1
|
Bamodu OA, Chung CC, Pisanic TR, Wu ATH. The intricate interplay between cancer stem cells and cell-of-origin of cancer: implications for therapeutic strategies. Front Oncol 2024; 14:1404628. [PMID: 38800385 PMCID: PMC11116576 DOI: 10.3389/fonc.2024.1404628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 04/25/2024] [Indexed: 05/29/2024] Open
Abstract
Background Cancer stem cells (CSCs) have emerged as pivotal players in tumorigenesis, disease progression, and resistance to therapies. Objective This comprehensive review delves into the intricate relationship between CSCs and the cell-of-origin in diverse cancer types. Design Comprehensive review of thematically-relevant literature. Methods We explore the underlying molecular mechanisms that drive the conversion of normal cells into CSCs and the impact of the cell-of-origin on CSC properties, tumor initiation, and therapeutic responses. Moreover, we discuss potential therapeutic interventions targeting CSCs based on their distinct cell-of-origin characteristics. Results Accruing evidence suggest that the cell-of-origin, the cell type from which the tumor originates, plays a crucial role in determining the properties of CSCs and their contribution to tumor heterogeneity. Conclusion By providing critical insights into the complex interplay between CSCs and their cellular origins, this article aims to enhance our understanding of cancer biology and pave the way for more effective and personalized cancer treatments.
Collapse
Affiliation(s)
- Oluwaseun Adebayo Bamodu
- Directorate of Postgraduate Studies, School of Clinical Medicine, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
- Ocean Road Cancer Institute, Dar es Salaam, Tanzania
| | - Chen-Chih Chung
- Department of Neurology, Taipei Medical University - Shuang Ho Hospital, New Taipei City, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University - Shuang Ho Hospital, New Taipei City, Taiwan
| | - Thomas R. Pisanic
- Johns Hopkins Institute for NanoBioTechnology, Baltimore, MD, United States
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Oncology - Cancer Genetics and Epigenetics, Johns Hopkins University, Baltimore, MD, United States
| | - Alexander T. H. Wu
- The Program for Translational Medicine, Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
- Clinical Research Center, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
2
|
Nam AR, Yoon J, Jin MH, Bang JH, Oh KS, Seo HR, Kim JM, Kim TY, Oh DY. ATR inhibition amplifies antitumor effects of olaparib in biliary tract cancer. Cancer Lett 2021; 516:38-47. [PMID: 34082024 DOI: 10.1016/j.canlet.2021.05.029] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/17/2021] [Accepted: 05/25/2021] [Indexed: 01/15/2023]
Abstract
Olaparib, a potent PARP inhibitor, has been shown to have great anti-tumor effects in some tumor types. Although biliary tract cancer (BTC) is a good candidate for DNA damage response (DDR)-targeted agents, targeted DDR inhibitors, including olaparib, are currently rarely evaluated in BTC. In our project, a total of ten BTC cell lines were used to assess the efficacy of olaparib. Olaparib alone showed moderate anti-proliferative effects in BTC cells and increased p-ATR and PD-L1 expression levels. In combination with an ATR inhibitor (AZD6738, ceralasertib) showed synergistic anti-proliferative effects and increased DNA strand breaks in vitro. PD-L1 induced by olaparib was also downregulated by ceralasertib through p-STAT-3 and YAP reduction with or without human primary peripheral blood mononuclear cells. In SNU478-xenograft models, the combination treatment significantly suppressed tumor growth. PD-L1 and YAP were strongly downregulated, similar to in vitro conditions, and expression of CXCR2 and CXCR4 was further reduced. In the current ongoing clinical trial (NCT04298021), BTC patients treated with olaparib and ceralasertib combination have shown tumor response. In conclusion, co-targeting of PARP and ATR might be a potential therapeutic approach for patients with BTC.
Collapse
Affiliation(s)
- Ah-Rong Nam
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Jeesun Yoon
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Mei-Hua Jin
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Ju-Hee Bang
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Kyoung-Seok Oh
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Hye-Rim Seo
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea; Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, South Korea
| | - Jae-Min Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea; Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, South Korea
| | - Tae-Yong Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea; Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Do-Youn Oh
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea; Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea; Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, South Korea.
| |
Collapse
|
3
|
Yu X, Lu W, Ng CW, Xu S, Wu Y, Chen S, Gao Y, Zhang Y, Zhang Q, Xu Y, Liu Y, Li S. Computer-aided assessment of the chemokine receptors CXCR3, CXCR4 and CXCR7 expression in gallbladder carcinoma. J Cell Mol Med 2020; 24:7670-7674. [PMID: 32512633 PMCID: PMC7339221 DOI: 10.1111/jcmm.15219] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 01/30/2020] [Accepted: 03/06/2020] [Indexed: 12/13/2022] Open
Abstract
Gallbladder carcinoma (GBC) is a vicious and invasive disease. The major challenge in the clinical treatment of GBC is the lack of a suitable prognosis method. Chemokine receptors such as CXCR3, CXCR4 and CXCR7 play vital roles in the process of tumour progression and metastasis. Their expression levels and distribution are proven to be indicative of the progression of GBC, but are hard to be decoded by conventional pathological methods, and therefore, not commonly used in the prognosis of GBC. In this study, we developed a computer‐aided image analysis method, which we used to quantitatively measure the expression levels of CXCR3, CXCR4 and CXCR7 in the nuclei and cytoplasm of glandular and interstitial cells from a cohort of 55 GBC patients. We found that CXCR3, CXCR4 and CXCR7 expressions are associated with the clinicopathological variables of GBC. Cytoplasmic CXCR3, nuclear CXCR7 and cytoplasmic CXCR7 were significant predictive factors of histology invasion, whereas cytoplasmic CXCR4 and nuclear CXCR4 were significantly correlated with T and N stage and were associated with the overall survival and disease‐free survival. These results suggest that the quantification and localisation of CXCR3, CXCR4 and CXCR7 expressions in different cell types should be considered using computer‐aided assessment to improve the accuracy of prognosis in GBC.
Collapse
Affiliation(s)
- Xiao Yu
- Department of Pathology and Forensic Medicine, Dalian Medical University, Dalian, China
| | - Wei Lu
- Department of General Surgery, School of Medicine, Xinhua Hospital, Shanghai JiaoTong University, Shanghai, China.,Institute of Biliary Tract Diseases Research, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Chan Way Ng
- Invitrocue Pte Ltd., Singapore City, Singapore
| | - Shuoyu Xu
- Department of Radiology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yao Wu
- Invitrocue Pte Ltd., Singapore City, Singapore
| | - Shili Chen
- Institute of Biliary Tract Diseases Research, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Yuren Gao
- Department of Urology, the second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yijian Zhang
- Institute of Biliary Tract Diseases Research, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Qingqing Zhang
- Department of Pathology and Forensic Medicine, Dalian Medical University, Dalian, China
| | - Yuzhen Xu
- Department of Gastrointestinal Surgery, Xu Zhou Center Hospital, Affiliated to Medical College of Southeast University, Jiangsu, China
| | - Yingbin Liu
- Department of General Surgery, School of Medicine, Xinhua Hospital, Shanghai JiaoTong University, Shanghai, China.,Institute of Biliary Tract Diseases Research, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Sheng Li
- National-Local Engineering Research Center for Drug Research and Development (R&D) of Neurodegenerative Diesases, Department of Biochemistry, Dalian Medical University, Dalian, China
| |
Collapse
|
4
|
Construction of a CXC Chemokine-Based Prediction Model for the Prognosis of Colon Cancer. BIOMED RESEARCH INTERNATIONAL 2020; 2020:6107865. [PMID: 32337262 PMCID: PMC7150705 DOI: 10.1155/2020/6107865] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 03/05/2020] [Indexed: 01/01/2023]
Abstract
Colon cancer is the third most common cancer, with a high incidence and mortality. Construction of a specific and sensitive prediction model for prognosis is urgently needed. In this study, profiles of patients with colon cancer with clinical and gene expression data were downloaded from Gene Expression Omnibus and The Cancer Genome Atlas (TCGA). CXC chemokines in patients with colon cancer were investigated by differential expression gene analysis, overall survival analysis, receiver operating characteristic analysis, gene set enrichment analysis (GSEA), and weighted gene coexpression network analysis. CXCL1, CXCL2, CXCL3, and CXCL11 were upregulated in patients with colon cancer and significantly correlated with prognosis. The area under curve (AUC) of the multigene forecast model of CXCL1, CXCL11, CXCL2, and CXCL3 was 0.705 in the GSE41258 dataset and 0.624 in TCGA. The prediction model was constructed using the risk score of the multigene model and three clinicopathological risk factors and exhibited 92.6% and 91.8% accuracy in predicting 3-year and 5-year overall survival of patients with colon cancer, respectively. In addition, by GSEA, expression of CXCL1, CXCL11, CXCL2, and CXCL3 was correlated with several signaling pathways, including NOD-like receptor, oxidative phosphorylation, mTORC1, interferon-gamma response, and IL6/JAK/STAT3 pathways. Patients with colon cancer will benefit from this prediction model for prognosis, and this will pave the way to improve the survival rate and optimize treatment for colon cancer.
Collapse
|
5
|
HOXB5 acts as an oncogenic driver in head and neck squamous cell carcinoma via EGFR/Akt/Wnt/β-catenin signaling axis. Eur J Surg Oncol 2019; 46:1066-1073. [PMID: 31864826 DOI: 10.1016/j.ejso.2019.12.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 10/26/2019] [Accepted: 12/11/2019] [Indexed: 12/19/2022] Open
Abstract
INTRODUCTION Identification of therapeutic targets in head and neck squamous cell carcinoma (HNSCC) is essential because most of the patients with advanced HNSCC have a poor prognosis. Homeobox genes constitute a large cluster of transcription factors with important regulatory roles in mammalian embryonic development and cell differentiation. The oncogenic role of homeobox B5 (HOXB5) in HNSCC has not been investigated. MATERIALS AND METHODS We used The Cancer Genome Atlas (TCGA) data to evaluate the correlations between HOXB5 expression and various HNSCC clinicopathological factors. We knocked down HOXB5 expression in HNSCC cell lines and explored the in vitro and in vivo effects on cell proliferation and motility, and HOXB5 signaling. RESULTS The Cancer Genome Atlas (TCGA) data shows that HOXB5 is overexpressed in HNSCC compared to normal tissues and significantly associates with tumor stage (P = 0.003), lymph node metastasis (P = 0.031), disease stage (P = 0.002), and angiolymphatic invasion (P = 0.004). Our results also show that HOXB5 expression is up-regulated in HNSCC cell lines, and HOXB5 knockdown significantly reduced cell proliferation and tumor growth in vitro and in vivo. Inhibition of HOXB5 decreases cell migration and invasion via suppression of epithelial-to-mesenchymal transition (EMT)-associated proteins expression. Moreover, HOXB5 directly binds to the promoter region of EGFR and consequently regulates the activity of the Akt/Wnt/β-catenin signaling axis. CONCLUSION HOXB5 may be a novel therapeutic target as an oncogenic driver by regulating EGFR transcription in HNSCC.
Collapse
|
6
|
Huang S, Wang H, Chen W, Zhan M, Xu S, Huang X, Lin R, Shen H, Wang J. Tamoxifen inhibits cell proliferation by impaired glucose metabolism in gallbladder cancer. J Cell Mol Med 2019; 24:1599-1613. [PMID: 31782270 PMCID: PMC6991689 DOI: 10.1111/jcmm.14851] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 10/27/2019] [Accepted: 11/02/2019] [Indexed: 02/05/2023] Open
Abstract
Gallbladder cancer (GBC) is the leading malignancy of biliary system showing refractory chemoresistance to current first‐line drugs. Growing epidemiological evidences have established that the incidence of GBC exhibits significant gender predominance with females two‐threefold higher than males, suggesting oestrogen/oestrogen receptors (ERs) signalling might be a critical driver of tumorigenesis in gallbladder. This study aims to evaluate the antitumour activity of tamoxifen (TAM), a major agent of hormonal therapy for breast cancer, in preclinical GBC model. Quantitative real‐time PCR was used to investigate mRNA levels. Protein expression was measured by immunohistochemistry and Western blot. Glycolytic levels were measured by glucose consumption and lactic acid measurement. The antitumour activity of TAM alone or with cisplatin was examined with CCK8 assay, colony formation, flow cytometry and in vivo models. The results revealed that ERɑ expression was higher in GBC tissues and predicted poor clinical outcomes. TAM was showed effective against a variety of GBC cell lines. Mechanical investigations revealed that TAM enabled potent reactive oxygen species (ROS) production by reduced nuclear factor Nrf2 expression and its target genes, leading to the activation of AMPK, which subsequently induced impaired glycolysis and survival advantages. Notably, TAM was demonstrated to sensitize GBC cells to cisplatin (CDDP) both in vitro and in vivo. In agreement with these findings, elimination of oestrogens by ovariectomy in nude mice prevented CDDP resistance. In summary, these results provide basis for TAM treatment for GBC and shed novel light on the potential application of endocrine therapy for patients with GBC.
Collapse
Affiliation(s)
- Shuai Huang
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hui Wang
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Chen
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ming Zhan
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Sunwang Xu
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xince Huang
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ruirong Lin
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hui Shen
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jian Wang
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
7
|
Zhang J, Chen J, Wo D, Yan H, Liu P, Ma E, Li L, Zheng L, Chen D, Yu Z, Liang C, Peng J, Ren DN, Zhu W. LRP6 Ectodomain Prevents SDF-1/CXCR4-Induced Breast Cancer Metastasis to Lung. Clin Cancer Res 2019; 25:4832-4845. [PMID: 31010839 DOI: 10.1158/1078-0432.ccr-18-3557] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 03/04/2019] [Accepted: 04/15/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Lung metastasis is an important cause of breast cancer-related deaths, in which SDF-1/CXCR4 signaling pathway plays a critical role. Single transmembrane protein LRP6 is viewed as an oncogene via activating the Wnt/β-catenin signaling pathway. Our work aims to investigate the relationship between SDF-1/CXCR4 and LRP6 in breast cancer lung metastasis. EXPERIMENTAL DESIGN We examined the expressions and functions of SDF-1/CXCR4 and LRP6 as well as their relationship in breast cancer in vitro and in vivo. RESULTS LRP6 ectodomain (LRP6N) directly bound to CXCR4 and competitively prevented SDF-1 binding to CXCR4. LRP6N prevented SDF-1/CXCR4-induced metastasis to lung and prolonged survival in mice bearing breast tumors, whereas LRP6 knockdown activated SDF-1/CXCR4 signal transduction and promoted lung metastasis and tumor death. Furthermore, patients with breast cancer with high CXCR4 expression had poor prognosis, which was exacerbated by low LRP6 expression but improved by high LRP6 expression. Interestingly, a secreted LRP6N was found in the serum of mice and humans, which was downregulated by the onset of cancer metastasis in both mice bearing breast cancer as well as in patients with breast cancer. CONCLUSIONS LRP6N might be a promising diagnostic marker for the early detection of breast cancer metastasis as well as an inhibitor of SDF-1/CXCR4-induced breast cancer metastasis. LRP6N also provides an interesting link between Wnt signaling and SDF-1/CXCR4 signaling, the two key pathways involved in cancer development.
Collapse
Affiliation(s)
- Jiankang Zhang
- Clinical and Translational Research Center, Research Institute of Heart Failure Shanghai East Hospital, Key Laboratory of Arrhythmias of Ministry of Education, Tongji University School of Medicine, Shanghai, China
| | - Jinxiao Chen
- Department of Plastic and Reconstructive Surgery, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Da Wo
- Department of Plastic and Reconstructive Surgery, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hongwei Yan
- Clinical and Translational Research Center, Research Institute of Heart Failure Shanghai East Hospital, Key Laboratory of Arrhythmias of Ministry of Education, Tongji University School of Medicine, Shanghai, China
| | - Peng Liu
- Clinical and Translational Research Center, Research Institute of Heart Failure Shanghai East Hospital, Key Laboratory of Arrhythmias of Ministry of Education, Tongji University School of Medicine, Shanghai, China
| | - En Ma
- Clinical and Translational Research Center, Research Institute of Heart Failure Shanghai East Hospital, Key Laboratory of Arrhythmias of Ministry of Education, Tongji University School of Medicine, Shanghai, China
| | - Limei Li
- Clinical and Translational Research Center, Research Institute of Heart Failure Shanghai East Hospital, Key Laboratory of Arrhythmias of Ministry of Education, Tongji University School of Medicine, Shanghai, China
| | - Liang Zheng
- Clinical and Translational Research Center, Research Institute of Heart Failure Shanghai East Hospital, Key Laboratory of Arrhythmias of Ministry of Education, Tongji University School of Medicine, Shanghai, China
| | - Daxin Chen
- Fujian Key Laboratory of Integrative Medicine on Geriatric, Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Zuoren Yu
- Clinical and Translational Research Center, Research Institute of Heart Failure Shanghai East Hospital, Key Laboratory of Arrhythmias of Ministry of Education, Tongji University School of Medicine, Shanghai, China
| | - Chunli Liang
- Department of Surgery East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jun Peng
- Fujian Key Laboratory of Integrative Medicine on Geriatric, Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China.
| | - Dan-Ni Ren
- Fujian Key Laboratory of Integrative Medicine on Geriatric, Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China.
| | - Weidong Zhu
- Clinical and Translational Research Center, Research Institute of Heart Failure Shanghai East Hospital, Key Laboratory of Arrhythmias of Ministry of Education, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
8
|
Nemunaitis JM, Brown-Glabeman U, Soares H, Belmonte J, Liem B, Nir I, Phuoc V, Gullapalli RR. Gallbladder cancer: review of a rare orphan gastrointestinal cancer with a focus on populations of New Mexico. BMC Cancer 2018; 18:665. [PMID: 29914418 PMCID: PMC6006713 DOI: 10.1186/s12885-018-4575-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 06/01/2018] [Indexed: 12/18/2022] Open
Abstract
Gallbladder cancer is a rare malignancy of the biliary tract with a poor prognosis, frequently presenting at an advanced stage. While rare in the United States overall, gallbladder cancer has an elevated incidence in geographically distinct locations of the globe including Chile, North India, Korea, Japan and the state of New Mexico in the United States. People with Native American ancestry have a much elevated incidence of gallbladder cancer compared to Hispanic and non-Hispanic white populations of New Mexico. Gallbladder cancer is also one of the few bi-gendered cancers with an elevated female incidence compared to men. Similar to other gastrointestinal cancers, gallbladder cancer etiology is likely multi-factorial involving a combination of genomic, immunological, and environmental factors. Understanding the interplay of these unique epidemiological factors is crucial in improving the prevention, early detection, and treatment of this lethal disease. Previous studies have failed to identify a distinct genomic mutational profile in gallbladder cancers, however, work to identify promising clinically actionable targets is this form of cancer is ongoing. Examples include, interest in the HER2/Neu signaling pathway and the recognition that chronic inflammation plays a crucial role in gallbladder cancer pathogenesis. In this review, we provide a comprehensive overview of gallbladder cancer epidemiology, risk factors, pathogenesis, and treatment with a specific focus on the rural and Native American populations of New Mexico. We conclude this review by discussing future research directions with the goal of improving clinical outcomes for patients of this lethal malignancy.
Collapse
Affiliation(s)
- Jacklyn M Nemunaitis
- Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM, USA.,Department of Internal Medicine, Division of Hematology and Oncology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Ursa Brown-Glabeman
- Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM, USA.,Department of Internal Medicine, Division of Hematology and Oncology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Heloisa Soares
- Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM, USA.,Department of Internal Medicine, Division of Hematology and Oncology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Jessica Belmonte
- Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM, USA.,Department of Internal Medicine, Division of Hematology and Oncology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Ben Liem
- Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM, USA.,Department of Internal Medicine, Division of Hematology and Oncology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Itzhak Nir
- Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM, USA.,Department of Surgery, Division of Surgical Oncology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Victor Phuoc
- Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM, USA.,Department of Surgery, Division of Surgical Oncology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Rama R Gullapalli
- Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM, USA. .,Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA. .,Department of Chemical and Biological Engineering, University of New Mexico, Room 333A, MSC08-4640, Albuquerque, NM, 87131, USA.
| |
Collapse
|
9
|
Guo JC, Li J, Zhou L, Yang JY, Zhang ZG, Liang ZY, Zhou WX, You L, Zhang TP, Zhao YP. CXCL12-CXCR7 axis contributes to the invasive phenotype of pancreatic cancer. Oncotarget 2018; 7:62006-62018. [PMID: 27542220 PMCID: PMC5308707 DOI: 10.18632/oncotarget.11330] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 07/27/2016] [Indexed: 12/26/2022] Open
Abstract
Chemokine (C-X-C motif) receptor 7 (CXCR7) and its ligand, chemokine (C-X-C motif) ligand 12 (CXCL12), were established to be involved in biological behaviors and associated with prognosis in many cancers. However, effects, underlying mechanisms of CXCL12-CXCR7 axis in invasive phenotype of pancreatic cancer (PC) and its clinicopathologic significances have not been comprehensively explored. In the present study, it was first found by tissue microarray-based immunohistochemistry that CXCL12 and CXCR7 staining scores were significantly associated with vessel invasion and overall survival in two independent cohorts of PC. Besides, co-expression of these proteins was an independent prognosticator in multivariate analysis in both cohorts. Then, migration and invasion, but not proliferation, were decreased in CXCR7-stably silenced PC cells, whereas opposite changes were observed in CXCR7-stably overexpressed cells, accompanied by alterations of mTOR and Rho/ROCK pathways. CXCL12 stimulated migration, invasion, CXCR7 expression and phosphorylation of key mTOR proteins. AMD3100 did not influence effects of CXCL12. Two mTOR inhibitors, rapamycin and Torin1, reversed enhanced invasive phenotypes and mTOR phosphorylation in CXCR7-overexpressed cells. Moreover, CXCR7 directly interacts with mTOR. Finally, liver metastasis, but not growth, was affected by CXCR7 status in orthotopically-implanted PC models in nude mice. Collectively, CXCL12-CXCR7 axis accelerates migration and invasion of PC cells through mTOR and Rho/ROCK pathways, and predicts poor prognosis of PC.
Collapse
Affiliation(s)
- Jun-Chao Guo
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, China
| | - Jian Li
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, China.,Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, China
| | - Li Zhou
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, China
| | - Jian-Yu Yang
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhi-Gang Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhi-Yong Liang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, China
| | - Wei-Xun Zhou
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, China
| | - Tai-Ping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, China
| | - Yu-Pei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
10
|
Rojas-Sepúlveda D, Tittarelli A, Gleisner MA, Ávalos I, Pereda C, Gallegos I, González FE, López MN, Butte JM, Roa JC, Fluxá P, Salazar-Onfray F. Tumor lysate-based vaccines: on the road to immunotherapy for gallbladder cancer. Cancer Immunol Immunother 2018; 67:1897-1910. [PMID: 29600445 PMCID: PMC6244977 DOI: 10.1007/s00262-018-2157-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 03/26/2018] [Indexed: 12/14/2022]
Abstract
Immunotherapy based on checkpoint blockers has proven survival benefits in patients with melanoma and other malignancies. Nevertheless, a significant proportion of treated patients remains refractory, suggesting that in combination with active immunizations, such as cancer vaccines, they could be helpful to improve response rates. During the last decade, we have used dendritic cell (DC) based vaccines where DCs loaded with an allogeneic heat-conditioned melanoma cell lysate were tested in a series of clinical trials. In these studies, 60% of stage IV melanoma DC-treated patients showed immunological responses correlating with improved survival. Further studies showed that an essential part of the clinical efficacy was associated with the use of conditioned lysates. Gallbladder cancer (GBC) is a high-incidence malignancy in South America. Here, we evaluated the feasibility of producing effective DCs using heat-conditioned cell lysates derived from gallbladder cancer cell lines (GBCCL). By characterizing nine different GBCCLs and several fresh tumor tissues, we found that they expressed some tumor-associated antigens such as CEA, MUC-1, CA19-9, Erb2, Survivin, and several carcinoembryonic antigens. Moreover, heat-shock treatment of GBCCLs induced calreticulin translocation and release of HMGB1 and ATP, both known to act as danger signals. Monocytes stimulated with combinations of conditioned lysates exhibited a potent increase of DC-maturation markers. Furthermore, conditioned lysate-matured DCs were capable of strongly inducing CD4+ and CD8+ T cell activation, in both allogeneic and autologous cell co-cultures. Finally, in vitro stimulated CD8+ T cells recognize HLA-matched GBCCLs. In summary, GBC cell lysate-loaded DCs may be considered for future immunotherapy approaches.
Collapse
Affiliation(s)
- Daniel Rojas-Sepúlveda
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 1027, building H, Third floor, 8380453, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, 8380453, Santiago, Chile.,Faculty of Science, Universidad San Sebastián, Lota 2465, 7510157, Santiago, Chile
| | - Andrés Tittarelli
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 1027, building H, Third floor, 8380453, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, 8380453, Santiago, Chile
| | - María Alejandra Gleisner
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 1027, building H, Third floor, 8380453, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, 8380453, Santiago, Chile
| | - Ignacio Ávalos
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 1027, building H, Third floor, 8380453, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, 8380453, Santiago, Chile
| | - Cristián Pereda
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 1027, building H, Third floor, 8380453, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, 8380453, Santiago, Chile
| | - Iván Gallegos
- Pathological Anatomy Service, Clinic Hospital, Universidad de Chile, 8380456, Santiago, Chile
| | - Fermín Eduardo González
- Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, 8380453, Santiago, Chile.,Department of Conservative Dentistry, Faculty of Dentistry, Universidad de Chile, 8380492, Santiago, Chile
| | - Mercedes Natalia López
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 1027, building H, Third floor, 8380453, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, 8380453, Santiago, Chile
| | - Jean Michel Butte
- Department of Surgery, Fundación Arturo López Pérez, Institute of Oncology, 7500921, Santiago, Chile
| | - Juan Carlos Roa
- Department of Pathology, School of Medicine, Pontificia Universidad Católica de Chile, 8330023, Santiago, Chile.,Center for Investigation in Translational Oncology (CITO), Advanced Center for Chronic Diseases (ACCDiS), School of Medicine, Pontificia Universidad Católica de Chile, 8330023, Santiago, Chile
| | - Paula Fluxá
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 1027, building H, Third floor, 8380453, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, 8380453, Santiago, Chile
| | - Flavio Salazar-Onfray
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 1027, building H, Third floor, 8380453, Santiago, Chile. .,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, 8380453, Santiago, Chile.
| |
Collapse
|
11
|
The Correlation Between Serum Chemokines and Clinical Outcome in Patients with Advanced Biliary Tract Cancer. Transl Oncol 2018; 11:353-357. [PMID: 29448202 PMCID: PMC5852407 DOI: 10.1016/j.tranon.2018.01.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/16/2018] [Accepted: 01/16/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Biliary tract cancers (BTCs) are known to have a dismal prognosis. A number of chemokines play important roles in the progress of BTCs. However, the serum levels of chemokines in BTCs have not yet been explored. METHODS The sera of healthy donors (n = 8) and patients with BTCs who were enrolled in second line sunitinib trials (n = 27) were collected. The concentrations of three kinds of chemokines (CXCL5, CXCL8 and CXCL12) were measured using ELISA assay. The median concentrations of chemokines were compared between healthy donors and BTC patients and the role of chemokines as a prognostic biomarker was examined. RESULTS BTC patients generally had higher serum levels of CXCL5 and CXCL12 compared to healthy donors. Patients with cholangiocarcinoma showed significantly higher levels of serum CXCL12 than patients with gallbladder cancer. In survival analysis, only CXCL12 level showed a prognostic impact on overall survival (median OS: 6.9 vs. 0.9 months in low CXCL12 vs. high CXCL12, respectively; P = .008). High CXCL5 levels were also correlated with poor survival without statistical insignificance (median OS: 6.2 vs. 2.0 months in low CXCL5 vs. high CXCL5, respectively; P = .070). CONCLUSIONS There was a significant difference in OS according to the level of CXCL12, suggesting that serum CXCL12 levels may be a useful surrogate marker for clinical outcome in advanced BTCs.
Collapse
|
12
|
Sharma A, Sharma KL, Gupta A, Yadav A, Kumar A. Gallbladder cancer epidemiology, pathogenesis and molecular genetics: Recent update. World J Gastroenterol 2017; 23:3978-3998. [PMID: 28652652 PMCID: PMC5473118 DOI: 10.3748/wjg.v23.i22.3978] [Citation(s) in RCA: 234] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Revised: 02/01/2017] [Accepted: 06/01/2017] [Indexed: 02/06/2023] Open
Abstract
Gallbladder cancer is a malignancy of biliary tract which is infrequent in developed countries but common in some specific geographical regions of developing countries. Late diagnosis and deprived prognosis are major problems for treatment of gallbladder carcinoma. The dramatic associations of this orphan cancer with various genetic and environmental factors are responsible for its poorly defined pathogenesis. An understanding to the relationship between epidemiology, molecular genetics and pathogenesis of gallbladder cancer can add new insights to its undetermined pathophysiology. Present review article provides a recent update regarding epidemiology, pathogenesis, and molecular genetics of gallbladder cancer. We systematically reviewed published literature on gallbladder cancer from online search engine PubMed (http://www.ncbi.nlm.nih.gov/pubmed). Various keywords used for retrieval of articles were Gallbladder, cancer Epidemiology, molecular genetics and bullion operators like AND, OR, NOT. Cross references were manually searched from various online search engines (http://www.ncbi.nlm.nih.gov/pubmed,https://scholar.google.co.in/, http://www.medline.com/home.jsp). Most of the articles published from 1982 to 2015 in peer reviewed journals have been included in this review.
Collapse
|
13
|
A meta-analysis of CXCL12 expression for cancer prognosis. Br J Cancer 2017; 117:124-135. [PMID: 28535157 PMCID: PMC5520200 DOI: 10.1038/bjc.2017.134] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 04/08/2017] [Accepted: 04/24/2017] [Indexed: 02/07/2023] Open
Abstract
Background: CXCL12 (SDF1) is reported to promote cancer progression in several preclinical models and this is corroborated by the analysis of human tissue specimens. However, the relationship between CXCL12 expression and cancer survival has not been systematically assessed. Methods: We conducted a systematic review and meta-analysis of studies that evaluated the association between CXCL12 expression and cancer survival. Results: Thirty-eight studies inclusive of 5807 patients were included in the analysis of overall, recurrence-free or cancer-specific survival, the majority of which were retrospective. The pooled hazard ratios (HRs) for overall and recurrence-free survival in patients with high CXCL12 expression were 1.39 (95% CI: 1.17–1.65, P=0.0002) and 1.12 (95% CI: 0.82–1.53, P=0.48) respectively, but with significant heterogeneity between studies. On subgroup analysis by cancer type, high CXCL12 expression was associated with reduced overall survival in patients with oesophagogastric (HR 2.08; 95% CI: 1.31–3.33, P=0.002), pancreatic (HR 1.54; 95% CI: 1.21–1.97, P=0.0005) and lung cancer (HR 1.37; 95% CI: 1.08–1.75, P=0.01), whereas in breast cancer patients high CXCL12 expression conferred an overall survival advantage (HR 0.5; 95% CI: 0.38–0.66, P<0.00001). Conclusions: Determination of CXCL12 expression has the potential to be of use as a cancer biomarker and adds prognostic information in various cancer types. Prospective or prospective–retrospective analyses of CXCL12 expression in clearly defined cancer cohorts are now required to advance our understanding of the relationship between CXCL12 expression and cancer outcome.
Collapse
|
14
|
Teng F, Tian WY, Wang YM, Zhang YF, Guo F, Zhao J, Gao C, Xue FX. Cancer-associated fibroblasts promote the progression of endometrial cancer via the SDF-1/CXCR4 axis. J Hematol Oncol 2016; 9:8. [PMID: 26851944 PMCID: PMC4744391 DOI: 10.1186/s13045-015-0231-4] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Accepted: 12/30/2015] [Indexed: 12/14/2022] Open
Abstract
Background Cancer-associated fibroblasts (CAFs) are believed to play an essential role in cancer initiation and development. However, little research has been undertaken to evaluate the role of CAFs in endometrial cancer (EC) progression. We aim to detect the functional contributions of CAFs to promote progression of EC. Methods Stromal fibroblasts were isolated from endometrioid adenocarcinomas and normal endometrial tissues. The conditioned media of cultured CAFs and normal fibroblasts (NFs) were collected to detect the level of stromal cell-derived factor-1alpha (SDF-1α), macrophage chemoattractant protein-1 (MCP-1), migration inhibitory factor (MIF), colony stimulating factor-1 (CSF-1), and interleukin-1 (IL-1) by ELISA. The CAFs or NFs were cocultured with EC cell lines to determine the proliferation, migration, and invasion by MTT assays and transwell chambers. Xenograft models were used to observe tumor growth. Matrix metalloproteinases (MMP)-2 and MMP-9 activity was evaluated by zymography. AMD3100 (a chemokine receptor 4 (CXCR4) antagonist) was used to block the SDF-1/CXCR4 axis. Neutralizing antibodies were used to detect PI3K/Akt and MAPK/Erk pathways by western blotting. SDF-1α and CXCR4 expressions were analyzed in xenotransplanted tumors and 348 cases by immunohistochemistry. Results CAFs promoted proliferation, migration, and invasion as well as in vivo tumorigenesis of admixed EC cells significantly more than NFs by secreting SDF-1α. These effects were significantly inhibited by AMD3100. CAFs promoted EC progression via the SDF-1α/CXCR4 axis to activate the PI3K/Akt and MAPK/Erk signalings in a paracrine-dependent manner or increase MMP-2 and MMP-9 secretion in an autocrine-dependent manner. SDF-1α and CXCR4 expression upregulation accompanied clinical EC development and progression. High SDF-1α expression levels were associated with deep myometrial invasion, lymph node metastasis, and poor prognosis in EC. Conclusions Our data indicated that CAFs derived from EC tissues promoted EC progression via the SDF-1/CXCR4 axis in a paracrine- or autocrine-dependent manner. SDF-1α is a novel independent poor prognostic factor for EC patients’ survival. Targeting the SDF-1/CXCR4 axis might provide a novel therapeutic strategy for EC treatment.
Collapse
Affiliation(s)
- Fei Teng
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, NO 154, Anshan Road, He Ping District, Tianjin, 300052, China.
| | - Wen-Yan Tian
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, NO 154, Anshan Road, He Ping District, Tianjin, 300052, China.
| | - Ying-Mei Wang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, NO 154, Anshan Road, He Ping District, Tianjin, 300052, China.
| | - Yan-Fang Zhang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, NO 154, Anshan Road, He Ping District, Tianjin, 300052, China.
| | - Fei Guo
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, NO 154, Anshan Road, He Ping District, Tianjin, 300052, China.
| | - Jing Zhao
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, NO 154, Anshan Road, He Ping District, Tianjin, 300052, China.
| | - Chao Gao
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, NO 154, Anshan Road, He Ping District, Tianjin, 300052, China.
| | - Feng-Xia Xue
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, NO 154, Anshan Road, He Ping District, Tianjin, 300052, China.
| |
Collapse
|
15
|
Huang J, Lin H, En Lin M. EVALUATION THE EXPRESSION OF THREE GENES TO EPITHELIAL OVARIAN CANCER RISK IN CHINESE POPULATION. AFRICAN JOURNAL OF TRADITIONAL, COMPLEMENTARY, AND ALTERNATIVE MEDICINES : AJTCAM 2016; 13:81-87. [PMID: 28852723 PMCID: PMC5566156 DOI: 10.21010/ajtcam.v13i4.12] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Ovarian cancer is associated with poor survival, because patients are diagnosed at an advanced stage of the disease, and in addition, tumors develop chemoresistance, which carries a poor prognosis for the patient. MATERIAL AND METHODS We hypothesize that high expression of SDF-1, survivin and smac is associated with ovarian cancers development and could be used as a biomarker to identify this disease. The expressions of SDF-1, survivin and smac in normal ovarian (NO) tissue, benign tumor (BT) tissue and epithelial ovarian cancer (EOC) tissue were immunohistochemically analysed. RESULTS Positive expressions of SDF-1, survivin and smac were significantly higher in EOC tissue than those in NO and BT tissues. SDF-1 expressions were significantly more weaker in advanced ovarian carcinomas (FIGO stage III-IV), and in high-grade carcinomas. There was a positive correlation between EOC patients with lymph node metastasis and with ascites and SDF-1 positivity (P < 0.05). Survivin expressions were significantly more stronger in advanced ovarian carcinomas (FIGO stage III-IV), and in high-grade carcinomas. There was a positive correlation between EOC patients with lymph node metastasis and with ascites and surviving positivity (P < 0.05). Smac expressions were significantly more stronger in advanced ovarian carcinomas (FIGO stage III-IV), and in high-grade carcinomas. There was a positive correlation between EOC patients with lymph node metastasis and with ascites and smac positivity (P < 0.05). CONCLUSION These results indicate that SDF-1, surviving and smac are closely associated with EOC metastasis.
Collapse
Affiliation(s)
- Ju Huang
- Department of Gynaecology, The First Affiliated Hospital of Shantou University Medical College, Shantou, PR China
| | - Hao Lin
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University
| | - Ming En Lin
- Department of Urology, The First Affiliated Hospital of Shantou University Medical College, Shantou, PR China
- Corresponding author E-mail: (ME Lin)
| |
Collapse
|
16
|
Ehling J, Tacke F. Role of chemokine pathways in hepatobiliary cancer. Cancer Lett 2015; 379:173-83. [PMID: 26123664 DOI: 10.1016/j.canlet.2015.06.017] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 06/09/2015] [Indexed: 12/17/2022]
Abstract
Persistent hepatic inflammation resulting from hepatitis B or C virus infections (HBV or HCV, respectively), obesity-associated non-alcoholic steatohepatitis (NASH) or alcohol abuse is a hallmark feature of chronic liver diseases and appears to be an essential prerequisite of hepatocarcinogenesis. The inflammatory processes in the liver are regulated by various chemokines, which orchestrate the interaction between parenchymal liver cells, Kupffer cells (resident macrophages), hepatic stellate cells (HSC), endothelial cells, and infiltrating immune cells. In consequence, these cellular interactions result in the re-modeling of the hepatic microenvironment toward a pro-inflammatory, pro-fibrotic, pro-angiogenic and thus pre-neoplastic milieu. Once developed, liver neoplasms provoke pro- and anti-tumor immune responses that are also critically regulated through differential activation of chemokine pathways. With respect to hepatobiliary cancers, including hepatocellular carcinoma (HCC), gallbladder cancer and cholangiocellular carcinoma (cholangiocarcinoma), together belonging to the highest causes of cancer-related deaths worldwide, this review article will give an overview of chemokine pathways involved in both the establishment of a pro-tumorigenic microenvironment as well as the development and progression of hepatobiliary cancer. Pharmaceutical targeting of chemokine pathways is a promising approach to treat or even prevent hepatobiliary cancer.
Collapse
Affiliation(s)
- Josef Ehling
- Department of Experimental Molecular Imaging, Helmholtz Institute for Biomedical Engineering, Medical Faculty, RWTH University, Aachen, Germany
| | - Frank Tacke
- Department of Medicine III, Medical Faculty, RWTH University, Aachen, Germany.
| |
Collapse
|
17
|
Kim HY, Moon JY, Ryu H, Choi YS, Song IC, Lee HJ, Yun HJ, Kim S, Jo DY. Bortezomib inhibits the survival and proliferation of bone marrow stromal cells. Blood Res 2015; 50:87-96. [PMID: 26157778 PMCID: PMC4486164 DOI: 10.5045/br.2015.50.2.87] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Revised: 05/24/2015] [Accepted: 05/27/2015] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Bortezomib is widely used for the treatment of multiple myeloma. Bone marrow stromal cells (BMSCs) endow myeloma cells with survival and growth advantages. However, the influence of bortezomib on BMSCs is not well elucidated. We examined the effects of bortezomib on the survival and growth of BMSCs in vitro. METHODS The effects of bortezomib on the survival and proliferation of the BMSC MS-5 cell line and on BMSCs obtained from healthy individuals (N=4) and newly diagnosed myeloma patients (N=5) were investigated in vitro. Transmembrane cell migration was evaluated using the Transwell system. A short interfering RNA strategy was used to knock down the expression of chemokine (CXC motif) ligand 12 (CXCL12) mRNA. To examine the effects of bortezomib-exposed BMSCs on the migration and localization of myeloma cells, MS-5 monolayers were treated with bortezomib for 24 hr, washed, and then overlaid with human RPMI8226 myeloma cells. RESULTS Bortezomib inhibited BMSC proliferation in a concentration-dependent manner, and induced cellular apoptosis. Bortezomib decreased CXCL12 production by BMSCs. Knockdown of CXCL12 mRNA in BMSCs revealed that CXCL12 served as an autocrine growth factor. Short-term bortezomib treatment of BMSC monolayers reduced the tendency of myeloma cells to locate to positions under the monolayers. CONCLUSION Bortezomib inhibits the survival and growth of BMSCs via downregulation of CXCL12, which may contribute to the clinical effects of this agent.
Collapse
Affiliation(s)
- Ha-Yon Kim
- Department of Drug Activity, New Drug Development Center, Medical Innovation Foundation, Osong, Korea
| | - Ji-Young Moon
- Department of Internal Medicine, School of Medicine, Chungnam National University, Daejeon, Korea
| | - Haewon Ryu
- Department of Internal Medicine, School of Medicine, Chungnam National University, Daejeon, Korea
| | - Yoon-Seok Choi
- Department of Internal Medicine, School of Medicine, Chungnam National University, Daejeon, Korea
| | - Ik-Chan Song
- Department of Internal Medicine, School of Medicine, Chungnam National University, Daejeon, Korea
| | - Hyo-Jin Lee
- Department of Internal Medicine, School of Medicine, Chungnam National University, Daejeon, Korea
| | - Hwan-Jung Yun
- Department of Internal Medicine, School of Medicine, Chungnam National University, Daejeon, Korea
| | - Samyong Kim
- Department of Internal Medicine, School of Medicine, Chungnam National University, Daejeon, Korea
| | - Deog-Yeon Jo
- Department of Internal Medicine, School of Medicine, Chungnam National University, Daejeon, Korea
| |
Collapse
|
18
|
Zhang C, Li J, Han Y, Jiang J. A meta-analysis for CXCR4 as a prognostic marker and potential drug target in non-small cell lung cancer. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:3267-78. [PMID: 26150700 PMCID: PMC4484670 DOI: 10.2147/dddt.s81564] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Background Recent reports have shown that C-X-C chemokine receptor type 4 (CXCR4) is a candidate oncogene in several types of human tumors, including non-small cell lung cancer (NSCLC). However, the correlation between CXCR4 expression and clinicopathological characteristics of NSCLC remains controversial and has not been emphasized. The aim of this study is to quantitatively evaluate the association of CXCR4 expression with the incidence of NSCLC and clinicopathological characteristics by performing a meta-analysis. Methods A detailed literature search was carried out for related research publications. Only articles in which CXCR4 expression was detected by immunohistochemical staining were included. Odds ratio (OR) and hazard ratio (HR) with 95% confidence intervals (CIs) were calculated and summarized. Results Final analysis of 1,872 NSCLC patients from 19 eligible studies was performed. We observed that CXCR4 expression was significantly higher in NSCLC than in normal lung tissue, based on the pooled OR from ten studies, including 678 NSCLCs and 189 normal lung tissues (OR =16.66, 95% CI =6.94–40.02, P<0.00001). CXCR4 expression was also significantly associated with clinical stages, metastatic status, and overall survival (OS) in NSCLC patients. In addition, CXCR4 mRNA high expression was found to correlate with worse OS of all NSCLC patients followed for 20 years, HR =1.24, P=0.0047. Conclusion The present meta-analysis indicated that CXCR4 protein expression is associated with an increased risk and worse survival in NSCLC patients. The aberrant CXCR4 protein and mRNA expression play an important role in the carcinogenesis and metastasis of NSCLC.
Collapse
Affiliation(s)
- Changyuan Zhang
- Department of Cardiothoracic Surgery, Inner Mongolia Autonomous Region People's Hospital, Inner Mongolia, People's Republic of China
| | - Jie Li
- Department of Oncology, Capital Medical University, Beijing, People's Republic of China
| | - Yi Han
- Department of Thoracic Surgery, Beijing Chest Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Jian Jiang
- Department of Thoracic Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
19
|
Sugihara H, Ishimoto T, Yasuda T, Izumi D, Eto K, Sawayama H, Miyake K, Kurashige J, Imamura Y, Hiyoshi Y, Iwatsuki M, Iwagami S, Baba Y, Sakamoto Y, Miyamoto Y, Yoshida N, Watanabe M, Takamori H, Baba H. Cancer-associated fibroblast-derived CXCL12 causes tumor progression in adenocarcinoma of the esophagogastric junction. Med Oncol 2015; 32:618. [DOI: 10.1007/s12032-015-0618-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 04/10/2015] [Indexed: 01/18/2023]
|
20
|
Mayr C, Neureiter D, Pichler M, Berr F, Wagner A, Kiesslich T, Namberger K. Cytotoxic effects of chemokine receptor 4 inhibition by AMD3100 in biliary tract cancer cells: Potential drug synergism with gemcitabine. Mol Med Rep 2015; 12:2247-52. [PMID: 25846744 DOI: 10.3892/mmr.2015.3589] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 02/27/2015] [Indexed: 12/21/2022] Open
Abstract
Biliary tract cancer (BTC) remains one of the most life-threatening types of cancer due to the lack of efficient therapies. Advanced tumour stages at the point of diagnosis and high chemoresistance are two of the predominant reasons for a 5-year survival rate of only ~5%. The present study investigated the effect of the chemokine receptor 4 (CXCR4) inhibitor AMD3100 (Plerixafor), alone and in combination with standard gemcitabine chemotherapy, on the proliferation of BTC cells. The expression of CXCR4 was analysed by reverse transcription-quantitative polymerase chain reaction in eight heterogeneously differentiated BTC cell lines. The effects of treatment with the CXCR4 antagonist, AMD3100, on cell viability and anchorage-independent growth, and the possible synergistic cytotoxic effects of AMD3100 with standard chemotherapeutics were assessed. The expression of CXCR4 was observed to a variable extent in all eight BTC cell lines, with SkChA-1 cells exhibiting the highest expression levels. Treatment with AMD3100 led to a marginal decrease in cell viability in the cell lines, with the exception of the CCSW-1 cells, and a significant reduction in the GBC, MzChA-1, SkChA.-1 and TFK-1 cell lines. The combined treatment of the SkChA-1 cells with varying concentrations of AMD3100 and standard gemcitabine chemotherapy revealed a more marked overall cytotoxicity, indicating a potential synergistic effect. In addition, AMD3100 significantly reduced anchorage-independent growth in the SkChA-1 cells. Overall, the results of the present study suggest that the inhibition of CXCR4 by AMD3100, in combination with gemcitabine, may be a suitable strategy for the future therapy of BTC.
Collapse
Affiliation(s)
- Christian Mayr
- Department of Internal Medicine I, Paracelsus Medical University/Salzburger Landeskliniken, Salzburg A‑5020, Austria
| | - Daniel Neureiter
- Institute of Pathology, Paracelsus Medical University/Salzburger Landeskliniken, Salzburg A‑5020, Austria
| | - Martin Pichler
- Department of Experimental Therapeutics, UT MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Frieder Berr
- Department of Internal Medicine I, Paracelsus Medical University/Salzburger Landeskliniken, Salzburg A‑5020, Austria
| | - Andrej Wagner
- Department of Internal Medicine I, Paracelsus Medical University/Salzburger Landeskliniken, Salzburg A‑5020, Austria
| | - Tobias Kiesslich
- Department of Internal Medicine I, Paracelsus Medical University/Salzburger Landeskliniken, Salzburg A‑5020, Austria
| | - Konrad Namberger
- Department of Hematology and Oncology, Klinikum Braunschweig, Braunschweig D‑38118, Germany
| |
Collapse
|
21
|
Shynlova O, Dorogin A, Li Y, Lye S. Inhibition of infection-mediated preterm birth by administration of broad spectrum chemokine inhibitor in mice. J Cell Mol Med 2014; 18:1816-29. [PMID: 24894878 PMCID: PMC4196657 DOI: 10.1111/jcmm.12307] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 03/25/2014] [Indexed: 12/15/2022] Open
Abstract
Preterm birth (PTB) is the single most important cause of perinatal and infant mortality worldwide. Maternal infection can result in PTB. We investigated the ability of a Broad Spectrum Chemokine Inhibitor (BSCI) to prevent infection-induced PTB in mice. PTB was initiated in pregnant mice by intraperitoneal injection of lipopolysaccharide (LPS; 50 μg). Half the mice received BSCI (10 mg/kg) 24 hrs prior to and immediately before LPS administration. The impact of LPS alone or LPS plus BSCI was assessed on (i) injection-to-delivery interval, foetal survival rate, placental and neonates' weight; (ii) amniotic fluid and maternal plasma cytokine levels (by Luminex assay); foetal and maternal tissue cytokine gene expression levels (by Real-Time RT-PCR); (iii) immune cells infiltration into the uterine tissue (by stereological immunohistochemistry). Pre-treatment with BSCI (i) decreased LPS-induced PTB (64% versus 100%, P < 0.05); (ii) significantly attenuated cytokine/chemokine expression in maternal tissues (plasma, liver, myometrium, decidua); (iii) significantly decreased neutrophil infiltration in the mouse myometrium. BSCI-treated mice in which PTB was delayed till term had live foetuses with normal placental and foetal weight. BSCI represents a promising new class of therapeutics for PTB. In a mouse model of preterm labour, BCSI suppresses systemic inflammation in maternal tissues which resulted in the reduced incidence of LPS-mediated PTB. These data provide support for efforts to target inflammatory responses as a means of preventing PTB.
Collapse
Affiliation(s)
- Oksana Shynlova
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada; Department of Obstetrics & Gynecology, University of Toronto, Toronto, ON, Canada
| | | | | | | |
Collapse
|
22
|
Lee HJ, Song IC, Yun HJ, Jo DY, Kim S. CXC chemokines and chemokine receptors in gastric cancer: From basic findings towards therapeutic targeting. World J Gastroenterol 2014; 20:1681-1693. [PMID: 24587647 PMCID: PMC3930968 DOI: 10.3748/wjg.v20.i7.1681] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 10/01/2013] [Accepted: 11/13/2013] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer is the fourth most common cancer, and the second-highest cause of cancer-related deaths worldwide. Despite extensive research to identify novel diagnostic and therapeutic agents, patients with advanced gastric cancer suffer from a poor quality of life and poor prognosis, and treatment is dependent mainly on conventional cytotoxic chemotherapy. To improve the quality of life and survival of gastric cancer patients, a better understanding of the underlying molecular pathologies, and their application towards the development of novel targeted therapies, is urgently needed. Chemokines are a group of small proteins associated with cytoskeletal rearrangements, the directional migration of several cell types during development and physiology, and the host immune response via interactions with G-protein coupled receptors. There is also growing evidence to suggest that chemokines not only play a role in the immune system, but are also involved in the development and progression of tumors. In gastric cancer, CXC chemokines and chemokine receptors regulate the trafficking of cells in and out of the tumor microenvironment. CXC chemokines and their receptors can also directly influence tumorigenesis by modulating tumor transformation, survival, growth, invasion and metastasis, as well as indirectly by regulating angiogenesis, and tumor-leukocyte interactions. In this review, we will focus on the roles of CXC chemokines and their receptors in the development, progression, and metastasis of gastric tumors, and discuss their therapeutic potential for gastric cancer.
Collapse
|
23
|
Gosalbez M, Hupe MC, Lokeshwar SD, Yates TJ, Shields J, Veerapen MK, Merseburger AS, Rosser CJ, Soloway MS, Lokeshwar VB. Differential expression of SDF-1 isoforms in bladder cancer. J Urol 2013; 191:1899-1905. [PMID: 24291546 DOI: 10.1016/j.juro.2013.11.053] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2013] [Indexed: 10/26/2022]
Abstract
PURPOSE SDF-1 is a ligand of the chemokine receptors CXCR4 and 7. The 6 known SDF-1 isoforms are generated by alternative mRNA splicing. While SDF-1 expression has been detected in various malignancies, only few groups have reported differential expression of SDF-1 isoforms and its clinical significance. We evaluated the expression of 3 SDF-1 isoforms (α, β and γ) in bladder cancer. MATERIALS AND METHODS Using quantitative polymerase chain reaction we measured SDF-1α, β and γ mRNA levels in 25 normal and 44 bladder cancer tissues, and in 210 urine specimens (28 normal, 74 benign, 57 bladder cancer, 35 bladder cancer history, 8 other cancer history and 8 other cancer) from consecutive patients. Levels were correlated with clinical outcome. RESULTS Of the SDF-1 isoforms only SDF-1β mRNA was significantly over expressed 2.5-fold to sixfold in bladder cancer compared to normal bladder tissues. SDF-1α was expressed in bladder tissues but SDF-1γ was undetectable. On multivariate analysis SDF-1β was an independent predictor of metastasis and disease specific mortality (p=0.017 and 0.043, respectively). In exfoliated urothelial cells only SDF-1β mRNA levels were differentially expressed with 91.2% sensitivity and 73.8% specificity for detecting bladder cancer. In patients with a bladder cancer history increased SDF-1β levels indicated a 4.3-fold increased risk of recurrence within 6 months (p=0.0001). CONCLUSIONS SDF-1 isoforms are differentially expressed in bladder tissues and exfoliated urothelial cells. SDF-1β mRNA levels in bladder cancer tissues predict a poor prognosis. Furthermore, SDF-1β mRNA levels in exfoliated cells detect bladder cancer with high sensitivity and they are a potential predictor of future recurrence.
Collapse
Affiliation(s)
- Miguel Gosalbez
- Department of Urology University of Miami - Miller School of Medicine, Miami, Florida, 33101
| | - Marie C Hupe
- Department of Urology University of Miami - Miller School of Medicine, Miami, Florida, 33101.,Department of Urology and Urologic Oncology Hannover Medical School (MHH), Hannover Germany
| | - Soum D Lokeshwar
- Sylvester Comprehensive Cancer Center, University of Miami - Miller School of Medicine, Miami, Florida, 33101
| | - Travis J Yates
- Department of Urology University of Miami - Miller School of Medicine, Miami, Florida, 33101
| | - John Shields
- Department of Urology University of Miami - Miller School of Medicine, Miami, Florida, 33101
| | - Muthu K Veerapen
- Department of Human Genetics and Hussman Institute of Human Genetics, University of Miami - Miller School of Medicine, Miami, Florida 33136
| | - Axel S Merseburger
- Department of Urology and Urologic Oncology Hannover Medical School (MHH), Hannover Germany
| | - Charles J Rosser
- University of Central Florida College of Medicine, Orlando, Florida
| | - Mark S Soloway
- Department of Urology University of Miami - Miller School of Medicine, Miami, Florida, 33101
| | - Vinata B Lokeshwar
- Department of Urology University of Miami - Miller School of Medicine, Miami, Florida, 33101.,Sylvester Comprehensive Cancer Center, University of Miami - Miller School of Medicine, Miami, Florida, 33101
| |
Collapse
|
24
|
Wu XS, Shi LB, Li ML, Ding Q, Weng H, Wu WG, Cao Y, Bao RF, Shu YJ, Ding QC, Mu JS, Gu J, Dong P, Liu YB. Evaluation of two inflammation-based prognostic scores in patients with resectable gallbladder carcinoma. Ann Surg Oncol 2013; 21:449-57. [PMID: 24081806 DOI: 10.1245/s10434-013-3292-z] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2013] [Indexed: 02/06/2023]
Abstract
BACKGROUND Survival after surgery for gallbladder cancer is generally poor. A number of inflammation-based prognostic scores have been established to help predict survival after surgery for several types of cancer. The objective of this study was to analyze and compare the utility of two inflammation-based prognostic scores, the Glasgow prognostic score (GPS) and the neutrophil-to-lymphocyte ratio (NLR), for predicting survival in patients with gallbladder cancer after surgery with curative intent. METHODS We retrospectively reviewed the medical records of 85 patients with histologically confirmed, resectable gallbladder carcinoma (GBC), who were to receive curative surgery in our department. Univariate and multivariate analyses were performed to evaluate the relationship between the variables to overall survival (OS). RESULTS A significant difference was detected in OS in patients with low and high GPS and NLR scores. Univariate analyses using clinicopathological characteristics revealed that tumor differentiation; tumor invasion; lymph node metastasis; tumor, node, metastasis classification system stage; positive margin status; combined common bile duct resection; serum levels of C-reactive protein, albumin, carbohydrate antigen 19-9 (CA19-9), carcinoembryonic antigen, and CA125; white blood cell count; and GPS and NLR were all associated with OS. Among these characteristics, multivariate analysis demonstrated that a high GPS was independently associated with poorer OS, together with tumor invasion, lymph node metastasis, and positive margin status. CONCLUSIONS GPS is superior to NLR with respect to its prognostic value for patients with GBC after surgery with curative intent. GPS is not only associated with tumor progression but is also an independent marker of poor prognosis.
Collapse
Affiliation(s)
- Xiang-Song Wu
- Department of General Surgery and Laboratory of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Institute of Biliary Tract Disease, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Qu QX, Huang Q, Xu J, Duan LT, Zhu YB, Zhang XG. CD40 signal regulates CXCR4 mediating ovarian carcinoma cell migration: implications for extrapelvic metastastic factors. Oncol Res 2013; 20:383-92. [PMID: 23924922 DOI: 10.3727/096504013x13657689382653] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Ovarian carcinomas are highly invasive, especially in the peritoneal cavity. SDF-1α and its receptor, CXCR4, play a crucial role in migration of cancer cells. Here, SDF-1α directed HO8910 cell migration, but not SKOV3 cells. After being educated to express CXCR4 in vivo or by treating with sCD40L, SDF-1α reexhibited the ability of directing SKOV3 cell migration, which could be antagonized by CXCR4-neutralizing antibody. Furthermore, concomitant expression of CXCR4/CD40 in ovarian carcinoma tissues had stronger correlation with pelvic metastasis than did each alone. It is suggest that SDF-1α acts through CXCR4 to induce ovarian cancer cell migration, which could be facilitated by CD40 activation. Simultaneously examining the expression of CXCR4 and CD40 will provide valuable diagnosis of pelvic metastasis for ovarian carcinomas.
Collapse
Affiliation(s)
- Qiu-Xia Qu
- Clinical Immunology Institute, The First Affiliated Hospital of Soochow University, Suzhou, China
| | | | | | | | | | | |
Collapse
|
26
|
Chronic inflammation and gallbladder cancer. Cancer Lett 2013; 345:242-8. [PMID: 23981574 DOI: 10.1016/j.canlet.2013.08.034] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 08/13/2013] [Accepted: 08/18/2013] [Indexed: 12/15/2022]
Abstract
Gallbladder cancer (GBC) is the most common biliary tract malignancy with an extremely poor prognosis. Epidemiological data have demonstrated that chronic inflammation resulting from infection of gallbladder or gallstones predispose individuals to GBC. Recent studies have begun to elucidate molecular mechanisms underlying the development of GBC in the setting of chronic inflammation. It is possible that persistently local inflammatory reactions may contribute to the development and progression of GBC through inducing genetic alterations, and subsequent promoting survival and proliferation of mutated sells, inhibiting apoptosis, stimulating angiogenesis and metastasis. This article reviews the current understanding of the involvement of chronic inflammation in gallbladder tumorigenesis.
Collapse
|
27
|
Oh YS, Kim HY, Song IC, Yun HJ, Jo DY, Kim S, Lee HJ. Hypoxia induces CXCR4 expression and biological activity in gastric cancer cells through activation of hypoxia-inducible factor-1α. Oncol Rep 2012; 28:2239-46. [PMID: 23023480 DOI: 10.3892/or.2012.2063] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Accepted: 09/07/2012] [Indexed: 01/13/2023] Open
Abstract
Given the important role of CXCR4 in cancer metastasis, microenvironmental factors that modulate CXCR4 may have an impact on the process of tumor expansion. Hypoxia is a common feature of solid tumors and a significant microenvironmental factor that drives aggressive behavior. CXCR4 is upregulated in several cancer cells under hypoxic conditions, suggesting a relationship between tumor hypoxia and CXCR4. However, the role of hypoxia in regulating CXCR4 in gastric cancer remains poorly understood. KATO III gastric cancer cells were exposed to hypoxia or normoxia. CXCR4 expression in cells transfected with shRNA specific for HIF-1α was investigated by western blotting and flow cytometry. Wound healing, migration and invasion assays were used to assess cell motility and the chemotactic response to CXCL12, a major CXCR4 ligand. CXCR4 expression at the protein level and in the cell membrane was significantly increased in KATO III cells following exposure to hypoxia. This upregulation of CXCR4 was implicated in increased cell motility and enhanced chemotactic responses (migration and invasion) to CXCL12 treatment in vitro. The increases in CXCR4 expression and metastatic potential in gastric cancer cells exposed to hypoxia were blocked by HIF-1α-specific shRNA. Our results indicate that hypoxia upregulates CXCR4 in gastric cancer cells in a HIF-1α-dependent manner, and that upregulation of CXCR4 plays a role in cancer cell migration and invasion. Thus, disrupting the hypoxia-HIF-1α-CXCR4 axis could be an attractive therapeutic strategy for the treatment of gastric cancer.
Collapse
Affiliation(s)
- Yoon Suk Oh
- Department of Internal Medicine and Cancer Research Institute, Chungnam National University School of Medicine, Daejeon 301-131, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|