Safety and immunogenicity of the PRAME cancer immunotherapeutic in metastatic melanoma: results of a phase I dose escalation study

R Gutzmer, L Rivoltini, E Levchenko, A Testori, J Utikal, P A Ascierto, L Demidov, J J Grob, R Ridolfi, D Schadendorf, P Queirolo, A Santoro, C Loquai, B Dreno, A Hauschild, E Schultz, T P Lesimple, N Vanhoutte, B Salaun, M Gillet, S Jarnjak, P M De Sousa Alves, J Louahed, V G Brichard, F F Lehmann, R Gutzmer, L Rivoltini, E Levchenko, A Testori, J Utikal, P A Ascierto, L Demidov, J J Grob, R Ridolfi, D Schadendorf, P Queirolo, A Santoro, C Loquai, B Dreno, A Hauschild, E Schultz, T P Lesimple, N Vanhoutte, B Salaun, M Gillet, S Jarnjak, P M De Sousa Alves, J Louahed, V G Brichard, F F Lehmann

Abstract

Purpose: The PRAME tumour antigen is expressed in several tumour types but in few normal adult tissues. A dose-escalation phase I/II study (NCT01149343) assessed the safety, immunogenicity and clinical activity of the PRAME immunotherapeutic (recombinant PRAME protein (recPRAME) with the AS15 immunostimulant) in patients with advanced melanoma. Here, we report the phase I dose-escalation study segment.

Patients and methods: Patients with stage IV PRAME-positive melanoma were enrolled to 3 consecutive cohorts to receive up to 24 intramuscular injections of the PRAME immunotherapeutic. The RecPRAME dose was 20, 100 or 500 µg in cohorts 1, 2 and 3, respectively, with a fixed dose of AS15. Adverse events (AEs), including predefined dose-limiting toxicity (DLT) and the anti-PRAME humoral response (ELISA), were coprimary end points. Cellular immune responses were evaluated using in vitro assays.

Results: 66 patients were treated (20, 24 and 22 in the respective cohorts). AEs considered by the investigator to be causally related were mostly grade 1 or 2 injection site symptoms, fatigue, chills, fever and headache. Two DLTs (grade 3 brain oedema and proteinuria) were recorded in two patients in two cohorts (cohorts 2 and 3). All patients had detectable anti-PRAME antibodies after four immunisations. Percentages of patients with predefined PRAME-specific-CD4+T-cell responses after four immunisations were similar in each cohort. No CD8+ T-cell responses were detected.

Conclusions: The PRAME immunotherapeutic had an acceptable safety profile and induced similar anti-PRAME-specific humoral and cellular immune responses in all cohorts. As per protocol, the phase II study segment was initiated to further evaluate the 500 µg PRAME immunotherapeutic dose.

Trial registration number: NCT01149343, Results.

Keywords: PRAME antigen; cancer immunotherapy; immunogenicity; metastatic melanoma; safety.

Conflict of interest statement

Employment: MG (GSK group of companies); NV (GSK group of companies); BS (GSK group of companies); SJ (GSK group of companies); PMDeSA (GSK group of companies); FFL (GSK group of companies); JL (GSK group of companies); VGB (GSK group of companies). Stock Ownership: SJ (GSK group of companies), FFL (GSK group of companies); JL (GSK group of companies); VGB (GSK group of companies); PMDeSA (GSK group of companies). Honoraria: BD (Roche, GSK, BMS), DS (GSK, Roche, BMS, Amgen, Novartis, Merck/MSD), JJG (GSK, BMS, Roche, Amgen, MSD, Novartis, Meda), AT (BMS, GSK, Amgen, Roche), AH (Amgen, BMS, Celgene, Eisai, GSK, MedImmune, MelaSciences, Merck Serono, MSD/Merck, Novartis, Oncosec, Roche Pharma), RG (GSK, Roche, BMS, MSD, Novartis, Pfizer, Janssen, Amgen, Merck Serono, Boehringer, Almirall Hermal), JU (Roche, GSK, BMS), PA (BMS, Roche/Genentech, GSK, Ventana), RR (BMS, GSK), ES (Novartis, BMS, DermaPharm), TL (GSK, BMS, Merck, Roche). Consultant or Advisory Role: BD (Roche, BMS, GSK), DS (GSK, Roche, BMS, Amgen, Novartis, Merck/MSD), JJG (GSK, BMS, Roche, Amgen, MSD, Novartis, Meda), AT (BMS, GSK, Amgen, Roche), AH (Amgen, BMS, Celgene, Eisai, GSK, MedImmune, MelaSciences, Merck Serono, MSD/Merck, Novartis, Oncosec, Roche Pharma), RG (GSK, BMS, Roche, Novartis, Almirall Hermal, MSD, Amgen), JU (Roche, GSK), PA (BMS, Roche/Genentech, MSD, GSK, Ventana, Novartis, Amgen), PQ (GSK, Roche, Bristol, MSD), CL (Roche, BMS, MSD), ES (BMS). Speakers’ Bureau: DS (GSK; Roche, BMS, Amgen, Novartis, Merck/MSD), JJG (GSK), RG (GSK), JU (Roche), CL (Roche, BMS, MSD, Leo), TL (GSK, Merck, Roche), LD (GSK, BMS, Roche, MSD). Research funding: BD (Roche, GSK), DS (Merck), JJG (Roche), AH (trial grants from Amgen, BMS, Celgene, Eisai, GSK, MelaSciences, Merck Serono, MSD/Merck, Novartis, Oncosec, Roche Pharma), RG (Roche, Novartis, Pfizer, Johnson & Johnson), PA (BMS, Roche/Genentech, Ventana). Patents, royalties, other intellectual property: ES (Royalties from Ludwig Institute for Cancer Research for contribution to a patent on human tumor antigen). Travel, accommodations, expenses: BD (Roche, BMS), DS (GSK, Roche, BMS, Amgen, Novartis, Merck/MSD), AT (Oncovision), RG (Roche, BMS), JU (Roche, GSK), PQ (Roche, GSK, MSD), CL (Roche, BMS, Leo), TL (Roche). EL, LR, AS declare that they have no conflict of interest.

Figures

Figure 1
Figure 1
Seropositivity rates and geometric mean antibody concentrations (GMCs) for anti-PRAME IgG antibodies (ATP cohort for immunogenicity). Footnote: N=number of patients with available results, n/%=number/percentage of patients with concentrations above the cut-off, vertical lines indicate 95% CIs, dotted line shows assay cut-off (12 E.U/mL), Pre=prior to dose 1, Post II=2 weeks after the second dose, Post IV=2 weeks after the fourth dose. ATP, according-to-protocol; PRAME, PReferentially expressed Antigen of Melanoma.
Figure 2
Figure 2
PRAME-specific CD4+ T-cell (TNF-α+/IFN-γ+) immunogenicity scores and cellular response prior to treatment and postdose 4 (ATP cohort for immunogenicity). Footnote: N=number of patients with available results, n/%=number/percentage of patients with immunogenicity score/response, vertical lines indicate 95% CIs, dotted line shows cut-off (2.68), Pre=prior to dose 1, Post IV=2 weeks after the fourth dose. See online supplementary data for details of the derivation of cut-offs and methods. ATP, according-to-protocol; IFN-γ, interferon-γ; TNF-α, tumour necrosis factor α; PRAME, PReferentially expressed Antigen of Melanoma.

References

    1. Balch CM, Gershenwald JE, Soong SJ et al. . Final version of 2009 AJCC melanoma staging and classification. J Clin Oncol 2009;27:6199–206. 10.1200/JCO.2009.23.4799
    1. Singh BP, Salama AKS. Updates in therapy for advanced melanoma. Cancers (Basel) 2016;8:pii:E17 10.3390/cancers8010017
    1. Ikeda H, Lethé B, Lehmann F et al. . Characterization of an antigen that is recognized on a melanoma showing partial HLA loss by CTL expressing an NK inhibitory receptor. Immunity 1997;6:199–208.
    1. Doolan P, Clynes M, Kennedy S et al. . Prevalence and prognostic and predictive relevance of PRAME in breast cancer. Breast Cancer Res Treat 2008;109:359–65. 10.1007/s10549-007-9643-3
    1. Kessler JH, Beekman NJ, Bres-Vloemans SA et al. . Efficient identification of novel HLA-A(*)0201-presented cytotoxic T lymphocyte epitopes in the widely expressed tumor antigen PRAME by proteasome-mediated digestion analysis. J Exp Med 2001;193:73–88.
    1. Quintarelli C, Dotti G, Hasan ST et al. . High-avidity cytotoxic T lymphocytes specific for a new PRAME-derived peptide can target leukemic and leukemic-precursor cells. Blood 2011;117:3353–62. 10.1182/blood-2010-08-300376
    1. Griffioen M, Kessler JH, Borghi M et al. . Detection and functional analysis of CD8+ T cells specific for PRAME: a target for T-cell therapy. Clin Cancer Res 2006;12:3130–6. 10.1158/1078-0432.CCR-05-2578
    1. Rezvani K, Yong ASM, Tawab A et al. . Ex vivo characterization of polyclonal memory CD8+ T-cell responses to PRAME-specific peptides in patients with acute lymphoblastic leukemia and acute and chronic myeloid leukemia. Blood 2009;113:2245–55. 10.1182/blood-2008-03-144071
    1. Weber JS, Vogelzang NJ, Ernstoff MS et al. . A phase 1 study of a vaccine targeting preferentially expressed antigen in melanoma and prostate-specific membrane antigen in patients with advanced solid tumors. J Immunother 2011;34:556–67. 10.1097/CJI.0b013e3182280db1
    1. Larkin J, Chiarion-Sileni V, Gonzalez R et al. . Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. N Engl J Med 2015;373:23–34. 10.1056/NEJMoa1504030
    1. Robert C, Schachter J, Long GV et al. . Pembrolizumab versus Ipilimumab in Advanced Melanoma. N Engl J Med 2015;372:2521–32. 10.1056/NEJMoa1503093
    1. Pujol J-L, De Pas T, Rittmeyer A et al. . Immunogenicity and safety of the PRAME cancer immunotherapeutic in non-small cell lung cancer (NSCLC): A phase I dose escalation study. Ann Oncol 2012;23(Suppl 9):ix386 (abs1182P).
    1. Kruit WHJ, Suciu S, Dreno B et al. . Selection of immunostimulant AS15 for active immunization with MAGE-A3 protein: results of a randomized Phase II study of the European Organisation for Research and Treatment of Cancer Melanoma Group in metastatic melanoma. J Clin Oncol 2013;31:2413–20. 10.1200/JCO.2012.43.7111
    1. Kruit WHJ, van Ojik HH, Brichard VG et al. . Phase 1/2 study of subcutaneous and intradermal immunization with a recombinant MAGE-3 protein in patients with detectable metastatic melanoma. Int J Cancer 2005;117:596–604. 10.1002/ijc.21264
    1. Vansteenkiste J, Zielinski M, Linder A et al. . Adjuvant MAGE-A3 immunotherapy in resected non-small-cell lung cancer: phase II randomized study results. J Clin Oncol 2013;31:2396–403. 10.1200/JCO.2012.43.7103
    1. Stockert E, Jäger E, Chen YT et al. . A survey of the humoral immune response of cancer patients to a panel of human tumor antigens. J Exp Med 1998;187:1349–54.
    1. Yuan J, Adamow M, Ginsberg BA et al. . Integrated NY-ESO-1 antibody and CD8+ T-cell responses correlate with clinical benefit in advanced melanoma patients treated with ipilimumab. Proc Natl Acad Sci USA 2011;108:16723–8. 10.1073/pnas.1110814108
    1. Valmori D, Souleimanian NE, Tosello V et al. . Vaccination with NY-ESO-1 protein and CpG in Montanide induces integrated antibody/Th1 responses and CD8 T cells through cross-priming. Proc Natl Acad Sci USA 2007;104:8947–52. 10.1073/pnas.0703395104
    1. Carrasco J, Van Pel A, Neyns B et al. . Vaccination of a melanoma patient with mature dendritic cells pulsed with MAGE-3 peptides triggers the activity of nonvaccine anti-tumor cells. J Immunol 2008;180:3585–93.
    1. Tomita Y, Yuno A, Tsukamoto H et al. . Identification of promiscuous KIF20A long peptides bearing both CD4+ and CD8+ T-cell epitopes: KIF20A-Specific CD4+ T-cell immunity in patients with malignant tumor. Clin Cancer Res 2013;19:4508–20. 10.1158/1078-0432.CCR-13-0197
    1. Schultz ES, Schuler-Thurner B, Stroobant V et al. . Functional analysis of tumor-specific Th cell responses detected in melanoma patients after dendritic cell-based immunotherapy. J Immunol 2004;172:1304–10.
    1. Braumüller H, Wieder T, Brenner E et al. . T helper-1-cell cytokines drive cancer into senescence. Nature 2013;494:361–5. 10.1038/nature11824
    1. Roeder C, Schuler-Thurner B, Berchtold S et al. . MAGE-A3 is a frequent tumor antigen of metastasized melanoma. Arch Dermatol Res 2005;296:314–19. 10.1007/s00403-004-0527-7

Source: PubMed

3
Subscribe