1
|
Zhang Q, Su C, Luo Y, Zheng F, Liang CL, Chen Y, Liu H, Qiu F, Liu Y, Feng W, Dai Z. Astragalus polysaccharide enhances antitumoral effects of chimeric antigen receptor- engineered (CAR) T cells by increasing CD122 +CXCR3 +PD-1 - memory T cells. Biomed Pharmacother 2024; 179:117401. [PMID: 39243425 DOI: 10.1016/j.biopha.2024.117401] [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: 06/27/2024] [Revised: 08/21/2024] [Accepted: 09/02/2024] [Indexed: 09/09/2024] Open
Abstract
Chimeric antigen receptor-engineered T (CAR-T) cell therapy of cancer has been a hotspot and promising. However, due to rapid exhaustion, CAR-T cells are less effective in solid tumors than in hematological ones. CD122+CXCR3+ memory T cells are characterized with longevity, self-renewal and great antitumoral capacity. Thus, it's compelling to induce memory CAR-T cells to enhance their efficacy on solid tumors. Astragalus polysaccharide (APS) has reportedly exhibited antitumoral effects. However, it's unclear if APS has an impact on CD8+ memory T cell generation or persistence. Using two human cancer cell lines, here we found that APS significantly improved the persistence of GPC3-targeted CAR-T cells and enhanced their suppression of tumor growth in both Huh7 and HepG2 xenograft models of hepatocellular carcinoma. APS increased CD122+/CXCR3+ memory T cells, but decreased their PD-1+ subset within CD8+ CAR-T cells in tumor-bearing mice, while these effects of APS were also confirmed with in vitro experiments. Moreover, APS augmented the expression of chemokines CXCL9/CXCL10 by the tumor in vivo and in vitro. It also enhanced the proliferation and chemotaxis/migration of CAR-T cells in vitro. Finally, APS promoted the phosphorylation of STAT5 in CD8+ CAR-T cells, whereas inhibition of STAT5 activation reversed these in vitro effects of APS. Therefore, APS enhanced the antitumoral effects of CD8+ CAR-T cells by promoting formation/persistence of CD122+/CXCR3+/PD-1- memory T cells and their migration to the tumor.
Collapse
Affiliation(s)
- Qunfang Zhang
- Immunology Program, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Section of Immunology, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China
| | - Chunzhao Su
- Immunology Program, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Yini Luo
- Immunology Program, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Fang Zheng
- Immunology Program, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Chun-Ling Liang
- Immunology Program, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Section of Immunology, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China
| | - Yuchao Chen
- Immunology Program, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Section of Immunology, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China
| | - Huazhen Liu
- Immunology Program, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Section of Immunology, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China
| | - Feifei Qiu
- Immunology Program, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Section of Immunology, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China
| | - Yunshan Liu
- Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Wenxuan Feng
- Immunology Program, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China.
| | - Zhenhua Dai
- Immunology Program, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Section of Immunology, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China.
| |
Collapse
|
2
|
Ceeraz S, Thompson CR, Beatson R, Choy EH. Harnessing CD8 +CD28 - Regulatory T Cells as a Tool to Treat Autoimmune Disease. Cells 2021; 10:2973. [PMID: 34831195 PMCID: PMC8616472 DOI: 10.3390/cells10112973] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 12/30/2022] Open
Abstract
T regulatory cell therapy presents a novel therapeutic strategy for patients with autoimmune diseases or who are undergoing transplantation. At present, the CD4+ Treg population has been extensively characterized, as a result of defined phenotypic and functional readouts. In this review article, we discuss the development and biology of CD8+ Tregs and their role in murine and human disease indications. A subset of CD8+ Tregs that lack the surface expression of CD28 (CD8+CD28- Treg) has proved efficacious in preclinical models. CD8+CD28- Tregs are present in healthy individuals, but their impaired functionality in disease renders them less effective in mediating immunosuppression. We primarily focus on harnessing CD8+ Treg cell therapy in the clinic to support current treatment for patients with autoimmune or inflammatory conditions.
Collapse
Affiliation(s)
| | | | - Richard Beatson
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 9RT, UK;
| | - Ernest H. Choy
- CREATE Centre, Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| |
Collapse
|
3
|
Rocamora-Reverte L, Melzer FL, Würzner R, Weinberger B. The Complex Role of Regulatory T Cells in Immunity and Aging. Front Immunol 2021; 11:616949. [PMID: 33584708 PMCID: PMC7873351 DOI: 10.3389/fimmu.2020.616949] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 12/07/2020] [Indexed: 12/20/2022] Open
Abstract
The immune system is a tightly regulated network which allows the development of defense mechanisms against foreign antigens and tolerance toward self-antigens. Regulatory T cells (Treg) contribute to immune homeostasis by maintaining unresponsiveness to self-antigens and suppressing exaggerated immune responses. Dysregulation of any of these processes can lead to serious consequences. Classically, Treg cell functions have been described in CD4+ T cells, but other immune cells also harbour the capacity to modulate immune responses. Regulatory functions have been described for different CD8+ T cell subsets, as well as other T cells such as γδT cells or NKT cells. In this review we describe the diverse populations of Treg cells and their role in different scenarios. Special attention is paid to the aging process, which is characterized by an altered composition of immune cells. Treg cells can contribute to the development of various age-related diseases but they are poorly characterized in aged individuals. The huge diversity of cells that display immune modulatory functions and the lack of universal markers to identify Treg make the expanding field of Treg research complex and challenging. There are still many open questions that need to be answered to solve the enigma of regulatory T cells.
Collapse
Affiliation(s)
- Lourdes Rocamora-Reverte
- Department of Immunology, Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck, Austria
| | - Franz Leonard Melzer
- Department of Immunology, Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck, Austria
| | - Reinhard Würzner
- Institute of Hygiene & Medical Microbiology, Department of Hygiene, Microbiology and Public Health, Medical University Innsbruck, Innsbruck, Austria
| | - Birgit Weinberger
- Department of Immunology, Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
4
|
Pseudomonas Aeruginosa Induced Cell Death in Acute Lung Injury and Acute Respiratory Distress Syndrome. Int J Mol Sci 2020; 21:ijms21155356. [PMID: 32731491 PMCID: PMC7432812 DOI: 10.3390/ijms21155356] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 07/21/2020] [Accepted: 07/23/2020] [Indexed: 12/27/2022] Open
Abstract
Pseudomonas aeruginosa is an important opportunistic pathogen responsible for the cause of acute lung injury and acute respiratory distress syndrome. P. aeruginosa isthe leading species isolated from patients with nosocomial infection and is detected in almost all the patients with long term ventilation in critical care units. P. aeruginosa infection is also the leading cause of deleterious chronic lung infections in patients suffering from cystic fibrosis as well as the major reason for morbidity in people with chronic obstructive pulmonary disease. P. aeruginosa infections are linked to diseases with high mortality rates and are challenging for treatment, for which no effective remedies have been developed. Massive lung epithelial cell death is a hallmark of severe acute lung injury and acute respiratory distress syndrome caused by P. aeruginosa infection. Lung epithelial cell death poses serious challenges to air barrier and structural integrity that may lead to edema, cytokine secretion, inflammatory infiltration, and hypoxia. Here we review different types of cell death caused by P. aeruginosa serving as a starting point for the diseases it is responsible for causing. We also review the different mechanisms of cell death and potential therapeutics in countering the serious challenges presented by this deadly bacterium.
Collapse
|
5
|
Elizondo CR, Bright JD, Byrne JA, Bright RK. Analysis of the CD8+ IL-10+ T cell response elicited by vaccination with the oncogenic tumor-self protein D52. Hum Vaccin Immunother 2020; 16:1413-1423. [PMID: 31769704 DOI: 10.1080/21645515.2019.1689746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Development of cancer vaccines targeting tumor self-antigens is complex and challenging due to the difficulty of overcoming immune tolerance to self-proteins. Vaccination against tumor self-protein D52 (D52) has been successful, although complete protection appears impaired by immune regulation. Our previous studies suggest that vaccine elicited CD8 + T cells producing interleukin 10 (IL-10) may have a negative impact on tumor protection. Understanding the role CD8+ IL-10 + T cells play in the immune response following vaccination with D52 could result in a more potent vaccine. To address this, we vaccinated IL-10 deficient mice with the murine orthologue of D52; vaccination of wild type (wt) C57BL/6J served as a control for comparison. In separate experiments, D52 vaccinated wt mice were administered IL-10R-specific mAb to neutralize IL-10 function. Interestingly, we observed similar protection against primary tumor challenge in the experimental groups compared to the controls. However, individual IL-10 deficient mice that rejected the primary tumor challenge were re-challenged 140 days post-primary challenge to access vaccine durability and immunologic memory against tumor recurrence. Mice deficient in IL-10 demonstrated a memory response in which 100% of the mice were protected from secondary tumor challenge, while wt mice had diminished recall response (25%) against tumor recurrence. These results with analysis of vaccine-elicited CD8 + T cells for tumor-specific killing and regulatory cell marker expression, add further support to our premise that CD8+ IL-10 + T cells elicited by D52 tumor-self protein vaccine contribute to the suppression of a memory CTL responses and durable tumor immunity.
Collapse
Affiliation(s)
- C Riccay Elizondo
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center , Lubbock, TX, USA
| | - Jennifer D Bright
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center , Lubbock, TX, USA
| | - Jennifer A Byrne
- Faculty of Medicine and Health, The University of Sydney , Westmead, Australia
| | - Robert K Bright
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center , Lubbock, TX, USA.,Cancer Center, Texas Tech University Health Sciences Center , Lubbock, TX, USA
| |
Collapse
|
6
|
Kinoshita M, Nakashima M, Nakashima H, Seki S. Immune Mechanisms Underlying Susceptibility to Endotoxin Shock in Aged Hosts: Implication in Age-Augmented Generalized Shwartzman Reaction. Int J Mol Sci 2019; 20:ijms20133260. [PMID: 31269748 PMCID: PMC6651521 DOI: 10.3390/ijms20133260] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 07/01/2019] [Accepted: 07/01/2019] [Indexed: 01/20/2023] Open
Abstract
In recent decades, the elderly population has been rapidly increasing in many countries. Such patients are susceptible to Gram-negative septic shock, namely endotoxin shock. Mortality due to endotoxin shock remains high despite recent advances in medical care. The generalized Shwartzman reaction is well recognized as an experimental endotoxin shock. Aged mice are similarly susceptible to the generalized Shwartzman reaction and show an increased mortality accompanied by the enhanced production of tumor necrosis factor (TNF). Consistent with the findings in the murine model, the in vitro Shwartzman reaction-like response is also age-dependently augmented in human peripheral blood mononuclear cells, as assessed by enhanced TNF production. Interestingly, age-dependently increased innate lymphocytes with T cell receptor-that intermediate expression, such as that of CD8+CD122+T cells in mice and CD57+T cells in humans, may collaborate with macrophages and induce the exacerbation of the Shwartzman reaction in elderly individuals. However, endotoxin tolerance in mice, which resembles a mirror phenomenon of the generalized Shwartzman reaction, drastically reduces the TNF production of macrophages while strongly activating their bactericidal activity in infection. Importantly, this effect can be induced in aged mice. The safe induction of endotoxin tolerance may be a potential therapeutic strategy for refractory septic shock in elderly patients.
Collapse
Affiliation(s)
- Manabu Kinoshita
- Department of Immunology and Microbiology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan.
| | - Masahiro Nakashima
- Department of Immunology and Microbiology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Hiroyuki Nakashima
- Department of Immunology and Microbiology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Shuhji Seki
- Department of Immunology and Microbiology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan.
| |
Collapse
|
7
|
Chen Q, Ning Z, Wang L, Ying H, Dong S, Zhang C, Shen X, Guo Y, Chen H, Zhu X, Shen Y, Shi W, Hua Y, Wang K, Lin J, Xu L, Chen L, Feng L, Zhang X, Xie J, Sun B, Sun Y, Gu W, Kang M, Tang Z, Chen Z, Chen Z, Liu L, Yu J, Li Z, Meng Z. Is chronic hepatitis B infection a protective factor for the progression of advanced pancreatic ductal adenocarcinoma? An analysis from a large multicenter cohort study. Oncotarget 2018; 7:85603-85612. [PMID: 27811354 PMCID: PMC5356762 DOI: 10.18632/oncotarget.13000] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 07/10/2016] [Indexed: 02/07/2023] Open
Abstract
PURPOSE Whether the progression of advanced pancreatic ductal adenocarcinoma (PDAC) patients could be affected by HBV exposure remains to be determined. Therefore, we conducted this study to assess the effect of HBV infection on PDAC progression among a large cohort in China. METHODS A multicenter cohort study was conducted to explore whether liver metastasis and overall survival in locally advanced and metastatic PDAC could be affected by HBV infection. In this study, we collected 1,526 advanced PDAC patients at three participating hospitals - Shanghai Cancer Center, Changhai Hospital and Ruijin Hospital from 2004 to 2013. The association between HBV status and advanced PDAC progression was then examined. RESULTS In multivariable Logistic regression model, chronic hepatitis B(CHB) infection was inversely associated with synchronous liver metastasis compared to non HBV infection (OR 0.41, 95% CI 0.19-0.85) for stage IV patients. In a multivariable Cox model, CHB infection (HR=0.11, 95% CI 0.02-0.82) is considered as a protective factor of metachronous liver metastasis compared to Non HBV infection for stage III patients. For stage IV patients, CHB infection was inversely associated with overall survival compared to non HBV infection (HR 0.70, 95% CI 0.51-0.95). Inactive carrier(IC) and resolved HBV infection showed no significant association with survival compared to non HBV infection. CONCLUSION This study indicated that CHB infection may serve as an independent factor which decrease synchronous or metachronous liver metastasis, and increase overall survival among advanced PDAC patients.
Collapse
Affiliation(s)
- Qiwen Chen
- Department of Integrative Oncology and Department of Oncology, Fudan University, Shanghai Cancer Center, Shanghai Medical College, Shanghai, China.,Institute of Clinical Epidemiology, Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai, China
| | - Zhouyu Ning
- Department of Integrative Oncology and Department of Oncology, Fudan University, Shanghai Cancer Center, Shanghai Medical College, Shanghai, China
| | - Lei Wang
- Digestive Endoscopy Center, Department of Gastroenterology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Haifeng Ying
- Department of Integrative Medicine of Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shu Dong
- Department of Integrative Oncology and Department of Oncology, Fudan University, Shanghai Cancer Center, Shanghai Medical College, Shanghai, China
| | - Chenyue Zhang
- Department of Integrative Oncology and Department of Oncology, Fudan University, Shanghai Cancer Center, Shanghai Medical College, Shanghai, China
| | - Xiaoheng Shen
- Department of Integrative Medicine of Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuanbiao Guo
- Department of Integrative Medicine of Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hao Chen
- Department of Integrative Oncology and Department of Oncology, Fudan University, Shanghai Cancer Center, Shanghai Medical College, Shanghai, China
| | - Xiaoyan Zhu
- Department of Integrative Oncology and Department of Oncology, Fudan University, Shanghai Cancer Center, Shanghai Medical College, Shanghai, China
| | - Yehua Shen
- Department of Integrative Oncology and Department of Oncology, Fudan University, Shanghai Cancer Center, Shanghai Medical College, Shanghai, China
| | - Weidong Shi
- Department of Integrative Oncology and Department of Oncology, Fudan University, Shanghai Cancer Center, Shanghai Medical College, Shanghai, China
| | - Yongqiang Hua
- Department of Integrative Oncology and Department of Oncology, Fudan University, Shanghai Cancer Center, Shanghai Medical College, Shanghai, China
| | - Kun Wang
- Department of Integrative Oncology and Department of Oncology, Fudan University, Shanghai Cancer Center, Shanghai Medical College, Shanghai, China
| | - Junhua Lin
- Department of Integrative Oncology and Department of Oncology, Fudan University, Shanghai Cancer Center, Shanghai Medical College, Shanghai, China
| | - Litao Xu
- Department of Integrative Oncology and Department of Oncology, Fudan University, Shanghai Cancer Center, Shanghai Medical College, Shanghai, China
| | - Lianyu Chen
- Department of Integrative Oncology and Department of Oncology, Fudan University, Shanghai Cancer Center, Shanghai Medical College, Shanghai, China
| | - Lanyun Feng
- Department of Integrative Oncology and Department of Oncology, Fudan University, Shanghai Cancer Center, Shanghai Medical College, Shanghai, China
| | - Xiumei Zhang
- Department of Integrative Oncology and Department of Oncology, Fudan University, Shanghai Cancer Center, Shanghai Medical College, Shanghai, China
| | - Jing Xie
- Department of Integrative Oncology and Department of Oncology, Fudan University, Shanghai Cancer Center, Shanghai Medical College, Shanghai, China
| | - Bo Sun
- Department of Endoscopy and Department of Oncology, Fudan University, Shanghai Cancer Center, Shanghai Medical College, Shanghai, China
| | - Yaqin Sun
- Department of Radiology and Department of Oncology, Fudan University, Shanghai Cancer Center, Shanghai Medical College, Shanghai, China
| | - Wenchao Gu
- Department of Radiology and Department of Oncology, Fudan University, Shanghai Cancer Center, Shanghai Medical College, Shanghai, China
| | - Mei Kang
- Institute of Clinical Epidemiology, Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai, China
| | - Zheng Tang
- Institute of Clinical Epidemiology, Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai, China
| | - Zhujun Chen
- Department of Clinical Laboratory and Department of Oncology, Fudan University, Shanghai Cancer Center, Shanghai Medical College, Shanghai, China
| | - Zhen Chen
- Department of Integrative Oncology and Department of Oncology, Fudan University, Shanghai Cancer Center, Shanghai Medical College, Shanghai, China
| | - Luming Liu
- Department of Integrative Oncology and Department of Oncology, Fudan University, Shanghai Cancer Center, Shanghai Medical College, Shanghai, China
| | - Jinming Yu
- Institute of Clinical Epidemiology, Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai, China
| | - Zhaoshen Li
- Digestive Endoscopy Center, Department of Gastroenterology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Zhiqiang Meng
- Department of Integrative Oncology and Department of Oncology, Fudan University, Shanghai Cancer Center, Shanghai Medical College, Shanghai, China
| |
Collapse
|
8
|
Abstract
Human stem cell-like memory T (Tscm) cells are long-lived, self-renewing memory lymphocytes that can differentiate into effector cells and mediate strong antitumour response in murine model. The distribution and function of Tscm cells in human lung cancer remain unknown. In this study, we investigated the properties of human Tscm cells in the blood and lymph node of non-small cell lung cancer (NSCLC) patients. There were more CD4 Tscm cells in blood from NSCLC patients than from healthy donors, fewer CD4 and CD8 TSCM cells in blood than in lymph node from NSCLC patients. To further analyze their properties, we stimulated peripheral blood mononuclear cells from NSCLC patients by mitogens to examine cytokine production. Our data suggest that both CD4 and CD8 Tscm cells in blood produced interferon-γ significantly increased in NSCLC patients compare with healthy subjects. In addition, fewer Tscm cells produced interferon-γ in lymph node than in blood from NSCLC patients. Our results strongly suggest that the distribution and function of CD4 Tscm cells in NSCLC patients is upregulated. Understanding of the properties of stem-like memory T cells will supply a good rationale for designing the new adoptive immunotherapy in cancer.
Collapse
|
9
|
Kim PS, Kwilas AR, Xu W, Alter S, Jeng EK, Wong HC, Schlom J, Hodge JW. IL-15 superagonist/IL-15RαSushi-Fc fusion complex (IL-15SA/IL-15RαSu-Fc; ALT-803) markedly enhances specific subpopulations of NK and memory CD8+ T cells, and mediates potent anti-tumor activity against murine breast and colon carcinomas. Oncotarget 2017; 7:16130-45. [PMID: 26910920 PMCID: PMC4941302 DOI: 10.18632/oncotarget.7470] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 02/11/2016] [Indexed: 02/07/2023] Open
Abstract
Interleukin (IL)-15-N72D superagonist-complexed with IL-15RαSushi-Fc fusion protein (IL-15SA/IL-15RαSu-Fc; ALT-803) has been reported to exhibit significant anti-tumor activity in murine myeloma, rat bladder cancer, and murine glioblastoma models. In this study, we examined the immunomodulatory and anti-tumor effects of IL-15SA/IL-15RαSu-Fc in tumor-free and highly metastatic tumor-bearing mice. Here, IL-15SA/IL-15RαSu-Fc significantly expanded natural killer (NK) and CD8+ T cells. In examining NK cell subsets, the greatest significant increase was in highly cytotoxic and migrating (CD11b+, CD27hi; high effector) NK cells, leading to enhanced function on a per-cell basis. CD8+ T cell subset analysis determined that IL-15SA/IL-15RαSu-Fc significantly increased IL-15 responding memory (CD122+, CD44+) CD8+ T cells, in particular those having the innate (NKG2D+, PD1−) phenotype. In 4T1 breast tumor–bearing mice, IL-15SA/IL-15RαSu-Fc induced significant anti-tumor activity against spontaneous pulmonary metastases, depending on CD8+ T and NK cells, and resulting in prolonged survival. Similar anti-tumor activity was seen in the experimental pulmonary metastasis model of CT26 colon carcinoma cells, particularly when IL-15SA/IL-15RαSu-Fc was combined with a cocktail of checkpoint inhibitors, anti-CTLA-4 and anti-PD-L1. Altogether, these studies showed for the first time that IL-15SA/IL-15RαSu-Fc (1) promoted the development of high effector NK cells and CD8+ T cell responders of the innate phenotype, (2) enhanced function of NK cells, and (3) played a vital role in reducing tumor metastasis and ultimately survival, especially in combination with checkpoint inhibitors.
Collapse
Affiliation(s)
- Peter S Kim
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Anna R Kwilas
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Wenxin Xu
- Altor BioScience Corporation, Miramar, FL, USA
| | - Sarah Alter
- Altor BioScience Corporation, Miramar, FL, USA
| | | | - Hing C Wong
- Altor BioScience Corporation, Miramar, FL, USA
| | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - James W Hodge
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
10
|
Brown DL. Immunopathology of the Hepatobiliary System. MOLECULAR AND INTEGRATIVE TOXICOLOGY 2017:329-417. [DOI: 10.1007/978-3-319-47385-7_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
11
|
Fan X, Liu Z, Jin H, Yan J, Liang HP. Alterations of dendritic cells in sepsis: featured role in immunoparalysis. BIOMED RESEARCH INTERNATIONAL 2015; 2015:903720. [PMID: 25821827 PMCID: PMC4363672 DOI: 10.1155/2015/903720] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Revised: 05/25/2014] [Accepted: 07/28/2014] [Indexed: 12/12/2022]
Abstract
Sepsis, the leading cause of mortality in intensive care unit, is characterized by hyperinflammatory response in the early stage and followed by a period of immunosuppression. This immune disorder is believed to be the potent factor that is tightly associated with high mortality in sepsis. Dendritic cells (DCs) serve as professional antigen-presenting cells that play a vital role in immune response by activating T lymphocytes. During the progression of sepsis, DCs have been reported to take part in the aberrant immune response and be necessary for survival. Therefore, a better understanding of the DCs pathology will be undoubtedly beneficial for resolving the problems occurring in sepsis. This review discusses effects of sepsis on DCs number and function, including surface molecules expression, cytokines secretion, and T cell activation, and the underlying mechanism as well as some potential therapeutic strategies.
Collapse
Affiliation(s)
- Xia Fan
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, The Third Military Medical University, Chongqing 400042, China
| | - Zheng Liu
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, The Third Military Medical University, Chongqing 400042, China
| | - He Jin
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, The Third Military Medical University, Chongqing 400042, China
| | - Jun Yan
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, The Third Military Medical University, Chongqing 400042, China
| | - Hua-ping Liang
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, The Third Military Medical University, Chongqing 400042, China
| |
Collapse
|
12
|
Arndt B, Witkowski L, Ellwart J, Seissler J. CD8+ CD122+ PD-1- effector cells promote the development of diabetes in NOD mice. J Leukoc Biol 2014; 97:111-20. [PMID: 25387835 DOI: 10.1189/jlb.3a0613-344rr] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
It is well established that CD4 and CD8 T cells are required for the initiation of autoimmune diabetes in NOD mice. However, different subsets of CD4 or CD8 cells may play different roles in the initiation of insulitis. In this study, we evaluated the role of the previously described CD8(+) CD122(+) in this process. We found that prediabetic NOD mice have an almost 50% reduction of CD8(+) CD122(+) T cells in their secondary lymphoid organs compared with BL/6 or Balb/c mouse strains. This reduction is explained by the lack of the regulatory CD8(+) CD122(+) PD-1(+) cell population in the NOD mice, as we found that all CD8(+) CD122(+) T cells from prediabetic NOD mice lack PD-1 expression and regulatory function. Depletion of CD8(+) CD122(+) PD-1(-) cells through injection of anti-CD122 mAb in prediabetic female NOD mice reduced the infiltration of mononuclear cells into the Langerhans islets and delayed the onset and decreased the incidence of overt diabetes. In addition, we found that transfer of highly purified and activated CD8(+) CD122(+) PD-1(-) cells, together with diabetogenic splenocytes from NOD donors to NOD SCID recipients, accelerates the diabetes development in these mice. Together, these results demonstrate that CD8(+) CD122(+) PD-1(-) T cells from NOD mice are effector cells that are involved in the pathogenesis of autoimmune diabetes.
Collapse
Affiliation(s)
- Börge Arndt
- *Medizinische Klinik und Poliklinik IV, Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, Munich, Germany; Medizinische Klinik und Poliklinik III, Campus Grosshadern, Klinikum der Ludwig-Maximilians-Universität, Munich, Germany; and Helmholtz Zentrum München, Institute of Molecular Immunology (Hämatologikum), Munich, Germany
| | - Lukas Witkowski
- *Medizinische Klinik und Poliklinik IV, Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, Munich, Germany; Medizinische Klinik und Poliklinik III, Campus Grosshadern, Klinikum der Ludwig-Maximilians-Universität, Munich, Germany; and Helmholtz Zentrum München, Institute of Molecular Immunology (Hämatologikum), Munich, Germany
| | - Joachim Ellwart
- *Medizinische Klinik und Poliklinik IV, Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, Munich, Germany; Medizinische Klinik und Poliklinik III, Campus Grosshadern, Klinikum der Ludwig-Maximilians-Universität, Munich, Germany; and Helmholtz Zentrum München, Institute of Molecular Immunology (Hämatologikum), Munich, Germany
| | - Jochen Seissler
- *Medizinische Klinik und Poliklinik IV, Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, Munich, Germany; Medizinische Klinik und Poliklinik III, Campus Grosshadern, Klinikum der Ludwig-Maximilians-Universität, Munich, Germany; and Helmholtz Zentrum München, Institute of Molecular Immunology (Hämatologikum), Munich, Germany
| |
Collapse
|
13
|
Lerret NM, Luo X. IL-15 expanded CD8+CD122+ cells: when do they suppress? Am J Transplant 2014; 14:7-8. [PMID: 24165560 DOI: 10.1111/ajt.12516] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 09/10/2013] [Indexed: 01/25/2023]
Affiliation(s)
- N M Lerret
- Division of Nephrology and Hypertension, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | | |
Collapse
|
14
|
Jagger A, Shimojima Y, Goronzy JJ, Weyand CM. Regulatory T cells and the immune aging process: a mini-review. Gerontology 2013; 60:130-7. [PMID: 24296590 DOI: 10.1159/000355303] [Citation(s) in RCA: 212] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 08/21/2013] [Indexed: 01/02/2023] Open
Abstract
Constant exposure to new and persisting antigens and the need to replace cellular attrition with newly built cells lead to profound remodeling of the immune system after the age of 50 years. The impact of the immunosenescence process varies amongst the different cellular subsets represented within the immune system. Emerging data suggest that progressive aging significantly affects frequencies, subset distribution and functional competence of regulatory T cells (Tregs). Given the central role of Tregs in immune homeostasis, age-related loss of Treg function would be predicted to cause excessive immunity, encountered in elderly humans as a syndrome of chronic, smoldering inflammation as well as the age-related increase in the risk for autoimmunity. Conversely, age-dependent gain of Treg activity would result in failing immunity, such as the rising risk of malignancies and infections amongst the elderly. Emerging data suggest that some Treg populations, specifically naturally occurring Tregs, seem to accumulate with advancing age, whereas inducible Tregs appear to be less available in the older host. More studies are necessary to elucidate functional competence of old Tregs, with an emphasis on comparing the efficacy of young and old Tregs for defined functional domains. Mechanisms of declining Treg inducibility are not understood, but may provide an opportunity for targeted immunomodulation in the elderly. On the horizon is the potential to develop novel therapeutic interventions that target Tregs to make the elderly more efficient in fighting cancers and infections and dampen the risk for senescence-associated inflammation.
Collapse
Affiliation(s)
- Ann Jagger
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, Calif., USA
| | | | | | | |
Collapse
|
15
|
Takigawa T, Miyazaki H, Kinoshita M, Kawarabayashi N, Nishiyama K, Hatsuse K, Ono S, Saitoh D, Seki S, Yamamoto J. Glucocorticoid receptor-dependent immunomodulatory effect of ursodeoxycholic acid on liver lymphocytes in mice. Am J Physiol Gastrointest Liver Physiol 2013; 305:G427-38. [PMID: 23868404 DOI: 10.1152/ajpgi.00205.2012] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Although ursodeoxycholic acid (UDCA) has long been used for patients with chronic cholestatic liver diseases, particularly primary biliary cirrhosis, it may modulate the host immune response. This study investigated the effect of UDCA feeding on experimental hepatitis, endotoxin shock, and bacterial infection in mice. C57BL/6 mice were fed a diet supplemented with or without 0.3% (wt/vol) UDCA for 4 wk. UDCA improved hepatocyte injury and survival in concanavalin-A (Con-A)-induced hepatitis by suppressing IFN-γ production by liver mononuclear cells (MNC), especially NK and NKT cells. UDCA also increased survival after lipopolysaccharide (LPS)-challenge; however, it increased mortality of mice following Escherichia coli infection due to the worsening of infection. UDCA-fed mice showed suppressed serum IL-18 levels and production of IL-18 from liver Kupffer cells, which together with IL-12 potently induce IFN-γ production. However, unlike normal mice, exogenous IL-18 pretreatment did not increase the serum IFN-γ levels after E. coli, LPS, or Con-A challenge in the UDCA-fed mice. Interestingly, however, glucocorticoid receptor (GR) expression was significantly upregulated in the liver MNC of the UDCA-fed mice but not in their whole liver tissue homogenates. Silencing GR in the liver MNC abrogated the suppressive effect of UDCA on LPS- or Con-A-induced IFN-γ production. Furthermore, RU486, a GR antagonist, restored the serum IFN-γ level in UDCA-fed mice after E. coli, LPS, or Con-A challenge. Taken together, these results suggest that IFN-γ-reducing immunomodulatory property of UDCA is mediated by elevated GR in the liver lymphocytes in an IL-12/18-independent manner.
Collapse
Affiliation(s)
- Toshimichi Takigawa
- Dept. of Immunology and Microbiology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513 Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Pagliari D, Cianci R, Frosali S, Landolfi R, Cammarota G, Newton EE, Pandolfi F. The role of IL-15 in gastrointestinal diseases: a bridge between innate and adaptive immune response. Cytokine Growth Factor Rev 2013; 24:455-66. [PMID: 23791986 DOI: 10.1016/j.cytogfr.2013.05.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 05/09/2013] [Accepted: 05/21/2013] [Indexed: 12/27/2022]
Abstract
IL-15 is a member of the IL-2 family of cytokines whose signaling pathways are a bridge between innate and adaptive immune response. IL-15 is part of the intestinal mucosal barrier, and functions to modulate gut homeostasis. IL-15 has pivotal roles in the control of development, proliferation and survival of both innate and adaptive immune cells. IL-15 becomes up-regulated in the inflamed tissue of intestinal inflammatory disease, such as IBD, Celiac Disease and related complications. Indeed, several studies have reported that IL-15 may participate to the pathogenesis of these diseases. Furthermore, although IL-15 seems to be responsible for inflammation and autoimmunity, it also may increase the immune response against cancer. For these reasons, we decided to study the intestinal mucosa as an 'immunological niche', in which immune response, inflammation and local homeostasis are modulated. Understanding the role of the IL-15/IL-15R system will provide a scientific basis for the development of new approaches that use IL-15 for immunotherapy of autoimmune diseases and malignancies. Indeed, a better understanding of the complexity of the mucosal immune system will contribute to the general understanding of immuno-pathology, which could lead to new therapeutical tools for widespread immuno-mediated diseases.
Collapse
Affiliation(s)
- Danilo Pagliari
- Institute of Internal Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
17
|
The Postoperative Serum Interleukin-15 Concentration Correlates with Organ Dysfunction and the Prognosis of Septic Patients Following Emergency Gastrointestinal Surgery. J Surg Res 2012; 175:e83-8. [DOI: 10.1016/j.jss.2011.12.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Revised: 11/29/2011] [Accepted: 12/01/2011] [Indexed: 02/02/2023]
|
18
|
The immunologic outcome of enhanced function of mouse liver lymphocytes and Kupffer cells by high-fat and high-cholesterol diet. Shock 2012; 36:484-93. [PMID: 21937954 DOI: 10.1097/shk.0b013e31822dc6e4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Dietary lipids/cholesterol may modulate liver immune function. We have recently found that mouse F4/80 Kupffer cells are classified into phagocytic CD68 Kupffer cells and cytokine-producing CD11b Kupffer cells. We here investigate how a high-fat and/or high-cholesterol diet affects innate immune liver mononuclear cells. For 4 weeks, C57BL/6 mice were fed a high-fat and high-cholesterol diet (HFCD), a high-cholesterol diet (HCD), a high-fat diet (HFD), or a control diet (CD). High-fat and high-cholesterol diet and HCD increased liver cholesterol levels; serum cholesterol levels increased in HFCD and HFD mice but not in HCD mice. The increased proportion of natural killer (NK) cells, downregulated NK1.1 expression of natural killer T cells, and enhanced CD69 and IL-12 receptor β mRNA expression of liver lymphocytes indicate the activation of them by HFCD. IL-12 production from Kupffer cells and interferon γ production from NK/natural killer T cells activated by LPS and/or IL-12 both increased. IL-12 pretreatment more effectively improved the survival of HFCD mice relative to the survival of CD mice upon injections of liver metastatic EL-4 cells. In contrast, HFCD mouse survival decreased after LPS injection and generalized Shwartzman reaction. Consistently in HFCD mice, Toll-like receptor 4 mRNA expression of whole Kupffer cells was upregulated, and CD11b Kupffer cells proportionally increased. Although the proportion of CD68 Kupffer cells decreased in HFCD mice, phagocytic activity of them was enhanced. Mice fed with HCD rather than those fed with HFD showed features closer to HFCD mice. Thus, enhanced function of mouse liver mononuclear cells is likely dependent on the liver cholesterol level, rather than the liver triglyceride level.
Collapse
|
19
|
Herndler-Brandstetter D, Landgraf K, Tzankov A, Jenewein B, Brunauer R, Laschober GT, Parson W, Kloss F, Gassner R, Lepperdinger G, Grubeck-Loebenstein B. The impact of aging on memory T cell phenotype and function in the human bone marrow. J Leukoc Biol 2011; 91:197-205. [PMID: 22013229 DOI: 10.1189/jlb.0611299] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Recently, the BM has been shown to play a key role in regulating the survival and function of memory T cells. However, the impact of aging on these processes has not yet been studied. We demonstrate that the number of CD4⁺ and CD8⁺ T cells in the BM is maintained during aging. However, the composition of the T cell pool in the aged BM is altered with a decline of naïve and an increase in T(EM) cells. In contrast to the PB, a highly activated CD8⁺CD28⁻ T cell population, which lacks the late differentiation marker CD57, accumulates in the BM of elderly persons. IL-6 and IL-15, which are both increased in the aged BM, efficiently induce the activation, proliferation, and differentiation of CD8⁺ T cells in vitro, highlighting a role of these cytokines in the age-dependent accumulation of highly activated CD8⁺CD28⁻ T cells in the BM. Yet, these age-related changes do not impair the maintenance of a high number of polyfunctional memory CD4⁺ and CD8⁺ T cells in the BM of elderly persons. In summary, aging leads to the accumulation of a highly activated CD8⁺CD28⁻ T cell population in the BM, which is driven by the age-related increase of IL-6 and IL-15. Despite these changes, the aged BM is a rich source of polyfunctional memory T cells and may thus represent an important line of defense to fight recurrent infections in old age.
Collapse
|
20
|
Inoue S, Unsinger J, Davis CG, Muenzer JT, Ferguson TA, Chang K, Osborne DF, Clark AT, Coopersmith CM, McDunn JE, Hotchkiss RS. IL-15 prevents apoptosis, reverses innate and adaptive immune dysfunction, and improves survival in sepsis. THE JOURNAL OF IMMUNOLOGY 2009; 184:1401-9. [PMID: 20026737 DOI: 10.4049/jimmunol.0902307] [Citation(s) in RCA: 162] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
IL-15 is a pluripotent antiapoptotic cytokine that signals to cells of both the innate and adaptive immune system and is regarded as a highly promising immunomodulatory agent in cancer therapy. Sepsis is a lethal condition in which apoptosis-induced depletion of immune cells and subsequent immunosuppression are thought to contribute to morbidity and mortality. This study tested the ability of IL-15 to block apoptosis, prevent immunosuppression, and improve survival in sepsis. Mice were made septic using cecal ligation and puncture or Pseudomonas aeruginosa pneumonia. The experiments comprised a 2 x 2 full factorial design with surgical sepsis versus sham and IL-15 versus vehicle. In addition to survival studies, splenic cellularity, canonical markers of activation and proliferation, intracellular pro- and antiapoptotic Bcl-2 family protein expression, and markers of immune cell apoptosis were evaluated by flow cytometry. Cytokine production was examined both in plasma of treated mice and splenocytes that were stimulated ex vivo. IL-15 blocked sepsis-induced apoptosis of NK cells, dendritic cells, and CD8 T cells. IL-15 also decreased sepsis-induced gut epithelial apoptosis. IL-15 therapy increased the abundance of antiapoptotic Bcl-2 while decreasing proapoptotic Bim and PUMA. IL-15 increased both circulating IFN-gamma, as well as the percentage of NK cells that produced IFN-gamma. Finally, IL-15 increased survival in both cecal ligation and puncture and P. aeruginosa pneumonia. In conclusion, IL-15 prevents two immunopathologic hallmarks of sepsis, namely, apoptosis and immunosuppression, and improves survival in two different models of sepsis. IL-15 represents a potentially novel therapy of this highly lethal disorder.
Collapse
Affiliation(s)
- Shigeaki Inoue
- Department of Anesthesiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|