251
|
Expression of V-set immunoregulatory receptor in malignant mesothelioma. Mod Pathol 2020; 33:263-270. [PMID: 31363159 DOI: 10.1038/s41379-019-0328-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/20/2019] [Accepted: 06/21/2019] [Indexed: 11/09/2022]
Abstract
Malignant mesothelioma is a highly lethal cancer. V-set immunoregulatory receptor (VSIR, also known as V-domain Ig suppressor T-cell activation, VISTA), a negative immune checkpoint regulator, was reported to be expressed in malignant mesothelioma; however, its detailed expression pattern and clinicopathological significance have not been elucidated. We examined the expression of VSIR and CD274 and CD8+ tumor-infiltrating lymphocytes in a total of 124 samples from 66 patients with malignant mesothelioma and analyzed the clinicopathological characteristics and their relationship with the immunohistochemical findings. A total of 553 non-small cell lung carcinomas were also evaluated for VSIR expression. VSIR expression was higher in epithelioid type mesothelioma (p < 0.001), whereas CD274 expression was higher in sarcomatoid type (p < 0.001). CD8+ tumor-infiltrating lymphocytes were more abundant in sarcomatoid mesotheliomas (p < 0.001), VSIR-low tumors (p = 0.045), and CD274-high tumors (p < 0.001). VSIR and CD274 were differentially expressed in each histological component of the biphasic type. VSIR expression was associated with favorable survival (p = 0.008). Two patients with VSIR-high tumors had received pembrolizumab; however, they showed progressive disease. No VSIR expression was observed in tumor cells of non-small cell lung carcinomas. In conclusion, VSIR expression may define a unique class of mesothelioma, characterized by predominantly epithelioid type and favorable prognosis. VSIR expression may be used as an immunohistochemical diagnostic marker for epithelioid mesothelioma. CD274 expression was associated with sarcomatoid mesothelioma and high infiltration of CD8+ lymphocytes. Because VSIR is a negative immune regulator and expressed in malignant mesothelioma, further study is warranted to investigate the therapeutic significance of VSIR blockade in this deadly cancer.
Collapse
|
252
|
Slater BT, Han X, Chen L, Xiong Y. Structural insight into T cell coinhibition by PD-1H (VISTA). Proc Natl Acad Sci U S A 2020; 117:1648-1657. [PMID: 31919279 PMCID: PMC6983362 DOI: 10.1073/pnas.1908711117] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Programmed death-1 homolog (PD-1H), a CD28/B7 family molecule, coinhibits T cell activation and is an attractive immunotherapeutic target for cancer and inflammatory diseases. The molecular basis of its function, however, is unknown. Bioinformatic analyses indicated that PD-1H has a very long Ig variable region (IgV)-like domain and extraordinarily high histidine content, suggesting that unique structural features may contribute to coinhibitory mechanisms. Here we present the 1.9-Å crystal structure of the human PD-1H extracellular domain. It reveals an elongated CC' loop and a striking concentration of histidine residues, located in the complementarity-determining region-like proximal half of the molecule. We show that surface-exposed histidine clusters are essential for robust inhibition of T cell activation. PD-1H exhibits a noncanonical IgV-like topology including an extra "H" β-strand and "clamping" disulfide, absent in known IgV-like structures, that likely restricts its orientation on the cell surface differently from other IgV-like domains. These results provide important insight into a molecular basis of T cell coinhibition by PD-1H.
Collapse
Affiliation(s)
| | - Xue Han
- Department of Immunobiology, Yale University, New Haven, CT 06511
| | - Lieping Chen
- Department of Immunobiology, Yale University, New Haven, CT 06511;
| | - Yong Xiong
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520
| |
Collapse
|
253
|
Guerrouahen BS, Maccalli C, Cugno C, Rutella S, Akporiaye ET. Reverting Immune Suppression to Enhance Cancer Immunotherapy. Front Oncol 2020; 9:1554. [PMID: 32039024 PMCID: PMC6985581 DOI: 10.3389/fonc.2019.01554] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 12/23/2019] [Indexed: 01/26/2023] Open
Abstract
Tumors employ strategies to escape immune control. The principle aim of most cancer immunotherapies is to restore effective immune surveillance. Among the different processes regulating immune escape, tumor microenvironment-associated soluble factors, and/or cell surface-bound molecules are mostly responsible for dysfunctional activity of tumor-specific CD8+T cells. These dynamic immunosuppressive networks prevent tumor rejection at several levels, limiting also the success of immunotherapies. Nevertheless, the recent clinical development of immune checkpoint inhibitors or of molecules modulating cellular targets and immunosuppressive enzymes highlights the great potential of approaches based on the selective disruption of immunosuppressive networks. Currently, the administration of different categories of immunotherapy in combination regimens is the ultimate modality for impacting the survival of cancer patients. With the advent of immune checkpoint inhibitors, designed to mount an effective antitumor immune response, profound changes occurred in cancer immunotherapy: from a global stimulation of the immune system to a specific targeting of an immune component. This review will specifically highlight the players, the mechanisms limiting an efficient antitumor response and the current immunotherapy modalities tailored to target immune suppressive pathways. We also discuss the ongoing challenges encountered by these strategies and provide suggestions for circumventing hurdles to new immunotherapeutic approaches, including the use of relevant biomarkers in the optimization of immunotherapy regimens and the identification of patients who can benefit from defined immune-based approaches.
Collapse
Affiliation(s)
- Bella S Guerrouahen
- Sidra Medicine, Member of Qatar Foundation, Research Department, Doha, Qatar
| | - Cristina Maccalli
- Sidra Medicine, Member of Qatar Foundation, Research Department, Doha, Qatar
| | - Chiara Cugno
- Sidra Medicine, Member of Qatar Foundation, Research Department, Doha, Qatar
| | - Sergio Rutella
- John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, United Kingdom
| | - Emmanuel T Akporiaye
- Veana Therapeutics, Inc., Portland, OR, United States.,Providence Cancer Center, Portland, OR, United States
| |
Collapse
|
254
|
ElTanbouly MA, Zhao Y, Nowak E, Li J, Schaafsma E, Le Mercier I, Ceeraz S, Lines JL, Peng C, Carriere C, Huang X, Day M, Koehn B, Lee SW, Silva Morales M, Hogquist KA, Jameson SC, Mueller D, Rothstein J, Blazar BR, Cheng C, Noelle RJ. VISTA is a checkpoint regulator for naïve T cell quiescence and peripheral tolerance. Science 2020; 367:eaay0524. [PMID: 31949051 PMCID: PMC7391053 DOI: 10.1126/science.aay0524] [Citation(s) in RCA: 179] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 07/30/2019] [Accepted: 12/02/2019] [Indexed: 12/18/2022]
Abstract
Negative checkpoint regulators (NCRs) temper the T cell immune response to self-antigens and limit the development of autoimmunity. Unlike all other NCRs that are expressed on activated T lymphocytes, V-type immunoglobulin domain-containing suppressor of T cell activation (VISTA) is expressed on naïve T cells. We report an unexpected heterogeneity within the naïve T cell compartment in mice, where loss of VISTA disrupted the major quiescent naïve T cell subset and enhanced self-reactivity. Agonistic VISTA engagement increased T cell tolerance by promoting antigen-induced peripheral T cell deletion. Although a critical player in naïve T cell homeostasis, the ability of VISTA to restrain naïve T cell responses was lost under inflammatory conditions. VISTA is therefore a distinctive NCR of naïve T cells that is critical for steady-state maintenance of quiescence and peripheral tolerance.
Collapse
Affiliation(s)
- Mohamed A ElTanbouly
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Yanding Zhao
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Elizabeth Nowak
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | | | - Evelien Schaafsma
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | | | - Sabrina Ceeraz
- Immunology Discovery, Janssen Research and Development LLC, Spring House, PA, USA
| | - J Louise Lines
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Changwei Peng
- Division of Rheumatic and Autoimmune Diseases, Center for Immunology, University of Minnesota, Minneapolis, MN, USA
- The Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | | | - Xin Huang
- ImmuNext Corporation, Lebanon, NH, USA
| | - Maria Day
- ImmuNext Corporation, Lebanon, NH, USA
| | - Brent Koehn
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Sam W Lee
- Yale University School of Medicine, New Haven, CT, USA
| | - Milagros Silva Morales
- Division of Rheumatic and Autoimmune Diseases, Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Kristin A Hogquist
- Division of Rheumatic and Autoimmune Diseases, Center for Immunology, University of Minnesota, Minneapolis, MN, USA
- The Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Stephen C Jameson
- Division of Rheumatic and Autoimmune Diseases, Center for Immunology, University of Minnesota, Minneapolis, MN, USA
- The Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Daniel Mueller
- Division of Rheumatic and Autoimmune Diseases, Center for Immunology, University of Minnesota, Minneapolis, MN, USA
- The Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | | | - Bruce R Blazar
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
- Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Chao Cheng
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, NH, USA.
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Randolph J Noelle
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA.
- ImmuNext Corporation, Lebanon, NH, USA
| |
Collapse
|
255
|
Wakeley ME, Gray CC, Monaghan SF, Heffernan DS, Ayala A. Check Point Inhibitors and Their Role in Immunosuppression in Sepsis. Crit Care Clin 2020; 36:69-88. [PMID: 31733683 PMCID: PMC6863093 DOI: 10.1016/j.ccc.2019.08.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Checkpoint regulators are a group of membrane-bound receptors or ligands expressed on immune cells to regulate the immune cell response to antigen presentation and other immune stimuli, such as cytokines, chemokines, and complement. In the context of profound immune activation, such as sepsis, the immune system can be rendered anergic by these receptors to prevent excessive inflammation and tissue damage. If this septic immunosuppression is prolonged, the host is unable to mount the appropriate immune response to a secondary insult or infection. This article describes the manner in which major regulators in the B7-CD28 family and their ligands mediate immunosuppression in sepsis.
Collapse
Affiliation(s)
- Michelle E Wakeley
- Division of Surgical Research, Department of Surgery, Brown University, Rhode Island Hospital, Room 242 Aldrich Building, 593 Eddy Street, Providence, RI 02903, USA
| | - Chyna C Gray
- Molecular Biology, Cell Biology and Biochemistry Department, Brown University, Rhode Island Hospital, Room 244 Aldrich Building, 593 Eddy Street, Providence, RI 02903, USA
| | - Sean F Monaghan
- Division of Surgical Research, Department of Surgery, Brown University, Rhode Island Hospital, Room 211 Middle House, 593 Eddy Street, Providence, RI 02903, USA; Division of Trauma and Surgical Critical Care, Department of Surgery, Brown University, Rhode Island Hospital, Room 211 Middle House, 593 Eddy Street, Providence, RI 02903, USA
| | - Daithi S Heffernan
- Division of Surgical Research, Department of Surgery, Brown University, Rhode Island Hospital, Room 205 Middle House, 593 Eddy Street, Providence, RI 02903, USA; Division of Trauma and Surgical Critical Care, Department of Surgery, Brown University, Rhode Island Hospital, Room 205 Middle House, 593 Eddy Street, Providence, RI 02903, USA
| | - Alfred Ayala
- Division of Surgical Research, Department of Surgery, Brown University, Rhode Island Hospital, Room 227 Aldrich Building, 593 Eddy Street, Providence, RI 02903, USA.
| |
Collapse
|
256
|
Abstract
Tumor development is characterized by the accumulation of mutational and epigenetic changes that transform normal cells and survival pathways into self-sustaining cells capable of untrammeled growth. Although multiple modalities including surgery, radiation, and chemotherapy are available for the treatment of cancer, the benefits conferred are often limited. The immune system is capable of specific, durable, and adaptable responses. However, cancers hijack immune mechanisms such as negative regulatory checkpoints that have evolved to limit inflammatory and immune responses to thwart effective antitumor immunity. The development of monoclonal antibodies against inhibitory receptors expressed by immune cells has produced durable responses in a broad array of advanced malignancies and heralded a new dawn in the cancer armamentarium. However, these remarkable responses are limited to a minority of patients and indications, highlighting the need for more effective and novel approaches. Preclinical and clinical studies with immune checkpoint blockade are exploring the therapeutic potential antibody-based therapy targeting multiple inhibitory receptors. In this chapter, we discuss the current understanding of the structure, ligand specificities, function, and signaling activities of various inhibitory receptors. Additionally, we discuss the current development status of various immune checkpoint inhibitors targeting these negative immune receptors and highlight conceptual gaps in knowledge.
Collapse
|
257
|
Kuang L, He Y. Potential value of V-domain Ig suppressor of T-cell activation for assessing progn osis in cervical cancer and as a target for therapy. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2020; 13:26-37. [PMID: 32055269 PMCID: PMC7013368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 12/19/2019] [Indexed: 06/10/2023]
Abstract
V-domain Ig suppressor of T-cell activation (VISTA), which belongs to the B7 family and is expressed predominantly on hematopoetic cells, myeloid, granulocytic and T cells, can suppresses T-cell activation in vivo and vitro. The blockade of VISTA has displayed brilliant results in certain murine tumor models. But to date, little is known about the expression and impact of VISTA in human cervical cancer (CC). To fill this gap of information, we systemically investigated the expression of VISTA on tumor cells, intratumoral immune cells (ICs) and vascular endothelial cells (VECs) in a group of patients with CC by performing immunohistochemical analysis. The associations between VISTA expression and different clinicopathologic features were evaluated using Fisher's exact test, and the analysis of overall survival in different groups was performed by the construction of Kaplan-Meier models. The results indicated that high expression of VISTA on ICs or VECs was significantly related to advanced tumor stage and the lymph node metastasis (LNM) of CC. Furthermore, we performed multivariate Cox regression analysis, which showed that there was no association between VISTA expression and the 5-year overall survival rate, and LNM was the only independent predicting factor of poor prognosis for CC.
Collapse
Affiliation(s)
- Ledi Kuang
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan UniversityChengdu, P. R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan UniversityChengdu, P. R. China
| | - Yuedong He
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan UniversityChengdu, P. R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan UniversityChengdu, P. R. China
| |
Collapse
|
258
|
Hargadon KM. Tumor microenvironmental influences on dendritic cell and T cell function: A focus on clinically relevant immunologic and metabolic checkpoints. Clin Transl Med 2020; 10:374-411. [PMID: 32508018 PMCID: PMC7240858 DOI: 10.1002/ctm2.37] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 04/23/2020] [Accepted: 04/23/2020] [Indexed: 12/11/2022] Open
Abstract
Cancer immunotherapy is fast becoming one of the most promising means of treating malignant disease. Cancer vaccines, adoptive cell transfer therapies, and immune checkpoint blockade have all shown varying levels of success in the clinical management of several cancer types in recent years. However, despite the clinical benefits often achieved by these regimens, an ongoing problem for many patients is the inherent or acquired resistance of their cancer to immunotherapy. It is now appreciated that dendritic cells and T lymphocytes both play key roles in antitumor immune responses and that the tumor microenvironment presents a number of barriers to the function of these cells that can ultimately limit the success of immunotherapy. In particular, the engagement of several immunologic and metabolic checkpoints within the hostile tumor microenvironment can severely compromise the antitumor functions of these important immune populations. This review highlights work from both preclinical and clinical studies that has shaped our understanding of the tumor microenvironment and its influence on dendritic cell and T cell function. It focuses on clinically relevant targeted and immunotherapeutic strategies that have emerged from these studies in an effort to prevent or overcome immune subversion within the tumor microenvironment. Emphasis is also placed on the potential of next-generation combinatorial regimens that target metabolic and immunologic impediments to dendritic cell and T lymphocyte function as strategies to improve antitumor immune reactivity and the clinical outcome of cancer immunotherapy going forward.
Collapse
Affiliation(s)
- Kristian M. Hargadon
- Hargadon LaboratoryDepartment of BiologyHampden‐Sydney CollegeHampden‐SydneyVirginiaUSA
| |
Collapse
|
259
|
Broughton TWK, ElTanbouly MA, Schaafsma E, Deng J, Sarde A, Croteau W, Li J, Nowak EC, Mabaera R, Smits NC, Kuta A, Noelle RJ, Lines JL. Defining the Signature of VISTA on Myeloid Cell Chemokine Responsiveness. Front Immunol 2019; 10:2641. [PMID: 31803182 PMCID: PMC6877598 DOI: 10.3389/fimmu.2019.02641] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 10/24/2019] [Indexed: 01/02/2023] Open
Abstract
The role of negative checkpoint regulators (NCRs) in human health and disease cannot be overstated. V-domain Ig-containing Suppressor of T-cell Activation (VISTA) is an Ig superfamily protein predominantly expressed within the hematopoietic compartment and has been studied for its role in the negative regulation of T cell responses. The findings presented in this study show that, unlike all other NCRs, VISTA deficiency dramatically impacts on macrophage cytokine and chemokine production, as well as the chemotactic response of VISTA-deficient macrophages. A select group of inflammatory chemokines, including CCL2, CCL3, CCL4, and CCL5, was strikingly elevated in culture supernatants from VISTA KO macrophages. VISTA deficiency also altered chemokine receptor recycling and profoundly disrupted myeloid chemotaxis. The impact of VISTA deficiency on chemotaxis in vivo was apparent with the reduced ability of both KO macrophages and MDSCs to migrate to the tumor microenvironment. This is the first demonstration of an NCR impacting on myeloid mediator production and chemotaxis, and will guide the use of anti-VISTA therapeutics to manipulate the chemotaxis of inflammatory macrophages or immunosuppressive MDSCs in inflammatory diseases and cancer.
Collapse
Affiliation(s)
- Thomas W. K. Broughton
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
- Division of Transplantation Immunology & Mucosal Biology, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Mohamed A. ElTanbouly
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Evelien Schaafsma
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Jie Deng
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Aurélien Sarde
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Walburga Croteau
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Jiannan Li
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Elizabeth C. Nowak
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Rodwell Mabaera
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
- Section of Hematology and Oncology, Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Nicole C. Smits
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Anna Kuta
- Immunext Corp., Lebanon, NH, United States
| | - Randolph J. Noelle
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - J. Louise Lines
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| |
Collapse
|
260
|
Ikarashi S, Tsuchiya A, Kawata Y, Kojima Y, Watanabe T, Takeuchi S, Igarashi K, Ideta-Otsuka M, Oki K, Takamura M, Terai S. Effects of Human Adipose Tissue-Derived and Umbilical Cord Tissue-Derived Mesenchymal Stem Cells in a Dextran Sulfate Sodium-Induced Mouse Model. Biores Open Access 2019; 8:185-199. [PMID: 31720090 PMCID: PMC6844129 DOI: 10.1089/biores.2019.0022] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Mesenchymal stem cells (MSCs) can be acquired from medical waste. MSCs are easily expanded and have multiple functions, including anti-inflammatory effects. We evaluated the effects of human adipose tissue-derived MSCs (AD-MSCs) and umbilical cord tissue-derived MSCs (UC-MSCs) in a dextran sulfate sodium (DSS)-induced mouse model. Human AD-MSCs and UC-MSCs (1 × 106 cells) were injected intravenously into a 7-day DSS-induced colitis model. The therapeutic effects of cell origin, injection timing, and supernatants obtained from MSC cultures were evaluated. We also analyzed messenger RNA (mRNA) expression in MSCs, tissues, and intestinal flora. AD-MSCs and UC-MSCs were found to show strong anti-inflammatory effects when injected on day 3 in a mouse model. On day 11, the mRNA levels of inflammatory factors in colon tissues were significantly decreased after injection of MSCs on day 3. Supernatants from MSCs culture decreased mRNA levels of tumor necrosis factor (Tnf)-α, but had reduced therapeutic effects compared with MSC cell injection. RNA sequencing using colon tissues obtained the day after cell injection revealed changes in the TNF-α/nuclear factor-κB and T cell receptor signaling pathways. Additional analyses showed that several factors, including chromosome 10 open reading frame 54, stanniocalcin-1, and TNF receptor superfamily member 11b were increased in MSCs after adding serum from DSS colitis mice. Furthermore, both AD-MSCs and UC-MSCs maintained the balance of intestinal flora. In conclusion, AD-MSCs and UC-MSCs showed therapeutic effects against inflammation after early cell injection while maintaining the intestinal flora. Although supernatants showed therapeutic effects, cell injection was more effective against inflammation.
Collapse
Affiliation(s)
- Shunzo Ikarashi
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Atsunori Tsuchiya
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Yuzo Kawata
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Yuichi Kojima
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Takayuki Watanabe
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Suguru Takeuchi
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Katsuhide Igarashi
- Laboratory of Biofunctional Science, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo, Japan.,Life Science Tokyo Advanced Research Center (L-StaR), Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo, Japan
| | - Maky Ideta-Otsuka
- Life Science Tokyo Advanced Research Center (L-StaR), Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo, Japan
| | | | - Masaaki Takamura
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Shuji Terai
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| |
Collapse
|
261
|
|
262
|
Mardomi A, Mohammadi N, Khosroshahi HT, Abediankenari S. An update on potentials and promises of T cell co-signaling molecules in transplantation. J Cell Physiol 2019; 235:4183-4197. [PMID: 31696513 DOI: 10.1002/jcp.29369] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 10/07/2019] [Indexed: 02/06/2023]
Abstract
The promising outcomes of immune-checkpoint based immunotherapies in cancer have provided a proportional perspective ahead of exploiting similar approaches in allotransplantation. Belatacept (CTLA-4-Ig) is an example of costimulation blockers successfully exploited in renal transplantation. Due to the wide range of regulatory molecules characterized in the past decades, some of these molecules might be candidates as immunomodulators in the case of tolerance induction in transplantation. Although there are numerous attempts on the apprehension of the effects of co-signaling molecules on immune response, the necessity for a better understanding is evident. By increasing the knowledge on the biology of co-signaling pathways, some pitfalls are recognized and improved approaches are proposed. The blockage of CD80/CD28 axis is an instance of evolution toward more efficacy. It is now evident that anti-CD28 antibodies are more effective than CD80 blockers in animal models of transplantation. Other co-signaling axes such as PD-1/PD-L1, CD40/CD154, 2B4/CD48, and others discussed in the present review are examples of critical immunomodulatory molecules in allogeneic transplantation. We review here the outcomes of recent experiences with co-signaling molecules in preclinical studies of solid organ transplantation.
Collapse
Affiliation(s)
- Alireza Mardomi
- Department of Immunology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran.,Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Nabiallah Mohammadi
- Department of Immunology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | | | - Saeid Abediankenari
- Department of Immunology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
263
|
Qin S, Xu L, Yi M, Yu S, Wu K, Luo S. Novel immune checkpoint targets: moving beyond PD-1 and CTLA-4. Mol Cancer 2019; 18:155. [PMID: 31690319 PMCID: PMC6833286 DOI: 10.1186/s12943-019-1091-2] [Citation(s) in RCA: 795] [Impact Index Per Article: 132.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Accepted: 10/18/2019] [Indexed: 02/10/2023] Open
Abstract
The emergence of immune checkpoint inhibitors (ICIs), mainly including anti-programmed cell death protein 1/programmed cell death ligand 1 (PD-1/PD-L1) and anti-cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) monoclonal antibodies (mAbs), has shaped therapeutic landscape of some type of cancers. Despite some ICIs have manifested compelling clinical effectiveness in certain tumor types, the majority of patients still showed de novo or adaptive resistance. At present, the overall efficiency of immune checkpoint therapy remains unsatisfactory. Exploring additional immune checkpoint molecules is a hot research topic. Recent studies have identified several new immune checkpoint targets, like lymphocyte activation gene-3 (LAG-3), T cell immunoglobulin and mucin-domain containing-3 (TIM-3), T cell immunoglobulin and ITIM domain (TIGIT), V-domain Ig suppressor of T cell activation (VISTA), and so on. The investigations about these molecules have generated promising results in preclinical studies and/or clinical trials. In this review, we discussed the structure and expression of these newly-characterized immune checkpoints molecules, presented the current progress and understanding of them. Moreover, we summarized the clinical data pertinent to these recent immune checkpoint molecules as well as their application prospects.
Collapse
Affiliation(s)
- Shuang Qin
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Linping Xu
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Ming Yi
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Shengnan Yu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China. .,Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China.
| | - Suxia Luo
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China.
| |
Collapse
|
264
|
Andrews LP, Yano H, Vignali DAA. Inhibitory receptors and ligands beyond PD-1, PD-L1 and CTLA-4: breakthroughs or backups. Nat Immunol 2019; 20:1425-1434. [PMID: 31611702 DOI: 10.1038/s41590-019-0512-0] [Citation(s) in RCA: 349] [Impact Index Per Article: 58.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 09/04/2019] [Indexed: 12/14/2022]
Abstract
Although immunotherapeutics targeting the inhibitory receptors (IRs) CTLA-4, PD-1 or PD-L1 have made substantial clinical progress in cancer, a considerable proportion of patients remain unresponsive to treatment. Targeting novel IR-ligand pathways in combination with current immunotherapies may improve clinical outcomes. New clinical immunotherapeutics target T cell-expressed IRs (LAG-3, TIM-3 and TIGIT) as well as inhibitory ligands in the B7 family (B7-H3, B7-H4 and B7-H5), although many of these targets have complex biologies and unclear mechanisms of action. With only modest clinical success in targeting these IRs, current immunotherapeutic design may not be optimal. This Review covers the biology of targeting novel IR-ligand pathways and the current clinical status of their immunotherapeutics, either as monotherapy or in combination with antibody to PD-1 or to its ligand PD-L1. Further understanding of the basic biology of these targets is imperative to the development of effective cancer immunotherapies.
Collapse
Affiliation(s)
- Lawrence P Andrews
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Hiroshi Yano
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Graduate Program of Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Dario A A Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA. .,Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
| |
Collapse
|
265
|
Murphy KA, Bhamidipati K, Rubin SJS, Kipp L, Robinson WH, Lanz TV. Immunomodulatory receptors are differentially expressed in B and T cell subsets relevant to autoimmune disease. Clin Immunol 2019; 209:108276. [PMID: 31669582 DOI: 10.1016/j.clim.2019.108276] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/15/2019] [Accepted: 10/17/2019] [Indexed: 12/15/2022]
Abstract
Inhibitory cell-surface receptors on lymphocytes, often called immune checkpoints, are powerful targets for cancer therapy. Despite their direct involvement in autoimmune pathology, they are currently not exploited therapeutically for autoimmune diseases. Understanding the expression pattern of these receptors in health and disease is essential for targeted drug design. Here, we designed three 23-colour flow cytometry panels for peripheral-blood T cells, including 15 lineage-defining markers and 21 immunomodulatory cell-surface receptors, and a 22-marker panel for B cells. Blood samples from healthy individuals, multiple sclerosis (MS), and lupus (SLE) patients were included in the study. Several receptors show differential expression on regulatory T cells (Treg) compared to T helper (Th) 1 and Th17 cells, and functional relevance of this difference could be shown for BTLA and CD5. Unbiased multiparametric analysis revealed a subset of activated CD8+ T cells and a subset of unswitched memory B cells that are diminished in MS and SLE, respectively.
Collapse
Affiliation(s)
- Katherine A Murphy
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Kartik Bhamidipati
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Samuel J S Rubin
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Lucas Kipp
- Division of Neuroimmunology, Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, United States
| | - William H Robinson
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States.
| | - Tobias V Lanz
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States; Department of Neurology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.
| |
Collapse
|
266
|
Hicks KC, Knudson KM, Lee KL, Hamilton DH, Hodge JW, Figg WD, Ordentlich P, Jones FR, Rabizadeh S, Soon-Shiong P, Schlom J, Gameiro SR. Cooperative Immune-Mediated Mechanisms of the HDAC Inhibitor Entinostat, an IL15 Superagonist, and a Cancer Vaccine Effectively Synergize as a Novel Cancer Therapy. Clin Cancer Res 2019; 26:704-716. [PMID: 31645354 DOI: 10.1158/1078-0432.ccr-19-0727] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 07/09/2019] [Accepted: 10/18/2019] [Indexed: 12/18/2022]
Abstract
PURPOSE Immunotherapy has demonstrated clinical efficacy in subsets of patients with solid carcinomas. Multimodal therapies using agents that can affect different arms of the immune system and/or tumor microenvironment (TME) might increase clinical responses. EXPERIMENTAL DESIGN We demonstrate that entinostat, a class I histone deacetylase inhibitor, enhances the antitumor efficacy of the IL15 superagonist N-803 plus vaccine in 4T1 triple-negative breast and MC38-CEA colon murine carcinoma models. A comprehensive immune and gene-expression analysis was performed in the periphery and/or TME of MC38-CEA tumor-bearing mice. RESULTS Although N-803 plus vaccine induced peripheral CD8+ T-cell activation and cytokine production, there was no reduction in tumor burden and poor tumor infiltration of CD8+ T cells with minimal levels of granzyme B. For the first time, we demonstrate that the addition of entinostat to N-803 plus vaccine promoted significant tumor control, correlating with increased expression of genes associated with tumor inflammation, enhanced infiltration of activated CD8+ T cells with maximal granzyme B, T-cell responses to multiple tumor-associated antigens, increased serum IFNγ, reduction of regulatory T cells in the TME, and decreased expression of the checkpoint V-domain Ig suppressor of T-cell activation (VISTA) on multiple immune subsets. CONCLUSIONS Collectively, these data demonstrate that the synergistic combination of entinostat, N-803, and vaccine elicits potent antitumor activity by generating a more inflamed TME. These findings thus form the rationale for the use of this combination of agents for patients harboring poorly or noninflamed solid carcinomas.
Collapse
Affiliation(s)
- Kristin C Hicks
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Karin M Knudson
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Karin L Lee
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Duane H Hamilton
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - James W Hodge
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - William D Figg
- Clinical Pharmacology Program, Office of the Clinical Director, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | | | | | - Shahrooz Rabizadeh
- ImmunityBio, Culver City, California.,NantOmics, LLC, Culver City, California.,NantWorks, Culver City, California
| | - Patrick Soon-Shiong
- ImmunityBio, Culver City, California.,NantOmics, LLC, Culver City, California.,NantWorks, Culver City, California
| | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, NCI, NIH, Bethesda, Maryland.
| | - Sofia R Gameiro
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| |
Collapse
|
267
|
VISTA is an acidic pH-selective ligand for PSGL-1. Nature 2019; 574:565-570. [PMID: 31645726 DOI: 10.1038/s41586-019-1674-5] [Citation(s) in RCA: 244] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 09/20/2019] [Indexed: 12/20/2022]
Abstract
Co-inhibitory immune receptors can contribute to T cell dysfunction in patients with cancer1,2. Blocking antibodies against cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death 1 (PD-1) partially reverse this effect and are becoming standard of care in an increasing number of malignancies3. However, many of the other axes by which tumours become inhospitable to T cells are not fully understood. Here we report that V-domain immunoglobulin suppressor of T cell activation (VISTA) engages and suppresses T cells selectively at acidic pH such as that found in tumour microenvironments. Multiple histidine residues along the rim of the VISTA extracellular domain mediate binding to the adhesion and co-inhibitory receptor P-selectin glycoprotein ligand-1 (PSGL-1). Antibodies engineered to selectively bind and block this interaction in acidic environments were sufficient to reverse VISTA-mediated immune suppression in vivo. These findings identify a mechanism by which VISTA may engender resistance to anti-tumour immune responses, as well as an unexpectedly determinative role for pH in immune co-receptor engagement.
Collapse
|
268
|
Chapoval AI, Chapoval SP, Shcherbakova NS, Shcherbakov DN. Immune Checkpoints of the B7 Family. Part 2. Representatives of the B7 Family B7-H3, B7-H4, B7-H5, B7-H6, B7-H7, and ILDR2 and Their Receptors. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2019. [DOI: 10.1134/s1068162019050091] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
269
|
Laplagne C, Domagala M, Le Naour A, Quemerais C, Hamel D, Fournié JJ, Couderc B, Bousquet C, Ferrand A, Poupot M. Latest Advances in Targeting the Tumor Microenvironment for Tumor Suppression. Int J Mol Sci 2019; 20:E4719. [PMID: 31547627 PMCID: PMC6801830 DOI: 10.3390/ijms20194719] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/18/2019] [Accepted: 09/19/2019] [Indexed: 12/13/2022] Open
Abstract
The tumor bulk is composed of a highly heterogeneous population of cancer cells, as well as a large variety of resident and infiltrating host cells, extracellular matrix proteins, and secreted proteins, collectively known as the tumor microenvironment (TME). The TME is essential for driving tumor development by promoting cancer cell survival, migration, metastasis, chemoresistance, and the ability to evade the immune system responses. Therapeutically targeting tumor-associated macrophages (TAMs), cancer-associated fibroblasts (CAFs), regulatory T-cells (T-regs), and mesenchymal stromal/stem cells (MSCs) is likely to have an impact in cancer treatment. In this review, we focus on describing the normal physiological functions of each of these cell types and their behavior in the cancer setting. Relying on the specific surface markers and secreted molecules in this context, we review the potential targeting of these cells inducing their depletion, reprogramming, or differentiation, or inhibiting their pro-tumor functions or recruitment. Different approaches were developed for this targeting, namely, immunotherapies, vaccines, small interfering RNA, or small molecules.
Collapse
Affiliation(s)
- Chloé Laplagne
- Centre de Recherches en Cancérologie de Toulouse, Inserm UMR1037, 31037 Toulouse, France.
- Université Toulouse III Paul-Sabatier, 31400 Toulouse, France.
- ERL 5294 CNRS, 31037 Toulouse, France.
| | - Marcin Domagala
- Centre de Recherches en Cancérologie de Toulouse, Inserm UMR1037, 31037 Toulouse, France.
- Université Toulouse III Paul-Sabatier, 31400 Toulouse, France.
- ERL 5294 CNRS, 31037 Toulouse, France.
| | - Augustin Le Naour
- Centre de Recherches en Cancérologie de Toulouse, Inserm UMR1037, 31037 Toulouse, France.
- Université Toulouse III Paul-Sabatier, 31400 Toulouse, France.
- Institut Claudius Regaud, IUCT-Oncopole, 31000 Toulouse, France.
| | - Christophe Quemerais
- Centre de Recherches en Cancérologie de Toulouse, Inserm UMR1037, 31037 Toulouse, France.
- Université Toulouse III Paul-Sabatier, 31400 Toulouse, France.
- ERL 5294 CNRS, 31037 Toulouse, France.
| | - Dimitri Hamel
- Université Toulouse III Paul-Sabatier, 31400 Toulouse, France.
- Institut de Recherche en Santé Digestive, Inserm U1220, INRA, ENVT, 31024 Toulouse, France.
| | - Jean-Jacques Fournié
- Centre de Recherches en Cancérologie de Toulouse, Inserm UMR1037, 31037 Toulouse, France.
- Université Toulouse III Paul-Sabatier, 31400 Toulouse, France.
- ERL 5294 CNRS, 31037 Toulouse, France.
| | - Bettina Couderc
- Centre de Recherches en Cancérologie de Toulouse, Inserm UMR1037, 31037 Toulouse, France.
- Université Toulouse III Paul-Sabatier, 31400 Toulouse, France.
- Institut Claudius Regaud, IUCT-Oncopole, 31000 Toulouse, France.
| | - Corinne Bousquet
- Centre de Recherches en Cancérologie de Toulouse, Inserm UMR1037, 31037 Toulouse, France.
- Université Toulouse III Paul-Sabatier, 31400 Toulouse, France.
- ERL 5294 CNRS, 31037 Toulouse, France.
| | - Audrey Ferrand
- Université Toulouse III Paul-Sabatier, 31400 Toulouse, France.
- Institut de Recherche en Santé Digestive, Inserm U1220, INRA, ENVT, 31024 Toulouse, France.
| | - Mary Poupot
- Centre de Recherches en Cancérologie de Toulouse, Inserm UMR1037, 31037 Toulouse, France.
- Université Toulouse III Paul-Sabatier, 31400 Toulouse, France.
- ERL 5294 CNRS, 31037 Toulouse, France.
| |
Collapse
|
270
|
Balancing cancer immunotherapy and immune-related adverse events: The emerging role of regulatory T cells. J Autoimmun 2019; 104:102310. [PMID: 31421963 DOI: 10.1016/j.jaut.2019.102310] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 07/24/2019] [Indexed: 02/06/2023]
Abstract
Advances in our understanding οf tumor immunity have prompted a paradigm shift in oncology, with the emergence of immunotherapy, where therapeutic agents are used to target immune cells rather than cancer cells. A real breakthrough in the field of immunotherapy came with the use of immune checkpoint inhibitors (ICI), namely antagonistic antibodies that block key immune regulatory molecules (checkpoint molecules), such as cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), programmed cell death protein (PD-1) and its ligand PD-L1, that under physiologic conditions suppress T cell effector function. However, despite the enormous success, a significant proportion of patients do not respond, while responses are frequently accompanied by life-threatening autoimmune related adverse events (irAEs). A major impediment in the effectiveness of ICI immunotherapy is the tumoral resistance, which is dependent on the immunosuppressive nature of tumor microenvironment (TME). Regulatory T cells (Tregs) are among the most abundant suppressive cells in the TME and their presence has been correlated with tumor progression, invasiveness as well as metastasis. Tregs are characterized by the expression of the transcription factor Foxp3 and various mechanisms ranging from cell-to-cell contact to secretion of inhibitory molecules have been implicated in their function. Notably, Tregs amply express most of the checkpoint molecules such as CTLA4, PD1 and LAG3 and therefore represent a direct target of ICI immunotherapy. Taking into consideration the critical role of Tregs in maintenance of immune homeostasis and avoidance of autoimmunity it is plausible that targeting of Tregs by ICI immunotherapy results in the development of irAEs. Since the use of ICI becomes common, and new immune checkpoint molecules are currently under clinical trials for the treatment of cancer, the occurrence of irAEs is expected to dramatically rise. Herein we review the current literature focusing on the role of Tregs in cancer evolution, ICI response and development of irAEs. Unraveling the complex mechanisms that hinder the tumor immune surveillance and in particular how ICI immunotherapy imprint on Treg activities to promote cancer regression while avoid development of irAEs, will empower the design of novel immunotherapeutic modalities in cancer with increased efficacy and diminished adverse events.
Collapse
|
271
|
Acquired resistance to cancer immunotherapy: Role of tumor-mediated immunosuppression. Semin Cancer Biol 2019; 65:13-27. [PMID: 31362073 DOI: 10.1016/j.semcancer.2019.07.017] [Citation(s) in RCA: 164] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 07/14/2019] [Accepted: 07/23/2019] [Indexed: 02/07/2023]
Abstract
In the tumor microenvironment (TME), tumor cells are constantly evolving to reduce neoantigen generation and the mutational burden to escape the anti-tumor response. This will lower tumor reactivity to the adaptive immune response and give rise to tumor intrinsic factors, such as altered expression of immune regulatory molecules on tumor cells. Tumor-extrinsic factors, such as immunosuppressive cells, soluble suppressive molecules or inhibitory receptors expressed by immune cells will alter the composition and activity of tumor-infiltrating lymphocytes (TILs) (by increasing T regulatory cells:T effector cells ratio and inhibiting T effector cell function) and promote tumor growth and metastasis. Together, these factors limit the response rates and clinical outcomes to a particular cancer therapy. Within the TME, the cross-talks between immune and non-immune cells result in the generation of positive feedback loops, which augment immunosuppression and support tumor growth and survival (termed as tumor-mediated immunosuppression). Cancer immunotherapies, such as immune checkpoint inhibitors (ICIs) and adoptive cell transfer (ACT), have shown therapeutic efficacy in hematologic cancers and different types of solid tumors. However, achieving durable response rates in some cancer patients remains a challenge as a result of acquired resistance and tumor immune evasion. This could be driven by the cellular and molecular suppressive network within the TME or due to the loss of tumor antigens. In this review, we describe the contribution of the immunosuppressive cellular and molecular tumor network to the development of acquired resistance against cancer immunotherapies. We also discuss potential combined therapeutic strategies which could help to overcome such resistance against cancer immunotherapies, and to enhance anti-tumor immune responses and improve clinical outcomes in patients.
Collapse
|
272
|
Xu W, Dong J, Zheng Y, Zhou J, Yuan Y, Ta HM, Miller HE, Olson M, Rajasekaran K, Ernstoff MS, Wang D, Malarkannan S, Wang L. Immune-Checkpoint Protein VISTA Regulates Antitumor Immunity by Controlling Myeloid Cell-Mediated Inflammation and Immunosuppression. Cancer Immunol Res 2019; 7:1497-1510. [PMID: 31340983 DOI: 10.1158/2326-6066.cir-18-0489] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 01/04/2019] [Accepted: 07/19/2019] [Indexed: 02/07/2023]
Abstract
Immune-checkpoint protein V-domain immunoglobulin suppressor of T-cell activation (VISTA) controls antitumor immunity and is a valuable target for cancer immunotherapy. This study identified a role of VISTA in regulating Toll-like receptor (TLR) signaling in myeloid cells and controlling myeloid cell-mediated inflammation and immunosuppression. VISTA modulated the polyubiquitination and protein expression of TRAF6. Consequently, VISTA dampened TLR-mediated activation of MAPK/AP-1 and IKK/NF-κB signaling cascades. At cellular levels, VISTA regulated the effector functions of myeloid-derived suppressor cells and tolerogenic dendritic cell (DC) subsets. Blocking VISTA augmented their ability to produce proinflammatory mediators and diminished their T cell-suppressive functions. These myeloid cell-dependent effects resulted in a stimulatory tumor microenvironment that promoted T-cell infiltration and activation. We conclude that VISTA is a critical myeloid cell-intrinsic immune-checkpoint protein and that the reprogramming of tolerogenic myeloid cells following VISTA blockade promotes the development of T cell-mediated antitumor immunity.
Collapse
Affiliation(s)
- Wenwen Xu
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Juan Dong
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Yongwei Zheng
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin.,Blood Research Institute, Milwaukee, Wisconsin
| | - Juan Zhou
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Immunology, Children's Hospital of Chongqing Medical University, Yuzhong District, Chongqing, P.R. China
| | - Ying Yuan
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Hieu Minh Ta
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Halli E Miller
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Michael Olson
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | | | | | - Demin Wang
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin.,Blood Research Institute, Milwaukee, Wisconsin
| | - Subramaniam Malarkannan
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin.,Blood Research Institute, Milwaukee, Wisconsin.,Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Li Wang
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, Cleveland, Ohio.
| |
Collapse
|
273
|
Toor SM, Sasidharan Nair V, Decock J, Elkord E. Immune checkpoints in the tumor microenvironment. Semin Cancer Biol 2019; 65:1-12. [PMID: 31265893 DOI: 10.1016/j.semcancer.2019.06.021] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 06/15/2019] [Accepted: 06/28/2019] [Indexed: 02/07/2023]
Abstract
Interactions between immune checkpoints (ICs) and their ligands negatively regulate T cell activation pathways involved in physiological immune responses against specific antigens. ICs and their ligands are frequently upregulated in the tumor microenvironment (TME) of various malignancies, and they represent significant barriers for induction of effective anti-tumor immune responses. Several IC inhibitors (ICIs) have been developed, with some currently in clinical trials and others have been approved for the treatment of different cancers. However, tumor cells are able to counteract the activity of ICIs and can commission additional inhibitory pathways via expression of other ICs/ligands within the TME. This review discusses the expression of various ICs/ligands in the TME and their impact on tumor immune evasion. Additionally, we discuss various regulatory mechanisms, including genetic and epigenetic, and other modulatory factors including hypoxia and the presence of immunosuppressive populations in the TME, which result in upregulation of ICs in various cancers. Moreover, we discuss the prognostic significance of ICs and their ligands, and the potential strategies to enhance treatment responses to ICIs. This review aims to advance our current knowledge on the role of ICs in the TME and the clinical benefits of targeting them.
Collapse
Affiliation(s)
- Salman M Toor
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar
| | - Varun Sasidharan Nair
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar
| | - Julie Decock
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar
| | - Eyad Elkord
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar; Institute of Cancer Sciences, University of Manchester, Manchester, United Kingdom.
| |
Collapse
|
274
|
Hsu MM, Balar AV. PD-1/PD-L1 Combinations in Advanced Urothelial Cancer: Rationale and Current Clinical Trials. Clin Genitourin Cancer 2019; 17:e618-e626. [PMID: 31005473 DOI: 10.1016/j.clgc.2019.03.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 01/14/2019] [Accepted: 03/17/2019] [Indexed: 12/27/2022]
Abstract
Chemotherapy is no longer the only viable option for patients with locally advanced or metastatic urothelial carcinoma. Immunotherapy, as checkpoint inhibition, has received United States Food and Drug Administration approval in the preceding several years, both in the second-line and first-line for cisplatin-ineligible patients. Those who respond often do so durably; however, response rates in the first line are 23% to 24%, and are lower in the second line. With a focus on urothelial carcinoma, this review discusses the tumor microenvironment and its negative influence on anti-tumor immunity, as well as measures to counteract immune suppression or evasion. The review then describes a range of current clinical trials implementing these measures in the form of programmed death-combination therapy, specifically in advanced bladder and urothelial cancers.
Collapse
Affiliation(s)
- Miles M Hsu
- New York University School of Medicine, New York, NY.
| | - Arjun V Balar
- New York University Perlmutter Cancer Center, New York, NY
| |
Collapse
|
275
|
Targeting Immune Checkpoints in Lung Cancer: Current Landscape and Future Prospects. Clin Drug Investig 2019; 39:341-353. [PMID: 30778885 DOI: 10.1007/s40261-018-00746-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Lung cancer is the most prevalent and deadly cancer worldwide. Immune checkpoint therapy, which targets regulatory pathways in T cells to boost anti-tumor immune response, has revolutionized lung cancer treatment paradigms. Inhibitors of the most established immune checkpoints such as programmed death-1 (PD-1)/PD-ligand 1 (PD-L1) have been approved by the US Food and Drug Administration in the management of lung cancer. Despite the pronounced survival benefits that have been seen with immune checkpoint inhibitors, not all lung cancer patients respond to single-agent immunotherapy due to the complexity of the immune microenvironment and tumor resistance. Alternative immune checkpoints beyond PD-1/PD-L1 must be sought so that more patients can benefit from immune checkpoint therapy. Additionally, novel combination strategies of immunotherapy and conventional treatments (e.g., chemotherapy, radiotherapy, and targeted therapy) have shown promise in some clinical trials. Meanwhile, identification of predictive biomarkers is pivotal in selecting eligible patients for immunotherapy and to guide individualized clinical decision-making. The future of immune checkpoint therapy in lung cancer is not devoid of challenges, and more prospective clinical studies are awaited to translate our understanding from bench to bedside.
Collapse
|
276
|
Deng J, Li J, Sarde A, Lines JL, Lee YC, Qian DC, Pechenick DA, Manivanh R, Le Mercier I, Lowrey CH, Varn FS, Cheng C, Leib DA, Noelle RJ, Mabaera R. Hypoxia-Induced VISTA Promotes the Suppressive Function of Myeloid-Derived Suppressor Cells in the Tumor Microenvironment. Cancer Immunol Res 2019; 7:1079-1090. [PMID: 31088847 DOI: 10.1158/2326-6066.cir-18-0507] [Citation(s) in RCA: 157] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 12/31/2018] [Accepted: 05/06/2019] [Indexed: 01/12/2023]
Abstract
Tumor hypoxia is a negative prognostic factor that is implicated in oncogenic signal activation, immune escape, and resistance to treatment. Identifying the mechanistic role of hypoxia in immune escape and resistance to immune-checkpoint inhibitors may aid the identification of therapeutic targets. We and others have shown that V-domain Ig suppressor of T-cell activation (VISTA), a negative checkpoint regulator in the B7 family, is highly expressed in the tumor microenvironment in tumor models and primary human cancers. In this study, we show that VISTA and HIF1α activity are correlated in a cohort of colorectal cancer patients. High VISTA expression was associated with worse overall survival. We used the CT26 colon cancer model to investigate the regulation of VISTA by hypoxia. Compared with less hypoxic tumor regions or draining lymph nodes, regions of profound hypoxia in the tumor microenvironment were associated with increased VISTA expression on tumor-infiltrating myeloid-derived suppressor cells (MDSC). Using chromatin immunoprecipitation and genetic silencing, we show that hypoxia-inducible factor (HIF)-1α binding to a conserved hypoxia response element in the VISTA promoter upregulated VISTA on myeloid cells. Further, antibody targeting or genetic ablation of VISTA under hypoxia relieved MDSC-mediated T-cell suppression, revealing VISTA as a mediator of MDSC function. Collectively, these data suggest that targeting VISTA may mitigate the deleterious effects of hypoxia on antitumor immunity.
Collapse
Affiliation(s)
- Jie Deng
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Jiannan Li
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Aurelien Sarde
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - J Louise Lines
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Yu-Chi Lee
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - David C Qian
- Department of Biomedical Data Sciences, Williamson Translational Research Building, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | | | - Richard Manivanh
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Isabelle Le Mercier
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Christopher H Lowrey
- Section of Hematology and Oncology, Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
| | - Frederick S Varn
- Department of Genetics, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Chao Cheng
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire.,Department of Genetics, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - David A Leib
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Randolph J Noelle
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Rodwell Mabaera
- Section of Hematology and Oncology, Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire.
| |
Collapse
|
277
|
Saleh R, Elkord E. Treg-mediated acquired resistance to immune checkpoint inhibitors. Cancer Lett 2019; 457:168-179. [PMID: 31078738 DOI: 10.1016/j.canlet.2019.05.003] [Citation(s) in RCA: 159] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/29/2019] [Accepted: 05/06/2019] [Indexed: 02/08/2023]
Abstract
T Regulatory cells (Tregs) act as a double-edged sword by regulating immune homeostasis (protective role) and inhibiting immune responses in different disease settings (pathological role). They contribute to cancer development and progression by suppressing T effector cell (Teff) functions. Decreased ratios of intratumoral CD8+ T cells to Tregs have been associated with poor prognosis in most cancer types. Targeting immune checkpoints (ICs), such as cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) and programmed cell death-1 (PD-1), by immune checkpoint inhibitors (ICIs) in cancer patients has been beneficial in inducing anti-tumor immune responses and improving clinical outcomes. However, response rates remain relatively low, ranging from 15 to 40% depending on cancer type. Additionally, a significant proportion of patients who initially demonstrates a clinical response can acquire resistance overtime. This acquired resistance could occur due to the emergence of compensatory mechanisms within the tumor microenvironment (TME) to evade the anti-tumor effects of ICIs. In this review, we describe the immunosuppressive role of Tregs in the TME, the effects of currently approved ICIs on Treg phenotype and function, and the mechanisms of acquired resistance to ICIs mediated by Tregs within the TME, such as the over-expression of ICs, the up-regulation of immunosuppressive molecules, and apoptotic Treg-induced immunosuppression. We also describe potential therapeutic strategies to target Tregs in combination with ICIs aiming to overcome such resistance and improve clinical outcomes. Elucidating the Treg-mediated acquired resistance mechanisms should benefit the designing of well-targeted therapeutic strategies to overcome resistance and maximize the therapeutic efficacy in cancer patients.
Collapse
Affiliation(s)
- Reem Saleh
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Eyad Elkord
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar.
| |
Collapse
|
278
|
Guo Q, Huang F, Goncalves C, Del Rincón SV, Miller WH. Translation of cancer immunotherapy from the bench to the bedside. Adv Cancer Res 2019; 143:1-62. [PMID: 31202357 DOI: 10.1016/bs.acr.2019.03.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
The tremendous success of immune checkpoint blockades has revolutionized cancer management. Our increased understanding of the cell types that compose the tumor microenvironment (TME), including those of the innate and adaptive immune system, has helped to shape additional immune modulatory strategies in cancer care. Pre-clinical and clinical investigations targeting novel checkpoint interactions and key pathways that regulate cancer immunity continue to increase rapidly. Various combinatorial drug regimens are being tested in attempt to achieve durable response and survival rates of patients with cancer. This review provides an overview of specific components of the TME, an introduction to novel immune checkpoints, followed by a survey of present day and future combination immune modulatory therapies. The idea that the immune system can recognize and destroy tumor cells was first described in the cancer immunosurveillance hypothesis of Burnet and Thomas. However, early experimental evidence failed to support the concept. It was not until the late 1990s when seminal papers clearly showed the existence of cancer immunosurveillance, leading to the cancer immunoediting hypothesis. In this century, progress in the understanding of negative regulators of the immune response led to the discovery that inhibition of these regulators in patients with cancer could lead to dramatic and durable remissions. Drs. Tasuku Honjo and James P. Allison were awarded the Nobel Prize in 2018 for their pioneering work in this field. We now see rapid advances in cancer immunology and emerging effective therapies revolutionizing cancer care across tumor types in the clinic, while pre-clinical research is moving from a focus on the malignant cells themselves to dissect the highly heterogenic and complex multi-cellular tumor microenvironment (TME).
Collapse
Affiliation(s)
- Qianyu Guo
- Division of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada; Jewish General Hospital, Segal Cancer Centre, Department of Oncology, Montreal, QC, Canada
| | - Fan Huang
- Division of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada; Jewish General Hospital, Segal Cancer Centre, Department of Oncology, Montreal, QC, Canada
| | - Christophe Goncalves
- Jewish General Hospital, Segal Cancer Centre, Department of Oncology, Montreal, QC, Canada
| | - Sonia V Del Rincón
- Division of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada; Jewish General Hospital, Segal Cancer Centre, Department of Oncology, Montreal, QC, Canada
| | - Wilson H Miller
- Division of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada; Jewish General Hospital, Segal Cancer Centre, Department of Oncology, Montreal, QC, Canada; Rossy Cancer Network, Montreal, QC, Canada.
| |
Collapse
|
279
|
Wylie B, Macri C, Mintern JD, Waithman J. Dendritic Cells and Cancer: From Biology to Therapeutic Intervention. Cancers (Basel) 2019; 11:E521. [PMID: 30979057 PMCID: PMC6521027 DOI: 10.3390/cancers11040521] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 04/05/2019] [Accepted: 04/07/2019] [Indexed: 12/16/2022] Open
Abstract
Inducing effective anti-tumor immunity has become a major therapeutic strategy against cancer. Dendritic cells (DC) are a heterogenous population of antigen presenting cells that infiltrate tumors. While DC play a critical role in the priming and maintenance of local immunity, their functions are often diminished, or suppressed, by factors encountered in the tumor microenvironment. Furthermore, DC populations with immunosuppressive activities are also recruited to tumors, limiting T cell infiltration and promoting tumor growth. Anti-cancer therapies can impact the function of tumor-associated DC and/or alter their phenotype. Therefore, the design of effective anti-cancer therapies for clinical translation should consider how best to boost tumor-associated DC function to drive anti-tumor immunity. In this review, we discuss the different subsets of tumor-infiltrating DC and their role in anti-tumor immunity. Moreover, we describe strategies to enhance DC function within tumors and harness these cells for effective tumor immunotherapy.
Collapse
Affiliation(s)
- Ben Wylie
- Phylogica, Harry Perkins Institute, QEII Medical Centre, Nedlands, WA 6009, Australia.
| | - Christophe Macri
- Department of Biochemistry and Molecular Biology, The University of Melbourne, Bio21, Molecular Science and Biotechnology Institute, Parkville, VIC 3010, Australia.
| | - Justine D Mintern
- Department of Biochemistry and Molecular Biology, The University of Melbourne, Bio21, Molecular Science and Biotechnology Institute, Parkville, VIC 3010, Australia.
| | - Jason Waithman
- Telethon Kids Institute, University of Western Australia, Northern Entrance, Perth Children's Hospital, Nedlands, WA 6009, Australia.
| |
Collapse
|
280
|
Jiang Y, Chen M, Nie H, Yuan Y. PD-1 and PD-L1 in cancer immunotherapy: clinical implications and future considerations. Hum Vaccin Immunother 2019; 15:1111-1122. [PMID: 30888929 DOI: 10.1080/21645515.2019.1571892] [Citation(s) in RCA: 385] [Impact Index Per Article: 64.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Programmed death-1 (PD-1) is a cell surface receptor that functions as a T cell checkpoint and plays a central role in regulating T cell exhaustion. Binding of PD-1 to its ligand, programmed death-ligand 1 (PD-L1), activates downstream signaling pathways and inhibits T cell activation. Moreover abnormally high PD-L1 expression on tumor cells and antigen-presenting cells in the tumor microenvironment mediates tumor immune escape, and the development of anti-PD-1/PD-L1 antibodies has recently become a hot topic in cancer immunotherapy. Here, we review the structure of PD-1 and PD-L1, the function of the PD-1/PD-L1 signaling pathway, the application of PD-1 or PD-L1 monoclonal antibodies and future directions for anti-PD-1/PD-L1 antibodies with combination therapies. Cancer immunotherapy using PD-1/PD-L1 immune checkpoint blockade may require more studies, and this approach may be curative for patients with many types of cancer in the future.
Collapse
Affiliation(s)
- Yongshuai Jiang
- a Shanghai Institute of Immunology, Department of Immunology and Microbiology , Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Ming Chen
- b Department of Gynecology, Shanghai Ninth People's Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Hong Nie
- a Shanghai Institute of Immunology, Department of Immunology and Microbiology , Shanghai Jiao Tong University School of Medicine , Shanghai , China.,c Department of Stomatology, Xin Hua Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Yuanyang Yuan
- a Shanghai Institute of Immunology, Department of Immunology and Microbiology , Shanghai Jiao Tong University School of Medicine , Shanghai , China
| |
Collapse
|
281
|
Immunological and clinical implications of immune checkpoint blockade in human cancer. Arch Pharm Res 2019; 42:567-581. [DOI: 10.1007/s12272-019-01140-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 02/28/2019] [Indexed: 12/20/2022]
|
282
|
Current Challenges in Cancer Immunotherapy: Multimodal Approaches to Improve Efficacy and Patient Response Rates. JOURNAL OF ONCOLOGY 2019; 2019:4508794. [PMID: 30941175 PMCID: PMC6420990 DOI: 10.1155/2019/4508794] [Citation(s) in RCA: 170] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 02/12/2019] [Indexed: 12/13/2022]
Abstract
Cancer immunotherapy is a promising innovative treatment for many forms of cancer, particularly melanoma. Although immunotherapy has been shown to be efficacious, patient response rates vary and, more often than not, only a small subset of the patients within a large cohort respond favourably to the treatment. This issue is particularly concerning and becomes a challenge of immunotherapy to improve the effectiveness and patient response rates. Here, we review the specific types of available immunotherapy options, their proposed mechanism(s) of action, and the reasons why the patient response to this treatment is variable. The potential favourable options to improve response rates to immunotherapy will be discussed with an emphasis on adopting a multimodal approach on the novel role that the gut microbiota may play in modulating the efficacy of cancer immunotherapy.
Collapse
|
283
|
Loeser H, Kraemer M, Gebauer F, Bruns C, Schröder W, Zander T, Persa OD, Alakus H, Hoelscher A, Buettner R, Lohneis P, Quaas A. The expression of the immune checkpoint regulator VISTA correlates with improved overall survival in pT1/2 tumor stages in esophageal adenocarcinoma. Oncoimmunology 2019; 8:e1581546. [PMID: 31069143 PMCID: PMC6492979 DOI: 10.1080/2162402x.2019.1581546] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 01/25/2019] [Accepted: 02/06/2019] [Indexed: 02/08/2023] Open
Abstract
Immune checkpoint modulation in cancer has been demonstrated as a high-value therapeutic strategy in many tumor entities. VISTA is an immune checkpoint receptor regulating T-cell function. To the best of our knowledge, nothing is known about the expression and prognostic impact of VISTA on tumor infiltrating lymphocytes (TILs) in the tumor microenvironment of esophageal adenocarcinoma (EAC). We analyzed in total 393 EACs within a test-cohort (n = 165) and a validation-cohort (n = 228) using a monoclonal antibody (clone D1L2G). These data were statistically correlated with clinical as well as molecular data. 22.2% of the tumor cohort presented with a VISTA expression on TILs. These patients demonstrated an improved median overall survival compared to patients without VISTA expression (202.2 months vs. 21.6 months; p < 0.0001). The favorable outcome of VISTA positive tumors is significant in the entire cohort but mainly driven by the general better prognosis of T1/T2 tumors. However, in the pT1/2 group, VISTA positive tumors show a tremendous survival benefit compared to VISTA negative tumors revealing real long-term survivors in this particular subgroup. The survival difference is independent of the T-stage. This unique characteristic could influence neoadjuvant therapy concepts for EAC, since a profit of therapy could be reduced in the already favorable subgroup of VISTA positive tumors. VISTA emerges as a prognostic biomarker for long-term survival especially in the group of early TNM-stages. Future studies have to show the relevance of VISTA positive TILs within a tumor concerning response to specific immune checkpoint inhibition.
Collapse
Affiliation(s)
- Heike Loeser
- Institute of Pathology, University of Cologne, Cologne, Germany
| | - Max Kraemer
- Institute of Pathology, University of Cologne, Cologne, Germany
| | - Florian Gebauer
- Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Christiane Bruns
- Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Wolfgang Schröder
- Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Thomas Zander
- Department I of Internal Medicine, Centre for Integrated Oncology (CIO), University of Cologne, Cologne, Germany
| | - Oana-Diana Persa
- Department of Dermatology, University of Cologne, Cologne, Germany
| | - Hakan Alakus
- Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Arnulf Hoelscher
- Centre for Esophageal and Gastric Surgery, AGAPLESION Markus Krankenhaus, Frankfurt, Germany
| | | | - Philipp Lohneis
- Institute of Pathology, University of Cologne, Cologne, Germany
| | - Alexander Quaas
- Institute of Pathology, University of Cologne, Cologne, Germany
| |
Collapse
|
284
|
Ohno T, Zhang C, Kondo Y, Kang S, Furusawa E, Tsuchiya K, Miyazaki Y, Azuma M. The immune checkpoint molecule VISTA regulates allergen-specific Th2-mediated immune responses. Int Immunol 2019; 30:3-11. [PMID: 29267882 DOI: 10.1093/intimm/dxx070] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 12/15/2017] [Indexed: 12/13/2022] Open
Abstract
V-domain immunoglobulin suppressor of T-cell activation (VISTA) is a novel immune checkpoint receptor and ligand that regulates T-cell activation. We investigated the functional involvement of VISTA in Th2 cell-mediated immune responses using an ovalbumin (OVA)-induced allergic asthma model. Treatment with an anti-VISTA monoclonal antibody (mAb) during allergen sensitization increased the production of antibodies, including total IgE, OVA-specific IgG1 and IgG2a and allergen-specific IL-5 and IL-13; it also increased the expression of IL-13 by splenic CD4+ T cells. However, treatment with the anti-VISTA mAb during sensitization did not accelerate asthmatic responses, including airway hyper-responsiveness (AHR) or the number of eosinophils in bronchoalveolar lavage (BAL) fluid. In contrast, treatment with the anti-VISTA mAb during allergen challenge significantly augmented AHR and BAL fluid eosinophilia. This treatment also increased the production of IL-5 and IL-13 in BAL fluid and the expression of IL-13 by CD4+ T cells in draining lymph nodes. These results suggest that VISTA is involved in the regulation of Th2 cell generation and Th2 cell-mediated antibody production and regulates asthmatic responses, especially in the effector phase.
Collapse
Affiliation(s)
- Tatsukuni Ohno
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo, Japan
| | - Chenyang Zhang
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo, Japan
| | - Yuta Kondo
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo, Japan
| | - Siwen Kang
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo, Japan
| | - Emi Furusawa
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo, Japan
| | - Kimitake Tsuchiya
- Department of Respiratory Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo, Japan
| | - Yasunari Miyazaki
- Department of Respiratory Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo, Japan
| | - Miyuki Azuma
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
285
|
Edwards J, Tasker A, Pires da Silva I, Quek C, Batten M, Ferguson A, Allen R, Allanson B, Saw RPM, Thompson JF, Menzies AM, Palendira U, Wilmott JS, Long GV, Scolyer RA. Prevalence and Cellular Distribution of Novel Immune Checkpoint Targets Across Longitudinal Specimens in Treatment-naïve Melanoma Patients: Implications for Clinical Trials. Clin Cancer Res 2019; 25:3247-3258. [PMID: 30777877 DOI: 10.1158/1078-0432.ccr-18-4011] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/18/2019] [Accepted: 02/13/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Immunotherapies targeting costimulating and coinhibitory checkpoint receptors beyond PD-1 and CTLA-4 have entered clinical trials. Little is known about the relative abundance, coexpression, and immune cells enriched for each specific drug target, limiting understanding of the biological basis of potential treatment outcomes and development of predictive biomarkers for personalized immunotherapy. We sought to assess the abundance of checkpoint receptors during melanoma disease progression and identify immune cells enriched for them.Experimental Design: Multiplex immunofluorescence staining for immune checkpoint receptors (ICOS, GITR, OX40, PD-1, TIM-3, VISTA) was performed on 96 melanoma biopsies from 41 treatment-naïve patients, including patient-matched primary tumors, nodal metastases, and distant metastases. Mass cytometry was conducted on tumor dissociates from 18 treatment-naïve melanoma metastases to explore immune subsets enriched for checkpoint receptors. RESULTS A small subset of tumor-infiltrating leukocytes expressed checkpoint receptors at any stage of melanoma disease. GITR and OX40 were the least abundant checkpoint receptors, with <1% of intratumoral T cells expressing either marker. ICOS, PD-1, TIM-3, and VISTA were most abundant, with TIM-3 and VISTA mostly expressed on non-T cells, and TIM-3 enriched on dendritic cells. Tumor-resident T cells (CD69+/CD103+/CD8+) were enriched for TIGIT (>70%) and other coinhibitory but not costimulatory receptors. The proportion of GITR+ T cells decreased from primary melanoma (>5%) to lymph node (<1%, P = 0.04) and distant metastases (<1%, P = 0.0005). CONCLUSIONS This study provides the first comprehensive assessment of immune checkpoint receptor expression in any cancer and provides important data for rational selection of targets for trials and predictive biomarker development.
Collapse
Affiliation(s)
- Jarem Edwards
- Melanoma Institute Australia, The University of Sydney, North Sydney, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.,Centenary Institute, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Annie Tasker
- Melanoma Institute Australia, The University of Sydney, North Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Inês Pires da Silva
- Melanoma Institute Australia, The University of Sydney, North Sydney, New South Wales, Australia
| | - Camelia Quek
- Melanoma Institute Australia, The University of Sydney, North Sydney, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Marcel Batten
- Melanoma Institute Australia, The University of Sydney, North Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Angela Ferguson
- Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.,Centenary Institute, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Ruth Allen
- Centenary Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Benjamin Allanson
- Melanoma Institute Australia, The University of Sydney, North Sydney, New South Wales, Australia.,Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Robyn P M Saw
- Melanoma Institute Australia, The University of Sydney, North Sydney, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.,Royal Prince Alfred Hospital, Sydney, New South Wales, Australia.,Mater Hospital, North Sydney, Australia
| | - John F Thompson
- Melanoma Institute Australia, The University of Sydney, North Sydney, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.,Royal Prince Alfred Hospital, Sydney, New South Wales, Australia.,Mater Hospital, North Sydney, Australia
| | - Alexander M Menzies
- Melanoma Institute Australia, The University of Sydney, North Sydney, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.,Mater Hospital, North Sydney, Australia.,Royal North Shore Hospital, Sydney, New South Wales Australia
| | - Umaimainthan Palendira
- Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.,Centenary Institute, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - James S Wilmott
- Melanoma Institute Australia, The University of Sydney, North Sydney, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, North Sydney, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Mater Hospital, North Sydney, Australia.,Royal North Shore Hospital, Sydney, New South Wales Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, The University of Sydney, North Sydney, New South Wales, Australia. .,Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| |
Collapse
|
286
|
Verma V, Sharma G, Singh A. Immunotherapy in extensive small cell lung cancer. Exp Hematol Oncol 2019; 8:5. [PMID: 30740266 PMCID: PMC6360752 DOI: 10.1186/s40164-019-0129-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 01/29/2019] [Indexed: 12/26/2022] Open
Abstract
Small cell lung cancer which constitutes about 15% of lung cancers is pathobiologically and clinically distinct from non small cell cancer. Histologically it is characterized by small cells with scant cytoplasm, absent or inconspicuous nucleoli, extensive necrosis, and expresses neuroendocrine markers. It is on a spectrum of neuroendocrine cancer that extend from typical carcinoids to large cell to small cell cancer. Clinically it behaves in a more malignant fashion with a rapid doubling time, early metastasis. They respond rapidly to cytotoxic treatment however tend to develop resistance soon. Immunotherapy with checkpoint inhibitors take advantage of PD 1 ligand-receptor axis between the tumor and T cells or CTLA4 on T cells which when engaged lead to inhibition of T cells. This inhibition helps tumors to evade immune surveillance. Checkpoint inhibitors break this axis by either binding to PD 1 ligands or PD 1 to CTLA4, thereby preventing tumors to evade the immune systems. This has led to remarkable responses in tumors. The immune related adverse effects can be severe however are experienced at much lower rates as compared to cytotoxic chemotherapy. Recently, CheckMate 032 has shown impressive response rates with Nivolumab and Nivolumab/Ipilimumab in relapsed small cell cancer. IMpower 133, a phase 3 trial showed that addition of Atezolizumab to Carbo/Etoposide led to a significant survival benefit in treatment naive extensive small cell cancer. This review will summarize recent developments and ongoing studies of immune therapy in extensive small cell cancer in addition to a brief summary of immune therapy landscape of Non small cell lung cancer. Investigational approaches to immune therapy have also been delineated.
Collapse
Affiliation(s)
- Vaibhav Verma
- Mon Health Medical Center, Morgantown, WV USA
- Morgantown, WV USA
| | - Geeti Sharma
- Steward Medical Group, Trumbull Medical Center, Youngstown, OH USA
| | | |
Collapse
|
287
|
Comparison of immune infiltrates in melanoma and pancreatic cancer highlights VISTA as a potential target in pancreatic cancer. Proc Natl Acad Sci U S A 2019; 116:1692-1697. [PMID: 30635425 DOI: 10.1073/pnas.1811067116] [Citation(s) in RCA: 244] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Immune checkpoint therapy (ICT) has transformed cancer treatment in recent years; however, treatment response is not uniform across tumor types. The tumor immune microenvironment plays a critical role in determining response to ICT; therefore, understanding the differential immune infiltration between ICT-sensitive and ICT-resistant tumor types will help to develop effective treatment strategies. We performed a comprehensive analysis of the immune tumor microenvironment of an ICT-sensitive tumor (melanoma, n = 44) and an ICT-resistant tumor (pancreatic cancer, n = 67). We found that a pancreatic tumor has minimal to moderate infiltration of CD3, CD4, and CD8 T cells; however, the immune infiltrates are predominantly present in the stromal area of the tumor and are excluded from tumoral area compared with melanoma, where the immune infiltrates are primarily present in the tumoral area. Metastatic pancreatic ductal adenocarcinomas (PDACs) had a lower infiltration of total T cells compared with resectable primary PDACs, suggesting that metastatic PDACs have poor immunogenicity. Further, a significantly higher number of CD68+ macrophages and VISTA+ cells (also known as V-domain immunoglobulin suppressor of T cell activation) were found in the pancreatic stromal area compared with melanoma. We identified VISTA as a potent inhibitory checkpoint that is predominantly expressed on CD68+ macrophages on PDACs. These data suggest that VISTA may be a relevant immunotherapy target for effective treatment of patients with pancreatic cancer.
Collapse
|
288
|
Immune checkpoint blockade and its combination therapy with small-molecule inhibitors for cancer treatment. Biochim Biophys Acta Rev Cancer 2018; 1871:199-224. [PMID: 30605718 DOI: 10.1016/j.bbcan.2018.12.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 12/13/2018] [Accepted: 12/14/2018] [Indexed: 02/05/2023]
Abstract
Initially understood for its physiological maintenance of self-tolerance, the immune checkpoint molecule has recently been recognized as a promising anti-cancer target. There has been considerable interest in the biology and the action mechanism of the immune checkpoint therapy, and their incorporation with other therapeutic regimens. Recently the small-molecule inhibitor (SMI) has been identified as an attractive combination partner for immune checkpoint inhibitors (ICIs) and is becoming a novel direction for the field of combination drug design. In this review, we provide a systematic discussion of the biology and function of major immune checkpoint molecules, and their interactions with corresponding targeting agents. With both preclinical studies and clinical trials, we especially highlight the ICI + SMI combination, with its recent advances as well as its application challenges.
Collapse
|
289
|
Shahbaz S, Bozorgmehr N, Koleva P, Namdar A, Jovel J, Fava RA, Elahi S. CD71+VISTA+ erythroid cells promote the development and function of regulatory T cells through TGF-β. PLoS Biol 2018; 16:e2006649. [PMID: 30550561 PMCID: PMC6310287 DOI: 10.1371/journal.pbio.2006649] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 12/28/2018] [Accepted: 11/20/2018] [Indexed: 02/07/2023] Open
Abstract
Cell-surface transferrin receptor (CD71+) erythroid cells are abundant in newborns with immunomodulatory properties. Here, we show that neonatal CD71+ erythroid cells express significant levels of V-domain Immunoglobulin (Ig) Suppressor of T Cell Activation (VISTA) and, via constitutive production of transforming growth factor (TGF)- β, play a pivotal role in promotion of naïve CD4+ T cells into regulatory T cells (Tregs). Interestingly, we discovered that CD71+VISTA+ erythroid cells produce significantly higher levels of TGF-β compared to CD71+VISTA− erythroid cells and CD71+ erythroid cells from the VISTA knock-out (KO) mice. As a result, CD71+VISTA+ erythroid cells—compared to CD71+VISTA− and CD71+ erythroid cells from the VISTA KO mice—significantly exceed promotion of naïve CD4+ T cells into induced Tregs (iTreg) via TGF-β in vitro. However, depletion of CD71+ erythroid cells had no significant effects on the frequency of Tregs in vivo. Surprisingly, we observed that the remaining and/or newly generated CD71+ erythroid cells following anti-CD71 antibody administration exhibit a different gene expression profile, evidenced by the up-regulation of VISTA, TGF-β1, TGF-β2, and program death ligand-1 (PDL-1), which may account as a compensatory mechanism for the maintenance of Treg population. We also observed that iTreg development by CD71+ erythroid cells is mediated through the inhibition of key signaling molecules phosphorylated protein kinase B (phospho-Akt) and phosphorylated mechanistic target of rapamycin (phospho-mTOR). Finally, we found that elimination of Tregs using forkhead box P3 (FOXP3)-diptheria toxin receptor (DTR) mice resulted in a significant expansion in the frequency of CD71+ erythroid cells in vivo. Collectively, these studies provide a novel, to our knowledge, insight into the cross-talk between CD71+ erythroid cells and Tregs in newborns. Our results highlight the biological role of CD71+ erythroid cells in the neonatal period and possibly beyond. The primary role of the red blood cells is to transport oxygen, but we know relatively little about the other functions they perform. Following maturation, red blood cells exit the bone marrow and enter blood circulation. Their immature counterparts are normally absent or in very low frequency in the blood of healthy adults. However, we showed previously that immature red blood cells are abundant in the spleens of neonatal mice and in human umbilical cord blood and that these cells possess immunological properties. In this report, we studied a subset of neonatal immature red blood cells that express a protein called V-domain Immunoglobulin (Ig) Suppressor of T Cell Activation (VISTA) on their surface. We found that the presence of VISTA enables the cells to repeatedly produce the regulatory cytokine TGF-β. TGF-β induces a subset of naïve lymphocytes—the CD4+ T cells—and converts them into regulatory T cells, also known as Tregs. Tregs modulate and suppress other immune cells. Our studies provide novel insights, to our knowledge, into the immunological role of immature red blood cells in newborns.
Collapse
Affiliation(s)
- Shima Shahbaz
- Department of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Najmeh Bozorgmehr
- Department of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Petya Koleva
- Department of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Afshin Namdar
- Department of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Juan Jovel
- The Applied Genomics Core, Office of Research, University of Alberta, Edmonton, Canada
| | - Roy A. Fava
- Department of Veterans Affairs Medical Center, Research Service, White River Junction, Vermont, United States of America
- Department of Medicine, Geisel School of Medicine at Dartmouth, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire, United States of America
| | - Shokrollah Elahi
- Department of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- * E-mail:
| |
Collapse
|
290
|
Sadreddini S, Baradaran B, Aghebati-Maleki A, Sadreddini S, Shanehbandi D, Fotouhi A, Aghebati-Maleki L. Immune checkpoint blockade opens a new way to cancer immunotherapy. J Cell Physiol 2018; 234:8541-8549. [PMID: 30511409 DOI: 10.1002/jcp.27816] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 11/01/2018] [Indexed: 12/20/2022]
Abstract
Among the main promising systems to triggering therapeutic antitumor immunity is the blockade of immune checkpoints. Immune checkpoint pathways regulate the control and eradication of infections, malignancies, and resistance against a host of autoantigens. Initiation point of the immune response is T cells, which have a critical role in this pathway. As several immune checkpoints are initiated by ligand-receptor interactions, they can be freely blocked by antibodies or modulated by recombinant forms of ligands or receptors. Antibodies against cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) were the first immunotherapeutics that achieved the US Food and Drug Administration approval. Preliminary clinical results with the blockers of additional immune checkpoint proteins, such as programmed cell death protein 1 (PD-1) indicate extensive and different chances to boost antitumor immunity with the objective of conferring permanent clinical effects. This study provides an overview of the immune checkpoint pathways, including CTLA-4, PD-1, lymphocyte activation gene 3, T-cell immunoglobulin and mucin domain 3, B7-H3, and diacylglycerol kinase α and implications of their inhibition in the cancer therapy.
Collapse
Affiliation(s)
- Sanam Sadreddini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Aghebati-Maleki
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sevil Sadreddini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Fotouhi
- Department of Orthopedic Surgery, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
291
|
Hernandez-Martinez JM, Vergara E, Zatarain-Barrón ZL, Barrón-Barrón F, Arrieta O. VISTA/PD-1H: a potential target for non-small cell lung cancer immunotherapy. J Thorac Dis 2018; 10:6378-6382. [PMID: 30746169 DOI: 10.21037/jtd.2018.11.39] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Juan-Manuel Hernandez-Martinez
- Thoracic Oncology Unit and Experimental Oncology Laboratory, Instituto Nacional de Cancerología de México (INCan), Mexico City, Mexico.,CONACYT-Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Edgar Vergara
- Thoracic Oncology Unit and Experimental Oncology Laboratory, Instituto Nacional de Cancerología de México (INCan), Mexico City, Mexico
| | - Zyanya Lucia Zatarain-Barrón
- Thoracic Oncology Unit and Experimental Oncology Laboratory, Instituto Nacional de Cancerología de México (INCan), Mexico City, Mexico
| | - Feliciano Barrón-Barrón
- Thoracic Oncology Unit and Experimental Oncology Laboratory, Instituto Nacional de Cancerología de México (INCan), Mexico City, Mexico
| | - Oscar Arrieta
- Thoracic Oncology Unit and Experimental Oncology Laboratory, Instituto Nacional de Cancerología de México (INCan), Mexico City, Mexico
| |
Collapse
|
292
|
VISTA expressed in tumour cells regulates T cell function. Br J Cancer 2018; 120:115-127. [PMID: 30382166 PMCID: PMC6325144 DOI: 10.1038/s41416-018-0313-5] [Citation(s) in RCA: 143] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 09/24/2018] [Accepted: 10/02/2018] [Indexed: 12/13/2022] Open
Abstract
Background V-domain Ig suppressor of T cell activation (VISTA) is a novel inhibitory immune-checkpoint protein. VISTA expression on tumour cells and the associated regulatory mechanisms remain unclear. We investigated VISTA expression and function in tumour cells, and evaluated its mechanism and activity. Methods VISTA in tumour cells was assessed by tissue microarray analysis, immunohistochemical staining and western blot. A series of in vitro assays were used to determine the function of tumour-expressed VISTA. In vivo efficacy was evaluated in syngeneic models. Results VISTA was highly expressed in human ovarian and endometrial cancers. Upregulation of VISTA in endometrial cancer was related to the methylation status of the VISTA promoter. VISTA in tumour cells suppressed T cell proliferation and cytokine production in vitro, and decreased the tumour-infiltrating CD8+ T cells in vivo. Anti-VISTA antibody prolonged the survival of tumour-bearing mice. Conclusions This is the first demonstration that VISTA is highly expressed in human ovarian and endometrial cancer cells, and that anti-VISTA antibody treatment significantly prolongs the survival of mice bearing tumours expressing high levels of VISTA. The data suggest that VISTA is a novel immunosuppressive factor within the tumour microenvironment, as well as a new target for cancer immunotherapy.
Collapse
|
293
|
Xie S, Huang J, Qiao Q, Zang W, Hong S, Tan H, Dong C, Yang Z, Ni L. Expression of the inhibitory B7 family molecule VISTA in human colorectal carcinoma tumors. Cancer Immunol Immunother 2018; 67:1685-1694. [PMID: 30128738 PMCID: PMC11028359 DOI: 10.1007/s00262-018-2227-8] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 08/02/2018] [Indexed: 10/28/2022]
Abstract
Colorectal carcinoma (CRC) is one of the most common malignancies in the world. PD-1/PD-L1 inhibitors have benefited cancer patients with multiple tumor types. However, their efficacy for CRC is low and this treatment in melanoma patients results in adaptive resistance through upregulation of VISTA, another checkpoint inhibitory pathway. Thus, there is an urgent need to explore additional co-inhibitory molecular pathways such as VISTA for CRC treatment. In this study, C10orf54 (encoding VISTA) expression was analyzed by RNA-seq data from 367 CRC patients in human cancer datasets. Moreover, 28 clinical CRC specimens were used to assess VISTA protein expression. Human cancer datasets showed that CRC tumors expressed higher levels of C10orf54 than CD274 (encoding PD-L1). Moreover, C10orf54 mRNA expression was significantly correlated with genes responsible for tumor immune evasion. VISTA protein expression was high in tumors compared with para-tumors and normal tissues, which is similar to PD-L1 expression. However, in contrast to PD-L1, VISTA was mainly expressed by tumor-infiltrating lymphocytes. This study is the first investigation of VISTA expression in human resected CRC tumors, and the results justify the need for future studies on the role of VISTA in anti-CRC immunity in clinical samples.
Collapse
Affiliation(s)
- Shan Xie
- Institute for Immunology and School of Medicine, Tsinghua University, Medical Research Building, No.30 Haidian Shuangqing Road, Beijing, 100084, China
| | - Jia Huang
- Department of General Surgery, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Qin Qiao
- Institute for Immunology and School of Medicine, Tsinghua University, Medical Research Building, No.30 Haidian Shuangqing Road, Beijing, 100084, China
| | - Wenjuan Zang
- Institute for Immunology and School of Medicine, Tsinghua University, Medical Research Building, No.30 Haidian Shuangqing Road, Beijing, 100084, China
| | - Shanjuan Hong
- Institute for Immunology and School of Medicine, Tsinghua University, Medical Research Building, No.30 Haidian Shuangqing Road, Beijing, 100084, China
| | - Haidong Tan
- Department of General Surgery, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Chen Dong
- Institute for Immunology and School of Medicine, Tsinghua University, Medical Research Building, No.30 Haidian Shuangqing Road, Beijing, 100084, China
| | - Zhiying Yang
- Department of General Surgery, China-Japan Friendship Hospital, Beijing, 100029, China.
| | - Ling Ni
- Institute for Immunology and School of Medicine, Tsinghua University, Medical Research Building, No.30 Haidian Shuangqing Road, Beijing, 100084, China.
| |
Collapse
|
294
|
Wang J, Wu G, Manick B, Hernandez V, Renelt M, Erickson C, Guan J, Singh R, Rollins S, Solorz A, Bi M, Li J, Grabowski D, Dirkx J, Tracy C, Stuart T, Ellinghuysen C, Desmond D, Foster C, Kalabokis V. VSIG-3 as a ligand of VISTA inhibits human T-cell function. Immunology 2018; 156:74-85. [PMID: 30220083 DOI: 10.1111/imm.13001] [Citation(s) in RCA: 185] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 08/27/2018] [Accepted: 08/31/2018] [Indexed: 12/12/2022] Open
Abstract
B7 family members and their receptors play a central role in the regulation of T-cell responses through T-cell co-stimulation and co-inhibition pathways that constitute attractive targets for the development of immunotherapeutic drugs. In this study, we report that VSIG-3/IGSF11 is a ligand of B7 family member VISTA/PD-1H and inhibits human T-cell functions through a novel VSIG-3/VISTA pathway. An extensive functional ELISA binding screening assay reveals that VSIG-3 binds to the new B7 family member VISTA but does not interact with other known members of the B7 family. Under the same experimental conditions, we did not observe any significant interaction between VSIG-8 and VISTA. In addition, VSIG-3 inhibits human T-cell proliferation in the presence of T-cell receptor signaling. Furthermore, VSIG-3 significantly reduces cytokine and chemokine production by human T cells including IFN-γ, IL-2, IL-17, CCL5/Rantes, CCL3/MIP-1α, and CXCL11/I-TAC. Anti-VISTA neutralization antibodies attenuate the binding of VSIG-3 and VISTA, as well as VSIG-3-induced T-cell inhibition. Hence, we have identified a novel ligand for VISTA that is able to inhibit human T-cell proliferation and cytokine production. This unique VSIG-3/VISTA co-inhibitory pathway may provide new strategies for the treatment of human cancers, autoimmune disorders, infection, and transplant rejection and may aid in the design of better vaccines.
Collapse
Affiliation(s)
- Jinghua Wang
- Department of Protein Development, R&D Systems, Bio-Techne, Minneapolis, MN, USA
| | - Guoping Wu
- Department of Protein Development, R&D Systems, Bio-Techne, Minneapolis, MN, USA
| | - Brian Manick
- Department of Protein Development, R&D Systems, Bio-Techne, Minneapolis, MN, USA
| | - Vida Hernandez
- Department of Protein Development, R&D Systems, Bio-Techne, Minneapolis, MN, USA
| | - Mark Renelt
- Department of Protein Development, R&D Systems, Bio-Techne, Minneapolis, MN, USA
| | - Christian Erickson
- Department of Protein Development, R&D Systems, Bio-Techne, Minneapolis, MN, USA
| | - Joanna Guan
- Department of Protein Development, R&D Systems, Bio-Techne, Minneapolis, MN, USA
| | - Ravinder Singh
- Department of Protein Development, R&D Systems, Bio-Techne, Minneapolis, MN, USA
| | - Simone Rollins
- Department of Protein Development, R&D Systems, Bio-Techne, Minneapolis, MN, USA
| | - Alicia Solorz
- Department of Protein Development, R&D Systems, Bio-Techne, Minneapolis, MN, USA
| | - Ming Bi
- Department of Protein Development, R&D Systems, Bio-Techne, Minneapolis, MN, USA
| | - Jun Li
- Department of Protein Development, R&D Systems, Bio-Techne, Minneapolis, MN, USA
| | - David Grabowski
- Department of Protein Development, R&D Systems, Bio-Techne, Minneapolis, MN, USA
| | - Janette Dirkx
- Department of Protein Development, R&D Systems, Bio-Techne, Minneapolis, MN, USA
| | - Camrin Tracy
- Department of Protein Development, R&D Systems, Bio-Techne, Minneapolis, MN, USA
| | - Thomas Stuart
- Department of Protein Development, R&D Systems, Bio-Techne, Minneapolis, MN, USA
| | - Chad Ellinghuysen
- Department of Protein Development, R&D Systems, Bio-Techne, Minneapolis, MN, USA
| | - Daniel Desmond
- Department of Protein Development, R&D Systems, Bio-Techne, Minneapolis, MN, USA
| | - Craig Foster
- Department of Protein Development, R&D Systems, Bio-Techne, Minneapolis, MN, USA
| | - Vassilios Kalabokis
- Department of Protein Development, R&D Systems, Bio-Techne, Minneapolis, MN, USA
| |
Collapse
|
295
|
De Sousa Linhares A, Leitner J, Grabmeier-Pfistershammer K, Steinberger P. Not All Immune Checkpoints Are Created Equal. Front Immunol 2018; 9:1909. [PMID: 30233564 PMCID: PMC6127213 DOI: 10.3389/fimmu.2018.01909] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 08/02/2018] [Indexed: 12/20/2022] Open
Abstract
Antibodies that block T cell inhibition via the immune checkpoints CTLA-4 and PD-1 have revolutionized cancer therapy during the last 15 years. T cells express additional inhibitory surface receptors that are considered to have potential as targets in cancer immunotherapy. Antibodies against LAG-3 and TIM-3 are currently clinically tested to evaluate their effectiveness in patients suffering from advanced solid tumors or hematologic malignancies. In addition, blockade of the inhibitory BTLA receptors on human T cells may have potential to unleash T cells to effectively combat cancer cells. Much research on these immune checkpoints has focused on mouse models. The analysis of animals that lack individual inhibitory receptors has shed some light on the role of these molecules in regulating T cells, but also immune responses in general. There are current intensive efforts to gauge the efficacy of antibodies targeting these molecules called immune checkpoint inhibitors alone or in different combinations in preclinical models of cancer. Differences between mouse and human immunology warrant studies on human immune cells to appreciate the potential of individual pathways in enhancing T cell responses. Results from clinical studies are not only highlighting the great benefit of immune checkpoint inhibitors for treating cancer but also yield precious information on their role in regulating T cells and other cells of the immune system. However, despite the clinical relevance of CTLA-4 and PD-1 and the high potential of the emerging immune checkpoints, there are still substantial gaps in our understanding of the biology of these molecules, which might prevent the full realization of their therapeutic potential. This review addresses PD-1, CTLA-4, BTLA, LAG-3, and TIM-3, which are considered major inhibitory immune checkpoints expressed on T cells. It provides summaries of our current conception of the role of these molecules in regulating T cell responses, and discussions about major ambiguities and gaps in our knowledge. We emphasize that each of these molecules harbors unique properties that set it apart from the others. Their distinct functional profiles should be taken into account in therapeutic strategies that aim to exploit these pathways to enhance immune responses to combat cancer.
Collapse
Affiliation(s)
- Annika De Sousa Linhares
- Division of Immune Receptors and T Cell Activation, Medical University of Vienna, Vienna, Austria
| | - Judith Leitner
- Division of Immune Receptors and T Cell Activation, Medical University of Vienna, Vienna, Austria
| | - Katharina Grabmeier-Pfistershammer
- Division of Clinical and Experimental Immunology, Center for Pathophysiology, Infectiology, and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Peter Steinberger
- Division of Immune Receptors and T Cell Activation, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
296
|
Lazăr DC, Avram MF, Romoșan I, Cornianu M, Tăban S, Goldiș A. Prognostic significance of tumor immune microenvironment and immunotherapy: Novel insights and future perspectives in gastric cancer. World J Gastroenterol 2018; 24:3583-3616. [PMID: 30166856 PMCID: PMC6113718 DOI: 10.3748/wjg.v24.i32.3583] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 06/05/2018] [Accepted: 06/27/2018] [Indexed: 02/06/2023] Open
Abstract
Despite a decrease in gastric cancer incidence, the development of novel biologic agents and combined therapeutic strategies, the prognosis of gastric cancer remains poor. Recently, the introduction of modern immunotherapy, especially using immune checkpoint inhibitors, led to an improved prognosis in many cancers. The use of immunotherapy was also associated with manageable adverse event profiles and promising results in the treatment of patients with gastric cancer, especially in heavily pretreated patients. These data have led to an accelerated approval of some checkpoint inhibitors in this setting. Understanding the complex relationship between the host immune microenvironment and tumor and the immune escape phenomenon leading to cancer occurrence and progression will subsequently lead to the identification of prognostic immune markers. Furthermore, this understanding will result in the discovery of both new mechanisms for blocking tumor immunosuppressive signals and pathways to stimulate the local immune response by targeting and modulating different subsets of immune cells. Due to the molecular heterogeneity of gastric cancers associated with different clinico-biologic parameters, immune markers expression and prognosis, novel immunotherapy algorithms should be personalized and addressed to selected subsets of gastric tumors, which have been proven to elicit the best clinical responses. Future perspectives in the treatment of gastric cancer include tailored dual immunotherapies or a combination of immunotherapy with other targeted agents with synergistic antitumor effects.
Collapse
Affiliation(s)
- Daniela Cornelia Lazăr
- Department of Internal Medicine I, University Medical Clinic, University of Medicine and Pharmacy “Victor Babeş”, Timişoara 300041, Timiş County, Romania
| | - Mihaela Flavia Avram
- Department of Surgery X, 1st Surgery Clinic, University of Medicine and Pharmacy “Victor Babeş”, Timişoara 300041, Timiş County, Romania
| | - Ioan Romoșan
- Department of Internal Medicine I, University Medical Clinic, University of Medicine and Pharmacy “Victor Babeş”, Timişoara 300041, Timiş County, Romania
| | - Mărioara Cornianu
- Department of Pathology, University of Medicine and Pharmacy “Victor Babeş”, Timişoara 300041, Timiş County, Romania
| | - Sorina Tăban
- Department of Pathology, University of Medicine and Pharmacy “Victor Babeş”, Timişoara 300041, Timiş County, Romania
| | - Adrian Goldiș
- Department of Gastroenterology and Hepatology, University of Medicine and Pharmacy “Victor Babeş”, Timişoara 300041, Timiş County, Romania
| |
Collapse
|
297
|
Popovic A, Jaffee EM, Zaidi N. Emerging strategies for combination checkpoint modulators in cancer immunotherapy. J Clin Invest 2018; 128:3209-3218. [PMID: 30067248 DOI: 10.1172/jci120775] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Current immune checkpoint-modulating agents have demonstrated clinical efficacy in certain tumor types, particularly those with a high burden of tumor-specific neoantigens, high tumor-mutational burden, and abundant tumor-infiltrating T cells. However, these tumors often stop responding, with signs of T cells exhaustion, decreased T cell effector function, and upregulated inhibitory checkpoints. To enhance antitumor immunity and rescue exhausted T cells, newer inhibitory and stimulatory checkpoint modulators are being tested as monotherapy or in combination with approved checkpoint inhibitors. In contrast, tumors with low tumor-mutational burden, low neoantigen burden, and a paucity of T cells are immunologically "cold," and therefore first require the addition of agents to facilitate the induction of T cells into tumors. Cold tumors also often recruit immunosuppressive cell subsets, including regulatory T cells, myeloid-derived suppressor cells, and macrophages, and secrete immunosuppressive soluble cytokines, chemokines, and metabolites. To unleash an optimal antitumor immune response, combinatorial therapeutics that combine immune checkpoints with other modalities, such as vaccines, are being developed. From current preclinical data, it appears that combinatorial strategies will provide robust and durable responses in patients with immunologically cold cancers.
Collapse
|
298
|
Wang L, Jia B, Claxton DF, Ehmann WC, Rybka WB, Mineishi S, Naik S, Khawaja MR, Sivik J, Han J, Hohl RJ, Zheng H. VISTA is highly expressed on MDSCs and mediates an inhibition of T cell response in patients with AML. Oncoimmunology 2018; 7:e1469594. [PMID: 30228937 PMCID: PMC6140587 DOI: 10.1080/2162402x.2018.1469594] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 04/18/2018] [Accepted: 04/20/2018] [Indexed: 01/26/2023] Open
Abstract
Treatment of acute myeloid leukemia (AML) remains challenging. Enhancement of anti-tumor responses by blocking negative immune regulators is a promising strategy for novel effective leukemia therapeutics. V-domain Ig suppressor of T-cell activation (VISTA) is a recently defined negative regulator mediating immune evasion in cancer. To investigate the effect of VISTA on anti-leukemia immune response in AML, we initiated a study using clinical samples collected from AML patients. Here we report that VISTA is highly expressed on myeloid-derived suppressor cells (MDSCs) in the peripheral blood of AML patients. Both the frequency and intensity of VISTA expression on MDSCs are significantly higher in newly diagnosed AML than in healthy controls. Importantly knockdown of VISTA by specific siRNA potently reduced the MDSCs-mediated inhibition of CD8 T cell activity in AML, suggesting a suppressive effect of VISTA on anti-leukemia T cell response. Furthermore, we observed a strong positive association between MDSC expression of VISTA and T cell expression of PD-1 in AML. These results support the strategy of VISTA-targeted treatment for AML and underscore the strong potential for combined blockade of VISTA and PD-1 pathways in effective leukemia control.
Collapse
Affiliation(s)
- Liru Wang
- Penn State Cancer Institute, Penn State University College of Medicine, Hershey, PA, USA.,Fu Xing Hospital, Capital Medical University, Beijing, China
| | - Bei Jia
- Penn State Cancer Institute, Penn State University College of Medicine, Hershey, PA, USA
| | - David F Claxton
- Penn State Cancer Institute, Penn State University College of Medicine, Hershey, PA, USA
| | - W Christopher Ehmann
- Penn State Cancer Institute, Penn State University College of Medicine, Hershey, PA, USA
| | - Witold B Rybka
- Penn State Cancer Institute, Penn State University College of Medicine, Hershey, PA, USA
| | - Shin Mineishi
- Penn State Cancer Institute, Penn State University College of Medicine, Hershey, PA, USA
| | - Seema Naik
- Penn State Cancer Institute, Penn State University College of Medicine, Hershey, PA, USA
| | - Muhammad R Khawaja
- Penn State Cancer Institute, Penn State University College of Medicine, Hershey, PA, USA
| | - Jeff Sivik
- Department of Medicine, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Junyan Han
- Penn State Cancer Institute, Penn State University College of Medicine, Hershey, PA, USA.,Institute of Infectious Diseases, Beijing Ditan Hospital, Beijing Key Laboratory of Emerging Infectious Diseases, Capital Medical University, Beijing, China
| | - Raymond J Hohl
- Penn State Cancer Institute, Penn State University College of Medicine, Hershey, PA, USA
| | - Hong Zheng
- Penn State Cancer Institute, Penn State University College of Medicine, Hershey, PA, USA.,Depatment of Microbiology and Immunology, Penn State University College of Medicine, Hershey, PA, USA
| |
Collapse
|
299
|
Borcherding N, Kolb R, Gullicksrud J, Vikas P, Zhu Y, Zhang W. Keeping Tumors in Check: A Mechanistic Review of Clinical Response and Resistance to Immune Checkpoint Blockade in Cancer. J Mol Biol 2018; 430:2014-2029. [PMID: 29800567 PMCID: PMC6071324 DOI: 10.1016/j.jmb.2018.05.030] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 05/15/2018] [Accepted: 05/16/2018] [Indexed: 12/26/2022]
Abstract
Immune checkpoints are a diverse set of inhibitory signals to the immune system that play a functional role in adaptive immune response and self-tolerance. Dysregulation of these pathways is a vital mechanism in the avoidance of immune destruction by tumor cells. Immune checkpoint blockade (ICB) refers to targeted strategies to disrupt the tumor co-opted immune suppression to enhance anti-tumor immunity. Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death 1 (PD-1) are two immune checkpoints that have the widest range of antibody-based therapies. These therapies have gone from promising approaches to Food and Drug Administration-approved first- and second-line agents for a number of immunogenic cancers. The burgeoning investigations of ICB efficacy in blood and solid cancers have underscored the importance of identifying the predictors of response and resistance to ICB. Identification of response correlates is made complicated by the observations of mixed reactions, or different responses in multiple lesions from the same patient, and delayed responses that can occur over a year after the induction therapy. Factors that can influence response and resistance in ICB can illuminate underlying molecular mechanisms of immune activation and suppression. These same response predictors can guide the identification of patients who would benefit from ICB, reduce off-target immune-relate adverse events, and facilitate the use of combinatorial therapies to increase efficacy. Here we review the underlying principles of immune checkpoint therapy and results of single-agent ICB clinical trials, and summarize the predictors of response and resistance.
Collapse
Affiliation(s)
- Nicholas Borcherding
- Department of Pathology, University of Iowa, College of Medicine, Iowa City, IA 52242-11, USA; Cancer Biology Graduate Program, University of Iowa, College of Medicine, Iowa City, IA 52242-11, USA; Medical Scientist Training Program, University of Iowa, College of Medicine, Iowa City, IA 52242-11, USA; Holden Comprehensive Cancer Center, University of Iowa, College of Medicine, Iowa City, IA 52242-11, USA
| | - Ryan Kolb
- Department of Pathology, University of Iowa, College of Medicine, Iowa City, IA 52242-11, USA; Holden Comprehensive Cancer Center, University of Iowa, College of Medicine, Iowa City, IA 52242-11, USA
| | - Jodi Gullicksrud
- Department of Pathobiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Praveen Vikas
- Holden Comprehensive Cancer Center, University of Iowa, College of Medicine, Iowa City, IA 52242-11, USA
| | - Yuwen Zhu
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Weizhou Zhang
- Department of Pathology, University of Iowa, College of Medicine, Iowa City, IA 52242-11, USA; Cancer Biology Graduate Program, University of Iowa, College of Medicine, Iowa City, IA 52242-11, USA; Medical Scientist Training Program, University of Iowa, College of Medicine, Iowa City, IA 52242-11, USA; Holden Comprehensive Cancer Center, University of Iowa, College of Medicine, Iowa City, IA 52242-11, USA.
| |
Collapse
|
300
|
Lanitis E, Dangaj D, Irving M, Coukos G. Mechanisms regulating T-cell infiltration and activity in solid tumors. Ann Oncol 2018; 28:xii18-xii32. [PMID: 29045511 DOI: 10.1093/annonc/mdx238] [Citation(s) in RCA: 268] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
T-lymphocytes play a critical role in cancer immunity as evidenced by their presence in resected tumor samples derived from long-surviving patients, and impressive clinical responses to various immunotherapies that reinvigorate them. Indeed, tumors can upregulate a wide array of defense mechanisms, both direct and indirect, to suppress the ability of Tcells to reach the tumor bed and mount curative responses upon infiltration. In addition, patient and tumor genetics, previous antigenic experience, and the microbiome, are all important factors in shaping the T-cell repertoire and sensitivity to immunotherapy. Here, we review the mechanisms that regulate T-cell homing, infiltration, and activity within the solid tumor bed. Finally, we summarize different immunotherapies and combinatorial treatment strategies that enable the immune system to overcome barriers for enhanced tumor control and improved patient outcome.
Collapse
Affiliation(s)
- E Lanitis
- The Ludwig Branch for Cancer Research of the University of Lausanne, Epalinges
| | - D Dangaj
- The Ludwig Branch for Cancer Research of the University of Lausanne, Epalinges
| | - M Irving
- The Ludwig Branch for Cancer Research of the University of Lausanne, Epalinges
| | - G Coukos
- The Ludwig Branch for Cancer Research of the University of Lausanne, Epalinges.,Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| |
Collapse
|