Skip to main content
Chinese Journal of Cancer logoLink to Chinese Journal of Cancer
. 2014 Jan;33(1):4–7. doi: 10.5732/cjc.013.10218

Molecular biology of high-grade gliomas: what should the clinician know?

Silvia Hofer 1, Elisabeth Rushing 2, Matthias Preusser 3, Christine Marosi 3
PMCID: PMC3905084  PMID: 24325789

Abstract

The current World Health Organization classification system of primary brain tumors is solely based on morphologic criteria. However, there is accumulating evidence that tumors with similar histology have distinct molecular signatures that significantly impact treatment response and survival. Recent practice-changing clinical trials have defined a role for routine assessment of O-6-methylguanine-DNA methyltransferase (MGMT) promoter methylation in glioblastoma patients, especially in the elderly, and 1p and 19q codeletions in patients with anaplastic glial tumors. Recently discovered molecular alterations including mutations in IDH-1/2, epidermal growth factor receptor (EGFR), and BRAF also have the potential to become targets for future drug development. This article aims to summarize current knowledge on the molecular biology of high-grade gliomas relevant to daily practice.

Keywords: High-grade glioma, molecular biology, IDH-1/2, MGMT, 1p/19q, EGFRvIII, diagnosis, prognosis, prediction


Gliomas account for approximately 70% of primary brain tumors in adults. The yearly incidence for Caucasians and Asians is about 6 cases per 100,000. Risk factors for gliomas are largely unknown, except for hereditary syndromes such as neurofibromatosis, tuberous sclerosis, Li Fraumeni syndrome, Turcot syndrome, and Cowden syndrome, as well as ionizing radiation to the head. Whether radiofrequency electromagnetic fields emitted by mobile phones induce gliomas remains unclear. Both inherited disorders and irradiation are rare occurrences, accounting for less than 10% of all gliomas and suggesting that complex genetic abnormalities combined with unknown environmental factors predispose individuals to glioma development.

For the last decades, the World Health Organization (WHO) histomorphologic classification of brain tumors together with clinical prognostic factors has guided clinicians in treating patients with high-grade gliomas. Tumor markers have not been readily available and their impact on decision making has not been supported by clinical trials. Patient- and tumor-related prognostic factors are still keys in decision making despite enormous progress in understanding the molecular biology of gliomas. Favorable clinical prognostic factors include young age, macroscopically complete tumor resection, and good Karnofsky performance status. Recursive partitioning analysis of large prospective trials refined clinical prognostic classes in the 1990s, which is still valid today[1],[2]. In one of the largest cohorts of Chinese glioma patients (n=1,235), the clinical characteristics and prognostic factors of patients with WHO grade II–IV glioma were similar to those of the Caucasian population[3].

Gliomas are classified using histomorphologic criteria and are designated as WHO grade I through IV according to their degree of malignancy[4]. WHO grade III and IV tumors are commonly lumped together as high-grade gliomas and comprise about 75% of all gliomas.

The WHO classification is based on subjective criteria and is imperfect in predicting patient outcome. Tumors may appear virtually identical by histology, yet still have very different outcomes. This is due, in part, to marked interobserver variability in making a diagnosis. Another contributing factor is whether the surgical specimen is representative of the overall lesion.

Progress in molecular techniques has allowed the identification of a number of markers and genetic profiles that characterize gliomas beyond their histologic criteria. So far, most have not had the awaited clinical impact, as data are not yet robust enough for clinical decision making.

A few molecular markers, however, have been introduced into the clinic in recent years and have been proven useful for identifying glioma subtypes (diagnosis), as well as guiding clinicians as to the course of the disease (prognostication) and on the choice of treatment (prediction). This notably holds true for patients with WHO grade III astrocytic and oligodendroglial gliomas, which may be difficult to distinguish on morphological criteria alone.

In 2013, three molecular markers were considered useful tools for the management of high-grade gliomas: 1p/19q chromosomal codeletion, O-6-methylguanine-DNA methyltransferase (MGMT) promoter methylation, and isocitrate dehydrogenase (IDH) 1 and 2 mutations. An additional biomarker, namely a specific mutation of the epidermal growth factor receptor (EGFR) variant III (EGFRvIII), serves as a potential target for yet to be proven experimental therapies (Tables 1 and 2).

Table 1. Role of glioma markers in clinical practice.

Marker Diagnostic Prognostic Predictive
1p/19q Oligodendroglial tumors1 WHO II-III WHO III2
IDH WHO > I; 2° GBM; exclusive for some glioma entities3 WHO II-IV No predictive role
MGMT No diagnostic role WHO III-IV WHO IV; alkylating agents, especially in elderly
EGFRvIII 1° GBM4 Not clearly defined WHO IV; vaccine or targeted therapies (experimental)

1Almost all oligodendroglial tumors have loss of heterozygosity 1p/19q. 2Predicitve for the treatment with radiotherapy and/or alkylating agents. 3Ependymoma and pilocytic astrocytoma do not have IDH mutation. 4EGFRvIII in ∼33% of primary GBM.

IDH, isocitrate dehydrogenase; MGMT, O-6-methylguanine-DNA methyltransferase; EGFRvIII, epidermal growth factor receptor variant III; GBM, gliblastoma multiforme. 2° GBM, secondary GBM progress from low-grade diffuse astrocytoma or anaplastic astrocytoma. 1° GBM, 90% of GBM develop rapidly de novo and are termed primary GBM.

Table 2. Overview of suitable methods for assessment of glioma markers in clinical practice.

Marker IHC FISH PCR/ Sequencing
1p/19q No Yes Yes
IDH/IDH Yes No Yes
MGMT No No Yes
EGFRvIII/EGFRvIII Yes No Yes

IDH, isocitrate dehydrogenase; MGMT, O-6-methylguanine methyltransferase; EGFR, epidermal growth factor receptor; IHC, immunohistochemistry; PCR, polymerase chain reaction; FISH, fluoresence in situ hybridization.

Molecular Marker, Clinically Useful for High-Grade Gliomas

1p/19q chromosomal codeletion

This codeletion is an unbalanced reciprocal translocation of 19q and 1p. Tumors that contain this translocation have been associated with an oligodendroglial phenotype, a better prognosis, and a better response to postoperative treatment, though the biological role of this marker remains unclear.

In 2012, follow-up results of more than 11–12 years in the Radiation Therapy Oncology Group (RTOG) 9402 and European Organization for Research and Treatment of Cancer (EORTC) 26951 trials demonstrated that an overall survival benefit from the addition of chemotherapy to radiotherapy was confined to patients with anaplastic oligodendroglial tumors with (versus without) 1p/19q co-deletion[5],[6]. The complete 1p/19q codeletion must be distinguished from partial 1p or 19q loss that, so far, lacks prognostic significance. Evidence suggests that 1p/19q codeletion is homogeneous within a tumor and does not change during disease evolution[7].

Currently, two international randomized trials are investigating sequence and the combination of radiotherapy and chemotherapy in WHO grade III tumors stratified according to 1p/19q status[8],[9].

MGMT promoter methylation

Methylation of the MGMT gene promoter results in epigenetic silencing of the methyltransferase, which loses its gene repair activity. MGMT methylation seems to be a prognostic factor prevalent throughout WHO grades II–IV gliomas, though with decreasing frequency as the malignant potential rises[10].

More than 15 years ago, reports indicated that high activity of the MGMT protein in glioma tissue was associated with resistance to alkylating agents, which, at that time, were largely nitrosoureas. In 2000, methylation of the promoter region of the MGMT gene was linked to improved outcomes. In 2005, MGMT promoter methylation assessed by a methylation-specific polymerase chain reaction was able to predict benefit from the addition of temozolomide (TMZ) chemotherapy to radiotherapy in the treatment of newly diagnosed glioblastoma multiforme (GBM)[11]. However, standardizing the MGMT assay for widespread clinical use was challenging, and treatment decisions continued to be performed without knowledge of the MGMT status. In 2012, two randomized trials performed in the growing population of elderly GBM patients demonstrated consistently that a methylated MGMT promoter is a powerful predictive biomarker for benefit from TMZ alone. In the German NOA-08 trial, patients older than 65 years were treated with either standard 6-week, fractionated (1.8–2.0 Gy) radiotherapy or dose-dense TMZ chemotherapy (week on/week off). Patients with tumors exhibiting methylated MGMT fared better if they were treated with TMZ alone than those treated with radiotherapy alone[12]. Similarly, the Nordic trial found standard-dose TMZ (5 out of 28 days) to be superior to radiotherapy in patients older than 60 years with methylated MGMT promoter[13]. Thus, at least in the elderly population, MGMT testing should become a standard procedure for decision making (chemotherapy vs. radiotherapy), though the test is not yet widely available.

Whether patients with MGMT promoter methylation of other age groups or other WHO grades should be treated with TMZ alone rather than chemoradiotherapy is an important question for future studies.

IDH1 and IDH2 mutations

Point mutations in the IDH1 and IDH2 genes, originally discovered in 2008, occur in the vast majority of low-grade gliomas (>80%) and secondary high-grade gliomas. The frequency of these mutations does not change during the progression from WHO grade II to WHO grades III or IV (so-called secondary GBM). Evidence has accumulated that primary and secondary GBM develop through different genetic pathways, though they remain largely histomorphologically indistinguishable at diagnosis.

IDH1/2 mutations, which occur early in gliomagenesis, change the function of the enzymes, causing them to produce 2-hydroxyglutarate, a possible oncometabolite, instead of α-ketoglutarate. The mutations are able to drive increased methylation in gliomas. Gliomas with a mutated IDH1 or, less frequently, mutated IDH2 are associated with better prognosis compared to their wild-type counterparts[14]. As with loss of heterozygosity 1p/19q, a given IDH status seems to be homogeneous within a tumor and does not change during disease evolution. Mutated IDH can easily be detected by immunohistochemistry and potentially even non-invasively by magnetic resonance spectroscopy. Non-tumoral glial cells (i.e. those involved in gliosis) never express mutated IDH, a fact that can be used to separate reactive gliosis from gliomas. Pilocytic astrocytoma (WHO grade I), ependymoma, and primary GBM (but not secondary GBM) do not harbor IDH mutations[15]. Of note, IDH mutations are not glioma-specific alterations. Furthermore, there is currently no drug that targets mutated IDH, although this remains an area of active research.

EGFRvIII

A tumor-specific mutant of the EGFR, EFGR variant III (EGFRvIII), causes constitutive activation of the receptor's tyrosine kinase activity and is frequently expressed in primary GBM (∼33%). This mutation confers enhanced tumorigenic behavior, at least in preclinical experiments[16]. Because it is localized solely on tumor tissue, EGFRvIII presents an ideal target for immunotherapy, reducing the risk of autoimmune toxicity. Immunohistochemical testing for EGFRvIII may be implemented if randomized trials demonstrate activity of EGFRvIII-targeted vaccination[17],[18].

Interrelations of various molecular markers

Among low-grade and anaplastic gliomas, nearly all with 1p/19q codeletion also harbor IDH1/2 mutations. However, some genetic markers, such as EGFR and IDH1, EGFR and TP53, TP53 and 1p/19q, are mutually exclusive. Molecularly, IDH1 and IDH2 mutations are heterozygous, affect only a single codon, and rarely occur together. Although TP53 mutations and 1p/19q codeletions are mutually exclusive, IDH1 mutations are common in both of these genotypes[19],[20].

Conclusions

The most recent clinical data from randomized phase III trials call for routine testing of 1p/19q for patients with WHO grade III gliomas and for assessing the MGMT methylation status, especially in elderly GBM patients too frail to undergo postoperative concomitant radiochemotherapy followed by chemotherapy—the standard treatment for GBM. Molecular marker determination, however, is technically demanding and requires reproducible and validated test procedures. This holds especially true for MGMT testing, where results sometimes may fall into a “gray zone.”

Outlook

BRAF mutations have been found in a fraction of high-grade glioma patients (e.g. epitheloid GBM) and may present a druggable treatment target for specific inhibitors such as vemurafenib or dabrafenib[21], [22]. Another focus of interest is immunosuppressive molecules (e.g., B7H1 and B7H4); further research is warranted to define the role of immunomodulatory drugs in high-grade glioma[23]. Moreover, we still lack biomarkers with predictive properties to select anti-angiogenic agents for treating gliomas.

Circulating microRNA (miRNA), small non-coding regulatory RNAs that modulate the expression of specific target genes, might be relevant in the future for diagnosis, prognosis, and therapy of gliomas[24]. For example, evaluating the circulating DNA of EGFRvIII in plasma may represent a strategy to screen patients for an anti-EGFRvIII therapy and monitor response to treatment[25].

The availability of high-throughput methods will most likely enrich the histomorphological WHO classification with a comprehensive molecular characterization of gliomas.

References

  • 1.Paravati AJ, Heron DE, Landsittel D, et al. Radiotherapy and temozolomide for newly diagnosed glioblastoma and anaplastic astrocytoma: validation of Radiation Therapy Oncology Group-Recursive Partitioning Analysis in the IMRT and temozolomide era. J Neurooncol. 2011;104:339–349. doi: 10.1007/s11060-010-0499-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Mirimanoff RO, Gorlia T, Mason W, et al. Radiotherapy and temozolomide for newly diagnosed glioblastoma: recursive partitioning analysis of the EORTC 26981/22981-NCIC CE3 phase III randomized trial. J Clin Oncol. 2006;24:2563–2569. doi: 10.1200/JCO.2005.04.5963. [DOI] [PubMed] [Google Scholar]
  • 3.Yang P, Wang Y, Peng X, et al. Management and survival rates in patients with glioma in China (2004-2010): a retrospective study from a single-institution. J Neurooncol. 2013;113:259–266. doi: 10.1007/s11060-013-1103-9. [DOI] [PubMed] [Google Scholar]
  • 4.Louis D, Ohgaki H, Wiestler O, et al. The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol. 2007;114:97–109. doi: 10.1007/s00401-007-0243-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Cairncross G, Wang M, Shaw E, et al. Phase III trial of chemoradiotherapy for anaplastic oligodendroglioma: long-term results of RTOG 9402. J Clin Oncol. 2013;31:337–343. doi: 10.1200/JCO.2012.43.2674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.van den Bent MJ, Brandes AA, Taphoorn MJ, 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. 2013;31:344–350. doi: 10.1200/JCO.2012.43.2229. [DOI] [PubMed] [Google Scholar]
  • 7.Weller M, Stupp R, Hegi 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. 2012;14(Suppl. 4):iv100–108. doi: 10.1093/neuonc/nos206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Radiation therapy with or without temozolomide in treating patients with anaplastic glioma. ClinicalTrials.gov Identifier: NCT00626990. Verified May 2011 by National Cancer Institute (NCI). ClinicalTrials.gov processed this record on December 01, 2013. http://clinicaltrials.gov/ct2/show/NCT00626990?term=NCT00626990&rank=1.
  • 9.Radiation therapy with concomitant and adjuvant temozolomide or radiation therapy with adjuvant PCV or temozolomide alone in treating patients with anaplastic glioma. ClinicalTrials.gov identifier: NCT00887146. Verified November 2013 by Alliance for Clinical Trials in Oncology. ClinicalTrials.gov processed this record on December 01, 2013. http://clinicaltrials.gov/ct2/show/NCT00887146?term=NCT00887146.&rank=1.
  • 10.Hofer S, Lassman AB. Molecular markers in gliomas: impact for the clinician. Targ Oncol. 2010;5:201–210. doi: 10.1007/s11523-010-0157-2. [DOI] [PubMed] [Google Scholar]
  • 11.Hegi ME, Diserens AC, Gorlia T, et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med. 2005;352:997–1003. doi: 10.1056/NEJMoa043331. [DOI] [PubMed] [Google Scholar]
  • 12.Wick W, Platten M, Meisner C, et al. Temozolomide chemotherapy alone versus radiotherapy alone for malignant astrocytoma in the elderly: the NOA-08 randomised, phase 3 trial. Lancet Oncol. 2012;13:707–715. doi: 10.1016/S1470-2045(12)70164-X. [DOI] [PubMed] [Google Scholar]
  • 13.Malmström A, Grønberg BH, Marosi C, 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;13:916–926. doi: 10.1016/S1470-2045(12)70265-6. [DOI] [PubMed] [Google Scholar]
  • 14.Zou P, Xu H, Chen P, et al. IDH1/IDH2 mutations define the prognosis and molecular profiles of patients with gliomas: a meta-analysis. PLoS One. 2013;8:e68782. doi: 10.1371/journal.pone.0068782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Capper D, Weissert S, Balss J, et al. Characterization of R132H mutation-specific IDH1 antibody binding in brain tumors. Brain Pathol. 2010;20:245–254. doi: 10.1111/j.1750-3639.2009.00352.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Gan HK, Cvrljevic AN, Johns TG. The epidermal growth factor receptor variant III (EGFRvIII): where wild things are altered. FEBS J. 2013;280:5350–5370. doi: 10.1111/febs.12393. [DOI] [PubMed] [Google Scholar]
  • 17.Sampson JH, Heimberger AB, Archer GE, 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. 2010;28:4722–4729. doi: 10.1200/JCO.2010.28.6963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Phase III study of rindopepimut/GM-CSF in patients with newly diagnosed glioblastoma. ClinicalTrials.gov identifier: NCT01480479. Verified November 2013 by Celldex Therapeutics. ClinicalTrials.gov processed this record on December 01, 2013. http://clinicaltrials.gov/ct2/show/NCT01480479?term=NCT01480479+.&rank=1.
  • 19.Kloosterhof NK, Bralten LB, Dubbink HJ, et al. Isocitrate dehydrogenase-1 mutations: a fundamentally new understanding of diffuse glioma? Lancet Oncol. 2011;12:83–91. doi: 10.1016/S1470-2045(10)70053-X. [DOI] [PubMed] [Google Scholar]
  • 20.Malzkorn B, Wolter M, Riemenschneider MJ, et al. Unraveling the glioma epigenome: from molecular mechanisms to novel biomarkers and therapeutic targets. Brain Pathol. 2011;21:619–632. doi: 10.1111/j.1750-3639.2011.00536.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Kleinschmidt-DeMasters BK, Aisner DL, Birks DK, et al. Epithelioid GBMs show a high percentage of BRAF V600E mutation. Am J Surg Pathol. 2013;37:685–698. doi: 10.1097/PAS.0b013e31827f9c5e. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Nicolaides TP, Li H, Solomon DA, et al. Targeted therapy for BRAFV600E malignant astrocytoma. Clin Cancer Res. 2011;17:7595–7604. doi: 10.1158/1078-0432.CCR-11-1456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Bloch O, Crane CA, Kaur R, et al. Gliomas promote immuno-suppression through induction of B7-H1 expression in tumor-associated macrophages. Clin Cancer Res. 2013;19:3165–3175. doi: 10.1158/1078-0432.CCR-12-3314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Illhan-Mutlu A, Wagner L, Preusser M. Circulating biomarkers of CNS tumors: an update. Biomark Med. 2013;7:267–285. doi: 10.2217/bmm.13.12. [DOI] [PubMed] [Google Scholar]
  • 25.Salkeni MA, Zarzour A, Ansay TY, et al. Detection of EGFRvIII mutant DNA in the peripheral blood of brain tumor patients. J Neurooncol. 2013;115:27–35. doi: 10.1007/s11060-013-1209-0. [DOI] [PubMed] [Google Scholar]

Articles from Chinese Journal of Cancer are provided here courtesy of BMC

RESOURCES