TNFα blockade overcomes resistance to anti-PD-1 in experimental melanoma

Florie Bertrand, Anne Montfort, Elie Marcheteau, Caroline Imbert, Julia Gilhodes, Thomas Filleron, Philippe Rochaix, Nathalie Andrieu-Abadie, Thierry Levade, Nicolas Meyer, Céline Colacios, Bruno Ségui, Florie Bertrand, Anne Montfort, Elie Marcheteau, Caroline Imbert, Julia Gilhodes, Thomas Filleron, Philippe Rochaix, Nathalie Andrieu-Abadie, Thierry Levade, Nicolas Meyer, Céline Colacios, Bruno Ségui

Abstract

Antibodies against programmed cell death-1 (PD-1) have considerably changed the treatment for melanoma. However, many patients do not display therapeutic response or eventually relapse. Moreover, patients treated with anti-PD-1 develop immune-related adverse events that can be cured with anti-tumor necrosis factor α (TNF) antibodies. Whether anti-TNF antibodies affect the anti-cancer immune response remains unknown. Our recent work has highlighted that TNFR1-dependent TNF signalling impairs the accumulation of CD8+ tumor-infiltrating T lymphocytes (CD8+ TILs) in mouse melanoma. Herein, our results indicate that TNF or TNFR1 blockade synergizes with anti-PD-1 on anti-cancer immune responses towards solid cancers. Mechanistically, TNF blockade prevents anti-PD-1-induced TIL cell death as well as PD-L1 and TIM-3 expression. TNF expression positively correlates with expression of PD-L1 and TIM-3 in human melanoma specimens. This study provides a strong rationale to develop a combination therapy based on the use of anti-PD-1 and anti-TNF in cancer patients.

Conflict of interest statement

Nicolas Meyer has worked as an investigator and/or consultant and/or speaker for: BMS, MSD, Amgen, Roche, GSK, Novartis, Pierre Fabre. The rest of the authors declare no competing financial interests.

Figures

Fig. 1
Fig. 1
TNF deficiency enhances anti-PD-1 response in a mouse melanoma model. C57BL/6 wild-type (WT) and TNF-deficient (TNF−/−) mice were intradermally and bilaterally grafted with 3 × 105 (ac) (n = 11 mice per group) or 1 × 106 (df) (n = 6 mice per group). B16K1 melanoma cells prior to intraperitoneal injection of anti-PD-1 antibodies (αPD-1, 10 mg kg−1) or a relevant isotype control (Iso, 10 mg kg−1) at days 6, 10 and 13. a and d Tumor volumes were determined with a calliper. Individual curves are depicted for each tumor. Numbers indicate complete tumor regression out of total tumors. Data are from three independent experiments. b and e Tumor volumes determined at the indicated days for individual tumors are depicted. Bars represent mean values ± s.e.m. (Mann–Whitney U test: *p < 0.05; **p < 0.01; ***p < 0.001). c and f Cumulative survival curves (Logrank test: *p < 0.05; **p < 0.01; ***p < 0.001). At day 60, surviving mice were challenged with a second B16K1 injection as indicated by the arrow; mice did not develop tumors and survived (c)
Fig. 2
Fig. 2
Immune cell infiltration of tumors from anti-PD-1 treated wild-type and TNF-deficient mice with established melanoma. ac, C57BL/6 wild-type (WT) and TNF-deficient (TNF−/−) mice were intradermally and bilaterally grafted with 1 × 106 B16K1 melanoma cells prior to intraperitoneal injection of anti-PD-1 antibodies (αPD-1, 10 mg kg−1) or vehicle (PBS) at day 7. a At day 10, mice were sacrificed and tumors were weighed. Data are means ± s.e.m. of at least 22 tumors per group from two independent experiments (Student’s t-test: *p < 0.05; ***p < 0.001). b and c TIL content was analysed by flow cytometry. The proportion of CD45+ and Thy1+ cells (b) CD4+ and CD8+ TILs (c) among total cells was determined. Data are means ± s.e.m. of at least 11 tumors per group from two independent experiments (Mann–Whitney U test: *p < 0.05; **p < 0.01; ***p < 0.001). d Proportion of dead CD8+ and CD4+ TILs from WT and TNF-deficient mice treated with anti-PD-1. Left panels: representative stainings. Right panels: values measured in 12 tumors per group from two independent experiments are represented as Tukey boxes (Student’s t-test: *p < 0.05; ***p < 0.001)
Fig. 3
Fig. 3
Anti-PD-1 triggers TIM-3 expression on CD8+ TILs in a TNF-dependent manner. WT and TNF-deficient mice were injected as described in the legend to Fig. 2. TIM-3 and PD-1 expression on CD8+ TILs was determined by flow cytometry at day 10. a Representative staining; values indicate the proportion of cells in the different quadrants. b Quantification of the proportion of TIM-3+ (left panel) and PD-1+ (right panel) among CD8+ TILs. c Quantification of the proportion of CD8+ TILs expressing or not TIM-3 and PD-1 (left panel), and proportion of cell death among CD8+ T cells of the indicated populations (right panel). Values in 5–6 mice per group from one experiment are represented as Tukey boxes (b: Student’s t-test: **p < 0.01; c: two-way Anova: ***p < 0.001). d Representative histograms of live/dead staining in CD8+ TILs expressing both TIM-3 and PD-1. Values are percentages of dead cells among the TIM-3+ PD-1+ CD8+ TILs
Fig. 4
Fig. 4
TNF induces TIM-3 expression on CD8+ T cells ex vivo. a WT or TNFR1-deficient CD8+ T cells were incubated with murine TNF for 2 days. TIM-3 expression was next analysed by flow cytometry. Upper panels: representative histograms. Lower panel: data are means ± s.e.m. of three independent experiments. b and c TILs from two human metastatic melanoma patients were cultured with or without autologous melanoma cells for two days in the presence 200 U ml−1 IL-2 +/−50 ng ml−1 human TNF. TIM-3 expression was next analysed by flow cytometry: histograms showing TIM-3 staining on TILs from patient 1 (b); bar graph depicting the fold increase in TIM-3 expression on TILs from patients 1 and 2 (c)
Fig. 5
Fig. 5
Anti-TNF treatment enhances anti-PD-1 response in a mouse melanoma model. C57BL/6 wild-type (WT) mice were intradermally grafted with 3 × 105 B16K1 melanoma cells followed by intraperitoneal injection of anti-TNF antibodies (αTNF, 10 mg kg−1) or vehicle (PBS) at days 6, 9, 13 and 16 and/or anti-PD-1 (10 mg kg−1) at days 6, 9 and 13 (n = 12 mice per group). Data are from two independent experiments. a Individual tumor volumes are depicted. Numbers indicate complete tumor regression out of total tumors. b Cumulative survival curves. At day 60, (arrow) surviving mice were challenged with a second B16K1 injection; these mice did not develop tumors and survived (Logrank test: **p < 0.01; ***p < 0.001). c and d C57BL/6 WT mice were intradermally and bilaterally grafted with 1 × 106 B16K1 melanoma cells prior to intraperitoneal injection of anti-TNF (αTNF, 10 mg kg−1) or vehicle at days 5 and 7, and with anti-PD-1 antibodies (αPD-1, 10 mg kg−1) or vehicle (PBS) at day 7. At day 10, CD8+ TILs (c) and the proportion of cell death in CD8+ TILs (d) were analysed by flow cytometry. Data from at least 5 tumors per group are represented as Tukey boxes (Mann–Whitney U test: *p < 0.05; **p < 0.01)
Fig. 6
Fig. 6
TNF blockade prevents TIM-3 up-regulation on TILs in response to anti-PD-1. WT mice were injected as described in the legend to Fig. 5. ac TIM-3 expression level on CD8+ TILs and CD4+ TILs was determined by flow cytometry on tumors from WT mice at day 10 after B16K1 graft following injection of vehicle (PBS), anti-PD-1 (αPD-1), anti-TNF (αTNF) or a combination of both. Representative stainings (a) mean fluorescence intensity (MFI) of TIM-3 on CD8+ (b) and CD4+ TILs (c) measured in at least 5 tumors per group from one experiment are represented as Tukey boxes. b, c: Mann–Whitney U test: *p < 0.05; **p < 0.01; ***p < 0.001). d and e C57BL/6 WT mice were intradermally and bilaterally grafted with 3 × 105 B16K1 melanoma cells prior to injection with vehicle, anti-PD-1 alone or combined with anti-TNF and/or anti-TIM-3 (10 mg kg−1 of each antibody) at days 6, 10, and 13 (d) (PBS (n = 12), αPD-1 (n = 9), αPD-1 + αTNF (n = 14), αPD-1 + αTIM-3 (n = 13), αPD-1 + αTNF + αTIM-3 (n = 15), data from two experiments) or at days 13, 16, and 19 (e) (n = 10 mice per group, data from one representative experiment out of two). Tumor volumes were determined with a calliper. Data are means ± s.e.m. (d) and e, two-way Anova: **p < 0.01, ***p < 0.001 at day 35 (d) and day 21 (e); p = 0.08 at day 35 when comparing anti-PD-1 alone and anti-PD-1 plus anti-TIM-3 (d)
Fig. 7
Fig. 7
TNF expression is associated with an immune escape gene signature in human metastatic melanoma. a Heatmap for a selected list of genes encoding immune escape proteins in metastatic melanoma patients from the TCGA melanoma cohort (n = 342), exhibiting high (80th percentile) and low (20th percentile) TNF expression in melanoma samples. Genes were clustered using a Euclidean distant matrix and average linkage clustering. b and c Correlation analysis between the expression of HAVCR2 (encoding TIM-3), PDCD1LG1 (encoding PD-L1), PDCD1LG2 (encoding PD-L2) and TNFA (encoding TNF) in melanoma samples from metastatic melanoma patients from the TCGA cohort (n = 342) (b) and from patients treated with anti-PD-1 (our analysis of data published in ref.) (n = 13) (c)

References

    1. Bertrand F, Colacios C, Segui B. TNF-R1, an immune checkpoint in melanoma? Genes Cancer. 2015;6:369–370.
    1. Bertrand F, et al. Targeting TNF alpha as a novel strategy to enhance CD8+ T cell-dependent immune response in melanoma? Oncoimmunology. 2016;5:e1068495. doi: 10.1080/2162402X.2015.1068495.
    1. Balkwill F. Tumour necrosis factor and cancer. Nat. Rev. Cancer. 2009;9:361–371. doi: 10.1038/nrc2628.
    1. Carswell EA, et al. An endotoxin-induced serum factor that causes necrosis of tumors. Proc. Natl. Acad. Sci. USA. 1975;72:3666–3670. doi: 10.1073/pnas.72.9.3666.
    1. Ratner A, Clark WR. Role of TNF-alpha in CD8+ cytotoxic T lymphocyte-mediated lysis. J. Immunol. 1993;150:4303–4314.
    1. Chopra M, et al. Tumor necrosis factor receptor 2-dependent homeostasis of regulatory T cells as a player in TNF-induced experimental metastasis. Carcinogenesis. 2013;34:1296–1303. doi: 10.1093/carcin/bgt038.
    1. Okubo Y, Mera T, Wang L, Faustman DL. Homogeneous expansion of human T-regulatory cells via tumor necrosis factor receptor 2. Sci. Rep. 2013;3:3153. doi: 10.1038/srep03153.
    1. Schioppa T, et al. B regulatory cells and the tumor-promoting actions of TNF-alpha during squamous carcinogenesis. Proc. Natl. Acad. Sci. USA. 2011;108:10662–10667. doi: 10.1073/pnas.1100994108.
    1. Zhao X, et al. TNF signaling drives myeloid-derived suppressor cell accumulation. J. Clin. Invest. 2012;122:4094–4104. doi: 10.1172/JCI64115.
    1. Sade-Feldman M, et al. Tumor necrosis factor-alpha blocks differentiation and enhances suppressive activity of immature myeloid cells during chronic inflammation. Immunity. 2013;38:541–554. doi: 10.1016/j.immuni.2013.02.007.
    1. Beyer M, et al. Tumor-necrosis factor impairs CD4(+) T cell-mediated immunological control in chronic viral infection. Nat. Immunol. 2016;17:593–603. doi: 10.1038/ni.3399.
    1. Chen X, Baumel M, Mannel DN, Howard OM, Oppenheim JJ. Interaction of TNF with TNF receptor type 2 promotes expansion and function of mouse CD4+ CD25+ T regulatory cells. J. Immunol. 2007;179:154–161. doi: 10.4049/jimmunol.179.1.154.
    1. Zheng L, et al. Induction of apoptosis in mature T cells by tumour necrosis factor. Nature. 1995;377:348–351. doi: 10.1038/377348a0.
    1. Bertrand F, et al. Blocking tumor necrosis factor alpha enhances CD8 T-cell-dependent immunity in experimental melanoma. Cancer Res. 2015;75:2619–2628. doi: 10.1158/0008-5472.CAN-14-2524.
    1. Donia M, et al. Aberrant expression of MHC class II in melanoma attracts inflammatory tumor-specific CD4+ T- cells, which dampen CD8+ T-cell antitumor reactivity. Cancer Res. 2015;75:3747–3759. doi: 10.1158/0008-5472.CAN-14-2956.
    1. Landsberg J, et al. Melanomas resist T-cell therapy through inflammation-induced reversible dedifferentiation. Nature. 2012;490:412–416. doi: 10.1038/nature11538.
    1. Lim SO, et al. Deubiquitination and Stabilization of PD-L1 by CSN5. Cancer Cell. 2016;30:925–939. doi: 10.1016/j.ccell.2016.10.010.
    1. Larkin J, et al. Combined nivolumab and Ipilimumab or monotherapy in untreated melanoma. N. Engl. J. Med. 2015;373:23–34. doi: 10.1056/NEJMoa1504030.
    1. Robert C, et al. Nivolumab in previously untreated melanoma without BRAF mutation. N. Engl. J. Med. 2015;372:320–330. doi: 10.1056/NEJMoa1412082.
    1. Robert C, et al. Pembrolizumab versus Ipilimumab in advanced melanoma. N. Engl. J. Med. 2015;372:2521–2532. doi: 10.1056/NEJMoa1503093.
    1. Postow MA, Callahan MK, Wolchok JD. Immune checkpoint blockade in cancer therapy. J. Clin. Oncol. 2015;33:1974–1982. doi: 10.1200/JCO.2014.59.4358.
    1. Koyama S, et al. Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints. Nat. Commun. 2016;7:10501. doi: 10.1038/ncomms10501.
    1. Chen PL, et al. Analysis of immune signatures in longitudinal tumor samples yields insight into biomarkers of response and mechanisms of resistance to immune checkpoint blockade. Cancer Discov. 2016;6:827–837. doi: 10.1158/-15-1545.
    1. Spain L, Diem S, Larkin J. Management of toxicities of immune checkpoint inhibitors. Cancer Treat. Rev. 2016;44:51–60. doi: 10.1016/j.ctrv.2016.02.001.
    1. Rizvi NA, et al. Activity and safety of nivolumab, an anti-PD-1 immune checkpoint inhibitor, for patients with advanced, refractory squamous non-small-cell lung cancer (CheckMate 063): a phase 2, single-arm trial. Lancet Oncol. 2015;16:257–265. doi: 10.1016/S1470-2045(15)70054-9.
    1. Borghaei H, et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N. Engl. J. Med. 2015;373:1627–1639. doi: 10.1056/NEJMoa1507643.
    1. Sabatos CA, et al. Interaction of Tim-3 and Tim-3 ligand regulates T helper type 1 responses and induction of peripheral tolerance. Nat. Immunol. 2003;4:1102–1110. doi: 10.1038/ni988.
    1. Fourcade J, et al. Upregulation of Tim-3 and PD-1 expression is associated with tumor antigen-specific CD8+ T cell dysfunction in melanoma patients. J. Exp. Med. 2010;207:2175–2186. doi: 10.1084/jem.20100637.
    1. Borghi A, Verstrepen L, Beyaert R. TRAF2 multitasking in TNF receptor-induced signaling to NF-kappaB, MAP kinases and cell death. Biochem. Pharmacol. 2016;116:1–10. doi: 10.1016/j.bcp.2016.03.009.
    1. Yoon SJ, et al. Activation of mitogen activated protein kinase-Erk kinase (MEK) increases T cell immunoglobulin mucin domain-3 (TIM-3) transcription in human T lymphocytes and a human mast cell line. Mol. Immunol. 2011;48:1778–1783. doi: 10.1016/j.molimm.2011.05.004.
    1. Mujib S, et al. Antigen-independent induction of Tim-3 expression on human T cells by the common gamma-chain cytokines IL-2, IL-7, IL-15, and IL-21 is associated with proliferation and is dependent on the phosphoinositide 3-kinase pathway. J. Immunol. 2012;188:3745–3756. doi: 10.4049/jimmunol.1102609.
    1. Cancer Genome Atlas Network. Genomic classification of cutaneous melanoma. Cell161, 1681–1696 (2015).
    1. Tumeh PC, et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 2014;515:568–571. doi: 10.1038/nature13954.
    1. Sharma P, Hu-Lieskovan S, Wargo JA, Ribas A. Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell. 2017;168:707–723. doi: 10.1016/j.cell.2017.01.017.
    1. Daud AI, et al. Tumor immune profiling predicts response to anti-PD-1 therapy in human melanoma. J. Clin. Invest. 2016;126:3447–3452. doi: 10.1172/JCI87324.
    1. Sakuishi K, et al. Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity. J. Exp. Med. 2010;207:2187–2194. doi: 10.1084/jem.20100643.
    1. Ngiow SF, et al. Anti-TIM3 antibody promotes T cell IFN-gamma-mediated antitumor immunity and suppresses established tumors. Cancer Res. 2011;71:3540–3551. doi: 10.1158/0008-5472.CAN-11-0096.
    1. Zhu C, et al. An IL-27/NFIL3 signalling axis drives Tim-3 and IL-10 expression and T-cell dysfunction. Nat. Commun. 2015;6:6072. doi: 10.1038/ncomms7072.
    1. Zingg D, et al. The histone methyltransferase Ezh2 controls mechanisms of adaptive resistance to tumor immunotherapy. Cell Rep. 2017;20:854–867. doi: 10.1016/j.celrep.2017.07.007.
    1. Reinhardt J, et al. MAPK signaling and inflammation link melanoma phenotype switching to induction of CD73 during immunotherapy. Cancer Res. 2017;77:4697–4709. doi: 10.1158/0008-5472.CAN-17-0395.
    1. Galluzzi L, et al. Essential versus accessory aspects of cell death: recommendations of the NCCD 2015. Cell. Death. Differ. 2015;22:58–73. doi: 10.1038/cdd.2014.137.
    1. Porgador A, Feldman M, Eisenbach L. H-2Kb transfection of B16 melanoma cells results in reduced tumourigenicity and metastatic competence. J. Immunogenet. 1989;16:291–303. doi: 10.1111/j.1744-313X.1989.tb00475.x.
    1. Pequeux C, et al. Stromal estrogen receptor-alpha promotes tumor growth by normalizing an increased angiogenesis. Cancer Res. 2012;72:3010–3019. doi: 10.1158/0008-5472.CAN-11-3768.
    1. Sheehan KC, et al. Monoclonal antibodies specific for murine p55 and p75 tumor necrosis factor receptors: identification of a novel in vivo role for p75. J. Exp. Med. 1995;181:607–617. doi: 10.1084/jem.181.2.607.

Source: PubMed

Подписаться