[en] The mainstay of treatment for adult patients with gliomas, glioneuronal and neuronal tumors consists of combinations of surgery, radiotherapy, and chemotherapy. For many systemic cancers, targeted treatments are a part of the standard of care, however, the predictive significance of most of these targets in central nervous system (CNS) tumors remains less well-studied. Despite that, there is increasing use of advanced molecular diagnostics that identify potential targets, and tumor-agnostic regulatory approvals on targets also present in CNS tumors have been granted. This raises the question of when and for which targets it is meaningful to test in adult patients with CNS tumors. This evidence-based guideline reviews the evidence available for targeted treatment for alterations in the RAS/MAPK pathway (BRAF, NF1), in growth factor receptors (EGFR, ALK, fibroblast growth factor receptor (FGFR), neurotrophic tyrosine receptor kinase (NTRK), platelet-derived growth factor receptor alpha, and ROS1), in cell cycle signaling (CDK4/6, MDM2/4, and TSC1/2) and altered genomic stability (mismatch repair, POLE, high tumor mutational burden (TMB), homologous recombination deficiency) in adult patients with gliomas, glioneuronal and neuronal tumors. At present, targeted treatment for BRAF p.V600E alterations is to be considered part of the standard of care for patients with recurrent gliomas, pending regulatory approval. For approved tumor agnostic treatments for NTRK fusions and high TMB, the evidence for efficacy in adult patients with CNS tumors is very limited, and treatment should preferably be given within prospective clinical registries and trials. For targeted treatment of CNS tumors with FGFR fusions or mutations, clinical trials are ongoing to confirm modest activity so far observed in basket trials. For all other reviewed targets, evidence of benefit in CNS tumors is currently lacking, and testing/treatment should be in the context of available clinical trials.
Disciplines :
Oncology
Author, co-author :
Capper, David ; Department of Neuropathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany. ; German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany.
Reifenberger, Guido; Institute of Neuropathology and German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Heinrich Heine University, Medical Faculty University Hospital Düsseldorf, Moorenstrasse 5, D-40225 Düsseldorf, Germany.
French, Pim J; Department of Neurology, Brain Tumor Center at Erasmus MC Cancer Institute, University Medical Center Rotterdam, The Netherlands.
Schweizer, Leonille ; Institute of Neurology (Edinger Institute), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany. ; German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, German Cancer Research Center (DKFZ), Heidelberg, Germany. ; Frankfurt Cancer Institute (FCI), Frankfurt am Main, Germany.
Weller, Michael ; Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland.
Touat, Mehdi; Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Service de Neurologie 2-Mazarin, Paris, France.
NICLOU, Simone P. ; Department of Cancer Research, NORLUX Neuro-Oncology Laboratory, Luxembourg Institute of Health, 6A, rue Nicolas Ernest-Barblé, L-1210 Luxembourg.
Euskirchen, Philipp ; Department of Neuropathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany. ; Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Klinik für Neurologie, Charitéplatz 1, 10117 Berlin, Germany. ; Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité Comprehensive Cancer Center, Charitéplatz 1, 10117 Berlin, Germany.
Haberler, Christine ; Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria.
Hegi, Monika E; Neuroscience Research Center, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.
Brandner, Sebastian ; Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London London, UK. ; Division of Neuropathology, The National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK.
Le Rhun, Emilie; Department of Neurosurgery and Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland.
Rudà, Roberta; Division of Neurology, Castelfranco Veneto/Treviso Hospital. ; Division of Neuro-Oncology, Department of Neuroscience, University of Turin, Turin, Italy.
Sanson, Marc; Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Service de Neurologie 2-Mazarin, Paris, France.
Tabatabai, Ghazaleh ; Department of Neurology & Interdisciplinary Neuro-Oncology, University Hospital Tübingen and Hertie Institute for Clinical Brain Research, Eberhard Karls University Tübingen. ; Center for Neuro-Oncology, Comprehensive Cancer Center Tübingen-Stuttgart, University Hospital Tübingen. ; German Cancer Consortium (DKTK), partner site Tübingen, Eberhard Karls University Tübingen.
Sahm, Felix ; Department of Neuropathology, University Hospital Heidelberg, CCU Neuropathology, Heidelberg, Germany. ; German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.
Wen, Patrick Y; Center For Neuro-Oncology, Dana-Farber Cancer institute and Harvard Medical School, Boston, USA.
Wesseling, Pieter; Department of Pathology, Amsterdam University Medical Centers/VUmc (De Boelelaan 1117, 1081 HV) Amsterdam, The Netherlands. ; Laboratory for Childhood Cancer Pathology, Princess Máxima Center for Pediatric Oncology (Heidelberglaan 25, 3584 CS Utrecht, The Netherlands.
Preusser, Matthias ; Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria.
van den Bent, Martin J ; Department of Neurology, Brain Tumor Center at Erasmus MC Cancer Institute, University Medical Center Rotterdam, The Netherlands.
Louis DN, Perry A, Wesseling P, et al. The 2021 WHO classification of tumors of the central nervous system: A summary. Neuro Oncol. 2021;23:1231 1251.
Marchiò C, Scaltriti M, Ladanyi M, et al. ESMO recommendations on the standard methods to detect NTRK fusions in daily practice and clinical research. Ann Oncol. 2019;30(9):1417 1427.
Capper D, Preusser M, Habel A, et al. Assessment of BRAF V600E mutation status by immunohistochemistry with a mutation-specific monoclonal antibody. Acta Neuropathol. 2011;122(1):11 19.
Schittenhelm J, Ziegler L, Sperveslage J, et al. FGFR3 overexpression is a useful detection tool for FGFR3 fusions and sequence variations in glioma. Neurooncol Pract. 2021;8:209 221.
Kwak Y, Kim SI, Park CK, et al. C-MET overexpression and amplification in gliomas. Int J Clin Exp Pathol. 2015;8:14932 14938.
Jó?wiak S, Kwiatkowski D, Kotulska K, et al. Tuberin and hamartin expression is reduced in the majority of subependymal giant cell astrocytomas in tuberous sclerosis complex consistent with a two-hit model of pathogenesis. J Child Neurol. 2004;19(2):102 106.
Reuss DE, Habel A, Hagenlocher C, et al. Neurofibromin specific antibody differentiates malignant peripheral nerve sheath tumors (MPNST) from other spindle cell neoplasms. Acta Neuropathol. 2014;127(4):565 572.
Touat M, Li YY, Boynton AN, et al. Mechanisms and therapeutic implications of hypermutation in gliomas. Nature. 2020;580(7804):517 523.
Capper D, Stichel D, Sahm F, et al. Practical implementation of DNA methylation and copy-number-based CNS tumor diagnostics: The Heidelberg experience. Acta Neuropathol. 2018;136(2):181 210.
Chalmers ZR, Connelly CF, Fabrizio D, et al. Analysis of 100, 000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 2017;9:34.
Vega DM, Yee LM, McShane LM, et al. Aligning tumor mutational burden (TMB) quantification across diagnostic platforms: phase II of the Friends of Cancer Research TMB Harmonization Project. Ann Oncol. 2021;32(12):1626 1636.
Zehir A, Benayed R, Shah RH, et al. Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10, 000 patients. Nat Med. 2017;23:703 713.
Merino DM, McShane LM, Fabrizio D, et al. Establishing guidelines to harmonize tumor mutational burden (TMB): in silico assessment of variation in TMB quantification across diagnostic platforms: phase I of the Friends of Cancer Research TMB Harmonization Project. J ImmunoTher Cancer. 2020;8:e000147.
Wimmer K, Kratz CP, Vasen HF, et al. Diagnostic criteria for constitutional mismatch repair deficiency syndrome: suggestions of the European consortium "care for CMMRD" (C4CMMRD). J Med Genet. 2014;51:355 365.
Bakry D, Aronson M, Durno C, et al. Genetic and clinical determinants of constitutional mismatch repair deficiency syndrome: report from the constitutional mismatch repair deficiency consortium. Eur J Cancer. 2014;50(5):987 996.
Aronson M, Colas C, Shuen A, et al. Diagnostic criteria for constitutional mismatch repair deficiency (CMMRD): recommendations from the international consensus working group. J Med Genet. 2022;59:318 327.
den Dunnen JT, Dalgleish R, Maglott DR, et al. HGVS recommendations for the description of sequence variants: 2016 update. Hum Mutat. 2016;37(6):564 569.
Li MM, Datto M, Duncavage EJ, et al. Standards and guidelines for the interpretation and reporting of sequence variants in cancer: A joint consensus Recommendation of the Association for Molecular Pathology, American Society of Clinical Oncology, and College of American Pathologists. J Mol Diagn. 2017;19:4 23.
Horak P, Griffith M, Danos AM, et al. Standards for the classification of pathogenicity of somatic variants in cancer (oncogenicity): joint recommendations of Clinical Genome Resource (ClinGen), Cancer Genomics Consortium (CGC), and Variant Interpretation for Cancer Consortium (VICC). Genet Med. 2022;24:986 998.
Richards S, Aziz N, Bale S, et al. Standards and guidelines for the interpretation of sequence variants: A joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med. 2015;17(5):405 424.
Mateo J, Chakravarty D, Dienstmann R, et al. A framework to rank genomic alterations as targets for cancer precision medicine: The ESMO Scale for Clinical Actionability of molecular Targets (ESCAT). Ann Oncol. 2018;29(9):1895 1902.
Leichsenring J, Horak P, Kreutzfeldt S, et al. Variant classification in precision oncology. Int J Cancer. 2019;145(11):2996 3010.
Dombi E, Baldwin A, Marcus LJ, et al. Activity of selumetinib in neurofibromatosis type 1-related plexiform neurofibromas. N Engl J Med. 2016;375(26):2550 2560.
Mullard A. BRAF plus MEK inhibitor combo secures tumour-Agnostic FDA approval. Nat Rev Drug Discov. 2022. https://dx.doi.org/10.1038/ d41573-022-00117-y
Italiano A, Nanda S, Briggs A, et al. Larotrectinib versus prior therapies in tropomyosin receptor kinase fusion cancer: An intra-patient comparative analysis. Cancers. 2020;12(11):3246.
CHMP) EMACfMPfHU. Guideline on the Evaluation of Anticancer Medicinal Products in Man; 2017. https://www.ema.europa.eu/en/ documents/scientific-guideline/guideline-evaluation-Anticancermedicinal-products-man-revision-5_en.pdf.
Bailey CH, Jameson G, Sima C, et al. Progression-free survival decreases with each subsequent therapy in patients presenting for phase I clinical trials. J Cancer. 2012;3:7 13.
Von Hoff DD, Stephenson JJ, Jr, Rosen P, et al. Pilot study using molecular profiling of patients tumors to find potential targets and select treatments for their refractory cancers. J Clin Oncol. 2010;28(33):4877 4883.
Mellinghoff IK, Ellingson BM, Touat M, et al. Ivosidenib in isocitrate dehydrogenase 1 mutated advanced glioma. J Clin Oncol. doi: 10.1200/ JCO.19.03327.
Cherny NI, Dafni U, Bogaerts J, et al. ESMO-magnitude of clinical benefit scale version 1.1. Ann Oncol. 2017;28(10):2340 2366.
Barthel FP, Johnson KC, Varn FS, et al. Longitudinal molecular trajectories of diffuse glioma in adults. Nature. 2019;576(7785):112 120.
Lassaletta A, Zapotocky M, Mistry M, et al. Therapeutic and prognostic implications of BRAF V600E in pediatric low-grade gliomas. J Clin Oncol. 2017;35:2934 2941.
Furuta T, Miyoshi H, Komaki S, et al. Clinicopathological and genetic association between epithelioid glioblastoma and pleomorphic xanthoastrocytoma. Neuropathology. 2018;38(3):218 227.
Nix JS, Blakeley J, Rodriguez FJ. An update on the central nervous system manifestations of neurofibromatosis type 1. Acta Neuropathol. 2020;139:625 641.
Miller TW, Traphagen NA, Li J, et al. Tumor pharmacokinetics and pharmacodynamics of the CDK4/6 inhibitor ribociclib in patients with recurrent glioblastoma. J Neurooncol. 2019;144(3):563 572.
Tien AC, Li J, Bao X, et al. A phase 0 trial of ribociclib in recurrent glioblastoma patients incorporating a tumor pharmacodynamicand pharmacokinetic-guided expansion cohort. Clin Cancer Res. 2019;25(19):5777 5786.
Taylor JW, Parikh M, Phillips JJ, et al. Phase-2 trial of palbociclib in adult patients with recurrent RB1-positive glioblastoma. J Neurooncol. 2018;140(2):477 483.
Golebiewska A, Hau AC, Oudin A, et al. Patient-derived organoids and orthotopic xenografts of primary and recurrent gliomas represent relevant patient avatars for precision oncology. Acta Neuropathol. 2020;140(6):919 949.
Suwala AK, Stichel D, Schrimpf D, et al. Primary mismatch repair deficient IDH-mutant astrocytoma (PMMRDIA) is a distinct type with a poor prognosis. Acta Neuropathol. 2021;141:85 100.
Marabelle A, Le DT, Ascierto PA, et al. Efficacy of pembrolizumab in patients with noncolorectal high microsatellite instability/mismatch repair-deficient cancer: results from the phase II KEYNOTE-158 study. J Clin Oncol. 2020;38:1 10.