Adoptive cell therapy with tumor-infiltrating lymphocytes supported by checkpoint inhibition across multiple solid cancer types

Anders Handrup Kverneland, Christopher Aled Chamberlain, Troels Holz Borch, Morten Nielsen, Sofie Kirial Mørk, Julie Westerlin Kjeldsen, Cathrine Lund Lorentzen, Lise Pyndt Jørgensen, Lene Buhl Riis, Christina Westmose Yde, Özcan Met, Marco Donia, Inge Marie Svane, Anders Handrup Kverneland, Christopher Aled Chamberlain, Troels Holz Borch, Morten Nielsen, Sofie Kirial Mørk, Julie Westerlin Kjeldsen, Cathrine Lund Lorentzen, Lise Pyndt Jørgensen, Lene Buhl Riis, Christina Westmose Yde, Özcan Met, Marco Donia, Inge Marie Svane

Abstract

Background: Adoptive cell therapy (ACT) with tumor-infiltrating lymphocytes (TILs) has shown remarkable results in malignant melanoma (MM), while studies on the potential in other cancer diagnoses are sparse. Further, the prospect of using checkpoint inhibitors (CPIs) to support TIL production and therapy remains to be explored.

Study design: TIL-based ACT with CPIs was evaluated in a clinical phase I/II trial. Ipilimumab (3 mg/kg) was administered prior to tumor resection and nivolumab (3 mg/kg, every 2 weeks ×4) in relation to TIL infusion. Preconditioning chemotherapy was given before TIL infusion and followed by low-dose (2 10e6 international units (UI) ×1 subcutaneous for 14 days) interleukin-2 stimulation.

Results: Twenty-five patients covering 10 different cancer diagnoses were treated with in vitro expanded TILs. Expansion of TILs was successful in 97% of recruited patients. Five patients had sizeable tumor regressions of 30%-63%, including two confirmed partial responses in patients with head-and-neck cancer and cholangiocarcinoma. Safety and feasibility were comparable to MM trials of ACT with the addition of expected CPI toxicity. In an exploratory analysis, tumor mutational burden and expression of the alpha-integrin CD103 (p=0.025) were associated with increased disease control. In vitro tumor reactivity was seen in both patients with an objective response and was associated with regressions in tumor size (p=0.028).

Conclusion: High success rates of TIL expansion were demonstrated across multiple solid cancers. TIL ACTs were found feasible, independent of previous therapy. Tumor regressions after ACT combined with CPIs were demonstrated in several cancer types supported by in vitro antitumor reactivity of the TILs.

Trial registration numbers: NCT03296137, and EudraCT No. 2017-002323-25.

Keywords: adoptive; immunotherapy; lymphocytes; tumor-infiltrating.

Conflict of interest statement

Competing interests: No, there are no competing interests.

© 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
Therapy regimen. Overview of the scheduled adoptive cell therapy in combination with conditioning chemotherapy, low-dose interleukin-2 and checkpoint inhibition. Created with BioRender.com. TIL, tumor-infiltrating lymphocyte.
Figure 2
Figure 2
Clinical efficacy and OS. (A) Best change in target lesion sum of all patients. The patients with disease control (PR and/or SD >4.5 months) are marked with light gray and listed with cancer diagnoses. (B) Swimmer’s plot showing the duration of response in the individual patients and indicating start of tumor regressions >30% of target lesions. (C) Kaplan-Meier plot of the OS in the benefit and non-benefit groups. The survival curves are compared with a log-rank test. *The patients with antitumor reactive cells in vitro. BOR, best overall response; CC, cholangiocarcinoma; CRC, colorectal cancer; HNSCC, head-and-neck squamous cell carcinoma; MM, malignant melanoma; OS, overall survival; PD, progressive disease; PR, partial response; SD, stable disease.
Figure 3
Figure 3
Computerized tomography (CT) scan images from patients with an objective response to therapy. Two patients had a partial response according to RECIST V.1.1. Images from baseline and 6 weeks after cell infusion in patient 8 with cholangiocarcinoma and patient 19 with HNSCC are shown. HNSCC, head-and-neck squamous cell carcinoma.
Figure 4
Figure 4
Phenotype analysis of TILs throughout expansion. (A) Single marker expression analysis of the unstimulated TILs before (unstimulated TILs), after initial in vitro expansion (young TILs) and after the REP TILs. Expression is shown as the fraction of CD3+ cells. (B) Comparison of CD103+ and CD8+CD103+TILs during the in vitro expansion according to disease control (partial response and/or stable disease >4.5 months). The difference was tested for significance with a non-parametric Mann-Whitney U test. CD4 expression is low in unstimulated TILs as the dissociation agent (collagenase IV) is known to cleave CD4. CM, central memory; EM, effector memory; EMRA, terminally differentiated effector memory; ns, not significant; REP, rapid expansion protocol; TIL, tumor-infiltrating lymphocyte.
Figure 5
Figure 5
In vitro antitumor reactivity of infusion product. Expression of reactivity markers in REP TILs from eight patients where antitumor in vitro reactivity was seen in more than 0.5% of CD3+ cells. The REP TILs are divided according to the dominant T-cell subset: either CD4 or CD8. Reactivity was assessed with intracellular flow cytometry after coculture with unstimulated autologous tumor material. REP TILs with expression of at least two markers (TNF-α, IFN-γ, CD107a or CD137) in more than 0.5% of all CD3+ cells were considered reactive. *Patients in the disease control group. **Patients in the disease control group with an objective response. CC, cholangiocarcinoma; CRC, colorectal cancer; HNSCC, head-and-neck squamous cell cancer; IFN-γ, interferon gamma; MM, malignant melanoma; REP, rapid expansion protocol; TIL, tumor-infiltrating lymphocyte; TNF-α, tumor necrosis factor-alpha.
Figure 6
Figure 6
TMB versus disease control of ACT. The resected tumor sample used for in vitro TIL expansion was examined for TMB. The TMB of 24/25 of the patients treated with ACT is shown together with the best overall response and the patients are divided according to disease control rate (PR and/or SD >4.5 months). Non-PR indicates SD or PD. ACC, adrenocorticocarcinoma; ACT, adoptive cell therapy; CC, cholangiocarcinoma; CRC, colorectal cancer; HNSCC, head-and-neck squamous cell carcinoma; MM, malignant melanoma; NCSLC, non-small cell lung cancer; PD, progressive disease; PR, partial response; SD, stable disease; TIL, tumor-infiltrating lymphocyte; TMB, tumor mutational burden.

References

    1. Dafni U, Michielin O, Lluesma SM, et al. . Efficacy of adoptive therapy with tumor-infiltrating lymphocytes and recombinant interleukin-2 in advanced cutaneous melanoma: a systematic review and meta-analysis. Ann Oncol 2019;30:1902–13. 10.1093/annonc/mdz398
    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. 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. 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. Hodi FS, O'Day SJ, McDermott DF, et al. . Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 2010;363:711–23. 10.1056/NEJMoa1003466
    1. Tran E, Turcotte S, Gros A. Cancer immunotherapy based on mutation-specific CD4+ T cells in a patient with epithelial cancer. Science 2014;344:641–5.
    1. Stevanović S, Draper LM, Langhan MM, et al. . Complete regression of metastatic cervical cancer after treatment with human papillomavirus-targeted tumor-infiltrating T cells. J Clin Oncol 2015;33:1543–50. 10.1200/JCO.2014.58.9093
    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. Kverneland AH, Pedersen M, Westergaard MCW, et al. . Adoptive cell therapy in combination with checkpoint inhibitors in ovarian cancer. Oncotarget 2020;11:2092–105. 10.18632/oncotarget.27604
    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. Maude SL, Laetsch TW, Buechner J, et al. . Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med 2018;378:439–48. 10.1056/NEJMoa1709866
    1. Yunger S, Bar El A, Zeltzer L-A, et al. . Tumor-infiltrating lymphocytes from human prostate tumors reveal anti-tumor reactivity and potential for adoptive cell therapy. Oncoimmunology 2019;8:e1672494. 10.1080/2162402X.2019.1672494
    1. Nielsen M, Krarup-Hansen A, Hovgaard D, et al. . In vitro 4-1BB stimulation promotes expansion of CD8+ tumor-infiltrating lymphocytes from various sarcoma subtypes. Cancer Immunol Immunother 2020;69:2179–91. 10.1007/s00262-020-02568-x
    1. Ben-Avi R, Farhi R, Ben-Nun A, et al. . Establishment of adoptive cell therapy with tumor infiltrating lymphocytes for non-small cell lung cancer patients. Cancer Immunol Immunother 2018;67:1221–30. 10.1007/s00262-018-2174-4
    1. Junker N, Andersen MH, Wenandy L, et al. . Bimodal ex vivo expansion of T cells from patients with head and neck squamous cell carcinoma: a prerequisite for adoptive cell transfer. Cytotherapy 2011;13:822–34. 10.3109/14653249.2011.563291
    1. Andersen R, Donia M, Westergaard MCW, et al. . Tumor infiltrating lymphocyte therapy for ovarian cancer and renal cell carcinoma. Hum Vaccin Immunother 2015;11:2790–5. 10.1080/21645515.2015.1075106
    1. Harao M, Forget M-A, Roszik J, et al. . 4-1BB-enhanced expansion of CD8+ TIL from triple-negative breast cancer unveils mutation-specific CD8+ T cells. Cancer Immunol Res 2017;5:439–45. 10.1158/2326-6066.CIR-16-0364
    1. Poch M, Hall M, Joerger A, et al. . Expansion of tumor infiltrating lymphocytes (TIL) from bladder cancer. Oncoimmunology 2018;7:1–7. 10.1080/2162402X.2018.1476816
    1. Hall M, Liu H, Malafa M, et al. . Expansion of tumor-infiltrating lymphocytes (TIL) from human pancreatic tumors. J Immunother Cancer 2016;4:61. 10.1186/s40425-016-0164-7
    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. Dudley ME, Wunderlich JR, Yang JC, et al. . A phase I study of nonmyeloablative chemotherapy and adoptive transfer of autologous tumor antigen-specific T lymphocytes in patients with metastatic melanoma. J Immunother 2002;25:243–51.
    1. Andersen R, Donia M, Ellebaek E, et al. . Long-Lasting complete responses in patients with metastatic melanoma after adoptive cell therapy with tumor-infiltrating lymphocytes and an attenuated IL2 regimen. Clin Cancer Res 2016;22:3734–45. 10.1158/1078-0432.CCR-15-1879
    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. 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. Hodi FS, Chiarion-Sileni V, Gonzalez R, et al. . Nivolumab plus ipilimumab or nivolumab alone versus ipilimumab alone in advanced melanoma (CheckMate 067): 4-year outcomes of a multicentre, randomised, phase 3 trial. Lancet Oncol 2018;19:1480–92. 10.1016/S1470-2045(18)30700-9
    1. Curran MA, Montalvo W, Yagita H, et al. . PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc Natl Acad Sci U S A 2010;107:4275–80. 10.1073/pnas.0915174107
    1. Kodumudi KN, Siegel J, Weber AM, et al. . Immune checkpoint blockade to improve tumor infiltrating lymphocytes for adoptive cell therapy. PLoS One 2016;11:1–13. 10.1371/journal.pone.0153053
    1. Bjoern J, Lyngaa R, Andersen R, et al. . Influence of ipilimumab on expanded tumour derived T cells from patients with metastatic melanoma. Oncotarget 2017;8:27062–74. 10.18632/oncotarget.16003
    1. Mullinax JE, Hall M, Prabhakaran S, et al. . Combination of ipilimumab and adoptive cell therapy with tumor-infiltrating lymphocytes for patients with metastatic melanoma. Front Oncol 2018;8:44. 10.3389/fonc.2018.00044
    1. Zippel D, Friedman-Eldar O, Rayman S, et al. . Tissue harvesting for adoptive tumor infiltrating lymphocyte therapy in metastatic melanoma. Anticancer Res 2019;39:4995–5001. 10.21873/anticanres.13689
    1. Friese C, Harbst K, Borch TH, et al. . CTLA-4 blockade boosts the expansion of tumor-reactive CD8+ tumor-infiltrating lymphocytes in ovarian cancer. Sci Rep 2020;10:3914. 10.1038/s41598-020-60738-4
    1. Donia M, Kjeldsen JW, Andersen R, et al. . PD-1+polyfunctional T cells dominate the periphery after tumor-infiltrating lymphocyte therapy for cancer. Clin Cancer Res 2017;23:5779–88. 10.1158/1078-0432.CCR-16-1692
    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. 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. 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;8:1–11. 10.1136/jitc-2020-000848
    1. Donia M, Larsen SM, Met O, et al. . Simplified protocol for clinical-grade tumor-infiltrating lymphocyte manufacturing with use of the wave bioreactor. Cytotherapy 2014;16:1117–20. 10.1016/j.jcyt.2014.02.004
    1. Westergaard MCW, Andersen R, Chong C, et al. . Tumour-reactive T cell subsets in the microenvironment of ovarian cancer. Br J Cancer 2019;120:424–34. 10.1038/s41416-019-0384-y
    1. Kverneland AH, Borch TH, Granhøj J, et al. . Bone marrow toxicity and immune reconstitution in melanoma and non-melanoma solid cancer patients after non-myeloablative conditioning with chemotherapy and checkpoint inhibition. Cytotherapy 2021;23:724–9. 10.1016/j.jcyt.2021.03.003
    1. Hellmann MD, Ciuleanu T-E, Pluzanski A, et al. . Nivolumab plus ipilimumab in lung cancer with a high tumor mutational burden. N Engl J Med 2018;378:2093–104. 10.1056/NEJMoa1801946
    1. Ellebaek E, Iversen TZ, Junker N. Adoptive cell therapy with autologous tumor infiltrating lymphocytes and low-dose interleukin-2 in metastatic melanoma patients. J Transl Med 2012;10:1–12.
    1. Nguyen LT, Saibil SD, Sotov V, et al. . Phase II clinical trial of adoptive cell therapy for patients with metastatic melanoma with autologous tumor-infiltrating lymphocytes and low-dose interleukin-2. Cancer Immunol Immunother 2019;68:773–85. 10.1007/s00262-019-02307-x
    1. Jakubowski CD, Azad NS. Immune checkpoint inhibitor therapy in biliary tract cancer (cholangiocarcinoma). Chin Clin Oncol 2020;9:2. 10.21037/cco.2019.12.10
    1. Ren L, Leisegang M, Deng B, et al. . Identification of neoantigen-specific T cells and their targets: implications for immunotherapy of head and neck squamous cell carcinoma. Oncoimmunology 2019;8:1–10. 10.1080/2162402X.2019.1568813
    1. Hald J, Rasmussen N, Claesson MH. Tumour-infiltrating lymphocytes mediate lysis of autologous squamous cell carcinomas of the head and neck. Cancer Immunol Immunother 1995;41:243–50. 10.1007/BF01516999
    1. Ferris RL, Licitra L. Pd-1 immunotherapy for recurrent or metastatic HNSCC. Lancet 2019;394:1882–4. 10.1016/S0140-6736(19)32539-5
    1. Besser MJ, Itzhaki O, Ben-Betzalel G, et al. . Comprehensive single institute experience with melanoma TIL: long term clinical results, toxicity profile, and prognostic factors of response. Mol Carcinog 2020;59:736–44. 10.1002/mc.23193
    1. Duhen T, Duhen R, Montler R, et al. . Co-expression of CD39 and CD103 identifies tumor-reactive CD8 T cells in human solid tumors. Nat Commun 2018;9:2724. 10.1038/s41467-018-05072-0
    1. Van Den Bulk J, Verdegaal EME, Ruano D. Neoantigen-specific immunity in low mutation burden colorectal cancers of the consensus molecular subtype 4. Genome Med 2019;11:1–15.
    1. Kortekaas KE, Santegoets SJ, Sturm G, et al. . CD39 identifies the CD4+ tumor-specific T-cell population in human cancer. Cancer Immunol Res 2020;8:1311–21. 10.1158/2326-6066.CIR-20-0270
    1. Djenidi F, Adam J, Goubar A, et al. . CD8+CD103+ tumor-infiltrating lymphocytes are tumor-specific tissue-resident memory T cells and a prognostic factor for survival in lung cancer patients. J Immunol 2015;194:3475–86. 10.4049/jimmunol.1402711
    1. Webb JR, Milne K, Nelson BH. PD-1 and CD103 are widely coexpressed on prognostically favorable intraepithelial CD8 T cells in human ovarian cancer. Cancer Immunol Res 2015;3:926–35. 10.1158/2326-6066.CIR-14-0239
    1. Gupta PK, Godec J, Wolski D, et al. . CD39 expression identifies terminally exhausted CD8+ T cells. PLoS Pathog 2015;11:e1005177. 10.1371/journal.ppat.1005177
    1. Simoni Y, Becht E, Fehlings M, et al. . Bystander CD8+ T cells are abundant and phenotypically distinct in human tumour infiltrates. Nature 2018;557:575–9. 10.1038/s41586-018-0130-2
    1. Krishna S, Lowery FJ, Copeland AR, et al. . Stem-like CD8 T cells mediate response of adoptive cell immunotherapy against human cancer. Science 2020;370:1328–34. 10.1126/science.abb9847
    1. Allard D, Allard B, Stagg J. On the mechanism of anti-CD39 immune checkpoint therapy. J Immunother Cancer 2020;8:1–11.
    1. Yu X, Huang X, Chen X, et al. . Characterization of a novel anti-human lymphocyte activation gene 3 (LAG-3) antibody for cancer immunotherapy. MAbs 2019;11:1139–48. 10.1080/19420862.2019.1629239
    1. Rizvi NA, Hellmann MD, Snyder A, et al. . Cancer immunology. mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015;348:124–8. 10.1126/science.aaa1348
    1. Yarchoan M, Hopkins A, Jaffee EM. Tumor mutational burden and response rate to PD-1 inhibition. N Engl J Med 2017;377:2500–1. 10.1056/NEJMc1713444
    1. Galon J, Bruni D. Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat Rev Drug Discov 2019;18:197–218. 10.1038/s41573-018-0007-y
    1. Wang X, Li M. Correlate tumor mutation burden with immune signatures in human cancers. BMC Immunol 2019;20:1–13.
    1. Carneiro BA, Konda B, Costa RB, et al. . Nivolumab in metastatic adrenocortical carcinoma: results of a phase 2 trial. J Clin Endocrinol Metab 2019;104:6193–200. 10.1210/jc.2019-00600
    1. Klein O, Senko C, Carlino MS, et al. . Combination immunotherapy with ipilimumab and nivolumab in patients with advanced adrenocortical carcinoma: a subgroup analysis of CA209-538. Oncoimmunology 2021;10:1908771. 10.1080/2162402X.2021.1908771

Source: PubMed

3
Subscribe