1
|
The chronological evolution of fluorescent GPCR probes for bioimaging. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2023.215040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
2
|
Kim NY, Jung YY, Yang MH, Um JY, Sethi G, Ahn KS. Isoimperatorin down-regulates epithelial mesenchymal transition through modulating NF-κB signaling and CXCR4 expression in colorectal and hepatocellular carcinoma cells. Cell Signal 2022; 99:110433. [DOI: 10.1016/j.cellsig.2022.110433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 07/17/2022] [Accepted: 08/01/2022] [Indexed: 11/03/2022]
|
3
|
Budi HS, Ahmad FN, Achmad H, Ansari MJ, Mikhailova MV, Suksatan W, Chupradit S, Shomali N, Marofi F. Human epidermal growth factor receptor 2 (HER2)-specific chimeric antigen receptor (CAR) for tumor immunotherapy; recent progress. Stem Cell Res Ther 2022; 13:40. [PMID: 35093187 PMCID: PMC8800342 DOI: 10.1186/s13287-022-02719-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 01/11/2022] [Indexed: 12/17/2022] Open
Abstract
Due to the overexpression or amplification of human epidermal growth factor receptor 2 (HER2) with poor prognosis in a myriad of human tumors, recent studies have focused on HER2-targeted therapies. Deregulation in HER2 signaling pathways is accompanied by sustained tumor cells growth concomitant with their migration and also tumor angiogenesis and metastasis by stimulation of proliferation of a network of blood vessels. A large number of studies have provided clear evidence that the emerging HER2-directed treatments could be the outcome of patients suffering from HER2 positive breast and also gastric/gastroesophageal cancers. Thanks to its great anti-tumor competence, immunotherapy using HER2-specific chimeric antigen receptor (CAR) expressing immune cell has recently attracted increasing attention. Human T cells and also natural killer (NK) cells can largely be found in the tumor microenvironment, mainly contributing to the tumor immune surveillance. Such properties make them perfect candidate for genetically modification to express constructed CARs. Herein, we will describe the potential targets of the HER2 signaling in tumor cells to clarify HER2-mediated tumorigenesis and also discuss recent findings respecting the HER2-specific CAR-expressing immune cells (CAR T and CAR NK cell) for the treatment of HER2-expressing tumors.
Collapse
Affiliation(s)
- Hendrik Setia Budi
- Department of Oral Biology, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, 60132 Indonesia
| | | | - Harun Achmad
- Department of Pediatric Dentistry, Faculty of Dentistry, Hasanuddin University, Makassar, Indonesia
| | - Mohammad Javed Ansari
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | | | - Wanich Suksatan
- Faculty of Nursing, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, 10210 Thailand
| | - Supat Chupradit
- Department of Occupational Therapy, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200 Thailand
| | - Navid Shomali
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Faroogh Marofi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
4
|
Baxter MA, Middleton F, Cagney HP, Petty RD. Resistance to immune checkpoint inhibitors in advanced gastro-oesophageal cancers. Br J Cancer 2021; 125:1068-1079. [PMID: 34230609 PMCID: PMC8505606 DOI: 10.1038/s41416-021-01425-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 03/17/2021] [Accepted: 04/22/2021] [Indexed: 12/13/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have altered the treatment paradigm across a range of tumour types, including gastro-oesophageal cancers. For patients with any cancer type who respond, ICIs can confer long-term disease control and significantly improve survival and quality of life, but for patients with gastro-oesophageal cancer, ICIs can be transformative, as durable responses in advanced disease have hitherto been rare, especially in those patients who are resistant to first-line cytotoxic therapies. Results from trials in patients with advanced-stage gastro-oesophageal cancer have raised hopes that ICIs will be successful as adjuvant and neoadjuvant treatments in early-stage disease, when the majority of patients relapse after potential curative treatments, and several trials are ongoing. Unfortunately, however, ICI-responding patients appear to constitute a minority subgroup within gastro-oesophageal cancer, and resistance to ICI therapy (whether primary or acquired) is common. Understanding the biological mechanisms of ICI resistance is a current major research challenge and involves investigation of both tumour and patient-specific factors. In this review, we discuss the mechanisms underlying ICI resistance and their potential specific applications of this knowledge towards precision medicine strategies in the management of gastro-oesophageal cancers in clinical practice.
Collapse
Affiliation(s)
- Mark A Baxter
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK.
- Tayside Cancer Centre, Ninewells Hospital and Medical School, NHS Tayside, Dundee, UK.
| | - Fearghas Middleton
- Tayside Cancer Centre, Ninewells Hospital and Medical School, NHS Tayside, Dundee, UK
| | - Hannah P Cagney
- School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Russell D Petty
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK.
- Tayside Cancer Centre, Ninewells Hospital and Medical School, NHS Tayside, Dundee, UK.
| |
Collapse
|
5
|
O'Donovan C, Davern M, Donlon NE, Lysaght J, Conroy MJ. Chemokine-targeted therapies: An opportunity to remodel immune profiles in gastro-oesophageal tumours. Cancer Lett 2021; 521:224-236. [PMID: 34506844 DOI: 10.1016/j.canlet.2021.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/18/2021] [Accepted: 09/05/2021] [Indexed: 02/07/2023]
Abstract
Immunotherapies are transforming outcomes for many cancer patients and are quickly becoming the fourth pillar of cancer therapy. However, their efficacy of only ∼25% in gastro-oesophageal cancer has been disappointing. This is attributed to factors such as insufficient patient stratification and the pro-tumourigenic immune landscape of gastro-oesophageal tumours. The chemokine profiles of solid tumours and the availability of effector immune cells greatly influence the immune infiltrate, producing 'cold' or 'immune-excluded' tumours in which immunotherapies are unable to reinvigorate the immune response. Other biological functions for chemokines have emerged, such as promoting cell survival, polarising T cell responses, and supporting several hallmarks of cancer. Therefore, chemokine networks may be exploited with therapeutic intent to mobilise and polarise anti-tumour immune cells, with further utility as combination treatments to augment the efficacy of current cancer immunotherapies. Few studies have demonstrated the clinical benefit of chemokine-targeted therapies as monotherapies, and this review proposes their consideration as combination treatments. Herein, we explore the anti-tumour and pro-tumour implications of chemokine signalling in gastro-oesophageal cancer and discuss their value as prognostic and predictive biomarkers in response to treatment.
Collapse
Affiliation(s)
- Cillian O'Donovan
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute, St. James's Hospital Campus, Dublin 8, Ireland
| | - Maria Davern
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute, St. James's Hospital Campus, Dublin 8, Ireland
| | - Noel E Donlon
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute, St. James's Hospital Campus, Dublin 8, Ireland
| | - Joanne Lysaght
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute, St. James's Hospital Campus, Dublin 8, Ireland
| | - Melissa J Conroy
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute, St. James's Hospital Campus, Dublin 8, Ireland; Department of Physiology, School of Medicine, Trinity College, Dublin, Ireland.
| |
Collapse
|
6
|
Beukinga RJ, Wang D, Karrenbeld A, Dijksterhuis WPM, Faber H, Burgerhof JGM, Mul VEM, Slart RHJA, Coppes RP, Plukker JTM. Addition of HER2 and CD44 to 18F-FDG PET-based clinico-radiomic models enhances prediction of neoadjuvant chemoradiotherapy response in esophageal cancer. Eur Radiol 2020; 31:3306-3314. [PMID: 33151397 PMCID: PMC8043921 DOI: 10.1007/s00330-020-07439-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 09/21/2020] [Accepted: 10/21/2020] [Indexed: 01/08/2023]
Abstract
OBJECTIVES To assess the complementary value of human epidermal growth factor receptor 2 (HER2)-related biological tumor markers to clinico-radiomic models in predicting complete response to neoadjuvant chemoradiotherapy (NCRT) in esophageal cancer patients. METHODS Expression of HER2 was assessed by immunohistochemistry in pre-treatment tumor biopsies of 96 patients with locally advanced esophageal cancer. Five other potentially active HER2-related biological tumor markers in esophageal cancer were examined in a sub-analysis on 43 patients. Patients received at least four of the five cycles of chemotherapy and full radiotherapy regimen followed by esophagectomy. Three reference clinico-radiomic models based on 18F-FDG PET were constructed to predict pathologic response, which was categorized into complete versus incomplete (Mandard tumor regression grade 1 vs. 2-5). The complementary value of the biological tumor markers was evaluated by internal validation through bootstrapping. RESULTS Pathologic examination revealed 21 (22%) complete and 75 (78%) incomplete responders. HER2 and cluster of differentiation 44 (CD44), analyzed in the sub-analysis, were univariably associated with pathologic response. Incorporation of HER2 and CD44 into the reference models improved the overall performance (R2s of 0.221, 0.270, and 0.225) and discrimination AUCs of 0.759, 0.857, and 0.816. All models exhibited moderate to good calibration. The remaining studied biological tumor markers did not yield model improvement. CONCLUSIONS Incorporation of HER2 and CD44 into clinico-radiomic prediction models improved NCRT response prediction in esophageal cancer. These biological tumor markers are promising in initial response evaluation. KEY POINTS • A multimodality approach, integrating independent genomic and radiomic information, is promising to improve prediction of γpCR in patients with esophageal cancer. • HER2 and CD44 are potential biological tumor markers in the initial work-up of patients with esophageal cancer. • Prediction models combining 18F-FDG PET radiomic features with HER2 and CD44 may be useful in the decision to omit surgery after neoadjuvant chemoradiotherapy in patients with esophageal cancer.
Collapse
Affiliation(s)
- Roelof J Beukinga
- Medical Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, 9700 RB, Groningen, The Netherlands.
| | - Da Wang
- Department of Surgical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Biomedical Sciences of Cells and Systems, Section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Arend Karrenbeld
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Willemieke P M Dijksterhuis
- Department of Surgical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Hette Faber
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Johannes G M Burgerhof
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Véronique E M Mul
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Riemer H J A Slart
- Medical Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, 9700 RB, Groningen, The Netherlands.,Faculty of Science and Technology, Department of Biomedical Photonic Imaging, University of Twente, Enschede, The Netherlands
| | - Robert P Coppes
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - John Th M Plukker
- Department of Surgical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
7
|
Zhu L, Dong L, Feng M, Yang F, Jiang W, Huang Z, Liu F, Wang L, Wang G, Li Q. Profiles of autophagy-related genes in esophageal adenocarcinoma. BMC Cancer 2020; 20:943. [PMID: 32998713 PMCID: PMC7528598 DOI: 10.1186/s12885-020-07416-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 09/15/2020] [Indexed: 12/13/2022] Open
Abstract
Background Several studies have demonstrated autophagy was involved in the process of esophageal adenocarcinoma (EAC). The aim of this study was to explore autophagy-related genes (ARGs) correlated with overall survival (OS) in EAC patients. Methods Expressions of ARGs in EAC and normal samples were downloaded from TCGA database. GO and KEGG enrichment analyses were used to investigate the ARGs bioinformatics functions. Univariate and multivariate cox regressions were performed to identify prognostic ARGs and the independent risk factors. ROC curve was established to evaluate the feasibility to predict the prognosis. Finally, the correlations between ARGs and clinical features were further explored. In addition, significantly different ARGs were verified in EAC specimens and normal esophageal mucosal tissues. Results Thirty significantly different ARGs were selected from EAC and normal tissues. Functional enrichments showed these ARGs were mainly related apoptosis. Multivariate cox regression analyses demonstrated eight ARGs were significantly associated with OS. Among these eight genes, BECN1 (HR = 0.321, P = 0.046), DAPK1 (HR = 0.636, P = 0.025) and CAPN1 (HR = 0.395, P = 0.004) played protective roles in survival. Gender (HR = 0.225, P = 0.032), stage (HR = 5.841, P = 0.008) and risk score (HR = 1.131, P < 0.001) were independent prognostic risk factors. ROC curves showed better efficacy to predict survival using the risk score. Additionally, we found BECN1, DAPK1, VAMP7 and SIRT1 genes were correlated significantly with survival status, gender, primary tumor and tumor stage (all P < 0.05). The experimental results confirmed the BIRC5 was overexpressed and the ITPR1, PRKN were downregulated in the EAC tissues compared with the normal esophageal mucosal tissues (all P < 0.05). Conclusion Our findings suggested that autophagy was involved in the process of EAC. Several ARGs probably could serve as diagnostic and prognostic biomarkers and may help facilitate therapeutic targets in EAC patients.
Collapse
Affiliation(s)
- Lei Zhu
- Department of Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China.,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Lin Dong
- Department of Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China.,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Minghao Feng
- Department of Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China.,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Fugui Yang
- Department of Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China.,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Wenhao Jiang
- Department of Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China.,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Zhiyuan Huang
- Department of Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China.,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Fabing Liu
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China.,Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 200080, China
| | - Lingwei Wang
- Department of Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China.,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Guangxue Wang
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China.
| | - Qinchuan Li
- Department of Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China. .,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China.
| |
Collapse
|
8
|
Zhou C, Fan N, Liu F, Fang N, Plum PS, Thieme R, Gockel I, Gromnitza S, Hillmer AM, Chon SH, Schlösser HA, Bruns CJ, Zhao Y. Linking Cancer Stem Cell Plasticity to Therapeutic Resistance-Mechanism and Novel Therapeutic Strategies in Esophageal Cancer. Cells 2020; 9:1481. [PMID: 32560537 PMCID: PMC7349233 DOI: 10.3390/cells9061481] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/07/2020] [Accepted: 06/10/2020] [Indexed: 12/24/2022] Open
Abstract
Esophageal cancer (EC) is an aggressive form of cancer, including squamous cell carcinoma (ESCC) and adenocarcinoma (EAC) as two predominant histological subtypes. Accumulating evidence supports the existence of cancer stem cells (CSCs) able to initiate and maintain EAC or ESCC. In this review, we aim to collect the current evidence on CSCs in esophageal cancer, including the biomarkers/characterization strategies of CSCs, heterogeneity of CSCs, and the key signaling pathways (Wnt/β-catenin, Notch, Hedgehog, YAP, JAK/STAT3) in modulating CSCs during esophageal cancer progression. Exploring the molecular mechanisms of therapy resistance in EC highlights DNA damage response (DDR), metabolic reprogramming, epithelial mesenchymal transition (EMT), and the role of the crosstalk of CSCs and their niche in the tumor progression. According to these molecular findings, potential therapeutic implications of targeting esophageal CSCs may provide novel strategies for the clinical management of esophageal cancer.
Collapse
Affiliation(s)
- Chenghui Zhou
- Department of General, Visceral, Cancer and Transplantation Surgery, University Hospital Cologne, 50937 Cologne, Germany
| | - Ningbo Fan
- Department of General, Visceral, Cancer and Transplantation Surgery, University Hospital Cologne, 50937 Cologne, Germany
| | - Fanyu Liu
- Department of General, Visceral, Cancer and Transplantation Surgery, University Hospital Cologne, 50937 Cologne, Germany
- Interfaculty Institute for Cell Biology, University of Tübingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany
| | - Nan Fang
- Singleron Biotechnologies, Yaogu Avenue 11, Nanjing 210000, China
| | - Patrick S Plum
- Department of General, Visceral, Cancer and Transplantation Surgery, University Hospital Cologne, 50937 Cologne, Germany
- Institute of Pathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - René Thieme
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital of Leipzig, 4107 Leipzig, Germany
| | - Ines Gockel
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital of Leipzig, 4107 Leipzig, Germany
| | - Sascha Gromnitza
- Institute of Pathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Axel M Hillmer
- Institute of Pathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50937 Cologne, Germany
| | - Seung-Hun Chon
- Department of General, Visceral, Cancer and Transplantation Surgery, University Hospital Cologne, 50937 Cologne, Germany
| | - Hans A Schlösser
- Department of General, Visceral, Cancer and Transplantation Surgery, University Hospital Cologne, 50937 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50937 Cologne, Germany
| | - Christiane J Bruns
- Department of General, Visceral, Cancer and Transplantation Surgery, University Hospital Cologne, 50937 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50937 Cologne, Germany
| | - Yue Zhao
- Department of General, Visceral, Cancer and Transplantation Surgery, University Hospital Cologne, 50937 Cologne, Germany
| |
Collapse
|
9
|
New Insights on the Emerging Genomic Landscape of CXCR4 in Cancer: A Lesson from WHIM. Vaccines (Basel) 2020; 8:vaccines8020164. [PMID: 32260318 PMCID: PMC7349554 DOI: 10.3390/vaccines8020164] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 03/30/2020] [Accepted: 03/31/2020] [Indexed: 12/16/2022] Open
Abstract
Deciphering the molecular alterations leading to disease initiation and progression is currently crucial to identify the most relevant targets for precision therapy in cancer patients. Cancers express a complex chemokine network influencing leucocyte infiltration and angiogenesis. Moreover, malignant cells also express a selective repertoire of chemokine receptors that sustain their growth and spread. At present, different cancer types have been shown to overexpress C-X-C chemokine receptor type 4 (CXCR4) and to respond to its ligand C-X-C motif chemokine 12 (CXCL12). The CXCL12/CXCR4 axis influences cancer biology, promoting survival, proliferation, and angiogenesis, and plays a pivotal role in directing migration of cancer cells to sites of metastases, making it a prognostic marker and a therapeutic target. More recently, mutations in the C-terminus of CXCR4 have been identified in the genomic landscape of patients affected by Waldenstrom's macroglobulinemia, a rare B cell neoplasm. These mutations closely resemble those occurring in Warts, Hypogammaglobulinemia, Immunodeficiency, and Myelokathexis (WHIM) syndrome, an immunodeficiency associated with CXCR4 aberrant expression and activity and with chemotherapy resistance in clinical trials. In this review, we summarize the current knowledge on the relevance of CXCR4 mutations in cancer biology, focusing on its importance as predictors of clinical presentation and response to therapy.
Collapse
|
10
|
Yue GGL, Li L, Lee JKM, Kwok HF, Wong ECW, Li M, Fung KP, Yu J, Chan AWH, Chiu PWY, Lau CBS. Multiple modulatory activities of Andrographis paniculata on immune responses and xenograft growth in esophageal cancer preclinical models. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 60:152886. [PMID: 30910259 DOI: 10.1016/j.phymed.2019.152886] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 02/20/2019] [Accepted: 03/09/2019] [Indexed: 05/25/2023]
Abstract
BACKGROUND Esophageal cancer (EC) is a malignant gastrointestinal cancer with high morbidity worldwide and is the fourth leading cause of cancer-related deaths in China. Even though surgery and/or chemotherapy/chemoradiation might achieve good therapeutic response, recurrence rate is high due to cancer metastasis. Hence, the use of alternative adjuvant treatments, such as herbal medicines, for metastatic EC remains a great desire of the patients. Our previous studies have demonstrated the anti-metastatic efficacy of hot water extract of Andrographis paniculata (APW) in human esophageal cancer cells and tumor-bearing nude mice. PURPOSE In the present study, the immunomodulatory activities of APW were further evaluated in human peripheral blood mononuclear cells (PBMCs) and in a carcinogen-induced esophageal tumorigenesis model using immune-competent C57BL/6 mice. Besides, the inhibitory effects of APW on esophageal cancer cell line-based xenografts and patient-derived xenografts (PDX) were examined so as to illustrate the potential multi-targeted efficacies of APW in esophageal cancer in pre-clinical models. RESULTS In vitro results showed that APW could stimulate proliferation of PBMCs, as well as TNF-α and IFN-γproductions. In mice with 4-nitroquinoline 1-oxide-induced tumorigenesis, 21-day oral treatment with APW (1600 mg/kg) decreased the level of dysplasia in esophagus and significantly modulated the population of regulatory T cells. The cytokines productions by spleen lymphocytes of APW-treated mice were shifted towards normal resting state (i.e. unchallenged with carcinogen). Furthermore, APW treatment suppressed the growth of cell line-based xenografts by significantly increasing apoptosis in tumors, without causing severe body weight loss as chemotherapeutics did. Most importantly, the inhibitory effects of APW treatment on esophageal patient-derived xenografts growth were demonstrated for the first time. Besides, several diterpenes were detected in the plasma after oral administration of APW in mice, suggesting that multi-components of APW were bioavailable and might have contributed towards the varied pharmacological activities demonstrated in our studies. CONCLUSION APW was shown to possess anti-tumor, anti-metastatic and immunomodulatory activities in esophageal cancer cell-based and animal models, including immunocompromised mice model and clinically relevant PDX model. Our findings illustrated the potential multi-targeted efficacies of APW in esophageal cancer management.
Collapse
Affiliation(s)
- Grace Gar-Lee Yue
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants (CUHK), The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Lin Li
- Department of Surgery, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Julia Kin-Ming Lee
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants (CUHK), The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Hin-Fai Kwok
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants (CUHK), The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Eric Chun-Wai Wong
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants (CUHK), The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Mingyue Li
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants (CUHK), The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong; School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Kwok-Pui Fung
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants (CUHK), The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong; School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Jun Yu
- Department of Medicine and Therapeutics and State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Anthony Wing-Hung Chan
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Philip Wai-Yan Chiu
- Department of Surgery, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong.
| | - Clara Bik-San Lau
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants (CUHK), The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong.
| |
Collapse
|
11
|
Zhou W, Guo S, Liu M, Burow ME, Wang G. Targeting CXCL12/CXCR4 Axis in Tumor Immunotherapy. Curr Med Chem 2019; 26:3026-3041. [PMID: 28875842 PMCID: PMC5949083 DOI: 10.2174/0929867324666170830111531] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 05/08/2017] [Accepted: 06/14/2017] [Indexed: 12/14/2022]
Abstract
Chemokines, which have chemotactic abilities, are comprised of a family of small cytokines with 8-10 kilodaltons. Chemokines work in immune cells by trafficking and regulating cell proliferation, migration, activation, differentiation, and homing. CXCR-4 is an alpha-chemokine receptor specific for stromal-derived-factor-1 (SDF-1, also known as CXCL12), which has been found to be expressed in more than 23 different types of cancers. Recently, the SDF-1/CXCR-4 signaling pathway has emerged as a potential therapeutic target for human tumor because of its critical role in tumor initiation and progression by activating multiple signaling pathways, such as ERK1/2, ras, p38 MAPK, PLC/ MAPK, and SAPK/ JNK, as well as regulating cancer stem cells. CXCL12/CXCR4 antagonists have been produced, which have shown encouraging results in anti-cancer activity. Here, we provide a brief overview of the CXCL12/CXCR4 axis as a molecular target for cancer treatment. We also review the potential utility of targeting CXCL12/CXCR4 axis in combination of immunotherapy and/or chemotherapy based on up-to-date literature and ongoing research progress.
Collapse
Affiliation(s)
- Weiqiang Zhou
- Key Laboratory of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No.146 North Huanghe St, Huanggu District, Shenyang, Liaoning Province 110034, P. R. China
| | - Shanchun Guo
- RCMI Cancer Research Center, Xavier University of Louisiana, New Orleans, LA 70125, USA
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA 70125, USA
| | - Mingli Liu
- Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Matthew E. Burow
- Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Guangdi Wang
- RCMI Cancer Research Center, Xavier University of Louisiana, New Orleans, LA 70125, USA
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA 70125, USA
| |
Collapse
|
12
|
Eckert F, Schilbach K, Klumpp L, Bardoscia L, Sezgin EC, Schwab M, Zips D, Huber SM. Potential Role of CXCR4 Targeting in the Context of Radiotherapy and Immunotherapy of Cancer. Front Immunol 2018; 9:3018. [PMID: 30622535 PMCID: PMC6308162 DOI: 10.3389/fimmu.2018.03018] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 12/06/2018] [Indexed: 12/28/2022] Open
Abstract
Cancer immunotherapy has been established as standard of care in different tumor entities. After the first reports on synergistic effects with radiotherapy and the induction of abscopal effects-tumor shrinkage outside the irradiated volume attributed to immunological effects of radiotherapy-several treatment combinations have been evaluated. Different immunotherapy strategies (e.g., immune checkpoint inhibition, vaccination, cytokine based therapies) have been combined with local tumor irradiation in preclinical models. Clinical trials are ongoing in different cancer entities with a broad range of immunotherapeutics and radiation schedules. SDF-1 (CXCL12)/CXCR4 signaling has been described to play a major role in tumor biology, especially in hypoxia adaptation, metastasis and migration. Local tumor irradiation is a known inducer of SDF-1 expression and release. CXCR4 also plays a major role in immunological processes. CXCR4 antagonists have been approved for the use of hematopoietic stem cell mobilization from the bone marrow. In addition, several groups reported an influence of the SDF-1/CXCR4 axis on intratumoral immune cell subsets and anti-tumor immune response. The aim of this review is to merge the knowledge on the role of SDF-1/CXCR4 in tumor biology, radiotherapy and immunotherapy of cancer and in combinatorial approaches.
Collapse
Affiliation(s)
- Franziska Eckert
- Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany
| | - Karin Schilbach
- Department of General Pediatrics/Pediatric Oncology, University Hospital Tuebingen, Tuebingen, Germany
| | - Lukas Klumpp
- Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany.,Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany
| | - Lilia Bardoscia
- Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany.,Department of Radiation Oncology, University of Brescia, Brescia, Italy
| | - Efe Cumhur Sezgin
- Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany.,Departments of Clinical Pharmacology, Pharmacy and Biochemistry, University Hospital and University Tuebingen, Tuebingen, Germany
| | - Daniel Zips
- Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany
| | - Stephan M Huber
- Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany
| |
Collapse
|
13
|
Zhao K, Wang Z, Hackert T, Pitzer C, Zöller M. Tspan8 and Tspan8/CD151 knockout mice unravel the contribution of tumor and host exosomes to tumor progression. J Exp Clin Cancer Res 2018; 37:312. [PMID: 30541597 PMCID: PMC6292129 DOI: 10.1186/s13046-018-0961-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 11/14/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The tetraspanins Tspan8 and CD151 promote metastasis, exosomes (Exo) being suggested to be important in the crosstalk between tumor and host. The contribution of Tspan8 and CD151 to host versus tumor-derived exosome (TEX) activities being not defined, we approached the questions using 3-methylcholanthrene-induced (MCA) tumors from wt, Tspan8ko, CD151ko and Tspan8/CD151 (db)ko mice, implanted into tetraspanin-competent and deficient hosts. METHODS Tumor growth and dissemination, hematopoiesis and angiogenesis were surveyed in wild type (wt), Tspan8ko, CD151ko and dbko mice bearing tetraspanin-competent and -deficient MCA tumors. In vitro studies using tumor cells, bone marrow cells (BMC) and endothelial cells (EC) elaborated the mechanism of serum (s)Exo- and TEX-induced target modulation. RESULTS Tumors grew in autochthonous and syngeneic hosts differing in Tspan8- and/or CD151-competence. However, Tspan8ko- and/or CD151ko-tumor cell dissemination and settlement in metastatic organs was significantly reduced in the autochthonous host, and less severely in the wt-host. Impaired wt-MCA tumor dissemination in the ko-host confirmed a contribution of host- and tumor-Tspan8/-CD151 to tumor cell dissemination, delivery of sExo and TEX being severely impaired by a Tspan8ko/CD151ko. Coculturing tumor cells, BMC and EC with sExo and TEX revealed minor defects in epithelial mesenchymal transition and apoptosis resistance of ko tumors. Strongly reduced migratory and invasive capacity of Tspan8ko/CD151ko-MCA relies on distorted associations with integrins and CAM and missing Tspan8/CD151-promoted recruitment of proteases. The defects, differing between Tspan8ko- and CD151ko-MCA, were rescued by wt-TEX and, less efficiently Tspan8ko- and CD151ko-TEX. Minor defects in hematopoietic progenitor maturation were based on the missing association of hematopoietic growth factors /- receptors with CD151 and, less pronounced, Tspan8. Rescue of impaired angiogenesis in ko mice by wt-sExo and promotion of angiogenesis by TEX depended on the association of Tspan8 and CD151 with GPCR and RTK in EC and tumor cells. CONCLUSIONS Tspan8-/CD151-TEX play central roles in tumor progression. Tspan8-/CD151-sExo and TEX contribute by stimulating angiogenesis. Tspan8 and CD151 fulfill these tasks by associating with function-relevant proteins, the additive impact of Tspan8 and CD151 relying on differences in preferred associations. The distinct Tspan8 and CD151 contributions suggest a blockade of TEX-Tspan8 and -CD151 promising for therapeutic intervention.
Collapse
Affiliation(s)
- Kun Zhao
- Pancreas Section, University Hospital of Surgery, Ruprecht-Karls-University, Heidelberg, Germany
| | - Zhe Wang
- Pancreas Section, University Hospital of Surgery, Ruprecht-Karls-University, Heidelberg, Germany
- Present Address: Department of Oncology, First Affiliated Hospital of Guangdong Pharmaceutical University, Guangdong, China
| | - Thilo Hackert
- Pancreas Section, University Hospital of Surgery, Ruprecht-Karls-University, Heidelberg, Germany
| | - Claudia Pitzer
- Interdisciplinary Neurobehavioral Core, Institute of Pharmacology, Ruprecht-Karls-University, Heidelberg, Germany
| | - Margot Zöller
- Pancreas Section, University Hospital of Surgery, Ruprecht-Karls-University, Heidelberg, Germany
| |
Collapse
|
14
|
Li W, Zhao W, Lu Z, Zhang W, Yang X. Long Noncoding RNA GAS5 Promotes Proliferation, Migration, and Invasion by Regulation of miR-301a in Esophageal Cancer. Oncol Res 2018; 26:1285-1294. [PMID: 29386089 PMCID: PMC7844703 DOI: 10.3727/096504018x15166193231711] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Long noncoding RNA (lncRNA) growth arrest-specific transcript 5 (GAS5) has been revealed to be associated with the progression of various cancers. However, the biological roles of GAS5 in esophageal cancer (EC) remain unclear. We aimed to thoroughly explore the functions of GAS5 in EC. The results showed that GAS5 expression was increased in EC cells (ECA109, TE-1, TE-3, and EC9706) compared to SHEE cells. Knockdown of GAS5 decreased cell viability, migration, and invasion and induced apoptosis in EC9706 cells. Moreover, miR-301a appeared to be directly sponged by GAS5, and miR-301a suppression obviously alleviated the protumor effects of GAS5. Furthermore, miR-301a positively regulated CXCR4 expression, and overexpression of CXCR4 induced apoptosis and abolished the promoting effect of miR-301a inhibition on cell viability, migration, and invasion. Besides, miR-301a blocked Wnt/β-catenin and NF-κB signaling pathways by regulation of CXCR4. Our results indicated that GAS5 promoted proliferation and metastasis and inhibited apoptosis by regulation of miR-301a in EC. These data contributed to our understanding of the mechanisms of miRNA-lncRNA interaction and provides a novel therapeutic strategy for EC.
Collapse
Affiliation(s)
- Wei Li
- Department of Gastroenterology, Shengli Oilfield Central Hospital, Dongying, P.R. China
| | - Weidong Zhao
- Department of Gastroenterology, Shengli Oilfield Central Hospital, Dongying, P.R. China
| | - Zhaohui Lu
- Department of Gastroenterology, Shengli Oilfield Central Hospital, Dongying, P.R. China
| | - Wen Zhang
- Department of Gastroenterology, Shengli Oilfield Central Hospital, Dongying, P.R. China
| | - Xuan Yang
- Department of Gastroenterology, Shengli Oilfield Central Hospital, Dongying, P.R. China
| |
Collapse
|
15
|
Drenckhan A, Freytag M, Supuran CT, Sauter G, Izbicki JR, Gros SJ. CAIX furthers tumour progression in the hypoxic tumour microenvironment of esophageal carcinoma and is a possible therapeutic target. J Enzyme Inhib Med Chem 2018; 33:1024-1033. [PMID: 29865880 PMCID: PMC6010094 DOI: 10.1080/14756366.2018.1475369] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The hypoxic tumour microenvironment of solid tumours represents an important starting point for modulating progression and metastatic spread. Carbonic anhydrase IX (CAIX) is a known HIF-1α-dependent key player in maintaining cell pH conditions under hypoxia. We show that CAIX is strongly expressed in esophageal carcinoma tissues. We hypothesize that a moderate CAIX expression facilitates metastases and thereby worsens prognosis. Selective inhibition of CAIX by specific CAIX inhibitors and a CAIX knockdown effectively inhibit proliferation and migration in vitro. In the orthotopic esophageal carcinoma model, the humanized HER2 antibody trastuzumab down-regulates CAIX, possibly through CAIX’s linkage with HER2 in the hypoxic microenvironment. Our results show CAIX to be an essential part of the tumour microenvironment and a possible master regulator of tumour progression. This makes CAIX a highly effective and feasible therapeutic target for selective cancer treatment.
Collapse
Affiliation(s)
- Astrid Drenckhan
- a Department of General, Visceral and Thoracic Surgery , University Medical Center Hamburg-Eppendorf , Hamburg , Germany
| | - Morton Freytag
- a Department of General, Visceral and Thoracic Surgery , University Medical Center Hamburg-Eppendorf , Hamburg , Germany
| | - Claudiu T Supuran
- b Department Neurofarba , Section of Pharmaceutical Sciences, University of Florence , Florence , Italy
| | - Guido Sauter
- c Department of Pathology , University Medical Center Hamburg-Eppendorf , Hamburg , Germany
| | - Jakob R Izbicki
- a Department of General, Visceral and Thoracic Surgery , University Medical Center Hamburg-Eppendorf , Hamburg , Germany
| | - Stephanie J Gros
- a Department of General, Visceral and Thoracic Surgery , University Medical Center Hamburg-Eppendorf , Hamburg , Germany.,d Department of Pediatric Surgery , Ûniversity Children's Hospital Basel , Basel , Switzerland
| |
Collapse
|
16
|
Lecavalier-Barsoum M, Chaudary N, Han K, Koritzinsky M, Hill R, Milosevic M. Targeting the CXCL12/CXCR4 pathway and myeloid cells to improve radiation treatment of locally advanced cervical cancer. Int J Cancer 2018; 143:1017-1028. [PMID: 29417588 DOI: 10.1002/ijc.31297] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 01/10/2018] [Accepted: 02/01/2018] [Indexed: 12/17/2022]
Abstract
Cervical cancer is the fourth most commonly diagnosed cancer and the fourth leading cause of cancer death in women worldwide. Approximately half of cervical cancer patients present with locally advanced disease, for which surgery is not an option. These cases are nonetheless potentially curable with radiotherapy and cisplatin chemotherapy. Unfortunately, some tumours are resistant to treatment, and lymph node and distant recurrences are major problems in patients with advanced disease at diagnosis. New targeted treatments that can overcome treatment resistance and reduce metastases are urgently needed. The CXCL12/CXCR4 chemokine pathway is ubiquitously expressed in many normal tissues and cancers, including cervical cancer. Emerging evidence indicates that it plays a central role in cervical cancer pathogenesis, malignant progression, the development of metastases and radiation treatment response. Pre-clinical studies of standard-of-care fractionated radiotherapy and concurrent weekly cisplatin plus the CXCR4 inhibitor Plerixafor (AMD3100) in patient-derived orthotopic cervical cancer xenografts have shown improved primary tumour response and reduced lymph node metastases with no increase in early or late side effects. These studies have pointed the way forward to future clinical trials of radiotherapy/cisplatin plus Plerixafor or other newly emerging CXCL12 or CXCR4 inhibitors in women with cervical cancer.
Collapse
Affiliation(s)
- Magali Lecavalier-Barsoum
- Department of Oncology, Segal Cancer Centre, Jewish General Hospital, McGill University, Montréal, Canada
| | - Naz Chaudary
- Princess Margaret Cancer Centre and Campbell Family Institute for Cancer Research, University Health Network, Toronto, Canada
| | - Kathy Han
- Radiation Medicine Program, University Health Network and Princess Margaret Cancer Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Marianne Koritzinsky
- Radiation Medicine Program, University Health Network and Princess Margaret Cancer Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Richard Hill
- Princess Margaret Cancer Centre and Campbell Family Institute for Cancer Research, University Health Network, Toronto, Canada.,Radiation Medicine Program, University Health Network and Princess Margaret Cancer Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Michael Milosevic
- Radiation Medicine Program, University Health Network and Princess Margaret Cancer Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Canada
| |
Collapse
|
17
|
MicroRNA-126 inhibits colon cancer cell proliferation and invasion by targeting the chemokine (C-X-C motif) receptor 4 and Ras homolog gene family, member A, signaling pathway. Oncotarget 2018; 7:60230-60244. [PMID: 27517626 PMCID: PMC5312381 DOI: 10.18632/oncotarget.11176] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 07/26/2016] [Indexed: 12/21/2022] Open
Abstract
MicroRNA-126 (miR-126) suppresses the migration, proliferation and invasion of colon cancer cells. However, the underlying mechanisms of miR-126 in colon cancer have not been fully elucidated. In this study, in vivo experiments revealed that miR-126 inhibits colon cancer growth and metastasis. Furthermore, miR-126 was down-regulated in human colon cancer tissue, and its expression was inversely correlated with TNM stage and metastasis of patients. Low level of miR-126 identified patients with poor prognosis. And we found that miR-126 expression was negatively correlated with the expression levels of chemokine (C-X-C motif) receptor 4 (CXCR4) and components of signaling pathway of Ras homolog gene family, member A (RhoA) in vitro and in vivo. Moreover, we verified that miR-126 negatively regulated CXCR4 and RhoA signaling in vitro. In addition, either in miR-126-overexpressing or in miR- 126-silenced colon cancer cells, the restoration of CXCR4 could significantly reverse the proliferation and invasion, as well as abolish the effects of miR-126 on RhoA signaling pathway. Collectively, these results demonstrated that miR-126 acts as a tumor suppressor by inactivating RhoA signaling via CXCR4 in colon cancer. And miR-126 may serve as a prognostic marker for monitoring and treating colon cancer.
Collapse
|
18
|
Fang HY, Münch NS, Schottelius M, Ingermann J, Liu H, Schauer M, Stangl S, Multhoff G, Steiger K, Gerngroß C, Jesinghaus M, Weichert W, Kühl AA, Sepulveda AR, Wester HJ, Wang TC, Quante M. CXCR4 Is a Potential Target for Diagnostic PET/CT Imaging in Barrett's Dysplasia and Esophageal Adenocarcinoma. Clin Cancer Res 2017; 24:1048-1061. [PMID: 29208671 DOI: 10.1158/1078-0432.ccr-17-1756] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Revised: 10/17/2017] [Accepted: 11/30/2017] [Indexed: 12/30/2022]
Abstract
Purpose: Barrett's esophagus represents an early stage in carcinogenesis leading to esophageal adenocarcinoma. Considerable evidence supports a major role for chronic inflammation and diverse chemokine pathways in the development of Barrett's esophagus and esophageal adenocarcinoma.Experimental Design: Here we utilized an IL1β transgenic mouse model of Barrett's esophagus and esophageal adenocarcinoma and human patient imaging to analyze the importance of CXCR4-expressing cells during esophageal carcinogenesis.Results: IL1β overexpression induces chronic esophageal inflammation and recapitulates the progression to Barrett's esophagus and esophageal adenocarcinoma. CXCR4 expression is increased in both epithelial and immune cells during disease progression in pL2-IL1β mice and also elevated in esophageal adenocarcinoma patient biopsy samples. Specific recruitment of CXCR4-positive (CXCR4+) immune cells correlated with dysplasia progression, suggesting that this immune population may be a key contributor to esophageal carcinogenesis. Similarly, with progression to dysplasia, there were increased numbers of CXCR4+ columnar epithelial cells at the squamocolumnar junction (SCJ). These findings were supported by stronger CXCR4-related signal intensity in ex vivo fluorescence imaging and autoradiography with advanced dysplasia. Pilot CXCR4-directed PET/CT imaging studies in patients with esophageal cancer demonstrate the potential utility of CXCR4 imaging for the diagnosis and staging of esophageal cancer.Conclusion: In conclusion, the recruitment of CXCR4+ immune cells and expansion of CXCR4+ epithelial cells in esophageal dysplasia and cancer highlight the potential of CXCR4 as a biomarker and molecular target for diagnostic imaging of the tumor microenvironment in esophageal adenocarcinoma. Clin Cancer Res; 24(5); 1048-61. ©2017 AACR.
Collapse
Affiliation(s)
- Hsin-Yu Fang
- II. Medizinische Klinik, Technische Universitat München, Munich, Germany
| | | | - Margret Schottelius
- Pharmazeutische Radiochemie, Technische Universitat München, Munich, Germany
| | - Jonas Ingermann
- II. Medizinische Klinik, Technische Universitat München, Munich, Germany
| | - Haibo Liu
- Division of Digestive and Liver Diseases, Columbia University Medical Center, New York, New York
| | - Michael Schauer
- II. Medizinische Klinik, Technische Universitat München, Munich, Germany
| | - Stefan Stangl
- Klinik für RadioOnkologie und Strahlentherapie, Technische Universitat München, Munich, Germany
| | - Gabriele Multhoff
- Klinik für RadioOnkologie und Strahlentherapie, Technische Universitat München, Munich, Germany
| | - Katja Steiger
- Institut für Pathologie, Technische Universitat München, Munich, Germany
| | - Carlos Gerngroß
- Nuklearmedizinische Klinik und Poliklinik, Technische Universitat München, Munich, Germany
| | - Moritz Jesinghaus
- Institut für Pathologie, Technische Universitat München, Munich, Germany
| | - Wilko Weichert
- Institut für Pathologie, Technische Universitat München, Munich, Germany
| | - Anja A Kühl
- iPATH.Berlin/Medizinische Klinik für Gastroenterologie, Infektiologie und Rheumatologie/Research Center ImmunoSciences, Charité-Campus Benjamin Franklin, Berlin, Germany
| | - Antonia R Sepulveda
- Division of Gastrointestinal Pathology, Columbia University Medical Center, New York, New York
| | - Hans-Jürgen Wester
- Pharmazeutische Radiochemie, Technische Universitat München, Munich, Germany
| | - Timothy C Wang
- Division of Digestive and Liver Diseases, Columbia University Medical Center, New York, New York
| | - Michael Quante
- II. Medizinische Klinik, Technische Universitat München, Munich, Germany.
| |
Collapse
|
19
|
Melsens E, De Vlieghere E, Descamps B, Vanhove C, De Wever O, Ceelen W, Pattyn P. Improved xenograft efficiency of esophageal adenocarcinoma cell lines through in vivo selection. Oncol Rep 2017; 38:71-81. [PMID: 28504813 PMCID: PMC5492849 DOI: 10.3892/or.2017.5640] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 11/18/2016] [Indexed: 01/29/2023] Open
Abstract
The present study aimed to investigate the orthotopic growth potential of two generally available esophageal adenocarcinoma cell lines, OE33 and OACM5 1.C, and a third in vivo selected subpopulation, OACM5 1.C SC1. One group of mice was subcutaneously injected in the hind legs. Tumor growth was measured with calipers. Another group was injected orthotopically in the distal esophageal wall through median laparotomy. Tumor development was evaluated macroscopically and confirmed microscopically. A subset of mice was evaluated with magnetic resonance imaging (MRI) to follow tumor progression. Additionally, functional cell line characteristics were evaluated in vitro (clonogenic, collagen invasion and sphere formation assays, and protein analysis of cell-cell adhesion and cytoskeletal proteins) to better understand xenograft behavior. OE33 cells were shown to be epithelial-like, whereas OACM5 1.C and OACM5 1.C SC1 were more mesenchymal-like. The three cell lines were non-invasive into native type I collagen gels. In vivo, OE33 cells led to 63.6 and 100% tumor nodules after orthotopic (n=12) and subcutaneous (n=8) injection, respectively. Adversely, OACM5 1.C cells did not lead to tumor formation after orthotopic injection (n=6) and only 50% of subcutaneous injections led to tumor nodules (n=8). However, the newly established cell line OACM5 1.C SC1 resulted in 33% tumor formation when orthotopically injected (n=6) and in 100% tumors when injected subcutaneously (n=8). The higher xenograft rate of OACM5 1.C SC1 (P<0.05) corresponded with a higher clonogenic potential compared to its parental cell line (P<0.0001). All models showed local tumor growth without metastasis formation. In conclusion, OACM5 1.C has a poor tumor take rate at an orthotopic and ectopic site. A subpopulation obtained through in vivo selection, OACM5 1.C SC1, gives a significant higher take rate, ectopically. Furthermore, OE33 establishes orthotopic (and subcutaneous) xenografts in mice. These models can be of interest for future studies, and their slow growth rates are a challenge for therapeutic intervention.
Collapse
Affiliation(s)
- Elodie Melsens
- Laboratory of Experimental Surgery, Department of Surgery, Ghent University Hospital, Ghent, Belgium
| | - Elly De Vlieghere
- Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Ghent, Belgium
| | - Benedicte Descamps
- Infinity (iMinds-IBiTech-MEDISIP), Department of Electronics and Information Systems, Ghent University, Ghent, Belgium
| | - Christian Vanhove
- Infinity (iMinds-IBiTech-MEDISIP), Department of Electronics and Information Systems, Ghent University, Ghent, Belgium
| | - Olivier De Wever
- Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Ghent, Belgium
| | - Wim Ceelen
- Laboratory of Experimental Surgery, Department of Surgery, Ghent University Hospital, Ghent, Belgium
| | - Piet Pattyn
- Laboratory of Experimental Surgery, Department of Surgery, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
20
|
The adjuvant value of Andrographis paniculata in metastatic esophageal cancer treatment - from preclinical perspectives. Sci Rep 2017; 7:854. [PMID: 28405006 PMCID: PMC5429803 DOI: 10.1038/s41598-017-00934-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 03/17/2017] [Indexed: 01/27/2023] Open
Abstract
Esophageal cancer (EC) is the fourth and sixth leading cause of cancer-related deaths in China and United States, respectively. The dismal prognosis of EC is mainly attributed to distant metastases, which may not be overcome by chemotherapy alone. Hence, the use of alternative adjuvant treatments, such as herbal medicines, for metastatic EC remains a great desire of patients. Our previous study demonstrated the in vivo anti-tumor and in vitro anti-invasion activities of Andrographis paniculata (AP) in esophageal cancer. In the present study, the chemical constituents of absorbed AP components through human intestinal Caco-2 cell monolayer were verified for the first time. The anti-migratory activities and suppressive effects on metastasis-related factors such as HER2, MMP2, MMP9, TM4SF3, CXCR4 of the absorbed AP components were revealed in esophageal cancer cells EC-109. The anti-tumor and anti-metastatic effects of AP water extract (1600 mg/kg) were further confirmed in metastatic esophageal xenograft-bearing mice. Besides, AP water extract acted synergistically with cisplatin plus 5-fluorouracil on inhibiting tumor nodule growth (with combination index <0.7). Meanwhile, chemotherapeutics-induced side-effects could also be reduced by AP water extract. The present findings provide evidence on safety and advantages of the combined use of AP with chemotherapeutics in pre-clinical setting.
Collapse
|
21
|
Duan Y, Zhang S, Wang L, Zhou X, He Q, Liu S, Yue K, Wang X. Targeted silencing of CXCR4 inhibits epithelial-mesenchymal transition in oral squamous cell carcinoma. Oncol Lett 2016; 12:2055-2061. [PMID: 27602138 DOI: 10.3892/ol.2016.4838] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 06/07/2016] [Indexed: 12/26/2022] Open
Abstract
Aberrant overexpression of C-X-C chemokine receptor type 4 (CXCR4) is a critical event during tumor metastasis. It has been previously reported that the expression of CXCR4 is linked with epithelial-mesenchymal transition (EMT) in oral squamous cell carcinoma (OSCC) tissues derived from patients. The present study addresses the role of CXCR4 in EMT in tongue squamous cell carcinoma (TSCCA) cells in vitro and in xenograft models. Small interfering (si) RNA sequences targeting the CXCR4 gene were transfected into TSCCA cells. Cell migration, invasion, apoptosis and EMT markers were determined in TSCCA cells using wound healing and Transwell assays, Annexin V/propdidum iodide double staining and western blot analysis, respectively. In vivo, tumor growth was assessed by subcutaneous inoculation of cells into BALB/c nude mice. Phenotypic EMT markers and regulatory factors were detected in the tumor tissues derived from the mice. In vitro, silencing of CXCR4 expression suppressed cell migration and invasion, and induced apoptosis. The protein expression of the EMT-associated markers N-cadherin and matrix metalloproteinases 2/9 were attenuated, while E-cadherin was increased. In vivo, CXCR4 siRNA inhibited tumor growth, and EMT-associated proteins had similar expression patterns to the experimental results observed in vitro. In conclusion, the present study demonstrated that CXCR4 silencing suppressed EMT in OSCC, thus affecting tumor metastasis.
Collapse
Affiliation(s)
- Yuansheng Duan
- Department of Maxillofacial and E.N.T Oncology, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, P.R. China; National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| | - Shu Zhang
- Department of Maxillofacial and E.N.T Oncology, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, P.R. China; National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| | - Longlong Wang
- Department of Thyroid Surgery, Baotou Cancer Hospital, Baotou, Inner Mongolia 014030, P.R. China
| | - Xuan Zhou
- Department of Maxillofacial and E.N.T Oncology, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, P.R. China; National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| | - Qinghua He
- Department of Maxillofacial and E.N.T Oncology, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, P.R. China; National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| | - Su Liu
- Department of Maxillofacial and E.N.T Oncology, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, P.R. China; National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| | - Kai Yue
- Department of Maxillofacial and E.N.T Oncology, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, P.R. China; National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| | - Xudong Wang
- Department of Maxillofacial and E.N.T Oncology, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, P.R. China; National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| |
Collapse
|
22
|
Kaifi JT, Li G, Clawson G, Kimchi ET, Staveley-O'Carroll KF. Perioperative circulating tumor cell detection: Current perspectives. Cancer Biol Ther 2016; 17:859-69. [PMID: 27045201 DOI: 10.1080/15384047.2016.1167296] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Primary cancer resections and in selected cases surgical metastasectomies significantly improve survival, however many patients develop recurrences. Circulating tumor cells (CTCs) function as an independent marker that could be used in the prognostication of different cancers. Sampling of blood and bone marrow compartments during cancer resections is a unique opportunity to increase individual tumor cell capture efficiency. This review will address the diagnostic and therapeutic potentials of perioperative tumor isolation and highlight the focus of future studies on characterization of single disseminated cancer cells to identify targets for molecular therapy and immune escape mechanisms.
Collapse
Affiliation(s)
- Jussuf T Kaifi
- a Hugh E. Stephenson Jr., M.D. , Department of Surgery , University of Missouri , Columbia , MO , USA.,b Ellis Fischel Cancer Center , University of Missouri , Columbia , MO , USA
| | - Guangfu Li
- a Hugh E. Stephenson Jr., M.D. , Department of Surgery , University of Missouri , Columbia , MO , USA.,c Department of Molecular Microbiology and Immunology , University of Missouri , Columbia , MO , USA
| | - Gary Clawson
- d Gittlen Cancer Research Foundation and Department of Pathology , Materials Research Institute, Penn State College of Medicine, Pennsylvania State University , Hershey , PA , USA
| | - Eric T Kimchi
- a Hugh E. Stephenson Jr., M.D. , Department of Surgery , University of Missouri , Columbia , MO , USA.,b Ellis Fischel Cancer Center , University of Missouri , Columbia , MO , USA.,c Department of Molecular Microbiology and Immunology , University of Missouri , Columbia , MO , USA
| | - Kevin F Staveley-O'Carroll
- a Hugh E. Stephenson Jr., M.D. , Department of Surgery , University of Missouri , Columbia , MO , USA.,b Ellis Fischel Cancer Center , University of Missouri , Columbia , MO , USA.,c Department of Molecular Microbiology and Immunology , University of Missouri , Columbia , MO , USA
| |
Collapse
|
23
|
Gravina GL, Mancini A, Muzi P, Ventura L, Biordi L, Ricevuto E, Pompili S, Mattei C, Di Cesare E, Jannini EA, Festuccia C. CXCR4 pharmacogical inhibition reduces bone and soft tissue metastatic burden by affecting tumor growth and tumorigenic potential in prostate cancer preclinical models. Prostate 2015; 75:1227-46. [PMID: 26073897 DOI: 10.1002/pros.23007] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 03/25/2015] [Indexed: 12/22/2022]
Abstract
BACKGROUND The majority of prostate cancer (Pca) patient morbidity can be attributed to bone metastatic events, which poses a significant clinical obstacle. Therefore, a better understanding of this phenomenon is imperative and might help to develop novel therapeutic strategies. Stromal cell-derived factor 1α (SDF-1α) and its receptor CXCR4 have been implicated as regulators of bone resorption and bone metastatic development, suggesting that agents able to suppress this signaling pathway may be used as pharmacological treatments. In this study we studied if two CXCR4 receptor antagonists, Plerixafor and CTE9908, may affect bone metastatic disease induced by Pca in preclinical experimental models METHODS To verify the hypothesis that CXCR4 inhibition affects Pca metastatic disease, selective CXCR4 compounds, Plerixafor, and CTE9908, were tested in preclinical models known to generate bone lesions. Additionally, the expression levels of CXCR4 and SDF-1α were analyzed in a number of human tissues derived from primary tumors, lymph-nodes and osseous metastases of Pca as well as in a wide panel of human Pca cell lines to non-tumorigenic and tumorigenic phenotype. RESULTS Bone-derived Pca cells express higher CXCR4 levels than other Pca cell lines. This differential expression was also observed in human Pca samples. In vitro evidence supports the hypothesis that factors produced by bone microenvironment differentially sustain CXCR4 and SDF1-α expression with respect to prostate microenvironment determining increased efficacy toward Plerixafor. The use of SDF1-α neutralizing antibodies greatly reduced the increase of CXCR4 expression in cells co-cultured with bone stromal cells (BMSc) and to a lesser extent in cells co-cultured with prostate stromal cells (HPSc) and partially reduced SDF1-α Plerixafor efficacy. SDF-1α induced tumor cell migration and invasion, as well as MMP-9, MMP-2, and uPA expression, which were reduced by Plerixafor. The incidence of X-ray detectable bone lesions was significantly reduced following Plerixafor and CTE9908 treatment Kaplan-Meier probability plots showed a significant improvement in the overall survival of mice treated with Plerixafor and CTE9908. The reduced intra-osseous growth of PC3 and PCb2 tumor cells after intratibial injection, as a result of Plerixafor and CTE9908 treatment, correlated with decreased osteolysis and serum levels of both mTRAP and type I collagen fragments (CTX), which were significantly lower with respect to controls. CONCLUSIONS Our report provides novel information on the potential activity of CXCR4 inhibitors on the formation and progression of Pca bone and soft tissue metastases and supports a biological rationale for the use of these inhibitors in men at high risk to develop clinically evident bone lesions.
Collapse
Affiliation(s)
- Giovanni Luca Gravina
- Division of Radiation Oncology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy
| | - Andrea Mancini
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy
| | - Paola Muzi
- Department of Life, Health and Environmental Sciences, L'Aquila, Italy
| | - Luca Ventura
- Pathology Department, San Salvatore Hospital L'Aquila, L'Aquila, Italy
| | - Leda Biordi
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Molecular Pathology, University of L'Aquila, L'Aquila, Italy
| | - Enrico Ricevuto
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Molecular Pathology, University of L'Aquila, L'Aquila, Italy
| | - Simona Pompili
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Human Anatomy, L'Aquila, Italy
| | - Claudia Mattei
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy
| | - Ernesto Di Cesare
- Division of Radiation Oncology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy
| | | | - Claudio Festuccia
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
24
|
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: 0.7] [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
|
25
|
Liao YX, Fu ZZ, Zhou CH, Shan LC, Wang ZY, Yin F, Zheng LP, Hua YQ, Cai ZD. AMD3100 reduces CXCR4-mediated survival and metastasis of osteosarcoma by inhibiting JNK and Akt, but not p38 or Erk1/2, pathways in in vitro and mouse experiments. Oncol Rep 2015; 34:33-42. [PMID: 25997540 PMCID: PMC4484610 DOI: 10.3892/or.2015.3992] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 04/29/2015] [Indexed: 12/27/2022] Open
Abstract
Osteosarcoma (OS) has an unfavorable prognosis and tends to metastasize to lung tissue. Although the CXCL12-CXCR4 axis appears to affect progression and metastasis in numerous tumors, its mechanism and downstream pathways in OS remain unclear. We used western blotting and flow cytometry to detect CXCR4 and CXCR7 expression in two OS cell lines (LM8 and Dunn). An MTT assay was used to evaluate the effects of CXCL12 and AMD3100, a specific CXCR4 antagonist, on cell viability. Flow cytometry was utilized to analyze changes in apoptosis induced by serum deprivation following treatment with CXCL12 and AMD3100. A Transwell assay was used to assess cell migration in response to CXCL12 and AMD3100. Western blotting was performed to identify the phosphorylation of signaling molecules (JNK, c-Jun, Akt, p38 and Erk1/2) and expression of caspase-3 and -8, and PARP. Mouse models were employed to evaluate AMD3100 inhibition of primary OS growth and lung metastasis in vivo. CXCR4 expression was detected in LM8 but not Dunn cells, and neither cell line expressed CXCR7. The addition of CXCL12 induced the survival and migration of serum-starved CXCR4+ LM8 cells activating JNK and Akt pathways, which were abrogated by adding AMD3100. However, similar results were not observed in CXCR4− Dunn cells. CXCL12 protected LM8, but not Dunn cells, from apoptosis induced by serum deprivation by suppressing PARP cleavage, which was partly reversed by AMD3100. In a mouse model, AMD3100 reduced primary tumor growth and lung metastasis compared with the controls. Thus, the CXCL12-CXCR4 axis regulated OS survival and metastasis through the JNK and Akt pathways, and blocking them with AMD3100 was found to be a potential OS treatment.
Collapse
Affiliation(s)
- Yu-Xin Liao
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Ze-Ze Fu
- Department of Orthopaedics, Shanghai First People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, P.R. China
| | - Cheng-Hao Zhou
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Lian-Cheng Shan
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Zhuo-Ying Wang
- Department of Orthopaedics, Shanghai First People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, P.R. China
| | - Fei Yin
- Department of Orthopaedics, Shanghai First People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, P.R. China
| | - Long-Po Zheng
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Ying-Qi Hua
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Zheng-Dong Cai
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| |
Collapse
|
26
|
Zhou XM, He L, Hou G, Jiang B, Wang YH, Zhao L. Clinicopathological significance of CXCR4 in non-small cell lung cancer. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:1349-58. [PMID: 25834393 PMCID: PMC4357617 DOI: 10.2147/dddt.s71060] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background Emerging evidence indicates 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 unclear. Here, we conducted a meta-analysis to quantitatively evaluate the association of CXCR4 expression with the incidence of NSCLC and clinicopatho-logical characteristics. Methods A detailed literature search was made from Medline and Web of Science for related research publications written in English and Chinese. The methodological quality of the studies was also evaluated. Analyses of pooled data were performed. Odds ratio (OR) and hazard ratio (HR) were calculated and summarized. Results The final analysis of 1,446 NSCLC patients from 13 eligible studies was performed. We observed that CXCR4 expression was significantly higher in NSCLC than in normal lung tissue from the pooled OR from five studies including 380 NSCLC and 118 normal lung tissue (OR=12.86, 95% confidence interval =3.63–45.59, P<0.0001). CXCR4 expression was not associated with smoking status and type of pathology. However, CXCR4 expression was significantly associated with clinical stages, metastatic status, and overall survival in NSCLC patients. Conclusion The results of this meta-analysis suggest that CXCR4 expression is associated with an increased risk and worse survival in NSCLC patients. The aberrant CXCR4 expression plays an important role in the carcinogenesis and metastasis of NSCLC.
Collapse
Affiliation(s)
- Xiao-Ming Zhou
- Department of Respiratory Medicine, The Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Lan He
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong SAR, People's Republic of China
| | - Gang Hou
- Department of Respiratory Medicine, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Bing Jiang
- Department of Ultrasonography, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Yuan-He Wang
- Department of Medical Oncology, Liaoning Cancer Hospital, Shenyang, People's Republic of China
| | - Li Zhao
- Department of Respiratory Medicine, The Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| |
Collapse
|
27
|
Trautmann F, Cojoc M, Kurth I, Melin N, Bouchez LC, Dubrovska A, Peitzsch C. CXCR4 as biomarker for radioresistant cancer stem cells. Int J Radiat Biol 2014; 90:687-99. [PMID: 24650104 DOI: 10.3109/09553002.2014.906766] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE Radioresistance of cancer cells remains a fundamental barrier for maximum efficient radiotherapy. Tumor heterogeneity and the existence of distinct cell subpopulations exhibiting different genotypes and biological behaviors raise difficulties to eradicate all tumorigenic cells. Recent evidence indicates that a distinct population of tumor cells, called cancer stem cells (CSC), is involved in tumor initiation and recurrence and is a putative cause of tumor radioresistance. There is an urgent need to identify the intrinsic molecular mechanisms regulating the generation and maintenance of resistance to radiotherapy, especially within the CSC subset. The chemokine C-X-C motif receptor 4 (CXCR4) has been found to be a prognostic marker in various types of cancer, being involved in chemotaxis, stemness and drug resistance. The interaction of CXCR4 with its ligand, the chemokine C-X-C motif ligand 12 (CXCL12), plays an important role in modulating the tumor microenvironment, angiogenesis and CSC niche. Moreover, the therapeutic inhibition of the CXCR4/CXCL12 signaling pathway is sensitizing the malignant cells to conventional anti-cancer therapy. CONTENT Within this review we are summarizing the role of the CXCR4/CXCL12 axis in the modulation of CSC properties, the regulation of the tumor microenvironment in response to irradiation, therapy resistance and tumor relapse. CONCLUSION In light of recent findings, the inhibition of the CXCR4/CXCL12 signaling pathway is a promising therapeutic option to refine radiotherapy.
Collapse
Affiliation(s)
- Franziska Trautmann
- OncoRay - National Center for Radiation Research in Oncology, Medizinische Fakultät Carl Gustav Carus der Technischen Universität and Helmholtz Zentrum Rossendorf , Dresden
| | | | | | | | | | | | | |
Collapse
|
28
|
Zhou J, Xiang Y, Yoshimura T, Chen K, Gong W, Huang J, Zhou Y, Yao X, Bian X, Wang JM. The role of chemoattractant receptors in shaping the tumor microenvironment. BIOMED RESEARCH INTERNATIONAL 2014; 2014:751392. [PMID: 25110692 PMCID: PMC4119707 DOI: 10.1155/2014/751392] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 06/17/2014] [Indexed: 12/13/2022]
Abstract
Chemoattractant receptors are a family of seven transmembrane G protein coupled receptors (GPCRs) initially found to mediate the chemotaxis and activation of immune cells. During the past decades, the functions of these GPCRs have been discovered to not only regulate leukocyte trafficking and promote immune responses, but also play important roles in homeostasis, development, angiogenesis, and tumor progression. Accumulating evidence indicates that chemoattractant GPCRs and their ligands promote the progression of malignant tumors based on their capacity to orchestrate the infiltration of the tumor microenvironment by immune cells, endothelial cells, fibroblasts, and mesenchymal cells. This facilitates the interaction of tumor cells with host cells, tumor cells with tumor cells, and host cells with host cells to provide a basis for the expansion of established tumors and development of distant metastasis. In addition, many malignant tumors of the nonhematopoietic origin express multiple chemoattractant GPCRs that increase the invasiveness and metastasis of tumor cells. Therefore, GPCRs and their ligands constitute targets for the development of novel antitumor therapeutics.
Collapse
Affiliation(s)
- Jiamin Zhou
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
- Endoscopic Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yi Xiang
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
- Department of Pulmonary Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Teizo Yoshimura
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Keqiang Chen
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Wanghua Gong
- Basic Research Program, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Jian Huang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Ye Zhou
- Department of Gastric Cancer and Soft Tissue Surgery, Fudan University Cancer Center, Shanghai 200032, China
| | - Xiaohong Yao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Xiuwu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Ji Ming Wang
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| |
Collapse
|
29
|
Papagerakis S, Pannone G, Zheng L, About I, Taqi N, Nguyen NPT, Matossian M, McAlpin B, Santoro A, McHugh J, Prince ME, Papagerakis P. Oral epithelial stem cells - implications in normal development and cancer metastasis. Exp Cell Res 2014; 325:111-29. [PMID: 24803391 DOI: 10.1016/j.yexcr.2014.04.021] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 04/25/2014] [Accepted: 04/28/2014] [Indexed: 12/18/2022]
Abstract
Oral mucosa is continuously exposed to environmental forces and has to be constantly renewed. Accordingly, the oral mucosa epithelium contains a large reservoir of epithelial stem cells necessary for tissue homeostasis. Despite considerable scientific advances in stem cell behavior in a number of tissues, fewer studies have been devoted to the stem cells in the oral epithelium. Most of oral mucosa stem cells studies are focused on identifying cancer stem cells (CSC) in oral squamous cell carcinomas (OSCCs) among other head and neck cancers. OSCCs are the most prevalent epithelial tumors of the head and neck region, marked by their aggressiveness and invasiveness. Due to their highly tumorigenic properties, it has been suggested that CSC may be the critical population of cancer cells in the development of OSCC metastasis. This review presents a brief overview of epithelium stem cells with implications in oral health, and the clinical implications of the CSC concept in OSCC metastatic dissemination.
Collapse
Affiliation(s)
- Silvana Papagerakis
- Department of Otolaryngology, Medical School, University of Michigan, Ann Arbor, MI, USA; Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, USA.
| | - Giuseppe Pannone
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Li Zheng
- Department of Otolaryngology, Medical School, University of Michigan, Ann Arbor, MI, USA; Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Imad About
- Aix-Marseille Université, CNRS, ISM UMR 7287, 13288, Marseille cedex 09, France
| | - Nawar Taqi
- Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Nghia P T Nguyen
- Department of Otolaryngology, Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Margarite Matossian
- Department of Otolaryngology, Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Blake McAlpin
- Department of Otolaryngology, Medical School, University of Michigan, Ann Arbor, MI, USA; Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Angela Santoro
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Jonathan McHugh
- Department of Pathology, Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Mark E Prince
- Department of Otolaryngology, Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Petros Papagerakis
- Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan, Ann Arbor, MI, USA; Center for Computational Medicine and Bioinformatics, School of Medicine, University of Michigan, Ann Arbor, MI, USA; Center for Organogenesis, School of Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
30
|
Herceptin resistance database for understanding mechanism of resistance in breast cancer patients. Sci Rep 2014; 4:4483. [PMID: 24670875 PMCID: PMC3967150 DOI: 10.1038/srep04483] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 03/07/2014] [Indexed: 01/15/2023] Open
Abstract
Monoclonal antibody Trastuzumab/Herceptin is considered as frontline therapy for Her2-positive breast cancer patients. However, it is not effective against several patients due to acquired or de novo resistance. In last one decade, several assays have been performed to understand the mechanism of Herceptin resistance with/without supplementary drugs. This manuscript describes a database HerceptinR, developed for understanding the mechanism of resistance at genetic level. HerceptinR maintains information about 2500 assays performed against various breast cancer cell lines (BCCs), for improving sensitivity of Herceptin with or without supplementary drugs. In order to understand Herceptin resistance at genetic level, we integrated genomic data of BCCs that include expression, mutations and copy number variations in different cell lines. HerceptinR will play a vital role in i) designing biomarkers to identify patients eligible for Herceptin treatment and ii) identification of appropriate supplementary drug for a particular patient. HerceptinR is available at http://crdd.osdd.net/raghava/herceptinr/.
Collapse
|
31
|
Wang TB, Hu BG, Liu DW, Shi HP, Dong WG. The influence of lentivirus-mediated CXCR4 RNA interference on hepatic metastasis of colorectal cancer. Int J Oncol 2014; 44:1861-9. [PMID: 24647809 PMCID: PMC4063541 DOI: 10.3892/ijo.2014.2348] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Accepted: 02/26/2014] [Indexed: 12/19/2022] Open
Abstract
The aim of this study was to construct a lentiviral vector of CXCR4-siRNA (Lenti-CXCR4-siRNA) and investigate whether the vector can inhibit the growth, migration, invasion and hepatic metastasis of colorectal cancer (CRC). RT-PCR and western blotting were employed to identify the ideal RNA interference sequence. Lenti-CXCR4-siRNA was constructed and transfected into the SW480 cell line. We used RT-PCR and western blotting to measure the expression of CXCR4 RNA and protein, respectively; the MTS assay to assess the proliferation of SW480 cells; transwell chambers to estimate the inhibitory effect on migration and invasion; and the Balb/c nude mouse model of CRC to examine the inhibition of hepatic metastasis. The relative expression of the CXCR4 gene and protein was 5.4 and 18.95%, respectively, in the siCXCR4 group. The genes in the expression plasmid pLenti-CXCR4-siRNA were in the correct order. In the SW480, nonsense control (NC) and the Lenti-CXCR4-siRNA groups CXCR4 RNA levels were, respectively, 0.54±0.06, 1.00±0.03 and 0.11±0.04 (P=0.0001); CXCR4 protein levels were 0.60±0.03, 0.72±0.03 and 0.18±0.02 (P=0.0001); the OD value was 1.38±0.04 (P=0.0050), 1.28±0.05 (P=0.0256) and 0.92±0.06; SW480 cell number in migration test was 32±6.85, 32.63±1.69 and 0.75±0.71 (P=0.0000); SW480 cell number in the invasion test was 29.13±10.3, 30.38±6.09 and 0.63±0.74 (P=0.0000); hepatic metastasis number was 7.10±3.98 (P=0.034), 7.50±4.09 (P=0.019) and (3.50±2.51); hepatic metastasis mean weight (in g) was 2.25±2.51 (P=0.000), 2.11±2.38 (P=0.000) and 1.45±2.07. Lenti-CXCR4-siRNA constructs were correctly constructed and effectively inhibit the expression of CXCR4 RNA and protein, reducing the proliferation, migration, invasion capacity of SW480 cells and hepatic metastasis of CRC.
Collapse
Affiliation(s)
- Tian-Bao Wang
- Department of Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Bao-Guang Hu
- Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, P.R. China
| | - Da-Wei Liu
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Han-Ping Shi
- Department of Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Wen-Guang Dong
- Department of Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
32
|
Ameis HM, Drenckhan A, von Loga K, Escherich G, Wenke K, Izbicki JR, Reinshagen K, Gros SJ. PGK1 as predictor of CXCR4 expression, bone marrow metastases and survival in neuroblastoma. PLoS One 2013; 8:e83701. [PMID: 24376734 PMCID: PMC3869792 DOI: 10.1371/journal.pone.0083701] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 11/06/2013] [Indexed: 12/03/2022] Open
Abstract
Background and Aim A close relationship between phosphoglycerate kinase 1 (PGK1) and the CXCR4/SDF1 axis (chemokine receptor 4/stromal cell derived factor 1) has been shown for several cancers. However, the role of PGK1 has not been investigated for neuroblastoma, and PGK1 might be a therapeutic target for this tumor entity. The aim of the current study was to evaluate the role of PGK1 expression in neuroblastoma patients, to determine the impact of PGK1 expression levels on survival, and to correlate PGK1 expression with CXCR4 expression and bone marrow dissemination. Materials and Methods Samples from 22 patients with neuroblastoma that were surgically treated at the University Medical Center Hamburg-Eppendorf were evaluated for expression of PGK1 and CXCR4 using immunohistochemistry. Results were correlated with clinical parameters, metastases and outcome of patients. Immunocytochemistry, proliferation and expression analysis of CXCR4 and PGK1 were performed in neuroblastoma cell lines. Results PGK1 is expressed in neuroblastoma cells. PGK1 expression is significantly positively correlated with CXCR4 expression and tumor dissemination to the bone marrow. Moreover the expression of PGK1 is significantly associated with a negative impact on survival in patients with neuroblastoma. PGK1 is downregulated by inhibition of CXCR4 in neuroblastoma cells. Conclusion PGK1 appears to play an important role for neuroblastoma, predicting survival and tumor dissemination. Further in vivo studies outstanding, it is a candidate target for novel therapeutic strategies.
Collapse
Affiliation(s)
- Helen M. Ameis
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf/Altona Children's Hospital, Hamburg, Germany
| | - Astrid Drenckhan
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katharina von Loga
- Department of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gabriele Escherich
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katharina Wenke
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf/Altona Children's Hospital, Hamburg, Germany
| | - Jakob R. Izbicki
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Konrad Reinshagen
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf/Altona Children's Hospital, Hamburg, Germany
| | - Stephanie J. Gros
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- * E-mail:
| |
Collapse
|
33
|
Clinicopathological and prognostic significance of chemokine receptor CXCR4 overexpression in patients with esophageal cancer: a meta-analysis. Tumour Biol 2013; 35:3709-15. [DOI: 10.1007/s13277-013-1490-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 11/28/2013] [Indexed: 02/06/2023] Open
|
34
|
Tachezy M, Zander H, Gebauer F, von Loga K, Pantel K, Izbicki JR, Bockhorn M. CXCR7 expression in esophageal cancer. J Transl Med 2013; 11:238. [PMID: 24074251 PMCID: PMC3851264 DOI: 10.1186/1479-5876-11-238] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 09/25/2013] [Indexed: 01/01/2023] Open
Abstract
Background The chemokine CXCL12 and its receptor CXCR4 play a major role in tumor invasion, proliferation and metastasis in different malignant diseases, including esophageal carcinoma, amongst others. CXCR7 was recently identified as a novel alternate receptor for CXCL12. The aim of this study was to evaluate the prognostic impact of expression of chemokine receptor CXCR7 in patients with esophageal carcinoma (EC). Methods Expression of CXCR7 in primary tumors, lymph nodes and distant metastases of 299 patients with EC was evaluated by immunohistochemistry on a tissue microarray and compared with clinical and histopathological data. Results In esophageal cancer sections, CXCR7-specific reactivity was apparent in 45% of the squamous cell carcinomas (ESCC), but only occasionally in adenocarcinomas. No correlation between CXCR4 and CXCR7 expression was evident. We correlated expression with clinical and histopathological characteristics, but could not find any association. Conclusions Contrary to the other known CXCL12 receptor, CXCR4, CXCR7 is expressed in ESCC only, underlining the divergent mechanisms and backgrounds of EAC and ESCC. The results of the study do not indicate a significant functional role for CXCR7 in EAC or ESCC of the esophagus. However, its variable expression in the main two main types of EC needs to be further investigated.
Collapse
Affiliation(s)
- Michael Tachezy
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany.
| | | | | | | | | | | | | |
Collapse
|
35
|
Hu T, Yao Y, Yu S, Guo H, Han L, Wang W, Tian T, Hao Y, Liu Z, Nan K, Wang S. Clinicopathologic significance of CXCR4 and Nrf2 in colorectal cancer. J Biomed Res 2013; 27:283-90. [PMID: 23885267 PMCID: PMC3721036 DOI: 10.7555/jbr.27.20130069] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2013] [Revised: 06/06/2013] [Accepted: 06/18/2013] [Indexed: 01/13/2023] Open
Abstract
The CXCR4 and Nrf2 signaling pathways are abnormally activated in response to cellular stress in various types of human cancers. In this study, we examined the expression of CXCR4 and Nrf2 in colorectal cancer (CRC) tissue specimens and investigated their correlation with patient clinicopathologic characteristics. We determined CXCR4 and Nrf2 expression in 76 CRC tissue specimens and paired normal tissue specimens by immunohistochemistry and real-time PCR. We found that the protein and mRNA transcript levels of CXCR4 were significantly higher in CRC tissue specimens than in paired normal tissues, while the expressions of Nrf2 protein and mRNA were increased in CRC tissues compared to distant non-cancerous tissues. High expression level of CXCR4 was positively correlated with poorly differentiated (P = 0.031), more advanced tumor-node-metastasis (TNM) stage (P = 0.019), lymph node metastasis (P = 0.007) and distant metastasis (P = 0.018). However, the expression of Nrf2 protein was positively correlated with larger tumor size (P = 0.049), more advanced TNM stage (P = 0.013), lymph node metastasis (P = 0.016) and distant metastasis (P = 0.023). Moreover, there was a strong relationship between CXCR4 and Nrf2 expression in CRC tissues, indicating that high Nrf2 expression may contribute to CXCR4 overexpression. In addition, combined expression of CXCR4 and Nrf2 strongly correlated with lymph node metastasis and distant metastasis (P = 0.003). Furthermore, we found that combined high expression of CXCR4 and Nrf2 had stronger correlation with lymph node metastasis and distant metastasis than any single molecule did. This study indicated that the abnormal expression of CXCR4 and Nrf2 contributed to the progression of CRC.
Collapse
|
36
|
Miyazaki H, Takabe K, Yeudall WA. Chemokines, chemokine receptors and the gastrointestinal system. World J Gastroenterol 2013; 19:2847-2863. [PMID: 23704819 PMCID: PMC3660811 DOI: 10.3748/wjg.v19.i19.2847] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 11/13/2012] [Accepted: 04/27/2013] [Indexed: 02/06/2023] Open
Abstract
The biological properties of tumor cells are known to be regulated by a multitude of cytokines and growth factors, which include epidermal growth factor receptor agonists and members of the transforming growth factor β family. Furthermore, the recent explosion of research in the field of chemokine function as mediators of tumor progression has led to the possibility that these small, immunomodulatory proteins also play key roles in carcinogenesis and may, therefore, be potential targets for novel therapeutic approaches. In this review, we will summarize recently reported findings in chemokine biology with a focus on the gastrointestinal tract.
Collapse
|
37
|
Drenckhan A, Kurschat N, Dohrmann T, Raabe N, Koenig AM, Reichelt U, Kaifi JT, Izbicki JR, Gros SJ. Effective inhibition of metastases and primary tumor growth with CTCE-9908 in esophageal cancer. J Surg Res 2012; 182:250-6. [PMID: 23117118 DOI: 10.1016/j.jss.2012.09.035] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Revised: 09/18/2012] [Accepted: 09/26/2012] [Indexed: 11/19/2022]
Abstract
BACKGROUND In spite of multimodular treatment, the therapeutic options for esophageal carcinoma are limited, and metastases remain the leading cause of tumor-related mortality. Expression of the chemokine receptor CXCR4 significantly correlates with poor survival rates in patients with esophageal carcinoma and is associated with lymph node and bone marrow metastases. The aim of this study was to evaluate the effect of the CXCR4 antagonist CTCE-9908 on metastatic homing and primary tumor growth in vitro and in vivo in an orthotopic xenograft model of esophageal cancer. MATERIALS AND METHODS OE19 cells were examined for stromal cell-derived factor 1 alpha-mediated migration under CTCE-9908 treatment. The CTCE-9908 treatment was further evaluated in an in vitro proliferation assay and orthotopic esophageal model, accompanied by magnetic resonance imaging. Tumor and metastases were immunohistochemically examined for CXCR4 expression. RESULTS CTCE-9908 has an inhibitory effect on stromal cell-derived factor 1 alpha-mediated migration and proliferation of OE19 cells. Treatment with CTCE-9908 in the orthotopic esophageal model leads to a reduction of metastatic spread and primary tumor growth. This was confirmed by magnetic resonsance imaging. Treatment with CTCE-9908 results in altered CXCR4 expression pattern exhibiting a high degree of variability. CONCLUSION CTCE-9908 effectively inhibits OE19 cell migration and proliferation in vitro, reduces metastases to lung, liver, and lymph nodes in vivo, and moreover leads to tumor growth reduction in an orthotopic model of esophageal carcinoma.
Collapse
Affiliation(s)
- Astrid Drenckhan
- Department of General, Visceral, and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|