1
|
Zhao J, Li L, Feng X, Yin H, Fan X, Gao C, Zhao M, Lu Q. Blockade of OX40/OX40L signaling using anti-OX40L alleviates murine lupus nephritis. Eur J Immunol 2024; 54:e2350915. [PMID: 38798163 DOI: 10.1002/eji.202350915] [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: 11/24/2023] [Revised: 04/24/2024] [Accepted: 04/26/2024] [Indexed: 05/29/2024]
Abstract
Genetic variants of the OX40 ligand (OX40L) locus are associated with the risk of systemic lupus erythematosus (SLE), it is unclear how the OX40L blockade delays the lupus phenotype. Therefore, we examined the effects of an anti-OX40L antibody in MRL/Lpr mice. Next, we investigated the effect of anti-OX40L on immunosuppression in keyhole limpet hemocyanin-immunized C57BL/6J mice. In vitro treatment of anti-OX40L in CD4+ T and B220+ B cells was used to explore the role of OX40L in the pathogenesis of SLE. Anti-OX40L alleviated murine lupus nephritis, accompanied by decreased production of anti-dsDNA and proteinuria, as well as lower frequencies of splenic T helper (Th) 1 and T-follicular helper cells (Tfh). In keyhole limpet hemocyanin-immunized mice, decreased levels of immunoglobulins and plasmablasts were observed in the anti-OX40L group. Anti-OX40L reduced the number and area of germinal centers. Compared with the control IgG group, anti-OX40L downregulated CD4+ T-cell differentiation into Th1 and Tfh cells and upregulated CD4+ T-cell differentiation into regulatory T cells in vitro. Furthermore, anti-OX40L inhibited toll-like receptor 7-mediated differentiation of antibody-secreting cells and antibody production through the regulation of the SPIB-BLIMP1-XBP1 axis in B cells. These results suggest that OX40L is a promising therapeutic target for SLE.
Collapse
Affiliation(s)
- Junpeng Zhao
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Liming Li
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Xiwei Feng
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Huiqi Yin
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Xinyu Fan
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Changxing Gao
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Ming Zhao
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Qianjin Lu
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
2
|
Schettini N, Pacetti L, Corazza M, Borghi A. The Role of OX40-OX40L Axis in the Pathogenesis of Atopic Dermatitis. Dermatitis 2024. [PMID: 38700255 DOI: 10.1089/derm.2024.0058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
OX40 is a co-stimulatory immune checkpoint molecule that promotes the activation and the effector function of T lymphocytes through interaction with its ligand (OX40L) on antigen-presenting cells. OX40-OX40L axis plays a crucial role in Th1 and Th2 cell expansion, particularly during the late phases or long-lasting response. Atopic dermatitis is characterized by an immune dysregulation of Th2 activity and by an overproduction of proinflammatory cytokines such as interleukin (IL)-4 and IL-13. Other molecules involved in its pathogenesis include thymic stromal lymphopoietin, IL-33, and IL-25, which contribute to the promotion of OX40L expression on dendritic cells. Lesional skin in atopic dermatitis exhibits a higher level of OX40L+-presenting cells compared with other dermatologic diseases or normal skin. Recent clinical trials using antagonizing anti-OX40 or anti-OX40L antibodies have shown symptom improvement and cutaneous manifestation alleviation in patients with atopic dermatitis. These findings suggest the relevance of the OX40-OX40L axis in atopic dermatitis pathogenesis.
Collapse
Affiliation(s)
- Natale Schettini
- From the Section of Dermatology and Infectious Diseases, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Lucrezia Pacetti
- From the Section of Dermatology and Infectious Diseases, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Monica Corazza
- From the Section of Dermatology and Infectious Diseases, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Alessandro Borghi
- From the Section of Dermatology and Infectious Diseases, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| |
Collapse
|
3
|
Sadrolashrafi K, Guo L, Kikuchi R, Hao A, Yamamoto RK, Tolson HC, Bilimoria SN, Yee DK, Armstrong AW. An OX-Tra'Ordinary Tale: The Role of OX40 and OX40L in Atopic Dermatitis. Cells 2024; 13:587. [PMID: 38607026 PMCID: PMC11011471 DOI: 10.3390/cells13070587] [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: 02/22/2024] [Revised: 03/26/2024] [Accepted: 03/26/2024] [Indexed: 04/13/2024] Open
Abstract
The transmembrane glycoprotein OX40 receptor (OX40) and its ligand, OX40L, are instrumental modulators of the adaptive immune response in humans. OX40 functions as a costimulatory molecule that promotes T cell activation, differentiation, and survival through ligation with OX40L. T cells play an integral role in the pathogenesis of several inflammatory skin conditions, including atopic dermatitis (AD). In particular, T helper 2 (TH2) cells strongly contribute to AD pathogenesis via the production of cytokines associated with type 2 inflammation (e.g., IL-4, IL-5, IL-13, and IL-31) that lead to skin barrier dysfunction and pruritus. The OX40-OX40L interaction also promotes the activation and proliferation of other T helper cell populations (e.g., TH1, TH22, and TH17), and AD patients have demonstrated higher levels of OX40 expression on peripheral blood mononuclear cells than healthy controls. As such, the OX40-OX40L pathway is a potential target for AD treatment. Novel therapies targeting the OX40 pathway are currently in development, several of which have demonstrated promising safety and efficacy results in patients with moderate-to-severe AD. Herein, we review the function of OX40 and the OX40-OX40L signaling pathway, their role in AD pathogenesis, and emerging therapies targeting OX40-OX40L that may offer insights into the future of AD management.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - April W. Armstrong
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
4
|
Rakké YS, Buschow SI, IJzermans JNM, Sprengers D. Engaging stimulatory immune checkpoint interactions in the tumour immune microenvironment of primary liver cancers - how to push the gas after having released the brake. Front Immunol 2024; 15:1357333. [PMID: 38440738 PMCID: PMC10910082 DOI: 10.3389/fimmu.2024.1357333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 01/31/2024] [Indexed: 03/06/2024] Open
Abstract
Hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA) are the first and second most common primary liver cancer (PLC). For decades, systemic therapies consisting of tyrosine kinase inhibitors (TKIs) or chemotherapy have formed the cornerstone of treating advanced-stage HCC and CCA, respectively. More recently, immunotherapy using immune checkpoint inhibition (ICI) has shown anti-tumour reactivity in some patients. The combination regimen of anti-PD-L1 and anti-VEGF antibodies has been approved as new first-line treatment of advanced-stage HCC. Furthermore, gemcibatine plus cisplatin (GEMCIS) with an anti-PD-L1 antibody is awaiting global approval for the treatment of advanced-stage CCA. As effective anti-tumour reactivity using ICI is achieved in a minor subset of both HCC and CCA patients only, alternative immune strategies to sensitise the tumour microenvironment of PLC are waited for. Here we discuss immune checkpoint stimulation (ICS) as additional tool to enhance anti-tumour reactivity. Up-to-date information on the clinical application of ICS in onco-immunology is provided. This review provides a rationale of the application of next-generation ICS either alone or in combination regimen to potentially enhance anti-tumour reactivity in PLC patients.
Collapse
Affiliation(s)
- Yannick S. Rakké
- Department of Surgery, Erasmus MC-Transplant Institute, University Medical Center, Rotterdam, Netherlands
| | - Sonja I. Buschow
- Department of Gastroenterology and Hepatology, Erasmus MC-Cancer Institute-University Medical Center, Rotterdam, Netherlands
| | - Jan N. M. IJzermans
- Department of Surgery, Erasmus MC-Transplant Institute, University Medical Center, Rotterdam, Netherlands
| | - Dave Sprengers
- Department of Gastroenterology and Hepatology, Erasmus MC-Cancer Institute-University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
5
|
Zhou G, Liu H, Yuan Y, Wang Q, Wang L, Wu J. Lentinan progress in inflammatory diseases and tumor diseases. Eur J Med Res 2024; 29:8. [PMID: 38172925 PMCID: PMC10763102 DOI: 10.1186/s40001-023-01585-7] [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: 11/02/2023] [Accepted: 12/10/2023] [Indexed: 01/05/2024] Open
Abstract
Shiitake mushrooms are a fungal food that has been recorded in Chinese medicine to nourish the blood and qi. Lentinan (lLNT) is an active substance extracted from shiitake mushrooms with powerful antioxidant, anti-inflammatory, anti-tumor functions. Inflammatory diseases and cancers are the leading causes of death worldwide, posing a serious threat to human life and health and posing enormous challenges to global health systems. There is still a lack of effective treatments for inflammatory diseases and cancer. LNT has been approved as an adjunct to chemotherapy in China and Japan. Studies have shown that LNT plays an important role in the treatment of inflammatory diseases as well as oncological diseases. Moreover, clinical experiments have confirmed that LNT combined with chemotherapy drugs has a significant effect in improving the prognosis of patients, enhancing their immune function and reducing the side effects of chemotherapy in lung cancer, colorectal cancer and gastric cancer. However, the relevant mechanism of action of the LNT signaling pathway in inflammatory diseases and cancer. Therefore, this article reviews the mechanism and clinical research of LNT in inflammatory diseases and tumor diseases in recent years.
Collapse
Affiliation(s)
- Guangda Zhou
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Jinan, 250062, China
| | - Haiyan Liu
- Department of Ultrasound, The Affiliated Taian City Central Hospital of Qingdao University, Taian, 271000, China
| | - Ying Yuan
- Department of Neurology, Xingtai Third Hospital, Xingtai, 054000, China
| | - Qian Wang
- Department of Central Laboratory, The Affiliated Taian City Central Hospital of Qingdao University, Taian, 271000, China.
| | - Lanping Wang
- Department of Surgery, The Second Affiliated Hospital of Shandong First Medical University, Taian, 271000, China.
| | - Jianghua Wu
- School of Nursing, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, China.
| |
Collapse
|
6
|
Jiang B, Zhang T, Deng M, Jin W, Hong Y, Chen X, Chen X, Wang J, Hou H, Gao Y, Gong W, Wang X, Li H, Zhou X, Feng Y, Zhang B, Jiang B, Lu X, Zhang L, Li Y, Song W, Sun H, Wang Z, Song X, Shen Z, Liu X, Li K, Wang L, Liu Y. BGB-A445, a novel non-ligand-blocking agonistic anti-OX40 antibody, exhibits superior immune activation and antitumor effects in preclinical models. Front Med 2023; 17:1170-1185. [PMID: 37747585 DOI: 10.1007/s11684-023-0996-8] [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: 10/14/2022] [Accepted: 03/06/2023] [Indexed: 09/26/2023]
Abstract
OX40 is a costimulatory receptor that is expressed primarily on activated CD4+, CD8+, and regulatory T cells. The ligation of OX40 to its sole ligand OX40L potentiates T cell expansion, differentiation, and activation and also promotes dendritic cells to mature to enhance their cytokine production. Therefore, the use of agonistic anti-OX40 antibodies for cancer immunotherapy has gained great interest. However, most of the agonistic anti-OX40 antibodies in the clinic are OX40L-competitive and show limited efficacy. Here, we discovered that BGB-A445, a non-ligand-competitive agonistic anti-OX40 antibody currently under clinical investigation, induced optimal T cell activation without impairing dendritic cell function. In addition, BGB-A445 dose-dependently and significantly depleted regulatory T cells in vitro and in vivo via antibody-dependent cellular cytotoxicity. In the MC38 syngeneic model established in humanized OX40 knock-in mice, BGB-A445 demonstrated robust and dose-dependent antitumor efficacy, whereas the ligand-competitive anti-OX40 antibody showed antitumor efficacy characterized by a hook effect. Furthermore, BGB-A445 demonstrated a strong combination antitumor effect with an anti-PD-1 antibody. Taken together, our findings show that BGB-A445, which does not block OX40-OX40L interaction in contrast to clinical-stage anti-OX40 antibodies, shows superior immune-stimulating effects and antitumor efficacy and thus warrants further clinical investigation.
Collapse
Affiliation(s)
- Beibei Jiang
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, 102206, China
| | - Tong Zhang
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, 102206, China
| | - Minjuan Deng
- Department of Discovery Biomarkers, BeiGene (Beijing) Co., Ltd., Beijing, 102206, China
| | - Wei Jin
- Department of Discovery Biomarkers, BeiGene (Beijing) Co., Ltd., Beijing, 102206, China
| | - Yuan Hong
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, 102206, China
| | - Xiaotong Chen
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, 102206, China
| | - Xin Chen
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, 102206, China
| | - Jing Wang
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, 102206, China
| | - Hongjia Hou
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, 102206, China
| | - Yajuan Gao
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, 102206, China
| | - Wenfeng Gong
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, 102206, China
| | - Xing Wang
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, 102206, China
| | - Haiying Li
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, 102206, China
| | - Xiaosui Zhou
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, 102206, China
| | - Yingcai Feng
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, 102206, China
| | - Bo Zhang
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, 102206, China
| | - Bin Jiang
- Department of Discovery Biomarkers, BeiGene (Beijing) Co., Ltd., Beijing, 102206, China
| | - Xueping Lu
- Department of Discovery Biomarkers, BeiGene (Beijing) Co., Ltd., Beijing, 102206, China
| | - Lijie Zhang
- Department of Discovery Biomarkers, BeiGene (Beijing) Co., Ltd., Beijing, 102206, China
| | - Yang Li
- Department of Discovery Biomarkers, BeiGene (Beijing) Co., Ltd., Beijing, 102206, China
| | - Weiwei Song
- Department of Discovery Biomarkers, BeiGene (Beijing) Co., Ltd., Beijing, 102206, China
| | - Hanzi Sun
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, 102206, China
| | - Zuobai Wang
- Department of Clinic Development, BeiGene (Beijing) Co., Ltd., Beijing, 102206, China
| | - Xiaomin Song
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, 102206, China
| | - Zhirong Shen
- Department of Discovery Biomarkers, BeiGene (Beijing) Co., Ltd., Beijing, 102206, China
| | - Xuesong Liu
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, 102206, China
| | - Kang Li
- Department of Biologics, BeiGene (Beijing) Co., Ltd., Beijing, 102206, China
| | - Lai Wang
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, 102206, China
| | - Ye Liu
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, 102206, China.
| |
Collapse
|
7
|
Steele H, Cheng J, Willicut A, Dell G, Breckenridge J, Culberson E, Ghastine A, Tardif V, Herro R. TNF superfamily control of tissue remodeling and fibrosis. Front Immunol 2023; 14:1219907. [PMID: 37465675 PMCID: PMC10351606 DOI: 10.3389/fimmu.2023.1219907] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 06/13/2023] [Indexed: 07/20/2023] Open
Abstract
Fibrosis is the result of extracellular matrix protein deposition and remains a leading cause of death in USA. Despite major advances in recent years, there remains an unmet need to develop therapeutic options that can effectively degrade or reverse fibrosis. The tumor necrosis super family (TNFSF) members, previously studied for their roles in inflammation and cell death, now represent attractive therapeutic targets for fibrotic diseases. In this review, we will summarize select TNFSF and their involvement in fibrosis of the lungs, the heart, the skin, the gastrointestinal tract, the kidney, and the liver. We will emphasize their direct activity on epithelial cells, fibroblasts, and smooth muscle cells. We will further report on major clinical trials targeting these ligands. Whether in isolation or in combination with other anti-TNFSF member or treatment, targeting this superfamily remains key to improve efficacy and selectivity of currently available therapies for fibrosis.
Collapse
Affiliation(s)
- Hope Steele
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- University of Cincinnati, Cincinnati, OH, United States
| | - Jason Cheng
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Ashley Willicut
- University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Garrison Dell
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- University of Cincinnati, Cincinnati, OH, United States
| | - Joey Breckenridge
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- University of Cincinnati, Cincinnati, OH, United States
| | - Erica Culberson
- University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Andrew Ghastine
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Virginie Tardif
- Normandy University, UniRouen, Institut National de la Santé et de la Recherche Médicale (INSERM), UMR1096 (EnVI Laboratory), Rouen, France
| | - Rana Herro
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
8
|
Manian M, Motallebnezhad M, Nedaeinia R, Salehi R, Khani L, Ferns GA, Jazayeri MH. Comparison of OX40 expression in patients with multiple sclerosis and neuromyelitis optica as an approach to diagnosis. ALLERGY, ASTHMA, AND CLINICAL IMMUNOLOGY : OFFICIAL JOURNAL OF THE CANADIAN SOCIETY OF ALLERGY AND CLINICAL IMMUNOLOGY 2023; 19:19. [PMID: 36899405 PMCID: PMC10007837 DOI: 10.1186/s13223-023-00772-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 02/13/2023] [Indexed: 03/12/2023]
Abstract
BACKGROUND Previous studies have shown that CD134 (OX40) co-stimulation is involved in the pathogenesis of experimental autoimmune encephalomyelitis (EAE) models and the antigen is expressed within multiple sclerosis lesions in humans. OX40 (CD134) is thought to be a secondary co-stimulatory immune checkpoint molecule that is expressed by T cells. This study aimed to evaluate the mRNA expression of OX40 and its serum levels in the peripheral blood of patients with Multiple Sclerosis (MS) or Neuromyelitis Optica (NMO). METHODS Patients with MS (n = 60), NMO (n = 20), and 20 healthy subjects were recruited from Sina Hospital, Tehran, Iran. The diagnoses were confirmed by a specialist in clinical neurology. Peripheral venous blood was obtained from all subjects, and mRNA quantification of OX40 was conducted using real-time PCR. Serum samples were also obtained and the concentration of OX40 was determined using an enzyme-linked immunosorbent assay (ELISA). RESULTS There was a significant correlation between the mRNA expression and serum levels of OX40 and disability as assessed using the expanded disability status scale (EDSS) in the patients with MS, but not in the patients with NMO. Expression of OX40 mRNA was significantly higher in the peripheral blood of MS patients compared to healthy individuals and NMO patients (*P < 0.05). In addition, serum OX40 concentrations were also significantly higher in patients with MS patients compared with healthy subjects (9.08 ± 2.48 vs. 1.49 ± 0.54 ng/ml; P = 0.041). CONCLUSIONS It appears that an increased expression of OX40 may be associated with the hyperactivation of T cells in patients with MS, and this may play a role in the pathogenesis of the disease.
Collapse
Affiliation(s)
- Mostafa Manian
- Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Morteza Motallebnezhad
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Shahid Hemmat Highway, P.O Box: 14665-354, Tehran, 1449614535, Iran
| | - Reza Nedaeinia
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Rasoul Salehi
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran.,Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Leila Khani
- Laboratory of Transcriptional Regulation, Institute of Medical Biology, Polish Academy of Science, Lodz, Poland.,Bio-Med-Chem Doctoral School of the University of Lodz, Lodz Institutes of the Polish Academy of Sciences, Lodz, Poland
| | - Gordon A Ferns
- Division of Medical Education, Brighton and Sussex Medical School, Falmer, Brighton, Sussex, BN1 9PH, UK
| | - Mir Hadi Jazayeri
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Shahid Hemmat Highway, P.O Box: 14665-354, Tehran, 1449614535, Iran. .,Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Zhao L, Zhang W, Liu M, Jia R, Wang J, Wang F, Xu Y. OX40L enhances the immunogenicity of dendritic cells and inhibits tumor metastasis in mice. Microbiol Immunol 2023; 67:79-89. [PMID: 36345699 DOI: 10.1111/1348-0421.13037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 10/22/2022] [Accepted: 11/06/2022] [Indexed: 11/10/2022]
Abstract
A well preserved immune system is a powerful tool to prevent foreign invasion or to suppress internal mutation, which must be tightly controlled by co-stimulatory molecules in different pathophysiological conditions. One such critical molecule is OX40L expressed on activated antigen-presenting cells (APCs). Consistently, its abnormality is associated with various immunological disorders such as autoinflammatory diseases and allergy. However, a comprehensive analysis of the immune-moderating role of OX40L in dendritic cells (DCs), the most powerful APCs to initiate immune responses in vivo, and investigation of its anti-tumor efficacy in the disease setting have not been performed properly. In this study, genetic approaches for both gain-of-function and reduction-of-function were employed to reveal that OX40L was required for the efficient presentation, but not uptake, of antigens by DCs to stimulate CD4+ , as well as CD8+ T cells in vivo. As a result, CD4+ T cells were promoted towards Th1, but inhibited on Treg differentiation, by the LPS-induced OX40L on DCs, which was supported by their altered expression of co-inhibitory receptor, PD-L1. CD8+ T cells, on the other hand, also enhanced their cytotoxicity towards target cells in response to OX40L expression on the DCs transferred in vivo. Finally, in a DC-mediated tumor immunity model, the strong immunogenic roles of OX40L on DCs led to better metastasis inhibition in vivo. Collectively, our results demonstrate that OX40L could serve as a potential target in the DC-based vaccine for enhanced anti-tumor efficacy in vivo.
Collapse
Affiliation(s)
- Lin Zhao
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, School of Life Sciences, Anhui Normal University, Wuhu, China
| | - Wenjie Zhang
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, School of Life Sciences, Anhui Normal University, Wuhu, China
| | - Meng Liu
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, School of Life Sciences, Anhui Normal University, Wuhu, China
| | - Ruoyu Jia
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, School of Life Sciences, Anhui Normal University, Wuhu, China
| | - Juncheng Wang
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, School of Life Sciences, Anhui Normal University, Wuhu, China
| | - Fengge Wang
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, School of Life Sciences, Anhui Normal University, Wuhu, China
| | - Yuekang Xu
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, School of Life Sciences, Anhui Normal University, Wuhu, China
| |
Collapse
|
10
|
Pan M, Zhao H, Jin R, Leung PSC, Shuai Z. Targeting immune checkpoints in anti-neutrophil cytoplasmic antibodies associated vasculitis: the potential therapeutic targets in the future. Front Immunol 2023; 14:1156212. [PMID: 37090741 PMCID: PMC10115969 DOI: 10.3389/fimmu.2023.1156212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/27/2023] [Indexed: 04/25/2023] Open
Abstract
Anti-neutrophil cytoplasmic autoantibodies (ANCA) associated vasculitis (AAV) is a necrotizing vasculitis mainly involving small blood vessels. It is demonstrated that T cells are important in the pathogenesis of AAV, including regulatory T cells (Treg) and helper T cells (Th), especially Th2, Th17, and follicular Th cells (Tfh). In addition, the exhaustion of T cells predicted the favorable prognosis of AAV. The immune checkpoints (ICs) consist of a group of co-stimulatory and co-inhibitory molecules expressed on the surface of T cells, which maintains a balance between the activation and exhaustion of T cells. CD28, inducible T-cell co-stimulator (ICOS), OX40, CD40L, glucocorticoid induced tumor necrosis factor receptor (GITR), and CD137 are the common co-stimulatory molecules, while the programmed cell death 1 (PD-1), cytotoxic T lymphocyte-associated molecule 4 (CTLA-4), T cell immunoglobulin (Ig) and mucin domain-containing protein 3 (TIM-3), B and T lymphocyte attenuator (BTLA), V-domain Ig suppressor of T cell activation (VISTA), T-cell Ig and ITIM domain (TIGIT), CD200, and lymphocyte activation gene 3 (LAG-3) belong to co-inhibitory molecules. If this balance was disrupted and the activation of T cells was increased, autoimmune diseases (AIDs) might be induced. Even in the treatment of malignant tumors, activation of T cells by immune checkpoint inhibitors (ICIs) may result in AIDs known as rheumatic immune-related adverse events (Rh-irAEs), suggesting the importance of ICs in AIDs. In this review, we summarized the features of AAV induced by immunotherapy using ICIs in patients with malignant tumors, and then reviewed the biological characteristics of different ICs. Our aim was to explore potential targets in ICs for future treatment of AAV.
Collapse
Affiliation(s)
- Menglu Pan
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Huanhuan Zhao
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ruimin Jin
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Patrick S. C. Leung
- Division of Rheumatology/Allergy and Clinical Immunology, University of California, Davis, Davis, CA, United States
- *Correspondence: Zongwen Shuai, ; Patrick S. C. Leung,
| | - Zongwen Shuai
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
- *Correspondence: Zongwen Shuai, ; Patrick S. C. Leung,
| |
Collapse
|
11
|
Padula L, Fisher E, Rivas K, Podack K, Frasca D, Kupritz J, Seavey MM, Jayaraman P, Dixon E, Jasuja R, Strbo N. Secreted heat shock protein gp96-Ig and OX40L-Fc combination vaccine enhances SARS-CoV-2 Spike (S) protein-specific B and T cell immune responses. Vaccine X 2022; 12:100202. [PMID: 35936992 PMCID: PMC9347141 DOI: 10.1016/j.jvacx.2022.100202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 04/15/2022] [Accepted: 07/29/2022] [Indexed: 11/30/2022] Open
Abstract
gp96-Ig-S-OX40L-Fc vaccine enhances S-specific IgG responses. gp96-Ig-S-OX40L-Fc vaccine enhances TFH cell responses. gp96-Ig-S-OX40L-Fc vaccine enhances lungs S-specific CD8 + T cell responses.
Encouraging protection results from current mRNA-based SARS-CoV-2 vaccine platforms are primarily due to the induction of SARS- CoV-2- specific B cell antibody and CD4 + T cell. Even though, current mRNA vaccine platforms are adept in inducing SARS-CoV2-specific CD8 + T cell, much less is known about CD8 T cells contribution to the overall vaccine protection. Our allogeneic cellular vaccine, based on a secreted form of the heat-shock protein gp96-Ig, achieves high frequencies of polyclonal CD8 + T cell responses to tumor and infectious antigens through antigen cross-priming in vivo. We and others have shown that gp96-Ig, in addition to antigen-specific CD8 + T cell anti-tumor and anti-pathogen immunity, primes antibody responses as well. Here, we generated a cell-based vaccine that expresses SARS-Cov-2 Spike (S) protein and simultaneously secretes gp96-Ig and OX40L-Fc fusion proteins. We show that co-secretion of gp96-Ig-S peptide complexes and the OX40L-Fc costimulatory fusion protein in allogeneic cell lines results in enhanced activation of S protein-specific IgG antibody responses. These findings were further strengthened by the observation that this vaccine platform induces T follicular helper cells (TFH) and protein-S -specific CD8 + T cells. Thus, a cell-based gp96-Ig vaccine/OX40-L fusion protein regimen provides encouraging translational data that this vaccine platform induces pathogen-specific CD8+, CD4 + T and B cell responses, and may cohesively work as a booster for FDA-approved vaccines. Our vaccine platform can be rapidly engineered and customized based on other current and future pathogen sequences.
Collapse
Affiliation(s)
- Laura Padula
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Eva Fisher
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Katelyn Rivas
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Kristin Podack
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Daniela Frasca
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Jonah Kupritz
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | | | | | - Eric Dixon
- Heat Biologics, Inc. Morrisville, NC, USA
| | | | - Natasa Strbo
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, USA
- Corresponding author at: Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, 1600 NW 10 Avenue, Miami, FL, 33136, USA.
| |
Collapse
|
12
|
Das A, Deka D, Banerjee A, Radhakrishnan AK, Zhang H, Sun XF, Pathak S. A Concise Review on the Role of Natural and Synthetically Derived Peptides in Colorectal Cancer. Curr Top Med Chem 2022; 22:2571-2588. [PMID: 35578849 DOI: 10.2174/1568026622666220516105049] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 03/29/2022] [Accepted: 04/06/2022] [Indexed: 01/20/2023]
Abstract
Colorectal cancer being the second leading cause of cancer-associated deaths has become a significant health concern around the globe. Though there are various cancer treatment approaches, many of them show adverse effects and some compromise the health of cancer patients. Hence, significant efforts are being made for the evolution of a novel biological therapeutic approach with better efficacy and minimal side effects. Current research suggests that the application of peptides in colorectal cancer therapeutics holds the possibility of the emergence of an anticancer reagent. The primary beneficial factors of peptides are their comparatively rapid and easy process of synthesis and the enormous potential for chemical alterations that can be evaluated for designing novel peptides and enhancing the delivery capacity of peptides. Peptides might be utilized as agents with cytotoxic activities or as a carrier of a specific drug or as cytotoxic agents that can efficiently target the tumor cells. Further, peptides can also be used as a tool for diagnostic purposes. The recent analysis aims at developing peptides that have the potential to efficiently target the tumor moieties without harming the nearby normal cells. Additionally, decreasing the adverse effects, and unfolding the other therapeutic properties of potential peptides, are also the subject matter of in-depth analysis. This review provides a concise summary of the function of both natural and synthetically derived peptides in colorectal cancer therapeutics that are recently being evaluated and their potent applications in the clinical field.
Collapse
Affiliation(s)
- Alakesh Das
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Kelambakkam, Chennai, India
| | - Dikshita Deka
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Kelambakkam, Chennai, India
| | - Antara Banerjee
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Kelambakkam, Chennai, India
| | - Arun Kumar Radhakrishnan
- Department of Pharmacology, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Kelambakkam, Chennai, India
| | - Hong Zhang
- School of Medicine, Department of Medical Sciences, Örebro University, Örebro, Sweden
| | - Xiao-Feng Sun
- Department of Oncology and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Surajit Pathak
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Kelambakkam, Chennai, India
| |
Collapse
|
13
|
Rastogi I, Jeon D, Moseman JE, Muralidhar A, Potluri HK, McNeel DG. Role of B cells as antigen presenting cells. Front Immunol 2022; 13:954936. [PMID: 36159874 PMCID: PMC9493130 DOI: 10.3389/fimmu.2022.954936] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 08/19/2022] [Indexed: 01/27/2023] Open
Abstract
B cells have been long studied for their role and function in the humoral immune system. Apart from generating antibodies and an antibody-mediated memory response against pathogens, B cells are also capable of generating cell-mediated immunity. It has been demonstrated by several groups that B cells can activate antigen-specific CD4 and CD8 T cells, and can have regulatory and cytotoxic effects. The function of B cells as professional antigen presenting cells (APCs) to activate T cells has been largely understudied. This, however, requires attention as several recent reports have demonstrated the importance of B cells within the tumor microenvironment, and B cells are increasingly being evaluated as cellular therapies. Antigen presentation through B cells can be through antigen-specific (B cell receptor (BCR) dependent) or antigen non-specific (BCR independent) mechanisms and can be modulated by a variety of intrinsic and external factors. This review will discuss the pathways and mechanisms by which B cells present antigens, and how B cells differ from other professional APCs.
Collapse
|
14
|
Cortellino S, Raveane A, Chiodoni C, Delfanti G, Pisati F, Spagnolo V, Visco E, Fragale G, Ferrante F, Magni S, Iannelli F, Zanardi F, Casorati G, Bertolini F, Dellabona P, Colombo MP, Tripodo C, Longo VD. Fasting renders immunotherapy effective against low-immunogenic breast cancer while reducing side effects. Cell Rep 2022; 40:111256. [PMID: 36001966 DOI: 10.1016/j.celrep.2022.111256] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 05/10/2022] [Accepted: 07/28/2022] [Indexed: 11/26/2022] Open
Abstract
Immunotherapy is improving the prognosis and survival of cancer patients, but despite encouraging outcomes in different cancers, the majority of tumors are resistant to it, and the immunotherapy combinations are often accompanied by severe side effects. Here, we show that a periodic fasting-mimicking diet (FMD) can act on the tumor microenvironment and increase the efficacy of immunotherapy (anti-PD-L1 and anti-OX40) against the poorly immunogenic triple-negative breast tumors (TNBCs) by expanding early exhausted effector T cells, switching the cancer metabolism from glycolytic to respiratory, and reducing collagen deposition. Furthermore, FMD reduces the occurrence of immune-related adverse events (irAEs) by preventing the hyperactivation of the immune response. These results indicate that FMD cycles have the potential to enhance the efficacy of anti-cancer immune responses, expand the portion of tumors sensitive to immunotherapy, and reduce its side effects.
Collapse
Affiliation(s)
| | - Alessandro Raveane
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, Milan 20141, Italy
| | - Claudia Chiodoni
- Department of Research, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Gloria Delfanti
- Experimental Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Federica Pisati
- IFOM, FIRC Institute of Molecular Oncology, Milan 20139, Italy
| | | | - Euplio Visco
- IFOM, FIRC Institute of Molecular Oncology, Milan 20139, Italy
| | | | | | - Serena Magni
- IFOM, FIRC Institute of Molecular Oncology, Milan 20139, Italy
| | - Fabio Iannelli
- IFOM, FIRC Institute of Molecular Oncology, Milan 20139, Italy
| | | | - Giulia Casorati
- Experimental Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Bertolini
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, Milan 20141, Italy; Onco-Tech Lab, European Institute of Oncology IRCCS and Politecnico di Milano, Milan, Italy
| | - Paolo Dellabona
- Experimental Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Mario P Colombo
- Department of Research, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Claudio Tripodo
- IFOM, FIRC Institute of Molecular Oncology, Milan 20139, Italy; University of Palermo School of Medicine, Palermo, Italy
| | - Valter D Longo
- IFOM, FIRC Institute of Molecular Oncology, Milan 20139, Italy; Longevity Institute and Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
15
|
Yadav R, Redmond WL. Current Clinical Trial Landscape of OX40 Agonists. Curr Oncol Rep 2022; 24:951-960. [PMID: 35352295 DOI: 10.1007/s11912-022-01265-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/26/2022] [Indexed: 12/17/2022]
Abstract
PURPOSE OF REVIEW Despite the efficacy of immune checkpoint blockade (ICB) immunotherapy, most cancer patients still develop progressive disease necessitating additional treatment options. One approach is ligation of the OX40 (CD134) costimulatory receptor which promotes T cell activation, effector function, and the generation of long-lived memory cells. RECENT FINDINGS Numerous preclinical studies have demonstrated that OX40 agonists alone or in combination with ICB (e.g., anti-PD-1, anti-PD-L1, and anti-CTLA-4) augment anti-tumor immunity. In this review, we discuss the impact of OX40 agonists on T cell function and the therapeutic potential of OX40 agonists alone or in conjunction with ICB for patients with advanced malignancies.
Collapse
Affiliation(s)
- Rashi Yadav
- Earle A. Chiles Research Institute, Providence Cancer Institute, 4805 NE Glisan St., 2N35, Portland, OR, 97213, USA
| | - William L Redmond
- Earle A. Chiles Research Institute, Providence Cancer Institute, 4805 NE Glisan St., 2N35, Portland, OR, 97213, USA.
| |
Collapse
|
16
|
Yang Y, Song S, Nie Y, Chen R, Chen P. Lentinan alleviates arsenic-induced hepatotoxicity in mice via downregulation of OX40/IL-17A and activation of Nrf2 signaling. BMC Pharmacol Toxicol 2022; 23:16. [PMID: 35313999 PMCID: PMC8939159 DOI: 10.1186/s40360-022-00557-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 03/10/2022] [Indexed: 02/05/2023] Open
Abstract
Background Arsenic, existing ubiquitously in soil, drinking water, or food, is well known to be an environmental pollutants concerned by European Food Safety Authority. Lentinan, a beta-1,6;1,3-glucan extracts from Lentinus edodes, which has the properties of antioxidant and immunomodulation, present study explored the pharmacological effects of Lentinan on arsenic induced hepatotoxicity in mice. Methods Mice experiments were performed by sodium arsenite (SA) treatment or Lentinan intervention, then histopathology, ELISA, Flow Cytometry, or Western-Blotting were applied to evaluate hepatic injury, oxidative stress, CD4+ type 17 helper T (Th17) cells, CD4+CD25+Foxp3+ regulatory T cells (Tregs), T cells receptor OX40/CD134, IL-17A, NLRP3, Nrf2, and NQO1. Results SA treatment showed hepatic pathological injury and the elevations of alanine aminotransferase (ALT) or aspartate aminotransferase (AST) in serum, and induced the increases of malondialdehyde (MDA), Th17 cells, OX40 or IL-17A in liver tissues, which were consistently ameliorated by Lentinan intervention. Further, immunoblotting experiments showed that Lentinan intervention downregulated the levels of OX40, IL-17A, and NLRP3 signals, while elevated the levels of anti-oxidative Nrf2, NQO1 signals compared to arsenic treatment group. For Tregs, Lentinan intervention showed no significant difference from SA treatment group. Conclusion Lentinan antagonizes SA-induced hepatotoxicity in mice, may be involved in the downregulations of pro-inflammatory OX40 or IL-17A and the activation of anti-oxidative Nrf2, NQO1 signals. Supplementary Information The online version contains supplementary material available at 10.1186/s40360-022-00557-7.
Collapse
Affiliation(s)
- Yuan Yang
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, China. .,Dong Medicine Key laboratory of Hunan Province, Department of Laboratory Medicine, Hunan University of Medicine, Huaihua, 418000, China. .,Department of Toxicology, School of Public Health, Guilin Medical University, Guilin, 541004, China.
| | - Shuang Song
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, China
| | - Yuanyuan Nie
- Dong Medicine Key laboratory of Hunan Province, Department of Laboratory Medicine, Hunan University of Medicine, Huaihua, 418000, China
| | - Rong Chen
- Dong Medicine Key laboratory of Hunan Province, Department of Laboratory Medicine, Hunan University of Medicine, Huaihua, 418000, China
| | - Peng Chen
- Dong Medicine Key laboratory of Hunan Province, Department of Laboratory Medicine, Hunan University of Medicine, Huaihua, 418000, China
| |
Collapse
|
17
|
Fu N, Xie F, Sun Z, Wang Q. The OX40/OX40L Axis Regulates T Follicular Helper Cell Differentiation: Implications for Autoimmune Diseases. Front Immunol 2021; 12:670637. [PMID: 34234777 PMCID: PMC8256170 DOI: 10.3389/fimmu.2021.670637] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 06/02/2021] [Indexed: 01/11/2023] Open
Abstract
T Follicular helper (Tfh) cells, a unique subset of CD4+ T cells, play an essential role in B cell development and the formation of germinal centers (GCs). Tfh differentiation depends on various factors including cytokines, transcription factors and multiple costimulatory molecules. Given that OX40 signaling is critical for costimulating T cell activation and function, its roles in regulating Tfh cells have attracted widespread attention. Recent data have shown that OX40/OX40L signaling can not only promote Tfh cell differentiation and maintain cell survival, but also enhance the helper function of Tfh for B cells. Moreover, upregulated OX40 signaling is related to abnormal Tfh activity that causes autoimmune diseases. This review describes the roles of OX40/OX40L in Tfh biology, including the mechanisms by which OX40 signaling regulates Tfh cell differentiation and functions, and their close relationship with autoimmune diseases.
Collapse
Affiliation(s)
- NanNan Fu
- School of Biology & Basic Medical Sciences, Medical College of Soochow University, Suzhou, China
| | - Fang Xie
- School of Biology & Basic Medical Sciences, Medical College of Soochow University, Suzhou, China
| | - ZhongWen Sun
- Department of Medical Technology, Suzhou Vocational Health College, Suzhou, China
| | - Qin Wang
- School of Biology & Basic Medical Sciences, Medical College of Soochow University, Suzhou, China
| |
Collapse
|
18
|
Quetglas JI, John LB, Kershaw MH, Alvarez-Vallina L, Melero I, Darcy PK, Smerdou C. Virotherapy, gene transfer and immunostimulatory monoclonal antibodies. Oncoimmunology 2021; 1:1344-1354. [PMID: 23243597 PMCID: PMC3518506 DOI: 10.4161/onci.21679] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Malignant cells are susceptible to viral infection and consequent cell death. Virus-induced cell death is endowed with features that are known to stimulate innate and adaptive immune responses. Thus danger signals emitted by cells succumbing to viral infection as well as viral nucleic acids are detected by specific receptors, and tumor cell antigens can be routed to professional antigen-presenting cells. The anticancer immune response triggered by viral infection is frequently insufficient to eradicate malignancy but may be further amplified. For this purpose, transgenes encoding cytokines as co-stimulatory molecules can be genetically engineered into viral vectors. Alternatively, or in addition, it is possible to use monoclonal antibodies that either block inhibitory receptors of immune effector cells, or act as agonists for co-stimulatory receptors. Combined strategies are based on the ignition of a local immune response at the malignant site plus systemic immune boosting. We have recently reported examples of this approach involving the Vaccinia virus or Semliki Forest virus, interleukin-12 and anti-CD137 monoclonal antibodies.
Collapse
Affiliation(s)
- José I Quetglas
- Division of Hepatology and Gene Therapy; Center for Applied Medical Research; University of Navarra; Pamplona, Spain
| | | | | | | | | | | | | |
Collapse
|
19
|
Li M, Wang L, Yu C, Wang J. Development of a robust reporter gene assay for measuring the bioactivity of OX40-targeted therapeutic antibodies. LUMINESCENCE 2021; 36:885-893. [PMID: 33382183 DOI: 10.1002/bio.4004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/12/2020] [Accepted: 12/27/2020] [Indexed: 11/12/2022]
Abstract
OX40 plays a prominent role in the onset and development of solid tumors, and OX40-targeted monoclonal antibodies (mAbs) have entered clinical trials for various tumors. Bioactivity determination of therapeutic mAbs is of great significance in product quality, however, mechanism of action-based bioassays to determine the bioactivity of anti-OX40 mAbs is still lacking. Here, we established a reporter gene assay system based on two cell lines, namely Jurkat-OX40-NFκB-Luc which stably expresses NFκB-controlled luciferase, and Raji cells which inherently express FcγRs. In the model, FcγRs on Raji cells could crosslink the Fc of anti-OX40 mAbs, which leads to the further crosslinking between Fab of anti-OX40 mAbs and OX40 on Jurkat-OX40-NFκB-Luc cells. OX40 crosslinking could activate Jurkat-OX40-NFκB-Luc cells, and induce the expression of NFκB-controlled luciferase, the extent of which could reflect the bioactivity of anti-OX40 mAbs in a dose-dependent manner. After the optimization of various assay conditions, the validation of the cell-based bioassay showed good assay performance characteristics, including specificity, accuracy, precision, linearity, and stability. This innovative assay that is based on the OX40-NFκB pathway can be a powerful pool to measure the bioactivity of OX40-targeted mAbs.
Collapse
Affiliation(s)
- Meng Li
- Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, National Institutes for Food and Drug Control, Beijing, China
| | - Lan Wang
- Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, National Institutes for Food and Drug Control, Beijing, China
| | - Chuanfei Yu
- Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, National Institutes for Food and Drug Control, Beijing, China
| | - Junzhi Wang
- Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, National Institutes for Food and Drug Control, Beijing, China
| |
Collapse
|
20
|
Markasz L, Olsson KW, Holmström G, Sindelar R. Cluster Analysis of Early Postnatal Biochemical Markers May Predict Development of Retinopathy of Prematurity. Transl Vis Sci Technol 2020; 9:14. [PMID: 33344058 PMCID: PMC7726592 DOI: 10.1167/tvst.9.13.14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 10/19/2020] [Indexed: 12/15/2022] Open
Abstract
Purpose Growth factors and inflammatory and angiogenetic proteins are involved in the development of retinopathy of prematurity (ROP). However, no early biochemical markers are in clinical use to predict ROP. By performing cluster analysis of multiple biomarkers, we aimed to determine patient groups with high and low risk for developing ROP. Methods In total, 202 protein markers in plasma were quantified by proximity extension assay from 35 extremely preterm infants on day 2 of life. Infants were sorted in groups by automated two-dimensional hierarchical clustering of all biomarkers. ROP was classified as stages I to III with or without surgical treatment. Predictive biomarkers were evaluated by analysis of variance and detected differences by two-sided paired t-test with Bonferroni corrections for multiple comparisons. Results Differences in 39 biochemical markers divided infants without ROP into two control groups (control 1, n = 7; control 2, n = 5; P < 0.05). Sixty-six biochemical markers defined differences between the control groups (n = 13) and all ROP infants (n = 23; P < 0.05). PARK7, VIM, MPO, CD69, and NEMO were markedly increased in control 1 compared to all ROP infants (P < 0.001). Lower TNFRSF4 and higher HER2 and GAL appeared in infants with ROP as compared to control 1 and/or 2 (P < 0.05, respectively). Conclusions Our data suggest that early elevated levels of PARK7, VIM, MPO, CD69, and NEMO may be associated with lower risk of developing ROP. Lower levels of TNFRSF4 with higher levels of HER2 and GAL may predict ROP development. Translational Relevance Cluster analysis of early postnatal biomarkers may help to identify infants with low or high risk of developing ROP.
Collapse
Affiliation(s)
- Laszlo Markasz
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Karl-Wilhelm Olsson
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden.,Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Gerd Holmström
- Department of Neuroscience/Ophthalmology, Uppsala University, Uppsala, Sweden
| | - Richard Sindelar
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| |
Collapse
|
21
|
Abstract
Therapeutic targeting of immune checkpoints has garnered significant attention in the area of cancer immunotherapy, in which efforts have focused in particular on cytotoxic T lymphocyte antigen 4 (CTLA4) and PD1, both of which are members of the CD28 family. In autoimmunity, these same pathways can be targeted to opposite effect: to curb the over-exuberant immune response. The CTLA4 checkpoint serves as an exemplar, whereby CTLA4 activity is blocked by antibodies in cancer immunotherapy and augmented by the provision of soluble CTLA4 in autoimmunity. Here, we review the targeting of co-stimulatory molecules in autoimmune diseases, focusing in particular on agents directed at members of the CD28 or tumour necrosis factor receptor families. We present the state of the art in co-stimulatory blockade approaches, including rational combinations of immune inhibitory agents, and discuss the future opportunities and challenges in this field.
Collapse
|
22
|
Th1 responses in vivo require cell-specific provision of OX40L dictated by environmental cues. Nat Commun 2020; 11:3421. [PMID: 32647184 PMCID: PMC7347572 DOI: 10.1038/s41467-020-17293-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 06/23/2020] [Indexed: 12/24/2022] Open
Abstract
The OX40-OX40L pathway provides crucial co-stimulatory signals for CD4 T cell responses, however the precise cellular interactions critical for OX40L provision in vivo and when these occur, remains unclear. Here, we demonstrate that provision of OX40L by dendritic cells (DCs), but not T cells, B cells nor group 3 innate lymphoid cells (ILC3s), is critical specifically for the effector Th1 response to an acute systemic infection with Listeria monocytogenes (Lm). OX40L expression by DCs is regulated by cross-talk with NK cells, with IFNγ signalling to the DC to enhance OX40L in a mechanism conserved in both mouse and human DCs. Strikingly, DC expression of OX40L is redundant in a chronic intestinal Th1 response and expression by ILC3s is necessary. Collectively these data reveal tissue specific compartmentalisation of the cellular provision of OX40L and define a mechanism controlling DC expression of OX40L in vivo. The OX40-OX40L axis is a crucial component of the costimulatory requirement of CD4 T cell responses. Here, the authors show context and cell type specific expression of OX40L for driving Th1 cell generation during acute and chronic models of infection.
Collapse
|
23
|
Chauhan SKS, Koehl U, Kloess S. Harnessing NK Cell Checkpoint-Modulating Immunotherapies. Cancers (Basel) 2020; 12:E1807. [PMID: 32640575 PMCID: PMC7408278 DOI: 10.3390/cancers12071807] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/30/2020] [Accepted: 07/02/2020] [Indexed: 12/11/2022] Open
Abstract
During the host immune response, the precise balance of the immune system, regulated by immune checkpoint, is required to avoid infection and cancer. These immune checkpoints are the mainstream regulator of the immune response and are crucial for self-tolerance. During the last decade, various new immune checkpoint molecules have been studied, providing an attractive path to evaluate their potential role as targets for effective therapeutic interventions. Checkpoint inhibitors have mainly been explored in T cells until now, but natural killer (NK) cells are a newly emerging target for the determination of checkpoint molecules. Simultaneously, an increasing number of therapeutic dimensions have been explored, including modulatory and inhibitory checkpoint molecules, either causing dysfunction or promoting effector functions. Furthermore, the combination of the immune checkpoint with other NK cell-based therapeutic strategies could also strengthen its efficacy as an antitumor therapy. In this review, we have undertaken a comprehensive review of the literature to date regarding underlying mechanisms of modulatory and inhibitory checkpoint molecules.
Collapse
Affiliation(s)
| | - Ulrike Koehl
- Institute of cellular therapeutics, Hannover Medical School, 30625 Hannover, Germany; (U.K.); (S.K.)
- Fraunhofer Institute for Cell Therapy and Immunology, 04103 Leipzig, Germany
- Institute of Clinical Immunology, University of Leipzig, 04103 Leipzig, Germany
| | - Stephan Kloess
- Institute of cellular therapeutics, Hannover Medical School, 30625 Hannover, Germany; (U.K.); (S.K.)
| |
Collapse
|
24
|
Kashima J, Okuma Y, Hosomi Y, Hishima T. High Serum OX40 and OX40 Ligand (OX40L) Levels Correlate with Reduced Survival in Patients with Advanced Lung Adenocarcinoma. Oncology 2020; 98:303-310. [PMID: 32097938 DOI: 10.1159/000505975] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 01/16/2020] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Interaction of OX40 and OX40 ligand (OX40L) is associated with immune activation. OX40-OX40L axis is also suggested to play a role in immunity against several solid malignancies. OBJECTIVE In this study, serum OX40 and OX40L levels in patients with advanced lung adenocarcinoma were assessed and their correlation with survival and clinicopathologic parameters was determined. METHODS Serum samples were collected from patients with advanced lung adenocarcinoma, then OX40 and OX40L were quantified via enzyme-linked immunosorbent assay. Immunohistochemical (IHC) analysis of OX40 and OX40L in resected primary lesions was also performed. The association between OX40 and OX40L levels and clinicopathologic status and patient survival was retrospectively analyzed. RESULTS A total of 56 patients were analyzed. Median serum OX40 and OX40L levels were 156.2 pg/mL and 186.6 pg/mL, respectively. IHC analysis in 5 patients indicated high positivity of OX40 in tumor-infiltrating lymphocytes and of OX40L in tumor cells in mucinous adenocarcinoma. Patients with a high OX40 level (≥152.2 pg/mL) had poorer prognosis than those with a low serum OX40 level (median survival, 7.36 vs. 21.19 months, respectively, p = 0.04). Patients with a high OX40L level (≥207.3 pg/mL) had poorer prognosis than those with a low serum OX40L level (median survival, 7.36 vs. 14.26 months, respectively, p = 0.04). In the subset of patients treated with immune checkpoint inhibitors (ICIs) (n = 12), those with a high OX40L level were found to have longer survival from ICI initiation than those with a low OX40L level (p = 0.023). CONCLUSIONS High OX40 and OX40L levels are associated with poor prognosis and may reflect the immune-exhausted status against lung adenocarcinoma.
Collapse
Affiliation(s)
- Jumpei Kashima
- Department of Pathology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan.,Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yusuke Okuma
- Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan, .,Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan,
| | - Yukio Hosomi
- Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Tsunekazu Hishima
- Department of Pathology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| |
Collapse
|
25
|
Alves Costa Silva C, Facchinetti F, Routy B, Derosa L. New pathways in immune stimulation: targeting OX40. ESMO Open 2020; 5:e000573. [PMID: 32392177 PMCID: PMC7046367 DOI: 10.1136/esmoopen-2019-000573] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 10/15/2019] [Accepted: 10/20/2019] [Indexed: 12/14/2022] Open
Abstract
Immune checkpoint blockers (ICB) reinvigorate the immune system by removing the molecular brakes responsible for the scarce activity of immune phenotypes against malignant cells. After having proven their remarkable role as monotherapy, combinations of anti-Programmed cell death 1 (PD-1)/Programmed death-ligand 1 (PD-L1) agents with cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) antibodies, chemotherapy and/or anti-angiogenic compounds provide unprecedented results and durable responses across a variety of tumour types. Nevertheless, the main drawbacks of ICB are represented by primary and acquired resistance, translating into disease progression, as well as by immune-related toxicities. In this sense, novel strategies to foster the immune system through its direct stimulation are being tested in order to provide additional clinical improvements in patients with cancer. In this scenario, the co-stimulatory molecule OX40 (CD134) belongs to the next generation of immune therapeutic targets. Preliminary results of early clinical trials evaluating OX40 stimulation by means of different agents are encouraging. Here we review the rationale of OX40 targeting, highlighting the combination of OX40-directed therapies with different anticancer agents as a potential strategy to foster the immune system against malignant phenotypes.
Collapse
Affiliation(s)
| | | | | | - Lisa Derosa
- INSERM U1015, Gustave Roussy Institute, Villejuif, France
| |
Collapse
|
26
|
Sun G, Jin H, Zhang C, Meng H, Zhao X, Wei D, Ou X, Wang Q, Li S, Wang T, Sun X, Shi W, Tian D, Liu K, Xu H, Tian Y, Li X, Guo W, Jia J, Zhang Z, Zhang D. OX40 Regulates Both Innate and Adaptive Immunity and Promotes Nonalcoholic Steatohepatitis. Cell Rep 2019; 25:3786-3799.e4. [PMID: 30590049 DOI: 10.1016/j.celrep.2018.12.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 07/26/2018] [Accepted: 11/30/2018] [Indexed: 02/08/2023] Open
Abstract
Both innate and adaptive immune cells are involved in the pathogenesis of nonalcoholic steatohepatitis (NASH), but the crosstalk between innate and adaptive immunity is largely unknown. Here we show that compared with WT mice, OX40-/- mice exhibit decreased liver fat accumulation, lobular inflammation, and focal necrosis after feeding with diets that induce NASH. Mechanistically, OX40 deficiency suppresses Th1 and Th17 differentiation, and OX40 deficiency in T cells inhibits monocyte migration, antigen presentation, and M1 polarization. Soluble OX40 stimulation alone upregulates antigen presentation, chemokine receptor expression, and proinflammatory cytokine secretion by liver monocytes. Furthermore, plasma soluble OX40 levels are positively associated with NASH in humans, suggesting clinical relevance of the findings. In conclusion, we show a mechanism for T cell regulation of innate immune cells. OX40 is a key regulator of both intrahepatic innate and adaptive immunity, generates two-way signals, and promotes both proinflammatory monocyte and macrophage and T cell function, resulting in NASH development.
Collapse
Affiliation(s)
- Guangyong Sun
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Clinical Research Institute, Beijing, 100050, China; Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, China
| | - Hua Jin
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Clinical Research Institute, Beijing, 100050, China; Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, China
| | - Chunpan Zhang
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Clinical Research Institute, Beijing, 100050, China; Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, China
| | - Hua Meng
- General Surgery Department, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; National Clinical Research Center for Digestive Diseases, Beijing, 100050, China
| | - Xinyan Zhao
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Dan Wei
- General Surgery Department, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; National Clinical Research Center for Digestive Diseases, Beijing, 100050, China
| | - Xiaojuan Ou
- National Clinical Research Center for Digestive Diseases, Beijing, 100050, China; Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Qianyi Wang
- National Clinical Research Center for Digestive Diseases, Beijing, 100050, China; Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Shuxiang Li
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Tianqi Wang
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Clinical Research Institute, Beijing, 100050, China; Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, China
| | - Xiaojing Sun
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Clinical Research Institute, Beijing, 100050, China; Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, China
| | - Wen Shi
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Clinical Research Institute, Beijing, 100050, China; Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, China
| | - Dan Tian
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Clinical Research Institute, Beijing, 100050, China; Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, China
| | - Kai Liu
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Clinical Research Institute, Beijing, 100050, China; Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, China
| | - Hufeng Xu
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Clinical Research Institute, Beijing, 100050, China; Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, China
| | - Yue Tian
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Clinical Research Institute, Beijing, 100050, China; Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, China
| | - Xinmin Li
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Clinical Research Institute, Beijing, 100050, China; Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, China
| | - Wei Guo
- General Surgery Department, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; National Clinical Research Center for Digestive Diseases, Beijing, 100050, China
| | - Jidong Jia
- National Clinical Research Center for Digestive Diseases, Beijing, 100050, China; Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, Beijing, 100050, China
| | - Zhongtao Zhang
- General Surgery Department, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; National Clinical Research Center for Digestive Diseases, Beijing, 100050, China.
| | - Dong Zhang
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Clinical Research Institute, Beijing, 100050, China; Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, China; National Clinical Research Center for Digestive Diseases, Beijing, 100050, China.
| |
Collapse
|
27
|
Mardomi A, Mohammadi N, Khosroshahi HT, Abediankenari S. An update on potentials and promises of T cell co-signaling molecules in transplantation. J Cell Physiol 2019; 235:4183-4197. [PMID: 31696513 DOI: 10.1002/jcp.29369] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 10/07/2019] [Indexed: 02/06/2023]
Abstract
The promising outcomes of immune-checkpoint based immunotherapies in cancer have provided a proportional perspective ahead of exploiting similar approaches in allotransplantation. Belatacept (CTLA-4-Ig) is an example of costimulation blockers successfully exploited in renal transplantation. Due to the wide range of regulatory molecules characterized in the past decades, some of these molecules might be candidates as immunomodulators in the case of tolerance induction in transplantation. Although there are numerous attempts on the apprehension of the effects of co-signaling molecules on immune response, the necessity for a better understanding is evident. By increasing the knowledge on the biology of co-signaling pathways, some pitfalls are recognized and improved approaches are proposed. The blockage of CD80/CD28 axis is an instance of evolution toward more efficacy. It is now evident that anti-CD28 antibodies are more effective than CD80 blockers in animal models of transplantation. Other co-signaling axes such as PD-1/PD-L1, CD40/CD154, 2B4/CD48, and others discussed in the present review are examples of critical immunomodulatory molecules in allogeneic transplantation. We review here the outcomes of recent experiences with co-signaling molecules in preclinical studies of solid organ transplantation.
Collapse
Affiliation(s)
- Alireza Mardomi
- Department of Immunology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran.,Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Nabiallah Mohammadi
- Department of Immunology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | | | - Saeid Abediankenari
- Department of Immunology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
28
|
Zeng Q, Zhou Y, Schwarz H. CD137L-DCs, Potent Immune-Stimulators-History, Characteristics, and Perspectives. Front Immunol 2019; 10:2216. [PMID: 31632390 PMCID: PMC6783506 DOI: 10.3389/fimmu.2019.02216] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 09/02/2019] [Indexed: 12/31/2022] Open
Abstract
Dendritic cell (DC)-based immunotherapies are being explored for over 20 years and found to be very safe. Most often, granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-4 (IL-4)-induced monocyte-derived DCs (moDCs) are being used, which have demonstrated some life-prolonging benefit to patients of multiple tumors. However, the limited clinical response and efficacy call for the development of more potent DCs. CD137L-DC may meet this demand. CD137L-DCs are a novel type of monocyte-derived inflammatory DCs that are induced by CD137 ligand (CD137L) agonists. CD137L is expressed on the surface of antigen-presenting cells, including monocytes, and signaling of CD137L into monocytes induces their differentiation to CD137L-DCs. CD137L-DCs preferentially induce type 1 T helper (Th1) cell polarization and strong type 1 CD8+ T cell (Tc1) responses against tumor-associated viral antigens. The in vitro T cell-stimulatory capacity of CD137L-DCs is superior to that of conventional moDCs. The transcriptomic profile of CD137L-DC is highly similar to that of in vivo DCs at sites of inflammation. The strict activation dependence of CD137 expression and its restricted expression on activated T cells, NK cells, and vascular endothelial cells at inflammatory sites make CD137 an ideally suited signal for the induction of monocyte-derived inflammatory DCs in vivo. These findings and their potency encouraged a phase I clinical trial of CD137L-DCs against Epstein-Barr virus-associated nasopharyngeal carcinoma. In this review, we introduce and summarize the history, the characteristics, and the transcriptional profile of CD137L-DC, and discuss the potential development and applications of CD137L-DC.
Collapse
Affiliation(s)
- Qun Zeng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Yubin Zhou
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Herbert Schwarz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| |
Collapse
|
29
|
Bourque J, Hawiger D. Immunomodulatory Bonds of the Partnership between Dendritic Cells and T Cells. Crit Rev Immunol 2019; 38:379-401. [PMID: 30792568 DOI: 10.1615/critrevimmunol.2018026790] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
By acquiring, processing, and presenting both foreign and self-antigens, dendritic cells (DCs) initiate T cell activation that is shaped through the immunomodulatory functions of a variety of cell-membrane-bound molecules including BTLA-HVEM, CD40-CD40L, CTLA-4-CD80/CD86, CD70-CD27, ICOS-ICOS-L, OX40-OX40L, and PD-L1-PD-1, as well as several key cytokines and enzymes such as interleukin-6 (IL-6), IL-12, IL-23, IL-27, transforming growth factor-beta 1 (TGF-β1), retinaldehyde dehydrogenase (Raldh), and indoleamine 2,3-dioxygenase (IDO). Some of these distinct immunomodulatory signals are mediated by specific subsets of DCs, therefore contributing to the functional specialization of DCs in the priming and regulation of immune responses. In addition to responding to the DC-mediated signals, T cells can reciprocally modulate the immunomodulatory capacities of DCs, further refining immune responses. Here, we review recent studies, particularly in experimental mouse systems, that have delineated the integrated mechanisms of crucial immunomodulatory pathways that enable specific populations of DCs and T cells to work intimately together as single functional units that are indispensable for the maintenance of immune homeostasis.
Collapse
Affiliation(s)
- Jessica Bourque
- Department of Molecular Microbiology and Immunology, St. Louis University School of Medicine, St. Louis, MO, USA
| | - Daniel Hawiger
- Department of Molecular Microbiology and Immunology, St. Louis University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
30
|
Immune Checkpoint Ligand Reverse Signaling: Looking Back to Go Forward in Cancer Therapy. Cancers (Basel) 2019; 11:cancers11050624. [PMID: 31060225 PMCID: PMC6563035 DOI: 10.3390/cancers11050624] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 04/30/2019] [Accepted: 05/02/2019] [Indexed: 02/06/2023] Open
Abstract
The so-called immune checkpoints are pathways that regulate the timing and intensity of the immune response to avoid an excessive reaction and to protect the host from autoimmunity. Immune checkpoint inhibitors (ICIs) are designed to target the negative regulatory pathways of T cells, and they have been shown to restore anti-tumor immune functions and achieve considerable clinical results. Indeed, several clinical trials have reported durable clinical response in different tumor types, such as melanoma, renal cell carcinoma (RCC) and non-small cell lung cancer (NSCLC). Nonetheless, after the initial enthusiasm, it is now evident that the majority of patients do not benefit from ICIs, due to innate or acquired tumor resistance. It is therefore mandatory to find ways to identify those patients who will respond and to find ways to induce response in those who at present do not benefit from ICIs. In this regard, the expression of programmed death ligand 1 (PD-L1) on neoplastic cells was the first, and most obvious, biomarker exploited to predict the activity of anti-programmed death 1 (PD-1) and/or anti-PD-L1 antibodies. As expected, a correlation was confirmed between the levels of PD-L1 and the efficacy of anti-PD-1 therapy in melanoma, NSCLC and RCC. However, further results from clinical trials showed that some patients display a clinical response regardless of tumor cell PD-L1 expression levels, while others do not benefit from ICI treatment despite the expression of PD-L1 on neoplastic elements. These findings strongly support the notion that other factors may be relevant for the efficacy of ICI-based treatment regimens. Furthermore, although the current dogma indicates that the PD-1/PD-L1 axis exerts its regulatory effects via the signal transduced in PD-1-expressing T cells, recent evidence suggests that a reverse signaling may also exist downstream of PD-L1 in both tumor and immune cells. The reverse signaling of PD-L1, but also of other immune checkpoints, might contribute to the pro-tumoral/immune suppressive environment associated with tumor development and progression. Clarifying this aspect could facilitate the prediction of patients’ clinical outcomes, which are so far unpredictable and result in response, resistance or even hyper-progressive disease in some cases.
Collapse
|
31
|
Abstract
Cancer remains the leading cause of death worldwide. Traditional treatments such as surgery, radiation, and chemotherapy have had limited efficacy, especially with late stage cancers. Cancer immunotherapy and targeted therapy have revolutionized how cancer is treated, especially in patients with late stage disease. In 2013 cancer immunotherapy was named the breakthrough of the year, partially due to the established efficacy of blockade of CTLA-4 and PD-1, both T cell co-inhibitory molecules involved in tumor-induced immunosuppression. Though early trials promised success, toxicity and tolerance to immunotherapy have hindered long-term successes. Optimizing the use of co-stimulatory and co-inhibitory pathways has the potential to increase the effectiveness of T cell-mediated antitumor immune response, leading to increased efficacy of cancer immunotherapy. This review will address major T cell co-stimulatory and co-inhibitory pathways and the role they play in regulating immune responses during cancer development and treatment.
Collapse
Affiliation(s)
- Rachel E O'Neill
- Department of Microbiology and Immunology, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD, United States
| | - Xuefang Cao
- Department of Microbiology and Immunology, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD, United States.
| |
Collapse
|
32
|
Lee WH, Seo D, Lim SG, Suk K. Reverse Signaling of Tumor Necrosis Factor Superfamily Proteins in Macrophages and Microglia: Superfamily Portrait in the Neuroimmune Interface. Front Immunol 2019; 10:262. [PMID: 30838001 PMCID: PMC6389649 DOI: 10.3389/fimmu.2019.00262] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 01/30/2019] [Indexed: 12/14/2022] Open
Abstract
The tumor necrosis factor (TNF) superfamily (TNFSF) is a protein superfamily of type II transmembrane proteins commonly containing the TNF homology domain. The superfamily contains more than 20 protein members, which can be released from the cell membrane by proteolytic cleavage. Members of the TNFSF function as cytokines and regulate diverse biological processes, including immune responses, proliferation, differentiation, apoptosis, and embryogenesis, by binding to TNFSF receptors. Many TNFSF proteins are also known to be responsible for the regulation of innate immunity and inflammation. Both receptor-mediated forward signaling and ligand-mediated reverse signaling play important roles in these processes. In this review, we discuss the functional expression and roles of various reverse signaling molecules and pathways of TNFSF members in macrophages and microglia in the central nervous system (CNS). A thorough understanding of the roles of TNFSF ligands and receptors in the activation of macrophages and microglia may improve the treatment of inflammatory diseases in the brain and periphery. In particular, TNFSF reverse signaling in microglia can be exploited to gain further insights into the functions of the neuroimmune interface in physiological and pathological processes in the CNS.
Collapse
Affiliation(s)
- Won-Ha Lee
- BK21 Plus KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu, South Korea
| | - Donggun Seo
- BK21 Plus KNU Biomedical Convergence Program, Department of Pharmacology, School of Medicine, Brain Science & Engineering Institute, Kyungpook National University, Daegu, South Korea
| | - Su-Geun Lim
- BK21 Plus KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu, South Korea
| | - Kyoungho Suk
- BK21 Plus KNU Biomedical Convergence Program, Department of Pharmacology, School of Medicine, Brain Science & Engineering Institute, Kyungpook National University, Daegu, South Korea
| |
Collapse
|
33
|
Rothfelder K, Hagelstein I, Roerden M, Blumenstock G, Hofmann M, Nuebling T, Jung G, Salih HR, Dörfel D. Expression of the Immune Checkpoint Modulator OX40 in Acute Lymphoblastic Leukemia Is Associated with BCR-ABL Positivity. Neoplasia 2018; 20:1150-1160. [PMID: 30300827 PMCID: PMC6175778 DOI: 10.1016/j.neo.2018.09.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 09/13/2018] [Accepted: 09/16/2018] [Indexed: 02/07/2023] Open
Abstract
OX40 and its ligand are members of the TNF/TNF receptor superfamily, which includes various molecules influencing cellular signaling and function of both tumor and immune cells. The ability of OX40 to promote proliferation and differentiation of activated T cells fueled present attempts to modulate this immune checkpoint to reinforce antitumor immunity. While we recently found evidence for the involvement of OX40 in pathophysiology of acute myeloid leukemia including natural killer (NK) cell immunosurveillance, less is known on its role in acute lymphoblastic leukemia (ALL). In the present study, OX40 expression on ALL cells was significantly associated with positivity for the adverse risk factor BCR-ABL. In line, signaling via OX40 increased metabolic activity of primary ALL cells and resulted in release of cytokines involved in disease pathophysiology. Furthermore, interaction of ALL-expressed OX40 with its cognate ligand on NK cells stimulated ALL cell lysis. The data presented thus not only identify the yet unknown involvement of OX40/OX40L in ALL pathophysiology and NK cell immunosurveillance but also point to the necessity to thoroughly consider the consequences of modulating the OX40/OX40L molecule system beyond its effects on T cells when developing OX40-targeting approaches for cancer immunotherapy.
Collapse
Affiliation(s)
- Kathrin Rothfelder
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner site Tübingen, Germany
| | - Ilona Hagelstein
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner site Tübingen, Germany
| | - Malte Roerden
- Department of Medical Oncology, Hematology, Immunology, Rheumatology and Pulmonology, University Hospital Tübingen, Tübingen, Germany
| | - Gunnar Blumenstock
- Institute for Clinical Epidemiology and Applied Biometry, Eberhard Karls University, Tübingen, Germany
| | - Martin Hofmann
- Department of Immunology, Eberhard Karls University, Tübingen, Germany
| | - Tina Nuebling
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner site Tübingen, Germany
| | - Gundram Jung
- Department of Immunology, Eberhard Karls University, Tübingen, Germany
| | - Helmut Rainer Salih
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner site Tübingen, Germany; Department of Medical Oncology, Hematology, Immunology, Rheumatology and Pulmonology, University Hospital Tübingen, Tübingen, Germany
| | - Daniela Dörfel
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner site Tübingen, Germany; Department of Medical Oncology, Hematology, Immunology, Rheumatology and Pulmonology, University Hospital Tübingen, Tübingen, Germany.
| |
Collapse
|
34
|
Wallach D. The Tumor Necrosis Factor Family: Family Conventions and Private Idiosyncrasies. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a028431. [PMID: 28847899 DOI: 10.1101/cshperspect.a028431] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The tumor necrosis factor (TNF) cytokine family and the TNF/nerve growth factor (NGF) family of their cognate receptors together control numerous immune functions, as well as tissue-homeostatic and embryonic-development processes. These diverse functions are dictated by both shared and distinct features of family members, and by interactions of some members with nonfamily ligands and coreceptors. The spectra of their activities are further expanded by the occurrence of the ligands and receptors in both membrane-anchored and soluble forms, by "re-anchoring" of soluble forms to extracellular matrix components, and by signaling initiation via intracellular domains (IDs) of both receptors and ligands. Much has been learned about shared features of the receptors as well as of the ligands; however, we still have only limited knowledge of the mechanistic basis for their functional heterogeneity and for the differences between their functions and those of similarly acting cytokines of other families.
Collapse
Affiliation(s)
- David Wallach
- Department of Biomolecular Sciences, The Weizmann Institute of Science, 76100 Rehovot, Israel
| |
Collapse
|
35
|
Han X, Vesely MD. Stimulating T Cells Against Cancer With Agonist Immunostimulatory Monoclonal Antibodies. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 342:1-25. [PMID: 30635089 DOI: 10.1016/bs.ircmb.2018.07.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Elimination of cancer cells through antitumor immunity has been a long-sought after goal since Sir F. Macfarlane Burnet postulated the theory of immune surveillance against tumors in the 1950s. Finally, the use of immunotherapeutics against established cancer is becoming a reality in the past 5years. Most notable are the monoclonal antibodies (mAbs) directed against inhibitory T-cell receptors cytotoxic T lymphocyte antigen-4 and programmed death-1. The next generation of mAbs targeting T cells is designed to stimulate costimulatory receptors on T cells. Here we review the recent progress on these immunostimulatory agonist antibodies against the costimulatory receptors CD137, GITR, OX40, and CD27.
Collapse
Affiliation(s)
- Xue Han
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, United States
| | - Matthew D Vesely
- Department of Dermatology, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
36
|
Tafalla C, Granja AG. Novel Insights on the Regulation of B Cell Functionality by Members of the Tumor Necrosis Factor Superfamily in Jawed Fish. Front Immunol 2018; 9:1285. [PMID: 29930556 PMCID: PMC6001812 DOI: 10.3389/fimmu.2018.01285] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 05/22/2018] [Indexed: 12/12/2022] Open
Abstract
Most ligands and receptors from the tumor necrosis factor (TNF) superfamily play very important roles in the immune system. In particular, many of these molecules are essential in the regulation of B cell biology and B cell-mediated immune responses. Hence, in mammals, it is known that many TNF family members play a key role on B cell development, maturation, homeostasis, activation, and differentiation, also influencing the ability of B cells to present antigens or act as regulators of immune responses. Evolutionarily, jawed fish (including cartilaginous and bony fish) constitute the first animal group in which an adaptive immune response based on B cells and immunoglobulins is present. However, until recently, not much was known about the expression of TNF ligands and receptors in these species. The sequences of many members of the TNF superfamily have been recently identified in different species of jawed fish, thus allowing posterior analysis on the role that these ligands and receptors have on B cell functionality. In this review, we summarize the current knowledge on the impact that the TNF family members have in different aspects of B cell functionality in fish, also providing an in depth comparison with functional aspects of TNF members in mammals, that will permit a further understanding of how B cell functionality is regulated in these distant animal groups.
Collapse
Affiliation(s)
| | - Aitor G Granja
- Animal Health Research Center (CISA-INIA), Madrid, Spain
| |
Collapse
|
37
|
Lei W, Zeng D, Liu G, Zhu Y, Wang J, Wu H, Jiang J, Huang J. Crucial role of OX40/OX40L signaling in a murine model of asthma. Mol Med Rep 2018; 17:4213-4220. [PMID: 29344664 PMCID: PMC5802192 DOI: 10.3892/mmr.2018.8453] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 08/31/2017] [Indexed: 12/15/2022] Open
Abstract
The aim of the present study was to explore the roles of OX40/OX40 ligand (OX40L) signaling and OX40+ T cells in ovalbumin (OVA)-induced mouse asthma model. Asthma was induced by OVA exposure and subsequent co-treatment with OX40L protein, neutralizing anti-OX40L blocking antibody, OX40+ T cells or PBS. The protein expression levels of interleukin (IL)-4, IL-6, IL-13, IL-17, tumor necrosis factor (TNF)-α and interferon (IFN)-γ in bronchoalveolar lavage fluid (BALF) were examined using murine cytokine-specific ELISA. Eosinophil accumulation as well as proliferation and apoptosis of T cells in BALF were detected by Cell Counting kit-8 and flow cytometric assays. Expression of the apoptosis-related protein cleaved caspase-3 was examined in OX40+ T cells using western blot assay. Flow cytometric analysis revealed that OVA-treated mice that were co-treated with OX40L or OX40+ T cells exhibited higher eosinophil infiltration compared with control mice treated only with OVA, whereas neutralizing anti-OX40L blocking antibody inhibited eosinophil infiltration. ELISA assays demonstrated that the expression of IL-4, IL-6, IL-13, IL-17, TNF-α and IFN-γ in BALF in OX40L-treated and OX40+ T cell-treated mice was increased compared with expression levels in control mice. Treatment with OX40L protein effectively reduced apoptosis of T cells and the expression of cleaved caspase-3 in T cells. OX40L-treated and OX40+ T cell-treated mice exhibited increased asthma through OX40/OX40L signaling, which probably promoted inflammatory factor expression, eosinophil infiltration and T cell proliferation.
Collapse
Affiliation(s)
- Wei Lei
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Daxiong Zeng
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Gaoqin Liu
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Yehan Zhu
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Jiajia Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Hongya Wu
- Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Junhong Jiang
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Jianan Huang
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
38
|
Nuebling T, Schumacher CE, Hofmann M, Hagelstein I, Schmiedel BJ, Maurer S, Federmann B, Rothfelder K, Roerden M, Dörfel D, Schneider P, Jung G, Salih HR. The Immune Checkpoint Modulator OX40 and Its Ligand OX40L in NK-Cell Immunosurveillance and Acute Myeloid Leukemia. Cancer Immunol Res 2018; 6:209-221. [PMID: 29321210 DOI: 10.1158/2326-6066.cir-17-0212] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 09/30/2017] [Accepted: 12/21/2017] [Indexed: 11/16/2022]
Abstract
The TNF receptor family member OX40 promotes activation and proliferation of T cells, which fuels efforts to modulate this immune checkpoint to reinforce antitumor immunity. Besides T cells, NK cells are a second cytotoxic lymphocyte subset that contributes to antitumor immunity, particularly in leukemia. Accordingly, these cells are being clinically evaluated for cancer treatment through multiple approaches, such as adoptive transfer of ex vivo expanded polyclonal NK cells (pNKC). Here, we analyzed whether and how OX40 and its ligand (OX40L) influence NK-cell function and antileukemia reactivity. We report that OX40 is expressed on leukemic blasts in a substantial percentage of patients with acute myeloid leukemia (AML) and that OX40 can, after stimulation with agonistic OX40 antibodies, mediate proliferation and release of cytokines that act as growth and survival factors for the leukemic cells. We also demonstrate that pNKC differentially express OX40L, depending on the protocol used for their generation. OX40L signaling promoted NK-cell activation, cytokine production, and cytotoxicity, and disruption of OX40-OX40L interaction impaired pNKC reactivity against primary AML cells. Together, our data implicate OX40/OX40L in disease pathophysiology of AML and in NK-cell immunosurveillance. Our findings indicate that effects of the OX40-OX40L receptor-ligand system in other immune cell subsets and also malignant cells should be taken into account when developing OX40-targeted approaches for cancer immunotherapy. Cancer Immunol Res; 6(2); 209-21. ©2018 AACR.
Collapse
Affiliation(s)
- Tina Nuebling
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Carla Emilia Schumacher
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Hematology and Oncology, Eberhard Karls University, Tuebingen, Germany
| | - Martin Hofmann
- Department of Immunology, Eberhard Karls University, Tuebingen, Germany
| | - Ilona Hagelstein
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Benjamin Joachim Schmiedel
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefanie Maurer
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Birgit Federmann
- Department of Pathology, Eberhard Karls University, Tuebingen, Germany
| | - Kathrin Rothfelder
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Malte Roerden
- Department of Hematology and Oncology, Eberhard Karls University, Tuebingen, Germany
| | - Daniela Dörfel
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Hematology and Oncology, Eberhard Karls University, Tuebingen, Germany
| | | | - Gundram Jung
- Department of Immunology, Eberhard Karls University, Tuebingen, Germany
| | - Helmut Rainer Salih
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany. .,Department of Hematology and Oncology, Eberhard Karls University, Tuebingen, Germany
| |
Collapse
|
39
|
|
40
|
Cortini A, Ellinghaus U, Malik TH, Cunninghame Graham DS, Botto M, Vyse TJ. B cell OX40L supports T follicular helper cell development and contributes to SLE pathogenesis. Ann Rheum Dis 2017; 76:2095-2103. [PMID: 28818832 PMCID: PMC5705841 DOI: 10.1136/annrheumdis-2017-211499] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 08/01/2017] [Indexed: 01/27/2023]
Abstract
Objectives TNFSF4 (encodes OX40L) is a susceptibility locus for systemic lupus erythematosus (SLE). Risk alleles increase TNFSF4 expression in cell lines, but the mechanism linking this effect to disease is unclear, and the OX40L-expressing cell types mediating the risk are not clearly established. Blockade of OX40L has been demonstrated to reduce disease severity in several models of autoimmunity, but not in SLE. We sought to investigate its potential therapeutic role in lupus. Methods We used a conditional knockout mouse system to investigate the function of OX40L on B and T lymphocytes in systemic autoimmunity. Results Physiologically, OX40L on both B and T cells contributed to the humoral immune response, but B cell OX40L supported the secondary humoral response and antibody affinity maturation. Our data also indicated that loss of B cell OX40L impeded the generation of splenic T follicular helper cells. We further show that in two models of SLE—a spontaneous congenic model and the H2-IAbm12 graft-versus-host-induced model—loss of B cell OX40L ameliorates the autoimmune phenotype. This improvement was, in each case, accompanied by a decline in T follicular helper cell numbers. Importantly, the germline knockout did not exhibit a markedly different phenotype from the B cell knockout in these models. Conclusions These findings contribute to a model in which genetically determined increased OX40L expression promotes human SLE by several mechanisms, contingent on its cellular expression. The improvement in pathology in two models of systemic autoimmunity indicates that OX40L is an excellent therapeutic target in SLE.
Collapse
Affiliation(s)
- Andrea Cortini
- Division of Medical and Molecular Genetics and Immunology, Infection and Inflammatory Disease, King's College London, London, UK
| | - Ursula Ellinghaus
- Division of Medical and Molecular Genetics and Immunology, Infection and Inflammatory Disease, King's College London, London, UK
| | - Talat H Malik
- Department of Medicine, Centre for Complement and Inflammation Research, Imperial College London, London, UK
| | - Deborah S Cunninghame Graham
- Division of Medical and Molecular Genetics and Immunology, Infection and Inflammatory Disease, King's College London, London, UK
| | - Marina Botto
- Department of Medicine, Centre for Complement and Inflammation Research, Imperial College London, London, UK
| | - Timothy James Vyse
- Division of Medical and Molecular Genetics and Immunology, Infection and Inflammatory Disease, King's College London, London, UK
| |
Collapse
|
41
|
Peripheral and central immune cell reservoirs in tissues from asymptomatic cats chronically infected with feline immunodeficiency virus. PLoS One 2017; 12:e0175327. [PMID: 28384338 PMCID: PMC5383277 DOI: 10.1371/journal.pone.0175327] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 03/23/2017] [Indexed: 11/21/2022] Open
Abstract
Feline immunodeficiency virus (FIV) infection in cats results in life-long viral persistence and progressive immunopathology. We have previously described a cohort of experimentally infected cats demonstrating a progressive decline of peripheral blood CD4+ T-cell over six years in the face of apparent peripheral viral latency. More recently we reported findings from this same cohort that revealed popliteal lymph node tissue as sites for ongoing viral replication suggesting that tissue reservoirs are important in FIV immunopathogenesis during the late asymptomatic phase of infection. Results reported herein characterize important tissue reservoirs of active viral replication during the late asymptomatic phase by examining biopsied specimens of spleen, mesenteric lymph node (MLN), and intestine from FIV-infected and uninfected control cats. Peripheral blood collected coincident with harvest of tissues demonstrated severe CD4+ T-cell depletion, undetectable plasma viral gag RNA and rarely detectable peripheral blood mononuclear cell (PBMC)-associated viral RNA (vRNA) by real-time PCR. However, vRNA was detectable in all three tissue sites from three of four FIV-infected cats despite the absence of detectable vRNA in plasma. A novel in situ hybridization assay identified B cell lymphoid follicular domains as microanatomical foci of ongoing FIV replication. Additionally, we demonstrated that CD4+ leukocyte depletion in tissues, and CD4+ and CD21+ leukocytes as important cellular reservoirs of ongoing replication. These findings revealed that tissue reservoirs support foci of ongoing viral replication, in spite of highly restricted viral replication in blood. Lentiviral eradication strategies will need address tissue viral reservoirs.
Collapse
|
42
|
Willoughby J, Griffiths J, Tews I, Cragg MS. OX40: Structure and function - What questions remain? Mol Immunol 2017; 83:13-22. [PMID: 28092803 DOI: 10.1016/j.molimm.2017.01.006] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 01/06/2017] [Accepted: 01/07/2017] [Indexed: 02/08/2023]
Abstract
OX40 is a type 1 transmembrane glycoprotein, reported nearly 30 years ago as a cell surface antigen expressed on activated T cells. Since its discovery, it has been validated as a bone fide costimulatory molecule for T cells and member of the TNF receptor family. However, many questions still remain relating to its function on different T cell sub-sets and with recent interest in its utility as a target for antibody-mediated immunotherapy, there is a growing need to gain a better understanding of its biology. Here, we review the expression pattern of OX40 and its ligand, discuss the structure of the receptor:ligand interaction, the downstream signalling it can elicit, its function on different T cell subsets and how antibodies might engage with it to provide effective immunotherapy.
Collapse
Affiliation(s)
- Jane Willoughby
- Antibody & Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, UK
| | - Jordana Griffiths
- Antibody & Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, UK; Biological Sciences, Life Science Building, University of Southampton, Highfield Campus, SO17 1BJ, UK
| | - Ivo Tews
- Biological Sciences, Life Science Building, University of Southampton, Highfield Campus, SO17 1BJ, UK; Institute for life Sciences, University of Southampton, Highfield Campus, SO17 1BJ, UK
| | - Mark S Cragg
- Antibody & Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, UK; Institute for life Sciences, University of Southampton, Highfield Campus, SO17 1BJ, UK.
| |
Collapse
|
43
|
Abstract
The tumour necrosis factor receptor OX40 (CD134) is activated by its cognate ligand OX40L (CD134L, CD252) and functions as a T cell co-stimulatory molecule. OX40-OX40L interactions have been proposed as a potential therapeutic target for treating autoimmunity. OX40 is expressed on activated T cells, and in the mouse at rest on regulatory T cells (Treg). OX40L is found on antigen-presenting cells, activated T cells and others including lymphoid tissue inducer cells, some endothelia and mast cells. Expression of both molecules is increased after antigen presentation occurs and also in response to multiple other pro-inflammatory factors including CD28 ligation, CD40L ligation and interferon-gamma signaling. Their interactions promote T cell survival, promote an effector T cell phenotype, promote T cell memory, tend to reduce regulatory function, increase effector cytokine production and enhance cell mobility. In some circumstances, OX40 agonism may be associated with increased tolerance, although timing with respect to antigenic stimulus is important. Further, recent work has suggested that OX40L blockade may be more effective than OX40 blockade in reducing autoimmunity. This article reviews the expression of OX40 and OX40L in health, the effects of their interactions and insights from their under- or over-expression. We then review OX40 and OX40L expression in human autoimmune disease, identified associations of variations in their genes (TNFRSF4 and TNFSF4, respectively) with autoimmunity, and data from animal models of human diseases. A rationale for blocking OX40-OX40L interaction in human autoimmunity is then presented along with commentary on the one trial of OX40L blockade in human disease conducted to date. Finally, we discuss potential problems with clinical use of OX40-OX40L directed pharmacotherapy.
Collapse
Affiliation(s)
- Gwilym J Webb
- MRC Centre for Immune Regulation, Institute of Biomedical Research, University of Birmingham, Birmingham, West Midlands, B15 2TT, UK. .,National Institute for Health Research Birmingham Liver Biomedical Research Unit, University of Birmingham, Birmingham, West Midlands, B15 2TT, UK.
| | - Gideon M Hirschfield
- National Institute for Health Research Birmingham Liver Biomedical Research Unit, University of Birmingham, Birmingham, West Midlands, B15 2TT, UK
| | - Peter J L Lane
- MRC Centre for Immune Regulation, Institute of Biomedical Research, University of Birmingham, Birmingham, West Midlands, B15 2TT, UK
| |
Collapse
|
44
|
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disorder with complex genetic underpinnings. This review attempts to assemble the myriad of genomic findings to build a clearer picture of the pathobiology of SLE to serve as a guide for therapeutics. Over 100 genes are now known for SLE, and several more penetrant ones have led to the emergence of more defined lupus phenotypes. Also discussed here are the targeted therapies that have come up on the horizon and the specific biologic mechanisms of more traditional therapies which have only recently been explored. The diagnostic toolbox has been enhanced by the addition of new antibodies, gene expression signatures, and mutation panels. This provides an opportunity to piece together the lupus puzzle and even revisit the clinical classification of SLE.
Collapse
|
45
|
Tahiliani V, Hutchinson TE, Abboud G, Croft M, Salek-Ardakani S. OX40 Cooperates with ICOS To Amplify Follicular Th Cell Development and Germinal Center Reactions during Infection. THE JOURNAL OF IMMUNOLOGY 2016; 198:218-228. [PMID: 27895177 DOI: 10.4049/jimmunol.1601356] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 10/31/2016] [Indexed: 11/19/2022]
Abstract
Cognate interactions between T follicular helper (Tfh) cells and B cells are essential for promoting protective Ab responses. Whereas costimulatory receptors such as ICOS are accepted as being important for the induction of Tfh cell fate decision, other molecules may play key roles in amplifying or maintaining the Tfh phenotype. In this study, with vaccinia virus infection in mice, we show that OX40 was expressed on Tfh cells that accumulated at the T/B borders in the white pulp of the spleen and that OX40-dependent signals directly shaped the magnitude and quality of the their response to viral Ags. OX40 deficiency in Tfh cells profoundly impaired the acquisition of germinal center (GC) B cell phenotype, plasma cell generation, and virus-specific Ab responses. Most significantly, we found that sustained interactions between OX40 and its ligand, OX40L, beyond the time of initial encounter with dendritic cells were required for the persistence of high numbers of Tfh and GC B cells. Interestingly, OX40 was coexpressed with ICOS on Tfh cells in and around the GC, and ICOS-ICOSL interactions were similarly crucial at late times for maintenance of the Tfh and GC B cells. Thus, OX40 and ICOS act in a cooperative, nonredundant manner to maximize and prolong the Tfh response that is generated after acute virus infection.
Collapse
Affiliation(s)
- Vikas Tahiliani
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32610; and
| | - Tarun E Hutchinson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32610; and
| | - Georges Abboud
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32610; and
| | - Michael Croft
- Division of Immune Regulation, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | - Shahram Salek-Ardakani
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32610; and
| |
Collapse
|
46
|
Wang Q, Shi BM, Xie F, Fu ZY, Chen YJ, An JN, Ma Y, Liu CP, Zhang XK, Zhang XG. Enhancement of CD4(+) T cell response and survival via coexpressed OX40/OX40L in Graves' disease. Mol Cell Endocrinol 2016; 430:115-24. [PMID: 27107937 DOI: 10.1016/j.mce.2016.04.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Revised: 04/11/2016] [Accepted: 04/19/2016] [Indexed: 12/21/2022]
Abstract
OX40/OX40L pathway plays a very important role in the antigen priming T cells and effector T cells. In the present study, we aimed to examine the involvement of OX40/OX40L pathway in the activation of autoreactive T cells in patients with Grave's disease (GD). We found that OX40 and OX40L were constitutively coexpressed on peripheral CD4(+) T cells from GD patients using flow cytometry analysis. The levels of OX40 and OX40L coexpression on CD4(+) T cells were shown to be correlated with TRAbs. Cell proliferation assay showed that blocking OX40/OX40L signal inhibited T cell proliferation and survival, which suggested that OX40/OX40L could enhance CD4(+) T cell proliferation and maintain their long-term survival in GD by self-enhancing loop of T cell activation independent of APCs. Confocal microscopy and coimmunoprecipitation analysis further revealed that OX40 and OX40L formed a functional complex, which may facilitate signal transduction from OX40L to OX40 and contribute to the pathogenesis of GD.
Collapse
Affiliation(s)
- Qin Wang
- Department of Immunology, Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Bi-Min Shi
- Department of Endocrinology, No. 1 Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Fang Xie
- Department of Pathology, Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Zhao-Yang Fu
- Department of Immunology, Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Yong-Jing Chen
- Department of Immunology, Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Jing-Nan An
- Department of Immunology, Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Yu Ma
- Department of Immunology, Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Cui-Ping Liu
- Clinical Immunology Research Laboratory of Jiangsu Province, No. 1 Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xue-Kun Zhang
- Department of Immunology, Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Xue-Guang Zhang
- Clinical Immunology Research Laboratory of Jiangsu Province, No. 1 Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; Stem Cell Research Laboratory of Jiangsu Province, China.
| |
Collapse
|
47
|
Cognasse F, Chavarin P, Acquart S, Sabido O, Beniguel L, Genin C, Richard Y, Garraud O. Differential Downstream Effects of Cd40 Ligation Mediated by Membrane or Soluble CD40L and Agonistic Ab: A Study on Purified Human B Cells. Int J Immunopathol Pharmacol 2016; 18:65-74. [PMID: 15698512 DOI: 10.1177/039463200501800108] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
With the addition of various cytokines, the CD40-CD40 ligand (CD40L) system can act as a T-helper cell surrogate to permit B lymphocytes to produce large amounts of polyclonal Ig. In the present study, we tested six CD40-CD40L stimulation models: (i, ii) soluble agonistic 89 and G28.5 mAbs; (iii, iv) ‘89’ and ‘G28.5’ bound via their Fc fragments on CDw32-transfected mouse fibroblasts; (v) purified, soluble, trimeric human CD40L molecules (sCD40L); and (vi) human CD40L expressed by a CD40L-transfected mouse fibroblastic cell line (LCD40L). Target B cells consisted of purified blood and tonsillar CD19+ lymphocytes cultured in the presence of CD40 stimuli and IL-2 and IL-10, added at the onset of each B cell culture. A) There was differential expression of CD69, CD80 and CD86 exposure to sCD40L and LCD40L was ensued by the strongest % MFI changes over control. B) In blood B cells, mAbs and sCD40L induced IgA, IgM and IgG production almost equally well; LCD40L proved less efficient. In contrast, in tonsil B cells, LCD40L induced significantly more IgA, IgG 1, IgG3and IgM production than other signals. Using certain CD40/CD40L stimuli to model in vitro Ig production, a system used regularly in many laboratories, may affect the interpretation based on the cell type and on the CD40/CD40L system used.
Collapse
Affiliation(s)
- F Cognasse
- GIMAP-EA 3064, Faculté de Médecine, Université de Saint-Etienne, France
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Di C, Lin X, Zhang Y, Zhong W, Yuan Y, Zhou T, Liu J, Xia Z. Basophil-associated OX40 ligand participates in the initiation of Th2 responses during airway inflammation. J Biol Chem 2015; 290:12523-36. [PMID: 25839234 DOI: 10.1074/jbc.m115.642637] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Indexed: 11/06/2022] Open
Abstract
Asthma is characterized by increased airway submucosal infiltration of T helper (Th) cells and myeloid cells that co-conspire to sustain a chronic inflammation. While recent studies have demonstrated that the myeloid basophils promote Th2 cells in response to various types of allergens, the underlying mechanisms are poorly understood. Here, we found for the first time that in a mouse model of allergic asthma basophils highly expressed OX40 ligand (OX40L) after activation. Interestingly, blockade of OX40-OX40L interaction suppressed basophils-primed Th2 cell differentiation in vitro and ameliorated ovalbumin (OVA)-induced allergic eosinophilic inflammation mediated by Th2 activation. In accordance, the adoptive transfer of basophils derived from mediastinal lymph nodes (MLN) of OVA-immunized mice triggered a robust Th2 response and eosinophilic inflammation in wild-type mice but largely muted in OX40(-/-) mice and mice receiving OX40L-blocked basophils. Taken together, our results reveal a critical role of OX40L presented by the activated basophils to initiate Th2 responses in an allergic asthma model, implicating OX40-OX40L signaling as a potential therapeutic target in the treatment of allergic airway inflammation.
Collapse
Affiliation(s)
- Caixia Di
- From the Department of Pediatrics, Ruijin Hospital affiliated with Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China
| | - Xiaoliang Lin
- From the Department of Pediatrics, Ruijin Hospital affiliated with Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China
| | - Yanjie Zhang
- From the Department of Pediatrics, Ruijin Hospital affiliated with Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China
| | - Wenwei Zhong
- Department of Pediatrics, Shanghai Children's Medical Center affiliated with Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai 200127, China, and
| | - Yufan Yuan
- From the Department of Pediatrics, Ruijin Hospital affiliated with Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China
| | - Tong Zhou
- From the Department of Pediatrics, Ruijin Hospital affiliated with Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China
| | - Junling Liu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Zhenwei Xia
- From the Department of Pediatrics, Ruijin Hospital affiliated with Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China,
| |
Collapse
|
49
|
Abstract
Using the immune system to control cancer has been investigated for over a century. Yet it is only over the last several years that therapeutic agents acting directly on the immune system have demonstrated improved overall survival for cancer patients in phase III clinical trials. Furthermore, it appears that some patients treated with such agents have been cured of metastatic cancer. This has led to increased interest and acceleration in the rate of progress in cancer immunotherapy. Most of the current immunotherapeutic success in cancer treatment is based on the use of immune-modulating antibodies targeting critical checkpoints (CTLA-4 and PD-1/PD-L1). Several other immune-modulating molecules targeting inhibitory or stimulatory pathways are being developed. The combined use of these medicines is the subject of intense investigation and holds important promise. Combination regimens include those that incorporate targeted therapies that act on growth signaling pathways, as well as standard chemotherapy and radiation therapy. In fact, these standard therapies have intrinsic immune-modulating properties that can support antitumor immunity. In the years ahead, adoptive T-cell therapy will also be an important part of treatment for some cancer patients. Other areas which are regaining interest are the use of oncolytic viruses that immunize patients against their own tumors and the use of vaccines against tumor antigens. Immunotherapy has demonstrated unprecedented durability in controlling multiple types of cancer and we expect its use to continue expanding rapidly.
Collapse
|
50
|
Abstract
Optimal T cell response is dependent not only on T cell receptor activation, but also on additional signaling from coreceptors. The main coreceptors include B7 and tumor necrosis factor family members. They exert costimulatory or coinhibitory effects, and their balance determines the fate of T cell response. In normal conditions, costimulators facilitate the development of protective immune response, whereas coinhibitors dampen inflammation to avoid organ/tissue damage from excessive immune reaction. In the tumor microenvironment, the balance is garbled: inhibitory pathways predominate, and T cell response is impaired. The importance of cosignaling in the tumor immune response has been experimentally and clinically demonstrated. New therapeutic strategies targeting T cell cosignaling, especially coinhibitory molecules, are under active experimental and clinical investigation. This review summarizes the functions of main T cell cosignaling axes and discusses their clinical application.
Collapse
|