1
|
Srivastava S, Tyagi A, Pawar VA, Khan NH, Arora K, Verma C, Kumar V. Revolutionizing Immunotherapy: Unveiling New Horizons, Confronting Challenges, and Navigating Therapeutic Frontiers in CAR-T Cell-Based Gene Therapies. Immunotargets Ther 2024; 13:413-433. [PMID: 39219644 PMCID: PMC11365499 DOI: 10.2147/itt.s474659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
The CAR-T cell therapy has marked the dawn of new era in the cancer therapeutics and cell engineering techniques. The review emphasizes on the challenges that obstruct the therapeutic efficiency caused by cell toxicities, immunosuppressive tumor environment, and decreased T cell infiltration. In the interest of achieving the overall survival (OS) and event-free survival (EFS) of patients, the conceptual background of potential target selection and various CAR-T cell design techniques are described which can minimize the off-target effects, reduce toxicity, and thus increase the resilience of CAR-T cell treatment in the haematological malignancies as well as in solid tumors. Furthermore, it delves into cutting-edge technologies like gene editing and synthetic biology, providing new opportunities to enhance the functionality of CAR-T cells and overcome mechanisms of immune evasion. This review provides a comprehensive understanding of the complex and diverse aspects of CAR-T cell-based gene treatments, including both scientific and clinical aspects. By effectively addressing the obstacles and utilizing the capabilities of cutting-edge technology, CAR-T cell therapy shows potential in fundamentally changing immunotherapy and reshaping the approach to cancer treatment.
Collapse
Affiliation(s)
- Shivani Srivastava
- Department of Pathology, School of Medicine, Yale University, New Haven, CT, USA
| | - Anuradha Tyagi
- Department of cBRN, Institute of Nuclear Medicine and Allied Science, Delhi, India
| | | | - Nawaid Hussain Khan
- Faculty of Medicine, Ala-Too International University, Bishkek, Kyrgyz Republic
| | - Kavita Arora
- Advanced Instrumentation Research Facility, Jawaharlal Nehru University, New Delhi, India
- School of Computational & Integrative Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Chaitenya Verma
- Department of Pathology, Wexner Medical Center, Ohio State University, Columbus, OH, USA
- Department of Biotechnology, SSET, Sharda University, Greater Noida, 201306, India
| | - Vinay Kumar
- Pennsylvania State University Hershey Medical Center, 500 University Dr, Heshey, PA, USA
| |
Collapse
|
2
|
Petrovic A, Jovanovic I, Stojanovic B, Dimitrijevic Stojanovic M, Stojanovic BS, Jurisevic M, Simovic Markovic B, Jovanovic M, Jovanovic M, Jovanovic M, Gajovic N. Harnessing Metformin's Immunomodulatory Effects on Immune Cells to Combat Breast Cancer. Int J Mol Sci 2024; 25:5869. [PMID: 38892058 PMCID: PMC11172298 DOI: 10.3390/ijms25115869] [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: 04/13/2024] [Revised: 05/12/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Metformin, a medication known for its anti-glycemic properties, also demonstrates potent immune system activation. In our study, using a 4T1 breast cancer model in BALB/C WT mice, we examined metformin's impact on the functional phenotype of multiple immune cells, with a specific emphasis on natural killer T (NKT) cells due to their understudied role in this context. Metformin administration delayed the appearance and growth of carcinoma. Furthermore, metformin increased the percentage of IFN-γ+ NKT cells, and enhanced CD107a expression, as measured by MFI, while decreasing PD-1+, FoxP3+, and IL-10+ NKT cells in spleens of metformin-treated mice. In primary tumors, metformin increased the percentage of NKp46+ NKT cells and increased FasL expression, while lowering the percentages of FoxP3+, PD-1+, and IL-10-producing NKT cells and KLRG1 expression. Activation markers increased, and immunosuppressive markers declined in T cells from both the spleen and tumors. Furthermore, metformin decreased IL-10+ and FoxP3+ Tregs, along with Gr-1+ myeloid-derived suppressor cells (MDSCs) in spleens, and in tumor tissue, it decreased IL-10+ and FoxP3+ Tregs, Gr-1+, NF-κB+, and iNOS+ MDSCs, and iNOS+ dendritic cells (DCs), while increasing the DCs quantity. Additionally, increased expression levels of MIP1a, STAT4, and NFAT in splenocytes were found. These comprehensive findings illustrate metformin's broad immunomodulatory impact across a variety of immune cells, including stimulating NKT cells and T cells, while inhibiting Tregs and MDSCs. This dynamic modulation may potentiate its use in cancer immunotherapy, highlighting its potential to modulate the tumor microenvironment across a spectrum of immune cell types.
Collapse
Affiliation(s)
- Andjela Petrovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia
| | - Ivan Jovanovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia
| | - Bojan Stojanovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia
| | - Milica Dimitrijevic Stojanovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia
- Department of Pathology, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia
| | - Bojana S Stojanovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia
- Department of Pathophysiology, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia
| | - Milena Jurisevic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia
| | - Bojana Simovic Markovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia
| | - Marina Jovanovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia
- Department of Otorhinolaryngology, Faculty of Medical Sciences, Svetozara Markovica 69, 34000 Kragujevac, Serbia
| | - Milan Jovanovic
- Department of Abdominal Surgery, Military Medical Academy, University of Defence, Crnotravska 17, 11000 Belgrade, Serbia
| | - Mihailo Jovanovic
- Clinic for Orthopaedics and Traumatology, University Clinical Center, Zmaj Jovina 30, 34000 Kragujevac, Serbia
| | - Nevena Gajovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia
| |
Collapse
|
3
|
Zhang H, Chen S, Zhang Y, Tian C, Pan J, Wang Y, Bai S, Wu Q, Su M, Xie D, Fu S, Li S, Zhang J, Chen Y, Zhu S, Qian Y, Bai L. Antigen Priming Induces Functional Reprogramming in iNKT Cells via Metabolic and Epigenetic Regulation: An Insight into iNKT Cell-Based Antitumor Immunotherapy. Cancer Immunol Res 2023; 11:1598-1610. [PMID: 37756568 DOI: 10.1158/2326-6066.cir-23-0448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/04/2023] [Accepted: 09/20/2023] [Indexed: 09/29/2023]
Abstract
Dysfunction of intratumoral invariant natural killer T (iNKT) cells hinders their antitumor efficacy, but the underlying mechanisms and the relationship with endogenous antigen priming remain to be explored. Here, we report that antigen priming leads to metabolic reprogramming and epigenetic remodeling, which causes functional reprogramming in iNKT cells, characterized by limited cytokine responses upon restimulation but constitutive high cytotoxicity. Mechanistically, impaired oxidative phosphorylation (OXPHOS) in antigen-primed iNKT cells inhibited T-cell receptor signaling, as well as elevation of glycolysis, upon restimulation via reducing mTORC1 activation, and thus led to impaired cytokine production. However, the metabolic reprogramming in antigen-primed iNKT cells was uncoupled with their enhanced cytotoxicity; instead, epigenetic remodeling explained their high expression of granzymes. Notably, intratumoral iNKT cells shared similar metabolic reprogramming and functional reprogramming with antigen-primed iNKT cells due to endogenous antigen priming in tumors, and thus recovery of OXPHOS in intratumoral iNKT cells by ZLN005 successfully enhanced their antitumor responses. Our study deciphers the influences of antigen priming-induced metabolic reprogramming and epigenetic remodeling on functionality of intratumoral iNKT cells, and proposes a way to enhance efficacy of iNKT cell-based antitumor immunotherapy by targeting cellular metabolism.
Collapse
Affiliation(s)
- Huimin Zhang
- Center for Advanced Interdisciplinary Science and Biomedicine, Institute of Health and Medicine, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Sanwei Chen
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuwei Zhang
- Center for Advanced Interdisciplinary Science and Biomedicine, Institute of Health and Medicine, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Chenxi Tian
- Center for Advanced Interdisciplinary Science and Biomedicine, Institute of Health and Medicine, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jun Pan
- Center for Advanced Interdisciplinary Science and Biomedicine, Institute of Health and Medicine, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yu Wang
- Center for Advanced Interdisciplinary Science and Biomedicine, Institute of Health and Medicine, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Shiyu Bai
- Center for Advanced Interdisciplinary Science and Biomedicine, Institute of Health and Medicine, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qielan Wu
- Center for Advanced Interdisciplinary Science and Biomedicine, Institute of Health and Medicine, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Miya Su
- Center for Advanced Interdisciplinary Science and Biomedicine, Institute of Health and Medicine, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Di Xie
- Center for Advanced Interdisciplinary Science and Biomedicine, Institute of Health and Medicine, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Sicheng Fu
- Center for Advanced Interdisciplinary Science and Biomedicine, Institute of Health and Medicine, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Shuhang Li
- Center for Advanced Interdisciplinary Science and Biomedicine, Institute of Health and Medicine, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jing Zhang
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China
| | - Yusheng Chen
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China
| | - Shasha Zhu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- National Health Commission Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Hefei, Anhui, China
| | - Yeben Qian
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Li Bai
- Center for Advanced Interdisciplinary Science and Biomedicine, Institute of Health and Medicine, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China
- National Synchrotron Radiation Laboratory, University of Science and Technology of China, Hefei, China
| |
Collapse
|
4
|
Li Z, Wang Q, Huang X, Yang M, Zhou S, Li Z, Fang Z, Tang Y, Chen Q, Hou H, Li L, Fei F, Wang Q, Wu Y, Gong A. Lactate in the tumor microenvironment: A rising star for targeted tumor therapy. Front Nutr 2023; 10:1113739. [PMID: 36875841 PMCID: PMC9978120 DOI: 10.3389/fnut.2023.1113739] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 01/30/2023] [Indexed: 02/18/2023] Open
Abstract
Metabolic reprogramming is one of fourteen hallmarks of tumor cells, among which aerobic glycolysis, often known as the "Warburg effect," is essential to the fast proliferation and aggressive metastasis of tumor cells. Lactate, on the other hand, as a ubiquitous molecule in the tumor microenvironment (TME), is generated primarily by tumor cells undergoing glycolysis. To prevent intracellular acidification, malignant cells often remove lactate along with H+, yet the acidification of TME is inevitable. Not only does the highly concentrated lactate within the TME serve as a substrate to supply energy to the malignant cells, but it also works as a signal to activate multiple pathways that enhance tumor metastasis and invasion, intratumoral angiogenesis, as well as immune escape. In this review, we aim to discuss the latest findings on lactate metabolism in tumor cells, particularly the capacity of extracellular lactate to influence cells in the tumor microenvironment. In addition, we examine current treatment techniques employing existing medications that target and interfere with lactate generation and transport in cancer therapy. New research shows that targeting lactate metabolism, lactate-regulated cells, and lactate action pathways are viable cancer therapy strategies.
Collapse
Affiliation(s)
- Zhangzuo Li
- Hematological Disease Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China.,Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Qi Wang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Xufeng Huang
- Faculty of Dentistry, University of Debrecen, Debrecen, Hungary
| | - Mengting Yang
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Shujing Zhou
- Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zhengrui Li
- School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China.,National Center for Stomatology and National Clinical Research Center for Oral Diseases, Shanghai, China.,Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Zhengzou Fang
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yidan Tang
- Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Qian Chen
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Hanjin Hou
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Li Li
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Fei Fei
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Qiaowei Wang
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yuqing Wu
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Aihua Gong
- Hematological Disease Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China.,Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
5
|
Qin Y, Xu G. Enhancing CAR T-cell therapies against solid tumors: Mechanisms and reversion of resistance. Front Immunol 2022; 13:1053120. [PMID: 36569859 PMCID: PMC9773088 DOI: 10.3389/fimmu.2022.1053120] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 11/21/2022] [Indexed: 12/13/2022] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy, belonging to adoptive immune cells therapy, utilizes engineered immunoreceptors to enhance tumor-specific killing. By now new generations of CAR T-cell therapies dramatically promote the effectiveness and robustness in leukemia cases. However, only a few CAR T-cell therapies gain FDA approval till now, which are applied to hematologic cancers. Targeting solid tumors through CAR T-cell therapies still faces many problems, such as tumor heterogeneity, antigen loss, infiltration inability and immunosuppressive micro-environment. Recent advances provide new insights about the mechanisms of CAR T-cell therapy resistance and give rise to potential reversal therapies. In this review, we mainly introduce existing barriers when treating solid tumors with CAR T-cells and discuss the methods to overcome these challenges.
Collapse
Affiliation(s)
- Yue Qin
- National Institute of Biological Sciences, Beijing, China,Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Guotai Xu
- National Institute of Biological Sciences, Beijing, China,Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China,*Correspondence: Guotai Xu,
| |
Collapse
|
6
|
Zimmerer JM, Ringwald BA, Chaudhari SR, Han J, Peterson CM, Warren RT, Hart MM, Abdel-Rasoul M, Bumgardner GL. Invariant NKT Cells Promote the Development of Highly Cytotoxic Multipotent CXCR3 +CCR4 +CD8 + T Cells That Mediate Rapid Hepatocyte Allograft Rejection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 207:3107-3121. [PMID: 34810223 PMCID: PMC9124232 DOI: 10.4049/jimmunol.2100334] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 10/13/2021] [Indexed: 12/22/2022]
Abstract
Hepatocyte transplant represents a treatment for metabolic disorders but is limited by immunogenicity. Our prior work identified the critical role of CD8+ T cells, with or without CD4+ T cell help, in mediating hepatocyte rejection. In this study, we evaluated the influence of invariant NKT (iNKT) cells, uniquely abundant in the liver, upon CD8-mediated immune responses in the presence and absence of CD4+ T cells. To investigate this, C57BL/6 (wild-type) and iNKT-deficient Jα18 knockout mice (cohorts CD4 depleted) were transplanted with allogeneic hepatocytes. Recipients were evaluated for alloprimed CD8+ T cell subset composition, allocytotoxicity, and hepatocyte rejection. We found that CD8-mediated allocytotoxicity was significantly decreased in iNKT-deficient recipients and was restored by adoptive transfer of iNKT cells. In the absence of both iNKT cells and CD4+ T cells, CD8-mediated allocytotoxicity and hepatocyte rejection was abrogated. iNKT cells enhance the proportion of a novel subset of multipotent, alloprimed CXCR3+CCR4+CD8+ cytolytic T cells that develop after hepatocyte transplant and are abundant in the liver. Alloprimed CXCR3+CCR4+CD8+ T cells express cytotoxic effector molecules (perforin/granzyme and Fas ligand) and are distinguished from alloprimed CXCR3+CCR4-CD8+ T cells by a higher proportion of cells expressing TNF-α and IFN-γ. Furthermore, alloprimed CXCR3+CCR4+CD8+ T cells mediate higher allocytotoxicity and more rapid allograft rejection. Our data demonstrate the important role of iNKT cells in promoting the development of highly cytotoxic, multipotent CXCR3+CCR4+CD8+ T cells that mediate rapid rejection of allogeneic hepatocytes engrafted in the liver. Targeting iNKT cells may be an efficacious therapy to prevent rejection of intrahepatic cellular transplants.
Collapse
Affiliation(s)
- Jason M Zimmerer
- Comprehensive Transplant Center, Department of Surgery, The Ohio State University College of Medicine, Columbus, OH
| | - Bryce A Ringwald
- Medical Student Research Program, The Ohio State University College of Medicine, Columbus, OH
| | - Sachi R Chaudhari
- Comprehensive Transplant Center, Department of Surgery, The Ohio State University College of Medicine, Columbus, OH
| | - Jing Han
- Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, OH; and
| | - Chelsea M Peterson
- Comprehensive Transplant Center, Department of Surgery, The Ohio State University College of Medicine, Columbus, OH
| | - Robert T Warren
- Comprehensive Transplant Center, Department of Surgery, The Ohio State University College of Medicine, Columbus, OH
| | - Madison M Hart
- Comprehensive Transplant Center, Department of Surgery, The Ohio State University College of Medicine, Columbus, OH
| | | | - Ginny L Bumgardner
- Comprehensive Transplant Center, Department of Surgery, The Ohio State University College of Medicine, Columbus, OH;
| |
Collapse
|
7
|
Kong R, Wang N, Han W, Bao W, Lu J. IFNγ-mediated repression of system xc - drives vulnerability to induced ferroptosis in hepatocellular carcinoma cells. J Leukoc Biol 2021; 110:301-314. [PMID: 34318944 DOI: 10.1002/jlb.3ma1220-815rrr] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 06/10/2021] [Accepted: 06/14/2021] [Indexed: 12/22/2022] Open
Abstract
IFNγ released from CD8+ T cells or natural killer cells plays a crucial role in antitumor host immunity. Several studies have found that IFNγ is involved in regulating tumor cell proliferation and apoptosis. However, few studies have examined its role in cell ferroptosis. Here, we found that IFNγ treatment enhanced glutathione depletion, promoted cell cycle arrested in G0/G1 phase, increased lipid peroxidation, and sensitized cells to ferroptosis activators. Additionally, IFNγ down-regulated the mRNA and protein levels of SLC3A2 and SLC7A11, two subunits of the glutamate-cystine antiporter system xc- via activating the JAK/STAT pathway in hepatocellular carcinoma (HCC) cell lines. Furthermore, IFNγ increased reactive oxygen species levels and decreased mitochondiral membrane potential in Bel7402 and HepG2 cells. These changes were accompanied by decreased system xc- activity. Cancer cells exposed to TGFβ1 for 48 h showed sensitization to IFNγ + erastin-induced ferroptosis, with decreased system xc- expression. In conclusion, IFNγ repressed system xc- activation via activating JAK/STAT signaling. Additionally, enhanced lipid peroxidation was associated with altered mitochondrial function in HCC cells. Our findings identified a role for IFNγ in sensitizing HCC cells to ferroptosis, which provided new insights for applying IFNγ as a cancer treatment.
Collapse
Affiliation(s)
- Rui Kong
- Department of Gastroenterology, Shanghai Tenth People's Hospital Affiliated to Tongji University, Tongji University, School of Medicine, Shanghai, China
| | - Nan Wang
- Department of Gastroenterology, Shanghai Tenth People's Hospital Affiliated to Tongji University, Tongji University, School of Medicine, Shanghai, China
| | - Wei Han
- Department of Gastroenterology, Shanghai Tenth People's Hospital Affiliated to Tongji University, Tongji University, School of Medicine, Shanghai, China
| | - Wen Bao
- Department of Gastroenterology, Shanghai Tenth People's Hospital Affiliated to Tongji University, Tongji University, School of Medicine, Shanghai, China
| | - Jie Lu
- Department of Gastroenterology, Shanghai Tenth People's Hospital Affiliated to Tongji University, Tongji University, School of Medicine, Shanghai, China
| |
Collapse
|
8
|
Gocher AM, Workman CJ, Vignali DAA. Interferon-γ: teammate or opponent in the tumour microenvironment? Nat Rev Immunol 2021; 22:158-172. [PMID: 34155388 DOI: 10.1038/s41577-021-00566-3] [Citation(s) in RCA: 255] [Impact Index Per Article: 85.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2021] [Indexed: 02/06/2023]
Abstract
Cancer immunotherapy offers substantive benefit to patients with various tumour types, in some cases leading to complete tumour clearance. However, many patients do not respond to immunotherapy, galvanizing the field to define the mechanisms of pre-existing and acquired resistance. Interferon-γ (IFNγ) is a cytokine that has both protumour and antitumour activities, suggesting that it may serve as a nexus for responsiveness to immunotherapy. Many cancer immunotherapies and chemotherapies induce IFNγ production by various cell types, including activated T cells and natural killer cells. Patients resistant to these therapies commonly have molecular aberrations in the IFNγ signalling pathway or express resistance molecules driven by IFNγ. Given that all nucleated cells can respond to IFNγ, the functional consequences of IFNγ production need to be carefully dissected on a cell-by-cell basis. Here, we review the cells that produce IFNγ and the different effects of IFNγ in the tumour microenvironment, highlighting the pleiotropic nature of this multifunctional and abundant cytokine.
Collapse
Affiliation(s)
- Angela M Gocher
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Creg J Workman
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Dario A A Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA. .,Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA. .,Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
| |
Collapse
|
9
|
Serum IL-5 and IFN- γ Are Novel Predictive Biomarkers for Anti-PD-1 Treatment in NSCLC and GC Patients. DISEASE MARKERS 2021; 2021:5526885. [PMID: 34239620 PMCID: PMC8235970 DOI: 10.1155/2021/5526885] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 05/20/2021] [Indexed: 11/17/2022]
Abstract
Background Because responses of patients with cancer to immunotherapy can vary in success, effective biomarkers are urgently needed for predicting clinical response with anti-PD-1 treatment. We aimed to evaluate the IL-5 and IFN-γ level with the response of anti-PD-1 blockade in non-small-cell lung cancer (NSCLC) and gastric cancer (GC). Methods Metastatic NSCLC and GC patients treated with anti-PD-1 monoclonal antibody were studied. Blood samples were taken before PD-1 McAb treatment, after the first cycle treatment, and during efficacy evaluation. The association between IL-5 and IFN-γ levels and clinical response were analyzed by the nonparametric Wilcoxon matched-pairs ranked tests. The progression-free survival (PFS) time was obtained by imaging evaluation and telephone follow-up of all the patients. Kaplan-Meier and the log rank test were used to plot the survival curve. Results IL-5 and IFN-γ levels were detected in the peripheral blood of 40 NSCLC and 35 GC patients who have received anti-PD-1 treatment. In effective group, IL-5 and IFN-γ levels at best response points significantly decreased (P < 0.001) compared with pretherapeutic levels in NSCLC and GC patients with lymph node or distant metastasis. Compared with pretherapeutic levels, IL-5 and IFN-γ levels largely increased as the tumor progresses (P < 0.01). Higher IL-5 and IFN-γ levels before treatment indicated shorter progression-free survival in patients with NSCLC metastasis (P = 0.007, P = 0.0111). Moreover, their levels also accurately reflected the pseudoprogression of two NSCLC patients to anti-PD-1 treatment. Conclusions Our results suggested that serum IL-5 and IFN-γ levels could be an effective indicator for predicting clinical efficacy and survival with anti-PD-1 blockade in NSCLC and GC patients.
Collapse
|
10
|
Expansion and CD2/CD3/CD28 stimulation enhance Th2 cytokine secretion of human invariant NKT cells with retained anti-tumor cytotoxicity. Cytotherapy 2020; 22:276-290. [PMID: 32238299 DOI: 10.1016/j.jcyt.2020.01.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 01/14/2020] [Accepted: 01/22/2020] [Indexed: 11/21/2022]
Abstract
BACKGROUND AIMS Key obstacles in human iNKT cell translational research and immunotherapy include the lack of robust protocols for dependable expansion of human iNKT cells and the paucity of data on phenotypes in post-expanded cells. METHODS We delineate expansion methods using interleukin (IL)-2, IL-7 and allogeneic feeder cells and anti-CD2/CD3/CD28 stimulation by which to dependably augment Th2 polarization and direct cytotoxicity of human peripheral blood CD3+Vα24+Vβ11+ iNKT cells. RESULTS Gene and protein expression profiling demonstrated augmented Th2 cytokine secretion (IL-4, IL-5, IL-13) in expanded iNKT cells stimulated with anti-CD2/CD3/CD28 antibodies. Cytotoxic effector molecules including granzyme B were increased in expanded iNKT cells after CD2/CD3/CD28 stimulation. Direct cytotoxicity assays using unstimulated expanded iNKT cell effectors revealed α-galactosyl ceramide (α-GalCer)-dependent killing of the T-ALL cell line Jurkat. Moreover, CD2/CD3/CD28 stimulation of expanded iNKT cells augmented their (α-GalCer-independent) killing of Jurkat cells. Co-culture of expanded iNKT cells with stimulated responder cells confirmed contact-dependent inhibition of activated CD4+ and CD8+ responder T cells. DISCUSSION These data establish a robust protocol to expand and novel pathways to enhance Th2 cytokine secretion and direct cytotoxicity in human iNKT cells, findings with direct implications for autoimmunity, vaccine augmentation and anti-infective immunity, cancer immunotherapy and transplantation.
Collapse
|
11
|
Fu S, He K, Tian C, Sun H, Zhu C, Bai S, Liu J, Wu Q, Xie D, Yue T, Shen Z, Dai Q, Yu X, Zhu S, Liu G, Zhou R, Duan S, Tian Z, Xu T, Wang H, Bai L. Impaired lipid biosynthesis hinders anti-tumor efficacy of intratumoral iNKT cells. Nat Commun 2020; 11:438. [PMID: 31974378 PMCID: PMC6978340 DOI: 10.1038/s41467-020-14332-x] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 12/30/2019] [Indexed: 12/14/2022] Open
Abstract
Dysfunction of invariant natural killer T (iNKT) cells in tumor microenvironment hinders their anti-tumor efficacy, and the underlying mechanisms remain unclear. Here we report that iNKT cells increase lipid biosynthesis after activation, and that is promoted by PPARγ and PLZF synergically through enhancing transcription of Srebf1. Among those lipids, cholesterol is required for the optimal IFN-γ production from iNKT cells. Lactic acid in tumor microenvironment reduces expression of PPARγ in intratumoral iNKT cells and consequently diminishes their cholesterol synthesis and IFN-γ production. Importantly, PPARγ agonist pioglitazone, a thiazolidinedione drug for type 2 diabetes, successfully restores IFN-γ production in tumor-infiltrating iNKT cells from both human patients and mouse models. Combination of pioglitazone and alpha-galactosylceramide treatments significantly enhances iNKT cell-mediated anti-tumor immune responses and prolongs survival of tumor-bearing mice. Our studies provide a strategy to augment the anti-tumor efficacy of iNKT cell-based immunotherapies via promoting their lipid biosynthesis. Lipid metabolism has been linked to iNKT function largely as it impacts processing and presentation of lipids they recognize. Here the authors show that iNKT-intrinsic lipid biosynthesis is important for their function but is impaired in tumors, and its restoration with PPARγ agonist drugs promotes anti-tumor iNKT response.
Collapse
Affiliation(s)
- Sicheng Fu
- Department of Oncology of The First Affiliated Hospital, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Kaixin He
- Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Chenxi Tian
- Department of Oncology of The First Affiliated Hospital, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Hua Sun
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230022, China
| | - Chenwen Zhu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230022, China
| | - Shiyu Bai
- Department of Oncology of The First Affiliated Hospital, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Jiwei Liu
- Department of Oncology of The First Affiliated Hospital, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Qielan Wu
- Department of Oncology of The First Affiliated Hospital, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Di Xie
- Department of Oncology of The First Affiliated Hospital, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Ting Yue
- Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Zhuxia Shen
- Department of Cardiology, Jing'an District Centre Hospital of Shanghai, Fudan University, Shanghai, 200040, China
| | - Qingqing Dai
- Department of Hepatopancreatobiliary Surgery and Organ Transplantation Center, Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Xiaojun Yu
- Department of Hepatopancreatobiliary Surgery and Organ Transplantation Center, Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Shu Zhu
- Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Gang Liu
- National Synchrotron Radiation Laboratory, University of Science and Technology of China, Hefei, 230027, China
| | - Rongbin Zhou
- Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Shengzhong Duan
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Zhigang Tian
- Department of Oncology of The First Affiliated Hospital, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Tao Xu
- National Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230022, China.
| | - Li Bai
- Department of Oncology of The First Affiliated Hospital, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China. .,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
| |
Collapse
|
12
|
Benam KH, Denney L, Ho LP. How the Respiratory Epithelium Senses and Reacts to Influenza Virus. Am J Respir Cell Mol Biol 2019; 60:259-268. [PMID: 30372120 DOI: 10.1165/rcmb.2018-0247tr] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The human lung is constantly exposed to the environment and potential pathogens. As the interface between host and environment, the respiratory epithelium has evolved sophisticated sensing mechanisms as part of its defense against pathogens. In this review, we examine how the respiratory epithelium senses and responds to influenza A virus, the biggest cause of respiratory viral deaths worldwide.
Collapse
Affiliation(s)
- Kambez H Benam
- 1 Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado - Anschutz Medical Campus, Aurora, Colorado.,2 Department of Bioengineering, University of Colorado Denver, Aurora, Colorado; and
| | - Laura Denney
- 3 Translational Lung Immunology Programme, MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Ling-Pei Ho
- 3 Translational Lung Immunology Programme, MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| |
Collapse
|
13
|
Wu SY, Chen CL, Tseng PC, Chiu CY, Lin YE, Lin CF. Fractionated ionizing radiation facilitates interferon-γ signaling and anticancer activity in lung adenocarcinoma cells. J Cell Physiol 2019; 234:16003-16010. [PMID: 30767202 DOI: 10.1002/jcp.28258] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 12/11/2018] [Accepted: 01/10/2019] [Indexed: 01/24/2023]
Abstract
Fractionated ionizing radiation (FIR) is a radiotherapy regimen that is regularly performed as part of lung cancer treatment. In contrast to the growth inhibition caused by DNA damage, immunomodulation in post-irradiated cancer cells is not well documented. Interferon (IFN)-γ confers anticancer activity by triggering both growth inhibition and cytotoxicity. This study investigated the priming effects of FIR with immunomodulation on the anticancer IFN-γ. Cell morphology, cell growth, and cytotoxicity were observed in FIR-treated A549 lung adenocarcinoma. Induction of p53 and epithelial-mesenchymal transition (EMT) were monitored. Following FIR, activation of IFN-γ signaling pathways were detected. FIR caused changes in cell morphology, inhibited cell growth, and induced cytotoxicity. While p53 was induced by FIR, no epithelial-mesenchymal transition could be found. Following IFN-γ stimulation, FIR-induced p53-associated cell cytotoxicity was significantly enhanced. Additionally, FIR increased the downstream response to IFN-γ by facilitating IFN-γ-induced signal transducer and activator of transcription 1 (STAT1) signaling without affecting the receptor expression. FIR-facilitated STAT1 activation through the mechanism involving mitogen-activated protein kinase activation and Src-homology 2 domain-containing tyrosine phosphatase 2 inactivation. These results demonstrate the FIR-facilitated IFN-γ signaling and its anticancer activity.
Collapse
Affiliation(s)
- Szu-Yuan Wu
- Department of Radiation Oncology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.,Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chia-Ling Chen
- Department of Respiratory Therapy, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Po-Chun Tseng
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chi-Yun Chiu
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yung-En Lin
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chiou-Feng Lin
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
14
|
Paul S, Chhatar S, Mishra A, Lal G. Natural killer T cell activation increases iNOS +CD206 - M1 macrophage and controls the growth of solid tumor. J Immunother Cancer 2019; 7:208. [PMID: 31387637 PMCID: PMC6685184 DOI: 10.1186/s40425-019-0697-7] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 07/30/2019] [Indexed: 01/12/2023] Open
Abstract
Background NKT cells play an important role in anti-tumor immunity. Alpha-galactosylceramide (α-GalCer), a synthetic glycolipid is presented to natural killer T (NKT) cells by most antigen-presenting cells through CD1d molecules leading to activation of NKT cells. However, the precise mechanisms of how α-GalCer-activated NKT regulate the polarization of the macrophages and effector T cells in the solid tumor are not studied adequately. Methods We induced solid tumor in C57BL/6 mice by subcutaneous injection of B16F10 cell line (1 X 106 cells) and monitored the tumor growth. Animals were given an intraperitoneal injection of α-GalCer (2 μg/injection) in 200 μl PBS on day + 1, + 5, + 10, + 15, and + 20 (with respect to tumor cell injection). Immune cells were characterized using flow cytometry and immunofluorescence staining. NK cells, Gr1+ cells, and F4/80+ macrophages in the mice were depleted by intravenous injection of cell-specific antibodies. Statistical analysis was performed using Student’s t-test or one-way ANOVA. Results Our results showed that intratumoral NKT cells have a lower frequency of CD69, CD25, CD122, and IFN-γR expression; produced less inflammatory cytokines such as IFN-γ, TNF-α, and GM-CSF; higher frequency CD62L+ NKT cells; and also showed reduced proliferation as compared to the splenic NKT cells. Mice treated with α-GalCer showed a significantly increased frequency of IFN-γ-producing NKT cells, CD8+ T cells, and effector Th1 cells. Depletion of NK cells in α-GalCer-treated mice showed a lower frequency of IFN-γ-producing CD4+ and CD8+ T cells in the tumor and prevented the α-GalCer-induced tumor growth. NKT cell activation with α-GalCer treatment significantly increased the iNOS+CD206− M1-macrophages and reduced the iNOS−CD206+ M2-macrophages in the spleen and tumor, and depletion of F4/80+ macrophages prevented the α-GalCer-induced reduction in the tumor growth. Conclusions We showed that activation of NKT cell with α-GalCer modulates the frequency of M1-macrophages and effector Th1 cells in the secondary lymphoid tissues and tumor microenvironment and inhibit tumor growth. The finding suggests that activation of NKT cells with α-GalCer may provide an effective anti-cancer outcome. Electronic supplementary material The online version of this article (10.1186/s40425-019-0697-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sourav Paul
- National Centre for Cell Science, NCCS Complex, Pune University Campus, Ganeshkhind, Pune, MH-411007, India
| | - Sushanta Chhatar
- National Centre for Cell Science, NCCS Complex, Pune University Campus, Ganeshkhind, Pune, MH-411007, India
| | - Amrita Mishra
- National Centre for Cell Science, NCCS Complex, Pune University Campus, Ganeshkhind, Pune, MH-411007, India
| | - Girdhari Lal
- National Centre for Cell Science, NCCS Complex, Pune University Campus, Ganeshkhind, Pune, MH-411007, India.
| |
Collapse
|
15
|
Shahrabi S, Zayeri ZD, Ansari N, Hadad EH, Rajaei E. Flip-flops of natural killer cells in autoimmune diseases versus cancers: Immunologic axis. J Cell Physiol 2019; 234:16998-17010. [PMID: 30864163 DOI: 10.1002/jcp.28421] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 02/03/2019] [Accepted: 02/14/2019] [Indexed: 12/25/2022]
Abstract
Natural killer (NK) cells play an essential role in the immune response to infections, inflammations, and malignancies. Recent studies suggest that NK cell surface receptors and cytokines are the key points of the disease development and protection. We hypothesized that the interactions between NK cell receptors and targeted cells construct an eventual niche, and this niche has an eventual profile in various autoimmune diseases and cancers. The NK cells preactivated with cytokines, such as interleukin-2 (IL-2), IL-12, IL-15, and IL-18 can have higher cytotoxicity; however, the toxic side effect of IL-2 should be considered. The vicissitudes of NK cell profile and its receptors obey the environmental communications and cell interactions. Our vision around the NK cells as an immune axis remained dual, and we still cannot judge the immune responses based on the NK cell flip-flop. A design of eventual niche to monitor the NK cell and targeted cell interaction is needed to strengthen our ability in diagnosis and treatment approaches based on the NK cells. Here, we have reviewed the shifts in the NK cells and their surface receptors in autoimmune diseases, solid tumors, and leukemia, and also discussed the effective chemokines that affect NK cell activation and proliferation. The main aim of this review is to present a broader vision of the NK cell changes in autoimmune disease and cancers.
Collapse
Affiliation(s)
- Saeid Shahrabi
- Department of Biochemistry and Hematology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Zeinab D Zayeri
- Golestan Hospital Clinical Research Development Unit, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Narges Ansari
- Isfahan Bone Metabolic Disorders Research Center, Department of Internal Medicine, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Elham H Hadad
- Research Center of Thalassemia and Hemoglobinopathy, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Elham Rajaei
- Golestan Hospital Clinical Research Development Unit, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
16
|
Shimizu T, Abu Lila AS, Kawaguchi Y, Shimazaki Y, Watanabe Y, Mima Y, Hashimoto Y, Okuhira K, Storm G, Ishima Y, Ishida T. A Novel Platform for Cancer Vaccines: Antigen-Selective Delivery to Splenic Marginal Zone B Cells via Repeated Injections of PEGylated Liposomes. THE JOURNAL OF IMMUNOLOGY 2018; 201:2969-2976. [DOI: 10.4049/jimmunol.1701351] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 09/12/2018] [Indexed: 12/14/2022]
|
17
|
Bedard M, Salio M, Cerundolo V. Harnessing the Power of Invariant Natural Killer T Cells in Cancer Immunotherapy. Front Immunol 2017; 8:1829. [PMID: 29326711 PMCID: PMC5741693 DOI: 10.3389/fimmu.2017.01829] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 12/04/2017] [Indexed: 12/19/2022] Open
Abstract
Invariant natural killer T (iNKT) cells are a distinct subset of innate-like lymphocytes bearing an invariant T-cell receptor, through which they recognize lipid antigens presented by monomorphic CD1d molecules. Upon activation, iNKT cells are capable of not only having a direct effector function but also transactivating NK cells, maturing dendritic cells, and activating B cells, through secretion of several cytokines and cognate TCR-CD1d interaction. Endowed with the ability to orchestrate an all-encompassing immune response, iNKT cells are critical in shaping immune responses against pathogens and cancer cells. In this review, we examine the critical role of iNKT cells in antitumor responses from two perspectives: (i) how iNKT cells potentiate antitumor immunity and (ii) how CD1d+ tumor cells may modulate their own expression of CD1d molecules. We further explore hypotheses to explain iNKT cell activation in the context of cancer and how the antitumor effects of iNKT cells can be exploited in different forms of cancer immunotherapy, including their role in the development of cancer vaccines.
Collapse
Affiliation(s)
- Melissa Bedard
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Mariolina Salio
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Vincenzo Cerundolo
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
18
|
Speir M, Hermans IF, Weinkove R. Engaging Natural Killer T Cells as 'Universal Helpers' for Vaccination. Drugs 2017; 77:1-15. [PMID: 28005229 DOI: 10.1007/s40265-016-0675-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Conventional vaccine adjuvants enhance peptide-specific T-cell and B-cell responses by modifying peptide stability or uptake or by binding to pattern-recognition receptors on antigen-presenting cells (APCs). This article discusses the application of a distinct mechanism of adjuvant activity: the activation of type I, or invariant, natural killer T (iNKT) cells to drive cellular and humoral immune responses. Using a semi-invariant T-cell receptor (TCR), iNKT cells recognize glycolipid antigens presented on cluster of differentiation (CD)-1d molecules. When their ligands are presented in concert with peptides, iNKT cells can provide T-cell help, 'licensing' APCs to augment peptide-specific T-cell and antibody responses. We discuss the potential benefits and limitations of exploiting iNKT cells as 'universal helpers' to enhance vaccine responses for the treatment and prevention of cancer and infectious diseases.
Collapse
Affiliation(s)
- Mary Speir
- Malaghan Institute of Medical Research, PO Box 7060, Wellington, 6242, New Zealand
| | - Ian F Hermans
- Malaghan Institute of Medical Research, PO Box 7060, Wellington, 6242, New Zealand. .,School of Biological Sciences, Victoria University Wellington, PO Box 600, Wellington, 6140, New Zealand. .,Maurice Wilkins Centre, Private Bag 92019, Auckland, New Zealand.
| | - Robert Weinkove
- Malaghan Institute of Medical Research, PO Box 7060, Wellington, 6242, New Zealand. .,Wellington Blood and Cancer Centre, Wellington Hospital, Private Bag 7902, Wellington, 6242, New Zealand. .,Department of Pathology and Molecular Medicine, University of Otago Wellington, Wellington, 6021, New Zealand.
| |
Collapse
|
19
|
Lin CF, Lin CM, Lee KY, Wu SY, Feng PH, Chen KY, Chuang HC, Chen CL, Wang YC, Tseng PC, Tsai TT. Escape from IFN-γ-dependent immunosurveillance in tumorigenesis. J Biomed Sci 2017; 24:10. [PMID: 28143527 PMCID: PMC5286687 DOI: 10.1186/s12929-017-0317-0] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 01/19/2017] [Indexed: 12/24/2022] Open
Abstract
Immune interferon (IFN), also known as IFN-γ, promotes not only immunomodulation but also antimicrobial and anticancer activity. After IFN-γ binds to the complex of IFN-γ receptor (IFNGR) 1-IFNGR2 and subsequently activates its downstream signaling pathways, IFN-γ immediately causes transcriptional stimulation of a variety of genes that are principally involved in its biological activities. Regarding IFN-γ-dependent immunosurveillance, IFN-γ can directly suppress tumorigenesis and infection and/or can modulate the immunological status in both cancer cells and infected cells. Regarding the anticancer effects of IFN-γ, cancer cells develop strategies to escape from IFN-γ-dependent cancer immunosurveillance. Immune evasion, including the recruitment of immunosuppressive cells, secretion of immunosuppressive factors, and suppression of cytotoxic T lymphocyte responses, is speculated to be elicited by the oncogenic microenvironment. All of these events effectively downregulate IFN-γ-expressing cells and IFN-γ production. In addition to these extrinsic pathways, cancer cells may develop cellular tolerance that manifests as hyporesponsiveness to IFN-γ stimulation. This review discusses the potential escape mechanisms from IFN-γ-dependent immunosurveillance in tumorigenesis.
Collapse
Affiliation(s)
- Chiou-Feng Lin
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan. .,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan.
| | - Chih-Ming Lin
- Department of Thoracic Surgery, Lotung Poh-Ai Hospital, Yilan, 265, Taiwan
| | - Kang-Yun Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, 110, Taiwan.,Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Szu-Yuan Wu
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan.,Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan.,Department of Radiation Oncology, Wan Fang Hospital, Taipei Medical University, Taipei Medical University, Taipei, 110, Taiwan.,Department of Biotechnology, Hung Kuang University, Taichung, 433, Taiwan
| | - Po-Hao Feng
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, 110, Taiwan.,Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Kuan-Yuan Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, 110, Taiwan.,Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Hsiao-Chi Chuang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Chia-Ling Chen
- Translational Research Center, Taipei Medical University, Taipei, 110, Taiwan
| | - Yu-Chih Wang
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Po-Chun Tseng
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Tsung-Ting Tsai
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| |
Collapse
|
20
|
Li J, Chen JN, Zeng TT, He F, Chen SP, Ma S, Bi J, Zhu XF, Guan XY. CD133+ liver cancer stem cells resist interferon-gamma-induced autophagy. BMC Cancer 2016; 16:15. [PMID: 26758620 PMCID: PMC4711109 DOI: 10.1186/s12885-016-2050-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 01/06/2016] [Indexed: 01/05/2023] Open
Abstract
Background Hepatocellular carcinoma (HCC) is one of the most fatal malignancies worldwide, and CD133 is a popular cancer stem cell (CSC) marker for HCC. CD133+ CSCs have been reported to resist conventional chemo- and radiotherapy, but little is known about their response to immune surveillance. Interferon-gamma (IFN-γ) is one of key cytokines that the immune system produce to eradicate cancer cells, so we investigated the function of IFN-γ on CD133+ HCC CSCs in this study. Methods The response of CD133+ cells to IFN-γ was performed with functional assays (cell proliferation assay and tumor formation in nude mice), flow cytometry, immunofluorescence staining and RNA interference. Results We found that IFN-γ inhibited the proliferation of cell lines with low percentage of CD133+ cells (wild-type human cells, BEL7402, QGY7701) but it did not affect the proliferation of cell lines with high percentage of CD133+ cells (wild-type human cells, Huh7, PLC8024) in vivo and in vitro (nude mice). Flow cytometry analysis demonstrated that the percentage of CD133+ cells increased after IFN-γ treatment of low CD133+ cell lines. Furthermore, IFN-γ induced the autophagy of low CD133+ cell lines to decrease proliferation. Conclusion CD133+ HCC CSCs resisted IFN-γ-induced autophagy, which might also be a mechanism through which CSCs resist immune eradication. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2050-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jian Li
- Department of Clinical Laboratory, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Room 102, 107 W. Yanjiang Road, Guangzhou, 510120, China. .,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China. .,State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, PR China.
| | - Jin-Na Chen
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| | - Ting-Ting Zeng
- State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, PR China.
| | - Fan He
- Department of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| | - Shu-Peng Chen
- State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, PR China.
| | - Stephanie Ma
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| | - Jiong Bi
- Department of Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Xiao-Feng Zhu
- State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, PR China.
| | - Xin-Yuan Guan
- State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, PR China. .,Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
21
|
Abstract
The observation that a subset of cancer patients show evidence for spontaneous CD8+ T cell priming against tumor-associated antigens has generated renewed interest in the innate immune pathways that might serve as a bridge to an adaptive immune response to tumors. Manipulation of this endogenous T cell response with therapeutic intent-for example, using blocking antibodies inhibiting PD-1/PD-L1 (programmed death-1/programmed death ligand 1) interactions-is showing impressive clinical results. As such, understanding the innate immune mechanisms that enable this T cell response has important clinical relevance. Defined innate immune interactions in the cancer context include recognition by innate cell populations (NK cells, NKT cells, and γδ T cells) and also by dendritic cells and macrophages in response to damage-associated molecular patterns (DAMPs). Recent evidence has indicated that the major DAMP driving host antitumor immune responses is tumor-derived DNA, sensed by the stimulator of interferon gene (STING) pathway and driving type I IFN production. A deeper knowledge of the clinically relevant innate immune pathways involved in the recognition of tumors is leading toward new therapeutic strategies for cancer treatment.
Collapse
|
22
|
Ando T, Ito H, Arioka Y, Ogiso H, Seishima M. Combination therapy with α-galactosylceramide and a Toll-like receptor agonist exerts an augmented suppressive effect on lung tumor metastasis in a mouse model. Oncol Rep 2014; 33:826-32. [PMID: 25434827 DOI: 10.3892/or.2014.3634] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 11/04/2014] [Indexed: 11/06/2022] Open
Abstract
α-galactosylceramide (GalCer), which is a natural killer T (NKT) cell ligand, has been reported to exert therapeutic effects against cancer in humans and mice. Toll-like receptor (TLR) agonists systemically or locally boost antitumor efficacy in mouse cancer models. In our previous study, the co-administration of GalCer and a TLR agonist synergistically enhanced interferon-γ (IFN-γ) production in mouse splenocytes in vitro and in vivo. The increased IFN-γ production promoted a tumor antigen-specific Th1 response. Therefore, co-treatment with GalCer and a TLR agonist is expected to exert an enhanced antitumor effect. In the present study, we examined the effect of GalCer and lipopolysaccharide (LPS) combination therapy in a mouse lung-metastasis model. GalCer and LPS combination therapy markedly decreased the number of lung metastatic tumor nodes. Co-treatment with GalCer and LPS enhanced the mRNA expression of CXCL9 and CXCL10 in mediastinal lymph nodes (MLNs) and increased the number of CD8+ cells in the MLNs. Furthermore, the depletion of CD8+ T cells canceled the antitumor effect of GalCer and LPS combination therapy. Thus, GalCer and LPS combination therapy significantly enhanced tumor antigen-specific immune responses and suppressed tumor growth in a mouse lung-metastasis model.
Collapse
Affiliation(s)
- Tatsuya Ando
- Department of Informative Clinical Medicine, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Hiroyasu Ito
- Department of Informative Clinical Medicine, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Yuko Arioka
- Department of Informative Clinical Medicine, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Hideyuki Ogiso
- Department of Informative Clinical Medicine, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Mitsuru Seishima
- Department of Informative Clinical Medicine, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| |
Collapse
|
23
|
Payne KK, Bear HD, Manjili MH. Adoptive cellular therapy of cancer: exploring innate and adaptive cellular crosstalk to improve anti-tumor efficacy. Future Oncol 2014; 10:1779-94. [DOI: 10.2217/fon.14.97] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
ABSTRACT The mammalian immune system has evolved to produce multi-tiered responses consisting of both innate and adaptive immune cells collaborating to elicit a functional response to a pathogen or neoplasm. Immune cells possess a shared ancestry, suggestive of a degree of coevolution that has resulted in optimal functionality as an orchestrated and highly collaborative unit. Therefore, the development of therapeutic modalities that harness the immune system should consider the crosstalk between cells of the innate and adaptive immune systems in order to elicit the most effective response. In this review, the authors will discuss the success achieved using adoptive cellular therapy in the treatment of cancer, recent trends that focus on purified T cells, T cells with genetically modified T-cell receptors and T cells modified to express chimeric antigen receptors, as well as the use of unfractionated immune cell reprogramming to achieve optimal cellular crosstalk upon infusion for adoptive cellular therapy.
Collapse
Affiliation(s)
- Kyle K Payne
- Department of Microbiology & Immunology, Virginia Commonwealth University, Massey Cancer Center, Richmond, VA 23298, USA
| | - Harry D Bear
- Department of Microbiology & Immunology, Virginia Commonwealth University, Massey Cancer Center, Richmond, VA 23298, USA
- Department of Surgery, Virginia Commonwealth University, Massey Cancer Center, Richmond, VA 23298, USA
| | - Masoud H Manjili
- Department of Microbiology & Immunology, Virginia Commonwealth University, Massey Cancer Center, Richmond, VA 23298, USA
| |
Collapse
|
24
|
Zhao Q, Tong L, He N, Feng G, Leng L, Sun W, Xu Y, Wang Y, Xiang R, Li Z. IFN-γ mediates graft-versus-breast cancer effects via enhancing cytotoxic T lymphocyte activity. Exp Ther Med 2014; 8:347-354. [PMID: 25009582 PMCID: PMC4079438 DOI: 10.3892/etm.2014.1760] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Accepted: 05/13/2014] [Indexed: 12/11/2022] Open
Abstract
Previous studies have demonstrated the beneficial effect of graft-versus-tumor (GVT) following hematopoietic stem cell transplantation (HSCT) on the incidence of leukemia relapse and the overall survival rate of patients with leukemia; however, detailed mechanisms underlying the effects GVT exhibits on solid tumors following allogeneic HSCT are yet to be elucidated. The aim of the present study was to investigate the immune mechanism underlying the effect of interferon (IFN)-γ on GVT following allogeneic HSCT in breast cancer therapy. An in situ breast cancer mouse model was established by injecting 5×104 4T1 cells into the mammary fat pads of BALB/c mice. The 4T1 cells were transfected with the firefly luciferase reporter gene in order to monitor the tumor progression in real time. An allogeneic HSCT model was then established by transplanting bone marrow mononuclear cells from C57BL/6 mice to the BALB/c mice. To investigate the influence of T lymphocyte proliferation following allogeneic bone marrow transplantation, the levels of CD3+CD8+ cytotoxic T lymphocytes (CTLs) and CD4+CD25+ regulatory T cells were determined. In addition, IFN-γ and granzyme B expression levels in splenic lymphocytes were analyzed using flow cytometry. Allogeneic HSCT was found to significantly promote the proliferation and cytotoxicity of CTLs and suppress the growth of breast cancer. Furthermore, the secretory levels of IFN-γ and granzyme B by T cells were elevated following allogeneic HSCT. These results indicated that alloreactive T cells increased the secretion of IFN-γ, which promoted the alloresponse of donor CTLs. In addition, the CTLs produced granzyme B, which exerted a tumor suppressive effect.
Collapse
Affiliation(s)
- Qianjie Zhao
- Department of Pathophysiology, School of Medicine, Nankai University, Ministry of Education, Tianjin 300071, P.R. China ; Key Laboratory of Bioactive Materials, College of Life Science, Nankai University, Ministry of Education, Tianjin 300071, P.R. China
| | - Lingling Tong
- Department of Pathophysiology, School of Medicine, Nankai University, Ministry of Education, Tianjin 300071, P.R. China
| | - Ningning He
- Department of Pathophysiology, School of Medicine, Nankai University, Ministry of Education, Tianjin 300071, P.R. China
| | - Guowei Feng
- Department of Pathophysiology, School of Medicine, Nankai University, Ministry of Education, Tianjin 300071, P.R. China
| | - Liang Leng
- Department of Pathophysiology, School of Medicine, Nankai University, Ministry of Education, Tianjin 300071, P.R. China
| | - Weijun Sun
- Department of Pathophysiology, School of Medicine, Nankai University, Ministry of Education, Tianjin 300071, P.R. China
| | - Yang Xu
- Department of Pathophysiology, School of Medicine, Nankai University, Ministry of Education, Tianjin 300071, P.R. China
| | - Yuebing Wang
- Department of Pathophysiology, School of Medicine, Nankai University, Ministry of Education, Tianjin 300071, P.R. China
| | - Rong Xiang
- Department of Pathophysiology, School of Medicine, Nankai University, Ministry of Education, Tianjin 300071, P.R. China
| | - Zongjin Li
- Department of Pathophysiology, School of Medicine, Nankai University, Ministry of Education, Tianjin 300071, P.R. China ; Key Laboratory of Bioactive Materials, College of Life Science, Nankai University, Ministry of Education, Tianjin 300071, P.R. China
| |
Collapse
|
25
|
Innate and adaptive immune cells in the tumor microenvironment. Nat Immunol 2013; 14:1014-22. [PMID: 24048123 DOI: 10.1038/ni.2703] [Citation(s) in RCA: 2861] [Impact Index Per Article: 260.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2013] [Accepted: 08/06/2013] [Indexed: 02/06/2023]
Abstract
Most tumor cells express antigens that can mediate recognition by host CD8(+) T cells. Cancers that are detected clinically must have evaded antitumor immune responses to grow progressively. Recent work has suggested two broad categories of tumor escape based on cellular and molecular characteristics of the tumor microenvironment. One major subset shows a T cell-inflamed phenotype consisting of infiltrating T cells, a broad chemokine profile and a type I interferon signature indicative of innate immune activation. These tumors appear to resist immune attack through the dominant inhibitory effects of immune system-suppressive pathways. The other major phenotype lacks this T cell-inflamed phenotype and appears to resist immune attack through immune system exclusion or ignorance. These two major phenotypes of tumor microenvironment may require distinct immunotherapeutic interventions for maximal therapeutic effect.
Collapse
|
26
|
Peripheral blood mononuclear cells of patients with breast cancer can be reprogrammed to enhance anti-HER-2/neu reactivity and overcome myeloid-derived suppressor cells. Breast Cancer Res Treat 2013; 142:45-57. [PMID: 24197563 DOI: 10.1007/s10549-013-2733-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 10/10/2013] [Indexed: 01/04/2023]
Abstract
Two major barriers in the immunotherapy of breast cancer include tumor-induced immune suppression and the establishment of long-lasting immune responses against the tumor. Recently, we demonstrated in an animal model of breast carcinoma that expanding and reprogramming tumor-sensitized lymphocytes, ex vivo, yielded T memory (Tm) cells as well as activated CD25+ NKT cells and NK cells. The presence of activated CD25+ NKT and NK cells rendered reprogrammed T cells resistant to MDSC-mediated suppression, and adoptive cellular therapy (ACT) of reprogrammed lymphocytes protected the host from tumor development and relapse. Here, we performed a pilot study to determine the clinical applicability of our protocol using peripheral blood mononuclear cells (PBMCs) of breast cancer patients, ex vivo. We show that bryostatin 1 and ionomycin combined with IL-2, IL-7, and IL-15 can expand and reprogram tumor-sensitized PBMCs. Reprogrammed lymphocytes contained activated CD25+ NKT and NK cells as well as Tm cells and displayed enhanced reactivity against HER-2/neu in the presence of MDSCs. The presence of activated NKT cells was highly correlated with the rescue of anti-HER-2/neu immune responses from MDSC suppression. Ex vivo blockade experiments suggest that the NKG2D pathway may play an important role in overcoming MDSC suppression. Our results show the feasibility of reprogramming tumor-sensitized immune cells, ex vivo, and provide rationale for ACT of breast cancer patients.
Collapse
|
27
|
Gajewski TF, Schreiber H, Fu YX. Innate and adaptive immune cells in the tumor microenvironment. Nat Immunol 2013. [PMID: 24048123 DOI: 10.1038/ni.2703.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Most tumor cells express antigens that can mediate recognition by host CD8(+) T cells. Cancers that are detected clinically must have evaded antitumor immune responses to grow progressively. Recent work has suggested two broad categories of tumor escape based on cellular and molecular characteristics of the tumor microenvironment. One major subset shows a T cell-inflamed phenotype consisting of infiltrating T cells, a broad chemokine profile and a type I interferon signature indicative of innate immune activation. These tumors appear to resist immune attack through the dominant inhibitory effects of immune system-suppressive pathways. The other major phenotype lacks this T cell-inflamed phenotype and appears to resist immune attack through immune system exclusion or ignorance. These two major phenotypes of tumor microenvironment may require distinct immunotherapeutic interventions for maximal therapeutic effect.
Collapse
|
28
|
Hoff P, Rakow A, Gaber T, Hahne M, Sentürk U, Strehl C, Fangradt M, Schmidt-Bleek K, Huscher D, Winkler T, Matziolis D, Matziolis G, Badakhshi H, Burmester GR, Duda GN, Perka C, Buttgereit F. Preoperative irradiation for the prevention of heterotopic ossification induces local inflammation in humans. Bone 2013; 55:93-101. [PMID: 23571050 DOI: 10.1016/j.bone.2013.03.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 03/28/2013] [Accepted: 03/29/2013] [Indexed: 11/25/2022]
Abstract
Radiation of the hip is an established method to prevent heterotopic ossification (HO) following total hip arthroplasty (THA) but the precise mechanism is unclear. As inflammatory processes are suggested to be involved in the pathogenesis of HO, we hypothesized that the preoperative irradiation impacts local immune components. Therefore, we quantified immune cell populations and cytokines in hematomas resulting from the transection of the femur in two groups of patients receiving THA: patients irradiated preoperatively (THA-X-hematoma: THA-X-H group) in the hip region (7 Gy) in order to prevent HO and patients who were not irradiated (THA-H group) but were postoperatively treated with non-steroidal anti-inflammatory drugs (NSAIDs). Radiation resulted in significantly increased frequencies of T cells, cytotoxic T cells, NKT cells and CD25+CD127- Treg cells, whereas the number of naive CD45RA-expressing cytotoxic T cells was reduced. These results indicate differential immune cell activation, corroborated by our findings of significantly higher concentrations of pro-inflammatory cytokines (e.g., IL-6, IFNγ) and chemokines (e.g., MCP-1, RANTES) in the THA-X-H group as compared to THA-H group. In contrast, the concentration of the angiogenic VEGF was significantly suppressed in the THA-X-H group. We conclude that preoperative irradiation results in significant changes in immune cell composition and cytokine secretion in THA-hematomas, establishing a specific - rather proinflammatory - milieu. This increase of inflammatory activity together with the observed suppression in VEGF secretion may contribute to the prevention of HO.
Collapse
Affiliation(s)
- Paula Hoff
- Department of Rheumatology and Clinical Immunology, Charité University Hospital, Berlin, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Transient Foxp3(+) regulatory T-cell depletion enhances therapeutic anticancer vaccination targeting the immune-stimulatory properties of NKT cells. Immunol Cell Biol 2012; 91:105-14. [PMID: 23090488 DOI: 10.1038/icb.2012.58] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The natural killer T (NKT) cell ligand, alpha-galactosylceramide (α-GalCer), represents a potential adjuvant to boost immunotherapeutic vaccination strategies against poorly immunogenic cancers. The objective of this study was to assess the therapeutic potential of an α-GalCer-loaded tumor-cell vaccine against solid tumors in mice and to enhance the effectiveness of this approach by removing immune suppression associated with the activity of Foxp3(+) regulatory T cells (Tregs). In the B16F10 melanoma model, we show that single vaccination with irradiated, α-GalCer-loaded tumor cells resulted in suppression of established subcutaneous (s.c.) B16F10 tumor growth, which was mediated by NKT cell-dependent IFN-γ production and enhanced in the absence of IL-17 A. Selective depletion of Foxp3(+) Tregs in transgenic DEpletion of REGulatory T cells (DEREG) mice led to significant inhibition of B16F10 tumor growth and enhanced survival of mice receiving vaccination. Short-term elimination of Foxp3(+) Tregs (<7 days) was sufficient to boost vaccine-induced immunity. Enhanced antitumor activity with combination therapy was associated with an increase in systemic NK cell and effector CD8(+) T-cell activation and IFN-γ production, as well as infiltration of effector CD8(+) T cells into the tumor. Overall, these findings demonstrate that transient depletion of Foxp3(+) Tregs constitutes a highly effective strategy to improve the therapeutic efficacy of anticancer vaccination with NKT cell adjuvants.
Collapse
|
30
|
Simon-Keller K, Mößinger K, Bohlender AL, Ströbel P, Marx A. Variable Resistance of RMS to Interferon γ Signaling. ISRN ONCOLOGY 2012; 2012:789152. [PMID: 22919516 PMCID: PMC3420146 DOI: 10.5402/2012/789152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 04/10/2012] [Indexed: 11/24/2022]
Abstract
Aims. Chimeric T cells directed to the γ-subunit of the fetal acetylcholine receptor (fAChR) produce large amounts of interferon-γ (IFNγ) on coculture with fAChR-expressing rhabdomyosarcoma (RMS) cells prior to RMS cell death. The aim of this study was to elucidate whether IFNγ blocks proliferation and survival of RMS cells and modulates expression of genes with relevance for cytotoxicity of chimeric T cells. Methods. Expression levels of IFNγ receptor (IFNGR), AChR, MHCI, MHCII, and CIITA (class II transactivator) by RMS were checked by flow cytometry, qRT-PCR, and western blot. Proliferation and cell survival were investigated by annexin V and propidium iodide staining and MTT (thiazolyl-blue-tetrazolium-bromide) assay. Key phosphorylation and binding sites of IFNGRs were checked by DNA sequencing. Results. IFNγ treatment blocked proliferation in 3 of 6 RMS cell lines, but reduced survival in only one. IFNGR was expressed at levels comparable to controls and binding sites for JAK and STAT1 were intact. Induction of several target genes (e.g., AChR, MHCI, and MHCII) by IFNγ was detected on the RNA level but not protein level. Conclusions. IFNγ does not significantly contribute to the killing of RMS cells by fAChR directed chimeric T cells. Signalling downstream of the IFNR receptor, including the posttranscriptional level, is impaired in most RMS cell lines.
Collapse
Affiliation(s)
- Katja Simon-Keller
- Institute of Pathology, University Medical Centre Mannheim, University of Heidelberg, 68135 Mannheim, Germany
| | | | | | | | | |
Collapse
|
31
|
Zeng J, Shahbazi M, Wu C, Toh HC, Wang S. Enhancing Immunostimulatory Function of Human Embryonic Stem Cell-Derived Dendritic Cells by CD1d Overexpression. THE JOURNAL OF IMMUNOLOGY 2012; 188:4297-304. [DOI: 10.4049/jimmunol.1102343] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
32
|
Han Z, Jing Y, Zhang S, Liu Y, Shi Y, Wei L. The role of immunosuppression of mesenchymal stem cells in tissue repair and tumor growth. Cell Biosci 2012; 2:8. [PMID: 22390479 PMCID: PMC3315743 DOI: 10.1186/2045-3701-2-8] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Accepted: 03/05/2012] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have acquired great interests for their potential use in the clinical therapy of many diseases because of their functions including multiple lineage differentiation, low immunogenicity and immunosuppression. Many studies suggest that MSCs are strongly immunosuppressive in vitro and in vivo. MSCs exert a profound inhibitory effect on the proliferation of T cells, B cells, dendritic cells and natural killer cells. In addition, several soluble factors have been reported to involved in the immunosuppressive effects by MSCs such as TGF-β, HGF, PGE2, IDO and iNOS. These results suggest that MSCs can be used in the therapy of immune disorder diseases, prevention of organ transplantation rejection and tissue injury. In recent study, we demonstrated that MSCs in tumor inflammatory microenvironment might be elicited of immunosuppressive function. Thus, the application of MSCs in cancer therapy might have negative effect by helping tumor cells escaping from the immune surveillance.
Collapse
Affiliation(s)
- Zhipeng Han
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, the Second Military Medicial University, Shanghai, China.
| | | | | | | | | | | |
Collapse
|
33
|
Allen CT, Judd NP, Bui JD, Uppaluri R. The clinical implications of antitumor immunity in head and neck cancer. Laryngoscope 2011; 122:144-57. [DOI: 10.1002/lary.21913] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
34
|
Sherman MH, Downes M, Evans RM. Nuclear receptors as modulators of the tumor microenvironment. Cancer Prev Res (Phila) 2011; 5:3-10. [PMID: 22135047 DOI: 10.1158/1940-6207.capr-11-0528] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Over the past several decades of cancer research, the inherent complexity of tumors has become increasingly appreciated. In addition to acquired cell-intrinsic properties, tumor initiation and growth is supported by an abundance of parenchymal, inflammatory, and stromal cell types, which infiltrate and surround the tumor. Accumulating evidence shows that numerous components of this supportive milieu, referred to collectively as the tumor microenvironment, are indeed critical during the process of multistep tumorigenesis. These findings highlight the important interplay between neoplastic cells and tumor-associated cell types, and suggest that therapy should target both neoplastic cells and supportive stromal cells to effectively attenuate tumor growth. The nuclear receptor superfamily encompasses a druggable class of molecules expressed in numerous stromal and parenchymal cell types, whose established physiologic roles suggest their potential as therapeutic and preventive targets in the context of the reactive tumor microenvironment. In this minireview, we discuss recent evidence that tumor-associated inflammation, angiogenesis, and fibrosis can be modulated at the transcriptional level by nuclear receptors and their ligands. As these processes have been widely implicated in cancer initiation, progression, and resistance to current therapy, nuclear receptor ligands targeting the tumor microenvironment may be potent antitumor agents in combination therapies, including for preventing cancer development within high-risk populations.
Collapse
Affiliation(s)
- Mara H Sherman
- Gene Expression Laboratory, Howard Hughes Medical Institute, 10010 N Torrey Pines Road, La Jolla, CA, USA
| | | | | |
Collapse
|
35
|
Palendira U, Low C, Chan A, Hislop AD, Ho E, Phan TG, Deenick E, Cook MC, Riminton DS, Choo S, Loh R, Alvaro F, Booth C, Gaspar HB, Moretta A, Khanna R, Rickinson AB, Tangye SG. Molecular pathogenesis of EBV susceptibility in XLP as revealed by analysis of female carriers with heterozygous expression of SAP. PLoS Biol 2011; 9:e1001187. [PMID: 22069374 PMCID: PMC3206011 DOI: 10.1371/journal.pbio.1001187] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2011] [Accepted: 09/16/2011] [Indexed: 11/18/2022] Open
Abstract
X-linked lymphoproliferative disease (XLP) is a primary immunodeficiency caused by mutations in SH2D1A which encodes SAP. SAP functions in signalling pathways elicited by the SLAM family of leukocyte receptors. A defining feature of XLP is exquisite sensitivity to infection with EBV, a B-lymphotropic virus, but not other viruses. Although previous studies have identified defects in lymphocytes from XLP patients, the unique role of SAP in controlling EBV infection remains unresolved. We describe a novel approach to this question using female XLP carriers who, due to random X-inactivation, contain both SAP(+) and SAP(-) cells. This represents the human equivalent of a mixed bone marrow chimera in mice. While memory CD8(+) T cells specific for CMV and influenza were distributed across SAP(+) and SAP(-) populations, EBV-specific cells were exclusively SAP(+). The preferential recruitment of SAP(+) cells by EBV reflected the tropism of EBV for B cells, and the requirement for SAP expression in CD8(+) T cells for them to respond to Ag-presentation by B cells, but not other cell types. The inability of SAP(-) clones to respond to Ag-presenting B cells was overcome by blocking the SLAM receptors NTB-A and 2B4, while ectopic expression of NTB-A on fibroblasts inhibited cytotoxicity of SAP(-) CD8(+) T cells, thereby demonstrating that SLAM receptors acquire inhibitory function in the absence of SAP. The innovative XLP carrier model allowed us to unravel the mechanisms underlying the unique susceptibility of XLP patients to EBV infection in the absence of a relevant animal model. We found that this reflected the nature of the Ag-presenting cell, rather than EBV itself. Our data also identified a pathological signalling pathway that could be targeted to treat patients with severe EBV infection. This system may allow the study of other human diseases where heterozygous gene expression from random X-chromosome inactivation can be exploited.
Collapse
MESH Headings
- Antigens, CD/immunology
- B-Lymphocytes/pathology
- B-Lymphocytes/virology
- CD48 Antigen
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/pathology
- CD8-Positive T-Lymphocytes/virology
- Dendritic Cells/immunology
- Epstein-Barr Virus Infections/genetics
- Epstein-Barr Virus Infections/immunology
- Epstein-Barr Virus Infections/virology
- Female
- Genotype
- Herpesvirus 4, Human/immunology
- Herpesvirus 4, Human/pathogenicity
- Humans
- Immunoglobulin Class Switching
- Influenza, Human/immunology
- Influenza, Human/virology
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/immunology
- Leukocytes, Mononuclear/immunology
- Leukocytes, Mononuclear/pathology
- Leukocytes, Mononuclear/virology
- Lymphoproliferative Disorders/genetics
- Lymphoproliferative Disorders/immunology
- Lymphoproliferative Disorders/pathology
- Lymphoproliferative Disorders/virology
- Orthomyxoviridae/immunology
- Orthomyxoviridae/pathogenicity
- Receptors, Cell Surface/immunology
- Receptors, Immunologic/immunology
- Signal Transduction
- Signaling Lymphocytic Activation Molecule Associated Protein
- Signaling Lymphocytic Activation Molecule Family
- Signaling Lymphocytic Activation Molecule Family Member 1
- X Chromosome Inactivation
Collapse
Affiliation(s)
- Umaimainthan Palendira
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- St. Vincent's Clinical School, University of New South Wales, Darlinghurst, New South Wales, Australia
| | - Carol Low
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Anna Chan
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Andrew D. Hislop
- School of Cancer Sciences and MRC Centre for Immune Regulation, University of Birmingham, Edgbaston, United Kingdom
| | - Edwin Ho
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Tri Giang Phan
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- St. Vincent's Clinical School, University of New South Wales, Darlinghurst, New South Wales, Australia
| | - Elissa Deenick
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- St. Vincent's Clinical School, University of New South Wales, Darlinghurst, New South Wales, Australia
| | - Matthew C. Cook
- Australian National University Medical School, Canberra, Australian Capital Territory, Australia
- John Curtin School of Medical Research, Canberra, Australian Capital Territory, Australia
- Department of Immunology, Canberra Hospital, Canberra, Australian Capital Territory, Australia
| | - D. Sean Riminton
- Department of Immunology, Concord Hospital, Sydney, New South Wales, Australia
| | - Sharon Choo
- Department of Allergy and Immunology, Royal Children's Hospital Melbourne, Melbourne, Victoria, Australia
| | - Richard Loh
- Department of Clinical Immunology, Princess Margaret Hospital for Children, Perth, Western Australia, Australia
| | - Frank Alvaro
- Pediatric Hematology, John Hunter Hospital, Newcastle, New South Wales, Australia
| | - Claire Booth
- Centre for Immunodeficiency, Molecular Immunology Unit, UCL Institute of Child Health, London, United Kingdom
| | - H. Bobby Gaspar
- Centre for Immunodeficiency, Molecular Immunology Unit, UCL Institute of Child Health, London, United Kingdom
| | - Alessandro Moretta
- Dipartimento di Medicina Sperimentale, Università di Genova, Genova, Italy
| | - Rajiv Khanna
- Tumour Immunology Laboratory, Division of Immunology, Queensland Institute of Medical Research, Brisbane, Queensland, Australia
| | - Alan B. Rickinson
- School of Cancer Sciences and MRC Centre for Immune Regulation, University of Birmingham, Edgbaston, United Kingdom
| | - Stuart G. Tangye
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- St. Vincent's Clinical School, University of New South Wales, Darlinghurst, New South Wales, Australia
- * E-mail:
| |
Collapse
|
36
|
Liu Y, Han ZP, Zhang SS, Jing YY, Bu XX, Wang CY, Sun K, Jiang GC, Zhao X, Li R, Gao L, Zhao QD, Wu MC, Wei LX. Effects of inflammatory factors on mesenchymal stem cells and their role in the promotion of tumor angiogenesis in colon cancer. J Biol Chem 2011; 286:25007-15. [PMID: 21592963 DOI: 10.1074/jbc.m110.213108] [Citation(s) in RCA: 132] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells (MSCs), which are modulated by cytokines present in the tumor microenvironment, play an important role in tumor progression. It is well documented that inflammation is an important part of the tumor microenvironment, so we investigated whether stimulation of MSCs by inflammatory cytokines would contribute to their ability to promote tumor growth. We first showed that MSCs could increase C26 colon cancer growth in mice. This growth-promoting effect was further accelerated when the MSCs were pre-stimulated by inflammatory factors IFN-γ and TNF-α. At the same time, we demonstrated that MSCs pre-stimulated by both inflammatory factors could promote tumor angiogenesis in vivo to a greater degree than untreated MSCs or MSCs pre-stimulated by either IFN-γ or TNF-α alone. A hen egg test-chorioallantoic membrane (HET-CAM) assay showed that treatment of MSC-conditioned medium can promote chorioallantoic membrane angiogenesis in vitro, especially treatment with conditioned medium of MSCs pretreated with IFN-γ and TNF-α together. This mechanism of promoting angiogenesis appears to take place via an increase in the expression of vascular endothelial growth factor (VEGF), which itself takes place through an increase in signaling in the hypoxia-inducible factor 1α (HIF-1α)-dependent pathway. Inhibition of HIF-1α in MSCs by siRNA was found to effectively reduce the ability of MSC to affect the growth of colon cancer in vivo in the inflammatory microenviroment. These results indicate that MSCs stimulated by inflammatory cytokines such as IFN-γ and TNF-α in the tumor microenvironment express higher levels of VEGF via the HIF-1α signaling pathway and that these MSCs then enhance tumor angiogenesis, finally leading to colon cancer growth in mice.
Collapse
Affiliation(s)
- Yan Liu
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai 200438, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Motohashi S, Nakayama T. Invariant natural killer T cell-based immunotherapy for cancer. Immunotherapy 2011; 1:73-82. [PMID: 20635975 DOI: 10.2217/1750743x.1.1.73] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Human Valpha24 invariant natural killer T (iNKT) cells are a distinct lymphocyte population, characterized by an invariant T-cell receptor Valpha24 chain paired mainly with Valpha11. Valpha24 iNKT cells are activated by a glycolipid ligand - alpha-galactosylceramide - and produce a large amount of Th1 and Th2 cytokines, thereby modulating the function of other cells. iNKT cells have the capability to control a wide variety of immune responses, including antitumor immunity. Abnormalities in the number and function of Valpha24 iNKT cells have been observed in patients with malignant diseases accompanied with a poor clinical outcome. Therefore, therapeutic strategies that focused on the restoration of Valpha24 iNKT cell population and function would be a reasonable rationale for the treatment of cancer. In this article, the progress to date in the clinical studies of iNKT cell-based immunotherapy is briefly reviewed and the role of Valpha24 iNKT cells in cancer immunotherapy is highlighted.
Collapse
Affiliation(s)
- Shinichiro Motohashi
- Department of Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | | |
Collapse
|
38
|
Tan JQ, Xiao W, Wang L, He YL. Type I natural killer T cells: naturally born for fighting. Acta Pharmacol Sin 2010; 31:1123-32. [PMID: 20694020 PMCID: PMC4002303 DOI: 10.1038/aps.2010.119] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2010] [Accepted: 07/09/2010] [Indexed: 12/26/2022] Open
Abstract
Type capital I, Ukrainian natural killer T cells (NKT cells), a subset of CD1d-restricted T cells with invariant Valphabeta TCR, are characterized by prompt production of large amounts of Th1 and/or Th2 cytokines upon primary stimulation through the TCR complex. The rapid release of cytokines implies that type capital I, Ukrainian NKT cells may play a critical role in modulating the upcoming immune responses, such as anti-tumor response, protection against infection, and autoimmunity. As a bridge between innate and adaptive immunity, type capital I, Ukrainian NKT cells differentiate and mature upon stimulations to achieve and maintain a homeostasis. Orchestrating with other arms of adaptive immunity, type capital I, Ukrainian NKT cells show strong cytotoxic effects in response to various tumors in a direct and/or indirect manner(s). This review will focus primarily on type capital I, Ukrainian NKT cell development, homeostasis, and effector functions, especially in anti-tumor immunity, and followed by their potential applications in treatment of cancers.
Collapse
Affiliation(s)
- Jin-quan Tan
- Department of Immunology Institute of Allergy and Immune-related Diseases and Center for Medical Research, Wuhan University School of Medicine, Wuhan 430071, China
- Laboratory of Allergy and Clinical Immunology, Institute of Allergy and Immune-related Diseases and Center for Medical Research, Wuhan University School of Medicine, Wuhan 430071, China
| | - Wei Xiao
- Department of Immunology Institute of Allergy and Immune-related Diseases and Center for Medical Research, Wuhan University School of Medicine, Wuhan 430071, China
| | - Lan Wang
- Department of Immunology Institute of Allergy and Immune-related Diseases and Center for Medical Research, Wuhan University School of Medicine, Wuhan 430071, China
| | - Yu-ling He
- Department of Immunology Institute of Allergy and Immune-related Diseases and Center for Medical Research, Wuhan University School of Medicine, Wuhan 430071, China
- Laboratory of Allergy and Clinical Immunology, Institute of Allergy and Immune-related Diseases and Center for Medical Research, Wuhan University School of Medicine, Wuhan 430071, China
| |
Collapse
|
39
|
Bontkes HJ, Moreno M, Hangalapura B, Lindenberg JJ, de Groot J, Lougheed S, van der Vliet HJJ, van den Eertwegh AJM, de Gruijl TD, von Blomberg BME, Scheper RJ. Attenuation of invariant natural killer T-cell anergy induction through intradermal delivery of alpha-galactosylceramide. Clin Immunol 2010; 136:364-74. [PMID: 20570567 DOI: 10.1016/j.clim.2010.04.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2009] [Revised: 04/05/2010] [Accepted: 04/28/2010] [Indexed: 11/28/2022]
Abstract
CD1d restricted, alpha-galactosylceramide (alphaGC) responsive invariant (i)NKT cells positively regulate immune responses. Both intravenous and intradermal administered alphaGC are known to activate iNKT cells. iNKT cells become unresponsive to a second intravenous alphaGC injection, whereas no data are available regarding potential anergy upon intradermal administration. Here, comparative analysis of two intradermal versus two intravenous injections in mice demonstrated that iNKT cell anergy was prevented by intradermal injection and when combined with a vaccine, superior tumor protection afforded by intradermally administered alphaGC. Moreover, human skin dendritic cells (DC) took up intradermally injected alphaGC and activated iNKT cells upon migration, while iNKT cells in human skin-draining lymph nodes expanded in response to alphaGC presented either by exogenously added DC or by CD1d positive antigen presenting cells in the lymph nodes. In conclusion, glycolipids such as alphaGC may greatly improve the efficacy of skin immunization strategies, targeting cutaneous and lymph node DC.
Collapse
Affiliation(s)
- Hetty J Bontkes
- Department of Pathology, VU University Medical Center, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Regulation of the induction and function of cytotoxic T lymphocytes by natural killer T cell. J Biomed Biotechnol 2010; 2010:641757. [PMID: 20508728 PMCID: PMC2873660 DOI: 10.1155/2010/641757] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2009] [Revised: 02/14/2010] [Accepted: 03/09/2010] [Indexed: 02/05/2023] Open
Abstract
Cytotoxic T lymphocytes (CTLs) play a crucial role in the infections and the antitumor immunity. Induction and activation of antigen-specific CTLs is an important strategy in immunotherapy for various diseases, and several researchers have focused on the modulation of CTL induction and function. Natural killer T (NKT) cells are an important focus area of researchers studying immunomodulatory responses to tumors and infectious diseases. CD1d-restricted NKT cells consist of type I NKT cells and type II NKT cells. α-galactosylceramide (α-GalCer)-activated type I NKT cells secrete both Th1 (e.g., IFN-γ) and Th2 cytokines, affect the expression of costimulatory molecules in immune cells, and regulate the host immune system. Type II NKT cells, however, are stimulated by sulfatide, a self-glycolipid derived from myelin, and play an immunosuppressive role in animal model of autoimmune diseases. CTL generation, activation, and suppression are strongly affected by activated type I and type II NKT cells. Thus, the regulation of these NKT cells leads to the modification of CTL function. CTLs contribute to antimicrobial responses, antitumor immune and autoimmune responses. Understanding the role of NKT cells in the regulation of CTL generation, activation, and suppression enable the development of novel treatment strategies for these diseases.
Collapse
|
41
|
Miotto D, Lo Cascio N, Stendardo M, Querzoli P, Pedriali M, De Rosa E, Fabbri LM, Mapp CE, Boschetto P. CD8+ T cells expressing IL-10 are associated with a favourable prognosis in lung cancer. Lung Cancer 2010; 69:355-60. [PMID: 20089329 DOI: 10.1016/j.lungcan.2009.12.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2009] [Revised: 11/20/2009] [Accepted: 12/21/2009] [Indexed: 02/07/2023]
Abstract
The dual role of tumour-infiltrating macrophages and lymphocytes on nonsmall cell lung cancer (NSCLC) progression and prognosis may be due to the differential activity of their phenotypes. To investigate the impact of inflammatory cells on NSCLC, we first quantified the number of macrophages (CD68+) and lymphocytes (CD8+ and CD4+) and the percentage of CD8+ cells expressing IL-10 (CD8+/IL-10+) in tumour stroma and epithelium. Then, we evaluated the possible relationships between the numbers of these cells and the clinicopathological features and the overall survival of patients. Paraffin-embedded sections of surgical specimens from 64 patients who had undergone surgery for NSCLC were immunostained with antibodies directed against CD68, CD4, CD8 and IL-10. The percentage of CD8+/IL-10+ cells was higher in cancer stroma of patients with stage I NSCLC than in those with stages II, III, and IV. High percentages of stromal CD8+/IL-10+ cells were associated with longer overall patient survival. In contrast, the number of CD68+, CD8+ and CD4+ cells did not differ between stage I NSCLC and stages II, III, and IV. In conclusion, the survival advantage of patients with stage I NSCLC may be related to the anti-tumour activity of the CD8+/IL-10+ cell phenotype.
Collapse
Affiliation(s)
- D Miotto
- Department of Clinical and Experimental Medicine, Section of Hygiene and Occupational Medicine, University of Ferrara, Via Fossato di Mortara 64/B, 44100 Ferrara, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Lobashevsky A, Manwaring J, Travis M, Nord B, Higgins N, Serov Y, Arnoff T, Hommel-Berrey G, Goggins W, Taber T, Carter C, Smith D, Wozniak T, O'Donnell J, Turrentine M. Effect of desensitization in solid organ transplant recipients depends on some cytokines genes polymorphism. Transpl Immunol 2009; 21:169-78. [DOI: 10.1016/j.trim.2009.03.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2009] [Revised: 03/12/2009] [Accepted: 03/16/2009] [Indexed: 10/20/2022]
|
43
|
Bonehill A, Van Nuffel AM, Corthals J, Tuyaerts S, Heirman C, François V, Colau D, van der Bruggen P, Neyns B, Thielemans K. Single-Step Antigen Loading and Activation of Dendritic Cells by mRNA Electroporation for the Purpose of Therapeutic Vaccination in Melanoma Patients. Clin Cancer Res 2009; 15:3366-75. [DOI: 10.1158/1078-0432.ccr-08-2982] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
44
|
Maccalli C, Scaramuzza S, Parmiani G. TNK cells (NKG2D+ CD8+ or CD4+ T lymphocytes) in the control of human tumors. Cancer Immunol Immunother 2009; 58:801-8. [PMID: 19089424 PMCID: PMC11030286 DOI: 10.1007/s00262-008-0635-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2008] [Accepted: 11/21/2008] [Indexed: 10/21/2022]
Abstract
Innate and adaptive immune responses have many interactions that are regulated by the balance of signals initiated by a variety of activatory and inhibitory receptors. Among these, the NKG2D molecule was identified as expressed by T lymphocytes, including most CD8+ cells and a minority of CD4+ cells, designated TNK cells in this paper. Tumor cells may overexpress the stress-inducible NKG2D ligands (NKG2DLs: MICA/B, ULBPs) and the NKG2D signaling has been shown to be involved in lymphocyte-mediated anti-tumor activity. Aberrant expression of NKG2DLs by cancer cells, such as the release of soluble form of NKG2DLs, can lead to the impairment of these immune responses. Here, we discuss the significance of NKG2D in TNK-mediated anti-tumor activity. Our studies demonstrate that NKG2D+ T cells (TNK) are commonly recruited at the tumor site in melanoma patients where they may exert anti-tumor activity by engaging both TCR and NKG2D. Moreover, NKG2D and TCR triggering was also observed by peripheral blood derived T lymphocyte- or T cell clone-mediated tumor recognition, both in melanoma and colorectal cancer (CRC) patients. Notably, heterogeneous expression of NKG2DLs was found in melanoma and CRC cells, with a decrease of these molecules along with tumor progression. Therefore, through the mechanisms that govern NKG2D engagement in anti-tumor activity and the expression of NKG2DLs by tumor cells that still need to be dissected, we showed that NKG2D expressing TNK cells are a relevant T cell subtype for immunosurveillance of tumors and we propose that new immunotherapeutic interventions for cancer patients should be aimed also at enhancing NKG2DLs expression by tumor cells.
Collapse
Affiliation(s)
- Cristina Maccalli
- Unit of Immuno-biotherapy of Melanoma and Solid Tumors, Department of Molecular Oncology, San Raffaele Scientific and University Institute, Via Olgettina 58, Milan, Italy.
| | | | | |
Collapse
|
45
|
Invariant natural killer T cells and immunotherapy of cancer. Clin Immunol 2008; 129:182-94. [PMID: 18783990 DOI: 10.1016/j.clim.2008.07.025] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2007] [Revised: 07/29/2008] [Accepted: 07/29/2008] [Indexed: 11/20/2022]
Abstract
Invariant CD1d restricted natural killer T (iNKT) cells are regulatory cells that express a canonical TCR-Valpha-chain (Valpha24.Jalpha18 in humans and Valpha14.Jalpha18 in mice) which recognizes glycolipid antigens presented by the monomorphic CD1d molecule. They can secrete a wide variety of both pro-inflammatory and anti-inflammatory cytokines very swiftly upon their activation. Evidence for the significance of iNKT cells in human cancer has been ambiguous. Still, the (pre-)clinical findings reviewed here, provide evidence for a distinct contribution of iNKT cells to natural anti-tumor immune responses in humans. Furthermore, clinical phase I studies that are discussed here have revealed that the infusion of cancer patients with ligand-loaded dendritic cells or cultured iNKT cells is well tolerated. We thus underscore the potential of iNKT cell based immunotherapy in conjunction with established modalities such as surgery and radiotherapy, as adjuvant therapy against carcinomas.
Collapse
|