Beyond the concept of cold and hot tumors for the development of novel predictive biomarkers and the rational design of immunotherapy combination
Eleonore De Guillebon
Department of Medical Oncology, Institut Curie, Paris, France
Search for more papers by this authorAntoine Dardenne
Department of Gastro-enterology and Gastro-intestinal Oncology, Hopital Européen Georges Pompidou, APHP, Paris, France
Search for more papers by this authorAntonin Saldmann
Université de Paris, PARCC, INSERM, Paris, France
Department of Immunology, AP-HP, Hopital Européen Georges Pompidou, Paris, France
Search for more papers by this authorSylvie Séguier
Université de Paris, PARCC, INSERM, Paris, France
Faculté de Chirurgie Dentaire, Hôpital Louis Mourier, Montrouge, France
Search for more papers by this authorCeleste Lebbe
Department of Dermatology, Saint-Louis University Hospital, Paris, France
Université de Paris, INSERM U976, Paris, France
Search for more papers by this authorCorresponding Author
Eric Tartour
Université de Paris, PARCC, INSERM, Paris, France
Department of Immunology, AP-HP, Hopital Européen Georges Pompidou, Paris, France
Equipe Labellisée Ligue Contre le Cancer, Paris, France
Correspondence to: Eleonore de Guillebon, E-mail: [email protected] or Eric Tartour, E-mail: [email protected]Search for more papers by this authorEleonore De Guillebon
Department of Medical Oncology, Institut Curie, Paris, France
Search for more papers by this authorAntoine Dardenne
Department of Gastro-enterology and Gastro-intestinal Oncology, Hopital Européen Georges Pompidou, APHP, Paris, France
Search for more papers by this authorAntonin Saldmann
Université de Paris, PARCC, INSERM, Paris, France
Department of Immunology, AP-HP, Hopital Européen Georges Pompidou, Paris, France
Search for more papers by this authorSylvie Séguier
Université de Paris, PARCC, INSERM, Paris, France
Faculté de Chirurgie Dentaire, Hôpital Louis Mourier, Montrouge, France
Search for more papers by this authorCeleste Lebbe
Department of Dermatology, Saint-Louis University Hospital, Paris, France
Université de Paris, INSERM U976, Paris, France
Search for more papers by this authorCorresponding Author
Eric Tartour
Université de Paris, PARCC, INSERM, Paris, France
Department of Immunology, AP-HP, Hopital Européen Georges Pompidou, Paris, France
Equipe Labellisée Ligue Contre le Cancer, Paris, France
Correspondence to: Eleonore de Guillebon, E-mail: [email protected] or Eric Tartour, E-mail: [email protected]Search for more papers by this authorAbstract
Immunotherapy has revolutionized the management of cancers. At the end of 2018, 1,716 clinical trials assessed regimen that combine program death-1 (PD-1)/program death ligand-1 (PD-L1) blockers with other cancer therapies (tyrosine kinase inhibitor, chemotherapy and radiotherapy). There is a contrast between these clinical dynamics and the difficulty of identifying biomarkers to better select patients that could benefit from immunotherapy. In this context, different tumor classifications have been proposed to try to better stratify patients. They rely on the characteristics of the tumor microenvironment and led first to divide them into hot and cold tumors. In this review, we aim to demonstrate the limitations of this classification focusing on the differential significance of subpopulations of intratumor CD8 + T cells. We also underline novel mechanisms of resistance to anti-PD-1/PD-L1 blockade, focusing on myeloid cells, hypoxia and tumor immunoediting under treatment. Understanding the mechanisms of resistance to immune-checkpoint inhibitor is indeed a powerful research driver that allows further identification of novel biomarkers, drug development and bring a rational to innovative therapeutic combinations.
Conflict of interest
Celeste Lebbe declares potential financial conflict of interest: Research grants (BMS, Roche), Honoraria (BMS, MSD, Novartis, Amgen, Roche, Pierre-Fabre, Pfizer, Incyte), Consultancy (BMS, MSD, Novartis, Amgen, Roche, Merck-Serono, Sanofi), Speakers bureau (BMS, MSD, Novartis, Amgen, Roche), Travel accommodations-meetings (BMS), Advisory board (BMS, Novartis, Amgen, Roche), Board (Avantis), advisory role (BMS, MSD, Novartis, Roche). Eric Tartour declares a potential conflict of interest: Research grants (Servier, Vaxeal), Honoraria (BMS, Merck-MSD), Advisory Board (BMS, Astra-Zeneca).
Supporting Information
Filename | Description |
---|---|
ijc32889-sup-0001-TableS1.pdfPDF document, 22.3 KB | Table S1 Description of the various IFNγ signatures |
Please note: The publisher is not responsible for the content or functionality of any supporting information supplied by the authors. Any queries (other than missing content) should be directed to the corresponding author for the article.
References
- 1Robert C, Long GV, Brady B, et al. Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med 2015; 372: 320–30.
- 2Rischin D, Harington K, Greil R, et al. Protocol-specified final analysis of the phase 3 KEYNOTE-048 trial of pembrolizumab (pembro) as first-line therapy for recurrent/metastatic head and neck squamous cell carcinoma (R/M HNSCC). J Clin Oncol 2019; 37: 6000.
- 3Motzer RJ, Tannir NM, McDermott DF, et al. Nivolumab plus Ipilimumab versus Sunitinib in advanced renal-cell carcinoma. N Engl J Med 2018; 378: 1277–90.
- 4Lenz H, Van Cutsem E, Limon M, et al. Durable clinical benefit with nivolumab (NIVO) plus low-dose ipilimumab (IPI) as first-line therapy in microsatellite instability-high/mismatch repair deficient (MSI-H/dMMR) metastatic colorectal cancer (mCRC). Ann Oncol 2018; 29:LBA18_PR.
- 5Gandhi L, Rodriguez-Abreu D, Gadgeel S, et al. Pembrolizumab plus chemotherapy in metastatic non-small-cell lung cancer. N Engl J Med 2018; 378: 2078–92.
- 6Rini BI, Plimack ER, Stus V, et al. Pembrolizumab plus Axitinib versus Sunitinib for advanced renal-cell carcinoma. N Engl J Med 2019; 380: 1116–27.
- 7Motzer RJ, Penkov K, Haanen J, et al. Avelumab plus Axitinib versus Sunitinib for advanced renal-cell carcinoma. N Engl J Med 2019; 380: 1103–15.
- 8Tang J, Shalabi A, Hubbard-Lucey VM. Comprehensive analysis of the clinical immuno-oncology landscape. Ann Oncol 2018; 29: 84–91.
- 9Hirsch L, Zitvogel L, Eggermont A, et al. PD-Loma: a cancer entity with a shared sensitivity to the PD-1/PD-L1 pathway blockade. Br J Cancer 2019; 120: 3–5.
- 10Marcus L, Lemery SJ, Keegan P, et al. FDA approval summary: Pembrolizumab for the treatment of microsatellite instability-high solid tumors. Clin Cancer Res 2019; 25: 3753–8.
- 11Havel JJ, Chowell D, Chan TA. The evolving landscape of biomarkers for checkpoint inhibitor immunotherapy. Nat Rev Cancer 2019; 19: 133–50.
- 12Nishino M, Ramaiya NH, Hatabu H, et al. Monitoring immune-checkpoint blockade: response evaluation and biomarker development. Nat Rev Clin Oncol 2017; 14: 655–68.
- 13Gajewski TF, Corrales L, Williams J, et al. Cancer immunotherapy targets based on understanding the T cell-inflamed versus non-T cell-inflamed tumor microenvironment. Adv Exp Med Biol 2017; 1036: 19–31.
- 14Galon J, Bruni D. Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat Rev Drug Discov 2019; 18: 197–218.
- 15Sharma P, Allison JP. The future of immune checkpoint therapy. Science 2015; 348: 56–61.
- 16Harlin H, Meng Y, Peterson AC, et al. Chemokine expression in melanoma metastases associated with CD8+ T-cell recruitment. Cancer Res 2009; 69: 3077–85.
- 17Taube JM, Anders RA, Young GD, et al. Colocalization of inflammatory response with b7-h1 expression in human melanocytic lesions supports an adaptive resistance mechanism of immune escape. Sci Transl Med 2012; 4:127ra137.
- 18Spranger S, Spaapen RM, Zha Y, et al. Up-regulation of PD-L1, IDO, and T(regs) in the melanoma tumor microenvironment is driven by CD8(+) T cells. Sci Transl Med 2013; 5:200ra116.
- 19Tumeh PC, Harview CL, Yearley JH, et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 2014; 515: 568–71.
- 20Hu-Lieskovan S, Lisberg A, Zaretsky JM, et al. Tumor characteristics associated with benefit from Pembrolizumab in advanced non-small cell lung cancer. Clin Cancer Res 2019; 25: 5061–8.
- 21Loi S, Adams S, Schmid P, et al. Relationship between tumor infiltrating lymphocyte (TIL) levels and response to pembrolizumab (pembro) in metastatic triple-negative breast cancer. Ann Oncol 2017; 28:LBA13.
- 22Wong PF, Wei W, Smithy JW, et al. Multiplex quantitative analysis of tumor-infiltrating lymphocytes and immunotherapy outcome in metastatic melanoma. Clin Cancer Res 2019; 25: 2442–9.
- 23Uryvaev A, Passhak M, Hershkovits D, et al. The role of tumor-infiltrating lymphocytes (TILs) as a predictive biomarker of response to anti-PD1 therapy in patients with metastatic non-small cell lung cancer or metastatic melanoma. Med Oncol 2018; 35: 25.
- 24Mami-Chouaib F, Blanc C, Corgnac S, et al. Resident memory T cells, critical components in tumor immunology. J Immunother Cancer 2018; 6: 87.
- 25Gide TN, Quek C, Menzies AM, et al. Distinct immune cell populations define response to anti-PD-1 Monotherapy and anti-PD-1/anti-CTLA-4 combined therapy. Cancer Cell 2019; 35: 238–255.e236.
- 26Herbst RS, Soria JC, Kowanetz M, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 2014; 515: 563–7.
- 27Ribas A, Robert C, Hodi S, et al. Association of response to programmed death receptor 1 (PD-1) blockade with pembrolizumab (MK-3475) with an interferon-inflammatory immune gene signature. J Clin Oncol 2015; 33: 3001.
- 28Rosenberg JE, Hoffman-Censits J, Powles T, et al. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial. Lancet 2016; 387: 1909–20.
- 29Seiwert T, Burtness B, Weiss J, et al. Inflamed-phenotype gene expression signatures to predict benefit from the anti-PD-1 antibody pembrolizumab in PD-L1+ head and neck cancer patients. J Clin Oncol 2015; 33: 6017.
- 30Ayers M, Lunceford J, Nebozhyn M, et al. IFN-gamma-related mRNA profile predicts clinical response to PD-1 blockade. J Clin Invest 2017; 127: 2930–40.
- 31Da Silva I, Menzies A, Newell F, et al. Comprehensive molecular profiling of metastatic melanoma to predict response to monotherapy and combination immunotherapy. J Clin Oncol 2019; 37: 9511.
- 32Fehrenbacher L, Spira A, Ballinger M, et al. Atezolizumab versus docetaxel for patients with previously treated non-small-cell lung cancer (POPLAR): a multicentre, open-label, phase 2 randomised controlled trial. Lancet 2016; 387: 1837–46.
- 33McDermott DF, Huseni MA, Atkins MB, et al. Clinical activity and molecular correlates of response to atezolizumab alone or in combination with bevacizumab versus sunitinib in renal cell carcinoma. Nat Med 2018; 24: 749–57.
- 34Damotte D, Arrondeau J, Boudou-Rouquette P, et al. The tumor inflammation signature is predictive of anti-PD1 treatment benefit in the CERTIM pan-cancer cohort. Cancer Res 2018; 78: 4546.
- 35Cristescu R, Mogg R, Ayers M, et al. Pan-tumor genomic biomarkers for PD-1 checkpoint blockade-based immunotherapy. Science 2018; 362:eaar3593.
- 36Ott PA, Bang YJ, Piha-Paul SA, et al. T-cell-inflamed gene-expression profile, programmed death ligand 1 expression, and tumor mutational burden predict efficacy in patients treated with Pembrolizumab across 20 cancers: KEYNOTE-028. J Clin Oncol 2019; 37: 318–27.
- 37Keenan TE, Burke KP, Van Allen EM. Genomic correlates of response to immune checkpoint blockade. Nat Med 2019; 25: 389–402.
- 38Samstein RM, Lee CH, Shoushtari AN, et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat Genet 2019; 51: 202–6.
- 39Yarchoan M, Hopkins A, Jaffee EM. Tumor mutational burden and response rate to PD-1 inhibition. N Engl J Med 2017; 377: 2500–1.
- 40Herbst R, Lopes G, Kowalski D, et al. Association between tissue TMB (tTMB) and clinical outcomes with pembrolizumab monotherapy (pembro) in PD-L1-positive advanced NSCLC in the KEYNOTE-010 and -042 trials. Ann Oncol 2019; 30(Suppl 5):LBA79.
- 41Paz-Ares L, Langer CJ, Novello S, et al. Pembrolizumab (pembro) plus platinum-based chemotherapy (chemo) for metastatic NSCLC: tissue TMB (tTMB) and outcomes in KEYNOTE-021, 189, and 407. Ann Oncol 2019; 30(Suppl 5): v851–934.
- 42Hegde PS, Karanikas V, Evers S. The where, the when, and the how of immune monitoring for cancer immunotherapies in the era of checkpoint inhibition. Clin Cancer Res 2016; 22: 1865–74.
- 43Kraman M, Bambrough PJ, Arnold JN, et al. Suppression of antitumor immunity by stromal cells expressing fibroblast activation protein-alpha. Science 2010; 330: 827–30.
- 44Alexandrov LB, Nik-Zainal S, Wedge DC, et al. Signatures of mutational processes in human cancer. Nature 2013; 500: 415–21.
- 45Bellone M, Calcinotto A. Ways to enhance lymphocyte trafficking into tumors and fitness of tumor infiltrating lymphocytes. Front Oncol 2013; 3: 231.
- 46Mariathasan S, Turley SJ, Nickles D, et al. TGFbeta attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature 2018; 554: 544–8.
- 47Tauriello DVF, Palomo-Ponce S, Stork D, et al. TGFbeta drives immune evasion in genetically reconstituted colon cancer metastasis. Nature 2018; 554: 538–43.
- 48Pascual-Garcia M, Bonfill-Teixidor E, Planas-Rigol E, et al. LIF regulates CXCL9 in tumor-associated macrophages and prevents CD8(+) T cell tumor-infiltration impairing anti-PD1 therapy. Nat Commun 2019; 10: 2416.
- 49Jerby-Arnon L, Shah P, Cuoco MS, et al. A cancer cell program promotes T cell exclusion and resistance to checkpoint blockade. Cell 2018; 175: 984–997.e924.
- 50Spranger S, Bao R, Gajewski TF. Melanoma-intrinsic beta-catenin signalling prevents anti-tumour immunity. Nature 2015; 523: 231–5.
- 51Peng W, Chen JQ, Liu C, et al. Loss of PTEN promotes resistance to T cell-mediated immunotherapy. Cancer Discov 2016; 6: 202–16.
- 52Koyama S, Akbay EA, Li YY, et al. STK11/LKB1 deficiency promotes neutrophil recruitment and Proinflammatory cytokine production to suppress T-cell activity in the lung tumor microenvironment. Cancer Res 2016; 76: 999–1008.
- 53Nghiem PT, Bhatia S, Lipson EJ, et al. PD-1 blockade with Pembrolizumab in advanced Merkel-cell carcinoma. N Engl J Med 2016; 374: 2542–52.
- 54Giraldo NA, Nguyen P, Engle EL, et al. Multidimensional, quantitative assessment of PD-1/PD-L1 expression in patients with Merkel cell carcinoma and association with response to pembrolizumab. J Immunother Cancer 2018; 6: 99.
- 55Miller BC, Sen DR, Al Abosy R, et al. Subsets of exhausted CD8(+) T cells differentially mediate tumor control and respond to checkpoint blockade. Nat Immunol 2019; 20: 326–36.
- 56Chen PL, Roh W, Reuben A, 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: 827–37.
- 57Hugo W, Zaretsky JM, Sun L, et al. Genomic and Transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma. Cell 2016; 165: 35–44.
- 58Ascierto ML, McMiller TL, Berger AE, et al. The Intratumoral balance between metabolic and immunologic gene expression is associated with anti-PD-1 response in patients with renal cell carcinoma. Cancer Immunol Res 2016; 4: 726–33.
- 59McDermott DF, Sosman JA, Sznol M, et al. Atezolizumab, an anti-programmed death-ligand 1 antibody, in metastatic renal cell carcinoma: Long-term safety, clinical activity, and immune correlates from a phase Ia study. J Clin Oncol 2016; 34: 833–42.
- 60Yost KE, Satpathy AT, Wells DK, et al. Clonal replacement of tumor-specific T cells following PD-1 blockade. Nat Med 2019; 25: 1251–9.
- 61Pauken KE, Sammons MA, Odorizzi PM, et al. Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade. Science 2016; 354: 1160–5.
- 62Scott AC, Dundar F, Zumbo P, et al. TOX is a critical regulator of tumour-specific T cell differentiation. Nature 2019; 571: 270–4.
- 63Campbell M, Yau C, Bolen J, et al. Analysis of immune cell infiltrates as predictors of response to the checkpoint inhibitor pembrolizumab in the neoadjuvant I-SPY 2 TRIAL. Cancer Res 2019; 79(13 suppl):CT003.
- 64Salmon H, Franciszkiewicz K, Damotte D, et al. Matrix architecture defines the preferential localization and migration of T cells into the stroma of human lung tumors. J Clin Invest 2012; 122: 899–910.
- 65Thommen DS, Schreiner J, Muller P, et al. Progression of lung cancer is associated with increased dysfunction of T cells defined by Coexpression of multiple inhibitory receptors. Cancer Immunol Res 2015; 3: 1344–55.
- 66Anderson AC, Joller N, Kuchroo VK. Lag-3, Tim-3, and TIGIT: co-inhibitory receptors with specialized functions in immune regulation. Immunity 2016; 44: 989–1004.
- 67Thommen DS, Koelzer VH, Herzig P, et al. A transcriptionally and functionally distinct PD-1(+) CD8(+) T cell pool with predictive potential in non-small-cell lung cancer treated with PD-1 blockade. Nat Med 2018; 24: 994–1004.
- 68Horton BL, Williams JB, Cabanov A, et al. Intratumoral CD8(+) T-cell apoptosis is a major component of T-cell dysfunction and impedes antitumor immunity. Cancer Immunol Res 2018; 6: 14–24.
- 69Datar I, Sanmamed MF, Wang J, et al. Expression analysis and significance of PD-1, LAG-3, and TIM-3 in human non-small cell lung cancer using spatially resolved and multiparametric single-cell analysis. Clin Cancer Res 2019; 25: 4663–73.
- 70Shayan G, Srivastava R, Li J, et al. Adaptive resistance to anti-PD1 therapy by Tim-3 upregulation is mediated by the PI3K-Akt pathway in head and neck cancer. Oncoimmunology 2017; 6:e1261779.
- 71Koyama S, Akbay EA, Li YY, et al. Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints. Nat Commun 2016; 7:10501.
- 72Zelba H, Bedke J, Hennenlotter J, et al. PD-1 and LAG-3 dominate checkpoint receptor-mediated T-cell inhibition in renal cell carcinoma. Cancer Immunol Res 2019; 7: 1891–9.
- 73Oweida A, Hararah MK, Phan A, et al. Resistance to radiotherapy and PD-L1 blockade is mediated by TIM-3 Upregulation and regulatory T-cell infiltration. Clin Cancer Res 2018; 24: 5368–80.
- 74Bertrand F, Montfort A, Marcheteau E, et al. TNFalpha blockade overcomes resistance to anti-PD-1 in experimental melanoma. Nat Commun 2017; 8: 2256.
- 75Wierz M, Pierson S, Guyonnet L, et al. Dual PD1/LAG3 immune checkpoint blockade limits tumor development in a murine model of chronic lymphocytic leukemia. Blood 2018; 131: 1617–21.
- 76Chauvin JM, Pagliano O, Fourcade J, et al. TIGIT and PD-1 impair tumor antigen-specific CD8(+) T cells in melanoma patients. J Clin Invest 2015; 125: 2046–58.
- 77Johnston RJ, Comps-Agrar L, Hackney J, et al. The immunoreceptor TIGIT regulates antitumor and antiviral CD8(+) T cell effector function. Cancer Cell 2014; 26: 923–37.
- 78Dixon KO, Schorer M, Nevin J, et al. Functional anti-TIGIT antibodies regulate development of autoimmunity and antitumor immunity. J Immunol 2018; 200: 3000–7.
- 79Fourcade J, Sun Z, Benallaoua M, 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: 2175–86.
- 80Sakuishi K, Apetoh L, Sullivan JM, et al. Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity. J Exp Med 2010; 207: 2187–94.
- 81Woo SR, Turnis ME, Goldberg MV, et al. Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape. Cancer Res 2012; 72: 917–27.
- 82Granier C, Dariane C, Combe P, et al. Tim-3 expression on tumor-infiltrating PD-1+CD8+ T cells correlates with poor clinical outcome in renal cell carcinoma. Cancer Res 2017; 77: 1075–82.
- 83Granier C, De Guillebon E, Blanc C, et al. Mechanisms of action and rationale for the use of checkpoint inhibitors in cancer. ESMO open 2017; 2:e000213.
- 84Fucikova J, Rakova J, Hensler M, et al. TIM-3 dictates functional orientation of the immune infiltrate in ovarian cancer. Clin Cancer Res 2019; 25: 4820–31.
- 85Hanna GJ, Lizotte P, Cavanaugh M, et al. Frameshift events predict anti-PD-1/L1 response in head and neck cancer. JCI Insight 2018; 3:e98811.
- 86Pignon JC, Jegede O, Shukla SA, et al. irRECIST for the evaluation of candidate biomarkers of response to Nivolumab in metastatic clear cell renal cell carcinoma: analysis of a phase II prospective clinical trial. Clin Cancer Res 2019; 25: 2174–84.
- 87Auslander N, Zhang G, Lee JS, et al. Robust prediction of response to immune checkpoint blockade therapy in metastatic melanoma. Nat Med 2018; 24: 1545–9.
- 88Daud AI, Loo K, Pauli ML, et al. Tumor immune profiling predicts response to anti-PD-1 therapy in human melanoma. J Clin Invest 2016; 126: 3447–52.
- 89Huang AC, Postow MA, Orlowski RJ, et al. T-cell invigoration to tumour burden ratio associated with anti-PD-1 response. Nature 2017; 545: 60–5.
- 90Duhen 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.
- 91Simoni 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.
- 92Ganesan AP, Clarke J, Wood O, et al. Tissue-resident memory features are linked to the magnitude of cytotoxic T cell responses in human lung cancer. Nat Immunol 2017; 18: 940–50.
- 93Nizard M, Roussel H, Diniz MO, et al. Induction of resident memory T cells enhances the efficacy of cancer vaccine. Nat Commun 2017; 8:15221.
- 94Granier C, Blanc C, Karaki S, et al. Tissue-resident memory T cells play a key role in the efficacy of cancer vaccines. Oncoimmunology 2017; 6:e1358841.
- 95Savas P, Virassamy B, Ye C, et al. Single-cell profiling of breast cancer T cells reveals a tissue-resident memory subset associated with improved prognosis. Nat Med 2018; 24: 986–93.
- 96Mackay LK, Rahimpour A, Ma JZ, et al. The developmental pathway for CD103(+)CD8+ tissue-resident memory T cells of skin. Nat Immunol 2013; 14: 1294–301.
- 97Kumar BV, Ma W, Miron M, et al. Human tissue-resident memory T cells are defined by Core transcriptional and functional signatures in lymphoid and mucosal sites. Cell Rep 2017; 20: 2921–34.
- 98Hombrink P, Helbig C, Backer RA, et al. Programs for the persistence, vigilance and control of human CD8+ lung-resident memory T cells. Nat Immunol 2016; 17: 1467–78.
- 99Boddupalli CS, Bar N, Kadaveru K, et al. Interlesional diversity of T cell receptors in melanoma with immune checkpoints enriched in tissue-resident memory T cells. JCI Insight 2016; 1:e88955.
- 100Wang P, Huang B, Gao Y, et al. CD103(+)CD8(+) T lymphocytes in non-small cell lung cancer are phenotypically and functionally primed to respond to PD-1 blockade. Cell Immunol 2018; 325: 48–55.
- 101Clarke J, Panwar B, Madrigal A, et al. Single-cell transcriptomic analysis of tissue-resident memory T cells in human lung cancer. J Exp Med 2019; 216: 2128–49.
- 102Pallett LJ, Davies J, Colbeck EJ, et al. IL-2(high) tissue-resident T cells in the human liver: sentinels for hepatotropic infection. J Exp Med 2017; 214: 1567–80.
- 103Blanc C, Hans S, Tran T, et al. Targeting resident memory T cells for cancer immunotherapy. Front Immunol 2018; 9: 1722.
- 104Edwards J, Wilmott JS, Madore J, et al. Palendira U.CD103(+) tumor-resident CD8(+) T cells are associated with improved survival in immunotherapy-naive melanoma patients and expand significantly during anti-PD-1 treatment. Clin Cancer Res 2018; 24: 3036–45.
- 105Wei SC, Levine JH, Cogdill AP, et al. Distinct cellular mechanisms underlie anti-CTLA-4 and anti-PD-1 checkpoint blockade. Cell 2017; 170: 1120–1133.e1117.
- 106Shields BD, Koss B, Taylor EM, et al. Loss of E-cadherin inhibits CD103 antitumor activity and reduces checkpoint blockade responsiveness in melanoma. Cancer Res 2019; 79: 1113–23.
- 107Kurtulus S, Madi A, Escobar G, et al. Checkpoint blockade immunotherapy induces dynamic changes in PD-1(−)CD8(+) tumor-infiltrating T cells. Immunity 2019; 50: 181–194.e186.
- 108Siddiqui I, Schaeuble K, Chennupati V, et al. Intratumoral Tcf1(+)PD-1(+)CD8(+) T cells with stem-like properties promote tumor control in response to vaccination and checkpoint blockade immunotherapy. Immunity 2019; 50: 195–211.e110.
- 109Thommen DS. The first shall (be) last: understanding durable T cell responses in immunotherapy. Immunity 2019; 50: 6–8.
- 110Chouaib S, Noman MZ, Kosmatopoulos K, et al. Hypoxic stress: obstacles and opportunities for innovative immunotherapy of cancer. Oncogene 2017; 36: 439–45.
- 111Tartour E, Pere H, Maillere B, et al. Angiogenesis and immunity: a bidirectional link potentially relevant for the monitoring of antiangiogenic therapy and the development of novel therapeutic combination with immunotherapy. Cancer Metastasis Rev 2011; 30: 83–95.
- 112Ben-Shoshan J, Maysel-Auslender S, Mor A, et al. Hypoxia controls CD4+CD25+ regulatory T-cell homeostasis via hypoxia-inducible factor-1alpha. Eur J Immunol 2008; 38: 2412–8.
- 113Facciabene A, Peng X, Hagemann IS, et al. Tumour hypoxia promotes tolerance and angiogenesis via CCL28 and T(reg) cells. Nature 2011; 475: 226–30.
- 114Noman MZ, Desantis G, Janji B, et al. PD-L1 is a novel direct target of HIF-1alpha, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J Exp Med 2014; 211: 781–90.
- 115Jayaprakash P, Ai M, Liu A, et al. Targeted hypoxia reduction restores T cell infiltration and sensitizes prostate cancer to immunotherapy. J Clin Invest 2018; 128: 5137–49.
- 116Peranzoni E, Lemoine J, Vimeux L, et al. Macrophages impede CD8 T cells from reaching tumor cells and limit the efficacy of anti-PD-1 treatment. Proc Natl Acad Sci USA 2018; 115: E4041–50.
- 117Rodriguez PC, Quiceno DG, Zabaleta J, et al. Arginase I production in the tumor microenvironment by mature myeloid cells inhibits T-cell receptor expression and antigen-specific T-cell responses. Cancer Res 2004; 64: 5839–49.
- 118Munn DH, Mellor AL. Indoleamine 2,3-dioxygenase and tumor-induced tolerance. J Clin Invest 2007; 117: 1147–54.
- 119Lin C, He H, Liu H, et al. Tumour-associated macrophages-derived CXCL8 determines immune evasion through autonomous PD-L1 expression in gastric cancer. Gut 2019; 68: 1764–73.
- 120Li J, Lee Y, Li Y, et al. Co-inhibitory molecule B7 superfamily member 1 expressed by tumor-infiltrating myeloid cells induces dysfunction of anti-tumor CD8(+) T cells. Immunity 2018; 48: 773–786.e775.
- 121Viitala M, Virtakoivu R, Tadayon S, et al. Immunotherapeutic blockade of macrophage Clever-1 reactivates the CD8(+) T-cell response against immunosuppressive tumors. Clin Cancer Res 2019; 25: 3289–303.
- 122Mantovani A, Bonecchi R. One clever macrophage checkpoint. Clin Cancer Res 2019; 25: 3202–4.
- 123Molon B, Ugel S, Del Pozzo F, et al. Chemokine nitration prevents intratumoral infiltration of antigen-specific T cells. J Exp Med 2011; 208: 1949–62.
- 124Pere H, Tanchot C, Bayry J, et al. Comprehensive analysis of current approaches to inhibit regulatory T cells in cancer. Oncoimmunology 2012; 1: 326–33.
- 125Li L, Wang L, Li J, et al. Metformin-induced reduction of CD39 and CD73 blocks myeloid-derived suppressor cell activity in patients with ovarian cancer. Cancer Res 2018; 78: 1779–91.
- 126Al-Khami AA, Zheng L, Del Valle L, et al. Exogenous lipid uptake induces metabolic and functional reprogramming of tumor-associated myeloid-derived suppressor cells. Oncoimmunology 2017; 6:e1344804.
- 127Fu Q, Xu L, Wang Y, et al. Tumor-associated macrophage-derived Interleukin-23 interlinks kidney cancer glutamine addiction with immune evasion. Eur Urol 2019; 75: 752–63.
- 128Gao J, Shi LZ, Zhao H, et al. Loss of IFN-gamma pathway genes in tumor cells as a mechanism of resistance to anti-CTLA-4 therapy. Cell 2016; 167: 397–404.e399.
- 129Miao D, Margolis CA, Vokes NI, et al. Genomic correlates of response to immune checkpoint blockade in microsatellite-stable solid tumors. Nat Genet 2018; 50: 1271–81.
- 130Zaretsky JM, Garcia-Diaz A, Shin DS, et al. Mutations associated with acquired resistance to PD-1 blockade in melanoma. N Engl J Med 2016; 375: 819–29.
- 131Jiang P, Gu S, Pan D, et al. Signatures of T cell dysfunction and exclusion predict cancer immunotherapy response. Nat Med 2018; 24: 1550–8.
- 132Luke JJ, Bao R, Sweis RF, et al. WNT/beta-catenin pathway activation correlates with immune exclusion across human cancers. Clin Cancer Res 2019; 25: 3074–83.
- 133Liu J, Jiang W, Zhao K, et al. Tumoral EHF predicts the efficacy of anti-PD1 therapy in pancreatic ductal adenocarcinoma. J Exp Med 2019; 216: 656–73.
- 134Wang J, Yu F, Jia X, et al. MicroRNA-155 deficiency enhances the recruitment and functions of myeloid-derived suppressor cells in tumor microenvironment and promotes solid tumor growth. Int J Cancer 2015; 136: E602–13.
- 135Hahne M, Rimoldi D, Schroter M, et al. Melanoma cell expression of Fas(Apo-1/CD95) ligand: implications for tumor immune escape. Science 1996; 274: 1363–6.
- 136Azuma T, Yao S, Zhu G, et al. B7-H1 is a ubiquitous antiapoptotic receptor on cancer cells. Blood 2008; 111: 3635–43.
- 137Loumagne L, Baudhuin J, Favier B, et al. In vivo evidence that secretion of HLA-G by immunogenic tumor cells allows their evasion from immunosurveillance. Int J Cancer 2014; 135: 2107–17.
- 138Jamal-Hanjani M, Wilson GA, McGranahan N, et al. Tracking the evolution of non-small-cell lung cancer. N Engl J Med 2017; 376: 2109–21.
- 139Wolf Y, Bartok O, Patkar S, et al. UVB-induced tumor heterogeneity diminishes immune response in melanoma. Cell 2019; 179: 219–235.e221.
- 140Galon J, Costes A, Sanchez-Cabo F, et al. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science 2006; 313: 1960–4.
- 141Dhodapkar KM. Role of tissue-resident memory in intra-tumor heterogeneity and response to immune checkpoint blockade. Front Immunol 2018; 9: 1655.
- 142Rashidian M, Ingram JR, Dougan M, et al. Predicting the response to CTLA-4 blockade by longitudinal noninvasive monitoring of CD8 T cells. J Exp Med 2017; 214: 2243–55.
- 143Dumauthioz N, Labiano S, Romero P. Tumor resident memory T cells: new players in immune surveillance and therapy. Front Immunol 2018; 9: 2076.
- 144Molodtsov A, Turk MJ. Tissue resident CD8 memory T cell responses in cancer and autoimmunity. Front Immunol 2018; 9: 2810.
- 145McDermott D, Huseni M, Rini B, et al. Molecular correlates of differential response to Atezolizumab +/− Bevacizumab vs Sunitnib in a Phase II study in untreated metastatic renal cell carcinoma (RCC) patients. Cancer res 2017; 77:CT081.
- 146Kim HR, Park SM, Seo SU, et al. The ratio of peripheral regulatory T cells to Lox-1(+) Polymorphonuclear myeloid-derived suppressor cells predicts the early response to anti-PD-1 therapy in patients with non-small cell lung cancer. Am J Respir Crit Care Med 2019; 199: 243–6.
- 147Papadopoulos K, Lakhari N, Johnson M, et al. First-in-human study of REGN3767 (R3767), a human LAG-3 monoclonal antibody (mAb), ± cemiplimab in patients (pts) with advanced malignancies. J Clin Oncol 2019; 37(Suppl 15): 2508.
- 148Mach N, Curigliano G, Santoro A, et al. Phase (Ph) II study of MBG453 + spartalizumab in patients (pts) with non-small cell lung cancer (NSCLC) and melanoma pretreated with anti-PD-1/L1 therapy. Ann Oncol 2019; 30(Suppl 5): 1202.
- 149Davar D, Boasberg P, Eroglu Z, et al. A phase 1 study of TSR-022, an anti-TIM-3 monoclonal antibody, in combination with TSR-042 (anti-PD-1) in patients with colorectal cancer and post-PD-1 NSCLC and melanoma. J Immunother Cancer 2018; 6(Suppl 1):021.
- 150Ascierto P, Melero I, Bhatia S, et al. Initial efficacy of anti-lymphocyte activation gene-3 (anti–LAG-3; BMS-986016) in combination with nivolumab (nivo) in pts with melanoma (MEL) previously treated with anti–PD-1/PD-L1 therapy. J Clin Oncol 2017; 35(Suppl 15): 9520.
- 151Yin T, Zhao ZB, Guo J, et al. Inhibition eliminates myeloid cell-mediated immunosuppression and enhances the efficacy of anti-PD-L1 therapy in breast cancer. Cancer Res 2019; 79: 3431–44.
- 152Wei Y, Zhao Q, Gao Z, et al. The local immune landscape determines tumor PD-L1 heterogeneity and sensitivity to therapy. J Clin Invest 2019; 129: 3347–60.
- 153Quaranta V, Schmid MC. Macrophage-mediated subversion of anti-tumour immunity. Cell 2019; 8:E747.
- 154Clavijo PE, Friedman J, Robbins Y, et al. Semaphorin4D inhibition improves response to immune-checkpoint blockade via attenuation of MDSC recruitment and function. Cancer Immunol Res 2019; 7: 282–91.
- 155Vigano S, Alatzoglou D, Irving M, et al. Targeting adenosine in cancer immunotherapy to enhance T-cell function. Front Immunol 2019; 10: 925.
- 156Gruosso T, O'ccnor M, Denis J, et al. AVID200 neutralizes TGF-beta1 and -beta3, the principal immunosuppressive TGF-beta isoforms overexpressed by tumors, and sensitizes tumors to immune-checkpoint inhibitors. Ann Oncol 2019; 30(suppl 5): 504.
- 157Etzerodt A, Tsalkitzi K, Maniecki M, et al. Specific targeting of CD163(+) TAMs mobilizes inflammatory monocytes and promotes T cell-mediated tumor regression. J Exp Med 2019; 216: 2394–411.
- 158Laplane L, Duluc D, Bikfalvi A, et al. Beyond the tumour microenvironment. Int J Cancer 2019; 145: 2611–8.