51
|
Angell TE, Lechner MG, Smith AM, Martin SE, Groshen SG, Maceri DR, Singer PA, Epstein AL. Circulating Myeloid-Derived Suppressor Cells Predict Differentiated Thyroid Cancer Diagnosis and Extent. Thyroid 2016; 26:381-9. [PMID: 26756227 PMCID: PMC4790214 DOI: 10.1089/thy.2015.0289] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND Establishing the preoperative diagnosis and long-term prognosis of differentiated thyroid cancer (DTC) remain challenging in some patients. Myeloid-derived suppressor cells (MDSC) are tumor-induced cells mediating immune tolerance that are detectable in the peripheral blood of cancer patients. The authors previously developed a novel clinical assay to detect the phenotypes of two human MDSC subsets in peripheral blood, and hypothesize that higher MDSC levels measured by this assay correlate positively with both malignancy and worse patient outcomes. METHODS A prospective observational pilot study was performed of patients undergoing thyroidectomy for a solitary thyroid nodule. The presence of a thyroid nodule >1 cm was confirmed sonographically, and fine-needle aspiration biopsy performed prior to surgery in all cases. Peripheral blood collected preoperatively was analyzed using a novel flow cytometry-based immunoassay to detect and quantify two subsets of human MDSC. Circulating MDSC levels were compared by histopathologic diagnosis, stage, and presence of persistent disease after treatment. RESULTS Of 50 patients included in this study, MDSC measurement was successful in 47 (94%). One patient was found to have a concurrent cancer, leaving 46 patients for primary analysis. The cytologic diagnoses were benign in five (10.8%), atypia or follicular lesion of undetermined significance in five (10.8%), suspicious for follicular neoplasm in five (10.8%), suspicious for malignant in three (6.5%), and malignant in 28 (60.1%) of the 46 nodules. Final histopathology was benign in 11 (24%) and DTC in 35 (76%), encompassing 34 PTC cases and one follicular thyroid carcinoma. Mean percentages of CD11b(+)HLA-DR(low)HIF1a(+) MDSC (CD11b(+)MDSC) were 14.0 ± 6.2% and 7.9 ± 3.6% in DTC versus benign nodules, respectively (p < 0.005). A cutoff of 12% yielded a specificity of 0.91, a sensitivity of 0.72, and a likelihood ratio of 7.9. Mean CD11b(+)MDSC levels increased linearly with higher TNM stage (p < 0.01), and were 19.4 ± 5.4 in patients with persistent cancer after surgery compared with 13.2 ± 6.8 in those without evidence of disease (p < 0.05). CONCLUSION MDSC measurement using this flow cytometry-based assay represents a novel approach for preoperatively assessing malignancy risk and cancer extent in patients with thyroid nodules. While further validation is needed, these data suggest that MDSC assessment may serve as a useful adjunct when cytology is indeterminate, and predict tumor stage and recurrence risk in cases of thyroid cancer.
Collapse
Affiliation(s)
- Trevor E. Angell
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital, Boston, Massachusetts
| | - Melissa G. Lechner
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Alison M. Smith
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Sue E. Martin
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Susan G. Groshen
- Department of Biostatistics, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Dennis R. Maceri
- Department of Otolaryngology, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Peter A. Singer
- Department of Endocrinology, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Alan L. Epstein
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California
| |
Collapse
|
52
|
Tagliamonte M, Petrizzo A, Napolitano M, Luciano A, Rea D, Barbieri A, Arra C, Maiolino P, Tornesello M, Ciliberto G, Buonaguro FM, Buonaguro L. A novel multi-drug metronomic chemotherapy significantly delays tumor growth in mice. J Transl Med 2016; 14:58. [PMID: 26911136 PMCID: PMC4766679 DOI: 10.1186/s12967-016-0812-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 02/09/2016] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The tumor immunosuppressive microenvironment represents a major obstacle to an effective tumor-specific cellular immune response. METHODS In the present study, the counterbalance effect of a novel metronomic chemotherapy protocol on such an immunosuppressive microenvironment was evaluated in a mouse model upon sub-cutaneous ectopic implantation of B16 melanoma cells. The chemotherapy consisted of a novel multi-drug cocktail including taxanes and alkylating agents, administered in a daily metronomic fashion. The newly designed strategy was shown to be safe, well tolerated and significantly efficacious. RESULTS Treated animals showed a remarkable delay in tumor growth and prolonged survival as compared to control group. Such an effect was directly correlated with CD4(+) T cell reduction and CD8(+) T cell increase. Furthermore, a significant reduction in the percentage of both CD25(+)FoxP3(+) and CD25(+)CD127(low) regulatory T cell population was found both in the spleens and in the tumor lesions. Finally, the metronomic chemotherapy induced an intrinsic CD8(+) T cell response specific to B16 naturally expressed Trp2 TAA. CONCLUSION The novel multi-drug daily metronomic chemotherapy evaluated in the present study was very effective in counterbalancing the immunosuppressive tumor microenvironment. Consequently, the intrinsic anti-tumor T cell immunity could exert its function, targeting specific TAA and significantly containing tumor growth. Overall, the results show that this represents a promising adjuvant approach to significantly enhance efficacy of intrinsic or vaccine-elicited tumor-specific cellular immunity.
Collapse
Affiliation(s)
- Maria Tagliamonte
- Laboratory of Molecular Biology and Viral Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione Pascale" - IRCCS, Naples, Italy.
| | - Annacarmen Petrizzo
- Laboratory of Molecular Biology and Viral Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione Pascale" - IRCCS, Naples, Italy.
| | - Maria Napolitano
- Laboratory of Clinical Immunology, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione Pascale" - IRCCS, Naples, Italy.
| | - Antonio Luciano
- Animal Facility, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione Pascale" - IRCCS, Naples, Italy.
| | - Domenica Rea
- Animal Facility, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione Pascale" - IRCCS, Naples, Italy.
| | - Antonio Barbieri
- Animal Facility, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione Pascale" - IRCCS, Naples, Italy.
| | - Claudio Arra
- Animal Facility, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione Pascale" - IRCCS, Naples, Italy.
| | - Piera Maiolino
- Pharmacy Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione Pascale" - IRCCS, Naples, Italy.
| | - Marialina Tornesello
- Laboratory of Molecular Biology and Viral Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione Pascale" - IRCCS, Naples, Italy.
| | - Gennaro Ciliberto
- Scientific Direction, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione Pascale" - IRCCS, Naples, Italy.
| | - Franco M Buonaguro
- Laboratory of Molecular Biology and Viral Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione Pascale" - IRCCS, Naples, Italy.
| | - Luigi Buonaguro
- Laboratory of Molecular Biology and Viral Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione Pascale" - IRCCS, Naples, Italy.
| |
Collapse
|
53
|
Dubeykovskaya Z, Si Y, Chen X, Worthley DL, Renz BW, Urbanska AM, Hayakawa Y, Xu T, Westphalen CB, Dubeykovskiy A, Chen D, Friedman RA, Asfaha S, Nagar K, Tailor Y, Muthupalani S, Fox JG, Kitajewski J, Wang TC. Neural innervation stimulates splenic TFF2 to arrest myeloid cell expansion and cancer. Nat Commun 2016; 7:10517. [PMID: 26841680 PMCID: PMC4742920 DOI: 10.1038/ncomms10517] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 12/16/2015] [Indexed: 12/31/2022] Open
Abstract
CD11b+Gr-1+ myeloid-derived suppressor cells (MDSCs) expand in the spleen during cancer and promote progression through suppression of cytotoxic T cells. An anti-inflammatory reflex arc involving the vagus nerve and memory T cells is necessary for resolution of acute inflammation. Failure of this neural circuit could promote procarcinogenic inflammation and altered tumour immunity. Here we show that splenic TFF2, a secreted anti-inflammatory peptide, is released by vagally modulated memory T cells to suppress the expansion of MDSCs through CXCR4. Splenic denervation interrupts the anti-inflammatory neural arc, resulting in the expansion of MDSCs and colorectal cancer. Deletion of Tff2 recapitulates splenic denervation to promote carcinogenesis. Colorectal carcinogenesis could be suppressed through transgenic overexpression of TFF2, adenoviral transfer of TFF2 or transplantation of TFF2-expressing bone marrow. TFF2 is important to the anti-inflammatory reflex arc and plays an essential role in arresting MDSC proliferation. TFF2 offers a potential approach to prevent and to treat cancer. During colorectal inflammation and cancer, myeloid cells accumulate in the spleen and suppress the host immunity response. In this study, the authors use a mouse model of colitis to demonstrate that upon vagus stimulation splenic memory T cells release TFF2, which suppresses the expansion of myeloid cells and cancer progression.
Collapse
Affiliation(s)
- Zina Dubeykovskaya
- Division of Digestive and Liver Disease, Department of Medicine and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York 10032, USA
| | - Yiling Si
- Division of Digestive and Liver Disease, Department of Medicine and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York 10032, USA
| | - Xiaowei Chen
- Division of Digestive and Liver Disease, Department of Medicine and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York 10032, USA
| | - Daniel L Worthley
- Division of Digestive and Liver Disease, Department of Medicine and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York 10032, USA
| | - Bernhard W Renz
- Division of Digestive and Liver Disease, Department of Medicine and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York 10032, USA.,Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, Hospital of the University of Munich, 81377 Munich, Germany
| | - Aleksandra M Urbanska
- Division of Digestive and Liver Disease, Department of Medicine and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York 10032, USA
| | - Yoku Hayakawa
- Division of Digestive and Liver Disease, Department of Medicine and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York 10032, USA
| | - Ting Xu
- Division of Digestive and Liver Disease, Department of Medicine and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York 10032, USA
| | - C Benedikt Westphalen
- Division of Digestive and Liver Disease, Department of Medicine and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York 10032, USA
| | - Alexander Dubeykovskiy
- Division of Digestive and Liver Disease, Department of Medicine and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York 10032, USA
| | - Duan Chen
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Pb 8905, N-7491 Trondheim, Norway
| | - Richard A Friedman
- Department of Biomedical Informatics, Irving Cancer Research Center, Columbia University, New York, New York 10032, USA
| | - Samuel Asfaha
- Division of Digestive and Liver Disease, Department of Medicine and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York 10032, USA
| | - Karan Nagar
- Division of Digestive and Liver Disease, Department of Medicine and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York 10032, USA
| | - Yagnesh Tailor
- Division of Digestive and Liver Disease, Department of Medicine and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York 10032, USA
| | - Sureshkumar Muthupalani
- Department of Biological Engineering, Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - James G Fox
- Department of Biological Engineering, Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Jan Kitajewski
- Department of Pathology and Cell Biology, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York 10032, USA
| | - Timothy C Wang
- Division of Digestive and Liver Disease, Department of Medicine and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York 10032, USA
| |
Collapse
|
54
|
Bartmann C, Junker M, Segerer SE, Häusler SF, Krockenberger M, Kämmerer U. CD33(+) /HLA-DR(neg) and CD33(+) /HLA-DR(+/-) Cells: Rare Populations in the Human Decidua with Characteristics of MDSC. Am J Reprod Immunol 2016; 75:539-56. [PMID: 26840716 DOI: 10.1111/aji.12492] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 12/28/2015] [Indexed: 12/31/2022] Open
Abstract
PROBLEM Human pregnancy needs a remarkable local immune tolerance toward the conceptus. Myeloid-derived suppressor cells (MDSC) are important players promoting cancer initiation and progression by suppressing T-cell functions and thus inducing immune tolerance. Therefore, MDSC were expected within decidua. METHODS Subpopulations of CD33(+) immune cells were isolated from human early pregnancy decidua and characterized phenotypically and functionally by microscopy, FACS analysis, RT-PCR, Western blotting and in the coculture with T cells. RESULTS Decidua harbors CD33(+) /HLA-DR(neg) and CD33(+) /HLA-DR(+/-) cells which both express arginase, iNOS and IDO and a typical cytokine profile. Both subtypes potently suppress T-cell proliferation and therefore fulfill the criteria of MDSC. CONCLUSION We characterized a new population of CD33(+) /HLA-DR(neg) and CD33(+) /HLA-DR(+/-) cells in human early pregnancy decidua with properties of classical MDSC and thus potentially being an important player in immune tolerance in pregnancy.
Collapse
Affiliation(s)
- Catharina Bartmann
- Department of OB/Gyn, University Hospital of Würzburg, Würzburg, Germany
| | - Markus Junker
- Department of OB/Gyn, University Hospital of Würzburg, Würzburg, Germany
| | | | | | | | - Ulrike Kämmerer
- Department of OB/Gyn, University Hospital of Würzburg, Würzburg, Germany
| |
Collapse
|
55
|
Zhao AM, Xu HJ, Kang XM, Zhao AM, Lu LM. New insights into myeloid-derived suppressor cells and their roles in feto-maternal immune cross-talk. J Reprod Immunol 2016; 113:35-41. [DOI: 10.1016/j.jri.2015.11.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 10/23/2015] [Accepted: 11/04/2015] [Indexed: 12/20/2022]
|
56
|
Hong HJ, Lim HX, Song JH, Lee A, Kim E, Cho D, Cohen EP, Kim TS. Aminoacyl-tRNA synthetase-interacting multifunctional protein 1 suppresses tumor growth in breast cancer-bearing mice by negatively regulating myeloid-derived suppressor cell functions. Cancer Immunol Immunother 2016; 65:61-72. [PMID: 26613952 PMCID: PMC11029743 DOI: 10.1007/s00262-015-1777-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 11/18/2015] [Indexed: 12/12/2022]
Abstract
Myeloid-derived suppressor cells (MDSCs) are one of the most important cell types that contribute to negative regulation of immune responses in the tumor microenvironment. Recently, aminoacyl-tRNA synthetase-interacting multifunctional protein 1 (AIMP1), a novel pleiotropic cytokine, was identified as an antitumor protein that inhibits angiogenesis and induces antitumor responses. However, the effect of AIMP1 on MDSCs in the tumor environment remains unclear. In the present study, we demonstrated that AIMP1 significantly inhibited tumor growth in 4T1 breast cancer-bearing mice and reduced MDSCs population of tumor sites and spleens of tumor-bearing mice. AIMP1 reduced expansion of MDSCs from bone marrow-derived cells in the tumor-conditioned media. AIMP1 also negatively regulated suppressive activities of MDSCs by inhibiting IL-6 and NO production, and Arg-1 expression. Furthermore, treatment of breast cancer-bearing mice with AIMP1 decreased the capacity of MDSCs to suppress T cell proliferation and Treg cell induction. Western blot and inhibition experiments showed that downregulation of MDSCs functions by AIMP1 may result from attenuated activation of STATs, Akt, and ERK. These findings indicate that AIMP1 plays an essential role in negative regulation of suppressive functions of MDSCs. Therefore, it has a significant potential as a therapeutic agent for cancer treatment.
Collapse
Affiliation(s)
- Hye-Jin Hong
- Division of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Room 607, Hana Science Building, Anam-dong 5-ga, Seongbuk-gu, Seoul, 136-701, Republic of Korea
| | - Hui Xuan Lim
- Division of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Room 607, Hana Science Building, Anam-dong 5-ga, Seongbuk-gu, Seoul, 136-701, Republic of Korea
| | - Ju Han Song
- Division of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Room 607, Hana Science Building, Anam-dong 5-ga, Seongbuk-gu, Seoul, 136-701, Republic of Korea
| | - Arim Lee
- Division of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Room 607, Hana Science Building, Anam-dong 5-ga, Seongbuk-gu, Seoul, 136-701, Republic of Korea
| | - Eugene Kim
- Division of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Room 607, Hana Science Building, Anam-dong 5-ga, Seongbuk-gu, Seoul, 136-701, Republic of Korea
| | - Daeho Cho
- Division of Life Science, Sookmyung Women's University, Seoul, 140-742, Republic of Korea
| | - Edward P Cohen
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL, 60612, USA
| | - Tae Sung Kim
- Division of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Room 607, Hana Science Building, Anam-dong 5-ga, Seongbuk-gu, Seoul, 136-701, Republic of Korea.
| |
Collapse
|
57
|
Células mieloides supresoras: potencial vínculo entre la enfermedad pulmonar obstructiva crónica y el cáncer de pulmón. Arch Bronconeumol 2016; 52:29-35. [DOI: 10.1016/j.arbres.2015.07.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 07/06/2015] [Accepted: 07/07/2015] [Indexed: 12/22/2022]
|
58
|
Santarpia M, González-Cao M, Viteri S, Karachaliou N, Altavilla G, Rosell R. Programmed cell death protein-1/programmed cell death ligand-1 pathway inhibition and predictive biomarkers: understanding transforming growth factor-beta role. Transl Lung Cancer Res 2015; 4:728-42. [PMID: 26798582 PMCID: PMC4700220 DOI: 10.3978/j.issn.2218-6751.2015.12.04] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 10/22/2015] [Indexed: 12/19/2022]
Abstract
A deeper understanding of the key role of the immune system in regulating tumor growth and progression has led to the development of a number of immunotherapies, including cancer vaccines and immune checkpoint inhibitors. Immune checkpoint inhibitors target molecular pathways involved in immunosuppression, such as cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) and programmed cell death protein-1 (PD-1)/programmed cell death ligand-1 (PD-L1) pathway, with the goal to enhance the host's own immune anticancer response. In phase I-III trials, anti-PD-1/PD-L1 antibodies have demonstrated to be effective treatment strategies by inducing significant durable tumor responses, with manageable toxicities, in patients with various malignancies, including those traditionally considered non-immunogenic, such as non-small cell lung cancer (NSCLC). Identification of predictive biomarkers to select patients for immune therapies is currently being investigated to improve their therapeutic efficacy. Transforming growth factor-β (TGF-β), a pleiotropic cytokine with immunosuppressive effects on multiple cell types of the innate and adaptive immune system, has emerged as one of the potential key factors modulating response to immune checkpoint inhibitors. However, due to the complexity of the anti-cancer immune response, the predictive value of many other factors related to cancer cells or tumor microenvironment needs to be further explored.
Collapse
|
59
|
Lin A, Yan WH. Human Leukocyte Antigen-G (HLA-G) Expression in Cancers: Roles in Immune Evasion, Metastasis and Target for Therapy. Mol Med 2015; 21:782-791. [PMID: 26322846 PMCID: PMC4749493 DOI: 10.2119/molmed.2015.00083] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 08/20/2015] [Indexed: 12/29/2022] Open
Abstract
Aberrant induction of human leukocyte antigen-G (HLA-G) expression has been observed in various malignancies and is strongly associated with tumor immune escape, metastasis and poor prognosis. To date, great achievements have been made in understanding the underlying mechanisms of HLA-G involved in tumor progression. HLA-G could lead to tumor evasion by inhibition of immune cell cytolysis, differentiation and proliferation and inhibition of cytokine production, induction of immune cell apoptosis, generation of regulatory cells and expansion of myeloid-derived suppressive cells and by impairment of chemotaxis. Moreover, HLA-G could arm tumor cells with a higher invasive and metastatic potential with the upregulation of tumor-promoting factor expression such as matrix metalloproteinases (MMPs), indicating that ectopic HLA-G expression could render multiple effects during the progression of malignancies. In this review, we summarized the mechanisms of HLA-G involved in promoting tumor cell immune escaping, metastasis and disease progression. Special attention will be paid to its significance as an attractive therapeutic target in cancers.
Collapse
Affiliation(s)
- Aifen Lin
- Medical Research Center, Taizhou Hospital of Zhejiang Province, Wenzhou Medical University, Linhai, Zhejiang, People’s Republic of China
| | - Wei-Hua Yan
- Medical Research Center, Taizhou Hospital of Zhejiang Province, Wenzhou Medical University, Linhai, Zhejiang, People’s Republic of China
| |
Collapse
|
60
|
Fernández A, Oliver L, Alvarez R, Fernández LE, Lee KP, Mesa C. Adjuvants and myeloid-derived suppressor cells: enemies or allies in therapeutic cancer vaccination. Hum Vaccin Immunother 2015; 10:3251-60. [PMID: 25483674 DOI: 10.4161/hv.29847] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Adjuvants are a critical but largely overlooked and poorly understood component included in vaccine formulations to stimulate and modulate the desired immune responses to an antigen. However, unlike in the protective infectious disease vaccines, adjuvants for cancer vaccines also need to overcome the effect of tumor-induced suppressive immune populations circulating in tumor-bearing individuals. Myeloid-derived suppressor cells (MDSC) are considered to be one of the key immunosuppressive populations that inhibit tumor-specific T cell responses in cancer patients. This review focuses on the different signals for the activation of the immune system induced by adjuvants, and the close relationship to the mechanisms of recruitment and activation of MDSC. This work explores the possibility that a cancer vaccine adjuvant may either strengthen or weaken the effect of tumor-induced MDSC, and the crucial need to address this in present and future cancer vaccines.
Collapse
Key Words
- APC, antigen-presenting cells
- ARG1, arginase 1
- CTL, cytotoxic T lymphocytes
- DC, dendritic cells
- G-MDSC, granulocytic MDSC
- GM-CSF, granulocyte macrophage colony-stimulating factor
- MDSC
- MDSC, myeloid-derived suppressor cells
- Mo-MDSC, monocytic MDSC
- NK, natural killer
- NOS2, inducible nitric oxide synthase
- TAM, tumor-associated macrophages
- TLR ligands
- TLR, Toll-like receptors
- Treg, regulatory T cells
- adjuvants
- cancer
- cytokines
- immunotherapy
Collapse
Affiliation(s)
- Audry Fernández
- a Immunobiology Division; Center of Molecular Immunology ; Havana , Cuba
| | | | | | | | | | | |
Collapse
|
61
|
Alleviation of skin inflammation after Lin(-) cell transplantation correlates with their differentiation into myeloid-derived suppressor cells. Sci Rep 2015; 5:14663. [PMID: 26441031 PMCID: PMC4594128 DOI: 10.1038/srep14663] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 09/03/2015] [Indexed: 12/11/2022] Open
Abstract
To understand the cellular mechanism underlying the therapeutic effects exerted by hematopoietic stem cell transplantation in the repair of tissue damage, we investigated the in vivo dynamics of bone marrow (BM) lineage-negative (Lin−) cells transplanted into mice with hyper sensitivity dermatitis. Longitudinal in vivo imaging and flow cytometry analyses revealed that Lin− cells home directly to inflamed skin within 6 h, where they undergo extensive expansion with the peak on day 14 post-transplantation, and preferential differentiation into CD11b+Ly6GintLy6C+ cells by day 7. Cells with phenotypic profiles of neutrophils, macrophages, and DCs appeared in inflamed skin on day 14. Progenies of transplanted Lin− cells showed similar kinetics of expansion and myeloid differentiation in BM. However, differentiation into CD11b+Ly6GintLy6C+ cells in the inflamed skin on day 7 was more skewed toward CD115+ cells (≥60%) with immune suppressive function and higher expression levels of iNOS, arginase, and IL-10, compared with those in the BM. Transplantation of Lin− cells reduced the levels of Cd3 transcript and CD4+/CD8+ cells in inflamed skin. These results demonstrate differentiation of transplanted Lin− cells into myeloid-derived suppressor cells in inflamed skin to be the basis of the alleviation of skin inflammation after Lin− cell transplantation.
Collapse
|
62
|
Fang Z, Li J, Yu X, Zhang D, Ren G, Shi B, Wang C, Kosinska AD, Wang S, Zhou X, Kozlowski M, Hu Y, Yuan Z. Polarization of Monocytic Myeloid-Derived Suppressor Cells by Hepatitis B Surface Antigen Is Mediated via ERK/IL-6/STAT3 Signaling Feedback and Restrains the Activation of T Cells in Chronic Hepatitis B Virus Infection. THE JOURNAL OF IMMUNOLOGY 2015; 195:4873-83. [PMID: 26416274 DOI: 10.4049/jimmunol.1501362] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 09/04/2015] [Indexed: 12/18/2022]
Abstract
Chronic hepatitis B virus (HBV) infection is characterized by T cell tolerance to virus. Although inhibition of T cell responses by myeloid-derived suppressor cells (MDSCs) has been observed in patients with chronic hepatitis B (CHB), the mechanism for expansion of MDSCs remains ambiguous. In this study, a significant increased frequency of monocytic MDSCs (mMDSCs) was shown positively correlated to level of HBsAg in the patients with CHB. We further found hepatitis B surface Ag (HBsAg) efficiently promoted differentiation of mMDSCs in vitro, and monocytes in PBMCs performed as the progenitors. This required the activation of ERK/IL-6/STAT3 signaling feedback. Importantly, the mMDSCs polarized by HBsAg in vitro acquired the ability to suppress T cell activation. Additionally, treatment of all-trans retinoic acid, an MDSC-targeted drug, restored the proliferation and IFN-γ production by HBV-specific CD4(+) and CD8(+) T cells in PBMCs from patients with CHB and prevented increase of viral load in mouse model. In summary, HBsAg maintains HBV persistence and suppresses T cell responses by promoting differentiation of monocytes into mMDSCs. A therapy aimed at the abrogation of MDSCs may help to disrupt immune suppression in patients with CHB.
Collapse
Affiliation(s)
- Zhong Fang
- Key Laboratory of Medical Molecular Virology, Shanghai Public Health Clinical Center, Shanghai Medical College of Fudan University, Shanghai 201508, People's Republic of China; Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, People's Republic of China; and
| | - Jin Li
- Key Laboratory of Medical Molecular Virology, Shanghai Public Health Clinical Center, Shanghai Medical College of Fudan University, Shanghai 201508, People's Republic of China
| | - Xiaoyu Yu
- Key Laboratory of Medical Molecular Virology, Shanghai Public Health Clinical Center, Shanghai Medical College of Fudan University, Shanghai 201508, People's Republic of China
| | - Dandan Zhang
- Key Laboratory of Medical Molecular Virology, Shanghai Public Health Clinical Center, Shanghai Medical College of Fudan University, Shanghai 201508, People's Republic of China
| | - Guangxu Ren
- Key Laboratory of Medical Molecular Virology, Shanghai Public Health Clinical Center, Shanghai Medical College of Fudan University, Shanghai 201508, People's Republic of China
| | - Bisheng Shi
- Key Laboratory of Medical Molecular Virology, Shanghai Public Health Clinical Center, Shanghai Medical College of Fudan University, Shanghai 201508, People's Republic of China
| | - Cong Wang
- Key Laboratory of Medical Molecular Virology, Shanghai Public Health Clinical Center, Shanghai Medical College of Fudan University, Shanghai 201508, People's Republic of China
| | - Anna D Kosinska
- Key Laboratory of Medical Molecular Virology, Shanghai Public Health Clinical Center, Shanghai Medical College of Fudan University, Shanghai 201508, People's Republic of China
| | - Sen Wang
- Key Laboratory of Medical Molecular Virology, Shanghai Public Health Clinical Center, Shanghai Medical College of Fudan University, Shanghai 201508, People's Republic of China
| | - Xiaohui Zhou
- Key Laboratory of Medical Molecular Virology, Shanghai Public Health Clinical Center, Shanghai Medical College of Fudan University, Shanghai 201508, People's Republic of China
| | - Maya Kozlowski
- Key Laboratory of Medical Molecular Virology, Shanghai Public Health Clinical Center, Shanghai Medical College of Fudan University, Shanghai 201508, People's Republic of China; Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Yunwen Hu
- Key Laboratory of Medical Molecular Virology, Shanghai Public Health Clinical Center, Shanghai Medical College of Fudan University, Shanghai 201508, People's Republic of China;
| | - Zhenghong Yuan
- Key Laboratory of Medical Molecular Virology, Shanghai Public Health Clinical Center, Shanghai Medical College of Fudan University, Shanghai 201508, People's Republic of China; Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, People's Republic of China; and
| |
Collapse
|
63
|
It takes two to tango: Understanding the interactions between engineered nanomaterials and the immune system. Eur J Pharm Biopharm 2015; 95:3-12. [DOI: 10.1016/j.ejpb.2015.03.007] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Revised: 02/13/2015] [Accepted: 03/03/2015] [Indexed: 01/21/2023]
|
64
|
Abstract
The regulatory approval of ipilimumab (Yervoy) in 2011 ushered in a new era of cancer immunotherapies with durable clinical effects. Most of these breakthrough medicines are monoclonal antibodies that block protein-protein interactions between T cell checkpoint receptors and their cognate ligands. In addition, genetically engineered autologous T cell therapies have also recently demonstrated significant clinical responses in haematological cancers. Conspicuously missing from this class of therapies are traditional small-molecule drugs, which have previously served as the backbone of targeted cancer therapies. Modulating the immune system through a small-molecule approach offers several unique advantages that are complementary to, and potentially synergistic with, biologic modalities. This Review highlights immuno-oncology pathways and mechanisms that can be best or solely targeted by small-molecule medicines. Agents aimed at these mechanisms--modulation of the immune response, trafficking to the tumour microenvironment and cellular infiltration--are poised to significantly extend the scope of immuno-oncology applications and enhance the opportunities for combination with tumour-targeted agents and biologic immunotherapies.
Collapse
|
65
|
Dynamic changes in myeloid derived suppressor cell subsets following renal transplant: A prospective study. Transpl Immunol 2015; 32:164-71. [DOI: 10.1016/j.trim.2015.05.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 04/21/2015] [Accepted: 05/04/2015] [Indexed: 01/19/2023]
|
66
|
Parker KH, Beury DW, Ostrand-Rosenberg S. Myeloid-Derived Suppressor Cells: Critical Cells Driving Immune Suppression in the Tumor Microenvironment. Adv Cancer Res 2015. [PMID: 26216631 DOI: 10.1016/bs.acr.2015.04.002] [Citation(s) in RCA: 377] [Impact Index Per Article: 41.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature myeloid cells that suppress innate and adaptive immunity. MDSCs are present in many disease settings; however, in cancer, they are a major obstacle for both natural antitumor immunity and immunotherapy. Tumor and host cells in the tumor microenvironment (TME) produce a myriad of pro-inflammatory mediators that activate MDSCs and drive their accumulation and suppressive activity. MDSCs utilize a variety of mechanisms to suppress T cell activation, induce other immune-suppressive cell populations, regulate inflammation in the TME, and promote the switching of the immune system to one that tolerates and enhances tumor growth. Because MDSCs are present in most cancer patients and are potent immune-suppressive cells, MDSCs have been the focus of intense research in recent years. This review describes the history and identification of MDSCs, the role of inflammation and intracellular signaling events governing MDSC accumulation and suppressive activity, immune-suppressive mechanisms utilized by MDSCs, and recent therapeutics that target MDSCs to enhance antitumor immunity.
Collapse
Affiliation(s)
- Katherine H Parker
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland, USA
| | - Daniel W Beury
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland, USA
| | - Suzanne Ostrand-Rosenberg
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland, USA.
| |
Collapse
|
67
|
Domagala-Kulawik J. The role of the immune system in non-small cell lung carcinoma and potential for therapeutic intervention. Transl Lung Cancer Res 2015; 4:177-90. [PMID: 25870800 DOI: 10.3978/j.issn.2218-6751.2015.01.11] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 01/21/2015] [Indexed: 12/27/2022]
Abstract
Over a hundred years after the first description of this disease, lung cancer represents one of the major challenges in oncology. Radical treatment cannot be introduced in more than 70% of cases and overall survival rate does not exceed 15%. The immunosurveillance of lung cancer may be effective in early oncogenesis but is inhibited in the course of developing a clinically detectable tumor. Very low and heterogonous antigenicity of lung cancer cells leads to passive escape from anti-cancer immune defense. The cytotoxic lymphocytes (CTLs) that play a main role in the anticancer response are actively suppressed in the tumor environment and following regulatory mechanisms inhibit the recognition of tumor antigens by antigen presenting cells. The population of regulatory T cells (Tregs) is augmented and the expression of transcription factor-Foxp3 is markedly increased on tumor cells and tumor infiltrating lymphocytes (TIL). It is accomplished by M2 macrophage polarization, the activity of myeloid derived suppressor cells (MDSCs) and a significantly elevated concentration of cytokines: transforming growth factor beta (TGFβ) and IL-10 in the tumor microenvironment. Very active suppression of immune protection is the predominant role of the programmed death 1 (PD-1)-PD-L1 pathway. The blockage of this pathway was found to be an effective treatment approach; therefore the monoclonal antibodies are being intensively investigated in lung cancer patients. Cytotoxic T lymphocyte antigen-4 (CTLA-4) is the molecule capable of inhibiting the activation signal. The antibody anti-CTLA-4 improves CTLs function in solid tumors and lung cancer patients may benefit from use of this agent. The second way in lung cancer immunotherapy is production of anti-cancer vaccines using recognized cancer antigens: MAGE-A3, membrane associated glycoprotein (MUC-1), and EGF. It was recently shown in ongoing clinical trials that combined therapies: immune- and chemotherapy, radiotherapy or targeted therapy seem to be effective. Immunotherapy in lung cancer has an individual character-there is a need to assess the patient's immune status prior to implementation of immunomodulating therapy.
Collapse
Affiliation(s)
- Joanna Domagala-Kulawik
- Department of Internal Diseases, Pneumonology and Allergology, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
68
|
Morris ME, Beare JE, Reed RM, Dale JR, LeBlanc AJ, Kaufman CL, Zheng H, Ng CK, Williams SK, Hoying JB. Systemically delivered adipose stromal vascular fraction cells disseminate to peripheral artery walls and reduce vasomotor tone through a CD11b+ cell-dependent mechanism. Stem Cells Transl Med 2015; 4:369-80. [PMID: 25722428 PMCID: PMC4367510 DOI: 10.5966/sctm.2014-0252] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 01/19/2015] [Indexed: 12/14/2022] Open
Abstract
Vasoactivity, an important aspect of tissue healing, is often compromised in disease and tissue injury. Dysfunction in the smaller vasoactive arteries is most impactful, given the role of these vessels in controlling downstream tissue perfusion. The adipose stromal vascular fraction (SVF) is a mix of homeostatic cells shown to promote tissue healing. Our objective was to test the hypothesis that autologous SVF cells therapeutically modulate peripheral artery vasoactivity in syngeneic mouse models of small artery function. Analysis of vasoactivity of saphenous arteries isolated from normal mice 1 week after intravenous injection of freshly isolated SVF cells revealed that pressure-dependent artery vasomotor tone was decreased by the SVF cell isolate, but not one depleted of CD11b(+) cells. Scavenging hydrogen peroxide in the vessel wall abrogated the artery relaxation promoted by the SVF cell isolate. Consistent with a CD11b(+) cell being the relevant cell type, SVF-derived F4/80-positive macrophages were present within the adventitia of the artery wall coincident with vasorelaxation. In a model of artery inflammation mimicking a common disease condition inducing vasoactive dysfunction, the SVF cells potentiated relaxation of saphenous arteries without structurally remodeling the artery via a CD11b(+) cell-dependent manner. Our findings demonstrate that freshly isolated, adipose SVF cells promote vasomotor relaxation in vasoactive arteries via a hydrogen peroxide-dependent mechanism that required CD11b(+) cells (most likely macrophages). Given the significant impact of small artery dysfunction in disease, we predict that the intravenous delivery of this therapeutic cell preparation would significantly improve tissue perfusion, particularly in diseases with diffuse vascular involvement.
Collapse
Affiliation(s)
- Marvin E Morris
- Cardiovascular Innovation Institute, Department of Surgery, Department of Physiology and Biophysics, and Department of Radiology, University of Louisville, Louisville, Kentucky, USA; Christina M. Kleinert Institute, Louisville, Kentucky, USA
| | - Jason E Beare
- Cardiovascular Innovation Institute, Department of Surgery, Department of Physiology and Biophysics, and Department of Radiology, University of Louisville, Louisville, Kentucky, USA; Christina M. Kleinert Institute, Louisville, Kentucky, USA
| | - Robert M Reed
- Cardiovascular Innovation Institute, Department of Surgery, Department of Physiology and Biophysics, and Department of Radiology, University of Louisville, Louisville, Kentucky, USA; Christina M. Kleinert Institute, Louisville, Kentucky, USA
| | - Jacob R Dale
- Cardiovascular Innovation Institute, Department of Surgery, Department of Physiology and Biophysics, and Department of Radiology, University of Louisville, Louisville, Kentucky, USA; Christina M. Kleinert Institute, Louisville, Kentucky, USA
| | - Amanda J LeBlanc
- Cardiovascular Innovation Institute, Department of Surgery, Department of Physiology and Biophysics, and Department of Radiology, University of Louisville, Louisville, Kentucky, USA; Christina M. Kleinert Institute, Louisville, Kentucky, USA
| | - Christina L Kaufman
- Cardiovascular Innovation Institute, Department of Surgery, Department of Physiology and Biophysics, and Department of Radiology, University of Louisville, Louisville, Kentucky, USA; Christina M. Kleinert Institute, Louisville, Kentucky, USA
| | - Huaiyu Zheng
- Cardiovascular Innovation Institute, Department of Surgery, Department of Physiology and Biophysics, and Department of Radiology, University of Louisville, Louisville, Kentucky, USA; Christina M. Kleinert Institute, Louisville, Kentucky, USA
| | - Chin K Ng
- Cardiovascular Innovation Institute, Department of Surgery, Department of Physiology and Biophysics, and Department of Radiology, University of Louisville, Louisville, Kentucky, USA; Christina M. Kleinert Institute, Louisville, Kentucky, USA
| | - Stuart K Williams
- Cardiovascular Innovation Institute, Department of Surgery, Department of Physiology and Biophysics, and Department of Radiology, University of Louisville, Louisville, Kentucky, USA; Christina M. Kleinert Institute, Louisville, Kentucky, USA
| | - James B Hoying
- Cardiovascular Innovation Institute, Department of Surgery, Department of Physiology and Biophysics, and Department of Radiology, University of Louisville, Louisville, Kentucky, USA; Christina M. Kleinert Institute, Louisville, Kentucky, USA
| |
Collapse
|
69
|
Abstract
PURPOSE OF REVIEW Suppressor cells regulate immune responses during chronic viral infection by limiting immunopathology associated with inflammation and immune activation. This dampening of adaptive immune responses can be harmful in HIV-1 infection as it also prevents the immune system from clearing the virus, leading to viral persistence and prolonged antigen expression that often leads to immune exhaustion. A current priority is to find the best strategy to target and manipulate key molecules such as CD39 that suppress anti-HIV-1 immune responses. RECENT FINDINGS New suppressor cell subsets and cellular markers have been identified and characterized in the past years. We are able to identify and measure regulatory T cells, regulatory B cells and myeloid-derived suppressor cells in HIV-1-infected patients. We can also measure antigen-specific regulatory T cells in patients, which is a valuable step forward. Targeting HIV-1-specific regulatory T cells could be beneficial if we aim to manipulate key inhibitory molecules such as CTLA-4 and/or PD-1 that have already proven their efficacy in cancer. New other possible targets to take into account are CD39 and Tim-3-Gal9 pathways that have recently attracted attention in the field. These new findings offer the possibility to recognize suppressor cells as future targets in therapeutic vaccines because it became obvious that good vaccines candidates should concurrently generate robust effector responses and inhibit specific pathways that lead to immune suppression and exhaustion. SUMMARY The recent advances on suppressor cells and the availability of new markers or assays will certainly open up new avenues for targeting molecules that are involved in immune suppression pathways, thus avoiding viral persistence and immune exhaustion.
Collapse
|
70
|
Clements DR, Sterea AM, Kim Y, Helson E, Dean CA, Nunokawa A, Coyle KM, Sharif T, Marcato P, Gujar SA, Lee PWK. Newly recruited CD11b+, GR-1+, Ly6C(high) myeloid cells augment tumor-associated immunosuppression immediately following the therapeutic administration of oncolytic reovirus. THE JOURNAL OF IMMUNOLOGY 2015; 194:4397-412. [PMID: 25825443 DOI: 10.4049/jimmunol.1402132] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 03/04/2015] [Indexed: 12/31/2022]
Abstract
Tumor-associated immunosuppression aids cancer cells to escape immune-mediated attack and subsequent elimination. Recently, however, many oncolytic viruses, including reovirus, have been reported to overturn such immunosuppression and promote the development of a clinically desired antitumor immunity, which is known to promote favorable patient outcomes. Contrary to this existing paradigm, in this article we demonstrate that reovirus augments tumor-associated immunosuppression immediately following its therapeutic administration. Our data show that reovirus induces preferential differentiation of highly suppressive CD11b(+), Gr-1(+), Ly6C(high) myeloid cells from bone marrow hematopoietic progenitor cells. Furthermore, reovirus administration in tumor-bearing hosts drives time-dependent recruitment of CD11b(+), Gr-1(+), Ly6C(high) myeloid cells in the tumor milieu, which is further supported by virus-induced increased expression of numerous immune factors involved in myeloid-derived suppressor cell survival and trafficking. Most importantly, CD11b(+), Gr-1(+), Ly6C(high) myeloid cells specifically potentiate the suppression of T cell proliferation and are associated with the absence of IFN-γ response in the tumor microenvironment early during oncotherapy. Considering that the qualitative traits of a specific antitumor immunity are largely dictated by the immunological events that precede its development, our findings are of critical importance and must be considered while devising complementary interventions aimed at promoting the optimum efficacy of oncolytic virus-based anticancer immunotherapies.
Collapse
Affiliation(s)
- Derek R Clements
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada B3H 4R2
| | - Andra M Sterea
- Department of Biology, Dalhousie University, Halifax, Nova Scotia, Canada B3H 4R2
| | - Youra Kim
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada B3H 4R2
| | - Erin Helson
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada B3H 4R2; and
| | - Cheryl A Dean
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada B3H 4R2; and
| | - Anna Nunokawa
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada B3H 4R2; and
| | - Krysta Mila Coyle
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada B3H 4R2
| | - Tanveer Sharif
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada B3H 4R2; and
| | - Paola Marcato
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada B3H 4R2
| | - Shashi A Gujar
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada B3H 4R2; and Strategy and Organizational Performance, Izaak Walton Killiam Health Centre, Halifax, Nova Scotia, Canada B3K 6R8
| | - Patrick W K Lee
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada B3H 4R2; Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada B3H 4R2; and
| |
Collapse
|
71
|
Zhang ZJ, Bulur PA, Dogan A, Gastineau DA, Dietz AB, Lin Y. Immune independent crosstalk between lymphoma and myeloid suppressor CD14 +HLA-DR low/neg monocytes mediates chemotherapy resistance. Oncoimmunology 2015; 4:e996470. [PMID: 26137410 PMCID: PMC4485750 DOI: 10.1080/2162402x.2014.996470] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 12/04/2014] [Accepted: 12/05/2014] [Indexed: 11/05/2022] Open
Abstract
We have previously reported a novel phenotype of myeloid suppressors in lymphoma patients characterized by a loss of HLA-DR expression on monocytes, CD14+HLA-DRlow/neg. These cells were directly immunosuppressive and were associated with poor clinical outcome. In this study, we found that lymphoma tumors could have more than 30% of their tumor occupied by CD14+ cells. This intimate spatial connection suggested substantial cell–cell communication. We examined cross talk between monocytes from healthy volunteers (normal) and lymphoma cells in co-culture to identify the mechanisms and consequences of these interactions. Normal CD14+HLA-DR+ monocytes lost their HLA-DR expression after co-culture with lymphoma cells. Lymphoma-converted CD14+HLA-DRlow/neg cells exhibited similar immunosuppressive functions as CD14+HLA-DRlow/neg monocytes from lymphoma patients. Unexpectedly monocyte additions to lymphoma cell cultures protected lymphoma from cytotoxic killing by chemotherapy drug doxorubicin (DOX). Monocyte mediated resistance to DOX killing was associated with decreased Caspase-3 activity and increased anti-apoptotic heat shock protein-27 (Hsp27) expression. Soluble Hsp27 was detected in supernatant and patient plasma. Increased Hsp27 in plasma correlated with increased proportion of CD14+HLA-DRlow/neg monocytes in patient blood and was associated with lack of clinical response to DOX. This is the first report to describe a non-immune function of CD14+HLA-DRlow/neg monocytes: enhanced lymphoma resistance to chemotherapy. It is also the first report in lymphoma of Hsp27 as a potential mediator of lymphoma and monocyte crosstalk and chemotherapy resistance. Together with previous reports of the prevalence of these myeloid suppressors in other cancers, our findings identify this pathway and these interactions as a potential novel therapeutic target.
Collapse
Affiliation(s)
| | - Peggy A Bulur
- Division of Transfusion Medicine; Mayo Clinic ; Rochester, MN; USA
| | - Ahmet Dogan
- Department of Pathology and Laboratory Medicine; Memorial Sloan Kettering Cancer Center ; New York, NY, USA
| | - Dennis A Gastineau
- Division of Hematology; Mayo Clinic ; Rochester, MN, USA ; Division of Transfusion Medicine; Mayo Clinic ; Rochester, MN; USA
| | - Allan B Dietz
- Division of Transfusion Medicine; Mayo Clinic ; Rochester, MN; USA ; Division of Experimental Pathology; Mayo Clinic ; Rochester, MN USA
| | - Yi Lin
- Division of Hematology; Mayo Clinic ; Rochester, MN, USA
| |
Collapse
|
72
|
Wang W, Jiao Z, Duan T, Liu M, Zhu B, Zhang Y, Xu Q, Wang R, Xiong Y, Xu H, Lu L. Functional characterization of myeloid-derived suppressor cell subpopulations during the development of experimental arthritis. Eur J Immunol 2014; 45:464-73. [PMID: 25352399 DOI: 10.1002/eji.201444799] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 10/05/2014] [Accepted: 10/21/2014] [Indexed: 12/22/2022]
Abstract
Recent evidence indicates the existence of subpopulations of myeloid-derived suppressor cells (MDSCs) with distinct phenotypes and functions. Here, we characterized the role of MDSC subpopulations in the pathogenesis of autoimmune arthritis in a collagen-induced arthritis (CIA) mouse model. The splenic CD11b(+) Gr-1(+) MDSC population expanded in CIA mice, and these cells could be subdivided into polymorphonuclear (PMN) and mononuclear (MO) MDSC subpopulations based on Ly6C and Ly6G expression. During CIA, the proportion of splenic MO-MDSCs was increased in association with the severity of joint inflammation, while PMN-MDSCs were decreased. MO-MDSCs expressed higher levels of surface CD40 and CD86 protein, but lower levels of Il10, Tgfb1, Ccr5, and Cxcr2 mRNA. PMN-MDSCs exhibited a more potent capacity to suppress polyclonal T-cell proliferation in vitro, compared with MO-MDSCs. Moreover, the adoptive transfer of PMN-MDSCs, but not MO-MDSCs, decreased joint inflammation, accompanied by reduced levels of serum cytokine secretion and the frequencies of Th1 and Th17 cells in draining lymph nodes. These results suggest that there could be a shift from potently suppressive PMN-MDSCs to poorly suppressive MO-MDSCs during the development of experimental arthritis, which might reflect the failure of expanded MDSCs to suppress autoimmune arthritis.
Collapse
Affiliation(s)
- Wenhong Wang
- Zhenjiang Key Laboratory of Medical Immunology, Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China; Department of Pathogenic Biology, School of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
73
|
Knaul JK, Jörg S, Oberbeck-Mueller D, Heinemann E, Scheuermann L, Brinkmann V, Mollenkopf HJ, Yeremeev V, Kaufmann SHE, Dorhoi A. Lung-Residing Myeloid-derived Suppressors Display Dual Functionality in Murine Pulmonary Tuberculosis. Am J Respir Crit Care Med 2014; 190:1053-66. [DOI: 10.1164/rccm.201405-0828oc] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
74
|
Gantt S, Gervassi A, Jaspan H, Horton H. The role of myeloid-derived suppressor cells in immune ontogeny. Front Immunol 2014; 5:387. [PMID: 25165466 PMCID: PMC4131407 DOI: 10.3389/fimmu.2014.00387] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 07/29/2014] [Indexed: 01/13/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSC) are a heterogeneous population of granulocytic or monocytic cells that suppress innate as well as adaptive immune responses. In healthy adults, immature myeloid cells differentiate into macrophages, dendritic cells, and granulocytes in the bone marrow and MDSC are rarely detected in peripheral blood. However, in certain pathologies, in particular malignancies and chronic infection, differentiation of these cells is altered resulting in accumulation of circulating suppressive myeloid cells. MDSC express suppressive factors such as arginase-1, reactive oxygen species, and inducible nitric oxide synthase, which have the ability to inhibit T cell proliferation and cytoxicity, induce the expansion of regulatory T cells, and block natural killer cell activation. It is increasingly recognized that MDSC alter the immune response to several cancers, and perhaps chronic viral infections, in clinically important ways. In this review, we outline the potential contribution of MDSC to the generation of feto-maternal tolerance and to the ineffective immune responses to many infections and vaccines observed in early post-natal life. Granulocytic MDSC are present in large numbers in pregnant women and in cord blood, and wane rapidly during infancy. Furthermore, cord blood MDSC suppress in vitro T cell and NK responses, suggesting that they may play a significant role in human immune ontogeny. However, there are currently no data that demonstrate in vivo effects of MDSC on feto-maternal tolerance or immune ontogeny. Studies are ongoing to evaluate the functional importance of MDSC, including their effects on control of infection and response to vaccination in infancy. Importantly, several pharmacologic interventions have the potential to reverse MDSC function. Understanding the role of MDSC in infant ontogeny and their mechanisms of action could lead to interventions that reduce mortality due to early-life infections.
Collapse
Affiliation(s)
- Soren Gantt
- Child and Family Research Institute, University of British Columbia , Vancouver, BC , Canada
| | | | - Heather Jaspan
- Seattle BioMed , Seattle, WA , USA ; Division of Immunology, University of Cape Town , Cape Town , South Africa
| | - Helen Horton
- Seattle BioMed , Seattle, WA , USA ; Janssen ID&V Research and Development , Antwerp , Belgium
| |
Collapse
|
75
|
Charles JF, Aliprantis AO. Osteoclasts: more than 'bone eaters'. Trends Mol Med 2014; 20:449-59. [PMID: 25008556 PMCID: PMC4119859 DOI: 10.1016/j.molmed.2014.06.001] [Citation(s) in RCA: 253] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 05/28/2014] [Accepted: 06/02/2014] [Indexed: 02/08/2023]
Abstract
As the only cells definitively shown to degrade bone, osteoclasts are key mediators of skeletal diseases including osteoporosis. Bone-forming osteoblasts, and hematopoietic and immune system cells, each influence osteoclast formation and function, but the reciprocal impact of osteoclasts on these cells is less well appreciated. We highlight here the functions that osteoclasts perform beyond bone resorption. First, we consider how osteoclast signals may contribute to bone formation by osteoblasts and to the pathology of bone lesions such as fibrous dysplasia and giant cell tumors. Second, we review the interaction of osteoclasts with the hematopoietic system, including the stem cell niche and adaptive immune cells. Connections between osteoclasts and other cells in the bone microenvironment are discussed within a clinically relevant framework.
Collapse
Affiliation(s)
- Julia F Charles
- Department of Medicine, Division of Rheumatology, Allergy, and Immunology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Antonios O Aliprantis
- Department of Medicine, Division of Rheumatology, Allergy, and Immunology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
76
|
Fernández A, Oliver L, Alvarez R, Fernández LE, Mesa C. GM3-containing nanoparticles in immunosuppressed hosts: Effect on myeloid-derived suppressor cells. World J Immunol 2014; 4:98-106. [DOI: 10.5411/wji.v4.i2.98] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Revised: 05/27/2014] [Accepted: 06/27/2014] [Indexed: 02/05/2023] Open
Abstract
Cancer vaccines to date have not broadly achieved a significant impact on the overall survival of patients. The negative effect on the immune system of the tumor itself and conventional anti-tumor treatments such as chemotherapy is, undoubtedly, a key reason for these disappointing results. Myeloid-derived suppressor cells (MDSCs) are considered a central node of the immunosuppressive network associated with tumors. These cells inhibit the effector function of natural killer and CD8+ T cells, expand regulatory T cells and can differentiate into tumor-associated macrophages within the tumor microenvironment. Thus, overcoming the suppressive effects of MDSCs is likely to be critical for cancer immunotherapy to generate effective anti-tumor immune responses. However, the capacity of cancer vaccines and particularly their adjuvants to overcome this inhibitory population has not been well characterized. Very small size proteoliposomes (VSSP) is a nanoparticulated adjuvant specifically designed to be formulated with vaccines used in the treatment of immunocompromised patients. This adjuvant contains immunostimulatory bacterial signals together with GM3 ganglioside. VSSP promotes dendritic cell maturation, antigen cross-presentation to CD8+ T cells, Th1 polarization, and enhances CD8+ T cell response in tumor-free mice. Currently, four cancer vaccines using VSSP as the adjuvant are in Phase I and II clinical trials. In this review, we summarize our work characterizing the unique ability of VSSP to stimulate antigen-specific CD8+ T cell responses in two immunocompromised scenarios; in tumor-bearing mice and during chemotherapy-induced leukopenia. Particular emphasis has been placed on the interaction of these nanoparticles with MDSCs, as well as comparison with other cancer vaccine adjuvants currently in preclinical or clinical studies.
Collapse
|
77
|
Hardwick NR, Carroll M, Kaltcheva T, Qian D, Lim D, Leong L, Chu P, Kim J, Chao J, Fakih M, Yen Y, Espenschied J, Ellenhorn JDI, Diamond DJ, Chung V. p53MVA therapy in patients with refractory gastrointestinal malignancies elevates p53-specific CD8+ T-cell responses. Clin Cancer Res 2014; 20:4459-70. [PMID: 24987057 DOI: 10.1158/1078-0432.ccr-13-3361] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE To conduct a phase I trial of a modified vaccinia Ankara (MVA) vaccine delivering wild-type human p53 (p53MVA) in patients with refractory gastrointestinal cancers. EXPERIMENTAL DESIGN Three patients were vaccinated with 1.0×10(8) plaque-forming unit (pfu) p53MVA followed by nine patients at 5.6×10(8) pfu. Toxicity was classified using the NCI Common Toxicity Criteria and clinical responses were assessed by CT scan. Peripheral blood samples were collected pre- and post-immunization for immunophenotyping, monitoring of p53MVA-induced immune response, and examination of PD1 checkpoint inhibition in vitro. RESULTS p53MVA immunization was well tolerated at both doses, with no adverse events above grade 2. CD4+ and CD8+ T cells showing enhanced recognition of a p53 overlapping peptide library were detectable after the first immunization, particularly in the CD8+ T-cell compartment (P=0.03). However, in most patients, this did not expand further with the second and third immunization. The frequency of PD1+ T cells detectable in patients' peripheral blood mononuclear cells (PBMC) was significantly higher than in healthy controls. Furthermore, the frequency of PD1+ CD8+ T cells showed an inverse correlation with the peak CD8+ p53 response (P=0.02) and antibody blockade of PD1 in vitro increased the p53 immune responses detected after the second or third immunizations. Induction of strong T-cell and antibody responses to the MVA backbone were also apparent. CONCLUSION p53MVA was well tolerated and induced robust CD8+ T-cell responses. Combination of p53MVA with immune checkpoint inhibition could help sustain immune responses and lead to enhanced clinical benefit.
Collapse
Affiliation(s)
- Nicola R Hardwick
- Division of Translational Vaccine Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, California
| | - Mary Carroll
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, California
| | - Teodora Kaltcheva
- Division of Translational Vaccine Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, California
| | - Dajun Qian
- Bioinformatics Core Facility, City of Hope National Medical Center, Duarte, California
| | - Dean Lim
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, California
| | - Lucille Leong
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, California
| | - Peiguo Chu
- Department of Pathology, City of Hope National Medical Center, Duarte, California
| | - Joseph Kim
- Department of Surgical Oncology, City of Hope National Medical Center, Duarte, California
| | - Joseph Chao
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, California
| | - Marwan Fakih
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, California
| | - Yun Yen
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, California
| | - Jonathan Espenschied
- Division of Cancer Etiology and Outcomes Research, City of Hope National Medical Center, Duarte, California
| | | | - Don J Diamond
- Division of Translational Vaccine Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, California.
| | - Vincent Chung
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, California
| |
Collapse
|