51
|
Hellsten R, Lilljebjörn L, Johansson M, Leandersson K, Bjartell A. The STAT3 inhibitor galiellalactone inhibits the generation of MDSC-like monocytes by prostate cancer cells and decreases immunosuppressive and tumorigenic factors. Prostate 2019; 79:1611-1621. [PMID: 31348843 PMCID: PMC6771992 DOI: 10.1002/pros.23885] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 07/02/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND The transcription factor signal transducer and activator of transcription 3 (STAT3) is implicated in cancer drug resistance, metastasis, and immunosuppression and has been identified as a promising therapeutic target for new anticancer drugs. Myeloid-derived suppressor cells (MDSCs) play a major role in the suppression of antitumor immunity and STAT3 is involved in the accumulation, generation, and function of MDSCs. Thus, targeting STAT3 holds the potential of reversing immunosuppression in cancer. This study aims to investigate the effect of the small molecule STAT3 inhibitor galiellalactone on prostate cancer cell- induced generation of MDSCs from monocytes and the effect on immunosuppressive factors and inflammatory cytokines. METHODS Primary human monocytes were cocultured with prostate cancer cells (DU145, PC3, and LNCaP-IL6) or with conditioned medium (CM) from prostate cancer cells in the presence or absence of the STAT3 inhibitor galiellalactone. Monocytes were analyzed by flow cytometry for an MDSC-like phenotype (CD14+ HLA-DR-/lo ). The secretion and gene expression of immunosuppressive factors and inflammatory cytokines from prostate cancer cells and monocytes were investigated. RESULTS Galiellalactone blocked the prostate cancer cell-induced generation of MDSC-like monocytes with an immunosuppressive phenotype ex vivo. Monocytes cultured with CM from prostate cancer cells showed increased expression of phosphorylated STAT3. Prostate cancer cells increased the expression of interleukin1β (IL1β), IL10, and IL6 in monocytes which was inhibited by galiellalactone. In addition, galiellalactone decreased indoleamine 2,3-dioxygenase gene expression in monocytes. Galiellalactone reduced the levels of IL8 and granulocyte macrophage-colony stimulating factor in prostate cancer cells per se. CONCLUSION The STAT3 inhibitor galiellalactone may prevent the prostate cancer cell-induced generation of MDSCs and reverse the immunosuppressive mechanisms caused by the interplay between prostate cancer cells and MDSCs. This is a potential new immunotherapeutic approach for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Rebecka Hellsten
- Division of Urological Cancers, Department of Translational MedicineLund UniversityMalmöSweden
| | - Lisa Lilljebjörn
- Division of Urological Cancers, Department of Translational MedicineLund UniversityMalmöSweden
| | | | - Karin Leandersson
- Cancer Immunology, Department of Translational MedicineLund UniversityMalmöSweden
| | - Anders Bjartell
- Division of Urological Cancers, Department of Translational MedicineLund UniversityMalmöSweden
| |
Collapse
|
52
|
Li YD, Lamano JB, Lamano JB, Quaggin-Smith J, Veliceasa D, Kaur G, Biyashev D, Unruh D, Bloch O. Tumor-induced peripheral immunosuppression promotes brain metastasis in patients with non-small cell lung cancer. Cancer Immunol Immunother 2019; 68:1501-1513. [PMID: 31489465 PMCID: PMC7037587 DOI: 10.1007/s00262-019-02384-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 08/24/2019] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Brain metastases are a significant source of morbidity and mortality for patients with lung cancer. Lung cancer can induce local and systemic immunosuppression, promoting tumor growth and dissemination. One mechanism of immunosuppression is tumor-induced expansion of programmed death-ligand 1 (PD-L1) expressing myeloid cells. Here, we investigate peripheral blood immune phenotype in NSCLC patients with or without brain metastasis. METHODS Peripheral blood was collected from patients with lung metastatic brain tumors and pre-metastatic lung cancer. Immunosuppressive monocytes, myeloid-derived suppressor cells (MDSCs), and regulatory T cells (Tregs) were quantified through flow cytometry. T cell reactivity was analyzed via ELISpot. Brain metastasis conditioned media was collected from tumor-derived cell cultures and analyzed for cytokines by ELISA. Naïve monocytes were stimulated with brain metastasis conditioned media to evaluate PD-L1 stimulation. RESULTS Patients with brain metastatic lung carcinoma demonstrated increased peripheral monocyte PD-L1, MDSC abundance, and Treg percentage compared to early stage pre-metastatic patients and healthy controls. Patients with elevated peripheral monocyte PD-L1 had less reactive T cells and worse survival. Brain metastasis conditioned media stimulation increased monocyte PD-L1, and conditioned media IL-6 levels correlated with PD-L1 induction. Treatment with anti-IL-6 or anti-IL-6 receptor antibodies reduced PD-L1 expression. In summary, patients with lung cancer and brain metastases exhibit multiple markers of peripheral immunosuppression. CONCLUSIONS The frequency of PD-L1+ myeloid cells correlated with the presence of brain metastases. Tumor-derived IL-6 was capable of inducing PD-L1+ myeloid cells in vitro, suggesting that monitoring of immunosuppressive factors in peripheral blood may identify new targets for therapeutic intervention in selected patients.
Collapse
Affiliation(s)
- Yuping D Li
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jonathan B Lamano
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jason B Lamano
- Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Jessica Quaggin-Smith
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Dorina Veliceasa
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Gurvinder Kaur
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Dauren Biyashev
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Dusten Unruh
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Orin Bloch
- Department of Neurological Surgery, University of California - Davis, 4860 Y Street, Suite 3740, Sacramento, CA, 95817, USA.
| |
Collapse
|
53
|
Calcinotto A, Kohli J, Zagato E, Pellegrini L, Demaria M, Alimonti A. Cellular Senescence: Aging, Cancer, and Injury. Physiol Rev 2019; 99:1047-1078. [PMID: 30648461 DOI: 10.1152/physrev.00020.2018] [Citation(s) in RCA: 669] [Impact Index Per Article: 133.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Cellular senescence is a permanent state of cell cycle arrest that occurs in proliferating cells subjected to different stresses. Senescence is, therefore, a cellular defense mechanism that prevents the cells to acquire an unnecessary damage. The senescent state is accompanied by a failure to re-enter the cell cycle in response to mitogenic stimuli, an enhanced secretory phenotype and resistance to cell death. Senescence takes place in several tissues during different physiological and pathological processes such as tissue remodeling, injury, cancer, and aging. Although senescence is one of the causative processes of aging and it is responsible of aging-related disorders, senescent cells can also play a positive role. In embryogenesis and tissue remodeling, senescent cells are required for the proper development of the embryo and tissue repair. In cancer, senescence works as a potent barrier to prevent tumorigenesis. Therefore, the identification and characterization of key features of senescence, the induction of senescence in cancer cells, or the elimination of senescent cells by pharmacological interventions in aging tissues is gaining consideration in several fields of research. Here, we describe the known key features of senescence, the cell-autonomous, and noncell-autonomous regulators of senescence, and we attempt to discuss the functional role of this fundamental process in different contexts in light of the development of novel therapeutic targets.
Collapse
Affiliation(s)
- Arianna Calcinotto
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland , Bellinzona , Switzerland ; University of Groningen, European Research Institute for the Biology of Ageing, University Medical Center Groningen , Groningen , The Netherlands ; IOR, Oncology Institute of Southern Switzerland , Bellinzona , Switzerland ; Università della Svizzera Italiana, Faculty of Biomedical Sciences , Lugano , Italy ; Faculty of Biology and Medicine, University of Lausanne UNIL , Lausanne , Switzerland ; and Department of Medicine, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Jaskaren Kohli
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland , Bellinzona , Switzerland ; University of Groningen, European Research Institute for the Biology of Ageing, University Medical Center Groningen , Groningen , The Netherlands ; IOR, Oncology Institute of Southern Switzerland , Bellinzona , Switzerland ; Università della Svizzera Italiana, Faculty of Biomedical Sciences , Lugano , Italy ; Faculty of Biology and Medicine, University of Lausanne UNIL , Lausanne , Switzerland ; and Department of Medicine, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Elena Zagato
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland , Bellinzona , Switzerland ; University of Groningen, European Research Institute for the Biology of Ageing, University Medical Center Groningen , Groningen , The Netherlands ; IOR, Oncology Institute of Southern Switzerland , Bellinzona , Switzerland ; Università della Svizzera Italiana, Faculty of Biomedical Sciences , Lugano , Italy ; Faculty of Biology and Medicine, University of Lausanne UNIL , Lausanne , Switzerland ; and Department of Medicine, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Laura Pellegrini
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland , Bellinzona , Switzerland ; University of Groningen, European Research Institute for the Biology of Ageing, University Medical Center Groningen , Groningen , The Netherlands ; IOR, Oncology Institute of Southern Switzerland , Bellinzona , Switzerland ; Università della Svizzera Italiana, Faculty of Biomedical Sciences , Lugano , Italy ; Faculty of Biology and Medicine, University of Lausanne UNIL , Lausanne , Switzerland ; and Department of Medicine, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Marco Demaria
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland , Bellinzona , Switzerland ; University of Groningen, European Research Institute for the Biology of Ageing, University Medical Center Groningen , Groningen , The Netherlands ; IOR, Oncology Institute of Southern Switzerland , Bellinzona , Switzerland ; Università della Svizzera Italiana, Faculty of Biomedical Sciences , Lugano , Italy ; Faculty of Biology and Medicine, University of Lausanne UNIL , Lausanne , Switzerland ; and Department of Medicine, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Andrea Alimonti
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland , Bellinzona , Switzerland ; University of Groningen, European Research Institute for the Biology of Ageing, University Medical Center Groningen , Groningen , The Netherlands ; IOR, Oncology Institute of Southern Switzerland , Bellinzona , Switzerland ; Università della Svizzera Italiana, Faculty of Biomedical Sciences , Lugano , Italy ; Faculty of Biology and Medicine, University of Lausanne UNIL , Lausanne , Switzerland ; and Department of Medicine, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| |
Collapse
|
54
|
Dong L, Zieren RC, Xue W, de Reijke TM, Pienta KJ. Metastatic prostate cancer remains incurable, why? Asian J Urol 2019; 6:26-41. [PMID: 30775246 PMCID: PMC6363601 DOI: 10.1016/j.ajur.2018.11.005] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 07/18/2018] [Accepted: 09/12/2018] [Indexed: 12/21/2022] Open
Abstract
Metastatic prostate cancer patients present in two ways-with already disseminated disease at the time of presentation or with disease recurrence after definitive local therapy. Androgen deprivation therapy is given as the most effective initial treatment to patients. However, after the initial response, almost all patients will eventually progress despite the low levels of testosterone. Disease at this stage is termed castration resistant prostate cancer (CRPC). Before 2010, the taxane docetaxel was the first and only life prolonging agent for metastatic CRPC (mCRPC). The last decade has witnessed robust progress in CRPC therapeutics development. Abiraterone, enzalutamide, apalutamide and sipuleucel-T have been evaluated as first- and second-line agents in mCRPC patients, while cabazitaxel was approved as a second-line treatment. Radium-223 dichloride was approved in symptomatic patients with bone metastases and no known visceral metastases pre- and post-docetaxel. However, despite significant advances, mCRPC remains a lethal disease. Both primary and acquired resistance have been observed in CRPC patients treated by these new agents. It could be solely cell intrinsic or it is possible that the clonal heterogeneity in treated tumors may result from the adaptive responses to the selective pressures within the tumor microenvironment. The aim of this review is to list current treatment agents of CRPC and summarize recent findings in therapeutic resistance mechanisms.
Collapse
Affiliation(s)
- Liang Dong
- The Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Richard C. Zieren
- The Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Urology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Wei Xue
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Theo M. de Reijke
- Department of Urology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Kenneth J. Pienta
- The Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
55
|
Rivas-Fuentes S, Iglesias AH, Trejo AG, Castro DYC, Figueroa NI, Pérez TA, Saldaña RB, Rosete PG. Restoration of Peripheral Intermediate and Classical Monocytes Expressing HLA-DR in Patients With Lung Adenocarcinoma After Platinum-Based Chemotherapy. Technol Cancer Res Treat 2018; 17:1533033818764720. [PMID: 29642771 PMCID: PMC5900825 DOI: 10.1177/1533033818764720] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Lung adenocarcinoma represents one of the lung cancer subtypes with major prevalence. Accumulating evidence indicates that the immune system plays an important role in the evolution of the neoplastic process; additionally, several reports suggest that chemotherapy has an immunomodulatory effect. In order to identify the peripheral subpopulations of leukocytes that may change after chemotherapy, we evaluated several peripheral immune subpopulations of monocytes and lymphocytes by multicolor flow cytometry. In addition, we also measured cytokines and growth factors on plasma in order to evaluate the pro-inflammatory context in patients with lung adenocarcinoma after chemotherapy. We found that HLA-DR+ classical and intermediate monocytes were decreased in patients before chemotherapy, compared to controls. After chemotherapy, the relative percentage of those subpopulations was restored. In addition, interleukin 1β, interleukin 12, and interleukin 5 were increased after chemotherapy compared to prechemotherapy levels, while MIP-1β was decreased.
Collapse
Affiliation(s)
- Selma Rivas-Fuentes
- 1 Department of Biochemistry Research, National Institute of Respiratory Diseases "Ismael Cosío Villegas," Mexico City, Mexico
| | - Anjarath Higuera Iglesias
- 2 Department of Clinical Epidemiology Research, National Institute of Respiratory Diseases "Ismael Cosío Villegas," Mexico City, Mexico
| | - Ana García Trejo
- 1 Department of Biochemistry Research, National Institute of Respiratory Diseases "Ismael Cosío Villegas," Mexico City, Mexico
| | - Daniel Yair Chavarría Castro
- 1 Department of Biochemistry Research, National Institute of Respiratory Diseases "Ismael Cosío Villegas," Mexico City, Mexico
| | - Norma Inclán Figueroa
- 1 Department of Biochemistry Research, National Institute of Respiratory Diseases "Ismael Cosío Villegas," Mexico City, Mexico
| | - Teresa Aguirre Pérez
- 3 Oncology Pneumology Service, National Institute of Respiratory Diseases "Ismael Cosío Villegas," Mexico City, Mexico
| | - Renata Báez Saldaña
- 3 Oncology Pneumology Service, National Institute of Respiratory Diseases "Ismael Cosío Villegas," Mexico City, Mexico
| | - Patricia Gorocica Rosete
- 1 Department of Biochemistry Research, National Institute of Respiratory Diseases "Ismael Cosío Villegas," Mexico City, Mexico
| |
Collapse
|
56
|
Abstract
Despite the high long-term survival in localized prostate cancer, metastatic prostate cancer remains largely incurable even after intensive multimodal therapy. The lethality of advanced disease is driven by the lack of therapeutic regimens capable of generating durable responses in the setting of extreme tumor heterogeneity on the genetic and cell biological levels. Here, we review available prostate cancer model systems, the prostate cancer genome atlas, cellular and functional heterogeneity in the tumor microenvironment, tumor-intrinsic and tumor-extrinsic mechanisms underlying therapeutic resistance, and technological advances focused on disease detection and management. These advances, along with an improved understanding of the adaptive responses to conventional cancer therapies, anti-androgen therapy, and immunotherapy, are catalyzing development of more effective therapeutic strategies for advanced disease. In particular, knowledge of the heterotypic interactions between and coevolution of cancer and host cells in the tumor microenvironment has illuminated novel therapeutic combinations with a strong potential for more durable therapeutic responses and eventual cures for advanced disease. Improved disease management will also benefit from artificial intelligence-based expert decision support systems for proper standard of care, prognostic determinant biomarkers to minimize overtreatment of localized disease, and new standards of care accelerated by next-generation adaptive clinical trials.
Collapse
Affiliation(s)
- Guocan Wang
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Di Zhao
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Denise J Spring
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Ronald A DePinho
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
57
|
Wang PF, Song SY, Wang TJ, Ji WJ, Li SW, Liu N, Yan CX. Prognostic role of pretreatment circulating MDSCs in patients with solid malignancies: A meta-analysis of 40 studies. Oncoimmunology 2018; 7:e1494113. [PMID: 30288362 PMCID: PMC6169582 DOI: 10.1080/2162402x.2018.1494113] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 06/19/2018] [Accepted: 06/24/2018] [Indexed: 12/13/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) have been shown to contribute to tumor progression, mainly through immune suppression. Inverse correlations have been observed between MDSC levels and patient survival for various malignancies. The purpose of this meta-analysis was to evaluate the prognostic value of pretreatment circulating MDSCs. We searched MEDLINE and EMBASE from their inceptions to September 2017 to identify relevant articles. Using a fixed or random effects model, pooled hazard ratios (HRs) were estimated for overall survival (OS) and combined disease-free survival, progression-free survival, and recurrence-free survival (DFS/PFS/RFS). A total of 40 studies comprising 2721 were included. For solid tumors, high levels of pretreatment circulating MDSCs were significantly associated with worse OS (HR = 1.796, 95% CI = 1.587-2.032) and DFS/PFS/RFS (HR = 2.459, 95% CI = 2.018-2.997). Breast cancer showed the largest association between high MDSC levels and worse OS (pooled HR = 3.053). Elevated MDSCs were also associated with worse OS for mixed-stage tumors (pooled HR = 1.659) and advanced-stage tumors (pooled HR = 2.337). Furthermore, both monocytic-MDSCs (M-MDSCs) and granulocytic or polymorphonuclear (PMN-MDSCs) showed negative associations with survival outcomes. Overall, high levels of pretreatment circulating MDSCs negatively influenced survival in most cancers. Pretreatment circulating MDSCs should be taken into account to further improve prognostic evaluation and develop novel therapeutic strategies.
Collapse
Affiliation(s)
- Peng-Fei Wang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Si-Ying Song
- School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Ting-Jian Wang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Wen-Jun Ji
- Department of Neurosurgery, Key Laboratory, The Second Hospital of Yulin, Xi’an Jiao tong University, Xi’an, China
| | - Shou-Wei Li
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Ning Liu
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Chang-Xiang Yan
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
58
|
Okla K, Wertel I, Wawruszak A, Bobiński M, Kotarski J. Blood-based analyses of cancer: Circulating myeloid-derived suppressor cells - is a new era coming? Crit Rev Clin Lab Sci 2018; 55:376-407. [PMID: 29927668 DOI: 10.1080/10408363.2018.1477729] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Progress in cancer treatment made by the beginning of the 21st century has shifted the paradigm from one-size-fits-all to tailor-made treatment. The popular vision, to study solid tumors through the relatively noninvasive sampling of blood, is one of the most thrilling and rapidly advancing fields in global cancer diagnostics. From this perspective, immune-cell analysis in cancer could play a pivotal role in oncology practice. This approach is driven both by rapid technological developments, including the analysis of circulating myeloid-derived suppressor cells (cMDSCs), and by the increasing application of (immune) therapies, the success or failure of which may depend on effective and timely measurements of relevant biomarkers. Although the implementation of these powerful noninvasive diagnostic capabilities in guiding precision cancer treatment is poised to change the ways in which we select and monitor cancer therapy, challenges remain. Here, we discuss the challenges associated with the analysis and clinical aspects of cMDSCs and assess whether the problems in implementing tumor-evolution monitoring as a global tool in personalized oncology can be overcome.
Collapse
Affiliation(s)
- Karolina Okla
- a 1st Chair and Department of Oncological Gynaecology and Gynaecology, Tumor Immunology Laboratory , Medical University of Lublin , Lublin , Poland
| | - Iwona Wertel
- a 1st Chair and Department of Oncological Gynaecology and Gynaecology, Tumor Immunology Laboratory , Medical University of Lublin , Lublin , Poland
| | - Anna Wawruszak
- b Department of Biochemistry and Molecular Biology , Medical University of Lublin , Lublin , Poland
| | - Marcin Bobiński
- a 1st Chair and Department of Oncological Gynaecology and Gynaecology, Tumor Immunology Laboratory , Medical University of Lublin , Lublin , Poland
| | - Jan Kotarski
- a 1st Chair and Department of Oncological Gynaecology and Gynaecology, Tumor Immunology Laboratory , Medical University of Lublin , Lublin , Poland
| |
Collapse
|
59
|
Weber R, Fleming V, Hu X, Nagibin V, Groth C, Altevogt P, Utikal J, Umansky V. Myeloid-Derived Suppressor Cells Hinder the Anti-Cancer Activity of Immune Checkpoint Inhibitors. Front Immunol 2018; 9:1310. [PMID: 29942309 PMCID: PMC6004385 DOI: 10.3389/fimmu.2018.01310] [Citation(s) in RCA: 383] [Impact Index Per Article: 63.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 05/25/2018] [Indexed: 12/20/2022] Open
Abstract
Immune checkpoint inhibitors (ICI) used for cancer immunotherapy were shown to boost the existing anti-tumor immune response by preventing the inhibition of T cells by tumor cells. Antibodies targeting two negative immune checkpoint pathways, namely cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), programmed cell death protein 1 (PD-1), and programmed cell death-ligand 1 (PD-L1), have been approved first for patients with melanoma, squamous non-small cell lung cancer (NSCLC), and renal cell carcinoma. Clinical trials are ongoing to verify the efficiency of these antibodies for other cancer types and to evaluate strategies to block other checkpoint molecules. However, a number of patients do not respond to this treatment possibly due to profound immunosuppression, which is mediated partly by myeloid-derived suppressor cells (MDSC). This heterogeneous population of immature myeloid cells can strongly inhibit anti-tumor activities of T and NK cells and stimulate regulatory T cells (Treg), leading to tumor progression. Moreover, MDSC can contribute to patient resistance to immune checkpoint inhibition. Accumulating evidence demonstrates that the frequency and immunosuppressive function of MDSC in cancer patients can be used as a predictive marker for therapy response. This review focuses on the role of MDSC in immune checkpoint inhibition and provides an analysis of combination strategies for MDSC targeting together with ICI to improve their therapeutic efficiency in cancer patients.
Collapse
Affiliation(s)
- Rebekka Weber
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany.,Faculty of Biosciences, Ruprecht-Karl University of Heidelberg, Heidelberg, Germany
| | - Viktor Fleming
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany.,Faculty of Biosciences, Ruprecht-Karl University of Heidelberg, Heidelberg, Germany
| | - Xiaoying Hu
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | - Vasyl Nagibin
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | - Christopher Groth
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | - Peter Altevogt
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | - Viktor Umansky
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| |
Collapse
|
60
|
Cioni B, Zwart W, Bergman AM. Androgen receptor moonlighting in the prostate cancer microenvironment. Endocr Relat Cancer 2018; 25:R331-R349. [PMID: 29618577 DOI: 10.1530/erc-18-0042] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 04/04/2018] [Indexed: 01/03/2023]
Abstract
Androgen receptor (AR) signaling is vital for the normal development of the prostate and is critically involved in prostate cancer (PCa). AR is not only found in epithelial prostate cells but is also expressed in various cells in the PCa-associated stroma, which constitute the tumor microenvironment (TME). In the TME, AR is expressed in fibroblasts, macrophages, lymphocytes and neutrophils. AR expression in the TME was shown to be decreased in higher-grade and metastatic PCa, suggesting that stromal AR plays a protective role against PCa progression. With that, the functionality of AR in stromal cells appears to deviate from the receptor's classical function as described in PCa cells. However, the biological action of AR in these cells and its effect on cancer progression remains to be fully understood. Here, we systematically review the pathological, genomic and biological literature on AR actions in various subsets of prostate stromal cells and aim to better understand the consequences of AR signaling in the TME in relation to PCa development and progression.
Collapse
Affiliation(s)
- B Cioni
- Division of OncogenomicsThe Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - W Zwart
- Division of OncogenomicsThe Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode InstituteThe Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - A M Bergman
- Division of OncogenomicsThe Netherlands Cancer Institute, Amsterdam, the Netherlands
- Division of Medical OncologyThe Netherlands Cancer Institute, Amsterdam, the Netherlands
| |
Collapse
|
61
|
Bernsmeier C, Triantafyllou E, Brenig R, Lebosse FJ, Singanayagam A, Patel VC, Pop OT, Khamri W, Nathwani R, Tidswell R, Weston CJ, Adams DH, Thursz MR, Wendon JA, Antoniades CG. CD14 + CD15 - HLA-DR - myeloid-derived suppressor cells impair antimicrobial responses in patients with acute-on-chronic liver failure. Gut 2018; 67:1155-1167. [PMID: 28592438 PMCID: PMC5969362 DOI: 10.1136/gutjnl-2017-314184] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 04/26/2017] [Accepted: 04/27/2017] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Immune paresis in patients with acute-on-chronic liver failure (ACLF) accounts for infection susceptibility and increased mortality. Immunosuppressive mononuclear CD14+HLA-DR- myeloid-derived suppressor cells (M-MDSCs) have recently been identified to quell antimicrobial responses in immune-mediated diseases. We sought to delineate the function and derivation of M-MDSC in patients with ACLF, and explore potential targets to augment antimicrobial responses. DESIGN Patients with ACLF (n=41) were compared with healthy subjects (n=25) and patients with cirrhosis (n=22) or acute liver failure (n=30). CD14+CD15-CD11b+HLA-DR- cells were identified as per definition of M-MDSC and detailed immunophenotypic analyses were performed. Suppression of T cell activation was assessed by mixed lymphocyte reaction. Assessment of innate immune function included cytokine expression in response to Toll-like receptor (TLR-2, TLR-4 and TLR-9) stimulation and phagocytosis assays using flow cytometry and live cell imaging-based techniques. RESULTS Circulating CD14+CD15-CD11b+HLA-DR- M-MDSCs were markedly expanded in patients with ACLF (55% of CD14+ cells). M-MDSC displayed immunosuppressive properties, significantly decreasing T cell proliferation (p=0.01), producing less tumour necrosis factor-alpha/interleukin-6 in response to TLR stimulation (all p<0.01), and reduced bacterial uptake of Escherichia coli (p<0.001). Persistently low expression of HLA-DR during disease evolution was linked to secondary infection and 28-day mortality. Recurrent TLR-2 and TLR-4 stimulation expanded M-MDSC in vitro. By contrast, TLR-3 agonism reconstituted HLA-DR expression and innate immune function ex vivo. CONCLUSION Immunosuppressive CD14+HLA-DR- M-MDSCs are expanded in patients with ACLF. They were depicted by suppressing T cell function, attenuated antimicrobial innate immune responses, linked to secondary infection, disease severity and prognosis. TLR-3 agonism reversed M-MDSC expansion and innate immune function and merits further evaluation as potential immunotherapeutic agent.
Collapse
Affiliation(s)
- Christine Bernsmeier
- Institute of Liver Studies, King’s College Hospital, King’s College London, London, UK,Liver Biology Laboratory, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Evangelos Triantafyllou
- Institute of Liver Studies, King’s College Hospital, King’s College London, London, UK,Division of Digestive Diseases, St. Mary’s Campus, Imperial College London, London, UK,Institute of Immunology and Immunotherapy, NIHR Biomedical Research Unit, Centre for Liver Research, University of Birmingham, Birmingham, UK
| | - Robert Brenig
- Liver Biology Laboratory, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Fanny J Lebosse
- Division of Digestive Diseases, St. Mary’s Campus, Imperial College London, London, UK
| | - Arjuna Singanayagam
- Institute of Liver Studies, King’s College Hospital, King’s College London, London, UK,Division of Digestive Diseases, St. Mary’s Campus, Imperial College London, London, UK,Institute of Immunology and Immunotherapy, NIHR Biomedical Research Unit, Centre for Liver Research, University of Birmingham, Birmingham, UK
| | - Vishal C Patel
- Institute of Liver Studies, King’s College Hospital, King’s College London, London, UK
| | - Oltin T Pop
- Institute of Liver Studies, King’s College Hospital, King’s College London, London, UK
| | - Wafa Khamri
- Division of Digestive Diseases, St. Mary’s Campus, Imperial College London, London, UK
| | - Rooshi Nathwani
- Division of Digestive Diseases, St. Mary’s Campus, Imperial College London, London, UK
| | - Robert Tidswell
- Institute of Liver Studies, King’s College Hospital, King’s College London, London, UK
| | - Christopher J Weston
- Institute of Immunology and Immunotherapy, NIHR Biomedical Research Unit, Centre for Liver Research, University of Birmingham, Birmingham, UK
| | - David H Adams
- Institute of Immunology and Immunotherapy, NIHR Biomedical Research Unit, Centre for Liver Research, University of Birmingham, Birmingham, UK
| | - Mark R Thursz
- Division of Digestive Diseases, St. Mary’s Campus, Imperial College London, London, UK
| | - Julia A Wendon
- Institute of Liver Studies, King’s College Hospital, King’s College London, London, UK
| | - Charalambos Gustav Antoniades
- Institute of Liver Studies, King’s College Hospital, King’s College London, London, UK,Division of Digestive Diseases, St. Mary’s Campus, Imperial College London, London, UK,Institute of Immunology and Immunotherapy, NIHR Biomedical Research Unit, Centre for Liver Research, University of Birmingham, Birmingham, UK
| |
Collapse
|
62
|
Profiling Prostate Cancer Therapeutic Resistance. Int J Mol Sci 2018; 19:ijms19030904. [PMID: 29562686 PMCID: PMC5877765 DOI: 10.3390/ijms19030904] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 03/16/2018] [Accepted: 03/16/2018] [Indexed: 02/06/2023] Open
Abstract
The major challenge in the treatment of patients with advanced lethal prostate cancer is therapeutic resistance to androgen-deprivation therapy (ADT) and chemotherapy. Overriding this resistance requires understanding of the driving mechanisms of the tumor microenvironment, not just the androgen receptor (AR)-signaling cascade, that facilitate therapeutic resistance in order to identify new drug targets. The tumor microenvironment enables key signaling pathways promoting cancer cell survival and invasion via resistance to anoikis. In particular, the process of epithelial-mesenchymal-transition (EMT), directed by transforming growth factor-β (TGF-β), confers stem cell properties and acquisition of a migratory and invasive phenotype via resistance to anoikis. Our lead agent DZ-50 may have a potentially high efficacy in advanced metastatic castration resistant prostate cancer (mCRPC) by eliciting an anoikis-driven therapeutic response. The plasticity of differentiated prostate tumor gland epithelium allows cells to de-differentiate into mesenchymal cells via EMT and re-differentiate via reversal to mesenchymal epithelial transition (MET) during tumor progression. A characteristic feature of EMT landscape is loss of E-cadherin, causing adherens junction breakdown, which circumvents anoikis, promoting metastasis and chemoresistance. The targetable interactions between androgens/AR and TGF-β signaling are being pursued towards optimized therapeutic regimens for the treatment of mCRPC. In this review, we discuss the recent evidence on targeting the EMT-MET dynamic interconversions to overcome therapeutic resistance in patients with recurrent therapeutically resistant prostate cancer. Exploitation of the phenotypic landscape and metabolic changes that characterize the prostate tumor microenvironment in advanced prostate cancer and consequential impact in conferring treatment resistance are also considered in the context of biomarker discovery.
Collapse
|
63
|
Boibessot C, Toren P. Sex steroids in the tumor microenvironment and prostate cancer progression. Endocr Relat Cancer 2018; 25:R179-R196. [PMID: 29317479 DOI: 10.1530/erc-17-0493] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 01/08/2018] [Indexed: 12/30/2022]
Abstract
Prostate cancer is uniquely dependent on androgens. Despite years of research on the relationship between androgens and prostate cancer, many questions remain as to the biological effects of androgens and other sex steroids during prostate cancer progression. This article reviews the clinical and basic research on the influence of sex steroids such as androgens, estrogens and progesterone within the prostate tumor microenvironment on the progression of prostate cancer. We review clinical studies to date evaluating serum sex steroids as prognostic biomarkers and discuss their respective biological effects within the prostate tumor microenvironment. We also review the link between genomic alterations and sex steroid levels within prostate tumors. Finally, we highlight the links between sex steroid levels and the function of the immune system within the tumor microenvironment. As the context of treatment of lethal prostate cancer evolves over time, an understanding of this underlying biology remains central to developing optimal treatment approaches.
Collapse
Affiliation(s)
- Clovis Boibessot
- Department of SurgeryLaval University, Quebec City, Quebec, Canada
| | - Paul Toren
- Department of SurgeryLaval University, Quebec City, Quebec, Canada
| |
Collapse
|
64
|
Lima DS, Lemes RPG, Matos DM. Immunosuppressive monocytes (CD14 +/HLA-DR low/-) increase in childhood precursor B-cell acute lymphoblastic leukemia after induction chemotherapy. Med Oncol 2018; 35:36. [PMID: 29429058 DOI: 10.1007/s12032-018-1092-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 01/31/2018] [Indexed: 01/12/2023]
Abstract
In tumor microenvironment, immunosuppression is a common event and results from the inhibition of activated immune cells and generation of cells with immunosuppressive capacity, as some subtypes of monocytes. The aim of this study was to evaluate the presence of immunosuppressive CD14+/HLA-DRlow/- monocytes in pediatric patients with the diagnosis of B-cell acute lymphoblastic leukemia (B-ALL) and, moreover, verify whether the chemotherapeutic treatment has any effect on these cells. Peripheral blood (PB) and bone marrow (BM) samples were collected from 15 untreated pediatric patients. The presence of CD14+/HLA-DRlow/- monocytes was evaluated at diagnosis and in the end of induction chemotherapy by flow cytometry. CD14+/HLA-DRlow/- monocytes increase was observed in 60% (9/15) of the patients at the end of the induction therapy. We were able to detect an increase in CD14+/HLA-DRlow/- monocytes values in BM and PB samples of pediatric patients with B-ALL. This increase was observed in the end of induction chemotherapy, which leads us to believe that these changes probably could have been induced by the inflammatory process engendered by the cytotoxic treatment or by drugs used in the chemotherapy treatment. This finding may be useful to guide new therapeutic approaches contemplating immunomodulatory drugs that act in the depletion of immunosuppressive monocytes.
Collapse
Affiliation(s)
- D S Lima
- Oncohematology Section, Albert Sabin Children Hospital, Ceará, Brazil
- Department of Clinical and Toxicological Analysis, Federal University of Ceará, UFC, Ceará, Brazil
| | - R P G Lemes
- Department of Clinical and Toxicological Analysis, Federal University of Ceará, UFC, Ceará, Brazil
| | - D M Matos
- Flow Cytometry Section, Clementino Fraga Laboratory, Ceará, Brazil.
| |
Collapse
|
65
|
Millrud CR, Mehmeti M, Leandersson K. Docetaxel promotes the generation of anti-tumorigenic human macrophages. Exp Cell Res 2017; 362:525-531. [PMID: 29269075 DOI: 10.1016/j.yexcr.2017.12.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 12/15/2017] [Accepted: 12/18/2017] [Indexed: 12/20/2022]
Abstract
The taxanes Docetaxel and Paclitaxel are two of the standard chemotherapies for patients with metastatic breast cancer. The functional effect of Docetaxel and Paclitaxel on human innate immune cells of the myeloid lineage is not well established, nor is the effects these agents have on differentiation of monocytes into macrophages and dendritic cells. Therefore, the aim with this project was to determine the effects of Docetaxel and Paclitaxel on primary human monocyte differentiation, activation and function. For this purpose, primary human monocytes were isolated from healthy donors and cultured with or without Docetaxel and Paclitaxel. We found that Docetaxel promoted the differentiation of primary human monocytes into pro-inflammatory macrophages with an M1 phenotype and an ability to present antigens to T cells. Monocytes treated with Docetaxel also displayed an elevated secretion of IL-8 and IL-1β, but did not promote generation of monocytic myeloid-derived suppressor cells. In conclusion, Docetaxel appears to have an immune stimulatory effect that would be beneficial for an anti-tumorigenic type of immune response, whereas Paclitaxel seems to have less effect on myeloid cells.
Collapse
Affiliation(s)
- Camilla Rydberg Millrud
- Cancer Immunology, Department of Translational Medicine, Lund University, Skånes University Hospital, Malmö, Sweden.
| | - Meliha Mehmeti
- Cancer Immunology, Department of Translational Medicine, Lund University, Skånes University Hospital, Malmö, Sweden
| | - Karin Leandersson
- Cancer Immunology, Department of Translational Medicine, Lund University, Skånes University Hospital, Malmö, Sweden
| |
Collapse
|
66
|
CD14+ HLA-DR-/low MDSCs are elevated in the periphery of early-stage breast cancer patients and suppress autologous T cell proliferation. Breast Cancer Res Treat 2017; 168:401-411. [PMID: 29230664 DOI: 10.1007/s10549-017-4594-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 11/22/2017] [Indexed: 02/07/2023]
Abstract
PURPOSE Despite the recent expansion in the use of immunotherapy for many cancer types, it is still not a standard treatment for breast cancer. Identifying differences in the immune systems of breast cancer patients compared to healthy women might provide insight into potential targets for immunotherapy and thus may assist its clinical implementation. METHODS Multi-colour flow cytometry was used to investigate myeloid and lymphoid populations in the peripheral blood of breast cancer patients (n = 40) and in the blood of healthy age-matched women (n = 25). We additionally performed functional testing to identify immune suppressive mechanisms used by circulating CD14+ myeloid cells from breast cancer patients. RESULTS Our results show that breast cancer patients have significantly elevated frequencies of cells with the monocytic myeloid-derived suppressor cell (mMDSC) phenotype CD14+ HLA-DR-/low compared with healthy women (p < 0.01). We also observed higher levels of earlier differentiated T cells and correspondingly lower levels of T cells in later stages of differentiation (p < 0.05). These disease-associated differences could already be detected in early-stage breast cancer patients in stages 1 and 2 (n = 33 of 40) (p < 0.05). Levels of circulating T cells correlated with certain clinical features and with patient age (p < 0.05). Functional tests showed that CD14+ myeloid cells from breast cancer patients more potently suppressed autologous T cell proliferation than CD14+ cells from healthy women (p < 0.01). Subsequent investigation determined that suppression was mediated in part by reactive oxygen species, because inhibiting this pathway partially restored T cell proliferation (p < 0.01). CONCLUSION Our results highlight the potential importance of cells with mMDSC phenotypes in breast cancer, identifiable already at early stages of disease. This may provide a basis for identifying possible new therapeutic targets to enhance anti-cancer immunity.
Collapse
|
67
|
Koga N, Moriya F, Waki K, Yamada A, Itoh K, Noguchi M. Immunological efficacy of herbal medicines in prostate cancer patients treated by personalized peptide vaccine. Cancer Sci 2017; 108:2326-2332. [PMID: 28898532 PMCID: PMC5715291 DOI: 10.1111/cas.13397] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 09/04/2017] [Accepted: 09/07/2017] [Indexed: 12/20/2022] Open
Abstract
This randomized phase II study investigated the immunological efficacy of herbal medicines (HM) using Hochu‐ekki‐to and Keishi‐bukuryo‐gan in combination with personalized peptide vaccination (PPV) for castration‐resistant prostate cancer (CRPC). Seventy patients with CRPC were assigned to two arms; PPV plus HM or PPV alone. Two to four peptides were chosen from 31 peptides derived from cancer antigens for a s.c. injection of PPV given eight times according to the patient's human leukocyte antigen type and levels of antigen‐specific IgG titer before PPV treatment. Peptide‐specific CTL, IgG, regulatory T cells (Treg), monocytic myeloid‐derived suppressor cells (Mo‐MDSC), and interleukin‐6 (IL‐6) responses were measured before and at the eighth vaccination. Clinical outcomes were also analyzed. Combination therapy of PPV with HM was well tolerated without severe adverse events. There was no significant change in antigen‐specific IgG, CTL, Treg or clinical outcomes. Combination therapy of PPV with HM stabilized the frequency of Mo‐MDSC (1.91%–1.92%, P = 0.96) and serum levels of IL‐6 (19.2 pg/mL to 16.1 pg/mL, P = 0.63) during the treatment. In contrast, the frequency of Mo‐MDSC and levels of IL‐6 in the PPV‐alone group were significantly increased (0.91%–1.49% for Mo‐MDSC and 9.2 pg/mL to 19.4 pg/mL for IL‐6, respectively). These results suggest that the combined use of HM has the potential to prevent the immunosuppression induced by Mo‐MDSC or IL‐6 during immunotherapy. More research is needed to validate the findings of the present study.
Collapse
Affiliation(s)
- Noriko Koga
- Division of Clinical Research, Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, Kurume, Japan
| | - Fukuko Moriya
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan
| | - Kayoko Waki
- Division of Cancer Vaccines, Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, Kurume, Japan
| | - Akira Yamada
- Division of Cancer Vaccines, Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, Kurume, Japan
| | - Kyogo Itoh
- Cancer Vaccine Center, Kurume University School of Medicine, Kurume, Japan
| | - Masanori Noguchi
- Division of Clinical Research, Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, Kurume, Japan.,Cancer Vaccine Center, Kurume University School of Medicine, Kurume, Japan.,Department of Urology, Kurume University School of Medicine, Kurume, Japan
| |
Collapse
|
68
|
Gao XH, Tian L, Wu J, Ma XL, Zhang CY, Zhou Y, Sun YF, Hu B, Qiu SJ, Zhou J, Fan J, Guo W, Yang XR. Circulating CD14 + HLA-DR -/low myeloid-derived suppressor cells predicted early recurrence of hepatocellular carcinoma after surgery. Hepatol Res 2017; 47:1061-1071. [PMID: 27764536 DOI: 10.1111/hepr.12831] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 10/16/2016] [Accepted: 10/18/2016] [Indexed: 12/23/2022]
Abstract
AIM Myeloid-derived suppressor cells (MDSCs) play an important role in tumor progression. The aim of the present study was to investigate the prognostic value of MDSCs for early recurrence of hepatocellular carcinoma (HCC) in patients undergoing curative resection. METHODS Myeloid-derived suppressor cells were measured by flow cytometry. The correlation between MDSCs and tumor recurrence was analyzed using a cohort of 183 patients who underwent curative resection between February 2014 and July 2015. Prognostic significance was further assessed using Kaplan-Meier survival estimates and log-rank tests. RESULTS In vivo, CD14+ HLA-DR-/low MDSCs inhibit T cell proliferation and secretion. The frequency of CD14+ HLA-DR-/low MDSCs was significantly higher in HCC patients (3.7 ± 5.3%, n = 183) than in chronic hepatitis patients (1.4 ± 0.6%, n = 25) and healthy controls (1.1 ± 0.5%, n = 50). High frequency of MDSCs was significantly correlated with recurrence (time to recurrence) (P < 0.001) and overall survival (P = 0.034). Patients with HCC in the high MDSC group were prone to more vascular invasion (P = 0.018) and high systemic immune-inflammation index (SII) (P = 0.009) than those in the low MDSC group. Scatter-plot analyses revealed a significant positive correlation between the SII level and the frequency of MDSCs (r = 0.188, P = 0.011). Patients with HCC with a high MDSC frequency and high SII level had significantly shorter time to recurrence (P < 0.001) and overall survival (P = 0.028) than those with a low MDSC frequency and low SII. CONCLUSIONS An increased frequency of MDSCs was correlated with early recurrence and predicted the prognosis of patients with HCC undergoing curative resection. The HCC patients with high frequency of MDSCs should be provided more advanced management and frequent monitoring.
Collapse
Affiliation(s)
- Xing-Hui Gao
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lu Tian
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiong Wu
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiao-Lu Ma
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chun-Yan Zhang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yan Zhou
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yun-Fan Sun
- Department of Liver Surgery, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Bo Hu
- Department of Liver Surgery, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shuang-Jian Qiu
- Department of Liver Surgery, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jian Zhou
- Department of Liver Surgery, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jia Fan
- Department of Liver Surgery, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wei Guo
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xin-Rong Yang
- Department of Liver Surgery, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
69
|
Ascites-derived IL-6 and IL-10 synergistically expand CD14 +HLA-DR -/low myeloid-derived suppressor cells in ovarian cancer patients. Oncotarget 2017; 8:76843-76856. [PMID: 29100353 PMCID: PMC5652747 DOI: 10.18632/oncotarget.20164] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 06/27/2017] [Indexed: 11/25/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSC) play a key immunosuppressive role in various types of cancer, including ovarian cancer (OC). In this study, we characterized CD14+HLA-DR–/lo MDSC with a typical monocytic phenotype (M-MDSC) in the peripheral blood (PB) and ascites from OC patients. Compared to healthy donors, OC patients had a significantly increased abundance of M-MDSC in both PB and ascites; importantly, their abundance in both compartments was inversely associated with the prognosis where OC patients with higher level of M-MDSC having a shorter relapse-free survival. Intriguingly, we demonstrated that M-MDSC could be readily induced by ascitic fluids (AF) from OC patients, which was predominantly dependent on IL-6, IL-10 and STAT3 activation as neutralization of IL-6 and/or IL-10 or inhibition of STAT3 abrogated MDSC's expansion while recombinant IL-6 and IL-10 recapitulated the expansive effect of AF; furthermore, predominantly elevated levels of IL-6 and IL-10 has been noted in the AF which was positively correlated with the abundance of M-MDSC as well as poor prognosis of OC patients. As expected, we observed that AF-driven STAT3 activation upregulated the expression of arginase (ARG1) and inducible nitric oxide synthase (iNOS) in induced M-MDSC through which these MDSC executed the immunosuppressive activity. Taken together, these results demonstrate that abundant M-MDSC are present in both periphery and ascites of OC patients whose accumulation and suppressive activity is critically attributable to ascites-derived IL-6 and IL-10 and their downstream STAT3 signal, thus providing a potentially novel therapeutic option by locally targeting MDSC to improve antitumor efficacy.
Collapse
|
70
|
Lopez-Bujanda Z, Drake CG. Myeloid-derived cells in prostate cancer progression: phenotype and prospective therapies. J Leukoc Biol 2017; 102:393-406. [PMID: 28550116 PMCID: PMC6608078 DOI: 10.1189/jlb.5vmr1116-491rr] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 04/17/2017] [Accepted: 04/19/2017] [Indexed: 12/21/2022] Open
Abstract
Prostate cancer is the second most common cause of cancer mortality in men in the United States. As is the case for other tumor types, accumulating evidence suggests an important role for myeloid-derived cells in the promotion and progression of prostate cancer. Here, we briefly describe myeloid-derived cells that interact with tumor cells and what is known about their immune suppressive function. We next discuss new evidence for tumor cell-mediated myeloid infiltration via the PI3K/PTEN/AKT signaling pathway and an alternative mechanism for immune evasion that may be regulated by an endoplasmic reticulum stress response. Finally, we discuss several interventions that target myeloid-derived cells to treat prostate cancer.
Collapse
Affiliation(s)
- Zoila Lopez-Bujanda
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York, USA
| | - Charles G Drake
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York, USA
| |
Collapse
|
71
|
Meirow Y, Baniyash M. Immune biomarkers for chronic inflammation related complications in non-cancerous and cancerous diseases. Cancer Immunol Immunother 2017; 66:1089-1101. [PMID: 28674756 PMCID: PMC11029284 DOI: 10.1007/s00262-017-2035-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 06/20/2017] [Indexed: 01/05/2023]
Abstract
Chronic inflammation arising in a diverse range of non-cancerous and cancerous diseases, dysregulates immunity and exposes patients to a variety of complications. These include immunosuppression, tissue damage, cardiovascular diseases and more. In cancer, chronic inflammation and related immunosuppression can directly support tumor growth and dramatically reduce the efficacies of traditional treatments, as well as novel immune-based therapies, which require a functional immune system. Nowadays, none of the immune biomarkers, regularly used by clinicians can sense a developing chronic inflammation, thus complications can only be detected upon their appearance. This review focuses on the necessity for such immune status biomarkers, which could predict complications prior to their appearance. Herein we bring examples for the use of cellular and molecular biomarkers in diagnosis, prognosis and follow-up of patients suffering from various cancers, for prediction of response to immune-based anti-cancer therapy and for prediction of cardiovascular disease in type 2 diabetes patients. Monitoring such biomarkers is expected to have a major clinical impact in addition to unraveling of the entangled complexity underlying dysregulated immunity in chronic inflammation. Thus, newly discovered biomarkers and those that are under investigation are projected to open a new era towards combating the silent damage induced by chronic inflammation.
Collapse
Affiliation(s)
- Yaron Meirow
- The Lautenberg Center for General and Tumor Immunology, Faculty of Medicine, Israel-Canada Medical Research Institute, The Hebrew University, POB 12272, 91120, Jerusalem, Israel
| | - Michal Baniyash
- The Lautenberg Center for General and Tumor Immunology, Faculty of Medicine, Israel-Canada Medical Research Institute, The Hebrew University, POB 12272, 91120, Jerusalem, Israel.
| |
Collapse
|
72
|
Hazeldine J, Naumann DN, Toman E, Davies D, Bishop JRB, Su Z, Hampson P, Dinsdale RJ, Crombie N, Duggal NA, Harrison P, Belli A, Lord JM. Prehospital immune responses and development of multiple organ dysfunction syndrome following traumatic injury: A prospective cohort study. PLoS Med 2017; 14:e1002338. [PMID: 28719602 PMCID: PMC5515405 DOI: 10.1371/journal.pmed.1002338] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 05/31/2017] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Almost all studies that have investigated the immune response to trauma have analysed blood samples acquired post-hospital admission. Thus, we know little of the immune status of patients in the immediate postinjury phase and how this might influence patient outcomes. The objective of this study was therefore to comprehensively assess the ultra-early, within 1-hour, immune response to trauma and perform an exploratory analysis of its relationship with the development of multiple organ dysfunction syndrome (MODS). METHODS AND FINDINGS The immune and inflammatory response to trauma was analysed in 89 adult trauma patients (mean age 41 years, range 18-90 years, 75 males) with a mean injury severity score (ISS) of 24 (range 9-66), from whom blood samples were acquired within 1 hour of injury (mean time to sample 42 minutes, range 17-60 minutes). Within minutes of trauma, a comprehensive leukocytosis, elevated serum pro- and anti-inflammatory cytokines, and evidence of innate cell activation that included neutrophil extracellular trap generation and elevated surface expression of toll-like receptor 2 and CD11b on monocytes and neutrophils, respectively, were observed. Features consistent with immune compromise were also detected, notably elevated numbers of immune suppressive CD16BRIGHT CD62LDIM neutrophils (82.07 x 106/l ± 18.94 control versus 1,092 x 106/l ± 165 trauma, p < 0.0005) and CD14+HLA-DRlow/- monocytes (34.96 x 106/l ± 4.48 control versus 95.72 x 106/l ± 8.0 trauma, p < 0.05) and reduced leukocyte cytokine secretion in response to lipopolysaccharide stimulation. Exploratory analysis via binary logistic regression found a potential association between absolute natural killer T (NKT) cell numbers and the subsequent development of MODS. Study limitations include the relatively small sample size and the absence of data relating to adaptive immune cell function. CONCLUSIONS Our study highlighted the dynamic and complex nature of the immune response to trauma, with immune alterations consistent with both activation and suppression evident within 1 hour of injury. The relationship of these changes, especially in NKT cell numbers, to patient outcomes such as MODS warrants further investigation.
Collapse
Affiliation(s)
- Jon Hazeldine
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
- NIHR Surgical Reconstruction and Microbiology Research Centre, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
- * E-mail:
| | - David N. Naumann
- NIHR Surgical Reconstruction and Microbiology Research Centre, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| | - Emma Toman
- NIHR Surgical Reconstruction and Microbiology Research Centre, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| | - David Davies
- NIHR Surgical Reconstruction and Microbiology Research Centre, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| | - Jonathan R. B. Bishop
- NIHR Surgical Reconstruction and Microbiology Research Centre, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| | - Zhangjie Su
- NIHR Surgical Reconstruction and Microbiology Research Centre, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| | - Peter Hampson
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
- Scar Free Foundation, Birmingham Centre for Burns Research, Birmingham, United Kingdom
| | - Robert J. Dinsdale
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
- Scar Free Foundation, Birmingham Centre for Burns Research, Birmingham, United Kingdom
| | - Nicholas Crombie
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
- Midlands Air Ambulance, Unit 16 Enterprise Trading Estate, Brierley Hill, West Midlands, United Kingdom
| | - Niharika Arora Duggal
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Paul Harrison
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
- Scar Free Foundation, Birmingham Centre for Burns Research, Birmingham, United Kingdom
| | - Antonio Belli
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
- NIHR Surgical Reconstruction and Microbiology Research Centre, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| | - Janet M. Lord
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
- NIHR Surgical Reconstruction and Microbiology Research Centre, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| |
Collapse
|
73
|
Chesney JA, Mitchell RA, Yaddanapudi K. Myeloid-derived suppressor cells-a new therapeutic target to overcome resistance to cancer immunotherapy. J Leukoc Biol 2017; 102:727-740. [PMID: 28546500 DOI: 10.1189/jlb.5vmr1116-458rrr] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 04/18/2017] [Accepted: 04/20/2017] [Indexed: 12/12/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature myeloid cells that accumulate during pathologic conditions, such as cancer. Patients diagnosed with advanced metastatic cancers have an average survival of 12-24 mo, a survival time that hasn't changed significantly in the past 30 yr. Despite some encouraging improvements in response rates and overall survival in patients receiving immunotherapies, such as immune checkpoint inhibitors, most patients will ultimately progress. MDSCs contribute to immunotherapeutic resistance by actively inhibiting antitumor T cell proliferation and cytotoxic activity as well as by promoting expansion of protumorigenic T regulatory cells, thereby, dampening the host immune responses against the tumor. In addition, MDSCs promote angiogenesis, tumor invasion, and metastasis. Thus, MDSCs are potential therapeutic targets in cases of multiple cancers. This review focuses on the phenotypic and functional characteristics of MDSCs and provides an overview of the mono- and combinatorial-therapeutic strategies that target MDSCs with an objective of enhancing the efficacy of cancer immunotherapies.
Collapse
Affiliation(s)
- Jason A Chesney
- Molecular Targets Program, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA; .,Department of Medicine, University of Louisville, Louisville, Kentucky, USA; and
| | - Robert A Mitchell
- Molecular Targets Program, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA; .,Department of Medicine, University of Louisville, Louisville, Kentucky, USA; and.,Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, USA
| | - Kavitha Yaddanapudi
- Molecular Targets Program, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA; .,Department of Medicine, University of Louisville, Louisville, Kentucky, USA; and.,Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
74
|
Myeloid Cell-Specific Knockout of NFI-A Improves Sepsis Survival. Infect Immun 2017; 85:IAI.00066-17. [PMID: 28167668 DOI: 10.1128/iai.00066-17] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 01/30/2017] [Indexed: 01/04/2023] Open
Abstract
Myeloid progenitor-derived suppressor cells (MDSCs) arise from myeloid progenitors and suppress both innate and adaptive immunity. MDSCs expand during the later phases of sepsis in mice, promote immunosuppression, and reduce survival. Here, we report that the myeloid differentiation-related transcription factor nuclear factor I-A (NFI-A) controls MDSC expansion during sepsis and impacts survival. Unlike MDSCs, myeloid cells with conditional deletion of the Nfia gene normally differentiated into effector cells during sepsis, cleared infecting bacteria, and did not express immunosuppressive mediators. In contrast, ectopic expression of NFI-A in myeloid progenitors from NFI-A myeloid cell-deficient mice impeded myeloid cell maturation and promoted immune repressor function. Importantly, surviving septic mice with conditionally deficient NFI-A myeloid cells were able to respond to challenge with bacterial endotoxin by mounting an acute inflammatory response. Together, these results support the concept of NFI-A as a master molecular transcriptome switch that controls myeloid cell differentiation and maturation and that malfunction of this switch during sepsis promotes MDSC expansion that adversely impacts sepsis outcome.
Collapse
|
75
|
Effective combinatorial immunotherapy for castration-resistant prostate cancer. Nature 2017; 543:728-732. [PMID: 28321130 PMCID: PMC5374023 DOI: 10.1038/nature21676] [Citation(s) in RCA: 393] [Impact Index Per Article: 56.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 01/31/2017] [Indexed: 12/12/2022]
Abstract
A significant fraction of advanced prostate cancer (PCa) patients treated with androgen deprivation therapy (ADT) experience relapse with relentless progression to lethal metastatic castration-resistant prostate cancer (mCRPC)1. Immune checkpoint blockade (ICB) using antibodies against cytotoxic-T-lymphocyte-associated protein 4 (CTLA4) or programmed cell death 1/programmed cell death 1 ligand 1 (PD1/PD-L1) generates durable therapeutic responses in a significant subset of patients across a variety of cancer types2. However, mCRPC showed overwhelming de novo resistance to ICB3–5, motivating a search for targeted therapies that overcome this resistance. Myeloid-derived suppressor cells (MDSCs) are known to play important roles in tumor immune evasion6. Circulating MDSC abundance correlates with PSA levels and metastasis in PCa patients7–9. Mouse models of PCa show that MDSCs (CD11b+ Gr1+) promote tumor initiation10 and progression11. These observations prompted us to hypothesize that robust immunotherapy responses in mCRPC may be elicited by the combined actions of ICB agents together with targeted agents that neutralize MDSCs yet preserve T cell function. Here we developed a novel chimeric mouse model of mCRPC to efficiently test combination therapies in an autochthonous setting. Combination of anti-CTLA4 and anti-PD1 engendered only modest efficacy. Targeted therapy against mCRPC-infiltrating MDSCs, using multikinase inhibitors such as cabozantinib and BEZ235, also showed minimal anti-tumor activities. Strikingly, primary and metastatic CRPC showed robust synergistic responses when ICB was combined with MDSC-targeted therapy. Mechanistically, combination therapy efficacy stemmed from the upregulation of IL-1ra and suppression of MDSC-promoting cytokines secreted by PCa cells. These observations illuminate a clinical path hypothesis for combining ICB with MDSC-targeted therapies in the treatment of mCRPC.
Collapse
|
76
|
Elliott LA, Doherty GA, Sheahan K, Ryan EJ. Human Tumor-Infiltrating Myeloid Cells: Phenotypic and Functional Diversity. Front Immunol 2017; 8:86. [PMID: 28220123 PMCID: PMC5292650 DOI: 10.3389/fimmu.2017.00086] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Accepted: 01/18/2017] [Indexed: 12/14/2022] Open
Abstract
Our current understanding of human tumor-resident myeloid cells is, for the most part, based on a large body of work in murine models or studies enumerating myeloid cells in patient tumor samples using immunohistochemistry (IHC). This has led to the establishment of the theory that, by and large, tumor-resident myeloid cells are either “protumor” M2 macrophages or myeloid-derived suppressor cells (MDSC). This concept has accelerated our understanding of myeloid cells in tumor progression and enabled the elucidation of many key regulatory mechanisms involved in cell recruitment, polarization, and activation. On the other hand, this paradigm does not embrace the complexity of the tumor-resident myeloid cell phenotype (IHC can only measure 1 or 2 markers per sample) and their possible divergent function in the hostile tumor microenvironment. Here, we examine the criteria that define human tumor-infiltrating myeloid cell subsets and provide a comprehensive and critical review of human myeloid cell nomenclature in cancer. We also highlight new evidence characterizing their contribution to cancer pathogenesis based on evidence derived from clinical studies drawing comparisons with murine studies where necessary. We then review the mechanisms in which myeloid cells are regulated by tumors in humans and how these are being targeted therapeutically.
Collapse
Affiliation(s)
- Louise A Elliott
- Centre for Colorectal Disease, St. Vincent's University Hospital, School of Medicine, University College Dublin , Dublin , Ireland
| | - Glen A Doherty
- Centre for Colorectal Disease, St. Vincent's University Hospital, School of Medicine, University College Dublin , Dublin , Ireland
| | - Kieran Sheahan
- Centre for Colorectal Disease, St. Vincent's University Hospital, School of Medicine, University College Dublin , Dublin , Ireland
| | - Elizabeth J Ryan
- Centre for Colorectal Disease, St. Vincent's University Hospital, School of Medicine, University College Dublin , Dublin , Ireland
| |
Collapse
|
77
|
Calcinotto A, Alimonti A. Aging tumour cells to cure cancer: “pro-senescence” therapy for cancer. Swiss Med Wkly 2017; 147:w14367. [DOI: 10.57187/smw.2017.14367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Robust scientific evidence demonstrates that senescence induction in cancer works as a potent weapon to eradicate tumorigenesis. Therapies that enhance senescence not only promote a stable cell growth arrest but also work as a strong stimulus for the activation of the antitumour immune response. However, recent advances suggest that if senescent tumour cells are not cleared from the tumours, they may promote tumour progression and metastasis. In this article, we focus on concepts that are relevant to a pro-senescence therapeutic approach, including caveats, and we propose therapeutic strategies that involve the combined use of pro-senescence therapies with immunotherapies to promote the clearance of senescent tumour cells. In our opinion, these approaches may avoid potential negative effects of pro-senescence therapies and may also enhance the efficacy of currently available immunotherapies.
Collapse
|
78
|
Javeed N, Gustafson MP, Dutta SK, Lin Y, Bamlet WR, Oberg AL, Petersen GM, Chari ST, Dietz AB, Mukhopadhyay D. Immunosuppressive CD14 +HLA-DR lo/neg monocytes are elevated in pancreatic cancer and "primed" by tumor-derived exosomes. Oncoimmunology 2016; 6:e1252013. [PMID: 28197368 DOI: 10.1080/2162402x.2016.1252013] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 10/17/2016] [Accepted: 10/19/2016] [Indexed: 01/06/2023] Open
Abstract
Immunological strategies to treat pancreatic cancer offer new therapeutic approaches to improve patient outcomes. Understanding alterations in the immune systems of pancreatic cancer patients will likely lead to advances in immunotherapy for the disease. We profiled peripheral blood leukocytes from pancreatic cancer patients (n = 22) and age-matched controls (n = 20) using flow cytometry. Immune profiling of pancreatic cancer patients identified phenotypic changes in various immune cell populations, including a population of immunosuppressive monocytes (CD14+HLA-DRlo/neg), which were shown to be increased in these patients. There was a correlation between the levels of CD14+ monocytes and the levels of CD14+HLA-DRlo/neg monocytes in peripheral blood from pancreatic cancer patients. HLA-DR downregulation of monocytes was shown to occur through pancreatic cancer-derived exosome interactions with monocytes. In an in vitro model, exosomes from patient-derived xenograft cell lines and patient plasma decreased HLA-DR expression on CD14+ monocytes. Additionally, tumor-derived exosomes caused immune suppression in monocytes through altered STAT3 signaling, induction of arginase expression, and reactive oxygen species. These findings provide novel insights into the mechanisms that govern immunosuppression in pancreatic cancer. Understanding monocyte-exosome interactions could lead to novel immunotherapies for this disease.
Collapse
Affiliation(s)
- Naureen Javeed
- Department of Biochemistry and Molecular Biology, Mayo Clinic , Rochester, MN, USA
| | - Michael P Gustafson
- Human Cellular Therapy Laboratory, Division of Transfusion Medicine, Department of Laboratory Medicine and Pathology, Mayo Clinic , Rochester, MN, USA
| | - Shamit K Dutta
- Department of Biochemistry and Molecular Biology, Mayo Clinic , Rochester, MN, USA
| | - Yi Lin
- Division of Hematology, Mayo Clinic , Rochester, MN, USA
| | - William R Bamlet
- Department of Health Sciences Research, Mayo Clinic , Rochester, MN, USA
| | - Ann L Oberg
- Department of Health Sciences Research, Mayo Clinic , Rochester, MN, USA
| | - Gloria M Petersen
- Department of Health Sciences Research, Mayo Clinic , Rochester, MN, USA
| | - Suresh T Chari
- Division of Gastroenterology and Hepatology, Mayo Clinic , Rochester, MN, USA
| | - Allan B Dietz
- Human Cellular Therapy Laboratory, Division of Transfusion Medicine, Department of Laboratory Medicine and Pathology, Mayo Clinic , Rochester, MN, USA
| | | |
Collapse
|
79
|
Shipp C, Speigl L, Janssen N, Martens A, Pawelec G. A clinical and biological perspective of human myeloid-derived suppressor cells in cancer. Cell Mol Life Sci 2016; 73:4043-61. [PMID: 27236468 PMCID: PMC11108339 DOI: 10.1007/s00018-016-2278-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 04/26/2016] [Accepted: 05/17/2016] [Indexed: 12/14/2022]
Abstract
Considering the large number of studies focused on myeloid-derived suppressor cells (MDSCs) to date, only a handful of well-defined relationships in human cancer have been established. The difficulty of assessing the impact of MDSCs in human cancer is partly due to the relatively small number of studies performed in humans. This is compounded in the literature by a common lack of clear indication of which species is being referred to for each characteristic described. These aspects may result in inappropriate extrapolation of animal studies to those in the human setting. This is especially the case for studies focused on investigating therapies which can be used to target MDSCs or those aimed at understanding their mechanism. Here, we attempt to rectify this by reviewing only studies on MDSC performed in humans. We survey studies which explore (1) whether MDSC levels are altered in cancer patients and if this is correlated with patient survival, (2) the so far identified mechanisms employed by MDSC to exert immune suppression, and (3) whether therapeutic agents can be used to target MDSCs by either altering their level, influencing their differentiation or inhibiting their suppressive function. Despite the fact that these studies clearly show that MDSCs are important in human cancer, the clinical employment of agents intended to target them has not yet been accomplished. We identify factors which have contributed to this and propose steps which may facilitate the translation of these therapies to the clinic in future.
Collapse
Affiliation(s)
- Christopher Shipp
- Second Department of Internal Medicine, University Hospital Tübingen, Waldhörnlestr. 22, 72072, Tübingen, Germany.
| | - Lisa Speigl
- Second Department of Internal Medicine, University Hospital Tübingen, Waldhörnlestr. 22, 72072, Tübingen, Germany
| | - Nicole Janssen
- Second Department of Internal Medicine, University Hospital Tübingen, Waldhörnlestr. 22, 72072, Tübingen, Germany
| | - Alexander Martens
- Second Department of Internal Medicine, University Hospital Tübingen, Waldhörnlestr. 22, 72072, Tübingen, Germany
- Department of Dermatology, University Hospital Tübingen, Liebermeisterstr. 24, 72076, Tübingen, Germany
| | - Graham Pawelec
- Second Department of Internal Medicine, University Hospital Tübingen, Waldhörnlestr. 22, 72072, Tübingen, Germany.
- School of Science and Technology, College of Arts and Science, Nottingham Trent University, Burton St, Nottingham, NG1 4BU, UK.
| |
Collapse
|
80
|
Okła K, Wertel I, Polak G, Surówka J, Wawruszak A, Kotarski J. Tumor-Associated Macrophages and Myeloid-Derived Suppressor Cells as Immunosuppressive Mechanism in Ovarian Cancer Patients: Progress and Challenges. Int Rev Immunol 2016; 35:372-385. [PMID: 27644763 DOI: 10.1080/08830185.2016.1206097] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cancers are complex masses of malignant cells and nonmalignant cells that create the tumor microenvironment (TME). Non-transformed cells of the TME such as tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs) have been observed in the TME of ovarian cancer (OC) patients. Although these subsets may contribute to each step of carcinogenesis and are commonly associated with poor prognosis, still little is known about creation of the protumor microenvironment in OC. In this review, we focused on the nature and prognostic significance of TAMs and MDSCs in OC patients. Moreover, we discuss the main problems and challenges that must be overcome by researchers and clinicians to enrich our knowledge about the immunosuppressive microenvironment of cancers.
Collapse
Affiliation(s)
- Karolina Okła
- a Department of Oncological Gynaecology and Gynaecology , Medical University , Lublin , Poland
| | - Iwona Wertel
- a Department of Oncological Gynaecology and Gynaecology , Medical University , Lublin , Poland
| | - Grzegorz Polak
- a Department of Oncological Gynaecology and Gynaecology , Medical University , Lublin , Poland
| | - Justyna Surówka
- a Department of Oncological Gynaecology and Gynaecology , Medical University , Lublin , Poland
| | - Anna Wawruszak
- b Department of Biochemistry and Molecular Biology , Medical University , Lublin , Poland
| | - Jan Kotarski
- a Department of Oncological Gynaecology and Gynaecology , Medical University , Lublin , Poland
| |
Collapse
|
81
|
Hepatic carcinoma-associated fibroblasts enhance immune suppression by facilitating the generation of myeloid-derived suppressor cells. Oncogene 2016; 36:1090-1101. [DOI: 10.1038/onc.2016.273] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Revised: 06/03/2016] [Accepted: 06/06/2016] [Indexed: 02/06/2023]
|
82
|
Lin D, Wang X, Choi SYC, Ci X, Dong X, Wang Y. Immune phenotypes of prostate cancer cells: Evidence of epithelial immune cell-like transition? Asian J Urol 2016; 3:195-202. [PMID: 29264187 PMCID: PMC5730833 DOI: 10.1016/j.ajur.2016.08.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Revised: 07/19/2016] [Accepted: 08/09/2016] [Indexed: 12/19/2022] Open
Abstract
Prostate cancers (PCa) have been reported to actively suppress antitumor immune responses by creating an immune-suppressive microenvironment. There is mounting evidence that PCas may undergo an ''Epithelial Immune Cell-like Transition'' (EIT) by expressing molecules conventionally associated with immune cells (e.g., a variety of cytokines/receptors, immune transcription factors, Ig motifs, and immune checkpoint molecules), which subsequently results in the suppression of anti-cancer immune activity within the tumor microenvironment. Recent progress within the field of immune therapy has underscored the importance of immune checkpoint molecules in cancer development, thus leading to the development of novel immunotherapeutic approaches. Here, we review the expression of select immune checkpoint molecules in PCa epithelial and associated immune cells, with particular emphasis on clinical data supporting the concept of an EIT-mediated phenotype in PCa. Furthermore, we summarize current advances in anti-immune checkpoint therapies, and provide perspectives on their potential applicability.
Collapse
Affiliation(s)
- Dong Lin
- Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC, Canada
| | - Xinya Wang
- Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | | | - Xinpei Ci
- Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Xin Dong
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC, Canada
| | - Yuzhuo Wang
- Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC, Canada
- Corresponding author. Department of Urologic Sciences/Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada. Fax: +1 604 675 8019.Department of Urologic Sciences/Vancouver Prostate CentreUniversity of British Columbia2660 Oak StreetVancouverBCV6H 3Z6Canada
| |
Collapse
|
83
|
McClure C, McPeak MB, Youssef D, Yao ZQ, McCall CE, El Gazzar M. Stat3 and C/EBPβ synergize to induce miR-21 and miR-181b expression during sepsis. Immunol Cell Biol 2016; 95:42-55. [PMID: 27430527 PMCID: PMC5209283 DOI: 10.1038/icb.2016.63] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 06/26/2016] [Accepted: 07/12/2016] [Indexed: 12/21/2022]
Abstract
Myeloid-derived suppressor cells (MDSCs) increase late sepsis immunosuppression and mortality in mice. We reported that microRNA (miR) 21 and miR-181b expression in Gr1+CD11b+ myeloid progenitors increase septic MDSCs in mice by arresting macrophage and dendritic cell differentiation. Here, we report how sepsis regulates miR-21 and miR-181b transcription. In vivo and in vitro binding studies have shown that C/EBPα transcription factor, which promotes normal myeloid cell differentiation, binds both miRNA promoters in Gr1+CD11b+ cells from sham mice. In contrast, in sepsis Gr1+CD11b+ MDSCs miR-21 and miR-181b promoters bind both transcription factors Stat3 and C/EBPβ, which co-imunoprecipitate as a single complex. Mechanistically, transcription factor Rb phosphorylation supports Stat3 and C/EBPβ accumulation at both miRNA promoters, and C/EBPβ or Stat3 depletion by siRNA in sepsis Gr1+CD11b+ MDSCs inhibits miR-21 and miR-181b expression. To further support this molecular path for MDSC accumulation, we found that Stat3 and C/EBP binding at miR-21 or miR-181b promoter was induced by IL-6, using a luciferase reporter gene transfection into naive Gr1+CD11b+ cells. Identifying how sepsis MDSCs are generated may inform new treatments to reverse sepsis immunosuppression.
Collapse
Affiliation(s)
- Clara McClure
- Department of Internal Medicine, East Tennessee State University College of Medicine, Johnson City, TN, USA
| | - Melissa B McPeak
- Department of Internal Medicine, East Tennessee State University College of Medicine, Johnson City, TN, USA
| | - Dima Youssef
- Department of Internal Medicine, East Tennessee State University College of Medicine, Johnson City, TN, USA
| | - Zhi Q Yao
- Department of Internal Medicine, East Tennessee State University College of Medicine, Johnson City, TN, USA
| | - Charles E McCall
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Mohamed El Gazzar
- Department of Internal Medicine, East Tennessee State University College of Medicine, Johnson City, TN, USA
| |
Collapse
|
84
|
Pyzer AR, Cole L, Rosenblatt J, Avigan DE. Myeloid-derived suppressor cells as effectors of immune suppression in cancer. Int J Cancer 2016; 139:1915-26. [PMID: 27299510 DOI: 10.1002/ijc.30232] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 06/07/2016] [Accepted: 06/08/2016] [Indexed: 12/11/2022]
Abstract
The tumor microenvironment consists of an immunosuppressive niche created by the complex interactions between cancer cells and surrounding stromal cells. A critical component of this environment are myeloid-derived suppressor cells (MDSCs), a heterogeneous group of immature myeloid cells arrested at different stages of differentiation and expanded in response to a variety of tumor factors. MDSCs exert diverse effects in modulating the interactions between immune effector cells and the malignant cells. An increased presence of MDSCs is associated with tumor progression, poorer outcomes, and decreased effectiveness of immunotherapeutic strategies. In this article, we will review our current understanding of the mechanisms that underlie MDSC expansion and their immune-suppressive function. Finally, we review the preclinical studies and clinical trials that have attempted to target MDSCs, in order to improve responses to cancer therapies.
Collapse
Affiliation(s)
- Athalia Rachel Pyzer
- Bone Marrow Transplant, Beth Israel Deaconess Medical Center, Center for Life Sciences, CLS724, Boston, MA
| | - Leandra Cole
- Bone Marrow Transplant, Beth Israel Deaconess Medical Center, Center for Life Sciences, CLS724, Boston, MA
| | - Jacalyn Rosenblatt
- Bone Marrow Transplant, Beth Israel Deaconess Medical Center, Center for Life Sciences, CLS724, Boston, MA
| | - David E Avigan
- Bone Marrow Transplant, Beth Israel Deaconess Medical Center, Center for Life Sciences, CLS724, Boston, MA
| |
Collapse
|
85
|
Khanolkar RC, Kalogeropoulos M, Lawrie A, Roghanian A, Vickers MA, Young NT. Leukocyte Ig-Like receptor B1 restrains dendritic cell function through increased expression of the NF-κB regulator ABIN1/TNIP1. J Leukoc Biol 2016; 100:737-746. [PMID: 27129285 DOI: 10.1189/jlb.1a0915-420rrr] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 04/06/2016] [Indexed: 12/14/2022] Open
Abstract
Inhibitory receptors of the human leukocyte immunoglobulin-like receptor family are constitutively expressed on all myeloid cell types and regulate their functional activity. We demonstrate that ligation of the human leukocyte antigen class I-specific receptor LILRB1, during the differentiation of monocytes to dendritic cells in vitro, results in increased expression of the nuclear factor κB inhibitor protein ABIN1 (also known as TNIP1). Similarly increased expression of ABIN1/TNIP1 was observed in the "immunosuppressive" monocyte populations of patients with non-Hodgkin lymphoma ex vivo. Reducing expression of ABIN1/TNIP1 using small interfering ribonucleic acid allows dendritic cells and immunosuppressive monocytes to respond to stimulation by allowing nuclear factor κB translocation to the nucleus (P < 0.001), increasing cell surface expression of antigen presentation and costimulatory molecules (P < 0.01), increasing phagocytic capacity (P < 0.001), secreting proinflammatory cytokines (P < 0.01), and an increasing ability to stimulate T cell responses (P < 0.05). Our study, therefore, identifies an important functional role for ABIN1/TNIP1 in mediating the effects of LILRB1 ligation-induced inhibitory effects on immune responses. Our findings suggest that inhibiting the LILRB1-ABIN1/TNIP1 pathway in antigen-presenting cells could be a therapeutic approach to stimulate antitumor immune responses. Conversely, stimulation of the pathway may also ameliorate autoimmune diseases in which TNIP1 is a susceptibility gene.
Collapse
Affiliation(s)
- Rahul C Khanolkar
- Division of Applied Medicine, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom;
| | - Michail Kalogeropoulos
- Division of Applied Medicine, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Alistair Lawrie
- Division of Applied Medicine, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Ali Roghanian
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA; and Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Mark A Vickers
- Division of Applied Medicine, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Neil T Young
- Division of Applied Medicine, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| |
Collapse
|
86
|
Yin J, Wang C, Huang M, Mao X, Zhou J, Zhang Y. Circulating CD14(+) HLA-DR(-/low) myeloid-derived suppressor cells in leukemia patients with allogeneic hematopoietic stem cell transplantation: novel clinical potential strategies for the prevention and cellular therapy of graft-versus-host disease. Cancer Med 2016; 5:1654-69. [PMID: 27109254 PMCID: PMC4944894 DOI: 10.1002/cam4.688] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 01/27/2016] [Accepted: 02/02/2016] [Indexed: 12/21/2022] Open
Abstract
Myeloid‐derived suppressor cells (MDSCs) are a heterogeneous cell population that includes immature myeloid cells and the progenitor cells of macrophages, dendritic cells (DCs), monocytes, and neutrophils. The expansion and functional importance of MDSCs in patients with cancer and noncancer pathogenic conditions has been recognized. As a result, there has been growing interest in understanding their roles in acute graft‐versus‐host disease (aGVHD) after allogenetic hematopoietic stem cell transplantation (allo‐HSCT). In order to evaluate possible effects of MDSCs on aGVHD development and clinical outcomes, this study systematically detected the dynamic changes of MDSCs accumulation in patients during the first 100 days after allo‐HSCT, and investigated the levels of other cell types and relative cytokines during MDSCs accumulation. Results showed that accumulation of MDSCs in the graft and in peripheral blood when engraftment might contribute to patients' overall immune suppression and result in the successful control of severe aGVHD and long‐term survival without influence on risk of recurrence after allo‐HSCT. But MDSCs levels in the graft had more favorable predictive abilities. Furthermore, MDSCs proportion significantly increased in patients developing aGVHD after allo‐HSCT. It might be caused by secondary inflammatory response, especially related to high concentrations of IL‐6 and TNF‐α. But this accumulation would not be able to counterbalance the aggravation of aGVHD and would not have influence on clinical outcomes and risk of relapse. Overall, MDSCs might be considered as potential new therapeutic option for aGVHD and achieve long‐term immunological tolerance and survival.
Collapse
Affiliation(s)
- Jin Yin
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chunyan Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Min Huang
- Clinical Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xia Mao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jianfeng Zhou
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yicheng Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
87
|
Wang D, An G, Xie S, Yao Y, Feng G. The clinical and prognostic significance of CD14+HLA-DR−/low myeloid-derived suppressor cells in hepatocellular carcinoma patients receiving radiotherapy. Tumour Biol 2016; 37:10427-33. [DOI: 10.1007/s13277-016-4916-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 01/25/2016] [Indexed: 12/17/2022] Open
|
88
|
Wichert S, Pettersson Å, Hellmark T, Johansson Å, Hansson M. Phagocyte function decreases after high-dose treatment with melphalan and autologous stem cell transplantation in patients with multiple myeloma. Exp Hematol 2016; 44:342-351.e5. [PMID: 26774385 DOI: 10.1016/j.exphem.2016.01.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 12/19/2015] [Accepted: 01/06/2016] [Indexed: 11/28/2022]
Abstract
High-dose melphalan with autologous hematopoietic stem cell transplantation (ASCT) is the standard of care for younger patients with newly diagnosed multiple myeloma and is aimed at achieving as deep and complete a response as possible after various combinations of induction therapy. However, it is frequently associated with infectious complications. This study investigated the effects of high-dose treatment with autologous stem cell support on patients' innate immunity, with a focus on subpopulations and functioning of recently released polymorphonuclear leukocytes (PMNs) and monocytes in peripheral blood. Flow cytometry-based analysis was used to measure the degree of PMN maturation and activation, before and after ASCT and compared with healthy controls. After high-dose treatment and ASCT, a smaller proportion of patients' PMNs had the capacity for oxidative burst. Moreover, patients' PMNs, both before and after ASCT, had a reduced capacity for phagocytosis. Eosinophils, which recently have been suggested to play a role in promoting malignant plasma cell proliferation, were markedly reduced after ASCT, with slow regeneration. HLA-DR expression by monocytes was significantly depressed after ASCT, a characteristic often attributed to monocytic myeloid-derived suppressor cells. Our results suggest that several aspects of phagocytic function are impaired for at least 20 days after ASCT.
Collapse
Affiliation(s)
- Stina Wichert
- Department of Hematology, Skåne University Hospital and Lund University, Lund, Sweden.
| | - Åsa Pettersson
- Department of Nephrology, Clinical Sciences in Lund, Lund University, Lund, Sweden
| | - Thomas Hellmark
- Department of Nephrology, Clinical Sciences in Lund, Lund University, Lund, Sweden
| | - Åsa Johansson
- Department of Hematology, Skåne University Hospital and Lund University, Lund, Sweden; Clinical Immunology and Transfusion Medicine, University and Regional Laboratories Region Skåne, Lund, Sweden
| | - Markus Hansson
- Department of Hematology, Skåne University Hospital and Lund University, Lund, Sweden
| |
Collapse
|
89
|
Younis RH, Han KL, Webb TJ. Human Head and Neck Squamous Cell Carcinoma-Associated Semaphorin 4D Induces Expansion of Myeloid-Derived Suppressor Cells. THE JOURNAL OF IMMUNOLOGY 2016; 196:1419-29. [PMID: 26740106 DOI: 10.4049/jimmunol.1501293] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 11/25/2015] [Indexed: 01/05/2023]
Abstract
One of the mechanisms by which malignancies can induce immune suppression is through the production of cytokines that affect the maturation and differentiation of inflammatory cells in the tumor microenvironment. Semaphorin 4D (Sema4D) is a proangiogenic cytokine produced by several malignancies, which has been described in the regulation of the immune system. In the present study, we examined the role of human head and neck squamous cell carcinoma (HNSCC)-secreted Sema4D on myeloid cell differentiation. CD33(+) cells cultured in HNSCC cell line-derived conditioned medium differentiated into myeloid derived suppressor cells (MDSC) (CD33(+)CD11b(+)HLA-DR(-/low)). The addition of anti-Sema4D Ab to HNSCC conditioned medium significantly reduced the expansion of the MDSC population. Similarly, knockdown of Sema4D in an HNSCC cell line resulted in a loss of MDSC function as shown by a decrease in the production of the immune-suppressive cytokines arginase-1, TGF-β, and IL-10 by MDSC, concomitant with recovery of T cell proliferation and IFN-γ production following stimulation of CD3/CD28. Importantly, CD33(+) myeloid and T cells cultured in conditioned medium of HNSCC cells in which Sema4D was knocked down promoted antitumor inflammatory profile, through recovery of the effector T cells (CD4(+)T-bet(+) and CD8(+)T-bet(+)), as well as a decrease in regulatory T cells (CD4(+)CD25(+)FOXP3(+)). We also showed that Sema4D was comparable to GM-CSF in its induction of MDSC. Collectively, this study describes a novel immunosuppressive role for Sema4D in HNSCC through induction of MDSC, and it highlights Sema4D as a therapeutic target for future studies to enhance the antitumorigenic inflammatory response in HNSCC and other epithelial malignancies.
Collapse
Affiliation(s)
- Rania H Younis
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, Baltimore, MD 21201; Marlene and Stewart Greenebaum Cancer Center, University of Maryland, Baltimore, Baltimore, MD 21201; and
| | - Kyu Lee Han
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, Baltimore, MD 21201
| | - Tonya J Webb
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland, Baltimore, Baltimore, MD 21201; and Department of Microbiology and Immunology, University of Maryland, Baltimore, Baltimore, MD 21201
| |
Collapse
|
90
|
Wang G, Lu X, Dey P, Deng P, Wu CC, Jiang S, Fang Z, Zhao K, Konaparthi R, Hua S, Zhang J, Li-Ning-Tapia EM, Kapoor A, Wu CJ, Patel NB, Guo Z, Ramamoorthy V, Tieu TN, Heffernan T, Zhao D, Shang X, Khadka S, Hou P, Hu B, Jin EJ, Yao W, Pan X, Ding Z, Shi Y, Li L, Chang Q, Troncoso P, Logothetis CJ, McArthur MJ, Chin L, Wang YA, DePinho RA. Targeting YAP-Dependent MDSC Infiltration Impairs Tumor Progression. Cancer Discov 2015; 6:80-95. [PMID: 26701088 DOI: 10.1158/2159-8290.cd-15-0224] [Citation(s) in RCA: 375] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 10/16/2015] [Indexed: 12/20/2022]
Abstract
UNLABELLED The signaling mechanisms between prostate cancer cells and infiltrating immune cells may illuminate novel therapeutic approaches. Here, utilizing a prostate adenocarcinoma model driven by loss of Pten and Smad4, we identify polymorphonuclear myeloid-derived suppressor cells (MDSC) as the major infiltrating immune cell type, and depletion of MDSCs blocks progression. Employing a novel dual reporter prostate cancer model, epithelial and stromal transcriptomic profiling identified CXCL5 as a cancer-secreted chemokine to attract CXCR2-expressing MDSCs, and, correspondingly, pharmacologic inhibition of CXCR2 impeded tumor progression. Integrated analyses identified hyperactivated Hippo-YAP signaling in driving CXCL5 upregulation in cancer cells through the YAP-TEAD complex and promoting MDSC recruitment. Clinicopathologic studies reveal upregulation and activation of YAP1 in a subset of human prostate tumors, and the YAP1 signature is enriched in primary prostate tumor samples with stronger expression of MDSC-relevant genes. Together, YAP-driven MDSC recruitment via heterotypic CXCL5-CXCR2 signaling reveals an effective therapeutic strategy for advanced prostate cancer. SIGNIFICANCE We demonstrate a critical role of MDSCs in prostate tumor progression and discover a cancer cell nonautonomous function of the Hippo-YAP pathway in regulation of CXCL5, a ligand for CXCR2-expressing MDSCs. Pharmacologic elimination of MDSCs or blocking the heterotypic CXCL5-CXCR2 signaling circuit elicits robust antitumor responses and prolongs survival.
Collapse
Affiliation(s)
- Guocan Wang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xin Lu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Prasenjit Dey
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Pingna Deng
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chia Chin Wu
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shan Jiang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zhuangna Fang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas. Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Kun Zhao
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ramakrishna Konaparthi
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sujun Hua
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jianhua Zhang
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elsa M Li-Ning-Tapia
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Avnish Kapoor
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chang-Jiun Wu
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Neelay Bhaskar Patel
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zhenglin Guo
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Vandhana Ramamoorthy
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Trang N Tieu
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tim Heffernan
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Di Zhao
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xiaoying Shang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sunada Khadka
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Pingping Hou
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Baoli Hu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Eun-Jung Jin
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Biological Science, College of Natural Sciences, Wonkwang University, Cheonbuk, Iksan, South Korea
| | - Wantong Yao
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xiaolu Pan
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zhihu Ding
- Sanofi Oncology, Cambridge, Massachusetts
| | - Yanxia Shi
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas. Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Liren Li
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas. Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Qing Chang
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Patricia Troncoso
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Christopher J Logothetis
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mark J McArthur
- Department of Veterinary Medicine and Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lynda Chin
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Y Alan Wang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Ronald A DePinho
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
91
|
Gato M, Blanco-Luquin I, Zudaire M, de Morentin XM, Perez-Valderrama E, Zabaleta A, Kochan G, Escors D, Fernandez-Irigoyen J, Santamaría E. Drafting the proteome landscape of myeloid-derived suppressor cells. Proteomics 2015; 16:367-78. [PMID: 26403437 DOI: 10.1002/pmic.201500229] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 08/18/2015] [Accepted: 09/21/2015] [Indexed: 01/12/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of cells that are defined by their myeloid origin, immature state, and ability to potently suppress T-cell responses. They regulate immune responses and the population significantly increases in the tumor microenvironment of patients with glioma and other malignant tumors. For their study, MDSCs are usually isolated from the spleen or directly of tumors from a large number of tumor-bearing mice although promising ex vivo differentiated MDSC production systems have been recently developed. During the last years, proteomics has emerged as a powerful approach to analyze MDSCs proteomes using shotgun-based mass spectrometry (MS), providing functional information about cellular homeostasis and metabolic state at a global level. Here, we will revise recent proteome profiling studies performed in MDSCs from different origins. Moreover, we will perform an integrative functional analysis of the protein compilation derived from these large-scale proteomic studies in order to obtain a comprehensive view of MDSCs biology. Finally, we will also discuss the potential application of high-throughput proteomic approaches to study global proteome dynamics and post-translational modifications (PTMs) during the differentiation process of MDSCs that will greatly boost the identification of novel MDSC-specific therapeutic targets to apply in cancer immunotherapy.
Collapse
Affiliation(s)
- María Gato
- Immunomodulation Laboratory, Navarrabiomed, Fundación Miguel Servet, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Idoia Blanco-Luquin
- Immunomodulation Laboratory, Navarrabiomed, Fundación Miguel Servet, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Maribel Zudaire
- Immunomodulation Laboratory, Navarrabiomed, Fundación Miguel Servet, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Xabier Martínez de Morentin
- Proteomics Unit, Navarrabiomed, Fundación Miguel Servet, ProteoRed-ISCIII, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Estela Perez-Valderrama
- Proteomics Unit, Navarrabiomed, Fundación Miguel Servet, ProteoRed-ISCIII, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Aintzane Zabaleta
- Biofunctional Nanomaterials Laboratory, CIC Biomagune, San Sebastian, Spain
| | - Grazyna Kochan
- Immunomodulation Laboratory, Navarrabiomed, Fundación Miguel Servet, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - David Escors
- Immunomodulation Laboratory, Navarrabiomed, Fundación Miguel Servet, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Joaquín Fernandez-Irigoyen
- Proteomics Unit, Navarrabiomed, Fundación Miguel Servet, ProteoRed-ISCIII, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Enrique Santamaría
- Proteomics Unit, Navarrabiomed, Fundación Miguel Servet, ProteoRed-ISCIII, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| |
Collapse
|
92
|
The immunobiology of myeloid-derived suppressor cells in cancer. Tumour Biol 2015; 37:1387-406. [PMID: 26611648 DOI: 10.1007/s13277-015-4477-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 11/19/2015] [Indexed: 12/31/2022] Open
Abstract
The tumor microenvironment is a complex and heterogeneous milieu in which multiple interactions occur between tumor and host cells. Immunosuppressive cells which are present in this microenvironment, such as regulatory T (Treg) cells and myeloid-derived suppressor cells (MDSCs), play an important role in tumor progression, via down-regulation of antitumor responses. MDSCs represent a heterogeneous group of cells originated from the myeloid lineage that are in the immature state. These cells markedly accumulate under pathologic conditions, such as cancer, infection, and inflammation, and use various mechanisms to inhibit both adaptive and innate immune responses. These immunosuppressive mechanisms include deprivation of T cells from essential amino acids, induction of oxidative stress, interference with viability and trafficking of T cells, induction of immunosuppressive cells, and finally polarizing immunity toward a tumor-promoting type 2 phenotype. In addition to suppression of antitumor immune responses, MDSCs can also enhance the tumor metastasis and angiogenesis. Previous studies have shown that increased frequency of MDSCs is related to the tumor progression. Moreover, various drugs that directly target these cells or reverse their suppressive activity can improve antitumor immune responses as well as increase the efficacy of immunotherapeutic intervention. In this review, we will first discuss on the immunobiology of MDSCs in an attempt to find the role of these cells in tumor progression and then discuss about therapeutic approaches to target these cells.
Collapse
|
93
|
Lesinski GB, Reville PK, Mace TA, Young GS, Ahn-Jarvis J, Thomas-Ahner J, Vodovotz Y, Ameen Z, Grainger E, Riedl K, Schwartz S, Clinton SK. Consumption of soy isoflavone enriched bread in men with prostate cancer is associated with reduced proinflammatory cytokines and immunosuppressive cells. Cancer Prev Res (Phila) 2015; 8:1036-44. [PMID: 26276751 PMCID: PMC4633400 DOI: 10.1158/1940-6207.capr-14-0464] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Accepted: 07/21/2015] [Indexed: 01/13/2023]
Abstract
We hypothesized that soy phytochemicals may have immunomodulatory properties that may affect prostate carcinogenesis and progression. A randomized, phase II trial was conducted in 32 patients with prostate cancer with asymptomatic biochemical recurrence but no measurable disease on standard staging studies. Patients were randomized to two slices of soy bread (34 mg isoflavones/slice) or soy bread containing almond powder daily as a source of β-glucosidase. Flow cytometry and bioplex assays were used to measure cytokines or immune cell phenotype in blood at baseline (day 0) and following intervention (day 56). Adequate blood samples were available at enrollment and day 56 and evaluated. Multiple plasma cytokines and chemokines were significantly decreased on day 56 versus baseline. Subgroup analysis indicated reduced TH1 (P = 0.028) and myeloid-derived suppressor cell (MDSC)-associated cytokines (P = 0.035). TH2 and TH17 cytokines were not significantly altered. Phenotypic analysis revealed no change in CD8(+) or CD4(+) T cells but showed increased CD56(+) natural killer (NK) cells (P = 0.038). The percentage of cells with a T regulatory cell phenotype (CD4(+)CD25(+)FoxP3(+)) was significantly decreased after 56 days of soy bread (P = 0.0136). Significantly decreased monocytic (CD33(+)HLADR(neg)CD14(+)) MDSC were observed in patients consuming soy bread (P = 0.0056). These data suggest that soy bread modulates systemic soluble and cellular biomarkers consistent with limiting inflammation and suppression of MDSCs. Additional studies to elucidate impact on the carcinogenic process or as a complement to immune-based therapy are required.
Collapse
Affiliation(s)
- Gregory B Lesinski
- Department of Internal Medicine, Division of Medical Oncology, The Arthur G. James and Richard Solove Research Institute, Columbus, Ohio. The Ohio State University Comprehensive Cancer Center, Columbus, Ohio.
| | - Patrick K Reville
- Department of Internal Medicine, Division of Medical Oncology, The Arthur G. James and Richard Solove Research Institute, Columbus, Ohio
| | - Thomas A Mace
- Department of Internal Medicine, Division of Medical Oncology, The Arthur G. James and Richard Solove Research Institute, Columbus, Ohio
| | - Gregory S Young
- Center for Biostatistics, The Ohio State University, Columbus, Ohio
| | - Jennifer Ahn-Jarvis
- College of Food, Agricultural and Environmental Science, Department of Food Science and Technology, The Ohio State University, Columbus, Ohio
| | - Jennifer Thomas-Ahner
- Department of Internal Medicine, Division of Medical Oncology, The Arthur G. James and Richard Solove Research Institute, Columbus, Ohio
| | - Yael Vodovotz
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio. College of Food, Agricultural and Environmental Science, Department of Food Science and Technology, The Ohio State University, Columbus, Ohio
| | - Zeenath Ameen
- Department of Internal Medicine, Division of Medical Oncology, The Arthur G. James and Richard Solove Research Institute, Columbus, Ohio
| | - Elizabeth Grainger
- Department of Internal Medicine, Division of Medical Oncology, The Arthur G. James and Richard Solove Research Institute, Columbus, Ohio
| | - Kenneth Riedl
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio. College of Food, Agricultural and Environmental Science, Department of Food Science and Technology, The Ohio State University, Columbus, Ohio
| | - Steven Schwartz
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio. College of Food, Agricultural and Environmental Science, Department of Food Science and Technology, The Ohio State University, Columbus, Ohio
| | - Steven K Clinton
- Department of Internal Medicine, Division of Medical Oncology, The Arthur G. James and Richard Solove Research Institute, Columbus, Ohio. The Ohio State University Comprehensive Cancer Center, Columbus, Ohio.
| |
Collapse
|
94
|
Meirow Y, Kanterman J, Baniyash M. Paving the Road to Tumor Development and Spreading: Myeloid-Derived Suppressor Cells are Ruling the Fate. Front Immunol 2015; 6:523. [PMID: 26528286 PMCID: PMC4601280 DOI: 10.3389/fimmu.2015.00523] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 09/24/2015] [Indexed: 01/19/2023] Open
Abstract
Cancer development is dependent on intrinsic cellular changes as well as inflammatory factors in the tumor macro and microenvironment. The inflammatory milieu nourishes the tumor and contributes to cancer progression. Numerous studies, including ours, have demonstrated that the tumor microenvironment is immunosuppressive, impairing the anticancer immune responses. Chronic inflammation was identified as the key process responsible for this immunosuppression via induction of immature myeloid-derived suppressor cells (MDSCs). Upon a prolonged immune response, MDSCs are polarized toward immunosuppressive cells meant to control the exacerbated immune response. In cancer, the chronic inflammatory response renders the MDSCs harmful. Polarized MDSCs suppress T-cells and natural killer cells, as well as antigen-presenting cells, abrogating the beneficial immune response. These changes in the immunological milieu could also lead to high frequency of mutations, enhanced cancer cell stemness, and angiogenesis, directly supporting tumor initiation, growth, and spreading. The presence of MDSCs in cancer poses a serious obstacle in a variety of immune-based therapies, which rely on the stimulation of antitumor immune responses. Cumulative data, including our own, suggest that the selection of an appropriate and effective anticancer therapy must take into consideration the host’s immune status as well as tumor-related parameters. Merging biomarkers for immune monitoring into the traditional patient’s categorization and follow-up can provide new predictive and diagnostic tools to the clinical practice. Chronic inflammation and MDSCs could serve as novel targets for therapeutic interventions, which can be combined with conventional cancer treatments such as chemotherapy, radiotherapy, and cancer cell-targeted and immune-based therapies. Intervention in environmental and tumor-specific inflammatory mechanisms will allow better clinical management of cancer toward more efficient treatment.
Collapse
Affiliation(s)
- Yaron Meirow
- The Lautenberg Center for General and Tumor Immunology, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University , Jerusalem , Israel
| | - Julia Kanterman
- The Lautenberg Center for General and Tumor Immunology, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University , Jerusalem , Israel
| | - Michal Baniyash
- The Lautenberg Center for General and Tumor Immunology, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University , Jerusalem , Israel
| |
Collapse
|
95
|
Stanojevic I, Miller K, Kandolf-Sekulovic L, Mijuskovic Z, Zolotarevski L, Jovic M, Gacevic M, Djukic M, Arsenijevic N, Vojvodic D. A subpopulation that may correspond to granulocytic myeloid-derived suppressor cells reflects the clinical stage and progression of cutaneous melanoma. Int Immunol 2015; 28:87-97. [PMID: 26391013 DOI: 10.1093/intimm/dxv053] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 09/11/2015] [Indexed: 12/12/2022] Open
Abstract
Seventy-eight melanoma patients and 10 healthy individuals were examined. Follow-up examinations of all melanoma patients were performed regularly every three months. Myeloid-derived suppressor cells (MDSC) were defined as lineage negative (CD3(-), CD19(-), CD56(-)), HLA-DR(-/low), CD11b(+) and CD33(+). Classification of granulocytic (GrMDSC) and monocytic (MoMDSC) subsets was based on the CD15 and CD14 expression, respectively. Unlike the MoMDSC, that were present in 60% of healthy controls and 15% of melanoma patients, the GrMDSC were present in all examined participants, and the melanoma patients were found to have statistically higher frequencies compared with healthy controls. Accordingly, we kept focused on GrMDSC frequencies in relation to the melanoma stages and course of the disease. The GrMDSC values are highest in stage IV melanoma patients, with statistical significance compared with stages IA, IB, IIA and IIB. Patients with progression had statistically higher GrMDSC counts comparing with those with stable disease (P = 0.0079). Patients who had progression-free interval (PFI) < 12 months showed significantly higher GrMDSC values compared with those with PFI > 12 months (P = 0.0333). GrMDSC showed significant negative correlation with PFI intervals (P = 0.0095). The GrMDSC subset was predominant in all our patients. We confirmed that GrMDSC do accumulate early in the peripheral blood of melanoma patients and their frequencies correlate narrowly with the clinical stage and the spread of the disease. The increase in GrMDSC frequencies correlates well with a progressive disease and could be considered a potential predictive biomarker of high-risk melanoma cases that are more likely to have a shorter PFI.
Collapse
Affiliation(s)
- Ivan Stanojevic
- Department of Clinical and Experimental Immunology, Institute for Medical Research, Military Medical Academy, Crnotravska 17, 11000 Belgrade, Serbia
| | - Karolina Miller
- Department of Histopathology, Dorset County Hospital NHS Foundation Trust, DT1 2JY Dorchester, UK
| | | | - Zeljko Mijuskovic
- Department of Dermatovenerology, Military Medical Academy, 11000 Belgrade, Serbia
| | | | - Milena Jovic
- Institute for Pathology, Military Medical Academy, 11000 Belgrade, Serbia
| | - Milomir Gacevic
- Clinic for Plastic and Reconstructive Surgery, Military Medical Academy, 11000 Belgrade, Serbia
| | - Mirjana Djukic
- Department of Toxicology, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia
| | | | - Danilo Vojvodic
- Department of Clinical and Experimental Immunology, Institute for Medical Research, Military Medical Academy, Crnotravska 17, 11000 Belgrade, Serbia
| |
Collapse
|
96
|
Pal SK, Kortylewski M. Breaking bad habits: Targeting MDSCs to alleviate immunosuppression in prostate cancer. Oncoimmunology 2015; 5:e1078060. [PMID: 27057455 DOI: 10.1080/2162402x.2015.1078060] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 07/22/2015] [Indexed: 10/23/2022] Open
Abstract
The myeloid-derived suppressor cells (MDSCs) contribute to tumor immune evasion and still remain an elusive therapeutic target. Our study identified granulocytic MDSCs accumulating in prostate cancer patients during disease progression. We demonstrate the feasibility of using STAT3siRNA-based strategy for targeting MDSCs to alleviate arginase-dependent suppression of T cell activity.
Collapse
Affiliation(s)
- Sumanta K Pal
- Department of Cancer Immunotherapeutics & Tumor Immunology at Beckman Research Institute, City of Hope National Medical Center , Duarte, CA, USA
| | - Marcin Kortylewski
- Department of Medical Oncology & Experimental Therapeutics, City of Hope National Medical Center , Duarte, CA, USA
| |
Collapse
|
97
|
Karakasheva TA, Waldron TJ, Eruslanov E, Kim SB, Lee JS, O'Brien S, Hicks PD, Basu D, Singhal S, Malavasi F, Rustgi AK. CD38-Expressing Myeloid-Derived Suppressor Cells Promote Tumor Growth in a Murine Model of Esophageal Cancer. Cancer Res 2015; 75:4074-85. [PMID: 26294209 DOI: 10.1158/0008-5472.can-14-3639] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 07/19/2015] [Indexed: 12/15/2022]
Abstract
Myeloid-derived suppressor cells (MDSC) are an immunosuppressive population of immature myeloid cells found in advanced-stage cancer patients and mouse tumor models. Production of inducible nitric oxide synthase (iNOS) and arginase, as well as other suppressive mechanisms, allows MDSCs to suppress T-cell-mediated tumor clearance and foster tumor progression. Using an unbiased global gene expression approach in conditional p120-catenin knockout mice (L2-cre;p120ctn(f/f)), a model of oral-esophageal cancer, we have identified CD38 as playing a vital role in MDSC biology, previously unknown. CD38 belongs to the ADP-ribosyl cyclase family and possesses both ectoenzyme and receptor functions. It has been described to function in lymphoid and early myeloid cell differentiation, cell activation, and neutrophil chemotaxis. We find that CD38 expression in MDSCs is evident in other mouse tumor models of esophageal carcinogenesis, and CD38(high) MDSCs are more immature than MDSCs lacking CD38 expression, suggesting a potential role for CD38 in the maturation halt found in MDSC populations. CD38(high) MDSCs also possess a greater capacity to suppress activated T cells, and promote tumor growth to a greater degree than CD38(low) MDSCs, likely as a result of increased iNOS production. In addition, we have identified novel tumor-derived factors, specifically IL6, IGFBP3, and CXCL16, which induce CD38 expression by MDSCs ex vivo. Finally, we have detected an expansion of CD38(+) MDSCs in peripheral blood of advanced-stage cancer patients and validated targeting CD38 in vivo as a novel approach to cancer therapy.
Collapse
Affiliation(s)
- Tatiana A Karakasheva
- Division of Gastroenterology, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania. Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Todd J Waldron
- Division of Gastroenterology, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania. Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Evgeniy Eruslanov
- Thoracic Surgery Research Laboratory, Department of Surgery, Hospital of the University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Sang-Bae Kim
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ju-Seog Lee
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shaun O'Brien
- Thoracic Surgery Research Laboratory, Department of Surgery, Hospital of the University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Philip D Hicks
- Division of Gastroenterology, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania. Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Devraj Basu
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania, Philadelphia, Pennsylvania. Surgery Service; Philadelphia Veterans Affairs Medical Center, Philadelphia, Pennsylvania
| | - Sunil Singhal
- Thoracic Surgery Research Laboratory, Department of Surgery, Hospital of the University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania. Surgery Service; Philadelphia Veterans Affairs Medical Center, Philadelphia, Pennsylvania
| | - Fabio Malavasi
- Lab of Immunogenetics, Department of Medical Sciences, University of Torino Medical School, Torino, Italy
| | - Anil K Rustgi
- Division of Gastroenterology, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania. Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania. Department of Genetics, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
98
|
Hingorani P, Maas ML, Gustafson MP, Dickman P, Adams RH, Watanabe M, Eshun F, Williams J, Seidel MJ, Dietz AB. Increased CTLA-4(+) T cells and an increased ratio of monocytes with loss of class II (CD14(+) HLA-DR(lo/neg)) found in aggressive pediatric sarcoma patients. J Immunother Cancer 2015; 3:35. [PMID: 26286851 PMCID: PMC4539889 DOI: 10.1186/s40425-015-0082-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 07/15/2015] [Indexed: 12/16/2022] Open
Abstract
Background There is little information regarding the composition of peripheral blood immunity in sarcoma patients and even less in the context of pediatric sarcomas. We describe the immune status using flow cytometry of peripheral blood in patients with osteosarcoma and Ewing sarcoma and demonstrate excessive CD14 in tumor tissues. Methods Peripheral blood from patients with OS and ES was collected at diagnosis or relapse, and used for immune phenotyping of 74 different leukocyte phenotypes. Blood from young adult healthy volunteers was collected as controls. Tumor tissues were analyzed by immunohistochemistry. Results Nineteen patients (average age = 14 y) and 16 controls (average age = 25y) were enrolled on study. Of the 74 phenotypes, 14 were different between sarcoma patients and HV. Sarcoma patients’ leukocytes contained a higher percentage of granulocytes (67 % sarcoma vs. 58 % HV; p = 0.003) and fewer lymphocytes (20 % sarcoma vs. 27 % HV; p = 0.001). Increased expression of CTLA-4 was seen in both T cells in sarcoma patients as compared to HV (p = 0.05). Increased CD14+ HLA-DRlo/neg immunosuppressive monocytes were seen in sarcoma patients (p = 0.03); primarily seen in OS. Increased tumor necrosis factor receptor II expression was seen on CD14+ cells derived from sarcoma patients as compared to HV (p = 0.01). Massive infiltration of CD14+ cells was seen in OS (>50 % of cells in the majority of tumors) compared to ES (<10-25 % of cells). In contrast, both OS and ES had limited T cell infiltration (generally <10 % of cells). Conclusions Pediatric sarcoma patients exhibit several immune phenotypic differences that were exacerbated in more severe disease. These phenotypes have the potential to contribute to immune suppression and may indicate potential targets for immune therapies. Electronic supplementary material The online version of this article (doi:10.1186/s40425-015-0082-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Pooja Hingorani
- Center for Cancer and Blood Disorders, Phoenix Children's Hospital, Phoenix, AZ USA
| | - Mary L Maas
- Human Cellular Therapy Lab, Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
| | - Michael P Gustafson
- Human Cellular Therapy Lab, Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
| | - Paul Dickman
- Department of Pathology, Phoenix Children's Hospital, Phoenix, AZ USA
| | - Roberta H Adams
- Center for Cancer and Blood Disorders, Phoenix Children's Hospital, Phoenix, AZ USA
| | - Masayo Watanabe
- Center for Cancer and Blood Disorders, Phoenix Children's Hospital, Phoenix, AZ USA
| | - Francis Eshun
- Center for Cancer and Blood Disorders, Phoenix Children's Hospital, Phoenix, AZ USA
| | - James Williams
- Center for Cancer and Blood Disorders, Phoenix Children's Hospital, Phoenix, AZ USA
| | | | - Allan B Dietz
- Human Cellular Therapy Lab, Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA.,Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, and Division of Immunology, Mayo Clinic, Rochester, MN USA
| |
Collapse
|
99
|
IL-10 induces the development of immunosuppressive CD14(+)HLA-DR(low/-) monocytes in B-cell non-Hodgkin lymphoma. Blood Cancer J 2015; 5:e328. [PMID: 26230952 PMCID: PMC4526782 DOI: 10.1038/bcj.2015.56] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 06/04/2015] [Accepted: 06/12/2015] [Indexed: 12/19/2022] Open
Abstract
The biological role of monocytes and macrophages in B-cell non-Hodgkin lymphoma (NHL) is not fully understood. We have previously reported that monocytes from patients with B-cell NHL have an immunosuppressive CD14+HLA-DRlow/− phenotype that correlates with a poor prognosis. However, the underlying mechanism by which CD14+HLA-DRlow/− monocytes develop in lymphoma is unknown. In the present study, we found that interleukin (IL)-10, which is increased in the serum of patients with B-cell NHL, induced the development of the CD4+HLA-DRlow/− population. Using peripheral blood samples from patients with B-cell NHL, we found that absolute numbers of CD14+ monocytic cells with an HLA-DRlow/− phenotype were higher than healthy controls and correlated with a higher International Prognostic Index score. IL-10 serum levels were elevated in lymphoma patients compared with controls and were associated with increased peripheral monocyte counts. Treatment of monocytes with IL-10 in vitro significantly decreased HLA-DR expression and resulted in the expansion of CD14+HLA-DRlow/− population. We found that lymphoma B cells produce IL-10 and supernatants from cultured lymphoma cells increased the CD14+HLA-DRlow/− population. Furthermore, we found that IL-10-induced CD14+HLA-DRlow/− monocytes inhibited the activation and proliferation of T cells. Taken together, these results suggest that elevated IL-10 serum levels contribute to increased numbers of immunosuppressive CD14+HLA-DRlow/− monocytes in B-cell NHL.
Collapse
|
100
|
Beyond the Immune Suppression: The Immunotherapy in Prostate Cancer. BIOMED RESEARCH INTERNATIONAL 2015; 2015:794968. [PMID: 26161414 PMCID: PMC4486485 DOI: 10.1155/2015/794968] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 02/06/2015] [Accepted: 02/06/2015] [Indexed: 11/18/2022]
Abstract
Prostate cancer (PCa) is the second most common cancer in men. As well in many other human cancers, inflammation and immune suppression have an important role in their development. We briefly describe the host components that interact with the tumor to generate an immune suppressive environment involved in PCa promotion and progression. Different tools provide to overcome the mechanisms of immunosuppression including vaccines and immune checkpoint blockades. With regard to this, we report results of most recent clinical trials investigating immunotherapy in metastatic PCa (Sipuleucel-T, ipilimumab, tasquinimod, Prostvac-VF, and GVAX) and provide possible future perspectives combining the immunotherapy to the traditional therapies.
Collapse
|