Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Primer
  • Published:

Glioma

Abstract

Gliomas are primary brain tumours that are thought to derive from neuroglial stem or progenitor cells. On the basis of their histological appearance, they have been traditionally classified as astrocytic, oligodendroglial or ependymal tumours and assigned WHO grades I–IV, which indicate different degrees of malignancy. Tremendous progress in genomic, transcriptomic and epigenetic profiling has resulted in new concepts of classifying and treating gliomas. Diffusely infiltrating gliomas in adults are now separated into three overarching tumour groups with distinct natural histories, responses to treatment and outcomes: isocitrate dehydrogenase (IDH)-mutant, 1p/19q co-deleted tumours with mostly oligodendroglial morphology that are associated with the best prognosis; IDH-mutant, 1p/19q non-co-deleted tumours with mostly astrocytic histology that are associated with intermediate outcome; and IDH wild-type, mostly higher WHO grade (III or IV) tumours that are associated with poor prognosis. Gliomas in children are molecularly distinct from those in adults, the majority being WHO grade I pilocytic astrocytomas characterized by circumscribed growth, favourable prognosis and frequent BRAF gene fusions or mutations. Ependymal tumours can be molecularly subdivided into distinct epigenetic subgroups according to location and prognosis. Although surgery, radiotherapy and alkylating agent chemotherapy are still the mainstay of treatment, individually tailored strategies based on tumour-intrinsic dominant signalling pathways and antigenic tumour profiles may ultimately improve outcome. For an illustrated summary of this Primer, visit: http://go.nature.com/TXY7Ri

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Relative frequency of primary brain and central nervous system tumours.
Figure 2: Brain cells and brain tumours.
Figure 3: Age-adjusted incidence of primary brain and central nervous system tumours by histology and age group.
Figure 4: Important genetic and epigenetic alterations in the different types of gliomas.
Figure 5: Biochemical consequences of glioma-associated isocitrate dehydrogenase mutations.
Figure 6: Neuroimaging and histological features of gliomas.
Figure 7: Molecular marker-based therapeutic approach to gliomas.

Similar content being viewed by others

References

  1. Gavrilovic, I. T. & Posner, J. B. Brain metastases: epidemiology and pathophysiology. J. Neurooncol. 75, 5–14 (2005).

    Article  PubMed  Google Scholar 

  2. Ferlay, J., Parkin, D. M. & Steliarova-Foucher, E. Estimates of cancer incidence and mortality in Europe in 2008. Eur. J. Cancer 46, 765–781 (2010).

    Article  CAS  PubMed  Google Scholar 

  3. Ostrom, Q. T. et al. CBTRUS statistical report: primary brain and central nervous system tumors diagnosed in the United States in 2007–2011. Neuro Oncol. 16, iv1–iv63 (2014). A comprehensive and updated report on the epidemiology of primary brain tumours in the United States.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Nakamura, H., Makino, K., Yano, S., Kuratsu, J. & Kumamoto Brain Tumor Research Group. Epidemiological study of primary intracranial tumors: a regional survey in Kumamoto prefecture in southern Japan—20-year study. Int. J. Clin. Oncol. 16, 314–321 (2011).

    Article  PubMed  Google Scholar 

  5. Kallio, M. The incidence of intracranial gliomas in southern Finland. Acta Neurol. Scand. 78, 480–483 (1988).

    Article  CAS  PubMed  Google Scholar 

  6. Bondy, M. L. et al. Brain tumor epidemiology: consensus from the Brain Tumor Epidemiology Consortium. Cancer 113, 1953–1968 (2008).

    Article  PubMed  Google Scholar 

  7. Connelly, J. M. & Malkin, M. G. Environmental risk factors for brain tumors. Curr. Neurol. Neurosci. Rep. 7, 208–214 (2007).

    Article  PubMed  Google Scholar 

  8. Ostrom, Q. T. & Barnholtz-Sloan, J. S. Current state of our knowledge on brain tumor epidemiology. Curr. Neurol. Neurosci. Rep. 11, 329–335 (2011).

    Article  PubMed  Google Scholar 

  9. Ron, E. et al. Tumors of the brain and nervous system after radiotherapy in childhood. N. Engl. J. Med. 319, 1033–1039 (1988).

    Article  CAS  PubMed  Google Scholar 

  10. Sadetzki, S. et al. Long-term follow-up for brain tumor development after childhood exposure to ionizing radiation for tinea capitis. Radiat. Res. 163, 424–432 (2005).

    Article  CAS  PubMed  Google Scholar 

  11. Neglia, J. P. et al. New primary neoplasms of the central nervous system in survivors of childhood cancer: a report from the Childhood Cancer Survivor Study. J. Natl Cancer Inst. 98, 1528–1537 (2006).

    Article  PubMed  Google Scholar 

  12. Brada, M. et al. Risk of second brain tumour after conservative surgery and radiotherapy for pituitary adenoma. BMJ 304, 1343–1346 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Minniti, G., Traish, D., Ashley, S., Gonsalves, A. & Brada, M. Risk of second brain tumor after conservative surgery and radiotherapy for pituitary adenoma: update after an additional 10 years. J. Clin. Endocrinol. Metab. 90, 800–804 (2005).

    Article  CAS  PubMed  Google Scholar 

  14. INTERPHONE Study Group. Brain tumour risk in relation to mobile telephone use: results of the INTERPHONE international case–control study. Int. J. Epidemiol. 39, 675–694 (2010).

    Article  Google Scholar 

  15. Ostrom, Q. T. et al. The epidemiology of glioma in adults: a “state of the science” review. Neuro Oncol. 16, 896–913 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Linos, E., Raine, T., Alonso, A. & Michaud, D. Atopy and risk of brain tumors: a meta-analysis. J. Natl Cancer Inst. 99, 1544–1550 (2007).

    Article  PubMed  Google Scholar 

  17. Ohgaki, H., Kim, Y. H. & Steinbach, J. P. Nervous system tumors associated with familial tumor syndromes. Curr. Opin. Neurol. 23, 583–591 (2010).

    Article  CAS  PubMed  Google Scholar 

  18. Bainbridge, M. N. et al. Germline mutations in shelterin complex genes are associated with familial glioma. J. Natl Cancer Inst. 107, 384 (2015).

    Article  CAS  PubMed  Google Scholar 

  19. Shete, S. et al. Genome-wide association study identifies five susceptibility loci for glioma. Nat. Genet. 41, 899–904 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Wrensch, M. et al. Variants in the CDKN2B and RTEL1 regions are associated with high-grade glioma susceptibility. Nat. Genet. 41, 905–908 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Jenkins, R. B. et al. A low-frequency variant at 8q24.21 is strongly associated with risk of oligodendroglial tumors and astrocytomas with IDH1 or IDH2 mutation. Nat. Genet. 44, 1122–1125 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Rice, T. et al. Inherited variant on chromosome 11q23 increases susceptibility to IDH-mutated but not IDH-normal gliomas regardless of grade or histology. Neuro Oncol. 15, 535–541 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Stacey, S. N. et al. A germline variant in the TP53 polyadenylation signal confers cancer susceptibility. Nat. Genet. 43, 1098–1103 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Andersson, U. et al. A comprehensive study of the association between the EGFR and ERBB2 genes and glioma risk. Acta Oncol. 49, 767–775 (2010).

    Article  CAS  PubMed  Google Scholar 

  25. Enciso-Mora, V. et al. Deciphering the 8q24.21 association for glioma. Hum. Mol. Genet. 22, 2293–2302 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Walsh, K. M. et al. Variants near TERT and TERC influencing telomere length are associated with high-grade glioma risk. Nat. Genet. 46, 731–735 (2014). The identification of risk alleles for glioma near TERC and TERT implicates a role for telomerase in gliomagenesis.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Rajaraman, P. et al. Genome-wide association study of glioma and meta-analysis. Hum. Genet. 131, 1877–1888 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Yunoue, S. et al. Neurofibromatosis type I tumor suppressor neurofibromin regulates neuronal differentiation via its GTPase-activating protein function toward Ras. J. Biol. Chem. 278, 26958–26969 (2003).

    Article  CAS  PubMed  Google Scholar 

  29. Pfister, S. et al. BRAF gene duplication constitutes a mechanism of MAPK pathway activation in low-grade astrocytomas. J. Clin. Invest. 118, 1739–1749 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Jones, D. T. et al. Tandem duplication producing a novel oncogenic BRAF fusion gene defines the majority of pilocytic astrocytomas. Cancer Res. 68, 8673–8677 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Jones, D. T. et al. Recurrent somatic alterations of FGFR1 and NTRK2 in pilocytic astrocytoma. Nat. Genet. 45, 927–932 (2013). This study provides a comprehensive overview of the mutational profile in pilocytic astrocytomas and demonstrates that these tumours are a single-pathway disease invariably driven by aberrant activation of MAPK pathway signalling.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Zhang, J. et al. Whole-genome sequencing identifies genetic alterations in pediatric low-grade gliomas. Nat. Genet. 45, 602–612 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Gronych, J. et al. An activated mutant BRAF kinase domain is sufficient to induce pilocytic astrocytoma in mice. J. Clin. Invest. 121, 1344–1348 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. National Cancer Institute. Selumetinib in treating young patients with recurrent or refractory low grade glioma. ClinicalTrials.gov[online], (2010).

  35. Jacob, K. et al. Genetic aberrations leading to MAPK pathway activation mediate oncogene-induced senescence in sporadic pilocytic astrocytomas. Clin. Cancer Res. 17, 4650–4660 (2011).

    Article  CAS  PubMed  Google Scholar 

  36. Raabe, E. H. et al. BRAF activation induces transformation and then senescence in human neural stem cells: a pilocytic astrocytoma model. Clin. Cancer Res. 17, 3590–3599 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Schindler, G. et al. Analysis of BRAF V600E mutation in 1,320 nervous system tumors reveals high mutation frequencies in pleomorphic xanthoastrocytoma, ganglioglioma and extra-cerebellar pilocytic astrocytoma. Acta Neuropathol. 121, 397–405 (2011).

    Article  CAS  PubMed  Google Scholar 

  38. Weber, R. G. et al. Frequent loss of chromosome 9, homozygous CDKN2A/p14ARF/CDKN2B deletion and low TSC1 mRNA expression in pleomorphic xanthoastrocytomas. Oncogene 26, 1088–1097 (2007).

    Article  CAS  PubMed  Google Scholar 

  39. Koelsche, C. et al. BRAF-mutated pleomorphic xanthoastrocytoma is associated with temporal location, reticulin fiber deposition and CD34 expression. Brain Pathol. 24, 221–229 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Robinson, J. P. et al. Activated BRAF induces gliomas in mice when combined with Ink4a/Arf loss or Akt activation. Oncogene 29, 335–344 (2010).

    Article  CAS  PubMed  Google Scholar 

  41. Huillard, E. et al. Cooperative interactions of BRAFV600E kinase and CDKN2A locus deficiency in pediatric malignant astrocytoma as a basis for rational therapy. Proc. Natl Acad. Sci. USA 109, 8710–8715 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Chan, J. A. et al. Pathogenesis of tuberous sclerosis subependymal giant cell astrocytomas: biallelic inactivation of TSC1 or TSC2 leads to mTOR activation. J. Neuropathol. Exp. Neurol. 63, 1236–1242 (2004).

    Article  CAS  PubMed  Google Scholar 

  43. Zhou, J. et al. Tsc1 mutant neural stem/progenitor cells exhibit migration deficits and give rise to subependymal lesions in the lateral ventricle. Genes Dev. 25, 1595–1600 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Krueger, D. A. et al. Everolimus for subependymal giant-cell astrocytomas in tuberous sclerosis. N. Engl. J. Med. 363, 1801–1811 (2010).

    Article  CAS  PubMed  Google Scholar 

  45. Franz, D. N. et al. Everolimus for subependymal giant cell astrocytoma in patients with tuberous sclerosis complex: 2-year open-label extension of the randomised EXIST-1 study. Lancet Oncol. 15, 1513–1520 (2014).

    Article  CAS  PubMed  Google Scholar 

  46. Sahm, F. et al. Farewell to oligoastrocytoma: in situ molecular genetics favor classification as either oligodendroglioma or astrocytoma. Acta Neuropathol. 128, 551–559 (2014).

    Article  CAS  PubMed  Google Scholar 

  47. Killela, P. J. et al. Mutations in IDH1, IDH2, and in the TERT promoter define clinically distinct subgroups of adult malignant gliomas. Oncotarget 5, 1515–1525 (2014).

    PubMed  PubMed Central  Google Scholar 

  48. Yan, H. et al. IDH1 and IDH2 mutations in gliomas. N. Engl. J. Med. 360, 765–773 (2009). This paper identifies IDH1 and IDH2 mutations as frequent genetic alterations in diffusely infiltrating astrocytic and oligodendroglial tumours.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Balss, J. et al. Analysis of the IDH1 codon 132 mutation in brain tumors. Acta Neuropathol. 116, 597–602 (2008). A report on frequent IDH1 mutations in diffusely infiltrating astrocytic and oligodendroglial tumours.

    Article  CAS  PubMed  Google Scholar 

  50. Watanabe, T., Vital, A., Nobusawa, S., Kleihues, P. & Ohgaki, H. Selective acquisition of IDH1 R132C mutations in astrocytomas associated with Li-Fraumeni syndrome. Acta Neuropathol. 117, 653–656 (2009).

    Article  CAS  PubMed  Google Scholar 

  51. Reitman, Z. J. & Yan, H. Isocitrate dehydrogenase 1 and 2 mutations in cancer: alterations at a crossroads of cellular metabolism. J. Natl Cancer Inst. 102, 932–941 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Xu, W. et al. Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of α-ketoglutarate-dependent dioxygenases. Cancer Cell 19, 17–30 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Turcan, S. et al. IDH1 mutation is sufficient to establish the glioma hypermethylator phenotype. Nature 483, 479–483 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Noushmehr, H. et al. Identification of a CpG island methylator phenotype that defines a distinct subgroup of glioma. Cancer Cell 17, 510–522 (2010). Identification of g-CIMP and its association with IDH mutation in gliomas.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Sasaki, M. et al. D-2-hydroxyglutarate produced by mutant IDH1 perturbs collagen maturation and basement membrane function. Genes Dev. 26, 2038–2049 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Bettegowda, C. et al. Mutations in CIC and FUBP1 contribute to human oligodendroglioma. Science 333, 1453–1455 (2011). Using large-scale exome sequencing, the authors found that 1p/19q co-deleted oligodendrogliomas frequently carry mutations in the CIC and FUBP1 genes on 19q and 1p, respectively.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Sahm, F. et al. CIC and FUBP1 mutations in oligodendrogliomas, oligoastrocytomas and astrocytomas. Acta Neuropathol. 123, 853–860 (2012).

    Article  CAS  PubMed  Google Scholar 

  58. Weber, R. G. et al. Characterization of genomic alterations associated with glioma progression by comparative genomic hybridization. Oncogene 13, 983–994 (1996).

    CAS  PubMed  Google Scholar 

  59. Ohgaki, H. et al. Genetic pathways to glioblastoma: a population-based study. Cancer Res. 64, 6892–6899 (2004).

    Article  CAS  PubMed  Google Scholar 

  60. Klink, B. et al. A novel, diffusely infiltrative xenograft model of human anaplastic oligodendroglioma with mutations in FUBP1, CIC, and IDH1. PLoS ONE 8, e59773 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Kelly, J. J. et al. Oligodendroglioma cell lines containing t(1;19)(q10;p10). Neuro Oncol. 12, 745–755 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Luchman, H. A. et al. An in vivo patient-derived model of endogenous IDH1-mutant glioma. Neuro Oncol. 14, 184–191 (2012).

    Article  CAS  PubMed  Google Scholar 

  63. Rohle, D. et al. An inhibitor of mutant IDH1 delays growth and promotes differentiation of glioma cells. Science 340, 626–630 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Ramkissoon, L. A. et al. Genomic analysis of diffuse pediatric low-grade gliomas identifies recurrent oncogenic truncating rearrangements in the transcription factor MYBL1. Proc. Natl Acad. Sci. USA 110, 8188–8193 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  65. Sturm, D. et al. Hotspot mutations in H3F3A and IDH1 define distinct epigenetic and biological subgroups of glioblastoma. Cancer Cell 22, 425–437 (2012). This study characterizes six biologically distinct subtypes of glioblastoma on the basis of DNA methylation profiles and associated genetic alterations.

    Article  CAS  PubMed  Google Scholar 

  66. Appin, C. L. & Brat, D. J. Molecular pathways in gliomagenesis and their relevance to neuropathologic diagnosis. Adv. Anat. Pathol. 22, 50–58 (2015).

    Article  CAS  PubMed  Google Scholar 

  67. Brennan, C. W. et al. The somatic genomic landscape of glioblastoma. Cell 155, 462–477 (2013). This paper provides a comprehensive and in-depth overview of the genetic, epigenetic and transcriptional profiles associated with glioblastoma.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Cancer Genome Atlas Research Network. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 455, 1061–1068 (2008).

    Article  CAS  Google Scholar 

  69. Plate, K. H., Breier, G., Weich, H. A. & Risau, W. Vascular endothelial growth factor is a potential tumour angiogenesis factor in human gliomas in vivo. Nature 359, 845–848 (1992).

    Article  CAS  PubMed  Google Scholar 

  70. Frei, K. et al. Transforming growth factor-β pathway activity in glioblastoma. Oncotarget 6, 5963–5977 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Buczkowicz, P. et al. Genomic analysis of diffuse intrinsic pontine gliomas identifies three molecular subgroups and recurrent activating ACVR1 mutations. Nat. Genet. 46, 451–456 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Hashizume, R. et al. Pharmacologic inhibition of histone demethylation as a therapy for pediatric brainstem glioma. Nat. Med. 20, 1394–1396 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Ebert, C. et al. Molecular genetic analysis of ependymal tumors. NF2 mutations and chromosome 22q loss occur preferentially in intramedullary spinal ependymomas. Am. J. Pathol. 155, 627–632 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Parker, M. et al. C11orf95RELA fusions drive oncogenic NF-κB signalling in ependymoma. Nature 506, 451–455 (2014). Identification of a specific fusion gene that is characteristic of a subset of supratentorial, mostly anaplastic ependymomas with poor clinical outcome.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Mack, S. C. et al. Epigenomic alterations define lethal CIMP-positive ependymomas of infancy. Nature 506, 445–450 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Wani, K. et al. A prognostic gene expression signature in infratentorial ependymoma. Acta Neuropathol. 123, 727–738 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Witt, H. et al. Delineation of two clinically and molecularly distinct subgroups of posterior fossa ependymoma. Cancer Cell 20, 143–157 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Pajtler, K. W. et al. Molecular classification of ependymal tumors across all CNS compartments, histopathological grades, and age groups. Cancer Cell 27, 728–743 (2015). This study provides compelling evidence for distinct molecular subtypes of ependymoma, including three subtypes each among spinal, infratentorial and supratentorial ependymomas.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. la Fougè re, C., Suchorska, B., Bartenstein, P., Kreth, F. W. & Tonn, J. C. Molecular imaging of gliomas with PET: opportunities and limitations. Neuro Oncol. 13, 806–819 (2011).

    Article  Google Scholar 

  81. World Health Organization. WHO Classification of Tumours of the Central Nervous System (eds Louis, D. N., Ohgaki, H., Wiestler, O. D. & Cavenee, W. K. ) (WHO Publications, 2007) This is the foundation of modern brain tumour diagnosis.

    Google Scholar 

  82. Weller, M. et al. Molecular neuro-oncology in clinical practice: a new horizon. Lancet Oncol. 14, e370–e379 (2013).

    Article  CAS  PubMed  Google Scholar 

  83. Weller, M. et al. EANO guideline for the diagnosis and treatment of anaplastic gliomas and glioblastoma. Lancet Oncol. 15, e395–e403 (2014).

    Article  PubMed  Google Scholar 

  84. Eigenbrod, S. et al. Molecular stereotactic biopsy technique improves diagnostic accuracy and enables personalized treatment strategies in glioma patients. Acta Neurochir. (Wien) 156, 1427–1440 (2014).

    Article  Google Scholar 

  85. Parsons, D. W. et al. An integrated genomic analysis of human glioblastoma multiforme. Science 321, 1807–1812 (2008). This paper is the first to identify IDH1 mutations in a subset of glioblastomas of younger patients and most secondary glioblastomas.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Ohgaki, H. & Kleihues, P. The definition of primary and secondary glioblastoma. Clin. Cancer Res. 19, 764–772 (2013).

    Article  CAS  PubMed  Google Scholar 

  87. Capper, D., Zentgraf, H., Balss, J., Hartmann, C. & von Deimling, A. Monoclonal antibody specific for IDH1 R132H mutation. Acta Neuropathol. 118, 599–601 (2009). A report on the development of a mutation-specific monoclonal antibody against IDHR132H that is suitable for immunohistochemistry on formalin-fixed paraffin-embedded tissue sections.

    Article  CAS  PubMed  Google Scholar 

  88. Felsberg, J. et al. Rapid and sensitive assessment of the IDH1 and IDH2 mutation status in cerebral gliomas based on DNA pyrosequencing. Acta Neuropathol. 119, 501–507 (2010).

    Article  CAS  PubMed  Google Scholar 

  89. Reuss, D. E. et al. ATRX and IDH1-R132H immunohistochemistry with subsequent copy number analysis and IDH sequencing as a basis for an “integrated” diagnostic approach for adult astrocytoma, oligodendroglioma and glioblastoma. Acta Neuropathol. 129, 133–146 (2015).

    Article  CAS  PubMed  Google Scholar 

  90. Weller, M. et al. Personalized care in neuro-oncology coming of age: why we need MGMT and 1p/19q testing for malignant glioma patients in clinical practice. Neuro Oncol. 14, iv100–iv108 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Weller, M. et al. MGMT promoter methylation in malignant gliomas: ready for personalized medicine? Nat. Rev. Neurol. 6, 39–51 (2010).

    Article  CAS  PubMed  Google Scholar 

  92. Wick, W. et al. MGMT testing — the challenges for biomarker-based glioma treatment. Nat. Rev. Neurol. 10, 372–385 (2014).

    Article  CAS  PubMed  Google Scholar 

  93. Hegi, M. E. et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. N. Engl. J. Med. 352, 997–1003 (2005). This study sets the basis for MGMT as a predictive biomarker for glioma treatment.

    Article  CAS  PubMed  Google Scholar 

  94. Stupp, R. et al. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol. 10, 459–466 (2009). A long-term follow-up of the study that established temozolomide as the standard of care in newly diagnosed glioblastoma.

    Article  CAS  PubMed  Google Scholar 

  95. Malmstrom, A. 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. 13, 916–926 (2012).

    Article  CAS  PubMed  Google Scholar 

  96. Wick, W. et al. Temozolomide chemotherapy alone versus radiotherapy alone for malignant astrocytoma in the elderly: the NOA-08 randomised, phase 3 trial. Lancet Oncol. 13, 707–715 (2012).

    Article  CAS  PubMed  Google Scholar 

  97. Bady, P. et al. MGMT methylation analysis of glioblastoma on the Infinium methylation BeadChip identifies two distinct CpG regions associated with gene silencing and outcome, yielding a prediction model for comparisons across datasets, tumor grades, and CIMP-status. Acta Neuropathol. 124, 547–560 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Labussiere, M. et al. TERT promoter mutations in gliomas, genetic associations and clinico-pathological correlations. Br. J. Cancer 111, 2024–2032 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Wick, W. et al. Prognostic or predictive value of MGMT promoter methylation in gliomas depends on IDH1 mutation. Neurology 81, 1515–1522 (2013).

    Article  CAS  PubMed  Google Scholar 

  100. Weller, M. et al. Molecular classification of diffuse cerebral WHO grade II/III gliomas using genome- and transcriptome-wide profiling improves stratification of prognostically distinct patient groups. Acta Neuropathol. 129, 679–693 (2015).

    Article  CAS  PubMed  Google Scholar 

  101. Suzuki, H. et al. Mutational landscape and clonal architecture in grade II and III gliomas. Nat. Genet. 47, 458–468 (2015). This study provides a comprehensive mutational profile of WHO grade II and III astrocytic and oligodendroglial tumours based on next-generation sequencing data.

    Article  CAS  PubMed  Google Scholar 

  102. The Cancer Genome Atlas Research Network. A comprehensive, integrative genomic analysis of diffuse lower-grade gliomas study of ICT-107 immunotherapy in glioblastoma multiforme (GBM). N. Engl. J. Med. 372, 2481–2498 (2015).

    Article  CAS  PubMed Central  Google Scholar 

  103. Eckel-Passow, J. E. et al. Glioma groups based on 1p/19q, IDH, and TERT promoter mutations in tumors. N. Engl. J. Med. 372, 2499–2508 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Aldape, K. D. et al. Immunohistochemical detection of EGFRvIII in high malignancy grade astrocytomas and evaluation of prognostic significance. J. Neuropathol. Exp. Neurol. 63, 700–707 (2004).

    Article  CAS  PubMed  Google Scholar 

  105. Hegi, M. E., Rajakannu, P. & Weller, M. Epidermal growth factor receptor: a re-emerging target in glioblastoma. Curr. Opin. Neurol. 25, 774–779 (2012).

    Article  CAS  PubMed  Google Scholar 

  106. Sampson, J. H. et al. Immunologic escape after prolonged progression-free survival with epidermal growth factor receptor variant III peptide vaccination in patients with newly diagnosed glioblastoma. J. Clin. Oncol. 28, 4722–4729 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  107. Korshunov, A. et al. Combined molecular analysis of BRAF and IDH1 distinguishes pilocytic astrocytoma from diffuse astrocytoma. Acta Neuropathol. 118, 401–405 (2009).

    Article  CAS  PubMed  Google Scholar 

  108. Tian, Y. et al. Detection of KIAA1549BRAF fusion transcripts in formalin-fixed paraffin-embedded pediatric low-grade gliomas. J. Mol. Diagn. 13, 669–677 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Lin, A. et al. BRAF alterations in primary glial and glioneuronal neoplasms of the central nervous system with identification of 2 novel KIAA1549:BRAF fusion variants. J. Neuropathol. Exp. Neurol. 71, 66–72 (2012).

    Article  CAS  PubMed  Google Scholar 

  110. Capper, D. et al. Assessment of BRAF V600E mutation status by immunohistochemistry with a mutation-specific monoclonal antibody. Acta Neuropathol. 122, 11–19 (2011).

    Article  CAS  PubMed  Google Scholar 

  111. Gutmann, D. H., Listernick, R. & Ferner, R. E. Screening for symptomatic optic pathway glioma in children with neurofibromatosis type 1. Eye (Lond.) 25, 818; author reply 818–819 (2011).

    Article  CAS  Google Scholar 

  112. Mandonnet, E. et al. Silent diffuse low-grade glioma: toward screening and preventive treatment? Cancer 120, 1758–1762 (2014).

    Article  PubMed  Google Scholar 

  113. Vernooij, M. W. et al. Incidental findings on brain MRI in the general population. N. Engl. J. Med. 357, 1821–1828 (2007).

    Article  CAS  PubMed  Google Scholar 

  114. Vogelbaum, M. A. et al. Application of novel response/progression measures for surgically delivered therapies for gliomas: Response Assessment in Neuro-Oncology (RANO) Working Group. Neurosurgery 70, 234–243; discussion 243–244 (2012).

    Article  PubMed  Google Scholar 

  115. Wen, P. Y. et al. Updated response assessment criteria for high-grade gliomas: Response Assessment in Neuro-Oncology Working Group. J. Clin. Oncol. 28, 1963–1972 (2010). This article began a process of refining and standardizing imaging criteria.

    Article  PubMed  Google Scholar 

  116. Pope, W. B. & Hessel, C. Response Assessment in Neuro-Oncology criteria: implementation challenges in multicenter neuro-oncology trials. AJNR Am. J. Neuroradiol. 32, 794–797 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Hygino da Cruz, L. C. Jr et al. Pseudoprogression and pseudoresponse: imaging challenges in the assessment of posttreatment glioma. AJNR Am. J. Neuroradiol. 32, 1978–1985 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  118. Chinot, O. L. et al. Response assessment criteria for glioblastoma: practical adaptation and implementation in clinical trials of antiangiogenic therapy. Curr. Neurol. Neurosci. Rep. 13, 347 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Radbruch, A. et al. Comparison of susceptibility weighted imaging and TOF-angiography for the detection of thrombi in acute stroke. PLoS ONE 8, e63459 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Kickingereder, P. et al. Primary central nervous system lymphoma and atypical glioblastoma: multiparametric differentiation by using diffusion-, perfusion-, and susceptibility-weighted MR imaging. Radiology 272, 843–850 (2014).

    Article  PubMed  Google Scholar 

  121. Ellingson, B. M. et al. Pretreatment ADC histogram analysis is a predictive imaging biomarker for bevacizumab treatment but not chemotherapy in recurrent glioblastoma. AJNR Am. J. Neuroradiol. 35, 673–679 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Ellingson, B. M. et al. Comparison between intensity normalization techniques for dynamic susceptibility contrast (DSC)-MRI estimates of cerebral blood volume (CBV) in human gliomas. J. Magn. Reson. Imaging 35, 1472–1477 (2012).

    Article  PubMed  Google Scholar 

  123. Roth, P., Wick, W. & Weller, M. Steroids in neurooncology: actions, indications, side-effects. Curr. Opin. Neurol. 23, 597–602 (2010).

    Article  CAS  PubMed  Google Scholar 

  124. Weller, M., Stupp, R. & Wick, W. Epilepsy meets cancer: when, why, and what to do about it? Lancet Oncol. 13, e375–e382 (2012).

    Article  PubMed  Google Scholar 

  125. Karajannis, M. A. et al. Phase II study of sorafenib in children with recurrent or progressive low-grade astrocytomas. Neuro Oncol. 16, 1408–1416 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Krueger, D. A. et al. Everolimus long-term safety and efficacy in subependymal giant cell astrocytoma. Neurology 80, 574–580 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Chamberlain, M. C. Salvage therapy with BRAF inhibitors for recurrent pleomorphic xanthoastrocytoma: a retrospective case series. J. Neurooncol. 114, 237–240 (2013).

    Article  CAS  PubMed  Google Scholar 

  128. Lee, E. Q., Ruland, S., LeBoeuf, N. R., Wen, P. Y. & Santagata, S. Successful treatment of a progressive BRAF V600E-mutated anaplastic pleomorphic xanthoastrocytoma with vemurafenib monotherapy. J. Clin. Oncol. http://dx.doi.org/10.1200/JCO.2013.51.1766 (2014).

  129. Kros, J. M. et al. Panel review of anaplastic oligodendroglioma from European Organization for Research and Treatment of Cancer Trial 26951: assessment of consensus in diagnosis, influence of 1p/19q loss, and correlations with outcome. J. Neuropathol. Exp. Neurol. 66, 545–551 (2007).

    Article  CAS  PubMed  Google Scholar 

  130. Claus, E. B. et al. Survival rates in patients with low-grade glioma after intraoperative magnetic resonance image guidance. Cancer 103, 1227–1233 (2005).

    Article  PubMed  Google Scholar 

  131. Chang, E. F. et al. Seizure characteristics and control following resection in 332 patients with low-grade gliomas. J. Neurosurg. 108, 227–235 (2008).

    Article  PubMed  Google Scholar 

  132. Pallud, J. et al. Epileptic seizures in diffuse low-grade gliomas in adults. Brain 137, 449–462 (2014).

    Article  PubMed  Google Scholar 

  133. De Witt Hamer, P. C., Robles, S. G., Zwinderman, A. H., Duffau, H. & Berger, M. S. Impact of intraoperative stimulation brain mapping on glioma surgery outcome: a meta-analysis. J. Clin. Oncol. 30, 2559–2565 (2012).

    Article  PubMed  Google Scholar 

  134. Smith, J. S. et al. Role of extent of resection in the long-term outcome of low-grade hemispheric gliomas. J. Clin. Oncol. 26, 1338–1345 (2008).

    Article  PubMed  Google Scholar 

  135. Ius, T. et al. Low-grade glioma surgery in eloquent areas: volumetric analysis of extent of resection and its impact on overall survival. A single-institution experience in 190 patients: clinical article. J. Neurosurg. 117, 1039–1052 (2012).

    Article  PubMed  Google Scholar 

  136. Jakola, A. S. et al. Comparison of a strategy favoring early surgical resection versus a strategy favoring watchful waiting in low-grade gliomas. JAMA 308, 1881–1888 (2012).

    Article  CAS  PubMed  Google Scholar 

  137. Pignatti, F. et al. Prognostic factors for survival in adult patients with cerebral low-grade glioma. J. Clin. Oncol. 20, 2076–2084 (2002). This paper provides a score aiding the decision about when to treat patients with WHO grade II gliomas.

    Article  PubMed  Google Scholar 

  138. Daniels, T. B. et al. Validation of EORTC prognostic factors for adults with low-grade glioma: a report using intergroup 86-72-51. Int. J. Radiat. Oncol. Biol. Phys. 81, 218–224 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  139. Soffietti, R. et al. Guidelines on management of low-grade gliomas: report of an EFNS-EANO Task Force. Eur. J. Neurol. 17, 1124–1133 (2010).

    Article  CAS  PubMed  Google Scholar 

  140. van den Bent, M. J. et al. Long-term efficacy of early versus delayed radiotherapy for low-grade astrocytoma and oligodendroglioma in adults: the EORTC 22845 randomised trial. Lancet 366, 985–990 (2005).

    Article  CAS  PubMed  Google Scholar 

  141. Shaw, E. G. et al. Randomized trial of radiation therapy plus procarbazine, lomustine, and vincristine chemotherapy for supratentorial adult low-grade glioma: initial results of RTOG 9802. J. Clin. Oncol. 30, 3065–3070 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. van den Bent, M. J. et al. Adjuvant procarbazine, lomustine, and vincristine chemotherapy in newly diagnosed anaplastic oligodendroglioma: long-term follow-up of EORTC brain tumor group study 26951. J. Clin. Oncol. 31, 344–350 (2013).

    Article  CAS  PubMed  Google Scholar 

  143. Cairncross, G. et al. Phase III trial of chemoradiotherapy for anaplastic oligodendroglioma: long-term results of RTOG 9402. J. Clin. Oncol. 31, 337–343 (2013). References 142 and 143 are long-term analyses of randomized trials in patients with anaplastic oligodendroglial tumours. They establish 1p/19q co-deletion as a predictive biomarker and chemoradiotherapy as the standard of care for postsurgical treatment of patients with 1p/19q co-deleted tumours.

    Article  CAS  PubMed  Google Scholar 

  144. Baumert, B. G. et al. Temozolomide chemotherapy versus radiotherapy in molecularly characterized (1p loss) low-grade glioma: a randomized phase III intergroup study by the EORTC/NCIC-CTG/TROG/MRC-CTU (EORTC 22033–26033). J. Clin. Oncol. Abstr. 31, 2007 (2013).

    Google Scholar 

  145. Wick, W. et al. NOA-04 randomized phase III trial of sequential radiochemotherapy of anaplastic glioma with procarbazine, lomustine, and vincristine or temozolomide. J. Clin. Oncol. 27, 5874–5880 (2009).

    Article  CAS  PubMed  Google Scholar 

  146. National Cancer Institute. Radiation therapy with or without temozolomide in treating patients with anaplastic glioma. ClinicalTrials.gov[online], 1 (2008).

  147. Turcan, S. et al. Efficient induction of differentiation and growth inhibition in IDH1 mutant glioma cells by the DNMT inhibitor decitabine. Oncotarget 4, 1729–1736 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  148. Castle, J. C. et al. Exploiting the mutanome for tumor vaccination. Cancer Res. 72, 1081–1091 (2012).

    Article  CAS  PubMed  Google Scholar 

  149. Schumacher, T. et al. A vaccine targeting mutant IDH1 induces antitumour immunity. Nature 512, 324–327 (2014).

    Article  CAS  PubMed  Google Scholar 

  150. Mohme, M., Neidert, M. C., Regli, L., Weller, M. & Martin, R. Immunological challenges for peptide-based immunotherapy in glioblastoma. Cancer Treat. Rev. 40, 248–258 (2014).

    Article  CAS  PubMed  Google Scholar 

  151. Han, S. J., Zygourakis, C., Lim, M. & Parsa, A. T. Immunotherapy for glioma: promises and challenges. Neurosurg. Clin. N. Am. 23, 357–370 (2012).

    Article  PubMed  Google Scholar 

  152. Sanai, N., Polley, M. Y., McDermott, M. W., Parsa, A. T. & Berger, M. S. An extent of resection threshold for newly diagnosed glioblastomas. J. Neurosurg. 115, 3–8 (2011).

    Article  PubMed  Google Scholar 

  153. Stummer, W. et al. Fluorescence-guided surgery with 5-aminolevulinic acid for resection of malignant glioma: a randomised controlled multicentre phase III trial. Lancet Oncol. 7, 392–401 (2006). This study establishes a role for extent of resection in newly diagnosed glioblastoma.

    Article  CAS  PubMed  Google Scholar 

  154. Stummer, W. et al. Extent of resection and survival in glioblastoma multiforme: identification of and adjustment for bias. Neurosurgery 62, 564–576; discussion 564–576 (2008).

    Article  PubMed  Google Scholar 

  155. Gilbert, M. R. et al. Dose-dense temozolomide for newly diagnosed glioblastoma: a randomized phase III clinical trial. J. Clin. Oncol. 31, 4085–4091 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Stupp, R. et al. 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 (2014).

    Article  CAS  PubMed  Google Scholar 

  157. Gilbert, M. R. et al. A randomized trial of bevacizumab for newly diagnosed glioblastoma. N. Engl. J. Med. 370, 699–708 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Chinot, O. L. et al. Bevacizumab plus radiotherapy–temozolomide for newly diagnosed glioblastoma. N. Engl. J. Med. 370, 709–722 (2014).

    Article  CAS  PubMed  Google Scholar 

  159. Gomez, D. R. et al. High failure rate in spinal ependymomas with long-term follow-up. Neuro Oncol. 7, 254–259 (2005).

    Article  PubMed  PubMed Central  Google Scholar 

  160. Merchant, T. E. et al. Preliminary results from a phase II trial of conformal radiation therapy and evaluation of radiation-related CNS effects for pediatric patients with localized ependymoma. J. Clin. Oncol. 22, 3156–3162 (2004).

    Article  PubMed  Google Scholar 

  161. Timmermann, B. et al. Combined postoperative irradiation and chemotherapy for anaplastic ependymomas in childhood: results of the German prospective trials HIT 88/89 and HIT 91. Int. J. Radiat. Oncol. Biol. Phys. 46, 287–295 (2000).

    Article  CAS  PubMed  Google Scholar 

  162. Brandes, A. A. et al. A multicenter retrospective study of chemotherapy for recurrent intracranial ependymal tumors in adults by the Gruppo Italiano Cooperativo di Neuro-Oncologia. Cancer 104, 143–148 (2005).

    Article  PubMed  Google Scholar 

  163. M. D. Anderson Cancer Center. Dose-dense temozolomide + lapatinib for recurrent ependymoma. ClinicalTrials.gov[online], (2009).

  164. University of Texas Southwestern Medical Center. Everolimus for children with recurrent or progressive ependymoma. ClinicalTrials.gov[online], (2014).

  165. M. D. Anderson Cancer Center. Carboplatin and bevacizumab for recurrent ependymoma. ClinicalTrials.gov[online], (2011).

  166. Ann & Robert H Lurie Children's Hospital of Chicago. Carboplatin as a radiosensitizer in treating childhood ependymoma. ClinicalTrials.gov[online], (2010).

  167. Dirven, L., Reijneveld, J. C. & Taphoorn, M. J. Health-related quality of life or quantity of life: a difficult trade-off in primary brain tumors? Semin. Oncol. 41, 541–552 (2014).

    Article  PubMed  Google Scholar 

  168. Klein, M. Neurocognitive functioning in adult WHO grade II gliomas: impact of old and new treatment modalities. Neuro Oncol. 14, iv17–iv24 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  169. Taphoorn, M. J. et al. Health-related quality of life in patients with glioblastoma: a randomised controlled trial. Lancet Oncol. 6, 937–944 (2005). This paper identifies the substantial impairment in quality of life that is associated with glioblastoma and its treatment.

    Article  PubMed  Google Scholar 

  170. Fliessbach, K. et al. Computer-based assessment of cognitive functions in brain tumor patients. J. Neurooncol. 100, 427–437 (2010).

    Article  PubMed  Google Scholar 

  171. Douw, L. et al. Cognitive and radiological effects of radiotherapy in patients with low-grade glioma: long-term follow-up. Lancet Neurol. 8, 810–818 (2009).

    Article  PubMed  Google Scholar 

  172. Cella, D. F. et al. The Functional Assessment of Cancer Therapy scale: development and validation of the general measure. J. Clin. Oncol. 11, 570–579 (1993).

    Article  CAS  PubMed  Google Scholar 

  173. Weitzner, M. A. et al. The Functional Assessment of Cancer Therapy (FACT) scale. Development of a brain subscale and revalidation of the general version (FACT-G) in patients with primary brain tumors. Cancer 75, 1151–1161 (1995).

    Article  CAS  PubMed  Google Scholar 

  174. Mauer, M. et al. The prognostic value of health-related quality-of-life data in predicting survival in glioblastoma cancer patients: results from an international randomised phase III EORTC Brain Tumour and Radiation Oncology Groups, and NCIC Clinical Trials Group study. Br. J. Cancer 97, 302–307 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Lin, E., Rosenthal, M. A., Le, B. H. & Eastman, P. Neuro-oncology and palliative care: a challenging interface. Neuro Oncol. 14, iv3–iv7 (2012). This paper highlights the role that palliative care may have in neuro-oncology.

    Article  PubMed  PubMed Central  Google Scholar 

  176. Patel, A. P. et al. Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science 344, 1396–1401 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Louis, D. N. et al. International Society of Neuropathology—Haarlem consensus guidelines for nervous system tumor classification and grading. Brain Pathol. 24, 429–435 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  178. Ozawa, T. et al. Most human non-GCIMP glioblastoma subtypes evolve from a common proneural-like precursor glioma. Cancer Cell 26, 288–300 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Sottoriva, A. et al. Intratumor heterogeneity in human glioblastoma reflects cancer evolutionary dynamics. Proc. Natl Acad. Sci. USA 110, 4009–4014 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  180. Celldex Therapeutics. Phase III study of rindopepimut/GM-CSF in patients with newly diagnosed glioblastoma (ACT IV). ClinicalTrials.gov[online], (2011).

  181. Northwest Biotherapeutics. Study of a drug [DCVax®-L] to treat newly diagnosed GBM brain cancer. ClinicalTrials.gov[online], (2002).

  182. Wen, P. et al. A randomized double blind placebo-controlled phase 2 trial of dendritic cell (DC) vaccine ICT-107 following standard treatment in newly diagnosed patients with GBM. Neuro Oncol. Abstr. 16, v22 (2014).

    Article  Google Scholar 

  183. Dutoit, V. et al. Exploiting the glioblastoma peptidome to discover novel tumour-associated antigens for immunotherapy. Brain 135, 1042–1054 (2012).

    Article  PubMed  Google Scholar 

  184. Britten, C. M. et al. The regulatory landscape for actively personalized cancer immunotherapies. Nat. Biotech. 31, 880–882 (2013).

    Article  CAS  Google Scholar 

  185. immatics Biotechnologies GmbH. GAPVAC Phase I trial in newly diagnosed glioblastoma patients. ClinicalTrials.gov[online], (2014).

  186. Rodon, J. et al. First-in-human dose study of the novel transforming growth factor-β receptor I kinase inhibitor LY2157299 monohydrate in patients with advanced cancer and glioma. Clin. Cancer Res. 21, 553–560 (2015).

    Article  CAS  PubMed  Google Scholar 

  187. Beatty, G. L. et al. Phase I study of the safety, pharmacokinetics (PK), and pharmacodynamics (PD) of the oral inhibitor of indoleamine 2,3-dioxygenase (IDO1) INCB024360 in patients (pts) with advanced malignancies. J. Clin. Oncol. Abstr. 31, 3025 (2013).

    Google Scholar 

  188. Hodi, F. S. et al. Improved survival with ipilimumab in patients with metastatic melanoma. N. Engl. J. Med. 363, 711–723 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  189. Ribas, A. et al. Phase III randomized clinical trial comparing tremelimumab with standard-of-care chemotherapy in patients with advanced melanoma. J. Clin. Oncol. 31, 616–622 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Wolchok, J. D. et al. Nivolumab plus ipilimumab in advanced melanoma. N. Engl. J. Med. 369, 122–133 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Mitchell, D. A. et al. Tetanus toxoid and CCL3 improve dendritic cell vaccines in mice and glioblastoma patients. Nature 519, 366–369 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Kaufmann, J. K. & Chiocca, E. A. Glioma virus therapies between bench and bedside. Neuro Oncol. 16, 334–351 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  193. Stupp, R. et al. Interim analysis of the EF-14 Trial: a prospective, multi-center trial of NovoTTF-100A together with temozolomide compared to temozolomide alone in patients with newly diagnosed GBM. Neuro Oncol. Abstr. 16, v167 (2014).

    Article  Google Scholar 

  194. Galanis, E. et al. Phase 2 trial design in neuro-oncology revisited: a report from the RANO group. Lancet Oncol. 13, e196–e204 (2012).

    Article  PubMed  Google Scholar 

  195. Gan, H. K. et al. A phase 1 study evaluating ABT-414 with temozolomide (TMZ) or concurrent radiotherapy (RT) and TMZ in glioblastoma (GBM). Neuro Oncol. Abstr. 16, v83 (2014).

    Article  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Contributions

Introduction (P.Y.W. and R.S.); Epidemiology (M. Brada and R.N.); Mechanisms/pathophysiology (G.R. and S.M.P.); Diagnosis, screening and prevention (K.A. and S.M.P.); Management (W.W. and M. Berger); Quality of life (M.R. and R.S.); Outlook (G.R., W.W. and M.W.); overview of Primer (M.W.).

Corresponding author

Correspondence to Michael Weller.

Ethics declarations

Competing interests

M. Berger serves as a consultant for Ivivi Health Sciences. M. Brada served on advisory boards for Merck Serono, Roche and AbbVie. R.N. has received honoraria for lectures or advisory board participation, or sponsorship for meetings from MSD, Roche, Chugai, Nobelpharma, Eisai and Novocure. G.R. has received research grants from Roche and Merck, as well as honoraria for lectures or advisory boards from Roche and Amgen. R.S. has served on advisory boards for AbbVie, Actelion, Merck Serono, MSD, Novartis, Pfizer and Roche, and is or has been the coordinating investigator for sponsored clinical trials evaluating temozolomide (MSD), cilengitide (Merck Serono) and Tumour Treating Fields (Novocure). M.W. has received research grants from Acceleron, Actelion, Alpinia Institute, Bayer, Isarna, MSD, Merck Serono, Novocure, PIQUR and Roche, and honoraria for lectures or advisory board participation from AbbVie, Celldex, Isarna, MagForce, MSD, Merck Serono, Novartis, Novocure, Pfizer, Roche and Teva. P.Y.W. has received research grants from Amgen, AngioChem, AstraZeneca, Exelixis, Genentech/Roche, GlaxoSmithKline, Merck, Novartis, Sanofi-Aventis and Vascular Biogenics, and honoraria for lectures or advisory board participation from AbbVie, Celldex, Foundation Medicine, Genentech/Roche, Merck, Novartis, Vascular Biogenics, Midatech and Monteris. W.W. has received research grants from Apogenix, Boehringer Ingelheim, Eli Lilly, immatics, MSD and Roche, as well as honoraria for lectures or advisory board participation from MSD and Roche. W.W. is or has been the coordinating investigator for sponsored clinical trials evaluating APG101 (Apogenix), bevacizumab (Roche), galunisertib (Eli Lilly), temozolomide (MSD) and temsirolimus (Pfizer). S.M.P. and M.R. and K.A. declare no competing interests.

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Weller, M., Wick, W., Aldape, K. et al. Glioma. Nat Rev Dis Primers 1, 15017 (2015). https://doi.org/10.1038/nrdp.2015.17

Download citation

  • Published:

  • DOI: https://doi.org/10.1038/nrdp.2015.17

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer