1
|
Fellhofer-Hofer J, Franz C, Vey JA, Kahlert C, Kalkum E, Mehrabi A, Halama N, Probst P, Klupp F. Chemokines as Prognostic Factor in Colorectal Cancer Patients: A Systematic Review and Meta-Analysis. Int J Mol Sci 2024; 25:5374. [PMID: 38791414 PMCID: PMC11121014 DOI: 10.3390/ijms25105374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Chemokines orchestrate many aspects of tumorigenic processes such as angiogenesis, apoptosis and metastatic spread, and related receptors are expressed on tumor cells as well as on inflammatory cells (e.g., tumor-infiltrating T cells, TILs) in the tumor microenvironment. Expressional changes of chemokines and their receptors in solid cancers are common and well known, especially in affecting colorectal cancer patient outcomes. Therefore, the aim of this current systematic review and meta-analysis was to classify chemokines as a prognostic biomarker in colorectal cancer patients. A systematic literature search was conducted in PubMed, CENTRAL and Web of Science. Information on the chemokine expression of 25 chemokines in colorectal cancer tissue and survival data of the patients were investigated. The hazard ratio of overall survival and disease-free survival with chemokine expression was examined. The risk of bias was analyzed using Quality in Prognosis Studies. Random effects meta-analysis was performed to determine the impact on overall respectively disease survival. For this purpose, the pooled hazard ratios (HR) and their 95% confidence intervals (CI) were used for calculation. Twenty-five chemokines were included, and the search revealed 5556 publications. A total of thirty-one publications were included in this systematic review and meta-analysis. Overexpression of chemokine receptor CXCR4 was associated with both a significantly reduced overall survival (HR = 2.70, 95%-CI: 1.57 to 4.66, p = 0.0003) as well as disease-free survival (HR = 2.68, 95%-CI: 1.41 to 5.08, p = 0.0026). All other chemokines showed either heterogeneous results or few studies were available. The overall risk of bias for CXCR4 was rated low. At the current level of evidence, this study demonstrates that CXCR4 overexpression in patients with colorectal cancer is associated with a significantly diminished overall as well as disease-free survival. Summed up, this systematic review and meta-analysis reveals CXCR4 as a promising prognostic biomarker. Nevertheless, more evidence is needed to evaluate CXCR4 and its antagonists serving as new therapeutic targets.
Collapse
Affiliation(s)
- Johanna Fellhofer-Hofer
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany; (J.F.-H.); (C.F.); (C.K.); (A.M.); (P.P.)
| | - Clemens Franz
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany; (J.F.-H.); (C.F.); (C.K.); (A.M.); (P.P.)
| | - Johannes A. Vey
- Institute of Medical Biometry (IMBI), University Hospital Heidelberg, Im Neuenheimer Feld 130/3, 69120 Heidelberg, Germany;
| | - Christoph Kahlert
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany; (J.F.-H.); (C.F.); (C.K.); (A.M.); (P.P.)
| | - Eva Kalkum
- Study Center of the German Society of Surgery (SDGC), University of Heidelberg, Im Neuenheimer Feld 130/3, 69120 Heidelberg, Germany;
| | - Arianeb Mehrabi
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany; (J.F.-H.); (C.F.); (C.K.); (A.M.); (P.P.)
| | - Niels Halama
- National Center for Tumor Diseases, Medical Oncology and Internal Medicine VI, Tissue Imaging and Analysis Center, Bioquant, University of Heidelberg, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany;
- Helmholtz Institute for Translational Oncology (HI-TRON), Department of Cancer Immunology & Cancer Immunotherapy, German Cancer Research Center (DKFZ), 55131 Mainz, Germany
| | - Pascal Probst
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany; (J.F.-H.); (C.F.); (C.K.); (A.M.); (P.P.)
- Department of Surgery, Cantonal Hospital Thurgau, Pfaffenholzstrasse 4, 8501 Frauenfeld, Switzerland
| | - Fee Klupp
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany; (J.F.-H.); (C.F.); (C.K.); (A.M.); (P.P.)
| |
Collapse
|
2
|
Huang H, Han L, Guo J, Zhang Y, Lin S, Chen S, Lin X, Cheng C, Guo Z, Qiu Y. Pretreatment MRI-Based Radiomics for Prediction of Rectal Cancer Outcome: A Discovery and Validation Study. Acad Radiol 2024; 31:1878-1888. [PMID: 37996362 DOI: 10.1016/j.acra.2023.10.055] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/22/2023] [Accepted: 10/29/2023] [Indexed: 11/25/2023]
Abstract
RATIONALE AND OBJECTIVES Accurate prediction of local recurrence or distant metastasis is critical for developing individualized therapies for locally advanced rectal cancer (LARC) patients after standard therapy. This study aims to develop and validate a multiparameter MRI-based radiomics signature (RS) for prognostic prediction in LARC patients receiving neoadjuvant chemoradiotherapy (nCRT) and total mesorectal excision (TME) and to explore the ability of RS for personalized survival risk stratification. MATERIALS AND METHODS In this multi-center study, 454 patients who received nCRT and TME and completed 3 years of follow-up participated. RS was constructed for prognostic prediction based on features extracted from pretreatment multiparameter MRI in a training cohort (TC; n = 298), which was tested in an internal validation cohort (IVC; n = 75) and further validated in an independent external validation cohort (EVC; n = 81). Furthermore, the ability of RS for personalized survival risk stratification was explored using the Kaplan-Meier survival curves. RESULTS The RS model showed satisfactory accuracy for prognostic prediction with AUCs of 0.83, 0.81 and 0.82 in the TC, IVC and EVC, respectively. In addition, RS helped to refine risk stratification for LARC patients on the basis of significantly different 3-year disease-free survival rates, independent of their pathological stage, pre-surgery CEA, and even treatment modality. CONCLUSIONS The proposed RS can be used not only to predict local recurrence or distant metastasis but also to serve as an effective postoperative survival risk stratification tool for clinicians to facilitate decision-making for LARC patients receiving standard treatment.
Collapse
Affiliation(s)
- Hongyan Huang
- Department of Radiology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen 518000, P.R. China
| | - Lujun Han
- Department of Medical Imaging, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou 510060, P.R. China
| | - Jianbo Guo
- Department of Radiology, Meizhou People's Hospital, No. 63 Huangtang Road, Meizhou 514000 P.R. China
| | - Yanyu Zhang
- Department of Radiology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Duobao AVE 56, Liwan district, Guangzhou, P.R. China
| | - Shiwei Lin
- Department of Radiology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen 518000, P.R. China
| | - Shengli Chen
- Department of Radiology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen 518000, P.R. China
| | - Xiaoshan Lin
- Department of Radiology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen 518000, P.R. China
| | - Caixue Cheng
- Department of Radiology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen 518000, P.R. China
| | - Zheng Guo
- Department of Hematology and Oncology, International Cancer Center, Shenzhen Key Laboratory of Precision Medicine for Hematological Malignancies, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University Health Science Center, Xueyuan AVE 1098, Nanshan District, Shenzhen, Guangdong 518000, P.R. China
| | - Yingwei Qiu
- Department of Radiology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen 518000, P.R. China.
| |
Collapse
|
3
|
Abstract
For our immune system to contain or eliminate malignant solid tumours, both myeloid and lymphoid haematopoietic cells must not only extravasate from the bloodstream into the tumour tissue but also further migrate to various specialized niches of the tumour microenvironment to functionally interact with each other, with non-haematopoietic stromal cells and, ultimately, with cancer cells. These interactions regulate local immune cell survival, proliferative expansion, differentiation and their execution of pro-tumour or antitumour effector functions, which collectively determine the outcome of spontaneous or therapeutically induced antitumour immune responses. None of these interactions occur randomly but are orchestrated and critically depend on migratory guidance cues provided by chemokines, a large family of chemotactic cytokines, and their receptors. Understanding the functional organization of the tumour immune microenvironment inevitably requires knowledge of the multifaceted roles of chemokines in the recruitment and positioning of its cellular constituents. Gaining such knowledge will not only generate new insights into the mechanisms underlying antitumour immunity or immune tolerance but also inform the development of biomarkers (or 'biopatterns') based on spatial tumour tissue analyses, as well as novel strategies to therapeutically engineer immune responses in patients with cancer. Here we will discuss recent observations on the role of chemokines in the tumour microenvironment in the context of our knowledge of their physiological functions in development, homeostasis and antimicrobial responses.
Collapse
Affiliation(s)
- Thorsten R Mempel
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Julia K Lill
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Lukas M Altenburger
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
4
|
Kim S, Yeo MK, Kim JS, Kim JY, Kim KH. Elevated CXCL12 in the plasma membrane of locally advanced rectal cancer after neoadjuvant chemoradiotherapy: a potential prognostic marker. J Cancer 2022; 13:162-173. [PMID: 34976180 PMCID: PMC8692683 DOI: 10.7150/jca.64082] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 11/10/2021] [Indexed: 02/07/2023] Open
Abstract
Background: Neoadjuvant chemoradiotherapy (nCRT) in locally advanced rectal cancer (LARC) has been shown to improve sphincter preservation and local pelvic control, but the efficacy of nCRT plateaus due to metastasis. CXC chemokine ligand 12 (CXCL12) has a critical impact on cancer development and metastasis. Methods: By investigating public databases containing LARC patient data, CXCL12, CXCR4 and FAPα expression was analyzed via the Tumor Immune Estimation Resource (TIMER) and GSEA. Immunohistochemistry was applied to a total of 121 surgically resected specimens consisting of 61 LARCs after nCRT and 60 LARCs with no nCRT and 16 cases with endoscopic resection of high-grade colorectal adenoma. Results: By investigating public databases containing LARC patient data, CXCL12 expression is correlated with poor prognosis, immune cell infiltration, epithelial- mesenchymal transition, and angiogenesis in LARC. Furthermore, radiation selectively induced CXCL12, CXCR4 and FAPα expression in tumor tissues. Immunohistochemistry results showed that the levels of CXCL12, CXCR4, and FAPα in LARC cells after nCRT were higher than in LARC cells untreated with nCRT (p < 0.001 for each). Elevated levels of CXCL12 in the plasma membrane of LARC cells after nCRT demonstrated an association with the period of freedom from recurrence (FFR) in univariate and multivariate survival analyses (p = 0.005 and p = 0.031, respectively). Conclusions: The expression of CXCL12 may influence the survival and invasive properties of LARC cells during nCRT and promote cancer recurrence. We suggest that CXCL12 expression in the plasma membrane of radioresistant LARC cells may be a predictive factor of recurrence and a viable therapeutic strategy to control radioresistant LARC recurrence.
Collapse
Affiliation(s)
- Sup Kim
- Department of Radiation Oncology, Chungnam National University School of Medicine, 288 Munhwa Street, Daejeon 35015, Korea.,Department of Radiation Oncology, Chungnam National University Hospital, 282 Munwha-ro, Daejeon 35015, Korea
| | - Min-Kyung Yeo
- Department of Pathology, Chungnam National University School of Medicine, 266 Munhwa Street, Daejeon 35015, Korea.,Department of Pathology, Chungnam National University Hospital, 282 Munwha-ro, Daejeon 35015, Korea
| | - Jun-Sang Kim
- Department of Radiation Oncology, Chungnam National University School of Medicine, 288 Munhwa Street, Daejeon 35015, Korea.,Department of Radiation Oncology, Chungnam National University Hospital, 282 Munwha-ro, Daejeon 35015, Korea
| | - Ji-Yeon Kim
- Department of Surgery, Division of Colorectal Surgery, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Kyung-Hee Kim
- Department of Pathology, Chungnam National University School of Medicine, 266 Munhwa Street, Daejeon 35015, Korea.,Department of Pathology, Chungnam National University Sejong Hospital, 20 Bodeum 7-ro, Sejong-si 30099, Korea
| |
Collapse
|
5
|
Jovani M, Liu EE, Paniagua SM, Lau ES, Li SX, Takvorian KS, Kreger BE, Splansky GL, de Boer RA, Joshi AD, Hwang SJ, Yao C, Huan T, Courchesne P, Larson MG, Levy D, Chan AT, Ho JE. Cardiovascular disease related circulating biomarkers and cancer incidence and mortality: is there an association? Cardiovasc Res 2021; 118:2317-2328. [PMID: 34469519 DOI: 10.1093/cvr/cvab282] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/25/2020] [Accepted: 08/30/2021] [Indexed: 12/14/2022] Open
Abstract
AIMS Recent studies suggest an association between cardiovascular disease (CVD) and cancer incidence/mortality, but the pathophysiological mechanisms underlying these associations are unclear. We aimed to examine biomarkers previously associated with CVD and study their association with incident cancer and cancer-related death in a prospective cohort study. METHODS AND RESULTS We used a proteomic platform to measure 71 cardiovascular biomarkers among 5,032 participants in the Framingham Heart Study who were free of cancer at baseline. We used multivariable-adjusted Cox models to examine the association of circulating protein biomarkers with risk of cancer incidence and mortality. To account for multiple testing, we set a 2-sided false discovery rate (FDR Q-value) <0.05.Growth differentiation factor-15 (GDF15; also known as macrophage inhibitory cytokine-1 [MIC1])) was associated with increased risk of incident cancer (hazards ratio [HR] per 1 standard deviation increment 1.31, 95% CI 1.17-1.47), incident gastrointestinal cancer (HR 1.85, 95% CI 1.37-2.50), incident colorectal cancer (HR 1.94, 95% CI 1.29-2.91) and cancer-related death (HR 2.15, 95% CI 1.72-2.70). Stromal cell-derived factor-1 (SFD1) showed an inverse association with cancer-related death (HR 0.75, 95% CI 0.65-0.86). Fibroblast growth factor-23 (FGF23) showed an association with colorectal cancer (HR 1.55, 95% CI 1.20-2.00), and granulin (GRN) was associated with hematologic cancer (HR 1.61, 95% CI 1.30-1.99). Other circulating biomarkers of inflammation, immune activation, metabolism, and fibrosis showed suggestive associations with future cancer diagnosis. CONCLUSION We observed several significant associations between circulating CVD biomarkers and cancer, supporting the idea that shared biological pathways underlie both diseases. Further investigations of specific mechanisms that lead to both CVD and cancer are warranted. TRANSLATIONAL PERSPECTIVE In our prospective cohort study, baseline levels of biomarkers previously associated with CVD were found to be associated with future development of cancer. In particular, GDF15 was associated with increased risk of cancer incidence and mortality, including gastrointestinal and colorectal cancers; SDF1 was inversely associated with cancer-related death, and FGF23 and GRN were associated with increased risk of colorectal and hematologic cancers, respectively. Other biomarkers of inflammation, immune activation, metabolism, and fibrosis showed suggestive associations. These results suggest potential shared biological pathways that underlie both development of cancer and CVD.
Collapse
Affiliation(s)
- Manol Jovani
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA.,Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA.,Harvard Medical School, Boston, MA.,Division of Gastroenterology; University of Kentucky Albert B. Chandler Hospital
| | - Elizabeth E Liu
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA
| | | | - Emily S Lau
- Corrigan Minehan Heart Center, Massachusetts General Hospital, Boston, MA.,Cardiology Division, Massachusetts General Hospital, Boston, MA.,Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Shawn X Li
- Department of Medicine, Massachusetts General Hospital, Boston, MA
| | | | - Bernard E Kreger
- General Internal Medicine, Department of Medicine, Boston University School of Medicine, Boston, MA.,The Framingham Heart Study, Framingham, MA
| | | | - Rudolf A de Boer
- Department of Cardiology, University Medical Centre Groningen, Groningen, The Netherlands
| | - Amit D Joshi
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA.,Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA.,Harvard Medical School, Boston, MA
| | - Shih-Jen Hwang
- The Framingham Heart Study, Framingham, MA.,Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda MD
| | - Chen Yao
- The Framingham Heart Study, Framingham, MA.,Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda MD
| | - Tianxiao Huan
- The Framingham Heart Study, Framingham, MA.,Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda MD
| | - Paul Courchesne
- The Framingham Heart Study, Framingham, MA.,Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda MD
| | - Martin G Larson
- The Framingham Heart Study, Framingham, MA.,Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Daniel Levy
- The Framingham Heart Study, Framingham, MA.,Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda MD
| | - Andrew T Chan
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA.,Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA.,Harvard Medical School, Boston, MA
| | - Jennifer E Ho
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA.,Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA.,Corrigan Minehan Heart Center, Massachusetts General Hospital, Boston, MA.,Cardiology Division, Massachusetts General Hospital, Boston, MA.,Department of Medicine, Massachusetts General Hospital, Boston, MA.,Harvard Medical School, Boston, MA
| |
Collapse
|
6
|
New CXCR4 Antagonist Peptide R (Pep R) Improves Standard Therapy in Colorectal Cancer. Cancers (Basel) 2020; 12:cancers12071952. [PMID: 32708431 PMCID: PMC7409147 DOI: 10.3390/cancers12071952] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/14/2020] [Accepted: 07/16/2020] [Indexed: 12/18/2022] Open
Abstract
The chemokine receptor CXCR4 is overexpressed and functional in colorectal cancer. To investigate the role of CXCR4 antagonism in potentiating colon cancer standard therapy, the new peptide CXCR4 antagonist Peptide R (Pep R) was employed. Human colon cancer HCT116 xenograft-bearing mice were treated with chemotherapeutic agents (CT) 5-Fluorouracil (5FU) and oxaliplatin (OX) or 5FU and radio chemotherapy (RT-CT) in the presence of Pep R. After two weeks, CT plus Pep R reduced by 4-fold the relative tumor volume (RTV) as compared to 2- and 1.6-fold reductions induced, respectively, by CT and Pep R. In vitro Pep R addition to CT/RT-CT impaired HCT116 cell growth and further reduced HCT116 and HT29 clonal capability. Thus, the hypothesis that Pep R could target the epithelial mesenchyme transition (EMT) process was evaluated. While CT decreased ECAD and increased ZEB-1 and CD90 expression, the addition of Pep R restored the pretreatment expression. In HCT116 and HT29 cells, CT/RT-CT induced a population of CD133+CXCR4+ cells, supposedly a stem-resistant cancer cell population, while Pep R reduced it. Taken together, the results showed that targeting CXCR4 ameliorates the effect of treatment in colon cancer through inhibition of cell growth and reversal of EMT treatment-induced markers, supporting further clinical studies.
Collapse
|
7
|
Liang T, Tong W, Ma S, Chang P. Standard therapies: solutions for improving therapeutic effects of immune checkpoint inhibitors on colorectal cancer. Oncoimmunology 2020; 9:1773205. [PMID: 32934878 PMCID: PMC7466849 DOI: 10.1080/2162402x.2020.1773205] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Immunotherapy using immune checkpoint inhibitors has opened a new era for cancer management. In colorectal cancer, patients with a phenotype of deficient mismatch repair or high microsatellite instability benefit from immunotherapy. However, the response of rest cases to immunotherapy alone is still poor. Nevertheless, preclinical data have revealed that either ionizing irradiation or chemotherapy can improve the tumoral immune milieu, because these approaches can induce immunogenic cell death among cancer cells. In this regard, combination use of standard therapy plus immunotherapy should be feasible. In this review, we will introduce the specific roles of standard therapies, including radiotherapy, chemotherapy, antiangiogenic and anti-EGFR therapy, in improving therapeutic effect of immune checkpoint inhibitors on colorectal cancer.
Collapse
Affiliation(s)
- Tingting Liang
- Oncology Department, The First Hospital of Jilin University, Changchun, P.R. China
| | - Weihua Tong
- Department of Gastrointestinal and Colorectal Surgery, The First Hospital of Jilin University, Changchun, P.R. China
| | - Siyang Ma
- Department of Radiation Oncology & Therapy, Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, P.R. China
| | - Pengyu Chang
- Department of Radiation Oncology & Therapy, Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, P.R. China
| |
Collapse
|
8
|
Li J, Zhang J, Hu H, Cai Y, Ling J, Wu Z, Deng Y. Gene Expression Signature to Predict Prognosis and Adjuvant Chemosensitivity of Colorectal Cancer Patients. Cancer Manag Res 2020; 12:3301-3310. [PMID: 32494194 PMCID: PMC7227814 DOI: 10.2147/cmar.s243490] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 04/16/2020] [Indexed: 12/22/2022] Open
Abstract
Purpose Molecular characteristics using gene-expression profiling can undoubtedly improve the prediction of treatment responses, and ultimately, the clinical outcome of cancer patients. We aimed at developing a genetic signature to improve the prediction of chemosensitivity and prognosis of patients with colorectal cancer (CRC). Patients and Methods We analyzed microarray data of 32 CRC patients to explore the potential functions and pathways involved in the disease relapse in CRC. Gene expression profiles and clinical follow-up information of GSE39582, GSE17536, and GSE103479 were downloaded from the Gene Expression Omnibus database (GEO) to identify prognostic genes. Eventually, a model of 15-mRNA signature was established, in which its efficacy for predicting chemosensitivity and prognosis was examined. Results Based on the proposed model of 15-mRNA signature, the test series patients could be classified into high-risk or low-risk subgroup with significantly different overall survival (OS) rate (hazard ratio [HR]=1.48, 95% confidence interval [CI]=1.30–1.70, P≤0.001). The prognostic value of this 15-mRNA signature was confirmed in another validation series. Further analysis revealed that the prognostic value of this signature was independent of the TNM stage and can predict adjuvant chemosensitivity of patients with early-stage CRC. Conclusion We identified a novel 15-mRNA signature in patients with CRC, which could be clinically helpful in the prognosis evaluation and the process of selection of patients with early-stage CRC for undergoing adjuvant chemotherapy.
Collapse
Affiliation(s)
- Jianxia Li
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangdong Province Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, Guangdong 510655, People's Republic of China
| | - Jianwei Zhang
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangdong Province Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, Guangdong 510655, People's Republic of China
| | - Huabin Hu
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangdong Province Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, Guangdong 510655, People's Republic of China
| | - Yue Cai
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangdong Province Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, Guangdong 510655, People's Republic of China
| | - Jiayu Ling
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangdong Province Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, Guangdong 510655, People's Republic of China
| | - Zehua Wu
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangdong Province Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, Guangdong 510655, People's Republic of China
| | - Yanhong Deng
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangdong Province Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, Guangdong 510655, People's Republic of China
| |
Collapse
|
9
|
Jiang Q, Sun Y, Liu X. CXCR4 as a prognostic biomarker in gastrointestinal cancer: a meta-analysis. Biomarkers 2019; 24:510-516. [PMID: 31244335 DOI: 10.1080/1354750x.2019.1637941] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Background: CXCR4 is a member of the C-X-C chemokine receptor family, which is associated with multiple types of cancer. Although it has been widely reported, the prognostic value of CXCR4 expression in gastrointestinal (GI) cancer remains controversial. Methods: A meta-analysis was conducted to investigate the relationship between CXCR4 and prognosis of patients with GI cancer. Subgroup analysis was also performed according to tumour subtypes and heterogeneity test. Results: A total of 24 studies including 3637 cases suggested that overexpression of CXCR4 is significantly associated with overall survival (OS) for patients with GI cancer (HR = 1.71, 95% CI = 1.45-2.03, p = 0.000). Subgroup analysis also indicated that high CXCR4 expression in oesophagus, gastric and colorectal cancer all predicted a worse prognosis (HR = 1.52, 95% CI = 1.26-1.84, p = 0.001 for oesophagus cancer; HR = 1.59, 95% CI = 1.10-2.30, p = 0.015 for gastric cancer; HR = 2.21, 95% CI = 1.56-3.14, p = 0.000 for colorectal cancer). Conclusions: CXCR4 may serve as a prognostic indicator in GI cancer patients.
Collapse
Affiliation(s)
- Qingtao Jiang
- a Department of Medicine, Jiangsu Health Vocational College , Nanjing , China
| | - Yun Sun
- b Center for Disease Prevention and Control of Changzhou , Changzhou , China
| | - Xin Liu
- c Institute of Occupational Disease Prevention, Jiangsu Provincial Center for Disease Prevention and Control , Nanjing , China
| |
Collapse
|
10
|
Wang XC, Yue X, Zhang RX, Liu TY, Pan ZZ, Yang MJ, Lu ZH, Wang ZY, Peng JH, Le LY, Wang GY, Peng QH, Meng Y, Huang W, Liu RY. Genome-wide RNAi Screening Identifies RFC4 as a Factor That Mediates Radioresistance in Colorectal Cancer by Facilitating Nonhomologous End Joining Repair. Clin Cancer Res 2019; 25:4567-4579. [PMID: 30979744 DOI: 10.1158/1078-0432.ccr-18-3735] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 01/14/2019] [Accepted: 04/09/2019] [Indexed: 12/24/2022]
Abstract
PURPOSE Neoadjuvant chemoradiotherapy (neoCRT) is a standard treatment for locally advanced rectal cancer (LARC); however, resistance to chemoradiotherapy is one of the main obstacles to improving treatment outcomes. The goal of this study was to identify factors involved in the radioresistance of colorectal cancer and to clarify the underlying mechanisms. EXPERIMENTAL DESIGN A genome-wide RNAi screen was used to search for candidate radioresistance genes. After RFC4 knockdown or overexpression, colorectal cancer cells exposed to X-rays both in vitro and in a mouse model were assayed for DNA damage, cytotoxicity, and apoptosis. Moreover, the regulatory effects and mechanisms of RFC4 in DNA repair were investigated in vitro. Finally, the relationships between RFC4 expression and clinical parameters and outcomes were investigated in 145 patients with LARC receiving neoCRT. RESULTS RFC4, NCAPH, SYNE3, LDLRAD2, NHP2, and FICD were identified as potential candidate radioresistance genes. RFC4 protected colorectal cancer cells from X-ray-induced DNA damage and apoptosis in vitro and in vivo. Mechanistically, RFC4 promoted nonhomologous end joining (NHEJ)-mediated DNA repair by interacting with Ku70/Ku80 but did not affect homologous recombination-mediated repair. Higher RFC4 expression in cancer tissue was associated with weaker tumor regression and poorer prognosis in patients with LARC treated with neoCRT, which likely resulted from the effect of RFC4 on radioresistance, not chemoresistance. CONCLUSIONS RFC4 was identified as a radioresistance factor that promotes NHEJ-mediated DNA repair in colorectal cancer cells. In addition, the expression level of RFC4 predicted radiotherapy responsiveness and the outcome of neoadjuvant radiotherapy in patients with LARC.
Collapse
Affiliation(s)
- Xue-Cen Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xin Yue
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Rong-Xin Zhang
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ting-Yu Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zhi-Zhong Pan
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Meng-Jie Yang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Zhen-Hai Lu
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zi-Yang Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jian-Hong Peng
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Li-Yuan Le
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Gao-Yuan Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Qi-Hua Peng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yuan Meng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wenlin Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China. .,Guangdong Provincial Key Laboratory of Tumor Targeted Drugs and Guangzhou Enterprise Key Laboratory of Gene Medicine, Guangzhou Doublle Bioproducts Co. Ltd., Guangzhou, China
| | - Ran-Yi Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
11
|
Fan H, Wang W, Yan J, Xiao L, Yang L. Prognostic significance of CXCR7 in cancer patients: a meta-analysis. Cancer Cell Int 2018; 18:212. [PMID: 30574021 PMCID: PMC6300004 DOI: 10.1186/s12935-018-0702-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 12/07/2018] [Indexed: 02/07/2023] Open
Abstract
Background CXC chemokine receptor 7 (CXCR7) is frequently overexpressed in a variety of tumors. Nevertheless, whether CXCR7 can be used as a tumor prognosis marker has not been systematically assessed. The current meta-analysis was performed to obtain an accurate evaluation of the relationship between CXCR7 level and the prognosis of cancer patients. Methods Embase, Web of Science, and PubMed were systematically searched according to a defined search strategy up to June 11, 2018. Then, the required data were extracted from all qualified studies which were screened out based on the defined inclusion and exclusion criteria. Finally, the hazard ratios (HR) with 95% confidence intervals (CI) were used to evaluate the prognostic significance of CXCR7 in tumor patients. Results A total of 28 original research studies comprising 33 cohorts and 5685 patients were included in this meta-analysis. The results showed that CXCR7 overexpression was significantly related to worse overall survival (OS) (HR 1.72; 95% CI 1.49–1.99), disease-free survival (DFS) (HR 5.58; 95% CI 3.16–9.85), progression-free survival (PFS) (HR 2.83; 95% CI 1.66–4.85) and recurrence-free survival (RFS) (HR 1.58; 95% CI 1.34–1.88) in cancer patients. Furthermore, for certain types of cancer, significant associations between higher CXCR7 expression and worse OS of glioma (HR 1.77; 95% CI 1.43–2.19), breast cancer (HR 1.45; 95% CI 1.28–1.63), esophageal cancer (HR 2.72; 95% CI 1.11–6.66) and pancreatic cancer (HR 1.46; 95% CI 1.12–1.90) were found. However, for lung cancer and hepatocellular cancer, there was no significant relationship between CXCR7 expression level and OS, (HR 2.40; 95% CI 0.34–17.07) and (HR 1.37; 95% CI 0.84–2.24) respectively. Conclusions Increased CXCR7 level could predict poor prognosis of tumor patients and might be regarded as a novel prognostic biomarker for tumor patients.
Collapse
Affiliation(s)
- Huiqian Fan
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weijun Wang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingjing Yan
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Xiao
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Yang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
12
|
Zhao Q, Zhang P, Qin G, Ren F, Zheng Y, Qiao Y, Sun T, Zhang Y. Role of CXCR7 as a Common Predictor for Prognosis in Solid Tumors: a Meta-Analysis. J Cancer 2018; 9:3138-3148. [PMID: 30210637 PMCID: PMC6134830 DOI: 10.7150/jca.25377] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 07/07/2018] [Indexed: 01/20/2023] Open
Abstract
Background: Accumulating evidence indicated that the CXC chemokine receptor (CXCR) 7 (CXCR7) was overexpressed in a variety of tumors. However, the value of the CXCR7 expression in predicting prognosis in solid tumors remains controversial. Therefore, we performed this meta-analysis to evaluate the correlation between CXCR7 expression and lymph node metastasis (LNM), tumor pathological grade and survival, including overall survival (OS), disease-free survival (DFS) and recurrence-free survival (RFS). Methods: Eligible studies were searched in PubMed, Web of Science, and PMC up to April 2018. A total of 27 studies were included in this meta-analysis. Odds ratio (OR), hazard ratio (HR) and 95 % confidence intervals (CI) were used as effect measures. Results: The meta-analysis showed that high expression of CXCR7 predicted a high risk of LNM (pooled OR = 2.22, 95%CI: 1.41-3.50), high tumor grade (pooled OR = 1.94, 95%CI: 1.20-3.13), poor OS (pooled HR = 1.66, 95%CI: 1.30-2.03), and poor DFS/RFS (pooled HR = 1.82, 95%CI: 1.21-2.43). Subgroup analysis showed that CXCR7 expression had a positive correlation with LNM in pan-adenocarcinoma subgroup (pooled OR = 3.73, 95%CI: 2.21-6.30), while no correlation was found in pan-squamous cancer subgroup (pooled OR = 1.29, 95%CI: 0.56-2.96). Subgroup analysis of tumor grade revealed that high expression of CXCR7 predicted high tumor grade both in pan-squamous cancer and pan-adenocarcinoma (pooled OR = 3.58, 95%CI: 1.39-9.22, pooled OR = 2.25, 95%CI: 1.20-4.20). As in OS group, we divided the data based on analysis method and it turned out that overexpressed CXCR7 predicted worse OS both in multivariate analysis (pooled HR =1.57, 95%CI: 1.12-2.01) and univariate analysis subgroup (pooled HR =1.86, 95%CI: 1.23-2.49). Conclusions: Our meta-analysis revealed that high expression of CXCR7 predicted unfavorable prognosis and may serve as potential targets of cancer therapy.
Collapse
Affiliation(s)
- Qitai Zhao
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.,Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Penghua Zhang
- Imaging Department, the Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Guohui Qin
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.,Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Feifei Ren
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.,Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.,School of Life Sciences, Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Yujia Zheng
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.,Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Yamin Qiao
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.,Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Ting Sun
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.,Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Yi Zhang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.,Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.,School of Life Sciences, Zhengzhou University, Zhengzhou 450052, Henan, China.,Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
13
|
Moreno MJ, Gallardo A, Novelli S, Mozos A, Aragó M, Pavón MÁ, Céspedes MV, Pallarès V, Falgàs A, Alcoceba M, Blanco O, Gonzalez-Díaz M, Sierra J, Mangues R, Casanova I. CXCR7 expression in diffuse large B-cell lymphoma identifies a subgroup of CXCR4+ patients with good prognosis. PLoS One 2018; 13:e0198789. [PMID: 29920526 PMCID: PMC6007902 DOI: 10.1371/journal.pone.0198789] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 04/14/2018] [Indexed: 01/10/2023] Open
Abstract
The CXCR4/CXCL12 axis has been extensively associated with different types of cancer correlating with higher aggressiveness and metastasis. In diffuse large B-cell lymphoma (DLBCL), the expression of the chemokine receptor CXCR4 is involved in the dissemination of malignant B cells and is a marker of poor prognosis. CXCR7 is a chemokine receptor that binds to the same ligand as CXCR4 and regulates de CXCR4-CXCL12 axis. These findings together with the report of CXCR7 prognostic value in several tumor types, led us to evaluate the expression of CXCR7 in diffuse large B-cell lymphoma biopsies. Here, we describe that CXCR7 receptor is an independent prognostic factor that associates with good clinical outcome. Moreover, the expression of CXCR7 associates with increased survival in CXCR4+ but not in CXCR4- DLBCL patients. Thus, the combined immunohistochemical evaluation of both CXCR7 and CXCR4 expression in DLBCL biopsies may improve their prognostic value as single markers. Finally, we show that CXCR7 overexpression in vitro is able to diminish DLBCL cell survival and increase their sensitivity to antitumor drugs. Hence, further studies on the CXCR7 receptor may establish its role in DLBCL and the molecular mechanisms that modulate CXCR4 activity.
Collapse
MESH Headings
- Adult
- Aged
- Biomarkers, Tumor
- Biopsy
- Cell Line, Tumor
- Chemokine CXCL12/physiology
- Drug Resistance, Neoplasm/genetics
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Kaplan-Meier Estimate
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lymphoma, Large B-Cell, Diffuse/pathology
- Male
- Middle Aged
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Neoplasm Proteins/physiology
- Prognosis
- Proportional Hazards Models
- Receptors, CXCR/biosynthesis
- Receptors, CXCR/genetics
- Receptors, CXCR/physiology
- Receptors, CXCR4/analysis
Collapse
Affiliation(s)
- María José Moreno
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomecidicina (CIBER-BBN), Barcelona, Spain
| | - Alberto Gallardo
- Department of Pathology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Silvana Novelli
- Department of Hematology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Ana Mozos
- Department of Pathology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Marc Aragó
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Miguel Ángel Pavón
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomecidicina (CIBER-BBN), Barcelona, Spain
| | - María Virtudes Céspedes
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomecidicina (CIBER-BBN), Barcelona, Spain
| | - Víctor Pallarès
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Department of Hematology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Aïda Falgàs
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Miguel Alcoceba
- Department of Hematology and Pathology, IBSAL-University Hospital, Center for Cancer Research-IBMCC (USAL-CSIC), Salamanca, Spain
- CIBER in Oncology (CIBER-ONC), Madrid, Spain
| | - Oscar Blanco
- Department of Hematology and Pathology, IBSAL-University Hospital, Center for Cancer Research-IBMCC (USAL-CSIC), Salamanca, Spain
| | - Marcos Gonzalez-Díaz
- Department of Hematology and Pathology, IBSAL-University Hospital, Center for Cancer Research-IBMCC (USAL-CSIC), Salamanca, Spain
- CIBER in Oncology (CIBER-ONC), Madrid, Spain
| | - Jorge Sierra
- Department of Hematology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Josep Carreras Research Institute, Barcelona, Spain
| | - Ramon Mangues
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomecidicina (CIBER-BBN), Barcelona, Spain
- Josep Carreras Research Institute, Barcelona, Spain
- * E-mail:
| | - Isolda Casanova
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomecidicina (CIBER-BBN), Barcelona, Spain
- Josep Carreras Research Institute, Barcelona, Spain
| |
Collapse
|
14
|
Zhao K, Li Z, Tian H. Twenty-gene-based prognostic model predicts lung adenocarcinoma survival. Onco Targets Ther 2018; 11:3415-3424. [PMID: 29928133 PMCID: PMC6003292 DOI: 10.2147/ott.s158638] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Introduction Lung adenocarcinoma (LAC) accounts for more than a half of non-small cell lung cancer with high morbidity and mortality. Progression of treatment has not accelerated the improvement of its prognosis. Hence, it is an urgent need to develop novel biomarkers for its early diagnosis and treatment. Materials and methods In this study, we proposed to identify LAC survival-related genes through comprehensive analysis of large-scale gene expression profiles. LAC gene expression data sets were obtained from The Cancer Genome Atlas (TCGA). Identification of differentially expressed genes (DEGs) in LAC compared with adjacent normal lung tissues was first performed followed by univariate Cox regression analysis to obtain genes that are significantly associated with LAC survival (SurGenes). Then, we conducted sure independence screening (SIS) for SurGenes to identify more reliable genes and the prognostic signature for LAC survival prediction. Another two lung cancer data sets from TCGA and Gene Expression Omnibus (GEO) were used for the validation of prognostic signature. Results A total of 20 genes were obtained, which were significantly associated with the overall survival (OS) of LAC patients. The prognostic signature, a weighted linear combination of the 20 genes, could successfully separate LAC samples with high OS from those with low OS and had robust predictive performance for survival (training set: p-value <2.2×10−16; testing set: p-value =2.04×10−5, area under the curve (AUC) =0.615). Combined with GEO data set, we obtained four genes, that is, FUT4, SLC25A42, IGFBP1, and KLHDC8B that are found in both the prognostic signature and DEGs of LAC in GEO data set. Discussion The prognostic signature combined with multi-gene expression profiles provides a moderate OS prediction for LAC and should be helpful for appropriate treatment method selection.
Collapse
Affiliation(s)
- Kai Zhao
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, China.,Department of Thoracic Surgery, Zibo Central Hospital, Zibo, Shandong Province, China
| | - Zulei Li
- Department of Thoracic Surgery, Zibo Central Hospital, Zibo, Shandong Province, China
| | - Hui Tian
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| |
Collapse
|
15
|
Nesseler JP, Schaue D, McBride WH, Nickers P. [Inflammatory and immune biomarkers of radiation response]. Cancer Radiother 2018; 22:180-192. [PMID: 29650389 DOI: 10.1016/j.canrad.2017.09.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 09/08/2017] [Indexed: 02/07/2023]
Abstract
In radiotherapy, the treatment is adapted to each individual to protect healthy tissues but delivers most of time a standard dose according to the tumor histology and site. The only biomarkers studied to individualize the treatment are the HPV status with radiation dose de-escalation strategies, and tumor hypoxia with dose escalation to hypoxic subvolumes using FMISO- or FAZA-PET imaging. In the last decades, evidence has grown about the contribution of the immune system to radiation tumor response. Many preclinical studies have identified some of the mechanisms involved. In this context, we have realised a systematic review to highlight potential inflammatory and immune biomarkers of radiotherapy response. Some are inside the tumor microenvironment, as lymphocyte infiltration or PD-L1 expression, others are circulating biomarkers, including different types of hematological cells, cytokines and chemokines.
Collapse
Affiliation(s)
- J P Nesseler
- Department of radiation oncology, David Geffen school of medicine, university of California at Los Angeles, 10833 Le Conte avenue, 90095-1714 Los Angeles, CA, États-Unis.
| | - D Schaue
- Department of radiation oncology, David Geffen school of medicine, university of California at Los Angeles, 10833 Le Conte avenue, 90095-1714 Los Angeles, CA, États-Unis
| | - W H McBride
- Department of radiation oncology, David Geffen school of medicine, university of California at Los Angeles, 10833 Le Conte avenue, 90095-1714 Los Angeles, CA, États-Unis
| | - P Nickers
- Départment de radiothérapie, centre François-Baclesse, rue Émile-Mayrisch, 4240 Esch-sur-Alzette, Luxembourg
| |
Collapse
|
16
|
Grizzi F, Basso G, Borroni EM, Cavalleri T, Bianchi P, Stifter S, Chiriva-Internati M, Malesci A, Laghi L. Evolving notions on immune response in colorectal cancer and their implications for biomarker development. Inflamm Res 2018; 67:375-389. [PMID: 29322204 DOI: 10.1007/s00011-017-1128-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 12/27/2017] [Accepted: 12/29/2017] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION Colorectal cancer (CRC) still represents the third most commonly diagnosed type of cancer in men and women worldwide. CRC is acknowledged as a heterogeneous disease that develops through a multi-step sequence of events driven by clonal selections; this observation is sustained by the fact that histologically similar tumors may have completely different outcomes, including a varied response to therapy. METHODS In "early" and "intermediate" stage of CRC (stages II and III, respectively) there is a compelling need for new biomarkers fit to assess the metastatic potential of their disease, selecting patients with aggressive disease that might benefit from adjuvant and targeted therapies. Therefore, we review the actual notions on immune response in colorectal cancer and their implications for biomarker development. RESULTS The recognition of the key role of immune cells in human cancer progression has recently drawn attention on the tumor immune microenvironment, as a source of new indicators of tumor outcome and response to therapy. Thus, beside consolidated histopathological biomarkers, immune endpoints are now emerging as potential biomarkers. CONCLUSIONS The introduction of immune signatures and cellular and molecular components of the immune system as biomarkers is particularly important considering the increasing use of immune-based cancer therapies as therapeutic strategies for cancer patients.
Collapse
Affiliation(s)
- Fabio Grizzi
- Department of Immunology and Inflammation, Humanitas Clinical and Research Center, Via Manzoni 56, 20089, Rozzano, Milan, Italy.
| | - Gianluca Basso
- Laboratory of Molecular Gastroenterology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Elena Monica Borroni
- Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Tommaso Cavalleri
- Laboratory of Molecular Gastroenterology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Paolo Bianchi
- Laboratory of Molecular Gastroenterology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Sanja Stifter
- Department of Pathology, School of Medicine, University of Rijeka, Rijeka, Croatia
| | | | - Alberto Malesci
- Laboratory of Molecular Gastroenterology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
- Department of Gastroenterology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
- Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Luigi Laghi
- Laboratory of Molecular Gastroenterology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
- Department of Gastroenterology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
- Hereditary Cancer Genetics Clinic, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| |
Collapse
|
17
|
Li YP, Pang J, Gao S, Bai PY, Wang WD, Kong P, Cui Y. Role of CXCR4 and SDF1 as prognostic factors for survival and the association with clinicopathology in colorectal cancer: A systematic meta-analysis. Tumour Biol 2017. [PMID: 28621237 DOI: 10.1177/1010428317706206] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Yao-ping Li
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, China
- Affiliated Provincial Hospital of Shanxi Medical University, Taiyuan, China
| | - Jing Pang
- Affiliated Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Sheng Gao
- Affiliated Tumor Hospital of Shanxi Medical University, Taiyuan, China
| | - Peng-yu Bai
- Affiliated Tumor Hospital of Shanxi Medical University, Taiyuan, China
| | - Wen-da Wang
- Affiliated Tumor Hospital of Shanxi Medical University, Taiyuan, China
| | - Pengzhou Kong
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yongping Cui
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
18
|
A meta-analysis of CXCL12 expression for cancer prognosis. Br J Cancer 2017; 117:124-135. [PMID: 28535157 PMCID: PMC5520200 DOI: 10.1038/bjc.2017.134] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 04/08/2017] [Accepted: 04/24/2017] [Indexed: 02/07/2023] Open
Abstract
Background: CXCL12 (SDF1) is reported to promote cancer progression in several preclinical models and this is corroborated by the analysis of human tissue specimens. However, the relationship between CXCL12 expression and cancer survival has not been systematically assessed. Methods: We conducted a systematic review and meta-analysis of studies that evaluated the association between CXCL12 expression and cancer survival. Results: Thirty-eight studies inclusive of 5807 patients were included in the analysis of overall, recurrence-free or cancer-specific survival, the majority of which were retrospective. The pooled hazard ratios (HRs) for overall and recurrence-free survival in patients with high CXCL12 expression were 1.39 (95% CI: 1.17–1.65, P=0.0002) and 1.12 (95% CI: 0.82–1.53, P=0.48) respectively, but with significant heterogeneity between studies. On subgroup analysis by cancer type, high CXCL12 expression was associated with reduced overall survival in patients with oesophagogastric (HR 2.08; 95% CI: 1.31–3.33, P=0.002), pancreatic (HR 1.54; 95% CI: 1.21–1.97, P=0.0005) and lung cancer (HR 1.37; 95% CI: 1.08–1.75, P=0.01), whereas in breast cancer patients high CXCL12 expression conferred an overall survival advantage (HR 0.5; 95% CI: 0.38–0.66, P<0.00001). Conclusions: Determination of CXCL12 expression has the potential to be of use as a cancer biomarker and adds prognostic information in various cancer types. Prospective or prospective–retrospective analyses of CXCL12 expression in clearly defined cancer cohorts are now required to advance our understanding of the relationship between CXCL12 expression and cancer outcome.
Collapse
|
19
|
Circelli L, Sciammarella C, Guadagno E, Tafuto S, del Basso de Caro M, Botti G, Pezzullo L, Aria M, Ramundo V, Tatangelo F, Losito NS, Ieranò C, D'Alterio C, Izzo F, Ciliberto G, Colao A, Faggiano A, Scala S. CXCR4/CXCL12/CXCR7 axis is functional in neuroendocrine tumors and signals on mTOR. Oncotarget 2017; 7:18865-75. [PMID: 26934559 PMCID: PMC4951335 DOI: 10.18632/oncotarget.7738] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 01/06/2016] [Indexed: 12/11/2022] Open
Abstract
Objective To evaluate the possible crosstalk between C-X-C chemokine receptor 4 (CXCR4)/C-X-C motif chemokine 12 (CXCL12)/C-X-C chemokine receptor 7 (CXCR7) axis with the mammalian target of rapamycin (mTOR) pathway in neuroendocrine tumors (NETs). Methods Sixty-one human NETs were included into the study. CXCR4/CXCL12/CXCR7 axis and mTOR pathway were assessed by qRT-PCR and immunohistochemistry (IHC). The effect of mTOR inhibitor, RAD001, was evaluated on CXCR4 pathway through proliferation and p-Erk and p-AKT induction. Results: CXCR4/CXCL12/CXCR7 axis and p-mTOR were found to be active and correlated with grading, Ki67 index and tumor stage. mTOR pathway activation significantly correlated with poor prognosis. In human NET cells, CXCL12 induced mTOR signalling while AMD3100 (CXCR4-antagonist) impaired it. The mTOR-antagonist, RAD001, impaired the CXCL12-dependent induction of CXCR4 downstream effectors. Combination of AMD3100 and RAD001 potentiate cell growth inhibition. Conclusions CXCR4/CXCL12/CXCR7 axis is active in NETs and signals on mTOR. CXCR4 might be considered a prognostic factor in NETs. Combined treatment with AMD3100 and RAD001 may provide clinical benefits in NET patients with drug-resistant.
Collapse
Affiliation(s)
- Luisa Circelli
- Molecolar Immunology and Immuneregulation, Istituto Nazionale per lo Studio e la Cura dei Tumori - IRCCS Naples "Fondazione G. Pascale", Naples, Italy
| | - Concetta Sciammarella
- Departments of Clinical Medicine and Surgery, "Federico II" University of Naples, Italy
| | - Elia Guadagno
- Advanced Biomedical Sciences, Division of Pathology, "Federico II" University of Naples, Italy
| | - Salvatore Tafuto
- Abdominal Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori - IRCCS Naples "Fondazione G. Pascale", Naples, Italy
| | | | - Giovanni Botti
- Molecolar Immunology and Immuneregulation, Istituto Nazionale per lo Studio e la Cura dei Tumori - IRCCS Naples "Fondazione G. Pascale", Naples, Italy
| | - Luciano Pezzullo
- Thyroid and Parathyroid Surgery Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori - IRCCS Naples "Fondazione G. Pascale", Naples, Italy
| | - Massimo Aria
- Economics and Statistics, "Federico II" University of Naples, Naples, Italy
| | - Valeria Ramundo
- Departments of Clinical Medicine and Surgery, "Federico II" University of Naples, Italy
| | - Fabiana Tatangelo
- Pathology, Istituto Nazionale per lo Studio e la Cura dei Tumori - IRCCS Naples "Fondazione G. Pascale", Naples, Italy
| | - Nunzia Simona Losito
- Pathology, Istituto Nazionale per lo Studio e la Cura dei Tumori - IRCCS Naples "Fondazione G. Pascale", Naples, Italy
| | - Caterina Ieranò
- Molecolar Immunology and Immuneregulation, Istituto Nazionale per lo Studio e la Cura dei Tumori - IRCCS Naples "Fondazione G. Pascale", Naples, Italy
| | - Crescenzo D'Alterio
- Molecolar Immunology and Immuneregulation, Istituto Nazionale per lo Studio e la Cura dei Tumori - IRCCS Naples "Fondazione G. Pascale", Naples, Italy
| | - Francesco Izzo
- Abdominal Surgery, Istituto Nazionale per lo Studio e la Cura dei Tumori - IRCCS Naples "Fondazione G. Pascale", Naples, Italy
| | - Gennaro Ciliberto
- Scientific Directorate, Istituto Nazionale per lo Studio e la Cura dei Tumori - IRCCS Naples "Fondazione G. Pascale", Naples, Italy
| | - Annamaria Colao
- Departments of Clinical Medicine and Surgery, "Federico II" University of Naples, Italy
| | - Antongiulio Faggiano
- Thyroid and Parathyroid Surgery Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori - IRCCS Naples "Fondazione G. Pascale", Naples, Italy
| | - Stefania Scala
- Molecolar Immunology and Immuneregulation, Istituto Nazionale per lo Studio e la Cura dei Tumori - IRCCS Naples "Fondazione G. Pascale", Naples, Italy
| |
Collapse
|
20
|
D'Alterio C, Nasti G, Polimeno M, Ottaiano A, Conson M, Circelli L, Botti G, Scognamiglio G, Santagata S, De Divitiis C, Nappi A, Napolitano M, Tatangelo F, Pacelli R, Izzo F, Vuttariello E, Botti G, Scala S. CXCR4-CXCL12-CXCR7, TLR2-TLR4, and PD-1/PD-L1 in colorectal cancer liver metastases from neoadjuvant-treated patients. Oncoimmunology 2016; 5:e1254313. [PMID: 28123896 DOI: 10.1080/2162402x.2016.1254313] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 10/20/2016] [Accepted: 10/21/2016] [Indexed: 12/20/2022] Open
Abstract
A neoadjuvant clinical trial was previously conducted in patients with resectable colorectal cancer liver metastases (CRLM). At a median follow up of 28 months, 20/33 patients were dead of disease, 8 were alive with disease and 5 were alive with no evidence of disease. To shed further insight into biological features accounting for different outcomes, the expression of CXCR4-CXCL12-CXCR7, TLR2-TLR4, and the programmed death receptor-1 (PD-1)/programmed death-1 ligand (PD-L1) was evaluated in excised liver metastases. Expression profiles were assessed through qPCR in metastatic and unaffected liver tissue of 33 CRLM neoadjuvant-treated patients. CXCR4 and CXCR7, TLR2/TLR4, and PD-1/PD-L1 mRNA were significantly overexpressed in metastatic compared to unaffected liver tissues. CXCR4 protein was negative/low in 10/31, and high in 21/31, CXCR7 was negative/low in 16/31 and high in 15/31, CXCL12 was negative/low in 14/31 and high in 17/31 CRLM. PD-1 was negative in 19/30 and positive in 11/30, PD-L1 was negative/low in 24/30 and high in 6/30 CRLM. Stromal PD-L1 expression, affected the progression-free survival (PFS) in the CRLM population. Patients overexpressing CXCR4 experienced a worse PFS and cancer specific survival (CSS) (p = 0.001 and p = 0.0008); in these patients, KRAS mutation identified a subgroup with a significantly worse CSS (p < 0.01). Thus, CXCR4 and PD-L1 expression discriminate patients with the worse PFS within the CRLM evaluated patients. Within the CXCR4 high expressing patients carrying Mut-KRAS in CRLM identifies the worst prognostic group. Thus, CXCR4 targeting plus anti-PD-1 therapy should be explored to improve the prognosis of Mut-KRAS-high CXCR4-CRLMs.
Collapse
Affiliation(s)
- Crescenzo D'Alterio
- Functional Genomics Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS , Napoli, Italy
| | - Guglielmo Nasti
- Gastrointestinal Medical Oncology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS , Napoli, Italy
| | - Marianeve Polimeno
- Functional Genomics Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS , Napoli, Italy
| | - Alessandro Ottaiano
- Gastrointestinal Medical Oncology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS , Napoli, Italy
| | - Manuel Conson
- Department of Advanced Biomedical Sciences, Federico II University School of Medicine , Naples, Italy
| | - Luisa Circelli
- Functional Genomics Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS , Napoli, Italy
| | - Giovanni Botti
- Functional Genomics Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS , Napoli, Italy
| | - Giosuè Scognamiglio
- Pathology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS , Napoli, Italy
| | - Sara Santagata
- Functional Genomics Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS , Napoli, Italy
| | - Chiara De Divitiis
- Gastrointestinal Medical Oncology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS , Napoli, Italy
| | - Anna Nappi
- Gastrointestinal Medical Oncology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS , Napoli, Italy
| | - Maria Napolitano
- Functional Genomics Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS , Napoli, Italy
| | - Fabiana Tatangelo
- Pathology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS , Napoli, Italy
| | - Roberto Pacelli
- Department of Advanced Biomedical Sciences, Federico II University School of Medicine , Naples, Italy
| | - Francesco Izzo
- Hepatobiliary Surgery Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS , Napoli, Italy
| | - Emilia Vuttariello
- Functional Genomics Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS , Napoli, Italy
| | - Gerardo Botti
- Department of Advanced Biomedical Sciences, Federico II University School of Medicine , Naples, Italy
| | - Stefania Scala
- Functional Genomics Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" - IRCCS , Napoli, Italy
| |
Collapse
|
21
|
Di Maro S, Trotta AM, Brancaccio D, Di Leva FS, La Pietra V, Ieranò C, Napolitano M, Portella L, D'Alterio C, Siciliano RA, Sementa D, Tomassi S, Carotenuto A, Novellino E, Scala S, Marinelli L. Exploring the N-Terminal Region of C-X-C Motif Chemokine 12 (CXCL12): Identification of Plasma-Stable Cyclic Peptides As Novel, Potent C-X-C Chemokine Receptor Type 4 (CXCR4) Antagonists. J Med Chem 2016; 59:8369-80. [PMID: 27571038 DOI: 10.1021/acs.jmedchem.6b00695] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
We previously reported the discovery of a CXCL12-mimetic cyclic peptide (2) as a selective CXCR4 antagonist showing promising in vitro and in vivo anticancer activity. However, further development of this peptide was hampered by its degradation in biological fluids as well as by its low micromolar affinity for the receptor. Herein, extensive chemical modifications led to the development of a new analogue (10) with enhanced potency, specificity, and plasma stability. A combined approach of Ala-amino acid scan, NMR, and molecular modeling unraveled the reasons behind the improved binding properties of 10 vs 2. Biological investigations on leukemia (CEM) and colon (HT29 and HCT116) cancer cell lines showed that 10 is able to impair CXCL12-mediated cell migration, ERK-phosphorylation, and CXCR4 internalization. These outcomes might pave the way for the future preclinical development of 10 in CXCR4 overexpressing leukemia and colon cancer.
Collapse
Affiliation(s)
- Salvatore Di Maro
- DiSTABiF, Second University of Naples , Via Vivaldi 43, 81100 Caserta, Italy
| | - Anna Maria Trotta
- Genomica Funzionale, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione "Giovanni Pascale", IRCCS-ITALY , Via M. Semmola, 80131 Naples, Italy
| | - Diego Brancaccio
- Dipartimento di Farmacia, Università degli Studi di Napoli "Federico II" , via D. Montesano 49, 80131 Naples, Italy.,Laboratory of Food Chemistry, Dipartimento di Agraria (QuaSic.A.Tec.), Università Mediterranea di Reggio Calabria , Reggio Calabria, loc. Feo di Vito, 89122 Reggio Calabria, Italy
| | - Francesco Saverio Di Leva
- Dipartimento di Farmacia, Università degli Studi di Napoli "Federico II" , via D. Montesano 49, 80131 Naples, Italy
| | - Valeria La Pietra
- Dipartimento di Farmacia, Università degli Studi di Napoli "Federico II" , via D. Montesano 49, 80131 Naples, Italy
| | - Caterina Ieranò
- Genomica Funzionale, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione "Giovanni Pascale", IRCCS-ITALY , Via M. Semmola, 80131 Naples, Italy
| | - Maria Napolitano
- Genomica Funzionale, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione "Giovanni Pascale", IRCCS-ITALY , Via M. Semmola, 80131 Naples, Italy
| | - Luigi Portella
- Genomica Funzionale, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione "Giovanni Pascale", IRCCS-ITALY , Via M. Semmola, 80131 Naples, Italy
| | - Crescenzo D'Alterio
- Genomica Funzionale, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione "Giovanni Pascale", IRCCS-ITALY , Via M. Semmola, 80131 Naples, Italy
| | - Rosa Anna Siciliano
- Istituto di Scienze dell'Alimentazione, CNR , Via Roma 64, 83100 Avellino, Italy
| | - Deborah Sementa
- Dipartimento di Farmacia, Università degli Studi di Napoli "Federico II" , via D. Montesano 49, 80131 Naples, Italy
| | - Stefano Tomassi
- Dipartimento di Farmacia, Università degli Studi di Napoli "Federico II" , via D. Montesano 49, 80131 Naples, Italy
| | - Alfonso Carotenuto
- Dipartimento di Farmacia, Università degli Studi di Napoli "Federico II" , via D. Montesano 49, 80131 Naples, Italy
| | - Ettore Novellino
- Dipartimento di Farmacia, Università degli Studi di Napoli "Federico II" , via D. Montesano 49, 80131 Naples, Italy
| | - Stefania Scala
- Genomica Funzionale, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione "Giovanni Pascale", IRCCS-ITALY , Via M. Semmola, 80131 Naples, Italy
| | - Luciana Marinelli
- Dipartimento di Farmacia, Università degli Studi di Napoli "Federico II" , via D. Montesano 49, 80131 Naples, Italy
| |
Collapse
|
22
|
CXCR4 over-expression and survival in cancer: a system review and meta-analysis. Oncotarget 2016; 6:5022-40. [PMID: 25669980 PMCID: PMC4467131 DOI: 10.18632/oncotarget.3217] [Citation(s) in RCA: 187] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Accepted: 12/28/2014] [Indexed: 01/11/2023] Open
Abstract
C-X-C chemokine receptor 4 (CXCR4) is frequently over-expressed in various types of cancer; many agents against CXCR4 are in clinical development currently despite variable data for the prognostic impact of CXCR4 expression. Here eighty-five studies with a total of 11,032 subjects were included to explore the association between CXCR4 and progression-free survival (PFS) or overall survival (OS) in subjects with cancer. Pooled analysis shows that CXCR4 over-expression is significantly associated with poorer PFS (HR 2.04; 95% CI, 1.72-2.42) and OS (HR=1.94; 95% CI, 1.71-2.20) irrespective of cancer types. Subgroup analysis indicates significant association between CXCR4 and shorter PFS in hematological malignancy, breast cancer, colorectal cancer, esophageal cancer, renal cancer, gynecologic cancer, pancreatic cancer and liver cancer; the prognostic effects remained consistent across age, risk of bias, levels of adjustment, median follow-up period, geographical area, detection methods, publication year and size of studies. CXCR4 over-expression predicts unfavorable OS in hematological malignancy, breast cancer, colorectal cancer, esophageal cancer, head and neck cancer, renal cancer, lung cancer, gynecologic cancer, liver cancer, prostate cancer and gallbladder cancer; these effects were independence of age, levels of adjustment, publication year, detection methods and follow-up period. In conclusion, CXCR4 over-expression is associated with poor prognosis in cancer.
Collapse
|
23
|
Stanisavljević L, Aßmus J, Storli KE, Leh SM, Dahl O, Myklebust MP. CXCR4, CXCL12 and the relative CXCL12-CXCR4 expression as prognostic factors in colon cancer. Tumour Biol 2015; 37:7441-52. [PMID: 26678887 DOI: 10.1007/s13277-015-4591-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 12/03/2015] [Indexed: 01/06/2023] Open
Abstract
The CXCL12-CXCR4 axis is proposed to mediate metastasis formation. In this study, we examined CXCL12, CXCR4 and the relative CXCL12-CXCR4 expression as prognostic factors in two cohorts of colon cancer patients. Immunohistochemistry (IHC) and in situ hybridization (ISH) were used to study CXCR4, CXCL12 and relative CXCL12-CXCR4 expression in tissue microarrays. Our study included totally 596 patients, 290 in cohort 1 and 306 in cohort 2. For tumour, node, metastasis (TNM) stage III, low nuclear expression of CXCR4 was a positive prognostic factor for 5-year disease-free survival (DFS) in cohort 1 (P = 0.007) and cohort 2 (P = 0.023). In multivariate analysis for stage III, nuclear expression of CXCR4 in cohort 1 was confirmed as a prognostic factor for DFS (hazard ratio (HR), 0.27; 95 % CI, 0.09 to 0.77). For TNM stage III, high cytoplasmic expression of CXCL12 was associated with better 5-year DFS in both cohorts (P = 0.006 and P = 0.006, respectively). We further validated the positive prognostic value of CXCL12 expression for 5-year DFS in stage III with ISH (P = 0.022). For TNM stage III, the relative CXCL12-CXCR4 expression (CXCL12 > CXCR4 vs CXCL12 = CXCR4 vs CXCL12 < CXCR4) was a prognostic factor for 5-year DFS in cohort 1 (92 % vs 46 % vs 31 %, respectively; P < 0.001) and cohort 2 (92 % vs 66 % vs 30 %, respectively; P = 0.006). In conclusion, CXCL12 and relative CXCL12-CXCR4 expression are independent prognostic factors for 5-year DFS in TNM stage III colon cancer.
Collapse
Affiliation(s)
- Luka Stanisavljević
- Section of Oncology, Department of Clinical Science, University of Bergen, Post box 7804, 5020, Bergen, Norway.
| | - Jörg Aßmus
- Centre for Clinical Research, Haukeland University Hospital, Bergen, Norway
| | | | - Sabine Maria Leh
- Department of Pathology, Haukeland University Hospital, Bergen, Norway.,Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Olav Dahl
- Section of Oncology, Department of Clinical Science, University of Bergen, Post box 7804, 5020, Bergen, Norway.,Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway
| | | |
Collapse
|
24
|
Brown GT, Cash B, Alnabulsi A, Samuel LM, Murray GI. The expression and prognostic significance of bcl-2-associated transcription factor 1 in rectal cancer following neoadjuvant therapy. Histopathology 2015; 68:556-66. [PMID: 26183150 DOI: 10.1111/his.12780] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 07/12/2015] [Indexed: 12/14/2022]
Abstract
AIMS bcl-2-associated transcription factor 1 (BCLAF1) is a nuclear protein that binds to bcl-related proteins and can induce apoptosis and autophagy. The aim of this study was to investigate the expression of BCLAF1 in a series of rectal cancers following neoadjuvant therapy. METHODS AND RESULTS Immunohistochemistry was performed on a post-neoadjuvant therapy rectal cancer tissue microarray. It contained rectal cancers (n = 248), lymph node metastases (n = 76), and non-neoplastic rectal mucosal samples (n = 73). A monoclonal antibody against BCLAF1 that we have developed was used. Non-neoplastic rectal epithelium showed nuclear localization of BCLAF1 in both crypt and surface epithelial cells, whereas rectal cancers showed both nuclear and cytoplasmic BCLAF1 expression. Most rectal cancers showed moderate or strong nuclear immunoreactivity, but showed weak cytoplasmic immunoreactivity. Cytoplasmic BCLAF1 expression was increased in primary rectal cancers as compared with non-neoplastic rectal mucosa (P = 0.008). Negative and weak nuclear BCLAF1 expression was associated with a poor prognosis [hazard ratio (HR) 0.502, 95% confidence interval (CI) 0.269-0.939, χ(2) = 4.876, P = 0.027]. Nuclear BCLAF1 expression was independently prognostic in a multivariate model (HR 0.431, 95% CI 0.221-0.840, P = 0.013). CONCLUSIONS This study has shown that both cytoplasmic BCLAF1 expression and nuclear BCLAF1 expression are increased in post-neoadjuvant therapy rectal cancer, and that negative and weak nuclear BCLAF1 expression are independently associated with a poor prognosis.
Collapse
Affiliation(s)
- Gordon T Brown
- Pathology, Division of Applied Medicine, School of Medicine and Dentistry, University of Aberdeen, Aberdeen, UK
| | - Beatriz Cash
- Vertebrate Antibodies, Tillydrone Avenue, Aberdeen, UK
| | | | - Leslie M Samuel
- Department of Clinical Oncology, Aberdeen Royal Infirmary, NHS Grampian, Aberdeen, UK
| | - Graeme I Murray
- Pathology, Division of Applied Medicine, School of Medicine and Dentistry, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
25
|
CXCL12 and CXCR4, but not CXCR7, are primarily expressed by the stroma in head and neck squamous cell carcinoma. Pathology 2015; 47:45-50. [PMID: 25474514 DOI: 10.1097/pat.0000000000000191] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The CXCL12/CXCR4 axis is involved in numerous models of metastatic dissemination, including head and neck squamous cell carcinoma (HNSCC). We assessed the relative expressions of CXCL12, CXCR4 and CXCR7 in the stroma and the tumour of HNSCC, and evaluated the methylation status of the CXCL12 promoter.Snap-frozen, HPV negative HNSCC samples were micro-dissected to isolate the tumoural and stromal compartments. The expression levels of CXCL12, CXCR4 and CXCR7 were assessed by qRT-PCR, and the methylation level of the CXCL12 promoter was evaluated by pyrosequencing.In total, 23 matched tumour/stroma samples were analysed. Higher expressions of CXCR4 and CXCL12 were observed in the stroma (p = 0.012 and p < 0.0001, respectively). No significant difference in expression was observed for CXCR7. A high methylation level (>40%) of the CXCL12 promoter was observed in only a few tumoural samples (5/23) and was associated with a lower expression of the gene (p = 0.03).Stromal cells, rather than the tumour itself, are mainly responsible for the expression of both CXCL12 and CXCR4 expression in HNSCC. CXCR7 expression did not differ between the two compartments and was not related to CXCL12 or CXCR4 expression. Finally, the methylation of the CXCL12 promoter could only explain the low intra-tumoural expression of this gene in 20% of cases.
Collapse
|
26
|
Scala S. Molecular Pathways: Targeting the CXCR4–CXCL12 Axis—Untapped Potential in the Tumor Microenvironment. Clin Cancer Res 2015. [DOI: 10.1158/1078-0432.ccr-14-0914] [Citation(s) in RCA: 189] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
27
|
Stacer AC, Fenner J, Cavnar SP, Xiao A, Zhao S, Chang SL, Salomonnson A, Luker KE, Luker GD. Endothelial CXCR7 regulates breast cancer metastasis. Oncogene 2015; 35:1716-24. [PMID: 26119946 PMCID: PMC4486335 DOI: 10.1038/onc.2015.236] [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: 07/21/2014] [Revised: 04/29/2015] [Accepted: 05/01/2015] [Indexed: 02/08/2023]
Abstract
Atypical chemokine receptor CXCR7 (ACKR3) functions as a scavenger receptor for chemokine CXCL12, a molecule that promotes multiple steps in tumor growth and metastasis in breast cancer and multiple other malignancies. While normal vascular endothelium expresses low levels of CXCR7, marked upregulation of CXCR7 occurs in tumor vasculature in breast cancer and other tumors. To investigate effects of endothelial CXCR7 in breast cancer, we conditionally deleted this receptor from vascular endothelium of adult mice, generating CXCR7ΔEND/ΔEND animals. CXCR7ΔEND/ΔEND mice appeared phenotypically normal, although these animals exhibited a modest 35 ± 3% increase in plasma CXCL12 as compared with control. Using two different syngeneic, orthotopic tumor implant models of breast cancer, we discovered that CXCR7ΔEND/ΔEND mice had significantly greater local recurrence of cancer following resection, elevated numbers of circulating tumor cells, and more spontaneous metastases. CXCR7ΔEND/ΔEND mice also showed greater experimental metastases following intracardiac injection of cancer cells. These results establish that endothelial CXCR7 limits breast cancer metastasis at multiple steps in the metastatic cascade, advancing understanding of CXCL12 pathways in tumor environments and informing ongoing drug development targeting CXCR7 in cancer.
Collapse
Affiliation(s)
- A C Stacer
- University of Michigan Center for Molecular Imaging, Department of Radiology, University of Michigan Medical School and College of Engineering, Ann Arbor, MI, USA
| | - J Fenner
- University of Michigan Center for Molecular Imaging, Department of Radiology, University of Michigan Medical School and College of Engineering, Ann Arbor, MI, USA
| | - S P Cavnar
- Department of Biomedical Engineering, University of Michigan Medical School and College of Engineering, Ann Arbor, MI, USA
| | - A Xiao
- University of Michigan Center for Molecular Imaging, Department of Radiology, University of Michigan Medical School and College of Engineering, Ann Arbor, MI, USA
| | - S Zhao
- Department of Radiation Oncology, University of Michigan Medical School and College of Engineering, Ann Arbor, MI, USA
| | - S L Chang
- Depatment of Chemical Engineering, University of Michigan Medical School and College of Engineering, Ann Arbor, MI, USA
| | - A Salomonnson
- University of Michigan Center for Molecular Imaging, Department of Radiology, University of Michigan Medical School and College of Engineering, Ann Arbor, MI, USA
| | - K E Luker
- University of Michigan Center for Molecular Imaging, Department of Radiology, University of Michigan Medical School and College of Engineering, Ann Arbor, MI, USA
| | - G D Luker
- University of Michigan Center for Molecular Imaging, Department of Radiology, University of Michigan Medical School and College of Engineering, Ann Arbor, MI, USA.,Department of Biomedical Engineering, University of Michigan Medical School and College of Engineering, Ann Arbor, MI, USA.,Department of Microbiology and Immunology, University of Michigan Medical School and College of Engineering, Ann Arbor, MI, USA
| |
Collapse
|
28
|
Yun HJ, Ryu H, Choi YS, Song IC, Jo DY, Kim S, Lee HJ. C-X-C motif receptor 7 in gastrointestinal cancer. Oncol Lett 2015; 10:1227-1232. [PMID: 26622655 DOI: 10.3892/ol.2015.3407] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 05/22/2015] [Indexed: 02/06/2023] Open
Abstract
Chemokine receptors are key mediators of normal physiology and numerous pathological conditions, including inflammation and cancer. This receptor family is an emerging target for anticancer drug development. C-X-C motif receptor 7 (CXCR7) is an atypical chemokine receptor that was first cloned from a canine cDNA library as an orphan receptor and was initially named receptor dog cDNA 1 (RDC1). Shortly after demonstrating that RDC1 binds with its ligand, stromal cell-derived factor-1α and interferon-inducible T-cell α chemoattractant, RDC1 was officially deorphanized and renamed CXCR7, as the seventh receptor in the CXC class of the chemokine receptor family. Recent accumulating evidence has demonstrated that CXCR7 expression is augmented in the majority of tumor cells compared with their normal counterparts and is involved in cell proliferation, survival, migration, invasion and angiogenesis during the initiation and progression of breast, lung and prostate cancer. In the present review, the expression and role of CXCR7, as well as its clinical relevance in cancer of the gastrointestinal system, were investigated. In addition, the potential of this chemokine receptor as a therapeutic target in the treatment of gastrointestinal cancer was discussed.
Collapse
Affiliation(s)
- Hwan-Jung Yun
- Division of Hematology/Oncology, Department of Internal Medicine, Chungnam National University Hospital, Daejeon 301-721, Republic of Korea ; Cancer Research Institute, Chungnam National University School of Medicine, Daejeon 301-747, Republic of Korea
| | - Hyewon Ryu
- Division of Hematology/Oncology, Department of Internal Medicine, Chungnam National University Hospital, Daejeon 301-721, Republic of Korea
| | - Yoon Seok Choi
- Division of Hematology/Oncology, Department of Internal Medicine, Chungnam National University Hospital, Daejeon 301-721, Republic of Korea
| | - Ik-Chan Song
- Division of Hematology/Oncology, Department of Internal Medicine, Chungnam National University Hospital, Daejeon 301-721, Republic of Korea
| | - Deog-Yeon Jo
- Division of Hematology/Oncology, Department of Internal Medicine, Chungnam National University Hospital, Daejeon 301-721, Republic of Korea ; Cancer Research Institute, Chungnam National University School of Medicine, Daejeon 301-747, Republic of Korea
| | - Samyong Kim
- Division of Hematology/Oncology, Department of Internal Medicine, Chungnam National University Hospital, Daejeon 301-721, Republic of Korea ; Cancer Research Institute, Chungnam National University School of Medicine, Daejeon 301-747, Republic of Korea
| | - Hyo Jin Lee
- Division of Hematology/Oncology, Department of Internal Medicine, Chungnam National University Hospital, Daejeon 301-721, Republic of Korea ; Cancer Research Institute, Chungnam National University School of Medicine, Daejeon 301-747, Republic of Korea
| |
Collapse
|
29
|
Rybinski B, Franco-Barraza J, Cukierman E. The wound healing, chronic fibrosis, and cancer progression triad. Physiol Genomics 2014; 46:223-44. [PMID: 24520152 PMCID: PMC4035661 DOI: 10.1152/physiolgenomics.00158.2013] [Citation(s) in RCA: 166] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 02/04/2014] [Indexed: 02/07/2023] Open
Abstract
For decades tumors have been recognized as "wounds that do not heal." Besides the commonalities that tumors and wounded tissues share, the process of wound healing also portrays similar characteristics with chronic fibrosis. In this review, we suggest a tight interrelationship, which is governed as a concurrence of cellular and microenvironmental reactivity among wound healing, chronic fibrosis, and cancer development/progression (i.e., the WHFC triad). It is clear that the same cell types, as well as soluble and matrix elements that drive wound healing (including regeneration) via distinct signaling pathways, also fuel chronic fibrosis and tumor progression. Hence, here we review the relationship between fibrosis and cancer through the lens of wound healing.
Collapse
Affiliation(s)
- Brad Rybinski
- Cancer Biology Program, Fox Chase Cancer Center/Temple Health, Philadelphia, Pennsylvania
| | | | | |
Collapse
|