Kobe University Repository : Kernel タイトル Tit le CD 244 + polymorphonuclear myeloid ‐ derived suppressor cells reflect the status of peritoneal disseminat ion in a colon cancer mouse model 著者

Despite the recent development of chemotherapeutic agents, the prognosis of colorectal cancer (CRC) patients with peritoneal dissemination (PD) remains poor. The tumor immune microenvironment (TIME) has drawn attention as a key contributing factor of tumor progression. Of TIME components, myeloid‐derived suppressor cells (MDSCs) are considered to play a responsible role in the immunosuppres‐ sive characteristics of the TIME. MDSCs are classified into two major subsets: Monocytic MDSCs (M‐MDSCs) and polymorphonuclear MDSCs (PMN‐MDSCs). Therefore, we hypothesize that MDSCs would play important roles in the PD‐relevant TIME and PD progression. To address this hypothesis, we established PD mouse models. As the PD nodules consisted scarcely of immune cells, we focused on the peritoneal cavity, but not PD nodule, to evaluate the PD‐relevant TIME. As a result, intraperitoneal PMN‐MDSCs were found to be substantially increased in association with PD progression. Based on these results, we phenotypi‐ cally and functionally verified the usefulness of CD244 for identifying PMN‐MDSCs. In addition, the concentrations of interleukin (IL)‐6 and granulocyte‐colony stimulating factor (G‐CSF) were significantly increased in the peritoneal cavity, both of which were produced by the tumors and thought to contribute to the increases in the PMN‐MDSCs. In vivo depletion of the PMN‐MDSCs by anti‐Ly6G monoclonal antibody (mAb) significantly inhibited the PD progression and reverted CD4+ and CD8+ T cells in the peritoneal cavity and the peripheral blood. Collectively, these results suggest that the targeted therapy for PMN‐MDSCs would provide not only new therapeutic value but also a novel strategy to synergize with T‐cell‐based immunotherapy for CRC‐derived PD.

[1]  D. Gabrilovich,et al.  Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity , 2021, Nature Reviews Immunology.

[2]  Ihor O. Arkhypov,et al.  IL-6 as a major regulator of MDSC activity and possible target for cancer immunotherapy. , 2020, Cellular immunology.

[3]  Yu Seong Lee,et al.  Oncolytic vaccinia virus reinvigorates peritoneal immunity and cooperates with immune checkpoint inhibitor to suppress peritoneal carcinomatosis in colon cancer , 2020, Journal for ImmunoTherapy of Cancer.

[4]  S. Abrams,et al.  Granulocytic Myeloid-Derived Suppressor Cells as Negative Regulators of Anticancer Immunity , 2020, Frontiers in Immunology.

[5]  K. Hoebe,et al.  CD244 represents a new therapeutic target in head and neck squamous cell carcinoma , 2020, Journal for ImmunoTherapy of Cancer.

[6]  O. De Wever,et al.  Targeting the Tumor Microenvironment in Colorectal Peritoneal Metastases. , 2020, Trends in cancer.

[7]  D. Gallego-Ortega,et al.  Myeloid-Derived Suppressor Cells as a Therapeutic Target for Cancer , 2020, Cells.

[8]  H. Rugo,et al.  NCCN Guidelines Insights: Hematopoietic Growth Factors, Version 1.2020. , 2020, Journal of the National Comprehensive Cancer Network : JNCCN.

[9]  J. Pouysségur,et al.  Tumor Microenvironment: A Metabolic Player that Shapes the Immune Response , 2019, International journal of molecular sciences.

[10]  A. Kotsakis,et al.  Myeloid-Derived Suppressor Cells: Major Figures that Shape the Immunosuppressive and Angiogenic Network in Cancer , 2019, Cells.

[11]  C. Yardin,et al.  Comparison of implantation sites for the development of peritoneal metastasis in a colorectal cancer mouse model using non-invasive bioluminescence imaging , 2019, PloS one.

[12]  J. Schlom,et al.  Inhibiting myeloid-derived suppressor cell trafficking enhances T cell immunotherapy. , 2019, JCI insight.

[13]  M. Kurachi CD8+ T cell exhaustion , 2019, Seminars in Immunopathology.

[14]  M. Masařík,et al.  Effect of tumor microenvironment on pathogenesis of the head and neck squamous cell carcinoma: a systematic review , 2019, Molecular Cancer.

[15]  E. Moon,et al.  CAR T Cells for Solid Tumors: New Strategies for Finding, Infiltrating, and Surviving in the Tumor Microenvironment , 2019, Front. Immunol..

[16]  J. Pollard,et al.  Deciphering myeloid-derived suppressor cells: isolation and markers in humans, mice and non-human primates , 2019, Cancer Immunology, Immunotherapy.

[17]  K. Hoebe,et al.  The Emerging Role of CD244 Signaling in Immune Cells of the Tumor Microenvironment , 2018, Front. Immunol..

[18]  A. Jemal,et al.  Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries , 2018, CA: a cancer journal for clinicians.

[19]  Sean C. Bendall,et al.  A Structured Tumor-Immune Microenvironment in Triple Negative Breast Cancer Revealed by Multiplexed Ion Beam Imaging , 2018, Cell.

[20]  G. Lyman,et al.  Overall survival and risk of second malignancies with cancer chemotherapy and G-CSF support , 2018, Annals of oncology : official journal of the European Society for Medical Oncology.

[21]  Y. Kakeji,et al.  Immunosuppression Induced by Perioperative Peritonitis Promotes Lung Metastasis , 2018, AntiCancer Research.

[22]  D. Valík,et al.  Myeloid-derived suppressor cells (MDSCs) in patients with solid tumors: considerations for granulocyte colony-stimulating factor treatment , 2018, Cancer Immunology, Immunotherapy.

[23]  R. Weinberg,et al.  Understanding the tumor immune microenvironment (TIME) for effective therapy , 2018, Nature Medicine.

[24]  Z. Fan,et al.  Highly Expressed Granulocyte Colony-Stimulating Factor (G-CSF) and Granulocyte Colony-Stimulating Factor Receptor (G-CSFR) in Human Gastric Cancer Leads to Poor Survival , 2018, Medical science monitor : international medical journal of experimental and clinical research.

[25]  D. Gabrilovich,et al.  Myeloid-derived suppressor cells coming of age , 2018, Nature Immunology.

[26]  Xiaomin Su,et al.  LncRNA RNCR3 promotes Chop expression by sponging miR-185-5p during MDSC differentiation , 2017, Oncotarget.

[27]  Laurence Zitvogel,et al.  The immune contexture in cancer prognosis and treatment , 2017, Nature Reviews Clinical Oncology.

[28]  Y. Kakeji,et al.  Frequency of Myeloid-derived Suppressor Cells in the Peripheral Blood Reflects the Status of Tumor Recurrence. , 2017, Anticancer research.

[29]  F. Pedeutour,et al.  Peritoneal carcinomatosis of colorectal cancer: novel clinical and molecular outcomes. , 2017, American journal of surgery.

[30]  C. Tournigand,et al.  Prognosis of patients with peritoneal metastatic colorectal cancer given systemic therapy: an analysis of individual patient data from prospective randomised trials from the Analysis and Research in Cancers of the Digestive System (ARCAD) database. , 2016, The Lancet. Oncology.

[31]  K. E. Visser,et al.  Neutrophils in cancer: neutral no more , 2016, Nature Reviews Cancer.

[32]  A. Biankin,et al.  CXCR2 Inhibition Profoundly Suppresses Metastases and Augments Immunotherapy in Pancreatic Ductal Adenocarcinoma , 2016, Cancer cell.

[33]  M. Gunzer,et al.  Survival of residual neutrophils and accelerated myelopoiesis limit the efficacy of antibody‐mediated depletion of Ly‐6G+ cells in tumor‐bearing mice , 2016, Journal of leukocyte biology.

[34]  D. Mougiakakos,et al.  TNF-Mediated Restriction of Arginase 1 Expression in Myeloid Cells Triggers Type 2 NO Synthase Activity at the Site of Infection. , 2016, Cell reports.

[35]  E. Morii,et al.  The significance of G-CSF expression and myeloid-derived suppressor cells in the chemoresistance of uterine cervical cancer , 2015, Scientific Reports.

[36]  Y. Kakeji,et al.  Early-phase Treatment by Low-dose 5-Fluorouracil or Primary Tumor Resection Inhibits MDSC-mediated Lung Metastasis Formation. , 2015, Anticancer research.

[37]  E. Wherry,et al.  Molecular and cellular insights into T cell exhaustion , 2015, Nature Reviews Immunology.

[38]  P. Sugarbaker Colorectal Cancer: Prevention and Management of Metastatic Disease , 2014, BioMed research international.

[39]  I. Pinchuk,et al.  G-CSF and G-CSFR are highly expressed in human gastric and colon cancers and promote carcinoma cell proliferation and migration , 2014, British Journal of Cancer.

[40]  Ada T Feldman,et al.  Tissue processing and hematoxylin and eosin staining. , 2014, Methods in molecular biology.

[41]  W. Pan,et al.  Monocyte/macrophage‐elicited natural killer cell dysfunction in hepatocellular carcinoma is mediated by CD48/2B4 interactions , 2013, Hepatology.

[42]  Xiaopei Huang,et al.  NK Cell Response to Vaccinia Virus Is Regulated by Myeloid-Derived Suppressor Cells , 2012, The Journal of Immunology.

[43]  R. Bleichrodt,et al.  Outcomes of colorectal cancer patients with peritoneal carcinomatosis treated with chemotherapy with and without targeted therapy. , 2012, European journal of surgical oncology : the journal of the European Society of Surgical Oncology and the British Association of Surgical Oncology.

[44]  F. Granath,et al.  Incidence, prevalence and risk factors for peritoneal carcinomatosis from colorectal cancer , 2012, The British journal of surgery.

[45]  I. Cuthill,et al.  Improving Bioscience Research Reporting: The ARRIVE Guidelines for Reporting Animal Research † , 2012, Osteoarthritis and cartilage.

[46]  D. Gabrilovich,et al.  Coordinated regulation of myeloid cells by tumours , 2012, Nature Reviews Immunology.

[47]  S. Biswas,et al.  Characterization of the nature of granulocytic myeloid‐derived suppressor cells in tumor‐bearing mice , 2012, Journal of leukocyte biology.

[48]  Qiang Hu,et al.  Tumor-Derived G-CSF Facilitates Neoplastic Growth through a Granulocytic Myeloid-Derived Suppressor Cell-Dependent Mechanism , 2011, PloS one.

[49]  D. Gabrilovich,et al.  Molecular mechanisms regulating myeloid-derived suppressor cell differentiation and function. , 2011, Trends in immunology.

[50]  K. Sugamura,et al.  Generation of a syngeneic mouse model to study the intraperitoneal dissemination of ovarian cancer with in vivo luciferase imaging. , 2009, Luminescence : the journal of biological and chemical luminescence.

[51]  Michelle Collazo,et al.  Subsets of Myeloid-Derived Suppressor Cells in Tumor-Bearing Mice1 , 2008, The Journal of Immunology.

[52]  H. Yamaue,et al.  Successful cancer vaccine therapy for carcinoembryonic antigen (CEA)‐expressing colon cancer using genetically modified dendritic cells that express CEA and T helper‐type 1 cytokines in CEA transgenic mice , 2007, International journal of cancer.

[53]  N. Kearney,et al.  EORTC guidelines for the use of granulocyte-colony stimulating factor to reduce the incidence of chemotherapy-induced febrile neutropenia in adult patients with lymphomas and solid tumours. , 2006, European journal of cancer.

[54]  J. Simpson,et al.  Mice transgenic for human carcinoembryonic antigen as a model for immunotherapy. , 1998, Cancer research.