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.

  • Review Article
  • Published:

Mechanisms of Disease: genetic predictors of response to treatment in brain tumors

Abstract

Brain tumors are currently diagnosed on the basis of their histology. The most common types in adults are astrocytomas, oligodendrogliomas and oligoastrocytomas or mixed tumors, which almost invariably lead to death. Improvements in outcome have been elusive despite intensive research. Recent findings indicate that response to conventional therapy, at least in some cases, correlates better with genetic characteristics than histopathology. An understanding of the molecular mechanisms that underlie the malignant phenotype of gliomas also provides the possibility of rational design of molecularly targeted therapies. This approach has proved successful in other areas of oncology. As many tumors have the same types of molecular abnormalities, molecular targeted therapies developed for nonbrain tumor types might be adapted for the treatment of brain tumors. There are a number of unique problems involved in treating tumors in the brain that must be overcome. The genetic predictors of response to conventional therapies, the genes and cellular mechanisms involved in glioma development, and potential therapeutic targets are reviewed. The possibility of designing tailored molecular therapy based on the molecular characteristics of the tumors is also explored.

Key Points

  • Patients with tumors that have hemizygous loss of 1p and 19q chromosomal arms have a survival advantage, which might be a predictor of response to conventional treatment with irradiation and alkylating agents

  • Methylation of MGMT in glioblastomas seems to predict a good response to alkylating agents, particularly temozolomide

  • Genetic and transcriptome studies indicate that technologies assessing global genetic changes and gene expression in brain tumors may provide information relevant for prognostication (prognostic indicators) and choice of therapy (therapy response indicators)

  • The introduction of molecularly targeted therapies will obligate a molecular (i.e. genetic and/or expression) analysis of tumor tissue to help determine the appropriate therapy in an individual case

  • Combinations of molecularly targeted therapies with or without conventional therapies are likely to be required for success

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: Adult diffuse astrocytic tumors
Figure 2: The proteins involved in the RB1 and p53 pathways with some interactions with the P13K/PKB pathway
Figure 3: Schematic diagram of the receptor tyrosine kinase and signaling pathways implicated in glioblastoma development
Figure 4: Histology of oligodendroglioma WHO grade II and anaplastic oligodendroglioma WHO grade III tumors

Similar content being viewed by others

References

  1. Hegi ME et al. (2005) MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med 352: 997–1003

    Article  CAS  Google Scholar 

  2. Carlson RW et al. (2006) NCCN Task Force Report: adjuvant therapy for breast cancer. J Natl Compr Canc Netw 4 (Suppl 1): S1–S26

    PubMed  Google Scholar 

  3. Tauchi T and Ohyashiki K (2006) The second generation of BCR-ABL tyrosine kinase inhibitors. Int J Hematol 83: 294–300

    Article  CAS  Google Scholar 

  4. Schwartzbaum JA et al. (2006) Epidemiology and molecular pathology of glioma. Nat Clin Pract Neurol 2: 494–503

    Article  Google Scholar 

  5. Corn B et al. (1994). Malignant oligodendroglioma arising after radiation therapy for lymphoma. Med Pediatr Oncol 22: 45–52

    Article  CAS  Google Scholar 

  6. Rollison DE et al. (2005) Investigation of human brain tumors for the presence of polyomavirus genome sequences by two independent laboratories. Int J Cancer 113: 769–774

    Article  CAS  Google Scholar 

  7. Kleihues P et al. (1995) Genetic and environmental factors in the etiology of human brain tumors. Toxicol Lett 82–83: 601–605

    Article  Google Scholar 

  8. Malmer B et al. (2001) Genetic epidemiology of glioma. Br J Cancer 84: 429–434

    Article  CAS  Google Scholar 

  9. Kleihues P and Cavenee WK (2000) Pathology and Genetics of Tumours of the Nervous System. Lyon: International Agency for Research on Cancer (series eds Kleihues P and Sobin LH)

    Google Scholar 

  10. Breedveld P et al. (2005) The effect of Bcrp1 (Abcg2) on the in vivo pharmacokinetics and brain penetration of imatinib mesylate (Gleevec): implications for the use of breast cancer resistance protein and P-glycoprotein inhibitors to enable the brain penetration of imatinib in patients. Cancer Res 65: 2577–2582

    Article  CAS  Google Scholar 

  11. Schinkel AH (1999) P-Glycoprotein, a gatekeeper in the blood-brain barrier. Adv Drug Deliv Rev 36: 179–194

    Article  CAS  Google Scholar 

  12. Kleihues P and Ohgaki H (1999) Primary and secondary glioblastomas: from concept to clinical diagnosis. Neuro-oncol 1: 44–51

    Article  CAS  Google Scholar 

  13. McCormack BM et al. (1992) Treatment and survival of low-grade astrocytoma in adults—1977–1988. Neurosurgery 31: 636–642

    CAS  PubMed  Google Scholar 

  14. Vertosick FT Jr et al. (1991) Survival of patients with well-differentiated astrocytomas diagnosed in the era of computed tomography. Neurosurgery 28: 496–501

    Article  Google Scholar 

  15. Stupp R et al. (2005) Optimal role of temozolomide in the treatment of malignant gliomas. Curr Neurol Neurosci Rep 5: 198–206

    Article  CAS  Google Scholar 

  16. Perry A et al. (1999) Clinicopathologic study of 85 similarly treated patients with anaplastic astrocytic tumors: an analysis of DNA content (ploidy), cellular proliferation, and p53 expression. Cancer 86: 672–683

    Article  CAS  Google Scholar 

  17. Rasheed A et al. (2000) Molecular markers of prognosis in astrocytic tumors. Cancer 94: 2688–2697

    Article  Google Scholar 

  18. Smith JS et al. (2001) PTEN mutation, EGFR amplification, and outcome in patients with anaplastic astrocytoma and glioblastoma multiforme. J Natl Cancer Inst 93: 1246–1256

    Article  CAS  Google Scholar 

  19. Ichimura K et al. (2000) Deregulation of the p14ARF/MDM2/p53 pathway is a prerequisite for human astrocytic gliomas with G1-S transition control gene abnormalities. Cancer Res 60: 417–424

    CAS  PubMed  Google Scholar 

  20. James CD et al. (1989) Mitotic recombination of chromosome 17 in astrocytomas. Proc Natl Acad Sci USA 86: 2858–2862

    Article  CAS  Google Scholar 

  21. Koschny R et al. (2002) Comparative genomic hybridization in glioma: a meta-analysis of 509 cases. Cancer Genet Cytogenet 135: 147–159

    Article  CAS  Google Scholar 

  22. Hermanson M et al. (1992) Platelet-derived growth factor and its receptors in human glioma tissue: expression of messenger RNA and protein suggests the presence of autocrine and paracrine loops. Cancer Res 52: 3213–3219

    CAS  PubMed  Google Scholar 

  23. Ekstrand AJ et al. (1991) Genes for epidermal growth factor receptor, transforming growth factor alpha, and epidermal growth factor and their expression in human gliomas in vivo. Cancer Res 51: 2164–2172

    CAS  PubMed  Google Scholar 

  24. Ohgaki H and Kleihues P (2005) Population-based studies on incidence, survival rates, and genetic alterations in astrocytic and oligodendroglial gliomas. J Neuropathol Exp Neurol 64: 479–489

    Article  CAS  Google Scholar 

  25. von Deimling A et al. (2000) Comprehensive allelotype and genetic anaysis of 466 human nervous system tumors. J Neuropathol Exp Neurol 59: 544–558

    Article  CAS  Google Scholar 

  26. von Deimling A et al. (1994) Loci associated with malignant progression in astrocytomas: a candidate on chromosome 19q. Cancer Res 54: 1397–1401

    CAS  PubMed  Google Scholar 

  27. Backlund LM et al. (2005) Mutations in Rb1 pathway-related genes are associated with poor prognosis in anaplastic astrocytomas. Br J Cancer 93: 124–130

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  29. Reifenberger J et al. (1996) Analysis of p53 mutation and epidermal growth factor receptor amplification in recurrent gliomas with malignant progression. J Neuropathol Exp Neurol 55: 822–831

    Article  CAS  Google Scholar 

  30. Reifenberger G et al. (1993) Amplification and overexpression of the MDM2 gene in a subset of human malignant gliomas without p53 mutations. Cancer Res 53: 2736–2739

    CAS  PubMed  Google Scholar 

  31. Riemenschneider MJ et al. (1999) Amplification and overexpression of the MDM4 (MDMX) gene from 1q32 in a subset of malignant gliomas without TP53 mutation or MDM2 amplification. Cancer Res 59: 6091–6096

    CAS  PubMed  Google Scholar 

  32. Sherr CJ and McCormick F (2002) The RB and p53 pathways in cancer. Cancer Cell 2: 103–112

    Article  CAS  Google Scholar 

  33. Schmidt EE et al. (1994) CDKN2 (p16/MTS1) gene deletion or CDK4 amplification occurs in the majority of glioblastomas. Cancer Res 54: 6321–6324

    CAS  PubMed  Google Scholar 

  34. Reifenberger G et al. (1994) Amplification of multiple genes from chromosomal region 12q13-14 in human malignant gliomas: preliminary mapping of the amplicons shows preferential involvement of CDK4, SAS, and MDM2. Cancer Res 54: 4299–4303

    CAS  PubMed  Google Scholar 

  35. Buschges R et al. (1999) Amplification and expression of cyclin D genes (CCND1, CCND2 and CCND3) in human malignant gliomas. Brain Pathol 9: 435–442; discussion 432–433

    Article  CAS  Google Scholar 

  36. Ichimura K et al. (1996) Human glioblastomas with no alterations of the CDKN2A (p16INK4A, MTS1) and CDK4 genes have frequent mutations of the retinoblastoma gene. Oncogene 13: 1065–1072

    CAS  PubMed  Google Scholar 

  37. Ichimura K et al. (2004) Molecular pathogenesis of astrocytic tumours. J Neurooncol 70: 137–160

    Article  Google Scholar 

  38. Nakamura M et al. (2001) Promoter hypermethylation of the RB1 gene in glioblastomas. Lab Invest 81: 77–82

    Article  CAS  Google Scholar 

  39. Biernat W et al. (1997) Alterations of cell cycle regulatory genes in primary (de novo) and secondary glioblastomas. Acta Neuropathol (Berl) 94: 303–309

    Article  CAS  Google Scholar 

  40. Libermann TA et al. (1985) Amplification, enhanced expression and possible rearrangement of EGF receptor gene in primary human brain tumours of glial origin. Nature 313: 144–147

    Article  CAS  Google Scholar 

  41. Liu L et al. (2005) Clinical significance of EGFR amplification and the aberrant EGFRvIII transcript in conventionally treated astrocytic gliomas. J Mol Med 83: 917–926

    Article  CAS  Google Scholar 

  42. Okada Y et al. (2003) Selection pressures of TP53 mutation and microenvironmental location influence epidermal growth factor receptor gene amplification in human glioblastomas. Cancer Res 63: 413–416

    CAS  PubMed  Google Scholar 

  43. Liu L et al. (2000) The complexity of the 7p12 amplicon in human astrocytic gliomas: detailed mapping of 246 tumors. J Neuropathol Exp Neurol 59: 1087–1093

    Article  CAS  Google Scholar 

  44. Huang HS et al. (1997) The enhanced tumorigenic activity of a mutant epidermal growth factor receptor common in human cancers is mediated by threshold levels of constitutive tyrosine phosphorylation and unattenuated signaling. J Biol Chem 272: 2927–2935

    Article  CAS  Google Scholar 

  45. Collins VP (1995) Gene amplification in human gliomas. Glia 15: 289–296

    Article  CAS  Google Scholar 

  46. Fleming TP et al. (1992) Amplification and/or overexpression of platelet-derived growth factor receptors and epidermal growth factor receptor in human glial tumors. Cancer Res 52: 4550–4553

    CAS  PubMed  Google Scholar 

  47. Knobbe CB et al. (2004) Mutation analysis of the Ras pathway genes NRAS, HRAS, KRAS and BRAF in glioblastomas. Acta Neuropathol (Berl) 108: 467–470

    Article  CAS  Google Scholar 

  48. Steck PA et al. (1997) Identification of a candidate tumour suppressor gene, MMAC1, at chromosome 10q23.3 that is mutated in multiple advanced cancers. Nat Genet 15: 356–362

    Article  CAS  Google Scholar 

  49. Schmidt E et al. (1999) Mutational profile of the PTEN/MMAC1 gene in primary human astrocytic tumors and xenografts. J Neuropathol Exp Neurol 58: 1170–1183

    Article  CAS  Google Scholar 

  50. Knobbe CB and Reifenberger G (2003) Genetic alterations and aberrant expression of genes related to the phosphatidyl-inositol-3′-kinase/protein kinase B (Akt) signal transduction pathway in glioblastomas. Brain Pathol 13: 507–518

    Article  CAS  Google Scholar 

  51. Samuels Y et al. (2004) High frequency of mutations of the PIK3CA gene in human cancers. Science 304: 554

    Article  CAS  Google Scholar 

  52. Knobbe CB et al. (2005) Genetic alteration and expression of the phosphoinositol-3-kinase/Akt pathway genes PIK3 CA and PIKE in human glioblastomas. Neuropathol Appl Neurobiol 31: 486–490

    Article  CAS  Google Scholar 

  53. Knobbe CB et al. (2004) Hypermethylation and transcriptional downregulation of the carboxyl-terminal modulator protein gene in glioblastomas. J Natl Cancer Inst 96: 483–486

    Article  CAS  Google Scholar 

  54. Rubio-Viqueira B and Hidalgo M (2006) Targeting mTOR for cancer treatment. Curr Opin Investig Drugs 7: 501–512

    CAS  PubMed  Google Scholar 

  55. Backlund LM et al. (2003) Short postoperative survival for glioblastoma patients with a dysfunctional Rb1 pathway in combination with no wild-type PTEN. Clin Cancer Res 9: 4151–4158

    PubMed  Google Scholar 

  56. Tso CL et al. (2006) Distinct transcription profiles of primary and secondary glioblastoma subgroups. Cancer Res 66: 159–167

    Article  CAS  Google Scholar 

  57. Phillips HS et al. (2006) Molecular subclasses of high-grade glioma predict prognosis, delineate a pattern of disease progression, and resemble stages in neurogenesis. Cancer Cell 9: 157–173

    Article  CAS  Google Scholar 

  58. Cairncross JG and Macdonald DR (1988) Successful chemotherapy for recurrent malignant oligodendroglioma. Ann Neurol 23: 360–364

    Article  CAS  Google Scholar 

  59. Reifenberger J et al. (1994) Molecular genetic analysis of oligodendroglial tumors shows preferential allelic deletions on 19q and 1p. Am J Pathol 145: 1175–1190

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Griffin CA et al. (2006) Identification of der(1;19)(q10;p10) in five oligodendrogliomas suggests mechanism of concurrent 1p and 19q loss. J Neuropathol Exp Neurol 65: 988–994

    Article  Google Scholar 

  61. Jenkins RB et al. (2006) A t(1;19)(q10;p10) mediates the combined deletions of 1p and 19q and predicts a better prognosis of patients with oligodendroglioma. Cancer Res 66: 9852–9861

    Article  CAS  Google Scholar 

  62. Cairncross JG 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  Google Scholar 

  63. Cairncross G et al. (2006) Phase III trial of chemotherapy plus radiotherapy compared with radiotherapy alone for pure and mixed anaplastic oligodendroglioma: Intergroup Radiation Therapy Oncology Group Trial 9402. J Clin Oncol 24: 2707–2714

    Article  CAS  Google Scholar 

  64. van den Bent MJ et al. (2006) Adjuvant procarbazine, lomustine, and vincristine improves progression-free survival but not overall survival in newly diagnosed anaplastic oligodendrogliomas and oligoastrocytomas: a randomized European Organisation for Research and Treatment of Cancer phase III trial. J Clin Oncol 24: 2715–2722

    Article  CAS  Google Scholar 

  65. Mollemann M et al. (2005) Frequent promoter hypermethylation and low expression of the MGMT gene in oligodendroglial tumors. Int J Cancer 113: 379–385

    Article  Google Scholar 

  66. Idbaih A et al. (2005) Two types of chromosome 1p losses with opposite significance in gliomas. Ann Neurol 58: 483–487

    Article  CAS  Google Scholar 

  67. Reifenberger G and Louis DN (2003) Oligodendroglioma: toward molecular definitions in diagnostic neuro-oncology. J Neuropathol Exp Neurol 62: 111–126

    Article  CAS  Google Scholar 

  68. Wolter M et al. (2001) Oligodendroglial tumors frequently demonstrate hypermethylation of the CDKN2A (MTS1, p16INK4a), p14ARF, and CDKN2B (MTS2, p15INK4b) tumor suppressor genes. J Neuropathol Exp Neurol 60: 1170–1180

    Article  CAS  Google Scholar 

  69. Cully M et al. (2006) Beyond PTEN mutations: the PI3K pathway as an integrator of multiple inputs during tumorigenesis. Nat Rev Cancer 6: 184–192

    Article  CAS  Google Scholar 

  70. Shaw RJ and Cantley LC (2006) Ras, PI(3) K and mTOR signalling controls tumour cell growth. Nature 441: 424–430

    Article  CAS  Google Scholar 

  71. Yohay KH (2006) The genetic and molecular pathogenesis of NF1 and NF2. Semin Pediatr Neurol 13: 21–26

    Article  Google Scholar 

  72. Galiatsatos P and Foulkes WD (2006) Familial adenomatous polyposis. Am J Gastroenterol 101: 385–398

    Article  Google Scholar 

  73. Hamilton SR et al. (1995) The molecular basis of Turcot's syndrome. N Engl J Med 332: 839–847

    Article  CAS  Google Scholar 

  74. Lackner C and Hoefler G (2005) Critical issues in the identification and management of patients with hereditary non-polyposis colorectal cancer. Eur J Gastroenterol Hepatol 17: 317–322

    Article  CAS  Google Scholar 

  75. Ess KC (2006) The neurobiology of tuberous sclerosis complex. Semin Pediatr Neurol 13: 37–42

    Article  Google Scholar 

  76. Inoki K et al. (2005) Dysregulation of the TSC-mTOR pathway in human disease. Nat Genet 37: 19–24

    Article  CAS  Google Scholar 

  77. Ino Y et al. (2000) Mutation analysis of the hCHK2 gene in primary human malignant gliomas. Neurogenetics 3: 45–46

    CAS  PubMed  Google Scholar 

  78. Varley J (2003) TP53, hChk2, and the Li-Fraumeni syndrome. Methods Mol Biol 222: 117–129

    CAS  PubMed  Google Scholar 

  79. Sansal I and Sellers WR (2004) The biology and clinical relevance of the PTEN tumor suppressor pathway. J Clin Oncol 22: 2954–2963

    Article  CAS  Google Scholar 

  80. Greene MH (1999) The genetics of hereditary melanoma and nevi. 1998 update. Cancer 86: 2464–2477

    Article  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Ethics declarations

Competing interests

The author declares no competing financial interests.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Collins, V. Mechanisms of Disease: genetic predictors of response to treatment in brain tumors. Nat Rev Clin Oncol 4, 362–374 (2007). https://doi.org/10.1038/ncponc0820

Download citation

  • Received:

  • Accepted:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncponc0820

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing