methylation pro- or in the form of a separate , smaller deletion event {2449).
files , suggesting that they have d ifferent cells of origin {2709) .
Consistent with this assumption , dermal skin - derived and Macroscopic appearance
Schwarm cell precursors in embryonic nerve roots were identi- Five macroscopic forms are distinguished : localized/nodular
fied as cells of origin tor dermal and plexiform neurofibromas, cutaneous , diffuse cutaneous, localized/nodular intraneural.
respectl'1ely, tn transgenic mou se models 1552 ,1819). More plexiform intraneural , and massive diffuse soft tissue neu-
rece1 tly, it tia~; tJeen ~huwn 1hat Nf1 loss in a neura l crest- rofibroma . Localized cutaneous neurofibromas can have d
cjenved HOY8/ lineage cell population leads to both dermal varie ty of gross appearances , including flat , sessile, globular
arid plex1form 11eurof1brurnd development 111 mice, and Hippo and peduncu lated, wh ereas diffuse neurofibromas typically
pathway ac11vat1on acts as a rnod1iicr [b53 l form large plaq ues {2335 f. lntraneural neurofibromas occur a::>
solitary fus iform masses or as ropy to worm -like growths when
plex11orm. Massive soft tissue neuro fibromas range 1n sl1ap~
from a relatively unitorm reg ional soft tissue enlargt:J rn~mt to

266 Gr<J'l:di arid !Jdf.1'> 11ir1dl 11C::r J( 1•11 11r11w.


B
fll, 6.09 Plexiform neurofibroma. A Gross appearance, demonstrating a tortuous architecture and a yellow, slightly heterogeneous cut surface. B By definition , involvement of
multiple fascicles must be present, which imparts a multlnodular appearance on low magnification. c Higher-power view of the adjacent enlarged nerve fi bres.

pendulous bag-like or cape-like masses. The skin overlying mas- Neurofibroma with atypia (ancient neurofibroma) is charac-
sive tumours commonly shows hyperpigmentation. Cut surfaces terized by scattered bizarre nuclei with smudgy chromatin in
of neurofibromas are most often uniformly tan or greyish-tan, the absence of other worrisome histological features. This is not
glistening, mucoid, semitranslucent, and firm . On neuroimaging, considered a premal ignant ch ange, an d should therefore not be
ANNUBP or malignant peripheral nerve sheath tumour (MPNST) confused with AN/ANNUBP as described below.
arising from a plexiform neurofibroma is suspected when there Cellular neurofibroma is defin ed by hypercellularity in the
is a distinct growing nodule and/or increased PET activity (1307) . absence of other worrisome features. Increased cell crowd-
ing leads to a generally blue appearance at low magnifica-
Histopathology tion . These cellular neurofi bromas may even show a fasc1cular
Neurofibromas are characterized by cytologically bland spin- growth pattern , but they lack the uniform cytological atypia,
dle cells with thin , wavy nuclei representing the neoplastic chromatin morphology, and mitotic activity seen in MPNST.
Schwann cell , immersed in a variably loose myxoid stroma . Plexiform neurofibroma is defin ed by its involvement of mul-
The tumour cells are typically smaller than those of schwan- tiple nerve fascicles , each surrounded by perineurium . It most
noma. Stromal collagen is characteristic , colourfully likened in often involves a large nerve or plexus , imparting a bag-of-worms
classic pathology descriptions to shredded carrots . A variety or ropy gross appearance. It is highly associated with NF1 and
of other cells are also identifiable in neurofibroma, including an increased risk of transformation to MPNST (264'1).
perineurial and perineurial-like cells , fibroblasts , and mast Pseudomeissnerian bodies or corpuscles are most often
cells. Even when localized at the gross level , neurofibromas seen in diffuse and pl exiform neurofibromas. They are delicate,
typically lack a capsule and tend to infiltrate adjacent soft tis- round , layered structures and are strongly labelled by S100
sues and parent nerves, in contrast to the more circumscribed immunohistochemistry. In the rare massive soft tissue subtype
schwannoma. Nerve fibres are easily identifiable in intraneu- limited to ind ividuals with neurofibromatosis, extensive infiltra-
ral subtypes , which are characterized by expansion of single tion of soft tissue and even skeletal muscle may be present.
(loc alized ) or multiple (plexiform) nerve fasc icles , but they may Pseu domeissnerian corpuscles are frequent in this subtype, as
be rare in cutaneous and soft tis sue locations. Entrapped gan- are cel lu lar areas containing cells with high N:C ratios . Although
glion ce lls may be conspicuou s in neurofibromas th at infiltrate they may be alarming at first glance , proliferative activi ty is very
dorsal root ganglia , and tl1ese should not be mistaken for gan- low. Other histological features that may be identifiable 1n individ -
glioneuroma. The ind ividual fascic les are recognized by the ual neurofibromas include Schwann cell nodules. S100 -positi ve
outlining perineurium that is com posed of EMA-pos itive cel ls. onion bulb- like Schwann cell proliferati ons , melani n pigment.

•.

f


.. •

•' '
• ' •

Cranial and Parasplnal nerve tumours 267


~ A .. • B ... . .
flg.&. 11 Atypica.I neurofibromat~us ne.oplasm of ~ncertain biological potential (ANNUBP). A Tumours designated as ANNUBP in patients with neurofibromatosls type 1 may
have several worrisome features, including cytological atypla. B ANNUBP with increased cellularity and mitotic activity, bu t otherwise falling short of a malignant diagnosis_

metaplastic bone, epithelioid change, and even glandular dif- Box&.02 Diagnostic criteria for neurofibroma
ferentiation .
Essential:
Markers of mature Schwann cells highlight the neoplastic
Infiltrative, low-cellularity spindle cell neoplasm associated with a variably myxoid to
component, including 8100, SOX10, and collagen IV, albeit to
collagenous stroma and a mixed cell population
a lesser extent than in schwannomas. Non-neoplastic compo-
nents are also intermixed, including scattered EMA-positive Desirable:
and GLUT1-positive perineurial cells, as well as CD34-positive S100 positivity in the Schwann cell population , with a lattice-like CD34 pattern.
stromal cells. lmmunostaining for p16 typically highlights a sub- highlighting the stromal component
set of tumour cells of neurofibroma, whereas complete loss of lntraneural localization
expression is often seen in foci of ANNUBP and MPNST NFP Patient has neurofibromatosis type 1
highlights entrapped axons . Staining for p53 is usually negative, Atypical histological features (nuclear enlargement, hypercellularity, architectural
and the Ki-67 proliferation index is low. H3 p.K28me3 (K27me3) loss, mitoses) for atypical neurofibroma I atypical neuroflbromatous neoplasm of
is retained in neurofibroma and ANNUBP, but it is frequently lost uncertain biological potential in the setting of neurofibromatosis type 1, often with
in MPNST loss of p16 expression
AN/ANNUBP is characterized by at least two of the follow-
ing worrisome features : cytological atypia, hypercellularity, loss
of neurofibroma architecture (on H&E and/or CD34 staining), note, conventional dermal and pl exiform neurofi brom as . AN/
and a mitotic count of > 0.2 mitoses/mm 2 and < 1.5 mitoses/ ANNUBP, and MPNST all exhibit distinct DNA methyl ation pro-
mm 2 (equating to > 1 mitosis/50 HPF and < 3 mitoses/10 HPF files (2709). The presence of SUZ12 or EEO mutations, leading
of 0.51 mm in diameter and 0 .2 mm 2 in area) {2109}. This is to H3 p.K28me3 (K27me3) loss, is restricted to MPNST
considered a premalignant or early malignant change that falls
short of the diagnostic criteria for MPNST but is associated Essential and desirable diagnostic criteria
with increased risk of progression to MPNST {1307}. The term See Box 6.02 .
"ANNUBP " is applied to NF1-associated tumours and is not cur-
rently applicable to sporadic lesions. Staging
Staging is not applicable, although one study showed that AN/
Cytology ANNUBP is associated with low recurrence rates even when
lntraoperative smears and FNA specimens are often paucicel- surgical margins are positive, suggesting that an overly aggres-
lular due to the increased collagen stroma in neurofibromas. sive surgical approach may not be necessary (255}.
Nevertheless, the presence of a mucin-rich background and
small spindled cells with thin , wavy nuclei can provide diagnos- Prognosis and prediction
tic clues . Identification of mitoses is a worrisome finding . Localized cutaneo us neurofibromas are consi stently benign
Plexlform neurofibroma, ANNUBP, an d solitary in traneural neu -
Diagnostic molecular pathology rofibroma arising in sizeable nerves can be precursor lesions ot
Molecular analyses do not have an established role in the MPNST. The lifetime risk for MPN ST in patients wi th NF1 is esti-
diagnosis ot neurofibrom a. However, chromosomal copy- mated at 9- 13% \870) . Diffuse cutaneous neurofi bromas rarely
number profiling may be helpful for the evaluation ot AN/ undergo malignan t transformati on 12821 ). Massive sof t tissue
ANNUBP (CDKN2A and/or COKN2 8 deletion) and its differen- neurofibromas , invariably benign , may never theless overlie an
tiation from MPNST (compl ex, highly rearranged genome). Of intraneural or pl exiform neurof1 broma- derived MPNST
Perineurioma Paulus W
Reuss DE
Stemmer-Rachamimov AO

Definition Epidemiology
Perineurioma is a benign tumour composed of neoplastic peri- lntraneural perineurioma typically affects young adults and ado-
neurial cells (C NS WHO grade 1). lescents (78). although rarely it occurs In children (913f . There
is no sex predilection . The incidence of soft tissue perineurioma
ICD-0 coding peaks in middle-aged adults, and the M:F ratio is 1:2 {1345) .
9571/0 Perineurioma Both the intraneural and soft tissue subtypes of perineurioma
are rare, accounting for approximately 1% of nerve sheath and
ICD-11 coding soft tissue neoplasms , respectively. More than 50 cases of
2A02 .3 & XHOXF? Benign neoplasm of cranial nerves & Peri- intraneural perineurioma and more than 300 cases of soft tissue
neurioma, NOS perineurioma have been described (919,1093 ,1158,1345,2618} .

Related terminology Etiology


Not recommended: localized hypertrophic neuropathy (for intra- Unknown
neural perineurioma).
Pathogenesis
Subtype(s) Like meningiomas, the majority of intraneural perineuriomas
Soft tissue perineurioma; intraneural perineurioma; reticular harbour missense mutations in TRAF7, affecting the WD40
perineurioma; sclerosing perineurioma domains of the protein. Some TRAF7-wildtype intraneural peri-
neuriomas were shown to carry large genomic deletions/dupli-
Localization cations, including deletions of 22q (1649] .
lntraneural perineuriomas primarily affect peripheral nerves of Soft tissue perineurlomas do not carry TRAF7 mutations but
the extremities; cranial nerve lesions are rare (82,593}. Soft tis- commonly show chromosomal alterations. The most frequently
sue perineuriomas are located in the deep soft tissue and are observed alterations are deletions of 22q (including the NF2
grossly not associated with nerves . One example involving the tumour suppressor gene) and deletions of 17q11 (encompassing
CNS arose within a lateral ventricle (1095}. the NF1 tumour suppressor gene) as well as 2p deletions (477} .
The sclerosing subtype of soft tissue perineurioma shows recur-
ClinicaJ features rent rearrangements or deletions of chromosome 10q (379} .
In intraneural perineu rioma , progressive muscle weakness
(with or without atrophy) is more frequent than sensory dis- Macroscopic appearance
turbances . Patients with soft tissue perineurioma present with lntraneural perineurioma produces a segmental , several-fold
nonspecific mass effects. Both intraneural and so'ft tissue enlargement of the affected nerve . Individual nerve fascicles
perineuriomas have been described in patients with neurofi- appear coarse and pale. Most lesions are < 100 mm long , but
bromatosis type 1 (NF1) and type 2 (NF2); however, these are one 400-mm -long sciatic nerve example has been reported
rare cases and it is unclear whether there is a true association (847) . Although multiple fascicles are often involved . a bag -of-
(2823,51,3423,24521 . worms plexiform growth is not seen . Involvement of two neigh-
bouring spinal nerves has been reported (847} .

Cranial and paraspinal nerve tumours 269


Box&.03 Diagnostic criteria for perineurioma
ranges from weak and focal to strong and diffuse [13451: clau-
lntraneural perineurioma
din-1 and GLUT1 are also often positive 1951 .3508) . CD34 ·s
Essential: expressed in about 60% of soft tissue penneuriomas {13451
Pseudo-onion bulb pattern on cross-section, with axons in the centre Staining for S100, SOX10 , and GFAP is negative .
AND Some rare subtypes such as reticular and sclerosing pen -
Tumour cells are positive for at least one perineurial antigen
neuriomas are described in the Soft tissue and bone tumours
(EMA, claudin-1, GLUT1) and negative for S100 volume of this series. The very rare malignant perineunoma is
usually considered a malignant peripheral nerve sheath tumour
Soft tissue perineurioma
with perineurial differentiation {1318 ,2128) and is discussed
Essential: in Malignant peripheral nerve sheath tumour (p. 273). Hybrrd
Slender spindle cells with bipolar cytoplasmic processes nerve sheath tumours may include a perineurioma component.
AND
Storiform and/or whorled architecture Cytology
There is little experience with cytological diagnosis of these
AND
lesions, but FNA specimens are said to be paucicellular, with
Tumour cells are positive for at least one perineurial antigen
smears containing fragments of myxoid stroma {1839).
(EMA, claudin-1 , GLUT1) and negative for S100

Diagnostic molecular pathology


Soft tissue perineurioma is solitary, generally small(< 70 mm), Molecular analyses do not have an establ ished role in the diag-
and well circumscribed but unencapsulated . The cut surface is nosis of perineurioma. Because of the rareness of perineurioma.
firm and greyish white and occasionally myxoid . a reference group for its DNA methylation-based classif ication
is not currently available.
Histopathology
lntraneural perineurioma consists of concentric layers of peri- Essential and desirable diagnostic criteria
neurial cells around axons, forming characteristic pseudo-onion See Box 6.03 .
bulbs. This distinctive architectural feature is best seen on cross-
section, wherein fascicles vary in cellularity. Perineurial cells Staging
accumulate in the endoneurium, but the perineurium is often Not relevant
affected as well. Particularly large whorls can envelop numerous
nerve fibres . In early lesions, axonal density and myelination may Prognosis and prediction
be almost normal, whereas in fully developed lesions, when most Long -term follow-up of intraneural perineuriomas indicates that
fibres are surrounded by perineurial cells and therefore widely they do not have a tendency to recur or metastasize. Radiologi-
separated , myelin is often scant or absent. At late stages, only cal follow-up studies have shown that intraneural penneu nomas
Schwann cell s without accompanying axons may remain at the only rarely grow in length and do not grow to involve new nerves
centre of the perineurial whorls . Hyalinization may be prominent. or nerve divisions, and that growth does not correlate with lin:-
Soft tissue perineuriomas are composed of sp indle cel ls with cal progression {3456}.
wavy or tapering nuclei, indi stinct nucleoli, and bipolar cyto- Soft tissue perineuriomas are usually amenable to gross
plasmic proc&sses . They typi c ally exhibit a storiform or whorled total removal. Recurrences are very infrequent, even 1n cases
growth pattern The stroma is usually collagenous . Mitoses are with histological atypia/degeneration , and there have been no
very rare to absent Necro0is 1s typi cally absent. Degenerative reported metastases (919,1158). Malignant progression ot ~ort
featu res 1nay 1 n c l ud~ nuclear pleomorphi sm, multi nucleated tissue perineurioma has not been documented . Malignant peri-
cell s and a myxo1 d matri x 11 3 45). neurial tumours with high mitotic activity and poor prognos1
lmmunot 11stochem1cally, tumour cell s of intraneural and soft including metastases correspond to malignant peripheral narv~
tissue peri neuriorna are cor1s1stently pos1t1ve tor MA , wh1ct1 sheath tumours [ 13 18 ).
Hybrid nerve sheath tumours Stemmer-Racham1mov AO
Reuss DE

Definition followed by hybrid neurofibroma/schwannoma. These tumours


Hybrid nerve sheath tumours are benign peripheral nerve occur over a wide age range , with a peak in young adults and
sheath tumours with combined features of more than one con- an equal sex distribution (13441. Hybrid neu rofibroma/penneu-
ventional type (neurofibroma, schwannoma, perineurioma). rioma is the rarest.

ICD-0 coding Etiology


9563/0 Hybrid nerve sheath tumour Hybrid schwannoma/perineurioma occurs sporadically [13441,
whereas hybrid neurofibroma/schwannoma is strongly associ-
IC0-11 coding ated with neurofibromatosis type 1 (NF1 ), neurofibromatosis
2A02.3 & XH01GO Benign neoplasm of cranial nerves & Hybrid type 2 (NF2), and schwannomatosis (1237}. A high prevalence
nerve sheath tumour of hybrid neurofibroma/schwannoma morphology (71 %) is
found in tumours from patients with schwan nomatosis, often
Related terminology leading to misdiagnosis as neurofibroma {1237,1967) . Hybrid
Acceptable: benign peripheral nerve sheath tumour NOS . neurofibroma/perineurioma has been descri bed in association
with NF1 [1410 ,1521 }.

I
Subtype(s)
Schwannoma/perineurioma; neurofibroma/schwannoma; neu- Pathogenesis
rofibroma/perineurioma The pathogenesis of the dual-differentiation characteristic of the
hybrid tumour is unknown . Activati ng ERBB2 mutations have
Localization been identified in a subset of neurofibroma/schwannoma hybrid I
Tumours show a wide anatomical distribution in somatic soft tis- tumours , which fall In a DNA methylation subcluster contain-
sue, most commonly occurring in the dermis or subcutaneous ing the majority of neurofibroma/schwannoma hybrid tumours
tissue {1344,898). Rare cases arise in cranial nerves . The most associated with sporad ic schwannomatosis [2719) .
commonly reported site for hybrid schwannoma I reticular peri-
neurioma is the fingers {2105) . Macroscopic appearance
Grossly, the tumours are well ci rcumscribed, with a firm cut sur-
Clinical features face . Their appearance is simi lar to that of other benign periph-
Hybrid nerve sheath tumours occur as painless masses in sub- eral nerve sheath tumours .
cutaneous tissue or dermis. When large peripheral nerves or
spinal nerves are involved , the tumours may be associated with H istopathology
pain or neurological deficit. Hybrid schwannoma/perineurioma shows a storiform or fascicu-
lar growth and is composed of Schwann cells with plump nuclei
Epidemiology and eosinophilic cytoplasm admixed with perineurial cells with
Hybrid nerve sheath tumours are rare . The most common sub- slender nuclei and delicate, elongated cytoplasmic processes
types show hybrid features of schwannoma and perineurioma, The Schwann cell component is often prominent. The tumours

Cranial and paraspinal nerve tumours 2 71


J
Box&.04 Diagnostic criteria for hybrid nerve sheath tumours
is a monomorphic Schwann cell population in the schwanno-
matous component (diffuse expression of S100 and SOX10)
Essential: (898,1237) .
Intermingled features of two types of benign neNe sheath tumours Hybrid neurofibroma/perineurioma contains a p lexiform neu-
AND rofibroma with areas of perineurial differentiation. These areas
Appropriate immunohistochemical staining for each component are often only recognizable with the aid of immunohistochem-
istry. The perineuriomatous areas are immunopositive for EMA
GLUT1, and claudin-1, and the neurofibromatous areas show
may show degenerative nuclear atypia (ancient change). a mixed population of cells that includes fibrob lasts (CD34).
Mitoses are rare . Rare cases show a biphasic appearance and 8chwann cells (8100 and SOX10) , and perineurial c ells (EMA
a lobulated growth pattern , either with separate schwannoma- claudin-1 , and GLUT1) {1521,23}.
tous and perineurial nodules or with schwannomatous nodules
surrounded by a perineurial component with a reticular growth Cytology
pattern and myxoid stroma . The two components can be high- Not clinically relevant
lighted by immunohistochemistry: 8100-positive Schwann cells
and EMA-positive peri neurial cells. Perineurial cells may also be Diagnostic molecular pathology
immunoreactive for claudin-1 and GLUT1 {1344) . Not clinically relevant
Hybrid neurofibroma/schwannoma is composed of schwan-
nomatous nodules within an oth erwise typical neurofibroma, or Essential and desirable diagnostic criteria
of 8chwann cell bundles dispersed in a myxoid background . See Box 6.04.
The tumours may have a plexiform architecture. The Schwann
cell nodular proliferations may exhi bit Verocay body formation Staging
or fascicular growth, and the neurofi broma component may Not clinically relevant
demonstrate myxoid change , collagen bundles, and mixed
cellular composition . Tumours may contain entrapp ed NFP- Prognosis and prediction
positive axons. lmmunostaining highlights the mixed popu - The tumours are benign and rarely recur local ly (1344} . ERBB2
lation of cells in the neurofibromatous component, incl uding mutations in a subset of neurofibroma/schwannoma hybrid
fibroblasts (CD34) , 8chwann cells (8100 and SOX10), and ~umou rs have been proposed as a potenti al therapeutic target
perineurial cells (EMA , claudin-1, and GLUT't), whereas there 1n unresectable tumours (2719}.

272 C1an1a\ ancJ para~plnd\ nerve turriour::.,


Malignant melanotic nerve sheath tumour Reuss DE
Folpe AL
Jo VY
Stemmer-Racham1mov AO

Definition
Malignant melanotic nerve sheath tumour (MMNST) is a periph-
eral nerve sheath tumour composed uniformly of tumour cells
~I with features of both Schwann cell and melanocytic differen -
I
I tiation, usually arising in association with spinal or autonomic
I nerves. It is variably associated with Carney complex and fre -
I quently shows aggressive clinical behaviour. PRKAR1A muta-
I

tions and loss of PRKAR1A protein expression are seen in the


overwhelming majority of cases .

ICD-0 coding
9540/3 Malignant melanotic nerve sheath tumour

ICD-11 coding
2A02 .0Y Other specified gliomas of spinal cord, cranial nerves ,
or other parts of the central nervous system

Related terminology
Acceptable: malignant melanotic Schwannian tumour.
- Not recommended: melanotic schwannoma ; psammomatous Flg.6.16 Malignant melanotic nerve sheath tumour. Tumour arising in a spinal nerve
melanotic schwannoma. root. The tumour is heavily pigmented and partially encapsulated.

Subtype(s) Etiology
None In some series, > 50% of patients with MMNSTs have evidence
of Carney complex, an autosomal dominant, sometimes familial .
Localization multiple neoplasia syndrome {470) . However, other series have
MMNST most often arises from spinal or autonomic nerves near noted an association with Carney complex in ~ 5% of affected
the midline . However, cases have been reported in the gastro- patients 13266,3219 ,3594 ,3375) . Other cases are considered
intestinal tract /551,557), as well as in bone, soft tissues , heart, sporadic and of unknown etiology.
bronchus , liver, and skin .
Pathogenesis
Clinical features Two genetic loci have been identified in Carney complex :
Presenting symptoms include pain , sensory abnormalities , and PRKAR1A (CNC1) and CNC2, mapping to 17q24 2 and 2p16 ,
mass effect. Bone erosion may be seen in spinal nerve root respectively. PRKAR1A inactivation is seen in roughly 50% of
tumours . Systemic symptoms, such as respiratory and liver fa il- Carney complex kindreds 12043,1639). PRKAR1A encodes ihe
ure, may be seen in patients with metastatic disease. Although type 1A regulatory (R1a) subunit of PKA , which inhibits PKA
it was once thought that psammoma bodies were more likely activity by binding to active catalytic subunits. PRKAR"IA acts as
in familial tumours, there are no clinical differences between a tumour suppressor gene. Loss of R1a leads to increased PKA
psammomatous and non-psammomatous MMNSTs, with both activity, which has been associated with secondary dysregula-
showing a variable association with Carney complex. loss of t1on of the ERK , TGF-p , and WNT signalling pathways (28301
PRKAR1A expression , and similar clinical behaviour [3219 , PRKAR1A mutations and loss of PRKAR1A protein expression
3375) . are seen in the overwhelming majority of studied MMNSTs. most
of which have occurred in patients lacking other stigmata of Car-
Epidemiology ney complex 13219,33751 . Chromosomal copy-number profiling
MMNST is rare and occurs chiefly in adults. The tumour typically of MMNST revealed recurrent whole -c hromosome losses and
develops at an earlier age (average- 22 .5 years) 1n patients with gains . The most frequent alterations are rnonosornies of cl1ro-
Carney complex than 1n sporadic cases (average: 33 .2 years) mosomes 1, 2, 17, 21, and 22q and whole-cliromosorne ga111s
1470,3219). Multiple tumours are seen in about 20% of patients; variably involving chromosomes 5. 6, 7. 8, and 9 [3375 1676 .
in such patients . there 1s a higher probability that other manifes - 4631 Inactivation of both alleles ot PRKAR IA through mutations
tations of Carney complex will also be present than in patients and/or loss of lleter0Lygos1ty ot t?q r1as beer 1 reported (33751
with a single tumour 1470,3219 ]

Grant I and paraspin al nerve tumours 273


Box&.05 Diagnostic criteria for malignant melanotic nerve sheath tumour
Essential: lmmunohistochemically, MMNSTs strongly express S100 and
SOX10, as well as various melanocytic markers , including
Fascicular to sheet-like proliferation of variably pigmented, relatively uniform
plump, spindled to epithelioid cells ' HMB45 , melan-A, and tyrosinase. Basement membrane mark-
ers (collagen IV, laminin) often show increased intercellular
AND
deposition compared with melanoma, although there are many
Coexpression of S100/SOX10 and melanocytic markers (e.g. HMB45, melan-A) exceptions . PRKAR1A expression is typically lost {3219.33751.
OR Ultrastructurally, the cells resemble Schwann cells with elabo-
Loss of PRKAR1A expression, or PRKAR1A mutation rate cytoplasmic processes that interdigitate or spiral in the
AND (for unresolved lesions) manner of mesaxons; however, pre-melanosomes and melano-
Methylation profile of malignant melanotic nerve sheath tumour somes are also present {3594,1365}.

Desirable: Cytology
Origin from a paraspinal or visceral autonomic nerve Not clinically relevant

Diagnostic molecular pathology


Gene expression analyses show MMNST to clearly segregate PRKAR1A immunoexpression is typically lost, corresponding to
from conventional schwannomas and melanomas {3219}. The gene inactivation {3219,3375}. MMNSTs show a distinct DNA
molecular distinctiveness of MMNST is further corroborated by methylation pattern [1676} and do not harbour mutations in
DNA methylation profiling {1676). GNAQ or GNA 11, which is useful for their differentiation from pn-
mary CNS melanocytomas and melanomas . MMNSTs also do
Macroscopic appearance not harbour BRAF, NRAS, or TERT promoter mutations, which is
Most MMNSTs are solitary, although multiple and multicentric useful for their differentiation from metastatic cutaneous mela-
tumours may be seen in patients with Carney complex. Grossly, nomas (1676,1770,3268}.
the tumours appear circumscribed or partially encapsulated,
and they are frequently heavily pigmented, with the appearance Essential and desirable diagnostic criteria
of dried tar. See Box 6.05 .

Histopathology Staging
The neoplastic ce ll s grow in short fascicles or sheets, vary in Not available
shape from polygo nal to spindled, and often have a syncytial
appearance Vag ue pa lisad ing or formation of whorled struc- Prognosis and prediction
tures may be present . Cell ul ar d etail is often difficult to discern, The behaviour of MMNST is difficult to predict and metastases
owing to the l1eavy pigment depos its. The melanin pigment may can occur in the absence of morphologically malignant fea-
be coarsely clumped or fine ly g ranular and varies from area to tures . In the past , it was thought that most of these lesions had
area . It stains pov1t1vely with the Fontana stain and negatively a benign, indolent course, with < 15% metastatic risk \4701
tor iron and PAS . In less pigmen ted areas, the tumour cells have However, more recent reports have shown frequently aggres-
eos1nophilic to amphophilic cytoplasm, round to ovoid nuclei sive behaviour, with local recurrence and metastatic rates of
(of1en with nuclear grooves and pseudoin clusion s), and usu- 26 - 44% 13266,3219,3375 ,1607). Additionally, only 53% of
ally small nucleoli . Occasional tumours show marked nuc lear patients followed for > 5 years have been reported to have
atypia with prominent macron ucleo li . Mitoses and necrosis rem ained di sease-free , suggesting that long-term follow-up is
can be pre sent, but they are not clea rly assoc iated with out- required In general , histopathological features are not predic -
come. Psammoma bodies are prese nt in roughly 50% of cases, ti ve of outcome, although there are limited data suggesting
although extensive sampl ing may be required to identify them.
more agg ress ive behaviour in mitotically active tumours {32191
274 Cranial and paraspinal nerve 1urnuur s
Malignant peripheral nerve sheath tumour Reuss DE
Hirose T
Jo VY
Rod riguez FJ
Stemmer-Rachamimov AO

Definition
Related terminology
Malign~nt peripheral nerve sheath tumour (MPNST) is a malig - Not recommended: mali gnant schwannoma , neurofibrosar-
nant spindle cell tumour often arising from a peripheral nerve, coma , neurogen ic sarcoma .
from a pre-existing benign nerve sheath tumour, or in a patient
with neurofibromatosis type 1 (NF1), and often showing limited Subtype(s) .
Schwannian differentiation. Molecular hallmarks are the com- Epithelioid malignant peripheral nerve sheath tumour ; penneu-
bined genetic inactivation of NF1, CDKN2A and/or CDKN28, rial malignant periphera l nerve sheath tumour
and SUZ12 or EEO genes, as well as complex genomic
rearrangements. Localization
The most common locations are ex tre mities, the trunk , and the
ICD-0 coding head and neck area {1817,796 }.
9540/3 Malignant peripheral nerve sheath tumour
Clinical features
ICD-11 coding MPNST occurs most commonl y in pati ents aged 20-50 years.
2A02.0Y & XH2XP8 Other specified gliomas of spinal cord , cra- MPNSTs in children are usually associated with NF1. The mean
nial nerves , or other parts of the central nervous system & age of patients with NF1-associated MPNST is about a decade
Mali gnant peripheral nerve sheath tumour younger than that of patients with sporad ic tumours . Patients
with MPNSTs often present with enlarging masses that may
cause pain or other neuropathic symptoms (91 2.3034}. PET-CT

Cranial and parasp 1nal nerve tumours 275


imaging is sensitive but not specific for the detection of MPNST in whi ch a subpopulation of Schwann cells alr13wJ·; r,::=.w ·&:.·, . ,
in patients with NF1 /375) . biallelic inactivation of NF1 /1 923) An intermediate 1w)1rJn 1', · r-~.
atypical neurofibromatou s neopl asm of uncertain r1101r;r11r,:o:
Epidemiology
potential (ANNUBP), wh ich fre quently harbours h<Jmnz-rJ'J·~-­
MPNSTs account for approximately 2- 10% of soft tissue sarco- CDKN2A and/or CDKN2B deletions /2109.4741 MP~ISTs ~r~
mas {371,3434) . with epithelioid MPNST being particularly rare molecularly characterized by two additional hallmarks 1 n;:v.111~­
(- 5% of all c ases). The estimated overa ll incidence is 1.46 cases tion of SUZ12 or EEO (core components of PRC2) /217 35%
per 1 mill ion person -years ; there is slightly g re ater risk in Black 1842) as well as complex genomic re arrangements 1nclud1r1
people and lower risk in Asian and Latino/a peopl e than in Wh ite numerous chromosomal deletion s and oncogene amplif1cat1Gric;
people; the M:F ratio is rough ly 1.2: 1 (2 15,2435 !. (1670,2709). In addition , some MPNSTs harbour mutations ,r.
TP53 \33 13,38 1,2984) .
Etiology PRC2 mediates the deposition of H3 p.K28me3 (K27rne3•.
About 50% of all MPNSTs are associ ated with NF1 . In th is set- an important repressive mark that plays a critical role 1n cel-
ting , they most common ly arise from deep-seated plexiform lular diffe rentiation and cellular identity (630. 2013}. Inactiva-
neurofibromas or large intraneural neurofibromas (912) . The tion of PRC2 lead s to a complete global loss of H3 p.K28rrie3
lifetime risk for MPNST in patients with NF1 is 10% (863 ,2052) . (K27me3) in tumo ur cells , wh ich can be demonstrated by
About 10% of all MPNSTs are associated with previous irradia- immunohistochemistry. About 80% of conventional high-grade
tion (950). Epithelioid MPNST is not associated with NF1 , but it MPNSTs show loss of H3 p .K28me3 (K27me3) 12822) . Conven-
has been associated w ith malignant transformation of schwan- tional MPNSTs with loss of H3 p.K28me3 (K27me3) and those
noma and has histolog ical and molecular features in common with retained H3 p.K28me3 (K27me3) form two distinct DN..U
with epithelioid schwannoma (2058 ,2820) . methylation classes (2709 ). Those with loss of H3 p K28me3
(K27me3) frequently display losses of 1p , 9p, 10q, 11 . 17p
Pathogenesis and segments of 17q incl uding NF1 and SUZ12 The most fre-
The pathogene sis of MPNST is complex and incompletely quent gains involve 7p, Sq , and larg er parts of 17q. Amplif ied
understood . Most available data are for NF1-associated oncogenes include EGFR, PDGFRA. and MET Tumours w1tn
tumours . In that setting , MPNSTs commonly develop from retained H3 p.K28me3 (K27me3) are predom inately parasp1na·
plexiform neurofibromas or localized intraneural neurofibromas , and show more frequent losses of 3q and gains of 5p 12709}.

Flg.6.20 Penneurial mal1griant peripheral nerve sheath tumour A Cellular whoils st1ow1ng con,·ent11c anangemen t of tu mour cells. B Storiform and whorling pattern . C lm-
munoreactiv1ty lor EMA.

276 Lr:cJ 111ul <: tr •U I Jd l cJ',p.r 1.11 11, ·r v\ n111 i. J L11


The pathogenesis of epithelioid MPNST is distinct from that may be increased, and tumour cell herniation into blood vessels
of conventional MPNST. Epithelioid MPNSTs are not associ- is a common featu re. Mitotic activity is usually brisk {2109}. Well-
~ted with NF1 and do not harbour most of the genetic altera- demarcated areas of necrosis are frequently present. About
tions present in conventional MPNST; they retain H3 p.K28me3 15% of MPNSTs show evidence of divergent differentiation .
(K27me3) and are driven by genetic inactivation of SMARCB1 There may be mesenchymal osseous, cartilaginous , or rhab-
in the vast majority of cases {1842,1478,2820). Recurrent chro- domyosarcomatous (skeletal muscle) heterologous elements ,
mos~mal alterations in epithelioid MPNST include loss of 22q , or epithel ial elements such as mucinous glands or islands of
deletion of 9p including CDKN2A and/or CDKN2B, and gain of squamous differentiation. Conventional MPNST with prominent
2q {2820} . rhabdomyosarcomatous differentiation has been termed "malig-
The pathogenesis of perineurial MPNST is currently unknown . nant triton tumour". Add itionally, MPNST may show a broader
morphological range and occasionally resemble various mes-
Macroscopic appearance enchymal tumours , including synovial sarcoma , solitary fibrous
Most MPNSTs are > 50 mm in diameter at diagnosis. They have tumour, and und ifferentiated pleomorphic sarcoma . MPNST
a tan-white, fleshy cut surface, often with areas of haemorrhage may diffusely infiltrate peri pheral nerve tissue as well as precur-
and necrosis. They may occur with fusiform enlargement of a sor neurofibromas. Therefore, it is not uncommon to find areas
peripheral nerve and may include a precursor neurofibroma. with different degrees of malignant progression in the same
tumour mass (e.g. neurofi broma , ANNUBP, low-grade MPNST,
Histopathology high-grade MPNST), especially in NF1-associated tumours.
Conventi onal high-grade MPNST appears as a hypercellular To improve reproduc ibility and association with clinical and
spindle cell tumour, with tumour cells arranged in interlacing molecular parameters , a consensus nomenclature for the spec-
fascicles . The nuclei are wavy or buckled and considerably trum of NF1-associated nerve sheath tumours has been pro-
larger and more atypical than in neurofibroma. There is pale posed (Table 6.01 ). The extent of mitotic activity and the pres-
eosinophilic and often indistinct cytoplasm . The cell density ence of necrosis is critical both for the distinction of ANNUBP
often alternates between highly cellular and less cellular areas, from MPNST and for the grading of MPNST (low-grade or high-
creati ng a marbled appearance. The cellularity around vessels grade) {2109) .

Table&.01 Proposed nomenclature for the spectrum of nerve sheath tumours associated with neurofibromatosis type 1

Diagnosis Proposed definition


---·-
Benign Schwann cell neoplasm with thin (often wavy) nuclei, wispy cell processes, and a myxoid to collagenous (shredded carrots) matrix;
Neurofibroma
immunohistochemistry includes extensive but not diffuse S100 and SOX10 positivity and a lattice-like CD34+ fibroblastic network
Plexiform neurofibroma Neurofibroma diffusely enlarging and replacing a nerve, often involving multiple nerve fascicles, delineated by EMA+ perineurial cells
Neuroflbroma with atypia
Neurofibroma with atypia alone, most commonly manifesting as scattered bizarre nuclei
(ancient neurofibroma)
CeUular neurofibroma Neurofibroma with hypercellularity but retained neurofibroma architecture and no mitoses

ANNUBP Schwann cell neoplasm with at least two of the followl.ng four features: cyto~ogical atypia, loss of neurofibroma architecture, hypercellularity,
and a mitotic count of > 0.2 m1toses/mm 2 and < 1.5 m1toses/mm 2 (> 1 m1tos1s/50 HPF and < 3 mitoses/1 oHPF•)
MPNST, low-grade Features of ANNUBP, but with a mitotic count of 1.5-4.5 mitoses/mm2 (3-9 mitoses/1 OHPP) and no necrosis
2
MPNST, high-grade MPNST with a mitotic count of ~ 5 mitoses/mm (~ 1Ornitoses/10 HPF•), or with a mitotic count of 1.5-4.S mitoses/mml
combined with necrosis
ANNUBP, atypical neurofibromatous neoplasm of uncertain biological potential; MPNST. malignant peripheral nerve sheath tumour.
2
·1 mm "' 5 HPF of 0.51 mm in diameter and 0.20 mm 2 1n area

C r rual and Pc ra Ptnal nerve tumours 277


lmmunohistochemically, the majority of MPN STs are negative
for Schwann cell markers like 8100 , SOX10 , and GFAP. Impor-
tantly, if positivity is present, the staining is typically not diffuse
but patchy or restricted to a subpopulation of cel ls , sometimes
attributable to entrapped neural remnants . Only a minority of
MPNSTs retain a Schwann cel l phenotype. Surrogate molecular
immunostains are of ten more helpful than lineage markers. As
many as 90% of conventional MPNSTs show loss of neurofibro-
min [2654,2441 ,2709} , although a commercial antibody is not
currently available . Complete loss of H3 p.K28me3 (K27me3) is
detectable in 50-80% of all MPNSTs, with the highest frequen-
cies reported in high-grade and radiation -i nduced MPNSTs
{2822 ,2576} . A small subset of MPNSTs demonstrate ATRX loss
and an alternative-lengthening-of-telomeres phenotype {2694,
1949}.
The tumour cells of epithelioid MPNST are predominantly
epithelioid with abundant eosinophilic cytoplasm and nuclei
with visible nucleoli . They often show a lobulated growth pat-
tern and a fibrotic or myxoid matrix. lmmunohistochemically,
they are consistently strongly and diffusely positive for S100
and SOX1 O but negative for melanocytic markers . Epithelioid Box&.06 Diagnostic criteria for conventional malignant peripheral nerve sheath tu-
MPNSTs retain H3 p.K28me3 (K27me3), but most show a loss mour (M PNST)
of SMARCB1 expression {2820) .
Essential:
Very rare malignant tumours with perineurial featu res have Histopathologically consistent malignant spindle cell tumour in a patient with Nf 1•
been described and termed "perineurial MPNST". They are OR in a pre-existing neurofibroma
composed of spindle cells arranged in intersecting fascicles
OR
or whorls and exhibit frank histological signs of malignancy,
Malignant spindle cell tumour associated with a peripheral nerve AND no more
such as frequent mitoses and necrosis. lmmunohistochemi- than focal/patchy S100/SOX10 expression AND no SS18::SSX (SSX1, SSX2, or
cally, tumour cells are 8100-negative but express EMA and SSX4) fusion gene present
(variably) CD34 [1318} . Perineurial MPNST is not associated
OR
with NF1 .
Malignant spindle cell tumour associated with a peripheral nerve AND evidence
of PRC2 inactivation (molecularly or via loss of H3 p.K28me3 [K27me3]
Cytology immunostaining)
Not cl inically relevant
OR
Tumour with features of ANNUBP in a patient with NF1 , but with a mitotic count
Diagnostic molecular pathology
of at least 1.5--4.5 mitoses/mm2 (3-9 mitoses/10 HPF of 0,51 mm In diameter and
DNA methylation-based classification may unequivocally 0.20 mm2 in area)
define a tumour as MPNST, especially one with retained H3
OR
p.K28me3 (K27me3). Other molecular data may provide sup-
portive information for the diagnosis of MPNST, such as muta- Unresolved lesion with the methylation profile of MPNST
tions in NF1 , SUZ12, or EEO, or the chromosomal copy-number
Desirable:
profi le. Chromosomal alterations are particularly helpful in bor-
derline cases to distinguish between ANNUBP (COKN2A and/ Loss of H3 p.K28me3 (K27me3)
or CDKN28 deletion and no or very few other alterations) and Loss of neurofibromin expression
MPNST (complex genomic profile) .
ANNUBP, atypical neurofibromatous neoplasm of uncertain biological potential; NF1 .
neurofibromatosis type 1.
Essential and desirable diagnostic criteria •Jn the setting of NF1 , a diagnosis other than conventional MPNST demands strong mo-
See Box 6.06 . lecular evidence (e.g. detection of a fusion gene pathognomonic for another tumour type).

Staging MPNST. Deep location, positive surgical margins , and high-


Not clinically relevant grade histology were additional factors associated with a poorer
outcome \18171. The prognosis of radiation-induced MPNST 1s
Prognosis and prediction even worse , with a reported 5-year overall survival rate of 23 .5%
In a recent study, the 5-year overall su rvival rate of MPNST (2104]. Epithelioid and perineurial MPNSTs seem to be less
as.,,oc1ated with NF1 was 35%, whereas it was 65% fo r sporadic aggressive than conven tional MPNST (1478 ,1318).

27Ll 1r11, .1\11 1 i1r1',i 11cilrlt,1.;1•!11•11>111~~


Cauda equina neuroendocrine tumour Brandner S
Kleinschm1dt-DeMasters BK
(previously paraganglioma) Sarkar C

Definition
Cauda e~~ina neuroendocrine tumour is a neuroendocrine neo-
plasm arising from specialized neural crest cells in the cauda
equina I filum terminale region .

ICD-0 coding
8693/3 ~auda equina neuroendocrine tumour (previously para-
gangiloma)

ICD-11 coding
2A02.0Y & XH1X68 Other specified gliomas of spinal cord, cra-
nial nerves, or other parts of the central nervous system &
Paraganglioma, benign
2A02.0Y & XHOEW6 Other specified gliomas of spinal cord ,
cranial nerves , or other parts of the central nervous system &

I
Paraganglioma, NOS

Related terminology
Fig. 6.23 Cauda equina neuroendocrine tumour. A well-circumscribed. encapsulated
tu mour with prominent vasculature on the surface .
.
Acceptable: paraganglioma of the cauda equina; cauda equina
paraganglioma.

Subtype(s}
None

Localization
The majority of spinal paragangliomas / neuroendocrine tumours
are located in the cauda equina region (1052) . Most neuroendo-
crine tumours of the cauda equina are entirely intradural and are
attached either to the filum terminale or (less often) to a caudal
nerve root {2992) .

Clinical features
Cauda equina neuroendocrine tumours exhibit no clinical fea-
tures that allow their distinction from other spinal cord tumours .
The most common presenting symptoms include a history
of low back pain and sciatica. Less common manifestations
are numbness, paraparesis, and sphincter symptoms (1052] .
Fully developed cauda equina syndrome is uncommon, as are
signs of increased intracranial pressure and papilloedema (21 ,
1052). Endocrinologically functional neuroendocrine tumours
of the cauda equina region , which are extremely rare (1052) ,
can lead to signs and symptoms of catecholamine hypersecre-
tion such as episodic or sustained hypertension, palpitations,
diaphoresis , and headache. Subarachnoid haemorrhage is
Fig. 6.24 Cauda equina neuroendocrine tumour. A Hypointense tumour on T2
another unusual presentation of cauda equina neuroendocrine weighted image with prominent flow voids from enlarged d1a1111ng veins superior
tumours (1878) Cerebrospinal fluid protein is usually markedly ly. B T1 -weighted image with marked contrast enhancemenr.
increased (2981,2992) .

Imaging occasionally partly cystic. and they can be hypo1ntense. 1su111-


MRI findings are nonspecific (21/. and the appearance of tense. or hyperinien se on T1- ancJ T2-we1gt1ted 11nagcs Gado
Paraganglioma is indistinguishable from that of schwannoma or l1nium enhancement may be present or absent. A low signal
ependymoma (2126) . Tumours are sharply c1rcumscnbed and intensity nm (cap sign) on T2-weighted images can be caused

Cranial and paraspinal nerve tumours 279


by subcapsul ar haemosiderin {186 1,3524,3517) . The hypervas- Pathogenesis
cular architecture can give a salt-and -pepper appearance on
Cauda equina neuroe ndocrine tumours are h1stogen8t1c;:i11 18r1
T2-weighted images. Serpentine flow voids along the tumour
molecularly distinct from paragangliomas and phaeo(;hrr.irn•Jr /
surface or spinal cord margin may also be seen at MRI {732,
2068 ). tomas outside the CN S. They overexpress the trnn sr:ript ori '8r: 1

tor HOXB13 f306A ), wh ich is developmentally expressed 1r ''"·"":!


Epidemiology caudal extent of the spinal cord and in the urogenital sirus 1 ~
coincides with dynamic ch anges associated with the format1c,r·
Neuroendocrine tumou rs of the cauda equi na regio n are
of the secondary neural tube f3585A,824AJ, provi ding Girc1Jrn-
uncommon , but nearly 300 cases have been reported si nce
stantial evidence of a cell of origin . Cauda equma neuroendo-
their initial description in 1970 1133 9,2 11 2) . In a series of
crine tumours have not been seen in people with heredit2r1
430 spinal tumours , < 3% we re parag ang liomas / neuroendo-
paraganglioma/phaeochromocytoma syndrome due to muta-
crine tumours (852 ,133} , and of cauda eq ui na region tumours , tions in SDH subunit genes (2165A J.
3.4-3 .8% are neuroendocrine tum ours (3460 ,3524] . Cauda
equ ina neuroendocrine tum ours gen erally affect adults, with Genetic profile
a peak incidence in the fo urth through si xth decades of life. The molecular alterations that drive tumorigenesis in cauda
Patient age ranges from 9 to 75 years (mean : 46- 47 years) equina neuroendocrine tumours are unknown. The mutations
(3517,1339), with a slight predom inance in male patients found in paragangliomas outside the cauda equina region are
(M :F ratio: 1.5:1) (1052,1339].
described in Chapter 14: Genetic tumour syndromes involvmg
the CNS. The genetic and epigenetic profiles (i .e. methylation
Etiology expression (1222,1858). microR NA (701 }, and metabolom1c s
Almost all neuroendocrine tumours of the cauda equina are 1488,1409}) of phaeochromocytomas/paragang liomas with suc -
sporadic . In contrast, as many as half of all phaeochromocyto- cinate dehydrogenase defects differ substantially from those
mas/paragangliomas outside the CNS in adults , and > 80% of with other genetic causes.
these tumours in chil dren , are inherited (1109] . The role of germ-
line mutations in th e pathogenesis of paragangliomas outside Macroscopic appearance
the cauda equina region is discussed in Chapter 14: Genetic Tumours are oval to sausage-shaped , del icately encapsu-
tumour syndromes involving the CNS. lated, soft, red-brown masses th at bl eed freely ; in 5 series

flg.6.25 Cauda eq~Hna r1eu1 oe ndocru;e wrnour A H&E sta1n1ng shows nes ts of uniform round or polygonal chie f cells surrounded by a capillary and fibre network The
nests of tumour cells are surrounded by a delicate suppor ting ret1cu lin fi bre network (ret1culin silve r stain) C Synaptophys1r1 1mmunosta1ni ng labels the neuroendocrine (chief )
cells. D Chromograrnn A immunosta1rnng labels the neuroe ndocr1 ne (c il1ef) cells.
of 59 spinal tumours, size ranged from 10 mm to 112 mm in lmmunophenotype
greatest dimension {2126,3517,2037}. Capsular calcification The neuroendocrine chief cells are immunoreact1ve for chro-

I
and cystic components may be found . An occasional tumour mogranin A and synaptophysin (1661,2992,2095). and they
penetrates the dura to invade bone . For tumours in the cauda show variable S100 immunoreactivity. lmmunoreactivity for .
equina. a macroscopically identifiable attachment may be cytokeratins (especially CAM5 .2, AE1/AE3 , and MNF116) has
found, either to the filum terminale or (less often) to a caudal been described in the majority of cauda equina neuroendocrine
nerve root {2992}. tumours {2332,556 ,3053 ,2095 .1 176,739,2610}, but it has also
been described in other locations {739}.
Histopathology Furthermore, gangliocytic neuroendocrine tumours / para-
Cauda equina neuroendocrine tumours I paragangliomas are gangliomas containing a variable mixture of epithelioid neu-
well-differentiated . They are composed of chief (type I) cells roendocrine cells, Schwann-l ike cells , and scattered ganglion-
disposed in nests or lobules (Zellballen), surrounded by an like cells can show cytokeratin positivity in the epithelioid cells
inconspicuous. single layer of sustentacular (type 11) cells. The {2889,2583} . Expression of serotonin (5-HT) and of various neu-
Zellballen are surrounded by a delicate capillary network and ropeptides (somatostatin , leu-enkephalin , and met-enkephalin)
a delicate supporting reticulin fibre network that may undergo has been demonstrated in neuroendocrine tumour of the cauda
sclerosis. The uniform round or polygonal chief cells have equina region {2162,2992) .
central, round to oval nuclei with finely stippled chromatin and Sustentacular cells show inconsistent S100 protein react1v1ty
inconspicuous nucleoli. Degenerative nuclear pleomorphism 12978,3241) and occasionally express GFAP 12996). They often
(endocrine anaplasia) is generally mild. Cytoplasm is usu - express the neural crest transcription factor SOX10 12276). The
ally eosinophilic and finely granular; in some instances, it is value of proliferation markers in cauda equina neuroendocrine
amphoph1lic or clear. Sustentacular cells are spindle-shaped; tumours has not been established .
encompassing the lobules , their long processes are often so Unlike paragangliomas of the autonomic nervous sys-
attenuated as to be undetectable by routine light microscopy tem {2978). cauda equina neuroendocrine tumours do not
and visible only on immunostains for 8100 . Approximately express the zinc finger transcription factor GATA3 (2610). They
25% of cauda equina neuroendocrine tumours contain mature express HOXB13 (a transcription factor that is developmentally
ganglion cells and a Schwann cell component (gangliocytic expressed in progenitor cells of the caudal spinal cord). a fea -
neuroendocrine tumours) . Ependymoma-like perivascular for- ture they share with myxopapillary ependymoma (306A}
mations are also common. Some tumours show architectural
features reminiscent of carcino1d tumours, including angi - Grading
omatous, adenomatous. and pseudorose1te patterns (2992}. Neuroendocrine tumours of the cauda equina correspond to
Tumours composed predominantly of spindle cells (2162] and CNS WHO grade 1
melanin-containing cells (melanotic neuroendocnne tumours)
(2162] have also been described at this site, as have onco - Cytology
cytic neuroendocrine tumours 11021,2395]. Foci of haemor- Not clinically relevant
rhagic necrosis may occur, and scattered m1tot1c figures can
be seen , but neither these features nor nuclear pleomorphism Diagnostic molecular pathology
is of prognostic significance (2992! . The DNA methylat1on profiles and chromosomal copy-number
profiles of cauda equina neuroendocrine tumours are distinct from
those ot paragangliomas arising from other locations 1261 0,2871] .

Crania l and paraspinal nerve tumours 281


Essential and desirable diagnostic criteria Box 8.07 01agnost1c criteria for cauda ~quina neurrJ'!ndnc:r r'! tiJrrrJ Jr

See Bo 6 07 Essential:
Well-demarcated tumour wi1h Zellballen architecture
Staging
AND
Not rele ant
Synaptophysin or chromogranin immunoreactivity in chief cells

Prognosis and prediction AND


The ast majority of cauda equ1na neuroendocrine tumours are Cauda equina location
slow-growing and cura ble by total excision ; only 4% recur after AND (for unresolved lesions)
gross total removal !3053) . Cerebrospinal fluid seeding of spinal Methylation profile of cauda equina neuroendocrlne tumour
paragangliomas has occasionally been documented (627,2692,
3053 ,3176 ,3190] , and metastasis outside the CNS (to the bone) Desirable:
from cauda equina neuroendocrine tumours has been reported S1CO-positive sustentacular cells
only once \2 162). In contrast, 10-20% of paragangliomas out- Cytokeratin-positive chief cells
side the CNS have metastatic potential (1811 ,639}. Reticulin silver stain showing typical architecture
Meningioma Sahm F
Brastianos PK
Perry A
Santagata S
Claus EB von Deimlinq A
Mawrin C

Definition
Men ingiomas comprise a family of neoplasms that are most
likely derived from the meningothelial cells of th e arachnoid
mater (CNS WHO grade 1, 2, or 3) .

ICD-0 coding
9530/0 Men ingioma

ICD-11 coding
2A01 .0Z Meningiomas , unspecified

Related terminology
None

Subtype(s)
See Box 7.01 .

Localization
Meningiomas typically arise in intracranial, intraspinal , or
orbital locations. The most common sites include the cerebral
convexities (with tumours often located parasagittally, in asso-
ciation with the falx cerebri and/or venous sinuses) , olfactory
grooves, sphenoid ridges, parasellar/suprasellar regions , optic
nerve sheath, petrous ridges , tentorium, and posterior fossa .
lntraventricular and epidural local ization is uncommon. Most
spinal meningiomas occur in the thoracic region . Tumour loca-
tion is strongly associated with the mutation spectrum: convex-
ity meningiomas and the majority of spinal meningiomas often Fig. 7.01 Meningioma. A Postcontrast T1-weighted MRI showing an extra-axial. left
carry a 22q deletion and/or NF2 mutations, whereas skull base convexity mass with homogeneous contrast enhancemen t and a dural tail sign. B Mul-
meningiomas harbour mutations in AKT1 , TRAF7, SMO, and/ tiple meningiomas. Postcontrast MRI shows several dural-based masses in the same
or PIK3CA {601 ,3451 ,3052 ,9,2777,353}. Higher-grade menin- patient, consistent with multiple meningiomas. Most such cases arise from a single
clone and are thought to represent dural spread.
giomas most commonly arise from the convexity and other

Box7.01 Subtypes of meningioma non- skull base sites . Rare primary meningiomas arise outside
the neuraxis (e.g . in the lung).
Meningothelial meningioma
Fibrous meningioma Clinical features
Transitional meningioma Meningiomas are usually slow-growing , occurring with neuro-
Psammomatous meningioma logical deficits that vary depending on tumour location . Clinical
Angiomatous meningioma signs and symptoms can arise from compression of adjacent
Microcyst1c mening1oma structures. Headaches, weakness , and seizures are common ,
Secretory mer11ng1orna although not specific for meningiomas . Higher-g rade tumours
Lymphoplasmacyte-rich mening1oma and those with molecular biomarkers of aggressive behaviour
progress more rapidly.
Metaplasttc mer 1ng1oma
Chordoid rner1ingiomc:.
Imaging
Clear cell rT\en1ngiorna
Meningiomas chmacteristica lly appear as isodense, uniformly
Rhabc!o1d rnening1orna
contrast-enhancing dural masses on MRI. Calcification is com -
Papillary menmg1oma mon and is best vis~alized on CT. A frequent imaging feature
Atypical mening1oma 1s a co ntrast-e nhancing dural tail sign at the tumour perimeter.
Anaplast1c \malignant) rnening1oma whi c h often corresponds to reactive fibrovascular tissue and
does not necessari ly predict dural involvement. Peritumour c I
cerebra l oedema can be prominent with certain hi stologica l
subtypes . such as s~cretory (3552), angiomatous/mi cro cysti c ,
lymp hoplas.m acyte -rich , and high-grade meningiomas 123 361.
c .yst formation may occu r wi th in or at the periphery of a menin -
g ~oma . ~e uro im~gi n.g features are not always specific for th e
d1agn?s 1~ of men1n g1oma or for estimating prognosis ; however,
q uant1tat1ve and qualitative imagi ng features from gadolinium -
~h anced MRI can suggest the histo logical grade of menin -
g 1omas and p red ict more-likely patient outcomes (638 ,2163) .

Spread
Meningiomas commonly invade adjacent anatomical stru c-
tures (especially the dura), although the rate and extent of local
spread ~re often ~ reater in the more aggressive subtypes. Th us,
depending. on their location and grade, some meningiomas pro - Fig. 7.02 Secretory meningioma. A small secretory meningioma on postcontrast
duce considerable patient morbidity and mortality. Extracranial T1 -weighted (A) and T2-welghted (B) MRI, showing extensive peritumoural brain
metastases (e.g . to lung , pleura, bone, and/or liver) are rare and oedema.
most often associated with CNS WHO grade 3 meningiomas. In
one series, the incidence of metastases from all meningiomas
was 0.67%, with a greater incidence in CNS WHO grade 2 (2%)
and grade 3 (9%) men ingiomas {670).
"'c0
I!!
Epidemiology GI
0.. 1.5
0
Incidence 0
0
0
Meningioma occurs in the USA at an average annual age-
adjusted rate of 8.58 cases per 100 000 population , account-
~
.
GI
0..
1.0

ing for 37.6% of CNS tumours (2344}. It is the most common .s<a
Q:
primary brain tumour in adults (estimated to occur in up to 1% ..,c
GI

I
0.5 ·
of the population) {3316} but the least common in children aged GI
"'O
(j
0-19 years. .5

Age, sex, and race distribution


The risk of mening ioma increases with age; the median age at
d iagnosis is 66 years (2344}. Across all ages, the incidence Age at Diagnosis (Years)

of g rad e 1 mening ioma is 2.32 times greater in women than in .,._ Combined ~ ~ Female .... Ma.Je

men , with the greatest risk d ifferential (3 .28) seen before meno-
pause and decreasing thereafter. The incidence is significantly Fig. 7.03 Meningioma. Incidence (number of cases per 100 000 person-years) of non-
higher in Black people than in White people (9.25 vs 7. 88 cases malignant meningioma by age at diagnosis and sex. Data are from the Central Brain
Tumor Registry of the United States (CBTRUS), 2012-2016, and include all 50 states
per 100 000 person-years) (2344] .
and Puerto Rico. Meningloma is defined by ICD-0-3 codes 9530-9534 and 9537-9539.

Etiology
Expo sure to ionizing rad iation is the primary established envi - for ~ 6 months (605 ,3435] . One study found enrichment of
ro nmental risk factor for mening ioma . The risk is higher in peo- PIK3 CA mutations in meningiomas of patients treated with pro-
ple who were exposed to ion izin g rad iation in childhood than gestln \792). A large case-control study showed that women
in those exposed in adulthood , and in peopl e exposed to high w ith meningioma were more likely than those without to report
levels of ionizing radiation, such as atomic bomb survivors and hormone-related conditions: uterine fibroids (odds ratio · 1 2,
patients treated with the rapeu tic radiation to the head. There 95% Cl : 1.0- 1.5), endometriosis (odds ratio: 1.5; 95% Cl 1.5-
is evidence that lower doses of ionizing radiation also increase 2.1), and breast cancer (odds ratio: 1.4, 95% Cl: o 8-2 3) [605) .
the risk of meningioma, including exposure to CT in child - Attempts to link specific chemicals, diet, occupation , head
hood or adolescence (2035 ,2434) . The Tinea capitis cohort trauma , and mobile phone use with rneningioma have been
study provided strong evidence of genetic susceptibility to the Inconclusive. However, allergic diseases such as asthma and
d evelopment of meningioma after exposure to ionizing radia - eczema have been fairly con '1stently associated with a reLiucecl
tion {949}. ri sk of meningioma 133801
Evidence of an association between hormones and menin - Several syndromes increase tt~e risk ot men1ng1orna develop-
g ioma risk is suggested by a number of findings, including the ment, most notably neurof 1bromntos1s type 2 ::i.nd , :..ll e ,lS'iO -
greater incidence of the disease in women tl1an in men and the ciations with 11aevo1d basal cell carc111oma sviv1rurnP 1 Gur11n
presence of hormone receptors in some rnen1ngiornas, as well syndrome) tiave been 1eporteLi Mernng1omas ha1,e alSl) ,een
as rep orts of a modestly increased risk associated with endog - reported 1n farrnil es with gerrnline defects 111 rJF 1. VHi_, PTEN.
enous/exog enous hormone use , bod y mass index, an d current PTCf-11, BAP1, SUFU, SMA RCE1, and CHEBBP \152,8/6, <.:, JL, 11,,
smoking , and a decreased risk associated witll breastteed1ng 1597,2888,2968 ,3003 ,3297,1154,1549,3319). Many of these

Menin gioma 285


Fig. !·~4 Menin~ioma . Postcontrast T1-weighted MRI. A Papillary meningioma. Occasional papillary meningiomas feature a cauliflower-like imaging appearance. B .Alyrrc ~
meningioma. An irregular interface between the atypical meningioma and adjacent brain . c Anaplastic meningioma. A centrally necrotic, contrast-enhancing tumour .v•tr rq(•e-;
mass effects.

syndromes are associated with increased radiosensitivity. Family Initiation and malignant progression of NF2-drive n mening•r:-
history studies suggest that inherited susceptibility not attribut- mas has been confirmed in genetically engineered mouse rr0 c -
able to established syndromes plays a role, with a positive family els . Inactivation of Nf2 by injection of an adenov1 r u s- encod 1 ;- ~
history associated with up to a four-fold personal risk of devel- recombinant Cre into the subdural space of mice harbouring
oping meningioma {606 ,644). Genome-wide association studies floxed Nf2 alleles (Nf2flox/flox) with arachnoid-specific ae te-
have recently detected SNPs on chromosomes 10 and 11 that tion of Nf2 results in the induction of mening iomas . proving th a t
are significantly associated with meningioma risk {764,607,828). inactivation of NF2 is an essential initial step for mernng1orra
The 10p12 SNP is located in the MLLT10 gene, a component in development (1530) . NF2 alterations are found in men ing 1ornas
several gene fusions resulting in forms of leukaemia {764). of all CNS WHO grades and thus represent an early event •n
meningioma development (1153). Progression of mening1ornas
Pathogenesis to CNS WHO grades 2 and 3 has been ach ieved in m ice o'J
Monosomy of chromosome 22 is the most frequently reported combined arachnoid-specific deletion of Nf2 along w ith Cdkn2a
genetic abnormality in meningioma, with > 50% of tumours and Cdkn2b (2489).
showing allelic losses in 22q12.2, the region encoding the In contrast, tumour initiation of non-NF2 men ingiomas has
NF2 gene. Higher-grade meningiomas exhibit more complex not been adequately modelled to date, but experimental evi -
genetic changes, with losses on 1p, 6p/q , 10q, 14q, and 18p/q, dence supports their role in oncogenesis . The mening ioma
and (less frequently) losses on 2p/q , 3p, 4p/q , 7p, and 8p/q, hotspot mutation AKT1 p.E17K , which is also found in b reas
as well as heterozygous or homozygous deletions of CDKN2A and urinary bladder cancer, leads to constitutive acrivat1on
and/or COKN28. Gains of chromosomal arms are less common of AKT1 and induces leukaemia in mice (which can not be
and mostly found in angiomatous, metaplastic, and microcystic induced by wildtype AKT1 alone) , suggesting that the AK' ~
men ing iomas. p.E17K mutation is an oncogen ic driver [472 ,690 ,199 -1) . Tr c
Genomic sequencing of two series of sporadic meningio- SMO hotspot mutations p.L412F and p.W535L are assoc.-
mas 1353,601) identified similar meningioma subsets, notable ated with increased SMO transactivat ing activity and devel-
for th eir distinct and mutually exclusive mutation distributions opment of basal cell carcinoma {1530 ,3490) . KLF4 has beer
as we ll as for their correlation with clinical behaviours and associated with context-dependent tumour suppression ill
anatomi cal locations . The first subset of meningiomas was oncogenesis 12740). and it may act as a tumour suppressor
defined by NF2 mutations and loss of chromosome 22. The in meningioma {3132) . Functionally, the KLF4 p .K409Q mu·a-
second subset lacked NF2 mutations and was characterized tion triggers the induction of HIF1a (3348}. TRAF7 interacrs
by recurrent oncogenic (p.E17K) mutations in AKT1 , as well as with MAP3K3 (MEKK3) and is involved in the regu lation Jt
alterations in TRAF? 1601}, KLF4 1601), or SMO 1353). These the TNF-a/NF-KB signal transduction pathway l 34 4) . Non-
findings have been confirmed and expanded , with oncogenic NF2 meningiomas with TRAF? mutation show upreg utanon .:-r
PIK3 CA mutat ion s al so identified 19.602,3052) . The accumu - the inhibitory immune checkpoint molecules PDL1 . 100 , ana
lation ot ad ditional copy-number losses , general genomic TOO (TD02) {1233). linking this mutation with supp ress~u
instob11ity, and emergence of TERT promoter mutatio ns was immune response i~ meningiomas . The oncogenic p ten-
largely rest rict8d o the group with NF2 mutation and/or loss t1al of PIK3CA mutations has been demonstrated in sever.J,
of ct1rornosome ?2q, wr1ereas cases with AKT1, KLF4 , SMO, tumour typ es \14031. and PIK3CA mutations activate sa\/tirJ1
PIK3CA, and/or Tf-1AF7 mutations had balanced copy-number proliferation -associated signalling pathways in men ing 1omJ"
prof1lv, \1153 601,3!13,1 15? 24881516,2390 1 YA P1 alterations 1831).. Moreover, PIK3CA mutations are convin ci ngl y 11n eu
occur in a subsr~t r.:.i1 prerJorn 11 1antly paed idtr ic meni ng1omas to ant1hormone treatm ent. Women with mening1oma wno ..:ll ::?
that do r1rJt havG NF?rnuta l 1or1s possibly r8su ltin g in ac tivation under long -term proges tin therapy carry PIK3CA mutGtlC' ''S
of ti 1t~ Hip f1U pathway \29301 more trequently than those not under hormone therapy . 1. : 1.:

28{) I /',, I 1;( I f JP Jf I 1d


high- d ose antiandrogen treatment with cyproterone leads to
an ennchment of P/K3CA -mutated skull base meningiomas
{24 87,25441 . P~LR2A mutations may drive meni ng ioma devel-
opm_e nt by altering the tran scri pti ona l mac hi nery and essential
meningeal genes (602) .

Macroscopic appearance
Meningiomas are generally sol id , globular, c ircu msc ribed
ma~es that have broad dural attachment. Some are lobulated
or b ilobed and others grow in a flat , carpet-like , en plaque pat-
tern , such ~s .those growing along the dura of the sphenoi d
bone. Menin~1omas are firm , rubbery, or (sometimes) gelati -
nous or cystic . Some men ingiomas, particularly the spinal
psammomatous subtype , can have a gritty texture due to an
abundance of psammoma bodies; others , such as the fibrous
subtyp~, can have a smooth surface . Most grade 1 meningi-
omas displace and compress the adjacent brain but are not
ad~eren~ or invasive and can be separated readily from the
b rain . H1gher-.grad~ men ingiomas, however, can be broadly
Fig. 7.05 Meningioma. This surgically resected meningioma shows the typical bos-
selated surface and dural attachment.
adherent and 1nvas1ve and may also feature areas of necrosis .
Mening iomas can also invade the dural sinuses , for exam-
p le, parasagittal meningiomas that can partly or completely Box 7.02 Criteria for assigning meningiomas to CNS WHO grades 2 and 3: these
obst~uct the superior sagittal sinus. Occasionally, meningio- criteria can be applied across all meningioma subtypes, but the CNS WHO grade 2 cri-
teria must be met for a diagnosis of atypical meningioma, and the CNS WHO grade 3
mas invade the skull and induce reactive hyperostosis of areas criteria must be met for a diagnosis of anaplas tic (malignant) men in giom~ __
such as the skull vault, the sphenoid bone, or the bones of
CNS WHO grade 2
the orbit. Men ing iomas may also attach or encase cerebral
arteries and/or cranial nerves , but they rarely infiltrate these 4 to 19 mitotic figures in 10 consecutive HPF of each 0.1 6 mm2(at least 2.5/mm 2)
structures. They may infiltrate through the cranium into the soft OR

I
tissues of the scalp and skin , and into extracranial compart- Unequivocal brain invasion (not only perivascular spread or indentation of brain
ments, such as the orbit. without pial breach)
OR
Histopathology Specific morphological subtype (chordoid or clear cell; see text)
The wi de morpho log ical spectrum of meningiomas is reflected OR
by the 15 subtypes described in Box 7.01 (p. 284) . The most
At least three of the following :
commonly encountered subtypes are meningothelial , fibrous ,
• Increased cellularity
and transition al men ing iomas. Most subtypes have a benign
clinical course and correspond to CNS WHO grade 1. How- • Small cells with high N:C ratio
ever, feature s of more agg ressive growth can arise in any of • Prominent nucleoli
these morpholog ic al patterns; in other words , the criteria defin- • Sheeting (uninterrupted patternless or sheet-like growth)
ing atypical or anap lasti c meni ngioma (see Box 7.02) should • Foci of spontaneous (non-iatrogenic) necrosis
be applied regard less of th e underlying subtype . Notably, two CNS WHO grade 3
subtypes - cho rd oid an d clear cell meningiomas - have been -
reported to have a hi gher likeli hood of recurrence than th e 20 or more mitotic figures in 10 consecutive HPF of each 0.16 mm2(at least 12.5/mm2)
average CNS WH O grade 1 meningi oma and have therefore OR
been assigned to CNS WHO grade 2, independent of the cri- Frank anaplasia (sarcoma-, carcinoma-, or melanoma-like appearance)
te ria otherwise applied for CNS WHO grade 2 atypical menin- OR
gioma ; nonetheless, larger and prospective studies would be TEAT promoter mutation
he Ipf ul to val idate these proposed CNS WHO grade 2 assign - OR
ments and to suggest additional prognostic biomarkers. In
Homozygous deletion of CDKN2A and/or CDKN2B
addition , historically, rhabdoid and papillary morphology qual -
ified for CNS WHO grade 3 irresp ective of any other indica-
tions for malignancy. Although papillary and rhabdoid features lmmunohistochemistry and pro/iteration
are often seen in combination with other aggressive features, lmmuno.h1stochem1stry can assist in esta blish in g a meningiorna
more recent studies suggest that the CN S WHO grade should d1ag~o~1s and can exclude other d iffere ntial cons1derat1 u11s
be assigned by applying the criteri a for CNS WHO grade 2 Men1ng1omas typically expres s EM A an d vimentin . However, the
atypical or CNS WHO grade 3 anapl ast1c rne ning1oma. not ~MA staining .can be faint , foca l, or even absent , particularly 111
on the basis of rhabdoid or papill ar y histolog y alone !3301] fibrou s and higher-grade subt yp · ·
Issues relating to grading meningi omas , as well as r ~co mmen ­ . . es , and v1me11 t1n pos1 t1v1ty Ila ~
~ow spe_c 1f1c.1ty. SSTR 2A 1s expressed strongly <lnd diffusely rn
dations for the use of parti c ular b1 oma rke rs. aro disc ussed 111 almost all cases, but it can also be expressed in neuroendo -
the description of each subtype be low c rin e neoplasms.

Meningioma 287
the cells are hardly appreciable with light microscopy, qivinq 1~ , r-:
impression of a syncytium , although ul trastructural i nv~ s t1 q:J .
tions have revealed that the tumour c ell s have separate delir....;:,tA
processes, demonstrating that the pattern is a pseudosyri r;y .
ium. The round to oval nuclei can have internal empty spac%
(nuclear holes) and pseudoincluslons (cytoplasmic invagina-
tions). Whorls and psammoma bodies are rare compared wi th
their occurrence in transitional , fibrous , and psammomatous
meningiomas.
The similarity to arachnoid cap cells warrants caution when
encountering small fragments that may also be compati ble w1m
meningothelial hyperplasia, which can occur in the vicinity of
other neoplasms such as optic gliomas .
Meningothelial meningiomas often harbour AKT1 p.E17K
mutations, frequently combined with TRAF7 mutations (also
Fig. 7.06 Meningothelial meningioma. Lobular growth pattern, syncytium-like appear- common in secretory meningioma), or SMO and PIK3CA muta-
ance due to poorly defined cell borders, and frequent clear nuclear holes, with occa- tions 1601,353,9,2777). AKT1, SMO, and PIK3CA mutations
sional intranuclear pseudoinclusions. are virtually absent in other subtypes. Conversely, NF2 muta-
tions are rare in meningothelial meningioma , as are deletions
lmmunohistochemistry for Ki-67 can highlight an uneven dis- of chromosomal arm 22q or other chromosomal alterations
tri bution of proliferation and guide assessment of mitotic counts. The DNA methylation profile of meningothelial meningiomas 1s
Studies suggest that cases with a proliferation index > 4% have similar to that of secretory meningioma {2783}. Meningiomas
recurrence rates similar to those of CNS WHO grade 2 (atypical) of this subtype are more common at the skull base than other
meningiomas, and that cases with an index > 20% are associ- subtypes, and the frequencies of AKT1, TRAF7, SMO, and/or
ated with mortality rates similar to those of CNS WHO grade 3 PIK3CA mutations In meningothelial meningioma at this location
(anaplastic) meningiomas. One study found that staining for the are particularly high .
mitosis marker phosphohistone H3 can stratify meningiomas
into three risk groups, defined by 0-2, 3-4, and ~ 5 labelled Fibrous meningioma
mitoses per 1000 tumour cells {2319); however, interlaboratory The fibrous subtype of meningioma has spindle cells in parallel.
differences that affect staining and interpretation limit the trans- storiform , or interlacing bundles in a collagen-rich matrix.
lation of these findings . Tumour cells form fascicles with varying amounts of intercel-
lular collagen . The collagen deposition can be extensive and
Meningothelial meningioma suggest the differential diagnosis of solitary fibrous tumour, but
In the meningothelial subtype of meningioma, epithelioid cells only solitary fibrous tumour shows nuclear staining for STAT6 .
form syncytia-like lobules, with some nuclei appearing to have EMA expression can be weak or absent, whereas 8100 staining
nuclear holes and pseudoinclusions. can be surprisingly strong. In contrast to that seen in schwan-
Meningothelial meningioma ce lls resemble the morphology of noma, though, SSTR2A expression is often strong and diffuse
arachnoid cap cells . They are largely monomorphic, with abun- in fibrous meningioma.
dant eosinophilic cytoplasm, and are arranged in lobules that Fibrous meningiomas typically show 22q deletion and
can be demarcated by fine collagen septa. Borders between mutation of the retained NF2 allele, similar to transitional and

. . , .·.
. .. "' '. .
·... :
.. ·
..
"'
./,;-'
~~ ..":
.· '
~'. "' .·, . .-. .- ,,,. ....- .~
/' , . •• .. 1~' , . - f . ' '\,.. '
A B ,, .- ~ . ' "
Fig. 7.07 Meningothelial hyperp1a.,ia A An optic nerve ghoma surrounded by meningothelial hyperpla~i~. On biopsy, th is ~~y be mista:en r . . - ~ . · •.' "'
1
1
smaller size. rnult1centnc11y, and lacK of dural invas1or1. B EMA immunostain1ng highlights the meningo1helial hyperplasia s d' h' o mernng1om.a, but it differs in its
urroun ing t is paraspinal neuroflbroma.
288 fVh_l 1 f \JIU 1 ' Id
psammomatous meningiomas. Their DNA methylation features
overlap those of transitional and psammomatous meningiomas .
They are frequently found at the convexity.

Transitional meningioma
The transitional subtype of meningioma contains meningothelial
and fibrous patterns as well as transitional features .
Lobular and fascicular areas appear side by side, with some
areas not clearly attributable to one or the other of the two pat-
terns (hence "transitional "). Whorl formation and psammoma
bodies are frequent in this subtype. Transitional meningiomas
share with fibrous and psammomatous meningiomas the fea-
tures of frequent 22q deletions and NF2 mutations, and they
have similar DNA methylation characteristics . They often arise
from th e convexity. Fig. 7.09 Transitional meningioma. This subtype contains numerous whorls.

Psammomatous meningioma lmmunohistochemistry for EMA or SSTR2A . Non-calcified foci


In the psammomatous subtype of meningloma, psammoma typically conform to the fibrous or transitional subtypes. Psam-
bodies predominate over the viable tumour cells. momatous meningiomas share molecular features with fibrous
The overlap of individual psammoma bodies can result in and transitional meningiomas, particularly 22q deletions, NF2
large, confluent, calcified masses . Actual meningioma cell s mutations, and epigenetic profiles . This subtype often occurs
can be rare to vi rtuall y absent, but they can be highli ghted by in the thoracic spine region of middle-aged to elderly women .

- d
Fig. 7.10 Psammomatous mening1oma. A Almo t completo replacement of the mening1oma by psammomatous calc1ftcat1ons (postdecalclt~cation specimen) .. 8 EMA imm~no­
sta1ning reveals mening1oma cells between psarnmoma bodies

Menmgioma 289 I
I .ii
( ~
. }' -~
1i"
-., ·~
;
I '
.
'

Fig. 7.12 Microcystic rneningiorna. A Cobweb-like background with numerous delicate processes. B Thin, wispy processes are evident on EMA irnrnunostaining.

Angiomatous meningioma meningiomas , with which microcystic areas can be combined


In the angiomatous subtype of meningioma, often-hyalinlzed (1599) . Cerebral oedema is frequent, as it is in angiomatous and
small blood vessels predominate over the intermixed meningi- secretory meningiomas.
oma cells .
Between the numerous vessels, the actual tumour cells may Secretory meningioma
be hard to find and to identify as meningioma cells. Blood ves- The secretory subtype of meningioma is characterized by foci
sels can be thin- or thick-walled, and variably hyalinized. Angi- of gland-like epithelial differentiation with PAS-positive eos1no-
omatous areas can also be intermixed with microcystic or even philic secretions and/or combined KLF4 and TRAF7 mutations.
metaplastic areas, and , like in these subtypes, the cells can The eosinophilic secretions (pseudopsammoma bodies)
show degenerative nuclear atypia. Hypervascular examples are positive for a variety of epithelial and secretory markers
may mimic haemangioblastoma but are typically inhibin-nega- including CEA. The surround ing cells can also be positive for
tive and SSTR2A-positive. CEA and cytokeratins . Elevated CEA levels in the blood can
Angiomatous , microcystic , and metaplastic meningiomas all occasionally be observed {1936). which drop with resection
have a high frequency of chromosome 5 gain (10) . and rise in the rare cases of recurrence. Peritumoural oedema
Like secretory and microcystic meningiomas, angiomatous is common.
meningiomas are often associated with cerebral oedema Combined KLF4 p.K409Q and TRAF7 (distributed across the
beyond that expected for the tumour size. WD40 domain) mutations genetically characterize this subtype
(601 ,2656). In a few instances, the KLF4 mutations may be iso-
M1crocystic menmgioma lated .
Thi:: rn1cmcystic subtype of meningioma has microcysts formed
by cell~ with thin elongated processes, creating a cobweb- like Lymphoplasmacyte-rich meningioma
backg1 uund on histology. Lymphoplasmacyte-rich meningioma is a rare subtype 1n which
The C/sts can e1pand to macroscopically or radiologicall y extensive chronic inflammatory infiltrates predominate over the
detE:ctabl8 m8crocyst~,. t 1ke i11 angiornatous meningioma , the meningothelial component .
prese1 ice of dE:geriernt1ve nuclear atypia in microcystic menin- Despite the name, plasma cells may be scant, and macro-
g1orna can raise the susp1c1on of a higher grade However, phages often predominate (1793) . In some cases. 1t ma be
m1crocyst1c menirig1ornas are typically be1ugn Gain of cl1ro- chall enging to distinguish this subtype from inflammatory disor-
rnosome ti 1s common, as 1t is 1n ang1ornatous and metaplastic ders with patct1y meningothelial hyperplasia .
290 lvh-011.11q1u111a
Fig. 7.14 Lymphoplasmacyte-rich meningioma. Marked tumour-associated chronic Fig. 7.15 Lipoma-like metaplastic meningioma. Lip1dized mening1oma cells resemble
I
inflammation. lipoma.

Metaplastic meningioma less often spindled). variably vacuolated cells embedded in a


The metaplastic subtype of meningioma has focal or extensive mucin-rich matrix.
mesenchymal components, includin g osseou s, cartil agi nous, Chordoid areas are of ten interspersed with more typical
lipomatous, myxoid. and xan thomatous tissue, either sing ly or meningioma; however, pure examples are also encountered.
in combinations . Chronic inflammatory infiltrates are often patchy when present,
These alterations have no known clinical relevance, and most but they may be prominent Chordoid meningiomas are char-
do not constitute true metaplasia (e.g. a lipomatous appearance acteristically large. supratentorial tumours , and the patients
as a result of lipid accumulation rather than true lipomatous may be young er, with an average age at presentation of about
metaplasia) . The morphological features of metaplastic menin- 45 years !5901. Chordo1d men1ng1omas often lack any other
gioma can overlap with those of ang1omatous and microcystic high -grade h1stopath ological features, but they have recur-
meningiomas, and gain of chromosome 5 is frequent in all three rence rates analogous to those of atypical mening1omas and
subtypes {2783) . have therefore been designated CNS WHO grade 2 [642.
Ossification in metaplastic meningioma can be hard to dis- 2934) . One study reported frequent ep1thel1al d1ff erentiat1on
tinguish from dystrophic ossification of psammoma bodies in wit11 NHERF1 -irnmunoreact1ve cytoplasmic m1crolum1na , similar
psammomatous meningioma, or from bone invasion . Remnants to that in secretory mening1omas ! 1059) Rarely, patients have
of the concentric inner structure of psammoma bodies or radio - associated haematological conditions. such as anaemia or
graphic imaging of adjacent bone, respectively, may assist in Castleman disease I 1590). Ct1romosome 2p deletions are over-
different1at1on . r·epresented . but DNA methylation profiles overlap with those of
other mening1oma subtypes 12934).
Chordoid meningioma
The chordo1d subtype of mening1oma predorrnnan tl y resembles Clear cell meningioma
chordoma. featuring cords or trabeculae of small. epithelio1d (or The clear cells btype of menin g1oma has a pre:...:::.
ternless or sheeting architecture containing round t
.,,. . .,
- -.......__ - - . . . B. - . -
Fig. 7.16 Chordoid meningloma. A Cords of small eplthelioid to vacuolated tumour cells embedded in a mucin-rich matrix. B Alcian blue highlights the mucin-rich matm:.

Fig. 7.17 Clear cell meningioma. A Sheets of rounded clear cells with block-like perivascular and interstitial collagenization . B Trichrome highlights the extensive collag&n
deposits, including larger coalescent forms. C Abundant intracytoplasmic glycogen is noted on PAS histochemistry. D SSTR2A-immunoreactive tumour cells.

cells witt1 clear. glycogen-rich cytop lasm and prominent angle and spine, especially the cauda equ1na region . It a.lsu
perivascular and interstitial collagen . tends to affect younger patients, including children and young
The per1vascula1 an d 1nterst1tial co llagen occasionally coa- adults (mean age in one series: 24 years) (3593). Clear ceil
lesces 1rito large acellular zones of collagen or forms brightly meningiomas are associated with more aggressive behaviour
eos1nopr1ilic. arn1ar11hoid-l1k collagen It shows prominent PAS- including recurrence and occasional cerebrospinal fluid seed -
pos1t1ve ai 1cl dict~,tas•-: sc~ns1t1ve cy toplasmic glycogen Whorl ing, and have therefore been designated CNS WHO grade 2
forrmnion is 1C1gu13 J11 :J p;,amrnorna bodies are inconspicuous . pending larger studies to confirm the higher rates of recur-
Clear cell rr,er11r1LJ1<>rfii.:1 t1cts a procl ivi ty for t11e ce rebellopontine rence {36201 Both germline (familial examples) and somatic

292 lv11•1 ''1(1 llll •


SMARCE1 mutations are common, with virtually all cases show- PBRM1 is predominantly mutant or deleted in papillary meningi-
ing loss of nuclear SMARCE1 expression by immunohistochem- oma , but it is also altered in single rhabdoid mening1oma cases
istry {1061,3148 ,575). {3449) . Similarly, BAP1 mutations or deletions, typically found
in rhabdoid meningiomas , have also been reported in papillary
Papillary meningioma meningiomas or rhabdoid meningiomas with partly papillary
The papillary subtype of meningioma is defined by the pres- features , which are then usually combined with alterations of
ence of a predominant perivascular pseudopapillary pattern . PBRM1 {3449,2888} .

I
In the papillary subtype, meningioma tumour cells surround
thin-walled blood vessels in a perivascular, pseudorosette-like Rhabdoid meningioma
pattern (i.e. a perivascular nucleus-free region). Some meningi- The rhabdoid subtype of meningioma is defined by the pres-
omas have cells with rhabdoid cytomorphology arranged in a ence of rhabdoid cells , which are plump cells with eccentric
papillary architecture, consistent with a molecular and genetic nuclei , open chromatin, macronucleoli, and prominent eosmo-
link between the papillary and rhabdoid subtypes {3485,3301 , philic paranuclear inclusions appearing either as discernible ·•·.
2888 ,3449) . Papillary meningiomas have been reported in chil- whorled fibrils or compact and waxy spheres.
dren {1957} and adults {1864 ,3592}. These tumours are com- Rhabdoid features are usually present at primary resection
monly associated with peritumoural oedema and bone hyper- but can become increasingly evident upon recurrence . Most
ostosis or destruction ; cyst formation can be seen {1910,1864, rhabdoid meningiomas are highly proliferative and have other
3555}. A papillary growth pattern has been associated with brain histological features of malignancy. Original cohorts of rhab-
invasion and aggressive clinical behaviour including dissemina- doid meningiomas comprised tumours with high rates of recur-
tion and metastasis, predominantly to the lung {2410,1957,2593, rence and death {1592,2474). supporting the designation as a
3485,1864). Focal papillary architecture, in the absence of any CNS WHO grade 3 malignancy. Most of the tumours in those
other features of higher grades, does not suffice for designating cohorts otherwise fulfilled the criteria for classif ication as CNS
tumours as CNS WHO grade 2 or 3. WHO grade 3 anaplastic/malignant meningioma, irrespective
Some meningiomas have cells with rhabdoid cytomorphology of rhabdoid cytology. A large portion of rhabdoid meningiomas,
arranged in a papillary architecture {3485,3301 ). Consistent with however, have since been diagnosed on the basis of rhabdoid
this occasionally observed morphological overlap, papillary cells alone, not fulfilling other criteria for CNS WHO grade 3 clas-
and rhabdoid meningiomas have the same genetic alterations: sification; of those, 50% have CNS WHO grade 1 features and

,
~ ..
.. ..,. . '·,,. •, ' : .... 'i!.~ .
, • ~'(~ ~ • ,·'>!.'.;.\I; . :: : •
. ' • .;, ""; .
'.-

'
: & # ., , •

... •• ~· ,· ~ti .. l·~ . : .


•• ,~.l.. IJ • ,('-
.... "'-'
I

•• ~
,, ..,; I •
........
''* fl"
t ..

," . •' ., ' ,.


J '

:
~·~ ,
~

..·,.: • .J. •.•. '.~ .••


~" ~ ~ ~ ~
·.
.
• I), ;.

I ~J :t· I
'i ~of • It
I
r
!I I ··~
.· - •_...
t "

• I',_}• ',
, ,. ' •, .
• ; ,,,• : . ,. 4 ..

fig 719 Rhabdoid meningioma A Eccentr1cally roe ate vesicu lar nuclei, prominent nucleoli, and eosrnophiltc globular/flbrillar paranuclear 1 ' .
• • · inc usrons. 1 Loss or nu tear l3 ri 1
immunoreact1vity, which has been associated w11h a rnoro aggressive biology.

Mernngioma 293
, .
I ~ .. I • ' ,I • • ~

.. .. '
.. . . .
'-
; ' ,• I .. I ,"'
.· J
~
'. /}
\

'•
" • •••• t ,· . ~ -
\ ,,. ,_I ,.· - : I ,· t • \ ... ::-- ~ •
• . ._ ~ <.\ • . D, :·. .. ',_.; . ·...., .. .
Fig. 7.20 Atypical meningioma, A Sheeting architecture and small cell formation. B Small cells with high N:C ratio. C Macronucleol i. D Increased mitotic activity. Note
the lack of nuclear atypia, despite the name "atypical meningioma". E Loss of nuclear H3 p.K28me3 (K27me3) immunostaining, which has been associated with a worse
prognosis.

50% have CNS WHO grade 2 features 13301 ). A meta-analysis papillary meningiomas arise in patients with germline mutations
showed that patient outcome is strongly correlated with CNS in the BAP1 gene as part of the BAP1 tumour predisposition syn-
WHO grade , independent of the rhabdoid features ; this work drome, in which family members may develop uveal and cutane-
suggests that rhabdoid meningiomas should be graded similarly ous melanoma, mesothelioma, and renal cell carcinoma, among
to non-rhabdoid meningiomas, but the authors cautioned that other tumours. Importantly, in this context, immunohistochem1cal
some of these tumours may still behave aggressively and that loss of BAP1 expression was associated with aggressive (consis-
close patient follow-up is required 13301 ). Some meningiomas tent with CNS WHO grade 3) clinical behaviour in these tumours
have cells with rhabdoid cytomorphology arranged in a papil- 12888). In addition, as discussed above under Papillary mening1-
lary architecture, suggesting a relationship between these two oma, there may be overlap between the histological and genetic
subtypes 13485,3301,2888,3449). A subset of rhabdoid and/or features of rhabdoid and papillary meningiomas .

. .,,_
• )
~ ., . ..
I

Atypical mernng10mci A Bra111 invasion through the pia. with irregular tongue-like protrusions into adjacent brain parenchyma a Brain invasion highltghteo' by "n·
trapped GFAP-pos1!1ve 1slarids ot gl10!1c brain parenchyma at the tumour periphery. , "

294 [Ji· · 1111l"l ' 1


.. . . . .
,..·:-·.,........
... .
~.,. , .., . .: ..:. .. . : :.

-~
·":..

- . -. ..•.- . . ...
--:. ·.~ -,,,.. . . ... ..... ,

Atypical meningioma
Atypical mening ioma is an intermediate-grade meningioma with
increased mitotic activity, brain invasion , and/or at least three of
WHO grade 2 meningiomas in some , but not all , studies 12473,
220 ,272/ . Larger series with longer follow-up times may be
needed to resolve this issue . Atypical mening iomas can be
I
.

the following : high cellularity, small cells with a high N:C ratio , further risk stratified based on the Inclusion of various ad di-
prominent nucleoli , sheeting (uninterrupted patternless or sheet- tional clinicopathological and genetic factors (773 ,2782,2783 ,
like growth) , and foci of spontaneous (non-iatrogenic) necrosis. 547,1569,405,206,936 ,1018}. However, some geneti c c han ges
Increased mitotic activity was defined in one large clin- (e.g . TERT promoter mutation or homozygous CDKN2A and/
icopathological series as ~ 2.5 mitoses/mm 2 (equating to or CDKN2B deletion) are evidence for d iagnosin g CNS WH O
~ 4 mitoses per/10 HPF of 0.16 mm 2 , as originally described) grade 3 meningioma (see below) , so cons ideration sh ould be
l 2475) . Despite the name "atypical meningioma ", nuclear atypia given to TERT, CDKN2A , and CDKN2B analysis in clinically
is not a usefu l criterion , as it is often considered degenerative aggressive atypical men ing iom as or those with borderlin e
in nature and not associated with patient outcome . Clinical risk CNS WHO grade 2/3 features
factors for atypical meningioma include male sex , non- skull
base location , and prior surgery [1539} . Atypical meningiomas Anap /astic (malignant) meningioma
have been associated with high recurren ce rates despite Anaplastic (malignant) meningioma is a high-grade meningi -
gross tota l resect ion j30), and bone involvement may be asso - oma with overtly malig nant cy tomorphology (anaplasia) that can
c iated with a further increa se in recurrence ri sk [1018) . Brain (1) resemble carcinoma , hi gh -grade sarcoma, or melanoma,
invasion by mening ioma is c harac terized by irregul ar, tongue - (2) display markedly elevated mitotic activity, (3) harbour a TER T
like protrusion s of tu mour ce ll s into underl yin g GFAP-pos itive promoter mutati on ; and/or (4) have a homozygou s COKN2A
parenchyma , without intervening leptomeninges . Extension and/or COKN2B deletion
along perivasc ular Virchow- Robin spaces does not constitute A ~i to tic cou nt of 2 12.5 mitoses/mm ' (equating to
brain invas ion because the pia 1s not breached . Such pe ri vas - ~ 20 mitoses/10 HPF of 0 16 mm 2 , as originally descri bed) wcis
cular spread and hyal1 nization 1s most commonly encoun - used to define markedly elevated m1tot1c activity 1n a st udy
' tered in children an d can mimic meningi oangiomatosis 11087, of 11 6 patients [2473) . Anaplast1 c rn en1ng1ornas account fci r
2469) . Brain invasion occurs most often in mening1om as wi th 1.- 3% ot meningiomas. Most of 1hese tumour s displ dy cx tt:n -
additional high -grade features . Nonet11eless. tl18 presen ce of s 1~e necro sis and c~ n invade br;:un In some c.Hl _-lp las trc Cd . L''•
brain invasion in clinica ll y totally re sec ted , otherwi se benign - rn eningothelral origin ca n be con f1rmed usin~J irnn iLmot :is tu
appearing meningiom as remains controversial, as 1t ha s been che-~11 s try 12070 ,791) or genetic tes ting j2783 ,266, 2j ::JlJ, .2 > 1.1,
f associated with re currence rates s11rn lar to those of other CNS Because malignant progression in meningiomas is a continuum

Mernng1oma 295
Fig. 7.23 Meningioma. A lntraoperative touch preparation of meningioma. Whorls and small psammoma bodies can be appreciated. B Secretory meningioma Pseudopsam-
moma bodies are evident on intraoperative smear.

of increasing anaplasia , determining the cut-off point between Other histopathological patterns
atypical and anaplastic meningioma can be challenging . Inter- The large number of subtypes covered above already illustrates
observer reproducibil ity is good for mitotic count but only fair the wide morpholog ical spectrum of mening iomas . However.
for overt anaplasia {1048} . The presence of a TERT promoter meningiomas can have a variety of morpholog ica l c haracteris-
mutation confers a high risk of recurrence and short interval to tics that even exceed those of the established subtypes . These
progression , irrespective of other histological features (1152, include meningiomas with oncocytic, mucinous , scleros1ng.
2782,1517}. Similarly, homozygous deletion of CDKN2A and/or whorling-sclerosing , GFAP-expressing , and granulofil amen-
COKN28 is associated with high-grade histopathology, elevated tous inclusion-bearing features , or the occurrence of menin-
risk of recurrence , and shorter time to progression {331 ,2939, gothelial rosettes (2717,1025,1 207,1342,250 ,69}. These patterns
2465,1724,2931}. Loss of H3 p.K28me3 (K27me3) is observed are rare, and the data on biological and clinical correlations are
in about 10-20% of anaplastic meningiomas and is associated too scarce to identify any relevant implications.
with shorter overall survival {1569 ,1048}.
Cytology
On intraoperative smear and touch preparations , characteris-
Aracluloldal cap cell (PGDS"') tic cytological features of meningioma are often apparent. with
oval , euchromatic nuclei (sometimes with intranuclear pseu-
doinclusions) and delicate cytoplasm visible. Whorls may be
____G_ra_de_ i _ _ _ _,I -+ Gradel I- prominent on touch preparations . Adequate smears may be
NF2 difficult in meningiomas with more copious collagen .
SMARCEr j
POI.RU
;
.s ~ Diagnostic molecular pathology
P/KJCA.

i0 ~ Jll.F4
Genetic changes (e.g. in AKT1, SMO, PIK3CA) are strongly
=
Q TIUF7
::e e SMO 11 TERTpromoler related to the subtypes of meningioma , but do not define th em .
A.KTJ The status of most genetic alterations immediately relevant to
subtyping and grading (including TERT promoter, SMARCE 1.
Loss : 22q KLF4 and TRAF7, and other alterations) can be assessed bv
Loss: lp I 6q / 10 / l4q / 18q
DNA sequencing . Because TERT mutations can arise during
~q / 9q I Uq / 15q / l 7q / 20q
progression , selection of tissue for DNA extraction should focus
Lo~
on the most malignant-appearing and proliferative reg ions.
TSLCI t PDGFR t IGFR j Homozygous deletion of CDKN2A and/or CDKN28 can be
assessed by in situ hybridization or calculated from vari ous
\'EGF t IGF high-throughput sequencing or hybridization assays; however.
CDKN2A/2B t
FISH probes are large, so small deletions can sometimes be
missed by this technique . Rare events such as TERT activation
lon:KT t
by gene fusion or such as gene fusions involving YAP 1 may
Fig. 7.24 Meningioma. Schematic showing the distribution and evolution of genomic in some cases be inferred from high-resolution copy-number
and expression characteristics in meningioma grades. Mutations are listed in the grey plots, but they can typically only be proved by RNA sequenc-
bars. with light grey indicating mutations occurring in meningiomas without NF2 altera- ing or in situ hybridization . BAP1 and PBRM1 can be aHecteo
tions. Cytogenet1c alterations are listed in the blue bars, and gene expression changes
by both mutation and deletion, thus requiring DNA sequencing
in green. Bar length indicates the relative frequency of an alteration within the given
iumour grade. 'SMARCE1 mutations have been found nea rly exclusively in clear cell and, if not already provided within the sequencing approacr1 ,
meningiomas. PGDS', prostaglandin 02 symhase-pos1t1ve precursor cell s in murine independent copy-number assessment. Alternatively to ONA-
mernngioma models. based methods, surrogate immunohistochemical stains can be

296 f\,l I I !_J •.ll I


used to detect some genetic alterations , including SMARCE1 Box7.03 Diagnostic criteria for men1ngloma: diagnosis frequently requires matching
loss (clear cell n:ieningiom~) : B~P1 loss (rhabdoid mening ioma), of several criteria
or posttranslat1on~I mod1f1cat1ons including H3 p.K28me3
Essential:
(K2~m~3) status (tnmethylation is lost in a subset of aggressive Classic histopathological features matching at least one of the meningioma
mening1omas).
subtypes
Met_hylo_m e profiling can provide information about tumour
OR
type i_n histological!~ challenging cases and define the epi-
Suggestive hlstopathological features combined wtth biallellc inactivation of NF2
genetic ~ubgroup; in addition, copy-number alterations are or other classic drivers of conventional meningioma (TRAF7, AKT1. KLF4, SMO,
reported in parallel with the DNA methylation results . PIK3CA) , clear cell meningioma (SMARCE1), or rhabdold meningloma (BAP1 )
OR
Essential and desirable diagnostic criteria
See Box 7.03. Suggestive histopathological features combined with one of the defined DNA
methylation classes of meningioma
Staging
Desirable:
Brain invasion is a criterion for the diagnosis of CNS WHO
Meningeal localization
grade 2 meningioma, and bone invasion has been associated
EMA immunoreactivity
with worse prognosis in atypical meningioma (1018).
Strong and diffuse SSTR2A immunoreactivity
Prognosis and prediction Classic copy-number alteratlons ot NF2-mutant meningioma, such as monosomy
22/22q in lower-grade menlngiomas, with additional losses of 1p, 6, 1Oq, 14q, and/
The major prognostic questions regarding meningiomas involve
or 18 in higher-grade meningiomas
estimates of recurrence, progression-free survival , and overall
survival.
Molecular features
Clinical factors A number of molecular features have prognostic significance
A major clinical predictor of recurrence and overall survival is in meningiomas. Higher-grade meningiomas are associated
the extent of resection {45}. which is influenced by the tumour with more complex copy-number changes and chromosomal
site, extent of invasion, attachment to critical intracranial struc- abnormalities (266,353 ,1851). DNA methylation patterns sepa-
tures , and availability of expert neurosurgical services . In most rate subgroups of meningiomas, including those with higher risk

I
cases, meningiomas can be removed entirely, as assessed of recurrence (1569,2390,2783). Meningiomas that have TERT
by operative or neuroradiological criteria; however, recurrence promoter mutations have a higher rate of malignant transforma- .
can occur even after complete resection . In one series, 20% tion, a shorter time to recurrence , and a lower overall survival rate
of gross totally resected benign meningiomas recurred within than those without (1152,2782,1517). In a meta-analysis com-
20 years {1429) . Rates of recurrence are significantly higher prising 677 patients, the median overall survival was 58 months

I
in CNS WHO grade 2 and 3 meningiomas than in CNS WHO in patients with meningiomas harbouring TERT mutations ver-
grade 1 meningiomas (1430); mortality rates are also higher, sus 160 months in the TERT-wildtype group (2124} . lntragenic
and especially so in patients with CNS WHO grade 3 tumours. deletions in the dystrophin-encoding and muscular dystrophy-
associated OMO gene are common in progressive/high-grade
Histopathology and grading meningiomas and are associated with shorter overall survival
Overall , CNS WHO grade is the most useful histopathologi- (1516). A subset of mening iomas with rhabdoid features have
cal predictor of recurrence , and (as mentioned above) some inactivation of BAP1 and a shorter time to recurrence than other
histological subtypes of meningioma are more likely to recur. meningiomas (2888}. In papillary meningiomas , mutations in
CNS WHO grade 1 meningiomas have recurrence rates of the chromatin modifier PBRM1 are enriched , suggesting that
about 7- 25% , whereas CNS WHO grade 2 meningiomas recur such mutations may be linked with aggressive tumou r behav-
in 29- 52% of cases and CNS WHO grade 3 meningiomas in iour 13449). Alterations in CDKN2A and/or COKN2B (which are
50- 94% . Even among CNS WHO grade 1 menlngiomas , how- cell -cyc le regulator genes) are frequently found in recurrent and
ever. the presence of some atypical features increases the risk progressive meningiomas and are associated with a poor prog-
of subsequent progression/recurrence 12009}. Malignant histo- nosis 11202,1153.2465\ .
logical features are associated with shorter survival times [45 , Several potentially clinically actionable mutations have been
622,2473] . Anaplastic meningioma is often fatal, with median described in meningiomas . including mutations in SMO, AK T1 .
survival times ranging from < 2 years to > 5 years , depend- and PIK3CA 1353,9,601 ,2777]. for which targeted therapies
ing on the extent of re section and the use of radiation therapy have sh?wn efficacy in other tumour types Furthermore, POL 1
12473,3064,1045,2334}. In one study, which found a median (wh1cr1 is. associated with response to immune checkpoint
overall survival of 2.6 years and a 5-year survival rate of 10%, blockade 1n other cancers) may be overexpressed in high-grade
de novo anaplastic menin g1omas had a better ou lcorne than mening1omas \ 790) Efficacy of immune checkpoint blockade
did secondary anaplastic meningiomas 12488). Patients with has been described 1n rare rnen1ng1omas with high tumour
meningiomas that show high mitotic coun ts have significantly mutation burdens due to the 1nact1vation of components of the
shorter overall survival than patients with rneningioma s showing mismatch repair apparatus {806AI. Ongoing precision rned1c1ne
overt anaplasia without a high mitotic cou nt , and those tumours trials for mening1omas will help us under-t3nd the 1mporrance of
are associated with significantly lower patien t survival rates than these alterations for predicting response to therapy
atypical meningiomas \24 88.1048).

Meningioma ~9
Ng HK
Mesenchymal, non-meningothelial
tumours involving the CNS: Introduction

The terminology and histological features of benign and malig- common in the CNS compared with other tissues Som8 r:c(").
~ant mesenchymal , non -meningothelial tumours originating mon soft tissue tumours that can exceptionally be found 1n ·r.~
in the CNS correspond to those of their soft tissue and bone CNS (e.g. leiomyoma , fibrosarcoma) and that we:e covered r
counterparts , and we have attempted to harmonize the terminol- previous editions are no longer included 1n t~1s ed1t1on beca•j<>e
ogy and diagnostic criteria presented in this classification with their histological and diagnostic features are 1dent1cal to those 0f
those in the Soft tissue and bone tumours volume of this series. their soft tissue counterparts . Please refer to the fifth-ed1t1on Ser
Mes~nchymal tumours arise more commonly in the meninges tissue and bone tumours volume of this series for these ent1i es
than in the CNS parenchyma or choroid plexus. In general, any {3426] . New lesions that have been added are intracranral rnes-
mesenchymal tumour may arise within or have an effect on the enchymal tumour, FET: :CREB ·fusion-positive ; CIC-rearranged
nervous system , but primary mesenchymal CNS tumours are sarcoma ; and primary intracranial sarcoma , DICER1 -mutar•
very rare . They can occur in patients of any age, and they arise Tumours of the peripheral nerves are covered in Chapter 6
more commonly in supratentorial locations than in infratentorial Cranial and paraspinal nerve tumours. Antiquated nosolog1cc1
or spinal locations. The clinical symptoms and neuroradiological terms, such as "spindle cell sarcoma", "pleomorphic sarcoma·
appearance of most tumours are nonspecific. Please refer to the "myxosarcoma", and "haemangiopericytoma", are discouragea
relevant sections in this chapter for details on individual lesions. Two relatively common vascular lesions in the CNS , artenove-
This chapter covers only those entibes that have special his- nous malformation and cavernous haemangioma, are covered
tological or molecular features, occur uniquely in the CNS, or in the section on haemangiomas because there was debate as
(although similar to their soft tissue counterparts) are re latively to whether arteriovenous malformation is truly neoplastic.

3CJCJ r .~. _.. , 1 ... , r ,1 1f


Solitary fibrous tumour Giannini C Fritchie K.J
Macagno N
Bouvier C
Dem icco EG Perry A
F1garella- Brang er D

Definition average annual age-adjusted inc idence rate of 0.12 cases per
Solitary f! br?us t~ mour (SFT) is a fibroblastic neoplasm with 100 000 population (2344) . Data from large series su ggest that
a genomic 1nvers1on at the 12q13 locus, leading to NAB2 and SFTs constitute < 1% of all CNS tumours (672 ,2066 ,2833) .
STAT6 gene fusion as well as STAT6 nuclear expre ssion .
Age and sex distribution . .
ICD-0 coding In two recently pub lished series comprising 265 pati ents .
8815/1 Solitary fibrous tumour peak incidence oc c urred between the fifth and the seventh
dec ades of life , with 18% of cases occurring in pati ents aged
ICD-11 coding < 40 years . The sex distrib ution was nearly equal (M:F _ratio·
2F7C & XH7E62 Neoplasms of uncertain behaviour of connec- 1.08 :1) {1966,988) . Primary C NS SFT has been reported 1n the
tive or other soft tissue & Solitary fibrous tumour, NOS paed iatric po pulation , but it is exceed ingly rare {2055 ,3193,
2BSY & XH1 HP3 Other specified malignant mesenchymal neo- 3530 ).
plasms & Solitary fibrous tumour, malignant
Etiology
Related terminology Genetic susceptibility
Not recommended: solitary fibrous tumour / haemangiopericy- There is no evidence of fam ilial c lustering of meningeal SFT.
toma; haemangiopericytoma.
Pathogenesis
Subtype(s) The histogenesis of CN S SFT remains a matter of debate. Its
None fibroblastic nature and the presence of a common NAB2:: STAT6
gene fusion {577,2688 ,2870} are stron g arguments for grouping
Localization CNS SFT with its pl eural and soft ti ss ue counterparts ; nevert he-
Most SFTs are dural based (often supratentorial), and about less, the precise cell of orig in has not been determined .
10% are sp inal. Skull base, parasagittal , and falcine locations The genetic hallmark of SFT at all anatomical sites is a para-
are especially common {2066 ,2833). Uncommon locations centri c inversion involving chro moso me 12q13, resulting in the

I
include the cerebellopontine angle (3138), pineal gland [3595), fus ion of the NA B2and STAT6ge nes (577,2688) . Demonstration I
and sellar reg ion (1508). II
I
I
Clinical features I

In most cases , the symptoms and signs are consistent with the
localization , mass effect, and increased intracranial pressure
due to tumour size (1125,2066). Massive intracranial haemor-
rhage (2027) and hypoglycaemia from tumours that release
insulin-like growth factor (2983) are rare complications.

Imaging
Plain CT images show so li tary, irregularly contoured masses
without calcificati ons or hyperostosis of the adjacent skull. On
MRI , the tumours are isointense on T1 -weighted images , show
high or mixed intensity on T2-weighted images, and have va ri-
able contrast enhancement. Dural contrast enhancement at
the periphery of the lesion (dural tail) and flow voids may be
observed (3384) . At present, no specific features on CT or MRI
can be used to distinguish SFT from meningiomas 1544) .

Epidemiology
The true incid ence and prevalence of this entity are difficult to
ascertain because of its inconsistent nomenclature . In the 2019 Flg.8.01 Solitary fibrous tumour (SFT ) A This la19e ,v1d multiloculated falcine tu-
statistical report published by the Central Brain Tumor Regis - mour in a 74 -year-old man shows diffuse pc:;rgadul1n1urn e1•n 2 n._emenr a This dural-
based posterior Iossa tumour 1n a 73-year ol d ri1an 1::; stro'lgly and dif fusely enhanc-
try of the United States (CBTRUS) , SFT is grouped with other
ing, and 1t has a small dural iail C A dural tail 110 mm 111 len gt11) 15 vis!b!e ,11 this
mesenchymal tumours of the mening es because of its rarity ; tum~ur _
of the upper lhorac1c spine 1n a 45 -year-old man. On imaging, SFTs may mirn,
as a group, mesenchymal tumours of the meninges have an mernng1oma.

M senchyrmd , no11 -mernngothel1al tumours involving the CNS 301


C;._ ~
Fig. 8.02 Solitary fibrous tumour. A The typical patternless architecture is shown. B This tumour is composed of cells with bland spindle-shaped nuclei and scant eosmophllic
cytoplasm immersed in a collagenous background. C There is often stromal and perivascular hyaline collagen deposition. D This tumour shows keloidal or arrnanthoid collagen.

of NAB2::STAT6 gene fusion is virtually pathognomonic of SFT. ovoid cells with little or no intervening stroma and less conspic-
Like their counterparts at other sites , CNS SFTs with fusions uous vasculature , albeit often interspersed with pale zones or
between NAB2 exon 5, 6, or 7 and STAT6 exon 16 or 17 tend foci of necrosis . Nuclei are monotonous and round to oval , and
to have a more cellular and more mitotically active phenotype they lack the pseudoinclusions typical of meningioma. Invasion
(corresponding to a higher grade) than do CNS SFTs with of brain parenchyma or engulfment of vessels or nerves may
fusions between NAB2 exon 4 and STAT6 exon 2 or 3 (which be present (2096}. Calcifications , including psammoma bodies.
have a hypocellular grade 1 phenotype) (989,3340,988,3561, are not seen . Myxoid stroma, giant cells , and/or a variably prom-
209,3104). TERT promoter mutations have been identified in inent adipocytic component (lipomatous SFT), as described in
10-30% of meningeal SFTs (3340 ,731) . TP53 mutation and soft tissue and other extracranial sites (741 ,952,1186,2254.5501.
overexpression of p16 have been reported in more aggressive can be seen in meningeal SFT, but only rarely. Papillary and
tumours (2396 ,1886). pseudopapillary patterns have also been reported (1422.35301
Dedifferentiated (anaplastic) SFT, in which conventional SFT
Macroscopic appearance areas are admixed with focal high-grade pleomorphic sarcoma
SFTs are usually dural-based, well -circumscribed , firm , white to forming eosinophilic amorphous osteoid (osteosarcoma), has
reddish-brown masses, depending on the degree of collagen- recently been reported in a case of recurrent meningeal tu mour
ous stroma and cellularity. Occasionally, they show infiltrative (1981 ,1533).
growth or they lack dural attachment (469,482 ,2096) . Variable In two separate studies , histological grading based on a
myxoid or haemorrhagic changes may be present. combination of mitotic activity and tumoural necrosis has been
found to correlate with prognosis {988 ,19661. In both studies.
Histopathology mitotic activity was evaluated in 1O adjacent high-power fields
SFT is composed of haphazardly arranged spindled to ovoid (400x ; 1 HPF = 0.22 mm 2 ) in the most proliferative zones. The
monomorphic cells admixed with hyalinized , dilated , thin - following grades were identified:
walled, branching (staghorn-shaped) blood vesse ls. SFT has a
wide histological spectrum, ranging from a hypocellular pheno- • CNS WHO grade 1: < 2.5 mitoses/mm 2 (< 5 mitoses/ 10 HPF)
type to a highly ce llular phenotype in a patternless architecture, • CNS WHO grade 2: ~ 2.5 mitoses/mm 2 (~ 5 mitoses/10 HPF)
and multipl0 phenotypes may coexist. The paucicellular end of without necrosis
the spectrum shows abundant stromal keloidal-type col lagen, • CNS WHO grade 3: ~ 2.5 mitoses/mm 2 (~ 5 mitoses/ 10 HPF)
whereas cellular tumours display densely packed round to with necrosis
302 r.~' ~-· H, l oyl I id! I r(Jf 1-1111 I 111 1[)11f l1dul tumour irivolv1ng the CNS
~~ .a: . i;.r·~~~-~~~i~~
Rg. 8.03 Solitary fibrous tumou r. A A highly cellular tumour with thin-walled, branching (staghorn) vessels. B There may be closely apposed cells with round to ovoid nuclei
arranged in a haphazard pattern , with limited intervening stroma. c Numerous mitoses (arrows) are present in this tumour. D Focal necrosis is present in this tum our.

As a consequence of NA82:: STAT6 fusion , diffuse and in the great majority of cases (72 1}. Both primary and metastatic

I
intense nuclear expression of STAT6 (C -term inal epitopes) con- monophasic synovial sarcomas can simulate SFT. lmmunore-
activity for EMA and TLE1 , and/or 55 18 gene rearrangement '
stitutes the immunohistochemical hallmark of SFT, with very I

high sensitivity and specificity {1681 ,2351 ,2870). STAT6 immu - detected by FISH analysis , supports this diagnosis (1900). I

nohistochemistry reliably differentiates meningeal SFT from a Mesenchymal chondrosarcoma , a rare mal ignant tumour with
variety of neoplasms, including mening ioma, meningeal Ewing a biph asic pattern , is composed of sheets and nests of poorly
sarcoma, mesenchymal chondrosarcoma, malignant periph - differentiate d small round ce lls interrupted by islands of well-
eral nerve sheath tumour, synovial sarcoma , and other sarco - differentiated hyal ine cartil ag e and a branching vasculature.
mas that may occur in the meninges {1965 ,531}. Rare cases , Because the cartilage islands can be extremely focal, this
includ ing ded ifferentiated SFTs, may require further molecular entity may be mistaken for malignant SFT if insufficien tl y sam-
or immunohistochemical workup because they may partially pl ed {3426). Malignant peripheral nerve sheath tumour rarely
or completely lack STAT6 nuclear expression by immunohisto- occurs in the meninges an d may resemble SFT, but it 1s usually
chemistry {664,2852,988). CD34 is typically diffusely positive negative for CD34 and STAT6 and may show focal expression
in grade 1 SFT, although little to no expression is common in of S1 00 and SOX1 0.
higher-grade tumou rs {2472). ALDH1 is a robust and specific
marker, staining > 84% of menin geal SFTs compared with only Cytology
1% of mening iomas {343 ,1965). Other markers , such as desmin , Not clin ically relevant
SMA , cytokeratin , EMA, and PR, may be rarely encountered as
a focal fi nding {2096 ,2472 ,2606,3458) . Diagnostic molecular pathology
T~ e NA B2 :: STAT6 fus ion can be detected by sequencing tech-
Differential diagnosis niq ues , RT- PCR , or proximity ligation assay (577.2688 ,1681 ,
The differential diagnosis incl udes both meningothelial and soft 2870}. Because NAB2 and STAT6 are 1n close pro 1m1ty on
tissue neoplasms . Fibrous meni ngioma is a close mimic of SFT ~h romosome 12q, it 1s diffic ult to detect their fusion by conven-
{469]. but it typical ly expresses EMA and is negative for CD34 tional cy~ogenet1c methods , and accounting for the diversity ot
and nuclear STAT6 expression . Dural -based Ewing sarcoma I breakpoints (occurring in both exons and 1ntrons) using PCR -
peripheral primitive neuroectodermal tumour shares the hyper- b.ased detection assays 1s chall en ging . Fortunately 1n1muno-
cellularity and CD99 positivity of SFT. but it lacks nuclear STAT6 h 1 stoc~em1ca l detection of strong nuclear STAT6 expression 1s
staining , and it is cha racteri zed by EWSR1 gene rearra ngement a sens1t1ve and specific surrogate for all fusion s {2870}.

Mesencl1ymal . non-meningoth lial tumours involving the CNS 303


"
Fig. 8.04 Solitary fibrous tumour. A Unusual patterns include a papillary pattern. The transition between a solid and papillary pattern is shown. B The unusual papillary arcri-
tecture is shown at higher power. C This tumour shows a markedly pleomorphic pattern with multinucleated tumour giant cells. D This markedly pleomorphic tumour with gian
cells shows STAT6 nuclear positivity, confirming the diagnosis.

BoxB.01 Diagnostic criteria for solitary fibrous tumour


Essential and desirable diagnostic criteria
See Box 8 .01 . Essential:
Variably cellular tumour composed of spindled to ovoid cells arranged around a
Staging branching and hyalinized vasculature
I ~ot rBlevant AND
Variable stromal collagen deposition
Pfognosis and prediction AND
r1.-0 i;.irge coriorts of patients with SFTs with confi rm ed
STAT6 nuclear expression
NAB2 _:,JAf6 gene fusion and/or nuclear overexpression of
S J-. 16 r1ave c_,rown that meningeal SFT has a high propensity
Desirable (In selected cases):
for recurrer1c8 and me1astas1s, which sometimes occur decades
Demonstration of NAB2::STAT6 gene fusion
&tter tht-~ 1nit1al d1agr1os1s {1966,988}. In one of these cohorts
(132 patients) 11966), recurrent disease occurred in 52 patients scheme as described above. In one series !1966}. progres-
after a median period of 36 months (of those cases, 14 recurred sion-fre~ survival (PFS) was associated with grade and extent
of surgery, and disease-specific survival was associated with
aft~r 10 years) and 16 patients died of the disease after a median
grade as well as its determinants (mitotic count and necrosi~) .
penod ~f 70 months (range: 22- 268 months). In the other cohort
On multivariate analysis, grade , extent of surgery, and m1tot1c
(1 33 patients) {988}, 42 patients experienced at least one adverse
activity remained independent prognosti~ facto rs for PF~ . while
~vent ~fter surgery (local recurrence or metastasis) and 29 died
necrosis was an independent prognostic factor for d1sease-
(including 20 of the disease). In both series, several CNS WHO
specific survival. The other series 1988} compared the .2016
grade. 1 tumours recurred over time, reflecting the experience
CNS WHO tumour grading scheme to the 2013 WHO soft tissue
of. tertiary referral centres . In one series, 6 (4 .5%) of the patients
grading scheme; on univariate analysis , mitotic count, n~cro­
wtth CNS WHO grade 1 tumours ultimately died, 2 with metasta-
sis, CNS WHO grade , and soft tissue ~.rade we~e assoc1a~ed
ses {1966} . Therefore, long-term follow-up is recommended for
with PFS but not overall survival. A mod1f1ed soft tissue grading
the entire meningeal SFT spectrum.
scheme incorporating necrosis was chose n, whi ch showed a
. Th~ widely u.sed risk model applied to SFT occurring at other
sites 1s not entirely applicable to meningeal SFT because older higher correlation with PFS . . . .
Although NAB2::STAT6 fusion type 1s associated with pheno-
~ge doe~ not correlate with worse outcome {1966,988} and
type, it is not predictive of outcome 1989,3340 ,988,3561,209 ,
intracrarnal tumour size is not prognostic {988}. Only mitotic
3104}. TEAT promoter mutations have not been shown to cor-
count and presence of necrosis appear to correlate with prog-
nosis, resulting in a histologically based three-tiered grading relate with worse outcomes {3340} .

I
I

Mcse ncr1ymal, nun-rn eningothell I tumou rs involving the CNS 305


Haemangiomas and Ha1nfellner J,A.
Bouvier C
vascular malformations Calon1e JE
Scio! R
Thway K

Definition
Haemangiomas are benign neoplastic vascular lesions with
multiple tightly packed capillary-sized and cavernous vessels .
They may be isolated, multiple , or part of a P/K3CA-related
overgrowth syndrome .
~avernous malformations (CMs) are angiographically occult
solitary or (rarely) multifocal vascular anomalies . Histologi-
cally, they comprise multiple tightly packed sinusoidal vessels
with fi.br~tic walls lacking arterial or venous features, and they
contain little or no interposed CNS tissue. Familial and some
sporadic CMs are associated with a mutation in KRIT1 (CCM1),
CCM2, or PDCD10 (CCM3).
Cerebral arteriovenous malformation (AVM) is a fast-flow vas-
cular anomaly consisting of arteriovenous connections through
a nidus or fistula of malformed arteries and veins instead of a
normal capillary bed . AVMs are typically sporadic; they show
interven ing brain parenchyma with gliosis between malformed :~
vessels , and they have frequent somatic KRAS or BRAF muta-
tions .
Capillary telangiectasia is an aggregation of individ ually dis-
persed dilated capillary-type vessels with interposed normal
brain parenchyma .

ICD-0 coding
9121 /0 Cavernous haemangioma
9131 /0 Capillary haemangioma
c
9123/0 Arteriovenous malformation -
- • ~ A -

. •·....:-i>
j._...~ ~". . . ..· ~
",,,. .. .-
ICD-11 coding .. . . '· .
2E81 .0Y Other specified neoplastic haemangioma
LA90 .3Y Other specified peripheral arteriovenous malforma- __,,.,, '' • '
tions

Related terminology · .~ ' ·


Acceptable: cavernous angioma; cerebral cavernous malfor- i "' ··. ,
"· ,.
mation; cavernoma.

Subtype(s)
None '".._
Fig. 8.06 ~ae~angioma. A Axial CT (bone window) of vertebral body L3 showing !he
Localization polka-dot s.1gn with th1ckened.tra~eculae. B Sagittal reconstruction of the CT {bona w
Haemangiomas arise preferentially in the spine; less frequently dow) showing the c?rduroy sign 1n vertebral body L3. C Presence of capillary-type a1 ·
1n the skull; and exceptionally in the CNS parenchyma, nerve cavernous vessels in the bone marrow space between coarse trabeculae of b ne 1n a
roots , and cauda equina (2944,938,1686,3566 }. Spinal haem- calvarial haemangioma. D Calvarlal haemangioma with cavernous vessels Jif1€'d by
ang1omas favour t11e thoracic and lumbar vertebrae . They are tened endothelium and separated by loose mesenchymal stroma with fibroblasts
often multiple, and they may involve vertebral bodies, pedicles,
arches, and sp1nous processes {3159) . Cerebral A~Ms can invo.lve the meninges . cortical reg 1 ns
CMs favour supratentonal locations, including the optic nerve and deep brain (insular region. basal ganglia. thalamus . rpu..:
or d113~n1 pineal gland, 3nd cavernous sinu s. Rare locations callosum, brainstem, and cerebellum) (3566} . In the spinal cu
1ncluac~ Hie cererJf·llopont1ne ang le, pons, ce rebellum , and spi- AVMs can. be ex tradural, intradural, or intramedullary, ur thev
nal L.urrJ \:3~Chl Spinal cord CMs ore usually intramedullary. can occur in the conus medullaris (2361 I
Cerebral capillary telangiectasia shows a predilection for the Imaging
pons and may extend into the middle cerebellar peduncle. The On CT, vertebral haemang iomas show a typical polka-dot, hon-
basal ganglia , cerebral hemispheres , and spinal cord may also eycomb, or corduroy pattern . On MRI , they are hypenntense
be affected {3566}. on T1- and T2-weighted images , and they show postcontrast
enhancement {3159) .
Clinical features On CT, calcifications are frequently visible in CMs . On MRI ,
Vertebral haemangiomas occur predominantly in male patients. T2-weighted images show mixed signal intensities centrally
They are usually asymptomatic, but they may also lead to ver- and a surrounding hypointense/low signal rim with blooming
tebral compression fracture , inducing compressive myelopathy (haemosiderin ring) . T1-weighted imaging with contrast shows
{3159 ). Pregnancy may induce progression (3369) . the associated developmental venous anomaly, if present. On
Patients with cerebral CMs more commonly present with angiography, CMs are occult, because they lack feeding arter-
seizures than with acute haemorrhage. The age range is wide . ies and draining veins . CMs are dynamic lesions that may anse
Male patients are affected in the second to third decades of de novo and may grow, contract, or remain the same size. In
life, and female patients often in the fourth to sixth decades. sporadic cases, single isolated lesions with or without associ-
lntralesional or perilesional haemorrhage may cause acute ated developmental venous anomaly are seen on MRI : 1n famil -
foc al neurological deficits that may improve without neurosur- ial cases , multiple CMs are seen (3567,3566).
g1cal intervention {946) . Most CMs remain asymptomatic, and On CT, vessels in AVMs are isodense and strongly enhancing
they can be incidental findings on MRI or autopsy (648). with contrast. Calcifications may be present, and haemorrhage
Patients with spinal CMs may present with pain, myelopathy, visible , with a nidus in the region. On T2-we1ghted MRI . AVMs
or radiculopathy; patients often also have intracranial lesions ap~ear as honeycomb flow voids due to rapid signal loss from
!1130). les1onal h.1gh flow. T2-weighted and FLA IR images show hyper-
AVMs can occur at any age, with no sex predilection . Patients 1ntens1ty 1n surrounding CNS parenchyma because of gl1os1s
with cerebral AVM most frequently present with acute intracer- due to 1schaem1a from the vascular steal pt1enorn2non ot the
ebral , intraventricular, or subarachnoid haemorrhage , typically AVM shunt. Angiography visualizes AVMs <-lS conglom erates ot
in the third to fourth decades of life. Other frequent manife s- tortuous vessels with early venous dra1naqe t-\VMs are usu.lily
tations are seizure, chronic headache. and progressive neuro- solitary, but n1ey rnay be multiple [3566).
logical deficit AVMs may also be asymptomatic and discovered On MRI with contrast, ect tic capil laries in cerebral capillary
1nc1dentally on imaging . AVM s n1ost common ly come to cl1n1cal telang1ec tas1as show mild to moderate enhancement with an
attention in the second to fourth decaues of Ille (450, 3566 ). irregular border. T1 -weighted postcontrast imaging may show

Mt"SPnd1 m ;..11 , 11 0 11 rne 111nyothel1 al tumours involving the CNS 307


' ... _
(, ! • •"1• II • \

. .. .. '
• I• ,

, ... , "'
' ;, ,. .J' ~
• f •• •• • ,
a, JI . ,f./.. . •
. , .. ~ , ' · .;. , ,

·. ..
.tA ;

~ . .. . . ,,
If" I I •• " " '. . ft,· '1
_1 .,... . . ' f' ... ,• •
J .,

. .... / ';;
,
. ' ' ..
t '',
...
,

B C _,,
Fig. 8.09 Artenovenous malformation. A T2-weighted axial MRI showing an arteriovenous malformation in the left temporo-occipital region with charactenstir, now r: 15 ~ ~~
nidus area (thick arrows) and dilated draining vein (thin arrows). B Digital subtraction angiography via the left vertebral artery (coronal proiection). The artenovenous rr.~r,,_,a ..,.,..
is visualized as a conglomerate of tortuous vessels: note the large nidus (asterisk). a feeding artery (thin arrow), and the drainrng veins (thick arrows). C CNS tissue M1t: t _-=
be ween two malformed vessels D Variably sized abnormal arteries with a large feeder (left) and interposed CNS tissue (elastica van Giesen).

Flg.8.10 Capillary telangiectasia. A T1 -weighted axial MRI of a capillary telangiectasia located in the pons, with faint brush-like enhancement (arrows) after contrast adrr"
istration. B Axial susceptibility-weighted MRI demonstrating susceptibility artefacts in the pons due to the capillary telangiectasia (thin arrows). Note the draining vein 1 t~c..1
arrow). C Cerebral lesion. Dilated capillary-type vessels with a flattened single-layer endothelium are separated by CNS tissue without gliosis.

a draining vein from the capillary telangiectasia. Susceptibility- (CCM3) (3567). The mutations most common ly result rn a rrt..!r: -
weighted imag ing shows signal intensity loss due to deoxyhae- cated protein ; rarely, missense mutations lead to protei n rn 's-
moglobin loss in stagnant vessels {3566) . folding {1880).
Most sporadic AVMs have somatic KRAS mutations or. less
Epidemiology frequently, a BRAFmutation {1149) .
Vertebral haemangiomas have a prevalence of 10-12% in the
general population {33691. Pathogenesis
For CM, the estimated population-based prevalence ranges The development of CM is thought to be a phenomenon ~ ·
from 0.16% to 0.9%. The estimated population prevalence of venous hypertension leading to erythrocyte extravasac1on ana
familial CM ranges from 0.01% to 0.03% [3567) . the release of angiogen ic growth factors. CMs may also be
For AVM , population-based studies approximate the inci - associated with developmental venous anomalies {3566)
dence to be about 1 case per 100 000 person -years [2338) . Familial CMs are associated with a mutation in KRITJ (CCM' I
The prevalence of capillary telangiectasia is 0.4- 0.7% in the CCM2, or PDCD10 (CCM3) . These genes are part of a signa-
general population (3566) . ling pathway that regulates cell proliferation . network forrna11<.. •1
and endothelial layer growth (3567,160) .
Etiology In AVM , dysregulation of angiogenesis , vasculogenes:::
lntraoss<::ous haernangioma c an be congenital or can develop and inflammation seems to be involved in pathogenesis t 50!
(je novo (r:: g due to traum a) {2944). Haemangiomas may also Sporadic AVMs may relate to abnormal ities 1n cerebral as-
br; part of a PIK3CA-related overgrowth syndrome (e.g . Klip - cu lar autoregulation or in venous architecture. AVMs ~ an ai - -
P'::I rr&nounay .. ynrfrorne) (1752]. occur in association with hereditary genetic syndrom es. 'uc.
McJ ~J CM 1r~ s1on ~J are sporadi c, anu they can be congenital as hereditary haemorrhagic telangiectasia syndrome L1 -1~r­
rJ r C1V~u1 r (:: cJ Ctv1<~ c;;ri ck:velop yea rs aft er brain or spinal irra Weber- Rendu disease) (1149).
(Jia!iori n -1riJ I hr~ (fl(;l(-(,IJIW basis of inh erited CMs (and of a Cerebral capillary telangiectasias may be congenita l 1n mg 1
r>' 1Jpr;rtir;1 1r if sprjr<Jd1c. U k>) 1!, ar 1 autosorn al dominant loss-of- (caused by a failure of capi ll ary involution during devel pmt?1h •
fur ,ct1 cr rri.J l at11ir 1 111 l< Rlf I (C C MI) (cum rr1 0 11 in tr1 Hispanic
1 or dc quir ed due to reactive angiogenesis after insults such ,1 -
p1Jp ul0t1cm1, CC M:) fvvt 1:d1 c: r1 c cid c-: •J 1r1alci..tVerr 11n), or POC010 venous twpertension or irr diation {35661 .

'
~!·~~ _?~~o~ic ~iteri~for ~a~ma~glomas and vascular malformations
small , poorly canalized channels lined by plump endothe-
Haemangloma
lial cells (giving a highly cellular solid appearance) to dilated
EINntllll: vessels lined with flattened endothelium . Blood- filled cavern-
lightly packed capillary-sized and cavernous vessels with a single layer of benign ous spaces and f1broendothelial papillae m1m1ck1ng oao1llary
endothelial cells endothelial hyperplasia may be observed The stroma displays
ANO haemorrhage, haemosiderin , fibroblasts. and oedema, but
Mesenchymaf stroma with fibroblasts foamy macrophages are absent. Reticu lin stain shows a deli-
AND cate network of reticulin fibres surrounding vessels lmmuno-
histochemically, the endothelial cells express ERG . C031 . and
Absence of foamy stromal cells
C034 . Single SMA-immunostaining cells may be present in the
Beslrable: subendothelial layer. VEGF-positive cells are seen 1n the solid ,
Typical neuroimaging findings immature-appearing areas without vessel lumen formation . The
Cavemous malformation Ki-67 (MIB1 ) index is usually< 10% (7,1686,9381 .
CMs are histologically well circumscribed , showing sinusoidal
Eaentta/: congested vessels (caverns) without intervening arteries, capil-
TlQhtty packed sinusoidal vessels wtth a single-layer attenuated endothelium and laries, or veins. The vessels comprise a single layer of attenu-
fbrotic vessel walls, lacking arterial or venous features ated endotheli um and tibrotic walls , without arterial or venous
AND features and without smooth muscle cells . Electron microscopy
Absence of prominent feeding arteries and draining veins shows defects in tight junctions between endothelial cells , per-
Deslrable: mitting leakage of blood components. The vessels are arranged
back to back with little or no interposed brain tissue. There may
Deposition of haemosiderln in surrounding CNS tissue
be hyalinization , calcification , cholesterol crystals. and micro-
Typk:al neuroimaging findings haemorrhages. The brain parenchyma abutting the lesion
Arterlovenous malformation shows haemosiderin deposition and gl iosis. Macrophages and
&Anfial: chronic inflammation may be present {648).
Histologically, AVMs are composed of variably sized abnor-
Aggregates of abnormal arteries and veins of variable diameters with direct mal arteries and veins with direct fistulous connections and
connections through a nidus or fistula, instead of a normal capillary bed
without normal intervening capillary beds. Between vessels,
Dnir.able: there is CNS tissue with gliosis.
Typical neuroimaging findings Cerebral capillary telangiectasias are localized aggregations
Capillary telanglectasia of thin-walled , dilated vessels without elastic fibres or smooth
muscle cells. They occur within brain parenchyma without adja-
Essential:
cent gliosis , calcification , or haemosiderin-laden macrophages .
Aggregation of capillary-type dilated vessels lined with a single benign endothelial
cell layer
Cytology
ANO Not clin ically relevant
Lack of tissue alterations in the intervening CNS parenchyma
Desirable: Diagnostic molecular pathology
Not clinically relevant
Typical neuroimaging findings

Essential and desirable diagnostic criteria


Macroscopic appearance See Box 8.02 .
Macroscopically, haemangiomas are soft, red , and lobular, and
they are associated with many small feeder and drainer vessels Staging
11686). Not relevant
CMs are macroscopically circumscribed and lobulated , with
a reddish-purple raspberry appearance. The surrounding brain Prognosis and prediction
tissue contains haemosiderin 1946). Haemangiomas of the neuraxis usually do not recur after com -
AVMs macroscopically show dilated surface draining veins, a plete resection (1686).
nidus located deep to these , and feeding arteries 12360). In CMs, the annual risk of clinically significant haemorrhage
Cerebral capillary telangiectas1as are vascular lesions without is 1.6-4.6%. Previous haemorrhage and brainstem location
mass effect and are usually small (ranging from only a few mil- are associated with increased risk Female sex , larger lesion
limetres up to -20 mm). size. and greater number of lesions are also associated w1tt1
increased risk of haemorrhage 135661
Histopathology In AVM , the risk of haemorrhage is 2 - 5% per year 111 AVM
Haemangiomas show a lobular pattern separated by fibrous haemorrtiage, 5- 25% of cases are tatdl. Age . hyperrensinr1
septa. Each lobule is fed by a separate artery and consists of and previous intracrarnal haemorc hage are assu 'l..::1tPc.i w1 t11 ,:i.r1
multiple capillary-sized vessels lined by single layers of bland increased risk of haemorrhage {450}.
endothelial cells with very rare m1tot1c act1v1ty. Rare cases with Capillary t~lang~ectasia is most often an incidental finding ,
high mitotic activity may be see11 Ves::;els vary 1n size frorn with only a minor risk of bleeding or progressive course.

Mest:r ichymal. 11011 -rr 1ernngothel1a1tumours in volving the CNS 309


Haemangioblastoma Tihan r
Fanburg -Smith JC
Vortrneyer AO
Zagzag D

Definition and hydrocephalus . Cerebellar deficits such as dysmetna 8~r:


Haemangioblastoma is a highly vascular tumour containing ataxia can also occur. Haemangioblastomas produce er ~hr,_
neoplastic stromal cells that have clear to vacuolated cytoplasm poietin , and this may cause secondary polycythaerrna (11 171
and characteristic immunohistochemical features (e.g. inhibin Haemangioblastoma is a frequent manifestation of VHL [11~5t
positivity) and molecular findings (e.g. VHL alterations) (CNS Therefore, genetic counselling and screening for VHL gerr--
WHO grade 1). line mutations are critical in the management of patients Mtr
haemangioblastoma (1115) .
ICD-0 coding
9161/1 Haemangioblastoma Imaging
Neuroimaging typically demonstrates contrast-enhancing noa-
ICD-11 coding ules that are frequently associated with cystic structures. T'"'e
2F7Y & XH6810 Neoplasms of uncertain behaviour of other solid component is usually peripheral in location within the ce'"-
specified site & Haemangioblastoma ebellar hemisphere. Flow voids may be seen w1th1n the nodu:e
due to enlarged feeding/draining vessels. Angiography 1s use-
Related terminology ful for identifying small lesions, showing a mass with a dense
Not recommended: angioblastoma; von Hippel- Lindau dis- tangle of vessels that may resemble arteriovenous malforma-
ease. tion. Spinal tumours can cause pain , hyperaesthesia, or incon-
tinence due to local compression, and they may be associated
Subtype(s) with a syrinx. Haemorrhage is a rare complication .
None
Epidemiology
Localization lntracranial haemangioblastoma is estimated to have ar
Haemangioblastomas typically occur in the cerebellum . Spo- annual incidence rate of 0.15 cases per 100 000 population
radic and multiple tumours are also found in the brainstem, (2244}. Tumours typically occur in adults , with an M:F ratio
spinal cord , cerebrum, retina, and peripheral nerves. Haeman- of 1:1. VHL-associated tumours occur at a younger age than
gioblastomas can also occur outside the CNS, such as in bone sporadic haemangioblastomas. Symptomatic presentation is
and soft tissue, liver, lung , pancreas , kidney, intestines , and skin usually in people aged 18- 30 years , and the disease has 95°b
(785,287,2181 ). penetrance by 60 years , although there is considerable vari-
ability.
Clinical features
Haemangioblastomas account for< 2% of all CNS tumours, and Etiology
they may occur sporadically (most commonly) or in association In both sporadic haemangioblastoma and VHL. allelic losses or
with von Hippel-Lindau syndrome (VHL) (2244). Symptoms are mutations of the VHL gene are found in stromal cells. VHL is
generally caused by the mass effect of the lesion through com- autosomal dominant, through a germline inactivating mutation 1n
pression of adjacent structures, and/or by the impairment of the VHL gene on chromosome 3p25 .3 with subsequent inactiva-
cerebrospinal fluid flow due to increased intracranial pressure tion of the second allele in tumours . In the majority of patients

Flg.8.11 Haema1191ubldstonia 111 the pos1e11u1 fossa 1n ci 5::' year old ma11. dt:rnonstrat1ng 1yp1cal solid cysllc teatures Axial MRI. A Tt weighted , contrast-enhanced 111
aye B r2we1ghtt:d 1111age c fl AIR 1111uye
Pathogenesis
The variably lipid-fi lled stromal cells release angiogeni c fac-
tors , including vascular endothelial growth factor (VEGF ), which
leads to the production of the rich vascular network present 1n
the tumour. HIF1 , a ubiquitously expressed and highly con-
served heterodimeric basic helix-loop- helix PAS transcription
factor composed of two subunits , HIF1 a and HIF1 P. plays an
essential role in oxygen homeostasis (2879) . In normoxic con -
ditions , after hydroxylation of HIF1a by the oxygen-dependent
prolyl hydroxylases at pral ine re si dues 402 and 564 within
the oxygen -dependent degradation domain , the protein com -
pl ex VCB -CUL2 (which includes VHL protein . an E3 ubiquitin
ligase) binds HIF1a and polyubiqu itinates it, targetin~ i'. f~r pro-
teasomal degradation . In contrast, when VHL protein 1s inacti-
flt.1.12 Haemangioblastoma. A,B The tumour is multicystic (wh ite arrow) and well vated or when cellular oxygen conce ntration decreases , HIF1 a
demarcated from the surrounding cerebellum (black arrows).
accumulates in the cytoplasm , tran slocates into the nucleus ,
and bin ds to hypoxia-response elements in the promoters of a
with V~L there is a family history uf the syndrome, so only one large battery of genes whose protein prod ucts function either
other d.1sease manifestation is necessary for the diagnosis. to increase oxygen availabil ity or to allow metabolic adaptation
Studies on sporadic tumours (including somatic mutation to oxyge n deprivation (1524}. For example, th e inactivation of
analyses.' assessments of allelic loss, deep-coverage DNA VHL protein in haemangioblastomas leads to th e accumulation
sequencing , and hypermethylation studies) have found a loss of HIF1 a in stromal cells (3572} and triggers increased tran -
or inactivation of the VHL gene in as many as 78% of cases scription of HIF1a-regulated genes (i nc lud ing those encoding
l1 114A,1837A,2890A}, suggesting that loss of function of VHL is VEGF, erythropoietin, glucose transporters. and glycolytic
a central event in haemangioblastoma formation . enzymes).

I
i
I

~ .
Flg.8.13 Haemangioblastoma. A lntradural, extramedullary localizallon 1s typical for spinal haemang1oblastornas Most tumours are well circumsc- b d b ,
encr h h · . h · bl n e ut t11ey may a1so
t hoac on t e spinal co rd parenchyma.
.
B Abu ndant vascu1anty o1 aemang10 astoma ·
is o1ten 1r1 the torrn of thin -walled vessels • some of whi'ch appear 'as h1ghi y branching
s ag orn vessels. C Neoplastic stromal cells with clear to vacuolated cytoplasm admixed with abundan1capillary vessels. D The stromal cells show "Id 1. 1 h.
and a rich capillary network. m1 nuc ear p eomorp ism

Mese11 c hyrnal , non-meningothelial tumours involving the CNS 311


. Mor~ tha~ 1000 unique VHL mu tations (substi tutions, dele- Histopathology
tions, 111sert1ons, and duplications) have been described in the Haemangioblastomas demonstrate two main u;rrpr,ri::- -.
Catalogue ?f Somatic Mutations in Cancer (COS MI C) , about (1) neoplastic stromal cells that are characteristically la r'J~ :::,..-!
25% of which are pathogenic . Recurrent DNA copy-number vacuolated but that can show con si derable cytolog1r:al 1;r':::-
losses of chromosomes 6q and 6, somatic gain-of-function tion, and (2) abundant reactive vascular cells . In m;:in1 turr r1 w":
EPAS1 (HIF2A) mutations , and loss of SD HB expression have vascular cells are more abundant than stromal cells. 1n 0tn-;r
been reported . Various cell signalling mechani sm s are upregu - tumours, stromal cells are more abundant and may reveal -:r:. ·':!
lated in CNS haemangioblastomas , including EGFR (HER1) , epithelioid aggregates that may be associated with extrarred•. . -
TGF-a, FGFR3, PDGFRA , and Notch si gnall ing pathways or lary haematopoiesis {3349A}. The most characteristic and rj ;-
receptors. Copy-number variations in various genes, including tinguishing morph ological feature of the stromal cell is nurrer-
gain of EGFR and microdel etion of FGFR1 , have been identi- ous lipid-containing vacuoles. Tumour cell nuclei can 1a y ,r
fied in haemangioblastomas. Recent whole-exome sequencing size, with occasional atypical and hyperchromatic nuc 1 ~ ·
of famil ial and sporadic CNS haemangioblastomas identified Mitotic figures are rare . In adjacent reactive tissues. par 1cular·/
multiple somatic single-nuc leotide variations and several copy- in cystic and syrinx wal ls , astrocytic glios1s and Rosenthal fibres
number variations associated with angiogenesis . Mutations are frequently observed . The tumour edge 1s ge nerally f!E
in BRCA2 (COSM3753648 , COSM5019704) have also been demarcated , but infiltratio n into surrounding neural trssues car
reported . be detected . Cellular tumours with clear cell aggregates rr21
resemble metastatic renal cell carcinoma .
Macroscopic appearance The neoplastic stromal cells frequently express a.-1nh1b1n
Haemangioblastomas are typically well-circumscribed pseu- (1321A], 02-40 (2741A], and brachyury (cytoplasmic expres-
doencapsulated masses that may be cystic with a mu ral solid sion), which may be helpful for differentiating haemang1oblas-
nodule or (l ess commonly) entirely solid (3363) . The re is a toma from metastatic renal clear ce ll carcinoma, particular·;
variegated yellow cut surface due to the high lipid content . in the context of VHL. Identification of stromal cells is further
Tumours are up to 125 mm in diameter in extraneuraxial loca- facilitated by positive immunoreactivity for NSE, NCAM1 [31 1H
tions an d generally< 30 mm in the cerebellum [287,785 ,1373]. S100 (1420), ezrin (315), CXCR4 (1 88 5,3570 1. aquapori'1-
11922), several carbonic anhydrase isozymes 12577). occasion- Box8.03 Diagnostic criteria for haemangloblastoma
ally GFAP {1593], and EGFR {313A) . Positivity of these markers
in addition to negative staining with RCCm , EMA , CD10, and Essential:
A tumour composed of large, multivacuolated, and lipidized stromal cells with
CAM5.2 anti b~d i es , may be helpful for exclud ing a metastatic
occasional hyperchromatic nuclei, as well as a rich capillary network
renal cell ca.rc1noma . Especially in patients with VHL , staining
tor these antigens may be helpful in the differentiation of haem- AND
a~giobla~toma tror:n many different tumours , but there is overlap Stromal cells with immunohistochemical positivity for markers such as inhibin
with the 1mmunoh1stochemical profile of renal cell carci noma. (at least focally)
Stromal cells lack endothelium-associated markers, such as OR
von Wlllebrand factor (factor VIII - related antigen), CD34, ERG , Loss or inactivation of the VHL gene
and CD31 {314,3464A}. OR
In a patient with van Hippel-lindau syndrome
Cytology
Cytological smear preparations often appear as cohesive tissue Desirable:
fragments and yield few individual cells. Stromal cells with spin- In patients with van Hippel-lindau syndrome, absence of immunohistochem1cal
dled morphology and cytoplasmic vacuoles can be identified at staining for markers of renal cell carcinoma
the periphery of the tissue fragment. Occasionally, tissue frag -
ments may demonstrate large irregular nuclei. Rare cells with
clear cytoplasm can be seen {623). Prognosis and prediction
Haemangioblastoma has excellent pro gnosis after complete exci-
Diagnostic molecular pathology sion. Permanent neurological deficits are rare {631}, and they can
Not clinically relevant be avoided when the haemangioblastoma is treated early \1116).
Sporadic tumours have a better prognosis than VHL-associated
Essential and desirable diagnostic criteria tumours because the latter show multiplicity and occur in com-
See Box 8.03. bination with other VHL-assoc iated neoplasms (2131 }. Tumours
may persist locally after incomplete excision (785). Features that
Staging affect outcome include age, size, growth pattern , treatment, mul-
No currently accepted staging system is recognized . tifocality, and location (e.g. brainstem vs cerebellum) [13731.

I
I

Mesenchymal , non -meningothelial tumours involving the CNS $13


Rhabdomyosarcoma Kleinschmidt-DeMasters Bl<
Bouvier C
Dry SM
Hainfellner JA
Rudzinski ER

Definition
Rhabdomyosarcomas are a family of malignant primitive neo-
plasms that show at least focal , predominantly skeletal muscle
differentiation and are rarely identified as a primary tumour in
the CNS .

ICD-0 coding
8910/3 Embryonal rhabdomyosarcoma
8920/3 Alveolar rhabdomyosarcoma
8901/3 Rhabdomyosarcoma, pleomorphic-type
8912/3 Spindle cell rhabdomyosarcoma

ICD-11 coding
2855.Z & XH83G1 Rhabdomyosarcoma & Embryonal rhabdo-
myosarcoma, NOS Flg.8.15 Rhabdomyosarcoma. Tumours occasionally contain elongate strap c
2655 .Z & XH7099 Rhabdomyosarcoma & Alveolar rhabdomyo- with eosinophilic cytoplasm, as seen here. Note, however, that the ma1ority of tumour
sarcoma cells are poorly differentiated and contain scant cytoplasm.
2655 .Z & XH5SX9 Rhabdomyosarcoma, primary site & Pleo-
morphic rhabdomyosarcoma, NOS
2855 .Z & XH7NM2 Rhabdomyosarcoma, primary site & Spin- that infratentorial I skull base sites (66% of cases) predominated
dle cell rhabdomyosarcoma over supratentorial locations (34% of cases) {3603).

Related terminology Clinical features


None Symptoms are referable to tumour location, but many patients
present with headache {3603} and mass effects such as nau-
Subtype(s) sea and vomiting {1501}. Supratentorial examples may cause
None hemiparesis (727} or extremity weakness {2379}. lnfratentona1
skull base examples are often associated with cranial nerve pal-
Localization sies (3603}. and intrasellar examples have mimicked symptoms
There is no stereotypical location for primary intracerebral seen in pituitary adenomas, such as bitemporal hemianops1a
rhabdomyosarcoma; examples have occurred in the cerebello- (3025}.
pontine angle {458,3544,2196) and in meningeal {2379,3494).
pineal 11812,1418}, posterior third ventricular and pineal (1501}, Epidemiology
and sellar 1803,135,3025) locations. A recent study of 12 exam- The majority of rhabdomyosarcomas develop in children , bur
ples plus cases published in the literature since 1946 found adults may also be affected (1269,740 ,2379,1501 ,135.30251.

Flg. 8.16 Alveolar rhabdomyosarcoma. A A h1gt1ly cellular !Uniour co mposed ol p1 imitive round cells with scant cytoplasm and hyperchromatic nuclei. Fibrovascular septa are
seen 1n this example of the solid type of tumour. B The alveolar type often contains scattered mult1nuclealed, wreath-like, giant tumour cells, as seen here.

314 Mesenct1ymal . no11 -rneningothel1al tumours 1nvulvir1g tt ·1e Cf\JS


c-mcoclin~ effnc tor:; of PnK (PTF N, PIK.1CA) .::.mrJ rJ0r1 r;•-, thr:it u )r 1
trol the cell cycle (FBXW7, CTN NB I) l?qrJc:. ,?rH9)
A t(2 ,13)(q36:q 14) translocation 1s fnunrJ 1ri thP, rnairmt·; •;f
alveolar rhabdomyosarcomas. ancJ a t(1 : 11J(p ~JF, .r~ 14) tr.:=rnslr;c<'.3-
lion is seen 1n a smaller subset. These transl0cat1on:, 11J1tapry;~
PAX3(at 2q36 .1) or PAX7(at 1p36.13) with the FOXOI qenFJ Cil
13q14.11, to generate chimeric genes that encorJ8 PA/3 FlJ/G 1
and PAX7:: FOX01 fusion proteins [202,693,1024 1. P/l .A3 n.rirJ
PAX? are transcription factors that play essential roles 1n rnyrj-
genesis [400) .
Please refer to the Soft tissue and bone tumours volume for
additional details of pathogenesis

Macroscopic appearance
Gross specimens are usually described as being moderately
vascular and firm; these characteristics can make it difficult to
ach ieve complete neurosurgical removal without complications
Etiology [3544} .
The etiology is unknown . Although system ic-site embryonal
rhabdomyosarcomas have been seen in patients with naevoid Histopathology
basal .c~ll carc inoma syndrome (Gorl in syndrome) 12564}, that The two most common types in systemic sites are embryonal
assoc1at1on has yet to be documented for primary intracerebral and alveolar rhabd omyosarcomas ; recently reported examples
examples . Patients w ith neurofibromatosis type 1 have a small of primary intrace reb ral rhabdomyosarcomas have also been
risk(< 1%) of developing systemic rhabdomyosarcomas {879} ; embryonal (1269,135,3 025) and alveolar (1501,740} when his-
one intracranial case has been described in association with tologically characterized . Pleomorphic rhabdomyosarcoma is
neurofibromatosis type 1, although the patient had received less commonly reported {1501}. No intracerebral cases of spin-
prior cranial irradiation for bilateral optic nerve gli oma [727}. A dle cell / sclerosing rha bdomyosarcoma have yet been con-
definite radiation-induced brainstem rhabdomyosarcoma has firmed ; however, spind le cell / sclerosing rhabdomyosarcoma is
been reported in a patient with neurofibromatosis type 2 {4 68}. freq uently parameningeal.
A meningeal rhabdomyosarcoma was found in a 15-month-old
infant with hypomelanosis of Ito 13494). Microscopy
Rhabdomyosarcomas manifest varying proportions of undiffer-
Pathogenesis enti ated small cells and strap c ells with cross-striations . Mitotic
Sporadic cases of embryonal rhabdomyosarcoma are aneuploid activity is often brisk.
with whole-chromosome gains including polysomy 8, followed Typical embryonal rhab domyosarcomas are composed of
in number by cases with extra copies of chromosomes 2, 11 , variably differentiated rhabd omyoblasts within a loose , myxoid
12, 13, and/or 20. In most embryonal rhabdomyosarcomas , a mesenchyme , wi th altern ating areas of dense and loose cellu-
genomic event such as chromosome loss or deletion , or uni- larity. The proportion s of myxoid matrix and spindled cells vary
parental d isomy, results in the loss of one of the two alleles at greatly between examples. Many tumour cells may be small
many chromosome 11 loci. This loss of heterozygosity involves with scant amphophilic cytoplasm, but differentiating rhabdo-
chromosomal region 11p15, which contains impri nted genes that myoblasts show larger cytoplasmic volume, more cytoplasmic
encode a growth factor (IGF2) and growth suppressors (H19 and eosinophilia, and elon gation . Terminal differentiation with cross-
CDKN1C) {2906}. Genomic studies of embryonal rhabdomyosar- stri ations or myotu be formation may be evident.
comas have identified somatic driver mutations in genes involved Alveol ar rhabdomyosarcomas are highly cellular and com-
in the RAS pathway (NRAS, KRAS, HRAS, NF1, FGFR4), genes posed of pri mitive round cells with scant cytoplasm and

~
... ' • " I
~ _, ..(.
B~ .
... ~
D
. .,_•..
Flg.8.18 Primary CNS alveolar rhabdomyosarcoma. A This tumour is composed of sheets of poorly difft:Hdilldfed ::ells B At l11gher magn1ricat1011. this tumour is monomorph1c
..
with round to ovoid nuclei and fine ch romatin with inconspicuous nucleoli. C Myoganin. (MYF4) stains ti~,, ma1or;:y ot luMour nuclei in tn1s P4 '(J: F0.'1<.01 fusion-positive alvevlar
rhabdomyosarcoma. D After chemotherapy, th is tumour showed cytod1tferent1at1on. Ditfeientiated turnour cellb Ir H' ciliundant eosin0ph1'1c cyt ~plJsm and ecc~ntrically located
nuclei with stippled chromatin and small nucleoli.

\{ 1,,1l }\ ' I ! r 1, ... 1! f 1ti~l,ll r 1.: ·~I •• itl) f •,l. \ \J . ._) J15
hypercl1ro111at1c nuclP1 <manged in nests separated by f 1bro- glc=ind ( 18121 and wh'3n srnall amQ•mts r,f r;tr. ~r '1 ' - .r_,
vascular s pta Loss of ce llular cohesion centrally mFly resul t Cartilage are presen t 114 1r;al M alt n~ n~nf ~r
~ ,
tr_1fY•' - --.>:',
, • I

1n irregular alveolar spaces and cystic change: the solid pat- mixed tumour com posed of ga nglirm (.. ~! Is r,r ,.. C:•;r r
tern lacks th es f ibrovasc ular septa Multinucleated , wreath-like one or more mesenchymal elements (1;c;.; a ll1 '~ ~ ,_/!,.
tumour giant ce ll s are freq uent, but overt rhabdomyob lastic dif- coma), also rarely occurs in the brain 124321 B~r · 'y ,.. . ::fr.,..:,
ferentiation 1s not typically seen . oma consisting of mature skeletal muscle sh01; ltj rr:! ~,- _....~
Spindle ce ll I sclerosing rhabdomyosarcoma is heteroge-
neous Purely spindle cell forms demonstrate fascicu lar, whor- Cytology
ling, or herringbone architectu re with uniform spindled cells in Cytological preparations of en:'br1onal rhabdl')(T" tr.r- - ~ --
intersecti ng fascicles . The sclerosing pattern contains round , demonstrate primitive round . spindled. a n~ st~ll~ e ~
oval, or (less often) spindled cells within a hyalinized/collagen- scattered rhabdomyoblasts. Fine-needle b•ops e.c:; rJ _ .,_
ized stroma typically showing cord -like or nested patterns sug- rhabdomyosarcoma are highly cellular and consist . _
gesting vascular or alveolar spaces. round cells with scant cytoplasm and variable rhabtjcr-.,..: _.:o::"
Pleomorphic rhabdomyosarcoma shows sheets of large, tic differentiation. Wreath-l ike multtnucleated giant ~~ c; ~' -
pleomorphic rhabdoid, spindled , or polygonal cells, often multi- seen (2393).
nucleated.
Diagnostic molecular pathology . .
/mmunophenotype Most alveolar rhabdomyosarcomas in systemic sites are -.:
All rhab domyosarcomas should show cytoplasmic immunore- acterized by the presence of PAX3:FOX01 or PAX7 H:,K
activity for desmin, although the extent of immunoreactivity is fusion , with a worse prognosis in those without ttie :. -
variable. The skeletal muscle- specific nuclear regulatory pro- Diverse molecular changes have been reported 1n sp
teins myogen in (MYF4) and MYOD1 are also positive in essen- cell / sclerosing rhabdomyosarcomas, suggesting "la; • .
tially all cases , although the number of immunopositive nuclei group may contain several subgroups. Congenital and 1""':r :
varies . Myogenin is usually diffusely positive in the alveolar type, spindle cell / sclerosing rhabdomyosarcomas usually c.crt
but the embryonal and spindle cell / sclerosing types may have rearrangements of NCOA2 and VGLL2, whereas those
only scattered positive cells. MSA and SMA immunoreactivity in older children and adults often show mutations in MYC.:-
is often present. Aberrant expression of keratins, S100, and {2354). Embryonal and pleomorphic rha bd o myosarc~ -
NFP has been reported in systemic examples, but few primary not show characteristic mutations or rearrangements. al ..
intracranial examples have been assessed for these markers. embryonal rhabdomyosarcomas often display who le-cr-:~­
A potential diagnostic pitfall for primary intracranial alveolar some gains , especially of chromosome 8.
rhabdomyosarcoma is that OLIG2 has been shown in systemic
PAX3::FOX01 or PAX7::FOX01 fusion-positive alveolar rhabdo- Essential and desirable diagnostic criteria
myosarcomas (2602}. Therefore, OLIG2 immunoreactivity in a See Box 8.04.
primary intracerebral alveolar rhabdomyosarcoma should not
be misinterpreted as representing the presence of a glial com- Staging
ponent. The Ki-67 labelling index is usually high. CNS rhabdomyosarcomas are not currently staged, b
their very limited numbers ; however. they almost always s:
Differential diagnosis clinically aggressive behaviour.
CNS metastases from systemic primary tumours must be
excluded /69 9}, as must parameningeal involvement by rhabdo- Prognosis and prediction
myosarcoma /3520}. Primary intracranial spindle cell sarcoma Prognosis is usually poor due to local recurrence. with ~4'\, S-'-
with rhabdomyosarcoma-like features and DICER1 mutations vival at 1 year {1995j ; long-term survival> 24 months 1s e:c:·--
(see Primary intracranial sarcoma, DICER1-mutant, p. 323) is tional (1418}. Metastasis occurs in< 20% of primary intrac~ .:.
now con sidered to be a distinct and separate entity (1677,2788}. examples (3603}.
Rhabdomyosarcomas must be differentiated from other brain
tumou rs that predominantly manifest other features but occa-
BoxB.04 Diagnostic criteria for rhabdomyosarcoma
sionally sh ow focal ske letal muscle differentiation. Examples of
primary CNS or peripheral nervous system tumours in which Essential:
th ere may be focal rhabdom yosarcomatous differentiation A malignant primitive tumour with at least focal immunohisttlci'lflmlieaJ
inc lude rare medulloblastomas with myoid elements (medullo- demonstration of skeletal muscle lineage
myoblastornas), glioblastornas with a sarcomatou s element AND
(i.e. g l1 osarcomas, especially postirrad iation exampl es), malig - Absence of non-rhabdomyosarcomatous components, a detailed In
nant peripheral nerve sheath tumours with a myoid com ponent
(rll3l1dn8nt triton tumou rs), and even rare mening iomas 11439 1. Desirable:
GF:rrr1 cell tumours with a prominent rh abdomyosarcomatous Confirmation of a FOXO 1gene fusion in diagnostically difficult cases (other lf\iJl
-- m or1t r1t n 1ay be difti cu lt to distinguish from primary intracer- alveolar rhabdomyosarcoma, in which such confirmation is essential rather lh!lll
f , 11 r!:dtJC1l)[ 1yosarcorna, 8bpeciall y when located in the pineal desirable)
lntracranial mesenchymal tumour , Kleinschm idt-DeMasters BK
Bouvier C
FET: :CREB fusion-positive Hainfellner JA
Perry A

Definition
tntracranial.me_senchymal tumour with FET: :CREB fusion (a pro-
visional entity) is a mesenchymal neoplasm arising intracranially
with variable histomorphology and fusion of a FET RNA-binding
protein family gene (usually EWSR1, rarely FUS) with a mem-
ber of the CREB family of transcription factors (CREB1 , ATF1,
or CREM).

ICD-0 coding
None

ICD-11 coding
2F7C & XH9362 Neoplasms of uncertain behaviour of connec-
tive or other soft tissue

Related terminology
Acceptable: intracranial mesenchymal tumour / angiomatoid Fig.8.19 lntracranial mesenchymal tumour, FET: :CREB tusion-posiiive. This 12-year-
fibrous histiocytoma. old boy presented with hemiparesis and headache and was found on neuroimag1ng to
Not recommended: intracranial myxoid variant of angioma- have a bulky left parietal tumour with heterogeneous signal features. The mass, as
toid fibrous histiocytoma; intracranial myxoid mesenchymal seen on T2-weighted axial MRI , is supratentorial, as are most examples of this tumour
type. On testing, an EWSR1::ATF1 fusion transcript was identified.
tumour with EWSR1::CREB family gene fus ions.

Subtype(s) Etiology
None The etiology is unknown, but no cases to date have been asso-
ciated with familial tumour predisposition syndromes.

I
.
Localization
These intracranial masses are more commonly located in Pathogenesis
supratentorial sites than in infratentorial sites. Most are extra- The cell of origin is unknown. These tumours harbour fusion of
axial, attached to the meninges or dura , or located intraven- a FET RNA-binding protein family gene (mostly EWSR 1) with
tricularly (2954). a member of the CREB family of transcription factors: CREB1
{3304,181,1552), ATF1 (1701,2873,1552,804}, or CREM (1701,
Clinical features 1039,181 ,1552}. One case with FUS:: CREM fusion has been
Tumours produce symptoms referable to mass effect and spe- identified {2954) .
cific location, including headache, nausea, vomiting, tinnitus , The relationship of these intracranial tumours to extracran1al
and occasionally seizures {3002} or focal neurological deficits . angiomatoid fibrous histiocytoma or to the many different types
Patients with anaemia {1230} or haemorrhage (2296) have been of extracranial tumours harbouring the same FET: .CREB fus ions
reported . (e.g . clear cell sarcoma of soft tissue , angiomatoid fibrous h1st1-
ocytoma, primary pulmonary myxoid sarcoma , hyalin1z1ng clear
Imaging cell carcinoma of the salivary gland , and gastrointestinal clear
Tumours are usually circumscribed extra-axial neoplasms with cell sarcoma) is uncertain .
attachment to the meninges or dura and compression of the
subiacent brain parenchyma . Other radiological characteris- Macroscopic appearance
tics include lobulated growth (often with both solid and cystic Examples have been described as partially encapsu lated ,
components). avid enhancement after contrast administration, focally haemorrhagic, tan brown, and focally gelat:nous i804l
1ntratumoural blood products , and substantial peritumoural
oedema Some demonstrate a dural lail or bony involvement of Histopathology
the overlying skull, mimicking meningioma 187,29541 Tumours demonstrate a wide morphological spectrum . usually
1nclud1ng a collagenous stroma with dense intercetlular matrix
Epidemiology highlighted by ret1culin staining Architecture ranges from syn -
Most cases occur 1n children or young adults, al:hr; uyh case::. in cvt1al or sr1eet-l1ke growth to reticular cord-like structures, and a
adults in their fifties and sixties have been rep ortPd 1 ' G.J1,1H4 subset ot tumours contain fibrous septa separating nodules of
1078.2954) . :urnour cells. f\lot all examples contain a myxoid stroma f2954J.
I,
I •• •
• "~ '
11
", _ • • :~ i.i iq > <~ e -' umours
1 inVO l\ltnQ the CNS 317
Tumour cell morphology varies from epithelioid/rh abdoid ce ll s to Dense lymphoplasmacyti c cu ff ing a l lhA ltJIT1()1Jr PC-!rif;h':r , ,
/ 1
stellate/s pindle cell s to monotonous round ce ll s. Mitoti c activity along fib rous septa an d haemos1cJe nn rJ r r1aAmatr;irJ1r1 ar""! r)t ii:. r
is generally low (typically< 5 mitoses/mm?). Haemangioma- like present. Addi tional morphologica l fea tures urn 1nr,li1rli:J m~r,,~: ­
collections of dilated thin-wall ed vessels are a frequ ent finding . goth elia l-like whor ls an d amianthoid -type fibres I 1F-i91 l'Jc,;. ,

Fig.8.20 lntracranial mesenchymal tumour, FET::CREB fusion-positive. A The morphological spectrum is wide. This example of an EWRS1:: ATF1 fusion-positive tumour
shows cords of small, cytologically uniform cells. B This example shows very prominent small vessels in an angiomatoid pattern . The tumour harboured an EWSR1::CREB fusion.
one of several types of characteristic fusions in this tumour type. C Architectural patterns in this tumour type include reticular, cord-like, spindled, and sheet-like: this examplewith
an EWSR1 ;:ATF1 fusion shows sheet-like architecture. D These tumours can show a wide range ot histological features, making the differential diagnosis broad in some cases.
This example with EWSR1::ATF1 fusion shows numerous clear cells .

.- .....' ,"'=,.
... . ·..
,-{
.-
. :f :~ ..·.. : ...~ :.:·:..=. ,.
·.. ;;.... I·. '":..
' .....,
I •"• ''.
• ,!
\ ·..;-. .....,,. ,.._ .. ,....
/.:. .. ·.. ~ ?' '"
. .."\.,. ... ,. •••: : f" Of I .. •,

• '
-- .
i ' .. ;·
J' • ' ' , ••
A .: .. ·. :.·
• • 1
,, 4 .~ ~ ' ' .. ~.· •• .. •

~ ·- -!'- .,., ,
... ' t~

. ,_ ... ' t

./ I' ,. • •
t "' ....

G. . .. ., r

'3 18
Tumours with EWSR1 :: CREB1 fusions more often have stellate/
spindle cell morphology, mucin -rich stroma , and haemangi-
orna-like vasculature , whereas tumours with EWSR1 :: ATF1
fusions are more commonly composed of sheets of epithelioid
cells with mucin -poor collagenous stroma 12954}.
The immunohistochemical profile is also variable , with the
most commonly reported immunoreactivities being for EMA ,
CD99, and desmin; all three are usually diffusely or focally posi-
tive, but in a minority of reports , CD99 or desmin were negative.
CD68 (8041. CD163 (87), and vimentin , when assessed , have
been positive. Variable positivity has been reported for synap -
tophysin , S100, and MUC4 12954).
Tumours have been negative for SSTR2A , OLIG2, GFAP, and
CAM5.2 {181); for myogenin , MYOD1 , and myoglobin 12296);
and for SMA (1552,1230), MSA , melan-A , HMB45 , MITF, nuclear
Fig. 8.22 lntracranial mesenchymal tumour, FET::CREB fusion-positive. Smear
STAT6, and CD34 {1230) . SMARCB1 (INl1) and SMARCA4
preparations from some examples may prompt diagnostic consideration of atypical
(BRG1) expression is retained 1181,1039}. Proliferation (Ki-67 teratoid/rhabdoid tumou r due to the rounded cytoplasmic profiles, prominent nucleoli,
labelling index) is generally low 12954). mitoses, and somewhat eccentrically located nuclei (at least 1n some cells). as 1n this
example. Fortunately, atypical teratoid/rhabdoid tumour can be excluded at the time of
Differential diagnosis permanent section if there is retention of nuclear immunoreactivity for SMARCB1 (INl1)
The broad morphological spectrum of these tumours and the and SMARCA4 (BRG1 ).
tact that their features overlap with those of other tumour enti-
BoxB.05 Diagnostic criteria for intracranial mesenchymal tumour, FET::CREB fu-
ties make diagnosis challenging {1039} . Only documentation of
sion-positive
a pathognomonic gene fusion provides diagnostic confidence.
The differential diagnosis is usually with sarcomas or menin- Essential:
gioma 187,10781. especially of chordoid, microcystic 11811. or Primary intracranial neoplasm
rhabdoid 11230} types . AND
Variable morphological features including spindle cells, mucin-rich stroma.
Cytology haemangioma-like vasculature, or epithelioid cells In a mucin-poor collagenous
Not relevant stroma
,. AND
t, ,' Diagnostic molecular pathology Demonstration or a FET::CREB family fusion
Diagnostic FET: :CREB family gene fusions may be detected
using FISH or DNA/RNA sequencing strategies . Demonstrat- Desirable:
ing EWSR1 rearrangement via break-apart FISH assay is not CD99, EMA, and desmin immunoreactivity
specific in isolation , and confirmation of a CREB family fusion
partner is recommended .
Prognosis and prediction
Essential and desirable diagnostic criteria The full spectrum of clinical behaviou r is not yet known, but it
See Box 8.05 . rang es from slow grow th to rapi d recurrences (2873,2296,1701 ,
2954) . Rarely, cerebrosp inal flui d dissemination or systemic
Staging metastases (including to pulmonary and thoracic lymph nodes)
Not applicable and bony metastases to spine have been seen {2954) .

~-h· ... ' 1L 1"·" 1 <ii r1<')!l rik r1111uothei1di lur ours rnv lving the CNS 319
CIC-rearranged sarcoma ( 1ri
r;rr Hf·
r)

r ,f 1 Jrrr f)
1r; r1 f lrAf V
(ri' J (j;; r

Definition Localization
CIC rear rnn ~wci se:11 ,01w1 o l 11 10 rwurn l rixis is fl h1gl1 grad8 , !hr~ rJ08rJ '/JI t1',','J<'~', II tr ~; /1: , r,
MrJ st (J(J(JI Jr 1r1 r '

poorly cirlfc'rc1111n !Prl smcoma cJo l111 d hy CIC fu sion with differ viscera (1?0 ~151\.'"~.'1(J5rJI 1nr,lurJ1rr3 tr c, ",r~r ','r • _ -
enl gorio pcir ln ors ancJ e1trn 81i8I UJrnrJ8r trrr8rit'", 1'3 r/-./~, ti/c=, 17 '; ,r 1,,
meta stasis rna'j 81S•J rf,r;ur '1 ?8~i
ICD-0 coding
93 7/3 CIC-rearranged arcoma Clinical features
Patients with CIC-rearran•J8d S8rr,rp·3 '",;:ir. rp=;' <'-;' /I
ICD-11 coding neurological def1c1ts or with gic:ib;:ill J r.:w:.wJ r •r -1r. -/ 2 - r
None sure. The symptoms are locat1rJn-dl'::pc::r·rJ-:;rt ;:i ,.. rJ ~ ·"=
effect.
Related terminology
Not recommended: CIC::DUX4, CIC:: NUTM1 sa rcoma ; CNS Epidemiology
Ewing sarcoma family tumour with CIC alteration. In a prospective reg istry, CIC-rearranged sarr:s,.,..,;;, tjc:~· • -=-~
using DNA methylation microarray accourti:;d 'ei r ~ .1t'=- · _
Subtype(s) newly diagnosed CNS tumours 1n patients aged -c 2· 1 ::..~ , ~
None There is a preference for adolescents and 'JOurg :J.d•_.. ·; -
older patients can be affected 1120 1808f

Etiology
Unknown

Pathogenesis
All CIC-rearranged sarcomas , irrespective of location un 'c,r~
contain an oncogenic gene fusion of CIC transcrrpt1o ra1 r':or;:.s
sor with various partners (most often OUX4, but also c:,,;1c.;
LEUTX, NUTM1, or NUTM2A) (1578 ,1424,1818.3066 3Go-
1371l . The t(4;19)(q35 ;q13) or t("10 ;19)(q26'q13) rar s rJ-:;::: · •
that results in CIC::DUX4 fusion in the ma1orrty of per1c"'~·::
tumours leads to the fusion of the C-terminus of CIC ~o ..... ~
N-terminus transactivating domain of DUX4 , and to the suo.e·
sion of the CIC transcriptional repressor function to one ria . .
activating {3003} . A subset of C/C::OUX4 cases contain .3 s•o::-
codon immediately after the breakpoin t, resulting 1n a en m-:::' ::
protein without a DUX4 sequence , suggesting that trLn:::ei·:::i
CIC may be sufficient for oncogenesis (1551 ,3542). CIC .-.J _ -

fusion leads to the pathological upregulation of norrr1::i.I ·arg~·:::


of CIC inhibition including the PEA3 family genes (e g .=~ ·
ETV4 , ETV5), CCND2, and MUC5AC (3003,1578 ,155 1 JJ..!
The majority of CNS CIC-rearranged sarcoma fusions are as.:2
ciated with NUTM1 (1818} .

Macroscopic appearance
The tumours are typically well-circumscribed, white or ran
masses, with frequent haemorrhage and necrosis .

Histopathology
CNS CIC-rearranged sarcomas display similar h1sto1CIJ c_:, ::?.\
Flg.8.23 CIC rea rranged sarcoma. A T2 weigh ted co ronal MRI shows a large cys-
tic tumour with several mural nodules 1n the right frontal lobe of a 35 - year--old worn· tures to those of their extra-CNS counterparts 11~0l T_, 1..'. :
a11 8 Tlie axial poslconl rast T1 -weighted 1111age shows enhancement of the capsule are composed of sheets of highly undifferen t1atec1 s J 1 • •• 'l
:111d de111011s11 at1011 of the solid components Nole tile stri ctly intra-axial localization of cells interposed with foci of necrosis , a var1J.bly l0bu\Jte1..' '='' '· rr
pattern , and desmoplastic stroma (774 , 1...\~t3 . 1818 3:i n
1
11tc le~1011 Tticse tu111uurs 111ay appear as cystic lesions or as solid masses .
.... . .........
"'.~ ~, ~~" ~~\;

Ftg.1.24 CIC-rearranged sarcoma. A A primary tumour in the cerebrum showing a well-circumscribed nodule m the brain parenchyma. B There 1s a diffuse proliferation of
small round cells with minimally pleomorphic nuclei and variably prominent nucleoli.

predominant round cell component may be interspersed with


epithelioid/spindle cell morphology. Myxoid chang e is common .
Cytological features also include prominent nucleoli and eosin-
ophilic cytoplasm . Tumours with CIC:: non- DUX4 fusion variants
Diagnostic molecular pathology
Suspected c ases require positive confirmation of CIC gene
fus ion eve nts. Break-apart FISH offers a simple approach
but does not inform on the binding partner and is affected by
I
.
.

have similar histology to that of CIC:: OUX4 tumou rs 11818,3066 ,


3067).

Grading
CIC-rearranged sarcomas are designated CNS WHO grade 4.

lmmunophenotype and differential diagnosis


CNS CIC-rearranged sarcomas can express patchy and weak
CD99; WT1 and ETV4 are frequently positive and NKX2-2 is
1yp1caily negative, which disting uishes CIC- rearranged sar-
coma from Ewing sarcoma . Scattered expression of cytokera-
un AE1/AE3, calretinin, a-SMA, and neurofilament has been
reported [1426,774,3507). Sarcomas with CIC:: NUTM1 fu sion
e1press NUT protein . Nuclear preservation of SMARCB1 and
SMARCA4 rules out atypica l teratoid/rhabdoid tumour

Cytology
Not clinically relevant

1'.l II ' I_/• \I l· r ' l t • 'lli l ~J ·. 'tll el d i lurnours involvino the CNS
I !• II
321
falsfl negative results ( 1424,35421 . NeY! generntion So/ttJ~r r,
19q13 2; ing of transcriptome (RNA sequen<:ing) or with .::Jnr,r-:.~~'l
c l Hl Ill_-i) multiplex PCR are practical yet sensitive appro~r)'·f!-; !1,..P '
1794 ,20051 . Detection of upregulat1on of ETV1 ET /4 ')r r:- r F.
Forward strand Forward strand complements FISH and RNA sequencing findmq5 11s5· 1 rt~
unique methylome (30591 permits the use of ON,11 mett"> f':,v·,;r
microarray profiling (16751 . RNA seq~ncmg and mett t~ ~~,
1aacaaaac ....aca~~~mai ___ ------ profiling is helpful in ruling out other tumours with ov~::iov·,..~
histology 13059) .
Exon 1-16 !
Exon 4-7
CIC:NUTM1
Essential and desirable diagnostic criteria
Rg.8.27 C!C-rearranged sarcoma. Schematic of chromosomal location, wildtype See Box 8 .06 .
RNA transcript, and exon structure resulting from a CIC::NUTM1 gene fusion.
Staging
Box 8.06 Diagnostic criteria for CIC-rearranged sarcoma Radiological survey and cerebrospinal fluid cy tology sh ~
be undertaken . There is no relevant staging system fo< CIC-
Essential:
rearranged sarcoma in the CNS.
Evidence of a CIC gene fusion
AND
Prognosis and prediction
Predominant round cell phenotype There is a lack of clinical data specific for CNS C/C-rearranget1
AND sarcomas; however, peripheral CIC-rearranged sarcomas ar".?
Mild nuclear pleomorphism characterized by a highly aggressive course that 1s marke 'I
AND worse than that of Ewing sarcoma (120,3543) , and they have
Variable admixture of epithelioid and/or spindle cells an inferior response to Ewing sarcoma chemotherapy reg

AND {120).
Variably myxoid stroma
AND
Variable C099 and frequent ETV4 and WT1 expression

Desirable:
DNA methylation pattern matching that of CIC-rearranged sarcoma

1
/i • I'• I 1 • " 1 1' •; 1!, J' .;t •.. ·'1 ti u Jr 11uu1 ~" 11wul 11no til e CNS
Prin1ary intracranial sarcoma, Solomon DA
Alexandrescu S
KoolM
Orr BA
O!CER1-mutant Foulkes WO
Haberler C
Pfister SM
Sturm D
Huang A von De1mling A
Kolsche C von Hoff K

Definition
Pnmary intracranial sarcoma , DICER1-mutant, is a primary intra-
cranial sarcoma composed of spindled or pleomorphic tumour
cells typically displaying eosinophilic cytoplasmic globules ,
1rnmunophenotypic evidence of myogenlc differentiation, and
occasionally foci of chondro1d differentiation . These tumours
are genetically defined by mutations in the DICER1 gene (either
somatic or germline as part of DICER1 syndrome) .

ICD-0 coding
9480/3 Primary intracranial sarcoma, DICER1-mutant

ICD-11 coding
None

Related terminology
Not recommended: primary intracranial spindle cell sarcoma
with rhabdomyosarcoma-like features , O/CER1-mutant.

Flg.8.28 Primary intracran ial sarcom a, O/CER1-mutant. Coronal T1-weighted post-


Subtype(s)
contrast MRI showing a heterogeneously enhancing mass with dural involvement and
None marked mass effect on the underlying brain parenchyma.

Localization 1836,2788,3391.1535) . Clear ethnicity biases have not been


The characteristic localization of these lesions is intracranial, reported to date, because of a lack of population-based studies .
often in supratentorial forebrain regions , and there is typically

I
leptomeningeal and sometimes dural involvement (1677,1836). Etiology
By definition, at least one pathogenic alteration in the OICER1 '

Clinical features gene located at chromosome 14q32 is present. These OICER1


Presenting symptoms include headaches, seizures, or focal neu- mutations can be either somatic or pr~sent in the germline as
rological signs representing the typical spectrum of clinical signs part of DICER1 syndrome {704,1677). An association with neu-
for tumours arising in the respective brain regions {1677,1836). rofibromatosis type 1 has also been observed {1836!. Therefore,
genetic counselling and germline testing may be warranted in
Epidemiology patients with this tumour entity. However, the exact incidence
The sex distribution has been reported as almost equal, and the of D/CER1-mutant primary intracranial sarcoma 1n DICER1 syn-
median age at diagnosis as 6 years (range: 2-76 years) (1677, drome and in neurofibromatosis type 1 remains to be elucidated.

..... .~ - .... -" ~ .... . . .


Flg.8.29 Primary intracranial sarcoma, DICER I-mutant. A These tumours frequently contain .cells .with prorrnnent eosinophilic cytoplas~ic globules. a These tumours are
typically composed of pleomorphic spindle cells arranged in fascicles or disorganized sheets. Brisk m1tot1c activity and eosinophillc cytoplasmic globules are common features.

L .8 '" l r•11 1< 11• non-men1ngotheltal tumours 11wolving the CNS 323
Fig. 8.31 Primary intracranial sarcoma, DICER1-mutant. A These tumours demonstrate immunophenotypic evidence of myogenic differentiation, most frequefltty w.01 SIZP.$-
sion of desmin as shown here. B These tumours demonstrate immunophenotypic evidence of myogenic differentiation, but this finding can often be focal rather than <iffuse. P.
example had desmin immunostaining in only a few small clusters of tumour cells. c While these tumours demonstrate immunophenotypic evidence of myogenic mrni!li!r.~Cll
with expression of desmin and SMA, they typically have limited or absent expression of myogenin, distinguishing them from rhabdomyosarcoma This example has ~
absence of nuclear staining for myogenin (a skeletal muscle-specific transcription factor), with only nonspecific background staining in the cytoplasmic globules.

Pathogenesis Histopathology
DICER1-mutant primary intracranial sarcoma is driven by DICER1-mutant primary intracranial sarcomas are matignar:
the genetic disruption of the DICER1 gene, which encodes a pleomorphic or spindle cell neoplasms often with tasc·
microRNA processing enzyme. The typical combination of or patternless growth , which may demonstrate myogenic -
events is a loss-of-function variant on one allele and one of a or cartilaginous differentiation {1677,1836,679}. Cyto
few recurrent missense mutations occurring in base pairs cod- eosinophilic globules and myxoid stroma are often pr
ing for metal-ion binding residues in the RNase lllb domain of (1836,1535}. These tumours typically have compact growth
OICER1 on the other allele (969,1677) . In addition to the dis- usually with involvement of the leptomeninges and dura bt.-
ruption of microRNA processing , the majority of these tumours they may also invade into surrounding brain tissue.
have d isruption of p53 signalling via inactivating mutations in
the TP53 tumour suppressor gene, ATRX mutation or deletion, Special stains and immunophenotype
an d activation of the MAPK signalling pathway via mutations in As with other sarcomas , there is abundant intercellular baserr:er.
KRAS, NF1, or PDGFRA (1677,1836,1535}. All tumours studied membrane deposition that can be highlighted with retJculir cc
to date have lacked the NAB2:: STAT6fusion that defines solitary collagen IV staining . The eosinophilic cytoplasmic globules star
fibrous tumou r, the PAX3::FOX01 and/or PAX7:: FOX01 fusions with PAS . The typical immunophenotype involves pos1tiv1ry C'
that characterize alveol ar rhabdomyosarcoma , and mutations in markers of myogenic differentiation (desmin . SMA. and occas:cn-
known genetic drivers of mening ioma (NF2, TRAF7, KLF4 , SMO, ally myogenin), which is often focal or patchy 11677.1836.7'05·
AKT1, SMARCB1) . The exact histogenesis and cell of origin are p53 expression is variable, and loss of ATRX expression occurs
unknown, as is the relationship with extracranial sarcomas of in a subset of tumours (1836). lmmunohistochemistry 1s typ1c
the kidney, uterine c ervix, and other sites that harbour DICER1 negative for GFAP, OLIG2, cytokeratins , EMA, 8100. SOX10. arc
mutation (966,3483.2049 ). SOX2 . These tumours frequently demonstrate nuclear pos1tro.i.
for TLE1 and loss of H3 p.K28me3 (K27me3) (72}.
Macroscopic appearance
O/CER1-mutant pr imary 1ntracranial sarcomas are typically uni- Differential diagnosis
focal, rr:lat1 .;ely 1,;11·t_:un 1scr1bed l e~1ons Tl1ey tend to be firm and Histologically, the resemblance to pleuropulmonary blasroma ~
often 1E d'l,re r.of·mor rr.oqL. striking . Because the brain is a known sanctuary for metas1at1c

3?4 •. F
Rg.1.33 Primary intracranial sarcoma, DICER1-mutant. A These tumours typically have a solid/compact growth pattern, but this example demonstrates invasion into the
underlymg brain parenchyma. B This tumour was located extra-axially and demonstrated meningioangiomatosis-like invasion through Virchow-Robin spaces into the subjacent
train parenchyma

I
pleuropulmonary blastoma 12085}, clinical history and imaging Box8.07 Diagnostic criteria for primary intracranial sarcoma. DICER1-mutant
studies are essential for ruling out pleuropulmonary blastoma
metastasis. Other tumours in the differential diagnosis based
Essential: .
'

Primary intracranial sarcoma


on location or histological features are anaplastic/malignant
AND
rnenin gioma, solitary fibrous tumour, gliosarcoma, and other
sarcoma subtypes such as rhabdomyosarcoma , fibrosarcoma, Pathogenic DICER1 mutation (either germline or somatic)
and synovial sarcoma. AND (for unresolved lesions)
DNA methylation profile aligned with primary intracranial sarcoma, OICER7-
Cytology mutant
Not relevant

Diagnostic molecular pathology Staging


The OICER1 mutations in these tumours are most often a hot- Staging is not clinically relevant to date , although spinal imaging
spot missense mutation in the RNase lllb domain on one allele and cerebrospinal f lu1d sampling should be considered
combined with a truncating mutation (frameshift , nonsense, or
splice-site) occurring in trans on the other allele. Some tumours Prognosis and prediction
harbour a single mutation accompanied by loss of heterozygos- The prognosis for patients with DICER 1-mutant primary intra-
1\y eliminating the remaining wildtype allele. OICER1-mutant pri- cranial sarcoma remains unknown, because only limited clini-
mary intracranial sarcoma harbours a DNA methylation profile cal data are available so far In one series of 22 patients, an
distinct from those of other CNS tumours !1677,1836]; however, aggressive clinical course was suspected, but long-term fol-
the epigenetic overlap with extracranial sarcomas harbouring low-up data were not suft1cient for reliable conclusions {1677}.
D!CER1 mutation is unknown . No prognostic or predictive factors have yet been reported,
and the prognostic relevance of tumours arising 1n the setting
Essential and desirable diagnostic criteria of gerrnltne DICER I or NF1 mutation has not yet been estab-
See Box 8.07. lished [704,1677,"1836)

M2 _e:- r,cn 1 r·1_: 1 riun 111er11119othelial tumours involving the CNS 325
Perry A Orr BA
Ewing sarcorna de Aiava E Par~ SH
Haberler C Sturm D
Jacques TS Yip S
KoolM Yoshida A

Definition
Ewing sarcoma of the nervous system is an extraosseous small
round cell sarcoma containing a fusion between one FET family
gene (usually EWSR1) and one ETS family gene (most often
FL/1) .

ICD-0 coding
9364/3 Ewing sarcoma

ICD-11 cod ing


2852.3 Ewing sarcoma of soft tissue
2852.Y Ewing sarcoma of bone and articular cartilage of other
specified sites

Related terminology
Not recommended: (peripheral) primitive neuroectodermal
tumour.

Subtype(s)
None

Localization
Roughly 12% of Ewing sarcomas are extraosseous tumours, a
small subset of which involve the craniospinal axis {1172}. Most
of the latter are meningeal (intracranial or spinal), paraspinal,
and/or peripheral nerve-associated masses, including those
that involve the cauda equina. Direct extension from adjacent
bone primaries can also occur. Fig.8.34 lntracranial Ewing sarcoma. A T2-weighted MRI of a dural-based so ma53
lesion in the right middle cranial Iossa, about 55 x 75 mm in size. showing heterogeflecbs
signal intensity. The mass effect on the right temporal lobe with impending transten..'0-
Clinical features rial (uncal) herniation is observed. Probable extra-axial location extending into the
Signs and symptoms vary with location and are typically due to temporal lobe with dural tear and adjacent parenchymal oedema was suspected. I CT
mass effect. They include localized pain , cranial/radicular neu- image reveals the multifocal calcifications and haemorrhages within the tumour. C The
ropathies, bone fractures , and/or fever, the last being more fre- mass lesion is mainly hypointense with multifocal hyperintensity on T1-weighted imag-
quent in patients with metastatic disease. Imaging studies are ing. D Contrast-enhanced T1-weighted imaging demonstrates multifocal enhanc:emern
necessary for defining the site of origin, extent of local disease, within the tumour and the medial displacement and thickening of the right temporal
and metastatic spread, but they do not otherwise show specific
diagnostic findings.

Epidemiology
Ewing sarcoma is most common ly encountered in children and
young adu lts, with older patients more commonly presenting
w1tt1 GJ(traosseous disease, including in the nervous system .
f-'u•s811ta!1on beyon d 50 years of age is rare {1443) .

t::.tiology
he 11Gpr11 of t: :v11,g sarcomas are sporadic and idiopathic,
,,[r· · i_,,, '.J t '-·"·tf111JIP~ have oeen reported in patients with
r
y.-·rrn n~ / i • ._?, PMC:,'), or RF. mutations : wheth er these were
1'ci1,,,,·- l• c c·1 ,,;ki flln1 1::, mclear (35961
A .
fll. l .31 Ewing sarcom~. A The focal Ho.mer ~right {neuroblaslic) .rosettes in this tumour are a sign of neuronal differentiation. B Sheets and nests of pnm11ive-appearing
small round cells with ~ehcate. chromatin •. high N.C ratios. and high m1tot1c count. C Cauda equina lesion. Native nerves are entrapped 1n this small round cell neoplasm. D A
rare form of neuronal d1fferent1at1on in Ewing sarcoma 1s ganglion cell maturation.

pathogenesis . . Histopathology
Ewing sarcomas uniformly conta1~ an onco.genic gene fusion Classic Ewing sarcoma ls com posed predominantly of mono-
ttiat combines a FET RNA-b1nd1.ng protein fam ily member morph ic . primitive-appearin g , and m1totically active small
(mostly EWSR1 and rarely FUS) with an ETS transcription fac - round cells arranged in sheets . The chromatin 1s delicate and
tor family member (FL/1 > ERG » ETV1 , ETV4 [E1AF] , and there are smal l amounts of c lear to amphophil1c cytoplasm .
FEV) p172J . The t(11;22)(q24 ;q12) translocation that results in wh ich is often glycogen -rich (PAS -positive and diastase-sen-
the EWSR1::FLl1 fusion transcript and protein product accounts sitive) . Like other sarcomas, Ewin g sarcoma 1s also ret1culin-
for roughly 85% of all cases. Additional STAG2 (in 15- 22% of rich . Evidence of neuronal differentiation most commonly man-
cases). CDKN2A (12%), and/or TP53 (7%) mutations are occa- ifests as Homer Wr ight rosettes with central neuropil (cases
sionally found , and they may be associated with a worse prog- previously referred to as "periphera l prim1t1ve neuroectodermal
nosis (382.3201 ,11721. tumour "), although rare cases may show ganglion cell differen-
tiation {3413 ). Postchemotherapy specimens are often exten-
Macroscopic appearance sively necrotic .
Ewing sarcoma has a soft. grey, fleshy appearance, often with
foci of necrosis and haemorrhage on the cut surface. Nerve Grading
roots may be entrapped within the tumour, especially in the Ewing sarcoma is considered CNS WHO grade 4.
cauda equina.

..•"' ... ...


., ... \ ,

.: . "' ' ....


D Scattered

327
Table8.01 Useful immunostalns for the diagnostic workup of Ewing sarcoma

lmmunostafn niltlvlty Specificity


e1Um1t eat1m1te
CD99 (diffuse membranous pattern) Nearly 100% 87% Can be positive in desmoplastic small round cell tumour, alveolar/emb<y0nal
rhabdomyosarcoma, and small cell osteosarcomi3

PAX7 (nuclear) Can be posltive in EWSR1 ::NFATC2 sarcoma, alveolar rhabdornyosarcoma. pr)()l'\y
Nearly 100% 88% differentiated synovial sarcoma, BCOR::CCNB3 sarcoma, small cell OS1eoearrmra.
and desmoplastic small round cell tumour
Can be positive in small cell carcinoma or neuroendocnne tumours, olfactory
NKX2-2 (nuclear) 93-100% 85-88% neuroblastoma. neuroblastoma, mesenchymal chondrosarcoma. CIC DUX sar':l}lr;;
synovlal sarcoma, and melanoma

lmmunophenotype and differential diagnosis


The three most useful immunostains for differentiating Ewing
sarcoma from other round cell sarcomas are summarized in
Table 8.01 11971,1615,3208). Otherwise, lineage markers are
mostly negative, except neuronal markers, which are variably
positive depending on the extent of neuronal differentiation.
Cytokeratin may be expressed focally. The differential diagnosis
for cases with Homer Wright rosettes and/or ganglionic differ-
entiation incl udes peripheral neuroblastoma, although patients
with Ewing sarcoma are typically much older and neuroblas-
tomas are virtually always CD99-negative. In contrast, the dif-
fuse CD99 positivity encountered in mesenchymal chondrosar-
coma is an occasional pitfall , but hyaline cartilage is seen only
in mesenchymal chondrosarcoma, as is the pathognomonic
HEY1 :: NCOA2 gene fusion . CD99 can also be expressed
in other tumour types, including solitary fi brous tumour and Fig.8.38 Ewing sarcoma of the dura. Locus-specific identifier (LSI) EWSR1 gen-:
atypical teratoid/rhabdoid tumour, as well as other embryonal (22q12) dual-colour break-apart FISH shows separated SpectrumOrange and Soectrurr-
CNS neoplasms. Solitary fibrous tumour is distinguished by Green signals in addition to a fused signal (normal) in the nuclei of Ewing sarcoma cells

1 1 - - - - - - - - - •n• - - - - - - - - - - ; 1 - - - - - - - - - ""•--
- -
.- ..- - - - - _ - - , ....... 1
.....
I
~-
I
~ .. -..a.. ..
I I
~- .......... .. a.aa.-.. a..u.IW .. a.t&U9 .. ~
I 1

_.....,_,_
...._._._, _ .... . Altadt\mll- ~

----------------~.;.,,"':';11 •

:.;= ==--:.t.• tuhl-1'...


Clpol4f>Ot
a,,... . ..... . ....
. . .. \t . . . . . . -)'et
MAL• lllrt • Ollftl.llllnS~ (~
Fn"',_
.....
-04---
IC •TctG!>O<i4

"'""°'
ZX -lt .. O
n- •n.o
.......
l.M.MGn - tllf"Zt.ll.tu.1'.l
. . . - A•W41

Fifi 8.3 L::11ng c;c,rc:oma !:WSR l .. FU1 fusion in the Integrative Genomics Viewer (IGV). The red bar on the chromosome track (below 29 683 900 bp of chromosome 22 and
or;:ow 1?86/1 400 t:p 0t l.hromo::.0 .1e 11J reµresents a breakpoint. The red reads are positive strands, and the blue reads are negative strands. The mated reads exlsl on a cbJ
ft'ru,1 ..J1r._r1o r r·e 'd,romrisome 11) Jpun rnouseover on chromosome 22 (H52V5CCXY:7:1212:288771:0).

, 1 :: 1 . 1• 1• 11 1• r 1'1;)1 , ... ,·I 11 1 111r .• ' '" J'\111r1 ll1E' Cf\] ~,


.,
1
a.oe Diagnostic criteria for Ewing sarcoma fusion - positive; myoepithel1al neoplasms) For example ,
EJsentlBI: CIC-.DUX4 fusion is frequently found in EWSR1 fus1on-negat1ve
SmaU round cell morphology small round cell sarcoma 13003.120) Also , EWSR I fusion has
been identified in other intra-axial CNS tumours, 1nclud1ng gan-
AND glioglioma and ependymoma /2928 ,34701. Given the sometimes
[)ifluse CD99 membranous expression
limited sensitivity of FISH 15411 and the diversity of gene fusion
AND partners and junctional breakpoints , next-generation sequenc-
FET::ETS fusion ing-based approaches are considered more efficient, compre-
hensive, and 1nformat1ve for fusion drivers.
Desirable:
NKX2-2 expression Essential and desirable diagnostic criteria
pAXf expression See Box 8.08 .

Staging
rts nuclear STAT6 immunoreactivity, whereas atypical teratoid/ There is no relevant staging system for Ewing sarcoma in the
mabdoid tumour shows a loss of SMARCB1 (INl1) expression CNS .
or. rarely, of SMARCA4 (BRG1) expression .
Prognosis and prediction
Cytology Patient outcome greatly improves with induction chemotherapy
Not relevant followed by tumour resection and rad iation therapy 11172,1615).
Nonetheless, roughly a quarter of cases come to clinical atten-
Diagnostic molecular pathology tion with metastatic disease and this is the strongest negative
Most Ewing sarcomas of the nervous system require molecular prognostic variable; the estimated 5-year overall survival rate
confirmation of a FET: :ETS-type gene fusion for definitive diag- in patients with localized disease is 70-80% , but it decreases
nosis. With classic histopathological features , a positive EWSR1 to about 30% in those with disseminated disease . In localized
break-apart FISH assay may suffice, but potential mimics with disease, complete response to induction chemotherapy (0%
other EWSR1 fusions must be excluded (e.g. desmoplastic small viable tumour in the posttherapy specimen) is associated with a
round cell tumour; intracranial mesenchymal tumour, FET::CREB favourable prognosis (57) .

I
.

·' fl ' '"' 11q ~t · · I tit rn oi.;;·s llWOI 1ny t .e C NS 329


I~
Kle1nschm1dt-OeMasteri; BK
Mesenchymal chondros rcoma Baumhoer D
Bouvier C
Flanagan AM
Hainfellner JA
Inwards CY

Definition Etiology
Mesenchymal chondrosarcoma is a rare, biphaslc, malignant Unknown
tumour composed of undifferentiated small, round or oval to
spindle -shaped cells and islands of well-di fferentiated hyaline Pathogenesis
cart ilage. The presence ot a HEY1 :: NCOA2 gene fusion is char- Almost all mesenchymal chond ro sarcomas show the · qr !
acteristic. specific HEY1 :: NCOA2 fusion transcript.

ICD-0 coding Macroscopic appearance


9240/3 Mesenchymal chondrosarcoma Typically, the tumours are lobular, grey-brown , firm rias&=--
{1787,2771 ,3314} with varying amounts of calcification 15421 ,
ICD-11 coding
2850.Z & XH8X47 Chondrosarcoma of bone and articular car- Histopathology
tilage of unspecified sites & Mesenchymal chondrosarcoma Mesenchymal chondrosarcoma shows primitive small O>Jro
cells intermingled with islands of well-differentiated !°l'fal "'e
Related terminology cartilage. The absence of the latter in core needle b1op&es
None can render the diagnosis particularly challenging 11898\. c 1S
examples show more spindling and less necrosis than muse to-
Subtype(s) skeletal tumours {888}. A staghorn , solitary fibrous tumour- ilce
None vascular pattern is often (but not invariably) present. Varymg
amounts of central calcification and/or enchondral oss1ficaIJcr
Localization of cartilaginous islands with eosinophilic osteoid-like matrrx are
lntracran ial location {18 98} is more frequent than intraspinal seen.
{53 5), and th ese are followed by frontoparietal and thoracic
cord locations. Approximately 60% have a dural attachment lmmunohistochemistry
{1898). The primitive small round cells can show expression of CD99
' Clinical features
EMA , desmin , myogenin , MYOD1 and NKX3-1 . whereas i fN
are negative for GFAP, keratins , SMA , and ER \888.953.3545l
Symptoms relate to intracranial mass effect or spinal cord com- SMARCB1 (INl1) is retained and Ki-67 labelling is generaily
pression {535). increased. 8100 and SOX10 can be positive in both comoc-
nents (889,3406}. ·
Epidemiology
Most mesenchymal chondrosarcomas arise in the second and Differential diagnosis
third decades of life, although patients at either end of the age The main differential for mesenchymal chondrosarcoma in '.~~
spectrum have been reported {2771,3314 ,1898). CNS is solitary fibrous tumour. The presence of hyaline carnlag-.?

fig. ll.,~Cs r.'.vs 3 • ,t1 " .:1 ' ~ •


1
A /,x,al CT shows a parttally calcified right paraspinal soft ti ssue mass that extends into the ad1acent neural foramen. B T2-we1ghte\1
~. '~ ' 1'~ :r,f '"' -;: ~ '.yµ.,-•i : C:·~n 1parn,o with adpcent paraspmal musculature and contains an irregular, very hyperintense necrotic centre. C Tl -weighted ax:ai
1
• -::.,..- ;r ':E:
;.: _!::-:, 'r~·-· ~-H-,i :hJ;;~ c..11 1r.·v 1 ~t·~ t:rt:dnc1ng 1111~ ot tumour surro unding a central non·enhancing necrotic core .

.. ,
flt.l.41 Mesenchymal chondrosarcoma. A Mesenchymal chondrosarcoma is a highly cellular biphasic tumour composed of areas of undifferentiated round cells with high N:C
ratios. as well as numerous staghorn vessels, and islands of hyallne cartilage. B Mesenchymal chondrosarcoma typically shows a blphasic pattern of small malignant cells with
scant cytoplasm and islands of hyaline cartilage, both seen here.

SOX9 positivity and HEY1: :NCOA2 fusion in the former versus


nuclear STAT6 expression and a NAB2:: STAT6 fusion in the lat-
ter usually allows an unequivocal diagnosis. Areas of ossification
and/or calcification of the cartilage component in mesenchymal
chondrosarcoma can mimic an osteosarcoma (either small cell or
chondroblastic) . On neuroimaging , dural-based examples most
often mimic meningioma (2787) . Histologically, meningioma with
metapfastic cartilage is distinguishable by lower-grade features ,
a meningothelial/fibroblastic architecture, and positive EMA and/
or SSTR2A immunolabelling. Other small round cell tumours ,
including Ewing sarcoma, monophasic synovial sarcoma, rhab-
domyosarcoma, and atypical teratoid/rhabdoid tumour, may
enter the differential diagnosis, particularly when there is insuf-
ficient hyaline cartilage in a small biopsy (1898 ,3254}

I
..
Flg.8.42 Mesenchymal chondrosarcoma. The small , poorly differentiated cells ex-
Cytology press diffuse cytoplasmic CD99.
Tumours show oval to spindled cells with high N:C ratios and
BoxB.09 Diagnostic criteria for mesenchymal chondrosarcoma
hyperchromatic nuclei . The cytoplasm may be vacuolated and
cells may be associated with myxoid stromal material and/or Essential:
necrotJc debris. lntraoperative smear preparations may feature Poorly differentiated tumour composed of small blue round cells with high N:C
a prominent perivascular arrangement of cells (3324). ratios and variable amounts of hyaline cartilage
AND (in cases lacking cartilage)
Diagnostic molecular pathology Demonstration of the characteristic fusion transcript (HEY1 :: NCOA2)
Mesenchymal chondrosarcoma typically harbours an und erly-
ing HEY1::NCOA2 gene fusion.
Prognosis and prediction
Essential and desirable diagnostic criteria Spinal examples show low re currence rates, and most patients
See Box 8.09. survive for> 2 years after diagnosis (535}; intracran1al examples
have a high rate of rec urrence and (rarely) can show leptome-
Staging ningeal disse mination (1994 I or metastasize outside the cranial
Not relevant vault [2754,1748 1.

1·.,.1 , .i::r1. ·., ,r ii ri rJr -m8 11ngothel1al tumours 1nvolv1ng the CNS 331
Chondrosarcon VIP.in :>r,r1m1dt [J~MF!':IP.r 1 8V
ou·11er r~
~ lt1nRq;:in MA
H-.i1nfCJllner .JJ.'
lnw;:irrJ c; C (
Rosi=.inr1Aff..J .A F.:

Definition
Chondrosa.rcomas a~e a family of malignant mesenchymal
tumours with cartilaginous differentiation, comprising conven-
tional central, dedifferentiated central, conventional peripheral
deditferentiated peripheral , and clear cell chondrosarcomas . '

ICD-0 coding
9220/3 Chondrosarcoma
9243/3 Dedifferentiated chondrosarcoma

ICD-11 coding
2850.Y Chondrosarcoma of bone or articular cartilage of other
specified sites
2850.Z & XHOFYO Chondrosarcoma of bone and articular car-
Flg. 8.43 Chondrosarcoma Tl-weighted (A} and T2-we1ghted 1 8) -:oronaJ
tilage of unspecified sites & Atypical cartilaginous tumour / 33-year-old man who presented with dysphag1a shows a skull base i8s1on that ::r'!t'-
chondrosarcoma, grade 1 sively involves left-sided basilar skull regions (nght side of images\.
850 .Z & XH6LT5 Chondrosarcoma of bone and articular carti-
lage of unspecified sites & Chondrosarcoma, grade 2
2850 .Z & XHOY34 Chondrosarcoma of bone and articular carti-
lage of unspecified sites & Chondrosarcoma, grade 3

Related terminology
None

Subtype(s)
None

Localization
Conventional chondrosarcoma is the most common tumour
type that arises in the cranial bones. The most common sites
are the skull base (spheno-occipital and sphenopetrosal syn- Flg.8.44 Chondrosarcoma. A CT of a 56-year-old man who presented with atai fll
chondroses). spine, and sacrum . Parafalcine examples are weakness demonstrates a heavily calcified skull base mass near the prepontme ~
uncommon; parenchymal intracranial, meningeal, and extraos- basal cisterns, behind the sella. Preoperative considerations included cran
oma and chondrosarcoma. Biopsy proved that the lesion was a low-grade -:t'l()(lCf()Sa'-
seous examples are rare. Peripheral chondrosarcoma is excep-
coma. B T1-weighted postcontrast coronal MRI of a 56-year-old man with a •
tionally rare at these sites. low-grade tumour shows the characteristic heterogeneous signal seen wittun the

Clinical features
Patients with chondrosarcoma present with a painful enlarging increased risk of developing chondrosarcoma, possibly assoc.-
mass . Neurological symptoms are site-dependent; skull base ated with pre-existing enchondroma. Individuals witn muit .e
examples may produce cranial nerve palsies . osteochondromas arising in association with germline E.XT C'f
EXT2 mutation have a greater propensity to develop a secooo-
Epidemiology ary peripheral chondrosarcoma 13312).
lntracranial chond rosarcomas comprise approximately 1% of all
chondrosarcomas !781]; in this site, they are less frequent than Pathogenesis
U1ordoma !1644). In one study of 200 patients with skull base Approximately 60% of central cartilaginous tumours r.ar
turr:c:Jurs, 1he age range was 10- 79 years (mean : 39 years), and an IOH1 or IOH2 mutation , the former being considerably
:\.r-; M·i- rat1ri w8.S 1.1 3 (2724). common . When this alteration occurs as an earl postzygc ~
mutation , it causes mosaic disorders including Oiiier 015CdS2
f·t1ulogy and Maffucci syndrome. The rate of IDH mutation is n1gr r
l/ 1 J ,, 1 , 1 11 ·.d.vJ 1rc .\,rf':'is are sporadic Individuals with enchon - skull base chondrosarcomas. whereas hondrosarcomds JI J' ·
'-' 1r,,/c <,1<, rr Jll1P1 rJ1',GdSe, Maffucci syndrome) have an facial skeleton lack IDH mutations [3118). The addinonal J · '
3 1tera t1ons
in conventional chondrosarcoma are similar to those lmmunophenotype . .
!DH-r utant and IDH -wi ldtype tumours and in central and
•="' . ·1 for S100 and 02-40 (podoplarnn) is typical ·
Immunoreac t 1v1 y .
penpheral chondrosarcoma (3426) . and immunoreactivity for ERG is possible; kerat1.n and brachy-
ury stains show no positivity. There is a goo d antibody for IDH1
0
Macroscopic appearance p.R132H , but this variant represents < 20 Yo of IDH mutations
-:nondrosarcomas are glistening, grey, firm lobular masses that in cartilaginous tumours. lmmunostaining can be abrogated by
are usually non-haemorrhagic but may have more mucinous harsh decalcification. Dedifferentiated chondrosarcomas may
areas reflecting myxoid change microscopically; skull base show loss of H3 p.K28me3 (K27me3) , but only in the dediffer-
umours are usually resected piecemeal. entiated areas [1990) .

Histopathology Proliferation and grading


·c rosoopy Proliferative activity is low in grade 1 neoplasms and high in
Hyabne-type conventional chondrosarcoma grows with an infil- grade 3 and dedifferentiated neoplasms. The gradin~ crit~ri a
tratJYe pattern. It is moderately cellular and composed of poly- from the Soft tissue and bone tumours volume of this series
hedral cells residing in lacunar spaces with in a solid basophilic can be used: for grade 1, nuclei are generally uniform in size,
"1atlix. In myxoid conventional chondrosarcoma , the tumour and binucleation is frequently seen . but mitoses are absent,
ceHs are spindle and stellate, and they float in a basophilic, floc - for grade 2, the re is increased cellularity, a greater. degree ?f
culent, mucinous matrix. The tumour cells are small to medium nuclear atypia, hyperchromasia , increased nuclear size, myxo1d
size. They do not form cohesive nests or aggregates, but matrix change, and the presence of mitoses; for grade 3, the
etongate cytoplasmic processes of adjacent cells may come tumour is highly cellular and more pleomorphic , mitoses are
into contact with one another and form complex interconnecting easily found, and the cells at the periphery of the mostly myxoid
'letworks . In a study of 200 skull base chondrosarcomas (the tumour lobu les are spindled and less differentiated . As noted in
largest such study to date), > 60% showed mixed hyaline and th is same reference, however, "histological grading is subject to
fTJYXOid features (2724). interobserver variability".
Grading schemes of conventional chondrosarcoma (CNS
WHO grades 1-3) are based on the degree of cellularity, Differential diagnosis
cytological atypia, and mitotic activity. Most skull base chon- Chondrosarcoma can be distinguished from chordoma by the
drosarcomas are low-grade (CNS WHO grade 1) [2724}; CNS absence of cohesive nests of cells , and the neoplastic chon -
0 grade 3 tumours are uncommon at this site {3118}. Dedif- drocytes are negative for cytokeratin and brachyury; approxi-
IErentiated chondrosarcomas often show abrupt transition mately 6% of chondrosarcomas are positive for EMA . SOX9 is
from a low-grade to a high-grade phenotype, usually with the common to both notochordal and cartil aginous differentiation
latter showing features of a pleomorphic spindle cell sarcoma, and is therefore not useful in the chordoma-chondrosarcoma
although osteosarcomatous or other lines of differentiation can differential diag nosis [2294} . The presence of osteoid matrix

I
.
occur. indicates a diagnosis of osteosarcoma, although a diagnosis of
a dedifferen ti ated chondrosarcoma must also be entertained .

~ J '/ -·
Ag.a.cs Chondrosarcoma. A Chondrosarcomas of the skull base show moderate hypercellularity, ind1v1dual turnour chond;ocytes wit·n· ·id .-.I
. . . .
h h .
m1 nuc ear yperc romas1a, an
d
acsence of m1tot1c acuv1ty. B This skull base chondrosarcoma shows the typical strong diffuse 1rnmunoreacliv1ty for s1oo in tumoural cel ls b th t d. .
. . . . , o 1n cy op 1asm an m nuc1e1.
Strong 1mmunosta1rnng for S100 can also be found 1n chordomas ot the skull base, chordoma often Is the main tumour type in the di1ferenti·al d S . ..
. . 1agnos1s so 100 1mmunostam1ng
d.oes not hetp d1stmgu1sh these two tumour types. '

M 2-:.e ... 1u · :l 'nal . non-mernngotheltal tum · 33


ours 1nvolv1n g the CNS 3
ct1ordoma Varlet P
Nielsen GP
Righi A
Tanaka S
Tirabosco R

Definition Localization
Chordomas are a family of p~imary ~~lignant bone neoplasms Chordomas almost always arise within the axial skeleton. par-
demonstrating notochord.al d1ff~rent1at1on , co.mprising conven - ticularly in the skull base and the sacrococcygeal region . The
tional, chondroid , poorly d1fferent1ated, and ded1fferentiated types. anatomical distribution varies depending on age {28761 and
histopathological type (detailed in Table 8 .02). Extra-axial loca-
ICD-0 coding tions are exceptional {2676,3200}.
9370/3 Chordoma
Clinical features
ICD-11 coding Patients with chordomas most commonly present with pain and
295y & XH9GHO Other specified mali g nant mesenchymal neo- site-related neurological symptoms .
plasms & Chordoma, NOS
ssY
2 & XH1708 Other specified mali gnant mesenchymal neo- Imaging
plasms & Chondroid chordoma Chordomas are lobular, lytic, destructive midline lesions, hypoin-
2B5Y & XH7303 Other specified malig nant mesenchymal neo- tense on T1-weighted MRI and hyperintense on T2-weighted
plasms & Dedifferentiated chordoma MRI, with enhancement on postcontrast imaging.

Related terminology Epidemiology


None Chordoma has an incidence of 0.088 cases per 100 000 per-
son-years , representing 0.5% of all primary CNS tumours {680) .
Subtype(s) Sex and age predominance accord ing to histopathological type
None are detailed in Table 8.02.

Table8.02 Characteristics of histopathological types of chordoma

Poorry differentiated chonk>ma,


Conventional chordoma Chondroid chordoma Dedifferentiated chordoma

I
SMARCB1-deficient
Adults (96%) Adults (86%) Adults (96%) Children (86%)
.
Age at diagnosis
Median: 55 years Median : 45 years Median: 61 years Median: 7 years
M:Fratlo 1.7 1.1 1.8 0.7
Prior irradiation No No Yes (25%) No
Localization Sacrococcygeal region (55%) Skull base (73%) Sacrococcygeal region (60%) Skull base (64%)
Conventional juxtaposed with
sarcomatous (91 %)
Chondroid juxtaposed with
Epithelio1d
Hlstopathology Classic Chondroid sarcomatous (2%)
No physaliphorous cells
Conventional chordoma
transformed into pure
sarcomatous tumour (7%)

SMAACB1 (IN11 ) preserved SMAACB1 (INl1) preserved SMAACB1 (INl1) preserved Loss of SMARCB1 t1Nl1)
Brachyury+ Brachyury+ Brachyury+/- • Brachyury+
lmmunohistochemical
profile Pancytokeralin+ Pancy1okeratin+ Pancy1okeratin- Pancytokeratin+
EMA+ EMA+ EMA- EMA+
S100+ S100+ S100-/+ SIOO+/-

Metastasis: 13% Metastasis: 9% Metastasis: 30% Metastasis: 30%


Local progression : 46% Local progression: 54% Local progression · 65% Local progression: 54%
Outcome Median PFS: 26.5 months Median PFS : 6 months Median PFS : 4 months
Median PFS: 24 months
Death during follow-up: 29% Death during follow-up: 42% Death during follow-up· 61 % Death during follow-up: 43%
Median OS : 48 months Median OS: 43 months Median OS : 15 months Median OS: 13 months
·oimmunopositive; -, immunoneg~tive· OS overall su~lval· PFS progression-free survival.
:P
s1t1ve in th . · • · ·
Not . e conventional or chondroid component. negative in the sarcomatous component.
0
57 ~ · .ata are based on the systematic review of 245 conventional chordomas (main references 13349.3144)\, 210 chondrotd chordomas (main refe<ences: {2876,3144}),
editterenliated chordomas (main references : [ 158 l ,3l 44)) and 65 poorly d1fferem1att>d chordomas 1ma1n ;e;arences . (1258 2912})

·) '·u' : 1 ·· rt :e r .1noothelial tumours involving the CNS 335


Box8.10 D1agnost1r. c11IE111n IN thoncirosA1coma
Diagnostic molecular pathofogy
Esstntl I. 101-(f or /0112 mu lat inn 1s prec;Anl 1n .::ipprr1Yim;::) t'3ly F. . 0 ..1., 0f r.~r -
A hlstolog1cally mal gnant tumour with cartilaginous dlfleren11atlon tral convent1onr~I 8nrJ cJed1fferent1Bted r,hrJr1rJrrJ!:iarr:rJm:1'"1
AND
lmmunoh1stochem1cal profile characteristic of chondrosarcoma
Essential and desirable diagnostic criteria
See Box 8 .10.

Staging
Cytology
Not applicable
C tolog rarely used because it 1s difficult to access skull
1s
base lesions with a needle, but when it is used, discohesrve Prognosis and prediction
round o elongate cells in a hyaline or myxoid matrix are seen . Most skull base chondrosarcomas are primary and low-grac~
The degree of cellularity and atypia varies with grade. In rare but they are locally destructive, requiring adjuvant therao1 fr;1t,y
in stances of high-grade tumours , examples may show necrosis proton beam therapy or ra d iosurgery). Metastases are rare
and pleomorph1c cells {1841 /.

3'.34 l ,,,,. I ' t ' ' I \I ' I ( . II . I I . .


, - - ••• · · ' • 1 '· • ,,,,_.,., !<"'d1 !1J' ! 'tJ1 1r· . . , ,,,, v1riq ll1fJCNS
hordoma Varlet P
Nielsen GP
Righi A
Tanaka S
Tirabosco R

Definition Localization
omas are a family of primary malignant bone neoplasms Chordomas almost always arise within the axial skeleton , par-
nstrating notochordal differentiation, comprising conven- ticularly in th e skull base and the sacrococcygeal re gion . The
nonal. chondroid, poorly differentiated, and dedifferentiated types. anatomical distribution varies depending on age {2876) and
histopathological type (detailed in Table 8.02). Extra-axial loca-
ICD-0 coding tions are exceptional {2676 ,3200) .
370/3 Chordoma
Clinical features
JCD-11 coding Patients with chordomas most commonly present with pain and
285Y & XH9GHO Other specified malignant mesenchymal neo- site-related neurolog ical symptoms .
plasms & Chordoma, NOS
285Y & XH1708 Other specified malignant mesenchymal neo- Imaging
plasms & Chondroid chordoma Chordomas are lobular, lytic, destructive midline lesions, hypoin-
285Y & XH7303 Other specified malignant mesenchymal neo- tense on T1 -weighted MRI and hyperintense on T2-weigh ted
plasms & Oedifferentiated chordoma MRI, with enhancement on postcontrast imaging .

Related terminology Epidemiology


None Chordoma has an incidence of 0.088 cases per 100 000 per-
son-years, representing 0.5% of all primary CNS tumours 1680).
Subtype(s) Sex and age predominance accord ing to hlstopathological type
None are detailed in Table 8.02.

Table8.02 Characteristics of histopathological types of chordoma

Poorly differentiated ehordbma,


Conventional chordoma Chondroid chordoma Dedlfferentlated chordoma

I
SMARCB1-deficlent
Adults (96%) Adults (86%) Adults (96%) Children (86%) '
Age at diagnosis
Median : SS years Median: 4S years Median: 61 years Median: 7 years
ll:f ratio 1.7 1.1 1.8 0.7
Prior Irradiation No No Yes (25%) No
Localll.ation Sacrococcygeal region (S5%) Skull base (73%) Sacrococcygeal region (60%) Skull base (64%)
Conventional juxtaposed with
sarcomatous (91%)
Chondroid juxtaposed with
Histopathology sarcomatous (2%) Epithelioid
Classic Chondrold
Conventional chordoma No physaliphorous cells
transformed into pure
sarcornatous tumour (7%)
SMARCB1 (INl1) preserved SMARCB1 (INl1) preserved SMARCB1 (INl1 ) preserved Loss of SMARC81 (INl1)
Brachyury+ Brachyury+ Brachyury+/- a Brachyury+
unoh stochemlcal Pancytokeratin-
pro e Pancytokeratin+ Pancytokeralin+ Pancytokeratin+
EMA+ EMA+ EMA- EMA+
S1 00+ S100+ 5100-/+ S100+/-
Metastasis: 13% Metastasis: 9% Metastasis. 30% Metastasis 30%
Local progression : 46% Local progression. 54°10 Local progression: 65% Local progression: 54%
Outcome Median PFS: 24 months Median PFS . 26.5 mon1hs Median PFS: 6 months Median PFS . 4 months
Death during follow-up: 29% Death dunng follow-up· 42% Death during follow-up: 61 % Death during follow-up: 43%
Median OS : 48 months Median OS: 43 months Median OS · 15 months Median OS: 13 months
- - - -
~immur\Opositive ; - , 1mmunonegative; OS, overall survival: PFS. progression-free survival
·, ~rtive in the conventional or chondroid component. negative 1r. the sarcoma!ous cornpone~I , ,
No.e Data are based on the systematic review of 245 con\ient10·1a: chorCc•meis \main reteron\;es. 13349 ·3144}), 2l0 chondroid chordomas (maln references: (2876,31441),
5? Oedifferenuated chordomas (main references: !1581 ,3144)1. and 65 poorly d1flerenl1atad chordomas (ma111 rett:rences · {1258.2912} ).

;.1 •. ·.
'•.J' 1 11 ' 1 ~ n 11 iq u ti 1 C:ll t al iumours involving rie CNS
1 1
' ' '\ - 335
Etiology <1 1r,1 ' i In , 1 .,, 1ti•:nt rif lhr' pr!p11l::1t1r1ri r1r1tr1r,hnrrJ;:rl rr:rr. r ~Jrt ' t, ,
lill' 1n, 11n11!y l)I l'11111lilllll 1l !. ill() !. I Hlldlill' 1111! lilll ' .i•.•,()( 111lll>tl'. r.1 1 ;1 r1nrl1(,) il <1rly 1r1 lhr• rJrlrintr11rl rJrrJrJ1','• rtr1rJ thr! r,ri' ,r, 11 fro
witli lttlH 1
lllll !; ~;1 ' il'lll~.1!; Ill l '11ldi( 'll ihl iH I ()/ l r lltlfli , 11 Ulf.(''· w11t1 mr ii Pr ,I il;ir rrlN JlrH W ,rnr, r if thf! r1rJlr ir, hrJrrJril turr rJ1 Jr -ii Ir -tr •,fr, rrr,. 1

~ll lllllllll' dtq1lll'.llH111 lll II)( ' /H\ I (ll ' llf ~ l 111vr' IH H•11 IC'fH>rlnrl t11i11 i l rl' rt<>I ( .()rTlpln1Ply I ir1rlr)r r; lr1rirJ f)IJI rJt 1r1lt1, 01t1rir ', r, f fF: /.
11 ~1ti!'l (j<'TH' (~ 1°/n ()f ( , rJ',()'">) m1rl filfC)(~A "", 1r1n;Jll1r r.J rr1 11t::Jl1 ' ;' i'" • 1.:;·
of l,(j',(''i) hrJVf) rr3(, f ntly tinr~ri r]r) ', r ,r hr!rl I 1 I -~ 1lttt 1 , ~~ ',
1
n
Pathogen si I Y.')f 1r1r:1<,l1vat1nq mut::1l1()n (1ri 1()% rJf r,:.1",l'.>'l) r, rJ/ r13~ r , ~r· .
llw 7/i \ / Ul'lll' 1 11n1d t'!' llH' p1ulc' 111 h1 ncllyu1y, n 11ol od1or( l rH>Vf)I umr,nr Cj()rl8 lrl chord()mrl 1;31 'fl I I Jr ltl.r(': r1 rr r, -:11 i C. J

t1sst1t spt'cif1c l1.111 ~w 11pl1 u 11 ln c trn c 1il1c;i l frn nol ocllrnd cluvc ' i u1I tcrritoirJ/rhahrJ01d t1Jrn01w; loss rJf SM/\R(.B i rn JI i •., 1rP, .
op111c11t. l l1t 11ntt)t .l101d u11dc ) 1qrn )~, 1q1rcm.;irn1 nnc l cl1 f~n pp nms s1on in chorrJomris results frrJm .::i hornr;;ygow, rJr~ k: 1 1 rJr 1,f ''"'-'
b L111tl1 to Lw ,0111D tlH' nucl eus pulposus nf th n intmvcrt8brn l SM/If ?CB f qene [I ?58)

.1'" I •
,, .
~ .
...
·.
,
l ..
..-
... ,
j.
• r ' ,, •
. •r•.e.• ~ • ,~. • •

Fig. 8.46 Conventional chordoma . A This shows a lobular architecture and abundant myxoid matrix. B Nests and cords of cells. sometimes with vacuolated bubbly cytopiasr:
~. .
'"- . a: , .
." .·•
(physaliphorous cells). C Diffuse nuclear expression of brachyury.

l ... .
.. .
•t
' ... ...
... , ·• 11\,
1

•....·.• .. ..
J
... .. ,., \
~

.. .. ' •"• .,. .,


'\

.; ..,_ , • · ·'~·
• ' .. t~'4
• J 4 I
~

(
~ . "'
: • "

•,
t
.. '
• •. ••'
;.. '•
.. •' I tJfl • ,


• ...o) • • '. ~ 'Ill "" .:.',
· -
...;, .
lj
.
, •>

..
.
4 I ..,
j

c. ' ,I
,. 0

•... ·. .: •
I
... ...
L

... . ,,
I ,I
.. '
.. .. ., C I~
Q~

/ " ' I ~· -
..,
: ~·.' ..~ ~
".. -- . .•.-.
~
-~
•"' .
., 0

c , . -· ' .. ~

Fiy. 8.47 Pr,r1r1y d1lter1o11'1ated ch urdoma A T2 weighted axial MRI showing a large enhancing tumour arising in the skull base B Spindle or ep1thalto1d cells wirn
111110
m~'l
rr 2.\' z c T• '= cnorrJ•, ma ce Is e1press nuclear brachyury. D Loss of SMARCBl (INll ) expression 1s essential for this d1agnos1s

I 'I , I 'I 11 1
ii I Jr r ,, 1 If '_, 1' 1 ! ' i , 1r 11 J ! I 1 • l' rJ
Macroscopic appearance Box8.11 Diagnostic criteria for chordoma
Chordomas are lob~lated, solid tumours with a gelatinous
Essential:
appearance, destroying bone and extending into surrounding
Midline axial bone tumour
soft tissue.
AND
Histopathology Lobules of cohesive and physaliphorous cells in a myxoid or chondroid matrix
Conventional chordoma is divided into lobules by fibrous septa. AND
Tumo.ur cell~ are arranged in cords or ribbons separated by a Brachyury immunopositivity
myxo1d matrix. Tumour cells are large with clear to eosinophilic AND (In the case of epithe/ioidlsolid forms)
cytopla~m characterized by vacuolated or bubbly cytoplasm Loss of SMARCB1 (INl1 ) expression to confirm the diagnosis of poorly
(physallphorous cells) . Anisokaryosis and nuclear inclusions differentiated chordoma
may be observed, but prominent nucleoli, mitoses and apop-
totic bodies are scarce or absent. Chondroid chordoma is a
subtype of conventional chordoma containing extracellular homozygous SMARCB1 deletions in poorly differentiated chor-
matrix mimicking hyaline cartilage . domas .
Dedifferentiated chordomas are biphasic tumours , composed
of conventional chordoma juxtaposed to high-grade sarcoma. Essential and desirable diagnostic criteria
Brachyury (nuclear) and cytokeratin expression are preserved See Box 8.11 .
in the conventional component but lost in the high-grade sarco-
matous component 11379,151). Staging
Poorly differentiated chordomas are epithelioid and solid, with Union for International Cancer Control (UICC) staging is accord-
focal rhabdoid morphology and without physaliphorous cells, ing to bone sarcoma protoc ols.
and they are characterized by a loss of SMARCB1 (INl1) expres-
sion but retained brachyury expression {2912,1258). Prognosis and prediction
Outcome data accord ing to type are detailed in Table 8.02
Cytology (p. 335). Dedifferentiated and poorly differentiated chordomas
Not clinically relevant appear to have the worst prog nosis 13604,228,3144\ . In con-
ventional chordomas , the main prognostic factors for worse pro-
Diagnostic molecular pathology gression-free and overall survival are age > 60 years, skull base
No diagnostic molecular markers have been reported for location , regional extens ion or metastasis at diagnosis , tumour
conventional chordomas , but FISH can be used to identify size> 80 mm , and incomplete resection (3604 ,228,3144 ,1367\.

r. lr . 'i'l 1, : I; • i 1.1 ;j( ,f ' I ! . ~~ ni rH..10thel 1 a l tumour


::::1
. I
s 1nvo v1ng tt1e
CNS 337
Melanocytic tumour · lntr duction

Primary men1 ~geal mel~nocytic tumours are rare , and they can histology, molecular fea tures. and cltnrcal behaviour ~J1~r· 1:--1 ,~~
b~ c1rcu.msc nbe.d or diffuse, and benign or malignant. Well- melanomas are usually highly aggressive and rad10r~:.1:,1.:.r,t
dlfferent1ated, c 1 rcumscrib~d tumours are called meningeal tumours with a poor prognosis, and they may cause "="~.r.arJJ:~
melanocytomas ; .their malignant counterparts are meningeal metastases . Still , the prognosis may be substantially bet•e• frir
melanomas . Meningeal melanocytomas with increased mitotic patients with primary meningeal melanoma (particularly rf r.0rr
acti~ity or invasion of the CNS parenchyma are considered inter- plete resection of the primary tumour can be ach1i::ved) t~.3n ~ ..
mediate-grad~ lesions . Diffuse meningeal melanocytic tumours those with CNS metastasis of cutaneous melanoma 11772 183
are charact~rized by the involvement of large expanses of the 1007). Meningeal melanocytosis may remain asymptoma re fr.1 ..
subarachno1d spa~e . with or. without focal nodularity. Based a variable period of time, but once symptoms develop. th.'=! pr0g-
on whether .th.e lesion has a benign or malignant histological nosis is usually poor; currently, the prognosis for NRA S-muta"
phenotype, 1t 1s called meningeal melanocytosis or meningeal meningeal melanoma and meningeal melanomatos1s 1n chlldrs11
melanomatosis, respectively {1772) .
is very poor 12233,1635,1772} .
. Molec.ular anal~sis is often very helpful for corroborating the Of note, in the fourth edition of the WHO classification of s/l1r
d1agnos1s of a primary meningeal melanocytic tumour. Analy- tumours (2018), the taxonomy of cutaneous melanocytic neo-
sis of GNAQ, GNA 11, PLCB4, and CYSLTR2, as well as meth- plasms is based , where possible , on different underlying evo-
ylation profiling , is especially useful for recognizing these neo- lutionary trajectories (pathways) associated with differences ri
plasms as primary CNS tumours and discriminating them from genetics , clinical presentation , and/or histopatholog1cal fealures
other pigmented CNS tumours such as malignant melanotic 1830). Perpendicular to this axis, another axis contains informa-
nerve sheath tumours 11676,637,1774,1164,1007,1772). The tion on the recognizable progression stages of the resp ective
presence of an additional SF3B1, EIF1AX, or BAP1 mutation neoplastic disorder. In this scheme, cutaneous melanocytomas
(BAP1 mutation leading to a loss of BAP1 protein expression are considered intermediate lesions because they have more
on immunohistochemistry); of chromosome 3 monosomy; or of pathogenic mutations than their fully benign counterparts. but
complex copy-number variations indicates aggressive behav- fewer than the melanomas they can produce. It is not yet pos-
iour consistent with meningeal melanoma {3269). In children , sible to follow exactly the same approach for primary meningeal
primary meningeal melanomas and meningeal melanocytosis melanocytic tumours because of their rarity and the relative lack
and melanomatosis are often NRAS-mutant and occasionally of data on their genetic evolution. Therefore, the term "melano-
BRAF-mutant (2233 ,1635,1637,2436,2791). cytoma" as in (intermediate-grade) meningeal melanocytoma
The lack of large clinical studies with adequate patient follow- has somewhat different connotations from those of cutaneous
up has hindered definitive assessment of the correlation between melanocytomas.
Diffuse meni~geal melanocyti c neopl asms: Gessi M
Bastian BC
Melanocytos1s and melanomatosis Kolsche C
Ku ster s-Vandevelci e HV
Reyes- Mug ica M

Definition
Me~i nge~I melanocyt~sis is a diffuse or multifocal meningeal
prohferat1on of cytolog1cally bland melanocytic cells that arises
from leptomeningeal melanocytes . Meningeal melanomatosis is
a diffuse or multifocal meningeal proliferation of melanoma cells
that arises from leptomeningeal melanocytes and often shows
CNS invasion .

ICD-0 coding
8728/0 Meningeal melanocytosis
8728/3 Meningeal melanomatosis

ICD-11 coding
2A01.0Y & XH8974 Other specified meningeal tumours &
Meningeal melanocytosis
2A01 .0Y & XH1 BP? Other specified meningeal tumours &
Meningeal melanomatosis

Related terminology
None

Subtype(s)
None

Localization
Meningeal melanocytosis and melanomatosis involve the
leptomeninges, often extending into Virchow-Robin spaces.
Meningeal melanomatosis frequently displays invasion of the
CNS parenchyma. The lesions generally involve large expanses
of the subarachnoid space, with focal or multifocal nodularity
occasionally present. The sites of highest frequency include
the temporal lobes, cerebellum , pons, medulla, and spinal cord
Fig. 9.01 Meningeal melanocytosis. A Autopsy findings in a child with meningeal
11523).
melanocytosis. The meninges of the lower spinal cord appear diffusely packed with
tumour tissue. The lesion harboured an NRAS p.061K mutation. B Melanocytic cells
Clinical features with benign features diffusely proliferate within the meninges between the nerve roots
Neurological symptoms associated with meningeal melanocy- of the lower spinal cord.
tosis or melanomatosis arise secondarily to either hydrocepha-
lus or local effects on the CNS parenchyma . Neuropsychiatric cysts , synngomye li a , b rain tumours (including astrocytoma ,
symptoms, bowel and bladder dysfunction , and sensory and choroid plexus papilloma , ep endymoma , and germinoma) . and
motor disturbances are common . Once malignant transforma- structural defects suc h as Dandy- Walker or Chiari malforma-
tion occurs , symptoms progress rapidly, with increasing intra- tions \261 1J. The incidence of neurological involvement , mela-
cranial pressure resulting in irritability, vom iting , lethargy, and noma , and death is significantly associated with the projected
seizures . Diffuse meningeal melanocytic tumours frequently ad ult size of the largest congenital melanocytic naevus (1637).
occur in the setting of neurocutaneous melanosi s, a syndrome
that is further characterized by giant or numerou s congenital Imaging
melanocytic naevi of the skin that usually involve the trun k or the CT and MRI of meningeal melanocytosis and melanomatosis typ-
head and neck (1523) . About 10- 15% of p atients with large con- ically show diffuse th ickening and enhancement of the leptome-
genital melanocytic naevi of the skin develop c linical symptoms ninges, often with focal or multifocal nodulanty !2519). Depending
related to meningeal melanocytosis (720). an d radiological ev i- on melanin content, they may have a characteristic appearance
dence of CNS involvement has been rep orted in as many as on MRI due to the paramagnetic p roperties of melanin, resutt-
23% of asymptomatic children with giant congeni tal naevi (965). ing in an isodense or hyperintense signal on T1 -weighted images
Other features are communicating hydrocephalus, arachnoid and a l1ypo1ntense signal on T2~wei ghted images {2959}

~1 e 1 0nOLVh: tumours 341


occurs without cutaneous melanocy 1c lesions Me1a.-1orr1a o-:",
~iris a bimodal aqe dis nbut1on and may bec0me mar11 fes ...
children with or without neurocutaneous melanosis as ,.,.~1 1 a>.
1n adults (mostly in the fourth decade of hfe) (1772}

Etiology
Diffuse meningeal melanocyt1c neoplasms assr;c1at~,.J N•tl" r·~ ;-
rocutaneous melanos1 s derive from melanocyte preciJrs0r r,~ ;.
that reach the CNS after acqu1nng postzygor1c c;omat": li'•Jf;,?-
tions , mostly of NRAS (chromosome 1p13) 11637.24361 Oif' ~c.~
melanocytosis may be associated with BRAF mutat1on5 •r a
minority of cases 12791 I. Copy-number variations found 1r r.eN '/
acquired or cl inicoradiologically progressive diffui:;e menmg~1
melanocytic neoplasms show overlap with those described -
cutaneous melanoma. even the in absence of maltgnar f~­ 1

tures at the histopathological level {1636,3281 I.

Pathogenesis
Menin geal melanomatosis and melanocytosis are mostly asso-
ciated with postzygotic somatic mutations in NRAS. wt;1c·,
pred ispose to oncogenesis as a first hit in a multistep process
(1 637,2436) . NRAS is part of the family of RAS GTPases ac · g
as a molecular switch that regulates the activation of the RAF/
MEK/ ERK and P13K/AKT/mTOR pathways . NRAS muta 1ons
mainly occur at codon 61. the catalytic centre of the GTPase
and cause constitutive activation of NRAS, resulting in cell
prol iferation and growth (1772}. Amplification of the muta ed
Fig. 9.02 Meningeal melanomatosis. A T1-weighted axial MRI revealing a hyperin- NRAS gene has also been described in an aggressive form of
tense. contrast-enhancing lesion outlining the gyri and sulci in the left tronto-parieto- neurocutaneous melanosis lead ing to CNS and widely d issemi-
occipital region in a 5-year-old child. B Macroscopy of meningeal melanomatosis in a nated congenital melanoma {2790 ).
child with neurocutaneous melanosis who succumbed at the age of 17 months due to
rapid disease progression. C Macroscopic appearance of the cerebral tissue shows Macroscopic appearance
brownish discolouration of the thickened leptomeninges and black discolouration of
Dependent on me lan in content , d iffu se meningeal melanocytic
the underlying cerebral cortex.
neoplasms appear as dense b lack replacement of the suba-
rachnoid space or as dusky cloud ing of th e meninges.
Epidemiology
Diffuse meningeal melanocytic neoplasms are rare , so a pop- Histopathology
ulation-based incidence is difficult to estimate. The incidence The pathological proliferation of leptomen in geal melanocytes
of neurocutaneous melanosis is reported as 0 .5-2 cases per and their production of melanin account for the main micro-
100 000 person -years . Melanocytosis mainly affects children , scopic findings in meningeal me lanocytosis and melanomato-
mostly in the context of neurocutaneous melanosis, and it rarely sis . The tumour cells may assume a variety of shapes. including

A ~ ~
Ftg. 9.03 Me'. ngeal me1anomatos1s. A Malignant melanocytic cells diffusely infiltrate the underlying brain parenchyma. The tumour carries an NRAS p.Q61R mutation. 1 A
s.,t)pupulat1on o! melanoma cells are heavily pigmented.
~p1~dl~d .. r?und, oval , and cuboidal. In meningeal melanocy- Box9.01 Diagnostic criteria for diffuse meningeal melanocytic neoplasms
tO~t s . 1nd1V1dual cells are cytologically bland and accumulate
Essential:
w1thm. the s~barachnoid and Virchow-Robin spaces . Lesions
Diffuse or multifocal primary meningeal melanocytic neoplasm
tha1 hrstolog1cally look like meningeal melanocytosis but show
u~eq u ivocal invasion of the CNS parenchyma should be con - AND
Stdered as meningeal melanomatosis {2959) . The presence • For meningeal melanocytosis: absence of CNS parnnc~yma l~vasion ,
of marked cytological atypia, mitotic activity, or necrosis also absence of marked cytological atypia, absence of m1tot1c act1v1ty. and
warrants a diagnosi s of meningeal melanomatosis. Distinction absence of necrosis
• For meningeal melanomatosis: Invasion of the CNS parenchyma and/or
from metastasis of cutaneous melanoma may be impossible marked cytological atypia and/or mitotic activity and/or necrosis
using microscopy alone; additional molecular testing may help
to solve th is diagnostic problem {637} . Desirable:
In children, meningeal melanocytosis/melanomatosis is often NAAS-mutant and
Cytology rarely BRAF-mutant
Diagnostic cerebrospinal fluid cytology in patients with menin-
geal melanomatosis may reveal atypical cells that often have
ep1thelioid features but immunocytochemically express melano- Staging
cyt1c markers and may contain melanin pigment {1689). Not relevant

Diagnostic molecular pathology Prognosis and prediction


See the Diagnostic molecular pathology subsection in Circum- Melanocytosis may remain asymptomatic for a variable period
scribed meningeal melanocytic neoplasms: Melanocytoma and of ti me, but once symptoms develop the prognosis is usually
melanoma (p. 347) . Mutation and DNA methylation profile analy- poor {1 772} . Melanomatosis is usually a very aggressive dis-
ses often help distinguish metastatic cutaneous melanoma from ease with a dismal prognosis {1635) . A particularly ominous
primary meningeal melanocytic tumours. In ch ildren , meningeal complication in patients with ventriculoperitoneal shunting to
melanocytosis and meningeal melanomatosis are often associ- relieve hydrocephalus is peritoneal melanomatosis (439) Dis-
ated with somatic mutations in NRAS. tinction of a primary (diffuse) meningeal melanocytic tumour
from metastatic spread derived from an extradural (usually
Essential and desirable diagnostic criteria cutaneous) melanoma is critical for guiding prognostication
See Box 9 .0 1. and therapy.
M
Circumscribed meningeal melanocytic GASSi
Bristl8 n Bl'.:
C
neoplasms: Melanocytoma and melanoma KDI c.h8
Kusters-Va nr:levefrJr:! HV

Definition
Circumscribed meningeal melanocytic neoplasms are tumours
that arise from leptomeningeal melanocytes and range histo-
logically from well-differentiated tumours (meningeal melanocy-
toma) to frankly malignant neoplasms with aggressive growth
properties (meningeal melanoma). Tumours with a bland histo-
logical appearance but increased mitotic activity or invasion of
the CNS parenchyma have been defined as melanocytoma of
intermediate grade.

ICD-0 coding
8728/1 Meningeal melanocytoma
8720/3 Meningeal melanoma

ICD-11 coding
Fig. 9.05 Meningeal melanocytoma. A Tl-weighted pre-contrast MRI rf?t"~
2A01 .0Y & XH3DN1 Other specified meningeal tumours & Mel- slightly hyperintense lesion at the T8-T9 level of the spinal cord. B The lesl()ri
anoma, meningeal strong contrast enhancement (Tl -weighted MRI). C On T2-weighted MRI. ~e
is hypointense.
Related terminology
Not recommended: melanocytoma (without site); melanoma or intramedullary localization have been reported 12183.1667'.
(without site) . Meningeal melanomas may occur throughout the neurax
but, like melanocytomas , they show a predilection for the sp.-
Subtype(s) nal canal and posterior fossa (1007,359) . A purely 1ntrapare .
None chymal location of a melanoma in the CNS is highly indlCalr1e
of metastatic disease.
Localization
Meningeal melanocytomas occur mostly in the cervical and Clinical features
thoracic spine. They can be dural based or associated with Patients with meningeal melanocytomas and melano as
nerve roots or spinal foramina {359,1121 }. Less frequently, present mostly with symptoms related to compression or e
they arise from the leptomeninges in the posterior fossa or spinal cord, cerebellum, or cerebrum by an extra-axial mass
supratentorial compartments . The trigeminal cave and cranial with focal neurological signs depending on location [359.3516.
base is a site with a peculiar predilection for primary menin- 1917}. Distant metastasis from meningeal melanocync tumours
geal melanocytic neoplasms that are associated with ipsilat- mostly melanomas , are rare; they have been reported 1n .1ver.
eral naevus of Ota I1761, 1129}. Rare cases of intraventricular bone, and lungs {1772,1771) .

Fig. 9.06 Meningeal melanocyloma A tumour from a 25-year-old patient with a lesion in the cavernous sinus. A Low-power view shows proliferating cells w1ttl abuooanl
pigment but no maior atypia or necrosis . B A higher-power view from the same tumour. C High-power view of the same tumour shows an absen~ oi mahgnant fa&ureS
proliferating pigmented cells The cells have a low N:C ratio, small nuclei, and inconspicuous nucleoli.
. ·-----:Ml·-- ~J~~~{
f11.9.07 Meningeal melanocytoma . A A melanocytoma, harbouring a GNAO p Q209L mutation, is composed of densely packed. slightly spindled or oval tumour cells conta1n-
gvariable melanin pigment. B The tumour does not show marked nuclear polymorphism.

Fig. 9.08 Meningeal melanocytoma, intermediate grade. A This tumour, affecting the tentorium, is composed of rounded cells without marked nuclear polymorphism but with
increased mitotic activity. The tumour carries GNA 11 and EIF1AXmutations . B This GNAQ-mutant tumour diffusely infiltrates CNS tissue of the Ilium terminale The surrounding
nervous tissue shows marked gliosis and Rosenthal fibres .

Imaging two relatively large series . the mean patient age at diagnosis
Meningeal melanocytomas and melanomas are isoattenu at- of meningeal melanocytoma and melanoma was 45 6 years
1ng to hyperattenuating , contrast-enhancing on CT, with an (range: 23 - 69 years) and 53 .7 years (range 15- 86 years) .
imaging appearance similar to that of mening iomas but usu- res pectively (1769,1164) . In other studies . the mean age at
ally without hyperostosis or intratumoural calcification \2959). the time of diagnosis of meningeal melanoma was found to
On MRI, they often show T1 hyperintensity due to the para- be 48 .5 years for adults and 5.4 years (median . 3 O years) for
magnetic properties of melanin pigment , and they are typi- cl1i ldren \2000 ,2233). For meningeal melanomas an annual
cally isointense to hypointense on T2-weight ed images \2957}. incidence of 0.005 cases per 100 000 population has been
hyperintense on FLA IR images , and th ey enh ance after reported 1·19'17).
t• gadolinium 11917) . CNS structures adjacent to a meningeal
melanoma are often T2-hyperintense as a resul t of vasogenic Etiology
oedema generated in response to rapid tumour growth. which In most well-d1tferentiated meningeal melanocytomas, copv-
may be accompanied by invasion by tumour cells into the CNS number variations are either absent or l1rn1ted in number. wh~n
parenchyma 1637) . present, they usually affect a single whole chromosome or large
parts of a single chromosome or a l1m1ted number of chromo-
Epidemiology somes {3268.1773) . The chromosomal alterations 1dent1f1ed in
Meningeal melanocytomas and melanomas are rare. account- melanocytoma may include gains of chromosome arms Sq and
ing for 0.06- 0.1% of mening ea l tumours . Melanocy tomas have 6p, loss of chrornosonie arms 1p and 6q, and monosomy ot
an estimated incidence of 1 case per 10 million person-years d1romosorne 3 this last a:terat1on fou nd 1n tum0urs with 1nter-
12344,1917) They can occ ur in patients of any age , but they rned!ate -~ 1r ac1 3 histology I 1164 ,17'7 31 Su h alterations. 1nclucf-
are most frequent in the fourtl1 and tift11 dacades of lite In 1nq monusOfllY < f : llromosorne 3. can be found in meningeal
intermediate grade and meningeal melanomas may carry an
additional EIF1AX, SF381 , or BAP1 mutation. again rn a mutu-
208 209 210 ally exclusive pattern and with a higher incidence reported in
G Q/L R melanomas 11769,1164,32681. Childhood meningeal melano-
G G C c NT A A G G mas in patients with neurocutaneous melanosis typically ~ar­
bour NRAS mutations (2436,1637) .

Pathogenesis
Mutually exclusive mutations in GNAQ, GNA11, PLCB4. and
CYSL TR2 are considered the first step in oncogenesis of menin-
geal melanocytic tumours not associated with neurocutaneous
GNAQ melanosis. The glutamine at codon 209 or arginine at codon 183
of GNAQ and GNA 11 is essential for GTP hydrolysis, and muta-
tions at these codons impair GTPase activity, leading to consti-
Fig.9.10 Meningeal melanocytoma. Detection of a GNAQ p.Q209L mutation by
tutive activation of downstream intracellular pathways including
Sanger sequencing.
the RAF/MEK/ERK and Hippo/YAP1 signalling pathways char
regulate cell growth and proliferation (212,1772). Like in uveaJ
melanomas as well , but (like cutaneous and uveal melanomas) melanomas, mutation in EIF1AX, SF381, or BAP1 is considered
meningeal melanomas usually have a more complex copy-num - to represent a next step in the oncogenic process {2397.2121
ber variation profile , with multiple large chromosomal gains and/ EIF1AX and SF381 encode eukaryotic translation initiation rac-
or losses 11773,1164). tor 1A (EIF1A) and splicing factor 3b subunit 1 (SF381). respec-
Mening eal melanocytomas and melanomas harbour mutu- tively, but their role in the oncogenesis of meningeal melanocytic
ally exclusive activating hotspot mutations in GNAQ, GNA 11, tumours is not yet fully understood . BAP1 is a well-charactenzed
PLCB4, or CYSL TR2. GNAO and GNA 11 mutations are most tumour suppressor gene. Carriers of germline BAP1 mutatJons
fre quent , ob served in about 60 - 70% of cases (1770,2176, are at risk of developing cutaneous, uveal, and meningeal meta-
1164,3268 ,3271) . Usually, meningeal melanocytomas and nomas, as well as mesotheliomas, clear cell renal cancer, and
melanomas do not harbour HRAS, KRAS, BRAF, or KIT muta- various other tumour types 1708). BAP1 (chromosome 3p21.1)
tions (1770 ,1676,3370 ,10711. TERT promoter mutations are encodes a nuclear ubiquitin hydrolase with multiple nuclear and
also usually ab sent l 10751. Meningeal melanocytomas of cytoplasmic substrates, regulating DNA repair, transcription.

346 Mdar1ocyt1c rumours


'2

oe

0.4
;.

0.0
·~-

_., 4 .-.,' '


I >
. i:

-41

-1.2

~ 6 ~ ~ ~ ~ ~ t 5
0 .. ...
i
... "'
~ ~
....
~ i "'
~ ~~~ s i
i 5 ~ i
flt.9.11 Meningeal melanoma. Copy-number variation plot shows multiple chromosomal changes.

and cell death. Loss-of-function hemizygous mutations com- Melanomas are more pleomorphic and m1totically active .
bined with chromosome 3 monosomy result in decreased or and they may have a high cell density. In addition , meningeal
absent BAP1 protein expression . melanomas often demonstrate unequivocal invasion of the CNS
parenchyma or coagulative necrosis . They may be composed
Macroscopic appearance of pleomorphic spindled or epithelioid cells (arranged in loose
Meningeal melanocytomas and melanomas are circumscribed nests , fascicles, or sheets) and display variable cytoplasmic
mass lesions that may be black, reddish-brown , blue, or mac- melanin (359,1007]. Some meningeal melanomas contain large
roscopically non-pigmented , depending on the melanin con- cells with bizarre nuclei, numerous (typical and atypical) mitotic
tent. figures, and large nucleoli ; others are highly cellular and less
pleomorphic, usually consisting of smaller, tightly packed spin-
Histopathology dle cells with a high N:C ratio . Meningeal melanomatos1s may
Circumscribed meningeal melanocytic tumours show a spec- arise from diffuse spreading of a primary meningeal melanoma
trum of histopathological features , ranging from bland-appear- through the subarachnoid space.
ing, low-grade, well-differentiated melanocytomas to overtly
malignant melanomas . Usually, all meningeal melanocytic Cytology
tumours strongly express S100, vimentin , melan-A (MART1), In patients with meningeal melanoma, cytological examination
HMB45, and MITF (3516} . Well-differentiated melanocytomas of cerebrospinal fluid may show atypical or frankly malignant

I
may show variable (sometimes high) cell density and are usu- cells , often with ep1thellold cytology and containing melanin pig-
ally composed of densely packed , slightly spindled or oval ment.
tumour cells containing variable (at times abundant) melanin . .
The tumour cells may form tight nests with a superficial resem - Diagnostic molecular pathology
blance to the whorls of meningioma. Heavily pigmented tumour Mutation analysis (including for GNAO, GNA11 , PLCB4. and
cells and intratumoural macrophages are especially seen at the CYSL TR2) and methylation profiling are useful for recogniz-
periphery of nests. Other melanocytomas may show storiform , ing meningeal melanocytic tumours as primary CNS tumours
vasocentric, or sheet-like arrangements. Only rare amelanotic and discriminating them from other pigmented CNS tumours
melanocytomas have been described . The nuclei are oval or such as malignant melanotic nerve sheath tumours [1676,637.
bean-shaped, occasionally showing grooves, with small eosin- 1774}. Primary meningeal melanomas in adults are rare . and
ophilic nucleoli . Cytological atypia, necrosis, and mitoses are when encountered, they raise susp1c1on of metastatic disease .
generally absent (on average < 0.5 mitoses/mm 2 , equating to In adults, the ident1f1cation BRAF, NRAS, or TERT promoter
< 1 mitosis/10 HPF of 0.5 mm in diameter and 0.2 mm 2 in area). mutations help differentiate a cutaneous melanoma metastasis
Melanocytomas generally do not show invasion of CNS paren- from a primary meningeal melanoma . Conversely, the pres-
chyma (359,1772,3370) . ence of GNAO or GNA 11 mutation 1n the absence of a uveal
Based on data from a relatively large study, meningeal mel- melanoma or a blue naevus-l1ke melanoma strongly favours a
anocytic tumours with the histology of melanocytoma but show- primary meningeal tumour. Combining mutation. copy-number,
ing CNS invasion or increased mitotic activity (0 .5-1 .5 mitoses/ and DNA methylation profiles has been described as a method
rnm 2 . equating to 1-3 mitoses/10 HPF of 0 5 mm in diameter of further distinguishing cutaneous melanoma metastases from
and 0 2 mm 2 in area) have been def med as intermediate-grade other melanocync tumours (1676 ,1164 ,1007)
melanocyt1c neoplasms (359] .
Essential and desirable diagnostic criteria Box9.02 Diagnostic criteria for circumscribed meningeal melanocy ir, !'lP.OO!asms
See Box 9.02 .
Essential:
Staging Circumscribed/localized primary melanocytic neoplasm in the meninges
Not relevant AND
• For melanocytoma: limited cytological atypia, (almost) no mlt~, no .
Prognosis and prediction necrosis, and (in cases of evaluable CNS parenchyma) no CNS 1nvasaoo
• For intermediate-grade melanocytoma: mitotic count of O.~ 1.5 mirosesl
3

The clinical behavio~r of _circumscribed meningeal melanocytic mm and/or CNS invasion, but limited cytological atypia and no necrosi$
2
tumours correlates with h1stopathological features. However the • For melanoma: mitotic counr > 1.5 mitoses/mm2 and/or necrosis. often
l~ck of large _cl.i~ical studies with adequate patient follow-up' has accompanied by marked cytological atypia
hindered def 1nit1ve assessment of the correlation between histol-
ogy, molecular fea'.ures , and clinical behaviour, in particular for Desirable:
melanocytom~s of intermediate grade. Although melanocytomas Demonstration of GNAO, GNA 11, PLCB4, or CYSLTR2 mutation corroborates the
lack anaplast1c features, in some patients local recurrence or CNS origin of the neoplasm, especially after exclusion of uveal or blue na~
leptomeningeal seeding occurs; intermediate-grade melanocytic like melanoma
tum?urs seem to be more recurrence prone. Malignant transfor- Additional molecular markers (SF381 , EIF1AX, and BAP1 mutations; chromosome
mation of a melanocytoma and metastatic spread outside the 3 monosomy; complex copy-number variations) indicating aggressive behaviour
CNS have been reported (2729,1771,1684). Some meningeal
•1 mm2 equates approximately to 5 HPF of 0.5 mm in diameter and 0.2 mm2 in area.
melanocytic tumours (not necessarily associated with worri-
some histology) harbour EIF1AX, SF3B1, and BAP1 mutations
and show aggressive clinical behaviour (1769,3268) . Therefore, prognosis, and it can rarely metastasize to distant organs (1772J.
the diagnosis of a meningeal melanoma should be considered Nevertheless, the prognosis tends to be better for patients with
in the presence of additional EIF1AX, SF3B1, or BAP1 mutations primary meningeal melanoma (particularly if complete resection
(BAP1 mutations leading to a loss of BAP1 protein expression at of the primary tumour can be achieved) than for patients wtttY
the immunohistochemical level); chromosome 3 monosomy; or CNS metastasis of cutaneous melanoma (983.1007). Currently
complex copy-number variations (3269}. Meningeal melanoma is the prognosis for NAAS-mutant meningeal melanoma in children
usually a highly aggressive and radioresistant tumour with a poor is very poor (2233,1635) .

. ; '

348
. '
Haematolymphoid tumours involving the Soffiett1 R

CNS : Introduction

The follow ing sections cover lymphomas and histiocytic vessels in the brain and thereby typically induc% preigr~>: 1r.;
~umo~rs that_may occur as solitary or multifocal CNS lesions neurocognitive deterioration (mimicking dementias) or '3i:. ;•e;
1n. primary 1ntraparenchymal and meningeal localizations . neurological deficits (mimicking cerebrovascular d 1s ;:;a~~
Virtual ly al l of these tumour types may also man ifest in other with a stroke -like appearance on MRI. Primary MALT lyrrip CJfT'.a
organs. Therefore , primary CNS manifestation needs to be of the dura is a rare lymp homa type that clinically and rad o r::;g _
disti ngu ished from secondary manifestation in the CNS cally may be mistaken for meningioma and has a s1m1lar' I gar:".!
i.e. metastases from systemic lesions. The way the different outcome after local treatment 11564 }.
tumour types are presented here follows the revised fourth- Despite impressive advances in our understanding IJ tr.~
ed ition volume of the WHO classification of CNS tumours with etiology and pathogenesis of primary CNS lymphomas ..,
th.e sections on less common CNS lymphoma types being particular for CNS-DLBCL, the mainstay of their d1agr:os"i:
slightly expanded . Of the primary CNS lymphomas, diffuse assessment remains tissue-based classification using h1stolog1-
large 8-cell lymphoma of the CNS (CNS-DLBCL) , previously cal and immunohistochemical analysis of biopsy spec mer5
cal led "primary CNS lymphoma", is the most common tumour Molecular pathology investigations , such as the demons rat:
type encountered . There has long been only modest insight of a clonal prol iferation of 8 cell s or (rarel y) T cells , are occa-
into the pathogenesis of CNS-DLBCL, mainly because of sionally helpful as adjunct methods, for example to distingu1s
limited tissue availability (because most patients undergo ste- neoplastic from inflammatory lymphoid infiltrates . In add1ion 10
reotactic biopsy rather than surgical resection), and a lack of individual cases, molecular testing may provide helpful in or a-
co rrelations between histological or molecular data and clini- tion by detecting diagnostically relevant translocations f gere
cal outcomes . Large-scale genomic investigations have char- fusions (e.g. by FISH) or an underlying EBV infection (e.g. bf
acterized the mutation profile and identified relevant genetic in situ hybridization). The importance of avoiding corticosteroid
dri vers in these tumours . In particular, the B-cell receptor, toll- administration before tissue biopsy in the diagnostic assess-
like receptor, and NF-KB pathways are frequently activated by ment of CNS lymphomas has long been rec ognized. H1gh!'t
recur rent mutations ; in addition , genes involved in chromatin potent corticosteroids like dexamethasone may induce rap d 1

structu re and modification , cell-cycle regulation , and immune tumour waning, impeding histolog ical diagnosis in as many as
recogni tion are commonly altered {2153,2150 ,2154,2205,349, 50% of cases {389).
3298). Among these various genetic changes, MYDBB and Histiocytic neoplasms may represent a clinical challenge
CD798 mutations are of potential clinical interest because because of their rarity, broad clinical spectrum (often mim ek-
th ey are frequent and may be detected in several body fluids ing non-neoplastic conditions), and varied histology (2641.
(plasm a, cerebrospinal fluid, vitreous fluid) . Liquid biopsy- For instance, Erdheim-Chester disease of the CNS. which
based detection of these mutations may assist disease moni- preferentially occurs in middle-aged adults, can be clm1cal')
toring under treatment {1305,2149 ,1306,1265), although their mistaken for various other diseases, such as multiple sclero-
detection in b lood (for non-invasive initial diagnosis) has not sis, neurosarcoidosis , CNS vascul itis, lgG4-related disease.
been proved to be a rel iable approach 12149). Genetically acti- and others . In addition to focal neurological deficits due o
vated pathways in CNS-DLBCL can be targeted by small mol- tumour-like masses , a peculiar clinical finding across severa!
ecules such as ibruti nib (1169 ,1908}, and the immune microen- histiocytoses (in as many as 30-50% of patients) is cognt 111e
vironment may be modulated by drugs such as lenalidomide impairment associated with brain and cerebellar atrophy ano
and pomalidomide 12146,1406) . An Increase in the mutation neurodegenerative lesions , whose pathophysiology 1s so11
burden, the presen ce of translocations involving the C0274 unknown . Comprehensive histolog ical and immunohistocherrtr·
(PO-U) and PDC0 1LG2 (PO-L2) loci in a subset of tumours cal assessment, complemented by molecular characterizauor
!53), and the expression of immune response biomarkers such as mutation analysis for BRAF p.V600E and other MAPI\
(2350) suggest a potential susceptibility of CNS-DLBCL to pathway gene alterations, is therefore of utmost irnportan 'e ror
immune checkpoint inhibitors , but cl inical evidence of this is confirming the diagnosis and guiding targeted treatment (753.
still limited 12220,1406). 755 ,1155,2707) . In addition to the common types of h1st1oc;tJc
Other lymphoid neoplasms (incl udi ng various types of low- tumours addressed in the individual sections of this "'hapter
LJrade B-cell lymphomas , as well as T-cell and NK/T-cell lym- ALK-positive histiocytosis has been identified as a no al y:...
r-,;t1r:,rnasJ rarely arise primarily in the CNS and may therefore of systemic histiocytic proliferative disorder that pred0Cl'1n· r tl ·
f;CJSG problems in differential diagnosis. Lymphomatoid granulo- occurs in young children and is driven by A LK fusions. m st
rr 1at0s1s is part of a grou p of EBV-associated 8 -cell lymphopro - commonly KIF5B::ALK (513 ,520} . Rare cases of ALK· s.t•1e
11!c r:.Jl1'.Je disorders that also includes other immunodeficiency- histiocytosis with exclusive involvement of the CNS have ~
: .. • ·, 1x,:med lyrnp~1omas . Because the histological features may reported (1952). underlining the importance of a th l'l
0'1erla 1.J, tt1e clinica l context is critical 12064) . lntravascular large molecular workup of CNS histiocytoses for diagnostic put
8-cell lymphoma (524) may obstruct small and medium-sized and for targeted therapy.
Nagane M
Prirnary d iffuse large B-cell lymphoma Deckert M
Batchelor T Paulus W
of the CNS Ferry JA
Hoang -Xuan K
Weller M

Definition
D•ffuse large B-cell lymphoma of the CNS (CNS-DLBCL) is a
OLBCL confined to the CNS at presentation . Its cytological fea-
tures. and many of its molecular features , correspond to those
of i s systemic counterparts .

ICD-0 coding
9680/3 Primary diffuse large B-cell lymphoma of the CNS

ICD-11 coding
2A81. 5 Primary diffuse large B-cell lymphoma of central nerv-
ous system

Related terminology
Acceptable: primary central nervous system lymphoma. Fig. 10.01 Primary diffuse large B-cell lymphoma of the CNS. A Single homoge-
neously enhancing lesion on postcontrast T1-weighted axial MRI. B Multiple enhanc-
Subtype(s) ing lesions with periventricular and subependymal location on postcontrast T1-we1ght-
None ed axial MRI.

Localization waning . Corticosteroids have been shown to prevent diagnosis


Primary CNS-DLBLCs are solitary brain lesions in 65% of in as many as 50 % of cases {389) .
cases , with the remaining cases being multifocal lesions.
Tumours are mainly located in the cerebral hemispheres Epidemiology
(38%), thalamus and basal ganglia (16%) , corpus callosum Primary CNS-DLBCL accounts for 2.4-3% of all brain tumours
(14%), periventricular region (12%), or cerebellum (9%) (1755, and 4-6% of all extranodal lymphomas (2847) . The overall
350}. The leptomeninges may be involved, but exclusive annual incidence rate is 0.47 cases per 100 000 population
meningeal involvement is unusual. Ocular manifestation (i .e. in (3327) . The incidence in patients aged > 60 years has been
the vitreous , retina, or optic nerve) occurs in as many as 20% reported to have increased over the past two decades (3327).
of patients and may antedate intracranial disease (1444} . Iso- CNS -DLBCL can affect patients of any age, with a peak inci-
lated spinal cord involvement occurs in < 1% of cases . Extra- dence during the fifth to seventh decades of life. The median
neural dissemination is rare . In cases with systemic spread , patient age is 66 years , and the M :F ratio is 3:2 (2347).
CNS-DLBCL has a propensity to home to the testis , an immu-
noprivileged organ (322 ,1445). Etiology
In immunocompetent individuals , etiological factors are
Clinical features unknown . There is no evidence that viruses such as EBV, HHV6
Patients present with cognitive dysfunction , psychomotor slow- !2429). HHV8 \2151 l. and the polyomaviruses SV40 and BK
ing, and focal neurological symptoms more frequently than with
headache, seizures, and cranial nerve palsies . Blurred vision
and eye floaters are symptoms of ocular involvement (213 ,1709,
2446) .
MRI is the most sensitive technique for detecting CNS -DLBCL .
Lesions are hypointense on Tl -weighted images , iso1ntense to
hypenntense on T2-weighted images , densely enhancing on
virus (2148,2178) play a role .

Genetic susceptibility
In immunocompetent individuals , genetic predispositions tu
CNS -DLBCL have not been described . About 8°·o of patients
have had a prior extracranial tumour [2652f . most of which amse
I
in the haematopoietic system In patients with CNS-DLBCL
oostcontrast images , and may manifest re stricted diffusion on and preceding extraneural lymphoma, comparative motecu:ar
diffusion-weighted images. Peritumoural oedema is relatively analyses of primary and secondary lymphomas m~:i.y co11t1rm
limited , and it is less seve re than in malignant gl1omas ancj brain or exclude a common clona l origin of tt1ese tumours. d1st1n-
metastases (1709) . Meningeal involvement may manifest as gu1sh1ng CNS relapse from an unrelated secondary cerebral
hyperinten se enhancement {1755) . With steroid therapy, lesions lymphoma . In 1ncJ1v1clual patients. assoc1at1 ns between CNS -
rnay vanish within hours (719J . DLBCL and other tumours (e .g carc111oma, mening1oma, and
Biopsy is the gold standard for establ1 sh111 g the cj1;:ignu-.:1s and gl1oma) or here?1tary tumour syndromes (e.g . neurofibromato-
classific ation of CNS -DLBCL It is 1mportc=1nt ;n 1111 111d1old cortl sis tvpe 1) are likely to be coincidental . Folate and methionine
costero1ds before biopsy because tho v 111c1t..JL·1;:: 1<:ip1J r!...orn<.u1 r' :0 1 ~~tiol1s rn have been proposed to be relevant to CNS-OLBCL

' .f·rn. tnl:;mphui i turnouts 1nvolv1ng the CNS 351


s~sceptibility. The G allele of the MTR:c.2756A>G p.D919G frequently Involve ch romosome bands 6q21 (in 52% 01 r::ai:-,c.:-. :
m1ssense ~olymorphism was found to be underrepresented 6p21 (in 37%), 8q12.1-q12.2 (in 32%), and 10q23 2 1 1?~7;:.i ;
among pat1e~ts with .CNS-DLBCL, suggesting that this allele COKN2A gene deletions are recurrent 1613,3491 Heterr1lf"Jr)1,-:.
has a protective function (1907) . or homozygous loss or partial uniparental disom1es of ch rr,rnr;
somal region 6p21 .32 affect 73% of CNS-DLBCLs. this re rJ11°Jr
Pathogenesis harbours the HLA class II-encoding HLA-DRB, HLA-DOA . ano
Tu~our cells ~orrespond to mature, late germinal-centre HLA-008 genes {1499,2674,28721 . Correspondingly, 55% 'lrt1
exit B c~lls derived from self-reactive/polyreactive precursor 46% of CNS-DLBCLs show loss of expression of HLA cla~s ,
cells. wh1c~ . ~ave escaped elimination , possibly fostered by and class II gene products, respectively 1322).
early .acqu1s1t1on ot MYDBB mutations. During a dysregulated Several important pathways (i .e. the 8-cell receptor, toll-lr1c'!
germinal-centre reaction, the tumour cells have increased self- receptor, and NF-KB pathways) are frequently activated due :r.,
reactivi~y/polyrea~tivity, allowing the tumour cell B-cell recep- genetic alterations affecting the genes CD79B (in 20- 83% 'j-1
tor to ~ind to ~u!t1ple CNS antigens {2156). which may, in part, cases), !NPP5D(in 25%), CBL (in 4%), BLNK(in 4%), CAR011 r,n
underlie the affinity and confinement of these lymphoma cells to 16%), MALT1 (in 43%), BCL2 (in 43%), and MYDBB (in > 50% 1
the C~S microenvironment. Tumour cells carry rearranged and which may foster proliferation and prevent apoptosis [1136.1732.
somat1cally mutated IG genes with evidence of ongoing somatic 2150,2154,2153,2872,2205,2223}. Epigenetic changes m~1
hypermutation [2152,2445,3189}. Consistent with the ongoing also contribute to pathogenesis, including epigenetic silencing
germinal-centre programme, they show persistent BCL6 activity by ONA methylation. ONA hypermethylation of DAPKt (in 8.4~
(392,2152,3189,2156). The process of somatic hypermutation is of cases), CDKN2A (in 75%), MGMT (in 52%), and RFC genes
not confined to its physiological targets (IG genes and BCL6) (in 30%) may be of potential therapeutic relevance {594.613.
but extends to other genes that have been implicated in tumor- 915,2872}. However, the ONA methylome does not unequivo-
igenesis, including BCL2, MYC, PIM1, PAX5, RHOH, KLHL 14, cally distinguish primary CNS-DLBCL from non-CNS DLBCL
OSBPL10, and SUSD2 {393,2157,3298}. These data indicate [3341}.
that aberrant somatic hypermutation has a major impact on the
pathogenesis of CNS-DLBCL. The fixed lgM/lgD phenotype Macroscopic appearance
of the tumour cells is in part due to miscarried IG class-switch CNS-DLBCLs occur as single or multiple masses ln the brain
rearrangements during which the Sµ region is deleted {2155). parenchyma, most frequently in the cerebral hemispheres.
PROM1 mutations also contribute to impaired JG class-switch Often , they are deep-seated and adjacent to the ventricular
recombination [645). system. The tumours are firm , friable, granular, haemorrhagic
Translocations affect the IG genes (in 38% of cases), BCL6 and greyish-tan or yellow, with central necrosis. They can
(in 17-47%), and ETV6 {2158,434,394} recurrently, whereas also be virtually indistinguishable from the adjacent neuropil.
MYC translocations are rare and translocations of the BCL2 Demarcation from surrounding parenchyma is variable. Some
gene are absent {392,434,2158} . FISH and genome-wide tumours appear well delineated, like carcinoma metastases.
SNP analyses have shown recurrent gains of genetic material, When diffuse borders and architectural effacement are present.
most frequently affecting 18q21-q23 (in 43% of cases), which the lesions may resemble gliomas. Meningeal involvement may
includes the BCL2and MALT1 genes; chromosome 12 (in 26%); resemble meningitis or meningioma, or it may be inconspicuous
and 10q23.21 (in 21%) {2872}. Losses of genetic material most macroscopically.

Histopathology
Model of the pathogenesis of CNS-DLBCL CNS-DLBCLs are highly cellular, diffusely growing, pattern-
less tumours . Centrally, large areas of geographical necrosis
ONA methytation Gain of genetic material
are common . Necrotic zones may harbour viable perivascular
DAPK1} Loss of /
18q21 ... 11c':fv·::on lymphoma Islands. At the periphery, an angiocentric infiltra-
CDKN2A . . . expression
Loss of genetic material tion pattern is frequent. Infiltration of cerebral blood vessels
MGMT
.....----......
SHMIASHM
Sµ deletion . . No CSR causes splitting of their argyrophilic fibre network. From these
CDKN2A . . Protifer111ion perivascular cuffs, tumour cells invade the CNS parenchyma.
1
6p21 (HLA) . . . :'~;: either with a well-delineated invasion front with small clusters,
or ~ith single tumour cells diffusely infiltrating the tissue. Cyto-
Polntmullltion
(not SHMIASIN) log1cally, CNS-DLBCLs consist of large atypical cells with
INPP5D
CDTflB BCR sign11Ulng large round, oval, irregular, or pleomorphic nuclei and disnnct
CSL .. ll~li~.;::n nucleoli, corresponding to centroblasts or immunoblasts.
BLNK

CARD11 .. ,.::J'v-::on Mitoti_c a.ctivity is brisk. Tumour cells are intermingled with
reactive inflammatory infiltrates consisting of mature, small T
MYDBB .,. ao~~·::on and B lymphocytes. C03-positive T cells predominantly cor-
PRDM1 ... ~l«~~.~~~"n 8
respond t~ ~OS-positive cytotoxic T cells {2536,2008), wh1cn
characteristically accumulate between tumour cells and ves-
Fig. 10.02 Prrmary difluse large 8-cell lyrnphoma of the CNS (CNS-DLBCL). Altera- sel walls {2536}. Intermingled with the tumour cells are reacn e
trons ot spcc.ifrc p;:.ihways contr1Dutmg to the pathogenesis of primary CNS-DLBCL. GFAP-positive astrocytes; prominently activated C045med
ASHM ati":narit cw,~w hyf1f.rn1utar1 or1, BCR. 8 ceil receptor; CSR, class-switch re-
curnb,;i.J1 ~fl S~:t1~ . •":IT.2L' iiy f,1:;1IT:!J\f:t(l' HI
(leukocyte common antigen), CD68+, HLA-DR+ microglta.
and macrophages .
1
, ,.,r 1. • 1rJ tut' . ' ' f,, . •11/1(1 ~ \f~~· ·r._ J ~:
.•
... ~

Af.10.03 Primary diffuse large 8-cell lymphoma of the CNS. A The lymphoma cells form dense sheets (right) and show the charactenstJc perivascular spread (left): B The lym-
phomacells inliltrate around blood vessels (left) and also into their walls (right). C The lymphoma cells are large, with vesicular nuclei. prominent nucleoli , and amphoph1ilc cytoplasm

R -
a~· l0.04 lmrnunophenotype of diffuse large 8-cell lymphoma of the CNS. A The tumour ceUs express the pan-8 -cell marker CD20 B Characteristic high proliferative
0~.'v~y evidenced by nuclear Ki -67 expression in the maiority of tu mour cells. C Strong nuclear expression ol MYC protein in the maiorlty of the tumour cells 1n the absence
01~c(sc rearrangement. D Expression of BCL 2 E St1ong nuclear staining of the tumour cells for IAF4 (MUM1). F The majoniy of the tumour cells show nuclear e>1pression

I lat=n mtol)mpho1d tumours rnvolvinQ the CNS 353


.
·~.I i I o
i ~ '. ~

... VI ID ,.... CO at 0 - ,.., )(. >-


~ ~ -5 ~.c_~~~~ ~ .s
Fig. 10.05 Diffuse large 8-cell lymphoma of the CNS. Copy-number profile. Note losses of chromosome arm 6q and homozygous deletion of CDKN2A on chromosome arm90
among other copy-number variations. The tumour also carried mutations in MYD88 and CD798, determined by next-generation sequencing (not shown).

lmmunophenotype Sentinel les ions


The tumour cells are mature B cells that are immunohistochemi- In rare cases , CNS-DLBCL has been repor ted to be preceded
cally positive for PAX5 , CD19, CD20 , CD22, and CD79a. lgM (for as long as 2 years) by demyel inating and inflammator /
and lgO, but not lgG , are expressed on the surface of tumour lesions similar to those occu rrin g in multiple sclerosis (63 .1386
cells {2155}, with either kappa or lambda light chain restriction . 1753}.
Most cells express BCL6 (60-80%) and IRF4 (MUM1) (90%),
whereas plasma cell markers (e.g. CD38 and CD138) are Cytology
negative. Fewer than 10% of all CNS-OLBCLs express CD10 Smear preparations per formed in the intraoperative setti g
{7181. whereas CD10 expression is more frequent in systemic show enlarged , highly atypical neoplastic cells that are dis-
DLBCL; therefore, C010 positivity in a CNS lymphoma with cohesive and have minimal detectab le cytoplasm, wh ic can
DLBCL characteristics should prompt a thorough investigation be useful in disting uishing CNS-DLBCL from metastatic neo-
for systemic DLBCL that might have spread to the CNS . HLA- plasms and high-grade gliomas . Anap lastic nuclear fea ures.
A, HLA-8, HLA-C, and HLA-DR are variably expressed , with apoptosis, and mitotic activity are typ ical. lntraoperative smear
approximately 50% of CNS-DLBCLs showing a loss of HLA is a useful techn ique for increasing diagnosti c security iri ti y
class I and/or class II expression {322,2675). BCL2 expression serial stereotactic biopsies to identify blasts {899 ). Final diag-
is common: 82% of CNS-DLBCLs have a BCL2high , MYChigh nosis requ ires further tumour cell characteri zation incl dir.g
phenotype {392}. The Ki-67 proliferation index is usually> 70% immunophenotyping .
or even > 90% {392}. Apoptotic cells may be frequent. Only The diagnostic value of cereb rosp inal fluid cytol ogy 1s li m -
isolated cases show evidence of EBV infection {2152), and the ited {1709). Meni ngeal disseminati on is d iag nosed in 15 .7% o'
presence of EBV should prompt evaluation for underlying immu- cases (of wh ich 12.2% are diagnosed by c erebrospinal fl id
nodeficiency. cytomorphology, 10.5% by PCR , and 4.1% by MRI) {1710)
Pleocytosis is found in 35 - 60% of cases and co rrelates wi:h
Corticoid-mitigated lymphoma meningeal dissem inati on 11 710). Ce ll counts may even be
Because the tumour cells are highly susceptible to steroid- normal. The cerebrospinal flu id harbours neoplastic cell s 1n a
induced apoptosis , CNS-DLBCL may vanish rap idly after cor- minority of patients with leptomen ingeal involvement , and tne1
ticosteroid treatment. Microscopically, neoplastic B cells may detection may requ ire re peated lu mbar puncture. Th e comb i-
then be present in only small numbers (or they may even be nation of cytolog ical and immunohistochemical analyses w1tn
absent) , and apoptotic debris may be abundant. Tissue sam- multiparameter flow cytometry may faci litate the detection o;
ples may show only nonspecific inflammatory and reactive cerebrosp inal fluid lymphoma ce lls {2858 ). PCR analysis o f
changes and/or necrosis; foamy macrophages are particularly the COR3 reg ion of th e IGH gene followed by sequenc1rig
frequent {389 ,718) . High numbers of large T cells with Ki-67 of the PCR products may identify a clo nal B-ce ll populat10
expression indicative of prominent activation may dominate, in the cerebrospinal fluid , but it does not enab le lymphoma
raising the possibility of T-cell lymphoma as a differential diag- classification . Elevated ce rebrosp inal flui d levels of severa
nosis. In some cases , PCR analysis of the CDR3 region of the microRNAs (miR-21 , miR-19 , miR-92a rniR -30) (196 ) and of
IGH gene may reveal a monoclonal B-cell population . However, CXCL13 plus IL-10 12742) have been repo~ted to rn st in guisr
pseudoclonality due to very low numbers of B cells poses a CNS-DLBCL from inflammatory and ot,...,e' C S oi sorders
problem.

354 Haeniatolyrnpnr-'1d h.vnours 1n 10111ng the CNS


.1 9 I Cumulative data suggest that pretherapeutic and post- Box 10.01 Diagnostic criteria for primary diffuse large 8-cell lymphoma of lh"! CNS
!reatment IL-10 levels 1n the cerebrosp inal fluid have prognos - (CNS-DLBCL)
:1c value 12246.2813) .
Essential:
Biopsy-proven mature large 8-cell lymphoma confined to the CNS at presentation
Diagnostic molecular pathology
PCR demonstrates clonal rearrangement of IG genes with the AND
introduction of somatic mutations . Expression of one or more 8-cell markers (CD20, CD19, CD22, CD79a. PAXS)

Essential and desirable diagnostic criteria Desirable:


See Box 10.01 lmmunohistochemical phenotype of late germinal-centre exit 8 cells (IRF4
[MUM1]+, 8CL6+/-, CD10-); CD10 expression does not exclude the diagnosis.
but it is uncommon and indicates possible systemic DLBCL
Staging
lmmunohistochemical positivity for 8CL2 and MYC
Staging at diagnosis includes CT of thorax and abdomen as
ell as bone marrow analysis to exclude systemic lymphoma . Absence of EBV-associated markers (in > 97% of cases)
Molecular detection of a clonal 8-cell populatJon in cases in which histology 1s not
Prognosis and prediction definitive, such as corticoid-mitigated CNS-DL8CL
CNS-DLBCL has a worse outcome than does systemic DLBCL.
However. a minority of younger patients may be cured . Older The transcobalamin missense variant TCN2:c .776C>G
patient age (> 65 years) is a major negative prognostic fac- p.P259R has been associated with shorter survival and neuro-
tor and is associated with reduced survival and an increased toxicity (1906). Most protocols report a median progression-free
risk of neurotoxicity {13,1709}. High-dose methotrexate-based survival of about 12 months and an OS of approximately 3 years.
f.X)lychemotherapy is currently the induction therapy of choice In a subgroup of elderly patients with CNS-DLBCL with a methyl-
{ 709}. The inclusion of consolidative whole-brain irradiation ated MGMT promoter, temozolom1de monotherapy appeared to
may improve progression-free survival, but it does not improve be therapeutically effective {1768f.
overall survival (OS) {3172}; moreover, it increases the risk of The presence of reactive perivascular C03 T-cell infiltrates
neurotoxicity resulting in cognitive dysfunction , especially in on biopsy has been associated with improved survival {2536).
patients aged > 60 years {13}. In eligible patients with CNS- LM02 protein expression by the tumour cells has been asso-
OLBCL. consolidative high-dose chemotherapy and autologous ciated with prolonged OS 11930}. BCL6 expression has been
s em cell transplantation after methotrexate-based polychemo- suggested as a prognostic marker in several studies, although
therapy is associated with prolonged survival and with mainte- conflicting conclusions as to whether it is a favourable or unfa-
nance of or improvement in cognitive outcomes and quality of vourable marker have been reported (2144,2570,2619,2990\ . In
fife. At recurrence, there is no standard approach and prognosis one study, del(6)(q22) was associated with inferior OS (434) .
1s poor.

Haematol /mpho1 tumours in olving the CNS 355


Immunodeficiency-associated CNS Deckert M
Batchelor T
~ J a g 8nP. ft
P a1Jf1J<j N
lymphomas Ferry JA
Hoang-Xuan K
Weller M

Definition
Immunodeficiency-associated CNS lymphomas comprise a
family of CNS lymphomas arising in patients with inherited or
acquired immunodeficiency, including that related to AIDS and
iatrogenic disease.

ICD-0 coding
None

ICD-11 coding
2832 Immunodeficiency-associated lymphoproliferative disor-
ders

Related terminology
Acceptable: AIDS-related diffuse large 8-cell lymphoma.

Subtype(s) Fig. 10.06 HIV-associated primary diffuse large B-cell lymphoma of the CNS ";
None case was in an HIV-infected patient In addition to the large tumour in the basatgar-
there are further foci in the contralateral insular region (arrowheads).
Localization
Immunodeficiency-associated CNS lymphomas typically mani-
fest in the CNS parenchyma.

Clinical features
The clinical presentation and imaging features of immunodefi-
ciency-associated CNS lymphoma may be similar to those of
CNS lymphoma in immunocompetent patients. Multiple lesions
and areas of necrosis occur more frequently in immunodefi-
ciency-associated CNS lymphoma than in CNS lymphoma of
immunocompetent patients (719}.

Epidemiology
Immunodeficiency-associated CNS lymphomas may develop
in rare hereditary immunodeficiency syndromes or (more com-
monly) in acquired immunodeficiency conditions related to
infectious , autoimmune, or neoplastic diseases, or to immu-
nosuppressive therapies. AIDS-related primary diffuse large
B-cell lymphoma of the CNS (CNS-DL8CL) has become less Fig. 10.07 HIV-associated primary diffuse large B-cell lymphoma of the CNS. i
common with the introduction of highly active antiretroviral in an HIV-infected patient.
therapy (HAART) (3327) . EBV-positive DLBCL of the elderly
is related to immunosenescence and occurs in patients aged infections that lead to immunodeficiency also increase tne ·
> 50 years (2356) . of CNS lymphomas, as does immunosenescence.

Etiology Pathogenesis
Immunodeficiency syndromes underlying immunodeficiency- E8V infection is important because most immunodehaen.:.
associated CNS lymphoma include ataxia telangiectasia, associated lymphomas are EBV-related.
Wiskott-Aldrich syndrome , and lgA deficiency. Other under-
lying conditions include autoimmune disorders (e.g. systemic Macroscopic appearance
lupus erythematosus and Sjogren syndrome), neoplastic dis- Multifocal presentation is more frequent than in CNS lympn011'.:i~
eases, and iatrogenic immunosuppression (either for the pur- in immunocompetent patients, as is a tendency to contain r--' ~
pose of organ transp lantation or due to treatme nt with immuno- and larger areas of necrosis. Tumours may simulate necrotL-~
suppressive drugs). Infectious disorders such as HIV and HTLV cerebral toxoplasmosis , which may occur concomitantly ! .J02~·
. -
ffl.18.0I EBV+ diffuse large 8-cell lymphoma of the CNS. A Large tumour cells clustering around a blood vessel are stained by the pan-8-cell marker CD20. B There is
stoog expression of EBV-encoded small RNA (EBER) in the nuclei of the tumour cells.

Histopathology Box 10.02 Diagnostic criteria for immunodeficiency-associated CNS lymphoma


Immunodeficiency-associated CNS lymphomas are typically Essential:
EBV-related, so in addition to 8-cell markers such as CD19, A 8-cell lymphoma confined to the CNS at presentation, in an immunodeficient
CD20, and CD79a, the lymphoma cells express the EBV- patient
associated proteins EBNA1-6 , LMP1 , and EBV-encoded small AND
RNAs 1 and 2 (EBER1 and EBER2) {1669}.
EBV positivity of tumour cells demonstrated by immunohistochem1stry or in situ
Otherwise, AIDS-related CNS-DLBCL shares the characteris- hybridization
tics of CNS-DLBCL in immunocompetent patients.
Desirable:
Cytology Demonstration of a clonal 8-cell population by PCR (in diagnostically difficult
Cytology is of limited value for immunodefic iency-associated cases)
CNS lymphomas .

Diagnostic molecular pathology Staging


Demonstration of a clonal B-ce ll popu lation may be helpful in Not clinically relevant
diagnostically ambiguous cases .
Prognosis and prediction
Essential and desirable diagnostic criteria Overall , outcome for immunodeficiency-associated CNS lym-
See Box 10.02. phoma is poor /1063) .

Haernatolympho1d tumours invot 1nq the CNS 357


Lymphomatoid granu lomatosis Deckert M
Batchelor T
N agan~ M
Paulu~ 'JI
Ferry JA Weller M
Hoang-Xuan K

Definition lesions along perivascular and periventncular spaces r alfP;;;,,


Lymphomatoid granulomatosis is an angiocentric and angiode- been reported (16231 . - # ,

structive lymphoproliferative disorder characterized by poly-


morphous lymphoid infiltrates composed of EBV-positive atypi- Epidemiology
cal B cells in a T cell- rich inflammatory background . This rare disease usually manifests in adults 1n the hf: ::. :!
sixth decade; a male predilection has been repor ed •5 :
ICD-0 coding 2485}.
9766/1 Lymphomatoid granulomatosis
9766/1 Lymphomatoid granulomatosis , grade 1 Etiology
9766/1 Lymphomatoid granulomatosis, grade 2 Lymphomatoid granulomatosis is an EBV-driven disease
9766/3 Lymphomatoid granulomatosis, grade 3 Immunodeficiency increases the risk of lymphoma a·d gr-~_,­
lomatosis.
ICD-11 coding
2A81 .3 Lymphomatoid granulomatosis Pathogenesis
Disease is hypothesized to result from defective immu os1..: __
Related terminology lance of EBV and an abnormal immune response towarc!s EB ·
None (2064).

Subtype(s) Macroscopic appearance


None Macroscopically, the lesions may resemble tumour o arc.-
like areas of necrosis.
Localization
The brain may show multiple focal intraparenchymal lesions . Histopathology
Leptomeninges and cranial nerves may also be involved (2423}. Lesions are characterized by polymorphous lymphoid 1 -
The spinal cord may also be affected (1620} . trates consisting of lymphocytes (including CD4+ and CD"'
T lymphocytes), and plasma cel ls intermingled with vary
Clinical features numbers of atypical EBV+, CD20+, CD30+/- , C0 15- large
This angiocentric an d angiodestructive lymphoproliferative neoplastic B cells . Grading of lymphomatoid granuloma.
disorder affects the brain in 26% of patients, who may present depends on the proportion of EBV-expressing CD20+ B ce
with focal neurological symptoms and/or headache or cogni- (see Table 10.01 ). Most cases affecting the CNS corres
tive impairment (2423}. Presentation may even be indistinct to grade 3, with large aggregates of EBV+ CD20+ B ce Is. a1':
from that of systemic manifestation (1623,1272). MRI may reveal they should be classified as EBV+ diffuse large B-cell
mass lesions throughout the CNS (1272}; multiple punctate phoma of the CNS . The infiltrates invade blood vesseJ

.. ~ ..
.. .. ..
..

•.

...
~rq. \O.t19 l yrnr.h0nia101d granulomatus1s. A Ang1ocentric lymphoid lesion harbouring some large cells with prominent nu~leoli. a Polymo . h • •I • h ·d fil•- ... ;
· · nc•r L t d d · d .hh . rp ous ymp 01 in 1uQlv
.., , , . . ; I ',11 111.a yrnp 10cy1es pre orninate. a m1xe wit 1st1ocytes and some plasma cells. Lymphocytes show some irregularity with slightly increased nuclear Slle ~
11
cr1;.,s8a nuclear basoph1l1a. C Enlarged CD20+ B cells as part of a lymphoid infiltrate within a blood vessel wall. Note that som 1' d h · ..i.....i - l'&iC:
par&nr:nirna. e en arge 8 ce 11 s ave also 1nvaU1;\l IUV Uil'Ol

358 H aL:111 a to lympt1r,1cl tur n0 urs involving tt1e CNS


Rg.10.10 .Lymphomatoi~ granulomatosis, CNS WHO grade 3. A An angiotropic lymphoma with infiltration of the blood vessel walls by blasts, lymphocytes, and hist1ocytes.
Blasts also invade the penvascular brain tissue, which is largely necrotic. e Note the high number of EBV-encoded small RNA (EBER)-positive cells . C Tumour cells express the
pan-B-cell marker CD20. D Tumour cells express the CD30 antigen .

Tablt10.01 Grading of lymphomatoid granulomatosis (adapted from the 2017 WHO classification of tumours of haematopoietic and lymphoid tissues)
--~~~~~~~~~~~~--..,

L.ymphald cell• Transformed B cells EBER+ lymphoid cells• Necrosis


--·- Focal or absent
Grade 1 Polymorphous background Rare or absent < 40 cells/mm2 (< 5 cells/HPF)
Grade2 Polymorphous background Single or in small clusters 4G--400 cells/mm2 (&-SO cells/HPF) More common

Grade 3 Polymorphous background Large atypical B cells or aggregates 2


> 400 cells/mm (> 50 cells/HPF) Extensive

EBER. EBV-encoded small RNA.


'Originally reported per HPF of 0.16 mm2•

lax10.03 Diagnostic criteria for lymphomatoid granulomatosis Essential and desirable diagnostic criteria
See Box 10.03 .
Essential:
Morphology of an lntracerebral polymorphous lymphoid infiltrate with atypical Staging
EBV+, CD20+, CD30+/- , CD15- large neoplastic B cells of variable numbers Not relevant
AND
Bklod vessel destruction Prognosis and prediction
Lymphomatoid granulomatosis of the CNS usually follows an
aggressive course because most CNS biopsies demonstrate
and may induce infarct-like necrosis of tumour and/or brain tis- EBV-positive diHuse large B-cell lymphoma of the CNS . Cur-
sue Rarely, CNS WHO grade 2 lesions may occur in the CNS . rent treatments of lymphomatoid granulomatosis are based on
its histological grade, although clinical courses and treatment
Cytology responses are largely unclear !1623], particularly in cases with
Not relevant CNS involvement. Since most cases of CNS lymphomato1d
granulomatosis correspond to diffuse large B-cell lymphoma of
Diagnostic molecular pathology the CNS, treatment options include corticosteroids , radiation . or
Detection of clonal rearrangement of IG genes may be helpful in chemotherapy !12721 .
d1agnost1cally difficult cases .

Haematolympho1d tumours 1r,vo lving me CNS 359


Deckert M f'lagane M
lntravascular large B-cell lymphoma Batchelo r T Paulus 'N
Ferry JA WeJler M
Hoang -Xuan K

Definition
•' .. ~

lntravas?ular large B-cell lymphoma is a distinctive type of


aggressive B-cell lymphoma characterized by exclusively intra-
vasc ular growth .

ICD-0 coding
9712/3 lntravascular large 8-cell lymphoma

ICD-11 coding
~
•t
2A81 .1 lntravascular large B-cell lymphoma

Related terminology , -
Not recommended: angiotropic large cell lymphoma. •
Subtype(s)
.••
-.
None
• :.-- •
Localization Fig. 10.11 lntravascular large B·cell lymphoma. Neoplastic lymphoma
The brain is nearly always involved ; spinal cord involvement is fined to the vessel lumen.
less common.
Etiology
Clinical features Unknown
Except for the solely cutaneous cases, CNS involvement occurs
in 75-85% of cases 1251). The hallmark intravascular growth Pathogenesis
leads to clin ical symptoms mim icking those of cerebral infarc- Absence of CD29 and CD54 (ICAM1) expression is a •a,
tion or subacute encephalopathy [954). to underlie the tumour cells' inability to migrate transvascutan
{2535}. Expression levels of the chemokine receptors CXCR5.
Epidemiology CCR6, and CCR? are decreased, and MMP2 and MMP9 are
lntravascular large 8-cell lymphoma is rare and usually mani- not expressed . Thus, the tumour cells express mo lecules
fests in adults. The median patient age is 70 years (range: enable their adhesion to the endothelium but not those invol eo
34-90 years). There is no sex predilection 12538). in extravasation {2538}.

..
• . •.'
,.
I

A • B
... •

Fig. 10.12 lntravascular large B-cell lymphoma. A lmmunostaining of vascular endothelium for CD34 highltghts the mtra~ascular I t h h I'" I~·
1ntravascular accumulat1on ot CD20+ lymphoma cells. °
oca ion 1 t e 1ymp oma ce ~ ~
Box 10.04 01agnost1c criteria for intravasr:ular large 8-r,P,ll iyrnoti'Jm:j
M.lltort)Acoplc appearance
~ C'"'SC PY reveals infarcts (acu1e and/or old) . necrosis and/or
Euentl81:
~, morrhage , although abnormalities may be 1nconsp•cuous Biopsy showing a 1ait;1e a-cell lymphoma with morpt1ofog1cal and 1mmunoli
tochemical con nement of neoplastic B cells to lhe blood v sel tum~n w out
H pathology I vasion of the surrounding tissue
~K"'Osrop1cally ,
large atypical B cells are present . confined to
l""e lumma of cerebral blood vessels ; they may occlude these
~els . but they do not invade into the brain parenc hyma ,
?.ttl'1ough . exceptionally. a few cells may extravasate. lmmuno- Essential and desirable diagnostic criteria
" tochem1cally, the neoplastic B cells typically show a strong See Box 10 04
ression of CD20 . However, exceptional C020-negative
cases have been reported that require immunostaining for addi- Staging
Staging should determine involvemen t of .other organs because
,al B-cell markers (e.g . CD79a and PAX5) 12538).
intravascular lym phoma may be widely d1ssern1nated

Cytology
Not relevant Prognosis and prediction
The prognosis of intravascu lar large 8 -c ell lyn p horna ' r ~o v r ~
the CNS IS poor Methotrexate -b ased chemotherapy IS ue,.., efi-
Olagnostic molecular pathology
cial in a subset of pati en ts [1582)
Detection of clonal rearrangement of the IG genes may be help-
ful for the diagnosis .

Ha •rr 1,1!olyrnph0id tumours 1nvolv 1ng the CNS 361


J' . . J

3306.1029) . No racial or geographical association s have been Box10.05 Diagnostic criteria for MALT lymphoma of the dura
identified .
Essential:
Etiology A dural-based lymphoma composed of tumour cells with r;iorph.ological
characteristics of marginal zone B cells with small or med1um-s1zed nuclei and
MALT lymphoma outside the CNS has been attributed to
pale cytoplasm
chronic inflammation (of either Infectious or autoimmune ori-
gin) {3171}. Regarding MALT lymphoma of the dura , it is still AND
unknown wheth~r the~e is an association with inflammatory dis- lmmunohistochem1cal expression of 8-cell markers (CD20, CD79a) with
demonstration of meshworks of CD21+, CD23+, CD35+ follicular dendritic cells
orders. One patient with hepatitis C who developed dural MALT
lymphoma has been reported {3328) . One patient with CNS
Desirable:
extranodal marginal zone lymphoma had a long history of white
FISH analysis for trisomies and MALT lymphoma-associated translocations
matter disease with some features of multiple sclerosis {2834},
Gene rearrangement studies to document clonality, if not already proved by
and another ~atient with extranodal marginal zone lymphoma
had Chlamydia psittaci infection {2537}. immunophenotyping

Pathogenesis
Trisomies, most often of chromosome 3, are occasionally lmmunohistochemistry
detected {3237,3306}. Inactivation of TNFAIP3 by mutation Neoplastic B cel ls are CD 20+, CD79a+, CD5- , CD10- , BCL6- ,
or loss appears common in cases with plasmacytic differen- CD23-, JRF4 (MUM1 )+/- , cyclin 01- , BCL2+, and they have a
tiation {1029}. Activating NOTCH2 mutations accompanied low proliferation in dex {1029,163 ,2274). A component of cl onal
by inactivating TBL 1XR1 mutations are common in cases with plasma cells is often found {1 536,3306). The monotypic plasma
monocytoid morphology {1029). Recurrent gains of 6p25 .3 and cells are often JgG4+ (3306 ,1029), but evidence of systemic
losses at 1p36.32 have been documented . IGH translocation JgG4-related disease has been absent to date {3306 ).
and MALT lymphoma-associated translocations are rare {163 ,
2274). but a translocation involving the MALT1 and IGH genes , Cytology
consistent with IGH::MALT1 fusion [t(14;18)(q32;q21)] , has been The cerebrosp inal fluid is occasionally invol ved [1428}.
identified in 1 case {263}.
Diagnostic molecular pathology
Macroscopic appearance IG genes are clo nally rearranged [1759,263 )
There is a solitary mass or plaque-like thickening of the dura
!1759,3237.1428,1536,3306}, often mimicking a meningioma Essential and desirable diagnostic criteria
!1 63}. See Box 10.05 .

Histopathology Staging
Dural MALT lymphomas share histological and immunohistologi- MALT lymphomas of the dura are typicall y localized at presen -
cal features with MALT lymphomas at other sites. They are com- tation {1759,3237,1428 ,3306}. Clinical stag in g is performed to
posed of small lymphocytes and marginal zone cells , often with exclude extracranial manifestations.
plasmacytic differentiation, sometimes with remnants of reactive
follicles with follicular colonization {1759,1534,3237,1428,3306). A Prognosis and prediction
subset of cases have tumour cells with abundant clear cytoplasm Patients are treated with res ection, radiation, and/or chemo-
(monocytoid morphology) !1029). Occasionally, associated amy- therapy, and they typ ically ac hieve c omplete remission (585 ,
lo1d deposition is seen {1847,3237). Dural MALT lymphomas 1029,163 ,2274 ,3071). The p rogn osis is very good . One case
may arise in association with meningothelium (just as marginal series reported 22 compl ete remi ssions and 1 partial response
zone lymphomas often arise in association with epithelium in in 23 evaluable patien ts. with a 3-year progression-free sur- •
other sites) , so entrapped meningothelial cells may be present vival rate of 89% ! 709) . Local and systemic relapses are rare :.
1
11759,1847). Infrequently, lymphoma cells invade Virchow- Robin !1 428,709).
spaces and the subjacent brain parenchyma /3306).

Haornaiolymphoid tumours involving the CNS 363


Other low-grade B-cell lymphomas Deckert M
Batchelor T
Nagane M
Paulus W
of the CNS Ferry JA
Hoang-Xuan K
Weller M

Definition 2274 ,3071}. Symptoms are variable, and they are occasio -
Oth er low-grade B-cell lymphomas of the CNS are those lym- ally present for months or years before diagnosis {2537.11881
phomas confined to the CNS at presentation that histologically The most common symptoms and signs include headache.
correspond to one of the types of systemic low-grade B-cell seizures, and speech impairment (2387,165,1188.307 11 The
lymphomas , most commonly extranodal marginal zone lym - clinical differential diagnosis can include glioma, intravascu-
phoma; other cases have been diagnosed as small lymphocytic lar lymphoma , and demyelinating disease (163 ,2274). Radio-
lymphoma, lymphoplasmacytic lymphoma, or low-grade B-cel l graphically, lesions may be well defined or infiltrative (2387.
lymphoma NOS. 3248,165).

ICD-0 coding Epidemiology


9671 /3 Lymphoplasmacytic lymphoma Low-grade B-cell lymphomas of the CNS are rare , and epide-
9690/3 Follicular lymphoma miological data are not available.

ICD-11 coding Etiology


2A85.Y Further specified mature B-cell neoplasms or lymphoma The etiology is unknown.

Related terminology Pathogenesis


None Limited data are available on genetic alterations in low-grade
B-cell lymphoma arising in the CNS . Clonal rearrangement
Subtype(s) of IGH and/or IGK has been reported {2950,1050 ,2274). One
None case with loss of heterozygosity on 6q (including the locus for
TNFAIP3) and one with MALT1 rearrangement are on record
Localization {2274) . In the few cases tested , MYDBB p.L265P was absent
Tumours may occur as a solitary lesion or (infrequently) as {2274}, and FISH for translocations of IGH, BCL2, and CCND1
multiple discrete lesions , or as diffuse involvement of the white was negative {1586,163).
matter of the brain. Lesions are mostly supratentorial (affecting
lobes of the cerebral cortex or basal ganglia) (2387,163,1188, Macroscopic appearance
2274). but the cerebellum and spinal cord also may be affected Limited data are available on macroscopy.
{2387,3248). Involvement of the leptomeninges and choroid
plexus has been re ported [1586 ,2537,1050,163). Histopathology
Most cases of low-grade B-cell lymphoma of the CNS cor-
Clinical features respond to extranodal marginal zone lymphoma {3248,22741.
Male and female patients are roughly equally affected . Patients other cases have been diagnosed as small lymphocytic lym-
are almost all adults, with only rare paediatric cases being phoma {165,1188}, lymphoplasmacytic lymphoma !2950). or
reported (age range : 5-79 years ; median : 49 years) [2387,163, low-grade B-cell lymphoma NOS {1446 ,2387).

.·.
.. ::,·. ..
' ..,.. ..
.. " \ ..
... .
.. . ..
.. •
.. '-:
• • ·~ I
-~

... . . ........
_ . . \..
\ ,.
. ....

. . c .
fl • 10.~~ Low-grao•. fl ce!l I/! 1priw1a A Pwvascular cuffs of small, monomorp.hic-appearlng lymphoid cell~ . a Perivas~ ta . ff . . f d' .· · ' •
ce:is wn1c'1 expre-s<:: I~ e par: 8 cell rnarkei CD£0 C Admixed with rhe lymphoma cells are some reactive CD3.T cells. u r cu so me 1um-s1zed, monomorphlc lymphoma
Diagnostic criteria for other low-grade B-cell lymphomas of the CNS Diagnostic molecular pathology
Clonal rearrangement of IGH and/or lGK genes has teen
~ a lymphoma confined to the CNS al presentation that htstologi- reported 12950.1050 2274}
.__,, ..........,.,,nds to one of the types of systemic low-grade B-cell lymphoma
Essential and desirable diagnostic criteria
~: See Box 10.06.
oemctJon of B-cell clonality by PCR
Staging
Extent of disease 1s established using lumbar puncture for
cytology and flow cytometry. complete blood count with differ-
tmmunophenotype ential, bone marrow biopsy with flow cytometry. and 1mag1ng to
Neoplastic c ell s are typically CD20+, CD3 -, CD5- , corn-, investigate sites of disease outside the CNS
C023-. BC L6- , BCL2+ , cyclin 01 - , TdT-, EBV-encoded small
RNA (EBER)- {2834 ,3248 ,2274) . When present , plasma cells Prognosis and prediction
(CD138+) often express monotypic light chain. The proliferation Low-grade B-cell lymphomas show a less aggressive course
index 1s low(< 10%) {165,1188). and have a better prognosis than do primary d1ff use large B-cell
lymphomas of the CNS. Patients have been treated with res~c­
Cytology tion , steroids, radiation, and/or chemotherapy Most are alive
Cerebrosp inal fluid is negative in most cases , but occasional and tree of disease or have stable disease on follow-up 13407.
cases have cerebrospinal fluid involved by lymphoma, with the 163,2274,3071 }. Progression outside the CNS is very rare . A
diagnosis confirmed by flow cytometry {2834 ,1050,1188}. small subset of patients succumb to lymphoma (22741 .

Haematolympho1d tumours involving the CNS 365


Anaplastic large cell lymphoma Deckert M
Batchelor T
Naqa P. M
Paul •Js 'fl
(ALK+/ALK-) Ferry JA
Hoang-Xuan K
'Neller M

Definition
Anaplastic large cell lymphoma (ALCL) is a distinctive CD30-
positive peripheral T-cell lymphoma that is rare in the CNS and
is separated into two distinct types: ALK-positive (ALK+ ALCL)
and ALK-negative (ALK- ALCL) .

ICD-0 coding
9714/3 Anaplastic large cell lymphoma (ALK+/ALK-)

ICD-11 coding
2A90 .A Anaplastic large cell lymphoma, ALK-positive
2A90.B Anaplastic large cell lymphoma, ALK-negative

Related terminology
None

Subtype(s) Fig. 10.15 CNS anaplastic large cell lymphoma. There is a polymorphic lymphoma..
None tous infiltrate with occasional markedly atypical hallmark cells.

Localization Epidemiology
ALK+ ALCL occurs as sin gle or multiple supratentorial pare n- ALK+ ALCL occurs from early childhood to young adulthOOd
chymal lesions with or without infratentorial involvement, and (m edian age: -17 years) , with a male preponderance. ALK-
rarely with spinal cord involvement. Extension to involve the ALCL affects adults (median age: 65 years), also with a ma.a
meninges and (rarely) the skull can occur (2751,2398,3447, preponderance (2275}.
776).
ALK- ALCL occurs as single or multiple lesions, usually Etiology
supratentorial (1057}. Unknown

Clinical features Pathogenesis


Patients present with headache, seizures, nausea, fever, or a ALK+ ALCL is driven by oncogenic ALK gene fusions . mos:
comb ination of these {251 ,2275,2398,1760) . They are often ini- commonly with NPM1 (in > 80% of cases) . ALK fusions i nc rea~
ti ally thought to have an infection (916,2398,1017,776). ALK expression, leading to aberrant activation of downstrea'T'
signalling pathways including JAK/STAT3 and others 13420!.

- ~
H~.. 'Ht1@ ,-, ,. + c!. a.r.1as11c ,.,ge c..·11 l'('li-''..•:irna
r; .•. 1"J1 sr1c~1 ey.oplasrn1c expres~1ori :>I AL K

366
Bu 10.07 Diagnostic criteria for anaplastic large cell lymphoma (ALK+IALK-) includes classic Hodgkin lymphoma and . diffuse large . B-cell
• I: lymphoma with pleomorphic cells . Hodgkin lymp~oma is 'lery
rare in the CNS [1064), typically has more admixed reactive
Biopsy showmg an aggressive lymphoma confined to the CNS that is histologically
cells and cells express PAX5 (weakly) and often CD15, 1n addi-
~ of CD30+, large neoplastic lymphoid cells that are negative for B-cell
l'l'\8J\ers and can be ALK+ or ALK- tion t~ CD30 . In contrast to ALCL , diffuse large B-cell lymphoma
is typically positive for CD20 and other B-cell antigens .
Desirable:
Expression of T cell-specific antigens. and/or gene rearrangement studies Cytology . .
st.owing clonal TR genes The cerebrospinal fluid may be involved . The l~r~e atypical
ASH showing ALK rearrangement (for ALK+ anaplastic large cell lymphoma) neopl astic cells may have cytoplasmic azuroph1l1c granules
{3447,2081 l.

ALK~ ALCL carries mutations or gene fusions of other receptor Diagnostic molecular pathology
tyrosin~ ~1nase genes that eventually activate signalling path- Molecular analysis demonstrates clonally rearranged TR genes
ways similar to those activated in ALK+ ALCL , Including JAK/ in the vast majority of tumours . ALK+ ALCL carries chromo-
STAT3 !653}. Clonal rearrangements of T-cell receptor genes somal translocations involving ALK, most commonly a t(2:5)
are present in the vast majority of cases of ALCL. (p23;q35) causing an oncogenic fusion with NPM1 (NPM1::ALK
fusion ).
Macroscopic appearance
Insufficient data available Essential and desirable diagnostic criteria
See Box 10.07.
Histopathology
ALK+ ALCL shows a diffuse proliferation of large atypical cells Staging
1th abundant cytoplasm, including hallmark cells with bean- Staging is requ ired to exclude a systemic primary lymphoma .
shaped nuclei and an eosinophilic paranuclear area (2081 ,
1057.2751 .1760}. Rare examples of the lymphohistiocytic and Prognosis and prediction
small cell patterns have been described (2539,3447}. Tumour ALK+ ALCL of the CNS has a prognosis similar to or worse than
cell s are CD30+, ALK+, and EMA+, and they may express one that of systemic ALK+ ALCL , although sustained remission is
or more T-cell antigens. possible . Treatment failures tend to occur in the CNS and are
ALK- ALCL histopathology is similar to that of ALK+ ALCL , rarely systemic. ALK- ALCL has a poor prognosis (1057,2275 ,
but ALK is not expressed (1057}. The differential diagnosis 3336 ,2081 }.

Haematolymphord l
· umour$ 1.nvoh11ng the CNS 367
T-cell and NK/T-cell lymphomas Deckert M f' laqaM M
Batchelor T Paulu s N
Ferry JA Weller M
Hoang-Xuan K

Definition headache, vomiting (1175}, weakness , paralysis, apha"'ia


Primary CNS T-cell and NK/T-cell lymphomas are groups of (2553), dysphagia , dysarthria (1866}, and mental deteriora !Of'
malignant non-Hodgkin lymphomas, including peripheral T-cell (2303) . Manifestations can mimic a cerebrovascular acc1de ~
lymphoma (PTCL) and NK/T-cell lymphoma (nasal type) , with (2303 ,2553} .
primary manifestation in the CNS .
Epidemiology
ICD-0 coding PTCLs accounted for 2- 4% of primary CNS lymphomas in a
9702/3 T-cell lymphoma European series {914), and as many as 17% of primary CNS
9719/3 NK/T-cell lymphoma lymphomas in an Asian series (583} . Extranodal NK/T-cell lym-
phoma is rare . Most reported patients were Asian {1175.2303
ICD-11 coding 2135,1866); rarely, patients were African -American 12553.21351
2B2Z Mature T-cell or NK-cell neoplasms, unspecified or Hispanic {2135) .

Related terminology Etiology


None Rare T-cell lymphomas {1809) and one extranodal NK!T-cel
lymphoma (614} of the CNS have been reported in HIV-pos1·ve
Subtype(s) patients , and one CNS NK/T-cell lymphoma has been reported
None in a transplant recipient {2135}, suggesting that immunodefi-
ciency might play a role in the etiology of the disease in a subset
Localization of patients. One patient with a CD4-positive primary CNS T-cell
In a series of 45 patients with primary CNS T-cell lymphoma, lymphoma was seropositive for HTLV-1 {1 931 }.
19% had positive cerebrospinal fluid cytology, 4% had eye
involvement, and 29% had multiple brain lesions (2902,2072}. Pathogenesis
Extranodal NK/T-cell lymphoma usually forms a single mass, Mutations of DNMT3A, TET2, and several JAK/STAT pathway
but it may also occur with multiple lesions (1175,2135) . Involved genes have been found in a subset of cases {2072} .
sites include the frontal and temporal lobes, cerebellum , pitui-
tary (2135,1866}, and (rarely) the leptomeninges (66}. Macroscopic appearance
Necrosis is common in extranodal NK/T-cell lymphomas {1175.
Clinical features 2135} and T-cell lymphomas [2072}.
Patients with primary CNS T-cell lymphoma are adults aged
21-81 years (median: sixth decade), with an M:F ratio of about Histopathology
1.5:1 (2072}. Patients present with headache, neurological Most T-cell lymphomas in the CNS are PTCL-NOS {2072,21601.
decline, seizures, and abnormalities of sensory and/or motor A minority of tumours (17% in one series) are anaplastic large
function {2072,2 160). cell lymphoma (see Anaplastic large cell lymphoma (ALK?/
Extranodal NK/T-cell lymphoma of the CNS affects patients ALK-), p. 366) {2072} . Most tumours in PTCL-NOS are com-
aged 21-77 years (median: fifth decade). with a male prepon- posed of small and/or medium-sized atypical lymphoid celts.
derance (M:F ratio: -2:1) (2135) . Patients present with symp- a minority are composed of medium-sized to large cells or
toms that are often rapidly progressive, including dizziness, mainly of large cells {583,1446 ,802,2072}. PTCL often snows
a prominent perivascular growth pattern and areas of necrosis Box 10.08 Diagnostic criteria for T-cell and NK/T-cell lymphomas
!583 , 1~4?,2902 , 80~.~072~ . Tumours are accompanied by reac -
T-cell lymphoma
t1v~ ghos1s an? a h1~t1ocyt1c reaction , but plasma cells, neutro-
ph1ls, and eos1noph1ls are usually inconspicuous {802 2072) Essentlal:
Extranod~I NK!T-cell lymphomas are composed of' medi~m ­ Biopsy-proven lymphoma manifesting in the CNS
sized, ~ed1um-s1~ed. and large, or large atypical , pleomorphic AND
lympho 1? cells . with irregular nuclei (featuring dark or coarse Expression of one or more T-cell markers 1n variable combmation (CD2, CD3 CD4,
chromatin and inconspicuous nucleoli) and a moderate amount CD8, CDS, CD7)
of p~le c~toplasm .{1175,2303 ,2135). accompanied by vascular Desirable:
proh~erat1on and h1stlocytic infiltrates (1866J . Mitotic figures are Abnormal T-cell immunophenotype with loss of one or more pan-T -cell antigens
readily found, and angiocentric growth and extensive necrosis
Demonstration of clonal TR gene rearrangement, if inflammation is considered in
are common {2135). Necrosis is coagulative and apoptotic bod -
the differential diagnosis
ies a.re ~lso seen (1866) . A single case of a collision tumour with -- - - - - - - - - -
mernng1oma has been reported (3515). NK/T-cell lymphoma _ __ __ _ ______ _
-------- - - -
Essential:
lmmunophenotype Biopsy-proven lymphoma manifesting in the CNS
Neoplastic cells in CNS PTCL are usually CD3-positive and AND
C056-negative; CDS is expressed more often than CD4 (2072) . NK-cell lmmunophenotype (typically CD3+, CD2+, CDS-, CD?+. CD56...)
Most tumours are of a~ T-cell lineage, but y8 lineage has been
OR
documented in a few cases {2072,2160). They typically express
Cytotoxic T-cell immunophenotype (typically CD3+, CD8+)
one or more cytotoxic granule proteins (TIA1 , granzyme B, and/
or perforin). CD2, CDS , and CO? are each lost in subsets of Desirable:
cases {2072}. Proliferation index is usually > 50% {2072). A Expression of cytotoxic molecules (perforin, granzyme BJ
case with aberrant coexpression of CD20 and CD79a has been Absence of clonal TR gene rearrangement in cases of N~-cell lin:_age
reported {1192}.
Extranodal NK/T-cell lymphoma is usually positive for CD3 ,
C056, cytotoxic granule protein , and EBV-encoded small RNA Essential and desirable diagnostic criteria
(EBER), and negative for COS and B-cell antigens {2135}. See Box 10.08 .
Occasional cases express CDS or lack C056 {2303,2135).
CD30 may be expressed {1866}. A single case showed aber- Staging
rant CD20 expression 11866). Staging is requ ired to exclude a systemic primary; for cases
of extranodal NK/T-cell lymphoma in particular, the poss1b1lity
Differential diagnosis of an occult primary in the upper respiratory tract should be
The differential diagnosis of T-cell lymphoma often includes a excluded.
reactive inflammatory process because of the often relatively
small size of tumour cells {2072,1192). Pan-T-cell antigen loss Prognosis and prediction
and clonal TR favour a PTCL; a polymorphous infiltrate with The outcome for patients with CNS T-cell lymphoma appears
admixed inflammatory cells favours a reactive process {802, similar to that for patients with primary diffuse large B-cell lym-
2072,1192}. phoma of the CNS . T-cell lymphomas with low-grade histologi-
The differential diagnosis of NK/T-cell lymphoma includes cal features may be associated with a more favourable outcome
B-cell lymphoma, especially lymphomatoid granulomatosis, than high-grade T-cell lymphomas (1446) . In one series, 20°0
because of the angiocentric growth and necrosis. lmmunohis- of patients with PTCL-NOS were alive without lymphoma ac last
tochemistry distinguishes the two entities. If the biopsy mainly follow-up , whereas 40% were alive with disease and 40~o had
shows necrosis, an infectious lesion can be considered in the died of the disease {2072). Methotrexate-based chemotherapy
differential diagnosis. is the most common treatment regimen used. In a cohort of
45 patients with primary CNS lymphoma of T-cell origin. the
Cytology median progression-free survival and overall survival were 22
Cerebrospinal fluid involvement is reported in extranodal NK/T- and 25 months, respectively [2902}
cell lymphoma with leptomeningeal disease; involvement can The prognosis of patients with extranodal NK/ T-cell lymphoma
be documented by cytology augmented by flow cytometry 1661 of the CNS 1s poor, and most patients experience rapidly pro-
gressive disease. Median survival 1s approximately 6 months
Diagnostic molecular pathology (range: 1- 18 months) [1175 ,2553 .2"1351 Long-term survival 1s
Clonal rearrangement of TR genes can be demonstrated in rare {2135) . Because primary CNS i\JK/ T-cell lymphomas are
most T-cell lymphomas (2072 ,11921. In extranodal NK/T-cell rare, no standard treatment protocol exists Methotrexate-based
lymphomas , TR genes are usually in germline configuration, but cl1emotherapy may be beneficial {25531.
they may be clonally rearranged 11175.2303)

Haematolympl101d tumours involving the CNS 369


Erdheim- Chester disease Pf3ultJS W
Chan JKC
ldba1 h A
Perry A
Sahm F

Definition
Erdheim- Chester disease of the CNS or the meninges, with
or without systemic lesions, pathologically corresponds to its
counterparts occurring elsewhere. It is a clonal histiocytosis
with foamy histiocytes, occasional Touton giant cells , chronic
inflammation , and variable fibrosis .

ICD-0 coding
9749/3 Erdheim-Chester disease

ICD-11 coding
2831.Y & XH1VJ3 Other specified histiocytic or dendritic cell
neoplasms & Erdheim-Chester disease

Flg.10.18 Erdheim-Chester disease. This 39-year-old man presented With ,


Related terminology
aches and severe proptosis. A T1 -we1ghted postcontrast axial MRI s
None proptosis with intensely enhancing 1ntraconal retrobulbar masses in both
cavernous sinus and pituitary gland are infiltrated and intensely enhancing. 1
Subtype(s) additional enhancing mass along the tentorium and postero1nfenor falx. I Tl·
None ed postcontrast sagittal MRI shows a mass infiltrating the pituitary gland anj
thalamus, extending inferiorly along the clivus. Multiple lobulated masses are
along the falx and tentorium. This is an important scan because 11 shows ~ _
Localization
hancing lesions in the pons, indicating parenchymal involvement.
All parts of the CNS may be involved, including the dura (in
15-25% of cases), cerebrum (60%), hypothalamus (3%), brain-
stem and cerebellum (25%), and spinal cord (12%) {1781,2402, Pathogenesis
492}. Neurodegenerative lesions are typically located in the Common alterations include BRAF p.V600E mutations (1~ as
posterior fossa , including in the brainstem, cerebellar pedun- many as 50% of cases) 1168,1241,1300.845}. MAP2K1 IT"\J a·
cles , and dentate nuclei. tions (in up to 30% of cases) , KRAS and/or NRAS mutations {""'
up to 20% of cases) {845,753,2389}. and other genetic
Clinical features tions pertaining to the MAPK pathway, with single c ases
Tumour-like lesions are characterized by symptoms related to iting ARAF mutations or BRAF, NTRK1 , ALK, or ETV3
tumour location , includin g increased intracranial pressure, focal (753,810). Single cases with CSF1R mutations or RET f ... ~
neurological deficits, and seizures. Neurodegenerative lesions have been reported {810). PIK3CA mutations can occur as s .-
may lead to cerebellar symptoms , pyramidal tract signs, pseu- gle events or in combination with other alterations, mostly !J
dobulbar palsy, or cognitive disturbances (1781] . Infiltration of BRAF p.V600E mutations (in about 11% of all cases) 18451 or
large arteries may induce stroke (892). general , EPHB2 (ERK) activation and a potentially therape-_!.-
cally relevant alteration of a MAPK pathway gene can be . _
Imaging in nearly all cases 1753).
On MRI , tumour lesions are isointense or hypointense on Patients with additional haematopoietic tumours may n
T1-wei g hted images , contrast enhancing, and associated with other mutation profiles. In patients with both Erdhe1m-Cti ~
mass effect and perilesional oedema. Diffuse CNS atrophy disease and Langerhans cell histiocytosis, the freque .
may ap pear over time , even without direct tumour cell infiltra- BRAF p.V600E mutation is even higher (82% of lesmns) ac
tion j754). More than 10% of patients are asymptomatic despite ing to one study {1300}. In patients with a concomitant m';
abnormal CNS MRI findings (2402). neoplasm, the alterations typical for Erdheim-Chester o
can overlap with mutations frequently found in myelo1a tum · ..•
Epidemiology cells (e .g . JAK2 mutations) {2388} .
Neurological com plication s occur in 30 - 50% of patients with
Erdheim-Chester disease, predominantly in men aged about Macroscopic appearance
50 years {2402,238 1) Erdheim- Chester disease manifests as widespread 1n1:1uc
parenchymal lesions (in 44% of cases) , dural thic" en 1~ :o ·
Etiology meningioma-like mass (37%) , or a comb1nat1on ot boll" t '~
Unknown I1781 l. Nerve root or pituitary stalk lesions also o cur 111 ~l

370 rlaemat o lyrnpr1o: d !t,1 i1ot u s 1n volv1n g tr1 e CNS


. .. .~ .
. •.•
f! • •

Rg.10.19 Erdheim-Chester disease. A Sheets of large foamy histiocytes are seen, along with a lymphoid infiltrate, bu t no obvious ernpenpolesis. B Sheets of ep1thelloid
h1stiocytes. C Strong CD163 immunoreactivity. D Extensive factor Xllla positivity.

Histopathology Box10.09 Diagnostic criteria for Erdhe1m-Chester disease


Like in juvenile xanthogranuloma, infiltrates are composed of
Essential:
foamy to epithelioid histiocytes with small nuclei , Touton-type
A population of foamy cells expressing histiocytic antigens
multinucleated giant cells, scant lymphocytes, rare eosinophils ,
and variable fibrosis or gliosis that is often rich in Rosenthal AND
fibres (piloid gliosis). This potential mimicry of pilocytic astrocy- Negativity for Langerhans cell markers (CD1 a and/or CD207 [langerin))
toma is exacerbated by histiocytes resembling astrocytes when
Desirable:
embedded in densely fibrillar CNS parenchyma .
Exclusion of other lines of differentiation (glial, epithelial , melanocyt1c, lymphocyt1c,
etc.)
lmmunophenotype
The neoplastic histiocytes are positive for CD68 , CD16 3, CD14 , Exclusion of reactive and demyelinating lesions

I
and factor Xllla; variably positive for S100; and negative fo r BRAF, MAP2K1, and KRAS or NRAS mutation status
CD1a . About half express BRAF p.V600E {2381) . Potential systemic manifestations on imaging
.
Cytology
Not clinically relevant Staging
Not appli cable
Diagnostic molecular pathology
Testing for potentially mutant genes (BRAF, some in combina- Prognosis and prediction
tion with P/K3CA, MAP2K1 , and KRAS or NRAS) should be Therapeutic options include surge ry, cladrib1ne 1rnmunomodu-
performed with sensitive assays , reflecting the frequ ently low lators, and molecular tmgeted the1 ap1es 1nclud111g BRAF inh1b1-
proportion of neoplastic cells . If none of these 1s detectecl, te st- iors and MEK inhibitors These therup1es yie ld a signi fic ant
ing can progress to the fusion s of NTRK 1, ALK, or ETV3, known tumour re sponse ra te In contrast, neurodegenerall ve lesions
from peripheral Erdheim - Chester di sease . are relatively resistant to tt1erapeut1c irnervent1ons

Essential and desirable diagnostic criteria


See Box 10.09.

Haemato lyrnpl loid tumours involving C11e CNS 371


Pauhis W
Rosai- Dorfman disease Chan .JKr;
ldbr.ith /I.
Per ry A
Sahm F

Definition
Rosai- Oorfman disease of the CNS or the meninges, with or
without systemic lesions , pathologically corresponds to its
counterparts occurring elsewhere . It is a clonal histiocytic pro-
liferation characterized by large 8100-positive histiocytes with
variable emperipolesis .

ICD-0 coding
9749/3 Rosai-Dorfman disease

ICD-11 coding
EK92 Histiocytoses of uncertain malignant potential

Related terminology
Not recommended: sinus histiocytosis with massive lymphad-
enopathy.

Subtype(s) Fig. 10.20 Rosai-Dorfman disease. Dural mass, mimicking a memngioma.


None

Localization loss - are absent in 70% of patients . and 52% have ro assc--
Rosai-Oorfman disease of the CNS forms solitary or multiple ated systemic disease {2582f . On MRI , Rosai-Oorfman o.se~
dural masses, especially in the cerebral convexity, cranial base, resembles mening ioma . Lesions are isointense or hypo te"Sol!
and cavernous sinuses , as well as parasagittal , suprasellar and on T1-weighted images, and they show homogeneous ccrt:e.Sr
petroclival regions {3 194). Parenchymal or intrasellar lesions enhancement. Hypointensity on T2-weighted images may
may also occur. in the differential diagnosis between Rosai-Dorfman disease
and classic meningioma .
Clinical features
Patients may exhibit signs of increased intracranial pressure or Epidemiology
focal neurological deficits. Patients with sellar lesions present The M:F ratio is estimated at 2:1, and the mean age of _
with signs of hypopituitarism and diabetes insipidus. The classic with CNS Rosai-Dorfman disease is approximately 40 yea!'S
systemic signs - cervical lymphadenopathy, fever, and weight {2799).

c
Fig. 10.21 Rosa1- Dorfman disease. This 42-year-old man presented with cervical lymphadenopathy and headaches Head and neck imaging {not shown) demor-su ~~
~e~.si~e enha~·~ing~ erdarged cervical lymph nodes. A This T2-weighted axial MRI shows a lobulated, nearly isointen;e duraJ-based mass alon the anterior falx i.;
;~~1 :,c.en~tr~n.a~ w.11 te m.atte.r is ~everely oedematous . B T1 -weighted, postco_ntrast axial MRI shows that the dural-based bifrontal lobulated ma;s enhances if!lell$&J
1
lht-i l,~S 0110 ~~~; ~~a~%~0:~r.;~'. 1 ~ 1t~:e1 1;1~\:~;:~~. shows another lobulated, intensely enhancing mass along the posterior falx cerebri. Both tentonal leaves are ~ll!n!ll:a.
1

372
Biolagy Box10.10 Diagnostic criteria for Rosai-Dorfman disease
Unknown
Essential:
Pathogenesis A population of large histiocytes with round nuclei, vesicular chromatin, distinct
nucleoli, and abundant pale cytoplasm
Two recent studies found BRAF p.V600E (in 12.5% of cases)
and mutations in KRAS or NRAS (in 25% and 12.5% of cases, AND
respectively) {810,753} . Single cases with ARAF, MAP2K1, Negativity for Langerhans cell markers (CD1 a and/or CD207 [langenn)) and
and CSF1R mutations have been reported (810}. However, the positivity for S100
numbers of Rosai-Dorfman cases were limited in these high-
Desirable:
throughput sequencing analyses of histiocytic disorders, and
Rosai-Dorfman samples were among those types in which no Emperipolesis
alteration could be detected in a large proportion . Exclusion of reactive and demyelinating lesions
Exclusion of other lines of differentiation (glial, epithelial , melanocytic, lymphocytic.
Macroscopic appearance meningothelial, etc.)
Rosai-Dorfman disease of the CNS is typically a firm, vaguely Nuclear cyclln 01 expression
lobulated, yellow to greyish-white dural mass. BRAF, MAP2K1 , and KRAS or NRAS mutation status
Potential systemic manifestations on imaging
Hlstopathology
Rosai-Dorfman disease occurs as a multinodular mass com-
posed of a mixed inflammatory infiltrate including large pale Diagnostic molecular pathology
h1stiocytes, numerous lymphocytes and plasma cells , and Testing for potentially mutant genes (BRAF, ARAF, KRAS and
variable fibrosis. Emperipolesis with histiocytic engulfment of NRAS, and the rare MAP2K1 and CSF1R) should be performed
intact lymphocytes, plasma cells , neutrophils , and occasionally with sensitive assays , reflec ting the frequently low proportion of
eosinophils is typical , but it may be inconspicuous or absent. neoplastic cells .
Notably, emperipolesis is not pathognomonic of Rosai-Dorf-
man disease. occasionally being encountered in other neoplas- Essential and desirable diagnostic criteria
tic or non-neoplastic disorders . See Box 10.10.

lmmunophenotype Staging
The neoplastic histiocytes are positive for CD11c, CD68 , CD163 , Not appl icable
fascin, and S100 ; variably positive for lysozyme; and negative
for CD1a and CD207 (langerin) . Expression of cyclin 01 (pos- Prognosis and prediction
sibly reflecting MAPK activation) can be diagnostically useful For resectab le lesions, surgery is the first therapeutic option .
1195}. particularly because most cases are negative for BRAF For non-resectable lesions, steroids, radiotherapy. and MAPK
p.V600E. signalling pathway inhibitors may be considered . Although little
is known about the long -term natural history of Rosa1-Dorfman
Cytology disease of the CNS . the overall prognosis appears to be favour-
Not applicable able 1n most cases

Haematolympho1d tumour5 1nvo'vrng the CNS 373


f.i;·:julu', II
Juvenile xanthogranuloma (,t1~Jri JVr,,
ldrJa 1h f ..
P8rr; f ;,,
:Jr:Jhrn F-

Definition Related terminology


Juvenile xanthogranuloma of the CNS or th e meninges, with or None
without systemic lesions , pathologically corresponds to its cu ta -
neous counterpart. It is a mostly paediatric , non- Langerhan s Subtype(s)
cell histiocytosis characteri zed by foamy histiocytes , occasional None
Touton giant cells, and inflammation .
Localization
ICD-0 coding Juvenile xanthogranuloma of the er JS localizes •o 'ri:: u::i r
9749/1 Juvenile xanthogranuloma (in 53% of cases) , 1ntradural e/trarnedullar J sp1n8 ( r 13%, '/
nerve roots (in 15%), with meningeal in1ol 1ernent als0 Gi:: r·g
ICD-11 coding common (722) .
2831 .0 Juvenile xan thogranuloma
Clinical features
Tumour lesions induce location-related neurological s; 1 rorc,rr5
whereas the rare neurodegenerative-like lesions induce rr-0 e
diffuse symptoms including cogn1t1ve disturbances .2503'.
On MRI , tumour lesions appear isointense to hypenr erse c,r
T1 -weighted images, with homogeneous contrast enreirse-
ment, and neurodegenerative-like lesions include non-gr arc-
ing hyperintense T2 signals and atrophy {2503,2371)

Epidemiology
CNS juvenile xanthogranuloma typically occurs 1n ch ildren and
young adults {2503 ,722 ,3365) . Neurological involvement 1s
seen in < 5% of patients with cutaneous juvenile xanthogran -
lama.

Etiology
Unknown

Pathogenesis
The frequen cy of BRAF p.V600E mutations is currently unclear
ARAF mutations occur in 18% of cases. KRAS and NRAS mL_a-
tions are also frequent (in as many as 20% of cases) 1810.753].
One study found that occasional cases can have combined
NRAS and ARAFmutations 1753).

Macroscopic appearance
Juvenile xanthogranuloma lesions are often rece ived as frag-
mented, soft, yellow to tan-pink biopsy specimens

Histopathology
Fig. 10.23 Juvenile xanthogranuloma. A 7-year-old boy with proptosis. A T2-weight- Juvenile xanthogranuloma (overlapping w1rh Erdhe1m-Cnesrer
ed axial MRI shows proptosis; both orbits are infiltrated with very hypointense soft disease) is composed of rounded to spindled . variably vacuo -
tissue masses. There is an extensive, lobulated, hypo1ntense mass involving the ten- lated histiocytes , scattered Touton and foreign body-type giant
torium and straight sinus. Moderate obstructive hydrocephalus is present and there is cells, lymphocytes , and occasional eosinopl11ls \25031
a lesion in the choroid plexus ot the left temporal horn . B T2-we1ghted coronal MRI
shows symmetrical hypointense masses along the tentorium. The glomus In each cho-
lmmunophenotype
roid plexus is enlarged and hypointense. C T1-we1ghted, contrast-enhanced axial MRI
with fat saturation . The orbital masses enhance intensely, as does the mass along the The neoplastic hist1ocytes of iuvenile ;.,,anthog•a·1 Jfoma are
straight sinus and tentorium. D T1 -we1ghted postcontrast MRI shows that the dural- CD1a-negative. C011c-posit1ve , CD14-pos,'.1ve. CD6.3-pos1t1ve
based lobulated masses enhance intensely and quite uniformly, as do the masses factor Xllla-pos1t1ve , lysozyme-negat1ve an 1 S ~ 1 c:gat1ve
-'

in the choroid plexuses. There 1s a smaller duraJ-based mass along the falx cerebn . BRAF p V600E protein 1s present 1n mut<:l'1' cw,,, -) 131
Cytology
Not relevant

Diagnostic molecular pathology


Testing for potentially mutant genes (BRAF, ARAF, KRAS, and
IRA ) should be performed with sensitive assays , reflecting
frequently low proportion of neoplastic cells . Mutations of
F1R and fusions involving an NTRK gene have been reported
r peripheral juvenile xanthogranuloma , so their assessment
ay be particularly useful in cases not limited to the CNS . For
TRK fusions, RNA sequencing or in situ hybridization can be
applied , depending on the tumour cell density.

Essential and desirable diagnostic criteria


See Box 10.11 .

Staging Box 10.11 Diagnostic criteria for juvenile xanthogranuloma


Not applicable
Essential: · f'lt t
. d f foamy cells and a mixed inflammatory 1n 1 ra e
A histiocyt1c tumour compose o
Prognosis and prediction
Therapy relies on maximally safe surgery when feasible . Cyto- Desirable: . .
toxic chemotherapies, radiotherapy, and molecular targeted Exclusion of other lines of differentiation (glial , epithelial, melanocyt1c, lymphocyt1c,
therapies including MAPK signalling pathway inhibitors are Langerhans cell , meningothelial , etc.)
therapeutic alternatives based on molecular characterization BRAF, ARAF, KRAS, and NRAS mutation status
{2503}. Drawing prognostic conclusions abou~ juv~nile xan-
Evidence of cutaneous manifestations
hogranuloma of the CNS is difficult because of its rarity.

Haematolympho1d tumours involvmg the CNS 375


P;:wlu s W
Langerhans cell histiocytosis 01rin .JKC
lrltv-11~1 /\
()<JJfJrrl f\()
f'F!rry /\
~ )Ahrrt F

Definition
Langerhans .ell h1stiocyto~is pf th . CN S ot thf) mAninqes 1s a
clonal proliferation of Lang rhans typ ells manifesting in the
CNS or th meninges, with 01 without sys l mic lesions, which
pathological! corresponds to its counterparts occurring else -
where

ICD-0 coding
9751/1 Langerhans cell histiocytosis

ICD-11 coding
2831.2Y & XH1J18 Other specified Langerhans cell histiocyto-
sis & Langerhans cell histiocytosis, NOS

Related terminology
Not recommended: histiocytosis X; eosinophilic granuloma;
Hand-Schuller-Christian disease; Letterer-Siwe disease; fig. 10.25 Langerhans cell h1stiocytos1s. X-ray showing bone uc8ncv at s•;e )f
1
J,_
Langerhans cell granulomatosis . ease.

Subtype(s)
None

Localization
The most common CNS involvement is via lesions of the crani-
ofacial bone and skull base (seen in 56% of cases) , with or
without soft tissue extension . lntracranial, extra-axial masses
are also common , particularly in the hypothalamic- pituitary
region (in 25-50% of patients), meninges (30%), and choroid
plexus (6%). A leukoencephalopathy-like pattern, with or with-
out dentate nucleus or basal ganglia neurodegeneration, is
seen in 36% of patients with Langerhans cell histiocytosis, and
cerebral atrophy occurs in 8%. Rare intraparenchymal CNS
masses have also been described (1291,1767,2557}. Langer- Fig. 10.26 Langerhans cell histiocytosls in a 46-year·old man with multiple cranial
hans cell sarcoma primarily occurring in the CNS has not been nerve palsies. A T1 -weighted postcontrast axial MRI shows a thickened . enhancing
reported. infundibulum with multiple patchy enhancing lesions 1n the pons and both temporal
lobes. B More cephalad T1-weighted postcontrast axial MRI shows a thickened. an -
hancing infundibulum with multiple patchy enhancing lesions in the pons and ooth
Clinical features temporal lobes.
Patients with circumscribed tumour lesions experience acute or
subacute, nonspecific and/or location-dependent neurological medullare and symmetrical T1 hyperintensity of the dentate
symptoms. Patients with neurodegenerative-like lesions present nuclei . Similar lesions are observed in supratentorial areas with
with a chronic and slowly progressing neurological pattern nonspecific white matter T2 hyperintensity and symmetrical T:
comb ining cerebellar syndrome, pyramidal tract signs, pseu- hyperintensity of the basal ganglia. Over time, diffuse CNS atro-
dobulbar palsy, and/or neuropsychiatric symptoms j1291}. phy may appear.
On MRI , tumour-like lesions are characterized by one or
multiple masses that appear hypointense on T1-weighted Epidemiology
images and contrast-enhanced after gadolinium infusion . On Most cases of Langerhans cell hist1ocytos1s occur 1n childhooa ,
T2-weighted images, lesions and perilesional oedema are with an annual incidence of 0.5 cases per 100 000 1ndiv1duals
hyperintense. Neurodegenerative lesions do not exert mass aged< 15 years and with an M :F ratio estimated ar 1 2 /12031
effect, are not contrast enhancing, and are not surrounded
by perilesional oedema . They are mainly located in the pos- Etiology
terior fossa with symmetrical T2 hyperintensity of the corpus Unknown

376 Ha.1:rnr.t~rJ1yrnnr101ci tumou rs involv1no the C f\J S


Rg.10.27 Langerhans cell histiocytosis. A In this lesion involving the skull bone and dura, Langerhans cells have typically oval, deeply grooved to contorted nuclei, delicate
nuclear membranes, fine chromatin, and ample lightly eosinophilic cytoplasm. There are admixed eosinophils. B CD1a expression by neoplastic Langerhans cells. C BRAF
p.VWOE expression in neoplastic Langerhans cells. D Langerhans cells are immunoreactive for CD207 (langerin).

Pathogenesis found in some {1168 ,2047] . Eosinophils may aggregate and


The most frequent molecular alteration is BRAF p.V600E muta- undergo necrosis , producing granulomas or abscesses
tion, which occurs in about 50% of cases (169,271 1,810). Single
cases of BRAF p.V6000 or ARAF mutations, or BRAF fusions, lmmunophenotype
have been reported {1550 ,2231,810 ). Of the BRAF-wil dtype Neoplastic Langerhans cells consistently express CD1a (sur-
cases, MAP2K1 mutations are found in about 25% (753,386) . face), CD207 (also known as langerin: granular cytoplasmic) ,
NRAS. KRAS, and PIK3CA mutations have been reported in S100 (nuclear and cytoplasmic) . and CD68 (Golgi dot-like stain-
single cases (1292,169,2711 ,810}. ing, sometimes weak) ; about 50-60% express BRAF p V600E

Macroscopic appearance
lntracranial Langerhan s cell histiocytosis lesions are of ten yel-
low or white and ran ge from discrete dural -based nodules to
granular parenchymal infiltrates . CNS lesions may be well delin-
eated or poorly defined .

Histopathology
lnftltrates include neoplastic Langerhans cells and vari-
able reactive macrophages, lymphocytes . plasma cel ls, and
eosinophils. The nuclei of Langerhans cells are typically
slightly eccentric , ovoid, and reniform or convoluted, with lin-
ear grooves and inconspicuous nu cleoli Tr1Pre is abundant
pale to eosinophilic cytoplasm, and Tou ton g1af"f t cells may be
&&en Copious col lagen depo si tion 1s c u1rr1•·11 In th13 neuro-
at:generat1 ve lesions of the cerebellum t)ff":1 11 :.lcrn ...:rnd b35al
ganglia, there are often no obv1ou ~: Lei.• 1•;il1rf1' f: lls , but
inflammation accom panie s severe r1 ou1 1f ·. d 1· •• I ~p.ri r .<:11 kiss
11111-i perivascula r BRAF p.V600E - po<,1". '· , 1'· " rl fl 101 qicy lll Flg.10.28 Langerhans cell h1stJ·t\l'\J1
. v ... ,
E · ·
osis. tectron mt.croscopy showing charactensttc
Phenotype (CD14+ CD33+ C01C 3 t ! '·: 1 :rL1 t•ll1PIL- 's
1 rod·shaped structures (B1rbeck granules).

t i (J1-~ rriulot rnplloi J turr o ·rs uw in_ me CNS 377


Box 10.12 D1agnost1c cr11enA for Langerh(lns ce ll histlocytosls 1rnrnunot11stochf)m1stry lndep8ndent ()f tr1e:i r,f Jl';(,if ir, ;:Jllc;r;jf,r,r
nct1vat1ori of the MAPK p8thway urn b8 1nfArr8rJ frrirr irr rr J" r,
Etnntlal:
histochemistry for p~1o sphoryl;:i tf.~rJ FRK
A population of cells showing cytological features of Langerhans cells and
expressing C01a andfor CD207 (langerln)
Essential and desirable diagnostic criteria
Deslr1ble: See Box 10 12.
s100 posihvlty
BRAFand MAP2K1 mutation status
Staging
Not applicable

p 156.27781. The Ki-67 proliferation ind ex is highly variable, but Prognosis and prediction
it can reacll 50% Tumour-like lesions are sens1t1ve to conventional ant1-turrei 1Jr
treatm ents including vinblastine or cladrib1ne and. 1n cases N1rr
Cytology an actionable target , to molecular targeted therapies (1 e ~11/iPV
Not relevant signalling pathway inhibitors). allowing a high tumour respors8
rate and a favourable prognosis 12241,27071 In contrast, neur0-
Diagnostic molecular pathology degenerative lesions are resistant or poorly sensitive to mult1oli:;
Testing tor potentially mutant genes (BRAF. MAP2K1 , ARAF. therapeutic strategies, including radiotherapy. d1ffereri·1at1rg
NRAS. KRAS, PIK3CA) should be performed with sensi- agents, immunosuppress1ve drugs , cytotoxic chemothemp res
tive assays, reflecting the frequently low proportion of and molecul ar targeted drugs . Neurological symptoms terd ·o
neoplastic cells . BRAF p.V600E can also be detected via worsen slowly over decades [ 1407] .
Hi tiocytic sarcoma Pau lu s W
Chan JKC
ldba1f-i A
Perry A
Sahm F

Definition Localization
,.; . ~t1ocyt1c sarcoma is a malignant proliferation of cells showing Hist1ocytic sarcoma can involve any site of the CNS and the
...,0 rph ological
and immunophenotypic features of tissue histio- meninges .
~; tes and exh ibiting no other lines of differentiation .
Clinical features
tCD-0 coding Neurological symptoms are related to tumour location Neuro-
9-55:3 Histiocytic sarcoma imag1ng mimics a malignant primary CNS tumour

ICD-11 coding Epidemiology


_8311 Histiocytic sarcoma Fewer than 100 cases have been reported . most of them 1n
adults l2045j .
Related terminology
one Etiology
Isolated cases of radiation-associated histiocyt1c sarcoma of
Subtype(s) the CNS have been reported (505.3484] .
•Jone
Pathogenesis
A stu dy of 28 histiocytic sarcomas found RAS/MAPK pathway
gene alte rations in 16 cases (57% , affecting mostly MAP2K1 .

.... -- . . ..
Flg.10.29 Histiocyt1c sarcoma of brain. A This example of t11 t.! 1u1. y11. :::,1•v1rr r. '~" . ;;'11~, i 11.:•o by n p10111111en1. lf1f1:11...it~ of neuuuphtls with areds ut ;;u~1~ q
turnour cells have oval or reniform nuclei and abundant ec;:;11 1J1)·1\if ' , ·.i .1! .. ,, t: •'u-11. ,~- .'lli11 u ,, s1 :i. 111rv fn1 ~, 100 111 d µroport10 11 ot tumour cells 13 ·u11 11 r ,,,1 0 ,
specific marker CD163 was diagnostic in this high -grade 11eq;u :o 11 1
Box 10.13 Diagnostic criteria for histiocytic sarcoma antigens , CD30. ALK , and other lymphoid markers, as well as
Essenttal: for glial , epithelial , and melanocytic antigens The tumour cells
A population of malignant cells expressing histlocytic antigens are negative for the follicular dendntic cell antigens CD23 and
AND CD35 . However, follicular dendritic cell sarcoma expre-:;s1ng
these antigens may primarily arise in the brain and must be
Exclusion of other lines of differentiation (glial, menlngothelial, epithelial,
melanocytic, lyrnphocytic, muscular, vascular, etc.) differentiated from histiocytic sarcoma 11257). Although MAPK
alterations are common, BRAF p.V600E expression is rare
11630,2892).

KRAS, and BRAF, but also NRAS, PTPN11, NF1, and CBL) and Cytology
Pl3K/AKT/mTOR pathway gene alterations in 6 cases (21%; in Not relevant
PTEN, MTOR, PIK3R1, and PIK3CA) {2892) . Alterations in both
pathways were not mutually exclusive. In addition , CDKN2A Diagnostic molecular pathology
and/or COKN28 was altered in 13 cases (46%), half of which Although the proportion of BRAF p.V600E-mutant cases
displayed homozygous deletions . Independently, a recent study is relatively small, a variety of other MAPK gene alterations
reported single cases with KRAS, NRAS, or CSF1R mutations , have been reported in non-CNS histiocytic sarcoma. so high-
or BRAF fusion (810) . A single case is described with concur- throughput sequencing may be advisable. Sequencing should
rent BRAF p.F595L and HRAS mutation {1707}. cover all the potentially affected genes of the MAPK pathway
(MAP2K1 , KRAS, NRAS, PTPN11 , NF1, and CBL) , the reported
Macroscopic appearance altered genes of the mTOR pathway (PTEN, MTOR, PIK3R1.
Histiocytic sarcomas are destructive, soft, fleshy, white masses and PIK3CA) , and CSF1R mutations. Rare BRAFfusions can be
with occasional yellow necrotic foci . detected via in situ hybridization or RNA sequencing. CDKN2A
and/or CDKN28 alterations often constitute homozygous dele-
Histopathology tions, which can be detected by in situ hybridization or array-
Histiocytic sarcoma is characterized by highly cellular, non- based approaches.
cohesive infiltrates of large, moderately pleomorphic , mitotically
active histiocytes , which have abundant eosinophilic cyto- Essential and desirable diagnostic criteria
plasm . variably indented to irregular nuclei, and often prominent See Box 10.13.
nucleoli . Occasional multinucleated or spindled forms are also
common, as is background reactive inflammation {534,558}. Staging
Not appli cable
lmmunophenotype
The tumour cells are typically positive for histiocytic markers Prognosis and prediction
(e.g. CD68 , CD163 , lysozyme , CD11c, and CD14); variably Survival time has been < 12 months after initial presentation in
positive for CD34; and negative for myeloid antigens, dendritic most reported patients.

r Ir j • I I I : ' I ,' I Ii "' I ~'


Germ cell tumours of the CNS Rosenblum MK
lch1rnura K
Lau CC
N1sh11<awa R
Pietscr1 T
Wong TT

Definition
Germ cell tumours of the CNS are a fam ily of morphological and
immunophenotypic homologues of gonadal and other extra-
neuraxial germ cell neoplasms sharing certain genetic features
(see Table 11 .01 for definitions of individual types) .

ICD-0 coding
9080/0 Mature teratoma
9080/3 Immature teratoma
9084/3 Teratoma with somatic-type malignancy
9064/3 Germinoma
9070/3 Embryonal carcinoma
9071/3 Yolk sac tumour
9100/3 Choriocarcinoma
9085/3 Mixed germ cell tumour
flg.11.01 Mature teratoma . Tl -weighted. contrast -en hanced sag1ttal MR! n a}- /'=~­
old boy shows a suprasellar tumour with strong contrast enhancemer \ ar c 'ee'h ~, ~
ICD-11 coding
posterior and inferior parts.
2A00 .1Y & XH1E13 Other specified embryonal tumours of brain
& Germinoma
2A00 .1Y & XH8 MB9 Other specified embryonal tumours of
brain & Embryonal carci noma, NOS
2A00 .1Y & XH09W7 Other spec ified embryonal tumours of
brain & Yolk sac tumour
2A00 .1Y & XH83G5 Other specifi ed embryonal tumours of brain
& Teratoma, NOS
2A00.1Y & XH8PK7 Other specified embryonal tumours of brain
& Choriocarcinoma, NOS
2A00 .1Y & XH2PS1 Other specified embryonal tumours of brain
& Mixed germ cell tumour

Related terminology Fig.11.02 Germinoma. Sagittal MRI of a 21-year-old man who developed headac."'a,
Teratoma with somatic-type malignancy nausea, and Parinaud syndrome. A T1-weighted image shows a contrast-en
Not recommended: malignant teratoma; teratoma with secondary tumour of the pineal reg ion. B T2-weighted image.
malignant component; teratoma with malignant transformation .
pituitary I infund ibular stalk) 13 10 8,3111 f. Bifocal and mult1foca!
Germinoma examples (nearly al l germinomas) typically involve these s1es.
Not recommended: sem inoma (used for germinoma in the tes- but they may also occ ur in the basal ganglia . thalami , or otner
tis); dysgerminoma (used for germinoma in the ovary). locations 13108}. A basal ganglionic location has been more fre-
quently reported in patients in eastern Asia than in those 1ri ti"e
Yolk sac tumour USA , with the reverse being recorded for a bifocal (posreri or
Not recommended: endoderm al sinus tumour. pituitary and pineal) presentation 13111 }. Germinornas can also
grow as diffu se periventricular lesions, and they can arise in tne
Choriocarcinoma cerebrum, cerebellum . posterior fossa structures. spinal c d
Not recommended: chorionepithelioma. and sella . Congenital holocranial examples (typically teratomasl
are encountered.
Subtype(s)
r'lone Clinical features
Pineal region tumours compress the ce rebral aqueduct, caus-
Localization
ing hydrocephalus and intracranial hypertension. and they '.ar
r"pp rox1mately 80 - 90% of CNS germ cell tumours arise in the produce paralysis of upward gaze and convergence tPar1nauu
1 1
r•· 1 J 111e, most frequently in the pineal region followed by the
syndrome) by compressing or invading the tecral plate Pre-
' Jt ,r asellar compartment (where they originate in the posterior
served pupillary accommodation with impaired constr C! IL 11
Table 11.01 Definitions and dia_g~ostic criteria for germ cell tumours of the CNS
'fypl Definition
Essentlal diagnostic criteria
A. germ cell tumour composed solely of fully • A germ cell tumour with components exhibiting d1fferentration along at least two of he
Mature teratoma d1fferent1ated, adult-type somatic tissue components three somatic tissue lines (ectoderm endoderm. mesoderm )
that recapitulate the differentiating potential of the Fully differentiated. adulHype histology (absence of fetal-type elements)
ectoderm, endoderm, and mesoderm • Absence of other germ cell tumour componen ts
A germ cell tumour containing incompletely • Identification of incompletely differentiated elements exhibiting differe11fiatlon along
differentia~ed, fetal-like somatic tissue components at least two of the three somatic tissue lines (ectoderm, endoderm , mes~rrn) rn a
Immature teratoma that recaprtulate the differentiating potential of teratoma, or the identification of any such elements within a tumour otherwise qualifying
the ectoderm, endoderm, and mesoderm; mature as a mature teratoma
elements may be admixed • Absence of other germ cell tumour components
A germ cell tumour of mature or immature
Teratoma with teratomatous type that develops a distinct secondary • ldentrflcation of a distinct histological component that has the cytological features.
somatic-type component resembling a somatic-type malignant architecture, mitotic activity, and disorderly growth pattern expected of a sarcoma.
maJlgnancy neoplasm (e.g. sarcoma or carcinoma) as seen in carcinoma, or other defined type of somatic cancer in a mature or immature teratoma
other organs and tissues

• A germ cell tumour containing large tumour cells with typical c~ologicaJ charactenstlcs
Nuclear OCT4 and widespread membranous KIT (or podoplamn (02-40])
immunoreactivity, or absence of 5-methylcytosine expression
A malignant germ cell tumour composed of cells • Absence of CD30 expression
Germlnoma • Absence of AFP expression . .
resembling primordial germ cells
hCG immunoreactivity in syncytiotrophoblastic giant cells (for the specific dragnosrs of
germinoma with syncytiotrophoblastic elements)
• Absence of other germ cell tumour components (except syncytiotrophoblastic giant cells
for the specific diagnosis of germinoma with syncytiotrophoblastic giant cells)
• A germ cell tumour with large epithelioid cells as described in the Histopathology
subsection
A malignant germ cell tumour composed of large • CD30 and OCT 4 expression
Embryonal • Absent or only focal, non-membranous KIT expression
epithelioid cells resembling those of the embryonic
carcinoma • Absence of hCG expression
germ disc
• Absence of AFP expression
• Absence of other germ cell tumour components
• Cytokeratin expression is desirable
• A germ cell tumour with epithelioid cells arranged in any of the patterns described 1n the
Histopathology subsection, with or without mesenchymal components
A malignant germ cell tumour that differentiates to • Absence of other germ cell tumour components
YQlk sac tumour resemble extraembryonic structures, including the • AFP expression
yolk sac, allantois, and extraembryonic mesenchyme • Absent or only focal, non-membranous KIT expression
• Absent or only focal CD30 expression
• Absence of P-hCG expression
• A germ cell tumour with both syncytiotrophoblaslic and cytotrophoblastic elements bur no
A malignant germ cell tumour that other germ cell tumour components
differentiates to resemble the trophoblastic • P-hCG expression
Chorlocarcinoma
cells of the extraembryonic chorion, including • Absence of KIT (or podoplanin [02-40]) expression
syncytiotrophoblastic and cytotrophoblastic elements • Absence of AFP expression
• Absence of OCT 4 expression
Mlxedgerm Malignant germ cell tumours harbouring at least two
• A germ cell tumour with at least two distinct germ cell tumour subtypes
cell tumours germ cell tumour subtypes in any combination

(Argyll Robertson pupil) is also frequent . Posterior p ituitary I of fat suggest teratom a, wh e reas haemorrhag e 1s common ly
suprasellar lesions produce visual disturbances by 1mp1nging assoc iate d with c hor1 ocarc1nomato us e lem ents.
on the optic chiasm , and they often c ause diabetes insipidu s,
delayed growth , and delayed sexual matu ration by d isrupting Epidemiology
the hypothalamic- pituitary axis . Ge rm ce ll tumour s prod uci ng CNS germ ce ll tumours principal ly affect child ren. and they are
hCG c an cause precocious pubert y in boys and (rarely) 1n girls more p revale nt 111 eastern Asia than m Europe and the USA Age -
12325 ,3020) adJusted annual 1nc1dence rates of O 45 cases per 100 000 pop-
ulot1011agoli < 15 ye::irs <JncJ O 49 cases per 100 OUO population
Imaging aqf d.: 19 Ytl<JrS h..J .. e bt.Of1 r~portecl trom Japan I 19891 c1nd [i'lt'
Teratomas excepted , germ ce ll tumours are usu...1llv sol1ci anu H,_,i,._iubi1L' · f ~ ,1r1:'"1 11 '-,-IGI r1-·spel:t1vely. triese rates (tf1e t11~1' 1t>St
contrast enhancing on CT/ MR I 1995 ,18841 lntr atun1 our«11 1 , ' r ~ '~' 1.•Ll• ·11• r 111 JrC· 11 1. 111 rr1pl<J those 111 G 1:1rr1 1a11y 11::i~>1JI dlh.1
calcification , and regions of low signal attenuat1c111 1_.l1c1r,1Cr·' .,' .' I ,,'I. ! ll :i ') ~.~'II. ,:,:II !1 C1hJUIS c1CC0Ullt tur :i i•'_. L'r

1 , I
• .t '
'·'' I'
Flg.11.03 Teratoma. Large immature teratoma of the cerebellum in a 4-week-old in- Fig. 11.04 Germinoma. Suprasellar tumour from a 7-year-old girl.
fant, with characteristic cysts and chondroid nodules.

all primary intracran ial neoplasms and for 8- 15% of paediat- reported to encompass KRAS, CCND2, and PROM14 (tti.s ra.s-
ric examp les in series from Japan 1350}; Taiwan , China /1321} ; gene being a regu lator of primordial germ cell spec1ficat1GriJ
and th e Republ ic of Korea 11546). In contrast, these tumours and losses of the RB1 locus could implicate the cyclin/COI<',
account for only 0 .3-0.6% of primary intracranial tumours in RB1/E2F pathway (3166 ,2861 ).
adults , and 3-4% of those affecting children , in series from As regards genetic drivers , gain-of-function mutatJons
India 11527}, Europe /656}, England {715), and North America involving genes encodin g MAPK pathway components (KrT
{1326 ,290 ). Incidence peaks in patients aged 10- 14 years , and RAS fam ily members) and (less freq uently) P13K/A KT/mTOP
a clear majority of all histolog ical types involve male patients pathway components (MTOR, PTEN, and PIK3 family merr-
{290 ,1321 ,2817,1989,1546}. More t han 90% of pineal examples bers) are the most com mon gene abnormalities ident1f1ed 1n
affect male patients , but there is no sex pred ilection in poste- CNS germ cell tumours occu rring beyond infancy and eart1
ri or pitu itary I suprasellar cases /2042 ,3110). Cases involving childhood 11005,3374 ,1397,286 1}. These occur across all sub-
members of the same famil y are rare {1160). Pure germinomas types , but they are variably rep resented . Germ1nomas exn1bi
outnumber oth er ty pes , with mixed lesions and teratomas being a particularly high freque ncy of MAPK pathway-activating Kl
next most com mon; embryonal carcinomas , yolk sac tumours , (exons 11 , 13, and 17) and RAS gene fami ly mutations (found
and choriocarc inomas occur uncommonly in pure form {1321 , in -60% of cases) as well as frequent coactivation of the RAS1
290 ,2042,3111 ). ERK and AKT pathways , wi th associated severe chromosornat
instability and global DNA hypo methylation sig natures simi-
Etiology lar to those of primord ial germ cells (and generally foreign to
Germline variants of the JMJD1C gene, wh ich encodes a other CNS germ cel l tumou rs) {1005 ,2861 ,1006}. KIT alte a-
jumonji domain-containing histone demethylase, have been tions have predom inated in germinomas derived from Japa-
associ ated with a hei ghtened risk of CNS germ cell tumours in nese patients /1005,1397,1 00 6), b ut they were outnumbered
Japanese people {1762). An increased risk of CNS germ cell by RAS lesions in a co hort reported from Germany (28611.
tumours in the settin g of Klinefelter syndrome (47,XXY) /1526 , Copy-number gains and amplifications of KIT and RAS genes
3450 ,23621 implicates X chromosome-associated genes that have been documented , as have loss-of-function mutat10ns m
presumab ly escape normal inactivation. An excess of CNS the tumour-suppressin g BCORL 1 and CBL genes (the latter a
germ cell tumours appears to occur in Down syndrome (tri- negative regulator of MAPK signall ing) {3374,139 7}. Spec · 1
somy 21) 12133}. microRNAs (e.g . mi R-302, miR-335 , miR -371-3 , and m1R-
Genetic observations arg ue against a shared eti ology (o r 654- 3p) appear to be upregulated in CNS germ cell tumours
pathogenesis) for all CNS germ cell tumou rs. Whereas intra- 13371,2377}.
cran1al teratomas of infancy re semble teratomas of the infant
testis in their generally diploid statu s and chromosomal integrity Pathogenesis
l26661. CNS germ ce ll tumours arisin g after early chi ldhood , CNS germ cell tumours arising beyond intancy and early ch1la-
irrespective of histological composition , share with their testicu- hood may share a unifying pathogenesis and cytog nes.s
lar counterparts in young men frequent aneuploidy with complex despite their morphological variety and epigenetic differences.
cnrornosomal anomalies and overlappin g copy-number altera- This is suggested by overlapping genetic alterations leading tc
tions (2851,3166,1005,3374,2861) . Particularly common are the activation of the MAPK and/or AKT/mTOR pathways (alb t
~Ja1ns on chromosomes 12p, 21q, Sq , 1q, 2p, and X, and losses with strikingly different mutation frequen c ies of KIT and other
ot 11q, 13q, 5q, 9q, and 10q The weight of evidence, however, individual genes) (1397,28611 and by the detection ot 1d l
1ndicc.ites that fewer CNS tumou rs exhibit isochromosome 12p, M TOR mutations in both the globally hypomethylated germino -
<t signature abnormality of testicu lar (and mediastinal) primaries
matous component and the highly methylated non-garm1r ~
12316,3068 ,3166,1005) Regions of pa rti cular gain have been
rn atous component of mixed lesions 110061. The similarity
38 4 (,\-Hiii L(• \i l 'J llllJUI:...
"NS and gonada l germ cell tumours in appearance an d immu- Immature teratomas may contain cysts, regions of calcif1ca-
0pheno~ ype, as well as in thei r chromosomal, genetic, and tion , an d c hondroid nodules , but they generally
.
. f·
have soft. fleshy
t I t
epigenetic la nds~ apes , accord s with the view that CNS germ components reflecting the high cellular.1ty o 1mma ure e emen s.
cell tumours . deri ve from primordial germ cell s that migrate to Teratomas with somatic-type malignancy may resemble
the ~NS d.unng development. Germinomas express a panoply mature or immature teratomas but are more likely to exhibit
of pnmord 1a.1germ cell- associated antigens {1324} and closely regional necrosis . Overgrowth by sarcomatous .components
res~m~le migratory primordial germ cell s at the E10.5 stage in may impart a fleshy appearance, whereas muco1d/gelat1nous
their wi despread DNA hypomethylation and in the differentially regions may reflect the presence of mucin-produc1ng adeno-
methylat~d statu.s of their imprinted genes (1006). Mutations of carcinoma . .
· IT l~ading to ligand-independent activation could save pri- Germinomas are usually solid , tan-wh ite, soft. and friable .
mordial germ ce!ls in the CNS from the apoptotic death th at Focal cystic change can occur, but haemorrhage and necrosis
follows physiological suppression of KIT signall ing in primordial are rare.
germ cells that fail to migrate properly from the midl ine (275 0}. Embryonal carcinomas are solid, grey-white , fn~ble masses
Su~h cells cou ld then spawn pure germinomas or, vi a epige- that may exhibit regional haemorrhage and n~cros1s . .
~etic repr?gran:ming , other germ cell tumour types. The abil- Yolk sac tumours are solid , grey-tan , and friable or gelatinous
ity of munne primordial germ cells to dedif'ferentiate into pluri- (due to myxoid alterations) . Focal haemorrhage may be seen .
potent stem cells and to generate teratomas in vivo has been Choriocarcinomas are typically solid, haemorrhagic , and
documented (2786}, but primordial germ cells have never been often extensively necrotic masses containing foci of grey-tan
Identified in the human CNS (901}, and alternative theories ot tumo ur tissue.
cytogenesis implicate other pluripotent ancestors (2327} . These The appearances of mixed germ cell tumours reflect the mac-
include embryonic stem cells and neural stem cells , which can roscopic features of the constituent germ cell tumour compo-
generate teratomas after activation of the POU5F1 (OCT4) pluri- nents, as described for these tumours in pure form .
polenc y gene {3122}. although why these stem cells would be
reprogrammed to the germ cell differentiation pathway remains Histopathology
unexp lained . The genetically distinct infantile CNS teratomas Mature teratoma
could derive, alternatively, from non-germinal stem cell ele- Mature teratomas harbour only fully differentiated, adult-type tis-
ments. Whether the characteristically pure and mature terato- sue elem ents that exhibit little if any mitotic activity. Ectodermal
mas of the spinal cord represent germ cell neoplasms {85} or components commonly include epidermis and skin adnexa,
complex malformations {1683) is debated. central nervous tissue, choroid plexus, and salivary gland acini .
Subsequent genetic events occurring with in the differenti- Smooth and striated muscle, cartilage , bone, and adipose tis-
ated tissue components of teratomas are presumed to drive the sue are typical mesodermal representatives . Glands that are
development of somatic cancers in these lesions; one enteric- lined by respiratory or enteric-type epithelia and are often cysti-
type adenocarcinoma arising from a mature teratoma exhibiting cally dilated are the usual endodermal participants , but hepatic
an ac quired KRAS mutation has been reported (1619). and pancreatic tissue may be encountered . Gut- and bronchus-
like structures re plete with mucosa and muscular coats or with
Macroscopic appearance carti laginous rings, respectively, can be formed. Exceptional
Mature teratomas typically have both sol id components an d intracranial teratomas contain remarkably organized , fetus-like
cysts of varying diameter that may contain mucinous material. bodies {22 18).
Areas of calc ification and chondroid nodules may be appreci - Re-resection specimens deriving from germ cell tumours
able. Haemorrhage and necrosis are typically absent. displaying progressive enlargement in the course of adjuvant

Ag. 11 .05 Mature teratoma. A Mature teratoma with glantJula i el .;mt-:'11~ ancl l1 carl1lag1nuu;:; nod le (lower rig ht)
amucosa that 1s partly of gastric type, witn muscularis mucos3 and sl:bnn .. uSil gl<rnc.Js
favnurNJ by CN ~) w~rrn (~8 11 t11rr'lfJIJr') 1 :~?'J?j Fr l'""".1':t; )¥,.. ~-.
( 1?8[>1 ;:rncJ lrnnmyosmr,(Jm;i l?'Jfi 1j h;:::)VP. bA'.m "Ir!· r.r ~.~rj ,..
sm:ondmy m;::iliqnarit r:nmr.;riri~nt'i, r!', h.=tt, ,) r:.;:irr,or 'l•1 : ·" r,"
assoc1atArl with" n 1ntrnrJur ~JI sp1r1'll ter::str:irr ~ 1111 F-, , 1 r , fA .-,,
geriP.SiS of n compoc;itA 1nt r.8Sellar turrtrJ1 Jr r,ry I~ r r1 rj e.r- '?" ·~
of Burkitt-lrke 8 -cell lymphoma '3nrl qArrr 1 n r, ,..,..,~ .... Jrr ~=­
(32621. Yolk sac tumour components r1rJv~ G~'1n th~ '".Po/. ,i~J!1
progenitors of Anterir:-type adAnf'J<:...3rorir)rri~r; tl'lr.r,.Jr k~
in se lected cases /9821 Cytologrr,al atyp1a ~lr;n tr1P." 11 ,,,,,
pronounced. should not prompt the d1agnr.tC>tS nf sc".>rr;~ •<>' '~­
malignant transformation , this being a featur~ 0f som~ r)r~ r
mature teratomas (especially after adjuvant therao1>

Germinoma
Germinomas are composed of large. undifferent•.::Jt~ -·'Y},."rr1
cells that have round, vesicular. and centrally po131t10red -, Y.:~
with prominent nucleoli , and abundant cytoplasm trat •s o!'i~
Fig. 11 .06 Immature teratoma. Undifferentiated neural component with neural tube- clear due to glycogen accumulation An example 9J<h1 • q
like formations in an immature teratoma. rhabdoid features has been described 131061 Tumour c 3
are disposed in sheets, lobules. or (in cases manites r'lg IJ'35-
therapy or recurrence after initially complete response to moplasia) regimented cords and trabeculae Mt otlc act , 1
treatment may be c omposed solely of mature teratomatous is variable, necrosis uncommon . Delicate fibrovascu1ar seo"a
elements , a seemingly paradoxical scenario termed "growing infiltrated by small lymphocytes are typical. with tumour r :
teratoma syndrome" (2355 ,3236) . Although the simple expan - occ asio nally being obscured by florid lymphoptasmacytic 3r-o
sion of cystic com ponents can play a role in this phenomenon , histiocytic infiltrates 13579) or an intense granulomatous reac-
Ki-67 imm unohistochemistry may reveal surprisingly elevated tion m im ickin g sarcoidosis or tuberculosis (1698}
labelling activity w ith in their ostensibly differentiated tissues Consistent c ell m em b rane and Golgi region immunoreact .,
{2355). for KIT and membranous labelling for 02-40 distinguish germ-
nomas from solid variants of yolk sac tumour. embryonal carc.-
Immature teratoma nomas , and other g erm cell neoplasms (1399.1034) . Comple!S
The id entification of even minor tissue components having loss of nuclear 5- methylcytosine immunoreactiv1ty by turno.,·
incompletely d ifferentiated , fetal-like appearances mandates cells is also uni q ue to germinomas , reflecting a global ONA
the class ification of a teratoma as immature. Commonly repre- hypomethylation foreign to other germ cell tumour types 1286. l
sented are hypercellular and mitotically active stromal elements Germ inomas share nuclear OCT4 expression with embrvo-
resemblin g embryon ic mesenchyme, glands lined by crowded nal carc inomas (1399 1. Inconsistent and nonspecific 1s PLAP
columnar c ells with clear subnuclear and apical cytoplasm (in expression [290 ,1321 J, an d similarly non-discrim1nat1ng IS reac -
mimicry of fetal gut and respiratory mucosa) , and primitive cen- tivity for LIN28A {457 1 as well as nuclear labelling for the tran-
tral neuroepitheli al elements that may form multilayered rosettes scription factors NANOG (28041. HESRG (33881. UTF1 123831
or canalic ul ar arrays of neural tube-l ike appearance. Abortive and SALL4 (1399 ). CD30 and AFP expression 1s typ1ca11.,.
retinal differentiation is reflected in the presence of clefts lined absent. Labelling of a minority of germinomas by the CAMS 2
by melanotic neuroepithelium . and AE1 /AE3 c ytokeratin antibodies (in dot-li ke or more dtfftise
The com ponents of immature teratomas exhibit the immuno- cytoplasm ic fash ion) may signal early differentiation along epi-
histochemi cal profiles expected of their somatic tissue coun- thelial/carcinomatous lines but is without demonstrated clrn+-
terparts . Retained expression of SMARCB1 (INl1), a general cal significance {2 118). Germinoma cell subsets may express
feature of CNS germ cell tumours {1208], may assist in distin- ~ - hCG , and syncytiotrophoblastic elements expressing hPt
guishing teratomas with multilayered neuroepithelial rosettes and ~ - hCG may be foun d in otherwise pure germmomas ard
from atypical teratoi d/rhabdoid tumours containing similar should not prom pt a diagnosis of choriocarcinoma. Tumours
structures (3373). For the d istinction of immature teratomas and havin g such components must be reported as germ1noma w•th
C19MC-altered embryonal tumours with multilayered rosettes , syncytiotrophoblastic giant cells .
see the Diagnostic molecular pathology subsection , below. lmmunohistochemical and other studies have sho n that
the reactive infiltrates within germinomas include T cells tbOtr'l
Teratoma with somatic-type malignancy CD4 + and CDS + elements being represented). 8 cells, plasma
The somatic-type cancers most com monly en c ountered in tera- ce ll s, and histiocytes, in varying proportions (3452 .3579 31 I
tomas with somatic-type malignancy are rhabdomyosarcomas PD1 -immunoreactive lymphocytes are commonly present but
and undifferentiated sarcomas {290,2042,2746), followed by variable in number (3579,34451 . One RNA sequencing in Sttt.
enteric-type adenocarcinomas /982,1619) and sq uamous carci- hybridization study {31091 and an immunohistochemical Jnar;-
nomas {2042). The possibility of a teratomatous derivati on m ust sis using the SP142 antibody (34451 reponec.i POU e press;· .1
also be kept in mind in the evaluation of primitive-appearing neu- by germinoma cells, but this was restricted to acr1vated rnac
roepithelial neoplasms arising in the age groups and locations phages in a series using the EIL3N antibody 135791

386 Gern1 cell tur11uur ~;


B Syncytiotrophoblast1c giant cells

• , .;J
. ..
Flg.11.0B Germinoma. A lmmunohlstochemistry for OCT4 s11 nvr, 'J ·,ir ,;,prc·:.!:. 101 111 g8rinino:n:i ct:lls ucyt,•~'l l; 111 L 1,x·, '":l . !,
cells show PLAP expression. D lmmunohistoch em1siry tor S-metn/r 1 ,c, •)i ru 11. Cu'11id:.t le inllarntriai()ly Ct:Jli~., ydr111111oma ce:I$ ::.llt)W u11 11 itl!t:~1 k'd , ,: ,
cells can express ~ - hCG. F Ki -67 immunoh1stochem1stry sno .... s rl!c,t Ill, '>t l.Jfl' rr1ir10 111d d:li J1e prul1l<:'ru1111~1
Embryonal carcinoma non-membranous KIT expression may be seen (1399], but .AFD
Embryonal carcinomas are composed of large epithelioid cells ~ -h CG , and hPL are typically not expressed {290,1321 1
with vesicular nuclei , macronucleoli, and clear to violet-hued
cytoplasm . These can form nests and sheets, line gland-like Yolk sac tumour
spaces, and be disposed in abortive or true papillae. Embryoid The neoplasm is composed of primitive-looking eprmeliat
bodies replete with germ discs and miniature amniotic cavities cells that may be associated with loose , variably cellular. and
may be encountered (rarely) . Conspicuous mitotic activity and frequently myxoid stromal components resembling extraern-
zones of necrosis are common . bryonic mesoblast. Epithelial elements may form solid sheets
Cell membrane immunoreactivity for CD30 , although poten- but are more common ly punctuated by irregular tissue spaces
tially shared by the epithelial and mesenchymal components (reticular pattern) or aligned in cuboid profile along sinusoidal
of teratomas, distinguishes embryonal carcinomas from other channels , in some cases draping fibrovascular projections to
germ cell tumours (1399) . Consistently displaying strong form papillae known as Schiller-Duval bodies. Flattened epithe-
/tokeratin expression , with nuclear OCT4 and SALL4 labelling lial elements may line eccentrically constricted cysts (polyve-
1399). and often being PLAP-positive and LIN28A-reactive sicular vitelline pattern), some examples containing enteric-type
[457). embryonal carcinomas also manifest nuclear expression glands with goblet cells or exh ibiting hepatocellular differentia-
of HESRG (3388). UTF1 (2383). and SOX2 (2805]. Focal and tion (hepatoid pattern). Diagnostically useful , but inconstantly

,, -
Fig. U.10 Yolk sac tumour. A The characteri_stic growth paHern with loose histoarchitecture is shown. B A Schiller-Duval body. c Epithelioid tumour cells show rrabecular an
s1nuso1dal patterns, with increased mitotic act1v11y. D There is strong , often globular PAS positivity. E There is focal AFP expression.

388 Germ cell tu rnours


J -
11.11 Choriocarcinoma. A The tumour is composed of multinucleated syncytiotrophoblastic giant cells and cytotrophoblasts. B Extensive haemorrhage surrounds the
cells. C Expression of ~-hCG.

present, are brightly eosinophilic , PAS -positive , diastase-resist- Mixed germ cell tumours
ant g lobules clustered in the cytoplasm of epithelial cells or Any combination of germ cell tumour subtypes can be encoun -
~xtracellul a r spaces. tered in mixed germ cell tu mours . Pathologists reporting such
Cytoplasmic immunoreactivity for AFP, although potentially lesions should specify the subtypes present and the relative
shared by the enteric glandular and hepatocellular components proportions of each . The ind ividual components display the
of teratomas, distinguishes yolk sac tumours from other germ same immunophenotypes as the subtypes in pure form (see
cell neoplasms {290,1321,1034). Hyaline globules are also Tab le 11.02).
eactive for AFP. Epithelial components consistently label for
cytokeratins . show intense nuclear expression of SALL4, often Cytology
express glypican-3 (GPC3), and may be PLAP-positive /2062). The cytological appearances of teratomas reflect their somatic-
Yolk sac tumours also express LIN28A 1457) but not ~ - hCG or type tissue components. In smear and squash preparations ,
PL. OCT4 expression is most exceptional , and KIT reactivity germinomas display large tumour c ells with deli cate , vacuo-
(rare) is focal, non-membranous, and without Golgi area accen- lated cytoplasm and prominent nucleoli admixed with small lym-
tuation when present {1399). phocytes {2239,1477). A tigroid extrace llular background may
be appreciated in material stained by the Giemsa method or
Cboriocarcinoma related methods /1477} . A pseudopapillary structuring of tumour
Syncytiotrophoblastic elements are represented by giant cells cells can be encountered in squash preparations / 156}. Embry-
containing multiple hyperchromatic or vesicular nuclei , which onal carcinomas show cohesive cl usters of large epithel ioid
am often clustered in knot-like fashion within a large expanse tumour cells with prominent nucleol i and abundant cytoplasm in
of basophilic or violaceous cytoplasm . Such cells surround or squash and smear preparations. Yolk sac tumou rs show cohe-
partia11y drape cytotrophoblastic components , which consist sive clusters of epithelioid cells with distinct nucleol i in smear
of cohesive sheets of large mononucleated cells with vesicular and squash preparations , wh ich may also contain spi ndled
nuclei and clear or lightly eosinophilic cytoplasm . Ectatic vas- mesenchymal elements and myxoi d material. The presence of
cu1ar channels , blood lakes, and haemorrhagic necrosis are syncytiotrophoblastic giant c ells in squash or smear prepara-
characteristic . Syncytiotrophoblastic cells exhibit diffuse cyto- tions should raise the question of choriocarcinoma, particularly
plasmic immunoreactivity for ~-hCG and hPL 1290,1321 ,1 034) . in a haemorrhagic and necrotic background . In mixed germ cell
Cytokeratin expression is the rule , with some choriocarcinomas tumours, cytolog ical appearances reflect the germ cell tumour
also expressing PLAP, but KIT and OCT4 labelling is not seen components present , as described in pure form
11399,1034 1.

11111111.02 Expression patterns of germ cell tumour markers in individual germ cell tumour components

OCT4 smc PUP KIT SAL 4 CD30 AFP ~-hCG LMWCK

Germinoma

~
Teratoma
carcinoma
Yolk sac tumour
+
+ +
+
+
+
+
+
+I-

+/-
+

+/-'
+
+
+
+
+
+

t/-J
- +' _.Tb

+
fl
.

- -
5mC, 5-methylcytosine; LMWCK, low-molecular-weight cytokerati11. .
1HlCG can be expressed in a proportion of typical germinoma c;ells and u1 syncyt101roph1c gram cells in other\\ ,sfl p... re l)erm1Po1r' I~ '( v~· <~c• at1· 1' ..!II t't xpr ~Sc.: . ' ..... , ,•, ',
ot lyptcal germinoma cells, often in a dot-like pattern. <KIT can be f0tnd :n rne~e nchyrnal or eµ1tl1e l1 01cl compor.i::ms le.g int!I , ·1:.i:-1!ds1 '<\H <'Jr, '1J t' 'J't-., _ _,,1 '. • -. ' i .. •

cr.Jffiponeots. "Cytokeratin is expressed in the epithelial compor 1ani:. of! rnroma

,, I I 1 1
-
Diagnostic molecular pathology
Molecular diagnostic methods currently pl ay only a minor role in
the diagnosis and subclassification of CN S germ ce ll tumours,
which generally rest on histopathologica l and immunoph eno-
typic features . Some of the microRNAs upregulated in these
lesions may be detected in th e ce rebrospin al fluid (CSF ) or
blood , making liquid biopsy pot entially feas ible (3 371 ,23771 .
Immature teratomas harbouring multil ayered neuroepithelial
rosettes and developing neuroectodermal tissues do not dem-
onstrate chromosome 19q13.42 amplification !2270), a feature
that distinguishes them from C19MC -altered embryonal tumours
with multilayered rosettes . Genome-wide copy-number analy-
sis may help in distinguishing immature teratomas from other
tumour types , the former generally having balanced genomes.

Essential and desirable diagnostic criteria Flg.11.12 Germinoma. Cytological touch preparation showing large p1~f"1~r. 'tr
mour cells with clear cytoplasm admixed with inflammatory cells
See Table 11 .01 (p . 383) .

Staging macrophage-derived NOS2 l3109 f to be negative progri~ie.


Extent of disease is established by craniospinal MRI and CSF indicators, whereas conspicuous immune cell 1nfiltrat1on ar<j
cytology. Current pretreatment recommendations also include high-level CD4 expression may carry positive 1mphcat10
the examination of serum and CSF levels of AFP and hCG (3109}.
(see Prognosis and prediction, below) (2179). Highly elevated Historically, dismal outcomes have characterized intracranial
AFP and hCG levels suggest the presence of aggressive yolk yolk sac tumours, embryonal carcinomas. and chonocarc.-
sac tumour and choriocarcinoma components, respectively, nomas in pure form , as well as mixed tumours 1n which these
although the correlation between marker elevation and histol- were prominently represented {2042,2817,2041) . An analyStS
ogy is imperfect (particularly in cases of limited biopsy or sub- of 153 patients treated from 1963 through 1994 found that ttie
total resection) . 3-year survival rates of patients with these pure and mixed
tumour types were 27% and 9.3% , respectively 120421. Mixoo
Prognosis and prediction tumours in which these aggressive elements were only minor
CNS germ cell tumours occurring as congenital lesions (usually components, immature teratomas. and teratomas with somatic
immature teratomas and often immense on discovery) are asso- malignant change were associated with a 3-year survival rate of
ciated with a high mortality rate . A literature survey of 90 fetal 70% in this series . Regimens combining irrad1at1on and chemo-
intracranial teratomas identi fied only 7 survivors after resection therapy have now pushed overall 5- and 10-year survival rates
!1417}. The prognosis of tumours arising beyond early child - for patients with aggressive lesions to 75- 80% 1440.31081
hood varies by histology. The best outcomes attach to pure and Logarithmic decreases of serum AFP and hCG in response
fully mature teratomas (curable by surgical means alone), pure to chemotherapy are favourable predictive indicators \15771
germinomas (curable by irradiation or chemoradiotherapy), and whereas CSF or serum AFP levels > 1000 ng/ml and residual
mixed tumours combining only these elements; 10-year survival disease on completion of treatment carry negative connota-
rates approach or exceed 90% in these cases !2042,33,441, tions !440). An important exception to the latter is the scenar.o
3108) . Comparable rates of control have been achieved with in which a tumour that is persistent or enlarging after tnerapy
appropriate radiation dosing !2910,2301) and radiochemo- is found to consist entirely of mature teratoma (growing tera-
therapy (441} of germinomas that harbour syncytiotrophoblas- toma syndrome), because in such cases radical resection rna
ti c elements or that are associated with elevated hCG levels effect disease control {1617). Unfortunately. long-term survivors
in the serum and/or CSF. Select series have found atypical of both germinoma and non-germinomatous tumours remam at
(e.g. basal ganglionic I thalamic) locations !3108), evidence of risk of premature death due to treatment-associated maltgnam
chromosomal instability (3108) , and high intratumoural levels of neoplasms, stroke, and other unwanted effects of therapy l ·41

39 0 Gem1 cell iurnours


Tumours of the sellar region ~ Introduction Lopes MBS

Tum ours of the sellar region include a diverse set of paediatric


and adult neoplasms that arise in the region extending from the
sella turcica inferiorly to the floor of the third ventricle superiorly. lll

Based on current understanding of the clinical , molecular, and


morphological features of these neoplasms , there have been
additions of new tumour types and modifications of existing
tumour types in the fifth -edition WHO classi fication of CNS
tumours .
In past editions , adamantinomatous and papillary craniopha-
ryngiomas were considered to be histological subtypes of cra-
niopharyngioma . In the current edition , adamantinomatous cra-
niopharyngioma and papillary craniopharyngioma are classified
as distinct tumour types, each discussed in their own section .
_ -- _ ...
-.-........
Although these tumours arise in the same general location and
both display squamous differentiation, it is now clear that they
should be classified separately, because they have distinct clini-
cal demographics, radiological features , histopathological find- Fig. 12.01 Tumours of the sellar region. Methylation profiling ot tu ours of!"~~ ,.
ings , genetic alterations, and methylation profiles (2173,1335). region demonstrates distinct clusters for adamantinomatous c amopharyng :1.. ::ac-
Pituicytoma, granular cell tumour of the sellar region , and spin- illary craniopharyngioma, and subtypes of pituitary adenoma / pituitary . eu~:r
crine tumour (PitNET). Pituicytoma, granular cell tumour, and sprndle cell onccc-,-- a
1e cell oncocytom a, which constitute a highly related family
cluster together.
J1 tumour types , are discussed together in a single section
(2094) . These neoplasms are all circumscribed and low-grade
and arise along the posterior pituitary or infundibulum. Because including the concept of classifying tumours by their anter ~·
of their shared immu noreactivity for thyroid transcription fac- pituitary cell lineage according to comb ined 1mmunoh1stocner-
tor 1 (TTF1) and close clustering on methylation profiling, they ical expression of pituitary hormones and transcri ption fac!crs
are all thought to have a shared histogenesis from pituicytes, as well as the definition of new subtypes, and mod1f1ca .., s
the primary glial cell of the posterior pituitary. Although these in histological grading . This chapter also introduces the n~ •
tumours may in fact represent morphological variations of the nomenclature for PitNETs proposed by the WHO Classif car"
same entity, their patient demographics and clinical outcomes of Tumours' endocrine group, which will be fu rther discussed ,"
differ, and they are still classified separately in the current edi- the fifth-edition classification of endocrine tumours 126781
tion (832) . Pituitary blastoma, a rare embryonal neoplasm of 1 n ·a~c,.
Pituitary adenomas I pituitary neuroendocrine tumours (Pit- composed of prim itive blastemal cells, neuroendocnne ce '.S
NETs) have been included in this fifth-edition volume because and Rathke pouch epithelium , has been added as a vrc..;r
they are typically resected by neurosurgeons and diagnosed type to th is fifth-edition volume on CNS tumours. These t fT'atr
by neuropathologists. We have followed the guidelines set forth are strongly associated with underlying germline vanat1on ,,
in the fourth-edition classification of endocrine tumours 11919). DICER1 !707}.

392 l u1 nuurs u f 11 if; ~;c~l l dr ruq1u 11


Adam anti nomatous cran iopharyng ioma Santagata S
Kleinschrnidt- DeMaste rs BK
Kornori T
Muller HL
Pietsch T

Definition
· mantinomatous craniopharyngioma is a mixed solid and
. . , - ic squamous epithelial tumour with stellate reticulum and
-e eratin. usually localized to the hypothalamic-pituitary axis
a characterized by activating CTNNB1 mutations.

tCD-0 coding
9351/1 Adamantinomatous craniopharyngioma

rcD-11 coding
2F7A.Y & XH15X9 Other specified neoplasms of uncertain
behaviour of endocrine glands & Craniopharyngioma, ada-
mantinomatous
Fig.12.02 Adamantinomatous craniopharyngioma. A T1-weighted postcontrast MRI
showing a 34 mm sellar mass with suprasellar extension stretching the optic nerves
Related terminology and optic chiasm in a 22-year-old man with a 1-year history of excessive thirst and
one urination, fatigue, and hair loss. B T1-weighted postcontrast MRI showing a 37 mm
predominantly cystic mass with a thin rim of enhancement extending into the third
SUbtype(s) ventricle. In prior imaging there was a 16 mm region of nodular enhancement inferiorly.
The 18-year-old man had a 2-month history of headache that had become acutely
r~ne
worse, with new onset of nausea and vomiting. The mass demonstrated peripheral
calcification on CT.
Localization
Ajamantinomatous craniopharyngiomas arise anywhere Imaging
c'ong the craniopharyngeal canal, but most occur in the sellar On MRI and CT images, adamantinomatous cran iopharyngio-
2!1d infundibulotuberal region {2173}. The majority (-95%) mas are intrasellar and parasellar tumours with sol id and cystic
nave a suprasellar component (purely suprasellar, 20-41 % components {3389} . Imaging features follow a 90% rule: about
:rl cases ; both suprasellar and intrasellar, 53-75%) , whereas 90% are predominantly cystic , about 90% have prominent cal-
ourely intrasellar craniopharyngiomas are less common (-5%) cifications , and about 90% take up contrast media in cyst walls
i1558 }. Occasionally, a tumour extends into the anterior (9%) , 11486,2173} .
middle (8%), or posterior (12%) fossa . Very rare examples
~ccur in the cerebellopontine angle and other ectopic sites Spread
1101 91 Local invasion of hypothalamic . visual tract , and vascular struc-
tures (including encasement of the internal carotid arteries) 1s
CHnical features common (occurring in -25% of cases) 13556,2173! Subarach -
Craniopharyngiomas are rarely detected incidentally (in < 2% noid dissemination or implantation along the spinal cord the
of cases) !311} . The diagnosis is often made years after initial surgical track, or the path of needle aspiration 1s rare (1331
manifestation of nonspecific symptoms related to increased 1708,2848,437).
1tracranial pressure, such as headache {1330 ,2171) . Primary
"'lanifestations include visual impairment (62-84%) 12575) and Epidemiology
endocrine deficits (52-87%) affecting growth hormone (75%) , Craniopharyngiomas constitu te 1 2-4.6% ot all 111tr3cran1al
~H or FSH (40%), TSH (25%) , ACTH (25%), and antidiuretic tumours , with an inciden ce of O 5-2 5 cases per 1 m11i1011
nr;rmone (17-2 7% of patients have central diabetes insipi- person-years 1409 2253,3562,2J_,~3) Acl:i.111anr111oma.tous cra-
jus at diagnosis) I1558,2173} . Reduced growth rates before niopharyng1oma;; art! the most l c•mn 1 n r on- k u1 ) n1theliJI
'.J tagnosis may occur in patients aged as young as 12 month s inlracerebr '11 ne0p!a:::;ms 1n ch ldr e1 (.'.K"~i 1t,rn,n~i 1 )I ' :i- • 1-' o
2171 I Weight gain , predictive of hypothalamic obesity, tends of 1ntracra111al lL rnuu1 ~ :n 1t"i1s cl<,_, • CJr 'P' 1 ..)• )...I "'-, )..__,.
) \ Th"'\' ~-.

trJ occur as a later manifestation, shortly before d1agnos1s ace 1unt T H 11 · ;.:-., Iv ~Ii ,~·::.r 1c·~1~ ,, rvr 1 1c1ri ,, 1 1, l: 1 se :n 2ri1l-
and during the first year after diagnosis {2170) Ail nm>t half drer1 and cibuut b J% •.1 l rar"<•~ ~1•rvnJ 1 r: 1_. ,ii 'qr 1, t', 11 ,l~iulr:>
0! patients develop hypothal amic syndrome f rorn di seaso- nr 1~)438,J!:>t)_> ?~l~ll.JJ

:reatment-related hypothalamic involvem ent 01 c!Rmage 12170, l twrt' i··~ ~ IJ111 'Lhl < ';t' ·',I I ,_.I
2171 .2172,313,8431; the hypothalamic syridr(,fl1~ 1s assoc i- done e PtJdk--; 1n \ ·t 11Ljrt't 1 (~ 1~ Vt , : ) Ji', I l I :~}

ated with morbid obesity, cognitive unpnu 1r '.t. pl-.r --.0nality RartJ neonatnl/rl't~tl CLI ,t=;., .__, ,·u. I' ;7 4 1 ••• , I k

changes, and psychiatric symptoms 12362 .:. 'Uf; sex pred1lect1on HOLJ.~~[1f''..! _ l~' 1 • • '
1
the WNT signalling pathway regulator ~ -r,ateni r, l.2877 · c-.F 7
~26 , 355,133~,1993 , 1147, 1234 , 129). T he~e are ai:ti ,iaiing ~n ~t'a­
t1ons , as evidenced by overexpress1on of ..,n.-"at ,, eri ir. t ar get ~
sue h as AXIN2 and LEF1 (1334). Many publications roo;;
CTNNB1 mutations in .about 60- 75% of samples 1 4 2 1~~ 6 7
2877). and ~ore ~ens1t1ve sequenci ng methods and analytrr....;3,
approaches 1dent1fy CTNNB1 mutations in as many as ~ ooo~,
of s~mples (355 , 1~9} by more rel iably identifying low all'3r.-:
fra~t10~ mutations 1n samples with small amounts of tumc•Jr
ep1thel1um . CTNNB1 mutations are clonal driver even s (355
128,129). but nuclear localization of P-catenin is observed ,r.
only some tumour cells . Additional recurrent mutations have ot
been reported . However, in a familial case of adaman rnama-
tous craniopharyngioma with wildtype CTNNB1 , germline and
somatic inactivating mutations were identified in APC f 142J
suggesting that mutations in other components of the W~ JT
signalling pathway may rarely contribute to the pathogenesis IJf
craniopharyngioma . Consistent with the distinct histology and
driver mutations in adamantinomatous craniopharyng1oma al1d
Fig. 12.03 Adamantinomatous craniopharyngioma. Solid and cystic mass with calci- papillary craniopharyngioma, the tumours also display distinct
fication , first diagnosed 4 years earlier.
methylation and transcriptional profiles 113351. Recurrent focal
deletions of Xp28 have been described in a subset of samplss
Etiology from male patients, and other recurrent gains have also been
The etiology is unknown . Occasional familial adenomatous described (1147}.
polyposis I-associated cases of adamantinomatous cranio-
pharyng ioma that lack CTNNB1 mutation and instead harbour Macroscopic appearance
germline APC mutation with somatic loss of heterozygosity have Craniopharyngiomas are sol id and cystic . The cyst fluid 1s darx.
been reported {2413} . greenish -brown , resembling machinery oil. Secondary changes
are common , such as fibrosis, gliosis, calcifications. and cho-
Pathogenesis lesterol deposition . The lobulated masses have irregular sur-
Craniopharyngiomas are proposed to arise from cellular ele- face s that strongly adhere to surrounding structures.
ments related to the Rathke pouch (craniopharyngeal duct),
which is integral to pituitary development {2173}. Expression of Histopathology
oncogen ic p-catenin in early embryonic precursors and in stem The well-differentiated tumour epithelium forms cords. lob les
cel l popu lations of the pituitary drive the formation of tumours ribbons , nodular whorls , and irreg ular trabeculae. Peripheral
resembli ng adamantinomatous craniopharyngioma {1047,110) . crowding and palisading are prominent. Degenerative fearures
SOX2-positive progenitors may also underlie the formation of such as fibrosis, calcification , and nodules/whorls of anucleate
papillary cran iopharyngiomas (1 263) and Rathke cleft cysts remnants of ghost-like squamous cells (termed "wet ke aun ·
{3761. Similar stem cell populations for adamantinomatous and are common . Loose microcystic areas of stellate reticulum often
papillary cran iopharyngiomas {1037} may explain shared pat- intermingle between the wet keratin and more densely arranged
terns of cytokeratin expression (1763,3143,3491,1815}; scat- areas of tumour epithelium. Cysts are often lined by an attenu-
tered cells expressing pituitary hormones {3093). chromogra- ated , flattened epithelium . Finger-like tumour protrusions e tenc
nin A 13504), and hCG {3101} ; and the occasional presence of into surrounding gliotic brain tissue with numerou s Rosenthai
mixed transi tional tumour and cyst phenotypes (2815 ,1128,935, fibres . A secondary degenerative feature is xanthogranu ma-
2845 ,200 1}. In adamantinomatous craniopharyngioma, SOX2- tous reaction to ruptured cyst material , whic h 1s characten
positive stem cel ls may contribute to the formation of epithelial by cholesterol clefts , haemosiderin deposits , xanthoma c . is
whorls with nuclear localized p-catenin . The whorls are quies- multinucleated giant cells , and lymphoplasmacytic infiltrate
cent and secrete numerous factors including sonic hedgehog , This extensive reaction can constitute a substantial (s mettm s
FGF, TGF-p, BMPs, and proinflammatory mediators 1425,109, near-total) component of the surgically excised material. r ~e ' ­
649,1138,651 ,127,3761 . These signalling centres are analogous sitating a careful search for residual, identifiable epitheh im d1 ·
to the enamel knot that controls tooth morphogenesis 1127,3761, wet keratin , as xanthogranulomas of the sellar region a.re a s c1-
an d they implicate paracrin e signalling in tumour formation and ated with a ruptured/haemorrhagic Rathke cleft cyst.
signal transduction via primary cilia 1649}. Histological and Histological malignant progression in adamam1nomatous
molecular parallels with odontogenic tumours suggest simi lar craniopharyngioma is exceedingly rare , and 1t general! . only
cells of origin and similar mechani sms of pathogenesis 1254, develops after multiple recurrences and rad1arron the rap iften
2433 ). and they explain the occasional presence of teeth in decades after first diagnosis {2701 ,2980.2211,3367 Hist -
adamantinomatous craniopharyn gioma 1225) . pathological features range from squamous cell carcrn
Adamantinomatous craniopharyngi omas are characterized ameloblastic or odontogenic ghost cell carcinoma, but larg
by mutations 1n exon 3 of C TNNB1 , the gene that encodes percentage lack specific histological features {270 1.2102)

394 Tumours ot the sellar region


lmmunophenotype Grading
p63 is expressed in all epithelial layers of adamantinomatous Adamantinomatous craniopharyng ioma is regarded as CN S
craniopharyngiomas {2142}; high-molecular weight cytokera- WHO grad e 1.
tins ( 34~E12 [K903]. CK5/6) and low- to intermediate-molecular-
weight cytokeratins (CK?, CK17, and CK19) are also expressed Differential diagnosis
13491 ). In contrast to Rathke cleft cysts , craniopharyngiomas The differential diagnosis incl ud e s pap illary cran 1opha ryngi-
in some stud ies lack CKB and CK20 expression except in rare oma , xanthogranuloma, Rath ke cleft cyst . epidermoid and
cells; the value of cytokeratin expression for distingu ishing these dermoid cysts . and p ilocytic astrocytoma . Xanthogranuloma of
two lesions is unclear {3491 ,1815). PDL1 is expressed in the the sellar region is a reactive lesion resulting from leakage, rup -
cyst-lining epitheli um of adamantinomatous c raniopharyngioma ture , or haemorrhage of a Rathke cleft cyst, indicated by stnps
1651). SOX2 is present in a small proportion of cells and SOX9 is of col umnar or ciliated columnar epithel ium in the resected
widely expressed (3175) . material (2427,94 ,1657,793}. They compri se cholesterol clefts.
Adamantinomatous cran iopharyngiomas have activati ng multinuc leated giant ce lls, macro phag es (xanthoma cells) lym-
mutations in CTNNB1 , w hich encodes the p-catenin protei n. phoplasmacyt1 c in fl ammation , fib rosis . eos1nophrl1c granular
The mutation is clonal and present across the tum our epithe- necroti c debris, an d haemos1denn deposits Ep1dermo1d cysts
hum {355,11 47,129); however, nuclear accumul ation of p-catenin are d ist1ng u1shed by tt1e presence of a single cavity lined by
tS spatially restricted and fo und in only a small perc ent age of keratin1zing squnmous ep1thel1um and filled with tlaky, dry l\era-
cells . most pronounced w ithin small ep 1thel1al whor ls 1426.425, t1n. Dermrnu c v , ts ddcl1t1ondlly ha.vt::: aJn8Xdl su uctures Rcithl-.e
651,129). Nuclear p-catenin is observed even wh en CTNNB I clett cysts tinve c1 we II l1n8cl by sirnpl columnar or cuboid.JI
mutations are not detected 111471 . ~pill 1ol1ur11 wt 11d 1 c1 tte11 1 ~• l 111 Hf' J. vv1tt1 rnucirh. 'US \:.JUblc;t cells
Myxo1 l/rnt.L,•''• )'.t •'t•11t ·1 1t · 1,.., \.;01~irn1.n R~)s .Hlidl t1t1rt;S
Proliferation ctrld Lh 11,,t" fJ I •
t 1, 1ct1 n,1nt11 'l r •1c1lt'd'' l ·1 u11 J-
Ki-67 is generally confined to palisad in g rtJg 1ons anc.l a!Jse11t pl1<1rvri:i1 1:r d 1' .r •11 i ,
~, .{ll•'-- 1[ Jill\ f>CJI l 't' dt• l:'
in epithelial whorls . The Ki- 67 proliferat ion 1r1dnx vanes w1dPly Cr llui.111tv :11111 t1.,111, '"· 1
' 1 {, ( ' [It.. • ii (. I >l , ,i I
between cases and provides no prognnslic 11 •t 1rrnc.tlturi [2t)UO <.'jli HI 1,1 Hf. ell••,, 1:
808,1929,354).
Flg.12.05 Malignant craniopharyngioma. High-power view of malignant craniopha- Fig. 12.06 Adamantinomatous craniopharyng1oma. Even though murati ;rs , ~ ~-;., 1
ryngioma emerging 19 years after resection and radiation treatment of previously of the CTNNB1 gene, which encodes p-catenln, are clonal and pr~~' ~S3 ,.. ~
grade 1 craniopharyngioma. Sheets of poorly differentiated cells show severe cyto- neoplastic epithelium, p-catenin protein only accumulates in the nucieiJs rl' a s-..tsi:; ~­
logical atypia, large vesicular nuclei, prominent nucleoli, mitotic activity, and necrosis. cells such as those scattered individually and In small clusters.

Box 12.01 Diagnostic criteria for adamantinomatous craniopharyngioma hypothalamic integrity can be preserved ) Gross total "ese<"r·~I")
Essential: is associated with better recurrence- free survival than suot'J·a.
Tumour in the sellar region resection, but many studies do not support an adva~age <:f
gross total resection over subtotal resection followed by 3CJJ-
AND
vant radiation (800,3033 ,3126 ,673 ,3368 .2328.2772! Ctr.er
Squamous non-keratinizing epithelium, benign
radiotherapeutic approaches and techn iques such as ~rom­
AND beam therapy are often considered (46,27621.
Stellate reticulum and/or wet keratin Overall survival rates that have been described 1n paec air•c
series are 83-96% at 5 years, 65-100% at 10 years. and or:
Desirable: at 20 years (1899.618}. In mixed cohorts including adu1.s and
Nuclear immunoreactivity for ~-catenin children , reported overall survival rates are 54-96% at 5 years.
Mutation in CTNNB 1 40-93% at 1O years, and 66-85% at 20 years 13290.2~5:
Absence of BRAF p.V600E mutation 2448}. Because overall survival rates are high. quality o' ;1·e
an essential consideration . Disease- and/or treatment-re a·ac
hypothalamic damage results in morbid obes1ry. meta
Cytology syndrome, circadian rhythm disturbances. memory de· -:s
Not relevant and neuropsycholog ical impairments (843.3033 .880.942.2 1 69
3282,313) . Although craniopharyng ioma corresponas "'
Diagnostic molecular pathology logically to CNS WHO grade 1, the prognosis is often \ orse
Demonstration of CTNNB1 mutation, as well as an absence of because of the large percentage of tumours that invade a-
BRAF p.V600E, may be helpful in selected cases. cent structures , often precluding safe gross total resec• on -
novel MRI-based grading system of presurg1cal hypotnai '
Essential and desirable diagnostic criteria involvement and surgical hypothalamic lesions shows tt'laI DS-
See Box 12.01 . terior hypothalamic involvement has a maior negaove 1m -~
on hypothalamic morbidity and quality of life 13033.3 3.:38 ''
Staging Accordingly, hypothalamus-sparing surgical and radtotNYS-
Not relevant peutic treatment strategies are recommended. Late rPorraial';
without tumour progression results from type 2 dtabeteS 1. ... -
Prognosis and prediction ebral and myocardial infarction. fracture , and se ·ere in 't
Current treatment strategies for craniopharyngioma are debated ; (3335 ,2323). Malignant transformation of craniooha g '·
they range from gross total resection to the extended transsphe- is rare and associated with a poor prognosis 12701 .:...98 ~-- 1
noidal endoscopic endonasal approach, through to limited sur- 3367). Although there are few studies of molecular aru1 '"'
gical app roaches focused on the preservation of hypothalamic dictive of worse outcome , tumours with TNN81 p T41 rJJ\; t ·
and visual integrity and quality of life {959,1304,130 ,1979,688) . tions or focal deletions of Xp28 may be associated witn d vsa
Safe total resection remains the goal when feas ible (i.e . when outcome (1147) _

396 ·1 UI f l()U I ~ ;(JI ii 1(-; St:: ll n 1 rrn_Jl[)fl


Papillary craniopharyngioma Santagata S
Kleinschmidt-DeMasters BK
Komori T
MOiier HL
Pietsch T

De AitiOn
Papillary cran iopharyngioma is a sol id or partially cystic, non-
ratinizing squamous epitheli al tumour that deve lops in the
undibulotuberal reg ion of the third ventricle floor, most often
· adults, and is characterized by BRAF p.V600E mutations.

rco-o coding
9352/1 Pap illary craniopharyngioma

ICD-11 coding
2F7A.Y & XH2BFO Other specified neoplasms of uncertain
behaviour of endocrine glands & Cran iopharyngioma , pap il-
lary

Related terminology
None

Subtype(s)
None

Localization
Papillary craniopharyngiomas arise anywhere along the hypo-
thalamic-pituitary axis, but there is a strong pred ilection fo r
intrinsic localization within the infundibulum and tuber cinereum
of the third ventricle floor. They can expand into the th ird ve ntri-
cle cavity, and they can be located entirely with in the ventric le
above an intact ventricu lar floor. lntrasellar involvement is not
common {2408 ,2380 ,3556 ,996 ,2574).

Clinical features
Primary manifestations in c lude headache (in 70% of cases)
and visual defic its (i n 63 %), the latter resulting from compres-
sion of the optic c hi asm, wh ich ofte n stabilizes or improves
after surge ry. Nearly all patients have some evidence of hypo-
pi uitansm (either partial hypopitu itarism or panhypopituita-
rism , in a 70:30 rati o) manifesting as hypothyroidism (80%) ,
hypogonadotropic hypogonadism (56%), hypocortlsolaemia Fig. 12.07 Papillary craniopharyng10111a A T1 weighted oostc1J11trasr :1.tP=11 snow111y
(50%), and growth hormone d eficiency (20%) . Hyperprolacti - a cys tic and solid mass with peript1eral and central enhancemtint. which prove(1 0n
naemia is also seen due to stalk effect (in 30% of cases) . Dia - biopsy to be a papillary craniophar yng1orna Although the cysr JnJ paprllarv rrn.:r al
betes insipidus is a primary manifestation in 25% of patients . nodule pattern rs archetypal tor this tumow on 1rnag111y. otn1:'r e.\amples 01 papillary
and in 70% of those patients it develops anew after surgery craniopharyng1orna can be rredomrnantiy so:1d or cystic: 8 rt -~~ergh rc>d postconrrast
coronal MRI ot a 28 rn 1n complex cvst1..: ma:;::; with 11odutJr :?1~r\rncemerll 111 the 111ru11
Hydrocephalus is common , occurring in 30% of cases . Preop -
dibulotubaral region ana cystir corr~p0nenb e\te1Jo.iin9 up 111!0 the !fwd ventr1c1a n1e
erative hypothalamic disturbances (in 63 % of cases) include mass involved thP p1!u1l.lry stall\ was oredo111111 rn!ly puslt'' ur tci rtre Jprrc cn1.1srn. a11d
weight gain and psychiatric and cognitive disturbances 1·1177, abulted and c1rspldC€(l t11e r;~ f)Jth,-11a1nu~ s~1 p..:r1 111 1 rt:e J1 yo:>,1r •Jill WL' · rn nrese1Hed
24071 as well as alterations in core bod y temperuture and with visual loss. 11eddache, ar•;1, 11!d °'l'd,i-_: r1t1 J:. !~1'hl1L'..;
sleep- wake cycles 136181 .
MRI rnos! nften ::rh•vVs tiu111.J,Jt', 1..'L.::; t->111 ·,· 11' •1,; •,\·'-1 •.'r'\
Imaging a sn1all µrop1··rt1u1 l '11 :\Se-' I,,, \'1' l1.. ' ' \ 'f' ·~ r1 •, -: t· 'i' ,, '
Papillary craniopharyngiomas are often f;o11d L 1 iI ·~urr,i:- tumours rner 11 lht1 p1lu1t~
I',· St :ii · 1..; l1ttt'n , ::.,1~, • 1l 1 , . , .1tt-'i_· "' ··' t" ~
are mixed solid/cystic or predomin antly c, .;t1l' r• .t~Y are ~Jbner l1ypotl1dlRmu~ 1<, ~1rtu1 , )\.\ tti. !•Ji•\• 1- , • 1
1
ally spherical (not lobulated or irregular) and c :.' . .r:r 11 ' 11 1 ts inf re · lesions utttJn ti ave ~1 :~·ltd l l 1111., ,., .~r· i .,t- 1 , ' ,
.•.;
quently seen 12812,1831,1777,9961 . Tl-Wt."!-lt • :i 1 r.stcJntrast t1yµointens 0;1 I 1-v..e1ghtecJ n.._•11:..'011tr;.is, 111 1

lurr •JUI'.,, !':·.· .


/\!most 8 11 papdlriry crar11r;ph:;irynq1rJrn 8~ h av~ BFIAF I; ·H ·J/,-:
m1 1tation s (J S5, 1RO:j ,28f.Jg . t147, 131S, 1~9'3 1?:14 1IP.arJ.rrJ tr,:~'
vat1on of tr1e MAPK/ERK p8thway l'lo oth~r r8o Jr r~r,t r' ;r~t r,r ·,
have been reported Only a relatively kwv rt 1Jrr•t"JP,r tjf r r-r ·, 1,.
onymous somatic mutation s are pre s~ n t 1n ~apr 1 1 ~r / r.r~r .t;.rJr,~
ryngiomas compared with other tumour typP,s 1n l?rqe r:r>,..,,.,..,_,
1355). Consistent with the distinct h1stol0gy and 1 n 1er rn1;1~,. r.'
in papillary craniopharyngioma (BR_AF p V600E mu;:; 1rF'~,1 ¥.'1
adamantinomatous craniopharyng1oma (CTNNB 1 m• t;:;t1r.,r-: 1
the tumours also display distinct methylation and trnn-;(: pt;or ;:;1
profiles {1335). Papillary craniopharyng1omas s ON s a J •
genomic copy-number profiles without recurrent chr0mfJ7r ~
gains or losses {2665 ,1147}.

Macroscopic appearance
Papillary craniopharyng iomas tend to be predominantly s0l!d ·y
mixed solid/cystic , but a small proportion can also b'3 rT'os· f
cystic; the cystic tumours generally have a cau liflower-trlt~ S-O 'l
Fig. 12.08 Papillary craniopharyngioma. Fixed coronal section of papillary cranio-
pharyngioma showing a solid and cystic mass involving the infundlbulotuberal region nodule. The cyst contents are typ ically described as vtscous
and third ventricle in an adult patient without prior surgical intervention. The tumour and yellow {657}. Calcifications are generally absent T1'i.q
mass has a cauliflower-like configuration. Calcifications are absent. tumours are generally circumscribed , spherical . and not ·de
adherent to surrounding brain tissue. The surface can ha•1e 3
Spread papillary pattern .
Local recurrence occurs in about 25% of patients who have
involvement of the hypothalamus and other vital neural and vas- Histopathology
cular structures {2380 ,2173}. Ectopic recurrence is a rare com- Papillary cran iopharyngiomas have non-keratirnzing m r
plication that occurs along the surgical track or at other sites squamous epithelium covering fibrovascular cores or a C't
in the CNS via cerebrospinal fluid spread in the subarachnoid wall. Stellate reticulum and flaky and wet keratin are abse •
space {3525,437}. [19,657}. Calcifications are rare . Crowding is presen in t
basal cell layer but pronounced palisading is absent. En e-
Epidemiology lial whorls and collagenous whorls are present, but they ar
Incidence distinct from those of adamantinomatous craniopharyn
Papillary craniopharyngiomas constitute 1.2-4.6% of all intra- Mitoses are infrequent. The tumour- brain interface is
cranial tumours, with an incidence of 0.5-2.5 cases per 1 mil- demarcated and invasive protrusions are absent 119}. li
lion person-years (409,2253,3562,2323}. They account for infiltrating neutrophils are common ; T cells and macropha
about 10% of all craniopharyngioma diagnoses and 12-33% of are also present throughout the fibrovascular cores aflO
those arising in adults {800,19,2438,2380,1777,2772). tumour epithelium. In as many as one th ird of cases. there are
single or small groups of PAS-positive goblet cells w1thi _
Age and sex distribution squamous epithel ium, and in a small number of cases the e
Papillary craniopharyngioma is principally a disease of adults are regions of ciliated epithelium . These histological features
(peak incidence in patients aged 30-59 years), with tumours overlap those of Rathke cleft cysts with extensive squaJ'T\Ot..5
arising in paediatric patients only rarely (657,2438 ,3562,329}. metaplasia .
There is no reported sex predilection (657,3562}. Histological malignant progression in craniopharyn
is exceedingly rare; it generally develops after multiple rec r-
Etiology rences of adamantinomatous craniopharyngioma and recetat
Unknown of radiation therapy, often decades after initial diagnosis 1270
2980,2211 ,3367). Malignant progression of papillary er .
Pathogenesis pharyngioma has been reported {1032,3367}. but none oft" se
Craniopharyngiomas have been proposed to arise from cellu- cases were tested for BRAF p.V600E mutations.
lar elements related to the Rathke pouch / craniopharyngeal
duct, which is integral to pituitary development {2173) . SOX2- lmmunophenotype
posi~ive progenitors may underlie the formation of papillary
p63 is. expressed in all epithelial layers 121421. High-
craniopharyngiomas (1263) and Rathke cleft cysts (376) . The
lar-we1ght cytokeratins (34~E12 (K903) and CK5/6) and I - ru
dev~lopment of papillary and adamantinomatous types of
intermediate-molecular-weight cytokeratins (CK?, C - ·
craniophar_vng1oma from similar stem cell populations {1037)
CK19) are expressed 134911. CK? expression is cominad !
may explain shared patterns of cytokeratin expression 11763,
the superficial epithelial layer {1763} . Some studies 111 1 a
3143 ,3491 ,1815); scattered cel ls expressing pituitary hormones
that craniopharyngiomas lack CK8 and CK20 e p ressr -
13093).' chromogranin A 13504). and hCG 131011; as well as the
occasional presence of mixed transitional tumour and cyst phe- except in rare cells , in contrast to Rathke cleft cysts. the v ltW
notypes 12815,1128,935 ,2845 ,2001). of cytokeratin expression for distinguishing these two 1e~ 'lf'S
is unclear 13491 ,18151. Primary cilia are present 1n the baSd1!\
398 Turnour~ of ti 18 sell;:tr fCQlur 1
,_
.
-
•it :
~-'
~

fig. 12.09 Papillary craniopharyngioma. A Low-power view of an H&E-stained section highlights papillary architecture. B Well-d1fferent1ated non-keratin1z1ng squamous epi-
thelium covering fibrovascular cores that contain a low density of fibroblasts and immune cells including lymphocytes, macrophages. and neutrophils. c Well-differentiated non-
keratinizing squamous epithelium with lntercellular bridges and abundant tumour-infiltrating neutrophlis, which are common in th ese tumours. D lmmunohistochemistry ror CK5 6
(an antibody against th e intermediate-weight keratins CK5 (58 kDa] and CK6 [56 kDa]) is positive throughout all layers. Staining for CK19 is also positive. Another marker commonly
used to assess squamous cell carcinomas of all types, p63, is also immunoreactive in almost all papillary and adamantinomatous craniopharyngiomas.

oriented tumour cells near the fibrovascular stroma !649 ).


POL1 is expressed in multiple layers of tum o ur ce ll s circum-
ferentially surrounding the fibrovascular stro ma /65 1J. SOX9
is vari ably expressed , and the express ion of SOX2 requires
furth er assessment {1263 ,3175) .
Nearly all papillary cran iopharyng iomas have BRAF p.V600E
mutations {355 ). which can be id entified across the tumour
epithelium usi ng mu tati on -specific antibodies, thereby dist1n -
guish1ng p ap illary c raniopharyngi oma from adamant1nomatou s
craniophar yngioma and Rathke cleft cyst (1498 ,2869.1622 1.
Rathke cleft c yst d isplays ant ibody cross -reac tivity 111 rnot1l e
cilia !1498) In pa pillary craniopharyngiorn a. p-ca tenin is co n-
fined to the cytoplasmic membrane (3551 .

Grading
Papillary c ran iopharyn g ioma is regard ed as Cr J~; WHO g1a<.fe 1

\ ' ' ;1 '


Proliferation
Ki-6_7 is 9en.erally co~fined to the basal layers (354,1263) . The Box 12.02 Diagnostic criteria for papillary cranrooliarr''.1",.. a
proltferat1on index vanes widely between cases and pro ·d Essential:
· · f . v1 es no
prognostic 1n ormat1on 1808,1929}. Tumour in the sellar region
AND
Differential diagnosis
Non-keratinizing mature squamous l!fll'lllellUm ai--.~oo tti~~--u~
!he differential ?iagnosis of papillary craniopharyngioma cyst wall
includes adamant1nomatous craniopharyngioma, xanthogranu-
lon:a, Rathke cleft cyst, epidermoid and dermoid cysts, and pilo- Desirable:
cyt1c astrocytoma . When accompanied by extensive squamous lmmunoreactivity for BRAF p.V600E
metaplasia, Rathke cleft cyst c an be diffi cult to discrimi nate Presence of BRAFp.VGOOE mutation
from papillary craniopharyngioma becau se the latter can also Absence of nuclear l}-catenin immunoreactiv1ty
contain ciliated columnar cells and goblet cells. Importantly, Absence of CTNNBt mutation
Rathke cleft cysts lack BRAF p.V600E mutations {1498,2869 ).

Cytology craniopharyngioma has a better prog nosis. in part cec:a


Not clinically relevant its well-demarcated spherical shape and fewer oo ..... 0' a- "';-
sion , which facilitate complete resection !3149 1 9 ~ c'"'r i:; ·- ~
Diagnostic molecular pathology studies have shown no significant difference 1 prog- _- 5
Demonstration of BRAF mutation confirms the diagnosis , between adamantinomatous and papillary era 1oor ar ..,g :;;..- ~
whereas CTNNB 1 mutation suggests adamantinomatous cra- (657,3410 ,800,2380) . Extent of resection 1s > 95 o ,,, 50
niopharyngioma {1234}. patients 11777}. Recurrence for papillary cranioor a,.. ·-g c. :l.
occurs in 20- 35% of patients !1777.2380 1. For crar :m- . --
Essential and desirable diagnostic criteria gioma as a whole, a higher recurrence- free s ~ r·.... a ~a·~ -
See Box 12.02. achieved with gross total resection than with sub o•a ' SSC?.:-
ti on , but many studies do not support the notion t a g-s..:-
Staging total resection has an advantage over subtotal rese.... : ~~ :; -
Not relevant lowed by adjuvant radiation therapy in terms of or og re::~ - ,. . -
free survival {800 ,673,2328,2772). In mixed co orts ~c _,...._
Prognosis and prediction ing adults and children, overall survival rates are 54-36
Studies often report outcom e measures that include patients 5 years , 40-93% at 10 years , and 66-85% at 20 j ears 3_ - .
with adamantinomatous an d pap il lary craniopharyngioma , 2455 ,2448). Rapid and dramatic tumour responses a ~ ~ e-=­
with papillary craniopharyn gioma often making up only reported in patients with BRAF p.V600E-mutam oa+ ~..
10- 20% of the total. Therefo re , information about prognosis cran iopharyngioma treated with BRAF and/or 1E f"l ..., ~ =:.: ·
and complications specifically for patients with papillary cra- {354,1515); a multicentre phase II ch nical tna 1s 01;;0 ~ "
niopharyngioma is limited . It has been proposed that papillary evaluate targeted treatment 1610}.

400 Tumours of tr1e sellar regio n


Pituicytoma, granular cell tumour of the Lopes MBS
Kleinschmidt-DeMasters BK
sellar region, and spindle cell oncocytoma Mete 0
Roncaroli FR
Shibuya M

Definition somatotroph adenomas/tumours (1402,2749,4441 A small


Pituicytoma, granular cell tumour of the sellar region, and spin- number of patients with pituicytomas and granular cell tumours
dle cell oncocytor:1a constitute a distinct family of low-grade have presented with hypercortisolism or acromegaly ~1t~out
n~plas~s that arise '.rom pituicytes of the posterior pituitary demonstration of a synchronous pituitary adeno'.11a I p1tu1tary
or 1nfun~1bulum: most likely representing a spectrum of a single neuroendocrine tumour (PitNET) 11402). Massive 1ntraoperat1ve
nosolog1cal entity, all showing expression of thyroid transcrip- bleeding or spontaneous haemorrhage can occur in ~p i ndle cell
tion factor 1 (TTF1 ). oncocytomas , possibly owing to their hypervascularity 11658).

ICD-0 coding Imaging . . .


9432/1 Pituicytoma No specific features have been identi.fie~ to ~1s~ 1 ngu1sh these
9582/0 Granular cell tumour of the sellar region three lesions from clinically non-functioning p1tu1tary tumours.
8290/0 Spindle cell oncocytoma They are isointense on T1-weighted MRI and show either homo-
geneous or heterogeneous enhancement. T2-weighted MRI
ICD-11 coding reveals various intensities 1647,2911}.
2F7A.Y & XH59V4 Other specified neoplasms of uncertain
behaviour of endocrine glands & Pituicytoma Epidemiology
2F7A.Y & XH2XW8 Other specified neoplasms of uncertain Pituicytoma, granular cell tumour, and spindle cell oncocytoma
behaviour of endocrine glands & Granular cell tumour of the are rare; there are no epidemiological data available at present.
sellar region A recent meta-analysis of literature published in either English
2F7A.Y & XH26P7 Other specified neoplasms of uncertain or Spanish identified about 270 cases of the entire group (1180}.
behaviour of endocrine glands & Spindle cell oncocytoma The majority of these tumours occurred in adults in the fifth and
sixth decades of life (median : 48 ± 21.8 years) \1180); patients
Related terminology with spindle cell oncocytoma were older (mean age: 61 .6 years)
Pituicytoma (2718) . Sex distribution varies in the literature; a slight male pre-
Not recommended: pilocytic astrocytoma of the posterior pitui- dominance is reported for pituicytoma 1647,27921 and a slight
tary; posterior pituitary astrocytoma; infundibuloma . female predominance has been reported for spindle cell onco-
cytoma 1647) and granular cell tumour \647,3600).
Granular cell tumour of the sellar region
Not recommended: Abrikossoff tumour; choristoma; granu-
lar cell myoblastoma; granular cell neuroma; granular cell
schwannoma.

Spindle cell oncocytoma


Not recommended: spindle cell oncocytoma of the adenohy-
pophysis.

Subtype(s)
None

Localization
Pitu1cytomas, granular cell tumours , and spindle cell oncocyto-
mas arise along the length of the posterior pituitary and infun -
dibulum, forming suprasellar or sellar/suprasellar masses . Spin -
dle cell oncocytomas occasionally extend into the cavern ous
sinus and invade the sellar floor 12911 l.

Clinical features
Symptoms are indistinguishable from those of other reyiom1I
,~ 1 ,,,, {1.~n, .J • ,_;i.;a!
lesions and include headaches, visual field fJefects, and hytpo- .... Ht·,. 11r\;(j ,

r!Y :P\iP::' ~.lfll1 ~J ·~ .... '1 J f ·: l..l::f )~lld? it.. 1 • r.. 1


pituitarism \29111 . Diabetes insipidus 1s u11..::ur111 r101 1 !G47) S1ud1es
1

re ...i n:1l·J ··1 l. >: ;t,? r·' hr .11, •._.,n !1.: , 111 r:.. µ · 'd·y !iJLr .. 'n-• .J , ~; '~:, r1 '1.
1
1

'.ituicytomas and granular cell tumours h ~1ve t"J8en repo rr ed


: ; ....

1
µresenct: o. I li~ 11 [Jt.:" UL .. ' ,, .;:! .1 ·~·.'ti"' ' Sl!•I r ~ .. ;.,' ..1, '..ti -:iS) ..-. ·.,
in association with synchronous tun cr1c~r.a l cut1crnrciph crnd Hov.evi:" , b1o~">Y pruvr:!O ti11: 1,, c. d µ.<·; .:v L ~;a

I
~ ! l . f ~ > ~ l f ; ; l i:,:!
1
i
• I
Etiology several mutations , including in the HAAS, SNO T ano c - .
The etiology of th ese neoplasms is unknown. No germline sus- genes in 4 spindle ce ll oncocytomas from 3 patients 12 .... -l .
ceptibility has been identified . DNA methylation classifica,tion The HRAS-mutant case also had a MEN 1 framesMt
demonstrates close clustering between the three tumour types , (2114). Similarly, constitutive MAPK aciivation was fo
with assignment to a single methylation class (460). 11 pituicytomas including HRAS somatic mutatJOns as
pathogenic BRAF p.V600E, NF1. and TSC 1 sequence
Pathogenesis (3323} . An additional case of a spindle cell ona:'JCVtorra
Ubiquitous nuclear TTF1 expression indicates a common BRAFp.V600E mutation has been shown to respond o
derivation of pituicytoma, granular cell tumour, and spindle cell inhibition of the MAPK/ERK signalling pathway 129861.
oncocytoma from the pitu itary infundibulum I forebrain gangli- study provided the fi rst whole-microRNA signarure of
onic eminence (ventral neuroectoderm) rather than from endo- cell oncocytomas, wi th distinct microRNA profiles di
crine cells of the anterior pitu itary or from folliculostellate cells ing primary tumours from recurrent tumours (1741) T
11829). Similarities between these tumours and the normal light , study also linked these tumours to an altered me
dark, granular, and oncocytic pituicyte subtypes are consistent notype related to lipid metabolism and the Krebs eycle p7 '
with an orig in from the posterior pituitary 13115,1829,2094) .
Macroscopic appearance
Genetic profile The three types of tumours cannot be disttngu1sheo o~ tr.~·r
The pathogenesis of these tumours has yet to be fully eluci- gross features. Their texture reportedly ranges from S1m1 a1 tc
dated. Methylation-based classification studies show close that of normal brain to firm and vascular, and tnetr col ur trOfT
clustering, suggesting they may be a single tu mour type with grey to yellow 12167}. Pituicytoma and spindle cell oncoq.roi 3
a shared histogenesis but distin ctive morphology 1460). IDH1 can occasionally be associated with haemorrhage I 24, t1 I
p.R132H mutation and KIAA15 49: :BRAF oncogene fusion
am absent 12094). A limited number of case series reported Hlstopathology
variable somatic alterati ons with some evidence suppor t- Pituicytomas
ing MAPK pathway ac tivation in p1tuicytoma and spind le ce ll Pituicytomas are composed of elongate bipolar spindle cells
rmcocytoma 12 11 4,3323 ). Whole-exome sequencing identified often arranged in soli d sheets and short fascicles. which can
'10? l1Jrr1ci1ir•, rJf tl1r! '.( . 11; 11 rf)l)IOl l
'"'3 .,. 3 ~ norm pattern 1365.2094 ,2093 ,1658 ,19191 Tumour
~t> 's ter ) to show d1st1nct cell borders These tumours lack
)S •I h1hc granular bodie.s. Rosenthal fibres, cytoplasmic
t'\: s·nop the coarse granularity. cytoplasmic vacuo lization . and
ahnized blood vess.els I1658) Like in spindle cell oncocyto-
s. rnf lamma~or.y infiltrates can sometimes be present !1658 ,
1
29} Some .p1tu1cytomas can display regions with ependymal
and oncoc tic change 12773,3548). These observations have
raised the possibility of morphological continuity among pitu-
1 • te-related tumours of the posterior lobe 12773.2911,20931

'ttJ1cytoma.s ma·y· be .distinguished from normal posterior pitui-


tary by the 1dent1f 1cat1on of Herring bodies and the presence of
a ns (NFP expression) 1n non-tumorous posterior lobe.
Atthough pituicytomas typically show a low mitotic activity
and low Ki-67 labelling index (often < 3%) \1215,33231. rare Flg.12.13 Granular cell tumour. Postcontrast coronal MRI shows homogeneous mild
examples with atypical features characterized by increased cel- enhancement within the mass and a relatively normal calibre of the more caudal stalk.
l larity. pleomorphism, mitotic activity, and elevated Ki-67 label-
h g index(> 5%) have been reported {3323,1215). pituicytomas from granular cell tumours and spindle cell onco-
Unlike granular cell tumours , pituicytomas lack the PAS-pos- cytomas , respectively 12715,1658). . . .
itive and d iastase-resistant intracytoplasmic granules {1658). Pituicytomas show ultrastructural characteristics of light and
Unlike schwannomas , reticulin histochemistry shows no pericel- dark pituicytes - two of five ultrastructural subtypes of non-
lular staining {1658). tumorous pituicytes \3115) that are enriched in intermediate fila-
By immunohistochemistry, pituicytomas invariably express ments (2094) .
TTF1, and they are negative for cytokeratins, pituitary hor-
mones and transcription factors , chromogranin A, synapto- Granular cell tumour of the sellar region
p ysm, and neurofilaments. They show strong reactivity for Granular cell tumours consist of densely packed polygonal cells
vimentin and 8100, but variable GFAP immunostaining . The with granular eosinophilic cytoplasm . The architecture 1s typically
tumours also express variable EMA , CD56 , galectin-3 , CD68 , nodular; sheets and/or spindled/fascicular patterns can also be
and BCL2 {2094 ,1098). Staining patterns for a1-antitrypsin and seen . PAS staining of cytoplasmic granules is resistant to diastase
antimitochondrial antibody can also assist in the distinction of digestion. The tumour cell nuclei are small, with inconspicuous


• • ••.. • .... • • 4:


•• !' "
•,.


•• ..*.
,,,

••
- ·-

• • •• "'• • •• •.

• •• • •
.•. • • ...
. '

• 0

~
, •• •
• •• •
•. ,. •
• . .' (
.." •
~

·r - "
'.l -
' •
Flg.12.14 Granular cell tumour of the sellar region . Tumours Jre ct1aractcr1 zed by eos1noph1llc polygonal cells with abundant granular eos111oph111c \..vtoµl-1s1'1 (Al Tri : "0: s::; :t1.~
marked PAS positivity (B), strong EMA expression (C), and nuclear P>(Jresslon of thyroid transcr1pt1on tactor 1 (TTF1) (0)

ir__ ,.,
I u1 rh_H r-.; r 1 .,
are negative for NFPs, cytokeratins. chromogranin A, synapto-
physin, desmin, SMA, and the pituitary hormones and transcrip -
tion fac tors !2094,1658) .
Ultrastructural analysis highlights the abundant cy oplasmic
lysosomal population of the tumour cells that confers the granu-
lar aspect of the cytoplasm seen on light microscopy. NeuroE~­
cretory granules are absent.

Spindle cell oncocytoma


Spindle cell oncocytomas are typically composed of inte·lar:-
ing fascicles and poorly defined lobules of spindle to eprth8'10rrj
cells with eosinophilic , variably granular cytoplasm Onc0cyi•i:.
changes can be focal to widespread . Whorls , myxoid changes
clear cells, osteoclastic-like giant cells , and follicle-like struc-
Fig. 12.15 Spindle cell oncocytoma. Tl-weighted , postcontrast coronal image dem- tures can also be features of these tumours (2733 .3261 .3548)
onstra'.es a lar~e , expansile, heterogeneously enhancing sellar mass with parasellar Mild to moderate nuclear atypia and (less commonly) marked
extension and invasion of the clivus. pleomorphism can be observed . Focal infiltrates of ma ure
lymphocytes are common .
nucleoli and evenly distributed chromatin. Perivascular lympho- Their immunoprofile includes TTF1 , vimentin , 8100, EMA.
cytic aggregates are common. Mitotic activity is usually incon- ANXA1, and galectin-3 expression . EMA expression var-
spicuous, and proliferative activity is usually very low. ies from diffuse to limited to a few tumour cells . Focal GFAP
Granular cell tumours show nuclear staining for TTF1 (1829, expression can be present. MU213-UC , an antibody aga1ns a
2094). The tumours are variably immunoreactive for SIOO and non-glycosylated 60-kDa mitochondrial protein, can help iden-
vimentin, but only occasionally immunoreactive for GFAP and tify oncocytic features (2715) . Chromogranin A is absent, but
EMA (2094,1658) . The tumours are also positive for CD68, synaptophysin (619) and faint and focal CD56 immunoreactivity
a1-antitrypsin, a1 -antichymotrypsin , and cathepsin B, and they have been described in some cases {2094,2004,1181). P1tu1tary

Fig. 12.16 Spindle cell oncocytoma. A Intersecting fascicles of tumour cells show the plump eosinophilic cytoplasm typical of this tumour. Increased cytoplasmic volume is du~
to increased mitochondrial content. B The clear cell appearance of the tumour cells can be seen, here with cells arranged in a nested pattern. C Tumour cells generally show
more oval nuclei than do those in schwannoma (which usually contain tapering nuclei with pointed ends). Note the absence of nuclear pseudoinclusions, as can be seen 1n some
meningiomas. Note also the uniformity of nuclear features and absence of mitotic activity. D There is diffuse nuclear immunoreactivity for thyroid transcription factor 1 (TTF1l in
spindle cell oncocytomas, but it is also in pituicytomas, in granular cell tumours of the posterior pituitary, and even in normal posterior pituitary gland, so this immunostain d~s
not distinguish these types of lesions from each other.
lu 12.0l Diagnostic criteria for pituic to 1
Y ma. granu ar cell tumour of the sellar region , and spindle cell oncocytoma
Pftuicytoma Spindle cell oncocytoma
Granular cell tumour

Essential: Essential:
spindle cell neoplasm in sheets and short Neoplasm composed of polygonal cells with granular Spindled or eplthelioid tumour with eosinophilic.
s cytoplasm granular cytoplasm
AND AND
Sellar or suprasellar location Sellar or suprasellar location
AND AND
TTF1 expression Nuclear ITF1 expression
Nuclear TIF1 expression
AND AND
~of pituitary hormone and transcription factor Absence of pituitary hormone and transcription factor Absence of pituitary hormone and transcription factor
"91Dras5:ion
expression expression
AND AND
Absence of neuronal and neuroendocrine marker Absence of neuronal and neuroendocrine marker Absence of neuronal and neuroendocrine marker
·on expression
expression

Desinlble: Desirable: Desirable:


Absence of interspersed reticulin fibres Absence of interspersed reticulin fibres Absence of interspersed reticulin fibres
PAS-positive/diastase-resistant Antimitochondrial antigen immunoreactiv1ty
CD68 or a1-antitrypsin immunoreactivity
m 1. ttlyroid transcription factor 1.

· ormones and transcription factors are absent. Other positive morphology, sharing glial and meningioma-like features (3322).
narkers include BCL2, CD44, nestin , and aB-crystallin {2094, Granular cell tumour cytological preparations are characterized
70} Cytokeratins, CD34, and markers of skeletal and smooth by the uniform appearance of the polygonal cells. which have
rruscle differentiation are absent. Phosphorylated ERK, AKT, round to ovoid nuclei and abundant eosinophilic granular cyto-
and S6 expression has been reported {70,2114} . Expression of plasm dispersed in a granular backgrou nd (2530) .
~STRs and DRD2 has also been documented {2995,1881).
Spindle cell oncocytomas usually show low proliferation . Diagnostic molecular pathology
.'totic activity is rarely reported in studies; when documented , No specific molecular test results are used in the diagnosis .
ses are usually limited to a few. The reported Ki-67 label-
mg index ranges from < 1% to 17%, although reports of values Essential and desirable diagnostic criteria
> 5% are few {1098}. See Box 12.03 .
Hallmark ultrastructural features include an increased num-
ber of often abnormal mitochondria, intermediate filaments , and Staging
~el l -to-cell junctions including well-formed desmosomes and Not applicable
1
1termediate-type junctions {2718 ,1174). A few cases may show
sparse small neurosecretory granules (667,2094 ,619,998) . Fol - Prognosis and prediction
licular structures {619) and intracytoplasmic lumina with micro- Pituicytoma, granular cell tumour, and spindle cell oncocyLoma
Ytilous projections are reported (3261 ,2167). are typically benign, slow-growing tumours, curable by gross
total surgical excision. However, there seems to be a "11gt12r
Cytology frequency of recurrence in spindle cell oncocyton13s rh~1n 1"
In cytological preparations , pituicytomas can display fibril- the other tum urs 11663,2004,324 1098,5U3 16581 f\IJ.l.~r1dnr
1ary to fine and wispy cytoplasm with occasional spindled cell transformation and distant met~lstase, t1ave lot b8en , eµ1..11 t<2Ll

/_, ' '-" ' JI


Pituitary adenoma I Lopes MBS
Asa SL
pituitary neuroendocrine tumour K leinsr::hmirJt-ueMast8rs BV
MeteO
Osarnura R'f
Villa C

Definition
Pituitary adenoma I pituitary neuroendocrine tumour (PitNET) is
a clonal neoplastic proliferation of anterior pituitary hormone-
producing cells .

ICD-0 coding
8272/3 Pituitary adenoma I pituitary neuroendocrine tumour
(Pit NET)

ICD-11 coding
2F37.Y & XH94UO Other specified benign neoplasm of endo-
crine glands & Pituitary adenoma, NOS
2F9A & XH94UO Neoplasms of unknown behaviour of endo-
crine glands & Pituitary adenoma, NOS Fig. 12.17 Pituitary adenoma I pituitary neuroendocrine tumour IP·t1 ET . A Cor-
cotroph adenoma/tumour. T1-weighted postcontrast coronal MRI demons rates-~
Related terminology classic appearance of a homogeneously hypoenhancing pituitary •esl!Jn : crrca'-
ible with corticotroph microadenoma. 8 Invasive adenomai tumour. T' -we·q.·~
Acceptable: PitNET; pituitary adenoma.
postcontrast coronal MRI demonstrates a very large heterogeneously 9r ·ar-':' -g
tumour centred in the sellar region with suprasellar and parasella1 ex ~el"s•or ar::
Subtype(s) invasion of the sphenoid sinus and clivus. Both internal carotid artenes are ~O'"'­
The types and subtypes of pituitary adenomas I PitNETs are pletely encased.
described in Table 12.01.
tumours tend to be hypointense, whereas sparsely grar.u 1ate-.J
Localization tumours are hyperintense {1278,2545) .
These tumours are usually identified in the sellar region, often
with suprasellar extension, but ectopic locations include the Epidemiology
sphenoid sinus , and rare clival and suprasellar tumours have Pituitary adenomas I PitNETs are identified incidentally 1n u,o 10
also been described {28) . Rarely, pituitary adenomas I PitNETs 20% of the population {882}. Clinically diagnosed tumours were
may arise in teratomas (56,161 ,3305) . once considered rare; however, recent population studies reocr:
a prevalence of 78-116 cases per 100 000 population 167 909
Clinical features 36,955). The Central Brain Tumor Registry of the Urn ed Sta:e-
Pituitary adenomas I PitNETs have a spectrum of clinical fea- (CBTRUS) reports that pituitary adenomas I PitNETs ac n
tures. They may be small, slow-growing, and found incidentally, for 16.5% of brain tumours , with an incidence of 3.94 cases p-er
or they may give rise to hormone excess syndromes, including 100 000 person-years {2347}; however. this database reoons
hyperprolactinaemia, acromegaly/gigantism, Cushing disease, surgically resected tumours and does not include those rea!eO
or hyperthyroidism {2089,1133). Large tumours may cause with medical therapy alone.
symptoms of an intracranial mass (e.g. headache, visual field The incidence of pituitary adenoma I PitNET increases 1!"
disturbances) and cause hypopituitarism (2089}. Some tumours age. Approximately 5% of patients are diagnosed before :r--
invade downwards and appear as a nasal or paranasal mass age of 20 years {1541 ,2168). Pituitary adenomas I P1r ETs
{1392). Occasional examples undergo acute haemorrhagic occur equally in both sexes , although some studies sho an
necrosis , resul ting in rapid expansion and causing a clinical overall female predominance of certain subtypes {36 ?.3-f"'
presentation termed "pituitary apoplexy" {1543). characterized 2090) . Cushing disease and prolactin-secreting tumours 3l
by severe headache, lethargy, and signs of increased intracra- more common in female patients (909}. whereas n n-rw ~­
nial pressure. tioning {909} and lactotroph tumours are more often s rgic.ill
resected in male patients {2090}.
Imaging
MRI with and without gadolinium is used to identify the sellar/ Etiology
suprasellar mass and to characterize size, optic chiasm com- Risk factors
pression , cavernous sinus and/or sphenoid sinus invasion, Risk factors for pituitary adenoma / P1tNET related m d '-
haemorrhage , or cystic changes. Most lesions are hypointense sure or lifestyle have not been definitively identi fied. Pr 1m1-
on T1 -weighted images and show variable gadolinium enhance- nary studies showed that environmental pollutants 1nflu~nce
ment 1771 ). On T2-weighted images, densely granulated the biological behaviour of somatotroph adenoma 'itun.<Jurs
and indu ce. proliferation
, \ '' 0 . in normal pituitary ce ll s aft er common recurrent somatic mutations that drive tumorigenes1s
•\; term 1ncu b a tton 111 vitro 11814,1141 ,2480,454 3624 3135 affect GNAS in as many as 40% of somatotroph adenomas/
. "4S .2599) . ' ' • tumours and lead to hormone hypersecret1on via upregula-
E ablished carcinogenic agents like X- and y-radiation do not tion of the cAMP/PKA pathway 11800,3000,2501 l USPB and
~ l to play a role in pituitary tumorigenesis 1617). Use of oral USP48 mutations rescue the EGFR (HER1) and CRH/SHH
tracepttves or m.enopa.usal hormone therapy is not signif i- pathways resulting in aberrant ACTH synthesis in approxi-
ly associated with an increase in the risk of tumours (242 mately 50% of corticotroph adenomas/tumours !2642, 1964,
6'13l l
2457,1270 ,897,2458 ,55) . Novel mutations associated with
sporadic tumours include USP48 !2819 ,?38) , NR3C1 (2819 ,
tic factors 1375,121), CABLES1 {1294 ,2736) in cort1cotro~h adenomas/
ary adenoma.s ~ PitNETs are monoclonal proliferations, tumours and TP53 f3133} in pituitary carc inomas Apart
the great maionty occur sporadically 11293). The most from th~se rare events , recurrent molecular alterations have

1.Z.01 Classification of pituitary neuroendocrine tumours (PitNETs) in the upcoming WHO Classification of Tumours volume Endocrine and neuroendocrine tumours 13425A)
Transcrlptlon Keratin Tumour su6types Hormone excess
factor(s) Hormone(s) (CAMS.2 or CK18) (if appllcable) syndromeb

GH, o-subunit Peri nuclear Densely granulated Florid acromegaly


somatotroph tumour
!mph tumours PIT1
GH Fibrous bodies (> 70%) Sparsely granulated Subtle acromegaly
somatotroph tumour
PAL (paranuclear) Weak or negative Sparsely granulated Hyperprolactinaemia<
lactotroph tumour
l.actotroph tumours PIT1, ERa
PAL (diffuse cytoplasmic) Weak or negative Densely granulated Hyperprolactinaemiac
lactotroph tumour
Mammosomatotroph GH (often predominant), Acromegaly and
PIT1, ERa Peri nuclear hyperprolactinaemiac
tumour PAL, a-subunit
Thyrotroph tumour PIT1, GATA2J3d a-subunit, TSH-P Weak or negative Hyperthyroidism
Acromegaly,
PIT1 , ERa, GH (often predominant),
re Pm ·lineage tumour GATA2f3d PAL, o-subunit, TSH-p
Perinuclear hyperprolactinaemiac, and
hyperthyroidism
PRL (predominant), Hyperprolactinaemiacand
AcidophH stem cell tumour PIT1, ERa Scattered fibrous bodies
GH (focal/variable) subclinical acromegaly
Acromegaly,
Immature PIT1 -lineage PIT1 , ERa, GH, PAL, a-subunit, TSH-p Focal/variable hyperprolactinaemia<, and
ll.lmour GATA2J3d
hyperthyroidism
T.PIT.(ineage tumours
Strong Densely granulated Florid Cushing, often
corticotroph tumour microtumour
TPIT (TBX19), ACTH and other POMC Variable Sparsely granulated Subtle Cushing, often
Cortiootroph tumours derivatives corticotroph tumour macrotumour
Neuro01 (P2)
Intense ring-like Crooke cell tumour Variable, Cushing
perinuclear
Sf1-tineage tumours
Hypogonad1sm (virtually
SF1, ERa, o-subunit, FSH-p, LH-13 Variable
Gonadotroph tumour GATA2f3d
all) or hypergonadism
(e cephonall
Tumours without distinct cell lineage
Unclassified plurihormonal Multiple Multiple Variable
tumours combinations combinations V::iriable

Null cell tumour None None Variable

a-subunit, glycoprotein hormones alpha subunit; GH, growth hormone PAL. prolaclln
'Mixed tu~o~rs also occur and can constitute ~ny combination ot tumo~rs s~o~n , the -~ost comrn?ri is in.x_ ll ~om<1t t. 1pn I 1)L1!1oph tu• r our f\l 1y !I., .· i !>pt· C' , 1._,, r,iJr1o•·?.i11'
non-functioning. cModerate hyperprolactmaemia can occur with a1 y sellar mass that has.suµr asellar exter1:,1on, 1merrup 1119 hn>oth 1a,1iic tni ,; d·µ<:1m. t !r• .l .. ~t. ui!t. 1 lrol ,,"1,.
lfle PAL level rarely exceeds 150 ng/mL; lactotroph tumours usually show a c;haractenstic correlation between tu1now '"Le and PRL lev,)I::,, w f.:lff'u· lt• ,., •
do not <lGATA2 and GATA3 are paralogues and show cross-reac1i111ty with some available ant1sera. ~ • ·

• j·-
not been identified in sporadi c tumou rs. Ins tead , epigene tic Pathogenesis
alterations may be relevant to tumori genesis in th e majority of Cell of origin
sporadic cases (3141 ,88 3,2 68 ,1267,88 4,36 13). Chromoso m al Pituitary adenomas I PitNETs ar13 r,ons•rJc;ri::<:J •r, r~~C/<:-,c-:r .=-..
alterations are common , whi c h is unu sual for tumours with clonal neoplastic proliferation (Jf h0 rrnon8-prr1rJu<:.. .-,,. - •.• -'"' ·
largely indolent behaviour 1267) . docrine anterior pituitary cells. , ·
A minority of tumours are as sociated with known famil ial
predisposition syndromes , implicating specific germline muta- Somatic genetic alterations
tions in the development of pituitary adenomas I PitNETs (see See Genetic factors in the Etiology subs8c.t1r,n ::ir:..r, ,~
Table 12.02) /3038 ,3013 ,3289} .

mRHA elustwr r,,,,.,,,


• t1
12
s 11),,,..,
•f.2:f)~
t3 • 2 ~,.,,..
I~ >"')....,.
tS
• t6
",,.,,,
.
·=
!"r1
WHO 2917 Tum<N lype • 2
")
I • Lacio
• h
Thyrolroph c - im-.
• Somatoeroph No
• NUkeft • Oa.tirllJ
I f'1untlofmooal P IT-1 • Y111
I • Mixed GH-PRl
Splwroid ,,..,..,,,

NMF signatures
I Granul.rlons No
• SpatSe • Doi.alJI
Rank4 I • Dense • Yes
Rank 1
Rank3 s~ ~
p......,,,
Rank2
1 - ~~lil'IJ
Rank6 · ~
· ~
Rank5 1

• =""~~
Acromegaly · ~
Cell composition
I •• Mixed
Silent
~
• Yes
Gonadotroph No
Corticotroph Su
Somatotroph Female
• Male
Lactotroph
L Thyrotroph

NKX2-2
ARX
0 Cortlcotroph
silent
Gonadotroph

HOXBS
DMRT2
LIN28B
MNXl
SOXll
SOX2
FOXL2
MKX
I DMRTA2
ZMAT4
PROMS
ZNFS36
RFX4
soxs ACTH
ESRl
LHX3
KCNIP3
TSHZ2
ISU
HMGA2
RAX2
ZBED2
ZNF804A TPIT
NEURODl
ASCLl
LMXlA
CUX2
DMRTAl
ZNF750
HIF3A
IRX2
ST18 SF1
Qll~~~:re tl!lllt:lllil• NEUROD4
~~rn~1
TBX19
PLAGLl
RORB
AR
PITX2 GATA3
,.._. .... ,,..~ ........ RXRG
~~m
ZFPM2
ZNF3858 P114 P054 8
FEV
E I I ~~t~~
Fig. 12.18 Transcriptome of pituitary neuroendocrine tumours (PltNETs). A Unsupervised classification of PitNETs identifies six main groups, corresponding lo corttcouoph
overt Cushing (t1 ). lactotroph (t2), silent corticotroph (t3), gonadotroph (t4), thyrotroph (t5), and somatotroph (t6) PitNETs. Pathological and clinical annotations are provided. Th¥
associati~n with transcriptome groups is detailed (p (x2) : chi -squared P values). B Heat map of the six non-negative matrix factorization (NMF) ranks used for generatlflg me
unsuper~i~ed cla~sification: C Proportion of gonadotroph, corticotroph, somatot roph, lactotroph, and thyrotroph canonical signatures in each PitNET. D Magrnficattan (20 l ..,f
H&E st.aimng and 1mmunoh1s~ochemistry for the corticotroph·related markers ACTH and TPIT and the gonadotroph-related markers SF1 and GATA3 performed on tissue socuons
of corhco'.r?ph of o~ert Cu.shing (P114), silent corticotroph (P054), and gonadotroph (P098) PitNETs. Scale bars represent 100 mm. E Expression profiles related to the top 50
most s1gmhcantly d1fferent1ally expressed transcription factors among the six transcriptome groups.

408 Tumours of tl1e sellar region


~ 12.02 Inherited genetic susceptibility to pituitary tumours
~~~--~~~--~~~-

Gene(s) Pituitary feslons Un order of frequency)

I isolated p1tuitary adenoma Somatotroph, lactotroph, rnammosomatotroph, corticotroph, and other tumours
AIP or unknown
Rarely, somatotroph hyperplasia
-finked acrogigantism GPR101• Mammosomatotroph adenomas and/or hyperplasia
associated with pituitary tumours

Lactotroph, non-functioning, somatotroph, and corticotroph tumours


Multiple endocrine neoplasia type 1 MEN1 Multiple or plurihormonal tumours
Somatotroph or mammosomatotroph hyperplasia
pie endocrine neoplasia type 4 CDKN1B Somatotroph, non-functioning, and corticotroph tumours

Camey complex Somatotroph, lactotroph, and corticotroph tumours


PRKAR1A
Mammosomatotroph or somatotroph hyperplasia

McCune-Albright syndrome Mammosomatotroph and somatotroph tumours


GNAS (mosaic)•
Mammosomatotroph or somatotroph hyperplasia
F~lial paraganglioma, phaeochromocytoma,
pituitary adenoma syndrome SDHA, SDHB, SOHC, SDHO Lactotroph, somatotroph, gonadotroph, and (rarely) corticotroph tumours

DICER1 syndrome Pituitary blastoma (majority ACTH-secreting; rarely, growth hormone-secreting


DICER1
and prolactin-secreting)

Neuroflbromatosis type 1 Corticotroph and somatotroph adenomas


NF1
Pituitary duplication
lynch syndrome MSH2, MSH6, MLH1, PMS2 Corticotroph and lactotroph tumours
USPB-related syndrome USPB' Corticotroph tumours
Tobemus sclerosis TSC1, TSC2 Corticotroph tumours
~n can also be somatic.

Gene expression Histopathology


A recent transcriptome-based classification identified distinct Pituitary adenomas I PitNETs are generally monomorphic , with
molecular subtypes of pituitary tumours, each associated with cells arranged in a variety of histolog ical patterns , including
specific secretion phenotypes, genetic alterations , and epige- diffuse, papillary, and trabecular arrangements . Cytologically,
11et1c profiles {2 232). In this classification, clinically aggressive tumour cells may be acidophilic , basoph ilic, or chromophobic ;
tumours did not appear as a distinct molecular entity. however, these tinctorial characteristics are nonspecific . Cells
may show densely or sparsely granulated cytoplasm according
Protein expression to the tumour subtype (see Histopathological subtypes, below).
A. limited number of transcription factors currently bridge the Nuclei tend to be bland with regularly distributed chromatin.
differing types of pituitary tumours and lineages. However, Mitotic activity is generally low. The presence of extensive cel-
recent multiomics and protein expression profiling studies lular pleomorphism, brisk mitotic activity, and necrosis 1s not
'lave shown less rigid pituitary lineage {2232,2670,635). For typical and should prompt consideration of alternative diagno-
1nstance , silent corticotroph tumours display both corticotroph
ses. Calcification or ossification is rare , except in lactotroph and
and gonadotroph signatures, suggesting a transdifferentiation thyrotroph subtypes .
state 12232,2092,635 ,2670) . In addition , SF1 is expressed
,., a subset of somatotroph tumours (mainly GNAS-wildtype Histopathological subtype::.
tumours) {2232) . Pituitary adenomas I P1tNET retied their derivation from six cel l
types (see Fig . 12.19, p. 410) with each li11ea.ge d1tferent1.Jt1ng
Macroscopic appearance into mulriple ~~ubtypes. Some are 1.. orn~"\m>ed of lass ditferenl1-
At autopsy, pituitary adenomas / PitNETs range from small pale ated cells
'.'IOdules within the gland to large cohesive hyperaemic masses
wrth pushing borders. Variable extension into the suprasellar 7j >i I ' l. , I .' , 11 I ;I

space and invasion into the sphenoid and/or cavernous sinus Ttw <:> 11bty1:...1<>. 1J\ltt11r; '' ,,
r111 <IC' \II\
are frequent; dura mater and bone inva sion may be seen fl 18) Cdfl l [' L ti• I dli I f I( 'If r 1t J '1... t-
Tumours with apoplexy may show haemorrt 1ag1r. nE-cr%is ahly r1r•·,;··~c; tiu,' 'l Ttl!\lf
I I·

I: j • 1 I > I ) ~ t
Undiff~~enttated pitult~ry J ? / Null cl'll
rumour
neuroendocrine cell

I TPIT lineage I
~--- ... · Poo: -
[ S£_1
I
llM~ge
.' dlffere11t1ated
.Y ' . PITl -linPage
I Cortlcotrophs I PIT! - lineage ~n~d~p~~
_____ .. ------ .. :
. ' _-_":, ______ _

. • · · . Demely granulated : .... : Sparsely granulated .. • · ·


precursor cell . · - · Mature
· ,.: . plurihonno nal

'. PITl-nneage
corticotro ph tumour '. ~· · 'corticotroph tumour .. :
'·.
- - -· -
- . - . ··- - - - - - - - . - -
- - - - .. - - - - - - - - . • .. t_u_rn~u_r . . -

Crooke cell
corticotroph tumour .. ·

somatotroph
• · -· •~-~~u_r_ -. '' -~cid~phil · . __...
. . . . . A: ::~~ ..... - - --~- .. - - ;
stem cell •
Thyrotroph
- - - liensely granulated - · · - •.. · · · Sparsely granulated Lactotrophs . _ . .. .... t~o_u_r _ .··
!Um<J1Jr
somatotroph . ; . • somatotroph tumour . - ;
__ ~u-~~u_r __ . _ · · ._ _ .. · .· ._
. - .. - ·. -. -_ ~ ·_ -. - - - - _,( •. - - - • • ••. ~ '!Ii. - . - - - - - - . - - -
.·• Densely granulated · •. . • Sparsely granulated · ·.
'.. lactotroph tumour _.' '.. . lactotroph tumour _· ·
·. . _ --- ·----- -------

Fig. 12.19 Pituitary neuroendocrine tumour (PitNET). Classification of PitNETs reflects derivation from six adenohypophyseal cell lypes of th ree lineages. W'th ~.
of some cell types and some tumours showing incomplete differentiation.

Densely granulated corticotroph adenomas/tumours are positivity and weak cytop lasmic ACTH react1 y ou: ,....~p~
strongly basophilic and PAS-positive , and they have intense cytokeratin reactivity (2090! . They are usually large 1 ~~ ._
cytoplasmic ACTH and cytokeratin reactivity. They are usually subtle clinical features of Cushing disease !52.26251
small and associated with florid Cushing disease, and they tend Crooke cell adenomas/tumours show Crooke ~yahrie ::
to harbour USPB mutations (1270) . (required in > 50% of tumour cells for the diagnosis) ·:
Sparsely granulated corticotroph adenomas/tumours are ing of abundant cytop lasmic accumulation of pate c dop "' 1

chromophobic or weakly basophilic, with faint or focal PAS hyaline material with focal basophilia. PAS and ACTH

F •
Fig. • Densely and sparsely granulated cori1cotroph adenomas/tumours. Densely granulated corticotroph tumours are composed of tumour .;tills w1tn ac:"~ ~ - . ·
12 20
granular cytoplasm (A) whereas sparsely g I t d 1· t h · · ··
t·ive 1or TPIT (C}, an d th,ey tend to display
. ran u a e cor ico rop tumours display a lightly basoph1l1c appearance (8). Irrespective of their subtypes. thtise tt:th.>i..1s .ll~ _x_,
diffuse cytopl · · t. t 1 · ·
densely granulated tumours are diffuse! . .. CAMS.2) (D). Consistent with their cycop1asm1( 1Jrai 11..1a111r l.}:i1:.
asmic ieac ivity or ow -molecular-weight cytokerat1n (_
identified when using PAS h·istoch . Yposiltve for ACTH (E), whereas sparsely granulated tumours are variably and weakly positive (f ). The same pa11ern 011~J 'J> 1t .-u ...
em1s 1ry (n ot shown).

410 lurnours of ti1e se!lar rc.;g 1u 11


Rg.12.21 Somatotroph adenomas/tumours. A-C A densely granulated somatotroph tumour showing eosinophilic, granular cytoplasm and a central nucleus with a prominent
nucleolus (A), as well as intense and diffuse growth hormone expression (B); cytokeratin (CAM5.2) staining reveals perinuclear deposition with very few fibrous bodies (C). D-F A
sparsely granulated somatotroph tumour with slightly eosinophilic cytoplasm and showing paranuclear fibrous bodies (D), as well as focal growth hormone expression in the
cytoplasm (E); cytokeratin (CAM5.2) staining highlights a large proportion (> 70%) of fibrous bodies (F).

in juxtanuclear or perimembranous distributions, and intense body pattern is the hallmark of the sp arsely granulated sub-
cytokeratin staining in a concentric pattern (1058,749 ,2089] . type , whereas the densely g ran ulated subtype has perinuclear
These are usually large, invasive tumours and about 76% occur cytokeratin and may have focal fi b rous bodies (2295) . The
with Cushing disease (749) . distinction is cl inically critical because the two subtypes have
different treatment responses [145,49]. Densely granulated
1 gonadotroph adenomas/tumours adenomas/tumours occur in about 30-50% of patients with
'Ttlese show a spectrum of morphologies and degrees of differ- acromegaly, whereas sparse ly g ranulated adenomas/tumours
entiation , from trabecular proliferations of elongated cells with account for about 15- 35% of acromegaly cases (2295,49,17361.
basal nuclei and prominent pseudorosettes to solid sheets of Densely granulated somatotroph adenomas/tumours are aci-
round cells . They can show prominent oncocytic change . They dophilic , with strong GH staining dispersed diffusely throughout
have a range of gonadotropin hormone expression: most often the entire tumou r; glycoprotein hormones a-subunit is typically
focal FSH, with scant LH and variable a-subunit immunoreac- expressed \2090).
tivity. About 40% are negative for cytokeratins \2090) . They Sparsely granulated somatotroph adenomas/tumours are
are characterized by variable nuclear expression of SF1 (142). less acid ophi lic than their densely granulated counterparts.
GATA3 12092,32431. and focal ER (3568,2924) . Hormone-neg - They often show cytoplasmic clearing due to fibrous bodies,
ative gonadotroph tumours with SF1 and GATA3 positivity alon e and eccentric nuclei that are often bllobed or contorted around
are the least differentiated and must be distinguished from null th e fi brous body GH expression 1s focal in the cytoplasm. and
cell tumours 12262,83 ,185,1210}. a-subunit expression is negative \2803,20901
Lactotroph adenomas/tumours are generally sparsely
P!Tt -lineage adenornas/rumuurs granulated with juxtanucle::lr l1orrno11e positivity in the Golg1
This is the most complex group . It comprises a rang e of tumours, complex; rare densely granulated ldctouoph tumours h3ve dif-
from those composed of a single c ell pop ulati on with the ability fuse cytoplasmic hormone st 11n:ng I'>U901 The turr1L1urs orten
to secrete one or more of the PIT1 -lineage hormones (growth sh w intense nurlec1r ~n sta1n1ng (3t:HJ8.C)t36l anci 3tJse11ce or
hormone [GH], prolactin , and TSH) to tu mours consisting of two u-subunit Spdr ~t.>ly gr anuldecl l,rti:'lrL)ph tu1 llL)ur s otlen show
types of cells with bihormonal or pl urihormonal secretory ab1li hyper µrola ' t111n rt111.1 tt 1r1t 1s f. ropur !1(111,11 to tht-~ir t•Jrrll.)ur s1L8
ties (147,985) . rtwrotwptl ~ 1tfunorr:d~ ·cw n ·)ut 'S di , 11s , illv kif )e ,1'1L1 -om·
Somatotroph adenomas/tumours that usually ,auso aero posed ot rolrn1veiy inurh..)rnurp!1,c, pli\t r1.:-:,I l'•"'ls 1,\icrl smne
megaly or gigantism are ch aracterized as densely or ~µar',f)ly LJPgree ur 11ucl!t1r pl<'L>rro'-1q1• 1,, 111 11,\J tui·i ·urs e pr~"S:'- TSH
granulated based on GH immu noexpre~.s1on <.md · ·flt)~1~1 attn drld u sut1un1t, ,111ci L:O't, 11 i~,-~ PIT! 11 i 'AC..li ~ 'J )II(: )1)291
pattern \3505) . The presence of a predoni111ant \ » -ou 1 t1L11(iu~ fl10y ri1c1y l1c.lvu tril ·risti _;t ru11 1;:tl ribrc ,-; I bUtil llkl ~a, ,1 , __ 1r,, •1

Turr,o · r~ <'I tr
I '.'t · ' '' ' It' 1 1/ 111 !11 ' I "• . c if f '1 l .f l•r )t • l(Jt '
1
lmmat11m fJI T1 ·ltnP.fl(j(J AdP.n r;ma1!tumo11rr f!r!rri ' r,r•- ~
1 his 1s a n mpl f mily, ch Ar Acte rJJ e ci AS follows known .::i s "silen t su t""Jt 1r;'9 '3 ~cJAr1rJm.: i · ;=jr1rJ ?.tt1 rc::?f~,·r-ir[ , , , • ~
/\ 1.m11 iosPma fL1froph AdenomAs/ tumour s Rre composed of a 2017 WHO c lass1ficat1on o f 13ndor,.rir1P. hJffl f )I; ( ) ~Cj 'pl1;r r.rirrrr
111 n L)m rphi . .II por ulation with eosinophili c cell cytopla sm nal PIT1 -posi t1 ve adenoma/tumour · fhe'>P. turr,r, w·~ ~r~ 0 . ; .
!90 .1'148) thr-i l co pr .sses GH , a-subunit prolact1n . and ER . posed of cells that do not show te r m1n~ I rJrff~ r~r.t 1 8 • r;r •, ,.,r.~
n1 often c use acrornegaly/g1gantism and hyperprolactinae- of the well -known PIT1 lineage anterior p1tu1tar / r.:~111 rr.~ 1 arr.)
mia. and th y are more common in the paediatric age group usually chromophobic, rather than ac• doph1l1c 1ri.-e 'T'ICJ'" d f~r
and oung r adults than in older populations . entiated tumours. They always express nuclear PIT l 12or~ ' ar1
A-11\ed somatotroph - la ctotroph adenomas/tumours are com- they may express focal ER and/or GATA3 120921 P1et nia r,~
1= osed of lwo discrete tumour ce ll populations , typically with immunonegative for hormones . but more nften they ari:; fcza •.1
one e pressing GH and the other expressing prolactin (see positive for one or more of GH . prolactin TSH. and ri~>•.JbJ
Table 12.01 , p. 407) . Many combinations of sparsely and/or 1855,2091 ,2090) . Cytokeratin patterns are variable and ' h~~
densely granulated somatotroph and lactotroph cells have been may be occasional fibrous bodies 12091.20901 The«>e turr01..1r:
described (2090) . Patien ts may present with acromegaly/gigan- are frequently clinically silent, but they may be assoc1 at~d 1trt
tism or with hyperprolactinaemia . acromegaly/gigantism , hyperprolact1naem1a. and/or yp~ ~­
Acidophilic stem cell adenomas/tumours are rare ; they most roidism 12091 ,855.1351 ). They tend to be aggressive and · .-a-
closely resemble lactotrophs but may express scant GH and be sive, with an increased risk of recurrence 12091 .8551.
associated with fugitive acromegaly (1349,1350). They are usu-
ally oncocytic , but they may have abundant clear to vacuolated Unc lassified p lunhormonal adenomas, t11r7101_, , ~
cytoplasm due to mitochondrial dilatation (1350) . Tumours pre- These are extremely rare, with only indivrdual case repo' s
dominantly express prolactin with variable intensity, and they published . Tumours show differentiation across more than OPg
often express focal to variable GH (1350 ,1349,2090). Staining lineage, expressing several comb inations of hormones (e.g
for cytokeratin highlights small, scattered fibrous bodies in GH/ACTH , prolactin/ACTH , and LH/ACTH) and correspond ng
about two thirds of these tumours (2090) . transcription factors (e.g. PIT1 /TPIT. PIT1/SF1 , TPIT/SF1) (1987
GH-producing plurihormonal adenomas/tumours are also 2802 ,2621 ,3217,24561. Multiple synchronous tumours of d --
rare; they may arise in the setting of acromegaly/gigantism , tinct lineages should not be confused for plurihormonal tumours
hyperprolactinaemia, and in some cases synchronous hyper- 12088}.
thyroidism. These are monomorphic eosinophilic tumours that
express variable amounts of TSH in addition to GH, prolactin, Null cell adenomas/tumours
and a-subunit (2090). GATA3 expression correlates with the These are anterior pituitary tumours that show no 1mmunohrsto-
TSH staining pattern (2092}. Intense acidophilia and abundant chemical expression of biomarkers of known anterior pituitary
GH and prolactin immunoexpression distinguish these tumours cell lineages . They are usually positive for chromogranin A and
from the immature PIT1-lineage pituitary adenomas I PitNETs cytokeratins but must be negative for pituitary transcription fac-
that may be plurihormonal (2091). tors and hormones. Recently, some cases expressing focal

Fig. 12.22 Immature PITHineage tumour. This tumour from a patient with hyperthyroidism 1s composed of pale ac1doph1tic cells with variable morphology, including polygonal
cells that resemble thyrotrophs. They have prominent nuclear inclusions that resemble irregular nucleoli (inset); these have been called spheridia. This tumour expressed nucl r
PIT1 with variable growth hormone (GH) and TSH but no prolactin (PRL); there is diffuse a-subunit (aSU) positivity. Tumours of this lineage have variable pos1t1Vtt~ for tne thretl
PIT1 -lineage hormones.
glycoprotein hormones a-subunit have been reported {2090). metastatic carcinomas {2092]. Pri mary sellar paragangl1omas
They represent < 5% of surgically resected pituitary tumours {1238 ,144] are distinguish ed from cytokeratin-negative p1tu1tary
12090,2262,83,185,1210}, and they may have a high incidence adenomas I PitNETs by their positivity for GATA3 (without SF1
of recurrence and cavernous sinus invasion {83 ,1210,185,2262] .

Grading
There is no formal grading system .

Invasion
Pituitary tumours may invade the surrounding tissues , includ -
ing the sphenoid and cavernous sinus, dura mater, and bone.
Invasion of the cavernous sinus prevents gross total tumour sur-
gical resection . MRI evaluation using the Knosp scale and its
modificat ions may predict the degree of invasion /1668,2107] .
Tumour invasion is closely related to tumour size, with about
55% of tumours > 10 mm showing histological dural invasion,
compared with about 24% of tumours < 10 mm (2063] . Non -
functioning tumours have a higher incidence of invasion than
functioning tumours {2063 ,3229] .

D;fferential diagnosis
Tumour metastases to the sell ar region , such as ca rcinomas
tespecrally breast and lung) and neuroendocnne tumours, can
·-·-
Flg. 12.24 Mixed ganglioc,tr 1ma-sorn<l!Ot1oph adenoma !umour .'\ ni1no11 ty
...... Jt p.1
t1ents with ctc101ne:,J?.ly may hJve a n11xe(i g:rng11ocyt011 a s01n<1t0tropn .iae110111,1
mimic pituitary adenomas / P1tNETs . Metastases ar8 irrir11u- ll.lm'.:IUI f"1~ IW•. Ceduiar >''t'1IH:'ll!S C~!1 Dd '><Jtlrl 111le1'1ll!\ed QI 111 1SOiJt.:>O ,Ht:' h. l~ Jll
nonegative for pituitary transcription factors. with the ~..:H.c-p ~1! 1 n'l 1 C' ''
1 11
·" 1'· '
1
n "ll.~l.k' ,1111 ;:;1.t.' S(> llo' wirh Jense N1ssl substance a111J Ilk\ :lr.:-
tion of ER . GATA3 , and SF1 . which can be ex presc;ec1 1n ·' ' k L d11 ,)f 1\ .. • 1'
1 lJ ~ '.J 1 • .._ ! _·;• do\~ llln{'IX

. '
Box 12.04 Diagnostic criteria for pituitary adenoma I pituitary neuroendocrine tumour (MRI and CT) and/or funr:tional (F0(; PF T ;:irirJfrir '/-J r':".J ;:. ..
(PitNET) imaging has been recommended 128211 -
Essential:
Prognosis and prediction
Sellar or suprasellar location
The 2017 WHO classification of tumours (jf ~nrJr;c.nr.~ rr,. ;..-.
AND categorized pituitary neuroendocrine tum0ur~ 3 , ~ ~,'J
,.J ~ ~

Histological features of a low-grade neuroendocrine tumour that displays adenomas and carcinomas" f 19191 . Notably. h'1 r~rrr· ·~ - 1 ,_
destruction of the normal anterior gland acinar structure cal adenoma" (defined in the 2004 WHO class.f·r:.~· .;101 ,,::.-
AND abandoned , because a definite correlation be wc:ien r,i ~i-::.. r,r; ...:;::
Subclassification based on immunoreactlvity for pituitary hormones and/or lineage- diagnosis and clinical behaviour could not be estat,lt:-,herJ ·- ~:.
specific transcription factors 2017 volume did not introduce a new tumour grad1rig s·6 :~,,.r­
but it emphasized the identification of h1gh-ns~ ad~ri:;F 3 -, ".J!
Desirable: recogn izin g (1) tumou rs with increased prolifera ion £ev3 k:'o/J
Reticulin fibre disruption by mitotic count and Ki-67 labelling index). whrch me:;· , ... c;.1
Low-molecular-weight cytokeratins, in particular for somatotroph and corticotroph relates to tumour recurrence 12050 .32281; and (2) the fo,1 0 1•
tumours ing five tumour subtypes that have an increased propers, :i:,r
Tumour proliferation as indicated by either mitotic count or Ki-67 (MIB1) early recurrence and resistance to treatment· sparsely g;;j. 'J·
expression lated somatotroph adenomas/tumours, silent corticot oph ade-
nomas/tumours , Crooke cell adenomas/tumours. plunnorMcr~
PIT1-positive adenomas/tumours . and lactotroph adenof""ias/
and PIT1) and tyrosine hydroxylase (144}. Sinonasal neuroendo- tumours in men 11919).
crine tumour and olfactory neuroblastoma can also involve the Pituitary carcinomas correspond to O 12-0.4% of all turro ::
sellar reg ion ; these do not express pituitary hormones or tran- {2775,86). They are characterized by craniospinal d1ssem1na-
scription factors . Spindle cell oncocytoma should be excluded tion and/or systemic metastases. The most common subtypes
in the differential diagnosis of pituitary adenoma I PitNET with are lactotroph and corticotroph carcinomas 12598.2461 .1 275'.
oncocytic features ; the former uniformly stain for thyroid tran- However, predictive markers of malignant progression are not
scription factor 1 (TTF1) {1829,2094}. The rare sellar neurocy- well established and no single histopathological marker ~as
tomas are negative for cytokeratins but express neurofilaments, been shown to reliably predict pituitary tumour behaviour.
TF1 , and hypothalamic hormones (143,146} . Pituitary hyper- Many pituitary adenomas I PitNETs are non-1nvas1ve and~
,Jasia should be distinguished using reticulin staining; reticulin exhibit expansive growth in the sellar region and surroundir.g
shows complete breakdown in tumours, whereas in hyperplasia tissue. A substantial subset (30-65% {2063.3220.3229 2483\l
the acini are expanded but intact. are invasive , with correspond ing residual tumour 12063.3565)
and regrowth after surgery (31201 . Therefore. a cons1oeraoia
Cytology number of patients with pituitary adenoma I PitNET reqwre chn..
At low magnification, pituitary adenomas I PitNETs are distin- cal surveillance and/or adjuvant treatment.
guished from normal anterior pituitary gland in cytological prepa- A subset of pituitary adenomas I PitNETs may display aggres-
rations by their hypercellularity and homogeneous appearance. sive clinical behaviour and rapid growth , with early and mu;:-pe
Because pituitary adenomas I PitNETs have less stroma than recurrences despite multimodal therapy 126241. The prevalence
normal tissue , their cells do not aggregate in large numbers; of these clinically aggressive pituitary adenomas I PitNETs 1s
rather, they are seen predominantly as individual cells and small unknown , probably because they are incons1scently defined
clusters . Tumour cells tend to have enlarged, pleomorphic, or {2624,3573,526 ,750 ,7231 . Pituitary tumour proliferation aiooe
atypical nuclei with variable amounts of cytoplasm and embed- does not always correlate with clinical behaviour 11167}; hcw-
ded in a granular background. Papillary formations are easily ever, the correlation of proliferation and radiological e'J1dence or
recognized on cytological touch or smear preparations . invasive growth appears to have prognostic value. and 1t 1de -
tifies tumours with aggressive potential 13229.150,1854.26'"3
Diagnostic molecular pathology 3228,545) .
Pituitary adenomas I PitNETs do not yet have specific molecular Tumour biomarkers may be relevant for guiding trearment
characteristics that are applied to routine diagnostic algorithms of pituitary adenomas / PitNETs . SSTR expression le'Vef m3Y
(see Prognosis and prediction, below, for prognostic biomark- pred ict the response to treatment with somatostatin a1 .a-
ers). logues {2140,3230,1393.16401. and a low expression of ER in
lactotroph tumours may predict resistance to dopamine agc-
Essential and desirable diagnostic criteria nists (729) . DR02 expression in gonadotroph tumours and 111
See Box 12.04 .
GNAS-mutant somatotroph tumours suggests potentiall"y ne....,
indications for dopamine treatment 1252 1,35831. MGMT pro 111
Staging
expression appears to be negatively correlat8d w1tn respl.)!1:io
Craniospin~I .spread may be observed at progression/transfor- to temozolomide {2623,20501 . In distinction fr m IDH-v. 1ldl)."'tJ
mation of p.1tu1tary_adenomas I PitN ETs 12300 ,1248 1; by definition, glioblastoma , MGMT promoter methylar1on status 1n p1tu1t "'
cerebrospinal flu1.d spread is indicative of pituitary carc inoma . adenomas I PitNET is not related to tumour r~sponse l 4 ~4i .
Cerebrosp1nal fluid cytology may be helpful for the detection of
Finally, losses ot MSH2 and MSH6 e press1on hav~ baen 11111<-e··
meningeal spread 112481. For monitoring progression, struct ural
to temozo lomide resistance 12391 .
414 Tumours of tl1e sellar regirni
/1tultary blastorna Jarzembowski JA
de Kock L
Mete 0
Rotondo F
Schultz KAP

Definition
Clinical features
..uitary blastoma is an embryonal neoplasm of the sellar Cushing syndrome is on e of the most common presenting
•eg1on, composed of primitive blastemal cells , neuroendocrine symptoms; the elevated and non-suppressible serum ACTH
..,ells, and Rathke pouch epithelium . level is due to its overexpression by tumour cells 1707,2828 ).
Other hormones may also be overexpressed (562). Ophthalmo-
ICD-0 coding plegia frequently occurs due to extension of the tumour 1n the
82 3/3 Pituitary blastoma suprasellar and parasellar reg ion (28 28) .

!CD-11 coding Epidemiology


2012.Y & XH5QV8 Other specified malignant neoplasms of Pituitary blastoma is an exceptionally rare tumou r, with fewer
other endocrine glands or related structures & Pituitary blas- than 20 published cases . It usually occu rs in chil.dren aged
toma < 2 years, with a median age of 9 months and a slight female
predominan ce {707,2828) . Patients may present with or subse-
Related terminology quently develop other O/CER1-related tumours 1707) .
Not recommended: pituitary embryoma.
Etiology
Subtype(s) Pituitary blastoma is linked to underlying germline and somatic
None variation in O/CER1, a critical gene in mic ro RNA processing
1707).
Localization
Pituitary blastoma originates within the sellar region and fre- Pathogenesis
Quently extends into the suprasellar region and invades the Pituitary blastoma appears to arise because of a genetic altera-
cavernous sinus {2828}. tion in O/CER1, which encodes an enzyme involved in microRNA

Flg.12.25 Pituitary blastoma. A The cellular compon ents of pitui tary bla::turn<t'> Ir"·, ud• 1'1 1•J · J .• :. i:_, : 1
M>otdal primitive Rathke pouch epithelium with rosetie or gland'foll1clt furroi::tion . 11 1·i SL 31 '-'L·'1 ' ,., ' ' '''"" .., 8 1' -' J• ·• •1c11 i) ' 1•1la 1 y n0L.!1J::;odr.,r1ne
tumour cells are arranged in diffuse sheets or lobules, and !hey arc van.:iDly pos live !t.r ..,,, 1H Li ,.,~"" J< 1 ···k '·" · ,. '' - ., ' 1' · ' P • 11 • • ' -~' ,; 1 '"tr ": ~1":1 1 ~e p, ·:ch
;;~~thel1um and blastemal undifferentiated cells are neg&.tive fc.r ACTH c CAM5 2 ~ t a ll !'.; d• ,.- c,l :·I"' l \ ' . '· ·~"
ji ;i• r ~ ':· 4;. 0 ::-~·~I; '=' ·L' ~ )(,d-, l~f)~ ~
'r

,? ',' ·J: :
e-.tpress1on distinguishes areas of neuroendocrine eel.~ .vh~rea<. 01h er cel1ular eie~llfnt~ ::.Ii\~~~~~ 1 '~c.·.:. 1.·, :-i.' ,::. :~-:. r r '- · J'· : - ·~·~ ~i:·- ,,,i\i : ' 1,,i- s 1,;!'1u1Jr
C::rnponems is illustrated in this photomicrograph. The 101>1:1E :om11ng cuboidal or ..,olurnr H P· '· F· " h · · - - •" · ' '· ·, · '· ~ -· "' . ;, 7 ~ ·,,: •er ar •..; ; :r ~> tfl :::: . :•.
'tie other ~ellular elements. f 5100 stains scattered fo 1 ~rd;:.s;.~!'ate cells

i,
• -' i ~. l t
processing ; mature microRNAs in turn regulate the translation of Box 12.05 Diagnostic criteria for pituitary bl;isroma
mRNA . Virtually all tumours (15 of 15 tested cases) harbour at
Essential:
least one DICER1 alteration , typically a germline loss-of-function
p~thogen i c vari.ant coupled with a somatic RNase Jllb hotspot Rathke pouch epithelial glands, primitive blasfomaloua certs. and
folliculostellate anterior pituitary cells
m1ssense mutation [2776 ,707) . In a minority of cases , the second
somatic alteration may be loss of the wildtype allele (707) . AND
DICER1 alterations
Macroscopic appearance
Desirable:
Descriptions of the macroscopic details of pituitary blastomas
are limited . Focal cystic or haemorrhagic changes and partial Diagnosed in children aged < 2 years
necrosis can be seen \2829) . Cushing syndrome
Personal or family history of DfCER1 syndrome
Histopathology
Pituitary blastomas are composed of three ce llular components:
(1) large, anterior pituitary neuroendocrine cells arranged in lob- tumours (PitNETs) with synchronous Rathke cleft cyst (2774
ules or diffuse sheets; (2) cuboidal or columnar primitive Rathke 694}.
pouch epithelium with rosette or gland/follicle formation; and (3)
small undifferentiated blastemal cells [2829,2828,707). Cytology
Ultrastructurally, pituitary blastomas resemble fetal pitui- Details of intraoperative cytological preparations have not bee~
tary gland of 10-12 weeks, but they are distinguished by (1) a described .
marked complex cellular proliferation of mature TPIT-lineage
corticotroph cells (some with Crooke hyaline change) , (2) PIT1 - Diagnostic molecular pathology
lineage somatotrophs with a background of anterior pituitary DICER1 variants may be considered a diagnostic molecut
cells with small secretory granules that simulate null cells , and marker of pituitary blastoma. Germline and tumour sequenong
(3) other elements including undifferentiated Rathke pouch epi- should be performed .
thel ial cells and folliculostell ate cells (2829,2828}.
Neuroendocrine cells variably express pro-opiomelanocortin Essential and desirable diagnostic criteria
derivatives including ACTH, p-endorphin, and MSH, and to a See Box 12.05.
lesser extent growth hormone {2829}. Rare LH-P [2829) and/
or FSH-P \2829 ,2776) staining has also been reported. Unlike Staging
neuroendocrine cells, blastemal and Rathke pouch cells rarely Staging should include brain and spine MRI with and wfthc.u
show pituitary transcription factor expression {2828) . EMA and contrast, endocrine evaluation (including ACTH level). and opll-
keratins are expressed in various components , with stronger thalmological evaluation . The role of cerebrosp1naJ fluid cyt
staining in the Rathke pouch epithelium {2829}. Galectin-3 vari- ogy is uncertain , and its use in individual situations may dep
ably stains cellular components [2829}. Proliferative heteroge- on the safety of lumbar puncture in the cl inical setting .
neity varies between cases and between tumour components
{2829 ,2828) . A high Ki-67 labelling index (up to 60%) and p53 Prognosis and prediction
expression are more frequent in the Rathke pouch epithelium Because of the rarity of this tumour, prognostic factors are n
{2829,2828,707) . Necrosis can occur {2828}. yet fully understood. In the largest series of 13 patients w1
The differential diagnosis includes sellar teratoma, germ cell pituitary blastoma, 5 children (38%) died: 4 from early or tate
tumou rs, and pituitary adenomas I pituitary neuroendocrine treatment-related complications and 1 from progression [ 7C:")

'~ I ri I : JI; 1 ,, ,, • \·I Ir c: '.-," 1 1r 11 , , , , 11


Metastases to the brain and spinal cord M1ttelbrnnn MGA
Ahluwalia MS
Rr; ...;~r1hl1;rr AV
fan:::tlt>i ,.)
parenchyma Bra stiano<J PK
Preusser WM
1 .N rnl-l,~r FP.

Definition Male.
Metastases to the brain and spinal cord parenchyma are
tumours originating outside the CNS and sp reading into the
Primary
brain and spinal cord parenchyma via a haematogenous route
tumours
or (less frequently) directly from adjacent anatomical structures .

ICD-0 coding
None

ICD-11 coding Brain


metastases
2050 Mal ignant neoplasm metastasis in brain , with extension
code for primary tumour type

Related terminology Fig. 13.01 Relative frequencies of primary tumours and of brain metastases dem":'d
None from them. Tumours with a high propensity to metastasize to the brain are lung cancer
breast cancer, renal cell carcinoma, and melanoma. In this series of brain metastases..
Subtype(s) about 14% of cases in male patients and 8% of cases in female patients were diag-
None nosed as carcinoma of unknown primary (CUP) {459,2568}. Data are based on cne
histology of archival tissue samples: 874 cases collected in 1990-201 1 at the ins ute
of Neurology (Neuropathology}, Medical University of Vienna. Metastases for wtildl
Localization surgery was not performed are not represented. The relative frequencies of bram me-
Approximately 80% of all brain metastases are located in the tastases may differ substantially in other regions of the world.
cerebral hemispheres, particularly in arterial border zones and
at the junction of the cerebral cortex and white matter; 15%
occur in the cerebellum and 5% occur in the brainstem . Fewer
than 50% occur as a single brain metastasis and very few as the
only (solitary) metastasis in the body {1049,2439} . Occasionally,
CNS metastases seed along ventricular walls or are located in
the pituitary gland or choroid plexus.

Non-parenchymal, non-diffuse meningeal metastases


In addition to having diffusely or multifocally disseminated
leptomeningeal metastases (see Metastases to the meninges,
p. 421) , 8-9% of patients with advanced cancer may also
present with circumscribed dural metastases {1791} . The dis-
tinction between diffuse and circumscribed meningeal metas-
tases is clinically important because the latter has a slightly
better prognosis and patients can usually undergo local treat- Fig. 13.02 Single brain metastasis from colorectal cancer. A Ring-enhancing 1 n
ment strategies 11822). Metastases to the dura and leptome- in the cerebellum on T1 -weighted, postcontrast axial MRI. B FLAIR MRI sriow1ng a
nin ges are most frequently linked to extension from or to other great extent of vasogenic oedema.
CN S compartments , with the vast majority affecting the spinal
cord via expansion from vertebral or paravertebral tissues into effects on the adjacent brain tissue . The symptoms may pro-
the epid ural space /1791,22 19,2 182}. Dural metastases are gress gradually and include headache , altered mencal status.
relativel y common in cancers of the prostate, breast, and lung , paresis, ataxia, visual changes , nausea, and sensory d1srur-
and in haematological malignancies 11791,2219). Spinal epi- bances . Some patients present with seizure. infarct. or haem-
dural metastases are most common in cancers of the prostate, orrhage (1830} . The interval between d iagnosis of the p r1rial)I
breast, lung, and kidney, as well as in non -Hodgkin lymphoma tumour and the CNS metastasis is frequently < 1 year ror lung
and multiple myeloma . lntramedullary spinal cord metastases carcinoma, but it can be many years for breast cancer aPO
are rnost common in small cell lung carcinoma /2182} . melanoma (248) .

Clinical features Imaging


I ~eurological symptoms of intracranial metastases are gener- On MRI , intraparenchymal metastases are general! circum-
ally caused by increased intracranial pressure and local tumour scribed and show mild T1 hypointensity. T2 hyper intens1t~. ana
diffuse or ring-like contrast enhancement with a surrounding in the various tumour types is still incompletely understood and
zone of parenchymal oedema . Haemorrhagic metastases and requires further study. Local spread of C~~S tumou~s may also
metastatic me~anom.as containing melanin pigment may dem- occur by direct extension from primary tumours in adiacent
onstrate hyperintens1ty on non-contrast MRI or CT (3551). anatomical structures (e .g . paranasal sinuses and bone) 128821-
Such tumours are not formally considered metastases because
Epidemiology they remain in continuity with the primary neoplasm .
The incidence rates reported in the literature probably under-
estimate the true incidence of brain metastases because of Macroscopic appearance
underdi~gnosis and inac~urate reporting (972,3099). In a large Metastases in the brain and spinal cord parenchyma often form
population- base~ stu?y 1n Sweden , the incidence of patients grossly circumscribed and rounded g:eyish-wh1te or tan masses
admitted to hospital with brain metastases doubled to 14 cases with variable central necrosis and pentumoural oedema . Metas-
per 100 000 person-years between 1987 and 2006 . More effi- tases of adenocarcinomas may contain collections of muco1d
cient control of disease spread outside the CNS and the use material. Haemorrhage is relatively frequent in metas ases of
of more a~v~nced neuroimaging techniques may have contrib- choriocarcinoma, hepatocellular carcinoma. melanoma. and
uted to this increase (2956 ,2353,3099) . Autopsy studies have clear cell renal cell carcinoma. Melanoma metastases with
shown that CNS metastases occur in about 25% of patients who abundant melanin pigment are brown to black in colour. Primary
die of cancer {1049). neoplasms in the head and neck region that extend intracrani-
ally by direct invas ion generally cause marked destruction of
Age and sex distribution the skull bones . However, in some cases. the skull is penetrated
CNS metastases are the most common CNS neoplasms in by relatively subtle perivascular or perineural invasion , without
adults, but metastases account for only about 2% of all pae- major bone destruction (28821.
diatric CNS tumours . As many as 30% of adults and 6-10%
of children with cancer develop brain metastases. The relative Histopathology
proportions of various primary tumours are different between The histological and immunohistochemical features of CNS
the two sexes, but for most tumour types , sex has no signifi- metastases are as diverse as those of the primary tumours from
cant independent effect on the occurrence of CNS metastasis which they arise. Most brain metastases are fairly well demar-
1201 ,972,3437} . The incidence of brain metastases has been cated, with variable perivascular growth (vascular cooption) in
reported to be highest among patients diagnosed with primary the adjacent CNS tissue (247}. On occasion, small cell carcino-
lung cancer at the age of 40-49 years ; with primary melanoma, mas and lymphomas may show more diffuse infiltration (pseu-
renal cancer, or colorectal cancer at the age of 50-59 years ; dogliomatous growth) in the adjacent brain parenchyma 1219,
and with breast cancer at the age of 20-39 years [3099). 2237) . Tumour necrosis may be extensive, leaving recognizable
tumour tissue only at the periphery of the lesion and around
Etiology blood vessels (2439) .
most common source of brain metastasis in adults is lung
cancer (especially adenocarcinoma and small cell carcinoma), Proliferation
~ed by breast cancer, melanoma , renal cell carcinoma , Metastatic CNS tumours show variable and often marked mitotic
and colorectal cancer (459,2252,2568). Tumours and their activity. The Ki-67 proliferation index may be sign if 1cantly higher
molecular subtypes vary in their propensity to metastasize to than that of the primary neoplasm [246) .
the CNS (201,661,248) . In as many as 10% of patients with brain
metastases, no primary tumour is found at presentation (2568) . lmmunophenotype
In children, the most common sources of CNS metastases are The immunoh istochemical characteristics of CNS metastases
leukaemias and lymphomas, followed by non-haematopoietic are generally similar to those of the tumours from which they
CNS neoplasms such as germ cell tumours , osteosarcoma, originate . lmmunoh1stochemical analysis 1s often ery hel pful
neuroblastoma, Ewing sarcoma, and rhabdomyosarc oma (661 , for distinguishing between primary CNS tumours and metasta-
3437}. Occasionally, primary neoplasms in the head and neck ses and for assessing the exact nature and origin of the meta-
region extend intracranially by direct invasion , sometimes along static neoplasm (particularly 1n cases with n unknown primary
cranial nerves, and manifest as intracranial tumours (2882). tumour) [229,24391 (see Table 13 .01 , p 420)

Pathogenesis Cytology
Before they manifest as haematogenous metastases in the Cytological fe atures depand mainly on the type or primary
CNS, tumour cells must successfully complete a series of tumour
steps: escape from the primary tumour, enter into and survive in
the blood stream, get arrested in brain capillaries. extravasate Diagnostic molecular pathology
mto the CNS, and colonize the perivascular niche that allows The use ot n1olecular markers for Cl\JS metastases 1s becom -
survival and subsequent growth 1n the CNS microenviroriment ing 1ncr as1114ly 1rw c rtant becquse the molecular proflies of
This process occurs via interactions with various cell types. µnrnary tU1nour hr Lllr 1 n ier 'l~l<-i~~es can vary and treatment has to
including neurons, and with the extracellular matrix 11609,2568. bt:' a• upt •cJ acn.rr!1n;;ly (?· 11 ,:JOO,.f In add1t1on some markers
3586}. An alternative, direct route to the brain us1 ny bri dg11.g ~· 1 h ~("_, PDL 1 2 1 ;.:. e u.u:itJ!e tw 1m1 unoh1stocr1~m1stry anrl 3re
vessels from the bone marrow has only been describecJ for lt:u , 11 10 1L vu':' d 111-: ur ia.J ·~cE.ctru111 of er Jt1es 1691). but shortcom
kaemia cells so far {3527}. The molecular basis of O~S sr.w ~!d 1n._. ot irw~ .::iprno:-11l1 (P c; ,n r-:: 1 urr.oural heterogeneity. ml1lt1p!e
Table 13.01 Diagnostic and theragnostic markers for CNS metastases

Primary tumour blagn-01tlo mart<ers


Uc
Melanoma Melan-A, HMB45, SOX10, BRAF p.V600E BRAF, NRAS. KIT. POL 1
Non-small cell lung carcinoma CK7, TIF1 , napsln A
EGFA, ALK, KRAS, BRAF, EAB82 (HER~ . MET ROSI PO T
Small cell lung carcinoma CK?, CD56, TIF1 None
Breast adenocarcinoma CK7, GCDFP-15, GAT A3, mammaglobin ER, PR, ERBB2 (HER2), PDL1 , gene expre iori pan
Ovarian carcinoma CK7, WT1 , PAX8 BRCA1 , BRCA2, CHEK2. PALB2. RADS IC and.or RA0510
Squamous cell carcinoma CK5/6, p63, p40 None
Renal cell carcinoma PAX8, CD10, RCCm None
Urothelial carcinoma CK5/6, CK7, CK20 PDL1

Colorectal carcinoma CK20, CDX2 KRAS, BRAF, NRAS, microsatellite instablllty (MLH1 ). mis atch ·epcir
immunohistochemistry (e.g. MSH2. MSH6. MLH1 . PMS2)
Gastric adenocarcinoma CK7, CK20 ERBB2 (HER2), POL 1
Prostate adenocarcinoma Pancytokeratln, PSA, PSAP, NKX3-1 None
Thyroid carcinoma TIF1, thyroglobulin, PAX8 None
B-cell lymphoma CD45, CD20, CD79a B-cell clonality (IG genes)
T-cell lymphoma CD45, CD3, CD4_, _
co_s_____ T-cell clonality (TR genes)
TIF1 , thyroid transcription factor 1.

different antibodies for distinct entities or treatment schemes, Box 13.01 Diagnostic criteria for metastases to the brain and spinal cord par~ :hyf"l
entity-specific evaluation algorithms) are associated with poor Essential:
1nterobserver reproducibility {2677) . In addition , there is increas-
Detection of malignant non-pnmary cells witf11n the brain or spinal cord
111g evidence that tumour cell expression of POL1 might be parenchyma
induced by infiltrating immune cells, and it may therefore (at least
partly) be considered an indirect effect, thus potentially being of Desirable:
lower predictive value than the immune cell infiltration itself (1951) . Fulfilment of specific diagnostic criteria for the primary tumour type
Molecular alterations that are frequently found , and that are of
high clinical importance because of the availability of targeted
drugs with activity against brain metastases, include EGFR muta- CT of the chest +/- abdomen is typically performed . FOG PE
tions and ALK fusions in lung cancer {1902), BRAF mutations in may be of additional value in cases of unknown pnmary t... ro
melanoma {2884), and ERBB2 overexpression in breast cancer {2982}. After treatment of brain metastases, regular tallow- o
(3265). For other treatable molecular alterations that are found at imaging is recommended , normally every 3 months
low frequencies in various cancers (e.g . NTRK fusions, involving
various oncogenic fusion partners), broad immunohistochemical Prognosis and prediction
screening might be useful in the future 12989}. The main establ ished prognostic factors for pauents w11h bra.iri
metastases are patient age , Karnofsky performance status
Essential and desirable diagnostic criteria number of brain metastases, and status of extracran1al dis.ease
See Box 13.01. Several prognostic scores taking these parameters into ace •un
have been described, but they require validation 1n independerir
Staging and prospective studies [1696 ,3006}. Other factors or rog-
Despite the high incidence of brain metastases , in certain nostic significance include the specific tumour type
cancers like metastatic melanoma , lung adenocarcinoma, and molecular drivers involved (e .g . ERBB2 [HER2] in be
ERBB2 (HER2)-positive and triple-negative breast cancer, brain cer) {3010 ,3007,3008) . Neuroradiological parame rs
1mag1ng is not .an established part of primary staging , but it peritumoural brain oedema may also provide progno
should be cons idered whenever clinically meaningful (at the lat- mation [3001) . In more recent studies, the repor d fJJJl'OW~ -t r l

est when neurological symptoms develop in patients with known


ment in the overall survival of patients with CNS me~:la!iQS
cancer) MRI .is recommended over CT when available, because
may be attributable to improvements in focal surgic
of its much h1gh~r sensitivi ty and the opportunity it provides to
for single lesions. stereotactic radiosurgery, and syst
exclude d1fferent1al diagnoses \446 ,68) . If the primary tumour is
pies . in combination with earlier detection of such rrnetas~i*i
not known, a skin exam1nat1on, gynaecological examination, and
12252.2982) .

420 Met astases to tlie C l\JS


Rosenblum 1W
Metastases to the meninges Mittelbronn MG A
Ahluwalia MS Tanal<a S
Brastianos PK VVinvler FA
Preusser WM

;._ etast~ses .to the meninges are tumours originating outside the

· S with diffuse and/or multifocal spread within the leptome-
~1 ges and the subarachnoid space.

ICD-0 coding

• ne
• I• • •·
ICD-11 coding oe••
2051 Malignant neoplasm metastasis in meninges •
•o ••• • 0
:
Related terminology A •• ••• ••
o~ ~ecommende?: leptomeningeal cancer; neoplastic menin-
g1t1s; (lepto)meningeal carcinomatosis .

Subtype(s)
ne

Localization
~ inges. For non-diffuse meningeal metastases , see Metasta-
ses to the brain and spinal cord parenchyma (p. 418) .

Clinical features
Many patients with leptomeningeal metastasis (LM) have multi- B
e and varied neurological symptoms at presentation , including Fig. 13.03 Metastases to the meninges. Cytological preparations showing metastatic
t-iea.dache, mental alteration, ataxia, cranial nerve dysfunction , cells within the cerebrospinal fluid . A Lobular breast carcinoma, showing single-file-
and radiculopathy. Clinical diagnosis can be made according like group and mitosis. B Melanoma metastasis; inset: S100 immunochem1stry.
to current diagnostic criteria {1822}. Cytological examination
reveals malignant cells in the initial cerebrospinal fluid (CSF) Imaging
sample in about 50% of such patients ; this proportion may In patients with LM , MRI can show focal or diffuse leptomenin-
1;icrease to ~ 80% when CSF sampling is repeated and ade- geal thickening and contrast enhancemen t (sometimes with dis-
q ate volumes (~ 10 ml) are available for cytological analysis persed tumour nodules in the subarachnoid space) . Enhance-
i506 ,1830}. Spinal metastases generally result in compression ment and enlargement of the cranial nerves and commun 1cat1ng
of the spinal cord or nerve roots, and they may cause back pain, hydrocephalus may also be found l1119).
oeakness of the extremities, sensory disturbances, and inconti-
n&nce over the course of hours, days , or weeks /1830) . Epidemiology
LM occurs in 4- 15% of patients with solid tumours (506,.J OSI,
however, this number might be underest1matec.i due co lad. o~
specific symptoms In cases of already existing br.:t1r metas-
tasis, rates of LM may increase to 33-54°'0 1n breast ca·1c8r.
56- 82% in lung cancer, and 87 96°0 11 n elanorna [ 1S~~r The
highest incidence rates or L r-...1 h~ve btk:n itJp )t ted 111 rl'e-' .J::>L1t1c
melanoma l?J-;~) a11t! lu19 c 1ril tA 1 ',)<\,) t 11! l\\1:'".i b t 1t'.1:3t
crncino;11a (5°'o) \ 11/Clt

Etiology
Mew ~ tdtl C '>JJ• kld u _•,.ii\ fl - . •1,. t' '< 1 'h. ___ • i id irl.·rn 1 Jll-
Cf\I " iD.Jl1u r1 1r1t r1~·-'. .: 1:>• 1

Pathogent:H:>IS
Flg.13.04 Leptomeningeal metastases from breast cam1.::ma A D•ffuse entlance-
Onr:c tr, cunlaL t \ it ~- r ,, L- )
ment of cortical sulci on T1-weighted, postcontrast a~1a1 MR 1 B Lint:ar enllancernent L• ! ti<1rt111 l' .. ._]or , Ii · fl
!rorn the brain or ~~·rn, .._;, m ; ir. l°t d1 tiy VIC°"t t~',~ ~ ~ Sr Ja.11
of tl1e spinal leptomeninges on T1-weighted , postcontrnsl S3.:)it! al MRI. i'11j

\i l
42·1
'· '~
Fig. 13.05 Cerebrospinal fluid (CSF) metastasis of B-cell lymphoma. The cellular Flg. 13.06 Diffuse meningeal metastasis. Cerebrospmal fluid shawm~ the ;::r~
component of normal CSF consists almost exclusively of cytologically atypical lym- of numerous metastatic breast adenocarcinoma cells. Note the i:lustermg ~r.a 'ar}I!
phocytes (60-70% of the cells) with round nuclei and sparse cytoplasm, mainly of size of the carcinoma cells in relation to the small lymphocytes.
the CD4+ memory phenotype, and monocytes with a bean-shaped nucleus (30-40%
of the cells), without associated haemorrhage (red blood cell) as in the present case;
apoptotic nuclei can be present. Finally. i mm u nocytoch~.~
neutrophils in the CSF indicate a barrier disturbance. When there is leptomeningeal
spread, tumour cells are primarily recognized by their large nuclei (here 4-5 times assessment for lineage-specific or even therapeutic m an< ~
larger than the nuclei of normal lymphocytes and monocytes), which are often irregu- can be an asset to the diagnostic procedure. The unequivoCiJ
lar and hyperchromatic. Frequently, the neoplastic cells display prominent nucleoli, a detection of malignant cells in the CSF should be reoor 'JC1
cytological feature that is absent in cells of the normal CSF. Abnormal binucleated or as ''positive", the detection of suspicious or atypical cetis
multinucleated cells are also observed. Mitoses are a frequent finding In leptomenin- "equivocal '', and the absence of malignant or equivocal cells as
geal spread of tumour cells. (Pappenheim stain) "negative" {1822) .
Box 13.02 Diagnostic criteria for metastases to the meninges
Diagnostic molecular pathology
Essential: Novel liquid biopsy approaches hold promise for more accurate
Unequivocal clinical and/or radiological evidence of leptomeningeal metastasis detection of, for example, circulating tumour cells and ceH-free
tumour DNA , so they may help improve diagnostic prec1s100
Desirable: therapeutic management, and treatment monitoring. There is
Presence of malignant cells within the cerebrospinal fluid increasing evidence that such liquid biopsies of CSF const1Me
lmmunohistochemical demonstration of the origin of the metastatic cells a reliable additional diagnostic tool , especially in cases that are
highly suspicious for LM but that have negative or equivocal
results in classic cytolog ical assessments of the CSF (3161
metastatic tumour cells may disseminate (seed) diffusely along
the leptomeninges. Essential and desirable diagnostic criteria
See Box 13.02.
Macroscopic appearance
LM may produce diffuse opacification of the membranes or Staging
manifest as multiple nodules {2439}. Central staging methods are MRI of the brain and spine, and CSF
diagnostics (cytology, as well as measurement of opening pres-
Histopathology sure, protein, glucose, and lactate). The highly vanable chnrcal
In LM , the tumour cells are dispersed in the subarachnoid presentation - with many possible combinations of solid menm-
(including Virchow- Robin) space and may invade the adjacent geal manifestation in various places throughout the CNS. an
CNS parenchyma and nerve roots {31 05}. or the presence of non-adherent (fluid) tumour cells 1n the CSF -
has complicated the development of generally accepted critena
Cytology for diagnosis, response assessment, and follow-up !18201
More than 90% of patients with LM show at least one of the Union for International Cancer Control (UICC) staging :>hou
following non-diagnostic pathological features in the CS F: be performed according to the criteria for the specific primary
increased opening pressure (> 200 mm H20), increased leuko- tumou r, when known .
cyte count (> 4/µL) , elevated protein (> 50 mg/dl) and lactate
(> 2.4 mmol/L) , and decreased glucose levels (< 60 mg/dl) Prognosis and prediction
{762 ,1822). However, the final proof of LM is sti ll provided by The prognosis is dismal for patients with LM , so pragmatic treat-
the highly specific detection of malignant cells in CSF samples ment approaches primarily focus on life prolonga11on w th an
by cytology. Normal CSF samples almost exclusively consist acceptable quality of life (1822} . Besides standard intr8\tenous.
of lymphocytes (60- 70%) and monocytes (30- 40%) , whereas or intra-CSF pharmacotherapy, focal radiotherapy can be used
> 50% of all patients with LM also show elevated numbers of for circumscribed, symptomatic lesions, whereas wno1~orai
granulocytes ind icating a barrier disturbance {762) . Neoplas- irradiation might be preferential for e tensively d1ssem1nated
tic cells within th e CSF are typically recognized by increased LM . The increasing availability of targeted treatments C1 11q-
nuclear an d total cell size , irregular cytoplasmic and nuclear uid biopsy approaches allow for more 1nd1viduahzed th~Y
shape, and prominent nucleoli . Furthermore , mitoses and according to molecular cancer profiles.
422 MPtas1a~f:s 1cJ 1he CN 0
Genetic tumour syndromes of the nervous Perry A

system: Introduction

Th e central and peripheral nervous systems are frequently encounters the fi rst clue, so it 1s en real!'/ '"rpr, r ·~r+ ,., .. c: ..., -:' -
impl icated in a wide range of genetic tumour predisposition of when to raise th e possibility of genetic tr..Jrrs Jr - ·r r.·-r- - -
syndromes . These disorders are often hig hly complex, and the patient's cl inical team for further wo r~uo Sonia. t •r ~ - •
patients are typically best serve d in specialized centres with man re lationships between syndromes and rurrr; 1.. . rs ;:. .,-c; r ,.. -4

broad multidisciplinary expertise. Traditionally, most of these been appreciated; for instance, most patholog1SiS "'rir 1 'f
patients' diagnoses have been based purely on clinical features ti ght associations between multiple and/or plex 1=,..r'T1 -e ·-.
and family histories . However, more and more often , a heritable bromas (sometimes with transformation to mahgr am G.:::r -- C::'~
pathogenic variant is first detected as part of a genomic screen - nerve sh eath tumour) and neurof1bromatos1s typi:; 1 . :Ji::' ,~ ­
ing assay, such as next-generation sequencing of tumour and/ bilateral vestibular schwannomas and ne rof or'" rr ~tr:,
or germline DNA. Not uncommon ly though , the patholog ist type 2, between haemangioblastomas and van Hippe -L ~ _J

Table 14.01 Tumour scenarios that should prompt a pathologist to consider a potential underlying genetic tumour syndrome
Tumour scenarJo Genetic tumour 1yndrome(s)
---~~~-~

Bilateral vestibular schwannomas NF2


Choroid plexus carcinoma Li-fraumenl syndrome
Dysplastic cerebellar gangliocytoma (Lhermitte--Ouclos disease) Cowden syndrome
Embryonal tumour with multilayered rosettes lacking C19MC alteration DICERf syndrome
Haemangioblastoma Von Hippel-lindau syndrome
Hybrid neurofibroma/schwannoma NF1 , NF2, and schwannomatosis
IDH- and H3-wildtype, p53-positive glioblastoma in a child Li-fraumeni syndrome
Constitutional mismatch repair deficiency, Lynch syndrome. and Li-Fraume
IDH-wildtype giant cell glioblastoma in a young patient
syndrome
IDH1 p.R132C/&-mutant astrocytoma in an adult Li-f raumeni syndrome
Malignant melanotic nerve sheath tumour Carney complex
Malignant peripheral nerve sheath tumour arising from a neurofibroma NF1
Meningioma in a child NF2
Multiple meningiomas NF2
Multiple neurofibromas, a plexiform neurofibroma, or a massive soft tissue
neurofibroma NF1

Multiple schwannomas or one with mosaic SMARCB1 (IN11) expression NF2 and schwannomatosis
Paraganglioma with loss of SDHB expression Familial paraganglioma syndromes (see Table 14.06, p. 465)
Pineoblastoma DICERf syndrome and familial retinoblastoma syndrome
Pituitary blastoma DICER1 syndrome
Primary intracranial sarcoma, D/CER1-mutant DICER1 syndrome
Rhabdoid and/or papillary meningioma BAP1 tumour predisposition syndrome
Rhabdoid tumour(s) in an infant Rhabdoid tumour predisposition syndrome
SHH-activated medulloblastoma Naevold basal cell carcinoma {Gartin) &¥Jl(lrom ELP1 -medun1oblaSt011l'l3 S\.1!0n:ime,
and GPR161 (Gorfln-like) s.yndrom
SHH-activated, TP53-mutant medulloblastoma (often the large cell / anaplastic
histological type) U- Fraumani syndrome and Fanconi anaemia
Subependymal giant cell astrocytoma
Tuberous sclerosis
WNT-activated medulloblastoma, CTNNBt-wildtype
Familial adenomatous polyposis
NF1, neurofibromatosis type 1; NF2, neurofibromatosis type 2; SHH, sonic hedgehog.

424 Genetic tumour sy r1drmnes 1nvolv11 19 thp Cl\ IS


s !ldrome. and betwe~n subependymAI gin.nt cell astrocytormi.s 19 syndrnrnes covered hP.rA ar~ ;:v1r I l t ,. n"il'/ rjcir ( rir;~fj
1 '' > .., •• · •
r r f ~ C";(
, -
and tuberous sclerosis . However, many pathologists are less volume s of th8 WHO r,lac:;sif1r;at1rJn ~ 1nr1Atr;e>IA ,'_ tt·,~rc~ ..., ,.., ~, J
aware of many of the oth~r s~ndromic associations highlighted rally a greater focus 0n nervous c:;yc;terr. finrJinr.r: ·r· tr 1 ,,. It 1rr.to.
,n Table 14.01 . The combination of colonic polyposis and brain For some of these syndromes. the r,ltn1r;8I qi;irJt":iltn~s ,r)r rl .:V.J- 1

tumours was historically designated as Turcot syndrome, but nosis have also evolved therefore tre riewAst ~>r rnry.:;i "'1e:11
in:s designation is no longer appropriate , because it is now used iterations are emphasized of en 1n tabl8 f0rm;=H P - ~r:,t,
~ ar that this outdated eponym in fact comprises multiple now cal diagnostic approaches utilizing surrogate 1 rnrrur0st.::i1rr
11 ~defined cancer predisposition syndromes, each with a dif- are also highlighted when appropriate along N•th n·..:: mr:iri::-
te ant pathogenesis. traditional molecular diagnostic echn1qUeS rhaf ma/ Q~ rgf)ljl
our understanding of genetic tumour syndromes has evolved site for a definitive diagnosis In most cases the mar.::roscooy
idly since the 2016 WHO classification of CNS tumours histopathology, and cytology descnpt1ons of 1nd1v1dual turno1Jr
su -:h that eight additional disorders are now covered in this ne~ types seen within a genetic syndrome are cover~d entire!/ 0r
ed1~1on: Carney complex, DICER1 syndrome, familial paragan- in greater detail within the earlier sec ions dedicated to the
~ oma syndrome, melanoma-astrocytoma syndrome, familial sporadic counterparts of those dist1nc entities In contrast the
noblastoma, BAP1 tumour predisposition syndrome, Fanconi clinical spectrum, pathogenesis, and molecular genetics of he
anaemia, and ELP1-medulloblastoma syndrome . Several of the syndromes are covered in greater de all here

.i '
LAq 1ue; f:.
Neurofibromatosis type 1 F1sh~r M J
Gutmarin [JH
Re1Jss DE
Rodriquez FJ

Definition
Neurofibromatosis type 1 (NF1) is an autosomal dominant disor-
der caused by sequence variation in the NF1 gene , diagnosed
clinically when at least two of the following are present: multiple
cafe-au-lait macules, skinfold freckling , iris hamartomas (Lisch
nodules), optic pathway glioma I pilocytic astrocytoma (OPG),
multiple neurofibromas or one plexiform neurofibroma , specific
osseous lesions , and an affected first-degree relative .

MIM numbering
162200 Neurofibromatosis, type I; NF1

ICD-11 coding Fig.14.01 Neurofibromatosis type 1 (NF1 ). A Multiple cafe-au-la1t spcts :r _


L020.10 Neurofibromatosis type 1 back in a child with NF1. B Cutaneous neurofibromas on the left am; ;:! M" 301.:·
with NF1 .
<elated terminology
Jt recommended: von Recklinghausen disease; peripheral growth relative to the rest of the tumour. or continued g ..... .., ..... : ·
neurofibromatosis . a plexiform neurofibroma into adulthood .
Specific bone abnormalities can be present 1n 1nd1v dua's t"'
Subtype(s) NF1 . These include severe scoliosis. spheno1d wing cr1sp as.a
Mosaic neurofibromatosis type 1, including segmental neurofi- non-ossifying fibromas, and congenital tibial bowing_ Prog"es-
bromatosis type 1; spinal neurofibromatosis; neurofibromato- sive tibial bowing can result in pathological fracture. • , t~
sis-Noonan syndrome; 17q11 .2 microdeletion syndrome development of a pseudarthrosis 12840).
Many children with NF1 exh ibit learning disabilrt1es_ a, -
Localization tion deficit hyperactivity disorder, and problems 1 h recp ~ ·
NF1 affects many different cell types and tissues in the body, cal social interactions (2165}. MRI brain morphometry re e ~
including both the central and peripheral nervous systems. a large grey matter volume and large corpus callos c-~
features associated w ith learning disabi lities 1216 }.
Clinical features About 15% of children with NF1 will develop an OPG ...
Multiple cafe-au-lait macules are usually present at birth , and can cause progressive vision loss in ~ 50% of affected ind .±..·
they increase in number during the first 2 years of life. Skin- als (1912}. The second most common locat1on for a g~ ma ~
fold freckling in the axillary, inguinal, and submammary regions the brainstem 11986); however. gliomas can develop "l ~t c'
occurs in > 80% of adults with NF1. Lisch nodules (asymp- locations (1985}. Young adults with NF1 are also prone to - .g-
tomatic iris hamartomas), present in > 90% of adults with NF1 , nant gliomas {2364) . In children, T2-weighted brain MRI s~ ~5
are best detected by slit-lamp examination. Lisch nodules and focal areas of high signal intensity that tend to d1sapped~ .~ . :n
skinfold freckling both usually develop before puberty. Cutane- age and are sometimes difficult to different1ale tram 1 ow-~race
ous neurofibromas , present in> 85% of adults with NF1 11199}. gliomas {1166) .
typically develop during puberty. In contrast, plexiform neurofi- Epilepsy is more common in individuals with NF1 tnan r· : ::
bromas are probably congenital lesions, arising in 30-50% of general population , and drug-resistant epilepsy requ1nrg ~r ­
children with NF1 {1199). Individual plexiform neurofibromas gery is often associated with dysembryoplast1c n~uf'"'cP•:ne. • .11
have variable growth rates , which can be relatively constant tumours {2340). Sleep disturbances are also rnofe ;: fTlmc1 ·•
for long periods of time , but they exhibit the highest growth people with NF1 11890).
potential during infancy and childhood . A subset of plexiform Other neoplasms observed at increased frequency ..i1e 11-.
neurofibromas transform into atypical neurofibroma / atypical nile myelomonocytic leukaemia. rhabdomyosarcoma. g1\J n 3
neurofibromatous neoplasm of uncertain biological potential tumours of the digits. gastrointestinal stromal tumours. µM )-
(ANNUBP) , which can be a premalignant lesion with increased chromocytoma. female breast cancer. and du0dendl ntik1fL1d!'
risk to progress to high-grade malignant peripheral nerve docrine tumours (somatostat1nomas) 1369).
sheath tumour (MPNST) {2109,23 1) . Possible transformation
to ANNUBP or MPNST should be considered in patients with Epidemiology
growing nodular plexiform neurofibromas, plexitorm neurof 1- NF1 is a common autosomal dominant disorder. w1rn d b1rrn
bromas in which an isolated portion exhibits disproportionate
incidence of 1 case per 3000 live b1rtt1s 13: 601.

426 Genetic tumour syndromes i1ivolv1rig tho CNS


EtiolOQY been detected in some low-grade gliomas in NF1 wher<:! ihe I
NF1 1s caused by heterozygous pathogeni c variants in th e NFt · · h stolog1cal features (ana-
are often associated with worrisome ' .
Jene. wl1ich encodes neurofibromin {3360,493 ,3334) . Half of · · ·t t.
plas1a and increased prol1 era ion a) nd more aggressive tumour
(re individuals with NF1 have unaffected parents; those cases behaviour. In the absence of a low-grade precursor such as
.~t NF1 represent de novo mutations . pilocytic astrocytoma, high-grade gliomas in f\JF1 are also char-
acterized by frequent TP53 mutations , as well as COKN2A.
Pathogenesis CDKN28 and ATRXmutations {675)
, eurofibromin primarily functions as a GAP for the RAS family In n eur~fi b romas and QPGs . murine studies have revealed
of oncogenes {1846) . The EVH1 domain of SPRED1 (the protein th at non -neop lastic (stromal) cells with a heterozygous Nf 1
• 11 Heated in Legius syndrome) binds to neurofibromin on both mutation, incl ud in g mast cells. macrophages.' neurons , T cells .
· es of the GAP-related domain and recruits neurofibromin to and mi c rogl ia, are critical for tumorigenes1s and continued
membrane where it can accelerate RAS inactivation {3047, growth .
1313,807}.
Most clinical features of NF1 , including cafe -au-lait macules Macroscopic appearance .
bony abnormalities, and benign tumours , result from a com~ Macroscopic appearances of nerve sheath neoplasms affecting
ptete loss of neurofibromin function (biallelic NF1 inactivation), people with NF1 incl ude small sessile or pedunculated growths
leading to increased RAS and RAS effector (MEK/ERK or AKT/ of the skin (cutaneou s neurofibromas), plaque-like th1cken1ng
mTOR) signalling . Atypical neurofibromas I ANNUBPs harbour of the skin (diffuse neurofib roma), fusiform segmental expan-
additional genetic alterations, including loss of the CDKN2A , sions of individual periphe ral nerve fascicles (intraneural neu-
CDKN28, and SMARCA2 regions on chromosome 9p (231 , rofibromas) , and multinod ular/multifascicular nerve expansions.
2449,3518). MPNSTs have a highly rearranged karyotype , described as a "bag of worms" (plexiform neurofibromas) .
and they frequently show additional aberrations in the PRC2 Massive soft tissue neurofi bromas occur exclusively in individu-
complex and, to a lesser extent, TP53 genes {712,1842,3598). als with NF1 and are characterized by large neurofibromatous
Low-grade gliomas in NF1 are typically characterized by bial- involvement of anatomical seg ments , even of entire limbs, with
telic NF1 loss alone, although other mutations are occasionally infiltration of soft tissue and skeleta l muscle. Fleshy masses and
seen. Alterations in COKN2A, CDKN28, and/or A TRX have areas of necrosis and haemorrhage characterize MPNST.

••

0
11 ~n "1~ tr " , "· •.L'JL~d ,p
.. f- ,f j1 \ l
1-1 ,,1 1 ''L'' r p:.1 ,,,,..3 ,\11t11 r,r-1
'l IJ!iy ,/t,,._[ 1J•J';' 11 " I> ll·l
·1 v·,-, rn, "'"1 .! 11 , - '::-d(j w

t J , , • . \.- ·. . . r ·~ " "


Histopathology Box14.01 Diagnostic criteria for neurofibrl)1t1:J!Ot;1' 'VP'?• 11c· 1
Nerve sheath tumours in NF1 are predominantly neurofibromas. Es ntlal:
which re semble their sporadic counterparts . Cells of the hae-
matopoietic lineage (including histiocytes and mast cells) are
A clinical diagnosis of NF I requires ltle presence
features:
°'
also encountered . which contribute to the tumour microenviron-
ment l1560J.
• Six or more cafe-au-lalt macutes {di > s m
adults)
Recent ly proposed criteria for ANNUBPs are used to
• Two or more cutaneous or subcutaneous neuimbrom.ss or
describe premalignant or worrisome changes in neurofibromas,
neurofibroma
and they include at least two of the following: cytolog ical atypia,
• Axillary/inguinal freckling
hypercellularity, loss of neurotibroma architecture, and a mitotic
count of > 0.2 mitoses/mm 2 and < 1.5 mitoses/mm 2 (equating • Optic pathway glioma I piloc:ytic astrocytoma
to > 1 mitosis/50 HPF and < 3 mitoses/10 HPF of 0.51 mm in • Two or more iris hamartomas (Lisch nodules)
diameter and 0.2 mm 2 in area) 12109). These changes may be • Distinctive bony abnormality (tibiat
present in clinically designated atypical neurofibromas, which, • First-degree relative with NF1 (by the above criteria)
in a subset of cases, later transform into MPNST 11307). In the future, these criteria may be revised to indude Qenl!TJC 1iem-:oalld
Most MPNSTs in individuals with NF1 are high-grade malig- common newly recognized clinical features. In young
nant spindle cell neoplasms with brisk mitotic activity and macules are usually the only clinical sign of NF1. such
necrosis , which either develop in a pre-existing plexiform neu- affected parent, a diagnosis of NF1 cannot be rendered. MUllJJle~rbm s.:::::.':3
with or without skinfold freckling can be seen in other coni:h1ioll1S. ...._ ...... ·~~
rofibroma or arise de nova. A wide spectrum of heterologous
syndrome (MIM number: 611431 ). Multiple neurofibromas can aISIJtIle ,as.s:x:a::=i1
differentiation (cartil aginous , osseous , rhabdomyoblastic , glan- with certain forms of Noonan syndrome, making malecufaranal!VSis:afllia
dular) may be present. increasingly more important for dlagnostic purposes (2083.2
lmmunohistochemical analyses frequently demonstrate p16 with multiple caf0-au-lait spots and malignant tumours (e.g, IEM:aerlrnaJ•. cue~
and H3 p.K28me3 (K27me3) loss {608), as well as decreased mismatch repair deficiency should also be considered {3062}.
or absent expression of Schwann cell markers (S100, SOX10).
Individuals with NF1 are also predisposed to a variety of gl ial p.R1809: these individuals do not develop neurofibromas ·y
neoplasms, with pilocytic astrocytomas being the predominant OPGs {1672,2710,2516 ,3257}. In addition . patients ..vrt ~ -
histological subtype . Difficult-to-classify gliomas with ambigu- sense mutations involving codons 844-848 appear to exr•t-i
ous features may also be encountered, as well as low-grade more severe disease symptomatology, including a higher c -
gliomas with morphological similarities to subependymal giant dence of externally visible plexiform neurofibromas. s~nc -
cell astrocytomas {2378} . High-grade astrocytomas in NF1 tend matic spinal nerve root neurofibromas, OPG . and MP ST ~5'3
to affect young adults, and their aggressiveness is similar to
that of their sporadic counterparts. High-grade astrocytomas in Essential and desirable diagnostic criteria
NF1 frequently have loss of ATRX expression , resulting in an See Box 14.01 .
alternative-lengthening-of-telomeres phenotype 12694,675).
Staging
Cytology Not relevant
Not clinically relevant
Prognosis and prediction
Diagnostic molecular pathology NF1 is an autosomal dominant disorder. where affec.ed a<
Although no mutation hotspots have been identified, a patho- harbour a 50% risk of disease transmission with eac'"' o•ag-
genic NF1 variant is detected in 95% of people with NF1 12084}. nancy. Individuals often have a shortened lifespan. mar ...
Nonsense, frameshift, and splice-site mutations, as well as to malignant disease and stroke 13260,875,2620}. The s
small insertions , small deletions, or small duplications, all result dard mortality ratio for many complications 1s higher in 'wO -
in NF1 inactivation. In 5-10% of cases a 17q11 .2 microdeletion aged < 50 years {3260). Genetic counselling is reccrn~a-oo
is identified , in which multiple genes are codeleted along with and includes the possibility of prenatal and/or preimptama· '
the NF1 gene . One of these deleted genes , SUZ12, a compo- genetic testing .
nent of the polycomb complex PRC2 , has been implicated in MEK inhibitors are approved for use in children ,3
MPNST development 1712,1842,3598). 2-18 years) with NF1 who have symptomatic, 1noperao1e 1a ,-
To date, the most striking genotype-phenotype correla- form neurofibromas, and they are currently being e c11 ... atea '
tions have involved individuals with a specific 3 bp deletion use in adults with plexiform neurofibromas a::; v ell as 1ri c
(c.2970_2972d el) or with a missense mutation affecting codon with low-grade glioma /1170 ,2462,1660}.

428 Gene ti c tu 1r1uLJr sy11cJ ron 1<~s 111vCllv11 i<.J 11 ie C N S


Neurof ibromatosis type 2 Stemmer-Rachamirnov AO
Kratz CP
Louis ON
Schuhmann MU

Definition
Neurofibromatosis type 2 (NF2) is an autosomal dominant dis-
ease caused by a pathological sequence variant of the NF2
gene, characterize~ by rr:ultiple benign tumours and dysplastic/
~amartomatous lesions 1~ the nervous system , including mul-
tiple schwannomas (particularly bilateral vestibular schwanno-
mas). meningiomas, and spinal ependymomas .

MIM numbering
101 000 Neurofibromatosis , type 11 ; NF2

ICD-11 coding
LD2D.11 Neurofibromatosis type 2

Related terminology
Acceptable: bilateral acoustic neurofibromatosis; central neu-
Fig.14.03 Neurofibromatosis type 2 (N F2) schwannomas. Postcontrast Tl-weighted
rofibromatosis. MRI of bilateral vestibular schwannomas, the hallmark of NF2. When large. vestibular
Not recommended: von Recklinghausen disease. schwannomas may compress the brainstem.

Subtype(s) with bilateral vestibular schwannomas and have no family his-


Wishart phenotype (severe); Gardner phenotype (mild) tory {210,871,1197) .
Because of the wide variability of symptoms and time of onset,
Localization it may be challenging to diagnose NF2 on the basis of clini-
Tumours and hamartomas associated with NF2 affect all loca- cal features alone. Particularly difficult to diagnose are genetic
tions of the central and peripheral nervous system, with predi- mosaics (accounting for 30% of sporadic cases) , in which
lections for cranial, paraspinal , and peripheral nerves, as well segmental involvement or milder disease may occur (1665!.
as the meninges and the spinal cord. and paediatric cases in which the full manifestations of the dis-
ease have not yet developed . The distinction from other forms
Clinical features is difficult in some cases. There is cl inical phenotyp1c overlap
Patients with NF2 often present in early adulthood, although between NF2 mosaic , early NF2, and schwannomatosis; some
the disease can manifest in childhood . The most common pre- cases that fulfil the clinical diagnostic criteria for schwannoma-
sentation in adults is with symptoms referable to a vestibular tosis are later proved to be NF2 !2524] . Molecular analysis of
schwannoma (hearing loss, tinnitus, or imbalance). Presen- LZTR1 and SMARCB1 mutations (the two schwannomatos1s
tation with vestibular schwannomas is uncommon in children genes) as well as NF2 mutations is helpful when a pari ent does
(1 5%) whereas cutaneous schwannomas (NF2 plaques) and not meet the clinical criteria for a defi ni te diag nosis and or has
ocular abnormalities are more frequent {865] . NF2 mosaics, in overlapping features of NF2 and schwannomatosis.
which a somatic mutation occurs during embryogenesis, have
variable phenotypes depending on the cell lineages involved. Schwannomas
They may be challenging to diagnose and have clinical symp- NF2-associated schwannomas differ from sporadic tumours 1n
toms overlapping with schwannomatos is !2524]. seve ral ways Th ey occur 1n younger par1ents (1n the third dec-
ade of lite. versus the sixth decade for spornd1c tumours) and
Clinical diagnostic criteria many pat11:rnts develop bilateral vest1bul<:lr schwannomas by
The original clinical diagnostic criteria for NF2 were estarJ- their fuurth c.1ecode ot life 1871,20251 Recent stuL11es vv1th h1gh-
hshed at the US National Institutes of Health (NIHJ Consensus re::,olution MRI sr10wed trtclt 1n mcinv co.st~s patients tKwe rnul ti-
Development Conference on neurofibromatos1 s in 1987 (22171 pl d1~crtto tun1our n0dul0<, .Jlonq both the supur 1or ....i~1d interior
Several revisions of these criteria have sinc e been proposed. 11est1bulc..r 1lt·c vt. b: :'nd1t -;. ,i_ Wl""Jl l as ·11\.)11 9 n-1 8 cu\'f1lear and
including the 1991 NIH criteria, the Manch ester cri teri a (see fm 1al '.wrvec.; (3ClJ 11 1 t11'; r1.1(1tr 19 r 'tutl'::> th · prtjv1ous hvporh-
Box 14.02, p . 432), the National Neurofibromarosis Foundat1n 1• es1~.· or s._;rw"• 1,1 wr•.J ·i. 191• 1 ·,t"·J -:1t tn' JlJ11 ·t1v11 oi t ermal dnd
(NNFF) criteria, the Baser criteria, and the Smith criteria Eacn pE.r1,Jr1t>rc.1l mv 1111,\l!l n 111 tr
1
(; ''l 1112\I aud1rc rv rnceitu: prrJoo...,ed

of these revisions expanded the orig inal crneri a, aim ing to aiso by f-ic.Jr·;r.>y C,•di!r -. in ·~crn~~ ,: "'S tun l' 11 s G>t:; ~! Cudlt.:~L'f't ·ce
identify patients with multiple NF2 features wr10 do not present or rrHil!ipt€ '1•Y ul(•.,,/tLr ,,1crr.:; t.:oC'h 11\·1lr1 Its O'Nrl som3t11· /\JF:J

' ' . : ~ 'S It ;. ' l ,1 11 r 1 j l t~


1 , ... , · ~ 429
A' ..
' I

Fig.14.04 Schwannoma/meningioma. A A collision tumour of schwannoma (lower half) and meningioma (upper half) is characteristic of neurofibromatos1s type 2 -,.~
meningioma component is highlighted with PR immunostaining. C The schwannoma component is highlighted with 8100 immunostaining.

mutation or deletion !744}. This explains the characteristic gross addition to having larger schwannomas , patients with IF2 r -. a, 1
and microscopic feature of multinodularity !2979) as well as have numerous small Schwann cell tumourlets on oenor- ~r::i..
the observed lower rates of surgical efficacy and higher rates nerves and spinal nerves , which despite their small size :;"c.
of surgical complications than in cases of sporadic vestibular biallelic NF2 inactivation suggestive of precursor neoo as.-:
schwannoma !584,864). In addition to the eighth cranial nerve , lesions !3030). Plexiform schwannomas are common a d ,.....a
other sensory nerves may be affected, including the fifth cra- involve large plexuses (brachia! , sacral) , skin , or subcutar2::;J-
nial nerve and spinal dorsal roots . However, motor nerves such tissues . Cutaneous plexiform schwannomas are corr~o1 ~
as the twelfth cranial nerve may also be involved (871,1941). In paediatric patients and are characteristically assoc1a ed ·N1'r 3
pigmented , plaque-like lesion 1871 ,2025,486 1.

Meningiomas
Multiple meningiomas are the second hallmark of NF2 ar' ;c-
ing half of all patients {1941) . NF2-associated mening1omas car:
occur throughout the meninges , but they are more cornr;a'l -
intracranial compartments (including along the falx cerebrn !rar
in spinal compartments (448,652}, and they may af ect s :es
such as the cerebral ventricles. NF2-associated mening.c,.....,as
occur earlier in life than sporadic meningiomas (24041 and ""'~h
be the presenting feature , especially in paediatric panems 865
871,2025).

G/iomas
Ependymomas account for mosi of the h1stologically Jag-
nosed gliomas in NF2 , and for almost all spinal gliomas 1- -os
2759). In most cases , NF2 spinal ependymomas are mu l pa
intramedullary, slow-growing , asymptomatic masses t:_ -cs
2759}. Most (70-80%) occur in the cervicomedu llar'y 1unc::0ri
or cervical spine; a minority occur in the thorac ic spire 12 '.?_
2044). Diffuse and pilocytic astrocytomas have beer repcnec
in NF2 , but they probably constitute misdiagnosed rar1cv.. ~
ependymomas \1216} .

Meningioangiomatosis
Meningioangiomatosis is a cortical lesion character .:eL1 t- 11
a plaque-like proliferation of perivascular meningotnc>lial anL1
fibroblast-like cells . It occurs both sporadically arll1 11' t=:...
Sporadic meningioangiomatosis is a single lesion that u 'L.J 1h
occurs in young adults or children, who present with sc-1z "~S
or persistent headaches . In contrast. NF2-assoc1ated mt::uwi-
gioangiomatosis may be mult1focal and often asymptomJ1 1c
diagnosed only at autopsy [3029) . Mening1oang1omatos1s tlld~·
be predominantly vascular (resembling a vascular 111dltor11 ia-
tion) or predominantly meningothelial , sometimes with an dS 0 -
ciated meningiorna, although most cases of the latter probe1bly
430 Genetic rumour Syr1ciro 1nes lil\rlJIVITl~J tile c~~s
· p'""E'5ent meningiomas with perivascular spread along Vir-
-:-~ w- R0bin spaces instead 12469).

u l al hamartias (also called microhamartomas)


These are circumscribed cellular clusters of the neocortex with
edium to large atypical nuclei . They are scattered through -
it the cortex and basal ganglia , and they show strong S100
. munoreactivity but only focal GFAP positivity. Glial hamar-
tTas are common in and pathognomonic of NF2 {2743 ,3440],
and they are not associated with mental retardation or gliomas .
The hamartias are usually intracortical, with a predilection for
;ie molecular and deeper cortical layers , but they have also
oeen observed in the basal ganglia, thalamus, cerebellum ,
and spinal cord {3440}. The fact that merlin (NF2) expression is
retained in glial hamartias suggests the possibility that haploin-
sufficiency during development underlies these malformations
{3028).

Peripheral neuropathies
Neuropathies not related to tumour masses are increasingly
recognized as a common feature of NF2 {641,1941 ). Monon-
europathies may be the presenting symptom in children 1865],
whereas progressive polyneuropathies are more common in
adults. Sural nerve biopsies from patients with NF2 suggest that
NF2 neuropathies are mostly axonal and may be due to focal
nerve compression by Schwann cell tumourlets, or that they
may be onion-bulb-like Schwann cell or perineurial cell prolif-
erations without associated axons {3011,3186).

Ophthalmological manifestations . .
Posterior lens opacities are common in children (Juvenile post~­
' tor cataract) and highly characteristic of NF2. A variet~ of ret1-
'1al abnormalities, including hamartomas , tufts, dysplas1as , and
epiretinal membranes, may also be found {509 ,1185).

Neurofibromas . . .
Cutaneous neurofibromas have been reported in pat1e_nts with
l..JF2 . However, on histological review, many such neurofibromas
prove to be schwannomas , including plexiform _schwannomas
and hybrid schwannomas/neurofibromas . Cafe-au-la1t spot~
rnay be present in patients with NF2, ~ut they ~re fewer in num
oor than in NF1 and not associated with freckling .

Epidemiology lation (8741


NF2 affects 2.5-4 individuals per 100 O00 popu ·
the C-terminu s. The pred ominant gene product, merlin (NF2),
There is no evidence of racial or sex preferen ce.
encoded by exons 1-·15 and 17, is a cytoskeletal protein with
intramolec u! ar interactions similar to those of the ERM proteins
Etiology . d by inacti va tion of (ezrin . radixin , and rnoes1n) [ 1194 ,3496]
!.JF2 1s an autosomal dominant di sease cause die occur-
the NF2 gene . About half of all NF2 cases are sporad b, newly Gtme mutaticu 1s
· t ·1 history and
r1ng in individual s with n_o ami Y
cause Y
% of de nova cases
1 50
f\lumerou qerrnline ,1r1li sorn,1t1c NF2 muta.tk)rlS ~iave been
acquired germli ne _mutat1on s. More thar enic variant is presen t detected 1r1 n8up1~1.;rn:;, supµ •r t1rig ttit:-> 11y~ uti1tJS1: triett f'JF:...1
are &omatic mosa1 ~s , where ,the path~g8 1665,3227) fu nctl 11s is ;:i tum lLH supp1 ,·_,_,01 ~l"' r e (IL:,, 10411 ~err11l1ne
oniy in some of the 1nd1v1dual s cells 18 ' NI-? rnurat1orn.) cl if lv1 c,~)fTH~\\ fh\l Tl )IT\ I

idf-•nt1fied 111 .-.pur, , 111 •,_ i1. lfll' 'lll<l;


Pathogenesis most frt' qtJt.:11! \:l -'1r11l1P,J r .L.iuti~>r1,' lit~
The NF2 gene ,., l '>7':: '.3??71 spans ~;plicc· 1urwt1on; t)r c11..; 1i. r1f' , ,\op
t hromosome 2 ,_ L ' '"' 4 •
The NF2 gene maps o c F? rnR~JA transcripts encode ::.>CJ"/ ?, ..::'/:·~,I 21t\,1 J·· l/1 C r.:d111(-' :1 'L1tJ,)11-.., ) ,, ~11 0. 1::>r•::r1'1 1'ly
11 0 kb, and consists of 17 exons N db nitl'rnative spl1c1ng m 1n cxun<; 1 8 IX}( 'l
a! !east two protein forms generate Y<
L i ~' \ l.l' ,..., • f ; : I I .,JI ·> ~' ,. ! ' ~31
Box 14.02 Clinical diagnostic criteria for neurofibromatosis type 2 (revised Manchester and ependymomas in NF2 is similar to that of non-syndrorr c
criteria) {872) tumours .
1. Bilateral vestibular schwannomas
Histopathology
OR
NF2 schwannomas may have a multilobular appeara.rce i:r
2. Family history of neurofibromatosis type 2 AND microscopic examination {3425}, which may reflect a mul er-
a. Unilateral vestibular schwannoma OR tric origin . NF2 vestibular schwannomas may entrap sever-ii
b. Any two of: meningioma, ependymoma, schwannoma, posterior subcapsular cranial nerve fibres (1431) and have higher proliferative acr .- ~J
opacities than sporadic schwannomas {117), although these fea ures d0
OR not necessarily connote more aggressive behaviour. A mosa c
3. Unilateral vestibular schwannoma AND pattern of immunostaining for SMARCB1 expression (ind1cat g
• Any two of: meningioma, ependymoma, schwannoma, posterior subcapsular patchy loss) has been reported in most syndrome-assoc1a•so
opacities schwannomas, including in both NF2 and schwannomatoss
OR {2421}. Hybrid schwannoma/neurofibroma tumours are corr-
4. Multiple meningiomas AND mon in NF2 (accounting for 30% of NF2 schwannomas) I 237
2159).
a. Unilateral vestibular schwannoma OR
All major subtypes of meningioma can occur in patJents
b. Any two of: ependymoma, schwannoma, posterior subcapsular opacities
with NF2, but the most common subtype is fibrous [116.194 1
Although many NF2-associated meningiomas are CNS Vl-iO
grade 1 tumours, a wide spectrum of tumours are encoun ered.
Gene expression including aggressive subtypes {116,2467}. Some repor s ave
The NF2 gene is expressed in most normal human tissues, characterized their growth as saltatory (760}. Collrsron tumo ~~
including brain (2734,3227) . The structural similarity of merlin of meningioma and schwannoma are characteristic of NF2
to the ERM proteins suggests that merlin links membrane-
associated proteins and the actin cytoskeleton, thus regulating Cytology
signal transmission from the extracellular environment to the cell Not relevant
12048} and influencing multiple downstream pathways, includ-
ing the MAPK , FAK/SRC , P13K/AKT, RAC/PAK/JNK , mTOR , Diagnostic molecular pathology
and WNT/p-catenin pathways . Many merlin binding partners A pathogenic variant of the NF2 gene is detected in 70-90Q
have been identified, including integrins and tyrosine receptor of patients with NF2 (and in 60% of patients with de no o ...
k1nases {1642,2180). In addition to its tumour suppressor func- most likely because of somatic mosaicism) [3359.87 1. Tre
rion at the cell membrane, merlin translocates to the nucleus risk of transmission to offspring in familial cases is 50° Rr
v1r1ere 1t suppresses the E3 ubiquitin ligase IL-17RB, which is of transmission in individuals with mosaicism is unknown Pre-
1r1volved 1n transcription activity {1879 ). natal diagnosis by mutation analysis and testing of cnildren :-it
patients with NF2 is possible when the mutation is known
Macroscopic appearance
t Jf-? >cr1t,anr1(.J,nas rna:y have a multilobular (bunch-of-grapes) Essential and desirable diagnostic criteria
ar,u ::ir~'ll e ur"' t.,oth gross and microscopic examination See Box 14.02 and Box 14.03 .
;'34/r,, r J1,..!lt1ple Schwa11n cell tumourlets may develop along
u 11rJ Ja1 r 1( r M~, particularly on spinal roots and the cauda Staging
<..;o iu, ;1 \ 1'::141,:m"m1 1!18 rJro~;s appearance of meningiomas Not relevant
!; '~ :,J o ~ I 1 ' I I \ f ' 1 "i r \ I ,{ · t J] \ ' Ji v 1 II• j rI lt I ( : f' j ~;
tox14.03 Diagnostic criteria for neurofibromatosls type 2 effect of maternal inheritance on severity has been noted , as
&l8tdfll: have families with genetic anticipation . Genotype- phenotype
Q11ca1 criteria as outlined in Box 14.02 correlations have been identified wherein truncating mutations
are associated with a more severe phenotype . whereas mis-
OR
sense mutations, large deletions , and somatic mosa1cism have
Actemoostrable germline pathological variant of the NF2 gene in addition to one of
dlnlcal criteria been associated with milder disease [211 ,878 ,1666.2077). In
addition to mutation type , the position of the mutation with in
the gene also affects the phenotype: splice-site mutation s
Prognosis and prediction upstream from exon 7 have more severe phenotypes [1.664)
TM clinical course in patients with NF2 varies widely between and mutations towards the 5' end of the gene are associated
and (to a lesser extent) within families {871 ,2025) . Some fami- with a higher risk of intracranial meningiomas [2965) . The
lies feature early-onset disease with diverse tumours and high life expectancy of people with NF2 is shorte~ed when com-
tu.mour load (V~ishart phenotype), whereas others present later, pared with lifespan in a White control population (69 years vs
with only vestibular schwannomas (Gardner phenotype). An 80 years , respectively) {3446) .

• t. ' ' "'I i •


Schwannomatos is Stemmer-RachamimrJV AO
Hulsebos TJM
Wesseilng P

Definition commonly in cranial nerves , including unilat13rall/ 1n tt"1B ,~..>


Schwannomatosis is a disorder caused by pathogenic variants tibular nerve (in patients with LZTR1 muta·1ons . schwar'"'0rr.~ 0 -
in SMARCB1 or LZTR1 (both on chromosome 22q11) and asso- sis 2). The tumours have a segmental distribution 1n ::luOJ 'Y/'~
ciated with inactivation of the NF2 gene in the tumours but not in of patients , presumed to be the result of gene ic rrs~~:-:.c;r­
the germline; it is characterized by multiple schwannomas (spi- (1967,2080 ,2966 ,2967) . Unlike in neurof1broma osrs ~ye~ 2
nal , cutaneous, and cranial) and , less commonly, meningiomas (NF2), severe chronic pain associated with the tum0l.r3 1'3 i:-.ar-
(cranial and spinal) . acteristic of the disease , and neurological deficits and pol re .
.J -

ropathy are uncommon (2080) . Patients with schwarir.omatGs.;


MIM numbering most often develop symptoms of pain . a mass. or both ,n tre
162091 Schwannomatosis 1; SWNTS1 second or third decade of life, but a formal diagnosis is usua 1
615670 Schwannomatosis 2; SWNTS2 delayed for years 12526}.

ICD-11 coding Meningiomas


LD2D .1Y Other specified neurofibromatoses Various studies have shown that SMARCB1 germline rruta-
tions also predispose indivi duals to the development of mul •o ~
Related terminology meningiomas , with the falx cerebri being a preferen11al loca.Lon
Not recommended: neurilemomatosis . for cranial meningiomas {167,592,3278) . The reported propor-
tion of patients with schwannomatosis who develop a men ""1gj-
Subtype(s) oma is 5% {2962). Occasionally, patients present with mu tio e
SMARCB1 schwannomatosis 1; LZTR1 schwannomatosis 2 meningiomas .

Localization Other tumours


Tumours associated with schwannomatosis affect the central Other tumours associated with schwannomatosis are rare. Trg
and peripheral nervous systems, with predilections for para- occurrence of malignant peripheral nerve sheath tumours r.as
spinal and peripheral nerves , as well as (less common ly) the been reported {1212,476 ,827) . Also . patients with SMARCB1-
meninges. related schwannomatosis have been described who developed
a leiomyoma, a leiomyosarcoma , or a renal cell carcinoma 1 ~
Clinical features a molecular profile similar to that of the schwannomas. as e
Schwannomas as the corresponding mosaic staining for the SMARCB 1 prote1
Patients with schwannomatosis typically have multiple schwan- {1377,827,2367,1376).
nomas that may develop in spinal nerve roots, and less
Epidemiology
Schwannomatosis was found to be almost as common as NF2.
with an estimated annual incidence of 1.25-2.5 cases per
100 000 population {118 ,2881 ).

Etiology
Schwannomatosis is often caused by mutations in SMARCB 1 or
LZTR1 , both located on chromosome 22q11 . The greac maiont
of schwannomatosis cases are sporadic , with only about s o
of patients having a positive family history (1970.2881 }. In t a
familial form, the disease displays an autosomal dominant pat-
tern of inheritance, with incomplete penetrance {19691 . Jn 2 -:.
the SMARCB1 gene on chromosome arm 22q11 was 1demr·
as a familial schwannomatosis-predispos1ng gene j 137 }, In
20 14, the LZTR1 gene was identified as a second causati ·
gene in schwannomatosis [2518) . In 2020 . the DG R gene
(which maps very close to LZTR1 on chromosome 22q 11) was
identified as the predisposing gene in a fam ily with niutttnvdul r
goitre ancl schwannomatosis [268 .11.
1 t"' lj, ,' 'I

, ;1 • J,', 11; •. i ·1· 11iJ t11j1 al I• 11 11,1 ;1 ~ 111 ·i prlll.::rit w 1i1 scilwanno111atosis .
1

,1 , , ,, ,1 , • . , .1 • , , , .• Ji.. , J , lt 1 r J':
Sternmer-Racham1mov AO
Schwannomatosis Hul sebos TJM
Wesseling P

Definition commonly in cranial nerves , including unilatera lly 1n the 'E-S-


Schwannomatosis is a disorder caused by pathogenic variants tibular nerve (in patients with LZTR1 mutations , schwanr')(T"3 0-
in SMARCB1 or LZTR1 (both on chromosome 22q11) and asso- sis 2) . The tumours have a segmental distribution _rn about 30~
ciated with inactivation of the NF2 gene in the tumours but not in of patients , presumed to be the result of genetic rnosa osm
the germline; it is characterized by multiple schwannomas (spi- (1967,2080 ,2966 ,2967) . Unlike in neurofibromatosis type 2
nal, cutaneous , and cranial) and , less commonly, meningiomas (NF2), severe chronic pain associated with the tumours is crrar-
(cran ial and spinal). acteristic of the disease, and neurological defic its and po!vneu-
ropathy are uncommon {2080) . Patients with schwannomaiOs;s
MIM numbering most often develop symptoms of pain , a mass, or both. '" tro'=
162091 Schwannomatosis 1; SWNTS1 second or th ird decade of life, but a formal diagnosis 1s usua'11
615670 Schwannomatosis 2; SWNTS2 delayed for years (2526).

ICD-11 coding Meningiomas


LD20.1Y Other specified neurofibromatoses Various studies have shown that SMARCB1 germline muta-
tions also predispose individuals to the development of multiple
Related terminology meningiomas, with the falx cerebri being a preferential locat1on
Not recommended: neurilemomatosis . for cranial meningiomas (167,592,3278) . The reported propor-
tion of patients with schwannomatosis who develop a mernng1-
Subtype(s) oma is 5% (2962). Occasionally, patients present with mul pie
SMARCB1 schwannomatosis 1; LZTR1 schwannomatosis 2 meningiomas .

Localization Other tumours


Tumours associated with schwannomatosis affect the central Other tumours associated with schwannomatosis are rare. Tha
and peripheral nervous systems, with predilections for para- occurrence of malignant peripheral nerve sheath tumours has
spinal and peripheral nerves, as well as (less commonly) the been reported (1212,476 ,827) . Also, patients with SMARC87-
meninges . related schwannomatosis have been described who developed
a leiomyoma, a leiomyosarcoma, or a renal cell carcinoma with
Clinical features a molecular profile similar to that of the schwannomas. as well
Schwannomas as the corresponding mosaic staining for the SMARCB1 protein
Patients with schwannomatosis typically have multiple schwan- {1377,827,2367,1376).
nomas that may develop in spinal nerve roots , and less
Epidemiology
Schwannomatosis was found to be almost as common as NF2
with an estimated annual incidence of 1.25-2.5 cases per
100 000 population {118 ,2881) .

Etiology
Schwannomatosis is often caused by mutations in SMARC8 1 or
LZTR1, both located on chromosome 22q11 . The great majon
of schwannomatosis cases are sporadic, with only about
of patients having a positive family history I1970,2881 ~ . In th
familial form , the disease displays an autosomal dominant pat-
tern of inheritance, with incomplete penetrance I19691. In 20 -
the SMARCB1 gene on chromosome arm 22q11 was id nbti
as a famil ial schwannomatosis-predisposing gene j13l 8 ), lfl
2014, the LZTR1 gene was identified as a second causatt"
gene in schwannomatosis 12518). In 2020, the OGCR8 gene
(which maps very close to LZTR1 on chromosome 22q 1H w·
iden tifi ed as the predisposing gene in a family with muJtinodu ar
goitre and schwannomatos1s [2681)
Flg.14.08 Schwannomas in schwannomatos1s. Tl -weighted coronal MRI showing
multiple bright, discrete peripheral tumours 1n a patient wnh schwannomatos1s.
Pathogenesis
According to the tumour suppressor gene model, both copies of Germllne Somatic
the SMARCB1 or LZTR1 gene are inactivated in the tumours of
patients with schwannomatosis . In addition , there is inactivation of
ISchwannoma 1 j
both copies (by mutation and deletion) of the NF2 gene, located ' - m ->.
distal to SMARCB1 and LZTR1 in chromosome region 22q12.2 Hit 1 Loss->22 mt · NF2 mutation
I
- ml -1.
1345,1212.2883,1213,1388,2366,2518,2966). A four-hit, three- +
step model of tumorigenesis has been proposed for schwanno-
matosis: first, an (inherited) SMARCB1 (or LZTR1) germline muta- SMARCBl •mtt+
tion occurs (hit 1); next, loss of the other chromosome 22 follows
with the ~ildtype copy. of SMARCB1 (and LZTR1) and one cop;
NF2 +tr I Schwannoma 2 I

of NF2 (hits 2 and 3); finally, a somatic mutation of the remaining NF2 mutation
copy of the NF2 gene occurs (hit 4) {2524,2883).

The SMARCB1 gene


SMARCB1 is located in chromosome region 22q11.23 and con- Fig. 14.09 The four-hit mechanism for the formation of tumours in schwannomato·
tains nine exons spanning 50 kb of genomic DNA [3318) . The sis. Schwannomatosis tumour pathogenesis involves three steps in which two genes
SMARCB1 protein is a core subunit of mammalian SWl/SNF (SMARCB2 and NF2) are lost as a result of three different hits. Different tumours will
chromatin remodelling complexes. These act as master regula- have a different NF2 mutation (som atic), but they will share the same SMARCB1 muta-
tors of transcription, using ATP for sliding the nucleosomes along tion (germline).
the DNA helix (3453) . In schwannomatosis 1, most germline
mutations in SMARCB1 are hypomorphic, non-truncating muta- of genomic DNA {2518) . It was suggested that LZTR1 may
tions, which are predicted to result in the synthesis of an altered function as a substrate adaptor in cullin-3 ub1quitin ligase com-
SMARCB1 protein with modified activity {2969). The mosaic stain- plexes, binding to cullin-3 and targeting substrates for ub1qu1t-
ing pattern seen in many schwannomatosis-associated schwan- ination [981), and it may function as a negative modulator of RAS
nomas suggests the absence of SMARCB1 protein in part of the activity (3027,2166) . The reported LZTR1 germline mutations in
tumour cells [2421). In contrast to schwannomatosis, in malignant schwannomatosis include non-truncating as well as truncating
rhabdoid tumours of the kidney and atypical teratoid/rhabdoid mutations , and they are found along the entire coding sequence
tumours of the CNS, the two copies of the SMARCB1 gene are of the gene, affecting the functionally important domains of
inactivated by a truncating mutation and deletion of the wildtype the LZTR1 protein (1388,2366,2518,2966) . lmmunostaining
gene. resulting in total loss of SMARCB1 expression in tumour of LZTR1-associated schwannomas with an LZTR 1-specif ic
cells. Because children with a rhabdoid tumour usually die before antibody demonstrates absent or reduced expression of the
the age of 3 years, familial inheritance of the predisposition is protein , consistent with its function as a tumour suppressor
extremely rare, and most cases are sporadic (2885 ,3151). A few {2366]. Germline LZTR1 mutations (but no germline NF2 muta-
families have been reported in which the affected individuals tions) were found in patients with schwannomatosis who had a
inherited a SMARCB1 mutation and developed schwannomato- unilateral vestibular schwannoma {2966 ,2964). Because these
sis or a rhabdoid tumour {476,816,3092). However, the schwan- patients phenotypically resemble patients with mosaic NF2 . this
nomas in these families (as well as the rhabdoid tumours) dis- causes the misdiagnosis with NF2 in a small number of cases
played total loss of SMARCB1 protein expression {476,3092). (1-2%) (866).

The LZTR1 gene Macroscopic appearance


The LZTR1 gene is situated proximal to SMARCB1 in chromo- Schwannomas in schwannomatosis are identic al to thei r non-
some region 22q11.21 and contains 21 exons, spanning 17 kb schwannomatosis counterparts .

: ;'. 1 1
1 r• ""' ~
Histopathology Box 14.04 Diagnostic criteria for schwa n nom~101i~
The histopathology of schwannomatosis-related tumours largely
Essential:
overlaps with that of their non -schwannomatosis co unterparts.
Most (70%) of the schwannomas associated with schwannoma- Two or more schwannomas (non-1mradermal and pa ti ~
tosis are hybrid schwannoma/neurofibroma tumours with promi - AND No bilateral vestlbular schwannomas o" higtl.qua / A
nent myxoid stroma, and they may sometimes be misdiagnosed OR
as neurofibromas or malignant peripheral nerve sheath tumours One schwannoma or meningioma
(2080) . Cutaneous schwannomas may be plexi form . AND One affected first-degree relative
lmmunohistochemically, almost all schwannomas of patients
Desirable:
with familial schwannomatosis show a mosaic pattern of stain -
Loss of heterozygosity I deletion of chromosome 22 and two drff~o/."f NF2
ing for SMARCB1 (with considerable intertumoural and intra-
tumoural heterogeneity; < 10% to > 50% immunonegative Germline SMARCB1 or LZTR1 mutation
nuclei) (2421). This mosaic staining is also frequently seen in Clinical history of pain, hybrid neurofibroma/schwannoma histology ~ •
the schwannomas of patients with sporadic schwannomatosis peripheral schwannomas
(55-100% of cases) and in most (non-vestibular) schwannomas
of patients with NF2, but it is rare in solitary, sporadic schwanno-
mas {1378 ,2421,443) . What causes this absence of SMARCB1 cannot be explained by the involvement of SMARCB 1 -:: · u -=-
staining in part of the tumour cells is unknown at present; the suggests the existence of additional causative genes 1 - : ;;,
synthesis of a mutant SMARCB1 protein is not a prerequisite Indeed, the OGCRB gene was recen tly identified as :1-e- C'o/! ~
because mosaic expression was found in the schwannomas of posing gene in a family with multinodular goitre anc s -- _,. .
patients with schwannomatosis with a germline LZTR1 mutation nomatosis (2681}. Patients with schwannomatos1s t o ca!t , - ::. ,~
(and no SMARCB1 mutation) and in those of patients lacking somatically acquired NF2 mutations in their schwan,,
germline mutations in both genes {443). no germline NF2 mutations 11436,1572,1 9691

Cytology Essential and desirable diagnostic criteria


Not relevant See Box 14.04.

Diagnostic molecular pathology Staging


In patients with schwannomatosis, SMARCB1 appears to be Not applicable
involved as a causative gene in about 50% of familial cases but
in < 10% of sporadic cases (345,1212,2735,2969). In patients Prognosis and prediction
without SMARCB1 germline mutations, LZTR1 mutations were Life expectancy in schwannomatosis is near normal (76 9 e ·si
found in about 40% of familial cases and 25% of sporadic cases it is higher than the mean life expectancy of patients .1th • f 2
{1388 ,2366 ,2966). The fact that most schwannomatosis cases (66.2 years) (866).

436 Genetic tu rn c11 1r s y mim111u~; 111vu 1v1r1u 1i1f: ( 'N~~


Von Hippel-Lindau syndrome Plate KH
Aldape KO
Neumann HPH
Vortmeyer AO
Zagzag D

oefinition
von Hippel-Lindau syndrome (VHL) Is an autosomal dominant
disorder caused by pathogenic germline variants of the VHL
tumour suppr~ssor gene (located on chromosome 3p25 .3)
and characterized by the development of haemangioblastoma
of the CNS and retina, clear c~ll renal cell carcinoma (RCC),
phaeochromocytoma, pancreatic neuroendocrine tumours , and
endolymphatic sac tumours (ELSTs).

MIM numbering
193300 Von Hippel-Lindau syndrome; VHLS

ICD-11 coding
None Fig. 14.11 Endolymphatic sac tumour in a patient with von Hippel-L1ndau syn-
drome. A This contrast-enhancing tumour (T1 -weighted postcon trast MRI) occurred
Related terminology In the cerebellopontine angle and was therefore resected by a neurosurgeon . B A CT
None of this same tumour shows destruction of the temporal bone, essentially excluding the
diagnostic consideration of choroid plexu s papilloma.
Subtype(s)
Von Hippel-Lindau syndrome types 1, 2A, 28 , and 2C peripheral nerves and even tissues outside the nervous system
\3352 ,3351 ,102,785,287,2181 ). ELSTs arise from the vestibular
Localization aqueduct and may invade through the temporal bone and into
Haemangioblastomas most often involve the retina , cerebel- the cerebellopontine angle {1118,789) .
lum, and spinal cord (especially paraspinal nerve roots) , but The sites of involvement in VHL are summarized in Table 14.02
they can occur anywhere along the craniospinal axis , including (p. 438) .

At: 14.12 Von H1ppel-Lindau syndrom e. A Transverse MRI of a 1...yst1c 'C't"tJcllcir ti:itiin_,r ·~l· ·b1o~ ;icHTid '-' 1 ' .Li -' -r "\ ~ ':~ ·' J ,1 •: ·.Jr -~ 11'-' 'l 8 S,1 y• l f\'RI 01 :1 1..., sr1c
brain stem haemangioblastoma; solid tumour sl1ows contrast er1r1,"111t.tirner·t C ' '• 11 "·i:::! ::!r ' 13 "c\.d 1',lf:li ,if'' ,.1 111 ''.i-- 1 "' _, 1l L' ''"c! 0 •L• :-111·1::'·1 1 -,~ .t::1:!~''"'1 1 .x:i 01
ography shows the tumour (arrow) with a parr of ieeder vessels E Contrast 'f1'iJrh.. ~d MRI s:, .,, ny "'U'l•P <.! '~"'· 11 ~:·, • :: ·~ . .,, ,, ~· ·1 .u ro!r -: • ·, :>~ - ::1t!•ar , ,1· 1. ~:1...1 JI
nephrectomy. F Contrast-enhanced MRI showing right adrenal phut:Ot.:IJ fnl.IL ,.,, "·' ar·: . . , G ·~· ' J ;r •. ,,. 'l ,, 1 .~··1 -.' '... . . I 1~._, ·11 .'I" ·:1 ...... J ~ ,,, L)J :1 ('-
,,

ma) (arrow). H Multiple pancreatic cysts on MRI 1 lwo pancraat11... 11t>uru0r11 J.:. ilt: 1' rr•oL..-.;",'"' ' ·" '· · ·~· 11 Ji· 1 1i-: •¥; r i y 1: 1.:1id1~· :--; 2 · ·'"" c1 ,, "·,
SCl!lllgraphy, planar anterior projection.
Table 14.02 Organ/tissue distribution and pathology of lesions in von Hippe I-Lindau
Ublquitin llgEtse a"tlVity
syndrome

Non·neopt~c
Oi:gan/tlssue Tumour(s)
111ton1

CNS Haemangioblastoma
Eye (retina) Haemangloblastoma
Kidney Clear cell renal cell carcinoma Cysts

Adrenal gland Phaeochromocytoma


HIF1a binding
Pancreas Neuroendocrine islet cell tumours Cysts A
Inner ear Endolymphatic sac tumour NORMOXIA HYPOXIA

Epididymis Papillary cystadenoma


- - - - - - - - - - - - --· - - - - - - - -

Clinical features
Retinal haemangioblastomas manifest at a mean age of 25 years
(earlier than RCC) and thus offer the possibility of an early
l
•••
HIF1a
P roteMOmal 09gradllborl

diagnosis {1813) . CNS haemangioblastomas develop mainly •••


in young adults (mean age: 29 years). They are predominantly
located in the cerebellum and next most commonly in the brain- B
stem or spinal cord {782} . Approximately 25% of all cases of Hemangiobl~loma
"Stroma" Tumor Cell
CNS haemangioblastoma are associated with hereditary VHL.
Key clinical characteristics of VHL-associated versus sporadic
haemangioblastoma are presented in Table 14.03 .
Renal cysts and clear cell RCCs are typically multifocal and
bilateral. The mean patient age at presentation is 37 years (vs
61 years for sporadic clear cell RCC), with a patient age at onset
of 16-67 years . Patients with VHL have a 70% chance of devel-
oping clear cell RCC by the age of 70 years . Metastatic RCC is
the leading cause of death from VHL.
Adrenal phaeochromocytomas arise in 20% of patients with
VHL; the mean age at onset is 30 years .
Pancreatic manifestations are predominantly multiple cysts
c
but also neuroendocrine tumours {1735). Fig. 14.13 Pathogenesis of von Hippel-Lindau syndrome. A The p oom4}(1 al
protein interacts with HIF1a, whereas the a domain interacts with o
Other VHL-associated tumours include ELSTs (associated
the VHL protein complex. In von Hippel-Undau syndrome. altera!Jons 1n
with hearing loss, tinnitus, and vertigo) and epididymal and prevent the formation of a functional VHL protein complex. I In n ,
broad ligament cystadenomas . complex binds to HlF1a and promotes ubiquitination and proteasomal r
VHL type 1 is characterized by haemangioblastomas and an oxygen-dependent manner. In hypoxia, binding of VHL protein to HIF1o .s'
RCCs (phaeochromocytomas are rare or absent), and it is resulting in the accumulation of HIF1a and increased ac!Jva1Jon ot targ 1 n ~
typically caused by VHL deletions, truncations, and missense Hlppel-Lindau syndrome, HIF1a accumulation results from tailed go Hli=1 "J •
mutations . Type 2A is characterized by haemangioblastomas the dysfunctional VHL protein complex. C HIF accumutanon in hae111311Qlt:oa:~Ta
stromal tumour cells leads to altered gene expression of H IF-r·esDonsr~
and phaeochromocytomas (RCCs are rare), and it is caused
results in increased vasculanzat1on due to angiogenesis and vascu eJ1
by missense mutations. Type 28 is characterized by a high fre- mation due to increased vascular permeability: and melabohc adaptabon
quency of haemangioblastomas, RCCs, and phaeochromocyto- ulation, extramedullary haematopoiesis, and lipid deposition (clear ceu ~mcl~"ta
mas, and it is mainly caused by missense mutations. Type 2C is
characterized by phaeochromocytomas (haemangioblastomas > 1000 mutations have been described 1n the VHL gene f--e
and RCCs are absent), and it is mainly caused by missense heterogeneous clinical manifestations of VH L are a refl :
mutations . the diversity of germline mutations.

Epidemiology Pathogenesis
VHL is estimated to have an annual incidence rate of 2.8 cases Mutational inactivation of the VHL tumour suppre r ger..:> 11
per 100 000 population. affected family members is responsible for their genet, s~­
ceptibility to tumour development at vanous organ s1 es ou· *fie
Etiology mechanisms by which the inactivation or loss of ttie sup
VHL is caused by heterozygous germline pathogenic sequence gene product (VHL protein) causes neoplastic rrans!or rr.~ n
variants of the VHL gene on chromosome 3p25 .3; these are are only partly understood [1150}. The cell ot ong1n 1nat3n d'
spread over the three exons . Missense mutations are most gioblast, stromal cell) is not well def 1ned . but current o
common , but nonsense mutations, microdeletions/insertions, points to a developmentally arrested ha~rnang -,o sc ,...re-.:.. _ -
spl ice-site mutations , and large deletions also occur. In total, sor \3350} . In accordance with the function d ~ ,..,L dS ..i ~~ - ' -

438 Genetic turr1uu1 syncJromes invo lving the CNS


::a:___ _~---------~~~
1'-03 Clinical characteristics of sporadic versus von Hippel-Lindau syndrome (VHL)-associated haemangiobla s':to:_m
VHL·assoclated
Sporadtc
Female sex 56%
41 %
patient age 23 years (range: 7--64 years)
44 years (range: 7-82 years)
tntraoranial location 73%
79%
location 75%
11%
ultiple haemangioblastomas 65%
5%

suppress?r gene, mutations are also common in sporadic Constitutive overexpre ssi o n of VEGF-A 13464.3051 ) explains
naemang1oblastomas (occurring in as many as 78% of cases) the extraord inary vascu larization of neop.lasms ass ociated
and ubiquitous in clear cell RCCs. with VHL due to increased an g iogenes1s/vasculogenes1s,
The VH.L protei~ has many different functions, and it is critically as well as the formation of cysts d ue to increased vascular
•Pvolved rn protein degradation . The a domain of the VHL pro- permeability (VEGF-A has also be en denominated "vascular
tein forms a complex with elongin B (transcription elongation permeab ility factor [VPF]") (1972) . Increased erythropo1etin
factor B. also known as TCEB2), elongin C (TCEB1), cullin-2 , expression is common in haemang ioblastomas (1737} and 1s
and RBX1; this is called VCB-CUL2 , and it has ubiquitin ligase responsible for intratumoural (extram ed ullary) haematopo1es1s
activity. thereby targeting cellular proteins for ubiquitination and and for the paraneoplastic eryth rocytos1s syndrome that can
proteasome-mediated degradation . The a domain of the gene occur in patients with VHL . HIF-depend ent downregulat1on of
involved in the binding to elongin B is frequently mutated in neo- carnitine palmitoyltransferase 1A lead s to enhanced li pid stor-
plasms associated with VHL . age , a characterist ic of VHL-depende nt tumou rs /2732) .
VHL protein plays a key role in cellular oxygen sensing , by
oolyubiquitination and proteasomal degradation of hypoxia- Macroscopic appearance
1nducible factors (HIF1 a and HIF2a) {3572,943}. which mediate The macroscopic appearance of VHL-associated tumours 1s
cellular responses to hypoxia. This leads to a loss of function similar to that of the ir sporadic counterp arts .
of VH L protein with a pseudohypoxic state characterized by
aliered expression of genes that drive vascularization , cyst for- Histopathology
mation, lipid storage, metabolic adaptation , and extramedul- The histopathology of VHL-associated tumours is sim ilar to that
lary erythropoiesis . The~ domain of VHL protein interacts with of their sporadic counterparts. Notab ly, however, ELSTs occur-
HIF1a. Binding of the hydroxylated subunit of the VHL protein ring in the cerebellopontine ang le may closely mimic choroi d
causes polyubiquitination and thereby targets HIF1a for protea- plexus papilloma; neuroimag ing is helpful in this differential
somal degradation . In the absence of functional VHL protein, because only ELSTs invade an d destroy temporal bone .
HIF1u accumulates and activates the transcription of several
hypoxia-inducible genes , including VEGFA, PDGFB, TGFA , Cytology
and EPO by binding to the respective hypoxia-responsive The cytology of VHL-associ ated tumours 1s sim il ar to th at of their
elements in the promoter region (leading to pseudohypoxia). sporadic coun terparts.

At.14.14 Endolymphatic sac tumour in a patient with vl1n H1r;pel- Lmdau syndrome Tt1e n1s1opc; tnology I:) re1111nisc"11r ut C'lk'r i,d "i~' ' .
- .. o\.J~ P<il)l 'l vni;i '.Ju• t·1- •' 1'1\'l• I "
1 t""
&ent1ally ex.eludes that diagnosis. A Low-power view B H1qh·poWB1view ' ' ' uc, ' • "') ' ~
Box 14.05 Diagnostic criteria for von Hippel- Llndau syndrome Essential and desirable diagnostic criterla
Essen tis/: See Box 14.05.
The clinical diagnosis of von Hippef.-Llndau syndrome is based on the presence
of haemangioblastoma in the CNS or retina and the presence of one of the typical Staging
extraneural tumours or a pertinent family history. By genetic testing, a VHL germllne Not relevant
variant can virtually always be identified.
Prognosis and prediction
The median life expectancy of pa ieri s 111 t"1 /Ml :-~ ~~ ,i;~·­
Diagnostic molecular pathology Clinical surveillance guidance has been p•Jb ·,.r~1 · 2~s a~-:.- -
Demonstration of a VHL germline sequence variant is desirable
to confirm the diagnosis.

440 Ger 1C!t1c turnour sync.Jr on r e~; 1 nvo1V1n~J ti 1e CNS


Tuberous sclerosis Lopes MBS
Rodriguez FJ
Santosh V
Sharma MC
Stemmer-Rachamimov AO

Definition
Tuberous sclerosis is a ~roup of autosomal dominant disorders
caused by a path?genic variant of TSC1 on 9q or rsc
2 on
16p and characterized by hamartomas and benign neoplastic
lesions that affect the CNS and various non-neural tissues .

MIM numbering
191100 Tuberous sclerosis 1; TSC1
613254 Tuberous sclerosis 2; TSC2

ICD-11 coding
LD20.2 Tuberous sclerosis

Related terminology
Fig.14.15 Tuberou s sclerosis . A T1-weighted postcontrast axial MRI demonstrates
None
two small enhanci ng subependymal nodules along the right caudate nucleus (ar-
rows). B T2-welghted coronal MRI demonstrates a minimally expans1le hypenntense
Subtype(s) lesion in the left frontal white matter compatible with a cortical/subcortical tuber (ar-
Tuberous sclerosis 1; tuberous sclerosis 2 row).

Localization
Major CNS manifestations of tuberous sclerosis include cortical
dysplasias (tubers and white matter glioneuronal hamartomas),
subependymal nodules, and subependymal giant cell astrocy-
tomas (SEGAs). Major extraneural manifestations include cuta-
neous angiofibromas, shagreen patches , subungual fibromas,
cardiac rhabdomyomas, pulmonary lymphangioleiomyomato-
sis, and renal angiomyolipomas.

Clinical features
Diagnostic criteria
The diagnosis of tuberous sclerosis is based primarily on clini-
cal features, and it may be challenging due to the considerable
variability in phenotype , patient age at symptom onset, and
Flg.14.16 Tuberous sclerosis. A Gross autopsy section from a patien t with tubarous
penetrance among mutation carriers. The diagnostic criteria for sclerosis illustrates axially cut cerebral hemispheric parenchyma with multiple sub-
tuberous sclerosis were revised in 2012 at the International Tuber- ependymal nodules in the lateral and third ventricles (arrows). B Gross image of a
ous Sclerosis Complex Consensus Conference and are based brain from a 57-year-old man showing an unusual tuber 1n the cerebellar hemisphere
on genetic testing and/or clinical manifestations {2286}. Clinical with extensive calc1fication.
manifestations are categorized as either major or minor. The diag-
nostic categories, which are based on the number of major/minor and more than half of c ard iac rhabci omyomas are associ -
manifestations present in a given individual , define disease likeli - ated with tuberou s sclerosis 126831 Cutaneous manitesta-
hood as being definite or possible 122861 (Box 14.06 , p. 444). t1ons include hypomela r1ot1c nodules, fac1,1I ang 1ot1 bronias.
Most patients have manifestations of tuberous sclerosis before and shagreen patches Ungual (or subungual) tibrumas otten
the age of 10 years, although some cases may manilesl much develoµ 1n ch1ldhoocl Renal 3.ng1omyol1porna.s develop by
later in life 137}. Confirmatory testing for TSC1 or TSC2 mutations the age ot 1.0 Ytk"lr~ 111 dS many ::u 80°{) Lt Pl'Ople with tuber-
may be helpful when a patient does not meet the clinical criteria ous ~:clerus1~~ F~81lc.ll \~y::.t~ ,m~ µ r 'S~~l)l 111 c1S rruny as 20010
for a definite diagnosis but the phenotype is cornpelling Ante- of' nflPCh'd 111d1v1rlu....1.ls. hl1t pulyl,y .t1L: ~<.idney d1st>ase 1Jnly
natal diagnosis by mutation analysis is possible when parents or occur •, ,r, .~l ~): · 1y111pt:<1n~110lt'nJrllyc1111..1t . .;1s ~~c'\/t'rt-;ly 1mp,11r5
Other family members are known to be affected 13481 lur1q lurH.1 1cJ11 mu r1 d)' Lh' 1.J k\' it 1::, t)fL ~"fl[ 111 ...1::: ni.:rny 35
40°/u ut wo1111;•1 wnl 1 tuln rotJ'; SC'lt''i , ~ 1 ,-; J\ il lht.' ptiL nc•ly(ltC
Clinical features ~eatLH•-'~ ol tl1b er 1 J•, ·; -.,, r.·~;is Ccl11 d!Sil )•'<:ur >porddicd'ly n 1

Cardiac rhab domyomas are often a presen ting featur E1 of 1nd1v1ouc.1ls W llli 0 L { tho v1: !1;-,t . ._, "j( tltl)11 I)·- t r0. ._JJ>i [!
,
b '--()'"/u
ll.[ __,, O.
t

tuberous sclerosis in newborns an d infants aged < 2 years. pat11:;r 1b w 1t i ly111pt 1a 11~ 1u1~1onw0 1 n •i•''-I" -10 n'Jl,.. 1.J•1e •uoer•;V:>
. ·
~ r • ...... ..., ., L

l_-:tt : . l !1( ' JP~ J\. •. , ' f '( 1. 1 .; ._,,_;


·' 44 1
'·'
sclerosis . Sporadic angiomyolipomas are typically so litary, small deletions and nonsense mutat1rms (8;:;r, h ;:;r,r,r,•Jr: ~ ,.1 '·,r
whereas tuberous sclerosis- associated angiomyolipomas are about 30% of all mutations 1n the gene) V1rr 1J;:;lf / ~t · ,., 1 ,!1.:'.-!".
often multiple or bilateral. result in a truncated gene produr:t arid m0r~ t~·.:jr r::j.f 'i' , ~
Neurological symptoms are among the most frequently changes affect exons 15 and 17 132861. I .::W:JP,- rJ~;c:;: ,r>,.., '>' --;
observed and serious (sometimes life -threatening) manifesta- TSC1 gene are rare .
tions of tuberous sclerosis 1654,3173) . The most common initial
signs of tuberous sclerosis are intractable epilepsy, including The TSC2 gene
infantile spasms (in 80- 90% of cases); cognitive impairment (in The TSC2 gene maps to ch romosome 16p 3 11 ~3P, . :yr] ':'J'
50%); and a combination of neurobehavioral disord ers known tains 42 exons, with exons 2- 42 encoding the ' 1.)rr, ·0 r -3 ·J·'j·~,
as tuberous sclerosis-associated neuropsychiatric disorders Gene expression: TSC2 encodes a large tran:.<:r•;:! 0 : 5 ,, 0
(TAND ; in > 60%), including autism spectrum disorder (in as which shows widespread expression in many tr3:; cs ,,.,.~ .. r! ' r
many as 40%) (1132,3173,2285). Cortical tubers and white the brain and other organs affected 1n uberous :::c1~r0s ~ •c.,:
matter glioneuronal hamartomas (see CNS manifestations, natively spliced mRNAs have been reported 134881 A r.r.--: ,,..
below) are both commonly associated with intractable epilepsy of the 180 kDa protein product tubenn has subs•ar~1=3' r 'jIT"':J
and learning difficulties in tuberous sclerosis (2286), although ogy with the catalytic domain of RAP 1GAP, a rnemb'3.. s r-
meticulous autopsy studies on small numbers of patients have RAS family.
also suggested that there may be more subtle degrees of corti- Gene mutations: The mutation spectrum of TSC2 is .v1di:;r ·r~~
cal and white matter disorganization (2010) . that of TSC1 ; it includes large deletions and m1ssense r- ~.a­
tions, and (less frequently) splice-junction mutations (76 6'32.
CNS manifestations 1492}. Exons 16, 33, and 40 have the highest number of..,., ·a-
CNS lesions in tuberous sclerosis include cerebral cortical tions . Large deletions in the TSC2 gene may extend 1 .o ·r-~
tubers, white matter glioneuronal heterotopia, subependymal adjacent PK01 gene, with a resulting phenotype o tub re ..:
hamartomatous nodules, and SEGAs. Schizencephaly, agen- sclerosis and polycystic kidney disease {310.32861 A .n€
esis of the corpus callosum, and cerebellar dysplasia are rare studies of genotype-phenotype correlations have oerror-
abnormalities. Cortical tubers can involve the cortex, subcorti- strated that TSC2 mutations are associated with a more se-.e;~
cal white matter, or both . They are both detected by CT or MRI , phenotype overall, with earlier seizure onset. a higher ri u -·
although MRI is considered the reference method for defining of tubers, and a lower cognition index. However. vrt.
CNS involvement in tuberous sclerosis (2760}. Diffusion tensor spectrum , TSC2 missense mutations are associated th
imaging and metabolic brain studies using AMT PET or FOG PET phenotypes (159,2798).
[2904,2760) in addition to intraoperative electrocorticography Like in other tumour suppressor gene syndromes. soma·."'
can identify epileptogenic tubers , facilitating tuberectomy as a inactivation of the wildtype allele (i.e. loss of heterozygos..
reasonable surgical approach to treating intractable seizures in the TSC1 or TSC2 locus) has been reported 1n kidney arc c ·-
these patients. Tubers resemble sporadic cortical dysplasias of diac lesions associated with tuberous sclerosis. as . . 11 as "
the cortex not associated with tuberous sclerosis, classified as SEGAs (512). However, there is conflicting evidence of etr::r
focal cortical dysplasia type 2b by the classification proposed a so-called second hit is required for cortical tuber foona ~
by the International League Against Epilepsy (ILAE) (305} . raising the possibility that some lesions in tuberous sc..ero- ~
may be due to haploinsufficiency 12256,3455}. Recent stu -
Epidemiology have predom inantly found activating mTOR mutations 1 •oc a;
The variability of the clinical manifestations of tuberous sclerosis cortical dysplasias, but rarely TSC1 and TSC2 mutano as
previously led to underdiagnosis. Recent data indicate that the well (179,1894}. In recent studies, loss of TSC1 1n pen"en r·c:..-
disorder affects as many as 25 000- 40 000 individuals in the lar zone neuronal stem cells was sufficient to cause abar~ar ·
USA and about 1-2 million individuals worldwide, with an esti- migration and giant cell phenotype, supportive of the r !
mated incidence of 1 case per 6000- 10 000 live births and a hypothesis for tuber formation [900 ,36061 .
population prevalence of about 1 in 20 000 (2286}. Most of the cases in which no mutation 1s iden fied 5~ d
patients with tuberous sclerosis) l662, 14921 were touna to ~
Etiology somatic mosaics when newer sequencing methods were
Tuberous sclerosis is caused by a pathogenic variant of TSC1 \3246). Mosaicism occurs when the pathogenic vanant oc~ .,~
on chromosome 9q or TSC2 on 16p. in embryogenesis, resulting in only some of the fetal ceO ·
carrying the pathogenic variants while other cell Imes
The TSC 1 gene two wildtype alleles. Clinical features of tubemus sci
The TSC1 gene maps to chromosome 9q34 1626) and con- milder in individuals with mosaicism and may ha e um1
tains 23 exons (3285), 21 of which carry coding information bution (e.g . they may involve only one organ).
(exons 3-23).
Gene expression: The TSC1-encoded protein, hamartin , has Inheritance and genetic heterogeneity
a molecular weight of 130 kDa . Hamartin is strongly expressed Most tuberous sclerosis cases (-60%) art:l spor
1n brain, kidney, and heart, all of which are tissues frequently no family history, indicating a high rate of de no" mut- ·
affected in tuberous sclerosis 12522). Its pattern of expression [2796) . Familial cases are inherited in an autosomal 001 1
overlaps with that of tuberin , the product of the TSC2 gene . fashion . In affected kindreds, the disease follows an uco :..
Gene mutations: Mutation analysis of large cohorts [530 ,32861 dominant pattern of inheritance. with high penetranc~ but corr
showed that the most common mutations in the TSC 1 gene are siderable phenotypic variability 1292 11.

442 Ge:r1 tic tun 1our syncJr ri rr1 !:'~ 11 iV<1lv1nu ti 10 CNS
Pathogenesis Macroscopic appearance
The impact of TSC1 and TSC2 sequence variants is mediated See individual tumour types within the class1f1r::::i.tion
by effects on signalling pathways involving tuberin and hamar-
tin. Tuberin, hamartin , and TBC107 form a heteromeric protein Hlstopathology
complex (known as TSC) that functions as a signalling node that Microscopically, CNS tubers consist of a d1s~rganized corte1
integrates growth factor and stress signals from the upstream with disrupted cortical lamination and conta1n1ng dysmorph1c .
P13K/AKT pathway and transmits signals downstream to coor- markedly enlarged neurons ; balloon cells (also des1gna er:J
dinate multiple cellular processes , including cell proliferation by some authors as "giant cells " {2138,20101) : dense f1bril-
and cell size (2586 ,626,1538,2523) . The complex negatively lary gliosis; calcification of blood vessel walls and_tor paren-
regulates the mTOR pathway (155 ,1033,3 158). Disruption of chyma; and myelin loss . The surrounding cortex, which usually
TSC causes upregulation of the mTOR pathway and increases appears normal in cytoarchitecture , shows changes on more
proliferation and cell growth through two effector molecules: detailed immunohistochemical and morphometric 1nvest1ga-
4E-BP1 and S6K1 (155,3158). The understanding of the basic tions [1387,2010). Oysmorphic neurons and balloon cells may
mechanism of mTOR pathway activation in tuberous sclerosis be seen in all cortical layers and in the underlying white mat-
lesions has led to the use of mTOR inhibitors in the treatment of ter. The dysmorphic neurons show alternd radial orientation in
manifestations of tuberous sclerosis . Several tuberous sclero- the cortex aberrant dendritic arborizat1on , and accumulation
sis-associated tumours (e.g . renal angiomyolipomas, SEGAs, of perikar~al fibrils. The perikaryal fibrils can be highlighted
and lymphangioleiomyomas) show a marked size reduction in using silver impregnation techniques , which show many neu-
response to treatment with mTOR inhibitors (e.g. everolimus), rons with neurofibrillary tangle-like morphology. Although the
and they regrow when treatment is stopped. mTOR inhibitors dysmorphic neurons express neuronal-associated proteins ,
were also shown to be effective in reducing seizure frequency they display cytoarchitectural features of immature or poorly
in children with refractory epilepsy (660 ,659) . differentiated neurons, such as reduced axonal projections

..

• •e..! •

Rg.14.17 Tuberous -sclerosi~. A,B Cortie-al tube; with dysmorphic enlarged neurons embedded in a densely fibrillary background. C Cortical tuber with large neuronal cells
highlighted by NeuN.

et .,, \ ' 11 l'h p tu: I' dS ;i1 1••: !-"' ' IT ltr'"' I t 1C \1 ,
1'11, 14.18 Tuberous sclerosis. A Balloon cell (with eos1noµh1l1c cytop cJ~
, ..
• · 1 I 1 t I t \:. 'i1

with a ganglion-like nucleus and strong immu11ureac11v1ty 1or lOO


Table14.04 Ma1or manifestations of tuberous sclerosis \2286) Box 14.06 Diagnostic criteria tor tuberous sclerosis (2012) \2286!

Manifestation Frequency Essential:


A. Genetic diagnostic criteria
CNS
Identification of either a TSC 1or a TSC2 pathogenic mutation tn 0
Cortical dysplasias (tubers and white matter normal tissue is sufficient to make a definitive dlagnosis of b.Jbe
- 90%
heterotoplas) according to strict criteria {1849,1850).
Subependymal nodule 80% B. Clinical diagnostic criteria
Subependymal giant cell astrocytoma 5-15% Major features:
1. Hypomelanotic macules (~ 3, at least 5 mm mdiameter)
Skin
2. Angiofibromas (~ 3) or fibrous cephalic plaque
Facial anglofibroma 75%
3. Ungual fibromas (~ 2)
Hypomelanotic macule 90%
4. Shagreen patch
Shagreen patch 50% 5. Multiple retinal hamartomas
Forehead plaque 25% 6. Cortical dysplasias (including tubers and cerebral white matter migr
Confetti skin (hypopigmented macules) 58% lines)
7. Subependymal nodules
Subungual fibroma 20%
8. Subependymal giant cell astrocytoma
Eye
9. Cardiac rhabdomyoma
Retinal hamartoma 30-50% 10. Lymphangioleiomyomatosis•
Retinal giant cell astrocytoma 2G-30% 11 . Angiomyolipomas (~ 2) 4
Retinal achromic patch 40% Minor features:
Kidney 1. Confetti-like skin lesions
2. Dental enamel pits (~ 3)
Anglomyolipoma• 80%
3. lntraoral fibromas (~ 2)
Isolated renal cyst 10-20%
4. Retinal achromic patch
Polycystic kidney cysts 2-3%
5. Multiple renal cysts
Heart 6. Non-renal hamartomas
Cardiac rhabdomyoma 50% Definite diagnosis: Two major featuresaOR one major feature with al least
minor features
Digestive system
Possible diagnosis: One major feature OR at least two minor features
Liver angiomyolipomas 10-15%
•A combination of the two major clinical features lymphangioleiomyomatosrs and an-
Lung giomyollpomas without other features does not meet the criteria for a definite dlagno
Lymphangioleiomyomatosis 3G-40% (female patients)
Pulmonary cysts 10-12% (male patients)
identical morphological phenotype express neuronal markers.
Micronodular pulmonary pneumocyte
40-S8% including connexin 26 , connex in 32, neurotilaments. class Ill
hyperplasia
~-tubulin , MAP2, and a-internexin (655 ,1317,35111 . However.
Other formation of well-defined synapses between balloon cells and
lntraoral fibroma 20-SOo/o adjacent neurons is not a consistent finding . As previously
mentioned , cortical dysplasias morphologically indistrngwsh-
Dental enamel pits Variable (up to 100%)
able from tubers may occur in chron ic focal epilepsies without
Bone cyst 40% clinical or genetic evidence of an underlying tuberous sclero-
Non-renal hamartomas Rare sis condition {301 ,3051 . The pathogenesis of these sporadic
lesions is unclear.
•May occur in other locations besides kidneys.
White matter gl ioneuronal heterotopias and radial migra-
tion lines are linear or flame -shaped bands radiating from he
periventricular zone to the subcort1cal white matter They are
and spine density {1317,1387). The other frequently observed composed of dysplastic glioneuronal cells , and they cane tePd
element in tubers and adjacent cortex and white matter is from the cortex down to the penventricutar region Sub pend~ -
the characteristi c balloon cell {1983,1317,1387) . Balloon cells mal glial nodules are elevated , often calcified nodules. The are
may resemble gemistocytic astrocytes with glassy eosino- composed of enlarged glioneuronal cells 1nd1st1ngu1shable lrom
philic cytoplasm and eccentric, often multinucleated nuclei ; those found in SEGAs, but they are smaller than those inc rt1 -
however, imm unohistochemical markers characteristic of glial cal tubers .
and neuronal phenotypes suggest a mixed glloneuronal ori- The proteins tuberin and hamartin are identifiable by 1mm -
gin of these cells . Many balloon cells express nestin mRNA nohistochemistry and western blotting 1n many rg ns nd
and protein {655) . Some balloon cells demonstrate immuno- tissu es throughout the body \1489); both proteins are wider ·
reactivity for vimentin and GFAP {1317). while others with an expressed throughout the CNS of the normal developing brain

444 Genetic tumour "'yridromes 1nvol 1ng the CN S


·y
(1488 '.3333} '. lmm~nos.t ai n ing a given tuber with anti-hamar tin
_ ...!di

or an~1-tu~e nn a nt1b~d 1es does not provide evidence of which


mutation 1s presen t 1n a given subject, and therefore is not of
diagnostic value.
I1
Extraneural manifestations
The extraneural manifestations of tuberous sclerosis and the
frequencies at wh ich they occur are summarized in Table 14.04 .

Cytology
Not relevant

Diagnostic molecular pathology


In t~ e re~i sed diagnostic criteria, demonstration of a patho- fig. 14.19 The mTOR molecular signalling pathway. The TSC genes are integral
members of the mTOR pathway, the inactivation of which leads to cell growth and
genic variant of TSC1 or TSC2 in DNA from normal tissue is survival. mTOR is modulated by the TSCH SC2 complex and regulated upstream by
an independent diagnostic criterion and is suffi cient for a several protein kinases, such as Pl3K, PDK1, AKT. and AM PK.
definitive diagnosis of tuberous sclerosis {1745) . Therefore,
genetic testing is recommended when tuberous sclerosis is Staging
suspected but cannot be clinically confirmed . On molecular Not relevant
testing , 75-90% of patients with tuberous sclerosis are fou nd
to be positive for a pathogenic genetic variant, so a negative Prognosis and prediction
result does not rule out the diagnosis {1745) . Genetic testing Tuberous sclerosis tends to shorten lifespan slightly (as com-
is recommended for family members of an affected patient, pared with lifespan in a White control population) \29031 . The
especially in babies , and may be offered as a preimplantation most common causes of death in the second decade of life are
or prenatal test. brain tumours and status epilepticus , followed by renal abnor-
malities {2286 ,99 ). In patients aged > 40 years . mortality is most
Essential and desirable diagnostic criteria commonly associated with re nal abnormalities (i.e. cystic dis-
Clinical and genetic diagnostic criteria are summarized in ease or neoplasm) or lymphangioleiomyomatosis.
Box 14.06.

1 1·ri•·1·1,·1l11r,,. 1 , 1 .- 1, . , 1 ,J'heCI~::> 445


Li- Fraumeni syndrome Orr BA
Hawkins CE
Kratz CP
Malkin 0
Solomon DA

Definition Clinical ·features


Li- Fraumeni syndrome (LFS) is an autosomal dominant disorder LFS is characterized by the early onset of a brrJr.ld :;a~rr #.-
caused by pathogenic sequence variants of the TP53 tu mour ot cancers and a high lifetime risk for cancer Breas• - ~""=!::' -:
suppressor gene on chromosome 17p13.1 and c haracterized the most common tumour in TP53 mu at1on earners (2-1 - 31 2 ...
by multiple primary neoplasms in c hildren an d adults, with a {2028 }, followe d by soft tissue sarcorras (1 6- , gc·,,1. orcin
predominance of soft tissue sarco mas, osteosarcomas, breast tumou rs (3 .5- 14%), osteosarcomas (12.6-13.4°1 ), and -dr~r,_
cancer, brain tumours , and adrenocortical carcinoma. corti cal tumours (6.5- 9.9%) (2251 ,3199,3361 However r. ~
all cancer types have been reported in TP53 muta11on carr -
MIM numbering ers , and tu mour patterns and penetrance demonstrate 15 • re:
151623 Li- Fraumeni synd rome ; LFS phases by age (93}.
Candidates for TP53 germline testing are 1dent1fied oa :: ~ -:::r"'
ICD-11 coding fulfilment of the classic LFS criteria, the Birch or Eeles er er 3
None for LFS-like syndrome (286,8261. or the 201 5 Chompre r;r, e-
ria (1869,336 } (Table 14.05). The classic clinical en ena us
Related terminology to identify individuals potentially harbouring patrogen c TP53
Not recommended: Li-Fraumen i syndrome , p53-associated. germline mutations or structural variants and thus affected o·~
LFS are: (1) occurrence of sarcoma before the age of 45 1,.ears
Subtype(s) (2) at least one first-degree relative with any tumour before r
None age of 45 years, and (3) a first- or second -degree relative 1. r,
cancer before the age of 45 years or a sarcoma at dny :ige
Localization (i869}.
Cancers of the breast, soft tissue, CNS , adrenal glands, and In general , tumours associated with a TP53 germhne mLJtato.... r
bone are the most frequent man ifestations of LFS , accounting develop earlier than their sporadic counterparts bu t t ere are
for about 80% of all tumours. The most common CNS tumour marked organ-specific differences 136071 For example adren-
manifestations of LFS are choroid plexus carcinoma, medullo- ocortical carcinoma associated with a TP53 germllne rruta 0"'
blastoma, and diffuse astrocytic gliomas . develops almost exclusively in ch ildren . in contrast ~o spora 1c

Frequency(%) p53 Frequency(%)


20 10 0 0 5 10
TP53 germ-line mutations TP53 somatic mutations

Bone

Ovary Kidney
Breast
Hemat opoieticl.3% ~l .3%
9%
system
1.8%
~
Col :--'-.
J
T125
Stomach 1.5%
2.l% Lung
Rl58
Ski n 3.l%
.____
R17S R175
3.3%

R213

G24S

R273
Tumor site distribution ...__..... ~ Tumor site distri bution of
of five hotspot TP53 R282 R282 five hotspot TP53
germ -line mutations somatic mutations
R337

'DW"*'lnl'tw_ _ _ _ • . . ,_

Fig. 14.20 Li-Fraumeni syndrome. Mutation landscape of TP53 germline and somatic mutations in human cancer. ++, lysine-rich basic C·temunal clOmam. 080, '
specific core DNA-binding domain; L, linker region; PP, proline domain; TAD, transcriptional activation domain; Tel, tetramerlzatton domain.

446 Genetic tumour syndromes 1nvolvi119 the CNS


YIMe1._D5 Chnical criteria for recommending TP53 germline testing
fdlllillll011•1 trll'·ftCOlnmendlng TP53 germllne testing Reference(s)
A proband with:

etasslc LFS criteria • a sarcoma diagnosed before the age of 45 years AND
{1 869)
• a first-degree relative with any cancer before the age of 45 years AND
• a first- or second-degree relative with any cancer before the age of 45 years or a sarcoma at any age
Birch criteria
A proband with:
• any childhood cancer or with a sarcoma, brain tumour, or adrenocortical carcinoma diagnosed before the age of
45 years AND
syndrome criteria • a first- or second-degree relative with a typical LFS cancer {sarcoma, breast cancer, brain tumour, {286,826}
adrenocorticaJ carcinoma, or leukaemia) at any age AND
• a first- or second-degree relative with any cancer before the age of 60 years
Eeles criteria
Two first- or second-degree relatives with LFS-related malignancies at any age
1. A proband with:
• a tumour belonging to the LFS tumour spectrum (soft tissue sarcoma, osteosarcoma, premenopausal breast
cancer, brain tumour, adrenocortical carcinoma, leukaemia, or bronchoalveolar lung cancer) before the age of
46 years AND
• at least one first- or second-degree relative with an LFS tumour (except breast cancer if the proband has breast
cancer) before the age of 56 years or with multiple tumours
OR
Cbompret criteria {587,337,3199}
2. A proband with multiple tumours (except multiple breast tumours) , two of which belong to the LFS tumour spectrum
and the first of which occurred before the age of 46 years
OR
3. A proband diagnosed with adrenocortical carcinoma or choroid plexus tumour, or anaplastic rhabdomyosarcoma of
embryonal subtype, irrespective of family history
OR
4. Breast cancer before the age of 31 years
----- - --- ---··-----------
LFS, Li-fraumeni syndrome.

adrenocortical carcinoma , which has a broad age distribution severity, with the most functionally severe mutations associated
with peak incidence in patients aged > 40 years {894) . Further- with early tumour onset {695] . Tumour patterns are generally
more , sonic hedgehog (SHH)-activated medulloblastoma and stable regardless of geographical or population demographics.
acute lymphoblastic leukaemia tend to occur at older ages than with the only notable exceptions being an excess of gastric can-
ao their sporadic counterparts. cers in south-eastern Asia, an excess of soft tissue sarcomas
in the western Pacifi c (695). and an excess of a low-penetrance
Nervous system neoplasms mutation at codon 334 in patients of Ashkenazi Jewish descent
In the 1245 individuals carrying a TP53 germline mutation who {2549] .
were included in the IARC TP53 Database as of July 2019,
a total of 2591 tumours were reported; 289 (11 .15%) of these Etiology
were located in the nervous system . The M:F ratio of patients LFS is caused by heterozygous germilne alterations (mutation
wrth brain tumours associated with TP53 germline mutation is rearrangem ent. or partial/complete deletion) 1n the TP53 gene
1 5·1 11394). As with sporadic brain tumours . the age of patients on chromosome 17p13.
with nervous system neoplasms associated with TP53 germ -
line mutations shows a bimodal distribution . The first incidence Pathogenesis
Peak is in children (mainly SHH -activated medulloblastomas. The p53 protein is a mult1tunct1onal tran scription factor 111volved
IDH-w1ldtype high -grade gliomas, and choroid plexus carcino- in a wide range ot b1olog1cal processos 11860) It, best ~r1ar­
rnas), and the second is in the third and fourth decacies of life acteri zed tunct1ons re 1n the control of 11-cvcle proqrt?ss1011
(mainly !DH -mutant diffuse astrocytic gl1ornas) DNA 1ntegr1ty, and t11e surviva l ot :ells e 1o::;e j tu [),-A clarn-
aging agents . Evicienc lndi ates that ~ b3 alsu rt="qul lies ot1ier
Epidemiology imp<. rtant pro<.~ ·ses , such , cell ox1dativt> tt1t:'t-c1h•) ':1111 tlh
TP53 germline variants have been est1mateo to occur .11 a rate cellular response _to nut11ent depr1vatio 11 . fernlitv. rt:rr•J1- l.i~1s
of about 1 in 500 to 1 in 5000 live births , and Tliey uL-r.ount tor and stem cell 111a111tenance The exteiit and cor' c'•1lier'1
1
'l'~ ..Jf
as many as 17% of all familial cancer cases t1 t34,2'J02,333. the b1ulog1cal respom;e el1c1ted by p53 vmy d~. ~urdii 1• 1 11J . 1 2 :.;·,:;
697.696]. There is a variant-dependent gr ad1e 1 11 or pnenotvpe a11d cell type [1566) The functions ot p5 3 rely r,,~1 : 11 1.1 r;n i1s

iun1 >LJr ~~yndron1es i!l,'ui\11 r1• J 11 .. ' -~4 7


transcriptional activity, but it can also act via interactions with was 1.55 (2331 1. Several reported farn1hes sr. r:w~r1:::. •c_--:•i. .
various proteins {2007,1727) .
~ble clustering of brain tumours 13398.1 6291, 3Jgg~:t r.J z ":(
In most human cancers, TP53 is inactivated through gene tlal for organ-specific or cell-spec1fic risk -
alterations that confer loss of the protein's tumour suppres- An .analysis of the IARC TP53 Database 1139.i) of g4=rr-1-~
sor function. Mutant p53 isoforms differ from each other in the mutations showed an association between brai11 u--~urs ,.!r d
extent to which they have lost suppressor function and in their missense mutations located in the p53 OMA-o "'d n '5 ;d~cc:i
capacity to inhibit wildtype p53 in a dominant negative manner that make contact with the minor groove of D ~A I2J2')! :- - ~
{1568,2482). In addition, some p53 mutants seem to exert onco- type of mutation was also associated with the pa ent age at
genic activity, but the molecular basis of this gain-of-function onset of brain tumours: truncating mu a ions w9re ac;-x..a•t)tj
phenotype is still unclear {1082). with early-onset brain tumours 12320.336}. Familial c.._s Fr r
may also be due to gene-environment 1nteractior.s lsr '="·qm-
Macroscopic appearance ple, exposure of families to similar enwo11rr.en·a1 care r.r:.gen >
The macroscopic appearance of tumours in LFS is similar to or lifestyle factors has been suggested 1n stomach ard hre s
that ot their sporadic counterparts. cancer \466}.

H istopathology Molecular features of CNS tumours In LFS


The most common brain tumours occurring in the setting of LFS (1) Medulloblastoma: Medulloblastomas assoc1a ed wit, LFS
are choroid plexus carcinomas, medulloblastomas, and diffuse demonstrate near uniform large cell I anaplas11c mcror-o'
astrocytic gliomas. Their histopathology is similar to that of their and belong to the sonic hedgehog (SHH)-activated . TP5~~ ­
sporadic counterparts, although the large cell I anaplastic pat- mutant molecular subtype. These tumours are charact r.zed
tern is observed more frequently in LFS-associated medullo- by chromothripsis and often have amplification of onccgenes
blastomas than in sporadic medulloblastomas (2622). including MYCN and GU2 {2622) .
(2) Diffuse astrocytic gliomas: Astrocytomas assoc.ated 't it~
lmmunophenotype LFS demonstrate distinct age-dependent molecular par erTl'i
lmmunohistochemistry can be used, albeit imperfectly, as a In the paediatric population, patients tend to present w1 h 1DH
surrogate marker of TP53 mutation, although somatic versus wildtype glioblastomas throughout the neurax1s charac er1zeo
germline mutations cannot be distinguished . The typical pattern by frequent NF1 mutations , MYCN amplification and cell-
of a missense TP53 mutation is strong, diffuse nuclear immu- cycle pathway gene alterations 12955} In young adults. 10\ 'er-
.nolabelling for p53 protein. Truncating mutations (nonsense, grade diffuse astrocytic gliomas with IDH1 mutattons loca1ed
ifameshift, or splice-site) are associated with an absence of within the cerebral hemispheres are most common. These are
nuclear staining in tumour cells. enriched for non-/OH1 p.R132H variants , including p R1 32C
and p.R132S, along with concurrent ATRX muta ·an or de eton
Cytology (3398,3070,2955).
Not relevant (3) Choroid plexus carcinoma: Choroid plexus care nomas
associated with LFS are not molecularly disunct from tne sun-
Diagnostic molecular pathology set of sporadic choroid plexus carcinomas haroot.rirg scrnanc
The TP53 gene on chromosome 17p13 has 11 exons spanning TP53 mutations. They are characterized by genomic 1ristao -
20 kb . Exon 1 is non-coding, and exons 5-8 are highly con- ity and cluster with the high-risk paediatric. type B meth 1 '. at1on
served among vertebrates. class [2507,3182).
Distribution of TP53 germline mutations: Most germline TP53
mutations are spread over exons 5-8, with major hotspots at Essential and desirable diagnostic criteria
codons 133, 175, 245, 248, 273 (all within the DNA-binding See Box 14.07.
domain) , and 337 (within the tetramerization domain). Missense
mutations are most common - accounting for > 85% of all germ- Staging
line TP53 pathogenic variants - but nonsense mutations, dele- Not clinically relevant
tions/insertions, and splice-site mutations also occur. Structural
variants with breakpoints located in intron 1 have also been Prognosis and prediction
identified in families fulfilling clinical criteria for LFS but lack- Medulloblastomas arising in the setting ot LFS are ~'OC13teo
ing TP53 point mutations 12661). Mutations observed at hotspot with a dismal prognosis {3392) . Choroid pie us care n rr.
codons consist of missense mutations that result in mutant pro- harbouring TP53 mutations , typically occumng 1n tM gerrTI •
teins with complete loss of function, dominant negative pheno- as part of LFS, are associated with a worse progn SI· t an are
types, and oncogenic (gain -of-function) activities . choroid plexus carcinomas without TP53 muta io {3097. 1 1 5!
Genotype/phenotype: Among 139 families with at least 1 case 20791 . IDH -wildtype high -grade gliomas th t ansa our 1
of brain tumour, the mean number of CNS tumours per family hood in the setting ot LFS are associated 1th a poor pr··'V"l•"'-.-.::"ee
whereas the !DH -mutant diffuse astrocyt1c glion s at '
Box 14.07 Diagnostic criteria for U-Fraumeni syndrome teenagers and young adults in the setting of LFS ar
Essential: with more favourable survival , similar to th t sociat d 1th t
Patho.genic germline alteration (mutation, rearrangement, or partiaVcomplete sporadic !DH -mutant astrocytoma counterparts !295 t '
deletion) in the TP53 gene surveillance has been associated with earty tumour oairec1rkm
and improved survival in patients with LFS l3326.11331

448 Genetic tumour syndromes 1nvo!v1ng the CNS


Cowden syndrome Eberhart CG
Eng CE

Definition polyposis {1274). In that series, 9 of ·127 patients were found


c wden syndrome is an autosomal dominant disorder caused to have colorectal cancer, all before the age of 50 years , with
mainly by germline pathogenic variants of PTEN, characterized a standardized incidence ratio of 224 (P < 0 .0001). Because
by multiple hamartomas involving tissues derived from all three of th is prospective study, the National Com prehensive Can~er
germ cell layers and a high risk of breast, thyroid, endometri al Network (NCCN) practice gu idel ines have advocated starti ng
'enal, and colon cancers . Adult-onset dysplastic cerebella ~ routine colon surveillance for patients when they are in their thir-
gangliocytoma (lherm itte-Duc los disease) is also considered ties . In an unselected series of 4 ch ildren with juvenile polyposis
to be pathognomonic. of infancy, germline 10q23 deletion also involved BMPR1A ,
upstream of PTEN. Subsequently, germ line deletion involving
MIM numbering both PTEN and BMPR1A was shown to characterize at least a
158350 Cowden syndrome 1; CWS 1 subset of juveni le polyposis of infancy (730).

ICD-11 coding Breast and thyroid cancer


LD2D.Y Other specified phakomatoses or hamartoneoplastic The two most com monly occurring cancers in Cowden syn -
syndromes drome are carcinomas of the breast an d thyroid 13019 ,3584) .
In the general population , the lifetime ri sks of breast and thy-
Related terminology roid cancers are approximately 11 % (among wome n) and 1%
Acceptable: Cowden disease; PTEN hamartoma tumour syn - (among both sexes) , respectively. In wome n with Cowden
drome. syndrome, a prospective series revealed lifeti me b reast can-
Not recommended: multiple hamartoma syndrome. cer risks of 85% with an elevated standard ized incid ence ratio
of 25.4 (31 25). The mean patient age at d iag nosis is about
Subtype(s) 10 years younger than that for breast cancer occurring in the
Bannayan-R iley-Ruvalcaba syndrome; Proteus syndrome general popu lation {1924 ,3019 ). Althoug h Rach el Cowden died
of breast cancer at 31 years of age {388 ,1918) and the young-
Localization est reported patient was aged 14 years at diagnosis \3019 ).
Dysplastic cerebe llar gangliocytoma affects the cerebel lum, the great majority of breast cance rs are d iagnosed 1n patients
and macrocephaly the cerebral hemispheres. aged > 30 years (range: 14- 65 years) {1924,3125) . The pre-
dominant histology is ductal adenocarcinoma . Most breast car-
c nnical features cinomas in Cowden synd ro me occur in a background of ductal
Dysplastic cerebellar gangliocytoma carcinoma in situ , atyp ical d uctal hyperplasia. adenosis, and
(Lhermitte-Duclos disease) sclerosis (2856) .
Adult-onset dysplastic cerebellar gangliocytoma, even in the The lifetime risk of epithelial thyroid cancer can be as high as
absence of other featu res or famil y history, is highly pred ictive 35% in patients with Cowden syndrome 13125). The youngest
of a germline pathog enic variant in PTEN {36091. and dysplastic patient reported was 8 years old at diagnosis. Based on this
cerebellar gangliocytoma is now considered pathog nomonic of and two other studies , routin e thyroid surveillance should begin
Cowden syndrome. For further details, see Oysplastic cerebel- at about 7 years of age. Histol og ically, follicular carcinoma pre-
lar gangliocytoma (Lhermitte- Duclos disease) (p . 146). dominates, althou gh p apillary histology has also been rarely
observed 11 232,1997,30 19,3584) . Medullary thyroid c arcinoma
Intestinal hamartomatous polyps and colorectal cancer has not been reported .
Several varieties of hamartomatous polyps are seen in th is syn -
drome, including hamartomas most similar to juvenile pol y ~s Other tumour types
composed of a mixture of con nective tissues normal ly present 1n A prospect ive series of individuals w 1tr1 -:owd tH1 syndr ome v-.1rh
the mucosa, lipomatous and g anglioneuromatou s lesions, and PTEN pathogenic variants 1evealed lifetirnt' m~~s or e11dometr1al
lymphoid hyperplasia 1467,3401). These polyps are found 1n the (~ 8 %). renal ce ll (30%), ·md colomc t ..tl (9q.o) c nc1nom,1s, ,1s we ll
stomach, duodenum, small bowel. and colon. The presar1ce of as rn ellm omd v ·q l312ti l
some ganglion tissue is not unusual 1n the iuveni le-l1ke polyps. Tt1A rno ~I 11 npur tarH t k r' yn tu1110,u !> 111 ' >wden S), idru1 !10
Lesions in which autonomic nerves are predominant (resulting tn are IT1t hilt.,rnrri on 1 s dr l j p , pill rn tou:; p ipi 11~' . -.J t tne s ' 1r 1
a ganglioneuroma-like appearance) have alsu been described. Benign tu i 1 ~ 1 u r •; 'tnd 01 .. ordurs ot tf 1 L)r '<.-• , t , , th
cl 1(. '" I
, l ,\ t;-. rt, Jc..'
but these seem to be exceptional j1805) A prospective study nex r rrh J S t c0111mrm u nd pr 1t1~t t 1lv ,_ 0 1 ~c·t 11 . tn~~ \.• '!llfJ >:h.'fl'
of a large series of patients with geirr.lme P7 EN pathogernc features n t tl1 8 ::;y· 1d romc' r li Jt ) u •d"t l 'I f ~ t
· '-' , 11 t::. <.l! l(j !tlll 'l'/ ,.\I 1 I'
variants found that ~ 90% of adults undergoing colono~copy ease of !he lir dt.: ' d re cur11• 11on 1,,n . 1 ( .
' ' • '> 'oJ Jo. . l\ tlt. r1 ~~· •111 Jf f '•'. < •,
1

had polyps with vari ous histological fe atures and even mixed are fol l1t,;ulctr ac1t:i n u rna~ anu n rulrl -. o , . · • •
r c ~ u 1 11 ~J u1tr ' •J' r. •t.' tr , ,
1

I ,,
,·J -
Chron ic lymphocytic thyroiditi s is a c ommon finding accompa-
which apparently interac:ts with the rrrfr;r,r r,;~' '"'.r ,. , ••
nying thyroid pathology in Cowd en syndrome with PTEN mu ta- cal impact is still unclear 12581 ) ·
tions .
PTEN is vir tually ubiqu1touc:;ly ay.....,.
p rP" t:t;)d 1-,r.Jr,~.,, ,_. -
·::>-> ~ 'J ,_.~ ,•
Other malignancies and benign tumours have also been studies of expres sion 1n human dev~10 p rr~• ' r1~ 11 ~ - , ~ ::: ·;:. , .:
reported in patients or fam ilies with Cowden syndrome . It formed , and only a single study has '!Ya m11~rj o- t:r .:;:, .,,-· N
remains to be determined whether these (e.g . sarcomas, lym - during human embryogenes1s using .:3 rr.r; ..- -:_c,0 .... a -::,... r
phomas , leukaemias , and meningiomas) are true components against the term inal 100 amino acids of P HJ ~ .. r - - - ·
of the syndrome. revealed high levels of expression of PTEr ~r'jf~ ; · -~ -., :
thyroid , and CNS - organs that are affecred .. -y · - ~ : r:. - _ - ;;y-
Epidemiology neoplasms of Cowden syndrom e It also re·1ec ".!'1 p·- - --
Before the identification of PTEN, the incidence of Cowden syn- expression in the developing autonomic ner r: J5 J' :icr- ~- '""
drome was estimated to be 1 case per 1 mill ion person-years gas trointestinal tract. Early embryonic dea 1n Prt;-r- - 'T - '=" --= _:
{3019) . After the gene was identified (1889), a molecular-based implies a crucial rol e for PTEN 1n early develop En· 7~ ,.. .:- ~ -
estimate of prevalence in the same population was 1 case per 3081 ). PTEN is a tumour suppressor and a dua'- 5P"'=': · r · , r _
200 000 population {2227) . Because of difficulties in recogniz- phosphatase that plays multiple roles 1n th e cell -: ·-= :!~ .-
ing th is syndrome , prevalence figures are likely to be underes- tosis , cell polarity, cell migration. and even geno" c : ·:::::: -
timates . One recent study estimated de novo PTEN mutation 12185,2901 ,3584}. The major substrate o PTEN s D ='J
frequency to be about 11 % at minimum and 48% at maximum in is part of the P13K pathway (665 .101 3.1871 1980 3C15
tested probands (2087}. PTEN is ample and functional. PIP3 is converted to PIP2
results in hypophosphoryl ated AKT. a known cell -s..;r. J3 ~ =
Etiology tor. Hypophosphorylated AKT 1s apoptotic. W en PTE'-1 c;
Approximately 85% of Cowden syndrome cases , as strictly cytoplasm , it predominantly signals via rts lipid c hcs;:;r ·"'t ~
defined by the International Cowden Consortium (ICC) cri- activity down the P13K/AKT pathway 121201 l'l cor :-ra-· .. -s--
teria , have a germline pathogenic variant in PTEN, including PTEN is in the nucleus. it predominantly signals 1a c- ·== •
intragenic mutations, promoter mutations, and large deletions/ phosphatase activity down the cyclin 01 I MAPK patri.-.a · ~ c -
rearrangements {1889,2018,3610} . If the diagnostic criteria are iting G 1 arrest at least in breast and gliai cells 110 3 · 1..l • '
relaxed , this mutation frequency drops to 10-50% {1961,2229, 2120) . It is also thoug ht that PTEN can dephosoror113 e ;:,... .
3232} . A formal study that ascertained 64 unrelated Cowden and inhibit integrin and MAPK signalllng 11 173.31 2 11
syndrome-like cases found a mutation frequency of 2% if the Bannayan-Riley-Ruvalcaba syndrome. wh1cn is :'larav -
criteria were not met, even if the diagnosis was made short of ized by macrocephaly, lipomatos1s. haemang1orna·os s
on ly one criterion {2019} . However, this study only looked at speckled penis, was previously thought to be clir.1ca 1,
the nine exons of PTEN; presumably, further mutations would tin ct, but it is now considered a likel y allelic variant or C: J , ••e ·•
have been identified in the promoter or in SOHB or SOHO. A syndrome {2020) . In a combined cohort of 16 soorad•c ")(" :!
single-centre study involving 37 unrelated families affected 27 familial cases , approximately 60% of the patter.ts _:::i.rr ec -
by Cowden syndrome (as strictly defined by the ICC criteria) germline pathogenic variant in PTEN (2021 I Of the 27 r.:cr a
fou nd a pathogenic variant frequency of 80% {2018}. Explora- cases studied , 11 were classified as exhib1tmg true OJe' a
tory genotype-phenotype analyses showed that the presence Cowden syndrome and Bannayan-Riley-Ruvalcaba s, c~
of a germ line pathogenic variant was associated with a familial and 10 of those 11 had a PTEN mutatmn . Another .... ~ .. _
risk of developing breast cancer [2018). Additionally, missense patients with Bannayan-Riley- Ruvalcaba synaroma ~-er e ~ . . . ... ·
mutations and/or mutations of the phosphatase core motif seem sequently found to harbour large germllne ae1encris G' =-=
to be associated with a surrogate for disease severity (mul - {3610). The overlapping mutation spec rum, the e '::>!er.:::: _•
tiorgan involvement) . A small study of 13 families with 8 PTEN true overlap between familial cases , and genorype- o'le'"'.:::: -
mutation-positi ve members did not show any genotype-phe- associations suggest that a germllne PTEN path ge •,c \ 3 .:: ,...
notype associations {2227 ). but this may be due to the small is associated with cancer. and they strongly suggest '":::1..: · ~~
sample size. two syndromes are allelic and part of a single spectr._
Recently, other Cowden syndrome predisposition genes molecular level. The aggregate term "PTEN na .• ano ~ .:1 -
have also been identified : the SDH genes , PIK3CA. KLLN, and syndrome" was first proposed in 1999 1202 I ana li.:JS : -
WWP1 . become even more apt, now that germline PTE1 C':3 ., ,c .::
variants have been identified 1n autism spec ru -.J ..., u
Pathogenesis macrocephaly, in Proteus syndrome , and 1n \ TE
PTEN, on 10q23, consists of 9 exon s spanning 120- 150 kb association (the c o-occurrence of several birth dere !~
of genomic distance, and it encodes a 1.2 kb transcript and macroc ephaly \427,2629.36081 In one ca 'e. tht> 1d"'1
a 403 amino acid and lipid dual-specificity phosphatase of a germline intragen1c PTEN mutation 1n a pat1d1H ' • " ! · -'
(dephosphorylating both protein an d lipid substrates), which is have juvenile polypos1s 123211 was subsequamlv .: ~ · -
homologous to the focal adhesion molecules tensin an d auxi lin to exc lude that spec ific clinical i 1agnos1s. the t1no119 1 '
l1872 ,2018,3023J. The amino acid sequence that is homolo- suggests a molecular designation of PTEN narn..ifh..r
gous to tensi n and auxilin is encoded by exon s 1- 6. A clas- syndrome 1850,1358 .1359.1764 .20-2.23291 fh 1~
sic phosphatase core motif is encoded within exon 5, which is has been further supported by the 1dent1f1 anon .,t ~ r •
the largest exon, con stituting 20% of the coding region (1 868, PTEN pathogenic variants 1n 1nd1v1duals with 1u 11 i.:
1872, 30231 . A longer isoform of PTEN has also been described , and of large deletions involving botn P TEN and - .
iuvenile polyposis of infancy 1730,3090). An impo1 tant fi11ciinw of . n Consortium (ICC) 3nrl N;:itlonal (,r;mprP.h~n~ 1vc:
Box 14.08 lnternat1onr1I Cowcle. 'teria for Cowd'3n syndrom'3 without knrJwn
the polyp ascer.tainment study was that the reasons for reforral Cancer Network (NCCN) operationa 1 en ·
hsted in the original ~athology report s were often inco rrecl , sug- family history of PTEN mutr1!1on
aesting that a re -review o.f all polyp histology by ga strointestinal Pathognomonlc criteria:
Ciathologists based in ma1or academic medical centres is a vit al · tom a (cerebellar tumours)
Adult dysplastic cerebellar ganghocy
~-tep for determining correct genetic etiology 13090).
Mucocutaneous lesions 0 •

Macroscopic appearance
. tnch1lemmomas,
• Facial . . anYnumber~ (at least two biopsy-proven tnchilem-
momasb)
0 •splastic cerebellar gangliocytomas, the main CNS lesions • Acral keratoses
associated with Cowden syndrome, are discrete lesions char-
acterized by hypertrophy of cerebellar gyri. • Papillomatous papules
Mucosal lesions (especially hamartomatous gastrointestinal polyps)
Hlstopathology Autism spectrum disorder and macrocephaJyb
o -splastic cerebellar gangliocytoma shows diffuse enlarge- Major criteria:
nent of the molecular and internal granular layers by ganglionic Breast cancer
. of various size, with relative preservation of the overall cer-
Thyroid cancer (non-medullary)
et>eHar architecture 111 ).
Macrocephaly (megalocephaly; i.e. 97th percentile and above)
Cytology Endometrial cancer
T~ cytology varies by tumour type. Mucocutaneous lesionsb
• One biopsy-proven trichilemmoma
Diagnostic molecular pathology • Multiple palmoplantar keratoses
Cowden syndrome is an autosomal dominant disorder, with • Multifocal cutaneous facial papules
age-related penetrance and variable expression {849,2255 , • Macular pigmentation of the glans penis
3125). The major Cowden syndrome susceptibility gene, PTEN,
Multiple gastrointestinal hamartomas or ganglloneuromasb
is located on 10q23.3 {1872,1889,2228) . Other predisposition
oenes in non-PTEN Cowden syndrome include the SDH genes •ICC criteria only, bNCCN 2010 criteria only (m?dified.from (3124,ll . .
Note: In 1996, the ICC (2228} compiled operational d1agn.ost1c cntena for Cowden
as well as PIK3CA, AKT1, KLLN, USF3, SEC23B, and WWP1 syndrome on the basis of the published literature and t~e1r own ?hnic~I expe~1ence .
1241 ,2248,2250,2330,2249,3534,1 843). (848,3584}. NCCN has also established a set of operational clinical d1agnos1Jc cntena
for identifying individuals with possible Cowden syndrome (2216} .
Essential and desirable diagnostic criteria
Pathognomonic and major diag nostic criteria are listed in Prognosis and prediction . . .
Sox 14.08. There have been no systematic studies to 1nd1cate wheth~r t~e
prognosis for patients with Cowden syndron:e and cancer is dif-
Staging ferent from that of non-syndromic patients with the same cancer
Stagin g varies by tumour type. types.

\ . l l •' {It [ A f JI S) f ) { ) 1 I( ~ , Ij I j ,; t ; ~
Constitutional mismatch repair Tabon U
Abedalth;:;r:f3t1~ -
deficiency syndrome Leg1uc; E
SolrJmon OP.

Definition
Constitutional mismatch repair deficiency syndrome (CMMRD)
is an autosomal recessive cancer predisposition syndrome
caused by biallelic germline mutations in one of four mismatch
repair genes (MLH1 , PMS2, MSH2, and MSH6) . Individuals with
CMMRD develop ultrahypermutated malignant gliomas, CNS
embryonal tumours , and a variety of other cancers during chil d-
hood and early adulthood .

MIM numbering
276300 Mismatch repair cancer syn drome 1; MMRCS1

ICD-11 coding
None
Fig. 14.21 Representative imaging of replication r9oa1r-'.!eL _- ~ _.,.
Related terminology mas. A T1 -weighted postcontrast MAI of a patient w11h co:ish ut•onaJ ,...,sr- • ·-::cc
Not recommended: mismatc h repair cancer syndrome; Turcot deficiency syndrome with a homozygous MSH2 patrn>genit ger,.... .e ,ara.-
syndrome; brain tumour polyposis syndrome type 1. two synchronous tumours (arrows). Both tumours had secondary ~ -
Acceptable: biall elic mism atch repair deficiency syndrome. and ultrahypermutation, one involving POLE and one 1rrvo '"9 POLD I ~ :: _
sequence of a patient with Lynch syndrome and MLHt tieterozygcus .
line variant. This gliomatosis-like pattern is typical for somatic murator .. -
Subtype(s)
hypermutation.
None

Localization Other CNS manifestations


The glioblastomas arisin g in th e setti ng of CMMRD, Lync h Agenesis of the corpus callosum and venous anQn"\~ - - '"'.._
syndrome, and polymerase proofreading defi ciency can occur been re ported in ch ildren with CMMRD Oevetoprne ·-
both in the cerebral hemi sp heres and in the posterior fossa, anomalies are extremely common and may point to
incl uding a gliomatosis-like dissem ination pattern in a subset possibility of CMMRD {291 7}.
of cases. Medulloblastomas centred in the posterior fossa , and
other embryonal CNS tumou rs, have also been reported 11 78 }. Extraneural manifestations
More than 90% of patients with CMMAO ha e cate-a..i-·- ·...,., -
Clinical features ules and other dermatological abnormallt1es su - . as
The combination of cafe -au -lait skin macules, consanguin- pigmented or hypopigmented areas 11781 Tnes ~are-.:j - -
ity, and specific brain , haematological , and gastrointestinal macules can mimic those found 1n panenrs 1tn '"'e~ •
cancers arisin g during childhood shou ld raise suspicion for tosis type 1. However, patients with CMMRD ry a·
CMMR D. A scoring system has been developed for determining other stigmata of neu rofibromarosis type 1. sue~ a a;
those patients in whom genetic testing for CMM RD should be groin freckling, c utaneous neurofibromas. ana Li' ·h •1.:
performed 13457). Importantly, family history of cancer is often {3457,813}. Haematological malignancies, pre ...._.:: •
uninformati ve, especially for those children with biallelic PMS2 leukaemia/lymphoma, occur in as many as 30
mutations, because of the substantially lower cancer risk asso- mostly in the first two decades ot life. wh reas gasrr
ciated with heterozygous mutations in this gene compared with polyposis and cancers are present 1n v1rtuall all
the other mismatch repai r genes . the second decade of life. Other can -ers (e.g unn
cancers and sarcomas) have also been re rt ·1
Nervous system neoplasms Multiple prlomatrixomas appear to b frequent n m1gn
Brain tumours, most common ly gliomas, occ ur in the first two gest CMMRO when present in combination with an ! l f
decades of life and account for 25- 40% of all CMMR D-assoc i- of the condition {576}. An increased frequenc or aed
ated cancers [178 ,3457) . Medu ll obl astoma and CNS embryo- system ic lupus erythematosus has been reported in
nal tumours have also been described in pati ents with CMMRD with CMMRO 1321 0); all 5 patients with CMMRD an P t<lU.
[1183}. Molecularly, all brain tumours arising in the setting of systemic lupus erythematosus were girls, and 4 ot them tia
CMMRD have a unique ultrahypermutation genotype, which b1allelic MSH6 mutations.
distinguishes them from the typically low somatic mutation bur-
dens in thei r sporadic paediatric counterpart s [335 ,447).

452 Genetic tumour syndromes involving tl1e CNS


[)itferen tiaf diagnosis
"'MMRD should not be confused with Lynch syndrome (also
1o..n0wn as l1ered1tary no~polyposis colorectal cancer syn-
t rorne) Whereas CMMRD 1~ an autosomal recessive syndrome

resulting from b1allelic germllne mutation in one of th e mismatch


repair genes .. Lynch syndrome is an autosomal dominant syn-
drome . resul~1ng from a heterozygous germline mutation. It
results 1n a different can.c er ~pectrum and different age of onset
(mostly colorectal , genitourinary, and sebaceous carcinomas
ouring adulthood). Hypermutant cancers including glioblasto-
A
mas. accompanied by some of the features of CMMRD includ- Hyptrmulilllon
ing cafe-au-lait macules , can also occur with germline muta-
f, ns in the POLE gene encoding DNA polymerase E which has
t>een termed "polymerase proofreading deficiency" {3267,114).

Epidemiology MMRD r1
ore than 200 kindr.e ds with CMMRD have been reported {178 , ~r
3457}. However, this syndrome is probably underdiagnosed
and highly prevalent in populations where consanguinity is B
'gh {96,811). In countries with a low level of consanguinity, the
Hy~rmut<1Uon
valence of this condition has been estimated at 1 case per
million children.

Etiology
CMMRD is caused by biallelic germline inactivation of one of mutant POLE

the four main mismatch repair genes: MLH1 at chromosome r V"i.-r


3p22.2. MSH2 at 2p21-p16.3, MSH6 at 2p16 , and PMS2 at
7p22. This can be the result either of two different mutations c
present in trans (compound heterozygous) or of the same muta-
tion present on both alleles (homozygous), the latter being com-
mon in consanguinity.

Pathogenesis
The genetic defect underlying CMMRD is the inability to rec-
ognize and repair DNA mismatches during replication . Rec-
ognition and repair of base-pair mismatches in human DNA is
mediated by heterodimers of MSH2 and MSH6, which form a D
sliding clamp on DNA. The C-terminus of PMS2 interacts with Flg.14.22 A model of replication repair deficiency and the consequent mutation ac-
MLH1. and this complex binds to MSH2/MSH6 heterodimers cumulation. A Replication repair in normal cells. B Mismatch repair deficiency from
to form a functional strand-specific mismatch recognition com- mutation in one on the four mismatch repair genes leads to hypermutation. C Muta-
plex 12946). Cells that are deficient in any of the above genes tions in the proofreading domain of the DNA polymerases POLE or POLD1overwhelm
are defective in the repair of mismatched bases and insertions/ the mismatch repair system and result in hypermutat1on. D Combined mismatch re-
deletions of single nucleotides, resulting in high mutation rates pair deficiency and polymerase mutations result in ultrahypermutation.
and microsatellite instability. Unlike in heterozygous carriers with
Lynch syndrome (in whom microsatellite instability is robustly and MSH2 mutations are most prevalent in Lynch syndrome.
observed in the resultant endometrial and colorectal cancers). PMS2 and MSH6 mutations predominate in CMMRD Heterozy-
glioblastomas arising in patients with CMMRD often lack clas- gous PMS2 mutation carrier parents are usually unaffected due
sic microsatellite instability and are characterized instead by to the substantiall y lower cance r risks The MLH1 and MSH2
extremely high rates of single-nucleotide mutations with a sig- group tends to have a younger age of first malignancy diagnosis
nificantly smaller component of small insertions/deletions (178 , and a more severe overal l cancer phenotype (3457).
29201. CMMRD-associated glioblastomas commonly acquire All tumour types are observed among spec1f1c CMMRD muta-
mutations in POLE or POLD1 to create complete replication tion carriers Some stu :11es suggest tnat brain tumours are rnore
deficiency and ultrahypermutation 1178,29201 . These tumours frequent 1n patients With biallel1c Ptv!S2 than 1n tl1o::>e with AIU-11
almost invariantl y inactivate tumour suppressor genes sucll as or MSH2 muratiuns. w1lt1 tl1" f\!1LH1 an(i MSH:? group more tre-
TP53 quentlv tiav1ng hat:rndl<..:lo~_i1cc-11mali!:Jnanc1t'S134571.

Genotype/phenotype . Macroscopic appear nee


In CMMRD, the genotype/phenotype correlation is ~ 1f f 1 c ul t to rhe macro•;copi 3U~i8cttdrlCf? of turrtdllfS !I~ --~Jr,1R['' 1:- -"''

ascertain due to the syndrome's rarity Whde:-...is germl 1r1e ML f-1 1 described tc1r tl1P 1nd1v1~Jt:al tu1111ur tyros
Fig. 14.23 Glioblastoma arising in the setting of constitutional mismatch repair deficiency syndrome.
pleomorphism with bizarre and multinucleated giant cells.

Histopathology
The glioblastomas arising in the setting of er 11::1C
severe nuclear pleomorph1sm and/or bizarre
giant cells (1183). Other brain tumours 1n C iARO ~v-~, -~-
phologically as sheets of primitive small blue ~e le, ·::.
differential diagnosis of a CNS embryonal tumo Jr Jr :r ...
blastoma depending on the location f178 1 831
brain tumours that arise in the setting of CMMRD ar
gl ioblastomas or whether true medulloblas omas o _r rr
xanthoastrocytomas , and other tumour types can
syndrome remains to be determined .
The finding of a paediatric high-grade glioma Jr qlio
with severe pleomorphism or giant cell feaures n )
suspicion for possible CMMRD and prompt 1m nu• o
cal testing for the mismatch repair proteins

Cytology
Not relevant
Fig. 14.24 Glioblastoma arising in the setting of constitutional mismatch repair defi-
ciency syndrome (CMMRD). This glioblastoma in the cerebral hemispheres of a child Diagnostic molecular pathology
with CMMRD demonstrates a complete absence of PMS2 protein both in tumour cells Detection of biallelic germline mutation (either horr:
and in normal cells, resulting from biallelic inactivation of the PMS2 mismatch repair compound heterozygous) in one of the four main m1
gene in the germline of this patient. genes is required for the diagnosis of CMMRD The
of variants of unknown significance and the tecri.
with sequencing PMS2. which has multiple pse ao
Box 14.09 Diagnostic criteria for constitutional mismatch repair deficiency syndrome
led to the development of several functional as -,.
Essential: aid in the rapid detection of CMMRD Microsaic · ~ -
Biallelic pathogenic germline mutation/deletion in one of the four main mismatch ity testing on glioblastomas is not a reltable esr Di:! ~-~ •
repair genes (MSH2, MSH6, PMS2, MLH1) typically demonstrate only a low level of m1c rosata 1• t_
OR ity despite being mismatch repa1r- dehc1ent afld u'tTo
A combination of the presence of two clinical criteria and positive results in two tated . lmmunohistochemistry demonstrates loss o• =
functional assays (see text) of the inactivated mismatch repair protein (and ·'1e.1 a ~
Desirable: ate, its heterodimer) 1n both tumour and norm I u ::iu- 1
Genomic profiling of the index brain tumour demonstrating an ultrahypermutated of CM MAD-associated cancers 1178). In vitro ce !-....
genotype with mutation signature characteristic of mismatch repair deficiency on normal fibroblasts and lymphoblasts can .:rec
Absence of expression of mismatch repair proteins in both tumour cells and normal ellite instability, resistance to several compounus
cells on immunohistochemistry to repair G- T mismatches 12920,30dl Rec dntl , ..: ·· -
tests based on microsatellite 1nstab11tty using r1~-.t q . -

454 Genetic tumuur s y11cimr110 ~~ rnvu l v 1n ~J llin CNS


s quenc 1ng of normal tissue repo t d .
, ~ ~ successful 1dent1fication
of patient s with CMMRD
11026 11 3 5 e8rly detection ml'.\y result 1n 1nUP.8<>ed s 1Jr ·11'18I fr1r brirl·· r)1~11~1.,-_
and heterozygous carrier s 1812 329'1! Th8 rnt"•.::rc:nr r8',1 ..., t8r · r.~
Essential and desirable diagnostic c 't . of mismatch repair- deficient cells to sev~ral rfJmmrin r:h<:?rr,r°J-
See Box 14.09. rr erra therapies, including temozolom1de , mal-res thP.m 1nr;ffAr,t11c:: .,
the management of gl1omas 1n the setting of CMMRO In r:rJn-
Staging trast, the ultrahypermutation phenotype of CMMRD -assr;r,1.:jfl'!rJ
Not applicable cancers results in a greater neoantigen burden 0n r:anr:er sells
which can be therapeutically exploited by immune checkpoint
Prognosis and prediction blockade {2920,1816) . Because the prognosis for children Ntth
Patients with CMMRD and their famil . glioblastornas arising in the setting of CMMRD 1s unfavourabf~
genetic counselling , because surv . Y members bene'.tt from imrnunotherapeutic approaches and prevention strategies are
81 11 ance protocols exist and
now being tested {335.8 .1183).

~ I
Familial adenomatous polyposis 1 Varlet P
Abedal thagaf 1MS
Pf ,..,t~r '}../•
Pii::t-:r:.h r
Ellison OW SolrJrrir.ir [Jfr.
Hawkins CE Tabori lJ
Leg ius E

Defi nition congenital retinal pigment epithelial hypertrophy eo d~r,.. .-:-•


Famili al adenomatous polyposis 1 (FAP1) is an autosomal cysts , dental abnormalities , and a high risk of de1elop nJ sit··-:·
dominant cancer syndrome caused by an inactivating germ- colonic tumours such as osteomas (50-90%), desrno1d -Jr"i·J'~
line pathogenic sequence variant in the tumour supp ressor (10-15%), thyroid cancers (2- 3%) , and hepatoblastorri:3 11~,
gene APC. In FAP1, gastrointestinal neoplasms pre dominate. {1855] . Therefore, FAP1 is a multiorga n cancer pred1spcc;. :.-::,..,
A subset of patients with FAP1 develop a primary brain tumour, syndrome. Timely diagnosis and awareness will help ''.l ean·1
which is also referred to as brain tumour po lyposis syn drome 2 detec tion of other cancers and appropriate interve tions ·
(BTP2). The principal brain tumour in BTP2 is medulloblastoma Medullob lastoma represents the only pnmary brain t~rno #,
with WNT activation . c learly associated with BTP2 13392,16161. No difference , 3 g~
at diagnosis has been found between patients with a syra ornic
MIM numbering medulloblastoma due to germline APC mutations and sooraaic
175100 Familial adenomatous poly posis 1; FAP1 medulloblastoma with somatic CTNNB1 mutations !3076 3392 1
Colonic symptoms may be mild or even absent and medullob'as-
ICD-11 coding toma can be the initial tu mour, predating manifestations or tre
2890.Y & XH1 CV4 Other specifi ed mal ignant neoplasms of polyposis syndrome. In this setting . fam ily history can be sugges-
colon & Adenomatous polyposis coli tive of FAP1 . The recommendation is therefore to care ully invest.
gate family history and offer genetic counselling for patients w 'h
Related terminology CTNNB1 wildtype WNT-activated medulloblastomas .
Not recommended: Turcot syn drome. Occasional FAP1-associated cases of ad amantinomato u~
craniopharyngioma that lack CTNNB1 mutation and instead
Subtype(s) harbour germline APC mutation with somatic loss ot heteroz,-
None gosity have been reported 12413).

Localization Epidemiology
For WNT-activated medulloblastoma arising in the setting of Only a small proportion of WNT-activated medulloblastcmas
BTP2, no differences in location (cerebellar midl ine I cerebel- are familial (approximately 5% of cases) . and these are so rar
lopontine angle) have been foun d from that of their sporadic exclusively due to germli ne APC pathogen ic variants. Medullo-
counterparts with somatic CTNNB1 mutations (3076}. blastomas are a rare manifestation of FAP1 , accounting tor 011
1% of all malignancies in FAP1 patients (1855]. FAP 1 occurs
Clinical features in 1- 3 per 10 000 births , with an almost 100% penetrance Ir
FAP1 is a common gastrointestinal polyposis syndrome and 20- 30% of cases , the disease is caused by a de nova m !at1ori
is characterized by the development of multiple adenomas in with no clin ical or genetic evidence of FAP1 1n the parents or
the colon and rectum , predisposin g to colorectal carc inoma. family {1391 }.
More than 70% of patients also develop multiple extracolonic The lifetime risk of developing a medulloblastoma 1n tne con-
manifestations, including gastric and d uodenal adenomas, text of FAP1 is about 1%, which is 92 times as high as that in tre
general population \1 581.

Etiology
FAP1 re sults from a heterozygous paihogenic variant 1n the
APC tu mour suppressor gene , located on chromosome ban
5q22 .2. A second hit (additional somatic mutation or dele •01 ·
loss of heterozygosity) in the APC gene is require for tt.. 1 \,; 1
form ation 11225).

Pathogenesis
A PC acts as a negative regulator of the WNT signalhng p t
playing an important role in ubiqu1tinat1on and degr
f3-catenin . APC loss leads to a nuclear translo t1on t ~ ~~
that impacts proliferation, differentiation , and m1gr· 1~ " 1
The activation of the WNT signalling p thway b APC
Fig. 14.25 Medulloblastoma in brain tumour polyposis syndrome 2. Classic medul- similar to that caused by common mutant oncog 1r' ~ -..:d
loblastorna with Homer Wright rosettes. proteins occurring in sporadic WNT-activated m dull 1

456 Genetic tumour syncJ rorri,;s irwolvmq ll it; Cl\J~~


BoX14.10 Diagnostic criteria for a familial adenomatous polyposis 1- · d . f .. h uld be offered genetic counselling and
brain tumour associate an d t he1r am1 11es s o
. e pathogenic variants because . the fre-
teste d for germ I1n . . .
Eslentfal: . APC mutations 1s very high 1n this setting
quency of germ I1ne
Qa:urrence of a brain tumour, typically WNT-activated medulloblastorna in a patient (3392).
wlh famffial adenomatous polyposis 1 '
OR Essential and desirable diagnostic criteria
l!ientiftea~n of a pathogenic germline mutation In the APC gene in a child with See Box 14.10.
WNT-actiVa.ted medutloblastoma lacklng CTNNB1 mutation
Staging . · I d · I
Staging involves cerebrospinal fluid cytolo~y, ~rania an spina
Macroscopic appearance MRI to exclud e metastasis, and postoperative 1mag1ng to evalu-
Not relevant ate gross residual disease.

Histopathology Prognosis and prediction . . .


In a series of 12 medulloblastomas in patients with BTP2, all had The overall and progression-free survival of patients with FAP1
a classic histology, including frequent Homer Wright rosettes with WNT-activated medul loblastoma is excellent (similar to that
and nuclear p-catenin accumulation (3076}. reported for sporadic WNT-activated medulloblastomas) (841 .
3076,3392}. Given the re latively low prevalence of CNS tumours
Cytology in patients with FAP1 . imag in g-based screening is not recom-
Not relevant mended for ch ildren of these families. However, identification of
a germline APC pathogen ic variant is key in initiating early sur-
Diagnostic molecular pathology veillance for gastrointesti nal and other FAP1-related neoplasms.
WNT-activated medulloblastomas without CTNNB1 mutation The guidelines are well established and include surveillance
should be tested for APC pathogen ic variants. These patients during ch ildhood (1 39 1}.

( ' :..··-'
Pietsr,~1 1
Naevoid basal cell carcinorna syndrom e F- ~Jer h2rr r,r;
!:lliYJn f JW
Evans 0(,R
Pa1tler K'N

Definition NBCCS-associated medulloblastoma


Naevoid basal cell carcinoma syndrome (NBCCS), also known In a review of 33 repor ted medullobla5tcrr.::, r • .:::-: ,_. ~-- ,. ::. .-4

as Gorlin syndrome, is a complex syndrome involving multiple with NBCCS, all but 1 tumour had de1e r ) a.rl .... - .. rl · -
organ systems . It is caused by germline mutations in genes < 5 years , and 22 c ases (66%) had aric;P,:- r• ~ • ~- - ;,~~
involved in the hedgehog signalling pathway (most commonly < 2 years 1100). Medulloblastornas asso:.1..j e~ ,, · , ~· -
PTCH1). The most common CNS finding is medulloblastoma of seem to be exclusively the extensively nad•J =:r or '1~ - - - --:.
the desmoplastic/nodular subtype . tic/nodular types (100,1042,2855.29631 Ii ha<: o*~ :;·-::. -
that nodular/desmoplast1c medulloblastomas rn ·r Jr "
MIM numbering should serve as a major criterion for the d1agr _ ~ .c;
109400 Basal cell naevus syndrome; BCNS (100,1042).

ICD-11 coding Epidemiology


LD20.4 Gorlin syndrome A UK prevalence of 3.2 cases per 100 000 ind
birth incidence of 5.3 cases per 100 000 b1r~hs .-a
Related terminology reported (869), although lower prevalence rates 'l,a: _
Acceptable: Gorlin syndrome; Gorlin-Goltz syndrome. reported from Italy (0.39 cases per 100 000 popu1a ,..
Not recommended: basal cell naevus syndrome; fifth phakoma- Japan (0.42 cases per 100 000 population) 19931 J~:: ~
tosis . (0.61 cases per 100 000 population) (28911. or
131 .:hil .·er ....
medulloblastomas. 6% had germline SUFU mutai nr;3 3 ·
Subtype(s) About 1-2% of patients with NBCCS with germltf'le PT['""' ::..,.,t·
None genie variants develop medulloblastoma, comoare • :r: 1 _
20% of patients with germline SUFU pathogenic vana ,·s 12 ~
Localization
Manifestations of NBCCS occur in the skin (basal cell carci- Etiology
nomas, sebaceous cysts), jaw (keratocysts), bone (congenital NBCCS results from inactivating germline mutatio~s n • ~
anomalies), and brain (macrocephaly, falx calcification, medul- human homologue of the Drosophila segment po1ar1~ ca·r· " ....
loblastoma) , as well as in the eyelid , mouth (cleft lip/palate), and gene (PTCH1) on 9q22 (1217,1491), its paralog PTCh2 or. .. o_
ovary (fibromas) . in rare cases {887,9941. or SUFU on 10q24 129631.
Numerous different PTCH1 germline pathogenic var a-·s a.;_-
Clinical features ciated with NBCCS have been reported 1993.19041 ::i.r ......;..g
The most frequent manifestations of NBCCS are multiple basal pathogenic variants are only detected in abour 50-60a..o ,. - -
cell carcinomas , as well as odontogenic keratocysts of the jaw. (2017}. However, the detection rate of specific pathog~ ' c ·.~ -
In one study, basal cell carcinomas and odontogenic keratocysts ants has increased significantly (with as many as 93'" O' : -
were found together in > 90% of affected individuals by the age found positive in one study in recent years) because or ·ri: ae-.-::-·
of 40 years {873}. Other frequent manifestations include calcifi- opment of improved methods of detection 1993). Ttia m
cation of the falx cerebri, palmar and plantar pits, and bifid or truncating) mutations are distributed over the ennre PTCi...; ....c; _-
fused ribs {1631 ,2891). These findings (apart from rib anomalies) ing region , with no mutation hotspots [3433) . M1ssen~~ m .... r~ c. s
are considered major diagnostic criteria (see Box 14.11). Minor cluster in a highly conserved region (the stero1-sens1no ..Jo rn- •
criteria include medulloblastoma {100}, ovarian fibroma, macro- and particularly in transmembrane domain 4. SVFU p .... -
cephaly, congenital facial abnormalities (e.g . cleft lip or palate), variants are only detected in approximately 5-6°"a iJ' r-
skeletal abnormalities (e.g. digit polydactyly), and radiological probands , and they are mostly truncating 129631
bone abnormalities (e.g . bridging of the sella turcica) (100,1631 ). The rate of new PTCH1 pathogenic variants nas '1Ct -
The clinical features manifest at different points in life. Macro - precisely determined . It has been estimatea mac 3 h1~" ' 1 ... -
cephaly and rib anomalies can be detected at birth, and medul- tion (14-81%) of cases are the result of new path en.: .
loblastomas typically develop within the first 3 years of life. Jaw (873,1144,2891 ,3432} . In 4 of 6 cases of NBCCS v'-lttn :i -
cysts do not usually become evident before the age of about SUFU pathogenic variant . the mutation was 1nh tel.
8 years , and basal cell carcinomas are usually found 1O years unaffected , healthy parent. In the otn8r 2 cases tne
later (993) . Radiation treatment of patients with NBCCS (e.g . was new (2963} .
craniospinal irradiation for the treatment of cerebellar medullo-
blastoma) induces multiple basal cell carcinomas of the skin as Pathogenesis
well as various other tumour types (e.g. meningioma) within the The PTCH1 gene encodes a 12-tranSfrodmbr:.in~ i.: - -~
radiation field {511 ,2324 ,3021 ). expressed on many progenitor cell types . It tLnc,. :> ..J

458 Genetic tumour syr1rl ro1118 f:: 1nvolv11ici thp CNS


lllllt'-11 Diagnostic criteria for naevoid basal cell carcinoma syndrome
Macroscopic appearance
crltvrla:
Not relevant
r (sheet-like) calcification of the falx
tocyst Histopathology .
OT plantar pits (~ 2) Practically all medulloblastomas associated with NBCCS have
basal cell carcinomas (> 5 in a lifetime) or a basal cell · · the morphological features of desmoplastic tumou rs . including
aged < 30 years carcinoma in a the desmoplast1c/nodular medulloblastoma and the medullo-
-degree relative with naevoid basal cell carcinoma syndrome
blastoma with extensive nodularity (2855 ,100,1042,391 .2963)
MiJOr criteria:
Cytology
iOOOd medulloblastoma•
Not relevant
. omesenteric or pleural cysts
~haly (occipitofrontal circumference> 97th percentile) Diagnostic molecular pathology
or palate A comprehensive mutation analysis of the PTCH1 and SUFU
•ettet>ral or rib anomalies genes by DNA sequencing along with a search for deletions/
duplications by appropriate methods (e .g. multiplex l1gat1on-
Preaxial or postaxial polydactyly
dependent probe amplification, targeted array-based analysis)
- or fused ribs
can identify most NBCCS-related germline pathogenic variants
<Nanan or cardiac tibromas {867). To detect more complex alterations , additional analysis
Orular anomalies on the transcript level may be necessary in some cases . The
Bequlrements for diagnosis: detection rate of specific pathogenic variants has increased
significantly thanks to improved methods of detection [9931 .
~r diagnostic criteria and one minor diagnostic criterion
Still , in 15-27% of index cases , neither PTCH1 nor SUFU patho-
OR
genic variants can be identified {876) .
major and three minor diagnostic criteria Because germline pathogenic variants are frequent in children
OR younger than 5 years with extensive nodular or desmoplastic/
- n of a heterozygous germnne PTCH1 or SUFU pathogenic variant on nodular medulloblastomas, human genetic counselling should
genetic testing and supporting clinical criteria be offered to the families , and early pathogenic variant analy-
blastoma with sonic hedgehog activation, mostly desmoplastic/nodular or sis of PTCH1 and SUFU should be performed with appropriate
~e nodular types. methods . The detection of a germline condition is important for
planning appropriate treatment that avoids radiation therapy
{1042,391 ,2963 ,1182,3392).
receptor for members of the hedgehog protein family of secreted
s:gnalling molecules (sonic hedgehog , IHH , and DHH ) (2014, Essential and desirable diagnostic criteria
3C43l PTCH1 controls another transmembrane protein , SMO The major and minor diagnostic criteria are listed in Box ·14.11 .
161 .3043). In the absence of its ligand , PTCH1 inhibits the activ-
~Y of SMO {61,3043}. Hedgehog signalling takes place in the Staging
primary cilium (157}. Binding of hedgehog proteins to PTCH1 Not relevant
can relieve its inhibition of SMO , allowing its translocation to
ne tip of the primary cilium , which results in the activation and Prognosis and prediction
rranslocation of GLI transcription factors into the cell nucleus The prognosis of NBCCS-associated medulloblastomas seems to
a'ld the transcription of a set of specific target genes control- be better than that of sporadic cases , and 1t has been suggested
ling the survival, differentiation , and proliferation of progenitor that therapy protocols be adjusted in patients aged < 5 years with
cells. In vertebrates , this pathway is critically involved in the NBCCS, to prevent the formation of radiation -induced secondary
development of various tissues and organ systems, such as tumours (100,3021). A recent retrospective review of patients with
lmos, gonads. bone, and the CNS {1140,1412). SUFU is located medulloblastoma with a SUFU germline pathogenic variant, how-
downstream in the hedgehog pathway. SUFU has been found ever. indicated a worse prognosis (11821. In another retrospective
\0 directly interact with GLI proteins , and it is a negative regula - series, no difference in survival was found between patients ''"ith
trJr of hedgehog signalling {3044) . PTCH1 and SUFU are classic medulloblastoma carrying a PTCl-11 pathogenic variant versus
tumour suppressor genes; the second allele of the mutant gene those carrying a SUFU pathogenic variant 133921
is lost in most NBCCS-related tumours . Inactivation of PTCH1 or Surveillance guidelines are well established and 1nclulie sur-
SUFU leads to the pathological activation of the sonic hedge - veillance during childhood [968) For earners of the SUFU path-
1og signalling pathway. In cases where PTCH1 and SUFU are ogenic variant. these include a recomrnendm1on TQr repedteLi
trndtype, a much less common source of a Gorl1n -like syndrome brain MRI screening for rnedullobl··1~hJ11u LH::>Vt'loµment w 1tt11n
to consider is that caused by pathogenic germline GPR161 vari- th e f 1rst 5 years of life (968)
a'lts 1232).

.,,
I\.... I I ~: I I j ..... I' ' ,-J .. j j ; + j\
' '
-1~9
Rh abdoid tumour predi sposition syndrome .Jur:lhns AR
Biegel JA
Eberr1;:Jrt (,G
Huang A
KoolM
Wesselrng P

Definition different location 19611. SMARCB1 muWi0 ,.. m~ 1 ,_,,. _. --


Rhabdoid tumour predisposition syndrome (RTPS) is a disor- ally underlie the oncogenes1s of other neopl.g~rr:- >· r - <:
der characterized by a markedly increased risk of developing proximal type of epithelioid sarcoma 12 39!. O·~ - ·~ a.; -::
malignant rhabdoid tumours (MRTs), including atypical teratoid/ sarcomas have not been described 1n R PS s~....... -~
rhabdoid tumours (AT/RTs), due to constitutional loss or inacti- somatic mutations in SMARCA4 also give r se to 1rr~ "/.!!.• ,,. ,_
vation of SMARCB1 (in the syndrome subtype RTPS1), or rarely cinoma of the ovary, hypercalcaemrc ype - a r~re ~ .J - -
SMARCA4 (in RTPS2). aggressive tumour in adolescents and young adt..'•s :t:,7 ~ - 3
The incidence of germline mutations is high 1 h;::)~,..=~ - _
MIM numbering type small cell carcinoma of the ovary. and 1n o e ~a..... ,
609322 Rhabdoid tumour predisposition syndrome 1; RTPS1 SMARCA4 germline mutation . AT/RT and ovana., sa,..-.:~#
613325 Rhabdoid tumour predisposition syndrome 2; RTPS2 diagnosed in a newborn and his mother. respect 1e• 1

ICD-11 coding Epidemiology


None AT/RTs generally occur in early childhood. bu t .ey s.~ c_o.::t-
sionally found in adults 12605}. RTPS is found 1 25-35·;) "J
Related terminology patients with AT/RT (273,816}; these patients are rr''J'e J'~ . •c;.
Not recommended: rhabdoid predisposition syndrome; familial present in their first year of life. Second primary ....1"'1o 'S :r
posterior fossa brain tumour syndrome of infancy. arise as late as 8 years after the primary diagno"'1s 26S
gesting that lifetime surveillance is required.
Subtype(s)
Rhabdoid tumour predisposition syndrome 1; rhabdoid tumour Etiology
predisposition syndrome 2 RTPS1 and RTPS2 are caused by germline SMARCB1
SMARCA4 mutations. respectively. De novo germl1 ne ;r _!a.
Localization can occur during oogenesis/spermatogenesis. or pos: g -
Both the nervous system and extraneural organs/tissues can cally during the early stages of embryogenesrs (mos c a "'
be involved. 1254,1453,2885}. Tumours in rare families with mult pie a ec-
children and no parental SMARCB ·t mutation are o ooa.oi 1
Clinical features to gonadal mosaicism . In some families , apparen ly u""'a4 ae.
The median age at tumour onset is younger in patients with carriers develop schwannomas in the fourth or fifth decade.
RTPS than in patients with sporadic tumours . To date, no other SMARCB1 has also been identified as a predis s
clinical features have been identified that distinguish RTPS- in fam ilial schwannomatosis, germline mutations 1n S
associated tumours from sporadic rhabdoid tumours. as predisposing in hypercalcaemic type small cell car~
of the ovary, and germline mutations in both S CB
Nervous system SMARCA4 have been identified to contribute to s no"
Individuals with RTPS1 often present with isolated AT/RTs or an such as Coffin-Siris syndrome. associated with
AT/RT with a synchronous renal or extrarenal MRT {1832}. features and intellectual disability.
In the past, a variety of other CNS tumours have been
reported to be associated with RTPS, including choroid plexus Pathogenesis
carcinoma 11069), medulloblastoma, and supratentorial primi- The SMARCB1 gene
tive neuroectodermal tumours (2885). However, because these The SMARCB1 gene was the first subunit of the SW SNF
tumours may be hard to distinguish from AT/RTs, and because plex found to be mutated in cancer. This gene is o ~affid
some AT/RTs lack well-developed rhabdoid cells, whether such chromosome region 22q 11. 23 and contains nine 0 . s s
tumours occur in RTPS is controversial 11208,1509,1511,3255). ning 50 kb of genomic DNA l3318I. Alternati'Ve s ~ r
exon 2 results in two transcripts and two proteins ith !
Extraneural manifestations of 385 and 376 amino acid residues , respectively Ti
By far the most common extraneural manifestation is MRT of homology domain in the second half of the pro in na
the kidney. Bilateral renal MRTs are almost always associated highly conserved structural motifs through wi11ch S RC l
with a germline SMARCB1 mutation, but infants with an isolated interacts with other proteins l3042}. The SMARCB 1 prot n
MRT may carry germline mutations as well. MRTs have been a core subunit of mammalian SWl/SNF chromatin re · tny
reported to originate in the head and neck region , paraspinal complexes, and it regulates gene expresoon via ATP-m&dtat
soft tissues, heart, mediastinum , and liver 134241. Children nucleosomal remodelling {3453}. The SMARCB1 protein func-
surviving a primary MRT can develop a second primary in a tions as a tumour suppressor via repression of CCND1 gene

460 Genetic tumour sy11drum e .~ irrvulvrng th ·CNS


~' ress1on . induction of the CDKN2A gene and hy h h -d predisposition syndrome
. . · pop osp o- Box14.12 Diagnos1ic criteria for rhabdo1 tumour
;a l()ri of retrnoblastoma protein , resulting in GO/G 1 cell-cycle
=~st 1262,2685] ..Loss of SMARC81 leads to activation of Essential:
. f r: SMARCBt or sMARCA4 mutation in a patient wrth
t.:H2. a h1stone lysine methyltra~sfe.rase and catalyt ic compo - Demonstration o a germirne . abd01·d t (AT/AT)
malignant rhabdoid tumour (MAT) or atypteaJ teratoid/rh umour
~t of the PRC2 complex , resulting 1n increased H3 K2 8
·? 3) k . . p. me 3
" ... me mar H.
s associated with repression of polyc
· .
b
om gene Desirable:
:argets. Th e 1ppo s1gnall1ng pathway is involved in the detri- Multiple MATs or AT!ATs
TS\tal _effects of SMARCB1 deficiency, and its main effector Siblings or other relatives with MAT or AT/RT
l P1) 1s overexpressed in AT/RTs {1462,3454] .
.fARCB1_is a classic tumour suppressor requiring biallelic
of func'.1on, with the second event typically being loss of Macroscopic appearance . .
,.:erozyg~s1ty I d~letion . The types of SMARCB1 mutations The macroscopic appearance of tumours 1n RTPS is similar to
observed 1n sp?rad1~ MRTs are similar to those observed in the that of' the ir sporad ic counterparts.
ger fine mutations 1n RTPS . However, single base deletions in
exon 9 ~cur ~o~t often in sporadic AT/RTs {273 ,816,3092]. The Histopathology . . . .
second inactivating event is most frequently a deletion of the The histopathology of tumours in RTPS 1s s1m1lar to that of their
type allele. often due to monosomy 22. Schwannomatosis sporadic counterparts.
mutations _are significantly more likely than sporadic mutations to
occur at_either end of the gene and to be non-truncating (2969]. Cytology .
Germhne SMARCB1 mutations may predispose individuals to The cytology of tumours in RTPS is similar to that of their spo-
the development of rhabdoid tumours or schwannomatosis. In radic counterparts.
rhab?oid tumours, the two copies of the SMARCB1 gene are
ctrvated by a truncating mutation and deletion of the wildtype Diagnostic molecular pathology
gene. resulting in the total loss of SMARCB1 expression in Loss of immunohistochemical sta ining for SMARC B1 (IN11) or
tumour cells. This is in contrast to the non-truncating SMARCB1 SMARCA4 (BRG1) expression all ows for the identification of
mutations and mosaic SMARCB1 expression in schwannomas MRTs and AT/RTs assoc iated with RTPS 1 and RTPS2 , respec -
patients with schwannomatosis {2969}. Because of the high tively. However, molecular genetic testin g of the patient 1s
mortality and morbidity associated with rhabdoid tumours , famil- needed to confirm a suspected germline alteration .
ial inhertta.nce of RTPS is extremely rare {2885,3151}. It has been
reported that as many as 35% of patients with a rhabdoid tumour Essential and desirable diagnostic criteria
carry a germline SMARCB1 alteration as the first hit (338,816}. See Box 14.12.
However, a recent study demonstrated a lower incidence, sug-
gesting that prior studies may have been biased by the inclusion Staging
of patients with multiple primary tumours, virtually all of whom Not relevant
have a germline SMARCB1 alteration (2630] . A few families
have been reported wherein affected individuals inherited a Prognosis and prediction
SMARCB1 mutation and developed either schwannomatosis or Transcriptome and DNA methylation p rofiling separate AT/RTs
2 rhabdoid tumour {476 ,816,3092}. In these families, both the into three molecular groups , which by consensus have been
rhabdoid tumours and schwannomas displayed total SMARCB1 designated "AT/ RT-TYR ", "AT/ RT-SHH ", and "AT/RT-MYC "
1oss 1476,3092} . Recently, a patient with co-occurrence of a (1320) . Although these subg roups show differences in patient
rhabdoid tumour and schwannomas was reported /1584) . age and in localization , no pattern of germline predisposition by
subgroup has been confirmed to date .
The SMARCA4 gene Because only a relatively small number of patients with
The SMARCA4 gene. located on chromosome 19p13.2 and RTPS have been reported in prospective therapeutic studies ,
encoding a catalytic subunit of the SWl/SNF complex, was the the impact of germline alterations on patient outcome remains
second member of this complex reported in a cancer predis- unclear. Al though earlier data suggested a poorer outcome
DOSit1on syndrome (274 ,2853} . More recently, other SWl/SNF for patients with SMARCB1 germline alterations, an increasing
SJJbunit genes have also been implicated in cancers . Collec- number of AT/RT survivors with germl1ne SN/ARCB1 alterations
!ivety, 20% of all human cancers contain a SWl/SNF mutation, are being seen, suggesting that other variables such as inten-
and because most of these tumours are not classic MRTs, the sity or type of therapy, as well as specific SMARCB 1 genotypes.
definition of RTPS may need adjustment in the future {1624). may influence outcome.

l Jl 'I l .' 1, .< ' ' ,


I t~ • .+ul
Legius E
Carney complex Folpe AL
.Jo VY
Reuss DE

Definition
Carn ey complex (C NC) is nn autosomal dominant syndrome
characterized by my omas , endocrinopathy, and pigmented
skin lesions : t11e main nervous system manifestation is malig -
nant melanolic nerve sheath tumou r. In > 70% of patients a
heterozygous inactivatin g pathoge nic va riant is detected in the
PRKAR1A gene coding for the type 1a regu latory (R1a) subunit
of PKA .

MIM numbering
160980 Carney complex, type 1; CNC1
605244 Carney complex , type 2; CNC 2 Fig. 14.26 Carney complex. A Lentigines on the lower eyelid. B Lentig1nes on rhe r

ICD-11 coding rare form of myxoma found in bone . A re cent study showed ver-
2F7A.O Multiple polyglandular tumours tebral nodular lesions on MR I in 31 .6% of patients (8 59) .
Malignant melanotic nerve sheath tumour is a rare but poten-
Related terminology tially lethal compl ication occurring in 8- 10% of ad ults [3049, •
Not recommended: LAMB syndrome (lentigines, atrial myxoma , 256,859} . Gastro intesti nal tract and the paraspinal sympathetic
mucocutaneous myoma, blue naevus) ; NAME syndrome chain are most freque ntly involved.
(naevi , atrial myxomas , myxoid neurofibromas , and ephelides); Endocrine neoplasms are characteristic for CNC . The most fre-
myxoma, spotty pigmentation, and endocrine overactivity. quent endocrine tumour is primary pigmented nodular ad reno-
cortical disease (PPNAD), whi ch frequently results in an ACTH-
Subtype(s) independent hypercortisolism (Cushing syndrome). PPNAD 1s
Isolated primary pigmented nodular adrenocortical disease reported in 26- 58% of patients in cohorts with CNC {256 ,3049,
caused by a specific splice mutation; severe Carney complex 859} . A recent study diagnosed PPNAD in 57% of patients , and
caused by a chromosomal microdeletion in an additional 11 .4% there was possible PPNAD {859). Another
study of 353 individuals found PPNAD in 58% of patients with a
Localization proven PRKAR1A pathogenic variant. The frequency was higher
Manifestations of CNC arise in the skin, endocrine organs in female patients (71 %; median age at onset: 30 years) than in • '
(adrenal cortex, thyroid , pituitary, testis , ovaries), peripheral and male patients (29%; median age at onset: 46 years) (256 ). Mul-
central nervous systems , heart, bone, breast, and (to a lesser tiple thyroid nodules were seen in 5-28% of patients 12 56 , 304~
extent) pancreas. 859} . Sometimes a thyroid papil lary or follicular carcinoma is
present. A somatotroph pituitar y adenoma I pituitary neuroen-
Clinical features docrine tumour (PitNET) is diagnosed in 10-18% of adul ts (256,
CNC was reported for the first time in 1985 as a complex of 3049,859] . Large cell calcifyi ng Sertoli cell tumours are reported
myxomas , spotty pigmentation, and endocrine overactivity (471}. in 33- 49% of male patients; they can be hormone-producing
Multiple lentigines might be present at birth (3049} but the typi- and are mostly benign (256,3049 ,859). They are more frequent in
cal appearance usually develops around puberty. Lentigines are male patients with a proven PRKA R 1A pathogenic variant (256J
predominantly localized on the lips, conjunctiva, eyelids, ears , than in those with CNC without a pathogen ic variant.
and external genitalia. Other pigmentation abnormalities such as Conditions associ ated with Carney comp lex are li sted in
hypopigmented macules, blue naevi, epithelioid blue naevi , and Box 14.13.
cafe -au-lait spots develop in early childhood . Skin and mucosal
myxomas are seen in 10-30% of patients {2034,256,859). Myxo- Epidemiology
mas of the breast are often bilateral and are seen in as many CNC is a rare autosom al dominant d isease known to affect
as 25% of women with CNC {256,3049,859}. Cardiac myxomas > 700 individuals worldwide (3050 }
can occur in childhood . The prevalence of cardiac myxomas
in different cohorts ranges from 22% to 53% (3049,256,859). Etiology
Recurrence after surgery was observed in 62% in one study CNC is caused by heterozygou s inactivating pathogenic vari -
{859}. Patients with cardiac myxomas present with symptoms of ants in the PRKAR1A gene in an estimated 70~'o o t patients ful-
systemic embolism , heart failure, or intracardiac obstruction of filling diagnostic crite ria [3049 , 1 639,256.~793) n1e p 8 netrance
blood flow. Myxomas can affect any chamber of the heart, ~nd of CNC caused by PRKAR1A pathogenic variai1ts is estimated
continued surveillance is advocated. Osteochondromyxoma is a to be virtu ally 100% (3049} .

462 GGr 18t1c tu1 r1our ~Y ' 1drornes 1nvolv1ng the CNS
Pathogenesis
Staging
!JAKAR1A codes for the R1a subunit of PKA. PKA is a hetero- Not relevant
rerramer ~1th two regulatory and two catalytic subunits. There
are four 1soforms of th~ regulatory subunits (R1a , R1p, R2a, Prognosis and prediction
R2~) an.d of the catalytic subunits (Ca, cp, Cy, PRKX) (3156) . CNC is an autosomal dominant disorder; affected parents har-
G-protein coupled receptors bound to a ligand will activate bour a 50% risk of disease transm ission with each pregnancy
actenylyl cyclase ~nd the synthesis of cAMP, which binds to the Genetic counselling is recommended . and it i n.elude~ the pos-
regulatory subunits of PKA . This allows the catalytic subunits sibility of prenatal and/or preimplantation genetic testi ng
dissociate ~nd p~osphorylate downstream targets . lnactivat- The average life expectancy is 50 ye~rs because of. excess
-~ pathogenic variants of PRKAR1A will result in an overacti- mortality related to cardiac myxoma , ~al1.gnant melanot1c. nerve
vanon of the ?AMP/PKA pathway. This pathway is essential in sheath tumour, postoperative compilcat1ons , and a variety of
many endocn~e cell types . Inactivation of the wildtype allele carcinomas [3049,3050).
CNG-assoc1ated tumours has been demonstrated , reflecting
e tu~our-suppressor function of the R1a protein (1639). Some Box 14.13 Conditions associated with Carney complex (3049}
mutations are not associated with nonsense-mediated RNA Intense freckling
cJecay, ~nd it ha.s b.een suggested that a specific splice vari- Blue naevus
ant leading to sk1pp1ng of exon 6 might have a dominant nega-
Cafe-au-lait spots
ve effect and that this is sufficient for tumorigenesis in CNC-
Elevated IGF1 levels, abnormal oral glucose tolerance test, or paradoxical growth
attected tissues [1171}, but the evidence for this is incomplete. honnone responses to TRH testing in the absence of clinical acromegaly

Macroscopic appearance Cardiomyopathy


The macroscopic appearance of CNC-associated tumours is Pilonldal sinus
similar to that of their sporadic counterparts . Cushing syndrome
Acromegaly
Histopathology Sudden death in extended family
The histopathology of CNC-associated tumours is similar to that Multiple skin tags
of their sporadic counterparts . Lipomas
Colonic polyps
Cytology
Hyperprolactinaemia
The cytology of CNC-associated tumours is sim ilar to that of
their sporadic counterparts. Thyroid nodule
Family history of carcinoma (especially of the thyroid, colon, pancreas, or ovary)
Diagnostic molecular pathology
Amutation in the PRKAR1A gene is identified in 70% of patients Box 14.14 Diagnostic criteria for Carney complex; diagnosis of Carney complex
fulfilli ng diagnostic criteria for CNC (3049,1639,256,2793 , requires two major criteri a or one major criterion plus one supplemental criterion (3049}
3321). Many types of mutations have been found , such as mis- Major diagnostic criteria:
sense, nonsense, splice , indels , and small and large deletions
Spotty skin pigmentation with typical distribution (lips, conjunctiva, inner or outer
in the PRKAR1A gene (1639,256 ,2793) . Genetic heterogeneity canthi, vaginal or penile mucosa)
is likely. Initial linkage studies in 1996 suggested a locus on
Cardiac myxoma0
chromosome 2 but a gene has never been identified in this
Myxoma (cutaneous and mucosal)•
region {3048}. There are a number of genotype-phenotype
correlations reported . Lentigines , cardiac myxomas, and thy- Breast myxomatosis• or fat-suppressed MRI findings suggestive of this diagnosis
m1d tumours are more frequent in patients with the hotspot Primary pigmented nodular adrenocortical disease• or paradoxical positive response
pathogenic variant PRKAR1A c.491_ 492del, and isolated of urinary glucocorticoid excretion to dexamethasone administration during the
Liddle test
PPNAO is frequently associated with splice variant PRKAR1A
c.709-7_709-2del {256) . CNC patients without a detectable Acromegaly due to growth hormone (GH)-producing pituitary adenoma I pituitary
neuroendocrine tumour (PitNET)•
PRKAR1A pathogenic variant usually represent sporadic cases
that occur later in life with a milder phenotype 1256). Large gene Large cell calcifying Sertoli cell tumour' or characteristic calc1ficat1on on testicular
ultrasound
0ele11ons might be associated with developmental delay and
other features not typically seen in CNC 12793). Thyroid follicular adenoma or carcmoma• or multiple. hypoecho1c nodules on thyroid
ultrasound in a young patient
Certain pathogenic variants affecting the 3' end of the gene
result in the synthesis of a mutant R1a protein unable to bind Mahgnant melanohc nerve sheath tumour
cAMP. The resulting PKA tetramer cannot be activated by cA MP Blue naevus ep1theho1d blue naevus (multiple)
and results in impaired signalling through the cAMP/PKA patt1- Br ast duct I adenoma Im ltiple)
'Nay 11905}. These variants do not cause CNC or tumours but 0 teochondromy oma'
an autosomal dominant form of acrodysostos1s with hormone Supplemental criteria:
resistance . Affected fir t-dvg1e re1ative
lnac1ivating mutation of th11 PRKAR14 ~c"
Essential and desirable diagnostic criteria
See Box 14.14. •These tumours all requir e hislologicai ~onrirmallon

~ c.n •CJ lll ,..,,; 1 H~r ( • 1 !.._.... , • '('d '· : . J , ~ '"=. \_ t J -.


Hi ll DA
O!CER1 syndrome AleYanrJr~ sr,,u S
1<61 s~he
C
Korshuno / A
Solomon DA

Definition are indistinguishable from those enr:oun e r~rJ ,,, tt- s--r -_'j,., .,~·':.
OICER1 syndrome is an autosomal dominant tumour predispo- counterparts (2769,706 ,1 677,3259,1836.705. 737 1 q1 ...1 ~~r~
sition syndrome caused by heterozygous germline pathogenic
sequence variants in the DICER1 gene, which encodes a micro- Extracranial manifestations
RNA-processing enzyme . It is characterized by increased inci- These include pleuropulmonary blastoma. pulnvjr;;r I -. w _
dence of benign and malignant neoplasms involving multiple thyroid gland neoplasia, ovarian sex-cord strorri a1~1..rrt:. ..r: ,._F--
organ systems . The CNS tumour manifestations associated with tic nephroma, renal anaplasti c sarcoma . c1liary med•.J r:~:: -.. i::
OICER1 syndrome are metastatic pleuropulmonary blastoma; lioma, nasal chondromesenchymal hamartoma. arid".:"':.'" ~ a
pineoblastoma; embryonal tumour with multilayered rosettes ; rhabdomyosarcoma of the uterine cervix {2572 3or. 75 2:152
pituitary blastoma; and primary intracranial sarcoma, DICER1- 3035,969,3483,3280) .
mutant.
Epidemiology
MIM numbering Pathogenic germline variants in DICER1 are estimated t '.JC::_,.
606241 DICER 1, ribonuclease Ill ; DICER1 in about 10 in 100 000 individuals {1 621}. DICER1 syr>d~orr~
has variable penetrance. Benign thyroid nodules and 1_.rg:, _·;
ICD-11 coding are the most common phenotypic manifestations occur rig -
None a large subset of individuals with pathogenic germhne vanar~
(1601). The exact incidence of specific neoplasms rn 1nd 'v10'v--::
Related terminology carrying germline DICER1 mutations remains uncertain n!•...-
Acceptable: pleuropulmonary blastoma familial tumour and tary blastoma is pathognomonic of DICER1 syndrome. vrr-- a
dysplasia syndrome . reported cases to date arising in the setting of this syndro'lle

subtype(s) Etiology
,\Jone DICER1 syndrome is caused by heterozygous germhne ioss-o•-
function variants in the OICER1 gene on chromosome 4q32 ·3
Localization (1309}. Mutations are most often transmitted in a familial mar--
The most commonly involved organs are the lungs, kidneys, ner, although approximately 13% of affected 1ndiv1duaJs rt
thyroid, ovaries, uterine cervix , eyes , and brain . pleuropulmonary blastoma harbour de nova mutatioris 372
Additionally, a subset of affected individuals acquire OICErl1
Clinical features mutations during postzygotic development and have a m .~
The clinical features are listed in Table 14.06 . phenotype. Individuals with somatic mosaic1sm for a DICE.R-1
RNase lllb hotspot mutation show an increased tumour 1 c -
CNS manifestations dence and are younger at presentation than individuals "' ·
The most common CNS manifestation of DICER1 syndrome germline loss-of-function truncating variants 13721.
is metastasis of pleuropulmonary blastoma to the cerebrum
(2571) . The CNS is the most frequent site of distant pleuropul- Pathogenesis
monary blastoma metastasis, with CNS metastasis occurring In addition to the germline DICER1 loss-of-function pathoge 1c
in 11 % of patients with advanced pleuropulmonary blastoma. variant, OICER1 syndrome-related tumours ty pically haroour ar1
The International Pleuropulmonary Blastoma/0/CER1 Regis- additional somatically acquired missense mutation in e ri 2
try recommends brain MRI surveillance every 3 months until or 25 encoding the RNase lllb cleavage domain, 1nv i an
36 months after a diagnosis of type II or type Ill pleuropulmonary of the following codons : p.E1705, p.01709, p .G1809. p..
blastoma (3280 ,2862}. The primary CNS tumour manifestations or p.E1813 . This leads to a unique combination of two its,
of DICER1 syndrome are pineoblastoma, pituitary blastoma, in contrast to the classic Knudson hypothesis, the
embryonal tumour with multilayered rosettes, and O/CER1- does not fully abrogate the function of the DICER1 gene
mutant primary intracranial sarcoma. Pituitary blastoma virtu- Some sporadic tumour counterparts harbour somanc b
ally always occurs in the setting of DICER1 syndrome, arises DICER1 alterations. These patients are not consider
in young ch ildren typically aged < 2 years , and often occurs dromic, although the possibility of unrecognized mo~aJCSSIT1
with Cushing syndrome and diabetes insipidus (2776,707}. should be entertained 1588,372} . However. the p·athoo1sne1S1S
The clinical features of pineoblastoma, embryonal tumour with of DICER1 syndrome-associated pineoblastoma di
multilayered rosettes , and DICER1-mutant primary intracranial the mechanism described above. with the somattc "
sarcoma that are associated with germline DICER1 mutations loss of heterozygosity of the DICERt allele t9 9,724}

464 Genetic tumou r syndromes i nvolvin~ tl1e CNS


-
___1_
~_0&~K-ey_c_h~rn-
· ca_1_p_he~n_o_ty_pe_s_a_s_so~c-ia_te.~d_w_it_h~ge_r~m-lln_e_o_1~CE~R~1~p~a~th=og~e:n:ic~va:r~ia~nt:s~~~~~~~~~~,...-~~---~~~~~-~~ -
OeathS assoclated With
Approximate ages of clinical
MaJig,nant (M) or benign (B) DICER1•muta.ted cases?
dlagnosls, range (peak)
Most frequent phenotypes
europulmonary blastoma (PPB)
Type I (cystic) PPB 0-24 months (8 months) M Yes, if it progresses to type II or Ill
Type 11 (cystic/solid) PPB M Yes, - 40%
12-60 months (31 months)
Type Ill (solid) PPB Yes, ~60%
1~72 months (44 months) M
Type Ir (cystic) PPB BorM None observed
Any age
MultinoduJar goitrea B No
5-40 years (10-20 years)
No (see anaplastic sarcoma of the
Cyslic nephroma G-48 months (undetermined) B kidney below)
Sertolt-leydig cell tumour of the ovary 2-45 years (10-25 years) M Yes,< 5%

tlOderale-fnK1uency phenotypes
Embryonal rhabdomyosarcoma of the cervix 4-45 years (10-20 years) M None observed

Rate phenotypes
Differentiated thyroid carcinomab 5-40 years (10-20 years) M None observed

Wilrns tumour° ~13 years (undetermined) M None observed


Juvenile hamartomatous Intestinal polypsb G-4 years (undetermined) B No

Ciiary body medulloepithelioma ~1 Oyears (undetermined) BorM None observed

Nasal chondromesenchymal hamartoma 6-18 years (undetermined) B No

Pituitary blastoma 0-24 months (undetermined) Undetermined Yes, -50%

Pineoblastoma 2-25 years (undetermined) M Yes

Very rare phenotypes


Anaplastic sarcoma of the kidney Estimated 2-20 years M Yes
Embryonal tumour with multilayered rosettesb Undetermined M Unknown

Pnmary intracranial sarcoma, DICER1-mutant Median 6 years M Unknown

Embryonal rhabdomyosarcoma of the bladder Estimated < 5 years M None observed


Embryonal rhabdomyosarcoma of the ovary Undetermined M None observed
Neuroblastomab Estimated < 5 years M Yes
Congenital phthisis bulbib Birth B No
Juvenile granulosa cell tumou~ Undetermined M None observed
Gynandroblastoma Undetermined M None observed
'.\ultinodular goitre occurring in patients aged< 18 years may warrant DICER1 testing, even if it occurs in the absence of other syndromic features 1n the patient or their family
tl"nese phenotypes may not be sufficiently associated with D/CER1 mutations to warrant testing In the absence of other personal or family history suggestive of DICER1 syndrome

lri general. the DICER1 alterations in ben ign and malignant Histopathology
syndrome-associated tumours are identical , and it is hypoth- The h1stopathology of tumours in DICER1 S\ ndrome is similar to
e~1zed rhat the variable malignant potential is due to th e pres- that of their sporadic counterparts .
crice of additional oncogenic alterations , such as TP53 and
NRAS RAF mutations (2580,1913,1865) . The mutations in Cytology
DICER1 are thought to promote tumorig enesis via th e disruption The cytology of tumours 1n DIC.. EFi'1 synurorne 1::; s1m1lar to that of
IJf rnicroRNA regulation of gene expression . permitting aberrant their sporadic counter par ts
oncofetal transc riptional programmes to persist beyond fetal
ljevelopment 1969). Diagnostic molecular pathology
Most germltn pathoyPrnc vark'lrits ar nur1~,t'fbl:' , 11L1lat1iJr1S
Macroscopic appearance small insert1on/del ,t1<)ns , 1Jr spl ice: s1 e •,u b ,.t 1tut:rn 1;:, r~~' , : 11•.1 111
The macroscopic appearance of tumour ~: 1n OICER1 syn-Jrome truncation ot t11e protein l ctrgu lield1ons :.:mu r uthl\1~'' '.I.~ ·~1~ ·· ·
15
similar to that of their sporadic counterp arts sens- variants make up d small perL.er itd~J e ut l~, 11 h:Jtiv.• v :i :.i '
There are now II-established clinical algorithms for the diag - lox14.15 DiaqMstii: 1;r1t~NI tor DICERr 'lV"tr11'!1T•
nosis of OICER1 syndrome beyond germline testing . The identi -
E f:
fication of a heterozygous germline DICER1 pathogenic variant
that is known or suspected to cause loss of funct ion establishes
the diagnosis . The hallmark manifestation of OICER1 syndrome DI lrable:
is pleuropulmonary blastoma, although any of the other mani- Genomic rumour testing demonstr.ittno
festations can appear first. In the presence of pleuropulmonary Involving the remaining DICER1 t • oft 'l
RNase lllb domain
blastoma or any other condition that has been described in the
setting of OICER1 syndrome, there should be a low threshold
for germline testing . Prognosis and prediction
No difference in clinical outcomes
Essential and desirable diagnostic criteria DICER1 syndrome-associated versu c;pnr .1 r - - rr ~ , •

See Box 14.15. embryonal tumour with multJlayered roset e,.


primary intracrarnal sarcoma
Staging
Not relevant

466 Genst1c tur nour ~yncJ rornes 1nvulv111q tt1G CNS


Farnilial paraganglioma syndromes Asa SL
Brandner S
Lloyd RV
I
oefinitlon
Familial paraganglioma syndromes are a group of inherited
cancer s~ndro~es characterized by the presence of paragan-
gliomas (1nclud1ng phaeochromocytoma) .

MIM numbering
"ee Table 14.07 (p . 468).

ICD-11 coding
one

Related terminology
4cceptable: familial paraganglioma-phaeochromocytoma syn-
dromes; hereditary paraganglioma-phaeochromocytoma syn-
dromes; hereditary phaeochromocytoma-paraganglioma .

Subtype(s)
Familial paraganglioma syndromes are shown in Table 14.07
(p. 468). Fig. 14.27 Paragangliomas in familial paraganglioma syndrome . Multiple para·aortic
and pelvic paragangliomas are identified with 68Ga-DOTATATE PET-CT 1n a patient
Localization with a pathogenic germline SDHB mutation.
Sympathetic-derived paragangliomas are usually intra-adrenal
(phaeochromocytoma) or they may be retroperitoneal, occur-
ring alongside the aorta and the inferior mesenteric artery, and
above the aortic bifurcation. Parasympathetic-derived paragan-
: I
gliomas commonly arise in the head-and-neck region , including Ousnr l
Krebs cycl....,.111&.t•d VHL/£PAS1-f•~ted
the carotid body and cervical branches of the glossopharyngeal PHw:kahYlll'lJd• pathWlly Pseudallypaxb palhw•v Owter 2
KINN Mfn•llnc pathway
and vagus nerves {2923). Familial paragangliomas may be mul - • lrmnaWlcba~ICaJ~'K>fype'
.s,,ci,.nt
r • lmman..rt')«ff'ftlfYPhttrklf'v~
· ~ Df.,,@N"°"'
1
I •Mot1J1".lll\'•Wf'y~t)./)9 · MD111<1!!W"!'"""''"·.,.,_,rr,u'
t1focaJ; they occur anywhere in the body with the exception of .::~Nor.idlt'l'.aJjne 1 •Gf'll'tot)p;r,flfotnrypH 1
AdJ.,_,..& ,..\ll~rc11""°" A.>l~UMGcMJr.n~

I · v~""'>-iucPru1atYpn
bone, bra in, and lymph nodes (144]. Spinal paragangliomas are •M°'tc.ommonU!J:Mof ptmlin.
dt-.,.u54~ndl\la,n&.\ t r.a"tcf
i Vl"L £P..UJ
~lt/t.110t11.lSCJtl •b.rll..:.t)olk ,...,,..r.u,?O
AH~U4~WJO'\J .fil,.\rL.\U..' l'MrMll-'
mfl• .utx:d~
usually non-familial, but one study identified an SOHO germline
mutation in one patient with recurrent sp inal paraganglioma and
cerebellar metastasis {2033) .

Clinical features
Clinical manifestations may be due to catecholamine excess
Flg.14.28 Familial paraganglioma syndromes. Biochemical and genetrc clusters of
and/or mass effects .
paragangliomas (144}.
Symptoms of adrenaline/noradrenaline excess include sweat-
;ng, palpitation, and anxiety; signs include hypertension and
tachycardia . These are generally associated with sympathetic indium -labelled so matostat1n PET. Cluster 2 tumours can be
11aragangliomas. Some parasympathetic paragangliorn as may imaged with W F-DOPA PET-CT or iobengudne trneto.ioclooen-
~ecrete dopamine with minimal clinical manifeslat1on s, wh ereas zyl g uani d 1ne, 123 1-MI BG) PET
Others, mainly those of the head and nec k and the cauda eq uina,
are non-secretory. There is strong genotype- phenotype correla· Epidemiology
lion in catecholamine profile. Cluster 1 tumours are those with 0 erall. 30 - 40% of paragangllomas m aaults ...ua hereditary
Dseudohypoxic pathogenesis , and they ter1cJ to be c l1nically silent cascade te:sting of incl 'X patients IGc1l1tates nsh rl:::'duct1ori strat-
and non -secretory or dopam1ne -secretiny. c 11 ister :::' comprises eg ies across mire kindreds !309 ,2024 1. Younger clQt" ar pn.~­
tumours with kinase signalling and rare pt'1<1t:uchror noc\!foinas sentC:Jt1on, multiple turnours . a11c xtn-cidrenal 1urr1 our~ J.re -;0
with WNT-pathway act1vat1on that are usu ally fur1c llL1 nal r~lf,~cet r1 1ly a_ssoc13ted wi th the µ re ence of a yerrnli .,,, rn~,r.:!t1on
Cluster 1 tumours express SSTRs. anJ tl 't;, are w~ II ..:1<:>u- I?"- 341 In 1..-011trast. CdU ia equ111 paragangllom~:i. s <::lJ1..! ~,1- 1_.r t..1dic
ahzed with 68Ga-DOTATATE PE T-CT or n,c less ~""f'rhiti'u c1nc.i t:xcc.;pt1orially rare 1n the fam ilial setting

'•.:it Il l "- ' '·'1h1u1 Sy·nd1•J.--.-lP"' r· - .


'' - ~ t l'/L1v1ny •'' C '- ! ,.
Table14.07 Familial paraganglioma syndromes
Chromosomal
Gene Syndrorrte Commonest locations Associated lesl0n1
location
Medullary thyroid carcinoma
RET 10q11.2 MEN2 Adrenal Parathyroid proliferations 171400. 162300. 164761
Mucocutaneous ganglioneuromas
Clear cell renal cell carcinoma
Haemangloblastomas 193300, 6085.17
VHL 3p25.3 VHL Adrenal, EA-PGL Neuroendocrine tumours
Pancreatic serous cystadenomas
Neurofibroma and MPNST
NF1 17q11.2 NF1 Adrenal, EA-PGL Ocular manifestations 162200
Duodenal neuroendocrine tumour
Renal cell carcinoma
SOHA 5p15.33 PGL5 EA-PGL, adrenal Gastrointestinal stromal tumour 614165, 600857
Pituitary tumour
Renal cell carcinoma
SDHB 1p36.13 PGL4 EA-PGL, A&T, H&N Gastrointestinal stromal tumour 606864", 115310. 185470
Pituitary tumour
Renal cell carcinoma 605373, 602413
SOHC 1q23.3 PGL3 EA-PGL, H&N Gastrointestinal stromal tumour
Renal cell carcinoma
SOHO 11q23 PGL1 EA-PGL, adrenal Gastrointestinal stromal tumour 168000, 602690
Pituitary tumour

SDHAF2 11q1 2.2 PGL2 EA-PGL, H&N Insufficient data 601650. 613019

TMEM127 2q11 .2 Unknown Adrenal, EA-PGL Renal cell carcinoma 171300. 613403

MAX 14q23.3 Unknown Adrenal, EA-PGL Insufficient data 17130°' 154950


Cutaneous and uterine leiomyomas
150800
t FH 1q43 HLRCC Adrenal, EA-PGL
Renal cell carcinoma
Duodenal neuroendocrine tumour
EPA St 2p21 PZS Adrenal, EA-PGL, A& T Polycythaemia 61 1783
Ocular manifestations

EGLN1 1q42.2 Unknown Adrenal, EA-PGL, A&T Polycythaemia 609070,609820

EGLN2 19q13.2 Unknown Adrenal, EA-PGL, A&T Polycythaemia 606424

MDH2 7q11.23 Unknown EA-PGL, A&T Insufficient data 617339


Ganglioneuroma/neuroblastoma
KIF18 1p36.22 Unknown Insufficient data Leiomyosarcoma 605995
Lung adenocarcinoma
Parathyroid proliferations
MEN1 11q13 Unknown Adrenal, EA-PGL, H&N Pituitary tumour 131100, 613733
Neuroendocrine tumours
A&T, abdomen and thorax; EA-PGL, extra-adrenal paraganglloma; H&N, head and neck; HLRCC, hereditary leiomyomatosis and renal cell carcinoma; MEN2, multiple endocnne
neoplasia type 2; MIM number, Mendelian Inheritance in Man number; MPNST, malignant peripheral nerve sheath tumour; NF1 , neurofibromatosis type 1; PGL 1-5, hereditary
paragangliomas 1-5; PZS, Pacak-Zhuang syndrome; VHL, van Hippel--lindau syndrome.
•Paraganglioma and gastric stromal sarcoma, also known as Carney-Stratakis syndrome.

Etiology Macroscopic appearance


Familial paragangliomas are caused by germline pathogenic Familial paragangliomas are often multifocal. Adrenal disease 1n
variants (Table 14.07) that predispose to tu mour development patients with multiple endocrine neoplasia type 2 is often bilat-
1939,3241 l. eral and grossly multinodular.

Pathogenesis Histopathology
See Table 14 07 Some SDH-associated tumours have a distinct pseudorosette
pattern (1632) . SDH-related paragangliomas from the head
and neck usually have small cells with clear cytoplasm Other
unique features include a prominent nested architecture with
well -formed. almost circular nests and monotonous cells with
Rf.14.29 SDH-related paraganglioma. A These tumours often have abundant granular eosinophilic cytoplasm . B They express cytoplasmic tyrosine hydroxylase. C Lack of
~c SDHB with intact stromal positivity indicates SDH-related disease. D Tumours associated with a pseudohypoxia pathway alteration also express inhibin.

acuolated eosinophilic cytoplasm {3242). Unlike sporadic


paragangliomas, they are rarely associated with a spindled
rrorphology or densely granular cytoplasm {3242) . VHL-asso-
ctated tumours may have clear cells with vacuolated cytoplasm
a~d stromal oedema 12617,1671).
lmmunohistochemistry localizes neuroendocrine markers
such as nuclear INSM1 {2723) and cytoplasmic synaptophysin
ard chromogranin . S100 highlights sustentacular cells. Para-
gangliomas express nuclear GATA3 (2978) and cytoplasmic
tyrosine hydroxylase {144) . With the exception of cauda equina
tumours, most paragangliomas are immunonegative for cyto-
r.eratins (739) . Loss of SDHB immunoreactivity in tumour cells
mtn granular cytoplasmic staining of stromal cells supports the
diagnosis of SDH-associated disease (1107,3283) ; in tumours
that lack SDHB immunoreactivity, loss of SDHA staining iden-
l!fies patients with SDHA mutations 11712). VHL-associated
:u-nours have membranous CAIX positivity 125151 and express
-
- (SDH) staining in SDH disease
Fig. 14.30 SDH disease. Succinate dehydrogenase
Familial head and neck paraganglioma 1mrnunostained for SDHB showing an SDH-
•rh1bin . FH mutations can also be screened by immunohisto - defic1ent paraganghorna with granular cytoplasmic sta1n1ng of endothelial cells and
chemistry {487) completely negative tumour cells.

Cytology Box 14.16 Diagnostic criteria for familial paraganglioma syndromes


The cytological diagnosis of paraga ngliomas is challenging, but
Essential:
1mmunoh1stochemistry may assist in the d1agnos1s and in deter-
Manifestation 1n rnulttp;e locatJons
rn1nat1on of familial disease {941 I.
ANO
Diagnostic molecular pathology Gem11111a mutation in a L.$< t•p•1t 1:,iy y1;·1

Not relevant Desirable:


Loss cf SDHB 1rn1 nunor -.:it:1 v1ty t1as ,1 hiQt1 pre,ti<:tive \ 'til'':? ltii ~-1>it1. :iL..!1 , ' '·
Essential and desirable diagnostic critenn SOHO niU!dltOn
See Box 14.16.
l ', I I I 11
t i t ; ,' ' '
' 'J . ; '
Fig. 14.32 Fumarate hydratase-deficient phaeochromocytoma arising In a patient with confirmed germllne FH mutation. Although there are no diagnostic morpnolog1caJ features
of fumarate hydratase deficiency on H&E (A,B) , staining for fumarate hydratase (C) shows loss of reactivity in the cytoplasm with staining in stromal elements, ano 1here •S " - t
staining for 2-succinocysteine (D).

Staging
multifocal ; the presentations may be asynch ronous r ,"fl-
Staging is available using the Union tor International Cancer icking metastasis . Catecholamine profile and SOhB rr L.t=t-
Control (UICC) eighth edition staging system.
tion increase risk of metastasis . Aggressiveness 1n sp1....r .::..Ol-
phaeochromocytoma is associated with MAMLJ tusions 1r
Prognosis and prediction
with ATRX and CSDE1 mutations but these are not roi-; ... 1 r..- ·
Most paragangliomas can be su rgi cally resected ; however,
in familial tumours . Five-year overall survival rates 1n p-.H•e 1 - 1

large tumours and some extra-adrenal tumour locations


with metastatic paraganglioma range from so~o to 7l 0 '1,'_
may preclude complete excision. Familial lesions are often
148 ,940 ,1223).

470 Genetic tL.mour syndromes 111volv111g tho CNS


Melanoma-astrocytoma syndrome Solomon DA
Reuss OE

Definition Etiology
Metanoma-astrocytoma syndrome is an autosomal domi- Melanoma-astrocytoma syndrome is an autosomal dominant
nant tumour predisposition syndrome caused by germline tumour predisposition syndrome caused by heterozygous
pathogenic variants of the CDKN2A tumour suppressor gene germline mutation or deletion of the CDKN2A tumour suppres-
encoding the p161NK4a and p14ARF cell-cycle regulators . The sor gene on chromosome 9p21 .3 l1532,173}.
syndrome is characterized by an increased risk of multiple neo -
pJasms including cutaneous melanoma , astrocytomas , nerve Pathogenesis . .
sheath tumours, pancreatic cancer, and squamous cell carci- Melanoma-astrocytoma syndrome is caused by genetic disrup-
noma of the oropharynx . tion of the COKN2A tumour suppressor gene, which encodes
the p161NK4a and p14ARF cell-cycle regulators 11532,173). The
MIM numbering p161NK4a protein binds and inhibits cyclin-dependent kinases 4
155755 Melanoma-astrocytoma syndrome and 6 to maintain cells in the resting G 1 phase of the cell cycle .
The structurally unrelated p14ARF prote in is produced from an
ICD-11 coding alternative reading frame and acts by antagonizing the p53
~one regulatory protein MOM2 . The neoplasms that arise in the set-
ting of melanoma-astrocytoma syndrome are thought to be due
Related terminology to abnormal prol iferation of melanocytes. astrocytes . and other
4ccept.able (depending on the tumour spectrum present in the cells after somatic inactivation of the remaining COKN2A allele,
kindred): melanoma and neural system tumour syndrome; typically via loss of heterozygosity (2809 ,507).
melanoma-pancreatic cancer syndrome; familial atypical
mole-melanoma (FAMM) syndrome; susceptibility to cutane- Macroscopic appearance
ous melanoma type 2 (CM2). The macroscopic appearance of tumours in melanoma-astrocy-
toma syndrome is as described for the individual tumour types.
Subtype(s)
None H istopathology
The astrocytomas arising in the setting of melanoma-astrocy-
Localization toma syndrome include both pleomorphic xanthoastrocytoma
The dysplastic naevi and melanomas arising in the setting of and diffuse astrocytic gliomas ranging from low-grade (diffuse
melanoma-astrocytoma syndrome are usually cutaneous and astrocytoma) to high-grade (glioblastoma) (507) . No histopatho-
nor mucosa!, acral , or uveal. The astrocytomas are usually logical features distinguishing these syndrome-associated
located in the cerebral hemispheres or cerebellum . astrocytomas from their sporadic counterparts have been iden-
tified to date. The nerve sheath tumours have been reported
Clinical features to histologically resemble either schwannoma or neurof1broma
Melanoma-astrocytoma syndrome is characterized by multiple (3291 ,2809) .
cutaneous dysplastic naevi and an increased risk of melanoma,
:istrocytomas , nerve sheath tumours , pancreatic cancer, and Cytology
squamous cell carcinoma of the oropharynx !1571,166,174, Not relevant
3553 .2874.430,3291,987,2809,507) . Nerve sheath tumours
1nclud1ng both paraspinal schwannoma-li ke neoplasms involv- Diagnostic molecular pathology
ing spinal nerve roots and cutaneous neurof1broma-like neo- Sequencing analysis of the CDKN2A gene 1n a con~t1tut1ondl
p1asms have been reported /174 ,3291.2809,5071 It is currently DNA sample assessing for p thogenic mutations or deletions 1s
·.mknown why some kindreds exclusively develop dysplastic required for the diagnosis of me la nor na-astrocyrumcl syndrome
'1aev1 and melanoma while others also develop astrocytornas, Genomic analy is of astro ytonn <tn other tumours arising 1n
Pancreatic cancer. and other neoplasms . the setting of melanorna-astrocytoma synliro1 ne demonstrates
somatic ina t1vntion of ll1e r\:·n1<11rl'nq c.Of N< •,._, 111 10 18 111.:1 loss 01
L

Epidemiology heterozygosiry or un ':1C u1red St'CL1nd n1L.tc1t1un/dale!lun (til17\


Melanoma-astrocytoma syndrome is rare w1ln < 100 gonet1- One study of a yndron11c ~I orn nph1L' xel1Hf1t•,1str•..11'YtL)JY1d
ca11y confirmed kindreds reported to dam Th e 1?.xact 1nc1derict> revealed CDl<..N.?A I 1omozyqou· JL?lcliiJn -tn,1 :3h'-V- p Vt cir,if
•s unknown, but this syndrome probat:,i y 8L'VJur1t~ for ~ U 1 ~ 0 of mutut1on, and .::1 sepdrato d1fru, e d~t 1 ocytornd til rn 1~-, Od 1en \' 1:-
astrocytom as and nerve sheath tu inour~; IDH - and h1stone. 13 wildtype With CL )KN:.!A l)\.11ll• 'ZV ll1L.') rj, it:-
t'.on CHKi P~PNI/ , NF1, ana ATRXmut~t1011s /bU7) 1t;t. r•Jl..:._L
lar P<llhogenesis of nerve st1eath tumours ar ,::; 11 '::.J 1r1 J1• , .~.• ~,;1'.J
Box 14.17 D1agnos1ic r.rilN1 for mr-ll!nomA itsfrt>r y•orn11 ~yn&l'lt."1 ~ J""' 'l r 1
'1 r1 'f' ' '' 1 I J P l

Essential: -, , d r ,.. !'" 17 11 r 1 ·r>• r ·~ t • ~· ,,,

Pathogenic rmlln vn nt of th
astrocytoma
Desirable:
Personal and'or f mlly h1~t rv of dyspl slr

l 1I lE'lanor 1<1 astrc Cyloma c::;yndrorn9 has r')f ee n195, ga·~


to a Qu\ Ol\N2A deletion rs not charac E>nst c cf e :r-e· sc -
rad1c schwannomas or yprcal neurol1brcrna sJgges~ .... g :!-13·
thE'lf pathogen SIS IS d1st1ncl from tha Of 1e:r SDOrad C CO "•er-
parts Neverth less . it 1s seen in atypical neurofrbror--·a I a' o ':.a
Familial retinoblastoma Eag le RC Jr
Jones OTW
I
'

Definition
Retinoblastoma is a malignant paediatric retinal neoplasm.
Familial cases are caused by germline RB1 pathogenic vari-
ants.

tCD-0 coding
9510/3 Retinoblastoma

MIM numbering
180200 Retinoblastoma; RB1

ICD-11 coding
2002.2 Retinoblastoma

Related terminology
Acceptable: trilateral retinoblastoma .
Not recommended: glioma retinae .

Subtype(s)
None

Fig. 14.33 Retinoblastoma. Low-power view showing replacement of the retina by


Localization
retinoblastoma.
Retinoblastoma is an intraocular tumour of the retina. In familial
retinoblastoma syndrome, synchronous or metachronous malig-
nant intracranial tumours (pineal or suprasellar) may develop. 45% of cases , with an approximate 5% risk of developing trilat-
eral retinoblastoma (703) .
Clinical features
Leukocoria (a white pupillary reflex) and strabismus caused Etiology
by visual loss are characteristic presenting manifestations. Familial retinoblastoma is caused by germl ine pathogenic
Eventually, tumour progression leads to extraocular extension sequence variants in the RB1 gene .
and involvement of adnexal structures. About 60% of patients
who have germline RB1 pathogenic variants develop bilat- Pathogenesis
eral tumours . Familial tumours tend to arise at an earlier age Most retinoblastomas are c aused by germli ne or somatic muta-
(-12 months) than do sporadic counterparts (-24 months). tions in the RB1 tumour su ppressor gene on chromosome 13
Germline carriers are at significant risk for secondary non-ocular (13q14.2) (788 ,58 ,352). Abo ut 40% have de novo or inherited
neoplasms including sarcomas and pineoblastoma; the combi- germl ine pathogenic variants that are transmi ssible as an auto-
nation of intraocular retinoblastoma and a histologically similar somal dominant trait. Ampli fi cation of the MYCN oncogene
brain tumour, most commonly in the pineal gland , is called causes rare cases of the non -hereditary form without RB1
trilateral retinoblastoma {2011,3616,1646,703}. For intracranial mutations (accounting for 2% of all cases of retinoblastoma)
tumours , a maximal safe surgical resection is encouraged at (2757) .
diagnosis, followed by craniospinal irradiation and high-dose
chemotherapy {4) . Macroscopic appearance
Reti noblastoma arises from and destroys the retina. The rumour
Epidemiology has a white or encephaloid appearance with lighter L.alc1t1c
Retinoblastoma is the most common primary intraocular tumour flecl<.s. Endophytic and exophytic tumours arise trom tha inner
m infants and the most common primary intraocular tumour and outer layers ot the retina causing vitreous 1nvas10n a.rid
worldwide 11645). It has a reported incidence of 5 6- 6 3 cases retina l detachment, respectively Rare cl it tu st= 1nriltr ...1r1ve 'et-
per 100 000 live births (422,3329,378 ,16451. There is no racial 1noblastomas (1 .5°~ of cases) lack Cdlcif1..._·c1t1l...1 l ,1'1(1 ~1 dis..:rt'(8
or sex predilection . The large majority (8 0%) of cases are diag- mass They occur in older ·.::-hildte'l (rne,1n dye r' ? 1 e,H~' .1·11...i
nosed before 3 years of age. and the mean age at cfi agnos1s is contribute \0 rn1sdiayriusis P::.1t1ents vv1lti 11.nitf'ti .Jl-2t::~;::; k, 11 'ti'
18 months (typically younger for bilateral retinoh lastom8) (3163 C..11 care lreqliently f're:::;ent witt 1 iJcu ~u dt=> ·:ruc'il' ' t~dr u,'L ,1·
131) Patients with heritable retinobldstoma ciccou1 it for about extension arid c:u1 t)rb1u1 1T1ass

I'..'•.·
Box14.18 Diagnostic criteria for familial retinoblastoma and sometimes trilateral) retinob18sft"'lrr"'
. .,, 1-J r.Ji:;f'1l•f1r, (' ...
Essential: s hould therefore be consid~red for r. ..,.. , '> · •~
. J.,,1t;n ~ ,11i..1 -.'ii"'$.- " r..-;
lntraocular small blue round cell tumour with typical histopathology and ocu Ia~ disease (particularly bilateral n::~11r •it°J .=J'"» ;rr.~ °''",.
demonstrated germline alteration of the RB 1gene ~al disease (pineoblastoma/ret1noblastorri;j) Gi:r,c_::~ • ;,,.. ~-
OR 1s us~d to determine if patients ha1e gerrn!1r-,:: r)r _,._...,,~. c.p. ,
mutations (25901 . lntracranial pineoblastorr~ ... re.• r.r· ~-·- --
Trilateral occurrence of intraocular small blue round cell tumours in both eyes and in
the pineal region ot
whether sporadic or germline . have a dis 1r,rt- l.i.- -_;.:=.: ' _::
p ro file from that of other pineal tumours and sr.-~r~ ,. ' ,..,.
copy-number features with ocular retirioblastcn- :i ~ 'J , 1r- ·_
Histopathology some 16 loss and 1q gain) 11865.2490 1
Retinoblastoma is a mitotically active small blue cell tumour
composed of primitive neuroblastic c ell s. Perivascul ar cuffs of Essential and desirable diagnostic criteria
viable cells, tumour necros is, and dystrophic calcification are See Box 14.18.
common {4221. Flexner-Wintersteiner rosettes are a character-
istic feature of retino b lastoma, b ut they may occur in other neo- Staging
plasms such as p in eobl astoma and medulloepithelioma. They Retinoblastoma is staged according to the Un :,n ;er r-•-rr-
represent early retin al d ifferentiation and have a central lumen tional Cancer Control (UICC) TNM class1f1cat1on e1gr•~ -=- · - r
{3231}. Rosettes are more c ommon in very young infants (815} . In b ilateral cases . each eye should be staged seo-ra'e.,
Photoreceptor differenti ation is found in 15-20% of retinoblas-
tomas {2097,815 ,757}, and it is characterized by aggregates of Prognosis and prediction
neoplastic photoreceptors called fleurettes . Massive posterior Untreated retinoblastomas are fatal. In developed ~c r. r P
uveal invasion (defin ed as > 3 mm in largest diameter) and ret- the survival rate of treated cases approaches 95"o ~c. ::
rolaminar optic nerve invasion are high-risk histopathological the survival rate is only 50% (2985,1645.4561 Ir a pro;oe ·•
markers that are in d ications for adjuvant chemotherapy (2814 , study of > 300 enuc leated eyes, the most s1gnif car. pre r· .
561 ). Involvement of anterior segment structures and severe factors for recurrence and death were extensive retro•<:l' n •
tumour anaplasia are thought to increase metastatic risk (2069). optic nerve invasion concomitant with massive ( 3 rim -:-. -
The histological fe atures of the intracranial tumour/pineoblas- roidal invasion 1561 }.
toma in trilateral retinob lastoma are essentially identical to those In patients with retinoblastoma syndrome. the i:;roq-..,
in retinoblastoma. sis depends on the stage at which the differef'lt r1.11T1 ·s 't
diagnosed and treated: the earlier the diagnos s tr.e o ~ :
Cytology the prognosis. It is important to screen survivors ot e·ea.~ ·,
Cytology shows small b lue cells , singly or in aggregates , mixed retinoblastoma for second malignancies. including ost r 'J
with necrotic tumour. Rosettes are found occasionally (529} . mas (typically in the first and second decade of life -1 ''
Fine-needle aspiration biopsy is discouraged due to concern tissue sarcomas (10- 50 years after ret1noblastor-a dt:::!c; r •

about needle track contaminati on and extraocular spread . Other epithelial tumours of the bladder, lung. and br t _,
melanoma, may arise after the second decade of It e Pa• c,.,..
Diagnostic molecular pathology with retinoblastoma syndrome may have more t an ~ ~ 5c:i-.:l IJ
Retinoblastoma syndrome is defined by a constitutional genetic primary mal ignancy, and screening should continue nro .... : j -~
alteration in the RB1 gene leadin g to a high risk for (often bilateral the patient 's life.

474 Genetic: turn o ur ~yr 1clior1 1w. 111 ·,, 1I IL• (j l"~ _.,)
;1 1, 111
')
BAP1 tumour predisposition syndrome Santagata S
Wesseling P

Definition predominance , and are frequently peritoneal rather. than. pleu-


BAP1 tumour predisposition syndrome is an autosomal domi- ral [218,2308,2414) . Histologically distinctive BAP1-ina~ t1vated
nant disorder caused by pathogenic germline variants in the naevi/melanocytomas occur in three quarters of germl1ne car-
BAP1 tumour suppressor gene. The syndrome is character- riers [1266 ,3486), yet they are not specific for the syndrome
ized by a predisposition to various tumours including uveal [431 ). An association between meningioma formation and
melanoma, mesothelioma, cutaneous melanoma, and renal cell BAP1 germline pathogenic variants was first reported_ in three
carc!no~a, with less frequ~ntly occurring tumours including affected families [6,559 ,3353/ and subsequently linked to
mening1oma, basal cell carcinoma, and cholangiocarcinoma. meningiomas with rhabdoid and papillary morphology (2888,
2890,3449).
MIM numbering Criteria for genetic counselling and testing include: (1) a
614327 Tumour predisposition syndrome; TPDS medical history of at least two BAP1 tumour predisposition syn-
drome-associated tumours, (2) one BAP1 tumour predisposi-
ICD-11 coding tion syndrome-associated tumour and a first- or second-degree
None relative with at least one BAP1 tumour predisposition syndrome-
associated tumour(s) , and/or (3) young age of tumour onset
Related terminology (2604,3361 ,527) . In the context of these criteria , loss of BAP1
None on immunohistochemistry or detection of BAP1 variants in rele-
vant tumours further supports a need for germline testing (1266,
Subtype(s) 3361/ . To identify tumours at early stages , screening includes
None eye and skin examinations and ultrasound or MRI [2604 ,3018,
3361,527) . BAP1-inactivated naevi/melanocytomas often pre-
Localization cede the onset of other tumours ; their recognition facilitates
Multiple sites and organ systems are involved, including the early syndrome detection .
eye [1235,6,1442). pleura, peritoneum (3169}. skin [3438,
3353). kidney {2540}. liver (2514). and meninges {6,559,3353, Epidemiology
2888,3361). More than 180 families have been described. with 140 unique
pathogenic germ line BAP1 variants , of which 104 are null vari-
Clinical features ants and 9 are missense variants in the UCH domain \3361) . The
The median age of tumour onset is younger in people with carrier frequency of germline pathogen ic variants is about 1 in
BAP1 tumour predisposition syndrome than in the general 50 000 in people without cancer and about 1 in 1900 among
population . Affected individuals can have multiple types of cancer patients , suggesting that the syndrome prevalence 1s
primary cancers {3361}. Mesotheliomas have substantially underestimated {2030) . The most prevalent founder variant is
different clinical features when arising in BAP1 tumour pre- BAP1 p.L573Wfs*2 (3361) . The lifetime risk of developing can-
disposition syndrome: they arise 20 years earlier, lack male cer is reported to be as high as 80 - 100% , wi th many carriers

Flg.14,34 BAPt meningioma. A recurrent meningioma arisiny in a 59-year-old man with a germlir1e BA>-1 1 µY1 7J. lrL.ncawi·' rn r· t· ,
. . ' I ' • ~ u ,l •Ufl 3 i!d ~ , J 1 ,, , 'I ' •
rna. A The tumour has mixed rhabdo1d and papillary features 8 BAP1 e.<press1on is ost 11111eoplast1c cells and reta.. 1e11 1n Jssoc 1at.... a
'"' non necµ:J li'L .1.: .,

... ,,, :.! . :J


Tumour type Prevalence of germline BAP1 mutations Prevatence of

Uveal melanoma 1-2% 40-45%


esothelioma 1-2% 23-64%
Cutaneous melanoma <1% 2-3% 21.!":
RenaJ cell carcinoma <1% 10-15% 5 -,.,
( ~

Basal cell carcinoma <1% 1-3% < •ry: _


lntrahepatic cholangiocarcinoma 1-2% 1&-25% 14 :
Meningioma (all histologies) < 1% <1%
-- - - - --- -- - -- --
BAP1-TPDS. BAP1 tumour predisposition syndrome.

developing multiple cancers; most families have at least two germline carriers (3169,2752,2890.2653 1. Tr·s rrc 1 ::-~.,- •
ypes of tumours in first- or second-degree relatives {2604 , with meningioma, where half of BAP1-mutart mer1~n r.: -
3361 j. Null variants predispose to earlier tumour formation than a small series arose in germline earners 12888 289CI 3~ ~._
do missense variants {3361}. mutant meningiomas account for< 1% of all rnenir.g1crr .
BAP1 is the most frequently mutated gene in sporadic The frequency of germline and somatic BAP1 rn.i a1 ·rs
mesothelioma (406} and in metastatic uveal melanoma (1235, various tumour types and the occurrence of those tur.ou c; -,
6,1442), but < 1-4% of these BAP1-mutant tumours arise in probands with null variants in BAP 1 are listed in Table • .~
to114.19 Diagnostic criteria for BAP1 tumour predisposition syndrome

tration of a germline pathogenic variant in BAP1


Cytology . ..
The cytology of tumours in BAP1 tumour pred1spos1t1on syn-
drome is similar to that of sporadic tumours .
I
.

Diagnostic molecular pathology .


Etiology Los s of BAP 1 immunoreactivity in tumour cell nuclei readily
BAP1 tu mo ~ r pre~isposition syndrome is caused by patho- identifies deficie nt tumours . Concordance between 1mmunohis-
genic germline variants in the BAP1 tumour su ppressor gene tochemistry and genotyping is high but incomplete { 33~ . 1704 ,
on chromos~me 3p21 .1. More data are requ ired to evaluate the 22 15,3270). In sequencing data, appra1s1ng pathogenic1t~ 1s
effect of en~1 ronmental mutagens (e.g. asbestos, uv rad iation) straightforward for indels and frameshift. nonsense. an.d splice-
on modulating the penetrance of germline mutations . De novo site mutations; guidelines for interpreting sequence variants can
BAP1 germline mutations are uncommon \3361 ,527) . assist in the class ificati on of missense mutations [2662 ,1877).

Pathogenesis Essential and desirable diagnostic criteria


The BAP1 gene on chromosome 3p21 .1 comprises 17 exons See Box 14.19.
en~d i ng_ ~ prot~ i n with ubiquitin carboxyl hydrolase activity
first 1dent1fied as interacting with BRCA1 [1468) . BAP1 interac- Staging
tors include ASXL1, ASXL2 , FOXK1 , FOXK2, HCFC1 , and YY1 Not relevant
consistent with roles in DNA damage response , transcription al
regulation , cell-cycle regulation , metabolism , inflammatory Prognosis and prediction
responses, and lineage differentiation (2890 ,560 ,3361 ,1775). Sporadic BAP 1-mutant tumours are often more aggressive than
Nuclear localization and deubiquitination activity are both nec- corresponding tumours lacking suc h mutations [1220,282). The
essary for BAP1-mediated tumour suppression {3310). prognosis of germline carriers with tumours is incompletely
characterize d because most stu dies of single tumour histology
Macroscopic appearance contain few patients with BAP1 tumou r pre disposition syndrome.
The macroscopic appearance of tumours in BAP1 tumou r pre- Uveal melanomas arising in BAP 1 tumour predisposition syn-
disposition syndrome is similar to that of sporadic tumours. drome have an increased risk of metastasis [2265 ,1191]. BAP1-
mutant meningiomas display aggressive behaviour with frequent
Histopathology recurrences (2888,2890}, althoug h it is unclear whether menin-
The histopathology of BAP1 tumour predisposition syndrome- giomas arising in BAP1 tumou r predisposition syndrome have
associated tumours is similar to that of thei r sporadic counter- a different prognosis than sporadic BAP1-mutant meningiomas.
parts. Many BAP1-mutant meningiomas have overt rhabdoid In contrast, patients wi th germline BAP1-related mesothelioma
cytomorphology, but the histology can be diverse, including have a better 5-year survival rate (47%) than those with sporadic
epithelioid-type cel ls and papillary growth \2888,2890). mesotheliomas (6.7%) (218,2308 ,2414) .

~i 7 7
Fanconi anaemia c;r,ir)r .. r)r
V rrll.1 r,p
r jf·

Definition Clinical features


Fanconi anaemia (FA) is a clini cally and genetically heteroge - FA is characterized by a range r;f or r -:.~' : ;
neous disorder caused by underlying genomic instability. Char- gressive bone marrow failure and 3 r.u~7-1 ;;.
acteristic clinical features include developmental abnormalities Classic features include abnorf"Tla t'"·'Jrr.Gs -
in major organ systems , early-onset bone marrow failure , and stature. skin hyperpigmentatron abn m~I h; .
a high predisposition to cancer. The predominant CNS tumour face , microcephaly). abnormal k1dne1s and je-:te:.-_
0

manifestation is medulloblastoma , resulting from biallelic patho- {717}, although th e absence of these fe-3t11~ .
genic germ line variants in either BRCA2 or PALB2. FA . The most common neoplasms assoc 3 ~1 N
lodysplastic syndrome and myelrnd :eu aerr;
MIM numbering carcinoma of the head and neck. Wilms tum
605724 Fanconi anaemia, com p lementation group 01; FANCD1 loblastoma .
610832 Fanconi anaemia, comple mentation group N; FA NCN
CNS tumours
ICD-11 coding Patients with FA due to b tallehc germline mu at 1:i :J
3A70 .0 Congenital aplastic anaemia PALB2 have a dramatically increased nsk of m-' gr- ,.,
ing childhood , with the predominant CNS tum ur ,_
Related terminology being medulloblastoma 1717.2297.131 .320 ~
None 2682,2108 ,3085,1662,33921. Rare examples o
with other CNS embryonal tumours or ghob as
Subtype(s) been reported /745,7691 . Children with meduJ',u.O>o.;,:~rr
1\Jone other childhood cancers that arise in the se 1ng o• - . . .
a family history of breast, ovarian. and pane e c con
Localization matern al and/or paternal lineages, caused cy h .... _
Manifestations of FA may develop in all organs and tissues . The carrier status for BRCA2 or PALB2 pathogenic • r
predominant CNS tu mour arisin g in the setting of FA is medul-
loblastoma. Epidemiology
FA is most often an autosomal recessive a1sor ·e
from homozygous or compound heterozygous gt:><!"1.,.•,n~
tions in the 22 different FANG genes The o e cc-
complementation group 8 (FANCB), wh1cn s X-
c omplementation group R (RA051), wh1cn is au _
nant. The estimated carrier frequency for FA rs 0
sons , while the approximate syndrome incidence
130 00 0 births !2725).

Etiology
FA is a heritable syndrome caused by germ 1ne •r:i.-·"'~'""''c_-- .,
deleterious mutations or deletions 1n the 11ar10• s -
FA can be caused by disrup tions tn 22 01H ,- t g
resu lt in a mostly s1m1lar clinical phenolype b t
ferences , including variable tumour pred1spo ..
on the specific causative gene , FA is dttfera ri
mentation groups A through W (e.g. comp1e1
is caused by biallelic germhne mutation in tr· ;:
date , only two complemen tation groups n "a
with an increased risk of CNS tumours O 1 an •
from biallelic germline muta tions 1n th~ BRt....
genes , respectively. Complemantauon gr yp '
Fig. 14.37 Medulloblastoma arising in the setting of Fanconi anaemia due to biallelic homozygous or compound heter~'r'QL u:::i Q~rr ,
germl1ne BRCA2 mutations . Tl-weighted postcontrast coronal MAI demonstrating a in the BRCA 2 gene n c hromosum~ !Jq 1 , i 1.J 1q, 1...
medulloblastoma in the cerebellar hemisphere ol a young child with Fanconi anaemia mentat1on group N 1s c au sed by h1.. 11
due to biallelic germline BRCA2 mutations .

478 Geneti c tun .uu r '> Yr 1d1 ( >1 1 1 u~, 1n ·/u1 , 111q ti lt- \ f\b
i
Rg.14.38 Medulloblastoma. Large cell I anaplastic histological type, SHH-activated and TP53-mutant molecular subtype, arising In the setting of Fanconi anaemia due to
biallelic germline BRCA2 mutations. A This medulloblastoma in a young child with Fanconi anaemia due to blallelic germline BRCA2 mutations demonstrates severe anapla-
s1a. B High power demonstrates the large cells with severe anaplasia. C This medulloblastoma demonstrates immunopositivity for GAB1 , indicative of SHH pathway act1va-
Mn. D This medulloblastoma also demonstrates strong nuclear staining for p53 protein in the majority of tumour cells, corresponding with the somatic TP53 mutation that 1s
present.

heterozygous germline mutations in the PALB2 gene on chro- causative of FA result in impaired homologous recomb1nat1on
mosome 16p12.2 {2633) . and crosslink repair, which drives chromosomal aberrations
such as amplifications, deletions , and translocat1ons that pro-
Pathogenesis mote tumorigenesis (979) .
The FANC genes encode proteins involved in the homologous
recombination of DNA double-strand breaks and in the repair of Macroscopic appearance
DNA crosslinks . The deleterious mutations in the FANC genes Not relevant

Rg.14,39 . ~ DH,~ , ,, .
Medulloblastoma , SHH-activated and TP5J mutant arr sing 1n the- s.etling of Fnrict.ni . ;:1r1'1t-r111u (:Ju'-· tv r 1o1.1e, ~ g' 1 , 11 °,"''- , ~ d ,
. .._ " - 1ilU!,J 0 ! • ~ 1 :' 11· I · I I 'J ,•Jf)/ .. (, 1
showing
DN the aneuploid genome ' with most chromosomes 1r1 the genome harbour 1n\l rnulttple rntr clctir.,11w , mJt ,• ~ ,. r '"rD-'r " 1:; ,,,"'"' "u'
, ,i n•.•" t'I 1.-r. 1 <}\If in1•
1
, .Jr- ,,,,-1111 dC•!!:)le .; ur,(;
· ,,
Abreak repair caused by the BRCA2 biallelic inac11vation. •

. \.·111 IJ ! ~ , r .- •r ~'1 i >. )'-;


1
,
1.i ...
segme ntal gains and losses j 1662,3392) . A di stinct mutation
Box14.20 Diagnostic criteria for Fanconi anaemia
signature has also been observed 1n medulloblastomas ari" -
Essential: ing in the setting of FA, featuring elevated numbers of large
Positive chromosomal breakage analysis after exposure to dlepoxybutane In (> 3 bp) insertions and deletions with overlapping m1croho~ol
leukocyte or fibroblast cultures (diepoxybutane test) ogy at breakpoint junctions characteristic of BRCA -def1c1en
Desirable: breast and ovarian cancers (3392) . The medulloblastomas that
Biallelic pathogenic germline mutations in a FANG gene arise in the setting of FA often belong to the SHH-activatecJ
molecular subtype, and they are frequently also TP53-mutant
Histopathology (2108 ,3392] . Notably, medulloblastomas can also arise 1n the
Medulloblastomas arising in the setting of FA are usually of the setting of heterozygous germline mutation in either BRCA2 or
large cell / anaplastic histological type, or they can have desmo- PALB2 (i .e. FA carrier status only); these medulloblastomao
plastic/nodular histology with superimposed anaplasia 13392}. have somatic/acquired inactivation of the remaining w1ldtype
allele (often via loss of heterozygosity), and they can belong to
Cytology the sonic hedgehog-act ivated , group 3, or group 4 molecular
Not relevant subtypes (1662,3392) .

Diagnostic molecular pathology Essential and desirable diagnostic criteria


Diagnosis of FA has traditionally employed chromosomal break- See Box 14.20 .
age analysis performed on patient-derived cultures of either
leukocytes from a peripheral blood sample or dermal fibroblasts Staging
from a skin biopsy after treatment with an agent that induces Not relevant
DNA interstrand crosslinks , such as diepoxybutane. Mutation
analysis of a constitutional DNA sample to assess for patho- Prognosis and prediction
genic mutations or deletions in the FANC genes is a comple- Medulloblastomas arising in the setting of FA due to biallelic
mentary diagnostic methodology that can identify the causative germline BRCA2 or PALB2 mutation have a dismal prognosis
gene abnormality. (3392) . This is probably a reflection of both the aggressive biol-
Medulloblastomas and other cancers arising in the setting ogy of these tumours and the fact that patients with FA cannoc
of FA are genomically characterized by a large number of tolerate high doses of certain genotoxic chemotherapies . In
intrachromosomal breaks, which can be visualized by copy- addition , patients with FA have a dramatically increased risk of
number analysis as several chromosomes having multiple developing metastases after therapy.

480
ELP1-medulloblastoma syndrorn e Pfi ster SM
Waszak SM

Definition Step 2 Step 3


Ste p 1
ELP1-medulloblastoma syndrome is an autosomal dominant (somatic) (somatic)
(germline)
disorder caused b~ pathogenic germline variants in the ELPt
ene and ?haractenzed by an increased risk of sonic hedgehog
(SHH)-act1vated medulloblastoma during childhood .
Chromosome 9q PTCH1
MIM numbering deletion mutation/deletion
155255 Medulloblastoma; MOB PTCH1 PTCH1
ELP1 ELP1

ICD-11 coding
None Wlldtype
Mutant

Related terminology fig . 14.40 Proposed three-step model of tumorigenesis in ELP1-medulloblastoma


Acceptable: ELP1-associ ated medulloblastoma. syndrome. Step 1: germline variant results in monoallelic inactivation of ELP1. Step 2:
som atic deletion of chromosome 9q results in biallelic inactivation of ELP1 and con-
Subtype(s) current monoallelic inactivation of PTCH1. Step 3: somatic PTCH1 mutation or focal
deletion on 9q22.32 results in biallelic inactivation of PTCH1.
None
pathogenic germline ELP1 variant; step 2, biallelic inactivation
Localization of ELP1 with concurrent monoallelic inactivation of PTCH1 due
Tumours arise in the cerebellum . to an acquired deletion of chromosome arm 9q ; step 3, biallelic
inactivation of PTCH1 due to an acquired mutation or focal dele-
Clinical features tion. ELP1-associated medulloblastomas also exhibit somatic
ELP1-medulloblastoma syndrome is characterized by SHH- alte rations that converge on the p53 pathway and include
activated TP53-wildtype medulloblastoma duri ng chi ldhood . am plification of the PPMW (31%) and MOM4 (10%) genes,
The median patient age at diagnosis of medulloblastoma is yet are mutually exclusive with somatic and germline TP53
6 years (reported range : 2-19 years). Patients with ELP1-asso- mutations. ELP1 is the largest subunit of the highly conserved
ciated medulloblastoma can have a fam ily history of medullo- eukaryotic Elongator complex /682). which catalyses transla-
blastoma (3393}. tional elongation through transfer RNA (tRNA) modifications at
wobble position 34 \1363,2391,2878 ,3238}. ELP1-associated
Epidemiology medulloblastomas are characterized by a destabilized Elonga-
Pathogenic germline ELP1 variants are estimated to affect 1 in tor complex. loss of Elongator-dependent tRNA modifications .
1000 people worldwide (3393}. ELP1 -medulloblastoma syn- codon-dependent dysregulation of protein expression , and an
drome accounts for 1 in 7 cases of SHH -activated medulloblas- unfolded protein response [3393). consistent with Elongator
toma in children and 1 in 3 cases of SHH subgrou p 3 medul- studies in model systems {1126 ,1788,2225) .
!oblastoma {491 ,2865 ,3083 ,3393}.
Macroscopic appearance
Etiology ELP1-associated medulloblastomas are similar in appearance
ELP1-medulloblastoma syndrome is caused by a heterozy- to sporadic medulloblastomas.
gous pathogenic germli ne sequence variant in the ELP1 gene,
located on chromosome 9 at position 9q31 .3. Histopathology
Patients with ELP1-medullob lastoma syn d rome are pri marily
Pathogenesis diagnosed with desmoplastic/nodular medulloblastoma (76°0)
£LP1-associated medulloblastomas are characterized by bial - and less common ly with class ic (18%) or large cel l / anaplast1 c
lelic inactivation of ELP1 due to somatic loss of chromosome (6 %) medulloblastoma [3393 )
arm 9q (100% of reported cases to date). The majority (84% of
reported cases) of ELP1-associated medulloblastomas acquire Cytology
an additional sequence variant or focal deletion in the tumour Evaluation ot cerebrosp1nal tlu1d cytology is requir e-.i fur :::;L'l.':J
suppressor gene PTCH1 , located on chromosome 9 at position 1ng .
9q22 3. and they are thus predisposed to con stitutive activa-
tion of SHH signalling during tumour development A tl1ree - Diagnostic molecular pathology
!i\ep model of tumorigenesis has been proposed for E:L P I - l v.c ' cll1h'.' J J,.r. p2tl ~01 ~ 1 11\ q,Jr r1 I r)t" \,1r1_111ts 111 th ,.., Eu-11 Jt't k '
associated medulloblastomas !339:3! . step 1. heterozyu .>us ll rt.• kn!_l·.\f! ' 1\ 1 .'k'lu,i ' ti(.1 1n,,,,::..;111 tt 11~i11::~t::·11 ...:;~; ~ t11 I l:a1k.1n,l'dl

l 11 I r
' ' l I I • • • "·'I J ( 11' l N::; 48 1
spli ce -site vari ants 1 :3;~9:3 1 Pathoqenir/ m1s':.~n5P, 1ar1~r, •r, , r ,~
stru ctural variants are 1dentif1ed 1n 10"/o of cac;ec; .Ar/·. Ar r,r. ,.,,
ELP1 gene and protein expression 1rt reseo~rJ turnr)•Jr rri~ ':' ::jl
allows for the ident1f1cat1on of patients with ELP 1 m~'11J'lr,t"J'P.'",
tom a syndrome. Patients with ELP 1-medullobla-;tr)rna c; rjrrp ~ r
show loss of chromosome arm 9q and typ1callif a c;rp·.;,t r:. 1

PTCH1 mutation in their SHH -activated medullobla,.,tr;rr .::l . 1~·


no germline PTCH1 mutation .

Essential and desirable diagnostic criteria


See Box 14.21.

Staging
Clinical staging procedures include MRI examina ior.s o tri.c:?
CNS with contrast agent. This is complemented by cerebrosp -
nal fluid cytology at the time of diagnosis. The postopera r'Je
staging system developed by Chang and others 1n 1969 15191
which defines the following degrees of metastatic spread rs st ·1
being used :
fig. 14.41 Histopathology of ELP1-associated medulloblastoma. Desmoplastic/
nodular pattern characterized by nodular. reticulin-free zones and intervening densely MO No evidence of gross subarachnoid or haematogenous
packed. poorly differentiated cells that produce an lntercellular network of reticulin-
metastasis
positive collagen fibres.
M1 Microscopic tumour cells found in the cerebrospmaJ f!uld
M2 Gross nodular seeding demonstrated in the cerebellar/
Box14.21 Diagnostic criteria for ELP1-medulloblastoma syndrome cerebral subarachnoid space or in the thtrd or lateral
ventricles
Essential:
M3 Gross nodular seeding in the spinal subarachnoid space
Heterozygous pathogenic germline variant in the ELP1 gene in the context of a sonic
M4 Metastasis outside the cerebrosp inal axis
hedgehog (SHH)-activated, TP53-wildtype medulloblastoma
Desirable: Prognosis and prediction
Loss of heterozygosity of the ELP1 gene in resected tumour material and a Prelimi nary data indicates that ELP1-associated medulloblas-
methylation profile consistent with SHH-activated medulloblastoma subgroup 3 toma is associated with a favourable clinical outcome (5-year
{3393) overall survival rate: 92%) {3393}.

•H'i/ " • I \I' \1 ' (' '• ' - I r ' J '.'I l'j \1it' (; I\)'..)
Contributors

ABEDALTHAGAFI, Malak S. CARNEIRO, Fatima


BIEGEL, Jaclyn A.
l\1ng Abdulaziz City for Science and
Children's Hospital Los Angeles lpat1mup/i3S
Technology (KACST) and Rua Julio Amaral de Carvalho . 45
Department of Pathology
King Fallad Medical City (KFMC) 4200-135 Porto
4650 Sunset Bouleva rd , MS 173
King Abdullah Road . Al Raed
Los Ange les CA 90027
Riyadh 11442 CHAN , John K.C.
BLOMCKE, Ingmar Queen Elizabeth Hospital
AHLUWALIA, Manmeet S.* University Hospitals Erlangen 30 Gascoigne Road
Cleveland Clinic Kowloon , Hong Kong SAR
Schwabachanlage 6
9500 Euclid Avenue, CA5 91054 Erlangen
Cleveland OH 44145 CHEUNG, Annie Nga-Yin
BOUVIER , Corinne University of Hong Kong
ALDAPE, Kenneth D. H6pital de la Timone 2 Queen Mary Hospital
National Cancer Institute 265 Rue Saint-Pierre Pok Fu Lam Road
9000 Rockville Pike 13385 Marseille Cedex 05 Hong Kong SAR
Bethesda MD 20892
BRANDNER, Sebastian CHIM ELLI, Leila
ALEXANDRESCU,Sanda UCL Queen Square Institute of Neurology and State Institute of Brain
Boston Children's Hospital National Hospital for Neurology and Rua do Resende 156
Harvard Medical School Neurosurgery, University College London Rio de Janeiro RJ 20231-092
300 Longwood Avenue, Bader 104 Hospitals NHS Foundation Trust
Boston MA 02115 Queen Square CIMINO, Patrick J.
London WC 1N 3BG University of Washington
ASA, Sylvia L. * 325 Ninth Avenue, Box 359791
Case Western Reserve University BRASTIANOS, Priscilla Kaliopi* 98104 Seattle WA
111 00 Euclid Avenue Massachusetts General Hospital and
Cleveland OH 44106 Harvard Medical School CLAUS, Elizabeth B.
55 Fruit Street, Yawkey 9E Yale School of Medicine
BAKER, Suzanne J. Boston MA 02114 60 College Street . Box 208034
St Jude Children 's Researc h Hospital New Haven CT 06520-8034
262 Danny Thomas Place BRAT, Daniel J.
Memphis TN 381 05 Northwestern University CLIFFORD, Steven C.
Feinberg School of Medicine Newcastle University Centre for Cancer
BANDOPADHAYAY, Pratiti* 303 East Chicago Avenue Herschel Building
Dana-Farber/Boston Children 's Ward Building , 3-140 Newcastle upon Tyne NE1 ?RU
Cancer and Blood Disorders Center Chicago IL 60611
450 Brookline Avenue , Mayer 658 COTTER. Jennifer A.
Dana-Farber Cancer Institute CAH ILL, Daniel Patrick* Ch ildren 's Hospital Los Angeles and
Boston MA 02215 Massachusetts General Hospital l<eck School of Med1c1ne ot USC
55 Fruit Street. Yawkey 9E 4650 Sunset Boulevard , Mall Stop #43
BASTIAN, Boris C.* Boston MA 02114 Los Angeles CA 90027
University of California, San Franc isco
1450 Third Street, #281 CAIRNCROSS, John Gregory* CREE, Ian A.
San Francisco CA 94143-3118 University of Calgary International Agency tor Re::,earch 011 Cancer
3280 Hospital Drive North-Wtist , HRIC 2AA-20 150 Cours A.lb8rt Thom3s
BATCHELOR, Tracy* Calgary AB T2N 4Z6 69372 Lyon
Brigham and Women's Hospital
60 Fenwood Road, Hale Bu ilding, 4th Floor CALONJE, Jaime E. DE ALAVA, Enrique
Boston MA 02115 St John's Institute of Dermatology H0s1-11tal LJ111 tJt'-'1t1no Virgen L1el Roc10-IB1S
St Thomas· Hosp1 tal Lin ers1tv ut Sevil1e
BAUMHOER, Daniel Westm111ster Bridge Roac.J tv1anu-JI ~ 1 u1 u ~. 11
U'1ivers1ty Hospital Basel London SE 1 ?El-I ·1 i'I I J S»v1llc
Schonbeinstrasse 40
4031 Basel CAPPER, David* uL K CK, L nne
Chan te - U 111var~~1tdts1Pe ..J1/111flc:1111
Char11«:1platz I h\. ,,, _r'I Ir , L'r-l
1011 7 Berlin ..'l''
~, I. I .. Ir ., Ir I I • ,- j
> t • ' ' > ' I Jr-- , j 1-1\•

'Indicates disclosure of interests (::, e€ f.1 ..lH'1)

C ontributors 483
DECKERT, Martina FIGARELLA-BRANGER, Dominique GILL, Anthony J .
Faculty of M dic1ne and Assistance Publique des H6p1 tau x de Royal North Shore Hoc;p1t8I
Universit Hospital of Cologne Marseille Pacific H1qhway
Uni ersity of Cologne 264 Rue Saint-Pierre St Leonards NSW 2065
Kerpene1 Straf3 6' 13005 Marseille
50924 Colo ne GUPTA, Klrti
FISHER, Michael J. Postgraduate Institute of Medri:al rrJ Jr,.;.;·
DEMICCO, Elizabeth G. Children's Hospital of Philadelphia and Resea rch
Uni ers1ty of Toronto 3501 Civic Center Boulevard 5th Floor , A Block (Research)
Mount S1na1 Hospital, 600 University Avenue Philadelphia PA 19104 PGIMER , Sector 12
Toronto ON MSG 1X5 Chandigarh 160012
FLANAGAN, Adrienne Margaret
DRY, Sarah M. Royal National Orthopaedic Hospital GUTMANN, David H.
University of California, Los Angeles (UCLA) Brackley Hill Wash ing ton University School of M~·c re::
13-222 CHS, 10833 Le Conte Avenue Stanmore , Middlesex HA? 4LP 660 South Euclid Avenue. Box 8111
Los Angeles CA 90095 St. Louis MO 63110
FOLPE, Andrew L.
EAGLE, Ralph C. Jr Mayo Clinic HABERLER, Christine
Wills Eye Hospital 200 First Street South-West Medical University of Vienna
840 Walnut Street, Suite 141 O Rochester MN 55905 Wahringer Gurtel 18-20
Philadelphia PA 19107 1090 Vienna
FOULKES, William D.
EBERHART, Charles G. McGill University HAINFELLNER, Johannes A.*
Johns Hopkins University Research Institute Medical University of Vienna
720 Rutland Avenue, Ross Building 558 McGill University Health Centre Wahringer Gurtel 18-20
Baltimore MD 21205 1001 Decarie Boulevard 1090 Vienna
Montreal QC H4A 3J 1
ELLISON, David W. HARTMANN, Christian*
St. Jude Children's Research Hospital FRITCHIE, Karen J. Institute of Pathology
262 Danny Thomas Place Cleveland Clinic Hannover Medical School
Memphis TN 38105 9500 Euclid Aven ue Carl-Neuberg-Stra/3e 1
Cleveland OH 44 195 30625 Hannover
ENG, Charis E.
Cleveland Clinic FULLER, Gregory N. HASSELBLATT, Martin
9500 Euclid Avenue, NE-50 University of Texas University Hospital Munster
Cleveland OH 44195 MD Anderson Cancer Center Pottkamp 2
1515 Holcombe Bou levard , Un it 85 48149 Munster
EVANS, D. Gareth R. Houston TX 77030
University of Manchester HAWKINS, Cynthia E.
Oxford Road GESSI, Marco Hospital for Sick Children
Manchester M 13 9WL Fondazione Policlinico Universitario 555 University Avenue
"Agostino Gemelli" IRCCS Toronto ON MSG 1XS
FANBURG-SMITH, Julie C. Universita Cattolica del Sacra Cuore
Penn State Health Largo Agostino Gemelli 8 HILL, 0 . Ashley*
Milton S. Hershey Medical Center 00168 Rome RM Children 's National Hospital
Pathology, Pediatrics, Orthopedics 111 Michigan Avenue North-West
500 University Drive , C7714 GIANGASPERO, Felice Washington DC 20010
Hershey PA 17033 Policlinico Umberto I, Sapienza University
and IRCCS Neuromed (Pozzilli) HIROSE, Takanori
FERRY, Judith A Viale Regina Elena 324 Kobe University Graduate School oi Med ""
Massachusetts General Hospital 00161 Rome RM 7-5-2 Kusunokl-cho , Chuo-ku. Hyogo
55 Fruit Street Prefecture
Boston MA 02114 GIANNINI, Caterina Kobe City 650-0017
Mayo Clinic
FIELD, Andrew S. (and Alma Mater Studiorum - HOANG-XUAN , Kha
University of NSW and University of Bologna) Hopital Universitaire P1tie Salpemere
Un1ve::rs1ty of Notre Darne Medical Scllools, 200 Fi rst Stree t South-We st Division Mazarin , 47 Boulevard de l'H
D"";::irirtrr18r1t rJf Ana1orn1cal Patl w logy Rochester MN 55905 75013 Paris
St 1/11 .u-11t , Hu'.1)11al Sydney
GILBERTSON, Richard James* HONAVAR, Mrinalini
University of Cambridge Pedro Hispano Hospital
Lt f<a Sh1ng Centre, Robinson Way Rua de Alfredo Cunha 365
Cambridge CB2 ORE 4464 -5 13 Matos1nhos
REYES-MUGICA, Miguel
SANTAGATA, Sandro SNUDERL, Matija
university of Pittsburgh Medical Center
Brigham and Women's Hos pital t'- IYU Langone Health
4401 Penn Avenue , Main Hospita l 8260 60 Fenwood Road , Hale 8002P 2'1 0 East 38th Street, 22nd Floor
one Chil dren's Hospital Drive
Bos ton MA 0211 5 f\lew York NY 10016
Pittsburgh PA 15224
SANTOSH, Vanl SOARES, Fernando Augusto
RIGHI. Alberto National Institute of Rede D'Or Hospitals
IRCCS. lstituto Ortopedico Ri zzoli
Mental Health and Neurosci ences Rua das Perobas 266
\·ra di Barb1ano 1/10 Hosur Road Sao Paulo SP 04321-120
40136 Bologna BO Bengaluru 560029
SOFFIETII , Riccardo
RODRIGUEZ, Fausto J. SARKAR , Chitra University of Turin and
,Johns Hopkins University All India Institute of Medical Sciences Ci ty of Health and Science Hosp1t.:31 Turrr
Sheikh Zayed Tower, Room M2101 An sari Nagar Via Cherasco 15
1800 Orleans Street New Delhi 110029 10 126 Turin TO
Baltimore MD 21231
SCHUHMANN, Martin Ulrich SOLOMON . David A.
RONCAROLI, Federico R. University Hospital TUbingen Unive rsity of California , San Francisco
University of Manchester Hoppe-Seyler-StraBe 3 5 13 Parn assus Avenue . HSW 451
Oxford Road 72076 TUbingen San Francisco CA 94143
Manchester M 13 9PT
SCHOLLER, Ulrich SRIGLEY, John R.
ROSENBERG, Andrew E. University of Hamburg Trillium Health Partners
Miller School of Medicine MartinistraBe 52 Cred it Val ley Hospital Site
University of Miami 20246 Hamburg 2200 Eglinto n Avenue West
1400 North-West 12th Avenue Mississauga ON L5M 2N 1
Miami FL 33136 SCHULTZ, Kris Ann P.
International PPB/0/CER1 Registry STEMMER-RACHAMIMOV, Anat Olga
ROSENBLUM, Marc K. Cancer and Blood Disorders Massachusetts General Hospital
Memorial Sloan Kettering Cancer Center Children's Minnesota 55 Fruit Street
275 York Avenue 2530 Chicago Avenue South Boston MA 02114
New York NY 10021 Minneapolis MN 55404
STURM , Dominik
ROTONDO, Fabio SCIOT, Raf Hopp Ch ildren 's Cancer Center Heidelberg
St Michael's Hospital Department of Pathology (KiTZ) , German Cancer Research Center
30 Bond Street University Hospital KULeuven (DKFZ), and Heidelberg Un1vers1ty Hosp1ral
Toronto ON M5B 1W8 Herestraat 49 Im Neuenheimer Feld 280
3000 Leuven 69120 Heidel berg
ROUS, Brian
Public Health England SHARMA, Mehar C. SUVA, Mario L.
Victoria House, Capital Park All India Institute of Medica l Sciences Massach usetts General Hospital
Fulbourn , Cambridge CB21 5XA Ansari Nagar 149 13th Street, Office 6.010
New Delhi 110029 Boston MA 02129
RUDA, Roberta
University of Turin and SHIBUYA, Makoto TABORI, Uri
City of Health and Science Hospital , Turin Tokyo Medical University Hospital for Sick Children
Via Cherasco 15 Hachioji Medical Center 555 University Avenue
10126 Turin TO 1163 Tatemachi Toronto ON MSG -1xs
Hachioji City 193-0998
RUDZINSKI, Erin R. TAN , Puay Hoon
Seattle Children 's Hospital SIEVERS, Philipp D1 v1sion of Pathology

I
1
4800 Sandpoint Way North-East
Se:attle WA 98105
Heidelberg University and
German Cance r Research Center (DKFZ)
Singapore General Ho::>p1tal
20 College Road .\cadt:H%1 Level 7
Im Neuenhei mer Feld 224 D1agno tics Tower
SAHM, Felix 69120 Heidelberg Sin~ -tµore 169Rbt:l

I Heidelberg University and


G~rrnan Cancer Research Center (DKFZ)
lrr, Neuenheimer Feld 224
SINGH, Rajendra
Northwell Health
TANA , Shinya
~k')l._",l 1..1l1 IJ~l'.v l)

l 69120 Heidelberg

SANSON, Marc
P,\,f::.~alpetnere Hospital - Sorbonne Univ«r ~.1tr•
19') 1 Marcus Avenue, Suite 300
Lakt.' SuL.ce:::s NY 11042
l )r <.H.ll!.:l!t.' l ~L'.11\ 11..11
N 15 v-:
I ·r f\ t' 1 . II ·~

47 8
3 Boulevard de l'H6p1tal
1'lJ13 Paris

Indicates disclosure of int eres ts ( set-: P tl/J',,

Contributors 487

You might also like