Skip to main content

Advertisement

Log in

Overview of DNA methylation in adult diffuse gliomas

  • Review Article
  • Published:
Brain Tumor Pathology Aims and scope Submit manuscript

Abstract

Adult diffuse gliomas form a heterogeneous group of tumors of the central nervous system that vary greatly in histology and prognosis. A significant advance during the last decade has been the identification of a set of genetic lesions that correlate well with histology and clinical outcome in diffuse gliomas. Most characteristic driver mutations consist of isocitrate dehydrogenase 1 (IDH1) and IDH2, and H3 histone family member 3A, which are strongly associated with DNA and histone methylation patterns. A well-characterized DNA methylation aberration is on the O6-methylguanine-DNA methyltransferase promoter. This aberration is associated with an improved response to the DNA alkylating agent, temozolomide. Methylation alterations are used for classification or treatment decisions of diffuse gliomas. This supports the importance of considering epigenomic aberrations in the pathogenesis of gliomas. Recent DNA methylation analyses revealed a small group of IDH mutant diffuse gliomas exhibiting decreased DNA hypermethylation resulting in substantial unfavorable prognosis comparable to glioblastoma. Thus, DNA methylation patterns may become a new standard that replaces the conventional grading system based on histological diagnosis. In this review, we summarize recent developments regarding the contributions of methylation patterns to the pathogenesis of adult diffuse glioma, the interactions between methylation patterns and driver mutations, and potential epigenomic targeted therapies.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2

Similar content being viewed by others

References

  1. Louis DN, Ohgaki H, Wiestler OD et al (2007) The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol 114:97–109

    Article  PubMed  PubMed Central  Google Scholar 

  2. Aldape K, Simmons ML, Davis RL et al (2000) Discrepancies in diagnoses of neuroepithelial neoplasms: the San Francisco Bay Area Adult Glioma Study. Cancer 88:2342–2349

    Article  CAS  PubMed  Google Scholar 

  3. van den Bent MJ (2010) Interobserver variation of the histopathological diagnosis in clinical trials on glioma: a clinician’s perspective. Acta Neuropathol 120:297–304

    Article  PubMed  PubMed Central  Google Scholar 

  4. Louis DN, Perry A, Reifenberger G et al (2016) The 2016 World Health Organization classification of tumors of the central nervous system: a summary. Acta Neuropathol 131:803–820

    Article  PubMed  Google Scholar 

  5. Yan H, Parsons DW, Jin G et al (2009) IDH1 and IDH2 mutations in gliomas. N Engl J Med 360:765–773

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Ichimura K (2012) Molecular pathogenesis of IDH mutations in gliomas. Brain Tumor Pathol 29:131–139

    Article  CAS  PubMed  Google Scholar 

  7. Cairncross JG, Ueki K, Zlatescu MC et al (1998) Specific genetic predictors of chemotherapeutic response and survival in patients with anaplastic oligodendrogliomas. J Natl Cancer Inst 90:1473–1479

    Article  CAS  PubMed  Google Scholar 

  8. Yamamichi A, Ohka F, Aoki K et al (2018) Immunohistochemical ATRX expression is not a surrogate for 1p19q codeletion. Brain Tumor Pathol 35:106–113

    Article  CAS  PubMed  Google Scholar 

  9. Parsons DW, Jones S, Zhang X et al (2008) An integrated genomic analysis of human glioblastoma multiforme. Science 321:1807–1812

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Schwartzentruber J, Korshunov A, Liu XY et al (2012) Driver mutations in histone H3.3 and chromatin remodelling genes in paediatric glioblastoma. Nature 482:226–231

    Article  CAS  PubMed  Google Scholar 

  11. Wu G, Broniscer A, McEachron TA et al (2012) Somatic histone H3 alterations in pediatric diffuse intrinsic pontine gliomas and non-brainstem glioblastomas. Nat Genet 44:251–253

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Wu G, Diaz AK, Paugh BS et al (2014) The genomic landscape of diffuse intrinsic pontine glioma and pediatric non-brainstem high-grade glioma. Nat Genet 46:444–450

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Hermann A, Gowher H, Jeltsch A (2004) Biochemistry and biology of mammalian DNA methyltransferases. Cell Mol Life Sci 61:2571–2587

    Article  CAS  PubMed  Google Scholar 

  14. Quina AS, Buschbeck M, Di Croce L (2006) Chromatin structure and epigenetics. Biochem Pharmacol 72:1563–1569

    Article  CAS  PubMed  Google Scholar 

  15. Goll MG, Bestor TH (2005) Eukaryotic cytosine methyltransferases. Annu Rev Biochem 74:481–514

    Article  CAS  PubMed  Google Scholar 

  16. Huang Y, Rao A (2014) Connections between TET proteins and aberrant DNA modification in cancer. Trends Genet 30:464–474

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Bird AP (1986) CpG-rich islands and the function of DNA methylation. Nature 321:209–213

    Article  CAS  PubMed  Google Scholar 

  18. Esteller M (2008) Epigenetics in cancer. N Engl J Med 358:1148–1159

    Article  CAS  PubMed  Google Scholar 

  19. Esteller M (2007) Epigenetic gene silencing in cancer: the DNA hypermethylome. Hum Mol Genet 16(Spec No 1):R50–R59

    Article  CAS  PubMed  Google Scholar 

  20. Berdasco M, Esteller M (2010) Aberrant epigenetic landscape in cancer: how cellular identity goes awry. Dev Cell 19:698–711

    Article  CAS  PubMed  Google Scholar 

  21. Costello JF, Berger MS, Huang HS et al (1996) Silencing of p16/CDKN2 expression in human gliomas by methylation and chromatin condensation. Cancer Res 56:2405–2410

    CAS  PubMed  Google Scholar 

  22. Watanabe T, Yokoo H, Yokoo M et al (2001) Concurrent inactivation of RB1 and TP53 pathways in anaplastic oligodendrogliomas. J Neuropathol Exp Neurol 60:1181–1189

    Article  CAS  PubMed  Google Scholar 

  23. Nakamura M, Yonekawa Y, Kleihues P et al (2001) Promoter hypermethylation of the RB1 gene in glioblastomas. Lab Invest 81:77–82

    Article  CAS  PubMed  Google Scholar 

  24. Bello MJ, Rey JA (2006) The p53/Mdm2/p14(ARF) cell cycle control pathway genes may be inactivated by genetic and epigenetic mechanisms in gliomas. Cancer Genet Cytogen 164:172–173

    Article  CAS  Google Scholar 

  25. Amatya VJ, Naumann U, Weller M et al (2005) TP53 promoter methylation in human gliomas. Acta neuropathologica 110:178–184

    Article  CAS  PubMed  Google Scholar 

  26. Lambiv WL, Vassallo I, Delorenzi M et al (2011) The Wnt inhibitory factor 1 (WIF1) is targeted in glioblastoma and has a tumor suppressing function potentially by induction of senescence. Neuro-Oncology 13:736–747

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Gotze S, Wolter M, Reifenberger G et al (2010) Frequent promoter hypermethylation of Wnt pathway inhibitor genes in malignant astrocytic gliomas. Int J Cancer 126:2584–2593

    PubMed  Google Scholar 

  28. Toyota M, Ahuja N, Ohe-Toyota M et al (1999) CpG island methylator phenotype in colorectal cancer. Proc Natl Acad Sci USA 96:8681–8686

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Noushmehr H, Weisenberger DJ, Diefes K et al (2010) Identification of a CpG island methylator phenotype that defines a distinct subgroup of glioma. Cancer Cell 17:510–522

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Turcan S, Rohle D, Goenka A et al (2012) IDH1 mutation is sufficient to establish the glioma hypermethylator phenotype. Nature 483:479–483

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Mardis ER, Ding L, Dooling DJ et al (2009) Recurring mutations found by sequencing an acute myeloid leukemia genome. N Engl J Med 361:1058–1066

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Amary MF, Bacsi K, Maggiani F et al (2011) IDH1 and IDH2 mutations are frequent events in central chondrosarcoma and central and periosteal chondromas but not in other mesenchymal tumours. J Pathol 224:334–343

    Article  CAS  PubMed  Google Scholar 

  33. Farshidfar F, Zheng S, Gingras MC et al (2017) Integrative genomic analysis of cholangiocarcinoma identifies distinct IDH-mutant molecular profiles. Cell Rep 19:2878–2880

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Suzuki H, Aoki K, Chiba K et al (2015) Mutational landscape and clonal architecture in grade II and III gliomas. Nat Genet 47:458–468

    Article  CAS  PubMed  Google Scholar 

  35. Hartmann C, Meyer J, Balss J et al (2009) Type and frequency of IDH1 and IDH2 mutations are related to astrocytic and oligodendroglial differentiation and age: a study of 1,010 diffuse gliomas. Acta Neuropathol 118:469–474

    Article  PubMed  Google Scholar 

  36. Dang L, White DW, Gross S et al (2010) Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature 465:966

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Ward PS, Patel J, Wise DR et al (2010) The common feature of leukemia-associated IDH1 and IDH2 mutations is a neomorphic enzyme activity converting alpha-ketoglutarate to 2-hydroxyglutarate. Cancer Cell 17:225–234

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Clark O, Yen K, Mellinghoff IK (2016) Molecular pathways: isocitrate dehydrogenase mutations in cancer. Clin Cancer Res 22:1837–1842

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Bunse L, Pusch S, Bunse T et al (2018) Suppression of antitumor T cell immunity by the oncometabolite (R)-2-hydroxyglutarate. Nat Med 24:1192–1203

    Article  CAS  PubMed  Google Scholar 

  40. Hegi ME, Diserens AC, Gorlia T et al (2005) MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med 352:997–1003

    Article  CAS  PubMed  Google Scholar 

  41. Stupp R, Mason WP, van den Bent MJ et al (2005) Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 352:987–996

    Article  CAS  PubMed  Google Scholar 

  42. Juratli TA, Kirsch M, Geiger K et al (2012) The prognostic value of IDH mutations and MGMT promoter status in secondary high-grade gliomas. J Neurooncol 110:325–333

    Article  CAS  PubMed  Google Scholar 

  43. Stupp R, Hegi ME, Gorlia T et al (2014) Cilengitide combined with standard treatment for patients with newly diagnosed glioblastoma with methylated MGMT promoter (CENTRIC EORTC 26071–22072 study): a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol 15:1100–1108

    Article  CAS  PubMed  Google Scholar 

  44. Baumert BG, Hegi ME, van den Bent MJ et al (2016) Temozolomide chemotherapy versus radiotherapy in high-risk low-grade glioma (EORTC 22033–26033): a randomised, open-label, phase 3 intergroup study. Lancet Oncol 17:1521–1532

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Bady P, Delorenzi M, Hegi ME (2016) 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 18:350–361

    Article  CAS  PubMed  Google Scholar 

  46. Fernandez AF, Assenov Y, Martin-Subero JI et al (2012) A DNA methylation fingerprint of 1628 human samples. Genome Res 22:407–419

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Moran S, Martinez-Cardus A, Sayols S et al (2016) Epigenetic profiling to classify cancer of unknown primary: a multicentre, retrospective analysis. Lancet Oncol 17:1386–1395

    Article  PubMed  Google Scholar 

  48. Sturm D, Orr BA, Toprak UH et al (2016) New brain tumor entities emerge from molecular classification of CNS-PNETs. Cell 164:1060–1072

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Sahm F, Schrimpf D, Stichel D et al (2017) DNA methylation-based classification and grading system for meningioma: a multicentre, retrospective analysis. Lancet Oncol 18:682–694

    Article  CAS  PubMed  Google Scholar 

  50. Capper D, Jones DTW, Sill M et al (2018) DNA methylation-based classification of central nervous system tumours. Nature 555:469–474

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Ceccarelli M, Barthel FP, Malta TM et al (2016) Molecular profiling reveals biologically discrete subsets and pathways of progression in diffuse glioma. Cell 164:550–563

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Verhaak RG, Hoadley KA, Purdom E et al (2010) Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 17:98–110

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Brat DJ, Aldape K, Colman H et al (2018) cIMPACT-NOW update 3: recommended diagnostic criteria for “Diffuse astrocytic glioma, IDH-wildtype, with molecular features of glioblastoma, WHO grade IV”. Acta Neuropathol 136:805–810

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Aoki K, Nakamura H, Suzuki H et al (2018) Prognostic relevance of genetic alterations in diffuse lower-grade gliomas. Neuro Oncol 20:66–77

    Article  CAS  PubMed  Google Scholar 

  55. Cancer Genome Atlas Research N, Brat DJ, Verhaak RG et al (2015) Comprehensive, integrative genomic analysis of diffuse lower-grade gliomas. N Engl J Med 372:2481–2498

    Article  CAS  Google Scholar 

  56. Olar A, Wani KM, Alfaro-Munoz KD et al (2015) IDH mutation status and role of WHO grade and mitotic index in overall survival in grade II-III diffuse gliomas. Acta Neuropathol 129:585–596

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. de Souza CF, Sabedot TS, Malta TM et al (2018) A distinct DNA methylation shift in a subset of glioma CpG island methylator phenotypes during tumor recurrence. Cell Rep 23:637–651

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Mazor T, Pankov A, Johnson BE et al (2015) DNA methylation and somatic mutations converge on the cell cycle and define similar evolutionary histories in brain tumors. Cancer Cell 28:307–317

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Bai H, Harmanci AS, Erson-Omay EZ et al (2016) Integrated genomic characterization of IDH1-mutant glioma malignant progression. Nat Genet 48:59–66

    Article  CAS  PubMed  Google Scholar 

  60. Yang X, Han H, De Carvalho DD et al (2014) Gene body methylation can alter gene expression and is a therapeutic target in cancer. Cancer Cell 26:577–590

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Singer M, Kosti I, Pachter L et al (2015) A diverse epigenetic landscape at human exons with implication for expression. Nucleic Acids Res 43:3498–3508

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Flavahan WA, Drier Y, Liau BB et al (2016) Insulator dysfunction and oncogene activation in IDH mutant gliomas. Nature 529:110–114

    Article  CAS  PubMed  Google Scholar 

  63. Malta TM, de Souza CF, Sabedot TS et al (2018) Glioma CpG island methylator phenotype (G-CIMP): biological and clinical implications. Neuro Oncol 20:608–620

    Article  CAS  PubMed  Google Scholar 

  64. Jones PA, Issa JP, Baylin S (2016) Targeting the cancer epigenome for therapy. Nat Rev Genet 17:630–641

    Article  CAS  PubMed  Google Scholar 

  65. Lewis PW, Muller MM, Koletsky MS et al (2013) Inhibition of PRC2 activity by a gain-of-function H3 mutation found in pediatric glioblastoma. Science 340:857–861

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Mund C, Brueckner B, Lyko F (2006) Reactivation of epigenetically silenced genes by DNA methyltransferase inhibitors basic concepts and clinical applications. Epigenetics-Us 1:7–13

    Google Scholar 

  67. Issa JP (2005) Optimizing therapy with methylation inhibitors in myelodysplastic syndromes: dose, duration, and patient selection. Nat Clin Pract Oncol 2(Suppl 1):S24–S29

    Article  CAS  PubMed  Google Scholar 

  68. Kaminskas E, Farrell A, Abraham S et al (2005) Approval summary: azacitidine for treatment of myelodysplastic syndrome subtypes. Clin Cancer Res 11:3604–3608

    Article  CAS  PubMed  Google Scholar 

  69. Tsai HC, Li H, Van Neste L et al (2012) Transient low doses of DNA-demethylating agents exert durable antitumor effects on hematological and epithelial tumor cells. Cancer Cell 21:430–446

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Chiappinelli KB, Strissel PL, Desrichard A et al (2017) Inhibiting DNA methylation causes an interferon response in cancer via dsRNA including endogenous retroviruses. Cell 169:361

    Article  CAS  PubMed  Google Scholar 

  71. Roulois D, Loo Yau H, Singhania R et al (2015) DNA-demethylating agents target colorectal cancer cells by inducing viral mimicry by endogenous transcripts. Cell 162:961–973

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Kantarjian HM, Roboz GJ, Kropf PL et al (2017) Guadecitabine (SGI-110) in treatment-naive patients with acute myeloid leukaemia: phase 2 results from a multicentre, randomised, phase 1/2 trial. Lancet Oncol 18:1317–1326

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Losman JA, Looper RE, Koivunen P et al (2013) R)-2-hydroxyglutarate is sufficient to promote leukemogenesis and its effects are reversible. Science 339:1621–1625

    Article  CAS  PubMed  Google Scholar 

  74. Stein EM (2015) IDH2 inhibition in AML: finally progress? Best Pract Res Cl Ha 28:112–115

    Article  Google Scholar 

  75. Yen K, Travins J, Wang F et al (2017) AG-221, a first-in-class therapy targeting acute myeloid leukemia harboring oncogenic IDH2 mutations. Cancer Discov 7:478–493

    Article  CAS  PubMed  Google Scholar 

  76. Stein EM, DiNardo CD, Pollyea DA et al (2017) Enasidenib in mutant IDH2 relapsed or refractory acute myeloid leukemia. Blood 130:722–731

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. DiNardo CD, Stein EM, de Botton S et al (2018) Durable remissions with ivosidenib in IDH1-mutated relapsed or refractory AML. New Engl J Med 378:2386–2398

    Article  CAS  PubMed  Google Scholar 

  78. Rohle D, Popovici-Muller J, Palaskas N et al (2013) An inhibitor of mutant IDH1 delays growth and promotes differentiation of glioma cells. Science 340:626–630

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Tateishi K, Wakimoto H, Iafrate AJ et al (2015) Extreme vulnerability of IDH1 mutant cancers to NAD + depletion. Cancer Cell 28:773–784

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Mazor T, Chesnelong C, Pankov A et al (2017) Clonal expansion and epigenetic reprogramming following deletion or amplification of mutant IDH1. Proc Natl Acad Sci USA 114:10743–10748

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Mellinghoff IK, Touat M, Maher E et al (2017) Ag-120, a first-in-class mutant Idh1 inhibitor in patients with recurrent or progressive Idh1 mutant glioma: updated results from the phase 1 non-enhancing glioma population. Neuro-Oncology 19:10–11

    Article  Google Scholar 

Download references

Acknowledgements

We would like to thank Editage (http://www.editage.jp) for English language editing.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Kosuke Aoki.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Aoki, K., Natsume, A. Overview of DNA methylation in adult diffuse gliomas. Brain Tumor Pathol 36, 84–91 (2019). https://doi.org/10.1007/s10014-019-00339-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10014-019-00339-w

Keywords

Navigation