PD-L1 blockade in combination with inhibition of MAPK oncogenic signaling in patients with advanced melanoma

Antoni Ribas, Alain Algazi, Paolo A Ascierto, Marcus O Butler, Sunandana Chandra, Michael Gordon, Leonel Hernandez-Aya, Donald Lawrence, Jose Lutzky, Wilson H Miller Jr, Katie M Campbell, Bruno Delafont, Shannon Marshall, Nancy Mueller, Caroline Robert, Antoni Ribas, Alain Algazi, Paolo A Ascierto, Marcus O Butler, Sunandana Chandra, Michael Gordon, Leonel Hernandez-Aya, Donald Lawrence, Jose Lutzky, Wilson H Miller Jr, Katie M Campbell, Bruno Delafont, Shannon Marshall, Nancy Mueller, Caroline Robert

Abstract

Combining PD-L1 blockade with inhibition of oncogenic mitogen-activated protein kinase (MAPK) signaling may result in long-lasting responses in patients with advanced melanoma. This phase 1, open-label, dose-escalation and -expansion study (NCT02027961) investigated safety, tolerability and preliminary efficacy of durvalumab (anti-PD-L1) combined with dabrafenib (BRAF inhibitor) and trametinib (MEK inhibitor) for patients with BRAF-mutated melanoma (cohort A, n = 26), or durvalumab and trametinib given concomitantly (cohort B, n = 20) or sequentially (cohort C, n = 22) for patients with BRAF-wild type melanoma. Adverse events and treatment discontinuation rates were more common than previously reported for these agents given as monotherapy. Objective responses were observed in 69.2% (cohort A), 20.0% (cohort B) and 31.8% (cohort C) of patients, with evidence of improved tumor immune infiltration and durable responses in a subset of patients with available biopsy samples. In conclusion, combined MAPK inhibition and anti-PD-L1 therapy may provide treatment options for patients with advanced melanoma.

Conflict of interest statement

A.R. has received honoraria from consulting with Amgen, BMS, Chugai, Genentech, Merck, Novartis, Roche and Sanofi, is/has been a member of the scientific advisory board and holds stock in Advaxis, Apricity, Arcus Biosciences, Bioncotech Therapeutics, Compugen, CytomX, Five Prime, FLX-Bio, ImaginAb, IsoPlexis, Kite-Gilead, Lutris Pharma, Merus, PACT Pharma, Rgenix and Tango Therapeutics, and has received research funding from Agilent and BMS through SU2C. A.A. reports grants from AstraZeneca, GSK and Novartis, during the conduct of the study, consulting fees, travel support and stock options from OncoSec, stock options for consulting from Valitor, honoraria and research funding to UCSF from Regeneron, honoraria from Array, and grants in the form of research funding to UCSF from Acerta, Amgen, AstraZeneca, BMS, Dynavax, Genentech, Idera, Incyte, ISA, LOXO, Merck, Novartis, Pfizer, Sensei, Tessa. P.A.A. reports grants or personal fees for advisory/consultancy work and research funding from BMS, Roche-Genentech and Array, personal fees for advisory/consultancy work and travel support from MSD, personal fees for advisory/consultancy work from Novartis, Merck Serono, Pierre Fabre, Incyte, Genmab, NewLink Genetics, Medimmune, AstraZeneca, Syndax, Sun Pharma, Sanofi, Idera, Ultimovacs, Sandoz, Immunocore, 4SC, Alkermes, and Nektar, and personal fees for consultancy work from Italfarmaco. M.B. reports he is contracted by his institute to complete work on this study for AstraZeneca, along with personal fees and honoraria for consultancy work from Merck, BMS, Novartis, Sanofi, Pfizer, Immunocore, Adaptimmune, and EMD Serono. S.C. has received honoraria from consulting with and/or has been a member of the scientific advisory board of BMS, Novartis, Array BioPharma, Regeneron, Sanofi-Genzyme, and Exicure. M.G. reports institutional research support from Medimmune, Merck, BMS, Amgen, Tesaro, Beigene, ABBVIE, Aeglea, Arcus, Astex, BluePrint, Calithera, CellDex, Corcept, Clovis, Eli Lilly, Endocyte, Five Prime, Genocea, Neon, Plexxicon, Revolution Medicine, Seattle Genetics, Serono, SynDevRx, and Tolero, and personal fees and institutional research support from Agenus, Imaging Endpoints, Tracon, Deciphera, and Salarius. L.H.-A. reports personal fees for study-related fees to institution from AstraZenaca, personal fees paid to institution for conduction of clinical trials from BMS, Merck, Amgen, Roche, Regeneron, Novartis, Immunocore, Merck-EMD, Corvus, Polynoma, Genentech. J.L. reports personal fees for advisory board consultancy/attendance from Castle, Array Pharmaceuticals and Kimera and personal fees for advisory board and speaker bureau work from Regeneron Pharmaceuticals. W.H.M. reports consultancy fees from BMS, Merck, Roche, Novartis, Amgen, and GSK. KMC is a shareholder in Geneoscopy LLC. B.D., S.M., and N.M. are all employees of AstraZeneca. C.R. has received honoraria from consulting with Amgen, BMS, MSD, Novartis, Roche, CureVac, Biother, Sanofi, Pierre Fabre, and Merck.

Figures

Fig. 1. Best percentage change from baseline…
Fig. 1. Best percentage change from baseline in tumor diameter (as-treated population).
Longitudinal tumor size was analyzed using a non-linear mixed-effects model to determine tumor growth rate constants and time to growth. Cohort A, n = 26; Cohort B, n = 20; Cohort C, n = 22. CR/PR, complete response/partial response.
Fig. 2. Duration of response (as-treated population).
Fig. 2. Duration of response (as-treated population).
Vertical lines indicate planned 12 months of treatment with durvalumab. Cohort A, n = 26; Cohort B, n = 20; Cohort C, n = 22.
Fig. 3. RNA-sequenced gene expression profiling, CD8+…
Fig. 3. RNA-sequenced gene expression profiling, CD8+ T-cell infiltration by IHC, and IFNγ levels in circulating blood.
Cohort A, n = 13; Cohort B, n = 12; Cohort C, n = 5. CR complete response, IHC immunohistochemistry, IFN-γ interferon-gamma, PR partial response, PD progressive disease, SD stable disease, TIL tumor-infiltrating lymphocyte.

References

    1. Hamid O, et al. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N. Engl. J. Med. 2013;369:134–144. doi: 10.1056/NEJMoa1305133.
    1. Robert C, et al. Nivolumab in previously untreated melanoma without BRAF mutation. N. Engl. J. Med. 2015;372:320–330. doi: 10.1056/NEJMoa1412082.
    1. Robert C, et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N. Engl. J. Med. 2011;364:2517–2526. doi: 10.1056/NEJMoa1104621.
    1. Schachter J, et al. Pembrolizumab versus ipilimumab for advanced melanoma: final overall survival results of a multicentre, randomised, open-label phase 3 study (KEYNOTE-006) Lancet. 2017;390:1853–1862. doi: 10.1016/S0140-6736(17)31601-X.
    1. Hodi FS, et al. Nivolumab plus ipilimumab or nivolumab alone versus ipilimumab alone in advanced melanoma (CheckMate 067): 4-year outcomes of a multicentre, randomised, phase 3 trial. Lancet Oncol. 2018;19:1480–1492. doi: 10.1016/S1470-2045(18)30700-9.
    1. McArthur GA, et al. Marked, homogeneous, and early [18F]fluorodeoxyglucose-positron emission tomography responses to vemurafenib in BRAF-mutant advanced melanoma. J. Clin. Oncol. 2012;30:1628–1634. doi: 10.1200/JCO.2011.39.1938.
    1. Long GV, et al. Dabrafenib and trametinib versus dabrafenib and placebo for Val600 BRAF-mutant melanoma: a multicentre, double-blind, phase 3 randomised controlled trial. Lancet. 2015;386:444–451. doi: 10.1016/S0140-6736(15)60898-4.
    1. Larkin J, et al. Combined vemurafenib and cobimetinib in BRAF-mutated melanoma. N. Engl. J. Med. 2014;371:1867–1876. doi: 10.1056/NEJMoa1408868.
    1. Dummer R, et al. Encorafenib plus binimetinib versus vemurafenib or encorafenib in patients with BRAF-mutant melanoma (COLUMBUS): a multicentre, open-label, randomised phase 3 trial. Lancet Oncol. 2018;19:603–615. doi: 10.1016/S1470-2045(18)30142-6.
    1. Hu-Lieskovan S, et al. Improved antitumor activity of immunotherapy with BRAF and MEK inhibitors in BRAF(V600E) melanoma. Sci. Transl. Med. 2015;7:279ra41. doi: 10.1126/scitranslmed.aaa4691.
    1. Deken MA, et al. Targeting the MAPK and PI3K pathways in combination with PD1 blockade in melanoma. Oncoimmunology. 2016;5:e1238557. doi: 10.1080/2162402X.2016.1238557.
    1. Knight DA, et al. Host immunity contributes to the anti-melanoma activity of BRAF inhibitors. J. Clin. Invest. 2013;123:1371–1381. doi: 10.1172/JCI66236.
    1. Wilmott JS, et al. Selective BRAF inhibitors induce marked T-cell infiltration into human metastatic melanoma. Clin. Cancer Res. 2012;18:1386–1394. doi: 10.1158/1078-0432.CCR-11-2479.
    1. Sullivan RJ, et al. Atezolizumab plus cobimetinib and vemurafenib in BRAF-mutated melanoma patients. Nat. Med. 2019;25:929–935. doi: 10.1038/s41591-019-0474-7.
    1. Ascierto PA, et al. Dabrafenib, trametinib and pembrolizumab or placebo in BRAF-mutant melanoma. Nat. Med. 2019;25:941–946. doi: 10.1038/s41591-019-0448-9.
    1. Ribas A, et al. Combined BRAF and MEK inhibition with PD-1 blockade immunotherapy in BRAF-mutant melanoma. Nat. Med. 2019;25:936–940. doi: 10.1038/s41591-019-0476-5.
    1. Gutzmer R, et al. Atezolizumab, vemurafenib, and cobimetinib as first-line treatment for unresectable advanced BRAFV600 mutationpositive melanoma (IMspire150): primary analysis of the randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2020;395:1835–1844. doi: 10.1016/S0140-6736(20)30934-X.
    1. Baverel PG, et al. Population pharmacokinetics of durvalumab in cancer patients and association with longitudinal biomarkers of disease status. Clin. Pharmacol. Ther. 2018;103:631–642. doi: 10.1002/cpt.982.
    1. Falchook GS, et al. Dose selection, pharmacokinetics, and pharmacodynamics of BRAF inhibitor dabrafenib (GSK2118436) Clin. Cancer Res. 2014;20:4449–4458. doi: 10.1158/1078-0432.CCR-14-0887.
    1. Infante JR, et al. Safety, pharmacokinetic, pharmacodynamic, and efficacy data for the oral MEK inhibitor trametinib: a phase 1 dose-escalation trial. Lancet Oncol. 2012;13:773–781. doi: 10.1016/S1470-2045(12)70270-X.
    1. Ouellet D, et al. Population pharmacokinetics of dabrafenib, a BRAF inhibitor: effect of dose, time, covariates, and relationship with its metabolites. J. Clin. Pharmacol. 2014;54:696–706. doi: 10.1002/jcph.263.
    1. Ouellet D, et al. Population pharmacokinetics and exposure–response of trametinib, a MEK inhibitor, in patients with BRAF V600 mutation positive melanoma. Cancer Chemother. Pharmacol. 2016;77:807–817. doi: 10.1007/s00280-016-2993-y.
    1. Tumeh PC, et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 2014;515:568–571. doi: 10.1038/nature13954.
    1. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15:550. doi: 10.1186/s13059-014-0550-8.
    1. Robert C, et al. Improved overall survival in melanoma with combined dabrafenib and trametinib. N. Engl. J. Med. 2015;372:30–39. doi: 10.1056/NEJMoa1412690.
    1. Urugel S, et al. Survival of patients with advanced metastatic melanoma: the impact of novel therapies-update 2017. Eur. J. Cancer. 2017;83:247–257. doi: 10.1016/j.ejca.2017.06.028.
    1. Hu-Lieskovan S, Robert L, Homet Moreno B, Ribas A. Combining targeted therapy with immunotherapy in BRAF-mutant melanoma: promise and challenges. J. Clin. Oncol. 2014;32:2248–2254. doi: 10.1200/JCO.2013.52.1377.
    1. Robert C, et al. Pembrolizumab versus ipilimumab in advanced melanoma. N. Engl. J. Med. 2015;372:2521–2532. doi: 10.1056/NEJMoa1503093.
    1. Flaherty KT, et al. Improved survival with MEK inhibition in BRAF-mutated melanoma. N. Eng. J. Med. 2012;367:107–114. doi: 10.1056/NEJMoa1203421.
    1. Schwartz LH, et al. RECIST 1.1-Update and clarification: from the RECIST committee. Eur. J. Cancer. 2016;62:132–137. doi: 10.1016/j.ejca.2016.03.081.
    1. Nishino M, et al. Developing a common language for tumor response to immunotherapy: immune-related response criteria using unidimensional measurements. Clin. Cancer Res. 2013;19:3936–3943. doi: 10.1158/1078-0432.CCR-13-0895.
    1. Del Curatolo A, et al. Therapeutic potential of combined BRAF/MEK blockade in BRAF-wild type preclinical tumor models. J. Exp. Clin. Cancer Res. 2018;37:140. doi: 10.1186/s13046-018-0820-5.
    1. Krepler C, et al. Personalized preclinical trials in BRAF inhibitor-resistant patient-derived xenograft models identify second-line combination therapies. Clin. Cancer Res. 2016;22:1592–1602. doi: 10.1158/1078-0432.CCR-15-1762.
    1. Kim D, Langmead B, Salzberg SL. HISAT: a fast spliced aligner with low memory requirements. Nat. Methods. 2015;12:357–360. doi: 10.1038/nmeth.3317.
    1. Anders S, Pyl PT, Huber W. HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics. 2015;31:166–169. doi: 10.1093/bioinformatics/btu638.
    1. Wickham, H. ggplot2: Elegant Graphics for Data Analysis. 2nd edn (Springer Nature, Switzerland, 2016).
    1. Healthwise Staff, University of Michigan Health. Lactic Acid Dehydrogenase, (2019).

Source: PubMed

3
Abonnieren