101
|
A core of kinase-regulated interactomes defines the neoplastic MDSC lineage. Oncotarget 2016; 6:27160-75. [PMID: 26320174 PMCID: PMC4694980 DOI: 10.18632/oncotarget.4746] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 07/13/2015] [Indexed: 12/17/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) differentiate from bone marrow precursors, expand in cancer-bearing hosts and accelerate tumor progression. MDSCs have become attractive therapeutic targets, as their elimination strongly enhances anti-neoplastic treatments. Here, immature myeloid dendritic cells (DCs), MDSCs modeling tumor-infiltrating subsets or modeling non-cancerous (NC)-MDSCs were compared by in-depth quantitative proteomics. We found that neoplastic MDSCs differentially expressed a core of kinases which controlled lineage-specific (PI3K-AKT and SRC kinases) and cancer-induced (ERK and PKC kinases) protein interaction networks (interactomes). These kinases contributed to some extent to myeloid differentiation. However, only AKT and ERK specifically drove MDSC differentiation from myeloid precursors. Interfering with AKT and ERK with selective small molecule inhibitors or shRNAs selectively hampered MDSC differentiation and viability. Thus, we provide compelling evidence that MDSCs constitute a distinct myeloid lineage distinguished by a “kinase signature” and well-defined interactomes. Our results define new opportunities for the development of anti-cancer treatments targeting these tumor-promoting immune cells.
Collapse
|
102
|
Abstract
Bone metastatic disease remains a significant and frequent problem for cancer patients that can lead to increased morbidity and mortality. Unfortunately, despite decades of research, bone metastases remain incurable. Current studies have demonstrated that many properties and cell types within the bone and bone marrow microenvironment contribute to tumor-induced bone disease. Furthermore, they have pointed to the importance of understanding how tumor cells interact with their microenvironment in order to help improve both the development of new therapeutics and the prediction of response to therapy.
Collapse
Affiliation(s)
- Denise Buenrostro
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37212, USA
- Center for Bone Biology, Vanderbilt University, 2215B Garland Avenue, 1235 MRBIV, Nashville, TN 37232, USA
- Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Patrick L. Mulcrone
- Center for Bone Biology, Vanderbilt University, 2215B Garland Avenue, 1235 MRBIV, Nashville, TN 37232, USA
- Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Philip Owens
- Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Julie A. Sterling
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37212, USA
- Center for Bone Biology, Vanderbilt University, 2215B Garland Avenue, 1235 MRBIV, Nashville, TN 37232, USA
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University, 2215B Garland Avenue, 1235 MRBIV, Nashville, TN 37232, USA
- Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
103
|
Novack DV, Mbalaviele G. Osteoclasts-Key Players in Skeletal Health and Disease. Microbiol Spectr 2016; 4:10.1128/microbiolspec.MCHD-0011-2015. [PMID: 27337470 PMCID: PMC4920143 DOI: 10.1128/microbiolspec.mchd-0011-2015] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Indexed: 12/12/2022] Open
Abstract
The differentiation of osteoclasts (OCs) from early myeloid progenitors is a tightly regulated process that is modulated by a variety of mediators present in the bone microenvironment. Once generated, the function of mature OCs depends on cytoskeletal features controlled by an αvβ3-containing complex at the bone-apposed membrane and the secretion of protons and acid-protease cathepsin K. OCs also have important interactions with other cells in the bone microenvironment, including osteoblasts and immune cells. Dysregulation of OC differentiation and/or function can cause bone pathology. In fact, many components of OC differentiation and activation have been targeted therapeutically with great success. However, questions remain about the identity and plasticity of OC precursors and the interplay between essential networks that control OC fate. In this review, we summarize the key principles of OC biology and highlight recently uncovered mechanisms regulating OC development and function in homeostatic and disease states.
Collapse
Affiliation(s)
- Deborah Veis Novack
- Musculoskeletal Research Center, Division of Bone and Mineral Diseases, Department of Medicine
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Gabriel Mbalaviele
- Musculoskeletal Research Center, Division of Bone and Mineral Diseases, Department of Medicine
| |
Collapse
|
104
|
Malek E, de Lima M, Letterio JJ, Kim BG, Finke JH, Driscoll JJ, Giralt SA. Myeloid-derived suppressor cells: The green light for myeloma immune escape. Blood Rev 2016; 30:341-8. [PMID: 27132116 DOI: 10.1016/j.blre.2016.04.002] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 03/28/2016] [Accepted: 04/01/2016] [Indexed: 01/04/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous, immature myeloid cell population with the ability to suppress innate and adaptive immune responses that promote tumor growth. MDSCs are increased in patients with multiple myeloma (MM) and have bidirectional interaction with tumors within the MM microenvironment. MM-MDSCs promote MM tumor growth and induce immune suppression; conversely, MM cells induce MDSC development and survival. Although the role of MDSCs in infections, inflammatory diseases and solid tumors has been extensively characterized, their tumor-promoting and immune-suppressive role in MM and the MM microenvironment is only beginning to emerge. The presence and activation of MDSCs in MM patients has been well documented; however, the direct actions and functional consequences of MDSCs on cancer cells is poorly defined. Immunosuppressive MDSCs play an important role in tumor progression primarily because of their capability to promote immune-escape, angiogenesis, drug resistance and metastasis. However, their role in the bone marrow (BM), the primary MM site, is poorly understood. MM remains an incurable malignancy, and it is likely that the BM microenvironment protects MM against chemotherapy agents and the host immune system. A growing body of evidence suggests that host immune cells with a suppressive phenotype contribute to a myeloma immunosuppressive network. Among the known suppressor cells, MDSCs and T regulatory cells (Tregs) have been found to be significantly increased in myeloma patients and their levels correlate with disease stage and clinical outcome. Furthermore, it has been shown that MDSC can mediate suppression of myeloma-specific T-cell responses through the induction of T-cell anergy and Treg development in the MM microenvironment. Here, we review clinical correlations and the preclinical proof-of-principle data on the role of MDSCs in myeloma immunotolerance and highlight the mechanistically relevant MDSC-targeted compounds and their potential utility in a new approach for anti-myeloma therapy.
Collapse
Affiliation(s)
- Ehsan Malek
- University Hospitals Case Medical Center, Seidman Cancer Center, Cleveland, OH, USA.
| | - Marcos de Lima
- University Hospitals Case Medical Center, Seidman Cancer Center, Cleveland, OH, USA
| | - John J Letterio
- Department of Pediatrics, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA; The Angie Fowler Adolescent & Young Adult Cancer Institute, Rainbow Babies & Children's Hospital, University Hospitals, Cleveland, OH, USA
| | - Byung-Gyu Kim
- Department of Pediatrics, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA; The Angie Fowler Adolescent & Young Adult Cancer Institute, Rainbow Babies & Children's Hospital, University Hospitals, Cleveland, OH, USA
| | - James H Finke
- Taussig Cancer Institute, Glickman Urological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - James J Driscoll
- Division of Hematology and Oncology, University of Cincinnati College of Medicine, Cincinnati, OH, USA; The Vontz Center for Molecular Studies, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Sergio A Giralt
- Adult Bone Marrow Transplant Service, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
105
|
Stiff A, Trikha P, Wesolowski R, Kendra K, Hsu V, Uppati S, McMichael E, Duggan M, Campbell A, Keller K, Landi I, Zhong Y, Dubovsky J, Howard JH, Yu L, Harrington B, Old M, Reiff S, Mace T, Tridandapani S, Muthusamy N, Caligiuri MA, Byrd JC, Carson WE. Myeloid-Derived Suppressor Cells Express Bruton's Tyrosine Kinase and Can Be Depleted in Tumor-Bearing Hosts by Ibrutinib Treatment. Cancer Res 2016; 76:2125-36. [PMID: 26880800 DOI: 10.1158/0008-5472.can-15-1490] [Citation(s) in RCA: 153] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 12/23/2015] [Indexed: 02/07/2023]
Abstract
Myeloid-derived suppressor cells (MDSC) are a heterogeneous group of immature myeloid cells that expand in tumor-bearing hosts in response to soluble factors produced by tumor and stromal cells. MDSC expansion has been linked to loss of immune effector cell function and reduced efficacy of immune-based cancer therapies, highlighting the MDSC population as an attractive therapeutic target. Ibrutinib, an irreversible inhibitor of Bruton's tyrosine kinase (BTK) and IL2-inducible T-cell kinase (ITK), is in clinical use for the treatment of B-cell malignancies. Here, we report that BTK is expressed by murine and human MDSCs, and that ibrutinib is able to inhibit BTK phosphorylation in these cells. Treatment of MDSCs with ibrutinib significantly impaired nitric oxide production and cell migration. In addition, ibrutinib inhibited in vitro generation of human MDSCs and reduced mRNA expression of indolamine 2,3-dioxygenase, an immunosuppressive factor. Treatment of mice bearing EMT6 mammary tumors with ibrutinib resulted in reduced frequency of MDSCs in both the spleen and tumor. Ibrutinib treatment also resulted in a significant reduction of MDSCs in wild-type mice bearing B16F10 melanoma tumors, but not in X-linked immunodeficiency mice (XID) harboring a BTK mutation, suggesting that BTK inhibition plays an important role in the observed reduction of MDSCs in vivo Finally, ibrutinib significantly enhanced the efficacy of anti-PD-L1 (CD274) therapy in a murine breast cancer model. Together, these results demonstrate that ibrutinib modulates MDSC function and generation, revealing a potential strategy for enhancing immune-based therapies in solid malignancies. Cancer Res; 76(8); 2125-36. ©2016 AACR.
Collapse
Affiliation(s)
- Andrew Stiff
- Medical Scientist Training Program, Columbus, Ohio. Biomedical Sciences Graduate Program, The Ohio State University, Columbus, Ohio
| | - Prashant Trikha
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Robert Wesolowski
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Kari Kendra
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Vincent Hsu
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Sarvani Uppati
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Elizabeth McMichael
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, Ohio
| | - Megan Duggan
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, Ohio
| | - Amanda Campbell
- Medical Scientist Training Program, Columbus, Ohio. Biomedical Sciences Graduate Program, The Ohio State University, Columbus, Ohio
| | - Karen Keller
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Ian Landi
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Yiming Zhong
- Division of Hematology, Department of Internal Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Jason Dubovsky
- Division of Hematology, Department of Internal Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - John Harrison Howard
- Department of Internal Medicine and Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Lianbo Yu
- Department of Biomedical Informatics, The Ohio State University, Columbus, Ohio
| | - Bonnie Harrington
- Division of Hematology, Department of Internal Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Matthew Old
- Department of Otolaryngology, The Ohio State University, Columbus, Ohio
| | - Sean Reiff
- Medical Scientist Training Program, Columbus, Ohio. Biomedical Sciences Graduate Program, The Ohio State University, Columbus, Ohio
| | - Thomas Mace
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Susheela Tridandapani
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, Ohio. Department of Internal Medicine and Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Natarajan Muthusamy
- Division of Hematology, Department of Internal Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Michael A Caligiuri
- Division of Hematology, Department of Internal Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - John C Byrd
- Division of Hematology, Department of Internal Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - William E Carson
- Division of Surgical Oncology, Department of Surgery and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.
| |
Collapse
|
106
|
Marvel D, Gabrilovich DI. Myeloid-derived suppressor cells in the tumor microenvironment: expect the unexpected. J Clin Invest 2015; 125:3356-64. [PMID: 26168215 DOI: 10.1172/jci80005] [Citation(s) in RCA: 813] [Impact Index Per Article: 81.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Our understanding of the role of myeloid-derived suppressor cells (MDSCs) in cancer is becoming increasingly complex. In addition to their eponymous role in suppressing immune responses, they directly support tumor growth, differentiation, and metastasis in a number of ways that are only now beginning to be appreciated. It is because of this increasingly complex role that these cells may become an important factor in the treatment of human cancer. In this Review, we discuss the most pertinent and controversial issues of MDSC biology and their role in promoting cancer progression and highlight how these cells may be used in the clinic, both as prognostic factors and as therapeutic targets.
Collapse
|
107
|
Shiozawa Y, Eber MR, Berry JE, Taichman RS. Bone marrow as a metastatic niche for disseminated tumor cells from solid tumors. BONEKEY REPORTS 2015; 4:689. [PMID: 26029360 PMCID: PMC4440229 DOI: 10.1038/bonekey.2015.57] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 03/20/2015] [Indexed: 02/07/2023]
Abstract
Bone marrow is a heterogeneous organ containing diverse cell types, and it is a preferred metastatic site for several solid tumors such as breast and prostate cancer. Recently, it has been shown that bone metastatic cancer cells interact with the bone marrow microenvironment to survive and grow, and thus this microenvironment is referred to as the 'metastatic niche'. Once cancer cells spread to distant organs such as bone, the prognosis for the patient is generally poor. There is an urgent need to establish a greater understanding of the mechanisms whereby the bone marrow niche influences bone metastasis. Here we discuss insights into the contribution of the bone marrow 'metastatic niche' to progression of bone metastatic disease, with a particular focus on cells of hematopoietic and mesenchymal origin.
Collapse
Affiliation(s)
- Yusuke Shiozawa
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - Matthew R Eber
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - Janice E Berry
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - Russell S Taichman
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| |
Collapse
|
108
|
The Impact of Immune System in Regulating Bone Metastasis Formation by Osteotropic Tumors. J Immunol Res 2015; 2015:143526. [PMID: 26064994 PMCID: PMC4433688 DOI: 10.1155/2015/143526] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 12/02/2014] [Indexed: 01/31/2023] Open
Abstract
Bone metastases are frequent and debilitating consequence for many tumors, such as breast, lung, prostate, and kidney cancer. Many studies report the importance of the immune system in the pathogenesis of bone metastasis. Indeed, bone and immune system are strictly linked to each other because bone regulates the hematopoietic stem cells from which all cells of the immune system derive, and many immunoregulatory cytokines influence the fate of bone cells. Furthermore, both cytokines and factors produced by immune and bone cells promote the growth of tumor cells in bone, contributing to supporting the vicious cycle of bone metastasis. This review summarizes the current knowledge on the interactions among bone, immune, and tumor cells aiming to provide an overview of the osteoimmunology field in bone metastasis from solid tumors.
Collapse
|
109
|
Mori G, D'Amelio P, Faccio R, Brunetti G. Bone-immune cell crosstalk: bone diseases. J Immunol Res 2015; 2015:108451. [PMID: 26000310 PMCID: PMC4427089 DOI: 10.1155/2015/108451] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 01/22/2015] [Accepted: 01/25/2015] [Indexed: 01/14/2023] Open
Abstract
Bone diseases are associated with great morbidity; thus, the understanding of the mechanisms leading to their development represents a great challenge to improve bone health. Recent reports suggest that a large number of molecules produced by immune cells affect bone cell activity. However, the mechanisms are incompletely understood. This review aims to shed new lights into the mechanisms of bone diseases involving immune cells. In particular, we focused our attention on the major pathogenic mechanism underlying periodontal disease, psoriatic arthritis, postmenopausal osteoporosis, glucocorticoid-induced osteoporosis, metastatic solid tumors, and multiple myeloma.
Collapse
Affiliation(s)
- Giorgio Mori
- Department of Clinical and Experimental Medicine, University of Foggia, 71100 Foggia, Italy
| | - Patrizia D'Amelio
- Department of Medical Science, Section of Gerontology and Bone Metabolism Diseases, University of Torino, 10126 Torino, Italy
| | - Roberta Faccio
- Department of Orthopedics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Giacomina Brunetti
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, Section of Human Anatomy and Histology, University of Bari, 70124 Bari, Italy
| |
Collapse
|
110
|
Chen THP, Swarnkar G, Mbalaviele G, Abu-Amer Y. Myeloid lineage skewing due to exacerbated NF-κB signaling facilitates osteopenia in Scurfy mice. Cell Death Dis 2015; 6:e1723. [PMID: 25880090 PMCID: PMC4650554 DOI: 10.1038/cddis.2015.87] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 01/29/2015] [Accepted: 02/16/2015] [Indexed: 01/03/2023]
Abstract
Immune surveillance through Foxp3+ regulatory T cells plays a crucial role in bone homeostasis. Scurfy, the mouse model of autoimmune IPEX syndrome, bears a loss-of-function mutation in Foxp3 that leads to multi-organ inflammation. Herein, we report that scurfy mice exhibit severe bone loss mediated by accelerated osteoclastogenesis. Mechanistically, Foxp3 deficiency results in the upregulation of NF-κB in T helper cells through the loss of repressive Foxp3/NEMO interaction, thereby unleashing NF-κB-mediated over-production of pro-osteoclastogenic cytokines. Flow cytometry analysis shows marked increase in lin-Sca-1+c-kit+ hematopoietic stem cells (LSK HSCs) and granulocyte/macrophage progenitors (GMPs) in bone marrow of scurfy mice with corresponding exacerbated osteoclastogenic potential, implying that osteoclast progenitors are affected at a very primitive stage in this disorder. Scurfy LSK HSCs exhibit greater sensitivity to M-CSF and contain abundant PU.1+ Sf LSK HSCs compared with WT. Accordingly, genetic or pharmacological inhibition of M-CSF or mTOR signaling, but not IL-17 signaling, attenuates osteoclastogenesis and osteopenia in scurfy. Thus, our study suggests that Foxp3 deficiency leads to osteopenia owing to dysregulated NF-κB activity and subsequent cytokine-mediated hyper-proliferation of myeloid precursors, and positions the NF-κB pathway as a potential target for therapeutic intervention for this disorder.
Collapse
Affiliation(s)
- T H-P Chen
- Department of Orthopaedic Surgery and Cell Biology & Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - G Swarnkar
- Department of Orthopaedic Surgery and Cell Biology & Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - G Mbalaviele
- Department of Internal Medicine, Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Y Abu-Amer
- Department of Orthopaedic Surgery and Cell Biology & Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
111
|
Bone specific immunity and its impact on metastasis. BONEKEY REPORTS 2015; 4:665. [PMID: 25908968 DOI: 10.1038/bonekey.2015.32] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Accepted: 02/13/2015] [Indexed: 02/07/2023]
Abstract
Bone is one of the most common sites of metastasis in solid malignancy. Contributing to this osteotropism are the dynamic interactions between tumor cells and the numerous cell types resident in the normal bone, particularly osteoclasts and osteoblasts, which create a tumor supporting microenvironment. However, disseminated cells are detected in the bone marrow long before evidence of metastatic outgrowth, and it is likely that prolonged survival is also reliant on immunoescape. Compared with other peripheral organs such as the lung and spleen, the bone marrow constitutes a unique immune cell compartment that likely provides an immune privileged niche for disseminated tumor cells. This includes the large proportions of immunosuppressive cells, including myeloid derived suppressor cells and regulatory T cells, that blunt the activity of cytotoxic lymphocytes involved in tumor immunosurveillance. This review highlights key aspects of the osteoimmune landscape and emerging mechanisms by which tumor cells create or co-opt an immunosuppressed niche to support their outgrowth in bone. Future studies in this field are likely to shed light on the differences in immunoregulation between the bone and other sites including the primary tumor, and the potential for immunotherapeutics in treating disseminated disease in the bone. However, more immunocompetent models, that recapitulate tumor heterogeneity and bone metastasis need to be developed to accelerate this field.
Collapse
|
112
|
Cook LM, Shay G, Araujo A, Aruajo A, Lynch CC. Integrating new discoveries into the "vicious cycle" paradigm of prostate to bone metastases. Cancer Metastasis Rev 2015; 33:511-25. [PMID: 24414228 DOI: 10.1007/s10555-014-9494-4] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In prostate to bone metastases, the "vicious cycle" paradigm has been traditionally used to illustrate how metastases manipulate the bone forming osteoblasts and resorbing osteoclasts in order to yield factors that facilitate growth and establishment. However, recent advances have illustrated that the cycle is far more complex than this simple interpretation. In this review, we will discuss the role of exosomes and hematopoietic/mesenchymal stem/stromal cells (MSC) that facilitate the establishment and activation of prostate metastases and how cells including myeloid-derived suppressor cells, macrophages, T cells, and nerve cells contribute to the momentum of the vicious cycle. The increased complexity of the tumor-bone microenvironment requires a system level approach. The evolution of computational models to interrogate the tumor-bone microenvironment is also discussed, and the application of this integrated approach should allow for the development of effective therapies to treat and cure prostate to bone metastases.
Collapse
Affiliation(s)
- Leah M Cook
- Department of Tumor Biology, Moffitt Cancer Center and Research Institute, 12902 Magnolia Dr., SRB-3, Tampa, FL, 33612, USA
| | | | | | | | | |
Collapse
|
113
|
Tabariès S, Ouellet V, Hsu BE, Annis MG, Rose AAN, Meunier L, Carmona E, Tam CE, Mes-Masson AM, Siegel PM. Granulocytic immune infiltrates are essential for the efficient formation of breast cancer liver metastases. Breast Cancer Res 2015; 17:45. [PMID: 25882816 PMCID: PMC4413545 DOI: 10.1186/s13058-015-0558-3] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2014] [Accepted: 03/10/2015] [Indexed: 12/18/2022] Open
Abstract
Introduction Breast cancer cells display preferences for specific metastatic sites including the bone, lung and liver. Metastasis is a complex process that relies, in part, on interactions between disseminated cancer cells and resident/infiltrating stromal cells that constitute the metastatic microenvironment. Distinct immune infiltrates can either impair the metastatic process or conversely, assist in the seeding, colonization and growth of disseminated cancer cells. Methods Using in vivo selection approaches, we previously isolated 4T1-derived breast cancer cells that preferentially metastasize to these organs and tissues. In this study, we examined whether the propensity of breast cancer cells to metastasize to the lung, liver or bone is associated with and dependent on distinct patterns of immune cell infiltration. Immunohistocytochemistry and immunohistofluorescence approaches were used to quantify innate immune cell infiltrates within distinct metastases and depletion of Gr1+ (Ly-6C and Ly-6G) or specifically Ly-6G+ cells was performed to functionally interrogate the role of Ly-6G+ infiltrates in promoting metastasis to these organs. Results We show that T lymphocytes (CD3+), myeloid-derived (Gr-1+) cells and neutrophils (Ly-6G+ or NE+) exhibit the most pronounced recruitment in lung and liver metastases, with markedly less recruitment within bone metastatic lesions. Interestingly, these infiltrating cell populations display different patterns of localization within soft tissue metastases. T lymphocytes and granulocytic immune infiltrates are localized around the periphery of liver metastases whereas they were dispersed throughout the lung metastases. Furthermore, Gr-1+ cell-depletion studies demonstrate that infiltrating myeloid-derived cells are essential for the formation of breast cancer liver metastases but dispensable for metastasis to the lung and bone. A specific role for the granulocytic component of the innate immune infiltrate was revealed through Ly-6G+ cell-depletion experiments, which resulted in significantly impaired formation of liver metastases. Finally, we demonstrate that the CD11b+/Ly-6G+ neutrophils that infiltrate and surround the liver metastases are polarized toward an N2 phenotype, which have previously been shown to enhance tumor growth and metastasis. Conclusions Our results demonstrate that the liver-metastatic potential of breast cancer cells is heavily reliant on interactions with infiltrating Ly-6G+ cells within the liver microenvironment. Electronic supplementary material The online version of this article (doi:10.1186/s13058-015-0558-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sébastien Tabariès
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue West, Room 513, Montréal, QC, H3A 1A3, Canada. .,Department of Medicine, McGill University, 3605 Rue de la Montagne, Montréal, QC, H3G 2M1, Canada.
| | - Véronique Ouellet
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CR-CHUM)/Institut du cancer de Montréal, 900 Saint Denis, Montréal, QC, H2X 0A9, Canada.
| | - Brian E Hsu
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue West, Room 513, Montréal, QC, H3A 1A3, Canada. .,Department of Medicine, McGill University, 3605 Rue de la Montagne, Montréal, QC, H3G 2M1, Canada.
| | - Matthew G Annis
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue West, Room 513, Montréal, QC, H3A 1A3, Canada. .,Department of Medicine, McGill University, 3605 Rue de la Montagne, Montréal, QC, H3G 2M1, Canada.
| | - April A N Rose
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue West, Room 513, Montréal, QC, H3A 1A3, Canada. .,Department of Medicine, McGill University, 3605 Rue de la Montagne, Montréal, QC, H3G 2M1, Canada.
| | - Liliane Meunier
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CR-CHUM)/Institut du cancer de Montréal, 900 Saint Denis, Montréal, QC, H2X 0A9, Canada.
| | - Euridice Carmona
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CR-CHUM)/Institut du cancer de Montréal, 900 Saint Denis, Montréal, QC, H2X 0A9, Canada.
| | - Christine E Tam
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue West, Room 513, Montréal, QC, H3A 1A3, Canada. .,Department of Medicine, McGill University, 3605 Rue de la Montagne, Montréal, QC, H3G 2M1, Canada.
| | - Anne-Marie Mes-Masson
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CR-CHUM)/Institut du cancer de Montréal, 900 Saint Denis, Montréal, QC, H2X 0A9, Canada. .,Department of Medecine, Université de Montréal, 2900 Boulevard Edouard-Montpetit, Montréal, QC, H3T 1J4, Canada.
| | - Peter M Siegel
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue West, Room 513, Montréal, QC, H3A 1A3, Canada. .,Department of Medicine, McGill University, 3605 Rue de la Montagne, Montréal, QC, H3G 2M1, Canada. .,Department of Biochemistry, McGill University, 3655 Promenade Sir William Osler, Montréal, QC, H3G 1Y6, Canada.
| |
Collapse
|
114
|
Younos IH, Abe F, Talmadge JE. Myeloid-derived suppressor cells: their role in the pathophysiology of hematologic malignancies and potential as therapeutic targets. Leuk Lymphoma 2015; 56:2251-63. [PMID: 25407654 DOI: 10.3109/10428194.2014.987141] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature myeloid cells at various stages of differentiation/maturation that have a role in cancer induction and progression. They function as vasculogenic and immunosuppressive cells, utilizing multiple mechanisms to block both innate and adaptive anti-tumor immunity. Recently, their mechanism of action and clinical importance have been defined, and the cross-talk between myeloid cells and cancer cells has been shown to contribute to tumor induction, progression, metastasis and tolerance. In this review, we focus on the role of MDSCs in hematologic malignancies and the therapeutic approaches targeting MDSCs that are currently in clinical studies.
Collapse
Affiliation(s)
- Ibrahim H Younos
- a Department of Clinical Pharmacology , Menoufia University , Al-Minufya , Egypt.,b Department of Pharmacology and Clinical Pharmacy , College of Medicine and Health Sciences, Sultan Qaboos University , Muscat , Oman
| | - Fuminori Abe
- c SBI Pharmaceuticals Co., Ltd. , Tokyo 106-6020 , Japan
| | - James E Talmadge
- d Department of Pathology and Microbiology , Nebraska Medical Center , Omaha , NE , USA
| |
Collapse
|
115
|
De Veirman K, Van Valckenborgh E, Lahmar Q, Geeraerts X, De Bruyne E, Menu E, Van Riet I, Vanderkerken K, Van Ginderachter JA. Myeloid-derived suppressor cells as therapeutic target in hematological malignancies. Front Oncol 2014; 4:349. [PMID: 25538893 PMCID: PMC4258607 DOI: 10.3389/fonc.2014.00349] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 11/23/2014] [Indexed: 12/29/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSC) are a heterogeneous population of immature myeloid cells that accumulate during pathological conditions such as cancer and are associated with a poor clinical outcome. MDSC expansion hampers the host anti-tumor immune response by inhibition of T cell proliferation, cytokine secretion, and recruitment of regulatory T cells. In addition, MDSC exert non-immunological functions including the promotion of angiogenesis, tumor invasion, and metastasis. Recent years, MDSC are considered as a potential target in solid tumors and hematological malignancies to enhance the effects of currently used immune modulating agents. This review focuses on the characteristics, distribution, functions, cell–cell interactions, and targeting of MDSC in hematological malignancies including multiple myeloma, lymphoma, and leukemia.
Collapse
Affiliation(s)
- Kim De Veirman
- Laboratory of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel , Brussels , Belgium
| | - Els Van Valckenborgh
- Laboratory of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel , Brussels , Belgium
| | - Qods Lahmar
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel , Brussels , Belgium ; Laboratory of Myeloid Cell Immunology, VIB , Brussels , Belgium
| | - Xenia Geeraerts
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel , Brussels , Belgium ; Laboratory of Myeloid Cell Immunology, VIB , Brussels , Belgium
| | - Elke De Bruyne
- Laboratory of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel , Brussels , Belgium
| | - Eline Menu
- Laboratory of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel , Brussels , Belgium
| | - Ivan Van Riet
- Laboratory of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel , Brussels , Belgium
| | - Karin Vanderkerken
- Laboratory of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel , Brussels , Belgium
| | - Jo A Van Ginderachter
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel , Brussels , Belgium ; Laboratory of Myeloid Cell Immunology, VIB , Brussels , Belgium
| |
Collapse
|
116
|
Botta C, Gullà A, Correale P, Tagliaferri P, Tassone P. Myeloid-derived suppressor cells in multiple myeloma: pre-clinical research and translational opportunities. Front Oncol 2014; 4:348. [PMID: 25538892 PMCID: PMC4258997 DOI: 10.3389/fonc.2014.00348] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 11/23/2014] [Indexed: 12/31/2022] Open
Abstract
Immunosuppressive cells have been reported to play an important role in tumor-progression mainly because of their capability to promote immune-escape, angiogenesis, and metastasis. Among them, myeloid-derived suppressor cells (MDSCs) have been recently identified as immature myeloid cells, induced by tumor-associated inflammation, able to impair both innate and adaptive immunity. While murine MDSCs are usually identified by the expression of CD11b and Gr1, human MDSCs represent a more heterogeneous population characterized by the expression of CD33 and CD11b, low or no HLA-DR, and variable CD14 and CD15. In particular, the last two may alternatively identify monocyte-like or granulocyte-like MDSC subsets with different immunosuppressive properties. Recently, a substantial increase of MDSCs has been found in peripheral blood and bone marrow (BM) of multiple myeloma (MM) patients with a role in disease progression and/or drug resistance. Pre-clinical models recapitulating the complexity of the MM-related BM microenvironment (BMM) are major tools for the study of the interactions between MM cells and cells of the BMM (including MDSCs) and for the development of new agents targeting MM-associated immune-suppressive cells. This review will focus on current strategies for human MDSCs generation and investigation of their immunosuppressive function in vitro and in vivo, taking into account the relevant relationship occurring within the MM–BMM. We will then provide trends in MDSC-associated research and suggest potential application for the treatment of MM.
Collapse
Affiliation(s)
- Cirino Botta
- Department of Experimental and Clinical Medicine, "Magna Graecia" University and Medical Oncology Unit, T. Campanella Cancer Center, "Salvatore Venuta" University Campus , Catanzaro , Italy
| | - Annamaria Gullà
- Department of Experimental and Clinical Medicine, "Magna Graecia" University and Medical Oncology Unit, T. Campanella Cancer Center, "Salvatore Venuta" University Campus , Catanzaro , Italy
| | | | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine, "Magna Graecia" University and Medical Oncology Unit, T. Campanella Cancer Center, "Salvatore Venuta" University Campus , Catanzaro , Italy
| | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, "Magna Graecia" University and Medical Oncology Unit, T. Campanella Cancer Center, "Salvatore Venuta" University Campus , Catanzaro , Italy ; Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University , Philadelphia, PA , USA
| |
Collapse
|
117
|
Capietto AH, Faccio R. Immune regulation of bone metastasis. BONEKEY REPORTS 2014; 3:600. [PMID: 25512853 DOI: 10.1038/bonekey.2014.95] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 09/18/2014] [Indexed: 12/17/2022]
Abstract
Metastases to bone occur in about 70% of patients with metastatic prostate and breast cancers. Unfortunately, bone metastases result in significant morbidity and mortality and treatment options are limited. Thus, significant effort has focused on understanding the mechanisms that drive tumor dissemination to bone. Bone metastases are typically characterized by a self-perpetuating 'vicious' cycle wherein tumor cells and bone-resorbing cells (osteoclasts) are locked in a cycle that leads to osteoclast-driven bone destruction and the release of bone-stored factors that in turn stimulate tumor cell proliferation and survival. To break this 'vicious' cycle, potent antiresorptive agents such as zoledronic acid (ZOL) have been used. However, in the clinical setting, ZOL failed to improve the overall survival of cancer patients even though it inhibited osteoclast resorptive activity. Thus, other cells in addition to osteoclasts are likely involved in modulating tumor growth in the bone. The immune system has the ability to eliminate tumor cells. Nevertheless, tumor cells can acquire the ability to escape immune control. Our recent observations indicated that a decline in the ability of the immune cells to recognize and kill the tumor drives tumor dissemination to bone even when osteoclasts are inhibited by potent antiresorptive agents. This review focuses on the antitumor and protumor effects of various immune cell populations involved in the bone metastatic process. We also discuss strategies to enhance antitumor immune responses and bypass cancer immune resistance.
Collapse
Affiliation(s)
- Aude-Hélène Capietto
- Department of Orthopedics, Washington University School of Medicine , St Louis, MO, USA
| | - Roberta Faccio
- Department of Orthopedics, Washington University School of Medicine , St Louis, MO, USA
| |
Collapse
|
118
|
Li H, Lu Y, Qian J, Zheng Y, Zhang M, Bi E, He J, Liu Z, Xu J, Gao JY, Yi Q. Human osteoclasts are inducible immunosuppressive cells in response to T cell-derived IFN-γ and CD40 ligand in vitro. J Bone Miner Res 2014; 29:2666-75. [PMID: 24916315 PMCID: PMC4262718 DOI: 10.1002/jbmr.2294] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 05/28/2014] [Accepted: 05/30/2014] [Indexed: 01/03/2023]
Abstract
Osteoclasts (OCs) are bone resorbing cells whose activity can be regulated by activated T cells and their cytokines. However, the immune function of OCs is largely unknown. In this study, we found that as bystanders, human OCs effectively suppressed T-cell proliferation induced by allogeneic, microbial antigenic, and T-cell receptor stimuli in vitro. Mechanism studies revealed that T cell-derived IFN-γ and CD40 ligand (CD40L) induced the expression of indoleamine 2,3-dioxygenase (IDO) in OCs, which mediated the immunosuppressive function on T-cell proliferation through depleting tryptophan. Neutralizing IFN-γ and blocking CD40L, or silencing or inhibiting IDO in OCs restored T-cell proliferation in the presence of OCs. Our data reveal a novel function of human OCs as inducible immunosuppressive cells, and a feedback loop between OCs and activated T cells. Thus, this study provides new insight into the mechanism of the immunosuppressive function of OCs, and may be helpful for developing novel therapeutic strategies for human diseases involving both the bone and immune systems.
Collapse
Affiliation(s)
- Haiyan Li
- Department of Cancer Biology, Lerner Research Institue, Cleveland Clinic, Cleveland, OH, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
119
|
Shen L, Sundstedt A, Ciesielski M, Miles KM, Celander M, Adelaiye R, Orillion A, Ciamporcero E, Ramakrishnan S, Ellis L, Fenstermaker R, Abrams SI, Eriksson H, Leanderson T, Olsson A, Pili R. Tasquinimod modulates suppressive myeloid cells and enhances cancer immunotherapies in murine models. Cancer Immunol Res 2014; 3:136-48. [PMID: 25370534 DOI: 10.1158/2326-6066.cir-14-0036] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A major barrier for cancer immunotherapy is the presence of suppressive cell populations in patients with cancer, such as myeloid-derived suppressor cells (MDSC) and tumor-associated macrophages (TAM), which contribute to the immunosuppressive microenvironment that promotes tumor growth and metastasis. Tasquinimod is a novel antitumor agent that is currently at an advanced stage of clinical development for treatment of castration-resistant prostate cancer. A target of tasquinimod is the inflammatory protein S100A9, which has been demonstrated to affect the accumulation and function of tumor-suppressive myeloid cells. Here, we report that tasquinimod provided a significant enhancement to the antitumor effects of two different immunotherapeutics in mouse models of cancer: a tumor vaccine (SurVaxM) for prostate cancer and a tumor-targeted superantigen (TTS) for melanoma. In the combination strategies, tasquinimod inhibited distinct MDSC populations and TAMs of the M2-polarized phenotype (CD206(+)). CD11b(+) myeloid cells isolated from tumors of treated mice expressed lower levels of arginase-1 and higher levels of inducible nitric oxide synthase (iNOS), and were less immunosuppressive ex vivo, which translated into a significantly reduced tumor-promoting capacity in vivo when these cells were coinjected with tumor cells. Tumor-specific CD8(+) T cells were increased markedly in the circulation and in tumors. Furthermore, T-cell effector functions, including cell-mediated cytotoxicity and IFNγ production, were potentiated. Taken together, these data suggest that pharmacologic targeting of suppressive myeloid cells by tasquinimod induces therapeutic benefit and provide the rationale for clinical testing of tasquinimod in combination with cancer immunotherapies.
Collapse
Affiliation(s)
- Li Shen
- Genitourinary Program, Roswell Park Cancer Institute, Buffalo, New York
| | | | - Michael Ciesielski
- Department of Neurosurgery, Roswell Park Cancer Institute, Buffalo, New York
| | | | | | - Remi Adelaiye
- Genitourinary Program, Roswell Park Cancer Institute, Buffalo, New York
| | - Ashley Orillion
- Genitourinary Program, Roswell Park Cancer Institute, Buffalo, New York
| | - Eric Ciamporcero
- Genitourinary Program, Roswell Park Cancer Institute, Buffalo, New York
| | | | - Leigh Ellis
- Genitourinary Program, Roswell Park Cancer Institute, Buffalo, New York
| | - Robert Fenstermaker
- Department of Neurosurgery, Roswell Park Cancer Institute, Buffalo, New York
| | - Scott I Abrams
- Department of Tumor Immunology, Roswell Park Cancer Institute, Buffalo, New York
| | | | - Tomas Leanderson
- Active Biotech AB, Lund, Sweden. Immunology Group, Lund University, Lund, Sweden
| | | | - Roberto Pili
- Genitourinary Program, Roswell Park Cancer Institute, Buffalo, New York.
| |
Collapse
|
120
|
Cancer and bone: A complex complex. Arch Biochem Biophys 2014; 561:159-66. [DOI: 10.1016/j.abb.2014.07.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 07/03/2014] [Accepted: 07/08/2014] [Indexed: 12/13/2022]
|
121
|
Sawant A, Ponnazhagan S. Myeloid-derived suppressor cells as a novel target for the control of osteolytic bone disease. Oncoimmunology 2014; 2:e24064. [PMID: 23762794 PMCID: PMC3667900 DOI: 10.4161/onci.24064] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Accepted: 02/21/2013] [Indexed: 11/29/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSC) from mice bearing bone metastases differentiate into functional osteoclasts in vitro and in vivo, through a signaling pathway that relies on nitric oxide. In addition, MDSC-targeting drugs have been shown to robustly inhibit osteolysis. Thus, MDSC stand out as novel osteoclast progenitors and hence as candidate targets for the control of osteolytic bone disease.
Collapse
Affiliation(s)
- Anandi Sawant
- Department of Pathology; University of Alabama at Birmingham; Birmingham, AL USA
| | | |
Collapse
|
122
|
Sawant A, Ponnazhagan S. Role of plasmacytoid dendritic cells in breast cancer bone dissemination. Oncoimmunology 2014; 2:e22983. [PMID: 23526329 PMCID: PMC3601166 DOI: 10.4161/onci.22983] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Elevated levels of plasmacytoid dendritic cells (pDC) have been observed as breast cancer disseminates to the bone. The selective depletion of pDC in mice led to a total abrogation of bone metastasis as well as to an increase in TH1 antitumor response, suggesting that pDC may be considered as a potential therapeutic target for metastatic breast cancer.
Collapse
Affiliation(s)
- Anandi Sawant
- Department of Pathology; The University of Alabama at Birmingham; Birmingham, AL USA
| | | |
Collapse
|
123
|
Chi N, Tan Z, Ma K, Bao L, Yun Z. Increased circulating myeloid-derived suppressor cells correlate with cancer stages, interleukin-8 and -6 in prostate cancer. Int J Clin Exp Med 2014; 7:3181-3192. [PMID: 25419348 PMCID: PMC4238489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Accepted: 09/25/2014] [Indexed: 06/04/2023]
Abstract
AIM Myeloid-derived suppressor cells (MDSCs) are a population of cells which negatively regulate immune response during tumor progression. In this study, we assessed the accumulation of MDSCs (CD33(+)CD11b(+)HLA-DR(-)CD14(-)) in patients with prostate cancer and its clinical relevance. METHODS We tested the frequency of MDSCs in the peripheral blood of patients with prostate cancer or benign prostate hyperplasia and healthy donors. Serumal interleukin-8, -6 and -10 were analyzed. Effects of MDSCs on the T cell response were determined. RESULTS MDSCs increased in cancer patients, and there was an association between MDSCs and cancer stages or overall survival. Elevated serumal interleukin-8 and -6 in cancer patients correlated with MDSCs. Moreover, accumulation of MDSCs was associated with defective T cell function. CONCLUSION Our study showed an increased population of MDSCs in patients with prostate cancer. Interleukin-8 and -6 in serum may play a new important role companied with MDSCs in prostate cancer.
Collapse
Affiliation(s)
- Ning Chi
- Department of Urology, Inner Mongolia People’s HospitalHohhot, Inner Mongolia Autonomous Region, China
| | - Zhaohui Tan
- Department of Urology, Inner Mongolia People’s HospitalHohhot, Inner Mongolia Autonomous Region, China
| | - Kewei Ma
- Department of Urology, Inner Mongolia People’s HospitalHohhot, Inner Mongolia Autonomous Region, China
| | - Lidao Bao
- Department of Pharmacy, The Affiliated Hospital of Inner Mongolia Medical UniversityHohhot, Inner Mongolia Autonomous Region, China
| | - Zhizhong Yun
- Department of Urology, Inner Mongolia People’s HospitalHohhot, Inner Mongolia Autonomous Region, China
| |
Collapse
|
124
|
Bolt AM, Sabourin V, Molina MF, Police AM, Negro Silva LF, Plourde D, Lemaire M, Ursini-Siegel J, Mann KK. Tungsten targets the tumor microenvironment to enhance breast cancer metastasis. Toxicol Sci 2014; 143:165-77. [PMID: 25324207 DOI: 10.1093/toxsci/kfu219] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The number of individuals exposed to high levels of tungsten is increasing, yet there is limited knowledge of the potential human health risks. Recently, a cohort of breast cancer patients was left with tungsten in their breasts following testing of a tungsten-based shield during intraoperative radiotherapy. While monitoring tungsten levels in the blood and urine of these patients, we utilized the 66Cl4 cell model, in vitro and in mice to study the effects of tungsten exposure on mammary tumor growth and metastasis. We still detect tungsten in the urine of patients' years after surgery (mean urinary tungsten concentration at least 20 months post-surgery = 1.76 ng/ml), even in those who have opted for mastectomy, indicating that tungsten does not remain in the breast. In addition, standard chelation therapy was ineffective at mobilizing tungsten. In the mouse model, tungsten slightly delayed primary tumor growth, but significantly enhanced lung metastasis. In vitro, tungsten did not enhance 66Cl4 proliferation or invasion, suggesting that tungsten was not directly acting on 66Cl4 primary tumor cells to enhance invasion. In contrast, tungsten changed the tumor microenvironment, enhancing parameters known to be important for cell invasion and metastasis including activated fibroblasts, matrix metalloproteinases, and myeloid-derived suppressor cells. We show, for the first time, that tungsten enhances metastasis in an animal model of breast cancer by targeting the microenvironment. Importantly, all these tumor microenvironmental changes are associated with a poor prognosis in humans.
Collapse
Affiliation(s)
- Alicia M Bolt
- *Lady Davis Institute for Medical Research, McGill University, Montréal, Québec, Canada, H3T 1E2, Department of Oncology, McGill University, Montréal, Québec, Canada, H2W 1S6, Pacific Breast Care Center, Costa Mesa, California, 92627, Division of Surgical Oncology, University of California, Irvine School of Medicine, Irvine, California, 92627; and Division of Experimental Medicine, McGill University, Montréal, Québec, Canada, H3A 1A3 *Lady Davis Institute for Medical Research, McGill University, Montréal, Québec, Canada, H3T 1E2, Department of Oncology, McGill University, Montréal, Québec, Canada, H2W 1S6, Pacific Breast Care Center, Costa Mesa, California, 92627, Division of Surgical Oncology, University of California, Irvine School of Medicine, Irvine, California, 92627; and Division of Experimental Medicine, McGill University, Montréal, Québec, Canada, H3A 1A3
| | - Valérie Sabourin
- *Lady Davis Institute for Medical Research, McGill University, Montréal, Québec, Canada, H3T 1E2, Department of Oncology, McGill University, Montréal, Québec, Canada, H2W 1S6, Pacific Breast Care Center, Costa Mesa, California, 92627, Division of Surgical Oncology, University of California, Irvine School of Medicine, Irvine, California, 92627; and Division of Experimental Medicine, McGill University, Montréal, Québec, Canada, H3A 1A3
| | - Manuel Flores Molina
- *Lady Davis Institute for Medical Research, McGill University, Montréal, Québec, Canada, H3T 1E2, Department of Oncology, McGill University, Montréal, Québec, Canada, H2W 1S6, Pacific Breast Care Center, Costa Mesa, California, 92627, Division of Surgical Oncology, University of California, Irvine School of Medicine, Irvine, California, 92627; and Division of Experimental Medicine, McGill University, Montréal, Québec, Canada, H3A 1A3
| | - Alice M Police
- *Lady Davis Institute for Medical Research, McGill University, Montréal, Québec, Canada, H3T 1E2, Department of Oncology, McGill University, Montréal, Québec, Canada, H2W 1S6, Pacific Breast Care Center, Costa Mesa, California, 92627, Division of Surgical Oncology, University of California, Irvine School of Medicine, Irvine, California, 92627; and Division of Experimental Medicine, McGill University, Montréal, Québec, Canada, H3A 1A3 *Lady Davis Institute for Medical Research, McGill University, Montréal, Québec, Canada, H3T 1E2, Department of Oncology, McGill University, Montréal, Québec, Canada, H2W 1S6, Pacific Breast Care Center, Costa Mesa, California, 92627, Division of Surgical Oncology, University of California, Irvine School of Medicine, Irvine, California, 92627; and Division of Experimental Medicine, McGill University, Montréal, Québec, Canada, H3A 1A3
| | - Luis Fernando Negro Silva
- *Lady Davis Institute for Medical Research, McGill University, Montréal, Québec, Canada, H3T 1E2, Department of Oncology, McGill University, Montréal, Québec, Canada, H2W 1S6, Pacific Breast Care Center, Costa Mesa, California, 92627, Division of Surgical Oncology, University of California, Irvine School of Medicine, Irvine, California, 92627; and Division of Experimental Medicine, McGill University, Montréal, Québec, Canada, H3A 1A3 *Lady Davis Institute for Medical Research, McGill University, Montréal, Québec, Canada, H3T 1E2, Department of Oncology, McGill University, Montréal, Québec, Canada, H2W 1S6, Pacific Breast Care Center, Costa Mesa, California, 92627, Division of Surgical Oncology, University of California, Irvine School of Medicine, Irvine, California, 92627; and Division of Experimental Medicine, McGill University, Montréal, Québec, Canada, H3A 1A3
| | - Dany Plourde
- *Lady Davis Institute for Medical Research, McGill University, Montréal, Québec, Canada, H3T 1E2, Department of Oncology, McGill University, Montréal, Québec, Canada, H2W 1S6, Pacific Breast Care Center, Costa Mesa, California, 92627, Division of Surgical Oncology, University of California, Irvine School of Medicine, Irvine, California, 92627; and Division of Experimental Medicine, McGill University, Montréal, Québec, Canada, H3A 1A3
| | - Maryse Lemaire
- *Lady Davis Institute for Medical Research, McGill University, Montréal, Québec, Canada, H3T 1E2, Department of Oncology, McGill University, Montréal, Québec, Canada, H2W 1S6, Pacific Breast Care Center, Costa Mesa, California, 92627, Division of Surgical Oncology, University of California, Irvine School of Medicine, Irvine, California, 92627; and Division of Experimental Medicine, McGill University, Montréal, Québec, Canada, H3A 1A3 *Lady Davis Institute for Medical Research, McGill University, Montréal, Québec, Canada, H3T 1E2, Department of Oncology, McGill University, Montréal, Québec, Canada, H2W 1S6, Pacific Breast Care Center, Costa Mesa, California, 92627, Division of Surgical Oncology, University of California, Irvine School of Medicine, Irvine, California, 92627; and Division of Experimental Medicine, McGill University, Montréal, Québec, Canada, H3A 1A3
| | - Josie Ursini-Siegel
- *Lady Davis Institute for Medical Research, McGill University, Montréal, Québec, Canada, H3T 1E2, Department of Oncology, McGill University, Montréal, Québec, Canada, H2W 1S6, Pacific Breast Care Center, Costa Mesa, California, 92627, Division of Surgical Oncology, University of California, Irvine School of Medicine, Irvine, California, 92627; and Division of Experimental Medicine, McGill University, Montréal, Québec, Canada, H3A 1A3 *Lady Davis Institute for Medical Research, McGill University, Montréal, Québec, Canada, H3T 1E2, Department of Oncology, McGill University, Montréal, Québec, Canada, H2W 1S6, Pacific Breast Care Center, Costa Mesa, California, 92627, Division of Surgical Oncology, University of California, Irvine School of Medicine, Irvine, California, 92627; and Division of Experimental Medicine, McGill University, Montréal, Québec, Canada, H3A 1A3 *Lady Davis Institute for Medical Research, McGill University, Montréal, Québec, Canada, H3T 1E2, Department of Oncology, McGill University, Montréal, Québec, Canada, H2W 1S6, Pacific Breast Care Center, Costa Mesa, California, 92627, Division of Surgical Oncology, University of California, Irvine School of Medicine, Irvine, California, 92627; and Division of Experimental Medicine, McGill University, Montréal, Québec, Canada, H3A 1A3
| | - Koren K Mann
- *Lady Davis Institute for Medical Research, McGill University, Montréal, Québec, Canada, H3T 1E2, Department of Oncology, McGill University, Montréal, Québec, Canada, H2W 1S6, Pacific Breast Care Center, Costa Mesa, California, 92627, Division of Surgical Oncology, University of California, Irvine School of Medicine, Irvine, California, 92627; and Division of Experimental Medicine, McGill University, Montréal, Québec, Canada, H3A 1A3 *Lady Davis Institute for Medical Research, McGill University, Montréal, Québec, Canada, H3T 1E2, Department of Oncology, McGill University, Montréal, Québec, Canada, H2W 1S6, Pacific Breast Care Center, Costa Mesa, California, 92627, Division of Surgical Oncology, University of California, Irvine School of Medicine, Irvine, California, 92627; and Division of Experimental Medicine, McGill University, Montréal, Québec, Canada, H3A 1A3 *Lady Davis Institute for Medical Research, McGill University, Montréal, Québec, Canada, H3T 1E2, Department of Oncology, McGill University, Montréal, Québec, Canada, H2W 1S6, Pacific Breast Care Center, Costa Mesa, California, 92627, Division of Surgical Oncology, University of California, Irvine School of Medicine, Irvine, California, 92627; and Division of Experimental Medicine, McGill University, Montréal, Québec, Canada, H3A 1A3
| |
Collapse
|
125
|
Condamine T, Ramachandran I, Youn JI, Gabrilovich DI. Regulation of tumor metastasis by myeloid-derived suppressor cells. Annu Rev Med 2014; 66:97-110. [PMID: 25341012 DOI: 10.1146/annurev-med-051013-052304] [Citation(s) in RCA: 384] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Accumulation of pathologically activated immature myeloid cells with potent immune-suppressive activity is one of the major immunological hallmarks of cancer. In recent years, it became clear that in addition to their immune-suppressive activity, myeloid-derived suppressor cells (MDSCs) influence tumor progression in a variety of ways. They are directly implicated in the promotion of tumor metastases by participating in the formation of premetastatic niches, promoting angiogenesis and tumor cell invasion. In this review, we discuss recent data describing various roles of MDSCs in the formation of tumor metastases.
Collapse
|
126
|
Charles JF, Aliprantis AO. Osteoclasts: more than 'bone eaters'. Trends Mol Med 2014; 20:449-59. [PMID: 25008556 PMCID: PMC4119859 DOI: 10.1016/j.molmed.2014.06.001] [Citation(s) in RCA: 265] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 05/28/2014] [Accepted: 06/02/2014] [Indexed: 02/08/2023]
Abstract
As the only cells definitively shown to degrade bone, osteoclasts are key mediators of skeletal diseases including osteoporosis. Bone-forming osteoblasts, and hematopoietic and immune system cells, each influence osteoclast formation and function, but the reciprocal impact of osteoclasts on these cells is less well appreciated. We highlight here the functions that osteoclasts perform beyond bone resorption. First, we consider how osteoclast signals may contribute to bone formation by osteoblasts and to the pathology of bone lesions such as fibrous dysplasia and giant cell tumors. Second, we review the interaction of osteoclasts with the hematopoietic system, including the stem cell niche and adaptive immune cells. Connections between osteoclasts and other cells in the bone microenvironment are discussed within a clinically relevant framework.
Collapse
Affiliation(s)
- Julia F Charles
- Department of Medicine, Division of Rheumatology, Allergy, and Immunology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Antonios O Aliprantis
- Department of Medicine, Division of Rheumatology, Allergy, and Immunology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
127
|
Bruno A, Pagani A, Pulze L, Albini A, Dallaglio K, Noonan DM, Mortara L. Orchestration of angiogenesis by immune cells. Front Oncol 2014; 4:131. [PMID: 25072019 PMCID: PMC4078768 DOI: 10.3389/fonc.2014.00131] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Accepted: 05/16/2014] [Indexed: 12/20/2022] Open
Abstract
It is widely accepted that the tumor microenvironment (TUMIC) plays a major role in cancer and is indispensable for tumor progression. The TUMIC involves many "players" going well beyond the malignant-transformed cells, including stromal, immune, and endothelial cells (ECs). The non-malignant cells can acquire tumor-promoting functions during carcinogenesis. In particular, these cells can "orchestrate" the "symphony" of the angiogenic switch, permitting the creation of new blood vessels that allows rapid expansion and progression toward malignancy. Considerable attention within the context of tumor angiogenesis should focus not only on the ECs, representing a fundamental unit, but also on immune cells and on the inflammatory tumor infiltrate. Immune cells infiltrating tumors typically show a tumor-induced polarization associated with attenuation of anti-tumor functions and generation of pro-tumor activities, among these angiogenesis. Here, we propose a scenario suggesting that the angiogenic switch is an immune switch arising from the pro-angiogenic polarization of immune cells. This view links immunity, inflammation, and angiogenesis to tumor progression. Here, we review the data in the literature and seek to identify the "conductors" of this "orchestra." We also suggest that interrupting the immune → inflammation → angiogenesis → tumor progression process can delay or prevent tumor insurgence and malignant disease.
Collapse
Affiliation(s)
- Antonino Bruno
- Scientific and Technology Pole, IRCCS MultiMedica , Milan , Italy
| | - Arianna Pagani
- Department of Biotechnology and Life Sciences, University of Insubria , Varese , Italy
| | - Laura Pulze
- Department of Biotechnology and Life Sciences, University of Insubria , Varese , Italy
| | - Adriana Albini
- Department of Research and Statistics, IRCCS Arcispedale Santa Maria Nuova , Reggio Emilia , Italy
| | - Katiuscia Dallaglio
- Department of Research and Statistics, IRCCS Arcispedale Santa Maria Nuova , Reggio Emilia , Italy
| | - Douglas M Noonan
- Scientific and Technology Pole, IRCCS MultiMedica , Milan , Italy ; Department of Biotechnology and Life Sciences, University of Insubria , Varese , Italy
| | - Lorenzo Mortara
- Department of Biotechnology and Life Sciences, University of Insubria , Varese , Italy
| |
Collapse
|
128
|
Dissecting the role of bone marrow stromal cells on bone metastases. BIOMED RESEARCH INTERNATIONAL 2014; 2014:875305. [PMID: 25054153 PMCID: PMC4099112 DOI: 10.1155/2014/875305] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 06/10/2014] [Indexed: 12/20/2022]
Abstract
Tumor-induced bone disease is a dynamic process that involves interactions with many cell types. Once metastatic cancer cells reach the bone, they are in contact with many different cell types that are present in the cell-rich bone marrow. These cells include the immune cells, myeloid cells, fibroblasts, osteoblasts, osteoclasts, and mesenchymal stem cells. Each of these cell populations can influence the behavior or gene expression of both the tumor cells and the bone microenvironment. Additionally, the tumor itself can alter the behavior of these bone marrow cells which further alters both the microenvironment and the tumor cells. While many groups focus on studying these interactions, much remains unknown. A better understanding of the interactions between the tumor cells and the bone microenvironment will improve our knowledge on how tumors establish in bone and may lead to improvements in diagnosing and treating bone metastases. This review details our current knowledge on the interactions between tumor cells that reside in bone and their microenvironment.
Collapse
|
129
|
Extracellular Membrane Vesicles Derived from 143B Osteosarcoma Cells Contain Pro-Osteoclastogenic Cargo: A Novel Communication Mechanism in Osteosarcoma Bone Microenvironment. Transl Oncol 2014; 7:331-40. [PMID: 25180057 PMCID: PMC4145399 DOI: 10.1016/j.tranon.2014.04.011] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 01/21/2014] [Accepted: 03/03/2014] [Indexed: 12/21/2022] Open
Abstract
The bone microenvironment (BME) is the main hub of all skeletal related pathological events in osteosarcoma leading to tumor induced bone destruction, and decreasing overall bone quality and bone strength. The role of extra-cellular membrane vesicles (EMVs) as mediators of intercellular communication in modulating osteosarcoma-BME is unknown, and needs to be investigated. It is our hypothesis that osteosarcoma-EMVs contain pro-osteoclastogenic cargo which increases osteoclastic activity, and dysregulated bone remodeling in the osteosarcoma-BME. In this study, EMVs were isolated from the conditioned media of 143B and HOS human osteosarcoma cell cultures using differential ultracentrifugation. Nano-particle tracking analysis determined EMVs in the size range of 50-200 nm in diameter. The EMV yield from 143B cells was relatively higher compared to HOS cells. Transmission electron microscopy confirmed the ultrastructure of 143B-EMVs and detected multivesicular bodies. Biochemical characterization of 143B-EMVs detected the expression of bioactive pro-osteoclastic cargo including matrix metalloproteinases-1 and -13 (MMP-1, -13), transforming growth factor-β (TGF-β), CD-9, and receptor activator of nuclear factor kappa-β ligand (RANKL). Detection of a protein signature that is uniquely pro-osteoclastic in 143B-EMVs is a novel finding, and is significant as EMVs represent an interesting mechanism for potentially mediating bone destruction in the osteosarcoma-BME. This study further demonstrates that 143B cells actively mobilize calcium in the presence of ionomycin, and forskolin, and induce cytoskeleton rearrangements leading to vesicular biogenesis. In conclusion, this study demonstrates that 143B osteosarcoma cells generate EMVs mainly by mechanisms involving increased intracellular calcium or cAMP levels, and contain pro-osteoclastic cargo.
Collapse
|
130
|
Immunological dysregulation in multiple myeloma microenvironment. BIOMED RESEARCH INTERNATIONAL 2014; 2014:198539. [PMID: 25013764 PMCID: PMC4071780 DOI: 10.1155/2014/198539] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 05/20/2014] [Indexed: 12/22/2022]
Abstract
Multiple Myeloma (MM) is a systemic hematologic disease due to uncontrolled proliferation of monoclonal plasma cells (PC) in bone marrow (BM). Emerging in other solid and liquid cancers, the host immune system and the microenvironment have a pivotal role for PC growth, proliferation, survival, migration, and resistance to drugs and are responsible for some clinical manifestations of MM. In MM, microenvironment is represented by the cellular component of a normal bone marrow together with extracellular matrix proteins, adhesion molecules, cytokines, and growth factors produced by both stromal cells and PC themselves. All these components are able to protect PC from cytotoxic effect of chemo- and radiotherapy. This review is focused on the role of immunome to sustain MM progression, the emerging role of myeloid derived suppressor cells, and their potential clinical implications as novel therapeutic target.
Collapse
|
131
|
Šućur A, Katavić V, Kelava T, Jajić Z, Kovačić N, Grčević D. Induction of osteoclast progenitors in inflammatory conditions: key to bone destruction in arthritis. INTERNATIONAL ORTHOPAEDICS 2014; 38:1893-903. [DOI: 10.1007/s00264-014-2386-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 05/13/2014] [Indexed: 12/14/2022]
|
132
|
Mao Y, Sarhan D, Steven A, Seliger B, Kiessling R, Lundqvist A. Inhibition of tumor-derived prostaglandin-e2 blocks the induction of myeloid-derived suppressor cells and recovers natural killer cell activity. Clin Cancer Res 2014; 20:4096-106. [PMID: 24907113 DOI: 10.1158/1078-0432.ccr-14-0635] [Citation(s) in RCA: 219] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
PURPOSE Increased frequencies of myeloid-derived suppressor cells (MDSC) correlate with poor prognosis in patients with cancers. Tumor-derived prostaglandin-E2 (PGE2) plays an important role in inducing MDSCs. However, the detailed mechanisms of this induction remain unknown. To develop targeted therapies for MDSCs, we sought to investigate the molecular basis of PGE2-regulated accumulation of MDSCs and their functional consequence on natural killer (NK) cell activity. EXPERIMENTAL DESIGN The effects of PGE2 in inducing phenotypic, signaling, and functional alternations on monocytes were analyzed in vitro. Suppression of NK-cell activity by PGE2-treated monocytes was compared with that of freshly isolated CD14(+)HLA-DR(low/-) monocytic MDSCs (moMDSC) from patients with melanoma. In addition, to explore the in vivo relevance of targeting PGE2 to reduce MDSC-mediated suppression of NK cells, we established a murine model, where tumor cells were disabled from cyclooxygenase-2 (COX-2) production. RESULTS Patient-derived moMDSCs inhibited NK-cell activity through the production of TGFβ. In vitro, binding of PGE2 to EP2 and EP4 receptors on monocytes activated the p38MAPK/ERK pathway and resulted in elevated secretion of TGFβ. Similar to moMDSCs, PGE2-treated monocytes potently suppressed NK-cell activity through production of TGFβ. Furthermore, silencing COX-2 in murine 4T1 tumor cells reduced the accumulation of CD11b(+)Gr1(+) MDSCs in the spleen, resulting in concomitant improved in vivo clearance of NK-cell sensitive YAC-1 cells. CONCLUSIONS Our results reveal an indispensable role of tumor-derived PGE2 in inducing MDSCs and suggest a favorable outcome of combining COX-2-targeted therapy and adoptive NK-cell transfer in patients with cancer.
Collapse
Affiliation(s)
- Yumeng Mao
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden; and
| | - Dhifaf Sarhan
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden; and
| | - André Steven
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Barbara Seliger
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Rolf Kiessling
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden; and
| | - Andreas Lundqvist
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden; and
| |
Collapse
|
133
|
Browne G, Taipaleenmäki H, Stein GS, Stein JL, Lian JB. MicroRNAs in the control of metastatic bone disease. Trends Endocrinol Metab 2014; 25:320-7. [PMID: 24811921 PMCID: PMC4075094 DOI: 10.1016/j.tem.2014.03.014] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 03/21/2014] [Accepted: 03/25/2014] [Indexed: 02/08/2023]
Abstract
Bone metastasis is a common and devastating complication of late-stage breast and prostate cancer. Complex interactions between tumor cells, bone cells, and a milieu of components in their microenvironment contribute to the osteolytic, osteoblastic, or mixed lesions present in patients with metastasis to bone. In the past decade microRNAs (miRNAs) have emerged as key players in cancer progression, but the importance of miRNAs in regulating cancer metastasis to bone is only now being appreciated. We emphasize here important concepts of bone biology and miRNAs in the context of breast and prostate cancer, and focus on recent advances that have improved our understanding of the role of specific miRNAs with direct involvement in metastatic bone disease.
Collapse
Affiliation(s)
- Gillian Browne
- Vermont Cancer Center and Department of Biochemistry, University of Vermont College of Medicine, Burlington, VT, 05405, USA
| | - Hanna Taipaleenmäki
- Heisenberg-Group for Molecular Skeletal Biology, Department of Trauma, Hand and Reconstructive Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gary S Stein
- Vermont Cancer Center and Department of Biochemistry, University of Vermont College of Medicine, Burlington, VT, 05405, USA
| | - Janet L Stein
- Vermont Cancer Center and Department of Biochemistry, University of Vermont College of Medicine, Burlington, VT, 05405, USA
| | - Jane B Lian
- Vermont Cancer Center and Department of Biochemistry, University of Vermont College of Medicine, Burlington, VT, 05405, USA.
| |
Collapse
|
134
|
Roato I. Bone metastases: When and how lung cancer interacts with bone. World J Clin Oncol 2014; 5:149-155. [PMID: 24829862 PMCID: PMC4014787 DOI: 10.5306/wjco.v5.i2.149] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 02/21/2014] [Accepted: 03/18/2014] [Indexed: 02/06/2023] Open
Abstract
Bone metastasis is a common and debilitating consequence of lung cancer: 30%-40% of patients with non-small cell lung cancer develop bone metastases during the course of their disease. Lung cancer cells find a favorable soil in the bone microenvironment due to factors released by the bone matrix, the immune system cells, and the same cancer cells. Many aspects of the cross-talk among lung tumor cells, the immune system, and bone cells are not clear, but this review aims to summarize the recent findings in this field, with particular attention to studies conducted to identify biomarkers for early detection of lung cancer bone metastases.
Collapse
|
135
|
Olechnowicz SWZ, Edwards CM. Contributions of the host microenvironment to cancer-induced bone disease. Cancer Res 2014; 74:1625-31. [PMID: 24599133 DOI: 10.1158/0008-5472.can-13-2645] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The bone marrow provides a specialized and highly supportive microenvironment for tumor growth and development of the associated bone disease. It is a preferred site for breast and prostate cancer bone metastasis and the hematologic malignancy, multiple myeloma. For many years, researchers have focused upon the interactions between tumor cells and the cells directly responsible for bone remodeling, namely osteoclasts and osteoblasts. However, there is ever-increasing evidence for a multitude of ways in which the bone marrow microenvironment can promote disease pathogenesis, including via cancer-associated fibroblasts, the hematopoietic stem cell niche, myeloid-derived suppressor cells, and the sympathetic nervous system. This review discusses the recent advances in our understanding of the contribution of the host microenvironment to the development of cancer-induced bone disease.
Collapse
Affiliation(s)
- Sam W Z Olechnowicz
- Authors' Affiliations: Nuffield Department of Surgical Sciences and Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
136
|
Abstract
The immune system and bone are intimately linked with significant physical and functionally related interactions. The innate immune system functions as an immediate response system to initiate protections against local challenges such as pathogens and cellular damage. Bone is a very specific microenvironment, in which infectious attack is less common but repair and regeneration are ongoing and important functions. Thus, in the bone the primary goal of innate immune and bone interactions is to maintain tissue integrity. Innate immune signals are critical for removal of damaged and apoptotic cells and to stimulate normal tissue repair and regeneration. In this review we focus on the innate immune mechanisms that function to regulate bone homeostasis.
Collapse
Affiliation(s)
- Julia F. Charles
- Department of Medicine, Division of Rheumatology, Allergy and Immunology, Brigham and Women’s Hospital, 75 Francis Street, Boston, MA, 02115 Phone: FAX:
| | - Mary C. Nakamura
- Department of Medicine, Division of Rheumatology, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA, 94143
- Arthritis/Immunology Section, Veterans Affairs Medical Center, 4150 Clement Street, 111R, San Francisco, CA 94121, Phone: 415 750-2104, FAX: 415 750-6920,
- corresponding author: Arthritis/Immunology Section, Veterans Affairs Medical Center, 4150 Clement Street, 111R, San Francisco, CA 94121
| |
Collapse
|
137
|
Diaz-Montero CM, Finke J, Montero AJ. Myeloid-derived suppressor cells in cancer: therapeutic, predictive, and prognostic implications. Semin Oncol 2014; 41:174-84. [PMID: 24787291 DOI: 10.1053/j.seminoncol.2014.02.003] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Immune evasion is a hallmark of cancer. While there are multiple different mechanisms that cancer cells employ, myeloid-derived suppressor cells (MDSCs) are one of the key drivers of tumor-mediated immune evasion. MDSCs begin as myeloid cells recruited to the tumor microenvironment, where they are transformed into potent immunosuppressive cells. However, our understanding of the clinical relevance of MDSCs in cancer patients has significantly lagged behind the preclinical literature in part due to the absence of a cognate molecule present in mice, as well as to the considerable heterogeneity of MDSCs. However, if one evaluates the clinical literature through the filter of clinically robust endpoints, such as overall survival, three important phenotypes emerge: promyelocytic, monocytic, and granulocytic. Based on these studies, MDSCs have clear prognostic importance in multiple solid tumors, and emerging data support the utility of circulating MDSCs as a predictive marker for cancer immunotherapy, and even as an early leading marker for predicting clinical response to systemic chemotherapy in patients with advanced solid tumors. More recent preclinical data in immunosuppressed murine models suggest that MDSCs play an important role in tumor progression and the metastatic process that is independent of their immunosuppressive properties. Consequently, targeting MDSCs either in combination with cancer immunotherapy or independently as part of an approach to inhibit the metastatic process appears to be a very clinically promising strategy. We review different approaches to target MDSCs that could potentially be tested in future clinical trials in cancer patients.
Collapse
Affiliation(s)
- C Marcela Diaz-Montero
- Lerner Research Institute Department of Immunology; Cleveland Clinic Foundation, Cleveland, OH
| | - Jim Finke
- Lerner Research Institute Department of Immunology; Cleveland Clinic Foundation, Cleveland, OH
| | - Alberto J Montero
- Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, OH.
| |
Collapse
|
138
|
Faccio R, Capietto AH, Stewart SA, Novack DV. Cellular Players in Breast Cancer Bone Metastases. Clin Rev Bone Miner Metab 2013. [DOI: 10.1007/s12018-013-9146-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
139
|
Capietto AH, Kim S, Sanford DE, Linehan DC, Hikida M, Kumosaki T, Novack DV, Faccio R. Down-regulation of PLCγ2-β-catenin pathway promotes activation and expansion of myeloid-derived suppressor cells in cancer. ACTA ACUST UNITED AC 2013; 210:2257-71. [PMID: 24127488 PMCID: PMC3804931 DOI: 10.1084/jem.20130281] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) favor tumor promotion, mainly by suppressing antitumor T cell responses in many cancers. Although the mechanism of T cell inhibition is established, the pathways leading to MDSC accumulation in bone marrow and secondary lymphoid organs of tumor-bearing hosts remain unclear. We demonstrate that down-regulation of PLCγ2 signaling in MDSCs is responsible for their aberrant expansion during tumor progression. PLCγ2(-/-) MDSCs show stronger immune-suppressive activity against CD8(+) T cells than WT MDSCs and potently promote tumor growth when adoptively transferred into WT mice. Mechanistically, PLCγ2(-/-) MDSCs display reduced β-catenin levels, and restoration of β-catenin expression decreases their expansion and tumor growth. Consistent with a negative role for β-catenin in MDSCs, its deletion in the myeloid population leads to MDSC accumulation and supports tumor progression, whereas expression of β-catenin constitutively active reduces MDSC numbers and protects from tumor growth. Further emphasizing the clinical relevance of these findings, MDSCs isolated from pancreatic cancer patients show reduced p-PLCγ2 and β-catenin levels compared with healthy controls, similar to tumor-bearing mice. Thus, for the first time, we demonstrate that down-regulation of PLCγ2-β-catenin pathway occurs in mice and humans and leads to MDSC-mediated tumor expansion, raising concerns about the efficacy of systemic β-catenin blockade as anti-cancer therapy.
Collapse
Affiliation(s)
- Aude-Hélène Capietto
- Department of Orthopedics, 2 Department of Surgery, 3 Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | | | | | | | | | | | | | | |
Collapse
|
140
|
Sawant A, Ponnazhagan S. Myeloid-derived suppressor cells as osteoclast progenitors: a novel target for controlling osteolytic bone metastasis. Cancer Res 2013; 73:4606-10. [PMID: 23887974 DOI: 10.1158/0008-5472.can-13-0305] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Immune cells and their secreted growth factors play major roles in tumor growth and metastasis. Interplay between the growing tumor and infiltrating immune cells determines the nature of immune response and ultimately, tumor fate. Increased infiltration of protumorigenic immune cells promotes tumor growth as well as dissemination to distant sites. These cells induce immunosuppression that inhibits proliferation and functions of cells of antitumor immune response. One population of immunosuppressive cells that is increasingly gaining attention is myeloid-derived suppressor cells (MDSC). MDSCs are immature myeloid progenitors that suppress T-cell effector functions and promote angiogenesis. MDSC numbers are elevated at both the primary tumor and metastatic sites, including bone. In addition to immunosuppressive functions of MDSCs, we and others have recently discovered a novel function for MDSCs as osteoclast progenitors. Osteolysis is a common complication in the carcinomas of breast, lung, prostate, and multiple myeloma with poor prognosis. Therefore, targeting the functions of MDSCs may exert dual therapeutic effects on immunosuppression and bone pathology. Cancer Res; 73(15); 4606-10. ©2013 AACR.
Collapse
Affiliation(s)
- Anandi Sawant
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | |
Collapse
|
141
|
The role of MDSCs in bone metastasis and cancer-related osteolysis. BONEKEY REPORTS 2013; 2:311. [PMID: 24422063 PMCID: PMC3722748 DOI: 10.1038/bonekey.2013.45] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
|