201
|
Yu J, Xie M, Ge S, Chai P, Zhou Y, Ruan J. Hierarchical Clustering of Cutaneous Melanoma Based on Immunogenomic Profiling. Front Oncol 2020; 10:580029. [PMID: 33330057 PMCID: PMC7735560 DOI: 10.3389/fonc.2020.580029] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 10/26/2020] [Indexed: 12/23/2022] Open
Abstract
Cutaneous melanoma is an aggressive malignancy with high heterogeneity. Several studies have been performed to identify cutaneous melanoma subtypes based on genomic profiling. However, few classifications based on assessments of immune-associated genes have limited clinical implications for cutaneous melanoma. Using 470 cutaneous melanoma samples from The Cancer Genome Atlas (TCGA), we calculated the enrichment levels of 29 immune-associated gene sets in each sample and hierarchically clustered them into Immunity High (Immunity_H, n=323, 68.7%), Immunity Medium (Immunity_M, n=135, 28.7%), and Immunity Low (Immunity_L, n=12, 2.6%) based on the ssGSEA score. The ESTIMATE algorithm was used to calculate stromal scores (range: -1,800.51-1,901.99), immune scores (range: -1,476.28-3,780.33), estimate scores (range: -2,618.28-5,098.14) and tumor purity (range: 0.216-0.976) and they were significantly correlated with immune subtypes (Kruskal-Wallis test, P < 0.001). The Immunity_H group tended to have higher expression levels of HLA and immune checkpoint genes (Kruskal-Wallis test, P < 0.05). The Immunity_H group had the highest level of naïve B cells, resting dendritic cells, M1 macrophages, resting NK cells, plasma cells, CD4 memory activated T cells, CD8 T cells, follicular helper T cells and regulatory T cells, and the Immunity_L group had better overall survival. The GO terms identified in the Immunity_H group were mainly immune related. In conclusion, immune signature-associated cutaneous melanoma subtypes play a role in cutaneous melanoma prognosis stratification. The construction of immune signature-associated cutaneous melanoma subtypes predicted possible patient outcomes and provided possible immunotherapy candidates.
Collapse
Affiliation(s)
| | | | | | - Peiwei Chai
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Yixiong Zhou
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Jing Ruan
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| |
Collapse
|
202
|
Wan L, Tan N, Zhang N, Xie X. Establishment of an immune microenvironment-based prognostic predictive model for gastric cancer. Life Sci 2020; 261:118402. [DOI: 10.1016/j.lfs.2020.118402] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 09/02/2020] [Accepted: 09/07/2020] [Indexed: 02/08/2023]
|
203
|
Guo X, Li M. LINC01089 is a tumor-suppressive lncRNA in gastric cancer and it regulates miR-27a-3p/TET1 axis. Cancer Cell Int 2020; 20:507. [PMID: 33088215 PMCID: PMC7568383 DOI: 10.1186/s12935-020-01561-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 09/17/2020] [Indexed: 12/21/2022] Open
Abstract
Background Gastric cancer (GC) is one of the most common malignancies around the world. Recently, the role of long non-coding RNA (lncRNA) in cancer biology has become a hot research topic. This work aimed to explore the biological function and underlying mechanism of LINC01089 in GC. Methods Quantitative real-time polymerase chain reaction (qRT-PCR) was employed to investigate the expression of LINC01089 in GC tissues and cells. The relationship between the expression level of LINC01089 and the clinicopathological parameters of GC was assessed. Cell models of LINC01089 overexpression, LINC01089 knockdown, miR-27a-3p overexpression, and miR-27a-3p inhibition were established by transfection. CCK-8 assay, BrdU assay, and Transwell assay were utilized to investigate the malignant biological behaviors of GC cell lines after transfection. Dual luciferase activity reporter assay, Pearson’s correlation analysis, and Western blot were utilized to the regulatory relationships among LINC01089, miR-27a-3p and tet methylcytosine dioxygenase 1 (TET1). Result LINC01089 down-regulation was observed in GC tissues and cell lines. Low expression level of LINC01089 in GC tissues was markedly linked to larger tumor size, higher T stage, as well as lymphatic metastasis of the patients. Functional experiments implied that LINC01089 overexpression impeded the proliferation, migration, as well as invasion of GC cells, whereas LINC01089 knockdown promoted the above malignant phenotypes. Additionally, up-regulation of miR-27a-3p was also observed in GC tissues. Functional experiments also showed that, miR-27a-3p overexpression boosted the malignant biological behaviors of GC cells; on the contrast, these phenotypes were impeded by miR-27a-3p inhibition. Moreover, LINC01089 interacted with and repressed miR-27a-3p, and miR-27a-3p antagonized the impact of LINC01089 on GC cells. Additionally, TET1 was verified as a target gene of miR-27a-3p, and could be positively regulated by LINC01089. Conclusion LINC01089 impedes the proliferation, migration, and invasion of GC cells by adsorbing miR-27a-3p and up-regulating the expression of TET1.
Collapse
Affiliation(s)
- Xufeng Guo
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430000 Hubei China
| | - Ming Li
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Zhangzhidong Road, Wuchang District, Wuhan, 430000 Hubei China
| |
Collapse
|
204
|
Wang L, Huang L, Xi L, Zhang SC, Zhang JX. High expression of squamous cell carcinoma antigen in poorly differentiated adenocarcinoma of the stomach: A case report. World J Clin Cases 2020; 8:4572-4578. [PMID: 33083420 PMCID: PMC7559675 DOI: 10.12998/wjcc.v8.i19.4572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 08/11/2020] [Accepted: 09/05/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Squamous cell carcinoma antigen (SCCA) is regarded as a specific indicator of epithelial malignancies and is widely used in the diagnosis of squamous cell carcinoma (SCC). However, the expression of SCCA in gastric adenocarcinoma has not been studied in detail. CASE SUMMARY A 52-year-old man was admitted to our hospital for a 2.5 cm × 2.5 cm ulcer at the antrum-body junction with dull pain and fullness in the upper abdomen for 2 mo. His pre-surgery serological testing results showed 0.51 ng/mL SCCA (reference interval, < 1.5 ng/mL) and 9.9 ng/mL carcinoembryonic antigen (reference range, < 4.7 ng/mL). He underwent radical distal gastrectomy and Roux-en Y anastomosis and was diagnosed with poorly differentiated mucinous adenocarcinoma (Lauren classification: Diffuse) by pathological examination of the resected lesion. Immunohistochemistry showed that SCCA was highly expressed in the cytoplasm of cancer cells. After surgery, the patient received an S-1 adjuvant chemotherapy regimen for six cycles containing tegafur, gimeracil, and oteracil potassium. He showed no sign of recurrence or metastasis within 24-mo follow-up. CONCLUSION This is a frontal report of SCCA overexpression in poorly differentiated adenocarcinoma of the stomach.
Collapse
Affiliation(s)
- Lin Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Lei Huang
- Department of Laboratory Medicine, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Lei Xi
- Department of Pathology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Shi-Chang Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Jie-Xin Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| |
Collapse
|
205
|
Pan Y, Fang Y, Xie M, Liu Y, Yu T, Wu X, Xu T, Ma P, Shu Y. LINC00675 Suppresses Cell Proliferation and Migration via Downregulating the H3K4me2 Level at the SPRY4 Promoter in Gastric Cancer. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 22:766-778. [PMID: 33230474 PMCID: PMC7595884 DOI: 10.1016/j.omtn.2020.09.038] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 09/30/2020] [Indexed: 02/07/2023]
Abstract
Accumulating evidence indicates that long noncoding RNAs (lncRNAs) are dysregulated in diverse tumors and take a pivotal role in modulating biological processes. In our study, a decreased expression level of LINC00675 in gastric cancer (GC) was first determined by data from The Cancer Genome Atlas (TCGA) and was identified using specimens from GC patients. Then, in vitro and in vivo functional experiments elaborated that LINC00675 could suppress cell proliferation and migration in GC. Multiple differentially expressed genes (DEGs) in LINC00675-overexpressing cells were identified through RNA sequencing analysis. An RNA-binding protein immunoprecipitation (RIP) assay was conducted to reveal that LINC00675 competitively bound with lysine-specific demethylase 1 (LSD1). A coimmunoprecipitation (coIP) assay indicated that LINC00675 overexpression may strengthen the binding of LSD1 and H3K4me2, whereas the chromatin immunoprecipitation (ChIP) assay results verified lower expression of H3K4me2 at the sprouty homolog 4 (SPRY4) promoter region. Together, our research identified that LINC00675 was remarkably downregulated in GC tissues and cells relative to nontumor tissues and cells. LINC00675 could repress GC tumorigenesis and metastasis via competitively binding with LSD1 and intensifying the binding of LSD1 and its target H3K4me2. Importantly, this contributed to attenuated binding of H3K4me2 at the promoter region of oncogene SPRY4 and suppressed SPRY4 transcription, thus suppressing GC cell proliferation and migration.
Collapse
Affiliation(s)
- Yutian Pan
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| | - Yuan Fang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| | - Mengyan Xie
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| | - Yu Liu
- Department of the Orthopaedics, RWTH Aachen University Clinic, Pauwelsstrasse 30, 52074 Aachen, Germany
| | - Tao Yu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| | - Xi Wu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| | - Tongpeng Xu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| | - Pei Ma
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| | - Yongqian Shu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China.,Department of Oncology, Affiliated Sir Run Hospital of Nanjing Medical University, Nanjing 211166, People's Republic of China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 211166, People's Republic of China
| |
Collapse
|
206
|
Shi XJ, Wei Y, Ji B. Systems Biology of Gastric Cancer: Perspectives on the Omics-Based Diagnosis and Treatment. Front Mol Biosci 2020; 7:203. [PMID: 33005629 PMCID: PMC7479200 DOI: 10.3389/fmolb.2020.00203] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/27/2020] [Indexed: 12/14/2022] Open
Abstract
Gastric cancer is the fifth most diagnosed cancer in the world, affecting more than a million people and causing nearly 783,000 deaths each year. The prognosis of advanced gastric cancer remains extremely poor despite the use of surgery and adjuvant therapy. Therefore, understanding the mechanism of gastric cancer development, and the discovery of novel diagnostic biomarkers and therapeutics are major goals in gastric cancer research. Here, we review recent progress in application of omics technologies in gastric cancer research, with special focus on the utilization of systems biology approaches to integrate multi-omics data. In addition, the association between gastrointestinal microbiota and gastric cancer are discussed, which may offer insights in exploring the novel microbiota-targeted therapeutics. Finally, the application of data-driven systems biology and machine learning approaches could provide a predictive understanding of gastric cancer, and pave the way to the development of novel biomarkers and rational design of cancer therapeutics.
Collapse
Affiliation(s)
- Xiao-Jing Shi
- Laboratory Animal Center, State Key Laboratory of Esophageal Cancer Prevention and Treatment, Academy of Medical Science, Zhengzhou University, Zhengzhou, China
| | - Yongjun Wei
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou, China
| | - Boyang Ji
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| |
Collapse
|
207
|
Predicting Peritoneal Dissemination of Gastric Cancer in the Era of Precision Medicine: Molecular Characterization and Biomarkers. Cancers (Basel) 2020; 12:cancers12082236. [PMID: 32785164 PMCID: PMC7547377 DOI: 10.3390/cancers12082236] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/29/2020] [Accepted: 08/05/2020] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer (GC) is a leading cause of worldwide cancer-related death. Being a highly heterogeneous disease, the current treatment of GC has been suboptimal due to the lack of subtype-dependent therapies. Peritoneal dissemination (PD) is a common pattern of GC metastasis associated with poor prognosis. Therefore, it is urgently necessary to identify patients at high risk of PD. PD is found to be associated with Lauren diffuse type GC. Molecular profiling of GC, especially diffuse type GC, has been utilized to identify molecular alterations and has given rise to various molecular classifications, shedding light on the underlying mechanism of PD and enabling identification of patients at higher PD risk. In addition, a series of diagnositc and prognostic biomarkers of PD from serum, peritoneal lavages and primary GCs have been reported. This comprehensive review summarizes findings on the multi-omic characteristics of diffuse type GC, the clinical significance of updating molecular classifications of GC in association with PD risk and research advances in PD-associated biomarkers.
Collapse
|
208
|
Li J, Zhang H, Liu M, Xiang Y, Li H, Huang F, Li H, Dai Z, Gu CJ, Liao X, Zhang T. miR‐133a‐3p/FOXP3 axis regulates cell proliferation and autophagy in gastric cancer. J Cell Biochem 2020; 121:3392-3405. [DOI: 10.1002/jcb.29613] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Accepted: 12/11/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Jia‐Peng Li
- College of Life and Health Sciences, Institute of Biology and MedicineWuhan University of Science and Technology Wuhan Hubei China
| | - Hui‐Min Zhang
- College of Life and Health Sciences, Institute of Biology and MedicineWuhan University of Science and Technology Wuhan Hubei China
| | - Mei‐Jun Liu
- College of Life and Health Sciences, Institute of Biology and MedicineWuhan University of Science and Technology Wuhan Hubei China
| | - Yuan Xiang
- College of Life and Health Sciences, Institute of Biology and MedicineWuhan University of Science and Technology Wuhan Hubei China
| | - Hui Li
- College of Life and Health Sciences, Institute of Biology and MedicineWuhan University of Science and Technology Wuhan Hubei China
| | - Feng Huang
- College of Life and Health Sciences, Institute of Biology and MedicineWuhan University of Science and Technology Wuhan Hubei China
| | - Han‐Han Li
- College of Life and Health Sciences, Institute of Biology and MedicineWuhan University of Science and Technology Wuhan Hubei China
| | - Zhou‐Tong Dai
- College of Life and Health Sciences, Institute of Biology and MedicineWuhan University of Science and Technology Wuhan Hubei China
| | - Chao Jiang Gu
- College of Life and Health Sciences, Institute of Biology and MedicineWuhan University of Science and Technology Wuhan Hubei China
| | - Xing‐Hua Liao
- College of Life and Health Sciences, Institute of Biology and MedicineWuhan University of Science and Technology Wuhan Hubei China
| | - Tong‐Cun Zhang
- College of Life and Health Sciences, Institute of Biology and MedicineWuhan University of Science and Technology Wuhan Hubei China
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin, College of BiotechnologyTianjin University of Science and Technology Tianjin China
| |
Collapse
|
209
|
Rodriquenz MG, Roviello G, D’Angelo A, Lavacchi D, Roviello F, Polom K. MSI and EBV Positive Gastric Cancer's Subgroups and Their Link With Novel Immunotherapy. J Clin Med 2020; 9:1427. [PMID: 32403403 PMCID: PMC7291039 DOI: 10.3390/jcm9051427] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/28/2020] [Accepted: 05/05/2020] [Indexed: 02/06/2023] Open
Abstract
Gastric cancers have been historically classified based on histomorphologic features. The Cancer Genome Atlas network reported the comprehensive identification of genetic alterations associated with gastric cancer, identifying four distinct subtypes- Epstein-Barr virus (EBV)-positive, microsatellite-unstable/instability (MSI), genomically stable and chromosomal instability. In particular, EBV-positive and MSI gastric cancers seem responsive to novel immunotherapies drugs. The aim of this review is to describe MSI and EBV positive gastric cancer's subgroups and their relationship with novel immunotherapy.
Collapse
Affiliation(s)
- Maria Grazia Rodriquenz
- Division of Medical Oncology, Department of Onco-Hematology, IRCCS-CROB, Referral Cancer Center of Basilicata, via Padre Pio 1, 85028 Rionero, Vulture (PZ), Italy;
| | - Giandomenico Roviello
- Department of Health Sciences, University of Florence, viale Pieraccini, 6, 50139 Florence, Italy;
| | - Alberto D’Angelo
- Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, UK;
| | - Daniele Lavacchi
- Azienda Ospedaliera Careggi University Hospital of Florence and University of Florence, 50134 Florence, Italy;
| | - Franco Roviello
- Department of Medical, Surgical and Neuro Sciences, Section of Surgery, Azienda Ospedaliera Universitaria Senese, University of Siena, 53100 Siena, Italy;
| | - Karol Polom
- Department of Surgical Oncology, Gdansk Medical University, 80-210 Gdansk, Poland
| |
Collapse
|
210
|
Outcomes of Radiotherapy for Mesenchymal and Non-Mesenchymal Subtypes of Gastric Cancer. Cancers (Basel) 2020; 12:cancers12040943. [PMID: 32290335 PMCID: PMC7226608 DOI: 10.3390/cancers12040943] [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: 03/25/2020] [Revised: 04/04/2020] [Accepted: 04/09/2020] [Indexed: 12/28/2022] Open
Abstract
Background: The purpose of this study was to evaluate the clinical outcomes following postoperative chemotherapy (XP) versus chemoradiotherapy (XP-RT) according to mesenchymal subtype based on RNA sequencing in gastric cancer (GC) in a cohort of the Adjuvant chemoRadioTherapy In Stomach Tumor (ARTIST) trial. Methods: Of the 458 patients enrolled in the ARTIST trial, formalin-fixed, paraffin-embedded (FFPE) specimens were available from 106 (23.1%) patients for RNA analysis. The mesenchymal subtype was classified according to a previously reported 71-gene MSS/EMT signature using the NanoString assay. Results: Of the 106 patients analyzed (50 in XP arm, 56 in XP-RT arm), 36 (34.0%) patients were categorized as mesenchymal subtype by NanoString assay. Recurrence-free survival (RFS, p = 0.009, hazard ratio (HR) = 2.11, 95% confidence interval (CI): 1.21-3.70) and overall survival (OS, p = 0.003, HR = 2.28, 95% CI: 1.31-3.96) were significantly lower in the mesenchymal subtype than in the non-mesenchymal subtype. In terms of post-operative radiotherapy (RT), mesenchymal subtype was not an independent variable to predict RFS or OS regardless to the assigned arm (XP with or without RT) in this patient cohort. However, there was a trend in the adjuvant XP arm, which showed higher OS than the XP-RT arm for the mesenchymal subtype and lower OS than the XP-RT arm for the non-mesenchymal subtype. Conclusions: We could not determine any significant differences between the mesenchymal and non-mesenchymal subtypes with respect to the effects of adjuvant XP with or without RT in gastric cancer following curative surgery.
Collapse
|
211
|
Lau HCH, Kranenburg O, Xiao H, Yu J. Organoid models of gastrointestinal cancers in basic and translational research. Nat Rev Gastroenterol Hepatol 2020; 17:203-222. [PMID: 32099092 DOI: 10.1038/s41575-019-0255-2] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/11/2019] [Indexed: 12/24/2022]
Abstract
Cancer is a major public health problem worldwide. Gastrointestinal cancers account for approximately one-third of the total global cancer incidence and mortality. Historically, the mechanisms of tumour initiation and progression in the gastrointestinal tract have been studied using cancer cell lines in vitro and animal models. Traditional cell culture methods are associated with a strong selection of aberrant genomic variants that no longer reflect the original tumours in terms of their (metastatic) behaviour or response to therapy. Organoid technology has emerged as a powerful alternative method for culturing gastrointestinal tumours and the corresponding normal tissues in a manner that preserves their genetic, phenotypic and behavioural traits. Importantly, accumulating evidence suggests that organoid cultures have great value in predicting the outcome of therapy in individual patients. Herein, we review the current literature on organoid models of the most common gastrointestinal cancers, including colorectal cancer, gastric cancer, oesophageal cancer, liver cancer and pancreatic cancer, and their value in modelling tumour initiation, metastatic progression and therapy response. We also explore the limitations of current organoid models and discuss how they could be improved to maximally benefit basic and translational research in the future, especially in the fields of drug discovery and personalized medicine.
Collapse
Affiliation(s)
- Harry Cheuk Hay Lau
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Onno Kranenburg
- UMC Utrecht Cancer Center, Utrecht Platform for Organoid Technology, Utrecht University, Utrecht, Netherlands
| | - Haipeng Xiao
- Department of Endocrinology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jun Yu
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Shatin, Hong Kong.
| |
Collapse
|
212
|
Du J, Liang Y, Li J, Zhao JM, Wang ZN, Lin XY. Gastric Cancer Cell-Derived Exosomal microRNA-23a Promotes Angiogenesis by Targeting PTEN. Front Oncol 2020; 10:326. [PMID: 32232005 PMCID: PMC7082307 DOI: 10.3389/fonc.2020.00326] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 02/25/2020] [Indexed: 12/24/2022] Open
Abstract
Hypoxia-exposed lung cancer-released exosomal microRNA-23a (miR-23a) has been shown to enhance angiogenesis as well as vascular permeability, contributing to the close correlation between exosomal miR-23a and tumorigenesis. The current study aimed to investigate whether gastric cancer (GC) cell-derived exosomal miR-23a could induce angiogenesis and to elucidate the potential mechanisms associated with the process. Differentially expressed miRNAs in GC were initially screened from the Gene Expression Omnibus database. Target genes were selected following miRNA-mRNA prediction and subsequently verified by dual luciferase reporter assay. RT-qPCR was conducted to detect miR-23a and PTEN expression in GC tissues, cells and exosomes. Human umbilical venous endothelial cells (HUVECs) were co-cultured with GC cell-derived exosomes to assess the angiogenesis mediated by exosomes in vitro. Additionally, PTEN was overexpressed in HUVECs to analyze the mechanism by which miR-23a regulates angiogenesis. miR-23a was highly expressed in GC tissues and cells and GC cell-derived exosomes. Angiogenesis was promoted by the co-culture of HUVECs and GC cells-derived exosomes, as evidenced by the increased expression of VEGF but decreased expression of TSP-1. PTEN was targeted by miR-23a and was lowly expressed in GC tissues. In a co-culture system, miR-23a carried by GC cells-derived exosomes promoted angiogenesis via the repression of PTEN. Collectively, GC cell-derived exosomal miR-23a could promote angiogenesis and provide blood supply for growth of GC cells. This study contributes to advancement of miRNA-targeted therapeutics.
Collapse
Affiliation(s)
- Jiang Du
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Science, China Medical University, Shenyang, China
| | - Yuan Liang
- Medical Oncology Department of Thoracic Cancer (2), Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Ji Li
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Science, China Medical University, Shenyang, China
| | - Jin-Ming Zhao
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Science, China Medical University, Shenyang, China
| | - Zhen-Ning Wang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Xu-Yong Lin
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Science, China Medical University, Shenyang, China
| |
Collapse
|
213
|
Yang J, Lian Y, Yang R, Lian Y, Wu J, Liu J, Wang K, Xu H. Upregulation of lncRNA LINC00460 Facilitates GC Progression through Epigenetically Silencing CCNG2 by EZH2/LSD1 and Indicates Poor Outcomes. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 19:1164-1175. [PMID: 32059342 PMCID: PMC7016164 DOI: 10.1016/j.omtn.2019.12.041] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 12/11/2019] [Accepted: 12/29/2019] [Indexed: 01/24/2023]
Abstract
Non-protein-coding functional elements in the human genome in the postgenomic biology field have been drawing great attention in recent years. Thousands of long non-coding RNAs (lncRNAs) have been found to be expressed in various tumors. Yet only a small proportion of these lncRNAs have been well characterized. We have demonstrated that LINC00460 could affect cell proliferation through epigenetic regulation of KLF2 and CUL4A in human colorectal cancer. However, the clinical significance and biological role of LINC00460 in gastric cancer (GC) remain largely unknown. In this research, we discovered that LINC00460 is remarkably upregulated in GC tissues compared to the non-tumor tissues. Additionally, LINC00460 served as an independent prognostic marker in GC. Functionally, proliferation of GC cells could be regulated by LINC00460 both in vitro and in vivo. RNA sequencing (RNA-seq) analysis for the whole transcriptome indicated that LINC00460 may serve as a key regulatory factor in the tumorigenesis of GC. What's more, the biological function of LINC00460 was mediated, to certain extent, by the direct interaction with enhancer of zeste homolog 2 (EZH2) and lysine (K)-specific demethylase 1A (LSD1) proteins. Further analyses indicated that LINC00460 promoted GC proliferation at least partly through the downregulation of tumor suppressor-gene Cyclin G2 (CCNG2), which is mediated by EZH2 and LSD1. In conclusion, our results suggested that LINC00460 acted as an oncogene in GC to inhibit the expression of CCNG2 at least partly by binding with EZH2 and LSD1. Our study could provide additional insights into the development of novel target therapeutic methods for GC.
Collapse
Affiliation(s)
- Jiebin Yang
- Department of Gastroenterology, Zhongshan Hospital, Xiamen University, Xiamen, Fujian, P.R. China; Department of Clinical Medicine, Fujian Medical University, Fuzhou, Fujian, P.R. China
| | - Yikai Lian
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, P.R. China; School of Medicine, Xiamen University, Xiamen, Fujian, P.R. China
| | - Renzhi Yang
- School of Medicine, Xiamen University, Xiamen, Fujian, P.R. China
| | - Yifan Lian
- Department of Gastroenterology, Zhongshan Hospital, Xiamen University, Xiamen, Fujian, P.R. China; School of Medicine, Xiamen University, Xiamen, Fujian, P.R. China
| | - Jingtong Wu
- Department of Gastroenterology, Zhongshan Hospital, Xiamen University, Xiamen, Fujian, P.R. China; School of Medicine, Xiamen University, Xiamen, Fujian, P.R. China
| | - Jingjing Liu
- Department of Gastroenterology, Zhongshan Hospital, Xiamen University, Xiamen, Fujian, P.R. China
| | - Keming Wang
- Department of Oncology, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, P.R. China.
| | - Hongzhi Xu
- Department of Gastroenterology, Zhongshan Hospital, Xiamen University, Xiamen, Fujian, P.R. China.
| |
Collapse
|
214
|
Zhou Q, Wu X, Wang X, Yu Z, Pan T, Li Z, Chang X, Jin Z, Li J, Zhu Z, Liu B, Su L. The reciprocal interaction between tumor cells and activated fibroblasts mediated by TNF-α/IL-33/ST2L signaling promotes gastric cancer metastasis. Oncogene 2020; 39:1414-1428. [PMID: 31659258 PMCID: PMC7018661 DOI: 10.1038/s41388-019-1078-x] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 10/13/2019] [Accepted: 10/15/2019] [Indexed: 01/25/2023]
Abstract
Gastric cancer (GC) is characterized by extensive local invasion, distant metastasis and poor prognosis. In most cases, GC progression is associated with aberrant expression of cytokines or activation of signaling cascades mediated by tumor-stroma interactions. However, the mechanisms by which these interactions contribute to GC progression are poorly understood. In this study, we find that IL-33 and its receptor ST2L are upregulated in the human GC and served as prognostic markers for poor survival of GC patients. In a co-culture model with GC cells and cancer-associated fibroblasts (CAFs), we further demonstrate that CAFs-derived IL-33 enhances the migration and invasion of GC cells by inducing the epithelial-mesenchymal transition (EMT) through activation of the ERK1/2-SP1-ZEB2 pathway in a ST2L-dependent manner. Furthermore, the secretion of IL-33 by CAFs can be induced by the proinflammatory cytokines TNF-α that is released by GC cells via TNFR2-NF-κB-IRF-1 pathway. Additionally, silencing of IL-33 expression in CAFs or ST2L expression in GC cells inhibits the peritoneal dissemination and metastatic potential of GC cells in nude mice. Taken together, these results characterize a critical role of the interaction between epithelial-stroma mediated by the TNF-α/IL-33/ST2L signaling in GC progression, and provide a rationale for targeting this pathway to treat GC metastasis.
Collapse
Affiliation(s)
- Quan Zhou
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, People's Republic of China
- Department of Urology, Center for Organ Transplantation, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, People's Republic of China
| | - Xiongyan Wu
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, People's Republic of China
| | - Xiaofeng Wang
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, People's Republic of China
| | - Zhenjia Yu
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, People's Republic of China
| | - Tao Pan
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, People's Republic of China
| | - Zhen Li
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, People's Republic of China
| | - Xinyu Chang
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, People's Republic of China
| | - Zhijian Jin
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, People's Republic of China
| | - Jianfang Li
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, People's Republic of China
| | - Zhenggang Zhu
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, People's Republic of China
| | - Bingya Liu
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, People's Republic of China.
| | - Liping Su
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, People's Republic of China.
| |
Collapse
|
215
|
Rizzo A, Mollica V, Ricci AD, Maggio I, Massucci M, Rojas Limpe FL, Fabio FD, Ardizzoni A. Third- and later-line treatment in advanced or metastatic gastric cancer: a systematic review and meta-analysis. Future Oncol 2019; 16:4409-4418. [PMID: 31793342 DOI: 10.2217/fon-2019-0429] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Aim: We performed a systematic review and meta-analysis to investigate the efficacy and safety of third-line (TLT) and salvage treatment (ST) in advanced or metastatic gastric cancer. Materials & methods: Eligible studies included randomized clinical trials assessing TLT and ST versus placebo or best supportive care. Outcomes of interest included: overall survival, objective response rate and disease control rate in TLT; progression-free survival in ST; grade 3-4 adverse events in ST. Results: The use of TLT and ST was superior to placebo or best supportive care in terms of prolonging overall survival and progression-free survival. Hematological toxicities were more frequent in ST. Conclusion: TLT and ST are considerable and tolerable treatment options for patients with advanced or metastatic gastric cancer. Given the substantial heterogeneities affecting the efficacy analyses, these results have to be interpreted cautiously.
Collapse
Affiliation(s)
- Alessandro Rizzo
- Division of Medical Oncology, S Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Veronica Mollica
- Division of Medical Oncology, S Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Angela Dalia Ricci
- Division of Medical Oncology, S Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Ilaria Maggio
- Division of Medical Oncology, S Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Maria Massucci
- Division of Medical Oncology, S Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | | | - Francesca Di Fabio
- Division of Medical Oncology, S Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Andrea Ardizzoni
- Division of Medical Oncology, S Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| |
Collapse
|
216
|
Jiang Y, Xie J, Huang W, Chen H, Xi S, Han Z, Huang L, Lin T, Zhao LY, Hu YF, Yu J, Cai SR, Li T, Li G. Tumor Immune Microenvironment and Chemosensitivity Signature for Predicting Response to Chemotherapy in Gastric Cancer. Cancer Immunol Res 2019; 7:2065-2073. [PMID: 31615816 DOI: 10.1158/2326-6066.cir-19-0311] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 08/08/2019] [Accepted: 10/10/2019] [Indexed: 12/29/2022]
Abstract
Current gastric cancer staging alone cannot predict prognosis and adjuvant chemotherapy benefits in stage II and III gastric cancer. Tumor immune microenvironment biomarkers and tumor-cell chemosensitivity might add predictive value to staging. This study aimed to construct a predictive signature integrating tumor immune microenvironment and chemosensitivity-related features to improve the prediction of survival and adjuvant chemotherapy benefits in patients with stage II to III gastric cancer. We used IHC to assess 26 features related to tumor, stroma, and chemosensitivity in tumors from 223 patients and evaluated the association of the features with disease-free survival (DFS) and overall survival (OS). Support vector machine (SVM)-based methods were used to develop the predictive signature, which we call the SVM signature. Validation of the signature was performed in two independent cohorts of 445 patients. The diagnostic signature integrated seven features: CD3+ cells at the invasive margin (CD3 IM), CD8+ cells at the IM (CD8 IM), CD45RO+ cells in the center of tumors (CD45RO CT), CD66b+ cells at the IM (CD66b IM), CD34+ cells, periostin, and cyclooxygenase-2. Patients fell into low- and high-SVM groups with significant differences in 5-year DFS and OS in the training and validation cohorts (all P < 0.001). The signature was an independent prognosis indicator in multivariate analysis in each cohort. The signature had better prognostic value than various clinicopathologic risk factors and single features. High-SVM patients exhibited a favorable response to adjuvant chemotherapy. Thus, this SVM signature predicted survival and has the potential for identifying patients with stage II and III gastric cancer who could benefit from adjuvant chemotherapy.
Collapse
Affiliation(s)
- Yuming Jiang
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jingjing Xie
- Research Center for Clinical Pharmacology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weicai Huang
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hao Chen
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Sujuan Xi
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Department of Infectious Disease, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhen Han
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lei Huang
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tian Lin
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Li-Ying Zhao
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yan-Feng Hu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiang Yu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shi-Rong Cai
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Tuanjie Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Guoxin Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
217
|
Li J, Wang L, He F, Li B, Han R. Long noncoding RNA LINC00629 restrains the progression of gastric cancer by upregulating AQP4 through competitively binding to miR-196b-5p. J Cell Physiol 2019; 235:2973-2985. [PMID: 31674022 DOI: 10.1002/jcp.29203] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 09/03/2019] [Indexed: 12/24/2022]
Abstract
Gastric cancer continues to be a common cancer in the world with high incidence and mortality. Accumulating evidence has implicated long noncoding RNAs (lncRNAs) in gastric cancer progression. Here, this study identified the potential role of a novel lncRNA, LINC00629 in gastric cancer and to elucidate the underlying mechanism. Initially, microarray-based gene expression profiling of gastric cancer was employed to identify differentially expressed genes. Next, the expression of LINC00629, microRNA-196b-5p (miR-196b-5p) and aquaporin 4 (AQP4) in clinical gastric cancer tissues was determined and the cell line presenting with the lowest LINC00629 expression was selected. The interaction among LINC00629, miR-196b-5p, and AQP4 was identified. Expression of LINC00629, miR-196b-5p, and AQP4 in gastric cancer cells were altered and then biological behaviors of gastric cancer cells were assessed by 5-ethynyl-2'-deoxyuridine and Transwell assays. Tumor formation in vivo was evaluated in nude mice. In gastric cancer, expression of LINC00629 and AQP4 was downregulated, and expression of miR-196b-5p was upregulated. Proliferation, invasion, and migration of gastric cancer cells were reduced after overexpression of LINC00629. LINC00629 competitively bound to miR-196b-5p, while AQP4 was a target of miR-196b-5p. Either downregulating miR-196b-5p or upregulating AQP4 could restrain the development of gastric cancer in vitro. LINC00629 overexpression repressed the growth of transplanted tumors in vivo. Taken together, LINC00629 competitively bound to miR-196b-5p to upregulate AQP4 expression, thereby inhibiting gastric cancer progression. Therefore, understanding of this mechanism may help to improve gastric cancer treatment.
Collapse
Affiliation(s)
- Jun Li
- Departement of Gastrointestinal Surgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Lei Wang
- Departement of Gastrointestinal Surgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Fang He
- Departement of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Bo Li
- Departement of Gastrointestinal Surgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Ruidong Han
- Departement of Gastrointestinal Surgery, General Hospital of Ningxia Medical University, Yinchuan, China
| |
Collapse
|
218
|
Abdi E, Latifi‐Navid S, Zahri S, Yazdanbod A, Pourfarzi F. Risk factors predisposing to cardia gastric adenocarcinoma: Insights and new perspectives. Cancer Med 2019; 8:6114-6126. [PMID: 31448582 PMCID: PMC6792520 DOI: 10.1002/cam4.2497] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 07/17/2019] [Accepted: 08/01/2019] [Indexed: 12/12/2022] Open
Abstract
Recent decades have seen an alarming increase in the incidence of cardia gastric adenocarcinoma (CGA) while noncardia gastric adenocarcinoma (NCGA) has decreased. In 2012, 260 000 CGA cases (age-standardised rate (ASR); 3.3/100 000) and 691 000 NCGA cases (ASR; 8.8/100 000) were reported worldwide. Compared with women, men had greater rates for both the subsites, especially for CGA. Recently, four molecular subtypes of GC have been proposed by the Cancer Genome Atlas (TCGA) and the Asian Cancer Research Group (ACRG); however, these classifications do not take into account predisposing germline variants and their possible interaction with somatic alterations in carcinogenesis. The etiology of adenocarcinoma of the cardia and the gastroesophageal junction (GEJ) is not known. It is thought that CGA is distinct from adenocarcinomas located in the esophagus or distal stomach, both epidemiologically and biologically. Moreover, CGA is often identified in the advanced stage having a poor prognosis. Therefore, understanding the risk and the role of predisposing factors in etiology of CGA can inform clinical practice and counseling for risk reduction. In this paper, we showed that GC family history, lifestyle, demographics, gastroesophageal reflux disease, Helicobacter pylori infection, and multiple genetic and epigenetic risk factors as well as several predisposing conditions may underlie susceptibility to CGA. However, several genome-wide association studies (GWASs) should be conducted to identify novel high-penetrance genes and pathways as well as causal germline variants predisposing to CGA. They must include different ethnic groups, especially from high-incidence countries for CGA, because some risk loci are ancestry-specific. In parallel, statistical methods can be developed to identify cancer predisposition genes (CPGs) from tumor sequencing data. It is also necessary to find novel long noncoding RNAs related to the risk of CGA. Taken altogether, new cancer risk prediction models, including all genetic and nongenetic factors influencing risk, should be developed to facilitate risk assessment, disease prevention, and early diagnosis and intervention of CGA in the future.
Collapse
Affiliation(s)
- Esmat Abdi
- Department of BiologyFaculty of SciencesUniversity of Mohaghegh ArdabiliArdabilIran
| | - Saeid Latifi‐Navid
- Department of BiologyFaculty of SciencesUniversity of Mohaghegh ArdabiliArdabilIran
| | - Saber Zahri
- Department of BiologyFaculty of SciencesUniversity of Mohaghegh ArdabiliArdabilIran
| | - Abbas Yazdanbod
- Digestive Diseases Research CenterArdabil University of Medical SciencesArdabilIran
| | - Farhad Pourfarzi
- Digestive Diseases Research CenterArdabil University of Medical SciencesArdabilIran
| |
Collapse
|
219
|
Choi RSY, Lai WYX, Lee LTC, Wong WLC, Pei XM, Tsang HF, Leung JJ, Cho WCS, Chu MKM, Wong EYL, Wong SCC. Current and future molecular diagnostics of gastric cancer. Expert Rev Mol Diagn 2019; 19:863-874. [PMID: 31448971 DOI: 10.1080/14737159.2019.1660645] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 08/23/2019] [Indexed: 12/14/2022]
Abstract
Introduction: Gastric cancer (GC) is the fifth most common cancer and confers the second-highest mortality among other cancers. Improving the survival rates of GC patients requires prompt and accurate diagnosis and effective treatment which is often preceded by the poorly understood pathogenic mechanisms. Area covered: This literature review aims to summarize current understanding of genetic and molecular alterations that promote carcinogenesis including (1) activation of oncogenes, (2) overexpression of growth factors, receptors and matrix metalloproteinases, (3) inactivation of tumor suppressor genes, DNA repair genes, and cell adhesion molecules and (4) alterations of cell-cycle regulators that regulate biological characteristics of cancer cells. Moreover, the significance of molecular biomarkers such as micro-RNAs (miRNAs) and long non-coding RNAs (lncRNAs) and advanced molecular techniques including droplet digital polymerase chain reaction (ddPCR), quantitative PCR (qPCR) and next-generation sequencing (NGS) are also discussed. Expert opinion: A GC-specific panel of biomarkers based on the NGS or ddPCR has the potential for diagnosis, prognosis, and monitoring treatment response in GC patients. Despite the requirements for validation in larger population in clinical studies, race-specific differences in the gene panel have also to be examined by performing the clinical trials in subjects with different races.
Collapse
Affiliation(s)
- Rachel Sin-Yu Choi
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, Hong Kong Polytechnic University , Hong Kong , Hong Kong Special Administrative Region, China
| | - Wing Yin Xenia Lai
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, Hong Kong Polytechnic University , Hong Kong , Hong Kong Special Administrative Region, China
| | - Lok Ting Claire Lee
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, Hong Kong Polytechnic University , Hong Kong , Hong Kong Special Administrative Region, China
| | - Wing Lam Christa Wong
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, Hong Kong Polytechnic University , Hong Kong , Hong Kong Special Administrative Region, China
| | - Xiao Meng Pei
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, Hong Kong Polytechnic University , Hong Kong , Hong Kong Special Administrative Region, China
| | - Hin Fung Tsang
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, Hong Kong Polytechnic University , Hong Kong , Hong Kong Special Administrative Region, China
| | - Joel Johnson Leung
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, Hong Kong Polytechnic University , Hong Kong , Hong Kong Special Administrative Region, China
| | - William Chi Shing Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital , Kowloon , Hong Kong Special Administrative Region, China
| | - Man Kee Maggie Chu
- Division of Life Science, The Hong Kong University of Science and Technology , Clear Water Bay , Hong Kong Special Administrative Region, China
| | - Elaine Yue Ling Wong
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, Hong Kong Polytechnic University , Hong Kong , Hong Kong Special Administrative Region, China
| | - Sze Chuen Cesar Wong
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, Hong Kong Polytechnic University , Hong Kong , Hong Kong Special Administrative Region, China
| |
Collapse
|
220
|
RAB13 as a novel prognosis marker promotes proliferation and chemotherapeutic resistance in gastric cancer. Biochem Biophys Res Commun 2019; 519:113-120. [PMID: 31474334 DOI: 10.1016/j.bbrc.2019.08.141] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 08/25/2019] [Indexed: 12/19/2022]
Abstract
Gastric cancer (GC) is still a major lethal gastrointestinal tumor. In this study, we clarified that RAB13, which is a member of Rab GTPase family and responsible for cargos delivery between the Golgi and the plasma membrane, plays critical roles in the proliferation and the chemotherapeutic resistance in GC cells. Analyzing RAB13 expression in GC specimens, we found that its mRNA level was higher in cancerous tissues compared with normal counterparts and this increase was further associated with malignant progression of GC. Moreover, increased RAB13 indicated poor overall survival (OS) and progression free survival (PFS) in GC patients. We then found that deletion of RAB13 inhibited the proliferation and promoted the apoptosis in AGS and NCI-N87 cells, the impairments of viability which was due to reduced amount of RAB13 anchoring the plasma membrane and attenuated cellular response to EGF treatment and the activation of downstream Akt/ERK/mTOR signaling pathways accordingly. Moreover, in vitro experiments showed that RAB13 deletion enhanced the sensitization of AGS and NCI-N87 cells toward cisplatin (CDDP) and 5-fluorouracil (5-FU) treatment respectively. Together, these data demonstrate that RAB13 promotes the proliferation and confers CDDP and 5-FU resistance to GC cells, which provides experimental support to target this protein in future clinical practice.
Collapse
|
221
|
Yin X, Zhang F, Guo Z, Kong W, Wang Y. Integrative analysis of miRNA and mRNA expression profiles reveals a novel mRNA/miRNA signature to improve risk classification for patients with gastric cancer. Oncol Lett 2019; 18:2330-2339. [PMID: 31402938 PMCID: PMC6676680 DOI: 10.3892/ol.2019.10536] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Accepted: 05/25/2019] [Indexed: 02/03/2023] Open
Abstract
Gastric cancer (GC) is one of the most common types of malignant cancer and is associated with poor prognosis. Although the prognosis of patients with GC is associated with grade, stage and lymph node metastases, these traditional clinical features are inadequate to predict the outcome of GC. Therefore, there has been an increased focus on identifying novel molecular biomarkers for early diagnosis and prognosis, in order to improve outcomes in GC. In the present study, an integrative analysis of microRNA (miRNA) expression profiles, mRNA expression profiles and clinical characteristics was performed in a large cohort of patients with GC in order to identify an integrative prognostic model for improving postoperative risk classification. An integrative mRNA/miRNA signature (IMMIS), comprised of three miRNAs and one mRNA, was identified from a large number of differentially expressed miRNAs and mRNAs using univariate and multivariate Cox regression analysis. The prognostic value of the IMMIS was validated in the discovery cohort, testing cohort and The Cancer Genome Atlas (TCGA) cohort. The present results suggested that the identified signature had a reliable predictive performance and could classify the patients into high- and low-risk groups with significantly different overall survival times. In the discovery cohort, the hazard ratio (HR) was 2.805 with a 95% CI=1.722–4.567 (P<0.001). The median overall survival time as 1.49 vs. 3.85 years. In the testing cohort, the HR was 1.625 with a 95% CI=1.004–2.638 (P=0.039) and the median overall survival time was 2.17 vs. 4.62 years. In the TCGA cohort, the HR was 2.139 with a 95% CI=1.519–3.012 (P<0.001) and the median overall survival time was 1.53 vs. 4.62 years. The IMMIS constituted a reliable independent prognostic factor compared with clinical covariates, including age, sex, grade and stage, as indicated by multivariate and stratified analyses. Furthermore, comparative analysis revealed that the predictive value of the IMMIS was superior to the mRNA-based signature alone. The present results suggested the potential value of the IMMIS as a promising novel biomarker for improving the clinical management of patients with GC.
Collapse
Affiliation(s)
- Xiang Yin
- Department of Minimally Invasive Tumor Surgery, Daqing Oilfield General Hospital, Daqing, Heilongjiang 163453, P.R. China
| | - Fumin Zhang
- Department of Minimally Invasive Tumor Surgery, Daqing Oilfield General Hospital, Daqing, Heilongjiang 163453, P.R. China
| | - Zhongwu Guo
- Department of Minimally Invasive Tumor Surgery, Daqing Oilfield General Hospital, Daqing, Heilongjiang 163453, P.R. China
| | - Weiyuan Kong
- Department of Minimally Invasive Tumor Surgery, Daqing Oilfield General Hospital, Daqing, Heilongjiang 163453, P.R. China
| | - Yuanyuan Wang
- Department of Gastroenterology, Daqing Long Nan Hospital, Daqing, Heilongjiang 163453, P.R. China
| |
Collapse
|
222
|
Lin S, Zhou R, Zeng D, Wu J, Wu J, Zhang J, Sun H, Zhu S, Shi M, Bin J, Liao Y, Liao W. A novel assessing system for predicting the prognosis of gastric cancer. Epigenomics 2019; 11:1251-1266. [PMID: 31364879 DOI: 10.2217/epi-2019-0151] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Aim: To develop novel diagnostic tools that can predict the prognosis of gastric cancer. Material & methods: Using RNA expression data from The Cancer Genome Atlas and Gene Expression Omnibus, we established protein-coding RNAs-noncoding RNAs-tumor microenvironment type (PNM) scores, which contain signatures of tumor protein coding genes (P), tumor noncoding genes (N) and immune/stroma cells in tumor microenvironment (M) to predict the prognosis of gastric cancer. Results & conclusion: Based on PNM scores, gastric cancer patients were divided into three subgroups and Kaplan-Meier survival curves revealed significant differences among the subgroups (p < 0.001). Our study showed that the PNM scores could be used as a robust predicting tool for the prognosis of gastric cancer.
Collapse
Affiliation(s)
- Siheng Lin
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Rui Zhou
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Dongqiang Zeng
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Jiani Wu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Jianhua Wu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Jingwen Zhang
- Department of Medicine Ultrasonics, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Huiying Sun
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Shaowei Zhu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Min Shi
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Jianping Bin
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Yulin Liao
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| |
Collapse
|
223
|
Low Expression of GLIS2 Gene Might Associate with Radiosensitivity of Gastric Cancer. JOURNAL OF ONCOLOGY 2019; 2019:2934925. [PMID: 31281358 PMCID: PMC6590498 DOI: 10.1155/2019/2934925] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/30/2019] [Accepted: 05/20/2019] [Indexed: 01/05/2023]
Abstract
Human gene GLIS family zinc finger 2 (GLIS2) is a member of GLI-similar zinc finger protein family. Previous studies indicated GLIS2 gene involved in tumorigenesis mechanisms. However, the association between GLIS2 expression and radiosensitivity of gastric cancer has not been well understood. In this study, we used the gastric cancer database in TCGA, and significant association was observed between the low expression of GLIS2 and radiosensitivity of patients with gastric cancer. The adjusted HR values for radiotherapy were 0.162(0.035-0.756) and 0.089(0.014-0.564), with p values 0.021 and 0.010, respectively, in training and testing data, for these patients with low expression of GLIS2, while for patients with high expression of GLIS2, there was no significant survival difference between radiotherapy and nonradiotherapy groups. The adjusted HR were 0.676(0.288-1.586) and 0.508(0.178-1.450), with p values 0.368 and 0.206 in training and testing data, respectively. Further study showed that, for low expression patients, radiotherapy did not significantly increase new tumor event rate and disease progression rate, which partially supported our assumption. These results suggested that low expression of GLIS2 might significantly associate with the radiosensitivity of patients with gastric cancer. The GLIS2 gene might be a potential effective molecular marker of gastric cancer for precise radiotherapy.
Collapse
|
224
|
Sex determining region Y-box 12 (SOX12) promotes gastric cancer metastasis by upregulating MMP7 and IGF1. Cancer Lett 2019; 452:103-118. [DOI: 10.1016/j.canlet.2019.03.035] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 03/02/2019] [Accepted: 03/21/2019] [Indexed: 02/07/2023]
|
225
|
Sammarco G, Varricchi G, Ferraro V, Ammendola M, De Fazio M, Altomare DF, Luposella M, Maltese L, Currò G, Marone G, Ranieri G, Memeo R. Mast Cells, Angiogenesis and Lymphangiogenesis in Human Gastric Cancer. Int J Mol Sci 2019; 20:2106. [PMID: 31035644 PMCID: PMC6540185 DOI: 10.3390/ijms20092106] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/15/2019] [Accepted: 04/19/2019] [Indexed: 12/12/2022] Open
Abstract
Gastric cancer is diagnosed in nearly one million new patients each year and it remains the second leading cause of cancer-related deaths worldwide. Although gastric cancer represents a heterogeneous group of diseases, chronic inflammation has been shown to play a role in tumorigenesis. Cancer development is a multistep process characterized by genetic and epigenetic alterations during tumour initiation and progression. The stromal microenvironment is important in maintaining normal tissue homeostasis or promoting tumour development. A plethora of immune cells (i.e., lymphocytes, macrophages, mast cells, monocytes, myeloid-derived suppressor cells, Treg cells, dendritic cells, neutrophils, eosinophils, natural killer (NK) and natural killer T (NKT) cells) are components of gastric cancer microenvironment. Mast cell density is increased in gastric cancer and there is a correlation with angiogenesis, the number of metastatic lymph nodes and the survival of these patients. Mast cells exert a protumorigenic role in gastric cancer through the release of angiogenic (VEGF-A, CXCL8, MMP-9) and lymphangiogenic factors (VEGF-C and VEGF-F). Gastric mast cells express the programmed death ligands (PD-L1 and PD-L2) which are relevant as immune checkpoints in cancer. Several clinical undergoing trials targeting immune checkpoints could be an innovative therapeutic strategy in gastric cancer. Elucidation of the role of subsets of mast cells in different human gastric cancers will demand studies of increasing complexity beyond those assessing merely mast cell density and microlocalization.
Collapse
Affiliation(s)
- Giuseppe Sammarco
- Department of Health Science, General Surgery, Magna Graecia University, Medicine School of Germaneto, 88100 Catanzaro, Italy.
| | - Gilda Varricchi
- Department of Translational Medical Sciences (DISMET) and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy.
- WAO Center of Excellence, 80131 Naples, Italy.
| | - Valentina Ferraro
- Department of Biomedical Sciences and Human Oncology, Unit of Endocrine, Digestive and Emergency Surgery, Aldo Moro University, 74124 Bari, Italy.
| | - Michele Ammendola
- Department of Health Science, General Surgery, Magna Graecia University, Medicine School of Germaneto, 88100 Catanzaro, Italy.
| | - Michele De Fazio
- Department of Emergency and Organ Transplantation, Aldo Moro University, 74124 Bari, Italy.
| | | | - Maria Luposella
- Cardiovascular Disease Unit, San Giovanni di Dio Hospital, 88900 Crotone, Italy.
| | - Lorenza Maltese
- Pathology Unit, Pugliese-Ciaccio Hospital, 88100 Catanzaro, Italy.
| | - Giuseppe Currò
- Department of Health Science, General Surgery, Magna Graecia University, Medicine School of Germaneto, 88100 Catanzaro, Italy.
- Department of Human Pathology of Adult and Evolutive Age G. Barresi, University of Messina, 98122 Messina, Italy.
| | - Gianni Marone
- Department of Translational Medical Sciences (DISMET) and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy.
- WAO Center of Excellence, 80131 Naples, Italy.
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council (CNR), 80131 Naples, Italy.
| | - Girolamo Ranieri
- Interventional Oncology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre, Istituto Tumori Giovanni Paolo II, 74124 Bari, Italy.
| | - Riccardo Memeo
- Department of Emergency and Organ Transplantation, Aldo Moro University, 74124 Bari, Italy.
| |
Collapse
|
226
|
Hao S, Lv J, Yang Q, Wang A, Li Z, Guo Y, Zhang G. Identification of Key Genes and Circular RNAs in Human Gastric Cancer. Med Sci Monit 2019; 25:2488-2504. [PMID: 30948703 PMCID: PMC6463957 DOI: 10.12659/msm.915382] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Accepted: 03/22/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Globally, gastric cancer (GC) is the third most common source of cancer-associated mortality. The aim of this study was to identify key genes and circular RNAs (circRNAs) in GC diagnosis, prognosis, and therapy and to further explore the potential molecular mechanisms of GC. MATERIAL AND METHODS Differentially expressed genes (DEGs) and circRNAs (DE circRNAs) between GC tissues and adjacent non-tumor tissues were identified from 3 mRNA and 3 circRNA expression profiles. Functional analyses were performed, and protein-protein interaction (PPI) networks were constructed. The significant modules and key genes in the PPI networks were identified. Kaplan-Meier analysis was performed to evaluate the prognostic value of these key genes. Potential miRNA-binding sites of the DE circRNAs and target genes of these miRNAs were predicted and used to construct DE circRNA-miRNA-mRNA networks. RESULTS A total of 196 upregulated and 311 downregulated genes were identified in GC. The results of functional analysis showed that these DEGs were significantly enriched in a variety of functions and pathways, including extracellular matrix-related pathways. Ten hub genes (COL1A1, COL3A1, COL1A2, COL5A2, FN1, THBS1, COL5A1, SPARC, COL18A1, and COL11A1) were identified via PPI network analysis. Kaplan-Meier analysis revealed that 7 of these were associated with a poor overall survival in GC patients. Furthermore, we identified 2 DE circRNAs, hsa_circ_0000332 and hsa_circ_0021087. To reveal the potential molecular mechanisms of circRNAs in GC, DE circRNA-microRNA-mRNA networks were constructed. CONCLUSIONS Key candidate genes and circRNAs were identified, and novel PPI and circRNA-microRNA-mRNA networks in GC were constructed. These may provide useful information for the exploration of potential biomarkers and targets for the diagnosis, prognosis, and therapy of GC.
Collapse
Affiliation(s)
- Shuhong Hao
- Department of Hematology and Oncology, The Second Hospital of Jilin University, Changchun, Jilin, P.R. China
| | - Junfeng Lv
- Department of Radiology, The Second Hospital of Jilin University, Changchun, Jilin, P.R. China
| | - Qiwei Yang
- Medical Research Center, The Second Hospital of Jilin University, Changchun, Jilin, P.R. China
| | - Ao Wang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, Jilin, P.R. China
| | - Zhaoyan Li
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, Jilin, P.R. China
| | - Yuchen Guo
- Department of Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun, Jilin, P.R. China
| | - Guizhen Zhang
- Medical Research Center, The Second Hospital of Jilin University, Changchun, Jilin, P.R. China
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, Jilin, P.R. China
| |
Collapse
|
227
|
Zhu C, Yang Q, Xu J, Zhao W, Zhang Z, Xu D, Zhang Y, Zhao E, Zhao G. Somatic mutation of DNAH genes implicated higher chemotherapy response rate in gastric adenocarcinoma patients. J Transl Med 2019; 17:109. [PMID: 30944005 PMCID: PMC6448266 DOI: 10.1186/s12967-019-1867-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Accepted: 03/29/2019] [Indexed: 12/13/2022] Open
Abstract
Background The dynein axonemal heavy chain (DNAH) family of genes encode the dynein axonemal heavy chain, which is involved in cell motility. Genomic variations of DNAH family members have been frequently reported in diverse kinds of malignant tumors. In this study, we analyzed the genomic database to evaluate the mutation status of DNAH genes in gastric adenocarcinoma and further identified the significance of mutant DNAH genes as effective molecular biomarkers for predicting chemotherapy response in gastric cancer patients. Methods We analyzed the clinical and genomic data of gastric cancer patients published in The Cancer Genome Atlas (TCGA) project. Data on chemotherapy response, overall survival (OS) and chemotherapy-free survival were retrieved. Then, we verified the results via targeted sequencing of gastric cancer patients with similar clinical characteristics but different chemotherapeutic outcomes. Results In total, 132 gastric adenocarcinoma patients undergoing chemotherapy treatment from TCGA were included in our study. Somatic mutations in all 13 members of the DNAH family of genes were associated with different chemotherapy responses. Compared with patients with wild-type DNAH genes (n = 59), a significantly higher proportion of those with mutations in DNAH genes (n = 73) (55.9% vs 80.8%) responded to chemotherapy (P = 0.002). Moreover, DNAH mutations were correlated with significantly better OS (P = 0.027), chemotherapy-free survival (P = 0.027), fluoropyrimidine-free survival (P = 0.048) and platinum-free survival (P = 0.014). DNAH mutation status was an independent risk factor for OS (P = 0.015), chemotherapy-free survival (P = 0.015) and platinum-free survival (P = 0.011). We identified somatic mutations in 27 (42.2%) of the 64 stage III gastric adenocarcinoma patients receiving fluoropyrimidine-based chemotherapy by targeted exon sequencing with strict screening conditions. In our own cohort, a significantly higher proportion of patients (n = 32) with DNAH mutations than patients with wild-type DNAH genes (n = 32) had a good prognosis (OS > 48 months) (70.4% vs 35.1%) (P = 0.005). Conclusions Dynein axonemal heavy chain gene mutations contribute positively to chemotherapy sensitivity in gastric cancer patients. Electronic supplementary material The online version of this article (10.1186/s12967-019-1867-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chunchao Zhu
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, People's Republic of China.,State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Qin Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Jia Xu
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, People's Republic of China.,State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Wenyi Zhao
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, People's Republic of China.,State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Zizhen Zhang
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, People's Republic of China.,State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Danhua Xu
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, People's Republic of China.,State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Yeqian Zhang
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, People's Republic of China.,State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Enhao Zhao
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, People's Republic of China.
| | - Gang Zhao
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, People's Republic of China.
| |
Collapse
|
228
|
Ahadi A, Safavi MS. miR-335-5p has an important role in the progression of gastric cancer by down-regulation of CEACAM5. Meta Gene 2019. [DOI: 10.1016/j.mgene.2018.10.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
|
229
|
Díaz Del Arco C, Estrada Muñoz L, Molina Roldán E, Cerón Nieto MÁ, Ortega Medina L, García Gómez de Las Heras S, Fernández Aceñero MJ. Immunohistochemical classification of gastric cancer based on new molecular biomarkers: a potential predictor of survival. Virchows Arch 2018; 473:687-695. [PMID: 30140949 DOI: 10.1007/s00428-018-2443-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 08/11/2018] [Accepted: 08/15/2018] [Indexed: 12/17/2022]
Abstract
Several classification systems have been described for stratifying patients with gastric carcinoma (GC). However, their prognostic value is low, and there is an urgent need for identification of molecular markers and development of new classifications. Retrospective study of 206 cases of GC diagnosed and surgically resected in our hospital between 2000 and 2017. Clinicopathological features of all cases were assessed and tissue microarrays were constructed for immunohistochemical (IHC) study. Patients were stratified based on IHC results. Mean patient age was 71 years and most patients were male (54.6%). Most tumors were located in the gastric antrum and body, and they were mostly fungoid or ulcerative lesions. GC were mainly intestinal-type tumors and 60.3% were diagnosed at pT3. 56.2% of patients showed recurrences and 29.4% died due to GC. According to our IHC classification, 23.5% of tumors showed microsatellite instability, 6% were E-cadherin negative, 53.5% were stable-p53 not overexpressed, and 17% were stable with p53 overexpression. IHC classification was significantly correlated with patient gender, gross morphology, Laurén classification, tumor necrosis, perineural infiltration, type of leading edge, and patient outcome. Multivariate analysis showed that IHC subtype was significantly and independently associated with overall survival, together with clinical symptoms, signet cell phenotype, tumor grade and vessel invasion. The application of IHC classifications based on molecular biomarkers in clinical practice can aid in the stratification of GC patients. More studies are needed to evaluate the reproducibility and clinical significance of these classifications.
Collapse
Affiliation(s)
- Cristina Díaz Del Arco
- Department of Surgical Pathology, Hospital Clínico San Carlos, 28040, Madrid, Spain.
- Complutense University of Madrid, Madrid, Spain.
| | - Lourdes Estrada Muñoz
- Department of Surgical Pathology, Hospital Rey Juan Carlos, Madrid, Spain
- Rey Juan Carlos University, Alcorcón, Madrid, Spain
| | - Elena Molina Roldán
- Department of Surgical Pathology, Hospital Clínico San Carlos, 28040, Madrid, Spain
| | | | - Luis Ortega Medina
- Department of Surgical Pathology, Hospital Clínico San Carlos, 28040, Madrid, Spain
- Complutense University of Madrid, Madrid, Spain
| | | | - Mª Jesús Fernández Aceñero
- Department of Surgical Pathology, Hospital Clínico San Carlos, 28040, Madrid, Spain
- Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
230
|
Ikoma N, Badgwell BD. ASO Author Reflections: Nodal Downstaging in Gastric Cancer. Ann Surg Oncol 2018; 25:751-752. [PMID: 30353393 PMCID: PMC7705585 DOI: 10.1245/s10434-018-6934-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Indexed: 08/30/2023]
Affiliation(s)
- Naruhiko Ikoma
- Department of Surgical Oncology, Unit 1484, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Brian D Badgwell
- Department of Surgical Oncology, Unit 1484, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
231
|
Jiang Y, Xie J, Han Z, Liu W, Xi S, Huang L, Huang W, Lin T, Zhao L, Hu Y, Yu J, Zhang Q, Li T, Cai S, Li G. Immunomarker Support Vector Machine Classifier for Prediction of Gastric Cancer Survival and Adjuvant Chemotherapeutic Benefit. Clin Cancer Res 2018; 24:5574-5584. [PMID: 30042208 DOI: 10.1158/1078-0432.ccr-18-0848] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 06/06/2018] [Accepted: 07/17/2018] [Indexed: 12/17/2022]
Abstract
Purpose: Current tumor-node-metastasis (TNM) staging system cannot provide adequate information for prediction of prognosis and chemotherapeutic benefits. We constructed a classifier to predict prognosis and identify a subset of patients who can benefit from adjuvant chemotherapy.Experimental Design: We detected expression of 15 immunohistochemistry (IHC) features in tumors from 251 gastric cancer (GC) patients and evaluated the association of their expression level with overall survival (OS) and disease-free survival (DFS). Then, integrating multiple clinicopathologic features and IHC features, we used support vector machine (SVM)-based methods to develop a prognostic classifier (GC-SVM classifier) with features. Further validation of the GC-SVM classifier was performed in two validation cohorts of 535 patients.Results: The GC-SVM classifier integrated patient sex, carcinoembryonic antigen, lymph node metastasis, and the protein expression level of eight features, including CD3invasive margin (IM), CD3center of tumor (CT), CD8IM, CD45ROCT, CD57IM, CD66bIM, CD68CT, and CD34. Significant differences were found between the high- and low-GC-SVM patients in 5-year OS and DFS in training and validation cohorts. Multivariate analysis revealed that the GC-SVM classifier was an independent prognostic factor. The classifier had higher predictive accuracy for OS and DFS than TNM stage and can complement the prognostic value of the TNM staging system. Further analysis revealed that stage II and III GC patients with high-GC-SVM were likely to benefit from adjuvant chemotherapy.Conclusions: The newly developed GC-SVM classifier was a powerful predictor of OS and DFS. Moreover, the GC-SVM classifier could predict which patients with stage II and III GC benefit from adjuvant chemotherapy. Clin Cancer Res; 24(22); 5574-84. ©2018 AACR.
Collapse
Affiliation(s)
- Yuming Jiang
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jingjing Xie
- Research Center for Clinical Pharmacology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhen Han
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wei Liu
- Guangdong Key Laboratory of Liver Disease Research, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Biotherapy Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Sujuan Xi
- Guangdong Key Laboratory of Liver Disease Research, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Department of Infectious Disease, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lei Huang
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Weicai Huang
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Tian Lin
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Liying Zhao
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yanfeng Hu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiang Yu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qi Zhang
- Guangdong Key Laboratory of Liver Disease Research, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
- Biotherapy Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Tuanjie Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.
- Guangdong Key Laboratory of Liver Disease Research, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shirong Cai
- Department of Gastrointestinal Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Guoxin Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
232
|
Jiang Y, Yuan Q, Lv W, Xi S, Huang W, Sun Z, Chen H, Zhao L, Liu W, Hu Y, Lu L, Ma J, Li T, Yu J, Wang Q, Li G. Radiomic signature of 18F fluorodeoxyglucose PET/CT for prediction of gastric cancer survival and chemotherapeutic benefits. Theranostics 2018; 8:5915-5928. [PMID: 30613271 PMCID: PMC6299427 DOI: 10.7150/thno.28018] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 10/22/2018] [Indexed: 12/13/2022] Open
Abstract
We aimed to evaluate whether radiomic feature-based fluorine 18 (18F) fluorodeoxyglucose (FDG) positron emission tomography (PET) imaging signatures allow prediction of gastric cancer (GC) survival and chemotherapy benefits. Methods: A total of 214 GC patients (training (n = 132) or validation (n = 82) cohort) were subjected to radiomic feature extraction (80 features). Radiomic features of patients in the training cohort were subjected to a LASSO cox analysis to predict disease-free survival (DFS) and overall survival (OS) and were validated in the validation cohort. A radiomics nomogram with the radiomic signature incorporated was constructed to demonstrate the incremental value of the radiomic signature to the TNM staging system for individualized survival estimation, which was then assessed with respect to calibration, discrimination, and clinical usefulness. The performance was assessed with concordance index (C-index) and integrated Brier scores. Results: Significant differences were found between the high- and low-radiomic score (Rad-score) patients in 5-year DFS and OS in training and validation cohorts. Multivariate analysis revealed that the Rad-score was an independent prognostic factor. Incorporating the Rad-score into the radiomics-based nomogram resulted in better performance (C-index: DFS, 0.800; OS, 0.786; in the training cohort) than TNM staging system and clinicopathologic nomogram. Further analysis revealed that patients with higher Rad-scores were prone to benefit from chemotherapy. Conclusion: The newly developed radiomic signature was a powerful predictor of OS and DFS. Moreover, the radiomic signature could predict which patients could benefit from chemotherapy.
Collapse
Affiliation(s)
- Yuming Jiang
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, China
| | - Qingyu Yuan
- Nanfang PET Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Wenbing Lv
- School of Biomedical Engineering and Guangdong Provincal Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Sujuan Xi
- Guangdong Key Laboratory of Liver Disease Research, the 3rd Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
- Department of Infectious Disease, the 3rd Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Weicai Huang
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, China
| | - Zepang Sun
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, China
| | - Hao Chen
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, China
| | - Liying Zhao
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, China
| | - Wei Liu
- Guangdong Key Laboratory of Liver Disease Research, the 3rd Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Yanfeng Hu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, China
| | - Lijun Lu
- School of Biomedical Engineering and Guangdong Provincal Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jianhua Ma
- School of Biomedical Engineering and Guangdong Provincal Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Tuanjie Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, China
| | - Jiang Yu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, China
| | - Quanshi Wang
- Nanfang PET Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Guoxin Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, China
| |
Collapse
|
233
|
Liu S, Liu JW, Sun LP, Gong YH, Xu Q, Jing JJ, Yuan Y. Association of IL10 gene promoter polymorphisms with risks of gastric cancer and atrophic gastritis. J Int Med Res 2018; 46:5155-5166. [PMID: 30205739 PMCID: PMC6300941 DOI: 10.1177/0300060518792785] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE To investigate the association between polymorphisms of the interleukin 10 ( IL10) gene and risk of gastric cancer (GC) and atrophic gastritis (AG). METHODS This study enrolled patients with GC, patients with AG and healthy control subjects. Demographic data were collected and the IL10 gene -1082A/G, -819C/T and -592A/C polymorphisms were genotyped. An enzyme-linked immunosorbent assay was performed to detect Helicobacter pylori infection. RESULTS The study enrolled 556 participants including 208 in the GC group, 116 in the AG group and 232 controls (CON group). In a recessive model of the IL10-819C/T polymorphism, a significantly decreased risk of GC was found compared with AG and non-cancer subjects, respectively (AG→GC: odds ratio OR 0.41; non-cancer→GC: OR 0.57). The CC genotype demonstrated a significantly increased risk of AG compared with CON. Similar significant results were detected in males and H. pylori-negative subgroups. The ACC haplotype was associated with a decreased risk of GC compared with AG. The ATC haplotype was associated with a decreased risk of AG compared with the CON group, but it was associated with an increased risk of GC compared with AG. CONCLUSION The IL10 gene promoter -819C/T (rs1800871) polymorphism was associated with the risk of GC and AG in a Chinese population.
Collapse
Affiliation(s)
- Sa Liu
- 1 Tumour Etiology and Screening Department of Cancer Institute and General Surgery, First Hospital of China Medical University, and Key Laboratory of Cancer Aetiology and Prevention (China Medical University), Liaoning Provincial Education Department, Shenyang, Liaoning Province, China.,2 The Second Department of Oncology, Fourth Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Jing-Wei Liu
- 1 Tumour Etiology and Screening Department of Cancer Institute and General Surgery, First Hospital of China Medical University, and Key Laboratory of Cancer Aetiology and Prevention (China Medical University), Liaoning Provincial Education Department, Shenyang, Liaoning Province, China
| | - Li-Ping Sun
- 1 Tumour Etiology and Screening Department of Cancer Institute and General Surgery, First Hospital of China Medical University, and Key Laboratory of Cancer Aetiology and Prevention (China Medical University), Liaoning Provincial Education Department, Shenyang, Liaoning Province, China
| | - Yue-Hua Gong
- 1 Tumour Etiology and Screening Department of Cancer Institute and General Surgery, First Hospital of China Medical University, and Key Laboratory of Cancer Aetiology and Prevention (China Medical University), Liaoning Provincial Education Department, Shenyang, Liaoning Province, China
| | - Qian Xu
- 1 Tumour Etiology and Screening Department of Cancer Institute and General Surgery, First Hospital of China Medical University, and Key Laboratory of Cancer Aetiology and Prevention (China Medical University), Liaoning Provincial Education Department, Shenyang, Liaoning Province, China
| | - Jing-Jing Jing
- 1 Tumour Etiology and Screening Department of Cancer Institute and General Surgery, First Hospital of China Medical University, and Key Laboratory of Cancer Aetiology and Prevention (China Medical University), Liaoning Provincial Education Department, Shenyang, Liaoning Province, China
| | - Yuan Yuan
- 1 Tumour Etiology and Screening Department of Cancer Institute and General Surgery, First Hospital of China Medical University, and Key Laboratory of Cancer Aetiology and Prevention (China Medical University), Liaoning Provincial Education Department, Shenyang, Liaoning Province, China
| |
Collapse
|
234
|
Real-Time Tumor Gene Expression Profiling to Direct Gastric Cancer Chemotherapy: Proof-of-Concept “3G” Trial. Clin Cancer Res 2018; 24:5272-5281. [DOI: 10.1158/1078-0432.ccr-18-0193] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 05/28/2018] [Accepted: 07/12/2018] [Indexed: 11/16/2022]
|
235
|
Gkolfinopoulos S, Papamichael D, Papadimitriou K, Papanastasopoulos P, Vassiliou V, Kountourakis P. Advances in molecular, genetic and immune signatures of gastric cancer: Are we ready to apply them in our patients' decision making? World J Gastrointest Oncol 2018; 10:172-183. [PMID: 30079143 PMCID: PMC6068857 DOI: 10.4251/wjgo.v10.i7.172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 05/16/2018] [Accepted: 06/13/2018] [Indexed: 02/05/2023] Open
Abstract
In the last few years we have witnessed a vast expansion of our knowledge regarding the molecular and genetic profile of gastric cancer. The molecular subtypes described have shed light on the pathogenesis of the disease, thus prompting the development of new therapeutic strategies and favoring a more individualized approach for treatment. Most of the clinical trials for so called targeted therapies could be considered, at best, partially successful. In addition, checkpoint inhibitors have recently been added to our armamentarium in later stages of the disease, and combinations with chemotherapy and targeted agents are currently under development. In view of the rapid advances of molecular oncology, a new challenge for the clinical oncologist arises: The appropriate patient selection for each new therapy, which can be made possible only through the implementation of predictive biomarkers in our therapy decision making.
Collapse
|
236
|
Battaglin F, Naseem M, Puccini A, Lenz HJ. Molecular biomarkers in gastro-esophageal cancer: recent developments, current trends and future directions. Cancer Cell Int 2018; 18:99. [PMID: 30008616 PMCID: PMC6042434 DOI: 10.1186/s12935-018-0594-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 06/28/2018] [Indexed: 12/12/2022] Open
Abstract
Gastro-esophageal adenocarcinomas (GEA) represent a severe global health burden and despite improvements in the multimodality treatment of these malignancies the prognosis of patients remains poor. HER2 overexpression/amplification has been the first predictive biomarker approved in clinical practice to guide patient selection for targeted treatment with trastuzumab in advanced gastric and gastro-esophageal junction cancers. More recently, immunotherapy has been approved for the treatment of GEA and PD-L1 expression is now a biomarker required for the administration of pembrolizumab in these diseases. Significant progress has been made in recent years in dissecting the genomic makeup of GEA in order to identify distinct molecular subtypes linked to distinct patterns of molecular alterations. GEA have been found to be highly heterogeneous malignances, representing a challenge for biomarkers discovery and targeted treatment development. The current review focuses on an overview of established and novel promising biomarkers in GEA, covering recent molecular classifications from TCGA and ACRG. Main elements of molecular heterogeneity are discussed, as well as emerging mechanisms of primary and secondary resistance to HER2 targeted treatment and recent biomarker-driven trials. Future perspectives on the role of epigenetics, miRNA/lncRNA and liquid biopsy, and patient-derived xenograft models as a new platform for molecular-targeted drug discovery in GEA are presented. Our knowledge on the genomic landscape of GEA continues to evolve, uncovering the high heterogeneity and deep complexity of these tumors. The availability of new technologies and the identification of promising novel biomarker will be critical to optimize targeted treatment development in a setting where therapeutic options are currently lacking. Nevertheless, clinical validation of novel biomarkers and treatment strategies still represents an issue.
Collapse
Affiliation(s)
- Francesca Battaglin
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlake Avenue, Suite 5410, Los Angeles, CA 90033 USA
- Medical Oncology Unit 1, Clinical and Experimental Oncology Department, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy
| | - Madiha Naseem
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlake Avenue, Suite 5410, Los Angeles, CA 90033 USA
| | - Alberto Puccini
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlake Avenue, Suite 5410, Los Angeles, CA 90033 USA
- Oncologia Medica 1, Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlake Avenue, Suite 5410, Los Angeles, CA 90033 USA
| |
Collapse
|
237
|
Alessandrini L, Manchi M, De Re V, Dolcetti R, Canzonieri V. Proposed Molecular and miRNA Classification of Gastric Cancer. Int J Mol Sci 2018; 19:E1683. [PMID: 29882766 PMCID: PMC6032377 DOI: 10.3390/ijms19061683] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 05/30/2018] [Accepted: 06/01/2018] [Indexed: 12/13/2022] Open
Abstract
Gastric cancer (GC) is a common malignant neoplasm worldwide and one of the main cause of cancer-related deaths. Despite some advances in therapies, long-term survival of patients with advanced disease remains poor. Different types of classification have been used to stratify patients with GC for shaping prognosis and treatment planning. Based on new knowledge of molecular pathways associated with different aspect of GC, new pathogenetic classifications for GC have been and continue to be proposed. These novel classifications create a new paradigm in the definition of cancer biology and allow the identification of relevant GC genomic subsets by using different techniques such as genomic screenings, functional studies and molecular or epigenetic characterization. An improved prognostic classification for GC is essential for the development of a proper therapy for a proper patient population. The aim of this review is to discuss the state-of-the-art on combining histological and molecular classifications of GC to give an overview of the emerging therapeutic possibilities connected to the latest discoveries regarding GC.
Collapse
Affiliation(s)
- Lara Alessandrini
- Pathology, IRCCS CRO National Cancer Institute, 33081 Aviano, Italy.
| | - Melissa Manchi
- Pathology, IRCCS CRO National Cancer Institute, 33081 Aviano, Italy.
| | - Valli De Re
- Immunopathology and Cancer Biomarkers, IRCCS CRO National Cancer Institute, 33081 Aviano, Italy.
| | - Riccardo Dolcetti
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, QLD 4102, Australia.
| | | |
Collapse
|
238
|
Xu X, Gao F, Wang J, Tao L, Ye J, Ding L, Ji W, Chen X. MiR-122-5p inhibits cell migration and invasion in gastric cancer by down-regulating DUSP4. Cancer Biol Ther 2018; 19:427-435. [PMID: 29509059 DOI: 10.1080/15384047.2018.1423925] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE To explore the relationship between miR-122-5p and DUSP4 and their effects on gastric cancer (GC) cell mobility and invasiveness. METHODS Abnormally expressed miRNAs and mRNAs were analyzed using microarrays. The miR-122-5p and DUSP4 mRNA expression levels in GC tissues and cells were determined by RT-qPCR. The target relationship between miR-122-5p and DUSP4 was validated by dual luciferase reporter assay. GC cell mobility and invasiveness were respectively observed by wound healing assay and transwell invasion assay. Western blot and immunohistochemistry were used for detection of the expressions of DUSP4 protein and MMP2 and MMP9 proteins related to cell invasion and migration. The migration and invasion abilities of gastric cancer cells in vivo were evaluated according to the number of lung metastatic nodules in mice. RESULTS The expression of miR-122-5p in GC tissues and cells was significantly down-regulated, whereas DUSP4 expression was up-regulated. Bioinformatics prediction strategies and dual luciferase reporter assay verified the binding sites of miR-122-5p on 3'UTR of DUSP4 and the target relationship between miR-122-5p and DUSP4. Overexpression of miR-122-5p and knockdown of DUSP4 in BGC-823 cells observantly suppressed GC cell mobility and invasiveness, whereas downregulation of miR-122-5p expression promoted cell metastasis. MiR-122-5p inhibited GC cell mobility and invasiveness and pulmonary tumor metastasis via downregulation of DUSP4. CONCLUSION MiR-122-5p restrained migration and invasion abilities of GC cells by repressing DUSP4.
Collapse
Affiliation(s)
- Xiaofeng Xu
- a Department of Laboratory Medicine , Jingjiang People's Hospital , Jingjiang , Jiangsu , China
| | - Feng Gao
- b Department of Gastroenterology , Jingjiang People's Hospital , Jingjiang , Jiangsu , China
| | - Jianjiang Wang
- b Department of Gastroenterology , Jingjiang People's Hospital , Jingjiang , Jiangsu , China
| | - Lan Tao
- c Department of Central Laboratory , Jingjiang People's Hospital , Jingjiang , Jiangsu , China
| | - Jinsong Ye
- a Department of Laboratory Medicine , Jingjiang People's Hospital , Jingjiang , Jiangsu , China
| | - Li Ding
- a Department of Laboratory Medicine , Jingjiang People's Hospital , Jingjiang , Jiangsu , China
| | - Wei Ji
- a Department of Laboratory Medicine , Jingjiang People's Hospital , Jingjiang , Jiangsu , China
| | - Xing Chen
- d Department of Science and Education , Jingjiang People's Hospital , Jingjiang , Jiangsu , China
| |
Collapse
|
239
|
Abstract
Ubiquilin 1 (UBQLN1) plays an essential role in the regulation of protein degradations which is involved in the pathophysiology of neurodegenerative diseases and cancer. This study aimed to investigate the expression level of UBQLN1 in gastric cancer and evaluated the relationship between its expression and clinicopathological characteristics, as well as prognostic of patients with gastric cancer. Immunohistochemistry (IHC) was used to detect the expression levels of UBQLN1 in 179 pairs of gastric cancer and adjacent normal tissues. The UBQLN1 was significantly upregulated in gastric cancer tissue. High UBQLN1 expression was associated with high histological grade, invasion, lymph node metastasis, and tumor node metastasis (TNM) stage III (P < .001). Multivariate Cox analysis showed that larger tumor size (HR = 3.125, 95%CI: 2.031-4.808, P < .001), histological grade 3 (HR = 15.313, 95%,CI: 8.075-29.041, P < .001), pT3 + pT4 (HR = 3.224, 95%CI: 1.389-7.483, P = .006), LNM (HR = 4.467, 95%CI: 2.404-8.302, P < .001), TNM stage III (HR = 2.152, 95%CI: 1.289-3.594, P = .003), and high UBQLN1 expression (HR = 2.547, 95%CI: 1.511-4.292, P < .001) were significantly associated with worse prognosis of patients with gastric cancer. In conclusion, high UBQLN1 expression was an independent worse prognostic factor for patients with gastric cancer.
Collapse
Affiliation(s)
- Jingjing Bao
- Department of Pathology, Taizhou People's Hospital, Taizhou, Jiangsu Province
| | - Xiaoqin Jiang
- Department of Pathology, Taizhou People's Hospital, Taizhou, Jiangsu Province
| | - Xiaowei Zhu
- Department of Pathology, Taizhou People's Hospital, Taizhou, Jiangsu Province
| | - Guihong Dai
- Department of Pathology, Taizhou People's Hospital, Taizhou, Jiangsu Province
| | - Rongrong Dou
- Department of Pathology, Taizhou People's Hospital, Taizhou, Jiangsu Province
| | - Xinyun Liu
- Department of Pathology, Taizhou People's Hospital, Taizhou, Jiangsu Province
| | - Haihui Sheng
- Shanghai Engineering Center for Molecular Medicine, National Engineering Center for Biochip at Shanghai, Shanghai
| | - Zongmin Liang
- Intensive Care Unit, Taizhou People's Hospital, Taizhou, Jiangsu Province, China
| | - Hong Yu
- Department of Pathology, Taizhou People's Hospital, Taizhou, Jiangsu Province
| |
Collapse
|
240
|
Xie Y, Shao Y, Sun W, Ye G, Zhang X, Xiao B, Guo J. Downregulated expression of hsa_circ_0074362 in gastric cancer and its potential diagnostic values. Biomark Med 2017; 12:11-20. [PMID: 29240459 DOI: 10.2217/bmm-2017-0114] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
AIM To explore the diagnostic value of hsa_circ_0074362 in the screening of gastric cancer. METHODS The expression levels of hsa_circ_0074362 in 127 gastric cancer tissues and paired adjacent normal tissues, 83 gastritis tissues and six gastric cancer cell lines were first detected by quantitative reverse transcription-polymerase chain reaction. Then, the relationship between its levels and clinicopathological factors of patients with gastric cancer was analyzed. Finally, a receiver operating characteristic curve was established. RESULTS Hsa_circ_0074362 levels were significantly downregulated in gastric cancer tissues, gastritis tissues and gastric cancer cell lines. Its levels were associated with lymphatic metastasis. CONCLUSION Hsa_circ_0074362 probably plays a role in the initiation of gastric cancer and may be a potential biomarker of gastric cancer.
Collapse
Affiliation(s)
- Yi Xie
- Department of Biochemistry & Molecular Biology, & Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo 315211, PR China
| | - Yongfu Shao
- Department of Gastroenterology, the Affiliated Hospital of Medical School of Ningbo University, Ningbo 315020, PR China
| | - Weiliang Sun
- Ningbo Yinzhou People's Hospital & the Affiliated Hospital, Medical School of Ningbo University, Ningbo 315040, PR China
| | - Guoliang Ye
- Department of Gastroenterology, the Affiliated Hospital of Medical School of Ningbo University, Ningbo 315020, PR China
| | - Xinjun Zhang
- Department of Gastroenterology, the Affiliated Hospital of Medical School of Ningbo University, Ningbo 315020, PR China
| | - Bingxiu Xiao
- Department of Biochemistry & Molecular Biology, & Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo 315211, PR China
| | - Junming Guo
- Department of Biochemistry & Molecular Biology, & Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo 315211, PR China
| |
Collapse
|
241
|
Rugge M, Genta RM, Di Mario F, El-Omar EM, El-Serag HB, Fassan M, Hunt RH, Kuipers EJ, Malfertheiner P, Sugano K, Graham DY. Gastric Cancer as Preventable Disease. Clin Gastroenterol Hepatol 2017; 15:1833-1843. [PMID: 28532700 DOI: 10.1016/j.cgh.2017.05.023] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 04/26/2017] [Accepted: 05/16/2017] [Indexed: 02/07/2023]
Abstract
Gastric cancer, 1 of the 5 most common causes of cancer death, is associated with a 5-year overall survival rate less than 30%. A minority of cancers occurs as part of syndromic diseases; more than 90% of adenocarcinomas are considered as the ultimate consequence of a longstanding mucosal inflammation. Helicobacter pylori infection is the leading etiology of non-self-limiting gastritis, which may result in atrophy of the gastric mucosa and impaired acid secretion. Gastric atrophy establishes a field of cancerization prone to further molecular and phenotypic changes, possibly resulting in cancer growth. This well-understood natural history provides the clinicopathologic rationale for primary and secondary cancer prevention strategies. A large body of evidence demonstrates that combined primary (H pylori eradication) and secondary (mainly endoscopy) prevention efforts may prevent or limit the progression of gastric oncogenesis. This approach, which is tailored to different country-specific gastric cancer incidence, socioeconomic, and cultural factors, requires that the complementary competences of gastroenterologists, oncologists, and pathologists be amalgamated into a common strategy of health policy.
Collapse
Affiliation(s)
- Massimo Rugge
- Department of Medicine (DIMED), University of Padua, Padua, Italy; Veneto Tumor Registry, Veneto Region, Padua, Italy.
| | - Robert M Genta
- Miraca Life Sciences Research Institute, Irving, and Departments of Pathology and Medicine, Baylor College of Medicine, Houston, Texas
| | - Francesco Di Mario
- Department of Clinical and Experimental Medicine, University of Parma, Parma, Italy
| | - Emad M El-Omar
- St George and Sutherland Clinical School, University of New South Wales, Sydney, Australia
| | - Hashem B El-Serag
- Department of Medicine, Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston, Texas
| | - Matteo Fassan
- Department of Medicine (DIMED), University of Padua, Padua, Italy
| | - Richard H Hunt
- Division of Gastroenterology, Department of Medicine and Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada
| | - Ernst J Kuipers
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | | | - Kentaro Sugano
- Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - David Y Graham
- Department of Medicine, Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
242
|
Nishijima TF, Kardos J, Chai S, Smith CC, Bortone DS, Selitsky SR, Parker JS, Sanoff HK, Lee MS, Vincent BG. Molecular and Clinical Characterization of a Claudin-Low Subtype of Gastric Cancer. JCO Precis Oncol 2017; 1:1-10. [DOI: 10.1200/po.17.00047] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose Claudin-low molecular subtypes have been identified in breast and bladder cancers and are characterized by low expression of claudins, enrichment for epithelial-to-mesenchymal transition (EMT), and tumor-initiating cell (TIC) features. We evaluated whether the claudin-low subtype also exists in gastric cancer. Materials and Methods Four hundred fifteen tumors from The Cancer Genome Atlas (TCGA) gastric cancer mRNA data set were clustered on the claudin, EMT, and TIC gene sets to identify claudin-low tumors. We derived a 24-gene predictor that classifies gastric cancer into claudin-low and non–claudin-low subtypes. This predictor was validated with the Asian Cancer Research Group (ACRG) data set. We characterized molecular and clinical features of claudin-low tumors. Results We identified 46 tumors that had consensus enrichment for claudin-low features in TCGA data set. Claudin-low tumors were most commonly diffuse histologic type (82%) and originally classified as TCGA genomically stable (GS) subtype (78%). Compared with GS subtype, claudin-low subtype had significant activation in Rho family of GTPases signaling, which appears to play a key role in its EMT and TIC properties. In the ACRG data set, 28 of 300 samples were classified as claudin-low tumors by the 24-gene predictor and were phenotypically similar to the initially derived claudin-low tumors. Clinically, claudin-low subtype had the worst overall survival. Of note, the hazard ratios that compared claudin-low versus GS subtype were 2.10 (95% CI, 1.07 to 4.11) in TCGA and 2.32 (95% CI, 1.18 to 4.55) in the ACRG cohorts, with adjustment for age and pathologic stage. Conclusion We identified a gastric claudin-low subtype that carries a poor prognosis likely related to therapeutic resistance as a result of its EMT and TIC phenotypes.
Collapse
Affiliation(s)
| | - Jordan Kardos
- All authors: University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Shengjie Chai
- All authors: University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Christof C. Smith
- All authors: University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Dante S. Bortone
- All authors: University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Sara R. Selitsky
- All authors: University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Joel S. Parker
- All authors: University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Hanna K. Sanoff
- All authors: University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Michael S. Lee
- All authors: University of North Carolina at Chapel Hill, Chapel Hill, NC
| | | |
Collapse
|
243
|
Huang G, Li S, Yang N, Zou Y, Zheng D, Xiao T. Recent progress in circular RNAs in human cancers. Cancer Lett 2017; 404:8-18. [PMID: 28705773 DOI: 10.1016/j.canlet.2017.07.002] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Revised: 06/30/2017] [Accepted: 07/03/2017] [Indexed: 12/14/2022]
Abstract
Circular RNAs (circRNAs) are a large class of endogenous RNAs, formed by exon skipping or back-splicing events as covalently closed loops, which are expressed abundantly in mammalian cells. Although their biological functions remain largely unknown, recent studies show that circRNAs have three main functions in mammalian cells. First, circRNAs can regulate transcription and RNA splicing. Second, circRNAs function as microRNA (miRNA) sponges. Third, they can be translated into protein driven by N6-methyladenosine modification. Taking advantage of RNA sequencing (RNA-seq) technology, the expressions of circRNAs were found to be dysregulated in all types of cancer cell lines, tumor tissues, and even plasma samples from patients, which correlated with certain clinical characteristics, suggesting the potential roles of circRNAs in tumor progression. Considering their conserved sequences and stable structures, circRNAs were deemed to be promising biomarkers for the early diagnosis and prognosis prediction of cancer. In this review, we describe briefly the formation and properties of circRNAs, and focus mainly on recent progress in research into their function, regulation, and clinical relevance in different cancers.
Collapse
Affiliation(s)
- Guanqun Huang
- Shenzhen Key Laboratory of Translational Medicine of Tumor, Department of Cell Biology and Genetics, Shenzhen University Health Sciences Center, Shenzhen, Guangdong, China
| | - Shuaihu Li
- Shenzhen Key Laboratory of Translational Medicine of Tumor, Department of Cell Biology and Genetics, Shenzhen University Health Sciences Center, Shenzhen, Guangdong, China
| | - Nuo Yang
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, China
| | - Yongdong Zou
- Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, China
| | - Duo Zheng
- Shenzhen Key Laboratory of Translational Medicine of Tumor, Department of Cell Biology and Genetics, Shenzhen University Health Sciences Center, Shenzhen, Guangdong, China; National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, China.
| | - Tian Xiao
- Shenzhen Key Laboratory of Translational Medicine of Tumor, Department of Cell Biology and Genetics, Shenzhen University Health Sciences Center, Shenzhen, Guangdong, China.
| |
Collapse
|
244
|
Steccanella F, Costanzo A, Petrelli F. Editorial on role of p53 in esophageal cancer from a meta-analysis of 16 studies by Fisher et al.. J Thorac Dis 2017; 9:1450-1452. [PMID: 28740655 DOI: 10.21037/jtd.2017.05.72] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Francesca Steccanella
- Surgical Oncology Unit, Surgery Department, Oncology Department, ASST Bergamo Ovest, Treviglio (BG), Italy
| | - Antonio Costanzo
- Surgical Oncology Unit, Surgery Department, Oncology Department, ASST Bergamo Ovest, Treviglio (BG), Italy
| | - Fausto Petrelli
- Medical Oncology Unit, Oncology Department, ASST Bergamo Ovest, Treviglio (BG), Italy
| |
Collapse
|
245
|
Garattini SK, Basile D, Cattaneo M, Fanotto V, Ongaro E, Bonotto M, Negri FV, Berenato R, Ermacora P, Cardellino GG, Giovannoni M, Pella N, Scartozzi M, Antonuzzo L, Silvestris N, Fasola G, Aprile G. Molecular classifications of gastric cancers: Novel insights and possible future applications. World J Gastrointest Oncol 2017; 9:194-208. [PMID: 28567184 PMCID: PMC5434387 DOI: 10.4251/wjgo.v9.i5.194] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 12/04/2016] [Accepted: 03/17/2017] [Indexed: 02/05/2023] Open
Abstract
Despite some notable advances in the systemic management of gastric cancer (GC), the prognosis of patients with advanced disease remains overall poor and their chance of cure is anecdotic. In a molecularly selected population, a median overall survival of 13.8 mo has been reached with the use of human epidermal growth factor 2 (HER2) inhibitors in combination with chemotherapy, which has soon after become the standard of care for patients with HER2-overexpressing GC. Moreover, oncologists have recognized the clinical utility of conceiving cancers as a collection of different molecularly-driven entities rather than a single disease. Several molecular drivers have been identified as having crucial roles in other tumors and new molecular classifications have been recently proposed for gastric cancer as well. Not only these classifications allow the identification of different tumor subtypes with unique features, but also they serve as springboard for the development of different therapeutic strategies. Hopefully, the application of standard systemic chemotherapy, specific targeted agents, immunotherapy or even surgery in specific cancer subgroups will help maximizing treatment outcomes and will avoid treating patients with minimal chance to respond, therefore diluting the average benefit. In this review, we aim at elucidating the aspects of GC molecular subtypes, and the possible future applications of such molecular analyses.
Collapse
|
246
|
Fang R, Xu J, Lin H, Xu X, Tian F. The histone demethylase lysine-specific demethylase-1-mediated epigenetic silence of KLF2 contributes to gastric cancer cell proliferation, migration, and invasion. Tumour Biol 2017; 39:1010428317698356. [PMID: 28381185 DOI: 10.1177/1010428317698356] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Gastric cancer is one of the most common malignancies and leading causes of cancer-related death worldwide. An increasing number of evidence has revealed that gastric tumorigenesis is a multistage pathological state, and epigenetic alterations are considered to play critical roles in the etiology of gastric cancer. Lysine-specific demethylase-1, a histone demethylase, has been linked to malignancy in several human cancers and considered to epigenetically regulate many tumor suppressor genes during tumorigenesis and cancer progression. However, its role and underlying targets in gastric cancer are still unclear. In this study, we detected the lysine-specific demethylase-1 expression level in gastric cancer tissues and cell lines and investigated the function and mechanism of lysine-specific demethylase-1 in the gastric cancer. The in vitro analysis shows that knockdown of lysine-specific demethylase-1 significantly inhibits gastric cancer cell proliferation, migration, and invasion and induces cell cycle G1 phase arrest and cell apoptosis. In vivo assays determine that lysine-specific demethylase-1 downregulation represses gastric cancer cell tumorigenesis. Mechanistic investigation reveals that tumor suppressor KLF2 is a key downstream target of lysine-specific demethylase-1 in gastric cancer. These findings indicate that lysine-specific demethylase-1 is an important oncogene in gastric cancer, and lysine-specific demethylase-1-mediated epigenetic repression of KLF2 plays a critical role in gastric cancer development and progression, which supports lysine-specific demethylase-1 as a potential therapeutic target in this disease.
Collapse
Affiliation(s)
- Ruizhong Fang
- 1 Department of Hyperbaric Oxygen, Yishui Central Hospital, Linyi, People's Republic of China
| | - Jian Xu
- 2 Department of Gastroenterology, Yishui Central Hospital, Linyi, People's Republic of China
| | - Hai Lin
- 2 Department of Gastroenterology, Yishui Central Hospital, Linyi, People's Republic of China
| | - Xiaoguang Xu
- 2 Department of Gastroenterology, Yishui Central Hospital, Linyi, People's Republic of China
| | - Feng Tian
- 2 Department of Gastroenterology, Yishui Central Hospital, Linyi, People's Republic of China
| |
Collapse
|
247
|
Lee HS, Kim WH, Kwak Y, Koh J, Bae JM, Kim KM, Chang MS, Han HS, Kim JM, Kim HW, Chang HK, Choi YH, Park JY, Gu MJ, Lhee MJ, Kim JY, Kim HS, Cho MY, The Gastrointestinal Pathology Study Group of Korean Society of Pathologists, The Molecular Pathology Study Group of Korean Society of Pathologists. Molecular Testing for Gastrointestinal Cancer. J Pathol Transl Med 2017; 51:103-121. [PMID: 28219002 PMCID: PMC5357760 DOI: 10.4132/jptm.2017.01.24] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 01/16/2017] [Accepted: 01/24/2017] [Indexed: 12/20/2022] Open
Abstract
With recent advances in molecular diagnostic methods and targeted cancer therapies, several molecular tests have been recommended for gastric cancer (GC) and colorectal cancer (CRC). Microsatellite instability analysis of gastrointestinal cancers is performed to screen for Lynch syndrome, predict favorable prognosis, and screen patients for immunotherapy. The epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor has been approved in metastatic CRCs with wildtype RAS (KRAS and NRAS exon 2-4). A BRAF mutation is required for predicting poor prognosis. Additionally, amplification of human epidermal growth factor receptor 2 (HER2) and MET is also associated with resistance to EGFR inhibitor in metastatic CRC patients. The BRAF V600E mutation is found in sporadic microsatellite unstable CRCs, and thus is helpful for ruling out Lynch syndrome. In addition, the KRAS mutation is a prognostic biomarker and the PIK3CA mutation is a molecular biomarker predicting response to phosphoinositide 3-kinase/AKT/mammalian target of rapamycin inhibitors and response to aspirin therapy in CRC patients. Additionally, HER2 testing should be performed in all recurrent or metastatic GCs. If the results of HER2 immunohistochemistry are equivocal, HER2 silver or fluorescence in situ hybridization testing are essential for confirmative determination of HER2 status. Epstein-Barr virus-positive GCs have distinct characteristics, including heavy lymphoid stroma, hypermethylation phenotype, and high expression of immune modulators. Recent advances in next-generation sequencing technologies enable us to examine various genetic alterations using a single test. Pathologists play a crucial role in ensuring reliable molecular testing and they should also take an integral role between molecular laboratories and clinicians.
Collapse
Affiliation(s)
- Hye Seung Lee
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | - Woo Ho Kim
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | - Yoonjin Kwak
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | - Jiwon Koh
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | - Jeong Mo Bae
- Department of Pathology, SMG-SNU Boramae Medical Center, Seoul, Korea
| | - Kyoung-Mee Kim
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Mee Soo Chang
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
- Department of Pathology, SMG-SNU Boramae Medical Center, Seoul, Korea
| | - Hye Seung Han
- Department of Pathology, Konkuk University School of Medicine, Seoul, Korea
| | - Joon Mee Kim
- Department of Pathology, Inha University School of Medicine, Incheon, Korea
| | - Hwal Woong Kim
- Department of Pathology, Seegene Medical Foundation, Busan, Korea
| | - Hee Kyung Chang
- Department of Pathology, Kosin University Gospel Hospital, Kosin University College of Medicine, Busan, Korea
| | - Young Hee Choi
- Department of Pathology, Hallym University Dongtan Sacred Heart Hospital, Hwaseong, Korea
| | - Ji Y. Park
- Department of Pathology, Catholic University of Daegu School of Medicine, Daegu, Korea
| | - Mi Jin Gu
- Department of Pathology, Yeungnam University College of Medicine, Daegu, Korea
| | - Min Jin Lhee
- Department of Pathology, Seoul Red Cross Hospital, Seoul, Korea
| | - Jung Yeon Kim
- Department of Pathology, Inje University Sanggye Paik Hospital, Seoul, Korea
| | - Hee Sung Kim
- Department of Pathology, Chung-Ang University College of Medicine, Seoul, Korea
| | - Mee-Yon Cho
- Department of Pathology, Wonju Severance Christian Hospital, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - The Gastrointestinal Pathology Study Group of Korean Society of Pathologists
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
- Department of Pathology, SMG-SNU Boramae Medical Center, Seoul, Korea
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Department of Pathology, Konkuk University School of Medicine, Seoul, Korea
- Department of Pathology, Inha University School of Medicine, Incheon, Korea
- Department of Pathology, Seegene Medical Foundation, Busan, Korea
- Department of Pathology, Kosin University Gospel Hospital, Kosin University College of Medicine, Busan, Korea
- Department of Pathology, Hallym University Dongtan Sacred Heart Hospital, Hwaseong, Korea
- Department of Pathology, Catholic University of Daegu School of Medicine, Daegu, Korea
- Department of Pathology, Yeungnam University College of Medicine, Daegu, Korea
- Department of Pathology, Seoul Red Cross Hospital, Seoul, Korea
- Department of Pathology, Inje University Sanggye Paik Hospital, Seoul, Korea
- Department of Pathology, Chung-Ang University College of Medicine, Seoul, Korea
- Department of Pathology, Wonju Severance Christian Hospital, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - The Molecular Pathology Study Group of Korean Society of Pathologists
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
- Department of Pathology, SMG-SNU Boramae Medical Center, Seoul, Korea
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Department of Pathology, Konkuk University School of Medicine, Seoul, Korea
- Department of Pathology, Inha University School of Medicine, Incheon, Korea
- Department of Pathology, Seegene Medical Foundation, Busan, Korea
- Department of Pathology, Kosin University Gospel Hospital, Kosin University College of Medicine, Busan, Korea
- Department of Pathology, Hallym University Dongtan Sacred Heart Hospital, Hwaseong, Korea
- Department of Pathology, Catholic University of Daegu School of Medicine, Daegu, Korea
- Department of Pathology, Yeungnam University College of Medicine, Daegu, Korea
- Department of Pathology, Seoul Red Cross Hospital, Seoul, Korea
- Department of Pathology, Inje University Sanggye Paik Hospital, Seoul, Korea
- Department of Pathology, Chung-Ang University College of Medicine, Seoul, Korea
- Department of Pathology, Wonju Severance Christian Hospital, Yonsei University Wonju College of Medicine, Wonju, Korea
| |
Collapse
|
248
|
Gurzu S, Sugimura H, Orlowska J, Szederjesi J, Szentirmay Z, Bara T, Bara T, Fetyko A, Jung I. Proposal of a Dukes-MAC-like staging system for gastric cancer. J Investig Med 2017; 65:316-322. [PMID: 27634644 DOI: 10.1136/jim-2016-000270] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/22/2016] [Indexed: 02/05/2023]
Abstract
The aim of this study was to present an epidemiological update regarding the classical prognostic parameters of gastric cancer (GC) in 3 countries from Eastern Europe and to suggest a modification of the pTNM staging system. In 333 consecutive cases which were diagnosed between 2003 and 2012 in 3 departments of pathology from Romania, Hungary, and Poland, the following parameters were analyzed: age and gender of patients, tumor localization, macroscopic and microscopic aspects including the degree of discohesivity, depth of tumor infiltration, and pTNM stage. From all of the studied parameters, the following proved to have independent prognostic value, indicating a lower survival rate: presence of distant metastases (p=0.001), lymph node positivity (p=0.0009), depth of tumor infiltration (p=0.04), age over 50 (p=0.02), proximally located tumors (p=0.03), and ulceroinfiltrative or diffusely infiltrative macroscopic aspect (p=0.0002). The pT2N1-3 staged cases showed a worse prognosis compared with the pT3N0 ones (p=0.02). Regardless of depth of invasion, the lymph node status remains the strongest indicator of the survival rate in GC. The pTN staging system should be adapted and a Dukes-MAC-like staging system should include the following groups: stage A1-T1N0, stage A2-T1N1-3, stage B1-T2N0, stage B2-T2N1-3, stage C1-T3N0, stage C2-T3N1-3, and stage D-T4N0-3. The grade of discohesivity/budding is not a prognostic factor in GC.
Collapse
Affiliation(s)
- Simona Gurzu
- Department of Pathology, University of Medicine and Pharmacy, Tirgu Mures, Romania
| | - Haruhiko Sugimura
- Department of Tumor Pathology, Hamamatsu University, Hamamatsu, Japan
| | - Janina Orlowska
- Department of Pathology, The Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland
| | - Janos Szederjesi
- Department of Intensive Care, University of Medicine and Pharmacy, Tirgu Mures, Romania
| | - Zoltan Szentirmay
- Department of Molecular Pathology, National Institute of Oncology, Budapest, Hungary
| | - Tivadar Bara
- Department of Surgery, University of Medicine and Pharmacy, Tirgu Mures, Romania
| | - Tivadar Bara
- Department of Surgery, University of Medicine and Pharmacy, Tirgu Mures, Romania
| | - Ananmaria Fetyko
- Department of Pathology, University of Medicine and Pharmacy, Tirgu Mures, Romania
| | - Ioan Jung
- Department of Pathology, University of Medicine and Pharmacy, Tirgu Mures, Romania
| |
Collapse
|
249
|
Mi Y, Zhang D, Jiang W, Weng J, Zhou C, Huang K, Tang H, Yu Y, Liu X, Cui W, Zhang M, Sun X, Zhou Z, Peng Z, Zhao S, Wen Y. miR-181a-5p promotes the progression of gastric cancer via RASSF6-mediated MAPK signalling activation. Cancer Lett 2016; 389:11-22. [PMID: 28043911 DOI: 10.1016/j.canlet.2016.12.033] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 12/21/2016] [Accepted: 12/22/2016] [Indexed: 02/05/2023]
Abstract
We previously discovered that Ras association domain family member 6 (RASSF6) was downregulated and predicted poor prognosis in GC patients. However, the mechanisms of the down regulation of RASSF6 in GC remained unclear. Increasing evidence indicates that dysregulation of microRNAs promotes the progression of cancer through the repression of tumour suppressors. Here, we identified miR-181a-5p as a novel regulator of RASSF6 in GC. Functionally, ectopic expression or silencing of miR-181a-5p, respectively, promoted or inhibited GC cell proliferation, colony formation and cell cycle transition, as well as enhanced or prevented the invasion, metastasis of GC cells and epithelial to mesenchymal transition of GC cells in vitro and in vivo. Molecularly, miR-181a-5p functioned as an onco-miRNA by activating the RASSF6-regulated MAKP pathway. Overexpression or silencing of RASSF6 could partially reverse the effects of the overexpression or repression of miR-181a-5p on GC progress caused by activation of the MAKP pathway in vitro and in vivo. Clinically, high miR-181a-5p expression predicted poor survival in GC patients, especially combined with low RASSF6 expression. Collectively, we identified miR-181a-5p as an onco-miRNA, which acts by directly repressing RASSF6 in GC.
Collapse
Affiliation(s)
- Yushuai Mi
- Department of General Surgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Dongyuan Zhang
- Department of General Surgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Weiliang Jiang
- Department of Gastroenterology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Junyong Weng
- Department of General Surgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Chongzhi Zhou
- Department of General Surgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Kejian Huang
- Department of General Surgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Huamei Tang
- Department of Pathology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Yang Yu
- Department of General Surgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Xisheng Liu
- Department of General Surgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Weiyingqi Cui
- Department of Oncology, Linköping University, Linköping, Sweden; Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.
| | - Meng Zhang
- Department of Pathology, Shanghai Cancer Center, Fudan University, Shanghai, China.
| | - Xiaofeng Sun
- Department of Oncology, Linköping University, Linköping, Sweden; Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.
| | - Zongguang Zhou
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Zhihai Peng
- Department of General Surgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Senlin Zhao
- Department of General Surgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Yugang Wen
- Department of General Surgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
250
|
Juxtaposed genes in 7q21-22 amplicon contribute for two major gastric cancer sub-Types by mutual exclusive expression. Mol Carcinog 2016; 56:1239-1250. [DOI: 10.1002/mc.22586] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 10/23/2016] [Accepted: 10/28/2016] [Indexed: 12/29/2022]
|