Skip to main content
Top
Published in: Journal of Hematology & Oncology 1/2018

Open Access 01-12-2018 | Review

Next generation of immune checkpoint therapy in cancer: new developments and challenges

Authors: Julian A. Marin-Acevedo, Bhagirathbhai Dholaria, Aixa E. Soyano, Keith L. Knutson, Saranya Chumsri, Yanyan Lou

Published in: Journal of Hematology & Oncology | Issue 1/2018

Login to get access

Abstract

Immune checkpoints consist of inhibitory and stimulatory pathways that maintain self-tolerance and assist with immune response. In cancer, immune checkpoint pathways are often activated to inhibit the nascent anti-tumor immune response. Immune checkpoint therapies act by blocking or stimulating these pathways and enhance the body’s immunological activity against tumors. Cytotoxic T lymphocyte-associated molecule-4 (CTLA-4), programmed cell death receptor-1 (PD-1), and programmed cell death ligand-1(PD-L1) are the most widely studied and recognized inhibitory checkpoint pathways. Drugs blocking these pathways are currently utilized for a wide variety of malignancies and have demonstrated durable clinical activities in a subset of cancer patients. This approach is rapidly extending beyond CTLA-4 and PD-1/PD-L1. New inhibitory pathways are under investigation, and drugs blocking LAG-3, TIM-3, TIGIT, VISTA, or B7/H3 are being investigated. Furthermore, agonists of stimulatory checkpoint pathways such as OX40, ICOS, GITR, 4-1BB, CD40, or molecules targeting tumor microenvironment components like IDO or TLR are under investigation. In this article, we have provided a comprehensive review of immune checkpoint pathways involved in cancer immunotherapy, and discuss their mechanisms and the therapeutic interventions currently under investigation in phase I/II clinical trials. We also reviewed the limitations, toxicities, and challenges and outline the possible future research directions.
Literature
2.
go back to reference Drake CG, Lipson EJ, Brahmer JR. Breathing new life into immunotherapy: review of melanoma, lung and kidney cancer. Nat Rev Clin Oncol. 2014;11:24–37.PubMedCrossRef Drake CG, Lipson EJ, Brahmer JR. Breathing new life into immunotherapy: review of melanoma, lung and kidney cancer. Nat Rev Clin Oncol. 2014;11:24–37.PubMedCrossRef
5.
go back to reference Sharma P, Allison JP. Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell. 2015;161:205–14.PubMedCrossRef Sharma P, Allison JP. Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell. 2015;161:205–14.PubMedCrossRef
7.
go back to reference Granier 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 Granier 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
8.
go back to reference Marin-Acevedo JA, Soyano AE, Dholaria B, et al. Cancer immunotherapy beyond immune checkpoint inhibitors. J Hematol Oncol. 2018;11 Marin-Acevedo JA, Soyano AE, Dholaria B, et al. Cancer immunotherapy beyond immune checkpoint inhibitors. J Hematol Oncol. 2018;11
9.
go back to reference Workman CJ, Dugger KJ, Vignali DA. Cutting edge: molecular analysis of the negative regulatory function of lymphocyte activation gene-3. J Immunol. 2002;169:5392–5.PubMedCrossRef Workman CJ, Dugger KJ, Vignali DA. Cutting edge: molecular analysis of the negative regulatory function of lymphocyte activation gene-3. J Immunol. 2002;169:5392–5.PubMedCrossRef
11.
go back to reference Anderson 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.PubMedPubMedCentralCrossRef Anderson 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.PubMedPubMedCentralCrossRef
13.
go back to reference Woo 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.PubMedCrossRef Woo 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.PubMedCrossRef
16.
go back to reference Brignone C, Escudier B, Grygar C, et al. A phase I pharmacokinetic and biological correlative study of IMP321, a novel MHC class II agonist, in patients with advanced renal cell carcinoma. Clin Cancer Res. 2009;15:6225–31.PubMedCrossRef Brignone C, Escudier B, Grygar C, et al. A phase I pharmacokinetic and biological correlative study of IMP321, a novel MHC class II agonist, in patients with advanced renal cell carcinoma. Clin Cancer Res. 2009;15:6225–31.PubMedCrossRef
17.
go back to reference Wang-Gillam A, Plambeck-Suess S, Goedegebuure P, et al. A phase I study of IMP321 and gemcitabine as the front-line therapy in patients with advanced pancreatic adenocarcinoma. Investig New Drugs. 2013;31:707–13.CrossRef Wang-Gillam A, Plambeck-Suess S, Goedegebuure P, et al. A phase I study of IMP321 and gemcitabine as the front-line therapy in patients with advanced pancreatic adenocarcinoma. Investig New Drugs. 2013;31:707–13.CrossRef
18.
go back to reference Brignone C, Gutierrez M, Mefti F, et al. First-line chemoimmunotherapy in metastatic breast carcinoma: combination of paclitaxel and IMP321 (LAG-3Ig) enhances immune responses and antitumor activity. J Transl Med. 2010;8:71.PubMedPubMedCentralCrossRef Brignone C, Gutierrez M, Mefti F, et al. First-line chemoimmunotherapy in metastatic breast carcinoma: combination of paclitaxel and IMP321 (LAG-3Ig) enhances immune responses and antitumor activity. J Transl Med. 2010;8:71.PubMedPubMedCentralCrossRef
19.
go back to reference Nguyen LT, Ohashi PS. Clinical blockade of PD1 and LAG3—potential mechanisms of action. Nat Rev Immunol. 2015;15:45–56.PubMedCrossRef Nguyen LT, Ohashi PS. Clinical blockade of PD1 and LAG3—potential mechanisms of action. Nat Rev Immunol. 2015;15:45–56.PubMedCrossRef
20.
go back to reference Ascierto PA, 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: 9520–9520. Ascierto PA, 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: 9520–9520.
21.
go back to reference Sakuishi K, Ngiow SF, Sullivan JM, et al. TIM3+FOXP3+ regulatory T cells are tissue-specific promoters of T-cell dysfunction in cancer. Oncoimmunology. 2013;2:e23849.PubMedPubMedCentralCrossRef Sakuishi K, Ngiow SF, Sullivan JM, et al. TIM3+FOXP3+ regulatory T cells are tissue-specific promoters of T-cell dysfunction in cancer. Oncoimmunology. 2013;2:e23849.PubMedPubMedCentralCrossRef
22.
go back to reference Du W, Yang M, Turner A, et al. TIM-3 as a Target for Cancer Immunotherapy and Mechanisms of Action. Int J Mol Sci. 2017;18:645.PubMedCentralCrossRef Du W, Yang M, Turner A, et al. TIM-3 as a Target for Cancer Immunotherapy and Mechanisms of Action. Int J Mol Sci. 2017;18:645.PubMedCentralCrossRef
23.
go back to reference Gorman JV, Starbeck-Miller G, Pham NL, et al. Tim-3 directly enhances CD8 T cell responses to acute Listeria monocytogenes infection. J Immunol. 2014;192:3133–42.PubMedPubMedCentralCrossRef Gorman JV, Starbeck-Miller G, Pham NL, et al. Tim-3 directly enhances CD8 T cell responses to acute Listeria monocytogenes infection. J Immunol. 2014;192:3133–42.PubMedPubMedCentralCrossRef
24.
go back to reference Ebrahim AH, Alalawi Z, Mirandola L, et al. Galectins in cancer: carcinogenesis, diagnosis and therapy. Ann Transl Med. 2014;2:88.PubMedPubMedCentral Ebrahim AH, Alalawi Z, Mirandola L, et al. Galectins in cancer: carcinogenesis, diagnosis and therapy. Ann Transl Med. 2014;2:88.PubMedPubMedCentral
25.
go back to reference Fiori V, Magnani M, Cianfriglia M. The expression and modulation of CEACAM1 and tumor cell transformation. Ann Ist Super Sanita. 2012;48:161–71.PubMedCrossRef Fiori V, Magnani M, Cianfriglia M. The expression and modulation of CEACAM1 and tumor cell transformation. Ann Ist Super Sanita. 2012;48:161–71.PubMedCrossRef
26.
go back to reference Ohue Y, Kurose K, Nishio Y et al. Abstract A101: role of TIM-3/Galectin-9 pathway in lung cancer. Cancer Immunol Res 2016; 4: A101-A101. Ohue Y, Kurose K, Nishio Y et al. Abstract A101: role of TIM-3/Galectin-9 pathway in lung cancer. Cancer Immunol Res 2016; 4: A101-A101.
27.
go back to reference Zhu C, Anderson AC, Schubart A, et al. The Tim-3 ligand galectin-9 negatively regulates T helper type 1 immunity. Nat Immunol. 2005;6:1245–52.PubMedCrossRef Zhu C, Anderson AC, Schubart A, et al. The Tim-3 ligand galectin-9 negatively regulates T helper type 1 immunity. Nat Immunol. 2005;6:1245–52.PubMedCrossRef
28.
go back to reference Yu X, Harden K, Gonzalez LC, et al. The surface protein TIGIT suppresses T cell activation by promoting the generation of mature immunoregulatory dendritic cells. Nat Immunol. 2009;10:48–57.PubMedCrossRef Yu X, Harden K, Gonzalez LC, et al. The surface protein TIGIT suppresses T cell activation by promoting the generation of mature immunoregulatory dendritic cells. Nat Immunol. 2009;10:48–57.PubMedCrossRef
29.
go back to reference Stanietsky N, Simic H, Arapovic J, et al. The interaction of TIGIT with PVR and PVRL2 inhibits human NK cell cytotoxicity. Proc Natl Acad Sci U S A. 2009;106:17858–63.PubMedPubMedCentralCrossRef Stanietsky N, Simic H, Arapovic J, et al. The interaction of TIGIT with PVR and PVRL2 inhibits human NK cell cytotoxicity. Proc Natl Acad Sci U S A. 2009;106:17858–63.PubMedPubMedCentralCrossRef
30.
go back to reference Casado JG, Pawelec G, Morgado S, et al. Expression of adhesion molecules and ligands for activating and costimulatory receptors involved in cell-mediated cytotoxicity in a large panel of human melanoma cell lines. Cancer Immunol Immunother. 2009;58:1517–26.PubMedCrossRef Casado JG, Pawelec G, Morgado S, et al. Expression of adhesion molecules and ligands for activating and costimulatory receptors involved in cell-mediated cytotoxicity in a large panel of human melanoma cell lines. Cancer Immunol Immunother. 2009;58:1517–26.PubMedCrossRef
31.
32.
go back to reference Chauvin 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.PubMedPubMedCentralCrossRef Chauvin 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.PubMedPubMedCentralCrossRef
34.
go back to reference Le Mercier I, Chen W, Lines JL, et al. VISTA regulates the development of protective antitumor immunity. Cancer Res. 2014;74:1933–44.PubMedCrossRef Le Mercier I, Chen W, Lines JL, et al. VISTA regulates the development of protective antitumor immunity. Cancer Res. 2014;74:1933–44.PubMedCrossRef
35.
go back to reference Lines JL, Sempere LF, Broughton T, et al. VISTA is a novel broad-spectrum negative checkpoint regulator for cancer immunotherapy. Cancer Immunol Res. 2014;2:510–7.PubMedPubMedCentralCrossRef Lines JL, Sempere LF, Broughton T, et al. VISTA is a novel broad-spectrum negative checkpoint regulator for cancer immunotherapy. Cancer Immunol Res. 2014;2:510–7.PubMedPubMedCentralCrossRef
36.
37.
go back to reference Castellanos JR, Purvis IJ, Labak CM, et al. B7-H3 role in the immune landscape of cancer. American Journal of Clinical and Experimental Immunology. 2017;6:66–75.PubMedPubMedCentral Castellanos JR, Purvis IJ, Labak CM, et al. B7-H3 role in the immune landscape of cancer. American Journal of Clinical and Experimental Immunology. 2017;6:66–75.PubMedPubMedCentral
38.
go back to reference Powderly J, Cote G, Flaherty K et al. Interim results of an ongoing phase I, dose escalation study of MGA271 (Fc-optimized humanized anti-B7-H3 monoclonal antibody) in patients with refractory B7-H3-expressing neoplasms or neoplasms whose vasculature expresses B7-H3. J Immunotherapy Cancer 2015; 3: O8-O8. Powderly J, Cote G, Flaherty K et al. Interim results of an ongoing phase I, dose escalation study of MGA271 (Fc-optimized humanized anti-B7-H3 monoclonal antibody) in patients with refractory B7-H3-expressing neoplasms or neoplasms whose vasculature expresses B7-H3. J Immunotherapy Cancer 2015; 3: O8-O8.
39.
go back to reference Weidle UH, Kontermann RE, Brinkmann U. Tumor-antigen-binding bispecific antibodies for cancer treatment. Semin Oncol. 2014;41:653–60.PubMedCrossRef Weidle UH, Kontermann RE, Brinkmann U. Tumor-antigen-binding bispecific antibodies for cancer treatment. Semin Oncol. 2014;41:653–60.PubMedCrossRef
40.
go back to reference Tolcher AW, Alley EW, Chichili G et al. Phase 1, first-in-human, open label, dose escalation ctudy of MGD009, a humanized B7-H3 x CD3 dual-affinity re-targeting (DART) protein in patients with B7-H3-expressing neoplasms or B7-H3 expressing tumor vasculature. J Clin Oncol 2016; 34: TPS3105-TPS3105. Tolcher AW, Alley EW, Chichili G et al. Phase 1, first-in-human, open label, dose escalation ctudy of MGD009, a humanized B7-H3 x CD3 dual-affinity re-targeting (DART) protein in patients with B7-H3-expressing neoplasms or B7-H3 expressing tumor vasculature. J Clin Oncol 2016; 34: TPS3105-TPS3105.
41.
go back to reference Kramer K, Kushner BH, Modak S, et al. Compartmental intrathecal radioimmunotherapy: results for treatment for metastatic CNS neuroblastoma. J Neuro-Oncol. 2010;97:409–18.CrossRef Kramer K, Kushner BH, Modak S, et al. Compartmental intrathecal radioimmunotherapy: results for treatment for metastatic CNS neuroblastoma. J Neuro-Oncol. 2010;97:409–18.CrossRef
42.
go back to reference Leone RD, Lo YC, Powell JD. A2aR antagonists: next generation checkpoint blockade for cancer immunotherapy. Comput Struct Biotechnol J. 2015;13:265–72.PubMedPubMedCentralCrossRef Leone RD, Lo YC, Powell JD. A2aR antagonists: next generation checkpoint blockade for cancer immunotherapy. Comput Struct Biotechnol J. 2015;13:265–72.PubMedPubMedCentralCrossRef
44.
go back to reference Vijayan D, Young A, Teng MWL, Smyth MJ. Targeting immunosuppressive adenosine in cancer (vol 17, pg 709, 2017). Nat Rev Cancer 2017; 17: 724–724. Vijayan D, Young A, Teng MWL, Smyth MJ. Targeting immunosuppressive adenosine in cancer (vol 17, pg 709, 2017). Nat Rev Cancer 2017; 17: 724–724.
45.
go back to reference Emens L, Powderly J, Fong L, et al. Abstract CT119: CPI-444, an oral adenosine A2a receptor (A2aR) antagonist, demonstrates clinical activity in patients with advanced solid tumors. Cancer Res. 2017;77:CT119. Emens L, Powderly J, Fong L, et al. Abstract CT119: CPI-444, an oral adenosine A2a receptor (A2aR) antagonist, demonstrates clinical activity in patients with advanced solid tumors. Cancer Res. 2017;77:CT119.
46.
go back to reference Antonioli L, Novitskiy SV, Sachsenmeier KF, et al. Switching off CD73: a way to boost the activity of conventional and targeted antineoplastic therapies. Drug Discov Today. 2017;22:1686–96.PubMedCrossRef Antonioli L, Novitskiy SV, Sachsenmeier KF, et al. Switching off CD73: a way to boost the activity of conventional and targeted antineoplastic therapies. Drug Discov Today. 2017;22:1686–96.PubMedCrossRef
47.
go back to reference Paulos CM, June CH. Putting the brakes on BTLA in T cell-mediated cancer immunotherapy. J Clin Invest. 2010;120:76–80.PubMedCrossRef Paulos CM, June CH. Putting the brakes on BTLA in T cell-mediated cancer immunotherapy. J Clin Invest. 2010;120:76–80.PubMedCrossRef
48.
go back to reference Malissen N, Macagno N, Granjeaud S et al. HVEM: a novel cosignaling molecule of major interest in melanoma. J Clin Oncol 2017; 35: e14591-e14591. Malissen N, Macagno N, Granjeaud S et al. HVEM: a novel cosignaling molecule of major interest in melanoma. J Clin Oncol 2017; 35: e14591-e14591.
49.
go back to reference Lan X, Li S, Gao H, et al. Increased BTLA and HVEM in gastric cancer are associated with progression and poor prognosis. Onco Targets Ther. 2017;10:919–26.PubMedPubMedCentralCrossRef Lan X, Li S, Gao H, et al. Increased BTLA and HVEM in gastric cancer are associated with progression and poor prognosis. Onco Targets Ther. 2017;10:919–26.PubMedPubMedCentralCrossRef
51.
go back to reference Wendt MK, Tian MZ, Schiemann WP. Deconstructing the mechanisms and consequences of TGF-beta-induced EMT during cancer progression. Cell Tissue Res. 2012;347:85–101.PubMedCrossRef Wendt MK, Tian MZ, Schiemann WP. Deconstructing the mechanisms and consequences of TGF-beta-induced EMT during cancer progression. Cell Tissue Res. 2012;347:85–101.PubMedCrossRef
52.
go back to reference Thomas DA, Massagué J. TGF-β directly targets cytotoxic T cell functions during tumor evasion of immune surveillance. Cancer Cell. 2005;8:369–80.PubMedCrossRef Thomas DA, Massagué J. TGF-β directly targets cytotoxic T cell functions during tumor evasion of immune surveillance. Cancer Cell. 2005;8:369–80.PubMedCrossRef
54.
go back to reference Neuzillet C, Tijeras-Raballand A, Cohen R, et al. Targeting the TGFbeta pathway for cancer therapy. Pharmacol Ther. 2015;147:22–31.PubMedCrossRef Neuzillet C, Tijeras-Raballand A, Cohen R, et al. Targeting the TGFbeta pathway for cancer therapy. Pharmacol Ther. 2015;147:22–31.PubMedCrossRef
55.
go back to reference Smith AL, Robin TP, Ford HL. Molecular pathways: targeting the TGF-beta pathway for cancer therapy. Clin Cancer Res. 2012;18:4514–21.PubMedCrossRef Smith AL, Robin TP, Ford HL. Molecular pathways: targeting the TGF-beta pathway for cancer therapy. Clin Cancer Res. 2012;18:4514–21.PubMedCrossRef
56.
go back to reference Bogdahn U, Hau P, Stockhammer G, et al. Targeted therapy for high-grade glioma with the TGF-beta2 inhibitor trabedersen: results of a randomized and controlled phase IIb study. Neuro-Oncology. 2011;13:132–42.PubMedCrossRef Bogdahn U, Hau P, Stockhammer G, et al. Targeted therapy for high-grade glioma with the TGF-beta2 inhibitor trabedersen: results of a randomized and controlled phase IIb study. Neuro-Oncology. 2011;13:132–42.PubMedCrossRef
57.
go back to reference Hwang L, Ng K, Wang W, Trieu VN. OT-101: an anti-TGF-beta-2 antisense-primed tumors to subsequent chemotherapies. J Clin Oncol 2016; 34: e15727-e15727. Hwang L, Ng K, Wang W, Trieu VN. OT-101: an anti-TGF-beta-2 antisense-primed tumors to subsequent chemotherapies. J Clin Oncol 2016; 34: e15727-e15727.
58.
go back to reference Gulley JL, Heery CR, Schlom J et al. Preliminary results from a phase 1 trial of M7824 (MSB0011359C), a bifunctional fusion protein targeting PD-L1 and TGF-β, in advanced solid tumors. J Clin Oncol 2017; 35: 3006–3006. Gulley JL, Heery CR, Schlom J et al. Preliminary results from a phase 1 trial of M7824 (MSB0011359C), a bifunctional fusion protein targeting PD-L1 and TGF-β, in advanced solid tumors. J Clin Oncol 2017; 35: 3006–3006.
59.
go back to reference Brandes AA, Carpentier AF, Kesari S, et al. A phase II randomized study of galunisertib monotherapy or galunisertib plus lomustine compared with lomustine monotherapy in patients with recurrent glioblastoma. Neuro-Oncology. 2016;18:1146–56.PubMedPubMedCentralCrossRef Brandes AA, Carpentier AF, Kesari S, et al. A phase II randomized study of galunisertib monotherapy or galunisertib plus lomustine compared with lomustine monotherapy in patients with recurrent glioblastoma. Neuro-Oncology. 2016;18:1146–56.PubMedPubMedCentralCrossRef
60.
go back to reference Long EO, Barber DF, Burshtyn DN, et al. Inhibition of natural killer cell activation signals by killer cell immunoglobulin-like receptors (CD158). Immunol Rev. 2001;181:223–33.PubMedCrossRef Long EO, Barber DF, Burshtyn DN, et al. Inhibition of natural killer cell activation signals by killer cell immunoglobulin-like receptors (CD158). Immunol Rev. 2001;181:223–33.PubMedCrossRef
61.
go back to reference Dahlberg CI, Sarhan D, Chrobok M, et al. Natural killer cell-based therapies targeting cancer: possible strategies to gain and sustain anti-tumor activity. Front Immunol. 2015;6:605.PubMedPubMedCentralCrossRef Dahlberg CI, Sarhan D, Chrobok M, et al. Natural killer cell-based therapies targeting cancer: possible strategies to gain and sustain anti-tumor activity. Front Immunol. 2015;6:605.PubMedPubMedCentralCrossRef
62.
go back to reference Muntasell A, Ochoa MC, Cordeiro L, et al. Targeting NK-cell checkpoints for cancer immunotherapy. Curr Opin Immunol. 2017;45:73–81.PubMedCrossRef Muntasell A, Ochoa MC, Cordeiro L, et al. Targeting NK-cell checkpoints for cancer immunotherapy. Curr Opin Immunol. 2017;45:73–81.PubMedCrossRef
64.
go back to reference Leichner R, Kang H, Haddad R, et al. Preliminary efficacy from a phase I/II study of the natural killer cell–targeted antibody lirilumab in combination with nivolumab in squamous cell carcinoma of the head and neck. Journal for Immunotherapy of Cancer. 2016;4 Leichner R, Kang H, Haddad R, et al. Preliminary efficacy from a phase I/II study of the natural killer cell–targeted antibody lirilumab in combination with nivolumab in squamous cell carcinoma of the head and neck. Journal for Immunotherapy of Cancer. 2016;4
65.
go back to reference Schmitt C, Marie-Cardine A, Bensussan A. Therapeutic antibodies to KIR3DL2 and other target antigens on cutaneous T-cell lymphomas. Front Immunol. 2017;8 Schmitt C, Marie-Cardine A, Bensussan A. Therapeutic antibodies to KIR3DL2 and other target antigens on cutaneous T-cell lymphomas. Front Immunol. 2017;8
66.
go back to reference Bagot M, Porcu P, Ram-Wolff C, et al. Phase I study of IPH4102, anti-KIR3DL2 Mab, in relapsed/refractory cutaneous T-cell lymphomas (CTCL): dose-escalation safety, biomarker and clinical activity results. Hematol Oncol. 2017;35:48–9.CrossRef Bagot M, Porcu P, Ram-Wolff C, et al. Phase I study of IPH4102, anti-KIR3DL2 Mab, in relapsed/refractory cutaneous T-cell lymphomas (CTCL): dose-escalation safety, biomarker and clinical activity results. Hematol Oncol. 2017;35:48–9.CrossRef
67.
go back to reference Gyori D, Chessa T, Hawkins PT, Stephens LR. Class (I) phosphoinositide 3-kinases in the tumor microenvironment. Cancers. 2017;9:24.PubMedCentralCrossRef Gyori D, Chessa T, Hawkins PT, Stephens LR. Class (I) phosphoinositide 3-kinases in the tumor microenvironment. Cancers. 2017;9:24.PubMedCentralCrossRef
68.
go back to reference Tolcher A, Hong D, Sullivan R et al. Abstract CT089: IPI-549-01—a phase 1/1b, first-in-human study of IPI-549, a PI3K-γ inhibitor, as monotherapy and in combination with nivolumab in patients with advanced solid tumors. Cancer Res 2017; 77: CT089-CT089. Tolcher A, Hong D, Sullivan R et al. Abstract CT089: IPI-549-01—a phase 1/1b, first-in-human study of IPI-549, a PI3K-γ inhibitor, as monotherapy and in combination with nivolumab in patients with advanced solid tumors. Cancer Res 2017; 77: CT089-CT089.
69.
go back to reference Liu XJ, Kwon H, Li ZH, Fu YX. Is CD47 an innate immune checkpoint for tumor evasion? J Hematol Oncol. 2017;10 Liu XJ, Kwon H, Li ZH, Fu YX. Is CD47 an innate immune checkpoint for tumor evasion? J Hematol Oncol. 2017;10
70.
go back to reference Weiskopf K. Cancer immunotherapy targeting the CD47/SIRP alpha axis. Eur J Cancer. 2017;76:100–9.PubMedCrossRef Weiskopf K. Cancer immunotherapy targeting the CD47/SIRP alpha axis. Eur J Cancer. 2017;76:100–9.PubMedCrossRef
71.
go back to reference Sikic BI, Narayanan S, Colevas AD et al. A first-in-human, first-in-class phase I trial of the anti-CD47 antibody Hu5F9-G4 in patients with advanced cancers. J Clin Oncol 2016; 34: 3019–3019. Sikic BI, Narayanan S, Colevas AD et al. A first-in-human, first-in-class phase I trial of the anti-CD47 antibody Hu5F9-G4 in patients with advanced cancers. J Clin Oncol 2016; 34: 3019–3019.
72.
go back to reference Thompson JA, Akilov O, Querfeld C et al. A phase 1 dose-escalation trial of intratumoral TTI-621, a novel immune checkpoint inhibitor targeting CD47, in subjects with relapsed or refractory percutaneously-accessible solid tumors and mycosis fungoides. J Clin Oncol 2017; 35: TPS3101-TPS3101. Thompson JA, Akilov O, Querfeld C et al. A phase 1 dose-escalation trial of intratumoral TTI-621, a novel immune checkpoint inhibitor targeting CD47, in subjects with relapsed or refractory percutaneously-accessible solid tumors and mycosis fungoides. J Clin Oncol 2017; 35: TPS3101-TPS3101.
73.
go back to reference Willoughby J, Griffiths J, Tews I, Cragg MS. OX40: structure and function—what questions remain? Mol Immunol. 2017;83:13–22.PubMedCrossRef Willoughby J, Griffiths J, Tews I, Cragg MS. OX40: structure and function—what questions remain? Mol Immunol. 2017;83:13–22.PubMedCrossRef
74.
go back to reference Aspeslagh S, Postel-Vinay S, Rusakiewicz S, et al. Rationale for anti-OX40 cancer immunotherapy. Eur J Cancer. 2016;52:50–66.PubMedCrossRef Aspeslagh S, Postel-Vinay S, Rusakiewicz S, et al. Rationale for anti-OX40 cancer immunotherapy. Eur J Cancer. 2016;52:50–66.PubMedCrossRef
75.
go back to reference Linch SN, McNamara MJ, Redmond WL. OX40 agonists and combination immunotherapy: putting the pedal to the metal. Front Oncol. 2015;5 Linch SN, McNamara MJ, Redmond WL. OX40 agonists and combination immunotherapy: putting the pedal to the metal. Front Oncol. 2015;5
76.
go back to reference Turner JG, Rakhmilevich AL, Burdelya L, et al. Anti-CD40 antibody induces antitumor and antimetastatic effects: the role of NK cells. J Immunol. 2001;166:89–94.PubMedCrossRef Turner JG, Rakhmilevich AL, Burdelya L, et al. Anti-CD40 antibody induces antitumor and antimetastatic effects: the role of NK cells. J Immunol. 2001;166:89–94.PubMedCrossRef
77.
go back to reference Curti BD, Kovacsovics-Bankowski M, Morris N, et al. OX40 is a potent immune stimulating target in late stage cancer patients. Cancer Res. 2013;73:7189–98.PubMedPubMedCentralCrossRef Curti BD, Kovacsovics-Bankowski M, Morris N, et al. OX40 is a potent immune stimulating target in late stage cancer patients. Cancer Res. 2013;73:7189–98.PubMedPubMedCentralCrossRef
78.
go back to reference Infante JR, Hansen AR, Pishvaian MJ et al. A phase Ib dose escalation study of the OX40 agonist MOXR0916 and the PD-L1 inhibitor atezolizumab in patients with advanced solid tumors. J Clin Oncol 2016; 34: 101–101. Infante JR, Hansen AR, Pishvaian MJ et al. A phase Ib dose escalation study of the OX40 agonist MOXR0916 and the PD-L1 inhibitor atezolizumab in patients with advanced solid tumors. J Clin Oncol 2016; 34: 101–101.
79.
go back to reference Hamid O, Thompson JA, Diab A et al. First in human (FIH) study of an OX40 agonist monoclonal antibody (mAb) PF-04518600 (PF-8600) in adult patients (pts) with select advanced solid tumors: preliminary safety and pharmacokinetic (PK)/pharmacodynamic results. J Clin Oncol 2016; 34: 3079–3079. Hamid O, Thompson JA, Diab A et al. First in human (FIH) study of an OX40 agonist monoclonal antibody (mAb) PF-04518600 (PF-8600) in adult patients (pts) with select advanced solid tumors: preliminary safety and pharmacokinetic (PK)/pharmacodynamic results. J Clin Oncol 2016; 34: 3079–3079.
80.
go back to reference Dempke WCM, Fenchel K, Uciechowski P, Dale SP. Second- and third-generation drugs for immuno-oncology treatment-The more the better? Eur J Cancer. 2017;74:55–72.PubMedCrossRef Dempke WCM, Fenchel K, Uciechowski P, Dale SP. Second- and third-generation drugs for immuno-oncology treatment-The more the better? Eur J Cancer. 2017;74:55–72.PubMedCrossRef
81.
go back to reference Knee DA, Hewes B, Brogdon JL. Rationale for anti-GITR cancer immunotherapy. Eur J Cancer. 2016;67:1–10.PubMedCrossRef Knee DA, Hewes B, Brogdon JL. Rationale for anti-GITR cancer immunotherapy. Eur J Cancer. 2016;67:1–10.PubMedCrossRef
82.
go back to reference Koon HB, Shepard DR, Merghoub T et al. First-in-human phase 1 single-dose study of TRX-518, an anti-human glucocorticoid-induced tumor necrosis factor receptor (GITR) monoclonal antibody in adults with advanced solid tumors. J Clin Oncol 2016; 34: 3017–3017. Koon HB, Shepard DR, Merghoub T et al. First-in-human phase 1 single-dose study of TRX-518, an anti-human glucocorticoid-induced tumor necrosis factor receptor (GITR) monoclonal antibody in adults with advanced solid tumors. J Clin Oncol 2016; 34: 3017–3017.
83.
go back to reference Siu LL, Steeghs N, Meniawy T et al. Preliminary results of a phase I/IIa study of BMS-986156 (glucocorticoid-induced tumor necrosis factor receptor–related gene [GITR] agonist), alone and in combination with nivolumab in pts with advanced solid tumors. J Clin Oncol 2017; 35: 104–104. Siu LL, Steeghs N, Meniawy T et al. Preliminary results of a phase I/IIa study of BMS-986156 (glucocorticoid-induced tumor necrosis factor receptor–related gene [GITR] agonist), alone and in combination with nivolumab in pts with advanced solid tumors. J Clin Oncol 2017; 35: 104–104.
84.
go back to reference Tran B, Carvajal RD, Marabelle A et al. Dose escalation results from a first-in-human, phase 1 study of the glucocorticoid-induced TNF receptor-related protein (GITR) agonist AMG 228 in patients (pts) with advanced solid tumors. J Clin Oncol 2017; 35: 2521–2521. Tran B, Carvajal RD, Marabelle A et al. Dose escalation results from a first-in-human, phase 1 study of the glucocorticoid-induced TNF receptor-related protein (GITR) agonist AMG 228 in patients (pts) with advanced solid tumors. J Clin Oncol 2017; 35: 2521–2521.
85.
go back to reference Sanmamed MF, Pastor F, Rodriguez A, et al. Agonists of co-stimulation in cancer immunotherapy directed against CD137, OX40, GITR, CD27, CD28, and ICOS. Semin Oncol. 2015;42:640–55.PubMedCrossRef Sanmamed MF, Pastor F, Rodriguez A, et al. Agonists of co-stimulation in cancer immunotherapy directed against CD137, OX40, GITR, CD27, CD28, and ICOS. Semin Oncol. 2015;42:640–55.PubMedCrossRef
86.
go back to reference Fan XZ, Quezada SA, Sepulveda MA, et al. Engagement of the ICOS pathway markedly enhances efficacy of CTLA-4 blockade in cancer immunotherapy. J Exp Med. 2014;211:715–25.PubMedPubMedCentralCrossRef Fan XZ, Quezada SA, Sepulveda MA, et al. Engagement of the ICOS pathway markedly enhances efficacy of CTLA-4 blockade in cancer immunotherapy. J Exp Med. 2014;211:715–25.PubMedPubMedCentralCrossRef
87.
go back to reference Harvey C, Elpek K, Duong E, et al. Efficacy of anti-ICOS agonist monoclonal antibodies in preclinical tumor models provides a rationale for clinical development as cancer immunotherapeutics. Journal for ImmunoTherapy of Cancer. 2015;3:O9.PubMedCentralCrossRef Harvey C, Elpek K, Duong E, et al. Efficacy of anti-ICOS agonist monoclonal antibodies in preclinical tumor models provides a rationale for clinical development as cancer immunotherapeutics. Journal for ImmunoTherapy of Cancer. 2015;3:O9.PubMedCentralCrossRef
88.
go back to reference Burris HA, Callahan MK, Tolcher AW et al. Phase 1 safety of ICOS agonist antibody JTX-2011 alone and with nivolumab (nivo) in advanced solid tumors; predicted vs observed pharmacokinetics (PK) in ICONIC. J Clin Oncol 2017; 35: 3033–3033. Burris HA, Callahan MK, Tolcher AW et al. Phase 1 safety of ICOS agonist antibody JTX-2011 alone and with nivolumab (nivo) in advanced solid tumors; predicted vs observed pharmacokinetics (PK) in ICONIC. J Clin Oncol 2017; 35: 3033–3033.
89.
90.
go back to reference Takeda K, Kojima Y, Uno T, et al. Combination therapy of established tumors by antibodies targeting immune activating and suppressing molecules. J Immunol. 2010;184:5493–501.PubMedCrossRef Takeda K, Kojima Y, Uno T, et al. Combination therapy of established tumors by antibodies targeting immune activating and suppressing molecules. J Immunol. 2010;184:5493–501.PubMedCrossRef
91.
go back to reference Tolcher AW, Sznol M, Hu-Lieskovan S et al. Phase Ib study of PF-05082566 in combination with pembrolizumab in patients with advanced solid tumors. J Clin Oncol 2016; 34: 3002–3002. Tolcher AW, Sznol M, Hu-Lieskovan S et al. Phase Ib study of PF-05082566 in combination with pembrolizumab in patients with advanced solid tumors. J Clin Oncol 2016; 34: 3002–3002.
92.
go back to reference Segal NH, Logan TF, Hodi FS, et al. Results from an integrated safety analysis of urelumab, an agonist anti-CD137 monoclonal antibody. Clin Cancer Res. 2017;23:1929–36.PubMedCrossRef Segal NH, Logan TF, Hodi FS, et al. Results from an integrated safety analysis of urelumab, an agonist anti-CD137 monoclonal antibody. Clin Cancer Res. 2017;23:1929–36.PubMedCrossRef
93.
go back to reference Massarelli E, Segal N, Ribrag V. Clinical safety and efficacy assessment of the CD137 agonist urelumab alone and in combination with nivolumab in patients with hematologic and solid tumor malignancies. J Immunother Cancer. 2016;4:O7.CrossRef Massarelli E, Segal N, Ribrag V. Clinical safety and efficacy assessment of the CD137 agonist urelumab alone and in combination with nivolumab in patients with hematologic and solid tumor malignancies. J Immunother Cancer. 2016;4:O7.CrossRef
94.
go back to reference Perez-Ruiz E, Etxeberria I, Rodriguez-Ruiz ME, Melero I. Anti-CD137 and PD-1/PD-L1 antibodies en route toward clinical synergy. Clin Cancer Res. 2017;23:5326–8.PubMedCrossRef Perez-Ruiz E, Etxeberria I, Rodriguez-Ruiz ME, Melero I. Anti-CD137 and PD-1/PD-L1 antibodies en route toward clinical synergy. Clin Cancer Res. 2017;23:5326–8.PubMedCrossRef
95.
go back to reference Denoeud J, Moser M. Role of CD27/CD70 pathway of activation in immunity and tolerance. J Leukoc Biol. 2011;89:195–203.PubMedCrossRef Denoeud J, Moser M. Role of CD27/CD70 pathway of activation in immunity and tolerance. J Leukoc Biol. 2011;89:195–203.PubMedCrossRef
96.
go back to reference van de Ven K, Borst J. Targeting the T-cell co-stimulatory CD27/CD70 pathway in cancer immunotherapy: rationale and potential. Immunotherapy. 2015;7:655–67.PubMedCrossRef van de Ven K, Borst J. Targeting the T-cell co-stimulatory CD27/CD70 pathway in cancer immunotherapy: rationale and potential. Immunotherapy. 2015;7:655–67.PubMedCrossRef
97.
go back to reference Michot J-M, Maerevoet M, Aftimos PG et al. Clinical response observed in a phase I study in T cell lymphoma patients treated with anti-CD70 SIMPLE antibody ARGX-110. J Clin Oncol 2016; 34: 7556–7556. Michot J-M, Maerevoet M, Aftimos PG et al. Clinical response observed in a phase I study in T cell lymphoma patients treated with anti-CD70 SIMPLE antibody ARGX-110. J Clin Oncol 2016; 34: 7556–7556.
98.
go back to reference Owonikoko TK, Hussain A, Stadler WM, et al. First-in-human multicenter phase I study of BMS-936561 (MDX-1203), an antibody-drug conjugate targeting CD70. Cancer Chemother Pharmacol. 2016;77:155–62.PubMedCrossRef Owonikoko TK, Hussain A, Stadler WM, et al. First-in-human multicenter phase I study of BMS-936561 (MDX-1203), an antibody-drug conjugate targeting CD70. Cancer Chemother Pharmacol. 2016;77:155–62.PubMedCrossRef
99.
go back to reference Sanborn RE, Pishvain MJ, Callahan MK et al. Abstract CT023: phase I results from the combination of an immune-activating anti-CD27 antibody (varlilumab) in combination with PD-1 blockade (nivolumab): activation across multiple immune pathways without untoward immune-related adverse events. Cancer Res 2016; 76: CT023-CT023. Sanborn RE, Pishvain MJ, Callahan MK et al. Abstract CT023: phase I results from the combination of an immune-activating anti-CD27 antibody (varlilumab) in combination with PD-1 blockade (nivolumab): activation across multiple immune pathways without untoward immune-related adverse events. Cancer Res 2016; 76: CT023-CT023.
100.
go back to reference Vonderheide RH. Prospect of targeting the CD40 pathway for cancer therapy. Clin Cancer Res. 2007;13:1083–8.PubMedCrossRef Vonderheide RH. Prospect of targeting the CD40 pathway for cancer therapy. Clin Cancer Res. 2007;13:1083–8.PubMedCrossRef
102.
go back to reference Dempke WCM, Fenchel K, Uciechowski P, Dale SP. Second- and third-generation drugs for immuno-oncology treatment-the more the better? Eur J Cancer. 2017;74:55–72.PubMedCrossRef Dempke WCM, Fenchel K, Uciechowski P, Dale SP. Second- and third-generation drugs for immuno-oncology treatment-the more the better? Eur J Cancer. 2017;74:55–72.PubMedCrossRef
103.
go back to reference Cabo M, Offringa R, Zitvogel L, et al. Trial watch: immunostimulatory monoclonal antibodies for oncological indications. Oncoimmunology. 2017;6:e1371896.PubMedCrossRef Cabo M, Offringa R, Zitvogel L, et al. Trial watch: immunostimulatory monoclonal antibodies for oncological indications. Oncoimmunology. 2017;6:e1371896.PubMedCrossRef
105.
go back to reference Toulmonde M, Penel N, Adam J, et al. Use of pd-1 targeting, macrophage infiltration, and ido pathway activation in sarcomas: a phase 2 clinical trial. JAMA Oncology. 2017; Toulmonde M, Penel N, Adam J, et al. Use of pd-1 targeting, macrophage infiltration, and ido pathway activation in sarcomas: a phase 2 clinical trial. JAMA Oncology. 2017;
106.
go back to reference Bilir C, Sarisozen C. Indoleamine 2,3-dioxygenase (IDO): only an enzyme or a checkpoint controller? Journal of Oncological Sciences. 2017;3:52–6.CrossRef Bilir C, Sarisozen C. Indoleamine 2,3-dioxygenase (IDO): only an enzyme or a checkpoint controller? Journal of Oncological Sciences. 2017;3:52–6.CrossRef
107.
go back to reference Siu LL, Gelmon K, Chu Q, et al. Abstract CT116: BMS-986205, an optimized indoleamine 2,3-dioxygenase 1 (IDO1) inhibitor, is well tolerated with potent pharmacodynamic (PD) activity, alone and in combination with nivolumab (nivo) in advanced cancers in a phase 1/2a trial. Cancer Res. 2017;77:CT116.CrossRef Siu LL, Gelmon K, Chu Q, et al. Abstract CT116: BMS-986205, an optimized indoleamine 2,3-dioxygenase 1 (IDO1) inhibitor, is well tolerated with potent pharmacodynamic (PD) activity, alone and in combination with nivolumab (nivo) in advanced cancers in a phase 1/2a trial. Cancer Res. 2017;77:CT116.CrossRef
108.
go back to reference Zakharia Y, Drabick JJ, Khleif S et al. Updates on phase 1b/2 trial of the indoleamine 2,3-dioxygenase pathway (IDO) inhibitor indoximod plus checkpoint inhibitors for the treatment of unresectable stage 3 or 4 melanoma. J Clin Oncol 2016; 34: 3075–3075. Zakharia Y, Drabick JJ, Khleif S et al. Updates on phase 1b/2 trial of the indoleamine 2,3-dioxygenase pathway (IDO) inhibitor indoximod plus checkpoint inhibitors for the treatment of unresectable stage 3 or 4 melanoma. J Clin Oncol 2016; 34: 3075–3075.
109.
go back to reference Bahary N, Garrido-Laguna I, Cinar P et al. Phase 2 trial of the indoleamine 2,3-dioxygenase pathway (IDO) inhibitor indoximod plus gemcitabine/nab-paclitaxel for the treatment of metastatic pancreas cancer: interim analysis. J Clin Oncol 2016; 34: 3020–3020. Bahary N, Garrido-Laguna I, Cinar P et al. Phase 2 trial of the indoleamine 2,3-dioxygenase pathway (IDO) inhibitor indoximod plus gemcitabine/nab-paclitaxel for the treatment of metastatic pancreas cancer: interim analysis. J Clin Oncol 2016; 34: 3020–3020.
110.
go back to reference Jha GG, Gupta S, Tagawa ST et al. A phase II randomized, double-blind study of sipuleucel-T followed by IDO pathway inhibitor, indoximod, or placebo in the treatment of patients with metastatic castration resistant prostate cancer (mCRPC). J Clin Oncol 2017; 35: 3066–3066. Jha GG, Gupta S, Tagawa ST et al. A phase II randomized, double-blind study of sipuleucel-T followed by IDO pathway inhibitor, indoximod, or placebo in the treatment of patients with metastatic castration resistant prostate cancer (mCRPC). J Clin Oncol 2017; 35: 3066–3066.
111.
go back to reference Hamid O, Bauer TM, Spira AI et al. Safety of epacadostat 100 mg bid plus pembrolizumab 200 mg Q3W in advanced solid tumors: phase 2 data from ECHO-202/KEYNOTE-037. J Clin Oncol 2017; 35: 3012–3012. Hamid O, Bauer TM, Spira AI et al. Safety of epacadostat 100 mg bid plus pembrolizumab 200 mg Q3W in advanced solid tumors: phase 2 data from ECHO-202/KEYNOTE-037. J Clin Oncol 2017; 35: 3012–3012.
112.
go back to reference Perez RP, Riese MJ, Lewis KD et al. Epacadostat plus nivolumab in patients with advanced solid tumors: preliminary phase I/II results of ECHO-204. J Clin Oncol 2017; 35: 3003–3003. Perez RP, Riese MJ, Lewis KD et al. Epacadostat plus nivolumab in patients with advanced solid tumors: preliminary phase I/II results of ECHO-204. J Clin Oncol 2017; 35: 3003–3003.
113.
go back to reference Beatty GL, Dwyer PJ, Clark J, et al. First-in-human phase I study of the oral inhibitor of indoleamine 2,3-dioxygenase-1 epacadostat (INCB024360) in patients with advanced solid malignancies. Clin Cancer Res. 2017;23:3269.PubMedCrossRef Beatty GL, Dwyer PJ, Clark J, et al. First-in-human phase I study of the oral inhibitor of indoleamine 2,3-dioxygenase-1 epacadostat (INCB024360) in patients with advanced solid malignancies. Clin Cancer Res. 2017;23:3269.PubMedCrossRef
114.
go back to reference Lu H. TLR agonists for cancer immunotherapy: tipping the balance between the immune stimulatory and inhibitory effects. Front Immunol. 2014;5:83.PubMedPubMedCentral Lu H. TLR agonists for cancer immunotherapy: tipping the balance between the immune stimulatory and inhibitory effects. Front Immunol. 2014;5:83.PubMedPubMedCentral
115.
116.
go back to reference Dowling JK, Mansell A. Toll-like receptors: the swiss army knife of immunity and vaccine development. Clinical & Translational Immunology. 2016;5 Dowling JK, Mansell A. Toll-like receptors: the swiss army knife of immunity and vaccine development. Clinical & Translational Immunology. 2016;5
117.
go back to reference Li K, Qu S, Chen X, et al. Promising targets for cancer immunotherapy: TLRs, RLRs, and STING-mediated innate immune pathways. Int J Mol Sci. 2017;18:404.PubMedCentralCrossRef Li K, Qu S, Chen X, et al. Promising targets for cancer immunotherapy: TLRs, RLRs, and STING-mediated innate immune pathways. Int J Mol Sci. 2017;18:404.PubMedCentralCrossRef
118.
go back to reference Gupta S, Grilley-Olson J, Hong D, et al. Abstract CT091: Safety and pharmacodynamic activity of MEDI9197, a TLR 7/8 agonist, administered intratumorally in subjects with solid tumors. Cancer Res. 2017;77:CT091. Gupta S, Grilley-Olson J, Hong D, et al. Abstract CT091: Safety and pharmacodynamic activity of MEDI9197, a TLR 7/8 agonist, administered intratumorally in subjects with solid tumors. Cancer Res. 2017;77:CT091.
119.
go back to reference Dredge K, Brennan T, Brown MP et al. An open-label, multi-center phase I study of the safety and tolerability of the novel immunomodulatory agent PG545 in subjects with advanced solid tumors. J Clin Oncol 2017; 35: 3083–3083. Dredge K, Brennan T, Brown MP et al. An open-label, multi-center phase I study of the safety and tolerability of the novel immunomodulatory agent PG545 in subjects with advanced solid tumors. J Clin Oncol 2017; 35: 3083–3083.
120.
go back to reference de la Torre AN, Contractor S, Castaneda I, et al. A phase I trial using local regional treatment, nonlethal irradiation, intratumoral and systemic polyinosinic-polycytidylic acid polylysine carboxymethylcellulose to treat liver cancer: in search of the abscopal effect. Journal of Hepatocellular Carcinoma. 2017;4 de la Torre AN, Contractor S, Castaneda I, et al. A phase I trial using local regional treatment, nonlethal irradiation, intratumoral and systemic polyinosinic-polycytidylic acid polylysine carboxymethylcellulose to treat liver cancer: in search of the abscopal effect. Journal of Hepatocellular Carcinoma. 2017;4
121.
go back to reference Tomala J, Kovar M. IL-2/anti-IL-2 mAb immunocomplexes: a renascence of IL-2 in cancer immunotherapy? Oncoimmunology. 2016;5 Tomala J, Kovar M. IL-2/anti-IL-2 mAb immunocomplexes: a renascence of IL-2 in cancer immunotherapy? Oncoimmunology. 2016;5
123.
go back to reference Su EW, Moore CJ, Suriano S et al. IL-2Rα mediates temporal regulation of IL-2 signaling and enhances immunotherapy. Sci Transl Med 2015; 7: 311ra170-311ra170. Su EW, Moore CJ, Suriano S et al. IL-2Rα mediates temporal regulation of IL-2 signaling and enhances immunotherapy. Sci Transl Med 2015; 7: 311ra170-311ra170.
124.
go back to reference Diab A, Tannir NM, Bernatchez C et al. A phase 1/2 study of a novel IL-2 cytokine, NKTR-214, and nivolumab in patients with select locally advanced or metastatic solid tumors. J Clin Oncol 2017; 35: e14040-e14040. Diab A, Tannir NM, Bernatchez C et al. A phase 1/2 study of a novel IL-2 cytokine, NKTR-214, and nivolumab in patients with select locally advanced or metastatic solid tumors. J Clin Oncol 2017; 35: e14040-e14040.
125.
go back to reference Bernatchez C, Haymaker CL, Hurwitz ME et al. Effect of a novel IL-2 cytokine immune agonist (NKTR-214) on proliferating CD8+T cells and PD-1 expression on immune cells in the tumor microenvironment in patients with prior checkpoint therapy. J Clin Oncol 2017; 35: 2545–2545. Bernatchez C, Haymaker CL, Hurwitz ME et al. Effect of a novel IL-2 cytokine immune agonist (NKTR-214) on proliferating CD8+T cells and PD-1 expression on immune cells in the tumor microenvironment in patients with prior checkpoint therapy. J Clin Oncol 2017; 35: 2545–2545.
127.
go back to reference Timosenko E, Hadjinicolaou AV, Cerundolo V. Modulation of cancer-specific immune responses by amino acid degrading enzymes. Immunotherapy. 2017;9:83–97.PubMedCrossRef Timosenko E, Hadjinicolaou AV, Cerundolo V. Modulation of cancer-specific immune responses by amino acid degrading enzymes. Immunotherapy. 2017;9:83–97.PubMedCrossRef
128.
go back to reference Papadopoulos KP, Tsai FY-C, Bauer TM et al. CX-1158-101: a first-in-human phase 1 study of CB-1158, a small molecule inhibitor of arginase, as monotherapy and in combination with an anti-PD-1 checkpoint inhibitor in patients (pts) with solid tumors. J Clin Oncol 2017; 35: 3005–3005. Papadopoulos KP, Tsai FY-C, Bauer TM et al. CX-1158-101: a first-in-human phase 1 study of CB-1158, a small molecule inhibitor of arginase, as monotherapy and in combination with an anti-PD-1 checkpoint inhibitor in patients (pts) with solid tumors. J Clin Oncol 2017; 35: 3005–3005.
129.
go back to reference Zhou H, Forveille S, Sauvat A, et al. The oncolytic peptide LTX-315 triggers immunogenic cell death. Cell Death Dis. 2016;7 Zhou H, Forveille S, Sauvat A, et al. The oncolytic peptide LTX-315 triggers immunogenic cell death. Cell Death Dis. 2016;7
130.
go back to reference Sveinbjornsson B, Camilio KA, Haug BE, Rekdal O. LTX-315: a first-in-class oncolytic peptide that reprograms the tumor microenvironment. Future Med Chem. 2017;9:1339–44.PubMedCrossRef Sveinbjornsson B, Camilio KA, Haug BE, Rekdal O. LTX-315: a first-in-class oncolytic peptide that reprograms the tumor microenvironment. Future Med Chem. 2017;9:1339–44.PubMedCrossRef
131.
go back to reference Yamazaki T, Pitt JM, Vétizou M, et al. The oncolytic peptide LTX-315 overcomes resistance of cancers to immunotherapy with CTLA4 checkpoint blockade. Cell Death Differ. 2016;23:1004–15.PubMedPubMedCentralCrossRef Yamazaki T, Pitt JM, Vétizou M, et al. The oncolytic peptide LTX-315 overcomes resistance of cancers to immunotherapy with CTLA4 checkpoint blockade. Cell Death Differ. 2016;23:1004–15.PubMedPubMedCentralCrossRef
132.
go back to reference Spicer JF, Baurain J-F, Awada A et al. LTX-315, an oncolytic peptide, to convert immunogenically ‘cold’ tumors to ‘hot’ in patients with advanced or metastatic tumours: results from an ongoing phase I study. J Clin Oncol 2017; 35: 3085–3085. Spicer JF, Baurain J-F, Awada A et al. LTX-315, an oncolytic peptide, to convert immunogenically ‘cold’ tumors to ‘hot’ in patients with advanced or metastatic tumours: results from an ongoing phase I study. J Clin Oncol 2017; 35: 3085–3085.
133.
go back to reference Mittal SK, Cho KJ, Ishido S, Roche PA. Interleukin 10 (IL-10)-mediated immunosuppression: MARCH-I INDUCTION REGULATES ANTIGEN PRESENTATION BY MACROPHAGES BUT NOT DENDRITIC CELLS. J Biol Chem. 2015;290:27158–67.PubMedPubMedCentralCrossRef Mittal SK, Cho KJ, Ishido S, Roche PA. Interleukin 10 (IL-10)-mediated immunosuppression: MARCH-I INDUCTION REGULATES ANTIGEN PRESENTATION BY MACROPHAGES BUT NOT DENDRITIC CELLS. J Biol Chem. 2015;290:27158–67.PubMedPubMedCentralCrossRef
134.
go back to reference Miotto D, Lo Cascio N, Stendardo M, et al. CD8+ T cells expressing IL-10 are associated with a favourable prognosis in lung cancer. Lung Cancer. 2010;69:355–60.PubMedCrossRef Miotto D, Lo Cascio N, Stendardo M, et al. CD8+ T cells expressing IL-10 are associated with a favourable prognosis in lung cancer. Lung Cancer. 2010;69:355–60.PubMedCrossRef
135.
go back to reference Zhang H, Wang Y, Hwang ES, He YW. Interleukin-10: an immune-activating cytokine in cancer immunotherapy. J Clin Oncol. 2016;34:3576.PubMedCrossRef Zhang H, Wang Y, Hwang ES, He YW. Interleukin-10: an immune-activating cytokine in cancer immunotherapy. J Clin Oncol. 2016;34:3576.PubMedCrossRef
136.
137.
go back to reference Naing A, Wong DJL, Infante JR et al. PEGylated human IL-10 (AM0010) in combination with pembrolizumab in anti-PD1 and CTLA-4 refractory melanoma. J Clin Oncol 2017; 35: 3084–3084. Naing A, Wong DJL, Infante JR et al. PEGylated human IL-10 (AM0010) in combination with pembrolizumab in anti-PD1 and CTLA-4 refractory melanoma. J Clin Oncol 2017; 35: 3084–3084.
138.
go back to reference Naidoo J, Page DB, Li BT, et al. Toxicities of the anti-PD-1 and anti-PD-L1 immune checkpoint antibodies. Ann Oncol. 2015;26:2375–91.PubMed Naidoo J, Page DB, Li BT, et al. Toxicities of the anti-PD-1 and anti-PD-L1 immune checkpoint antibodies. Ann Oncol. 2015;26:2375–91.PubMed
139.
go back to reference Michot JM, Pruvost R, Mateus C, et al. Fever reaction and haemophagocytic syndrome induced by immune checkpoint inhibitors. Ann Oncol. 2018;29:518–20.PubMedCrossRef Michot JM, Pruvost R, Mateus C, et al. Fever reaction and haemophagocytic syndrome induced by immune checkpoint inhibitors. Ann Oncol. 2018;29:518–20.PubMedCrossRef
140.
go back to reference Picchi H, Mateus C, Chouaid C, et al. Infectious complications associated with the use of immune checkpoint inhibitors in oncology: reactivation of tuberculosis after anti PD-1 treatment. Clin Microbiol Infect. 2017; Picchi H, Mateus C, Chouaid C, et al. Infectious complications associated with the use of immune checkpoint inhibitors in oncology: reactivation of tuberculosis after anti PD-1 treatment. Clin Microbiol Infect. 2017;
141.
go back to reference Champiat S, Lambotte O, Barreau E, et al. Management of immune checkpoint blockade dysimmune toxicities: a collaborative position paper. Ann Oncol. 2016;27:559–74.PubMedCrossRef Champiat S, Lambotte O, Barreau E, et al. Management of immune checkpoint blockade dysimmune toxicities: a collaborative position paper. Ann Oncol. 2016;27:559–74.PubMedCrossRef
142.
go back to reference Dine J, Gordon R, Shames Y, et al. Immune checkpoint inhibitors: an innovation in immunotherapy for the treatment and management of patients with cancer. Asia-Pacific Journal of Oncology Nursing. 2017;4:127–35.PubMedPubMedCentralCrossRef Dine J, Gordon R, Shames Y, et al. Immune checkpoint inhibitors: an innovation in immunotherapy for the treatment and management of patients with cancer. Asia-Pacific Journal of Oncology Nursing. 2017;4:127–35.PubMedPubMedCentralCrossRef
143.
go back to reference Linardou H, Gogas H. Toxicity management of immunotherapy for patients with metastatic melanoma. Annals of Translational Medicine. 2016;4 Linardou H, Gogas H. Toxicity management of immunotherapy for patients with metastatic melanoma. Annals of Translational Medicine. 2016;4
144.
go back to reference Gupta A, De Felice KM, Loftus EV Jr, Khanna S. Systematic review: colitis associated with anti-CTLA-4 therapy. Aliment Pharmacol Ther. 2015;42:406–17. Gupta A, De Felice KM, Loftus EV Jr, Khanna S. Systematic review: colitis associated with anti-CTLA-4 therapy. Aliment Pharmacol Ther. 2015;42:406–17.
145.
146.
go back to reference Zaretsky 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.PubMedPubMedCentralCrossRef Zaretsky 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.PubMedPubMedCentralCrossRef
148.
go back to reference Taube 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.CrossRef Taube 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.CrossRef
149.
150.
go back to reference Lou Y, Diao L, Cuentas ER, et al. Epithelial-mesenchymal transition is associated with a distinct tumor microenvironment including elevation of inflammatory signals and multiple immune checkpoints in lung adenocarcinoma. Clin Cancer Res. 2016;22:3630–42.PubMedPubMedCentralCrossRef Lou Y, Diao L, Cuentas ER, et al. Epithelial-mesenchymal transition is associated with a distinct tumor microenvironment including elevation of inflammatory signals and multiple immune checkpoints in lung adenocarcinoma. Clin Cancer Res. 2016;22:3630–42.PubMedPubMedCentralCrossRef
151.
go back to reference Miao D, Margolis CA, Gao W, et al. Genomic correlates of response to immune checkpoint therapies in clear cell renal cell carcinoma. Science. 2018;359:801–6.PubMedCrossRef Miao D, Margolis CA, Gao W, et al. Genomic correlates of response to immune checkpoint therapies in clear cell renal cell carcinoma. Science. 2018;359:801–6.PubMedCrossRef
152.
go back to reference Luksza M, Riaz N, Makarov V, et al. A neoantigen fitness model predicts tumour response to checkpoint blockade immunotherapy. Nature. 2017;551:517.PubMed Luksza M, Riaz N, Makarov V, et al. A neoantigen fitness model predicts tumour response to checkpoint blockade immunotherapy. Nature. 2017;551:517.PubMed
Metadata
Title
Next generation of immune checkpoint therapy in cancer: new developments and challenges
Authors
Julian A. Marin-Acevedo
Bhagirathbhai Dholaria
Aixa E. Soyano
Keith L. Knutson
Saranya Chumsri
Yanyan Lou
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Journal of Hematology & Oncology / Issue 1/2018
Electronic ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-018-0582-8

Other articles of this Issue 1/2018

Journal of Hematology & Oncology 1/2018 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