cancer cell

147

Upload: manbasti

Post on 19-Oct-2015

156 views

Category:

Documents


0 download

DESCRIPTION

Cancer cell

TRANSCRIPT

  • tO li

    a

    ,

    e tu ae gs

    with mutations in H3F3A. This gene

    encodes the replication-independent his-

    These findings build on earlier studies

    et al., 2010). Tumors with IDH1mutations

    fall primarily into the proneural expression

    emphasizes the differences between

    ciated with a methylation pattern that is

    distinct from both that of H3F3A G34

    in IDH1 and H3F3A K27M mutant groups(Noushmehr et al., 2010; Parsons et al.,

    2008; Verhaak et al., 2010). Patients

    without IDH1 mutation are older and

    this tumor into six subgroups, two of

    which are primarily pediatric, and the

    remainder of which are primarily adult.

    to differentiation signals, which are hall-

    marks of pre-cancerous cells (Turcan

    et al., 2012). Sturm et al. (2012) show inhave more rapidly progressive disease,

    whereas those with IDH1 mutation

    frequently have an antecedent low grade

    Methylation groups largely correlate with

    the pattern of expression profiling that

    was previously reported, emphasizing

    H3F3A G34 mutated glioblastoma a

    similar decrease in expression of OLIG2,

    an important neuro-developmental gene.that showed that adult GBM could be

    subdivided into three epigenetic sub-

    groups, one of which correlated with

    mutations in the metabolic gene IDH1

    adult and pediatric GBM. Sturm et al.

    (2012) now show that an expanded anal-

    ysis of the methylome of GBM, which

    includes pediatric GBM cases, can divide

    (Lu et al., 2012). IDH1mutation also leads

    to increased neural stem cell marker

    expression and decreased expression of

    mature differentiation genes in responsetone H3.3, which predominantly binds

    transcriptionally active loci and telomeres.

    H3.3 is frequentlymethylatedat or near the

    residues that are mutated. Alterations in

    histonemethylationaffect theaccessibility

    of the associated DNA and may promote

    changes in DNA methylation (Turcan

    et al., 2012). H3F3A mutations therefore

    provide a potential mechanism underlying

    the global methylation changes observed

    in these pediatric GBM subgroups.

    profile, and in epigenetic analyses, CIMP+

    tumors segregate into that same group

    (Noushmehr et al., 2010).

    IDH1 is rarely altered in pediatric

    GBMs. Indeed, pediatric GBM contain

    significantly different genomic alterations

    from adult tumors (Paugh et al., 2010).

    The recent discovery that mutations in

    the histone gene H3F3A occur predomi-

    nantly in pediatric high grade gliomas

    (Schwartzentruber et al., 2012) further

    and the CIMP+ phenotype associated

    with IDH1 mutation.

    There is also increasing evidence that

    alterations in H3F3A and IDH1 interfere

    with the normal differentiation of neural

    progenitors. The abnormal metabolite

    2HG produced by mutant IDH1 leads

    to increased repressive methylation at

    H3K27 and inhibits immortalized neural

    cell differentiation, suggesting a potential

    common mechanism for tumorigenesisCancer Cell

    Previews

    Methylome AlteraNew Therapeutic

    Eric H. Raabe1,2,* and Charles G. Eberh1Division of Pediatric Oncology2Division of PathologyJohns Hopkins University School of Medicine*Correspondence: [email protected]://dx.doi.org/10.1016/j.ccr.2012.10.001

    In this issue of Cancer Cell, Sturmforme divide adult and pediatric tprofiles, and most importantly, difftherapeutics for this dreaded disea

    Glioblastoma multiforme (GBM) is the

    most aggressive brain tumor and is asso-

    ciated with very poor overall survival.

    GBM occurs in adults much more fre-

    quently than in children or adolescents,

    and pediatric GBM has genetic abnormal-

    ities that make it distinct from adult

    tumors, suggesting that although the

    microscopic appearance and grim prog-

    nosis are shared (Figure 1), pediatric and

    adult GBM have different underlying biol-

    ogies (Paugh et al., 2010). In this issue of

    Cancer Cell, Sturm et al. (2012) show that

    pediatric GBM contains two epigenetic

    subgroups that are distinct from those

    found in adult tumors. The epigenetic

    profiles of these groups correlate tightlyions Markpportunities in G

    rt2

    Baltimore, MD 21287, USA

    t al. report that global DNA methylamors into subgroups that have chrent clinical behaviors. These findine.

    glioma, are younger, have more frequent

    TP53 mutations, and are less likely to

    have receptor tyrosine kinase amplifica-

    tion (Parsons et al., 2008). The IDH1

    mutation is a gain of function alteration

    that creates a novel onco-metabolite, 2-

    hydroxyglutarate (2HG), which interferes

    with the normal cellular methylation

    machinery. This in turn leads to wide-

    spread increases in global methylation

    known as the CpG-island methylator

    phenotype (CIMP) (Turcan et al., 2012).

    mRNA expression profiling identified four

    subgroups of adult GBM (proneural,

    neural, classical, and mesenchymal) and

    determined that each subgroup contains

    distinct pathway alterations (VerhaakCancer Cell 22oblastoma

    ion patterns in glioblastoma multi-racteristic DNA mutations, mRNAs suggest novel opportunities for

    the importance of epigenetic regulation

    in GBM (Verhaak et al., 2010).

    Sturm et al. (2012) show that three of

    the epigenetic subgroups (two pediatric

    and one adult) correlate with mutations

    in H3F3A and IDH1. H3F3A and IDH1

    mutations are non-overlapping, sug-

    gesting that they represent different paths

    to achieve widespread alterations in

    genomic methylation. The two mutations

    in H3F3A are seven amino acid residues

    apart and are associated with dramati-

    cally different methylation profiles. The

    H3F3A G34 mutation is associated with

    a hypomethylation phenotype, which is

    most prominent at the ends of chromo-

    somes. The H3F3A K27 mutation is asso-, October 16, 2012 2012 Elsevier Inc. 417

  • Loss of OLIG2 is associated

    with increased methylation

    at the OLIG2 locus, which

    occurs despite the global hy-

    pomethylation of the genome

    in H3F3A G34 mutated glio-

    blastoma. Although there

    are intriguing similarities in

    the pathways between IDH1

    ., 2007). It

    tification of

    nslate into

    for these

    hanges in

    entified by

    epigenetic

    therapeutic

    logy Group

    ring chro-

    eacetylase

    560). How-

    have not

    pediatric

    that the

    GBM is

    chromosome ends and features the

    alternative lengthening of telomeres

    (ALT) phenotype. Although the precise

    mechanism of ALT remains unknown,

    the presence of promyelocytic leukemia

    bodies and evidence of heterologous

    recombination between telomeres pro-

    vide enticing targets for therapy (Heaphy

    et al., 2011). The recent increases in our

    understanding of the genetics of GBM,

    and in particular, the integration of

    both pediatric and adult tumors into an

    overall epigenomic classification scheme

    by Sturm et al. (2012), set the stage for

    translation of molecular advances into

    improved care for patients with this

    disease.

    REFERENCES

    Heaphy, C.M., Subhawong, A.P.,Hong, S.M., Goggins, M.G., Mont-gomery, E.A., Gabrielson, E., Netto,G.J., Epstein, J.I., Lotan, T.L.,Westra, W.H., et al. (2011). Am. J.Pathol. 179, 16081615.

    Louis, D.N., Ohgaki, H., Wiestler,O.D., and Cavenee, W.K. (2007).

    wicz-Brice, M., Zhang, J., Bax, D.A., Coyle, B.,Barrow, J., Hcol. 28, 3061

    SchwartzentrJones, D.T., Pbasso, A.M.,Nature 482, 2

    Sturm, D., WiD.-A., JonesTonjes, M., SiCell 22, this is

    Turcan,S.,RoF., Yilmaz, E.Ward, P.S., et

    Verhaak, R.GV., Qi, Y., WiGolub, T., MAtlas Researc98110.

    Si dOm inh oua tom 0n oat one aM

    Cancer Cell

    Previewsin the literature (Louis et al

    remains to be seen how iden

    methylation alterations will tra

    improved therapeutic options

    patients. The significant c

    global methylation patterns id

    Sturm et al. (2012) make

    altering agents an attractive

    modality. The Childrens Onco

    is currently investigating alte

    matin structure with histone d

    inhibitors in GBM (NCT01236

    ever demethylating agents

    been widely investigated in

    GBM.

    Sturm et al. (2012) show

    G34 subgroup of pediatricand H3F3Amutated glioblas-

    toma, the disparate methyla-

    tion signatures and clinical

    course of these three groups

    involving children and young

    adults suggest that the

    mechanism by which these

    alterations lead to transfor-

    mation of normal cells will

    require extensive study.

    Although this study shows

    that different GBM meth-

    ylation subgroups have

    somewhat variable clinical

    courses, the overall out-

    comes of children and adults

    with GBM is extremely poor,

    with few long-term survivors reported

    Figure 1.DivergentA pediatricleft) and a(right) haveeosin photoissue of Caarise in anand behavbar = 100 m418 Cancer Cell 22, October 16, 2012 2012hypomethylated preferentially at the

    milar Appearances, Different Methylomes, anutcomesidline GBM of the pons (a diffuse intrinsic pontemispheric glioblastoma multiforme arising in a ysimilar histologic appearance on high power hemaicrographs. Yet, as demonstrated by Sturm et al. (2cer Cell, these two tumors affect different patient pomically distinct regions, have different methylatidifferently. For both images, magnification = 2003.Elsevier Inc.argrave, D., et al. (2010). J. Clin. On-3068.

    uber, J., Korshunov, A., Liu, X.Y.,faff, E., Jacob, K., Sturm, D., Fonte-Quang, D.A., Tonjes, M., et al. (2012).26231.

    tt, H., Hovestadt, V., Khuong-Quang,, D.T.W., Konerman, C., Pfaff, E.,ll, M., Bender, S., et al. (2012). Cancersue, 425437.

    hle,D.,Goenka,A.,Walsh, L.A., Fang,, Campos, C., Fabius, A.W., Lu, C.,al. (2012). Nature 483, 479483.

    ., Hoadley, K.A., Purdom, E., Wang,lkerson, M.D., Miller, C.R., Ding, L.,esirov, J.P., et al.; Cancer Genomeh Network. (2010). Cancer Cell 17,WHO Classification of Tumours ofthe Central Nervous System (Lyon,France: IARC Press).

    Lu, C., Ward, P.S., Kapoor, G.S.,Rohle, D., Turcan, S., Abdel-Wahab,O., Edwards, C.R., Khanin, R., Fig-ueroa, M.E., Melnick, A., et al.(2012). Nature 483, 474478.

    Noushmehr, H., Weisenberger, D.J.,Diefes, K., Phillips, H.S., Pujara, K.,Berman, B.P., Pan, F., Pelloski,C.E., Sulman, E.P., Bhat, K.P.,et al.; Cancer Genome AtlasResearch Network. (2010). CancerCell 17, 510522.

    Parsons, D.W., Jones, S., Zhang, X.,Lin, J.C., Leary, R.J., Angenendt, P.,Mankoo, P., Carter, H., Siu, I.M.,Gallia, G.L., et al. (2008). Science321, 18071812.

    Paugh, B.S., Qu, C., Jones, C., Liu, Z., Adamo-

    e glioma,ng adultxylin and12) in thispulations,profiles,nd scale

  • Ttio E-

    s dt res e

    CDKs in DNA replication and chromo-

    specific and dependent on specific onco-

    Because the genetic modification was

    obtained using a small-molecule inhibitor

    Both lung and breast cancer cells display

    display high levels of cyclin D2/D3 andgenic alterations. These studies were per-

    formed using germline cyclin D1 knockout

    of CDK4/6 (PD 0332991) currently being

    studied in clinical trials. Importantly, this

    cyclin D3-CDK6 complexes in agree-

    ment with the modulation of cyclinsome segregation.

    The pioneering work by P. Sicinskis

    group in 2001 (Yu et al., 2001) showed

    that a specific interphase cyclin, cyclin

    D1, was required for RAS- and HER2-

    induced mammary tumors, but was

    dispensable for WNT- or MYC-induced

    mammary tumors. This work showed for

    the first time that the therapeutic value

    of a cell cycle regulator may be cell-type

    present since conception, the remaining

    question was whether the acute inhibition

    of cyclin D1-CDK complexes would be

    effective in already developed breast

    tumors. A new study in this issue of

    Cancer Cell by Choi et al. (2012) now

    shows that conditional genetic ablation

    of cyclin D1 in adult mice that bear tumors

    inhibits the growth of HER2-positive

    mammary carcinomas. A similar effect is

    characteristics of cell cycle arrest and

    senescence after genetic ablation or

    chemical inhibition of cyclin D1-CDK4

    complexes (Choi et al., 2012; Puyol

    et al., 2010). Similar treatments, however,

    result in apoptotic cell death in mouse

    and human leukemic cells (Choi et al.,

    2012; Sawai et al., 2012). This response

    seems to be associated with the

    finding that Notch1-induced tumorsCell Cycle-Based

    Marcos Malumbres1,*1Cell Division and Cancer Group, Spanish Na*Correspondence: [email protected]://dx.doi.org/10.1016/j.ccr.2012.09.024

    Targeted therapies directed againIn this issue of Cancer Cell, Choi ecyclin-dependent kinase complextumors and leukemias.

    The real problem is to understand

    why cancer cells grow when their

    normal counterparts would not.

    (Hunt, 2008)

    Tumor cells invariably display defects in

    the machinery that controls the cell divi-

    sion cycle. Yet, whether the cell cycle is

    a useful therapeutic target is still being

    intensely debated mostly due to the

    essential role of this process in tissue

    homeostasis and the difficulties of finding

    a therapeutic window (Malumbres and

    Barbacid, 2009). Progression through

    the cell cycle is driven by several protein

    kinases, including cyclin-dependent

    kinases (CDKs). Molecular analysis of

    tumor cells has provided strong evidence

    that the activity of these kinases is de-

    regulated in human tumors suggesting

    their potential therapeutic use. However,

    the clinical benefit of the first generation

    of CDK inhibitors has been limited due to

    toxicity and lack of specificity, possibly

    derived from the critical function of

    Cancer Cell

    Previewsmice, which raised some important

    questions at that time. Since cyclin D1

    knockout mice displayed minor defects

    in mammary gland development, it was

    argued that the effect observed could be

    due to special requirements for cyclin D1herapies Move Fo

    nal Cancer Research Centre (CNIO), Madrid

    t cell cycle regulators have beenal. and Sawai et al. rekindle the theby demonstrating their requirem

    in the cell of origin of these tumors. Two

    studies in 2006 suggested that this was

    not the case, as a similar therapeutic

    benefit was observed using CDK4-defi-

    cient mice or knockin mice expressing

    a mutant cyclin D1, which does not bind

    CDKs but maintains other CDK-indepen-

    dent functions (Landis et al., 2006; Yu

    et al., 2006). None of these models

    displayed defects in mammary gland

    development; yet, they were resistant to

    HER2-induced mammary gland carci-

    nomas. In addition, these studies sug-

    gested that CDK4, one of the kinase

    partners of cyclin D1, was the critical

    enzymatic activity to be targeted in

    HER2-positive mammary gland carci-

    nomas. Subsequent studies by Barbacid

    and colleagues (Puyol et al., 2010)

    showed that CDK4, but not CDK2 or

    CDK6, was critical for the development

    of K-RAS-induced lung tumors, whereas

    lack of this interphase kinase did not

    affect the development of lungs in germ-

    line knockout mice.effect is not accompanied by toxicities

    associated with the acute inhibition of cy-

    clin D1-CDK complexes in adult individ-

    uals (Puyol et al., 2010; Choi et al., 2012;

    Sawai et al., 2012), suggesting that side-

    effects of such treatments will be minimal.

    Cancer Cell 22rward

    28029, Spain

    ifficult to translate into the clinic.apeutic value of inhibiting specificnts in the maintenance of breast

    Based on the previous results, cyclin

    D1-CDK4 complexes seem to be critical

    targets in the proliferation of epithelial

    tumors, at least breast and lung carci-

    nomas (Figure 1). What about other

    interphase CDK complexes? Previous

    studies established a critical role for

    cyclin D3 and, at least partially, CDK6

    in lymphocytes and in the development

    of T cell leukemias (Hu et al., 2009;

    Sicinska et al., 2003). The relevance of

    these findings in cancer therapy has

    now been addressed using conditional

    knockouts or acute treatments with

    kinase inhibitors. Genetic ablation of

    cyclin D3 or inhibition of CDK4/6

    complexes in Notch1-induced T cell

    acute lymphoblastic leukemia (T-ALL)

    results in tumor regression without

    causing major abnormalities in other

    tissues (Choi et al., 2012; Sawai

    et al., 2012; in this issue of Cancer Cell).

    Importantly, the response of leukemic

    cells to these treatments is dramatically

    different from that of epithelial cells.D3 expression by the Notch1 pathway

    (Choi et al., 2012; Joshi et al., 2009).

    In fact, Notch1-negative T-ALLs or

    other types of leukemic cells did not

    undergo apoptosis following CDK4/6

    inhibition (Choi et al., 2012). Whether the

    , October 16, 2012 2012 Elsevier Inc. 419

  • susceptibility to apoptosis is

    provided by the activation of

    Notch1 or the specific

    inhibition of cyclin D3-(and

    possibly CDK6) complexes

    is not clear at present. Inter-

    estingly, cyclin D2 cannot

    compensate for the lack of

    cyclin D3 when expressed

    from the Ccnd3 locus sug-

    gesting intrinsic differences

    st tissues

    tration of

    f mamma-

    ts on the

    activities

    Barbacid,

    in breast,

    rong case

    r contexts

    ecific cy-

    ave thera-

    on these

    that many

    human tumors may be sensitive to

    specific cyclin-CDK complexes, as long

    as we identify the specific complexes

    that mediate the response to specific

    oncogenic pathways in each specific

    cell type. A long road in which difficult

    questions such as why cancer cells

    grow when their normal counterparts

    would not (Hunt, 2008) need to be

    addressed in a cell-type- and onco-

    gene-specific manner. Cancer patients

    will undoubtedly benefit from these

    studies.

    REFERENCES

    Choi, Y.J., Li, X., Hydbring, P.,Sanda, T., Stefano, J., Christie,A.L., Signoretti, S., Look, A.T.,Kung, A.L., von Boehmer, H., andSicinski, P. (2012). Cancer Cell 22,this issue, 438451.

    Hu, M.G., Deshpande, A., Enos, M.,Mao, D., Hinds, E.A., Hu, G.F.,Chang, R., Guo, Z., Dose, M., Mao,C., et al. (2009). Cancer Res. 69,810818.

    Strikoudis, A.M.A., Clark, Missue, 45246

    Sicinska, E., Aowski, C., YuGeng, Y., vo(2003). Cance

    Yu, Q., Geng411, 101710

    Yu, Q., Sicinsgozdzon, A., KHarris, L.N.,Cancer Cell 9

    A ncis Cp ied cifiK y

    fo orsill c

    complexes in similar pathologies induced by other oncogenes (e.g., mammarygland or lung tumors without activation of the RAS pathway) or in othertumor types.

    Cancer Cell

    Previewsneered mice with cell cycle

    mutations. The discovery

    that individual interphase

    cyclins or CDKs were dis-

    pensable for the develop-

    ment and homeostasis of mo

    was considered a demons

    the developmental plasticity o

    lian tissues and raised doub

    usefulness of inhibiting these

    in tumors (Malumbres and

    2009). The recent studies

    lung, and T-ALL make a st

    for the identification of cellula

    in which the inhibition of sp

    clin-CDK complexes may h

    peutic value (Figure 1). Based

    results, one could proposein the function of these two

    proteins (Sawai et al., 2012).

    The molecular basis for the

    differential response to

    CDK4/6 inhibitionsenes-

    cence versus apoptosis

    may be considered a new

    avenue of high interest in

    the therapeutic evaluation of

    these cell cycle regulators.

    It is been a long road20

    yearssince the initial gene-

    ration of genetically-engi-

    Figure 1.MalignanSince theCDK6 combe discardactivate CDpensableresearch w420 Cancer Cell 22, October 16, 2012 2012n Initial Road Map for the Treatment of Humaes with Specific CDK4/6 Inhibitorsmall-molecule inhibitor PD 0332991 inhibits bothlexes, the participation of CDK4 in human leukemat this time. Similarly, it is not clear which spe

    4 in lung tumors, although this information is likelr the selection of small-molecule kinase inhibitbe necessary to evaluate the relevance of specificElsevier Inc.Puyol, M., Martn, A., Dubus, P.,Mulero, F., Pizcueta, P., Khan, G.,Guerra, C., Santamara, D., andBarbacid, M. (2010). Cancer Cell18, 6373.

    Sawai, C., Freund, J., Oh, P.,Ndiaye-Lobry, D., Bretz, J.C.,, Genesca, L., Trimarchi, T., Kelliher,., et al. (2012). Cancer Cell 22, this5.

    ifantis, I., Le Cam, L., Swat, W., Bor-, Q., Ferrando, A.A., Levin, S.D.,n Boehmer, H., and Sicinski, P.r Cell 4, 451461.

    , Y., and Sicinski, P. (2001). Nature21.

    ka, E., Geng, Y., Ahnstrom, M., Za-ong, Y., Gardner, H., Kiyokawa, H.,Stal, O., and Sicinski, P. (2006)., 2332.Hunt, T. (2008). Cell Cycle 7, 37893790.

    Joshi, I., Minter, L.M., Telfer, J.,Demarest, R.M., Capobianco, A.J.,Aster, J.C., Sicinski, P., Fauq, A.,Golde, T.E., and Osborne, B.A.(2009). Blood 113, 16891698.

    Landis, M.W., Pawlyk, B.S., Li, T.,Sicinski, P., and Hinds, P.W.(2006). Cancer Cell 9, 1322.

    Malumbres, M., and Barbacid, M.(2009).Nat.Rev.Cancer9,153166.

    DK4 andas cannotc cyclinsto be dis-. Furtheryclin-CDK

  • yil e

    a wd me n

    protein 4 gene (Fabp4, also called aP2)

    The authors validated tumors as true

    et al. (2012) was shown to be active in

    of SmoM in myogenic lineages, Hatley

    given that most Hedgehog pathway inhib-

    develop into both muscle and brown fateRMS by histology and immunohisto-

    chemistry (Desmin, MyoD, and Myoge-

    et al. (2012) activated the SmoM2 allele in

    early muscle development, employing

    tissue via PRDM16 and PPARg signaling

    (Seale et al., 2008). Furthermore, satelliteto drive Cre. Eighty percent of aP2-Cre;

    SmoM2/+ mice developed eRMS in the

    head and ventral neck by 2 months of

    age. aP2-Cre Cdkn2aFlox/Flox mice did

    not develop eRMS, whereas deletion of

    Cdkn2a in aP2-Cre;SmoM2/+ mice de-

    creased latency and increased tumor

    penetrance, with all mice having tumors

    by 55 days. This finding implicates

    Cdkn2a locus loss as a secondary factor

    (disease modifier) of eRMS progression.

    adipose tissue but not in skeletal muscle,

    at least as evidenced from recombination

    in the sternocleidomastoid muscle (SCM)

    of aP2-Cre;R26-LacZ reporter mice and

    aP2-Cre;R26-YFP mice and whole-tissue

    examination. Interestingly, it was found

    that eRMS tumors in situ were completely

    surrounded by non-neoplastic adipose

    tissue adjacent to and clearly separated

    from the SCM at P14.

    To compare the tumorigenic potential

    itors currently in clinical trials are Smooth-

    ened antogonists, this model may have

    highest value as a genetic model of

    eRMS for the time being and as a preclin-

    ical therapeutic model only when GLI

    inhibitors emerge as clinical candidate

    agents.

    Taking a look onto mesenchymal

    progenitor cell biology, the plasticity of

    these stem cells may not be so surprising.

    It is known that Myf5 expressing cell canThe Not-so-Skinn

    Ken Kikuchi1 and Charles Keller1,*1Pediatric Cancer Biology Program, Pape FamPortland, OR 97239 USA*Correspondence: [email protected]://dx.doi.org/10.1016/j.ccr.2012.09.018

    The childhood cancer embryonal rhthis issue of Cancer Cell, Hatley anorigin for Sonic Hedgehog-drivenmouse model.

    One of the most intriguing clinical and

    scientific questions in pediatric oncology

    is how rhabdomyosarcoma, a tumor with

    a myogenic phenotype, can arise in tissue

    without hypaxial-like skeletal muscle

    elements. This conundrum is especially

    evident for the embryonal subtype of

    rhabdomyosarcoma (eRMS), which can

    arise from the salivary glands, skull base

    (parameninges), biliary tree, and genito-

    urinary tract (bladder/prostate) (Gurney

    et al., 1999; Shapiro and Bhattacharyya,

    2006). The work by Hatley et al. (2012) in

    this issue of Cancer Cell begins to

    address this conundrum by highlighting

    the plasticity of cells traditionally thought

    to be in the adipogenic lineage.

    In this paper, Hatley et al. (2012) condi-

    tionally activate a mutant Smoothened

    (SmoM2) allele to drive Hedgehog sig-

    naling in the adipogenic lineage using

    a 5.4 kb promoter/enhancer fragment

    from the adipocyte fatty acid binding

    Cancer Cell

    Previewsnin). Similarly, an embryonal muscle

    gene signature (MyoD1, Myogenin, Pax7,

    Myf5, Myh3, and Myh8) was evident by

    RT-PCR assay. To further affirm the diag-

    nosis, Hatley et al. (2012) performed gene

    expression profiling of aP2-Cre;SmoM2/+on Muscle Cance

    y Pediatric Research Institute, Department of P

    bdomyosarcoma can arise in tissuecolleagues report that non-skeletalmbryonal rhabdomyosarcoma in a

    tumors in comparison to human eRMS

    and tumors of previously reported mouse

    eRMS models. Overall, 67% of the probe

    pairs and 58% of the ortholog gene

    pairs showed agreement in gene expres-

    sion between their mouse tumors and

    previously published eRMS models (Ru-

    bin et al., 2011) or human tumors, respec-

    tively. Hatley et al. (2012) concluded that,

    despite being of an adipogenic lineage of

    origin, aP2-Cre;SmoM2/+ mouse tumors

    are an accurate preclinical eRMS model

    based on these histological and transcrip-

    tome parameters.

    The most interesting aspect of this

    paper is that these tumors originated

    from aP2 expressing cells. Previously,

    activity of the aP2 promoter in transgenic

    mice has been documented as specific

    for the adipocyte lineage and to be

    distinct from any skeletal muscle lineage

    (Urs et al., 2006). Moreover, the specific

    aP2-Cre transgenic line used by HatleyPax3-, Myf5-, or MyoD1-Cre. All of these

    crosses resulted in embryonic lethality

    without tumor formation. However, acti-

    vation of SmoM2 with Myogenin-Cre,

    which is specific for myoblast-stage

    muscle differentiation, resulted in tongue

    Cancer Cell 22r

    diatrics, Oregon Health & Science University,

    ithout skeletal muscle elements. Inuscle progenitors can be a cell ofadipocyte-restricted conditional

    tumors in 100% of the mice. Mice with

    activation of SmoM2 in terminally differen-

    tiated skeletal muscle (MCK-Cre) were

    viable with no evidence of tumorigenesis.

    This result is the best by far in asking

    whether differentiated versus differenti-

    ating myofibers can transform into rhab-

    domyosarcoma. To address the tumori-

    genic potential of SmoM2 in postnatal

    muscle stem cells (satellite cells), a Pax7-

    CreERT2 was employed, and mice were

    aged until 150 dayswithout develop-

    ment of tumors. It would have been

    intriguing to see if prior reports of non-

    myogenic sarcomas arising from a satel-

    lite cell lineage (Rubin et al., 2011) would

    have been observed if the mice had

    been aged longer. Another caveat of

    these findings is that the promoter of

    SmoM2 in this system was Rosa26, not

    the native Smo promoter, and it is there-

    fore difficult to say that this is amolecularly

    physiological eRMS model. Furthermore,cells can undergo adipogenesis under

    certain experimental conditions (Asakura

    et al., 2001; Shefer et al., 2004) or may

    apparently undergo transdifferentiation

    into fibroblasts (Brack et al., 2007).

    Hedgehog activation can also reduce fat

    , October 16, 2012 2012 Elsevier Inc. 421

  • formation via PPARg (Suh

    et al., 2006). Such reports

    lly in rhab-

    p53 status

    was unex-

    of interest

    s of these

    969 histor-

    Fraumeni

    syndrome

    to germline

    S tumors

    and neck,

    lso in the

    and retro-

    is cranial-

    of specific

    and neck

    eRMS patients it models, and closer

    examination of the preneoplastic and

    early neoplastic lesions will invariably

    help us understand the microenvironment

    in which these tumor initiating cells

    transform.

    Overall, the heterogeneity in human

    rhabdomyosarcoma phenotypes may

    result from the balance between genetic

    factors (mutational profile of the tumor

    including the initial mutation[s] and modi-

    fiers) and epigenetic factors (the cell of

    origin). In an era of precision medicine,

    this transgenic model representing the

    subset of human head and neck ERMS

    not derived from myogenic precursors

    may be particularly valuable in defining

    therapeutic targets for select

    patients. Applying knowledge

    Graff, J., GaCancer Cell 2

    Rubin, B.P., ND.P., Pal, R.,B.R., Chen,177191.

    Seale, P., BjoS., Kuang, S.,H.M., Erdjum454, 961967

    Shapiro, N.LOtolaryngol. H

    Shefer, G.,Reuveni, Z. (2

    Suh, J.M., Gaand Graff, J.M

    Urs, S., Harr(2006). Transg

    Figure 1. Model of Skeletal Myogenesis and Possible CellularOrigins of eRMSPostnatal muscle maintenance and regeneration are regulated by Pax3, Pax7,and muscle regulatory factors (MyoD, Myf5, Myogenin, and Myf6). Studiesdescribed in the text suggest that the cells of origin for eRMS include differen-tiating myoblasts and adipocytes.

    Cancer Cell

    Previewsof Hedgehog signaling was

    evident by RT-PCR studies

    of aP2-Cre;SmoM2/+ tumors,

    yet, by contrast, less than

    30% of human eRMS exhibit

    a gene expression signature

    consistent with a Hedgehog

    pathway on overdrive state

    (Rubin et al., 2011), and only

    rarely is Hedgehog overdrive

    a solitary signaling abnor-

    mality. Instead, p53 loss of

    function was most frequent

    (Rubin et al., 2011)implying

    that p53 loss precedes

    Hedgehog overdrive tempora

    domyosarcomagenesis. The

    of aP2-Cre;SmoM2/+ tumors

    plored, but it would have been

    to have known the p53 statu

    tumors, particularly given the 1

    ical precedent of Li and

    describing a familial eRMS

    now known to be attributable

    p53 mutation.

    In the Hatley model, eRM

    occurred from only the head

    yet human eRMS occur a

    urogenital tract, extremities,

    peritoneum. Nevertheless, th

    oriented eRMS model may be

    interest for the subset of headlend one to speculate that

    the origin of aP2-Cre;

    SmoM2/+ mice tumors might

    be cells that have undergone

    a transdifferentiation event

    from the adipocyte to the

    myogenic lineage (Figure 1).

    Hatley et al. (2012) recapit-

    ulate another important find-

    ing by gene expression pro-

    filing that aP2-Cre;SmoM2/+

    tumors show an activated

    satellite cell phenotype re-

    flective of the eRMS tumor

    pathology rather than the

    lineage of origin for the

    tumor. Certainly, activation422 Cancer Cell 22, October 16, 2012 2012 Elsevier Inc.Brack, A.S., Conboy, M.J., Roy, S.,Lee, M., Kuo, C.J., Keller, C., andRando, T.A. (2007). Science 317,807810.

    Gurney, J.G., Young, J.L., Jr.,Roffers, S.D., Smith, M.A., andBunin, G.R. (1999). Soft TissueSarcomas. In Cancer Incidence andSurvival among Children andAdolescents: United States SEERProgram 19751995, NationalCancer Institute, SEER Program.NIH Pub. No. 994649. Ries LAG,SM, J.G. Gurney, M. Linet, T. Tamra,J.L. Young, and G.R. Bunin, eds.(Bethesda, MD: National CancerInstitute).

    Hatley, M.E., Tang, W., Garcia, M.R.,Finkelstein, D., Millay, D.P., Liu, N.,lindo, R.L., and Olson, E.N. (2012).2, this issue, 536546.

    ishijo, K., Chen, H.I., Yi, X., Schuetze,Prajapati, S.I., Abraham, J., Arenkiel,Q.R., et al. (2011). Cancer Cell 19,

    rk, B., Yang, W., Kajimura, S., Chin,Scime`, A., Devarakonda, S., Conroe,ent-Bromage, H., et al. (2008). Nature.

    ., and Bhattacharyya, N. (2006).ead Neck Surg. 134, 631634.

    Wleklinski-Lee, M., and Yablonka-004). J. Cell Sci. 117, 53935404.

    o, X., McKay, J., McKay, R., Salo, Z.,. (2006). Cell Metab. 3, 2534.

    ington, A., Liaw, L., and Small, D.enic Res. 15, 647653.from these specialized pre-

    clinical models to clinical use

    will no doubt require novel

    statistical designs for clinical

    trials, given that the overall

    annual incidence of pediatric

    rhabdomyosarcoma is only in

    the hundreds, not thousands.

    Personalized rhabdomyosar-

    coma science inches ever

    closer.

    REFERENCES

    Asakura, A., Komaki, M., and Rud-nicki, M. (2001). Differentiation 68,245253.

  • o h

    , n

    hw

    rk r. i tnd c

    genes and tumor suppressors have been

    inefficient because most of the energy

    informatics study on diffuse large B cell

    approach revealed three major DLBCL

    DLBCLs. PPARg antagonists suppressed

    same type. This point should be empha-that could be generated from glucose

    is lost when the cell secretes lactate.

    subtypes: the OxPhos subset; the B cell

    receptor (BCR) subset, characterized by

    sized, because it tempers the paradigm

    that tumors are monolithic in their prefer-demonstrated to stimulate glycolysis

    (Ward and Thompson, 2012).

    Cells have two ways to produce adeno-

    sine triphosphate (ATP) for energy: gly-

    colysis and oxidative phosphorylation

    (OxPhos) (Figure 1). In glycolysis, glucose

    is converted to pyruvate, generating

    NADH from NAD+ and ATP from ADP. If

    the pyruvate is reduced to lactate, NAD+

    is regenerated and glycolysis continues.

    Although glycolysis is rapid, it is deemed

    lymphoma (DLBCL) revealed that some

    30% of these tumors belonged to a

    subset defined by high expression of

    genes involved in OxPhos (Monti et al.,

    2005). DLBCL is the most common non-

    Hodgkins lymphoma in Western popu-

    lations and is characterized by rapid

    growth. Several attempts have been

    made to classify DLBCL by molecular

    typing, particularly through shared

    patterns of gene expression. One such

    fatty acid oxidation and killed OxPhos

    cells in culture. Inhibitors of fatty acid

    oxidation had the same effect, as did

    silencing of an enzyme required to

    synthesize the antioxidant glutathione.

    None of these treatments affected non-

    OxPhos cells.

    The work illustrates several key prin-

    ciples in cancer metabolism. First,

    it demonstrates significant metabolic

    heterogeneity among tumors of theA Mitochondrial P

    Ralph J. DeBerardinis1,*1Childrens Medical Center Research InstituteDallas, TX 75390-8502, USA*Correspondence: ralph.deberardinis@utsouthttp://dx.doi.org/10.1016/j.ccr.2012.09.023

    Deregulated energetics is a hallmais unknown. A study by Caro et alis defined by reliance on mitochoimpaired.

    Altered metabolism was among the first

    cancer biomarkers (Koppenol et al.,

    2011). By 1925, Otto Warburg had ob-

    served that tumors rapidly took up

    glucose and converted it to lactic acid,

    which was released into the milieu. This

    was a departure from the other organs

    he studied, which imported less glucose,

    oxidized a higher fraction of it to carbon

    dioxide, and secreted much less lactate.

    This remarkable phenotype, called the

    Warburg effect, provides the rationale

    for two modern imaging technologies in

    cancer diagnosis: 18fluoro-2-deoxyglu-

    cose positron emission tomography

    (FDG-PET) to identify tumors by rapid

    glucose import and 1H magnetic reso-

    nance spectroscopy to identify areas of

    elevated lactate. Over the last two

    decades, the Warburg effect has been

    mechanistically tied to the molecular

    basis of transformation, as tumor-pro-

    moting mutations in many different onco-

    Cancer Cell

    PreviewsIn contrast, OxPhos is highly efficient.

    When substrates like pyruvate are

    oxidized in the mitochondria, reducing

    equivalents are delivered to the elec-

    tron transport chain, creating a proton

    gradient coupled to ATP synthesiswer Play in Lymp

    University of Texas Southwestern Medical Ce

    estern.edu

    of malignancy, but metabolic heten this issue of Cancer Cell demonsrial energy generation and is sele

    (Figure 1). This system yields nearly 20

    times as many ATP molecules per

    glucose as the Warburg effect. Other

    oxidizable substrates, like fatty acids,

    produce an even higher energy yield.

    Warburg considered tumor glycolysis

    to be a metabolic anomaly, famously

    postulating that it was the consequence

    of irreversible defects in respiration (i.e.,

    OxPhos) which were the root cause of

    cancer (Warburg, 1956). This model has

    been disproven as a general principle,

    because most cancer cells do not have

    static defects in their respiratory

    machinery, and more pointedly, because

    nonmalignant cells also display the

    Warburg effect if stimulated to grow

    (Wang et al., 1978). Nevertheless, the

    concept that glycolysis is a universal

    feature of aggressive tumor growth, and

    that OxPhos is counter-productive to

    tumorigenesis still pervades the literature.

    It was therefore surprising when a bio-expression of genes involved in B cell

    receptor signaling; and the host response

    (HR) subset, expressing markers of infil-

    trating inflammatory cells (Monti et al.,

    2005). Now, a study in this issue of

    Cancer Cell (Caro et al., 2012) examines

    Cancer Cell 22oma

    ter, 5323 Harry Hines Boulevard,

    ogeneity among individual tumorsrates that a subset of lymphomastively killed when this pathway is

    the metabolic phenotypes of these sub-

    types and found that OxPhos cell lines

    and primary biopsies contained elevated

    expression of many mitochondrial pro-

    teins. OxPhos cells displayed enhanced

    glucose oxidation relative to lactate

    formation, better defenses against oxida-

    tive stress, and a robust ability to oxidize

    fatty acids in the mitochondria. Providing

    exogenous fatty acids stimulated survival

    and growth in OxPhos cells, but not in

    cells from other subtypes. Overall, cells

    from OxPhos tumors generated a higher

    fraction of their ATP in the mitochondria,

    and this activity was required for cellular

    fitness.

    Importantly, inhibition of mitochondrial

    metabolism selectively killed OxPhos

    cells. The nuclear receptor peroxisome

    proliferator-activated receptor gamma

    (PPARg), which regulates the expression

    of many genes encoding mitochondrial

    proteins, was overexpressed in OxPhosence for glycolytic energy production.

    Second, metabolic heterogeneity is pro-

    grammed, and it persists when cells are

    adapted to culture. This argues strongly

    against the concept that the glycolytic

    phenotype of most cancer cell lines is

    , October 16, 2012 2012 Elsevier Inc. 423

  • (Fig-

    ority

    cted

    des

    of

    hen

    ther

    glio-

    sion

    ito-

    cur-

    factors that stimulate cell growth and

    may contribute to tumorigenesis (Wein-

    berg et al., 2010).

    It remains to be seen whether the

    OxPhos phenotype is driven by tumori-

    genic mutations or other features of

    DLBCL biology, and whether it bestows

    some context-specific advantage to this

    subset. Regardless of the mechanism, it

    is encouraging that molecular phenotyp-

    ing led to the discovery of metabolic

    R.M., Mashimo, T., Raisanen, J., Hatanpaa,K.J., Jindal, A., Jeffrey, F.M., Choi, C.,Madden, C., et al. (2012). NMR Biomed.

    WangScienc

    Warbu

    Ward,Cell 2

    WeinbWeinbman,del, N8788

    Zu, X.

    ta

    CitrateOxaloacetate

    ATP

    ATPATPATP

    ATP

    Reducing equivalents

    ATP

    ATPATP

    ElectronTransport Chain

    TCACycle

    ADP, Pi

    Glycolysis OxPhosfraction of ATP generation

    Non-OxPhoscells

    OxPhoscells

    mitochondrion

    Figure 1. Variable Reliance on OxidativePhosphorylation in DLBCLCells produce ATP from glycolysis (blue pathway) and oxida-tive phosphorylation (OxPhos, green). Cancer cell lines varyaccording to what fraction of their total ATP comes fromeach pathway, as shown on the spectrum at the bottom. Asubset of DLBCL tumors was defined by high expression ofgenes related to OxPhos. These cells produced most of theirATP from OxPhos and required this pathway for survival andgrowth. Non-OxPhos cells, which did not share this geneexpression signature, produced a higher fraction of their ATPfrom glycolysis, and were resistant to OxPhos inhibition.Abbreviation: TCA, tricarboxylic.

    Cancer Cell

    Previewsclinical description of the OxPhos

    DLBCL is lacking, patients with

    these tumors do not appear to

    survive any longer than those with

    other types (Monti et al., 2005). The

    aggressive course of DLBCL as a

    whole suggests that a preference

    for OxPhos does not prevent rapid

    cell growth in the large subset

    of tumors using that metabolic plat-

    form. Thus, metabolic programming

    to use the mitochondria rather than

    glycolysis as themajor site of energy

    formation is an effective strategy for

    cancer cell growth.

    The findings also emphasize that

    cancer cells, like normal cells, use

    multiple pathways concurrently to

    produce energy (Figure 1). The

    vast majority of cancer cells use

    both glycolysis and OxPhos

    together, although the balance

    between the two varies widely (Zu

    and Guppy, 2004). DLBCL cells

    are no exception. Despite their

    ability to oxidize pyruvate and

    fatty acids in the mitochondria,

    OxPhos cells still consumed glu-

    cose and secreted a modest

    amount of lactate. Conversely, the

    non-OxPhos cells still produced

    half of their ATP in the mitochondria

    ure 1). Along these lines, a large maj

    of DLBCL tumors are readily dete

    by FDG-PET, and this likely inclu

    OxPhos tumors. Thus, avid uptake

    glucose analogs may occur even w

    the mitochondria are fully active. Ano

    recent study on metabolic flux in

    blastoma reached this same conclu

    (Maher et al., 2012). Besides ATP, m

    chondria produce biosynthetic prean unavoidable consequence of

    culture conditions. Even the

    suppression of OxPhos in other

    DLBCL subtypes was programmed,

    because these cells could be stimu-

    lated to activate fatty acid oxidation

    simply by silencing Syk, a kinase in

    BCR signaling. Third, high rates of

    OxPhos can support growth in

    cancer cell lines. This may also be

    true in vivo. Although a complete

    Lacsors, reactive oxygen species, and other

    424 Cancer Cell 22, October 16, 2012 2012Glucose

    Pyruvate

    NAD+

    NADH

    te

    Acetyl-CoA

    Fatty Acids

    ADP, Pi

    ATPvulnerabilities in DLBCL. In a sense, the Res. C

    Elsevier Inc.Published online March 15, 2012. http://dx.doi.org/10.1002/nbm.2794.

    Monti, S., Savage, K.J., Kutok, J.L.,Feuerhake, F., Kurtin, P., Mihm, M., Wu,B., Pasqualucci, L., Neuberg, D., Aguiar,R.C., et al. (2005). Blood 105, 18511861.

    , T., Sheppard, J.R., and Foker, J.E. (1978).e 201, 155157.

    rg, O. (1956). Science 123, 309314.

    P.S., and Thompson, C.B. (2012). Cancer1, 297308.

    erg, F., Hamanaka, R., Wheaton, W.W.,erg, S., Joseph, J., Lopez, M., Kalyanara-B., Mutlu, G.M., Budinger, G.R., and Chan-.S. (2010). Proc. Natl. Acad. Sci. USA 107,8793.

    L., and Guppy, M. (2004). Biochem. Biophys.currently in use for cancer. New

    approaches to image tumor meta-

    bolism noninvasively (Kurhanewicz

    et al., 2011) may soon make it

    possible to stratify patients to

    therapeutic regimens solely by

    probing metabolism in vivo.

    REFERENCES

    Caro, P., Kishan, A.U., Norberg, E., Stanley,I.A., Chapuy, B., Ficarro, S.B., Polak, K.,Tondera, D., Gounarides, J., Yin, H.,et al. (2012). Cancer Cell 22, this issue,547560.

    Koppenol, W.H., Bounds, P.L., and Dang,C.V. (2011). Nat. Rev. Cancer 11,325337.

    Kurhanewicz, J., Vigneron, D.B., Brindle, K.,Chekmenev, E.Y., Comment, A., Cunning-ham, C.H., Deberardinis, R.J., Green, G.G.,Leach, M.O., Rajan, S.S., et al. (2011).Neoplasia 13, 8197.

    Maher, E.A., Marin-Valencia, I., Bachoo,new work fortifies Warburgs histor-

    ical claim that metabolism is an

    actionable biomarker (even if

    he would have been surprised by

    these particular results). PPARg

    antagonism may provide a starting

    point for targeting OxPhos tumors,

    and it would also be interesting

    to determine whether OxPhos pre-

    dicts sensitivity to therapiesommun. 313, 459465.

  • Cancer CellDepartment of Neurosurgery, Medical and Health Science Center,

    35Program in Developmental and Stem Cell Biology, Division of Neurosurgery, Arthur and Sonia Labatt Brain Tumour Research Centre,

    Hospital for Sick Children, University of Toronto, Toronto, ON M4N 1X8, Canada36Department of Oncogenomics, AMC, University of Amsterdam, Amsterdam 1105 AZ, The Netherlands

    37Departments of Neurology and Neurosurgery, Henry Ford Hospital, Detroit, MI 48202, USA38Department of Neuro-Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA39Division of Molecular Histopathology, Department of Pathology, University of Cambridge, Cambridge, CB2 0QQ, United KingdomArticle

    Hotspot Mutations in H3F3Aand IDH1 Define Distinct Epigeneticand Biological Subgroups of GlioblastomaDominik Sturm,1,42 Hendrik Witt,1,7,42 Volker Hovestadt,2,42 Dong-Anh Khuong-Quang,11,42 David T.W. Jones,1

    Carolin Konermann,3 Elke Pfaff,1 Martje Tonjes,2 Martin Sill,4 Sebastian Bender,1 Marcel Kool,1 Marc Zapatka,2

    Natalia Becker,4 Manuela Zucknick,4 Thomas Hielscher,4 Xiao-Yang Liu,11 Adam M. Fontebasso,12 Marina Ryzhova,13

    Steffen Albrecht,14 Karine Jacob,11 Marietta Wolter,15 Martin Ebinger,16 Martin U. Schuhmann,17 Timothy van Meter,18

    Michael C. Fruhwald,19 Holger Hauch,20 Arnulf Pekrun,21 Bernhard Radlwimmer,2 Tim Niehues,22

    Gregor von Komorowski,23 Matthias Durken,23 Andreas E. Kulozik,7 Jenny Madden,24 Andrew Donson,24

    Nicholas K. Foreman,24 Rachid Drissi,25 Maryam Fouladi,25 Wolfram Scheurlen,26 Andreas von Deimling,5,9

    Camelia Monoranu,27 Wolfgang Roggendorf,27 Christel Herold-Mende,8 Andreas Unterberg,8 Christof M. Kramm,28

    Jorg Felsberg,15 Christian Hartmann,29 Benedikt Wiestler,10 Wolfgang Wick,10 Till Milde,6,7 Olaf Witt,6,7

    Anders M. Lindroth,3 Jeremy Schwartzentruber,30 Damien Faury,11 Adam Fleming,11 Magdalena Zakrzewska,31

    Pawel P. Liberski,31 Krzysztof Zakrzewski,32 Peter Hauser,33 Miklos Garami,33 Almos Klekner,34 Laszlo Bognar,34

    Sorana Morrissy,35 Florence Cavalli,35 Michael D. Taylor,35 Peter van Sluis,36 Jan Koster,36 Rogier Versteeg,36

    Richard Volckmann,36 Tom Mikkelsen,37 Kenneth Aldape,38 Guido Reifenberger,15 V. Peter Collins,39 Jacek Majewski,40

    Andrey Korshunov,5 Peter Lichter,2 Christoph Plass,3,41,* Nada Jabado,11,41,* and Stefan M. Pfister1,7,41,*1Division of Pediatric Neurooncology2Division of Molecular Genetics3Division of Epigenetics and Cancer Risk Factors4Division of Biostatistics5Clinical Cooperation Unit Neuropathology6Clinical Cooperation Unit Pediatric Oncology

    German Cancer Research Center (DKFZ) Heidelberg, 69120 Heidelberg, Germany7Department of Pediatric Oncology, Hematology, and Immunology8Department of Neurosurgery9Department of Neuropathology10Department of Neurooncology

    Heidelberg University Hospital, 69120 Heidelberg, Germany11Departments of Pediatrics and Human Genetics, McGill University12Division of Experimental Medicine, Montreal Childrens Hospital

    McGill University Health Center Research Institute, Montreal, QC H3Z 2Z3, Canada13NN Burdenko Neurosurgical Institute, Moscow, 125047, Russia14Department of Pathology, Montreal Childrens Hospital, McGill University Health Center Research Institute,Montreal, QCH3H 1P3, Canada15Department of Neuropathology, Heinrich-Heine-University, 40225 Dusseldorf, Germany16Department of Hematology and Oncology, Childrens University Hospital Tubingen, 72076 Tubingen, Germany17Department of Neurosurgery, University Hospital Tubingen, 76076 Tubingen, Germany18Virginia Commonwealth University, Richmond, VA 23298, USA19Pediatric Hospital, Klinikum Augsburg, 86156 Augsburg, Germany20Pediatric Hospital, Klinikum Heilbronn, 74078 Heilbronn, Germany21Prof.-Hess-Kinderklinik, Klinikum Bremen-Mitte, 28177 Bremen, Germany22Childrens Hospital, Helios Clinics, 47805 Krefeld, Germany23Childrens University Hospital, 68135 Mannheim, Germany24Department of Pediatrics, University of Colorado Denver, Aurora, CO 80045, USA25Division of Oncology, Cincinnati Childrens Hospital Medical Center, Cincinnati, OH 45229, USA26Cnopfsche Kinderklinik, Nurnberg Childrens Hospital, 90419 Nurnberg, Germany27Department of Neuropathology, Institute of Pathology, University Wurzburg, 97080 Wurzburg, Germany28University Childrens Hospital, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany29Institute of Pathology, Department of Neuropathology, Hannover Medical School, 30175 Hannover, Germany30McGill University and Genome Quebec Innovation Centre, Montreal, QC H3A 1A4, Canada31Department of Molecular Pathology and Neuropathology, Medical University of Lodz 92-216 Poland32Department of Neurosurgery, Polish Mothers Memorial Hospital Research Institute, Lodz 93-338 Poland332nd Department of Paediatrics, Semmelweis University, Budapest H-1094 Hungary34 University of Debrecen, H-4032 Debrecen, HungaryCancer Cell 22, 425437, October 16, 2012 2012 Elsevier Inc. 425

  • 40Department of Human Genetics, McGill University, Montreal, QC H3Z 2Z3, Canada41These authors contributed equally to this work42These authors contributed equally to this work

    *Correspondence: [email protected] (C.P.), [email protected] (N.J.), [email protected] (S.M.P.)

    http://dx.doi.org/10.1016/j.ccr.2012.08.024

    is

    Htuoioc

    .

    shown to be associated with a distinct Glioma-CpG-Island

    heterogeneity of glioblastoma across all age groups. An over-

    view of all GBM samples subjected to various analyses is given

    Cancer Cell

    Epigenetic and Biological Subgroups of GlioblastomaSignificance

    GBM is the most common and also the most devastating brain tumor, with a 5-year survival rate below 10%. We presentstrong evidence that GBM can be subclassified into multiple groups, indistinguishable by histological appearance, butcorrelating with molecular-genetic factors as well as key clinical variables such as patient age and tumor location. We iden-Mutations in a protein complex comprised of H3.3 and ATRX/ in Figure S1A available online.one-third of pediatric GBMs (Schwartzentruber et al., 2012)GBM, which arise from a preceding lower-grade lesion. How-

    ever, stepwise transformation from less-malignant gliomas into

    GBM rarely occurs in children (Broniscer et al., 2007). Further-

    more, IDH1 or IDH2 mutations, which are found in up to 98%

    of adult secondary GBM, are very rare in childhood GBM

    (

  • MGB

    Col

    or s

    cale

    (bet

    a-va

    lues

    )

    0.5

    1

    ylat

    ion

    prob

    es (n

    = 8

    ,000

    )

    Met

    hyla

    ted

    Cancer Cell

    Epigenetic and Biological Subgroups of GlioblastomaWe investigated a cohort of GBMs from children (n = 59) and

    adult patients (n = 77) for genome-wide DNA methylation

    patterns using the Illumina 450k methylation array, and comple-

    mented our data with unpublished profiles of 74 adult GBM

    samples generated by The Cancer Genome Atlas (TCGA)

    Consortium (McLendon et al., 2008) (Table S1). Consensus

    clustering using the 8,000 most variant probes across the data

    set robustly identified six distinct DNA methylation clusters

    (Figures 1 and S1B). Based on correlations with mutational

    status, DNA copy-number aberrations, and gene expression

    signatures, as outlined below, we have labeled these subgroups

    IDH, K27, G34, RTK I (PDGFRA), Mesenchymal, and

    RTK II (Classic).

    A striking finding of this integrated analysis is that H3F3A K27

    and G34 mutations were exclusively distributed to the K27

    (18/18) andG34 (18/18) clusters, respectively (p < 0.001; Fishers

    exact test) (Figure 1). The IDH group contained 88% of IDH1-

    mutated tumors (23/26) (p < 0.001) and displayed concerted,

    0

    21

    Age60

    90

    PDGFRA ampl.EGFR ampl.

    CDKN2A del.Chr. 10 lossChr. 7 gain

    TP53H3F3A

    IDH1TCGA ExpressionTCGA MethylationMethylation cluster

    0

    G34R/V

    MUT

    WTNA

    K27M

    Mut

    atio

    nsC

    ytog

    enet

    ics

    IDH K27 G34 RTK I 'PDGFRA

    DN

    A m

    eth

    G-CIMP+Cluster #2Cluster #3

    ProneuralNeuralClassicalMesenchymal

    TCGA subgroups

    Met

    hyla

    tion

    Exp

    ress

    ion

    Unm

    ethy

    late

    d

    Figure 1. Methylation Profiling Reveals the Existence of Six Epigenetic

    Heatmap of methylation levels in six GBM subgroups identified by unsupervised

    represents a probe; each column represents a sample. The level of DNA methylat

    (n = 210), patient age, subgroup association, mutational status, and cytogenetic

    See also Figure S1 and Tables S1 and S2.

    C samples (n = 210) Controlsglobal hypermethylation (Figures 1, 2A, and 2B), thereby ex-

    panding the previously described link between IDH1 mutation

    and G-CIMP+ tumors to a pediatric setting (Noushmehr et al.,

    2010). In contrast, tumors in the G34 cluster specifically showed

    widespread hypomethylation across the whole genome, and

    especially in nonpromoter regions, when compared with all other

    subgroups (Figures 2A and 2B). This suggests the existence of

    a more global version of a CpG hypomethylator phenotype

    (CHOP), as proposed for a small number of genes in gastric

    cancer (Kaneda et al., 2002). More detailed mapping of differen-

    tially methylated regions revealed that the hypomethylation

    observed in H3F3A G34-mutated tumors was particularly prom-

    inent at chromosome ends (Figures 2C and 2D), potentially link-

    ing subtelomeric demethylation to alternative lengthening of

    telomeres, which is most frequently observed in this subgroup

    (Schwartzentruber et al., 2012).

    Of note, all mutations inH3F3A and IDH1were mutually exclu-

    sive (p < 0.001) (Figure 1). To further test this observation, we

    ' Mesenchymal RTK II 'Classic'

    p < 0.001

    p < 0.001

    p < 0.001

    p < 0.001

    p < 0.001

    p < 0.001

    p < 0.001

    p < 0.001

    p < 0.001

    p < 0.001

    Feta

    l nor

    mal

    bra

    in (n

    =4)

    Adu

    lt no

    rmal

    bra

    in (n

    =2)

    WG

    A-D

    NA

    (n=2

    )M

    .Sss

    I-DN

    A (n

    =2)

    GBM Subgroups

    k-means consensus clustering, and control samples as indicated. Each row

    ion (beta-value) is represented with a color scale as depicted. For each sample

    aberrations are indicated.

    ancer Cell 22, 425437, October 16, 2012 2012 Elsevier Inc. 427

  • AB

    0

    5

    10

    0

    0.2

    0.4

    0.0e+00 5.0e+06 1.0e+07 1.5e+07 2.0e+07 2.5e+07 3.0e+07

    -0.04

    0.00

    0.04

    Distance to chromosome end [bp]

    Num

    ber o

    f pro

    bes

    (x 1

    ,000

    )N

    orm

    aliz

    ed D

    NA

    Met

    hyla

    tion Fr

    actio

    n C

    pG Is

    land

    G34IDH

    K27RTK IMESRTK II

    C D

    ***

    IDH

    K27

    G34

    RTK

    I

    ME

    S

    RTK

    II

    Con

    trol

    0.35

    0.40

    0.45

    0.50

    0.55

    0.60

    Mea

    n m

    ethy

    latio

    n (m

    ean

    beta

    val

    ue)

    ******

    **

    ****

    ***

    IDH

    K27

    G34

    RTK

    I

    ME

    S

    RTK

    II

    Con

    trol

    0.20

    0.25

    0.30

    0.35

    ******

    ******

    ******

    ******

    ****

    IDH

    K27

    G34

    RTK

    I

    ME

    S

    RTK

    II

    Con

    trol

    0.45

    0.50

    0.55

    0.60

    0.65

    0.70

    0.75

    *

    ****

    ***

    0.0 0.2 0.4 0.6 0.8 1.0

    0.0

    0.2

    0.4

    0.6

    0.8

    1.0

    Overall

    Pro

    babi

    lty F

    n(pr

    obes

    ) Hypomethylation

    Hypermethylation

    DNA methylation (beta-value)0.0 0.2 0.4 0.6 0.8 1.0

    Non-promoter

    0.0

    0.2

    0.4

    0.6

    0.8

    1.0

    DNA methylation (beta-value)

    K27IDH

    G34RTK IMESRTK IIControl

    0.0 0.2 0.4 0.6 0.8 1.0

    Promoter

    0.0

    0.2

    0.4

    0.6

    0.8

    1.0

    DNA methylation (beta-value)

    K27IDH

    G34RTK IMESRTK IIControl

    -0.04

    -0.03

    -0.02

    -0.01

    0.00

    0.01

    0.02

    Chromosome end (4Mb)

    IDH

    K27

    G34

    RTK

    I

    ME

    S

    RTK

    II

    *********

    *** ***Mea

    n m

    ethy

    latio

    n (m

    ean

    beta

    val

    ue)

    15

    Figure 2. Global DNA Methylation Patterns in GBM Subgroups

    (A) Distinct patterns of global DNA methylation in GBM subgroups as identified by consensus clustering. The empirical cumulative distribution function for DNA

    methylation levels (beta-values) is plotted individually for each subgroup.

    (B) Overall DNAmethylation levels (mean beta-values) of individual GBMmethylation subgroups. Significant differences (***p < 0.001; **p < 0.01; *p < 0.05) to IDH

    and G34 subgroups are indicated.

    (C) Upper panel: Probe density in respect of distance to chromosome end. The fraction of probes located within CpG-Islands (red line) remains stable. Lower

    panel: Mean methylation value per subgroup within windows of 500kb, normalized to control samples. Individual samples are normalized by the mean overall

    methylation value.

    (D) Mean methylation value within 4 Mb to the chromosome end normalized to the mean overall methylation value and to control samples. Significant differences

    (***p < 0.001) between subgroups compared to G34 tumors are indicated. MES, Mesenchymal.

    See also Figure S2.

    Cancer Cell

    Epigenetic and Biological Subgroups of Glioblastoma

    428 Cancer Cell 22, 425437, October 16, 2012 2012 Elsevier Inc.

  • extended the targeted sequencing analysis of H3F3A and IDH1

    to include 460 GBM samples from patients covering a broad

    age range (Figure S1C; Table S2). Even in this expanded series,

    no co-occurring mutations in H3F3A and IDH1 were detected

    (p < 0.001), and the age distribution confirmed reported associ-

    ations of certain mutations with GBM in children (H3F3A K27),

    adolescent patients (H3F3A G34), and young adult patients

    (IDH1) (Khuong-Quang et al., 2012; Schwartzentruber et al.,

    2012; Yan et al., 2009) (Figure S1C; Table S2). As we have

    shown, TP53 mutations largely overlap with H3F3A mutations

    in pediatric GBM (Schwartzentruber et al., 2012), similar to the

    association of TP53 and IDH1 mutations in adults (Yan et al.,

    2009). This observation also holds true in our larger cohort,

    with a high enrichment of TP53 mutations in the G34 (18/18),

    IDH (22/24), and K27 (13/18) clusters (p < 0.001) (Figure 1).

    Since pediatric GBMs have been shown to display a distinct

    spectrum of focal copy-number aberrations (CNAs) compared

    with their adult counterparts (Bax et al., 2010; Paugh et al.,

    2010; Qu et al., 2010), we integrated DNA methylation clusters

    with copy-number data derived from the methylation arrays

    (Figures 1 and S1D). Interestingly, PDGFRA amplification was

    significantly more common in the RTK I PDGFRA cluster

    than any other subgroup (11/33; p < 0.001), hence our pro-

    posed name for this group. The RTK II Classic cluster demon-

    strated a very high frequency of whole chromosome 7 gain

    (50/56; p < 0.001) and whole chromosome 10 loss (56/56;

    p < 0.001), as well as frequent homozygous deletion of CDKN2A

    (35/56; p < 0.001) and amplification of EGFR (39/56; p < 0.001)

    (Figures 1 and S1D)all hallmark CNAs of adult GBM

    (Louis et al., 2007), as reflected by the complete absence of pedi-

    atric patients in this cluster. Overall, tumors from the IDH, K27,

    and G34 clusters were mostly devoid of the detected CNAs

    associated with the other GBM subgroups (amplifications of

    PDGFRA and EGFR, deletion of CDKN2A, chromosome 7 gain,

    and chromosome 10 loss) (Figure 1; Table S1), in keeping

    with a previously reported finding in G-CIMP+ tumors (Noush-

    mehr et al., 2010).

    To additionally place the methylation subgroups proposed

    here into the context of previous GBM classification systems,

    we used the gene expression signature described by the

    TCGA to classify 122 of the above tumors with available tran-

    scriptome data into one of four gene expression subtypes:

    Proneural, Neural, Mesenchymal, and Classical (Verhaak et al.,

    2010) (Figure 1; Table S1). This further confirmed the prototypic

    nature of tumors in the RTK II Classic cluster, which was

    clearly enriched for Classical expression profiles (p < 0.001).

    The RTK I PDGFRA cluster was highly enriched for Proneu-

    ral expression (p = 0.01), further substantiating the previously

    reported association of PDGFRA amplification with this expres-

    sion subtype (Verhaak et al., 2010). As expected, all tumors in

    the IDH cluster displayed Proneural expression patterns. Inter-

    estingly, the K27 cluster also showed a clear enrichment of

    tumors with a Proneural signature (p < 0.01), indicating that

    Cancer Cell

    Epigenetic and Biological Subgroups of Glioblastomathis expression subtype can be divided into subgroups harboring

    distinct genomic aberrations based on methylation profiling and

    targeted gene sequencing. Mesenchymal gene expression

    was mostly restricted to one methylation subgroup (p < 0.001),

    that showed a much lower incidence of typical GBM-related

    CNAs, generally fewer copy-number changes, and no character-

    Cistic point mutations. We therefore termed this methylation

    cluster, which displayed the largest similarity with normal brain

    methylation patterns, Mesenchymal. Copy-number aberra-

    tions in these samples were, however, observed at a similar

    amplitude as in other cases, indicating an absence of excess

    stromal contamination.

    Our finding of six GBM methylation clusters is different from

    a TCGA study using Illumina 27k arrays, which identified three

    methylation clusters in an adult GBM cohort (Noushmehr et al.,

    2010). Applying their signature to our data set, however, showed

    that two clusters (G-CIMP+ and Cluster #3) mapped almost

    exactly to two of our subgroups (IDH and RTK II Classic,,

    respectively, p < 0.001) (Figure 1). By adding pediatric cases to

    the study cohort, we demonstrate that TCGAmethylation Cluster

    #2 can be further divided into four biologically distinct sub-

    groups, defined by a clear enrichment for mutations (K27,

    G34), CNAs (PDGFRA), and/or gene expression signatures

    (Mesenchymal). The same DNA methylation clusters were

    apparent when restricting our analyses to the pediatric popula-

    tion, with the exception of the RTK II Classic cluster, which

    is not represented in the pediatric population (Figure S1E).

    Notably, by analyzing tumors from patients spanning a broad

    age spectrum, we further observed a clear age-dependent

    increase in overall DNA methylation levels (Figure S2A), even

    after adjusting our analysis to exclude tumors with age-related

    mutations in IDH1 or H3F3A (Figure S2B).

    GBM Subgroups Show Correlations withClinicopathological VariablesThe DNA methylation clusters described here were closely

    associated with specific age groups, pointing toward the biolog-

    ical diversity of epigenetic GBM subgroups (Figure 1). While the

    K27 cluster predominantly consisted of childhood patients

    (median age 10.5 years, range 523 years), patients in the G34

    cluster were found mostly around the threshold between the

    adolescent and adult populations (median age 18 years, range

    942 years), as previously suggested (Schwartzentruber et al.,

    2012). The RTK I PDGFRA cluster also harbored a proportion

    of pediatric patients (median age 36 years, range 874 years), in

    line with reports of PDGFRA CNAs being more prevalent in

    childhood high-grade gliomas (Bax et al., 2010; Paugh et al.,

    2010; Qu et al., 2010). The Mesenchymal cluster displayed a

    widespread age distribution (median age 47, range 285 years).

    The IDH and RTK II Classic clusters were mostly comprised of

    younger adult (median age 40 years, range 1371 years) and

    older adult (median age 58, range 3681 years) patients, respec-

    tively, reflecting the established differences in patient age

    between IDH1-mutated/G-CIMP+ and IDH1 WT adult GBM

    (Noushmehr et al., 2010; Yan et al., 2009).

    The epigenetic GBM subgroups identified here also showed

    mutation-specific patterns of tumor location in the central

    nervous system (Figure 3A). While K27-mutated tumors were

    predominantly seen in midline locations, e.g., thalamus, pons,and the spinal cord (21/25 cases with available data), tumors

    from all other subgroups almost exclusively arose in the cerebral

    hemispheres (86/92, p < 0.001). To further investigate this asso-

    ciation of mutation type and location, we investigated the tran-

    scriptomic profiles of H3F3A-mutated samples (n = 13). Gene

    signatures characteristic for K27 and G34 mutant GBMs were

    ancer Cell 22, 425437, October 16, 2012 2012 Elsevier Inc. 429

  • AFrontal lobe

    Parietal lobeBasal gangliaapplied to a published series of 1,340 transcriptomic profiles

    representing multiple regions of the developing and adult human

    brain (Kang et al., 2011; Figure S3). The G34 mutant signature

    appeared to be most strongly expressed in early embryonic

    regions and early- to mid-fetal stages of neocortex and striatum

    development. In contrast, the K27 signature most closely

    matched with mid- to late-fetal stages of striatum and thalamus

    development. Thus, G34 and K27 mutant GBMs seem to show

    expression patterns of early developmental stages correlating

    with their subsequent tumor location, possibly indicating

    different cellular origins and/or time of tumor initiation for these

    two subgroups.

    B

    Pons

    Thalamus

    IDH (36)

    G34 (22)RTK I 'PDGFRA' (15)Mesenchymal (10)

    K27 (27)

    RTK II 'Classic' (9)

    n = 119

    2

    2

    7

    11

    Surv

    ival

    pro

    babi

    liy

    Overall survival (months)

    censored

    K27 (14)

    RTK I 'PDGFRA' (23)Mesenchymal (50)RTK II 'Classic' (43)

    IDH (10)

    G34 (12)

    n = 152

    0 12 24 36 48 60 72 84 96 108 1200

    0.2

    0.4

    0.6

    0.8

    1

    Figure 3. Epigenetic Subgroups of GBM Correlate with Distinct Clinica(A) Location of 119GBMs in the human central nervous systemgrouped bymethyl

    Circles without numbers represent single cases. Different colors indicate methyla

    sagittal view (left panel), tumors occurring in the cerebral and cerebellar hemisph

    (B and C) Kaplan-Meier survival curves for GBM subgroups defined by methylati

    rank tests between subgroups.

    See also Figure S3.

    430 Cancer Cell 22, 425437, October 16, 2012 2012 Elsevier Inc.Frontal lobe Parietal lobe

    2 32

    62143

    35 42

    Cancer Cell

    Epigenetic and Biological Subgroups of GlioblastomaCorrelating our proposed methylation clusters with patient

    survival indicated differences between mutation-defined sub-

    groups, but this was somewhat restricted by the low number

    of patients with available survival data in each subgroup (Fig-

    ure 3B). We therefore enlarged our survival analysis to include

    all tumors with known H3F3A and IDH1 mutation status (Fig-

    ure 3C). As expected, patients with IDH1 mutant tumors had a

    significantly longer overall survival (OS) than patients with

    H3F3A and IDH1 WT tumors (p < 0.001) (Noushmehr et al.,

    2010; Parsons et al., 2008; Yan et al., 2009). Notably, G34mutant

    GBM patients also showed a trend toward a better OS than WT

    tumor patients, with marginal statistical significance (p = 0.05). In

    Cerebellum

    Brainstem

    Temporal lobe

    Occipital lobe

    Occipital lobe

    Spinal cord

    3

    2

    2

    3

    7

    3

    2

    102

    2

    3

    3

    5

    = 1 Case

    = 5 Cases

    C

    Overall survival (months)

    censored

    H3F3A K27 (24)IDH1 MUT (47)

    H3F3A G34 (13)H3F3A/IDH1 WT (199)n = 283

    0 12 24 36 48 60 72 84 96 108 1200

    0.2

    0.4

    0.6

    0.8

    1

    p=0.05 p

  • A B

    Sig

    nific

    ance

    (-Lo

    g 10 p

    -val

    ue)

    2

    4

    6

    8

    710475

    OLIG1 OLIG2

    FOXG1

    1

    2

    3

    4

    5

    87

    Sig

    nific

    ance

    (-Lo

    g 10 p

    -val

    ue)

    FOXG1

    Cancer Cell

    Epigenetic and Biological Subgroups of Glioblastomacontrast, patients with K27 mutations tended toward an even

    shorter OS than patients with WT tumors, although this did not

    reach statistical significance (p = 0.12). Comparing the two

    H3F3A mutations, patients harboring G34-mutated tumors

    clearly had a longer OS than patients with tumors carrying the

    K27 mutation (p < 0.01). While this association may be partly

    linked to G34-mutated tumors being more accessible to surgery

    than the midline K27-mutated tumors, the better prognosis of

    G34 versus K27 was independent of location for those cases

    where both mutation type and tumor site information were avail-

    able (p = 0.02; HR = 0.20, 95% CI = 0.050.77; Cox proportional

    hazards model).

    Integrating Methylome and Transcriptome DataIdentifies Marker Genes of GBM SubgroupsA combined analysis of DNA methylation and gene expression

    data was used to identify subgroup-specific differentially regu-

    D E

    OLIG2OLIG1

    Chromosome 21

    IDH

    K27

    G34

    Mes

    ench

    ymal

    RTK

    IIR

    TK I

    1.0

    0.5

    0

    4

    6

    8

    10

    12

    IDH

    K27

    G34

    OLI

    G1

    expr

    essi

    on (L

    og2)

    OLI

    G2

    expr

    essi

    on (L

    og2)

    *** ***

    4

    6

    8

    10

    12 *** ***

    DNA methylation difference (beta-values)

    0

    0.6 0.4 0.2 0.0 0.2 0.4 0.6 -10 -5

    0

    Gene expression diffe

    Figure 4. Identification of Marker Genes Affected by Differential Methy

    (A andB) Volcanoplots illustratingdifferences inDNAmethylation (A) andgeneexpr

    values (A) and Log2 fold change in gene expression values (B) are plotted on the x ax

    (C) Starburst plot integrating DNA methylation (x axis) and gene expression (y ax

    (D) Methylation levels at the OLIG1 and OLIG2 loci across all 210 GBM samples in

    CpG-site. Light blue bars indicate promoter regions. Methylation levels are repre

    (E) Mean gene expression levels ofOLIG1 (upper panel) andOLIG2 (lower panel) a

    *p < 0.05) between subgroups compared to G34 tumors are indicated.

    (F) Inverse correlation of promoter methylation (x axis) and gene expression (y ax

    coefficient, r). MES, Mesenchymal.

    See also Figure S4 and Table S3.

    CC53

    254

    OLIG1

    OLIG2

    53

    -4

    2

    0

    2

    4

    1014

    39

    Gen

    e ex

    pres

    sion

    hang

    e >=

    0 L

    og10

    (pv

    alue

    , BH

    ), fo

    ld c

    hang

    e =0 Log10 (pvalue, BH), Difference

  • in2,

    m

    pr

    s

    (DBA

    DC

    48%

    IDH1 (2/2)

    WT (9/9)

    7%15%

    G34 (6/6)

    26%K27 (8/8)

    4%

    ATRX+ ATRX-

    6

    14 8

    17

    26

    3 2

    52

    OLI

    G2+

    /FO

    XG1-

    OLI

    G2-

    /FO

    XG1+

    OLI

    G2-

    /FO

    XG1-

    OLI

    G2+

    /FO

    XG1+

    0%

    20%

    40%

    60%

    80%

    100%

    ALT+ ALT-

    OLI

    G2+

    /FO

    XG1-

    OLI

    G2-

    /FO

    XG1+

    OLI

    G2-

    /FO

    XG1-

    OLI

    G2+

    /FO

    XG1+

    0%

    20%

    40%

    60%

    80%

    100%

    23

    4 3

    51

    9

    13 7

    18

    OLIG2- / FOXG1+

    OLIG2+ / FOXG1-

    OLIG2- / FOXG1-

    OLIG2+ / FOXG1+

    IDH1R132H

    n=143

    Figure 5. Identification of H3F3A-Mutated GBMs by Differential Prote(A) Classification of 143 pediatric GBMs according to protein expression of OLIG

    with known H3F3A and IDH1 mutation status as predicted by immunohistoche

    (B) Typical pattern of OLIG2/FOXG1+ cells with concomitant loss of ATRXcontrasting staining results for comparison. Scale bars represent 100 mm unles

    (C and D) Correlation of GBMs as classified in (A) with ATRX loss (C), and ALT

    See also Figure S5 and Table S4.hypomethylated (1653/1946, 85%, Figure S4B) in this subgroup,

    in contrast to the hypermethylator G-CIMP pattern observed in

    the IDH subgroup (Figure S4C). Hypermethylation and concur-

    rent downregulation of TP73 antisense RNA 1 (TP73-AS1) was

    identified as a unique characteristic of this IDH/G-CIMP+ cluster

    (Figures S4D and S4F). Interestingly, inactivation of this gene by

    promoter methylation has been reported as a common mecha-

    nism in a high proportion of oligodendrogliomas, 80% of which

    are also known to harbor IDH1 mutations (Pang et al., 2010).

    Immunohistochemical Analysis Correctly SubclassifiesMutation-Defined GBM SubgroupsIn an attempt to subgroup GBM samples based on differential

    protein expressionamethod which is likely to be more suitable

    for possible clinical applicationwe used commercially avail-

    able antibodies against OLIG2, FOXG1, and mutated IDH1

    (R132H) to stain a tissue-microarray (TMA) with cores from 143

    pediatric GBMs, and classified tumors according to their protein

    expression patterns (Figures 5A and 5B; Table S4). The resulting

    fractions of tumors with predicted mutations in IDH1 (IDH1R132H,

    n = 6) andH3F3A (OLIG2+/FOXG1 for K27, n = 37, and OLIG2/FOXG1+ for G34, n = 21) were consistent with the frequency of

    each mutation in the pediatric population as detected by tar-

    geted gene sequencing (Figure S1C). Our approach correctly

    classified GBMs with known H3F3A and IDH1 mutation status,

    and revealed a frequent association between OLIG2/FOXG1+

    tumors (assumed to be G34-mutated), loss of ATRX protein

    expression, and an ALT phenotype (Figures 5BD), as previously

    reported for H3F3A G34-mutated tumors (Schwartzentruber

    et al., 2012). The putative H3F3A mutant groups also did not

    overlap with tumors harboring IDH1 (R132H) mutations, and

    only one case with EGFR amplification and homozygous

    432 Cancer Cell 22, 425437, October 16, 2012 2012 Elsevier Inc.OLIG2 FOXG1

    ATRX ALT

    5 m 10 m

    Expression PatternsFOXG1, andmutated IDH1 (IDH1R132H). Numbers in brackets indicate samples

    istry and verified by targeted gene sequencing, respectively.

    otein expression and ALT as observed in G34-mutated GBMs. Insets show

    indicated differently.

    ).

    Cancer Cell

    Epigenetic and Biological Subgroups of GlioblastomaCDKN2A deletion was detected therein (Figure S5A). The corre-

    lation with clinicopathological variables, such as tumor location

    and patient survival, also reflected our findings from the array-

    based analysis (Figures S5B and S5C). Of note, rare tumors

    represented on the TMA occurring in the basal ganglia and the

    spinal cord were almost always found in the OLIG2+/FOXG1

    subgroup (and therefore predicted to harbor the H3F3A K27

    mutation), further strengthening our hypothesis of the H3F3A

    K27 mutation as a unifying characteristic of midline GBM.

    DISCUSSION

    We have identified six biological subgroups of GBM based on

    global DNA methylation patterns, which correlate with specific

    molecular-genetic alterations and key clinical parameters. Our

    findings suggest that at least 30%40% of pediatric/young adult

    GBMs are likely characterized by disrupted epigenetic regula-

    tory mechanisms, associated with recurrent and mutually exclu-

    sive mutations in either H3F3A or IDH1, and aberrant DNA

    methylation patterns. Placing these subgroups into the context

    of previous molecular GBM classification schemes described

    by the TCGA (Noushmehr et al., 2010; Verhaak et al., 2010)

    revealed a clear correlation with DNA methylation clusters and

    a corresponding enrichment for previously established expres-

    sion signatures in different epigenetic subgroups. We have

    also demonstrated that our proposed classification can refine

    that described by the TCGA for adult GBM, to give a stratification

    system that is applicable across all ages, and defines additional

    biologically meaningful subgroups. A simplified graphical sum-

    mary of the key molecular and biological characteristics of the

    GBM subgroups as identified by our integrated classification

    strategy is given in Figure 6.

  • AP

    loGMutations /Cytogenetics

    IDH1mut H3F3Amut K27

    TP53mut

    H3F3

    TTP53mut

    Epigenetic and BioIDH K27

    Cancer Cell

    Epigenetic and Biological Subgroups of GlioblastomaWe and others have recently described a high frequency of

    H3F3A K27 mutations in thalamic GBMs and in diffuse intrinsic

    pontine gliomas (DIPGs), suggesting that the latter likely repre-

    sent an anatomically-defined subset of K27 mutant GBM

    (Khuong-Quang et al., 2012; Schwartzentruber et al., 2012; Wu

    et al., 2012). We now extend this observation to a larger

    subgroup of GBM, characterized by the K27M mutation, which

    almost exclusively occurs in midline locations, including rare

    tumors in the basal ganglia and the spinal cord. This is in line

    with a recent study by Puget et al. (2012) in which gene expres-

    sion patterns of brainstem gliomas were found to resemble

    midline/thalamic tumors, indicating a closely related origin. The

    K27 subgroup also displays markedly lower expression of the

    ventral telencephalic marker FOXG1 than other subgroups.

    Conversely, non-K27 tumors were restricted to hemispheric

    AgeDistribution

    (years)

    TumorLocation

    PatientSurvival(months)

    DNAMethylation

    GeneExpression

    IHC ProteinMarker

    Proneural Proneural

    IDH1R132H

    0 12060 0 12060 0 6

    CIMP+

    -0.6 0.60 -0.6 0

    0 9030 60 0 9030 60 0 30

    Mix

    OLIG2+/FOXG1- OLIG2-/

    -0.6 0.60

    Figure 6. Graphical Summary of Key Molecular and Biological Charac

    Simplified schematic representation of key genetic and epigenetic findings in si

    clinical patient data.

    Cmut G34

    53mut

    PDGFRA ampl.

    CDKN2A del.CDKN2A del.

    7+

    10-CNVlow

    EGFR ampl.

    gical Subgroups of Glioblastoma34 RTK I RTK IIMESENCHYMALlocations, further underlining the biological divergence of epige-

    netic GBM subgroups. While recurrent focal amplification of

    PDGFRA has been suggested as a key oncogenic event in pedi-

    atric DIPGs in some studies (Paugh et al., 2011; Puget et al.,

    2012; Zarghooni et al., 2010), midline-associated tumors in the

    K27 or OLIG2+/FOXG1 subgroups (including ten brainstemgliomas with known PDGFRA copy-number status) lacked this

    common feature in our series. PDGFRA amplification was,

    however, enriched in a subgroup of supratentorial hemispheric

    GBMs. In part, this discrepancy may be explained by the use

    of autopsy (and therefore post radio/chemotherapy) material

    in previous study cohorts of DIPGs, which might have been

    confounded by the higher incidence of PDGFRA amplifications

    observed in radiation-induced gliomas (Paugh et al., 2010).

    Nevertheless, amplifications of PDGFRA have also been

    Proneural ClassicalMesenchymal

    0 120601200 0 12060 0 12060

    CHOP+

    0.6

    9060 0 9030 60 0 9030 60 0 9030 60

    ed

    FOXG1+ OLIG2+/FOXG1+ OLIG2+/FOXG1+ OLIG2+/FOXG1+

    -0.6 0.60 -0.6 0.60 -0.6 0.60

    teristics of GBM Subgroups

    x GBM subgroups as identified by methylation profiling and correlations with

    ancer Cell 22, 425437, October 16, 2012 2012 Elsevier Inc. 433

  • future work at a basic and translational/targeted therapeuticdetected in small numbers of pretreatment samples (Paugh

    et al., 2011; Puget et al., 2012; Zarghooni et al., 2010), and

    post-treatment samples were not found to show increased

    widespread genomic instability (Paugh et al., 2011). This partic-

    ularly clinically challenging subset of tumors clearly warrants

    further investigation, underlining the importance of routine

    stereotactic biopsy of DIPGs at the time of primary diagnosis.

    OLIG2 has previously been reported as a universal marker

    for diffuse gliomas (Ligon et al., 2004), and OLIG2-positive

    progenitor-like cells of the subventricular zone have been sug-

    gested as potential glioma-initiating cells (Wang et al., 2009).

    There is also evidence that OLIG2-mediated modification of

    p53 function is required for complete inactivation of the latter

    in malignant gliomas, which typically show indirect loss of

    p53 activity through MDM2 amplification or p14ARF deletion

    (Mehta et al., 2011). Here, we describe a distinct subgroup of

    GBM, harboring the H3F3A G34 mutation, in which OLIG1

    and OLIG2 protein expression is absent. Given the 100%mutation frequency of TP53 in this subgroup, this may indicate

    a different pathogenesis of G34-mutated GBM, in which

    direct inactivation of p53 is required rather than via an OLIG2-

    dependent mechanism.

    The previously reported association of H3F3A mutations,

    particular the G34 mutation, with loss of ATRX and ALT

    (Schwartzentruber et al., 2012) is further expanded upon here.

    Interestingly, the global CHOP that we observed in G34 mutants

    was particularly pronounced in subtelomeric regions, suggesting

    a possible mechanistic link with ALT in these tumors (Gonzalo

    et al., 2006). Whether this is a more general phenotype that

    can be observed in clinically and etiologically distinct subgroups

    of other human cancers, remains to be investigated.

    The close link between H3F3A mutation type, tumor location,

    and differential expression of key neuronal lineagemarkers leads

    us to speculate that there may be differences in the cell of origin

    and/or the time of tumor development between these GBM

    subgroups. Although supported by the differential expression

    of mutant-specific gene signatures at different stages of human

    brain development, this remains to be formally shown. Also

    requiring further validation in larger, prospective cohorts is the

    association of the G34 mutation with better overall survival

    compared with H3F3A and IDH1 WT tumors, and that of K27

    mutation with potentially poorer prognosis, as observed in our

    series and a recent cohort of pediatric DIPGs (Khuong-Quang

    et al., 2012).

    Given the location of the H3F3A mutations at or near critical

    regulatory histone residues, and their distinct methylation pro-

    files, we consider it likely that the H3.3 mutations are directly

    involved in producing widespread aberrant DNA methylation

    and deregulation of gene expression. This has recently been

    shown for IDH1 mutations, which alone are sufficient to induce

    the global epigenetic reprogramming of the G-CIMP phenotype

    in normal astrocytes (Turcan et al., 2012). Overproduction of

    the oncometabolite 2-hydroxyglutarate in IDH1-mutated cellsinhibits the TET family of 5-methlycytosine hydroxylases and

    H3K27-specific demethylases. This is thought to lead to de-

    creased 5-hydroxylmethycytosine and increased H3K27methyl-

    ation (Xu et al., 2011), resulting in aberrant DNA and histone

    methylation, and a block to differentiation (Christensen et al.,

    2011; Dang et al., 2009; Lu et al., 2012).

    434 Cancer Cell 22, 425437, October 16, 2012 2012 Elsevier Inc.level, particularly in a pediatric and young adult setting.

    EXPERIMENTAL PROCEDURES

    Patients and Tumor Samples

    Primary tumor samples for methylation (n = 136; Table S1), mutation (n = 460;

    Table S2), and gene expression (n = 69) analysis and all clinical data were

    collected at the DKFZ (Heidelberg, Germany) and at McGill University (Mon-

    treal, Canada). Paraffin-embedded samples (n = 143; Table S4) for TMA anal-

    ysis were collected from the Burdenko Neurosurgical Institute (Moscow,

    Russia) and from the Department of Neuropathology, University of Wurzburg

    (Germany). Patient clinical details can be found in Table S1 for the methylation

    analysis data set and in Table S4 for the TMA cohort. All of the tumors were

    banked at the time of primary diagnosis between 1994 and 2011 in accordance

    with research ethics board approval from the respective institutes. Informed

    consent was obtained from all patients included in this study. An overview of

    all samples included in different data collections is given in Figure S1A. All of

    the samples were independently reviewed by senior pediatric neuropatholo-

    gists (S.A. and A.K.) according to the WHO guidelines. Detailed information

    about samples provided by TCGA can be found elsewhere (http://

    cancergenome.nih.gov).

    DNA Methylation Profiling

    For genome-wide assessment of DNAmethylationGBM samples (n = 136) and

    controls (n = 10; four fetal and two