1
|
Tan SN, Hao J, Ge J, Yang Y, Liu L, Huang J, Lin M, Zhao X, Wang G, Yang Z, Ni L, Dong C. Regulatory T cells converted from Th1 cells in tumors suppress cancer immunity via CD39. J Exp Med 2025; 222:e20240445. [PMID: 39907686 PMCID: PMC11797014 DOI: 10.1084/jem.20240445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 10/17/2024] [Accepted: 01/10/2025] [Indexed: 02/06/2025] Open
Abstract
Regulatory T (Treg) cells are known to impede antitumor immunity, yet the regulatory mechanisms and functional roles of these cells remain poorly understood. In this study, through the characterization of multiple cancer models, we identified a substantial presence of peripherally induced Treg cells in the tumor microenvironment (TME). Depletion of these cells triggered antitumor responses and provided potent therapeutic effects by increasing functional CD8+ T cells. Fate-mapping and transfer experiments revealed that IFN-γ-expressing T helper (Th) 1 cells differentiated into Treg cells in response to TGF-β signaling in tumors. Pseudotime trajectory analysis further revealed the terminal differentiation of Th1-like Treg cells from Th1 cells in the TME. Tumor-resident Treg cells highly expressed T-bet, which was essential for their functions in the TME. Additionally, CD39 was highly expressed by T-bet+ Treg cells in both mouse and human tumors, and was necessary for Treg cell-mediated suppression of CD8+ T cell responses. Our study elucidated the developmental pathway of intratumoral Treg cells and highlighted novel strategies for targeting them in cancer patients.
Collapse
Affiliation(s)
- Sang-Nee Tan
- School of Medicine, Westlake University, Hangzhou, China
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Jing Hao
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
- Shanghai Immune Therapy Institute, Shanghai Jiao Tong University School of Medicine-affiliated Renji Hospital, Shanghai, China
| | - Jing Ge
- Shanghai Immune Therapy Institute, Shanghai Jiao Tong University School of Medicine-affiliated Renji Hospital, Shanghai, China
| | - Yazheng Yang
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Liguo Liu
- Department of Hepatobiliary Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Jia Huang
- Department of Hepatobiliary Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Meng Lin
- School of Medicine, Westlake University, Hangzhou, China
| | - Xiaohong Zhao
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Genyu Wang
- School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Zhiying Yang
- Department of Hepatobiliary Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Ling Ni
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Chen Dong
- School of Medicine, Westlake University, Hangzhou, China
- Shanghai Immune Therapy Institute, Shanghai Jiao Tong University School of Medicine-affiliated Renji Hospital, Shanghai, China
| |
Collapse
|
2
|
Oka K, Yamakawa M, Kawamura Y, Kutsukake N, Miura K. The Naked Mole-Rat as a Model for Healthy Aging. Annu Rev Anim Biosci 2023; 11:207-226. [PMID: 36318672 DOI: 10.1146/annurev-animal-050322-074744] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Naked mole-rats (NMRs, Heterocephalus glaber) are the longest-lived rodents with a maximum life span exceeding 37 years. They exhibit a delayed aging phenotype and resistance to age-related functional decline/diseases. Specifically, they do not display increased mortality with age, maintain several physiological functions until nearly the end of their lifetime, and rarely develop cancer and Alzheimer's disease. NMRs live in a hypoxic environment in underground colonies in East Africa and are highly tolerant of hypoxia. These unique characteristics of NMRs have attracted considerable interest from zoological and biomedical researchers. This review summarizes previous studies of the ecology, hypoxia tolerance, longevity/delayed aging, and cancer resistance of NMRs and discusses possible mechanisms contributing to their healthy aging. In addition, we discuss current issues and future perspectives to fully elucidate the mechanisms underlying delayed aging and resistance to age-related diseases in NMRs.
Collapse
Affiliation(s)
- Kaori Oka
- Department of Aging and Longevity Research, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan; , ,
| | - Masanori Yamakawa
- Department of Evolutionary Studies of Biosystems, Sokendai (The Graduate University for Advanced Studies), Kanagawa, Japan; ,
| | - Yoshimi Kawamura
- Department of Aging and Longevity Research, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan; , ,
| | - Nobuyuki Kutsukake
- Department of Evolutionary Studies of Biosystems, Sokendai (The Graduate University for Advanced Studies), Kanagawa, Japan; , .,Research Center for Integrative Evolutionary Science, Sokendai (The Graduate University for Advanced Studies), Kanagawa, Japan
| | - Kyoko Miura
- Department of Aging and Longevity Research, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan; , , .,Center for Metabolic Regulation of Healthy Aging, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
3
|
Pereira Gonçalves J, Fwu Shing T, Augusto Fonseca Alves G, Eduardo Fonseca-Alves C. Immunology of Canine Melanoma. Vet Med Sci 2022. [DOI: 10.5772/intechopen.108430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Malignant melanoma is one of the most important tumors in dogs and is highly metastatic and aggressive disease. In recent years, molecular knowledge regarding canine melanoma has increased, and some chromosomal imbalances and tyrosine kinase pathways have been identified to be dysregulated. Mxoreover, canine melanoma is an immunogenic tumor that provides opportunities to administer immunotherapy to the patient. Podoplanin and chondroitin sulfate proteoglycan-4 (CSPG4) are markers against which monoclonal antibodies have been developed and tested in dogs in vivo with promising results. Owing to the importance of canine melanoma in the veterinary oncology field, this chapter reviews the most important aspects related to immunological involvement in the prognosis and treatment of canine melanoma.
Collapse
|
4
|
Connolly KA, Fitzgerald B, Damo M, Joshi NS. Novel Mouse Models for Cancer Immunology. ANNUAL REVIEW OF CANCER BIOLOGY 2022; 6:269-291. [PMID: 36875867 PMCID: PMC9979244 DOI: 10.1146/annurev-cancerbio-070620-105523] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mouse models for the study of cancer immunology provide excellent systems in which to test biological mechanisms of the immune response against cancer. Historically, these models have been designed to have different strengths based on the current major research questions at the time. As such, many mouse models of immunology used today were not originally developed to study questions currently plaguing the relatively new field of cancer immunology, but instead have been adapted for such purposes. In this review, we discuss various mouse model of cancer immunology in a historical context as a means to provide a fuller perspective of each model's strengths. From this outlook, we discuss the current state of the art and strategies for tackling future modeling challenges.
Collapse
Affiliation(s)
- Kelli A. Connolly
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Brittany Fitzgerald
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Martina Damo
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Nikhil S. Joshi
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06519, USA
| |
Collapse
|
5
|
Nave O. A mathematical model for treatment using chemo-immunotherapy. Heliyon 2022; 8:e09288. [PMID: 35520602 PMCID: PMC9065634 DOI: 10.1016/j.heliyon.2022.e09288] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 10/18/2021] [Accepted: 04/12/2022] [Indexed: 11/15/2022] Open
Abstract
In this study, we investigated a mathematical model for chemoimmunotherapy (a combination of chemotherapy and immunotherapy) for brain cancer. In most cases, the standard protocol for cancer treatment is fixed in terms of treatment time intervals and dosages. We offer a wide range of non-fixed protocols, which essentially vary in terms of time intervals and dosages (i.e., personalised medicine). The functions that describe this treatment are explicit and analytical. Hence, the parameters of the function can be easily changed and a new protocol can be obtained. We compared different protocols and obtained an optimal solution. In addition, we applied the singular perturbed vector field (SPVF) method to determine the hierarchy of the system of equations, which enabled us to identify the equilibrium points of the mathematical model and investigate their stability.
Collapse
Affiliation(s)
- Ophir Nave
- Department of Mathematics, Jerusalem College of Technology, Israel
| |
Collapse
|
6
|
Oka K, Fujioka S, Kawamura Y, Komohara Y, Chujo T, Sekiguchi K, Yamamura Y, Oiwa Y, Omamiuda-Ishikawa N, Komaki S, Sutoh Y, Sakurai S, Tomizawa K, Bono H, Shimizu A, Araki K, Yamamoto T, Yamada Y, Oshiumi H, Miura K. Resistance to chemical carcinogenesis induction via a dampened inflammatory response in naked mole-rats. Commun Biol 2022; 5:287. [PMID: 35354912 PMCID: PMC8967925 DOI: 10.1038/s42003-022-03241-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/09/2022] [Indexed: 12/13/2022] Open
Abstract
Naked mole-rats (NMRs) have a very low spontaneous carcinogenesis rate, which has prompted studies on the responsible mechanisms to provide clues for human cancer prevention. However, it remains unknown whether and how NMR tissues respond to experimental carcinogenesis induction. Here, we show that NMRs exhibit extraordinary resistance against potent chemical carcinogenesis induction through a dampened inflammatory response. Although carcinogenic insults damaged skin cells of both NMRs and mice, NMR skin showed markedly lower immune cell infiltration. NMRs harbour loss-of-function mutations in RIPK3 and MLKL genes, which are essential for necroptosis, a type of necrotic cell death that activates strong inflammation. In mice, disruption of Ripk3 reduced immune cell infiltration and delayed carcinogenesis. Therefore, necroptosis deficiency may serve as a cancer resistance mechanism via attenuating the inflammatory response in NMRs. Our study sheds light on the importance of a dampened inflammatory response as a non-cell-autonomous cancer resistance mechanism in NMRs. Naked mole rats are found to be resistant to cancer development through dampened inflammatory response due to genetically determined impaired necroptosis, with essential necroptosis genes RIPK3 and MLKL containing mutations causing premature stop codons.
Collapse
Affiliation(s)
- Kaori Oka
- Department of Aging and Longevity Research, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-0811, Japan.,Biomedical Animal Research Laboratory, Institute for Genetic Medicine, Hokkaido University, Sapporo, 060-0815, Japan
| | - Shusuke Fujioka
- Department of Aging and Longevity Research, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-0811, Japan.,Biomedical Animal Research Laboratory, Institute for Genetic Medicine, Hokkaido University, Sapporo, 060-0815, Japan
| | - Yoshimi Kawamura
- Department of Aging and Longevity Research, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-0811, Japan.,Biomedical Animal Research Laboratory, Institute for Genetic Medicine, Hokkaido University, Sapporo, 060-0815, Japan
| | - Yoshihiro Komohara
- Department of Cell Pathology, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Takeshi Chujo
- Department of Molecular Physiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Koki Sekiguchi
- Department of Aging and Longevity Research, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-0811, Japan
| | - Yuki Yamamura
- Department of Aging and Longevity Research, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-0811, Japan
| | - Yuki Oiwa
- Department of Aging and Longevity Research, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-0811, Japan.,Biomedical Animal Research Laboratory, Institute for Genetic Medicine, Hokkaido University, Sapporo, 060-0815, Japan
| | - Natsuko Omamiuda-Ishikawa
- Department of Aging and Longevity Research, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-0811, Japan
| | - Shohei Komaki
- Division of Biomedical Information Analysis, Iwate Tohoku Medical Megabank Organization, Disaster Reconstruction Center, Iwate Medical University, Iwate, 028-3694, Japan
| | - Yoichi Sutoh
- Division of Biomedical Information Analysis, Iwate Tohoku Medical Megabank Organization, Disaster Reconstruction Center, Iwate Medical University, Iwate, 028-3694, Japan
| | - Satoko Sakurai
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan
| | - Kazuhito Tomizawa
- Department of Molecular Physiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-8556, Japan.,Center for Metabolic Regulation of Healthy Aging, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Hidemasa Bono
- Program of Biomedical Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, 739-0046, Japan
| | - Atsushi Shimizu
- Division of Biomedical Information Analysis, Iwate Tohoku Medical Megabank Organization, Disaster Reconstruction Center, Iwate Medical University, Iwate, 028-3694, Japan.,Division of Biomedical Information Analysis, Institute for Biomedical Sciences, Iwate Medical University, Iwate, 028-3694, Japan
| | - Kimi Araki
- Center for Metabolic Regulation of Healthy Aging, Kumamoto University, Kumamoto, 860-8556, Japan.,Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, 860-0811, Japan
| | - Takuya Yamamoto
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan.,Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, 606-8501, Japan.,Medical-risk Avoidance based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto, 606-8507, Japan.,AMED-CREST, AMED, Tokyo, 100-0004, Japan
| | - Yasuhiro Yamada
- AMED-CREST, AMED, Tokyo, 100-0004, Japan.,Division of Stem Cell Pathology, Center for Experimental Medicine and Systems Biology, Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Hiroyuki Oshiumi
- Department of Immunology, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Kyoko Miura
- Department of Aging and Longevity Research, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-0811, Japan. .,Biomedical Animal Research Laboratory, Institute for Genetic Medicine, Hokkaido University, Sapporo, 060-0815, Japan. .,Center for Metabolic Regulation of Healthy Aging, Kumamoto University, Kumamoto, 860-8556, Japan.
| |
Collapse
|
7
|
Vienne M, Etiennot M, Escalière B, Galluso J, Spinelli L, Guia S, Fenis A, Vivier E, Kerdiles YM. Type 1 Innate Lymphoid Cells Limit the Antitumoral Immune Response. Front Immunol 2021; 12:768989. [PMID: 34868026 PMCID: PMC8637113 DOI: 10.3389/fimmu.2021.768989] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/26/2021] [Indexed: 12/11/2022] Open
Abstract
Natural killer (NK) cells are known to be able to kill established tumor cell lines, but important caveats remain regarding their roles in the detection and elimination of developing primary tumors. Using a genetic model of selective ILC1 and NK cell deficiency, we showed that these cells were dispensable for tumor immunosurveillance and immunoediting in the MCA-induced carcinogenesis model. However, we were able to generate primary cell lines derived from MCA-induced tumors with graded sensitivity to NK1.1+ cells (including NK cells and ILC1). This differential sensitivity was associated neither with a modulation of intratumoral NK cell frequency, nor the capacity of tumor cells to activate NK cells. Instead, ILC1 infiltration into the tumor was found to be a critical determinant of NK1.1+ cell-dependent tumor growth. Finally, bulk tumor RNAseq analysis identified a gene expression signature associated with tumor sensitivity to NK1.1+ cells. ILC1 therefore appear to play an active role in inhibiting the antitumoral immune response, prompting to evaluate the differential tumor infiltration of ILC1 and NK cells in patients to optimize the harnessing of immunity in cancer therapies.
Collapse
Affiliation(s)
- Margaux Vienne
- Aix-Marseille Univ, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre d'Immunologie de Marseille-Luminy (CIML), Marseille, France
| | - Marion Etiennot
- Aix-Marseille Univ, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre d'Immunologie de Marseille-Luminy (CIML), Marseille, France
| | - Bertrand Escalière
- Aix-Marseille Univ, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre d'Immunologie de Marseille-Luminy (CIML), Marseille, France
| | - Justine Galluso
- Aix-Marseille Univ, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre d'Immunologie de Marseille-Luminy (CIML), Marseille, France
| | - Lionel Spinelli
- Aix-Marseille Univ, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre d'Immunologie de Marseille-Luminy (CIML), Marseille, France
| | - Sophie Guia
- Aix-Marseille Univ, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre d'Immunologie de Marseille-Luminy (CIML), Marseille, France
| | | | - Eric Vivier
- Aix-Marseille Univ, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre d'Immunologie de Marseille-Luminy (CIML), Marseille, France.,Innate Pharma, Marseille, France.,APHM, Hôpital de la Timone, Marseille-Immunopôle, Marseille, France
| | - Yann M Kerdiles
- Aix-Marseille Univ, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre d'Immunologie de Marseille-Luminy (CIML), Marseille, France
| |
Collapse
|
8
|
Li M, Li X, Goldsmith JR, Shi S, Zhang L, Zamani A, Wan L, Sun H, Li T, Yu J, Etwebi Z, Bou-Dargham MJ, Chen YH. Decoupling tumor cell metastasis from growth by cellular pilot protein TNFAIP8. Oncogene 2021; 40:6456-6468. [PMID: 34608264 PMCID: PMC8604770 DOI: 10.1038/s41388-021-02035-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 08/28/2021] [Accepted: 09/22/2021] [Indexed: 02/08/2023]
Abstract
Cancer metastasis accounts for nearly 90% of all cancer deaths. Metastatic cancer progression requires both cancer cell migration to the site of the metastasis and subsequent proliferation after colonization. However, it has long been recognized that cancer cell migration and proliferation can be uncoupled; but the mechanism underlying this paradox is not well understood. Here we report that TNFAIP8 (tumor necrosis factor-α-induced protein 8), a "professional" transfer protein of phosphoinositide second messengers, promotes cancer cell migration or metastasis but inhibits its proliferation or cancer growth. TNFAIP8-deficient mice developed larger tumors, but TNFAIP8-deficient tumor cells completely lost their ability to migrate toward chemoattractants and were defective in colonizing lung tissues as compared to wild-type counterparts. Mechanistically, TNFAIP8 served as a cellular "pilot" of tumor cell migration by locally amplifying PI3K-AKT and Rac signals on the cell membrane facing chemoattractant; at the same time, TNFAIP8 also acted as a global inhibitor of tumor cell growth and proliferation by regulating Hippo signaling pathway. These findings help explain the migration-proliferation paradox of cancer cells that characterizes many cancers.
Collapse
Affiliation(s)
- Mingyue Li
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA,Corresponding authors: Dr. Youhai H. Chen, 713 Stellar-Chance Laboratories, 422 Curie Blvd. Philadelphia, PA 19104, 215-898-4671, ; Dr. Mingyue Li, 712 Stellar-Chance Laboratories, 422 Curie Blvd., Philadelphia, PA 19104, 215-898-7962,
| | - Xinyuan Li
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jason R. Goldsmith
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Songlin Shi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Li Zhang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ali Zamani
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lin Wan
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Honghong Sun
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ting Li
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jiyeon Yu
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zienab Etwebi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mayassa J. Bou-Dargham
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Youhai H. Chen
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA,Faculty of Pharmaceutical Sciences, CAS Shenzhen Institute of Advanced Technology, Shenzhen, China,Corresponding authors: Dr. Youhai H. Chen, 713 Stellar-Chance Laboratories, 422 Curie Blvd. Philadelphia, PA 19104, 215-898-4671, ; Dr. Mingyue Li, 712 Stellar-Chance Laboratories, 422 Curie Blvd., Philadelphia, PA 19104, 215-898-7962,
| |
Collapse
|
9
|
Lauder SN, Milutinovic S, Pires A, Smart K, Godkin A, Gallimore A. Using methylcholanthrene-induced fibrosarcomas to study tumor immunology. Methods Cell Biol 2020; 163:59-75. [PMID: 33785169 DOI: 10.1016/bs.mcb.2020.09.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Mouse models of cancer are essential in furthering our understanding both of the mechanisms that drive tumor development and the immune response that develops in parallel, and also in providing a platform for testing novel anti-cancer therapies. The majority of solid tumor models available rely on the injection of existing cancer cell lines into naïve hosts which, while providing quick and reproducible model systems, typically lack the development of a tumor microenvironment that recapitulates those seen in human cancers. Administration of the carcinogen 3-methylcholanthrene (MCA), allows tumors to develop in situ, forming a tumor microenvironment with an established stroma and vasculature. This article provides a detailed set of protocols for the administration of MCA into mice and the subsequent monitoring of tumors. Protocols are also provided for some of the routinely used downstream applications that can be used for MCA tumors.
Collapse
Affiliation(s)
- S N Lauder
- Division of Infection Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - S Milutinovic
- Division of Infection Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - A Pires
- Division of Infection Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - K Smart
- Division of Infection Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - A Godkin
- Division of Infection Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - A Gallimore
- Division of Infection Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom.
| |
Collapse
|
10
|
Devalaraja S, To TKJ, Folkert IW, Natesan R, Alam MZ, Li M, Tada Y, Budagyan K, Dang MT, Zhai L, Lobel GP, Ciotti GE, Eisinger-Mathason TSK, Asangani IA, Weber K, Simon MC, Haldar M. Tumor-Derived Retinoic Acid Regulates Intratumoral Monocyte Differentiation to Promote Immune Suppression. Cell 2020; 180:1098-1114.e16. [PMID: 32169218 PMCID: PMC7194250 DOI: 10.1016/j.cell.2020.02.042] [Citation(s) in RCA: 161] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 12/13/2019] [Accepted: 02/19/2020] [Indexed: 12/13/2022]
Abstract
The immunosuppressive tumor microenvironment (TME) is a major barrier to immunotherapy. Within solid tumors, why monocytes preferentially differentiate into immunosuppressive tumor-associated macrophages (TAMs) rather than immunostimulatory dendritic cells (DCs) remains unclear. Using multiple murine sarcoma models, we find that the TME induces tumor cells to produce retinoic acid (RA), which polarizes intratumoral monocyte differentiation toward TAMs and away from DCs via suppression of DC-promoting transcription factor Irf4. Genetic inhibition of RA production in tumor cells or pharmacologic inhibition of RA signaling within TME increases stimulatory monocyte-derived cells, enhances T cell-dependent anti-tumor immunity, and synergizes with immune checkpoint blockade. Furthermore, an RA-responsive gene signature in human monocytes correlates with an immunosuppressive TME in multiple human tumors. RA has been considered as an anti-cancer agent, whereas our work demonstrates its tumorigenic capability via myeloid-mediated immune suppression and provides proof of concept for targeting this pathway for tumor immunotherapy.
Collapse
Affiliation(s)
- Samir Devalaraja
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19014, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tsun Ki Jerrick To
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19014, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ian W Folkert
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ramakrishnan Natesan
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Md Zahidul Alam
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19014, USA
| | - Minghong Li
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19014, USA
| | - Yuma Tada
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19014, USA
| | - Konstantin Budagyan
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19104, USA
| | - Mai T Dang
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Li Zhai
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19014, USA
| | - Graham P Lobel
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19014, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gabrielle E Ciotti
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19014, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - T S Karin Eisinger-Mathason
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19014, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Irfan A Asangani
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kristy Weber
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - M Celeste Simon
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Malay Haldar
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19014, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
11
|
Henke N, Ferreirós N, Geisslinger G, Ding MG, Essler S, Fuhrmann DC, Geis T, Namgaladze D, Dehne N, Brüne B. Loss of HIF-1α in macrophages attenuates AhR/ARNT-mediated tumorigenesis in a PAH-driven tumor model. Oncotarget 2017; 7:25915-29. [PMID: 27015123 PMCID: PMC5041954 DOI: 10.18632/oncotarget.8297] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 03/11/2016] [Indexed: 01/04/2023] Open
Abstract
Activation of hypoxia-inducible factor (HIF) and macrophage infiltration of solid tumors independently promote tumor progression. As little is known how myeloid HIF affects tumor development, we injected the polycyclic aromatic hydrocarbon (PAH) and procarcinogen 3-methylcholanthrene (MCA; 100 μg/100 μl) subcutaneously into myeloid-specific Hif-1α and Hif-2α knockout mice (C57BL/6J) to induce fibrosarcomas (n = 16). Deletion of Hif-1α but not Hif-2α in macrophages diminished tumor outgrowth in the MCA-model. While analysis of the tumor initiation phase showed comparable inflammation after MCA-injection, metabolism of MCA was impaired in the absence of Hif-1α. An ex vivo macrophage/fibroblast coculture recapitulated reduced DNA damage after MCA-stimulation in fibroblasts of cocultures with Hif-1αLysM−/− macrophages compared to wild type macrophages. A loss of myeloid Hif-1α decreased RNA levels of arylhydrocarbon receptor (AhR)/arylhydrocarbon receptor nuclear translocator (ARNT) targets such as Cyp1a1 because of reduced Arnt but unchanged Ahr expression. Cocultures using Hif-1αLysM−/− macrophages stimulated with the carcinogen 7,12-dimethylbenz[a]anthracene (DMBA; 2 μg/ml) also attenuated a DNA damage response in fibroblasts, while the DNA damage-inducing metabolite DMBA-trans-3,4-dihydrodiol remained effective in the absence of Hif-1α. In chemical-induced carcinogenesis, HIF-1α in macrophages maintains ARNT expression to facilitate PAH-biotransformation. This implies a metabolic activation of PAHs in stromal cells, i.e. myeloid-derived cells, to be crucial for tumor initiation.
Collapse
Affiliation(s)
- Nina Henke
- Institute of Biochemistry I, Goethe-University Frankfurt, 60590 Frankfurt, Germany
| | - Nerea Ferreirós
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt, Goethe-University Frankfurt, 60590 Frankfurt, Germany
| | - Gerd Geisslinger
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt, Goethe-University Frankfurt, 60590 Frankfurt, Germany
| | - Martina G Ding
- Institute of Biochemistry I, Goethe-University Frankfurt, 60590 Frankfurt, Germany
| | - Silke Essler
- Institute of Biochemistry I, Goethe-University Frankfurt, 60590 Frankfurt, Germany
| | - Dominik C Fuhrmann
- Institute of Biochemistry I, Goethe-University Frankfurt, 60590 Frankfurt, Germany
| | - Theresa Geis
- Institute of Biochemistry I, Goethe-University Frankfurt, 60590 Frankfurt, Germany
| | - Dmitry Namgaladze
- Institute of Biochemistry I, Goethe-University Frankfurt, 60590 Frankfurt, Germany
| | - Nathalie Dehne
- Institute of Biochemistry I, Goethe-University Frankfurt, 60590 Frankfurt, Germany
| | - Bernhard Brüne
- Institute of Biochemistry I, Goethe-University Frankfurt, 60590 Frankfurt, Germany
| |
Collapse
|
12
|
Becht E, Giraldo NA, Germain C, de Reyniès A, Laurent-Puig P, Zucman-Rossi J, Dieu-Nosjean MC, Sautès-Fridman C, Fridman WH. Immune Contexture, Immunoscore, and Malignant Cell Molecular Subgroups for Prognostic and Theranostic Classifications of Cancers. Adv Immunol 2016; 130:95-190. [DOI: 10.1016/bs.ai.2015.12.002] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
13
|
Chinyengetere F, Sekula DJ, Lu Y, Giustini AJ, Sanglikar A, Kawakami M, Ma T, Burkett SS, Eisenberg BL, Wells WA, Hoopes PJ, Demicco EG, Lazar AJ, Torres KE, Memoli V, Freemantle SJ, Dmitrovsky E. Mice null for the deubiquitinase USP18 spontaneously develop leiomyosarcomas. BMC Cancer 2015; 15:886. [PMID: 26555296 PMCID: PMC4640382 DOI: 10.1186/s12885-015-1883-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 10/30/2015] [Indexed: 11/10/2022] Open
Abstract
Background USP18 (ubiquitin-specific protease 18) removes ubiquitin-like modifier interferon stimulated gene 15 (ISG15) from conjugated proteins. USP18 null mice in a FVB/N background develop tumors as early as 2 months of age. These tumors are leiomyosarcomas and thus represent a new murine model for this disease. Methods Heterozygous USP18 +/− FVB/N mice were bred to generate wild-type, heterozygous and homozygous cohorts. Tumors were characterized immunohistochemically and two cell lines were derived from independent tumors. Cell lines were karyotyped and their responses to restoration of USP18 activity assessed. Drug testing and tumorigenic assays were also performed. USP18 immunohistochemical staining in a large series of human leiomyosacomas was examined. Results USP18 −/− FVB/N mice spontaneously develop tumors predominantly on the back of the neck with most tumors evident between 6–12 months (80 % penetrance). Immunohistochemical characterization of the tumors confirmed they were leiomyosarcomas, which originate from smooth muscle. Restoration of USP18 activity in sarcoma-derived cell lines did not reduce anchorage dependent or independent growth or xenograft tumor formation demonstrating that these cells no longer require USP18 suppression for tumorigenesis. Karyotyping revealed that both tumor-derived cell lines were aneuploid with extra copies of chromosomes 3 and 15. Chromosome 15 contains the Myc locus and MYC is also amplified in human leiomyosarcomas. MYC protein levels were elevated in both murine leiomyosarcoma cell lines. Stabilized P53 protein was detected in a subset of these murine tumors, another feature of human leiomyosarcomas. Immunohistochemical analyses of USP18 in human leiomyosarcomas revealed a range of staining intensities with the highest USP18 expression in normal vascular smooth muscle. USP18 tissue array analysis of primary leiomyosarcomas from 89 patients with a clinical database revealed cases with reduced USP18 levels had a significantly decreased time to metastasis (P = 0.0441). Conclusions USP18 null mice develop leiomyosarcoma recapitulating key features of clinical leiomyosarcomas and patients with reduced-USP18 tumor levels have an unfavorable outcome. USP18 null mice and the derived cell lines represent clinically-relevant models of leiomyosarcoma and can provide insights into both leiomyosarcoma biology and therapy. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1883-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fadzai Chinyengetere
- Department of Pharmacology and Toxicology, Dartmouth, Hanover, NH, USA. .,Geisel School of Medicine, Dartmouth, Hanover, NH, USA. .,Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA.
| | - David J Sekula
- Department of Pharmacology and Toxicology, Dartmouth, Hanover, NH, USA. .,Geisel School of Medicine, Dartmouth, Hanover, NH, USA. .,Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA.
| | - Yun Lu
- Department of Pharmacology and Toxicology, Dartmouth, Hanover, NH, USA. .,Geisel School of Medicine, Dartmouth, Hanover, NH, USA. .,Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA.
| | - Andrew J Giustini
- Geisel School of Medicine, Dartmouth, Hanover, NH, USA. .,Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA. .,Thayer School of Engineering, Dartmouth College, Hanover, NH, USA.
| | | | - Masanori Kawakami
- Department of Pharmacology and Toxicology, Dartmouth, Hanover, NH, USA. .,Geisel School of Medicine, Dartmouth, Hanover, NH, USA. .,Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA.
| | - Tian Ma
- Department of Pharmacology and Toxicology, Dartmouth, Hanover, NH, USA. .,Geisel School of Medicine, Dartmouth, Hanover, NH, USA. .,Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA.
| | - Sandra S Burkett
- Comparative Molecular Cytogenetics Core, Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA.
| | - Burton L Eisenberg
- Department of Surgery, Dartmouth, Hanover, NH, USA. .,Norris Cotton Cancer Center, Lebanon, NH, USA. .,Geisel School of Medicine, Dartmouth, Hanover, NH, USA. .,Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA.
| | - Wendy A Wells
- Department of Pathology, Dartmouth, Hanover, NH, USA. .,Norris Cotton Cancer Center, Lebanon, NH, USA. .,Geisel School of Medicine, Dartmouth, Hanover, NH, USA. .,Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA.
| | - Paul J Hoopes
- Department of Surgery, Dartmouth, Hanover, NH, USA. .,Norris Cotton Cancer Center, Lebanon, NH, USA. .,Geisel School of Medicine, Dartmouth, Hanover, NH, USA. .,Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA. .,Thayer School of Engineering, Dartmouth College, Hanover, NH, USA.
| | | | - Alexander J Lazar
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. .,Sarcoma Research Center, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Keila E Torres
- Sarcoma Research Center, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. .,Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Vincent Memoli
- Department of Pathology, Dartmouth, Hanover, NH, USA. .,Norris Cotton Cancer Center, Lebanon, NH, USA. .,Geisel School of Medicine, Dartmouth, Hanover, NH, USA. .,Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA.
| | - Sarah J Freemantle
- Department of Pharmacology and Toxicology, Dartmouth, Hanover, NH, USA. .,Geisel School of Medicine, Dartmouth, Hanover, NH, USA. .,Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA.
| | - Ethan Dmitrovsky
- Department of Pharmacology and Toxicology, Dartmouth, Hanover, NH, USA. .,Department of Medicine, Dartmouth, Hanover, NH, USA. .,Norris Cotton Cancer Center, Lebanon, NH, USA. .,Geisel School of Medicine, Dartmouth, Hanover, NH, USA. .,Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA. .,Present address: MD Anderson Cancer Center, Houston, TX, 77030-4009, USA.
| |
Collapse
|
14
|
Thiery J, Lieberman J. Perforin: a key pore-forming protein for immune control of viruses and cancer. Subcell Biochem 2014; 80:197-220. [PMID: 24798013 DOI: 10.1007/978-94-017-8881-6_10] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Perforin (PFN) is the key pore-forming molecule in the cytotoxic granules of immune killer cells. Expressed only in killer cells, PFN is the rate-limiting molecule for cytotoxic function, delivering the death-inducing granule serine proteases (granzymes) into target cells marked for immune elimination. In this chapter we describe our current understanding of how PFN accomplishes this task. We discuss where PFN is expressed and how its expression is regulated, the biogenesis and storage of PFN in killer cells and how they are protected from potential damage, how it is released, how it delivers Granzymes into target cells and the consequences of PFN deficiency.
Collapse
Affiliation(s)
- Jerome Thiery
- INSERM U753, University Paris Sud and Gustave Roussy Cancer Campus, Villejuif, France,
| | | |
Collapse
|
15
|
Natividad KDT, Junankar SR, Mohd Redzwan N, Nair R, Wirasinha RC, King C, Brink R, Swarbrick A, Batten M. Interleukin-27 signaling promotes immunity against endogenously arising murine tumors. PLoS One 2013; 8:e57469. [PMID: 23554861 PMCID: PMC3595259 DOI: 10.1371/journal.pone.0057469] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Accepted: 01/21/2013] [Indexed: 12/29/2022] Open
Abstract
Interleukin-27 (IL-27) is a pleiotropic cytokine but its immunosuppressive effects predominate during many in vivo immunological challenges. Despite this, evidence from tumor cell line transfer models suggested that IL-27 could promote immune responses in the tumor context. However, the role of IL-27 in immunity against tumors that develop in situ and in tumor immunosurveillance remain undefined. In this study, we demonstrate that tumor development and growth are accelerated in IL-27 receptor α (Il27ra)-deficient mice. Enhanced tumor growth in both carcinogen-induced fibrosarcoma and oncogene-driven mammary carcinoma was associated with decreased interferon-γ production by CD4 and CD8 T cells and increased numbers of regulatory T-cells (Treg). This is the first study to show that IL-27 promotes protective immune responses against endogenous tumors, which is critical as the basis for future development of an IL-27 based therapeutic agent.
Collapse
MESH Headings
- Animals
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/pathology
- Immune Tolerance/genetics
- Interferon-gamma/genetics
- Interferon-gamma/immunology
- Interleukins/genetics
- Interleukins/immunology
- Male
- Mice
- Mice, Knockout
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/pathology
- Neoplasms, Experimental/therapy
- Receptors, Cytokine/genetics
- Receptors, Cytokine/immunology
- Receptors, Interleukin
- Signal Transduction/genetics
- Signal Transduction/immunology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/pathology
Collapse
Affiliation(s)
- Karlo D. T. Natividad
- Immunological Diseases Division, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Simon R. Junankar
- Cancer Division, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Norhanani Mohd Redzwan
- Immunological Diseases Division, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Radhika Nair
- Cancer Division, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Rushika C. Wirasinha
- Immunological Diseases Division, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Cecile King
- Immunological Diseases Division, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St. Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | - Robert Brink
- Immunological Diseases Division, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St. Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | - Alexander Swarbrick
- Cancer Division, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St. Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | - Marcel Batten
- Immunological Diseases Division, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St. Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia
- * E-mail:
| |
Collapse
|
16
|
Oleinika K, Nibbs RJ, Graham GJ, Fraser AR. Suppression, subversion and escape: the role of regulatory T cells in cancer progression. Clin Exp Immunol 2013. [PMID: 23199321 DOI: 10.1111/j.1365-2249.2012.04657.x] [Citation(s) in RCA: 156] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Regulatory T cells (T(regs) ) are crucial in mediating immune homeostasis and promoting the establishment and maintenance of peripheral tolerance. However, in the context of cancer their role is more complex, and they are thought to contribute to the progress of many tumours. As cancer cells express both self- and tumour-associated antigens, T(regs) are key to dampening effector cell responses, and therefore represent one of the main obstacles to effective anti-tumour responses. Suppression mechanisms employed by T(regs) are thought to contribute significantly to the failure of current therapies that rely on induction or potentiation of anti-tumour responses. This review will focus on the current evidence supporting the central role of T(regs) in establishing tumour-specific tolerance and promoting cancer escape. We outline the mechanisms underlying their suppressive function and discuss the potential routes of T(regs) accumulation within the tumour, including enhanced recruitment, in-situ or local proliferation, and de-novo differentiation. In addition, we review some of the cancer treatment strategies that act, at least in part, to eliminate or interfere with the function of T(regs) . The role of T(regs) is being recognized increasingly in cancer, and controlling the function of these suppressive cells in the tumour microenvironment without compromising peripheral tolerance represents a significant challenge for cancer therapies.
Collapse
Affiliation(s)
- K Oleinika
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | | | | | | |
Collapse
|
17
|
Balza E, Castellani P, Delfino L, Truini M, Rubartelli A. The pharmacologic inhibition of the xc- antioxidant system improves the antitumor efficacy of COX inhibitors in the in vivo model of 3-MCA tumorigenesis. Carcinogenesis 2012; 34:620-6. [PMID: 23161574 DOI: 10.1093/carcin/bgs360] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The chemopreventive and therapeutic efficacy of the cyclooxygenase (COX) inhibitor ibuprofen (IB) and of sulfasalazine (SASP), a drug that targets the antioxidant xc- system, were exploited in the experimental model of 3-methylcholantrene (3-MCA)-induced mouse sarcoma. The chemopreventive treatments gave unsatisfactory results because administration of IB one day after the 3-MCA injection only slightly delayed the tumor development, whereas SASP dispensed under the same conditions resulted in accelerated tumorigenesis. Similarly, the therapeutic treatment with either drug, administrated daily from the tumor detection, decreased the proliferation rate of tumor cells and increased the survival of treated mice only at a low extent. Remarkably, the combined chemopreventive treatment with IB and therapeutic treatment with SASP displayed a better efficacy, with strong delay of sarcoma growth, reduced tumor size and increased survival of treated mice. The two drugs target not only tumor cells but also tumor-associated macrophages that were dramatically decreased in the tumor infiltrate of mice subjected to the combined treatment. The synergistic effects of the association between a broad anti-inflammatory compound, such as IB, and a redox-directed drug, such as SASP, shed new light in the role of inflammation and of the redox response in chemical tumorigenesis and point to the combined chemopreventive plus therapeutic treatment with IB and SASP as a promising novel approach for antitumor therapy.
Collapse
Affiliation(s)
- Enrica Balza
- Unit of Cellular Biology, IRCCS AOU San Martino-IST, Genoa 16132, Italy
| | | | | | | | | |
Collapse
|
18
|
Briesemeister D, Friese C, Isern CC, Dietz E, Blankenstein T, Thoene-Reineke C, Kammertoens T. Differences in serum cytokine levels between wild type mice and mice with a targeted mutation suggests necessity of using control littermates. Cytokine 2012; 60:626-33. [PMID: 22902947 DOI: 10.1016/j.cyto.2012.07.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2011] [Revised: 07/12/2012] [Accepted: 07/14/2012] [Indexed: 01/09/2023]
Abstract
To enhance protection from pathogens, housing conditions have been improved constantly. We wanted to test whether various environmental conditions and caging systems affect serum cytokine levels of immunodeficient mice differently than they affect immunocompetent control animals. We compared serum cytokine levels of immunodeficient and immunocompetent mice kept in three different environments: a specific pathogen free (SPF) breeding barrier with open cages. An SPF experimental unit with individually ventilated cages. An experimental semi-barrier with open cages. Serum from Rag1(-/-), μMT(-/-), IFN-γR(-/-), IFN-γ(-/-), IL-4(-/-), the heterozygous controls and wild type C57BL/6 or BALB/c mice was analyzed for the presence of 10 cytokines (IL-1α, IL-2, IL-4, IL-5, IL-6, IL-10, IL-17, IFN-γ, TNF-α and GM-CSF). No major changes in cytokine levels were detected in mice exposed to different housing conditions. However, irrespective of immunodeficiency at 4 weeks of age a number of mice from the breeding colonies with a targeted mutation (TM), both -/- and +/- mice, showed a statistically significant elevation of some cytokines (primarily IL-1α, IL-5) when compared to wild type BALB/c and C57BL/6 mice. We conclude that under SPF conditions, immunodeficient mice can be kept either in open caging or IVC systems without affecting serum cytokine levels. The more important conclusion, however, stems from the observation that there is a significant difference in serum cytokine levels between wild type and mice carrying either one or two alleles of a targeted mutation (either -/- and +/- mice). This suggests an altered base-line inflammatory responsiveness in the TM-breeding colonies.
Collapse
Affiliation(s)
- Dana Briesemeister
- Institute of Immunology, Charité Campus Benjamin Franklin, 12200 Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
19
|
Träger U, Sierro S, Djordjevic G, Bouzo B, Khandwala S, Meloni A, Mortensen M, Simon AK. The immune response to melanoma is limited by thymic selection of self-antigens. PLoS One 2012; 7:e35005. [PMID: 22506061 PMCID: PMC3323626 DOI: 10.1371/journal.pone.0035005] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 03/12/2012] [Indexed: 12/18/2022] Open
Abstract
The expression of melanoma-associated antigens (MAA) being limited to normal melanocytes and melanomas, MAAs are ideal targets for immunotherapy and melanoma vaccines. As MAAs are derived from self, immune responses to these may be limited by thymic tolerance. The extent to which self-tolerance prevents efficient immune responses to MAAs remains unknown. The autoimmune regulator (AIRE) controls the expression of tissue-specific self-antigens in thymic epithelial cells (TECs). The level of antigens expressed in the TECs determines the fate of auto-reactive thymocytes. Deficiency in AIRE leads in both humans (APECED patients) and mice to enlarged autoreactive immune repertoires. Here we show increased IgG levels to melanoma cells in APECED patients correlating with autoimmune skin features. Similarly, the enlarged T cell repertoire in AIRE−/− mice enables them to mount anti-MAA and anti-melanoma responses as shown by increased anti-melanoma antibodies, and enhanced CD4+ and MAA-specific CD8+ T cell responses after melanoma challenge. We show that thymic expression of gp100 is under the control of AIRE, leading to increased gp100-specific CD8+ T cell frequencies in AIRE−/− mice. TRP-2 (tyrosinase-related protein), on the other hand, is absent from TECs and consequently TRP-2 specific CD8+ T cells were found in both AIRE−/− and AIRE+/+ mice. This study emphasizes the importance of investigating thymic expression of self-antigens prior to their inclusion in vaccination and immunotherapy strategies.
Collapse
Affiliation(s)
- Ulrike Träger
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- NIHR Biomedical Centre, Oxford, United Kingdom
| | - Sophie Sierro
- Ludwig Institute for Cancer Research, Epalinges, Switzerland
| | - Gordana Djordjevic
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- NIHR Biomedical Centre, Oxford, United Kingdom
| | - Basma Bouzo
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- NIHR Biomedical Centre, Oxford, United Kingdom
| | - Shivani Khandwala
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- NIHR Biomedical Centre, Oxford, United Kingdom
| | - Antonella Meloni
- Pediatric Clinic II, Ospedale Microcitemico and Department of Biomedical and Biotechnological Science, University of Cagliari, Cagliari, Italy
| | - Monika Mortensen
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- NIHR Biomedical Centre, Oxford, United Kingdom
- Apoptosis Department and Center for Genotoxic Stress Research, Institute of Cancer Biology, Danish Cancer Society, Copenhagen, Denmark
| | - Anna Katharina Simon
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- NIHR Biomedical Centre, Oxford, United Kingdom
- * E-mail:
| |
Collapse
|
20
|
Kammertoens T, Qin Z, Briesemeister D, Bendelac A, Blankenstein T. B-cells and IL-4 promote methylcholanthrene-induced carcinogenesis but there is no evidence for a role of T/NKT-cells and their effector molecules (Fas-ligand, TNF-α, perforin). Int J Cancer 2012; 131:1499-508. [PMID: 22212899 DOI: 10.1002/ijc.27411] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2011] [Accepted: 12/02/2011] [Indexed: 12/11/2022]
Abstract
Mice deficient either in subtypes of immune cells, cytokines or lytic pathways have been subjected to chemical carcinogenesis by methylcholanthrene to evaluate whether these components of the immune system affect tumor development. Inbred mice of the same genotype but from different sources differed in tumor development in magnitude comparable to that previously attributed to differences in immunocompetence. This suggested that genetic drift between separate inbred colonies of mice and/or environmental factors (e.g., transport of the animals) influenced carcinogenesis. Therefore, littermates were used as control in subsequent experiments. Although deficiency of T-cells, NKT-cells, perforin, Fas-ligand, TNF-α-receptor failed to reveal significant differences in tumor development, the presence of B-cells and IL-4 enhanced tumor development under similar experimental conditions.
Collapse
Affiliation(s)
- Thomas Kammertoens
- Institut für Immunologie, Charité, Campus Benjamin-Franklin, Berlin, Germany
| | | | | | | | | |
Collapse
|
21
|
Murphy MA, O'Leary JJ, Cahill DJ. Assessment of the humoral immune response to cancer. J Proteomics 2012; 75:4573-9. [PMID: 22300580 DOI: 10.1016/j.jprot.2012.01.021] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Revised: 01/13/2012] [Accepted: 01/16/2012] [Indexed: 12/21/2022]
Abstract
One of the deadly hallmarks of cancer is its ability to prosper within the constraints of the host immune system. Recent advances in immunoproteomics and high-throughput technologies have lead to profiling of the antibody repertoire in cancer patients. This in turn has lead to the identification of tumour associated antigens/autoantibodies. Autoantibodies are extremely attractive and promising biomarker entities, however there has been relatively little discussion on how to interpret the humoral immune response. It may be that autoantibody profiles hold the key to ultimately uncovering neoplastic associated pathways and through the process of immunosculpting the tumour may have yielded an immune response in the early stages of malignant tumour development. The aim of this review is to discuss the utility of the autoantibody response that is elicited as a result of malignancy and discuss the advantages and limitations of autoantibody profiling. This article is part of a Special Issue entitled: Translational Proteomics.
Collapse
Affiliation(s)
- Mairead Anne Murphy
- Department of Histopathology, School of Medicine, Trinity College, Institute of Molecular Medicine, St James's Hospital, Dublin 8, Ireland
| | | | | |
Collapse
|
22
|
Fox BA, Schendel DJ, Butterfield LH, Aamdal S, Allison JP, Ascierto PA, Atkins MB, Bartunkova J, Bergmann L, Berinstein N, Bonorino CC, Borden E, Bramson JL, Britten CM, Cao X, Carson WE, Chang AE, Characiejus D, Choudhury AR, Coukos G, de Gruijl T, Dillman RO, Dolstra H, Dranoff G, Durrant LG, Finke JH, Galon J, Gollob JA, Gouttefangeas C, Grizzi F, Guida M, Håkansson L, Hege K, Herberman RB, Hodi FS, Hoos A, Huber C, Hwu P, Imai K, Jaffee EM, Janetzki S, June CH, Kalinski P, Kaufman HL, Kawakami K, Kawakami Y, Keilholtz U, Khleif SN, Kiessling R, Kotlan B, Kroemer G, Lapointe R, Levitsky HI, Lotze MT, Maccalli C, Maio M, Marschner JP, Mastrangelo MJ, Masucci G, Melero I, Melief C, Murphy WJ, Nelson B, Nicolini A, Nishimura MI, Odunsi K, Ohashi PS, O'Donnell-Tormey J, Old LJ, Ottensmeier C, Papamichail M, Parmiani G, Pawelec G, Proietti E, Qin S, Rees R, Ribas A, Ridolfi R, Ritter G, Rivoltini L, Romero PJ, Salem ML, Scheper RJ, Seliger B, Sharma P, Shiku H, Singh-Jasuja H, Song W, Straten PT, Tahara H, Tian Z, van Der Burg SH, von Hoegen P, Wang E, Welters MJP, Winter H, Withington T, Wolchok JD, Xiao W, Zitvogel L, et alFox BA, Schendel DJ, Butterfield LH, Aamdal S, Allison JP, Ascierto PA, Atkins MB, Bartunkova J, Bergmann L, Berinstein N, Bonorino CC, Borden E, Bramson JL, Britten CM, Cao X, Carson WE, Chang AE, Characiejus D, Choudhury AR, Coukos G, de Gruijl T, Dillman RO, Dolstra H, Dranoff G, Durrant LG, Finke JH, Galon J, Gollob JA, Gouttefangeas C, Grizzi F, Guida M, Håkansson L, Hege K, Herberman RB, Hodi FS, Hoos A, Huber C, Hwu P, Imai K, Jaffee EM, Janetzki S, June CH, Kalinski P, Kaufman HL, Kawakami K, Kawakami Y, Keilholtz U, Khleif SN, Kiessling R, Kotlan B, Kroemer G, Lapointe R, Levitsky HI, Lotze MT, Maccalli C, Maio M, Marschner JP, Mastrangelo MJ, Masucci G, Melero I, Melief C, Murphy WJ, Nelson B, Nicolini A, Nishimura MI, Odunsi K, Ohashi PS, O'Donnell-Tormey J, Old LJ, Ottensmeier C, Papamichail M, Parmiani G, Pawelec G, Proietti E, Qin S, Rees R, Ribas A, Ridolfi R, Ritter G, Rivoltini L, Romero PJ, Salem ML, Scheper RJ, Seliger B, Sharma P, Shiku H, Singh-Jasuja H, Song W, Straten PT, Tahara H, Tian Z, van Der Burg SH, von Hoegen P, Wang E, Welters MJP, Winter H, Withington T, Wolchok JD, Xiao W, Zitvogel L, Zwierzina H, Marincola FM, Gajewski TF, Wigginton JM, Disis ML. Defining the critical hurdles in cancer immunotherapy. J Transl Med 2011; 9:214. [PMID: 22168571 PMCID: PMC3338100 DOI: 10.1186/1479-5876-9-214] [Show More Authors] [Citation(s) in RCA: 117] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Accepted: 12/14/2011] [Indexed: 02/07/2023] Open
Abstract
Scientific discoveries that provide strong evidence of antitumor effects in preclinical models often encounter significant delays before being tested in patients with cancer. While some of these delays have a scientific basis, others do not. We need to do better. Innovative strategies need to move into early stage clinical trials as quickly as it is safe, and if successful, these therapies should efficiently obtain regulatory approval and widespread clinical application. In late 2009 and 2010 the Society for Immunotherapy of Cancer (SITC), convened an "Immunotherapy Summit" with representatives from immunotherapy organizations representing Europe, Japan, China and North America to discuss collaborations to improve development and delivery of cancer immunotherapy. One of the concepts raised by SITC and defined as critical by all parties was the need to identify hurdles that impede effective translation of cancer immunotherapy. With consensus on these hurdles, international working groups could be developed to make recommendations vetted by the participating organizations. These recommendations could then be considered by regulatory bodies, governmental and private funding agencies, pharmaceutical companies and academic institutions to facilitate changes necessary to accelerate clinical translation of novel immune-based cancer therapies. The critical hurdles identified by representatives of the collaborating organizations, now organized as the World Immunotherapy Council, are presented and discussed in this report. Some of the identified hurdles impede all investigators; others hinder investigators only in certain regions or institutions or are more relevant to specific types of immunotherapy or first-in-humans studies. Each of these hurdles can significantly delay clinical translation of promising advances in immunotherapy yet if overcome, have the potential to improve outcomes of patients with cancer.
Collapse
Affiliation(s)
- Bernard A Fox
- Earle A. Chiles Research Institute, Robert W. Franz Research Center, Providence Cancer Center, Providence Portland Medical Center, Portland, OR, USA
- Department of Molecular Microbiology and Immunology and Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Dolores J Schendel
- Institute of Molecular Immunology and Clinical Cooperation Group "Immune Monitoring", Helmholtz Centre Munich, German Research Center for Environmental Health, Munich, Germany
| | - Lisa H Butterfield
- Departments of Medicine, Division of Hematology Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
- Department of Surgery University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Steinar Aamdal
- Department of Clinical Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - James P Allison
- Memorial Sloan-Kettering Cancer Center, New York, NY, USA
- Howard Hughes Medical Institute, New York, NY, USA
| | - Paolo Antonio Ascierto
- Medical Oncology and Innovative Therapy, Instituto Nazionale Tumori-Fondazione 'G. Pascale', Naples, Italy
| | - Michael B Atkins
- Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Jirina Bartunkova
- Institute of Immunology, FOCIS Center of Excellence, 2nd Medical School, Charles University, Prague, Czech Republic
| | - Lothar Bergmann
- Goethe Universität Frankfurt Am Main,Medizinische Klinik II, Frankfurt Am Main, Germany
| | | | - Cristina C Bonorino
- Instituto Nacional para o Controle do Câncer, Instituto de Pesquisas Biomédicas, PUCRS Faculdade de Biociências, PUCRS, Porto Alegre RS Brazil
| | - Ernest Borden
- Department of Translational Hematology and Oncology Research, Cleveland Clinic, Cleveland, OH, USA
- Department of Solid Tumor Oncology, Cleveland Clinic, Cleveland, OH, USA
| | | | - Cedrik M Britten
- University Medical Center Mainz, III. Medical Department, Mainz, Germany
- Ribological GmbH, Mainz, Germany
| | - Xuetao Cao
- Chinese Academy of Medical Sciences, Beijing, China
- Institute of Immunology, National Key Laboratory of Medical Immunology, Second Military Medical University, Shanghai, China
| | | | - Alfred E Chang
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI
| | | | | | - George Coukos
- Ovarian Cancer Research Center, University of Pennsylvania Medical Center, Philadelphia, A, USA
| | - Tanja de Gruijl
- Department of Medical Oncology, VU Medical Center, Cancer Center Amsterdam Amsterdam, The Netherlands
| | - Robert O Dillman
- Hoag Institute for Research and Education, Hoag Cancer Institute, Newport Beach, CA, USA
| | - Harry Dolstra
- Department of Laboratory Medicine, Nijmegen Centre for Molecular Life Sciences, Radboud University, Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Glenn Dranoff
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Lindy G Durrant
- Academic Department of Clinical Oncology, University of Nottingham, Nottingham, UK
| | - James H Finke
- Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Jerome Galon
- INSERM U872, Cordeliers Research Center, Paris, France
| | | | - Cécile Gouttefangeas
- Institute for Cell Biology, Department of Immunology, University of Tuebingen, Tuebingen, Germany
| | | | | | - Leif Håkansson
- University of Lund, Lund, Sweden
- CanImGuide Therapeutics AB, Hoellviken, Sweden
| | - Kristen Hege
- University of California, San Francisco, CA and Celgene Corporation, San Francisco, CA, USA
| | | | - F Stephen Hodi
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Axel Hoos
- Bristol-Myers Squibb Company, Wallingford, Connecticut, USA
| | - Christoph Huber
- Translational Oncology & Immunology Centre TRON at the Mainz University Medical Center, Mainz, Germany
| | - Patrick Hwu
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Kohzoh Imai
- The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Elizabeth M Jaffee
- Department of Oncology, the Sidney Kimmel Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | | | - Carl H June
- Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Pawel Kalinski
- Department of Surgery University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Howard L Kaufman
- Rush University Cancer Center, Rush University Medical Center, Chicago, IL, USA
| | - Koji Kawakami
- School of Medicine and Public Health, Kyoto University, Kyoto, Japan
| | - Yutaka Kawakami
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Ulrich Keilholtz
- Dept. of Hematology and Medical Oncology, Charité Comprehensive Cancer Center, Berlin, Germany
| | | | - Rolf Kiessling
- Department of Oncology - Pathology, Cancer Center Karolinska, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Beatrix Kotlan
- Department of Molecular Immunology and Toxicology, Center of Surgical and Molecular Tumor pathology, National Institute of Oncology, Budapest, Hungary
| | - Guido Kroemer
- INSERM, U848, Institut Gustave Roussy, Villejuif, France
| | - Rejean Lapointe
- Research Center, University Hospital, Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Institut du Cancer de, Montréal, Montréal, Québec, Canada
| | - Hyam I Levitsky
- School of Medicine, Oncology Center, Johns Hopkins University, Baltimore, MD, USA
| | - Michael T Lotze
- Departments of Medicine, Division of Hematology Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
- Department of Surgery University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Cristina Maccalli
- Department of Molecular Oncology, Foundation San Raffaele Scientific Institute, Milan, Italy
| | - Michele Maio
- Medical Oncology and Immunotherapy, Department of Oncology, University, Hospital of Siena, Istituto Toscano Tumori, Siena, Italy
| | | | | | - Giuseppe Masucci
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | - Ignacio Melero
- Department of Immunology, CIMA, CUN and Medical School University of Navarra, Pamplona, Spain
| | - Cornelius Melief
- Deptartment of Immunohematology and Blood Transfusion, Leiden University Medical Centre, Leiden, the Netherlands
| | - William J Murphy
- University of California-Davis Medical Center, Sacramento, CA, USA
| | - Brad Nelson
- Deeley Research Centre, BC Cancer Agency, Victoria, BC, Canada
| | - Andrea Nicolini
- Department of Internal Medicine, University of Pisa, Santa Chiara Hospital, Pisa, Italy
| | - Michael I Nishimura
- Oncology Institute, Loyola University Medical Center, Cardinal Bernardin Cancer Center, Maywood, IL, USA
| | - Kunle Odunsi
- Department of Gynecologic Oncology, Tumor Immunology and Immunotherapy Program, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Pamela S Ohashi
- Ontario Cancer Institute/University Health Network, Toronto, ON, Canada
| | | | - Lloyd J Old
- Ludwig Institute for Cancer Research, New York, NY, USA
| | - Christian Ottensmeier
- Experimental Cancer Medicine Centre, University of Southampton Faculty of Medicine, Southampton, UK
| | - Michael Papamichail
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, Athens, Greece
| | - Giorgio Parmiani
- Unit of Immuno-Biotherapy of Melanoma and Solid Tumors, San Raffaele Scientific Institute, Milan, Italy
| | - Graham Pawelec
- Center for Medical Research, University of Tuebingen, Tuebingen, Germany
| | | | - Shukui Qin
- Chinese PLA Cancer Center, Nanjing, China
| | - Robert Rees
- The John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Antoni Ribas
- Department of Medicine, Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California, USA
| | - Ruggero Ridolfi
- Immunoterapia e Terapia Cellulare Somatica, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (I.R.S.T.), Meldola (FC), Italy
| | - Gerd Ritter
- Memorial Sloan-Kettering Cancer Center, New York, NY, USA
- Ludwig Institute for Cancer Research, New York, NY, USA
| | - Licia Rivoltini
- Unit of Immunotherapy of Human Tumors, IRCCS Foundation, Istituto Nazionale Tumori, Milan, Italy
| | - Pedro J Romero
- Division of Clinical Onco-Immunology, Ludwig Center for Cancer Research of the University of Lausanne, Epalinges, Switzerland
| | - Mohamed L Salem
- Immunology and Biotechnology Unit, Department of Zoology, Faculty of Science, Tanta University, Egypt
| | - Rik J Scheper
- Dept. of Pathology, VU University Medical Center, Amsterdam, The Netherlands
| | | | | | - Hiroshi Shiku
- Department of Cancer Vaccine, Mie University Graduate School of Medicine, Mie, Japan
- Department of Immuno-gene Therapy, Mie University Graduate School of Medicine, Mie, Japan
| | | | - Wenru Song
- Millennium: The Takeda Oncology Company, Cambridge, MA, USA
| | - Per Thor Straten
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, Herlev Hospital, Herlev, Denmark
| | - Hideaki Tahara
- Department of Surgery and Bioengineering, Advanced Clinical Research Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Zhigang Tian
- Institute of Immunology, School of Life Sciences, University of Science & Technology of China, Hefei, China
- Institute of Immunopharmacology & Immunotherapy, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Sjoerd H van Der Burg
- Experimental Cancer Immunology and Therapy, Department of Clinical Oncology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Ena Wang
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center, NIH, Bethesda, MD, USA
- Center for Human Immunology (CHI), NIH, Bethesda, MD, USA
| | - Marij JP Welters
- Experimental Cancer Immunology and Therapy, Department of Clinical Oncology (K1-P), Leiden University Medical Center, Leiden, The Netherlands
| | - Hauke Winter
- Department of Surgery, Klinikum Grosshadern, Ludwig Maximilians University, Munich, Germany
| | | | - Jedd D Wolchok
- Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Weihua Xiao
- Institute of Immunology, School of Life Science, University of Science and Technology of China, Hefei, China
| | - Laurence Zitvogel
- Institut Gustave Roussy, Center of Clinical Investigations CICBT507, Villejuif, France
| | - Heinz Zwierzina
- Department Haematology and Oncology Innsbruck Medical University, Innsbruck, Austria
| | - Francesco M Marincola
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center, NIH, Bethesda, MD, USA
- Center for Human Immunology (CHI), NIH, Bethesda, MD, USA
| | | | - Jon M Wigginton
- Discovery Medicine-Oncology, Bristol-Myers Squibb Company, Princeton, New Jersey, USA
| | - Mary L Disis
- Tumor Vaccine Group, Center for Translational Medicine in Women's Health, University of Washington, Seattle, WA, USA
| |
Collapse
|
23
|
Wilke CM, Wei S, Wang L, Kryczek I, Kao J, Zou W. Dual biological effects of the cytokines interleukin-10 and interferon-γ. Cancer Immunol Immunother 2011; 60:1529-41. [PMID: 21918895 PMCID: PMC11029274 DOI: 10.1007/s00262-011-1104-5] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Accepted: 08/23/2011] [Indexed: 12/21/2022]
Abstract
It is generally thought that each cytokine exerts either immune stimulatory (inflammatory) or immune inhibitory (antiinflammatory or regulatory) biological activities. However, multiple cytokines can enact both inhibitory and stimulatory effects on the immune system. Two of these cytokines are interleukin (IL)-10 and interferon-gamma (IFNγ). IL-10 has demonstrated antitumor immunity even though it has been known for years as an immunoregulatory protein. Generally perceived as an immune stimulatory cytokine, IFNγ can also induce inhibitory molecule expression including B7-H1 (PD-L1), indoleamine 2,3-dioxygenase (IDO), and arginase on multiple cell populations (dendritic cells, tumor cells, and vascular endothelial cells). In this review, we will summarize current knowledge of the dual roles of both of these cytokines and stress the previously underappreciated stimulatory role of IL-10 and inhibitory role of IFNγ in the context of malignancy. Our progressive understanding of the dual effects of these cytokines is important for dissecting cytokine-associated pathology and provides new avenues for developing effective immune therapy against human diseases, including cancer.
Collapse
Affiliation(s)
- Cailin Moira Wilke
- Department of Surgery, University of Michigan School of Medicine, C560B MSRB II/Box 0669, 1150 W. Medical Center Drive, Ann Arbor, MI 48109-0669 USA
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI USA
| | - Shuang Wei
- Department of Surgery, University of Michigan School of Medicine, C560B MSRB II/Box 0669, 1150 W. Medical Center Drive, Ann Arbor, MI 48109-0669 USA
| | - Lin Wang
- Department of Surgery, University of Michigan School of Medicine, C560B MSRB II/Box 0669, 1150 W. Medical Center Drive, Ann Arbor, MI 48109-0669 USA
- Central Laboratory, Union Hospital, Tongji Medical School, Huazhong University of Science and Technology, Wuhan, China
| | - Ilona Kryczek
- Department of Surgery, University of Michigan School of Medicine, C560B MSRB II/Box 0669, 1150 W. Medical Center Drive, Ann Arbor, MI 48109-0669 USA
| | - John Kao
- Department of Medicine, University of Michigan, Ann Arbor, MI USA
- University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI USA
| | - Weiping Zou
- Department of Surgery, University of Michigan School of Medicine, C560B MSRB II/Box 0669, 1150 W. Medical Center Drive, Ann Arbor, MI 48109-0669 USA
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI USA
- University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI USA
- Graduate Program in Cancer Biology, University of Michigan, Ann Arbor, MI USA
| |
Collapse
|
24
|
[Prostate carcinoma: vaccination as a new option for treatment]. Urologe A 2011; 51:44-9. [PMID: 21989588 DOI: 10.1007/s00120-011-2712-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Immune therapy and tumor cell vaccination is a challenging option in prostate cancer therapy, especially as side effects rarely occur. This review highlights recent developments in vaccination therapy of prostate cancer. The FDA approved antigen presenting cell vaccine Sipuleucel-T is described and new strategies of immune therapy like RNA and peptide vaccination are discussed in detail. Currently the effect of prostate cancer vaccination has still limitations, at least partially due to the immune suppressive effects of the tumor microenvironment and regulatory T cells, which suppress the immune effector function. To overcome these hurdles the concept of immune checkpoint modulation, which has the aim to break tolerance mechanisms, is discussed. Potential clinical therapies of checkpoint modulation are outlined.
Collapse
|
25
|
Foster AD, Sivarapatna A, Gress RE. The aging immune system and its relationship with cancer. ACTA ACUST UNITED AC 2011; 7:707-718. [PMID: 22121388 DOI: 10.2217/ahe.11.56] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The incidence of most common cancers increases with age. This occurs in association with, and is possibly caused by a decline in immune function, termed immune senescence. Although the size of the T-cell compartment is quantitatively maintained into older age, several deleterious changes (including significant changes to T-cell subsets) occur over time that significantly impair immunity. This article highlights some of the recent findings regarding the aging immune system, with an emphasis on the T-cell compartment and its role in cancer.
Collapse
Affiliation(s)
- Anthony D Foster
- National Cancer Institute (NCI), Experimental Transplantation & Immunology Branch (ETIB), 10 Center Dr. 10 CRC, 3-3330 Bethesda, MD 20814, USA
| | | | | |
Collapse
|
26
|
Soto AM, Sonnenschein C. The tissue organization field theory of cancer: a testable replacement for the somatic mutation theory. Bioessays 2011; 33:332-40. [PMID: 21503935 DOI: 10.1002/bies.201100025] [Citation(s) in RCA: 233] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The somatic mutation theory (SMT) of cancer has been and remains the prevalent theory attempting to explain how neoplasms arise and progress. This theory proposes that cancer is a clonal, cell-based disease, and implicitly assumes that quiescence is the default state of cells in multicellular organisms. The SMT has not been rigorously tested, and several lines of evidence raise questions that are not addressed by this theory. Herein, we propose experimental strategies that may validate the SMT. We also call attention to an alternative theory of carcinogenesis, the tissue organization field theory (TOFT), which posits that cancer is a tissue-based disease and that proliferation is the default state of all cells. Based on epistemological and experimental evidence, we argue that the TOFT compellingly explains carcinogenesis, while placing it within an evolutionarily relevant context.
Collapse
Affiliation(s)
- Ana M Soto
- Department of Anatomy and Cellular Biology, Tufts University School of Medicine, Boston, MA, USA
| | | |
Collapse
|
27
|
Nesbeth Y, Conejo-Garcia JR. Harnessing the effect of adoptively transferred tumor-reactive T cells on endogenous (host-derived) antitumor immunity. Clin Dev Immunol 2010; 2010:139304. [PMID: 21076522 PMCID: PMC2975067 DOI: 10.1155/2010/139304] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2010] [Accepted: 08/05/2010] [Indexed: 12/21/2022]
Abstract
Adoptive T cell transfer therapy, the ex vivo activation, expansion, and subsequent administration of tumor-reactive T cells, is already the most effective therapy against certain types of cancer. However, recent evidence in animal models and clinical trials suggests that host conditioning interventions tailored for some of the most aggressive and frequent epithelial cancers will be needed to maximize the benefit of this approach. Similarly, the subsets, stage of differentiation, and ex vivo expansion procedure of tumor-reactive T cells to be adoptively transferred influence their in vivo effectiveness and may need to be adapted for different types of cancer and host conditioning interventions. The effects of adoptively transferred tumor-reactive T cells on the mechanisms of endogenous (host-derived) antitumor immunity, and how to maximize their combined effects, are further discussed.
Collapse
Affiliation(s)
- Yolanda Nesbeth
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, NH 03756, USA
| | | |
Collapse
|
28
|
The initial immune reaction to a new tumor antigen is always stimulatory and probably necessary for the tumor's growth. Clin Dev Immunol 2010; 2010. [PMID: 20811480 PMCID: PMC2926581 DOI: 10.1155/2010/851728] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Accepted: 06/03/2010] [Indexed: 12/20/2022]
Abstract
All nascent neoplasms probably elicit at least a weak immune reaction. However, the initial effect of the weak immune reaction on a nascent tumor is always stimulatory rather than inhibitory to tumor growth, assuming only that exposure to the tumor antigens did not antedate the initiation of the neoplasm (as may occur in some virally induced tumors). This conclusion derives from the observation that the relationship between the magnitude of an adaptive immune reaction and tumor growth is not linear but varies such that while large quantities of antitumor immune reactants tend to inhibit tumor growth, smaller quantities of the same reactants are, for unknown reasons, stimulatory. Any immune reaction must presumably be small before it can become large; hence the initial reaction to the first presentation of a tumor antigen must always be small and in the stimulatory portion of this nonlinear relationship. In mouse-skin carcinogenesis experiments it was found that premalignant papillomas were variously immunogenic, but that the carcinomas that arose in them were, presumably because of induced immune tolerance, nonimmunogenic in the animal of origin.
Collapse
|
29
|
Voskoboinik I, Dunstone MA, Baran K, Whisstock JC, Trapani JA. Perforin: structure, function, and role in human immunopathology. Immunol Rev 2010; 235:35-54. [PMID: 20536554 DOI: 10.1111/j.0105-2896.2010.00896.x] [Citation(s) in RCA: 151] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The secretory granule-mediated cell death pathway is the key mechanism for elimination of virus-infected and transformed target cells by cytotoxic lymphocytes. The formation of the immunological synapse between an effector and a target cell leads to exocytic trafficking of the secretory granules and the release of their contents, which include pro-apoptotic serine proteases, granzymes, and pore-forming perforin into the synapse. There, perforin polymerizes and forms a transmembrane pore that allows the delivery of granzymes into the cytosol, where they initiate various apoptotic death pathways. Unlike relatively redundant individual granzymes, functional perforin is absolutely essential for cytotoxic lymphocyte function and immune regulation in the host. Nevertheless, perforin is still the least studied and understood cytotoxic molecule in the immune system. In this review, we discuss the current state of affairs in the perforin field: the protein's structure and function as well as its role in immune-mediated diseases.
Collapse
Affiliation(s)
- Ilia Voskoboinik
- Cancer Cell Death Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Vic. 8006, Australia
| | | | | | | | | |
Collapse
|
30
|
Schreiber TH, Raez L, Rosenblatt JD, Podack ER. Tumor immunogenicity and responsiveness to cancer vaccine therapy: the state of the art. Semin Immunol 2010; 22:105-12. [PMID: 20226686 PMCID: PMC2884069 DOI: 10.1016/j.smim.2010.02.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2009] [Accepted: 02/15/2010] [Indexed: 12/21/2022]
Abstract
Despite enormous effort, promising pre-clinical data in animal studies and over 900 clinical trials in the United States, no cancer vaccine has ever been approved for clinical use. Over the past decade a great deal of progress has been in both laboratory and clinical studies defining the interactions between developing tumors and the immune system. The results of these studies provide a rationale that may help explain the failure of recent therapeutic cancer vaccines in terms of vaccine principles, in selecting which tumors are the most appropriate to target and instruct the design and implementation of state-of-the-art cancer vaccines.
Collapse
Affiliation(s)
- Taylor H. Schreiber
- Sheila and David Fuente Graduate Program in Cancer Biology, University of Miami Leonard Miller School of Medicine and the Sylvester Comprehensive Cancer Center, Miami FL
| | - Luis Raez
- Department of Medicine, University of Miami Leonard Miller School of Medicine and the Sylvester Comprehensive Cancer Center, Miami FL
| | - Joseph D. Rosenblatt
- Sheila and David Fuente Graduate Program in Cancer Biology, University of Miami Leonard Miller School of Medicine and the Sylvester Comprehensive Cancer Center, Miami FL
- Department of Microbiology and Immunology, University of Miami Leonard Miller School of Medicine and the Sylvester Comprehensive Cancer Center, Miami FL
- Department of Medicine, University of Miami Leonard Miller School of Medicine and the Sylvester Comprehensive Cancer Center, Miami FL
| | - Eckhard R. Podack
- Sheila and David Fuente Graduate Program in Cancer Biology, University of Miami Leonard Miller School of Medicine and the Sylvester Comprehensive Cancer Center, Miami FL
- Department of Microbiology and Immunology, University of Miami Leonard Miller School of Medicine and the Sylvester Comprehensive Cancer Center, Miami FL
- Department of Medicine, University of Miami Leonard Miller School of Medicine and the Sylvester Comprehensive Cancer Center, Miami FL
| |
Collapse
|
31
|
Abstract
The tolerance state that exists between renal cell carcinoma (RCC) and the host's immune system would be an ideal situation in the setting of human kidney transplantation, in which graft tolerance is the ultimate goal of immunosuppressive therapy. On the other hand, acute rejection, as it appears in renal allografts, would be the optimal immunologic situation in patients with RCC. Analysis of the underlying mechanisms of acute allograft rejection and local pro-tumor immunosuppression could help to identify potential therapeutic targets for inducing immune tolerance in allograft recipients and immune rejection in RCC patients. Experimental kidney transplantation might be a suitable model in which to analyze these processes. Macrophages are a prominent and vital cell type in the cellular infiltrate seen in both RCC and renal allografts. Depending on their polarization, they can initiate and promote either proinflammatory or pro-tumor responses, which lead to tissue rejection or acceptance, respectively. Improved understanding of macrophage biology could lead to therapeutic modification of their function in order to promote a desirable immunologic response in either RCC or transplant tissue.
Collapse
|