chromatin remodeling defects in pediatric and young adult glioblastoma: a tale of a variant histone...

7
MINI-SYMPOSIUM: When Genetics Meets Epigenetics—A New Option for Therapeutic Intervention in Brain Tumors? Chromatin Remodeling Defects in Pediatric and Young Adult Glioblastoma: A Tale of a Variant Histone 3 Tail Adam M. Fontebasso 1 ; Xiao-Yang Liu 2 ; Dominik Sturm 3 ; Nada Jabado 1,2,4 1 Division of Experimental Medicine, McGill University and McGill University Health Centre, Montreal, QC, Canada. 2 Department of Human Genetics, McGill University and McGill University Health Centre, Montreal, QC, Canada. 3 Division of Pediatric Neuro-oncology, German Cancer Research Centre (DKFZ), Heidelberg, Germany. 4 Department of Pediatrics, McGill University and the McGill University Health Centre Research Institute, Montreal, QC, Canada. Keywords ATRX, chromatin remodeling, glioblastoma, H3F3A, pediatric, TP53, young adult. Corresponding author: Nada Jabado, MD, PhD, Department of Pediatrics, McGill University and McGill University Health Centre, 4060 Ste-Catherine West, PT-239, Montreal, Quebec, Canada H3Z 2Z3 (E-mail: [email protected]) Received 28 December 2012 Accepted 29 December 2012 doi:10.1111/bpa.12023 Abstract Primary brain tumors occur in 8 out of 100 000 people and are the leading cause of cancer-related death in children. Among brain tumors, high-grade astrocytomas (HGAs) including glioblastoma multiforme (GBM) are aggressive and are lethal human cancers. Despite decades of concerted therapeutic efforts, HGAs remain essentially incurable in adults and children. Recent discoveries have revolutionized our understanding of these tumors in children and young adults. Recurrent somatic driver mutations in the tail of histone 3 variant 3 (H3.3), leading to amino acid substitutions at key residues, namely lysine (K) 27 (K27M) and glycine 34 (G34R/G34V), were identified as a new molecular mechanism in pediatric GBM. These mutations represent the pediatric counterpart of the recurrent mutations in isocitrate dehydrogenases (IDH) identified in young adult gliomas and provide a much-needed new pathway that can be targeted for therapeutic development. This review will provide an overview of the potential role of these mutations in altering chromatin structure and affecting specific molecular pathways ultimately leading to gliom- agenesis. The distinct changes in chromatin structure and the specific downstream events induced by each mutation need characterizing independently if progress is to be made in tackling this devastating cancer. Primary brain tumors (originating in the brain) are the leading cause of cancer-related death in children under the age of 20, now surpassing leukemia, and the third leading cause of cancer-related death in young adults aged 20–39 years (61). Astrocytomas are the most common primary brain tumor across the lifespan, and the highest grade, glioblastoma multiforme (GBM), remains essen- tially incurable despite decades of concerted therapeutic efforts (14). Similar histology of pediatric and adult tumors has caused current childhood GBM treatments to be fueled by adult studies, and show, across the board, little therapeutic advance. One impedi- ment to treatment is that GBM is diagnosed as a single entity by pathologists who cannot discriminate potential genetic drivers and molecular subtypes. This impacts the design and outcome of clini- cal trials and it likely contributes to the apparent inherent resist- ance of GBM to adjuvant therapies and poor progress in improving survival or the quality of life of patients and their families. Other crucial obstacles to progress are the lack of reliable in vitro and in vivo models for pediatric GBM based on the lack of identified genetic drivers specific to children until very recently. Conse- quently, better stratification of patients based on tumor biology, improved identification of relevant therapeutic targets and the design of experimental “companion” models to test compounds affecting specific genetic/molecular drivers are essential for thera- peutic breakthroughs in this deadly disease. This review will provide a short overview of gliomas across the lifespan and recent molecular advances that led to the identification of recurrent muta- tions in H3F3A, which are genetic drivers specific to pediatric GBM, their role in chromatin remodeling and the presence of at least six distinct molecular subgroups in GBM across ages that need to be tackled separately to achieve future therapeutic success in this deadly cancer. GENERAL OVERVIEW OF GLIOMAS ACROSS AGE Gliomas are heterogeneous and are classified by the World Health Organization (WHO) according to the presumed cell of origin (41). WHO grade I and II gliomas are commonly referred to as low-grade gliomas (LGG), while WHO grade III (anaplastic) and IV (GBM) tumors are regarded as high-grade gliomas (41). Tumor grade, age at diagnosis and degree of surgical resection are regarded as the most important prognostic factors across the lifespan (35). The frequency, anatomic location, progression mode and pathologic spectrum of gliomas differ in children and adults. Oligodendroglial differentiation and progression of lower grade gliomas to higher grade tumors are rare in children, and the vast majority of pediatric GBM occur de novo. A significant proportion of pediatric gliomas are low-grade tumors with a high prevalence Brain Pathology ISSN 1015-6305 210 Brain Pathology 23 (2013) 210–216 © 2013 The Authors; Brain Pathology © 2013 International Society of Neuropathology

Upload: nada

Post on 06-Dec-2016

217 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Chromatin Remodeling Defects in Pediatric and Young Adult Glioblastoma: A Tale of a Variant Histone 3 Tail

M I N I - S Y M P O S I U M : W h e n G e n e t i c s M e e t s E p i g e n e t i c s — A N e w O p t i o n f o rT h e r a p e u t i c I n t e r v e n t i o n i n B r a i n Tu m o r s ?

Chromatin Remodeling Defects in Pediatric and Young AdultGlioblastoma: A Tale of a Variant Histone 3 TailAdam M. Fontebasso1; Xiao-Yang Liu2; Dominik Sturm3; Nada Jabado1,2,4

1 Division of Experimental Medicine, McGill University and McGill University Health Centre, Montreal, QC, Canada.2 Department of Human Genetics, McGill University and McGill University Health Centre, Montreal, QC, Canada.3 Division of Pediatric Neuro-oncology, German Cancer Research Centre (DKFZ), Heidelberg, Germany.4 Department of Pediatrics, McGill University and the McGill University Health Centre Research Institute, Montreal, QC, Canada.

Keywords

ATRX, chromatin remodeling, glioblastoma,H3F3A, pediatric, TP53, young adult.

Corresponding author:

Nada Jabado, MD, PhD, Department ofPediatrics, McGill University and McGillUniversity Health Centre, 4060 Ste-CatherineWest, PT-239, Montreal, Quebec, Canada H3Z2Z3 (E-mail: [email protected])

Received 28 December 2012Accepted 29 December 2012

doi:10.1111/bpa.12023

AbstractPrimary brain tumors occur in 8 out of 100 000 people and are the leading cause ofcancer-related death in children. Among brain tumors, high-grade astrocytomas (HGAs)including glioblastoma multiforme (GBM) are aggressive and are lethal human cancers.Despite decades of concerted therapeutic efforts, HGAs remain essentially incurable inadults and children. Recent discoveries have revolutionized our understanding of thesetumors in children and young adults. Recurrent somatic driver mutations in the tail ofhistone 3 variant 3 (H3.3), leading to amino acid substitutions at key residues, namelylysine (K) 27 (K27M) and glycine 34 (G34R/G34V), were identified as a new molecularmechanism in pediatric GBM. These mutations represent the pediatric counterpart of therecurrent mutations in isocitrate dehydrogenases (IDH) identified in young adult gliomasand provide a much-needed new pathway that can be targeted for therapeutic development.This review will provide an overview of the potential role of these mutations in alteringchromatin structure and affecting specific molecular pathways ultimately leading to gliom-agenesis. The distinct changes in chromatin structure and the specific downstream eventsinduced by each mutation need characterizing independently if progress is to be made intackling this devastating cancer.

Primary brain tumors (originating in the brain) are the leadingcause of cancer-related death in children under the age of 20, nowsurpassing leukemia, and the third leading cause of cancer-relateddeath in young adults aged 20–39 years (61). Astrocytomas are themost common primary brain tumor across the lifespan, and thehighest grade, glioblastoma multiforme (GBM), remains essen-tially incurable despite decades of concerted therapeutic efforts(14). Similar histology of pediatric and adult tumors has causedcurrent childhood GBM treatments to be fueled by adult studies,and show, across the board, little therapeutic advance. One impedi-ment to treatment is that GBM is diagnosed as a single entity bypathologists who cannot discriminate potential genetic drivers andmolecular subtypes. This impacts the design and outcome of clini-cal trials and it likely contributes to the apparent inherent resist-ance of GBM to adjuvant therapies and poor progress in improvingsurvival or the quality of life of patients and their families. Othercrucial obstacles to progress are the lack of reliable in vitro and invivo models for pediatric GBM based on the lack of identifiedgenetic drivers specific to children until very recently. Conse-quently, better stratification of patients based on tumor biology,improved identification of relevant therapeutic targets and thedesign of experimental “companion” models to test compoundsaffecting specific genetic/molecular drivers are essential for thera-peutic breakthroughs in this deadly disease. This review will

provide a short overview of gliomas across the lifespan and recentmolecular advances that led to the identification of recurrent muta-tions in H3F3A, which are genetic drivers specific to pediatricGBM, their role in chromatin remodeling and the presence of atleast six distinct molecular subgroups in GBM across ages thatneed to be tackled separately to achieve future therapeutic successin this deadly cancer.

GENERAL OVERVIEW OF GLIOMASACROSS AGEGliomas are heterogeneous and are classified by the World HealthOrganization (WHO) according to the presumed cell of origin(41). WHO grade I and II gliomas are commonly referred to aslow-grade gliomas (LGG), while WHO grade III (anaplastic) andIV (GBM) tumors are regarded as high-grade gliomas (41). Tumorgrade, age at diagnosis and degree of surgical resection areregarded as the most important prognostic factors across thelifespan (35). The frequency, anatomic location, progression modeand pathologic spectrum of gliomas differ in children and adults.Oligodendroglial differentiation and progression of lower gradegliomas to higher grade tumors are rare in children, and the vastmajority of pediatric GBM occur de novo. A significant proportionof pediatric gliomas are low-grade tumors with a high prevalence

Brain Pathology ISSN 1015-6305

210 Brain Pathology 23 (2013) 210–216

© 2013 The Authors; Brain Pathology © 2013 International Society of Neuropathology

Page 2: Chromatin Remodeling Defects in Pediatric and Young Adult Glioblastoma: A Tale of a Variant Histone 3 Tail

of grade I astrocytomas and rare grade II astrocytomas, whereas inadults, high-grade gliomas represent the vast majority of primarycentral nervous system tumors. Tumors in the optic pathway, and ininfra-tentorial locations such as the cerebellum and brainstem arethe most common localizations in children, whereas supra-tentorial tumors are more frequent in adults and include locationssuch as the thalamus and cerebral cortex. This is of importance asthe cell of origin, the microenvironment and chromatin conforma-tion will differ based on age, cell type and anatomical localizationand are, as we have shown, relevant in pathogenesis (8, 36, 40, 59,60). We and others have shown that grade I tumors, which repre-sent ~25% of all pediatric brain tumors, are characterizedby genetic alterations in the mitogen-activated protein kinase(MAPK) pathway (27, 31, 32, 53) and constitute one biologicalparadigm (13, 24, 27, 28). Grade II astrocytomas are rare in chil-dren (<5% of brain cancers) and high-grade astrocytomas (HGA,grades III and IV) account for ~10%–15% of all pediatric braintumors when combined with diffuse intrinsic pontine gliomas(DIPG), which are high-grade gliomas that occur in the brainstem,with an incidence close to 0.6/100 000 in children aged 0–19 yearsand a dismal mortality rate close to 90% at 5 years (58). Thera-peutic advances remain low and this high mortality occurs regard-less of patient’s age. This is despite numerous clinical trialscombining surgery, radiotherapy, chemotherapy and, in recentyears, the development of many therapies including temozolo-mide, and targeted agents such as receptor tyrosine kinase andangiogenesis inhibitors, suggesting that these efforts are not suf-ficient and/or adapted to counter tumor progression.

GBM TUMORS IN CHILDREN ANDYOUNG ADULTS ARE NOT A SINGLEDISEASE AND RESULT FROMMUTATIONS IN GENES AFFECTINGCHROMATIN REWIRINGThe Cancer Genome Atlas project (TCGA) and other groupsperformed genomic and epigenomic analysis and targetedsequencing of adult GBM samples (1, 7, 9, 48, 50, 68). Theyrevealed GBM in adults to be highly heterogeneous and identifiedthe crucial role for isocitrate dehydrogenase 1 or 2 (IDH) meta-bolic pathways in the genesis of secondary GBM, while EGFRamplification and gain of function mutations as well as PTENand/or CDKN2A/B loss target de novo adult GBM. Indeed, recur-rent IDH1 and 2 mutations are found in up to 80% of grade II andIII adult gliomas and secondary GBM but rarely in GBM occur-ring de novo (primary GBM) (50, 68). Pediatric GBM are mor-phologically indistinguishable from adult GBM, which promptedclinicians and investigators to view them as the same disease. Wehave, however, disproved this assumption and our group and othersdemonstrated the importance of analyzing the unique biologyof these tumors in children. Indeed, while mutations in TP53,CDKN2A and PIK3CA are common to all HGA, PTEN mutationsand EGFR amplifications occur in less than 10% of pediatric GBM(54, 55), while IDH mutations are rare in children (less than 5% ofGBM) and their frequency is higher in adolescents (56). Pediatricand adult HGA have different gene expression profiles and DNAcopy number alterations (2, 20, 23, 52, 57) including a higherproportion of amplification of PDGFRA in children, especiallyafter radiation therapy (52). Up to recently, DIPG, which occur

mainly in children aged between 5 and 10 years and have a par-ticularly grim prognosis with an overall survival rate at 2 years lessthan 10%, have been considered to be molecularly distinct fromsupra-tentorial GBM (51, 69).

Using next-generation sequencing technology, we recently iden-tified a new molecular mechanism driving GBM in children,namely two recurrent heterozygous somatic mutations in H3F3A,which encodes the replication-independent histone 3 variant H3.3(59). These mutations lead to amino acid changes in key residues(K27M, G34R/G34V) and were identified in 35% of supra-tentorial pediatric and young adult HGA (59). This is the firstreport to identify mutations in a regulatory histone in humans andthese mutations are the pediatric counterpart of the recurrent IDHmutations identified in young adult GBM, which indirectly affectthese histone marks (50, 68). H3.3 mutations significantly over-lapped with mutations in TP53 and in ATRX (a-thalassemia/mental retardation syndrome-X-linked) (59) and less frequentlyin the ATRX heterodimer DAXX, which encodes subunits of achromatin remodeling complex required for H3.3 incorporation atpericentric heterochromatin and telomeres (16, 39). Using an inde-pendent cohort of ~790 gliomas across age, group and grade, wefurther showed H3.3 mutations to be specific to high-grade tumors,to be prevalent in children (incidence <3.4% in adult HGA) and tobe mutually exclusive with IDH mutations (59, 62). We furtheridentified that K27M-H3.3 characterize 71% of brainstem high-grade gliomas (DIPG) (34) and 80% of thalamic pediatric GBM(62). G34R/V-H3.3 in HGA and K27M-H3.3 in HGA and DIPGwere independently identified by another group, which also iden-tified K27M in the related canonical histone H3.1 in 18% of DIPG(67). H3F3A and ATRX/DAXX were not part of the close to ~600genes initially sequenced in adult GBM by the adult consortiaincluding TCGA. This and their relative specificity to childrenexplain why these mutations were not previously identified despitetheir staggering frequency. Rather than selecting and sequencinggenes thought to be important, the use of unbiased genome-widesequencing methods to cover all protein-coding genes in samplesfrom diseased and normal tissue, and the enrichment in pediatricsamples helped in identifying these mutations. Next-generationsequencing technologies have currently become the method ofchoice to interrogate a tumor genome based on coverage andcost-effectiveness. Remarkably, H3F3A mutations seem for thetime being specific to pediatric HGA and have not yet been iden-tified in other pediatric tumors including other brain tumors orleukemia (17) or in available datasets on adult cancers. However,one needs to bear in mind that most of these samples are from adultcohorts and that possibly enrichment of a specific rare tumorsubset may still reveal presence of these mutations that mayotherwise be overlooked even in children.

H3F3A AND IDH MUTATIONS ARISE INDISTINCT ANATOMICAL LOCATIONSOF THE BRAIN AND MAY ARISE FROMDISTINCT DEVELOPMENTAL TIMEPOINTS AND CELLULAR ORIGINSOur findings indicate neuroanatomical and age specificities of themutation type and the combination of mutations in this settingrelative to older age GBM and point to a developmental defect atthe origin of pediatric and young adult HGA (Figure 1) (34, 59,

Fontebasso et al Chromatin Remodeling Defects in Pediatric and Young Adult Glioblastoma

211Brain Pathology 23 (2013) 210–216

© 2013 The Authors; Brain Pathology © 2013 International Society of Neuropathology

Page 3: Chromatin Remodeling Defects in Pediatric and Young Adult Glioblastoma: A Tale of a Variant Histone 3 Tail

62). Indeed, K27M-H3.3 mutations characterize midline GBM andoccur in younger children with brainstem and thalamic tumors(70%–80% of all HGA cases in these regions, median age 10.5years, range 5–23 years) (34, 59, 62). They overlap with TP53mutations in 80% of cases regardless of location, while ATRXmutations occur in only 50% of cases and favor older children andthalamic location (only 20% of DIPG are mutant for ATRX) (34).G34R/V-H3.3 mutations are mainly found in HGA located withinthe cerebral hemispheres (34, 59, 62). They occur in patientsaround the threshold between the adolescent and adult populations(median age 18 years, range 9–42 years) and almost always overlapwith mutant TP53/ATRX. IDH alterations affect younger adults(median age 40 years, range 13–71 years) and occur in the cortex,mainly in the frontal lobes suggesting that these tumors also arisefrom a neural precursor population that is spatially and temporallyrestricted in the brain (38). IDH alterations are gain of function,heterozygous, somatic mutations and have been shown to beinitiating events in gliomas (50, 65, 66). Intriguingly, they alsoseem to require association with other genetic events to achievefull-blown tumorigenesis. They are associated with two mutuallyexclusive genetic alterations, TP53 alterations and 1p19q co-deletions (5, 49) that respectively characterize astrocytic and oli-godendroglial IDH-mutant gliomas. We (34, 40, 59) and others(29, 33) recently showed that mutations in ATRX characterize IDH-and TP53-mutant astrocytomas in young adults with low- andhigh-grade tumors. Furthermore, ATRX alterations are specific toastrocytic tumors and are mutually exclusive with CIC mutationsand loss of 1p19q, which characterize oligodendrogliomas. ATRXinactivating mutations thus characterize older children with GBMand adult IDH-mutant gliomas of the astrocytic lineage arguing forthe importance of an H3F3A & ATRX & TP53 or an ATRX &IDH1/2 & TP53 mutant phenotype in their early development andprogression and have now been found in pediatric and adult GBM(25, 59), older patients with neuroblastomas (10, 45) and in

pancreatic neuroendocrine tumors (29). Mutations in ATRX/DAXXmay interfere with H3.3 incorporation at these loci, thus compro-mising the structural integrity of the chromosome. Supportiveof this hypothesis is the presence of alternative lengthening oftelomeres (ALT), a telomerase-independent telomere maintenancemechanism, in tumors with ATRX mutations (25, 59). ALT seemsto be promoted by loss of ATRX that may increase the rate ofhomologous recombination at telomeres and pericentric hetero-chromatin and thus lead to increased lengthening of the telomeresthrough an alternate mechanism to increased telomerase activity,providing cells with the capacity for unlimited cellular prolifera-tion. Based on the role of ATRX-DAXX in H3.3 deposition inspecific areas of the chromatin, in telomere maintenance and ingenomic stability (6, 26), these findings further support the role ofchromatin modifications in the genesis of HGA in children andyoung adults.

ALTERATIONS IN THE HISTONE CODEUNDERLIE PEDIATRIC AND YOUNGADULT GBM AND INDUCE DEFECTS INCHROMATIN REMODELINGVirtually all DNA processes are regulated by the histone code,including replication and repair, regulation of gene expression andcentromere and telomere maintenance (11). Accordingly, muta-tions in genes affecting histone post-translational modifications(PTMs) are increasingly described in cancer (11). Histonespackage and organize DNA at the level of the fundamental unit ofchromatin, the nucleosome. Nucleosomes can be modulated by alarge variety of covalent PTMs mostly occurring in the N-terminaltails of histones, and also by the incorporation of histone variants(18, 21, 22). H3.3 is a universal, replication-independent histonepredominantly incorporated into transcription sites and associatedwith active and open chromatin [reviewed in (18, 63)]. H3.3 not

Figure 1. Neuroanatomical and age specificity of isocitrate dehydroge-nase (IDH), K27M-H3.3 and G34R/V-H3.3 mutations in high-grade astro-cytomas (HGA). K27M-H3.3 or H3.1 (yellow stars) occur mainly inbrainstem HGA and K27M-H3.3 mainly thalamic HGA (70%–80% of allGBM in these locations). This H3.3 mutation is inconsistently associatedwith ATRX mutations. G34R/V-H3.3 occur mainly in the cerebral hemi-spheres similar to IDH mutations which have been previously shown to

have a predilection for the frontal cortex. Both H3.3 mutations aresignificantly associated with ATRX mutations (purple filled circles) andTP53 mutations (not represented here for clarity issues). IDH and H3.3mutations represent distinct disease entities that possibly arise fromseparate cellular origins as the result of largely non-overlapping sets ofmolecular events. Note: the size of the shape illustrating each mutationis approximately proportional to the % identified in (34, 59, 62).

Chromatin Remodeling Defects in Pediatric and Young Adult Glioblastoma Fontebasso et al

212 Brain Pathology 23 (2013) 210–216

© 2013 The Authors; Brain Pathology © 2013 International Society of Neuropathology

Page 4: Chromatin Remodeling Defects in Pediatric and Young Adult Glioblastoma: A Tale of a Variant Histone 3 Tail

only functions as a neutral replacement histone, but in additionparticipates in the epigenetic transmission of active chromatinstates and is associated with chromatin assembly factors in large-scale replication-independent chromatin remodeling mechanisms(37, 42). Its role in histone replacement at active genes and pro-moters is conserved in the single histone H3 present in yeast,indicating its importance throughout evolution (18). This histonevariant is actively loaded in the developing brain, replacing otherresident histones 3.1 or 3.2 (4). Several PTMs regulate histonefunction in the nucleosome. Reversible methylation of severalhistone lysine residues is mediated by distinct histone methyltrans-ferases or demethylases to specific histone Lys (K) or Arg residues.Polycomb repressor complex 2 (PRC2) recruitment is associatedwith methylation at K27 that represses transcription and cell dif-ferentiation (3). Gain of function mutations in EZH2, the mainK27 trimethyltransferase, has been identified in leukemias andlymphomas (44, 46, 70), and overexpression of this gene was alsoidentified in many cancers including medulloblastoma (30, 47).Thus, altered methylation or loss of acetylation of K27 is a poten-tial driver of gliomagenesis. Methylation of K36 has been widelyassociated with active chromatin but also with transcriptionalrepression, alternative splicing, DNA replication and repair, DNAmethylation and imprinting, and the transmission of memory ofgene expression from parents to offspring [reviewed in (4)].H3.3K36 methylation is potentially disrupted by the G34R/V-H3.3mutation. Indeed, glycine 34 of histone H3 (H3G34) lies in closeproximity to lysine 36 (H3K36), a residue that regulates transcrip-tional elongation. The G34R/V-H3.3 mutation may impact theability of histone-modifying complexes to methylate or acetylateH3K36, thereby altering the transcription of several target genes.Strikingly, our gene expression analysis revealed different geneexpression patterns between samples with the K27M-H3.3 muta-tion vs. samples with the G34R/V-H3.3 mutation, suggesting thateach mutation favors the expression of a specific genetic program.IDH mutations that give a neomorphic function to these enzymesenable the generation of high quantities of 2-hydroxyglutarate(15), an oncometabolite. This oncometabolite, in turn, competi-tively inhibits the activity of histone demethylases (43), affectingchromatin structure through alteration of histone PTMs. Interest-ingly, the oncometabolite produced by IDH1 mutations impairshistone demethylation, affecting UTX (H3K27me) and KDM4A/JMJD2A (H3K36/K9me) resulting in a block to cell differentiation(12), further supporting a role in gliomas for altered methylation ofthese residues.

INTEGRATED EPIGENETIC ANDGENETIC ANALYSIS IDENTIFIES ATLEAST SIX BIOLOGICALLY DISTINCTGBM SUBGROUPS ACROSS AGESAltogether, findings from several groups converge to indicate thatalterations of the histone code are at the origin of pediatric andyoung adult HGA. DNA methylation patterns are better correlatedwith histone lysine methylation patterns than with the underlyinggenome sequence context (19) and, in recent years, a distinctglioma-CpG-island methylator phenotype (G-CIMP) was identi-fied and found to correlate with IDH1-mutant gliomas (48, 64). Ourinvestigation of the genome-wide DNA methylation patterns usingthe Illumina 450k methylation array in a cohort of 210 GBM from

children and adult patients (62) sub-classified GBM into at least sixdistinct epigenetic groups, indistinguishable by histological appear-ance, but correlating with molecular genetic factors as well as keyclinical variables such as patient age and tumor location. Indeed,IDH1 mutations characterized, as expected, a mutation-definedsubgroup which included pediatric IDH-mutant GBM, while eachH3F3A mutation defined an epigenetic subgroup of GBM with adistinct global methylation pattern. The last three epigenetic sub-groups were enriched for hallmark genetic events of adult GBMand/or established transcriptomic signatures. In line with reportsof PDGFRA copy number alterations being more prevalent inchildhood high-grade gliomas (2, 52, 57), an epigenetic cluster, wenamed receptor tyrosine kinase (RTK) I “PDGFRA,” harbored aproportion of pediatric patients (median age 36 years, range 8–74 years). A mesenchymal cluster displayed a widespread agedistribution (median age 47, range 2–85 years), while an RTK II“Classic” cluster was mostly composed of older adults (medianage 58, range 36–81 years) patients. Interestingly, the two H3F3Amutations gave rise to GBMs with differential regulation of tran-scription factors—OLIG1, OLIG2 and FOXG1, which may reflectdifferent cellular origins. Last, our results demonstrate differentialsurvival for each of these six subgroups, with worse prognosis andrapid death for K27M tumors, which behave like DIPG regardlessof their localization within the brain, and a slightly improvedsurvival for IDH-mutant GBM which show the longest overallsurvival followed by G34R/V-H3.3 GBM, while wild-type tumorsfor these mutations have the expected bad prognosis seen in GBM.

CONCLUSIONS AND FUTUREPERSPECTIVESCurrently, there are no effective treatments for pediatric HGAwhich carry the highest rate of morbidity and mortality in child-hood cancer. Collectively, recent findings have changed the land-scape for this disease, providing at last relevant targets that willdrive new trial models based on well-characterized gene subsets,thus providing hope for future change in the management andoutcome of this deadly group of cancers. Defects in chromatinremodeling are central in the genesis of pediatric and young adultHGA, and age and brain-location specific defects in chromatinstructure underlie the genesis of these tumors. This provides alaunching point for the identification of diagnostics and therapiesspecific to the molecular subtypes in children and young adults.Although histologically indistinguishable, there are at least sixdistinct disease entities that may arise from separate cell types oforigin as the result of largely non-overlapping sets of molecularevents. The development of the normal brain is a very dynamic andcomplex process, involving numerous extracellular factors that arepresent at precise times in specific brain locations. This is of majorsignificance as the human brain continues to develop post-natally,reaching completion in the early to mid-20s. What is unique aboutthe developing brain that enables K27M-H3.3 mutations to betumorigenic mainly in children and G34R/V-H3.3 and IDH muta-tions to affect mainly adolescents and younger adults? Why theneed for ATRX mutations that seem to be mutation (G34R/V-H3.3and IDH more than K27M-H3.3), and/or location (cortex andthalamus more than the brainstem) and/or age (older more thanyoung children) specific? Why the need for added TP53 muta-tions? Is it possible that the different extracellular factors in the

Fontebasso et al Chromatin Remodeling Defects in Pediatric and Young Adult Glioblastoma

213Brain Pathology 23 (2013) 210–216

© 2013 The Authors; Brain Pathology © 2013 International Society of Neuropathology

Page 5: Chromatin Remodeling Defects in Pediatric and Young Adult Glioblastoma: A Tale of a Variant Histone 3 Tail

developing brain tissue contribute to the transformation of cellswith K27M-H3.3 and G34R/V-H3.3 mutations, allowing them toform tumors? The timing, the contribution and the geneticprogram that are altered by these mutations/combinations of muta-tions need further investigation to help address and better targettheir effects in HGA. Further dissection of the effects downstreamof each H3F3A mutation in primary tumors and the generation ofmuch needed “in vitro” and “in vivo” models of these mutationsand their interaction(s) with ATRX and TP53 alterations are thuswarranted. This will help us better tackle this cancer as uncoveringwhat they induce on the epigenetic/genetic level has major poten-tial to improve our understanding of HGA, brain development andpotentially other diseases affecting the epigenome. Optimal clini-cal management should account for the distinction between theseGBM disease subtypes, and as routine histopathology is unable todistinguish these genetic subgroups, use of molecular tools wouldresult in better stratification of patients and enable better therapeu-tic choices as they become available.

CONFLICT OF INTERESTThe authors declare no conflict of interest.

ACKNOWLEDGMENTSThis work was funded in part by Genome Canada and the Cana-dian Institute for Health Research (CIHR) with co-funding fromGenome BC, Genome Quebec, CIHR-ICR (Institute for CancerResearch) and C17, through the Genome Canada/CIHR jointATID Competition (project title: The Canadian Paediatric CancerGenome Consortium: Translating next generation sequencingtechnologies into improved therapies for high-risk childhoodcancer) and the PedBrain project contributing to the InternationalCancer Genome Consortium funded by the German Cancer Aid(109252). AMF and XYL are the recipient of studentship awardsfrom the CIHR. NJ is the recipient of a Chercheur Clinicien Awardfrom Fonds de Recherche en Santé du Québec and a “Next Gen-eration Champion of Genetics” award from the Canadian GeneCure Foundation.

REFERENCES1. Cancer Genome Atlas Research Network (2008) Comprehensive

genomic characterization defines human glioblastoma genes andcore pathways. Nature 455:1061–1068.

2. Bax DA, Mackay A, Little SE, Carvalho D, Viana-Pereira M,Tamber N et al (2010) A distinct spectrum of copy numberaberrations in pediatric high-grade gliomas. Clin Cancer Res16:3368–3377.

3. Bernstein BE, Mikkelsen TS, Xie X, Kamal M, Huebert DJ,Cuff J et al (2006) A bivalent chromatin structure marks keydevelopmental genes in embryonic stem cells. Cell 125:315–326.

4. Bosch A, Suau P (1995) Changes in core histone variantcomposition in differentiating neurons: the roles of differentialturnover and synthesis rates. Eur J Cell Biol 68:220–225.

5. Bourne TD, Schiff D (2010) Update on molecular findings,management and outcome in low-grade gliomas. Nat Rev Neurol6:695–701.

6. Bower K, Napier CE, Cole SL, Dagg RA, Lau LM, Duncan EL et al(2012) Loss of wild-type ATRX expression in somatic cell hybridssegregates with activation of alternative lengthening of telomeres.Plos One 7:e50062.

7. Bredel M, Scholtens DM, Harsh GR, Bredel C, Chandler JP,Renfrow JJ et al (2009) A network model of a cooperative geneticlandscape in brain tumours. JAMA 302:261–275.

8. Cha TL, Zhou BP, Xia W, Wu Y, Yang CC, Chen CT et al (2005)Akt-mediated phosphorylation of EZH2 suppresses methylation oflysine 27 in histone H3. Science 310:306–310.

9. Chen Y, Lin MC, Yao H, Wang H, Zhang AQ, Yu J et al (2007)Lentivirus-mediated RNA interference targeting enhancer of zestehomolog 2 inhibits hepatocellular carcinoma growth throughdown-regulation of stathmin. Hepatology 46:200–208.

10. Cheung NK, Zhang J, Lu C, Parker M, Bahrami A, Tickoo SK et al(2012) Association of age at diagnosis and genetic mutations inpatients with neuroblastoma. JAMA 307:1062–1071.

11. Chi P, Allis CD, Wang GG (2010) Covalent histone modifications—miswritten, misinterpreted and mis-erased in human cancers. NatRev Cancer 10:457–469.

12. Chowdhury R, Yeoh KK, Tian YM, Hillringhaus L, Bagg EA, RoseNR et al (2011) The oncometabolite 2-hydroxyglutarate inhibitshistone lysine demethylases. EMBO Rep 12:463–469.

13. Cin H, Meyer C, Herr R, Janzarik WG, Lambert S, Jones DT et al(2011) Oncogenic FAM131B-BRAF fusion resulting from 7q34deletion comprises an alternative mechanism of MAPK pathwayactivation in pilocytic astrocytoma. Acta Neuropathol 121:763–774.

14. Cloughesy TF, Mischel PS (2011) New strategies in the moleculartargeting of glioblastoma: how do you hit a moving target? ClinCancer Res 17:6–11.

15. Dang L, White DW, Gross S, Bennett BD, Bittinger MA, DriggersEM et al (2009) Cancer-associated IDH1 mutations produce2-hydroxyglutarate. Nature 462:739–744.

16. Dhayalan A, Tamas R, Bock I, Tattermusch A, Dimitrova E,Kudithipudi S et al (2011) The ATRX-ADD domain binds to H3 tailpeptides and reads the combined methylation state of K4 and K9.Hum Mol Genet 20:2195–2203.

17. Downing JR, Wilson RK, Zhang J, Mardis ER, Pui CH, Ding Let al (2012) The pediatric cancer genome project. Nat Genet44:619–622.

18. Elsaesser SJ, Goldberg AD, Allis CD (2010) New functions for anold variant: no substitute for histone H3.3. Curr Opin Genet Dev20:110–117.

19. Eyler CE, Foo WC, LaFiura KM, McLendon RE, Hjelmeland AB,Rich JN (2008) Brain cancer stem cells display preferentialsensitivity to Akt inhibition. Stem Cells 26:3027–3036.

20. Faury D, Nantel A, Dunn SE, Guiot MC, Haque T, Hauser P et al(2007) Molecular profiling identifies prognostic subgroups ofpediatric glioblastoma and shows increased YB-1 expression intumours. J Clin Oncol 25:1196–1208.

21. Goldberg AD, Banaszynski LA, Noh KM, Lewis PW, Elsaesser SJ,Stadler S et al (2010) Distinct factors control histone variant H3.3localization at specific genomic regions. Cell 140:678–691.

22. Hake SB, Allis CD (2006) Histone H3 variants and their potentialrole in indexing mammalian genomes: the “H3 barcode hypothesis”.Proc Natl Acad Sci U S A 103:6428–6435.

23. Haque T, Faury D, Albrecht S, Lopez-Aguilar E, Hauser P, GaramiM et al (2007) Gene expression profiling from formalin-fixedparaffin-embedded tumours of pediatric glioblastoma. Clin CancerRes 13:6284–6292.

24. Hawkins C, Walker E, Mohamed N, Zhang C, Jacob K, Shirinian Met al (2011) BRAF-KIAA1549 fusion predicts better clinicaloutcome in pediatric low-grade astrocytoma. Clin Cancer Res17:4790–4798.

Chromatin Remodeling Defects in Pediatric and Young Adult Glioblastoma Fontebasso et al

214 Brain Pathology 23 (2013) 210–216

© 2013 The Authors; Brain Pathology © 2013 International Society of Neuropathology

Page 6: Chromatin Remodeling Defects in Pediatric and Young Adult Glioblastoma: A Tale of a Variant Histone 3 Tail

25. Heaphy CM, de Wilde RF, Jiao Y, Klein AP, Edil BH, Shi C et al(2011) Altered telomeres in tumours with ATRX and DAXXmutations. Science 333:425.

26. Huh MS, Price O’Dea T, Ouazia D, McKay BC, Parise G, Parks RJet al (2012) Compromised genomic integrity impedes musclegrowth after Atrx inactivation. J Clin Invest 122:4412–4423.

27. Jacob K, Albrecht S, Sollier C, Faury D, Sader E, Montpetit Aet al (2009) Duplication of 7q34 is specific to juvenile pilocyticastrocytomas and a hallmark of cerebellar and optic pathwaytumours. Br J Cancer 101:722–733.

28. Jacob K, Quang-Khuong DA, Jones DT, Witt H, Lambert S,Albrecht S et al (2011) Genetic aberrations leading to MAPKpathway activation mediate oncogene-induced senescence insporadic pilocytic astrocytomas. Clin Cancer Res 17:4650–4660.

29. Jiao Y, Killela PJ, Reitman ZJ, Rasheed AB, Heaphy CM, de WildeRF et al (2012) Frequent ATRX, CIC, and FUBP1 mutations refinethe classification of malignant gliomas. Oncotarget 3:709–722.

30. Jones DT, Jager N, Kool M, Zichner T, Hutter B, Sultan M et al(2012) Dissecting the genomic complexity underlyingmedulloblastoma. Nature 488:100–105.

31. Jones DT, Kocialkowski S, Liu L, Pearson DM, Backlund LM,Ichimura K, Collins VP (2008) Tandem duplication producing anovel oncogenic BRAF fusion gene defines the majority of pilocyticastrocytomas. Cancer Res 68:8673–8677.

32. Jones DT, Kocialkowski S, Liu L, Pearson DM, Ichimura K, CollinsVP (2009) Oncogenic RAF1 rearrangement and a novel BRAFmutation as alternatives to KIAA1549:BRAF fusion in activating theMAPK pathway in pilocytic astrocytoma. Oncogene 28:2119–2123.

33. Kannan K, Inagaki A, Silber J, Gorovets D, Zhang J, KastenhuberER et al (2012) Whole-exome sequencing identifies ATRX mutationas a key molecular determinant in lower-grade glioma. Oncotarget3:1194–1203.

34. Khuong-Quang DA, Buczkowicz P, Rakopoulos P, Liu XY,Fontebasso AM, Bouffet E et al (2012) K27M mutation in histoneH3.3 defines clinically and biologically distinct subgroups ofpediatric diffuse intrinsic pontine gliomas. Acta Neuropathol124:439–447.

35. Kieran MW, Walker D, Frappaz D, Prados M (2010) Brain tumours:from childhood through adolescence into adulthood. J Clin Oncol28:4783–4789.

36. Kridel R, Sehn LH, Gascoyne RD (2012) Pathogenesis of follicularlymphoma. J Clin Invest 122:3424–3431.

37. Lacoste N, Almouzni G (2008) Epigenetic memory: H3.3 steps inthe groove. Nat Cell Biol 10:7–9.

38. Lai A, Kharbanda S, Pope WB, Tran A, Solis OE, Peale F et al(2011) Evidence for sequenced molecular evolution of IDH1 mutantglioblastoma from a distinct cell of origin. J Clin Oncol29:4482–4490.

39. Lewis PW, Elsaesser SJ, Noh KM, Stadler SC (2010) Daxx is anH3.3-specific histone chaperone and cooperates with ATRX inreplication-independent chromatin assembly at telomeres. Proc NatlAcad Sci U S A 107:14075–14080.

40. Liu XY, Gerges N, Korshunov A, Sabha N, Khuong-Quang DA,Fontebasso AM et al (2012) Frequent ATRX mutations and loss ofexpression in adult diffuse astrocytic tumours carrying IDH1/IDH2and TP53 mutations. Acta Neuropathol 124:615–625.

41. Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC,Jouvet A et al (2007) The 2007 WHO classification of tumours ofthe central nervous system. Acta Neuropathol 114:97–109.

42. Loyola A, Almouzni G (2007) Marking histone H3 variants: how,when and why? Trends Biochem Sci 32:425–433.

43. Lu C, Ward PS, Kapoor GS, Rohle D, Turcan S, Abdel-Wahab Oet al (2012) IDH mutation impairs histone demethylation and resultsin a block to cell differentiation. Nature 483:474–478.

44. Martinez-Garcia E, Licht JD (2010) Deregulation of H3K27methylation in cancer. Nat Genet 42:100–101.

45. Molenaar JJ, Koster J, Zwijnenburg DA, van Sluis P, Valentijn LJ,van der Ploeg I et al (2012) Sequencing of neuroblastoma identifieschromothripsis and defects in neuritogenesis genes. Nature483:589–593.

46. Morin RD, Johnson NA, Severson TM, Mungall AJ, An J, Goya Ret al (2010) Somatic mutations altering EZH2 (Tyr641) in follicularand diffuse large B-cell lymphomas of germinal-center origin. NatGenet 42:181–185.

47. Northcott PA, Shih DJ, Peacock J, Garzia L, Morrissy AS, ZichnerT et al (2012) Subgroup-specific structural variation across 1,000medulloblastoma genomes. Nature 488:49–56.

48. Noushmehr H, Weisenberger DJ, Diefes K, Phillips HS, Pujara K,Berman BP et al (2010) Identification of a CpG island methylatorphenotype that defines a distinct subgroup of glioma. Cancer Cell17:510–522.

49. Ohgaki H, Kleihues P (2011) Genetic profile of astrocyticand oligodendroglial gliomas. Brain Tumour Pathol 28:177–183.

50. Parsons DW, Jones S, Zhang X, Lin JC, Leary RJ, Angenendt Pet al (2008) An integrated genomic analysis of human glioblastomamultiforme. Science 321:1807–1812.

51. Paugh BS, Broniscer A, Qu C, Miller CP, Zhang J, TatevossianRG et al (2011) Genome-wide analyses identify recurrentamplifications of receptor tyrosine kinases and cell-cycleregulatory genes in diffuse intrinsic pontine glioma. J ClinOncol 29:3999–4006.

52. Paugh BS, Qu C, Jones C, Liu Z, Adamowicz-Brice M, Zhang Jet al (2010) Integrated molecular genetic profiling of pediatrichigh-grade gliomas reveals key differences with the adult disease.J Clin Oncol 28:3061–3068.

53. Pfister S, Janzarik WG, Remke M, Ernst A, Werft W, Becker Net al (2008) BRAF gene duplication constitutes a mechanism ofMAPK pathway activation in low-grade astrocytomas. J Clin Invest118:1739–1749.

54. Pollack IF, Finkelstein SD, Woods J, Burnham J, Holmes EJ,Hamilton RL et al (2002) Expression of p53 and prognosisin children with malignant gliomas. N Engl J Med 346:420–427.

55. Pollack IF, Hamilton RL, James CD, Finkelstein SD, Burnham J,Yates AJ et al (2006) Rarity of PTEN deletions and EGFRamplification in malignant gliomas of childhood: resultsfrom the Children’s Cancer Group 945 cohort. J Neurosurg105(Suppl):418–424.

56. Pollack IF, Hamilton RL, Sobol RW, Nikiforova MN,Lyons-Weiler MA, Laframboise WA et al (2010) IDH1 mutationsare common in malignant gliomas arising in adolescents: areport from the Children’s Oncology Group. Childs Nerv Syst27:87–94.

57. Qu HQ, Jacob K, Fatet S, Ge B, Barnett D, Delattre O et al (2010)Genome-wide profiling using single-nucleotide polymorphism arraysidentifies novel chromosomal imbalances in pediatric glioblastomas.Neuro Oncol 12:153–163.

58. Rood BR, Macdonald TJ (2005) Pediatric high-grade glioma:molecular genetic clues for innovative therapeutic approaches.J Neurooncol 75:267–272.

59. Schwartzentruber J, Korshunov A, Liu XY, Jones DT, Pfaff E,Jacob K et al (2012) Driver mutations in histone H3.3 andchromatin remodelling genes in paediatric glioblastoma. Nature482:226–231.

60. So AY, Jung JW, Lee S, Kim HS, Kang KS (2011) DNAmethyltransferase controls stem cell aging by regulating BMI1 andEZH2 through microRNAs. Plos One 6:e19503.

Fontebasso et al Chromatin Remodeling Defects in Pediatric and Young Adult Glioblastoma

215Brain Pathology 23 (2013) 210–216

© 2013 The Authors; Brain Pathology © 2013 International Society of Neuropathology

Page 7: Chromatin Remodeling Defects in Pediatric and Young Adult Glioblastoma: A Tale of a Variant Histone 3 Tail

61. States CCBTRotU (2009) CBTRUS Statistical Report: PrimaryBrain and Central Nervous System Tumours Diagnosed in EighteenStates in 2002–2006.

62. Sturm D, Witt H, Hovestadt V, Khuong-Quang DA, Jones DT,Konermann C et al (2012) Hotspot mutations in H3F3A and IDH1define distinct epigenetic and biological subgroups of glioblastoma.Cancer Cell 22:425–437.

63. Talbert PB, Henikoff S (2010) Histone variants—ancient wrapartists of the epigenome. Nat Rev Mol Cell Biol 11:264–275.

64. Turcan S, Rohle D, Goenka A, Walsh LA, Fang F, Yilmaz E et al(2012) IDH1 mutation is sufficient to establish the gliomahypermethylator phenotype. Nature 483:479–483.

65. Verhaak RG, Hoadley KA, Purdom E, Wang V, Qi Y, WilkersonMD et al (2010) Integrated genomic analysis identifies clinicallyrelevant subtypes of glioblastoma characterized by abnormalities inPDGFRA, IDH1, EGFR, and NF1. Cancer Cell 17:98–110.

66. Weller M, Felsberg J, Hartmann C, Berger H, Steinbach JP,Schramm J et al (2009) Molecular predictors of progression-free

and overall survival in patients with newly diagnosed glioblastoma:a prospective translational study of the German Glioma Network. JClin Oncol 27:5743–5750.

67. Wu G, Broniscer A, McEachron TA, Lu C, Paugh BS, Becksfort Jet al (2012) Somatic histone H3 alterations in pediatric diffuseintrinsic pontine gliomas and non-brainstem glioblastomas. NatGenet 44:251–253.

68. Yan H, Parsons DW, Jin G, McLendon R, Rasheed BA, Yuan Wet al (2009) IDH1 and IDH2 mutations in gliomas. N Engl J Med360:765–773.

69. Zarghooni M, Bartels U, Lee E, Buczkowicz P, Morrison A, HuangA et al (2010) Whole-genome profiling of pediatric diffuse intrinsicpontine gliomas highlights platelet-derived growth factor receptoralpha and poly (ADP-ribose) polymerase as potential therapeutictargets. J Clin Oncol 28:1337–1344.

70. Zhang J, Ding L, Holmfeldt L, Wu G, Heatley SL, Payne-Turner Det al (2012) The genetic basis of early T-cell precursor acutelymphoblastic leukaemia. Nature 481:157–163.

Chromatin Remodeling Defects in Pediatric and Young Adult Glioblastoma Fontebasso et al

216 Brain Pathology 23 (2013) 210–216

© 2013 The Authors; Brain Pathology © 2013 International Society of Neuropathology