1
|
Abdo EL, Ajib I, El Mounzer J, Husseini M, Kalaoun G, Matta TM, Mosleh R, Nasr F, Richani N, Khalil A, Shayya A, Ghanem H, Faour WH. Molecular biology of the novel anticancer medications: a focus on kinases inhibitors, biologics and CAR T-cell therapy. Inflamm Res 2025; 74:41. [PMID: 39960501 DOI: 10.1007/s00011-025-02008-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 01/28/2025] [Accepted: 02/10/2025] [Indexed: 05/09/2025] Open
Abstract
INTRODUCTION Cancer treatment underwent significant changes in the last few years with the introduction of novel treatments targeting the immune system. OBJECTIVES The objective of this review is to discuss novel anticancer drugs including kinase inhibitors, biologics and cellular therapy with CAR-T cells. METHODS Most recent research articles were extracted from PubMed using keywords such as "kinases inhibitors", "CAR-T cell therapy". RESULTS AND DISCUSSION The number of kinase inhibitors is significantly increasing due to their demonstrated effectiveness in combination with biologics. CAR-T represented a major breakthrough in the field. Also, it focused on their mechanisms of action and the rational of their use either alone or in combination in relation to their modes of action.
Collapse
Affiliation(s)
- Elia-Luna Abdo
- Gilbert & Rose-Marie Chagoury School of Medicine, Lebanese American University, Room 4722, P.O. Box 36, Byblos, Lebanon
| | - Imad Ajib
- Gilbert & Rose-Marie Chagoury School of Medicine, Lebanese American University, Room 4722, P.O. Box 36, Byblos, Lebanon
| | - Jason El Mounzer
- Gilbert & Rose-Marie Chagoury School of Medicine, Lebanese American University, Room 4722, P.O. Box 36, Byblos, Lebanon
| | - Mohammad Husseini
- Gilbert & Rose-Marie Chagoury School of Medicine, Lebanese American University, Room 4722, P.O. Box 36, Byblos, Lebanon
| | - Gharam Kalaoun
- Gilbert & Rose-Marie Chagoury School of Medicine, Lebanese American University, Room 4722, P.O. Box 36, Byblos, Lebanon
| | - Tatiana-Maria Matta
- Gilbert & Rose-Marie Chagoury School of Medicine, Lebanese American University, Room 4722, P.O. Box 36, Byblos, Lebanon
| | - Reine Mosleh
- Gilbert & Rose-Marie Chagoury School of Medicine, Lebanese American University, Room 4722, P.O. Box 36, Byblos, Lebanon
| | - Fidel Nasr
- Gilbert & Rose-Marie Chagoury School of Medicine, Lebanese American University, Room 4722, P.O. Box 36, Byblos, Lebanon
| | - Nour Richani
- Gilbert & Rose-Marie Chagoury School of Medicine, Lebanese American University, Room 4722, P.O. Box 36, Byblos, Lebanon
| | - Alia Khalil
- Gilbert & Rose-Marie Chagoury School of Medicine, Lebanese American University, Room 4722, P.O. Box 36, Byblos, Lebanon
| | - Anwar Shayya
- Gilbert & Rose-Marie Chagoury School of Medicine, Lebanese American University, Room 4722, P.O. Box 36, Byblos, Lebanon
- Department of Hematology-Oncology, Lebanese American University Medical Center- Rizk Hospital, Beirut, Lebanon
| | - Hady Ghanem
- Gilbert & Rose-Marie Chagoury School of Medicine, Lebanese American University, Room 4722, P.O. Box 36, Byblos, Lebanon
- Department of Hematology-Oncology, Lebanese American University Medical Center- Rizk Hospital, Beirut, Lebanon
| | - Wissam H Faour
- Gilbert & Rose-Marie Chagoury School of Medicine, Lebanese American University, Room 4722, P.O. Box 36, Byblos, Lebanon.
| |
Collapse
|
2
|
Zimmerer JM, Chaudhari S, Koneru K, Han JL, Abdel-Rasoul M, Uwase H, Yi T, Breuer CK, Bumgardner GL. Germinal Center B Cells are Uniquely Targeted by Antibody-Suppressor CXCR5 +CD8 + T Cells. Transplant Direct 2025; 11:e1742. [PMID: 39802197 PMCID: PMC11723704 DOI: 10.1097/txd.0000000000001742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/16/2024] [Accepted: 10/31/2024] [Indexed: 01/16/2025] Open
Abstract
Background Alloprimed antibody-suppressor CXCR5+CD8+ T cells (CD8+ TAb-supp cells) downregulate alloantibody production, mediate cytotoxicity of IgG+ B cells, and prolong allograft survival. The purpose of this investigation was to determine which immune-cell subsets are susceptible to CD8+ TAb-supp cell-mediated cytotoxicity or noncytotoxic suppression. Methods Alloprimed immune-cell subsets were evaluated for susceptibility to CD8+ TAb-supp cell-mediated in vitro cytotoxicity and/or suppression of intracellular cytokine expression. In vivo CD8-mediated cytotoxicity to wild-type germinal center (GC) B cells or wild-type CD4+ T follicular helper cells (TFH cells) was assessed in RAG1 knockout mice. The impact of in vivo adoptive transfer of CD8+ TAb-supp cells into hepatocyte or kidney transplant recipients on the quantity of lymphoid immune-cell subsets was assessed. Results CD8+ TAb-supp cells mediated allospecific cytotoxicity to alloprimed GC B cells but not alloprimed extrafollicular plasmablasts, marginal zone B cells, follicular B cells, or plasma cells. CD8+ TAb-supp cells did not mediate cytotoxicity to alloprimed dendritic cells, macrophages, CD4+ TFH cells, CD4+ T follicular regulatory cells, or CD4+ regulatory T cell. CD8+ TAb-supp cells did not suppress CD4+ TFH cell, T follicular regulatory cell, or regulatory T-cell cytokine expression. Adoptive transfer of CD8+ TAb-supp cells into hepatocyte or kidney transplant recipients reduced alloantibody production and the quantity of GC B cells, TFH cells, and plasma cells (but not other B-cell, T-cell, or antigen-presenting cell subsets). The reduction of TFH-cell quantity was dependent on CD8+ TAb-supp cell-mediated major histocompatibility complex-I-dependent cytotoxic killing of GC B cells. Conclusions The primary targets of CD8+ TAb-supp cells are GC B cells with downstream reduction of TFH and plasma cells.
Collapse
Affiliation(s)
- Jason M. Zimmerer
- Department of Surgery, Comprehensive Transplant Center, and the College of Medicine, The Ohio State University, Columbus, OH
| | - Sachi Chaudhari
- Department of Surgery, Comprehensive Transplant Center, and the College of Medicine, The Ohio State University, Columbus, OH
| | - Kavya Koneru
- Department of Surgery, Comprehensive Transplant Center, and the College of Medicine, The Ohio State University, Columbus, OH
| | - Jing L. Han
- Department of Surgery, Comprehensive Transplant Center, and the College of Medicine, The Ohio State University, Columbus, OH
- Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, OH
| | - Mahmoud Abdel-Rasoul
- Department of Biomedical Informatics, Center for Biostatistics, The Ohio State University, Columbus, OH
| | - Hope Uwase
- Department of Surgery, Comprehensive Transplant Center, and the College of Medicine, The Ohio State University, Columbus, OH
| | - Tai Yi
- Center for Regenerative Medicine, The Research Institute at Nationwide Children’s Hospital, Columbus, OH
| | - Christopher K. Breuer
- Center for Regenerative Medicine, The Research Institute at Nationwide Children’s Hospital, Columbus, OH
| | - Ginny L. Bumgardner
- Department of Surgery, Comprehensive Transplant Center, and the College of Medicine, The Ohio State University, Columbus, OH
| |
Collapse
|
3
|
Messmer JM, Thommek C, Piechutta M, Venkataramani V, Wehner R, Westphal D, Schubert M, Mayer CD, Effern M, Berghoff AS, Hinze D, Helfrich I, Schadendorf D, Wick W, Hölzel M, Karreman MA, Winkler F. T lymphocyte recruitment to melanoma brain tumors depends on distinct venous vessels. Immunity 2024; 57:2688-2703.e11. [PMID: 39368486 DOI: 10.1016/j.immuni.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/14/2024] [Accepted: 09/06/2024] [Indexed: 10/07/2024]
Abstract
To improve immunotherapy for brain tumors, it is important to determine the principal intracranial site of T cell recruitment from the bloodstream and their intracranial route to brain tumors. Using intravital microscopy in mouse models of intracranial melanoma, we discovered that circulating T cells preferably adhered and extravasated at a distinct type of venous blood vessel in the tumor vicinity, peritumoral venous vessels (PVVs). Other vascular structures were excluded as alternative T cell routes to intracranial melanomas. Anti-PD-1/CTLA-4 immune checkpoint inhibitors increased intracranial T cell motility, facilitating migration from PVVs to the tumor and subsequently inhibiting intracranial tumor growth. The endothelial adhesion molecule ICAM-1 was particularly expressed on PVVs, and, in samples of human brain metastases, ICAM-1 positivity of PVV-like vessels correlated with intratumoral T cell infiltration. These findings uncover a distinct mechanism by which the immune system can access and control brain tumors and potentially influence other brain pathologies.
Collapse
Affiliation(s)
- Julia M Messmer
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany; Institute of Experimental Oncology (IEO), Medical Faculty, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Calvin Thommek
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Manuel Piechutta
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Varun Venkataramani
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, INF 400, 69120 Heidelberg, Germany; Department of Functional Neuroanatomy, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Rebekka Wehner
- Faculty of Medicine Carl Gustav Carus, Institute of Immunology, TU Dresden, 01307 Dresden, Germany; Partner Site Dresden, National Center for Tumor Diseases (NCT), 01307 Dresden, Germany; German Cancer Consortium (DKTK), partner site Dresden, 01307 Dresden, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dana Westphal
- Partner Site Dresden, National Center for Tumor Diseases (NCT), 01307 Dresden, Germany; Department of Dermatology, Medical Faculty and University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Marc Schubert
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, INF 400, 69120 Heidelberg, Germany
| | - Chanté D Mayer
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, INF 400, 69120 Heidelberg, Germany
| | - Maike Effern
- Institute of Experimental Oncology (IEO), Medical Faculty, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Anna S Berghoff
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Vienna, Austria; Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Daniel Hinze
- Institute of Experimental Oncology (IEO), Medical Faculty, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Iris Helfrich
- Medical Faculty of the Ludwig Maximilian University of Munich, Department of Dermatology and Allergology, Frauenlobstrasse 9-11, 80377 Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany; Department of Dermatology, University Hospital Essen, Hufelandstraße 55, 45147 Essen, Germany
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, Hufelandstraße 55, 45147 Essen, Germany
| | - Wolfgang Wick
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, INF 400, 69120 Heidelberg, Germany
| | - Michael Hölzel
- Institute of Experimental Oncology (IEO), Medical Faculty, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Matthia A Karreman
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, INF 400, 69120 Heidelberg, Germany.
| | - Frank Winkler
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, INF 400, 69120 Heidelberg, Germany.
| |
Collapse
|
4
|
Yang X, Li X, Xu H, Du S, Wang C, He H. Predicting CTLA4 expression and prognosis in clear cell renal cell carcinoma using a pathomics signature of histopathological images and machine learning. Heliyon 2024; 10:e34877. [PMID: 39145002 PMCID: PMC11320204 DOI: 10.1016/j.heliyon.2024.e34877] [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] [Received: 08/08/2023] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 08/16/2024] Open
Abstract
Background CTLA4, an immune checkpoint, plays an important role in tumor immunotherapy. The purpose of this study was to develop a pathomics signature to evaluate CTLA4 expression and predict clinical outcomes in clear cell renal cell carcinoma (ccRCC) patients. Methods A total of 354 patients from the TCGA-KIRC dataset were enrolled in this study. The patients were stratified into two groups based on the level of CTLA4 expression, and overall survival rates were analyzed between groups. Pathological features were identified using machine learning algorithms, and a gradient boosting machine (GBM) was employed to construct the pathomics signatures for predicting prognosis and CTLA4 expression. The predictive performance of the model was subsequently assessed. Enrichment analysis was performed on diferentially expressed genes related to the pathomics score (PS). Additionally, correlations between PS and TMB, as well as immune infiltration profiles associated with different PS values, were explored. In vitro experiments, CTLA4 knockdown was performed to investigate its impact on cell proliferation, migration, invasion, TGF-β signaling pathway, and macrophage polarization. Results High expression of CTLA4 was associated with an unfavorable prognosis in ccRCC patients. The pathomics signature displayed good performance in the validation set (AUC = 0.737; P < 0.001 in the log-rank test). The PS was positively correlated with CTLA4 expression. We next explored the underlying mechanism and found the associations between the pathomics signature and TGF-β signaling pathways, TMB, and Tregs. Further in vitro experiments demonstrated that CTLA4 knockdown inhibited cell proliferation, migration, invasion, TGF-β expression, and macrophage M2 polarization. Conclusion High expression of CTLA4 was found to correlate with poor prognosis in ccRCC patients. The pathomics signature established by our group using machine learning effectively predicted both patient prognosis and CTLA4 expression levels in ccRCC cases.
Collapse
Affiliation(s)
- Xiaoqun Yang
- Department of Pathology, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiangyun Li
- Department of Pathology, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Haimin Xu
- Department of Pathology, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Silin Du
- University Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Chaofu Wang
- Department of Pathology, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hongchao He
- Department of Urology, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| |
Collapse
|
5
|
Qiu J, Cheng Z, Jiang Z, Gan L, Zhang Z, Xie Z. Immunomodulatory Precision: A Narrative Review Exploring the Critical Role of Immune Checkpoint Inhibitors in Cancer Treatment. Int J Mol Sci 2024; 25:5490. [PMID: 38791528 PMCID: PMC11122264 DOI: 10.3390/ijms25105490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 05/11/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
An immune checkpoint is a signaling pathway that regulates the recognition of antigens by T-cell receptors (TCRs) during an immune response. These checkpoints play a pivotal role in suppressing excessive immune responses and maintaining immune homeostasis against viral or microbial infections. There are several FDA-approved immune checkpoint inhibitors (ICIs), including ipilimumab, pembrolizumab, and avelumab. These ICIs target cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), programmed cell death protein 1 (PD-1), and programmed death ligand 1 (PD-L1). Furthermore, ongoing efforts are focused on developing new ICIs with emerging potential. In comparison to conventional treatments, ICIs offer the advantages of reduced side effects and durable responses. There is growing interest in the potential of combining different ICIs with chemotherapy, radiation therapy, or targeted therapies. This article comprehensively reviews the classification, mechanism of action, application, and combination strategies of ICIs in various cancers and discusses their current limitations. Our objective is to contribute to the future development of more effective anticancer drugs targeting immune checkpoints.
Collapse
Affiliation(s)
- Junyu Qiu
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (J.Q.); (Z.C.); (Z.J.); (L.G.); (Z.Z.)
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Zilin Cheng
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (J.Q.); (Z.C.); (Z.J.); (L.G.); (Z.Z.)
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Zheng Jiang
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (J.Q.); (Z.C.); (Z.J.); (L.G.); (Z.Z.)
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Luhan Gan
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (J.Q.); (Z.C.); (Z.J.); (L.G.); (Z.Z.)
- Huan Kui School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Zixuan Zhang
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (J.Q.); (Z.C.); (Z.J.); (L.G.); (Z.Z.)
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Zhenzhen Xie
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (J.Q.); (Z.C.); (Z.J.); (L.G.); (Z.Z.)
| |
Collapse
|
6
|
Ding R, Yu X, Hu Z, Dong Y, Huang H, Zhang Y, Han Q, Ni ZY, Zhao R, Ye Y, Zou Q. Lactate modulates RNA splicing to promote CTLA-4 expression in tumor-infiltrating regulatory T cells. Immunity 2024; 57:528-540.e6. [PMID: 38417442 DOI: 10.1016/j.immuni.2024.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/04/2024] [Accepted: 01/30/2024] [Indexed: 03/01/2024]
Abstract
RNA splicing is involved in cancer initiation and progression, but how it influences host antitumor immunity in the metabolically abnormal tumor microenvironment (TME) remains unclear. Here, we demonstrate that lactate modulates Foxp3-dependent RNA splicing to maintain the phenotypic and functional status of tumor-infiltrating regulatory T (Treg) cells via CTLA-4. RNA splicing in Treg cells was correlated with the Treg cell signatures in the TME. Ubiquitin-specific peptidase 39 (USP39), a component of the RNA splicing machinery, maintained RNA-splicing-mediated CTLA-4 expression to control Treg cell function. Mechanistically, lactate promoted USP39-mediated RNA splicing to facilitate CTLA-4 expression in a Foxp3-dependent manner. Moreover, the efficiency of CTLA-4 RNA splicing was increased in tumor-infiltrating Treg cells from patients with colorectal cancer. These findings highlight the immunological relevance of RNA splicing in Treg cells and provide important insights into the environmental mechanism governing CTLA-4 expression in Treg cells.
Collapse
Affiliation(s)
- Rui Ding
- Hongqiao International Institute of Medicine, Tongren Hospital & Shanghai Institute of Immunology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiaoyan Yu
- Hongqiao International Institute of Medicine, Tongren Hospital & Shanghai Institute of Immunology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhilin Hu
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease, the School of Basic Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Yu Dong
- Hongqiao International Institute of Medicine, Tongren Hospital & Shanghai Institute of Immunology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Haiyan Huang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yuerong Zhang
- Hongqiao International Institute of Medicine, Tongren Hospital & Shanghai Institute of Immunology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qiaoqiao Han
- Hongqiao International Institute of Medicine, Tongren Hospital & Shanghai Institute of Immunology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhi-Yu Ni
- Clinical Medical College, Hebei University of Engineering, Handan 056038, Hebei, China; Central Laboratory, Hebei Collaborative Innovation Center of Tumor Microecological Metabolism Regulation, Affiliated Hospital of Hebei University, Baoding 071000, Hebei, China; Affiliated Hospital of Hebei Engineering University, Handan 056002, Hebei, China.
| | - Ren Zhao
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Youqiong Ye
- Hongqiao International Institute of Medicine, Tongren Hospital & Shanghai Institute of Immunology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Qiang Zou
- Hongqiao International Institute of Medicine, Tongren Hospital & Shanghai Institute of Immunology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
7
|
Deng W, Zhao Z, Zou T, Kuang T, Wang J. Research Advances in Fusion Protein-Based Drugs for Diabetes Treatment. Diabetes Metab Syndr Obes 2024; 17:343-362. [PMID: 38288338 PMCID: PMC10823413 DOI: 10.2147/dmso.s421527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 12/22/2023] [Indexed: 01/31/2024] Open
Abstract
Diabetes mellitus (DM) is a chronic metabolic disease characterized by elevated blood glucose levels, resulting in multi-organ dysfunction and various complications. Fusion proteins can form multifunctional complexes by combining the target proteins with partner proteins. It has significant advantages in improving the performance of the target proteins, extending their biological half-life, and enhancing patient drug compliance. Fusion protein-based drugs have emerged as promising new drugs in diabetes therapeutics. However, there has not been a systematic review of fusion protein-based drugs for diabetes therapeutics. Hence, we conducted a comprehensive review of published literature on diabetic fusion protein-based drugs for diabetes, with a primary focus on immunoglobulin G (IgG) fragment crystallizable (Fc) region, albumin, and transferrin (TF). This review aims to provide a reference for the subsequent development and clinical application of fusion protein-based drugs in diabetes therapeutics.
Collapse
Affiliation(s)
- Wenying Deng
- School of Basic Medical Sciences, University of South China, Hengyang, Hunan Province, 421001, People’s Republic of China
| | - Zeyi Zhao
- School of Basic Medical Sciences, University of South China, Hengyang, Hunan Province, 421001, People’s Republic of China
| | - Tao Zou
- Department of Cardiovascular Medicine, First Affiliated Hospital of University of South China, Hengyang, Hunan Province, 421001, People’s Republic of China
| | - Tongdong Kuang
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi Province, 541199, People’s Republic of China
| | - Jing Wang
- School of Basic Medical Sciences, University of South China, Hengyang, Hunan Province, 421001, People’s Republic of China
| |
Collapse
|
8
|
Liu Z, Wang D, Zhang J, Xiang P, Zeng Z, Xiong W, Shi L. cGAS-STING signaling in the tumor microenvironment. Cancer Lett 2023; 577:216409. [PMID: 37748723 DOI: 10.1016/j.canlet.2023.216409] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/21/2023] [Accepted: 09/21/2023] [Indexed: 09/27/2023]
Abstract
The cGAS-STING signaling is an important pathway involved in the regulation of tumor microenvironment, which affects many cellular functions including immune activation. Its role in combating tumor progression is widely recognized, especially with its function in inducing innate and adaptive immune responses, on which many immunotherapies have been developed. However, a growing number of findings also suggest a diversity of its roles in shaping tumor microenvironment, including functions that promote tumor progression. Here, we summarize the functions of the cGAS-STING signaling in tumor microenvironment to maintain tumor survival and proliferation through facilitating the forming of an immunosuppressive tumor microenvironment and discuss the current advances of STING-related immunotherapies.
Collapse
Affiliation(s)
- Ziqi Liu
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China; Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Dan Wang
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Jiarong Zhang
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Pingjuan Xiang
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China; Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China; Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.
| | - Lei Shi
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China; Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China; Department of Pathology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
9
|
Hossen MM, Ma Y, Yin Z, Xia Y, Du J, Huang JY, Huang JJ, Zou L, Ye Z, Huang Z. Current understanding of CTLA-4: from mechanism to autoimmune diseases. Front Immunol 2023; 14:1198365. [PMID: 37497212 PMCID: PMC10367421 DOI: 10.3389/fimmu.2023.1198365] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 06/19/2023] [Indexed: 07/28/2023] Open
Abstract
Autoimmune diseases (ADs) are characterized by the production of autoreactive lymphocytes, immune responses to self-antigens, and inflammation in related tissues and organs. Cytotoxic T-lymphocyte antigen 4 (CTLA-4) is majorly expressed in activated T cells and works as a critical regulator in the inflammatory response. In this review, we first describe the structure, expression, and how the signaling pathways of CTLA-4 participate in reducing effector T-cell activity and enhancing the immunomodulatory ability of regulatory T (Treg) cells to reduce immune response, maintain immune homeostasis, and maintain autoimmune silence. We then focused on the correlation between CTLA-4 and different ADs and how this molecule regulates the immune activity of the diseases and inhibits the onset, progression, and pathology of various ADs. Finally, we summarized the current progress of CTLA-4 as a therapeutic target for various ADs.
Collapse
Affiliation(s)
- Md Munnaf Hossen
- Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
- Department of Immunology, Biological Therapy Institute, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Health Science Center, Shenzhen University, Shenzhen, China
- Joint Research Laboratory for Rheumatology of Shenzhen University Health Science Center and Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| | - Yanmei Ma
- Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
- Department of Immunology, Biological Therapy Institute, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Health Science Center, Shenzhen University, Shenzhen, China
- Joint Research Laboratory for Rheumatology of Shenzhen University Health Science Center and Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| | - Zhihua Yin
- Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
- Joint Research Laboratory for Rheumatology of Shenzhen University Health Science Center and Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| | - Yuhao Xia
- Department of Immunology, Biological Therapy Institute, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Health Science Center, Shenzhen University, Shenzhen, China
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jing Du
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jim Yi Huang
- Department of Psychology, University of Oklahoma, Norman, OK, United States
| | - Jennifer Jin Huang
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, United States
| | - Linghua Zou
- Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
- Department of Rehabilitation Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| | - Zhizhong Ye
- Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
- Joint Research Laboratory for Rheumatology of Shenzhen University Health Science Center and Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| | - Zhong Huang
- Department of Immunology, Biological Therapy Institute, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Health Science Center, Shenzhen University, Shenzhen, China
- Joint Research Laboratory for Rheumatology of Shenzhen University Health Science Center and Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| |
Collapse
|
10
|
Bouqdayr M, Abbad A, Baba H, Saih A, Wakrim L, Kettani A. Computational analysis of structural and functional evaluation of the deleterious missense variants in the human CTLA4 gene. J Biomol Struct Dyn 2023; 41:14179-14196. [PMID: 36764830 DOI: 10.1080/07391102.2023.2178509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 02/04/2023] [Indexed: 02/12/2023]
Abstract
CTLA-4 is an immune checkpoint receptor that negatively regulates the T-cell function expressed after T-cell activation to break the immune response. The current study predicted the genomic analysis to explore the functional variations of missense SNPs in the human CTLA4 gene using PolyPhen2, SIFT, PANTHER, PROVEAN, Fathmm, Mutation Assessor, PhD-SNP, SNPs&GO, SNAP2, and MutPred2. Phylogenetic conservation protein was predicted by ConSurf. Protein structural analysis was carried out by I-Mutant3, MUpro, iStable2, PremPS, and ERIS servers. Molecular dynamics trajectory analysis (RMSD, RMSF, Rg, SASA, H-bonds, and PCA) was performed to analyze the dynamic behavior of native and mutant CTLA-4 at the atomic level. Our in-silico analysis suggested that C58S, G118R, P137Q, P137R, P137L, P138T, and G146L variants were predicted to be the most deleterious missense variants and highly conserved residues. Moreover, the molecular dynamics analysis proposed a decrease in the protein stability and compactness with the P137R and P138T highlighting the impact of these variants on the function of the CTLA-4 protein.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Meryem Bouqdayr
- Laboratory of Biology and Health, Faculty of Sciences Ben M'sick, Hassan II University of Casablanca, Casablanca, Morocco
- Virology Unit, Immunovirology Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Anass Abbad
- Medical Virology and BSL-3 Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Hanâ Baba
- Laboratory of Biology and Health, Faculty of Sciences Ben M'sick, Hassan II University of Casablanca, Casablanca, Morocco
- Virology Unit, Immunovirology Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Asmae Saih
- Laboratory of Biology and Health, Faculty of Sciences Ben M'sick, Hassan II University of Casablanca, Casablanca, Morocco
- Virology Unit, Immunovirology Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Lahcen Wakrim
- Virology Unit, Immunovirology Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Anass Kettani
- Laboratory of Biology and Health, Faculty of Sciences Ben M'sick, Hassan II University of Casablanca, Casablanca, Morocco
| |
Collapse
|
11
|
Chen CH, Weng TH, Huang HH, Huang LY, Huang KY, Chen PR, Yeh KY, Huang CT, Chien YT, Chuang PY, Lin YL, Tsai NM, Liu SJ, Su YC, Weng SL, Liao KW. A flexible liposomal polymer complex as a platform of specific and regulable immune regulation for individual cancer immunotherapy. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2023; 42:29. [PMID: 36691089 PMCID: PMC9869520 DOI: 10.1186/s13046-023-02601-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/16/2023] [Indexed: 01/24/2023]
Abstract
BACKGROUND The applicability and therapeutic efficacy of specific personalized immunotherapy for cancer patients is limited by the genetic diversity of the host or the tumor. Side-effects such as immune-related adverse events (IRAEs) derived from the administration of immunotherapy have also been observed. Therefore, regulatory immunotherapy is required for cancer patients and should be developed. METHODS The cationic lipo-PEG-PEI complex (LPPC) can stably and irreplaceably adsorb various proteins on its surface without covalent linkage, and the bound proteins maintain their original functions. In this study, LPPC was developed as an immunoregulatory platform for personalized immunotherapy for tumors to address the barriers related to the heterogenetic characteristics of MHC molecules or tumor associated antigens (TAAs) in the patient population. Here, the immune-suppressive and highly metastatic melanoma, B16F10 cells were used to examine the effects of this platform. Adsorption of anti-CD3 antibodies, HLA-A2/peptide, or dendritic cells' membrane proteins (MP) could flexibly provide pan-T-cell responses, specific Th1 responses, or specific Th1 and Th2 responses, depending on the host needs. Furthermore, with regulatory antibodies, the immuno-LPPC complex properly mediated immune responses by adsorbing positive or negative antibodies, such as anti-CD28 or anti-CTLA4 antibodies. RESULTS The results clearly showed that treatment with LPPC/MP/CD28 complexes activated specific Th1 and Th2 responses, including cytokine release, CTL and prevented T-cell apoptosis. Moreover, LPPC/MP/CD28 complexes could eliminate metastatic B16F10 melanoma cells in the lung more efficiently than LPPC/MP. Interestingly, the melanoma resistance of mice treated with LPPC/MP/CD28 complexes would be reversed to susceptible after administration with LPPC/MP/CTLA4 complexes. NGS data revealed that LPPC/MP/CD28 complexes could enhance the gene expression of cytokine and chemokine pathways to strengthen immune activation than LPPC/MP, and that LPPC/MP/CTLA4 could abolish the LPPC/MP complex-mediated gene expression back to un-treatment. CONCLUSIONS Overall, we proved a convenient and flexible immunotherapy platform for developing personalized cancer therapy.
Collapse
Affiliation(s)
- Chia-Hung Chen
- grid.413593.90000 0004 0573 007XDepartment of Medical Research, Hsinchu MacKay Memorial Hospital, Hsinchu City, 30071 Taiwan
| | - Tzu-Han Weng
- grid.413593.90000 0004 0573 007XDepartment of Dermatology, MacKay Memorial Hospital, Taipei City, 10449 Taiwan
| | - Hsiao-Hsuan Huang
- grid.260539.b0000 0001 2059 7017Industrial Development Graduate Program of College of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu City, 30068 Taiwan
| | - Ling-Ya Huang
- grid.260539.b0000 0001 2059 7017Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu City, 30068 Taiwan
| | - Kai-Yao Huang
- grid.413593.90000 0004 0573 007XDepartment of Medical Research, Hsinchu MacKay Memorial Hospital, Hsinchu City, 30071 Taiwan ,grid.452449.a0000 0004 1762 5613Department of Medicine, MacKay Medical College, 25245 New Taipei City, Taiwan
| | - Pin-Rong Chen
- grid.260539.b0000 0001 2059 7017Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu City, 30068 Taiwan
| | - Kuang-Yu Yeh
- grid.260539.b0000 0001 2059 7017Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu City, 30068 Taiwan
| | - Chi-Ting Huang
- grid.260539.b0000 0001 2059 7017Department of Biological Science and Technology, National Yang Ming Chiao Tung University, 30068 Hsinchu City, Taiwan
| | - Yu-Tzu Chien
- grid.260539.b0000 0001 2059 7017Department of Biological Science and Technology, National Yang Ming Chiao Tung University, 30068 Hsinchu City, Taiwan
| | - Po-Ya Chuang
- grid.260539.b0000 0001 2059 7017Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu City, 30068 Taiwan
| | - Yu-Ling Lin
- grid.28665.3f0000 0001 2287 1366Agricultural Biotechnology Research Center, Academia Sinica, Taipei, 11529 Taiwan
| | - Nu-Man Tsai
- grid.411641.70000 0004 0532 2041Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung City, 40201 Taiwan ,grid.411645.30000 0004 0638 9256Department of Pathology and Clinical Laboratory, Chung Shan Medical University Hospital, Taichung City, 40201 Taiwan
| | - Shih-Jen Liu
- grid.59784.370000000406229172National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, 350401 Miaoli, Taiwan
| | - Yu-Cheng Su
- grid.260539.b0000 0001 2059 7017Department of Biological Science and Technology, National Yang Ming Chiao Tung University, 30068 Hsinchu City, Taiwan
| | - Shun-Long Weng
- grid.452449.a0000 0004 1762 5613Department of Medicine, MacKay Medical College, 25245 New Taipei City, Taiwan ,grid.413593.90000 0004 0573 007XDepartment of Obstetrics and Gynecology, Hsinchu MacKay Memorial Hospital, Hsinchu City, 30071 Taiwan ,grid.507991.30000 0004 0639 3191MacKay Junior College of Medicine, Nursing and Management, Taipei City, 11260 Taiwan
| | - Kuang-Wen Liao
- grid.260539.b0000 0001 2059 7017Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu City, 30068 Taiwan ,grid.260539.b0000 0001 2059 7017Department of Biological Science and Technology, National Yang Ming Chiao Tung University, 30068 Hsinchu City, Taiwan ,grid.412019.f0000 0000 9476 5696Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung City, 80708 Taiwan ,grid.412019.f0000 0000 9476 5696College of Dental Medicine, Kaohsiung Medical University School of Dentistry, Kaohsiung City, 80708 Taiwan ,grid.64523.360000 0004 0532 3255Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan City, 70101 Taiwan ,grid.260539.b0000 0001 2059 7017Center for Intelligent Drug Systems and Smart Bio-Devices, National Yang Ming Chiao Tung University, Hsinchu City, 30068 Taiwan
| |
Collapse
|
12
|
Molon B, Liboni C, Viola A. CD28 and chemokine receptors: Signalling amplifiers at the immunological synapse. Front Immunol 2022; 13:938004. [PMID: 35983040 PMCID: PMC9379342 DOI: 10.3389/fimmu.2022.938004] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/08/2022] [Indexed: 01/14/2023] Open
Abstract
T cells are master regulators of the immune response tuning, among others, B cells, macrophages and NK cells. To exert their functions requiring high sensibility and specificity, T cells need to integrate different stimuli from the surrounding microenvironment. A finely tuned signalling compartmentalization orchestrated in dynamic platforms is an essential requirement for the proper and efficient response of these cells to distinct triggers. During years, several studies have depicted the pivotal role of the cytoskeleton and lipid microdomains in controlling signalling compartmentalization during T cell activation and functions. Here, we discuss mechanisms responsible for signalling amplification and compartmentalization in T cell activation, focusing on the role of CD28, chemokine receptors and the actin cytoskeleton. We also take into account the detrimental effect of mutations carried by distinct signalling proteins giving rise to syndromes characterized by defects in T cell functionality.
Collapse
Affiliation(s)
- Barbara Molon
- Pediatric Research Institute “Città della Speranza”, Corso Stati Uniti, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- *Correspondence: Barbara Molon,
| | - Cristina Liboni
- Pediatric Research Institute “Città della Speranza”, Corso Stati Uniti, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Antonella Viola
- Pediatric Research Institute “Città della Speranza”, Corso Stati Uniti, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| |
Collapse
|
13
|
Henriques-Pons A, Beghini DG, Silva VDS, Iwao Horita S, da Silva FAB. Pulmonary Mesenchymal Stem Cells in Mild Cases of COVID-19 Are Dedicated to Proliferation; In Severe Cases, They Control Inflammation, Make Cell Dispersion, and Tissue Regeneration. Front Immunol 2022; 12:780900. [PMID: 35095855 PMCID: PMC8793136 DOI: 10.3389/fimmu.2021.780900] [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: 09/22/2021] [Accepted: 12/17/2021] [Indexed: 12/29/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent adult stem cells present in virtually all tissues; they have potent self-renewal capacity and differentiate into multiple cell types. For many reasons, these cells are a promising therapeutic alternative to treat patients with severe COVID-19 and pulmonary post-COVID sequelae. These cells are not only essential for tissue regeneration; they can also alter the pulmonary environment through the paracrine secretion of several mediators. They can control or promote inflammation, induce other stem cells differentiation, restrain the virus load, and much more. In this work, we performed single-cell RNA-seq data analysis of MSCs in bronchoalveolar lavage samples from control individuals and COVID-19 patients with mild and severe clinical conditions. When we compared samples from mild cases with control individuals, most genes transcriptionally upregulated in COVID-19 were involved in cell proliferation. However, a new set of genes with distinct biological functions was upregulated when we compared severely affected with mild COVID-19 patients. In this analysis, the cells upregulated genes related to cell dispersion/migration and induced the γ-activated sequence (GAS) genes, probably triggered by IFNGR1 and IFNGR2. Then, IRF-1 was upregulated, one of the GAS target genes, leading to the interferon-stimulated response (ISR) and the overexpression of many signature target genes. The MSCs also upregulated genes involved in the mesenchymal-epithelial transition, virus control, cell chemotaxis, and used the cytoplasmic RNA danger sensors RIG-1, MDA5, and PKR. In a non-comparative analysis, we observed that MSCs from severe cases do not express many NF-κB upstream receptors, such as Toll-like (TLRs) TLR-3, -7, and -8; tumor necrosis factor (TNFR1 or TNFR2), RANK, CD40, and IL-1R1. Indeed, many NF-κB inhibitors were upregulated, including PPP2CB, OPTN, NFKBIA, and FHL2, suggesting that MSCs do not play a role in the "cytokine storm" observed. Therefore, lung MSCs in COVID-19 sense immune danger and act protectively in concert with the pulmonary environment, confirming their therapeutic potential in cell-based therapy for COVID-19. The transcription of MSCs senescence markers is discussed.
Collapse
Affiliation(s)
- Andrea Henriques-Pons
- Fundação Oswaldo Cruz, Instituto Oswaldo Cruz, Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Rio de Janeiro, Brazil
| | - Daniela Gois Beghini
- Fundação Oswaldo Cruz, Instituto Oswaldo Cruz, Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Rio de Janeiro, Brazil
| | | | - Samuel Iwao Horita
- Fundação Oswaldo Cruz, Instituto Oswaldo Cruz, Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Rio de Janeiro, Brazil
| | | |
Collapse
|
14
|
Barros L, Ferreira C, Veldhoen M. The fellowship of regulatory and tissue-resident memory cells. Mucosal Immunol 2022; 15:64-73. [PMID: 34608235 PMCID: PMC8488068 DOI: 10.1038/s41385-021-00456-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 02/04/2023]
Abstract
T cells located in non-lymphoid tissues have come to prominence in recent years. CD8+ tissue-resident memory (Trm) cells are important for tissue immune surveillance, provide an important line of defence against invading pathogens and show promise in cancer therapies. These cells differ in phenotype from other memory populations, are adapted to the tissue they home to where they found their cognate antigen and have different metabolic requirements for survival and activation. CD4+ Foxp3+ regulatory T (Treg) cells also consist of specialised populations, found in non-lymphoid tissues, with distinct transcriptional programmes. These cells have equally adapted to function in the tissue they made their home. Both Trm and Treg cells have functions beyond immune defence, involving tissue homeostasis, repair and turnover. They are part of a multicellular communication network. Intriguingly, occupying the same niche, Treg cells are important in the establishment of Trm cells, which may have implications to harness the immune surveillance and tissue homeostasis properties of Trm cells for future therapies.
Collapse
Affiliation(s)
- Leandro Barros
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Av. Professor Egas Moniz, Lisbon, 1649-028, Portugal
| | - Cristina Ferreira
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Av. Professor Egas Moniz, Lisbon, 1649-028, Portugal
| | - Marc Veldhoen
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Av. Professor Egas Moniz, Lisbon, 1649-028, Portugal.
| |
Collapse
|
15
|
Kanda Y, Okazaki T, Katakai T. Motility Dynamics of T Cells in Tumor-Draining Lymph Nodes: A Rational Indicator of Antitumor Response and Immune Checkpoint Blockade. Cancers (Basel) 2021; 13:4616. [PMID: 34572844 PMCID: PMC8465463 DOI: 10.3390/cancers13184616] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/12/2021] [Accepted: 09/13/2021] [Indexed: 01/22/2023] Open
Abstract
The migration status of T cells within the densely packed tissue environment of lymph nodes reflects the ongoing activation state of adaptive immune responses. Upon encountering antigen-presenting dendritic cells, actively migrating T cells that are specific to cognate antigens slow down and are eventually arrested on dendritic cells to form immunological synapses. This dynamic transition of T cell motility is a fundamental strategy for the efficient scanning of antigens, followed by obtaining the adequate activation signals. After receiving antigenic stimuli, T cells begin to proliferate, and the expression of immunoregulatory receptors (such as CTLA-4 and PD-1) is induced on their surface. Recent findings have revealed that these 'immune checkpoint' molecules control the activation as well as motility of T cells in various situations. Therefore, the outcome of tumor immunotherapy using checkpoint inhibitors is assumed to be closely related to the alteration of T cell motility, particularly in tumor-draining lymph nodes (TDLNs). In this review, we discuss the migration dynamics of T cells during their activation in TDLNs, and the roles of checkpoint molecules in T cell motility, to provide some insight into the effect of tumor immunotherapy via checkpoint blockade, in terms of T cell dynamics and the importance of TDLNs.
Collapse
Affiliation(s)
- Yasuhiro Kanda
- Department of Immunology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 950-8510, Japan;
| | - Taku Okazaki
- Laboratory of Molecular Immunology, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo 113-0032, Japan;
| | - Tomoya Katakai
- Department of Immunology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 950-8510, Japan;
| |
Collapse
|
16
|
Sobhani N, Tardiel-Cyril DR, Davtyan A, Generali D, Roudi R, Li Y. CTLA-4 in Regulatory T Cells for Cancer Immunotherapy. Cancers (Basel) 2021; 13:1440. [PMID: 33809974 PMCID: PMC8005092 DOI: 10.3390/cancers13061440] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 12/14/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have obtained durable responses in many cancers, making it possible to foresee their potential in improving the health of cancer patients. However, immunotherapies are currently limited to a minority of patients and there is a need to develop a better understanding of the basic molecular mechanisms and functions of pivotal immune regulatory molecules. Immune checkpoint cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and regulatory T (Treg) cells play pivotal roles in hindering the anticancer immunity. Treg cells suppress antigen-presenting cells (APCs) by depleting immune stimulating cytokines, producing immunosuppressive cytokines and constitutively expressing CTLA-4. CTLA-4 molecules bind to CD80 and CD86 with a higher affinity than CD28 and act as competitive inhibitors of CD28 in APCs. The purpose of this review is to summarize state-of-the-art understanding of the molecular mechanisms underlining CTLA-4 immune regulation and the correlation of the ICI response with CTLA-4 expression in Treg cells from preclinical and clinical studies for possibly improving CTLA-4-based immunotherapies, while highlighting the knowledge gap.
Collapse
Affiliation(s)
- Navid Sobhani
- Department of Medicine, Section of Epidemiology and Population Sciences, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Dana Rae Tardiel-Cyril
- Department of Medicine, Section of Epidemiology and Population Sciences, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Aram Davtyan
- Atomwise, 717 Market St, San Francisco, CA 94103, USA;
| | - Daniele Generali
- Department of Medical, Surgery and Health Sciences, University of Trieste, 34147 Trieste, Italy;
| | - Raheleh Roudi
- Department of Medicine, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
| | - Yong Li
- Department of Medicine, Section of Epidemiology and Population Sciences, Baylor College of Medicine, Houston, TX 77030, USA;
| |
Collapse
|
17
|
Chang M, Hou Z, Wang M, Li C, Lin J. Recent Advances in Hyperthermia Therapy-Based Synergistic Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2004788. [PMID: 33289219 DOI: 10.1002/adma.202004788] [Citation(s) in RCA: 233] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/17/2020] [Indexed: 06/12/2023]
Abstract
The past decades have witnessed hyperthermia therapy (HTT) as an emerging strategy against malignant tumors. Nanomaterial-based photothermal therapy (PTT) and magnetic hyperthermia (MHT), as highly effective and noninvasive treatment models, offer advantages over other strategies in the treatment of different types of tumors. However, both PTT and MHT cannot completely cure cancer due to recurrence and distal metastasis. In recent years, cancer immunotherapy has attracted widespread attention owing to its capability to activate the body's own natural defense to identify, attack, and eradicate cancer cells. Significant efforts have been devoted to studying the activated immune responses caused by hyperthermia-ablated tumors. In this article, the synergistic mechanism of HTT in immunotherapy, including immunogenic cell death and reversal of the immunosuppressive tumor microenvironment is discussed. The reports of the combination of HTT or HTT-based multimodal therapy with immunotherapy, including immunoadjuvant exploitation, immune checkpoint blockade therapy, and adoptive cellular immunotherapy are summarized. As highlighted, these strategies could achieve synergistically enhanced therapeutic outcomes against both primary tumors and metastatic lesions, prevent cancer recurrence, and prolong the survival period. Finally, current challenges and prospective developments in HTT-synergized immunotherapy are also reviewed.
Collapse
Affiliation(s)
- Mengyu Chang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Sciences and Technology of China, Hefei, 230026, P. R. China
| | - Zhiyao Hou
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangdong, 511436, P. R. China
- Department of Abdominal Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, P. R. China
| | - Man Wang
- Institute of Molecular Sciences and Engineering, Shandong University, Qingdao, 266237, P. R. China
| | - Chunxia Li
- Institute of Molecular Sciences and Engineering, Shandong University, Qingdao, 266237, P. R. China
| | - Jun Lin
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Sciences and Technology of China, Hefei, 230026, P. R. China
| |
Collapse
|
18
|
Weinmann A, Galle PR. Role of immunotherapy in the management of hepatocellular carcinoma: current standards and future directions. Curr Oncol 2020; 27:S152-S164. [PMID: 33343209 PMCID: PMC7739523 DOI: 10.3747/co.27.7315] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The multikinase inhibitor sorafenib was the only approved systemic therapy in advanced hepatocellular carcinoma (hcc) for about a decade. In recent years, the number of approved agents has increased significantly as a result of a number of positive phase iii clinical trials. Lenvatinib as a first-line treatment, and regorafenib, cabozantinib, and ramucirumab in the second-line setting are now approved by the U.S. Food and Drug Administration (fda) and the European Medicines Agency. In phase ii studies, immunotherapy with nivolumab and monotherapy using pembrolizumab yielded impressive results for overall survival in therapy-naïve and pretreated patients, leading to the accelerated approval by the fda of nivolumab and pembrolizumab for second-line treatment. However, phase iii trials of nivolumab in the first line and pembrolizumab in the second line as single agents failed to reach statistical significance, although clinical benefit for a subset of patients with long durations of response could be demonstrated. Despite that setback, immunotherapy for hcc is a promising therapeutic approach, and the combination of immunotherapy with other treatment modalities such as monoclonal antibodies, tyrosine kinase inhibitors, or local therapies has the potential to increase the overall response rate and survival. Recently, the results of a phase iii trial of combination atezolizumab-bevacizumab compared with sorafenib showed a highly significant survival benefit and median overall survival that was not reached in the immunotherapy arm, making the combination the preferred standard of care in first-line therapy. Despite the impressive results and generally good toxicity profile of immunotherapy, patients who respond to therapy constitute only a subset of the overall population, and response rates are still limited. This review focuses on the currently reported results and ongoing clinical trials of checkpoint inhibitor-based immunotherapy in hcc.
Collapse
Affiliation(s)
- A Weinmann
- Department of Internal Medicine i, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany
| | - P R Galle
- Department of Internal Medicine i, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
19
|
Park S, Shi Y, Kim BC, Jo MH, Cruz LO, Gou Z, Ha T, Lu LF, Reich DH, Chen Y. Force-dependent trans-endocytosis by breast cancer cells depletes costimulatory receptor CD80 and attenuates T cell activation. Biosens Bioelectron 2020; 165:112389. [PMID: 32729511 DOI: 10.1016/j.bios.2020.112389] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 06/08/2020] [Accepted: 06/10/2020] [Indexed: 12/11/2022]
Abstract
In this study, we investigated the biophysical interaction between cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and CD80. CTLA-4 is a key molecule in immunosuppression, and CD80 is a costimulatory receptor promoting T cell activation. We observed that after cell-cell contact was established between breast cancer cells and antigen presenting cells (APCs), CTLA-4 expressed on the breast cancer cells bind to CD80 expressed on the APCs, and underwent trans-endocytosis to deplete CD80. Force measurement and live cell imaging revealed that upon binding to CD80, forces generated by breast cancer cells and transmitted via CTLA-4 were sufficiently strong to displace CD80 from the surface of APCs to be internalized by breast cancer cells. We further demonstrated that because of the force-dependent trans-endocytosis of CD80, the capacity of APCs to activate T cells was significantly attenuated. Furthermore, inhibiting force generation in cancer cells would increase the T cell activating capacity of APCs. Our results provide a possible mechanism behind the immunosuppression commonly seen in breast cancer patients, and may lead to a new strategy to restore anti-tumor immunity by inhibiting pathways of force-generation.
Collapse
Affiliation(s)
- Seungman Park
- Department of Mechanical Engineering, Johns Hopkins University, MD, 21218, USA; Center for Cell Dynamics, Johns Hopkins University, MD, 21218, USA; Institute for NanoBioTechnology, Johns Hopkins University, MD, 21218, USA
| | - Yu Shi
- Department of Physics & Astronomy, Johns Hopkins University, MD, 21218, USA
| | - Byoung Choul Kim
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA; Howard Hughes Medical Institute, Baltimore, MD, 21205, USA; Division of Nano-Bioengineering, Incheon National University, Incheon, 22012, South Korea
| | - Myung Hyun Jo
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Leilani O Cruz
- Division of Biological Science, University of California, San Diego, CA, 92093, USA
| | - Zheming Gou
- Department of Mechanical Engineering, Johns Hopkins University, MD, 21218, USA
| | - Taekjip Ha
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA; Howard Hughes Medical Institute, Baltimore, MD, 21205, USA
| | - Li-Fan Lu
- Division of Biological Science, University of California, San Diego, CA, 92093, USA
| | - Daniel H Reich
- Department of Physics & Astronomy, Johns Hopkins University, MD, 21218, USA
| | - Yun Chen
- Department of Mechanical Engineering, Johns Hopkins University, MD, 21218, USA; Center for Cell Dynamics, Johns Hopkins University, MD, 21218, USA; Institute for NanoBioTechnology, Johns Hopkins University, MD, 21218, USA.
| |
Collapse
|
20
|
Aref S, El Agdar M, El Sebaie A, Abouzeid T, Sabry M, Ibrahim L. Prognostic Value of CD200 Expression and Soluble CTLA-4 Concentrations in Intermediate and High-Risk Myelodysplastic Syndrome Patients. Asian Pac J Cancer Prev 2020; 21:2225-2230. [PMID: 32856848 PMCID: PMC7771941 DOI: 10.31557/apjcp.2020.21.8.2225] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE This study was designed in order to identify the prognostic relevance of CD200 expression and soluble Cytotoxic T-lymphocyte antigen-4 (CTLA-4) levels in myelodysplastic syndrome (MDS) patients. METHODS The study included 57 MDS (37 intermediate and 20 high risk) patients and 10 controls. For all of included patients; CD200 expression was identified by flowcytometry on CD33 positive cells and soluble CTLA-4 (CD152) concentration was determined by ELISA. RESULTS CD200 positive expression was detected in 32/57 (56.1%) of MDS cases, the mean serum CTLA-4 concentrations were significantly higher in MDS patients as compared to controls (P<0.01). Significant association between high CD200 positive expression; high CTLA-4 concentration levels and MDS risk stages being higher in high risk MDS group as compared to intermediate risk one (P < 0.01). After 36-month follow-up; the subgroup of MDS patients with high expression of CD200; and high serum CTLA-4 concentrations showed high death rate and high frequency of acute myeloid leukemia transformation. CONCLUSIONS CD200 positive expression could be considered as a new prognostic marker for risk stratification of MDS patients. CD200 expression may exert its effect through upregulation of CTLA-4.<br />.
Collapse
Affiliation(s)
- Salah Aref
- Hematology Unit, Clinical Pathology Department, Faculty of Medicine, Mansoura University, Egypt.,Hematology Unit, Mansoura University Oncology Center, Faculty of Medicine, Mansoura University, Egypt
| | - Mohamed El Agdar
- Hematology Unit, Mansoura University Oncology Center, Faculty of Medicine, Mansoura University, Egypt
| | - Ahmed El Sebaie
- Hematology Unit, Clinical Pathology Department, Faculty of Medicine, Mansoura University, Egypt
| | - Tarek Abouzeid
- Clinical Hematology unit, Mansoura University Oncology Center, Faculty of Medicine, Mansoura University, Egypt
| | - Mohamed Sabry
- Hematology Unit, Mansoura University Oncology Center, Faculty of Medicine, Mansoura University, Egypt
| | - Lamiaa Ibrahim
- Hematology Unit, Clinical Pathology Department, Faculty of Medicine, Mansoura University, Egypt
| |
Collapse
|
21
|
Lau D, Garçon F, Chandra A, Lechermann LM, Aloj L, Chilvers ER, Corrie PG, Okkenhaug K, Gallagher FA. Intravital Imaging of Adoptive T-Cell Morphology, Mobility and Trafficking Following Immune Checkpoint Inhibition in a Mouse Melanoma Model. Front Immunol 2020; 11:1514. [PMID: 32793206 PMCID: PMC7387409 DOI: 10.3389/fimmu.2020.01514] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 06/09/2020] [Indexed: 12/13/2022] Open
Abstract
Efficient T-cell targeting, infiltration and activation within tumors is crucial for successful adoptive T-cell therapy. Intravital microscopy is a powerful tool for the visualization of T-cell behavior within tumors, as well as spatial and temporal heterogeneity in response to immunotherapy. Here we describe an experimental approach for intravital imaging of adoptive T-cell morphology, mobility and trafficking in a skin-flap tumor model, following immune modulation with immune checkpoint inhibitors (ICIs) targeting PD-L1 and CTLA-4. A syngeneic model of ovalbumin and mCherry-expressing amelanotic mouse melanoma was used in conjunction with adoptively transferred OT-1+ cytotoxic T-cells expressing GFP to image antigen-specific live T-cell behavior within the tumor microenvironment. Dynamic image analysis of T-cell motility showed distinct CD8+ T-cell migration patterns and morpho-dynamics within different tumor compartments in response to ICIs: this approach was used to cluster T-cell behavior into four groups based on velocity and meandering index. The results showed that most T-cells within the tumor periphery demonstrated Lévy-like trajectories, consistent with tumor cell searching strategies. T-cells adjacent to tumor cells had reduced velocity and appeared to probe the local environment, consistent with cell-cell interactions. An increased number of T-cells were detected following treatment, traveling at lower mean velocities than controls, and demonstrating reduced displacement consistent with target engagement. Histogram-based analysis of immunofluorescent images from harvested tumors showed that in the ICI-treated mice there was a higher density of CD31+ vessels compared to untreated controls and a greater infiltration of T-cells towards the tumor core, consistent with increased cellular trafficking post-treatment.
Collapse
Affiliation(s)
- Doreen Lau
- Cancer Research UK Cambridge Centre, Cambridge, United Kingdom
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
| | - Fabien Garçon
- Laboratory of Lymphocyte Signaling and Development, The Babraham Institute, Cambridge, United Kingdom
| | - Anita Chandra
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | | | - Luigi Aloj
- Cancer Research UK Cambridge Centre, Cambridge, United Kingdom
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
- Department of Nuclear Medicine, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Edwin R. Chilvers
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Pippa G. Corrie
- Department of Oncology, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Klaus Okkenhaug
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Ferdia A. Gallagher
- Cancer Research UK Cambridge Centre, Cambridge, United Kingdom
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
22
|
Sun F, Ganguli A, Nguyen J, Brisbin R, Shanmugam K, Hirschberg DL, Wheeler MB, Bashir R, Nash DM, Cunningham BT. Smartphone-based multiplex 30-minute nucleic acid test of live virus from nasal swab extract. LAB ON A CHIP 2020; 20:1621-1627. [PMID: 32334422 DOI: 10.1039/d0lc00304b] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Rapid, sensitive and specific detection and reporting of infectious pathogens is important for patient management and epidemic surveillance. We demonstrated a point-of-care system integrated with a smartphone for detecting live virus from nasal swab media, using a panel of equine respiratory infectious diseases as a model system for corresponding human diseases such as COVID-19. Specific nucleic acid sequences of five pathogens were amplified by loop-mediated isothermal amplification on a microfluidic chip and detected at the end of reactions by the smartphone. Pathogen-spiked horse nasal swab samples were correctly diagnosed using our system, with a limit of detection comparable to that of the traditional lab-based test, polymerase chain reaction, with results achieved in ∼30 minutes.
Collapse
Affiliation(s)
- Fu Sun
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Illinois, USA.
| | - Anurup Ganguli
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Illinois, USA
| | | | - Ryan Brisbin
- Department of Interdisciplinary Arts and Sciences & The Center for Urban Waters, University of Washington Tacoma, Washington, USA
| | - Krithika Shanmugam
- Department of Interdisciplinary Arts and Sciences & The Center for Urban Waters, University of Washington Tacoma, Washington, USA
| | - David L Hirschberg
- RAIN Incubator, Tacoma, Washington, USA and Department of Interdisciplinary Arts and Sciences & The Center for Urban Waters, University of Washington Tacoma, Washington, USA
| | - Matthew B Wheeler
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Rashid Bashir
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Illinois, USA. and Department of Bioengineering, University of Illinois at Urbana-Champaign, Illinois, USA
| | | | - Brian T Cunningham
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Illinois, USA. and Department of Bioengineering, University of Illinois at Urbana-Champaign, Illinois, USA
| |
Collapse
|
23
|
Van Coillie S, Wiernicki B, Xu J. Molecular and Cellular Functions of CTLA-4. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1248:7-32. [PMID: 32185705 DOI: 10.1007/978-981-15-3266-5_2] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) is an inhibitory receptor belonging to the CD28 immunoglobulin subfamily, expressed primarily by T-cells. Its ligands, CD80 and CD86, are typically found on the surface of antigen-presenting cells and can either bind CD28 or CTLA-4, resulting in a costimulatory or a co-inhibitory response, respectively. Because of its dampening effect, CTLA-4 is a crucial regulator of T-cell homeostasis and self-tolerance. The mechanisms by which CTLA-4 exerts its inhibitory function can be categorized as either cell-intrinsic (affects the CTLA-4 expressing T-cell) or cell-extrinsic (affects secondary cells). Research from the last decade has shown that CTLA-4 mainly acts in a cell-extrinsic manner via its competition with CD28, CTLA-4-mediated trans-endocytosis of CD80 and CD86, and its direct tolerogenic effects on the interacting cell. Nonetheless, intrinsic CTLA-4 signaling has been implicated in T-cell motility and the regulation of CTLA-4 its subcellular localization amongst others. CTLA-4 is well recognized as a key immune checkpoint and has gained significant momentum as a therapeutic target in the field of autoimmunity and cancer. In this chapter, we describe the role of costimulation in immune response induction as well as the main mechanisms by which CTLA-4 can inhibit this process.
Collapse
Affiliation(s)
- Samya Van Coillie
- Molecular Signaling and Cell Death Unit, VIB-UGent Center for Inflammation Research, Zwijnaarde, 9052, Ghent, Belgium.
| | - Bartosz Wiernicki
- Molecular Signaling and Cell Death Unit, VIB-UGent Center for Inflammation Research, Zwijnaarde, 9052, Ghent, Belgium
| | - Jie Xu
- Institutes of Biomedical Sciences, Zhongshan-Xuhui Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
24
|
Yeung MY, Grimmig T, Sayegh MH. Costimulation Blockade in Transplantation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1189:267-312. [PMID: 31758538 DOI: 10.1007/978-981-32-9717-3_10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
T cells play a pivotal role in orchestrating immune responses directed against a foreign (allogeneic) graft. For T cells to become fully activated, the T-cell receptor (TCR) must interact with the major histocompatibility complex (MHC) plus peptide complex on antigen-presenting cells (APCs), followed by a second "positive" costimulatory signal. In the absence of this second signal, T cells become anergic or undergo deletion. By blocking positive costimulatory signaling, T-cell allo-responses can be aborted, thus preventing graft rejection and promoting long-term allograft survival and possibly tolerance (Alegre ML, Najafian N, Curr Mol Med 6:843-857, 2006; Li XC, Rothstein DM, Sayegh MH, Immunol Rev 229:271-293, 2009). In addition, costimulatory molecules can provide negative "coinhibitory" signals that inhibit T-cell activation and terminate immune responses; strategies to promote these pathways can also lead to graft tolerance (Boenisch O, Sayegh MH, Najafian N, Curr Opin Organ Transplant 13:373-378, 2008). However, T-cell costimulation involves an incredibly complex array of interactions that may act simultaneously or at different times in the immune response and whose relative importance varies depending on the different T-cell subsets and activation status. In transplantation, the presence of foreign alloantigen incites not only destructive T effector cells but also protective regulatory T cells, the balance of which ultimately determines the fate of the allograft (Lechler RI, Garden OA, Turka LA, Nat Rev Immunol 3:147-158, 2003). Since the processes of alloantigen-specific rejection and regulation both require activation of T cells, costimulatory interactions may have opposing or synergistic roles depending on the cell being targeted. Such complexities present both challenges and opportunities in targeting T-cell costimulatory pathways for therapeutic purposes. In this chapter, we summarize our current knowledge of the various costimulatory pathways in transplantation and review the current state and challenges of harnessing these pathways to promote graft tolerance (summarized in Table 10.1).
Collapse
Affiliation(s)
- Melissa Y Yeung
- Department of Medicine, Renal Division, Brigham and Women's Hospital, Boston, MA, USA. .,Harvard Medical School, Boston, MA, USA.
| | - Tanja Grimmig
- Department of Surgery, Molecular Oncology and Immunology, University of Wuerzburg, Wuerzburg, Germany
| | - Mohamed H Sayegh
- Department of Medicine, Renal Division, Brigham and Women's Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA.,Department of Medicine and Immunology, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
25
|
Mitsuiki N, Schwab C, Grimbacher B. What did we learn from CTLA-4 insufficiency on the human immune system? Immunol Rev 2019; 287:33-49. [PMID: 30565239 DOI: 10.1111/imr.12721] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Accepted: 09/16/2018] [Indexed: 02/07/2023]
Abstract
Cytotoxic-T-lymphocyte-antigen-4 (CTLA-4) is a negative immune regulator constitutively expressed on regulatory T (Treg) cells and upregulated on activated T cells. CTLA-4 inhibits T cell activation by various suppressive functions including competition with CD28, regulation of the inhibitory function of Treg cells, such as transendocytosis, and the control of adhesion and motility. Intrinsic CTLA-4 signaling has been controversially discussed, but so far no distinct signaling pathway has been identified. The CTLA-4-mediated Treg suppression plays an important role in the maintenance of peripheral tolerance and the prevention of autoimmune diseases. Human CTLA-4 insufficiency is caused by heterozygous germline mutations in CTLA4 and characterized by a complex immune dysregulation syndrome. Clinical studies on CTLA4 mutation carriers showed a reduced penetrance and variable expressivity, suggesting modifying factor(s). One hundred and forty-eight CTLA4 mutation carriers have been reported; patients showed hypogammaglobulinemia, recurrent infectious diseases, various autoimmune diseases, and lymphocytic infiltration into multiple organs. The CTLA-4 expression level in Treg cells was reduced, while the frequency of Treg cells was increased in CTLA-4-insufficient patients. The transendocytosis assay is a specific functional test for the assessment of newly identified CTLA4 gene variants. Immunoglobulin substitution, corticosteroids, immunosuppressive therapy, and targeted therapy such as with CTLA-4 fusion proteins and mechanistic target of rapamycin (mTOR) inhibitors were applied; patients with life-threatening, treatment-resistant symptoms underwent hematopoietic stem cell transplantation. The fact that in humans CTLA-4 insufficiency causes severe disease taught us that the amount of CTLA-4 molecules present in/on T cells matters for immune homeostasis. However, whether the pathology-causing activated T lymphocytes in CTLA-4-insufficient patients are antigen-specific is an unsolved question. CTLA-4, in addition, has a role in autoimmune diseases and cancer. Anti-CTLA-4 drugs are employed as checkpoint inhibitors to target various forms of cancer. Thus, clinical research on human CTLA-4 insufficiency might provide us a deeper understanding of the mechanism(s) of the CTLA-4 molecule and immune dysregulation disorders.
Collapse
Affiliation(s)
- Noriko Mitsuiki
- Center for Chronic Immunodeficiency (CCI), Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Charlotte Schwab
- Center for Chronic Immunodeficiency (CCI), Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Bodo Grimbacher
- Center for Chronic Immunodeficiency (CCI), Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
26
|
Disorders of CTLA-4 expression, how they lead to CVID and dysregulated immune responses. Curr Opin Allergy Clin Immunol 2019; 19:578-585. [DOI: 10.1097/aci.0000000000000590] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
27
|
Czaja AJ. Immune inhibitory proteins and their pathogenic and therapeutic implications in autoimmunity and autoimmune hepatitis. Autoimmunity 2019; 52:144-160. [PMID: 31298041 DOI: 10.1080/08916934.2019.1641200] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Key inhibitory proteins can blunt immune responses to self-antigens, and deficiencies in this repertoire may promote autoimmunity. The goals of this review are to describe the key immune inhibitory proteins, indicate their possible impact on the development of autoimmune disease, especially autoimmune hepatitis, and encourage studies to clarify their pathogenic role and candidacy as therapeutic targets. English abstracts were identified in PubMed by multiple search terms. Full length articles were selected for review, and secondary and tertiary bibliographies were developed. Cytotoxic T lymphocyte antigen-4 impairs ligation of CD28 to B7 ligands on antigen presenting cells and inhibits the adaptive immune response by increasing anti-inflammatory cytokines, generating regulatory T cells, and reducing T cell activation and proliferation. Programed cell death antigen-1 inhibits T cell selection, activation, and proliferation by binding with two ligands at different phases and locations of the immune response. A soluble alternatively spliced variant of this protein can dampen the inhibitory signal. Autoimmune hepatitis has been associated with polymorphisms of the cytotoxic T lymphocyte antigen-4 gene, reduced hepatic expression of a ligand of programed cell death antigen-1, an interfering soluble variant of this key inhibitory protein, and antibodies against it. Findings have been associated with laboratory indices of liver injury and suboptimal treatment response. Abatacept, belatacept, CD28 blockade, and induction of T cell exhaustion are management considerations that require scrutiny. In conclusion, deficiencies in key immune inhibitory proteins may promote the occurrence of autoimmune diseases, such as autoimmune hepatitis, and emerging interventions may overcome these deficiencies. Investigations should define the nature, impact and management of these inhibitory disturbances in autoimmune hepatitis.
Collapse
Affiliation(s)
- Albert J Czaja
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science , Rochester , MN , USA
| |
Collapse
|
28
|
Tumor mechanisms of resistance to immune attack. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 164:61-100. [PMID: 31383409 DOI: 10.1016/bs.pmbts.2019.03.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The immune system plays a key role in the interactions between host and tumor. Immune selection pressure is a driving force behind the sculpting and evolution of malignant cancer cells to escape this immune attack. Several common tumor cell-based mechanisms of resistance to immune attack have been identified and can be broadly categorized into three main classes: loss of antigenicity, loss of immunogenicity, and creation of an immunosuppressive microenvironment. In this review, we will discuss in detail the relevant literature associated with each class of resistance and will describe the relevance of these mechanisms to human cancer patients. To conclude, we will outline the implications these mechanisms have for the treatment of cancer using currently available therapeutic approaches. Immunotherapy has been a successful addition to current treatment approaches, but many patients either do not respond or quickly become resistant. This reflects the ability of tumors to continue to adapt to immune selection pressure at all stages of development. Additional study of immune escape mechanisms and immunotherapy resistance mechanisms will be needed to inform future treatment approaches.
Collapse
|
29
|
Haas OA. Primary Immunodeficiency and Cancer Predisposition Revisited: Embedding Two Closely Related Concepts Into an Integrative Conceptual Framework. Front Immunol 2019; 9:3136. [PMID: 30809233 PMCID: PMC6379258 DOI: 10.3389/fimmu.2018.03136] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 12/19/2018] [Indexed: 12/13/2022] Open
Abstract
Common understanding suggests that the normal function of a "healthy" immune system safe-guards and protects against the development of malignancies, whereas a genetically impaired one might increase the likelihood of their manifestation. This view is primarily based on and apparently supported by an increased incidence of such diseases in patients with specific forms of immunodeficiencies that are caused by high penetrant gene defects. As I will review and discuss herein, such constellations merely represent the tip of an iceberg. The overall situation is by far more varied and complex, especially if one takes into account the growing difficulties to define what actually constitutes an immunodeficiency and what defines a cancer predisposition. The enormous advances in genome sequencing, in bioinformatic analyses and in the functional in vitro and in vivo assessment of novel findings together with the availability of large databases provide us with a wealth of information that steadily increases the number of sequence variants that concur with clinically more or less recognizable immunological problems and their consequences. Since many of the newly identified hard-core defects are exceedingly rare, their tumor predisposing effect is difficult to ascertain. The analyses of large data sets, on the other hand, continuously supply us with low penetrant variants that, at least in statistical terms, are clearly tumor predisposing, although their specific relevance for the respective carriers still needs to be carefully assessed on an individual basis. Finally, defects and variants that affect the same gene families and pathways in both a constitutional and somatic setting underscore the fact that immunodeficiencies and cancer predisposition can be viewed as two closely related errors of development. Depending on the particular genetic and/or environmental context as well as the respective stage of development, the same changes can have either a neutral, predisposing and, in some instances, even a protective effect. To understand the interaction between the immune system, be it "normal" or "deficient" and tumor predisposition and development on a systemic level, one therefore needs to focus on the structure and dynamic functional organization of the entire immune system rather than on its isolated individual components alone.
Collapse
Affiliation(s)
- Oskar A. Haas
- Department of Clinical Genetics, Children's Cancer Research Institute, Vienna, Austria
| |
Collapse
|
30
|
Co-signal Molecules in T-Cell Activation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1189:3-23. [DOI: 10.1007/978-981-32-9717-3_1] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
31
|
Brunner-Weinzierl MC, Rudd CE. CTLA-4 and PD-1 Control of T-Cell Motility and Migration: Implications for Tumor Immunotherapy. Front Immunol 2018; 9:2737. [PMID: 30542345 PMCID: PMC6277866 DOI: 10.3389/fimmu.2018.02737] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 11/06/2018] [Indexed: 12/12/2022] Open
Abstract
CTLA-4 is a co-receptor on T-cells that controls peripheral tolerance and the development of autoimmunity. Immune check-point blockade (ICB) uses monoclonal antibodies (MAbs) to block the binding of inhibitory receptors (IRs) to their natural ligands. A humanized antibody to CTLA-4 was first approved clinically followed by the use of antibody blockade against PD-1 and its ligand PD-L1. Effective anti-tumor immunity requires the activation of tumor-specific effector T-cells, the blockade of regulatory cells and the migration of T-cells into the tumor. Here, we review data implicating CTLA-4 and PD-1 in the motility of T-cells with a specific reference to the potential exploitation of these pathways for more effective tumor infiltration and eradication.
Collapse
Affiliation(s)
- Monika C Brunner-Weinzierl
- Department of Experimental Pediatrics, University Hospital, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, Magdeburg, Germany
| | - Christopher E Rudd
- Research Center-Maisonneuve-Rosemont Hospital (CRHMR), Montreal, QC, Canada.,Département de Medicine, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
32
|
Ninomiya K, Hotta K. Pembrolizumab for the first-line treatment of non-small cell lung cancer. Expert Opin Biol Ther 2018; 18:1015-1021. [PMID: 30207786 DOI: 10.1080/14712598.2018.1522300] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Platinum-based chemotherapy had long played a role as standard therapy for the first-line treatment of advanced or recurrent non-small cell lung cancer (NSCLC). However, immune checkpoint inhibitors such as pembrolizumab, a monoclonal antibody that prevents programmed death protein 1 (PD-1) receptor, have brought a paradigm shift in this field. Areas covered: In this article, we review the relevant literatures and ongoing trials on the first-line treatment of pembrolizumab. Especially, in two pivotal phase III trials, KEYNOTE-024 and -189, both pembrolizumab monotherapy and combined pembrolizumab plus chemotherapy significantly prolonged overall survival (OS) compared to the existing platinum-based chemotherapy. Currently, multiple trials with combination therapy of pembrolizumab and other agents have been conducted, and further evidences are expected to be created. Expert opinion: Immune checkpoint inhibitors that block the PD-1/PD-L1 pathway are essential drugs for advanced or recurrent NSCLC, among which pembrolizumab becomes one of the standards of care in the first-line of NSCLC. For further improvement in efficacy of pembrolizumab, it is necessary to clarify the identification of biomarkers exclusive to PD-L1 expression, predictive factors for patients who benefit most from the agent.
Collapse
Affiliation(s)
- Kiichiro Ninomiya
- a Department of Hematology, Oncology and Respiratory Medicine , Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences , Okayama , Japan
| | - Katsuyuki Hotta
- b Center of Innovative Clinical Medicine , Okayama University Hospital , Okayama , Japan
| |
Collapse
|
33
|
Steele KE, Tan TH, Korn R, Dacosta K, Brown C, Kuziora M, Zimmermann J, Laffin B, Widmaier M, Rognoni L, Cardenes R, Schneider K, Boutrin A, Martin P, Zha J, Wiestler T. Measuring multiple parameters of CD8+ tumor-infiltrating lymphocytes in human cancers by image analysis. J Immunother Cancer 2018; 6:20. [PMID: 29510739 PMCID: PMC5839005 DOI: 10.1186/s40425-018-0326-x] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 02/14/2018] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Immuno-oncology and cancer immunotherapies are areas of intense research. The numbers and locations of CD8+ tumor-infiltrating lymphocytes (TILs) are important measures of the immune response to cancer with prognostic, pharmacodynamic, and predictive potential. We describe the development, validation, and application of advanced image analysis methods to characterize multiple immunohistochemistry-derived CD8 parameters in clinical and nonclinical tumor tissues. METHODS Commercial resection tumors from nine cancer types, and paired screening/on-drug biopsies of non-small-cell lung carcinoma (NSCLC) patients enrolled in a phase 1/2 clinical trial investigating the PD-L1 antibody therapy durvalumab (NCT01693562), were immunostained for CD8. Additional NCT01693562 samples were immunostained with a CD8/PD-L1 dual immunohistochemistry assay. Whole-slide scanning was performed, tumor regions were annotated by a pathologist, and images were analyzed with customized algorithms using Definiens Developer XD software. Validation of image analysis data used cell-by-cell comparison to pathologist scoring across a range of CD8+ TIL densities of all nine cancers, relying primarily on 95% confidence in having at least moderate agreement regarding Lin concordance correlation coefficient (CCC = 0.88-0.99, CCC_lower = 0.65-0.96). RESULTS We found substantial variability in CD8+ TILs between individual patients and across the nine types of human cancer. Diffuse large B-cell lymphoma had several-fold more CD8+ TILs than some other cancers. TIL densities were significantly higher in the invasive margin versus tumor center for carcinomas of head and neck, kidney and pancreas, and NSCLC; the reverse was true only for prostate cancer. In paired patient biopsies, there were significantly increased CD8+ TILs 6 weeks after onset of durvalumab therapy (mean of 365 cells/mm2 over baseline; P = 0.009), consistent with immune activation. Image analysis accurately enumerated CD8+ TILs in PD-L1+ regions of lung tumors using the dual assay and also measured elongate CD8+ lymphocytes which constituted a fraction of overall TILs. CONCLUSIONS Validated image analysis accurately enumerates CD8+ TILs, permitting comparisons of CD8 parameters among tumor regions, individual patients, and cancer types. It also enables the more complex digital solutions needed to better understand cancer immunity, like analysis of multiplex immunohistochemistry and spatial evaluation of the various components comprising the tumor microenvironment. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT01693562 . Study code: CD-ON-MEDI4736-1108. Interventional study (ongoing but not currently recruiting). Actual study start date: August 29, 2012. Primary completion date: June 23, 2017 (final data collection date for primary outcome measure).
Collapse
Affiliation(s)
- Keith E Steele
- MedImmune, One MedImmune Way, Gaithersburg, MD, 20878, USA.
| | - Tze Heng Tan
- Professional Services, Definiens AG, Bernhard-Wicki-Strasse 5, 80636, Munich, Germany
| | - René Korn
- Professional Services, Definiens AG, Bernhard-Wicki-Strasse 5, 80636, Munich, Germany
| | - Karma Dacosta
- MedImmune, One MedImmune Way, Gaithersburg, MD, 20878, USA
| | - Charles Brown
- MedImmune, One MedImmune Way, Gaithersburg, MD, 20878, USA
| | | | - Johannes Zimmermann
- Professional Services, Definiens AG, Bernhard-Wicki-Strasse 5, 80636, Munich, Germany
| | - Brian Laffin
- Professional Services, Definiens AG, Bernhard-Wicki-Strasse 5, 80636, Munich, Germany
- Present address: Brian Laffin-BMS US Medical Oncology, 3401 Princeton Pike, Lawrence Township, NJ, 08648, USA
| | - Moritz Widmaier
- Professional Services, Definiens AG, Bernhard-Wicki-Strasse 5, 80636, Munich, Germany
| | - Lorenz Rognoni
- Professional Services, Definiens AG, Bernhard-Wicki-Strasse 5, 80636, Munich, Germany
| | - Ruben Cardenes
- Professional Services, Definiens AG, Bernhard-Wicki-Strasse 5, 80636, Munich, Germany
| | - Katrin Schneider
- Professional Services, Definiens AG, Bernhard-Wicki-Strasse 5, 80636, Munich, Germany
| | | | - Philip Martin
- MedImmune, One MedImmune Way, Gaithersburg, MD, 20878, USA
| | - Jiping Zha
- MedImmune, One MedImmune Way, Gaithersburg, MD, 20878, USA
- Present address: Jiping Zha - NGM Biopharmaceuticals, 333 Oyster Point Boulevard, South San Francisco, CA, 94080, USA
| | - Tobias Wiestler
- Professional Services, Definiens AG, Bernhard-Wicki-Strasse 5, 80636, Munich, Germany
| |
Collapse
|
34
|
Rowshanravan B, Halliday N, Sansom DM. CTLA-4: a moving target in immunotherapy. Blood 2018; 131:58-67. [PMID: 29118008 PMCID: PMC6317697 DOI: 10.1182/blood-2017-06-741033] [Citation(s) in RCA: 816] [Impact Index Per Article: 116.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 07/31/2017] [Indexed: 02/08/2023] Open
Abstract
CD28 and CTLA-4 are members of a family of immunoglobulin-related receptors that are responsible for various aspects of T-cell immune regulation. The family includes CD28, CTLA-4, and ICOS as well as other proteins, including PD-1, BTLA, and TIGIT. These receptors have both stimulatory (CD28, ICOS) and inhibitory roles (CTLA-4, PD-1, BTLA, and TIGIT) in T-cell function. Increasingly, these pathways are targeted as part of immune modulatory strategies to treat cancers, referred to generically as immune checkpoint blockade, and conversely to treat autoimmunity and CTLA-4 deficiency. Here, we focus on the biology of the CD28/CTLA-4 pathway as a framework for understanding the impacts of therapeutic manipulation of this pathway.
Collapse
Affiliation(s)
- Behzad Rowshanravan
- Institute of Immunity and Transplantation, Division of Infection & Immunity, University College London, Royal Free Hospital, London, United Kingdom
| | - Neil Halliday
- Institute of Immunity and Transplantation, Division of Infection & Immunity, University College London, Royal Free Hospital, London, United Kingdom
| | - David M Sansom
- Institute of Immunity and Transplantation, Division of Infection & Immunity, University College London, Royal Free Hospital, London, United Kingdom
| |
Collapse
|
35
|
Krueger J, Rudd CE. Two Strings in One Bow: PD-1 Negatively Regulates via Co-receptor CD28 on T Cells. Immunity 2017; 46:529-531. [PMID: 28423334 DOI: 10.1016/j.immuni.2017.04.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The identity of PD-1 dependency on other receptors and signaling has been unclear. In a recent issue of Science, Hui et al. (2017) and Kamphorst et al. (2017) now show that CD28 expression is a target of PD-1-associated phosphatases and is needed for T cell expansion in anti-PD-1 immunotherapy.
Collapse
Affiliation(s)
- Janna Krueger
- Division of Immunology-Oncology, Hospital Maisonneuve-Rosemont Research Center, Montreal, QC H1T 2M4, Canada
| | - Christopher E Rudd
- Division of Immunology-Oncology, Hospital Maisonneuve-Rosemont Research Center, Montreal, QC H1T 2M4, Canada; Department of Medicine, Université de Montréal, Montreal, QC H3C 3J7, Canada.
| |
Collapse
|
36
|
Ghatalia P, Zibelman M, Geynisman DM, Plimack ER. Checkpoint Inhibitors for the Treatment of Renal Cell Carcinoma. Curr Treat Options Oncol 2017; 18:7. [PMID: 28210995 DOI: 10.1007/s11864-017-0458-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OPINION STATEMENT The advent of checkpoint inhibitors has revolutionized systemic therapy for many malignancies, including renal cell carcinoma (RCC) where multiple PD-1, PD-L1, and CTLA-4 inhibitors have demonstrated responses and improved survival for patients in clinical trials. Durable benefit with manageable toxicity can be achieved with these agents-but unfortunately for only a minority of individuals. Efforts are ongoing to understand mechanisms driving the response and resistance to checkpoint inhibitors in order to personalize therapy and extend benefit to more patients. In particular, combination immunotherapy is an area of active study with multiple ongoing trials in RCC. Novel immunotherapeutic agents are being explored as well. Clinically, there are nuances related to the use of immunotherapy that are important to understand in order to provide optimal care to patients. Potential autoimmune toxicities are important to identify early so they can be best mitigated with immunosuppression, and careful review of imaging with clinical correlation is important to ensure responding patients are not taken off treatment prematurely due to "pseudo-progression." Lastly, although immunotherapy is an important new tool, it exists among other active agents in the treatment of RCC, and further study is needed to understand where it best fits in the treatment paradigm. In this article, we review the most recent data for immune checkpoint inhibitors in metastatic renal cell carcinoma and more broadly discuss the rapidly evolving landscape of immunotherapy in RCC, including combination immunotherapies.
Collapse
Affiliation(s)
- Pooja Ghatalia
- Department of Hematology/Oncology, Fox Chase Cancer Center, Temple Health, 333 Cottman Avenue, Philadelphia, PA, 19111-2497, USA
| | - Matthew Zibelman
- Department of Hematology/Oncology, Fox Chase Cancer Center, Temple Health, 333 Cottman Avenue, Philadelphia, PA, 19111-2497, USA
| | - Daniel M Geynisman
- Department of Hematology/Oncology, Fox Chase Cancer Center, Temple Health, 333 Cottman Avenue, Philadelphia, PA, 19111-2497, USA
| | - Elizabeth R Plimack
- Department of Hematology/Oncology, Fox Chase Cancer Center, Temple Health, 333 Cottman Avenue, Philadelphia, PA, 19111-2497, USA.
| |
Collapse
|
37
|
Multiscale Modeling of Complex Formation and CD80 Depletion during Immune Synapse Development. Biophys J 2017; 112:997-1009. [PMID: 28297658 DOI: 10.1016/j.bpj.2016.12.052] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 12/22/2016] [Accepted: 12/27/2016] [Indexed: 11/24/2022] Open
Abstract
The mechanisms that discriminate self- and foreign antigen before T cell activation are unresolved. As part of the immune system's adaptive response to specific infections or neoplasms, antigen-presenting cells (APC) and effector T cells form transcellular molecular complexes. CTLA4 expression on regulatory or effector T cells reduces T cell activation. The CTLA4 transendocytosis hypothesis proposes that CTLA4 depletes CD80 and CD86 proteins from the APC membrane, rendering the APC incapable of activating T cells. We developed a multiscale spatiotemporal model for the interaction of a T cell and APC. Formation of the immune complex between T cell and APC starts with formation of the transmembrane complexes between the major histocompatibility complex and the T cell receptor (Signal 1) and between CD80 or CD86 and CD28 (Signal 2) at the opposing membrane surfaces of the interacting cells. By 0.01 s after contact simulation, an increasing concentration gradient of the free membrane proteins develops between the opposing surfaces and spherical parts of each cell's membrane, reaching a maximum at ∼30 s. Over several hours, diffusion across the gradient equalizes the free protein concentrations. During this phase, CTLA4 surface expression and its complexation with CD80/CD86 cause internalization and degradation of CD80/CD86. The simulation results show reasonable agreement with reported experimental data and indicate that key molecular processes take place over a very broad timescale, covering five orders of magnitude. Besides the fast complexation reactions, diffusion-limited processes, especially lateral diffusion in cell membranes and geometrical constraints, considerably slow down evolution of the synapse. Our results are consistent with the CTLA4 transendocytosis hypothesis and suggest the importance of lateral diffusion of surface proteins in contributing to a gradual increase in Signal 1 and Signal 2.
Collapse
|
38
|
Crepeau RL, Ford ML. Challenges and opportunities in targeting the CD28/CTLA-4 pathway in transplantation and autoimmunity. Expert Opin Biol Ther 2017; 17:1001-1012. [PMID: 28525959 DOI: 10.1080/14712598.2017.1333595] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION T cell activation is a complex process that requires multiple cell signaling pathways, including a primary recognition signal and additional costimulatory signals. One of the best-characterized costimulatory pathways includes the Ig superfamily members CD28 and CTLA-4 and their ligands CD80 and CD86. Areas covered: This review discusses past, current and future biological therapies that have been utilized to block the CD28/CTLA-4 cosignaling pathway in the settings of autoimmunity and transplantation, as well the challenges facing successful implementation of these therapies. Expert opinion: The development of CD28 blockers Abatacept and Belatacept provided a more targeted therapy approach for transplant rejection and autoimmune disease relative to calcineurin inhibitors and anti-proliferatives, but overall efficacy may be limited due to their collateral effect of simultaneously blocking CTLA-4 coinhibitory signals. As such, current investigations into the potential of selective CD28 blockade to block the costimulatory potential of CD28 while exploiting the coinhibitory effects of CTLA-4 are promising. However, as selective CD28 blockade inhibits the activity of both effector and regulatory T cells, an important goal for the future is the design of therapies that will maximize the attenuation of effector responses while preserving the suppressive function of T regulatory cells.
Collapse
Affiliation(s)
- Rebecca L Crepeau
- a Emory Transplant Center and Department of Surgery , Emory University , Atlanta , GA , USA
| | - Mandy L Ford
- a Emory Transplant Center and Department of Surgery , Emory University , Atlanta , GA , USA
| |
Collapse
|
39
|
Hui E, Cheung J, Zhu J, Su X, Taylor MJ, Wallweber HA, Sasmal DK, Huang J, Kim JM, Mellman I, Vale RD. T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science 2017; 355:1428-1433. [PMID: 28280247 DOI: 10.1126/science.aaf1292] [Citation(s) in RCA: 1197] [Impact Index Per Article: 149.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 11/09/2016] [Accepted: 02/17/2017] [Indexed: 12/16/2022]
Abstract
Programmed cell death-1 (PD-1) is a coinhibitory receptor that suppresses T cell activation and is an important cancer immunotherapy target. Upon activation by its ligand PD-L1, PD-1 is thought to suppress signaling through the T cell receptor (TCR). By titrating PD-1 signaling in a biochemical reconstitution system, we demonstrate that the co-receptor CD28 is strongly preferred over the TCR as a target for dephosphorylation by PD-1-recruited Shp2 phosphatase. We also show that CD28, but not the TCR, is preferentially dephosphorylated in response to PD-1 activation by PD-L1 in an intact cell system. These results reveal that PD-1 suppresses T cell function primarily by inactivating CD28 signaling, suggesting that costimulatory pathways play key roles in regulating effector T cell function and responses to anti-PD-L1/PD-1 therapy.
Collapse
Affiliation(s)
- Enfu Hui
- Department of Cellular and Molecular Pharmacology and the Howard Hughes Medical Institute, University of California, San Francisco, CA 94158, USA
| | - Jeanne Cheung
- Department of Cancer Immunology, Genentech, South San Francisco, CA 94080, USA
| | - Jing Zhu
- Department of Cancer Immunology, Genentech, South San Francisco, CA 94080, USA
| | - Xiaolei Su
- Department of Cellular and Molecular Pharmacology and the Howard Hughes Medical Institute, University of California, San Francisco, CA 94158, USA
| | - Marcus J Taylor
- Department of Cellular and Molecular Pharmacology and the Howard Hughes Medical Institute, University of California, San Francisco, CA 94158, USA
| | - Heidi A Wallweber
- Department of Cancer Immunology, Genentech, South San Francisco, CA 94080, USA
| | - Dibyendu K Sasmal
- Institute for Molecular Engineering, University of Chicago, IL 60637, USA
| | - Jun Huang
- Institute for Molecular Engineering, University of Chicago, IL 60637, USA
| | - Jeong M Kim
- Department of Cancer Immunology, Genentech, South San Francisco, CA 94080, USA
| | - Ira Mellman
- Department of Cancer Immunology, Genentech, South San Francisco, CA 94080, USA.
| | - Ronald D Vale
- Department of Cellular and Molecular Pharmacology and the Howard Hughes Medical Institute, University of California, San Francisco, CA 94158, USA.
| |
Collapse
|
40
|
Thind K, Padrnos LJ, Ramanathan RK, Borad MJ. Immunotherapy in pancreatic cancer treatment: a new frontier. Therap Adv Gastroenterol 2017; 10:168-194. [PMID: 28286568 PMCID: PMC5330603 DOI: 10.1177/1756283x16667909] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Pancreatic cancer is a highly aggressive and lethal cancer characterized by high invasiveness, local and extensive dissemination at time of diagnosis and resistance to treatment. Few therapies have shown efficacy in the past and even standard of care therapies yield only modest improvements in the mortality of patients with advanced or metastatic disease. Efforts have been undertaken to study the pancreatic tumor microenvironment and have established its complex and immunosuppressive nature which could explain the high resistance to chemotherapy. Novel therapies targeting the tumor microenvironment with an aim to decrease this resistance, improve immune tolerance and increase the efficacy of the current treatment have shown some promising preliminary results in preclinical and clinical trials. We review the current advances in the field of immunotherapy and their effectiveness as a potential treatment strategy in the pancreatic cancer.
Collapse
Affiliation(s)
- Komal Thind
- Department of Internal Medicine, Cleveland Clinic Akron General, Akron, OH, USA
| | - Leslie J. Padrnos
- Division of Hematology/Oncology, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | | | - Mitesh J. Borad
- Division of Hematology/Oncology, Mayo Clinic Arizona, 5777 E. Mayo Boulevard, Phoenix, AZ 85054, USA
| |
Collapse
|
41
|
Tanhapour M, Vaisi-Raygani A, Khazaei M, Rahimi Z, Pourmotabbed T. Cytotoxic T-lymphocyte Associated Antigen-4 (CTLA-4) Polymorphism, Cancer, and Autoimmune Diseases. AIMS MEDICAL SCIENCE 2017. [DOI: 10.3934/medsci.2017.4.395] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
|
42
|
Carlo MI, Voss MH, Motzer RJ. Checkpoint inhibitors and other novel immunotherapies for advanced renal cell carcinoma. Nat Rev Urol 2016; 13:420-31. [PMID: 27324121 PMCID: PMC5532875 DOI: 10.1038/nrurol.2016.103] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The management of advanced renal cell carcinoma (RCC) has dramatically changed over the past decade. Therapies that target the vascular endothelial growth factor (VEGF) and mammalian target of rapamycin (mTOR) pathways have considerably expanded treatment options; however, most patients with advanced RCC still have limited overall survival. Increased understanding of the mechanisms of T cell-antigen recognition and function has led to the development of novel immunotherapies to treat cancer, chief among them inhibitors of checkpoint receptors - molecules whose function is to restrain the host immune response. In 2015, the FDA approved the first checkpoint inhibitor nivolumab for patients with advanced RCC following treatment with antiangiogenic therapy based on improved overall survival compared with the standard of care. Ongoing phase III trials are comparing checkpoint-inhibitor-based combination regimens with antiangiogenesis agents in the first-line setting. The field is evolving rapidly, with many clinical trials already testing several checkpoint inhibitors alone, in combination, or with other targeted therapies. In addition, different novel immune therapies are being investigated including vaccines, T-cell agonists, and chimeric antigen receptor T cells. Determining which patients will benefit from these therapies and which combination approaches will result in better response will be important as this field evolves.
Collapse
Affiliation(s)
- Maria I Carlo
- Memorial Sloan Kettering Cancer Center 1275 York Avenue New York, New York 10065, USA
| | - Martin H Voss
- Memorial Sloan Kettering Cancer Center 1275 York Avenue New York, New York 10065, USA
| | - Robert J Motzer
- Memorial Sloan Kettering Cancer Center 1275 York Avenue New York, New York 10065, USA
| |
Collapse
|
43
|
Ting WH, Chien MN, Lo FS, Wang CH, Huang CY, Lin CL, Lin WS, Chang TY, Yang HW, Chen WF, Lien YP, Cheng BW, Lin CH, Chen CC, Wu YL, Hung CM, Li HJ, Chan CI, Lee YJ. Association of Cytotoxic T-Lymphocyte-Associated Protein 4 (CTLA4) Gene Polymorphisms with Autoimmune Thyroid Disease in Children and Adults: Case-Control Study. PLoS One 2016; 11:e0154394. [PMID: 27111218 PMCID: PMC4844099 DOI: 10.1371/journal.pone.0154394] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Accepted: 04/11/2016] [Indexed: 11/18/2022] Open
Abstract
Autoimmune thyroid disease (AITD), including Graves disease (GD) and Hashimoto disease (HD), is an organ-specific autoimmune disease with a strong genetic component. Although the cytotoxic T-lymphocyte-associated protein 4 (CTLA4) polymorphism has been reported to be associated with AITD in adults, few studies have focused on children. The aim of our study was to investigate whether the CTLA4 polymorphisms, including -318C/T (rs5742909), +49A/G (rs231775), and CT60 (rs3087243), were associated with GD and HD in Han Chinese adults and children. We studied 289 adult GD, 265 pediatric GD, 229 pediatric HD patients, and 1058 healthy controls and then compared genotype, allele, carrier, and haplotype frequencies between patients and controls. We found that CTLA4 SNPs +49A/G and CT60 were associated with GD in adults and children. Allele G of +49A/G was significantly associated with GD in adults (odds ratio [OR], 1.50; 95% confidence interval [CI], 1.21–1.84; corrected P value [Pc] < 0.001) and children (OR, 1.42; 95% CI, 1.15–1.77; Pc = 0.002). Allele G of CT60 also significantly increased risk of GD in adults (OR, 1.63; 95% CI, 1.27–2.09; Pc < 0.001) and GD in children (OR, 1.58; 95% CI, 1.22–2.04; Pc < 0.001). Significant linkage disequilibrium was found between +49A/G and CT60 in GD and control subjects (D’ = 0.92). Our results showed that CTLA4 was associated with both GD and HD and played an equivalent role in both adult and pediatric GD in Han Chinese population.
Collapse
Affiliation(s)
- Wei-Hsin Ting
- Department of Pediatrics, MacKay Children’s Hospital, Taipei, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
- MacKay Junior College of Medicine, Nursing and Management, New Taipei City, Taiwan
| | - Ming-Nan Chien
- Department of Endocrinology and Metabolism, MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
- Institute of Mechatronic Engineering, National Taipei University of Technology, Taipei, Taiwan
- MacKay Junior College of Medicine, Nursing and Management, New Taipei City, Taiwan
| | - Fu-Sung Lo
- Department of Pediatrics, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan
- College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Chao-Hung Wang
- Department of Endocrinology and Metabolism, MacKay Memorial Hospital, Taipei, Taiwan
| | - Chi-Yu Huang
- Department of Pediatrics, MacKay Children’s Hospital, Taipei, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
| | - Chiung-Ling Lin
- Department of Medical Research, MacKay Memorial Hospital Tamsui, New Taipei City, Taiwan
| | - Wen-Shan Lin
- Department of Medical Research, MacKay Memorial Hospital Tamsui, New Taipei City, Taiwan
| | - Tzu-Yang Chang
- Department of Medical Research, MacKay Memorial Hospital Tamsui, New Taipei City, Taiwan
| | - Horng-Woei Yang
- Department of Medical Research, MacKay Memorial Hospital Tamsui, New Taipei City, Taiwan
| | - Wei-Fang Chen
- Department of Medical Research, MacKay Memorial Hospital Tamsui, New Taipei City, Taiwan
| | - Ya-Ping Lien
- Department of Medical Research, MacKay Memorial Hospital Tamsui, New Taipei City, Taiwan
| | - Bi-Wen Cheng
- Department of Pediatrics, MacKay Memorial Hospital HsinChu, Hsin-Chu, Taiwan
| | - Chao-Hsu Lin
- Department of Pediatrics, MacKay Memorial Hospital HsinChu, Hsin-Chu, Taiwan
| | - Chia-Ching Chen
- Department of Pediatrics, Chiayi Christian Hospital, Chia-Yi, Taiwan
| | - Yi-Lei Wu
- Department of Pediatrics, Changhua Christian Hospital, Chang-Hua, Taiwan
| | - Chen-Mei Hung
- Department of Pediatrics, Hsinchu Cathay General Hospital, Hsin-Chu, Taiwan
| | - Hsin-Jung Li
- Department of Pediatrics, St. Martin De Porres Hospital, Chia-Yi, Taiwan
| | - Chon-In Chan
- Department of Pediatrics, MacKay Children’s Hospital, Taipei, Taiwan
| | - Yann-Jinn Lee
- Department of Pediatrics, MacKay Children’s Hospital, Taipei, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
- Department of Medical Research, MacKay Memorial Hospital Tamsui, New Taipei City, Taiwan
- Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Institute of Biomedical Sciences, MacKay Medical College, New Taipei City, Taiwan
- * E-mail:
| |
Collapse
|
44
|
Sckisel GD, Mirsoian A, Bouchlaka MN, Tietze JK, Chen M, Blazar BR, Murphy WJ. Late administration of murine CTLA-4 blockade prolongs CD8-mediated anti-tumor effects following stimulatory cancer immunotherapy. Cancer Immunol Immunother 2015; 64:1541-52. [PMID: 26423422 DOI: 10.1007/s00262-015-1759-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 09/14/2015] [Indexed: 12/19/2022]
Abstract
We have demonstrated that immunostimulatory therapies such as interleukin-2 (IL-2) and anti-CD40 (αCD40) can be combined to deliver synergistic anti-tumor effects. While this strategy has shown success, efficacy varies depending on a number of factors including tumor type and severe toxicities can be seen. We sought to determine whether blockade of negative regulators such as cytotoxic T lymphocyte antigen-4 (CTLA-4) could simultaneously prolong CD8(+) T cell responses and augment T cell anti-tumor effects. We devised a regimen in which anti-CTLA-4 was administered late so as to delay contraction and minimize toxicities. This late administration both enhanced and prolonged CD8 T cell activation without the need for additional IL-2. The quality of the T cell response was improved with increased frequency of effector/effector memory phenotype cells along with improved lytic ability and bystander expansion. This enhanced CD8 response translated to improved anti-tumor responses both at the primary and metastatic sites. Importantly, toxicities were not exacerbated with combination. This study provides a platform for rational design of immunotherapy combinations to maximize anti-tumor immunity while minimizing toxicities.
Collapse
Affiliation(s)
- Gail D Sckisel
- Department of Dermatology, University of California, Davis, School of Medicine, IRC Building Rm 1630, 2921 Stockton Blvd., Sacramento, CA, 95817, USA
| | - Annie Mirsoian
- Department of Dermatology, University of California, Davis, School of Medicine, IRC Building Rm 1630, 2921 Stockton Blvd., Sacramento, CA, 95817, USA
| | - Myriam N Bouchlaka
- Department of Dermatology, University of California, Davis, School of Medicine, IRC Building Rm 1630, 2921 Stockton Blvd., Sacramento, CA, 95817, USA
| | - Julia K Tietze
- Department of Dermatology, University of California, Davis, School of Medicine, IRC Building Rm 1630, 2921 Stockton Blvd., Sacramento, CA, 95817, USA
| | - Mingyi Chen
- Department of Pathology, University of California, School of Medicine, Sacramento, CA, USA
| | - Bruce R Blazar
- Department of Pediatrics, Division of Bone Marrow Transplantation, University of Minnesota Cancer Center, Minneapolis, MN, USA
| | - William J Murphy
- Department of Dermatology, University of California, Davis, School of Medicine, IRC Building Rm 1630, 2921 Stockton Blvd., Sacramento, CA, 95817, USA. .,Department of Internal Medicine, University of California, Davis, School of Medicine, Sacramento, CA, USA.
| |
Collapse
|
45
|
Jeffery LE, Qureshi OS, Gardner D, Hou TZ, Briggs Z, Soskic B, Baker J, Raza K, Sansom DM. Vitamin D Antagonises the Suppressive Effect of Inflammatory Cytokines on CTLA-4 Expression and Regulatory Function. PLoS One 2015; 10:e0131539. [PMID: 26134669 PMCID: PMC4489761 DOI: 10.1371/journal.pone.0131539] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 06/03/2015] [Indexed: 12/12/2022] Open
Abstract
The immune suppressive protein CTLA-4 is constitutively expressed by Tregs and induced in effector T cells upon activation. Its crucial role in adaptive immunity is apparent from the fatal autoimmune pathology seen in CTLA-4 knockout mice. However, little is known regarding factors that regulate CTLA-4 expression and their effect upon its function to remove CD80 and CD86 from antigen presenting cells by transendocytosis. Th17 cells are emerging as significant players in autoimmunity as well as other diseases. Therefore, in this study we have examined the effects of Th17 polarising conditions on CTLA-4 expression and function in human T cells and show that Th17 conditions can suppress the expression of CTLA-4 and its transendocytic function. In contrast to Th17 cells, vitamin D is inversely associated with autoimmune disease. We have previously shown a striking ability of 1,25 dihydroxyvitamin D3 (1,25(OH)2D3) to enhance CTLA-4, however, its effects upon B7 transendocytosis and its activity in the context of inflammation remained unknown. Here we show that induction of CTLA-4 by 1,25(OH)2D3 can actually be enhanced in the presence of Th17 polarising cytokines. Furthermore, its transendocytic function was maintained such that T cells generated in the presence of Th17 conditions and 1,25(OH)2D3 were highly effective at capturing CTLA-4 ligands from antigen presenting cells and suppressing T cell division. Taken together, these data reveal an inhibitory effect of Th17 polarising conditions upon CTLA-4-mediated regulation and show that 1,25(OH)2D3 counteracts this effect. Given the importance of CTLA-4-mediated suppression in the control of autoimmune diseases, our novel data highlight the importance of vitamin D in inflammatory settings.
Collapse
Affiliation(s)
- Louisa E. Jeffery
- Medical Research Council Centre for Immune Regulation, School of Immunity and Infection, Institute of Biomedical Research, University of Birmingham Medical School, Birmingham, United Kingdom
| | - Omar S. Qureshi
- Medical Research Council Centre for Immune Regulation, School of Immunity and Infection, Institute of Biomedical Research, University of Birmingham Medical School, Birmingham, United Kingdom
| | - David Gardner
- Medical Research Council Centre for Immune Regulation, School of Immunity and Infection, Institute of Biomedical Research, University of Birmingham Medical School, Birmingham, United Kingdom
| | - Tie Z. Hou
- UCL Institute of Immunity and Transplantation, Royal Free Campus, University College London, London, United Kingdom
| | - Zoe Briggs
- Medical Research Council Centre for Immune Regulation, School of Immunity and Infection, Institute of Biomedical Research, University of Birmingham Medical School, Birmingham, United Kingdom
| | - Blagoje Soskic
- UCL Institute of Immunity and Transplantation, Royal Free Campus, University College London, London, United Kingdom
| | - Jennifer Baker
- Medical Research Council Centre for Immune Regulation, School of Immunity and Infection, Institute of Biomedical Research, University of Birmingham Medical School, Birmingham, United Kingdom
| | - Karim Raza
- Medical Research Council Centre for Immune Regulation, School of Immunity and Infection, Institute of Biomedical Research, University of Birmingham Medical School, Birmingham, United Kingdom
- Department of Rheumatology, Sandwell and West Birmingham Hospitals NHS Trust, Birmingham, United Kingdom
- * E-mail:
| | - David M. Sansom
- UCL Institute of Immunity and Transplantation, Royal Free Campus, University College London, London, United Kingdom
| |
Collapse
|
46
|
Chan SL, Chan AWH, Yeo W. Novel therapeutic targets and predictive markers for hepatocellular carcinoma. Expert Opin Ther Targets 2015; 19:973-983. [PMID: 25910512 DOI: 10.1517/14728222.2015.1031109] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Following the approval of sorafenib as the first systemic agent for treatment of advanced hepatocellular carcinoma (HCC), there have been an increasing number of targeted treatments under testing for the cancer. However, most of the recently published drug trials in HCC failed to produce remarkable results. The researchers are actively pursuing novel therapeutic targets as well as predictive biomarker for treatment of HCC. AREAS COVERED This review discusses a number of potential novel targets for drug development of HCC. Focus is put on the underlying rationale for therapeutic development of the target and the possibility of using a predictive biomarker to select patients for drug testing. EXPERT OPINION Future direction of drug development will be discussed. Notably, a clinical trial on drug testing in HCC should be shifted from all-comers approach to selected populations based on underlying viral etiology and molecular targets. A study to evaluate predictive biomarker is crucial to the development of targeted agents for HCC. Design of clinical trials on HCC should introduce measures to encourage acquisition of tumor and plasma samples for biomarker development.
Collapse
Affiliation(s)
- Stephen L Chan
- The Chinese University of Hong Kong, Hong Kong Cancer Institute and Prince of Wales Hospital, State Key Laboratory of Oncology in South China, Sir YK Pao Center for Cancer, Department of Clinical Oncology , Hong Kong , China +852 2632 2118 ; +852 2648 7097 ;
| | | | | |
Collapse
|
47
|
Abstract
The fate of T and B lymphocytes, the key cells that direct the adaptive immune response, is regulated by a diverse network of signal transduction pathways. The T- and B-cell antigen receptors are coupled to intracellular tyrosine kinases and adaptor molecules to control the metabolism of inositol phospholipids and calcium release. The production of inositol polyphosphates and lipid second messengers directs the activity of downstream guanine-nucleotide-binding proteins and protein and lipid kinases/phosphatases that control lymphocyte transcriptional and metabolic programs. Lymphocyte activation is modulated by costimulatory molecules and cytokines that elicit intracellular signaling that is integrated with the antigen-receptor-controlled pathways.
Collapse
Affiliation(s)
- Doreen Cantrell
- College of Life Sciences, Wellcome Trust Biocentre, University of Dundee, Dundee DD1 5EH, Scotland, United Kingdom
| |
Collapse
|
48
|
Beyersdorf N, Kerkau T, Hünig T. CD28 co-stimulation in T-cell homeostasis: a recent perspective. Immunotargets Ther 2015; 4:111-22. [PMID: 27471717 PMCID: PMC4918251 DOI: 10.2147/itt.s61647] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
T-cells play a key role within the adaptive immune system mediating cellular immunity and orchestrating the immune response as a whole. Their activation requires not only recognition of antigen/major histocompatibility complexes by the T-cell receptor but in addition co-stimulation via the CD28 molecule through binding to CD80, CD86, or as recently discovered, inducible co-stimulator ligand expressed by antigen-presenting cells. Apart from tight control of the co-stimulatory signal by the T-cell receptor complex, expression of the inhibitory receptor cytotoxic T-lymphocyte antigen-4 (CTLA-4) sharing its ligands with CD28 is required to avoid inappropriate or prolonged T-cell activation. CD4(+) Foxp3(+) regulatory T (Treg) cells, which are crucial inhibitors of autoimmunity, add another level of complexity in that they differ from conventional non-regulatory CD4(+) T-cells by strongly depending on CD28 signaling for their generation and homeostasis. Moreover, CTLA-4 is constitutively expressed by Treg cells where it serves as a key mediator of suppression, while conventional CD4(+) T-cells express CTLA-4 only after activation. Here, we discuss recent insights into the molecular events underlying CD28-mediated co-stimulation, its impact on gene regulation, and the differential role of CD28 expression on Treg cells versus conventional CD4(+) and CD8(+) T-cells. Moreover, we summarize the exciting therapeutic options which have arisen from our current understanding of T-cell co-stimulation. Some of these have already been translated into the clinic, while others are expected to follow soon due to promising preclinical results. In particular, we discuss the failed 2006 trial of the CD28 superagonist TGN1412, and the return of this potent T-cell activator to clinical development.
Collapse
Affiliation(s)
- Niklas Beyersdorf
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Thomas Kerkau
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Thomas Hünig
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
49
|
Rahimian S, Fransen MF, Kleinovink JW, Amidi M, Ossendorp F, Hennink WE. Polymeric microparticles for sustained and local delivery of antiCD40 and antiCTLA-4 in immunotherapy of cancer. Biomaterials 2015; 61:33-40. [PMID: 25993015 DOI: 10.1016/j.biomaterials.2015.04.043] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 04/30/2015] [Indexed: 01/24/2023]
Abstract
This study investigated the feasibility of the use of polymeric microparticles for sustained and local delivery of immunomodulatory antibodies in immunotherapy of cancer. Local delivery of potent immunomodulatory antibodies avoids unwanted systemic side effects while retaining their anti-tumor effects. Microparticles based on poly(lactic-co-hydroxymethyl-glycolic acid) (pLHMGA) and loaded with two distinct types of immunomodulatory antibodies (CTLA-4 antibody blocking inhibitory receptors on T cells or CD40 agonistic antibody stimulating dendritic cells) were prepared by double emulsion solvent evaporation technique. The obtained particles had a diameter of 12-15 μm to avoid engulfment by phagocytes and were slightly porous as shown by SEM analysis. The loading efficiency of the antibodies in the microparticles was >85%. The in vitro release profile of antiCD40 and antiCTLA-4 from microparticles showed a burst release of about 20% followed by a sustained release of the content up to 80% of the loading in around 30 days. The therapeutic efficacy of the microparticulate formulations was studied in colon carcinoma tumor model (MC-38). Mice bearing subcutaneous MC-38 tumors were treated with the same dose of immunomodulatory antibodies formulated either in incomplete Freund's adjuvant (IFA) or in microparticles. The antibody-loaded microparticles showed comparable therapeutic efficacy to the IFA formulation with no local adverse effects. The biodegradable microparticles were fully resorbed in vivo and no remnants of inflammatory depots as observed with IFA were present in the cured mice. Moreover the microparticles exhibited lower antibody serum levels in comparison with IFA formulations which lowers the probability of systemic adverse effects. In conclusion, pLHMGA microparticles are excellent delivery systems in providing long-lasting and non-toxic antibody therapy for immunotherapy of cancer.
Collapse
Affiliation(s)
- Sima Rahimian
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Marieke F Fransen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Jan Willem Kleinovink
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Maryam Amidi
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Ferry Ossendorp
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands.
| | - Wim E Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
50
|
Hou TZ, Qureshi OS, Wang CJ, Baker J, Young SP, Walker LSK, Sansom DM. A transendocytosis model of CTLA-4 function predicts its suppressive behavior on regulatory T cells. THE JOURNAL OF IMMUNOLOGY 2015; 194:2148-59. [PMID: 25632005 DOI: 10.4049/jimmunol.1401876] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Manipulation of the CD28/CTLA-4 pathway is at the heart of a number of immunomodulatory approaches used in both autoimmunity and cancer. Although it is clear that CTLA-4 is a critical regulator of T cell responses, the immunological contexts in which CTLA-4 controls immune responses are not well defined. In this study, we show that whereas CD80/CD86-dependent activation of resting human T cells caused extensive T cell proliferation and robust CTLA-4 expression, in this context CTLA-4 blocking Abs had no impact on the response. In contrast, in settings where CTLA-4(+) cells were present as "regulators," inhibition of resting T cell responses was dependent on CTLA-4 expression and specifically related to the number of APC. At low numbers of APC or low levels of ligand, CTLA-4-dependent suppression was highly effective whereas at higher APC numbers or high levels of ligand, inhibition was lost. Accordingly, the degree of suppression correlated with the level of CD86 expression remaining on the APC. These data reveal clear rules for the inhibitory function of CTLA-4 on regulatory T cells, which are predicted by its ability to remove ligands from APC.
Collapse
Affiliation(s)
- Tie Zheng Hou
- Division of Infection and Immunity, Department of Immunology, Institute of Immunity and Transplantation, University College London, Royal Free Hospital, London NW3 2PF, United Kingdom; and
| | - Omar S Qureshi
- School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Chun Jing Wang
- Division of Infection and Immunity, Department of Immunology, Institute of Immunity and Transplantation, University College London, Royal Free Hospital, London NW3 2PF, United Kingdom; and
| | - Jennifer Baker
- School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Stephen P Young
- School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Lucy S K Walker
- Division of Infection and Immunity, Department of Immunology, Institute of Immunity and Transplantation, University College London, Royal Free Hospital, London NW3 2PF, United Kingdom; and
| | - David M Sansom
- Division of Infection and Immunity, Department of Immunology, Institute of Immunity and Transplantation, University College London, Royal Free Hospital, London NW3 2PF, United Kingdom; and
| |
Collapse
|