1
|
Ebigbo A, Messmann H, Lee SH. Artificial Intelligence Applications in Image-Based Diagnosis of Early Esophageal and Gastric Neoplasms. Gastroenterology 2025:S0016-5085(25)00471-8. [PMID: 40043857 DOI: 10.1053/j.gastro.2025.01.253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 01/14/2025] [Accepted: 01/22/2025] [Indexed: 04/03/2025]
Abstract
Artificial intelligence (AI) holds the potential to transform the management of upper gastrointestinal (GI) conditions, such as Barrett's esophagus, esophageal squamous cell cancer, and early gastric cancer. Advancements in deep learning and convolutional neural networks offer improved diagnostic accuracy and reduced diagnostic variability across different clinical settings, particularly where human error or fatigue may impair diagnostic precision. Deep learning models have shown the potential to improve early cancer detection and lesion characterization, predict invasion depth, and delineate lesion margins with remarkable accuracy, all contributing to effective treatment planning. Several challenges, however, limit the broad application of AI in GI endoscopy, particularly in the upper GI tract. Subtle lesion morphology and restricted diversity in training datasets, which are often sourced from specialized centers, may constrain the generalizability of AI models in various clinical settings. Furthermore, the "black box" nature of some AI systems can impede explainability and clinician trust. To address these issues, efforts are underway to incorporate multimodal data, such as combining endoscopic and histopathologic imaging, to bolster model robustness and transparency. In the future, AI promises substantial advancements in automated real-time endoscopic guidance, personalized risk assessment, and optimized biopsy decision making. As it evolves, it would substantially impact not only early diagnosis and prognosis, but also the cost-effectiveness of managing upper GI diseases, ultimately leading to improved patient outcomes and more efficient health care delivery.
Collapse
Affiliation(s)
- Alanna Ebigbo
- Department of Gastroenterology, University Hospital Augsburg, Augsburg, Germany.
| | - Helmut Messmann
- Department of Gastroenterology, University Hospital Augsburg, Augsburg, Germany.
| | - Sung Hak Lee
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul, South Korea; Seoul St. Mary's Hospital, Seoul, South Korea.
| |
Collapse
|
2
|
Xue J, Qin S, Ren N, Guo B, Shi X, Jia E. Extracellular vesicle biomarkers in circulation for the diagnosis of gastric cancer: A systematic review and meta‑analysis. Oncol Lett 2023; 26:423. [PMID: 37664665 PMCID: PMC10472029 DOI: 10.3892/ol.2023.14009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 06/14/2023] [Indexed: 09/05/2023] Open
Abstract
The prognosis of a gastric cancer (GC) diagnosis is poor due to the current lack of effective early diagnostic methods. Extracellular vesicle (EV) biomarkers have previously demonstrated strong diagnostic efficiency for certain types of cancer, including pancreatic and lung cancer. The present review aimed to summarize the diagnostic value of circulating EV biomarkers for early stage GC. The PubMed, Medline and Web of Science databases were searched from May 1983 to September 18, 2022. All studies that reported the diagnostic performance of EV biomarkers for GC were included for analysis. Overall, 27 studies were selected containing 2,831 patients with GC and 2,117 controls. A total of 58 EV RNAs were reported in 26 studies, including 39 microRNAs (miRNAs), 10 long non-coding RNAs (lncRNAs), five circular RNAs, three PIWI-interacting RNAs and one mRNA, in addition to one protein in the remaining study. Meta-analysis of the aforementioned studies demonstrated that the pooled sensitivity, specificity and AUC value of the total RNAs were 84, 67% and 0.822, respectively. The diagnostic values of miRNAs were consistent with the total RNA, as the pooled sensitivity, specificity and AUC value were 84, 67% and 0.808, respectively. The pooled sensitivity, specificity and AUC values of lncRNAs were 89, 69% and 0.872, respectively, markedly higher compared with that of miRNAs. A total of five studies reported the diagnostic performance of EV RNA panels for early stage GC and reported powerful diagnostic values with a pooled sensitivity, specificity and AUC value of 80, 77% and 0.879, respectively. Circulating EV RNAs could have the potential to be used in the future as effective, noninvasive biomarkers for early GC diagnosis. Further research in this field is necessary to translate these findings into clinical practice.
Collapse
Affiliation(s)
- Jinru Xue
- Department of Thoracic Surgery, China-Japan Union Hospital of Jilin University, Jilin, Changchun 130000, P.R. China
| | - Shaoyou Qin
- Department of Gastroenterology, China-Japan Union Hospital of Jilin University, Jilin, Changchun 130000, P.R. China
| | - Na Ren
- Department of Thoracic Surgery, China-Japan Union Hospital of Jilin University, Jilin, Changchun 130000, P.R. China
| | - Bo Guo
- Department of Thoracic Surgery, China-Japan Union Hospital of Jilin University, Jilin, Changchun 130000, P.R. China
| | - Xianquan Shi
- Department of Ultrasound, Beijing Friendship Hospital of Capital Medical University, Beijing 100050, P.R. China
| | - Erna Jia
- Department of Gastroenterology, China-Japan Union Hospital of Jilin University, Jilin, Changchun 130000, P.R. China
| |
Collapse
|
3
|
Albahli S, Nazir T. A Circular Box-Based Deep Learning Model for the Identification of Signet Ring Cells from Histopathological Images. Bioengineering (Basel) 2023; 10:1147. [PMID: 37892876 PMCID: PMC10604551 DOI: 10.3390/bioengineering10101147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/16/2023] [Accepted: 09/18/2023] [Indexed: 10/29/2023] Open
Abstract
Signet ring cell (SRC) carcinoma is a particularly serious type of cancer that is a leading cause of death all over the world. SRC carcinoma has a more deceptive onset than other carcinomas and is mostly encountered in its later stages. Thus, the recognition of SRCs at their initial stages is a challenge because of different variants and sizes and illumination changes. The recognition process of SRCs at their early stages is costly because of the requirement for medical experts. A timely diagnosis is important because the level of the disease determines the severity, cure, and survival rate of victims. To tackle the current challenges, a deep learning (DL)-based methodology is proposed in this paper, i.e., custom CircleNet with ResNet-34 for SRC recognition and classification. We chose this method because of the circular shapes of SRCs and achieved better performance due to the CircleNet method. We utilized a challenging dataset for experimentation and performed augmentation to increase the dataset samples. The experiments were conducted using 35,000 images and attained 96.40% accuracy. We performed a comparative analysis and confirmed that our method outperforms the other methods.
Collapse
Affiliation(s)
- Saleh Albahli
- Department of Information Technology, College of Computer, Qassim University, Buraydah 51452, Saudi Arabia;
| | - Tahira Nazir
- Faculty of Computing, Riphah International University, Islamabad 44600, Pakistan
| |
Collapse
|
4
|
Huang Y, Liang L, Zhao YX, Yao BH, Zhang RM, Song L, Zhang ZT. RUNX2 Reverses p53-Induced Chemotherapy Resistance in Gastric Cancer. Pharmgenomics Pers Med 2023; 16:253-261. [PMID: 37009416 PMCID: PMC10065424 DOI: 10.2147/pgpm.s394393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 02/25/2023] [Indexed: 03/29/2023] Open
Abstract
Objective Gastric cancer is one of the most common malignancies worldwide; however, its overall mortality has not improved significantly over the last decade. Chemoresistance plays a critical role in this issue. This study aimed to clarify the role and mechanism of runt-related transcription factor 2 (RUNX2) in platinum-based chemotherapy resistance. Methods First, a drug-resistant model of gastric cancer cells was established to evaluate the relative expression level of the RUNX2 as a potential biomarker of chemotherapy resistance. Next, exogenous silencing was conducted to study whether RUNX2 could reverse drug resistance and understand the underlying mechanisms. Simultaneously, the correlation between the clinical outcomes of 40 patients after chemotherapy and the RUNX2 expression levels in tumor samples was analyzed. Results We discovered that RUNX2 was significantly expressed in drug-resistant gastric cancer cells and tissues; it was also reversibly resistant to transformation treatment by exogenous RUNX2 silencing. It is confirmed that RUNX2 negatively regulates the apoptosis pathway of the p53 to reduce the chemotherapeutic effects of gastric cancer. Conclusion RUNX2 is a possible target for platinum-based chemotherapy resistance.
Collapse
Affiliation(s)
- Yuan Huang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, People’s Republic of China
| | - Lu Liang
- Department of General Surgery, Baotou Central Hospital, Baotou, 014000, People’s Republic of Chin
| | - Yong-Xiang Zhao
- Department of Pediatrics and Urology Surgery, Baotou No.4 Hospital, Baotou, 014000, People’s Republic of China
| | - Bi-Hui Yao
- Department of General Surgery, Baotou Central Hospital, Baotou, 014000, People’s Republic of Chin
| | - Rui-Min Zhang
- Department of Pediatrics and Urology Surgery, Baotou No.4 Hospital, Baotou, 014000, People’s Republic of China
| | - Lei Song
- Department of General Surgery, Baotou Central Hospital, Baotou, 014000, People’s Republic of Chin
| | - Zhong-Tao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, People’s Republic of China
- Correspondence: Zhong-Tao Zhang, Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, No. 95 Yongan Road, Xicheng District, Beijing, 100050, People’s Republic of China, Tel +8613801060364, Email
| |
Collapse
|
5
|
Cao R, Tang L, Fang M, Zhong L, Wang S, Gong L, Li J, Dong D, Tian J. Artificial intelligence in gastric cancer: applications and challenges. Gastroenterol Rep (Oxf) 2022; 10:goac064. [PMID: 36457374 PMCID: PMC9707405 DOI: 10.1093/gastro/goac064] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/27/2022] [Accepted: 10/18/2022] [Indexed: 08/10/2023] Open
Abstract
Gastric cancer (GC) is one of the most common malignant tumors with high mortality. Accurate diagnosis and treatment decisions for GC rely heavily on human experts' careful judgments on medical images. However, the improvement of the accuracy is hindered by imaging conditions, limited experience, objective criteria, and inter-observer discrepancies. Recently, the developments of machine learning, especially deep-learning algorithms, have been facilitating computers to extract more information from data automatically. Researchers are exploring the far-reaching applications of artificial intelligence (AI) in various clinical practices, including GC. Herein, we aim to provide a broad framework to summarize current research on AI in GC. In the screening of GC, AI can identify precancerous diseases and assist in early cancer detection with endoscopic examination and pathological confirmation. In the diagnosis of GC, AI can support tumor-node-metastasis (TNM) staging and subtype classification. For treatment decisions, AI can help with surgical margin determination and prognosis prediction. Meanwhile, current approaches are challenged by data scarcity and poor interpretability. To tackle these problems, more regulated data, unified processing procedures, and advanced algorithms are urgently needed to build more accurate and robust AI models for GC.
Collapse
Affiliation(s)
| | | | - Mengjie Fang
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, P. R. China
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, P. R. China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, P. R. China
| | - Lianzhen Zhong
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, P. R. China
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, P. R. China
| | - Siwen Wang
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, P. R. China
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, P. R. China
| | - Lixin Gong
- College of Medicine and Biological Information Engineering School, Northeastern University, Shenyang, Liaoning, P. R. China
| | - Jiazheng Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Radiology Department, Peking University Cancer Hospital & Institute, Beijing, P. R. China
| | - Di Dong
- Corresponding authors. Di Dong, CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, 95 Zhongguancun East Road, Beijing 100190, P. R. China. Tel: +86-13811833760; ; Jie Tian, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, P. R. China. Tel: +86-10-82618465;
| | - Jie Tian
- Corresponding authors. Di Dong, CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, 95 Zhongguancun East Road, Beijing 100190, P. R. China. Tel: +86-13811833760; ; Jie Tian, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, P. R. China. Tel: +86-10-82618465;
| |
Collapse
|
6
|
Cui MY, Yi X, Zhu DX, Wu J. The Role of Lipid Metabolism in Gastric Cancer. Front Oncol 2022; 12:916661. [PMID: 35785165 PMCID: PMC9240397 DOI: 10.3389/fonc.2022.916661] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 05/23/2022] [Indexed: 11/20/2022] Open
Abstract
Gastric cancer has been one of the most common cancers worldwide with extensive metastasis and high mortality. Chemotherapy has been found as a main treatment for metastatic gastric cancer, whereas drug resistance limits the effectiveness of chemotherapy and leads to treatment failure. Chemotherapy resistance in gastric cancer has a complex and multifactorial mechanism, among which lipid metabolism plays a vital role. Increased synthesis of new lipids or uptake of exogenous lipids can facilitate the rapid growth of cancer cells and tumor formation. Lipids form the structural basis of biofilms while serving as signal molecules and energy sources. It is noteworthy that lipid metabolism is capable of inducing drug resistance in gastric cancer cells by reshaping the tumor micro-environment. In this study, new mechanisms of lipid metabolism in gastric cancer and the metabolic pathways correlated with chemotherapy resistance are reviewed. In particular, we discuss the effects of lipid metabolism on autophagy, biomarkers treatment and drug resistance in gastric cancer from the perspective of lipid metabolism. In brief, new insights can be gained into the development of promising therapies through an in-depth investigation of the mechanism of lipid metabolism reprogramming and resensitization to chemotherapy in gastric cancer cells, and scientific treatment can be provided by applying lipid-key enzyme inhibitors as cancer chemical sensitizers in clinical settings.
Collapse
Affiliation(s)
| | | | | | - Jun Wu
- *Correspondence: Jun Wu, ; Dan-Xia Zhu,
| |
Collapse
|
7
|
Budak C, Mençik V. Detection of ring cell cancer in histopathological images with region of interest determined by SLIC superpixels method. Neural Comput Appl 2022. [DOI: 10.1007/s00521-022-07183-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
8
|
Zhang Y, Liu C, Ma X, Xu L, Wang X, Wang X, Cao J, Ma A, Gao T. Effect of Dietary Modification on Gastric Mucosa, Gastrointestinal Symptoms and Nutritional Status of Patients With Early Gastric Cancer After Endoscopic Submucosal Dissection Surgery: A Retrospective Cohort Study. Front Nutr 2022; 9:741630. [PMID: 35399675 PMCID: PMC8984610 DOI: 10.3389/fnut.2022.741630] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 02/14/2022] [Indexed: 12/15/2022] Open
Abstract
Food is an important factor affecting the treatment of patients with early gastric cancer (EGC). We have established a hospital cohort to guide dietary patterns and observe the health status of patients with EGC after endoscopic submucosal dissection (ESD) after dietary modification. A total of 273 patients with EGC who underwent ESD were recruited to the cohort. They were given dietary instruction and education through a dietary manual and were followed up for 12 months. If the dietary pattern changed to the “traditional food” pattern (high consumption of vegetables, wheat products, and red meat) after the nutritional guidance, subjects were defined as the improvement diet group. Dietary patterns focused on “alcohol and fish” (drink a lot of wine and beer and eating freshwater and marine fish) or “coarse cereals” (mainly whole grains, beans and poultry) were the main ones in the unimproved diet group. The nutritional status, gastric mucosa, and gastrointestinal symptoms of the two groups of patients before and after the dietary instruction were compared. Compared with the unimproved diet group, the endoscopic performance score and the symptom score in the improved diet group were decreased by an average of 1.31 and 1.90, respectively. Except for lymphocyte count (P = 0.227), total protein (P < 0.000), albumin (P = 0.003), globulin (P = 0.014), red blood cell count (P < 0.000), and hemoglobin (P < 0.000) values were improved to varying degrees. After changing the diet, the intake of wheat products and vegetables in the improved diet group increased by 15.58 and 17.52%, respectively, while the intake of alcohol, fish, and pickled products was reduced by 43.36, 36.43, and 31.41%, respectively. After 1 year of dietary adjustment, the nutritional status, gastric mucosa, and gastrointestinal symptoms of patients with EGC after ESD eating the "traditional food" diet were all improved.
Collapse
Affiliation(s)
- Yebing Zhang
- Institute of Nutrition and Health, School of Public Health, Qingdao University, Qingdao, China
| | - Chengxia Liu
- Binzhou Medical University Hospital, Binzhou, China
| | - Xingbin Ma
- Binzhou Medical University Hospital, Binzhou, China
| | - Lei Xu
- Huai’an Center for Disease Control and Prevention, Huai’an, China
| | - Xiuhua Wang
- Binzhou Medical University Hospital, Binzhou, China
| | - Xin Wang
- Binzhou Medical University Hospital, Binzhou, China
| | - Jingrun Cao
- Binzhou Medical University Hospital, Binzhou, China
| | - Aiguo Ma
- Institute of Nutrition and Health, School of Public Health, Qingdao University, Qingdao, China
- *Correspondence: Aiguo Ma,
| | - Tao Gao
- Binzhou Medical University Hospital, Binzhou, China
- Tao Gao,
| |
Collapse
|
9
|
Feng XY, Xu X, Zhang Y, Xu YM, She Q, Deng B. Application of convolutional neural network in detecting and classifying gastric cancer. Artif Intell Gastrointest Endosc 2021. [DOI: 10.37126/aige.v2.i3.70] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
10
|
Feng XY, Xu X, Zhang Y, Xu YM, She Q, Deng B. Application of convolutional neural network in detecting and classifying gastric cancer. Artif Intell Gastrointest Endosc 2021; 2:71-78. [DOI: 10.37126/aige.v2.i3.71] [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: 04/27/2021] [Revised: 05/21/2021] [Accepted: 06/07/2021] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer (GC) is the fifth most common cancer in the world, and at present, esophagogastroduodenoscopy is recognized as an acceptable method for the screening and monitoring of GC. Convolutional neural networks (CNNs) are a type of deep learning model and have been widely used for image analysis. This paper reviews the application and prospects of CNNs in detecting and classifying GC, aiming to introduce a computer-aided diagnosis system and to provide evidence for subsequent studies.
Collapse
Affiliation(s)
- Xin-Yi Feng
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou 225000, Jiangsu Province, China
| | - Xi Xu
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou 225000, Jiangsu Province, China
| | - Yun Zhang
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou 225000, Jiangsu Province, China
| | - Ye-Min Xu
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou 225000, Jiangsu Province, China
| | - Qiang She
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou 225000, Jiangsu Province, China
| | - Bin Deng
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou 225000, Jiangsu Province, China
| |
Collapse
|
11
|
Hsiao YJ, Wen YC, Lai WY, Lin YY, Yang YP, Chien Y, Yarmishyn AA, Hwang DK, Lin TC, Chang YC, Lin TY, Chang KJ, Chiou SH, Jheng YC. Application of artificial intelligence-driven endoscopic screening and diagnosis of gastric cancer. World J Gastroenterol 2021; 27:2979-2993. [PMID: 34168402 PMCID: PMC8192292 DOI: 10.3748/wjg.v27.i22.2979] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/10/2021] [Accepted: 04/22/2021] [Indexed: 02/06/2023] Open
Abstract
The landscape of gastrointestinal endoscopy continues to evolve as new technologies and techniques become available. The advent of image-enhanced and magnifying endoscopies has highlighted the step toward perfecting endoscopic screening and diagnosis of gastric lesions. Simultaneously, with the development of convolutional neural network, artificial intelligence (AI) has made unprecedented breakthroughs in medical imaging, including the ongoing trials of computer-aided detection of colorectal polyps and gastrointestinal bleeding. In the past demi-decade, applications of AI systems in gastric cancer have also emerged. With AI's efficient computational power and learning capacities, endoscopists can improve their diagnostic accuracies and avoid the missing or mischaracterization of gastric neoplastic changes. So far, several AI systems that incorporated both traditional and novel endoscopy technologies have been developed for various purposes, with most systems achieving an accuracy of more than 80%. However, their feasibility, effectiveness, and safety in clinical practice remain to be seen as there have been no clinical trials yet. Nonetheless, AI-assisted endoscopies shed light on more accurate and sensitive ways for early detection, treatment guidance and prognosis prediction of gastric lesions. This review summarizes the current status of various AI applications in gastric cancer and pinpoints directions for future research and clinical practice implementation from a clinical perspective.
Collapse
Affiliation(s)
- Yu-Jer Hsiao
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Yuan-Chih Wen
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Medical Education, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Wei-Yi Lai
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei 112304, Taiwan
- Institute of Pharmacology, National Yang-Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Yi-Ying Lin
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei 112304, Taiwan
- Institute of Pharmacology, National Yang-Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Yi-Ping Yang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Internal Medicine, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Critical Center, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Yueh Chien
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | | | - De-Kuang Hwang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Institute of Clinical Medicine, National Yang-Ming Chiao Tung University, Taipei 112201, Taiwan
| | - Tai-Chi Lin
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Institute of Clinical Medicine, National Yang-Ming Chiao Tung University, Taipei 112201, Taiwan
| | - Yun-Chia Chang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Ting-Yi Lin
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Department of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Kao-Jung Chang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei 112304, Taiwan
- Institute of Clinical Medicine, National Yang-Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Shih-Hwa Chiou
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Institute of Pharmacology, National Yang-Ming Chiao Tung University, Taipei 112304, Taiwan
- Institute of Clinical Medicine, National Yang-Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Ying-Chun Jheng
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Big Data Center, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| |
Collapse
|
12
|
Guo X, Lv X, Ru Y, Zhou F, Wang N, Xi H, Zhang K, Li J, Chang R, Xie T, Wang X, Li B, Chen Y, Yang Y, Chen L, Chen L. Circulating Exosomal Gastric Cancer-Associated Long Noncoding RNA1 as a Biomarker for Early Detection and Monitoring Progression of Gastric Cancer: A Multiphase Study. JAMA Surg 2021; 155:572-579. [PMID: 32520332 PMCID: PMC7287948 DOI: 10.1001/jamasurg.2020.1133] [Citation(s) in RCA: 123] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Questions What role does circulating exosomal long noncoding RNA-GC1 (lncRNA-GC1) play in gastric cancer (GC), and does lncRNA-GC1 exhibit sufficient diagnostic performance for detecting early-stage GC and for monitoring disease progression? Findings In this multiphase study involving 826 participants, including patients with GC, patients with gastric precancerous lesions, and healthy donor individuals, circulating exosomal lncRNA-GC1 served as a noninvasive biomarker for detecting early-stage GC and for monitoring disease progression. Meaning For patients with gastric cancer, detection of circulating exosomal lncRNA-GC1 may improve the early diagnostic rate and monitor disease progression. Importance The gastric cancer (GC)–associated long noncoding RNA1 (lncRNA-GC1) plays an important role in gastric carcinogenesis. However, exosomal lncRNA-GC1 and its potential role in GC are poorly understood. Objective To evaluate the diagnostic value of circulating exosomal lncRNA-GC1 for early detection and monitoring progression of GC. Design, Setting, and Participants We performed a multiphase investigation of circulating exosomal lncRNA-GC1 for early detection of GC involving consecutive patients with GC (n = 522), patients with gastric precancerous lesions (n = 85), and healthy donor individuals (HDs; n = 219) from December 2016 to February 2019 at Chinese People’s Liberation Army General Hospital, China. LncRNA-GC1 was measured by reverse transcription–polymerase chain reaction by independent researchers who had no access to patients’ information. Receiver operating characteristic curves were used to calculate diagnostic efficiency in comparison between lncRNA-GC1 and 3 traditional biomarkers (carcinoembryonic antigen [CEA], cancer antigen 72-4 [CA72-4], and CA19-9). Main Outcomes and Measures Assessment of diagnostic efficiency on the basis of area under curve (AUC), specificity, and sensitivity. Results Of the 826 patients included in the study, 508 were men (61.5%), and the median age of all patients was 60 years (range, 28-82 years). In the test phase, lncRNA-GC1 achieved better diagnostic performance than the standard biomarkers CEA, CA72-4, and CA19-9 (AUC = 0.9033) for distinguishing between the patients with GC and HDs. Additionally, exosomal lncRNA-GC1 levels were significantly higher in culture media from GC cells compared with those of normal gastric epithelial cells (t = 5.310; P = .002). In the verification phase, lncRNA-GC1 retained its diagnostic efficiency in discriminating patients with GC from those with gastric precancerous lesions as well from HDs. Moreover, lncRNA-GC1 exhibited a higher AUC compared with those of CEA, CA72-4, and CA19-9 for early detection of GC with sufficient specificity and sensitivity, especially for patients with GC with negative standard biomarkers. Moreover, the levels of circulating exosomal lncRNA-GC1 were significantly associated with GC from early to advanced stages (HD vs stage I, t = 20.98; P < .001; stage I vs stage II, t = 2.787; P = .006; stage II vs stage III, t = 4.471; P < .001; stage III vs stage IV, t = 1.023; P = .30), independent of pathological grading and Lauren classification (pathological grading: HD vs G1, t = 21.09; P < .001; G1 vs G2, t = 0.3718; P = .71; G2 vs G3, t = 0.3598; P = .72; Lauren classification: t = 24.81; P <.001). In the supplemental phase, the levels of circulating exosomal lncRNA-GC1 were consistent with those in GC tissues and cells and were higher compared with those in normal tissues and cells. Furthermore, the levels of circulating lncRNA-GC1 were unchanged after exosomes were treated with RNase and remained constant after prolonged exposure to room temperature or after repeated freezing and thawing (t = 1.443; P = .39). Total circulating lncRNA-GC1 was nearly all packaged within exosomes rather than a free form in plasma. Conclusions and Relevence Circulating exosomal lncRNA-GC1 may serve as a noninvasive biomarker for detecting early-stage GC and for monitoring disease progression. Combining circulating exosomal lncRNA-GC1 detection with endoscopy could improve the early diagnostic rate of GC.
Collapse
Affiliation(s)
- Xin Guo
- Department of Endoscopic Surgery, Chinese People's Liberation Army 986th Hospital, Fourth Military Medical University, Shaanxi, China.,Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Xiaohui Lv
- Department of Gynecology and Obstetrics, Xijing Hospital, Fourth Military Medical University, Shaanxi, China
| | - Yi Ru
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Shaanxi, China
| | - Fuxing Zhou
- Department of Gynecology and Obstetrics, Xijing Hospital, Fourth Military Medical University, Shaanxi, China
| | - Ning Wang
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Hongqing Xi
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Kecheng Zhang
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Jiyang Li
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Rongyan Chang
- Department of Endoscopic Surgery, Chinese People's Liberation Army 986th Hospital, Fourth Military Medical University, Shaanxi, China
| | - Tianyu Xie
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Xinxin Wang
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Baohai Li
- Department of Ophthalmology, Chinese People's Liberation Army 305th Hospital, Nanjing, China
| | - Yong Chen
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Shaanxi, China
| | - Yanling Yang
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Shaanxi, China
| | - Lubin Chen
- Department of Endoscopic Surgery, Chinese People's Liberation Army 986th Hospital, Fourth Military Medical University, Shaanxi, China
| | - Lin Chen
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
13
|
Prognosis of Stage IIB Early Gastric Cancer Has a Unique and Dismal Property Putatively Requiring Postoperative Adjuvant Chemotherapy. Int Surg 2021. [DOI: 10.9738/intsurg-d-15-00177.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Pathological T1 (pT1) gastric cancer showed excellent prognosis; however, lymph node metastasis sometimes reflects patients with dismal prognosis. In this study, we investigated prognosis of pT1 gastric cancer with lymph node metastasis to identify prognostic factors. Among 1442 gastric cancer patients between 2002 and 2010, 73 (5%) of pT1 with lymph node metastasis were identified. Univariate prognostic factors were applied to multivariate Cox proportional hazards model. First, among the 1442 patients, pT1 was composed of 333 patients with pT1a and 423 patients with pT1b, which included 9 (2.7%) and 64 cases (15.1%) with lymph node metastasis, respectively. Secondly, 10 (13.7%) patients of the 73 patients with lymph node metastasis showed tumor relapse. Univariate negative prognostic factors were tumor size (P = 0.03), intraoperative bleeding (P = 0.03), and perioperative transfusion (POT; P = 0.001), as well as 14th JGCA (Japanese Gastric Cancer Association) Stage (P < 0.0001), and multivariate analysis identified 14th JGCA Stage (P = 0.0004) and POT (P = 0.03) as independent prognostic factors. Third, pT1 gastric cancer representing pN3 (Stage IIB) was rare (n = 4) and unique entity from a prognostic point of view, exhibiting dismal prognosis (0% at 5 years). We thereafter identified 17 such cases from 5204 gastric cancer cases including the earliest cases. Prognosis of the 17 patients was unique in that recurrences occurred even 5 years after curative operation, and the frequent recurrent sites were bone. pT1 gastric cancer prognosis is robustly affected by pN3 and POT, and Stage IIB disease showed unique prognosis requiring special attention even after 5 years of operation.
Collapse
|
14
|
Chen W, Yan H, Li X, Ge K, Wu J. Circulating tumor DNA detection and its application status in gastric cancer: a narrative review. Transl Cancer Res 2021; 10:529-536. [PMID: 35116282 PMCID: PMC8797971 DOI: 10.21037/tcr-20-2856] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 11/20/2020] [Indexed: 12/30/2022]
Abstract
Circulating tumor DNA (ctDNA) is the small genomic fragment released by tumor cells into the circulating system, which carries the gene variation features, such as mutation, insertion, deletion, rearrangement, copy number variation (CNV) and methylation, rendering it an important biomarker. It can be used not only to diagnose certain types of solid tumors, but also to monitor the therapeutic response and explore the minimal residual disease (MRD) and resistant mutation of targeted therapy. Therefore, ctDNA detection may become the preferred non-invasive tumor screening method. For patients who cannot receive further gene detection due to insufficient or restricted sample collection with the defined pathological diagnosis, ctDNA detection can be carried out to determine the gene mutation type, with no need for repeated sampling. Gastric cancer (GC) is a malignancy with extremely high morbidity and mortality, and its genesis and development are the consequence of interactions of multiple factors, including environment, diet, heredity, helicobacter pylori infection, chronic inflammatory infiltration, and precancerous lesion. As the research on GC moves forward, the existing research mainly focuses on genetic and epigenetic changes, including DNA methylation, histone modification, non-coding RNA changes, gene mutation, gene heterozygosity loss and microsatellite instability. This paper aimed to summarize the contents of ctDNA detection, its application status in GC and clinical significance.
Collapse
Affiliation(s)
- Wenyu Chen
- Department of Oncology, Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Haijiao Yan
- Department of Oncology, Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Xiaodong Li
- Department of Oncology, Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Kele Ge
- Department of Oncology, Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jun Wu
- Department of Oncology, Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
15
|
Prediction of novel miRNA biomarker candidates for diagnostic and prognostic analysis of STAD and LIHC: An integrated in silico approach. INFORMATICS IN MEDICINE UNLOCKED 2021. [DOI: 10.1016/j.imu.2021.100581] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
16
|
Iyer P, Moslim M, Farma JM, Denlinger CS. Diffuse gastric cancer: histologic, molecular, and genetic basis of disease. Transl Gastroenterol Hepatol 2020; 5:52. [PMID: 33073047 DOI: 10.21037/tgh.2020.01.02] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 01/15/2020] [Indexed: 12/24/2022] Open
Abstract
Diffuse gastric cancer (DGC) is a distinct histopathologic and molecular disease, characterized by mutations in CDH1, RHOA, and others. In addition, DGC is associated with familial syndromes, including hereditary DGC and germline mutation in CDH1. Clinically, this subtype of gastric adenocarcinoma is associated with a poor prognosis and possible resistance to available systemic therapies. An understanding of the genetic and molecular underpinnings of DGC may help inform of its clinical behavior and aid in screening, diagnosis, and response to treatment. In this review, we will review the current histologic, molecular, and genetic landscape of DGC and its relevance to clinical practice.
Collapse
Affiliation(s)
- Pritish Iyer
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Maitham Moslim
- Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Jeffrey M Farma
- Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Crystal S Denlinger
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| |
Collapse
|
17
|
Lee KF, Tsai MM, Tsai CY, Huang CG, Ou YH, Hsieh CC, Hsieh HL, Wang CS, Lin KH. DEK Is a Potential Biomarker Associated with Malignant Phenotype in Gastric Cancer Tissues and Plasma. Int J Mol Sci 2019; 20:5689. [PMID: 31766266 PMCID: PMC6888682 DOI: 10.3390/ijms20225689] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 11/08/2019] [Accepted: 11/09/2019] [Indexed: 12/13/2022] Open
Abstract
Gastric cancer (GC) is the second most widespread cause of cancer-related mortality worldwide. The discovery of novel biomarkers of oncoproteins can facilitate the development of therapeutic strategies for GC treatment. In this study, we identified novel biomarkers by integrating isobaric tags for relative and absolute quantitation (iTRAQ), a human plasma proteome database, and public Oncomine datasets to search for aberrantly expressed oncogene-associated proteins in GC tissues and plasma. One of the most significantly upregulated biomarkers, DEK, was selected and its expression validated. Our immunohistochemistry (IHC) (n = 92) and quantitative real-time polymerase chain reaction (qRT-PCR) (n = 72) analyses disclosed a marked increase in DEK expression in tumor tissue, compared with paired nontumor mucosa. Importantly, significantly higher preoperative plasma DEK levels were detected in GC patients than in healthy controls via enzyme-linked immunosorbent assay (ELISA). In clinicopathological analysis, higher expression of DEK in both tissue and plasma was significantly associated with advanced stage and poorer survival outcomes of GC patients. Data from receiver operating characteristic (ROC) curve analysis disclosed a better diagnostic accuracy of plasma DEK than carcinoembryonic antigen (CEA), carbohydrate antigen 19.9 (CA 19.9), and C-reactive protein (CRP), highlighting its potential as an effective plasma biomarker for GC. Plasma DEK is also more sensitive in tumor detection than the other three biomarkers. Knockdown of DEK resulted in inhibition of GC cell migration via a mechanism involving modulation of matrix metalloproteinase MMP-2/MMP-9 level and vice versa. Our results collectively support plasma DEK as a useful biomarker for making diagnosis and prognosis of GC patients.
Collapse
Affiliation(s)
- Kam-Fai Lee
- Department of Pathology, Chang Gung Memorial Hospital, Chiayi 613, Taiwan;
| | - Ming-Ming Tsai
- Department of Nursing, Division of Basic Medical Sciences, Chang-Gung University of Science and Technology, Taoyuan 333, Taiwan; (M.-M.T.); (H.-L.H.)
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan
- Department of General Surgery, Chang Gung Memorial Hospital, Chia-yi 613, Taiwan;
| | - Chung-Ying Tsai
- Department of Biochemistry, College of Medicine, Chang-Gung University, Taoyuan 333, Taiwan; (C.-Y.T.); (Y.-H.O.)
- Kidney Research Center and Department of Nephrology, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Chung-Guei Huang
- Department of Medical Biotechnology and Laboratory Science, and Graduate Institute of Biomedical Science, Chang Gung University, Taoyuan 333, Taiwan;
- Department of Laboratory Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan
| | - Yu-Hsiang Ou
- Department of Biochemistry, College of Medicine, Chang-Gung University, Taoyuan 333, Taiwan; (C.-Y.T.); (Y.-H.O.)
- Department of Laboratory Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan
| | - Ching-Chuan Hsieh
- Department of General Surgery, Chang Gung Memorial Hospital, Chia-yi 613, Taiwan;
| | - Hsi-Lung Hsieh
- Department of Nursing, Division of Basic Medical Sciences, Chang-Gung University of Science and Technology, Taoyuan 333, Taiwan; (M.-M.T.); (H.-L.H.)
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan
- Department of Neurology, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Chia-Siu Wang
- Department of General Surgery, Chang Gung Memorial Hospital, Chia-yi 613, Taiwan;
| | - Kwang-Huei Lin
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan
- Department of Biochemistry, College of Medicine, Chang-Gung University, Taoyuan 333, Taiwan; (C.-Y.T.); (Y.-H.O.)
- Liver Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan
| |
Collapse
|
18
|
Dong Z, Sun X, Xu J, Han X, Xing Z, Wang D, Ge J, Meng L, Xu X. Serum Membrane Type 1-Matrix Metalloproteinase (MT1-MMP) mRNA Protected by Exosomes as a Potential Biomarker for Gastric Cancer. Med Sci Monit 2019; 25:7770-7783. [PMID: 31619663 PMCID: PMC6820360 DOI: 10.12659/msm.918486] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Our previous research revealed that membrane type 1-matrix metalloproteinase (MT1-MMP) is overexpressed and plays a crucial role in gastric cancer (GC) progression. Exosomes are important for GC carcinogenesis, and the exosomal contents are ideal biomarkers. However, the expression of exosomal MT1-MMP mRNA in serum and its potential significance in GC remains unclear. Material/Methods The expression of exosomal MT1-MMP mRNA in serum of patients with GC, chronic gastritis, or atypical hyperplasia, and healthy controls was detected using real-time quantitative RT-PCR. Serum CEA, CA19-9, and CA72-4 were also measured by electrochemiluminescence assay. Results Exosomes were isolated and identified in serum, and serum exosomal MT1-MMP mRNA was found to be higher in patients with GC compared with healthy controls and patients with chronic gastritis or atypical hyperplasia (all P<0.05). Serum exosomal MT1-MMP mRNA was significantly correlated with tumor diameter, differentiation, Borrmann type, invasion depth, lymphatic metastasis, distal metastasis, and TNM stage. The AUC of exosomal MT1-MMP mRNA was 0.788 (95% CI: 0.714–0.850) with 63.9% sensitivity and 87.1% specificity, and was higher than that of CEA (0.655 (95% CI: 0.573–0.730)). The combination of 2 markers gave an AUC of 0.821 (95% CI: 0.750–0.878), which was better than with the individual marker. The sensitivity, specificity, and positive and negative predictive values were 75.6%, 83.9%, 94.7%, and 47.3%, respectively. Moreover, the multiple logistic regression model showed that tumor diameter, differentiation, invasion depth, and exosomal MT1-MMP mRNA were the risk factors for lymphatic metastasis in GC. Conclusions Our results characterized serum exosomal MT1-MMP mRNA in GC, providing a foundation for discovering serum exosomes-targeted biomarkers for GC diagnosis.
Collapse
Affiliation(s)
- Zhaogang Dong
- Department of Clinical Laboratory Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Xiaoyan Sun
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China (mainland)
| | - Jingjing Xu
- School of Microelectronics, Shandong University, Jinan, Shandong, China (mainland)
| | - Xia Han
- Department of Urology Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Zhaoquan Xing
- Department of Urology Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Ding Wang
- Department of Clinical Laboratory Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Jian Ge
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China (mainland)
| | - Liwei Meng
- Department of Urology Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Xiaofei Xu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| |
Collapse
|
19
|
Yoon HJ, Kim S, Kim JH, Keum JS, Oh SI, Jo J, Chun J, Youn YH, Park H, Kwon IG, Choi SH, Noh SH. A Lesion-Based Convolutional Neural Network Improves Endoscopic Detection and Depth Prediction of Early Gastric Cancer. J Clin Med 2019; 8:jcm8091310. [PMID: 31454949 PMCID: PMC6781189 DOI: 10.3390/jcm8091310] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 08/22/2019] [Accepted: 08/22/2019] [Indexed: 02/07/2023] Open
Abstract
In early gastric cancer (EGC), tumor invasion depth is an important factor for determining the treatment method. However, as endoscopic ultrasonography has limitations when measuring the exact depth in a clinical setting as endoscopists often depend on gross findings and personal experience. The present study aimed to develop a model optimized for EGC detection and depth prediction, and we investigated factors affecting artificial intelligence (AI) diagnosis. We employed a visual geometry group(VGG)-16 model for the classification of endoscopic images as EGC (T1a or T1b) or non-EGC. To induce the model to activate EGC regions during training, we proposed a novel loss function that simultaneously measured classification and localization errors. We experimented with 11,539 endoscopic images (896 T1a-EGC, 809 T1b-EGC, and 9834 non-EGC). The areas under the curves of receiver operating characteristic curves for EGC detection and depth prediction were 0.981 and 0.851, respectively. Among the factors affecting AI prediction of tumor depth, only histologic differentiation was significantly associated, where undifferentiated-type histology exhibited a lower AI accuracy. Thus, the lesion-based model is an appropriate training method for AI in EGC. However, further improvements and validation are required, especially for undifferentiated-type histology.
Collapse
Affiliation(s)
- Hong Jin Yoon
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Korea
| | - Seunghyup Kim
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Korea
| | - Jie-Hyun Kim
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Korea.
| | | | | | - Junik Jo
- SELVAS AI Inc., Seoul 08594, Korea
| | - Jaeyoung Chun
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Korea
| | - Young Hoon Youn
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Korea
| | - Hyojin Park
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Korea
| | - In Gyu Kwon
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Korea
| | - Seung Ho Choi
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Korea
| | - Sung Hoon Noh
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Korea
| |
Collapse
|
20
|
Wang C, Wang M, Xing B, Chi Z, Wang H, Lie C, Dong H. C-terminal of E1A binding protein 1 enhances the migration of gastric epithelial cells and has a clinicopathologic significance in human gastric carcinoma. Onco Targets Ther 2019; 12:5189-5200. [PMID: 31308691 PMCID: PMC6616302 DOI: 10.2147/ott.s203479] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 05/31/2019] [Indexed: 12/25/2022] Open
Abstract
Background Recent studies have claimed that the C-terminal of E1A binding proteins (CtBPs) influence tumorigenesis through participating in cell signal transduction in various human tumors. However, the detailed expression profiles of CtBP isoforms in human gastric cancer (GC) and the molecular mechanisms of CtBP involvement in tumor cell phenotypes warrant further investigation. Materials and methods The expression of CtBPs in GC cell lines and a human gastric epithelial cell line were explored via RT-qPCR and Western blotting assays. Moreover, the expression profiles of CtBPs in GC and histologically noncancerous tissues were explored by immunohistochemistry. To explore the effects of CtBP1 on the metastatic phenotype in GC, gastric epithelial cells were transfected with a eukaryotic expression plasmid to overexpress CTBP1, and the endogenous CtBP1 or JAK1 in GC cells was silenced through an RNA interference (RNAi) method. These transfections were validated via Western blotting, and the activation state of the JAK1/Stat3 signaling pathway was also explored via Western blotting. Furthermore, the malignant phenotype of GC cells was evaluated via a Cell Counting Kit-8 (CCK8) assay, colony formation assay, transwell assay, and wound-healing experiment. Results Our data revealed that the expression of CtBP1, but not CTBP2, was upregulated in 102 GC tissue samples compared with 98 noncancerous tissue samples, and the elevated expression level of CtBP1 was notably associated with distant metastasis. CTBP1 modulated cell migration and invasion through the JAK1/Stat3 signaling pathway in gastric epithelial cells. In addition, genetic silence of CtBP1 expression in GC cells notably constrained cell proliferation, invasion and migration abilities through inhibiting the activation of the JAK1/Stat3 pathway in GC cells. Conclusion Our data reveal that the knockout of CtBP1 notably constrains distant metastasis in GC through the JAK1/Stat3 pathway, suggesting that targeting CtBP1 is a practical anti-tumor approach to restrain tumor progression in GC.
Collapse
Affiliation(s)
- Can Wang
- Second Department of Gastrointestinal Surgery, Jilin Provincial Cancer Hospital, Changchun, Jilin 130012, People's Republic of China
| | - Min Wang
- Department of Pathology, Jilin Provincial Cancer Hospital, Changchun 130012, People's Republic of China
| | - Bocheng Xing
- Second Department of Gastrointestinal Surgery, Jilin Provincial Cancer Hospital, Changchun, Jilin 130012, People's Republic of China
| | - Zhaocheng Chi
- Second Department of Gastrointestinal Surgery, Jilin Provincial Cancer Hospital, Changchun, Jilin 130012, People's Republic of China
| | - Hongyu Wang
- Internal Medicine of Abdominal Tumors, Jilin Provincial Cancer Hospital, Changchun 130012, People's Republic of China
| | - Chunxiao Lie
- Second Department of Gastrointestinal Surgery, Jilin Provincial Cancer Hospital, Changchun, Jilin 130012, People's Republic of China
| | - Han Dong
- Department of Geriatric Medicine, First Hospital of Jilin University, Changchun, Jilin 130012, People's Republic of China
| |
Collapse
|
21
|
Iqbal M, Roberts A, Starr J, Mody K, Kasi PM. Feasibility and clinical value of circulating tumor DNA testing in patients with gastric adenocarcinomas. J Gastrointest Oncol 2019; 10:400-406. [PMID: 31183188 DOI: 10.21037/jgo.2019.01.14] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Gastric cancer is one of the leading causes of cancer worldwide, and this trend appears to be rising. Most patients are diagnosed at an advanced stage and thus prognosis is poor. Liquid biopsy, or circulating tumor DNA (ctDNA) testing, is emerging as a promising prognostic and/or predictive biomarker for patients with various types of malignancies. Its value and utility for patients with gastrointestinal malignancies, particularly gastric cancer is still being explored. There is ongoing research in other tumor types to suggest that ctDNA testing can be potentially used to identify tumor specific genomic alterations, predict tumor mutation burden, as well as help assess clinical response. We report on the feasibility and clinical value of ctDNA testing in patients with gastric cancers in a real time clinical context by reporting data on cohort of patients with gastric cancers (including those with gastroesophageal junction adenocarcinomas) treated at our institution.
Collapse
Affiliation(s)
- Madiha Iqbal
- Department of Hematology and Oncology, Mayo Clinic, Jacksonville, FL, USA
| | | | - Jason Starr
- Department of Hematology and Oncology, Mayo Clinic, Jacksonville, FL, USA
| | - Kabir Mody
- Department of Hematology and Oncology, Mayo Clinic, Jacksonville, FL, USA
| | | |
Collapse
|
22
|
S-1 combined with paclitaxel may benefit advanced gastric cancer: Evidence from a systematic review and meta-analysis. Int J Surg 2019; 62:34-43. [PMID: 30641155 DOI: 10.1016/j.ijsu.2018.11.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 10/13/2018] [Accepted: 11/07/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND Gastric cancer, as one of the increasingly common malignancies, has experienced high morbidity throughout many countries at present. Currently, chemotherapy regimen with more efficacy and safety for advanced gastric cancer (AGC) is needed. We aimed to assess the clinical efficacy and safety of S-1 combined with paclitaxel (PTX) for AGC by performing a systematic review and meta-analysis of the published studies. METHOD All published randomized controlled trials (RCTs) of S-1 combined with PTX for AGC were searched. Studies that included patients with locally advanced or metastases' gastric cancers were included. We searched the databases included Cochrane Library of Clinical Comparative Trials, MEDLINE, Embase, American Society of Clinical Oncology meeting abstracts and China National Knowledge Internet (CNKI) from 2000 to 2018. We searched the database up to January 2018. The first endpoint was overall survival (OS). Other endpoints were progression-free survival (PFS), objective response rate (ORR) and disease control rate (DCR). Safety analyses were also performed. RESULTS A total of 7 trials (including 1407 patients, 711 patients in intervention group and 696 patients in control group) were included in the present analysis. S-1 combined with PTX significantly improved the OS [HR = 0.78, 95% CI: 0.60-0.97, P = 0.000],PFS [HR = 0.70, 95% CI: 0.55-0.85, P = 0.000], ORR [RR = 1.30, 95%CI: 1.05-1.60, P = 0.017] and DCR [RR = 1.15, 95%CI: 1.04-1.27, P = 0.008] of patients with AGC. The grade 3 or 4 haematological and non-hematologic toxicities were anemia [RR = 1.71, 95% CI: 1.04-2.79, P = 0.03], neutropenia [RR = 1.65, 95% CI: 1.32-2.06, P < 0.0001] and anorexia [RR = 1.66, 95% CI: 1.05-2.64, P = 0.03] respectively. CONCLUSION S-1 combined with PTX may be a good choice for patients with AGC. S-1 plus PTX experienced more efficacy and safety when compared with S-1 alone or S-1 plus other drugs.
Collapse
|
23
|
Xu XC, Zhang WB, Li CX, Gao H, Pei Q, Cao BW, He TH. Up-Regulation of MiR-1915 Inhibits Proliferation, Invasion, and Migration of Helicobacter pylori-Infected Gastric Cancer Cells via Targeting RAGE. Yonsei Med J 2019; 60:38-47. [PMID: 30554489 PMCID: PMC6298885 DOI: 10.3349/ymj.2019.60.1.38] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 11/05/2018] [Accepted: 11/20/2018] [Indexed: 01/10/2023] Open
Abstract
PURPOSE Helicobacter pylori (HP)-infected gastric cancer (GC) is known to be a fatal malignant tumor, but the molecular mechanisms underlying its proliferation, invasion, and migration remain far from being completely understood. Our aim in this study was to explore miR-1915 expression and its molecular mechanisms in regulating proliferation, invasion, and migration of HP-infected GC cells. MATERIALS AND METHODS Quantitative real-time PCR and western blot analysis were performed to determine miR-1915 and receptor for advanced glycation end product (RAGE) expression in HP-infected GC tissues and gastritis tissues, as well as human gastric mucosal cell line GES-1 and human GC cell lines SGC-7901 and MKN45. CCK8 assay and transwell assay were performed to detect the proliferation, invasion, and migration capabilities. MiR-1915 mimics and miR-1915 inhibitor were transfected into GC cells to determine the target relationship between miR-1915 and RAGE. RESULTS MiR-1915 was under-expressed, while RAGE was over-expressed in HP-infected GC tissues and GC cells. Over-expressed miR-1915 could attenuate cellular proliferation, invasion, and migration capacities. RAGE was confirmed to be the target gene of miR-1915 by bioinformatics analysis and luciferase reporter assay. Moreover, HP-infected GC cellular proliferation, invasion, and migration were inhibited after treatment with pcDNA-RAGE. CONCLUSION MiR-1915 exerted tumor-suppressive effects on cellular proliferation, invasion, and migration of HP-infected GC cells via targeting RAGE, which provided an innovative target candidate for treatment of HP-infected GC.
Collapse
Affiliation(s)
- Xin Cai Xu
- Department of Gastrointestinal Tumor, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Wen Bin Zhang
- Department of Gastrointestinal Tumor, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China.
| | - Chun Xing Li
- Department of Gastrointestinal Tumor, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Hua Gao
- Department of Gastrointestinal Tumor, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Qi Pei
- Department of Gastrointestinal Tumor, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Bo Wei Cao
- Department of Gastrointestinal Tumor, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Tie Han He
- Department of Gastrointestinal Tumor, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
24
|
Qin SC, Zhao Z, Sheng JX, Xu XH, Yao J, Lu JJ, Chen B, Zhao GD, Wang XY, Yang YD. Dowregulation of OTX1 attenuates gastric cancer cell proliferation, migration and invasion. Oncol Rep 2018; 40:1907-1916. [PMID: 30066897 PMCID: PMC6111461 DOI: 10.3892/or.2018.6596] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Accepted: 07/17/2018] [Indexed: 12/16/2022] Open
Abstract
Orthodenticle homolog 1 (OTX1) has previously been revealed to be tightly associated with the development and progression of several human tumors. However, the functional roles and underlying molecular mechanisms of OTX1 in gastric cancer (GC) remain poorly understood. In the present study, we observed that OTX1 was highly expressed in GC tissues compared with adjacent non-tumor tissues based on a large cohort of samples from The Cancer Genome Atlas (TCGA) database. An immunohistochemical analysis indicated that OTX1 levels were increased in tumors that became metastatic compared with those in tumors that did not. This finding was significantly associated with patients who had shorter overall survival times. The knockdown of OTX1 significantly inhibited the proliferation, migration and invasion of SGC-7901 and MGC-803 cells. Furthermore, the knockdown of OTX1 induced cell cycle arrest in the G0/G1 phase and reduced the expression of cyclin D1. In addition, the inhibition of OTX1 led to increased GC cell apoptosis by upregulating cleaved PARP, cleaved caspase-3 and Bax. In conclusion, our data indicated that OTX1 functions as a key regulator in tumor growth and metastasis of GC cells. Thus, OTX1 may be a promising novel target for molecular therapy directed toward GC.
Collapse
Affiliation(s)
- Shi-Chen Qin
- Department of General Surgery, Haimen People's Hospital, Haimen, Jiangsu 226100, P.R. China
| | - Zhong Zhao
- Department of General Surgery, Haimen People's Hospital, Haimen, Jiangsu 226100, P.R. China
| | - Jin-Xin Sheng
- Department of General Surgery, Haimen People's Hospital, Haimen, Jiangsu 226100, P.R. China
| | - Xiang-Hui Xu
- Department of General Surgery, Haimen People's Hospital, Haimen, Jiangsu 226100, P.R. China
| | - Jie Yao
- Department of General Surgery, Haimen People's Hospital, Haimen, Jiangsu 226100, P.R. China
| | - Jin-Jun Lu
- Department of General Surgery, Haimen People's Hospital, Haimen, Jiangsu 226100, P.R. China
| | - Bin Chen
- Department of General Surgery, Haimen People's Hospital, Haimen, Jiangsu 226100, P.R. China
| | - Guo-Dong Zhao
- Department of General Surgery, Haimen People's Hospital, Haimen, Jiangsu 226100, P.R. China
| | - Xiao-Yong Wang
- Department of General Surgery, Haimen People's Hospital, Haimen, Jiangsu 226100, P.R. China
| | - Yan-Dong Yang
- Department of General Surgery, Haimen People's Hospital, Haimen, Jiangsu 226100, P.R. China
| |
Collapse
|
25
|
miR-589 promotes gastric cancer aggressiveness by a LIFR-PI3K/AKT-c-Jun regulatory feedback loop. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:152. [PMID: 30012200 PMCID: PMC6048856 DOI: 10.1186/s13046-018-0821-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 07/01/2018] [Indexed: 12/15/2022]
Abstract
Background As novel biomarkers for various cancers, microRNAs negatively regulate genes expression via promoting mRNA degradation and suppressing mRNA translation. miR-589 has been reported to be deregulated in several human cancer types. However, its biological role has not been functionally characterized in gastric cancer. Here, we aim to investigate the biological effect of miR-589 on gastric cancer and to reveal the possible mechanism. Methods Real-time PCR was performed to evaluate the expression of miR-589 in 34 paired normal and stomach tumor specimens, as well as gastric cell lines. Functional assays, such as wound healing, transwell assays and in vivo assays, were used to detect the biological effect of miR-589 and LIFR. We determined the role of miR-589 in gastric cancer tumorigenesis in vivo using xenograft nude models. Dual-luciferase report assays and Chromatin immunoprecipitation (ChIP) assay were performed for target evaluation, and the relationships were confirmed by western blot assay. Result MiR-589 expression was significantly higher in tumor tissues and gastric cancer cells than those in matched normal tissues and gastric epithelial cells, respectively. Clinically, overexpression of miR-589 is associated with tumor metastasis, invasion and poor prognosis of GC patients. Gain- and loss-of function experiments showed that miR-589 promoted cell migration, metastasis and invasion in vitro and lung metastasis in vivo. Mechanistically, we found that miR-589 directly targeted LIFR to activate PI3K/AKT/c-Jun signaling. Meanwhile, c-Jun bound to the promoter region of miR-589 and activated its transcription. Thus miR-589 regulated its expression in a feedback loop that promoted cell migration, metastasis and invasion. Conclusion Our study identified miR-589, as an oncogene, markedly induced cell metastasis and invasion via an atypical miR-589-LIFR-PI3K/AKT-c-Jun feedback loop, which suggested miR-589 as a potential biomarker and/or therapeutic target for the gastric cancer management. Electronic supplementary material The online version of this article (10.1186/s13046-018-0821-4) contains supplementary material, which is available to authorized users.
Collapse
|
26
|
Park SJ, Park YS, Jung IS, Yoon H, Shin CM, Ahn SH, Park DJ, Kim HH, Kim N, Lee DH. Is endoscopic surveillance necessary for patients who undergo total gastrectomy for gastric cancer? PLoS One 2018; 13:e0196170. [PMID: 29856747 PMCID: PMC5983473 DOI: 10.1371/journal.pone.0196170] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 04/06/2018] [Indexed: 01/29/2023] Open
Abstract
There have been only a few reports investigating the clinical efficacy of follow-up endoscopy for detection of recurrent gastric cancer after total gastrectomy (TG). We reviewed the records of 747 patients undergoing TG from 2003 to 2012 and enrolled 267 patients (70 with early gastric cancer (EGC) and 197 with advanced gastric cancer (AGC)), who received one or more follow-up endoscopy and contrast abdominal computed tomography (CT) scan. We found no tumor recurrence in the 70 EGC patients during the mean follow-up periods of 42.1 ± 18 and 43.2 ± 19 months by endoscopy and contrast abdominal CT scan. In 197 AGC patients, 59 patients (29.8%) had confirmed tumor recurrence during mean follow-up periods of 40.5 ± 21 and 45.3 ± 22 months. The most common pattern of tumor recurrence was distant metastasis (n = 35) followed by peritoneal metastasis (n = 11). Among the other 13 cases with loco-regional recurrence, seven cases were regional lymph node metastases, four were anastomosis site recurrences, and two were duodenal stump and jejunal loop site recurrences. Three of the four cases of anastomosis site recurrence were found by both endoscopy and contrast abdominal CT scan; one case was missed by contrast abdominal CT scan. However, the two cases with duodenal stump and jejunal loop recurrences were detected by contrast abdominal CT scan only. An annual follow-up endoscopy for gastric cancer after TG might have a limited role in the detection of tumor recurrence, especially in patients with EGC. Contrast abdominal CT scan may be sufficient as a follow-up method for recurrent gastric cancer after TG.
Collapse
Affiliation(s)
- Sung Jae Park
- Department of Internal medicine, Seoul National University Bundang Hospital, Seongnam-si, Republic of Korea
- Department of Internal medicine, Seoul Medical Center, Seoul, Republic of Korea
| | - Young Soo Park
- Department of Internal medicine, Seoul National University Bundang Hospital, Seongnam-si, Republic of Korea
- * E-mail:
| | - In Sub Jung
- Department of Internal medicine, Seoul National University Bundang Hospital, Seongnam-si, Republic of Korea
| | - Hyuk Yoon
- Department of Internal medicine, Seoul National University Bundang Hospital, Seongnam-si, Republic of Korea
| | - Cheol Min Shin
- Department of Internal medicine, Seoul National University Bundang Hospital, Seongnam-si, Republic of Korea
| | - Sang-Hoon Ahn
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam-si, Republic of Korea
| | - Do Joong Park
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam-si, Republic of Korea
| | - Hyung Ho Kim
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam-si, Republic of Korea
| | - Nayoung Kim
- Department of Internal medicine, Seoul National University Bundang Hospital, Seongnam-si, Republic of Korea
| | - Dong Ho Lee
- Department of Internal medicine, Seoul National University Bundang Hospital, Seongnam-si, Republic of Korea
| |
Collapse
|
27
|
Chen W, Yu Y, Yang N, Zhu J, Li K, Li R, Su W, Luo L, Hu L, Chen G, Deng H. Effects of Yangzheng Sanjie Decoction-containing serum mediated by microRNA-7 on cell proliferation and apoptosis in gastric cancer. Oncol Lett 2018; 15:3621-3629. [PMID: 29467883 PMCID: PMC5796316 DOI: 10.3892/ol.2018.7757] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 05/16/2017] [Indexed: 12/12/2022] Open
Abstract
Gastric cancer (GC) is one of the most common types of cancer and a leading cause of cancer-associated mortality. MicroRNAs (miRNAs/miRs) are demonstrated to function as oncomiRs or tumor-suppressor-miRs in GC. miR-7 has been identified to be a tumor suppressor of GC by targeting epidermal growth factor receptor (EGFR). In our previous study, Yangzheng Sanjie Decoction (YZSJD), a traditional Chinese formula, was identified to be effective in alleviating the symptoms and even postponing turnover of precancerous lesions. To elucidate the mechanism of YZSJD, the present study evaluated the effects of YZSJD of the GC MKN-45 cell line and investigated the underlying molecular mechanisms using YZSJD-containing serum (YCS). The expression of miR-7 in GC, normal and adjacent tissue samples was examined. The results demonstrated that YCS inhibited proliferation by inducing cell cycle arrest at the S phase, and significantly induced apoptosis compared with the control group. miR-7 was significantly downregulated in GC tissues compared with the matched ones. Using reverse transcription-quantitative polymerase chain reaction and western blot analysis, the expression of miR-7 was inversely associated with EGFR. This indicates that YCS inhibits proliferation and induces apoptosis of GC cells mediated by miR-7 targeting EGFR, which may be one of the mechanisms whereby YZSJD exerts its effects on GC.
Collapse
Affiliation(s)
- Wanqun Chen
- Department of Gastroenterology, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China.,Discipline of Integrated Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China.,Institute of Gastroenterology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Yaya Yu
- Department of Gastroenterology, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China.,Discipline of Integrated Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Naikun Yang
- Department of Gastroenterology, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China
| | - Jingli Zhu
- Department of Gastroenterology, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China
| | - Ke Li
- Pharmaceutical Research Institute, Hunan Academy of Chinese Medicine, Changsha, Hunan 410013, P.R. China
| | - Ruocun Li
- Pharmaceutical Research Institute, Hunan Academy of Chinese Medicine, Changsha, Hunan 410013, P.R. China
| | - Wenqiao Su
- Pharmaceutical Research Institute, Hunan Academy of Chinese Medicine, Changsha, Hunan 410013, P.R. China
| | - Lina Luo
- Department of Gastroenterology, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China.,Discipline of Integrated Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Ling Hu
- Institute of Gastroenterology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Gengxin Chen
- Department of Gastroenterology, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China.,Discipline of Integrated Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Haixia Deng
- Academy of Chinese Medical Sciences, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| |
Collapse
|
28
|
F-box protein 11 promotes the growth and metastasis of gastric cancer via PI3K/AKT pathway-mediated EMT. Biomed Pharmacother 2018; 98:416-423. [DOI: 10.1016/j.biopha.2017.12.088] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 12/03/2017] [Accepted: 12/18/2017] [Indexed: 11/17/2022] Open
|
29
|
Li Q, Qu F, Li R, He X, Zhai Y, Chen W, Zheng Y. A functional polymorphism of SSBP1 gene predicts prognosis and response to chemotherapy in resected gastric cancer patients. Oncotarget 2017; 8:110861-110876. [PMID: 29340022 PMCID: PMC5762290 DOI: 10.18632/oncotarget.22864] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 11/03/2017] [Indexed: 12/17/2022] Open
Abstract
Growing evidence has indicated that single-stranded DNA-binding proteins 1 (SSBP1) is involved in tumor initiation and progression. However, effects of single nucleotide polymorphisms (SNPs) in SSBP1 gene on gastric cancer (GC) prognosis are still unknown. In present study, two functional SNPs from SSBP1 were selected and genotyped in a large cohorts of 1030 resected GC patients (326 in the training set, 704 in the validation set) to explore the association of SNPs with patients’ survival. The rs6976500 G allele (CG/GG) genotypes were found significantly associated with both worse overall survival (OS) and recurrence-free survival (RFS) in the training and the independent validation set when compared to C allele genotype, which reaching a more robust statistical significance in the pooled analysis. Furthermore, integration of rs6976500 genotypes and TNM stage significantly improved the prognosis prediction models based on TNM stage alone. In addition, only carriers with at least one G allele of rs6976500 gained significant survival benefit from FOLFOX-based ACT. Mechanistically, SNP rs6976500 G allele genotype could significantly decrease promoter transcriptional activity and markedly reduce expression level of SSBP1 compared with the C allele genotype in GC cells. This was further substantiated by immunohistochemical assay in 70 GC tissue samples. Our study presents the first evidence that SNP rs6976500 G allele genotypes might contribute to GC prognosis by attenuating SSBP1 promoter activity and gene expression, and provides the guidance in refining therapeutic decisions of GC patients. Further exploration on its function is needed to clarify the exact biological mechanism behind.
Collapse
Affiliation(s)
- Qiuchen Li
- Department of Gastroenterology, First Affiliated Hospital of the Medical College, Shihezi University, Shihezi, Xinjiang, 832008, China
| | - Falin Qu
- Department of General Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Renli Li
- Department of General Surgery, The Fourth Hospital of Chinese PLA, Xining, Qinghai, 810007, China
| | - Xianli He
- Department of General Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Yulong Zhai
- Department of General Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Weigang Chen
- Department of Gastroenterology, First Affiliated Hospital of the Medical College, Shihezi University, Shihezi, Xinjiang, 832008, China
| | - Yong Zheng
- Department of Gastroenterology, First Affiliated Hospital of the Medical College, Shihezi University, Shihezi, Xinjiang, 832008, China
| |
Collapse
|
30
|
Sun ZP, Li AQ, Jia WH, Ye S, Van Eps G, Yu JM, Yang WJ. MicroRNA expression profiling in exosomes derived from gastric cancer stem-like cells. Oncotarget 2017; 8:93839-93855. [PMID: 29212193 PMCID: PMC5706839 DOI: 10.18632/oncotarget.21288] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 08/07/2017] [Indexed: 12/21/2022] Open
Abstract
Cancer stem-like cells (CSCs) have been identified as the initial cell in formation of cancer. Quiescent CSCs can "hide out" from traditional cancer therapy which may produce an initial response but are often unsuccessful in curing patients. Thus, levels of CSC in patients may be used as an indicator to measure the chance of recurrence of cancer after therapy. The goals of our work are to develop specific exosomal miRNA clusters for gastric CSCs that can potentially predict which patients are at high risk for developing gastric cancer (GC) in order to diagnose GC at an early stage. Here, upon sorting gastric CSCs, we initially isolated and characterized exosomes secreted by both gastric CSCs and their differentiated cells (DCs). By deep sequencing of each exosomal miRNA library, 11 typical differentially expressed miRNAs were identified as signature miRNAs for CSC. Gene target prediction, GO annotation and KEGG pathway enrichment analysis showed possible functions associated with these signature miRNAs. Hence, upon research of exosomal miRNAs that would influence behavior of tumor cells and their microenvironment, this study shows that a specific miRNA signature is present in CSCs, and implies that a potential miRNA biomarker reflecting the stage of gastric cancer progression and metastasis could be developed in the foreseeable future.
Collapse
Affiliation(s)
- Zhan-Peng Sun
- College of Life Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Conservation Biology for Endangered Wildlife of The Ministry of Education, Zhejiang University, Hangzhou, China
| | - An-Qi Li
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Wen-Huan Jia
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Sen Ye
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Grace Van Eps
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Jian-Min Yu
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Wei-Jun Yang
- College of Life Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Conservation Biology for Endangered Wildlife of The Ministry of Education, Zhejiang University, Hangzhou, China
| |
Collapse
|
31
|
Wang Z, Jin G, Wu Q, Wang R, Li Y. Immature colon carcinoma transcript-1 promotes proliferation of gastric cancer cells. Acta Biochim Biophys Sin (Shanghai) 2017; 49:979-988. [PMID: 29036264 DOI: 10.1093/abbs/gmx099] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Indexed: 11/14/2022] Open
Abstract
Gastric cancer is the fourth most common malignant tumor and has been considered as one of the leading causes of cancer-related death worldwide. The identification of the molecular mechanism during gastric cancer progression is urgently needed, which will help to develop more effective treatment strategies. As a component of the human mitoribosome, immature colon carcinoma transcript-1 (ICT1) might be involved in tumor formation and progression. However, its biological function and the corresponding mechanism in gastric cancer have been poorly characterized. To study the mechanism of ICT1 in gastric cancer, we first investigated the mRNA levels of ICT1 in human normal and gastric cancer tissues using datasets from the publicly available Oncomine database. The results showed that ICT1 is overexpressed in gastric cancer tissues. Then in order to study the role of ICT1 in gastric cancer, two shRNAs were used to silence ICT1 in MGC80-3 and AGS cells. Functional analysis showed ICT1 knockdown significantly inhibited the proliferation of gastric cancer cells and induced apoptosis. Further, mechanistic study demonstrated that ICT1 silencing induced cell-cycle arrest at G2/M phase via the suppression of cyclin A2 and cyclin B1. In addition, ICT1 silencing also increased cleaved caspase-3 and activated PARP in gastric cancer cells. These findings suggest that ICT1 may play a crucial role in promoting gastric cancer proliferation in vitro.
Collapse
Affiliation(s)
- Zishu Wang
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, China
| | - Gongsheng Jin
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, China
| | - Qiong Wu
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, China
| | - Rui Wang
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, China
| | - Yumei Li
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, China
| |
Collapse
|
32
|
Tao Y, Song Y, Han T, Wang C, Zhao T, Gu Y. miR-205 regulation of ICT1 has an oncogenic potential via promoting the migration and invasion of gastric cancer cells. Biomed Pharmacother 2017; 96:191-197. [PMID: 28987942 DOI: 10.1016/j.biopha.2017.09.147] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 09/21/2017] [Accepted: 09/26/2017] [Indexed: 01/16/2023] Open
Abstract
Immature colon carcinoma transcript-1 (ICT1) is a newly identified oncogene, which regulates mobility, apoptosis, cell cycle progression and proliferation of cancer cells. Nevertheless, the role of ICT1 and its clinical significance in gastric cancer (GC) is largely uncovered. Here, we found that ICT1 displayed higher expression in GC tissues compared to corresponding tumor-adjacent tissues. Further investigation confirmed ICT1 overexpression in GC cell lines. Clinical data disclosed that high ICT1 expression correlated with distant metastasis and advanced tumor-node-metastasis (TNM) stage. The Cancer Genome Atlas (TCGA) data further demonstrated that GC tissues with metastasis showed a significant higher level of ICT1 compared to those without metastasis. Furthermore, ICT1 overexpression notably predicted poor prognosis of GC patients. Functionally, we demonstrated that ICT1 knockdown suppressed invasion and migration of MGC-803 and BGC-823 cells in vitro. ICT1 overexpression promoted the mobility of SGC-7901 cells. Mechanistically, microRNA-205 (miR-205) was recognized as a direct down-regulator and inversely modulated ICT1 abundance in GC cells. miR-205 expression was down-regulated and negatively associated with ICT1 level in GC tissues. Underexpression of miR-205 indicated an obvious shorter survival of GC patients. miR-205 overexpression inhibited migration and invasion of MGC-803 cells, while these inhibitory effects were reversed by ICT1 restoration. Taken together, we have the earliest evidence that miR-205 regulation of ICT1 functions as an oncogene and prognostic biomarker in GC.
Collapse
Affiliation(s)
- Youmao Tao
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University,No.126 Xiantai Street, Changchun, Jilin Province 130033, China
| | - Yan Song
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University,No.126 Xiantai Street, Changchun, Jilin Province 130033, China
| | - Tao Han
- Department of Vascular Surgery, China-Japan Union Hospital of Jilin University,No.126 Xiantai Street, Changchun, Jilin Province 130033, China
| | - Changjing Wang
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University,No.126 Xiantai Street, Changchun, Jilin Province 130033, China
| | - Tongquan Zhao
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University,No.126 Xiantai Street, Changchun, Jilin Province 130033, China
| | - Ye Gu
- Department of Pediatric Surgery, The Second Hospital of Jilin University,No.218 Ziqiang Street, Changchun, Jilin Province 130022, China.
| |
Collapse
|
33
|
Ma DH, Li BS, Liu JJ, Xiao YF, Yong X, Wang SM, Wu YY, Zhu HB, Wang DX, Yang SM. miR-93-5p/IFNAR1 axis promotes gastric cancer metastasis through activating the STAT3 signaling pathway. Cancer Lett 2017; 408:23-32. [PMID: 28842285 DOI: 10.1016/j.canlet.2017.08.017] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Revised: 08/11/2017] [Accepted: 08/13/2017] [Indexed: 01/23/2023]
Abstract
Aberrant expression of microRNAs (miRNAs) plays an important role in gastric cancer (GC) development. miR-93-5p has shown opposing functions in different types of cancers, but the exact expression pattern and molecular mechanism of miR-93-5p in GC development remain to be elucidated. Here, we reported that miR-93-5p expression was increased in GC tissues compared with the adjacent normal tissues and that its overexpression was correlated with distant metastasis and poor survival in GC patients. miR-93-5p knockdown inhibited the migration, invasion and proliferation of GC cells in vitro and in vivo, while its overexpression displayed an opposite result. Using an mRNA microarray, we found that miR-93-5p significantly downregulated IFNAR1 expression in GC cells, which was further identified as a direct target of miR-93-5p. IFNAR1 knockdown promoted GC cell migration and invasion, but its restoration could rescue GC cell migration and invasion induced by miR-93-5p overexpression. Moreover, miR-93-5p-IFNAR1 axis increased MMP9 expression via STAT3 pathway in GC cells. Taken together, we reveal that miR-93-5p overexpression is associated with the poor survival of GC patients and miR-93-5p-IFNAR1 axis promotes GC metastasis through activation of STAT3 pathway.
Collapse
Affiliation(s)
- Dong-Hong Ma
- Department of Gastroenterology, No. 254 Hospital of PLA, Tianjin, 300142, PR China
| | - Bo-Sheng Li
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, PR China
| | - Jing-Jing Liu
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, PR China
| | - Yu-Feng Xiao
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, PR China
| | - Xin Yong
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, PR China
| | - Shu-Ming Wang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, PR China
| | - Yu-Yun Wu
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, PR China
| | - Hong-Bin Zhu
- Department of Gastroenterology, No. 254 Hospital of PLA, Tianjin, 300142, PR China
| | - Dong-Xu Wang
- Department of Gastroenterology, No. 254 Hospital of PLA, Tianjin, 300142, PR China.
| | - Shi-Ming Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, PR China.
| |
Collapse
|
34
|
Jin J, Lv H, Wu J, Li D, Chen K, Zhang F, Han J, Feng J, Zhang N, Yu H, Su D, Ying L. Regenerating Family Member 4 (Reg4) Enhances 5-Fluorouracil Resistance of Gastric Cancer Through Activating MAPK/Erk/Bim Signaling Pathway. Med Sci Monit 2017; 23:3715-3721. [PMID: 28759561 PMCID: PMC5549713 DOI: 10.12659/msm.903134] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 01/19/2017] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Reg4, a member of the Reg multigene family, is highly upregulated in many gastrointestinal cancers including gastric cancer (GC). The enhanced expression of Reg4 is associated with the resistance of GC to 5-fluorouracil (5-FU), while the underlying mechanism is not clear. The aim of the present study was to explore the resistant mechanism underlying 5-FU resistance. MATERIAL AND METHODS Reg4 expression was assessed by Western blot analysis for SGC-7901, BGC-823, AGS, MKN28, and MKN45. Synthetic short single strand RNA oligonucleotides and Flag-Reg4 plasmid were used to investigate the biological function of Reg4 in vitro. The cell viability assay was performed by MTT. Flow cytometry was carried out to measure the apoptosis caused by 5-FU. Reverse-transcriptase quantitative polymerase chain reaction (RT-qPCR) was used to examine the expression of 5-FU metabolism related enzymes. The effect of Reg4 on intracellular signaling was evaluated by Western blot. RESULTS Western blot analysis of 5 GC cells showed that Reg4 was low or null in SGC-7901 and BGC-823, while high in AGS, MKN28, and MKN45. Over-expression of flag-Reg4 in SGC-7901 led to an increase in cell viability and lower apoptosis with 5-FU treatment. In contrast, siRNA knockdown of Reg4 enhanced 5-FU induced apoptosis. However, over-expression or knockdown of Reg4 had no significant influence on the expression of 5-FU metabolic enzymes. Further investigation revealed that Reg4 could activate Erk1/2-Bim-caspase3 cascade. CONCLUSIONS Reg4 inhibited apoptosis through regulating MAPK/Erk/Bim signaling pathway and thereby enhanced the resistance of GC to 5-FU.
Collapse
Affiliation(s)
- Jiaoyue Jin
- Cancer Research Institute, Zhejiang Cancer Hospital and Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology of Zhejiang Province, Hangzhou, Zhejiang, P.R. China
| | - Hang Lv
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Diagnosis and Treatment of Digestive System Tumor, Zhejiang Provincial Hospital of Traditional Chinese Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P.R. China
| | - Junzhou Wu
- Cancer Research Institute, Zhejiang Cancer Hospital and Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology of Zhejiang Province, Hangzhou, Zhejiang, P.R. China
| | - Dan Li
- Cancer Research Institute, Zhejiang Cancer Hospital and Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology of Zhejiang Province, Hangzhou, Zhejiang, P.R. China
| | - Kaiyan Chen
- Cancer Research Institute, Zhejiang Cancer Hospital and Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology of Zhejiang Province, Hangzhou, Zhejiang, P.R. China
| | - Fanrong Zhang
- Cancer Research Institute, Zhejiang Cancer Hospital and Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology of Zhejiang Province, Hangzhou, Zhejiang, P.R. China
| | - Jing Han
- Tissue Bank, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, P.R. China
| | - Jianguo Feng
- Cancer Research Institute, Zhejiang Cancer Hospital and Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology of Zhejiang Province, Hangzhou, Zhejiang, P.R. China
| | - Nan Zhang
- Cancer Research Institute, Zhejiang Cancer Hospital and Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology of Zhejiang Province, Hangzhou, Zhejiang, P.R. China
| | - Herbert Yu
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, U.S.A
| | - Dan Su
- Cancer Research Institute, Zhejiang Cancer Hospital and Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology of Zhejiang Province, Hangzhou, Zhejiang, P.R. China
| | - Lisha Ying
- Cancer Research Institute, Zhejiang Cancer Hospital and Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology of Zhejiang Province, Hangzhou, Zhejiang, P.R. China
| |
Collapse
|
35
|
Tao Y, Sun C, Zhang T, Song Y. SMURF1 promotes the proliferation, migration and invasion of gastric cancer cells. Oncol Rep 2017; 38:1806-1814. [PMID: 28731194 DOI: 10.3892/or.2017.5825] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 07/06/2017] [Indexed: 11/06/2022] Open
Abstract
Smad ubiquitin regulatory factor 1 (SMURF1), a well-known E3 ubiquitin ligase, targets substrate proteins for ubiquitination and proteasomal degradation. Accumulating studies have shown that SMURF1 acts as an oncogenic factor in human malignancies. However, the clinical significance of SMURF1 and its role in gastric cancer (GC) remain unclear. The expression of SMURF1 was detected in 68 cases of GC and corresponding tumor-adjacent specimens. Our results revealed that SMURF1 was prominently overexpressed in GC specimens compared to corresponding tumor-adjacent tissues. Furthermore, increased levels of SMURF1 mRNA were also observed in GC cell lines. Clinicopathological detection ascertained that SMURF1-positive expression was associated with large tumor size, more lymph nodes and distant metastasis as well as advanced tumor-node-metastasis (TNM) stage of GC. Notably, GC patients with SMURF1 positive‑expressing tumors exhibited a significant decreased survival. Further experiments illustrated that SMURF1 knockdown significantly inhibited proliferation, migration and invasion of MGC-803 cells, while SMURF1 overexpression prominently promoted these behaviors in SGC-7901 cells. In vivo studies revealed that SMURF1 knockdown markedly inhibited tumor growth and liver metastasis of GC. Mechanically, SMURF1 inversely regulated the expression of DOC-2/DAB2 interactive protein (DAB2IP) in GC tissues and cells. Furthermore, DAB2IP restoration revealed similar effects to SMURF1 knockdown on MGC-803 cells with decreased proliferation, migration and invasion. In addition, the PI3K/Akt pathway and its downstream targets including c-Myc and ZEB1 were potentially involved in the oncogenic role of the SMURF1/DABIP axis. Collectively, the present study revealed the first evidence that SMURF1 can be potentially used as a clinical biomarker and target for novel treatment of human GC.
Collapse
Affiliation(s)
- Youmao Tao
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital, Jilin University, Changchun, Jilin 130033, P.R. China
| | - Caixia Sun
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital, Jilin University, Changchun, Jilin 130033, P.R. China
| | - Tao Zhang
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital, Jilin University, Changchun, Jilin 130033, P.R. China
| | - Yan Song
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital, Jilin University, Changchun, Jilin 130033, P.R. China
| |
Collapse
|
36
|
Jing X, Cui X, Liang H, Hao C, Han C. Diagnostic accuracy of ELISA for detecting serum Midkine in cancer patients. PLoS One 2017; 12:e0180511. [PMID: 28686647 PMCID: PMC5501560 DOI: 10.1371/journal.pone.0180511] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 06/18/2017] [Indexed: 12/21/2022] Open
Abstract
Midkine (MK) has been reported as the potential novel diagnostic biomarker for cancer in several studies, but their results were controversial. Therefore, we performed a diagnostic meta-analysis to assess the diagnostic value of serum MK in cancer patients. A systematic electronic and manual search was performed for relevant literatures through several databases up to June 1, 2017. The quality of the studies included in the meta-analysis was assessed using the Quality Assessment of Diagnostic Accuracy Studies (QUADAS-2) tool. All analyses were conducted using stata12.0 software. Ten studies collectively included 1119 cancer patients and 1441 controls met the eligible criteria. The summary estimates were: sensitivity 0.78 (95% CI = 0.68-0.85), specificity 0.83 (95% CI = 0.72-0.90), positive likelihood ratio 4,54 (95% CI = 2.64-7.80), negative likelihood 0.27 (95% CI = 0.18-0.40), diagnostic odds ratio 16.79 (95% CI = 7.17-39.33), and area under the curve 0.87 (95% CI = 0.84-0.89). Publication bias was suggested by Deeks' funnel plot asymmetry test (P = 0.92). According to our results, serum MK has greater diagnostic value in diagnosing cancer, however, more reliable studies in larger cohort should be conducted to evaluate the diagnostic accuracy of serum MK.
Collapse
Affiliation(s)
- Xuan Jing
- Department of Clinical Laboratory, Shanxi Provincial People's Hospital, Affiliate of Shanxi Medical University, Taiyuan, P.R. China
- * E-mail:
| | - Xiangrong Cui
- Reproductive Medicine Center, Children's Hospital of Shanxi and Women Health Center of Shanxi, Affiliate of Shanxi Medical University, Taiyuan, Shanxi, P.R. China
| | - Hongping Liang
- Department of Clinical Laboratory, Shanxi Provincial People's Hospital, Affiliate of Shanxi Medical University, Taiyuan, P.R. China
| | - Chonghua Hao
- Department of Clinical Laboratory, Shanxi Provincial People's Hospital, Affiliate of Shanxi Medical University, Taiyuan, P.R. China
| | - Chongyang Han
- Department of Nephrology, Shanxi Provincial People's Hospital, Affiliate of Shanxi Medical University, Taiyuan, P.R. China
| |
Collapse
|
37
|
Zheng L, Chen J, Zhou Z, He Z. Knockdown of long non-coding RNA HOXD-AS1 inhibits gastric cancer cell growth via inactivating the JAK2/STAT3 pathway. Tumour Biol 2017; 39:1010428317705335. [PMID: 28475004 DOI: 10.1177/1010428317705335] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Long non-coding RNA HOXD-AS1 (HOXD cluster antisense RNA 1) has been demonstrated to be closely associated with the progression of several tumors. However, the biological function of HOXD-AS1 and the underlying molecular mechanism in gastric cancer are still unclear. The expression of HOXD-AS1 in gastric cancer cell lines was evaluated by quantitative real-time polymerase chain reaction. The association of HOXD-AS1 expression and clinical parameters was statistically analyzed by chi-square test. Cell viability, colony formation capacity, and phosphorylation of Janus kinase 2 and signal transducer and activator of transcription 3 in treated SGC-7901 and BGC-823 cells were detected by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, colony formation assay, and western blot analysis, respectively. The results indicated that HOXD-AS1 was significantly upregulated in gastric cancer cells and clinically involved in tumor size, invasion depth, tumor-node-metastasis stages, regional lymph nodes, lymphatic metastasis, as well as distant metastasis. HOXD-AS1 knockdown dramatically inhibited gastric cancer cell proliferation, colony formation capacity, and phosphorylation of Janus kinase 2 and signal transducer and activator of transcription 3 in vitro. In addition, HOXD-AS1 overexpression significantly promoted gastric cancer cell proliferation and colony formation capacity, whereas both Janus kinase small interfering RNAs and Janus kinase 2 inhibitor AG490 overturned these effects. Furthermore, xenograft assays confirmed the biological function of HOXD-AS1 in vivo. Taken together, our data elucidate that knockdown of HOXD-AS1 dramatically suppresses gastric cancer cell growth by inactivating the Janus kinase 2/signal transducer and activator of transcription 3 pathway in vitro and in vivo, contributing to a better understanding of gastric cancer pathogenesis and providing a possible theoretical foundation for long non-coding RNA-directed diagnosis and therapy against this disease.
Collapse
Affiliation(s)
- Li Zheng
- Department of General Surgery, Huaihe Hospital of Henan University, Kaifeng, China
| | - Jiangtao Chen
- Department of General Surgery, Huaihe Hospital of Henan University, Kaifeng, China
| | - Zhongyong Zhou
- Department of General Surgery, Huaihe Hospital of Henan University, Kaifeng, China
| | - Zhikuan He
- Department of General Surgery, Huaihe Hospital of Henan University, Kaifeng, China
| |
Collapse
|
38
|
Shen L, Dong X, Yu M, Luo Z, Wu S. β3GnT8 Promotes Gastric Cancer Invasion by Regulating the Glycosylation of CD147. J Cancer 2017; 8:314-322. [PMID: 28243336 PMCID: PMC5327381 DOI: 10.7150/jca.16526] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Accepted: 10/17/2016] [Indexed: 12/17/2022] Open
Abstract
β1, 3-N-acetylglucosminyltransferase 8(β3GnT8) synthesizes a unique cabohydrate structure known as polylactosamine, and plays a vital role in progression of various human cancer types. However, its involvement in gastric cancer remains unclear. In this study, we analyzed the expression and clinical significance of β3GnT8 by Western blot in 6 paired fresh gastric cancer tissues, noncancerous tissues and immunohistochemistry on 110 paraffin-embedded slices. β3GnT8 was found to be over-expressed in gastric cancer tissues, which correlated with lymph node metastasis and TNM stage. Forced the expression of β3GnT8 promoted migration and invasion of gastric cancer cells, whereas β3GnT8 knockdown led to the opposite results. Further studies showed that the regulated β3GnT8 could convert the heterogeneous N-glycosylated forms of CD147 and change the polylactosamine structures carried on CD147. In addition, our data suggested the annexin A2 (ANXA2) to be an essential interaction partner of β3GnT8 during the process of CD147 glycosylation. Collectively, these results provide a novel molecular mechanism for β3GnT8 in promotion of gastric cancer invasion and metastasis. Targeting β3GnT8 could serve as a new strategy for future gastric cancer therapy.
Collapse
Affiliation(s)
- Li Shen
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China; Department of Biochemistry and Molecular Biology, Soochow University, Suzhou 215123, Jiangsu, China
| | - Xiaoxia Dong
- Department of pharmacology, School of Basic Medicine, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Meiyun Yu
- Department of Biochemistry and Molecular Biology, Soochow University, Suzhou 215123, Jiangsu, China
| | - Zhiguo Luo
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China; Institute of Cancer Research, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Shiliang Wu
- Department of Biochemistry and Molecular Biology, Soochow University, Suzhou 215123, Jiangsu, China
| |
Collapse
|
39
|
Li M, Ke J, Wang Q, Qian H, Yang L, Zhang X, Xiao J, Ding H, Shan X, Liu Q, Xiao Y, Bao B, Huang H. Upregulation of ROCK2 in gastric cancer cell promotes tumor cell proliferation, metastasis and invasion. Clin Exp Med 2016; 17:519-529. [PMID: 27921230 DOI: 10.1007/s10238-016-0444-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Accepted: 11/19/2016] [Indexed: 12/27/2022]
Abstract
Rho-associated coiled-coil-containing protein kinase 2 (ROCK2) has been known as an effector for the small GTPase Rho and plays an important role in tumor progression and metastasis. However, the effect of ROCK2 in gastric cancer (GC) has not been identified. This study showed that ROCK2 expression significantly increased in clinical GC tissues compared with adjacent non-cancer tissues. Immunohistochemistrical analysis showed that high expression of ROCK2 was correlated with tumor grade, tumor-node-metastasis stage, infiltration depth, lymph node invasion and Ki-67, and predicted poor prognosis in 135 gastric cancer specimens. In addition, we found that upregulated ROCK2 promoted proliferation, metastasis and invasion of GC cells, while ROCK2 knockdown led to the opposite results in vitro by Cell Counting Kit-8 (CCK-8) assay, colony formation assays, flow cytometric analysis and trans-well assays. Our findings supported that ROCK2 was a significant protein in the progress of GC and would provide a novel promising therapeutic strategy against human GC.
Collapse
Affiliation(s)
- Manhua Li
- Department of Gastroenterology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, Jiangsu, China.,Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 19 QiXiu Road, Nantong, 226001, Jiangsu, China
| | - Jing Ke
- Department of General Surgery, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, Jiangsu, China
| | - Qiuhong Wang
- Department of Clinical Laboratory,Nantong Maternal and Child Health Care Hospital, Affiliated to Nantong University, 339 Shiji Avenue, Nantong, 226001, Jiangsu, China
| | - Hongyan Qian
- Cancer Research Center Nantong, Nantong Tumor Hospital, Nantong, 226001, Jiangsu, China
| | - Lei Yang
- Department of Oncology, Nantong Tumor Hospital, Nantong, 226363, Jiangsu, China
| | - Xunlei Zhang
- Department of Oncology, Nantong Tumor Hospital, Nantong, 226363, Jiangsu, China
| | - Jinzhang Xiao
- Department of Oncology, Nantong Tumor Hospital, Nantong, 226363, Jiangsu, China
| | - Haifang Ding
- Department of Gastroenterology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, Jiangsu, China
| | - Xiaohang Shan
- Department of Gastroenterology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, Jiangsu, China
| | - Qingqing Liu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, Jiangsu, China
| | - Ying Xiao
- Department of Gastroenterology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, Jiangsu, China
| | - Bojun Bao
- Department of Gastroenterology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, Jiangsu, China.
| | - Hua Huang
- Department of Pathology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
40
|
hsa-miR-376c-3p Regulates Gastric Tumor Growth Both In Vitro and In Vivo. BIOMED RESEARCH INTERNATIONAL 2016; 2016:9604257. [PMID: 27965982 PMCID: PMC5124681 DOI: 10.1155/2016/9604257] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 08/10/2016] [Indexed: 11/18/2022]
Abstract
Background. In recent studies, aberrant expression of various microRNAs (miRNAs) is reported to be associated with gastric cancer metastasis. Method. Overexpression construct and inhibitor of hsa-miR-376c-3p were expressed in human gastric adenocarcinoma cell line SGC-7901. The expression level of tumor related genes was detected by qPCR, western blot, and immunostaining. Cell apoptosis was determined by flow cytometry. Xenograft of SGC-7901 cells was used to elucidate the function of hsa-miR-376c-3p in gastric tumor growth in vivo. Result. Expression of hsa-miR-376c-3p was detected in SGC-7901 cells. Downregulation of hsa-miR-376c-3p increased the expression level of BCL-2 and decreased the expression of smad4 and BAD. On the contrary, overexpression of hsa-miR-376c-3p increased the expression of BAD and smad4, while it led to the decreasing expression level of BCL-2. Overexpression of hsa-miR-376c-3p also promoted cell apoptosis in vitro and inhibited gastric tumor growth in vivo. Furthermore, the expression of BCL-2 was higher and expression of smad4 and BAD was lower in tumor tissue than the tissue adjacent to tumor from gastric cancer patients. Conclusion. This study demonstrated that hsa-miR-376c-3p plays an important role in the inhibition of gastric tumor growth and tumor related gene expression both in vitro and in vivo.
Collapse
|
41
|
Wang H, Xiong M, Hu Y, Sun Y, Ma Q. MicroRNA-19b inhibits proliferation of gastric cancer cells by targeting B-cell CLL/lymphoma 3. Oncol Rep 2016; 36:2079-86. [PMID: 27572553 DOI: 10.3892/or.2016.5029] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 08/16/2016] [Indexed: 11/05/2022] Open
Abstract
Previous studies reported that the aberrant expression of miR-19b in gastric cancer tissues and circulating miR-19b is a potential biomarker to indicate progression of gastric cancer. However, the prognostic significance of miR-19b, and its role and underlying mechanisms in gastric cancer remain poorly investigated. In the present study, we demonstrated that the expression of miR-19b was aberrantly downregulated in both gastric cancer tissues and cell lines. Clinical association analyses disclosed that the reduced expression of miR-19b was significantly associated with adverse clinicopathological characteristics including poor differentiation, large tumor size and advanced tumor-node-metastasis (TNM) stage. Gastric cancer patients with low expression of miR-19b had prominent shorter overall survival and disease-free survival. Gain-of-function studies indicated that miR-19b overexpression inhibited cell proliferation and cell cycle progression in MGC-803 cells. While miR-19b silencing promoted cell proliferation and cell cycle progression in SGC-7901 cells. Furthermore, in vivo experiments showed that miR-19b overexpression suppressed the tumor growth of MGC-803 cells. Notably, miR-19b inversely regulated B-cell CLL/lymphoma 3 (BCL3) abundance in gastric cancer cells. BCL3 was identified as a direct target of miR-19b using luciferase reporter assays. Moreover, BCL3 knockdown abolished the effects of miR-19b knockdown on gastric cancer cells. In conclusion, our data suggest that miR-19b may potentially serve as a novel prognostic biomarker and therapeutic target for gastric cancer.
Collapse
Affiliation(s)
- Huan Wang
- Department of Medical Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Mei Xiong
- Department of Medical Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Yongwu Hu
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yusheng Sun
- Department of Traumatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Qing Ma
- Department of Medical Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| |
Collapse
|
42
|
Abnormal expression of FOSB correlates with tumor progression and poor survival in patients with gastric cancer. Int J Oncol 2016; 49:1489-1496. [DOI: 10.3892/ijo.2016.3661] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 07/20/2016] [Indexed: 11/05/2022] Open
|
43
|
Circulating microRNA-196a/b are novel biomarkers associated with metastatic gastric cancer. Eur J Cancer 2016; 64:137-48. [PMID: 27420607 DOI: 10.1016/j.ejca.2016.05.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 04/28/2016] [Accepted: 05/05/2016] [Indexed: 02/07/2023]
Abstract
miR-196a and/or miR-196b, involved in cancer initiation and progression, are frequently upregulated in tumour tissues. However, the clinical significance of these microRNAs in gastric cancer (GC) remains to be clarified. In the current study, we investigated the potential utility of circulating miR-196a/b as novel biomarkers for early detection and/or metastatic prognosis of GC. The quantitative real time-polymerase chain reaction data revealed markedly higher pre-operative circulating miR-196a and miR-196b levels in GC patients than healthy controls. Receiver-operating characteristics curve analysis showed that circulating miR-196a, miR-196b and combined miR-196a and miR-196b (miR-196a/b) are more effective than carcinoembryonic antigen or carbohydrate antigen 19-9 alone in distinguishing GC patients from healthy controls, with higher sensitivity and specificity. Circulating miR-196a exhibited higher diagnostic capacity than combined miR-196a/b or miR-196b alone, highlighting its potential as an effective plasma biomarker for GC. In clinicopathological analysis, elevated circulating miR-196a/b levels were highly correlated with metastatic potential or more advanced stages of disease and poorer survival. In addition, the expression levels of circulating miR-196a/b were reduced after surgical resection in GC patients. Taken together, we propose that circulating miR-196a/b serve as a more sensitive and specific novel biomarker than carbohydrate antigen 19-9 for GC monitor, diagnosis and prognosis.
Collapse
|
44
|
Nanthanangkul S, Suwanrungruang K, Wiangnon S, Promthet S. Survival of Stomach Cancer Cases in Khon Kaen, Thailand 2000-2012. Asian Pac J Cancer Prev 2016; 17:2125-9. [DOI: 10.7314/apjcp.2016.17.4.2125] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
45
|
Xue L, Chen XL, Lin PP, Xu YW, Zhang WH, Liu K, Chen XZ, Yang K, Zhang B, Chen ZX, Chen JP, Zhou ZG, Hu JK. Impact of capillary invasion on the prognosis of gastric adenocarcinoma patients: A retrospective cohort study. Oncotarget 2016; 7:31215-31225. [PMID: 27145279 PMCID: PMC5058751 DOI: 10.18632/oncotarget.9101] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 04/11/2016] [Indexed: 02/05/2023] Open
Abstract
Capillary invasion (CI) has been found to play an important role in metastasis and recurrence of gastric adenocarcinoma (GAC). However, the prognostic significance of CI is still controversial. From January 2005 to December 2011, 1398 patients with GAC who underwent gastrectomy were retrospectively enrolled and divided into CI (+) and CI (-) groups. Clinicopathological features and survival outcomes were compared between these groups. In our study, 227 (16.2%) patients were CI (+). Patients with CI (+) had significantly more advanced tumors and worse prognosis than those with CI (-) (p < 0.001). CI was demonstrated as an independent prognostic factor (p = 0.023) in patients with GAC. When stratified by TNM stage, the prognosis of CI (+) group in stage III was remarkably worse than CI (-) group (p = 0.006), while the differences were not significant in stage I-II and stage IV (both p > 0.05). The nomograms indicated that CI was part of the individual prognostic prediction system. The predictive accuracy of CI and other characteristics was better than TNM alone (p < 0.001). Our finding suggested that CI was an independent prognostic factor in patients with GAC, and the nomogram based on CI and other clinicopathological factors was a valuable and accurate tool in individual prognostic prediction.
Collapse
Affiliation(s)
- Lian Xue
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, China
- Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, China
| | - Xiao-Long Chen
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, China
- Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, China
| | - Pan-Pan Lin
- West China School of Medicine, Sichuan University, China
| | - Yuan-Wei Xu
- West China School of Medicine, Sichuan University, China
| | - Wei-Han Zhang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, China
- Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, China
| | - Kai Liu
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, China
- Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, China
| | - Xin-Zu Chen
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, China
- Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, China
| | - Kun Yang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, China
- Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, China
| | - Bo Zhang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, China
| | - Zhi-Xin Chen
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, China
| | - Jia-Ping Chen
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, China
| | - Zong-Guang Zhou
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, China
- Laboratory of Digestive Surgery, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center West China Hospital, Sichuan University, China
| | - Jian-Kun Hu
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, China
- Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, China
| |
Collapse
|
46
|
Cheng DQ, Gu XD, Li ZY, Xiang JB, Chen ZY. Expression of C4.4A is a potential independent prognostic factor for patients with gastric cancer. Asian Pac J Cancer Prev 2016; 15:3895-9. [PMID: 24935570 DOI: 10.7314/apjcp.2014.15.9.3895] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
C4.4A, a metastasis-associated gene, encodes a glycolipid-anchored membrane protein which is overexpressed in several human malignancies. However, there are few data available on C4.4A expression and its relationship with progression in gastric cancer. Our study was designed to explore the expression of C4.4A in gastric cancer and to correlate it with clinical outcome. C4.4A expression was studied by quantitative real-time RT-PCR and immunohistochemistry for assessment of correlations with clinicopathological factors. C4.4A mRNA expression was significantly up-regulated in gastric cancer as compared with noncancerous tissue (p<0.05)., being observed in 107 (88.4%) of the 121 gastric cancer cases by immunohistochemistry. We found that the expression of C4.4A mRNA was correlated with size of the tumor, depth of invasion, lymph node metastasis, distant metastasis and TNM stage. Moreover, patients with overexpression of C4.4A has a significantly worse survival (p<0.05). Further multivariable analysis indicated that the expression of C4.4A was an independent prognostic indicator for gastric cancer (p<0.05). In conclusion, overexpression of C4.4A correlates with metastatic potential of gastric cancer and C4.4A could be a novel independent prognostic marker for predicting outcome.
Collapse
Affiliation(s)
- Da-Qing Cheng
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China E-mail :
| | | | | | | | | |
Collapse
|
47
|
Xu Y, Huang Y, Wang H, Liu Y. Inhibition of the peritoneal metastasis of human gastric cancer cells by dextran sulphate in vivo and in vitro. Oncol Lett 2016; 11:2384-2390. [PMID: 27073484 DOI: 10.3892/ol.2016.4234] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 01/14/2016] [Indexed: 11/06/2022] Open
Abstract
The present study investigated the inhibitory effects of dextran sulphate (DS) on the peritoneal metastasis of gastric cancer by observing the adhesion and implantation of human gastric cancer cells in the omenta of nude mice. DS or PBS was added to the culture medium of gastric cancer MKN1 cells. The adhesion of the cancer cells to the culture dishes, and the morphological changes of fixed and living cancer cells were observed using fluorescence staining and confocal microscopy. In addition, the expression of integrin β1 was measured using immunohistochemistry and reverse transcription-polymerase chain reaction (RT-PCR). Gastric cancer BGC-823 cells were peritoneally injected into nude mice to develop an animal model. DS and PBS were peritoneally injected into the experimental and control groups, respectively, concurrently with the tumour cells. Haematoxylin and eosin staining was performed, and the number of carcinoma nodules with celiac implantation was counted. Integrin expression was determined by immunohistochemistry and RT-PCR. The results showed that MKN1 cells strongly expressed integrin β1 in the cell membrane and clustered in vitro. DS inhibited the expression of integrin β1 and reduced cluster formation. In addition, the number of pseudopodia formed by the cells decreased, and the cells maintained a rounded shape. The expression of integrin β1 in the adherent and free cells in the experimental group was reduced to 74 and 38% of the levels in the control group, respectively. In the in vivo study, significantly fewer tumour nodules were observed in the experimental group than in the control group (P<0.01). Integrin β1 expression in the experimental group was decreased significantly compared with that in the control group (P<0.01). The present study indicates that DS inhibits the adhesion of human gastric cancer cells, accompanied by a decrease in the expression of integrin β1. DS may inhibit the metastatic celiac implantation of human gastric cancer cells by downregulating integrin β1.
Collapse
Affiliation(s)
- Yuanyi Xu
- Department of Pathology, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Yunning Huang
- Department of Gastrointestinal Surgery, Ningxia People's Hospital, Yinchuan, Ningxia 750001, P.R. China
| | - Honghong Wang
- Department of Pathology, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Yong Liu
- Department of Pathology, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| |
Collapse
|
48
|
Yang L, Zhu J, Huang H, Yang Q, Cai J, Wang Q, Zhu J, Shao M, Xiao J, Cao J, Gu X, Zhang S, Wang Y. PFTK1 Promotes Gastric Cancer Progression by Regulating Proliferation, Migration and Invasion. PLoS One 2015; 10:e0140451. [PMID: 26488471 PMCID: PMC4619205 DOI: 10.1371/journal.pone.0140451] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 09/25/2015] [Indexed: 11/18/2022] Open
Abstract
PFTK1, also known as PFTAIRE1, CDK14, is a novel member of Cdc2-related serine/threonine protein kinases. Recent studies show that PFTK1 is highly expressed in several malignant tumors such as hepatocellular carcinoma, esophageal cancer, breast cancer, and involved in regulation of cell cycle, tumors proliferation, migration, and invasion that further influence the prognosis of tumors. However, the expression and physiological significance of PFTK1 in gastric cancer remain unclear. In this study, we analyzed the expression and clinical significance of PFTK1 by Western blot in 8 paired fresh gastric cancer tissues, nontumorous gastric mucosal tissues and immunohistochemistry on 161 paraffinembedded slices. High PFTK1 expression was correlated with the tumor grade, lymph node invasion as well as Ki-67. Through Cell Counting Kit (CCK)-8 assay, flow cytometry, colony formation, wound healing and transwell assays, the vitro studies demonstrated that PFTK1 overexpression promoted proliferation, migration and invasion of gastric cancer cells, while PFTK1 knockdown led to the opposite results. Our findings for the first time supported that PFTK1 might play an important role in the regulation of gastric cancer proliferation, migration and would provide a novel promising therapeutic strategy against human gastric cancer.
Collapse
Affiliation(s)
- Lei Yang
- Department of Oncology, Nantong Tumor Hospital, Nantong, Jiangsu, China
| | - Jia Zhu
- Department of Oncology, Nantong Tumor Hospital, Nantong, Jiangsu, China
| | - Hua Huang
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Qichang Yang
- Department of Pathology, Nantong first people's hospital, Nantong, Jiangsu, China
| | - Jing Cai
- Department of Oncology, Nantong Tumor Hospital, Nantong, Jiangsu, China
| | - Qiuhong Wang
- Department of Oncology, Nantong Tumor Hospital, Nantong, Jiangsu, China
| | - Junya Zhu
- Department of Pathogen Biology, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong 226001, Jiangsu, China
| | - Mengting Shao
- Department of Pathogen Biology, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong 226001, Jiangsu, China
| | - Jinzhang Xiao
- Department of Oncology, Nantong Tumor Hospital, Nantong, Jiangsu, China
| | - Jie Cao
- Department of Pathology, Nantong first people's hospital, Nantong, Jiangsu, China
| | - Xiaodan Gu
- Department of Pathogen Biology, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong 226001, Jiangsu, China
| | - Shusen Zhang
- Department of Pathogen Biology, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong 226001, Jiangsu, China
| | - Yingying Wang
- Department of Pathogen Biology, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong 226001, Jiangsu, China
- * E-mail:
| |
Collapse
|
49
|
Shim CN, Lee SK. Endoscopic submucosal dissection for undifferentiated-type early gastric cancer: do we have enough data to support this? World J Gastroenterol 2015. [PMID: 24744583 DOI: 10.3748/wjg.v20.i14.393] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Although endoscopic submucosal dissection (ESD) is now accepted for treatment of early gastric cancers (EGC) with negligible risk of lymph node (LN) metastasis, ESD for intramucosal undifferentiated type EGC without ulceration and with diameter ≤ 2 cm is regarded as an investigational treatment according to the Japanese gastric cancer treatment guidelines. This consideration was largely based on the analysis of surgically resected EGCs that contained undifferentiated type EGCs; however, results from several institutes showed some discrepancies in sample size and incidence of LN metastasis. Recently, some reports about the safety and efficacy of ESD for undifferentiated type EGC meeting the expanded criteria have been published. Nonetheless, only limited data are available regarding long-term outcomes of ESD for EGC with undifferentiated histology so far. At the same time, endoscopists cannot ignore the patients' desire to guarantee quality of life after the relatively non-invasive endoscopic treatment when compared to conventional surgery. To satisfy the needs of patients and provide solid evidence to support ESD for undifferentiated EGC, we need more delicate tools to predict undetected LN metastasis and more data that can reveal predictive factors for LN metastasis.
Collapse
Affiliation(s)
- Choong Nam Shim
- Choong Nam Shim, Sang Kil Lee, Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul 120-752, South Korea
| | - Sang Kil Lee
- Choong Nam Shim, Sang Kil Lee, Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul 120-752, South Korea
| |
Collapse
|
50
|
Zhang J, Song Y, Zhang C, Zhi X, Fu H, Ma Y, Chen Y, Pan F, Wang K, Ni J, Jin W, He X, Su H, Cui D. Circulating MiR-16-5p and MiR-19b-3p as Two Novel Potential Biomarkers to Indicate Progression of Gastric Cancer. Theranostics 2015; 5:733-45. [PMID: 25897338 PMCID: PMC4402497 DOI: 10.7150/thno.10305] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2014] [Accepted: 02/26/2015] [Indexed: 12/12/2022] Open
Abstract
Gastric cancer (GC) is the second most common cancer in China and the second leading cause of cancer-related death in the world. Identifying circulating biomarkers is helpful to improve theranostics of gastric cancer. Herein, we are for the first time to report miR-16-5p and miR-19b-3p were identified to be the novel potential plasma biomarkers to detect gastric cancer. Differentially expressed miRNAs were initially screened out by genome-wide miRNA profiling microarrays between 16 plasma samples of gastric cancer and 18 matched normal controls, and then were quantified and validated by quantitative reverse transcription-PCR method between 155 gastric cancer cases and 111 normal controls. Additionally, 30 plasma samples from precancerous lesions and 18 paired samples from gastric cancer patients with gastrectomy were further detected. Results showed that based on two normalization methods, miR-16-5p and miR-19b-3p in plasma were found to be capable of distinguishing normal population from GC cases with different TNM stages and differentiation grades, particularly including the early cancer cases (P<0.05). And the two miRNAs were down-regulated in GC cases (FC<0.5). Especially, the down-regulation degree was correlated with the progression of the GC cases from the early stage to the advanced stage (0.2< rs<0.3, P<0.01). And the same weak down-regulation of the two biomarkers as the early GC occurred initially in the precancerous diseases (P<0.05). The corresponding performance of the two miRNAs to detect GC in ROC analysis gradually performed better with the disease progression from the earlier stages or lower grades to the advanced stages (TNM Ⅳ stage: AUC=0.832 for miR-16-5p; TNM Ⅲ stage: AUC=0.822 for miR-19b-3p) or high grade (Poorly differentiated: AUC=0.801, 0.791 respectively for miR-16-5p and miR-19b-3p). Additionally, miR-19b-3p remained down-regulated in patient plasma within 9 days after gastrectomy. In conclusion, miR-19b-3p and miR-16-5p maybe prospective biomarkers to detect gastric cancer and indicate its progression, and thus may own great potential in applications such as early screening and progression evaluation of gastric cancer in the near future.
Collapse
|