Elsevier

Seminars in Cancer Biology

Volume 51, August 2018, Pages 50-58
Seminars in Cancer Biology

Review
Glioma epigenetics: From subclassification to novel treatment options

https://doi.org/10.1016/j.semcancer.2017.11.010Get rights and content

Abstract

Gliomas are the most common malignant primary brain tumors, of which glioblastoma is the most malignant form (WHO grade IV), and notorious for treatment resistance. Over the last decade mutations in epigenetic regulator genes have been identified as key drivers of subtypes of gliomas with distinct clinical features. Most characteristic are mutations in IDH1 or IDH2 in lower grade gliomas, and histone 3 mutations in pediatric high grade gliomas that are also associated with characteristic DNA methylation patterns. Furthermore, in adult glioblastoma patients epigenetic silencing of the DNA repair gene MGMT by promoter methylation is predictive for benefit from alkylating agent therapy. These epigenetic alterations are used as biomarkers and play a central role for classification of gliomas (WHO 2016) and treatment decisions. Here we review the pivotal role of epigenetic alterations in the etiology and biology of gliomas. We summarize the complex interactions between “driver” mutations, DNA methylation, histone post-translational modifications, and overall chromatin organization, and how they inform current efforts of testing epigenetic compounds and combinations in preclinical and clinical studies.

Introduction

Gliomas are among the most common primary brain tumors in adults and account for over 70% of malignant brain tumors, of which glioblastoma is the most common and most malignant (World Health Organization [WHO] grade IV) with an incidence rate of 3.2 per 100 000 population [1]. The median survival is less than 2 years with the current standard of care of maximal safe resection, followed by combined radio-chemotherapy with the alkylating agent Temozolomide [2] that may be modestly improved with the addition of Tumor Treating Fields [3]. Glioblastomas are notorious for resistance to therapy, and despite numerous efforts, the addition of targeted agents against genetic or biological hallmarks of gliomas have largely failed [4]. Lower grade gliomas (LGG) WHO grade II and III are less common and affect younger patients. They have a better prognosis and show some sensitivity to therapy that both depend on the molecular subtype [5], [6]. After resection LGG patients may first just be followed according to a “wait and see” strategy that depends on clinical and molecular risk factors, before entering treatment with different schemes of radio- or chemotherapy, or a combination thereof [6], [7], [8]. The optimal therapy is debated, however treatment related effects on cognitive function require risk-adapted (molecularly driven) treatment strategies, given that LGG patients may live more than 15 years [9].

Section snippets

Epigenetic subtypes of gliomas

Insights into the molecular landscape of diffuse gliomas have revealed characteristic genetic and epigenetic profiles which have clarified their etiologic evolution [5], [10], [11], [12], [13], [14], [15], [16] and allowed their classification into distinct molecular subtypes that have been integrated into the 2016 WHO classification (Fig. 1) [17]. Mutations in the epigenetic modulator genes isocitrate dehydrogenase 1 or 2 (IDH1 or IDH2), and in the histone genes H3F3A or HIST1H3B have become

Targeting the glioma epigenome

Several different approaches of targeting epigenetic alterations have been or are being tested in clinical trials: those targeting mutant IDH either by small molecule inhibitors, or as target for vaccination in the respective patient population; and those targeting epigenetic modifiers affecting large parts of the epigenome such as BETi, HDACi, DNMTi, and EZH2i (Fig. 3). Respective clinical trials, as available on clinicaltrials.gov, are summarized in Table 1.

Conclusions

Discoveries of the last decade have completely changed our view on the genomic and epigenetic landscape of human gliomas. Driver mutations in epigenetic regulator genes have clarified their etiology and defined molecular subtypes with distinct biology. Consequently, epigenetic biomarkers play now a central role in tumor classification and decision making for stratified therapies. A whole arsenal of epigenetic drugs promises to target epigenetically deregulated pathways by interfering on several

Funding sources

Olga Gusyatiner is funded by the Swiss National Science Foundation (31003A_163297 to M. Hegi).

Conflict of interest statement

Olga Gusyatiner and Monika Hegi receive institutional research funding for institutional research project from Orion Corporation. M. Hegi received research funding for institutional research project from Novocure. She served as a consultant to Bristol-Myers Squibb. The Institution receives free testing from MDxHealth.

References (107)

  • Y.W. Zhang et al.

    Acetylation enhances TET2 function in protecting against abnormal DNA methylation during oxidative stress

    Mol. Cell

    (2017)
  • D. Sturm et al.

    New brain tumor entities emerge from molecular classification of CNS-PNETs

    Cell

    (2016)
  • S. Moran et al.

    Epigenetic profiling to classify cancer of unknown primary: a multicentre, retrospective analysis

    Lancet Oncol.

    (2016)
  • W. Xu et al.

    Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of [alpha]-ketoglutarate-dependent dioxygenases

    Cancer Cell

    (2011)
  • M.E. Figueroa et al.

    Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation

    Cancer Cell

    (2010)
  • P. Wang et al.

    Oncometabolite D-2-Hydroxyglutarate inhibits ALKBH DNA repair enzymes and sensitizes IDH mutant cells to alkylating agents

    Cell Rep.

    (2015)
  • K. Tateishi et al.

    Extreme vulnerability of IDH1 mutant cancers to nad+ depletion

    Cancer Cell

    (2015)
  • A. Malmstrom et al.

    Temozolomide versus standard 6-week radiotherapy versus hypofractionated radiotherapy in patients older than 60 years with glioblastoma: the Nordic randomised, phase 3 trial

    Lancet Oncol.

    (2012)
  • P. Bady et al.

    Sensitivity analysis of the MGMT-STP27 model and impact of genetic and epigenetic context to predict the MGMT methylation status in gliomas and other tumors

    J. Mol. Diagn.

    (2016)
  • I. Vlassenbroeck et al.

    Validation of real-time methylation-specific PCR to determine O6-methylguanine-DNA methyltransferase gene promoter methylation in glioma

    J. Mol. Diagn.

    (2008)
  • T. Kouzarides

    Chromatin modifications and their function

    Cell

    (2007)
  • S. Bender et al.

    Reduced H3K27me3 and DNA hypomethylation are major drivers of gene expression in K27 M mutant pediatric high-grade gliomas

    Cancer Cell

    (2013)
  • J.H. Gibcus et al.

    The hierarchy of the 3D genome

    Mol. Cell

    (2013)
  • H.M. Kantarjian et al.

    Guadecitabine (SGI-110) in treatment-naive patients with acute myeloid leukaemia: phase 2 results from a multicentre, randomised, phase 1/2 trial

    Lancet Oncol.

    (2017)
  • K.B. Chiappinelli et al.

    Inhibiting DNA methylation causes an interferon response in cancer via dsRNA including endogenous retroviruses

    Cell

    (2015)
  • S. Ropero et al.

    The role of histone deacetylases (HDACs) in human cancer

    Mol. Oncol.

    (2007)
  • J. Loven et al.

    Selective inhibition of tumor oncogenes by disruption of super-enhancers

    Cell

    (2013)
  • D. Hnisz et al.

    Super-enhancers in the control of cell identity and disease

    Cell

    (2013)
  • Q.T. Ostrom et al.

    CBTRUS Statistical report: primary brain and other central nervous system tumors diagnosed in the United States in 2009–2013

    Neuro Oncol.

    (2016)
  • R. Stupp et al.

    Maintenance therapy with tumor-treating fields plus temozolomide vs temozolomide alone for glioblastoma: a randomized clinical trial

    JAMA

    (2015)
  • H. Suzuki et al.

    Mutational landscape and clonal architecture in grade II and III gliomas

    Nat. Genet.

    (2015)
  • J.C. Buckner et al.

    Radiation plus procarbazine, CCNU, and vincristine in low-grade glioma

    N. Engl. J. Med.

    (2016)
  • M.J. van den Bent et al.

    Interim results from the CATNON trial (EORTC study 26053-22054) of treatment with concurrent and adjuvant temozolomide for 1p/19q non-co-deleted anaplastic glioma: a phase 3, randomised, open-label intergroup study

    Lancet

    (2017)
  • R. Ruda et al.

    Controversies in management of low-grade gliomas in light of new data from clinical trials

    Neuro Oncol.

    (2017)
  • H. Yan et al.

    IDH1 and IDH2 mutations in gliomas

    N. Engl. J. Med.

    (2009)
  • H. Noushmehr et al.

    Identification of a CpG island methylator phenotype that defines a distinct subgroup of glioma

    Cancer Cell

    (2010)
  • D.J. Brat et al.

    Comprehensive, integrative genomic analysis of diffuse lower-grade gliomas

    N. Engl. J. Med.

    (2015)
  • D.N. Louis et al.

    WHO Classification of tumours of the central nervous system

  • J. Schwartzentruber et al.

    Driver mutations in histone H3.3 and chromatin remodelling genes in paediatric glioblastoma

    Nature

    (2012)
  • G. Wu et al.

    Somatic histone H3 alterations in pediatric diffuse intrinsic pontine gliomas and non-brainstem glioblastomas

    Nat. Genet.

    (2012)
  • G. Reifenberger et al.

    Advances in the molecular genetics of gliomas ? implications for classification and therapy

    Nat. Rev. Clin. Oncol.

    (2016)
  • D.E. Reuss et al.

    Adult IDH wild type astrocytomas biologically and clinically resolve into other tumor entities

    Acta Neuropathol.

    (2015)
  • H. Ohgaki et al.

    The definition of primary and secondary glioblastoma

    Clin. Cancer Res.

    (2013)
  • D. Sturm et al.

    Pediatric gliomas: current concepts on diagnosis, biology, and clinical management

    J. Clin. Oncol.

    (2017)
  • A. Hermann et al.

    Biochemistry and biology of mammalian DNA methyltransferases

    Cell. Mol. Life Sci.

    (2004)
  • L. Scourzic et al.

    TET proteins and the control of cytosine demethylation in cancer

    Genome Med.

    (2015)
  • A. Portela et al.

    Epigenetic modifications and human disease

    Nat. Biotech.

    (2010)
  • A.M. Deaton et al.

    CpG islands and the regulation of transcription

    Genes Dev.

    (2011)
  • S. Fukushima et al.

    Genome-wide methylation profiles in primary intracranial germ cell tumors indicate a primordial germ cell origin for germinomas

    Acta Neuropathol.

    (2017)
  • J. Eckel-Passow et al.

    PATH-47. Clinical sensitivity and specificity of illumina methylation array for classifying adult gliomas into WHO groups

    Neuro-oncol

    (2017)
  • Cited by (366)

    View all citing articles on Scopus
    View full text