201
|
Ustyanovska Avtenyuk N, Visser N, Bremer E, Wiersma VR. The Neutrophil: The Underdog That Packs a Punch in the Fight against Cancer. Int J Mol Sci 2020; 21:E7820. [PMID: 33105656 PMCID: PMC7659937 DOI: 10.3390/ijms21217820] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/19/2020] [Accepted: 10/19/2020] [Indexed: 02/07/2023] Open
Abstract
The advent of immunotherapy has had a major impact on the outcome and overall survival in many types of cancer. Current immunotherapeutic strategies typically aim to (re)activate anticancer T cell immunity, although the targeting of macrophage-mediated anticancer innate immunity has also emerged in recent years. Neutrophils, although comprising ≈ 60% of all white blood cells in the circulation, are still largely overlooked in this respect. Nevertheless, neutrophils have evident anticancer activity and can induce phagocytosis, trogocytosis, as well as the direct cytotoxic elimination of cancer cells. Furthermore, therapeutic tumor-targeting monoclonal antibodies trigger anticancer immune responses through all innate Fc-receptor expressing cells, including neutrophils. Indeed, the depletion of neutrophils strongly reduced the efficacy of monoclonal antibody treatment and increased tumor progression in various preclinical studies. In addition, the infusion of neutrophils in murine cancer models reduced tumor progression. However, evidence on the anticancer effects of neutrophils is fragmentary and mostly obtained in in vitro assays or murine models with reports on anticancer neutrophil activity in humans lagging behind. In this review, we aim to give an overview of the available knowledge of anticancer activity by neutrophils. Furthermore, we will describe strategies being explored for the therapeutic activation of anticancer neutrophil activity.
Collapse
Affiliation(s)
| | | | - Edwin Bremer
- Department of Hematology, Cancer Research Center Groningen, University Medical Center Groningen (UMCG), University of Groningen, Hanzeplein 1/DA13, 9713 GZ Groningen, The Netherlands; (N.U.A.); (N.V.)
| | - Valerie R. Wiersma
- Department of Hematology, Cancer Research Center Groningen, University Medical Center Groningen (UMCG), University of Groningen, Hanzeplein 1/DA13, 9713 GZ Groningen, The Netherlands; (N.U.A.); (N.V.)
| |
Collapse
|
202
|
Wang Z, Yang C, Li L, Zhang Z, Pan J, Su K, Chen W, Li J, Qiu F, Huang J. CD62L dim Neutrophils Specifically Migrate to the Lung and Participate in the Formation of the Pre-Metastatic Niche of Breast Cancer. Front Oncol 2020; 10:540484. [PMID: 33178575 PMCID: PMC7593663 DOI: 10.3389/fonc.2020.540484] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 09/24/2020] [Indexed: 12/14/2022] Open
Abstract
Lung metastasis is one of the leading causes of death in patients with breast cancer. The mechanism of tumor metastasis remains controversial. Recently, the formation of a pre-metastatic niche has been considered a key factor contributing to breast cancer metastasis, which might also explain the tendency of organ metastasis. Our study initially re-examined the critical time of the niche formation and simultaneously detected a novel subset of neutrophils, CD62Ldim neutrophils, which had not previously been reported in tumor metastasis; the number of these cells progressively increased during breast cancer progression and was closely related to the formation of the pre-metastatic niche. Furthermore, we explored the mechanism of their aggregation in the pre-metastatic niche in the lung and found that they were specifically chemoattracted by the CXCL12-CXCR4 signaling pathway. Compared to the CD62Lhi neutrophils, CD62Ldim neutrophils exhibited stronger adhesion and increased survival. The results provide new insights into the subsequent targeted treatment of breast cancer metastasis.
Collapse
Affiliation(s)
- Zhen Wang
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Breast Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chenghui Yang
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Breast Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lili Li
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhigang Zhang
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Pan
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Breast Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ke Su
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Breast Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wuzhen Chen
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Breast Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinfan Li
- Department of Pathology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fuming Qiu
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Huang
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Breast Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
203
|
Zonneville J, Colligan S, Grant S, Miller A, Wallace P, Abrams SI, Bakin AV. Blockade of p38 kinase impedes the mobilization of protumorigenic myeloid populations to impact breast cancer metastasis. Int J Cancer 2020; 147:2279-2292. [PMID: 32452014 PMCID: PMC7484223 DOI: 10.1002/ijc.33050] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/27/2020] [Accepted: 05/04/2020] [Indexed: 12/31/2022]
Abstract
Patients with metastatic breast cancer (MBC) have limited therapeutic options and novel treatments are critically needed. Prior research implicates tumor-induced mobilization of myeloid cell populations in metastatic progression, as well as being an unfavorable outcome in MBC; however, the underlying mechanisms for these relationships remain unknown. Here, we provide evidence for a novel mechanism by which p38 promotes metastasis. Using triple-negative breast cancer models, we showed that a selective inhibitor of p38 (p38i) significantly reduced tumor growth, angiogenesis, and lung metastasis. Importantly, p38i decreased the accumulation of myeloid populations, namely, myeloid-derived suppressor cells (MDSCs) and CD163+ tumor-associated macrophages (TAMs). p38 controlled the expression of tumor-derived chemokines/cytokines that facilitated the recruitment of protumor myeloid populations. Depletion of MDSCs was accompanied by reduced TAM infiltration and phenocopied the antimetastatic effects of p38i. Reciprocally, p38i increased tumor infiltration by cytotoxic CD8+ T cells. Furthermore, the CD163+ /CD8+ expression ratio inversely correlated with metastasis-free survival in breast cancer, suggesting that targeting p38 may improve clinical outcomes. Overall, our study highlights a previously unknown p38-driven pathway as a therapeutic target in MBC.
Collapse
MESH Headings
- Animals
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/metabolism
- Antineoplastic Agents/pharmacology
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/pathology
- Carcinogenesis/drug effects
- Carcinogenesis/metabolism
- Carcinogenesis/pathology
- Cell Line, Tumor
- Chemokines/metabolism
- Cytokines/metabolism
- Female
- Humans
- Lung Neoplasms/drug therapy
- Lung Neoplasms/metabolism
- MAP Kinase Signaling System/drug effects
- Macrophages/drug effects
- Macrophages/metabolism
- Macrophages/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, SCID
- Mice, Transgenic
- Myeloid Cells/drug effects
- Myeloid Cells/metabolism
- Myeloid Cells/pathology
- Myeloid-Derived Suppressor Cells/drug effects
- Myeloid-Derived Suppressor Cells/metabolism
- Myeloid-Derived Suppressor Cells/pathology
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Receptors, Cell Surface/metabolism
- Triple Negative Breast Neoplasms/drug therapy
- Triple Negative Breast Neoplasms/metabolism
- Triple Negative Breast Neoplasms/pathology
Collapse
Affiliation(s)
- Justin Zonneville
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263
| | - Sean Colligan
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263
| | - Sydney Grant
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263
| | | | - Paul Wallace
- Department of Flow & Image Cytometry, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263
| | - Scott I. Abrams
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263
| | - Andrei V. Bakin
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263
- Sechenov Medical University, Moscow, Russia 119991
| |
Collapse
|
204
|
Ishida Y, Kuninaka Y, Yamamoto Y, Nosaka M, Kimura A, Furukawa F, Mukaida N, Kondo T. Pivotal Involvement of the CX3CL1-CX3CR1 Axis for the Recruitment of M2 Tumor-Associated Macrophages in Skin Carcinogenesis. J Invest Dermatol 2020; 140:1951-1961.e6. [PMID: 32179066 DOI: 10.1016/j.jid.2020.02.023] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 02/18/2020] [Accepted: 02/26/2020] [Indexed: 12/19/2022]
Abstract
We previously revealed the crucial roles of a chemokine, CX3CL1, and its receptor, CX3CR1, in skin wound healing. Although repeated wounds frequently develop into skin cancer, the roles of CX3CL1 in skin carcinogenesis remain elusive. Here, we proved that CX3CL1 protein expression and CX3CR1+ macrophages were observed in human skin cancer tissues. Similarly, we observed the enhancement of CX3CL1 expression and the abundant accumulation of CX3CR1+ tumor-associated macrophages with M2-like phenotypes in the skin carcinogenesis process induced by the combined treatment with 7,12-dimethylbenz[a]anthracene and 12-O-tetradecanoylphorbol-13-acetate. In this mouse skin carcinogenesis process, CX3CR1+ tumor-associated macrophages exhibited M2-like phenotypes with the expression of Wnt3a and angiogenic molecules including VEGF and matrix metalloproteinase 9. Compared with wild-type mice, CX3CR1-deficient mice showed fewer numbers of skin tumors with a lower incidence. Concomitantly, M2-macrophage numbers and neovascularization were reduced with the depressed expression of angiogenic factors and Wnt3a. Thus, the CX3CL1-CX3CR1 axis can crucially contribute to skin carcinogenesis by regulating the accumulation and functions of tumor-associated macrophages. Thus, this axis can be a good target for preventing and/or treating skin cancers.
Collapse
Affiliation(s)
- Yuko Ishida
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Yumi Kuninaka
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Yuki Yamamoto
- Department of Dermatology, Wakayama Medical University, Wakayama, Japan
| | - Mizuho Nosaka
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Akihiko Kimura
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Fukumi Furukawa
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Naofumi Mukaida
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Toshikazu Kondo
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan.
| |
Collapse
|
205
|
Castro F, Pinto ML, Pereira CL, Serre K, Barbosa MA, Vermaelen K, Gärtner F, Gonçalves RM, De Wever O, Oliveira MJ. Chitosan/γ-PGA nanoparticles-based immunotherapy as adjuvant to radiotherapy in breast cancer. Biomaterials 2020; 257:120218. [DOI: 10.1016/j.biomaterials.2020.120218] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/05/2020] [Accepted: 06/22/2020] [Indexed: 02/07/2023]
|
206
|
van Elsas M, Kleinovink JW, Moerland M, Feiss G, Beyrend G, Arens R, Mei H, Nibbering PH, Jirka SM, van Hall T, van der Burg SH. Host genetics and tumor environment determine the functional impact of neutrophils in mouse tumor models. J Immunother Cancer 2020; 8:jitc-2020-000877. [PMID: 32998952 PMCID: PMC7528431 DOI: 10.1136/jitc-2020-000877] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2020] [Indexed: 01/06/2023] Open
Abstract
Background Neutrophils have been reported to have protumor, antitumor or neutral effects in cancer progression. The underlying causes for this functional variability are not clear. Methods We studied the role of neutrophils in six different mouse tumor models by intratumoral injection of antimicrobial peptides or vaccination. Changes in systemic and intratumoral immune cells were analyzed by flow-cytometry and mass-cytometry. The role of neutrophils was studied by antibody-mediated neutrophil depletion. Neutrophils from different mouse strains were compared by RNA sequencing. Results The antimicrobial peptide Omiganan reduced the growth of TC-1 tumors in BL/6 mice and CT26 tumors in BALB/c mice. No significant effects were observed in B16F10, MC38 and 4T1 tumors. Growth delay was associated with increased abundance of neutrophils in TC-1 but not CT26 tumors. Systemic neutrophil depletion abrogated Omiganan efficacy in TC-1 but further reduced growth of CT26, indicating that neutrophils were required for the antitumor effect in TC-1 but suppressed tumor control in CT26. Neutrophils were also required for a therapeutic vaccine-induced T-cell mediated control of RMA tumors in BL/6 mice. Clearly, the circulating and intratumoral neutrophils differed in the expression of Ly6G and CD62L, between TC-1 and CT26 and between blood neutrophils of tumor-naïve BL/6 and BALB/c mice. RNA-sequencing revealed that neutrophils from BL/6 mice but not BALB/c mice displayed a robust profile of immune activation, matching their opposing roles in TC-1 and RMA versus CT26. Conclusions Neutrophil functionality differs strongly between mouse strains and tumor types, with consequences for tumor progression and therapy.
Collapse
Affiliation(s)
- Marit van Elsas
- Medical Oncology, Oncode institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Jan Willem Kleinovink
- Medical Oncology, Oncode institute, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Gary Feiss
- Cutanea Life Sciences, Wayne, Pennsylvania, USA
| | - Guillaume Beyrend
- Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Ramon Arens
- Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Hailiang Mei
- Sequencing Analysis Support Core, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter H Nibbering
- Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Thorbald van Hall
- Medical Oncology, Oncode institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Sjoerd H van der Burg
- Medical Oncology, Oncode institute, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
207
|
Long W, Chen J, Gao C, Lin Z, Xie X, Dai H. Brief review on the roles of neutrophils in cancer development. J Leukoc Biol 2020; 109:407-413. [PMID: 32970873 PMCID: PMC7891660 DOI: 10.1002/jlb.4mr0820-011r] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 08/25/2020] [Accepted: 09/01/2020] [Indexed: 12/14/2022] Open
Abstract
Neutrophils, which are traditionally regarded as a hallmark of inflammation, are also a member of the intratumoral immune cells. The roles of neutrophils in cancer development are diverse and undefined. So far, they are known to be involved in tumor initiation and tumor cell proliferation and metastasis. They show heterogeneity in both phenotypes and functions during early versus late stage of cancer development. Because they are also associated with the clinical outcomes of various types of solid tumors, cancer treatments that target neutrophils might be highly effective. In this review, we briefly cover the latest findings on the multiple roles of neutrophils in cancer development and point out the future directions as well.
Collapse
Affiliation(s)
- Wang Long
- Department of Kidney Transplantation, The Second Xiangya Hospital of Central South University, Changsha, China.,Department of Pathological Cell Biology, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University, Tokyo, Japan
| | - Jingjing Chen
- Department of Kidney Transplantation, The Second Xiangya Hospital of Central South University, Changsha, China.,Clinical Research Center for Organ Transplantation in Hunan Province, Changsha, China
| | - Chen Gao
- Department of Kidney Transplantation, The Second Xiangya Hospital of Central South University, Changsha, China.,Clinical Research Center for Organ Transplantation in Hunan Province, Changsha, China
| | - Zhi Lin
- Department of Kidney Transplantation, The Second Xiangya Hospital of Central South University, Changsha, China.,Clinical Research Center for Organ Transplantation in Hunan Province, Changsha, China
| | - Xubiao Xie
- Department of Kidney Transplantation, The Second Xiangya Hospital of Central South University, Changsha, China.,Clinical Research Center for Organ Transplantation in Hunan Province, Changsha, China
| | - Helong Dai
- Department of Kidney Transplantation, The Second Xiangya Hospital of Central South University, Changsha, China.,Clinical Research Center for Organ Transplantation in Hunan Province, Changsha, China.,Clinical Immunology Center, Central South University, Changsha, China
| |
Collapse
|
208
|
Wu M, Ma M, Tan Z, Zheng H, Liu X. Neutrophil: A New Player in Metastatic Cancers. Front Immunol 2020; 11:565165. [PMID: 33101283 PMCID: PMC7546851 DOI: 10.3389/fimmu.2020.565165] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 08/12/2020] [Indexed: 12/11/2022] Open
Abstract
The interaction between cancer cells and immune cells is important for the cancer development. However, much attention has been given to T cells and macrophages. Being the most abundant leukocytes in the blood, the functions of neutrophils in cancer have been underdetermined. They have long been considered an “audience” in the development of cancer. However, emerging evidence indicate that neutrophils are a heterogeneous population with plasticity, and subpopulation of neutrophils (such as low density neutrophils, polymorphonuclear-myeloid-derived suppressor cells) are actively involved in cancer growth and metastasis. Here, we review the current understanding of the role of neutrophils in cancer development, with a specific focus on their pro-metastatic functions. We also discuss the potential and challenges of neutrophils as therapeutic targets. A better understanding the role of neutrophils in cancer will discover new mechanisms of metastasis and develop new immunotherapies by targeting neutrophils.
Collapse
Affiliation(s)
- Mengyue Wu
- Department of Pathophysiology, Anhui Medical University, Hefei, China
| | - Mutian Ma
- Department of Pathophysiology, Anhui Medical University, Hefei, China
| | - Zhenya Tan
- Department of Pathophysiology, Anhui Medical University, Hefei, China
| | - Hong Zheng
- Department of Pathophysiology, Anhui Medical University, Hefei, China
| | - Xia Liu
- Department of Toxicology and Cancer Biology, Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
209
|
Karagiannidis I, Jerman SJ, Jacenik D, Phinney BB, Yao R, Prossnitz ER, Beswick EJ. G-CSF and G-CSFR Modulate CD4 and CD8 T Cell Responses to Promote Colon Tumor Growth and Are Potential Therapeutic Targets. Front Immunol 2020; 11:1885. [PMID: 33042110 PMCID: PMC7522314 DOI: 10.3389/fimmu.2020.01885] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 07/13/2020] [Indexed: 01/01/2023] Open
Abstract
Cytokines are known to shape the tumor microenvironment and although progress has been made in understanding their role in carcinogenesis, much remains to learn regarding their role in tumor growth and progression. We have identified granulocyte colony-stimulating factor (G-CSF) as one such cytokine, showing that G-CSF is linked with metastasis in human gastrointestinal tumors and neutralizing G-CSF in a mouse model of colitis-associated cancer is protective. Here, we set out to identify the role of G-CSF and its receptor, G-CSFR, in CD4+ and CD8+ T cell responses in the tumor microenvironment. MC38 colon cancer cells were injected into WT, G-CSFR-/- mice, or Rag2-/- mice. Flow cytometry, Real Time PCR and Multiplex cytokine array analysis were used for in vitro T cell phenotype analysis. Adoptive transfer of WT or G-CSFR-/- CD4+ of CD8+ T cells were performed. Mouse tumor size, cytokine expression, T cell phenotype, and cytotoxic activity were analyzed. We established that in G-CSFR-/- mice, tumor growth of MC38 colon cancer cells is significantly decreased. T cell phenotype and cytokine production were also altered, as both in vitro and in vivo approaches revealed that the G-CSF/G-CSFR stimulate IL-10-producing, FoxP3-expressing CD4+ and CD8+ T cells, whereas G-CSFR-/- T cells exhibit increased IFNγ and IL-17A production, leading to increased cytotoxic activity in the tumor microenvironment. Furthermore, peritumoral injection of recombinant IFNγ or IL-17A inhibited colon and pancreas tumor growth compared to controls. Taken together, our data reveal an unknown mechanism by which G-CSF, through its receptor G-CSFR, promotes an inhibitory Treg phenotype that limits tumor immune responses and furthermore suggest that targeting this cytokine/receptor axis could represent a novel therapeutic approach for gastrointestinal, and likely other tumors with high expression of these factors.
Collapse
MESH Headings
- Animals
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cell Line, Tumor
- Coculture Techniques
- Colonic Neoplasms/immunology
- Colonic Neoplasms/metabolism
- Colonic Neoplasms/pathology
- Cytotoxicity, Immunologic
- DNA-Binding Proteins/deficiency
- DNA-Binding Proteins/genetics
- Female
- Granulocyte Colony-Stimulating Factor/metabolism
- Interferon-gamma/metabolism
- Interleukin-10/metabolism
- Interleukin-17/metabolism
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Phenotype
- Receptors, Colony-Stimulating Factor/deficiency
- Receptors, Colony-Stimulating Factor/genetics
- Receptors, Colony-Stimulating Factor/metabolism
- Signal Transduction
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Tumor Burden
- Tumor Microenvironment
Collapse
Affiliation(s)
- Ioannis Karagiannidis
- Division of Gastroenterology, Department of Internal Medicine, University of Utah, Salt Lake City, UT, United States
| | - Stephanie J. Jerman
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center Albuquerque, Albuquerque, NM, United States
| | - Damian Jacenik
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Brandon B. Phinney
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center Albuquerque, Albuquerque, NM, United States
| | - Ruoxin Yao
- Division of Gastroenterology, Department of Internal Medicine, University of Utah, Salt Lake City, UT, United States
| | - Eric R. Prossnitz
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Ellen J. Beswick
- Division of Gastroenterology, Department of Internal Medicine, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
210
|
Bakherad H, Farahmand M, Setayesh N, Ebrahim-Habibi A. Engineering an anti-granulocyte colony stimulating factor receptor nanobody for improved affinity. Life Sci 2020; 257:118052. [PMID: 32634431 DOI: 10.1016/j.lfs.2020.118052] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 06/23/2020] [Accepted: 07/01/2020] [Indexed: 12/21/2022]
Abstract
AIMS Granulocyte colony-stimulating factor (G-CSF) is a cytokine that induces proliferation and differentiation of hematopoietic precursor cells and activation of mature neutrophils. G-CSF is overexpressed in several malignant tumors and blocking its binding to the receptor can lead to significant decrease in tumor growth, vascularization and metastasis. Furthermore, targeting G-CSF receptor has shown therapeutic benefit in other diseases such as rheumatoid arthritis, progressive neurodegenerative disorder and uveitis. Camelid single-chain antibodies (nanobodies) have exceptional properties making them appropriate for tumor imaging and therapeutic application. In this study we aim to use the rational design approach to engineer a previously described G-CSF-R targeting nanobody (VHH1), to improve its affinity toward G-CSF-R. MAIN METHODS We redesigned the complementary determining region 3 (CDR3) domain of the VHH1 nanobody to mimic G-CSF interaction to its receptor and developed five new engineered nanobodies. Binding affinity of the engineered nanobodies was evaluated by ELISA (Enzyme-linked immunosorbent assay) on NFS60 cells. KEY FINDINGS Enzyme-linked immunosorbent assay (ELISA) confirmed the specificity of the engineered nanobodies and ELISA-based determination of affinity revealed that two of the engineered nanobodies (1c and 5a) bind to G-CSF-R on the surface of NFS60 cells in a dose-dependent manner and with a higher potency compared to the parental nanobody. SIGNIFICANCE Additional studies are required to better characterize these nanobodies and assess their interaction with G-CSF-R in vitro and in vivo. These newly developed nanobodies could be beneficial in tumor imaging and therapy and make a basis for development of additional engineered nanobodies.
Collapse
Affiliation(s)
- Hamid Bakherad
- Department of Pharmaceutical Biotechnology and Isfahan Pharmaceutical Sciences Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Mohammad Farahmand
- Department of Pharmaceutical Biotechnology and Biotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Neda Setayesh
- Department of Pharmaceutical Biotechnology and Biotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Azadeh Ebrahim-Habibi
- Biosensor Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran; Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
211
|
Morris AH, Orbach SM, Bushnell GG, Oakes RS, Jeruss JS, Shea LD. Engineered Niches to Analyze Mechanisms of Metastasis and Guide Precision Medicine. Cancer Res 2020; 80:3786-3794. [PMID: 32409307 PMCID: PMC7501202 DOI: 10.1158/0008-5472.can-20-0079] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 03/04/2020] [Accepted: 05/11/2020] [Indexed: 12/20/2022]
Abstract
Cancer metastasis poses a challenging problem both clinically and scientifically, as the stochastic nature of metastatic lesion formation introduces complexity for both early detection and the study of metastasis in preclinical models. Engineered metastatic niches represent an emerging approach to address this stochasticity by creating bioengineered sites where cancer can preferentially metastasize. As the engineered niche captures the earliest metastatic cells at a nonvital location, both noninvasive and biopsy-based monitoring of these sites can be performed routinely to detect metastasis early and monitor alterations in the forming metastatic niche. The engineered metastatic niche also provides a new platform technology that serves as a tunable site to molecularly dissect metastatic disease mechanisms. Ultimately, linking the engineered niches with advances in sensor development and synthetic biology can provide enabling tools for preclinical cancer models and fosters the potential to impact the future of clinical cancer care.
Collapse
Affiliation(s)
- Aaron H Morris
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Sophia M Orbach
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Grace G Bushnell
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
- Department of Internal Medicine, Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Robert S Oakes
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| | - Jacqueline S Jeruss
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan.
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan.
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
212
|
Farahi N, Gillett D, Southwood M, Rassl D, Tregay N, Hill U, Preston SD, Loutsios C, Lok LSC, Heard S, Buscombe J, Rintoul RC, Peters AM, Summers C, Chilvers ER. Lesson of the month: novel method to quantify neutrophil uptake in early lung cancer using SPECT-CT. Thorax 2020; 75:1020-1023. [PMID: 32887739 PMCID: PMC7569370 DOI: 10.1136/thoraxjnl-2020-214642] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 06/25/2020] [Accepted: 07/14/2020] [Indexed: 12/22/2022]
Abstract
Neutrophils play an important role in the lung tumour microenvironment. We hypothesised that radiolabelled neutrophils coupled to single-photon emission CT (SPECT) may non-invasively quantify neutrophil uptake in tumours from patients with non-small cell lung cancer. We demonstrated increased uptake of radiolabelled neutrophils from the blood into tumours compared with non-specific uptake using radiolabelled transferrin. Moreover, indium-111-neutrophil activity in the tumour biopsies also correlated with myeloperoxidase (MPO)-positive neutrophils. Our data support the utility of imaging with In-111-labelled neutrophils and SPECT-CT to quantify neutrophil uptake in lung cancer.
Collapse
Affiliation(s)
- Neda Farahi
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Daniel Gillett
- Department of Nuclear Medicine, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Mark Southwood
- Department of Pathology, Royal Papworth Hospital NHS Foundation Trust, Cambridge, UK
| | - Doris Rassl
- Department of Pathology, Royal Papworth Hospital NHS Foundation Trust, Cambridge, UK
| | - Nicola Tregay
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Uta Hill
- Cambridge Centre for Lung Infection, Royal Papworth Hospital NHS Foundation Trust, Cambridge, UK
| | - Stephen Denis Preston
- Department of Pathology, Royal Papworth Hospital NHS Foundation Trust, Cambridge, UK
| | | | | | - Sarah Heard
- Department of Nuclear Medicine, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - John Buscombe
- Department of Nuclear Medicine, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Robert Campbell Rintoul
- Department of Thoracic Oncology, Royal Papworth Hospital NHS Foundation Trust, Cambridge, UK.,Department of Oncology, University of Cambridge, Cambridge, UK
| | - A Michael Peters
- Department of Nuclear Medicine, King's College Hospital NHS Foundation Trust, London, UK
| | | | | |
Collapse
|
213
|
Jaillon S, Ponzetta A, Di Mitri D, Santoni A, Bonecchi R, Mantovani A. Neutrophil diversity and plasticity in tumour progression and therapy. Nat Rev Cancer 2020; 20:485-503. [PMID: 32694624 DOI: 10.1038/s41568-020-0281-y] [Citation(s) in RCA: 720] [Impact Index Per Article: 144.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/04/2020] [Indexed: 12/11/2022]
Abstract
Neutrophils play a key role in defence against infection and in the activation and regulation of innate and adaptive immunity. In cancer, tumour-associated neutrophils (TANs) have emerged as an important component of the tumour microenvironment. Here, they can exert dual functions. TANs can be part of tumour-promoting inflammation by driving angiogenesis, extracellular matrix remodelling, metastasis and immunosuppression. Conversely, neutrophils can also mediate antitumour responses by direct killing of tumour cells and by participating in cellular networks that mediate antitumour resistance. Neutrophil diversity and plasticity underlie the dual potential of TANs in the tumour microenvironment. Myeloid checkpoints as well as the tumour and tissue contexture shape neutrophil function in response to conventional therapies and immunotherapy. We surmise that neutrophils can provide tools to tailor current immunotherapy strategies and pave the way to myeloid cell-centred therapeutic strategies, which would be complementary to current approaches.
Collapse
Affiliation(s)
- Sebastien Jaillon
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy.
- Humanitas Clinical and Research Center IRCCS, Rozzano (MI), Italy.
| | - Andrea Ponzetta
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy
- Humanitas Clinical and Research Center IRCCS, Rozzano (MI), Italy
| | - Diletta Di Mitri
- Humanitas Clinical and Research Center IRCCS, Rozzano (MI), Italy
| | - Angela Santoni
- Dipartimento di Medicina Molecolare Istituto Pasteur-Fondazione Cenci Bolognetti, Università di Roma 'La Sapienza', Rome, Italy
- IRCCS Neuromed, Pozzilli (IS), Italy
| | - Raffaella Bonecchi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy
- Humanitas Clinical and Research Center IRCCS, Rozzano (MI), Italy
| | - Alberto Mantovani
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy.
- Humanitas Clinical and Research Center IRCCS, Rozzano (MI), Italy.
- The William Harvey Research Institute, Queen Mary University of London, London, UK.
| |
Collapse
|
214
|
Li P, Lu M, Shi J, Hua L, Gong Z, Li Q, Shultz LD, Ren G. Dual roles of neutrophils in metastatic colonization are governed by the host NK cell status. Nat Commun 2020; 11:4387. [PMID: 32873795 PMCID: PMC7463263 DOI: 10.1038/s41467-020-18125-0] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 08/04/2020] [Indexed: 12/11/2022] Open
Abstract
The role of neutrophils in solid tumor metastasis remains largely controversial. In preclinical models of solid tumors, both pro-metastatic and anti-metastatic effects of neutrophils have been reported. In this study, using mouse models of breast cancer, we demonstrate that the metastasis-modulating effects of neutrophils are dictated by the status of host natural killer (NK) cells. In NK cell-deficient mice, granulocyte colony-stimulating factor-expanded neutrophils show an inhibitory effect on the metastatic colonization of breast tumor cells in the lung. In contrast, in NK cell-competent mice, neutrophils facilitate metastatic colonization in the same tumor models. In an ex vivo neutrophil-NK cell-tumor cell tri-cell co-culture system, neutrophils are shown to potentially suppress the tumoricidal activity of NK cells, while neutrophils themselves are tumoricidal. Intriguingly, these two modulatory effects by neutrophils are both mediated by reactive oxygen species. Collectively, the absence or presence of NK cells, governs the net tumor-modulatory effects of neutrophils. The role of neutrophils in the regulation of tumour growth and metastasis remains controversial. Here, the authors demonstrate that neutrophils, by exerting inhibitory effects on cytotoxic NK cells, show a net pro-metastatic effect in immune-competent mice, while they are tumoricidal and anti-metastatic in NK cell-deficient hosts.
Collapse
Affiliation(s)
- Peishan Li
- Key Laboratory of Experimental Teratology, Ministry of Education and Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 250012, Jinan, Shandong, China.,The Jackson Laboratory, Bar Harbor, ME, 04609, USA
| | - Ming Lu
- The Jackson Laboratory, Bar Harbor, ME, 04609, USA.,Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, 200040, Shanghai, China
| | - Jiayuan Shi
- The Jackson Laboratory, Bar Harbor, ME, 04609, USA
| | - Li Hua
- The Jackson Laboratory, Bar Harbor, ME, 04609, USA
| | - Zheng Gong
- The Jackson Laboratory, Bar Harbor, ME, 04609, USA
| | - Qing Li
- The Jackson Laboratory, Bar Harbor, ME, 04609, USA
| | | | - Guangwen Ren
- The Jackson Laboratory, Bar Harbor, ME, 04609, USA.
| |
Collapse
|
215
|
Liu L, Wu Y, Zhang C, Zhou C, Li Y, Zeng Y, Zhang C, Li R, Luo D, Wang L, Zhang L, Tu S, Deng H, Luo S, Chen YG, Xiong X, Yan X. Cancer-associated adipocyte-derived G-CSF promotes breast cancer malignancy via Stat3 signaling. J Mol Cell Biol 2020; 12:723-737. [PMID: 32242230 PMCID: PMC7749739 DOI: 10.1093/jmcb/mjaa016] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 02/13/2020] [Accepted: 03/31/2020] [Indexed: 12/25/2022] Open
Abstract
Adipocyte is the most predominant cell type in the tumor microenvironment of breast cancer and plays a pivotal role in cancer progression, yet the underlying mechanisms and functional mediators remain elusive. We isolated primary preadipocytes from mammary fat pads of human breast cancer patients and generated mature adipocytes and cancer-associated adipocytes (CAAs) in vitro. The CAAs exhibited significantly different gene expression profiles as assessed by transcriptome sequencing. One of the highly expressed genes in CAAs is granulocyte colony-stimulating factor (G-CSF). Treatment with recombinant human G-CSF protein or stable expression of human G-CSF in triple-negative breast cancer (TNBC) cell lines enhanced epithelial-mesenchymal transition, migration, and invasion of cancer cells, by activating Stat3. Accordantly, targeting G-CSF/Stat3 signaling with G-CSF-neutralizing antibody, a chemical inhibitor, or siRNAs for Stat3 could all abrogate CAA- or G-CSF-induced migration and invasion of breast cancer cells. The pro-invasive genes MMP2 and MMP9 were identified as target genes of G-CSF in TNBC cells. Furthermore, in human breast cancer tissues, elevated G-CSF expression in adipocytes is well correlated with activated Stat3 signal in cancer cells. Together, our results suggest a novel strategy to intervene with invasive breast cancers by targeting CAA-derived G-CSF.
Collapse
Affiliation(s)
- Li Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
| | - Yudong Wu
- Department of Breast Surgery, Jiangxi Provincial Cancer Hospital, Nanchang 330029, China
| | - Cheng Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
| | - Chong Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
| | - Yining Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
| | - Yi Zeng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
| | - Chunbo Zhang
- School of Pharmacy, Nanchang University, Nanchang 330006, China
| | - Rong Li
- School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
| | - Daya Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
| | - Lieliang Wang
- Department of Breast Surgery, Jiangxi Provincial Cancer Hospital, Nanchang 330029, China
| | - Long Zhang
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China
| | - Shuo Tu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
| | - Huan Deng
- Department of Pathology, The Fourth Affiliated Hospital of Nanchang University, Nanchang 330003, China
| | - Shiwen Luo
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Ye-Guang Chen
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xiangyang Xiong
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
| | - Xiaohua Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
- Institute of Biomedical Sciences, Nanchang University Medical College, Nanchang 330031, China
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology, Nanchang University Medical College, Nanchang 330006, China
| |
Collapse
|
216
|
Flerin NC, Pinioti S, Menga A, Castegna A, Mazzone M. Impact of Immunometabolism on Cancer Metastasis: A Focus on T Cells and Macrophages. Cold Spring Harb Perspect Med 2020; 10:a037044. [PMID: 31615868 PMCID: PMC7461771 DOI: 10.1101/cshperspect.a037044] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Despite improved treatment options, cancer remains the leading cause of morbidity and mortality worldwide, with 90% of this mortality correlated to the development of metastasis. Since metastasis has such an impact on treatment success, disease outcome, and global health, it is important to understand the different steps and factors playing key roles in this process, how these factors relate to immune cell function and how we can target metabolic processes at different steps of metastasis in order to improve cancer treatment and patient prognosis. Recent insights in immunometabolism direct to promising therapeutic targets for cancer treatment, however, the specific contribution of metabolism on antitumor immunity in different metastatic niches warrant further investigation. Here, we provide an overview of what is so far known in the field of immunometabolism at different steps of the metastatic cascade, and what may represent the next steps forward. Focusing on metabolic checkpoints in order to translate these findings from in vitro and mouse studies to the clinic has the potential to revolutionize cancer immunotherapy and greatly improve patient prognosis.
Collapse
Affiliation(s)
- Nina C Flerin
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven B3000, Belgium
- Department of Oncology, Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, KU Leuven, Leuven B3000, Belgium
| | - Sotiria Pinioti
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven B3000, Belgium
- Department of Oncology, Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, KU Leuven, Leuven B3000, Belgium
| | - Alessio Menga
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven B3000, Belgium
- Department of Oncology, Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, KU Leuven, Leuven B3000, Belgium
| | - Alessandra Castegna
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari 70125, Italy
- IBIOM-CNR, Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council, Italy
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven B3000, Belgium
- Department of Oncology, Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, KU Leuven, Leuven B3000, Belgium
| |
Collapse
|
217
|
Suppressing neutrophil-dependent angiogenesis abrogates resistance to anti-VEGF antibody in a genetic model of colorectal cancer. Proc Natl Acad Sci U S A 2020; 117:21598-21608. [PMID: 32817421 PMCID: PMC7474657 DOI: 10.1073/pnas.2008112117] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Using mouse models that recapitulate key genetic abnormalities accumulating during colorectal cancer (CRC) tumorigenesis, we report that chemically induced colitis promoted development of colon tumors that were largely resistant to anti-VEGF antibody treatment. Serum G-CSF levels were markedly elevated after induction of colitis. Inhibition of G-CSF or Bv8/PROK2 increased the efficacy of anti-VEGF antibody and prevented onset of resistance. To verify the potential clinical relevance of these findings, we examined a series of CRC specimens and found that tumor-infiltrating neutrophils strongly expressed Bv8/PROK2. CRC patients had significantly higher plasma Bv8/PROK2 levels than healthy volunteers and high plasma Bv8/PROK2 levels were inversely correlated with overall survival. These findings establish Bv8/PROK2 as a translational target in CRC, in combination with anti-VEGF agents. We tested cis-ApcΔ716/Smad4+/− and cis-ApcΔ716/Smad4+/−KrasG12D mice, which recapitulate key genetic abnormalities accumulating during colorectal cancer (CRC) tumorigenesis in humans, for responsiveness to anti-VEGF therapy. We found that even tumors in cis-ApcΔ716/Smad4+/−KrasG12D mice, although highly aggressive, were suppressed by anti-VEGF treatment. We tested the hypothesis that inflammation, a major risk factor and trigger for CRC, may affect responsiveness to anti-VEGF. Chemically induced colitis (CIC) in cis-ApcΔ716/Smad4+/− and cis-ApcΔ716/Smad4+/−KrasG12D mice promoted development of colon tumors that were largely resistant to anti-VEGF treatment. The myeloid growth factor G-CSF was markedly increased in the serum after induction of colitis. Antibodies blocking G-CSF, or its target Bv8/PROK2, suppressed tumor progression and myeloid cell infiltration when combined with anti-VEGF in CIC-associated CRC and in anti-VEGF-resistant CRC liver metastasis models. In a series of CRC specimens, tumor-infiltrating neutrophils strongly expressed Bv8/PROK2. CRC patients had significantly higher plasma Bv8/PROK2 levels than healthy volunteers and high plasma Bv8/PROK2 levels were inversely correlated with overall survival. Our findings establish Bv8/PROK2 as a translational target in CRC, in combination with anti-VEGF agents.
Collapse
|
218
|
Li S, Sun Y, Huang J, Wang B, Gong Y, Fang Y, Liu Y, Wang S, Guo Y, Wang H, Xu Z, Guo Y. Anti-tumor effects and mechanisms of Astragalus membranaceus (AM) and its specific immunopotentiation: Status and prospect. JOURNAL OF ETHNOPHARMACOLOGY 2020; 258:112797. [PMID: 32243990 DOI: 10.1016/j.jep.2020.112797] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/10/2020] [Accepted: 03/23/2020] [Indexed: 06/11/2023]
Abstract
With cancer deaths increasing, the initiation, pathophysiology and curative management of cancer is receiving increasing attention. Traditional therapies such as surgery and chemoradiotherapy are often accompanied by suppression of host immunity, which increase the risk of metastasis. Astragalus membranceus (AM) is commonly utilized as one herbal medicine of traditional Chinese medicines (TCMs) with a variety of biological activities. Studies have shown that the active ingredients of AM and AM-based TCMs, combined with chemotherapy, can enhance anti-tumor efficacy in cancer patients, in addition to reduce complications and avoid side effects induced by chemotherapy. By using various cancer models and cell lines, AM has been found to be capable of shrinking or stabilizing tumors by direct anti-proliferation or pro-apoptosis effect on tumor cells. Further, AM ameliorates immunosuppression by activating M1 macrophages and T cells tumor-kill function in tumor microenvironment (TME). AM is also found to improve systemic immunity which may help promoting efficacy of chemotherapy and preventing metastasis. Thereby this review contributes to an understanding of AM as an adjunctive therapy in the whole course of cancer treatment, at the same time providing useful information for development of more effective anti-tumor medication. The combination of AM and immune checkpoint therapies has a promising therapeutic prospect, and the observation of direct efficacy and mechanisms on tumor growth and metastasis of AM combined with chemotherapies or other therapies require more in vivo validations and further clinical investigation as well.
Collapse
Affiliation(s)
- Shanshan Li
- Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yi Sun
- Nephropathy and Rheumatology Department, Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300250, China
| | - Jin Huang
- Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Bin Wang
- Tianjin Medical University Cancer Institute of Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Yinan Gong
- Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yuxin Fang
- Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Acu-moxibustion and Tuina Department, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yangyang Liu
- Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Acu-moxibustion and Tuina Department, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Shenjun Wang
- Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Acu-moxibustion and Tuina Department, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yi Guo
- Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Hong Wang
- Acu-moxibustion and Tuina Department, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Zhifang Xu
- Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Acu-moxibustion and Tuina Department, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Yongming Guo
- Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Acu-moxibustion and Tuina Department, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
219
|
Myeloid-derived suppressor cell depletion therapy targets IL-17A-expressing mammary carcinomas. Sci Rep 2020; 10:13343. [PMID: 32770025 PMCID: PMC7414122 DOI: 10.1038/s41598-020-70231-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 07/20/2020] [Indexed: 12/11/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an invasive subtype of breast cancer but paradoxically associated with increased tumor-infiltrating leukocytes. The molecular and cellular mechanisms underlying TNBC immunobiology are incompletely understood. Interleukin (IL)-17A is a pro-inflammatory cytokine that has both pro- and anti-tumor effects and found in 40-80% of TNBC samples. We report here that IL-17A mRNA and protein are detectable in some human TNBC cell lines and further upregulated by IL-23 and LPS stimulation. Furthermore, the impact of tumor-derived IL-17A in host immune response and tumor growth was examined using murine TNBC 4T1 mammary carcinoma cells transduced with an adenoviral vector expressing IL-17A (AdIL-17A) or control vector (Addl). Compared to Addl-transduction, AdIL-17A-transduction enhanced 4T1 tumor growth and lung metastasis in vivo, which was associated with a marked expansion of myeloid-derived suppressor cells (MDSCs). However, AdIL-17A-transduction also induced strong organ-specific and time-dependent immune activation indicated by dynamic changes of NK cells, B cells, CD4, and CD8 T cells in peripheral blood, lung, and tumor site, as well as the plasma levels of IFNγ. Such findings highlight that tumor-associated IL-17A induces concurrent immune activation and immune suppression. Administration of anti-Gr1 or anti-G-CSF antibody effectively depleted MDSCs in vivo, markedly reducing the growth of AdIL-17A-transduced 4T1 tumors, and eliminating lung metastasis. Collectively, our study demonstrates that MDSC depletion is an effective and practical approach for treating IL-17A-enriched mammary carcinomas.
Collapse
|
220
|
Bakherad H, Setayesh N, Mousavi Gargari SL, Ebrahimizadeh W, Mavandadnejad F, Faghfuri E, Ebrahimi S, Heiat M, Shahpari M, Sepehrizadeh Z. Expression of recombinant G-CSF receptor domains and their inhibitory role on G-CSF function. Res Pharm Sci 2020; 15:381-389. [PMID: 33312216 PMCID: PMC7714017 DOI: 10.4103/1735-5362.293516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 03/28/2020] [Accepted: 08/18/2020] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND AND PURPOSE Granulocyte colony-stimulating factor (G-CSF) is routinely used in combination with chemotherapy to battle neutropenia. However, studies suggest that this chemokine may increase the risk of metastasis and malignancy in many cancers. To counteract the adverse effects of G-CSF in cancer, antibodies have been used to block its action. However, antibodies are large and complex molecules which makes their production expensive. Thus in this study, we aim to construct different structure variants of the G-CSF receptor containing different domains and select the best variant that prevents the adverse actions of this chemokine. These novel structures are smaller than antibodies and easier to produce. EXPERIMENTAL APPROACH Different domains of the G-CSF receptor were designed and cloned into the pET28a expression vector. These recombinant receptor subunits were then expressed in Escherichia coli and purified using standard affinity chromatography techniques. Interaction of recombinant receptor subunits with G-CSF was assessed using enzyme-linked immunosorbent assay and NFS60 cells. FINDINGS / RESULTS Two recombinant receptor subunits containing D1 + D2 + D3 domains and D2 domain showed the strongest inhibitory activity to G-CSF. CONCLUSION AND IMPLICATIONS These novel recombinant receptor variants could be candidates for further studies in the development of novel therapeutics.
Collapse
Affiliation(s)
- Hamid Bakherad
- Department of Pharmaceutical Biotechnology and Isfahan Pharmaceutical Sciences Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Neda Setayesh
- Department of Pharmaceutical Biotechnology and Pharmaceutical Biotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, I.R. Iran
| | | | - Walead Ebrahimizadeh
- Department of Surgery, Division of Urology, McGill University and the Research Institute of the McGill University Health Centre (RI MUHC), Montreal, Quebec, Canada
| | - Faranak Mavandadnejad
- Department of Pharmaceutical Biotechnology and Pharmaceutical Biotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, I.R. Iran
| | - Elnaz Faghfuri
- Digestive Disease Research Center, Ardabil University of Medical Sciences, Ardabil, I.R. Iran
| | - Soheila Ebrahimi
- Department of Pharmaceutical Biotechnology and Pharmaceutical Biotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, I.R. Iran
| | - Mohammad Heiat
- Baqiyatallah Research Center for Gastroenterology and Liver Diseases, Baqiyatallah University of Medical Sciences, Tehran, I.R. Iran
| | - Mona Shahpari
- Department of Pharmaceutical Biotechnology and Pharmaceutical Biotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, I.R. Iran
| | - Zargham Sepehrizadeh
- Department of Pharmaceutical Biotechnology and Pharmaceutical Biotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, I.R. Iran
| |
Collapse
|
221
|
Hsu BE, Tabariès S, Johnson RM, Andrzejewski S, Senecal J, Lehuédé C, Annis MG, Ma EH, Völs S, Ramsay L, Froment R, Monast A, Watson IR, Granot Z, Jones RG, St-Pierre J, Siegel PM. Immature Low-Density Neutrophils Exhibit Metabolic Flexibility that Facilitates Breast Cancer Liver Metastasis. Cell Rep 2020; 27:3902-3915.e6. [PMID: 31242422 DOI: 10.1016/j.celrep.2019.05.091] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 02/13/2019] [Accepted: 05/22/2019] [Indexed: 02/07/2023] Open
Abstract
Neutrophils are phenotypically heterogeneous and exert either anti- or pro-metastatic functions. We show that cancer-cell-derived G-CSF is necessary, but not sufficient, to mobilize immature low-density neutrophils (iLDNs) that promote liver metastasis. In contrast, mature high-density neutrophils inhibit the formation of liver metastases. Transcriptomic and metabolomic analyses of high- and low-density neutrophils reveal engagement of numerous metabolic pathways specifically in low-density neutrophils. iLDNs exhibit enhanced global bioenergetic capacity, through their ability to engage mitochondrial-dependent ATP production, and remain capable of executing pro-metastatic neutrophil functions, including NETosis, under nutrient-deprived conditions. We demonstrate that NETosis is an important neutrophil function that promotes breast cancer liver metastasis. iLDNs rely on the catabolism of glutamate and proline to support mitochondrial-dependent metabolism in the absence of glucose, which enables sustained NETosis. These data reveal that distinct pro-metastatic neutrophil populations exhibit a high degree of metabolic flexibility, which facilitates the formation of liver metastases.
Collapse
Affiliation(s)
- Brian E Hsu
- Goodman Cancer Research Centre, McGill University, Montreal, Québec, QC H3A 1A3, Canada; Department of Medicine, McGill University, Montreal, Québec, QC H3G 1Y6, Canada
| | - Sébastien Tabariès
- Goodman Cancer Research Centre, McGill University, Montreal, Québec, QC H3A 1A3, Canada
| | | | - Sylvia Andrzejewski
- Goodman Cancer Research Centre, McGill University, Montreal, Québec, QC H3A 1A3, Canada
| | - Julien Senecal
- Goodman Cancer Research Centre, McGill University, Montreal, Québec, QC H3A 1A3, Canada; Department of Medicine, McGill University, Montreal, Québec, QC H3G 1Y6, Canada
| | - Camille Lehuédé
- Goodman Cancer Research Centre, McGill University, Montreal, Québec, QC H3A 1A3, Canada
| | - Matthew G Annis
- Goodman Cancer Research Centre, McGill University, Montreal, Québec, QC H3A 1A3, Canada
| | - Eric H Ma
- Goodman Cancer Research Centre, McGill University, Montreal, Québec, QC H3A 1A3, Canada; Department of Physiology, McGill University, Montreal, Québec, QC H3G 1Y6, Canada
| | - Sandra Völs
- Department of Developmental Biology and Cancer Research, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - LeeAnn Ramsay
- Goodman Cancer Research Centre, McGill University, Montreal, Québec, QC H3A 1A3, Canada
| | - Remi Froment
- Department of Pathology and Microbiology, Université de Montréal, Saint Hyacinth, Québec, QC J2S 2M2, Canada
| | - Anie Monast
- Goodman Cancer Research Centre, McGill University, Montreal, Québec, QC H3A 1A3, Canada
| | - Ian R Watson
- Goodman Cancer Research Centre, McGill University, Montreal, Québec, QC H3A 1A3, Canada
| | - Zvi Granot
- Department of Developmental Biology and Cancer Research, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Russell G Jones
- Goodman Cancer Research Centre, McGill University, Montreal, Québec, QC H3A 1A3, Canada; Department of Physiology, McGill University, Montreal, Québec, QC H3G 1Y6, Canada
| | - Julie St-Pierre
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Peter M Siegel
- Goodman Cancer Research Centre, McGill University, Montreal, Québec, QC H3A 1A3, Canada; Department of Medicine, McGill University, Montreal, Québec, QC H3G 1Y6, Canada.
| |
Collapse
|
222
|
Li Y, Wang D, Li X. The blood cells in NSCLC and the changes after RFA. Int J Hyperthermia 2020; 37:753-762. [PMID: 32619369 DOI: 10.1080/02656736.2020.1782486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Lung cancer has attracted a lot of attention because of its high morbidity and mortality. The emergence of RFA provides a new treatment for unresectable NSCLC patients. In addition to killing in situ lung tumors, RFA also provides new immuno-activated antigens, for the treatment of lung cancer. It changes the tumor microenvironment and activates the entire immune system of patients. The peripheral blood cell count is easy to achieve and the blood cells are important in tumor immunity, which changes after RFA. On the one hand, the changes in blood cells identify the immune changes of NSCLC; on the other hand, it provides support and suspicion for the treatment of RFA.
Collapse
Affiliation(s)
- Yunfang Li
- Department of Minimally Invasive Tumor Therapies Center, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, P.R. China.,Graduate School of Perking Union Medical College, China Academy of Medical Sciences, Beijing, China
| | - Dongdong Wang
- Minimally Invasive Interventional Therapy Center Department, Qingdao Municipal Hospital, Qingdao, China
| | - Xiaoguang Li
- Department of Minimally Invasive Tumor Therapies Center, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, P.R. China.,Graduate School of Perking Union Medical College, China Academy of Medical Sciences, Beijing, China
| |
Collapse
|
223
|
Liu K, Sun E, Wang L, Nian X, Ma C. Gene Expression in Human Polymorphonuclear Neutrophils (PMNs) Stimulated by Bacillus Calmette-Guérin (BCG). Inflammation 2020; 43:2098-2108. [PMID: 32578048 DOI: 10.1007/s10753-020-01277-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Neutrophils are the most abundant leukocytes in the blood. Moreover, neutrophils form the first line of host immune defense against bacterial and fungal invasion, and also play an important part in inflammatory and immune system responses. Intravesical bacillus Calmette-Guérin (BCG) has been shown to reduce and delay tumor progression to muscle-invasive disease after transurethral resection of bladder tumors (TRUBTs). Following intravesical BCG, neutrophils gather around tissues infected by BCG in the early stage of inflammatory and immune responses. In our previous study, we reported that BCG induced the formation of neutrophil extracellular traps (NETs), which play an important role in tumor treatment. Therefore, in the present study, we analyzed the gene expression profile of neutrophils stimulated by BCG through high-throughput arrays, which helped us determine the potential roles of neutrophils in BCG immunotherapy. The results showed that the expression of neutrophil genes led to changes in the early stage of BCG stimulation. The changed genes were involved in many functions of neutrophils such as mobility, proliferation, and secretion of cytokines, chemokines, and adhesion molecules. These changes in neutrophil biological functions may play an essential role in BCG induction of inflammatory and immune responses, and in anti-tumor processes.
Collapse
Affiliation(s)
- Kangkang Liu
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, No. 23, PingJiang Road, Hexi District, Tianjin, 300211, People's Republic of China
| | - Erlin Sun
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, No. 23, PingJiang Road, Hexi District, Tianjin, 300211, People's Republic of China.
| | - Lining Wang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, No. 23, PingJiang Road, Hexi District, Tianjin, 300211, People's Republic of China
| | - Xuewu Nian
- Department of Urology, Tianjin Nankai hospital, Tianjin, 300100, People's Republic of China
| | - Chengquan Ma
- Department of Urology, Tianjin Nankai hospital, Tianjin, 300100, People's Republic of China
| |
Collapse
|
224
|
Tumor microenvironment and epithelial mesenchymal transition as targets to overcome tumor multidrug resistance. Drug Resist Updat 2020; 53:100715. [PMID: 32679188 DOI: 10.1016/j.drup.2020.100715] [Citation(s) in RCA: 332] [Impact Index Per Article: 66.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 05/29/2020] [Accepted: 06/07/2020] [Indexed: 12/11/2022]
Abstract
It is well established that multifactorial drug resistance hinders successful cancer treatment. Tumor cell interactions with the tumor microenvironment (TME) are crucial in epithelial-mesenchymal transition (EMT) and multidrug resistance (MDR). TME-induced factors secreted by cancer cells and cancer-associated fibroblasts (CAFs) create an inflammatory microenvironment by recruiting immune cells. CD11b+/Gr-1+ myeloid-derived suppressor cells (MDSCs) and inflammatory tumor associated macrophages (TAMs) are main immune cell types which further enhance chronic inflammation. Chronic inflammation nurtures tumor-initiating/cancer stem-like cells (CSCs), induces both EMT and MDR leading to tumor relapses. Pro-thrombotic microenvironment created by inflammatory cytokines and chemokines from TAMs, MDSCs and CAFs is also involved in EMT and MDR. MDSCs are the most common mediators of immunosuppression and are also involved in resistance to targeted therapies, e.g. BRAF inhibitors and oncolytic viruses-based therapies. Expansion of both cancer and stroma cells causes hypoxia by hypoxia-inducible transcription factors (e.g. HIF-1α) resulting in drug resistance. TME factors induce the expression of transcriptional EMT factors, MDR and metabolic adaptation of cancer cells. Promoters of several ATP-binding cassette (ABC) transporter genes contain binding sites for canonical EMT transcription factors, e.g. ZEB, TWIST and SNAIL. Changes in glycolysis, oxidative phosphorylation and autophagy during EMT also promote MDR. Conclusively, EMT signaling simultaneously increases MDR. Owing to the multifactorial nature of MDR, targeting one mechanism seems to be non-sufficient to overcome resistance. Targeting inflammatory processes by immune modulatory compounds such as mTOR inhibitors, demethylating agents, low-dosed histone deacetylase inhibitors may decrease MDR. Targeting EMT and metabolic adaptation by small molecular inhibitors might also reverse MDR. In this review, we summarize evidence for TME components as causative factors of EMT and anticancer drug resistance.
Collapse
|
225
|
Nguyen TT, Wolter WR, Anderson B, Schroeder VA, Gao M, Gooyit M, Suckow MA, Chang M. Limitations of Knockout Mice and Other Tools in Assessment of the Involvement of Matrix Metalloproteinases in Wound Healing and the Means to Overcome Them. ACS Pharmacol Transl Sci 2020; 3:489-495. [PMID: 32566914 DOI: 10.1021/acsptsci.9b00109] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Indexed: 01/22/2023]
Abstract
Matrix metalloproteinases (MMPs) play important roles in wound healing, but attribution of their functions in repair of wounds has been challenging. Commonly used tools such as MMP-knockout mice and zymography often confound analysis, which is complicated further as these enzymes exist in three distinct forms with only one being catalytically competent. With the use of topical exogenously administered recombinant MMP-8 and MMP-13 to diabetic and nondiabetic mouse wounds, we show that these proteinases facilitate wound repair by upregulating IL-6 and increasing neutrophil trafficking with an early onset of inflammation. Furthermore, by spatiotemporal control in the use of a selective MMP-2 inhibitor, along with immunoprecipitation and Western blotting, we provide definitive demonstration that MMP-2 does not affect wound healing, contrary to reports. MMP-2 is found in wounds complexed with TIMPs, which is catalytically incompetent.
Collapse
Affiliation(s)
- Trung T Nguyen
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - William R Wolter
- Freimann Life Sciences Center and Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Bowen Anderson
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Valerie A Schroeder
- Freimann Life Sciences Center and Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Ming Gao
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Major Gooyit
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Mark A Suckow
- Freimann Life Sciences Center and Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Mayland Chang
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| |
Collapse
|
226
|
Gruber-Bzura BM. Anticancer activity of vitamin D – molecular mechanisms. POSTEP HIG MED DOSW 2020. [DOI: 10.5604/01.3001.0014.1882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
A large number of studies have pointed to the relations between blood levels of 25-hydroxy vitamin D with cancer incidence and survival. The phenomenon of the multidirectional activity of vitamin D is possibly due to the presence of VDR in most nonskeletal human cells, including cancer cells. A wide range of the genes regulated by VDR are related with cell proliferation, apoptosis, differentiation, angiogenesis and metastasis. In some preclinical studies, colon, lung and BC have all demonstrated downregulation of VDR expression as compared to normal cells, and well-differentiated tumors have shown more VDR expression when compared to their poorly differentiated counterparts. Generally, higher tumor VDR expression has been noted as correlating with better prognosis in cancer patients. However, vitamin D pathway genetic polymorphisms also may influence cancer risk. VDR polymorphisms have received the most attention, but this influence has also been observed in genes related to vitamin D metabolism or signalling, such as: CYP27B1, CYP24A1, VDBP or RXRA. Even though the associations between most of them and cancers were not significant, some studies show that VDR polymorphisms may be a better or poor prognostic factor to assess the risk of cancer. The aim of this paper was to present the molecular pathways affected by vitamin D, which are included in carcinogenesis. The literature survey comprised of research compiled from mostly the last five years and it proves vitamin D as the most phenomenal among other vitamins.
Collapse
Affiliation(s)
- Beata M. Gruber-Bzura
- Department of Biochemistry and Biopharmaceuticals, National Medicines Institute, Warsaw, Poland
| |
Collapse
|
227
|
|
228
|
Armbrecht L, Rutschmann O, Szczerba BM, Nikoloff J, Aceto N, Dittrich PS. Quantification of Protein Secretion from Circulating Tumor Cells in Microfluidic Chambers. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1903237. [PMID: 32537399 PMCID: PMC7284199 DOI: 10.1002/advs.201903237] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 02/28/2020] [Accepted: 03/11/2020] [Indexed: 05/05/2023]
Abstract
Cancer cells can be released from a cancerous lesion and migrate into the circulatory system, from whereon they may form metastases at distant sites. Today, it is possible to infer cancer progression and treatment efficacy by determining the number of circulating tumor cells (CTCs) in the patient's blood at multiple time points; further valuable information about CTC phenotypes remains inaccessible. In this article, a microfluidic method for integrated capture, isolation, and analysis of membrane markers as well as quantification of proteins secreted by single CTCs and CTC clusters is introduced. CTCs are isolated from whole blood with extraordinary efficiencies above 95% using dedicated trapping structures that allow co-capture of functionalized magnetic beads to assess protein secretion. The patform is tested with multiple breast cancer cell lines spiked into human blood and mouse-model-derived CTCs. In addition to immunostaining, the secretion level of granulocyte growth stimulating factor (G-CSF), which is shown to be involved in neutrophil recruitment, is quantified The bead-based assay provides a limit of detection of 1.5 ng mL-1 or less than 3700 molecules per cell. Employing barcoded magnetic beads, this platform can be adapted for multiplexed analysis and can enable comprehensive functional CTC profiling in the future.
Collapse
Affiliation(s)
- Lucas Armbrecht
- Department for Biosystems Science and EngineeringBioanalytics GroupETH ZurichMattenstrasse 26BaselCH‐4058Switzerland
| | - Ophélie Rutschmann
- Department for Biosystems Science and EngineeringBioanalytics GroupETH ZurichMattenstrasse 26BaselCH‐4058Switzerland
| | - Barbara Maria Szczerba
- Department of BiomedicineCancer Metastasis LabUniversity of Basel and University Hospital BaselMattenstrasse 28BaselCH‐4058Switzerland
| | - Jonas Nikoloff
- Department for Biosystems Science and EngineeringBioanalytics GroupETH ZurichMattenstrasse 26BaselCH‐4058Switzerland
| | - Nicola Aceto
- Department of BiomedicineCancer Metastasis LabUniversity of Basel and University Hospital BaselMattenstrasse 28BaselCH‐4058Switzerland
| | - Petra S. Dittrich
- Department for Biosystems Science and EngineeringBioanalytics GroupETH ZurichMattenstrasse 26BaselCH‐4058Switzerland
| |
Collapse
|
229
|
Mukaida N, Sasaki SI, Baba T. Two-Faced Roles of Tumor-Associated Neutrophils in Cancer Development and Progression. Int J Mol Sci 2020; 21:3457. [PMID: 32422991 PMCID: PMC7278934 DOI: 10.3390/ijms21103457] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/11/2020] [Accepted: 05/13/2020] [Indexed: 02/07/2023] Open
Abstract
Neutrophils are the most abundant circulating leukocytes in humans. Neutrophil infiltration into tumor tissues has long been observed but its roles have been ignored due to the presumed short life cycle and metabolic incompetence of neutrophils. Recent advances in neutrophil biology research have revealed that neutrophils have a longer life cycle with a potential to express various bioactive molecules. Clinical studies have simultaneously unraveled an increase in the neutrophil-lymphocyte ratio (NLR), a ratio of absolute neutrophil to absolute lymphocyte numbers in cancer patient peripheral blood and an association of higher NLR with more advanced or aggressive disease. As a consequence, tumor-associated neutrophils (TANs) have emerged as important players in tumor microenvironment. The elucidation of the roles of TANs, however, has been hampered by their multitude of plasticity in terms of phenotypes and functionality. Difficulties are further enhanced by the presence of a related cell population-polymorphonuclear leukocyte (PMN)-myeloid-derived suppressor cells (MDSCs)-and various dissimilar aspects of neutrophil biology between humans and mice. Here, we discuss TAN biology in various tumorigenesis processes, and particularly focus on the context-dependent functional heterogeneity of TANs.
Collapse
Affiliation(s)
- Naofumi Mukaida
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan; (S.-i.S.); (T.B.)
| | | | | |
Collapse
|
230
|
Abstract
Tumours reprogram host physiology, metabolism and immune responses during cancer progression. The release of soluble factors, exosomes and metabolites from tumours leads to systemic changes in distant organs, where cancer cells metastasize and grow. These tumour-derived circulating factors also profoundly impact tissues that are rarely inhabited by metastatic cancer cells such as skeletal muscle and adipose tissue. In fact, the majority of patients with metastatic cancer develop a debilitating muscle-wasting syndrome, known as cachexia, that is associated with decreased tolerance to antineoplastic therapy, poor prognosis and accelerated death, with no approved treatments. In this Perspective, we discuss the development of cachexia in the context of metastatic progression. We briefly discuss how circulating factors either directly or indirectly promote cachexia development and examine how signals from the metastatic process can trigger and amplify this process. Finally, we highlight promising therapeutic opportunities for targeting cachexia in the context of metastatic cancers.
Collapse
Affiliation(s)
- Anup K Biswas
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Swarnali Acharyya
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, Columbia University, New York, NY, USA.
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA.
- Herbert Irving Comprehensive Cancer Center, New York, NY, USA.
| |
Collapse
|
231
|
Inflammation-Based Scores Increase the Prognostic Value of Circulating Tumor Cells in Primary Breast Cancer. Cancers (Basel) 2020; 12:cancers12051134. [PMID: 32369910 PMCID: PMC7281016 DOI: 10.3390/cancers12051134] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/16/2020] [Accepted: 04/29/2020] [Indexed: 12/26/2022] Open
Abstract
A correlation between circulating tumor cells (CTCs) and monocytes in metastatic breast cancer (BC), where CTCs and monocyte-to-lymphocyte ratio (MLR) were predictors of overall survival (OS), was recently shown. Herein, we aimed to assess the association between CTCs and the complete blood count (CBC)-derived inflammation-based scores in 284 primary BC patients. CTCs were determined in CD45-depleted peripheral blood mononuclear cells by real time-PCR. This method allowed us to detect a subset of CTCs with an epithelial-to-mesenchymal transition phenotype (CTC EMT), previously associated with inferior outcomes in primary BC. In the present study, CTC EMT positivity (hazard ratio (HR) = 2.4; 95% CI 1.20–4.66, p = 0.013) and elevated neutrophil-to-lymphocyte ratio (NLR) (HR = 2.20; 95% CI 1.07–4.55; p = 0.033) were associated with shorter progression-free survival (PFS) in primary BC patients. Multivariate analysis showed that CTC EMT-positive patients with NLR ≥ 3 had 8.6 times increased risk of disease recurrence (95% CI 2.35–31.48, p = 0.001) compared with CTC EMT-negative patients with NLR < 3. Similarly, disease recurrence was 13.14 times more likely in CTC EMT-positive patients with MLR ≥ 0.34 (95% CI 4.35–39.67, p < 0.001). Given its low methodological and financial demands, the CBC-derived inflammation-based score determination could, after broader validation, significantly improve the prognostication of BC patients.
Collapse
|
232
|
Oliver AJ, Darcy PK, Trapani JA, Kershaw MH, Slaney CY. Cross-talk between tumors at anatomically distinct sites. FEBS J 2020; 288:81-90. [PMID: 32248616 DOI: 10.1111/febs.15316] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/14/2020] [Accepted: 03/27/2020] [Indexed: 12/15/2022]
Abstract
Cancer tissue is not homogenous, and individual metastases at different anatomical locations can differ from the primary tumor and from one another in both their morphology and cellular composition, even within an individual patient. Tumors are composed of cancer cells and a range of other cell types, which, together with a variety of secreted molecules, collectively comprise the tumor microenvironment (TME). Cells of the TME can communicate with each other and with distant tissues in a form of molecular cross-talk to influence their growth and function. Cross-talk between cancer cells and local immune cells is well described and can lead to the induction of local immunosuppression. Recently, it has become apparent that tumors located remotely from each other, can engage in cross-talk that can influence their responsiveness to various therapies, including immunotherapy. In this article, we review studies that describe how tumors systemically communicate with distant tissues through motile cells, extracellular vesicles, and secreted molecules that can affect their function. In addition, we summarize evidence from mouse studies and the clinic that indicate an ability of some tumors to influence the progression and therapeutic responses of other tumors in different anatomical locations.
Collapse
Affiliation(s)
- Amanda J Oliver
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Phillip K Darcy
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Joseph A Trapani
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Michael H Kershaw
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Clare Y Slaney
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| |
Collapse
|
233
|
Elliot A, Myllymäki H, Feng Y. Inflammatory Responses during Tumour Initiation: From Zebrafish Transgenic Models of Cancer to Evidence from Mouse and Man. Cells 2020; 9:cells9041018. [PMID: 32325966 PMCID: PMC7226149 DOI: 10.3390/cells9041018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 04/08/2020] [Accepted: 04/14/2020] [Indexed: 12/12/2022] Open
Abstract
The zebrafish is now an important model organism for cancer biology studies and provides unique and complementary opportunities in comparison to the mammalian equivalent. The translucency of zebrafish has allowed in vivo live imaging studies of tumour initiation and progression at the cellular level, providing novel insights into our understanding of cancer. Here we summarise the available transgenic zebrafish tumour models and discuss what we have gleaned from them with respect to cancer inflammation. In particular, we focus on the host inflammatory response towards transformed cells during the pre-neoplastic stage of tumour development. We discuss features of tumour-associated macrophages and neutrophils in mammalian models and present evidence that supports the idea that these inflammatory cells promote early stage tumour development and progression. Direct live imaging of tumour initiation in zebrafish models has shown that the intrinsic inflammation induced by pre-neoplastic cells is tumour promoting. Signals mediating leukocyte recruitment to pre-neoplastic cells in zebrafish correspond to the signals that mediate leukocyte recruitment in mammalian tumours. The activation state of macrophages and neutrophils recruited to pre-neoplastic cells in zebrafish appears to be heterogenous, as seen in mammalian models, which provides an opportunity to study the plasticity of innate immune cells during tumour initiation. Although several potential mechanisms are described that might mediate the trophic function of innate immune cells during tumour initiation in zebrafish, there are several unknowns that are yet to be resolved. Rapid advancement of genetic tools and imaging technologies for zebrafish will facilitate research into the mechanisms that modulate leukocyte function during tumour initiation and identify targets for cancer prevention.
Collapse
Affiliation(s)
| | | | - Yi Feng
- Correspondence: ; Tel.: +44-(0)131-242-6685
| |
Collapse
|
234
|
Long Y, Lu Z, Xu S, Li M, Wang X, Zhang Z, He Q. Self-Delivery Micellar Nanoparticles Prevent Premetastatic Niche Formation by Interfering with the Early Recruitment and Vascular Destruction of Granulocytic Myeloid-Derived Suppressor Cells. NANO LETTERS 2020; 20:2219-2229. [PMID: 31823615 DOI: 10.1021/acs.nanolett.9b03883] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Distal metastases of tumors result from the interaction between "seeds" (circulating tumor cells, CTCs) and "soil" (premetastatic niche, PMN). Various strategies focus on CTC inhibition, but only a few strategies inhibit PMN formation. The main predisposition of PMN formation in melanoma lies in the pulmonary recruitment of granulocytic myeloid-derived suppressor cells (G-MDSCs, CD11b+Ly6G+ cells) induced by tumors, which increase vascular permeability by secreting matrix metalloproteinase-9 (MMP-9) and result in immunosuppression by secreting interleukin-10 (IL-10) in premetastatic lungs. Here, a micellar hypotoxic low molecular weight heparin-tocopherol succinate nanoparticle (LMWH-TOS nanoparticle, LT NP) was established and investigated for its influence on PMN formation in this study. We first demonstrated that the hydrophilic segment LMWH in LT NPs can inhibit early pulmonary recruitment of G-MDSCs through interrupting their extravasation by inhibiting P-selectin/PSGL-1-mediated adhesion between vascular endothelial cells and G-MDSCs. In addition, the hydrophobic segment (TOS) in LT NPs significantly inhibited the expression of MMP-9 in G-MDSCs. As a result, the drug-free nanoparticles could maintain the normal microenvironment of lungs, thus effectively inhibiting implantation and colonization of CTCs. Further, phenylboronic acid (PBA)-modified and doxorubicin/immunopotentiator α-galactosylceramide (αGC)-coloaded nanoparticles (PLT/DOX/αGC NPs) were exploited. PBA modification achieved targeted chemotherapy by binding to overexpressed sialic acid residues on the tumor cell surface. This nanosystem effectively inhibited the postoperative metastasis and tumor recurrence simultaneously. Our work provides a proof of concept that the prevention of PMN formation through interfering G-MDSCs with self-delivery nanosystems is a safe and effective antimetastasis strategy.
Collapse
Affiliation(s)
- Yang Long
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zhengze Lu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Shanshan Xu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Man Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xuhui Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zhirong Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Qin He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
235
|
The complexity of neutrophils in health and disease: Focus on cancer. Semin Immunol 2020; 48:101409. [PMID: 32958359 PMCID: PMC7500440 DOI: 10.1016/j.smim.2020.101409] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/21/2020] [Accepted: 09/04/2020] [Indexed: 12/14/2022]
Abstract
Neutrophils are essential soldiers of the immune response and their role have long been restricted to their activities in defence against microbial infections and during the acute phase of the inflammatory response. However, increasing number of investigations showed that neutrophils are endowed with plasticity and can participate in the orchestration of both innate and adaptive immune responses. Neutrophils have an impact on a broad range of disorders, including infections, chronic inflammations, and cancer. Neutrophils are present in the tumour microenvironment and have been reported to mediate both pro-tumour and anti-tumour responses. Neutrophils can contribute to genetic instability, tumour cell proliferation, angiogenesis and suppression of the anti-tumour immune response. In contrast, neutrophils are reported to mediate anti-tumour resistance by direct killing of tumour cells or by engaging cooperative interactions with other immune cells. Here we discuss the current understandings of neutrophils biology and functions in health and diseases, with a specific focus on their role in cancer biology and their prognostic significance in human cancer.
Collapse
|
236
|
Sällberg M, Pasetto A. Liver, Tumor and Viral Hepatitis: Key Players in the Complex Balance Between Tolerance and Immune Activation. Front Immunol 2020; 11:552. [PMID: 32292409 PMCID: PMC7119224 DOI: 10.3389/fimmu.2020.00552] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 03/10/2020] [Indexed: 12/12/2022] Open
Abstract
Liver cancer is the third most common cause of cancer related death in the World. From an epidemiological point of view the risk factors associated to primary liver cancer are mainly viral hepatitis infection and alcohol consumption. Even though there is a clear correlation between liver inflammation, cirrhosis and cancer, other emerging liver diseases (like fatty liver) could also lead to liver cancer. Moreover, the liver is the major site of metastasis from colon, breast, ovarian and other cancers. In this review we will address the peculiar status of the liver as organ that has to balance between tolerance and immune activation. We will focus on macrophages and other key cellular components of the liver microenvironment that play a central role during tumor progression. We will also discuss how current and future therapies may affect the balance toward immune activation.
Collapse
Affiliation(s)
- Matti Sällberg
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anna Pasetto
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
237
|
Liu L, Liu Y, Yan X, Zhou C, Xiong X. The role of granulocyte colony‑stimulating factor in breast cancer development: A review. Mol Med Rep 2020; 21:2019-2029. [PMID: 32186767 PMCID: PMC7115204 DOI: 10.3892/mmr.2020.11017] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 02/25/2020] [Indexed: 12/17/2022] Open
Abstract
Granulocyte-colony-stimulating factor (G-CSF) is a member of the hematopoietic growth factor family that primarily affects the neutrophil lineage. G-CSF serves as a powerful mobilizer of peripheral blood stem cells and recombinant human G-CSF (rhG-CSF) has been used to treat granulocytopenia and neutropenia after chemotherapy for cancer patients. However, recent studies have found that G-CSF plays an important role in cancer progression. G-CSF expression is increased in different types of cancer cells, such as lung cancer, gastric cancer, colorectal cancer, invasive bladder carcinoma, glioma and breast cancer. However, it is unclear whether treatment with G-CSF has an adverse effect. The current review provides an overview of G-CSF in malignant breast cancer development and the data presented in this review are expected to provide new ideas for cancer therapy.
Collapse
Affiliation(s)
- Li Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yangyang Liu
- Department of Anesthesiology, First Clinical Medical College, School of Medicine, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xiaohua Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Chong Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xiangyang Xiong
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
238
|
Li B, Lu J, Peng DZ, Zhang XY, You Z. Elevated platelet distribution width predicts poor prognosis in hilar cholangiocarcinoma. Medicine (Baltimore) 2020; 99:e19400. [PMID: 32195935 PMCID: PMC7220385 DOI: 10.1097/md.0000000000019400] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Although the platelet distribution width (PDW) has been reported as a reliable predictor of prognosis in several types of cancer, to our knowledge the prognostic value of PDW in hilar cholangiocarcinoma (HC) has not been studied. The aim of the study was to investigate the prognostic value of PDW in HC patients. A retrospective analysis of 292 consecutively recruited HC patients undergoing radical resection with at least a 5-year follow-up. The optimal cutoff value of PDW was determined by receiver operating characteristic (ROC) curve. Survival analysis by the Kaplan-Meier method and the difference between the clinico-pathologic variables and survival was evaluated by log-rank analysis. Multivariate analysis identified independent prognostic risk factors of overall survival (OS). ROC curve analysis suggested that the optimal cutoff value for the PDW was 16.55. There were significant associations of high PDW with high white blood cell (P < .001) and high neutril-to-lymph ratio (P < .001). In a multivariate analysis, the PDW was an independent prognostic factor for overall survival (HR = 2.521, 95% CI 1.832-3.470, P < .001). In conclusions, our findings indicate that PDW may have clinical significance in predicting OS after surgery in HC patients.
Collapse
Affiliation(s)
- Bei Li
- Department of Biliary Surgery, West China Hospital of Sichuan University, No. 37 Guo Xue Xiang
- West China-Washington Mitochondria Metabolism Center, West China Hospital of Sichuan University, Chengdu, Sichuan Province, China
| | - Jiong Lu
- Department of Biliary Surgery, West China Hospital of Sichuan University, No. 37 Guo Xue Xiang
| | - Ding-Zhong Peng
- Department of Biliary Surgery, West China Hospital of Sichuan University, No. 37 Guo Xue Xiang
| | - Xin-Yi Zhang
- Department of Biliary Surgery, West China Hospital of Sichuan University, No. 37 Guo Xue Xiang
| | - Zhen You
- Department of Biliary Surgery, West China Hospital of Sichuan University, No. 37 Guo Xue Xiang
| |
Collapse
|
239
|
Haibe Y, Kreidieh M, El Hajj H, Khalifeh I, Mukherji D, Temraz S, Shamseddine A. Resistance Mechanisms to Anti-angiogenic Therapies in Cancer. Front Oncol 2020; 10:221. [PMID: 32175278 PMCID: PMC7056882 DOI: 10.3389/fonc.2020.00221] [Citation(s) in RCA: 250] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 02/10/2020] [Indexed: 12/12/2022] Open
Abstract
Tumor growth and metastasis rely on tumor vascular network for the adequate supply of oxygen and nutrients. Tumor angiogenesis relies on a highly complex program of growth factor signaling, endothelial cell (EC) proliferation, extracellular matrix (ECM) remodeling, and stromal cell interactions. Numerous pro-angiogenic drivers have been identified, the most important of which is the vascular endothelial growth factor (VEGF). The importance of pro-angiogenic inducers in tumor growth, invasion and extravasation make them an excellent therapeutic target in several types of cancers. Hence, the number of anti-angiogenic agents developed for cancer treatment has risen over the past decade, with at least eighty drugs being investigated in preclinical studies and phase I-III clinical trials. To date, the most common approaches to the inhibition of the VEGF axis include the blockade of VEGF receptors (VEGFRs) or ligands by neutralizing antibodies, as well as the inhibition of receptor tyrosine kinase (RTK) enzymes. Despite promising preclinical results, anti-angiogenic monotherapies led only to mild clinical benefits. The minimal benefits could be secondary to primary or acquired resistance, through the activation of alternative mechanisms that sustain tumor vascularization and growth. Mechanisms of resistance are categorized into VEGF-dependent alterations, non-VEGF pathways and stromal cell interactions. Thus, complementary approaches such as the combination of these inhibitors with agents targeting alternative mechanisms of blood vessel formation are urgently needed. This review provides an updated overview on the pathophysiology of angiogenesis during tumor growth. It also sheds light on the different pro-angiogenic and anti-angiogenic agents that have been developed to date. Finally, it highlights the preclinical evidence for mechanisms of angiogenic resistance and suggests novel therapeutic approaches that might be exploited with the ultimate aim of overcoming resistance and improving clinical outcomes for patients with cancer.
Collapse
Affiliation(s)
- Yolla Haibe
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut-Medical Center, Beirut, Lebanon
| | - Malek Kreidieh
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut-Medical Center, Beirut, Lebanon
| | - Hiba El Hajj
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut-Medical Center, Beirut, Lebanon
- Department of Experimental Pathology, Immunology and Microbiology, American University of Beirut-Medical Center, Beirut, Lebanon
| | - Ibrahim Khalifeh
- Department of Pathology and Laboratory Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Deborah Mukherji
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut-Medical Center, Beirut, Lebanon
| | - Sally Temraz
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut-Medical Center, Beirut, Lebanon
| | - Ali Shamseddine
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut-Medical Center, Beirut, Lebanon
| |
Collapse
|
240
|
Huang T, Li Y, Zhou Y, Lu B, Zhang Y, Tang D, Gan Y, He Z, Chen Z, Yu W, Li P. Stroke Exacerbates Cancer Progression by Upregulating LCN2 in PMN-MDSC. Front Immunol 2020; 11:299. [PMID: 32153594 PMCID: PMC7050632 DOI: 10.3389/fimmu.2020.00299] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 02/06/2020] [Indexed: 12/20/2022] Open
Abstract
Acute ischemic stroke (AIS) is common in patients with cancer, and mounting clinical evidence suggests that it may shorten the survival of cancer patients. But how stroke affects the progression of cancer remains unclear. We inoculated B16 tumor cells (2 × 105) subcutaneously before distal middle cerebral artery occlusion (dMCAO) or sham surgery in C57BL/6 mice and found that compared to sham operated mice, dMCAO mice developed significantly increased tumor volume and were accompanied by lower survival rate. To explore the underlying mechanism, we performed RNA-sequencing analysis of the tumor tissue from mice with or without stroke and found prominent upregulation of lipocalin 2 (LCN2) in the tumor from stroke mice compared to those from sham mice. Using quantitative reverse transcription-PCR, we confirmed increased mRNA expression of LCN2 as well as anti-inflammatory cytokines-Arg1, IL-10, and decreased mRNA level of pro-inflammatory cytokines-IL-6, IL-23 in the tumor of cancer-bearing stroke mice. Both immunofluorescence staining and flow cytometry analysis revealed that increased expression of LCN2 was mainly derived from the polymorphonuclear myeloid derived suppressor cells (PMN-MDSCs) in the tumor. We also found that stroke reduced the PMN-MDSCs in the peripheral blood, but increased PMN-MDSCs in the tumor of the cancer-bearing mice after stroke. In conclusion, cerebral ischemic stroke may exacerbate cancer progression by increasing LCN2 expression in PMN-MDSCs, which turns out to be a promising therapeutic target to suppress cancer progression after ischemic stroke.
Collapse
Affiliation(s)
- Tingting Huang
- Department of Anesthesiology, Renji Hospital, School of Medicine Shanghai Jiaotong University, Shanghai, China
| | - Yan Li
- Department of Anesthesiology, Renji Hospital, School of Medicine Shanghai Jiaotong University, Shanghai, China
| | - Yuxi Zhou
- Department of Anesthesiology, Renji Hospital, School of Medicine Shanghai Jiaotong University, Shanghai, China
| | - Bingwei Lu
- Department of Anesthesiology, Renji Hospital, School of Medicine Shanghai Jiaotong University, Shanghai, China
| | - Yueman Zhang
- Department of Anesthesiology, Renji Hospital, School of Medicine Shanghai Jiaotong University, Shanghai, China
| | - Dan Tang
- Department of Anesthesiology, Renji Hospital, School of Medicine Shanghai Jiaotong University, Shanghai, China
| | - Yu Gan
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, Shanghai, China
| | - Zhengzhou He
- Department of Anesthesiology, Renji Hospital, School of Medicine Shanghai Jiaotong University, Shanghai, China
| | - Zengai Chen
- Department of Radiology, Renji Hospital, School of Medicine Shanghai Jiaotong University, Shanghai, China
| | - Weifeng Yu
- Department of Anesthesiology, Renji Hospital, School of Medicine Shanghai Jiaotong University, Shanghai, China
| | - Peiying Li
- Department of Anesthesiology, Renji Hospital, School of Medicine Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
241
|
Trovato R, Canè S, Petrova V, Sartoris S, Ugel S, De Sanctis F. The Engagement Between MDSCs and Metastases: Partners in Crime. Front Oncol 2020; 10:165. [PMID: 32133298 PMCID: PMC7040035 DOI: 10.3389/fonc.2020.00165] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 01/30/2020] [Indexed: 12/18/2022] Open
Abstract
Tumor metastases represent the major cause of cancer-related mortality, confirming the urgent need to identify key molecular pathways and cell-associated networks during the early phases of the metastatic process to develop new strategies to either prevent or control distal cancer spread. Several data revealed the ability of cancer cells to establish a favorable microenvironment, before their arrival in distant organs, by manipulating the cell composition and function of the new host tissue where cancer cells can survive and outgrow. This predetermined environment is termed “pre-metastatic niche” (pMN). pMN development requires that tumor-derived soluble factors, like cytokines, growth-factors and extracellular vesicles, genetically and epigenetically re-program not only resident cells (i.e., fibroblasts) but also non-resident cells such as bone marrow-derived cells. Indeed, by promoting an “emergency” myelopoiesis, cancer cells switch the steady state production of blood cells toward the generation of pro-tumor circulating myeloid cells defined as myeloid-derived suppressor cells (MDSCs) able to sustain tumor growth and dissemination. MDSCs are a heterogeneous subset of myeloid cells with immunosuppressive properties that sustain metastatic process. In this review, we discuss current understandings of how MDSCs shape and promote metastatic dissemination acting in each fundamental steps of cancer progression from primary tumor to metastatic disease.
Collapse
Affiliation(s)
- Rosalinda Trovato
- Section of Immunology, Department of Medicine, University of Verona, Verona, Italy
| | - Stefania Canè
- Section of Immunology, Department of Medicine, University of Verona, Verona, Italy
| | - Varvara Petrova
- Section of Immunology, Department of Medicine, University of Verona, Verona, Italy
| | - Silvia Sartoris
- Section of Immunology, Department of Medicine, University of Verona, Verona, Italy
| | - Stefano Ugel
- Section of Immunology, Department of Medicine, University of Verona, Verona, Italy
| | - Francesco De Sanctis
- Section of Immunology, Department of Medicine, University of Verona, Verona, Italy
| |
Collapse
|
242
|
Garner H, de Visser KE. Immune crosstalk in cancer progression and metastatic spread: a complex conversation. Nat Rev Immunol 2020; 20:483-497. [PMID: 32024984 DOI: 10.1038/s41577-019-0271-z] [Citation(s) in RCA: 276] [Impact Index Per Article: 55.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2019] [Indexed: 12/12/2022]
Abstract
Metastatic disease is responsible for approximately 90% of cancer deaths. For successful dissemination and metastasis, cancer cells must evade detection and destruction by the immune system. This process is enabled by factors secreted by the primary tumour that shape both the intratumoural microenvironment and the systemic immune landscape. Here, we review the evidence of aberrant immune cell crosstalk in metastasis formation and the role that primary tumours play in hijacking these interactions in order to enhance their metastatic potential. Moreover, we highlight the intriguing parallels between the inflammatory pathways underlying inflammatory disorders and cancer progression.
Collapse
Affiliation(s)
- Hannah Garner
- Division of Tumour Biology & Immunology, Oncode Institute, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Karin E de Visser
- Division of Tumour Biology & Immunology, Oncode Institute, Netherlands Cancer Institute, Amsterdam, Netherlands. .,Department of Immunohematology and Blood Transfusion, Leiden University Medical Centre, Leiden, Netherlands.
| |
Collapse
|
243
|
Charan M, Dravid P, Cam M, Setty B, Roberts RD, Houghton PJ, Cam H. Tumor secreted ANGPTL2 facilitates recruitment of neutrophils to the lung to promote lung pre-metastatic niche formation and targeting ANGPTL2 signaling affects metastatic disease. Oncotarget 2020; 11:510-522. [PMID: 32082485 PMCID: PMC7007290 DOI: 10.18632/oncotarget.27433] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 12/28/2019] [Indexed: 12/28/2022] Open
Abstract
The pre-metastatic niche (PMN) represents an abnormal microenvironment devoid of cancer cells, but favoring tumor growth. Little is known about the mechanisms that generate the PMN or their effects on host cells within metastasis-prone organs. Here, we investigated by using spontaneous metastatic models whether lung epithelial cells are essential for primary tumor induced neutrophil recruitment in lung and subsequently initiating PMN formation in osteosarcoma. We found that serum levels of ANGPTL2 in osteosarcoma patients are significantly higher compared to those in healthy controls and that ANGPTL2 secretion by tumor cells plays an essential role in osteosarcoma metastasis. We determined that tumor-derived ANGPTL2 stimulates lung epithelial cells, which is essential for primary tumor-induced neutrophil recruitment in lung and subsequent pre-metastatic niche formation. Lastly, we identified that a p63 isoform, ΔNp63, drives high level of ANGPTL2 secretion and pharmaceutical inhibition of ANGPTL2 signaling by a non–RGD-based integrin binding peptide (ATN-161) diminished metastatic load in lungs likely due to reduction of the lung pre-metastatic niche formation.
Collapse
Affiliation(s)
- Manish Charan
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Piyush Dravid
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Maren Cam
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Bhuvana Setty
- Department of Hematology & Oncology, Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | - Ryan D Roberts
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | - Peter J Houghton
- Greehey Children's Cancer Research Institute, Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Hakan Cam
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
244
|
Disease progression model of 4T1 metastatic breast cancer. J Pharmacokinet Pharmacodyn 2020; 47:105-116. [PMID: 31970615 DOI: 10.1007/s10928-020-09673-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 01/09/2020] [Indexed: 12/18/2022]
Abstract
Cancer metastasis is the main cause of death in various types of cancer. However, in the field of pharmacometrics, cancer disease progression models focus on the growth of primary tumors with tumor volume or weight as target values, while the metastasis process is less mentioned. We propose a series of mathematical models to quantitatively describe and predict the disease progression of 4T1 breast cancer in the aspect of primary breast tumor, lung metastasis and white blood cell. The 4T1 cells were injected into breast fat pad of female BALB/c mice to establish an animal model of breast cancer metastasis. The number and volume of lung metastases at different times were measured. Based on the above data, a disease progression model of breast cancer lung metastasis was established and parameter values were estimated. The white blood cell growth and the primary tumor growth of 4T1 mouse are also modeled. The established models can describe the lung metastasis of 4T1 breast cancer in three aspects: (1) the increase in metastasis number; (2) the growth of metastasis volume; (3) metastasis number-size distribution at different time points. Compared with the prior metastasis models based on von Forester equation, our models distinguished the growth rate of primary tumor and metastasis and got parameter values for 4T1 mouse model. And the current models optimized the metastasis number-size distribution model by utilizing logistic function instead of the prior power function. This study provides a comprehensive description of lung metastasis progression for 4T1 breast cancer model, as well as an alternative disease progression model structure for further pharmacodynamics modeling.
Collapse
|
245
|
Feng Z, Yu Q, Zhang T, Tie W, Li J, Zhou X. Updates on mechanistic insights and targeting of tumour metastasis. J Cell Mol Med 2020; 24:2076-2086. [PMID: 31957271 PMCID: PMC7011147 DOI: 10.1111/jcmm.14931] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 11/15/2019] [Accepted: 12/16/2019] [Indexed: 02/05/2023] Open
Abstract
Malignant tumours are one of the major diseases that seriously endanger human health. The characteristics of their invasion and metastasis are one of the main causes of death in cancer patients, and these features cannot be separated from the participation of various molecules-related cells living in the tumour microenvironment and specific structures. Tumour invasion can approximately be divided into several specific steps according to the movement of tumour cells. In each step, there are different actions in the tumour microenvironment that mediate the interactions among substances. Researchers are attempting to clarify every mechanism of the tumour dissemination. However, there is still a long way to the final determination. Here, we review these interactions in tumour invasion and metastasis at the structural, molecular and cellular levels. We also discuss the ongoing studies and the promise of targeting metastasis in tumour therapy.
Collapse
Affiliation(s)
- Zeru Feng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China College of Stomatology, Sichuan University, Chengdu, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Qiuxuan Yu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China College of Stomatology, Sichuan University, Chengdu, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Ting Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China College of Stomatology, Sichuan University, Chengdu, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Wanpeng Tie
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China College of Stomatology, Sichuan University, Chengdu, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Jing Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China College of Stomatology, Sichuan University, Chengdu, China
| | - Xikun Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| |
Collapse
|
246
|
Shaul ME, Eyal O, Guglietta S, Aloni P, Zlotnik A, Forkosh E, Levy L, Weber LM, Levin Y, Pomerantz A, Nechushtan H, Eruslanov E, Singhal S, Robinson MD, Krieg C, Fridlender ZG. Circulating neutrophil subsets in advanced lung cancer patients exhibit unique immune signature and relate to prognosis. FASEB J 2020; 34:4204-4218. [PMID: 31957112 DOI: 10.1096/fj.201902467r] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/19/2019] [Accepted: 01/06/2020] [Indexed: 12/31/2022]
Abstract
The accumulation of circulating low-density neutrophils (LDN) has been described in cancer patients and associated with tumor-supportive properties, as opposed to the high-density neutrophils (HDN). Here we aimed to evaluate the clinical significance of circulating LDN in lung cancer patients, and further assessed its diagnostic vs prognostic value. Using mass cytometry (CyTOF), we identified major subpopulations within the circulating LDN/HDN subsets and determined phenotypic modulations of these subsets along tumor progression. LDN were highly enriched in the low-density (LD) fraction of advanced lung cancer patients (median 7.0%; range 0.2%-80%, n = 64), but not in early stage patients (0.7%; 0.05%-6%; n = 35), healthy individuals (0.8%; 0%-3.5%; n = 15), or stable chronic obstructive pulmonary disease (COPD) patients (1.2%; 0.3%-7.4%, n = 13). Elevated LDN (>10%) remarkably related with poorer prognosis in late stage patients. We identified three main neutrophil subsets which proportions are markedly modified in cancer patients, with CD66b+ /CD10low /CXCR4+ /PDL1inter subset almost exclusively found in advanced lung cancer patients. We found substantial variability in subsets between patients, and demonstrated that HDN and LDN retain a degree of inherent spontaneous plasticity. Deep phenotypic characterization of cancer-related circulating neutrophils and their modulation along tumor progression is an important advancement in understanding the role of myeloid cells in lung cancer.
Collapse
Affiliation(s)
- Merav E Shaul
- Institute of Pulmonary Medicine, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Ophir Eyal
- Institute of Pulmonary Medicine, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Silvia Guglietta
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA.,Department of Dermatology, Medical University of South Carolina, Charleston, SC, USA
| | - Pazzit Aloni
- Institute of Pulmonary Medicine, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Asaf Zlotnik
- Institute of Pulmonary Medicine, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Ester Forkosh
- Institute of Pulmonary Medicine, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Liran Levy
- Institute of Pulmonary Medicine, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Lukas M Weber
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland.,SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland
| | - Yonathan Levin
- Institute of Pulmonary Medicine, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Alon Pomerantz
- Institute of Pulmonary Medicine, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Hovav Nechushtan
- Sharrett Institute of Oncology, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Evgeniy Eruslanov
- Thoracic Oncology Research Laboratory, University of Pennsylvania, Philadelphia, PA, USA
| | - Sunil Singhal
- Thoracic Oncology Research Laboratory, University of Pennsylvania, Philadelphia, PA, USA
| | - Mark D Robinson
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland.,SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland
| | - Carsten Krieg
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA.,Department of Dermatology, Medical University of South Carolina, Charleston, SC, USA
| | - Zvi G Fridlender
- Institute of Pulmonary Medicine, Hadassah Hebrew University Hospital, Jerusalem, Israel
| |
Collapse
|
247
|
Qin L, Li JY, Huang WJ, Zhang ML, Wang RT, Shen W. Higher platelet distribution width is associated with unfavorable prognosis in ovarian cancer. Cancer Biomark 2020; 28:365-370. [PMID: 32417761 DOI: 10.3233/cbm-191190] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Ovarian cancer (OC) is one of the most malignant gynecological cancers. Platelets play a profound role in cancer growth and metastasis. Platelet distribution width (PDW) is an indicator of platelet activation and is altered in malignancies. However, the prognostic value of PDW in OC remains unclear. This present study aimed to investigate the predictive significance of PDW in OC. METHODS 221 OC patients, between January 2013 and December 2013, were included in this study. The correlations between PDW and clinicopathological features were analyzed. Kaplan-Meier method and Cox regression were used to evaluate the prognostic impact of PDW. RESULTS Of the 221 patients, increased PDW levels were observed in 163 (73.6%) patients. Kaplan-Meier analysis revealed that higher PDW levels were associated with poor progression-free survival and overall survival (both p< 0.001). Cox-regression analysis confirmed the independent predictive value of PDW on overall survival (HR = 2.820, 95% CI = 1.776-4.476, p< 0.001). CONCLUSION Higher PDW levels predict poor prognosis in patients with OC. Elevated PDW may be a novel target for therapy.
Collapse
Affiliation(s)
- Ling Qin
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | - Jia-Yu Li
- Department of Obstetrics and Gynecology, King Chulalongkorn Memorial Hospital, Chulalongkorn University, Bangkok, Thailand
| | - Wen-Juan Huang
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | - Meng-Lin Zhang
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | - Rui-Tao Wang
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | - Wenjie Shen
- Department of Gynaecology and Obstetrics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
248
|
Jing W, Guo X, Wang G, Bi Y, Han L, Zhu Q, Qiu C, Tanaka M, Zhao Y. Breast cancer cells promote CD169 + macrophage-associated immunosuppression through JAK2-mediated PD-L1 upregulation on macrophages. Int Immunopharmacol 2019; 78:106012. [PMID: 31865052 DOI: 10.1016/j.intimp.2019.106012] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 09/27/2019] [Accepted: 10/27/2019] [Indexed: 01/10/2023]
Abstract
Macrophages are recognized as one of the major cell types in tumor microenvironment, and macrophage infiltration has been predominantly associated with poor prognosis among patients with breast cancer. Using the murine models of triple-negative breast cancer in CD169-DTR mice, we found that CD169+ macrophages support tumor growth and metastasis. CD169+ macrophage depletion resulted in increased accumulation of CD8+ T cells within tumor, and produced significant expansion of CD8+ T cells in circulation and spleen. In addition, we observed that CD169+ macrophage depletion alleviated tumor-induced splenomegaly in mice, but had no improvement in bone loss and repression of bone marrow erythropoiesis in tumor-bearing mice. Cancer cells and tumor associated macrophages exploit the upregulation of the immunosuppressive protein PD-L1 to subvert T cell-mediated immune surveillance. Within the tumor microenvironment, our understanding of the regulation of PD-L1 protein expression is limited. We showed that there was a 5-fold higher relative expression of PD-L1 on macrophages as compared with 4T1 tumor cells; coculture of macrophages with 4T1 cells augmented PD-L1 levels on macrophages, but did not upregulate the expression of PD-L1 on 4T1 cells. JAK2/STAT3 signaling pathway was activated in macrophages after coculture, and we further identified the JAK2 as a critical regulator of PD-L1 expression in macrophages during coculture with 4T1 cells. Collectively, our data reveal that breast cancer cells and CD169+ macrophages exhibit bidirectional interactions that play a critical role in tumor progression, and inhibition of JAK2 signaling pathway in CD169+ macrophages may be potential strategy to block tumor microenvironment-derived immune escape.
Collapse
Affiliation(s)
- Weiqiang Jing
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Xing Guo
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Ganyu Wang
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Yuxuan Bi
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Lihui Han
- Department of Immunology, Shandong Provincial Key Laboratory of Infection & Immunology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Qingfen Zhu
- Shandong Institute for Food and Drug Control, Jinan, China.
| | - Chunhong Qiu
- Department of Cell Biology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Masato Tanaka
- Laboratory of Immune Regulation, School of Life Science, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Yunxue Zhao
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China; Department of Immunology, Shandong Provincial Key Laboratory of Infection & Immunology, School of Basic Medical Sciences, Shandong University, Jinan, China.
| |
Collapse
|
249
|
Snoderly HT, Boone BA, Bennewitz MF. Neutrophil extracellular traps in breast cancer and beyond: current perspectives on NET stimuli, thrombosis and metastasis, and clinical utility for diagnosis and treatment. Breast Cancer Res 2019; 21:145. [PMID: 31852512 PMCID: PMC6921561 DOI: 10.1186/s13058-019-1237-6] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 12/04/2019] [Indexed: 12/30/2022] Open
Abstract
The formation of neutrophil extracellular traps (NETs), known as NETosis, was first observed as a novel immune response to bacterial infection, but has since been found to occur abnormally in a variety of other inflammatory disease states including cancer. Breast cancer is the most commonly diagnosed malignancy in women. In breast cancer, NETosis has been linked to increased disease progression, metastasis, and complications such as venous thromboembolism. NET-targeted therapies have shown success in preclinical cancer models and may prove valuable clinical targets in slowing or halting tumor progression in breast cancer patients. We will briefly outline the mechanisms by which NETs may form in the tumor microenvironment and circulation, including the crosstalk between neutrophils, tumor cells, endothelial cells, and platelets as well as the role of cancer-associated extracellular vesicles in modulating neutrophil behavior and NET extrusion. The prognostic implications of cancer-associated NETosis will be explored in addition to development of novel therapeutics aimed at targeting NET interactions to improve outcomes in patients with breast cancer.
Collapse
Affiliation(s)
- Hunter T Snoderly
- Department of Chemical and Biomedical Engineering, West Virginia University, 1306 Evansdale Drive, ESB 521, Morgantown, WV, 26506, USA
| | - Brian A Boone
- Department of Surgery, West Virginia University, Morgantown, WV, 26506, USA
| | - Margaret F Bennewitz
- Department of Chemical and Biomedical Engineering, West Virginia University, 1306 Evansdale Drive, ESB 521, Morgantown, WV, 26506, USA.
| |
Collapse
|
250
|
Zhang X, Qin YY, Chen M, Wu YY, Lin FQ. Combined Use of Mean Platelet Volume/Platelet Count Ratio and Platelet Distribution Width to Distinguish Between Patients with Nasopharyngeal Carcinoma, Those with Benign Tumors of the Nasopharynx, and Healthy Subjects. Cancer Manag Res 2019; 11:10375-10382. [PMID: 31849526 PMCID: PMC6913058 DOI: 10.2147/cmar.s226050] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 11/20/2019] [Indexed: 01/13/2023] Open
Abstract
Purpose For the diagnosis of nasopharyngeal carcinoma (NPC), reliable early indicators with sensitivity and specificity should be sought. This study evaluated the effect of the combined use of mean platelet volume/platelet count ratio (MPV/PC ratio) and platelet distribution width (PDW) for differential diagnosis of NPC. In this study, MPV/PC ratio was used for the first time to diagnostically evaluate NPC. Patients and methods We retrospectively analyzed various hematological indices of three subject groups (208, 185, and 162 patients with NPC, benign tumors of the nasopharynx, and healthy subjects, respectively) and evaluated the value of combined use of MPV/PC ratio and PDW for differential diagnosis of the three groups using the one-way analysis of variance. Results Comparison of laboratory variables between the three groups showed a significant difference in MPV/PC ratio and PDW (P<0.001, all). The MPV/PC ratio in the NPC group was significantly lower than the other two groups (P<0.001); MPV/PC ratio also showed a statistically significant difference in different stages (P=0.034) and serosal invasions (P<0.001) of the NPC group. Receiver operating characteristic curve (ROC) analysis showed that areas under the curve (AUC) of either patients with benign tumors of the nasopharynx (AUCMPV/PCratio+PDW: 0.708) or healthy subjects (AUCMPV/PCratio+PDW: 0.909) were larger than those of MPV/PC ratio (AUCMPV/PCratio: 0.665, 0.869, respectively) and PDW (AUCPDW:0.614, 0.716, respectively) use alone (P<0.05, all). Conclusion MPV/PC ratio and PDW may be used as indexes of NPC. MPV/PC ratio combined with PDW could be considered as meaningful laboratory indexes for differential diagnosis of NPC, benign tumors of the nasopharynx, and healthy subjects. This finding could enhance the detection of NPC.
Collapse
Affiliation(s)
- Xuan Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Yuan-Yuan Qin
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Min Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Yang-Yang Wu
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Fa-Quan Lin
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| |
Collapse
|