EANO guidelines on the diagnosis and treatment of diffuse gliomas of adulthood

Michael Weller, Martin van den Bent, Matthias Preusser, Emilie Le Rhun, Jörg C Tonn, Giuseppe Minniti, Martin Bendszus, Carmen Balana, Olivier Chinot, Linda Dirven, Pim French, Monika E Hegi, Asgeir S Jakola, Michael Platten, Patrick Roth, Roberta Rudà, Susan Short, Marion Smits, Martin J B Taphoorn, Andreas von Deimling, Manfred Westphal, Riccardo Soffietti, Guido Reifenberger, Wolfgang Wick, Michael Weller, Martin van den Bent, Matthias Preusser, Emilie Le Rhun, Jörg C Tonn, Giuseppe Minniti, Martin Bendszus, Carmen Balana, Olivier Chinot, Linda Dirven, Pim French, Monika E Hegi, Asgeir S Jakola, Michael Platten, Patrick Roth, Roberta Rudà, Susan Short, Marion Smits, Martin J B Taphoorn, Andreas von Deimling, Manfred Westphal, Riccardo Soffietti, Guido Reifenberger, Wolfgang Wick

Abstract

In response to major changes in diagnostic algorithms and the publication of mature results from various large clinical trials, the European Association of Neuro-Oncology (EANO) recognized the need to provide updated guidelines for the diagnosis and management of adult patients with diffuse gliomas. Through these evidence-based guidelines, a task force of EANO provides recommendations for the diagnosis, treatment and follow-up of adult patients with diffuse gliomas. The diagnostic component is based on the 2016 update of the WHO Classification of Tumors of the Central Nervous System and the subsequent recommendations of the Consortium to Inform Molecular and Practical Approaches to CNS Tumour Taxonomy - Not Officially WHO (cIMPACT-NOW). With regard to therapy, we formulated recommendations based on the results from the latest practice-changing clinical trials and also provide guidance for neuropathological and neuroradiological assessment. In these guidelines, we define the role of the major treatment modalities of surgery, radiotherapy and systemic pharmacotherapy, covering current advances and cognizant that unnecessary interventions and expenses should be avoided. This document is intended to be a source of reference for professionals involved in the management of adult patients with diffuse gliomas, for patients and caregivers, and for health-care providers.

Conflict of interest statement

M.W. reports grants and personal fees from Abbvie, Merck (EMD) and MSD, grants from Adastra, Dracen and Novocure, personal fees from Basilea, BMS, Celgene, Medac, Nerviano Medical Sciences, Orbus, Roche and Tocagen. M.vdB. reports personal fees from Abbvie, Agios, Bayer, Carthera, Genenta, Karyopharm and Nerviano Medical Sciences. M.Preusser reports grants and personal fees from Abbvie, Daiichi Sankyo, GlaxoSmithKline and MSD, grants from Böhringer Ingelheim, Bristol Myers Squibb, Novocure and Roche, and personal fees from AstraZeneca, Bayer, BMJ Journals, BMS, CMC Contrast, Gerson Lehrman Group, Lilly, Medahead, MedMedia, Mundipharma, Novartis, Roche, Sanofi and Tocagen. E.L.R. reports personal fees from Abbvie, Daiichy Sankyo and Tocagen. J.C.T. reports personal fees from BrainLab and Carthera. M.B. reports grants and personal fees from Novartis, grants from Siemens, and personal fees from B. Braun, Bayer, Boehringer Ingelheim, Grifols, Merck, Springer, Teva and Vascular Dynamics. C.B. reports personal fees from Celgene, Karyopharm, Lipopharma and Pharmamar, and other support from Abbvie. O.C. reports personal fees and travel support from Abbvie. M.E.H. reports funding from Bristol Myers Squibb and Novocure. A.S.J. reports personal fees from INOMED. M.Pl. reports non-financial support from Pfizer and Roche, personal fees and supply of reagents from Bayer, and personal fees from Affiris, Apogenix and Novartis. In addition, M.Pl has six patents licensed to Bayer (EP2753315B1, WO2018146010A1, WO2019101643A1, WO2019101647A1, WO2019101641A1 and WO2019101642A1), one patent issued (EP2800580B1) and three patents pending (US20180155403A1, US20180246118A1 and US20170254803A1). P.R. reports grants and personal fees from Novocure and personal fees from Bristol Myers Squibb, Debiopharm, Medac, Merck, MSD, QED and Roche. S.S. reports personal fees from Hox Therapeutics and Tocagen and personal fees and support of reagents from Abbvie. M.S. reports other support from GE Healthcare and Parexel. A.vD. receives royalties for the diagnostic use of IDH1 R132H mutant-specific antibody H09; all terms are being managed by the German Cancer Research Center in accordance with its conflict of interest policies. R.S. reports personal fees from AstraZeneca, EISAI, Merck and Puma Biotechnologies. G.R. reports personal fees from Abbvie. G.M., L.D., P.F., R.R., M.J.B.T., M.W. and W.W. declare no competing interests.

Figures

Fig. 1. Diagnostic algorithm for the integrated…
Fig. 1. Diagnostic algorithm for the integrated classification of the major diffuse gliomas in adults.
Tissue specimens obtained through biopsy sampling in patients with diffuse gliomas are routinely assessed by immunohistochemistry for the presence of R132H-mutant IDH1 and loss of nuclear ATRX. In patients aged >55 years with a histologically typical glioblastoma, without a pre-existing lower grade glioma, with a non-midline tumour location and with retained nuclear ATRX expression, immunohistochemical negativity for IDH1 R132H suffices for the classification as IDH-wild-type glioblastoma. In all other instances of diffuse gliomas, a lack of IDH1 R132H immunopositivity should be followed by IDH1 and IDH2 DNA sequencing to detect or exclude the presence of non-canonical mutations. IDH-wild-type diffuse astrocytic gliomas without microvascular proliferation or necrosis should be tested for EGFR amplification, TERT promoter mutation and a +7/–10 cytogenetic signature as molecular characteristics of IDH-wild-type glioblastomas. In addition, the presence of histone H3.3 G34R/V mutations should be assessed by immunohistochemistry or DNA sequencing to identify H3.3 G34-mutant diffuse hemispheric gliomas, in particular in young patients with IDH-wild-type gliomas (such as those <50 years of age with nuclear ATRX loss in tumour cells). Diffuse gliomas of the thalamus, brainstem or spinal cord should be evaluated for histone H3 K27M mutations and loss of nuclear K27-trimethylated histone H3 (H3K27me3) to identify H3 K27M-mutant diffuse midline gliomas. The presence and absence of the diagnostically most relevant molecular alterations for each tumour type are highlighted with red and green boxes. MVP, microvascular proliferation.
Fig. 2. Clinical pathway for IDH-mutant gliomas.
Fig. 2. Clinical pathway for IDH-mutant gliomas.
KPS, Karnofsky performance status; PCV, procarbazine, lomustine and vincristine.
Fig. 3. Clinical pathway for IDH-wild-type glioblastomas,…
Fig. 3. Clinical pathway for IDH-wild-type glioblastomas, WHO grade 4.
KPS, Karnofsky performance status.

References

    1. Louis, D. N., Ohgaki, H., Wiestler, O. D. & Cavenee, W. K. (eds) World Health Organization Histological Classification of Tumors of the Central Nervous System (International Agency for Research on Cancer, 2016)
    1. Brat DJ, et al. cIMPACT-NOW update 3: recommended diagnostic criteria for ‘Diffuse astrocytic glioma, IDH-wildtype, with molecular features of glioblastoma, WHO grade IV’. Acta Neuropathol. 2018;136:805–810.
    1. Brat DJ, et al. cIMPACT-NOW update 5: recommended grading criteria and terminologies for IDH-mutant astrocytomas. Acta Neuropathol. 2020;139:603–608.
    1. Louis DN, et al. cIMPACT-NOW update 6: new entity and diagnostic principle recommendations of the cIMPACT-Utrecht meeting on future CNS tumor classification and grading. Brain Pathol. 2020;30:844–856.
    1. Weller M, et al. European association for neuro-oncology (EANO) guideline on the diagnosis and treatment of adult astrocytic and oligodendroglial gliomas. Lancet Oncol. 2017;18:e315–e329.
    1. Brainin M, et al. Guidance for the preparation of neurological management guidelines by EFNS scientific task forces-revised recommendations 2004. Eur. J. Neurol. 2004;11:577–581.
    1. Ostrom QT, et al. CBTRUS statistical report: primary brain and other central nervous system tumors diagnosed in the United States in 2012-2016. Neuro-Oncology. 2019;21(Suppl. 5):v1–v100.
    1. Rice T, et al. Understanding inherited genetic risk of adult glioma - a review. Neurooncol. Pract. 2016;3:10–16.
    1. Posti JP, et al. Presenting symptoms of glioma in adults. Acta Neurol. Scand. 2015;131:88–93.
    1. Rasmussen BK, et al. Epidemiology of glioma: clinical characteristics, symptoms, and predictors of glioma patients grade I-IV in the the danish neuro-oncology registry. J. Neurooncol. 2017;135:571–579.
    1. Peeters MCM, et al. Prediagnostic symptoms and signs of adult glioma: the patients’ view. J. Neurooncol. 2020;146:293–301.
    1. Pace, A. et al. Determining medical decision-making capacity in brain tumor patients: why and how? Neurooncol. Pract.10.1093/nop/npaa040 (2020).
    1. Nayak L, et al. The neurologic assessment in neuro-oncology (NANO) scale: a tool to assess neurologic function for integration into the response assessment in neuro-oncology (RANO) criteria. Neuro-Oncology. 2017;19:625–635.
    1. van den Bent MJ, et al. Bevacizumab and temozolomide in patients with first recurrence of WHO grade II and III glioma, without 1p/19q co-deletion (TAVAREC): a randomised controlled phase 2 EORTC trial. Lancet Oncol. 2018;19:1170–1179.
    1. Folstein MF, Folstein SE, McHugh PR. ‘Mini-mental state’. A practical method for grading the cognitive state of patients for the clinician. J. Psychiatr. Res. 1975;12:189–198.
    1. Nasreddine ZS, et al. The montreal cognitive assessment, MoCA: a brief screening tool for mild cognitive impairment. J. Am. Geriatr. Soc. 2005;53:695–699.
    1. Ellingson BM, Wen PY, Cloughesy TF. modified criteria for radiographic response assessment in glioblastoma clinical trials. Neurotherapeutics. 2017;14:307–320.
    1. Albert NL, et al. Response assessment in neuro-oncology working group and european association for neuro-oncology recommendations for the clinical use of PET imaging in gliomas. Neuro-Oncology. 2016;18:1199–1208.
    1. Le Rhun E, Seoane J, Salzet M, Soffietti R, Weller M. Liquid biopsies for diagnosing and monitoring primary tumors of the central nervous system. Cancer Lett. 2020;480:24–28.
    1. Roth, P. et al. Neurological and vascular complications of primary and secondary brain tumors: EANO-ESMO Clinical Practice Guidelines for prophylaxis, diagnosis, treatment and follow-up. Ann. Oncol.10.1093/annonc/mdx221 (2020).
    1. Williams M, Treasure P, Greenberg D, Brodbelt A, Collins P. Surgeon volume and 30 day mortality for brain tumours in England. Br. J. Cancer. 2016;115:1379–1382.
    1. Eigenbrod S, et al. Molecular stereotactic biopsy technique improves diagnostic accuracy and enables personalized treatment strategies in glioma patients. Acta Neurochir. 2014;156:1427–1440.
    1. Hamisch CA, et al. Frame-based stereotactic biopsy of deep-seated and midline structures in 511 procedures: feasibility, risk profile, and diagnostic yield. Acta Neurochir. 2019;161:2065–2071.
    1. Grasbon-Frodl EM, et al. Intratumoral homogeneity of MGMT promoter hypermethylation as demonstrated in serial stereotactic specimens from anaplastic astrocytomas and glioblastomas. Int. J. Cancer. 2007;121:2458–2464.
    1. Millesi M, et al. Is intraoperative pathology needed if 5-aminolevulinic-acid-induced tissue fluorescence is found in stereotactic brain tumor biopsy? Neurosurgery. 2020;86:366–373.
    1. Shirahata M, et al. Novel, improved grading system(s) for IDH-mutant astrocytic gliomas. Acta Neuropathol. 2018;136:153–166.
    1. Banan R, et al. Infratentorial IDH-mutant astrocytoma is a distinct subtype. Acta Neuropathol. 2020;140:569–581.
    1. Reinhardt A, et al. Tumors diagnosed as cerebellar glioblastoma comprise distinct molecular entities. Acta Neuropathol. Commun. 2019;7:163.
    1. Appay R, et al. CDKN2A homozygous deletion is a strong adverse prognosis factor in diffuse malignant IDH-mutant gliomas. Neuro-Oncology. 2019;21:1519–1528.
    1. Tesileanu CMS, et al. Survival of diffuse astrocytic glioma, IDH1/2 wildtype, with molecular features of glioblastoma, WHO grade IV: a confirmation of the cIMPACT-NOW criteria. Neuro-Oncology. 2020;22:515–523.
    1. Reuss DE, et al. Adult IDH wild type astrocytomas biologically and clinically resolve into other tumor entities. Acta Neuropathol. 2015;130:407–417.
    1. Wick W, et al. Prognostic or predictive value of MGMT promoter methylation in gliomas depends on IDH1 mutation. Neurology. 2013;81:1515–1522.
    1. Bady P, Delorenzi M, Hegi ME. Sensitivity analysis of the MGMT-STP27 model and impact of genetic and epigenetic context to predict the MGMT methylation status in gliomas and other tumors. J. Mol. Diagn. 2016;18:350–361.
    1. Preusser M, et al. Anti-O6-methylguanine-methyltransferase (MGMT) immunohistochemistry in glioblastoma multiforme: observer variability and lack of association with patient survival impede its use as clinical biomarker. Brain Pathol. 2008;18:520–532.
    1. Sahm F, et al. Next-generation sequencing in routine brain tumor diagnostics enables an integrated diagnosis and identifies actionable targets. Acta Neuropathol. 2016;131:903–910.
    1. Zacher A, et al. Molecular diagnostics of gliomas using next generation sequencing of a glioma-tailored gene panel. Brain Pathol. 2017;27:146–159.
    1. Capper D, et al. DNA methylation-based classification of central nervous system tumours. Nature. 2018;555:469–474.
    1. Ferguson SD, et al. Targetable gene fusions associate with the IDH wild-type astrocytic lineage in adult gliomas. J. Neuropathol. Exp. Neurol. 2018;77:437–442.
    1. Stichel D, et al. Routine RNA sequencing of formalin-fixed paraffin-embedded specimens in neuropathology diagnostics identifies diagnostically and therapeutically relevant gene fusions. Acta Neuropathol. 2019;138:827–835.
    1. Stummer W, et al. Fluorescence-guided surgery with 5-aminolevulinic acid for resection of malignant glioma: a randomised controlled multicentre phase III trial. Lancet Oncol. 2006;7:392–401.
    1. De Witt Hamer PC, Robles SG, Zwinderman AH, Duffau H, Berger MS. Impact of intraoperative stimulation brain mapping on glioma surgery outcome: a meta-analysis. J. Clin. Oncol. 2012;30:2559–2565.
    1. Brennum J, Maier CM, Almdal K, Engelmann CM, Gjerris M. Primo non nocere or maximum survival in grade 2 gliomas? A medical ethical question. Acta Neurochir. 2015;157:155–164.
    1. Gulati S, Jakola AS, Nerland US, Weber C, Solheim O. The risk of getting worse: surgically acquired deficits, perioperative complications, and functional outcomes after primary resection of glioblastoma. World Neurosurg. 2011;76:572–579.
    1. Bette S, et al. Patterns and time dependence of unspecific enhancement in postoperative magnetic resonance imaging after glioblastoma resection. World Neurosurg. 2016;90:440–447.
    1. Grabowski MM, et al. Residual tumor volume versus extent of resection: predictors of survival after surgery for glioblastoma. J. Neurosurg. 2014;121:1115–1123.
    1. Molinaro, A. M. et al. Association of maximal extent of resection of contrast-enhanced and non-contrast-enhanced tumor with survival within molecular subgroups of patients with newly diagnosed glioblastoma. JAMA Oncol.10.1001/jamaoncol.2019.6143 (2020).
    1. Wijnenga MMJ, et al. The impact of surgery in molecularly defined low-grade glioma: an integrated clinical, radiological, and molecular analysis. Neuro-Oncology. 2018;20:103–112.
    1. Press RH, et al. Optimal timing of chemoradiotherapy after surgical resection of glioblastoma: Stratification by validated prognostic classification. Cancer. 2020;126:3255–3264.
    1. Beiko J, et al. IDH1 mutant malignant astrocytomas are more amenable to surgical resection and have a survival benefit associated with maximal surgical resection. Neuro-Oncology. 2014;16:81–91.
    1. Walker MD, et al. Evaluation of BCNU and/or radiotherapy in the treatment of anaplastic gliomas. A cooperative clinical trial. J. Neurosurg. 1978;49:333–343.
    1. Keime-Guibert F, et al. Radiotherapy for glioblastoma in the elderly. N. Engl. J. Med. 2007;356:1527–1535.
    1. Breen WG, et al. Final report from Intergroup NCCTG 86-72-51 (Alliance): a phase III randomized clinical trial of high-dose versus low-dose radiation for adult low-grade glioma. Neuro-Oncology. 2020;22:830–837.
    1. Khan L, et al. External beam radiation dose escalation for high grade glioma. Cochrane Database Syst. Rev. 2020;5:CD011475.
    1. Roa W, et al. Abbreviated course of radiation therapy in older patients with glioblastoma multiforme: a prospective randomized clinical trial. J. Clin. Oncol. 2004;22:1583–1588.
    1. Niyazi M, et al. ESTRO-ACROP guideline ‘target delineation of glioblastomas’. Radiother. Oncol. 2016;118:35–42.
    1. Harrabi SB, et al. Dosimetric advantages of proton therapy over conventional radiotherapy with photons in young patients and adults with low-grade glioma. Strahlenther. Onkol. 2016;192:759–769.
    1. Malouff TD, Peterson JL, Mahajan A, Trifiletti DM. Carbon ion radiotherapy in the treatment of gliomas: a review. J. Neurooncol. 2019;145:191–199.
    1. Nachbichler SB, Kreth F-W. Brachytherapy of intracranial gliomas. Prog. Neurol. Surg. 2018;31:72–86.
    1. Stupp R, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N. Engl. J. Med. 2005;352:987–996.
    1. Weller M, Le Rhun E. How did lomustine become standard of care in recurrent glioblastoma? Cancer Treat. Rev. 2020;87:102029.
    1. Brem H, et al. Placebo-controlled trial of safety and efficacy of intraoperative controlled delivery by biodegradable polymers of chemotherapy for recurrent gliomas. The Polymer-brain Tumor Treatment Group. Lancet. 1995;345:1008–1012.
    1. Westphal M, et al. A phase 3 trial of local chemotherapy with biodegradable carmustine (BCNU) wafers (Gliadel wafers) in patients with primary malignant glioma. Neuro-Oncology. 2003;5:79–88.
    1. Wen, P. Y. et al. Glioblastoma in adults: a Society for Neuro-Oncology (SNO) and European Society of Neuro-Oncology (EANO) consensus review on current management and future directions. Neuro-Oncology10.1093/neuonc/noaa106 (2020).
    1. Touat M, et al. Mechanisms and therapeutic implications of hypermutation in gliomas. Nature. 2020;580:517–523.
    1. Wick W, et al. Lomustine and bevacizumab in progressive glioblastoma. N. Engl. J. Med. 2017;377:1954–1963.
    1. Friedman HS, et al. Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J. Clin. Oncol. 2009;27:4733–4740.
    1. Kreisl TN, et al. Phase II trial of single-agent bevacizumab followed by bevacizumab plus irinotecan at tumor progression in recurrent glioblastoma. J. Clin. Oncol. 2009;27:740–745.
    1. Wen PY, et al. Updated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working group. J. Clin. Oncol. 2010;28:1963–1972.
    1. van den Bent MJ, et al. Response assessment in neuro-oncology (a report of the RANO group): assessment of outcome in trials of diffuse low-grade gliomas. Lancet Oncol. 2011;12:583–593.
    1. Radbruch A, et al. Pseudoprogression in patients with glioblastoma: clinical relevance despite low incidence. Neuro-Oncology. 2015;17:151–159.
    1. Ellingson BM, et al. Consensus recommendations for a standardized Brain Tumor Imaging Protocol in clinical trials. Neuro-Oncology. 2015;17:1188–1198.
    1. Gui C, Lau JC, Kosteniuk SE, Lee DH, Megyesi JF. Radiology reporting of low-grade glioma growth underestimates tumor expansion. Acta Neurochir. 2019;161:569–576.
    1. Zikou A, et al. Radiation necrosis, pseudoprogression, pseudoresponse, and tumor recurrence: imaging challenges for the evaluation of treated gliomas. Contrast Media Mol. Imaging. 2018;2018:6828396.
    1. Pace A, et al. European association for neuro-oncology (EANO) guidelines for palliative care in adults with glioma. Lancet Oncol. 2017;18:e330–e340.
    1. Buckner JC, et al. Radiation plus procarbazine, CCNU, and vincristine in low-grade glioma. N. Engl. J. Med. 2016;374:1344–1355.
    1. Wahl M, et al. Chemotherapy for adult low-grade gliomas: clinical outcomes by molecular subtype in a phase II study of adjuvant temozolomide. Neuro-Oncology. 2017;19:242–251.
    1. Rudà R, et al. Efficacy of initial temozolomide for high-risk low grade gliomas in a phase II AINO (Italian Association for Neuro-Oncology) study: a post-hoc analysis within molecular subgroups of WHO 2016. J. Neurooncol. 2019;145:115–123.
    1. Wick W, et al. NOA-04 randomized phase III trial of sequential radiochemotherapy of anaplastic glioma with procarbazine, lomustine, and vincristine or temozolomide. J. Clin. Oncol. 2009;27:5874–5880.
    1. Cairncross G, et al. Phase III trial of chemoradiotherapy for anaplastic oligodendroglioma: long-term results of RTOG 9402. J. Clin. Oncol. 2013;31:337–343.
    1. van den Bent 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.
    1. Habets EJJ, et al. Health-related quality of life and cognitive functioning in long-term anaplastic oligodendroglioma and oligoastrocytoma survivors. J. Neurooncol. 2014;116:161–168.
    1. Wick W, et al. Long-term analysis of the NOA-04 randomized phase III trial of sequential radiochemotherapy of anaplastic glioma with PCV or temozolomide. Neuro-Oncology. 2016;18:1529–1537.
    1. US National Library of Medicine. (2020).
    1. Jakola AS, et al. Surgical resection versus watchful waiting in low-grade gliomas. Ann. Oncol. 2017;28:1942–1948.
    1. van den Bent MJ, et al. Long-term efficacy of early versus delayed radiotherapy for low-grade astrocytoma and oligodendroglioma in adults: the EORTC 22845 randomised trial. Lancet. 2005;366:985–990.
    1. Baumert BG, et al. Temozolomide chemotherapy versus radiotherapy in high-risk low-grade glioma (EORTC 22033-26033): a randomised, open-label, phase 3 intergroup study. Lancet Oncol. 2016;17:1521–1532.
    1. Bell EH, et al. Comprehensive genomic analysis in NRG oncology/RTOG 9802: A phase III trial of radiation versus radiation plus procarbazine, lomustine (CCNU), and vincristine in high-risk low-grade glioma. J. Clin. Oncol. 2020;38:3407–3417.
    1. van den Bent, M. J. et al. Interim results from the CATNON trial (EORTC study 26053-22054) of treatment with concurrent and adjuvant temozolomide for 1p/19q non-co-deleted anaplastic glioma: a phase 3, randomised, open-label intergroup study. Lancet10.1016/S0140-6736(17)31442-3 (2017).
    1. van den Bent M, et al. ACTR-11. Second interim and 1st molecular analysis of the EORTC randomized phase III intergroup CATNON trial on concurrent and adjuvant temozolomide in anaplastic glioma without 1p/19q codeletion. Neuro-Oncology. 2019;21:vi14–vi14.
    1. Yung WK, et al. Multicenter phase II trial of temozolomide in patients with anaplastic astrocytoma or anaplastic oligoastrocytoma at first relapse. Temodal Brain Tumor Group. J. Clin. Oncol. 1999;17:2762–2771.
    1. Brada M, et al. Temozolomide versus procarbazine, lomustine, and vincristine in recurrent high-grade glioma. J. Clin. Oncol. 2010;28:4601–4608.
    1. Vuorinen V, Hinkka S, Färkkilä M, Jääskeläinen J. Debulking or biopsy of malignant glioma in elderly people - a randomised study. Acta Neurochir. 2003;145:5–10.
    1. Roa W, et al. International atomic energy agency randomized phase III study of radiation therapy in elderly and/or frail patients with newly diagnosed glioblastoma multiforme. J. Clin. Oncol. 2015;33:4145–4150.
    1. Perry JR, et al. Short-course radiation plus temozolomide in elderly patients with glioblastoma. N. Engl. J. Med. 2017;376:1027–1037.
    1. Hegi ME, et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. N. Engl. J. Med. 2005;352:997–1003.
    1. Wick W, et al. Temozolomide chemotherapy alone versus radiotherapy alone for malignant astrocytoma in the elderly: the NOA-08 randomised, phase 3 trial. Lancet Oncol. 2012;13:707–715.
    1. Wick A, et al. Superiority of temozolomide over radiotherapy for elderly patients with RTK II methylation class, MGMT promoter-methylated malignant astrocytoma. Neuro-Oncology. 2020 doi: 10.1093/neuonc/noaa033.
    1. Malmström A, et al. Temozolomide versus standard 6-week radiotherapy versus hypofractionated radiotherapy in patients older than 60 years with glioblastoma: the Nordic randomised, phase 3 trial. Lancet Oncol. 2012;13:916–926.
    1. Weller M. Where does O6 -methylguanine DNA methyltransferase promoter methylation assessment place temozolomide in the future standards of care for glioblastoma? Cancer. 2018;124:1316–1318.
    1. US National Library of Medicine. (2020).
    1. Stupp R, et al. Effect of tumor-treating fields plus maintenance temozolomide vs maintenance temozolomide alone on survival in patients with glioblastoma: a randomized clinical trial. JAMA. 2017;318:2306–2316.
    1. Taphoorn MJB, et al. Influence of treatment with tumor-treating fields on health-related quality of life of patients with newly diagnosed glioblastoma: a secondary analysis of a randomized clinical trial. JAMA Oncol. 2018;4:495–504.
    1. Wick W. TTFields: where does all the skepticism come from? Neuro-Oncology. 2016;18:303–305.
    1. Bernard-Arnoux F, et al. The cost-effectiveness of tumor-treating fields therapy in patients with newly diagnosed glioblastoma. Neuro-Oncology. 2016;18:1129–1136.
    1. Gilbert MR, et al. Dose-dense temozolomide for newly diagnosed glioblastoma: a randomized phase III clinical trial. J. Clin. Oncol. 2013;31:4085–4091.
    1. Balana, C. et al. A phase II randomized, multicenter, open-label trial of continuing adjuvant temozolomide beyond six cycles in patients with glioblastoma (GEINO 14-01). Neuro-Oncology10.1093/neuonc/noaa107 (2020).
    1. Herrlinger U, et al. Lomustine-temozolomide combination therapy versus standard temozolomide therapy in patients with newly diagnosed glioblastoma with methylated MGMT promoter (CeTeG/NOA-09): a randomised, open-label, phase 3 trial. Lancet. 2019;393:678–688.
    1. Chinot OL, et al. Bevacizumab plus radiotherapy-temozolomide for newly diagnosed glioblastoma. N. Engl. J. Med. 2014;370:709–722.
    1. Gilbert MR, Sulman EP, Mehta MP. Bevacizumab for newly diagnosed glioblastoma. N. Engl. J. Med. 2014;370:2048–2049.
    1. Wirsching H-G, et al. Bevacizumab plus hypofractionated radiotherapy versus radiotherapy alone in elderly patients with glioblastoma: the randomized, open-label, phase II ARTE trial. Ann. Oncol. 2018;29:1423–1430.
    1. Weller M, et al. Rindopepimut with temozolomide for patients with newly diagnosed, EGFRvIII-expressing glioblastoma (ACT IV): a randomised, double-blind, international phase 3 trial. Lancet Oncol. 2017;18:1373–1385.
    1. Suchorska B, et al. Complete resection of contrast-enhancing tumor volume is associated with improved survival in recurrent glioblastoma-results from the DIRECTOR trial. Neuro-Oncology. 2016;18:549–556.
    1. Ryu S, et al. The role of radiotherapy in the management of progressive glioblastoma: a systematic review and evidence-based clinical practice guideline. J. Neurooncol. 2014;118:489–499.
    1. Tsien C, et al. ACTR-32. NRG ONCOLOGY RTOG 1205: randomized phase II trial of concurrent bevacizumab and re-irradiation vs. bevacizumab alone as treatment for recurrent glioblastoma. Neuro-Oncology. 2019;21:vi20–vi20.
    1. Batchelor TT, et al. Phase II study of cediranib, an oral pan-vascular endothelial growth factor receptor tyrosine kinase inhibitor, in patients with recurrent glioblastoma. J. Clin. Oncol. 2010;28:2817–2823.
    1. Becker Buxton, M. et al. GBM AGILE: A global, phase II/III adaptive platform trial to evaluate multiple regimens in newly diagnosed and recurrent glioblastoma. J. Clin. Oncol.10.1200/JCO.2020.38.15_suppl.TPS2579 (2020).
    1. Perry JR, et al. Phase II trial of continuous dose-intense temozolomide in recurrent malignant glioma: RESCUE study. J. Clin. Oncol. 2010;28:2051–2057.
    1. Weller M, et al. MGMT promoter methylation is a strong prognostic biomarker for benefit from dose-intensified temozolomide rechallenge in progressive glioblastoma: the DIRECTOR trial. Clin. Cancer Res. 2015;21:2057–2064.
    1. Reardon, D. A. et al. Effect of nivolumab vs bevacizumab in patients with recurrent glioblastoma: the CheckMate 143 phase 3 randomized clinical trial. JAMA Oncol.10.1001/jamaoncol.2020.1024 (2020).
    1. Stupp R, et al. NovoTTF-100A versus physician’s choice chemotherapy in recurrent glioblastoma: a randomised phase III trial of a novel treatment modality. Eur. J. Cancer. 2012;48:2192–2202.
    1. Kaley T, et al. BRAF inhibition in BRAFV600-mutant gliomas: results from the VE-BASKET study. J. Clin. Oncol. 2018;36:3477–3484.
    1. Sturm D, et al. Hotspot mutations in H3F3A and IDH1 define distinct epigenetic and biological subgroups of glioblastoma. Cancer Cell. 2012;22:425–437.
    1. Pitter KL, et al. Corticosteroids compromise survival in glioblastoma. Brain. 2016;139:1458–1471.
    1. Hovey EJ, et al. Continuing or ceasing bevacizumab beyond progression in recurrent glioblastoma: an exploratory randomized phase II trial. Neurooncol Pract. 2017;4:171–181.
    1. US National Library of Medicine. (2020).

Source: PubMed

3
Suscribir