51
|
Jinushi M, Baghdadi M. Role of Innate Immunity in Cancers and Antitumor Response. CANCER IMMUNOLOGY 2020:11-28. [DOI: 10.1007/978-3-030-30845-2_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
52
|
Omene C, Ma L, Moore J, Ouyang H, Illa-Bochaca I, Chou W, Patel MS, Sebastiano C, Demaria S, Mao JH, Karagoz K, Gatza ML, Barcellos-Hoff MH. Aggressive Mammary Cancers Lacking Lymphocytic Infiltration Arise in Irradiated Mice and Can Be Prevented by Dietary Intervention. Cancer Immunol Res 2019; 8:217-229. [PMID: 31831632 DOI: 10.1158/2326-6066.cir-19-0253] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 07/26/2019] [Accepted: 11/27/2019] [Indexed: 01/06/2023]
Abstract
Because the incidence of breast cancer increases decades after ionizing radiation exposure, aging has been implicated in the evolution of the tumor microenvironment and tumor progression. Here, we investigated radiation-induced carcinogenesis using a model in which the mammary glands of 10-month-old BALB/c mice were transplanted with Trp53-null mammary tissue 3 days after exposure to low doses of sparsely ionizing γ-radiation or densely ionizing particle radiation. Mammary transplants in aged, irradiated hosts gave rise to significantly more tumors that grew more rapidly than those in sham-irradiated mice, with the most pronounced effects seen in mice irradiated with densely ionizing particle radiation. Tumor transcriptomes identified a characteristic immune signature of these aggressive cancers. Consistent with this, fast-growing tumors exhibited an immunosuppressive tumor microenvironment with few infiltrating lymphocytes, abundant immunosuppressive myeloid cells, and high COX-2 and TGFβ. Only irradiated hosts gave rise to tumors lacking cytotoxic CD8+ lymphocytes (defined here as immune desert), which also occurred in younger irradiated hosts. These data suggest that host irradiation may promote immunosuppression. To test this, young chimera mice were fed chow containing a honeybee-derived compound with anti-inflammatory and immunomodulatory properties, caffeic acid phenethyl ester (CAPE). CAPE prevented the detrimental effects of host irradiation on tumor growth rate, immune signature, and immunosuppression. These data indicated that low-dose radiation, particularly densely ionizing exposure of aged mice, promoted more aggressive cancers by suppressing antitumor immunity. Dietary intervention with a nontoxic immunomodulatory agent could prevent systemic effects of radiation that fuel carcinogenesis, supporting the potential of this strategy for cancer prevention.
Collapse
Affiliation(s)
- Coral Omene
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Lin Ma
- University of California, San Francisco, San Francisco, California
| | - Jade Moore
- University of California, San Francisco, San Francisco, California
| | - Haoxu Ouyang
- New York University School of Medicine, New York, New York
| | | | - William Chou
- University of California, San Francisco, San Francisco, California
| | - Manan S Patel
- New York University School of Medicine, New York, New York
| | | | - Sandra Demaria
- New York University School of Medicine, New York, New York
| | - Jian-Hua Mao
- Lawrence Berkeley National Laboratory, Berkeley, California
| | - Kubra Karagoz
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Michael L Gatza
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | | |
Collapse
|
53
|
Abstract
NKT cells are a small but influential member of the T cell family, recognizing lipids presented by the non-classical MHC-like molecule CD1d rather than peptides presented by classical MHC molecules. They bridge between the innate and adaptive immune systems, serving as rapid responders but also allowing the T cell immune system to recognize lipid antigens, for example derived from tumors or bacteria. They also serve as potent regulatory cells, controlling other immune responses. Type I NKT cells use a semi-invariant T cell receptor (TCR) whereas type II use diverse TCRs. Most often, type I NKT cells promote tumor immunity whereas type II tend to suppress it, and the two subtypes crossregulate each other, forming an immunoregulatory axis. Lack of tools to study these important cells has limited the understanding of these, but newer tools have allowed great advances, especially in mouse models. These range from transgenic and knock-out mice to CD1d tetramers carrying ligands for type I or II NKT cells, to antibodies and NKT cell hybridomas. Here we describe these complementary tools and approaches and their use to study NKT cells and their role in the immunology and immunotherapy of cancer.
Collapse
Affiliation(s)
- Jay A Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States.
| | - Purevdorj B Olkhanud
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Masaki Terabe
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
54
|
Wan J, Cai W, Wang H, Cheng J, Su Z, Wang S, Xu H. Role of type 2 innate lymphoid cell and its related cytokines in tumor immunity. J Cell Physiol 2019; 235:3249-3257. [PMID: 31625163 DOI: 10.1002/jcp.29287] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 09/27/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Jie Wan
- Department of Immunology Jiangsu University Zhenjiang China
| | - Wei Cai
- Department of Immunology Jiangsu University Zhenjiang China
| | - Huixuan Wang
- Department of Immunology Jiangsu University Zhenjiang China
| | - Jianjun Cheng
- Department of Immunology Jiangsu University Zhenjiang China
| | - Zhaoliang Su
- Department of Immunology Jiangsu University Zhenjiang China
- The Central Laboratory The Fourth Affiliated Hospital of Jiangsu University Zhenjiang China
| | - Shengjun Wang
- Department of Immunology Jiangsu University Zhenjiang China
- Department of Laboratory Medicine, The Affiliated People's Hospital Jiangsu University Zhenjiang China
| | - Huaxi Xu
- Department of Immunology Jiangsu University Zhenjiang China
| |
Collapse
|
55
|
Regulation of Immunity in Breast Cancer. Cancers (Basel) 2019; 11:cancers11081080. [PMID: 31366131 PMCID: PMC6721298 DOI: 10.3390/cancers11081080] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 07/24/2019] [Accepted: 07/26/2019] [Indexed: 12/24/2022] Open
Abstract
Breast cancer affects millions of women worldwide, leading to many deaths and significant economic burden. Although there are numerous treatment options available, the huge potentials of immunotherapy in the management of localized and metastatic breast cancer is currently being explored. However, there are significant gaps in understanding the complex interactions between the immune system and breast cancer. The immune system can be pro-tumorigenic and anti-tumorigenic depending on the cells involved and the conditions of the tumor microenvironment. In this review, we discuss current knowledge of breast cancer, including treatment options. We also give a brief overview of the immune system and comprehensively highlight the roles of different cells of the immune system in breast tumorigenesis, including recent research discoveries. Lastly, we discuss some immunotherapeutic strategies for the management of breast cancer.
Collapse
|
56
|
Huang S, Wang Z, Zhou J, Huang J, Zhou L, Luo J, Wan YY, Long H, Zhu B. EZH2 Inhibitor GSK126 Suppresses Antitumor Immunity by Driving Production of Myeloid-Derived Suppressor Cells. Cancer Res 2019; 79:2009-2020. [PMID: 30737232 DOI: 10.1158/0008-5472.can-18-2395] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 12/01/2018] [Accepted: 02/05/2019] [Indexed: 11/16/2022]
Abstract
Enhancer of zeste homolog (EZH2) is a key epigenetic regulator of gene expression and is frequently overexpressed in various cancer types, suggesting a role in oncogenesis. The therapeutic potential of EZH2 inhibitors is currently being explored, but their effect on antitumor immunity is largely unknown. Here we report that suppressing EZH2 activity using EZH2 inhibitor GSK126 resulted in increased numbers of myeloid-derived suppressor cells (MDSC) and fewer CD4+ and IFNγ+CD8+ T cells, which are involved in antitumor immunity. Addition of a neutralizing antibody against the myeloid differentiation antigen GR-1 or gemcitabine/5-fluorouracil-depleted MDSCs alleviated MDSC-mediated immunosuppression and increased CD4+ and CD8+ T-cell tumor infiltration and GSK126 therapeutic efficacy. Mechanistically, we identified a novel pathway of MDSC production in cancer in which EZH2 inhibition directs myeloid differentiation from primitive hematopoietic progenitor cells. These findings suggest that modulating the tumor immune microenvironment may improve the efficacy of EZH2 inhibitors. SIGNIFICANCE: This study uncovers a potential mechanism behind disappointing results of a phase I clinical trial of EZH2 inhibitor GSK126 and identifies a translatable combinational strategy to overcome it.
Collapse
Affiliation(s)
- Shuo Huang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China.,Chongqing Key Laboratory of Immunotherapy, Chongqing, China
| | - Zhongyu Wang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China.,Chongqing Key Laboratory of Immunotherapy, Chongqing, China
| | - Jie Zhou
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China.,Chongqing Key Laboratory of Immunotherapy, Chongqing, China
| | - Jiani Huang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China.,Chongqing Key Laboratory of Immunotherapy, Chongqing, China
| | - Li Zhou
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China.,Chongqing Key Laboratory of Immunotherapy, Chongqing, China
| | - Jing Luo
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China.,Chongqing Key Laboratory of Immunotherapy, Chongqing, China
| | - Yisong Y Wan
- Department of Microbiology and Immunology, Lineberger Comprehensive Cancer Centre, University of North Carolina at Chapel Hill, Chapel Hill, North California
| | - Haixia Long
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China. .,Chongqing Key Laboratory of Immunotherapy, Chongqing, China
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China. .,Chongqing Key Laboratory of Immunotherapy, Chongqing, China
| |
Collapse
|
57
|
The mechanistic study behind suppression of GVHD while retaining GVL activities by myeloid-derived suppressor cells. Leukemia 2019; 33:2078-2089. [PMID: 30737483 PMCID: PMC6687551 DOI: 10.1038/s41375-019-0394-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 01/09/2019] [Accepted: 01/18/2019] [Indexed: 12/25/2022]
Abstract
Graft-versus-host disease (GVHD) is a major barrier to the widespread use of allogeneic hematopoietic stem cell transplantation (allo-HSCT) for treating hematologic malignancies. Myeloid-derived suppressor cells (MDSCs) have been recognized as crucial immunosuppressive cells in various pathologic settings. Here, we investigated whether the unique functional properties of MDSCs could be harnessed to control allo-HSCT-associated GVHD. Using multiple murine GVHD/GVL models including both MHC-mismatched and miHA-mismatched, we demonstrated that treatment with CD115+ MDSCs efficiently suppressed GVHD but did not significantly impair graft-versus-leukemia (GVL) activity, leading to 80% and 67% protection in treated mice in GVHD and GVL models, respectively. The mechanism for this dissociation of GVHD from GVL, specifically the emergence of donor-derived NKG2D+ CD8 T cells with a memory phenotype in MDSC-treated recipient mice, was identified. NKG2D expression on donor T cells was required for eradication of allogeneic lymphoma cells. Furthermore, long-term surviving MDSC recipients that exhibited cytolytic activities against allogeneic leukemia cells had a significantly increased percentage of T regulatory cells and, more importantly, NKG2D+ CD8 T cells. These findings indicate that MDSCs can be used as a novel cell-based therapy to suppress GVHD while maintaining GVL activities through selective induction of NKG2D+ CD8 memory T cells.
Collapse
|
58
|
Impairment of Vα24-Jα18+Vβ11+ natural killer T cells in adult acute lymphoblastic leukemia patients. Exp Cell Res 2019; 374:69-75. [DOI: 10.1016/j.yexcr.2018.11.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 11/08/2018] [Accepted: 11/09/2018] [Indexed: 11/19/2022]
|
59
|
Valdes-Mora F, Handler K, Law AMK, Salomon R, Oakes SR, Ormandy CJ, Gallego-Ortega D. Single-Cell Transcriptomics in Cancer Immunobiology: The Future of Precision Oncology. Front Immunol 2018; 9:2582. [PMID: 30483257 PMCID: PMC6240655 DOI: 10.3389/fimmu.2018.02582] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 10/19/2018] [Indexed: 12/21/2022] Open
Abstract
Cancer is a heterogeneous and complex disease. Tumors are formed by cancer cells and a myriad of non-cancerous cell types that together with the extracellular matrix form the tumor microenvironment. These cancer-associated cells and components contribute to shape the progression of cancer and are deeply involved in patient outcome. The immune system is an essential part of the tumor microenvironment, and induction of cancer immunotolerance is a necessary step involved in tumor formation and growth. Immune mechanisms are intimately associated with cancer progression, invasion, and metastasis; as well as to tumor dormancy and modulation of sensitivity to drug therapy. Transcriptome analyses have been extensively used to understand the heterogeneity of tumors, classifying tumors into molecular subtypes and establishing signatures that predict response to therapy and patient outcomes. However, the classification of the tumor cell diversity and specially the identification of rare populations has been limited in these transcriptomic analyses of bulk tumor cell populations. Massively-parallel single-cell RNAseq analysis has emerged as a powerful method to unravel heterogeneity and to study rare cell populations in cancer, through unsupervised sampling and modeling of transcriptional states in single cells. In this context, the study of the role of the immune system in cancer would benefit from single cell approaches, as it will enable the characterization and/or discovery of the cell types and pathways involved in cancer immunotolerance otherwise missed in bulk transcriptomic information. Thus, the analysis of gene expression patterns at single cell resolution holds the potential to provide key information to develop precise and personalized cancer treatment including immunotherapy. This review is focused on the latest single-cell RNAseq methodologies able to agnostically study thousands of tumor cells as well as targeted single-cell RNAseq to study rare populations within tumors. In particular, we will discuss methods to study the immune system in cancer. We will also discuss the current challenges to the study of cancer at the single cell level and the potential solutions to the current approaches.
Collapse
Affiliation(s)
- Fatima Valdes-Mora
- Genomics and Epigenetics Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Kristina Handler
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Andrew M K Law
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Robert Salomon
- Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Samantha R Oakes
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia.,The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Christopher J Ormandy
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia.,The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - David Gallego-Ortega
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia.,The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| |
Collapse
|
60
|
Yen CH, Hsiao HH. NRF2 Is One of the Players Involved in Bone Marrow Mediated Drug Resistance in Multiple Myeloma. Int J Mol Sci 2018; 19:E3503. [PMID: 30405034 PMCID: PMC6274683 DOI: 10.3390/ijms19113503] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 10/28/2018] [Accepted: 11/04/2018] [Indexed: 02/07/2023] Open
Abstract
Multiple myeloma with clonal plasma expansion in bone marrow is the second most common hematologic malignancy in the world. Though the improvement of outcomes from the achievement of novel agents in recent decades, the disease progresses and leads to death eventually due to the elusive nature of myeloma cells and resistance mechanisms to therapeutic agents. In addition to the molecular and genetic basis of resistance pathomechanisms, the bone marrow microenvironment also contributes to disease progression and confers drug resistance in myeloma cells. In this review, we focus on the current state of the literature in terms of critical bone marrow microenvironment components, including soluble factors, cell adhesion mechanisms, and other cellular components. Transcriptional factor nuclear factor erythroid-derived-2-like 2 (NRF2), a central regulator for anti-oxidative stresses and detoxification, is implicated in chemoresistance in several cancers. The functional roles of NRF2 in myeloid-derived suppressor cells and multiple myeloma cells, and the potential of targeting NRF2 for overcoming microenvironment-mediated drug resistance in multiple myeloma are also discussed.
Collapse
Affiliation(s)
- Chia-Hung Yen
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
- Center for Infectious Disease and Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan.
| | - Hui-Hua Hsiao
- Division of Hematology-Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan.
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| |
Collapse
|
61
|
Stolk D, van der Vliet HJ, de Gruijl TD, van Kooyk Y, Exley MA. Positive & Negative Roles of Innate Effector Cells in Controlling Cancer Progression. Front Immunol 2018; 9:1990. [PMID: 30298063 PMCID: PMC6161645 DOI: 10.3389/fimmu.2018.01990] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 08/13/2018] [Indexed: 12/29/2022] Open
Abstract
Innate immune cells are active at the front line of host defense against pathogens and now appear to play a range of roles under non-infectious conditions as well, most notably in cancer. Establishing the balance of innate immune responses is critical for the “flavor” of these responses and subsequent adaptive immunity and can be either “good or bad” in controlling cancer progression. The importance of innate NK cells in tumor immune responses has already been extensively studied over the last few decades, but more recently several relatively mono- or oligo-clonal [i.e., (semi-) invariant] innate T cell subsets received substantial interest in tumor immunology including invariant natural killer T (iNKT), γδ-T and mucosal associated invariant T (MAIT) cells. These subsets produce high levels of various pro- and/or anti-inflammatory cytokines/chemokines reflecting their capacity to suppress or stimulate immune responses. Survival of patients with cancer has been linked to the frequencies and activation status of NK, iNKT, and γδ-T cells. It has become clear that NK, iNKT, γδ-T as well as MAIT cells all have physiological roles in anti-tumor responses, which emphasize their possible relevance for tumor immunotherapy. A variety of clinical trials has focused on manipulating NK, iNKT, and γδ-T cell functions as a cancer immunotherapeutic approach demonstrating their safety and potential for achieving beneficial therapeutic effects, while the exploration of MAIT cell related therapies is still in its infancy. Current issues limiting the full therapeutic potential of these innate cell subsets appear to be related to defects and suppressive properties of these subsets that, with the right stimulus, might be reversed. In general, how innate lymphocytes are activated appears to control their subsequent abilities and consequent impact on adaptive immunity. Controlling these potent regulators and mediators of the immune system should enable their protective roles to dominate and their deleterious potential (in the specific context of cancer) to be mitigated.
Collapse
Affiliation(s)
- Dorian Stolk
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, Netherlands
| | - Hans J van der Vliet
- Department of Medical Oncology, VU University Medical Center, Amsterdam, Netherlands
| | - Tanja D de Gruijl
- Department of Medical Oncology, VU University Medical Center, Amsterdam, Netherlands
| | - Yvette van Kooyk
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, Netherlands
| | - Mark A Exley
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom.,Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States.,Agenus, Inc., Lexington, MA, United States
| |
Collapse
|
62
|
Chen X, Wang L, Li P, Song M, Qin G, Gao Q, Zhang Z, Yue D, Wang D, Nan S, Qi Y, Li F, Yang L, Huang L, Zhang M, Zhang B, Gao Y, Zhang Y. Dual TGF-β and PD-1 blockade synergistically enhances MAGE-A3-specific CD8 + T cell response in esophageal squamous cell carcinoma. Int J Cancer 2018; 143:2561-2574. [PMID: 29981155 DOI: 10.1002/ijc.31730] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 06/16/2018] [Accepted: 06/21/2018] [Indexed: 12/26/2022]
Abstract
PD-1 is highly expressed on tumor-infiltrated antigen-specific T cells and limit the antitumor function. Blocking of PD-1/PD-L1 signaling has shown unprecedented curative efficacies in patients with advanced cancer. However, only a limited population of patients benefited from such therapies. Our study aimed to explore biological properties, functional regulation and reversal of MAGE-A3-specific CD8+ T cells in patients with esophageal squamous cell carcinoma (ESCC). The underlying principle of deficiency and restoring MAGE-A3-specific CD8+ T cells function in tumor microenvironment (TME) was evaluated. MAGE-A3-specific CD8+ T cells could lyse HLA-A2+ /MAGE-A3+ tumor cells. Tetramer+ T cell frequency was higher in elder patients, but lower in patients with lymph node metastasis and late tumor stage (p < 0.05). CD107ahigh expression on functional T cells was an independent prognostic factor in Cox regression analysis. PD-1 was highly expressed on dysfunctional antigen-specific CD8+ T cells and tumor infiltrating T lymphocytes (p < 0.05). Myeloid-derived suppressor cells (MDSCs) derived-TGF-β mediated PD-1high expression on CD8+ T cells, which led to be resistance to PD-1/PD-L1 blockade in TME. Dual PD-1/PD-L1 and TGF-β signaling pathway blockades synergistically restored the function and antitumor ability of antigen-specific CD8+ T cells in vitro/vivo assay. The presence of functional MAGE-A3-specific CD8+ T cells had an independent prognostic impact on survival of patients with ESCC. Furthermore, MDSCs-derived TGF-β increased PD-1 expression on T cells and decreased the sensitivity to PD-1/PD-L1 blockade. Combining T cell-based therapy with dual PD-1/PD-L1 and TGF-β signaling pathway blockade could be considered a promising strategy for cancer treatment.
Collapse
Affiliation(s)
- Xinfeng Chen
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Liping Wang
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Pupu Li
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Mengjia Song
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Guohui Qin
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qun Gao
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhen Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Dongli Yue
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Dan Wang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shufeng Nan
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yu Qi
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Feng Li
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Li Yang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lan Huang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Mingzhi Zhang
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Bin Zhang
- Department of Hematology/Oncology, School of Medicine, Northwestern University, Chicago, IL
| | - Yanfeng Gao
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China.,Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, Henan, China
| |
Collapse
|
63
|
Terabe M, Berzofsky JA. Tissue-Specific Roles of NKT Cells in Tumor Immunity. Front Immunol 2018; 9:1838. [PMID: 30158927 PMCID: PMC6104122 DOI: 10.3389/fimmu.2018.01838] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 07/25/2018] [Indexed: 01/07/2023] Open
Abstract
NKT cells are an unusual population of T cells recognizing lipids presented by CD1d, a non-classical class-I-like molecule, rather than peptides presented by conventional MHC molecules. Type I NKT cells use a semi-invariant T cell receptor and almost all recognize a common prototype lipid, α-galactosylceramide (α-GalCer). Type II NKT cells are any lipid-specific CD1d-restricted T cells that use other receptors and generally don't recognize α-GalCer. They play important regulatory roles in immunity, including tumor immunity. In contrast to type I NKT cells that most have found to promote antitumor immunity, type II NKT cells suppress tumor immunity and the two subsets cross-regulate each other, forming an immunoregulatory axis. They also can promote other regulatory cells including regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs), and can induce MDSCs to secrete TGF-β, one of the most immunosuppressive cytokines known. In some tumors, both Tregs and type II NKT cells can suppress immunosurveillance, and the balance between these is determined by a type I NKT cell. We have also seen that regulation of tumor immunity can depend on the tissue microenvironment, so the same tumor in the same animal in different tissues may be regulated by different cells, such as type II NKT cells in the lung vs Tregs in the skin. Also, the effector T cells that protect those sites when Tregs are removed do not always act between tissues even in the same animal. Thus, metastases may require different immunotherapy from primary tumors. Newly improved sulfatide-CD1d tetramers are starting to allow better characterization of the elusive type II NKT cells to better understand their function and control it to overcome immunosuppression.
Collapse
Affiliation(s)
- Masaki Terabe
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Jay A Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
64
|
Shen Y, Li J, Wang SQ, Jiang W. Ambiguous roles of innate lymphoid cells in chronic development of liver diseases. World J Gastroenterol 2018; 24:1962-1977. [PMID: 29760540 PMCID: PMC5949710 DOI: 10.3748/wjg.v24.i18.1962] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 04/25/2018] [Accepted: 05/06/2018] [Indexed: 02/06/2023] Open
Abstract
Innate lymphoid cells (ILCs) are defined as a distinct arm of innate immunity. According to their profile of secreted cytokines and lineage-specific transcriptional factors, ILCs can be categorized into the following three groups: group 1 ILCs (including natural killer (NK) cells and ILC1s) are dependent on T-bet and can produce interferon-γ; group 2 ILCs (ILC2s) are dependent on GATA3 and can produce type 2 cytokines, including interleukin (IL)-5 and IL-13; and, group 3 ILCs (including lymphoid tissue-like cells and ILC3s) are dependent on RORγt and can produce IL-22 and IL-17. Collaborative with adaptive immunity, ILCs are highly reactive innate effectors that promptly orchestrate immunity, inflammation and tissue repair. Dysregulation of ILCs might result in inflammatory disorders. Evidence regarding the function of intrahepatic ILCs is emerging from longitudinal studies of inflammatory liver diseases wherein they exert both physiological and pathological functions, including immune homeostasis, defenses and surveillance. Their overall effect on the liver depends on the balance of their proinflammatory and antiinflammatory populations, specific microenvironment and stages of immune responses. Here, we review the current data about ILCs in chronic liver disease progression, to reveal their roles in different stages as well as to discuss their therapeutic potency as intervention targets.
Collapse
Affiliation(s)
- Yue Shen
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jing Li
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of Gastroenterology, Tongji Hospital, Tongji University, Shanghai 200000, China
| | - Si-Qi Wang
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Wei Jiang
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
65
|
Servais L, Wéra O, Dibato Epoh J, Delierneux C, Bouznad N, Rahmouni S, Mazzucchelli G, Baiwir D, Delvenne P, Lancellotti P, Oury C. Platelets contribute to the initiation of colitis-associated cancer by promoting immunosuppression. J Thromb Haemost 2018; 16:762-777. [PMID: 29369476 DOI: 10.1111/jth.13959] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Indexed: 02/06/2023]
Abstract
Essentials Inflammation plays a key role in the development of colorectal cancer. Understanding mechanisms of cancer initiation might reveal new anticancer preventive strategy. Hyperactive platelets promote tumor formation by fostering immune evasion of cancer. Platelet inhibition by clopidogrel prevents carcinogenesis by restoring antitumor immunity. SUMMARY Background Clinical and experimental evidence support a role for inflammation in the development of colorectal cancer, although the mechanisms are not fully understood. Beyond thrombosis and hemostasis, platelets are key actors in inflammation; they have also been shown to be involved in cancer. However, whether platelets participate in the link between inflammation and cancer is unknown. Objective To investigate the contribution of platelets and platelet-derived proteins to inflammation-elicited colorectal tumor development. Methods We used a clinically relevant mouse model of colitis-associated cancer. Platelet secretion and platelet reactivity to thrombin were assessed at each stage of carcinogenesis. We conducted an unbiased proteomic analysis of releasates of platelets isolated at the pretumoral stage to identify soluble factors that might act on tumor development. Plasma levels of the identified proteins were measured during the course of carcinogenesis. We then treated the mice with clopidogrel to efficiently inhibit platelet release reaction. Results At the pretumoral stage, hyperactive platelets constituted a major source of circulating protumoral serum amyloid A (SAA) proteins. Clopidogrel prevented the early elevation of the plasma SAA protein level, decreased colitis severity, and delayed the formation of dysplastic lesions and adenocarcinoma. Platelet inhibition hindered the expansion and function of immunosuppressive myeloid cells, as well as their infiltration into tumors, but increased the number of tissue CD8+ T cells. Platelets and releasates of platelets from mice with cancer were both able to polarize myeloid cells towards an immunosuppressive phenotype. Conclusions Thus, platelets promote the initiation of colitis-associated cancer by enhancing myeloid cell-dependent immunosuppression. Antiplatelet agents may help to prevent inflammation-elicited carcinogenesis by restoring antitumor immunity.
Collapse
Affiliation(s)
- L Servais
- Laboratory of Thrombosis and Hemostasis and Valvular Heart Disease, GIGA-Cardiovascular Sciences, Department of Cardiology, University of Liège, CHU Sart-Tilman, Liège, Belgium
| | - O Wéra
- Laboratory of Thrombosis and Hemostasis and Valvular Heart Disease, GIGA-Cardiovascular Sciences, Department of Cardiology, University of Liège, CHU Sart-Tilman, Liège, Belgium
| | - J Dibato Epoh
- Laboratory of Thrombosis and Hemostasis and Valvular Heart Disease, GIGA-Cardiovascular Sciences, Department of Cardiology, University of Liège, CHU Sart-Tilman, Liège, Belgium
| | - C Delierneux
- Laboratory of Thrombosis and Hemostasis and Valvular Heart Disease, GIGA-Cardiovascular Sciences, Department of Cardiology, University of Liège, CHU Sart-Tilman, Liège, Belgium
| | - N Bouznad
- Laboratory of Thrombosis and Hemostasis and Valvular Heart Disease, GIGA-Cardiovascular Sciences, Department of Cardiology, University of Liège, CHU Sart-Tilman, Liège, Belgium
| | - S Rahmouni
- Immunology and Infectious Diseases Unit, GIGA-R, University of Liège, Liège, Belgium
| | - G Mazzucchelli
- Laboratory of Mass Spectrometry, Chemistry Department, GIGA-R, CART, University of Liège, Liège, Belgium
| | - D Baiwir
- Laboratory of Mass Spectrometry, Chemistry Department, GIGA-R, CART, University of Liège, Liège, Belgium
| | - P Delvenne
- Department of Pathology, Laboratory of Experimental Pathology, University of Liège, Liège, Belgium
| | - P Lancellotti
- Laboratory of Thrombosis and Hemostasis and Valvular Heart Disease, GIGA-Cardiovascular Sciences, Department of Cardiology, University of Liège, CHU Sart-Tilman, Liège, Belgium
- Gruppo Villa Maria Care and Reseach, Anthea Hospital, Bari, Italy
| | - C Oury
- Laboratory of Thrombosis and Hemostasis and Valvular Heart Disease, GIGA-Cardiovascular Sciences, Department of Cardiology, University of Liège, CHU Sart-Tilman, Liège, Belgium
| |
Collapse
|
66
|
Yadav AS, Pandey PR, Butti R, Radharani NNV, Roy S, Bhalara SR, Gorain M, Kundu GC, Kumar D. The Biology and Therapeutic Implications of Tumor Dormancy and Reactivation. Front Oncol 2018; 8:72. [PMID: 29616190 PMCID: PMC5868535 DOI: 10.3389/fonc.2018.00072] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 03/02/2018] [Indexed: 01/06/2023] Open
Abstract
Advancements in the early detection of cancer coupled with improved surgery, radiotherapy, and adjuvant therapy led to substantial increase in patient survival. Nevertheless, cancer metastasis is the leading cause of death in several cancer patients. The majority of these deaths are associated with metastatic relapse kinetics after a variable period of clinical remission. Most of the cancer recurrences are thought to be associated with the reactivation of dormant disseminated tumor cells (DTCs). In this review, we have summarized the cellular and molecular mechanisms related to DTCs and the role of microenvironmental niche. These mechanisms regulate the dormant state and help in the reactivation, which leads to metastatic outgrowth. Identification of novel therapeutic targets to eliminate these dormant tumor cells will be highly useful in controlling the metastatic relapse-related death with several cancers.
Collapse
Affiliation(s)
- Amit S. Yadav
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science, Pune, India
| | - Poonam R. Pandey
- Laboratory of Genetics, National Institute on Aging-Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Ramesh Butti
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science, Pune, India
| | - N. N. V. Radharani
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science, Pune, India
| | - Shamayita Roy
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science, Pune, India
| | - Shaileshkumar R. Bhalara
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science, Pune, India
| | - Mahadeo Gorain
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science, Pune, India
| | - Gopal C. Kundu
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science, Pune, India
| | - Dhiraj Kumar
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science, Pune, India
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
67
|
Maeng H, Terabe M, Berzofsky JA. Cancer vaccines: translation from mice to human clinical trials. Curr Opin Immunol 2018; 51:111-122. [PMID: 29554495 DOI: 10.1016/j.coi.2018.03.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 02/13/2018] [Accepted: 03/01/2018] [Indexed: 01/22/2023]
Abstract
Therapeutic cancer vaccines have been a long-sought approach to harness the exquisite specificity of the immune system to treat cancer, but until recently have not had much success as single agents in clinical trials. However, new understanding of the immunoregulatory mechanisms exploited by cancers has allowed the development of approaches to potentiate the effect of vaccines by removing the brakes while the vaccines step on the accelerator. Thus, vaccines that had induced a strong T cell response but no clinical therapeutic effect may now reach their full potential. Here, we review a number of promising approaches to cancer vaccines developed initially in mouse models and their translation into clinical trials, along with combinations of vaccines with other therapies that might allow cancer vaccines to finally achieve clinical efficacy against many types of cancer.
Collapse
Affiliation(s)
- Hoyoung Maeng
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, United States
| | - Masaki Terabe
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, United States
| | - Jay A Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, United States.
| |
Collapse
|
68
|
Krijgsman D, Hokland M, Kuppen PJK. The Role of Natural Killer T Cells in Cancer-A Phenotypical and Functional Approach. Front Immunol 2018. [PMID: 29535734 PMCID: PMC5835336 DOI: 10.3389/fimmu.2018.00367] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Natural killer T (NKT) cells are a subset of CD1d-restricted T cells at the interface between the innate and adaptive immune system. NKT cells can be subdivided into functional subsets that respond rapidly to a wide variety of glycolipids and stress-related proteins using T- or natural killer (NK) cell-like effector mechanisms. Because of their major modulating effects on immune responses via secretion of cytokines, NKT cells are also considered important players in tumor immunosurveillance. During early tumor development, T helper (TH)1-like NKT cell subsets have the potential to rapidly stimulate tumor-specific T cells and effector NK cells that can eliminate tumor cells. In case of tumor progression, NKT cells may become overstimulated and anergic leading to deletion of a part of the NKT cell population in patients via activation-induced cell death. In addition, the remaining NKT cells become hyporesponsive, or switch to immunosuppressive TH2-/T regulatory-like NKT cell subsets, thereby facilitating tumor progression and immune escape. In this review, we discuss this important role of NKT cells in tumor development and we conclude that there should be three important focuses of future research in cancer patients in relation with NKT cells: (1) expansion of the NKT cell population, (2) prevention and breaking of NKT cell anergy, and (3) skewing of NKT cells toward TH1-like subsets with antitumor activity.
Collapse
Affiliation(s)
- Daniëlle Krijgsman
- Department of Surgery, Leiden University Medical Center, Leiden, Netherlands.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Peter J K Kuppen
- Department of Surgery, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
69
|
Kato S, Berzofsky JA, Terabe M. Possible Therapeutic Application of Targeting Type II Natural Killer T Cell-Mediated Suppression of Tumor Immunity. Front Immunol 2018; 9:314. [PMID: 29520281 PMCID: PMC5827362 DOI: 10.3389/fimmu.2018.00314] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 02/05/2018] [Indexed: 12/17/2022] Open
Abstract
Natural killer T (NKT) cells are a unique T cell subset that exhibits characteristics from both the innate immune cells and T cells. There are at least two subsets of NKT cells, type I and type II. These two subsets of NKT cells have opposite functions in antitumor immunity. Type I NKT cells usually enhance and type II NKT cells suppress antitumor immunity. In addition, these two subsets of NKT cells cross-regulate each other. In this review, we mainly focus on immunosuppressive NKT cells, type II NKT cells. After summarizing their definition, experimental tools to study them, and subsets of them, we will discuss possible therapeutic applications of type II NKT cell pathway targeted therapies.
Collapse
Affiliation(s)
- Shingo Kato
- Department of Gastroenterology and Hepatology, Yokohama City University School of Medicine, Yokohama, Japan
| | - Jay A. Berzofsky
- Molecular Immunogenetics and Vaccine Research Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Masaki Terabe
- Molecular Immunogenetics and Vaccine Research Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
70
|
Khan MA, Aljarbou AN, Aldebasi YH, Alorainy MS, Rahmani AH, Younus H, Khan A. Liposomal formulation of glycosphingolipids from Sphingomonas paucimobilis induces antitumour immunity in mice. J Drug Target 2018; 26:709-719. [PMID: 29307241 DOI: 10.1080/1061186x.2018.1424857] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Natural Killer T (NKT) cells play an important role in host's anti-tumour immune response. Glycosphingolipids (GSLs) isolated from Sphingomonas paucimobilis have the ability to stimulate NKT cells. In this study, the activity of free GSLs or GSLs-incorporated liposomes (glycosphingosomes) was investigated against dimethyl-α-benzanthracene (DMBA)-induced tumours in mice. The anti-tumour immunity of GSLs- or glycosphingosomes-loaded bone marrow-derived dendritic cells (BMDCs) was investigated in tumour-bearing mice. The Immunotherapeutic potential of co-administration of liposomal doxorubicin (Lip-Dox) and GSLs or glycosphingosomes was assessed by measuring cytokine levels and VEGF in the tumour tissues. Pretreatment with glycosphingosomes significantly delayed the frequency of tumour formation. Immunotherapy with glycosphingosomes-loaded BMDCs increased serum IFN-γ level and survival rate in mice. The effect of immunotherapy was dependent on effector functions of NK cells because the depletion of NK cells abolished the effects of immunotherapy. There was reduced tumour growth with low expression of VEGF in the group of mice treated with glycosphingosomes and Lip-Dox combination. Moreover, the splenocytes secreted higher levels of IFN-γ, IL-12 and lower TGF-β level. The results of this study indicate that glycosphingosomes can induce better antitumour immunity and may be considered a novel formulation in antitumour therapy.
Collapse
Affiliation(s)
- Masood A Khan
- a College of Applied Medical Sciences , Qassim University , Buraydah , Saudi Arabia
| | - Ahmed N Aljarbou
- b College of Pharmacy , Qassim University , Buraydah , Saudi Arabia
| | - Yousef H Aldebasi
- a College of Applied Medical Sciences , Qassim University , Buraydah , Saudi Arabia
| | | | - Arshad H Rahmani
- a College of Applied Medical Sciences , Qassim University , Buraydah , Saudi Arabia
| | - Hina Younus
- d Interdisciplinary Biotechnology Unit , Aligarh Muslim University , Aligarh , India
| | - Arif Khan
- a College of Applied Medical Sciences , Qassim University , Buraydah , Saudi Arabia
| |
Collapse
|
71
|
Transforming Growth Factor β is a Poor Prognostic Factor and Inhibits the Favorable Prognostic Value of CD8+ CTL in Human Hepatocellular Carcinoma. J Immunother 2018; 40:175-186. [PMID: 28394813 DOI: 10.1097/cji.0000000000000166] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
It is widely understood that transforming growth factor β (TGF-β) has dual functions in tumors-tumor promoter or tumor suppressor. As a tumor promoter, TGF-β drives tumor initiation and progression partially by suppressing the antitumor responses of CD8 cytotoxic T lymphocytes (CTLs) in the tumor microenvironment. Here, we investigated the prognostic value of measuring TGF-β and CD8 CTLs levels and their relationship in human hepatocellular carcinoma (HCC). Immunohistochemical staining was conducted to analyze the prognostic value of TGF-β expression and/or CD8 CTLs levels in 407 HCC patients. The relationship between TGF-β and CD8 T-cell was also evaluated using HCC cell lines and patients' peripheral blood. Lower TGF-β expression or a higher CD8 CTL density was associated with better overall survival and recurrence-free survival, and the patients with low TGF-β expression and more CD8 CTLs had the best prognosis. Although there was no correlation between TGF-β expression and the density of CD8 CTLs, the survival of patients with more CD8 CTL cells was only significantly improved when the tumor expressed low levels of TGF-β. Furthermore, the TGF-β levels was not associated with the proportion of CD8 T cells, but negatively related to interferon γ secretion by CD8 T cells in peripheral blood of HCC patients. Higher TGF-β also resulted in decreased interferon γ secreted by CD8 T cells in vitro. In conclusion, our study suggests that TGF-β is a poor prognostic factor for patients and negatively affect the prognostic value of CD8 CTLs through suppressing antitumor activity of CD8 T-cell in HCC.
Collapse
|
72
|
Wang Y, Cardell SL. The Yin and Yang of Invariant Natural Killer T Cells in Tumor Immunity-Suppression of Tumor Immunity in the Intestine. Front Immunol 2018; 8:1945. [PMID: 29375569 PMCID: PMC5767593 DOI: 10.3389/fimmu.2017.01945] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 12/18/2017] [Indexed: 12/11/2022] Open
Abstract
CD1d-restricted invariant natural killer T (iNKT) cells are known as early responding, potent regulatory cells of immune responses. Besides their established role in the regulation of inflammation and autoimmune disease, numerous studies have shown that iNKT cells have important functions in tumor immunosurveillance and control of tumor metastasis. Tumor-infiltrating T helper 1 (TH1)/cytotoxic T lymphocytes have been associated with a positive prognosis. However, inflammation has a dual role in cancer and chronic inflammation is believed to be a driving force in many cancers as exemplified in patients with inflammatory bowel disease that have an increased risk of colorectal cancer. Indeed, NKT cells promote intestinal inflammation in human ulcerative colitis, and the associated animal model, indicating that NKT cells may favor tumor development in intestinal tissue. In contrast to other cancers, recent data from animal models suggest that iNKT cells promote tumor formation in the intestine by supporting an immunoregulatory tumor microenvironment and suppressing TH1 antitumor immunity. Here, we review the role of iNKT cells in suppression of tumor immunity in light of iNKT-cell regulation of intestinal inflammation. We also discuss suppression of immunity in other situations as well as factors that may influence whether iNKT cells have a protective or an immunosuppressive and tumor-promoting role in tumor immunity.
Collapse
Affiliation(s)
- Ying Wang
- Department of Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden
| | - Susanna L Cardell
- Department of Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
73
|
Unique invariant natural killer T cells promote intestinal polyps by suppressing TH1 immunity and promoting regulatory T cells. Mucosal Immunol 2018; 11:131-143. [PMID: 28401935 PMCID: PMC5638666 DOI: 10.1038/mi.2017.34] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 03/14/2017] [Indexed: 02/04/2023]
Abstract
CD1d-restricted invariant natural killer T (iNKT) cells are known as potent early regulatory cells of immune responses. Besides the established roles in the regulation of inflammation and autoimmune disease, studies have shown that iNKT cells have important roles in tumor surveillance and the control of tumor metastasis. Here we found that the absence of iNKT cells markedly decreased the total number of intestinal polyps in APCMin/+ mice, a model for colorectal cancer. Polyp iNKT cells were enriched for interleukin-10 (IL-10)- and IL-17-producing cells, showed a distinct phenotype being CD4+, NK1.1- CD44int, and PD-1lo, and they were negative for the NKT cell transcription factor promyelocytic leukemia zinc-finger. The absence of iNKT cells was associated with a reduced frequency of regulatory T (Tregs) cells and lower expression levels of FoxP3 protein and transcript uniquely in the polyps, and a switch to an inflammatory macrophage phenotype. Moreover, in iNKT cell-deficient APCMin/+ mice, expression of T-helper (TH) 1-associated genes, such as IFN-γ and Nos2, was increased in polyps, concomitantly with elevated frequencies of conventional CD4+ and CD8+ T cells in this tissue. The results suggest that a population of regulatory iNKT cells locally promote intestinal polyp formation by enhancing Treg cells and immunosuppression of antitumor TH1 immunity.
Collapse
|
74
|
Biology of Myeloid-Derived Suppressor Cells. Oncoimmunology 2018. [DOI: 10.1007/978-3-319-62431-0_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
75
|
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: 4.6] [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
|
76
|
Shissler SC, Lee MS, Webb TJ. Mixed Signals: Co-Stimulation in Invariant Natural Killer T Cell-Mediated Cancer Immunotherapy. Front Immunol 2017; 8:1447. [PMID: 29163518 PMCID: PMC5671952 DOI: 10.3389/fimmu.2017.01447] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 10/17/2017] [Indexed: 12/31/2022] Open
Abstract
Invariant natural killer T (iNKT) cells are an integral component of the immune system and play an important role in antitumor immunity. Upon activation, iNKT cells can directly kill malignant cells as well as rapidly produce cytokines that stimulate other immune cells, making them a front line defense against tumorigenesis. Unfortunately, iNKT cell number and activity are reduced in multiple cancer types. This anergy is often associated with upregulation of co-inhibitory markers such as programmed death-1. Similar to conventional T cells, iNKT cells are influenced by the conditions of their activation. Conventional T cells receive signals through the following three types of receptors: (1) T cell receptor (TCR), (2) co-stimulation molecules, and (3) cytokine receptors. Unlike conventional T cells, which recognize peptide antigen presented by MHC class I or II, the TCRs of iNKT cells recognize lipid antigen in the context of the antigen presentation molecule CD1d (Signal 1). Co-stimulatory molecules can positively and negatively influence iNKT cell activation and function and skew the immune response (Signal 2). This study will review the background of iNKT cells and their co-stimulatory requirements for general function and in antitumor immunity. We will explore the impact of monoclonal antibody administration for both blocking inhibitory pathways and engaging stimulatory pathways on iNKT cell-mediated antitumor immunity. This review will highlight the incorporation of co-stimulatory molecules in antitumor dendritic cell vaccine strategies. The use of co-stimulatory intracellular signaling domains in chimeric antigen receptor-iNKT therapy will be assessed. Finally, we will explore the influence of innate-like receptors and modification of immunosuppressive cytokines (Signal 3) on cancer immunotherapy.
Collapse
Affiliation(s)
- Susannah C Shissler
- Department of Microbiology and Immunology, Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Michael S Lee
- Department of Microbiology and Immunology, Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Tonya J Webb
- Department of Microbiology and Immunology, Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
77
|
Weng X, He Y, Visvabharathy L, Liao CM, Tan X, Balakumar A, Wang CR. Crosstalk between type II NKT cells and T cells leads to spontaneous chronic inflammatory liver disease. J Hepatol 2017; 67:791-800. [PMID: 28596110 PMCID: PMC5605413 DOI: 10.1016/j.jhep.2017.05.024] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Revised: 04/28/2017] [Accepted: 05/23/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIM Natural killer T (NKT) cells are CD1d-restricted innate-like T cells that modulate innate and adaptive immune responses. Unlike the well-characterized invariant/type I NKT cells, type II NKT cells with a diverse T cell receptor repertoire are poorly understood. This study defines the pathogenic role of type II NKT cells in the etiology of chronic liver inflammation. METHODS Transgenic mice with the Lck promoter directing CD1d overexpression on T cells in Jα18 wild-type (Lck-CD1dTgJα18+; type I NKT cell sufficient) and Jα18-deficient (Lck-CD1dTgJα18o, type I NKT cell deficient) mice were analyzed for liver pathology and crosstalk between type II NKT cells and conventional T cells. CD1d expression on T cells in peripheral blood samples and liver sections from autoimmune hepatitis patients and healthy individuals were also examined. RESULTS Lck-CD1dTgJα18o and Lck-CD1dTgJα18+ mice developed similar degrees of liver pathology resembling chronic autoimmune hepatitis in humans. Increased CD1d expression on T cells promoted the activation of type II NKT cells and other T cells. This resulted in Th1-skewing and impaired Th2 cytokine production in type II NKT cells. Dysfunction of type II NKT cells was accompanied by conventional T cell activation and pro-inflammatory cytokine production, leading to a hepatic T/B lymphocyte infiltration, elevated autoantibodies and hepatic injury in Lck-CD1dTg mice. A similar mechanism could be extended to humans as CD1d expression is upregulated on activated human T cells and increased presence of CD1d-expressing T cells was observed in autoimmune hepatitis patients. CONCLUSIONS Our data reveals enhanced crosstalk between type II NKT cells and conventional T cells, leading to a Th1-skewed inflammatory milieu, and consequently, to the development of chronic autoimmune liver disease. Lay summary: CD1d overexpression on T cells enhances crosstalk between type II NKT cells and T cells, resulting in their aberrant activation and leading to the development of chronic autoimmune liver disease.
Collapse
Affiliation(s)
- Xiufang Weng
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States; Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Ying He
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| | - Lavanya Visvabharathy
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| | - Chia-Min Liao
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| | - Xiaosheng Tan
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Arjun Balakumar
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| | - Chyung-Ru Wang
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States.
| |
Collapse
|
78
|
Nair S, Dhodapkar MV. Natural Killer T Cells in Cancer Immunotherapy. Front Immunol 2017; 8:1178. [PMID: 29018445 PMCID: PMC5614937 DOI: 10.3389/fimmu.2017.01178] [Citation(s) in RCA: 173] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 09/06/2017] [Indexed: 12/27/2022] Open
Abstract
Natural killer T (NKT) cells are specialized CD1d-restricted T cells that recognize lipid antigens. Following stimulation, NKT cells lead to downstream activation of both innate and adaptive immune cells in the tumor microenvironment. This has impelled the development of NKT cell-targeted immunotherapies for treating cancer. In this review, we provide a brief overview of the stimulatory and regulatory functions of NKT cells in tumor immunity as well as highlight preclinical and clinical studies based on NKT cells. Finally, we discuss future perspectives to better harness the potential of NKT cells for cancer therapy.
Collapse
Affiliation(s)
- Shiny Nair
- Yale University, New Haven, CT, United States
| | | |
Collapse
|
79
|
Hogg A, Sui Y, Ben-Sasson SZ, Paul WE, Berzofsky JA. Role of CD4 T cell helper subsets in immune response and deviation of CD8 T cells in mice. Eur J Immunol 2017; 47:2059-2069. [PMID: 28741316 DOI: 10.1002/eji.201747091] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 06/15/2017] [Accepted: 07/19/2017] [Indexed: 02/02/2023]
Abstract
The ability of different CD4+ T cell subsets to help CD8+ T-cell response is not fully understood. Here, we found using the murine system that Th17 cells induced by IL-1β, unlike Th1, were not effective helpers for antiviral CD8 responses as measured by IFNγ-producing cells or protection against virus infection. However, they skewed CD8 responses to a Tc17 phenotype. Thus, the apparent lack of help was actually immune deviation. This skewing depended on both IL-21 and IL-23. To overcome this effect, we inhibited Th17 induction by blocking TGF-β. Anti-TGF-β allowed the IL-1β adjuvant to enhance CD8+ T-cell responses without skewing the phenotype to Tc17, thereby providing an approach to harness the benefit of common IL-1-inducing adjuvants like alum without immune deviation.
Collapse
Affiliation(s)
- Alison Hogg
- Vaccine Branch, Center for Cancer Research, National Cancer Institute
| | - Yongjun Sui
- Vaccine Branch, Center for Cancer Research, National Cancer Institute
| | - Shlomo Z Ben-Sasson
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.,Lautenberg Center for General and Tumor Immunology, Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| | - William E Paul
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jay A Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute
| |
Collapse
|
80
|
Benson Z, Manjili SH, Habibi M, Guruli G, Toor AA, Payne KK, Manjili MH. Conditioning neoadjuvant therapies for improved immunotherapy of cancer. Biochem Pharmacol 2017; 145:12-17. [PMID: 28803721 DOI: 10.1016/j.bcp.2017.08.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 08/08/2017] [Indexed: 12/19/2022]
Abstract
Recent advances in the treatment of melanoma and non-small cell lung cancer (NSCLC) by combining conventional therapies with anti-PD1/PD-L1 immunotherapies, have renewed interests in immunotherapy of cancer. The emerging concept of conventional cancer therapies combined with immunotherapy differs from the classical concept in that it is not simply taking advantage of their additive anti-tumor effects, but it is to use certain therapeutic regimens to condition the tumor microenvironment for optimal response to immunotherapy. To this end, low dose immunogenic chemotherapies, epigenetic modulators and inhibitors of cell cycle progression are potential candidates for rendering tumors highly responsive to immunotherapy. Next generation immunotherapeutics are therefore predicted to be highly effective against cancer, when they are used following appropriate immune modulatory compounds or targeted delivery of tumor cell cycle inhibitors using nanotechnology.
Collapse
Affiliation(s)
- Zachary Benson
- Department of Internal Medicine, Virginia Commonwealth University School of Medicine, USA
| | - Saeed H Manjili
- Department of Biomedical Engineering, Virginia Commonwealth University School of Engineering, USA
| | - Mehran Habibi
- Department of Surgery, The Johns Hopkins School of Medicine, USA
| | - Georgi Guruli
- Division of Urology, Department of Surgery, Virginia Commonwealth University School of Medicine, USA; Massey Cancer Center, USA
| | - Amir A Toor
- Massey Cancer Center, USA; Bone Marrow Transplant Program, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, USA
| | - Kyle K Payne
- Translational Tumor Immunology Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Masoud H Manjili
- Massey Cancer Center, USA; Department of Microbiology & Immunology, Virginia Commonwealth University School of Medicine, USA.
| |
Collapse
|
81
|
Chesney JA, Mitchell RA, Yaddanapudi K. Myeloid-derived suppressor cells-a new therapeutic target to overcome resistance to cancer immunotherapy. J Leukoc Biol 2017; 102:727-740. [PMID: 28546500 DOI: 10.1189/jlb.5vmr1116-458rrr] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 04/18/2017] [Accepted: 04/20/2017] [Indexed: 12/12/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature myeloid cells that accumulate during pathologic conditions, such as cancer. Patients diagnosed with advanced metastatic cancers have an average survival of 12-24 mo, a survival time that hasn't changed significantly in the past 30 yr. Despite some encouraging improvements in response rates and overall survival in patients receiving immunotherapies, such as immune checkpoint inhibitors, most patients will ultimately progress. MDSCs contribute to immunotherapeutic resistance by actively inhibiting antitumor T cell proliferation and cytotoxic activity as well as by promoting expansion of protumorigenic T regulatory cells, thereby, dampening the host immune responses against the tumor. In addition, MDSCs promote angiogenesis, tumor invasion, and metastasis. Thus, MDSCs are potential therapeutic targets in cases of multiple cancers. This review focuses on the phenotypic and functional characteristics of MDSCs and provides an overview of the mono- and combinatorial-therapeutic strategies that target MDSCs with an objective of enhancing the efficacy of cancer immunotherapies.
Collapse
Affiliation(s)
- Jason A Chesney
- Molecular Targets Program, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA; .,Department of Medicine, University of Louisville, Louisville, Kentucky, USA; and
| | - Robert A Mitchell
- Molecular Targets Program, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA; .,Department of Medicine, University of Louisville, Louisville, Kentucky, USA; and.,Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, USA
| | - Kavitha Yaddanapudi
- Molecular Targets Program, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA; .,Department of Medicine, University of Louisville, Louisville, Kentucky, USA; and.,Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
82
|
Masuda J, Takayama E, Strober W, Satoh A, Morimoto Y, Honjo Y, Ichinohe T, Tokuno SI, Ishizuka T, Nakata T, Mizutani A, Umemura N, Kitani A, Fuss IJ, Shigehiro T, Kawaki H, Mizuno-Kamiya M, Kondoh N, Seno M. Tumor growth limited to subcutaneous site vs tumor growth in pulmonary site exhibit differential effects on systemic immunities. Oncol Rep 2017; 38:449-455. [PMID: 28535011 DOI: 10.3892/or.2017.5646] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 05/08/2017] [Indexed: 12/23/2022] Open
Abstract
To evaluate systemic immunity associated with tumor growth limited to a subcutaneous site versus growth proceeding at multiple tumor sites, we established syngeneic mouse subcutaneous and pulmonary tumor models by local subcutaneous and intravenous injection of colon carcinoma CT26 cells. We found that splenic myeloid-derived suppressor cell (MDSC) levels were significantly increased in the subcutaneous tumor model but not in the pulmonary tumor model. Furthermore, both CD4+ and CD8+ T cells as well as CD4+ Foxp3+ T cells were significantly decreased in the subcutaneous tumor model and were largely unchanged in the pulmonary tumor model. In addition, the subcutaneous model, but not the pulmonary model, displayed a Th1 polarization bias. This bias was characterized by decreased IL-4, IL-9, and IL-10 production, whereas the pulmonary model displayed increased production of IL-10. These results suggest that the mode of tumor development has differential effects on systemic immunity that may, in turn, influence approaches to treatment of cancer patients.
Collapse
Affiliation(s)
- Junko Masuda
- Division of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama, Japan
| | - Eiji Takayama
- Department of Oral Biochemistry, Asahi University School of Dentistry, Gifu, Japan
| | - Warren Strober
- Mucosal Immunity Section, Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ayano Satoh
- Division of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama, Japan
| | - Yuji Morimoto
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Saitama, Japan
| | - Yasuko Honjo
- Department of Hematology and Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Tatsuo Ichinohe
- Department of Hematology and Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Shin-Ichi Tokuno
- Verbal Analysis of Pathophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Toshiaki Ishizuka
- Department of Pharmacology, National Defense Medical College, Saitama, Japan
| | - Takahiro Nakata
- Department of Molecular and Cellular Anatomy, Faculty of Health Promotional Science, Tokoha University, Hamamatsu, Japan
| | - Akifumi Mizutani
- Division of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama, Japan
| | - Naoki Umemura
- Department of Oral Biochemistry, Asahi University School of Dentistry, Gifu, Japan
| | - Atsushi Kitani
- Mucosal Immunity Section, Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ivan J Fuss
- Mucosal Immunity Section, Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Tsukasa Shigehiro
- Division of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama, Japan
| | - Harumi Kawaki
- Department of Oral Biochemistry, Asahi University School of Dentistry, Gifu, Japan
| | - Masako Mizuno-Kamiya
- Department of Oral Biochemistry, Asahi University School of Dentistry, Gifu, Japan
| | - Nobuo Kondoh
- Department of Oral Biochemistry, Asahi University School of Dentistry, Gifu, Japan
| | - Masaharu Seno
- Division of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama, Japan
| |
Collapse
|
83
|
Expansion of CD11b+Ly6Ghigh and CD11b+CD49d+ myeloid cells with suppressive potential in mice with chronic inflammation and light-at-night-induced circadian disruption. Inflamm Res 2017; 66:711-724. [DOI: 10.1007/s00011-017-1052-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 03/24/2017] [Accepted: 04/22/2017] [Indexed: 12/20/2022] Open
|
84
|
Poh AR, Love CG, Masson F, Preaudet A, Tsui C, Whitehead L, Monard S, Khakham Y, Burstroem L, Lessene G, Sieber O, Lowell C, Putoczki TL, O'Donoghue RJJ, Ernst M. Inhibition of Hematopoietic Cell Kinase Activity Suppresses Myeloid Cell-Mediated Colon Cancer Progression. Cancer Cell 2017; 31:563-575.e5. [PMID: 28399411 PMCID: PMC5479329 DOI: 10.1016/j.ccell.2017.03.006] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 01/08/2017] [Accepted: 03/16/2017] [Indexed: 12/17/2022]
Abstract
Aberrant activation of the SRC family kinase hematopoietic cell kinase (HCK) triggers hematological malignancies as a tumor cell-intrinsic oncogene. Here we find that high HCK levels correlate with reduced survival of colorectal cancer patients. Likewise, increased Hck activity in mice promotes the growth of endogenous colonic malignancies and of human colorectal cancer cell xenografts. Furthermore, tumor-associated macrophages of the corresponding tumors show a pronounced alternatively activated endotype, which occurs independently of mature lymphocytes or of Stat6-dependent Th2 cytokine signaling. Accordingly, pharmacological inhibition or genetic reduction of Hck activity suppresses alternative activation of tumor-associated macrophages and the growth of colon cancer xenografts. Thus, Hck may serve as a promising therapeutic target for solid malignancies.
Collapse
Affiliation(s)
- Ashleigh R Poh
- Olivia Newton-John Cancer Research Institute, La Trobe University School of Cancer Medicine, Heidelberg, VIC 3084, Australia; The Walter and Eliza Hall Institute of Medical Research, Department of Medical Biology, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Christopher G Love
- The Walter and Eliza Hall Institute of Medical Research, Department of Medical Biology, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Frederick Masson
- Olivia Newton-John Cancer Research Institute, La Trobe University School of Cancer Medicine, Heidelberg, VIC 3084, Australia
| | - Adele Preaudet
- The Walter and Eliza Hall Institute of Medical Research, Department of Medical Biology, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Cary Tsui
- The Walter and Eliza Hall Institute of Medical Research, Department of Medical Biology, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Lachlan Whitehead
- The Walter and Eliza Hall Institute of Medical Research, Department of Medical Biology, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Simon Monard
- The Walter and Eliza Hall Institute of Medical Research, Department of Medical Biology, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Yelena Khakham
- The Walter and Eliza Hall Institute of Medical Research, Department of Medical Biology, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Lotta Burstroem
- The Walter and Eliza Hall Institute of Medical Research, Department of Medical Biology, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Guillaume Lessene
- The Walter and Eliza Hall Institute of Medical Research, Department of Medical Biology, University of Melbourne, Melbourne, VIC 3052, Australia; Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Oliver Sieber
- The Walter and Eliza Hall Institute of Medical Research, Department of Medical Biology, University of Melbourne, Melbourne, VIC 3052, Australia; Department of Colorectal Surgery, Royal Melbourne Hospital, Melbourne, VIC 3050, Australia; School of Biomedical Sciences, Monash University, Clayton, VIC 3800, Australia
| | - Clifford Lowell
- Department of Pathology and Laboratory Medicine, University of California, San Francisco, CA 94143, USA
| | - Tracy L Putoczki
- Olivia Newton-John Cancer Research Institute, La Trobe University School of Cancer Medicine, Heidelberg, VIC 3084, Australia; The Walter and Eliza Hall Institute of Medical Research, Department of Medical Biology, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Robert J J O'Donoghue
- Olivia Newton-John Cancer Research Institute, La Trobe University School of Cancer Medicine, Heidelberg, VIC 3084, Australia; The Walter and Eliza Hall Institute of Medical Research, Department of Medical Biology, University of Melbourne, Melbourne, VIC 3052, Australia.
| | - Matthias Ernst
- Olivia Newton-John Cancer Research Institute, La Trobe University School of Cancer Medicine, Heidelberg, VIC 3084, Australia; The Walter and Eliza Hall Institute of Medical Research, Department of Medical Biology, University of Melbourne, Melbourne, VIC 3052, Australia.
| |
Collapse
|
85
|
Terabe M, Robertson FC, Clark K, De Ravin E, Bloom A, Venzon DJ, Kato S, Mirza A, Berzofsky JA. Blockade of only TGF-β 1 and 2 is sufficient to enhance the efficacy of vaccine and PD-1 checkpoint blockade immunotherapy. Oncoimmunology 2017. [PMID: 28638730 DOI: 10.1080/2162402x.2017.1308616] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Checkpoint inhibition has established immunotherapy as a major modality of cancer treatment. However, the success of cancer immunotherapy is still limited as immune regulation of tumor immunity is very complicated and mechanisms involved may also differ among cancer types. Beside checkpoints, other good candidates for immunotherapy are immunosuppressive cytokines. TGF-β is a very potent immunosuppressive cytokine involved in suppression of tumor immunity and also necessary for the function of some regulatory cells. TGF-β has three isoforms, TGF-β 1, 2 and 3. It has been demonstrated in multiple mouse tumor models that inhibition of all three isoforms of TGF-β facilitates natural tumor immunosurveillance and tumor vaccine efficacy. However, individual isoforms of TGF-β are not well studied yet. Here, by using monoclonal antibodies (mAbs) specific for TGF-β isoforms, we asked whether it is necessary to inhibit TGF-β3 to enhance tumor immunity. We found that blockade of TGF-β1 and 2 and of all isoforms provided similar effects on tumor natural immunosurveillance and therapeutic vaccine-induced tumor immunity. The protection was CD8+ T cell-dependent. Blockade of TGF-β increased vaccine-induced Th1-type response measured by IFNγ production or T-bet expression in both tumor draining lymph nodes and tumors, although it did not increase tumor antigen-specific CD8+ T cell numbers. Therefore, protection correlated with qualitative rather than quantitative changes in T cells. Furthermore, when combined with PD-1 blockade, blockade of TGF-β1 and 2 further increased vaccine efficacy. In conclusion, blocking TGF-β1 and 2 is sufficient to enhance tumor immunity, and it can be further enhanced with PD-1 blockade.
Collapse
Affiliation(s)
- Masaki Terabe
- Vaccine Branch and Data Management Section, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Faith C Robertson
- Vaccine Branch and Data Management Section, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Katharine Clark
- Vaccine Branch and Data Management Section, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Emma De Ravin
- Vaccine Branch and Data Management Section, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Anja Bloom
- Vaccine Branch and Data Management Section, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - David J Venzon
- Data Management Section, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Shingo Kato
- Vaccine Branch and Data Management Section, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | - Jay A Berzofsky
- Vaccine Branch and Data Management Section, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
86
|
Brown JM, Recht L, Strober S. The Promise of Targeting Macrophages in Cancer Therapy. Clin Cancer Res 2017; 23:3241-3250. [PMID: 28341752 DOI: 10.1158/1078-0432.ccr-16-3122] [Citation(s) in RCA: 248] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 01/17/2017] [Accepted: 03/17/2017] [Indexed: 12/14/2022]
Abstract
Cancer therapy has developed around the concept of killing, or stopping the growth of, the cancer cells. Molecularly targeted therapy is the modern expression of this paradigm. Increasingly, however, the realization that the cancer has co-opted the normal cells of the stroma for its own survival has led to the concept that the tumor microenvironment (TME) could be targeted for effective therapy. In this review, we outline the importance of tumor-associated macrophages (TAM), a major component of the TME, in the response of tumors to cancer therapy. We discuss the normal role of macrophages in wound healing, the major phenotypes of TAMs, and their role in blunting the efficacy of cancer treatment by radiation and anticancer drugs, both by promoting tumor angiogenesis and by suppressing antitumor immunity. Finally, we review the many preclinical studies that have shown that the response of tumors to irradiation and anticancer drugs can be improved, sometimes markedly so, by depleting TAMs from tumors or by suppressing their polarization from an M1 to an M2 phenotype. The data clearly support the validity of clinical testing of combining targeting TAMs with conventional therapy. Clin Cancer Res; 23(13); 3241-50. ©2017 AACR.
Collapse
Affiliation(s)
- J Martin Brown
- Department of Radiation Oncology, Stanford University, Stanford, California.
| | - Lawrence Recht
- Department of Neurology, Stanford University, Stanford, California
| | - Samuel Strober
- Department of Medicine, Stanford University, Stanford, California
| |
Collapse
|
87
|
Musolino C, Allegra A, Pioggia G, Gangemi S. Immature myeloid-derived suppressor cells: A bridge between inflammation and cancer (Review). Oncol Rep 2016; 37:671-683. [PMID: 27922687 DOI: 10.3892/or.2016.5291] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 04/29/2015] [Indexed: 11/06/2022] Open
Abstract
Chronic inflammation is considered to be one of the hallmarks of tumor initiation and progression. Changes occurring in the microenvironment of progressing tumors resemble the process of chronic inflammation, which begins with ischemia followed by interstitial and cellular edema, appearance of immune cells, growth of blood vessels and tissue repair, and development of inflammatory infiltrates. Moreover, long‑term production and accumulation of inflammatory factors lead to local and systemic immunosuppression associated with cancer progression. Of the several mechanisms described to explain this anergy, the accumulation of myeloid cells in the tumor, spleen, and peripheral blood of cancer patients has gained considerable interest. A population of suppressive CD11b+Gr-1+ cells has in fact been designated as myeloid-derived suppressor cells (MDSCs). MDSCs are a unique category of the myeloid lineage, and they induce the prevention of the development of cytotoxic T lymphocytes (CTLs) in vitro, and the induction of antigen-specific CD8+ T-cell tolerance in vivo. Therapeutic approaches directed toward the manipulation of the MDSC population and their function may improve chemoimmune-enhancing therapy for advanced malignancies.
Collapse
Affiliation(s)
- Caterina Musolino
- Division of Hematology, Department of General Surgery, Pathological Anatomy and Oncology, University of Messina, Messina, Italy
| | - Alessandro Allegra
- Division of Hematology, Department of General Surgery, Pathological Anatomy and Oncology, University of Messina, Messina, Italy
| | - Govanni Pioggia
- Institute of Clinical Physiology, IFN CNR, Messina Unit, Messina, Italy
| | - Sebastiano Gangemi
- School and Division of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University Hospital 'G. Martino', Messina, Italy
| |
Collapse
|
88
|
The Crosstalk between Myeloid Derived Suppressor Cells and Immune Cells: To Establish Immune Tolerance in Transplantation. J Immunol Res 2016; 2016:4986797. [PMID: 27868073 PMCID: PMC5102737 DOI: 10.1155/2016/4986797] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 09/25/2016] [Indexed: 12/16/2022] Open
Abstract
Myeloid derived suppressor cells (MDSCs) are a heterogeneous population of myeloid precursor and progenitor cells and endowed with a robust immunosuppressive activity in multiple pathophysiological conditions. Recent studies have uncovered the crosstalk between MDSCs and immune cells (i.e., natural killer cells, dendritic cells, macrophages, natural killer T cells, and regulatory T cells) and its role in the establishment and maintenance of immune tolerant microenvironment in transplantation. Considering their strong immunosuppressive capability, MDSCs could become a prospective clinical regimen during transplantation tolerance induction, resulting in long-term graft survival with decreased or without immunosuppressive drugs. The review summarized recent research advances in this field and looked ahead at the research directions in the future.
Collapse
|
89
|
Korbelik M, Banáth J, Zhang W. Mreg Activity in Tumor Response to Photodynamic Therapy and Photodynamic Therapy-Generated Cancer Vaccines. Cancers (Basel) 2016; 8:cancers8100094. [PMID: 27754452 PMCID: PMC5082384 DOI: 10.3390/cancers8100094] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Revised: 10/10/2016] [Accepted: 10/11/2016] [Indexed: 01/20/2023] Open
Abstract
Myeloid regulatory cells (Mregs) are, together with regulatory T cells (Tregs), a dominant effector population responsible for restriction of the duration and strength of antitumor immune response. Photodynamic therapy (PDT) and cancer vaccines generated by PDT are modalities whose effectiveness in tumor destruction is closely dependent on the associated antitumor immune response. The present study investigated whether the immunodepletion of granulocytic Mregs in host mice by anti-GR1 antibody would improve the response of tumors to PDT or PDT vaccines in these animals. Anti-GR1 administration immediately after Temoporfin-PDT of mouse SCCVII tumors abrogated curative effect of PDT. The opposite effect, increasing PDT-mediated tumor cure-rates was attained by delaying anti-GR1 treatment to 1 h post PDT. With PDT vaccines, multiple anti-GR1 administrations (days 0, 4, and 8 post vaccination) improved the therapy response with SCCVII tumors. The results with PDT suggest that neutrophils (boosting antitumor effect of this therapy) that are engaged immediately after photodynamic light treatment are within one hour replaced with a different myeloid population, presumably Mregs that hampers the therapy-mediated antitumor effect. Anti-GR1 antibody, when used with optimal timing, can improve the efficacy of both PDT of tumors in situ and PDT-generated cancer vaccines.
Collapse
Affiliation(s)
- Mladen Korbelik
- British Columbia Cancer Agency, Vancouver, BC V5Z 4E6, Canada.
| | - Judith Banáth
- British Columbia Cancer Agency, Vancouver, BC V5Z 4E6, Canada.
| | - Wei Zhang
- British Columbia Cancer Agency, Vancouver, BC V5Z 4E6, Canada.
| |
Collapse
|
90
|
Okła K, Wertel I, Polak G, Surówka J, Wawruszak A, Kotarski J. Tumor-Associated Macrophages and Myeloid-Derived Suppressor Cells as Immunosuppressive Mechanism in Ovarian Cancer Patients: Progress and Challenges. Int Rev Immunol 2016; 35:372-385. [PMID: 27644763 DOI: 10.1080/08830185.2016.1206097] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cancers are complex masses of malignant cells and nonmalignant cells that create the tumor microenvironment (TME). Non-transformed cells of the TME such as tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs) have been observed in the TME of ovarian cancer (OC) patients. Although these subsets may contribute to each step of carcinogenesis and are commonly associated with poor prognosis, still little is known about creation of the protumor microenvironment in OC. In this review, we focused on the nature and prognostic significance of TAMs and MDSCs in OC patients. Moreover, we discuss the main problems and challenges that must be overcome by researchers and clinicians to enrich our knowledge about the immunosuppressive microenvironment of cancers.
Collapse
Affiliation(s)
- Karolina Okła
- a Department of Oncological Gynaecology and Gynaecology , Medical University , Lublin , Poland
| | - Iwona Wertel
- a Department of Oncological Gynaecology and Gynaecology , Medical University , Lublin , Poland
| | - Grzegorz Polak
- a Department of Oncological Gynaecology and Gynaecology , Medical University , Lublin , Poland
| | - Justyna Surówka
- a Department of Oncological Gynaecology and Gynaecology , Medical University , Lublin , Poland
| | - Anna Wawruszak
- b Department of Biochemistry and Molecular Biology , Medical University , Lublin , Poland
| | - Jan Kotarski
- a Department of Oncological Gynaecology and Gynaecology , Medical University , Lublin , Poland
| |
Collapse
|
91
|
Singel KL, Segal BH. Neutrophils in the tumor microenvironment: trying to heal the wound that cannot heal. Immunol Rev 2016; 273:329-43. [PMID: 27558344 PMCID: PMC5477672 DOI: 10.1111/imr.12459] [Citation(s) in RCA: 134] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Neutrophils are the first responders to infection and injury and are critical for antimicrobial host defense. Through the generation of reactive oxidants, activation of granular constituents and neutrophil extracellular traps, neutrophils target microbes and prevent their dissemination. While these pathways are beneficial in the context of trauma and infection, their off-target effects in the context of tumor are variable. Tumor-derived factors have been shown to reprogram the marrow, skewing toward the expansion of myelopoiesis. This can result in stimulation of both neutrophilic leukocytosis and the release of immature granulocytic populations that accumulate in circulation and in the tumor microenvironment. While activated neutrophils have been shown to kill tumor cells, there is growing evidence for neutrophil activation driving tumor progression and metastasis through a number of pathways, including stimulation of thrombosis and angiogenesis, stromal remodeling, and impairment of T cell-dependent anti-tumor immunity. There is also growing appreciation of neutrophil heterogeneity in cancer, with distinct neutrophil populations promoting cancer control or progression. In addition to the effects of tumor on neutrophil responses, anti-neoplastic treatment, including surgery, chemotherapy, and growth factors, can influence neutrophil responses. Future directions for research are expected to result in more mechanistic knowledge of neutrophil biology in the tumor microenvironment that may be exploited as prognostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Kelly L. Singel
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Brahm H. Segal
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY, USA
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
- Department of Medicine, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY, USA
| |
Collapse
|
92
|
Dysregulation of TGFβ1 Activity in Cancer and Its Influence on the Quality of Anti-Tumor Immunity. J Clin Med 2016; 5:jcm5090076. [PMID: 27589814 PMCID: PMC5039479 DOI: 10.3390/jcm5090076] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 08/26/2016] [Accepted: 08/29/2016] [Indexed: 01/01/2023] Open
Abstract
TGFβ1 is a pleiotropic cytokine that exhibits a variety of physiologic and immune regulatory functions. Although its influence on multiple cell types is critical for the regulation of numerous biologic processes in the host, dysregulation of both TGFβ1 expression and activity is frequently observed in cancer and contributes to various aspects of cancer progression. This review focuses on TGFβ1’s contribution to tumor immune suppression and escape, with emphasis on the influence of this regulatory cytokine on the differentiation and function of dendritic cells and T cells. Clinical trials targeting TGFβ1 in cancer patients are also reviewed, and strategies for future therapeutic interventions that build on our current understanding of immune regulation by TGFβ1 are discussed.
Collapse
|
93
|
Bagchi S, Li S, Wang CR. CD1b-autoreactive T cells recognize phospholipid antigens and contribute to antitumor immunity against a CD1b + T cell lymphoma. Oncoimmunology 2016; 5:e1213932. [PMID: 27757307 DOI: 10.1080/2162402x.2016.1213932] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 07/08/2016] [Accepted: 07/11/2016] [Indexed: 12/21/2022] Open
Abstract
Adoptive immunotherapy for cancer treatment is an emerging field of study. Till now, several tumor-derived, peptide-specific T cell responses have been harnessed for treating cancers. However, the contribution of lipid-specific T cells in tumor immunity has been understudied. CD1 molecules, which present self- and foreign lipid antigens to T cells, are divided into group 1 (CD1a, CD1b, and CD1c) and group 2 (CD1d). Although the role of CD1d-restricted natural killer T cells (NKT) in several tumor models has been well established, the contribution of group 1 CD1-restricted T cells in tumor immunity remains obscure due to the lack of group 1 CD1 expression in mice. In this study, we used a double transgenic mouse model expressing human group 1 CD1 molecules (hCD1Tg) and a CD1b-restricted, self-lipid reactive T cell receptor (HJ1Tg) to study the potential role of group 1 CD1-restricted autoreactive T cells in antitumor response. We found that HJ1 T cells recognized phospholipids and responded more potently to lipid extracted from tumor cells than the equivalent amount of lipids extracted from normal cells. Additionally, the autoreactivity of HJ1 T cells was enhanced upon treatment with various intracellular toll-like receptor (TLR) agonists, including CpG oligodeoxynucleotides (ODN), R848, and poly (I:C). Interestingly, the adoptive transfer of HJ1 T cells conferred protection against the CD1b-transfected murine T cell lymphoma (RMA-S/CD1b) and CpG ODN enhanced the antitumor effect. Thus, this study, for the first time, demonstrates the antitumor potential of CD1b-autoreactive T cells and their potential use in adoptive immunotherapy.
Collapse
Affiliation(s)
- Sreya Bagchi
- Department of Microbiology and Immunology, Northwestern University , Chicago, IL, USA
| | - Sha Li
- Department of Microbiology and Immunology, Northwestern University, Chicago, IL, USA; Department of Microbiology, Cornell University, Ithaca, NY, USA
| | - Chyung-Ru Wang
- Department of Microbiology and Immunology, Northwestern University , Chicago, IL, USA
| |
Collapse
|
94
|
Pyzer AR, Cole L, Rosenblatt J, Avigan DE. Myeloid-derived suppressor cells as effectors of immune suppression in cancer. Int J Cancer 2016; 139:1915-26. [PMID: 27299510 DOI: 10.1002/ijc.30232] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 06/07/2016] [Accepted: 06/08/2016] [Indexed: 12/11/2022]
Abstract
The tumor microenvironment consists of an immunosuppressive niche created by the complex interactions between cancer cells and surrounding stromal cells. A critical component of this environment are myeloid-derived suppressor cells (MDSCs), a heterogeneous group of immature myeloid cells arrested at different stages of differentiation and expanded in response to a variety of tumor factors. MDSCs exert diverse effects in modulating the interactions between immune effector cells and the malignant cells. An increased presence of MDSCs is associated with tumor progression, poorer outcomes, and decreased effectiveness of immunotherapeutic strategies. In this article, we will review our current understanding of the mechanisms that underlie MDSC expansion and their immune-suppressive function. Finally, we review the preclinical studies and clinical trials that have attempted to target MDSCs, in order to improve responses to cancer therapies.
Collapse
Affiliation(s)
- Athalia Rachel Pyzer
- Bone Marrow Transplant, Beth Israel Deaconess Medical Center, Center for Life Sciences, CLS724, Boston, MA
| | - Leandra Cole
- Bone Marrow Transplant, Beth Israel Deaconess Medical Center, Center for Life Sciences, CLS724, Boston, MA
| | - Jacalyn Rosenblatt
- Bone Marrow Transplant, Beth Israel Deaconess Medical Center, Center for Life Sciences, CLS724, Boston, MA
| | - David E Avigan
- Bone Marrow Transplant, Beth Israel Deaconess Medical Center, Center for Life Sciences, CLS724, Boston, MA
| |
Collapse
|
95
|
Delitto D, Wallet SM, Hughes SJ. Targeting tumor tolerance: A new hope for pancreatic cancer therapy? Pharmacol Ther 2016; 166:9-29. [PMID: 27343757 DOI: 10.1016/j.pharmthera.2016.06.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Accepted: 06/09/2016] [Indexed: 01/18/2023]
Abstract
With a 5-year survival rate of just 8%, pancreatic cancer (PC) is projected to be the second leading cause of cancer deaths by 2030. Most PC patients are not eligible for surgery with curative intent upon diagnosis, emphasizing a need for more effective therapies. However, PC is notoriously resistant to chemoradiation regimens. As an alternative, immune modulating strategies have recently achieved success in melanoma, prompting their application to other solid tumors. For such therapeutic approaches to succeed, a state of immunologic tolerance must be reversed in the tumor microenvironment and that has been especially challenging in PC. Nonetheless, knowledge of the PC immune microenvironment has advanced considerably over the past decade, yielding new insights and perspectives to guide multimodal therapies. In this review, we catalog the historical groundwork and discuss the evolution of the cancer immunology field to its present state with a specific focus on PC. Strategies currently employing immune modulation in PC are reviewed, specifically highlighting 66 clinical trials across the United States and Europe.
Collapse
Affiliation(s)
- Daniel Delitto
- Department of Surgery, University of Florida, Gainesville, FL, USA
| | - Shannon M Wallet
- Department of Oral Biology, University of Florida, Gainesville, FL, USA
| | - Steven J Hughes
- Department of Surgery, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
96
|
Parchment RE, Voth AR, Doroshow JH, Berzofsky JA. Immuno-pharmacodynamics for evaluating mechanism of action and developing immunotherapy combinations. Semin Oncol 2016; 43:501-13. [PMID: 27663482 DOI: 10.1053/j.seminoncol.2016.06.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Immunotherapy has become a major modality of cancer treatment, with multiple new classes of immunotherapeutics recently entering the clinic and obtaining market approval from regulatory agencies. While the promise of these therapies is great, so is the number of possible combinations not only with each other but also with small molecule therapeutics. Furthermore, the observation of unusual dose-response relationships suggests a critical dependency of drug effectiveness on the dosage regimen (dose and schedule). Clinical pharmacodynamic (PD) biomarkers will be useful endpoints for confirming drug mechanism of action, evaluating combination therapies for synergy or antagonism, and identifying optimal dosage regimens. In contrast to conventional PD in which drug action occurs entirely within a single target cell (ie, is self-contained within the malignant cell), immunotherapy involves a complex mechanism of action with sequential steps that propagate through multiple cell types, both normal and malignant. Its intercellular pharmacology begins with molecular target engagement either on an immune effector cell or a malignant cell, followed by stimulatory biochemical and biological signals in immune effector cells, and then finally ends with activation of cell death mechanisms in malignant cells lying within a certain distance from the activated effector cells (immune cell-tumor cell proximity). Evaluating such "trans-cellular pharmacology," in which different steps of drug action are distributed across multiple cell types, requires novel microscopy and image analysis tools capable of quantifying PD-biomarker responses, mapping the responses onto the cellular geography of the tumor using phenotypic biomarkers to identify specific cell types, and finally analyzing the spatial relationships between biomarkers in the context of each cell's biological role. We have termed this form of nearest neighbor image analysis of drug action "proximity PD microscopy," to indicate the importance of the location of the PD-biomarker response within the cellular landscape of a tumor specimen. We discuss herein the major modes of immunotherapy, and lay out a blueprint for using PD assessment to optimize dosage regimens of single agents and guide development of combination immunotherapy regimens, using PD1/PD-L1 immune checkpoint inhibition as a case study.
Collapse
Affiliation(s)
- Ralph E Parchment
- Clinical Pharmacodynamics Program, Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc, Frederick National Laboratory for Cancer Research, Frederick, MD.
| | - Andrea Regier Voth
- Clinical Pharmacodynamics Program, Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - James H Doroshow
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | | |
Collapse
|
97
|
Poly (I:C) enhances the anti-tumor activity of canine parvovirus NS1 protein by inducing a potent anti-tumor immune response. Tumour Biol 2016; 37:12089-12102. [PMID: 27209409 DOI: 10.1007/s13277-016-5093-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 05/15/2016] [Indexed: 12/26/2022] Open
Abstract
The canine parvovirus NS1 (CPV2.NS1) protein selectively induces apoptosis in the malignant cells. However, for an effective in vivo tumor treatment strategy, an oncolytic agent also needs to induce a potent anti-tumor immune response. In the present study, we used poly (I:C), a TLR3 ligand, as an adjuvant along with CPV2.NS1 to find out if the combination can enhance the oncolytic activity by inducing a potent anti-tumor immune response. The 4T1 mammary carcinoma cells were used to induce mammary tumor in Balb/c mice. The results suggested that poly (I:C), when given along with CPV2.NS1, not only significantly reduced the tumor growth but also augmented the immune response against tumor antigen(s) as indicated by the increase in blood CD4+ and CD8+ counts and infiltration of immune cells in the tumor tissue. Further, blood serum analysis of the cytokines revealed that Th1 cytokines (IFN-γ and IL-2) were significantly upregulated in the treatment group indicating activation of cell-mediated immune response. The present study reports the efficacy of CPV2.NS1 along with poly (I:C) not only in inhibiting the mammary tumor growth but also in generating an active anti-tumor immune response without any visible toxicity. The results of our study may help in developing CPV2.NS1 and poly (I: C) combination as a cancer therapeutic regime to treat various malignancies.
Collapse
|
98
|
The spleen as an extramedullary source of inflammatory cells responding to acetaminophen-induced liver injury. Toxicol Appl Pharmacol 2016; 304:110-20. [PMID: 27163765 DOI: 10.1016/j.taap.2016.04.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 04/25/2016] [Accepted: 04/27/2016] [Indexed: 12/18/2022]
Abstract
Macrophages have been shown to play a role in acetaminophen (APAP)-induced hepatotoxicity, contributing to both pro- and anti-inflammatory processes. In these studies, we analyzed the role of the spleen as an extramedullary source of hepatic macrophages. APAP administration (300mg/kg, i.p.) to control mice resulted in an increase in CD11b(+) infiltrating Ly6G(+) granulocytic and Ly6G(-) monocytic cells in the spleen and the liver. The majority of the Ly6G(+) cells were also positive for the monocyte/macrophage activation marker, Ly6C, suggesting a myeloid derived suppressor cell (MDSC) phenotype. By comparison, Ly6G(-) cells consisted of 3 subpopulations expressing high, intermediate, and low levels of Ly6C. Splenectomy was associated with increases in mature (F4/80(+)) and immature (F4/80(-)) pro-inflammatory Ly6C(hi) macrophages and mature anti-inflammatory (Ly6C(lo)) macrophages in the liver after APAP; increases in MDSCs were also noted in the livers of splenectomized (SPX) mice after APAP. This was associated with increases in APAP-induced expression of chemokine receptors regulating pro-inflammatory (CCR2) and anti-inflammatory (CX3CR1) macrophage trafficking. In contrast, APAP-induced increases in pro-inflammatory galectin-3(+) macrophages were blunted in livers of SPX mice relative to control mice, along with hepatic expression of TNF-α, as well as the anti-inflammatory macrophage markers, FIZZ-1 and YM-1. These data demonstrate that multiple subpopulations of pro- and anti-inflammatory cells respond to APAP-induced injury, and that these cells originate from distinct hematopoietic reservoirs.
Collapse
|
99
|
Favreau M, Vanderkerken K, Elewaut D, Venken K, Menu E. Does an NKT-cell-based immunotherapeutic approach have a future in multiple myeloma? Oncotarget 2016; 7:23128-40. [PMID: 26895468 PMCID: PMC5029615 DOI: 10.18632/oncotarget.7440] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 02/05/2016] [Indexed: 12/21/2022] Open
Abstract
Natural killer T (NKT) cells constitute a unique subset of innate-like T lymphocytes which differ from conventional T cells by recognizing lipid antigens presented by the non-polymorphic major histocompatibility complex (MHC) I-like molecule CD1d. Despite being a relatively infrequent population of lymphocytes, NKT cells can respond rapidly upon activation with glycosphingolipids by production of cytokines which aim to polarize different axes of the immune system. Due to their dual effector capacities, NKT cells can play a vital role in cancer immunity, infection, inflammation and autoimmune diseases. It is believed that modulation of their activity towards immune activation can be a useful tool in anti-tumor immunotherapeutic strategies. Here we summarize the characteristics of NKT cells and discuss their involvement in immunosurveillance. Furthermore, an update is given about their role and the progress that has been made in the field of multiple myeloma (MM). Finally, some challenges are discussed that are currently hampering further progress.
Collapse
Affiliation(s)
- Mérédis Favreau
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Laboratory for Molecular Immunology and Inflammation, Department of Rheumatology, Faculty of Medicine and Health Sciences, VIB Inflammation Research Center and Ghent University, Ghent, Belgium
| | - Karin Vanderkerken
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Dirk Elewaut
- Laboratory for Molecular Immunology and Inflammation, Department of Rheumatology, Faculty of Medicine and Health Sciences, VIB Inflammation Research Center and Ghent University, Ghent, Belgium
| | - Koen Venken
- Laboratory for Molecular Immunology and Inflammation, Department of Rheumatology, Faculty of Medicine and Health Sciences, VIB Inflammation Research Center and Ghent University, Ghent, Belgium
| | - Eline Menu
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| |
Collapse
|
100
|
De Vlaeminck Y, González-Rascón A, Goyvaerts C, Breckpot K. Cancer-Associated Myeloid Regulatory Cells. Front Immunol 2016; 7:113. [PMID: 27065074 PMCID: PMC4810015 DOI: 10.3389/fimmu.2016.00113] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 03/14/2016] [Indexed: 12/25/2022] Open
Abstract
Myeloid cells are critically involved in the pathophysiology of cancers. In the tumor microenvironment (TME), they comprise tumor-associated macrophages (TAMs), neutrophils (TANs), dendritic cells, and myeloid-derived suppressor cells, which are further subdivided into a monocytic subset and a granulocytic subset. Some of these myeloid cells, in particular TAMs and TANs, are divided into type 1 or type 2 cells, according to the paradigm of T helper type 1 or type 2 cells. Type 1-activated cells are generally characterized as cells that aid tumor rejection, while all other myeloid cells are shown to favor tumor progression. Moreover, these cells are often at the basis of resistance to various therapies. Much research has been devoted to study the biology of myeloid cells. This endeavor has proven to be challenging, as the markers used to categorize myeloid cells in the TME are not restricted to particular subsets. Also from a functional and metabolic point of view, myeloid cells share many features. Finally, myeloid cells are endowed with a certain level of plasticity, which further complicates studying them outside their environment. In this article, we challenge the exclusive use of cell markers to unambiguously identify myeloid cell subsets in the TME. We further propose to divide myeloid cells into myeloid regulatory or stimulatory cells according to their pro- or antitumor function, because we contend that for therapeutic purposes it is not targeting the cell subsets but rather targeting their protumor traits; hence, myeloid regulatory cells will push antitumor immunotherapy to the next level.
Collapse
Affiliation(s)
- Yannick De Vlaeminck
- Laboratory of Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel , Brussels , Belgium
| | - Anna González-Rascón
- Laboratory of Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium; Centro de Investigación en Alimentación y Desarrollo, Hermosillo, Mexico
| | - Cleo Goyvaerts
- Laboratory of Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel , Brussels , Belgium
| | - Karine Breckpot
- Laboratory of Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel , Brussels , Belgium
| |
Collapse
|