Multicenter, double-blind, placebo-controlled trial of seviprotimut-L polyvalent melanoma vaccine in patients with post-resection melanoma at high risk of recurrence

Craig L Slingluff, Karl D Lewis, Robert Andtbacka, John Hyngstrom, Mohammed Milhem, Svetomir N Markovic, Tawnya Bowles, Omid Hamid, Leonel Hernandez-Aya, Joel Claveau, Sekwon Jang, Prejesh Philips, Shernan G Holtan, Montaser F Shaheen, Brendan Curti, William Schmidt, Marcus O Butler, Juan Paramo, Jose Lutzky, Arvinda Padmanabhan, Sajeve Thomas, Daniel Milton, Andrew Pecora, Takami Sato, Eddy Hsueh, Suprith Badarinath, John Keech, Sujith Kalmadi, Pallavi Kumar, Robert Weber, Edward Levine, Adam Berger, Anna Bar, J Thaddeus Beck, Jeffrey B Travers, Catalin Mihalcioiu, Brian Gastman, Peter Beitsch, Suthee Rapisuwon, John Glaspy, Edward C McCarron, Vinay Gupta, Deepti Behl, Brent Blumenstein, Joanna J Peterkin, Craig L Slingluff, Karl D Lewis, Robert Andtbacka, John Hyngstrom, Mohammed Milhem, Svetomir N Markovic, Tawnya Bowles, Omid Hamid, Leonel Hernandez-Aya, Joel Claveau, Sekwon Jang, Prejesh Philips, Shernan G Holtan, Montaser F Shaheen, Brendan Curti, William Schmidt, Marcus O Butler, Juan Paramo, Jose Lutzky, Arvinda Padmanabhan, Sajeve Thomas, Daniel Milton, Andrew Pecora, Takami Sato, Eddy Hsueh, Suprith Badarinath, John Keech, Sujith Kalmadi, Pallavi Kumar, Robert Weber, Edward Levine, Adam Berger, Anna Bar, J Thaddeus Beck, Jeffrey B Travers, Catalin Mihalcioiu, Brian Gastman, Peter Beitsch, Suthee Rapisuwon, John Glaspy, Edward C McCarron, Vinay Gupta, Deepti Behl, Brent Blumenstein, Joanna J Peterkin

Abstract

Background: Most patients with advanced melanomas relapse after checkpoint blockade therapy. Thus, immunotherapies are needed that can be applied safely early, in the adjuvant setting. Seviprotimut-L is a vaccine containing human melanoma antigens, plus alum. To assess the efficacy of seviprotimut-L, the Melanoma Antigen Vaccine Immunotherapy Study (MAVIS) was initiated as a three-part multicenter, double-blind, placebo-controlled phase III trial. Results from part B1 are reported here.

Methods: Patients with AJCC V.7 stage IIB-III cutaneous melanoma after resection were randomized 2:1, with stage stratification (IIB/C, IIIA, IIIB/C), to seviprotimut-L 40 mcg or placebo. Recurrence-free survival (RFS) was the primary endpoint. For an hypothesized HR of 0.625, one-sided alpha of 0.10, and power 80%, target enrollment was 325 patients.

Results: For randomized patients (n=347), arms were well-balanced, and treatment-emergent adverse events were similar for seviprotimut-L and placebo. For the primary intent-to-treat endpoint of RFS, the estimated HR was 0.881 (95% CI: 0.629 to 1.233), with stratified logrank p=0.46. However, estimated HRs were not uniform over the stage randomized strata, with HRs (95% CIs) for stages IIB/IIC, IIIA, IIIB/IIIC of 0.67 (95% CI: 0.37 to 1.19), 0.72 (95% CI: 0.35 to 1.50), and 1.19 (95% CI: 0.72 to 1.97), respectively. In the stage IIB/IIC stratum, the effect on RFS was greatest for patients <60 years old (HR=0.324 (95% CI: 0.121 to 0.864)) and those with ulcerated primary melanomas (HR=0.493 (95% CI: 0.255 to 0.952)).

Conclusions: Seviprotimut-L is very well tolerated. Exploratory efficacy model estimation supports further study in stage IIB/IIC patients, especially younger patients and those with ulcerated melanomas.

Trial registration number: NCT01546571.

Keywords: active; immunotherapy; melanoma; vaccination.

Conflict of interest statement

Competing interests: CLS discloses research funding to his University from Merck, Celldex, and GlaxoSmithKline; research support in kind to his University from Theraclion, 3M, Merck, and Celldex; Scientific Advisory Board role with Immatics (prior), and Curevac; principal investigator role for Polynoma with compensation to his university, related to this manuscript; and patent royalties as co-inventor of peptides for use in cancer vaccines (patents held by the UVA Licensing and Ventures Group). KDL received research funding from Polynoma. JH served on an advisory board for Nektar and BMS in 2020. SNM has received research grants from BMS and from Sorrento Pharma. TB is receiving time and effort support from Genentech for a melanoma prevention study and is the principal investigator for melanoma clinical studies sponsored by Genentech, Amgen, and Replimune. JC serves as consultant/speaker for Amgen, BMS, Merck, Novartis, Pfizer, Roche. SGH serves as an advisor to Incyte. BC has received honoraria from Merck (DSMB member), Clinigen (Ad board), Nektar (Ad Board), research support from Galectin Therapeutics, Clinigen. BMS (to Institution) and AstraZeneca (to Institution). MOB discloses roles on Advisory boards (Merck, BMS, Novartis, EMD Serono, Sanofi, Pfizer, Immunocore, GSK); safety Review Committees (GSK, Adaptimmune), and research funding from Merck and Takara Bio. JL has received research funding to his institution from BMS, Immunocore, Polynoma, Iovance and has participated in advisory boards from Regeneron and Array. DM directly is on speaker’s bureaus for Pfizer, Merck, Astellas, and SeaGen. TS serves on Advisory boards for Immunocore and Castle Biosciences. EH is on the speakers’ bureaus for Amgen and Castle Biosciences. SK is on the speakers’ bureau for Janssen, Pfizer, Roche/Genentech, AstraZeneca. RW is on a Speaker’s bureau for AstraZeneca. ABe has served as an advisor to Cardinal Health and Castle Biosciences. ABa is a consultant/speaker for Regeneron, Castle Biosciences, and receives research funding to her institution from Polynoma, Castle Biosciences, Pellepharm. CM has received grants, ad board and speaker fees from Novartis, BMS, Merck and Pfizer. VG is an investor in Medocity. DB: Research support- Natera; served on advisory boards for Merck, AstraZeneca, Novartis, Eli Lilly, Janssen, Oncocyte; speaker’s bureau-Janssen, Guardant. JJP is a paid consultant for Polynoma.

© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Recurrence-free survival (RFS) by arm, stage, and age. Kaplan-Meier estimates of RFS are plotted, with analysis results in the legend and risk set and censoring accumulation below the time axis. The designation ‘(cut-off)’ indicates possible data truncation based on designated cut-off date. (A) RFS by arm, stratified by stage, for the intent-to-treat (ITT) data set, (B) RFS by arm and stage for the ITT data set, (C) RFS by arm and age for the ITT data set, (D) RFS by arm and age for Stage IIB/C stratum. P values by logrank test. P value, HR and 95% CI are based on a univariate Cox regression model assuming proportional hazards with treatment, age, and treatment×age as factors, stratified for the randomisation stratification variable of disease stage.
Figure 2
Figure 2
Recurrence-free survival (RFS) by arm, age, and ulceration for stage IIB/IIC stratum. Kaplan-Meier estimates of RFS are plotted for stage IIB/IIC patients (A) by arm and ulceration, and (B) by arm, age and ulceration. Shown in (C) is a forest plot for stage IIB/IIC patients as a function of age, ulceration, and both. In (D) is the Kaplan-Meier estimate for RFS for stage IIB/IIC patients under age 60, by arm and ulceration. P values by logrank test.
Figure 3
Figure 3
Survival effect modification by arm, age, and stage. Kaplan-Meier estimates of overall survival (OS) are plotted (A) for all intent-to-treat (ITT) patients stratified by stage, by arm, (B) for all ITT patients by arm and stage, and (C) for all ITT patients by arm and age. Shown in (D) is a forest plot for all ITT patients by stage and by age. In (E) is the Kaplan-Meier estimate for OS for stage IIB/IIC patients by arm and age. P values by logrank test.

References

    1. Kirkwood JM, Manola J, Ibrahim J, et al. . A pooled analysis of eastern cooperative oncology group and intergroup trials of adjuvant high-dose interferon for melanoma. Clin Cancer Res 2004;10:1670–7. 10.1158/1078-0432.CCR-1103-3
    1. Swetter SM, Thompson JA, Albertini MR. Melanoma: cutaneous, version 2.2021. NCCN clinical practice guidelines in oncology: national comprehensive cancer network. Network NCC, 2021.
    1. Reynolds SR, Oratz R, Shapiro RL, et al. . Stimulation of CD8+ T cell responses to MAGE-3 and Melan A/MART-1 by immunization to a polyvalent melanoma vaccine. Int J Cancer 1997;72:972–6. 10.1002/(SICI)1097-0215(19970917)72:6&lt;972::AID-IJC9&gt;;2-M
    1. Reynolds SR, Celis E, Sette A, et al. . HLA-independent heterogeneity of CD8+ T cell responses to MAGE-3, Melan-A/MART-1, gp100, tyrosinase, MC1R, and TRP-2 in vaccine-treated melanoma patients. J Immunol 1998;161:6970–6.
    1. Miller K, Abeles G, Oratz R, et al. . Improved survival of patients with melanoma with an antibody response to immunization to a polyvalent melanoma vaccine. Cancer 1995;75:495–502. 10.1002/1097-0142(19950115)75:2&lt;495::AID-CNCR2820750212&gt;;2-S
    1. Reynolds SR, Celis E, Sette A, et al. . Identification of HLA-A*03, A*11 and B*07-restricted melanoma-associated peptides that are immunogenic in vivo by vaccine-induced immune response (VIIR) analysis. J Immunol Methods 2000;244:59–67. 10.1016/S0022-1759(00)00254-4
    1. Bystryn JC, Zeleniuch-Jacquotte A, Oratz R, et al. . Double-blind trial of a polyvalent, shed-antigen, melanoma vaccine. Clin Cancer Res 2001;7:1882–7.
    1. Faries MB, Thompson JF, Cochran AJ, et al. . Completion dissection or observation for sentinel-node metastasis in melanoma. N Engl J Med 2017;376:2211–22. 10.1056/NEJMoa1613210
    1. Gabellini C, Trisciuoglio D, Desideri M, et al. . Functional activity of CXCL8 receptors, CXCR1 and CXCR2, on human malignant melanoma progression. Eur J Cancer 2009;45:2618–27. 10.1016/j.ejca.2009.07.007
    1. Korch C, Hall EM, Dirks WG, et al. . Authentication of M14 melanoma cell line proves misidentification of MDA-MB-435 breast cancer cell line. Int J Cancer 2018;142:561–72. 10.1002/ijc.31067
    1. Carey TE, Takahashi T, Resnick LA, et al. . Cell surface antigens of human malignant melanoma: mixed hemadsorption assays for humoral immunity to cultured autologous melanoma cells. Proc Natl Acad Sci U S A 1976;73:3278–82. 10.1073/pnas.73.9.3278
    1. Xing F, Persaud Y, Pratilas CA, et al. . Concurrent loss of the PTEN and RB1 tumor suppressors attenuates RAF dependence in melanomas harboring (V600E)BRAF. Oncogene 2012;31:446–57. 10.1038/onc.2011.250
    1. Fogh J, Fogh JM, Orfeo T. One hundred and twenty-seven cultured human tumor cell lines producing tumors in nude mice. J Natl Cancer Inst 1977;59:221–6. 10.1093/jnci/59.1.221
    1. Adler A, Oraz R, Bystryn JC. In vitro cell-mediated immune responses induced by a polyvalent allogeneic melanoma vaccine. Cancer Biother 1995;10:211–24. 10.1089/cbr.1995.10.211
    1. Hoos A, Eggermont AMM, Janetzki S, et al. . Improved endpoints for cancer immunotherapy trials. J Natl Cancer Inst 2010;102:1388–97. 10.1093/jnci/djq310
    1. Grossmann KF, Othus M, Patel SP, et al. . Final analysis of overall survival (OS) and relapse-free-survival (RFS) in the intergroup S1404 phase III randomized trial comparing either high-dose interferon (HDI) or ipilimumab to pembrolizumab in patients with high-risk resected melanoma. J Clin Oncol 2021;39:9501–01. 10.1200/JCO.2021.39.15_suppl.9501
    1. Schwartzentruber DJ, Lawson DH, Richards JM, et al. . Gp100 peptide vaccine and interleukin-2 in patients with advanced melanoma. N Engl J Med 2011;364:2119–27. 10.1056/NEJMoa1012863
    1. Slingluff CL, Petroni GR, Olson W, et al. . Helper T-cell responses and clinical activity of a melanoma vaccine with multiple peptides from MAGE and melanocytic differentiation antigens. J Clin Oncol 2008;26:4973–80. 10.1200/JCO.2008.17.3161
    1. Hunder NN, Wallen H, Cao J, et al. . Treatment of metastatic melanoma with autologous CD4+ T cells against NY-ESO-1. N Engl J Med 2008;358:2698–703. 10.1056/NEJMoa0800251
    1. Robbins PF, Kassim SH, Tran TLN, et al. . A pilot trial using lymphocytes genetically engineered with an NY-ESO-1-reactive T-cell receptor: long-term follow-up and correlates with response. Clin Cancer Res 2015;21:1019–27. 10.1158/1078-0432.CCR-14-2708
    1. Jones D, Pereira ER, Padera TP. Growth and immune evasion of lymph node metastasis. Front Oncol 2018;8:36. 10.3389/fonc.2018.00036
    1. Pawelec G. Immunosenescence and cancer. Biogerontology 2017;18:717–21. 10.1007/s10522-017-9682-z
    1. Ramirez AG, Wages NA, Hu Y, et al. . Defining the effects of age and gender on immune response and outcomes to melanoma vaccination: a retrospective analysis of a single-institution clinical trials' experience. Cancer Immunol Immunother 2015;64:1531–9. 10.1007/s00262-015-1758-5
    1. Kugel CH, Douglass SM, Webster MR, et al. . Age correlates with response to anti-PD1, reflecting age-related differences in intratumoral effector and regulatory T-cell populations. Clin Cancer Res 2018;24:5347–56. 10.1158/1078-0432.CCR-18-1116
    1. Eggermont AMM, Suciu S, Testori A, et al. . Ulceration and stage are predictive of interferon efficacy in melanoma: results of the phase III adjuvant trials EORTC 18952 and EORTC 18991. Eur J Cancer 2012;48:218–25. 10.1016/j.ejca.2011.09.028
    1. Baurain J, Stas M, Hammouch F, et al. . Association of primary melanoma ulceration and clinical benefit of adjuvant vaccination with tumor-specific antigenic peptides. J Clin Oncol 2009;27:3022–22. 10.1200/jco.2009.27.15_suppl.3022
    1. Faries MB, Mozzillo N, Kashani-Sabet M, et al. . Long-Term survival after complete surgical resection and adjuvant immunotherapy for distant melanoma metastases. Ann Surg Oncol 2017;24:3991–4000. 10.1245/s10434-017-6072-3
    1. Morton DL, Mozzillo N, Thompson JF, et al. . An international, randomized, phase III trial of Bacillus Calmette-Guerin (BCG) plus allogeneic melanoma vaccine (MCV) or placebo after complete resection of melanoma metastatic to regional or distant sites. J Clin Oncol 2007;25:8508. 10.1200/jco.2007.25.18_suppl.8508
    1. Testori A, Richards J, Whitman E, et al. . Phase III comparison of vitespen, an autologous tumor-derived heat shock protein gp96 peptide complex vaccine, with physician's choice of treatment for stage IV melanoma: the C-100-21 Study Group. J Clin Oncol 2008;26:955–62. 10.1200/JCO.2007.11.9941
    1. Cheever MA, Higano CS. Provenge (Sipuleucel-T) in prostate cancer: the first FDA-approved therapeutic cancer vaccine. Clin Cancer Res 2011;17:3520–6. 10.1158/1078-0432.CCR-10-3126
    1. Schwartzentruber DJ, Lawson D, Richards J, et al. . A phase III multi-institutional randomized study of immunization with the gp100: 209–217(210M) peptide followed by high-dose IL-2 compared with high-dose IL-2 alone in patients with metastatic melanoma. JCO 2009;27:CRA9011. 10.1200/jco.2009.27.15_suppl.cra9011
    1. Merck announces phase 3 KEYNOTE-716 trial of anti-PD-1 therapy Keytruda to treat stage II melanoma meets primary endpoints. Available:
    1. Chen C, Wu B, Zhang C, et al. . Immune-related adverse events associated with immune checkpoint inhibitors: an updated comprehensive disproportionality analysis of the FDA adverse event reporting system. Int Immunopharmacol 2021;95:107498. 10.1016/j.intimp.2021.107498
    1. Arora P, Talamo L, Dillon P, et al. . Severe combined cardiac and neuromuscular toxicity from immune checkpoint blockade: an institutional case series. Cardiooncology 2020;6:21. 10.1186/s40959-020-00076-6
    1. Zhang R, Cai X-L, Liu L, et al. . Type 1 diabetes induced by immune checkpoint inhibitors. Chin Med J 2020;133:2595–8. 10.1097/CM9.0000000000000972
    1. Chang L-S, Barroso-Sousa R, Tolaney SM, et al. . Endocrine toxicity of cancer immunotherapy targeting immune checkpoints. Endocr Rev 2019;40:17–65. 10.1210/er.2018-00006
    1. Long GV, Hauschild A, Santinami M, et al. . Adjuvant dabrafenib plus trametinib in stage III BRAF-mutated melanoma. N Engl J Med 2017;377:1813–23. 10.1056/NEJMoa1708539

Source: PubMed

3
订阅