Impact of initial treatment and prognostic factors on postprogression survival in BRAF-mutated metastatic melanoma treated with dacarbazine or vemurafenib ± cobimetinib: a pooled analysis of four clinical trials

Paolo A Ascierto, Antoni Ribas, James Larkin, Grant A McArthur, Karl D Lewis, Axel Hauschild, Keith T Flaherty, Edward McKenna, Qian Zhu, Yong Mun, Brigitte Dréno, Paolo A Ascierto, Antoni Ribas, James Larkin, Grant A McArthur, Karl D Lewis, Axel Hauschild, Keith T Flaherty, Edward McKenna, Qian Zhu, Yong Mun, Brigitte Dréno

Abstract

Background: We sought to identify patient subgroups with distinct postprogression overall survival (ppOS) outcomes and investigate the impact of original treatment assignment and initial postprogression treatment (ppRx) on ppOS.

Methods: Recursive partitioning analysis (RPA) was performed to model relationships between prespecified covariates and ppOS in patients with BRAFV600-mutated metastatic melanoma who had experienced progressive disease (PD) following treatment with cobimetinib plus vemurafenib, vemurafenib monotherapy, or dacarbazine in the BRIM-2, BRIM-3, BRIM-7, and coBRIM studies. Prognostic subgroups identified by RPA were then applied to pooled treatment cohorts. The primary endpoint was ppOS, defined as time from first PD to death from any cause.

Results: RPA identified baseline lactate dehydrogenase (LDH), baseline disease stage, Eastern Cooperative Oncology Group performance status at PD, and ppRx as significant prognostic factors for ppOS. Median ppOS was longest in patients with normal baseline LDH, stage M1c disease at baseline, and ppRx with immunotherapy or targeted therapy (12.2 months; 95% CI 10.3-16.1) and shortest in those with elevated baseline LDH > 2 × upper limit of normal (2.3 months; 95% CI 1.8-2.7). Original treatment assignment did not impact ppOS. Across treatment cohorts, patients treated with immunotherapy or targeted therapy after PD had better ppOS than those given other treatments.

Conclusion: A combination of factors at baseline (LDH, disease stage) and PD (performance status, ppRx) impact ppOS outcomes. ppRx with immunotherapy or targeted therapy is an independent prognostic factor for improved overall survival following progression regardless of original treatment. Trial registration The trials included in this analysis are registered with ClinicalTrials.gov: NCT00949702 (BRIM-2), NCT01006980 (BRIM-3), NCT01271803 (BRIM-7), and NCT01689519 (coBRIM).

Keywords: Cobimetinib; Dacarbazine; Melanoma; Survival analysis; Vemurafenib.

Conflict of interest statement

PAA reports consulting/advisory role for Bristol-Myers Squibb, Roche/Genentech, Merck Sharp & Dohme, Array, Novartis, Merck Serono, Pierre Fabre, Incyte, Genmab, NewLink Genetics, MedImmune, AstraZeneca, Syndax, Sun Pharma, Sanofi, Idera, Ultimovacs, Sandoz, and Immunocore; research funding from Bristol-Myers Squibb, Roche/Genentech, and Array; and travel, accommodations, or expenses from Merck Sharp & Dohme. AR reports honoraria from Amgen, Chugai, Genentech, Merck, and Novartis; consulting/advisory role for Amgen, Chugai, Genentech, Merck, and Novartis; and stock or other ownership in Advaxis, Arcus, Bioncotech, Compugen, CytomX, Five Prime, FLX-Bio, ImaginAb, IsoPlexis, Kite-Gilead, Merus, Rgenix, Lutris, PACT Pharma, and Tango Therapeutics. JL reports honoraria from Achilles Therapeutics, AstraZeneca, Boston Biomedical, Bristol-Myers Squibb, Eisai, EUSA Pharma, GlaxoSmithKline, Ipsen, Imugene, Incyte, Onctura, Kymab, Merck Serono, Merck Sharp & Dohme, Nektar, Novartis, Pierre Fabre, Pfizer, Roche/Genentech, Secarna, and Vitaccess; consulting/advisory role for Achilles Therapeutics, AstraZeneca, Boston Biomedical, Bristol-Myers Squibb, Eisai, EUSA Pharma, GlaxoSmithKline, Ipsen, Imugene, Incyte, Onctura, Kymab, Merck Serono, Merck Sharp & Dohme, Nektar, Novartis, Pierre Fabre, Pfizer, Roche/Genentech, Secarna, and Vitaccess; research funding from Achilles Therapeutics, Aveo, Bristol-Myers Squibb, Merck Sharp & Dohme, Nektar, Novartis, Pfizer, Roche/Genentech, Covance, and Immunocore. GAM reports steering committee membership and principal investigator roles for Roche/Genentech. KDL reports a consulting/advisory role for Roche, Merck, Regeneron, and Array; and research funding from Roche, Merck, Array, Regeneron, Iovance, Incyte, Neon, Nektar, and Bristol-Myers Squibb. AH reports honoraria from Amgen, Bristol-Myers Squibb, Merck Serono, Merck Sharp & Dohme, Philogen, Pierre Fabre, Provectus, Regeneron, Roche, OncoSec, Sanofi-Genzyme, and Novartis Pharma; consulting/advisory role for Amgen, Bristol-Myers Squibb, Merck Serono, Merck Sharp & Dohme, Philogen, Pierre Fabre, Provectus, Regeneron, Roche, OncoSec, Sanofi-Genzyme, and Novartis Pharma; research funding from Amgen, Bristol-Myers Squibb, Merck Serono, Merck Sharp & Dohme, Philogen, Pierre Fabre, Provectus, Regeneron, Roche, Sanofi-Genzyme, and Novartis Pharma. KTF reports consulting/advisory role for Roche, X4 Pharmaceutical, Apricity, PIC Therapeutics, Sanofi, Amgen, Asana, Adaptimmune, Fount Therapeutics, Aeglea, Array BioPharma, Shattuck Labs, Arch Oncology, Tolero Pharmaceuticals, Oncoceutics, Fog Pharma, Neon Therapeutics, Tvardi, Novartis, Genentech, Bristol-Myers Squibb, Merck, Takeda, Verastem, Checkmate, Boston Biomedical, Pierre Fabre, Cell Medica, and Debiopharm; research funding from Roche; stock ownership for Loxo Oncology, Clovis Oncology, Strata Oncology, Vivid Biosciences, X4 Pharmaceuticals, Apricity, PIC Therapeutics, Fount Therapeutics, Shattuck Labs, Oncoceutics, Fog Pharma, and Tvardi; and served on the board of directors for Loxo Oncology, Clovis Oncology, Strata Oncology, and Vivid Biosciences. EM and YM report employment with Genentech and stock or other ownership in Genentech. QZ reports employment with Edwards Lifesciences; stock or other ownership in Edwards Lifesciences; and a consulting/advisory role with Genentech. BD reports honoraria from Bristol-Myers Squibb, Roche, Novartis, and Pierre Fabre; consulting/advisory roles for Bristol-Myers Squibb, Roche, Novartis, and Pierre Fabre; research funding from Bristol-Myers Squibb and Roche; and travel, accommodations, or expenses from Roche and Bristol-Myers Squibb.

Figures

Fig. 1
Fig. 1
Recursive partitioning analysis for prognostic subgroups in all pooled patients. Recursive partitioning decision tree and ppOS outcomes by identified prognostic subgroups for all pooled patients. Data are presented as percentage (95% CI). ECOG PS Eastern Cooperative Oncology Group performance status, IT immunotherapy, LDH lactate dehydrogenase, PD progressive disease, ppOS postprogression overall survival, ppRx postprogression treatment, TT targeted therapy, ULN upper limit of normal
Fig. 2
Fig. 2
Postprogression overall survival by treatment cohort. Kaplan–Meier curves of ppOS in a cobimetinib plus vemurafenib cohort, b vemurafenib monotherapy cohort, and c dacarbazine cohort. CI confidence interval, ECOG PS Eastern Cooperative Oncology Group performance status, IT immunotherapy, LDH lactate dehydrogenase, NE not estimable, PD progressive disease, ppOS postprogression overall survival, ppRx initial postprogression treatment, TT targeted therapy, ULN upper limit of normal
Fig. 3
Fig. 3
Postprogression overall survival by initial postprogression treatment. Kaplan–Meier curves of ppOS in all pooled patients receiving initial postprogression treatment with immunotherapy or targeted therapy (n = 300). ppOS postprogression overall survival
Fig. 4
Fig. 4
Postprogression treatment patterns by initial postprogression treatment. Postprogression treatment patterns in patients who received initial postprogression treatment with a immunotherapy (n = 218), b targeted therapy (n = 82), and c other (n = 727). CTLA-4 cytotoxic T lymphocyte–associated antigen 4, PD-1 programmed death receptor 1

References

    1. Rozeman EA, Dekker TJA, Haanen J, Blank CU. Advanced melanoma: current treatment options, biomarkers, and future perspectives. Am J Clin Dermatol. 2018;19:303–317. doi: 10.1007/s40257-017-0325-6.
    1. Sosman JA, Kim KB, Schuchter L, Gonzalez R, Pavlick AC, Weber JS, et al. Survival in BRAFV600-mutant advanced melanoma treated with vemurafenib. N Engl J Med. 2012;366:707–714. doi: 10.1056/NEJMoa1112302.
    1. Chapman PB, Hauschild A, Robert C, Haanen JB, Ascierto P, Larkin J, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med. 2011;364:2507–2516. doi: 10.1056/NEJMoa1103782.
    1. McArthur GA, Chapman PB, Robert C, Larkin J, Haanen JB, Dummer R, et al. Safety and efficacy of vemurafenib in BRAFv600E and BRAFv600K mutation-positive melanoma (BRIM-3): extended follow-up of a phase 3, randomised, open-label study. Lancet Oncol. 2014;15:323–332. doi: 10.1016/S1470-2045(14)70012-9.
    1. Ribas A, Gonzalez R, Pavlick A, Hamid O, Gajewski TF, Daud A, et al. Combination of vemurafenib and cobimetinib in patients with advanced BRAF(V600)-mutated melanoma: a phase 1b study. Lancet Oncol. 2014;15:954–965. doi: 10.1016/S1470-2045(14)70301-8.
    1. Larkin J, Ascierto PA, Dreno B, Atkinson V, Liszkay G, Maio M, et al. Combined vemurafenib and cobimetinib in BRAF-mutated melanoma. N Engl J Med. 2014;371:1867–1876. doi: 10.1056/NEJMoa1408868.
    1. Ascierto PA, McArthur GA, Dreno B, Atkinson V, Liszkay G, Di Giacomo AM, et al. Cobimetinib combined with vemurafenib in advanced BRAF V600-mutant melanoma (coBRIM): updated efficacy results from a randomised, double-blind, phase 3 trial. Lancet Oncol. 2016;17:1248–1260. doi: 10.1016/S1470-2045(16)30122-X.
    1. Chapman PB, Robert C, Larkin J, Haanen JB, Ribas A, Hogg D, et al. Vemurafenib in patients with BRAFV600 mutation-positive metastatic melanoma: final overall survival results of the randomized BRIM-3 study. Ann Oncol. 2017;28:2581–2587. doi: 10.1093/annonc/mdx339.
    1. Dréno B, Ascierto PA, McArthur GA, Atkinson V, Liszkay G, Di Giacomo AM, et al. Efficacy and safety of cobimetinib (C) combined with vemurafenib (V) in patients (pts) with BRAF V600-mutation–positive metastatic melanoma: analysis from the 4-year extended follow-up follow-up of the phase 3 coBRIM study. J Clin Oncol. 2018;36:9522. doi: 10.1200/JCO.2018.36.15_suppl.9522.
    1. Menzies AM, Wilmott JS, Drummond M, Lo S, Lyle M, Chan MM, et al. Clinicopathologic features associated with efficacy and long-term survival in metastatic melanoma patients treated with BRAF or combined BRAF and MEK inhibitors. Cancer. 2016;121:3826–3835. doi: 10.1002/cncr.29586.
    1. Long GV, Weber JS, Infante JR, Kim KB, Daud A, Gonzalez R, et al. Overall survival and durable responses in patients with BRAF V600-mutant metastatic melanoma receiving dabrafenib combined with trametinib. J Clin Oncol. 2016;34:871–878. doi: 10.1200/JCO.2015.62.9345.
    1. Schadendorf D, Hodi FS, Robert C, Weber JS, Margolin K, Hamid O, et al. Pooled analysis of long-term survival data from phase II and phase III trials of ipilimumab in unresectable or metastatic melanoma. J Clin Oncol. 2015;33:1889–1894. doi: 10.1200/JCO.2014.56.2736.
    1. Dickson PV, Gershenwald JE. Staging and prognosis of cutaneous melanoma. Surg Oncol Clin N Am. 2011;20:1–17. doi: 10.1016/j.soc.2010.09.007.
    1. Balch CM, Gershenwald JE, Soong SJ, Thompson JF, Atkins MB, Byrd DR, et al. Final version of 2009 AJCC melanoma staging and classification. J Clin Oncol. 2009;27:6199–6206. doi: 10.1200/JCO.2009.23.4799.
    1. Long GV, Grob JJ, Nathan P, Ribas A, Robert C, Schadendorf D, et al. Factors predictive of response, disease progression, and overall survival after dabrafenib and trametinib combination treatment: a pooled analysis of individual patient data from randomised trials. Lancet Oncol. 2016;17:1743–1754. doi: 10.1016/S1470-2045(16)30578-2.
    1. Long GV, Blank C, Ribas A, Mortier L, Carlino MS, Lotem M, et al. Impact of baseline serum lactate dehydrogenase concentration on the efficacy of pembrolizumab and ipilimumab in patients with advanced melanoma: data from KEYNOTE-006. Eur J Cancer. 2017;72:S122–S123. doi: 10.1016/S0959-8049(17)30482-3.
    1. Schadendorf D, Long GV, Stroiakovski D, Karaszewska B, Hauschild A, Levchenko E, et al. Three-year pooled analysis of factors associated with clinical outcomes across dabrafenib and trametinib combination therapy phase 3 randomised trials. Eur J Cancer. 2017;82:45–55. doi: 10.1016/j.ejca.2017.05.033.
    1. Hauschild A, Larkin J, Ribas A, Dreno B, Flaherty KT, Ascierto PA, et al. Modeled prognostic subgroups for survival and treatment outcomes in BRAF V600-mutated metastatic melanoma: pooled analysis of 4 randomized clinical trials. JAMA Oncol. 2018;4:1382–1388. doi: 10.1001/jamaoncol.2018.2668.
    1. Ackerman A, Klein O, McDermott DF, Wang W, Ibrahim N, Lawrence DP, et al. Outcomes of patients with metastatic melanoma treated with immunotherapy prior to or after BRAF inhibitors. Cancer. 2014;120:1695–1701. doi: 10.1002/cncr.28620.
    1. Ascierto PA, Simeone E, Sileni VC, Del Vecchio M, Marchetti P, Cappellini GC, et al. Sequential treatment with ipilimumab and BRAF inhibitors in patients with metastatic melanoma: data from the Italian cohort of the ipilimumab expanded access program. Cancer Invest. 2014;32:144–149. doi: 10.3109/07357907.2014.885984.
    1. Polkowska M, Ekk-Cierniakowski P, Czepielewska E, Wysoczanski W, Matusewicz W, Kozlowska-Wojciechowska M. Survival of melanoma patients treated with novel drugs: retrospective analysis of real-world data. J Cancer Res Clin Oncol. 2017;143:2087–2094. doi: 10.1007/s00432-017-2453-z.
    1. Simeone E, Grimaldi AM, Festino L, Giannarelli D, Vanella V, Palla M, et al. Correlation between previous treatment with BRAF inhibitors and clinical response to pembrolizumab in patients with advanced melanoma. Oncoimmunology. 2017;6:e1283462. doi: 10.1080/2162402X.2017.1283462.
    1. Long GV, Atkinson V, Lo S, Sandhu S, Guminski AD, Brown MP, et al. Combination nivolumab and ipilimumab or nivolumab alone in melanoma brain metastases: a multicentre randomised phase 2 study. Lancet Oncol. 2018;19:672–681. doi: 10.1016/S1470-2045(18)30139-6.
    1. Aya F, Fernandez-Martinez A, Gaba L, Victoria I, Tosca M, Pineda E, et al. Sequential treatment with immunotherapy and BRAF inhibitors in BRAF-mutant advanced melanoma. Clin Transl Oncol. 2017;19:119–124. doi: 10.1007/s12094-016-1514-0.
    1. Larkin J, Lao CD, Urba WJ, McDermott DF, Horak C, Jiang J, et al. Efficacy and safety of nivolumab in patients with BRAF V600 mutant and BRAF wild-type advanced melanoma: a pooled analysis of 4 clinical trials. JAMA Oncol. 2015;1:433–440. doi: 10.1001/jamaoncol.2015.1184.
    1. Robert C, Schachter J, Long GV, Arance A, Grob JJ, Mortier L, et al. Pembrolizumab versus ipilimumab in advanced melanoma. N Engl J Med. 2015;372:2521–2532. doi: 10.1056/NEJMoa1503093.
    1. Johnson DB, Pectasides E, Feld E, Ye F, Zhao S, Johnpulle R, et al. Sequencing treatment in BRAFV600 mutant melanoma: anti-PD-1 before and after BRAF inhibition. J Immunother. 2017;40:31–35. doi: 10.1097/CJI.0000000000000148.
    1. Hothorn T, Hornik K, Zeileis A. Unbiased recursive partitioning: a conditional inference framework. J Comput Graph Stat. 2006;15:651–674. doi: 10.1198/106186006X133933.
    1. Chan MM, Haydu LE, Menzies AM, Azer MW, Klein O, Lyle M, et al. The nature and management of metastatic melanoma after progression on BRAF inhibitors: effects of extended BRAF inhibition. Cancer. 2014;120:3142–3153. doi: 10.1002/cncr.28851.
    1. Weber JS, Gibney G, Sullivan RJ, Sosman JA, Slingluff CL, Jr, Lawrence DP, et al. Sequential administration of nivolumab and ipilimumab with a planned switch in patients with advanced melanoma (CheckMate 064): an open-label, randomised, phase 2 trial. Lancet Oncol. 2016;17:943–955. doi: 10.1016/S1470-2045(16)30126-7.

Source: PubMed

3
Abonnere