Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2021

01-12-2021 | Melanoma | Review

Emerging dynamics pathways of response and resistance to PD-1 and CTLA-4 blockade: tackling uncertainty by confronting complexity

Authors: Allan Relecom, Maysaloun Merhi, Varghese Inchakalody, Shahab Uddin, Darawan Rinchai, Davide Bedognetti, Said Dermime

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2021

Login to get access

Abstract

Immune checkpoint inhibitors provide considerable therapeutic benefit in a range of solid cancers as well as in a subgroup of hematological malignancies. Response rates are however suboptimal, and despite considerable efforts, predicting response to immune checkpoint inhibitors ahead of their administration in a given patient remains elusive. The study of the dynamics of the immune system and of the tumor under immune checkpoint blockade brought insight into the mechanisms of action of these therapeutic agents. Equally relevant are the mechanisms of adaptive resistance to immune checkpoint inhibitors that have been uncovered through this approach. In this review, we discuss the dynamics of the immune system and of the tumor under immune checkpoint blockade emanating from recent studies on animal models and humans. We will focus on mechanisms of action and of resistance conveying information predictive of therapeutic response.
Literature
1.
go back to reference Larkin J, et al. Five-Year Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N Engl J Med. 2019;381:1535–46.PubMedCrossRef Larkin J, et al. Five-Year Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N Engl J Med. 2019;381:1535–46.PubMedCrossRef
2.
go back to reference Maio M, et al. Five-year survival rates for treatment-naive patients with advanced melanoma who received ipilimumab plus dacarbazine in a phase III trial. J Clin Oncol. 2015;33:1191–6.PubMedPubMedCentralCrossRef Maio M, et al. Five-year survival rates for treatment-naive patients with advanced melanoma who received ipilimumab plus dacarbazine in a phase III trial. J Clin Oncol. 2015;33:1191–6.PubMedPubMedCentralCrossRef
4.
go back to reference Robert C, et al. Pembrolizumab versus ipilimumab in advanced melanoma (KEYNOTE-006): post-hoc 5-year results from an open-label, multicentre, randomised, controlled, phase 3 study. Lancet Oncol. 2019;20:1239–51.PubMedCrossRef Robert C, et al. Pembrolizumab versus ipilimumab in advanced melanoma (KEYNOTE-006): post-hoc 5-year results from an open-label, multicentre, randomised, controlled, phase 3 study. Lancet Oncol. 2019;20:1239–51.PubMedCrossRef
7.
go back to reference K Choucair et al., TMB: a promising immune-response biomarker, and potential spearhead in advancing targeted therapy trials. Cancer Gene Ther, (2020). K Choucair et al., TMB: a promising immune-response biomarker, and potential spearhead in advancing targeted therapy trials. Cancer Gene Ther, (2020).
8.
go back to reference D Chowell et al. Patient HLA class I genotype influences cancer response to checkpoint blockade immunotherapy. Science. 2018;359:582–7.CrossRefPubMed D Chowell et al. Patient HLA class I genotype influences cancer response to checkpoint blockade immunotherapy. Science. 2018;359:582–7.CrossRefPubMed
9.
go back to reference Shaikh FY, Gills JJ, Sears CL. Impact of the microbiome on checkpoint inhibitor treatment in patients with non-small cell lung cancer and melanoma. EBioMedicine. 2019;48:642–7.PubMedPubMedCentralCrossRef Shaikh FY, Gills JJ, Sears CL. Impact of the microbiome on checkpoint inhibitor treatment in patients with non-small cell lung cancer and melanoma. EBioMedicine. 2019;48:642–7.PubMedPubMedCentralCrossRef
14.
go back to reference Hodgkin PD. A probabilistic view of immunology: drawing parallels with physics. Immunol Cell Biol. 2007;85:295–9.PubMedCrossRef Hodgkin PD. A probabilistic view of immunology: drawing parallels with physics. Immunol Cell Biol. 2007;85:295–9.PubMedCrossRef
15.
go back to reference D Duneau et al., Stochastic variation in the initial phase of bacterial infection predicts the probability of survival in D. melanogaster. Elife 6, (2017). D Duneau et al., Stochastic variation in the initial phase of bacterial infection predicts the probability of survival in D. melanogaster. Elife 6, (2017).
16.
go back to reference WJ Lesterhuis et al. Dynamic versus static biomarkers in cancer immune checkpoint blockade: unravelling complexity. Nat Rev Drug Discov. 2017;16:264–72.PubMedCrossRef WJ Lesterhuis et al. Dynamic versus static biomarkers in cancer immune checkpoint blockade: unravelling complexity. Nat Rev Drug Discov. 2017;16:264–72.PubMedCrossRef
17.
go back to reference TL Walunas et al. CTLA-4 can function as a negative regulator of T cell activation. Immunity. 1994;1:405–13.CrossRefPubMed TL Walunas et al. CTLA-4 can function as a negative regulator of T cell activation. Immunity. 1994;1:405–13.CrossRefPubMed
18.
go back to reference P Waterhouse et al. Lymphoproliferative disorders with early lethality in mice deficient in Ctla-4. Science. 1995;270:985–8.PubMedCrossRef P Waterhouse et al. Lymphoproliferative disorders with early lethality in mice deficient in Ctla-4. Science. 1995;270:985–8.PubMedCrossRef
21.
go back to reference Fehlings M, et al. Checkpoint blockade immunotherapy reshapes the high-dimensional phenotypic heterogeneity of murine intratumoural neoantigen-specific CD8(+) T cells. Nat Commun. 2017;8:562.PubMedPubMedCentralCrossRef Fehlings M, et al. Checkpoint blockade immunotherapy reshapes the high-dimensional phenotypic heterogeneity of murine intratumoural neoantigen-specific CD8(+) T cells. Nat Commun. 2017;8:562.PubMedPubMedCentralCrossRef
22.
23.
go back to reference Klein O, et al. Melan-A-specific cytotoxic T cells are associated with tumor regression and autoimmunity following treatment with anti-CTLA-4. Clin Cancer Res. 2009;15:2507–13.PubMedCrossRef Klein O, et al. Melan-A-specific cytotoxic T cells are associated with tumor regression and autoimmunity following treatment with anti-CTLA-4. Clin Cancer Res. 2009;15:2507–13.PubMedCrossRef
24.
go back to reference PL Chen 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.PubMedPubMedCentralCrossRef PL Chen 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.PubMedPubMedCentralCrossRef
25.
go back to reference Ribas A, et al. Intratumoral immune cell infiltrates, FoxP3, and indoleamine 2,3-dioxygenase in patients with melanoma undergoing CTLA4 blockade. Clin Cancer Res. 2009;15:390–9.PubMedCrossRef Ribas A, et al. Intratumoral immune cell infiltrates, FoxP3, and indoleamine 2,3-dioxygenase in patients with melanoma undergoing CTLA4 blockade. Clin Cancer Res. 2009;15:390–9.PubMedCrossRef
26.
go back to reference RR Ji et al. An immune-active tumor microenvironment favors clinical response to ipilimumab. Cancer Immunol Immunother. 2012;61:1019–31.PubMedCrossRef RR Ji et al. An immune-active tumor microenvironment favors clinical response to ipilimumab. Cancer Immunol Immunother. 2012;61:1019–31.PubMedCrossRef
27.
go back to reference Pedicord VA, Montalvo W, Leiner IM, Allison JP. Single dose of anti-CTLA-4 enhances CD8 + T-cell memory formation, function, and maintenance. Proc Natl Acad Sci U S A. 2011;108:266–71.PubMedCrossRef Pedicord VA, Montalvo W, Leiner IM, Allison JP. Single dose of anti-CTLA-4 enhances CD8 + T-cell memory formation, function, and maintenance. Proc Natl Acad Sci U S A. 2011;108:266–71.PubMedCrossRef
29.
go back to reference de Coana YP, et al. Ipilimumab treatment decreases monocytic MDSCs and increases CD8 effector memory T cells in long-term survivors with advanced melanoma. Oncotarget. 2017;8:21539–53.PubMedPubMedCentralCrossRef de Coana YP, et al. Ipilimumab treatment decreases monocytic MDSCs and increases CD8 effector memory T cells in long-term survivors with advanced melanoma. Oncotarget. 2017;8:21539–53.PubMedPubMedCentralCrossRef
30.
go back to reference RR Huang et al. CTLA4 blockade induces frequent tumor infiltration by activated lymphocytes regardless of clinical responses in humans. Clin Cancer Res. 2011;17:4101–9.PubMedPubMedCentralCrossRef RR Huang et al. CTLA4 blockade induces frequent tumor infiltration by activated lymphocytes regardless of clinical responses in humans. Clin Cancer Res. 2011;17:4101–9.PubMedPubMedCentralCrossRef
33.
go back to reference Hosoi A, et al. Increased diversity with reduced “diversity evenness” of tumor infiltrating T-cells for the successful cancer immunotherapy. Sci Rep. 2018;8:1058.PubMedPubMedCentralCrossRef Hosoi A, et al. Increased diversity with reduced “diversity evenness” of tumor infiltrating T-cells for the successful cancer immunotherapy. Sci Rep. 2018;8:1058.PubMedPubMedCentralCrossRef
34.
go back to reference Fu T, He Q, Sharma P. The ICOS/ICOSL pathway is required for optimal antitumor responses mediated by anti-CTLA-4 therapy. Cancer Res. 2011;71:5445–54.PubMedCrossRef Fu T, He Q, Sharma P. The ICOS/ICOSL pathway is required for optimal antitumor responses mediated by anti-CTLA-4 therapy. Cancer Res. 2011;71:5445–54.PubMedCrossRef
35.
go back to reference Jiao S, et al. Differences in Tumor Microenvironment Dictate T Helper Lineage Polarization and Response to Immune Checkpoint Therapy. Cell. 2019;179:1177–90 e1113.PubMedCrossRef Jiao S, et al. Differences in Tumor Microenvironment Dictate T Helper Lineage Polarization and Response to Immune Checkpoint Therapy. Cell. 2019;179:1177–90 e1113.PubMedCrossRef
36.
go back to reference E von Euw et al. CTLA4 blockade increases Th17 cells in patients with metastatic melanoma. J Transl Med. 2009;7:35.CrossRef E von Euw et al. CTLA4 blockade increases Th17 cells in patients with metastatic melanoma. J Transl Med. 2009;7:35.CrossRef
37.
go back to reference Yasuda K, Takeuchi Y, Hirota K. The pathogenicity of Th17 cells in autoimmune diseases. Semin Immunopathol. 2019;41:283–97.PubMedCrossRef Yasuda K, Takeuchi Y, Hirota K. The pathogenicity of Th17 cells in autoimmune diseases. Semin Immunopathol. 2019;41:283–97.PubMedCrossRef
38.
go back to reference Chen H, et al. Anti-CTLA-4 therapy results in higher CD4 + ICOShi T cell frequency and IFN-gamma levels in both nonmalignant and malignant prostate tissues. Proc Natl Acad Sci U S A. 2009;106:2729–34.PubMedPubMedCentralCrossRef Chen H, et al. Anti-CTLA-4 therapy results in higher CD4 + ICOShi T cell frequency and IFN-gamma levels in both nonmalignant and malignant prostate tissues. Proc Natl Acad Sci U S A. 2009;106:2729–34.PubMedPubMedCentralCrossRef
39.
go back to reference CI Liakou et al. CTLA-4 blockade increases IFNgamma-producing CD4 + ICOShi cells to shift the ratio of effector to regulatory T cells in cancer patients. Proc Natl Acad Sci U S A. 2008;105:14987–92.PubMedPubMedCentralCrossRef CI Liakou et al. CTLA-4 blockade increases IFNgamma-producing CD4 + ICOShi cells to shift the ratio of effector to regulatory T cells in cancer patients. Proc Natl Acad Sci U S A. 2008;105:14987–92.PubMedPubMedCentralCrossRef
40.
go back to reference BC Carthon et al. Preoperative CTLA-4 blockade: tolerability and immune monitoring in the setting of a presurgical clinical trial. Clin Cancer Res. 2010;16:2861–71.PubMedPubMedCentralCrossRef BC Carthon et al. Preoperative CTLA-4 blockade: tolerability and immune monitoring in the setting of a presurgical clinical trial. Clin Cancer Res. 2010;16:2861–71.PubMedPubMedCentralCrossRef
41.
go back to reference Ng Tang D, et al. Increased frequency of ICOS + CD4 T cells as a pharmacodynamic biomarker for anti-CTLA-4 therapy. Cancer Immunol Res. 2013;1:229–34.PubMedCrossRef Ng Tang D, et al. Increased frequency of ICOS + CD4 T cells as a pharmacodynamic biomarker for anti-CTLA-4 therapy. Cancer Immunol Res. 2013;1:229–34.PubMedCrossRef
42.
go back to reference JS Weber et al. Ipilimumab increases activated T cells and enhances humoral immunity in patients with advanced melanoma. J Immunother. 2012;35:89–97.PubMedCrossRef JS Weber et al. Ipilimumab increases activated T cells and enhances humoral immunity in patients with advanced melanoma. J Immunother. 2012;35:89–97.PubMedCrossRef
43.
go back to reference Kitano S, et al. Enhancement of tumor-reactive cytotoxic CD4 + T cell responses after ipilimumab treatment in four advanced melanoma patients. Cancer Immunol Res. 2013;1:235–44.PubMedCrossRef Kitano S, et al. Enhancement of tumor-reactive cytotoxic CD4 + T cell responses after ipilimumab treatment in four advanced melanoma patients. Cancer Immunol Res. 2013;1:235–44.PubMedCrossRef
44.
go back to reference RH Vonderheide et al. Tremelimumab in combination with exemestane in patients with advanced breast cancer and treatment-associated modulation of inducible costimulator expression on patient T cells. Clin Cancer Res. 2010;16:3485–94.PubMedCrossRef RH Vonderheide et al. Tremelimumab in combination with exemestane in patients with advanced breast cancer and treatment-associated modulation of inducible costimulator expression on patient T cells. Clin Cancer Res. 2010;16:3485–94.PubMedCrossRef
45.
go back to reference R Das et al. Combination therapy with anti-CTLA-4 and anti-PD-1 leads to distinct immunologic changes in vivo. J Immunol. 2015;194:950–9.PubMedCrossRef R Das et al. Combination therapy with anti-CTLA-4 and anti-PD-1 leads to distinct immunologic changes in vivo. J Immunol. 2015;194:950–9.PubMedCrossRef
46.
go back to reference B Kavanagh et al. CTLA4 blockade expands FoxP3 + regulatory and activated effector CD4 + T cells in a dose-dependent fashion. Blood. 2008;112:1175–83.PubMedPubMedCentralCrossRef B Kavanagh et al. CTLA4 blockade expands FoxP3 + regulatory and activated effector CD4 + T cells in a dose-dependent fashion. Blood. 2008;112:1175–83.PubMedPubMedCentralCrossRef
48.
go back to reference Pico de Coana Y, et al. Ipilimumab treatment results in an early decrease in the frequency of circulating granulocytic myeloid-derived suppressor cells as well as their Arginase1 production. Cancer Immunol Res. 2013;1:158–62.PubMedCrossRef Pico de Coana Y, et al. Ipilimumab treatment results in an early decrease in the frequency of circulating granulocytic myeloid-derived suppressor cells as well as their Arginase1 production. Cancer Immunol Res. 2013;1:158–62.PubMedCrossRef
49.
go back to reference GQ Phan et al. Cancer regression and autoimmunity induced by cytotoxic T lymphocyte-associated antigen 4 blockade in patients with metastatic melanoma. Proc Natl Acad Sci U S A. 2003;100:8372–7.PubMedPubMedCentralCrossRef GQ Phan et al. Cancer regression and autoimmunity induced by cytotoxic T lymphocyte-associated antigen 4 blockade in patients with metastatic melanoma. Proc Natl Acad Sci U S A. 2003;100:8372–7.PubMedPubMedCentralCrossRef
50.
go back to reference Wistuba-Hamprecht K, et al. Peripheral CD8 effector-memory type 1 T-cells correlate with outcome in ipilimumab-treated stage IV melanoma patients. Eur J Cancer. 2017;73:61–70.PubMedPubMedCentralCrossRef Wistuba-Hamprecht K, et al. Peripheral CD8 effector-memory type 1 T-cells correlate with outcome in ipilimumab-treated stage IV melanoma patients. Eur J Cancer. 2017;73:61–70.PubMedPubMedCentralCrossRef
51.
go back to reference J Yuan et al. CTLA-4 blockade enhances polyfunctional NY-ESO-1 specific T cell responses in metastatic melanoma patients with clinical benefit. Proc Natl Acad Sci U S A. 2008;105:20410–5.PubMedPubMedCentralCrossRef J Yuan et al. CTLA-4 blockade enhances polyfunctional NY-ESO-1 specific T cell responses in metastatic melanoma patients with clinical benefit. Proc Natl Acad Sci U S A. 2008;105:20410–5.PubMedPubMedCentralCrossRef
52.
go back to reference FS Hodi et al. Immunologic and clinical effects of antibody blockade of cytotoxic T lymphocyte-associated antigen 4 in previously vaccinated cancer patients. Proc Natl Acad Sci U S A. 2008;105:3005–10.PubMedPubMedCentralCrossRef FS Hodi et al. Immunologic and clinical effects of antibody blockade of cytotoxic T lymphocyte-associated antigen 4 in previously vaccinated cancer patients. Proc Natl Acad Sci U S A. 2008;105:3005–10.PubMedPubMedCentralCrossRef
53.
go back to reference Martens A, et al. Increases in Absolute Lymphocytes and Circulating CD4 + and CD8 + T Cells Are Associated with Positive Clinical Outcome of Melanoma Patients Treated with Ipilimumab. Clin Cancer Res. 2016;22:4848–58.PubMedPubMedCentralCrossRef Martens A, et al. Increases in Absolute Lymphocytes and Circulating CD4 + and CD8 + T Cells Are Associated with Positive Clinical Outcome of Melanoma Patients Treated with Ipilimumab. Clin Cancer Res. 2016;22:4848–58.PubMedPubMedCentralCrossRef
54.
go back to reference S Kelderman et al. Lactate dehydrogenase as a selection criterion for ipilimumab treatment in metastatic melanoma. Cancer Immunol Immunother. 2014;63:449–58.PubMed S Kelderman et al. Lactate dehydrogenase as a selection criterion for ipilimumab treatment in metastatic melanoma. Cancer Immunol Immunother. 2014;63:449–58.PubMed
55.
go back to reference GY Ku et al. Single-institution experience with ipilimumab in advanced melanoma patients in the compassionate use setting: lymphocyte count after 2 doses correlates with survival. Cancer. 2010;116:1767–75.PubMedCrossRef GY Ku et al. Single-institution experience with ipilimumab in advanced melanoma patients in the compassionate use setting: lymphocyte count after 2 doses correlates with survival. Cancer. 2010;116:1767–75.PubMedCrossRef
56.
go back to reference J Delyon et al. Experience in daily practice with ipilimumab for the treatment of patients with metastatic melanoma: an early increase in lymphocyte and eosinophil counts is associated with improved survival. Ann Oncol. 2013;24:1697–703.PubMedCrossRef J Delyon et al. Experience in daily practice with ipilimumab for the treatment of patients with metastatic melanoma: an early increase in lymphocyte and eosinophil counts is associated with improved survival. Ann Oncol. 2013;24:1697–703.PubMedCrossRef
57.
go back to reference AA Sarnaik et al. Extended dose ipilimumab with a peptide vaccine: immune correlates associated with clinical benefit in patients with resected high-risk stage IIIc/IV melanoma. Clin Cancer Res. 2011;17:896–906.CrossRefPubMed AA Sarnaik et al. Extended dose ipilimumab with a peptide vaccine: immune correlates associated with clinical benefit in patients with resected high-risk stage IIIc/IV melanoma. Clin Cancer Res. 2011;17:896–906.CrossRefPubMed
58.
go back to reference Ribas A, et al. Imaging of CTLA4 blockade-induced cell replication with (18)F-FLT PET in patients with advanced melanoma treated with tremelimumab. J Nucl Med. 2010;51:340–6.PubMedCrossRef Ribas A, et al. Imaging of CTLA4 blockade-induced cell replication with (18)F-FLT PET in patients with advanced melanoma treated with tremelimumab. J Nucl Med. 2010;51:340–6.PubMedCrossRef
60.
go back to reference Cha E, et al. Improved survival with T cell clonotype stability after anti-CTLA-4 treatment in cancer patients. Sci Transl Med. 2014;6:238ra270.CrossRef Cha E, et al. Improved survival with T cell clonotype stability after anti-CTLA-4 treatment in cancer patients. Sci Transl Med. 2014;6:238ra270.CrossRef
61.
go back to reference DY Oh et al. Immune Toxicities Elicted by CTLA-4 Blockade in Cancer Patients Are Associated with Early Diversification of the T-cell Repertoire. Cancer Res. 2017;77:1322–30.PubMedCrossRef DY Oh et al. Immune Toxicities Elicted by CTLA-4 Blockade in Cancer Patients Are Associated with Early Diversification of the T-cell Repertoire. Cancer Res. 2017;77:1322–30.PubMedCrossRef
62.
go back to reference Arakawa A, et al. Clonality of CD4(+) Blood T Cells Predicts Longer Survival With CTLA4 or PD-1 Checkpoint Inhibition in Advanced Melanoma. Front Immunol. 2019;10:1336.PubMedPubMedCentralCrossRef Arakawa A, et al. Clonality of CD4(+) Blood T Cells Predicts Longer Survival With CTLA4 or PD-1 Checkpoint Inhibition in Advanced Melanoma. Front Immunol. 2019;10:1336.PubMedPubMedCentralCrossRef
63.
go back to reference Messaoudi I, Guevara Patino JA, Dyall R, LeMaoult J. J. Nikolich-Zugich, Direct link between mhc polymorphism, T cell avidity, and diversity in immune defense. Science. 2002;298:1797–800. Messaoudi I, Guevara Patino JA, Dyall R, LeMaoult J. J. Nikolich-Zugich, Direct link between mhc polymorphism, T cell avidity, and diversity in immune defense. Science. 2002;298:1797–800.
64.
go back to reference P Kvistborg et al. Anti-CTLA-4 therapy broadens the melanoma-reactive CD8 + T cell response. Sci Transl Med. 2014;6:254ra128.PubMedCrossRef P Kvistborg et al. Anti-CTLA-4 therapy broadens the melanoma-reactive CD8 + T cell response. Sci Transl Med. 2014;6:254ra128.PubMedCrossRef
65.
go back to reference SK Subudhi et al. Clonal expansion of CD8 T cells in the systemic circulation precedes development of ipilimumab-induced toxicities. Proc Natl Acad Sci U S A. 2016;113:11919–24.PubMedPubMedCentralCrossRef SK Subudhi et al. Clonal expansion of CD8 T cells in the systemic circulation precedes development of ipilimumab-induced toxicities. Proc Natl Acad Sci U S A. 2016;113:11919–24.PubMedPubMedCentralCrossRef
66.
go back to reference MA Postow et al. Peripheral T cell receptor diversity is associated with clinical outcomes following ipilimumab treatment in metastatic melanoma. J Immunother Cancer. 2015;3:23.PubMedPubMedCentralCrossRef MA Postow et al. Peripheral T cell receptor diversity is associated with clinical outcomes following ipilimumab treatment in metastatic melanoma. J Immunother Cancer. 2015;3:23.PubMedPubMedCentralCrossRef
67.
go back to reference DB Page et al. Deep Sequencing of T-cell Receptor DNA as a Biomarker of Clonally Expanded TILs in Breast Cancer after Immunotherapy. Cancer Immunol Res. 2016;4:835–44.PubMedPubMedCentralCrossRef DB Page et al. Deep Sequencing of T-cell Receptor DNA as a Biomarker of Clonally Expanded TILs in Breast Cancer after Immunotherapy. Cancer Immunol Res. 2016;4:835–44.PubMedPubMedCentralCrossRef
68.
go back to reference Mougiakakos D, Choudhury A, Lladser A, Kiessling R, Johansson CC. Regulatory T cells in cancer. Adv Cancer Res. 2010;107:57–117.PubMedCrossRef Mougiakakos D, Choudhury A, Lladser A, Kiessling R, Johansson CC. Regulatory T cells in cancer. Adv Cancer Res. 2010;107:57–117.PubMedCrossRef
69.
go back to reference Wei T, Zhong W, Li Q. Role of heterogeneous regulatory T cells in the tumor microenvironment. Pharmacol Res. 2020;153:104659.PubMedCrossRef Wei T, Zhong W, Li Q. Role of heterogeneous regulatory T cells in the tumor microenvironment. Pharmacol Res. 2020;153:104659.PubMedCrossRef
71.
go back to reference T Takahashi et al. Immunologic self-tolerance maintained by CD25(+)CD4(+) regulatory T cells constitutively expressing cytotoxic T lymphocyte-associated antigen 4. J Exp Med. 2000;192:303–10.PubMedPubMedCentralCrossRef T Takahashi et al. Immunologic self-tolerance maintained by CD25(+)CD4(+) regulatory T cells constitutively expressing cytotoxic T lymphocyte-associated antigen 4. J Exp Med. 2000;192:303–10.PubMedPubMedCentralCrossRef
72.
go back to reference Zheng Y, Rudensky AY. Foxp3 in control of the regulatory T cell lineage. Nat Immunol. 2007;8:457–62.PubMedCrossRef Zheng Y, Rudensky AY. Foxp3 in control of the regulatory T cell lineage. Nat Immunol. 2007;8:457–62.PubMedCrossRef
73.
go back to reference Chen C, Rowell EA, Thomas RM, Hancock WW, Wells AD. Transcriptional regulation by Foxp3 is associated with direct promoter occupancy and modulation of histone acetylation. J Biol Chem. 2006;281:36828–34.PubMedCrossRef Chen C, Rowell EA, Thomas RM, Hancock WW, Wells AD. Transcriptional regulation by Foxp3 is associated with direct promoter occupancy and modulation of histone acetylation. J Biol Chem. 2006;281:36828–34.PubMedCrossRef
74.
go back to reference S Read et al. Blockade of CTLA-4 on CD4 + CD25 + regulatory T cells abrogates their function in vivo. J Immunol. 2006;177:4376–83.PubMedCrossRef S Read et al. Blockade of CTLA-4 on CD4 + CD25 + regulatory T cells abrogates their function in vivo. J Immunol. 2006;177:4376–83.PubMedCrossRef
76.
77.
go back to reference Peggs KS, Quezada SA, Chambers CA, Korman AJ, Allison JP. Blockade of CTLA-4 on both effector and regulatory T cell compartments contributes to the antitumor activity of anti-CTLA-4 antibodies. J Exp Med. 2009;206:1717–25.PubMedPubMedCentralCrossRef Peggs KS, Quezada SA, Chambers CA, Korman AJ, Allison JP. Blockade of CTLA-4 on both effector and regulatory T cell compartments contributes to the antitumor activity of anti-CTLA-4 antibodies. J Exp Med. 2009;206:1717–25.PubMedPubMedCentralCrossRef
78.
go back to reference MJ Selby et al. Anti-CTLA-4 antibodies of IgG2a isotype enhance antitumor activity through reduction of intratumoral regulatory T cells. Cancer Immunol Res. 2013;1:32–42.PubMedCrossRef MJ Selby et al. Anti-CTLA-4 antibodies of IgG2a isotype enhance antitumor activity through reduction of intratumoral regulatory T cells. Cancer Immunol Res. 2013;1:32–42.PubMedCrossRef
79.
go back to reference Tang F, Du X, Liu M, Zheng P, Liu Y. Anti-CTLA-4 antibodies in cancer immunotherapy: selective depletion of intratumoral regulatory T cells or checkpoint blockade? Cell Biosci. 2018;8:30.PubMedPubMedCentralCrossRef Tang F, Du X, Liu M, Zheng P, Liu Y. Anti-CTLA-4 antibodies in cancer immunotherapy: selective depletion of intratumoral regulatory T cells or checkpoint blockade? Cell Biosci. 2018;8:30.PubMedPubMedCentralCrossRef
80.
go back to reference TR Simpson et al. Fc-dependent depletion of tumor-infiltrating regulatory T cells co-defines the efficacy of anti-CTLA-4 therapy against melanoma. J Exp Med. 2013;210:1695–710.PubMedPubMedCentralCrossRef TR Simpson et al. Fc-dependent depletion of tumor-infiltrating regulatory T cells co-defines the efficacy of anti-CTLA-4 therapy against melanoma. J Exp Med. 2013;210:1695–710.PubMedPubMedCentralCrossRef
81.
83.
go back to reference Pages F, et al. International validation of the consensus Immunoscore for the classification of colon cancer: a prognostic and accuracy study. Lancet. 2018;391:2128–39.PubMedCrossRef Pages F, et al. International validation of the consensus Immunoscore for the classification of colon cancer: a prognostic and accuracy study. Lancet. 2018;391:2128–39.PubMedCrossRef
84.
go back to reference Saito T, et al. Two FOXP3(+)CD4(+) T cell subpopulations distinctly control the prognosis of colorectal cancers. Nat Med. 2016;22:679–84.CrossRefPubMed Saito T, et al. Two FOXP3(+)CD4(+) T cell subpopulations distinctly control the prognosis of colorectal cancers. Nat Med. 2016;22:679–84.CrossRefPubMed
85.
go back to reference Quezada SA, Peggs KS, Curran MA, Allison JP. CTLA4 blockade and GM-CSF combination immunotherapy alters the intratumor balance of effector and regulatory T cells. J Clin Invest. 2006;116:1935–45.PubMedPubMedCentralCrossRef Quezada SA, Peggs KS, Curran MA, Allison JP. CTLA4 blockade and GM-CSF combination immunotherapy alters the intratumor balance of effector and regulatory T cells. J Clin Invest. 2006;116:1935–45.PubMedPubMedCentralCrossRef
87.
go back to reference J Bjoern et al. Immunological correlates of treatment and response in stage IV malignant melanoma patients treated with Ipilimumab. Oncoimmunology. 2016;5:e1100788.PubMedCrossRef J Bjoern et al. Immunological correlates of treatment and response in stage IV malignant melanoma patients treated with Ipilimumab. Oncoimmunology. 2016;5:e1100788.PubMedCrossRef
88.
go back to reference AA Tarhini et al. Immune monitoring of the circulation and the tumor microenvironment in patients with regionally advanced melanoma receiving neoadjuvant ipilimumab. PLoS One. 2014;9:e87705.PubMedPubMedCentralCrossRef AA Tarhini et al. Immune monitoring of the circulation and the tumor microenvironment in patients with regionally advanced melanoma receiving neoadjuvant ipilimumab. PLoS One. 2014;9:e87705.PubMedPubMedCentralCrossRef
89.
go back to reference S Khan et al. Tremelimumab (anti-CTLA4) mediates immune responses mainly by direct activation of T effector cells rather than by affecting T regulatory cells. Clin Immunol. 2011;138:85–96.PubMedCrossRef S Khan et al. Tremelimumab (anti-CTLA4) mediates immune responses mainly by direct activation of T effector cells rather than by affecting T regulatory cells. Clin Immunol. 2011;138:85–96.PubMedCrossRef
90.
go back to reference E Simeone et al. Immunological and biological changes during ipilimumab treatment and their potential correlation with clinical response and survival in patients with advanced melanoma. Cancer Immunol Immunother. 2014;63:675–83.PubMedCrossRef E Simeone et al. Immunological and biological changes during ipilimumab treatment and their potential correlation with clinical response and survival in patients with advanced melanoma. Cancer Immunol Immunother. 2014;63:675–83.PubMedCrossRef
91.
go back to reference Maker AV, Attia P, Rosenberg SA. Analysis of the cellular mechanism of antitumor responses and autoimmunity in patients treated with CTLA-4 blockade. J Immunol. 2005;175:7746–54.PubMedCrossRef Maker AV, Attia P, Rosenberg SA. Analysis of the cellular mechanism of antitumor responses and autoimmunity in patients treated with CTLA-4 blockade. J Immunol. 2005;175:7746–54.PubMedCrossRef
92.
go back to reference Romano E, et al. Ipilimumab-dependent cell-mediated cytotoxicity of regulatory T cells ex vivo by nonclassical monocytes in melanoma patients. Proc Natl Acad Sci U S A. 2015;112:6140–5.PubMedPubMedCentralCrossRef Romano E, et al. Ipilimumab-dependent cell-mediated cytotoxicity of regulatory T cells ex vivo by nonclassical monocytes in melanoma patients. Proc Natl Acad Sci U S A. 2015;112:6140–5.PubMedPubMedCentralCrossRef
93.
go back to reference Sharma A, et al. Anti-CTLA-4 Immunotherapy Does Not Deplete FOXP3(+) Regulatory T Cells (Tregs) in Human Cancers-Response. Clin Cancer Res. 2019;25:3469–70.PubMedPubMedCentralCrossRef Sharma A, et al. Anti-CTLA-4 Immunotherapy Does Not Deplete FOXP3(+) Regulatory T Cells (Tregs) in Human Cancers-Response. Clin Cancer Res. 2019;25:3469–70.PubMedPubMedCentralCrossRef
94.
go back to reference Quezada SA, Peggs KS. Lost in Translation: Deciphering the Mechanism of Action of Anti-human CTLA-4. Clin Cancer Res. 2019;25:1130–2.PubMedCrossRef Quezada SA, Peggs KS. Lost in Translation: Deciphering the Mechanism of Action of Anti-human CTLA-4. Clin Cancer Res. 2019;25:1130–2.PubMedCrossRef
98.
go back to reference V Verma et al. PD-1 blockade in subprimed CD8 cells induces dysfunctional PD-1(+)CD38(hi) cells and anti-PD-1 resistance. Nat Immunol. 2019;20:1231–43.PubMedPubMedCentralCrossRef V Verma et al. PD-1 blockade in subprimed CD8 cells induces dysfunctional PD-1(+)CD38(hi) cells and anti-PD-1 resistance. Nat Immunol. 2019;20:1231–43.PubMedPubMedCentralCrossRef
99.
go back to reference CS Grasso et al., Conserved Interferon-gamma Signaling Drives Clinical Response to Immune Checkpoint Blockade Therapy in Melanoma. Cancer Cell, (2020). CS Grasso et al., Conserved Interferon-gamma Signaling Drives Clinical Response to Immune Checkpoint Blockade Therapy in Melanoma. Cancer Cell, (2020).
101.
go back to reference AO Kamphorst et al. Proliferation of PD-1 + CD8 T cells in peripheral blood after PD-1-targeted therapy in lung cancer patients. Proc Natl Acad Sci U S A. 2017;114:4993–8.PubMedPubMedCentralCrossRef AO Kamphorst et al. Proliferation of PD-1 + CD8 T cells in peripheral blood after PD-1-targeted therapy in lung cancer patients. Proc Natl Acad Sci U S A. 2017;114:4993–8.PubMedPubMedCentralCrossRef
102.
go back to reference KH Kim et al. The First-week Proliferative Response of Peripheral Blood PD-1(+)CD8(+) T Cells Predicts the Response to Anti-PD-1 Therapy in Solid Tumors. Clin Cancer Res. 2019;25:2144–54.PubMedCrossRef KH Kim et al. The First-week Proliferative Response of Peripheral Blood PD-1(+)CD8(+) T Cells Predicts the Response to Anti-PD-1 Therapy in Solid Tumors. Clin Cancer Res. 2019;25:2144–54.PubMedCrossRef
103.
go back to reference Kamada T, et al. PD-1(+) regulatory T cells amplified by PD-1 blockade promote hyperprogression of cancer. Proc Natl Acad Sci U S A. 2019;116:9999–10008.PubMedPubMedCentralCrossRef Kamada T, et al. PD-1(+) regulatory T cells amplified by PD-1 blockade promote hyperprogression of cancer. Proc Natl Acad Sci U S A. 2019;116:9999–10008.PubMedPubMedCentralCrossRef
104.
go back to reference DM Woods et al. Decreased Suppression and Increased Phosphorylated STAT3 in Regulatory T Cells are Associated with Benefit from Adjuvant PD-1 Blockade in Resected Metastatic Melanoma. Clin Cancer Res. 2018;24:6236–47.PubMedPubMedCentralCrossRef DM Woods et al. Decreased Suppression and Increased Phosphorylated STAT3 in Regulatory T Cells are Associated with Benefit from Adjuvant PD-1 Blockade in Resected Metastatic Melanoma. Clin Cancer Res. 2018;24:6236–47.PubMedPubMedCentralCrossRef
105.
go back to reference Zappasodi R, et al. Non-conventional Inhibitory CD4(+)Foxp3(-)PD-1(hi) T Cells as a Biomarker of Immune Checkpoint Blockade Activity. Cancer Cell. 2018;34:691.PubMedPubMedCentralCrossRef Zappasodi R, et al. Non-conventional Inhibitory CD4(+)Foxp3(-)PD-1(hi) T Cells as a Biomarker of Immune Checkpoint Blockade Activity. Cancer Cell. 2018;34:691.PubMedPubMedCentralCrossRef
107.
go back to reference J Han et al. TCR Repertoire Diversity of Peripheral PD-1(+)CD8(+) T Cells Predicts Clinical Outcomes after Immunotherapy in Patients with Non-Small Cell Lung Cancer. Cancer Immunol Res. 2020;8:146–54. J Han et al. TCR Repertoire Diversity of Peripheral PD-1(+)CD8(+) T Cells Predicts Clinical Outcomes after Immunotherapy in Patients with Non-Small Cell Lung Cancer. Cancer Immunol Res. 2020;8:146–54.
108.
go back to reference Snyder A, et al. Contribution of systemic and somatic factors to clinical response and resistance to PD-L1 blockade in urothelial cancer: An exploratory multi-omic analysis. PLoS Med. 2017;14:e1002309.PubMedPubMedCentralCrossRef Snyder A, et al. Contribution of systemic and somatic factors to clinical response and resistance to PD-L1 blockade in urothelial cancer: An exploratory multi-omic analysis. PLoS Med. 2017;14:e1002309.PubMedPubMedCentralCrossRef
109.
go back to reference Gebhardt C, et al. Myeloid Cells and Related Chronic Inflammatory Factors as Novel Predictive Markers in Melanoma Treatment with Ipilimumab. Clin Cancer Res. 2015;21:5453–9.PubMedCrossRef Gebhardt C, et al. Myeloid Cells and Related Chronic Inflammatory Factors as Novel Predictive Markers in Melanoma Treatment with Ipilimumab. Clin Cancer Res. 2015;21:5453–9.PubMedCrossRef
110.
111.
112.
go back to reference Y Latchman et al. PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat Immunol. 2001;2:261–8.CrossRefPubMed Y Latchman et al. PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat Immunol. 2001;2:261–8.CrossRefPubMed
113.
114.
go back to reference SD Blackburn et al. Coregulation of CD8 + T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nat Immunol. 2009;10:29–37.PubMedCrossRef SD Blackburn et al. Coregulation of CD8 + T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nat Immunol. 2009;10:29–37.PubMedCrossRef
115.
go back to reference DS Thommen 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.PubMedPubMedCentralCrossRef DS Thommen 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.PubMedPubMedCentralCrossRef
116.
go back to reference H Li et al. Dysfunctional CD8 T Cells Form a Proliferative, Dynamically Regulated Compartment within Human Melanoma. Cell. 2019;176:775–89 e718.PubMedCrossRef H Li et al. Dysfunctional CD8 T Cells Form a Proliferative, Dynamically Regulated Compartment within Human Melanoma. Cell. 2019;176:775–89 e718.PubMedCrossRef
117.
121.
go back to reference RR Jadhav et al. Epigenetic signature of PD-1 + TCF1 + CD8 T cells that act as resource cells during chronic viral infection and respond to PD-1 blockade. Proc Natl Acad Sci U S A. 2019;116:14113–8.PubMedPubMedCentralCrossRef RR Jadhav et al. Epigenetic signature of PD-1 + TCF1 + CD8 T cells that act as resource cells during chronic viral infection and respond to PD-1 blockade. Proc Natl Acad Sci U S A. 2019;116:14113–8.PubMedPubMedCentralCrossRef
122.
go back to reference I Siddiqui 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 50, 195–211 e110 (2019). I Siddiqui 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 50, 195–211 e110 (2019).
123.
go back to reference BC Miller et al. Subsets of exhausted CD8(+) T cells differentially mediate tumor control and respond to checkpoint blockade. Nat Immunol. 2019;20:326–36.PubMedPubMedCentralCrossRef BC Miller et al. Subsets of exhausted CD8(+) T cells differentially mediate tumor control and respond to checkpoint blockade. Nat Immunol. 2019;20:326–36.PubMedPubMedCentralCrossRef
125.
126.
127.
go back to reference Kurtulus S, et al. Checkpoint Blockade Immunotherapy Induces Dynamic Changes in PD-1(-)CD8(+) Tumor-Infiltrating T Cells. Immunity. 2019;50:181–94 e186.PubMedPubMedCentralCrossRef Kurtulus S, et al. Checkpoint Blockade Immunotherapy Induces Dynamic Changes in PD-1(-)CD8(+) Tumor-Infiltrating T Cells. Immunity. 2019;50:181–94 e186.PubMedPubMedCentralCrossRef
128.
go back to reference AP Ganesan 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.PubMedPubMedCentralCrossRef AP Ganesan 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.PubMedPubMedCentralCrossRef
130.
go back to reference Forde PM, Chaft JE, Pardoll DM. Neoadjuvant PD-1 Blockade in Resectable Lung Cancer. N Engl J Med. 2018;379:e14.PubMedCrossRef Forde PM, Chaft JE, Pardoll DM. Neoadjuvant PD-1 Blockade in Resectable Lung Cancer. N Engl J Med. 2018;379:e14.PubMedCrossRef
131.
go back to reference M Merhi et al., Persistent anti-NY-ESO-1-specific T cells and expression of differential biomarkers in a patient with metastatic gastric cancer benefiting from combined radioimmunotherapy treatment: a case report. J Immunother Cancer 8, (2020). M Merhi et al., Persistent anti-NY-ESO-1-specific T cells and expression of differential biomarkers in a patient with metastatic gastric cancer benefiting from combined radioimmunotherapy treatment: a case report. J Immunother Cancer 8, (2020).
132.
go back to reference Sakaguchi S, Miyara M, Costantino CM, Hafler DA. FOXP3 + regulatory T cells in the human immune system. Nat Rev Immunol. 2010;10:490–500.PubMedCrossRef Sakaguchi S, Miyara M, Costantino CM, Hafler DA. FOXP3 + regulatory T cells in the human immune system. Nat Rev Immunol. 2010;10:490–500.PubMedCrossRef
133.
go back to reference DE Lowther et al., PD-1 marks dysfunctional regulatory T cells in malignant gliomas. JCI Insight 1, (2016). DE Lowther et al., PD-1 marks dysfunctional regulatory T cells in malignant gliomas. JCI Insight 1, (2016).
134.
go back to reference Togashi Y, Shitara K, Nishikawa H. Regulatory T cells in cancer immunosuppression - implications for anticancer therapy. Nat Rev Clin Oncol. 2019;16:356–71.PubMedCrossRef Togashi Y, Shitara K, Nishikawa H. Regulatory T cells in cancer immunosuppression - implications for anticancer therapy. Nat Rev Clin Oncol. 2019;16:356–71.PubMedCrossRef
135.
go back to reference Duraiswamy J, Kaluza KM, Freeman GJ, Coukos G. Dual blockade of PD-1 and CTLA-4 combined with tumor vaccine effectively restores T-cell rejection function in tumors. Cancer Res. 2013;73:3591–603.PubMedPubMedCentralCrossRef Duraiswamy J, Kaluza KM, Freeman GJ, Coukos G. Dual blockade of PD-1 and CTLA-4 combined with tumor vaccine effectively restores T-cell rejection function in tumors. Cancer Res. 2013;73:3591–603.PubMedPubMedCentralCrossRef
136.
go back to reference Q Zhou et al. Program death-1 signaling and regulatory T cells collaborate to resist the function of adoptively transferred cytotoxic T lymphocytes in advanced acute myeloid leukemia. Blood. 2010;116:2484–93.PubMedPubMedCentralCrossRef Q Zhou et al. Program death-1 signaling and regulatory T cells collaborate to resist the function of adoptively transferred cytotoxic T lymphocytes in advanced acute myeloid leukemia. Blood. 2010;116:2484–93.PubMedPubMedCentralCrossRef
137.
go back to reference HJ Park et al. PD-1 upregulated on regulatory T cells during chronic virus infection enhances the suppression of CD8 + T cell immune response via the interaction with PD-L1 expressed on CD8 + T cells. J Immunol. 2015;194:5801–11.PubMedCrossRef HJ Park et al. PD-1 upregulated on regulatory T cells during chronic virus infection enhances the suppression of CD8 + T cell immune response via the interaction with PD-L1 expressed on CD8 + T cells. J Immunol. 2015;194:5801–11.PubMedCrossRef
138.
go back to reference Kitazawa Y, et al. Involvement of the programmed death-1/programmed death-1 ligand pathway in CD4 + CD25 + regulatory T-cell activity to suppress alloimmune responses. Transplantation. 2007;83:774–82.PubMedCrossRef Kitazawa Y, et al. Involvement of the programmed death-1/programmed death-1 ligand pathway in CD4 + CD25 + regulatory T-cell activity to suppress alloimmune responses. Transplantation. 2007;83:774–82.PubMedCrossRef
140.
go back to reference W Wang et al. PD1 blockade reverses the suppression of melanoma antigen-specific CTL by CD4 + CD25(Hi) regulatory T cells. Int Immunol. 2009;21:1065–77.PubMedPubMedCentralCrossRef W Wang et al. PD1 blockade reverses the suppression of melanoma antigen-specific CTL by CD4 + CD25(Hi) regulatory T cells. Int Immunol. 2009;21:1065–77.PubMedPubMedCentralCrossRef
142.
143.
go back to reference Stathopoulou C, et al. PD-1 Inhibitory Receptor Downregulates Asparaginyl Endopeptidase and Maintains Foxp3 Transcription Factor Stability in Induced Regulatory T Cells. Immunity. 2018;49:247–63 e247.PubMedPubMedCentralCrossRef Stathopoulou C, et al. PD-1 Inhibitory Receptor Downregulates Asparaginyl Endopeptidase and Maintains Foxp3 Transcription Factor Stability in Induced Regulatory T Cells. Immunity. 2018;49:247–63 e247.PubMedPubMedCentralCrossRef
144.
go back to reference Wong M, La Cava A, Hahn BH. Blockade of programmed death-1 in young (New Zealand Black x New Zealand White)F1 mice promotes the suppressive capacity of CD4 + regulatory T cells protecting from lupus-like disease. J Immunol. 2013;190:5402–10.PubMedCrossRef Wong M, La Cava A, Hahn BH. Blockade of programmed death-1 in young (New Zealand Black x New Zealand White)F1 mice promotes the suppressive capacity of CD4 + regulatory T cells protecting from lupus-like disease. J Immunol. 2013;190:5402–10.PubMedCrossRef
145.
go back to reference Dodagatta-Marri E, et al. alpha-PD-1 therapy elevates Treg/Th balance and increases tumor cell pSmad3 that are both targeted by alpha-TGFbeta antibody to promote durable rejection and immunity in squamous cell carcinomas. J Immunother Cancer. 2019;7:62.PubMedPubMedCentralCrossRef Dodagatta-Marri E, et al. alpha-PD-1 therapy elevates Treg/Th balance and increases tumor cell pSmad3 that are both targeted by alpha-TGFbeta antibody to promote durable rejection and immunity in squamous cell carcinomas. J Immunother Cancer. 2019;7:62.PubMedPubMedCentralCrossRef
146.
go back to reference Franceschini D, et al. PD-L1 negatively regulates CD4 + CD25 + Foxp3 + Tregs by limiting STAT-5 phosphorylation in patients chronically infected with HCV. J Clin Invest. 2009;119:551–64.PubMedPubMedCentralCrossRef Franceschini D, et al. PD-L1 negatively regulates CD4 + CD25 + Foxp3 + Tregs by limiting STAT-5 phosphorylation in patients chronically infected with HCV. J Clin Invest. 2009;119:551–64.PubMedPubMedCentralCrossRef
147.
go back to reference Penaloza-MacMaster P, Provine NM, Blass E, Barouch DH. CD4 T Cell Depletion Substantially Augments the Rescue Potential of PD-L1 Blockade for Deeply Exhausted CD8 T Cells. J Immunol. 2015;195:1054–63.PubMedPubMedCentralCrossRef Penaloza-MacMaster P, Provine NM, Blass E, Barouch DH. CD4 T Cell Depletion Substantially Augments the Rescue Potential of PD-L1 Blockade for Deeply Exhausted CD8 T Cells. J Immunol. 2015;195:1054–63.PubMedPubMedCentralCrossRef
148.
150.
go back to reference R Cabrita et al. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature. 2020;577:561–5.PubMedCrossRef R Cabrita et al. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature. 2020;577:561–5.PubMedCrossRef
151.
go back to reference Erdag G, et al. Immunotype and immunohistologic characteristics of tumor-infiltrating immune cells are associated with clinical outcome in metastatic melanoma. Cancer Res. 2012;72:1070–80.PubMedPubMedCentralCrossRef Erdag G, et al. Immunotype and immunohistologic characteristics of tumor-infiltrating immune cells are associated with clinical outcome in metastatic melanoma. Cancer Res. 2012;72:1070–80.PubMedPubMedCentralCrossRef
152.
153.
154.
go back to reference JS Nielsen et al. CD20 + tumor-infiltrating lymphocytes have an atypical CD27- memory phenotype and together with CD8 + T cells promote favorable prognosis in ovarian cancer. Clin Cancer Res. 2012;18:3281–92.PubMedCrossRef JS Nielsen et al. CD20 + tumor-infiltrating lymphocytes have an atypical CD27- memory phenotype and together with CD8 + T cells promote favorable prognosis in ovarian cancer. Clin Cancer Res. 2012;18:3281–92.PubMedCrossRef
156.
go back to reference X Wang et al. PD-1-expressing B cells suppress CD4(+) and CD8(+) T cells via PD-1/PD-L1-dependent pathway. Mol Immunol. 2019;109:20–6.PubMedCrossRef X Wang et al. PD-1-expressing B cells suppress CD4(+) and CD8(+) T cells via PD-1/PD-L1-dependent pathway. Mol Immunol. 2019;109:20–6.PubMedCrossRef
157.
go back to reference Wouters MCA, Nelson BH. Prognostic Significance of Tumor-Infiltrating B Cells and Plasma Cells in Human Cancer. Clin Cancer Res. 2018;24:6125–35.PubMedCrossRef Wouters MCA, Nelson BH. Prognostic Significance of Tumor-Infiltrating B Cells and Plasma Cells in Human Cancer. Clin Cancer Res. 2018;24:6125–35.PubMedCrossRef
158.
go back to reference Neyt K, Perros F, GeurtsvanKessel CH, Hammad H, Lambrecht BN. Tertiary lymphoid organs in infection and autoimmunity. Trends Immunol. 2012;33:297–305.PubMedPubMedCentralCrossRef Neyt K, Perros F, GeurtsvanKessel CH, Hammad H, Lambrecht BN. Tertiary lymphoid organs in infection and autoimmunity. Trends Immunol. 2012;33:297–305.PubMedPubMedCentralCrossRef
159.
go back to reference FK Baddoura et al. Lymphoid neogenesis in murine cardiac allografts undergoing chronic rejection. Am J Transplant. 2005;5:510–6.PubMedCrossRef FK Baddoura et al. Lymphoid neogenesis in murine cardiac allografts undergoing chronic rejection. Am J Transplant. 2005;5:510–6.PubMedCrossRef
161.
go back to reference F Petitprez et al. B cells are associated with survival and immunotherapy response in sarcoma. Nature. 2020;577:556–60.PubMedCrossRef F Petitprez et al. B cells are associated with survival and immunotherapy response in sarcoma. Nature. 2020;577:556–60.PubMedCrossRef
162.
go back to reference DP Hollern et al., B Cells and T Follicular Helper Cells Mediate Response to Checkpoint Inhibitors in High Mutation Burden Mouse Models of Breast Cancer. Cell 179, 1191–206 e1121 (2019). DP Hollern et al., B Cells and T Follicular Helper Cells Mediate Response to Checkpoint Inhibitors in High Mutation Burden Mouse Models of Breast Cancer. Cell 179, 1191–206 e1121 (2019).
163.
go back to reference G Bindea et al. Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer. Immunity. 2013;39:782–95.PubMedCrossRef G Bindea et al. Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer. Immunity. 2013;39:782–95.PubMedCrossRef
166.
go back to reference Nakamura K, Smyth MJ. Myeloid immunosuppression and immune checkpoints in the tumor microenvironment. Cell Mol Immunol. 2020;17:1–12.PubMedCrossRef Nakamura K, Smyth MJ. Myeloid immunosuppression and immune checkpoints in the tumor microenvironment. Cell Mol Immunol. 2020;17:1–12.PubMedCrossRef
168.
go back to reference MM Gubin et al. High-Dimensional Analysis Delineates Myeloid and Lymphoid Compartment Remodeling during Successful Immune-Checkpoint Cancer Therapy. Cell. 2018;175:1443.PubMedPubMedCentralCrossRef MM Gubin et al. High-Dimensional Analysis Delineates Myeloid and Lymphoid Compartment Remodeling during Successful Immune-Checkpoint Cancer Therapy. Cell. 2018;175:1443.PubMedPubMedCentralCrossRef
169.
go back to reference Xiong H, et al. Anti-PD-L1 Treatment Results in Functional Remodeling of the Macrophage Compartment. Cancer Res. 2019;79:1493–506.PubMedCrossRef Xiong H, et al. Anti-PD-L1 Treatment Results in Functional Remodeling of the Macrophage Compartment. Cancer Res. 2019;79:1493–506.PubMedCrossRef
170.
go back to reference Y Zhu et al. CSF1/CSF1R blockade reprograms tumor-infiltrating macrophages and improves response to T-cell checkpoint immunotherapy in pancreatic cancer models. Cancer Res. 2014;74:5057–69.PubMedPubMedCentralCrossRef Y Zhu et al. CSF1/CSF1R blockade reprograms tumor-infiltrating macrophages and improves response to T-cell checkpoint immunotherapy in pancreatic cancer models. Cancer Res. 2014;74:5057–69.PubMedPubMedCentralCrossRef
171.
go back to reference PA Beavis et al. Dual PD-1 and CTLA-4 Checkpoint Blockade Promotes Antitumor Immune Responses through CD4(+)Foxp3(-) Cell-Mediated Modulation of CD103(+) Dendritic Cells. Cancer Immunol Res. 2018;6:1069–81.PubMedCrossRef PA Beavis et al. Dual PD-1 and CTLA-4 Checkpoint Blockade Promotes Antitumor Immune Responses through CD4(+)Foxp3(-) Cell-Mediated Modulation of CD103(+) Dendritic Cells. Cancer Immunol Res. 2018;6:1069–81.PubMedCrossRef
172.
go back to reference S Laurent et al. CTLA-4 is expressed by human monocyte-derived dendritic cells and regulates their functions. Hum Immunol. 2010;71:934–41.PubMedCrossRef S Laurent et al. CTLA-4 is expressed by human monocyte-derived dendritic cells and regulates their functions. Hum Immunol. 2010;71:934–41.PubMedCrossRef
173.
go back to reference Y Liu et al. Regulation of arginase I activity and expression by both PD-1 and CTLA-4 on the myeloid-derived suppressor cells. Cancer Immunol Immunother. 2009;58:687–97.PubMedCrossRef Y Liu et al. Regulation of arginase I activity and expression by both PD-1 and CTLA-4 on the myeloid-derived suppressor cells. Cancer Immunol Immunother. 2009;58:687–97.PubMedCrossRef
174.
go back to reference PC Rodriguez 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.PubMedCrossRef PC Rodriguez 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.PubMedCrossRef
175.
go back to reference Strauss L, et al., Targeted deletion of PD-1 in myeloid cells induces antitumor immunity. Sci Immunol 5, (2020). Strauss L, et al., Targeted deletion of PD-1 in myeloid cells induces antitumor immunity. Sci Immunol 5, (2020).
176.
go back to reference SH Kim et al. Phenformin Inhibits Myeloid-Derived Suppressor Cells and Enhances the Anti-Tumor Activity of PD-1 Blockade in Melanoma. J Invest Dermatol. 2017;137:1740–8.PubMedCrossRef SH Kim et al. Phenformin Inhibits Myeloid-Derived Suppressor Cells and Enhances the Anti-Tumor Activity of PD-1 Blockade in Melanoma. J Invest Dermatol. 2017;137:1740–8.PubMedCrossRef
177.
go back to reference K Kim et al. Eradication of metastatic mouse cancers resistant to immune checkpoint blockade by suppression of myeloid-derived cells. Proc Natl Acad Sci U S A. 2014;111:11774–9.PubMedPubMedCentralCrossRef K Kim et al. Eradication of metastatic mouse cancers resistant to immune checkpoint blockade by suppression of myeloid-derived cells. Proc Natl Acad Sci U S A. 2014;111:11774–9.PubMedPubMedCentralCrossRef
178.
go back to reference Orillion A, et al. Entinostat Neutralizes Myeloid-Derived Suppressor Cells and Enhances the Antitumor Effect of PD-1 Inhibition in Murine Models of Lung and Renal Cell Carcinoma. Clin Cancer Res. 2017;23:5187–201.PubMedPubMedCentralCrossRef Orillion A, et al. Entinostat Neutralizes Myeloid-Derived Suppressor Cells and Enhances the Antitumor Effect of PD-1 Inhibition in Murine Models of Lung and Renal Cell Carcinoma. Clin Cancer Res. 2017;23:5187–201.PubMedPubMedCentralCrossRef
179.
go back to reference PE Clavijo et al. Resistance to CTLA-4 checkpoint inhibition reversed through selective elimination of granulocytic myeloid cells. Oncotarget. 2017;8:55804–20.PubMedPubMedCentralCrossRef PE Clavijo et al. Resistance to CTLA-4 checkpoint inhibition reversed through selective elimination of granulocytic myeloid cells. Oncotarget. 2017;8:55804–20.PubMedPubMedCentralCrossRef
180.
go back to reference Davis RJ, et al. Anti-PD-L1 Efficacy Can Be Enhanced by Inhibition of Myeloid-Derived Suppressor Cells with a Selective Inhibitor of PI3Kdelta/gamma. Cancer Res. 2017;77:2607–19.PubMedPubMedCentralCrossRef Davis RJ, et al. Anti-PD-L1 Efficacy Can Be Enhanced by Inhibition of Myeloid-Derived Suppressor Cells with a Selective Inhibitor of PI3Kdelta/gamma. Cancer Res. 2017;77:2607–19.PubMedPubMedCentralCrossRef
181.
go back to reference Meyer C, et al. Frequencies of circulating MDSC correlate with clinical outcome of melanoma patients treated with ipilimumab. Cancer Immunol Immunother. 2014;63:247–57.PubMedCrossRef Meyer C, et al. Frequencies of circulating MDSC correlate with clinical outcome of melanoma patients treated with ipilimumab. Cancer Immunol Immunother. 2014;63:247–57.PubMedCrossRef
183.
go back to reference Krieg C, et al. High-dimensional single-cell analysis predicts response to anti-PD-1 immunotherapy. Nat Med. 2018;24:144–53.PubMedCrossRef Krieg C, et al. High-dimensional single-cell analysis predicts response to anti-PD-1 immunotherapy. Nat Med. 2018;24:144–53.PubMedCrossRef
184.
go back to reference M Bonomi et al. Circulating immune biomarkers as predictors of the response to pembrolizumab and weekly low dose carboplatin and paclitaxel in NSCLC and poor PS: An interim analysis. Oncol Lett. 2019;17:1349–56.PubMed M Bonomi et al. Circulating immune biomarkers as predictors of the response to pembrolizumab and weekly low dose carboplatin and paclitaxel in NSCLC and poor PS: An interim analysis. Oncol Lett. 2019;17:1349–56.PubMed
185.
go back to reference Biswas SK, Mantovani A. Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm. Nat Immunol. 2010;11:889–96.PubMedCrossRef Biswas SK, Mantovani A. Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm. Nat Immunol. 2010;11:889–96.PubMedCrossRef
186.
go back to reference S Koyama et al. Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints. Nat Commun. 2016;7:10501.PubMedPubMedCentralCrossRef S Koyama et al. Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints. Nat Commun. 2016;7:10501.PubMedPubMedCentralCrossRef
187.
go back to reference Curran MA, Montalvo W, Yagita H, Allison JP. 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.PubMedPubMedCentralCrossRef Curran MA, Montalvo W, Yagita H, Allison JP. 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.PubMedPubMedCentralCrossRef
188.
go back to reference J Gao et al. VISTA is an inhibitory immune checkpoint that is increased after ipilimumab therapy in patients with prostate cancer. Nat Med. 2017;23:551–5.PubMedPubMedCentralCrossRef J Gao et al. VISTA is an inhibitory immune checkpoint that is increased after ipilimumab therapy in patients with prostate cancer. Nat Med. 2017;23:551–5.PubMedPubMedCentralCrossRef
189.
go back to reference Reese Z, Straubhar A, Pal SK, Agarwal N. Ipilimumab in the treatment of prostate cancer. Future Oncol. 2015;11:27–37.PubMedCrossRef Reese Z, Straubhar A, Pal SK, Agarwal N. Ipilimumab in the treatment of prostate cancer. Future Oncol. 2015;11:27–37.PubMedCrossRef
190.
192.
go back to reference MD Hellmann et al. Nivolumab plus Ipilimumab in Advanced Non-Small-Cell Lung Cancer. N Engl J Med. 2019;381:2020–31.PubMedCrossRef MD Hellmann et al. Nivolumab plus Ipilimumab in Advanced Non-Small-Cell Lung Cancer. N Engl J Med. 2019;381:2020–31.PubMedCrossRef
193.
go back to reference S Spranger et al. Mechanism of tumor rejection with doublets of CTLA-4, PD-1/PD-L1, or IDO blockade involves restored IL-2 production and proliferation of CD8(+) T cells directly within the tumor microenvironment. J Immunother Cancer. 2014;2:3.PubMedPubMedCentralCrossRef S Spranger et al. Mechanism of tumor rejection with doublets of CTLA-4, PD-1/PD-L1, or IDO blockade involves restored IL-2 production and proliferation of CD8(+) T cells directly within the tumor microenvironment. J Immunother Cancer. 2014;2:3.PubMedPubMedCentralCrossRef
194.
go back to reference SC Wei et al. Negative Co-stimulation Constrains T Cell Differentiation by Imposing Boundaries on Possible Cell States. Immunity. 2019;50:1084–98 e1010.PubMedPubMedCentralCrossRef SC Wei et al. Negative Co-stimulation Constrains T Cell Differentiation by Imposing Boundaries on Possible Cell States. Immunity. 2019;50:1084–98 e1010.PubMedPubMedCentralCrossRef
195.
go back to reference JS Weber et al. Sequential administration of nivolumab and ipilimumab with a planned switch in patients with advanced melanoma (CheckMate 064): an open-label, randomised, phase 2 trial. Lancet Oncol. 2016;17:943–55.PubMedPubMedCentralCrossRef JS Weber et al. Sequential administration of nivolumab and ipilimumab with a planned switch in patients with advanced melanoma (CheckMate 064): an open-label, randomised, phase 2 trial. Lancet Oncol. 2016;17:943–55.PubMedPubMedCentralCrossRef
196.
go back to reference Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion. Science. 2011;331:1565–70.PubMedCrossRef Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion. Science. 2011;331:1565–70.PubMedCrossRef
197.
go back to reference O’Donnell JS, Teng MWL, Smyth MJ. Cancer immunoediting and resistance to T cell-based immunotherapy. Nat Rev Clin Oncol. 2019;16:151–67.PubMedCrossRef O’Donnell JS, Teng MWL, Smyth MJ. Cancer immunoediting and resistance to T cell-based immunotherapy. Nat Rev Clin Oncol. 2019;16:151–67.PubMedCrossRef
198.
go back to reference Dunn GP, Old LJ, Schreiber RD. The immunobiology of cancer immunosurveillance and immunoediting. Immunity. 2004;21:137–48.PubMedCrossRef Dunn GP, Old LJ, Schreiber RD. The immunobiology of cancer immunosurveillance and immunoediting. Immunity. 2004;21:137–48.PubMedCrossRef
199.
go back to reference Rooney MS, Shukla SA, Wu CJ, Getz G, Hacohen N. Molecular and genetic properties of tumors associated with local immune cytolytic activity. Cell. 2015;160:48–61.PubMedPubMedCentralCrossRef Rooney MS, Shukla SA, Wu CJ, Getz G, Hacohen N. Molecular and genetic properties of tumors associated with local immune cytolytic activity. Cell. 2015;160:48–61.PubMedPubMedCentralCrossRef
203.
go back to reference Nsengimana J, et al. beta-Catenin-mediated immune evasion pathway frequently operates in primary cutaneous melanomas. J Clin Invest. 2018;128:2048–63.PubMedPubMedCentralCrossRef Nsengimana J, et al. beta-Catenin-mediated immune evasion pathway frequently operates in primary cutaneous melanomas. J Clin Invest. 2018;128:2048–63.PubMedPubMedCentralCrossRef
205.
go back to reference Gao J, et al, Loss of IFN-gamma Pathway Genes in Tumor Cells as a Mechanism of Resistance to Anti-CTLA-4 Therapy. Cell167, 397–404 e399 (2016). Gao J, et al, Loss of IFN-gamma Pathway Genes in Tumor Cells as a Mechanism of Resistance to Anti-CTLA-4 Therapy. Cell167, 397–404 e399 (2016).
206.
go back to reference A Jimenez-Sanchez et al. Heterogeneous Tumor-Immune Microenvironments among Differentially Growing Metastases in an Ovarian Cancer Patient. Cell. 2017;170:927–38 e920.PubMedPubMedCentralCrossRef A Jimenez-Sanchez et al. Heterogeneous Tumor-Immune Microenvironments among Differentially Growing Metastases in an Ovarian Cancer Patient. Cell. 2017;170:927–38 e920.PubMedPubMedCentralCrossRef
207.
go back to reference M Angelova et al. Evolution of Metastases in Space and Time under Immune Selection. Cell. 2018;175:751–65 e716.PubMedCrossRef M Angelova et al. Evolution of Metastases in Space and Time under Immune Selection. Cell. 2018;175:751–65 e716.PubMedCrossRef
208.
go back to reference Westerhoff HV, Palsson BO. The evolution of molecular biology into systems biology. Nat Biotechnol. 2004;22:1249–52.PubMedCrossRef Westerhoff HV, Palsson BO. The evolution of molecular biology into systems biology. Nat Biotechnol. 2004;22:1249–52.PubMedCrossRef
209.
go back to reference Chaussabel D. Assessment of immune status using blood transcriptomics and potential implications for global health. Semin Immunol. 2015;27:58–66.PubMedCrossRef Chaussabel D. Assessment of immune status using blood transcriptomics and potential implications for global health. Semin Immunol. 2015;27:58–66.PubMedCrossRef
210.
211.
go back to reference Chaussabel D, Pulendran B. A vision and a prescription for big data-enabled medicine. Nat Immunol. 2015;16:435–9.PubMedCrossRef Chaussabel D, Pulendran B. A vision and a prescription for big data-enabled medicine. Nat Immunol. 2015;16:435–9.PubMedCrossRef
212.
go back to reference Nakaya HI, Pulendran B. Vaccinology in the era of high-throughput biology. Philos Trans R Soc Lond B Biol Sci 370, (2015). Nakaya HI, Pulendran B. Vaccinology in the era of high-throughput biology. Philos Trans R Soc Lond B Biol Sci 370, (2015).
213.
go back to reference Obermoser G, et al. Systems scale interactive exploration reveals quantitative and qualitative differences in response to influenza and pneumococcal vaccines. Immunity. 2013;38:831–44.PubMedPubMedCentralCrossRef Obermoser G, et al. Systems scale interactive exploration reveals quantitative and qualitative differences in response to influenza and pneumococcal vaccines. Immunity. 2013;38:831–44.PubMedPubMedCentralCrossRef
214.
go back to reference M Abend et al. Examining Radiation-Induced In Vivo and In Vitro Gene Expression Changes of the Peripheral Blood in Different Laboratories for Biodosimetry Purposes: First RENEB Gene Expression Study. Radiat Res. 2016;185:109–23.PubMedCrossRef M Abend et al. Examining Radiation-Induced In Vivo and In Vitro Gene Expression Changes of the Peripheral Blood in Different Laboratories for Biodosimetry Purposes: First RENEB Gene Expression Study. Radiat Res. 2016;185:109–23.PubMedCrossRef
215.
go back to reference MC Panelli et al. Gene-expression profiling of the response of peripheral blood mononuclear cells and melanoma metastases to systemic IL-2 administration. Genome Biol. 2002;3:RESEARCH0035.PubMedPubMedCentralCrossRef MC Panelli et al. Gene-expression profiling of the response of peripheral blood mononuclear cells and melanoma metastases to systemic IL-2 administration. Genome Biol. 2002;3:RESEARCH0035.PubMedPubMedCentralCrossRef
216.
Metadata
Title
Emerging dynamics pathways of response and resistance to PD-1 and CTLA-4 blockade: tackling uncertainty by confronting complexity
Authors
Allan Relecom
Maysaloun Merhi
Varghese Inchakalody
Shahab Uddin
Darawan Rinchai
Davide Bedognetti
Said Dermime
Publication date
01-12-2021
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2021
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-021-01872-3

Other articles of this Issue 1/2021

Journal of Experimental & Clinical Cancer Research 1/2021 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine