1
|
Song J, Xie D, Wei X, Liu B, Yao F, Ye W. A cuproptosis-related lncRNAs signature predicts prognosis and reveals pivotal interactions between immune cells in colon cancer. Heliyon 2024; 10:e34586. [PMID: 39114018 PMCID: PMC11305305 DOI: 10.1016/j.heliyon.2024.e34586] [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: 09/17/2023] [Revised: 07/11/2024] [Accepted: 07/11/2024] [Indexed: 08/10/2024] Open
Abstract
Copper-mediated cell death presents distinct pathways from established apoptosis processes, suggesting alternative therapeutic approaches for colon cancer. Our research aims to develop a predictive framework utilizing long-noncoding RNAs (lncRNAs) related to cuproptosis to predict colon cancer outcomes while examining immune interactions and intercellular signaling. We obtained colon cancer-related human mRNA expression profiles and clinical information from the Cancer Genome Atlas repository. To isolate lncRNAs involved in cuproptosis, we applied Cox proportional hazards modeling alongside the least absolute shrinkage and selection operator technique. We elucidated the underlying mechanisms by examining the tumor mutational burden, the extent of immune cell penetration, and intercellular communication dynamics. Based on the model, drugs were predicted and validated with cytological experiments. A 13 lncRNA-cuproptosis-associated risk model was constructed. Two colon cancer cell lines were used to validate the predicted representative mRNAs with high correlation coefficients with copper-induced cell death. Survival enhancement in the low-risk cohort was evidenced by the trends in Kaplan-Meier survival estimates. Analysis of immune cell infiltration suggested that survival was induced by the increased infiltration of naïve CD4+ T cells and a reduction of M2 macrophages within the low-risk faction. Decreased infiltration of naïve B cells, resting NK cells, and M0 macrophages was significantly associated with better overall survival. Combined single-cell analysis suggested that CCL5-ACKR1, CCL2-ACKR1, and CCL5-CCR1 pathways play key roles in mediating intercellular dialogues among immune constituents within the neoplastic microhabitat. We identified three drugs with a high sensitivity in the high-risk group. In summary, this discovery establishes the possibility of using 13 cuproptosis-associated lncRNAs as a risk model to assess the prognosis, unravel the immune mechanisms and cell communication, and improve treatment options, which may provide a new idea for treating colon cancer.
Collapse
Affiliation(s)
- Jingru Song
- Department of Gastroenterology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310007, Zhejiang, China
| | - Dong Xie
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xia Wei
- Department of Gastroenterology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310007, Zhejiang, China
| | - Binbin Liu
- Department of Gastroenterology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310007, Zhejiang, China
| | - Fang Yao
- Department of Gastroenterology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310007, Zhejiang, China
| | - Wei Ye
- Department of Gastroenterology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310007, Zhejiang, China
| |
Collapse
|
2
|
González-Arriagada WA, Coletta RD, Lozano-Burgos C, García C, Maripillán J, Alcayaga-Miranda F, Godínez-Pacheco B, Oyarce-Pezoa S, Martínez-Flores R, García IE. CR5/CCL5 axis is linked to a poor outcome, and inhibition reduces metastasis in oral squamous cell carcinoma. J Cancer Res Clin Oncol 2023; 149:17335-17346. [PMID: 37831273 DOI: 10.1007/s00432-023-05443-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 09/21/2023] [Indexed: 10/14/2023]
Abstract
PURPOSE The CCR5/CCL5 axis is essential for interactions between malignant cells and microenvironment components, promoting tumor progression in oral squamous cell carcinoma (OSCC). This study aims to evaluate the association of CCL5 and CCR5 with the behavior of oral cancer and assess the therapeutic potential of a CCR5 antagonist. METHODS A retrospective study to analyze CCR5 and CCL5 expression on paraffin-embedded tissues was performed. In cell lines, rhCCL5 was added to induce CCR5-related pathways, and Maraviroc and shRNA against CCR5 were used to neutralize the receptor. Finally, an in vivo murine orthotopic xenograft model of tongue cancer was used to evaluate Maraviroc as an oncologic therapy. After 15 days, the mice were killed, and the primary tumors and cervical lymph nodes were analyzed. RESULTS The expression of CCR5 was associated with clinical stage and metastasis, and CCL5 was related to overall survival. Adding rhCCL5 induced cell proliferation, while shRNA and Maraviroc reduced it in a dose-dependent manner. Maraviroc treatment also increased apoptosis and modified cytoskeletal organization. In vivo, Maraviroc reduced neck metastasis. CONCLUSIONS The effects of CCR5 antagonists in OSCC have been poorly studied, and this study reports in vitro and in vivo evidence for the effects of Maraviroc in OSCC. Our results suggest that the CCR5/CCL5 axis plays a role in oral cancer behavior, and that its inhibition is a promising new therapy alternative.
Collapse
Affiliation(s)
- Wilfredo Alejandro González-Arriagada
- Facultad de Odontología, Universidad de los Andes, Santiago, Chile.
- Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago, Chile.
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile.
| | - Ricardo D Coletta
- Oral Pathology and Graduate Program in Oral Biology, Piracicaba Dental School, State University of Campinas, Piracicaba, Brazil
| | | | - Cynthia García
- PhD Program in Biomedicine, Universidad de los Andes, Santiago, Chile
| | - Jaime Maripillán
- Laboratorio de Fisiología Molecular y Biofísica, Facultad de Odontología, Universidad de Valparaíso, Valparaíso, Chile
- Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| | - Francisca Alcayaga-Miranda
- Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | | | | | - René Martínez-Flores
- Facultad de Odontología, Unidad de Patología y Medicina Oral, Universidad Andres Bello, Santiago, Chile
| | - Isaac E García
- Laboratorio de Fisiología Molecular y Biofísica, Facultad de Odontología, Universidad de Valparaíso, Valparaíso, Chile
- Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
- Centro de Investigación Interoperativa en Ciencias Odontológicas y Médicas, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
3
|
Qin R, Ren W, Ya G, Wang B, He J, Ren S, Jiang L, Zhao S. Role of chemokines in the crosstalk between tumor and tumor-associated macrophages. Clin Exp Med 2023; 23:1359-1373. [PMID: 36173487 PMCID: PMC10460746 DOI: 10.1007/s10238-022-00888-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 09/07/2022] [Indexed: 11/03/2022]
Abstract
Tumor microenvironment (TME) consists of a dynamic network of non-tumoral stromal cells, including cancer-associated fibroblasts, endothelial cells, tumor-associated macrophages (TAMs), B and T cells. In the TME, TAMs support tumor initiation, progression, invasion and metastasis by promoting angiogenesis and immunosuppression of the tumor cells. There is close crosstalk between TAMs and tumor cells. Notably, chemokines are a significant messenger mediating the crosstalk between tumor cells and TAMs. TAMs can promote tumor progression via secretion of chemokines. Various chemokines secreted by tumors are involved in the generation and polarization of TAMs, the infiltration of TAMs in tumors, and the development of TAMs' suppressive function. This paper reviews CCL2-CCR2, CCL3/5-CCR5, CCL15-CCR1, CCL18-CCR8, CX3CL1/CCL26-CX3CR1, CXCL8-CXCR1/2, CXCL12-CXCR4/CXCR7 signaling pathways, their role in the recruitment, polarization and exertion of TAMs, and their correlation with tumor development, metastasis and prognosis. Furthermore, we present the current research progress on modulating the effects of TAMs with chemokine antagonists and discuss the prospects and potential challenges of using chemokine antagonists as therapeutic tools for cancer treatment. The TAMs targeting by chemokine receptor antagonists in combination with chemotherapy drugs, immune checkpoint inhibitors or radiotherapy appears to be a promising approach.
Collapse
Affiliation(s)
- Rui Qin
- The First Clinical Medical Institute, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Weihong Ren
- Department of Laboratory Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China.
| | - Guoqi Ya
- The First Clinical Medical Institute, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Bei Wang
- The First Clinical Medical Institute, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Jiao He
- The First Clinical Medical Institute, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Shaoxin Ren
- The First Clinical Medical Institute, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Lu Jiang
- Department of Laboratory Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Shuo Zhao
- Department of Laboratory Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
4
|
Li YT, Yuan WZ, Jin WL. Vagus innervation in the gastrointestinal tumor: Current understanding and challenges. Biochim Biophys Acta Rev Cancer 2023; 1878:188884. [PMID: 36990250 DOI: 10.1016/j.bbcan.2023.188884] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 02/17/2023] [Accepted: 02/28/2023] [Indexed: 03/30/2023]
Abstract
The vagus nerve (VN) is the main parasympathetic nerve of the autonomic nervous system. It is widely distributed in the gastrointestinal tract and maintains gastrointestinal homeostasis with the sympathetic nerve under physiological conditions. The VN communicates with various components of the tumor microenvironment to positively and dynamically affect the progression of gastrointestinal tumors (GITs). The intervention in vagus innervation delays GIT progression. Developments in adeno-associated virus vectors, nanotechnology, and in vivo neurobiological techniques have enabled the creation of precisely regulated "tumor neurotherapies". Furthermore, the combination of neurobiological techniques and single cell sequencing may reveal more insights into VN and GIT. The present review aimed to summarize the mechanisms of communication between the VN and the gastrointestinal TME and to explore the potential and challenges of VN-based tumor neurotherapy in GITs.
Collapse
|
5
|
González-Arriagada WA, García IE, Martínez-Flores R, Morales-Pison S, Coletta RD. Therapeutic Perspectives of HIV-Associated Chemokine Receptor (CCR5 and CXCR4) Antagonists in Carcinomas. Int J Mol Sci 2022; 24:ijms24010478. [PMID: 36613922 PMCID: PMC9820365 DOI: 10.3390/ijms24010478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 12/29/2022] Open
Abstract
The interaction between malignant cells and the tumor microenvironment is critical for tumor progression, and the chemokine ligand/receptor axes play a crucial role in this process. The CXCR4/CXCL12 and CCR5/CCL5 axes, both related to HIV, have been associated with the early (epithelial-mesenchymal transition and invasion) and late events (migration and metastasis) of cancer progression. In addition, these axes can also modulate the immune response against tumors. Thus, antagonists against the receptors of these axes have been proposed in cancer therapy. Although preclinical studies have shown promising results, clinical trials are needed to include these drugs in the oncological treatment protocols. New alternatives for these antagonists, such as dual CXCR4/CCR5 antagonists or combined therapy in association with immunotherapy, need to be studied in cancer therapy.
Collapse
Affiliation(s)
- Wilfredo Alejandro González-Arriagada
- Facultad de Odontología, Universidad de Los Andes, Santiago 7620086, Chile
- Centro de Investigación e Innovación Biomédica (CIIB), Universidad de los Andes, Santiago 7620086, Chile
- Patología Oral y Maxilofacial, Hospital El Carmen Luis Valentín Ferrada, Maipú 9251521, Chile
- Correspondence: ; Tel.: +562-2618-1000
| | - Isaac E. García
- Laboratorio de Fisiología y Biofísica, Facultad de Odontología, Universidad de Valparaíso, Valparaíso 2360004, Chile
- Centro de Investigación en Ciencias Odontológicas y Médicas, Universidad de Valparaíso, Valparaíso 2360004, Chile
- Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso, Valparaíso 2381850, Chile
| | - René Martínez-Flores
- Unidad de Patología y Medicina Oral, Facultad de Odontología, Universidad Andres Bello, Viña del Mar 2531015, Chile
| | - Sebastián Morales-Pison
- Centro de Oncología de Precisión (COP), Facultad de Medicina y Ciencias de la Salud, Universidad Mayor, Santiago 7560908, Chile
| | - Ricardo D. Coletta
- Department of Oral Diagnosis and Graduate Program in Oral Biology, Piracicaba Dental School, University of Campinas, Piracicaba 13414-903, SP, Brazil
| |
Collapse
|
6
|
Haag GM, Springfeld C, Grün B, Apostolidis L, Zschäbitz S, Dietrich M, Berger AK, Weber TF, Zoernig I, Schaaf M, Waberer L, Müller DW, Al-Batran SE, Halama N, Jaeger D. Pembrolizumab and maraviroc in refractory mismatch repair proficient/microsatellite-stable metastatic colorectal cancer – The PICCASSO phase I trial. Eur J Cancer 2022; 167:112-122. [DOI: 10.1016/j.ejca.2022.03.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 03/13/2022] [Indexed: 12/20/2022]
|
7
|
Yang J, Sontag D, Gong Y, Minuk GY. Alterations in chemokine receptor CCR5 activity influence tumor cell biology in human cholangiocarcinoma cell lines. Ann Hepatol 2021; 21:100265. [PMID: 33045415 DOI: 10.1016/j.aohep.2020.09.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/08/2020] [Accepted: 09/18/2020] [Indexed: 02/07/2023]
Abstract
INTRODUCTION AND OBJECTIVES Intrahepatic (I-CCA) and extrahepatic (E-CCA) cholangiocarcinoma (CCA) have different growth patterns and risks for tumor metastasis. Inhibition and/or activation of the chemokine receptor CCR subclasses have been reported to alter tumor cell biology in non-CCA cancers. In this study we documented CCR expression profiles in representative human I-CCA and E-CCA cell lines and the in vitro effects of CCR antagonists and agonists on tumor cell biology. MATERIALS AND METHODS CCR expression profiles were documented by real-time reverse transcription polymerase chain reaction; cell proliferation by WST-1; spheroid formation by sphere dimensions in anchorage-free medium; cell migration by wound healing and invasion by Transwell invasion chambers. RESULTS All 10 CCR motifs (CCR1-10) were expressed in the I-CCA, HuCCT1 cell line and six (CCR4, 5, 6, 8, 9 and 10) in the E-CCA, KMBC cell line. In HuCCT1 cells, CCR5 expression was most abundant whereas in KMBC cells, CCR6 followed by CCR5 were most abundant. The CCR5 antagonist Maraviroc significantly inhibited cell proliferation, migration and invasion in HuCCT1 cells, and spheroid formation and invasion in KMBC cells. The CCR5 agonist RANTES had no effect on HuCCT1 cells but increased cell proliferation, migration and invasion of KMBC cells. CONCLUSION These results suggest that CCR expression profiles differ in I-CCA and E-CCA. They also indicate that CCR5 antagonists and agonists have cell-specific effects but in general, CCR5 inactivation inhibits CCA tumor cell aggressiveness. Additional research is required to determine whether CCR5 inactivation is of value in the treatment of CCA in humans.
Collapse
MESH Headings
- Bile Duct Neoplasms/genetics
- Bile Duct Neoplasms/metabolism
- Bile Duct Neoplasms/pathology
- Bile Ducts, Extrahepatic/metabolism
- Bile Ducts, Extrahepatic/pathology
- Bile Ducts, Intrahepatic/metabolism
- Bile Ducts, Intrahepatic/pathology
- Biomarkers, Tumor/biosynthesis
- Biomarkers, Tumor/genetics
- Cell Line, Tumor
- Cell Proliferation/genetics
- Cholangiocarcinoma/genetics
- Cholangiocarcinoma/metabolism
- Cholangiocarcinoma/pathology
- DNA, Neoplasm/genetics
- DNA, Neoplasm/metabolism
- Gene Expression Regulation, Neoplastic
- Humans
- Receptors, CCR5/biosynthesis
- Receptors, CCR5/genetics
- Signal Transduction
Collapse
Affiliation(s)
- Jiaqi Yang
- Section of Hepatology, Department of Internal Medicine, Rudy Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - David Sontag
- College of Pharmacy, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Yuewen Gong
- College of Pharmacy, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Gerald Y Minuk
- Section of Hepatology, Department of Internal Medicine, Rudy Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada; College of Pharmacy, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
8
|
Abstract
INTRODUCTION Chemokines and their cognate receptors play a major role in modulating inflammatory responses. Depending on their ligand binding, chemokine receptors can stimulate both immune activating and inhibitory signaling pathways. The CC chemokine receptor 5 (CCR5) promotes immune responses by recruiting immune cells to the sites of inflammation/tumor, and is involved in stimulating tumor cell proliferation, invasion and migration through various mechanisms. Moreover, CCR5 also contributes to an immune-suppressive tumor microenvironment by recruiting regulatory T cells and myeloid-derived suppressor cells facilitating tumor development and progression. In summary, cells expressing CCR5 modulate immune response and tumor progression. Expression of CCR5 is increased in various malignancies and associated with poor outcome. Experimental data show promising efficacy signals with CCR5 antagonists in preclinical tumor models. Therefore, CCR5 has been recognized as a potential therapeutic target for cancer. AREAS COVERED In this review, we focus on the role of CCR5 in cancer progression and discuss its impact and potential as a therapeutic target for cancer. EXPERT OPINION Beyond immune-checkpoint inhibitors, potentially synergistic immune-modulatory drugs such as CCR5 antagonists are a promising approach to enlarge our treatment armamentarium against cancer.
Collapse
Affiliation(s)
- Hossein Hemmatazad
- Department of Radiation Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Martin D Berger
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
9
|
Kim ES, Nam SM, Song HK, Lee S, Kim K, Lim HK, Lee H, Kang KT, Kwon YJ, Chun YJ, Park SY, Jung J, Moon A. CCL8 mediates crosstalk between endothelial colony forming cells and triple-negative breast cancer cells through IL-8, aggravating invasion and tumorigenicity. Oncogene 2021; 40:3245-3259. [PMID: 33833397 DOI: 10.1038/s41388-021-01758-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 03/02/2021] [Accepted: 03/15/2021] [Indexed: 02/01/2023]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive type of breast cancer with a poor prognosis for which no effective therapeutic measures are currently available. The present study aimed to investigate whether interactions with endothelial colony-forming cells (ECFCs) promote aggressive progression of TNBC cells. Herein, using an indirect co-culture system, we showed that co-culture increased the invasive and migratory phenotypes of both MDA-MB-231 TNBC cells and ECFCs. Through a cytokine antibody array and RT-PCR analysis, we revealed that co-culture markedly induced secretion of the chemokine C-C motif ligand (CCL)8 from ECFCs and that of interleukin (IL)-8 from MDA-MB-231 cells. CCL8 was crucial for ECFC-induced IL-8 secretion and invasion of MDA-MB-231 cells as well as for MDA-MB-231-enhanced MMP-2 secretion and angiogenesis of ECFCs. We suggest c-Jun as a transcription factor for CCL8-induced IL-8 expression in MDA-MB-231 cells. IL-8 was important for co-culture-induced CCL8 and MMP-2 upregulation and invasion of ECFCs. Notably, our findings reveal a positive feedback loop between CCL8 and IL-8, which contributes to the aggressive phenotypes of both ECFC and TNBC cells. Using an MDA-MB-231 cell-based xenograft model, we show that tumor growth and metastasis are increased by co-injected ECFCs in vivo. Increased expression of IL-8 was observed in tissues with bone metastases in mice injected with conditioned media from co-cultured cells. High IL-8 levels are correlated with poor recurrence-free survival in TNBC patients. Together, these results suggest that CCL8 and IL-8 mediate the crosstalk between ECFCs and TNBC, leading to aggravation of tumorigenicity in TNBC.
Collapse
Affiliation(s)
- Eun-Sook Kim
- Duksung Innovative Drug Center, College of Pharmacy, Duksung Women's University, Seoul, Korea
| | - Su-Min Nam
- Duksung Innovative Drug Center, College of Pharmacy, Duksung Women's University, Seoul, Korea
| | - Hye Kyung Song
- College of Chemistry, Duksung Women's University, Seoul, Korea
| | - Seungeun Lee
- Duksung Innovative Drug Center, College of Pharmacy, Duksung Women's University, Seoul, Korea
| | - Kyoungmee Kim
- Duksung Innovative Drug Center, College of Pharmacy, Duksung Women's University, Seoul, Korea
| | - Hyun Kyung Lim
- Duksung Innovative Drug Center, College of Pharmacy, Duksung Women's University, Seoul, Korea
| | - Hyunsook Lee
- Duksung Innovative Drug Center, College of Pharmacy, Duksung Women's University, Seoul, Korea
| | - Kyu-Tae Kang
- Duksung Innovative Drug Center, College of Pharmacy, Duksung Women's University, Seoul, Korea
| | - Yeo-Jung Kwon
- College of Pharmacy, Chung-Ang University, Seoul, Korea
| | | | - So Yeon Park
- Department of Pathology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Joohee Jung
- Duksung Innovative Drug Center, College of Pharmacy, Duksung Women's University, Seoul, Korea.
| | - Aree Moon
- Duksung Innovative Drug Center, College of Pharmacy, Duksung Women's University, Seoul, Korea.
| |
Collapse
|
10
|
Wang N, Wang S, Wang X, Zheng Y, Yang B, Zhang J, Pan B, Gao J, Wang Z. Research trends in pharmacological modulation of tumor-associated macrophages. Clin Transl Med 2021; 11:e288. [PMID: 33463063 PMCID: PMC7805405 DOI: 10.1002/ctm2.288] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 12/27/2020] [Accepted: 12/29/2020] [Indexed: 02/06/2023] Open
Abstract
As one of the most abundant immune cell populations in the tumor microenvironment (TME), tumor-associated macrophages (TAMs) play important roles in multiple solid malignancies, including breast cancer, prostate cancer, liver cancer, lung cancer, ovarian cancer, gastric cancer, pancreatic cancer, and colorectal cancer. TAMs could contribute to carcinogenesis, neoangiogenesis, immune-suppressive TME remodeling, cancer chemoresistance, recurrence, and metastasis. Therefore, reprogramming of the immune-suppressive TAMs by pharmacological approaches has attracted considerable research attention in recent years. In this review, the promising pharmaceutical targets, as well as the existing modulatory strategies of TAMs were summarized. The chemokine-chemokine receptor signaling, tyrosine kinase receptor signaling, metabolic signaling, and exosomal signaling have been highlighted in determining the biological functions of TAMs. Besides, both preclinical research and clinical trials have suggested the chemokine-chemokine receptor blockers, tyrosine kinase inhibitors, bisphosphonates, as well as the exosomal or nanoparticle-based targeting delivery systems as the promising pharmacological approaches for TAMs deletion or reprogramming. Lastly, the combined therapies of TAMs-targeting strategies with traditional treatments or immunotherapies as well as the exosome-like nanovesicles for cancer therapy are prospected.
Collapse
Affiliation(s)
- Neng Wang
- The Research Center for Integrative MedicineSchool of Basic Medical SciencesGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Shengqi Wang
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Xuan Wang
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Yifeng Zheng
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Bowen Yang
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Juping Zhang
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Bo Pan
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Jianli Gao
- Academy of Traditional Chinese MedicineZhejiang Chinese Medical UniversityHangzhouZhejiangChina
| | - Zhiyu Wang
- The Research Center for Integrative MedicineSchool of Basic Medical SciencesGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| |
Collapse
|
11
|
Antineoplastic effects of targeting CCR5 and its therapeutic potential for colorectal cancer liver metastasis. J Cancer Res Clin Oncol 2020; 147:73-91. [PMID: 32902795 PMCID: PMC7810651 DOI: 10.1007/s00432-020-03382-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 09/01/2020] [Indexed: 12/12/2022]
Abstract
Purpose Liver metastasis is observed in up to 50% of colorectal cancer (CRC) patients. Available treatment options are limited and disease recurrence is often. Chemokine receptor 5 (CCR5) has attracted attention as novel therapeutic target for treating cancers. In this study, we reinforced the importance of CCR5 as therapeutic target in CRC and its liver metastasis by applying in vitro, in vivo and clinical investigations. Methods By targeting CCR5 via siRNAs or an FDA approved antagonist (maraviroc), we investigated the ensuing antineoplastic effects in three CRC cell lines. An animal model for CRC liver metastasis was used to evaluate time-dependent expressional modulation of the CCR5 axis by cDNA microarray. The model was also used to evaluate the in vivo efficacy of targeting CCR5 by maraviroc. Circulatory and tumor associated levels of CCR5 and its cognate ligands (CCL3, CCL4, CCL5) were analyzed by ELISA, qRT-PCR and immunohistochemistry. Results Targeting the CCR5 inhibited proliferative, migratory and clonogenic properties and interfered with cell cycle-related signaling cascades. In vivo findings showed significant induction of the CCR5 axis during the early liver colonization phase. Treatment with maraviroc significantly inhibited CRC liver metastasis in the animal model. Differential expression profiles of circulatory and tumor associated CCR5/ligands were observed in CRC patients and healthy controls. Conclusion The findings indicate that targeting the CCR5 axis can be an effective strategy for treating CRC liver metastasis. Electronic supplementary material The online version of this article (10.1007/s00432-020-03382-9) contains supplementary material, which is available to authorized users.
Collapse
|
12
|
The CCL5/CCR5 Axis in Cancer Progression. Cancers (Basel) 2020; 12:cancers12071765. [PMID: 32630699 PMCID: PMC7407580 DOI: 10.3390/cancers12071765] [Citation(s) in RCA: 198] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/25/2020] [Accepted: 06/30/2020] [Indexed: 02/07/2023] Open
Abstract
Tumor cells can “hijack” chemokine networks to support tumor progression. In this context, the C-C chemokine ligand 5/C-C chemokine receptor type 5 (CCL5/CCR5) axis is gaining increasing attention, since abnormal expression and activity of CCL5 and its receptor CCR5 have been found in hematological malignancies and solid tumors. Numerous preclinical in vitro and in vivo studies have shown a key role of the CCL5/CCR5 axis in cancer, and thus provided the rationale for clinical trials using the repurposed drug maraviroc, a CCR5 antagonist used to treat HIV/AIDS. This review summarizes current knowledge on the role of the CCL5/CCR5 axis in cancer. First, it describes the involvement of the CCL5/CCR5 axis in cancer progression, including autocrine and paracrine tumor growth, ECM (extracellular matrix) remodeling and migration, cancer stem cell expansion, DNA damage repair, metabolic reprogramming, and angiogenesis. Then, it focuses on individual hematological and solid tumors in which CCL5 and CCR5 have been studied preclinically. Finally, it discusses clinical trials of strategies to counteract the CCL5/CCR5 axis in different cancers using maraviroc or therapeutic monoclonal antibodies.
Collapse
|
13
|
Reyes ME, de La Fuente M, Hermoso M, Ili CG, Brebi P. Role of CC Chemokines Subfamily in the Platinum Drugs Resistance Promotion in Cancer. Front Immunol 2020; 11:901. [PMID: 32499779 PMCID: PMC7243460 DOI: 10.3389/fimmu.2020.00901] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 04/20/2020] [Indexed: 12/11/2022] Open
Abstract
Cancer is a significant medical issue, being one of the main causes of mortality around the world. The therapies for this pathology depend on the stage in which the cancer is found, but it is usually diagnosed at an advanced stage in which the treatment is chemotherapy. Platinum drugs are among the most commonly used in therapy, unfortunately, one of the main obstacles to this treatment is the development of chemoresistance, which is the ability of cancer cells to evade the effects of drugs. Although some molecular mechanisms involved in resistance to platinum drugs are described, elucidation is still required of others. Secretion of inflammatory mediators such as cytokines and chemokines, by tumor microenvironment components or tumor cells, show direct influence on proliferation, metastasis and progression of cancer and are related to chemoresistance and poor prognosis. In this review, the general mechanisms associated with resistance to platinum drugs, inflammation on cancer development and chemoresistance in various types of cancer will be approached with special emphasis on the current history of CC chemokines subfamily-mediated chemoresistance.
Collapse
Affiliation(s)
- Maria E. Reyes
- Laboratorio de Biología Integrativa (LIBi), Centro de Excelencia en Medicina Traslacional-Scientific and Technological Bioresource Nucleus (CEMT-BIOREN), Universidad de la Frontera, Temuco, Chile
| | - Marjorie de La Fuente
- Laboratorio de Inmunidad Innata, Programa de Inmunología, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Dirección Académica, Clínica Las Condes, Santiago, Chile
| | - Marcela Hermoso
- Laboratorio de Inmunidad Innata, Programa de Inmunología, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Carmen G. Ili
- Laboratorio de Biología Integrativa (LIBi), Centro de Excelencia en Medicina Traslacional-Scientific and Technological Bioresource Nucleus (CEMT-BIOREN), Universidad de la Frontera, Temuco, Chile
| | - Priscilla Brebi
- Laboratorio de Biología Integrativa (LIBi), Centro de Excelencia en Medicina Traslacional-Scientific and Technological Bioresource Nucleus (CEMT-BIOREN), Universidad de la Frontera, Temuco, Chile
| |
Collapse
|
14
|
Kaushik I, Ramachandran S, Prasad S, Srivastava SK. Drug rechanneling: A novel paradigm for cancer treatment. Semin Cancer Biol 2020; 68:279-290. [PMID: 32437876 DOI: 10.1016/j.semcancer.2020.03.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 01/15/2020] [Accepted: 03/18/2020] [Indexed: 12/13/2022]
Abstract
Cancer continues to be one of the leading contributors towards global disease burden. According to NIH, cancer incidence rate per year will increase to 23.6 million by 2030. Even though cancer continues to be a major proportion of the disease burden worldwide, it has the lowest clinical trial success rate amongst other diseases. Hence, there is an unmet need for novel, affordable and effective anti-neoplastic medications. As a result, a growing interest has sparkled amongst researchers towards drug repurposing. Drug repurposing follows the principle of polypharmacology, which states, "any drug with multiple targets or off targets can present several modes of action". Drug repurposing also known as drug rechanneling, or drug repositioning is an economic and reliable approach that identifies new disease treatment of already approved drugs. Repurposing guarantees expedited access of drugs to the patients as these drugs are already FDA approved and their safety and toxicity profile is completely established. Epidemiological studies have identified the decreased occurrence of oncological or non-oncological conditions in patients undergoing treatment with FDA approved drugs. Data from multiple experimental studies and clinical observations have depicted that several non-neoplastic drugs have potential anticancer activity. In this review, we have summarized the potential anti-cancer effects of anti-psychotic, anti-malarial, anti-viral and anti-emetic drugs with a brief overview on their mechanism and pathways in different cancer types. This review highlights promising evidences for the repurposing of drugs in oncology.
Collapse
Affiliation(s)
- Itishree Kaushik
- Department of Immunotherapeutics and Biotechnology, and Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Sharavan Ramachandran
- Department of Immunotherapeutics and Biotechnology, and Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Sahdeo Prasad
- Department of Immunotherapeutics and Biotechnology, and Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Sanjay K Srivastava
- Department of Immunotherapeutics and Biotechnology, and Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA.
| |
Collapse
|
15
|
Ellwanger JH, Kaminski VDL, Rodrigues AG, Kulmann-Leal B, Chies JAB. CCR5 and CCR5Δ32 in bacterial and parasitic infections: Thinking chemokine receptors outside the HIV box. Int J Immunogenet 2020; 47:261-285. [PMID: 32212259 DOI: 10.1111/iji.12485] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/02/2020] [Accepted: 03/06/2020] [Indexed: 12/14/2022]
Abstract
The CCR5 molecule was reported in 1996 as the main HIV-1 co-receptor. In that same year, the CCR5Δ32 genetic variant was described as a strong protective factor against HIV-1 infection. These findings led to extensive research regarding the CCR5, culminating in critical scientific advances, such as the development of CCR5 inhibitors for the treatment of HIV infection. Recently, the research landscape surrounding CCR5 has begun to change. Different research groups have realized that, since CCR5 has such important effects in the chemokine system, it could also affect other different physiological systems. Therefore, the effect of reduced CCR5 expression due to the presence of the CCR5Δ32 variant began to be further studied. Several studies have investigated the role of CCR5 and the impacts of CCR5Δ32 on autoimmune and inflammatory diseases, various types of cancer, and viral diseases. However, the role of CCR5 in diseases caused by bacteria and parasites is still poorly understood. Therefore, the aim of this article is to review the role of CCR5 and the effects of CCR5Δ32 on bacterial (brucellosis, osteomyelitis, pneumonia, tuberculosis and infection by Chlamydia trachomatis) and parasitic infections (toxoplasmosis, leishmaniasis, Chagas disease and schistosomiasis). Basic information about each of these infections was also addressed. The neglected role of CCR5 in fungal disease and emerging studies regarding the action of CCR5 on regulatory T cells are briefly covered in this review. Considering the "renaissance of CCR5 research," this article is useful for updating researchers who develop studies involving CCR5 and CCR5Δ32 in different infectious diseases.
Collapse
Affiliation(s)
- Joel Henrique Ellwanger
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - Valéria de Lima Kaminski
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - Andressa Gonçalves Rodrigues
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - Bruna Kulmann-Leal
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - José Artur Bogo Chies
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| |
Collapse
|
16
|
Bansal SS, Ismahil MA, Goel M, Zhou G, Rokosh G, Hamid T, Prabhu SD. Dysfunctional and Proinflammatory Regulatory T-Lymphocytes Are Essential for Adverse Cardiac Remodeling in Ischemic Cardiomyopathy. Circulation 2019; 139:206-221. [PMID: 30586716 DOI: 10.1161/circulationaha.118.036065] [Citation(s) in RCA: 198] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Heart failure (HF) is a state of inappropriately sustained inflammation, suggesting the loss of normal immunosuppressive mechanisms. Regulatory T-lymphocytes (Tregs) are considered key suppressors of immune responses; however, their role in HF is unknown. We hypothesized that Tregs are dysfunctional in ischemic cardiomyopathy and HF, and they promote immune activation and left ventricular (LV) remodeling. METHODS Adult male wild-type C57BL/6 mice, Foxp3-diphtheria toxin receptor transgenic mice, and tumor necrosis factor (TNF) α receptor-1 (TNFR1)-/- mice underwent nonreperfused myocardial infarction to induce HF or sham operation. LV remodeling was assessed by echocardiography as well as histological and molecular phenotyping. Alterations in Treg profile and function were examined by flow cytometry, immunostaining, and in vitro cell assays. RESULTS Compared with wild-type sham mice, CD4+Foxp3+ Tregs in wild-type HF mice robustly expanded in the heart, circulation, spleen, and lymph nodes in a phasic manner after myocardial infarction, beyond the early phase of wound healing, and exhibited proinflammatory T helper 1-type features with interferon-γ, TNFα, and TNFR1 expression, loss of immunomodulatory capacity, heightened proliferation, and potentiated antiangiogenic and profibrotic properties. Selective Treg ablation in Foxp3-diphtheria toxin receptor mice with ischemic cardiomyopathy reversed LV remodeling and dysfunction, alleviating hypertrophy and fibrosis, while suppressing circulating CD4+ T cells and systemic inflammation and enhancing tissue neovascularization. Tregs reconstituted after ablation exhibited restoration of immunosuppressive capacity and normalized TNFR1 expression. Treg dysfunction was also tightly coupled to Treg-endothelial cell contact- and TNFR1-dependent inhibition of angiogenesis and the mobilization and tissue infiltration of CD34+Flk1+ circulating angiogenic cells in a C-C chemokine ligand 5/C-C chemokine receptor 5-dependent manner. Anti-CD25-mediated Treg depletion in wild-type mice imparted similar benefits on LV remodeling, circulating angiogenic cells, and tissue neovascularization. CONCLUSIONS Proinflammatory and antiangiogenic Tregs play an essential pathogenetic role in chronic ischemic HF to promote immune activation and pathological LV remodeling. The restoration of normal Treg function may be a viable approach to therapeutic immunomodulation in this disease.
Collapse
Affiliation(s)
- Shyam S Bansal
- Division of Cardiovascular Disease and Comprehensive Cardiovascular Center, University of Alabama at Birmingham (S.S.B., M.A.I., M.G., G.Z., G.R., T.H., S.D.P.).,The current affiliation for S.S.B. is Department of Physiology and Cell Biology, The Dorothy M Davis Heart & Lung Research Institute, Ohio State University Wexner Medical Center, Columbus
| | - Mohamed Ameen Ismahil
- Division of Cardiovascular Disease and Comprehensive Cardiovascular Center, University of Alabama at Birmingham (S.S.B., M.A.I., M.G., G.Z., G.R., T.H., S.D.P.)
| | - Mehak Goel
- Division of Cardiovascular Disease and Comprehensive Cardiovascular Center, University of Alabama at Birmingham (S.S.B., M.A.I., M.G., G.Z., G.R., T.H., S.D.P.)
| | - Guihua Zhou
- Division of Cardiovascular Disease and Comprehensive Cardiovascular Center, University of Alabama at Birmingham (S.S.B., M.A.I., M.G., G.Z., G.R., T.H., S.D.P.)
| | - Gregg Rokosh
- Division of Cardiovascular Disease and Comprehensive Cardiovascular Center, University of Alabama at Birmingham (S.S.B., M.A.I., M.G., G.Z., G.R., T.H., S.D.P.)
| | - Tariq Hamid
- Division of Cardiovascular Disease and Comprehensive Cardiovascular Center, University of Alabama at Birmingham (S.S.B., M.A.I., M.G., G.Z., G.R., T.H., S.D.P.)
| | - Sumanth D Prabhu
- Division of Cardiovascular Disease and Comprehensive Cardiovascular Center, University of Alabama at Birmingham (S.S.B., M.A.I., M.G., G.Z., G.R., T.H., S.D.P.).,Medical Service, Birmingham VAMC, AL (S.D.P.)
| |
Collapse
|
17
|
Suarez-Carmona M, Chaorentong P, Kather JN, Rothenheber R, Ahmed A, Berthel A, Heinzelmann A, Moraleda R, Valous NA, Kosaloglu Z, Eurich R, Wolf J, Grauling-Halama S, Hundemer M, Lasitschka F, Klupp F, Kahlert C, Ulrich A, Schneider M, Falk C, Jäger D, Zoernig I, Halama N. CCR5 status and metastatic progression in colorectal cancer. Oncoimmunology 2019; 8:e1626193. [PMID: 31428524 PMCID: PMC6685512 DOI: 10.1080/2162402x.2019.1626193] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 05/27/2019] [Accepted: 05/29/2019] [Indexed: 12/18/2022] Open
Abstract
Multiple reports have highlighted the importance of the local immunological cellular composition (i.e. the density of effector T cells and macrophage polarization state) in predicting clinical outcome in advanced metastatic stage of colorectal cancer. However, in spite of the general association between a high effector T cell density and improved outcome, our recent work has revealed a specific lymphocyte-driven cancer cell-supporting signal. Indeed, lymphocyte-derived CCL5 supports CCR5-positive tumor cell proliferation and thereby fosters tumor growth in metastatic liver lesions. Upon systematic analysis of CCR5 expression by tumor cells using immunohistochemistry, we observed that the intensity of CCR5 increases with primary tumor size and peaks in T4 tumors. In liver metastases however, though CCR5 expression intensity is globally heightened compared to primary tumors, alterations in the expression patterns appear, leading to “patchiness” of the stain. CCR5 patchiness is, therefore, a signature of liver metastases in our cohort (n = 97 specimens) and relates to globally decreased expression intensity, but does not influence the extent of the response to CCR5 inhibitor Maraviroc in patients. Moreover, CCR5 patchiness relates to a poor immune landscape characterized by a low cytotoxic-to-regulatory T cell ratio at the invasive margin and enriched cellular and molecular markers of macrophage M2 polarization. Finally, because higher numbers of PD-1- and CTLA-4-positive cells surround tumors with patchy CCR5 expression, one can speculate that these tumors potentially respond to immune checkpoint blockade. This hypothesis is corroborated by the prolonged disease-free survival and disease-specific survival observed in patients with low gene expression of CCR5 in metastases from two publically available cohorts. These observations highlight the complex role of the CCL5-CCR5 axis in CRC metastatic progression and warrant further investigations.
Collapse
Affiliation(s)
- Meggy Suarez-Carmona
- Department of Translational Immunotherapy, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Medical oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.,Helmholtz Center for Translational Oncology (HITRON), Mainz, Germany
| | - Pornpimol Chaorentong
- Clinical Cooperation Unit Applied Tumor Immunity, National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany
| | - Jakob Nikolas Kather
- Medical oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.,Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Rebecca Rothenheber
- Medical oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Azaz Ahmed
- Medical oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Anna Berthel
- Medical oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Anita Heinzelmann
- Medical oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Rodrigo Moraleda
- Medical oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit Applied Tumor Immunity, National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany
| | - Nektarios A Valous
- Clinical Cooperation Unit Applied Tumor Immunity, National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany
| | - Zeynep Kosaloglu
- Clinical Cooperation Unit Applied Tumor Immunity, National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany
| | - Rosa Eurich
- Medical oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Jana Wolf
- Medical oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Silke Grauling-Halama
- Department of Translational Immunotherapy, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Medical oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.,Helmholtz Center for Translational Oncology (HITRON), Mainz, Germany
| | - Michael Hundemer
- Department of Hematology, University Hospital Heidelberg, Heidelberg, Germany
| | - Felix Lasitschka
- Institute for Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Fee Klupp
- Department of Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Christoph Kahlert
- Department of Surgery, University Hospital Dresden, Dresden, Germany
| | - Alexis Ulrich
- Department of Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Martin Schneider
- Department of Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Christine Falk
- Institute of Transplant Immunology, Integrated Research and Treatment Center Transplantation, Hannover Medical School Hannover, Hanover, Germany
| | - Dirk Jäger
- Medical oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.,Helmholtz Center for Translational Oncology (HITRON), Mainz, Germany.,Department of Internal Medicine VI, University Hospital Heidelberg, Heidelberg, Germany
| | - Inka Zoernig
- Medical oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit Applied Tumor Immunity, National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany.,Department of Internal Medicine VI, University Hospital Heidelberg, Heidelberg, Germany
| | - Niels Halama
- Department of Translational Immunotherapy, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Medical oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.,Helmholtz Center for Translational Oncology (HITRON), Mainz, Germany.,Clinical Cooperation Unit Applied Tumor Immunity, National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany.,Department of Internal Medicine VI, University Hospital Heidelberg, Heidelberg, Germany.,Institute of Immunology, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
18
|
Guo P, Bi K, Lu Z, Wang K, Xu Y, Wu H, Cao Y, Jiang H. CCR5/CCR5 ligand-induced myeloid-derived suppressor cells are related to the progression of endometriosis. Reprod Biomed Online 2019; 39:704-711. [PMID: 31427176 DOI: 10.1016/j.rbmo.2019.05.014] [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] [Received: 02/21/2019] [Revised: 04/25/2019] [Accepted: 05/22/2019] [Indexed: 11/19/2022]
Abstract
RESEARCH QUESTION Immunological disorders have been reported to promote the progression of endometriosis. Several recent studies have shown that myeloid-derived suppressor cells (MDSC) drive the progression of endometriosis. The aim of this case-control study was to test whether CCR5 and its ligands drive MDSC accumulation and play a role in the progression of endometriosis. DESIGN Thirty-six endometriosis patients and 20 controls were recruited. All subjects underwent laparoscopy. An ELISA kit was used to define CCR5 ligands in plasma and peritoneal fluid from endometriosis patients; flow cytometry was then used to characterize CCR5+MDSC in peripheral blood and peritoneal fluid. RESULTS Data showed that endometriosis patients displayed a significantly higher production of plasma CCL3 (P = 0.046) and peritoneal fluid CCL3/5 (P = 0.042/0.036) compared with those from the uterine leiomyoma group. Furthermore, the concentrations of peritoneal fluid CCL5 were elevated in late stage patients compared with those from the uterine leiomyoma group. Accumulation of blood CCR5+Mo-MDSC was detected in endometriosis patients compared with those from both the ovarian dermoid cysts and uterine leiomyoma groups. Endometriosis patients also showed an elevation of CCR5+MDSC and CCR5+Mo-MDSC in peritoneal fluid samples compared with uterine leiomyoma samples. It was also found that enrichment of CCR5+MDSC (r = 0.6807; P < 0.0001) and CCR5+Mo-MDSC (r = 0.6893; P < 0.0001) were correlated with enhanced production of CCL5 in peritoneal fluid from endometriosis patients. CONCLUSIONS This study showed that CCR5 and its ligands could drive the progression of endometriosis by enhancing the accumulation of MDSC. These findings might produce a promising treatment that targets CCR5+MDSC for endometriosis patients.
Collapse
Affiliation(s)
- Peipei Guo
- Reproductive Medicine Centre, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, PR China; Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, PR China
| | - Kaihuan Bi
- Reproductive Medicine Centre, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, PR China
| | - Zhimin Lu
- Reproductive Medicine Centre, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, PR China; Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Centre, Anhui Medical University, Hefei, PR China
| | - Kangxia Wang
- Reproductive Medicine Centre, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, PR China; Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, PR China
| | - Yuping Xu
- Reproductive Medicine Centre, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, PR China; Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Centre, Anhui Medical University, Hefei, PR China
| | - Huan Wu
- Reproductive Medicine Centre, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, PR China; Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Centre, Anhui Medical University, Hefei, PR China
| | - Yunxia Cao
- Reproductive Medicine Centre, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, PR China; Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, PR China; Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Centre, Anhui Medical University, Hefei, PR China.
| | - Huanhuan Jiang
- Reproductive Medicine Centre, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, PR China; Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, PR China; Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Centre, Anhui Medical University, Hefei, PR China.
| |
Collapse
|
19
|
Pervaiz A, Zepp M, Mahmood S, Ali DM, Berger MR, Adwan H. CCR5 blockage by maraviroc: a potential therapeutic option for metastatic breast cancer. Cell Oncol (Dordr) 2019; 42:93-106. [PMID: 30456574 DOI: 10.1007/s13402-018-0415-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/12/2018] [Indexed: 01/01/2023] Open
Abstract
PURPOSE Bone metastasis is observed in up to 70% of breast cancer patients. The currently available treatment options are palliative in nature. Chemokine receptor 5 (CCR5) has gained attention as therapeutic target in various malignancies. Here, we investigated the effects of targeting CCR5 by its antagonist maraviroc in metastatic breast cancer cells. METHODS In response to maraviroc exposure, cytotoxicity was assessed using an MTT proliferation assay, whereas the effects on colony formation and migration were assessed using colony formation, transwell chamber migration and scratch wound healing assays, respectively. Apoptosis-related activities were investigated using nuclear staining, annexin-V FITC staining and Western blotting. Cell cycle changes were analysed using flow cytometry and qRT-PCR for cell cycle relevant genes. A nude rat model for breast cancer bone metastasis was used to evaluate the in vivo efficacy of CCR5 targeting by maraviroc. Circulatory levels of the three cognate ligands for CCR5 (CCL3, CCL4, CCL5) were analysed in sera of breast cancer patients using ELISA. RESULTS We found that blockade of CCR5 attenuated the proliferation, colony formation and migration of metastatic breast cancer cells, and induced apoptosis and arrest in the G1 phase of the cell cycle. Expression profiling highlighted the involvement of cell cycle related signalling cascades. We also found that treatment with maraviroc significantly inhibited bone metastasis in nude rats implanted with MDA-MB-231 breast cancer cells. Finally, we found that the circulatory levels of three cognate ligands for the CCR5 receptor varied between breast cancer patients and healthy controls. CONCLUSION Our findings indicate that targeting CCR5 may be an effective strategy to combat breast cancer bone metastasis.
Collapse
Affiliation(s)
- Asim Pervaiz
- Institute of Biomedical and Allied Health Sciences, University of Health Sciences, Lahore, Pakistan
- Toxicology and Chemotherapy Unit, German Cancer Research Centre (DKFZ), Im Neuenheimer Feld 581, 69120, Heidelberg, Germany
| | - Michael Zepp
- Toxicology and Chemotherapy Unit, German Cancer Research Centre (DKFZ), Im Neuenheimer Feld 581, 69120, Heidelberg, Germany
| | - Saqib Mahmood
- Institute of Biomedical and Allied Health Sciences, University of Health Sciences, Lahore, Pakistan
| | - Doaa Mohamed Ali
- Toxicology and Chemotherapy Unit, German Cancer Research Centre (DKFZ), Im Neuenheimer Feld 581, 69120, Heidelberg, Germany
| | - Martin R Berger
- Toxicology and Chemotherapy Unit, German Cancer Research Centre (DKFZ), Im Neuenheimer Feld 581, 69120, Heidelberg, Germany.
| | - Hassan Adwan
- Toxicology and Chemotherapy Unit, German Cancer Research Centre (DKFZ), Im Neuenheimer Feld 581, 69120, Heidelberg, Germany
- German University of Cairo, Cairo, Egypt
| |
Collapse
|
20
|
Liu J, Wang C, Ma X, Tian Y, Wang C, Fu Y, Luo Y. High expression of CCR5 in melanoma enhances epithelial-mesenchymal transition and metastasis via TGFβ1. J Pathol 2019; 247:481-493. [PMID: 30474221 DOI: 10.1002/path.5207] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 10/16/2018] [Accepted: 10/30/2018] [Indexed: 02/06/2023]
Abstract
Chemokine receptors are highly expressed in various cancers and play crucial roles in tumor progression. However, their expression patterns and functions in melanoma are unclear. The present study aimed to identify the chemokine receptors that play critical roles in melanoma progression and unravel the underlying molecular mechanisms. We found that CCR5 was more abundant in melanoma cells than normal cells and was positively associated with tumor malignancy in clinical patients. Animal experiments suggested that CCR5 deficiency in B16/F10 or A375 cells suppressed primary tumor growth and lung metastasis, whereas CCR5 overexpression in B16/F0 cells enhanced primary tumor growth and lung metastasis. CCR5 played a critical role in proliferation and migration of melanoma cells in vitro. Importantly, CCR5 was required for maintenance of the mesenchymal phenotype of metastatic melanoma cells. Mechanistically, CCR5 positively regulated expression of TGFβ1, which in turn induced epithelial-mesenchymal transition and migration via PI3K/AKT/GSK3β signaling. Collectively, our results establish a critical role of CCR5 expressed by melanoma cells in cancer progression and reveal the novel mechanisms controlling this process, which suggests the prognostic value of CCR5 in melanoma patients and provides novel insights into CCR5-targeted strategies for melanoma treatment. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Jie Liu
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing, PR China.,Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing, PR China.,Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, PR China
| | - Caihong Wang
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing, PR China.,Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing, PR China.,Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, PR China
| | - Xuhui Ma
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing, PR China.,Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing, PR China.,Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, PR China
| | - Yang Tian
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing, PR China.,Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing, PR China.,Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, PR China
| | - Chunying Wang
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing, PR China.,Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing, PR China.,Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, PR China
| | - Yan Fu
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing, PR China.,Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing, PR China.,Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, PR China
| | - Yongzhang Luo
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing, PR China.,Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing, PR China.,Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, PR China
| |
Collapse
|
21
|
Lin HY, Ho Y, Liu HL. Structure-Based Pharmacophore Modeling to Discover Novel CCR5 Inhibitors for HIV-1/Cancers Therapy. ACTA ACUST UNITED AC 2019. [DOI: 10.4236/jbise.2019.121002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
22
|
Casagrande N, Borghese C, Visser L, Mongiat M, Colombatti A, Aldinucci D. CCR5 antagonism by maraviroc inhibits Hodgkin lymphoma microenvironment interactions and xenograft growth. Haematologica 2018; 104:564-575. [PMID: 30309853 PMCID: PMC6395337 DOI: 10.3324/haematol.2018.196725] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 10/09/2018] [Indexed: 12/14/2022] Open
Abstract
Classic Hodgkin lymphoma tumor cells express a functional CCR5 receptor, and tumor tissues express high CCL5 levels, suggesting that CCL5-CCR5 signaling is involved in tumor-microenvironment formation and tumor growth. Using the CCR5 antagonist, maraviroc, and a neutralizing anti-CCL5 antibody, we found that CCL5 secreted by classic Hodgkin lymphoma cells recruited mesenchymal stromal cells and monocytes. The “education” of mesenchymal stromal cells by tumor cell-conditioned medium enhanced mesenchymal stromal cells’ proliferation and CCL5 secretion. In turn, educated mesenchymal stromal cell-conditioned medium increased the clonogenic growth of tumor cells and monocyte migration, but these effects were reduced by maraviroc. Monocyte education by tumor cell-conditioned medium induced their growth and reprogrammed them towards immunosuppressive tumor-associated macrophages that expressed IDO and PD-L1 and secreted IL-10, CCL17 and TGF-β. Educated monocyte-conditioned medium slowed the growth of phytohemagglutinin-activated lymphocytes. Maraviroc decreased tumor cell growth and synergized with doxorubicin and brentuximab vedotin. A three-dimensional heterospheroid assay showed that maraviroc counteracted both the formation and viability of heterospheroids generated by co-cultivation of tumor cells with mesenchymal stromal cells and monocytes. In mice bearing tumor cell xenografts, maraviroc reduced tumor growth by more than 50% and inhibited monocyte accumulation, without weight loss. Finally, in classic Hodgkin lymphoma human tumor tissues, CCL5 and CD68 expression correlated positively, and patients with high CCL5 levels had poor prognosis. In conclusion, since the present challenges are to find molecules counteracting the formation of the immunosuppressive tumor microenvironment or new, less toxic drug combinations, the repurposed drug maraviroc may represent a new opportunity for classic Hodgkin lym phoma treatment.
Collapse
Affiliation(s)
- Naike Casagrande
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Italy
| | - Cinzia Borghese
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Italy
| | - Lydia Visser
- Department of Pathology and Medical Biology, University Medical Center Groningen (UMcG), the Netherlands
| | - Maurizio Mongiat
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Italy
| | - Alfonso Colombatti
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Italy
| | - Donatella Aldinucci
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Italy
| |
Collapse
|
23
|
Rafat M, Aguilera TA, Vilalta M, Bronsart LL, Soto LA, von Eyben R, Golla MA, Ahrari Y, Melemenidis S, Afghahi A, Jenkins MJ, Kurian AW, Horst KC, Giaccia AJ, Graves EE. Macrophages Promote Circulating Tumor Cell-Mediated Local Recurrence following Radiotherapy in Immunosuppressed Patients. Cancer Res 2018; 78:4241-4252. [PMID: 29880480 PMCID: PMC6072588 DOI: 10.1158/0008-5472.can-17-3623] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 04/09/2018] [Accepted: 05/25/2018] [Indexed: 01/07/2023]
Abstract
Although radiotherapy (RT) decreases the incidence of locoregional recurrence in breast cancer, patients with triple-negative breast cancer (TNBC) have increased risk of local recurrence following breast-conserving therapy. The relationship between RT and local recurrence is unknown. Here, we tested the hypothesis that recurrence in some instances is due to the attraction of circulating tumor cells to irradiated tissues. To evaluate the effect of absolute lymphocyte count on local recurrence after RT in patients with TNBC, we analyzed radiation effects on tumor and immune cell recruitment to tissues in an orthotopic breast cancer model. Recurrent patients exhibited a prolonged low absolute lymphocyte count when compared with nonrecurrent patients following RT. Recruitment of tumor cells to irradiated normal tissues was enhanced in the absence of CD8+ T cells. Macrophages (CD11b+F480+) preceded tumor cell infiltration and were recruited to tissues following RT. Tumor cell recruitment was mitigated by inhibiting macrophage infiltration using maraviroc, an FDA-approved CCR5 receptor antagonist. Our work poses the intriguing possibility that excessive macrophage infiltration in the absence of lymphocytes promotes local recurrence after RT. This combination thus defines a high-risk group of patients with TNBC.Significance: This study establishes the importance of macrophages in driving tumor cell recruitment to sites of local radiation therapy and suggests that this mechanism contributes to local recurrence in women with TNBC that are also immunosuppressed.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/15/4241/F1.large.jpg Cancer Res; 78(15); 4241-52. ©2018 AACR.
Collapse
Affiliation(s)
- Marjan Rafat
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Todd A Aguilera
- Department of Radiation Oncology, Harold C. Simmons Comprehensive Cancer Center, U.T. Southwestern Medical Center, Dallas, Texas
| | - Marta Vilalta
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Laura L Bronsart
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Luis A Soto
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Rie von Eyben
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Meghana A Golla
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Yasaman Ahrari
- Department of Radiation Oncology, Stanford University, Stanford, California
| | | | - Anosheh Afghahi
- Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Melissa J Jenkins
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Allison W Kurian
- Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Kathleen C Horst
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Amato J Giaccia
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Edward E Graves
- Department of Radiation Oncology, Stanford University, Stanford, California.
| |
Collapse
|
24
|
Inhibition of the CCL5/CCR5 Axis against the Progression of Gastric Cancer. Int J Mol Sci 2018; 19:ijms19051477. [PMID: 29772686 PMCID: PMC5983686 DOI: 10.3390/ijms19051477] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 05/11/2018] [Accepted: 05/14/2018] [Indexed: 12/14/2022] Open
Abstract
Despite the progress made in molecular and clinical research, patients with advanced-stage gastric cancer (GC) have a bad prognosis and very low survival rates. Furthermore, it is challenging to find the complex molecular mechanisms that are involved in the development of GC, its progression, and its resistance to therapy. The interactions of chemokines, also known as chemotactic cytokines, with their receptors regulate immune and inflammatory responses. However, updated research demonstrates that cancer cells subvert the normal chemokine role, transforming them into fundamental constituents of the tumor microenvironment (TME) with tumor-promoting effects. C-C chemokine ligand 5 (CCL5) is a chemotactic cytokine, and its expression and secretion are regulated in T cells. C-C chemokine receptor type 5 (CCR5) is expressed in T cells, macrophages, other leukocytes, and certain types of cancer cells. The interaction between CCL5 and CCR5 plays an active role in recruiting leukocytes into target sites. This review summarizes recent information on the role of the CCL5 chemokine and its receptor CCR5 in GC cell proliferation, metastasis formation, and in the building of an immunosuppressive TME. Moreover, it highlights the development of new therapeutic strategies to inhibit the CCL5/CCR5 axis in different ways and their possible clinical relevance in the treatment of GC.
Collapse
|
25
|
Wu YC, Shen YC, Chang JWC, Hsieh JJ, Chu Y, Wang CH. Autocrine CCL5 promotes tumor progression in esophageal squamous cell carcinoma in vitro. Cytokine 2018; 110:94-103. [PMID: 29705397 DOI: 10.1016/j.cyto.2018.04.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 04/03/2018] [Accepted: 04/20/2018] [Indexed: 01/04/2023]
Abstract
The pro-tumoral effects of CCL5 have been identified in numerous cancer types. We successfully cultivated 4 esophageal squamous cell carcinoma (ESCC) cell lines, including TWES-1, TWES-3 and a pair of cell lines derived from primary lesion (TWES-4PT) and metastatic lymph node (TWES-4LN) of the same patient. Whole genome screening showed that TWES-4LN expressed higher levels of CCL5 compared to that of TWES-4PT; quantification of protein secretion displayed comparable results, suggesting that CCL5 could be associated with lymph node metastasis in ESCC. CCL5 knockdown by siRNA significantly reduced basal growth rate, tumor migration and invasiveness in the paired cell lines; whereas this treatment induced cell apoptosis in TWES-1 and TWES-3. CCR5 antagonist maraviroc significantly inhibited tumor migration and invasion in the paired cell lines without affecting tumor growth. Collectively, these results suggest that CCL5 autocrine loop may promote ESCC progression; targeting the CCL5/CCR5 axis could be a potential therapeutic strategy for this deadly disease.
Collapse
Affiliation(s)
- Yi-Cheng Wu
- Division of Thoracic and Cardiovascular Surgery, Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Taoyuan 333, Taiwan
| | - Yung-Chi Shen
- Division of Hematology/Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Keelung 204, Taiwan
| | - John Wen-Cheng Chang
- Division of Hematology/Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Taoyuan 333, Taiwan
| | - Jia-Juan Hsieh
- Division of Hematology/Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Taoyuan 333, Taiwan
| | - Yen Chu
- Department of Medical Research and Development, Division of Thoracic and Cardiovascular Surgery, Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Taoyuan 333, Taiwan.
| | - Cheng-Hsu Wang
- Division of Hematology/Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Keelung 204, Taiwan.
| |
Collapse
|
26
|
Carino A, Graziosi L, D'Amore C, Cipriani S, Marchianò S, Marino E, Zampella A, Rende M, Mosci P, Distrutti E, Donini A, Fiorucci S. The bile acid receptor GPBAR1 (TGR5) is expressed in human gastric cancers and promotes epithelial-mesenchymal transition in gastric cancer cell lines. Oncotarget 2018; 7:61021-61035. [PMID: 27409173 PMCID: PMC5308633 DOI: 10.18632/oncotarget.10477] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 06/09/2016] [Indexed: 12/13/2022] Open
Abstract
GPBAR1 (also known as TGR5) is a bile acid activated receptor expressed in several adenocarcinomas and its activation by secondary bile acids increases intestinal cell proliferation. Here, we have examined the expression of GPBAR1 in human gastric adenocarcinomas and investigated whether its activation promotes the acquisition of a pro-metastatic phenotype. By immunohistochemistry and RT-PCR analysis we found that expression of GPBAR1 associates with advanced gastric cancers (Stage III-IV). GPBAR1 expression in tumors correlates with the expression of N-cadherin, a markers of epithelial-mesenchymal transition (EMT) (r=0.52; P<0.01). Expression of GPBAR1, mRNA and protein, was detected in cancer cell lines, with MKN 45 having the higher expression. Exposure of MKN45 cells to GPBAR1 ligands, TLCA, oleanolic acid or 6-ECDCA (a dual FXR and GPBAR1 ligand) increased the expression of genes associated with EMT including KDKN2A, HRAS, IGB3, MMP10 and MMP13 and downregulated the expression of CD44 and FAT1 (P<0.01 versus control cells). GPBAR1 activation in MKN45 cells associated with EGF-R and ERK1 phosphorylation. These effects were inhibited by DFN406, a GPBAR1 antagonist, and cetuximab. GPBAR1 ligands increase MKN45 migration, adhesion to peritoneum and wound healing. Pretreating MKN45 cells with TLCA increased propensity toward peritoneal dissemination in vivo. These effects were abrogated by cetuximab. In summary, we report that GPBAR1 is expressed in advanced gastric cancers and its expression correlates with markers of EMT. GPBAR1 activation in MKN45 cells promotes EMT. These data suggest that GPBAR1 antagonist might have utility in the treatment of gastric cancers.
Collapse
Affiliation(s)
- Adriana Carino
- Dipartimento di Scienze Chirurgiche e Biomediche, Università degli Studi di Perugia, Perugia, Italy
| | | | - Claudio D'Amore
- Dipartimento di Scienze Chirurgiche e Biomediche, Università degli Studi di Perugia, Perugia, Italy
| | - Sabrina Cipriani
- Dipartimento di Medicina, Università degli Studi di Perugia, Perugia, Italy
| | - Silvia Marchianò
- Dipartimento di Scienze Chirurgiche e Biomediche, Università degli Studi di Perugia, Perugia, Italy
| | | | - Angela Zampella
- Dipartimento di Farmacia, Università di Napoli, Napoli, Italy
| | - Mario Rende
- Dipartimento di Scienze Chirurgiche e Biomediche, Università degli Studi di Perugia, Perugia, Italy
| | - Paolo Mosci
- Dipartimento di Medicina Veterinaria, Università degli Studi di Perugia, Perugia, Italy
| | | | - Annibale Donini
- Dipartimento di Scienze Chirurgiche e Biomediche, Università degli Studi di Perugia, Perugia, Italy
| | - Stefano Fiorucci
- Dipartimento di Scienze Chirurgiche e Biomediche, Università degli Studi di Perugia, Perugia, Italy
| |
Collapse
|
27
|
Tanabe Y, Sasaki S, Mukaida N, Baba T. Blockade of the chemokine receptor, CCR5, reduces the growth of orthotopically injected colon cancer cells via limiting cancer-associated fibroblast accumulation. Oncotarget 2018; 7:48335-48345. [PMID: 27340784 PMCID: PMC5217021 DOI: 10.18632/oncotarget.10227] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 06/07/2016] [Indexed: 12/19/2022] Open
Abstract
We previously demonstrated that cancer-associated fibroblasts (CAFs) accumulate at tumor sites through the interaction between a chemokine, CCL3, and its receptor, CCR5, in the late phase of colitis-associated colon carcinogenesis. Here we examined the effect of a CCR5 antagonist, maraviroc, on tumor growth arising from the orthotopic injection of mouse or human colon cancer cell lines into the cecal wall by focusing on CAFs. Orthotopic injection of either cell line caused tumor formation together with leukocyte infiltration and fibroblast accumulation. Concomitant oral administration of maraviroc reduced tumor formation with few effects on leukocyte infiltration. In contrast, maraviroc reduced the intratumor number of α-smooth muscle actin-positive fibroblasts, which express epidermal growth factor, a crucial growth factor for colon cancer cell growth. These observations suggest that maraviroc or other CCR5 antagonists might act as novel anti-CRC drugs to dampen CAFs, an essential cell component for tumor progression.
Collapse
Affiliation(s)
- Yamato Tanabe
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Soichiro Sasaki
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Naofumi Mukaida
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Tomohisa Baba
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
28
|
Gao T, Shen Z, Ma C, Li Y, Kang X, Sun M. The CCL5/CCR5 Chemotactic Pathway Promotes Perineural Invasion in Salivary Adenoid Cystic Carcinoma. J Oral Maxillofac Surg 2018; 76:1708-1718. [PMID: 29549020 DOI: 10.1016/j.joms.2018.02.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 02/06/2018] [Accepted: 02/11/2018] [Indexed: 01/10/2023]
Abstract
PURPOSE Perineural invasion (PNI) is a hallmark of salivary adenoid cystic carcinoma (SACC) and represents an important risk factor for local recurrence and poor survival. However, the mechanism of PNI has yet to be explored. We sought to examine the CCL5-CCR5 ligand-receptor interaction between nerves and SACC cells. MATERIALS AND METHODS CCL5/CCR5 expression was determined by immunohistochemistry in SACC tissue specimens. The correlations between CCL5/CCR5 expression and clinicopathologic features were investigated. Dorsal root ganglia (DRG) and SACC cells cocultured in vitro were used to evaluate the effects of CCL5/CCR5 on PNI progression and pathogenesis. RESULTS CCR5 expression was significantly elevated in SACC tissues and associated with distant metastasis, PNI, and TNM grade (P < .05). DRG and SACC cells cocultured in vitro showed that the activation of the CCL5/CCR5 axis significantly increased SACC cell invasion and promoted the outgrowth of the DRG. SACC cell lines expressing CCR5 migrated in response to CCL5 derived from DRG, eventually leading to PNI. More importantly, further study showed that blocking of CCL5 or CCR5 effectively inhibited the invasive capacity and PNI activity of SACC cells (P < .05). CONCLUSIONS Our results suggest a pivotal role of CCL5/CCR5 axis in tumor-nerve interactions during PNI of SACC. The CCL5/CCR5 pathway might prove to be an attractive new target for the treatment of SACC with PNI.
Collapse
Affiliation(s)
- Tao Gao
- Attending Physician, State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases; and Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Zhiyuan Shen
- Resident, State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases; Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an, China; and Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Department of Oral and Maxillofacial surgery, Stomatological Hospital of Shandong University, Jinan, China
| | - Chao Ma
- Resident, State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases; and Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Yun Li
- Resident, State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases; and Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Xiangfeng Kang
- Resident, Department of Pediatrics, The First Hospital of Yulin, Shaanxi, China
| | - Moyi Sun
- Professor, State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases; and Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
29
|
Ryu H, Baek SW, Moon JY, Jo IS, Kim N, Lee HJ. C-C motif chemokine receptors in gastric cancer. Mol Clin Oncol 2018; 8:3-8. [PMID: 29285394 PMCID: PMC5738695 DOI: 10.3892/mco.2017.1470] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 06/06/2017] [Indexed: 12/20/2022] Open
Abstract
Gastric cancer is the fifth most common cancer and the third leading cause of cancer-associated mortality worldwide. Despite recent advances in molecular and clinical research, patients with gastric cancer at an advanced stage have a dismal prognosis and poor survival rates, and systemic treatment relies predominantly on traditional cytotoxic chemotherapy. To improve patients' quality of life and survival, an improved understanding of the complex molecular mechanisms involved in gastric cancer progression and treatment resistance, and of its clinical application in the development of novel targeted therapies, is urgently required. Chemokines are a group of small chemotactic cytokines that interact with seven-transmembrane G-protein-coupled receptors, and this interaction serves a crucial role in various physiological processes, including organ development and the host immune response, to recruit cells to specific sites in the body. There is also accumulating evidence that chemokines and chemokine receptors (CCRs) contribute to tumor development and progression, as well as metastasis. However, research regarding the functional roles of chemokines and their receptors in cancer is dynamic and context-dependent, and much remains to be elucidated, although various aspects have been explored extensively. In gastric cancer, C-C motif CCRs are involved in the biological behavior of tumor cells, including the processes of growth, invasion and survival, as well as the epithelial-mesenchymal transition. In the present review, attention is given to the clinical relevance of C-C motif CCRs in the development, progression, and metastasis of gastric cancer, particularly CCR7 and CCR5, which have been investigated extensively, as well as their potential therapeutic implications.
Collapse
Affiliation(s)
- Hyewon Ryu
- Department of Internal Medicine, Chungnam National University Hospital, Daejeon 35015, Republic of Korea
| | - Seung Woo Baek
- Department of Internal Medicine, Chungnam National University Hospital, Daejeon 35015, Republic of Korea
| | - Ji Young Moon
- Department of Internal Medicine, Chungnam National University Hospital, Daejeon 35015, Republic of Korea
| | - In-Sook Jo
- Department of Medical Science, School of Medicine Chungnam National University and Chungnam National University Hospital, Daejeon 35015, Republic of Korea
| | - Nayoung Kim
- Department of Medical Science, School of Medicine Chungnam National University and Chungnam National University Hospital, Daejeon 35015, Republic of Korea
| | - Hyo Jin Lee
- Department of Internal Medicine, School of Medicine Chungnam National University and Chungnam National University Hospital, Daejeon 35015, Republic of Korea
- Cancer Research Institute, Chungnam National University, Daejeon 35015, Republic of Korea
| |
Collapse
|
30
|
CCR5 Directs the Mobilization of CD11b+Gr1+Ly6Clow Polymorphonuclear Myeloid Cells from the Bone Marrow to the Blood to Support Tumor Development. Cell Rep 2017; 21:2212-2222. [DOI: 10.1016/j.celrep.2017.10.104] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 08/13/2017] [Accepted: 10/18/2017] [Indexed: 12/24/2022] Open
|
31
|
CCR5+ Myeloid-Derived Suppressor Cells Are Enriched and Activated in Melanoma Lesions. Cancer Res 2017; 78:157-167. [DOI: 10.1158/0008-5472.can-17-0348] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 02/15/2017] [Accepted: 10/25/2017] [Indexed: 11/16/2022]
|
32
|
贾 搏, 邱 小, 褚 洪, 孙 翔, 盘 杰, 王 治, 赵 建. [Effect of macrophage inflammatory protein-1β on proliferation and apoptosis of human tongue squamous cell carcinoma CAL-27 cells in vitro]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2017; 37:1104-1109. [PMID: 28801293 PMCID: PMC6765725 DOI: 10.3969/j.issn.1673-4254.2017.08.17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Indexed: 06/07/2023]
Abstract
OBJECTIVE To detect CCR5 protein expression in different human tongue squamous cell carcinoma cells and observe the effect of macrophage inflammatory protein-1β (MIP-1β) on the proliferation and apoptosis of CAL-27 cells. METHODS Western blotting and immunofluorescence staining were used to detect the expression of the CCR5, the receptor of MIP-1β, in 3 human tongue squamous cell carcinoma cells UM-1, CAL-27, and Tca-8113. CCK-8 assay was used to assess the proliferation of CAL-27 cells stimulated with 10, 20, and 40 ng/mL MIP-1β for 12, 24, or 48 h. The apoptosis of the cells stimulated with MIP-1β (10, 20, and 40 ng/mL) for 24 h was analyzed using flow cytometry with Annexin V/PI double staining. RESULTS CCR5 expression was detected both on the membrane and in the cytoplasm in all the 3 tongue squamous cell carcinoma cell lines. At the concentrations of 10, 20, and 40 ng/mL, MIP-1β stimulation for 12 and 24 h significantly promoted the proliferation of CAL-27 cells (P<0.05); MIP-1β stimulation for 48 h at the concentrations 10 and 20 ng/mL, but not at 40 ng/mL, promoted the proliferation of CAL-27 cells (P<0.05). MIP-1β stimulation at 40 ng/mL for 24 produced the most obvious apoptosis-inducing effect in CAL -27 cells (P<0.05), while MIP-1β at 10 or 20 ng/mL did not induce obvious apoptosis in the cells (P>0.05). CONCLUSION CCR5 is expressed in all the 3 human tongue squamous cell carcinoma cells. MIP-1β can promote the proliferation of CAL-27 cells but high concentrations of MIP-1β also induced cell apoptosis. Prolonged stimulation of the cells with a high concentration of MIP-1β shows attenuated effect in promoting cell proliferation probably as a result of cell apoptosis induced by MIP-1β.
Collapse
Affiliation(s)
- 搏 贾
- 南方医科大学口腔医院//广东省口腔医院, 口腔颌面外科, 广东 广州 510280Department of Oral Surgery, Guangdong Provincial Stomatological Hospital/Stomatological Hospital of Southern Medical University, Guangzhou 510280, China
| | - 小玲 邱
- 南方医科大学口腔医院//广东省口腔医院, 牙体牙髓科,广东 广州 510280Department of Endodontics, Guangdong Provincial Stomatological Hospital/Stomatological Hospital of Southern Medical University, Guangzhou 510280, China
| | - 洪星 褚
- 南方医科大学口腔医院//广东省口腔医院, 口腔颌面外科, 广东 广州 510280Department of Oral Surgery, Guangdong Provincial Stomatological Hospital/Stomatological Hospital of Southern Medical University, Guangzhou 510280, China
| | - 翔 孙
- 南方医科大学口腔医院//广东省口腔医院, 口腔颌面外科, 广东 广州 510280Department of Oral Surgery, Guangdong Provincial Stomatological Hospital/Stomatological Hospital of Southern Medical University, Guangzhou 510280, China
| | - 杰 盘
- 南方医科大学口腔医院//广东省口腔医院, 口腔颌面外科, 广东 广州 510280Department of Oral Surgery, Guangdong Provincial Stomatological Hospital/Stomatological Hospital of Southern Medical University, Guangzhou 510280, China
| | - 治平 王
- 南方医科大学口腔医院//广东省口腔医院, 口腔颌面外科, 广东 广州 510280Department of Oral Surgery, Guangdong Provincial Stomatological Hospital/Stomatological Hospital of Southern Medical University, Guangzhou 510280, China
| | - 建江 赵
- 南方医科大学口腔医院//广东省口腔医院, 口腔颌面外科, 广东 广州 510280Department of Oral Surgery, Guangdong Provincial Stomatological Hospital/Stomatological Hospital of Southern Medical University, Guangzhou 510280, China
| |
Collapse
|
33
|
Umansky V, Blattner C, Gebhardt C, Utikal J. CCR5 in recruitment and activation of myeloid-derived suppressor cells in melanoma. Cancer Immunol Immunother 2017; 66:1015-1023. [PMID: 28382399 PMCID: PMC11029643 DOI: 10.1007/s00262-017-1988-9] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 03/13/2017] [Indexed: 12/14/2022]
Abstract
Malignant melanoma is characterized by the development of chronic inflammation in the tumor microenvironment, leading to the accumulation of myeloid-derived suppressor cells (MDSCs). Using ret transgenic mouse melanoma model, we found a significant migration of MDSCs expressing C-C chemokine receptor (CCR)5 into primary tumors and metastatic lymph nodes, which was correlated with tumor progression. An increased CCR5 expression on MDSCs was associated with elevated concentrations of CCR5 ligands in melanoma microenvironment. In vitro experiments showed that the upregulation of CCR5 expression on CD11b+Gr1+ immature myeloid cells was induced by CCR5 ligands, IL-6, GM-CSF, and other inflammatory factors. Furthermore, CCR5+ MDSCs infiltrating melanoma lesions displayed a stronger immunosuppressive pattern than their CCR5- counterparts. Targeting CCR5/CCR5 ligand signaling via a fusion protein mCCR5-Ig, which selectively binds and neutralizes all three CCR5 ligands, increased the survival of tumor-bearing mice. This was associated with a reduced migration and immunosuppressive potential of tumor MDSCs. In melanoma patients, circulating CCR5+ MDSCs were increased as compared to healthy donors. Like in melanoma-bearing mice, we observed an enrichment of these cells and CCR5 ligands in tumors as compared to the peripheral blood. Our findings define a critical role for CCR5 not only in the recruitment but also in the activation of MDSCs in tumor lesions, suggesting that novel strategies of melanoma treatment could be based on blocking CCR5/CCR5 ligand interactions.
Collapse
Affiliation(s)
- Viktor Umansky
- Skin Cancer Unit (G300), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht Karl University of Heidelberg, Mannheim, Germany.
| | - Carolin Blattner
- Skin Cancer Unit (G300), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht Karl University of Heidelberg, Mannheim, Germany
| | - Christoffer Gebhardt
- Skin Cancer Unit (G300), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht Karl University of Heidelberg, Mannheim, Germany
| | - Jochen Utikal
- Skin Cancer Unit (G300), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht Karl University of Heidelberg, Mannheim, Germany
| |
Collapse
|
34
|
Lappano R, Maggiolini M. Pharmacotherapeutic Targeting of G Protein-Coupled Receptors in Oncology: Examples of Approved Therapies and Emerging Concepts. Drugs 2017; 77:951-965. [PMID: 28401445 DOI: 10.1007/s40265-017-0738-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
G protein-coupled receptors (GPCRs) are involved in numerous physio-pathological processes, including the stimulation of cancer progression. In this regard, it should be mentioned that although GPCRs may represent major pharmaceutical targets, only a few drugs acting as GPCR inhibitors are currently used in anti-tumor therapies. For instance, certain pro-malignancy effects mediated by GPCRs are actually counteracted by the use of small molecules and peptides that function as receptor antagonists or inverse agonists. Recently, humanized monoclonal antibodies targeting GPCRs have also been developed. Here, we review the current GPCR-targeted therapies for cancer treatment, summarizing the clinical studies that led to their official approval. We provide a broad overview of the mechanisms of action of the available anti-cancer drugs targeting gonadotropin-releasing hormone, somatostatin, chemokine, and Smoothened receptors. In addition, we discuss the anti-tumor potential of novel non-approved molecules and antibodies able to target some of the aforementioned GPCRs in different experimental models and clinical trials. Likewise, we focus on the repurposing in cancer patients of non-oncological GPCR-based drugs, elucidating the rationale behind this approach and providing clinical evidence on their safety and efficacy.
Collapse
Affiliation(s)
- Rosamaria Lappano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy.
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy.
| |
Collapse
|
35
|
Atretkhany KSN, Drutskaya MS, Nedospasov SA, Grivennikov SI, Kuprash DV. Chemokines, cytokines and exosomes help tumors to shape inflammatory microenvironment. Pharmacol Ther 2016; 168:98-112. [PMID: 27613100 DOI: 10.1016/j.pharmthera.2016.09.011] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Relationship between inflammation and cancer is now well-established and represents a paradigm that our immune response does not necessarily serves solely to protect us from infections and cancer. Many specific mechanisms that link chronic inflammation to cancer promotion and metastasis have been uncovered in the recent years. Here we are focusing on the effects that tumors may exert on inflammatory cascades, tuning the immune system ability to cause tumor promotion or regression. In particular, we discuss the contributions of chemokines, cytokines and exosomes to the processes such as induction of inflammation and tumorigenesis. Overall, tumor-elicited inflammation is a key driver of tumor progression and an essential component of tumor microenvironment.
Collapse
Affiliation(s)
- K-S N Atretkhany
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Vavilova Str. 32, Russia; Biological Faculty, Lomonosov Moscow State University, 119234, Moscow, Russia
| | - M S Drutskaya
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Vavilova Str. 32, Russia; Biological Faculty, Lomonosov Moscow State University, 119234, Moscow, Russia
| | - S A Nedospasov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Vavilova Str. 32, Russia; Biological Faculty, Lomonosov Moscow State University, 119234, Moscow, Russia; German Rheumatology Research Center (DRFZ), Berlin, Germany
| | - S I Grivennikov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Vavilova Str. 32, Russia; Fox Chase Cancer Center, Cancer Prevention and Control Program, Philadelphia, PA, USA.
| | - D V Kuprash
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Vavilova Str. 32, Russia; Biological Faculty, Lomonosov Moscow State University, 119234, Moscow, Russia.
| |
Collapse
|
36
|
RNA Sequencing of Tumor-Associated Microglia Reveals Ccl5 as a Stromal Chemokine Critical for Neurofibromatosis-1 Glioma Growth. Neoplasia 2016; 17:776-88. [PMID: 26585233 PMCID: PMC4656811 DOI: 10.1016/j.neo.2015.10.002] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 10/06/2015] [Accepted: 10/14/2015] [Indexed: 12/31/2022] Open
Abstract
Solid cancers develop within a supportive microenvironment that promotes tumor formation and growth through the elaboration of mitogens and chemokines. Within these tumors, monocytes (macrophages and microglia) represent rich sources of these stromal factors. Leveraging a genetically engineered mouse model of neurofibromatosis type 1 (NF1) low-grade brain tumor (optic glioma), we have previously demonstrated that microglia are essential for glioma formation and maintenance. To identify potential tumor-associated microglial factors that support glioma growth (gliomagens), we initiated a comprehensive large-scale discovery effort using optimized RNA-sequencing methods focused specifically on glioma-associated microglia. Candidate microglial gliomagens were prioritized to identify potential secreted or membrane-bound proteins, which were next validated by quantitative real-time polymerase chain reaction as well as by RNA fluorescence in situ hybridization following minocycline-mediated microglial inactivation in vivo. Using these selection criteria, chemokine (C-C motif) ligand 5 (Ccl5) was identified as a chemokine highly expressed in genetically engineered Nf1 mouse optic gliomas relative to nonneoplastic optic nerves. As a candidate gliomagen, recombinant Ccl5 increased Nf1-deficient optic nerve astrocyte growth in vitro. Importantly, consistent with its critical role in maintaining tumor growth, treatment with Ccl5 neutralizing antibodies reduced Nf1 mouse optic glioma growth and improved retinal dysfunction in vivo. Collectively, these findings establish Ccl5 as an important microglial growth factor for low-grade glioma maintenance relevant to the development of future stroma-targeted brain tumor therapies.
Collapse
|
37
|
Woollard SM, Kanmogne GD. Maraviroc: a review of its use in HIV infection and beyond. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:5447-68. [PMID: 26491256 PMCID: PMC4598208 DOI: 10.2147/dddt.s90580] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The human immunodeficiency virus-1 (HIV-1) enters target cells by binding its envelope glycoprotein gp120 to the CD4 receptor and/or coreceptors such as C-C chemokine receptor type 5 (CCR5; R5) and C-X-C chemokine receptor type 4 (CXCR4; X4), and R5-tropic viruses predominate during the early stages of infection. CCR5 antagonists bind to CCR5 to prevent viral entry. Maraviroc (MVC) is the only CCR5 antagonist currently approved by the United States Food and Drug Administration, the European Commission, Health Canada, and several other countries for the treatment of patients infected with R5-tropic HIV-1. MVC has been shown to be effective at inhibiting HIV-1 entry into cells and is well tolerated. With expanding MVC use by HIV-1-infected humans, different clinical outcomes post-approval have been observed with MVC monotherapy or combination therapy with other antiretroviral drugs, with MVC use in humans infected with dual-R5- and X4-tropic HIV-1, infected with different HIV-1 genotype or infected with HIV-2. This review discuss the role of CCR5 in HIV-1 infection, the development of the CCR5 antagonist MVC, its pharmacokinetics, pharmacodynamics, drug–drug interactions, and the implications of these interactions on treatment outcomes, including viral mutations and drug resistance, and the mechanisms associated with the development of resistance to MVC. This review also discusses available studies investigating the use of MVC in the treatment of other diseases such as cancer, graft-versus-host disease, and inflammatory diseases.
Collapse
Affiliation(s)
- Shawna M Woollard
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Georgette D Kanmogne
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
38
|
Métifiot M, Amrane S, Mergny JL, Andreola ML. Anticancer molecule AS1411 exhibits low nanomolar antiviral activity against HIV-1. Biochimie 2015; 118:173-5. [PMID: 26363100 DOI: 10.1016/j.biochi.2015.09.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 09/07/2015] [Indexed: 11/29/2022]
Abstract
During clinical trials, a number of fully characterized molecules are dropped along the way because they do not provide enough benefit for the patient. Some of them show limited side effects and might be of great use for other applications. AS1411 is a nucleolin-targeting aptamer that underwent phase II clinical trials as anticancer agent. Here, we show that AS1411 exhibits extremely potent antiviral activity and is therefore an attractive new lead as anti-HIV agent.
Collapse
Affiliation(s)
- Mathieu Métifiot
- Laboratoire MFP, CNRS UMR-5234, Université de Bordeaux, FR Transbiomed, 146 Rue Léo Saignat, 33076 Bordeaux, France
| | - Samir Amrane
- INSERM U869, IECB, ARNA Laboratory, Université de Bordeaux, 2 Rue Robert Escarpit, 33600 Pessac, France
| | - Jean-Louis Mergny
- INSERM U869, IECB, ARNA Laboratory, Université de Bordeaux, 2 Rue Robert Escarpit, 33600 Pessac, France.
| | - Marie-Line Andreola
- Laboratoire MFP, CNRS UMR-5234, Université de Bordeaux, FR Transbiomed, 146 Rue Léo Saignat, 33076 Bordeaux, France.
| |
Collapse
|
39
|
The key role of astrocyte elevated gene-1 in CCR6-induced EMT in cervical cancer. Tumour Biol 2015; 36:9763-7. [PMID: 26156805 DOI: 10.1007/s13277-015-3760-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 07/02/2015] [Indexed: 01/05/2023] Open
Abstract
In recent years, astrocyte elevated gene-1 (AEG-1) has been recommended as an important mediator that is involved in the epithelial-to-mesenchymal transition (EMT) process. However, the mechanisms underlying the chemokine (C-C motif) ligand 20 (CCL20)/chemokine (C-C motif) receptor 6 (CCR6)-AEG-1 pathway-mediated EMT in cervical cancer (CC) have not been well featured till now. We used immunohistochemistry and immunoblotting to assess the expression of AEG-1 in 94 cervical cancer tissues and cells. Subsequently, cervical cancer SiHa cells were treated with si-AEG-1 and then subjected to in vitro assays. We observed that AEG-1 proteins were highly expressed in cervical cancer tissues and closely correlated with International Federation of Gynecology and Obstetrics (FIGO) stage and metastasis. Importantly, we validated the expression of AEG-1, p-Erk1/2, p-Akt, vimentin, N-cadherin, and matrix metalloproteinase 2 (MMP2) increased in SiHa with CCL20 treatment in a concentration-dependent manner. When cells were treated with si-AEG-1, the expression of p-Erk1/2, p-Akt, vimentin, N-cadherin, and MMP2 was also downregulated. Using the cell cycle assay, the knockdown of AEG-1 inhibited the entry of G1 into S phase. In conclusion, AEG-1 mediates CCL20/CCR6-induced EMT development via both Erk1/2 and Akt signaling pathway in cervical cancer, which indicates that CCL20/CCR6-AEG-1-EMT pathway could be suggested as a useful target to affect the progression of cervical cancer.
Collapse
|
40
|
Ganetsky A, Miano TA, Hughes ME, Vonderheide RH, Porter DL, Reshef R. Lack of a significant pharmacokinetic interaction between maraviroc and tacrolimus in allogeneic HSCT recipients. J Antimicrob Chemother 2015; 70:2078-83. [PMID: 25881619 PMCID: PMC4472330 DOI: 10.1093/jac/dkv082] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Revised: 02/20/2015] [Accepted: 03/13/2015] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES Emerging data suggest that the combination of tacrolimus and the CCR5 antagonist maraviroc, both cytochrome P450-3A4 substrates, may be effective in preventing graft-versus-host disease in patients undergoing allogeneic HSCT. This study evaluated whether a pharmacokinetic interaction exists between these agents. METHODS The study included 36 allogeneic HSCT recipients who received maraviroc + tacrolimus and 43 recipients of tacrolimus alone. We used a difference-in-differences analysis to examine the change in the concentration/dose ratios of tacrolimus after the discontinuation of maraviroc. In addition, we analysed the concentrations and dose requirements of tacrolimus in the two groups. RESULTS There was no significant difference in tacrolimus concentration/dose ratios in patients receiving maraviroc + tacrolimus compared with tacrolimus alone. Upon discontinuation of maraviroc, the change in concentration/dose ratio was small and not significant relative to the control group, and the effect estimate was further attenuated after adjustment for confounders [-0.35 (ng/mL)/(mg/day); P = 0.46]. In addition, the change in mean tacrolimus dose after discontinuation of maraviroc was similar between the groups (0.12 mg/day; P = 0.56), as was the change in mean tacrolimus concentration (0.02 ng/mL; P = 0.97). CONCLUSIONS Our findings do not support a significant inhibitory effect of maraviroc on the metabolism of tacrolimus. These data demonstrate that this drug combination is safe and imply that the protective effect of maraviroc against graft-versus-host disease was not mediated through an increase in tacrolimus concentrations. These findings are important for the design of clinical trials that evaluate maraviroc in combination with cytochrome P450-3A4 substrates.
Collapse
Affiliation(s)
- Alex Ganetsky
- Department of Pharmacy, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Todd A Miano
- Center for Pharmacoepidemiology Research and Training, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Mitchell E Hughes
- Department of Pharmacy, Hospital of the University of Pennsylvania, Philadelphia, PA, USA Philadelphia College of Pharmacy, University of the Sciences, Philadelphia, PA, USA
| | - Robert H Vonderheide
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA Abramson Cancer Center and the Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - David L Porter
- Abramson Cancer Center and the Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Ran Reshef
- Abramson Cancer Center and the Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
41
|
Shi P, Fang C, Pang X. Astrocyte elevated gene-1 regulates CCL3/CCR5-induced epithelial-to-mesenchymal transition via Erk1/2 and Akt signaling in cardiac myxoma. Oncol Rep 2015; 34:1319-26. [PMID: 26134542 DOI: 10.3892/or.2015.4081] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 06/09/2015] [Indexed: 11/06/2022] Open
Abstract
In recent years, astrocyte elevated gene-1 (AEG-1) has been reported as a key mediator that is involved in the epithelial-to-mesenchymal transition (EMT) process. However, the mechanisms underlying CCL3/CCR5-AEG-1 pathway-mediated EMT in cardiac myxoma (CM) has not been well featured till now. We used immnohistochemistry and immunoblotting to assess the expression of CCR5 and AEG-1 in 30 cases of CM tissues and cells. Subsequently, cultured CM cells were treated with si-AEG-1 or si-CCR5 and then subjected to in vitro assays. We observed that CCR5 and AEG-1 proteins were highly expressed in CM tissues (73.3 and 76.7%, respectively) and closely correlated with tumor size (>5 cm). Importantly, we validated the expression of AEG-1, p-Erk1/2, p-Akt, vimentin, N-cadherin and MMP2 increased in the CM cell with CCL3 treatment in a time- and concentration-dependent manner. When CM cells were treated with si-CCR5, the expression of AEG-1, p-Erk1/2, p-Akt, vimentin, N-cadherin and MMP2 was downregulated. In addition, when CM cells were treated with si-AEG-1, the expression of p-Erk1/2, p-Akt, vimentin, N-cadherin and MMP2 was also downregulated. Using the cell cycle and proliferation assay, the knockdown of AEG-1 inhibited the entry of G1 into S phase and the proliferation capacity of CM cells. In conclusion, AEG-1 mediates CCL3/CCR5-induced EMT development via both Erk1/2 and Akt signaling pathway in CM patients, which indicates CCL3/CCR5-AEG-1-EMT pathway could be suggested as a useful target to affect the progression of CM.
Collapse
Affiliation(s)
- Ping Shi
- Department of Emergency Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, P.R. China
| | - Changcun Fang
- Department of Cardiosurgery, Qilu Hospital of Shandong University, Jinan, Shandong, P.R. China
| | - Xinyan Pang
- Department of Cardiosurgery, Qilu Hospital of Shandong University, Jinan, Shandong, P.R. China
| |
Collapse
|
42
|
Pervaiz A, Ansari S, Berger MR, Adwan H. CCR5 blockage by maraviroc induces cytotoxic and apoptotic effects in colorectal cancer cells. Med Oncol 2015; 32:158. [PMID: 25840792 DOI: 10.1007/s12032-015-0607-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 03/26/2015] [Indexed: 12/21/2022]
Abstract
Alterations in the expression of C-C chemokine receptor type 5 (CCR5 or CD195) have been correlated with disease progression in different cancers. Recently, a few investigations have reported the blockage of this receptor by an antagonist (maraviroc) and its antineoplastic effects on tumor cell growth. However, little is known about the mechanistic reasons behind these antineoplastic effects of CCR5 blockage by maraviroc. In this study, we blocked the CCR5 receptor by maraviroc in SW480 and SW620 colorectal cancer cells to study the resulting changes in biological properties and related pathways. This blockage induced significantly reduced proliferation and a profound arrest in G1 phase of the cell cycle. Concomitantly, maraviroc caused significant signs of apoptosis at morphological level. Significant modulation of multiple apoptosis-relevant genes was also noticed at mRNA levels. In addition, we found remarkable increases in cleaved caspases at protein level. These modulations led us to propose a signaling pathway for the observed apoptotic effects. In conclusion, blocking the CCR5 by maraviroc induces significant cytotoxic and apoptotic effects in colorectal cancer cells. Thus, maraviroc can be considered a model compound, which may foster the development of further CCR5 antagonists to be used for the treatment of colorectal cancer.
Collapse
Affiliation(s)
- Asim Pervaiz
- Toxicology and Chemotherapy Unit, German Cancer Research Center, Im Neuenheimer Feld 581, 69120, Heidelberg, Germany
| | | | | | | |
Collapse
|