101
|
Yogarajah T, Ong KC, Perera D, Wong KT. AIM2 Inflammasome-Mediated Pyroptosis in Enterovirus A71-Infected Neuronal Cells Restricts Viral Replication. Sci Rep 2017; 7:5845. [PMID: 28724943 PMCID: PMC5517550 DOI: 10.1038/s41598-017-05589-2] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 05/31/2017] [Indexed: 01/17/2023] Open
Abstract
Encephalomyelitis is a well-known complication of hand, foot, and mouth disease (HFMD) due to Enterovirus 71 (EV71) infection. Viral RNA/antigens could be detected in the central nervous system (CNS) neurons in fatal encephalomyelitis but the mechanisms of neuronal cell death is not clearly understood. We investigated the role of absent in melanoma 2 (AIM2) inflammasome in neuronal cell death, and its relationship to viral replication. Our transcriptomic analysis, RT-qPCR, Western blot, immunofluorescence and flow cytometry studies consistently showed AIM2 gene up-regulation and protein expression in EV-A71-infected SK-N-SH cells. Downstream AIM2-induced genes, CARD16, caspase-1 and IL-1β were also up-regulated and caspase-1 was activated to form cleaved caspase-1 p20 subunits. As evidenced by 7-AAD positivity, pyroptosis was confirmed in infected cells. Overall, these findings have a strong correlation with decreases in viral titers, copy numbers and proteins, and reduced proportions of infected cells. AIM2 and viral antigens were detected by immunohistochemistry in infected neurons in inflamed areas of the CNS in EV-A71 encephalomyelitis. In infected AIM2-knockdown cells, AIM2 and related downstream gene expressions, and pyroptosis were suppressed, resulting in significantly increased virus infection. These results support the notion that AIM2 inflammasome-mediated pyroptosis is an important mechanism of neuronal cell death and it could play an important role in limiting EV-A71 replication.
Collapse
Affiliation(s)
- Thinesshwary Yogarajah
- Department of Pathology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Kien Chai Ong
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - David Perera
- Institute of Health and Community Medicine, University Malaysia Sarawak, Sarawak, Malaysia
| | - Kum Thong Wong
- Department of Pathology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.
| |
Collapse
|
102
|
Post-chemotherapy PD-L1 expression correlates with clinical outcomes in Japanese bladder cancer patients treated with total cystectomy. Med Oncol 2017; 34:117. [DOI: 10.1007/s12032-017-0977-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Accepted: 05/09/2017] [Indexed: 10/19/2022]
|
103
|
Kamphorst AO, Wieland A, Nasti T, Yang S, Zhang R, Barber DL, Konieczny BT, Daugherty CZ, Koenig L, Yu K, Sica GL, Sharpe AH, Freeman GJ, Blazar BR, Turka LA, Owonikoko TK, Pillai RN, Ramalingam SS, Araki K, Ahmed R. Rescue of exhausted CD8 T cells by PD-1-targeted therapies is CD28-dependent. Science 2017; 355:1423-1427. [PMID: 28280249 PMCID: PMC5595217 DOI: 10.1126/science.aaf0683] [Citation(s) in RCA: 708] [Impact Index Per Article: 101.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 11/09/2016] [Accepted: 01/06/2017] [Indexed: 12/12/2022]
Abstract
Programmed cell death-1 (PD-1)-targeted therapies enhance T cell responses and show efficacy in multiple cancers, but the role of costimulatory molecules in this T cell rescue remains elusive. Here, we demonstrate that the CD28/B7 costimulatory pathway is essential for effective PD-1 therapy during chronic viral infection. Conditional gene deletion showed a cell-intrinsic requirement of CD28 for CD8 T cell proliferation after PD-1 blockade. B7-costimulation was also necessary for effective PD-1 therapy in tumor-bearing mice. In addition, we found that CD8 T cells proliferating in blood after PD-1 therapy of lung cancer patients were predominantly CD28-positive. Taken together, these data demonstrate CD28-costimulation requirement for CD8 T cell rescue and suggest an important role for the CD28/B7 pathway in PD-1 therapy of cancer patients.
Collapse
Affiliation(s)
- Alice O Kamphorst
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Andreas Wieland
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Tahseen Nasti
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Shu Yang
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA
- Xiangya School of Medicine, Central South University, Changsha, Hunan Province, China, 410013
| | - Ruan Zhang
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02144, USA
| | - Daniel L Barber
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA
| | - Bogumila T Konieczny
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Candace Z Daugherty
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Lydia Koenig
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ke Yu
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Gabriel L Sica
- Department of Pathology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Arlene H Sharpe
- Department of Microbiology and Immunobiology and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Woman's Hospital, Boston, MA 02115, USA
| | - Gordon J Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Bruce R Blazar
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN 55455, USA
| | - Laurence A Turka
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02144, USA
| | - Taofeek K Owonikoko
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Rathi N Pillai
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Suresh S Ramalingam
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Koichi Araki
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Rafi Ahmed
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA.
| |
Collapse
|
104
|
Zhong F, Cheng X, Sun S, Zhou J. Transcriptional activation of PD-L1 by Sox2 contributes to the proliferation of hepatocellular carcinoma cells. Oncol Rep 2017; 37:3061-3067. [PMID: 28339084 DOI: 10.3892/or.2017.5523] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 03/03/2017] [Indexed: 11/06/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common and lethal malignancies in the world. Sox2 is a potential oncogene in the pathogenesis of HCC, however, the actual mechanisms of Sox2 functions in HCC has not emerged yet. In this study, we explored the expression, function and the relationship between Sox2 and PD-L1 in HCC. We found that both Sox2 and PD-L1 were expressed at a markedly higher level in HCC tissues in comparison to adjacent non-tumor tissues. Moreover, the expression levels of both genes were correlated with each other. Knockdown of Sox2 reduced the cell proliferation ability and induces apoptosis of HCC cells, suggesting the function of Sox2 in regulating both the cell proliferation and apoptosis. Noteworthy, the depletion of Sox2 also reduced the expression of PD-L1. Further analysis showed that there is a consensus Sox2 binding site in the promoter region of PD-L1. Through in vitro EMSA assay and in vivo chromatin immunoprecipitation assays, we demonstrated that Sox2 directly bound to the PD-L1 promoter through the consensus Sox2 motif. Further evidence by luciferase reporter assays revealed that Sox2 promoted the transcription activity of PD-L1 promoter region through the Sox2 motif. Collectively, our data provide a novel insight into the function and the interplay of Sox2 and PD-L1 in HCC.
Collapse
Affiliation(s)
- Feng Zhong
- Department of Hepatobiliary Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong 518100, P.R. China
| | - Xinsheng Cheng
- Department of Hepatobiliary Surgery, Nanshan Hospital, Guangdong Medical College, Shenzhen, Guangdong 518052, P.R. China
| | - Shibo Sun
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jie Zhou
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
105
|
Rittmeyer A, Barlesi F, Waterkamp D, Park K, Ciardiello F, von Pawel J, Gadgeel SM, Hida T, Kowalski DM, Dols MC, Cortinovis DL, Leach J, Polikoff J, Barrios C, Kabbinavar F, Frontera OA, De Marinis F, Turna H, Lee JS, Ballinger M, Kowanetz M, He P, Chen DS, Sandler A, Gandara DR. Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial. LANCET (LONDON, ENGLAND) 2017. [PMID: 27979383 DOI: 10.1016/s0140-6736(16] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
BACKGROUND Atezolizumab is a humanised antiprogrammed death-ligand 1 (PD-L1) monoclonal antibody that inhibits PD-L1 and programmed death-1 (PD-1) and PD-L1 and B7-1 interactions, reinvigorating anticancer immunity. We assessed its efficacy and safety versus docetaxel in previously treated patients with non-small-cell lung cancer. METHODS We did a randomised, open-label, phase 3 trial (OAK) in 194 academic or community oncology centres in 31 countries. We enrolled patients who had squamous or non-squamous non-small-cell lung cancer, were 18 years or older, had measurable disease per Response Evaluation Criteria in Solid Tumors, and had an Eastern Cooperative Oncology Group performance status of 0 or 1. Patients had received one to two previous cytotoxic chemotherapy regimens (one or more platinum based combination therapies) for stage IIIB or IV non-small-cell lung cancer. Patients with a history of autoimmune disease and those who had received previous treatments with docetaxel, CD137 agonists, anti-CTLA4, or therapies targeting the PD-L1 and PD-1 pathway were excluded. Patients were randomly assigned (1:1) to intravenously receive either atezolizumab 1200 mg or docetaxel 75 mg/m2 every 3 weeks by permuted block randomisation (block size of eight) via an interactive voice or web response system. Coprimary endpoints were overall survival in the intention-to-treat (ITT) and PD-L1-expression population TC1/2/3 or IC1/2/3 (≥1% PD-L1 on tumour cells or tumour-infiltrating immune cells). The primary efficacy analysis was done in the first 850 of 1225 enrolled patients. This study is registered with ClinicalTrials.gov, number NCT02008227. FINDINGS Between March 11, 2014, and April 29, 2015, 1225 patients were recruited. In the primary population, 425 patients were randomly assigned to receive atezolizumab and 425 patients were assigned to receive docetaxel. Overall survival was significantly longer with atezolizumab in the ITT and PD-L1-expression populations. In the ITT population, overall survival was improved with atezolizumab compared with docetaxel (median overall survival was 13·8 months [95% CI 11·8-15·7] vs 9·6 months [8·6-11·2]; hazard ratio [HR] 0·73 [95% CI 0·62-0·87], p=0·0003). Overall survival in the TC1/2/3 or IC1/2/3 population was improved with atezolizumab (n=241) compared with docetaxel (n=222; median overall survival was 15·7 months [95% CI 12·6-18·0] with atezolizumab vs 10·3 months [8·8-12·0] with docetaxel; HR 0·74 [95% CI 0·58-0·93]; p=0·0102). Patients in the PD-L1 low or undetectable subgroup (TC0 and IC0) also had improved survival with atezolizumab (median overall survival 12·6 months vs 8·9 months; HR 0·75 [95% CI 0·59-0·96]). Overall survival improvement was similar in patients with squamous (HR 0·73 [95% CI 0·54-0·98]; n=112 in the atezolizumab group and n=110 in the docetaxel group) or non-squamous (0·73 [0·60-0·89]; n=313 and n=315) histology. Fewer patients had treatment-related grade 3 or 4 adverse events with atezolizumab (90 [15%] of 609 patients) versus docetaxel (247 [43%] of 578 patients). One treatment-related death from a respiratory tract infection was reported in the docetaxel group. INTERPRETATION To our knowledge, OAK is the first randomised phase 3 study to report results of a PD-L1-targeted therapy, with atezolizumab treatment resulting in a clinically relevant improvement of overall survival versus docetaxel in previously treated non-small-cell lung cancer, regardless of PD-L1 expression or histology, with a favourable safety profile. FUNDING F. Hoffmann-La Roche Ltd, Genentech, Inc.
Collapse
Affiliation(s)
| | - Fabrice Barlesi
- Aix Marseille Universite, Assistance Publique Hôpitaux de Marseille, Marseille, France
| | | | - Keunchil Park
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | | | | | | | | | | | - Manuel Cobo Dols
- Medical Oncology Section, Hospital Regional Universitario Carlos Haya, Málaga, Spain
| | | | | | | | | | | | | | | | - Hande Turna
- Istanbul University Cerrahpasa Medical Faculty Hospital, Medical Oncology, Istanbul, Turkey
| | - Jong-Seok Lee
- Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | | | | | - Pei He
- Genentech, Inc., South San Francisco, CA, USA
| | | | | | | | | |
Collapse
|
106
|
Rittmeyer A, Barlesi F, Waterkamp D, Park K, Ciardiello F, von Pawel J, Gadgeel SM, Hida T, Kowalski DM, Dols MC, Cortinovis DL, Leach J, Polikoff J, Barrios C, Kabbinavar F, Frontera OA, De Marinis F, Turna H, Lee JS, Ballinger M, Kowanetz M, He P, Chen DS, Sandler A, Gandara DR. Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial. Lancet 2017; 389:255-265. [PMID: 27979383 PMCID: PMC6886121 DOI: 10.1016/s0140-6736(16)32517-x] [Citation(s) in RCA: 3518] [Impact Index Per Article: 502.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 11/14/2016] [Accepted: 11/15/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND Atezolizumab is a humanised antiprogrammed death-ligand 1 (PD-L1) monoclonal antibody that inhibits PD-L1 and programmed death-1 (PD-1) and PD-L1 and B7-1 interactions, reinvigorating anticancer immunity. We assessed its efficacy and safety versus docetaxel in previously treated patients with non-small-cell lung cancer. METHODS We did a randomised, open-label, phase 3 trial (OAK) in 194 academic or community oncology centres in 31 countries. We enrolled patients who had squamous or non-squamous non-small-cell lung cancer, were 18 years or older, had measurable disease per Response Evaluation Criteria in Solid Tumors, and had an Eastern Cooperative Oncology Group performance status of 0 or 1. Patients had received one to two previous cytotoxic chemotherapy regimens (one or more platinum based combination therapies) for stage IIIB or IV non-small-cell lung cancer. Patients with a history of autoimmune disease and those who had received previous treatments with docetaxel, CD137 agonists, anti-CTLA4, or therapies targeting the PD-L1 and PD-1 pathway were excluded. Patients were randomly assigned (1:1) to intravenously receive either atezolizumab 1200 mg or docetaxel 75 mg/m2 every 3 weeks by permuted block randomisation (block size of eight) via an interactive voice or web response system. Coprimary endpoints were overall survival in the intention-to-treat (ITT) and PD-L1-expression population TC1/2/3 or IC1/2/3 (≥1% PD-L1 on tumour cells or tumour-infiltrating immune cells). The primary efficacy analysis was done in the first 850 of 1225 enrolled patients. This study is registered with ClinicalTrials.gov, number NCT02008227. FINDINGS Between March 11, 2014, and April 29, 2015, 1225 patients were recruited. In the primary population, 425 patients were randomly assigned to receive atezolizumab and 425 patients were assigned to receive docetaxel. Overall survival was significantly longer with atezolizumab in the ITT and PD-L1-expression populations. In the ITT population, overall survival was improved with atezolizumab compared with docetaxel (median overall survival was 13·8 months [95% CI 11·8-15·7] vs 9·6 months [8·6-11·2]; hazard ratio [HR] 0·73 [95% CI 0·62-0·87], p=0·0003). Overall survival in the TC1/2/3 or IC1/2/3 population was improved with atezolizumab (n=241) compared with docetaxel (n=222; median overall survival was 15·7 months [95% CI 12·6-18·0] with atezolizumab vs 10·3 months [8·8-12·0] with docetaxel; HR 0·74 [95% CI 0·58-0·93]; p=0·0102). Patients in the PD-L1 low or undetectable subgroup (TC0 and IC0) also had improved survival with atezolizumab (median overall survival 12·6 months vs 8·9 months; HR 0·75 [95% CI 0·59-0·96]). Overall survival improvement was similar in patients with squamous (HR 0·73 [95% CI 0·54-0·98]; n=112 in the atezolizumab group and n=110 in the docetaxel group) or non-squamous (0·73 [0·60-0·89]; n=313 and n=315) histology. Fewer patients had treatment-related grade 3 or 4 adverse events with atezolizumab (90 [15%] of 609 patients) versus docetaxel (247 [43%] of 578 patients). One treatment-related death from a respiratory tract infection was reported in the docetaxel group. INTERPRETATION To our knowledge, OAK is the first randomised phase 3 study to report results of a PD-L1-targeted therapy, with atezolizumab treatment resulting in a clinically relevant improvement of overall survival versus docetaxel in previously treated non-small-cell lung cancer, regardless of PD-L1 expression or histology, with a favourable safety profile. FUNDING F. Hoffmann-La Roche Ltd, Genentech, Inc.
Collapse
Affiliation(s)
| | - Fabrice Barlesi
- Aix Marseille Universite, Assistance Publique Hôpitaux de Marseille, Marseille, France
| | | | - Keunchil Park
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | | | | | | | | | | | - Manuel Cobo Dols
- Medical Oncology Section, Hospital Regional Universitario Carlos Haya, Málaga, Spain
| | | | | | | | | | | | | | | | - Hande Turna
- Istanbul University Cerrahpasa Medical Faculty Hospital, Medical Oncology, Istanbul, Turkey
| | - Jong-Seok Lee
- Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | | | | | - Pei He
- Genentech, Inc., South San Francisco, CA, USA
| | | | | | | |
Collapse
|
107
|
Immunosuppression in liver tumors: opening the portal to effective immunotherapy. Cancer Gene Ther 2016; 24:114-120. [DOI: 10.1038/cgt.2016.54] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 09/01/2016] [Indexed: 12/11/2022]
|
108
|
Enterovirus A71 and coxsackievirus A16 show different replication kinetics in human neuronal and non-neuronal cell lines. Arch Virol 2016; 162:727-737. [DOI: 10.1007/s00705-016-3157-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 10/19/2016] [Indexed: 01/15/2023]
|
109
|
Malvezzi P, Jouve T, Rostaing L. Costimulation Blockade in Kidney Transplantation: An Update. Transplantation 2016; 100:2315-2323. [PMID: 27472094 PMCID: PMC5084636 DOI: 10.1097/tp.0000000000001344] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 05/04/2016] [Accepted: 05/10/2016] [Indexed: 12/15/2022]
Abstract
In the setting of solid-organ transplantation, calcineurin inhibitor (CNI)-based therapy remains the cornerstone of immunosuppression. However, long-term use of CNIs is associated with some degree of nephrotoxicity. This has led to exploring the blockade of some costimulation pathways as an efficient immunosuppressive tool instead of using CNIs. The only agent already in clinical use and approved by the health authorities for kidney transplant patients is belatacept (Nulojix), a fusion protein that interferes with cytotoxic T lymphocyte-associated protein 4. Belatacept has been demonstrated to be as efficient as cyclosporine-based immunosuppression and is associated with significantly better renal function, that is, no nephrotoxicity. However, in the immediate posttransplant period, significantly more mild/moderate episodes of acute rejection have been reported, favored by the fact that cytotoxic T lymphocyte-associated protein pathway has an inhibitory effect on the alloimmune response; thereby its inhibition is detrimental in this regard. This has led to the development of antibodies that target CD28. The most advanced is FR104, it has shown promise in nonhuman primate models of autoimmune diseases and allotransplantation. In addition, research into blocking the CD40-CD154 pathway is underway. A phase II study testing ASK1240, that is, anti-CD40 antibody has been completed, and the results are pending.
Collapse
Affiliation(s)
- Paolo Malvezzi
- Clinique Universitaire de Néphrologie, Unité de Transplantation Rénale, CHU Grenoble, France
| | - Thomas Jouve
- Clinique Universitaire de Néphrologie, Unité de Transplantation Rénale, CHU Grenoble, France
- Université Joseph Fourier, Grenoble, France
| | - Lionel Rostaing
- Clinique Universitaire de Néphrologie, Unité de Transplantation Rénale, CHU Grenoble, France
- UniversitéToulouse III Paul Sabatier, Toulouse, France
- INSERM U563, IFR-BMT, CHU Purpan, Toulouse, France
| |
Collapse
|
110
|
Clark CA, Gupta HB, Sareddy G, Pandeswara S, Lao S, Yuan B, Drerup JM, Padron A, Conejo-Garcia J, Murthy K, Liu Y, Turk MJ, Thedieck K, Hurez V, Li R, Vadlamudi R, Curiel TJ. Tumor-Intrinsic PD-L1 Signals Regulate Cell Growth, Pathogenesis, and Autophagy in Ovarian Cancer and Melanoma. Cancer Res 2016; 76:6964-6974. [PMID: 27671674 DOI: 10.1158/0008-5472.can-16-0258] [Citation(s) in RCA: 277] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 09/16/2016] [Accepted: 09/16/2016] [Indexed: 01/09/2023]
Abstract
PD-L1 antibodies produce efficacious clinical responses in diverse human cancers, but the basis for their effects remains unclear, leaving a gap in the understanding of how to rationally leverage therapeutic activity. PD-L1 is widely expressed in tumor cells, but its contributions to tumor pathogenicity are incompletely understood. In this study, we evaluated the hypothesis that PD-L1 exerts tumor cell-intrinsic signals that are critical for pathogenesis. Using RNAi methodology, we attenuated PD-L1 in the murine ovarian cell line ID8agg and the melanoma cell line B16 (termed PD-L1lo cells), which express basal PD-L1. We observed that PD-L1lo cells proliferated more weakly than control cells in vitro As expected, PD-L1lo cells formed tumors in immunocompetent mice relatively more slowly, but unexpectedly, they also formed tumors more slowly in immunodeficient NSG mice. RNA sequencing analysis identified a number of genes involved in autophagy and mTOR signaling that were affected by PD-L1 expression. In support of a functional role, PD-L1 attenuation augmented autophagy and blunted the ability of autophagy inhibitors to limit proliferation in vitro and in vivo in NSG mice. PD-L1 attenuation also reduced mTORC1 activity and augmented the antiproliferative effects of the mTORC1 inhibitor rapamycin. PD-L1lo cells were also relatively deficient in metastasis to the lung, and we found that anti-PD-L1 administration could block tumor cell growth and metastasis in NSG mice. This therapeutic effect was observed with B16 cells but not ID8agg cells, illustrating tumor- or compartmental-specific effects in the therapeutic setting. Overall, our findings extend understanding of PD-L1 functions, illustrate nonimmune effects of anti-PD-L1 immunotherapy, and suggest broader uses for PD-L1 as a biomarker for assessing cancer therapeutic responses. Cancer Res; 76(23); 6964-74. ©2016 AACR.
Collapse
Affiliation(s)
- Curtis A Clark
- The Graduate School of Biomedical Sciences, University of Texas Health Science Center, San Antonio, Texas.,Department of Medicine, University of Texas Health Science Center, San Antonio, Texas
| | - Harshita B Gupta
- Department of Medicine, University of Texas Health Science Center, San Antonio, Texas
| | - Gangadhara Sareddy
- Cancer Therapy & Research Center, University of Texas Health Science Center, San Antonio, Texas.,Department of Obstetrics and Gynecology, University of Texas Health Science Center, San Antonio, Texas
| | - Srilakshmi Pandeswara
- Department of Medicine, University of Texas Health Science Center, San Antonio, Texas
| | - Shunhua Lao
- Department of Medicine, University of Texas Health Science Center, San Antonio, Texas
| | - Bin Yuan
- Department of Medicine, University of Texas Health Science Center, San Antonio, Texas.,Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, Texas
| | - Justin M Drerup
- The Graduate School of Biomedical Sciences, University of Texas Health Science Center, San Antonio, Texas.,Department of Medicine, University of Texas Health Science Center, San Antonio, Texas
| | - Alvaro Padron
- Department of Medicine, University of Texas Health Science Center, San Antonio, Texas
| | - José Conejo-Garcia
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania
| | - Kruthi Murthy
- The Graduate School of Biomedical Sciences, University of Texas Health Science Center, San Antonio, Texas.,Department of Medicine, University of Texas Health Science Center, San Antonio, Texas
| | - Yang Liu
- Department of Medicine, University of Texas Health Science Center, San Antonio, Texas.,Xiangya School of Medicine, Central South University, Changsha, Hunan, PR China
| | - Mary Jo Turk
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Kathrin Thedieck
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands and Department for Neuroscience, School of Medicine and Health Sciences, University Oldenburg, Oldenburg, Germany
| | - Vincent Hurez
- Department of Medicine, University of Texas Health Science Center, San Antonio, Texas
| | - Rong Li
- The Graduate School of Biomedical Sciences, University of Texas Health Science Center, San Antonio, Texas.,Cancer Therapy & Research Center, University of Texas Health Science Center, San Antonio, Texas.,Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, Texas
| | - Ratna Vadlamudi
- The Graduate School of Biomedical Sciences, University of Texas Health Science Center, San Antonio, Texas.,Cancer Therapy & Research Center, University of Texas Health Science Center, San Antonio, Texas.,Department of Obstetrics and Gynecology, University of Texas Health Science Center, San Antonio, Texas
| | - Tyler J Curiel
- The Graduate School of Biomedical Sciences, University of Texas Health Science Center, San Antonio, Texas. .,Department of Medicine, University of Texas Health Science Center, San Antonio, Texas.,Cancer Therapy & Research Center, University of Texas Health Science Center, San Antonio, Texas
| |
Collapse
|
111
|
The future of cancer treatment: immunomodulation, CARs and combination immunotherapy. Nat Rev Clin Oncol 2016; 13:273-90. [PMID: 26977780 DOI: 10.1038/nrclinonc.2016.25] [Citation(s) in RCA: 736] [Impact Index Per Article: 92.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In the past decade, advances in the use of monoclonal antibodies (mAbs) and adoptive cellular therapy to treat cancer by modulating the immune response have led to unprecedented responses in patients with advanced-stage tumours that would otherwise have been fatal. To date, three immune-checkpoint-blocking mAbs have been approved in the USA for the treatment of patients with several types of cancer, and more patients will benefit from immunomodulatory mAb therapy in the months and years ahead. Concurrently, the adoptive transfer of genetically modified lymphocytes to treat patients with haematological malignancies has yielded dramatic results, and we anticipate that this approach will rapidly become the standard of care for an increasing number of patients. In this Review, we highlight the latest advances in immunotherapy and discuss the role that it will have in the future of cancer treatment, including settings for which testing combination strategies and 'armoured' CAR T cells are recommended.
Collapse
|
112
|
Xia B, Herbst RS. Immune checkpoint therapy for non-small-cell lung cancer: an update. Immunotherapy 2016; 8:279-98. [DOI: 10.2217/imt.15.123] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The role of immunotherapy in treatment of non-small-cell lung cancer (NSCLC) has been gaining interest over the past few years. This has been driven primarily by promising results from trials evaluating antagonist antibodies that target co-inhibitory immune checkpoints expressed on tumor cells and immune cells within the tumor microenvironment. Immune checkpoints exist to dampen or terminate immune activity to guard against autoimmunity and allow for self-tolerance. However, tumors can take advantage of these immune checkpoint pathways to evade destruction. Antibodies that block inhibitory checkpoints, such as anti-CTLA-4, anti-PD1 and anti-PD-L1 antibodies have demonstrated delayed tumor growth and increased survival. Novel therapies are now investigating combining checkpoint inhibitors with chemotherapy, targeted therapy, radiation and vaccines to produce synergistic antitumor activity.
Collapse
Affiliation(s)
- Bing Xia
- Yale Comprehensive Cancer Center, Yale School of Medicine, 333 Cedar Street WWW221, New Haven, CT 06520, USA
| | - Roy S Herbst
- Yale Comprehensive Cancer Center, Yale School of Medicine, 333 Cedar Street WWW221, New Haven, CT 06520, USA
| |
Collapse
|
113
|
Predictive biomarkers in PD-1/PD-L1 checkpoint blockade immunotherapy. Cancer Treat Rev 2015; 41:868-76. [PMID: 26589760 DOI: 10.1016/j.ctrv.2015.11.001] [Citation(s) in RCA: 298] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Revised: 10/29/2015] [Accepted: 11/01/2015] [Indexed: 12/30/2022]
Abstract
Checkpoint blockades turn on a new paradigm shift in immunotherapy for cancer. Remarkable clinical efficacy, durable response and low toxicity of programmed death 1 (PD-1)/programmed death ligand-1 (PD-L1) checkpoint blockades have been observed in various malignancies. However, a lot of cancer patients failed to respond to the PD-1/PD-L1 checkpoint blockades. It is crucial to identify a biomarker to predict the response to checkpoint blockades. The overexpression of PD-L1 is an important and widely-explored predictive biomarker for the response to PD-1/PD-L1 antibodies. However PD-L1 staining cannot be used to accurately select patients for PD-1/PD-L1 pathway blockade due to the low prediction accuracy and dynamic changes. Tumor-infiltrating immune cells and molecules in the tumor microenvironment, or along with PD-L1 expression, may be important in predicting clinical benefits of PD-1/PD-L1 checkpoint blockades. Gene analysis has proven to be new approach for judging the potential clinical benefit of immune checkpoint inhibitors, such as mutational landscape and mismatch-repair deficiency. Further preclinical and clinical studies are necessary to carry out before its application in clinical practice. Challenges should be overcome to identify patients accurately who will benefit from PD-1/PD-L1 checkpoint blockades. In this review, we focus on the predictive biomarkers for checkpoint blockades of PD-1/PD-L1 pathway.
Collapse
|
114
|
Nduom EK, Wei J, Yaghi NK, Huang N, Kong LY, Gabrusiewicz K, Ling X, Zhou S, Ivan C, Chen JQ, Burks JK, Fuller GN, Calin GA, Conrad CA, Creasy C, Ritthipichai K, Radvanyi L, Heimberger AB. PD-L1 expression and prognostic impact in glioblastoma. Neuro Oncol 2015; 18:195-205. [PMID: 26323609 DOI: 10.1093/neuonc/nov172] [Citation(s) in RCA: 426] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 07/25/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Therapeutic targeting of the immune checkpoints cytotoxic T-lymphocyte-associated molecule-4 (CTLA-4) and PD-1/PD-L1 has demonstrated tumor regression in clinical trials, and phase 2 trials are ongoing in glioblastoma (GBM). Previous reports have suggested that responses are more frequent in patients with tumors that express PD-L1; however, this has been disputed. At issue is the validation of PD-L1 biomarker assays and prognostic impact. METHODS Using immunohistochemical analysis, we measured the incidence of PD-L1 expression in 94 patients with GBM. We categorized our results according to the total number of PD-L1-expressing cells within the GBMs and then validated this finding in ex vivo GBM flow cytometry with further analysis of the T cell populations. We then evaluated the association between PD-L1 expression and median survival time using the protein expression datasets and mRNA from The Cancer Genome Atlas. RESULTS The median percentage of PD-L1-expressing cells in GBM by cell surface staining is 2.77% (range: 0%-86.6%; n = 92), which is similar to the percentage found by ex vivo flow cytometry. The majority of GBM patients (61%) had tumors with at least 1% or more PD-L1-positive cells, and 38% had at least 5% or greater PD-L1 expression. PD-L1 is commonly expressed on the GBM-infiltrating T cells. Expression of both PD-L1 and PD-1 are negative prognosticators for GBM outcome. CONCLUSIONS The incidence of PD-L1 expression in GBM patients is frequent but is confined to a minority subpopulation, similar to other malignancies that have been profiled for PD-L1 expression. Higher expression of PD-L1 is correlated with worse outcome.
Collapse
Affiliation(s)
- Edjah K Nduom
- Departments of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (E.K.N., J.W., N.K.Y., N.H., L.-Y.K., K.G., X.L., A.B.H.); Department of Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (S.Z.); Center for RNA Interference and Non-Coding RNAs, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.I.); Flow Cytometry and Cell Imaging Core Facility, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.K.B.); Neuropathology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (G.N.F.); Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (G.A.C.); Neuro-Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.A.C.); Melanoma Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.C., K.R.); Lion Biotechnologies, Woodland Hills, California (J.Q.C., L.R.); Dept. of Immunology, H. Lee Moffitt Cancer Center, Tampa, Florida (J.Q.C., L.R.)
| | - Jun Wei
- Departments of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (E.K.N., J.W., N.K.Y., N.H., L.-Y.K., K.G., X.L., A.B.H.); Department of Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (S.Z.); Center for RNA Interference and Non-Coding RNAs, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.I.); Flow Cytometry and Cell Imaging Core Facility, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.K.B.); Neuropathology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (G.N.F.); Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (G.A.C.); Neuro-Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.A.C.); Melanoma Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.C., K.R.); Lion Biotechnologies, Woodland Hills, California (J.Q.C., L.R.); Dept. of Immunology, H. Lee Moffitt Cancer Center, Tampa, Florida (J.Q.C., L.R.)
| | - Nasser K Yaghi
- Departments of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (E.K.N., J.W., N.K.Y., N.H., L.-Y.K., K.G., X.L., A.B.H.); Department of Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (S.Z.); Center for RNA Interference and Non-Coding RNAs, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.I.); Flow Cytometry and Cell Imaging Core Facility, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.K.B.); Neuropathology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (G.N.F.); Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (G.A.C.); Neuro-Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.A.C.); Melanoma Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.C., K.R.); Lion Biotechnologies, Woodland Hills, California (J.Q.C., L.R.); Dept. of Immunology, H. Lee Moffitt Cancer Center, Tampa, Florida (J.Q.C., L.R.)
| | - Neal Huang
- Departments of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (E.K.N., J.W., N.K.Y., N.H., L.-Y.K., K.G., X.L., A.B.H.); Department of Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (S.Z.); Center for RNA Interference and Non-Coding RNAs, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.I.); Flow Cytometry and Cell Imaging Core Facility, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.K.B.); Neuropathology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (G.N.F.); Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (G.A.C.); Neuro-Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.A.C.); Melanoma Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.C., K.R.); Lion Biotechnologies, Woodland Hills, California (J.Q.C., L.R.); Dept. of Immunology, H. Lee Moffitt Cancer Center, Tampa, Florida (J.Q.C., L.R.)
| | - Ling-Yuan Kong
- Departments of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (E.K.N., J.W., N.K.Y., N.H., L.-Y.K., K.G., X.L., A.B.H.); Department of Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (S.Z.); Center for RNA Interference and Non-Coding RNAs, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.I.); Flow Cytometry and Cell Imaging Core Facility, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.K.B.); Neuropathology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (G.N.F.); Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (G.A.C.); Neuro-Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.A.C.); Melanoma Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.C., K.R.); Lion Biotechnologies, Woodland Hills, California (J.Q.C., L.R.); Dept. of Immunology, H. Lee Moffitt Cancer Center, Tampa, Florida (J.Q.C., L.R.)
| | - Konrad Gabrusiewicz
- Departments of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (E.K.N., J.W., N.K.Y., N.H., L.-Y.K., K.G., X.L., A.B.H.); Department of Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (S.Z.); Center for RNA Interference and Non-Coding RNAs, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.I.); Flow Cytometry and Cell Imaging Core Facility, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.K.B.); Neuropathology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (G.N.F.); Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (G.A.C.); Neuro-Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.A.C.); Melanoma Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.C., K.R.); Lion Biotechnologies, Woodland Hills, California (J.Q.C., L.R.); Dept. of Immunology, H. Lee Moffitt Cancer Center, Tampa, Florida (J.Q.C., L.R.)
| | - Xiaoyang Ling
- Departments of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (E.K.N., J.W., N.K.Y., N.H., L.-Y.K., K.G., X.L., A.B.H.); Department of Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (S.Z.); Center for RNA Interference and Non-Coding RNAs, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.I.); Flow Cytometry and Cell Imaging Core Facility, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.K.B.); Neuropathology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (G.N.F.); Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (G.A.C.); Neuro-Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.A.C.); Melanoma Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.C., K.R.); Lion Biotechnologies, Woodland Hills, California (J.Q.C., L.R.); Dept. of Immunology, H. Lee Moffitt Cancer Center, Tampa, Florida (J.Q.C., L.R.)
| | - Shouhao Zhou
- Departments of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (E.K.N., J.W., N.K.Y., N.H., L.-Y.K., K.G., X.L., A.B.H.); Department of Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (S.Z.); Center for RNA Interference and Non-Coding RNAs, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.I.); Flow Cytometry and Cell Imaging Core Facility, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.K.B.); Neuropathology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (G.N.F.); Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (G.A.C.); Neuro-Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.A.C.); Melanoma Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.C., K.R.); Lion Biotechnologies, Woodland Hills, California (J.Q.C., L.R.); Dept. of Immunology, H. Lee Moffitt Cancer Center, Tampa, Florida (J.Q.C., L.R.)
| | - Cristina Ivan
- Departments of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (E.K.N., J.W., N.K.Y., N.H., L.-Y.K., K.G., X.L., A.B.H.); Department of Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (S.Z.); Center for RNA Interference and Non-Coding RNAs, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.I.); Flow Cytometry and Cell Imaging Core Facility, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.K.B.); Neuropathology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (G.N.F.); Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (G.A.C.); Neuro-Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.A.C.); Melanoma Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.C., K.R.); Lion Biotechnologies, Woodland Hills, California (J.Q.C., L.R.); Dept. of Immunology, H. Lee Moffitt Cancer Center, Tampa, Florida (J.Q.C., L.R.)
| | - Jie Qing Chen
- Departments of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (E.K.N., J.W., N.K.Y., N.H., L.-Y.K., K.G., X.L., A.B.H.); Department of Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (S.Z.); Center for RNA Interference and Non-Coding RNAs, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.I.); Flow Cytometry and Cell Imaging Core Facility, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.K.B.); Neuropathology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (G.N.F.); Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (G.A.C.); Neuro-Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.A.C.); Melanoma Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.C., K.R.); Lion Biotechnologies, Woodland Hills, California (J.Q.C., L.R.); Dept. of Immunology, H. Lee Moffitt Cancer Center, Tampa, Florida (J.Q.C., L.R.)
| | - Jared K Burks
- Departments of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (E.K.N., J.W., N.K.Y., N.H., L.-Y.K., K.G., X.L., A.B.H.); Department of Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (S.Z.); Center for RNA Interference and Non-Coding RNAs, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.I.); Flow Cytometry and Cell Imaging Core Facility, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.K.B.); Neuropathology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (G.N.F.); Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (G.A.C.); Neuro-Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.A.C.); Melanoma Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.C., K.R.); Lion Biotechnologies, Woodland Hills, California (J.Q.C., L.R.); Dept. of Immunology, H. Lee Moffitt Cancer Center, Tampa, Florida (J.Q.C., L.R.)
| | - Greg N Fuller
- Departments of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (E.K.N., J.W., N.K.Y., N.H., L.-Y.K., K.G., X.L., A.B.H.); Department of Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (S.Z.); Center for RNA Interference and Non-Coding RNAs, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.I.); Flow Cytometry and Cell Imaging Core Facility, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.K.B.); Neuropathology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (G.N.F.); Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (G.A.C.); Neuro-Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.A.C.); Melanoma Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.C., K.R.); Lion Biotechnologies, Woodland Hills, California (J.Q.C., L.R.); Dept. of Immunology, H. Lee Moffitt Cancer Center, Tampa, Florida (J.Q.C., L.R.)
| | - George A Calin
- Departments of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (E.K.N., J.W., N.K.Y., N.H., L.-Y.K., K.G., X.L., A.B.H.); Department of Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (S.Z.); Center for RNA Interference and Non-Coding RNAs, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.I.); Flow Cytometry and Cell Imaging Core Facility, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.K.B.); Neuropathology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (G.N.F.); Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (G.A.C.); Neuro-Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.A.C.); Melanoma Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.C., K.R.); Lion Biotechnologies, Woodland Hills, California (J.Q.C., L.R.); Dept. of Immunology, H. Lee Moffitt Cancer Center, Tampa, Florida (J.Q.C., L.R.)
| | - Charles A Conrad
- Departments of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (E.K.N., J.W., N.K.Y., N.H., L.-Y.K., K.G., X.L., A.B.H.); Department of Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (S.Z.); Center for RNA Interference and Non-Coding RNAs, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.I.); Flow Cytometry and Cell Imaging Core Facility, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.K.B.); Neuropathology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (G.N.F.); Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (G.A.C.); Neuro-Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.A.C.); Melanoma Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.C., K.R.); Lion Biotechnologies, Woodland Hills, California (J.Q.C., L.R.); Dept. of Immunology, H. Lee Moffitt Cancer Center, Tampa, Florida (J.Q.C., L.R.)
| | - Caitlin Creasy
- Departments of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (E.K.N., J.W., N.K.Y., N.H., L.-Y.K., K.G., X.L., A.B.H.); Department of Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (S.Z.); Center for RNA Interference and Non-Coding RNAs, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.I.); Flow Cytometry and Cell Imaging Core Facility, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.K.B.); Neuropathology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (G.N.F.); Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (G.A.C.); Neuro-Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.A.C.); Melanoma Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.C., K.R.); Lion Biotechnologies, Woodland Hills, California (J.Q.C., L.R.); Dept. of Immunology, H. Lee Moffitt Cancer Center, Tampa, Florida (J.Q.C., L.R.)
| | - Krit Ritthipichai
- Departments of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (E.K.N., J.W., N.K.Y., N.H., L.-Y.K., K.G., X.L., A.B.H.); Department of Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (S.Z.); Center for RNA Interference and Non-Coding RNAs, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.I.); Flow Cytometry and Cell Imaging Core Facility, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.K.B.); Neuropathology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (G.N.F.); Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (G.A.C.); Neuro-Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.A.C.); Melanoma Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.C., K.R.); Lion Biotechnologies, Woodland Hills, California (J.Q.C., L.R.); Dept. of Immunology, H. Lee Moffitt Cancer Center, Tampa, Florida (J.Q.C., L.R.)
| | - Laszlo Radvanyi
- Departments of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (E.K.N., J.W., N.K.Y., N.H., L.-Y.K., K.G., X.L., A.B.H.); Department of Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (S.Z.); Center for RNA Interference and Non-Coding RNAs, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.I.); Flow Cytometry and Cell Imaging Core Facility, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.K.B.); Neuropathology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (G.N.F.); Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (G.A.C.); Neuro-Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.A.C.); Melanoma Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.C., K.R.); Lion Biotechnologies, Woodland Hills, California (J.Q.C., L.R.); Dept. of Immunology, H. Lee Moffitt Cancer Center, Tampa, Florida (J.Q.C., L.R.)
| | - Amy B Heimberger
- Departments of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (E.K.N., J.W., N.K.Y., N.H., L.-Y.K., K.G., X.L., A.B.H.); Department of Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (S.Z.); Center for RNA Interference and Non-Coding RNAs, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.I.); Flow Cytometry and Cell Imaging Core Facility, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.K.B.); Neuropathology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (G.N.F.); Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (G.A.C.); Neuro-Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.A.C.); Melanoma Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (C.C., K.R.); Lion Biotechnologies, Woodland Hills, California (J.Q.C., L.R.); Dept. of Immunology, H. Lee Moffitt Cancer Center, Tampa, Florida (J.Q.C., L.R.)
| |
Collapse
|
115
|
Bodhankar S, Chen Y, Lapato A, Dotson AL, Wang J, Vandenbark AA, Saugstad JA, Offner H. PD-L1 Monoclonal Antibody Treats Ischemic Stroke by Controlling Central Nervous System Inflammation. Stroke 2015; 46:2926-34. [PMID: 26306753 DOI: 10.1161/strokeaha.115.010592] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 08/05/2015] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Both pathogenic and regulatory immune processes are involved in the middle cerebral artery occlusion (MCAO) model of experimental stroke, including interactions involving the programmed death 1 (PD-1) receptor and its 2 ligands, PD-L1 and PD-L2. Although PD-1 reduced stroke severity, PD-L1 and PD-L2 appeared to play pathogenic roles, suggesting the use of anti-PD-L monoclonal antibody therapy for MCAO. METHODS Male C57BL/6 mice were treated with a single dose of anti-PD-L1 monoclonal antibody 4 hours after MCAO and evaluated for clinical, histological and immunologic changes after 96 hours of reperfusion. RESULTS Blockade of the PD-L1 checkpoint using a single injection of 200 μg anti-PD-L1 monoclonal antibody given intravenously 4 hours after occlusion significantly reduced MCAO infarct volumes and improved neurological outcomes after 96 hours of reperfusion. Treatment partially reversed splenic atrophy and decreased central nervous system infiltrating immune cells concomitant with enhanced appearance of CD8(+) regulatory T cells in the lesioned central nervous system hemisphere. CONCLUSIONS This study demonstrates for the first time the beneficial therapeutic effects of PD-L1 checkpoint blockade on MCAO, thus validating proposed mechanisms obtained in our previous studies using PD-1- and PD-L-deficient mice. These results provide strong support for the use of available humanized anti-PD-L1 antibodies for treatment of human stroke subjects.
Collapse
Affiliation(s)
- Sheetal Bodhankar
- From the Neuroimmunology Research, VA Portland Health Care System, OR (S.B., A.L., A.L.D., A.A.V., H.O.); and Departments of Neurology (S.B., A.L., A.L.D., A.A.V., J.A.S., H.O.), Anesthesiology and Perioperative Medicine (Y.C., J.W., J.A.S., H.O.), Molecular Microbiology and Immunology (A.A.V.), and Medical and Molecular Genetics (J.A.S.), Oregon Health and Science University, Portland
| | - Yingxin Chen
- From the Neuroimmunology Research, VA Portland Health Care System, OR (S.B., A.L., A.L.D., A.A.V., H.O.); and Departments of Neurology (S.B., A.L., A.L.D., A.A.V., J.A.S., H.O.), Anesthesiology and Perioperative Medicine (Y.C., J.W., J.A.S., H.O.), Molecular Microbiology and Immunology (A.A.V.), and Medical and Molecular Genetics (J.A.S.), Oregon Health and Science University, Portland
| | - Andrew Lapato
- From the Neuroimmunology Research, VA Portland Health Care System, OR (S.B., A.L., A.L.D., A.A.V., H.O.); and Departments of Neurology (S.B., A.L., A.L.D., A.A.V., J.A.S., H.O.), Anesthesiology and Perioperative Medicine (Y.C., J.W., J.A.S., H.O.), Molecular Microbiology and Immunology (A.A.V.), and Medical and Molecular Genetics (J.A.S.), Oregon Health and Science University, Portland
| | - Abby L Dotson
- From the Neuroimmunology Research, VA Portland Health Care System, OR (S.B., A.L., A.L.D., A.A.V., H.O.); and Departments of Neurology (S.B., A.L., A.L.D., A.A.V., J.A.S., H.O.), Anesthesiology and Perioperative Medicine (Y.C., J.W., J.A.S., H.O.), Molecular Microbiology and Immunology (A.A.V.), and Medical and Molecular Genetics (J.A.S.), Oregon Health and Science University, Portland
| | - Jianming Wang
- From the Neuroimmunology Research, VA Portland Health Care System, OR (S.B., A.L., A.L.D., A.A.V., H.O.); and Departments of Neurology (S.B., A.L., A.L.D., A.A.V., J.A.S., H.O.), Anesthesiology and Perioperative Medicine (Y.C., J.W., J.A.S., H.O.), Molecular Microbiology and Immunology (A.A.V.), and Medical and Molecular Genetics (J.A.S.), Oregon Health and Science University, Portland
| | - Arthur A Vandenbark
- From the Neuroimmunology Research, VA Portland Health Care System, OR (S.B., A.L., A.L.D., A.A.V., H.O.); and Departments of Neurology (S.B., A.L., A.L.D., A.A.V., J.A.S., H.O.), Anesthesiology and Perioperative Medicine (Y.C., J.W., J.A.S., H.O.), Molecular Microbiology and Immunology (A.A.V.), and Medical and Molecular Genetics (J.A.S.), Oregon Health and Science University, Portland
| | - Julie A Saugstad
- From the Neuroimmunology Research, VA Portland Health Care System, OR (S.B., A.L., A.L.D., A.A.V., H.O.); and Departments of Neurology (S.B., A.L., A.L.D., A.A.V., J.A.S., H.O.), Anesthesiology and Perioperative Medicine (Y.C., J.W., J.A.S., H.O.), Molecular Microbiology and Immunology (A.A.V.), and Medical and Molecular Genetics (J.A.S.), Oregon Health and Science University, Portland
| | - Halina Offner
- From the Neuroimmunology Research, VA Portland Health Care System, OR (S.B., A.L., A.L.D., A.A.V., H.O.); and Departments of Neurology (S.B., A.L., A.L.D., A.A.V., J.A.S., H.O.), Anesthesiology and Perioperative Medicine (Y.C., J.W., J.A.S., H.O.), Molecular Microbiology and Immunology (A.A.V.), and Medical and Molecular Genetics (J.A.S.), Oregon Health and Science University, Portland.
| |
Collapse
|
116
|
Development of PD-1/PD-L1 Pathway in Tumor Immune Microenvironment and Treatment for Non-Small Cell Lung Cancer. Sci Rep 2015; 5:13110. [PMID: 26279307 PMCID: PMC4538573 DOI: 10.1038/srep13110] [Citation(s) in RCA: 269] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 07/15/2015] [Indexed: 02/07/2023] Open
Abstract
Lung cancer is currently the leading cause of cancer-related death in worldwide, non-small cell lung cancer (NSCLC) accounts for about 85% of all lung cancers. Surgery, platinum-based chemotherapy, molecular targeted agents and radiotherapy are the main treatment of NSCLC. With the strategies of treatment constantly improving, the prognosis of NSCLC patients is not as good as before, new sort of treatments are needed to be exploited. Programmed death 1 (PD-1) and its ligand PD-L1 play a key role in tumor immune escape and the formation of tumor microenvironment, closely related with tumor generation and development. Blockading the PD-1/PD-L1 pathway could reverse the tumor microenvironment and enhance the endogenous antitumor immune responses. Utilizing the PD-1 and/or PD-L1 inhibitors has shown benefits in clinical trials of NSCLC. In this review, we discuss the basic principle of PD-1/PD-L1 pathway and its role in the tumorigenesis and development of NSCLC. The clinical development of PD-1/PD-L1 pathway inhibitors and the main problems in the present studies and the research direction in the future will also be discussed.
Collapse
|
117
|
Madorsky Rowdo FP, Baron A, Urrutia M, Mordoh J. Immunotherapy in Cancer: A Combat between Tumors and the Immune System; You Win Some, You Lose Some. Front Immunol 2015; 6:127. [PMID: 25859247 PMCID: PMC4374472 DOI: 10.3389/fimmu.2015.00127] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 03/08/2015] [Indexed: 12/18/2022] Open
Abstract
Cancer immunotherapy has emerged as a treatment modality, mainly as the result of discoveries in the immune response regulation, including mechanisms that turn off immune responses. Immunogenic cutaneous melanoma is a canonical model for therapeutic immunotherapy studies. “Passive” immunotherapy with monoclonal antibodies (mAbs) has outpaced “active” immunotherapy with anti-tumor vaccines, and mAbs that antagonize the off responses have been recently introduced in clinical practice. Despite these recent successes, many unresolved practical and theoretical questions remain. Notably unknown are the identity of the lymphocytes that eliminate tumor cells, which white cells enter into tumors, through which endothelium, in what order, and how they perform their task. The parameters of size and location that could be used to determine in which tumors the immune response may be sufficient to eradicate the tumor are yet unknown. Immunotherapy has been so far more efficient to treat solid and hematologic tumors located outside the central nervous system, than primary brain tumors and brain metastases. In contrast to recent advances with mAbs, anti-tumor vaccine development has been lagging behind. The multiplicity of antigens that must be targeted to achieve significant clinical response is partially responsible for this lag, especially in melanoma, one of the most mutated tumors. Further hampering vaccination results is the fact that tumor elimination by the immune system is the result of a race between tumors with different growth rates and the relatively slow development of the adaptive immune response. The enhancement of the native arm of the immune response or the administration of targeted chemotherapy to slow tumor development, are approaches that should be studied. Finally, criteria used to analyze patient response to immunotherapeutic treatments must be perfected, and the patient populations that could benefit the most from this approach must be better defined.
Collapse
Affiliation(s)
| | - Antonela Baron
- Laboratorio de Cancerología, Fundación Instituto Leloir - IIBBA-CONICET , Buenos Aires , Argentina
| | - Mariela Urrutia
- Laboratorio de Cancerología, Fundación Instituto Leloir - IIBBA-CONICET , Buenos Aires , Argentina
| | - José Mordoh
- Laboratorio de Cancerología, Fundación Instituto Leloir - IIBBA-CONICET , Buenos Aires , Argentina ; Centro de Investigaciones Oncológicas, Fundación Cáncer and Instituto Alexander Fleming , Buenos Aires , Argentina
| |
Collapse
|
118
|
PD-L1 checkpoint blockade prevents immune dysfunction and leukemia development in a mouse model of chronic lymphocytic leukemia. Blood 2015; 126:203-11. [PMID: 25800048 DOI: 10.1182/blood-2015-01-622936] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Accepted: 03/14/2015] [Indexed: 01/12/2023] Open
Abstract
Blockade of the programmed cell death 1 (PD-1)/programmed death-ligand 1 (PD-L1) immune checkpoint augments antitumor immunity and induces durable responses in patients with solid cancers, but data on clinical efficacy in leukemias are sparse. Chronic lymphocytic leukemia (CLL) is associated with a tumor-supportive microenvironment and a dysfunctional immune system, as shown by "exhausted" T cells, defective immunologic synapse formation, and immunosuppressive myeloid cells. These defects involve aberrant expression of PD-L1 and are closely mirrored in the Eµ-TCL1 mouse model for CLL. In this study, we treated mice after adoptive transfer of Eµ-TCL1 CLL with PD-L1-blocking antibodies, which prevented CLL development and was accompanied by a reactivation of immune effector functions. This included restoration of mature macrophages and major histocompatibility complex class II-expressing dendritic cells and prevention of aberrant and exhaustion-like T-cell phenotypes. In addition, PD-L1 blockade restored CD8 T-cell cytotoxicity and immune synapse formation and normalized T-cell cytokines and proliferation ex vivo and in vivo. Our data demonstrate that early PD-L1 blockade effectively corrects leukemia-induced immune dysfunction and thus prevents CLL development in mice. Targeting PD-L1/PD-1 interactions should therefore be further explored in clinical studies with CLL patients, ideally in combination with novel compounds to help eliminate CLL.
Collapse
|
119
|
Blake SJP, Ching ALH, Kenna TJ, Galea R, Large J, Yagita H, Steptoe RJ. Blockade of PD-1/PD-L1 promotes adoptive T-cell immunotherapy in a tolerogenic environment. PLoS One 2015; 10:e0119483. [PMID: 25741704 PMCID: PMC4351071 DOI: 10.1371/journal.pone.0119483] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 01/19/2015] [Indexed: 12/22/2022] Open
Abstract
Adoptive cellular immunotherapy using in vitro expanded CD8+ T cells shows promise for tumour immunotherapy but is limited by eventual loss of function of the transferred T cells through factors that likely include inactivation by tolerogenic dendritic cells (DC). The co-inhibitory receptor programmed death-1 (PD-1), in addition to controlling T-cell responsiveness at effector sites in malignancies and chronic viral diseases is an important modulator of dendritic cell-induced tolerance in naive T cell populations. The most potent therapeutic capacity amongst CD8+ T cells appears to lie within Tcm or Tcm-like cells but memory T cells express elevated levels of PD-1. Based on established trafficking patterns for Tcm it is likely Tcm-like cells interact with lymphoid-tissue DC that present tumour-derived antigens and may be inherently tolerogenic to develop therapeutic effector function. As little is understood of the effect of PD-1/PD-L1 blockade on Tcm-like CD8+ T cells, particularly in relation to inactivation by DC, we explored the effects of PD-1/PD-L1 blockade in a mouse model where resting DC tolerise effector and memory CD8+ T cells. Blockade of PD-1/PD-L1 promoted effector differentiation of adoptively-transferred Tcm-phenotype cells interacting with tolerising DC. In tumour-bearing mice with tolerising DC, effector activity was increased in both lymphoid tissues and the tumour-site and anti-tumour activity was promoted. Our findings suggest PD-1/PD-L1 blockade may be a useful adjunct for adoptive immunotherapy by promoting effector differentiation in the host of transferred Tcm-like cells.
Collapse
Affiliation(s)
| | - Alan L. H. Ching
- UQ Diamantina Institute, University of Queensland, Brisbane, Australia
| | - Tony J. Kenna
- UQ Diamantina Institute, University of Queensland, Brisbane, Australia
| | - Ryan Galea
- UQ Diamantina Institute, University of Queensland, Brisbane, Australia
| | - Justin Large
- UQ Diamantina Institute, University of Queensland, Brisbane, Australia
| | - Hideo Yagita
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | - Raymond J. Steptoe
- UQ Diamantina Institute, University of Queensland, Brisbane, Australia
- * E-mail:
| |
Collapse
|
120
|
Li R, Lee J, Kim MS, Liu V, Moulik M, Li H, Yi Q, Xie A, Chen W, Yang L, Li Y, Tsai TH, Oka K, Chan L, Yechoor V. PD-L1-driven tolerance protects neurogenin3-induced islet neogenesis to reverse established type 1 diabetes in NOD mice. Diabetes 2015; 64:529-40. [PMID: 25332429 PMCID: PMC4303975 DOI: 10.2337/db13-1737] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A breakdown in self-tolerance underlies autoimmune destruction of β-cells and type 1 diabetes. A cure by restoring β-cell mass is limited by the availability of transplantable β-cells and the need for chronic immunosuppression. Evidence indicates that inhibiting costimulation through the PD-1/PD-L1 pathway is central to immune tolerance. We therefore tested whether induction of islet neogenesis in the liver, protected by PD-L1-driven tolerance, reverses diabetes in NOD mice. We demonstrated a robust induction of neo-islets in the liver of diabetic NOD mice by gene transfer of Neurogenin3, the islet-defining factor, along with betacellulin, an islet growth factor. These neo-islets expressed all the major pancreatic hormones and transcription factors. However, an enduring restoration of glucose-stimulated insulin secretion and euglycemia occurs only when tolerance is also induced by the targeted overexpression of PD-L1 in the neo-islets, which results in inhibition of proliferation and increased apoptosis of infiltrating CD4(+) T cells. Further analysis revealed an inhibition of cytokine production from lymphocytes isolated from the liver but not from the spleen of treated mice, indicating that treatment did not result in generalized immunosuppression. This treatment strategy leads to persistence of functional neo-islets that resist autoimmune destruction and consequently an enduring reversal of diabetes in NOD mice.
Collapse
Affiliation(s)
- Rongying Li
- Division of Diabetes, Endocrinology and Metabolism, Diabetes and Endocrinology Research Center, and Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Jeongkyung Lee
- Division of Diabetes, Endocrinology and Metabolism, Diabetes and Endocrinology Research Center, and Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Mi-sun Kim
- Division of Diabetes, Endocrinology and Metabolism, Diabetes and Endocrinology Research Center, and Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Victoria Liu
- Division of Diabetes, Endocrinology and Metabolism, Diabetes and Endocrinology Research Center, and Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Mousumi Moulik
- Division of Cardiology, Department of Pediatrics, University of Texas Medical School at Houston, Houston, TX
| | - Haiyan Li
- Department of Cancer Biology, Cleveland Clinic, Lerner Research Institute, Cleveland, OH
| | - Qing Yi
- Department of Cancer Biology, Cleveland Clinic, Lerner Research Institute, Cleveland, OH
| | - Aini Xie
- Division of Diabetes, Endocrinology and Metabolism, Diabetes and Endocrinology Research Center, and Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Wenhao Chen
- Division of Diabetes, Endocrinology and Metabolism, Diabetes and Endocrinology Research Center, and Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Lina Yang
- Division of Diabetes, Endocrinology and Metabolism, Diabetes and Endocrinology Research Center, and Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Yimin Li
- Division of Diabetes, Endocrinology and Metabolism, Diabetes and Endocrinology Research Center, and Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Tsung Huang Tsai
- Division of Diabetes, Endocrinology and Metabolism, Diabetes and Endocrinology Research Center, and Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Kazuhiro Oka
- Division of Diabetes, Endocrinology and Metabolism, Diabetes and Endocrinology Research Center, and Department of Medicine, Baylor College of Medicine, Houston, TX Division of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
| | - Lawrence Chan
- Division of Diabetes, Endocrinology and Metabolism, Diabetes and Endocrinology Research Center, and Department of Medicine, Baylor College of Medicine, Houston, TX Division of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
| | - Vijay Yechoor
- Division of Diabetes, Endocrinology and Metabolism, Diabetes and Endocrinology Research Center, and Department of Medicine, Baylor College of Medicine, Houston, TX Division of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
| |
Collapse
|
121
|
Ostrand-Rosenberg S, Horn LA, Haile ST. The programmed death-1 immune-suppressive pathway: barrier to antitumor immunity. THE JOURNAL OF IMMUNOLOGY 2015; 193:3835-41. [PMID: 25281753 DOI: 10.4049/jimmunol.1401572] [Citation(s) in RCA: 161] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Programmed death ligand 1 (PD-L1, also known as B7 homolog 1 or CD274) is a major obstacle to antitumor immunity because it tolerizes/anergizes tumor-reactive T cells by binding to its receptor programmed death-1 (CD279), renders tumor cells resistant to CD8(+) T cell- and FasL-mediated lysis, and tolerizes T cells by reverse signaling through T cell-expressed CD80. PD-L1 is abundant in the tumor microenvironment, where it is expressed by many malignant cells, as well as by immune cells and vascular endothelial cells. The critical role of PD-L1 in obstructing antitumor immunity has been demonstrated in multiple animal models and in recent clinical trials. This article reviews the mechanisms by which PD-L1 impairs antitumor immunity and discusses established and experimental strategies for maintaining T cell activation in the presence of PD-L1-expressing cells in the tumor microenvironment.
Collapse
Affiliation(s)
| | - Lucas A Horn
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, MD 21250
| | - Samuel T Haile
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, MD 21250
| |
Collapse
|
122
|
Deng R, Cassady K, Li X, Yao S, Zhang M, Racine J, Lin J, Chen L, Zeng D. B7H1/CD80 interaction augments PD-1-dependent T cell apoptosis and ameliorates graft-versus-host disease. THE JOURNAL OF IMMUNOLOGY 2014; 194:560-74. [PMID: 25488990 DOI: 10.4049/jimmunol.1402157] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Interactions of B7H1 (programmed death ligand 1 [PD-L1]) with its two ligands, PD-1 and CD80, on T cells play a pivotal role in controlling T cell activation, proliferation, anergy, and apoptosis. However, the interactions between the two pathways remain unknown. Using an alloimmune response model of graft-versus-host disease (GVHD), we report in this study that: 1) Comparison of proliferation and apoptosis of wild-type (WT) and PD-1(-/-)CD4(+) conventional T (Tcon) cells in WT and B7H1(-/-) recipients revealed that B7H1/CD80 interaction per se augments T cell proliferation, and this interaction augments T cell apoptosis mediated by B7H1/PD-1 interaction. This observation was recapitulated in an in vitro MLR assay. 2) Specific blockade of the B7H1/CD80 axis by anti-B7H1 mAb reduces WT-alloreactive Tcon cell proliferation, IL-2 production, expression of PD-1, and apoptosis, resulting in worsening GVHD. In contrast, specific blockade of B7H1/CD80 interaction reduces donor PD-1(-/-) Tcon cell proliferation without an impact on apoptosis, resulting in ameliorating GVHD. 3) B7H1 fused to an Ig Fc domain (B7H1-Ig), when produced in vivo by hydrodynamic injection of B7H1-Ig plasmid, ameliorates GVHD by augmenting proliferation and apoptosis of WT- alloreactive Tcon cells. Conversely, B7H1-Ig treatment has no impact on apoptosis but augments PD-1(-/-) T cell proliferation and worsens GVHD. These results indicate that B7H1/CD80 interaction augments Tcon cell proliferation, IL-2 production, and expression of PD-1, which leads to increased apoptosis mediated by the B7H1/PD-1 pathway. Additionally, by engaging both PD-1 and CD80, B7H1-Ig can be a powerful therapeutic reagent for downregulating the T cell immune response.
Collapse
Affiliation(s)
- Ruishu Deng
- Department of Diabetes Research, Beckman Research Institute, City of Hope, Duarte, CA 91010; Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope, Duarte, CA 91010
| | - Kaniel Cassady
- Department of Diabetes Research, Beckman Research Institute, City of Hope, Duarte, CA 91010; Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope, Duarte, CA 91010; Irell and Manella Graduate School of Biological Sciences, City of Hope, Duarte, CA 91010
| | - Xiaofan Li
- Department of Diabetes Research, Beckman Research Institute, City of Hope, Duarte, CA 91010; Department of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Sheng Yao
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520; and
| | - Mingfeng Zhang
- Department of Diabetes Research, Beckman Research Institute, City of Hope, Duarte, CA 91010; Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope, Duarte, CA 91010
| | - Jeremy Racine
- Department of Diabetes Research, Beckman Research Institute, City of Hope, Duarte, CA 91010; Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope, Duarte, CA 91010; Irell and Manella Graduate School of Biological Sciences, City of Hope, Duarte, CA 91010
| | - Jeffrey Lin
- Eugene and Ruth Roberts Summer Student Academy of City of Hope, Duarte, CA 91010
| | - Lieping Chen
- Department of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Defu Zeng
- Department of Diabetes Research, Beckman Research Institute, City of Hope, Duarte, CA 91010; Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope, Duarte, CA 91010; Irell and Manella Graduate School of Biological Sciences, City of Hope, Duarte, CA 91010;
| |
Collapse
|
123
|
Herbst RS, Soria JC, Kowanetz M, Fine GD, Hamid O, Gordon MS, Sosman JA, McDermott DF, Powderly JD, Gettinger SN, Kohrt HEK, Horn L, Lawrence DP, Rost S, Leabman M, Xiao Y, Mokatrin A, Koeppen H, Hegde PS, Mellman I, Chen DS, Hodi FS. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 2014; 515:563-7. [PMID: 25428504 PMCID: PMC4836193 DOI: 10.1038/nature14011] [Citation(s) in RCA: 4014] [Impact Index Per Article: 401.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 10/31/2014] [Indexed: 11/09/2022]
Abstract
The development of human cancer is a multistep process characterized by the accumulation of genetic and epigenetic alterations that drive or reflect tumour progression. These changes distinguish cancer cells from their normal counterparts, allowing tumours to be recognized as foreign by the immune system. However, tumours are rarely rejected spontaneously, reflecting their ability to maintain an immunosuppressive microenvironment. Programmed death-ligand 1 (PD-L1; also called B7-H1 or CD274), which is expressed on many cancer and immune cells, plays an important part in blocking the 'cancer immunity cycle' by binding programmed death-1 (PD-1) and B7.1 (CD80), both of which are negative regulators of T-lymphocyte activation. Binding of PD-L1 to its receptors suppresses T-cell migration, proliferation and secretion of cytotoxic mediators, and restricts tumour cell killing. The PD-L1-PD-1 axis protects the host from overactive T-effector cells not only in cancer but also during microbial infections. Blocking PD-L1 should therefore enhance anticancer immunity, but little is known about predictive factors of efficacy. This study was designed to evaluate the safety, activity and biomarkers of PD-L1 inhibition using the engineered humanized antibody MPDL3280A. Here we show that across multiple cancer types, responses (as evaluated by Response Evaluation Criteria in Solid Tumours, version 1.1) were observed in patients with tumours expressing high levels of PD-L1, especially when PD-L1 was expressed by tumour-infiltrating immune cells. Furthermore, responses were associated with T-helper type 1 (TH1) gene expression, CTLA4 expression and the absence of fractalkine (CX3CL1) in baseline tumour specimens. Together, these data suggest that MPDL3280A is most effective in patients in which pre-existing immunity is suppressed by PD-L1, and is re-invigorated on antibody treatment.
Collapse
Affiliation(s)
- Roy S Herbst
- Yale Comprehensive Cancer Center, Yale School of Medicine, 333 Cedar Street, WWW221, New Haven, Connecticut 06520, USA
| | - Jean-Charles Soria
- Gustave Roussy South-Paris University, 114 Rue Edouard Vaillant, 94805 Villefuij, Cedex, France
| | - Marcin Kowanetz
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, USA
| | - Gregg D Fine
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, USA
| | - Omid Hamid
- The Angeles Clinic and Research Institute, 11818 Wilshire Blvd, Los Angeles, California 90025, USA
| | - Michael S Gordon
- Pinnacle Oncology Hematology, 9055 E Del Camino Dr 100, Scottsdale, Arizona 85258, USA
| | - Jeffery A Sosman
- Vanderbilt-Ingram Cancer Center, 2220 Pierce Avenue, Nashville, Tennessee 37212, USA
| | - David F McDermott
- Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Shapiro 9, Boston, Massachusetts 02215, USA
| | - John D Powderly
- Carolina BioOncology Institute, 9801 W. Kincey Ave, Suite 145, Huntersville, North Carolina 28078, USA
| | - Scott N Gettinger
- Yale Comprehensive Cancer Center, Yale School of Medicine, 333 Cedar Street, WWW221, New Haven, Connecticut 06520, USA
| | - Holbrook E K Kohrt
- Stanford University, CCSR Bldg Room 1110, Stanford, California 94305, USA
| | - Leora Horn
- Vanderbilt-Ingram Cancer Center, 1301 Medical Center Dr, Suite 1710, Nashville, Tennessee 37212, USA
| | - Donald P Lawrence
- Massachusetts General Hospital, 55 Fruit Street, YAW 9E, Boston, Massachusetts 02114, USA
| | - Sandra Rost
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, USA
| | - Maya Leabman
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, USA
| | - Yuanyuan Xiao
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, USA
| | - Ahmad Mokatrin
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, USA
| | - Hartmut Koeppen
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, USA
| | - Priti S Hegde
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, USA
| | - Ira Mellman
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, USA
| | - Daniel S Chen
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, USA
| | - F Stephen Hodi
- Dana-Farber/Brigham and Women's Cancer Center, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
| |
Collapse
|
124
|
Ellestad KK, Thangavelu G, Ewen CL, Boon L, Anderson CC. PD-1 is not required for natural or peripherally induced regulatory T cells: Severe autoimmunity despite normal production of regulatory T cells. Eur J Immunol 2014; 44:3560-72. [DOI: 10.1002/eji.201444688] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 08/06/2014] [Accepted: 09/16/2014] [Indexed: 12/15/2022]
Affiliation(s)
- Kristofor K. Ellestad
- Department of Medical Microbiology and Immunology; University of Alberta; Edmonton AB Canada
- Alberta Diabetes and Transplant Institutes; University of Alberta; Edmonton AB Canada
| | - Govindarajan Thangavelu
- Alberta Diabetes and Transplant Institutes; University of Alberta; Edmonton AB Canada
- Department of Surgery; University of Alberta; Edmonton AB Canada
| | - Catherine L. Ewen
- Alberta Diabetes and Transplant Institutes; University of Alberta; Edmonton AB Canada
| | | | - Colin C. Anderson
- Department of Medical Microbiology and Immunology; University of Alberta; Edmonton AB Canada
- Alberta Diabetes and Transplant Institutes; University of Alberta; Edmonton AB Canada
- Department of Surgery; University of Alberta; Edmonton AB Canada
| |
Collapse
|
125
|
The perspective of immunotherapy: new molecules and new mechanisms of action in immune modulation. Curr Opin Oncol 2014; 26:204-14. [PMID: 24424272 DOI: 10.1097/cco.0000000000000054] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
PURPOSE OF REVIEW Targeting CTLA-4, the patriarch of immune checkpoint modulators, is currently the only immunotherapeutic approach that has achieved significant clinical benefit in melanoma phase III trials. In this review, recent new ideas about the mechanism of action of anti-CTLA antibodies, other new molecules to target, and rationales for combination therapies will be discussed. RECENT FINDINGS Although the clinical efficacy of the anti-CTLA-4 monoclonal antibody (mAb) ipilimumab is meanwhile without doubt, its mechanism of action is still not fully understood. Recent data indicate that, besides modulation of the TCR signal, CTLA-4 mAbs can mediate regulatory T-cell depletion in an Fc gamma receptor dependent manner.Blockade of the molecules PD-1 and PD-L1 has given promising clinical responses (and this beyond melanoma), whereas their complex expression and interaction pattern makes a clear statement about the mechanism of action challenging.Additional other co-inhibitory and co-stimulatory molecules have been identified recently. Combinations of immune checkpoint modulators themselves or with other therapies, such as adoptive cell therapy, targeted therapy or radiotherapy, will improve the outcomes further. SUMMARY Immune checkpoint blockade is currently the most promising systemic therapeutic approach to achieve long-lasting responses or even cure in melanoma and other malignancies.
Collapse
|
126
|
Ikebuchi R, Konnai S, Okagawa T, Yokoyama K, Nakajima C, Suzuki Y, Murata S, Ohashi K. Influence of PD-L1 cross-linking on cell death in PD-L1-expressing cell lines and bovine lymphocytes. Immunology 2014; 142:551-61. [PMID: 24405267 DOI: 10.1111/imm.12243] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 12/27/2013] [Accepted: 01/06/2014] [Indexed: 11/30/2022] Open
Abstract
Programmed death-ligand 1 (PD-L1) blockade is accepted as a novel strategy for the reactivation of exhausted T cells that express programmed death-1 (PD-1). However, the mechanism of PD-L1-mediated inhibitory signalling after PD-L1 cross-linking by anti-PD-L1 monoclonal antibody (mAb) or PD-1-immunogloblin fusion protein (PD-1-Ig) is still unknown, although it may induce cell death of PD-L1(+) cells required for regular immune reactions. In this study, PD-1-Ig or anti-PD-L1 mAb treatment was tested in cell lines that expressed PD-L1 and bovine lymphocytes to investigate whether the treatment induces immune reactivation or PD-L1-mediated cell death. PD-L1 cross-linking by PD-1-Ig or anti-PD-L1 mAb primarily increased the number of dead cells in PD-L1(high) cells, but not in PD-L1(low) cells; these cells were prepared from Cos-7 cells in which bovine PD-L1 expression was induced by transfection. The PD-L1-mediated cell death also occurred in Cos-7 and HeLa cells transfected with vectors only encoding the extracellular region of PD-L1. In bovine lymphocytes, the anti-PD-L1 mAb treatment up-regulated interferon-γ (IFN-γ) production, whereas PD-1-Ig treatment decreased this cytokine production and cell proliferation. The IFN-γ production in B-cell-depleted peripheral blood mononuclear cells was not reduced by PD-1-Ig treatment and the percentages of dead cells in PD-L1(+) B cells were increased by PD-1-Ig treatment, indicating that PD-1-Ig-induced immunosuppression in bovine lymphocytes could be caused by PD-L1-mediated B-cell death. This study provides novel information for the understanding of signalling through PD-L1.
Collapse
Affiliation(s)
- Ryoyo Ikebuchi
- Department of Disease Control, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
127
|
Dai S, Jia R, Zhang X, Fang Q, Huang L. The PD-1/PD-Ls pathway and autoimmune diseases. Cell Immunol 2014; 290:72-9. [PMID: 24908630 DOI: 10.1016/j.cellimm.2014.05.006] [Citation(s) in RCA: 270] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 04/17/2014] [Accepted: 05/07/2014] [Indexed: 12/27/2022]
Abstract
The programmed death (PD)-1/PD-1 ligands (PD-Ls) pathway, is a new member of the B7/CD28 family, and consists of the PD-1 receptor and its ligands PD-L1 (B7-H1, CD274) and PD-L2 (B7-DC, CD273). Recently, it is reported that PD-1, PD-L1 and PD-L2 also have soluble forms aside from their membrane bound forms. The soluble forms increase the diversity and complexity of PD-1/PD-Ls pathway in both composition and function. The PD-1/PD-Ls pathway is broadly expressed and exerts a wider range of immunoregulatory roles in T-cell activation and tolerance compared with other B7/CD28 family members. Studies show that the PD-1/PD-Ls pathway regulates the induction and maintenance of peripheral tolerance and protects tissues from autoimmune attack in physiological conditions. In addition, it is also involved in various diseases mediated by T cells, such as autoimmunity, tumor immunity, chronic viral infections, and transplantation immunity. In this review, we will summarize the relevance of the soluble forms and the latest researches on the role of PD-1/PD-Ls pathway in autoimmune diseases.
Collapse
Affiliation(s)
- Suya Dai
- Department of Laboratory Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, People's Republic of China
| | - Ru Jia
- Department of Laboratory Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, People's Republic of China
| | - Xiao Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, People's Republic of China
| | - Qiwen Fang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, People's Republic of China
| | - Lijuan Huang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, People's Republic of China.
| |
Collapse
|
128
|
Cooper ZA, Juneja VR, Sage PT, Frederick DT, Piris A, Mitra D, Lo JA, Hodi FS, Freeman GJ, Bosenberg MW, McMahon M, Flaherty KT, Fisher DE, Sharpe AH, Wargo JA. Response to BRAF inhibition in melanoma is enhanced when combined with immune checkpoint blockade. Cancer Immunol Res 2014; 2:643-54. [PMID: 24903021 DOI: 10.1158/2326-6066.cir-13-0215] [Citation(s) in RCA: 207] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BRAF-targeted therapy results in objective responses in the majority of patients; however, the responses are short lived (∼6 months). In contrast, treatment with immune checkpoint inhibitors results in a lower response rate, but the responses tend to be more durable. BRAF inhibition results in a more favorable tumor microenvironment in patients, with an increase in CD8(+) T-cell infiltrate and a decrease in immunosuppressive cytokines. There is also increased expression of the immunomodulatory molecule PDL1, which may contribute to the resistance. On the basis of these findings, we hypothesized that BRAF-targeted therapy may synergize with the PD1 pathway blockade to enhance antitumor immunity. To test this hypothesis, we developed a BRAF(V600E)/Pten(-/-) syngeneic tumor graft immunocompetent mouse model in which BRAF inhibition leads to a significant increase in the intratumoral CD8(+) T-cell density and cytokine production, similar to the effects of BRAF inhibition in patients. In this model, CD8(+) T cells were found to play a critical role in the therapeutic effect of BRAF inhibition. Administration of anti-PD1 or anti-PDL1 together with a BRAF inhibitor led to an enhanced response, significantly prolonging survival and slowing tumor growth, as well as significantly increasing the number and activity of tumor-infiltrating lymphocytes. These results demonstrate synergy between combined BRAF-targeted therapy and immune checkpoint blockade. Although clinical trials combining these two strategies are ongoing, important questions still remain unanswered. Further studies using this new melanoma mouse model may provide therapeutic insights, including optimal timing and sequence of therapy.
Collapse
Affiliation(s)
- Zachary A Cooper
- Authors' Affiliations: Departments of Surgical Oncology and Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Vikram R Juneja
- Harvard-MIT Division of Health Sciences and Technology, Cambridge; Department of Microbiology and Immunobiology; Harvard Medical School; Divisions of
| | - Peter T Sage
- Department of Microbiology and Immunobiology; Harvard Medical School; Divisions of
| | | | - Adriano Piris
- Harvard Medical School; Divisions of Dermatopathology, and
| | | | | | - F Stephen Hodi
- Harvard Medical School; Divisions of Department of Medical Oncology, Dana-Farber Cancer Institute
| | - Gordon J Freeman
- Harvard Medical School; Divisions of Department of Medical Oncology, Dana-Farber Cancer Institute
| | - Marcus W Bosenberg
- Department of Dermatology, Yale University School of Medicine, New Haven, Connecticut
| | - Martin McMahon
- Helen Diller Family Comprehensive Cancer Center; and Department of Cell and Molecular Pharmacology, University of California San Francisco, San Francisco, California
| | | | - David E Fisher
- Harvard Medical School; Divisions of Dermatology, Massachusetts General Hospital
| | - Arlene H Sharpe
- Department of Microbiology and Immunobiology; Harvard Medical School; Divisions of Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Jennifer A Wargo
- Authors' Affiliations: Departments of Surgical Oncology and Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas;
| |
Collapse
|
129
|
Kochupurakkal NM, Kruger AJ, Tripathi S, Zhu B, Adams LT, Rainbow DB, Rossini A, Greiner DL, Sayegh MH, Wicker LS, Guleria I. Blockade of the programmed death-1 (PD1) pathway undermines potent genetic protection from type 1 diabetes. PLoS One 2014; 9:e89561. [PMID: 24586872 PMCID: PMC3938467 DOI: 10.1371/journal.pone.0089561] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 01/22/2014] [Indexed: 01/07/2023] Open
Abstract
Aims/Hypothesis Inhibition of PD1-PDL1 signaling in NOD mice accelerates onset of type 1 diabetes implicating this pathway in suppressing the emergence of pancreatic beta cell reactive T-cells. However, the molecular mechanism by which PD1 signaling protects from type 1 diabetes is not clear. We hypothesized that differential susceptibility of Idd mouse strains to type 1 diabetes when challenged with anti PDL1 will identify genomic loci that collaborate with PD1 signaling in suppressing type 1 diabetes. Methods Anti PDL1 was administered to NOD and various Idd mouse strains at 10 weeks of age and onset of disease was monitored by measuring blood glucose levels. Additionally, histological evaluation of the pancreas was performed to determine degree of insulitis. Statistical analysis of the data was performed using Log-Rank and Student's t-test. Results Blockade of PDL1 rapidly precipitated type 1 diabetes in nearly all NOD Idd congenic strains tested, despite the fact that all are moderately (Idd5, Idd3 and Idd10/18) or highly (Idd3/10/18 and Idd9) protected from spontaneous type 1 diabetes by virtue of their protective Idd genes. Only the Idd3/5 strain, which is nearly 100% protected from spontaneous disease, remained normoglycemic following PDL1 blockade. Conclusions These results indicate that multiple Idd loci collaborate with PD1 signaling. Anti PDL1 treatment undermines a large portion of the genetic protection mediated by Idd genes in the NOD model of type 1 diabetes. Basal insulitis correlated with higher susceptibility to type 1 diabetes. These findings have important implications since the PD1 pathway is a target for immunotherapy.
Collapse
Affiliation(s)
- Nora M. Kochupurakkal
- Transplantation Research Center, Brigham and Women's Hospital and Children's Hospital Boston, Harvard Medical School Renal Division, Boston, Massachusetts, United States of America
| | - Annie J. Kruger
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Sudipta Tripathi
- Transplantation Research Center, Brigham and Women's Hospital and Children's Hospital Boston, Harvard Medical School Renal Division, Boston, Massachusetts, United States of America
| | - Bing Zhu
- Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - La Tonya Adams
- Transplantation Research Center, Brigham and Women's Hospital and Children's Hospital Boston, Harvard Medical School Renal Division, Boston, Massachusetts, United States of America
| | - Daniel B. Rainbow
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Aldo Rossini
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Dale L. Greiner
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Mohamed H. Sayegh
- Transplantation Research Center, Brigham and Women's Hospital and Children's Hospital Boston, Harvard Medical School Renal Division, Boston, Massachusetts, United States of America
| | - Linda S. Wicker
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Indira Guleria
- Transplantation Research Center, Brigham and Women's Hospital and Children's Hospital Boston, Harvard Medical School Renal Division, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
130
|
Mott KR, Allen SJ, Zandian M, Akbari O, Hamrah P, Maazi H, Wechsler SL, Sharpe AH, Freeman GJ, Ghiasi H. Inclusion of CD80 in HSV targets the recombinant virus to PD-L1 on DCs and allows productive infection and robust immune responses. PLoS One 2014; 9:e87617. [PMID: 24475315 PMCID: PMC3903765 DOI: 10.1371/journal.pone.0087617] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 12/20/2013] [Indexed: 12/31/2022] Open
Abstract
CD80 plays a critical role in stimulation of T cells and subsequent control of infection. To investigate the effect of CD80 on HSV-1 infection, we constructed a recombinant HSV-1 virus that expresses two copies of the CD80 gene in place of the latency associated transcript (LAT). This mutant virus (HSV-CD80) expressed high levels of CD80 and had similar virus replication kinetics as control viruses in rabbit skin cells. In contrast to parental virus, this CD80 expressing recombinant virus replicated efficiently in immature dendritic cells (DCs). Additionally, the susceptibility of immature DCs to HSV-CD80 infection was mediated by CD80 binding to PD-L1 on DCs. This interaction also contributed to a significant increase in T cell activation. Taken together, these results suggest that inclusion of CD80 as a vaccine adjuvant may promote increased vaccine efficacy by enhancing the immune response directly and also indirectly by targeting to DC.
Collapse
Affiliation(s)
- Kevin R. Mott
- Center for Neurobiology and Vaccine Development, Ophthalmology Research, Department of Surgery, Cedars-Sinai Burns & Allen Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Sariah J. Allen
- Center for Neurobiology and Vaccine Development, Ophthalmology Research, Department of Surgery, Cedars-Sinai Burns & Allen Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Mandana Zandian
- Center for Neurobiology and Vaccine Development, Ophthalmology Research, Department of Surgery, Cedars-Sinai Burns & Allen Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Omid Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Pedram Hamrah
- Massachusetts Eye & Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Hadi Maazi
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Steven L. Wechsler
- Gavin Herbert Eye Institute, the Department of Ophthalmology, the Department of Microbiology and Molecular Genetics, and the Center for Virus Research, University of California Irvine, School of Medicine, Irvine, California, United States of America
| | - Arlene H. Sharpe
- Department of Pathology, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Gordon J. Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Homayon Ghiasi
- Center for Neurobiology and Vaccine Development, Ophthalmology Research, Department of Surgery, Cedars-Sinai Burns & Allen Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
131
|
Fankhauser SC, Starnbach MN. PD-L1 limits the mucosal CD8+ T cell response to Chlamydia trachomatis. THE JOURNAL OF IMMUNOLOGY 2013; 192:1079-90. [PMID: 24353266 DOI: 10.4049/jimmunol.1301657] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Chlamydia trachomatis infection is the most common bacterial sexually transmitted disease in the United States. Repeated infections with C. trachomatis lead to serious sequelae, such as infertility. It is unclear why the adaptive immune system, specifically the CD8(+) T cell response, is unable to protect against subsequent C. trachomatis infections. In this article, we characterize the mucosal CD8(+) T cell response to C. trachomatis in the murine genital tract. We demonstrate that the immunoinhibitory ligand, PD-L1, contributes to the defective CD8(+) T cell response. Deletion or inhibition of PD-L1 restores the CD8(+) T cell response and enhances C. trachomatis clearance.
Collapse
Affiliation(s)
- Sarah C Fankhauser
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115
| | | |
Collapse
|
132
|
Tang PA, Heng DYC. Programmed death 1 pathway inhibition in metastatic renal cell cancer and prostate cancer. Curr Oncol Rep 2013; 15:98-104. [PMID: 23263823 DOI: 10.1007/s11912-012-0284-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Programmed death 1 (PD-1) is a T cell co-inhibitory receptor with two ligands, PD-L1 and PD-L2. In cancer, this pathway plays a major role in immune resistance in the tumor environment. Blockade of this pathway can enhance antitumor immune responses. This review discusses the preclinical rationale for PD-1 pathway inhibition in advanced renal cell carcinoma and prostate cancer, in addition to the clinical activity and toxicity of the anti-PD-L1 antibody BMS-936559, as well as anti-PD-1 antibodies MK-3475 and BMS-936558.
Collapse
Affiliation(s)
- Patricia A Tang
- Tom Baker Cancer Center, University of Calgary, Calgary, AB, Canada
| | | |
Collapse
|
133
|
Mukherjee G, Geliebter A, Babad J, Santamaria P, Serreze DV, Freeman GJ, Tarbell KV, Sharpe A, DiLorenzo TP. DEC-205-mediated antigen targeting to steady-state dendritic cells induces deletion of diabetogenic CD8⁺ T cells independently of PD-1 and PD-L1. Int Immunol 2013; 25:651-60. [PMID: 24021877 DOI: 10.1093/intimm/dxt031] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
CD8⁺ T cells specific for islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP) have been implicated in type 1 diabetes in both humans and non-obese diabetic (NOD) mice, in which T cells specific for IGRP₂₀₆₋₂₁₄ are highly prevalent. We sought to manipulate these pathogenic T cells by exploiting the ability of steady-state dendritic cells (DCs) to present antigens in a tolerogenic manner. The endocytic receptor DEC-205 was utilized to deliver an IGRP₂₀₆₋₂₁₄ mimotope to DCs in NOD mice, and the impact of this delivery on a polyclonal population of endogenous islet-reactive cognate T cells was determined. Assessment of islet-infiltrating CD8⁺ T cells showed a decrease in the percentage, and the absolute number, of endogenous IGRP₂₀₆₋₂₁₄-specific T cells when the mimotope was delivered to DCs, compared with delivery of a specificity control. Employing an adoptive transfer system, deletion of CD8⁺ T cells as a result of DEC-205-mediated antigen targeting was found to occur independently of programmed death-1 (PD-1) and its ligand (PD-L1), both often implicated in the regulation of peripheral T-cell tolerance. Given its promise for the manipulation of self-reactive polyclonal T cells demonstrated here, the distinctive characteristics of this antigen delivery system will be important to appreciate as its potential as an intervention for autoimmune diseases continues to be investigated.
Collapse
Affiliation(s)
- Gayatri Mukherjee
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
134
|
A missing PD-L1/PD-1 coinhibition regulates diabetes induction by preproinsulin-specific CD8 T-cells in an epitope-specific manner. PLoS One 2013; 8:e71746. [PMID: 23977133 PMCID: PMC3747217 DOI: 10.1371/journal.pone.0071746] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Accepted: 07/02/2013] [Indexed: 11/19/2022] Open
Abstract
Coinhibitory PD-1/PD-L1 (B7-H1) interactions provide critical signals for the regulation of autoreactive T-cell responses. We established mouse models, expressing the costimulator molecule B7.1 (CD80) on pancreatic beta cells (RIP-B7.1 tg mice) or are deficient in coinhibitory PD-L1 or PD-1 molecules (PD-L1−/− and PD-1−/− mice), to study induction of preproinsulin (ppins)-specific CD8 T-cell responses and experimental autoimmune diabetes (EAD) by DNA-based immunization. RIP-B7.1 tg mice allowed us to identify two CD8 T-cell specificities: pCI/ppins DNA exclusively induced Kb/A12–21-specific CD8 T-cells and EAD, whereas pCI/ppinsΔA12–21 DNA (encoding ppins without the COOH-terminal A12–21 epitope) elicited Kb/B22–29-specific CD8 T-cells and EAD. Specific expression/processing of mutant ppinsΔA12–21 (but not ppins) in non-beta cells, targeted by intramuscular DNA-injection, thus facilitated induction of Kb/B22–29-specific CD8 T-cells. The A12–21 epitope binds Kb molecules with a very low avidity as compared with B22–29. Interestingly, immunization of coinhibition-deficient PD-L1−/− or PD-1−/− mice with pCI/ppins induced Kb/A12–21-monospecific CD8 T-cells and EAD but injections with pCI/ppinsΔA12–21 did neither recruit Kb/B22–29-specific CD8 T-cells into the pancreatic target tissue nor induce EAD. PpinsΔA12–21/(Kb/B22–29)-mediated EAD was efficiently restored in RIP-B7.1+/PD-L1−/− mice, differing from PD-L1−/− mice only in the tg B7.1 expression in beta cells. Alternatively, an ongoing beta cell destruction and tissue inflammation, initiated by ppins/(Kb/A12–21)-specific CD8 T-cells in pCI/ppins+pCI/ppinsΔA12–21 co-immunized PD-L1−/− mice, facilitated the expansion of ppinsΔA12–21/(Kb/B22–29)-specific CD8 T-cells. CD8 T-cells specific for the high-affinity Kb/B22–29- (but not the low-affinity Kb/A12–21)-epitope thus require stimulatory ´help from beta cells or inflamed islets to expand in PD-L1-deficient mice. The new PD-1/PD-L1 diabetes models may be valuable tools to study under well controlled experimental conditions distinct hierarchies of autoreactive CD8 T-cell responses, which trigger the initial steps of beta cell destruction or emerge during the pathogenic progression of EAD.
Collapse
|
135
|
Pauken KE, Jenkins MK, Azuma M, Fife BT. PD-1, but not PD-L1, expressed by islet-reactive CD4+ T cells suppresses infiltration of the pancreas during type 1 diabetes. Diabetes 2013; 62:2859-69. [PMID: 23545706 PMCID: PMC3717847 DOI: 10.2337/db12-1475] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The inhibitory receptor programmed death-1 (PD-1) constrains type 1 diabetes (T1D) in the nonobese diabetic (NOD) mouse. However, how PD-1 influences diabetogenic CD4(+) T cells during natural diabetes is not fully understood. To address this question, we developed a novel model to investigate antigen-specific CD4(+) T cells under physiological conditions in vivo. We transferred a low number of naïve CD4(+) T cells from the BDC2.5 mouse into prediabetic NOD mice to mimic a physiological precursor frequency and allowed the cells to become primed by endogenous autoantigen. Transferred BDC2.5 T cells became activated, differentiated into T-bet(+) IFN-γ-producing cells, and infiltrated the pancreas. In this model, loss of PD-1, but not programmed death ligand-1 (PD-L1), on the antigen-specific CD4(+) T cell resulted in increased cell numbers in the spleen, pancreas-draining lymph node, and pancreas. PD-1 deficiency also increased expression of the chemokine receptor CXCR3. Lastly, histological data showed that a loss of PD-1 caused BDC2.5 cells to penetrate deep into the islet core, resulting in conversion from peri-insulitis to destructive insulitis. These data support a model by which PD-1 regulates islet-reactive CD4(+) T cells in a cell intrinsic manner by suppressing proliferation, inhibiting infiltration of the pancreas, and limiting diabetes.
Collapse
Affiliation(s)
- Kristen E. Pauken
- Department of Medicine, Center for Immunology, University of Minnesota, Minneapolis, Minnesota
| | - Marc K. Jenkins
- Department of Microbiology, Center for Immunology, University of Minnesota, Minneapolis, Minnesota
| | - Miyuki Azuma
- Department of Molecular Immunology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Brian T. Fife
- Department of Medicine, Center for Immunology, University of Minnesota, Minneapolis, Minnesota
- Corresponding author: Brian T. Fife,
| |
Collapse
|
136
|
HHLA2 is a member of the B7 family and inhibits human CD4 and CD8 T-cell function. Proc Natl Acad Sci U S A 2013; 110:9879-84. [PMID: 23716685 DOI: 10.1073/pnas.1303524110] [Citation(s) in RCA: 155] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
T-cell costimulation and coinhibition generated by engagement of the B7 family and their receptor CD28 family are of central importance in regulating the T-cell response, making these pathways very attractive therapeutic targets. Here we describe HERV-H LTR-associating protein 2 (HHLA2) as a member of the B7 family that shares 10-18% amino acid identity and 23-33% similarity to other human B7 proteins and phylogenetically forms a subfamily with B7x and B7-H3 within the family. HHLA2 is expressed in humans but not in mice, which is unique within the B7 and CD28 families. HHLA2 protein is constitutively expressed on the surface of human monocytes and is induced on B cells after stimulation with LPS and IFN-γ. HHLA2 does not interact with other known members of the CD28 family or the B7 family, but does bind a putative receptor that is constitutively expressed not only on resting and activated CD4 and CD8 T cells but also on antigen-presenting cells. HHLA2 inhibits proliferation of both CD4 and CD8 T cells in the presence of T-cell receptor signaling. In addition, HHLA2 significantly reduces cytokine production by T cells including IFN-γ, TNF-α, IL-5, IL-10, IL-13, IL-17A, and IL-22. Thus, we have identified a unique B7 pathway that is able to inhibit human CD4 and CD8 T-cell proliferation and cytokine production. This unique human T-cell coinhibitory pathway may afford unique strategies for the treatment of human cancers, autoimmune disorders, infection, and transplant rejection and may help to design better vaccines.
Collapse
|
137
|
Intlekofer AM, Thompson CB. At the bench: preclinical rationale for CTLA-4 and PD-1 blockade as cancer immunotherapy. J Leukoc Biol 2013; 94:25-39. [PMID: 23625198 DOI: 10.1189/jlb.1212621] [Citation(s) in RCA: 278] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Tumors can avoid immune surveillance by stimulating immune inhibitory receptors that function to turn off established immune responses. By blocking the ability of tumors to stimulate inhibitory receptors on T cells, sustained, anti-tumor immune responses can be generated in animals. Thus, therapeutic blockade of immune inhibitory checkpoints provides a potential method to boost anti-tumor immunity. The CTLA-4 and PD-1Rs represent two T cell-inhibitory receptors with independent mechanisms of action. Preclinical investigations revealed that CTLA-4 enforces an activation threshold and attenuates proliferation of tumor-specific T lymphocytes. In contrast, PD-1 functions primarily as a stop signal that limits T cell effector function within a tumor. The unique mechanisms and sites of action of CTLA-4 and PD-1 suggest that although blockade of either has the potential to promote anti-tumor immune responses, combined blockade of both might offer even more potent anti-tumor activity. See related review At the Bedside: CTLA-4 and PD-1 blocking antibodies in cancer immunotherapy.
Collapse
Affiliation(s)
- Andrew M Intlekofer
- Department of Medicine, Cancer Biology and Genetics Program, and Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
| | | |
Collapse
|
138
|
Elhag OAO, Hu XJ, Wen-Ying Z, Li X, Yuan YZ, Deng LF, Liu DL, Liu YL, Hui G. Reconstructed adeno-associated virus with the extracellular domain of murine PD-1 induces antitumor immunity. Asian Pac J Cancer Prev 2013; 13:4031-6. [PMID: 23098512 DOI: 10.7314/apjcp.2012.13.8.4031] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The negative signaling provided by interactions of the co-inhibitory molecule, programmed death-1 (PD-1), and its ligands, B7-H1 (PD-L1) and B7-DC (PD-L2), is a critical mechanism contributing to tumor evasion; blockade of this pathway has been proven to enhance cytotoxic activity and mediate antitumor therapy. Here we evaluated the anti-tumor efficacy of AAV-mediated delivery of the extracellular domain of murine PD-1 (sPD-1) to a tumor site. MATERIAL AND METHODS An rAAV vector was constructed in which the expression of sPD-1, a known negative regulator of TCR signals, is driven by human cytomegalovirus immediate early promoter (CMV-P), using a triple plasmid transfection system. Tumor-bearing mice were then treated with the AAV/sPD1 construct and expression of sPD-1 in tumor tissues was determined by semi quantitative RT-PCR, and tumor weights and cytotoxic activity of splenocytes were measured. RESULTS Analysis of tumor homogenates revealed sPD-1 mRNA to be significantly overexpressed in rAAV/sPD-1 treated mice as compared with control levels. Its use for local gene therapy at the inoculation site of H22 hepatoma cells could inhibit tumor growth, also enhancing lysis of tumor cells by lymphocytes stimulated specifically with an antigen. In addition, PD-1 was also found expressed on the surfaces of activated CD8+ T cells. CONCLUSION This study confirmed that expression of the soluble extracellular domain of PD-1 molecule could reduce tumor microenvironment inhibitory effects on T cells and enhance cytotoxicity. This suggests that it might be a potential target for development of therapies to augment T-cell responses in patients with malignancies.
Collapse
Affiliation(s)
- Osama A O Elhag
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, College of Life Sciences, Central China Normal University, Hubei, China
| | | | | | | | | | | | | | | | | |
Collapse
|
139
|
Manipulating the PD-1 pathway to improve immunity. Curr Opin Immunol 2013; 25:381-8. [PMID: 23582509 DOI: 10.1016/j.coi.2013.03.003] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 03/05/2013] [Indexed: 12/13/2022]
Abstract
PD-1 is an inhibitory receptor induced in T cells by antigen stimulation and sustained PD-1 expression plays a key role in T cell dysfunction. Blocking PD-1 signaling rescues exhausted T cells and is an effective treatment for chronic infections and cancer. Nonetheless, combining PD-1 pathway blockade to therapeutic vaccination should further improve T cell rescue. PD-1 is induced shortly after T cell priming, but little is known about the role of PD-1 in the initiation of immune responses. In addition, the PD-1 pathway may also modulate humoral responses, since both B cells and Tfh cells express PD-1. Therefore, even though much progress has been achieved by manipulation of the PD-1 pathway to rescue exhausted T cells, this powerful immunotherapy could still be further exploited.
Collapse
|
140
|
Abstract
Co-stimulatory and co-inhibitory receptors have a pivotal role in T cell biology, as they determine the functional outcome of T cell receptor (TCR) signalling. The classic definition of T cell co-stimulation continues to evolve through the identification of new co-stimulatory and co-inhibitory receptors, the biochemical characterization of their downstream signalling events and the delineation of their immunological functions. Notably, it has been recently appreciated that co-stimulatory and co-inhibitory receptors display great diversity in expression, structure and function, and that their functions are largely context dependent. Here, we focus on some of these emerging concepts and review the mechanisms through which T cell activation, differentiation and function is controlled by co-stimulatory and co-inhibitory receptors.
Collapse
Affiliation(s)
- Lieping Chen
- Department of Immunobiology and Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut 06519, USA.
| | | |
Collapse
|
141
|
Kulpa DA, Lawani M, Cooper A, Peretz Y, Ahlers J, Sékaly RP. PD-1 coinhibitory signals: the link between pathogenesis and protection. Semin Immunol 2013; 25:219-27. [PMID: 23548749 DOI: 10.1016/j.smim.2013.02.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 02/15/2013] [Indexed: 12/31/2022]
Abstract
In the majority of HIV-1 infected individuals, the adaptive immune response drives virus escape resulting in persistent viremia and a lack of immune-mediated control. The expression of negative regulatory molecules such as PD-1 during chronic HIV infection provides a useful marker to differentiate functional memory T cell subsets and the frequency of T cells with an exhausted phenotype. In addition, cell-based measurements of virus persistence equate with activation markers and the frequency of CD4 T cells expressing PD-1. High-level expression of PD-1 and its ligands PD-L1 and PD-L2 are found on hematopoietic and non-hematopoietic cells, and are upregulated by chronic antigen stimulation, Type 1 and Type II interferons (IFNs), and homeostatic cytokines. In HIV infected subjects, PD-1 levels on CD4 and CD8 T cells continue to remain high following combination anti-retroviral therapy (cART). System biology approaches have begun to elucidate signal transduction pathways regulated by PD-1 expression in CD4 and CD8 T cell subsets that become dysfunctional through chronic TCR activation and PD-1 signaling. In this review, we summarize our current understanding of transcriptional signatures and signal transduction pathways associated with immune exhaustion with a focus on recent work in our laboratory characterizing the role of PD-1 in T cell dysfunction and HIV pathogenesis. We also highlight the therapeutic potential of blocking PD-1-PD-L1 and other immune checkpoints for activating potent cellular immune responses against chronic viral infections and cancer.
Collapse
Affiliation(s)
- Deanna A Kulpa
- Division of Infectious Diseases, Vaccine and Gene Therapy Institute-Florida (VGTI-FL), Port Saint Lucie, FL, United States
| | | | | | | | | | | |
Collapse
|
142
|
Villaflor VM, Salgia R. Targeted agents in non-small cell lung cancer therapy: What is there on the horizon? J Carcinog 2013; 12:7. [PMID: 23599689 PMCID: PMC3622362 DOI: 10.4103/1477-3163.109253] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Accepted: 01/10/2013] [Indexed: 02/06/2023] Open
Abstract
Lung cancer is a heterogeneous group of diseases. There has been much research in lung cancer over the past decade which has advanced our ability to treat these patients with a more personalized approach. The scope of this paper is to review the literature and give a broad understanding of the current molecular targets for which we currently have therapies as well as other targets for which we may soon have therapies. Additionally, we will cover some of the issues of resistance with these targeted therapies. The molecular targets we intend to discuss are epidermal growth factor receptor (EGFR), Vascular endothelial growth factor (VEGF), anaplastic large-cell lymphoma kinase (ALK), KRAS, C-MET/RON, PIK3CA. ROS-1, RET Fibroblast growth factor receptor (FGFR). Ephrins and their receptors, BRAF, and immunotherapies/vaccines. This manuscript only summarizes the work which has been done to date and in no way is meant to be comprehensive.
Collapse
Affiliation(s)
- Victoria M Villaflor
- Department of Medicine, Section of Hematology/Oncology University of Chicago, Chicago, IL, USA
| | | |
Collapse
|
143
|
Lack of PD-L1 expression by iNKT cells improves the course of influenza A infection. PLoS One 2013; 8:e59599. [PMID: 23555047 PMCID: PMC3598698 DOI: 10.1371/journal.pone.0059599] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Accepted: 02/19/2013] [Indexed: 01/12/2023] Open
Abstract
There is evidence indicating that invariant Natural Killer T (iNKT) cells play an important role in defense against influenza A virus (IAV). However, the effect of inhibitory receptor, programmed death-1 (PD-1), and its ligands, programmed death ligand (PD-L) 1 and 2 on iNKT cells in protection against IAV remains to be elucidated. Here we investigated the effects of these co-stimulatory molecules on iNKT cells in the response to influenza. We discovered that compare to the wild type, PD-L1 deficient mice show reduced sensitivity to IAV infection as evident by reduced weight loss, decreased pulmonary inflammation and cellular infiltration. In contrast, PD-L2 deficient mice showed augmented weight loss, pulmonary inflammation and cellular infiltration compare to the wild type mice after influenza infection. Adoptive transfer of iNKT cells from wild type, PD-L1 or PD-L2 deficient mice into iNKT cell deficient mice recapitulated these findings. Interestingly, in our transfer system PD-L1−/−-derived iNKT cells produced high levels of interferon-gamma whereas PD-L2−/−-derived iNKT cells produced high amounts of interleukin-4 and 13 suggesting a role for these cytokines in sensitivity to influenza. We identified that PD-L1 negatively regulates the frequency of iNKT cell subsets in the lungs of IAV infected mice. Altogether, these results demonstrate that lack of PD-L1 expression by iNKT cells reduces the sensitivity to IAV and that the presence of PD-L2 is important for dampening the deleterious inflammatory responses after IAV infection. Our findings potentially have clinical implications for developing new therapies for influenza.
Collapse
|
144
|
Peraino JS, Schenk M, Zhang H, Li G, Hermanrud CE, Neville DM, Sachs DH, Huang CA, Duran-Struuck R, Wang Z. A truncated diphtheria toxin based recombinant porcine CTLA-4 fusion toxin. J Immunol Methods 2013; 391:103-11. [PMID: 23470981 DOI: 10.1016/j.jim.2013.02.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 02/26/2013] [Accepted: 02/27/2013] [Indexed: 01/19/2023]
Abstract
Targeted cell therapies are possible through the generation of recombinant fusion proteins that combine a toxin, such as diphtheria toxin (DT), with an antibody or other molecule that confers specificity. Upon binding of the fusion protein to the cell of interest, the diphtheria toxin is internalized which results in protein synthesis inhibition and subsequent cell death. We have recently expressed and purified the recombinant soluble porcine CTLA-4 both with and without N-glycosylation in yeast Pichia pastoris for in vivo use in our preclinical swine model. The glycosylated and non-N-glycosylated versions of this recombinant protein each bind to a porcine CD80 expressing B-cell lymphoma line (LCL13271) with equal affinity (K(D)=13 nM). In this study we have linked each of the glycosylated and non-N-glycosylated soluble porcine CTLA-4 proteins to the truncated diphtheria toxin DT390 through genetic engineering yielding three versions of the porcine CTLA-4 fusion toxins: 1) monovalent glycosylated soluble porcine CTLA-4 fusion toxin; 2) monovalent non-N-glycosylated soluble porcine CTLA-4 fusion toxin and 3) bivalent non-N-glycosylated soluble porcine CTLA-4 fusion toxin. Protein synthesis inhibition analysis demonstrated that while all three fusion toxins are capable of inhibiting protein synthesis in vitro, the non-N-glycosylated porcine CTLA-4 isoforms function most efficiently. Binding analysis using flow cytometry of the porcine CTLA-4 fusion toxins to LCL13271 cells also demonstrated that the non-N-glycosylated porcine CTLA-4 isoforms bind to these cells with higher affinity compared to the glycosylated fusion toxin. The monovalent non-N-glycosylated porcine CTLA-4 fusion toxin was tested in vivo. NSG (NOD/SCID IL-2 receptor γ(-)/(-)) mice were injected with porcine CD80(+) LCL13271 tumor cells. All animals succumbed to tumors and those treated with the monovalent non-N-glycosylated porcine CTLA-4 fusion toxin survived longer based on a symptomatic scoring system compared to the untreated controls. This recombinant protein may therefore provide a novel approach for in vivo depletion of porcine antigen presenting cells (APCs) for studies investigating the induction of transplantation tolerance, autoimmune disease and cancer treatment.
Collapse
Affiliation(s)
- Jaclyn Stromp Peraino
- Transplantation Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
145
|
Frebel H, Oxenius A. The risks of targeting co-inhibitory pathways to modulate pathogen-directed T cell responses. Trends Immunol 2013; 34:193-9. [PMID: 23333205 PMCID: PMC7106470 DOI: 10.1016/j.it.2012.12.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 12/18/2012] [Accepted: 12/21/2012] [Indexed: 12/22/2022]
Abstract
The identification of T cell co-inhibition as a central mechanism in the regulation of adaptive immunity during infectious diseases provides new opportunities for immunotherapeutic interventions. However, the fact that T cell activity is frequently downregulated during pathogen-directed responses suggests a pivotal physiological role of co-inhibitory pathways during infectious disease. Reports of exacerbated immunopathology in conditions of impaired co-inhibition foster the view that downregulation of T cell activity is an essential negative feedback mechanism that protects from excessive pathogen-directed immunity. Thus, targeting co-inhibitory pathways can bear detrimental potential through the deregulation of physiological processes. Here, we summarize recent preclinical and clinical interventions that report immune-related adverse events after targeting co-inhibitory pathways.
Collapse
Affiliation(s)
- Helge Frebel
- Institute of Microbiology, ETH Zurich, Wolfgang-Pauli-Str. 10, 8093 Zurich, Switzerland
| | | |
Collapse
|
146
|
Chen DS, Irving BA, Hodi FS. Molecular pathways: next-generation immunotherapy--inhibiting programmed death-ligand 1 and programmed death-1. Clin Cancer Res 2012; 18:6580-7. [PMID: 23087408 DOI: 10.1158/1078-0432.ccr-12-1362] [Citation(s) in RCA: 465] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The aim of T-cell-based immune therapy for cancer has been to generate durable clinical benefit for patients. Following a generation of therapies that largely showed minimal activity, substantial toxicity, and no biomarkers to identify which patients benefit from treatment, early studies are showing signs that programmed death-ligand 1 (PD-L1) and programmed death-1 (PD-1) inhibitors are highly active. Preclinical and early data from clinical studies suggest that targeting this pathway can induce durable clinical responses in patients in a variety of tumor types, including lung and colon cancer. Furthermore, correlations with tumor PD-L1 expression may enable selection of patients most likely to benefit from treatment. The emerging data not only offer the hope of better cancer therapy but also provide evidence that changes our understanding of how the host immune system interacts with human cancer.
Collapse
Affiliation(s)
- Daniel S Chen
- Stanford Medical Oncology, Stanford, California, USA
| | | | | |
Collapse
|
147
|
Dépis F, Hatterer E, Lamacchia C, Waldburger JM, Gabay C, Reith W, Kosco-Vilbois M, Dean Y. Long-term amelioration of established collagen-induced arthritis achieved with short-term therapy combining anti-CD3 and anti-tumor necrosis factor treatments. ACTA ACUST UNITED AC 2012; 64:3189-98. [DOI: 10.1002/art.34497] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
148
|
Brahmer JR, Tykodi SS, Chow LQM, Hwu WJ, Topalian SL, Hwu P, Drake CG, Camacho LH, Kauh J, Odunsi K, Pitot HC, Hamid O, Bhatia S, Martins R, Eaton K, Chen S, Salay TM, Alaparthy S, Grosso JF, Korman AJ, Parker SM, Agrawal S, Goldberg SM, Pardoll DM, Gupta A, Wigginton JM. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med 2012; 366:2455-65. [PMID: 22658128 PMCID: PMC3563263 DOI: 10.1056/nejmoa1200694] [Citation(s) in RCA: 6073] [Impact Index Per Article: 506.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Programmed death 1 (PD-1) protein, a T-cell coinhibitory receptor, and one of its ligands, PD-L1, play a pivotal role in the ability of tumor cells to evade the host's immune system. Blockade of interactions between PD-1 and PD-L1 enhances immune function in vitro and mediates antitumor activity in preclinical models. METHODS In this multicenter phase 1 trial, we administered intravenous anti-PD-L1 antibody (at escalating doses ranging from 0.3 to 10 mg per kilogram of body weight) to patients with selected advanced cancers. Anti-PD-L1 antibody was administered every 14 days in 6-week cycles for up to 16 cycles or until the patient had a complete response or confirmed disease progression. RESULTS As of February 24, 2012, a total of 207 patients--75 with non-small-cell lung cancer, 55 with melanoma, 18 with colorectal cancer, 17 with renal-cell cancer, 17 with ovarian cancer, 14 with pancreatic cancer, 7 with gastric cancer, and 4 with breast cancer--had received anti-PD-L1 antibody. The median duration of therapy was 12 weeks (range, 2 to 111). Grade 3 or 4 toxic effects that investigators considered to be related to treatment occurred in 9% of patients. Among patients with a response that could be evaluated, an objective response (a complete or partial response) was observed in 9 of 52 patients with melanoma, 2 of 17 with renal-cell cancer, 5 of 49 with non-small-cell lung cancer, and 1 of 17 with ovarian cancer. Responses lasted for 1 year or more in 8 of 16 patients with at least 1 year of follow-up. CONCLUSIONS Antibody-mediated blockade of PD-L1 induced durable tumor regression (objective response rate of 6 to 17%) and prolonged stabilization of disease (rates of 12 to 41% at 24 weeks) in patients with advanced cancers, including non-small-cell lung cancer, melanoma, and renal-cell cancer. (Funded by Bristol-Myers Squibb and others; ClinicalTrials.gov number, NCT00729664.).
Collapse
Affiliation(s)
- Julie R Brahmer
- Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21231, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
149
|
Abstract
Among the most promising approaches to activating therapeutic antitumour immunity is the blockade of immune checkpoints. Immune checkpoints refer to a plethora of inhibitory pathways hardwired into the immune system that are crucial for maintaining self-tolerance and modulating the duration and amplitude of physiological immune responses in peripheral tissues in order to minimize collateral tissue damage. It is now clear that tumours co-opt certain immune-checkpoint pathways as a major mechanism of immune resistance, particularly against T cells that are specific for tumour antigens. Because many of the immune checkpoints are initiated by ligand-receptor interactions, they can be readily blocked by antibodies or modulated by recombinant forms of ligands or receptors. Cytotoxic T-lymphocyte-associated antigen 4 (CTLA4) antibodies were the first of this class of immunotherapeutics to achieve US Food and Drug Administration (FDA) approval. Preliminary clinical findings with blockers of additional immune-checkpoint proteins, such as programmed cell death protein 1 (PD1), indicate broad and diverse opportunities to enhance antitumour immunity with the potential to produce durable clinical responses.
Collapse
Affiliation(s)
- Drew M Pardoll
- Johns Hopkins University School of Medicine, Sidney Kimmel Comprehensive Cancer Center, CRB1 Room 444, 1650 Orleans Street, Baltimore, Maryland 21287, USA.
| |
Collapse
|
150
|
Topalian SL, Drake CG, Pardoll DM. Targeting the PD-1/B7-H1(PD-L1) pathway to activate anti-tumor immunity. Curr Opin Immunol 2012; 24:207-12. [PMID: 22236695 DOI: 10.1016/j.coi.2011.12.009] [Citation(s) in RCA: 1029] [Impact Index Per Article: 85.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Revised: 12/19/2011] [Accepted: 12/21/2011] [Indexed: 02/06/2023]
Abstract
Genetic alterations and epigenetic dysregulation in cancer cells create a vast array of neoepitopes potentially recognizable by the immune system. Immune checkpoint blockade has the capacity to enhance and sustain endogenous immunity against non-mutated tumor-associated antigens as well as uniquely mutant antigens, establishing durable tumor control. Recent evidence from preclinical models highlights the pivotal role of the Programmed Death-1 (PD-1) T cell co-receptor and its ligands, B7-H1/PD-L1 and B7-DC/PD-L2, in maintaining an immunosuppressive tumor microenvironment. Encouraging early clinical results using blocking agents against components of the PD-1 pathway have validated its importance as a target for cancer immunotherapy.
Collapse
Affiliation(s)
- Suzanne L Topalian
- Department of Surgery, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA.
| | | | | |
Collapse
|