418
Views
102
CrossRef citations to date
0
Altmetric
Review

Personalized treatment strategies in glioblastoma: MGMT promoter methylation status

, &
Pages 1363-1372 | Published online: 27 Sep 2013

Abstract

The identification of molecular genetic biomarkers considerably increased our current understanding of glioma genesis, prognostic evaluation, and treatment planning. In glioblastoma, the most malignant intrinsic brain tumor entity in adults, the promoter methylation status of the gene encoding for the repair enzyme O6-methylguanine-DNA methyltransferase (MGMT) indicates increased efficacy of current standard of care, which is concomitant and adjuvant chemoradiotherapy with the alkylating agent temozolomide. In the elderly, MGMT promoter methylation status has recently been introduced to be a predictive biomarker that can be used for stratification of treatment regimes. This review gives a short summery of epidemiological, clinical, diagnostic, and treatment aspects of patients who are currently diagnosed with glioblastoma. The most important molecular genetic markers and epigenetic alterations in glioblastoma are summarized. Special focus is given to the physiological function of DNA methylation–in particular, of the MGMT gene promoter, its clinical relevance, technical aspects of status assessment, its correlation with MGMT mRNA and protein expressions, and its place within the management cascade of glioblastoma patients.

Glioblastoma

Diffuse gliomas are heterogeneous neoplasms that account for half of all intrinsic brain tumors. Histological grading of these tumors according to the classification system of the World Health Organization (WHO) provides a basis for defining groups of patients for clinical assessment, but also predicts the clinical behavior of the respective neoplasm with direct impact on the applied treatment regimes. WHO grade IV gliomas are characterized by high cellularity, cellular pleomorphism, nuclear atypia, brisk mitotic activity, microvascular proliferation, and necrosis.Citation1 They account for 50%–60% of all astrocytic gliomas, with an increasing incidence of currently three to five new cases per 100,000 patient-years.Citation2 The cause for glioblastoma development is not clear. Some authors speculate that cytomegalovirus infection may drive the oncogenic process by modulating growth factor and receptor expressions in gliomas.Citation3 Clinically, these tumors are rapidly progressive and ultimately fatal, with a median survival of only 15 months in trial populations.Citation4 Primary glioblastomas develop with only a short clinical history and without evidence of a previous lesion of lower malignancy. They account for the vast majority of glioblastomas in older patients. Morphologically, they cannot be distinguished from about 5%–10% of glioblastomas that develop in younger patients below the age of 50 years by progression from a pre-existing lower-grade glioma.Citation5,Citation6 These secondary glioblastomas differ significantly in their genetic profiles from primary glioblastomas.Citation7,Citation8

The diagnostic challenge

Routine diagnosis critically relies on magnetic resonance imaging (MRI) that frequently exhibits contrast enhancement of the lesion in T1-weighted sequences accompanied by perifocal edema as being depicted on T2-/fluid-attenuated inversion recovery-weighted images. Lack of focal contrast enhancement, however, does not exclude grade IV histology.Citation9,Citation10 The vast majority of glioblastomas develop in the cerebral hemispheres. However, the highly infiltrative growing pattern frequently causes tumor spread into the basal ganglia or even the contralateral hemisphere, with unfavorable effects on patients’ outcome. Glioblastomas with primary location in the cerebellum, brain stem, or myelon are rare. Metabolic imaging such as positron emissions tomography (PET) utilizing amino acid tracers (eg, O-[2-{18F}fluoroethyl]-1-tyrosine) is increasingly used to assess differential diagnosis,Citation11,Citation12 biological tumor volume, and intratumoral heterogeneity,Citation10 and to monitor therapy.Citation13Citation15 Mainly due to its limited availability and associated costs, metabolic imaging has not yet been routinely implemented into a current standard of care algorithm for glioblastoma patients. Ultimately, tissue samples are needed for definite histological diagnosis. Neuropathological evaluation of gliomas, however, suffers from significant interobserver variability, particularly in the distinction of grade III and grade IV histology and the determination of an oligodendroglial cell component.Citation16

Current treatment concepts

More recently, some progress has been achieved in the treatment of glioblastoma patients. A companion prospective multicenter study performed by the European Organization for Research and Treatment of Cancer (EORTC) and the National Cancer Institute of Canada (NCIC) has shown that the addition of the alkylating agent temozolomide to radiotherapy improved 2-year survival of patients with newly diagnosed malignant glioma (mainly glioblastoma) from 11.0% to 27.3%, 3-year survival from 4.4% to 16.0%, and 5-year survival from 1.9% to 9.8%, respectively.Citation17,Citation18 Temozolomide is an orally administered chemotherapy with limited side effects and a good penetration of the blood–brain barrier. It prevents replication by alkyl group-mediated crosslinking of DNA.Citation19 Currently, radiotherapy with concomitant and adjuvant temozolomide is the gold standard for patients with newly diagnosed glioblastoma.Citation4 Most recent results from the Phase III study AVAglio, evaluating the addition of the antiangiogenic agent bevacizumab to combined chemoradiotherapy with temozolomide, have indicated favorable effects on progression-free survival (median 10.6 months versus 6.2 months) and duration of good clinical performance (Karnofsky performance status ≥70; 9 months versus 6 months).Citation20,Citation21 However, no effect on overall survival was observed. In the elderly, the best treatment strategy is highly influenced by the patient and tumor-related factors and ranges from combined chemoradiotherapy with temozolomide for highly selected patients with good clinical performance status to either radiotherapy or temozolomide treatment alone, and supportive care only.Citation22Citation28

Open tumor resection also represents one of the mainstays in glioblastoma treatment and is usually considered the first step within the management algorithm.Citation29,Citation30 The highly infiltrative growing pattern of glioblastomas into surrounding brain tissues explains that curative resection generally cannot be achieved. However, there is good evidence that overall survival is improved if early postoperative MRI depicts that no residual contrast-enhancing tumor mass has been left in situ (“gross total resection”).Citation31Citation35 In a community setting, complete resection is achieved in about 40%–45% of patients, a similar proportion receives incomplete resection, and about 10%–20% are diagnosed by biopsy only.Citation21 The number of patients without residual tumors can be significantly increased if microsurgical tumor removal is performed with the aid of 5-aminolevulinic acid and intraoperative MRI.Citation31,Citation36 The prognostic impact of incomplete resection is poorly defined.Citation30 Favorable impacts on survival rates have been suggested if at least 78% of contrast-enhancing tumor volume has been successfully removed.Citation37 However, this and other studies did not compare the impact of incomplete resection with biopsy only, and there is some evidence that biopsy only with early transferral to chemoradiotherapy might be appropriate in selected patients in whom even partial resection bears a considerable risk of surgery-related sequalae.Citation38 In patients suffering from clinical deterioration due to a large space-occupying tumor burden, decompressive surgery should be attempted.Citation30

Although the use of temozolomide has improved outcome considerably, almost all patients suffer from recurrent disease. Local tumor progression (within 3 cm from the margin) is the predominant pattern (93.5%) of treatment failure.Citation39Citation42 In general, tumor recurrence is associated with poor outcome as treatment options are limited.Citation43 Approximately one-quarter of patients with glioblastoma develop a type of recurrence that allows for local treatment including repeated surgical resection.Citation44 Although reoperation might improve post-recurrence survival in selected cases, its value must be counterbalanced by the risk of surgery-related morbidity and mortality.Citation45,Citation46 Other local treatment options include stereotactic radiosurgery for small lesions, hypofractionated radiotherapy protocols, even for larger recurrent tumors, and any combination of local irradiation and chemotherapy.Citation43 Systemic therapy is generally considered essential for recurrent tumors. Various antineoplastic agents have been tested. Efficacy, however, is generally hampered by the broad chemoresistance of glioblastoma cells, in parallel with the limited bioavailability of most drugs inside the central nervous system. The most important approaches include rechallenge regimes with dose-intensified temozolomide and non-temozolomide-containing regimes.Citation43,Citation47 Since 2009 the monoclonal antibody to vascular endothelial growth factor bevacizumab has gained US Food and Drug Administration approval for recurrent glioblastoma based on response rate, with favorable effects relative to historical controls from noncomparative Phase II trials.Citation48,Citation49 The rejection in Europe, however, was based on the absence of a randomized trial with a bevacizumab-free control arm.Citation47 Other targeted approaches address the epidermal growth factor receptor, mammalian target of rapamycin, histone deacetylase, and many other structures in recurrent glioblastoma. However, postrecurrence survival rarely exceeds 6–9 months in most clinical study populations.

The prognostic network of glioblastoma patients

In glioblastoma, the patient’s prognosis and response to therapy are highly influenced by clinical and molecular genetic factors that are increasingly used for prognostic profiling and individualized risk-adapted treatment considerations.Citation50 Classical phenotypical traits correlating with favorable survival are younger age, good performance status, and noneloquent circumscribed tumor formations.Citation8,Citation35,Citation51Citation53 However, these conventional prognostic factors do not necessarily account for the highly variable clinical courses of glioblastoma patients.

Molecular biomarkers have improved our understanding of glioma development, are increasingly exploited for glioblastoma subclassification, and have gained prognostic/predictive relevance. From a clinical perspective, a biomarker should be highly sensitive and specific in providing information relevant for diagnosis, prognosis, or therapy of a disease. So far, only a few biomarkers have gained clinical relevance in glioblastoma patients.

Genetic markers relevant for glioma patients

Mutations in the gene encoding for the Krebs cycle enzyme, isocitrate dehydrogenase (IDH), have been identified as one of the earliest molecular events in the pathway of glioma genesis. They are frequent in grade II/III astrocytomas, oligoastrocytomas, and oligodendrogliomas, and can be used to differentiate primary and secondary glioblastomas.Citation6,Citation54,Citation55 These findings suggest that histologically differently appearing tumor subclasses might share common precursor cells, and that histologically indistinguishable tumors (eg, primary versus secondary glioblastomas) could be separated into biologically distinct subclasses. IDH point mutations are predominantly found at the arginine 132 (IDH1), but can also occur at arginine 172 (IDH2). These mutations cause alterations in the active site of the IDH enzyme and result in increased production of 2-hydroxyglutarate, which is associated with an increased risk of cancer and glioma progression.Citation56 IDH1 mutations are associated with favorable outcome in WHO grade III and IV malignant gliomas.Citation57,Citation58 Notably, patients with IDH1 wild-type anaplastic astrocytoma WHO grade III experience even worse outcome than those with glioblastomas WHO grade IV harboring an IDH1 mutation.Citation59 The prognostic impact of the IDH1 status in WHO grade II astrocytomas is more controversial but might be associated with unfavorable outcome until tumor progression occurs.Citation60 TP53 mutations are frequent in 70% of secondary glioblastomas and seldom in primary glioblastomas.Citation61 TP53 modulates cell-cycle control in tumor cells and has been associated with unfavorable outcome in grade II gliomas.Citation62,Citation63 Loss of heterozygosity on chromosome arms 1p and 19q (LOH 1p/19q) is frequent in oligodendroglial tumors, and has been associated with increased chemosensitivity in grade III gliomas.Citation64,Citation65 Whether 1p- and 19q-codeleted tumors have a less aggressive natural course than noncodeleted tumor has not been clarified so far. In glioblastoma, LOH 1p/19q is rare (~5%).Citation50 However, an oligodendroglioma-like component (GBM-O) is seen in 15% of all glioblastomas and has been associated with improved clinical outcome.Citation66 Whether a favorable outcome in these patients is also influenced by LOH 1p/19q has not been unequivocally clarified.Citation67,Citation68 Other molecular genetic markers, such as PTEN mutation, epidermal growth factor receptor variant III and CDK 4 amplifications, and CDKN2 A homozygous deletion, can be used to separate primary from secondary glioblastomas, indicating different biology and cells of origin.Citation8,Citation69 However, these markers have not yet gained prognostic relevance in clinical trials.Citation70

Besides a mutated IDH1 status and LOH 1p/19q, methylation of the promoter region of the O6-methylguanine-DNA methyltransferase (MGMT) gene has been correlated with favorable outcome in malignant glioma patients.

Physiological function of DNA methylation and its role in glioma formation

One of the first and most important epigenetic modifications studied in humans is DNA methylation, which describes the covalent addition of a methyl group preferentially at the 5′-position of a cytosine or guanine nucleotide. These CpG dinucleotides tend to cluster to so-called CpG islands, being located in the promoter regions of more than half of all human genes, or to CpG island shores, which are regions of lower CpG density that lie in close proximity to CpG islands.Citation71,Citation72 DNA methylation is mediated by the DNA methyltransferase family of enzymes. These enzymes catalyze the transfer of a methyl group from S-adenosyl methionine to DNA and are responsible for maintaining the methylation pattern (eg, during DNA replication).Citation73,Citation74 Generally, CpG methylation is closely associated with transcriptional inactivation. Less frequently, when occurring at gene bodies, it can cause transcriptional activation. Genome-wide methylation patterns have been associated with tumor initiation and progression in several cancers, including colon, breast, and lung.Citation75,Citation76

DNA methylation in glioblastoma

The genome of glioblastoma cells shows broad hypomethylation with specific areas of hypermethylation.Citation77Citation80 This characteristic pattern has been associated with increased genetic instability, silencing of tumor suppressors such as TP53 and PTEN, and activation of oncogenes. Hypermethylation mostly occurs at the promoter CpG island of genes that are associated with tumor suppression,Citation81,Citation82 DNA repair,Citation83 cell-cycle regulation,Citation84 apoptosis,Citation85,Citation86 invasion,Citation87,Citation88 and migration.Citation89 Interestingly, the methylation patterns differ between gliomas of WHO grade II–IV.Citation90

The Cancer Genome Atlas project has identified a glioma CpG island methylation phenotype that correlated with younger age, a proneural gene expression profile,Citation91 and longer overall survival in glioblastoma patients.Citation92 Moreover, a high frequency of IDH1 mutations indicates a link between metabolic alterations and epigenetic modification in these tumors.Citation93,Citation94 Increased production of the metabolite 2-hydroxyglutarate interferes with α-ketoglutarate-dependent enzyme, including histone demethylases and the TET family of 5-methylcytosine hydroxylases,Citation56,Citation95 which causes genome-wide histone modifications,Citation96 and alterations of DNA methylation statusCitation97 in malignant glioma cells.Citation98,Citation99

MGMT promoter methylation in glioblastoma

In glioblastoma, promoter methylation of the gene encoding for MGMT is undoubtedly the genetic fingerprint with highest impact on clinical practice. The MGMT gene is located at chromosome 10q26 and codes for a ubiquitously expressed suicide DNA repair enzyme that removes alkyl adducts from the O6-position of guanine.Citation100 As O6-alkylated guanine leads to double-strand breaks and base mispairing, thereby inducing apoptosis and cell death, MGMT protects normal cells from carcinogens. Unfortunately, it also protects tumor cells from normally lethal effects of chemotherapy with alkylating agents such as temozolomide.Citation100 MGMT is consumed when counteracting TMZ-induced DNA damage, and it has been predicted that the intracellular level of MGMT correlates with chemoresistance.Citation101 However, analysis of MGMT protein expression in glioblastoma tissue by immunohistochemistry failed to correlate with survival under chemoradiotherapy.Citation102

Clinical relevance

Methylation of the MGMT promoter is found in 35%–45% of malignant gliomas (WHO grades III and IV) and in about 80% of WHO grade II gliomas.Citation60,Citation103 MGMT methylated and unmethylated glioblastomas seem to differ in primary location,Citation104 pattern of contrast enhancementCitation105 and the apparent diffusion coefficient in MRI analysis,Citation106 the incidence of pseudoprogression,Citation107,Citation108 and pattern of recurrence as determined by 18FET-PET imaging.Citation109 However, none of these methods sufficiently allows for noninvasive determination of MGMT promoter methylation status for the individual patient.Citation104,Citation105,Citation110

The methylation status of the MGMT promoter has been identified as a strong and independent predictive factor of favorable survival in glioblastoma patients undergoing chemotherapy with alkylating agents.Citation103,Citation111Citation113 The median survival for patients with a methylated MGMT promoter was 21.7 months compared with 12.7 months for patients without. Accordingly, a high frequency of MGMT promoter methylation was noted in long-term survivors of glioblastoma who received repetitive alkylating chemotherapy during the course of the disease.Citation23,Citation114 Generally, carriers of the methylated form of the MGMT promoter respond substantially better to therapy with temozolomide as compared with those with an unmethylated MGMT promoter.Citation17,Citation58 This predominant influence of MGMT promoter methylation is independent of surgical treatment. Moreover, a methylated MGMT promoter status seems to stratify outcome even in the recurrent disease.Citation115 Postrecurrence survival was better in those MGMT methylated patients who receive alkylating therapy for recurrent disease.Citation115 However, a methylated MGMT promoter status does not influence outcome in solely irradiated glioblastoma patients (without alkylating therapy), whereas in grade III gliomas favorable outcome in methylated patients seems to be irrespective of the applied treatment regime.Citation58,Citation116 It is currently unclear, however, whether this observation indicates a true prognostic value for MGMT methylation in all grade III gliomas.Citation117 In grade II gliomas, MGMT promoter methylation shows a Janus head-like correlation with shortened progression-free survival but prolonged overall survival under radio and/or chemotherapy.Citation61

Determination of the MGMT promoter methylation status

Currently, there is no consensus about the most suitable technique for determination of the MGMT promoter methylation status. This concerns both the tissue sampling technique as well as the molecular assays currently used in the clinical setting.Citation30 MGMT promoter methylation status can be efficiently determined even from small (1 mm³) formalin-fixed, paraffin-embedded tissue samples obtained from stereotactic biopsy procedures.Citation23 Analysis of MGMT promoter methylation status of multiple tissue samples harvested from serial biopsy procedures throughout entire tumor volumes has proven that this molecular fingerprint is homogeneous in glioblastoma formations.Citation23 Notably, only vital (non-necrotic) tumor specimens must be used for subsequent analysis in order to avoid false-negative results.Citation23,Citation118 Analyses from paired tumor samples harvested from primary and recurrent glioblastomas revealed that MGMT promoter methylation status is unchanged during the course of the disease.Citation115

In respect of the currently available methods for MGMT testing, nonquantitative methylation-specific polymerase chain reaction (MSP)Citation119 seems to be the most appropriate one in a clinical setting.Citation120,Citation121 Commonly, two pairs of primers–each specific to either the methylated or the unmethylated MGMT promoter region–are used for MSP as originally described by Esteller et al.Citation122 However, discrepancies exist between sequencing analysesCitation123 and results from MSP. Tumors classified as “unmethylated” by MSP could turn out to be “methylated” or at least “partially methylated” using sequencing analysis.Citation124 A recent study systematically analyzed the impact of specific CpG sites within the MGMT promoter on the transcriptional regulation of MGMT using a luciferase reporter assay.Citation125 This study revealed that MSP is located within the optimal region for MGMT testing. However, the study also showed that substitution of a single CpG outside the MSP region almost completely inhibited the promoter activity. Also, there was a high variability regarding the methylated positions. Hence, promoter methylation-mediated gene silencing seems to be strongly dependent on the location of the methylated CpGs and the extent of the overall CPG island methylation, which considerably complicates classification.Citation117,Citation126 Given these facts, determination of all individual CpG sites of the recently identified optimal region might predict the transcriptional activity and sensitivity to alkylating substances more confidently.Citation117 More clinical data are needed to support this assumption.

MGMT determination by immunohistochemistry has also been suggested. However, this method lacks standardization, reproducibility, and correlation with outcome.

MGMT promoter methylation and RNA expression

Differential regulation of MGMT mRNA expression might also explain why MGMT promoter methylation is not unequivocally linked to a favorable treatment response. It has been shown that not all patients with a methylated promoter reveal similar response to temozolomide treatment and that a considerable number of unmethylated tumors experienced a surprisingly favorable course of the disease.Citation127 These observations could be explained by discordant correlations between MGMT promoter methylation (as being determined by both MSP and sequencing analysis) and MGMT mRNA expression pattern. High (low) mRNA MGMT expression was detected in approximately 25% of glioblastomas despite a methylated (unmethylated) MGMT promoter. Notably, those patients with low transcriptional activity exhibited a better treatment response, which was independent of MGMT promoter methylation. These observations were confirmed by another study verifying low MGMT immunostaining in 23% of unmethylated patients and high MGMT expression in 8% of methylated patients.Citation128 The underlying mechanisms of discordance still remain unclear. It has been hypothesized that low MGMT expression levels combined with an unmethylated promoter might result from transcript destabilization and/or transcription-repressing factors, such as miRNA regulation or histone modifications.Citation127,Citation129 Additionally, variable outcomes in the unmethylated glioblastoma population could be mediated by further factors, such as a heterogeneous expression pattern of the DNA repair enzyme APNG, which confers resistance to temozolomide treatment.Citation130

MGMT for decision making in glioblastoma patients

MGMT promoter methylation status has been established as an important clinical biomarker in neuro-oncology. Accordingly, determination of MGMT promoter methylation status is of utmost interest for prognostication of adult patients suffering from newly diagnosed glioblastoma. This also includes those patients with nonresectable tumors that undergo biopsy only. However, with a lack of established alternative treatment options and in the absence of any clinical consequence of routine determination of the MGMT promoter, methylation status does not yet add to the management of glioblastoma patients outside clinical trials. In the absence of potent alternative drugs, temozolomide chemotherapy should not be withheld from unmethylated glioblastoma patients younger than 70 years of age in general practice. Moreover, discordant responses even within the subgroups of methylated and unmethylated patients indicate that treatment decision in respect of chemotherapy cannot be based on this biomarker alone.

The effects of MGMT promoter methylation in malignant gliomas seem to also be dependent on WHO grading. In anaplastic glioma, favorable progression-free survival in MGMT methylated patients was also seen under radiation therapy alone.Citation58 This difference might be explained by the high incidence of other favorable molecular markers in WHO grade III gliomas, such as IDH1 mutation, 1p/19q deletion, or yet to be identified novel aberrations.

MGMT in the elderly

In the older glioblastoma patient, MGMT promoter methylation status is on the verge of entering clinical decision making. Combined chemoradiotherapy comprising temozolomide might be too toxic for the elderly, with increased side effects.Citation22,Citation131,Citation132 For the elderly with malignant glioma, two recently published Phase III trials have evaluated the place of dose-dense/conventional temozolomide regimes alone as compared with conventional/hypofractionated radiotherapy.Citation25,Citation133 Overall survival in methylated patients was better if temozolomide treatment was applied, whereas in unmethylated patients radiotherapy alone was more effective. Thus, MGMT promoter methylation is an important biomarker for personalized treatment strategies in the elderly subpopulation.

MGMT and clinical trials

MGMT promoter methylation testing has recently been introduced as a marker for patient selection within clinical trials.Citation113,Citation134Citation137 Results from a Phase I/IIA trial for cilengitide have shown that treatment effects were better in MGMT promoter methylated patients.Citation138 Accordingly, the ongoing Cilengitide, Temozolomide, and Radiation Therapy in Treating Patients With Newly Diagnosed Glioblastoma and Methylated Gene Promoter Status (CENTRIC) study (NCT00689221) was one of the first prospective multicenter studies that evaluated alternative treatment (cilengitide, temozolomide, and radiation therapy) in MGMT methylated glioblastoma patients only. The Radiation Therapy Oncology Group (RTOG) 0825 trial also incorporated MGMT promoter methylation testing for primary glioblastoma patients who underwent bevacizumab administered with radiotherapy compared with conventional concurrent chemoradiotherapy (temozolomide).Citation139

Conclusion

There is an urgent need to identify biomarkers in malignant glioma patients in order to indicate patients at risk of tumor relapse, treatment failure, or adverse events, and to allow for prognostication and clinical decision making.Citation122 Currently, the methylation status of the MGMT gene promoter seems to separate different subtypes of malignant glioma patients and will certainly influence future studies in respect of stratification of patient groups who are more likely to respond to a certain therapy. The usefulness of MGMT testing in a routine clinical setting will be of fundamental relevance when distinct treatment strategies for methylated and unmethylated glioblastoma patients will be available.

Disclosure

The authors report no conflicts of interest in this work.

References

  • LouisDNOhgakiHWiestlerODThe 2007 WHO classification of tumours of the central nervous systemActa Neuropathol200711429710917618441
  • CrocettiETramaAStillerCEpidemiology of glial and non-glial brain tumours in EuropeEur J Cancer201248101532154222227039
  • SampsonJHMitchellDAIs cytomegalovirus a therapeutic target in glioblastoma?Clin Cancer Res201117144619462121632859
  • PreusserMde RibaupierreSWöhrerACurrent concepts and management of glioblastomaAnn Neurol201170192121786296
  • SchererHJA critical review: the pathology of cerebral gliomasJ Neurol Psychiatry19403214717721610973
  • NobusawaSWatanabeTKleihuesPOhgakiHIDH1 mutations as molecular signature and predictive factor of secondary glioblastomasClin Cancer Res200915196002600719755387
  • WatanabeKTachibanaOSataKYonekawaYKleihuesPOhgakiHOverexpression of the EGF receptor and p53 mutations are mutually exclusive in the evolution of primary and secondary glioblastomasBrain Pathol1996632172238864278
  • OhgakiHDessenPJourdeBGenetic pathways to glioblastoma: a population-based studyCancer Res200464196892689915466178
  • BarkerFG2ndChangSMHuhnSLAge and the risk of anaplasia in magnetic resonance-nonenhancing supratentorial cerebral tumorsCancer19978059369419307194
  • KunzMThonNEigenbrodSHot spots in dynamic (18)FET-PET delineate malignant tumor parts within suspected WHO grade II gliomasNeuro Oncol201113330731621292686
  • PöpperlGKrethFWMehrkensJHFET PET for the evaluation of untreated gliomas: correlation of FET uptake and uptake kinetics with tumour gradingEur J Nucl Med Mol Imaging200734121933194217763848
  • JansenNLSchwartzCGrauteVPrediction of oligodendroglial histology and LOH 1p/19q using dynamic [18F]FET-PET imaging in intracranial WHO grade II and III gliomasNeuro Oncol201214121473148023090986
  • PöpperlGGoldbrunnerRGildehausFJO-(2-[18F]fluoroethyl)-L-tyrosine PET for monitoring the effects of convection-enhanced delivery of paclitaxel in patients with recurrent glioblastomaEur J Nucl Med Mol Imaging20053291018102515877226
  • JansenNLSuchorskaBSchwarzSB[18F]fluoroethyltyrosine-positron emission tomography-based therapy monitoring after stereotactic iodine-125 brachytherapy in patients with recurrent high-grade gliomaMol Imaging201312313714723490440
  • GotzIGrosuAL[(18)F]FET-PET imaging for treatment and response monitoring of radiation therapy in malignant glioma patients: a reviewFront Oncol2013310423630666
  • van den BentMJInterobserver variation of the histopathological diagnosis in clinical trials on glioma: a clinician’s perspectiveActa Neuropathol2010120329730420644945
  • StuppRMasonWPvan den BentMJRadiotherapy plus concomitant and adjuvant temozolomide for glioblastomaN Engl J Med20053521098799615758009
  • StuppRHegiMEMasonWPvan den BentMJTaphoornMJJanzerRCEffects 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 trialLancet Oncol200910545946619269895
  • ThomasRPRechtLNagpalSAdvances in the management of glioblastoma: the role of temozolomide and MGMT testingClin Pharmacol201351923293540
  • ChinotOLde La Motte RougeTMooreNAVAglio: Phase 3 trial of bevacizumab plus temozolomide and radiotherapy in newly diagnosed glioblastoma multiformeAdv Ther201128433434021432029
  • ChinotOWickWMasonWPhase III trial of bevacizumab added to standard radiotherapy and temozolomide for newly-diagnosed glioblastoma: mature progression-free survival and preliminary overall survival results in AVAglioNeuro Oncol20121410110322013168
  • FiorentinoABalducciMDe BonisPCan elderly patients with newly diagnosed glioblastoma be enrolled in chemoradiotherapy trials?Am J Clin Oncol252013 [Epub ahead of print.]
  • ThonNEigenbrodSGrasbon-FrodlEMPredominant influence of MGMT methylation in non-resectable glioblastoma after radiotherapy plus temozolomideJ Neurol Neurosurg Psychiatry201182444144620861061
  • WiestlerBClausRHartliebSAMalignant astrocytomas of elderly patients lack favorable molecular markers: an analsis of the NOA-08 study collectiveNeuro Oncol20131581017102623595628
  • WickWPlattenMMeisnerCTemozolomide chemotherapy alone versus radiotherapy alone for malignant astrocytoma in the elderly: the NOA-08 randomised, phase 3 trialLancet Oncol201213770771522578793
  • Gállego Pérez-LarrayaJDucrayFChinotOTemozolomide in elderly patients with newly diagnosed glioblastoma and poor performance status: an ANOCEF phase II trialJ Clin Oncol201129223050305521709196
  • GulatiSJakolaASJohannesenTBSolheimOSurvival and treatment patterns of glioblastoma in the elderly: a population-based studyWorld Neurosurg201278551852622381305
  • NiyaziMSchwarzSBSuchorskaBBelkaCRadiotherapy with and without temozolomide in elderly patients with glioblastomaStrahlenther Onkol2012188215415922231634
  • StuppRTonnJCBradaMPentheroudakisGHigh-grade malignant glioma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-upAnn Oncol201021Suppl 5v190v19320555079
  • TonnJCThonNSchnellOKrethFWPersonalized surgical therapyAnn Oncol201223Suppl 10x28x3222987977
  • StummerWPichlmeierUMeinelTWiestlerODZanellaFReulenHJFluorescence-guided surgery with 5-aminolevulinic acid for resection of malignant glioma: a randomised controlled multicentre phase III trialLancet Oncol20067539240116648043
  • StummerWTonnJCMehdornHMCounterbalancing risks and gains from extended resections in malignant glioma surgery: a supplemental analysis from the randomized 5-aminolevulinic acid glioma resection study. Clinical articleJ Neurosurg2011114361362320397896
  • LawsERParneyIFHuangWSurvival following surgery and prognostic factors for recently diagnosed malignant glioma: data from the Glioma Outcomes ProjectJ Neurosurg200399346747312959431
  • OszvaldAGüresirESetzerMGlioblastoma therapy in the elderly and the importance of the extent of resection regardless of ageJ Neurosurg2012116235736421942727
  • LacroixMAbi-SaidDFourneyDRA multivariate analysis of 416 patients with glioblastoma multiforme: prognosis, extent of resection, and survivalJ Neurosurg200195219019811780887
  • SenftCBinkAFranzKVatterHGasserTSeifertVIntraoperative MRI guidance and extent of resection in glioma surgery: a randomised, controlled trialLancet Oncol20111211997100321868284
  • SanaiNPolleyMYMcDermottMWParsaATBergerMSAn extent of resection threshold for newly diagnosed glioblastomasJ Neurosurg201111513821417701
  • GulatiSJakolaASNerlandUSWeberCSolheimOThe risk of getting worse: surgically acquired deficits, perioperative complications, and functional outcomes after primary resection of glioblastomaWorld Neurosurg201176657257922251506
  • BashirRHochbergFOotRRegrowth patterns of glioblastoma multiforme related to planning of interstitial brachytherapy radiation fieldsNeurosurgery198823127302845294
  • JansenEPDewitLGvanHMBartelinkHTarget volumes in radiotherapy for high-grade malignant glioma of the brainRadiother Oncol200056215115610927133
  • WallnerKEGalicichJHKrolGArbitEMalkinMGPatterns of failure following treatment for glioblastoma multiforme and anaplastic astrocytomaInt J Radiat Oncol Biol Phys1989166140514092542195
  • AydinHSillenbergIvonLHPatterns of failure following CT-based 3-D irradiation for malignant gliomaStrahlenther Onkol2001177842443111544905
  • NiyaziMSiefertASchwarzSBTherapeutic options for recurrent malignant gliomaRadiother Oncol201198111421159396
  • MandlESDirvenCMBuisDRPostmaTJVandertopWPRepeated surgery for glioblastoma multiforme: only in combination with other salvage therapySurg Neurol200869550650918262245
  • GuyotatJSignorelliFFrappazDMadarassyGRicciACBretPIs reoperation for recurrence of glioblastoma justified?Oncol Rep20007489990410854567
  • BarkerFG2ndChangSMGutinPHSurvival and functional status after resection of recurrent glioblastoma multiformeNeurosurgery19984247097209574634
  • WellerMCloughesyTPerryJRWickWStandards of care for treatment of recurrent glioblastoma: are we there yet?Neuro Oncol201315142723136223
  • FriedmanHSPradosMDWenPYBevacizumab alone and in combination with irinotecan in recurrent glioblastomaJ Clin Oncol200927284733474019720927
  • KreislTNKimLMooreKPhase II trial of single-agent bevacizumab followed by bevacizumab plus irinotecan at tumor progression in recurrent glioblastomaJ Clin Oncol200927574074519114704
  • TabatabaiGStuppRvan den BentMJMolecular diagnostics of gliomas: the clinical perspectiveActa Neuropathol2010120558559220862485
  • GorliaTvan den BentMJHegiMENomograms for predicting survival of patients with newly diagnosed glioblastoma: prognostic factor analysis of EORTC and NCIC trial 26981-22981/CE.3Lancet Oncol200891293818082451
  • MirimanoffROGorliaTMasonWRadiotherapy and temozolomide for newly diagnosed glioblastoma: recursive partitioning analysis of the EORTC 26981/22981-NCIC CE3 phase III randomized trialJ Clin Oncol200624162563256916735709
  • BucknerJCFactors influencing survival in high-grade gliomasSemin Oncol2003306 Suppl 19101414765378
  • IchimuraKPearsonDMKocialkowskiSIDH1 mutations are present in the majority of common adult gliomas but rare in primary glioblastomasNeuro Oncol200911434134719435942
  • YanHParsonsDWJinGIDH1 and IDH2 mutations in gliomasN Engl J Med2009360876577319228619
  • DangLWhiteDWGrossSCancer-associated IDH1 mutations produce 2-hydroxyglutarateNature2010465730096620559394
  • ParsonsDWJonesSZhangXAn integrated genomic analysis of human glioblastoma multiformeScience200832158971807181218772396
  • WickWHartmannCEngelCNOA-04 randomized phase III trial of sequential chemoradiotherapy of anaplastic glioma with procarbazine, lomustine, and vincristine or temozolomideJ Clin Oncol200927355874588019901110
  • HartmannCHentschelBWickWPatients with IDH1 wild type anaplastic astrocytomas exhibit worse prognosis than IDH1-mutated glioblastomas, and IDH1 mutation status accounts for the unfavorable prognostic effect of higher age: implications for classification of gliomasActa Neuropathol2010120670771821088844
  • ThonNEigenbrodSKrethSIDH1 mutations in grade II astrocytomas are associated with unfavorable progression-free survival and prolonged postrecurrence survivalCancer2012118245246021717448
  • OhgakiHKleihuesPGenetic pathways to primary and secondary glioblastomaAm J Pathol200717051445145317456751
  • StanderMPeraudALerochBKrethFWPrognostic impact of TP53 mutation status for adult patients with supratentorial World Health Organization Grade II astrocytoma or oligoastrocytoma: a long-term analysisCancer200410151028103515329912
  • AgarwalMLAgarwalATaylorWRStarkGRp53 controls both the G2/M and the G1 cell cycle checkpoints and mediates reversible growth arrest in human fibroblastsProc Natl Acad Sci U S A19959218849384977667317
  • CairncrossGWangMShawEPhase III trial of chemoradiotherapy for anaplastic oligodendroglioma: long-term results of RTOG 9402J Clin Oncol201331333734323071247
  • van den BentMJBrandesAATaphoornMJAdjuvant procarbazine, lomustine, and vincristine chemotherapy in newly diagnosed anaplastic oligodendroglioma: long-term follow-up of EORTC brain tumor group study 26951J Clin Oncol201331334435023071237
  • HegiMEJanzerRCLambivWLPresence of an oligodendroglioma-like component in newly diagnosed glioblastoma identifies a pathogenetically heterogeneous subgroup and lacks prognostic value: central pathology review of the EORTC_26981/NCIC_CE.3 trialActa Neuropathol2012123684185222249618
  • Boots-SprengerSHSijbenARijntjesJSignificance of complete 1p/19q co-deletion, IDH1 mutation and MGMT promoter methylation in gliomas: use with cautionMod Pathol201326792292923429602
  • WellerMStuppRHegiMEPersonalized care in neuro-oncology coming of age: why we need MGMT and 1p/19q testing for malignant glioma patients in clinical practiceNeuro Oncol201214Suppl 4iv100iv10823095825
  • Cancer Genome Atlas Research NetworkComprehensive genomic characterization defines human glioblastoma genes and core pathwaysNature200845572161061106818772890
  • WellerMFelsbergJHartmannCMolecular predictors of progression-free and overall survival in patients with newly diagnosed glioblastoma: a prospective translational study of the German Glioma NetworkJ Clin Oncol200927345743575019805672
  • SuzukiMMBirdADNA methylation landscapes: provocative insights from epigenomicsNat Rev Genet20089646547618463664
  • IrizarryRALadd-AcostaCWenBThe human colon cancer methylome shows similar hypo- and hypermethylation at conserved tissue-specific CpG island shoresNat Genet200941217818619151715
  • ChenZXRiggsADDNA methylation and demethylation in mammalsJ Biol Chem201128621183471835321454628
  • HellmanAChessAGene body-specific methylation on the active X chromosomeScience200731558151141114317322062
  • EstellerMCancer epigenomics: DNA methylomes and histone-modification mapsNat Rev Genet20078428629817339880
  • RobertsonKDDNA methylation and human diseaseNat Rev Genet20056859761016136652
  • EstellerMCornPGBaylinSBHermanJGA gene hypermethylation profile of human cancerCancer Res20016183225322911309270
  • CadieuxBChingTTVandenbergSRCostelloJFGenome-wide hypomethylation in human glioblastomas associated with specific copy number alteration, methylenetetrahydrofolate reductase allele status, and increased proliferationCancer Res200666178469847616951158
  • MartinezRMartin-SuberoJIRohdeVA microarray-based DNA methylation study of glioblastoma multiformeEpigenetics20094425526419550145
  • WuXRauchTAZhongXCpG island hypermethylation in human astrocytomasCancer Res20107072718272720233874
  • AgnihotriSWolfAMunozDMA GATA4-regulated tumor suppressor network represses formation of malignant human astrocytomasJ Exp Med2011208468970221464220
  • TepelMRoerigPWolterMFrequent promoter hypermethylation and transcriptional downregulation of the NDRG2 gene at 14q11.2 in primary glioblastomaInt J Cancer200812392080208618709645
  • NakamuraMWatanabeTYonekawaYKleihuesPOhgakiHPromoter methylation of the DNA repair gene MGMT in astrocytomas is frequently associated with G:C –>A:T mutations of the TP53 tumor suppressor geneCarcinogenesis200122101715171911577014
  • NakamuraMWatanabeTKlangbyUp14ARF deletion and methylation in genetic pathways to glioblastomasBrain Pathol200111215916811303791
  • StoneARBoboWBratDJDeviNSVan MeirEGVertinoPMAberrant methylation and down-regulation of TMS1/ASC in human glioblastomaAm J Pathol200416541151116115466382
  • KoslaKPluciennikEKurzykAMolecular analysis of WWOX expression correlation with proliferation and apoptosis in glioblastoma multiformeJ Neurooncol2011101220721320535528
  • WahaAGüntnerSHuangTHEpigenetic silencing of the protocadherin family member PCDH-gamma-A11 in astrocytomasNeoplasia20057319319915799819
  • LindemannCHackmannODelicSSchmidtNReifenbergerGRiemenschneiderMJSOCS3 promoter methylation is mutually exclusive to EGFR amplification in gliomas and promotes glioma cell invasion through STAT3 and FAK activationActa Neuropathol2011122224125121590492
  • AlonsoMMDiez-ValleRManterolaLGenetic and epigenetic modifications of Sox2 contribute to the invasive phenotype of malignant gliomasPLoS One2011611e2674022069467
  • UhlmannKRohdeKZellerCDistinct methylation profiles of glioma subtypesInt J Cancer20031061525912794756
  • VerhaakRGHoadleyKAPurdomEIntegrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1Cancer Cell20101719811020129251
  • NoushmehrHWeisenbergerDJDiefesKIdentification of a CpG island methylator phenotype that defines a distinct subgroup of gliomaCancer Cell201017551052220399149
  • FigueroaMEAbdel-WahabOLuCLeukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiationCancer Cell201018655356721130701
  • WilliamsKChristensenJPedersenMTTET1 and hydroxymethylcytosine in transcription and DNA methylation fidelityNature2011473734734334821490601
  • GuoJUSuYZhongCMingGLSongHHydroxylation of 5-methylcytosine by TET1 promotes active DNA demethylation in the adult brainCell2011145342343421496894
  • LuCWardPSKapoorGSIDH mutation impairs histone demethylation and results in a block to cell differentiationNature2012483739047447822343901
  • XuWYangHLiuYOncometabolite 2-hydroxyglutarate is a competitive inhibitor of alpha-ketoglutarate-dependent dioxygenasesCancer Cell2011191173021251613
  • JinSGJiangYQiuR5-Hydroxymethylcytosine is strongly depleted in human cancers but its levels do not correlate with IDH1 mutationsCancer Res201171247360736522052461
  • KrausTFGlobischDWagnerMLow values of 5-hydroxymethylcytosine (5hmC), the “sixth base,” are associated with anaplasia in human brain tumorsInt J Cancer201213171577159022234893
  • PeggAERepair of O(6)-alkylguanine by alkyltransferasesMutat Res20004622–38310010767620
  • GersonSLMGMT: its role in cancer aetiology and cancer therapeuticsNat Rev Cancer20044429630715057289
  • PreusserMCharles JanzerRFelsbergJAnti-O6-methylguanine-methyltransferase (MGMT) immunohistochemistry in glioblastoma multiforme: observer variability and lack of association with patient survival impede its use as clinical biomarkerBrain Pathol200818452053218400046
  • HegiMEDiserensACGorliaTMGMT gene silencing and benefit from temozolomide in glioblastomaN Engl J Med200535210997100315758010
  • EllingsonBMCloughesyTFPopeWBAnatomic localization of O6-methylguanine DNA methyltransferase (MGMT) promoter methylated and unmethylated tumors: a radiographic study in 358 de novo human glioblastomasNeuroimage201259290891622001163
  • LevnerIDrabyczSRoldanGde RoblesPCairncrossJGMitchellRPredicting MGMT methylation status of glioblastomas from MRI textureMed Image Comput Comput Assist Interv200912Pt 252253020426152
  • RomanoACalabriaLFTavantiFApparent diffusion coefficient obtained by magnetic resonance imaging as a prognostic marker in glioblastomas: correlation with MGMT promoter methylation statusEur Radiol201323251352022875158
  • ParkCKKimJYimSYUsefulness of MS-MLPA for detection of MGMT promoter methylation in the evaluation of pseudoprogression in glioblastoma patientsNeuro Oncol201113219520221075779
  • BrandesAAFranceschiETosoniAMGMT promoter methylation status can predict the incidence and outcome of pseudoprogression after concomitant chemoradiotherapy in newly diagnosed glioblastoma patientsJ Clin Oncol200826132192219718445844
  • NiyaziMSchnellOSuchorskaBFET-PET assessed recurrence pattern after radio-chemotherapy in newly diagnosed patients with glioblastoma is influenced by MGMT methylation statusRadiother Oncol20121041788222673727
  • DrabyczSRoldánGde RoblesPAn analysis of image texture, tumor location, and MGMT promoter methylation in glioblastoma using magnetic resonance imagingNeuroimage20104921398140519796694
  • AhluwaliaMSAmerican Society of Clinical Oncology. 2011 CNS tumors updateExpert Rev Anticancer Ther201111101495149721999122
  • ReifenbergerGHentschelBFelsbergJPredictive impact of MGMT promoter methylation in glioblastoma of the elderlyInt J Cancer201213161342135022139906
  • GilbertMRWangMAldapeKRTOG 0525: a randomized phase III trial comparing standard adjuvant temozolomide (TMZ) with a dose-dense (dd) schedule in newly diagnosed glioblastoma (GBM)Neuro Oncol2011135121123368
  • BaurMPreusserMPiribauerMFrequent MGMT (0(6)-methylguanine-DNA methyltransferase) hypermethylation in long-term survivors of glioblastoma: a single institution experienceRadiol Oncol201044211312022933901
  • FelsbergJThonNEigenbrodSPromoter methylation and expression of MGMT and the DNA mismatch repair genes MLH1, MSH2, MSH6 and PMS2 in paired primary and recurrent glioblastomasInt J Cancer2011129365967021425258
  • van den BentMJDubbinkHJSansonMMGMT promoter methylation is prognostic but not predictive for outcome to adjuvant PCV chemotherapy in anaplastic oligodendroglial tumors: a report from EORTC Brain Tumor Group Study 26951J Clin Oncol200927355881588619901104
  • BadyPSciuscioDDiserensACMGMT 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-statusActa Neuropathol2012124454756022810491
  • Della PuppaAPersanoLMasiGMGMT expression and promoter methylation status may depend on the site of surgical sample collection within glioblastoma: a possible pitfall in stratification of patients?J Neurooncol20121061334121725802
  • HermanJGGraffJRMyohanenSNelkinBDBaylinSBMethylation-specific PCR: a novel PCR assay for methylation status of CpG islandsProc Natl Acad Sci U S A19969318982198268790415
  • ChristiansAHartmannCBennerAMeyerJPrognostic value of three different methods of MGMT promoter methylation analysis in a prospective trial on newly diagnosed glioblastomaPLoS One201273e3344922428052
  • McDonaldKLAwGKleihuesPRole of biomarkers in the clinical management of glioblastomas: what are the barriers and how can we overcome them?Front Neurol2012318823346075
  • EstellerMGarcia-FoncillasJAndionEInactivation of the DNA-repair gene MGMT and the clinical response of gliomas to alkylating agentsN Engl J Med2000343191350135411070098
  • MollemannMWolterMFelsbergJCollinsVPReifenbergerGFrequent promoter hypermethylation and low expression of the MGMT gene in oligodendroglial tumorsInt J Cancer2005113337938515455350
  • Grasbon-FrodlEMKrethFWRuiterMIntratumoral homogeneity of MGMT promoter hypermethylation as demonstrated in serial stereotactic specimens from anaplastic astrocytomas and glioblastomasInt J Cancer2007121112458246417691113
  • MalleyDSHamoudiRAKocialkowskiSPearsonDMCollinsVPIchimuraKA distinct region of the MGMT CpG island critical for transcriptional regulation is preferentially methylated in glioblastoma cells and xenograftsActa Neuropathol2011121565166121287394
  • van VlodropIJNiessenHEDerksSAnalysis of promoter CpG island hypermethylation in cancer: location, location, location!Clin Cancer Res201117134225423121558408
  • KrethSThonNEigenbrodSO-methylguanine-DNA methyltransferase (MGMT) mRNA expression predicts outcome in malignant glioma independent of MGMT promoter methylationPLoS One201162e1715621365007
  • LalezariSChouAPTranACombined analysis of O6-methylguanine-DNA methyltransferase protein expression and promoter methylation provides optimized prognostication of glioblastoma outcomeNeuro Oncol201315337038123328811
  • KrethSLimbeckEHinskeLCIn human glioblastomas transcript elongation by alternative polyadenylation and miRNA targeting is a potent mechanism of MGMT silencingActa Neuropathol2013125567168123340988
  • AgnihotriSGajadharASTernamianCAlkylpurine-DNA-N-glycosylase confers resistance to temozolomide in xenograft models of glioblastoma multiforme and is associated with poor survival in patientsJ Clin Invest2012122125326622156195
  • WellerMPlattenMRothPWickWGeriatric neuro-oncology: from mythology to biologyCurr Opin Neurol201124659960421968549
  • LaperriereNWellerMStuppROptimal management of elderly patients with glioblastomaCancer Treat Rev201339435035722722053
  • MalmströmAGrønbergBHMarosiCTemozolomide versus standard 6-week radiotherapy versus hypofractionated radiotherapy in patients older than 60 years with glioblastoma: the Nordic randomised, phase 3 trialLancet Oncol201213991692622877848
  • GrossmanSAYeXChamberlainMTalampanel with standard radiation and temozolomide in patients with newly diagnosed glioblastoma: a multicenter phase II trialJ Clin Oncol200927254155416119636006
  • PeereboomDMShepardDRAhluwaliaMSPhase II trial of erlotinib with temozolomide and radiation in patients with newly diagnosed glioblastoma multiformeJ Neurooncol2010981939919960228
  • WeilerMHartmannCWiewrodtDChemoradiotherapy of newly diagnosed glioblastoma with intensified temozolomideInt J Radiat Oncol Biol Phys201077367067619836157
  • LaiATranANghiemphuPLPhase II study of bevacizumab plus temozolomide during and after radiation therapy for patients with newly diagnosed glioblastoma multiformeJ Clin Oncol201129214214821135282
  • StuppRHegiMENeynsBPhase I/IIa study of cilengitide and temozolomide with concomitant radiotherapy followed by cilengitide and temozolomide maintenance therapy in patients with newly diagnosed glioblastomaJ Clin Oncol201028162712271820439646
  • ColmanHZhangLSulmanEPA multigene predictor of outcome in glioblastomaNeuro Oncol2010121495720150367