1
|
Yu C, Wang J. The impact of Helicobacter pylori on gastric cancer formation and early warning signal identification. J Chem Phys 2024; 161:235102. [PMID: 39704568 DOI: 10.1063/5.0243016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 11/25/2024] [Indexed: 12/21/2024] Open
Abstract
Gastric cancer is highly prevalent in Asia and is characterized by poor prognosis post-surgery and a high recurrence rate within five years. Research has highlighted the role of Helicobacter pylori in initiating or accelerating gastric cancer development. However, quantitative analysis of its impact on gastric cancer carcinogenesis is still lacking. This study employs gene regulatory networks and landscape and flux theory, integrating genetic and epigenetic factors, to quantitatively elucidate how Helicobacter pylori influences gastric cancer progression. Varied Helicobacter pylori infection concentrations lead to significant shifts in system thermodynamic and dynamic driving forces, altering gene expression levels. Quantitative analysis of entropy production rate and mean-flux in the gastric cancer system reveals the global changes in thermodynamic and dynamic driving forces. Coupled with autocorrelation, cross correlation, and variance analysis, we pinpoint critical stages of Helicobacter pylori infection, serving as early warning signals for gastric cancer. This approach bridges theoretical and clinical realms, dynamically assessing Helicobacter pylori's impact on gastric cancer and identifying crucial early warning signals, with significant clinical and translational implications.
Collapse
Affiliation(s)
- Chong Yu
- Department of Statistics, Jilin University of Finance and Economics Changchun, Jilin 130117, China
| | - Jin Wang
- Joint Research Centre on Medicine, the Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang 315700, People's Republic of China
- Center for Theoretical Interdisciplinary Sciences Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, People's Republic of China
- Department of Chemistry and of Physics and Astronomy State University of New York at Stony Brook, Stony Brook, New York 11794-3400, USA
| |
Collapse
|
2
|
Fernández Aceñero MJ, Díaz del Arco C. Hereditary Gastrointestinal Tumor Syndromes: When Risk Comes with Your Genes. Curr Issues Mol Biol 2024; 46:6440-6471. [PMID: 39057027 PMCID: PMC11275188 DOI: 10.3390/cimb46070385] [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: 05/27/2024] [Revised: 06/19/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
Despite recent campaigns for screening and the latest advances in cancer therapy and molecular biology, gastrointestinal (GI) neoplasms remain among the most frequent and lethal human tumors. Most GI neoplasms are sporadic, but there are some well-known familial syndromes associated with a significant risk of developing both benign and malignant GI tumors. Although some of these entities were described more than a century ago based on clinical grounds, the increasing molecular information obtained with high-throughput techniques has shed light on the pathogenesis of several of them. The vast amount of information gained from next-generation sequencing has led to the identification of some high-risk genetic variants, although others remain to be discovered. The opportunity for genetic assessment and counseling in these families has dramatically changed the management of these syndromes, though it has also resulted in significant psychological distress for the affected patients, especially those with indeterminate variants. Herein, we aim to summarize the most relevant hereditary cancer syndromes involving the stomach and colon, with an emphasis on new molecular findings, novel entities, and recent changes in the management of these patients.
Collapse
Affiliation(s)
- María Jesús Fernández Aceñero
- Department of Legal Medicine, Psychiatry and Pathology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
- Department of Pathology, Hospital Clínico San Carlos, Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Cristina Díaz del Arco
- Department of Legal Medicine, Psychiatry and Pathology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
- Department of Pathology, Hospital Clínico San Carlos, Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| |
Collapse
|
3
|
Cao D, Shi F, Sheng J, Zhu J, Yin H, Qin S, Yao J, Zhu L, Lu J, Wang X. Machine learning-driven SERS analysis platform for rapid and accurate detection of precancerous lesions of gastric cancer. Mikrochim Acta 2024; 191:415. [PMID: 38907752 DOI: 10.1007/s00604-024-06508-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 06/13/2024] [Indexed: 06/24/2024]
Abstract
A novel approach is proposed leveraging surface-enhanced Raman spectroscopy (SERS) combined with machine learning (ML) techniques, principal component analysis (PCA)-centroid displacement-based nearest neighbor (CDNN). This label-free approach can identify slight abnormalities between SERS spectra of gastric lesions at different stages, offering a promising avenue for detection and prevention of precancerous lesion of gastric cancer (PLGC). The agaric-shaped nanoarray substrate was prepared using gas-liquid interface self-assembly and reactive ion etching (RIE) technology to measure SERS spectra of serum from mice model with gastric lesions at different stages, and then a SERS spectral recognition model was trained and constructed using the PCA-CDNN algorithm. The results showed that the agaric-shaped nanoarray substrate has good uniformity, stability, cleanliness, and SERS enhancement effect. The trained PCA-CDNN model not only found the most important features of PLGC, but also achieved satisfactory classification results with accuracy, area under curve (AUC), sensitivity, and specificity up to 100%. This demonstrated the enormous potential of this analysis platform in the diagnosis of PLGC.
Collapse
Affiliation(s)
- Dawei Cao
- School of Information Engineering, Yangzhou Polytechnic Institute, Yangzhou, 225002, China
| | - Fanfeng Shi
- School of Information Engineering, Yangzhou Polytechnic Institute, Yangzhou, 225002, China
| | - JinXin Sheng
- Department of General Surgery, Nantong Haimen People's Hospital, Nantong, 226100, China
| | - Jinhua Zhu
- Department of Gastroenterology, Yangzhong People's Hospital, Zhenjiang, 212200, China
| | - Hongjun Yin
- Department of Gastroenterology, Yangzhong People's Hospital, Zhenjiang, 212200, China
| | - ShiChen Qin
- Department of General Surgery, Nantong Haimen People's Hospital, Nantong, 226100, China
| | - Jie Yao
- Department of General Surgery, Nantong Haimen People's Hospital, Nantong, 226100, China
| | - LiangFei Zhu
- Department of General Surgery, Nantong Haimen People's Hospital, Nantong, 226100, China
| | - JinJun Lu
- Department of General Surgery, Nantong Haimen People's Hospital, Nantong, 226100, China
| | - XiaoYong Wang
- Department of General Surgery, Nantong Haimen People's Hospital, Nantong, 226100, China.
| |
Collapse
|
4
|
Shah D, Bentrem D. Environmental and Genetic Risk Factors for Gastric Cancer. Cancer Treat Res 2024; 192:1-17. [PMID: 39212913 DOI: 10.1007/978-3-031-61238-1_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Gastric cancer is a heterogeneous and prevalent disease. The traditional environmental exposures associated with elevated risk of gastric cancer are less prevalent in the USA today. Genetic risks and risks associated with inflammation remain. Most cases are sporadic, and familial clustering is observed in about 10% of the cases. Hereditary gastric cancer accounts for a very low percentage of cases. Here we review the genetic and environmental risk factors associated with the disease. In addition, we will review screening guidelines and current modalities that are available for screening in high-risk populations.
Collapse
Affiliation(s)
- Dhavan Shah
- Northwestern Quality Improvement, Research, and Education in Surgery, Department of Surgery, Feinberg School of Medicine, Northwestern University, Evanston, USA
| | - David Bentrem
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Evanston, USA.
- Jesse Brown VA Medical Center, Chicago, USA.
| |
Collapse
|
5
|
Zhang X, Zhang H, Wang Z, Ma X, Luo J, Zhu Y. PWSC: a novel clustering method based on polynomial weight-adjusted sparse clustering for sparse biomedical data and its application in cancer subtyping. BMC Bioinformatics 2023; 24:490. [PMID: 38129803 PMCID: PMC10740247 DOI: 10.1186/s12859-023-05595-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 12/04/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Clustering analysis is widely used to interpret biomedical data and uncover new knowledge and patterns. However, conventional clustering methods are not effective when dealing with sparse biomedical data. To overcome this limitation, we propose a hierarchical clustering method called polynomial weight-adjusted sparse clustering (PWSC). RESULTS The PWSC algorithm adjusts feature weights using a polynomial function, redefines the distances between samples, and performs hierarchical clustering analysis based on these adjusted distances. Additionally, we incorporate a consensus clustering approach to determine the optimal number of classifications. This consensus approach utilizes relative change in the cumulative distribution function to identify the best number of clusters, resulting in more stable clustering results. Leveraging the PWSC algorithm, we successfully classified a cohort of gastric cancer patients, enabling categorization of patients carrying different types of altered genes. Further evaluation using Entropy showed a significant improvement (p = 2.905e-05), while using the Calinski-Harabasz index demonstrates a remarkable 100% improvement in the quality of the best classification compared to conventional algorithms. Similarly, significantly increased entropy (p = 0.0336) and comparable CHI, were observed when classifying another colorectal cancer cohort with microbial abundance. The above attempts in cancer subtyping demonstrate that PWSC is highly applicable to different types of biomedical data. To facilitate its application, we have developed a user-friendly tool that implements the PWSC algorithm, which canbe accessed at http://pwsc.aiyimed.com/ . CONCLUSIONS PWSC addresses the limitations of conventional approaches when clustering sparse biomedical data. By adjusting feature weights and employing consensus clustering, we achieve improved clustering results compared to conventional methods. The PWSC algorithm provides a valuable tool for researchers in the field, enabling more accurate and stable clustering analysis. Its application can enhance our understanding of complex biological systems and contribute to advancements in various biomedical disciplines.
Collapse
Affiliation(s)
- Xiaomeng Zhang
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Hongtao Zhang
- School of Mathematics and Statistics, Wuhan University, Wuhan, 430070, Hubei Province, China
| | - Zhihao Wang
- School of Mathematics and Statistics, Wuhan University, Wuhan, 430070, Hubei Province, China
| | - Xiaofei Ma
- School of Mathematics and Statistics, Wuhan University, Wuhan, 430070, Hubei Province, China
| | - Jiancheng Luo
- School of Mathematics and Statistics, Wuhan University, Wuhan, 430070, Hubei Province, China.
| | - Yingying Zhu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China.
| |
Collapse
|
6
|
Wu F, Guo X, Ren Y, Peng Y, Lai Z, Xu J. CircRNA0007766 accelerates cancer progression via miR-34c-5p/cyclin D1 axis in adenocarcinoma of the esophagogastric junction (AEG). Cell Signal 2023; 112:110912. [PMID: 37802173 DOI: 10.1016/j.cellsig.2023.110912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/12/2023] [Accepted: 10/02/2023] [Indexed: 10/08/2023]
Abstract
Growing empirical evidence shows that circular RNAs (circRNAs) are implicated in tumor pathogenesis. However, little is known about the mechanism by which circRNAs contribute to the progression of adenocarcinoma of the esophagogastric junction (AEG). We conducted RNA high-throughput sequencing and bioinformatic analyses on 22 AEG tissues and their matching healthy gastric mucosal tissues and found that circRNA0007766 may act as a tumor promoter in AEG pathogenesis. BaseScope® in situ hybridization revealed that circRNA0007766 was strongly upregulated in AEG. We then constructed co-expression and ceRNA networks to elucidate the relationships among specific circRNAs, microRNAs (miRNAs), and mRNAs. We also demonstrated that circRNA0007766 acted as the sponge of miR-34c-5p, thereby positively regulating cyclin D1. In vivo and in vitro experiments demonstrated the roles of circRNA0007766 in promoting AEG progression and invasion. AEG tissues are characterized by circRNA0007766 upregulation which is correlated with lymph node metastasis and poor survival. To the best of our knowledge, the present study is one of the first to show that the circRNA0007766/miR-34c-5p/cyclin D1 axis is important in AEG progression. Furthermore, the results of this work imply that circRNA0007766 is potentially a novel AEG biomarker.
Collapse
Affiliation(s)
- Feng Wu
- First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China.
| | - Xin Guo
- Medical ICU, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital,Chinese Academy of Medical Sciences, Taiyuan, Shanxi Province, China
| | - Yifan Ren
- First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Yuting Peng
- Faculty of Graduate Studies, Shanxi Medical University, Taiyuan,Shanxi Province, China
| | - Zhiyong Lai
- Department of Hepatobiliary and Pancreatic Surgery, First Hospital of Shanxi Medical University, Taiyuan,Shanxi Province, China.
| | - Jun Xu
- Department of Hepatobiliary and Pancreatic Surgery, First Hospital of Shanxi Medical University, Taiyuan,Shanxi Province, China; Institute of Liver Diseases and Organ Transplantation, Shanxi Medical University, Taiyuan,Shanxi Province, China.
| |
Collapse
|
7
|
Shen WN, Qu XD, Chen SS, Liu YH, Gao MJ, Shi YQ. Relationship between gastric xanthoma, gastric precancerous lesions, and gastric cancer: A retrospective study. J Dig Dis 2023; 24:340-347. [PMID: 37417403 DOI: 10.1111/1751-2980.13202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 06/27/2023] [Accepted: 07/05/2023] [Indexed: 07/08/2023]
Abstract
OBJECTIVE To evaluate the relationship between gastric cancer and its precancerous lesions and gastric xanthoma. METHODS Medical records of 47 736 patients who underwent gastroscopy in our center from January 2020 to December 2021 were reviewed. Patients' age, sex, endoscopic and histopathological findings, and the presence, number and location of gastric xanthoma were recorded. To investigate the detection rate of gastric xanthoma at different stages of gastric lesions, the participants were further divided into the chronic gastritis group (n = 42 758), the precancerous lesion group (n = 3672), and the gastric cancer group (n = 1306), respectively. RESULTS The overall detection rate of gastric xanthoma was 2.85%, and it was most commonly observed in the gastric antrum (52.50%). In addition, gastric xanthoma was more common in men and more likely to be single lesion. It was most detected in the precancerous lesion group (8.39%), followed by the gastric cancer group (5.44%), and least in the chronic gastritis group (2.29%). Multivariate analysis showed that gastric xanthoma was closely related to precancerous lesions (odds ratio [OR] 3.197, 95% confidence interval [CI] 2.791-3.662, P < 0.001) and gastric cancer (OR 1.794, 95% CI 1.394-2.309, P < 0.001). CONCLUSION Gastric xanthoma is closely related to gastric precancerous lesions and gastric cancer.
Collapse
Affiliation(s)
- Wei Na Shen
- Xi'an Medical University, Xi'an, Shaanxi Province, China
| | - Xiao Dong Qu
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi Province, China
| | - Sha Sha Chen
- Xi'an Medical University, Xi'an, Shaanxi Province, China
| | - Yu Huan Liu
- Xi'an Medical University, Xi'an, Shaanxi Province, China
| | - Meng Jie Gao
- Xi'an Medical University, Xi'an, Shaanxi Province, China
| | - Yong Quan Shi
- Xi'an Medical University, Xi'an, Shaanxi Province, China
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi Province, China
| |
Collapse
|
8
|
Sun J, Meng M. Chemoprotective Effect of Scutellarin against Gastric Cancer in Rats: An in vitro and in vivo Study. J Oleo Sci 2022; 71:1003-1012. [PMID: 35781253 DOI: 10.5650/jos.ess21399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
This study evaluated the chemoprotective effect of scutellarin (SC) in vitro and in vivo against gastric carcinogenesis in rats and celllines and examined the underlying mechanism. Gastric cancer celllines (AGS) was used for the in vitro study and lactate dehydrogenase (LDH) profile, histone deacetylase (HDAC) assay, cell cycle & apoptosis ratio and antioxidant parameters were measured. N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) was used to induce gastric carcinogenesis in rats and the rats received the different doses of SC (10, 20 and 30 mg/kg). The body weight and tumor incidence were measured at regular time intervals. The antioxidant and pro-inflammatory cytokines were estimated. The finding of data showed that the drug was effective against AGS cell line. Supplementation of scutellarin revealed an upregulation in body weight compared with the MNNG group rats. Moreover, it also reduced the incidence of tumor. It also altered the significant DNA density, LDH content, mucus content and acidity. Scutellarin treated rats showed improved activity in enzymatic and non-enzymatic antioxidant profile and reversed the content of cytokines compared with MNNG induced gastric cancer group rats. This research reveals the chemoprotective property of the scutellarin and highlights the promising role of drug by alteration of inflammatory pathway by minimizing its adverse effect.
Collapse
Affiliation(s)
- Jiu Sun
- Department of General Surgery, The first people's Hospital of Yibin
| | - Meng Meng
- Department of Gastrointestinal Surgery, Shandong Provincial Third Hospital, Shandong University
| |
Collapse
|
9
|
Shah D, Bentrem D. Environmental and genetic risk factors for gastric cancer. J Surg Oncol 2022; 125:1096-1103. [PMID: 35481919 PMCID: PMC9322002 DOI: 10.1002/jso.26869] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/10/2022] [Accepted: 03/12/2022] [Indexed: 12/11/2022]
Abstract
Gastric cancer is a heterogeneous and prevalent disease. The traditional environmental exposures associated with an elevated risk of gastric cancer are less prevalent in the United States today. Genetic risks and risks associated with inflammation remain. Most cases are sporadic and familial clustering is observed in about 10% of the cases. Hereditary gastric cancer accounts for a very low percentage of cases. Here we review the genetic and environmental risk factors associated with the disease.
Collapse
Affiliation(s)
- Dhavan Shah
- Department of Surgery, Surgical Outcome and Quality Improvement Center, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - David Bentrem
- Department of Surgery, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
- Jesse Brown VA Medical CenterChicagoIllinoisUSA
| |
Collapse
|
10
|
Maheshwari U, Sharma M, Goel V, Goyal P, Jain P, Agarwal C, Jajodia A, Talwar V, BP A, Joga S, Doval DC, Pasricha S, Koyyala VPB. Clinical Profile and Outcomes of Treatment in Gastric Cancer in Young Patients in India. ASIAN JOURNAL OF ONCOLOGY 2022. [DOI: 10.1055/s-0042-1744451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Abstract
Introduction Gastric cancer poses an enormous burden across the globe and India in terms of cancer-related mortality. There is paucity of epidemiological and survival data among young gastric cancer patients in India. In this study, we retrospectively analyzed the general characteristics, clinical profile, and survival data of gastric cancer in young patients < 30 years at tertiary care institution at New Delhi, India.
Materials and Methods Young gastric cancer patients (≤30 years) who were registered over a period of 7 years (2010–2017) were analyzed at a tertiary care center. Total of 2,735 patients of gastric cancers were registered out of which 70 cases were younger than 30 years, of which 63 patients were available for final analysis and data was missing for the remaining 7 cases. All patients underwent standard diagnostic and staging investigation and were staged as per American Joint Committee on Cancer 7 staging system. Lymph node ratio was calculated as number of positive nodes by the number of lymph nodes removed and were categorized as ≤0.6 and >0.6. Minimum follow-up of 1 year was required for inclusion in the study. Twelve patients were lost to follow-up and were not included for survival analysis.
Results Younger patients (≤30 years) with gastric cancer were 2.5% of total gastric cancer patients. Mean age was 24.9 years with males being involved twice as commonly as females (2.15:1). Positive family history was present in 14.2% patients and smoking was present in 57.1% patients. Metastatic disease at presentation was present in 69.8% patients, while only 6.4% patients presented with stage I/II disease. Fourteen patients underwent surgery, out of which six patients underwent partial gastrectomy and remaining eight underwent total gastrectomy with D2 nodal dissection. Median overall survival was 10.8 months (8.8–12.8) and 2-year overall survival was 15.1%.
Conclusion Incidence of stomach cancer in young patients is more than expected and more than global average in India. Most of these young patients are presenting in advanced stage and survival is poor compared with typical aged patients
Collapse
Affiliation(s)
| | - Manish Sharma
- Department of Medical Oncology, Rajiv Gandhi Cancer Institute and Research Centre (RGCIRC), New Delhi, India
| | - Varun Goel
- Department of Medical Oncology, Rajiv Gandhi Cancer Institute and Research Centre (RGCIRC), New Delhi, India
| | - Pankaj Goyal
- Department of Medical Oncology, Rajiv Gandhi Cancer Institute and Research Centre (RGCIRC), New Delhi, India
| | - Parveen Jain
- Department of Medical Oncology, Rajiv Gandhi Cancer Institute and Research Centre (RGCIRC), New Delhi, India
| | - Chaturbhuj Agarwal
- Department of Medical Oncology, Rajiv Gandhi Cancer Institute and Research Centre (RGCIRC), New Delhi, India
| | - Ankush Jajodia
- Department of Radiology, Rajiv Gandhi Cancer Institute and Research Centre (RGCIRC), New Delhi, India
| | - Vineet Talwar
- Department of Medical Oncology, Rajiv Gandhi Cancer Institute and Research Centre (RGCIRC), New Delhi, India
| | - Amrit BP
- Department of Medical Oncology, Rajiv Gandhi Cancer Institute and Research Centre (RGCIRC), New Delhi, India
| | - Srujana Joga
- Department of Medical Oncology, Rajiv Gandhi Cancer Institute and Research Centre (RGCIRC), New Delhi, India
| | - Dinesh Chandra Doval
- Department of Medical Oncology, Rajiv Gandhi Cancer Institute and Research Centre (RGCIRC), New Delhi, India
| | - Sunil Pasricha
- Department of Pathology, Rajiv Gandhi Cancer Institute and Research Centre (RGCIRC), New Delhi, India
| | | |
Collapse
|
11
|
Hereditary diffuse gastric cancer (HDGC). An overview. Clin Res Hepatol Gastroenterol 2022; 46:101820. [PMID: 34656755 DOI: 10.1016/j.clinre.2021.101820] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/02/2021] [Accepted: 09/25/2021] [Indexed: 02/06/2023]
Abstract
It is estimated that up to 10% of gastric carcinomas show familial aggregation. In contrast, around 1-3 % (approximately 33,000 yearly) are genuinely hereditary. Hereditary diffuse gastric cancer (HDGC) is a rare malignancy characterized by autosomal dominant inheritance of pathological variants of the CDH1 and CTNNA1 genes encoding the adhesion molecules E-cadherin and α-catenin, respectively. The multifocal nature of the disease and the difficulty of visualizing precursor lesions by endoscopy underscore the need to be aware of this malignancy as surgical prevention can be fully protective. Here, we provide an overview of the main epidemiological, clinical, genetic, and pathological features of HDGC, as well as updated guidelines for its diagnosis, genetic testing, counseling, surveillance, and management. We conclude that HDGC is a rare, highly penetrant disease that is difficult to diagnose and manage, so it is necessary to correctly identify it to offer patients and their families' adequate management following the recommendations of the IGCL. A critical point is identifying a mutation in HDGC families to determine whether unaffected relatives are at risk for cancer.
Collapse
|
12
|
Ilic M, Ilic I. Epidemiology of stomach cancer. World J Gastroenterol 2022; 28:1187-1203. [PMID: 35431510 PMCID: PMC8968487 DOI: 10.3748/wjg.v28.i12.1187] [Citation(s) in RCA: 168] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/30/2021] [Accepted: 02/16/2022] [Indexed: 02/06/2023] Open
Abstract
Despite a decline in incidence and mortality during the last decades, stomach cancer is one of the main health challenges worldwide. According to the GLOBOCAN 2020 estimates, stomach cancer caused approximately 800000 deaths (accounting for 7.7% of all cancer deaths), and ranks as the fourth leading cause of cancer deaths in both genders combined. About 1.1 million new cases of stomach cancer were diagnosed in 2020 (accounting for 5.6% of all cancer cases). About 75% of all new cases and all deaths from stomach cancer are reported in Asia. Stomach cancer is one of the most lethal malignant tumors, with a five-year survival rate of around 20%. There are some well-established risk factors for stomach cancer: Helicobacter pylori infection, dietary factors, tobacco, obesity, and radiation. To date, the most important way of preventing stomach cancer is reduced exposure to risk factors, as well as screening and early detection. Further research on risk factors can help identify various opportunities for more effective prevention. Screening programs for stomach cancer have been implemented in a few countries, either as a national or opportunistic screening of high-risk individuals only. Generally, due to its high aggressiveness and heterogeneity, stomach cancer still remains a severe global health problem.
Collapse
Affiliation(s)
- Milena Ilic
- Department of Epidemiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac 34000, Serbia
| | - Irena Ilic
- Faculty of Medicine, University of Belgrade, Belgrade 11000, Serbia
| |
Collapse
|
13
|
Luu MN, Quach DT, Hiyama T. Screening and surveillance for gastric cancer: Does family history play an important role in shaping our strategy? Asia Pac J Clin Oncol 2021; 18:353-362. [PMID: 34811928 DOI: 10.1111/ajco.13704] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 09/23/2021] [Indexed: 12/24/2022]
Abstract
Family history is an important risk factor of gastric cancer. No guidelines have been developed that target gastric cancer with a family history; only hereditary familial gastric cancer is targeted. We review the available evidence regarding the familial aggregation mechanisms of gastric cancer and a strategy of screening and surveillance for gastric cancer in individuals with a positive family history of the disease. As there is a synergic effect of Helicobacter pylori infection and family history on the increased risk of gastric cancer, Helicobacter pylori eradication should be considered in all infected individuals with a family history of gastric cancer. Currently, there is weak evidence indicating that suitable timing to initiate eradication therapy is at the age of 20, when precancerous lesions, including significant gastric atrophy and intestinal metaplasia, have not been established. Reasonable timing to initiate screening for gastric cancer in individuals with a family history of gastric cancer is 10 years prior to the age of onset of gastric cancer in affected relatives. A 2-year surveillance interval, instead of the 3-year interval recommended in the present guidelines, may be better to detect early gastric cancer in those individuals who have already developed precancerous gastric lesions.
Collapse
Affiliation(s)
- Mai Ngoc Luu
- Department of Internal Medicine, University of Medicine and Pharmacy, at Ho Chi Minh City, Ho Chi Minh, Vietnam
| | - Duc Trong Quach
- Department of Internal Medicine, University of Medicine and Pharmacy, at Ho Chi Minh City, Ho Chi Minh, Vietnam
| | - Toru Hiyama
- Health Service Center, Hiroshima University, Higashihiroshima, Japan
| |
Collapse
|
14
|
Zhang D, Chen I, Liao X. Small Intestinal Adenocarcinomas Arising in Enteritis Cystica Profunda With Metaplasia: A Report of 2 Cases. Appl Immunohistochem Mol Morphol 2021; 29:759-764. [PMID: 34132683 DOI: 10.1097/pai.0000000000000956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 05/23/2021] [Indexed: 11/25/2022]
Abstract
Enteritis cystica profunda (ECP) is an uncommon benign condition arising after mucosal damage. We describe 2 cases of small intestinal adenocarcinomas associated with ECP at the distal ileum, one in a background of active Crohn ileitis (case 1), the other 22 years after pelvic radiation therapy (case 2). Both patients presented with small bowel obstruction and received ileocectomy. Macroscopic examination identified an indurated/strictured area in the distal ileum. Histologically, both cases showed a low-grade tubuloglandular adenocarcinoma arising in a background of chronic ischemic stricture and ECP lined by flat cuboidal cells with mild cytologic atypia resembling pancreatobiliary-type epithelium. There was no conventional dysplasia in the surface or adjacent mucosa. Immunohistochemically, both ECP with metaplasia and invasive carcinomas were diffusely positive for CK7 and CK19, while focally positive for CDX2 or CK20. Both cases showed normal wild-type p53 expression. Case 2 was also mismatch repair protein proficient, with membranous β-catenin staining, and retained nuclear SMAD4 expression. In summary, the 2 cases uniquely exhibits "enteritis-metaplasia-carcinoma" sequence, which has not been reported before. This process appears to bypass conventional dysplasia, be slow and indolent, independent of p53, APC/β-catenin, and SMAD4/TGFβ signaling pathways.
Collapse
Affiliation(s)
- Dongwei Zhang
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY
| | | | | |
Collapse
|
15
|
Pan Z, Fu Z, Luo C, Bao Y, Wang M, Cao W, Xu X. CDH1 germline mutations in a Chinese cohort with hereditary diffuse gastric cancer. J Cancer Res Clin Oncol 2021; 148:2145-2151. [PMID: 34537906 DOI: 10.1007/s00432-021-03775-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 08/17/2021] [Indexed: 12/14/2022]
Abstract
PURPOSE Germline mutations in CDH1 are associated with hereditary diffuse gastric cancer (HDGC) and have been identified in multiple ethnicities. However, CDH1 germline mutations have seldom been documented in Chinese patients with HDGC, and their frequency remains unclear. Here, we aimed to examine the frequency of CDH1 germline mutations in Chinese patients with HDGC. In total, 285 patients who met the International Gastric Cancer Linkage Consortium 2015 testing criteria of HDGC for CDH1 germline mutations were recruited. METHODS All 16 CDH1 exons, including neighboring intronic sequences, were amplified using polymerase chain reaction and screened using Sanger sequencing. Variants were analyzed using Mutation Surveyor V4.0, SIFT, and PolyPhen-2 software. RESULTS Three nonsense and nine missense CDH1 germline mutations were identified in 21 of 285 index cases (7.4%). Two CDH1 germline mutations, N405Y (Asn405Tyr) and W409X (Trp409Ter), were identified as new variants. In addition, up to 28.6% of CDH1 mutations in the 21 indicated patients were identified as c.1775G>C (E551Q). The frequency of CDH1 mutations was 6.5% (7/108) in HDGC and 7.9% (14/177) in early onset diffuse gastric cancer (EODGC). The mutation detection rates of CDH1 in males and females were 6.7% (4/60) and 8.5% (10/117) in EODGC and 4.6% (3/65) and 9.3% (4/43) in HDGC, respectively. CONCLUSION These data reveal, for the first time, the type and frequency of CDH1 germline mutations in Chinese HDGC and demonstrate that germline CDH1 mutations are a noteworthy contributor to the high frequency of HDGC in Chinese.
Collapse
Affiliation(s)
- Zhiwen Pan
- Clinical Laboratory Department, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China.,Zhejiang Key Laboratory of Diagnosis & Treatment Technology on Thoracic Oncology (Lung and Esophagus), Hangzhou, Zhejiang, China
| | - Zhixuan Fu
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Cong Luo
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Yejiang Bao
- Clinical Laboratory Department, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Mingli Wang
- Clinical Laboratory Department, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Wenming Cao
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Xiaohong Xu
- Clinical Laboratory Department, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China.
| |
Collapse
|
16
|
da Costa AC, Santa-Cruz F, Mattos LAR, Rêgo Aquino MA, Martins CR, Bandeira Ferraz ÁA, Figueiredo JL. Cathepsin S as a target in gastric cancer. Mol Clin Oncol 2020; 12:99-103. [PMID: 31929878 PMCID: PMC6951222 DOI: 10.3892/mco.2019.1958] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 09/19/2019] [Indexed: 02/06/2023] Open
Abstract
Cathepsin S (Cat S) is a protein expressed in some epithelial cells, which appears to be associated with cancer metastasis and recurrence. The abnormal expression of Cat S has been reported to be associated with the progression of certain types of gastrointestinal neoplasms, including gastric cancer (GC). There is a need to identify novel biomarkers and therapeutic targets associated with the growth, invasion and migration of GC cells, in order to develop non-invasive diagnostic and prognostic procedures and design new therapeutic approaches. The aim of the present study was to assess the association between Cat S and oncogenic processes implicated in the development of GC, focusing on the diagnostic and therapeutic potential of this molecule in GC. A search was conducted through the PubMed and Cochrane Central Register of Controlled Trials electronic databases for relevant literature published between 2003 and 2018, using the mesh terms 'cathepsin S' and 'cancer' and 'gastric cancer'.
Collapse
Affiliation(s)
- Adriano Carneiro da Costa
- Unidade de Oncologia, Hospital das Clínicas da Universidade Federal de Pernambuco, Recife, PE 50670-901, Brazil
| | - Fernando Santa-Cruz
- Centro de Ciências Médicas, Universidade Federal de Pernambuco, Recife, PE 50670-901, Brazil
| | - Luiz Alberto Reis Mattos
- Unidade de Oncologia, Hospital das Clínicas da Universidade Federal de Pernambuco, Recife, PE 50670-901, Brazil
| | | | - Camila Ramos Martins
- Curso de Medicina, Centro Universitário de João Pessoa, Recife, PE 50670-901, Brazil
| | | | - José Luiz Figueiredo
- Departamento de Cirurgia, Universidade Federal de Pernambuco, Recife, PE 50670-901, Brazil
| |
Collapse
|
17
|
Yang L, Ying X, Liu S, Lyu G, Xu Z, Zhang X, Li H, Li Q, Wang N, Ji J. Gastric cancer: Epidemiology, risk factors and prevention strategies. Chin J Cancer Res 2020; 32:695-704. [PMID: 33446993 PMCID: PMC7797232 DOI: 10.21147/j.issn.1000-9604.2020.06.03] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Gastric cancer (GC) is a global health problem, with more than 1 million people newly diagnosed with GC worldwide each year. GC is more prevalent in less developed countries than in more developed countries. About half of all GC cases worldwide occur in East Asia, notably China. Globally, overall incidence rates of GC are declining, which is potentially attributed to a decrease in Helicobacter pylori (H. pylori) infection and the use of refrigeration to preserve foods rather than salt. GC is a multifactorial disease, and its occurrence and development were impacted by environmental and genetic factors. H. pylori infection is the primary risk factor for GC, especially for non-cardia. The prognosis of GC is poor due to stages at the first diagnosis. The 5-year survival rate is less than 10% when patients are diagnosed at an advanced stage, but the rate is as high as 85% if patients are detected at an earlier stage. Endoscopic screening can potentially prevent GC by early diagnosis and early treatment and has been widely adopted in screening programs in East Asian countries, such as Japan and Korea. This review summarizes updated epidemiological aspects, risk factors, and prevention strategies of GC in recent years to help researchers determine the most effective intervention strategies for reducing risk of GC.
Collapse
Affiliation(s)
- Lei Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Beijing Office for Cancer Prevention and Control, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xiangji Ying
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Center of Gastrointestinal Cancer, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Shuo Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Beijing Office for Cancer Prevention and Control, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Guoqing Lyu
- Department of Gastrointestinal Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Zekuan Xu
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Xi Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Beijing Office for Cancer Prevention and Control, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Huichao Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Beijing Office for Cancer Prevention and Control, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Qingyu Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Beijing Office for Cancer Prevention and Control, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Ning Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Beijing Office for Cancer Prevention and Control, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Jiafu Ji
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Center of Gastrointestinal Cancer, Peking University Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
18
|
Gao J, Ren W, Xiao C, Wang L, Huang Q, Zhang Z, Dang Y, Weng P, Wang H, Fang X, Zhuang M, Lin L, Chen S. Involvement of SLC39A6 in gastric adenocarcinoma and correlation of the SLC39A6 polymorphism rs1050631 with clinical outcomes after resection. BMC Cancer 2019; 19:1069. [PMID: 31703635 PMCID: PMC6839152 DOI: 10.1186/s12885-019-6222-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 09/30/2019] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND The single-nucleotide polymorphism SLC39A6 rs1050631 is strongly implicated in esophageal squamous cell carcinoma, leading us to question whether it may also play a role in gastric adenocarcima (GA). METHODS We genotyped the SLC39A6 rs1050631 in 512 patients who underwent GA resection. All study subjects lived in an area of China with high GA incidence. Genotypes were examined for possible correlation with survival and recurrence. The potential involvement of SLC39A6 in gastric cancer was explored in clinical samples and cell culture studies. RESULTS Multivariable analysis showed that patients with the CT + TT genotype at SLC39A6 rs1050631 were at greater risk of recurrence (hazard ratio, HR 1.387, p = 0.004) and death (HR 1.429, p = 0.002) than patients with CC genotype. Median recurrence-free and overall survival were significantly shorter in patients with the CT + TT genotype (20, 27 months) than in patients with the CC genotype (36, 43 months, p = 0.001, p < 0.001). Patients with the CT + TT genotype who were male or ≥ 60 years, or who had a tumor ≥5 cm or a moderately differentiated tumor were at significantly higher risk of recurrence and death. SLC39A6 was overexpressed in tissues from GA patients and in GA cell lines, and SLC39A6 knockdown in GA cell lines inhibited their proliferation, migration and invasion. CONCLUSION SLC39A6 rs1050631 correlates with post-resection prognosis of GA patients and SLC39A6 may participate in GA onset or progression.
Collapse
Affiliation(s)
- Jian Gao
- Department of General Surgery, Dongfang Hospital (900 Hospital of the Joint Logistics Team), 156 North Xi-er Huan Road, Fuzhou, 350025, Fujian, China
| | - Wenjun Ren
- Department of General Surgery, Dongfang Hospital (900 Hospital of the Joint Logistics Team), 156 North Xi-er Huan Road, Fuzhou, 350025, Fujian, China
| | - Chunhong Xiao
- Department of General Surgery, Dongfang Hospital (900 Hospital of the Joint Logistics Team), 156 North Xi-er Huan Road, Fuzhou, 350025, Fujian, China.,China Clinical Institute of Fuzhou General Hospital (900 Hospital of the Joint Logistics Team), Fujian Medical University, 156 North Xi-er Huan Road, Fuzhou, 350025, Fujian, China
| | - Lie Wang
- Department of General Surgery, Dongfang Hospital (900 Hospital of the Joint Logistics Team), 156 North Xi-er Huan Road, Fuzhou, 350025, Fujian, China.,China Clinical Institute of Fuzhou General Hospital (900 Hospital of the Joint Logistics Team), Fujian Medical University, 156 North Xi-er Huan Road, Fuzhou, 350025, Fujian, China
| | - Qiaojia Huang
- Department of Experimental Medicine, Dongfang Hospital (900 Hospital of the Joint Logistics Team), 156 North Xi-er Huan Road, Fuzhou, 350025, Fujian, China
| | - Zaizhong Zhang
- Department of General Surgery, Dongfang Hospital (900 Hospital of the Joint Logistics Team), 156 North Xi-er Huan Road, Fuzhou, 350025, Fujian, China.,China Clinical Institute of Fuzhou General Hospital (900 Hospital of the Joint Logistics Team), Fujian Medical University, 156 North Xi-er Huan Road, Fuzhou, 350025, Fujian, China
| | - Yuan Dang
- Fujian Meiya Aijiankang Health Management Co, Ltd. 4602#, Building 1, Shimao International Center, 108 Guangda Road, Fuzhou, 350025, Fujian, China
| | - Pengcheng Weng
- Union Medical College, Fujian Medical University, 29 XinQuan Road, Fuzhou, 350025, Fujian, China
| | - Hui Wang
- Union Medical College, Fujian Medical University, 29 XinQuan Road, Fuzhou, 350025, Fujian, China
| | - Xuehong Fang
- Union Medical College, Fujian Medical University, 29 XinQuan Road, Fuzhou, 350025, Fujian, China
| | - Minxian Zhuang
- Union Medical College, Fujian Medical University, 29 XinQuan Road, Fuzhou, 350025, Fujian, China
| | - Liying Lin
- Department of General Surgery, Dongfang Hospital (900 Hospital of the Joint Logistics Team), 156 North Xi-er Huan Road, Fuzhou, 350025, Fujian, China. .,China Clinical Institute of Fuzhou General Hospital (900 Hospital of the Joint Logistics Team), Fujian Medical University, 156 North Xi-er Huan Road, Fuzhou, 350025, Fujian, China.
| | - Shaoquan Chen
- Department of General Surgery, Dongfang Hospital (900 Hospital of the Joint Logistics Team), 156 North Xi-er Huan Road, Fuzhou, 350025, Fujian, China. .,China Clinical Institute of Fuzhou General Hospital (900 Hospital of the Joint Logistics Team), Fujian Medical University, 156 North Xi-er Huan Road, Fuzhou, 350025, Fujian, China.
| |
Collapse
|
19
|
Identification of different gene expressions between diffuse- and intestinal-type spheroid-forming gastric cancer cells. Gastric Cancer 2019; 22:967-979. [PMID: 30726523 DOI: 10.1007/s10120-019-00935-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 02/01/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Three-dimensional in vitro spheroid models are unique because they are considered for enrichment of specific cell populations with self-renewal ability. In this study, we explored the different mechanisms of gastric cancer spheroid-forming cells according to the Lauren classification. METHODS We isolated and enriched cells with self-renewal ability using spheroid-forming methods from gastric cancer cell lines. The expression of candidate target genes was investigated using western blot and qRT-PCR analysis. Lentiviral shRNA knockdown of target gene expression was performed and the effects on spheroid, colony forming, and tumorigenic ability were analyzed. RESULTS The SNU-638, SNU-484, MKN-28, and NCI-N87 successfully formed spheroid from single cell and enriched for self-renewal ability from 11 gastric cancer cell lines, including diffuse and intestinal types. The expression of SOX2 and E-cadherin increased in spheroid-forming cells in a diffuse-type cell line (SNU-638 and SNU-484), but not in the intestinal type (MKN-28 and NCI-N87). In contrast, ERBB3 expression was only increased in intestinal-type spheroid cells. The depletion of each candidate target gene expression suppressed self-renewal ability to grow as spheroids and colonies in a soft agar assay. In particular, down-regulated ERBB3 in the intestinal-type cell lines inhibited tumor growth in a mouse xenograft model. We found that high ERBB3 gene expression correlates with decreased survival in the intestinal type of gastric cancer. CONCLUSIONS Our results suggest that diffuse- and intestinal-type spheroid-forming cells express genes differently. Our data suggest that these candidate genes from spheroid-forming cells can be used in applications in targeted therapy.
Collapse
|
20
|
Signal V, Gurney J, Inns S, McLeod M, Sika-Paotonu D, Sowerbutts S, Teng A, Sarfati D. Helicobacter pylori, stomach cancer and its prevention in New Zealand. J R Soc N Z 2019. [DOI: 10.1080/03036758.2019.1650081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Virginia Signal
- Cancer and Chronic Conditions (C3) Research Group, Department of Public Health, University of Otago, Wellington, New Zealand
| | - Jason Gurney
- Cancer and Chronic Conditions (C3) Research Group, Department of Public Health, University of Otago, Wellington, New Zealand
| | - Stephen Inns
- Department of Medicine, University of Otago, Wellington, New Zealand
| | - Melissa McLeod
- Cancer and Chronic Conditions (C3) Research Group, Department of Public Health, University of Otago, Wellington, New Zealand
| | - Dianne Sika-Paotonu
- Department of Pathology & Molecular Medicine, University of Otago, Wellington, New Zealand
- Dean’s Department, University of Otago, Wellington, New Zealand
- Wesfarmers Centre for Vaccines & Infectious Diseases, Telethon Kids Institute, Perth, Australia
- Faculty of Health, Victoria University of Wellington, Wellington, New Zealand
| | - Sam Sowerbutts
- Department of Medicine, University of Otago, Wellington, New Zealand
| | - Andrea Teng
- Department of Public Health, University of Otago, Wellington, New Zealand
| | - Diana Sarfati
- Cancer and Chronic Conditions (C3) Research Group, Department of Public Health, University of Otago, Wellington, New Zealand
| |
Collapse
|
21
|
De Scalzi AM, Bonanni B, Galimberti V, Veronesi P, Pravettoni G, Corso G. E-cadherin germline mutations in Māori population. Future Oncol 2019; 15:1291-1294. [DOI: 10.2217/fon-2018-0834] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
| | - Bernardo Bonanni
- Division of Cancer Prevention & Genetics, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Viviana Galimberti
- Division of Breast Surgery, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Paolo Veronesi
- Division of Breast Surgery, European Institute of Oncology IRCCS, 20141 Milan, Italy
- Faculty of Medicine, University of Milan, Italy
| | - Gabriella Pravettoni
- Faculty of Medicine, University of Milan, Italy
- Applied Research Division for Cognitive and Psychological Science, European Institute of Oncology, 20141 Milan, Italy
| | - Giovanni Corso
- Division of Breast Surgery, European Institute of Oncology IRCCS, 20141 Milan, Italy
- Faculty of Medicine, University of Milan, Italy
| |
Collapse
|
22
|
Cavatorta O, Scida S, Miraglia C, Barchi A, Nouvenne A, Leandro G, Meschi T, De' Angelis GL, Di Mario F. Epidemiology of gastric cancer and risk factors. ACTA BIO-MEDICA : ATENEI PARMENSIS 2018; 89:82-87. [PMID: 30561423 PMCID: PMC6502220 DOI: 10.23750/abm.v89i8-s.7966] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Indexed: 12/16/2022]
Abstract
Gastric cancer is, still nowadays, an important healthcare problem worldwide. In Italy, it represents the fifth tumour by frequency in both men and women over 70 years old. A crucial point is represented by the percentage of early gastric cancers usually found, which is actually very low, and it carries to a worse morbidity and mortality. The most important focus in this oncological disease, is to perform an effective detection of the most common precancerous lesion linked with this neoplasia, chronic atrophic gastritis, in order to avoid the future outcome of gastric cancer itself. (www.actabiomedica.it)
Collapse
Affiliation(s)
- Ottavia Cavatorta
- Department of Medicine and Surgery, University of Parma, Parma, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Ichikawa H, Wakai T, Nagahashi M, Shimada Y, Hanyu T, Kano Y, Muneoka Y, Ishikawa T, Takizawa K, Tajima Y, Sakata J, Kobayashi T, Kemeyama H, Yabusaki H, Nakagawa S, Sato N, Kawasaki T, Homma K, Okuda S, Lyle S, Takabe K. Pathogenic germline BRCA1/2 mutations and familial predisposition to gastric cancer. JCO Precis Oncol 2018; 2. [PMID: 31608315 PMCID: PMC6788804 DOI: 10.1200/po.18.00097] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Affiliation(s)
- Hiroshi Ichikawa
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| | - Toshifumi Wakai
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| | - Masayuki Nagahashi
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| | - Yoshifumi Shimada
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| | - Takaaki Hanyu
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| | - Yosuke Kano
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| | - Yusuke Muneoka
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| | - Takashi Ishikawa
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| | - Kazuyasu Takizawa
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| | - Yosuke Tajima
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| | - Jun Sakata
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| | - Takashi Kobayashi
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| | - Hitoshi Kemeyama
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| | - Hiroshi Yabusaki
- Department of Gastroenterological Surgery, Niigata Cancer Center Hospital, 2-15-3 Kawagishi-cho, Chuo-ku, Niigata City, Niigata 951-8566, Japan
| | - Satoru Nakagawa
- Department of Gastroenterological Surgery, Niigata Cancer Center Hospital, 2-15-3 Kawagishi-cho, Chuo-ku, Niigata City, Niigata 951-8566, Japan
| | - Nobuaki Sato
- Department of Breast Oncology, Niigata Cancer Center Hospital, 2-15-3 Kawagishi-cho, Chuo-ku, Niigata City, Niigata 951-8566, Japan
| | - Takashi Kawasaki
- Department of Pathology, Niigata Cancer Center Hospital, 2-15-3 Kawagishi-cho, Chuo-ku, Niigata City, Niigata 951-8566, Japan
| | - Keiichi Homma
- Department of Pathology, Niigata Cancer Center Hospital, 2-15-3 Kawagishi-cho, Chuo-ku, Niigata City, Niigata 951-8566, Japan
| | - Shujiro Okuda
- Division of Bioinformatics, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| | - Stephen Lyle
- University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, Massachusetts 01655, USA
| | - Kazuaki Takabe
- Breast Surgery, Roswell Park Cancer Institute, Elm & Carlton Streets, Buffalo, New York 14263, USA.,Department of Surgery, University at Buffalo the State University of New York, 100 High Street, Buffalo New York 14203, USA
| |
Collapse
|
24
|
Abbas M, Faggian A, Sintali DN, Khan GJ, Naeem S, Shi M, Dingding C. Current and future biomarkers in gastric cancer. Biomed Pharmacother 2018; 103:1688-1700. [DOI: 10.1016/j.biopha.2018.04.178] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 04/24/2018] [Accepted: 04/24/2018] [Indexed: 02/06/2023] Open
|
25
|
Jing K, Mao Q, Ma P. Decreased expression of TROAP suppresses cellular proliferation, migration and invasion in gastric cancer. Mol Med Rep 2018; 18:3020-3026. [PMID: 29956806 DOI: 10.3892/mmr.2018.9230] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 11/11/2017] [Indexed: 11/06/2022] Open
Abstract
Trophinin associated protein (TROAP) is a cytoplasmic protein required for spindle assembly and cell invasion; however, its biological function in cancer remains to be elucidated. In the present study, by analyzing three independent datasets from the Oncomine database, it was identified that TROAP mRNA expression was upregulated in gastric cancer (GC) tissues compared with normal counterparts. Furthermore, elevated expression of TROAP was associated with poor survival in patients with GC, as predicted using Kaplan‑Meier analysis. TROAP was knocked down to verify its functional role in gastric cancer cell lines, SGC‑7901 and MGC80‑3. MTT assay was used to analyze cell proliferation. Cell cycle progression, and migration and invasion were determined using flow cytometry and Transwell assay, respectively. In vitro experiments demonstrated that knockdown of TROAP significantly suppressed cell proliferation, G1 to S cell cycle transition, and the migration and invasion ability of GC cells. The results of the present study suggest that TROAP is overexpressed in GC and serves an oncogenic role in gastric cancer by affecting cell proliferation and invasion.
Collapse
Affiliation(s)
- Ke Jing
- Department of Gastrointestinal Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Qinsheng Mao
- Department of Gastrointestinal Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Peng Ma
- Department of Gastrointestinal Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
26
|
SSAT State-of-the-Art Conference: Current Surgical Management of Gastric Tumors. J Gastrointest Surg 2018; 22:32-42. [PMID: 28808875 PMCID: PMC5742030 DOI: 10.1007/s11605-017-3533-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 07/31/2017] [Indexed: 01/31/2023]
Abstract
INTRODUCTION The current era of gastric surgery is marked by low morbidity and mortality rates, innovative strategies to approach resections with a minimally invasive fashion or hyperthermic intraperitoneal chemotherapy (HIPEC), as well as improved understanding of the biology of sporadic and hereditary stromal, neuroendocrine, and epithelial malignancies. METHODS In 2017, the Society for Surgery of the Alimentary Tract convened a State-of-the-Art Conference on Current Surgical Management of Gastric Tumors with both international experts and emerging leaders in the field of gastric surgery. RESULTS Martin D. McCarter, MD of the University of Colorado discussed the current management of gastric gastrointestinal stromal tumors (GIST). Kaitlyn J. Kelly, MD of the University of California, San Diego discussed the management of gastric carcinoid tumors. Jeffrey A. Norton of Stanford University discussed recent advances in the management of gastric adenocarcinoma including a focus on hereditary diffuse gastric cancer (HDGC). Joseph Kim, MD of Stony Brook University discussed a systematic approach to minimally invasive gastrectomy for cancer. Joyce Wong, MD of Pennsylvania State University discussed the role for cytoreductive surgery (CRS) and HIPEC for gastric adenocarcinoma. CONCLUSIONS This review provides gastrointestinal surgeons with a concise update on the current surgical management of gastric tumors.
Collapse
|
27
|
Zhang H, Feng M, Feng Y, Bu Z, Li Z, Jia S, Ji J. Germline mutations in hereditary diffuse gastric cancer. Chin J Cancer Res 2018; 30:122-130. [PMID: 29545726 DOI: 10.21147/j.issn.1000-9604.2018.01.13] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer is one of the leading causes of cancer-related deaths worldwide. Among which, about 1%-3% of gastric cancer patients were characterized by inherited gastric cancer predisposition syndromes, knowing as hereditary diffuse gastric cancer (HDGC). Studies reported that CDH1 germline mutations are the main cause of HDGC. With the help of rapid development of genetic testing technologies and data analysis tools, more and more researchers focus on seeking candidate susceptibility genes for hereditary cancer syndromes. In addition, National Comprehensive Cancer Network (NCCN) guidelines recommend that the patients of HDGC carrying CDH1 mutations should undergo prophylactic gastrectomy or routine endoscopic surveillances. Therefore, genetic counseling plays a key role in helping individuals with pathogenic mutations make appropriate risk management plans. Moreover, experienced and professional genetic counselors as well as a systematic multidisciplinary team (MDT) are also required to facilitate the development of genetic counseling and benefit pathogenic mutation carriers who are in need of regular and standardized risk management solutions. In this review, we provided an overview about the germline mutations of several genes identified in HDGC, suggesting that these genes may potentially act as susceptibility genes for this malignant cancer syndrome. Furthermore, we introduced information for prevention, diagnosis and risk management of HDGC. Investigations on key factors that may have effect on risk management decision-making and genetic data collection of more cancer syndrome family pedigrees are required for the development of HDGC therapeutic strategies.
Collapse
Affiliation(s)
- Hao Zhang
- Surgery Laboratory, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China.,Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Molecular Diagnostics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Mengmeng Feng
- Surgery Laboratory, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China
| | - Yi Feng
- Surgery Laboratory, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China
| | - Zhaode Bu
- Surgery Laboratory, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China
| | - Ziyu Li
- Surgery Laboratory, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China
| | - Shuqin Jia
- Surgery Laboratory, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China.,Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Molecular Diagnostics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Jiafu Ji
- Surgery Laboratory, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China
| |
Collapse
|
28
|
Strong VE, Russo A, Yoon SS, Brennan MF, Coit DG, Zheng CH, Li P, Huang CM. Comparison of Young Patients with Gastric Cancer in the United States and China. Ann Surg Oncol 2017; 24:3964-3971. [DOI: 10.1245/s10434-017-6073-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2023]
|
29
|
Total Gastrectomy for Hereditary Diffuse Gastric Cancer at a Single Center: Postsurgical Outcomes in 41 Patients. Ann Surg 2017; 266:1006-1012. [PMID: 27759617 DOI: 10.1097/sla.0000000000002030] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE The aim of this study was to describe postoperative outcomes of total gastrectomy at our institution for patients with hereditary diffuse gastric cancer (HDGC). BACKGROUND HDGC, which is mainly caused by germline mutations in the E-cadherin gene (CDH1), renders a lifetime risk of gastric cancer of up to 70%, prompting a recommendation for prophylactic total gastrectomy. METHODS A prospective gastric cancer database identified 41 patients with CDH1 mutation who underwent total gastrectomy during 2005 to 2015. Perioperative, histopathologic, and long-term data were collected. RESULTS Of the 41 patients undergoing total gastrectomy, median age was 47 years (range 20 to 71). There were 14 men and 27 women, with 25 open operations and 16 minimally invasive operations. Median length of stay was 7 days (range 4 to 50). In total, 11 patients (27%) experienced a complication requiring intervention, and there was 1 peri-operative mortality (2.5%). Thirty-five patients (85%) demonstrated 1 or more foci of intramucosal signet ring cell gastric cancer in the examined specimen. At 16 months median follow-up, the median weight loss was 4.7 kg (15% of preoperative weight). By 6 to 12 months postoperatively, weight patterns stabilized. Overall outcome was reported to be "as expected" by 40% of patients and "better than expected" by 45%. Patient-reported outcomes were similar to those of other patients undergoing total gastrectomy. CONCLUSION Total gastrectomy should be considered for all CDH1 mutation carriers because of the high risk of invasive diffuse-type gastric cancer and lack of reliable surveillance options. Although most patients have durable weight loss after total gastrectomy, weights stabilize at about 6 to 12 months postoperatively, and patients report outcomes as being good to better than their preoperative expectations. No patients have developed gastric cancer recurrence after resections.
Collapse
|
30
|
Min KW, Kwon MJ, Kim DH, Son BK, Kim EK, Oh YH, Wi YC. Persistent elevation of postoperative neutrophil-to-lymphocyte ratio: A better predictor of survival in gastric cancer than elevated preoperative neutrophil-to-lymphocyte ratio. Sci Rep 2017; 7:13967. [PMID: 29070814 PMCID: PMC5656603 DOI: 10.1038/s41598-017-13969-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 09/25/2017] [Indexed: 02/06/2023] Open
Abstract
Postoperative neutrophil-to-lymphocyte ratio change (NLRc) reflects the dynamic change of balance between host inflammatory response and immune response after treatment. In gastric cancer, an elevated initial NLR (iNLR) is reported to be a prognostic predictor, but the clinical application of the NLRc remains unclear. The NLRc was assessed in 734 patients undergoing total/subtotal gastrectomy and endoscopic submucosal dissection for gastric adenocarcinoma. The iNLR and NLRc were recorded within 10 days of the first diagnosis and 3–6 months after surgery, respectively. Using receiver operating characteristic (ROC) curves, we investigated the relationship between NLRc or iNLR and patient survival. The analysis revealed a higher predictive power for correlating patient survival with the NLRc compared with iNLR. NLRc was defined as negative (lower than iNLR) and positive (higher than iNLR). A positive NLRc was frequently observed in patients with advanced AJCC stage, local recurrence, distant metastasis, perineural invasion, and adjuvant chemotherapy (all p < 0.05). Univariate and multivariate analyses revealed a significant relationship between patient survival and NLRc (all p < 0.05) but no association between survival and iNLR. The NLRc could be a better indicator than iNLR for predicting survival in patients with gastric cancer.
Collapse
Affiliation(s)
- Kyueng-Whan Min
- Department of Pathology, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Gyeonggi-do, Republic of Korea
| | - Mi Jung Kwon
- Department of Pathology, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Gyeonggi-do, Republic of Korea
| | - Dong-Hoon Kim
- Department of Pathology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| | - Byoung Kwan Son
- Departments of Internal medicine, Eulji Hospital, Eulji University School of medicine, Seoul, Republic of Korea.
| | - Eun-Kyung Kim
- Department of Pathology, Eulji Hospital, Eulji University School of medicine, Seoul, Republic of Korea
| | - Young Ha Oh
- Department of Pathology, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Gyeonggi-do, Republic of Korea
| | - Young Chan Wi
- Department of Pathology, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Gyeonggi-do, Republic of Korea
| |
Collapse
|
31
|
Diagnostic, Predictive, Prognostic, and Therapeutic Molecular Biomarkers in Third Millennium: A Breakthrough in Gastric Cancer. BIOMED RESEARCH INTERNATIONAL 2017; 2017:7869802. [PMID: 29094049 PMCID: PMC5637861 DOI: 10.1155/2017/7869802] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 07/12/2017] [Indexed: 02/08/2023]
Abstract
Introduction Gastric cancer is the fifth most common cancer and the third cause of cancer death. The clinical outcomes of the patients are still not encouraging with a low rate of 5 years' survival. Often the disease is diagnosed at advanced stages and this obviously negatively affects patients outcomes. A deep understanding of molecular basis of gastric cancer can lead to the identification of diagnostic, predictive, prognostic, and therapeutic biomarkers. Main Body This paper aims to give a global view on the molecular classification and mechanisms involved in the development of the tumour and on the biomarkers for gastric cancer. We discuss the role of E-cadherin, HER2, fibroblast growth factor receptor (FGFR), MET, human epidermal growth factor receptor (EGFR), hepatocyte growth factor receptor (HGFR), mammalian target of rapamycin (mTOR), microsatellite instability (MSI), PD-L1, and TP53. We have also considered in this manuscript new emerging biomarkers as matrix metalloproteases (MMPs), microRNAs, and long noncoding RNAs (lncRNAs). Conclusions Identifying and validating diagnostic, prognostic, predictive, and therapeutic biomarkers will have a huge impact on patients outcomes as they will allow early detection of tumours and also guide the choice of a targeted therapy based on specific molecular features of the cancer.
Collapse
|
32
|
Hallowell N, Lawton J, Badger S, Richardson S, Hardwick RH, Caldas C, Fitzgerald RC. The Psychosocial Impact of Undergoing Prophylactic Total Gastrectomy (PTG) to Manage the Risk of Hereditary Diffuse Gastric Cancer (HDGC). J Genet Couns 2017; 26:752-762. [PMID: 27837291 DOI: 10.1007/s10897-016-0045-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 10/26/2016] [Indexed: 12/13/2022]
Abstract
Individuals identified as at high risk of developing Hereditary Diffuse Gastric Cancer (HDGC) are advised to undergo prophylactic surgery - have their stomach removed - in their early twenties. Research with (older) cancer patients who undergo gastrectomy for curative reasons suggests that gastric resection has a number of physical and psychosocial sequelae. Because it is difficult to extrapolate the findings of studies of older cancer patients to younger healthy patients who are considering prophylactic total gastrectomy (PTG), the aim of this qualitative interview study was to determine the psychosocial implications of undergoing prophylactic surgery to manage genetic risk. Fourteen men and 13 women from the UK's Familial Gastric Cancer study who had undergone PTG were invited to participate in qualitative interviews. Most reported that undergoing surgery and convalescence was easier than anticipated. There was evidence that age affected experiences of PTG, with younger patients tending to report faster recovery times and more transient aftereffects. All saw the benefits of risk reduction as outweighing the costs of surgery. Surgery was described as having a range of physical impacts (disrupted appetite, weight loss, fatigue, GI symptoms) that had related psychological, social and economic implications. Those considering PTG need to be aware that its impact on quality of life is difficult to predict and negative sequelae may be ongoing for some individuals.
Collapse
Affiliation(s)
- Nina Hallowell
- Ethox Centre, Nuffield Department of Population Health, University of Oxford, Old Road, Oxford, OX3 7LF, UK.
| | - Julia Lawton
- Centre for Population Health Sciences, University of Edinburgh, Edinburgh, UK
| | - Shirlene Badger
- PHG Foundation, Cambridge, UK
- Institute of Public Health, University of Cambridge, Cambridge, UK
| | | | - Richard H Hardwick
- Cambridge University Hospitals Trust, Addenbrookes Hospital, Cambridge, UK
| | - Carlos Caldas
- Cambridge University Hospitals Trust, Addenbrookes Hospital, Cambridge, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Rebecca C Fitzgerald
- Cambridge University Hospitals Trust, Addenbrookes Hospital, Cambridge, UK
- MRC Cancer Unit, University of Cambridge, Cambridge, UK
| |
Collapse
|
33
|
Shah MA. Future Directions in Improving Outcomes for Patients with Gastric and Esophageal Cancer. Hematol Oncol Clin North Am 2017; 31:545-552. [PMID: 28501093 DOI: 10.1016/j.hoc.2017.01.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
"This issue of Hematology/Oncology Clinics of North America provides an update to the current understanding of the physiology of gastric and esophageal cancers and the state-of-the-art management of disease. Over the past 10 years, we have witnessed dramatic changes in both our understanding of the disease and its management. We have 2 new biological agents approved to treat advanced disease, with several more prospects under development. In this article, the author looks to the future, attempting to answer the question of which advancements will play the biggest role in improving patient outcomes in this still-devastating disease.
Collapse
Affiliation(s)
- Manish A Shah
- Weill Cornell Medicine/New York-Presbyterian Hospital, Division of Hematology and Medical Oncology, 1305 York Avenue, New York, NY 10021, USA.
| |
Collapse
|
34
|
Wilson RD, Langlois S. Facteurs génétiques à prendre en considération dans le cadre de l'examen gynécologique annuel. JOURNAL OF OBSTETRICS AND GYNAECOLOGY CANADA 2017; 38:S22-S33. [PMID: 28063537 DOI: 10.1016/j.jogc.2016.09.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIF Offrir aux médecins un survol des troubles génétiques courants qui devraient être pris en considération dans le cadre de l'examen gynécologique annuel d'une patiente, et ce, afin de déterminer le risque que court celle-ci ou d'en venir à procéder à des examens particuliers ou à orienter la patiente vers un autre service de sous-spécialité, en fonction de ses antécédents personnels ou familiaux. OPTIONS Ces renseignements d'ordre génétique peuvent être utilisés aux fins de la sensibilisation des patientes et du dépistage ou du diagnostic de possibles maladies et/ou mutations. ISSUES L'utilisation de ces renseignements d'ordre génétique pourrait mener à l'amélioration de l'évaluation des risques et des avantages et à celle de la prise en charge dans le cadre de l'examen gynécologique annuel. RéSULTATS: Les études publiées en anglais, jusques et y compris en mai 2010, ont été récupérées par l'intermédiaire de recherches menées dans PubMed et la Cochrane Library au moyen d'un vocabulaire contrôlé (« gynaecological diagnosis », « genetic inheritance ») et de mots clés (« genetic risk », « genetic mutation », « inheritance », « family history », « uterus », « ovary », « endometrial », « vagina », « colon », « gastric », « renal », « breast », « cardiac », « thrombophilia », « diabetes », « epilepsy », « leiomyomata uteri ») appropriés. D'autres sources ont été identifiées par l'intermédiaire de recherches menées dans les sites Web d'organismes s'intéressant à l'évaluation des technologies dans le domaine de la santé et d'organismes connexes, dans des collections de directives cliniques, dans des registres d'essais cliniques et auprès de sociétés de spécialité médicale nationales et internationales. VALEURS Le niveau des résultats ne permet pas la formulation de recommandations factuelles. AVANTAGES, DéSAVANTAGES ET COûTS: La présente opinion de comité améliorera l'utilisation de nouvelles connaissances génétiques et leur application aux soins gynécologiques offerts annuellement aux femmes. Les occasions de gestion du risque et de diagnostic, pour ce qui est des troubles gynécologiques génétiques, s'en trouveront améliorées. Une compréhension plus exhaustive des troubles génétiques pourrait entraîner une hausse de l'anxiété et du stress psychologique chez les femmes et les membres de leur famille. COMMANDITAIRE Société des obstétriciens et gynécologues du Canada. RECOMMANDATIONS Le niveau des résultats ne permet pas la formulation de recommandations factuelles.
Collapse
|
35
|
Hallowell N, Badger S, Richardson S, Caldas C, Hardwick RH, Fitzgerald RC, Lawton J. An investigation of the factors effecting high-risk individuals' decision-making about prophylactic total gastrectomy and surveillance for hereditary diffuse gastric cancer (HDGC). Fam Cancer 2016; 15:665-76. [PMID: 27256430 PMCID: PMC5935221 DOI: 10.1007/s10689-016-9910-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hereditary diffuse gastric cancer has an early onset and poor prognosis, therefore, individuals who carry a pathogenic (CDH1) mutation in the E-cadherin gene (CDH1) are offered endoscopic surveillance and advised to undergo prophylactic total gastrectomy (PTG) in their early to mid-twenties. Patients not ready or fit to undergo gastrectomy, or in whom the genetic testing result is unknown or ambiguous, are offered surveillance. Little is known about the factors that influence decisions to undergo or decline PTG, making it difficult to provide optimal support for those facing these decisions. Qualitative interviews were carried out with 35 high-risk individuals from the Familial Gastric Cancer Study in the UK. Twenty-seven had previously undergone PTG and eight had been identified as carrying a pathogenic CDH1 mutation but had declined surgery at the time of interview. The interviews explored the experience of decision-making and factors influencing risk-management decisions. The data suggest that decisions to proceed with PTG are influenced by a number of potentially competing factors: objective risk confirmation by genetic testing and/or receiving a positive biopsy; perceived familial cancer burden and associated risk perceptions; perceptions of post-surgical life; an increasing inability to tolerate endoscopic procedures; a concern that surveillance could miss a cancer developing and individual's life stage. These findings have implications for advising this patient group.
Collapse
Affiliation(s)
- Nina Hallowell
- Ethox Centre, Nuffield Department of Population Health, University of Oxford, Oxford, UK.
| | - Shirlene Badger
- PHG Foundation and Institute of Public Health, University of Cambridge, Cambridge, UK
| | | | - Carlos Caldas
- Cambridge University Hospitals Trust, Addenbrookes Hospital, Cambridge, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Richard H Hardwick
- Cambridge University Hospitals Trust, Addenbrookes Hospital, Cambridge, UK
| | - Rebecca C Fitzgerald
- Cambridge University Hospitals Trust, Addenbrookes Hospital, Cambridge, UK
- MRC Cancer Unit, University of Cambridge, Cambridge, UK
| | - Julia Lawton
- Centre for Population Health Sciences, University of Edinburgh, Cambridge, UK
| |
Collapse
|
36
|
Zhuang W. MiR-155-5p inhibits proliferation and invasion of gastric cancer cells by downregulation of SOX4. Shijie Huaren Xiaohua Zazhi 2016; 24:3476-3481. [DOI: 10.11569/wcjd.v24.i23.3476] [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] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the mechanism of miR-155-5p to inhibit the proliferation and invasion of gastric cancer (GC) cells.
METHODS: The expression of miR-155-5p in GC cells was detected by quantitative real-time PCR. The miR-155-5p mimics and inhibitor were designed and transfected into BGC-823 cells. Then, the role of miR-155-5p in GC cell proliferation and invasion in vitro was explored. Luciferase reporter assay and Western blot were performed to confirm the target gene of miR-155-5p.
RESULTS: MiR-155-5p expression was significantly down-regulated in GC cells (P < 0.05). Overexpression of miR-155-5p inhibited GC cell proliferation and invasion in vitro, while down-regulated expression of miR-155-5p promoted GC cell proliferation and invasion. SOX4 was identified as a target of miR-155-5p in GC cells, and SOX4 expression levels were inversely correlated with miR-155-5p. Knockdown of Sox4 inhibited the proliferation and invasion of GC cells (P < 0.05).
CONCLUSION: MiR-155-5p could inhibit GC cell proliferation and invasion partially by down-regulating SOX4. MiR-155-5p might be a potential therapeutic target for GC treatment in the future.
Collapse
|
37
|
Baniak N, Senger JL, Ahmed S, Kanthan SC, Kanthan R. Gastric biomarkers: a global review. World J Surg Oncol 2016; 14:212. [PMID: 27514667 PMCID: PMC4982433 DOI: 10.1186/s12957-016-0969-3] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 08/02/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Gastric cancer is an aggressive disease with a poor 5-year survival and large global burden of disease. The disease is biologically and genetically heterogeneous with a poorly understood carcinogenesis at the molecular level. Despite the many prognostic, predictive, and therapeutic biomarkers investigated to date, gastric cancer continues to be detected at an advanced stage with resultant poor clinical outcomes. MAIN BODY This is a global review of gastric biomarkers with an emphasis on HER2, E-cadherin, fibroblast growth factor receptor, mammalian target of rapamycin, and hepatocyte growth factor receptor as well as sections on microRNAs, long noncoding RNAs, matrix metalloproteinases, PD-L1, TP53, and microsatellite instability. CONCLUSION A deeper understanding of the pathogenesis and biological features of gastric cancer, including the identification and characterization of diagnostic, prognostic, predictive, and therapeutic biomarkers, hopefully will provide improved clinical outcomes.
Collapse
Affiliation(s)
- Nick Baniak
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, 103 Hospital Drive, Saskatoon, SK S7N 0W8 Canada
| | - Jenna-Lynn Senger
- Department of Surgery, University of Alberta, 116 St & 85 Ave, Edmonton, T6G 2R3, T6G 2B7 AB Canada
| | - Shahid Ahmed
- Division of Medical Oncology, University of Saskatchewan, 103 Hospital Drive, Saskatoon, SK S7N 0W8 Canada
| | - S. C. Kanthan
- Department of General Surgery, University of Saskatchewan, 103 Hospital Drive, Saskatoon, SK S7N 0W8 Canada
| | - Rani Kanthan
- Department of General Surgery, University of Saskatchewan, 103 Hospital Drive, Saskatoon, SK S7N 0W8 Canada
| |
Collapse
|
38
|
Davidson M, Smyth EC, Cunningham D. Clinical role of ramucirumab alone or in combination with paclitaxel for gastric and gastro-esophageal junction adenocarcinoma. Onco Targets Ther 2016; 9:4539-48. [PMID: 27524910 PMCID: PMC4966750 DOI: 10.2147/ott.s84153] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cancers of the stomach and gastro-esophageal junction represent a significant challenge in oncology. Despite some recent advances in genetic categorization and the development of novel agents, outcomes remain poor. The vascular endothelial growth factor receptor 2 monoclonal antibody ramucirumab is the first targeted therapy to improve survival in a molecularly unselected population, and represents a valuable new treatment option. This review describes the current treatment landscape for advanced disease, evaluates existing and ongoing research into ramucirumab, and discusses its current and potential future therapeutic role.
Collapse
Affiliation(s)
- Michael Davidson
- Department of Gastrointestinal Cancer Research, The Royal Marsden Hospital NHS Foundation Trust, London, UK
| | - Elizabeth C Smyth
- Department of Gastrointestinal Cancer Research, The Royal Marsden Hospital NHS Foundation Trust, London, UK
| | - David Cunningham
- Department of Gastrointestinal Cancer Research, The Royal Marsden Hospital NHS Foundation Trust, London, UK
| |
Collapse
|
39
|
Kim GH, Liang PS, Bang SJ, Hwang JH. Screening and surveillance for gastric cancer in the United States: Is it needed? Gastrointest Endosc 2016; 84:18-28. [PMID: 26940296 DOI: 10.1016/j.gie.2016.02.028] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 02/19/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS Although the incidence of gastric cancer in the United States is relatively low, the incidence of gastric cancer is higher than for esophageal cancer, for which clear guidelines for screening and surveillance exist. With the increasing availability of endoscopic therapy, such as endoscopic submucosal dissection, for treating advanced dysplasia and early gastric cancer, establishing guidelines for screening and surveillance of patients who are at high risk of developing gastric cancer has the potential to diagnose and treat gastric cancer at an earlier stage and improve mortality from gastric cancer. The aims of this article were to review the data regarding the risk factors for developing gastric cancer, methods for gastric cancer screening, and results of national screening programs. METHODS A review of the existing literature related to the aims was performed. RESULTS Risk factors for gastric cancer that were identified include race/ethnicity (East Asian, Russian, or South American), first-degree relative diagnosed with gastric cancer, positive Helicobacter pylori status, and presence of atrophic gastritis or intestinal metaplasia. Endoscopy has the highest rate of detecting gastric cancer compared with other gastric cancer screening methods. The national screening program in Japan has demonstrated a mortality reduction from gastric cancer based on cohort data. CONCLUSIONS Gastric cancer screening with endoscopy should be considered in individuals who are immigrants from regions associated with a high risk of gastric cancer (East Asia, Russia, or South America) or who have a family history of gastric cancer. Those with findings of atrophic gastritis or intestinal metaplasia on screening endoscopy should undergo surveillance endoscopy every 1 to 2 years. Large prospective multicenter studies are needed to further identify additional risk factors for developing gastric cancer and to assess whether gastric cancer screening programs for high-risk populations in the United States would result in improved mortality.
Collapse
Affiliation(s)
- Gwang Ha Kim
- Department of Internal Medicine, Pusan National University School of Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
| | - Peter S Liang
- Division of Gastroenterology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Sung Jo Bang
- Department of Internal Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Joo Ha Hwang
- Division of Gastroenterology, Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
40
|
Chang KK, Cho SJ, Yoon C, Lee JH, Park DJ, Yoon SS. Increased RhoA Activity Predicts Worse Overall Survival in Patients Undergoing Surgical Resection for Lauren Diffuse-Type Gastric Adenocarcinoma. Ann Surg Oncol 2016; 23:4238-4246. [PMID: 27364501 DOI: 10.1245/s10434-016-5357-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Indexed: 01/16/2023]
Abstract
BACKGROUND Several studies have reported a high rate of RHOA mutations in the Lauren diffuse-type gastric adenocarcinoma (GA) but not in intestinal-type GA. The aim of this study was to determine if RhoA activity is prognostic for overall survival (OS) in patients with resectable GA. METHODS Retrospective review was performed on a prospective database of GA patients who underwent potentially curative resection between 2003 and 2012 at a single institution. Tissue microarrays were constructed from surgical specimens and analyzed for phosphorylated RhoA, a marker of inactive RhoA signaling. OS was estimated by the Kaplan-Meier method, and multivariate analysis was performed by Cox proportional hazards regression modeling. RESULTS One hundred thirty-six patients with diffuse-type GA and 129 patients with intestinal-type GA were examined. Compared to intestinal-type GA, diffuse-type GA tumors were significantly associated with increased tumor size and advanced tumor, node, metastasis (TNM) classification system stage. In patients with diffuse-type GA, high RhoA activity was associated with significantly worse OS when compared to low RhoA activity (5-year OS 52.5 vs. 81.0 %, p = 0.017). This difference in OS was not observed in patients with intestinal-type GA (5-year OS 83.9 vs. 81.6 %, p = 0.766). On multivariate analysis of diffuse-type GA patients, high RhoA activity was an independent negative prognostic factor for OS (hazard ratio 2.38, 95 % confidence interval 1.07-5.28). CONCLUSIONS Increased RhoA activity is predictive of worse OS in patients with diffuse-type GA who undergo potentially curative surgical resection. Along with findings from genomic studies, these results suggest RhoA may be a novel therapeutic target in diffuse-type GA.
Collapse
Affiliation(s)
- Kevin K Chang
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Soo-Jeong Cho
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Changhwan Yoon
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jun Ho Lee
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Do Joong Park
- Department of Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, South Korea
| | - Sam S Yoon
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
41
|
Mesic A, Rogar M, Hudler P, Juvan R, Komel R. Association of the AURKA and AURKC gene polymorphisms with an increased risk of gastric cancer. IUBMB Life 2016; 68:634-44. [PMID: 27270838 DOI: 10.1002/iub.1521] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 05/18/2016] [Indexed: 12/19/2022]
Abstract
Single nucleotide polymorphisms (SNPs) in mitotic checkpoint genes can contribute to susceptibility of human cancer, including gastric cancer (GC). We aimed to investigate the effects of Aurora kinase A (AURKA), Aurora kinase B (AURKB), and Aurora kinase C (AURKC) gene polymorphisms on GC risk in Slovenian population. We genotyped four SNPs in AURKA (rs2273535 and rs1047972), AURKB (rs2241909), and AURKC (rs758099) in a total of 128 GC patients and 372 healthy controls using TaqMan allelic discrimination assays to evaluate their effects on GC risk. Our results showed that genotype frequencies between cases and controls were significantly different for rs1047972 and rs758099 (P < 0.05). Our study demonstrated that AURKA rs1047972 TT and (CC + CT) genotypes were significantly associated with an increased risk of gastric cancer. Our results additionally revealed that AURKC rs758099 TT and (CC + CT) genotypes were also associated with increased GC risk. In stratified analysis, genotypes TT and (CC + CT) of AURKA rs1047972 SNP were associated with increased risk of both, intestinal and diffuse, types of GC. In addition, AURKC rs758099 TT and (CC + CT) genotypes were positively associated with increased intestinal type GC risk, but not with an increased diffuse type GC risk. Based on these results, we can conclude that AURKA rs1047972 and AURKC rs758099 polymorphisms could affect the risk of GC development. Further larger studies are needed to confirm these findings. © 2016 IUBMB Life, 68(8):634-644, 2016.
Collapse
Affiliation(s)
- Aner Mesic
- Department of Biology, Faculty of Science, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Marija Rogar
- Medical Centre for Molecular Biology, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Petra Hudler
- Medical Centre for Molecular Biology, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Robert Juvan
- Clinical Department for Abdominal Surgery, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Radovan Komel
- Medical Centre for Molecular Biology, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
42
|
Seol SY, Kim C, Lim JY, Yoon SO, Hong SW, Kim JW, Choi SH, Cho JY. Overexpression of Endoplasmic Reticulum Oxidoreductin 1-α (ERO1L) Is Associated with Poor Prognosis of Gastric Cancer. Cancer Res Treat 2016; 48:1196-1209. [PMID: 26987398 PMCID: PMC5080819 DOI: 10.4143/crt.2015.189] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 02/17/2016] [Indexed: 12/14/2022] Open
Abstract
Purpose Gastric cancer is the second leading cause of cancer-related death worldwide. Although surgery is the standard curative treatment for gastric cancer, relapse occurs in a large number of patients, except in the case of early diagnosed gastric cancer. Following previous studies that identified endoplasmic reticulum oxidoreductin 1-α (ERO1L) as a potential marker for gastric cancer, we investigated the functional role of ERO1L in gastric cancer. Materials and Methods For validation of microarray data, the mRNA expression level of ERO1L was measured by quantitative real-time reverse transcription polymerase chain reaction in 56 independent stage III gastric cancer patients. Immunohistochemical staining was performed to examine the protein expression level of ERO1L in 231 gastric cancer patients. Correlation between gene expression and cancer prognosis was evaluated. Results Patients with high ERO1L expression had poorer survival than those with low expression (p < 0.01). Functional assays demonstrated that ERO1L knockdown inhibited cell proliferation, migration, invasion, and chemoresistance. In addition, involvement of inactivation of Akt and JNK signaling in molecular mechanisms of ERO1L inhibition was demonstrated. Conclusion High expression of ERO1L is associated with poor prognosis of patients with gastric cancer. These results indicate that ERO1L expression may be a clinically promising therapeutic target for prevention of gastric cancer.
Collapse
Affiliation(s)
- So-Young Seol
- Department of Medical Oncology, Gangnam Severance Cancer Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Chul Kim
- Samsung Bioepis Co., Ltd., Incheon, Korea
| | - Jae Yun Lim
- Department of Medical Oncology, Gangnam Severance Cancer Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Sun Och Yoon
- Department of Pathology, Gangnam Severance Cancer Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Soon Won Hong
- Department of Pathology, Gangnam Severance Cancer Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jong Won Kim
- Department of Surgery, Gangnam Severance Cancer Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Seung Ho Choi
- Department of Surgery, Gangnam Severance Cancer Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jae Yong Cho
- Department of Medical Oncology, Gangnam Severance Cancer Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
43
|
Yoon C, Cho SJ, Aksoy BA, Park DJ, Schultz N, Ryeom SW, Yoon SS. RETRACTED: Chemotherapy Resistance in Diffuse-Type Gastric Adenocarcinoma Is Mediated by RhoA Activation in Cancer Stem-Like Cells. Clin Cancer Res 2016; 22:971-83. [PMID: 26482039 PMCID: PMC4823002 DOI: 10.1158/1078-0432.ccr-15-1356] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 09/28/2015] [Indexed: 12/21/2022]
Abstract
PURPOSE The Lauren diffuse type of gastric adenocarcinoma (DGA), as opposed to the intestinal type (IGA), often harbors mutations in RHOA, but little is known about the role of RhoA in DGA. EXPERIMENTAL DESIGN We examined RhoA activity and RhoA pathway inhibition in DGA cell lines and in two mouse xenograft models. RhoA activity was also assessed in patient tumor samples. RESULTS RhoA activity was higher in DGA compared with IGA cell lines and was further increased when grown as spheroids to enrich for cancer stem-like cells (CSCs) or when sorted using the gastric CSC marker CD44. RhoA shRNA or the RhoA inhibitor Rhosin decreased expression of the stem cell transcription factor, Sox2, and decreased spheroid formation by 78% to 81%. DGA spheroid cells had 3- to 5-fold greater migration and invasion than monolayer cells, and this activity was Rho-dependent. Diffuse GA spheroid cells were resistant in a cytotoxicity assay to 5-fluorouracil and cisplatin chemotherapy, and this resistance could be reversed with RhoA pathway inhibition. In two xenograft models, cisplatin inhibited tumor growth by 40% to 50%, RhoA inhibition by 32% to 60%, and the combination by 77% to 83%. In 288 patient tumors, increased RhoA activity correlated with worse overall survival in DGA patients (P = 0.017) but not in IGA patients (P = 0.612). CONCLUSIONS RhoA signaling promotes CSC phenotypes in DGA cells. Increased RhoA activity is correlated with worse overall survival in DGA patients, and RhoA inhibition can reverse chemotherapy resistance in DGA CSC and in tumor xenografts. Thus, the RhoA pathway is a promising new target in DGA patients.
Collapse
Affiliation(s)
- Changhwan Yoon
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Soo-Jeong Cho
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Bülent Arman Aksoy
- Computational Biology Center, Memorial Sloan-Kettering Cancer Center, New York, New York. Tri-Institutional Training Program in Computational Biology and Medicine, New York, New York
| | - Do Joong Park
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Nikolaus Schultz
- Computational Biology Center, Memorial Sloan-Kettering Cancer Center, New York, New York. Tri-Institutional Training Program in Computational Biology and Medicine, New York, New York
| | - Sandra W Ryeom
- Department of Cancer Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Sam S Yoon
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York.
| |
Collapse
|
44
|
Xu K, Zhao YC. MEF2D/Wnt/β-catenin pathway regulates the proliferation of gastric cancer cells and is regulated by microRNA-19. Tumour Biol 2016; 37:9059-69. [PMID: 26762410 DOI: 10.1007/s13277-015-4766-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 12/29/2015] [Indexed: 01/06/2023] Open
Abstract
The underlying molecular pathogenesis in gastric cancer remains poorly unknown. The transcription factor myocyte enhancer factor 2D (MEF2D) participates in the initiation and development of many human cancers. However, its potential roles in gastric cancer have surprisingly not been studied. In present study, we first explored MEF2's expression in gastric cancer, finding that only MEF2D rather than MEF2A, 2B, or 2C was elevated in gastric cancer clinical specimens. Furthermore, immunohistochemical analysis on the tissue samples obtained from 260 patients with gastric cancer revealed that MEF2D expression was significantly associated with the clinical stage, vascular invasion, metastasis, and tumor size. Gastric cancer patients with MEF2D expression showed a significantly shorter overall survival time compared with that of patients lacking of MEF2D. Multivariate analysis revealed that MEF2D expression was an independent prognostic factor for overall survival. These results indicated that MEF2D was a prognostic marker for gastric cancer. Notably, MEF2D silencing was able to reduce the proliferation and survival of gastric cancer cells. Further study revealed that MEF2D suppression significantly inactivated the oncogenic Wnt/β-catenin pathway. Downregulation of MEF2D inhibited the tumorigenesis of gastric cancer cells in nude mice. Finally, MEF2D is a direct target of miR-19, which was found to be decreased in gastric cancer clinical specimens. Collectively, we found that miR-19/MEF2D/Wnt/β-catenin regulatory network contributes to the growth of gastric cancer, hinting a new promising target for gastric cancer treatment.
Collapse
Affiliation(s)
- Kai Xu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying-Chao Zhao
- Department of Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
45
|
Recapitulating Human Gastric Cancer Pathogenesis: Experimental Models of Gastric Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 908:441-78. [PMID: 27573785 DOI: 10.1007/978-3-319-41388-4_22] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This review focuses on the various experimental models to study gastric cancer pathogenesis, with the role of genetically engineered mouse models (GEMMs) used as the major examples. We review differences in human stomach anatomy compared to the stomachs of the experimental models, including the mouse and invertebrate models such as Drosophila and C. elegans. The contribution of major signaling pathways, e.g., Notch, Hedgehog, AKT/PI3K is discussed in the context of their potential contribution to foregut tumorigenesis. We critically examine the rationale behind specific GEMMs, chemical carcinogens, dietary promoters, Helicobacter infection, and direct mutagenesis of relevant oncogenes and tumor suppressor that have been developed to study gastric cancer pathogenesis. Despite species differences, more efficient and effective models to test specific genes and pathways disrupted in human gastric carcinogenesis have yet to emerge. As we better understand these species differences, "humanized" versions of mouse models will more closely approximate human gastric cancer pathogenesis. Towards that end, epigenetic marks on chromatin, the gut microbiota, and ways of manipulating the immune system will likely move center stage, permitting greater overlap between rodent and human cancer phenotypes thus providing a unified progression model.
Collapse
|
46
|
Sun Y, Huang XJ, Chen L. New progress in study of risk factors for gastric cancer. Shijie Huaren Xiaohua Zazhi 2015; 23:4831-4837. [DOI: 10.11569/wcjd.v23.i30.4831] [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] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer is one of the most common malignant tumors, and its etiology is not clear yet. Numerous studies show that the development of gastric cancer is a complex process related with many factors, such as demographic, lifestyle and diet, infectious, hereditary, socioeconomic, and mental factors. Early prevention can effectively reduce the incidence of gastric cancer. Here we make a review of the new progress in research of risk factors for gastric cancer.
Collapse
|
47
|
Sugimoto S, Komatsu H, Morohoshi Y, Kanai T. Recognition of and recent issues in hereditary diffuse gastric cancer. J Gastroenterol 2015; 50:831-43. [PMID: 26049741 DOI: 10.1007/s00535-015-1093-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 05/21/2015] [Indexed: 02/04/2023]
Abstract
In East Asian countries, gastric cancer incidence is high, but detection rates for germline CDH1 mutations that cause hereditary diffuse gastric cancers (HDGCs) are low. Consequently, screens and genetic testing for HDGC are often considered unimportant. Since the first germline truncating CDH1 mutations in Japanese patients were reported, some HDGC cases have been reported, and some of these involve large germline rearrangements and de novo mutation of CDH1. New methods for mutation detection--such as multiplex ligation-dependent probe amplification, array comparative genomic hybridization, and exome sequencing--have become available, as have new experimental models, including novel gene-knockout mice and gastric organoids. Because of these advances, searches for candidate genes (e.g., CTNNA1, MAP3K6) and our understanding of HDGC pathogenesis have improved in recent years; moreover, there have been substantial changes in the field since the current HDGC consensus guidelines were released. This review focuses on recent issues and advances in the study of HDGC. For example, lobular breast cancer cases and de novo occurrences of DGC are unlikely to meet the existing criteria for genetic testing, but current evidence indicates that some such cases may be good candidates for genetic testing. It is important to recognize that HDGC is a syndrome and that lobular breast cancer can be the first manifestation of this syndrome. CDH1 testing, including analyses of large genomic rearrangements, should be recommended even in countries where few HDGC cases have been reported.
Collapse
Affiliation(s)
- Shinya Sugimoto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan,
| | | | | | | |
Collapse
|
48
|
Kim DY, Lee JH, Kim KY, Kang DB, Park WC, Chae SC, Lee JK. Association between genetic polymorphisms in cortactin and susceptibility to gastric cancer. Ann Surg Treat Res 2015; 89:74-80. [PMID: 26236696 PMCID: PMC4518033 DOI: 10.4174/astr.2015.89.2.74] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 03/12/2015] [Accepted: 03/19/2015] [Indexed: 01/14/2023] Open
Abstract
PURPOSE Overexpression of cortactin (CTTN) in human tumors has been proposed to result in increased cell migration and metastatic potential. Here, we determined the frequencies of CTTN g.-9101C>T, g.-8748C>T, and g.72C>T polymorphisms in apparently healthy subjects and gastric cancer patients, respectively, and the influence of the CTTN polymorphisms on gastric cancer susceptibility. METHODS Blood samples were collected from 267 patients and 533 controls. CTTN g.-8748C>T and g.-9101C>T polymorphisms were determined using polymerase chain reaction-restriction fragment length polymorphism; the g.72C>T polymorphism was determined using the TaqMan method. RESULTS Genotype frequencies of the CTTN g.-9101C>T polymorphism were 97.5% (TT), 2.5% (TC), and 0% (CC) in the patient group, and 98.6% (TT), 1.4% (TC), and 0% (CC) in the control group. Genotype frequencies of the CTTN g.-8748C>T polymorphism were 93.3% (TT), 6.8% (TC), and 0% (CC) in the patient group, and 94.2% (TT), 5.8% (TC), and 0% (CC) in the control group. Genotype frequencies of the CTTN g.72C>T polymorphism were 82.4% (CC), 17.2% (CT), and 0.4% (TT) in the patient group, and 78.0% (CC), 20.1% (CT), and 1.9% (TT) in the control group. Genotype and allele frequencies of the CTTN g.-9101C>T polymorphism differed significantly between the advanced gastric cancer and control groups. Patients with advanced gastric cancer, possessing the TC genotype, had a significantly poorer prognosis than the group with the TT genotype. CONCLUSION The CTTN g.-9101C>T polymorphism might influence advanced gastric cancer susceptibility. However, the role of the CTTN g.-9101C>T, g.-8748C>T, and g.72C>T polymorphisms requires careful interpretation and confirmation through larger studies.
Collapse
Affiliation(s)
- Dae Yong Kim
- Department of Surgery, Institute of Medical Science, Wonkwang University School of Medicine, Iksan, Korea
| | - Joo Hyun Lee
- Department of Surgery, Institute of Medical Science, Wonkwang University School of Medicine, Iksan, Korea
| | - Keun Young Kim
- Department of Surgery, Institute of Medical Science, Wonkwang University School of Medicine, Iksan, Korea
| | - Dong Baek Kang
- Department of Surgery, Institute of Medical Science, Wonkwang University School of Medicine, Iksan, Korea
| | - Won Cheol Park
- Department of Surgery, Institute of Medical Science, Wonkwang University School of Medicine, Iksan, Korea
| | - Soo Cheon Chae
- Department of Pathology, Institute of Medical Science, Wonkwang University School of Medicine, Iksan, Korea
| | - Jeong Kyun Lee
- Department of Surgery, Institute of Medical Science, Wonkwang University School of Medicine, Iksan, Korea
| |
Collapse
|
49
|
Abstract
Cancers of the stomach and esophagus are among the most challenging cancers of the GI tract to treat, associated with poor median survivals for metastatic disease and significant, sometimes prolonged, deteriorations in patient performance status as the diseases progress. However, in the past decade, we have begun to better understand disease biology and carcinogenesis, leading to the identification of subtypes of these diseases. There is also an increasing awareness of the global heterogeneity of disease and its impact on drug development. Our improved understanding of the molecular underpinnings of gastric and esophageal cancers has been accompanied with the development of novel therapeutic strategies. Recent actively investigated targets in this disease include human epidermal growth factor receptor 2, angiogenesis, MET, and immune checkpoint inhibition, with approvals of two new targeted agents, trastuzumab and ramucirumab. Improvements in our ability to deliver cytotoxic therapy, which is better tolerated and allows patients an opportunity to benefit from second- and more advanced lines of therapy, have also been observed. In this review, the current state-of-the-art management of advanced and metastatic gastric and esophageal adenocarcinomas, specifically highlighting the development of targeted therapies in these diseases, is described.
Collapse
Affiliation(s)
- Manish A Shah
- From Weill Cornell Medical College of Cornell University, New York-Presbyterian Hospital, New York, NY.
| |
Collapse
|
50
|
Oliveira C, Pinheiro H, Figueiredo J, Seruca R, Carneiro F. Familial gastric cancer: genetic susceptibility, pathology, and implications for management. Lancet Oncol 2015; 16:e60-70. [PMID: 25638682 DOI: 10.1016/s1470-2045(14)71016-2] [Citation(s) in RCA: 258] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Familial gastric cancer comprises at least three major syndromes: hereditary diffuse gastric cancer, gastric adenocarcinoma and proximal polyposis of the stomach, and familial intestinal gastric cancer. The risk of development of gastric cancer is high in families affected b-y these syndromes, but only hereditary diffuse gastric cancer is genetically explained (caused by germline alterations of CDH1, which encodes E-cadherin). Gastric cancer is also associated with a range of several cancer-associated syndromes with known genetic causes, such as Lynch, Li-Fraumeni, Peutz-Jeghers, hereditary breast-ovarian cancer syndromes, familial adenomatous polyposis, and juvenile polyposis. We present contemporary knowledge on the genetics, pathogenesis, and clinical features of familial gastric cancer, and discuss research and technological developments, which together are expected to open avenues for new genetic testing approaches and novel therapeutic strategies.
Collapse
Affiliation(s)
- Carla Oliveira
- Ipatimub-Institute of Molecular Pathology and Immunology & Instituto Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal; Department of Pathology and Oncology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Hugo Pinheiro
- Ipatimub-Institute of Molecular Pathology and Immunology & Instituto Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
| | - Joana Figueiredo
- Ipatimub-Institute of Molecular Pathology and Immunology & Instituto Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
| | - Raquel Seruca
- Ipatimub-Institute of Molecular Pathology and Immunology & Instituto Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal; Department of Pathology and Oncology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Fátima Carneiro
- Ipatimub-Institute of Molecular Pathology and Immunology & Instituto Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal; Department of Pathology and Oncology, Faculty of Medicine, University of Porto, Porto, Portugal; Centro Hospitalar S João, Porto, Portugal.
| |
Collapse
|