151
|
Wang D, DuBois RN. Myeloid-derived suppressor cells link inflammation to cancer. Oncoimmunology 2014; 3:e28581. [PMID: 25054088 PMCID: PMC4092003 DOI: 10.4161/onci.28581] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 03/18/2014] [Indexed: 11/19/2022] Open
Abstract
Our recent work has provided the first evidence that MDSCs promote chronic colonic inflammation and colitis-associated carcinogenesis. Our findings not only reveal a novel function of MDSCs in connecting inflammation to cancer, but also provide a rationale for developing effective therapeutic strategies to subvert inflammation- and tumor-induced immunosuppression.
Collapse
Affiliation(s)
- Dingzhi Wang
- Laboratory for Inflammation and Cancer; the Biodesign Institute at Arizona State University; Tempe, AZ USA
| | - Raymond N DuBois
- Laboratory for Inflammation and Cancer; the Biodesign Institute at Arizona State University; Tempe, AZ USA ; Department of Chemistry and Biochemistry; Arizona State University; Tempe, AZ USA ; Department of Research and Division of Gastroenterology; Mayo Clinic; Scottsdale, AZ USA
| |
Collapse
|
152
|
Lack of significant elevation of myeloid-derived suppressor cells in peripheral blood of chronically hepatitis C virus-infected individuals. J Virol 2014; 88:7678-82. [PMID: 24741087 DOI: 10.1128/jvi.00113-14] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSC) are immature myeloid cells with immunosuppressive function. Compared to the level in healthy controls (HC), no elevation of MDSC in chronic hepatitis C (cHEP-C) patients was found, and there was no difference in MDSC based on genotype or viral load (P > 0.25). Moreover, MDSC of cHEP-C patients inhibited CD8 T cell function as efficiently as MDSC of HC did. Since we detected neither quantitative nor qualitative differences in MDSC of cHEP-C patients relative to those of HC, we postulate that MDSC in peripheral blood are most likely not significant regarding immune dysfunction in cHEP-C.
Collapse
|
153
|
Trikha P, Carson WE. Signaling pathways involved in MDSC regulation. Biochim Biophys Acta Rev Cancer 2014; 1846:55-65. [PMID: 24727385 DOI: 10.1016/j.bbcan.2014.04.003] [Citation(s) in RCA: 129] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 02/03/2014] [Accepted: 04/04/2014] [Indexed: 02/06/2023]
Abstract
The immune system has evolved mechanisms to protect the host from the deleterious effects of inflammation. The generation of immune suppressive cells like myeloid derived suppressor cells (MDSCs) that can counteract T cell responses represents one such strategy. There is an accumulation of immature myeloid cells or MDSCs in bone marrow (BM) and lymphoid organs under pathological conditions such as cancer. MDSCs represent a population of heterogeneous myeloid cells comprising of macrophages, granulocytes and dendritic cells that are at early stages of development. Although, the precise signaling pathways and molecular mechanisms that lead to MDSC generation and expansion in cancer remains to be elucidated. It is widely believed that perturbation of signaling pathways involved during normal hematopoietic and myeloid development under pathological conditions such as tumorogenesis contributes to the development of suppressive myeloid cells. In this review we discuss the role played by key signaling pathways such as PI3K, Ras, Jak/Stat and TGFb during myeloid development and how their deregulation under pathological conditions can lead to the generation of suppressive myeloid cells or MDSCs. Targeting these pathways should help in elucidating mechanisms that lead to the expansion of MDSCs in cancer and point to methods for eliminating these cells from the tumor microenvironment.
Collapse
Affiliation(s)
- Prashant Trikha
- Comprehensive Cancer Center, The Ohio State University, USA.
| | - William E Carson
- Comprehensive Cancer Center, The Ohio State University, USA; Department of Surgery, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
154
|
Occhipinti S, Sponton L, Rolla S, Caorsi C, Novarino A, Donadio M, Bustreo S, Satolli MA, Pecchioni C, Marchini C, Amici A, Cavallo F, Cappello P, Pierobon D, Novelli F, Giovarelli M. Chimeric rat/human HER2 efficiently circumvents HER2 tolerance in cancer patients. Clin Cancer Res 2014; 20:2910-21. [PMID: 24668647 DOI: 10.1158/1078-0432.ccr-13-2663] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Despite the great success of HER2 vaccine strategies in animal models, effective clinical results have not yet been obtained. We studied the feasibility of using DNA coding for chimeric rat/human HER2 as a tool to break the unresponsiveness of T cells from patients with HER2-overexpressing tumors (HER2-CP). EXPERIMENTAL DESIGN Dendritic cells (DCs) generated from patients with HER2-overexpressing breast (n = 28) and pancreatic (n = 16) cancer were transfected with DNA plasmids that express human HER2 or heterologous rat sequences in separate plasmids or as chimeric constructs encoding rat/human HER2 fusion proteins and used to activate autologous T cells. Activation was evaluated by IFN-γ ELISPOT assay, perforin expression, and ability to halt HER2+ tumor growth in vivo. RESULTS Specific sustained proliferation and IFN-γ production by CD4 and CD8 T cells from HER2-CP was observed after stimulation with autologous DCs transfected with chimeric rat/human HER2 plasmids. Instead, T cells from healthy donors (n = 22) could be easily stimulated with autologous DCs transfected with any human, rat, or chimeric rat/human HER2 plasmid. Chimeric HER2-transfected DCs from HER2-CP were also able to induce a sustained T-cell response that significantly hindered the in vivo growth of HER2(+) tumors. The efficacy of chimeric plasmids in overcoming tumor-induced T-cell dysfunction relies on their ability to circumvent suppressor effects exerted by regulatory T cells (Treg) and/or interleukin (IL)-10 and TGF-β1. CONCLUSIONS These results provide the proof of concept that chimeric rat/human HER2 plasmids can be used as effective vaccines for any HER2-CP with the advantage of being not limited to specific MHC. Clin Cancer Res; 20(11); 2910-21. ©2014 AACR.
Collapse
Affiliation(s)
- Sergio Occhipinti
- Authors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, ItalyAuthors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, Italy
| | - Laura Sponton
- Authors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, ItalyAuthors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, Italy
| | - Simona Rolla
- Authors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, ItalyAuthors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, Italy
| | - Cristiana Caorsi
- Authors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, Italy
| | - Anna Novarino
- Authors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, Italy
| | - Michela Donadio
- Authors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, Italy
| | - Sara Bustreo
- Authors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, Italy
| | - Maria Antonietta Satolli
- Authors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, Italy
| | - Carla Pecchioni
- Authors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, Italy
| | - Cristina Marchini
- Authors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, Italy
| | - Augusto Amici
- Authors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, Italy
| | - Federica Cavallo
- Authors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, Italy
| | - Paola Cappello
- Authors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, ItalyAuthors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, Italy
| | - Daniele Pierobon
- Authors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, ItalyAuthors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, Italy
| | - Francesco Novelli
- Authors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, ItalyAuthors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, Italy
| | - Mirella Giovarelli
- Authors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, ItalyAuthors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, Italy
| |
Collapse
|
155
|
Meyer C, Cagnon L, Costa-Nunes CM, Baumgaertner P, Montandon N, Leyvraz L, Michielin O, Romano E, Speiser DE. Frequencies of circulating MDSC correlate with clinical outcome of melanoma patients treated with ipilimumab. Cancer Immunol Immunother 2014; 63:247-57. [PMID: 24357148 PMCID: PMC11029062 DOI: 10.1007/s00262-013-1508-5] [Citation(s) in RCA: 454] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 12/05/2013] [Indexed: 12/18/2022]
Abstract
Metastatic melanoma has a poor prognosis with high resistance to chemotherapy and radiation. Recently, the anti-CTLA-4 antibody ipilimumab has demonstrated clinical efficacy, being the first agent to significantly prolong the overall survival of inoperable stage III/IV melanoma patients. A major aim of patient immune monitoring is the identification of biomarkers that predict clinical outcome. We studied circulating myeloid-derived suppressor cells (MDSC) in ipilimumab-treated patients to detect alterations in the myeloid cell compartment and possible correlations with clinical outcome. Lin(-) CD14(+) HLA-DR(-) monocytic MDSC were enriched in peripheral blood of melanoma patients compared to healthy donors (HD). Tumor resection did not significantly alter MDSC frequencies. During ipilimumab treatment, MDSC frequencies did not change significantly compared to baseline levels. We observed high inter-patient differences. MDSC frequencies in ipilimumab-treated patients were independent of baseline serum lactate dehydrogenase levels but tended to increase in patients with severe metastatic disease (M1c) compared to patients with metastases in skin or lymph nodes only (M1a), who had frequencies comparable to HD. Interestingly, clinical responders to ipilimumab therapy showed significantly less lin(-) CD14(+) HLA-DR(-) cells as compared to non-responders. The data suggest that the frequency of monocytic MDSC may be used as predictive marker of response, as low frequencies identify patients more likely benefitting from ipilimumab treatment. Prospective clinical trials assessing MDSC frequencies as potential biomarkers are warranted to validate these observations.
Collapse
Affiliation(s)
- Christiane Meyer
- Ludwig Center for Cancer Research of the University of Lausanne, Lausanne, Switzerland
| | - Laurène Cagnon
- Ludwig Center for Cancer Research of the University of Lausanne, Lausanne, Switzerland
| | - Carla M. Costa-Nunes
- Ludwig Center for Cancer Research of the University of Lausanne, Lausanne, Switzerland
| | - Petra Baumgaertner
- Ludwig Center for Cancer Research of the University of Lausanne, Lausanne, Switzerland
| | - Nicole Montandon
- Ludwig Center for Cancer Research of the University of Lausanne, Lausanne, Switzerland
| | - Loredana Leyvraz
- Ludwig Center for Cancer Research of the University of Lausanne, Lausanne, Switzerland
| | - Olivier Michielin
- Ludwig Center for Cancer Research of the University of Lausanne, Lausanne, Switzerland
- Service of Medical Oncology, Department of Oncology, Lausanne University Hospital Center (CHUV), Lausanne, Switzerland
| | - Emanuela Romano
- Service of Medical Oncology, Department of Oncology, Lausanne University Hospital Center (CHUV), Lausanne, Switzerland
| | - Daniel E. Speiser
- Ludwig Center for Cancer Research of the University of Lausanne, Lausanne, Switzerland
- Service of Medical Oncology, Department of Oncology, Lausanne University Hospital Center (CHUV), Lausanne, Switzerland
| |
Collapse
|
156
|
Tarhini AA, Edington H, Butterfield LH, Lin Y, Shuai Y, Tawbi H, Sander C, Yin Y, Holtzman M, Johnson J, Rao UNM, Kirkwood JM. Immune monitoring of the circulation and the tumor microenvironment in patients with regionally advanced melanoma receiving neoadjuvant ipilimumab. PLoS One 2014; 9:e87705. [PMID: 24498358 PMCID: PMC3912016 DOI: 10.1371/journal.pone.0087705] [Citation(s) in RCA: 249] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 01/02/2014] [Indexed: 11/21/2022] Open
Abstract
We evaluated neoadjuvant ipilimumab in patients with surgically operable regionally advanced melanoma in order to define markers of activity in the blood and tumor as assessed at baseline (before ipilimumab) and early on-treatment. Patients were treated with ipilimumab (10 mg/kg intravenously every 3 weeks ×2 doses) bracketing surgery. Tumor and blood biospecimens were obtained at baseline and at surgery. Flow cytometry and immunohistochemistry for select biomarkers were performed. Thirty five patients were enrolled; IIIB (3; N2b), IIIC (32; N2c, N3), IV (2). Worst toxicities included Grade 3 diarrhea/colitis (5; 14%), hepatitis (2; 6%), rash (1; 3%), elevated lipase (3; 9%). Median follow up was 18 months: among 33 evaluable patients, median progression free survival (PFS) was 11 months, 95% CI (6.2–19.2). There was a significant decrease in circulating myeloid derived suppressor cells (MDSC). Greater decrease in circulating monocyte gate MDSC Lin1−/HLA-DR−/CD33+/CD11b+ was associated with improved PFS (p = 0.03). There was a significant increase in circulating regulatory T cells (Treg; CD4+CD25hi+Foxp3+) that, unexpectedly, was associated with improved PFS (HR = 0.57; p = 0.034). Baseline evidence of fully activated type I CD4+ and CD8+ antigen-specific T cell immunity against cancer-testis (NY-ESO-1) and melanocytic lineage (MART-1, gp100) antigens was detected and was significantly potentiated after ipilimumab. In tumor, there was a significant increase in CD8+ T cells after ipilimumab (p = 0.02). Ipilimumab induced increased tumor infiltration by fully activated (CD69+) CD3+/CD4+ and CD3+/CD8+ T cells with evidence of induction/potentiation of memory T cells (CD45RO+). The change in Treg observed within the tumor showed an inverse relationship with clinical benefit and greater decrease in tumor MDSC subset Lin1−/HLA-DR−/CD33+/CD11b+ was associated with improved PFS at one year. Neoadjuvant evaluation revealed a significant immunomodulating role for ipilimumab on Treg, MDSC and effector T cells in the circulation and tumor microenvironment that warrants further pursuit in the quest for optimizing melanoma immunotherapy.
Collapse
Affiliation(s)
- Ahmad A. Tarhini
- Department of Medicine, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| | - Howard Edington
- Department of Surgery, West Penn Allegheny Health System, Pittsburgh, Pennsylvania, United States of America
| | - Lisa H. Butterfield
- Department of Medicine, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, United States of America
| | - Yan Lin
- Department of Biostatistics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, United States of America
| | - Yongli Shuai
- Department of Biostatistics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, United States of America
| | - Hussein Tawbi
- Department of Medicine, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, United States of America
| | - Cindy Sander
- Department of Medicine, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, United States of America
| | - Yan Yin
- Department of Medicine, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, United States of America
| | - Matthew Holtzman
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States of America
| | - Jonas Johnson
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Uma N. M. Rao
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - John M. Kirkwood
- Department of Medicine, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
157
|
Increased levels of granulocytic myeloid-derived suppressor cells in peripheral blood and tumour tissue of pancreatic cancer patients. J Immunol Res 2014; 2014:879897. [PMID: 24741628 PMCID: PMC3987936 DOI: 10.1155/2014/879897] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 12/01/2013] [Accepted: 12/03/2013] [Indexed: 12/15/2022] Open
Abstract
Pancreatic cancer (PC) often presents late with poor survival. While role of immunosuppressive cells in preclinical studies provided help to develop immunotherapeutic agents, these cells remain under investigation in PC. The aim of this study was to characterise the different subsets of myeloid-derived suppressor cells (MDSCs) and evaluate their level and function in the circulation and tissue of PC patients. Significant increases in circulating and tumour-infiltrating granulocytic (Lin-HLA-DR-CD33+CD11b+CD15+), but not monocytic (Lin-HLA-DR-CD14+), MDSCs were detected in PC patients when compared with healthy donors and patients with chronic pancreatitis. The circulating MDSCs from PC patients expressed arginase 1, which represents their functional state. Blood levels of MDSCs showed no association with PC stage or preoperative levels of tumour markers. These findings provide a first characterisation of the phenotype of different subsets of peripheral and local MDSCs in PC patients and suggest that the frequency and contribution of these cells are predominantly granulocytic. This information demonstrates that MDSCs play a role in pancreatic cancer and future large validation studies may help in the development of new immunotherapeutic strategies to inhibit or eliminate MDSC function.
Collapse
|
158
|
Mirghorbani M, Van Gool S, Rezaei N. Myeloid-derived suppressor cells in glioma. Expert Rev Neurother 2014; 13:1395-406. [DOI: 10.1586/14737175.2013.857603] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
159
|
Bueno V, Sant'Anna OA, Lord JM. Ageing and myeloid-derived suppressor cells: possible involvement in immunosenescence and age-related disease. AGE (DORDRECHT, NETHERLANDS) 2014; 36:9729. [PMID: 25399072 PMCID: PMC4233024 DOI: 10.1007/s11357-014-9729-x] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 11/04/2014] [Indexed: 05/02/2023]
Abstract
Infections, cancer and autoimmune diseases occur more frequently in the elderly, and although many factors contribute to this, the age-related remodelling of the immune system, termed immunosenescence, plays a major role. Over the last two decades, studies have evaluated the effect of ageing on both the adaptive and innate arms of the immune system and demonstrated compromised function in several cells including lymphocytes (naïve, effector and memory), regulatory T and B cells, monocytes, neutrophils and NK cells. In addition, a well-documented feature of ageing is the increase in systemic inflammatory status (inflammageing), with raised serum levels of IL6, TNFα and CRP as well as reduced IL10. Recently, myeloid-derived suppressor cells have been the focus of many reports as these cells show immunosuppressive properties and are present in higher frequency during infections, cancer and autoimmunity. Importantly, there have been publications showing increased numbers of myeloid-derived suppressor cells in aged mice and humans. In this review, we discuss the current literature on myeloid-derived suppressor cells, their possible role in altered immune function in the elderly, and whether it may be possible to manipulate these cells to alleviate age-related immune dysfunction.
Collapse
Affiliation(s)
- Valquiria Bueno
- Department of Microbiology Immunology and Parasitology, UNIFESP Federal University of São Paulo, São Paulo, Brazil,
| | | | | |
Collapse
|
160
|
Maeda A, Kawamura T, Ueno T, Usui N, Miyagawa S. Monocytic suppressor cells derived from human peripheral blood suppress xenogenic immune reactions. Xenotransplantation 2013; 21:46-56. [DOI: 10.1111/xen.12067] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Accepted: 09/16/2013] [Indexed: 11/29/2022]
Affiliation(s)
- Akira Maeda
- Department of Surgery; Osaka University Graduate School of Medicine; Osaka Japan
| | - Takuji Kawamura
- Department of Surgery; Osaka University Graduate School of Medicine; Osaka Japan
| | - Takehisa Ueno
- Department of Surgery; Osaka University Graduate School of Medicine; Osaka Japan
| | - Noriaki Usui
- Department of Surgery; Osaka University Graduate School of Medicine; Osaka Japan
| | - Shuji Miyagawa
- Department of Surgery; Osaka University Graduate School of Medicine; Osaka Japan
| |
Collapse
|
161
|
Weide B, Martens A, Zelba H, Stutz C, Derhovanessian E, Di Giacomo AM, Maio M, Sucker A, Schilling B, Schadendorf D, Büttner P, Garbe C, Pawelec G. Myeloid-derived suppressor cells predict survival of patients with advanced melanoma: comparison with regulatory T cells and NY-ESO-1- or melan-A-specific T cells. Clin Cancer Res 2013; 20:1601-9. [PMID: 24323899 DOI: 10.1158/1078-0432.ccr-13-2508] [Citation(s) in RCA: 213] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To analyze the prognostic relevance and relative impact of circulating myeloid-derived suppressor cells (MDSC) and regulatory T cells (Treg) compared with functional tumor antigen-specific T cells in patients with melanoma with distant metastasis. EXPERIMENTAL DESIGN The percentage of CD14(+)CD11b(+)HLA-DR(-/low) MDSCs, CD4(+)CD25(+)FoxP3(+) Tregs, and the presence of NY-ESO-1- or Melan-A-specific T cells was analyzed in 94 patients and validated in an additional cohort of 39 patients by flow cytometry. Univariate survival differences were calculated according to Kaplan-Meier and log-rank tests. Multivariate analyses were performed using Cox regression models. RESULTS NY-ESO-1-specific T cells, the M-category, and the frequency of MDSCs were associated with survival. The absence of NY-ESO-1-specific T cells and the M-category M1c independently increased the risk of death. In a second Cox model not considering results on antigen-specific T cells, a frequency of >11% MDSCs showed independent impact. Its association with survival was confirmed in the additional patient cohort. Median survival of patients with a lower frequency of MDSCs was 13 months versus 8 months for others (P < 0.001, combined cohorts). We observed a strong correlation between high levels of MDSCs and the absence of melanoma antigen-specific T cells implying a causal and clinically relevant interaction. No prognostic impact was observed for Tregs. CONCLUSIONS Circulating CD14(+)CD11b(+)HLA-DR(-/low) MDSCs have a negative impact on survival and inversely correlate with the presence of functional antigen-specific T cells in patients with advanced melanoma. Our findings provide a rationale to investigate MDSC-depleting strategies in the therapeutic setting especially in combination with vaccination or T-cell transfer approaches.
Collapse
Affiliation(s)
- Benjamin Weide
- Authors' Affiliations: Department of Dermatology, University Medical Center; Department of Internal Medicine II, Section for Transplantation Immunology and Immunohematology, University of Tübingen, Tübingen; German Cancer Research Center (DKFZ); German Cancer Consortium (DKTK), Heidelberg; Department of Dermatology, University Hospital, West German Cancer Center, University Duisburg-Essen, Essen, Germany; Department of Medical Oncology and Immunotherapy, University Hospital of Siena, Istituto Toscano Tumori, Italy; and Skin Cancer Research Group, School of Public Health, Tropical Medicine and Rehabilitation Sciences, James Cook University, Townsville, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
162
|
Abstract
T cells are the master regulators of adaptive immune responses and maintenance of their tolerance is critical to prevent autoimmunity. However, in the case of carcinogenesis, the tumor microenvironment aids T-cell tolerance, which contributes to uncontrolled tumor growth. Recently, there has been significant progress in understanding the intrinsic extracellular (positive and negative costimulatory molecules on APCs) and intracellular mechanisms (E3 ubiquitin ligases, transcriptional and epigenetic repressors), as well as extrinsic mechanisms (Tregs and tolerogenic dendritic cells) that are required for the implementation and maintenance of T-cell tolerance. Ultimately, understanding and manipulating T-cell tolerance will help to break the tolerance state in cancer.
Collapse
Affiliation(s)
- Roza Nurieva
- Department of Immunology & Center for Inflammation & Cancer, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Junmei Wang
- Department of Immunology & Center for Inflammation & Cancer, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Anupama Sahoo
- Department of Immunology & Center for Inflammation & Cancer, MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
163
|
Katoh H, Wang D, Daikoku T, Sun H, Dey SK, DuBois RN. CXCR2-expressing myeloid-derived suppressor cells are essential to promote colitis-associated tumorigenesis. Cancer Cell 2013; 24:631-44. [PMID: 24229710 PMCID: PMC3928012 DOI: 10.1016/j.ccr.2013.10.009] [Citation(s) in RCA: 393] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 09/09/2013] [Accepted: 10/03/2013] [Indexed: 12/21/2022]
Abstract
A large body of evidence indicates that chronic inflammation is one of several key risk factors for cancer initiation, progression, and metastasis. However, the underlying mechanisms responsible for the contribution of inflammation and inflammatory mediators to cancer remain elusive. Here, we present genetic evidence that loss of CXCR2 dramatically suppresses chronic colonic inflammation and colitis-associated tumorigenesis through inhibiting infiltration of myeloid-derived suppressor cells (MDSCs) into colonic mucosa and tumors in a mouse model of colitis-associated cancer. CXCR2 ligands were elevated in inflamed colonic mucosa and tumors and induced MDSC chemotaxis. Adoptive transfer of wild-type MDSCs into Cxcr2(-/-) mice restored AOM/DSS-induced tumor progression. MDSCs accelerated tumor growth by inhibiting CD8(+) T cell cytotoxic activity.
Collapse
Affiliation(s)
- Hiroshi Katoh
- Laboratory for Inflammation and Cancer, the Biodesign Institute at Arizona State University, Tempe, AZ 85287
| | - Dingzhi Wang
- Laboratory for Inflammation and Cancer, the Biodesign Institute at Arizona State University, Tempe, AZ 85287
| | - Takiko Daikoku
- Cincinnati Children’s Hospital Medical Center, University of Cincinnati, OH 45229
| | - Haiyan Sun
- Laboratory for Inflammation and Cancer, the Biodesign Institute at Arizona State University, Tempe, AZ 85287
| | - Sudhansu K. Dey
- Cincinnati Children’s Hospital Medical Center, University of Cincinnati, OH 45229
| | - Raymond N. DuBois
- Laboratory for Inflammation and Cancer, the Biodesign Institute at Arizona State University, Tempe, AZ 85287
- Department of Chemistry and Biology, Arizona State University, Tempe, AZ 85287
- Correspondence to: Raymond N. DuBois, MD. Ph.D., Executive Director of the Biodesign Institute at Arizona State University, PO Box 875001, 1001, S. McAllister Ave. Tempe, AZ 85287, Tel: 480-965-1228 and Fax: 480-727-9550,
| |
Collapse
|
164
|
Myeloid-derived suppressor cells are associated with disease progression and decreased overall survival in advanced-stage melanoma patients. Cancer Immunol Immunother 2013; 62:1711-22. [PMID: 24072401 DOI: 10.1007/s00262-013-1475-x] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 09/12/2013] [Indexed: 12/15/2022]
Abstract
Myeloid-derived suppressor cells are increased in the peripheral blood of advanced-stage cancer patients; however, no studies have shown a correlation of these immunosuppressive cells with clinical outcomes in melanoma patients. We characterized the frequency and suppressive function of multiple subsets of myeloid-derived suppressor cells in the peripheral blood of 34 patients with Stage IV melanoma, 20 patients with Stage I melanoma, and 15 healthy donors. The frequency of CD14+ MDSCs (Lin- CD11b+ HLA-DR- CD14+ CD33+) and CD14- MDSCs (Lin- CD11b+ HLA-DR- CD14- CD33+) was increased in the peripheral blood of Stage IV melanoma patients relative to healthy donors. The frequency of CD14+ and CD14- MDSCs correlated with each other and with the increased frequency of regulatory T cells, but not with classically defined monocytes. CD14- MDSCs isolated from the peripheral blood of Stage IV melanoma patients suppressed T cell activation more than those isolated from healthy donors, and the frequency of these cells correlated with disease progression and decreased overall survival. Our study provides the first evidence that the frequency of CD14- MDSCs negatively correlates with clinical outcomes in advanced-stage melanoma patients. These data indicate that suppressive MDSCs should be considered as targets for future immunotherapies.
Collapse
|
165
|
Waight JD, Netherby C, Hensen ML, Miller A, Hu Q, Liu S, Bogner PN, Farren MR, Lee KP, Liu K, Abrams SI. Myeloid-derived suppressor cell development is regulated by a STAT/IRF-8 axis. J Clin Invest 2013; 123:4464-78. [PMID: 24091328 DOI: 10.1172/jci68189] [Citation(s) in RCA: 271] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2012] [Accepted: 07/18/2013] [Indexed: 12/13/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) comprise immature myeloid populations produced in diverse pathologies, including neoplasia. Because MDSCs can impair antitumor immunity, these cells have emerged as a significant barrier to cancer therapy. Although much research has focused on how MDSCs promote tumor progression, it remains unclear how MDSCs develop and why the MDSC response is heavily granulocytic. Given that MDSCs are a manifestation of aberrant myelopoiesis, we hypothesized that MDSCs arise from perturbations in the regulation of interferon regulatory factor-8 (IRF-8), an integral transcriptional component of myeloid differentiation and lineage commitment. Overall, we demonstrated that (a) Irf8-deficient mice generated myeloid populations highly homologous to tumor-induced MDSCs with respect to phenotype, function, and gene expression profiles; (b) IRF-8 overexpression in mice attenuated MDSC accumulation and enhanced immunotherapeutic efficacy; (c) the MDSC-inducing factors G-CSF and GM-CSF facilitated IRF-8 downregulation via STAT3- and STAT5-dependent pathways; and (d) IRF-8 levels in MDSCs of breast cancer patients declined with increasing MDSC frequency, implicating IRF-8 as a negative regulator in human MDSC biology. Together, our results reveal a previously unrecognized role for IRF-8 expression in MDSC subset development, which may provide new avenues to target MDSCs in neoplasia.
Collapse
|
166
|
du Plessis N, Loebenberg L, Kriel M, von Groote-Bidlingmaier F, Ribechini E, Loxton AG, van Helden PD, Lutz MB, Walzl G. Increased Frequency of Myeloid-derived Suppressor Cells during Active Tuberculosis and after RecentMycobacterium tuberculosisInfection Suppresses T-Cell Function. Am J Respir Crit Care Med 2013; 188:724-32. [DOI: 10.1164/rccm.201302-0249oc] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
167
|
Kohanbash G, McKaveney K, Sakaki M, Ueda R, Mintz AH, Amankulor N, Fujita M, Ohlfest JR, Okada H. GM-CSF promotes the immunosuppressive activity of glioma-infiltrating myeloid cells through interleukin-4 receptor-α. Cancer Res 2013; 73:6413-23. [PMID: 24030977 DOI: 10.1158/0008-5472.can-12-4124] [Citation(s) in RCA: 145] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Malignant gliomas are lethal cancers in the brain and heavily infiltrated by myeloid cells. Interleukin-4 receptor-α (IL-4Rα) mediates the immunosuppressive functions of myeloid cells, and polymorphisms in the IL-4Rα gene are associated with altered glioma risk and prognosis. In this study, we sought to evaluate a hypothesized causal role for IL-4Rα and myeloid suppressor cells in glioma development. In both mouse de novo gliomas and human glioblastoma cases, IL-4Rα was upregulated on glioma-infiltrating myeloid cells but not in the periphery or in normal brain. Mice genetically deficient for IL-4Rα exhibited a slower growth of glioma associated with reduced production in the glioma microenvironment of arginase, a marker of myeloid suppressor cells, which is critical for their T-cell inhibitory function. Supporting this result, investigations using bone marrow-derived myeloid cells showed that IL-4Rα mediates IL-13-induced production of arginase. Furthermore, glioma-derived myeloid cells suppressed T-cell proliferation in an IL-4Rα-dependent manner, consistent with their identification as myeloid-derived suppressor cells (MDSC). Granulocyte macrophage colony-stimulating factor (GM-CSF) plays a central role for the induction of IL-4Rα expression on myeloid cells, and we found that GM-CSF is upregulated in both human and mouse glioma microenvironments compared with normal brain or peripheral blood samples. Together, our findings establish a GM-CSF-induced mechanism of immunosuppression in the glioma microenvironment via upregulation of IL-4Rα on MDSCs.
Collapse
Affiliation(s)
- Gary Kohanbash
- Authors' Affiliations: Department of Infectious Diseases and Microbiology, University of Pittsburgh Graduate School of Public Health; Brain Tumor Program, University of Pittsburgh Cancer Institute, Hillman Cancer Center; Departments of Neurological Surgery, Surgery, and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota
| | | | | | | | | | | | | | | | | |
Collapse
|
168
|
Activated human hepatic stellate cells induce myeloid derived suppressor cells from peripheral blood monocytes in a CD44-dependent fashion. J Hepatol 2013; 59:528-35. [PMID: 23665041 DOI: 10.1016/j.jhep.2013.04.033] [Citation(s) in RCA: 116] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 04/02/2013] [Accepted: 04/20/2013] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Myeloid derived suppressor cells (MDSCs) are a heterogeneous population of cells associated with the suppression of immunity. However, little is known about how or where MDSCs are induced and from which cells they originate. The liver is known for its immune regulatory functions. Here, we investigated the capacity of human hepatic stellate cells (HSCs) to transform peripheral blood monocytes into MDSCs. METHODS We cultured freshly isolated human monocytes from healthy donors on primary human HSCs or an HSC cell-line and characterized the phenotype and function of resulting CD14(+)HLA-DR(-/low) monocytes by flow cytometry, quantitative PCR, and functional assays. We analyzed the molecular mechanisms underlying the induction and function of the CD14(+)HLA-DR(-/low) cells by using blocking antibodies or knock-down technology. RESULTS Mature peripheral blood monocytes co-cultured with HSCs downregulated HLA-DR and developed a phenotypic and functional profile similar to MDSCs. Only activated but not freshly isolated HSCs were capable of inducing CD14(+)HLA-DR(-/low) cells. Such CD14(+)HLA-DR(-/low) monocyte-derived MDSCs suppressed T-cell proliferation in an arginase-1 dependent fashion. HSC-induced development of CD14(+)HLA-DR(-/low) monocyte-derived MDSCs was not mediated by soluble factors, but required physical interaction and was abrogated by blocking CD44. CONCLUSIONS Our study shows that activated human HSCs convert mature peripheral blood monocytes into MDSCs. As HSCs are activated during chronic inflammation, the subsequent local induction of MDSCs may prevent ensuing excessive liver injury. HSC-induced MDSCs functionally and phenotypically resemble those isolated from liver cancer patients. Thus, our data suggest that local generation of MDSCs by liver-resident HSCs may contribute to immune suppression during inflammation and cancer in the liver.
Collapse
|
169
|
Huang A, Zhang B, Wang B, Zhang F, Fan KX, Guo YJ. Increased CD14(+)HLA-DR (-/low) myeloid-derived suppressor cells correlate with extrathoracic metastasis and poor response to chemotherapy in non-small cell lung cancer patients. Cancer Immunol Immunother 2013; 62:1439-51. [PMID: 23760662 PMCID: PMC11028777 DOI: 10.1007/s00262-013-1450-6] [Citation(s) in RCA: 169] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 06/04/2013] [Indexed: 01/21/2023]
Abstract
Accumulating evidence has demonstrated that myeloid-derived suppressor cells (MDSCs), a heterogeneous population of cells, play an important role in the subversion, inhibition, and downregulation of the immune response to cancer. However, the characteristics of these cells, particularly clinical relevance, in malignant tumors remain unclear due to a lack of specific markers. In this study, we characterized peripheral CD14(+)HLA-DR(-/low) cells, a new human MDSC subpopulation, in 89 patients with non-small cell lung cancer (NSCLC). As expected, both frequency and absolute number of CD14(+)HLA-DR(-/low) cells were significantly increased in the peripheral blood of NSCLC patients compared with that of the healthy controls and indicated an association with metastasis, response to chemotherapy, and progression-free survival. These cells showed decreased expression of CD16 and CD86 compared with HLA-DR(+) monocytes. Unlike classical monocytes, these populations showed significantly decreased allostimulatory activity and showed the ability to inhibit autologous T cell proliferation and IFN-γ production in a cell-contact-dependent manner. Furthermore, we demonstrated that CD14(+)HLA-DR(-/low) cells expressed the NADPH oxidase component gp91(phox) and generated high level of reactive oxygen species (ROS). Moreover, inactivation of ROS reversed their immunosuppressive capacity on T cell response. These results prove, for the first time, the existence of ROS-producing CD14(+)HLA-DR(-/low) myeloid-derived suppressor cells in NSCLC patients, which mediate tumor immunosuppression and might thus represent a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Ang Huang
- Cancer Center, Chinese PLA General Hospital, Beijing, 100853 People’s Republic of China
- Department of Infectious Diseases, Beijing 302 Hospital, Beijing, 100039 People’s Republic of China
| | - Bo Zhang
- International Joint Cancer Institute, The Second Military Medical University, 800 Xiang Yin Road, New Building West 10-11th Floor, Shanghai, 200433 People’s Republic of China
| | - Bo Wang
- Cancer Center, Chinese PLA General Hospital, Beijing, 100853 People’s Republic of China
| | - Fan Zhang
- Cancer Center, Chinese PLA General Hospital, Beijing, 100853 People’s Republic of China
| | - Ke-Xing Fan
- Cancer Center, Chinese PLA General Hospital, Beijing, 100853 People’s Republic of China
- International Joint Cancer Institute, The Second Military Medical University, 800 Xiang Yin Road, New Building West 10-11th Floor, Shanghai, 200433 People’s Republic of China
| | - Ya-Jun Guo
- Cancer Center, Chinese PLA General Hospital, Beijing, 100853 People’s Republic of China
- International Joint Cancer Institute, The Second Military Medical University, 800 Xiang Yin Road, New Building West 10-11th Floor, Shanghai, 200433 People’s Republic of China
| |
Collapse
|
170
|
Obermajer N, Wong JL, Edwards RP, Chen K, Scott M, Khader S, Kolls JK, Odunsi K, Billiar TR, Kalinski P. Induction and stability of human Th17 cells require endogenous NOS2 and cGMP-dependent NO signaling. J Exp Med 2013; 210:1433-445. [PMID: 23797095 PMCID: PMC3698515 DOI: 10.1084/jem.20121277] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 05/30/2013] [Indexed: 01/24/2023] Open
Abstract
Nitric oxide (NO) is a ubiquitous mediator of inflammation and immunity, involved in the pathogenesis and control of infectious diseases, autoimmunity, and cancer. We observed that the expression of nitric oxide synthase-2 (NOS2/iNOS) positively correlates with Th17 responses in patients with ovarian cancer (OvCa). Although high concentrations of exogenous NO indiscriminately suppress the proliferation and differentiation of Th1, Th2, and Th17 cells, the physiological NO concentrations produced by patients’ myeloid-derived suppressor cells (MDSCs) support the development of RORγt(Rorc)+IL-23R⁺IL-17⁺ Th17 cells. Moreover, the development of Th17 cells from naive-, memory-, or tumor-infiltrating CD4+ T cells, driven by IL-1β/IL-6/IL-23/NO-producing MDSCs or by recombinant cytokines (IL-1β/IL-6/IL-23), is associated with the induction of endogenous NOS2 and NO production, and critically depends on NOS2 activity and the canonical cyclic guanosine monophosphate (cGMP)–cGMP-dependent protein kinase (cGK) pathway of NO signaling within CD4⁺ T cells. Inhibition of NOS2 or cGMP–cGK signaling abolishes the de novo induction of Th17 cells and selectively suppresses IL-17 production by established Th17 cells isolated from OvCa patients. Our data indicate that, apart from its previously recognized role as an effector mediator of Th17-associated inflammation, NO is also critically required for the induction and stability of human Th17 responses, providing new targets to manipulate Th17 responses in cancer, autoimmunity, and inflammatory diseases.
Collapse
|
171
|
Fujii W, Ashihara E, Hirai H, Nagahara H, Kajitani N, Fujioka K, Murakami K, Seno T, Yamamoto A, Ishino H, Kohno M, Maekawa T, Kawahito Y. Myeloid-derived suppressor cells play crucial roles in the regulation of mouse collagen-induced arthritis. THE JOURNAL OF IMMUNOLOGY 2013; 191:1073-81. [PMID: 23804709 DOI: 10.4049/jimmunol.1203535] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Myeloid-derived suppressor cells (MDSCs) are of myeloid origin and are able to suppress T cell responses. The role of MDSCs in autoimmune diseases remains controversial, and little is known about the function of MDSCs in autoimmune arthritis. In this study, we clarify that MDSCs play crucial roles in the regulation of proinflammatory immune response in a collagen-induced arthritis (CIA) mouse model. MDSCs accumulated in the spleens of mice with CIA when arthritis severity peaked. These MDSCs inhibited the proliferation of CD4(+) T cells and their differentiation into Th17 cells in vitro. Moreover, MDSCs inhibited the production of IFN-γ, IL-2, TNF-α, and IL-6 by CD4(+) T cells in vitro, whereas they promoted the production of IL-10. Adoptive transfer of MDSCs reduced the severity of CIA in vivo, which was accompanied by a decrease in the number of CD4(+) T cells and Th17 cells in the draining lymph nodes. However, depletion of MDSCs abrogated the spontaneous improvement of CIA. In conclusion, MDSCs in CIA suppress the progression of CIA by inhibiting the proinflammatory immune response of CD4(+) T cells. These observations suggest that MDSCs play crucial roles in the regulation of autoimmune arthritis, which could be exploited in new cell-based therapies for human rheumatoid arthritis.
Collapse
Affiliation(s)
- Wataru Fujii
- Inflammation and Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
172
|
Khaled YS, Ammori BJ, Elkord E. Myeloid-derived suppressor cells in cancer: recent progress and prospects. Immunol Cell Biol 2013; 91:493-502. [PMID: 23797066 DOI: 10.1038/icb.2013.29] [Citation(s) in RCA: 174] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2013] [Revised: 05/26/2013] [Accepted: 05/27/2013] [Indexed: 02/07/2023]
Abstract
Immunosuppressive cells, mainly myeloid-derived suppressor cells (MDSCs) and T regulatory cells, downregulate antitumour immunity and cancer immunotherapy. MDSCs are a heterogeneous group of immature myeloid cells that negatively regulate the immune responses during tumour progression, inflammation and infection. Whilst there have been extensive laboratory investigations aimed at characterising the MDSC subsets in cancer, there remains a significant gap in our understanding of their phenotypical and functional heterogeneity. In this article, we review data concerning the phenotypical and functional role of MDSCs in cancers. Importantly, we analyse the value of MDSCs as a prognostic factor in various clinical settings and the possible therapeutic approaches towards elimination of their immunosuppressive activity and enhancement of beneficial antitumour immune responses. MDSCs promote tumour immune evasion by inhibiting T-cell responses, as well as by supporting tumour progression. Accumulation of MDSCs is associated with the progression of human cancers, and their elimination was shown to improve anti-tumour immune responses. Phenotypical characterisation of MDSCs has been poorly investigated in many human cancers and lacks comprehensive clinicopathological correlation data. Although the need for effective therapeutic agents to eliminate the MDSC suppressive effect is immense, their role has been examined only in a few clinical settings.
Collapse
Affiliation(s)
- Yazan S Khaled
- 1] Institutes of Cancer and Cardiovascular Sciences, University of Manchester, Manchester, UK [2] Biomedical Research Centre, School of Environment and Life Sciences, University of Salford, Manchester, UK [3] Department of Upper Gastrointestinal Surgery, Salford Royal Foundation Trust, Manchester, UK [4] Department of Hepatobiliary Surgery, North Manchester General Hospital, Manchester, UK [5] Section of Translational Anaesthetic and Surgical Sciences, Leeds Institute of Molecular Medicine, Leeds, UK
| | | | | |
Collapse
|
173
|
Abstract
Fibrocytes are hematopoietic stem cell-derived fibroblast precursors that are implicated in chronic inflammation, fibrosis, and wound healing. Myeloid-derived suppressor cells (MDSCs) expand in cancer-bearing hosts and contribute to tumor immune evasion. They are typically described as CD11b⁺HLA-DR⁻ in humans. We report abnormal expansions of CD11b⁺HLA-DR⁺ myeloid cells in peripheral blood mononuclear fractions of subjects with metastatic pediatric sarcomas. Like classical fibrocytes, they display cell surface α smooth muscle actin, collagen I/V, and mediate angiogenesis. However, classical fibrocytes serve as antigen presenters and augment immune reactivity, whereas fibrocytes from cancer subjects suppressed anti-CD3-mediated T-cell proliferation, primarily via indoleamine oxidase (IDO). The degree of fibrocyte expansion observed in individual subjects directly correlated with the frequency of circulating GATA3⁺CD4⁺ cells (R = 0.80) and monocytes from healthy donors cultured with IL-4 differentiated into fibrocytes with the same phenotypic profile and immunosuppressive properties as those observed in patients with cancer. We thus describe a novel subset of cancer-induced myeloid-derived suppressor cells, which bear the phenotypic and functional hallmarks of fibrocytes but mediate immune suppression. These cells are likely expanded in response to Th2 immune deviation and may contribute to tumor progression via both immune evasion and angiogenesis.
Collapse
|
174
|
Vasquez-Dunddel D, Pan F, Zeng Q, Gorbounov M, Albesiano E, Fu J, Blosser RL, Tam AJ, Bruno T, Zhang H, Pardoll D, Kim Y. STAT3 regulates arginase-I in myeloid-derived suppressor cells from cancer patients. J Clin Invest 2013; 123:1580-9. [PMID: 23454751 DOI: 10.1172/jci60083] [Citation(s) in RCA: 428] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Accepted: 01/04/2013] [Indexed: 12/27/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSC) play a key immunosuppressive role in various types of cancer, including head and neck squamous cell carcinoma (HNSCC). In this study, we characterized CD14+HLA-DR(-/lo) cells sorted from the tumors, draining lymph nodes, and peripheral blood of HNSCC patients. CD14+HLA-DR(-/lo) cells were phenotyped as CD11b+, CD33+, CD34+, arginase-I+, and ROS+. In all 3 compartments, they suppressed autologous, antigen-independent T cell proliferation in a differential manner. The abundance of MDSC correlated with stage, but did not correlate with previous treatment with radiation or subsites of HNSCC. Interestingly, MDSC from all 3 compartments showed high phosphorylated STAT3 levels that correlated with arginase-I expression levels and activity. Stattic, a STAT3-specific inhibitor, and STAT3-targeted siRNA abrogated MDSC’s suppressive function. Inhibition of STAT3 signaling also resulted in decreased arginase-I activity. Analysis of the human arginase-I promoter region showed multiple STAT3-binding elements, and ChIP demonstrated that phosphorylated STAT3 binds to multiple sites in the arginase-I promoter. Finally, rescue of arginase-I activity after STAT3 blockade restored MDSC’s suppressive function. Taken together, these results demonstrate that the suppressive function of arginase-I in both infiltrating and circulating MDSC is a downstream target of activated STAT3.
Collapse
Affiliation(s)
- David Vasquez-Dunddel
- Department of Otolaryngology-Head and Neck Surgery, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, Maryland 21231, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
175
|
Schilling B, Sucker A, Griewank K, Zhao F, Weide B, Görgens A, Giebel B, Schadendorf D, Paschen A. Vemurafenib reverses immunosuppression by myeloid derived suppressor cells. Int J Cancer 2013; 133:1653-63. [PMID: 23526263 DOI: 10.1002/ijc.28168] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Accepted: 03/08/2013] [Indexed: 01/03/2023]
Abstract
Myeloid derived suppressor cells (MDSCs) suppress innate and adaptive immunity, thereby limiting anti-tumor immune responses in cancer patients. In patients with advanced melanoma, the phenotype and function of MDSCs remains controversial. In our study, we further explored two distinct subpopulations of MDSCs and investigated the impact of Vemurafenib on these cells. Flow cytometry analysis revealed that in comparison to healthy donors and patients with localized disease, PBMCs from patients with metastatic melanoma showed an increased frequency of CD14(+) HLA-DR(-/low) monocytic MDSCs (moMDSCs) and of a previously unrecognized population of CD14(-) CD66b(+) Arginase1(+) granulocytic MDSCs (grMDSCs). In vitro, both populations suppressed autologous T-cell proliferation, which was tested in CFSE-based proliferation assays. Vemurafenib treatment of melanoma patients reduced the frequency of both moMDSCs and grMDSCs. According to our in vivo finding, conditioned medium (CM) from Vemurafenib treated melanoma cells was less active in inducing moMDSCs in vitro than CM from untreated melanoma cells. In conclusion, patients with advanced melanoma show increased levels of moMDSCs, and of a population of CD14(-) CD66b(+) Arginase1(+) grMDSCs. Both MDSCs are distinct populations capable of suppressing autologous T-cell responses independently of each other. In vitro as well as in vivo, Vemurafenib inhibits the generation of human moMDSCs. Thus, Vemurafenib decreases immunosuppression in patients with advanced melanoma, indicating its potential as part of future immunotherapies.
Collapse
Affiliation(s)
- Bastian Schilling
- Department of Dermatology, University Hospital, University Duisburg-Essen, Essen, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
176
|
Differing patterns of circulating regulatory T cells and myeloid-derived suppressor cells in metastatic melanoma patients receiving anti-CTLA4 antibody and interferon-α or TLR-9 agonist and GM-CSF with peptide vaccination. J Immunother 2013; 35:702-10. [PMID: 23090079 DOI: 10.1097/cji.0b013e318272569b] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Changes in the biomarkers of host suppressor immune response were evaluated in patients with melanoma enrolled in 2 trials. Two similar cohorts of patients participating in the 2 studies were evaluated. The first (IFN/treme) tested interferon (IFN)-α2b and tremelimumab in metastatic melanoma and reported a response rate of 24%, 6.4 months median progression-free survival, and 21 months median overall survival. The second [toll-like receptor 9 (TLR)/GM] tested vaccination with MART-1, gp100, tyrosinase given with TLR-9 agonist and granulocyte-macrophage colony-stimulating factor and reported 9% response rate, median progression-free survival of 1.9 months, and median overall survival of 13.4 months. We monitored circulating T regulatory cells (T-reg) and myeloid-derived suppressor cells (MDSC) utilizing multicolor flow cytometry. In "IFN/treme," changes in circulating T-reg and MDSC were compared between baseline, day 29 (end of IFN-α induction) and day 85 (1 course). The CD4(+)CD25hi(+)CD39(+) T-reg percentage was increased most at day 85 (P = 0.018) and less significantly at day 29 (P = 0.09). There was a decrease in the percentage of MDSC populations taken in aggregate, which was most significant for monocytic MDSC (HLA-DR(+) low/CD14(+)) at day 29 (P < 0.0001) and day 85 (P = 0.001). In "TLR-9/GM," changes in T-reg and MDSC were compared between baseline and day 50 (4 vaccinations) and day 90 (8 vaccinations). There were no significant changes in T-reg or MDSC, except for a trend towards decreased (HLA-DR(+) low/CD14(+)) MDSC at day 50 (P = 0.07). Therefore, IFN/treme significantly downregulated MDSC suggesting a role on the significant clinical activity observed in this trial. T-reg findings suggest that IFN/treme induced clinically significant antitumor responses by inhibiting CTLA4 suppressive effects on T effectors, and less so by affecting T-reg.
Collapse
|
177
|
Eisenstein S, Coakley BA, Briley-Saebo K, Ma G, Chen HM, Meseck M, Ward S, Divino C, Woo S, Chen SH, Pan PY. Myeloid-derived suppressor cells as a vehicle for tumor-specific oncolytic viral therapy. Cancer Res 2013; 73:5003-15. [PMID: 23536556 DOI: 10.1158/0008-5472.can-12-1597] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
One of the several impediments to effective oncolytic virus therapy of cancer remains a lack of tumor-specific targeting. Myeloid-derived suppressor cells (MDSC) are immature myeloid cells induced by tumor factors in tumor-bearing hosts. The biodistribution kinetics of MDSC and other immune cell types in a murine hepatic colon cancer model was investigated through the use of tracking markers and MRI. MDSCs were superior to other immune cell types in preferential migration to tumors in comparison with other tissues. On the basis of this observation, we engineered a strain of vesicular stomatitis virus (VSV), an oncolytic rhabdovirus that bound MDSCs and used them as a delivery vehicle. Improving VSV-binding efficiency to MDSCs extended the long-term survival of mice bearing metastatic colon tumors compared with systemic administration of wild-type VSV alone. Survival was further extended by multiple injections of the engineered virus without significant toxicity. Notably, direct tumor killing was accentuated by promoting MDSC differentiation towards the classically activated M1-like phenotype. Our results offer a preclinical proof-of-concept for using MDSCs to facilitate and enhance the tumor-killing activity of tumor-targeted oncolytic therapeutics.
Collapse
Affiliation(s)
- Samuel Eisenstein
- Department of Oncological Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
178
|
Najjar YG, Finke JH. Clinical perspectives on targeting of myeloid derived suppressor cells in the treatment of cancer. Front Oncol 2013; 3:49. [PMID: 23508517 PMCID: PMC3597982 DOI: 10.3389/fonc.2013.00049] [Citation(s) in RCA: 121] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Accepted: 02/26/2013] [Indexed: 12/29/2022] Open
Abstract
Tumors escape immune recognition by several mechanisms, and induction of myeloid derived suppressor cells (MDSC) is thought to play a major role in tumor mediated immune evasion. MDSC arise from myeloid progenitor cells that do not differentiate into mature dendritic cells, granulocytes, or macrophages, and are characterized by the ability to suppress T cell and natural killer cell function. They are increased in patients with cancer including renal cell carcinoma (RCC), and their levels have been shown to correlate with prognosis and overall survival. Multiple methods of inhibiting MDSCs are currently under investigation. These can broadly be categorized into methods that (a) promote differentiation of MDSC into mature, non-suppressive cells (all trans retinoic acid, vitamin D), (b) decrease MDSC levels (sunitinib, gemcitabine, 5-FU, CDDO-Me), or (c) functionally inhibit MDSC (PDE-5 inhibitors, cyclooxygenase 2 inhibitors). Recently, several pre-clinical tumor models of combination therapy involving sunitinib plus vaccines and/or adoptive therapy have shown promise in MDSC inhibition and improved outcomes in the tumor bearing host. Current clinical trials are underway in RCC patients to assess not only the impact on clinical outcome, but how this combination can enhance anti-tumor immunity and reduce immune suppression. Decreasing immune suppression by MDSC in the cancer host may improve outcomes and prolong survival in this patient population.
Collapse
Affiliation(s)
- Yana G Najjar
- Department of Internal Medicine, The Cleveland Clinic Foundation Cleveland, OH, USA
| | | |
Collapse
|
179
|
Lindau D, Gielen P, Kroesen M, Wesseling P, Adema GJ. The immunosuppressive tumour network: myeloid-derived suppressor cells, regulatory T cells and natural killer T cells. Immunology 2013; 138:105-15. [PMID: 23216602 DOI: 10.1111/imm.12036] [Citation(s) in RCA: 630] [Impact Index Per Article: 52.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Revised: 10/25/2012] [Accepted: 10/29/2012] [Indexed: 12/12/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSC) and regulatory T (Treg) cells are major components of the immune suppressive tumour microenvironment (TME). Both cell types expand systematically in preclinical tumour models and promote T-cell dysfunction that in turn favours tumour progression. Clinical reports show a positive correlation between elevated levels of both suppressors and tumour burden. Recent studies further revealed that MDSCs can modulate the de novo development and induction of Treg cells. The overlapping target cell population of Treg cells and MDSCs is indicative for the importance and flexibility of immune suppression under pathological conditions. It also suggests the existence of common pathways that can be used for clinical interventions aiming to manipulate the TME. Elimination or reprogramming of the immune suppressive TME is one of the major current challenges in immunotherapy of cancer. Interestingly, recent findings suggest that natural killer T (NKT) cells can acquire the ability to convert immunosuppressive MDSCs into immunity-promoting antigen-presenting cells. Here we will review the cross-talk between MDSCs and other immune cells, focusing on Treg cells and NKT cells. We will consider its impact on basic and applied cancer research and discuss how targeting MDSCs may pave the way for future immunocombination therapies.
Collapse
Affiliation(s)
- Dennis Lindau
- Department of Tumour Immunology, Nijmegen Centre for Molecular Life Sciences, Nijmegen, the Netherlands
| | | | | | | | | |
Collapse
|
180
|
Vasquez-Dunddel D, Pan F, Zeng Q, Gorbounov M, Albesiano E, Fu J, Blosser RL, Tam AJ, Bruno T, Zhang H, Pardoll D, Kim Y. STAT3 regulates arginase-I in myeloid-derived suppressor cells from cancer patients. J Clin Invest 2013. [PMID: 23454751 DOI: 10.1172/jci60083.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSC) play a key immunosuppressive role in various types of cancer, including head and neck squamous cell carcinoma (HNSCC). In this study, we characterized CD14+HLA-DR(-/lo) cells sorted from the tumors, draining lymph nodes, and peripheral blood of HNSCC patients. CD14+HLA-DR(-/lo) cells were phenotyped as CD11b+, CD33+, CD34+, arginase-I+, and ROS+. In all 3 compartments, they suppressed autologous, antigen-independent T cell proliferation in a differential manner. The abundance of MDSC correlated with stage, but did not correlate with previous treatment with radiation or subsites of HNSCC. Interestingly, MDSC from all 3 compartments showed high phosphorylated STAT3 levels that correlated with arginase-I expression levels and activity. Stattic, a STAT3-specific inhibitor, and STAT3-targeted siRNA abrogated MDSC’s suppressive function. Inhibition of STAT3 signaling also resulted in decreased arginase-I activity. Analysis of the human arginase-I promoter region showed multiple STAT3-binding elements, and ChIP demonstrated that phosphorylated STAT3 binds to multiple sites in the arginase-I promoter. Finally, rescue of arginase-I activity after STAT3 blockade restored MDSC’s suppressive function. Taken together, these results demonstrate that the suppressive function of arginase-I in both infiltrating and circulating MDSC is a downstream target of activated STAT3.
Collapse
Affiliation(s)
- David Vasquez-Dunddel
- Department of Otolaryngology-Head and Neck Surgery, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, Maryland 21231, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
181
|
Yu J, Du W, Yan F, Wang Y, Li H, Cao S, Yu W, Shen C, Liu J, Ren X. Myeloid-derived suppressor cells suppress antitumor immune responses through IDO expression and correlate with lymph node metastasis in patients with breast cancer. THE JOURNAL OF IMMUNOLOGY 2013; 190:3783-97. [PMID: 23440412 DOI: 10.4049/jimmunol.1201449] [Citation(s) in RCA: 468] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Myeloid-derived suppressor cells (MDSCs) represent heterogeneous immunosuppressive cells in multiple cancer types and display potent immunosuppressive activity on T cells. We have shown the increased expression of IDO in breast cancer. Because IDO plays a pivotal role in immune tolerance via suppressing T cell function, the aim of this study was to investigate the expression of IDO in MDSCs in breast cancer and its role in MDSC-mediated inhibition of immune surveillance. The proportion of MDSCs with the phenotype of CD45(+)CD13(+)CD33(+)CD14(-)CD15(-) significantly increased in primary cancer tissues and patients' peripheral blood. IDO expression was significantly upregulated in MDSCs isolated from fresh breast cancer tissues (fresh MDSCs [fMDSCs]), which correlated with increased infiltration of Foxp3(+) regulatory T cells in tumors and lymph node metastasis in patients. fMDSCs inhibited IL-2 and anti-CD3/CD28 mAb-induced T cell amplification and Th1 polarization but stimulated apoptosis in T cells in an IDO-dependent manner. CD33(+) progenitors isolated from healthy donors' umbilical cord blood were cocultured with breast cancer cell line MDA-MB-231 cells to induce MDSCs. IDO expression was upregulated in induced MDSCs, which required phosphorylation of STAT3, but not STAT1. IDO was required for induced MDSCs' immunosuppressive activity on T cells, which was blocked by IDO inhibitor 1-methyl-L-tryptophan or STAT3 antagonist JSI-124. Consistently, increased STAT3 phosphorylation level was found in fMDSCs. Together, our findings suggest that STAT3-dependent IDO expression mediates immunosuppressive effects of MDSCs in breast cancer. Thus, inhibition of MDSC-induced T cell suppression by blocking IDO may represent a previously unrecognized mechanism underlying immunotherapy for breast cancer.
Collapse
Affiliation(s)
- Jinpu Yu
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
182
|
Immune suppression by neutrophils and granulocytic myeloid-derived suppressor cells: similarities and differences. Cell Mol Life Sci 2013; 70:3813-27. [PMID: 23423530 PMCID: PMC3781313 DOI: 10.1007/s00018-013-1286-4] [Citation(s) in RCA: 298] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 01/14/2013] [Accepted: 01/30/2013] [Indexed: 02/06/2023]
Abstract
Neutrophils are essential effector cells in the host defense against invading pathogens. Recently, novel neutrophil functions have emerged in addition to their classical anti-microbial role. One of these functions is the suppression of T cell responses. In this respect, neutrophils share similarities with granulocytic myeloid-derived suppressor cells (G-MDSCs). In this review, we will discuss the similarities and differences between neutrophils and G-MDSCs. Various types of G-MDSCs have been described, ranging from immature to mature cells shaping the immune response by different immune suppressive mechanisms. However, all types of G-MDSCs share distinct features of neutrophils, such as surface markers and morphology. We propose that G-MDSCs are heterogeneous and represent novel phenotypes of neutrophils, capable of suppressing the immune response. In this review, we will attempt to clarify the differences and similarities between neutrophils and G-MDSCs and attempt to facilitate further research.
Collapse
|
183
|
Abstract
Myeloid-derived suppressor cells (MDSCs) were initially reported as suppressor of the adaptive immune responses against cancer and other diseases. However, emerging evidence suggest that MDSCs may also support anti-tumor immune responses under certain conditions or may inhibit tumor growth. In the present mini-review, we suggest that such opposing functions of MDSCs are due to phenotypic plasticity of the myeloid cells, allowing them to produce a diverse form of morphology, physiological state, and function in response to environmental conditions. Therefore, they can be manipulated by means of immune modulators to overcome their immune suppressive function.
Collapse
Affiliation(s)
- Masoud H Manjili
- Department of Microbiology & Immunology, Virginia Commonwealth University, Massey Cancer Center, Richmond, Virginia 232989, USA.
| |
Collapse
|
184
|
Kohanbash G, Okada H. Myeloid-derived suppressor cells (MDSCs) in gliomas and glioma-development. Immunol Invest 2013; 41:658-79. [PMID: 23017140 DOI: 10.3109/08820139.2012.689591] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Myeloid derived suppressor cells (MDSCs) are a heterogeneous population of cells that inhibit anti-tumor immunity through a variety of mechanisms. Malignant gliomas are heavily infiltrated by myeloid cells, some of which appear to share biological functions of MDSCs. Our data with mouse de novo gliomas indicate critical roles of these cells in glioma development. This review summarizes the current understanding of MDSC biology in gliomas and discusses therapeutic interventions that can safely reverse the suppressive effects of MDSCs. The insight gained from these findings may lead to the development of novel immunotherapeutic strategies for gliomas.
Collapse
Affiliation(s)
- Gary Kohanbash
- Department of Infectious Diseases and Microbiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA 15261, USA
| | | |
Collapse
|
185
|
Solito S, Pinton L, Damuzzo V, Mandruzzato S. Highlights on molecular mechanisms of MDSC-mediated immune suppression: paving the way for new working hypotheses. Immunol Invest 2013; 41:722-37. [PMID: 23017143 DOI: 10.3109/08820139.2012.678023] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
MDSCs have been recognized in the last years as tolerogenic cells, potentially dangerous in the context of neoplasia, since they are able to induce tolerance to a variety of anti-tumor effectors, including CD4(+) and CD8(+) T cells. It is currently believed that the origin of MDSCs is due to an arrest of the myeloid differentiation process caused by tumor-secreted factors released in the tumor microenvironment that are able to exert an effect on myeloid progenitors, rendering them unable to terminally differentiate into dendritic cells, granulocytes and macrophages. As a consequence, these immature myeloid cells acquire suppressive activity through the activation of several mechanisms, controlled by different transcription factors. The lack of consensus about the phenotypical characterization of human MDSCs is the result of the existence of different MDSC subsets, most likely depending on the tumor in which they expand and on the tumor specific cytokine cocktail driving their activation. This, in turn, might also influence the mechanisms of MDSC-mediated immune suppression. In this review article we address the role of tumor-derived factors (TDFs) in MDSC-recruitment and activation, discuss the complex heterogeneity of MDSC phenotype and analyze the crosstalk between activated T cells and MDSCs.
Collapse
Affiliation(s)
- Samantha Solito
- Department of Surgery, Oncology and Gastroenterology, Oncology and Immunology Section, University of Padova, Padova, Italy
| | | | | | | |
Collapse
|
186
|
Increased level of myeloid-derived suppressor cells, programmed death receptor ligand 1/programmed death receptor 1, and soluble CD25 in Sokal high risk chronic myeloid leukemia. PLoS One 2013; 8:e55818. [PMID: 23383287 PMCID: PMC3561335 DOI: 10.1371/journal.pone.0055818] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 01/02/2013] [Indexed: 12/16/2022] Open
Abstract
Immunotherapy (eg interferon α) in combination with tyrosine kinase inhibitors is currently in clinical trials for treatment of chronic myeloid leukemia (CML). Cancer patients commonly have problems with so called immune escape mechanisms that may hamper immunotherapy. Hence, to study the function of the immune system in CML is of interest. In the present paper we have identified immune escape mechanisms in CML with focus on those that directly hamper T cells since these cells are important to control tumor progression. CML patient samples were investigated for the presence of myeloid-derived suppressor cells (MDSCs), expression of programmed death receptor ligand 1/programmed death receptor 1 (PD-L1/PD-1), arginase 1 and soluble CD25. MDSC levels were increased in samples from Sokal high risk patients (p<0,05) and the cells were present on both CD34 negative and CD34 positive cell populations. Furthermore, expression of the MDSC-associated molecule arginase 1, known to inhibit T cells, was increased in the patients (p = 0,0079). Myeloid cells upregulated PD-L1 (p<0,05) and the receptor PD-1 was present on T cells. However, PD-L1 blockade did not increase T cell proliferation but upregulated IL-2 secretion. Finally, soluble CD25 was increased in high risk patients (p<0,0001). In conclusion T cells in CML patients may be under the control of different immune escape mechanisms that could hamper the use of immunotherapy in these patients. These escape mechanisms should be monitored in trials to understand their importance and how to overcome the immune suppression.
Collapse
|
187
|
Wang L, Chang EWY, Wong SC, Ong SM, Chong DQY, Ling KL. Increased myeloid-derived suppressor cells in gastric cancer correlate with cancer stage and plasma S100A8/A9 proinflammatory proteins. THE JOURNAL OF IMMUNOLOGY 2012; 190:794-804. [PMID: 23248262 DOI: 10.4049/jimmunol.1202088] [Citation(s) in RCA: 201] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Immune dysfunction may contribute to tumor progression in gastric cancer (GC) patients. One mechanism of immune dysfunction is the suppression of T cell activation and impairment of the efficacy of cancer immunotherapy by myeloid-derived suppressor cells (MDSCs). We assessed the phenotype and immunosuppressive function of MDSCs in GC patients. We further investigated the role of S100A8/A9 in GC and the relationship between S100A8/A9 and MDSC function. Lastly, the effect of MDSCs on survival rates and its potential as a prognostic factor in GC patients were investigated. MDSCs from PBMCs of GC patients were identified by comparing the expression of specific surface markers with PBMCs from healthy individuals. The ability of MDSCs to suppress T lymphocyte response and the effect of S100A8/A9 and RAGE blocking were tested in vitro by (autologous) MLR. GC patients had significantly more MDSCs than healthy individuals. These MDSCs suppressed both T lymphocyte proliferation and IFN-γ production and had high arginase-I expression. Levels of S100A8/A9 in plasma were higher in GC patients compared with healthy individuals, and they correlated with MDSC levels in the blood. Blocking of S100A8/A9 itself and the S100A8/A9 receptor RAGE on MDSCs from GC patients abrogated T cell effector function. We found that high levels of MDSCs correlated with more advanced cancer stage and with reduced survival (p = 0.006). S100A8/A9 has been identified as a potential target to modulate antitumor immunity by reversing MDSC-mediated immunosuppression.
Collapse
Affiliation(s)
- Linda Wang
- Department of Gastroenterology, Singapore General Hospital, Singapore 169608, Republic of Singapore.
| | | | | | | | | | | |
Collapse
|
188
|
Jiao ZJ, Gao JJ, Hua SH, Chen DY, Wang WH, Wang H, Wang XH, Xu HX. Correlation between circulating myeloid-derived suppressor cells and Th17 cells in esophageal cancer. World J Gastroenterol 2012; 18:5454-61. [PMID: 23082063 PMCID: PMC3471115 DOI: 10.3748/wjg.v18.i38.5454] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2012] [Revised: 03/06/2012] [Accepted: 03/20/2012] [Indexed: 02/06/2023] Open
Abstract
AIM: To perform a comprehensive investigation into the potential correlation between circulating myeloid-derived suppressor cells (MDSCs) and Th17 cells in esophageal cancer (ECA).
METHODS: A total of 31 patients newly diagnosed with ECA and 26 healthy subjects were included in the current study. The frequencies of MDSCs and Th17 cells in peripheral blood were determined by flow cytometry. The mRNA expression of cytokines, arginase 1 (Arg1) and inducible NO synthase (iNOS) in peripheral blood mononuclear cells (PBMCs) and plasma Arg1 were assessed by real-time polymerase chain reaction and enzyme-linked immunosorbent assay, respectively.
RESULTS: There was an increased prevalence of MDSCs in the peripheral blood from ECA patients (15.21% ± 2.25%) when compared with healthy control (HC) (1.10% ± 0.12%, P < 0.0001). The plasma levels of Arg1 in ECA patients were significantly higher than those in HC (28.28 ± 4.10 ng/mL vs 9.57 ± 1.51 ng/mL, P = 0.0003). iNOS mRNA levels in the peripheral blood of ECA patients also showed a threefold increase compared with HC (P = 0.0162). The frequencies of Th17 cells (CD4+IL-17A+) were significantly elevated in ECA patients versus HC (3.50% ± 0.33% vs 1.82% ± 0.19%, P = 0.0001). Increased mRNA expression of IL-17 and ROR-γt was also observed in ECA patients compared with HC (P = 0.0041 and P = 0.0004, respectively), while the mRNA expression of IL-6 and tumor necrosis factor-α (TNF-α) showed significant decreases (P = 0.0049 and P < 0.0001, respectively). No obvious correlations were found between the frequencies of MDSCs and Th17 cells in the peripheral blood from ECA patients(r = -0.1725, P = 0.3534). Arg1 mRNA levels were positively correlated with levels of IL-6 (r = 0.6404, P = 0.0031) and TNF-α (r = 0.7646, P = 0.0001). Similarly, iNOS mRNA levels were also positively correlated with levels of IL-6 (r = 0.6782, P = 0.0007) and TNF-α (r = 0.7633, P < 0.0001).
CONCLUSION: This study reveals the relationship between circulating MDSCs and Th17 cells, which may lead to new immunotherapy approaches for ECA based on the associated metabolites and cytokines.
Collapse
|
189
|
Botti G, Cerrone M, Scognamiglio G, Anniciello A, Ascierto PA, Cantile M. Microenvironment and tumor progression of melanoma: New therapeutic prospectives. J Immunotoxicol 2012; 10:235-52. [DOI: 10.3109/1547691x.2012.723767] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
190
|
Abstract
A large body of evidence indicates that genetic mutations, epigenetic changes, chronic inflammation, diet, and lifestyle are key risk factors for colorectal cancer (CRC). Prevention of CRC has long been considered a plausible approach for the population and individuals at high risk for developing this disease. A significant effort has been made in the development of novel drugs for both prevention and treatment over the past two decades. This review highlights recent advances in our understanding of the role of nonsteroidal anti-inflammatory drugs in CRC prevention and adjuvant treatment. Moreover, we focus on the molecular mechanisms underlying the antitumor effects of these drugs in CRC. The knowledge of how anti-inflammatory agents inhibit cancer formation and progression may provide a rationale for the development of more effective chemopreventive and chemotherapeutic agents with less toxicity.
Collapse
Affiliation(s)
- Dingzhi Wang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030-4009, USA
| | | |
Collapse
|
191
|
Yang B, Wang X, Ren X. Amino acid metabolism related to immune tolerance by MDSCs. Int Rev Immunol 2012; 31:177-83. [PMID: 22587019 DOI: 10.3109/08830185.2012.679989] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Myeloid-derived suppressor cells (MDSCs) are present in most cancer patients. Due to their significant role in blocking immune responses, MDSCs are strategic obstacles to immunotherapy that require activation of the host's cell-mediated and innate immune responses. Following a brief description of the immunosuppressive MDSCs, the authors review the discovered mechanisms of amino acid metabolism that MDSCs use to suppress the activation of T cells. Given the heterogeneity of MDSCs and the variety of suppressive mechanisms employed by MDSCs, it is essential to understand which group of these cells and mechanisms are dominant.
Collapse
Affiliation(s)
- Baohong Yang
- Department of Biotherapy, Tianjin Cancer Institute and Hospital, Tianjin Medical University, China
| | | | | |
Collapse
|
192
|
Feng PH, Lee KY, Chang YL, Chan YF, Kuo LW, Lin TY, Chung FT, Kuo CS, Yu CT, Lin SM, Wang CH, Chou CL, Huang CD, Kuo HP. CD14(+)S100A9(+) monocytic myeloid-derived suppressor cells and their clinical relevance in non-small cell lung cancer. Am J Respir Crit Care Med 2012; 186:1025-36. [PMID: 22955317 DOI: 10.1164/rccm.201204-0636oc] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Myeloid-derived suppressor cells (MDSCs) are a heterogeneous family of myeloid cells that suppress T-cell immunity in tumor-bearing hosts. Their clinical relevance remains unclear. OBJECTIVES To identify subtypes of myeloid-derived suppressor cells in patients with non-small cell lung cancer (NSCLC) and their clinical relevance. METHODS CD11b(+)CD14(-) and CD11b(+)CD14(+) cells, determined and phenotyped by fluorescence-activated cell sorter analysis, in the peripheral blood mononuclear cells (PBMCs) of treatment-naive patients with advanced NSCLC were correlated with clinical data. T-cell activation in response to CD3/CD28 costimulation was determined by carboxy-fluorescein diacetate succinimidyl ester (CFSE) staining and ELISA analysis of IFN-γ. The percentage of CD11b(+)CD14(+)S100A9(+) cells in PBMCs was correlated with and tested as a predictor for treatment response in a cohort of patients prospectively receiving first-line cisplatin-based chemotherapy. MEASUREMENTS AND MAIN RESULTS Patients with NSCLC had a significantly higher ratio of CD11b(+)CD14(+) cells than healthy subjects, which was correlated with poor performance status and poor response to chemotherapy. The depletion of these cells in the PBMC reversed the suppression of CD8(+) and CD4(+) T cells. Isolated CD11b(+)CD14(+) cells suppressed CD8(+) T-cell proliferation and IFN-γ production, and the former effect was attenuated by the inducible nitric oxide synthase (iNOS) inhibitor aminoguanidine hydrochloride, arginase inhibitor N-hydroxy-nor-l-arginine (nor-NOHA), and blocking antibodies for IL-4Rα(+) and IL-10. CD11b(+)CD14(+) cells were monocyte-like, expressing CD33(+), CD15(-/low), IL-4Rα(+), and S100A9(+) and producing iNOS, arginase, and several cytokines. The ratio of S100A9(+) cells positively correlated with the suppressive ability of the CD11b(+)CD14(+) cells, was associated with poor response to chemotherapy, and predicted shorter progression-free survival. CONCLUSIONS CD14(+)S100A9(+) inflammatory monocytes in patients with NSCLC are a distinct subset of MDSCs, which suppress T cells by arginase, iNOS, and the IL-13/IL-4Rα axis. The amount of these inflammatory monocytes is associated with poor response to chemotherapy. Clinical trial registered with www.clinicaltrials.gov (NCT 01204307).
Collapse
Affiliation(s)
- Po-Hao Feng
- Pulmonary Research Center, Chang Gung Medical Foundation, Chang Gung University, 199, Tun-Hwa North Road, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
193
|
Schmid MC, Varner JA. Myeloid cells in tumor inflammation. Vasc Cell 2012; 4:14. [PMID: 22938502 PMCID: PMC3479419 DOI: 10.1186/2045-824x-4-14] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Accepted: 06/25/2012] [Indexed: 02/08/2023] Open
Abstract
Bone marrow derived myeloid cells progressively accumulate in tumors, where they establish an inflammatory microenvironment that is favorable for tumor growth and spread. These cells are comprised primarily of monocytic and granulocytic myeloid derived suppressor cells (MDSCs) or tumor-associated macrophages (TAMs), which are generally associated with a poor clinical outcome. MDSCs and TAMs promote tumor progression by stimulating immunosuppression, neovascularization, metastasis and resistance to anti-cancer therapy. Strategies to target the tumor-promoting functions of myeloid cells could provide substantial therapeutic benefit to cancer patients.
Collapse
Affiliation(s)
- Michael C Schmid
- Moores UCSD Cancer Center, University of California, San Diego, 3855 Health Sciences Drive, La Jolla, CA, 92093-0912, USA.
| | | |
Collapse
|
194
|
Immunity and malignant mesothelioma: From mesothelial cell damage to tumor development and immune response-based therapies. Cancer Lett 2012; 322:18-34. [DOI: 10.1016/j.canlet.2012.02.034] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Revised: 02/24/2012] [Accepted: 02/24/2012] [Indexed: 11/22/2022]
|
195
|
Dumitru CA, Moses K, Trellakis S, Lang S, Brandau S. Neutrophils and granulocytic myeloid-derived suppressor cells: immunophenotyping, cell biology and clinical relevance in human oncology. Cancer Immunol Immunother 2012; 61:1155-67. [PMID: 22692756 PMCID: PMC11028504 DOI: 10.1007/s00262-012-1294-5] [Citation(s) in RCA: 302] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Accepted: 05/28/2012] [Indexed: 02/08/2023]
Abstract
Accumulating evidence indicates that myeloid cells are critically involved in the pathophysiology of human cancers. In contrast to the well-characterized tumor-associated macrophages, the significance of granulocytes in cancer has only recently begun to emerge. A number of studies found increased numbers of neutrophil granulocytes and granulocytic myeloid-derived suppressor cells (GrMDSCs) both in the peripheral blood and in the tumor tissues of patients with different types of cancer. Most importantly, granulocytes have been linked to poor clinical outcome in cancer patients which suggests that these cells might have important tumor-promoting effects. In this review, we will address in detail the following major topics: (1) neutrophils and GrMDSCs in the peripheral blood of cancer patients-phenotype and functional changes; (2) neutrophils and GrMDSCs in the tumor tissue-potential mechanisms of tumor progression and (3) relevance of neutrophils and GrMDSCs for the clinical outcome of cancer patients. Furthermore, we will discuss the advantages and disadvantages of the current strategies used for identification and monitoring of human MDSCs. We propose a six-color immunophenotyping protocol that discriminates between monocytic MDSCs (MoMDSCs), two subsets of GrMDSCs and two subsets of immature myeloid cells in human cancer patients, thus, allowing for an improved characterization and understanding of these multifaceted cells.
Collapse
Affiliation(s)
- Claudia A. Dumitru
- Department of Otorhinolaryngology, University of Duisburg-Essen, Hufelandstraße 55, 45122 Essen, Germany
| | - Katrin Moses
- Department of Otorhinolaryngology, University of Duisburg-Essen, Hufelandstraße 55, 45122 Essen, Germany
| | - Sokratis Trellakis
- Department of Otorhinolaryngology, University of Duisburg-Essen, Hufelandstraße 55, 45122 Essen, Germany
| | - Stephan Lang
- Department of Otorhinolaryngology, University of Duisburg-Essen, Hufelandstraße 55, 45122 Essen, Germany
| | - Sven Brandau
- Department of Otorhinolaryngology, University of Duisburg-Essen, Hufelandstraße 55, 45122 Essen, Germany
| |
Collapse
|
196
|
Sinha P, Parker KH, Horn L, Ostrand-Rosenberg S. Tumor-induced myeloid-derived suppressor cell function is independent of IFN-γ and IL-4Rα. Eur J Immunol 2012; 42:2052-9. [PMID: 22673957 PMCID: PMC3673533 DOI: 10.1002/eji.201142230] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Myeloid-derived suppressor cells (MDSCs) are present in most cancer patients and experimental animals where they exert a profound immune suppression and are a significant obstacle to immunotherapy. IFN-γ and IL-4 receptor alpha (IL-4Rα) have been implicated as essential molecules for MDSC development and immunosuppressive function. If IFN-γ and IL-4Rα are critical regulators of MDSCs, then they are potential targets for preventing MDSC accumulation or inhibiting MDSC function. Because data supporting a role for IFN-γ and IL-4Rα are not definitive, we have examined MDSCs induced in IFN-γ-deficient, IFN-γR-deficient, and IL-4Rα-deficient mice carrying three C57BL/6-derived (B16 melanoma, MC38 colon carcinoma, and 3LL lung adenocarcinoma), and three BALB/c-derived (4T1 and TS/A mammary carcinomas, and CT26 colon carcinoma) tumors. We report that although MDSCs express functional IFN-γR and IL-4Rα, and have the potential to signal through the STAT1 and STAT6 pathways, respectively, neither IFN-γ nor IL-4Rα impacts the phenotype, accumulation, or T-cell suppressive potency of MDSCs, although IFN-γ and IL-4Rα modestly alter MDSC-macrophage IL-10 crosstalk. Therefore, neither IFN-γ nor IL-4Rα is a key regulator of MDSCs and targeting these molecules is unlikely to significantly alter MDSC accumulation or function.
Collapse
Affiliation(s)
- Pratima Sinha
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, MD 21250, USA
| | | | | | | |
Collapse
|
197
|
Chronic progressive HIV-1 infection is associated with elevated levels of myeloid-derived suppressor cells. AIDS 2012; 26:F31-7. [PMID: 22526518 DOI: 10.1097/qad.0b013e328354b43f] [Citation(s) in RCA: 127] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVES Myeloid-derived suppressor cells (MDSCs) have been described as suppressors of T-cell functions in many tumor models. However, MDSC in HIV-1 infection have not been studied to date. As impaired T-cell function is a hallmark of chronic progressive HIV-1 infection, we hypothesized that MDSC also play a role here. METHODS Surface staining and flow cytometry analysis were performed on freshly isolated peripheral blood mononuclear cells (PBMC) of HIV-infected individuals and compared to healthy controls and individuals with lung carcinoma. MDSC of late-stage HIV-infected individuals were isolated using magnetic beads and cocultured with the respective CD8 T cells for evaluation of proliferative capacity. RESULTS We found that chronically HIV-infected HAART-naive individuals had significantly higher CD11bCD14CD33CD15 MDSC levels than healthy controls (P = 0.01). MDSC frequencies showed a positive correlation with viral load (r = 0.24, P = 0.0002) and a negative correlation with CD4 cell count (r = 0.29, P < 0.0001). Initiation of HAART led to a rapid drop in MDSC levels. MDSC from HIV-infected progressors restricted the proliferative capacity of CD8 T cells from healthy donors and of Gag/Nef-specific CD8 T cells from HIV-controllers in vitro. Furthermore, CD11bCD14CD33CD15 MDSC induced the expansion of CD4CD25FoxP3 regulatory T cells when coincubated with PBMC from controllers in vitro. CONCLUSION We conclude that chronic uncontrolled HIV-infection is associated with elevated levels of MDSC, which potentially contribute to the impaired T-cell responses characteristic for the progressive disease stage.
Collapse
|
198
|
Multipeptide immune response to cancer vaccine IMA901 after single-dose cyclophosphamide associates with longer patient survival. Nat Med 2012; 18:1254-61. [PMID: 22842478 DOI: 10.1038/nm.2883] [Citation(s) in RCA: 625] [Impact Index Per Article: 48.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Accepted: 06/20/2012] [Indexed: 12/13/2022]
Abstract
IMA901 is the first therapeutic vaccine for renal cell cancer (RCC) consisting of multiple tumor-associated peptides (TUMAPs) confirmed to be naturally presented in human cancer tissue. We treated a total of 96 human leukocyte antigen A (HLA-A)*02(+) subjects with advanced RCC with IMA901 in two consecutive studies. In the phase 1 study, the T cell responses of the patients to multiple TUMAPs were associated with better disease control and lower numbers of prevaccine forkhead box P3 (FOXP3)(+) regulatory T (T(reg)) cells. The randomized phase 2 trial showed that a single dose of cyclophosphamide reduced the number of T(reg) cells and confirmed that immune responses to multiple TUMAPs were associated with longer overall survival. Furthermore, among six predefined populations of myeloid-derived suppressor cells, two were prognostic for overall survival, and among over 300 serum biomarkers, we identified apolipoprotein A-I (APOA1) and chemokine (C-C motif) ligand 17 (CCL17) as being predictive for both immune response to IMA901 and overall survival. A randomized phase 3 study to determine the clinical benefit of treatment with IMA901 is ongoing.
Collapse
|
199
|
Zhao F, Hoechst B, Duffy A, Gamrekelashvili J, Fioravanti S, Manns MP, Greten TF, Korangy F. S100A9 a new marker for monocytic human myeloid-derived suppressor cells. Immunology 2012; 136:176-83. [PMID: 22304731 DOI: 10.1111/j.1365-2567.2012.03566.x] [Citation(s) in RCA: 171] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSC) are a heterogeneous population of cells that negatively regulate the immune response during tumour progression, inflammation and infection. Only limited data are available on human MDSC because of the lack of specific markers. We have identified members of the S100 protein family-S100A8, S100A9 and S100A12 - specifically expressed in CD14(+) HLA-DR(-/low) MDSC. S100A9 staining in combination with anti-CD14 could be used to identify MDSC in whole blood from patients with colon cancer. An increase in the population of CD14(+) S100A9(high) MDSC was observed in the peripheral blood from colon cancer patients in comparison with healthy controls. Finally, nitric oxide synthase expression, a hallmark of MDSC, was induced in CD14(+) S100A9(high) upon lipopolysaccharide/interferon-γ stimulation. We propose S100 proteins as useful markers for the analysis and further characterization of human MDSC.
Collapse
Affiliation(s)
- Fei Zhao
- National Institutes of Health, National Cancer Institute, Medical Oncology Branch, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
200
|
Poschke I, Kiessling R. On the armament and appearances of human myeloid-derived suppressor cells. Clin Immunol 2012; 144:250-68. [PMID: 22858650 DOI: 10.1016/j.clim.2012.06.003] [Citation(s) in RCA: 149] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Revised: 06/10/2012] [Accepted: 06/12/2012] [Indexed: 02/08/2023]
Abstract
Myeloid-derived suppressor cells (MDSC) have frequently been observed in patients with cancer. This heterogeneous population of myeloid cells can exert potent suppression of lymphocyte function and thereby poses a significant hurdle to natural or therapeutically induced anti-tumor immunity. On the other hand, the natural function of MDSC is not yet well understood and their role in infection, inflammation and autoimmune disease is still puzzling. Understanding MDSC biology will provide the tools necessary for therapeutic targeting of this population, but also permit exploitation of their strong tolerogenic function in the treatment of inflammatory conditions and the prevention of graft rejection.
Collapse
Affiliation(s)
- Isabel Poschke
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.
| | | |
Collapse
|