A monocentric phase I study of vemurafenib plus cobimetinib plus PEG-interferon (VEMUPLINT) in advanced melanoma patients harboring the V600BRAF mutation

Ester Simeone, Giosuè Scognamiglio, Mariaelena Capone, Diana Giannarelli, Antonio M Grimaldi, Domenico Mallardo, Gabriele Madonna, Marcello Curvietto, Assunta Esposito, Fabio Sandomenico, Francesco Sabbatino, Nicholas L Bayless, Sarah Warren, SuFey Ong, Gerardo Botti, Keith T Flaherty, Soldano Ferrone, Paolo A Ascierto, Ester Simeone, Giosuè Scognamiglio, Mariaelena Capone, Diana Giannarelli, Antonio M Grimaldi, Domenico Mallardo, Gabriele Madonna, Marcello Curvietto, Assunta Esposito, Fabio Sandomenico, Francesco Sabbatino, Nicholas L Bayless, Sarah Warren, SuFey Ong, Gerardo Botti, Keith T Flaherty, Soldano Ferrone, Paolo A Ascierto

Abstract

Background: Studies carried out in vitro and in a mouse model have shown that BRAF inhibitors enhance the effects of IFN-α on BRAFV600E melanoma cells through the inhibition of ERK. Therefore, the combination of vemurafenib and IFN-α in patients with BRAFV600E melanoma may provide therapeutic benefits; MEK inhibition may prevent the reactivation of the MAPK pathway induced by BRAF inhibitor resistance.

Patients and methods: In a phase I study, adult patients with advanced BRAFV600-mutated melanoma were treated with vemurafenib + PEG-IFN-α-2b or vemurafenib + cobimetinib + PEG-IFN-α-2b, to assess the safety of the combination and the upregulation of IFN-α/β receptor-1 (IFNAR1).

Results: Eight patients were treated; 59 adverse events with four serious ones (three related to study treatments) were reported. Patients with a pre-treatment IFNAR1 expression on ≤ 35% melanoma cells had a median progression-free survival of 12.0 months (range: 5.6-18.4 months) and a median overall survival of 31.0 months (range: 19.8-42.2 months), while patients with a pre-treatment IFNAR1 expression on > 35% of melanoma cells had a median progression-free survival of 4.0 months (range: 0-8.8; p = 0.03), and a median overall survival of 5 months (p = 0.02). Following treatment, responders had higher levels of growth-suppressor genes, including GAS1 and DUSP1, and genes involved in a metabolically robust immune response, including FAP.

Conclusion: Our study supports the overall safety of the vemurafenib + PEG-IFN-α-2b + cobimetinib combination. IFNAR1 expression levels correlated with response to treatment, including survival. Vemurafenib + PEG-IFN-α-2b + cobimetinib would have difficulty finding a niche in the current treatment scenario for advanced melanoma, but we speculate that our findings may contribute to identify subjects particularly responsive to treatment.

Trial registration: The study was registered at clinicaltrials.gov (NCT01959633). Registered 10 October 2013, https://ichgcp.net/clinical-trials-registry/NCT01959633.

Keywords: BRAF inhibitor; Interferon; MAP kinase; Malignant melanoma.

Conflict of interest statement

E. Simeone received honoraria from Bristol Myers Squibb, Novartis and Merck Sharp & Dohme. The other authors declare no potential competing interests. A.M.Grimaldi has/had an advisory and consultant role Bristol Myers Squibb, Merck Sharp & Dohme and Novartis. He received travel support from Bristol Myers Squibb, Merck Serono, Pierre Fabre, Roche-Genentech and Novartis. S Warren and S. Ong are/were employees and stockholders in NanoString Technologies. K.T. Flaherty served(s) on the Board of Directors of Clovis Oncology, Strata Oncology, Vivid Biosciences, Checkmate Pharmaceuticals, and Loxo Oncology; Corporate Advisory Board of X4 Pharmaceuticals; Scientific Advisory Boards of PIC Therapeutics, Sanofi, Amgen, Asana, Adaptimmune, Fount, Aeglea, Shattuck Labs, Tolero, Apricity, Oncoceutics, Fog Pharma, Neon, Tvardi, xCures, Monopteros, Vibliome, and consultant to Lilly, Novartis, Genentech, BMS, Merck, Takeda, Verastem, Boston Biomedical, Pierre Fabre, and Debiopharm; as well as research funding from Novartis and Sanofi. P.A. Ascierto has/had a consultant/advisory role for Bristol Myer Squibb, Roche-Genentech, Merck Sharp & Dohme, Array, Novartis, Merck Serono, Pierre Fabre, Incyte, NewLink Genetics, Genmab, Medimmune, AstraZeneca, Syndax, SunPharma, Sanofi, Idera, Ultimovacs, Sandoz, Immunocore, 4SC, Alkermes, Italfarmaco, Nektar, Boehringer-Ingelheim. He also received research funds from Bristol Myers-Squibb, Roche-Genentech, Array, and travel support from MSD.

Figures

Fig. 1
Fig. 1
Patient disposition. Group A: patients who received vemurafenib plus PEG-IFN-α-2b, group B: patients who received vemurafenib + cobimetinib + PEG-IFN-α-2b
Fig. 2
Fig. 2
Change in tumor size during the study. Group A: patients who received vemurafenib plus PEG-IFN-α-2b, group B: patients who received vemurafenib + cobimetinib + PEG-IFN-α-2b
Fig. 3
Fig. 3
Tumor response during study treatment according to baseline staging. a patients who received vemurafenib plus PEG-IFN-α-2b (patients 1 and 3 had durable response); b patients who received vemurafenib + cobimetinib + PEG-IFN-α-2b (patient 1 had durable response). CR: complete response, DP: disease progression, PR: partial response, SD: stable disease
Fig. 4
Fig. 4
Proportion of IFNAR1-positive cells in tumor samples, in the immunohistochemical analysis
Fig. 5
Fig. 5
PFS (a) and OS (b) according to the proportion of IFNAR1-positive cells in pretreatment specimens
Fig. 6
Fig. 6
Differential gene expression by Nanostring. a Pretreatment: Responders vs. non-responders. b Post-treatment vs. pretreatment in responders

References

    1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin. 2019;69(1):7–34. doi: 10.3322/caac.21551.
    1. Schadendorf D, Fisher DE, Garbe C, Gershenwald JE, Grob JJ, Halpern A, Herlyn M, Marchetti MA, McArthur G, Ribas A, Roesch A, Hauschild A. Melanoma. Nat Rev Dis Primers. 2015;1:15003. doi: 10.1038/nrdp.2015.3.
    1. Ugurel S, Röhmel J, Ascierto PA, Flaherty KT, Grob JJ, Hauschild A, Larkin J, Long GV, Lorigan P, McArthur GA, Ribas A, Robert C, Schadendorf D, Garbe C. 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. Ottaviano M, De Placido S, Ascierto PA. Recent success and limitations of immune checkpoint inhibitors for cancer: a lesson from melanoma. Virchows Arch. 2019;474(4):421–432. doi: 10.1007/s00428-019-02538-4.
    1. Ascierto PA, Dummer R. Immunological effects of BRAF+MEK inhibition. Oncoimmunology. 2018;7(9):e1468955. doi: 10.1080/2162402X.2018.1468955.
    1. Schummer P, Schilling B, Gesierich A. Long-term outcomes in BRAF-mutated melanoma treated with combined targeted therapy or immune checkpoint blockade: are we approaching a true cure? Am J Clin Dermatol. 2020 doi: 10.1007/s40257-020-00509-z.
    1. Pisanu ME, Maugeri-Saccà M, Fattore L, Bruschini S, De Vitis C, Tabbì E, Bellei B, Migliano E, Kovacs D, Camera E, Picardo M, Jakopin Z, Cippitelli C, Bartolazzi A, Raffa S, Torrisi MR, Fulciniti F, Ascierto PA, Ciliberto G, Mancini R. Inhibition of steeroyl-CoA desaturase 1 reverts BRAF and MEK inhibition-induced selection of cancer stem cells in BRAF-mutated melanoma. J Exp Clin Cancer Res. 2018;37(1):318. doi: 10.1186/s13046-018-0989-7.
    1. Junttila MR, de Sauvage FJ. Influence of tumour micro-environment heterogeneity on therapeutic response. Nature. 2013;501(7467):346–354. doi: 10.1038/nature12626.
    1. Wan PT, Garnett MJ, Roe SM, Lee S, Niculescu-Duvaz D, Good VM, Jones CM, Marshall CJ, Springer CJ, Barford D, Marais R, Cancer Genome Project Mechanism of activation of the RAF-ERK signaling pathway by oncogenic mutations of B-RAF. Cell. 2004;116(6):855–867. doi: 10.1016/s0092-8674(04)00215-6.
    1. Cohen C, Zavala-Pompa A, Sequeira JH, Shoji M, Sexton DG, Cotsonis G, Cerimele F, Govindarajan B, Macaron N, Arbiser JL. Mitogen-activated protein kinase activation is an early event in melanoma progression. Clin Cancer Res. 2002;8(12):3728–3733.
    1. Sapkota B, Hill CE, Pollack BP. Vemurafenib enhances MHC induction in BRAFV600E homozygous melanoma cells. Oncoimmunology. 2013;2(1):e22890. doi: 10.4161/onci.22890.
    1. Boni A, Cogdill AP, Dang P, Udayakumar D, Njauw CN, Sloss CM, Ferrone CR, Flaherty KT, Lawrence DP, Fisher DE, Tsao H, Wargo JA. Selective BRAFV600E inhibition enhances T-cell recognition of melanoma without affecting lymphocyte function. Cancer Res. 2010;70(13):5213–5219. doi: 10.1158/0008-5472.CAN-10-0118.
    1. Mimura K, Kua LF, Shiraishi K, Kee Siang L, Shabbir A, Komachi M, Suzuki Y, Nakano T, Yong WP, So J, Kono K. Inhibition of mitogen-activated protein kinase pathway can induce upregulation of human leukocyte antigen class I without PD-L1-upregulation in contrast to interferon-γ treatment. Cancer Sci. 2014;105(10):1236–1244. doi: 10.1111/cas.12503.
    1. Frederick DT, Piris A, Cogdill AP, Cooper ZA, Lezcano C, Ferrone CR, Mitra D, Boni A, Newton LP, Liu C, Peng W, Sullivan RJ, Lawrence DP, Hodi FS, Overwijk WW, Lizée G, Murphy GF, Hwu P, Flaherty KT, Fisher DE, Wargo JA. BRAF inhibition is associated with enhanced melanoma antigen expression and a more favorable tumor microenvironment in patients with metastatic melanoma. Clin Cancer Res. 2013;19(5):1225–1231. doi: 10.1158/1078-0432.CCR-12-1630.
    1. Sabbatino F, Wang Y, Scognamiglio G, Favoino E, Feldman SA, Villani V, Flaherty KT, Nota S, Giannarelli D, Simeone E, Anniciello AM, Palmieri G, Pepe S, Botti G, Ascierto PA, Ferrone CR, Ferrone S. Antitumor activity of BRAF inhibitor and IFNα combination in BRAF-mutant melanoma. J Natl Cancer Inst. 2016;108(7):djv435. doi: 10.1093/jnci/djv435.
    1. Liu J, Suresh Kumar KG, Yu D, Molton SA, McMahon M, Herlyn M, Thomas-Tikhonenko A, Fuchs SY. Oncogenic BRAF regulates beta-Trcp expression and NF-kappaB activity in human melanoma cells. Oncogene. 2007;26(13):1954–1958. doi: 10.1038/sj.onc.1209994.
    1. Taniguchi T, Takaoka A. A weak signal for strong responses: interferon-alpha/beta revisited. Nat Rev Mol Cell Biol. 2001;2(5):378–386. doi: 10.1038/35073080.
    1. Colamonici OR, Porterfield B, Domanski P, Handa RK, Flex S, Samuel CE, Pine R, Diaz MO. Ligand-independent anti-oncogenic activity of the alpha subunit of the type I interferon receptor. J Biol Chem. 1994;269(44):27275–27279. doi: 10.1016/S0021-9258(18)46980-3.
    1. Mocellin S, Pasquali S, Rossi CR, Nitti D. Interferon alpha adjuvant therapy in patients with high-risk melanoma: a systematic review and meta-analysis. J Natl Cancer Inst. 2010;102(7):493–501. doi: 10.1093/jnci/djq009.
    1. Spiegelman VS, Tang W, Chan AM, Igarashi M, Aaronson SA, Sassoon DA, Katoh M, Slaga TJ, Fuchs SY. Induction of homologue of Slimb ubiquitin ligase receptor by mitogen signaling. J Biol Chem. 2002;277(39):36624–36630. doi: 10.1074/jbc.M204524200.
    1. Kumar KG, Tang W, Ravindranath AK, Clark WA, Croze E, Fuchs SY. SCF(HOS) ubiquitin ligase mediates the ligand-induced down-regulation of the interferon-alpha receptor. EMBO J. 2003;22(20):5480–5490. doi: 10.1093/emboj/cdg524.
    1. McArthur G, Larkin J, Dréno B, Ascierto P, et al. 25LBA Impact of baseline genetic heterogeneities on progression-free survival (PFS) in patients (pts) with advanced BRAFV600-mutated melanoma treated with cobimetinib (COBI) + vemurafenib (VEM) in the phase 3 coBRIM study. Eur J Cancer. 2015;51(suppl 3):S722–S723. doi: 10.1016/S0959-8049(16)31945-1.
    1. Sernee MF, Ploegh HL, Schust DJ. Why certain antibodies cross-react with HLA-A and HLA-G: epitope mapping of two common MHC class I reagents. Mol Immunol. 1998;35:177–188. doi: 10.1016/S0161-5890(98)00026-1.
    1. Stam NJ, Spits H, Ploegh HL. Monoclonal antibodies raised against denatured HLA-B locus heavy chains permit biochemical characterization of certain HLA-C locus products. J Immunol. 1986;137(7):2299–2306.
    1. Perosa F, Luccarelli G, Prete M, Favoino E, Ferrone S, Dammacco F. Beta 2-microglobulin-free HLA class I heavy chain epitope mimicry by monoclonal antibody HC-10-specific peptide. J Immunol. 2003;171(4):1918–1926. doi: 10.4049/jimmunol.171.4.1918.
    1. Garrido F, Cabrera T, Accolla RS, Bensa JC, Bodmer W, Dohr G, Drouet M, Fauchet R, Ferrara GB, Ferrone S, Giacomini P, Kageshita T, Koopman L, Maio M, Marincola M, Mazzilli C, Morel PA, Murray A, Papasteriades CRH, Salvaneschi L, Stern PL, Ziegler A. HLA and cancer Charron D. eds. HLA. Genetic Diversity of HLA. Functional and Medical Implication, Vol. I: 445–452, EDK Paris 1997.
    1. Berghoff AS, Ricken G, Widhalm G, Rajky O, Dieckmann K, Birner P, Bartsch R, Höller C, Preusser M. Tumour-infiltrating lymphocytes and expression of programmed death ligand 1 (PD-L1) in melanoma brain metastases. Histopathology. 2015;66(2):289–299. doi: 10.1111/his.12537.
    1. Ascierto PA, Grimaldi AM, Acquavella N, Borgognoni L, Calabrò L, Cascinelli N, Cesano A, Del Vecchio M, Eggermont AM, Faries M, Ferrone S, Fox BA, Gajewski TF, Galon J, Gnjatic S, Gogas H, Kashani-Sabet M, Kaufman HL, Larkin J, Lo RS, Mantovani A, Margolin K, Melief C, McArthur G, Palmieri G, Puzanov I, Ribas A, Seliger B, Sosman J, Suenaert P, Tarhini AA, Trinchieri G, Vidal-Vanaclocha F, Wang E, Ciliberto G, Mozzillo N, Marincola FM, Thurin M. Future perspectives in melanoma research. Meeting report from the "Melanoma Bridge Napoli, December 2nd-4th 2012". J Transl Med. 2013;11:137. doi: 10.1186/1479-5876-11-137.
    1. McArthur GA, Chapman PB, Robert C, Larkin J, Haanen JB, Dummer R, Ribas A, Hogg D, Hamid O, Ascierto PA, Garbe C, Testori A, Maio M, Lorigan P, Lebbé C, Jouary T, Schadendorf D, O'Day SJ, Kirkwood JM, Eggermont AM, Dréno B, Sosman JA, Flaherty KT, Yin M, Caro I, Cheng S, Trunzer K, Hauschild A. Safety and efficacy of vemurafenib in BRAF(V600E) and BRAF(V600K) mutation-positive melanoma (BRIM-3): extended follow-up of a phase 3, randomised, open-label study. Lancet Oncol. 2014;15(3):323–332. doi: 10.1016/S1470-2045(14)70012-9.
    1. Ribas A, Hodi FS, Callahan M, Konto C, Wolchok J. Hepatotoxicity with combination of vemurafenib and ipilimumab. N Engl J Med. 2013;368(14):1365–1366. doi: 10.1056/NEJMc1302338.
    1. Minor DR, Puzanov I, Callahan MK, Hug BA, Hoos A. Severe gastrointestinal toxicity with administration of trametinib in combination with dabrafenib and ipilimumab. Pigment Cell Melanoma Res. 2015;28(5):611–612. doi: 10.1111/pcmr.12383.
    1. Yan Y, Wongchenko MJ, Robert C, Larkin J, Ascierto PA, Dréno B, Maio M, Garbe C, Chapman PB, Sosman JA, Shi Z, Koeppen H, Hsu JJ, Chang I, Caro I, Rooney I, McArthur GA, Ribas A. Genomic features of exceptional response in vemurafenib ± cobimetinib-treated patients with BRAFV600-mutated metastatic melanoma. Clin Cancer Res. 2019;25(11):3239–3246. doi: 10.1158/1078-0432.CCR-18-0720.
    1. Ascierto PA, Ferrucci PF, Fisher R, Del Vecchio M, Atkinson V, Schmidt H, Schachter J, Queirolo P, Long GV, Di Giacomo AM, Svane IM, Lotem M, Bar-Sela G, Couture F, Mookerjee B, Ghori R, Ibrahim N, Moreno BH, Ribas A. Dabrafenib, trametinib and pembrolizumab or placebo in BRAF-mutant melanoma. Nat Med. 2019;25(6):941–946. doi: 10.1038/s41591-019-0448-9.
    1. Ascierto PA, Ferrucci PF, Stephens R, Del Vecchio M, Atkinson V, Schmidt H, Schachter J, Queirolo P, Long GV, Di Giacomo AM, Svane I, Lotem M, Bar-Sela G, Couture F, Mookerjee BP, Ghori R, Ibrahim N, Homet Moreno B, Ribas A. KEYNOTE-022 part 3: phase II randomized study of 1L dabrafenib (D) and trametinib (T) plus pembrolizumab (Pembro) or placebo (PBO) for BRAF-mutant advanced melanoma. Ann Oncol. 2018;29(Supplement 8):viii442–viii466. doi: 10.1093/annonc/mdy289.

Source: PubMed

3
Abonner