Spartalizumab or placebo in combination with dabrafenib and trametinib in patients with BRAF V600-mutant melanoma: exploratory biomarker analyses from a randomized phase 3 trial (COMBI-i)

Hussein A Tawbi, Caroline Robert, Jan C Brase, Daniel Gusenleitner, Eduard Gasal, James Garrett, Alexander Savchenko, Güllü Görgün, Keith T Flaherty, Antoni Ribas, Reinhard Dummer, Dirk Schadendorf, Georgina V Long, Paul D Nathan, Paolo A Ascierto, Hussein A Tawbi, Caroline Robert, Jan C Brase, Daniel Gusenleitner, Eduard Gasal, James Garrett, Alexander Savchenko, Güllü Görgün, Keith T Flaherty, Antoni Ribas, Reinhard Dummer, Dirk Schadendorf, Georgina V Long, Paul D Nathan, Paolo A Ascierto

Abstract

Background: The randomized phase 3 COMBI-i trial did not meet its primary endpoint of improved progression-free survival (PFS) with spartalizumab plus dabrafenib and trametinib (sparta-DabTram) vs placebo plus dabrafenib and trametinib (placebo-DabTram) in the overall population of patients with unresectable/metastatic BRAF V600-mutant melanoma. This prespecified exploratory biomarker analysis was performed to identify subgroups that may derive greater treatment benefit from sparta-DabTram.

Methods: In COMBI-i (ClinicalTrials.gov, NCT02967692), 532 patients received spartalizumab 400 mg intravenously every 4 weeks plus dabrafenib 150 mg orally two times daily and trametinib 2 mg orally one time daily or placebo-DabTram. Baseline/on-treatment pharmacodynamic markers were assessed via flow cytometry-based immunophenotyping and plasma cytokine profiling. Baseline programmed death ligand 1 (PD-L1) status and T-cell phenotype were assessed via immunohistochemistry; BRAF V600 mutation type, tumor mutational burden (TMB), and circulating tumor DNA (ctDNA) via DNA sequencing; gene expression signatures via RNA sequencing; and CD4+/CD8+ T-cell ratio via immunophenotyping.

Results: Extensive biomarker analyses were possible in approximately 64% to 90% of the intention-to-treat population, depending on sample availability and assay. Subgroups based on PD-L1 status/TMB or T-cell inflammation did not show significant differences in PFS benefit with sparta-DabTram vs placebo-DabTram, although T-cell inflammation was prognostic across treatment arms. Subgroups defined by BRAF V600K mutation (HR 0.45 (95% CI 0.21 to 0.99)), detectable ctDNA shedding (HR 0.75 (95% CI 0.58 to 0.96)), or CD4+/CD8+ ratio above median (HR 0.58 (95% CI 0.40 to 0.84)) derived greater PFS benefit with sparta-DabTram vs placebo-DabTram. In a multivariate analysis, ctDNA emerged as strongly prognostic (p=0.007), while its predictive trend did not reach significance; in contrast, CD4+/CD8+ ratio was strongly predictive (interaction p=0.0131).

Conclusions: These results support the feasibility of large-scale comprehensive biomarker analyses in the context of a global phase 3 study. T-cell inflammation was prognostic but not predictive of sparta-DabTram benefit, as patients with high T-cell inflammation already benefit from targeted therapy alone. Baseline ctDNA shedding also emerged as a strong independent prognostic variable, with predictive trends consistent with established measures of disease burden such as lactate dehydrogenase levels. CD4+/CD8+ T-cell ratio was significantly predictive of PFS benefit with sparta-DabTram but requires further validation as a biomarker in melanoma. Taken together with previous observations, further study of checkpoint inhibitor plus targeted therapy combination in patients with higher disease burden may be warranted.

Trial registration number: NCT02967692.

Keywords: Drug Therapy, Combination; Melanoma; Tumor Biomarkers.

Conflict of interest statement

Competing interests: HAT reports personal consulting fees and institutional research support from Novartis; institutional research support and personal consulting fees from Merck, Bristol Myers Squibb, and Genentech; institutional research support from GSK and Celgene; and consulting fees from Eisai and Iovance. CR reports consulting fees from and advisory board participation with Bristol Myers Squibb, Roche, Pierre Fabre, Novartis, Amgen, Sanofi, Merck, Merck Sharp & Dohme, and AstraZeneca. JCB reports stock ownership of and employment with Novartis; also, he is a coinventor on a patent application related to reported biomarker subgroups of interest. DG reports employment with Novartis and that they are a coinventor on a patent application related to reported biomarker subgroups of interest. EG reports stock ownership of and employment with Novartis. JG, AS, and GG report employment with Novartis. KTF reports grants or contracts from Novartis and Sanofi; consulting fees from, advisory board participation with, stock ownership of, and a leadership or fiduciary role with Clovis Oncology and Strata Oncology; consulting fees from and a leadership or fiduciary role with Vivid Biosciences and Checkmate Pharmaceuticals; consulting fees from, advisory board participation with, and stock ownership of X4 Pharmaceuticals, PIC Therapeutics, Sanofi, Amgen, Asana, Adaptimmune, Fount, Aeglea, Shattuck Labs, Tolero, Apricity, Oncoceutics, FogPharma, Neon, Tvardi, xCures, Monopteros, and Vibliome; stock ownership of and a leadership or fiduciary role with Loxo Oncology; and consulting fees from Lilly, Novartis, Genentech, Bristol Myers Squibb, Merck, Takeda, Verastem, Boston Biomedical, Pierre Fabre, and Debiopharm. AR reports clinical trial funding and consulting fees from and advisory board participation with Novartis; consulting fees from Amgen, Bristol Myers Squibb, Chugai, Genentech, Merck, Roche, Sanofi, and Vedanta; advisory board participation with and stock ownership of Advaxis, Apricity, Arcus, Compugen, CytomX, Five Prime, Highlight Therapeutics, ImaginAb, IsoPlexis, Kalthera, Kite-Gilead, Merus, PACT Pharma, RAPT, Rgenix, and Tango; and research funding from Agilent and Bristol Myers Squibb through Stand Up To Cancer (SU2C). RD reports intermittent, project-focused consulting fees from and advisory relationships with Novartis, Merck Sharp & Dohme, Bristol Myers Squibb, Roche, Amgen, Takeda, Pierre Fabre, Sun Pharma, Sanofi, CatalYm, Second Genome, Regeneron, Alligator, MaxiVAX SA, and touchlME. DS reports clinical trial funding to their institution from Novartis; institutional research grants and personal consulting fees, payments or honoraria, and support for attending meetings/travel from and advisory board participation with Novartis and Bristol Myers Squibb; institutional research grants and personal consulting fees and support for attending meetings/travel from and advisory board participation with Amgen; consulting fees, payments or honoraria, and support for attending meetings/travel from and advisory board participation with Pierre Fabre, Sanofi-Genzyme, and Merck Serono; consulting fees and support for attending meetings/travel from and advisory board participation with Merck Sharp & Dohme, Roche, Incyte, Array BioPharma, Pfizer, Regeneron, 4SC, InflaRx, NeraCare, Ultimovacs, Sun Pharma, Philogen, Immunocore, and Sandoz-Hexal; and leadership or fiduciary roles in the Dermatologic Cooperative Oncology Group, German Cancer Society, Hilfe-Stiftung, Deutsche Hautkrebsstiftung, NVKH eV, and EUMelaReg. GVL reports consulting fees from and advisory board participation with Aduro Biotech, Amgen, Array BioPharma, Boehringer Ingelheim, Bristol Myers Squibb, Hexel AG, Highlight Therapeutics, Merck Sharp & Dohme, Novartis Pharma AG, OncoSec, Pierre Fabre, QBiotics Group, Regeneron, SkylineDx BV, and Specialised Therapeutics Australia Pty Ltd; and personal payments or honoraria from Bristol Myers Squibb and Pierre Fabre. PDN reports consulting fees from AstraZeneca, Bristol Myers Squibb, Immunocore, Ipsen, Merck Sharp & Dohme, Merck, Novartis, Pfizer, and 4SC; personal payments or honoraria from Novartis, Merck, and Pfizer; and support for meeting attendance/travel from Bristol Myers Squibb. PAA reports research funding from, consulting fees from, and advisory board participation with Bristol Myers Squibb, Roche Genentech, Array BioPharma, and Sanofi; consulting fees from and advisory board participation with Novartis, Merck Serono, MedImmune, Sun Pharma, Idera, Sandoz, and 4SC; consulting fees from Italfarmaco, Pfizer, OncoSec, Takis, and Lunaphore; advisory board participation with Pierre Fabre, Incyte, AstraZeneca, Syndax, Ultimovacs, Immunocore, Alkermes, Nektar, Boehringer Ingelheim, Eisai, Regeneron, Daiichi Sankyo, Nouscom, and Seagen; consulting fees from, advisory board participation with, and support for meeting attendance/travel from Merck Sharp & Dohme; and leadership or fiduciary roles with the Society for Immunotherapy of Cancer, Society for Melanoma Research, Melanoma Foundation, and the Campania Society of Immunotherapy of Cancer. All authors acknowledge funding for medical writing support by ArticulateScience from Novartis, related to the present study.

© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY. Published by BMJ.

Figures

Figure 1
Figure 1
Characterization (A) and prognostic impact (B and C) of T-cell phenotypes. Representative samples of the inflamed, excluded, and desert T-cell phenotypes derived from digital pathology immunohistochemistry of tumor-infiltrating and stroma-infiltrating lymphocytes are shown in (A). Based on average CD8 density values, the top third of tumors were defined as inflamed; the bottom third of stroma and other samples were defined as desert; and samples between these thresholds were defined as excluded. Shown in (B and C) are Kaplan-Meier estimates of progression-free survival based on these phenotypes in the placebo-DabTram (B; inflamed, n=73; excluded, n=72; desert, n=62) and sparta-DabTram (C; inflamed, n=66; excluded, n=91; desert, n=52) treatment arms. CD, cluster of differentiation; placebo-DabTram, placebo plus dabrafenib and trametinib; sparta-DabTram, spartalizumab plus dabrafenib and trametinib.
Figure 2
Figure 2
Immunophenotyping of peripheral blood mononuclear cells using markers for T-cell activation and proliferation (N=323) and cytokine profiling (N=468) of plasma samples taken at baseline and after 4 weeks of treatment. Shown are proliferating CD8+/PD-1+ T cells (A), activated cytotoxic CD8+ T cells (B), and plasma IFN-γ (C). CD, cluster of differentiation; HLA, human leukocyte antigen; IFN, interferon; PD-1, programmed death receptor 1; placebo-DabTram, placebo plus dabrafenib and trametinib; sparta-DabTram, spartalizumab plus dabrafenib and trametinib.
Figure 3
Figure 3
Progression-free survival based on baseline peripheral CD4+/CD8+ T cell ratios. Shown are Kaplan-Meier estimates of progression-free survival in patients randomized to either the sparta-DabTram or placebo-DabTram arm with peripheral blood mononuclear cell samples reflecting baseline CD4+/CD8+ T-cell ratios at or above the median (N=204) (A) or below the median (N=204) (B) value of 2.9 at baseline. CD, cluster of differentiation; placebo-DabTram, placebo plus dabrafenib and trametinib; sparta-DabTram, spartalizumab plus dabrafenib and trametinib.
Figure 4
Figure 4
Predictive and prognostic value of baseline and on-treatment ctDNA shedding. (A) Kaplan-Meier estimates of progression-free survival (left) and overall survival (right) based on baseline and on-treatment ctDNA shedding in either the placebo-DabTram (top) or sparta-DabTram (bottom) arm. ‘No Shed’ indicates no ctDNA shedding observed at baseline or week 8 (placebo-DabTram, n=53; sparta-DabTram, n=56), ‘Loss at W8’ indicates shedding observed at baseline but not at week 8 (placebo-DabTram, n=122; sparta-DabTram, n=136), and ‘Shed at W8’ indicates shedding observed at both baseline and week 8 (placebo-DabTram, n=27; sparta-DabTram, n=16). (B) Kaplan-Meier estimates of progression-free survival (left) and overall survival (right) based on treatment with placebo-DabTram or sparta-DabTram in patients without (top; N=138) or with (bottom; N=342) baseline ctDNA shedding. ctDNA, circulating tumor DNA; placebo-DabTram, placebo plus dabrafenib and trametinib; sparta-DabTram, spartalizumab plus dabrafenib and trametinib.

References

    1. Ascierto PA, Dummer R, Gogas HJ, et al. . Update on tolerability and overall survival in COLUMBUS: landmark analysis of a randomised phase 3 trial of encorafenib plus binimetinib vs vemurafenib or encorafenib in patients with BRAF V600-mutant melanoma. Eur J Cancer 2020;126:33–44. 10.1016/j.ejca.2019.11.016
    1. Larkin J, Chiarion-Sileni V, Gonzalez R, et al. . Five-year survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med 2019;381:1535–46. 10.1056/NEJMoa1910836
    1. Robert C, Grob JJ, Stroyakovskiy D, et al. . Five-year outcomes with dabrafenib plus trametinib in metastatic melanoma. N Engl J Med 2019;381:626–36. 10.1056/NEJMoa1904059
    1. Robert C, Ribas A, Schachter J, et al. . Pembrolizumab versus ipilimumab in advanced melanoma (KEYNOTE-006): post-hoc 5-year results from an open-label, multicentre, randomised, controlled, phase 3 study. Lancet Oncol 2019;20:1239–51. 10.1016/S1470-2045(19)30388-2
    1. Ascierto PA, Dréno B, Larkin J, et al. . 5-year outcomes with cobimetinib plus vemurafenib in BRAFV600 mutation-positive advanced melanoma: extended follow-up of the coBRIM study. Clin Cancer Res 2021;27:5225–35. 10.1158/1078-0432.CCR-21-0809
    1. Ascierto PA, Dummer R. Immunological effects of BRAF+MEK inhibition. Oncoimmunology 2018;7:e1468955. 10.1080/2162402X.2018.1468955
    1. Wilmott JS, Long GV, Howle JR, et al. . Selective BRAF inhibitors induce marked T-cell infiltration into human metastatic melanoma. Clin Cancer Res 2012;18:1386–94. 10.1158/1078-0432.CCR-11-2479
    1. Frederick DT, Piris A, Cogdill AP, et al. . BRAF inhibition is associated with enhanced melanoma antigen expression and a more favorable tumor microenvironment in patients with metastatic melanoma. Clin Cancer Res 2013;19:1225–31. 10.1158/1078-0432.CCR-12-1630
    1. Ferrucci PF, Di Giacomo AM, Del Vecchio M, et al. . KEYNOTE-022 part 3: a randomized, double-blind, phase 2 study of pembrolizumab, dabrafenib, and trametinib in BRAF-mutant melanoma. J Immunother Cancer 2020;8:e001806. 10.1136/jitc-2020-001806
    1. Gutzmer R, Stroyakovskiy D, Gogas H, et al. . Atezolizumab, vemurafenib, and cobimetinib as first-line treatment for unresectable advanced BRAFV600 mutation-positive melanoma (IMspire150): primary analysis of the randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2020;395:1835–44. 10.1016/S0140-6736(20)30934-X
    1. Dummer R, Long GV, Robert C, et al. . Randomized phase III trial evaluating spartalizumab plus dabrafenib and trametinib for BRAF V600–mutant unresectable or metastatic melanoma. J Clin Oncol 2022;40:1428–38. 10.1200/JCO.21.01601
    1. Dummer R, Lebbé C, Atkinson V, et al. . Combined PD-1, BRAF and MEK inhibition in advanced BRAF-mutant melanoma: safety run-in and biomarker cohorts of COMBI-i. Nat Med 2020;26:1557–63. 10.1038/s41591-020-1082-2
    1. Liberzon A, Birger C, Thorvaldsdóttir H, et al. . The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst 2015;1:417–25. 10.1016/j.cels.2015.12.004
    1. Subramanian A, Tamayo P, Mootha VK, et al. . Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A 2005;102:15545–50. 10.1073/pnas.0506580102
    1. Cristescu R, Mogg R, Ayers M, et al. . Pan-tumor genomic biomarkers for PD-1 checkpoint blockade-based immunotherapy. Science 2018;362:eaar3593. 10.1126/science.aar3593
    1. Ribas A, Robert C, Schachter J. Tumor mutational burden (TMB), T cell-inflamed gene expression profile (GEP) and PD-L1 are independently associated with response to pembrolizumab (pembro) in patients with advanced melanoma in the KEYNOTE (KN)-006 study [abstract]. Cancer Res 2019;79:4217.
    1. Huang AC, Postow MA, Orlowski RJ, et al. . T-cell invigoration to tumour burden ratio associated with anti-PD-1 response. Nature 2017;545:60–5. 10.1038/nature22079
    1. Robert C, Lewis KD, Gutzmer R. Biomarkers of treatment benefit with atezolizumab plus vemurafenib plus cobimetinib in BRAFV600 mutation–positive melanoma. Ann Oncol 2022;33:544–55.
    1. Lee JH, Long GV, Boyd S, et al. . Circulating tumour DNA predicts response to anti-PD1 antibodies in metastatic melanoma. Ann Oncol 2017;28:1130–6. 10.1093/annonc/mdx026
    1. Santiago-Walker A, Gagnon R, Mazumdar J, et al. . Correlation of BRAF mutation status in circulating-free DNA and tumor and association with clinical outcome across four BRAFi and MEKi clinical trials. Clin Cancer Res 2016;22:567–74. 10.1158/1078-0432.CCR-15-0321
    1. Syeda MM, Wiggins JM, Corless BC, et al. . Circulating tumour DNA in patients with advanced melanoma treated with dabrafenib or dabrafenib plus trametinib: a clinical validation study. Lancet Oncol 2021;22:370–80. 10.1016/S1470-2045(20)30726-9
    1. Das D, Sarkar B, Mukhopadhyay S, et al. . An altered ratio of CD4+ and CD8+ T lymphocytes in cervical cancer tissues and peripheral blood – a prognostic clue? Asian Pac J Cancer Prev 2018;19:471–8. 10.22034/APJCP.2018.19.2.471
    1. Waki K, Kawano K, Tsuda N, et al. . CD4/CD8 ratio is a prognostic factor in IgG nonresponders among peptide vaccine-treated ovarian cancer patients. Cancer Sci 2020;111:1124–31. 10.1111/cas.14349
    1. Yang J, Xu J, E Y, et al. . Predictive and prognostic value of circulating blood lymphocyte subsets in metastatic breast cancer. Cancer Med 2019;8:492–500. 10.1002/cam4.1891
    1. Ariyan CE, Brady MS, Siegelbaum RH, et al. . Robust antitumor responses result from local chemotherapy and CTLA-4 blockade. Cancer Immunol Res 2018;6:189–200. 10.1158/2326-6066.CIR-17-0356
    1. Tang C, Welsh JW, de Groot P, et al. . Ipilimumab with stereotactic ablative radiation therapy: phase I results and immunologic correlates from peripheral T cells. Clin Cancer Res 2017;23:1388–96. 10.1158/1078-0432.CCR-16-1432
    1. Pires da Silva I, Wang KYX, Wilmott JS, et al. . Distinct molecular profiles and immunotherapy treatment outcomes of V600E and V600K BRAF-mutant melanoma. Clin Cancer Res 2019;25:1272–9. 10.1158/1078-0432.CCR-18-1680
    1. Daud AI, Wolchok JD, Robert C, et al. . Programmed death-ligand 1 expression and response to the anti-programmed death 1 antibody pembrolizumab in melanoma. J Clin Oncol 2016;34:4102–9. 10.1200/JCO.2016.67.2477
    1. Hodi FS, Wolchok JD, Schadendorf D, et al. . TMB and inflammatory gene expression associated with clinical outcomes following immunotherapy in advanced melanoma. Cancer Immunol Res 2021;9:1202–13. 10.1158/2326-6066.CIR-20-0983
    1. Dummer R, Brase JC, Garrett J, et al. . Adjuvant dabrafenib plus trametinib versus placebo in patients with resected, BRAFV600-mutant, stage III melanoma (COMBI-AD): exploratory biomarker analyses from a randomised, phase 3 trial. Lancet Oncol 2020;21:358–72. 10.1016/S1470-2045(20)30062-0

Source: PubMed

3
Předplatit