101
|
Li X, Peng W, Wu J, Yeung SCJ, Yang R. Advances in immune checkpoint inhibitors induced-cardiotoxicity. Front Immunol 2023; 14:1130438. [PMID: 36911712 PMCID: PMC9995967 DOI: 10.3389/fimmu.2023.1130438] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 02/13/2023] [Indexed: 02/25/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) are approved as the first-line drug for treating many cancers and has shown significant survival benefits; however, it also causes immune-related adverse events (irAEs) while activating the immune system, involving multiple organs. Among them, cardiovascular immune-related adverse events (CV-irAE) are rare, but common causes of death in ICIs treated cancer patients, which manifest as myocardial, pericardial, vascular and other cardiovascular toxicities. Therefore, it is important that irAEs, especially CV-irAE should be carefully recognized and monitored during the whole ICIs treatment because early detection and treatment of CV-irAE can significantly reduce the mortality of such patients. Consequently, it is urgent to fully understand the mechanism and management strategies of CV-irAE. The effects of ICIs are multifaceted and the exact mechanism of CV-irAE is still elusive. Generally, T cells identify tumor cell antigens as well as antigen in cardiomyocytes that are the same as or homologous to those on tumor cells, thus causing myocardial damage. In addition, ICIs promote formation of cardiac troponin I (cTnI) that induces cardiac dysfunction and myocardial dilatation; moreover, ICIs also increase the production of cytokines, which promote infiltration of inflammation-linked molecules into off-target tissues. Currently, the management and treatment of cardiovascular toxicity are largely dependent on glucocorticoids, more strategies for prevention and treatment of CV-irAE, such as predictive markers are being explored. This review discusses risk factors, potential pathophysiological mechanisms, clinical manifestations, and management and treatment of CV-irAE, guiding the development of more effective prevention, treatment and management strategies in the future.
Collapse
Affiliation(s)
- Xiang Li
- Department of the Second Medical Oncology, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Wenying Peng
- Department of the Second Medical Oncology, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Jiao Wu
- Department of the Second Medical Oncology, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Sai-Ching Jim Yeung
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, TX, United States
| | - Runxiang Yang
- Department of the Second Medical Oncology, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
102
|
Wong P, Cina DP, Sherwood KR, Fenninger F, Sapir-Pichhadze R, Polychronakos C, Lan J, Keown PA. Clinical application of immune repertoire sequencing in solid organ transplant. Front Immunol 2023; 14:1100479. [PMID: 36865546 PMCID: PMC9971933 DOI: 10.3389/fimmu.2023.1100479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 01/25/2023] [Indexed: 02/16/2023] Open
Abstract
Background Measurement of T cell receptor (TCR) or B cell receptor (BCR) gene utilization may be valuable in monitoring the dynamic changes in donor-reactive clonal populations following transplantation and enabling adjustment in therapy to avoid the consequences of excess immune suppression or to prevent rejection with contingent graft damage and to indicate the development of tolerance. Objective We performed a review of current literature to examine research in immune repertoire sequencing in organ transplantation and to assess the feasibility of this technology for clinical application in immune monitoring. Methods We searched MEDLINE and PubMed Central for English-language studies published between 2010 and 2021 that examined T cell/B cell repertoire dynamics upon immune activation. Manual filtering of the search results was performed based on relevancy and predefined inclusion criteria. Data were extracted based on study and methodology characteristics. Results Our initial search yielded 1933 articles of which 37 met the inclusion criteria; 16 of these were kidney transplant studies (43%) and 21 were other or general transplantation studies (57%). The predominant method for repertoire characterization was sequencing the CDR3 region of the TCR β chain. Repertoires of transplant recipients were found to have decreased diversity in both rejectors and non-rejectors when compared to healthy controls. Rejectors and those with opportunistic infections were more likely to have clonal expansion in T or B cell populations. Mixed lymphocyte culture followed by TCR sequencing was used in 6 studies to define an alloreactive repertoire and in specialized transplant settings to track tolerance. Conclusion Methodological approaches to immune repertoire sequencing are becoming established and offer considerable potential as a novel clinical tool for pre- and post-transplant immune monitoring.
Collapse
Affiliation(s)
- Paaksum Wong
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Davide P Cina
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Karen R Sherwood
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Franz Fenninger
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Ruth Sapir-Pichhadze
- Department of Medicine, Division of Nephrology, McGill University, Montreal, QC, Canada.,Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, QC, Canada
| | - Constantin Polychronakos
- Department of Pediatrics, The Research Institute of the McGill University Health Centre and the Montreal Children's Hospital, Montreal, QC, Canada
| | - James Lan
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Paul A Keown
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
103
|
Zhang Q, Geng M, Li K, Gao H, Jiao X, Ai K, Wei X, Yang J. TGF-β1 suppresses the T-cell response in teleost fish by initiating Smad3- and Foxp3-mediated transcriptional networks. J Biol Chem 2022; 299:102843. [PMID: 36581209 PMCID: PMC9860442 DOI: 10.1016/j.jbc.2022.102843] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 12/17/2022] [Accepted: 12/19/2022] [Indexed: 12/27/2022] Open
Abstract
Transforming growth factor-β1 (TGF-β1) can suppress the activation, proliferation, and function of many T-cell subsets, protecting organisms from inflammatory and autoimmune disease caused by an overexuberant immune response. However, whether and how TGF-β1 regulates T-cell immunity in early vertebrates remain unknown. Here, using a Nile tilapia (Oreochromis niloticus) model, we investigated suppression of the T-cell response by TGF-β1 in teleost species. Tilapia encodes an evolutionarily conserved TGF-β1, the expression of which in lymphocytes is significantly induced during the immune response following Edwardsiella piscicida infection. Once activated, tilapia T cells increase TGF-β1 production, which in turn suppresses proinflammatory cytokine expression and inhibits T-cell activation. Notably, we found administration of TGF-β1 cripples the proliferation of tilapia T cells, reduces the potential capacity of Th1/2 differentiation, and impairs the cytotoxic function, rendering the fish more vulnerable to bacterial infection. Mechanistically, TGF-β1 initiates the TGF-βR/Smad signaling pathway and triggers the phosphorylation and nuclear translocation of Smad2/3. Smad3 subsequently interacts with several transcriptional partners to repress transcription of cytokines IL-2 and IFN-γ but promote transcription of immune checkpoint regulator CTLA4 and transcription factor Foxp3. Furthermore, TGF-β1/Smad signaling further utilizes Foxp3 to achieve the cascade regulation of these T-cell genes. Taken together, our findings reveal a detailed mechanism by which TGF-β1 suppresses the T cell-based immunity in Nile tilapia and support the notion that TGF-β1 had already been employed to inhibit the T-cell response early in vertebrate evolution, thus providing novel insights into the evolution of the adaptive immune system.
Collapse
Affiliation(s)
- Qian Zhang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, China
| | - Ming Geng
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, China
| | - Kang Li
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, China
| | - Haiyou Gao
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, China
| | - Xinying Jiao
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, China
| | - Kete Ai
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, China
| | - Xiumei Wei
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, China.
| | - Jialong Yang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.
| |
Collapse
|
104
|
Au L, Larkin J, Turajlic S. Relatlimab and nivolumab in the treatment of melanoma. Cell 2022; 185:4866-4869. [PMID: 36563660 DOI: 10.1016/j.cell.2022.12.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 11/23/2022] [Accepted: 12/01/2022] [Indexed: 12/24/2022]
Abstract
Combined LAG-3 and PD-1 blockade is an emerging strategy for the treatment of melanoma. Tawbi et al. and Amaria et al. report in The New England Journal of Medicine and Nature respectively on two clinical trials evaluating relatlimab and nivolumab as front-line treatment for metastatic, and resectable, high-risk, node-positive melanoma.
Collapse
Affiliation(s)
- Lewis Au
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, VIC 3010, Australia; Cancer Dynamics Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - James Larkin
- Renal and Skin Units, The Royal Marsden NHS Foundation Trust, London SW3 6JJ, UK; Melanoma and Kidney Cancer Team, The Institute of Cancer Research, London SW7 3RP, UK.
| | - Samra Turajlic
- Cancer Dynamics Laboratory, The Francis Crick Institute, London NW1 1AT, UK; Renal and Skin Units, The Royal Marsden NHS Foundation Trust, London SW3 6JJ, UK; Melanoma and Kidney Cancer Team, The Institute of Cancer Research, London SW7 3RP, UK.
| |
Collapse
|
105
|
Andrews LP, Cillo AR, Karapetyan L, Kirkwood JM, Workman CJ, Vignali DAA. Molecular Pathways and Mechanisms of LAG3 in Cancer Therapy. Clin Cancer Res 2022; 28:5030-5039. [PMID: 35579997 PMCID: PMC9669281 DOI: 10.1158/1078-0432.ccr-21-2390] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/29/2022] [Accepted: 05/05/2022] [Indexed: 01/24/2023]
Abstract
Immunotherapy targeting coinhibitory receptors has been highly successful in treating a wide variety of malignancies; however, only a subset of patients exhibits durable responses. The first FDA-approved immunotherapeutics targeting coinhibitory receptors PD1 and CTLA4, alone or in combination, significantly improved survival but were also accompanied by substantial toxicity in combination. The third FDA-approved immune checkpoint inhibitor targets LAG3, a coinhibitory receptor expressed on activated CD4+ and CD8+ T cells, especially in settings of long-term antigenic stimulation, such as chronic viral infection or cancer. Mechanistically, LAG3 expression limits both the expansion of activated T cells and the size of the memory pool, suggesting that LAG3 may be a promising target for immunotherapy. Importantly, the mechanism(s) by which LAG3 contributes to CD8+ T-cell exhaustion may be distinct from those governed by PD1, indicating that the combination of anti-LAG3 and anti-PD1 may synergistically enhance antitumor immunity. Clinical studies evaluating the role of anti-LAG3 in combination with anti-PD1 are underway, and recent phase III trial results in metastatic melanoma demonstrate both the efficacy and safety of this combination. Further ongoing clinical trials are evaluating this combination across multiple tumor types and the adjuvant setting, with accompanying translational and biomarker-focused studies designed to elucidate the molecular pathways that lead to improved antitumor T-cell responses following dual blockade of PD1 and LAG3. Overall, LAG3 plays an important role in limiting T-cell activation and has now become part of the repertoire of combinatorial immunotherapeutics available for the treatment of metastatic melanoma.
Collapse
Affiliation(s)
- Lawrence P Andrews
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Anthony R Cillo
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Lilit Karapetyan
- Division of Hematology and Oncology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - John M Kirkwood
- Division of Hematology and Oncology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Creg J Workman
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Dario A A Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| |
Collapse
|
106
|
Perez-Santos M, Anaya-Ruiz M, Villafaña-Diaz L, Sánchez Esgua G. Approaches for development of LAG-3 inhibitors and the promise they hold as anticancer agents. Expert Opin Drug Discov 2022; 17:1341-1355. [PMID: 36399656 DOI: 10.1080/17460441.2022.2148652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
INTRODUCTION LAG-3 is considered to be the third point of immunological control in relation to clinical trials that address cancer treatment, only behind PD-1 and CTLA-4, due to its role as a suppressor of the immune response and enhancer of differentiation of Treg cells. AREAS COVERED The authors focus on emphasizing the strategy of development of LAG-3 inhibitors to develop anticancer therapeutics, especially from the perspective of designing new monoclonal and bispecific antibodies against LAG-3. This article also covers details of patents and clinical trials of LAG-3 inhibitors reported in the literature. In addition, we highlight as future research challenges the design and development of peptides and small molecules as inhibitors of LAG-3 function. EXPERT OPINION Three approaches have been used for the development of LAG-3 inhibitors, and they include inhibitory LAG-3 binding peptides and antagonist monoclonal and multispecific antibodies. These approaches include more than 100 clinical trials of 21 molecules that bind to LAG-3 and block its binding to MHC II. However, these approaches do not cover the design and development of peptides and small molecules that could inhibit the function of LAG-3, for which it is necessary to develop new alternatives that cover this gap.
Collapse
Affiliation(s)
- Martin Perez-Santos
- Dirección de Innovación y Transferencia de Conocimiento, Benemérita Universidad Autónoma de Puebla, Puebla CP, México
| | - Maricruz Anaya-Ruiz
- Laboratorio de Biología Celular, Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Metepec, Puebla CP, México
| | - Luis Villafaña-Diaz
- Centro de Investigación en Inteligencia de Negocios, Universidad Popular Autónoma del Estado de Puebla, Puebla, México
| | - Gabriela Sánchez Esgua
- Dirección de Innovación y Transferencia de Conocimiento, Benemérita Universidad Autónoma de Puebla, Puebla CP, México
| |
Collapse
|
107
|
Hivroz C. [LAG3 intracytoplasmic acidic environment locally disrupts the association of the Lck kinase to CD4 and CD8, two co-receptors of the TCR]. Med Sci (Paris) 2022; 38:1068-1071. [PMID: 36692275 DOI: 10.1051/medsci/2022157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Claire Hivroz
- Institut Curie, université PSL, Inserm U932, Immunité et cancer, équipe Analyse intégrative de l'activation des lymphocytes T, Paris, France
| |
Collapse
|
108
|
De Re V, Repetto O, Mussolin L, Brisotto G, Elia C, Lopci E, d’Amore ESG, Burnelli R, Mascarin M. Promising drugs and treatment options for pediatric and adolescent patients with Hodgkin lymphoma. Front Cell Dev Biol 2022; 10:965803. [PMID: 36506094 PMCID: PMC9729954 DOI: 10.3389/fcell.2022.965803] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 11/07/2022] [Indexed: 11/25/2022] Open
Abstract
Currently-available therapies for newly-diagnosed pediatric and adolescent patients with Hodgkin lymphoma result in >95% survival at 5 years. Long-term survivors may suffer from long-term treatment-related side effects, however, so the past 20 years have seen clinical trials for children and adolescents with HL gradually abandon the regimens used in adults in an effort to improve this situation. Narrower-field radiotherapy can reduce long-term toxicity while maintaining good tumor control. Various risk-adapted chemo-radiotherapy strategies have been used. Early assessment of tumor response with interim positron emission tomography and/or measuring metabolic tumor volume has been used both to limit RT in patients with favorable characteristics and to adopt more aggressive therapies in patients with a poor response. Most classical Hodgkin's lymphoma relapses occur within 3 years of initial treatment, while relapses occurring 5 years or more after diagnosis are rare. As the outcome for patients with relapsed/refractory classical Hodgkin lymphoma remains unsatisfactory, new drugs have been proposed for its prevention or treatment. This review summarizes the important advances made in recent years in the management of pediatric and adolescent with classical Hodgkin lymphoma, and the novel targeted treatments for relapsed and refractory classical Hodgkin lymphoma.
Collapse
Affiliation(s)
- Valli De Re
- Immunopatologia e Biomarcatori Oncologici, Dipartimento di Ricerca e Diagnostica Avanzata dei Tumori, CRO Aviano, National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, Aviano, Italy,*Correspondence: Valli De Re, ; Maurizio Mascarin,
| | - Ombretta Repetto
- Immunopatologia e Biomarcatori Oncologici, Dipartimento di Ricerca e Diagnostica Avanzata dei Tumori, CRO Aviano, National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, Aviano, Italy
| | - Lara Mussolin
- Pediatric Hemato-Oncology Unit, Department of Women’s and Children’s Health, University of Padua, Padua, Italy
| | - Giulia Brisotto
- Immunopatologia e Biomarcatori Oncologici, Dipartimento di Ricerca e Diagnostica Avanzata dei Tumori, CRO Aviano, National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, Aviano, Italy
| | - Caterina Elia
- AYA Oncology and Pediatric Radiotherapy Unit, Centro di Riferimento Oncologico IRCCS, Aviano, Italy
| | - Egesta Lopci
- Nuclear Medicine, IRCCS—Humanitas Research Hospital, Rozzano, MI, Italy
| | | | - Roberta Burnelli
- Pediatric Hematology-Oncology Unit, Azienda Ospedaliera Universitaria, Ospedale Sant’Anna, Ferrara, Italy
| | - Maurizio Mascarin
- AYA Oncology and Pediatric Radiotherapy Unit, Centro di Riferimento Oncologico IRCCS, Aviano, Italy,*Correspondence: Valli De Re, ; Maurizio Mascarin,
| |
Collapse
|
109
|
Mollavelioglu B, Cetin Aktas E, Cabioglu N, Abbasov A, Onder S, Emiroglu S, Tükenmez M, Muslumanoglu M, Igci A, Deniz G, Ozmen V. High co-expression of immune checkpoint receptors PD-1, CTLA-4, LAG-3, TIM-3, and TIGIT on tumor-infiltrating lymphocytes in early-stage breast cancer. World J Surg Oncol 2022; 20:349. [PMID: 36271406 DOI: 10.1186/s12957-022-02810-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 09/21/2022] [Indexed: 11/10/2022] Open
Abstract
High expression of immune checkpoint receptors (ICRs) in the tumor microenvironment regulates the anti-tumor response. In this study, the differential expressions of ICRs on tumor-infiltrating lymphocytes (TILs) in patients with early-stage breast cancer were investigated.The study included 32 patients who underwent surgery with a diagnosis of early-stage breast cancer between September 2018 and March 2020. TIL isolation was performed using a MACS tumor separation device and tumor separation kit. PD-1, CTLA-4, LAG-3, TIM-3, and TIGIT expression of cytotoxic T and natural killer (NK) cells on TILs and peripheral blood lymphocytes (PBLs) were determined by flow cytometry.Patients with a high Ki-67 index, high TIL density, and HER-2 positivity were more likely to have increased CD16+CD56dim NK cells on TILs. Patients with T2 tumors were more likely to have increased expression of PD-1, LAG-3, and TIGIT on tumor-infiltrating CD8+ cytotoxic T cells than those with T1 tumors. PD-1, CTLA-4, TIGIT, LAG-3, and TIM-3 expression of CD8+ T and CD16-CD56bright NK cells in TILs showed significant positive correlations with each other. PD1+CD8+, TIGIT+CD16+, and CTLA-4+CD56+ cells in PBLs and TILs were found to be negatively correlated, whereas only TIM-3+ expression of CD8+ T and CD16+CD56dim cells in PBLs and TILs showed positive correlations.Our results suggest that CD16+CD56dim NK cells on TILs may play a major role in the immune response against HER2-positive or highly proliferating breast tumors in patients with early-stage breast cancer. Furthermore, various ICRs were found to be highly co-expressed with each other on TILs, including PD-1, CTLA-4, LAG-3, TIM-3, and TIGIT. These receptors may synergistically suppress the response to the tumor, which may trigger immune escape mechanisms in the early stage of carcinogenesis. However, ICR expressions other than TIM3 on PBLs were not found to accompany their counterparts on TILs.
Collapse
Affiliation(s)
- Baran Mollavelioglu
- Istanbul Faculty of Medicine, Department of General Surgery, Istanbul University, Istanbul, Turkey
| | - Esin Cetin Aktas
- Department of Immunology, Istanbul University, Aziz Sancar Institute of Experimental Medicine, Istanbul, Turkey
| | - Neslihan Cabioglu
- Istanbul Faculty of Medicine, Department of General Surgery, Istanbul University, Istanbul, Turkey
| | - Aykhan Abbasov
- Istanbul Faculty of Medicine, Department of General Surgery, Istanbul University, Istanbul, Turkey
| | - Semen Onder
- Department of Pathology, Istanbul University Istanbul Faculty of Medicine, Istanbul, Turkey
| | - Selman Emiroglu
- Istanbul Faculty of Medicine, Department of General Surgery, Istanbul University, Istanbul, Turkey
| | - Mustafa Tükenmez
- Istanbul Faculty of Medicine, Department of General Surgery, Istanbul University, Istanbul, Turkey
| | - Mahmut Muslumanoglu
- Istanbul Faculty of Medicine, Department of General Surgery, Istanbul University, Istanbul, Turkey
| | - Abdullah Igci
- Istanbul Faculty of Medicine, Department of General Surgery, Istanbul University, Istanbul, Turkey
| | - Gunnur Deniz
- Department of Immunology, Istanbul University, Aziz Sancar Institute of Experimental Medicine, Istanbul, Turkey
| | - Vahit Ozmen
- Istanbul Faculty of Medicine, Department of General Surgery, Istanbul University, Istanbul, Turkey.
| |
Collapse
|
110
|
Matsumura Y, Watanabe R, Fujimoto M. Suppressive mechanisms of regulatory B cells in mice and humans. Int Immunol 2022; 35:55-65. [PMID: 36153768 PMCID: PMC9918854 DOI: 10.1093/intimm/dxac048] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 09/23/2022] [Indexed: 11/14/2022] Open
Abstract
B cells include immune-suppressive fractions, called regulatory B cells (Bregs), which regulate inflammation primarily through an interleukin 10 (IL-10)-mediated inhibitory mechanism. Several B-cell fractions have been reported as IL-10-producing Bregs in murine disease models and human inflammatory responses including autoimmune diseases, infectious diseases, cancer and organ-transplant rejection. Although the suppressive functions of Bregs have been explored through the hallmark molecule IL-10, inhibitory cytokines and membrane-binding molecules other than IL-10 have also been demonstrated to contribute to Breg activities. Transcription factors and surface antigens that are characteristically expressed in Bregs are also being elucidated. Nevertheless, defining Bregs is still challenging because their active periods and differentiation stages vary among disease models. The identity of the diverse Breg fractions is also under debate. In the first place, since regulatory functions of Bregs are mostly evaluated by ex vivo stimulation, the actual in vivo phenotypes and functions may not be reflected by the ex vivo observations. In this article, we provide a historical overview of studies that established the characteristics of Bregs and review the various suppressive mechanisms that have been reported to be used by Bregs in murine and human disease conditions. We are only part-way through but the common phenotypes and functions of Bregs are still emerging.
Collapse
Affiliation(s)
- Yutaka Matsumura
- Department of Dermatology, Graduate School of Medicine, Faculty of Medicine, Osaka University, Osaka, 565-0871, Japan
| | - Rei Watanabe
- Department of Dermatology, Graduate School of Medicine, Faculty of Medicine, Osaka University, Osaka, 565-0871, Japan,Department of Integrative Medicine for Allergic and Immunological Diseases, Graduate School of Medicine/Faculty of Medicine, Osaka University, Osaka, 565-0871, Japan
| | | |
Collapse
|
111
|
Huang J, Huang Q, Xue J, Liu H, Guo Y, Chen H, Zhou L. Fibrinogen like protein-1 knockdown suppresses the proliferation and metastasis of TU-686 cells and sensitizes laryngeal cancer to LAG-3 blockade. J Int Med Res 2022; 50:3000605221126874. [PMID: 36173010 PMCID: PMC9528049 DOI: 10.1177/03000605221126874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE To detect the expression of fibrinogen like protein-1 (FGL-1) in laryngeal cancer and evaluate its effect on tumor proliferation, metastasis, and antitumor immunity. METHODS ELISA and immunohistochemistry were performed to detect FGL-1 expression in laryngeal cancer. The effects of FGL-1 knockdown on the proliferation, cell cycle progression, apoptosis, migration, and invasion of laryngeal cancer cells were evaluated by the CCK-8, colony formation, flow cytometry, Transwell migration, and western blot assays. We detected changes in tumorigenesis and drug response in vivo following FGL-1 knockdown as well as the effects of anti-LAG3 immunotherapy. Immunohistochemistry was performed to determine CD8 and LAG-3 expression in mouse tumor tissues. RESULTS FGL-1 was highly expressed in the plasma and tumor tissues of laryngeal cancer patients. FGL-1 knockdown suppressed the proliferation of TU-686 cells and inhibited the migration and invasion of laryngeal cancer by blocking epithelial-to-mesenchymal transition. Moreover, silencing FGL-1 inhibited tumorigenicity in vivo and synergized with anti-LAG3 immunotherapy. CONCLUSIONS We confirmed the high expression of FGL-1 in laryngeal cancer and identified FGL-1 as a potential marker for immunotherapy in laryngeal cancer.
Collapse
Affiliation(s)
- Jiameng Huang
- ENT Institute and Otorhinolaryngology Department, Affiliated Eye and ENT Hospital, Fudan University, Shanghai, China
| | - Qiang Huang
- ENT Institute and Otorhinolaryngology Department, Affiliated Eye and ENT Hospital, Fudan University, Shanghai, China
| | - Jiyao Xue
- ENT Institute and Otorhinolaryngology Department, Affiliated Eye and ENT Hospital, Fudan University, Shanghai, China
| | - Huiqin Liu
- ENT Institute and Otorhinolaryngology Department, Affiliated Eye and ENT Hospital, Fudan University, Shanghai, China
| | - Yang Guo
- ENT Institute and Otorhinolaryngology Department, Affiliated Eye and ENT Hospital, Fudan University, Shanghai, China
| | - Hui Chen
- ENT Institute and Otorhinolaryngology Department, Affiliated Eye and ENT Hospital, Fudan University, Shanghai, China
| | - Liang Zhou
- ENT Institute and Otorhinolaryngology Department, Affiliated Eye and ENT Hospital, Fudan University, Shanghai, China
| |
Collapse
|
112
|
Sauer N, Szlasa W, Jonderko L, Oślizło M, Kunachowicz D, Kulbacka J, Karłowicz-Bodalska K. LAG-3 as a Potent Target for Novel Anticancer Therapies of a Wide Range of Tumors. Int J Mol Sci 2022; 23:9958. [PMID: 36077354 PMCID: PMC9456311 DOI: 10.3390/ijms23179958] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/22/2022] [Accepted: 08/29/2022] [Indexed: 12/20/2022] Open
Abstract
LAG-3 (Lymphocyte activation gene 3) protein is a checkpoint receptor that interacts with LSEC-tin, Galectin-3 and FGL1. This interaction leads to reduced production of IL-2 and IFN-γ. LAG-3 is widely expressed in different tumor types and modulates the tumor microenvironment through immunosuppressive effects. Differential expression in various tumor types influences patient prognosis, which is often associated with coexpression with immune checkpoint inhibitors, such as TIM-3, PD-1 and CTLA-4. Here, we discuss expression profiles in different tumor types. To date, many clinical trials have been conducted using LAG-3 inhibitors, which can be divided into anti-LAG-3 monoclonal antibodies, anti-LAG-3 bispecifics and soluble LAG-3-Ig fusion proteins. LAG-3 inhibitors supress T-cell proliferation and activation by disallowing for the interaction between LAG-3 to MHC-II. The process enhances anti-tumor immune response. In this paper, we will review the current state of knowledge on the structure, function and expression of LAG-3 in various types of cancer, as well as its correlation with overall prognosis, involvement in cell-based therapies and experimental medicine. We will consider the role of compounds targeting LAG-3 in clinical trials both as monotherapy and in combination, which will provide data relating to the efficacy and safety of proposed drug candidates.
Collapse
Affiliation(s)
- Natalia Sauer
- Faculty of Pharmacy, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Wojciech Szlasa
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Laura Jonderko
- Faculty of Pharmacy, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | | | | | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | | |
Collapse
|
113
|
Kuzevanova A, Apanovich N, Mansorunov D, Korotaeva A, Karpukhin A. The Features of Checkpoint Receptor—Ligand Interaction in Cancer and the Therapeutic Effectiveness of Their Inhibition. Biomedicines 2022; 10:biomedicines10092081. [PMID: 36140182 PMCID: PMC9495440 DOI: 10.3390/biomedicines10092081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/14/2022] [Accepted: 08/22/2022] [Indexed: 12/12/2022] Open
Abstract
To date, certain problems have been identified in cancer immunotherapy using the inhibition of immune checkpoints (ICs). Despite the excellent effect of cancer therapy in some cases when blocking the PD-L1 (programmed death-ligand 1) ligand and the immune cell receptors PD-1 (programmed cell death protein 1) and CTLA4 (cytotoxic T-lymphocyte-associated protein 4) with antibodies, the proportion of patients responding to such therapy is still far from desirable. This situation has stimulated the exploration of additional receptors and ligands as targets for immunotherapy. In our article, based on the analysis of the available data, the TIM-3 (T-cell immunoglobulin and mucin domain-3), LAG-3 (lymphocyte-activation gene 3), TIGIT (T-cell immunoreceptor with Ig and immunoreceptor tyrosine-based inhibitory motif (ITIM) domains), VISTA (V-domain Ig suppressor of T-cell activation), and BTLA (B- and T-lymphocyte attenuator) receptors and their ligands are comprehensively considered. Data on the relationship between receptor expression and the clinical characteristics of tumors are presented and are analyzed together with the results of preclinical and clinical studies on the therapeutic efficacy of their blocking. Such a comprehensive analysis makes it possible to assess the prospects of receptors of this series as targets for anticancer therapy. The expression of the LAG-3 receptor shows the most unambiguous relationship with the clinical characteristics of cancer. Its inhibition is the most effective of the analyzed series in terms of the antitumor response. The expression of TIGIT and BTLA correlates well with clinical characteristics and demonstrates antitumor efficacy in preclinical and clinical studies, which indicates their high promise as targets for anticancer therapy. At the same time, the relationship of VISTA and TIM-3 expression with the clinical characteristics of the tumor is contradictory, and the results on the antitumor effectiveness of their inhibition are inconsistent.
Collapse
|
114
|
Lamberti MJ, Montico B, Ravo M, Nigro A, Giurato G, Iorio R, Tarallo R, Weisz A, Stellato C, Steffan A, Dolcetti R, Casolaro V, Faè DA, Dal Col J. Integration of miRNA:mRNA Co-Expression Revealed Crucial Mechanisms Modulated in Immunogenic Cancer Cell Death. Biomedicines 2022; 10:biomedicines10081896. [PMID: 36009442 PMCID: PMC9405340 DOI: 10.3390/biomedicines10081896] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/23/2022] [Accepted: 07/29/2022] [Indexed: 11/21/2022] Open
Abstract
Immunogenic cell death (ICD) in cancer represents a functionally unique therapeutic response that can induce tumor-targeting immune responses. ICD is characterized by the exposure and release of numerous damage-associated molecular patterns (DAMPs), which confer adjuvanticity to dying cancer cells. The spatiotemporally defined emission of DAMPs during ICD has been well described, whereas the epigenetic mechanisms that regulate ICD hallmarks have not yet been deeply elucidated. Here, we aimed to examine the involvement of miRNAs and their putative targets using well-established in vitro models of ICD. To this end, B cell lymphoma (Mino) and breast cancer (MDA-MB-231) cell lines were exposed to two different ICD inducers, the combination of retinoic acid (RA) and interferon-alpha (IFN-α) and doxorubicin, and to non ICD inducers such as gamma irradiation. Then, miRNA and mRNA profiles were studied by next generation sequencing. Co-expression analysis identified 16 miRNAs differentially modulated in cells undergoing ICD. Integrated miRNA-mRNA functional analysis revealed candidate miRNAs, mRNAs, and modulated pathways associated with Immune System Process (GO Term). Specifically, ICD induced a distinctive transcriptional signature hallmarked by regulation of antigen presentation, a crucial step for proper activation of immune system antitumor response. Interestingly, the major histocompatibility complex class I (MHC-I) pathway was upregulated whereas class II (MHC-II) was downregulated. Analysis of MHC-II associated transcripts and HLA-DR surface expression confirmed inhibition of this pathway by ICD on lymphoma cells. miR-4284 and miR-212-3p were the strongest miRNAs upregulated by ICD associated with this event and miR-212-3p overexpression was able to downregulate surface expression of HLA-DR. It is well known that MHC-II expression on tumor cells facilitates the recruitment of CD4+ T cells. However, the interaction between tumor MHC-II and inhibitory coreceptors on tumor-associated lymphocytes could provide an immunosuppressive signal that directly represses effector cytotoxic activity. In this context, MHC-II downregulation by ICD could enhance antitumor immunity. Overall, we found that the miRNA profile was significantly altered during ICD. Several miRNAs are predicted to be involved in the regulation of MHC-I and II pathways, whose implication in ICD is demonstrated herein for the first time, which could eventually modulate tumor recognition and attack by the immune system.
Collapse
Affiliation(s)
- María Julia Lamberti
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, SA, Italy
- INBIAS, CONICET-UNRC, Río Cuarto, Córdoba 5800, Argentina
- Correspondence: (M.J.L.); (J.D.C.); Tel.: +54-358-4676437 (M.J.L.); +39-089-965210 (J.D.C.)
| | - Barbara Montico
- Immunopathology and Cancer Biomarkers, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, PN, Italy
| | - Maria Ravo
- Genomix Life Srl, 84081 Baronissi, SA, Italy
| | - Annunziata Nigro
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, SA, Italy
| | - Giorgio Giurato
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, SA, Italy
| | | | - Roberta Tarallo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, SA, Italy
| | - Alessandro Weisz
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, SA, Italy
| | - Cristiana Stellato
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, SA, Italy
| | - Agostino Steffan
- Immunopathology and Cancer Biomarkers, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, PN, Italy
| | - Riccardo Dolcetti
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3010, Australia
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Vincenzo Casolaro
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, SA, Italy
| | - Damiana Antonia Faè
- Immunopathology and Cancer Biomarkers, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, PN, Italy
| | - Jessica Dal Col
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, SA, Italy
- Correspondence: (M.J.L.); (J.D.C.); Tel.: +54-358-4676437 (M.J.L.); +39-089-965210 (J.D.C.)
| |
Collapse
|
115
|
Sung E, Ko M, Won JY, Jo Y, Park E, Kim H, Choi E, Jung UJ, Jeon J, Kim Y, Ahn H, Choi DS, Choi S, Hong Y, Park H, Lee H, Son YG, Park K, Won J, Oh SJ, Lee S, Kim KP, Yoo C, Song HK, Jin HS, Jung J, Park Y. LAG-3xPD-L1 bispecific antibody potentiates antitumor responses of T cells through dendritic cell activation. Mol Ther 2022; 30:2800-2816. [PMID: 35526096 PMCID: PMC9372323 DOI: 10.1016/j.ymthe.2022.05.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 04/08/2022] [Accepted: 05/03/2022] [Indexed: 11/26/2022] Open
Abstract
Several preclinical studies demonstrate that antitumor efficacy of programmed cell death-1 (PD-1)/programmed death-ligand 1 (PD-L1) blockade can be improved by combination with other checkpoint inhibitors. Lymphocyte-activation gene 3 (LAG-3) is an inhibitory checkpoint receptor involved in T cell exhaustion and tumor immune escape. Here, we describe ABL501, a bispecific antibody targeting LAG-3 and PD-L1 in modulating immune cell responses against tumors. ABL501 that efficiently inhibits both LAG-3 and PD-L1 pathways enhances the activation of effector CD4+ and CD8+ T cells with a higher degree than a combination of single anti-LAG-3 and anti-PD-L1. The augmented effector T cell responses by ABL501 resulted in mitigating regulatory-T-cell-mediated immunosuppression. Mechanistically, the simultaneous binding of ABL501 to LAG-3 and PD-L1 promotes dendritic cell (DC) activation and tumor cell conjugation with T cells that subsequently mounts effective CD8+ T cell responses. ABL501 demonstrates its potent in vivo antitumor efficacy in a humanized xenograft model and with knockin mice expressing human orthologs. The immune profiling analysis of peripheral blood reveals an increased abundance of LAG-3hiPD-1hi memory CD4+ T cell subset in relapsed cholangiocarcinoma patients after gemcitabine plus cisplatin therapy, which are more responsive to ABL501. This study supports the clinical evaluation of ABL501 as a novel cancer immunotherapeutic, and a first-in-human trial has started (NCT05101109).
Collapse
Affiliation(s)
| | - Minkyung Ko
- Theragnosis Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, South Korea
| | - Ju-Young Won
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Yunju Jo
- Theragnosis Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, South Korea; Department of Life Sciences, Korea University, Seoul 02481, South Korea
| | | | | | - Eunji Choi
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | | | | | | | - Hyejin Ahn
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Da-Som Choi
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Seunghyun Choi
- Theragnosis Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, South Korea
| | | | | | | | | | | | | | - Soo Jin Oh
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Seonmin Lee
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Kyu-Pyo Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Changhoon Yoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Hyun Kyu Song
- Department of Life Sciences, Korea University, Seoul 02481, South Korea
| | - Hyung-Seung Jin
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea.
| | - Jaeho Jung
- ABL Bio Inc., Seongnam 13488, South Korea.
| | - Yoon Park
- Theragnosis Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, South Korea.
| |
Collapse
|
116
|
Huo JL, Wang YT, Fu WJ, Lu N, Liu ZS. The promising immune checkpoint LAG-3 in cancer immunotherapy: from basic research to clinical application. Front Immunol 2022; 13:956090. [PMID: 35958563 PMCID: PMC9361790 DOI: 10.3389/fimmu.2022.956090] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 07/07/2022] [Indexed: 12/13/2022] Open
Abstract
LAG-3, a type of immune checkpoint receptor protein belonging to the immunoglobulin superfamily, is confirmed to be expressed on activated immune cells, mainly including activated T cells. LAG-3 can negatively regulate the function of T cells, exerting important effects on maintaining the homeostasis of the immune system under normal physiological conditions and promoting tumor cells immune escape in the tumor microenvironment. Given its important biological roles, LAG-3 has been regarded as a promising target for cancer immunotherapy. To date, many LAG-3 inhibitors have been reported, which can be divided into monoclonal antibody, double antibody, and small molecule drug, some of which have entered the clinical research stage. LAG-3 inhibitors can negatively regulate and suppress T cell proliferation and activation through combination with MHC II ligand. Besides, LAG-3 inhibitors can also affect T cell function via binding to Galectin-3 and LSECtin. In addition, LAG-3 inhibitors can prevent the FGL1-LAG-3 interaction, thereby enhancing the human body’s antitumor immune effect. In this review, we will describe the function of LAG-3 and summarize the latest LAG-3 inhibitors in the clinic for cancer therapy.
Collapse
Affiliation(s)
- Jin-Ling Huo
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Research Institute of Nephrology, Zhengzhou University, Henan Province Research Center For Kidney Disease, Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Ya-Tao Wang
- Department of Orthopedics, First People’s Hospital of Shangqiu, Shangqiu, China
| | - Wen-Jia Fu
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Research Institute of Nephrology, Zhengzhou University, Henan Province Research Center For Kidney Disease, Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Nan Lu
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore, Singapore
- *Correspondence: Nan Lu, ; Zhang-Suo Liu,
| | - Zhang-Suo Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Research Institute of Nephrology, Zhengzhou University, Henan Province Research Center For Kidney Disease, Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
- *Correspondence: Nan Lu, ; Zhang-Suo Liu,
| |
Collapse
|
117
|
Lao Y, Shen D, Zhang W, He R, Jiang M. Immune Checkpoint Inhibitors in Cancer Therapy—How to Overcome Drug Resistance? Cancers (Basel) 2022; 14:cancers14153575. [PMID: 35892835 PMCID: PMC9331941 DOI: 10.3390/cancers14153575] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/15/2022] [Accepted: 07/19/2022] [Indexed: 12/04/2022] Open
Abstract
Simple Summary Immune checkpoint inhibitors (ICIs) are an important strategy in cancer therapy. However, with the widespread clinical use of ICIs, people gradually found that ICIs may not be effective enough to eliminate tumor tissue for certain patients. The resistance to ICI treatment makes some patients unable to benefit from their antitumor effects. Therefore, it is vital to understand their antitumor and drug resistance mechanisms to better narrow the ICI-resistant patient population. This review outlines the antitumor action sites and mechanisms of different types of ICIs and lists the main reason of ICI resistance based on recent studies. Finally, we propose current and future solutions for resistance to ICIs. Abstract Immune checkpoint inhibitors (ICIs), antagonists used to remove tumor suppression of immune cells, have been widely used in clinical settings. Their high antitumor effect makes them crucial for treating cancer after surgery, radiotherapy, chemotherapy, and targeted therapy. However, with the advent of ICIs and their use by a large number of patients, more clinical data have gradually shown that some cancer patients still have resistance to ICI treatment, which makes some patients unable to benefit from their antitumor effect. Therefore, it is vital to understand their antitumor and drug resistance mechanisms. In this review, we focused on the antitumor action sites and mechanisms of different types of ICIs. We then listed the main possible mechanisms of ICI resistance based on recent studies. Finally, we proposed current and future solutions for the resistance of ICIs, providing theoretical support for improving their clinical antitumor effect.
Collapse
Affiliation(s)
- Yefang Lao
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China;
| | - Daoming Shen
- Department of Internal Medicine, Xiangcheng People’s Hospital, Suzhou 215131, China;
| | - Weili Zhang
- Department of Gastroenterology, Xiangcheng People’s Hospital, Suzhou 215131, China;
| | - Rui He
- Department of Pneumoconiosis, Shanghai Pulmonary Hospital, Shanghai 200433, China
- Correspondence: (R.H.); (M.J.); Tel.: +86-18862185684 (R.H.); +86-13776022109 (M.J.)
| | - Min Jiang
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China;
- Correspondence: (R.H.); (M.J.); Tel.: +86-18862185684 (R.H.); +86-13776022109 (M.J.)
| |
Collapse
|
118
|
Aristin Revilla S, Kranenburg O, Coffer PJ. Colorectal Cancer-Infiltrating Regulatory T Cells: Functional Heterogeneity, Metabolic Adaptation, and Therapeutic Targeting. Front Immunol 2022; 13:903564. [PMID: 35874729 PMCID: PMC9304750 DOI: 10.3389/fimmu.2022.903564] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 06/06/2022] [Indexed: 11/18/2022] Open
Abstract
Colorectal cancer (CRC) is a heterogeneous disease with one of the highest rates of incidence and mortality among cancers worldwide. Understanding the CRC tumor microenvironment (TME) is essential to improve diagnosis and treatment. Within the CRC TME, tumor-infiltrating lymphocytes (TILs) consist of a heterogeneous mixture of adaptive immune cells composed of mainly anti-tumor effector T cells (CD4+ and CD8+ subpopulations), and suppressive regulatory CD4+ T (Treg) cells. The balance between these two populations is critical in anti-tumor immunity. In general, while tumor antigen-specific T cell responses are observed, tumor clearance frequently does not occur. Treg cells are considered to play an important role in tumor immune escape by hampering effective anti-tumor immune responses. Therefore, CRC-tumors with increased numbers of Treg cells have been associated with promoting tumor development, immunotherapy failure, and a poorer prognosis. Enrichment of Treg cells in CRC can have multiple causes including their differentiation, recruitment, and preferential transcriptional and metabolic adaptation to the TME. Targeting tumor-associated Treg cell may be an effective addition to current immunotherapy approaches. Strategies for depleting Treg cells, such as low-dose cyclophosphamide treatment, or targeting one or more checkpoint receptors such as CTLA-4 with PD-1 with monoclonal antibodies, have been explored. These have resulted in activation of anti-tumor immune responses in CRC-patients. Overall, it seems likely that CRC-associated Treg cells play an important role in determining the success of such therapeutic approaches. Here, we review our understanding of the role of Treg cells in CRC, the possible mechanisms that support their homeostasis in the tumor microenvironment, and current approaches for manipulating Treg cells function in cancer.
Collapse
Affiliation(s)
- Sonia Aristin Revilla
- Center Molecular Medicine, University Medical Center Utrecht, Utrecht, Netherlands
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, Netherlands
- Laboratory Translational Oncology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Onno Kranenburg
- Laboratory Translational Oncology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Paul J. Coffer
- Center Molecular Medicine, University Medical Center Utrecht, Utrecht, Netherlands
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, Netherlands
- *Correspondence: Paul J. Coffer,
| |
Collapse
|
119
|
Mørup SB, Nazaryan-Petersen L, Gabrielaite M, Reekie J, Marquart HV, Hartling HJ, Marvig RL, Katzenstein TL, Masmas TN, Lundgren J, Murray DD, Helleberg M, Borgwardt L. Added Value of Reanalysis of Whole Exome- and Whole Genome Sequencing Data From Patients Suspected of Primary Immune Deficiency Using an Extended Gene Panel and Structural Variation Calling. Front Immunol 2022; 13:906328. [PMID: 35874679 PMCID: PMC9302041 DOI: 10.3389/fimmu.2022.906328] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/13/2022] [Indexed: 11/13/2022] Open
Abstract
Background Knowledge of the genetic variation underlying Primary Immune Deficiency (PID) is increasing. Reanalysis of genome-wide sequencing data from undiagnosed patients with suspected PID may improve the diagnostic rate. Methods We included patients monitored at the Department of Infectious Diseases or the Child and Adolescent Department, Rigshospitalet, Denmark, for a suspected PID, who had been analysed previously using a targeted PID gene panel (457 PID-related genes) on whole exome- (WES) or whole genome sequencing (WGS) data. A literature review was performed to extend the PID gene panel used for reanalysis of single nucleotide variation (SNV) and small indels. Structural variant (SV) calling was added on WGS data. Results Genetic data from 94 patients (86 adults) including 36 WES and 58 WGS was reanalysed a median of 23 months after the initial analysis. The extended gene panel included 208 additional PID-related genes. Genetic reanalysis led to a small increase in the proportion of patients with new suspicious PID related variants of uncertain significance (VUS). The proportion of patients with a causal genetic diagnosis was constant. In total, five patients (5%, including three WES and two WGS) had a new suspicious PID VUS identified due to reanalysis. Among these, two patients had a variant added due to the expansion of the PID gene panel, and three patients had a variant reclassified to a VUS in a gene included in the initial PID gene panel. The total proportion of patients with PID related VUS, likely pathogenic, and pathogenic variants increased from 43 (46%) to 47 (50%), as one patient had a VUS detected in both initial- and reanalysis. In addition, we detected new suspicious SNVs and SVs of uncertain significance in PID candidate genes with unknown inheritance and/or as heterozygous variants in genes with autosomal recessive inheritance in 8 patients. Conclusion These data indicate a possible diagnostic gain of reassessing WES/WGS data from patients with suspected PID. Reasons for the possible gain included improved knowledge of genotype-phenotype correlation, expanding the gene panel, and adding SV analyses. Future studies of genotype-phenotype correlations may provide additional knowledge on the impact of the new suspicious VUSs.
Collapse
Affiliation(s)
- Sara Bohnstedt Mørup
- Centre of Excellence for Health, Immunity, and Infections, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Lusine Nazaryan-Petersen
- Center for Genomic Medicine, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Migle Gabrielaite
- Center for Genomic Medicine, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Joanne Reekie
- Centre of Excellence for Health, Immunity, and Infections, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Hanne V. Marquart
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Hans Jakob Hartling
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Rasmus L. Marvig
- Center for Genomic Medicine, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Terese L. Katzenstein
- Department of Infectious Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Tania N. Masmas
- The Child and Adolescent Department, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Jens Lundgren
- Centre of Excellence for Health, Immunity, and Infections, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Infectious Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Daniel D. Murray
- Centre of Excellence for Health, Immunity, and Infections, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Marie Helleberg
- Centre of Excellence for Health, Immunity, and Infections, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Infectious Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Line Borgwardt
- Center for Genomic Medicine, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
120
|
LAG3 ectodomain structure reveals functional interfaces for ligand and antibody recognition. Nat Immunol 2022; 23:1031-1041. [PMID: 35761082 DOI: 10.1038/s41590-022-01238-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 05/11/2022] [Indexed: 01/05/2023]
Abstract
The immune checkpoint receptor lymphocyte activation gene 3 protein (LAG3) inhibits T cell function upon binding to major histocompatibility complex class II (MHC class II) or fibrinogen-like protein 1 (FGL1). Despite the emergence of LAG3 as a target for next-generation immunotherapies, we have little information describing the molecular structure of the LAG3 protein or how it engages cellular ligands. Here we determined the structures of human and murine LAG3 ectodomains, revealing a dimeric assembly mediated by Ig domain 2. Epitope mapping indicates that a potent LAG3 antagonist antibody blocks interactions with MHC class II and FGL1 by binding to a flexible 'loop 2' region in LAG3 domain 1. We also defined the LAG3-FGL1 interface by mapping mutations onto structures of LAG3 and FGL1 and established that FGL1 cross-linking induces the formation of higher-order LAG3 oligomers. These insights can guide LAG3-based drug development and implicate ligand-mediated LAG3 clustering as a mechanism for disrupting T cell activation.
Collapse
|
121
|
Petersen J, Rossjohn J. Overcoming the LAG3 phase problem. Nat Immunol 2022; 23:993-995. [PMID: 35761086 PMCID: PMC9243723 DOI: 10.1038/s41590-022-01239-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- Jan Petersen
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Jamie Rossjohn
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia. .,Institute of Infection and Immunity, Cardiff University, School of Medicine, Cardiff, UK.
| |
Collapse
|
122
|
Liu YF, Zhang ZC, Wang SY, Fu SQ, Cheng XF, Chen R, Sun T. Immune checkpoint inhibitor-based therapy for advanced clear cell renal cell carcinoma: A narrative review. Int Immunopharmacol 2022; 110:108900. [PMID: 35753122 DOI: 10.1016/j.intimp.2022.108900] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 05/19/2022] [Accepted: 05/24/2022] [Indexed: 12/12/2022]
Abstract
The prognosis for advanced clear cell renal cell carcinoma (ccRCC) is not satisfactory, even though its treatment has evolved rapidly over the past 20 years. Systemic ccRCC treatment options mainly involve antiangiogenic therapy, immune checkpoint blockade, or a combination of these therapies, and as more clinical evidence becomes available, immune checkpoint inhibitors (ICIs) are increasingly dominant. Conventional ICIs lead to the restoration of T-cell activation and a reduction in T-cell depletion by specifically blocking programmed cell death 1 (PD-1), programmed cell death 1 ligand 1 (PD-L1) or cytotoxic T lymphocyte antigen 4 (CTLA-4), ultimately enhancing the antitumor immune response. There is no doubt that these therapies have achieved some clinical efficacy in the overall ccRCC population, but response rates and durability remain a great challenge. Therefore, novel immune checkpoints or new combination therapeutic strategies based on ICIs continue to be sought and developed. This review will provide a comprehensive overview of ICI-based therapeutic strategies in advanced ccRCC, including their mechanisms of action and the latest clinical evidence.
Collapse
Affiliation(s)
- Yi-Fu Liu
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang 330000, Jiangxi Province, China; Jiangxi Institute of Urology, Nanchang 330000, Jiangxi Province, China
| | - Zhi-Cheng Zhang
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang 330000, Jiangxi Province, China; Jiangxi Institute of Urology, Nanchang 330000, Jiangxi Province, China
| | - Si-Yuan Wang
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang 330000, Jiangxi Province, China; Jiangxi Institute of Urology, Nanchang 330000, Jiangxi Province, China
| | - Sheng-Qiang Fu
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang 330000, Jiangxi Province, China; Jiangxi Institute of Urology, Nanchang 330000, Jiangxi Province, China
| | - Xiao-Feng Cheng
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang 330000, Jiangxi Province, China; Jiangxi Institute of Urology, Nanchang 330000, Jiangxi Province, China
| | - Ru Chen
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang 330000, Jiangxi Province, China; Jiangxi Institute of Urology, Nanchang 330000, Jiangxi Province, China
| | - Ting Sun
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang 330000, Jiangxi Province, China; Jiangxi Institute of Urology, Nanchang 330000, Jiangxi Province, China.
| |
Collapse
|
123
|
Greisen SR, Aspari M, Deleuran B. Co-Inhibitory Molecules – Their Role in Health and Autoimmunity; Highlighted by Immune Related Adverse Events. Front Immunol 2022; 13:883733. [PMID: 35784333 PMCID: PMC9243421 DOI: 10.3389/fimmu.2022.883733] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/10/2022] [Indexed: 12/18/2022] Open
Abstract
Immune checkpoint receptors are key players in regulating the immune response. They are responsible for both generating an immune response sufficient to kill invading pathogens, balancing the same response, and protecting against tissue destruction or the development of autoimmune events. The central role of the co-inhibitory receptors also referred to as inhibitory immune checkpoints, including PD-1 and CTLA-4 has become especially evident with the cancer treatments targeting these receptors. Blocking these pathways enhances the immune activity, resulting in both an increased chance of cancer clearance, at the same time induction of immune-related adverse events (irAE). Some of these irAE progress into actual autoimmune diseases with autoantibodies and symptoms, undistinguished from the naturally occurring diseases. This review will take advantage of the lessons learned from immune checkpoint blockade and relate this knowledge to our understanding of the same pathways in naturally occurring autoimmune diseases, mainly focusing on rheumatic diseases.
Collapse
Affiliation(s)
- Stinne R. Greisen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark
- *Correspondence: Stinne R. Greisen,
| | - Maithri Aspari
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Bent Deleuran
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
124
|
Li YJ, Chen Z. Cell-based therapies for rheumatoid arthritis: opportunities and challenges. Ther Adv Musculoskelet Dis 2022; 14:1759720X221100294. [PMID: 35634355 PMCID: PMC9131381 DOI: 10.1177/1759720x221100294] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 04/26/2022] [Indexed: 11/18/2022] Open
Abstract
Rheumatoid arthritis (RA) is the most common immune-mediated inflammatory disease characterized by chronic synovitis that hardly resolves spontaneously. The current treatment of RA consists of nonsteroidal anti-inflammatory drugs (NSAIDs), glucocorticoids, conventional disease-modifying antirheumatic drugs (cDMARDs), biologic and targeted synthetic DMARDs. Although the treat-to-target strategy has been intensively applied in the past decade, clinical unmet needs still exist since a substantial proportion of patients are refractory or even develop severe adverse effects to current therapies. In recent years, with the deeper understanding of immunopathogenesis of the disease, cell-based therapies have exhibited effective and promising interventions to RA. Several cell-based therapies, such as mesenchymal stem cells (MSC), adoptive transfer of regulatory T cells (Treg), and chimeric antigen receptor (CAR)-T cell therapy as well as their beneficial effects have been documented and verified so far. In this review, we summarize the current evidence and discuss the prospect as well as challenges for these three types of cellular therapies in RA.
Collapse
Affiliation(s)
- Yu-Jing Li
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Second Clinical Medical School, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | | |
Collapse
|
125
|
Hivroz C. LAG3 disrupts the TCR signal by local acidification. Nat Immunol 2022; 23:649-651. [PMID: 35477982 DOI: 10.1038/s41590-022-01196-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Claire Hivroz
- Institut Curie, PSL University, Inserm U932, Immunity and Cancer, Paris, France.
| |
Collapse
|
126
|
Wei Y, Li Z. LAG3-PD-1 Combo Overcome the Disadvantage of Drug Resistance. Front Oncol 2022; 12:831407. [PMID: 35494015 PMCID: PMC9048820 DOI: 10.3389/fonc.2022.831407] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/22/2022] [Indexed: 12/13/2022] Open
Abstract
Although PD-1 blockade therapy has been promising in cancer treatment, only 4% (pancreatic cancer) to 70% (melanoma) of patients have a positive response to this blockade therapy, which is one of its important disadvantages. Therefore, it is important to seek out new targets for cancer immunotherapy to improve the overall response rate in patients. Lymphocyte activation gene-3 (LAG-3), an immune checkpoint receptor, is mainly expressed in activated immune cells. LAG-3 maintains the body’s immune homeostasis under physiological conditions while mediating tumour immune escape. Several preclinical and clinical examinations have shown that LAG-3 blockade effectively alleviates the patient’s tolerance to PD-1 immune checkpoint inhibitors. Moreover, the combination of LAG-3 and PD-1 blockade has good clinical efficacy in cancers. Hence, synchronous LAG-3 and PD-1 inhibition may be a potential new strategy for tumour immunotherapy.
Collapse
|
127
|
Maruhashi T, Sugiura D, Okazaki IM, Shimizu K, Maeda TK, Ikubo J, Yoshikawa H, Maenaka K, Ishimaru N, Kosako H, Takemoto T, Okazaki T. Binding of LAG-3 to stable peptide-MHC class II limits T cell function and suppresses autoimmunity and anti-cancer immunity. Immunity 2022; 55:912-924.e8. [PMID: 35413245 DOI: 10.1016/j.immuni.2022.03.013] [Citation(s) in RCA: 104] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 01/25/2022] [Accepted: 03/16/2022] [Indexed: 12/12/2022]
Abstract
Lymphocyte activation gene-3 (LAG-3) is a potent inhibitory co-receptor; yet, its functional ligand remains elusive, with distinct potential ligands identified. Here, we investigated the relative contribution of potential ligands, stable peptide-MHC class II complexes (pMHCII) and fibrinogen-like protein 1 (FGL1), to LAG-3 activity in vitro and in vivo. Binding of LAG-3 to stable pMHCII but not to FGL1 induced T cell suppression in vitro. Consistently, LAG-3 mutants lacking FGL1-binding capacity but not those lacking stable pMHCII-binding capacity retained suppressive activity in vitro. Accordingly, targeted disruption of stable pMHCII- but not FGL1-binding capacity of LAG-3 in NOD mice recapitulated diabetes exacerbation by LAG-3 deficiency. Additionally, the loss of stable pMHCII-binding capacity of LAG-3 augmented anti-cancer immunity comparably with LAG-3 deficiency in C57BL/6 mice. These results identify stable pMHCII as a functional ligand of LAG-3 both in autoimmunity and anti-cancer immunity. Thus, stable pMHCII-LAG-3 interaction is a potential therapeutic target in human diseases.
Collapse
Affiliation(s)
- Takumi Maruhashi
- Laboratory of Molecular Immunology, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo 113-0032, Japan
| | - Daisuke Sugiura
- Laboratory of Molecular Immunology, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo 113-0032, Japan
| | - Il-Mi Okazaki
- Laboratory of Molecular Immunology, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo 113-0032, Japan; Division of Immune Regulation, Institute of Advanced Medical Sciences, Tokushima University, Tokushima 770-8503, Japan
| | - Kenji Shimizu
- Laboratory of Molecular Immunology, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo 113-0032, Japan
| | - Takeo K Maeda
- Division of Immune Regulation, Institute of Advanced Medical Sciences, Tokushima University, Tokushima 770-8503, Japan
| | - Jun Ikubo
- Division of Immune Regulation, Institute of Advanced Medical Sciences, Tokushima University, Tokushima 770-8503, Japan
| | - Harunori Yoshikawa
- Division of Cell Signaling, Fujii Memorial Institute of Medical Sciences, Institute of Advanced Medical Sciences, Tokushima University, Tokushima 770-8503, Japan
| | - Katsumi Maenaka
- Laboratory of Biomolecular Science, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Naozumi Ishimaru
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8504, Japan
| | - Hidetaka Kosako
- Division of Cell Signaling, Fujii Memorial Institute of Medical Sciences, Institute of Advanced Medical Sciences, Tokushima University, Tokushima 770-8503, Japan
| | - Tatsuya Takemoto
- Laboratory of Embryology, Institute of Advanced Medical Sciences, Tokushima University, Tokushima 770-8503, Japan
| | - Taku Okazaki
- Laboratory of Molecular Immunology, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo 113-0032, Japan; Division of Immune Regulation, Institute of Advanced Medical Sciences, Tokushima University, Tokushima 770-8503, Japan.
| |
Collapse
|
128
|
Strategies targeting tumor immune and stromal microenvironment and their clinical relevance. Adv Drug Deliv Rev 2022; 183:114137. [PMID: 35143893 DOI: 10.1016/j.addr.2022.114137] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/23/2022] [Accepted: 02/03/2022] [Indexed: 12/13/2022]
Abstract
The critical role of tumor microenvironment (TME) in tumor initiation and development has been well-recognized after more than a century of studies. Numerous therapeutic approaches targeting TME are rapidly developed including those leveraging nanotechnology, which have been further accelerated since the emergence of immune checkpoint blockade therapies in the past decade. While there are many reviews focusing on TME remodeling therapies via drug delivery and engineering strategies in animal models, state-of-the-art evaluation of clinical development states of TME-targeted therapeutics is rarely found. Here, we illustrate opportunities for integrating nano-delivery system for the development of TME-specific therapeutic regimen, followed by a comprehensive summary of the most up to date approved or clinically evaluated therapeutics targeting cellular and extracellular components within tumor immune and stromal microenvironment, including small molecule and monoclonal antibody drugs as well as nanomedicines. In the end, we also discuss challenges and possible solutions for clinical translation of TME-targeted nanomedicines.
Collapse
|
129
|
Walwyn-Brown K, Pugh J, Cocker AT, Beyzaie N, Singer BB, Olive D, Guethlein LA, Parham P, Djaoud Z. Phosphoantigen-stimulated γδ T cells suppress natural killer cell-responses to missing-self. Cancer Immunol Res 2022; 10:558-570. [PMID: 35263761 DOI: 10.1158/2326-6066.cir-21-0696] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 01/14/2022] [Accepted: 03/04/2022] [Indexed: 11/16/2022]
Abstract
γδ T cells stimulated by phosphoantigens (pAg) are potent effectors that secrete Th1 cytokines and kill tumor cells. Consequently, they are considered candidates for use in cancer immunotherapy. However, they have proven only moderately effective in several clinical trials. We studied the consequences of pAg-stimulated γδ T-cell interactions with Natural Killer (NK) cells and CD8+ T cells, major innate and adaptive effectors, respectively. We found that pAg-stimulated γδ T cells suppressed NK-cell responses to "missing-self" but had no effect on antigen-specific CD8+ T-cell responses. Extensive analysis of the secreted cytokines showed that pAg-stimulated γδ T cells had a pro-inflammatory profile. CMV-pp65-specific CD8+ T cells primed with pAg-stimulated γδ T cells showed little effect on responses to pp65-loaded target cells. By contrast, NK cells primed similarly with γδ T cells had impaired capacity to degranulate and produce IFNγ in response to HLA class I-deficient targets. This effect depended on BTN3A1 and required direct contact between NK cells and γδ T cells. γδ T cell-priming of NK cells also led to a downregulation of NKG2D and NKp44 on NK cells. Every NK-cell subset was affected by γδ T cell-mediated immunosuppression, but the strongest effect was on KIR+NKG2A- NK cells. We therefore report a previously unknown function for γδ T cells, as brakes of NK-cell responses to "missing-self". This provides a new perspective for optimizing the use of γδ T cells in cancer immunotherapy and for assessing their role in immune responses to pAg-producing pathogens.
Collapse
Affiliation(s)
| | | | | | | | | | - Daniel Olive
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, CRCM,, Marseille, France
| | | | | | | |
Collapse
|
130
|
Abstract
Exhaustion of T cells occurs in response to long-term exposure to self and foreign antigens. It limits T cell capacity to proliferate and produce cytokines, leading to an impaired ability to clear chronic infections or eradicate tumors. T-cell exhaustion is associated with a specific transcriptional, epigenetic, and metabolic program and characteristic cell surface markers' expression. Recent studies have begun to elucidate the role of T-cell exhaustion in transplant. Higher levels of exhausted T cells have been associated with better graft function in kidney transplant recipients. In contrast, reinvigorating exhausted T cells by immune checkpoint blockade therapies, while promoting tumor clearance, increases the risk of acute rejection. Lymphocyte depletion and high alloantigen load have been identified as major drivers of T-cell exhaustion. This could account, at least in part, for the reduced rates of acute rejection in organ transplant recipients induced with thymoglobulin and for the pro-tolerogenic effects of a large organ such as the liver. Among the drugs that are widely used for maintenance immunosuppression, calcineurin inhibitors have a contrasting inhibitory effect on exhaustion of T cells, while the influence of mTOR inhibitors is still unclear. Harnessing or encouraging the natural processes of exhaustion may provide a novel strategy to promote graft survival and transplantation tolerance.
Collapse
|
131
|
Tian J, Liu Y, Zhang T, Yue L, Xiao Y, Guo C. LAG-3 is a promising inhibitory immune checkpoint for antitumor immunotherapy. Expert Rev Anticancer Ther 2022; 22:289-296. [PMID: 35132925 DOI: 10.1080/14737140.2022.2039124] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
INTRODUCTION Lymphocyte activation gene-3 (LAG-3) is a member of the immunoglobulin superfamily. Engagement of LAG-3 by its ligands to trigger downstream signaling can inhibit immune responses and regulate the pathogenesis of many diseases, including cancer and inflammatory diseases. AREAS COVERED We used keywords to search for relevant publications in PubMed and information on websites. After systematic analysis, we discuss the biological characteristics of LAG-3 and its ligands, LAG-3 related signaling, its roles in the pathogenesis of tumors, and its blockages for the treatment of cancers, as well as current challenges and future directions of research. EXPERT OPINION Although the mechanisms underlying the action of LAG-3/ligand-related signaling in tumor development are not fully understood, advances in scientific research and LAG-3-based immunotherapies are promising. Further studies to explore its biological roles and molecular mechanisms may aid in developing new LAG-3- and ligand-based therapeutic drugs to benefit patients with different types of cancers.
Collapse
Affiliation(s)
- Jin Tian
- Institute of Clinical Medicine, Qingdao University Medical College, Qingdao, China.,Department of Oncology, Qingdao Municipal Hospital, Qingdao, China
| | - Yang Liu
- Department of Oncology, Qingdao Municipal Hospital, Qingdao, China
| | - TengLong Zhang
- Department of Oncology, Qingdao Municipal Hospital, Qingdao, China
| | - Lu Yue
- Department of Oncology, Qingdao Municipal Hospital, Qingdao, China
| | - YaNan Xiao
- Department of Endocrinology, Qingdao Municipal Hospital West Campus, Qingdao, China
| | - ChengYe Guo
- Department of Oncology, Qingdao Municipal Hospital, Qingdao, China
| |
Collapse
|
132
|
Sugiura D, Okazaki IM, Maeda TK, Maruhashi T, Shimizu K, Arakaki R, Takemoto T, Ishimaru N, Okazaki T. PD-1 agonism by anti-CD80 inhibits T cell activation and alleviates autoimmunity. Nat Immunol 2022; 23:399-410. [PMID: 35145298 DOI: 10.1038/s41590-021-01125-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 12/20/2021] [Indexed: 12/11/2022]
Abstract
Targeted blockade of the checkpoint molecule programmed cell death 1 (PD-1) can activate tumor-specific T cells to destroy tumors, whereas targeted potentiation of PD-1 is expected to suppress autoreactive T cells and alleviate autoimmune diseases. However, the development of methods to potentiate PD-1 remains challenging. Here we succeeded in eliciting PD-1 function by targeting the cis-PD-L1-CD80 duplex, formed by binding of CD80 to the PD-1 ligand PD-L1, that attenuates PD-L1-PD-1 binding and abrogates PD-1 function. By generating anti-CD80 antibodies that detach CD80 from the cis-PD-L1-CD80 duplex and enable PD-L1 to engage PD-1 in the presence of CD80, we demonstrate that the targeted dissociation of cis-PD-L1-CD80 duplex elicits PD-1 function in the condition where PD-1 function is otherwise restricted. We demonstrate using murine models that the removal of PD-1 restriction is effective in alleviating autoimmune disease symptoms. Our findings establish a method to potentiate PD-1 function and propose the removal of restraining mechanisms as an efficient strategy to potentiate the function of inhibitory molecules.
Collapse
Affiliation(s)
- Daisuke Sugiura
- Laboratory of Molecular Immunology, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan.,Laboratory for Immune Regulation, Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan
| | - Il-Mi Okazaki
- Laboratory of Molecular Immunology, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan.,Laboratory for Immune Regulation, Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan
| | - Takeo K Maeda
- Laboratory for Immune Regulation, Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan
| | - Takumi Maruhashi
- Laboratory of Molecular Immunology, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan.,Laboratory for Immune Regulation, Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan
| | - Kenji Shimizu
- Laboratory of Molecular Immunology, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan.,Laboratory for Immune Regulation, Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan
| | - Rieko Arakaki
- Department of Oral Molecular Pathology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Tatsuya Takemoto
- Laboratory for Embryology, Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan
| | - Naozumi Ishimaru
- Department of Oral Molecular Pathology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Taku Okazaki
- Laboratory of Molecular Immunology, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan. .,Laboratory for Immune Regulation, Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan.
| |
Collapse
|
133
|
Carloni R, Rizzo A, Ricci AD, Frega G, Federico AD, Palloni A, Marco MD, Gadaleta-Caldarola G, Brandi G. Dual immune checkpoint blockade in hepatocellular carcinoma: where do we stand? Future Oncol 2022; 18:1023-1034. [PMID: 35109664 DOI: 10.2217/fon-2021-0905] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Hepatocellular carcinoma (HCC) represents the fourth most common cause of cancer-related death. Surgery, local ablative therapies and liver transplantation are the only potentially curative strategies, but the majority of patients present with advanced disease at diagnosis or develop recurrence after surgery. In recent years, immunotherapy for HCC has received growing interest, and one of the most promising strategies is the association of two immune checkpoint inhibitors (ICIs), which has already demonstrated its potential in other solid tumors such as melanoma and renal cell carcinoma. Herein, we discuss the role and the biologic rationale of dual immune checkpoint blockade in HCC patients, focusing on the two ICI combinations: nivolumab plus ipilimumab and durvalumab plus tremelimumab.
Collapse
Affiliation(s)
- Riccardo Carloni
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni 15, Bologna, Italy
| | - Alessandro Rizzo
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni 15, Bologna, Italy.,Medical Oncology Unit, 'Mons. R. Dimiccoli' Hospital, Barletta (BT), Azienda Sanitaria Locale Barletta, 76121, Italy
| | - Angela Dalia Ricci
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni 15, Bologna, Italy.,Medical Oncology Unit, 'Mons. R. Dimiccoli' Hospital, Barletta (BT), Azienda Sanitaria Locale Barletta, 76121, Italy
| | - Giorgio Frega
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni 15, Bologna, Italy
| | - Alessandro Di Federico
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni 15, Bologna, Italy
| | - Andrea Palloni
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni 15, Bologna, Italy
| | - Mariacristina Di Marco
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni 15, Bologna, Italy
| | - Gennaro Gadaleta-Caldarola
- Medical Oncology Unit, 'Mons. R. Dimiccoli' Hospital, Barletta (BT), Azienda Sanitaria Locale Barletta, 76121, Italy
| | - Giovanni Brandi
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni 15, Bologna, Italy
| |
Collapse
|
134
|
Meier JA, Savoldo B, Grover NS. The Emerging Role of CAR T Cell Therapy in Relapsed/Refractory Hodgkin Lymphoma. J Pers Med 2022; 12:197. [PMID: 35207685 PMCID: PMC8877886 DOI: 10.3390/jpm12020197] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 01/27/2023] Open
Abstract
Treatment for Hodgkin lymphoma (HL) has evolved considerably from the time it was originally described in the 19th century with many patients now being cured with frontline therapy. Despite these advances, upwards of 10% of patients experience progressive disease after initial therapy with an even higher percentage relapsing. Until recently there had been limited therapeutic options for relapsed and/or refractory HL outside of highly intensive chemotherapy with stem cell rescue. Improved understanding of the pathophysiology of HL, coupled with the emergence of more targeted therapeutics, has reshaped how we view the treatment of relapsed/refractory HL and its prognosis. With this, there has been an increased focus on immunotherapies that can reprogram the immune system to better overcome the immunosuppressive milieu found in HL for improved cancer cell killing. In particular, chimeric antigen receptor (CAR) T cells are emerging as a valuable therapeutic tool in this area. Building on the success of antibody-drug conjugates directed against CD30, CAR T cells engineered to recognize the same antigen are now reaching patients. Though still in its infancy, CAR T therapy for relapsed/refractory HL has shown exceptional promise in early-stage clinical trials with the potential for durable responses even in patients who had progressed through multiple lines of prior therapy. Here we will review currently available data on the use of CAR T cells in HL, strategies to optimize their effectiveness, and how this therapy may fit into the treatment paradigm of HL going forward.
Collapse
Affiliation(s)
- Jeremy A. Meier
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (J.A.M.); (B.S.)
- Department of Medicine, Division of Hematology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Barbara Savoldo
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (J.A.M.); (B.S.)
- Department of Pediatrics, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Natalie S. Grover
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (J.A.M.); (B.S.)
- Department of Medicine, Division of Hematology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
135
|
Ye Y, Zhang Y, Yang N, Gao Q, Ding X, Kuang X, Bao R, Zhang Z, Sun C, Zhou B, Wang L, Hu Q, Lin C, Gao J, Lou Y, Lin SH, Diao L, Liu H, Chen X, Mills GB, Han L. Profiling of immune features to predict immunotherapy efficacy. Innovation (N Y) 2022; 3:100194. [PMID: 34977836 PMCID: PMC8688727 DOI: 10.1016/j.xinn.2021.100194] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/30/2021] [Indexed: 12/14/2022] Open
Abstract
Immune checkpoint blockade (ICB) therapies exhibit substantial clinical benefit in different cancers, but relatively low response rates in the majority of patients highlight the need to understand mutual relationships among immune features. Here, we reveal overall positive correlations among immune checkpoints and immune cell populations. Clinically, patients benefiting from ICB exhibited increases for both immune stimulatory and inhibitory features after initiation of therapy, suggesting that the activation of the immune microenvironment might serve as the biomarker to predict immune response. As proof-of-concept, we demonstrated that the immune activation score (IS Δ) based on dynamic alteration of interleukins in patient plasma as early as two cycles (4-6 weeks) after starting immunotherapy can accurately predict immunotherapy efficacy. Our results reveal a systematic landscape of associations among immune features and provide a noninvasive, cost-effective, and time-efficient approach based on dynamic profiling of pre- and on-treatment plasma to predict immunotherapy efficacy.
Collapse
Affiliation(s)
- Youqiong Ye
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX 77030, USA
| | - Yongchang Zhang
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410008, China
| | - Nong Yang
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410008, China
| | - Qian Gao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Xinyu Ding
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xinwei Kuang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Rujuan Bao
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhao Zhang
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX 77030, USA
| | - Chaoyang Sun
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Bingying Zhou
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Li Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Qingsong Hu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chunru Lin
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jianjun Gao
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yanyan Lou
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Steven H. Lin
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lixia Diao
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hong Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Gordon B. Mills
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Leng Han
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX 77030, USA
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030, USA
- Department of Translational Medical Sciences, College of Medicine, Texas A&M University, Houston, TX 77030 USA
| |
Collapse
|
136
|
Burnell SEA, Capitani L, MacLachlan BJ, Mason GH, Gallimore AM, Godkin A. Seven mysteries of LAG-3: a multi-faceted immune receptor of increasing complexity. IMMUNOTHERAPY ADVANCES 2021; 2:ltab025. [PMID: 35265944 PMCID: PMC8895726 DOI: 10.1093/immadv/ltab025] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 12/17/2021] [Indexed: 12/17/2022] Open
Abstract
Despite three decades of research to its name and increasing interest in immunotherapies that target it, LAG-3 remains an elusive co-inhibitory receptor in comparison to the well-established PD-1 and CTLA-4. As such, LAG-3 targeting therapies have yet to achieve the clinical success of therapies targeting other checkpoints. This could, in part, be attributed to the many unanswered questions that remain regarding LAG-3 biology. Of these, we address: (i) the function of the many LAG-3-ligand interactions, (ii) the hurdles that remain to acquire a high-resolution structure of LAG-3, (iii) the under-studied LAG-3 signal transduction mechanism, (iv) the elusive soluble form of LAG-3, (v) the implications of the lack of (significant) phenotype of LAG-3 knockout mice, (vi) the reports of LAG-3 expression on the epithelium, and (vii) the conflicting reports of LAG-3 expression (and potential contributions to pathology) in the brain. These mysteries which surround LAG-3 highlight how the ever-evolving study of its biology continues to reveal ever-increasing complexity in its role as an immune receptor. Importantly, answering the questions which shroud LAG-3 in mystery will allow the maximum therapeutic benefit of LAG-3 targeting immunotherapies in cancer, autoimmunity and beyond.
Collapse
Affiliation(s)
- Stephanie E A Burnell
- Division of Infection and Immunity, Henry Wellcome Building, Cardiff University, Cardiff, UK
| | - Lorenzo Capitani
- Division of Infection and Immunity, Henry Wellcome Building, Cardiff University, Cardiff, UK
| | - Bruce J MacLachlan
- Division of Infection and Immunity, Henry Wellcome Building, Cardiff University, Cardiff, UK
| | - Georgina H Mason
- Division of Infection and Immunity, Henry Wellcome Building, Cardiff University, Cardiff, UK
| | - Awen M Gallimore
- Division of Infection and Immunity, Henry Wellcome Building, Cardiff University, Cardiff, UK
| | - Andrew Godkin
- Division of Infection and Immunity, Henry Wellcome Building, Cardiff University, Cardiff, UK
- Department of Gastroenterology and Hepatology, University Hospital of Wales, Heath Park, Cardiff, UK
| |
Collapse
|
137
|
Immunosuppression and outcomes in adult patients with de novo acute myeloid leukemia with normal karyotypes. Proc Natl Acad Sci U S A 2021; 118:2116427118. [PMID: 34845035 PMCID: PMC8673586 DOI: 10.1073/pnas.2116427118] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2021] [Indexed: 11/18/2022] Open
Abstract
Acute myeloid leukemia (AML) patients rarely have long first remissions (LFRs; >5 y) after standard-of-care chemotherapy, unless classified as favorable risk at presentation. Identification of the mechanisms responsible for long vs. more typical, standard remissions may help to define prognostic determinants for chemotherapy responses. Using exome sequencing, RNA-sequencing, and functional immunologic studies, we characterized 28 normal karyotype (NK)-AML patients with >5 y first remissions after chemotherapy (LFRs) and compared them to a well-matched group of 31 NK-AML patients who relapsed within 2 y (standard first remissions [SFRs]). Our combined analyses indicated that genetic-risk profiling at presentation (as defined by European LeukemiaNet [ELN] 2017 criteria) was not sufficient to explain the outcomes of many SFR cases. Single-cell RNA-sequencing studies of 15 AML samples showed that SFR AML cells differentially expressed many genes associated with immune suppression. The bone marrow of SFR cases had significantly fewer CD4+ Th1 cells; these T cells expressed an exhaustion signature and were resistant to activation by T cell receptor stimulation in the presence of autologous AML cells. T cell activation could be restored by removing the AML cells or blocking the inhibitory major histocompatibility complex class II receptor, LAG3. Most LFR cases did not display these features, suggesting that their AML cells were not as immunosuppressive. These findings were confirmed and extended in an independent set of 50 AML cases representing all ELN 2017 risk groups. AML cell-mediated suppression of CD4+ T cell activation at presentation is strongly associated with unfavorable outcomes in AML patients treated with standard chemotherapy.
Collapse
|
138
|
Multifaceted Roles of Chemokines and Chemokine Receptors in Tumor Immunity. Cancers (Basel) 2021; 13:cancers13236132. [PMID: 34885241 PMCID: PMC8656932 DOI: 10.3390/cancers13236132] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/26/2021] [Accepted: 12/02/2021] [Indexed: 12/22/2022] Open
Abstract
Simple Summary Various immune cells are involved in host immune responses to cancer. T-helper (Th) 1 cells, cytotoxic CD8+ T cells, and natural killer cells are the major effector cells in anti-tumor immunity, whereas cells such as regulatory T cells and myeloid-derived suppressor cells are negatively involved in anti-tumor immunity. Th2 cells and Th17 cells have been shown to have both pro-tumor and anti-tumor activities. The migratory properties of various immune cells are essential for their function and critically regulated by the chemokine superfamily. In this review, we summarize the roles of various immune cells in tumor immunity and their migratory regulation by the chemokine superfamily. We also assess the therapeutic possibilities of targeting chemokines and chemokine receptors in cancer immunotherapy. Abstract Various immune cells are involved in host tumor immune responses. In particular, there are many T cell subsets with different roles in tumor immunity. T-helper (Th) 1 cells are involved in cellular immunity and thus play the major role in host anti-tumor immunity by inducing and activating cytotoxic T lymphocytes (CTLs). On the other hand, Th2 cells are involved in humoral immunity and suppressive to Th1 responses. Regulatory T (Treg) cells negatively regulate immune responses and contribute to immune evasion of tumor cells. Th17 cells are involved in inflammatory responses and may play a role in tumor progression. However, recent studies have also shown that Th17 cells are capable of directly inducting CTLs and thus may promote anti-tumor immunity. Besides these T cell subsets, there are many other innate immune cells such as dendritic cells (DCs), natural killer (NK) cells, and myeloid-derived suppressor cells (MDSCs) that are involved in host immune responses to cancer. The migratory properties of various immune cells are critical for their functions and largely regulated by the chemokine superfamily. Thus, chemokines and chemokine receptors play vital roles in the orchestration of host immune responses to cancer. In this review, we overview the various immune cells involved in host responses to cancer and their migratory properties regulated by the chemokine superfamily. Understanding the roles of chemokines and chemokine receptors in host immune responses to cancer may provide new therapeutic opportunities for cancer immunotherapy.
Collapse
|
139
|
Elliot TAE, Jennings EK, Lecky DAJ, Thawait N, Flores-Langarica A, Copland A, Maslowski KM, Wraith DC, Bending D. Antigen and checkpoint receptor engagement recalibrates T cell receptor signal strength. Immunity 2021; 54:2481-2496.e6. [PMID: 34534438 PMCID: PMC8585507 DOI: 10.1016/j.immuni.2021.08.020] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 06/21/2021] [Accepted: 08/17/2021] [Indexed: 12/17/2022]
Abstract
How T cell receptor (TCR) signal strength modulates T cell function and to what extent this is modified by immune checkpoint blockade (ICB) are key questions in immunology. Using Nr4a3-Tocky mice, we characterized early quantitative and qualitative changes that occur in CD4+ T cells in relation to TCR signaling strength. We captured how dose- and time-dependent programming of distinct co-inhibitory receptors rapidly recalibrates T cell activation thresholds and visualized the immediate effects of ICB on T cell re-activation. Our findings reveal that anti-PD1 immunotherapy leads to an increased TCR signal strength. We defined a strong TCR signal metric of five genes upregulated by anti-PD1 in T cells (TCR.strong), which was superior to a canonical T cell activation gene signature in stratifying melanoma patient outcomes to anti-PD1 therapy. Our study therefore reveals how analysis of TCR signal strength-and its manipulation-can provide powerful metrics for monitoring outcomes to immunotherapy.
Collapse
Affiliation(s)
- Thomas A E Elliot
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Emma K Jennings
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - David A J Lecky
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Natasha Thawait
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Adriana Flores-Langarica
- Infrastructure and Facilities, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Alastair Copland
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Kendle M Maslowski
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - David C Wraith
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - David Bending
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK.
| |
Collapse
|
140
|
Garcia Cruz D, Giri RR, Gamiotea Turro D, Balsbaugh JL, Adler AJ, Rodriguez A. Lymphocyte Activation Gene-3 Regulates Dendritic Cell Metabolic Programing and T Cell Priming Function. THE JOURNAL OF IMMUNOLOGY 2021; 207:2374-2384. [PMID: 34588222 DOI: 10.4049/jimmunol.2001188] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 09/01/2021] [Indexed: 12/15/2022]
Abstract
Deficiency of lymphocyte activation gene-3 (LAG3) is significantly associated with increased cardiovascular disease risk with in vitro results demonstrating increased TNF-α and decreased IL-10 secretion from LAG3-deficient human B lymphoblasts. The hypothesis tested in this study was that Lag3 deficiency in dendritic cells (DCs) would significantly affect cytokine expression, alter cellular metabolism, and prime naive T cells to greater effector differentiation. Experimental approaches used included differentiation of murine bone marrow-derived DCs (BMDCs) to measure secreted cytokines, cellular metabolism, RNA sequencing, whole cell proteomics, adoptive OT-II CD4+Lag3 +/+ donor cells into wild-type (WT) C57BL/6 and Lag3 -/- recipient mice, and ex vivo measurements of IFN-γ from cultured splenocytes. Results showed that Lag3 -/- BMDCs secreted more TNF-α, were more glycolytic, used fewer fatty acids for mitochondrial respiration, and glycolysis was significantly reduced by exogenous IL-10 treatment. Under basal conditions, RNA sequencing revealed increased expression of CD40 and CD86 and other cytokine-signaling targets as compared with WT. Whole cell proteomics identified a significant number of proteins up- and downregulated in Lag3 -/- BMDCs, with significant differences noted in exogenous IL-10 responsiveness compared with WT cells. Ex vivo, IFN-γ expression was significantly higher in Lag3 -/- mice as compared with WT. With in vivo adoptive T cell and in vitro BMDC:T coculture experiments, Lag3 -/- BMDCs showed greater T cell effector differentiation and proliferation, respectively, compared with WT BMDCs. In conclusion, Lag3 deficiency in DCs is associated with an inflammatory phenotype that provides a plausible mechanism for increased cardiovascular disease risk in humans with LAG3 deficiency.
Collapse
Affiliation(s)
| | | | | | - Jeremy L Balsbaugh
- Center for Open Research Resources and Equipment, University of Connecticut, Storrs, CT; and
| | - Adam J Adler
- Department of Immunology, UConn Health, Farmington, CT
| | | |
Collapse
|
141
|
Gertel S, Polachek A, Furer V, Levartovsky D, Elkayam O. CD4 + LAG-3 + T cells are decreased in active psoriatic arthritis patients and their restoration in vitro is mediated by TNF inhibitors. Clin Exp Immunol 2021; 206:173-183. [PMID: 34309834 PMCID: PMC8506121 DOI: 10.1111/cei.13646] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 07/04/2021] [Accepted: 07/19/2021] [Indexed: 12/25/2022] Open
Abstract
Psoriatic arthritis (PsA) is a chronic inflammatory disease associated with T cell dysregulation. The lymphocyte-activation gene (LAG)-3 is one of the regulatory receptors expressed on T cells in a soluble form. LAG-3 expression on T cells was analyzed in vitro in PsA patients with minimal disease activity (MDA), active disease (non-MDA) and healthy controls. In cultured in-vitro peripheral blood mononuclear cells (PBMCs), LAG-3 expression on CD4+ T cells was similar in both MDA PsA patients (7.5 ± 0.9) (n = 14) and healthy controls (7.8 ± 0.6) (n = 15), but significantly lower in non-MDA PsA patients (3.1 ± 0.3) (n = 13) (p < 0.0001). An inverse correlation between PsA clinical disease activity and %CD4+ LAG-3+ T cells in vitro was observed (composite psoriatic disease activity index r = -0.47, p < 0.02 and psoriatic arthritis disease activity score, r = -0.51, p < 0.008). In-vitro co-culture of CD4+ T cells with anti-tumor necrosis factor (TNF) or anti-interleukin (IL)-17A had no effect on LAG-3+ expression in MDA PsA patients and healthy controls. In non-MDA patients, anti-TNF, but not anti-IL-17A, restored the %CD4+ LAG-3+ T cells (7.9 ± 0.9 and 3.2 ± 0.4, respectively) (p < 0.0004). Lower soluble LAG-3 levels were found in sera of naive to biological PsA patients (n = 39) compared to healthy controls (n = 35) (p < 0.03). Impaired LAG-3 on CD4+ T cells may reflect active PsA disease state. Anti-TNFs have potency to up-regulate the CD4+ LAG-3+ T cells in vitro.
Collapse
Affiliation(s)
- Smadar Gertel
- Department of RheumatologyTel Aviv Sourasky Medical CenterTel AvivIsrael
- Sackler Faculty of MedicineTel Aviv UniversityTel AvivIsrael
| | - Ari Polachek
- Department of RheumatologyTel Aviv Sourasky Medical CenterTel AvivIsrael
- Sackler Faculty of MedicineTel Aviv UniversityTel AvivIsrael
| | - Victoria Furer
- Department of RheumatologyTel Aviv Sourasky Medical CenterTel AvivIsrael
- Sackler Faculty of MedicineTel Aviv UniversityTel AvivIsrael
| | - David Levartovsky
- Department of RheumatologyTel Aviv Sourasky Medical CenterTel AvivIsrael
- Sackler Faculty of MedicineTel Aviv UniversityTel AvivIsrael
| | - Ori Elkayam
- Department of RheumatologyTel Aviv Sourasky Medical CenterTel AvivIsrael
- Sackler Faculty of MedicineTel Aviv UniversityTel AvivIsrael
| |
Collapse
|
142
|
Ma LR, Li JX, Tang L, Li RZ, Yang JS, Sun A, Leung ELH, Yan PY. Immune checkpoints and immunotherapy in non-small cell lung cancer: Novel study progression, challenges and solutions. Oncol Lett 2021; 22:787. [PMID: 34594428 PMCID: PMC8456509 DOI: 10.3892/ol.2021.13048] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 06/04/2021] [Indexed: 12/15/2022] Open
Abstract
Lung cancer is the most common type of cancer with the highest mortality rate worldwide. Non-small cell lung cancer (NSCLC) accounts for ~85% of the total number of lung cancer cases. In the past two decades, immunotherapy has become a more promising treatment method than traditional treatments (surgery, radiotherapy and chemotherapy). Immunotherapy has been shown to improve the survival rate of patients and to have a superior effect when controlling lung cancer than traditional therapy. However, only a small number of patients can benefit from immunotherapy, and not all patients who qualify experience long-term benefits. In the clinic, the objective response rate of programmed cell death protein 1 treatment without the prior screening of patients is only 15-20%. Immunotherapy is associated with both opportunities and challenges for patients with NSCLC. The current challenges of immunotherapy include the lack of accurate biomarkers, inevitable resistance and insufficient understanding of immune checkpoints. In previous years, several methods for overcoming the challenges posed by immunotherapy have been proposed, but combination therapy is the most suitable choice. A large number of studies have shown that the combination of drugs can significantly improve their efficacy, compared with monotherapy, and that some therapeutic combinations have been approved by the Food and Drug Administration for the treatment of NSCLC. Traditional Chinese medicine (TCM) is a traditional medical practice in China that can play an important role in immunotherapy. Most agents used in TCM originate from plants, and have the advantages of low toxicity and multiple targets. In addition, TCM includes a unique class of drugs that can improve autoimmunity. Therefore, TCM may be a promising treatment method for all types of cancer.
Collapse
Affiliation(s)
- Lin-Rui Ma
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 999078, P.R. China
| | - Jia-Xin Li
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 999078, P.R. China
| | - Ling Tang
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China.,Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China.,School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Run-Ze Li
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 999078, P.R. China
| | - Jia-Shun Yang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510630, P.R. China
| | - Ao Sun
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 999078, P.R. China
| | - Elaine Lai-Han Leung
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 999078, P.R. China.,Department of Integrated Chinese and Western Medicine, Zhuhai Hospital of Integrated Traditional Chinese and Western Medicine, Zhuhai 519000, P.R. China
| | - Pei-Yu Yan
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 999078, P.R. China
| |
Collapse
|
143
|
Souri Z, Wierenga APA, Kroes WGM, van der Velden PA, Verdijk RM, Eikmans M, Luyten GPM, Jager MJ. LAG3 and Its Ligands Show Increased Expression in High-Risk Uveal Melanoma. Cancers (Basel) 2021; 13:cancers13174445. [PMID: 34503258 PMCID: PMC8430821 DOI: 10.3390/cancers13174445] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/20/2021] [Accepted: 08/31/2021] [Indexed: 01/10/2023] Open
Abstract
Uveal melanoma (UM) is a rare ocular malignancy which originates in the uveal tract, and often gives rise to metastases. Potential targets for immune checkpoint inhibition are lymphocyte-activation gene 3 (LAG3) and its ligands. We set out to analyse the distribution of these molecules in UM. The expression of mRNA was determined using an Illumina array in 64 primary UM from Leiden. The T lymphocyte fraction was determined by digital droplet PCR. In a second cohort of 15 cases from Leiden, mRNA expression was studied by Fluidigm qPCR, while a third cohort consisted of 80 UM from TCGA. In the first Leiden cohort, LAG3 expression was associated with the presence of epithelioid cells (p = 0.002), monosomy of chromosome 3 (p = 0.004), and loss of BAP1 staining (p = 0.001). In this Leiden cohort as well as in the TCGA cohort, LAG3 expression correlated positively with the expression of its ligands: LSECtin, Galectin-3, and the HLA class II molecules HLA-DR, HLA-DQ, and HLA-DP (all p < 0.001). Furthermore, ligands Galectin-3 and HLA class II were increased in monosomy 3 tumours and the expression of LAG3 correlated with the presence of an inflammatory phenotype (T cell fraction, macrophages, HLA-A and HLA-B expression: all p < 0.001). High expression levels of LAG3 (p = 0.01), Galectin-3 (p = 0.001), HLA-DRA1 (p = 0.002), HLA-DQA1 (p = 0.04), HLA-DQB2 (p = 0.03), and HLA-DPA1 (p = 0.007) were associated with bad survival. We conclude that expression of the LAG ligands Galectin-3 and HLA class II strongly correlates with LAG3 expression and all are increased in UM with Monosomy 3/BAP1 loss. The distribution suggests a potential benefit of monoclonal antibodies against LAG3 or Galectin-3 as adjuvant treatment in patients with high-risk UM.
Collapse
Affiliation(s)
- Zahra Souri
- Department of Ophthalmology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (Z.S.); (A.P.A.W.); (P.A.v.d.V.); (G.P.M.L.)
| | - Annemijn P. A. Wierenga
- Department of Ophthalmology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (Z.S.); (A.P.A.W.); (P.A.v.d.V.); (G.P.M.L.)
| | - Wilma G. M. Kroes
- Department of Clinical Genetics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
| | - Pieter A. van der Velden
- Department of Ophthalmology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (Z.S.); (A.P.A.W.); (P.A.v.d.V.); (G.P.M.L.)
| | - Robert M. Verdijk
- Department of Pathology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands;
- Department of Pathology, Section Ophthalmic Pathology, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Michael Eikmans
- Department of Immunology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands;
| | - Gregorius P. M. Luyten
- Department of Ophthalmology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (Z.S.); (A.P.A.W.); (P.A.v.d.V.); (G.P.M.L.)
| | - Martine J. Jager
- Department of Ophthalmology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (Z.S.); (A.P.A.W.); (P.A.v.d.V.); (G.P.M.L.)
- Correspondence:
| |
Collapse
|
144
|
Moon J, Oh YM, Ha SJ. Perspectives on immune checkpoint ligands: expression, regulation, and clinical implications. BMB Rep 2021. [PMID: 34078531 PMCID: PMC8411045 DOI: 10.5483/bmbrep.2021.54.8.054] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In the tumor microenvironment, immune checkpoint ligands (ICLs) must be expressed in order to trigger the inhibitory signal via immune checkpoint receptors (ICRs). Although ICL expression frequently occurs in a manner intrinsic to tumor cells, extrinsic factors derived from the tumor microenvironment can fine-tune ICL expression by tumor cells or prompt non-tumor cells, including immune cells. Considering the extensive interaction between T cells and other immune cells within the tumor microenvironment, ICL expression on immune cells can be as significant as that of ICLs on tumor cells in promoting anti-tumor immune responses. Here, we introduce various regulators known to induce or suppress ICL expression in either tumor cells or immune cells, and concise mechanisms relevant to their induction. Finally, we focus on the clinical significance of understanding the mechanisms of ICLs for an optimized immunotherapy for individual cancer patients.
Collapse
Affiliation(s)
- Jihyun Moon
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul 03722, Korea
- Brain Korea 21 (BK21) FOUR Program, Yonsei Education & Research Center for Biosystems, Yonsei University, Seoul 03722, Korea
| | - Yoo Min Oh
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul 03722, Korea
- Brain Korea 21 (BK21) FOUR Program, Yonsei Education & Research Center for Biosystems, Yonsei University, Seoul 03722, Korea
| | - Sang-Jun Ha
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul 03722, Korea
- Brain Korea 21 (BK21) FOUR Program, Yonsei Education & Research Center for Biosystems, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
145
|
Abstract
Anti-PD-1 therapies can activate tumor-specific T cells to destroy tumors. However, whether and how T cells with different antigen specificity and affinity are differentially regulated by PD-1 remain vaguely understood. Upon antigen stimulation, a variety of genes is induced in T cells. Recently, we found that T cell receptor (TCR) signal strength required for the induction of genes varies across different genes and PD-1 preferentially inhibits the induction of genes that require stronger TCR signal. As each T cell has its own response characteristics, inducibility of genes likely differs across different T cells. Accordingly, the inhibitory effects of PD-1 are also expected to differ across different T cells. In the current study, we investigated whether and how factors that modulate T cell responsiveness to antigenic stimuli influence PD-1 function. By analyzing TCRs with different affinities to peptide-MHC complexes (pMHC) and pMHCs with different affinities to TCR, we demonstrated that PD-1 inhibits the expression of TCR-inducible genes efficiently when TCR:pMHC affinity is low. In contrast, affinities of peptides to MHC and MHC expression levels did not affect PD-1 sensitivity of TCR-inducible genes although they markedly altered the dose responsiveness of T cells by changing the efficiency of pMHC formation, suggesting that the strength of individual TCR signal is the key determinant of PD-1 sensitivity. Accordingly, we observed a preferential expansion of T cells with low-affinity to tumor-antigen in PD-1-deficient mice upon inoculation of tumor cells. These results demonstrate that PD-1 imposes qualitative control of T cell responses by preferentially suppressing low-affinity T cells.
Collapse
|
146
|
Recent Advances in Glioma Therapy: Combining Vascular Normalization and Immune Checkpoint Blockade. Cancers (Basel) 2021; 13:cancers13153686. [PMID: 34359588 PMCID: PMC8345045 DOI: 10.3390/cancers13153686] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/09/2021] [Accepted: 07/19/2021] [Indexed: 12/28/2022] Open
Abstract
Glioblastoma (GBM) accounts for more than 50% of all primary malignancies of the brain. Current standard treatment regimen for GBM includes maximal surgical resection followed by radiation and adjuvant chemotherapy. However, due to the heterogeneity of the tumor cells, tumor recurrence is often inevitable. The prognosis of patients with glioma is, thus, dismal. Glioma is a highly angiogenic tumor yet immunologically cold. As such, evolving studies have focused on designing strategies that specifically target the tyrosine kinase receptors of angiokines and encourage immune infiltration. Recent promising results from immunotherapies on other cancer types have prompted further investigations of this therapy in GBM. In this article, we reviewed the pathological angiogenesis and immune reactivity in glioma, as well as its target for drug development, and we discussed future directions in glioma therapy.
Collapse
|
147
|
Solé P, Santamaria P. Re-Programming Autoreactive T Cells Into T-Regulatory Type 1 Cells for the Treatment of Autoimmunity. Front Immunol 2021; 12:684240. [PMID: 34335585 PMCID: PMC8320845 DOI: 10.3389/fimmu.2021.684240] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/22/2021] [Indexed: 12/21/2022] Open
Abstract
Systemic delivery of peptide-major histocompatibility complex (pMHC) class II-based nanomedicines can re-program cognate autoantigen-experienced CD4+ T cells into disease-suppressing T-regulatory type 1 (TR1)-like cells. In turn, these TR1-like cells trigger the formation of complex regulatory cell networks that can effectively suppress organ-specific autoimmunity without impairing normal immunity. In this review, we summarize our current understanding of the transcriptional, phenotypic and functional make up of TR1-like cells as described in the literature. The true identity and direct precursors of these cells remain unclear, in particular whether TR1-like cells comprise a single terminally-differentiated lymphocyte population with distinct transcriptional and epigenetic features, or a collection of phenotypically different subsets sharing key regulatory properties. We propose that detailed transcriptional and epigenetic characterization of homogeneous pools of TR1-like cells will unravel this conundrum.
Collapse
Affiliation(s)
- Patricia Solé
- Institut D'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Pere Santamaria
- Institut D'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain.,Julia McFarlane Diabetes Research Centre (JMDRC) and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
148
|
Mei Y, Bi WL, Agolia J, Hu C, Giantini Larsen AM, Meredith DM, Al Abdulmohsen S, Bale T, Dunn GP, Abedalthagafi M, Dunn IF. Immune profiling of pituitary tumors reveals variations in immune infiltration and checkpoint molecule expression. Pituitary 2021; 24:359-373. [PMID: 33492612 DOI: 10.1007/s11102-020-01114-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/27/2020] [Indexed: 12/30/2022]
Abstract
PURPOSE Pituitary tumors are the second most common primary brain tumors. Functional tumors demonstrate increased PD-L1 expression, but expression of other checkpoint regulators has not been characterized. We sought to characterize the immune microenvironment of human pituitary tumors to identify new treatment opportunities. METHODS 72 pituitary tumors were evaluated for expression of the immune regulatory markers programmed death ligand 1 (PD-L1), programmed death ligand 2 (PD-L2), V-domain Ig suppressor of T cell activation (VISTA), lymphocyte activation gene 3 (LAG3) and tumor necrosis factor receptor superfamily member 4 (OX40) by immunohistochemistry (IHC). Lymphocyte infiltration, macrophage infiltration, and angiogenesis were analyzed using IHC. Expression of pituitary tumor initiating cell marker CD15 and mismatch repair proteins MutS protein homolog 2 (MSH2) and MutS protein homolog 6 (MSH6) was also assessed. RESULTS Pituitary tumors were infiltrated by macrophages and T cells, and they expressed varying levels of PD-L1, PD-L2, VISTA, LAG3, and OX40. Functional tumors and tumors with high expression of tumor stem cell markers had higher immune cell infiltration and greater expression of immunosuppressive checkpoint regulators. Increased PD-L1 and LAG3 and reduced VISTA were observed in primary tumors compared to recurrent tumors. CONCLUSION Immune cell infiltration and checkpoint regulator expression vary depending on functional status and presence of pituitary tumor initiating cells. Functional tumors may have a particularly immunosuppressive microenvironment. Further studies of immune checkpoint blockade of pituitary tumors, particularly functional tumors, are warranted, though combination therapy may be required.
Collapse
Affiliation(s)
- Yu Mei
- Center for Skull Base and Pituitary Surgery, Department of Neurosurgery, Harvard Medical School, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Wenya Linda Bi
- Center for Skull Base and Pituitary Surgery, Department of Neurosurgery, Harvard Medical School, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA, 02115, USA.
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
| | - James Agolia
- Center for Skull Base and Pituitary Surgery, Department of Neurosurgery, Harvard Medical School, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Changchen Hu
- Center for Skull Base and Pituitary Surgery, Department of Neurosurgery, Harvard Medical School, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA, 02115, USA
- Department of Neurosurgery, Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, China
| | | | - David M Meredith
- Department of Pathology, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Sally Al Abdulmohsen
- Center for Skull Base and Pituitary Surgery, Department of Neurosurgery, Harvard Medical School, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA, 02115, USA
- King Fahad Medical City and King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Tejus Bale
- Department of Neuropathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gavin P Dunn
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, MO, USA
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA
| | - Malak Abedalthagafi
- King Fahad Medical City and King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Ian F Dunn
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, HHDC Suite 4000, 1000 N. Lincoln Blvd, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
149
|
The critical role of CD4+ T cells in PD-1 blockade against MHC-II-expressing tumors such as classic Hodgkin lymphoma. Blood Adv 2021; 4:4069-4082. [PMID: 32870971 DOI: 10.1182/bloodadvances.2020002098] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 08/02/2020] [Indexed: 12/18/2022] Open
Abstract
Classic Hodgkin lymphoma (cHL) responds markedly to PD-1 blockade therapy, and the clinical responses are reportedly dependent on expression of major histocompatibility complex class II (MHC-II). This dependence is different from other solid tumors, in which the MHC class I (MHC-I)/CD8+ T-cell axis plays a critical role. In this study, we investigated the role of the MHC-II/CD4+ T-cell axis in the antitumor effect of PD-1 blockade on cHL. In cHL, MHC-I expression was frequently lost, but MHC-II expression was maintained. CD4+ T cells highly infiltrated the tumor microenvironment of MHC-II-expressing cHL, regardless of MHC-I expression status. Consequently, CD4+ T-cell, but not CD8+ T-cell, infiltration was a good prognostic factor in cHL, and PD-1 blockade showed antitumor efficacy against MHC-II-expressing cHL associated with CD4+ T-cell infiltration. Murine lymphoma and solid tumor models revealed the critical role of antitumor effects mediated by CD4+ T cells: an anti-PD-1 monoclonal antibody exerted antitumor effects on MHC-I-MHC-II+ tumors but not on MHC-I-MHC-II- tumors, in a cytotoxic CD4+ T-cell-dependent manner. Furthermore, LAG-3, which reportedly binds to MHC-II, was highly expressed by tumor-infiltrating CD4+ T cells in MHC-II-expressing tumors. Therefore, the combination of LAG-3 blockade with PD-1 blockade showed a far stronger antitumor immunity compared with either treatment alone. We propose that PD-1 blockade therapies have antitumor effects on MHC-II-expressing tumors such as cHL that are mediated by cytotoxic CD4+ T cells and that LAG-3 could be a candidate for combination therapy with PD-1 blockade.
Collapse
|
150
|
Abstract
Immunotherapy has become the mainstay for lung cancer treatment, providing sustained therapeutic responses and improved prognosis compared with those obtained with surgery, chemotherapy, radiotherapy, and targeted therapy. It has the potential for anti-tumor treatment and killing tumor cells by activating human immunity and has moved the targets of anti-cancer therapy from malignant tumor cells to immune cell subsets. Two kinds of immune checkpoints, cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and programmed death-1 (PD-1)/programmed death ligand 1 (PD-L1), are the main targets of current immunotherapy in lung cancer. Despite the successful outcomes achieved by immune checkpoint inhibitors, a small portion of lung cancer patients remain unresponsive to checkpoint immunotherapy or may ultimately become resistant to these agents as a result of the complex immune modulatory network in the tumor microenvironment. Therefore, it is imperative to exploit novel immunotherapy targets to further expand the proportion of patients benefiting from immunotherapy. This review summarizes the molecular features, biological function, and clinical significance of several novel checkpoints that have important roles in lung cancer immune responses beyond the CTLA-4 and PD-1/PD-L1 axes, including the markers of co-inhibitory and co-stimulatory T lymphocyte pathways and inhibitory markers of macrophages and natural killer cells.
Collapse
|