101
|
Signal transducer and activator of transcription proteins: regulators of myeloid-derived suppressor cell-mediated immunosuppression in cancer. Arch Pharm Res 2016; 39:1597-1608. [DOI: 10.1007/s12272-016-0822-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 08/18/2016] [Indexed: 12/31/2022]
|
102
|
Pyter LM, Husain Y, Calero H, McKim DB, Lin HY, Godbout JP, Sheridan JF, Engeland CG, Marucha PT. Tumors Alter Inflammation and Impair Dermal Wound Healing in Female Mice. PLoS One 2016; 11:e0161537. [PMID: 27548621 PMCID: PMC4993492 DOI: 10.1371/journal.pone.0161537] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 08/08/2016] [Indexed: 12/29/2022] Open
Abstract
Tissue repair is an integral component of cancer treatment (e.g., due to surgery, chemotherapy, radiation). Previous work has emphasized the immunosuppressive effects of tumors on adaptive immunity and has shown that surgery incites cancer metastases. However, the extent to which and how tumors may alter the clinically-relevant innate immune process of wound healing remains an untapped potential area of improvement for treatment, quality of life, and ultimately, mortality of cancer patients. In this study, 3.5 mm full-thickness dermal excisional wounds were placed on the dorsum of immunocompetent female mice with and without non-malignant flank AT-84 murine oral squamous cell carcinomas. Wound closure rate, inflammatory cell number and inflammatory signaling in wounds, and circulating myeloid cell concentrations were compared between tumor-bearing and tumor-free mice. Tumors delayed wound closure, suppressed inflammatory signaling, and altered myeloid cell trafficking in wounds. An in vitro scratch “wounding” assay of adult dermal fibroblasts treated with tumor cell-conditioned media supported the in vivo findings. This study demonstrates that tumors are sufficient to disrupt fundamental and clinically-relevant innate immune functions. The understanding of these underlying mechanisms provides potential for therapeutic interventions capable of improving the treatment of cancer while reducing morbidities and mortality.
Collapse
Affiliation(s)
- Leah M. Pyter
- Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, OH, United States of America
- Department of Psychiatry and Behavioral Health, Ohio State University, Columbus, OH, United States of America
- Department of Neuroscience, Ohio State University, Columbus, OH, United States of America
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, Chicago, IL, United States of America
- * E-mail:
| | - Yasmin Husain
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Humberto Calero
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Daniel B. McKim
- Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, OH, United States of America
| | - Hsin-Yun Lin
- Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, OH, United States of America
- Department of Psychiatry and Behavioral Health, Ohio State University, Columbus, OH, United States of America
| | - Jonathan P. Godbout
- Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, OH, United States of America
- Department of Neuroscience, Ohio State University, Columbus, OH, United States of America
| | - John F. Sheridan
- Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, OH, United States of America
- Deparment of Biosciences, College of Dentistry, Ohio State University, Columbus, OH, United States of America
| | - Christopher G. Engeland
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, Chicago, IL, United States of America
- Department of Biobehavioral Health and College of Nursing, Pennsylvania State University, University Park, PA, United States of America
| | - Phillip T. Marucha
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, Chicago, IL, United States of America
- College of Dentistry, Oregon Health and Sciences University, Portland, OR, United States of America
| |
Collapse
|
103
|
Pyzer AR, Cole L, Rosenblatt J, Avigan DE. Myeloid-derived suppressor cells as effectors of immune suppression in cancer. Int J Cancer 2016; 139:1915-26. [PMID: 27299510 DOI: 10.1002/ijc.30232] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 06/07/2016] [Accepted: 06/08/2016] [Indexed: 12/11/2022]
Abstract
The tumor microenvironment consists of an immunosuppressive niche created by the complex interactions between cancer cells and surrounding stromal cells. A critical component of this environment are myeloid-derived suppressor cells (MDSCs), a heterogeneous group of immature myeloid cells arrested at different stages of differentiation and expanded in response to a variety of tumor factors. MDSCs exert diverse effects in modulating the interactions between immune effector cells and the malignant cells. An increased presence of MDSCs is associated with tumor progression, poorer outcomes, and decreased effectiveness of immunotherapeutic strategies. In this article, we will review our current understanding of the mechanisms that underlie MDSC expansion and their immune-suppressive function. Finally, we review the preclinical studies and clinical trials that have attempted to target MDSCs, in order to improve responses to cancer therapies.
Collapse
Affiliation(s)
- Athalia Rachel Pyzer
- Bone Marrow Transplant, Beth Israel Deaconess Medical Center, Center for Life Sciences, CLS724, Boston, MA
| | - Leandra Cole
- Bone Marrow Transplant, Beth Israel Deaconess Medical Center, Center for Life Sciences, CLS724, Boston, MA
| | - Jacalyn Rosenblatt
- Bone Marrow Transplant, Beth Israel Deaconess Medical Center, Center for Life Sciences, CLS724, Boston, MA
| | - David E Avigan
- Bone Marrow Transplant, Beth Israel Deaconess Medical Center, Center for Life Sciences, CLS724, Boston, MA
| |
Collapse
|
104
|
Keskinov AA, Tapias V, Watkins SC, Ma Y, Shurin MR, Shurin GV. Impact of the Sensory Neurons on Melanoma Growth In Vivo. PLoS One 2016; 11:e0156095. [PMID: 27227315 PMCID: PMC4882065 DOI: 10.1371/journal.pone.0156095] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 05/09/2016] [Indexed: 12/29/2022] Open
Abstract
Nerve endings are often identified within solid tumors, but their impact on the tumor growth and progression remains poorly understood. Emerging data suggests that the central nervous system may affect cancer development and spreading via the hypothalamic-pituitary-adrenal axis and autonomous nervous system. However, the role of the afferent sensory neurons in tumor growth is unclear, except some reports on perineural invasion in prostate and pancreatic cancer and cancer-related pain syndrome. Here, we provide the results of primary testing of the concept that the interaction between melanoma cells and sensory neurons may induce the formation of tumor-supporting microenvironment via attraction of immune regulatory cells by the tumor-activated dorsal root ganglion (DRG) neurons. We report that despite DRG cells not directly up-regulating proliferation of melanoma cells in vitro, presence of DRG neurons allows tumors to grow significantly faster in vivo. This effect has been associated with increased production of chemokines by tumor-activated DRG neurons and attraction of myeloid-derived suppressor cells both in vitro and in vivo. These initial proof-of-concept results justify further investigations of the sensory (afferent) nervous system in the context of tumorigenesis and the local protumorigenic immunoenvironment.
Collapse
Affiliation(s)
- Anton A. Keskinov
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania,United States of America
- * E-mail:
| | - Victor Tapias
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Simon C. Watkins
- Department of Cell Biology and Physiology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States of America
| | - Yang Ma
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania,United States of America
| | - Michael R. Shurin
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania,United States of America
- Department of Immunology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania,United States of America
| | - Galina V. Shurin
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania,United States of America
| |
Collapse
|
105
|
Yin J, Wang C, Huang M, Mao X, Zhou J, Zhang Y. Circulating CD14(+) HLA-DR(-/low) myeloid-derived suppressor cells in leukemia patients with allogeneic hematopoietic stem cell transplantation: novel clinical potential strategies for the prevention and cellular therapy of graft-versus-host disease. Cancer Med 2016; 5:1654-69. [PMID: 27109254 PMCID: PMC4944894 DOI: 10.1002/cam4.688] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 01/27/2016] [Accepted: 02/02/2016] [Indexed: 12/21/2022] Open
Abstract
Myeloid‐derived suppressor cells (MDSCs) are a heterogeneous cell population that includes immature myeloid cells and the progenitor cells of macrophages, dendritic cells (DCs), monocytes, and neutrophils. The expansion and functional importance of MDSCs in patients with cancer and noncancer pathogenic conditions has been recognized. As a result, there has been growing interest in understanding their roles in acute graft‐versus‐host disease (aGVHD) after allogenetic hematopoietic stem cell transplantation (allo‐HSCT). In order to evaluate possible effects of MDSCs on aGVHD development and clinical outcomes, this study systematically detected the dynamic changes of MDSCs accumulation in patients during the first 100 days after allo‐HSCT, and investigated the levels of other cell types and relative cytokines during MDSCs accumulation. Results showed that accumulation of MDSCs in the graft and in peripheral blood when engraftment might contribute to patients' overall immune suppression and result in the successful control of severe aGVHD and long‐term survival without influence on risk of recurrence after allo‐HSCT. But MDSCs levels in the graft had more favorable predictive abilities. Furthermore, MDSCs proportion significantly increased in patients developing aGVHD after allo‐HSCT. It might be caused by secondary inflammatory response, especially related to high concentrations of IL‐6 and TNF‐α. But this accumulation would not be able to counterbalance the aggravation of aGVHD and would not have influence on clinical outcomes and risk of relapse. Overall, MDSCs might be considered as potential new therapeutic option for aGVHD and achieve long‐term immunological tolerance and survival.
Collapse
Affiliation(s)
- Jin Yin
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chunyan Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Min Huang
- Clinical Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xia Mao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jianfeng Zhou
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yicheng Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
106
|
Sasso MS, Lollo G, Pitorre M, Solito S, Pinton L, Valpione S, Bastiat G, Mandruzzato S, Bronte V, Marigo I, Benoit JP. Low dose gemcitabine-loaded lipid nanocapsules target monocytic myeloid-derived suppressor cells and potentiate cancer immunotherapy. Biomaterials 2016; 96:47-62. [PMID: 27135716 DOI: 10.1016/j.biomaterials.2016.04.010] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 04/15/2016] [Accepted: 04/18/2016] [Indexed: 12/21/2022]
Abstract
Tumor-induced expansion of myeloid-derived suppressor cells (MDSCs) is known to impair the efficacy of cancer immunotherapy. Among pharmacological approaches for MDSC modulation, chemotherapy with selected drugs has a considerable interest due to the possibility of a rapid translation to the clinic. However, such approach is poorly selective and may be associated with dose-dependent toxicities. In the present study, we showed that lipid nanocapsules (LNCs) loaded with a lauroyl-modified form of gemcitabine (GemC12) efficiently target the monocytic (M-) MDSC subset. Subcutaneous administration of GemC12-loaded LNCs reduced the percentage of spleen and tumor-infiltrating M-MDSCs in lymphoma and melanoma-bearing mice, with enhanced efficacy when compared to free gemcitabine. Consistently, fluorochrome-labeled LNCs were preferentially uptaken by monocytic cells rather than by other immune cells, in both tumor-bearing mice and human blood samples from healthy donors and melanoma patients. Very low dose administration of GemC12-loaded LNCs attenuated tumor-associated immunosuppression and increased the efficacy of adoptive T cell therapy. Overall, our results show that GemC12-LNCs have monocyte-targeting properties that can be useful for immunomodulatory purposes, and unveil new possibilities for the exploitation of nanoparticulate drug formulations in cancer immunotherapy.
Collapse
Affiliation(s)
- Maria Stella Sasso
- Section of Oncology and Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy
| | - Giovanna Lollo
- LUNAM Université - Micro et Nanomédecines Biomimétiques, F-49933 Angers, France; INSERM U1066, IBS-CHU, 4 Rue Larrey, F-49933 Angers Cedex 9, France
| | - Marion Pitorre
- LUNAM Université - Micro et Nanomédecines Biomimétiques, F-49933 Angers, France; INSERM U1066, IBS-CHU, 4 Rue Larrey, F-49933 Angers Cedex 9, France
| | - Samantha Solito
- Section of Oncology and Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy
| | - Laura Pinton
- Section of Oncology and Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy
| | - Sara Valpione
- Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
| | - Guillaume Bastiat
- LUNAM Université - Micro et Nanomédecines Biomimétiques, F-49933 Angers, France; INSERM U1066, IBS-CHU, 4 Rue Larrey, F-49933 Angers Cedex 9, France
| | - Susanna Mandruzzato
- Section of Oncology and Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy; Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
| | - Vincenzo Bronte
- Immunology Section, Department of Medicine, University of Verona, 37135 Verona, Italy
| | - Ilaria Marigo
- Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy.
| | - Jean-Pierre Benoit
- LUNAM Université - Micro et Nanomédecines Biomimétiques, F-49933 Angers, France; INSERM U1066, IBS-CHU, 4 Rue Larrey, F-49933 Angers Cedex 9, France.
| |
Collapse
|
107
|
Ost M, Singh A, Peschel A, Mehling R, Rieber N, Hartl D. Myeloid-Derived Suppressor Cells in Bacterial Infections. Front Cell Infect Microbiol 2016; 6:37. [PMID: 27066459 PMCID: PMC4814452 DOI: 10.3389/fcimb.2016.00037] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Accepted: 03/15/2016] [Indexed: 01/05/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) comprise monocytic and granulocytic innate immune cells with the capability of suppressing T- and NK-cell responses. While the role of MDSCs has been studied in depth in malignant diseases, the understanding of their regulation and function in infectious disease conditions has just begun to evolve. Here we summarize and discuss the current view how MDSCs participate in bacterial infections and how this knowledge could be exploited for potential future therapeutics.
Collapse
Affiliation(s)
- Michael Ost
- Children's Hospital, University of Tübingen Tübingen, Germany
| | - Anurag Singh
- Children's Hospital, University of Tübingen Tübingen, Germany
| | - Andreas Peschel
- Infection Biology Department, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen Tübingen, Germany
| | - Roman Mehling
- Children's Hospital, University of Tübingen Tübingen, Germany
| | - Nikolaus Rieber
- Children's Hospital, University of TübingenTübingen, Germany; Department of Pediatrics, Kinderklinik München Schwabing, Klinikum Schwabing, StKM GmbH und Klinikum rechts der Isar, Technische Universität MünchenMunich, Germany
| | - Dominik Hartl
- Children's Hospital, University of Tübingen Tübingen, Germany
| |
Collapse
|
108
|
Wu T, Zhao Y, Wang H, Li Y, Shao L, Wang R, Lu J, Yang Z, Wang J, Zhao Y. mTOR masters monocytic myeloid-derived suppressor cells in mice with allografts or tumors. Sci Rep 2016; 6:20250. [PMID: 26833095 PMCID: PMC4735296 DOI: 10.1038/srep20250] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 12/30/2015] [Indexed: 01/06/2023] Open
Abstract
CD11b(+) Gr1(+) myeloid-derived suppressor cells (MDSCs) play critical roles in controlling the processes of tumors, infections, autoimmunity and graft rejection. Immunosuppressive drug rapamycin (RPM), targeting on the key cellular metabolism molecule mTOR, is currently used in clinics to treat patients with allo-grafts, autoimmune diseases and tumors. However, the effect of RPM on MDSCs has not been studied. RPM significantly decreases the cell number and the immunosuppressive ability on T cells of CD11b(+) Ly6C(high) monocytic MDSCs (M-MDSCs) in both allo-grafts-transplanted and tumor-bearing mice respectively. Mice with a myeloid-specific deletion of mTOR have poor M-MDSCs after grafting with allo-skin tissue or a tumor. Grafting of allo-skin or tumors significantly activates glycolysis pathways in myeloid precursor cells in bone marrow, which is inhibited by RPM or mTOR deletion. 2-deoxyglucose (2-DG), an inhibitor of the glycolytic pathway, inhibits M-MDSC differentiation from precursors, while enhancing glycolysis by metformin significantly rescues the RPM-caused deficiency of M-MDSCs. Therefore, we offer evidence supporting that mTOR is an intrinsic factor essential for the differentiation and immunosuppressive function of M-MDSCs and that these metabolism-relevant medicines may impact MDSCs-mediated immunosuppression or immune tolerance induction, which is of considerable clinical importance in treating graft rejection, autoimmune diseases and cancers.
Collapse
Affiliation(s)
- Tingting Wu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yang Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Hao Wang
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, China
| | - Yang Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Lijuan Shao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Department of Oncology, the Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Ruoyu Wang
- Department of Oncology, the Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Jun Lu
- Hepatology and Cancer Biotherapy Ward, Beijing YouAn Hospital, Capital Medical University, Beijing, China
| | - Zhongzhou Yang
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Junjie Wang
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, China
| | - Yong Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
109
|
Mandruzzato S, Brandau S, Britten CM, Bronte V, Damuzzo V, Gouttefangeas C, Maurer D, Ottensmeier C, van der Burg SH, Welters MJP, Walter S. Toward harmonized phenotyping of human myeloid-derived suppressor cells by flow cytometry: results from an interim study. Cancer Immunol Immunother 2016; 65:161-9. [PMID: 26728481 PMCID: PMC4726716 DOI: 10.1007/s00262-015-1782-5] [Citation(s) in RCA: 171] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 12/12/2015] [Indexed: 01/02/2023]
Abstract
There is an increasing interest for monitoring circulating myeloid-derived suppressor cells (MDSCs) in cancer patients, but there are also divergences in their phenotypic definition. To overcome this obstacle, the Cancer Immunoguiding Program under the umbrella of the Association of Cancer Immunotherapy is coordinating a proficiency panel program that aims at harmonizing MDSC phenotyping. After a consultation period, a two-stage approach was designed to harmonize MDSC phenotype. In the first step, an international consortium of 23 laboratories immunophenotyped 10 putative MDSC subsets on pretested, peripheral blood mononuclear cells of healthy donors to assess the level of concordance and define robust marker combinations for the identification of circulating MDSCs. At this stage, no mandatory requirements to standardize reagents or protocols were introduced. Data analysis revealed a small intra-laboratory, but very high inter-laboratory variance for all MDSC subsets, especially for the granulocytic subsets. In particular, the use of a dead-cell marker altered significantly the reported percentage of granulocytic MDSCs, confirming that these cells are especially sensitive to cryopreservation and/or thawing. Importantly, the gating strategy was heterogeneous and associated with high inter-center variance. Overall, our results document the high variability in MDSC phenotyping in the multicenter setting if no harmonization/standardization measures are applied. Although the observed variability depended on a number of identified parameters, the main parameter associated with variation was the gating strategy. Based on these findings, we propose further efforts to harmonize marker combinations and gating parameters to identify strategies for a robust enumeration of MDSC subsets.
Collapse
Affiliation(s)
- Susanna Mandruzzato
- Section of Oncology and Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Gattamelata, 64, 35128, Padua, Italy.
- Veneto Institute of Oncology IOV - IRCCS, Padua, Italy.
| | - Sven Brandau
- Department of Otorhinolaryngology, University Hospital Essen, Essen, Germany
| | - Cedrik M Britten
- TRON Translationale Onkologie an der Universitätsmedizin der Johannes Gutenberg-Universität Mainz GmbH, Mainz, Germany
- Cell Therapy Group, Immuno-Oncology and Combinations, GlaxoSmithKline, Stevenage, UK
| | - Vincenzo Bronte
- Section of Immunology, Department of Pathology and Diagnostics, Verona University Hospital, Verona, Italy
| | - Vera Damuzzo
- Section of Oncology and Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Gattamelata, 64, 35128, Padua, Italy
- Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Cécile Gouttefangeas
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany
| | | | - Christian Ottensmeier
- Cancer Sciences Unit, Faculty of Medicine, Experimental Cancer Medicine Centre, Southampton General Hospital, University of Southampton, Tremona Road, Southampton, UK
| | - Sjoerd H van der Burg
- Department of Clinical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marij J P Welters
- Department of Clinical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
110
|
Gato M, Blanco-Luquin I, Zudaire M, de Morentin XM, Perez-Valderrama E, Zabaleta A, Kochan G, Escors D, Fernandez-Irigoyen J, Santamaría E. Drafting the proteome landscape of myeloid-derived suppressor cells. Proteomics 2015; 16:367-78. [PMID: 26403437 DOI: 10.1002/pmic.201500229] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 08/18/2015] [Accepted: 09/21/2015] [Indexed: 01/12/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of cells that are defined by their myeloid origin, immature state, and ability to potently suppress T-cell responses. They regulate immune responses and the population significantly increases in the tumor microenvironment of patients with glioma and other malignant tumors. For their study, MDSCs are usually isolated from the spleen or directly of tumors from a large number of tumor-bearing mice although promising ex vivo differentiated MDSC production systems have been recently developed. During the last years, proteomics has emerged as a powerful approach to analyze MDSCs proteomes using shotgun-based mass spectrometry (MS), providing functional information about cellular homeostasis and metabolic state at a global level. Here, we will revise recent proteome profiling studies performed in MDSCs from different origins. Moreover, we will perform an integrative functional analysis of the protein compilation derived from these large-scale proteomic studies in order to obtain a comprehensive view of MDSCs biology. Finally, we will also discuss the potential application of high-throughput proteomic approaches to study global proteome dynamics and post-translational modifications (PTMs) during the differentiation process of MDSCs that will greatly boost the identification of novel MDSC-specific therapeutic targets to apply in cancer immunotherapy.
Collapse
Affiliation(s)
- María Gato
- Immunomodulation Laboratory, Navarrabiomed, Fundación Miguel Servet, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Idoia Blanco-Luquin
- Immunomodulation Laboratory, Navarrabiomed, Fundación Miguel Servet, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Maribel Zudaire
- Immunomodulation Laboratory, Navarrabiomed, Fundación Miguel Servet, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Xabier Martínez de Morentin
- Proteomics Unit, Navarrabiomed, Fundación Miguel Servet, ProteoRed-ISCIII, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Estela Perez-Valderrama
- Proteomics Unit, Navarrabiomed, Fundación Miguel Servet, ProteoRed-ISCIII, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Aintzane Zabaleta
- Biofunctional Nanomaterials Laboratory, CIC Biomagune, San Sebastian, Spain
| | - Grazyna Kochan
- Immunomodulation Laboratory, Navarrabiomed, Fundación Miguel Servet, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - David Escors
- Immunomodulation Laboratory, Navarrabiomed, Fundación Miguel Servet, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Joaquín Fernandez-Irigoyen
- Proteomics Unit, Navarrabiomed, Fundación Miguel Servet, ProteoRed-ISCIII, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Enrique Santamaría
- Proteomics Unit, Navarrabiomed, Fundación Miguel Servet, ProteoRed-ISCIII, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| |
Collapse
|
111
|
Zheng R, Chen S, Chen S. Correlation between myeloid-derived suppressor cells and S100A8/A9 in tumor and autoimmune diseases. Int Immunopharmacol 2015; 29:919-925. [PMID: 26508452 DOI: 10.1016/j.intimp.2015.10.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 10/11/2015] [Accepted: 10/12/2015] [Indexed: 02/05/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of cells that constitute an important component of immune regulatory system. Two calcium-binding proteins S100A8 and S100A9 act as important mediators in acute and chronic inflammation. In recent years, many researchers have found that MDSCs and S100A8/A9 operated with one another through a positive feedback loop to promote tumor development and metastasis. However, the correlation between MDSCs and S100A8/A9 in autoimmune diseases (AIDs) remains unknown. In this review, we discussed the co-operation of MDSCs and S100A8/A9 in tumor environment, and also, the role of these two components in AIDs.
Collapse
Affiliation(s)
- Ruoting Zheng
- Department of Endocrinology and Rheumatology, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515000, China
| | - Shiyi Chen
- Department of Endocrinology and Rheumatology, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515000, China
| | - Shenren Chen
- Department of Endocrinology and Rheumatology, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515000, China.
| |
Collapse
|
112
|
The immunobiology of myeloid-derived suppressor cells in cancer. Tumour Biol 2015; 37:1387-406. [PMID: 26611648 DOI: 10.1007/s13277-015-4477-9] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 11/19/2015] [Indexed: 12/31/2022] Open
Abstract
The tumor microenvironment is a complex and heterogeneous milieu in which multiple interactions occur between tumor and host cells. Immunosuppressive cells which are present in this microenvironment, such as regulatory T (Treg) cells and myeloid-derived suppressor cells (MDSCs), play an important role in tumor progression, via down-regulation of antitumor responses. MDSCs represent a heterogeneous group of cells originated from the myeloid lineage that are in the immature state. These cells markedly accumulate under pathologic conditions, such as cancer, infection, and inflammation, and use various mechanisms to inhibit both adaptive and innate immune responses. These immunosuppressive mechanisms include deprivation of T cells from essential amino acids, induction of oxidative stress, interference with viability and trafficking of T cells, induction of immunosuppressive cells, and finally polarizing immunity toward a tumor-promoting type 2 phenotype. In addition to suppression of antitumor immune responses, MDSCs can also enhance the tumor metastasis and angiogenesis. Previous studies have shown that increased frequency of MDSCs is related to the tumor progression. Moreover, various drugs that directly target these cells or reverse their suppressive activity can improve antitumor immune responses as well as increase the efficacy of immunotherapeutic intervention. In this review, we will first discuss on the immunobiology of MDSCs in an attempt to find the role of these cells in tumor progression and then discuss about therapeutic approaches to target these cells.
Collapse
|
113
|
Stanojevic I, Miller K, Kandolf-Sekulovic L, Mijuskovic Z, Zolotarevski L, Jovic M, Gacevic M, Djukic M, Arsenijevic N, Vojvodic D. A subpopulation that may correspond to granulocytic myeloid-derived suppressor cells reflects the clinical stage and progression of cutaneous melanoma. Int Immunol 2015; 28:87-97. [PMID: 26391013 DOI: 10.1093/intimm/dxv053] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 09/11/2015] [Indexed: 12/12/2022] Open
Abstract
Seventy-eight melanoma patients and 10 healthy individuals were examined. Follow-up examinations of all melanoma patients were performed regularly every three months. Myeloid-derived suppressor cells (MDSC) were defined as lineage negative (CD3(-), CD19(-), CD56(-)), HLA-DR(-/low), CD11b(+) and CD33(+). Classification of granulocytic (GrMDSC) and monocytic (MoMDSC) subsets was based on the CD15 and CD14 expression, respectively. Unlike the MoMDSC, that were present in 60% of healthy controls and 15% of melanoma patients, the GrMDSC were present in all examined participants, and the melanoma patients were found to have statistically higher frequencies compared with healthy controls. Accordingly, we kept focused on GrMDSC frequencies in relation to the melanoma stages and course of the disease. The GrMDSC values are highest in stage IV melanoma patients, with statistical significance compared with stages IA, IB, IIA and IIB. Patients with progression had statistically higher GrMDSC counts comparing with those with stable disease (P = 0.0079). Patients who had progression-free interval (PFI) < 12 months showed significantly higher GrMDSC values compared with those with PFI > 12 months (P = 0.0333). GrMDSC showed significant negative correlation with PFI intervals (P = 0.0095). The GrMDSC subset was predominant in all our patients. We confirmed that GrMDSC do accumulate early in the peripheral blood of melanoma patients and their frequencies correlate narrowly with the clinical stage and the spread of the disease. The increase in GrMDSC frequencies correlates well with a progressive disease and could be considered a potential predictive biomarker of high-risk melanoma cases that are more likely to have a shorter PFI.
Collapse
Affiliation(s)
- Ivan Stanojevic
- Department of Clinical and Experimental Immunology, Institute for Medical Research, Military Medical Academy, Crnotravska 17, 11000 Belgrade, Serbia
| | - Karolina Miller
- Department of Histopathology, Dorset County Hospital NHS Foundation Trust, DT1 2JY Dorchester, UK
| | | | - Zeljko Mijuskovic
- Department of Dermatovenerology, Military Medical Academy, 11000 Belgrade, Serbia
| | | | - Milena Jovic
- Institute for Pathology, Military Medical Academy, 11000 Belgrade, Serbia
| | - Milomir Gacevic
- Clinic for Plastic and Reconstructive Surgery, Military Medical Academy, 11000 Belgrade, Serbia
| | - Mirjana Djukic
- Department of Toxicology, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia
| | | | - Danilo Vojvodic
- Department of Clinical and Experimental Immunology, Institute for Medical Research, Military Medical Academy, Crnotravska 17, 11000 Belgrade, Serbia
| |
Collapse
|
114
|
Wang D, DuBois RN. Immunosuppression associated with chronic inflammation in the tumor microenvironment. Carcinogenesis 2015; 36:1085-93. [PMID: 26354776 DOI: 10.1093/carcin/bgv123] [Citation(s) in RCA: 270] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Accepted: 08/16/2015] [Indexed: 02/06/2023] Open
Abstract
Chronic inflammation contributes to cancer development via multiple mechanisms. One potential mechanism is that chronic inflammation can generate an immunosuppressive microenvironment that allows advantages for tumor formation and progression. The immunosuppressive environment in certain chronic inflammatory diseases and solid cancers is characterized by accumulation of proinflammatory mediators, infiltration of immune suppressor cells and activation of immune checkpoint pathways in effector T cells. In this review, we highlight recent advances in our understanding of how immunosuppression contributes to cancer and how proinflammatory mediators induce the immunosuppressive microenvironment via induction of immunosuppressive cells and activation of immune checkpoint pathways.
Collapse
Affiliation(s)
- Dingzhi Wang
- Laboratory for Inflammation and Cancer, The Biodesign Institute and
| | - Raymond N DuBois
- Laboratory for Inflammation and Cancer, The Biodesign Institute and Department of Chemistry and Biochemistry, Arizona State University, PO Box 875001, 1001 S. McAllister Ave., Tempe, AZ 85287, USA and Department of Research and Division of Gastroenterology, Mayo Clinic, Scottsdale, AZ 85259, USA
| |
Collapse
|
115
|
Ugel S, De Sanctis F, Mandruzzato S, Bronte V. Tumor-induced myeloid deviation: when myeloid-derived suppressor cells meet tumor-associated macrophages. J Clin Invest 2015; 125:3365-76. [PMID: 26325033 DOI: 10.1172/jci80006] [Citation(s) in RCA: 441] [Impact Index Per Article: 44.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The generation of an inflammatory environment is favorable and often decisive for the growth of both primary tumors and metastases. Tumor cells either express membrane molecules or release tumor-derived soluble factors able to alter myelopoiesis. Tumor-reprogrammed myeloid cells not only create a tolerogenic environment by blocking T cell functions and proliferation, but also directly drive tumor growth by promoting cancer stemness, angiogenesis, stroma deposition, epithelial-to-mesenchymal transition, and metastasis formation. In this Review, we discuss the interplay between immunosuppressive and protumoral myeloid cells and detail their immune-regulatory mechanisms, the molecular pathways involved in their differentiation, as well as their potential role as prognostic and diagnostic biomarkers and prospective targets for innovative approaches to treat tumor-bearing hosts.
Collapse
|
116
|
Mei S, Xin J, Liu Y, Zhang Y, Liang X, Su X, Yan H, Huang Y, Yang R. MicroRNA-200c Promotes Suppressive Potential of Myeloid-Derived Suppressor Cells by Modulating PTEN and FOG2 Expression. PLoS One 2015; 10:e0135867. [PMID: 26285119 PMCID: PMC4540422 DOI: 10.1371/journal.pone.0135867] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 07/27/2015] [Indexed: 01/05/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) constitute one of the major populations that potently suppress anti-tumor immune responses and favor tumor growth in tumor microenvironment. However, the mechanism(s) regulating the differentiation and suppressive function of tumor-associated MDSCs remain(s) unclear. Here, we identified a microRNA-200c (miR-200c), whose expression was dramatically induced by tumor-derived factors. Meanwhile, we also demonstrated that GM-CSF was a main inducer of miR-200c in tumor environment, and miR-200c in turn promoted the expansion and immune suppressive activity of MDSCs via targeting phosphatase and tensin homolog (PTEN) and friend of Gata 2 (FOG2), which can lead to STAT3 and PI3K/Akt activation. Finally, we examined in vivo suppressive function of miR-200c transfected MDSCs and found that miR-200c could remarkably promote tumor growth via modifying MDSCs. Thus, GM-CSF induced miR-200c in tumor environment plays a critical role in governing the expansion and functions of tumor-associated MDSCs and serves as a potential target in immunotherapy against tumor.
Collapse
Affiliation(s)
- Shiyue Mei
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, P. R. China
| | - Jiaxuan Xin
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, P. R. China
| | - Yu Liu
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, P. R. China
| | - Yuan Zhang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, P. R. China
| | - Xue Liang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, P. R. China
| | - Xiaomin Su
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, P. R. China
| | - Hui Yan
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, P. R. China
| | - Yugang Huang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, P. R. China
| | - Rongcun Yang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, P. R. China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, P. R. China
- Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, P. R. China
- * E-mail:
| |
Collapse
|
117
|
De Sanctis F, Solito S, Ugel S, Molon B, Bronte V, Marigo I. MDSCs in cancer: Conceiving new prognostic and therapeutic targets. Biochim Biophys Acta Rev Cancer 2015; 1865:35-48. [PMID: 26255541 DOI: 10.1016/j.bbcan.2015.08.001] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 07/31/2015] [Accepted: 08/01/2015] [Indexed: 12/30/2022]
Abstract
The incomplete clinical efficacy of anti-tumor immunotherapy can depend on the presence of an immunosuppressive environment in the host that supports tumor progression. Tumor-derived cytokines and growth factors induce an altered hematopoiesis that modifies the myeloid cell differentiation process, promoting proliferation and expansion of cells with immunosuppressive skills, namely myeloid derived suppressor cells (MDSCs). MDSCs promote tumor growth not only by shaping immune responses towards tumor tolerance, but also by supporting several processes necessary for the neoplastic progression such as tumor angiogenesis, cancer stemness, and metastasis dissemination. Thus, MDSC targeting represents a promising tool to eliminate host immune dysfunctions and increase the efficacy of immune-based cancer therapies.
Collapse
Affiliation(s)
- Francesco De Sanctis
- Immunology Section, Department of Pathology and Diagnostics, University of Verona, 37134 Verona, Italy
| | - Samantha Solito
- Section of Oncology and Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy
| | - Stefano Ugel
- Immunology Section, Department of Pathology and Diagnostics, University of Verona, 37134 Verona, Italy
| | - Barbara Molon
- Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
| | - Vincenzo Bronte
- Immunology Section, Department of Pathology and Diagnostics, University of Verona, 37134 Verona, Italy.
| | - Ilaria Marigo
- Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
| |
Collapse
|
118
|
Bodogai M, Moritoh K, Lee-Chang C, Hollander CM, Sherman-Baust CA, Wersto RP, Araki Y, Miyoshi I, Yang L, Trinchieri G, Biragyn A. Immunosuppressive and Prometastatic Functions of Myeloid-Derived Suppressive Cells Rely upon Education from Tumor-Associated B Cells. Cancer Res 2015; 75:3456-65. [PMID: 26183924 DOI: 10.1158/0008-5472.can-14-3077] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 06/13/2015] [Indexed: 12/30/2022]
Abstract
Myeloid-derived suppressive cells (MDSC) have been reported to promote metastasis, but the loss of cancer-induced B cells/B regulatory cells (tBreg) can block metastasis despite MDSC expansion in cancer. Here, using multiple murine tumor models and human MDSC, we show that MDSC populations that expand in cancer have only partially primed regulatory function and limited prometastatic activity unless they are fully educated by tBregs. Cancer-induced tBregs directly activate the regulatory function of both the monocyte and granulocyte subpopulations of MDSC, relying, in part, on TgfβR1/TgfβR2 signaling. MDSC fully educated in this manner exhibit an increased production of reactive oxygen species and NO and more efficiently suppress CD4(+) and CD8(+) T cells, thereby promoting tumor growth and metastasis. Thus, loss of tBregs or TgfβR deficiency in MDSC is sufficient to disable their suppressive function and to block metastasis. Overall, our data indicate that cancer-induced B cells/B regulatory cells are important regulators of the immunosuppressive and prometastatic functions of MDSC.
Collapse
Affiliation(s)
- Monica Bodogai
- Immune Regulation Section, Laboratory of Molecular Biology and Immunology, National Institute on Aging, Baltimore, Maryland
| | - Kanako Moritoh
- Immune Regulation Section, Laboratory of Molecular Biology and Immunology, National Institute on Aging, Baltimore, Maryland
| | - Catalina Lee-Chang
- Immune Regulation Section, Laboratory of Molecular Biology and Immunology, National Institute on Aging, Baltimore, Maryland
| | - Christine M Hollander
- Tumor Microenvironment Section, Laboratory of Cancer Biology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Cheryl A Sherman-Baust
- Immune Regulation Section, Laboratory of Molecular Biology and Immunology, National Institute on Aging, Baltimore, Maryland
| | - Robert P Wersto
- Flow Cytometry Unit, National Institute on Aging, Baltimore, Maryland
| | - Yoshihiko Araki
- Juntendo University Graduate School of Medicine, Chiba, Japan
| | - Ichiro Miyoshi
- Center for Experimental Animal Science, Nagoya City University Graduate School of Medicine, Nagoya, Japan
| | - Li Yang
- Tumor Microenvironment Section, Laboratory of Cancer Biology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Giorgio Trinchieri
- Cancer Immunobiology Section, Laboratory of Experimental Immunology, National Cancer Institute, Frederick, Maryland
| | - Arya Biragyn
- Immune Regulation Section, Laboratory of Molecular Biology and Immunology, National Institute on Aging, Baltimore, Maryland.
| |
Collapse
|
119
|
Myeloid-Derived Suppressor Cells and Therapeutic Strategies in Cancer. Mediators Inflamm 2015; 2015:159269. [PMID: 26078490 PMCID: PMC4452485 DOI: 10.1155/2015/159269] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 03/23/2015] [Indexed: 01/04/2023] Open
Abstract
Development of solid cancer depends on escape from host immunosurveillance. Various types of immune cells contribute to tumor-induced immune suppression, including tumor associated macrophages, regulatory T cells, type 2 NKT cells, and myeloid-derived suppressor cells (MDSCs). Growing body of evidences shows that MDSCs play pivotal roles among these immunosuppressive cells in multiple steps of cancer progression. MDSCs are immature myeloid cells that arise from myeloid progenitor cells and comprise a heterogeneous immune cell population. MDSCs are characterized by the ability to suppress both adaptive and innate immunities mainly through direct inhibition of the cytotoxic functions of T cells and NK cells. In clinical settings, the number of circulating MDSCs is associated with clinical stages and response to treatment in several cancers. Moreover, MDSCs are reported to contribute to chemoresistant phenotype. Collectively, targeting MDSCs could potentially provide a rationale for novel treatment strategies in cancer. This review summarizes recent understandings of MDSCs in cancer and discusses promissing clinical approaches in cancer patients.
Collapse
|
120
|
Increase in Both CD14-Positive and CD15-Positive Myeloid-Derived Suppressor Cell Subpopulations in the Blood of Patients With Glioma But Predominance of CD15-Positive Myeloid-Derived Suppressor Cells in Glioma Tissue. J Neuropathol Exp Neurol 2015; 74:390-400. [DOI: 10.1097/nen.0000000000000183] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
|
121
|
Papaspyridonos M, Matei I, Huang Y, do Rosario Andre M, Brazier-Mitouart H, Waite JC, Chan AS, Kalter J, Ramos I, Wu Q, Williams C, Wolchok JD, Chapman PB, Peinado H, Anandasabapathy N, Ocean AJ, Kaplan RN, Greenfield JP, Bromberg J, Skokos D, Lyden D. Id1 suppresses anti-tumour immune responses and promotes tumour progression by impairing myeloid cell maturation. Nat Commun 2015; 6:6840. [PMID: 25924227 PMCID: PMC4423225 DOI: 10.1038/ncomms7840] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Accepted: 03/04/2015] [Indexed: 12/15/2022] Open
Abstract
A central mechanism of tumour progression and metastasis involves the generation of an immunosuppressive ‘macroenvironment' mediated in part through tumour-secreted factors. Here we demonstrate that upregulation of the Inhibitor of Differentiation 1 (Id1), in response to tumour-derived factors, such as TGFβ, is responsible for the switch from dendritic cell (DC) differentiation to myeloid-derived suppressor cell expansion during tumour progression. Genetic inactivation of Id1 largely corrects the myeloid imbalance, whereas Id1 overexpression in the absence of tumour-derived factors re-creates it. Id1 overexpression leads to systemic immunosuppression by downregulation of key molecules involved in DC differentiation and suppression of CD8 T-cell proliferation, thus promoting primary tumour growth and metastatic progression. Furthermore, advanced melanoma patients have increased plasma TGFβ levels and express higher levels of ID1 in myeloid peripheral blood cells. This study reveals a critical role for Id1 in suppressing the anti-tumour immune response during tumour progression and metastasis. Tumour progression is promoted by the generation of an immunosuppressive macroenvironment. Here, the authors demonstrate that the Inhibitor of Differentiation 1 promotes the switch from dendritic cell differentiation towards myeloid-derived suppressor cell expansion during tumour progression.
Collapse
Affiliation(s)
- Marianna Papaspyridonos
- Children's Cancer and Blood Foundation Laboratories and Departments of Pediatrics and Cell and Developmental Biology, Weill Cornell Medical College, 413 East 69th Street, New York City, New York 10021, USA
| | - Irina Matei
- Children's Cancer and Blood Foundation Laboratories and Departments of Pediatrics and Cell and Developmental Biology, Weill Cornell Medical College, 413 East 69th Street, New York City, New York 10021, USA
| | - Yujie Huang
- 1] Children's Cancer and Blood Foundation Laboratories and Departments of Pediatrics and Cell and Developmental Biology, Weill Cornell Medical College, 413 East 69th Street, New York City, New York 10021, USA [2] Department of Neurosurgery, Weill Cornell Medical College, 1300 York Avenue, New York City, New York 10065, USA
| | - Maria do Rosario Andre
- 1] Children's Cancer and Blood Foundation Laboratories and Departments of Pediatrics and Cell and Developmental Biology, Weill Cornell Medical College, 413 East 69th Street, New York City, New York 10021, USA [2] Department of Genetics, Oncology and Human Toxicology, Faculdade de Ciência Médicas, Universidade Nova de Lisboa, Rua da Junqueira 100, 1349-008 Lisbon, Portugal
| | - Helene Brazier-Mitouart
- Children's Cancer and Blood Foundation Laboratories and Departments of Pediatrics and Cell and Developmental Biology, Weill Cornell Medical College, 413 East 69th Street, New York City, New York 10021, USA
| | | | - April S Chan
- Children's Cancer and Blood Foundation Laboratories and Departments of Pediatrics and Cell and Developmental Biology, Weill Cornell Medical College, 413 East 69th Street, New York City, New York 10021, USA
| | - Julie Kalter
- Regeneron Pharmaceuticals, Tarrytown, New York 10591, USA
| | - Ilyssa Ramos
- Regeneron Pharmaceuticals, Tarrytown, New York 10591, USA
| | - Qi Wu
- Regeneron Pharmaceuticals, Tarrytown, New York 10591, USA
| | - Caitlin Williams
- Children's Cancer and Blood Foundation Laboratories and Departments of Pediatrics and Cell and Developmental Biology, Weill Cornell Medical College, 413 East 69th Street, New York City, New York 10021, USA
| | - Jedd D Wolchok
- 1] Melanoma and Immunotherapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York City, New York 10065, USA [2] Ludwig Center for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York City, New York 10065, USA
| | - Paul B Chapman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York City, New York 10065, USA
| | - Hector Peinado
- 1] Children's Cancer and Blood Foundation Laboratories and Departments of Pediatrics and Cell and Developmental Biology, Weill Cornell Medical College, 413 East 69th Street, New York City, New York 10021, USA [2] Tumor Metastasis Laboratory, Fundación Centro Nacional de Investigaciones Oncológicas, Calle Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Niroshana Anandasabapathy
- Brigham and Women's Hospital, Department of Dermatology, Harvard Medical School, 221 Longwood Avenue EBRC, Room 513, Boston, Massachusetts 02118, USA
| | - Allyson J Ocean
- Department of Medicine, Weill Cornell Medical College and Medical Oncology/Solid Tumor Program, 1305 York Avenue, New York City, New York 10021, USA
| | - Rosandra N Kaplan
- Center for Cancer Research, Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Building 10-Hatfield CRC, Room 1-3940, Bethesda, Maryland 20892, USA
| | - Jeffrey P Greenfield
- Department of Neurosurgery, Weill Cornell Medical College, 1300 York Avenue, New York City, New York 10065, USA
| | - Jacqueline Bromberg
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York City, New York 10065, USA
| | | | - David Lyden
- 1] Children's Cancer and Blood Foundation Laboratories and Departments of Pediatrics and Cell and Developmental Biology, Weill Cornell Medical College, 413 East 69th Street, New York City, New York 10021, USA [2] Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York City, New York 10065, USA
| |
Collapse
|
122
|
Pan W, Sun Q, Wang Y, Wang J, Cao S, Ren X. Highlights on mechanisms of drugs targeting MDSCs: providing a novel perspective on cancer treatment. Tumour Biol 2015; 36:3159-69. [DOI: 10.1007/s13277-015-3363-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 03/19/2015] [Indexed: 12/22/2022] Open
|
123
|
Höchst B, Mikulec J, Baccega T, Metzger C, Welz M, Peusquens J, Tacke F, Knolle P, Kurts C, Diehl L, Ludwig-Portugall I. Differential induction of Ly6G and Ly6C positive myeloid derived suppressor cells in chronic kidney and liver inflammation and fibrosis. PLoS One 2015; 10:e0119662. [PMID: 25738302 PMCID: PMC4349817 DOI: 10.1371/journal.pone.0119662] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 01/28/2015] [Indexed: 12/20/2022] Open
Abstract
CD11b+Gr1+ myeloid derived suppressor cells (MDSC) are known to be very potent suppressors of T cell immunity and can be further stratified into granulocytic MDSC and monocytic MDSC in mice based on expression of Ly6G or Ly6C, respectively. Here, using these markers and functional assays, we aimed to identify whether MDSC are induced during chronic inflammation leading to fibrosis in both kidney and liver and whether additional markers could more specifically identify these MDSC subsets. In an adenine-induced model of kidney inflammation/fibrosis suppressive Ly6Gpos MDSC were induced. The suppressive function within the Ly6G+ MDSC population was exclusively present in IFNγRβ expressing cells. In contrast, in chronic inflammation in the liver induced by bile duct ligation, suppressive capacity was exclusively present in the Ly6Cpos MDSC subset. Gene expression analyses confirmed the differential origins and regulation of those MDSC subsets. Additionally, depletion of MDSC in either kidney or liver fibrosis enhanced fibrosis markers, indicating a protective role for MDSC in organ fibrosis. Thus, our data demonstrate that during liver inflammation and kidney fibrosis MDSC with similar function arise bearing a distinct marker profile and arising from different cell populations.
Collapse
Affiliation(s)
- Bastian Höchst
- Institute for Molecular Medicine, University of Bonn, Bonn, Germany
- Institute of Molecular Immunology, Technische Universität München, Munich, Germany
| | - Julita Mikulec
- Institute for Molecular Medicine, University of Bonn, Bonn, Germany
| | - Tania Baccega
- Institute for Experimental Immunology, University of Bonn, Bonn, Germany
| | | | - Meike Welz
- Institute for Molecular Medicine, University of Bonn, Bonn, Germany
| | - Julia Peusquens
- Department of Medicine III, University Hospital Aachen, Aachen, Germany
| | - Frank Tacke
- Department of Medicine III, University Hospital Aachen, Aachen, Germany
| | - Percy Knolle
- Institute for Molecular Medicine, University of Bonn, Bonn, Germany
- Institute of Molecular Immunology, Technische Universität München, Munich, Germany
| | - Christian Kurts
- Institute for Experimental Immunology, University of Bonn, Bonn, Germany
| | - Linda Diehl
- Institute for Molecular Medicine, University of Bonn, Bonn, Germany
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- * E-mail: (LD); (ILP)
| | - Isis Ludwig-Portugall
- Institute for Experimental Immunology, University of Bonn, Bonn, Germany
- * E-mail: (LD); (ILP)
| |
Collapse
|
124
|
Damuzzo V, Pinton L, Desantis G, Solito S, Marigo I, Bronte V, Mandruzzato S. Complexity and challenges in defining myeloid-derived suppressor cells. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2015; 88:77-91. [PMID: 25504825 PMCID: PMC4405078 DOI: 10.1002/cyto.b.21206] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 11/14/2014] [Accepted: 11/18/2014] [Indexed: 12/23/2022]
Abstract
Study of myeloid cells endowed with suppressive activity is an active field of research which has particular importance in cancer, in view of the negative regulatory capacity of these cells to the host's immune response. The expansion of these cells, called myeloid-derived suppressor cells (MDSCs), has been documented in many models of tumor-bearing mice and in patients with tumors of various origin, and their presence is associated with disease progression and reduced survival. For this reason, monitoring this type of cell expansion is of clinical importance, and flow cytometry is the technique of choice for their identification. Over the years, it has been demonstrated that MDSCs comprise a group of immature myeloid cells belonging both to monocytic and granulocytic lineages, with several stages of differentiation; their occurrence depends on tumor-derived soluble factors, which guide their expansion and determine their block of differentiation. Because of their heterogeneous composition, accurate phenotyping of these cells requires a multicolor approach, so that the expansion of all MDSC subsets can be appreciated. This review article focuses on identifying MDSCs and discusses problems associated with phenotyping circulating and tumor-associated MDSCs in humans and in mouse models.
Collapse
Affiliation(s)
- Vera Damuzzo
- Section of Oncology and Immunology, Department of Surgery, Oncology and Gastroenterology, University of PadovaPadova, Italy
| | - Laura Pinton
- Section of Oncology and Immunology, Department of Surgery, Oncology and Gastroenterology, University of PadovaPadova, Italy
| | | | - Samantha Solito
- Section of Oncology and Immunology, Department of Surgery, Oncology and Gastroenterology, University of PadovaPadova, Italy
| | | | - Vincenzo Bronte
- Section of Immunology, Department of Pathology, Verona University HospitalVerona, Italy
| | - Susanna Mandruzzato
- Section of Oncology and Immunology, Department of Surgery, Oncology and Gastroenterology, University of PadovaPadova, Italy
- Veneto Institute of Oncology IOV—IRCCSPadova, Italy
| |
Collapse
|
125
|
Ning G, She L, Lu L, Liu Y, Zeng Y, Yan Y, Lin C. Analysis of monocytic and granulocytic myeloid-derived suppressor cells subsets in patients with hepatitis C virus infection and their clinical significance. BIOMED RESEARCH INTERNATIONAL 2015; 2015:385378. [PMID: 25815313 PMCID: PMC4359884 DOI: 10.1155/2015/385378] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 02/06/2015] [Accepted: 02/14/2015] [Indexed: 01/08/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) have been shown to inhibit T-cell responses in many diseases, but, in hepatitis C virus (HCV) infected patients, MDSCs are still poorly studied. In this assay, we investigated the phenotype and frequency of two new populations of MDSCs denoted as monocytic and granulocytic MDSCs (M-MDSCs and G-MDSCs) in HCV infected patients and analyzed their clinical significance in these patients respectively. We found that the frequency of CD14(+)HLA-DR(-/low) cells (M-MDSCs) from HCV infected patients (mean ± SE, 3.134% ± 0.340%) was significantly increased when compared to healthy controls (mean ± SE, 1.764% ± 0.461%) (Z = -2.438, P = 0.015), while there was no statistical difference between the frequency of HLA-DR(-/low)CD33(+)CD11b(+)CD15(+) (G-MDSCs) of HCV infected patients and healthy donors (0.201% ± 0.038% versus 0.096% ± 0.026%, P > 0.05), which suggested that HCV infection could cause the proliferation of M-MDSCs instead of G-MDSCs. Besides, we found that the frequency of M-MDSCs in HCV infected patients had certain relevance with age (r = 0.358, P = 0.003); patients older than 40 years old group (mean ± SE, 3.673% ± 0.456%) had a significantly higher frequency of M-MDSCs than that of age less than 40 years old group (mean ± SE, 2.363% ± 0.482%) (Z = -2.685, P = 0.007). The frequency of M-MDSCs, however, had no correlation with HCV RNA loads, aspartate aminotransferase (AST), alanine aminotransferase (ALT), and the level of liver inflammation degree.
Collapse
Affiliation(s)
- Gang Ning
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| | - Lanhui She
- Intranet of Guangzhou Woman and Children's Medical Center, Guangzhou 510630, China
| | - Lirong Lu
- Department of Infectious Diseases, First Affiliated Hospital of Kunming Medical University, Yunnan, Kunming 650032, China
| | - Ying Liu
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| | - Yingfu Zeng
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| | - Ying Yan
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| | - Chaoshuang Lin
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| |
Collapse
|
126
|
Draghiciu O, Lubbers J, Nijman HW, Daemen T. Myeloid derived suppressor cells-An overview of combat strategies to increase immunotherapy efficacy. Oncoimmunology 2015; 4:e954829. [PMID: 25949858 PMCID: PMC4368153 DOI: 10.4161/21624011.2014.954829] [Citation(s) in RCA: 217] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 07/07/2014] [Indexed: 01/08/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) contribute to tumor-mediated immune escape and negatively correlate with overall survival of cancer patients. Nowadays, a variety of methods to target MDSCs are being investigated. Based on the intervention stage of MDSCs, namely development, expansion and activation, function and turnover, these methods can be divided into: (I) prevention or differentiation to mature cells, (II) blockade of MDSC expansion and activation, (III) inhibition of MDSC suppressive activity or (IV) depletion of intratumoral MDSCs. This review describes effective mono- or multimodal-therapies that target MDSCs for the benefit of cancer treatment.
Collapse
Key Words
- 5-FU, 5-fluorouracil
- 5-Fluorouracil
- ADAM17, metalloproteinase domain-containing protein 17
- APCs, antigen presenting cells
- ARG1, arginase-1
- ATRA, all-trans retinoic acid
- CCL2, chemokine (C-C motif) ligand 2
- CD62L, L-selectin
- CDDO-Me, bardoxolone methyl
- COX2, cyclooxygenase 2
- CTLs, cytotoxic T lymphocytes
- CXCL12, chemokine (C-X-C motif) ligand 12
- CXCL15, chemokine (C-X-C motif) ligand 15
- DCs, dendritic cells
- ERK1/2, extracellular signal-regulated kinases
- Flt3, Fms-like tyrosine kinase 3
- FoxP3, forkhead box P3
- GITR, anti-glucocorticoid tumor necrosis factor receptor
- GM-CSF/CSF2, granulocyte monocyte colony stimulating factor
- GSH, glutathione
- HIF-1α, hypoxia inducible factor 1α
- HLA, human leukocyte antigen
- HNSCC, head and neck squamous cell carcinoma
- HPV-16, human papillomavirus 16
- HSCs, hematopoietic stem cells
- ICT, 3, 5, 7-trihydroxy-4′-emthoxy-8-(3-hydroxy-3-methylbutyl)-flavone
- IFNγ, interferon γ
- IL-10, interleukin 10
- IL-13, interleukin 13
- IL-1β, interleukin 1 β
- IL-4, interleukin 4
- IL-6, interleukin 6
- IMCs, immature myeloid cells
- JAK2, Janus kinase 2
- MDSCs, myeloid-derived suppressor cells
- MMPs, metalloproteinases (e.g., MMP9)
- Myd88, myeloid differentiation primary response protein 88
- NAC, N-acetyl cysteine
- NADPH, nicotinamide adenine dinucleotide phosphate-oxidase NK cells, natural killer cells
- NO, nitric oxide
- NOHA, N-hydroxy-L-Arginine
- NSAID, nonsteroidal anti-inflammatory drugs
- ODN, oligodeoxynucleotides
- PDE-5, phosphodiesterase type 5
- PGE2, prostaglandin E2
- RNS, reactive nitrogen species
- ROS, reactive oxygen species
- SCF, stem cell factor
- STAT3, signal transducer and activator of transcription 3
- TAMs, tumor-associated macrophages
- TCR, T cell receptor
- TGFβ, transforming growth factor β
- TNFα, tumor necrosis factor α
- Tregs, regulatory T cells
- VEGFR, vascular endothelial growth factor receptor
- WA, withaferin A
- WRE, Withaferin somnifera
- all-trans retinoic acid
- bisphosphonates
- c-kit, Mast/stem cell growth factor receptor
- gemcitabine
- iNOS2, inducible nitric oxid synthase 2
- immune suppressive mechanisms
- mRCC, metastatic renal cell carcinoma
- myeloid-derived suppressor cells
- sunitinib therapeutic vaccination
Collapse
Affiliation(s)
- Oana Draghiciu
- Department of Medical Microbiology; Tumor Virology and Cancer Immunotherapy; University of Groningen; University Medical Center Groningen ; Groningen, The Netherlands
| | - Joyce Lubbers
- Department of Medical Microbiology; Tumor Virology and Cancer Immunotherapy; University of Groningen; University Medical Center Groningen ; Groningen, The Netherlands
| | - Hans W Nijman
- Department of Gynecology; University of Groningen; University Medical Center Groningen ; Groningen, The Netherlands
| | - Toos Daemen
- Department of Medical Microbiology; Tumor Virology and Cancer Immunotherapy; University of Groningen; University Medical Center Groningen ; Groningen, The Netherlands
| |
Collapse
|
127
|
Diakos CI, Charles KA, McMillan DC, Clarke SJ. Cancer-related inflammation and treatment effectiveness. Lancet Oncol 2015; 15:e493-503. [PMID: 25281468 DOI: 10.1016/s1470-2045(14)70263-3] [Citation(s) in RCA: 1604] [Impact Index Per Article: 160.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Inflammation is a recognised hallmark of cancer that substantially contributes to the development and progression of malignancies. In established cancers, there is increasing evidence for the roles that local immune response and systemic inflammation have in progression of tumours and survival of patients with cancer. This knowledge provides an opportunity to target these inflammatory responses to improve patient outcomes. In this Review, we examine the complex interplay between local immune responses and systemic inflammation, and their influence on clinical outcomes, and propose potential anti-inflammatory interventions for patients with cancer.
Collapse
Affiliation(s)
- Connie I Diakos
- Kolling Institute of Medical Research, University of Sydney, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Kellie A Charles
- School of Medical Sciences (Pharmacology), Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Donald C McMillan
- Academic Unit of Surgery, School of Medicine, University of Glasgow, Royal Infirmary, Glasgow, UK
| | - Stephen J Clarke
- Kolling Institute of Medical Research, University of Sydney, Royal North Shore Hospital, Sydney, NSW, Australia.
| |
Collapse
|
128
|
Ohshio Y, Hanaoka J, Kontani K, Teramoto K. Tranilast inhibits the function of cancer-associated fibroblasts responsible for the induction of immune suppressor cell types. Scand J Immunol 2015; 80:408-16. [PMID: 25224016 DOI: 10.1111/sji.12242] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 09/08/2014] [Indexed: 01/02/2023]
Abstract
Cancer-associated fibroblasts (CAFs) are the dominant stromal component in the tumour microenvironment (TME), playing critical roles in generation of pro-tumourigenic TME; however, their contribution to suppression of antitumour immune responses has not been fully understood. To elucidate the interaction between CAFs and immune suppressor cells, we examined whether inhibition of CAFs function would impair the induction of immune suppressor cell types in vitro. In this study, we applied an anti-allergic and antifibrotic agent tranilast, which is used clinically, and evaluated a potential of tranilast to serve as a CAFs inhibitor. CAFs that had been isolated from E.G7 or LLC1 tumour-bearing mice were cultured in the presence of tranilast, and thereafter, CAFs functions on the secretion of some soluble factors as well as the induction of immune suppressor cells were evaluated. As a result, tranilast inhibited the proliferation of CAFs and reduced the levels of stromal cell-derived factor-1, prostaglandin E2 and transforming growth factor-β1 from CAFs in a dose-dependent manner. On the other hand, tranilast exerted no inhibitory effects on immune cells at doses under 100 μm. The induction of regulatory T cells and myeloid-derived suppressor cells from their progenitor cells was suppressed in the medium that CAFs had been cultured in the presence of tranilast; however, these findings were not observed when those progenitor cells were cultured in the medium containing tranilast alone. These data demonstrate that tranilast inhibits CAFs function, which is responsible for the induction of immune suppressor cells, and possesses a potential to serve as a specific CAFs inhibitor.
Collapse
Affiliation(s)
- Y Ohshio
- Department of Surgery, Shiga University of Medical Science, Otsu, Japan
| | | | | | | |
Collapse
|
129
|
Chevolet I, Speeckaert R, Schreuer M, Neyns B, Krysko O, Bachert C, Van Gele M, van Geel N, Brochez L. Clinical significance of plasmacytoid dendritic cells and myeloid-derived suppressor cells in melanoma. J Transl Med 2015; 13:9. [PMID: 25592374 PMCID: PMC4326397 DOI: 10.1186/s12967-014-0376-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 12/26/2014] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Immune markers in the peripheral blood of melanoma patients could provide prognostic information. However, there is currently no consensus on which circulating cell types have more clinical impact. We therefore evaluated myeloid-derived suppressor cells (MDSC), dendritic cells (DC), cytotoxic T-cells and regulatory T-cells (Treg) in a series of blood samples of melanoma patients in different stages of disease. METHODS Flow cytometry was performed on peripheral blood mononuclear cells of 69 stage I to IV melanoma patients with a median follow-up of 39 months after diagnosis to measure the percentage of monocytic MDSCs (mMDSCs), polymorphonuclear MDSCs (pmnMDSCs), myeloid DCs (mDCs), plasmacytoid DCs (pDCs), cytotoxic T-cells and Tregs. We also assessed the expression of PD-L1 and CTLA-4 in cytotoxic T-cells and Tregs respectively. The impact of cell frequencies on prognosis was tested with multivariate Cox regression modelling. RESULTS Circulating pDC levels were decreased in patients with advanced (P = 0.001) or active (P = 0.002) disease. Low pDC levels conferred an independent negative impact on overall (P = 0.025) and progression-free survival (P = 0.036). Even before relapse, a decrease in pDC levels was observed (P = 0.002, correlation coefficient 0.898). High levels of circulating MDSCs (>4.13%) have an independent negative prognostic impact on OS (P = 0.012). MDSC levels were associated with decreased CD3+ (P < 0.001) and CD3 + CD8+ (P = 0.017) T-cell levels. Conversely, patients with high MDSC levels had more PD-L1+ T-cells (P = 0.033) and more CTLA-4 expression by Tregs (P = 0.003). pDCs and MDSCs were inversely correlated (P = 0.004). The impact of pDC levels on prognosis and prediction of the presence of systemic disease was stronger than that of MDSC levels. CONCLUSION We demonstrated that circulating pDC and MDSC levels are inversely correlated but have an independent prognostic value in melanoma patients. These cell types represent a single immunologic system and should be evaluated together. Both are key players in the immunological climate in melanoma patients, as they are correlated with circulating cytotoxic and regulatory T-cells. Circulating pDC and MDSC levels should be considered in future immunoprofiling efforts as they could impact disease management.
Collapse
Affiliation(s)
- Ines Chevolet
- Department of Dermatology, Ghent University Hospital, De Pintelaan 185, 9000, Ghent, Belgium.
| | - Reinhart Speeckaert
- Department of Dermatology, Ghent University Hospital, De Pintelaan 185, 9000, Ghent, Belgium.
| | - Max Schreuer
- Department of Medical Oncology, UZ-Brussel, Brussels, Belgium.
- Department of Medical Oncology, Ghent University Hospital, Ghent, Belgium.
| | - Bart Neyns
- Department of Medical Oncology, UZ-Brussel, Brussels, Belgium.
| | - Olga Krysko
- Upper Airways Research Laboratory, Ghent University Hospital, Ghent, Belgium.
| | - Claus Bachert
- Upper Airways Research Laboratory, Ghent University Hospital, Ghent, Belgium.
| | - Mireille Van Gele
- Department of Dermatology, Ghent University Hospital, De Pintelaan 185, 9000, Ghent, Belgium.
| | - Nanja van Geel
- Department of Dermatology, Ghent University Hospital, De Pintelaan 185, 9000, Ghent, Belgium.
| | - Lieve Brochez
- Department of Dermatology, Ghent University Hospital, De Pintelaan 185, 9000, Ghent, Belgium.
| |
Collapse
|
130
|
Ohshio Y, Teramoto K, Hanaoka J, Tezuka N, Itoh Y, Asai T, Daigo Y, Ogasawara K. Cancer-associated fibroblast-targeted strategy enhances antitumor immune responses in dendritic cell-based vaccine. Cancer Sci 2015; 106:134-42. [PMID: 25483888 PMCID: PMC4399032 DOI: 10.1111/cas.12584] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 11/18/2014] [Accepted: 11/28/2014] [Indexed: 12/13/2022] Open
Abstract
Given the close interaction between tumor cells and stromal cells in the tumor microenvironment (TME), TME-targeted strategies would be promising for developing integrated cancer immunotherapy. Cancer-associated fibroblasts (CAFs) are the dominant stromal component, playing critical roles in generation of the pro-tumorigenic TME. We focused on the immunosuppressive trait of CAFs, and systematically explored the alteration of tumor-associated immune responses by CAF-targeted therapy. C57BL/6 mice s.c. bearing syngeneic E.G7 lymphoma, LLC1 Lewis lung cancer, or B16F1 melanoma were treated with an anti-fibrotic agent, tranilast, to inhibit CAF function. The infiltration of immune suppressor cell types, including regulatory T cells and myeloid-derived suppressor cells, in the TME was effectively decreased through reduction of stromal cell-derived factor-1, prostaglandin E2 , and transforming growth factor-β. In tumor-draining lymph nodes, these immune suppressor cell types were significantly decreased, leading to activation of tumor-associated antigen-specific CD8(+) T cells. In addition, CAF-targeted therapy synergistically enhanced multiple types of systemic antitumor immune responses such as the cytotoxic CD8(+) T cell response, natural killer activity, and antitumor humoral immunity in combination with dendritic cell-based vaccines; however, the suppressive effect on tumor growth was not observed in tumor-bearing SCID mice. These data indicate that systemic antitumor immune responses by various immunologic cell types are required to bring out the efficacy of CAF-targeted therapy, and these effects are enhanced when combined with effector-stimulatory immunotherapy such as dendritic cell-based vaccines. Our mouse model provides a novel rationale with TME-targeted strategy for the development of cell-based cancer immunotherapy.
Collapse
Affiliation(s)
- Yasuhiko Ohshio
- Department of Surgery, Shiga University of Medical Science, Shiga, Japan
| | | | | | | | | | | | | | | |
Collapse
|
131
|
Kumar V, Gabrilovich DI. Hypoxia-inducible factors in regulation of immune responses in tumour microenvironment. Immunology 2015; 143:512-9. [PMID: 25196648 DOI: 10.1111/imm.12380] [Citation(s) in RCA: 263] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 08/21/2014] [Accepted: 08/29/2014] [Indexed: 12/14/2022] Open
Abstract
Hypoxia is one of the hallmarks of the tumour microenvironment. It is the result of insufficient blood supply to support proliferating tumour cells. In response to hypoxia, the cellular machinery uses mechanisms whereby the low level of oxygen is sensed and counterbalanced by changing the transcription of numerous genes. Hypoxia-inducible factors (HIF) play a critical role in the regulation of cellular responses to hypoxia. In recent years ample evidence has indicated that HIF play a prominent role in tumour immune responses. Up-regulation of HIF1α promotes immune suppressive activity of myeloid-derived suppressive cells (MDSC) and tumour-associated macrophages (TAM) and rapid differentiation of MDSC to TAM. HIF1α does not affect MDSC differentiation to dendritic cells (DC) but instead causes DC activation. HIF inhibit effector functions of tumour-infiltrating lymphocytes. HIF1α inhibits regulatory T (Treg) cell development by switching the balance towards T helper type 17 cells. However, as a major part of Treg cell differentiation does not take place in the tumour site, a functionally more important role of HIF1α is in the promotion of Treg cell recruitment to the tumour site in response to chemokines. As a result, the presence of Treg cells inside tumours is increased. Hence, HIF play a largely negative role in the regulation of immune responses inside tumours. It appears that therapeutic strategies targeting HIF in the immune system could be beneficial for anti-tumour immune responses.
Collapse
|
132
|
Maenhout SK, Thielemans K, Aerts JL. Location, location, location: functional and phenotypic heterogeneity between tumor-infiltrating and non-infiltrating myeloid-derived suppressor cells. Oncoimmunology 2014; 3:e956579. [PMID: 25941577 DOI: 10.4161/21624011.2014.956579] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 08/16/2014] [Indexed: 12/26/2022] Open
Abstract
An increasing number of studies is focusing on the role of myeloid-derived suppressor cells (MDSCs) in the suppression of antitumor immune responses. Although the main site of action for MDSCs is most likely the tumor microenvironment, the study of these cells has been largely restricted to MDSCs derived from peripheral lymphoid organs. Only in a minority of studies MDSCs isolated from the tumor microenvironment have been characterized. This review will give an overview of the data available on the phenotypical and functional differences between tumor-derived MDSCs and MDSCs isolated from the spleen of tumor-bearing mice or from the peripheral blood of cancer patients.
Collapse
Key Words
- ATRA, all-trans retinoic acid
- Bv8, Bombina variagata peptide 8
- CTLA-4, cytotoxic T-lymphocyte antigen-4
- GM-CSF, granulocyte-macrophage colony-stimulating factor
- IFN-γ, interferon gamma
- IL, interleukin
- IL-4Rα, interleukin-4 receptor alpha
- LPS, lipopolysaccharide
- M-CSF, macrophage-colony stimulating factor
- MAPK, mitogen-activated protein kinases
- MDSCs, myeloid-derived suppressor cells
- NS cells, natural suppressor cells
- PD-L1, programmed death-ligand 1
- PHA, phytohemagglutinin
- ROS, reactive oxygen species
- TAMs, tumor-associated macrophages
- Treg, regulatory T cells
- VEGF, vascular endothelial growth factor.
- iNOS, inducible nitric oxide synthase
- immunosuppression
- myeloid-derived suppressor cells
- siRNA, small interfering ribonucleic acid
- tumor immunology
- tumor microenvironment
- tumor models
Collapse
Affiliation(s)
- Sarah K Maenhout
- Laboratory of Molecular and Cellular Therapy; Department of Immunology-Physiology ; Vrije Universiteit Brussel ; Brussels, Belgium
| | - Kris Thielemans
- Laboratory of Molecular and Cellular Therapy; Department of Immunology-Physiology ; Vrije Universiteit Brussel ; Brussels, Belgium
| | - Joeri L Aerts
- Laboratory of Molecular and Cellular Therapy; Department of Immunology-Physiology ; Vrije Universiteit Brussel ; Brussels, Belgium
| |
Collapse
|
133
|
Wang Y, Jin TH, Farhana A, Freeman J, Estell K, Zmijewski JW, Gaggar A, Thannickal VJ, Schwiebert LM, Steyn AJC, Deshane JS. Exposure to cigarette smoke impacts myeloid-derived regulatory cell function and exacerbates airway hyper-responsiveness. J Transl Med 2014; 94:1312-25. [PMID: 25365203 PMCID: PMC4245361 DOI: 10.1038/labinvest.2014.126] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 09/08/2014] [Accepted: 09/12/2014] [Indexed: 01/26/2023] Open
Abstract
Cigarette smoking enhances oxidative stress and airway inflammation in asthma, the mechanisms of which are largely unknown. Myeloid-derived regulatory cells (MDRC) are free radical producing immature myeloid cells with immunoregulatory properties that have recently been demonstrated as critical regulators of allergic airway inflammation. NO (nitric oxide)-producing immunosuppressive MDRC suppress T-cell proliferation and airway-hyper responsiveness (AHR), while the O2(•-) (superoxide)-producing MDRC are proinflammatory. We hypothesized that cigarette smoke (CS) exposure may impact MDRC function and contribute to exacerbations in asthma. Exposure of bone marrow (BM)-derived NO-producing MDRC to CS reduced the production of NO and its metabolites and inhibited their potential to suppress T-cell proliferation. Production of immunoregulatory cytokine IL-10 was significantly inhibited, while proinflammatory cytokines IL-6, IL-1β, TNF-α and IL-33 were enhanced in CS-exposed BM-MDRC. Additionally, CS exposure increased NF-κB activation and induced BM-MDRC-mediated production of O2(•-), via NF-κB-dependent pathway. Intratracheal transfer of smoke-exposed MDRC-producing proinflammatory cytokines increased NF-κB activation, reactive oxygen species and mucin production in vivo and exacerbated AHR in C57BL/6 mice, mice deficient in Type I IFNR and MyD88, both with reduced numbers of endogenous MDRC. Thus CS exposure modulates MDRC function and contributes to asthma exacerbation and identifies MDRC as potential targets for asthma therapy.
Collapse
Affiliation(s)
- Yong Wang
- Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Tong Huan Jin
- Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Aisha Farhana
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jason Freeman
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kim Estell
- Department of Cell Developmental and Integrative Biology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jaroslaw W Zmijewski
- Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Amit Gaggar
- Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Victor J Thannickal
- Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lisa M Schwiebert
- Department of Cell Developmental and Integrative Biology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Adrie J C Steyn
- 1] Department of Microbiology, The University of Alabama at Birmingham, Birmingham, AL, USA [2] KwaZulu-Natal Research Institute for Tuberculosis and HIV, Durban, South Africa
| | - Jessy S Deshane
- Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
134
|
Messmer MN, Netherby CS, Banik D, Abrams SI. Tumor-induced myeloid dysfunction and its implications for cancer immunotherapy. Cancer Immunol Immunother 2014; 64:1-13. [PMID: 25432147 DOI: 10.1007/s00262-014-1639-3] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 11/19/2014] [Indexed: 01/27/2023]
Abstract
Immune function relies on an appropriate balance of the lymphoid and myeloid responses. In the case of neoplasia, this balance is readily perturbed by the dramatic expansion of immature or dysfunctional myeloid cells accompanied by a reciprocal decline in the quantity/quality of the lymphoid response. In this review, we seek to: (1) define the nature of the atypical myelopoiesis observed in cancer patients and the impact of this perturbation on clinical outcomes; (2) examine the potential mechanisms underlying these clinical manifestations; and (3) explore potential strategies to restore normal myeloid cell differentiation to improve activation of the host antitumor immune response. We posit that fundamental alterations in myeloid homeostasis triggered by the neoplastic process represent critical checkpoints that govern therapeutic efficacy, as well as offer novel cellular-based biomarkers for tracking changes in disease status or relapse.
Collapse
Affiliation(s)
- Michelle N Messmer
- Department of Immunology, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | | | | | | |
Collapse
|
135
|
A circulating subpopulation of monocytic myeloid-derived suppressor cells as an independent prognostic/predictive factor in untreated non-small lung cancer patients. J Immunol Res 2014; 2014:659294. [PMID: 25436215 PMCID: PMC4243712 DOI: 10.1155/2014/659294] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 09/19/2014] [Accepted: 09/29/2014] [Indexed: 12/30/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) represent a heterogeneous population of cells with immunosuppressive properties and might confer to worse prognosis in cancer patients. The presence of phenotypically newly described subpopulations of MDSCs and their association with the clinical outcome were investigated in non-small cell lung cancer (NSCLC) patients. The percentages and correlation between MDSCs and distinct immune cells in the peripheral blood of 110 chemotherapy-naive patients before treatment and healthy controls were investigated using flow cytometry. Two monocytic [CD14(+)CD15(-)CD11b(+)CD33(+)HLA-DR(-)Lin(-) and CD14(+)CD15(+)CD11b(+)CD33(+)HLA-DR(-)Lin(-)] and a granulocytic [CD14(-)CD15(+)CD11b(+)CD33(+)HLA-DR(-)Lin(-)] subpopulations of MDSCs were identified, expressing inducible nitric oxide synthase, and reactive oxygen species, respectively. Increased percentages of both monocytic-MDSCs' subpopulations were inversely correlated to dendritic/monocyte levels (P ≤ 0.04), while granulocytic-MDSCs were inversely correlated to CD4(+) T cells (P = 0.006). Increased percentages of monocytic-MDSCs were associated with worse response to treatment (P = 0.02) and patients with normal levels of CD14(+)CD15(+)CD11b(+)CD33(+)HLA-DR(-)Lin(-) had longer overall survival and progression free-survival compared to those with high levels (P = 0.008 and P = 0.005, resp.). Multivariate analysis revealed that the increased percentages of CD14(+)CD15(+)CD11b(+)CD33(+)HLA-DR(-)Lin(-) MDSCs were independently associated with decreased progression free-survival and overall survival. The data provide evidence that increased percentages of new monocytic-MDSCs' subpopulations in advanced NSCLC patients are associated with an unfavourable clinical outcome.
Collapse
|
136
|
Rudolph BM, Loquai C, Gerwe A, Bacher N, Steinbrink K, Grabbe S, Tuettenberg A. Increased frequencies of CD11b(+) CD33(+) CD14(+) HLA-DR(low) myeloid-derived suppressor cells are an early event in melanoma patients. Exp Dermatol 2014; 23:202-4. [PMID: 24495013 DOI: 10.1111/exd.12336] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/30/2014] [Indexed: 11/28/2022]
Abstract
Myeloid-derived suppressor cells (MDSC) are a heterogeneous cell population characterized by immunosuppressive activity. Elevated levels of MDSC in peripheral blood are found in inflammatory diseases as well as in malignant tumors where they are supposed to be major contributors to mechanisms of tumor-associated tolerance. We investigated the frequency and function of MDSC in peripheral blood of melanoma patients and observed an accumulation of CD11b(+) CD33(+) CD14(+) HLA-DR(low) MDSC in all stages of disease (I-IV), including early stage I patients. Disease progression and enhanced tumor burden did not result in a further increase in frequencies or change in phenotype of MDSC. By investigation of specific MDSC-associated cytokines in patients' sera, we found an accumulation of IL-8 in all stages of disease. T-cell proliferation assays revealed that MDSC critically contribute to suppressed antigen-specific T-cell reactivity and thus might explain the frequently observed transient effects of immunotherapeutic strategies in melanoma patients.
Collapse
Affiliation(s)
- Berenice M Rudolph
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | | | | | | | | | | | | |
Collapse
|
137
|
Jiang J, Guo W, Liang X. Phenotypes, accumulation, and functions of myeloid-derived suppressor cells and associated treatment strategies in cancer patients. Hum Immunol 2014; 75:1128-37. [DOI: 10.1016/j.humimm.2014.09.025] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Revised: 09/27/2014] [Accepted: 09/27/2014] [Indexed: 02/07/2023]
|
138
|
Abstract
Chronic inflammation predisposes patients with inflammatory bowel disease to the risk of developing colitis-associated cancer (CAC). Growing evidence strongly suggests that CAC development is multifactorial and is attributed to concurrent, dynamic dysregulations in host immunity, enteric microbiota, and epithelial restitution during the course of chronic inflammation. This article discusses the recent advances in understanding the different forms of CAC that may develop in patients with inflammatory bowel disease and animal models, as well as molecular alterations and other processes that orchestrate the development of CAC.
Collapse
|
139
|
Waldron TJ, Quatromoni JG, Karakasheva TA, Singhal S, Rustgi AK. Myeloid derived suppressor cells: Targets for therapy. Oncoimmunology 2014; 2:e24117. [PMID: 23734336 PMCID: PMC3654606 DOI: 10.4161/onci.24117] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 02/22/2013] [Accepted: 02/26/2013] [Indexed: 01/14/2023] Open
Abstract
The goal of achieving measurable response with cancer immunotherapy requires counteracting the immunosuppressive characteristics of tumors. One of the mechanisms that tumors utilize to escape immunosurveillance is the activation of myeloid derived suppressor cells (MDSCs). Upon activation by tumor-derived signals, MDSCs inhibit the ability of the host to mount an anti-tumor immune response via their capacity to suppress both the innate and adaptive immune systems. Despite their relatively recent discovery and characterization, anti-MDSC agents have been identified, which may improve immunotherapy efficacy.
Collapse
Affiliation(s)
- Todd J Waldron
- Gastroenterology Division; Department of Medicine; University of Pennsylvania; Philadelphia, PA USA ; Abramson Cancer Center; University of Pennsylvania; Philadelphia, PA USA
| | | | | | | | | |
Collapse
|
140
|
De Vita M, Catzola V, Buzzonetti A, Fossati M, Battaglia A, Zamai L, Fattorossi A. Unexpected interference in cell surface staining by monoclonal antibodies to unrelated antigens. CYTOMETRY PART B-CLINICAL CYTOMETRY 2014; 88:352-4. [DOI: 10.1002/cyto.b.21197] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 09/01/2014] [Accepted: 09/29/2014] [Indexed: 11/10/2022]
Affiliation(s)
- Martina De Vita
- Laboratorio di Immunologia dei tumori; Unità di Ginecologia Oncologica; Università Cattolica del Sacro Cuore; Roma Italia
| | - Valentina Catzola
- Laboratorio di Immunologia dei tumori; Unità di Ginecologia Oncologica; Università Cattolica del Sacro Cuore; Roma Italia
| | - Alexia Buzzonetti
- Laboratorio di Immunologia dei tumori; Unità di Ginecologia Oncologica; Università Cattolica del Sacro Cuore; Roma Italia
| | - Marco Fossati
- Laboratorio di Immunologia dei tumori; Unità di Ginecologia Oncologica; Università Cattolica del Sacro Cuore; Roma Italia
| | - Alessandra Battaglia
- Laboratorio di Immunologia dei tumori; Unità di Ginecologia Oncologica; Università Cattolica del Sacro Cuore; Roma Italia
| | - Loris Zamai
- Dipartimento di Scienze della Terra; della Vita e dell'Ambiente; Università di Urbino “Carlo Bo,”; Urbino Italia
| | - Andrea Fattorossi
- Laboratorio di Immunologia dei tumori; Unità di Ginecologia Oncologica; Università Cattolica del Sacro Cuore; Roma Italia
| |
Collapse
|
141
|
Zoso A, Mazza EMC, Bicciato S, Mandruzzato S, Bronte V, Serafini P, Inverardi L. Human fibrocytic myeloid-derived suppressor cells express IDO and promote tolerance via Treg-cell expansion. Eur J Immunol 2014; 44:3307-19. [PMID: 25113564 DOI: 10.1002/eji.201444522] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 07/02/2014] [Accepted: 08/08/2014] [Indexed: 01/03/2023]
Abstract
By restraining T-cell activation and promoting Treg-cell expansion, myeloid-derived suppressor cells (MDSCs) and tolerogenic DCs can control self-reactive and antigraft effector T cells in autoimmunity and transplantation. Their therapeutic use and characterization, however, is limited by their scarce availability in the peripheral blood of tumor-free donors. In the present study, we describe and characterize a novel population of human myeloid suppressor cells, named fibrocytic MDSC, which are differentiated from umbilical cord blood precursors by 4-day culture with FDA-approved cytokines (recombinant human-GM-CSF and recombinant human-G-CSF). This MDSC subset, characterized by the expression of MDSC-, DC-, and fibrocyte-associated markers, promotes Treg-cell expansion and induces normoglycemia in a xenogeneic mouse model of Type 1 diabetes. In order to exert their protolerogenic function, fibrocytic MDSCs require direct contact with activated T cells, which leads to the production and secretion of IDO. This new myeloid subset may have an important role in the in vitro and in vivo production of Treg cells for the treatment of autoimmune diseases, and in either the prevention or control of allograft rejection.
Collapse
Affiliation(s)
- Alessia Zoso
- Diabetes Research Institute, University of Miami, Miami, FL, USA
| | | | | | | | | | | | | |
Collapse
|
142
|
Weed DT, Vella JL, Reis IM, De la Fuente AC, Gomez C, Sargi Z, Nazarian R, Califano J, Borrello I, Serafini P. Tadalafil reduces myeloid-derived suppressor cells and regulatory T cells and promotes tumor immunity in patients with head and neck squamous cell carcinoma. Clin Cancer Res 2014; 21:39-48. [PMID: 25320361 DOI: 10.1158/1078-0432.ccr-14-1711] [Citation(s) in RCA: 213] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE Myeloid-derived suppressor cells (MDSC) and regulatory T cells (Treg) play a key role in the progression of head and neck squamous cell carcinoma (HNSCC). On the basis of our preclinical data demonstrating that phosphodiesterase-5 (PDE5) inhibition can modulate these cell populations, we evaluated whether the PDE5 inhibitor tadalafil can revert tumor-induced immunosuppression and promote tumor immunity in patients with HNSCC. EXPERIMENTAL DESIGN First, we functionally and phenotypically characterized MDSCs in HNSCCs and determined, retrospectively, whether their presence at the tumor site correlates with recurrence. Then, we performed a prospective single-center, double-blinded, randomized, three-arm study in which patients with HNSCC undergoing definitive surgical resection of oral and oropharyngeal tumors were treated with tadalafil 10 mg/day, 20 mg/day, or placebo for at least 20 days preoperatively. Blood and tumor MDSC and Treg presence and CD8(+) T-cell reactivity to tumor antigens were evaluated before and after treatment. RESULTS MDSCs were characterized in HNSCC and their intratumoral presence significantly correlates with recurrence. Tadalafil treatment was well tolerated and significantly reduced both MDSCs and Treg concentrations in the blood and in the tumor (P < 0.05). In addition, the concentration of blood CD8(+) T cells reactive to autologous tumor antigens significantly increased after treatment (P < 0.05). Tadalafil immunomodulatory activity was maximized at an intermediate dose but not at higher doses. Mechanistic analysis suggests a possible off-target effect on PDE11 at high dosages that, by increasing intracellular cAMP, may negatively affect antitumor immunity. CONCLUSIONS Tadalafil seems to beneficially modulate the tumor micro- and macro-environment in patients with HNSCC by lowering MDSCs and Tregs and increasing tumor-specific CD8(+) T cells in a dose-dependent fashion.
Collapse
Affiliation(s)
- Donald T Weed
- Department of Otolaryngology, University of Miami, Miller School of Medicine, Miami, Florida
| | - Jennifer L Vella
- Department of Microbiology and Immunology, University of Miami, Miller School of Medicine, Miami, Florida
| | - Isildinha M Reis
- Department of Public Health Sciences and Sylvester Biostatistics and Bioinformatics Core Resource, University of Miami, Miller School of Medicine, Miami, Florida
| | - Adriana C De la Fuente
- Department of Microbiology and Immunology, University of Miami, Miller School of Medicine, Miami, Florida
| | - Carmen Gomez
- Department of Pathology, University of Miami, Miller School of Medicine, Miami, Florida
| | - Zoukaa Sargi
- Department of Otolaryngology, University of Miami, Miller School of Medicine, Miami, Florida
| | - Ronen Nazarian
- Department of Otolaryngology, University of Miami, Miller School of Medicine, Miami, Florida
| | - Joseph Califano
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University, Baltimore, Maryland.,Milton J. Dance Head and Neck Center, Greater Baltimore Medical Center, Baltimore, Maryland
| | - Ivan Borrello
- Oncology Department, Johns Hopkins University, Baltimore, Maryland
| | - Paolo Serafini
- Department of Microbiology and Immunology, University of Miami, Miller School of Medicine, Miami, Florida
| |
Collapse
|
143
|
Busch A, Zeh D, Janzen V, Mügge LO, Wolf D, Fingerhut L, Hahn-Ast C, Maurer O, Brossart P, von Lilienfeld-Toal M. Treatment with lenalidomide induces immunoactivating and counter-regulatory immunosuppressive changes in myeloma patients. Clin Exp Immunol 2014; 177:439-53. [PMID: 24712857 DOI: 10.1111/cei.12343] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2014] [Indexed: 01/13/2023] Open
Abstract
Lenalidomide activates the immune system, but the exact immunomodulatory mechanisms of lenalidomide in vivo are poorly defined. In an observational study we assessed the impact of lenalidomide on different populations of immune cells in multiple myeloma patients. Lenalidomide therapy was associated with increased amounts of a CD8(+) T cell subset, phenotypically staged between classical central memory T cells (TCM) and effector memory T cells (TEM), consequently termed TCM/TEM. The moderate expression of perforin/granzyme and phenotypical profile of these cells identifies them as not yet terminally differentiated, which makes them promising candidates for the anti-tumour response. In addition, lenalidomide-treated patients showed higher abundance of CD14(+) myeloid cells co-expressing CD15. This population was able to inhibit both CD4(+) and CD8(+) T cell proliferation in vitro and could thus be defined as a so far undescribed novel myeloid-derived suppressor cell (MDSC) subtype. We observed a striking correlation between levels of TCM/TEM, mature regulatory T cells (T(regs)) and CD14(+) CD15(+) MDSCs. In summary, lenalidomide induces both activating and inhibitory components of the immune system, indicating the existence of potential counter-regulatory mechanisms. These findings provide new insights into the immunomodulatory action of lenalidomide.
Collapse
Affiliation(s)
- A Busch
- Department of Hematology, Oncology and Rheumatology, University Hospital Bonn, Bonn
| | | | | | | | | | | | | | | | | | | |
Collapse
|
144
|
Abstract
It is widely accepted that intake of dietary fats and chronic inflammation are risk factors for developing colorectal cancer. Arachidonic acid is a major component of animal fats, and the bioactive lipids produced from this substrate play critical roles in a variety of biologic processes, including cancer. Cyclooxygenase-derived prostaglandin E2 is a known proinflammatory lipid mediator that promotes tumor progression. Metabolism of arachidonic acid by the cyclooxygenase pathway provides one mechanism for the contribution of dietary fats and chronic inflammation to carcinogenesis. In this review, we highlight recent advances in our understanding of how a proinflammatory mediator prostaglandin E2 promotes colorectal cancer immune evasion. These findings may provide a rationale for the development of new therapeutic approaches to subvert tumor-induced immunosuppression.
Collapse
|
145
|
Rigoni A, Colombo MP, Pucillo C. The Role of Mast Cells in Molding the Tumor Microenvironment. CANCER MICROENVIRONMENT 2014; 8:167-76. [PMID: 25194694 DOI: 10.1007/s12307-014-0152-8] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 08/20/2014] [Indexed: 12/20/2022]
Abstract
Mast cells (MCs) are granulocytic immune cells that reside in tissues exposed to the external environment. MCs are best known for their activity in allergic reactions, but they have been involved in different physiological and pathological conditions. In particular, MC infiltration has been shown in several types of human tumors and in animal cancer models. Nevertheless, the role of MCs in the tumor microenvironment is still debated because they have been associated either to good or poor prognosis depending on tumor type and tissue localization. This dichotomous role relies on MC capacity to secrete a broad spectrum of molecules with modulatory functions, which may condition the final tumor outcome also promoting angiogenesis and tissue remodeling. In this review, we analyze the multifaceted role of mast cell in tumor progression and inhibition considering their ability to interact with: i) immune cells, ii) tumor cells and iii) the extracellular matrix. Eventually, the current MC targeting strategies to treat cancer patients are discussed. Deciphering the actual role of MCs in tumor onset and progression is crucial to identify MC-targeted treatments aimed at killing cancer cells or at making the tumor vulnerable to selected anti-cancer drugs.
Collapse
Affiliation(s)
- A Rigoni
- Molecular Immunology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, via Amadeo 42, 20133, Milan, Italy
| | - M P Colombo
- Molecular Immunology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, via Amadeo 42, 20133, Milan, Italy.
| | - C Pucillo
- Department of Medical and Biological Sciences, University of Udine, 33100, Udine, Italy
| |
Collapse
|
146
|
Wu T, Zhao Y, Zhao Y. The roles of myeloid-derived suppressor cells in transplantation. Expert Rev Clin Immunol 2014; 10:1385-94. [DOI: 10.1586/1744666x.2014.948424] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
147
|
Kitano S, Postow MA, Ziegler CGK, Kuk D, Panageas KS, Cortez C, Rasalan T, Adamow M, Yuan J, Wong P, Altan-Bonnet G, Wolchok JD, Lesokhin AM. Computational algorithm-driven evaluation of monocytic myeloid-derived suppressor cell frequency for prediction of clinical outcomes. Cancer Immunol Res 2014; 2:812-21. [PMID: 24844912 PMCID: PMC4125466 DOI: 10.1158/2326-6066.cir-14-0013] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Evaluation of myeloid-derived suppressor cells (MDSC), a cell type implicated in T-cell suppression, may inform immune status. However, a uniform methodology is necessary for prospective testing as a biomarker. We report the use of a computational algorithm-driven analysis of whole blood and cryopreserved samples for monocytic MDSC (m-MDSC) quantity that removes variables related to blood processing and user definitions. Applying these methods to samples from patients with melanoma identifies differing frequency distribution of m-MDSC relative to that in healthy donors. Patients with a pretreatment m-MDSC frequency outside a preliminary definition of healthy donor range (<14.9%) were significantly more likely to achieve prolonged overall survival following treatment with ipilimumab, an antibody that promotes T-cell activation and proliferation. m-MDSC frequencies were inversely correlated with peripheral CD8(+) T-cell expansion following ipilimumab. Algorithm-driven analysis may enable not only development of a novel pretreatment biomarker for ipilimumab therapy, but also prospective validation of peripheral blood m-MDSCs as a biomarker in multiple disease settings.
Collapse
Affiliation(s)
- Shigehisa Kitano
- Department of Experimental Therapeutics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Tsukiji, Tokyo, Japan
| | - Michael A Postow
- Memorial Sloan-Kettering Cancer Center; Weill-Cornell Medical and Graduate Schools; and
| | | | | | | | - Czrina Cortez
- Memorial Sloan-Kettering Cancer Center; Ludwig Collaborative and Swim Across America Lab, New York, New York
| | - Teresa Rasalan
- Ludwig Center for Cancer Immunotherapy; Memorial Sloan-Kettering Cancer Center; Ludwig Collaborative and Swim Across America Lab, New York, New York
| | - Mathew Adamow
- Ludwig Center for Cancer Immunotherapy; Memorial Sloan-Kettering Cancer Center
| | - Jianda Yuan
- Ludwig Center for Cancer Immunotherapy; Memorial Sloan-Kettering Cancer Center
| | - Philip Wong
- Ludwig Center for Cancer Immunotherapy; Memorial Sloan-Kettering Cancer Center
| | | | - Jedd D Wolchok
- Ludwig Center for Cancer Immunotherapy; Memorial Sloan-Kettering Cancer Center; Weill-Cornell Medical and Graduate Schools; and Ludwig Collaborative and Swim Across America Lab, New York, New York
| | - Alexander M Lesokhin
- Memorial Sloan-Kettering Cancer Center; Weill-Cornell Medical and Graduate Schools; and Ludwig Collaborative and Swim Across America Lab, New York, New York
| |
Collapse
|
148
|
Solito S, Marigo I, Pinton L, Damuzzo V, Mandruzzato S, Bronte V. Myeloid-derived suppressor cell heterogeneity in human cancers. Ann N Y Acad Sci 2014; 1319:47-65. [DOI: 10.1111/nyas.12469] [Citation(s) in RCA: 302] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Samantha Solito
- Department of Surgery; Oncology and Gastroenterology; Oncology and Immunology Section; University of Padova; Padova Italy
| | | | - Laura Pinton
- Department of Surgery; Oncology and Gastroenterology; Oncology and Immunology Section; University of Padova; Padova Italy
| | - Vera Damuzzo
- Department of Surgery; Oncology and Gastroenterology; Oncology and Immunology Section; University of Padova; Padova Italy
| | - Susanna Mandruzzato
- Department of Surgery; Oncology and Gastroenterology; Oncology and Immunology Section; University of Padova; Padova Italy
- Istituto Oncologico Veneto; IOV-IRCCS; Padova Italy
| | - Vincenzo Bronte
- Pathology and Diagnostics; Verona University Hospital; Verona Italy
| |
Collapse
|
149
|
Janols H, Bergenfelz C, Allaoui R, Larsson AM, Rydén L, Björnsson S, Janciauskiene S, Wullt M, Bredberg A, Leandersson K. A high frequency of MDSCs in sepsis patients, with the granulocytic subtype dominating in gram-positive cases. J Leukoc Biol 2014; 96:685-93. [DOI: 10.1189/jlb.5hi0214-074r] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
150
|
Zhao T, Du H, Ding X, Walls K, Yan C. Activation of mTOR pathway in myeloid-derived suppressor cells stimulates cancer cell proliferation and metastasis in lal(-/-) mice. Oncogene 2014; 34:1938-48. [PMID: 24882582 PMCID: PMC4254377 DOI: 10.1038/onc.2014.143] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 03/06/2014] [Accepted: 04/03/2014] [Indexed: 12/15/2022]
Abstract
Inflammation critically contributes to cancer metastasis, in which myeloid-derived suppressor cells (MDSCs) are an important participant. Although MDSCs are known to suppress immune surveillance, their roles in directly stimulating cancer cell proliferation and metastasis currently remain unclear. Lysosomal acid lipase (LAL) deficiency causes systemic expansion and infiltration of MDSCs in multiple organs and subsequent inflammation. In the LAL-deficient (lal−/−) mouse model, melanoma metastasized massively in allogeneic lal−/− mice, which was suppressed in allogeneic lal+/+ mice due to immune rejection. Here we report for the first time that MDSCs from lal−/− mice directly stimulated B16 melanoma cell in vitro proliferation, and in vivo growth and metastasis. Cytokines i.e., IL-1β and TNFα from MDSCs are required for B16 melanoma cell proliferation in vitro. Myeloid-specific expression of human LAL (hLAL) in lal−/− mice rescues these malignant phenotypes in vitro and in vivo. The tumor-promoting function of lal−/− MDSCs is mediated, at least in part, through over-activation of the mammalian target of rapamycin (mTOR) pathway. Knockdown of mTOR, Raptor or Rictor in lal−/− MDSCs suppressed their stimulation on proliferation of cancer cells, including B16 melanoma, LLC and Tramp-C2 cancer cells. Our results indicate that LAL plays a critical role in regulating MDSCs ability to directly stimulate cancer cell proliferation, and overcome immune rejection of cancer metastasis in allogeneic mice through modulation of the mTOR pathway, which provides a mechanistic basis for targeting MDSCs to reduce the risk of cancer metastasis. Therefore, MDSCs possess dual functions to facilitate cancer metastasis: suppress immune surveillance, and stimulate cancer cell proliferation and growth.
Collapse
Affiliation(s)
- T Zhao
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - H Du
- 1] Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA [2] IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - X Ding
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - K Walls
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - C Yan
- 1] Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA [2] IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA [3] Center for Immunobiology, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|