Randomized phase II trial of lymphodepletion plus adoptive cell transfer of tumor-infiltrating lymphocytes, with or without dendritic cell vaccination, in patients with metastatic melanoma

Chantal Saberian, Rodabe N Amaria, Amer M Najjar, Laszlo G Radvanyi, Cara L Haymaker, Marie-Andrée Forget, Roland L Bassett, Silvana C Faria, Isabella C Glitza, Enrique Alvarez, Sapna Parshottam, Victor Prieto, Gregory Lizée, Michael K Wong, Jennifer L McQuade, Adi Diab, Cassian Yee, Hussein A Tawbi, Sapna Patel, Elizabeth J Shpall, Michael A Davies, Patrick Hwu, Chantale Bernatchez, Chantal Saberian, Rodabe N Amaria, Amer M Najjar, Laszlo G Radvanyi, Cara L Haymaker, Marie-Andrée Forget, Roland L Bassett, Silvana C Faria, Isabella C Glitza, Enrique Alvarez, Sapna Parshottam, Victor Prieto, Gregory Lizée, Michael K Wong, Jennifer L McQuade, Adi Diab, Cassian Yee, Hussein A Tawbi, Sapna Patel, Elizabeth J Shpall, Michael A Davies, Patrick Hwu, Chantale Bernatchez

Abstract

Background: The adoptive transfer of tumor-infiltrating lymphocytes (TIL) has demonstrated robust efficacy in metastatic melanoma patients. Tumor antigen-loaded dendritic cells (DCs) are believed to optimally activate antigen-specific T lymphocytes. We hypothesized that the combined transfer of TIL, containing a melanoma antigen recognized by T cells 1 (MART-1) specific population, with MART-1-pulsed DC will result in enhanced proliferation and prolonged survival of transferred MART-1 specific T cells in vivo ultimately leading to improved clinical responses.

Design: We tested the combination of TIL and DC in a phase II clinical trial of patients with advanced stage IV melanoma. HLA-A0201 patients whose early TIL cultures demonstrated reactivity to MART-1 peptide were randomly assigned to receive TIL alone or TIL +DC pulsed with MART-1 peptide. The primary endpoint was to evaluate the persistence of MART-1 TIL in the two arms. Secondary endpoints were to evaluate clinical response and survival.

Results: Ten patients were given TIL alone while eight patients received TIL+DC vaccine. Infused MART-1 reactive CD8+ TIL were tracked in the blood over time by flow cytometry and results show good persistence in both arms, with no difference in the persistence of MART-1 between the two arms. The objective response rate was 30% (3/10) in the TIL arm and 50% (4/8) in the TIL+DC arm. All treatments were well tolerated.

Conclusions: The combination of TIL +DC showed no difference in the persistence of MART-1 TIL compared with TIL therapy alone. Although more patients showed a clinical response to TIL+DC therapy, this study was not powered to resolve differences between groups.

Trial registration number: NCT00338377.

Keywords: adaptive immunity; dendritic cells; lymphocytes; melanoma; tumor-infiltrating; vaccination.

Conflict of interest statement

Competing interests: CLH is on the SAB for Briacell. PH consulting agreement with Immatics, Dragonfly, Sanofi, GSK. CB is on the SAB of Myst therapeutics and received research funding from Iovance biotherapeutics.

© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY. Published by BMJ.

Figures

Figure 1
Figure 1
The infusion of a larger proportion of CD8+ TIL tends to associate with longer survival. (A) Total number of TIL infused by arm (left panel) or responsecircles represent patients treated with TIL alone while squares denote patients who received TIL +DC. Mean and SD are shown. Statistics were calculated with double sided Mann-Whitney U test. (B) %CD8+ TIL infused by arm (left panel) or response (right panel). Circles represent patients who responded (CR or PR) while squares denote patients who did not respond (SD or PD). Mean and SD are shown. Statistics were calculated with double sided Mann-Whitney U test. (C) %CD8+ TIL infused and PFS D) %CD8+ TIL infused and OS. (E) Kaplan-Meier plot of OS stratified by %CD8+ TIL infused (≥50% or <50%). CR, complete response; DC, dendritic cell; ns, not significant; OS, overall survival; PFS, progression-free survival; PD, progressive disease; PR, partial response; SD, stable disease; TIL, tumor-infiltrating lymphocytes.
Figure 2
Figure 2
MART-1 reactivity of TIL #302 infusion product. (A) Flow cytometry evaluation of MART-1 reactivity in infusion product from TIL #302. Gating strategy demonstrates the gating of viable (7AAD negative) lymphocytic population. The cells were stained for CD8 and either no tetramer (left plot), gp100 dextramer (middle plot) or MART-1 dextramer (right plot). (B) Nanostring analysis of TCR clonotypes. (C) Sequencing data of the CDR3 region of the alpha chain of the TCR. (D) Sequencing data of the CDR3 region of the beta chain of the TCR. (E) Alignment of the CDR3 sequence of the alpha and beta chains of the TCR with published sequences from CDR3 regions of MART-1 recognizing TCRs. MART-1, melanoma antigen recognized by T cells 1; TIL, tumor-infiltrating lymphocytes.
Figure 3
Figure 3
Persistence of MART-1 reactive CD8+ TIL after infusion. (A) %MART-1 reactive CD8+ TIL infused by arm (left panel) and response (right panel). Circles represent patients treated with TIL alone while triangles denote patients who received TIL+DC. Statistics were calculated with double-sided Mann-Whitney U test. (B) %MART-1 reactive CD8+ TIL, as a proportion of total CD8+ T cells, circulating in the blood at 1 month post TIL infusion, by arm. Circles represent patients treated with TIL alone while triangles denote patients who received TIL+DC. Statistics were calculated with double sided Mann-Whitney U test. (C) Longitudinal tracking of MART-1 reactive CD8+ T cells in the blood of patients treated in the TIL arm. Dotted lines indicate Responder patients. Values less than 0.001% (or undetected) were represented as 0.001% to allow visualization on a logarithmic axis (D) longitudinal tracking of MART-1 reactive CD8+ T cells in the blood of patients treated in the TIL+DC arm by flow cytometry. Dotted lines indicate Responder patients. values less than 0.001% (or undetected) were represented as 0.001% to allow visualization on a logarithmic axis. DC, dendritic cell; ns, not significant; MART-1, melanoma antigen recognized by T cells 1; TIL, tumor-infiltrating lymphocytes.
Figure 4
Figure 4
Clinical response to TIL with or without DC co-vaccination. (A) Waterfall plot (B) spider plot depicting the tumor burden of every patient at every time point collected. Patients in the TIL arm are denoted by a black curve while patients in the TIL +DC arm are denoted by a blue curve. (C) Kaplan-Meier plot showing pfs by treatment arm. (red curve is for TIL alone and blue curve is for TIL+DC patients). The median PFS duration was 0.26 years in the TIL arm and 0.49 years in the TIL +DC arm. The 2-year PFS rate was 0% (95% CI 1.6% to 64%) in the TIL arm and 13% in the TIL +DC arm (95% CI 13% to 78%). (D) Kaplan-Meier plot of os by treatment arm. (Red curve is for TIL alone and blue curve is for TIL +DC patients). The median OS duration was 4.1 years in the TIL arm and 2.0 years in the TIL+DC arm. at 2 years, the OS rate was 58% (95% CI 34% to 100%) in the TIL arm and 50% in the TIL +DC arm (95% CI 25% to 100%). DC, dendritic cell; OS, overall survival; PFS, progression-free survival; TIL, tumor-infiltrating lymphocytes.
Figure 5
Figure 5
Long-term systemic tumor control following local radiation of the brain in patients who had received TIL. Systemic tumor burden decreases over time in three patients who received local intracranial radiation of the brain within a month of being treated with TIL therapy (Patients #228, 232 and 312). TIL, tumor-infiltrating lymphocytes. Abbreviations: FDG, Fluorodeoxyglucose. NED, No evidence of disease.

References

    1. Rosenberg SA, Restifo NP. Adoptive cell transfer as personalized immunotherapy for human cancer. Science 2015;348:62–8. 10.1126/science.aaa4967
    1. Forget M-A, Haymaker C, Hess KR, et al. . Prospective analysis of adoptive TIL therapy in patients with metastatic melanoma: response, impact of anti-CTLA4, and biomarkers to predict clinical outcome. Clin Cancer Res 2018;24:4416–28. 10.1158/1078-0432.CCR-17-3649
    1. Pedersen M, Westergaard MCW, Milne K, et al. . Adoptive cell therapy with tumor-infiltrating lymphocytes in patients with metastatic ovarian cancer: a pilot study. Oncoimmunology 2018;7:e1502905. 10.1080/2162402X.2018.1502905
    1. Tran E, Robbins PF, Lu Y-C, et al. . T-Cell transfer therapy targeting mutant KRAS in cancer. N Engl J Med 2016;375:2255–62. 10.1056/NEJMoa1609279
    1. Tran E, Turcotte S, Gros A, et al. . Cancer immunotherapy based on mutation-specific CD4+ T cells in a patient with epithelial cancer. Science 2014;344:641–5. 10.1126/science.1251102
    1. Zacharakis N, Chinnasamy H, Black M, et al. . Immune recognition of somatic mutations leading to complete durable regression in metastatic breast cancer. Nat Med 2018;24:724–30. 10.1038/s41591-018-0040-8
    1. Stevanović S, Helman SR, Wunderlich JR, et al. . A phase II study of tumor-infiltrating lymphocyte therapy for human papillomavirus-associated epithelial cancers. Clin Cancer Res 2019;25:1486–93. 10.1158/1078-0432.CCR-18-2722
    1. Radvanyi LG, Bernatchez C, Zhang M, et al. . Specific lymphocyte subsets predict response to adoptive cell therapy using expanded autologous tumor-infiltrating lymphocytes in metastatic melanoma patients. Clin Cancer Res 2012;18:6758–70. 10.1158/1078-0432.CCR-12-1177
    1. Rosenberg SA, Yang JC, Sherry RM, et al. . Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin Cancer Res 2011;17:4550–7. 10.1158/1078-0432.CCR-11-0116
    1. Besser MJ, Shapira-Frommer R, Itzhaki O, et al. . Adoptive transfer of tumor-infiltrating lymphocytes in patients with metastatic melanoma: intent-to-treat analysis and efficacy after failure to prior immunotherapies. Clin Cancer Res 2013;19:4792–800. 10.1158/1078-0432.CCR-13-0380
    1. Goff SL, Dudley ME, Citrin DE, et al. . Randomized, prospective evaluation comparing intensity of lymphodepletion before adoptive transfer of tumor-infiltrating lymphocytes for patients with metastatic melanoma. J Clin Oncol 2016;34:2389–97. 10.1200/JCO.2016.66.7220
    1. Pilon-Thomas S, Kuhn L, Ellwanger S, et al. . Efficacy of adoptive cell transfer of tumor-infiltrating lymphocytes after lymphopenia induction for metastatic melanoma. J Immunother 2012;35:615–20. 10.1097/CJI.0b013e31826e8f5f
    1. van den Berg JH, Heemskerk B, van Rooij N, et al. . Tumor infiltrating lymphocytes (TIL) therapy in metastatic melanoma: boosting of neoantigen-specific T cell reactivity and long-term follow-up. J Immunother Cancer 2020;8:e000848. 10.1136/jitc-2020-000848
    1. Robbins PF, Dudley ME, Wunderlich J, et al. . Cutting edge: persistence of transferred lymphocyte clonotypes correlates with cancer regression in patients receiving cell transfer therapy. J Immunol 2004;173:7125–30. 10.4049/jimmunol.173.12.7125
    1. Sabado RL, Balan S, Bhardwaj N. Dendritic cell-based immunotherapy. Cell Res 2017;27:74–95. 10.1038/cr.2016.157
    1. Spranger S, Dai D, Horton B, et al. . Tumor-Residing Batf3 dendritic cells are required for effector T cell trafficking and adoptive T cell therapy. Cancer Cell 2017;31:711–23. 10.1016/j.ccell.2017.04.003
    1. Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature 1998;392:245–52. 10.1038/32588
    1. Lizée G, Basha G, Jefferies WA. Tails of wonder: endocytic-sorting motifs key for exogenous antigen presentation. Trends Immunol 2005;26:141–9. 10.1016/j.it.2005.01.005
    1. Cerundolo V, Hermans IF, Salio M. Dendritic cells: a journey from laboratory to clinic. Nat Immunol 2004;5:7–10. 10.1038/ni0104-7
    1. Ridgway D. The first 1000 dendritic cell vaccinees. Cancer Invest 2003;21:873–86. 10.1081/CNV-120025091
    1. Lizée G, Cantu MA, Hwu P. Less Yin, more Yang: confronting the barriers to cancer immunotherapy. Clin Cancer Res 2007;13:5250–5. 10.1158/1078-0432.CCR-07-1722
    1. Kvistborg P, Shu CJ, Heemskerk B, et al. . TIL therapy broadens the tumor-reactive CD8(+) T cell compartment in melanoma patients. Oncoimmunology 2012;1:409–18. 10.4161/onci.18851
    1. Prickett TD, Crystal JS, Cohen CJ, et al. . Durable complete response from metastatic melanoma after transfer of autologous T cells recognizing 10 mutated tumor antigens. Cancer Immunol Res 2016;4:669–78. 10.1158/2326-6066.CIR-15-0215
    1. Lou Y, Wang G, Lizée G, et al. . Dendritic cells strongly boost the antitumor activity of adoptively transferred T cells in vivo. Cancer Res 2004;64:6783–90. 10.1158/0008-5472.CAN-04-1621
    1. Tanyi JL, Bobisse S, Ophir E, et al. . Personalized cancer vaccine effectively mobilizes antitumor T cell immunity in ovarian cancer. Sci Transl Med 2018;10. 10.1126/scitranslmed.aao5931. [Epub ahead of print: 11 Apr 2018].
    1. Wilgenhof S, Corthals J, Heirman C, et al. . Phase II study of autologous monocyte-derived mRNA electroporated dendritic cells (TriMixDC-MEL) plus ipilimumab in patients with pretreated advanced melanoma. J Clin Oncol 2016;34:1330–8. 10.1200/JCO.2015.63.4121
    1. Nesselhut J, Marx D, Lange H, et al. . Systemic treatment with anti-PD-1 antibody nivolumab in combination with vaccine therapy in advanced pancreatic cancer. JCO 2016;34:3092. 10.1200/JCO.2016.34.15_suppl.3092
    1. Ribas A, Comin-Anduix B, Chmielowski B, et al. . Dendritic cell vaccination combined with CTLA4 blockade in patients with metastatic melanoma. Clin Cancer Res 2009;15:6267–76. 10.1158/1078-0432.CCR-09-1254
    1. Poschke I, Lövgren T, Adamson L, et al. . A phase I clinical trial combining dendritic cell vaccination with adoptive T cell transfer in patients with stage IV melanoma. Cancer Immunol Immunother 2014;63:1061–71. 10.1007/s00262-014-1575-2
    1. Lövgren T, Wolodarski M, Wickström S, et al. . Complete and long-lasting clinical responses in immune checkpoint inhibitor-resistant, metastasized melanoma treated with adoptive T cell transfer combined with DC vaccination. Oncoimmunology 2020;9:1792058. 10.1080/2162402X.2020.1792058
    1. Cole DJ, Weil DP, Shamamian P, et al. . Identification of MART-1-specific T-cell receptors: T cells utilizing distinct T-cell receptor variable and joining regions recognize the same tumor epitope. Cancer Res 1994;54:5265–8.
    1. Johnson LA, Heemskerk B, Powell DJ, et al. . Gene transfer of tumor-reactive TCR confers both high avidity and tumor reactivity to nonreactive peripheral blood mononuclear cells and tumor-infiltrating lymphocytes. J Immunol 2006;177:6548–59. 10.4049/jimmunol.177.9.6548
    1. Fisher B, Packard BS, Read EJ, et al. . Tumor localization of adoptively transferred indium-111 labeled tumor infiltrating lymphocytes in patients with metastatic melanoma. J Clin Oncol 1989;7:250–61. 10.1200/JCO.1989.7.2.250
    1. Okada N, Tsujino M, Hagiwara Y, et al. . Administration route-dependent vaccine efficiency of murine dendritic cells pulsed with antigens. Br J Cancer 2001;84:1564–70. 10.1054/bjoc.2001.1801
    1. Lesterhuis WJ, de Vries IJM, Schreibelt G, et al. . Route of administration modulates the induction of dendritic cell vaccine-induced antigen-specific T cells in advanced melanoma patients. Clin Cancer Res 2011;17:5725–35. 10.1158/1078-0432.CCR-11-1261
    1. Fong L, Brockstedt D, Benike C, et al. . Dendritic cells injected via different routes induce immunity in cancer patients. J Immunol 2001;166:4254–9. 10.4049/jimmunol.166.6.4254
    1. Borch TH, Andersen R, Ellebaek E, et al. . Future role for adoptive T-cell therapy in checkpoint inhibitor-resistant metastatic melanoma. J Immunother Cancer 2020;8:e000668. 10.1136/jitc-2020-000668
    1. Sarnaik A, Khushalani NI, Chesney JA, et al. . Safety and efficacy of cryopreserved autologous tumor infiltrating lymphocyte therapy (LN-144, lifileucel) in advanced metastatic melanoma patients who progressed on multiple prior therapies including anti-PD-1. JCO 2019;37:2518. 10.1200/JCO.2019.37.15_suppl.2518
    1. Fetsch PA, Marincola FM, Filie A. Melanoma-Associated antigen recognized by T cells (MART-1): the advent of a preferred immunocytochemical antibody for the diagnosis of metastatic malignant melanoma with fine-needle aspiration. Cancer 1999;87.
    1. Pinto S, Sommermeyer D, Michel C, et al. . Misinitiation of intrathymic MART-1 transcription and biased TCR usage explain the high frequency of MART-1-specific T cells. Eur J Immunol 2014;44:2811–21. 10.1002/eji.201444499
    1. Dudley ME, Wunderlich JR, Robbins PF, et al. . Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes. Science 2002;298:850–4. 10.1126/science.1076514
    1. Kucuk A, Topkan E, Ikiz ED. Combination of stereotactic radiosurgery with immunotherapy: is it a new treatment option for brain metastases? Int J Oncol 2020:9–19.
    1. Chow MT, Möller A, Smyth MJ. Inflammation and immune surveillance in cancer. seminars in cancer biology. Elsevier, 2012.

Source: PubMed

3
Abonner