Association of homogeneous inflamed gene signature with a better outcome in patients with metastatic melanoma treated with MAGE-A3 immunotherapeutic

Jean-François Baurain, Caroline Robert, Laurent Mortier, Bart Neyns, Florent Grange, Céleste Lebbe, Fernando Ulloa-Montoya, Pedro Miguel De Sousa Alves, Marc Gillet, Jamila Louahed, Silvija Jarnjak, Frédéric F Lehmann, Jean-François Baurain, Caroline Robert, Laurent Mortier, Bart Neyns, Florent Grange, Céleste Lebbe, Fernando Ulloa-Montoya, Pedro Miguel De Sousa Alves, Marc Gillet, Jamila Louahed, Silvija Jarnjak, Frédéric F Lehmann

Abstract

Purpose: This study assessed clinical activity, safety and immunogenicity of MAGE-A3 immunotherapeutic in patients with MAGE-A3-positive metastatic melanoma.

Patients and methods: In this open-label, multicentre, uncontrolled, Phase II study (ClinicalTrials.gov NCT00896480), patients received ≤24 doses of MAGE-A3 immunotherapeutic (4-cycle schedule). At screening, two skin lesions were biopsied for MAGE-A3 expression analysis and presence/absence of a previously identified gene signature (GS) associated with favourable clinical outcome. Clinical activity was assessed in terms of clinical response, time-to-treatment failure (TTF) and progression-free survival (PFS). Adverse events (AEs) and serious AEs (SAEs) were recorded. MAGE-A3-specific immune responses were assessed. Clinical activity and immunogenicity were analysed overall and separately in patients with 2/2 (GS+/+), 1/2 (GS+/-) or 0/2 (GS-/-) biopsies presenting GS.

Results: Of 49 screened patients, 32 had MAGE-A3-positive tumours; 24 (8 GS+/+, 8 GS+/-, 8 GS-/-) were treated. Two complete (GS+/+ patients) and two partial responses (one GS+/+, one GS+/-) were reported; of note, one of the two complete responses was unlikely to be related to the study treatment. Median TTF and PFS were 14.8 and 7.2 months for GS+/+, 2.3 and 2.8 months for GS+/- and 2.4 and 2.9 months for GS-/- patients. Three grade 3 AEs and two SAEs unrelated to treatment were reported. All patients were seropositive for MAGE-A3 antibodies on vaccination with no differences between the different GS profiles. MAGE-A3-specific CD4+ and CD8+ T cell immunogenicity was detected; 12/16 (75.0%) of patients presented CD4+ T cell responses.

Conclusion: Treatment with MAGE-A3 immunotherapeutic showed signs of clinical activity in GS+/+ patients. Treatment was well tolerated and immunogenic. No differences in immune responses according to GS status were observed.

Trial registration number: NCT00896480 (Results).

Keywords: clinical activity; gene signature; mage-a3 immunotherapeutic; melanoma; safety.

Conflict of interest statement

Competing interests: MG, JL, FU-M and SJ are employees of the GSK group of companies. FFL and PMDSA were employees of the GSK group of companies during the conduct of the study. FFL, JL, PMDSA, FU-M and SJ hold shares in the GSK group of companies as part of their employee remuneration.

Figures

Figure 1
Figure 1
Participant flow. Of the 24 patients included in the study, 3 patients completed the study. Patients may have more than one reason for elimination from ATP population. ATP, according-to-protocol; GS+/+, patients presenting the gene signature on both biopsies; GS+/-, patients presenting the gene signature on one biopsy and not on the other one; GS-/-, patients without the gene signature; N, number of patients.
Figure 2
Figure 2
TTF and PFS (total treated population). TTF was longer in patients for whom, out of the two biopsied lesions, both were GS-positive (GS+/+) (14.8 months) than in patients with only one GS-positive biopsy (GS+/-) (2.3 months) and in patients with both biopsies being GS-negative (GS-/-) (2.4 months) (A). PFS was longer for GS+/+ patients (7.2 months) than for GS+/- patients (2.8 months) and GS-/- patients (2.9 months) (B). PFS, progression-free survival; TTF, time-to-treatment failure.
Figure 3
Figure 3
MAGE-A3-specific geometric mean titres (GMCs) (ATP population for immunogenicity). In the overall population, 2 weeks postdose 2, all patients were seropositive for MAGE-A3-specific antibodies (GMC 1865.7 EU/mL). The antibody levels further increased up to postdose 6 timepoint (GMC 9080.5 EU/mL); thereafter, a plateau was observed (A). There were no marked differences in MAGE-A3-specific antibody profiles between patients presenting the different gene signature on the two biopsies (B). The error bars represent 95% CI. ATP, according-to-protocol; EU, ELISA units; GMC, geometric mean concentration; GS+/+, patients presenting the gene signature on both biopsies; GS+/-, patients presenting the gene signature on one biopsy and not on the other one; GS-/-, patients without the gene signature; Pre, baseline; Post-, postdose number indicated by Arabic numerals.
Figure 4
Figure 4
MAGE-A3-specific cellular responses (ATP population for immunogenicity). Prior to the first MAGE-A3 immunotherapeutic administration, MAGE-A3-specific double-positive TNF-α/IFN-γ-producing (TNF-α/IFN-γ++) CD4+ T cells with immunogenicity score above the assay cut-off value (1.24) were found in one patient; the highest proportion of patients with these cells was observed postdose 12 (9/10 patients (90.0%)) (A). The highest proportion of CD4+ T cell responders was observed postdose 6 at the end of cycle 1 (8/13 patients (61.5%)) (B). CMI response was defined as GMR is above the cut-off value (1.24) and at least a 4-fold increase after immunisation as compared with the patient’s baseline value. The numbers in brackets indicate numbers of patients with GMR ≥cut-off value (A) or number of patients with CMI response (B). ATP, according-to-protocol; CMI, cell-mediated immune; GM, geometric mean of MAGE-A3-specific immunogenicity score (GMR) calculated on all patients; GMR, geometric mean ratio; IFN-γ, interferon gamma; Pre, baseline; Post-, postdose number indicated by Arabic numerals; TNF-α, tumour necrosis factor-alpha.

References

    1. WHO. Skin cancers, 2015. accessed on 11 May 2015.
    1. Agarwala SS. Current systemic therapy for metastatic melanoma. Expert Rev Anticancer Ther 2009;9:587–95. 10.1586/era.09.25
    1. Davies MA, Liu P, McIntyre S, et al. . Prognostic factors for survival in melanoma patients with brain metastases. Cancer 2011;117:1687–96. 10.1002/cncr.25634
    1. Tas F. Metastatic behavior in melanoma: timing, pattern, survival, and influencing factors. J Oncol 2012;2012:1–9. 10.1155/2012/647684
    1. Kaufman HL, Margolin K, Sullivan R. Management of Metastatic Melanoma in 2018. JAMA Oncol 2018;4:857 10.1001/jamaoncol.2018.0170
    1. Haass NK. Dynamic tumor heterogeneity in melanoma therapy: how do we address this in a novel model system? Melanoma Manag 2015;2:93–5. 10.2217/mmt.15.1
    1. Somasundaram R, Villanueva J, Herlyn M. Intratumoral heterogeneity as a therapy resistance mechanism: role of melanoma subpopulations. Adv Pharmacol 2012;65:335–59. 10.1016/B978-0-12-397927-8.00011-7
    1. Griewank KG, Scolyer RA, Thompson JF, et al. . Genetic alterations and personalized medicine in melanoma: progress and future prospects. J Natl Cancer Inst 2014;106 djt435 10.1093/jnci/djt435
    1. Palmieri G, Ombra M, Colombino M, et al. . Multiple Molecular Pathways in Melanomagenesis: Characterization of Therapeutic Targets. Front Oncol 2015;5:183 10.3389/fonc.2015.00183
    1. Colombino M, Sini M, Lissia A, et al. . Discrepant alterations in main candidate genes among multiple primary melanomas. J Transl Med 2014;12:117 10.1186/1479-5876-12-117
    1. Balkwill FR, Capasso M, Hagemann T. The tumor microenvironment at a glance. J Cell Sci 2012;125(Pt 23):5591–6. 10.1242/jcs.116392
    1. Chen F, Zhuang X, Lin L, et al. . New horizons in tumor microenvironment biology: challenges and opportunities. BMC Med 2015;13:45 10.1186/s12916-015-0278-7
    1. Mbeunkui F, Johann DJ. Cancer and the tumor microenvironment: a review of an essential relationship. Cancer Chemother Pharmacol 2009;63:571–82. 10.1007/s00280-008-0881-9
    1. Sun Y. Tumor microenvironment and cancer therapy resistance. Cancer Lett 2016;380:205–15. 10.1016/j.canlet.2015.07.044
    1. Whiteside TL. The tumor microenvironment and its role in promoting tumor growth. Oncogene 2008;27:5904–12. 10.1038/onc.2008.271
    1. Shiao SL, Ganesan AP, Rugo HS, et al. . Immune microenvironments in solid tumors: new targets for therapy. Genes Dev 2011;25:2559–72. 10.1101/gad.169029.111
    1. A Schindler K, Postow MA. Current options and future directions in the systemic treatment of metastatic melanoma. J Community Support Oncol 2014;12:20–6. 10.12788/jcso.0005
    1. Azijli K, Stelloo E, Peters GJ, et al. . New developments in the treatment of metastatic melanoma: immune checkpoint inhibitors and targeted therapies. Anticancer Res 2014;34:1493–505.
    1. Johnson DB, Sosman JA. Therapeutic Advances and Treatment Options in Metastatic Melanoma. JAMA Oncol 2015;1:380–6. 10.1001/jamaoncol.2015.0565
    1. Menzies AM, Long GV. Recent advances in melanoma systemic therapy. BRAF inhibitors, CTLA4 antibodies and beyond. Eur J Cancer 2013;49:3229–41. 10.1016/j.ejca.2013.06.027
    1. Aris M, Barrio MM. Combining immunotherapy with oncogene-targeted therapy: a new road for melanoma treatment. Front Immunol 2015;6:46 10.3389/fimmu.2015.00046
    1. Sun Y. Translational horizons in the tumor microenvironment: harnessing breakthroughs and targeting cures. Med Res Rev 2015;35:n/a–36. 10.1002/med.21338
    1. van der Burg SH. Correlates of immune and clinical activity of novel cancer vaccines. Semin Immunol 2018. 10.1016/j.smim.2018.04.001
    1. Jungbluth AA, Silva WA, Iversen K, et al. . Expression of cancer-testis (CT) antigens in placenta. Cancer Immun 2007;7:15.
    1. Roeder C, Schuler-Thurner B, Berchtold S, et al. . MAGE-A3 is a frequent tumor antigen of metastasized melanoma. Arch Dermatol Res 2005;296:314–9. 10.1007/s00403-004-0527-7
    1. Esfandiary A, Ghafouri-Fard S. MAGE-A3: an immunogenic target used in clinical practice. Immunotherapy 2015;7:683–704. 10.2217/imt.15.29
    1. Reuschenbach M, von Knebel Doeberitz M, Wentzensen N. A systematic review of humoral immune responses against tumor antigens. Cancer Immunol Immunother 2009;58:1535–44. 10.1007/s00262-009-0733-4
    1. Blankenstein T, Coulie PG, Gilboa E, et al. . The determinants of tumour immunogenicity. Nat Rev Cancer 2012;12:307–13. 10.1038/nrc3246
    1. Mellman I, Coukos G, Dranoff G. Cancer immunotherapy comes of age. Nature 2011;480:480–9. 10.1038/nature10673
    1. Ohue Y, Wada H, Oka M, et al. . Antibody response to cancer/testis (CT) antigens: A prognostic marker in cancer patients. Oncoimmunology 2014;3:e970032 10.4161/21624011.2014.970032
    1. Kruit WH, 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. Pujol JL, Vansteenkiste JF, De Pas TM, et al. . Safety and Immunogenicity of MAGE-A3 Cancer Immunotherapeutic with or without Adjuvant Chemotherapy in Patients with Resected Stage IB to III MAGE-A3-Positive Non-Small-Cell Lung Cancer. J Thorac Oncol 2015;10:1458–67. 10.1097/JTO.0000000000000653
    1. Vansteenkiste JF, Cho BC, Vanakesa T, et al. . Efficacy of the MAGE-A3 cancer immunotherapeutic as adjuvant therapy in patients with resected MAGE-A3-positive non-small-cell lung cancer (MAGRIT): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 2016;17:822–35. 10.1016/S1470-2045(16)00099-1
    1. Saiag P, Gutzmer R, Ascierto PA, et al. . Prospective assessment of a gene signature potentially predictive of clinical benefit in metastatic melanoma patients following MAGE-A3 immunotherapeutic (PREDICT). Annals of Oncology 2016;27:1947–53. 10.1093/annonc/mdw291
    1. Vantomme V, Dantinne C, Amrani N, et al. . Immunologic analysis of a phase I/II study of vaccination with MAGE-3 protein combined with the AS02B adjuvant in patients with MAGE-3-positive tumors. J Immunother 2004;27:124–35. 10.1097/00002371-200403000-00006
    1. Ulloa-Montoya F, Louahed J, Dizier B, et al. . Predictive gene signature in MAGE-A3 antigen-specific cancer immunotherapy. J Clin Oncol 2013;31:2388–95. 10.1200/JCO.2012.44.3762
    1. Hachey SJ, Boiko AD. Therapeutic implications of melanoma heterogeneity. Exp Dermatol 2016;25:497–500. 10.1111/exd.13002
    1. Chiou VL, BM. and Immune-Related Response in Solid Tumors. J Clin Oncol 2015;33:3541–3.
    1. Chiou VL, Burotto M. Pseudoprogression and Immune-Related Response in Solid Tumors. J Clin Oncol 2015;33:3541–3. 10.1200/JCO.2015.61.6870
    1. Robert C, Thomas L, Bondarenko I, et al. . Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med 2011;364:2517–26. 10.1056/NEJMoa1104621
    1. McGranahan N, Swanton C. Biological and therapeutic impact of intratumor heterogeneity in cancer evolution. Cancer Cell 2015;27:15–26. 10.1016/j.ccell.2014.12.001
    1. Swanton C. Intratumor heterogeneity: evolution through space and time. Cancer Res 2012;72:4875–82. 10.1158/0008-5472.CAN-12-2217
    1. Yap TA, Gerlinger M, Futreal PA, et al. . Intratumor heterogeneity: seeing the wood for the trees. Sci Transl Med 2012;4:127ps10 10.1126/scitranslmed.3003854
    1. Marchand M, Punt CJ, Aamdal S, et al. . Immunisation of metastatic cancer patients with MAGE-3 protein combined with adjuvant SBAS-2: a clinical report. Eur J Cancer 2003;39:70–7. 10.1016/S0959-8049(02)00479-3

Source: PubMed

3
Suscribir