BRAF Inhibition in BRAFV600-Mutant Gliomas: Results From the VE-BASKET Study

Thomas Kaley, Mehdi Touat, Vivek Subbiah, Antoine Hollebecque, Jordi Rodon, A Craig Lockhart, Vicki Keedy, Franck Bielle, Ralf-Dieter Hofheinz, Florence Joly, Jean-Yves Blay, Ian Chau, Igor Puzanov, Noopur S Raje, Jurgen Wolf, Lisa M DeAngelis, Martina Makrutzki, Todd Riehl, Bethany Pitcher, Jose Baselga, David M Hyman, Thomas Kaley, Mehdi Touat, Vivek Subbiah, Antoine Hollebecque, Jordi Rodon, A Craig Lockhart, Vicki Keedy, Franck Bielle, Ralf-Dieter Hofheinz, Florence Joly, Jean-Yves Blay, Ian Chau, Igor Puzanov, Noopur S Raje, Jurgen Wolf, Lisa M DeAngelis, Martina Makrutzki, Todd Riehl, Bethany Pitcher, Jose Baselga, David M Hyman

Abstract

Purpose: BRAFV600 mutations are frequently found in several glioma subtypes, including pleomorphic xanthoastrocytoma (PXA) and ganglioglioma and much less commonly in glioblastoma. We sought to determine the activity of vemurafenib, a selective inhibitor of BRAFV600, in patients with gliomas that harbor this mutation.

Patients and methods: The VE-BASKET study was an open-label, nonrandomized, multicohort study for BRAFV600-mutant nonmelanoma cancers. Patients with BRAFV600-mutant glioma received vemurafenib 960 mg twice per day continuously until disease progression, withdrawal, or intolerable adverse effects. Key end points included confirmed objective response rate by RECIST version 1.1, progression-free survival, overall survival, and safety.

Results: Twenty-four patients (median age, 32 years; 18 female and six male patients) with glioma, including malignant diffuse glioma (n = 11; six glioblastoma and five anaplastic astrocytoma), PXA (n = 7), anaplastic ganglioglioma (n = 3), pilocytic astrocytoma (n = 2), and high-grade glioma, not otherwise specified (n = 1), were treated. Confirmed objective response rate was 25% (95% CI, 10% to 47%) and median progression-free survival was 5.5 months (95% CI, 3.7 to 9.6 months). In malignant diffuse glioma, best response included one partial response and five patients with stable disease, two of whom had disease stabilization that lasted more than 1 year. In PXA, best response included one complete response, two partial responses, and three patients with stable disease. Additional partial responses were observed in patients with pilocytic astrocytoma and anaplastic ganglioglioma (one each). The safety profile of vemurafenib was generally consistent with that of previously published studies.

Conclusion: Vemurafenib demonstrated evidence of durable antitumor activity in some patients with BRAFV600-mutant gliomas, although efficacy seemed to vary qualitatively by histologic subtype. Additional study is needed to determine the optimal use of vemurafenib in patients with primary brain tumors and to identify the mechanisms driving differential responses across histologic subsets.

Trial registration: ClinicalTrials.gov NCT01524978.

Figures

Fig 1.
Fig 1.
Integrated efficacy and treatment duration by patient. Maximal decrease in sum of the longest diameters (SLD), confirmed best response, treatment duration, and prior regimens in patients with (A) PXA, (B) malignant diffuse glioma, and (C) other tumor types. Numbers that appear above individual waterfall bars indicate the percent maximal increase in SLD from baseline. (*) Unchanged from baseline. (†) Patient had no postbaseline assessments. PD was symptomatic deterioration. (§) Patients with secondary malignant diffuse glioma. CR, complete response; D, deleted; MGMT, methylguanine-DNA-methyltransferase gene promoter methylation; IDH1, isocitrate dehydrogenase 1 gene; N, no; NA, not available; NOS, not otherwise specified; PA, pilocytic astrocytoma; PD, progressive disease; PR, partial response; PXA, pleomorphic xanthoastrocytoma; SD, stable disease; UM, unmethylated; WT, wild type; Y, yes.
Fig 2.
Fig 2.
Progression-free survival (PFS) and overall survival (OS) curves in the (A) cohort overall, as well as in patients with (B) pleomorphic xanthoastrocytoma (PXA), (C) malignant diffuse glioma, and (D) other tumor types. NR, not reached.
Fig 3.
Fig 3.
Time to events in individual patients. (*) Anaplastic astrocytoma, n = 5; glioblastoma, n = 6. (†) Anaplastic ganglioglioma, n = 3; pilocytic astrocytoma, n = 2; high-grade glioma, not otherwise specified, n = 1. The first patient with pleomorphic xanthoastrocytoma (PXA; top swimmer lane) experienced his or her first progression at 9 months but continued on treatment beyond progression.

References

    1. Louis DN, Ohgaki H, Wiestler OD, et al. : World Health Organization Histological Classification of Tumours of the Central Nervous System (ed 2). Lyon, France, International Agency for Research on Cancer, 2016
    1. Stupp R, Hegi ME, Mason WP, et al. : Effects 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 trial. Lancet Oncol 10:459-466, 2009
    1. Stupp R, Taillibert S, Kanner AA, et al. : Maintenance therapy with tumor-treating fields plus temozolomide vs temozolomide alone for glioblastoma: A randomized clinical trial. JAMA 314:2535-2543, 2015
    1. Wick W, Gorlia T, Bendszus M, et al. : Lomustine and bevacizumab in progressive glioblastoma. N Engl J Med 377:1954-1963, 2017
    1. Wong ET, Hess KR, Gleason MJ, et al. : Outcomes and prognostic factors in recurrent glioma patients enrolled onto phase II clinical trials. J Clin Oncol 17:2572-2578, 1999
    1. Lamborn KR, Chang SM, Prados MD: Prognostic factors for survival of patients with glioblastoma: Recursive partitioning analysis. Neuro-oncol 6:227-235, 2004
    1. Gilbert MR, Dignam JJ, Armstrong TS, et al. : A randomized trial of bevacizumab for newly diagnosed glioblastoma. N Engl J Med 370:699-708, 2014
    1. Chinot OL, Wick W, Mason W, et al. : Bevacizumab plus radiotherapy-temozolomide for newly diagnosed glioblastoma. N Engl J Med 370:709-722, 2014
    1. Kreisl TN, Kim L, Moore K, et al. : Phase II trial of single-agent bevacizumab followed by bevacizumab plus irinotecan at tumor progression in recurrent glioblastoma. J Clin Oncol 27:740-745, 2009
    1. Friedman HS, Prados MD, Wen PY, et al. : Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J Clin Oncol 27:4733-4740, 2009
    1. Wen PY, Macdonald DR, Reardon DA, et al. : Updated response assessment criteria for high-grade gliomas: Response assessment in neuro-oncology working group. J Clin Oncol 28:1963-1972, 2010
    1. Hyman DM, Taylor BS, Baselga J: Implementing genome-driven oncology. Cell 168:584-599, 2017
    1. Ascierto PA, McArthur GA, Dréno B, et al. : Cobimetinib combined with vemurafenib in advanced BRAFV600-mutant melanoma (coBRIM): Updated efficacy results from a randomised, double-blind, phase 3 trial. Lancet Oncol 17:1248-1260, 2016
    1. Robert C, Karaszewska B, Schachter J, et al. : Improved overall survival in melanoma with combined dabrafenib and trametinib. N Engl J Med 372:30-39, 2015
    1. Planchard D, Besse B, Groen HJM, et al. : Dabrafenib plus trametinib in patients with previously treated BRAFV600E-mutant metastatic non–small-cell lung cancer: An open-label, multicentre phase 2 trial. Lancet Oncol 17:984-993, 2016
    1. Planchard D, Smit EF, Groen HJM, et al. : Dabrafenib plus trametinib in patients with previously untreated BRAFV600E-mutant metastatic non–small-cell lung cancer: An open-label, phase 2 trial. Lancet Oncol 18:1307-1316, 2017
    1. Diamond EL, Subbiah V, Lockhart AC, et al. : Vemurafenib for BRAFV600-mutant Erdheim–Chester disease and Langerhans cell histiocytosis: Analysis of data from the histology-independent phase 2 open-label VE-BASKET study. JAMA Oncol 4:384-388, 2018
    1. Subbiah V, Kreitman RJ, Wainberg ZA, et al. : Dabrafenib and trametinib treatment in patients with locally advanced or metastatic BRAFV600-mutant anaplastic thyroid cancer. J Clin Oncol 36:7-13, 2018
    1. Brose MS, Cabanillas ME, Cohen EE, et al. : Vemurafenib in patients with BRAFV600E-positive metastatic or unresectable papillary thyroid cancer refractory to radioactive iodine: A non-randomised, multicentre, open-label, phase 2 trial. Lancet Oncol 17:1272-1282, 2016
    1. Corcoran RB, Atreya CE, Falchook GS, et al. : Combined BRAF and MEK inhibition with dabrafenib and trametinib in BRAFV600-mutant colorectal cancer. J Clin Oncol 33:4023-4031, 2015
    1. Tiacci E, Park JH, De Carolis L, et al. : Targeting mutant BRAF in relapsed or refractory hairy-cell leukemia. N Engl J Med 373:1733-1747, 2015
    1. Chi AS, Batchelor TT, Yang D, et al. : BRAFV600E mutation identifies a subset of low-grade diffusely infiltrating gliomas in adults. J Clin Oncol 31:e233-e236, 2013
    1. Brennan CW, Verhaak RG, McKenna A, et al. : The somatic genomic landscape of glioblastoma. Cell 155:462-477, 2013. [Erratum: Cell 157:753, 2014]
    1. Dougherty MJ, Santi M, Brose MS, et al. : Activating mutations in BRAF characterize a spectrum of pediatric low-grade gliomas. Neuro-oncol 12:621-630, 2010
    1. Schindler G, Capper D, Meyer J, et al. : Analysis of BRAFV600E mutation in 1,320 nervous system tumors reveals high mutation frequencies in pleomorphic xanthoastrocytoma, ganglioglioma and extra-cerebellar pilocytic astrocytoma. Acta Neuropathol 121:397-405, 2011
    1. Dias-Santagata D, Lam Q, Vernovsky K, et al. : BRAFV600E mutations are common in pleomorphic xanthoastrocytoma: Diagnostic and therapeutic implications. PLoS One 6:e17948, 2011
    1. Chamberlain MC: Salvage therapy with BRAF inhibitors for recurrent pleomorphic xanthoastrocytoma: A retrospective case series. J Neurooncol 114:237-240, 2013
    1. Bautista F, Paci A, Minard-Colin V, et al. : Vemurafenib in pediatric patients with BRAFV600E mutated high-grade gliomas. Pediatr Blood Cancer 61:1101-1103, 2014
    1. Burger MC, Ronellenfitsch MW, Lorenz NI, et al. : Dabrafenib in patients with recurrent, BRAFV600E mutated malignant glioma and leptomeningeal disease. Oncol Rep 38:3291-3296, 2017
    1. Johanns TM, Ferguson CJ, Grierson PM, et al. : Rapid clinical and radiographic response with combined dabrafenib and trametinib in adults with BRAF-mutated high-grade glioma. J Natl Compr Canc Netw 16:4-10, 2018
    1. Brown NF, Carter T, Kitchen N, et al. : Dabrafenib and trametinib in BRAFV600E mutated glioma. CNS Oncol 6:291-296, 2017
    1. Migliorini D, Aguiar D, Vargas MI, et al. : BRAF/MEK double blockade in refractory anaplastic pleomorphic xanthoastrocytoma. Neurology 88:1291-1293, 2017
    1. Hyman DM, Puzanov I, Subbiah V, et al. : Vemurafenib in multiple nonmelanoma cancers with BRAFV600 mutations. N Engl J Med 373:726-736, 2015
    1. Eisenhauer EA, Therasse P, Bogaerts J, et al. : New response evaluation criteria in solid tumours: Revised RECIST guideline (version 1.1). Eur J Cancer 45:228-247, 2009
    1. US National Institutes of Health : Common Terminology Criteria for Adverse Events (CTCAE), version 4.0.
    1. Lin Y, Shih WJ: Adaptive two-stage designs for single-arm phase IIA cancer clinical trials. Biometrics 60:482-490, 2004
    1. Touat M, Idbaih A, Sanson M, et al. : Glioblastoma targeted therapy: Updated approaches from recent biological insights. Ann Oncol 28:1457-1472, 2017
    1. Qazi MA, Vora P, Venugopal C, et al. : Intratumoral heterogeneity: Pathways to treatment resistance and relapse in human glioblastoma. Ann Oncol 28:1448-1456, 2017
    1. Koelsche C, Wöhrer A, Jeibmann A, et al. : Mutant BRAF V600E protein in ganglioglioma is predominantly expressed by neuronal tumor cells. Acta Neuropathol 125:891-900, 2013
    1. Mistry M, Zhukova N, Merico D, et al. : BRAF mutation and CDKN2A deletion define a clinically distinct subgroup of childhood secondary high-grade glioma. J Clin Oncol 33:1015-1022, 2015
    1. Huillard E, Hashizume R, Phillips JJ, et al. : Cooperative interactions of BRAFV600E kinase and CDKN2A locus deficiency in pediatric malignant astrocytoma as a basis for rational therapy. Proc Natl Acad Sci USA 109:8710-8715, 2012
    1. Lassaletta A, Zapotocky M, Mistry M, et al. : Therapeutic and prognostic implications of BRAFV600E in pediatric low-grade gliomas. J Clin Oncol 35:2934-2941, 2017
    1. Cancer Genome Atlas Research Network. Brat DJ, Verhaak RG, et al. : Comprehensive, integrative genomic analysis of diffuse lower-grade gliomas. N Engl J Med 372:2481-2498, 2015
    1. Zehir A, Benayed R, Shah RH, et al. : Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat Med 23:703-713, 2017
    1. Galanis E, Buckner JC, Maurer MJ, et al. : Validation of neuroradiologic response assessment in gliomas: Measurement by RECIST, two-dimensional, computer-assisted tumor area, and computer-assisted tumor volume methods. Neuro Oncol 8:156-165, 2006
    1. Shah GD, Kesari S, Xu R, et al. : Comparison of linear and volumetric criteria in assessing tumor response in adult high-grade gliomas. Neuro Oncol 8:38-46, 2006

Source: PubMed

3
Sottoscrivi