1
|
Ahmed RA, Aldalbahi AA, Alhumaidan NI, Alotaibi TA, Alharbi MA, Alharbi MA, Alzahrani MMM, Althobaiti AA, Alzelfawi L, Almouaalamy NA. An approach to COVID‑19 and oncology: From impact, staging and management to vaccine outcomes in cancer patients: A systematic review and meta‑analysis. Exp Ther Med 2025; 29:37. [PMID: 39776889 PMCID: PMC11705223 DOI: 10.3892/etm.2024.12787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/15/2024] [Indexed: 01/11/2025] Open
Abstract
The COVID-19 pandemic has had a global impact, with >771 million confirmed cases and 6 million deaths reported by October 2023. Cancer patients, due to their immunosuppressed status, face an increased infection risk and higher COVID-19 complications. The present study aimed to assess clinical outcomes in COVID-19-infected cancer patients, focusing on mortality rates and other aspects, providing valuable insight for better protection and outcomes. This systematic review was conducted by searching the PubMed, Cochrane and Embase databases from August 2023 following the PRISMA guidelines. Studies from 2020 to 2023 pertaining to the impact of COVID-19 on patients previously diagnosed with malignancies were considered. Inclusion criteria entailed a pre-existing malignancy diagnosis, confirmed COVID-19 infection and an impact of COVID-19 on any aspect of the patient's cancer management. Studies written in English were exclusively reviewed. Post-COVID-19 malignancy diagnoses, case reports, review articles and data-insufficient studies were excluded. Screening and consensus on eligibility were carried out by a team of four authors, with disputes resolved by a non-screening author. Data extraction was performed by a five-author team, detailing study and population characteristics, as well as cancer patient outcomes related to COVID-19. Cross-checking was conducted by the same team, with conflicts resolved by a third author. The review of 27 studies explored COVID-19's impact on oncology, revealing diverse sample sizes (1,807,559 to 177 participants). Studies spanned various cancer types, including gastric adenocarcinoma, breast, lung, gynecologic, colorectal and non-melanoma skin cancer. Mortality rates were higher among cancer patients with COVID-19 compared to those without. Gastric adenocarcinoma exhibited a 5.9% mortality rate. Thoracic cancer patients faced elevated mortality and gastrectomies decreased. A meta-analysis (10 studies, 5,151 patients) showed a 19.1% mortality rate for COVID-19-infected cancer patients, contrasting with 1% for non-COVID-19 cancer patients (5 studies, 54,528 patients). The odds ratio for mortality in non-COVID-19 vs. COVID-19 cancer patients was 0.1036 (3 studies, 3,496 patients). Cancer patients consistently faced elevated mortality during the pandemic, with specific cancers showing unique impacts. Gastric adenocarcinoma exhibited a significant COVID-19 mortality rate. Patients with thoracic cancer faced increased risks, influencing surgical trends. Meta-analysis revealed an overall elevated mortality rate among COVID-19-infected cancer patients compared to non-COVID-19 counterparts.
Collapse
Affiliation(s)
- Ruqayyah Ali Ahmed
- Department of Medicine and Surgery, Batterjee Medical College for Science and Technology, Jeddah 21442, Saudi Arabia
| | | | | | | | | | - Mohammed A. Alharbi
- College of Medicine, Imam Abdulrahman bin Faisal University, Dammam 31441, Saudi Arabia
| | | | | | - Lama Alzelfawi
- College of Medicine, Princess Noura University, Riyadh 11564, Saudi Arabia
| | - Nabil A. Almouaalamy
- Oncology Department, Princess Noorah Oncology Center, King Abdul Aziz Medical City, Ministry of National Guard-Health Affairs, King Abdullah International Medical Research Centre, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Makkah-Jeddah Highway Road, Jeddah 22384, Saudi Arabia
| |
Collapse
|
2
|
Wu Z, Yang X, Yuan Z, Guo Y, Wang X, Qu L. Identification of a novel histone acetylation-related long non-coding RNA model combined with qRT-PCR experiments for prognosis and therapy in gastric cancer. Heliyon 2024; 10:e36615. [PMID: 39263162 PMCID: PMC11387370 DOI: 10.1016/j.heliyon.2024.e36615] [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: 10/28/2023] [Revised: 08/12/2024] [Accepted: 08/19/2024] [Indexed: 09/13/2024] Open
Abstract
Gastric cancer (GC) is considered a global health crisis due to the scarcity of early diagnostic methods. Numerous studies have substantiated the involvement of histone acetylation imbalance in the progression of diverse tumor types. The potential roles of long non-coding RNA (lncRNA) in improving prognostic, predictive as well as therapeutic approaches in cancers have made it a major hotspot in recent years. Nevertheless, existent studies have never concerned the prognostic and clinical value of histone acetylation-related lncRNAs (HARlncs) in GC. Based on the aforementioned rationale, we developed a prognostic model incorporating four HARlncs-AC114730.1, AL445250.1, LINC01778, and AL163953.1-which demonstrated potential as an independent predictor of prognosis. Subsequently, GC patients were stratified into high-risk and low-risk groups. The low-risk group exhibited significantly higher overall survival (OS) compared to the high-risk group. Based on the analyses of the tumor microenvironment (TME) and immune responses, significant differences were observed between the two risk groups in terms of immune cell infiltration, immune checkpoint (ICP) expression, and other TME alterations. Furthermore, the sensitivity of GC patients to some chemotherapeutic drugs and the discrepant biological behaviors of three tumor clusters were studied in this model. In summary, we developed an effective HARlncs model with the objective of offering novel prognostic prediction methods and identifying potential therapeutic targets for GC patients.
Collapse
Affiliation(s)
- Zhixuan Wu
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, 318000, China
| | - Xuejia Yang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Ziwei Yuan
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Yangyang Guo
- Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, 318000, China
| | - Xiaowu Wang
- Department of Burns and Skin Repair Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Ruian, 325200, China
- Department of Thyroid and Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, 315000, China
| | - Liangchen Qu
- Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, 318000, China
| |
Collapse
|
3
|
Iglesias G, Talavera E, Troya J, Díaz-Álvarez A, García-Remesal M. Artificial intelligence model for tumoral clinical decision support systems. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 253:108228. [PMID: 38810378 DOI: 10.1016/j.cmpb.2024.108228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 04/21/2024] [Accepted: 05/13/2024] [Indexed: 05/31/2024]
Abstract
BACKGROUND AND OBJECTIVE Comparative diagnostic in brain tumor evaluation makes possible to use the available information of a medical center to compare similar cases when a new patient is evaluated. By leveraging Artificial Intelligence models, the proposed system is able of retrieving the most similar cases of brain tumors for a given query. The primary objective is to enhance the diagnostic process by generating more accurate representations of medical images, with a particular focus on patient-specific normal features and pathologies. A key distinction from previous models lies in its ability to produce enriched image descriptors solely from binary information, eliminating the need for costly and difficult to obtain tumor segmentation. METHODS The proposed model uses Artificial Intelligence to detect patient features to recommend the most similar cases from a database. The system not only suggests similar cases but also balances the representation of healthy and abnormal features in its design. This not only encourages the generalization of its use but also aids clinicians in their decision-making processes. This generalization makes possible for future research in different medical diagnosis areas with almost not any change in the system. RESULTS We conducted a comparative analysis of our approach in relation to similar studies. The proposed architecture obtains a Dice coefficient of 0.474 in both tumoral and healthy regions of the patients, which outperforms previous literature. Our proposed model excels at extracting and combining anatomical and pathological features from brain Magnetic Resonances (MRs), achieving state-of-the-art results while relying on less expensive label information. This substantially reduces the overall cost of the training process. Our findings highlight the significant potential for improving the efficiency and accuracy of comparative diagnostics and the treatment of tumoral pathologies. CONCLUSIONS This paper provides substantial grounds for further exploration of the broader applicability and optimization of the proposed architecture to enhance clinical decision-making. The novel approach presented in this work marks a significant advancement in the field of medical diagnosis, particularly in the context of Artificial Intelligence-assisted image retrieval, and promises to reduce costs and improve the quality of patient care using Artificial Intelligence as a support tool instead of a black box system.
Collapse
Affiliation(s)
- Guillermo Iglesias
- Departamento de Sistemas Informáticos, Escuela Técnica Superior de Ingeniería de Sistemas Informáticos, Universidad Politécnica de Madrid, Spain.
| | - Edgar Talavera
- Departamento de Sistemas Informáticos, Escuela Técnica Superior de Ingeniería de Sistemas Informáticos, Universidad Politécnica de Madrid, Spain.
| | - Jesús Troya
- Infanta Leonor University Hospital. Madrid., Spain
| | - Alberto Díaz-Álvarez
- Departamento de Sistemas Informáticos, Escuela Técnica Superior de Ingeniería de Sistemas Informáticos, Universidad Politécnica de Madrid, Spain.
| | - Miguel García-Remesal
- Biomedical Informatics Group, Departamento de Inteligencia Artificial, Escuela Técnica Superior de Ingenieros Informáticos, Universidad Politécnica de Madrid, Spain.
| |
Collapse
|
4
|
Li J, Liu Y, Zhou P, Fan QQ, Liu HM. Mass spectrometry-based pseudotargeted metabolomics reveals metabolic variations in a2-induced gastric cancer cell. EUROPEAN JOURNAL OF MASS SPECTROMETRY (CHICHESTER, ENGLAND) 2024; 30:199-206. [PMID: 38656147 DOI: 10.1177/14690667241248444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Gastric cancer (GC) is one of the most malignant tumors with high morbidity and mortality in the world. Compound a2, a Jiyuan oridonin derivative, exhibited excellent anti-proliferative activity against GC cells. To investigate the gastric cellular response to a2 therapy as a novel drug candidate, we adopted a pseudotargeted metabolomics method to explore metabolic variation in a2-induced MGC-803 gastric cells using liquid chromatography tandem mass spectrometry combined with multivariate statistical analysis. The results showed that a2 treatment induced significant metabolic changes in the levels of aminoacyl-tRNA biosynthesis, alanine, aspartate and glutamate metabolism, pyrimidine metabolism, and tricarboxylic acid cycle, approximately 80% of the metabolites were down-regulated in the low-dose and high-dose groups including aspartate, tryptophan, sedoheptulose 7-phosphate, succinate, 2'-deoxyadenosine, uridine, cytidine, etc. which can provide evidence for a new therapy of GC.
Collapse
Affiliation(s)
- Juan Li
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou, China
| | - Ying Liu
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, China
| | - Piao Zhou
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, China
| | - Qi-Qi Fan
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, China
| | - Hong-Min Liu
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
5
|
Díaz Del Arco C, Fernández Aceñero MJ, Ortega Medina L. Molecular Classifications in Gastric Cancer: A Call for Interdisciplinary Collaboration. Int J Mol Sci 2024; 25:2649. [PMID: 38473896 DOI: 10.3390/ijms25052649] [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: 01/11/2024] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Gastric cancer (GC) is a heterogeneous disease, often diagnosed at advanced stages, with a 5-year survival rate of approximately 20%. Despite notable technological advancements in cancer research over the past decades, their impact on GC management and outcomes has been limited. Numerous molecular alterations have been identified in GC, leading to various molecular classifications, such as those developed by The Cancer Genome Atlas (TCGA) and the Asian Cancer Research Group (ACRG). Other authors have proposed alternative perspectives, including immune, proteomic, or epigenetic-based classifications. However, molecular stratification has not yet transitioned into clinical practice for GC, and little attention has been paid to alternative molecular classifications. In this review, we explore diverse molecular classifications in GC from a practical point of view, emphasizing their relationships with clinicopathological factors, prognosis, and therapeutic approaches. We have focused on classifications beyond those of TCGA and the ACRG, which have been less extensively reviewed previously. Additionally, we discuss the challenges that must be overcome to ensure their impact on patient treatment and prognosis. This review aims to serve as a practical framework to understand the molecular landscape of GC, facilitate the development of consensus molecular categories, and guide the design of innovative molecular studies in the field.
Collapse
Affiliation(s)
- 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
| | - 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
| | - Luis Ortega Medina
- 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
|
6
|
Wen F, Zhao F, Huang W, Liang Y, Sun R, Lin Y, Zhang W. A novel ferroptosis-related gene signature for overall survival prediction in patients with gastric cancer. Sci Rep 2024; 14:4422. [PMID: 38388534 PMCID: PMC10883968 DOI: 10.1038/s41598-024-53515-0] [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: 08/18/2022] [Accepted: 02/01/2024] [Indexed: 02/24/2024] Open
Abstract
The global diagnosis rate and mortality of gastric cancer (GC) are among the highest. Ferroptosis and iron-metabolism have a profound impact on tumor development and are closely linked to cancer treatment and patient's prognosis. In this study, we identified six PRDEGs (prognostic ferroptosis- and iron metabolism-related differentially expressed genes) using LASSO-penalized Cox regression analysis. The TCGA cohort was used to establish a prognostic risk model, which allowed us to categorize GC patients into the high- and the low-risk groups based on the median value of the risk scores. Our study demonstrated that patients in the low-risk group had a higher probability of survival compared to those in the high-risk group. Furthermore, the low-risk group exhibited a higher tumor mutation burden (TMB) and a longer 5-year survival period when compared to the high-risk group. In summary, the prognostic risk model, based on the six genes associated with ferroptosis and iron-metabolism, performs well in predicting the prognosis of GC patients.
Collapse
Affiliation(s)
- Fang Wen
- Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
- College of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Fan Zhao
- Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
- College of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Wenjie Huang
- Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Yan Liang
- Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
- College of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Ruolan Sun
- Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
- College of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Yize Lin
- Clinical Laboratory Department, Hospital of the Office of the People's Government of the Tibet Autonomous Region in Chengdu, Chengdu, 850015, Sichuan, China
| | - Weihua Zhang
- Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China.
- College of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China.
| |
Collapse
|
7
|
Xiu Z, Yang Q, Xie F, Han F, He W, Liao W. Revolutionizing digestive system tumor organoids research: Exploring the potential of tumor organoids. J Tissue Eng 2024; 15:20417314241255470. [PMID: 38808253 PMCID: PMC11131411 DOI: 10.1177/20417314241255470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 05/02/2024] [Indexed: 05/30/2024] Open
Abstract
Digestive system tumors are the leading cause of cancer-related deaths worldwide. Despite ongoing research, our understanding of their mechanisms and treatment remain inadequate. One promising tool for clinical applications is the use of gastrointestinal tract tumor organoids, which serve as an important in vitro model. Tumor organoids exhibit a genotype similar to the patient's tumor and effectively mimic various biological processes, including tissue renewal, stem cell, and ecological niche functions, and tissue response to drugs, mutations, or injury. As such, they are valuable for drug screening, developing novel drugs, assessing patient outcomes, and supporting immunotherapy. In addition, innovative materials and techniques can be used to optimize tumor organoid culture systems. Several applications of digestive system tumor organoids have been described and have shown promising results in related aspects. In this review, we discuss the current progress, limitations, and prospects of this model for digestive system tumors.
Collapse
Affiliation(s)
- Zhian Xiu
- Department of Medical Laboratory, Clinical Medical College, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| | - Qian Yang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fusheng Xie
- Department of Medical Laboratory, Clinical Medical College, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| | - Feng Han
- Department of Medical Laboratory, Clinical Medical College, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| | - Weiwei He
- Department of Medical Laboratory, Clinical Medical College, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| | - Weifang Liao
- Department of Medical Laboratory, Clinical Medical College, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| |
Collapse
|
8
|
Zhang C, Huang W, Xu X, Zuo S. Flexible endoscopic instrument for diagnosis and treatment of early gastric cancer. Med Biol Eng Comput 2023; 61:2815-2828. [PMID: 37608080 DOI: 10.1007/s11517-023-02911-1] [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: 11/15/2022] [Accepted: 08/14/2023] [Indexed: 08/24/2023]
Abstract
Gastric cancer is a common cancer endangering human life and health worldwide. Early detection and diagnosis of gastric cancer that is normally performed by flexible endoscope can significantly improve the survival rate of patients. However, current endoscopic instruments have some problems, such as limitation of degrees of freedom (DOFs) and lack of surgical triangulation. Meanwhile, the lack of an intraoperative technique for the real-time evaluation of early gastric cancer is also a serious problem. To solve these problems, we have developed a dual-bending flexible endoscopic instrument for the diagnosis and treatment of early gastric cancer. This instrument has a compact structure with a maximum outer diameter of 3 mm and an insertion length of 1220 mm. It has 5 DOFs with a dual-bending function, which can form a surgical operation triangulation to easily perform the endoscopic procedure. Apart from the surgical forceps, the end of the instrument can be equipped with different endoscopic devices to meet the needs of diagnosis and treatment, such as endomicroscopic probes, electrosurgical knives, and laser ablation optical fibers. It is verified that the instrument can carry these devices to complete corresponding tasks, demonstrating the great potential of this instrument in clinical applications.
Collapse
Affiliation(s)
- Chi Zhang
- Key Laboratory of Mechanism Theory and Equipment Design of Ministry of Education, Tianjin University, Tianjin, 300072, China
| | - Weihao Huang
- Key Laboratory of Mechanism Theory and Equipment Design of Ministry of Education, Tianjin University, Tianjin, 300072, China
| | - Xingfeng Xu
- Key Laboratory of Mechanism Theory and Equipment Design of Ministry of Education, Tianjin University, Tianjin, 300072, China
| | - Siyang Zuo
- Key Laboratory of Mechanism Theory and Equipment Design of Ministry of Education, Tianjin University, Tianjin, 300072, China.
| |
Collapse
|
9
|
Huang RJ, Laszkowska M, In H, Hwang JH, Epplein M. Controlling Gastric Cancer in a World of Heterogeneous Risk. Gastroenterology 2023; 164:736-751. [PMID: 36706842 PMCID: PMC10270664 DOI: 10.1053/j.gastro.2023.01.018] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 01/12/2023] [Accepted: 01/17/2023] [Indexed: 01/29/2023]
Abstract
Gastric cancer (GC) is a leading cause of global mortality but also a cancer whose footprint is highly unequal. This review aims to define global disease epidemiology, critically appraise strategies of prevention and disease attenuation, and assess how these strategies could be applied to improve outcomes from GC in a world of variable risk and disease burden. Strategies of primary prevention focus on improving the detection and eradication of the main environmental risk factor, Helicobacter pylori. In certain countries of high incidence, endoscopic or radiographic screening of the asymptomatic general population has been adopted as a means of secondary prevention. By contrast, identification and targeted surveillance of individuals with precancerous lesions (such as intestinal metaplasia) is being increasingly embraced in nations of low incidence. This review also highlights existing knowledge gaps in GC prevention as well as the role of emerging technologies for early detection and risk stratification.
Collapse
Affiliation(s)
- Robert J Huang
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California.
| | - Monika Laszkowska
- Gastroenterology, Hepatology, and Nutrition Service, Department of Subspecialty Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Haejin In
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Joo Ha Hwang
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California
| | - Meira Epplein
- Duke University, Department of Population Health Sciences, and Cancer Risk, Detection, and Interception Program, Duke Cancer Institute, Durham, North Carolina
| |
Collapse
|
10
|
A nucleolin-activated polyvalent aptamer nanoprobe for the detection of cancer cells. Anal Bioanal Chem 2023; 415:2217-2226. [PMID: 36864310 DOI: 10.1007/s00216-023-04629-3] [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: 01/07/2023] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 03/04/2023]
Abstract
Sensitive detection of cancer cells plays a critical role in early cancer diagnosis. Nucleolin, overexpressed on the surface of cancer cells, is regarded as a candidate biomarker for cancer diagnosis. Thus, cancer cells can be detected through the detection of membrane nucleolin. Herein, we designed a nucleolin-activated polyvalent aptamer nanoprobe (PAN) to detect cancer cells. In brief, a long single-stranded DNA with many repeated sequences was synthesized through rolling circle amplification (RCA). Then the RCA product acted as a scaffold chain to combine with multiple AS1411 sequences, which was doubly modified with fluorophore and quenching group, respectively. The fluorescence of PAN was initially quenched. Upon binding to target protein, the conformation of PAN changed, leading to the recovery of fluorescence. The fluorescence signal of cancer cells treated with PAN was much brighter compared with that of monovalent aptamer nanoprobes (MAN) at the same concentration. Furthermore, the binding affinity of PAN to B16 cells was proved to be 30 times higher than that of MAN by calculating the dissociation constants. The results indicated that PAN could specifically detect target cells, and this design concept has potential to become promising in cancer diagnosis.
Collapse
|
11
|
Guo Y, Guo L, Su Y, Xiong Y. CRISPR-Cas system manipulating nanoparticles signal transduction for cancer diagnosis. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1851. [PMID: 36199268 DOI: 10.1002/wnan.1851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 07/30/2022] [Accepted: 08/10/2022] [Indexed: 11/05/2022]
Abstract
Early diagnosis of cancer is important to improve the survival rate and relieve patient pain. Sensitive detection of cancer related biomarkers in body fluids is a critical approach for the early diagnosis of cancer. The clustered regularly interspaced short palindromic repeat-associated protein (CRISPR-Cas) system has emerged as a molecular manipulation technology because of its simple detection procedure, high base resolution, and isothermal signal amplification. Recently, various nanomaterials with unique optical and electrical characteristics have been introduced as the novel signal transducers to enhance the detection performance of CRISPR-Cas-based nanosensors. This review summarizes the working mechanisms of the CRISPR-Cas system for biosensing. It also enumerates the strategies of CRISPR-manipulated nanosensors based on various signal models for cancer diagnosis, including colorimetric, fluorescence, electrochemical, electrochemiluminescence, pressure, and other signals. Finally, the prospects and challenges of CRISPR-Cas-based nanosensors for cancer diagnostic are also discussed. This article is categorized under: Diagnostic Tools > Biosensing.
Collapse
Affiliation(s)
- Yuqian Guo
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, People's Republic of China
| | - Liang Guo
- Jiangxi-OAI Joint Research Institute, Nanchang University, Nanchang, People's Republic of China
| | - Yu Su
- School of Food Science and Technology, Nanchang University, Nanchang, People's Republic of China
| | - Yonghua Xiong
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, People's Republic of China.,School of Food Science and Technology, Nanchang University, Nanchang, People's Republic of China
| |
Collapse
|
12
|
Deng S, Gu J, Jiang Z, Cao Y, Mao F, Xue Y, Wang J, Dai K, Qin L, Liu K, Wu K, He Q, Cai K. Application of nanotechnology in the early diagnosis and comprehensive treatment of gastrointestinal cancer. J Nanobiotechnology 2022; 20:415. [PMID: 36109734 PMCID: PMC9479390 DOI: 10.1186/s12951-022-01613-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 08/30/2022] [Indexed: 02/08/2023] Open
Abstract
Gastrointestinal cancer (GIC) is a common malignant tumour of the digestive system that seriously threatens human health. Due to the unique organ structure of the gastrointestinal tract, endoscopic and MRI diagnoses of GIC in the clinic share the problem of low sensitivity. The ineffectiveness of drugs and high recurrence rates in surgical and drug therapies are the main factors that impact the curative effect in GIC patients. Therefore, there is an urgent need to improve diagnostic accuracies and treatment efficiencies. Nanotechnology is widely used in the diagnosis and treatment of GIC by virtue of its unique size advantages and extensive modifiability. In the diagnosis and treatment of clinical GIC, surface-enhanced Raman scattering (SERS) nanoparticles, electrochemical nanobiosensors and magnetic nanoparticles, intraoperative imaging nanoparticles, drug delivery systems and other multifunctional nanoparticles have successfully improved the diagnosis and treatment of GIC. It is important to further improve the coordinated development of nanotechnology and GIC diagnosis and treatment. Herein, starting from the clinical diagnosis and treatment of GIC, this review summarizes which nanotechnologies have been applied in clinical diagnosis and treatment of GIC in recent years, and which cannot be applied in clinical practice. We also point out which challenges must be overcome by nanotechnology in the development of the clinical diagnosis and treatment of GIC and discuss how to quickly and safely combine the latest nanotechnology developed in the laboratory with clinical applications. Finally, we hope that this review can provide valuable reference information for researchers who are conducting cross-research on GIC and nanotechnology.
Collapse
Affiliation(s)
- Shenghe Deng
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Junnan Gu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Zhenxing Jiang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Yinghao Cao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Fuwei Mao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Yifan Xue
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Jun Wang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Kun Dai
- Department of Neonatal Intensive Care Unit, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Le Qin
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Ke Liu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Ke Wu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Qianyuan He
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China.
| | - Kailin Cai
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China.
| |
Collapse
|
13
|
Han Y, Yoo HJ, Jee SH, Lee JH. High serum levels of L-carnitine and citric acid negatively correlated with alkaline phosphatase are detectable in Koreans before gastric cancer onset. Metabolomics 2022; 18:62. [PMID: 35900644 DOI: 10.1007/s11306-022-01922-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 07/14/2022] [Indexed: 10/16/2022]
Abstract
INTRODUCTION Monitoring metabolic biomarkers could be utilized as an effective tool for the early detection of gastric cancer (GC) risk. OBJECTIVE We aimed to discover predictive serum biomarkers for GC and investigate biomarker-related metabolism. METHODS Subjects were randomly selected from the Korean Cancer Prevention Study-II cohort and matched by age and sex. We analyzed baseline serum samples of 160 subjects (discovery set; control and GC occurrence group, 80 each) via nontargeted screening. Identified putative biomarkers were validated in baseline serum samples of 140 subjects (validation set; control and GC occurrence group, 70 each) using targeted metabolites analysis. RESULTS The final analysis was conducted on the discovery set (control, n = 52 vs. GC occurrence, n = 50) and the validation set (control, n = 43 vs. GC occurrence, n = 44) applying exclusion conditions. Eighteen putative metabolite sets differed between two groups found on nontargeted metabolic screening. We focused on fatty acid-related energy metabolism. In targeted analysis, levels of decanoyl-L-carnitine (p = 0.019), L-carnitine (p = 0.033), and citric acid (p = 0.025) were significantly lower in the GC occurrence group, even after adjusting for age, sex, and smoking status. Additionally, L-carnitine and citric acid were confirmed to have an independently significant relationship to GC development. Notably, alkaline phosphatase showed a significant correlation with these two biomarkers. CONCLUSION Changes in serum L-carnitine and citric acid levels that may result from alterations of fatty-acid-related energy metabolism are expected to be valuable biomarkers for the early diagnosis of GC risk.
Collapse
Affiliation(s)
- Youngmin Han
- National Leading Research Laboratory of Clinical Nutrigenetics/Nutrigenomics, Department of Food and Nutrition, College of Human Ecology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- Institute for Health Promotion, Graduate School of Public Health, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Hye Jin Yoo
- Research Center for Silver Science, Institute of Symbiotic Life-TECH, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Sun Ha Jee
- Institute for Health Promotion, Graduate School of Public Health, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| | - Jong Ho Lee
- National Leading Research Laboratory of Clinical Nutrigenetics/Nutrigenomics, Department of Food and Nutrition, College of Human Ecology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
- Research Center for Silver Science, Institute of Symbiotic Life-TECH, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|
14
|
Guo W, Cao P, Wang X, Hu M, Feng Y. Medicinal Plants for the Treatment of Gastrointestinal Cancers From the Metabolomics Perspective. Front Pharmacol 2022; 13:909755. [PMID: 35833022 PMCID: PMC9271783 DOI: 10.3389/fphar.2022.909755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/23/2022] [Indexed: 12/27/2022] Open
Abstract
Gastrointestinal cancer (GIC), primarily including colorectal cancer, gastric cancer, liver cancer, pancreatic cancer, and esophageal cancer, is one of the most common causes of cancer-related deaths with increasing prevalence and poor prognosis. Medicinal plants have been shown to be a great resource for the treatment of GIC. Due to their complex manifestations of multi-component and multi-target, the underlying mechanisms how they function against GIC remain to be completely deciphered. Cell metabolism is of primary importance in the initialization and development of GIC, which is reported to be a potential target. As an essential supplement to the newest “omics” sciences, metabolomics focuses on the systematic study of the small exogenous and endogenous metabolites involved in extensive biochemical metabolic pathways of living system. In good agreement with the systemic perspective of medicinal plants, metabolomics offers a new insight into the efficacy assessment and action mechanism investigation of medicinal plants as adjuvant therapeutics for GIC therapy. In this review, the metabolomics investigations on metabolism-targeting therapies for GIC in the recent 10 years were systematically reviewed from five aspects of carbohydrate, lipid, amino acid, and nucleotide metabolisms, as well as other altered metabolisms (microbial metabolism, inflammation, and oxidation), with particular attention to the potential of active compounds, extracts, and formulae from medicinal plants. Meanwhile, the current perspectives and future challenges of metabolism-targeting therapies of medicinal plants for GIC were also discussed. In conclusion, the understanding of the action mechanisms of medicinal plants in GIC from the metabolomics perspective will contribute to the clinical application of potential candidates from the resourceful medicinal plants as novel and efficient adjuvant therapeutics for GIC therapy.
Collapse
Affiliation(s)
- Wei Guo
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, China
| | - Peng Cao
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, China
| | - Xuanbin Wang
- Laboratory of Chinese Herbal Pharmacology, Department of Pharmacy, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Min Hu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, China
- *Correspondence: Min Hu, ; Yibin Feng,
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- *Correspondence: Min Hu, ; Yibin Feng,
| |
Collapse
|
15
|
Prasad SK, Bhat S, Shashank D, C R A, R S, Rachtanapun P, Devegowda D, Santhekadur PK, Sommano SR. Bacteria-Mediated Oncogenesis and the Underlying Molecular Intricacies: What We Know So Far. Front Oncol 2022; 12:836004. [PMID: 35480118 PMCID: PMC9036991 DOI: 10.3389/fonc.2022.836004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/22/2022] [Indexed: 01/10/2023] Open
Abstract
Cancers are known to have multifactorial etiology. Certain bacteria and viruses are proven carcinogens. Lately, there has been in-depth research investigating carcinogenic capabilities of some bacteria. Reports indicate that chronic inflammation and harmful bacterial metabolites to be strong promoters of neoplasticity. Helicobacter pylori-induced gastric adenocarcinoma is the best illustration of the chronic inflammation paradigm of oncogenesis. Chronic inflammation, which produces excessive reactive oxygen species (ROS) is hypothesized to cause cancerous cell proliferation. Other possible bacteria-dependent mechanisms and virulence factors have also been suspected of playing a vital role in the bacteria-induced-cancer(s). Numerous attempts have been made to explore and establish the possible relationship between the two. With the growing concerns on anti-microbial resistance and over-dependence of mankind on antibiotics to treat bacterial infections, it must be deemed critical to understand and identify carcinogenic bacteria, to establish their role in causing cancer.
Collapse
Affiliation(s)
- Shashanka K Prasad
- Department of Biotechnology and Bioinformatics, Faculty of Life Sciences, Jagadguru Sri Shivarathreeshwara (JSS) Academy of Higher Education and Research (JSSAHER), Mysuru, India
| | - Smitha Bhat
- Department of Biotechnology and Bioinformatics, Faculty of Life Sciences, Jagadguru Sri Shivarathreeshwara (JSS) Academy of Higher Education and Research (JSSAHER), Mysuru, India
| | - Dharini Shashank
- Department of General Surgery, Adichunchanagiri Institute of Medical Sciences, Mandya, India
| | - Akshatha C R
- Department of Medical Oncology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| | - Sindhu R
- Department of Microbiology, Faculty of Life Sciences, Jagadguru Sri Shivarathreeshwara (JSS) Academy of Higher Education and Research (JSSAHER), Mysuru, India
| | - Pornchai Rachtanapun
- School of Agro-Industry, Faculty of Agro-Industry, Chiang Mai University, Chiang Mai, Thailand
- Cluster of Agro Bio-Circular-Green Industry (Agro BCG), Chiang Mai University, Chiang Mai, Thailand
| | - Devananda Devegowda
- Centre of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education and Research (JSSAHER), Mysuru, India
| | - Prasanna K Santhekadur
- Centre of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education and Research (JSSAHER), Mysuru, India
| | - Sarana Rose Sommano
- Cluster of Agro Bio-Circular-Green Industry (Agro BCG), Chiang Mai University, Chiang Mai, Thailand
- Department of Plant and Soil Sciences, Faculty of Agriculture, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
16
|
Modular Point-of-Care Breath Analyzer and Shape Taxonomy-Based Machine Learning for Gastric Cancer Detection. Diagnostics (Basel) 2022; 12:diagnostics12020491. [PMID: 35204584 PMCID: PMC8871298 DOI: 10.3390/diagnostics12020491] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/01/2022] [Accepted: 02/11/2022] [Indexed: 01/17/2023] Open
Abstract
Background: Gastric cancer is one of the deadliest malignant diseases, and the non-invasive screening and diagnostics options for it are limited. In this article, we present a multi-modular device for breath analysis coupled with a machine learning approach for the detection of cancer-specific breath from the shapes of sensor response curves (taxonomies of clusters). Methods: We analyzed the breaths of 54 gastric cancer patients and 85 control group participants. The analysis was carried out using a breath analyzer with gold nanoparticle and metal oxide sensors. The response of the sensors was analyzed on the basis of the curve shapes and other features commonly used for comparison. These features were then used to train machine learning models using Naïve Bayes classifiers, Support Vector Machines and Random Forests. Results: The accuracy of the trained models reached 77.8% (sensitivity: up to 66.54%; specificity: up to 92.39%). The use of the proposed shape-based features improved the accuracy in most cases, especially the overall accuracy and sensitivity. Conclusions: The results show that this point-of-care breath analyzer and data analysis approach constitute a promising combination for the detection of gastric cancer-specific breath. The cluster taxonomy-based sensor reaction curve representation improved the results, and could be used in other similar applications.
Collapse
|
17
|
Dai R, Liu M, Xiang X, Li Y, Xi Z, Xu H. OMICS Applications for Medicinal Plants in Gastrointestinal Cancers: Current Advancements and Future Perspectives. Front Pharmacol 2022; 13:842203. [PMID: 35185591 PMCID: PMC8855055 DOI: 10.3389/fphar.2022.842203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 01/20/2022] [Indexed: 12/24/2022] Open
Abstract
Gastrointestinal cancers refer to a group of deadly malignancies of the gastrointestinal tract and organs of the digestive system. Over the past decades, considerable amounts of medicinal plants have exhibited potent anticancer effects on different types of gastrointestinal cancers. OMICS, systems biology approaches covering genomics, transcriptomics, proteomics and metabolomics, are broadly applied to comprehensively reflect the molecular profiles in mechanistic studies of medicinal plants. Single- and multi-OMICS approaches facilitate the unravelling of signalling interaction networks and key molecular targets of medicinal plants with anti-gastrointestinal cancer potential. Hence, this review summarizes the applications of various OMICS and advanced bioinformatics approaches in examining therapeutic targets, signalling pathways, and the tumour microenvironment in response to anticancer medicinal plants. Advances and prospects in this field are also discussed.
Collapse
Affiliation(s)
- Rongchen Dai
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, China
| | - Mengfan Liu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, China
| | - Xincheng Xiang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, China
| | - Yang Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, China
| | - Zhichao Xi
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, China
- *Correspondence: Zhichao Xi, ; Hongxi Xu,
| | - Hongxi Xu
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Zhichao Xi, ; Hongxi Xu,
| |
Collapse
|
18
|
Wang R, Liu H, Song J, Wu Q. Activity of Melatonin Against Gastric Cancer Growth in a Chick Embryo Tumor Xenograft Model. Cancer Manag Res 2021; 13:8803-8808. [PMID: 34853535 PMCID: PMC8627858 DOI: 10.2147/cmar.s329728] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 10/05/2021] [Indexed: 12/14/2022] Open
Abstract
Purpose Previous studies have shown the antitumor activity of melatonin against a wide range of human cancers; however, the impact of melatonin on gastric cancer growth remains to be illustrated. This study aimed to investigate the activity of melatonin against gastric cancer growth in a chick embryo tumor xenograft model and explore the possible mechanisms. Materials and Methods The growth of gastric cancer SGC-7901 cells was measured using MTT assay, and a chick embryo tumor xenograft model was generated to observe the effect of melatonin on gastric cancer growth in vivo. In addition, the VEGF and angiogenin secretion was measured in the supernatant of chick embryo tumor xenograft models with ELISA. Results MLT treatment inhibited the growth of SGC-7901 cells at a concentration-dependent manner, and treatment with MLT at 1 mM was found to markedly reduce the volume and weight of tumors bearing the allantois of chicken embryos. ELISA showed that MLT at concentrations of 0.0041, 0.012, 0.037 and 0.11 had no remarkable impact on VEGF and angiopoietin secretion, while MLT at 1 mM significantly suppressed VEGF and angiopoietin production in chick embryo tumor xenograft models with SGC-7901 cells (P = 0.023). Conclusion Our data demonstrate that MLT inhibits gastric cancer growth in vitro at a concentration-dependent manner, and suppresses angiogenesis of the chick embryo tumor xenograft model with SGC-7901 cells through inhibiting VEGF and angiogenin secretion. Further studies are needed to investigate the therapeutic potential of MLT for gastric cancer as compared to drugs clinically approved.
Collapse
Affiliation(s)
- Rixiong Wang
- Department of Oncology, the First Affiliated Hospital of Fujian Medical University, Fuzhou City, People's Republic of China.,Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital of Fujian Medical University, Fuzhou City, People's Republic of China
| | - Hui Liu
- Department of Human Anatomy, Histology and Embryology, Fujian Medical University, Fuzhou City, People's Republic of China.,Key Laboratory of the Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou City, People's Republic of China
| | - Jun Song
- Department of Human Anatomy, Histology and Embryology, Fujian Medical University, Fuzhou City, People's Republic of China.,Key Laboratory of the Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou City, People's Republic of China
| | - Qing Wu
- Department of Oncology, the First Affiliated Hospital of Fujian Medical University, Fuzhou City, People's Republic of China.,Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital of Fujian Medical University, Fuzhou City, People's Republic of China
| |
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW Artificial intelligence is becoming rapidly integrated into modern technology including medicine. Artificial intelligence has a wide range of potential in gastroenterology, particularly with endoscopy, given the required analysis of large datasets of images. The aim of this review is to summarize the advances of artificial intelligence in gastroenterology (GI) endoscopy over the past year. RECENT FINDINGS Computer-aided detection (CADe) systems during real-time colonoscopy have resulted in increased adenoma detection rate with no significant increase in procedure times. Deep learning techniques have been utilized to accurately assess bowel preparation quality, which would impact surveillance colonoscopy recommendations. For the upper GI tract, CADe systems have been developed to aid in improving the diagnosis of Barrett's neoplasia during real-time endoscopy. Artificial intelligence-assisted real-time endoscopy has been shown to reduce blind spots during EGD. SUMMARY The application of artificial intelligence in gastroenterology endoscopy remains promising. Advances over the past year include improved detection of GI neoplasia during endoscopy and characterization of lesions. Further research including randomized, multicenter trials are needed to further evaluate the use of artificial intelligence for real-time endoscopy.
Collapse
|
20
|
El Halabi M, Horanieh R, Tamim H, Mukherji D, Jdiaa S, Temraz S, Shamseddine A, Barada K. The impact of age on prognosis in patients with gastric cancer: experience in a tertiary care centre. J Gastrointest Oncol 2021; 11:1233-1241. [PMID: 33456996 DOI: 10.21037/jgo-20-139] [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] [Indexed: 12/11/2022] Open
Abstract
Background Gastric cancer (GC) is a leading cause of cancer-related death in the world and most patients have advanced disease upon presentation. The effect of age on prognosis in GC is controversial. We aimed to determine the impact of age on survival in patients with GC. Methods This was a retrospective study of the medical records of Lebanese patients diagnosed with GC at the American University of Beirut Medical Center (AUBMC) between 2005 and 2014. Patients were divided into young (<65 years) and older groups (≥65 years). A multivariate analysis was done to determine the independent predictors of survival. Kaplan-Meier method was used for analysis of long-term survival outcomes. Results The sample consisted of 156 patients. The mean age was 62.15 (SD 13.54). Most patients presented with stage 4 disease (62.2%) and poorly differentiated histology (66.4%). The most common symptoms were abdominal pain and weight loss. On bivariate analysis, advanced stage (P=0.02) and higher grade (P=0.04) were associated with increased mortality. Patients <65 years of age were significantly more likely to have poorly differentiated tumours, while patients ≥65 years had more comorbidities (P=0.001). The 5-year DFS were 35% and 37% for patients <65 years of age and ≥65 years of age, respectively (P=0.15). Conclusions Higher grade and advanced stage are associated with worse survival in patients with GC, but age did not seem to have an impact. Screening high risk patients and early diagnosis are necessary to improve survival.
Collapse
Affiliation(s)
- Maan El Halabi
- Department of Internal Medicine, American University of Beirut Medical Centre, Beirut, Lebanon.,Division of Gastroenterology, American University of Beirut Medical Centre, Beirut, Lebanon
| | - Renee Horanieh
- Department of Internal Medicine, American University of Beirut Medical Centre, Beirut, Lebanon
| | - Hani Tamim
- Clinical Research Institute, American University of Beirut Medical Centre, Beirut, Lebanon
| | - Deborah Mukherji
- Department of Internal Medicine, American University of Beirut Medical Centre, Beirut, Lebanon.,Division of Hematology and Oncology, American University of Beirut Medical Centre, Beirut, Lebanon
| | - Sara Jdiaa
- Department of Internal Medicine, American University of Beirut Medical Centre, Beirut, Lebanon
| | - Sally Temraz
- Department of Internal Medicine, American University of Beirut Medical Centre, Beirut, Lebanon.,Division of Hematology and Oncology, American University of Beirut Medical Centre, Beirut, Lebanon
| | - Ali Shamseddine
- Department of Internal Medicine, American University of Beirut Medical Centre, Beirut, Lebanon.,Division of Hematology and Oncology, American University of Beirut Medical Centre, Beirut, Lebanon
| | - Kassem Barada
- Department of Internal Medicine, American University of Beirut Medical Centre, Beirut, Lebanon.,Division of Gastroenterology, American University of Beirut Medical Centre, Beirut, Lebanon
| |
Collapse
|
21
|
Wen F, Huang J, Lu X, Huang W, Wang Y, Bai Y, Ruan S, Gu S, Chen X, Shu P. Identification and prognostic value of metabolism-related genes in gastric cancer. Aging (Albany NY) 2020; 12:17647-17661. [PMID: 32920549 PMCID: PMC7521523 DOI: 10.18632/aging.103838] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 07/14/2020] [Indexed: 02/07/2023]
Abstract
Gastric cancer (GC) is one of the most commonly occurring cancers, and metabolism-related genes (MRGs) are associated with its development. Transcriptome data and the relevant clinical data were downloaded from The Cancer Genome Atlas and Gene Expression Omnibus databases, and we identified 194 MRGs differentially expressed between GC and adjacent nontumor tissues. Through univariate Cox and lasso regression analyses we identified 13 potential prognostic differentially expressed MRGs (PDEMRGs). These PDEMRGs (CKMT2, ME1, GSTA2, ASAH1, GGT5, RDH12, NNMT, POLR1A, ACYP1, GLA, OPLAH, DCK, and POLD3) were used to build a Cox regression risk model to predict the prognosis of GC patients. Further univariate and multivariate Cox regression analyses showed that this model could serve as an independent prognostic parameter. Gene Set Enrichment Analysis showed significant enrichment pathways that could potentially contribute to pathogenesis. This model also revealed the probability of genetic alterations of PDEMRGs. We have thus identified a valuable metabolic model for predicting the prognosis of GC patients. The PDEMRGs in this model reflect the dysregulated metabolic microenvironment of GC and provide useful noninvasive biomarkers.
Collapse
Affiliation(s)
- Fang Wen
- Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China,Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, Jiangsu Province, China,Department of Oncology, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| | - Jiani Huang
- Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China,College of Traditional Chinese Medicine, College of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiaona Lu
- Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China,Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, Jiangsu Province, China,Department of Oncology, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| | - Wenjie Huang
- Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China,Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, Jiangsu Province, China,Department of Oncology, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| | - Yulan Wang
- Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China,Department of Hematology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| | - Yingfeng Bai
- Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China,College of Traditional Chinese Medicine, College of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Shuai Ruan
- Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China,Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, Jiangsu Province, China,Department of Oncology, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| | - Suping Gu
- Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China,Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, Jiangsu Province, China,Department of Oncology, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| | - Xiaoxue Chen
- Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China,Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, Jiangsu Province, China,Department of Oncology, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| | - Peng Shu
- Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China,Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, Jiangsu Province, China,Department of Oncology, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| |
Collapse
|
22
|
Aznab M, Ahmadi SM, Khazaei S, Ahmadi SM, Khazaei M, Hamdi H. Relationship between the Expression of the Thymidylate Synthase and the Prognosis of Gastric Cancer Patients Treated with Combinational Chemotherapy Regimen Including Fluorouracil, Docetaxel and Cisplatin. Int J Hematol Oncol Stem Cell Res 2020; 14:181-187. [PMID: 33024525 PMCID: PMC7521394 DOI: 10.18502/ijhoscr.v14i3.3727] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Background: Thymidylate synthase is one of the target enzymes of 5-fluorouracil. However, the clinical and prognostic significance of TS expression in gastric cancer has remained controversial. In this study, the expression of thymidylate synthase was evaluated in gastric cancer patients treated with combinational chemotherapy; moreover, the association between TS expression and clinicopathologic characteristics and overall survival of the patients were also assessed. Materials and Methods: In this descriptive study, 89 pathological samples were gathered from patients at Kermanshah hospitals during 2008-2017. The survival status of patients was recorded and their overall survival was evaluated individually. Results: The average survival period for low and high thymidylate synthase groups was 54 and 50 months, respectively, meaning higher survival time in the lower thymidylate group. But this difference was not statistically significant (log Rank=0.88). In addition, sex, stage, recurrence, and survival had no significant difference between the low and high expression of thymidylate synthase groups (p=0.89). Conclusion: The results clearly indicated that the level of thymidylate synthase is not a significant modulator of 5- fluorouracil in gastric cancer patients. Nevertheless, evaluation of the level of the enzymes and markers as well as their effects are highly recommended for accurate selection of chemotherapeutical strategies.
Collapse
Affiliation(s)
- Mozaffar Aznab
- Department of Internal Medicine, Taleghani Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | | - Sedigheh Khazaei
- Molecular Pathology Research Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Seyed Mojtaba Ahmadi
- Department of Clinical Psychology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mansour Khazaei
- Taleghani Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | |
Collapse
|
23
|
In H, Solsky I, Castle PE, Schechter CB, Parides M, Friedmann P, Wylie-Rosett J, Kemeny MM, Rapkin BD. Utilizing Cultural and Ethnic Variables in Screening Models to Identify Individuals at High Risk for Gastric Cancer: A Pilot Study. Cancer Prev Res (Phila) 2020; 13:687-698. [PMID: 32409594 DOI: 10.1158/1940-6207.capr-19-0490] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 02/26/2020] [Accepted: 04/23/2020] [Indexed: 11/16/2022]
Abstract
Identifying persons at high risk for gastric cancer is needed for targeted interventions for prevention and control in low-incidence regions. Combining ethnic/cultural factors with conventional gastric cancer risk factors may enhance identification of high-risk persons. Data from a prior case-control study (40 gastric cancer cases and 100 controls) were used. A "conventional model" using risk factors included in the Harvard Cancer Risk Index's gastric cancer module was compared with a "parsimonious model" created from the most predictive variables of the conventional model as well as ethnic/cultural and socioeconomic variables. Model probability cutoffs aimed to identify a cohort with at least 10 times the baseline risk using Bayes' Theorem applied to baseline U.S. gastric cancer incidence. The parsimonious model included age, U.S. generation, race, cultural food at ages 15-18 years, excessive salt, education, alcohol, and family history. This 11-item model enriched the baseline risk by 10-fold, at the 0.5 probability level cutoff, with an estimated sensitivity of 72% [95% confidence interval (CI), 64-80], specificity of 94% (95% CI, 90-97), and ability to identify a subcohort with gastric cancer prevalence of 128.5 per 100,000. The conventional model was only able to reach a risk level of 9.8 times baseline with a corresponding sensitivity of 31% (95% CI, 23-39) and specificity of 97% (95% CI, 94-99). Cultural and ethnic data may add important information to models for identifying U.S. individuals at high risk for gastric cancer, who then could be targeted for interventions to prevent and control gastric cancer. The findings of this pilot study remain to be validated in an external dataset.
Collapse
Affiliation(s)
- Haejin In
- Department of Surgery, Montefiore Medical Center, New York, New York. .,Department of Surgery, Albert Einstein College of Medicine, New York, New York.,Department of Epidemiology & Population Health, Albert Einstein College of Medicine, New York, New York
| | - Ian Solsky
- Department of Surgery, Montefiore Medical Center, New York, New York
| | - Philip E Castle
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, New York, New York
| | - Clyde B Schechter
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, New York, New York.,Department of Family and Social Medicine, Albert Einstein College of Medicine, New York, New York
| | - Michael Parides
- Department of Surgery, Montefiore Medical Center, New York, New York
| | | | - Judith Wylie-Rosett
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, New York, New York
| | | | - Bruce D Rapkin
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, New York, New York
| |
Collapse
|
24
|
Zeng W, Zhang S, Yang L, Wei W, Gao J, Guo N, Wu F. Serum miR-101-3p combined with pepsinogen contributes to the early diagnosis of gastric cancer. BMC MEDICAL GENETICS 2020; 21:28. [PMID: 32041551 PMCID: PMC7011268 DOI: 10.1186/s12881-020-0967-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 02/03/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND This study aimed to explore the diagnostic value of serum miR-101-3p combined with pepsinogen (PG) on early diagnosis of gastric cancer (GC). METHODS A total of 61 atrophic gastritis (AG) and 86 GC patients, and 50 healthy volunteers were enrolled. The serum expression of miR-101-3p was measured by qRT-PCR. The serum content of carcinoembryonic antigen (CEA) was measured by Electrochemiluminescence immunoassay. The serum contents of PGI and PGII were measured by Enzyme linked immunosorbent assay. The diagnostic value of serum markers on AG and GC was analyzed by receiver operating characteristic (ROC) analysis. RESULTS The expression of miR-101-3p, the content of PGI and the ratio of PGI/II were significantly decreased, and the content of PGII was significantly increased in AG patients compared with those in normal controls. The changes of the above serum indicators were more obvious in GC patients than those in AG patients. The content of CEA was significantly higher in GC patients than that in AG patients. In addition, the expression of miR-101-3p was negatively associated with the submucosal infiltration in GC patients. MiR-101-3p exhibited high diagnostic value on AG (AUC 0.8493, sensitivity 80.33%, specificity 80%) and GC (AUC 0.8749, sensitivity 72.09%, specificity 86.49%). MiR-101-3p + PGI + PGI/II (AUC 0.856, sensitivity 80.23%, specificity 77.05%) exhibited a high diagnostic value in distinguishing between AG and GC. CONCLUSIONS MiR-101-3p was a potential diagnostic marker for AG and GC. MiR-101-3p + PGI + PGI/II was effective in distinguishing between AG and GC.
Collapse
Affiliation(s)
- Weiwei Zeng
- Department of Gastroenterology, Dongying People's Hospital, No. 317, Chengnan First Road, Dongying City, 257091, Shandong Province, China.
| | - Shuxiang Zhang
- Department of Gastroenterology, Dongying People's Hospital, No. 317, Chengnan First Road, Dongying City, 257091, Shandong Province, China
| | - Lei Yang
- Department of Gastroenterology, Dongying People's Hospital, No. 317, Chengnan First Road, Dongying City, 257091, Shandong Province, China
| | - Wenchao Wei
- Department of Gastroenterology, Dongying People's Hospital, No. 317, Chengnan First Road, Dongying City, 257091, Shandong Province, China
| | - Jie Gao
- Department of Gastroenterology, Dongying People's Hospital, No. 317, Chengnan First Road, Dongying City, 257091, Shandong Province, China
| | - Ni Guo
- Department of Gastroenterology, Dongying People's Hospital, No. 317, Chengnan First Road, Dongying City, 257091, Shandong Province, China
| | - Fengting Wu
- Department of Gastroenterology, Dongying People's Hospital, No. 317, Chengnan First Road, Dongying City, 257091, Shandong Province, China
| |
Collapse
|
25
|
Zhenming Y, Lei S. Diagnostic value of blue laser imaging combined with magnifying endoscopy for precancerous and early gastric cancer lesions. TURKISH JOURNAL OF GASTROENTEROLOGY 2020; 30:549-556. [PMID: 31144661 DOI: 10.5152/tjg.2019.18210] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND/AIMS Blue laser imaging (BLI) is a new technique for detailed examination of upper gastrointestinal lesions. This study aimed to evaluate the diagnostic value of BLI combined with magnifying endoscopy for precancerous and early gastric cancer lesions. MATERIALS AND METHODS A total of 249 gastric lesions detected via conventional white light endoscopy (WLE) based on assessments of mucosal shape and color were included in this study. The accuracy of diagnosis of precancerous or early cancer lesions white light magnification alone, BLI-contrast magnification, and BLI-bright magnification was determined according to the VS criteria. RESULTS For white light magnification alone, BLI-contrast magnification, and BLI-bright magnification, the concordance rates for lesions were 76.7%, 85.1%, and 86.7%, respectively, and the Kappa values were 0.571, 0.730, and 0.760, respectively. For the screening of high-grade intraepithelial neoplasia or early gastric cancer, the diagnostic sensitivities of white light magnification alone, BLI-contrast magnification, and BLI-bright magnification were 72.0%, 92.0%, and 92.0%, respectively; the specificities were 95.5%, 98.2%, and 99.1%, respectively; the consistencies were 93.2%, 97.6%, and 98.4%, respectively; and the Kappa values were 0.642, 0.871, and 0.911, respectively. For diagnoses of high-grade intraepithelial neoplasia or early gastric cancer, the concordance between endoscopic and pathological diagnosis was significantly higher for BLI-contrast and BLI-bright magnification than for white light magnification alone (p<0.05). CONCLUSION BLI combined with magnifying endoscopy may improve diagnostic accuracy for early gastric cancer and precancerous lesions.
Collapse
Affiliation(s)
- Yang Zhenming
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shen Lei
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
26
|
Su X, Zhang J, Luo X, Yang W, Liu Y, Liu Y, Shan Z. LncRNA LINC01116 Promotes Cancer Cell Proliferation, Migration And Invasion In Gastric Cancer By Positively Interacting With lncRNA CASC11. Onco Targets Ther 2019; 12:8117-8123. [PMID: 31632064 PMCID: PMC6781852 DOI: 10.2147/ott.s208133] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Accepted: 07/18/2019] [Indexed: 01/14/2023] Open
Abstract
Purpose The oncogenic roles of lncRNA LINC01116 have been reported in several types of cancer, while its involvement in gastric cancer is unknown. This study aimed to investigate the involvement of LINC01116 in gastric cancer. Methods Gene expression was detected by qPCR. Correlations were analyzed by linear regression. Overexpression and siRNA silencing techniques were used to analyze gene functions. Cell invasion and migration were analyzed by Transwell assays. Results LINC01116 and lncRNA CASC11 were both upregulated in cancer tissues compared to cancer-adjacent tissues. Expression levels of LINC01116 and CASC11 were increased with the increase in clinical stages. Expression levels of LINC01116 and CASC11 were positively correlated. Overexpression of LINC01116 mediated the upregulated CASC11 in gastric cancer cells, and CASC11 overexpression also led to overexpressed LINC01116. Overexpression of LINC01116 and CASC11 led to promoted invasion and migration of gastric cancer cells. Rescue experiments showed that CASC11 knockdown attenuated the effects of LINC01116 overexpression. Overexpression of LINC01116 failed to significantly affect cancer cell proliferation. Conclusion LINC01116 promoted cancer cell invasion and migration in gastric cancer by positively interacting with CASC11.
Collapse
Affiliation(s)
- Xiaohui Su
- Department of Gastric Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang City, Liaoning Province 110042, People's Republic of China
| | - Jianjun Zhang
- Department of Gastric Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang City, Liaoning Province 110042, People's Republic of China
| | - Xianfeng Luo
- Department of Gastric Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang City, Liaoning Province 110042, People's Republic of China
| | - Wei Yang
- Department of Gastric Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang City, Liaoning Province 110042, People's Republic of China
| | - Yanqing Liu
- Department of Gastric Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang City, Liaoning Province 110042, People's Republic of China
| | - Yang Liu
- Department of Gastric Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang City, Liaoning Province 110042, People's Republic of China
| | - Zexing Shan
- Department of Gastric Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang City, Liaoning Province 110042, People's Republic of China
| |
Collapse
|
27
|
Yan H, Li M, Cao L, Chen H, Lai H, Guan Q, Chen H, Zhou W, Zheng B, Guo Z, Zheng C. A robust qualitative transcriptional signature for the correct pathological diagnosis of gastric cancer. J Transl Med 2019; 17:63. [PMID: 30819200 PMCID: PMC6394047 DOI: 10.1186/s12967-019-1816-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 02/21/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Currently, pathological examination of gastroscopy biopsy specimens is the gold standard for gastric cancer (GC) diagnosis. However, it has a false-negative rate of 10-20% due to inaccurate sampling locations and/or insufficient sampling amount. A signature should be developed to aid the early diagnosis of GC using biopsy specimens even when they are sampled from inaccurate locations. METHODS We extracted a robust qualitative transcriptional signature, based on the within-sample relative expression orderings (REOs) of gene pairs, to discriminate both GC tissues and adjacent-normal tissues from non-GC gastritis, intestinal metaplasia and normal gastric tissues. RESULTS A signature consisting of two gene pairs for GC diagnosis was identified and validated in data of both biopsy specimens and surgical resection specimens pooled from publicly available datasets measured by different laboratories with different platforms. For gastroscopy biopsy specimens, 96.20% of 79 non-GC tissues were correctly identified as non-GC, and 96.84% of 158 GC tissues and six of seven adjacent-normal tissues were correctly identified as GC. For surgical resection specimens, 98.37% of 2560 GC tissues and 97.28% of 221 adjacent-normal tissues were correctly identified as GC. Especially, 97.67% of the 257 GC patients at stage I were exactly diagnosed as GC. We additionally measured 21 GC tissues from seven different GC patients, each with three specimens sampled from three tumor locations with different proportions of the tumor epithelial cell. All these GC tissues were correctly identified as GC, even when the proportion of the tumor epithelial cell was as low as 14%. CONCLUSIONS The qualitative transcriptional signature can distinguish both GC and adjacent-normal tissues from normal, gastritis and intestinal metaplasia tissues of non-GC patients even using inaccurately sampled biopsy specimens, which can be applied robustly at the individual level to aid the early GC diagnosis.
Collapse
Affiliation(s)
- Haidan Yan
- Department of Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China
| | - Meifeng Li
- Department of Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China
| | - Longlong Cao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, China
| | - Haifeng Chen
- Department of General Surgery, Fuzhou Second Hospital Affiliated To Xiamen University, Xiamen, 350007, China
| | - Hungming Lai
- Department of Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China
| | - Qingzhou Guan
- Department of Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China
| | - Huxing Chen
- Department of Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China
| | - Wenbin Zhou
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150086, China
| | - Baotong Zheng
- Department of Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China
| | - Zheng Guo
- Department of Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China. .,Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150086, China.
| | - Chaohui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, China.
| |
Collapse
|
28
|
Pirini F, Noazin S, Jahuira-Arias MH, Rodriguez-Torres S, Friess L, Michailidi C, Cok J, Combe J, Vargas G, Prado W, Soudry E, Pérez J, Yudin T, Mancinelli A, Unger H, Ili-Gangas C, Brebi-Mieville P, Berg DE, Hayashi M, Sidransky D, Gilman RH, Guerrero-Preston R. Early detection of gastric cancer using global, genome-wide and IRF4, ELMO1, CLIP4 and MSC DNA methylation in endoscopic biopsies. Oncotarget 2018; 8:38501-38516. [PMID: 28418867 PMCID: PMC5503549 DOI: 10.18632/oncotarget.16258] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 02/24/2017] [Indexed: 12/15/2022] Open
Abstract
Clinically useful molecular tools to triage gastric cancer patients are not currently available. We aimed to develop a molecular tool to predict gastric cancer risk in endoscopy-driven biopsies obtained from high-risk gastric cancer clinics in low resource settings. We discovered and validated a DNA methylation biomarker panel in endoscopic samples obtained from 362 patients seen between 2004 and 2009 in three high-risk gastric cancer clinics in Lima, Perú, and validated it in 306 samples from the Cancer Genome Atlas project (“TCGA”). Global, epigenome wide and gene-specific DNA methylation analyses were used in a Phase I Biomarker Development Trial to identify a continuous biomarker panel that combines a Global DNA Methylation Index (GDMI) and promoter DNA methylation levels of IRF4, ELMO1, CLIP4 and MSC. We observed an inverse association between the GDMI and histological progression to gastric cancer, when comparing gastritis patients without metaplasia (mean = 5.74, 95% CI, 4.97−6.50), gastritis patients with metaplasia (mean = 4.81, 95% CI, 3.77−5.84), and gastric cancer cases (mean = 3.38, 95% CI, 2.82−3.94), respectively (p < 0.0001). Promoter methylation of IRF4 (p < 0.0001), ELMO1 (p < 0.0001), CLIP4 (p < 0.0001), and MSC (p < 0.0001), is also associated with increasing severity from gastritis with no metaplasia to gastritis with metaplasia and gastric cancer. Our findings suggest that IRF4, ELMO1, CLIP4 and MSC promoter methylation coupled with a GDMI>4 are useful molecular tools for gastric cancer risk stratification in endoscopic biopsies.
Collapse
Affiliation(s)
- Francesca Pirini
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Sassan Noazin
- The Johns Hopkins University, Bloomberg School of Public Health, Department of International Health, Baltimore, MD, USA
| | - Martha H Jahuira-Arias
- The Johns Hopkins University, School of Medicine, Otolaryngology Department, Head and Neck Cancer Research Division, Baltimore, MD, USA.,Universidad Peruana Cayetano Heredia, Lima, Perú
| | - Sebastian Rodriguez-Torres
- The Johns Hopkins University, School of Medicine, Otolaryngology Department, Head and Neck Cancer Research Division, Baltimore, MD, USA
| | - Leah Friess
- The Johns Hopkins University, School of Medicine, Otolaryngology Department, Head and Neck Cancer Research Division, Baltimore, MD, USA
| | - Christina Michailidi
- The Johns Hopkins University, School of Medicine, Otolaryngology Department, Head and Neck Cancer Research Division, Baltimore, MD, USA
| | - Jaime Cok
- Hospital Nacional Cayetano Heredia, Pathology Department, Lima, Perú
| | - Juan Combe
- Instituto Nacional de Enfermedades Neoplásicas, Gastroenterology Department, Lima, Perú
| | - Gloria Vargas
- Hospital Nacional Arzobispo Loayza, Gastroenterology Department, Lima, Perú
| | - William Prado
- Hospital Nacional Dos de Mayo, Gastroenterology Department, Lima, Perú
| | - Ethan Soudry
- The Johns Hopkins University, School of Medicine, Otolaryngology Department, Head and Neck Cancer Research Division, Baltimore, MD, USA
| | - Jimena Pérez
- The Johns Hopkins University, School of Medicine, Otolaryngology Department, Head and Neck Cancer Research Division, Baltimore, MD, USA
| | - Tikki Yudin
- The Johns Hopkins University, School of Medicine, Otolaryngology Department, Head and Neck Cancer Research Division, Baltimore, MD, USA
| | - Andrea Mancinelli
- The Johns Hopkins University, School of Medicine, Otolaryngology Department, Head and Neck Cancer Research Division, Baltimore, MD, USA
| | - Helen Unger
- The Johns Hopkins University, School of Medicine, Otolaryngology Department, Head and Neck Cancer Research Division, Baltimore, MD, USA
| | - Carmen Ili-Gangas
- Laboratory of Molecular Pathology, Department of Pathological Anatomy, School of Medicine, Universidad de La Frontera, Temuco, Chile.,Center of Excellence in Translational Medicine - Scientific and Technological Bioresource Nucleus (CEMT-BIOREN), Universidad de La Frontera, Temuco, Chile
| | - Priscilla Brebi-Mieville
- Laboratory of Molecular Pathology, Department of Pathological Anatomy, School of Medicine, Universidad de La Frontera, Temuco, Chile.,Center of Excellence in Translational Medicine - Scientific and Technological Bioresource Nucleus (CEMT-BIOREN), Universidad de La Frontera, Temuco, Chile
| | - Douglas E Berg
- Washington University Medical School, Department of Molecular Microbiology, St Louis, MO, USA.,University of California San Diego, Department of Medicine, La Jolla, CA, USA
| | - Masamichi Hayashi
- The Johns Hopkins University, School of Medicine, Otolaryngology Department, Head and Neck Cancer Research Division, Baltimore, MD, USA.,Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - David Sidransky
- The Johns Hopkins University, School of Medicine, Otolaryngology Department, Head and Neck Cancer Research Division, Baltimore, MD, USA
| | - Robert H Gilman
- The Johns Hopkins University, Bloomberg School of Public Health, Department of International Health, Baltimore, MD, USA.,Universidad Peruana Cayetano Heredia, Lima, Perú
| | - Rafael Guerrero-Preston
- The Johns Hopkins University, School of Medicine, Otolaryngology Department, Head and Neck Cancer Research Division, Baltimore, MD, USA.,University of Puerto Rico School of Medicine, Department of Obstetrics and Gynecology, San Juan, Puerto Rico
| |
Collapse
|
29
|
Sun Q, Lu NN, Feng L. Apigetrin inhibits gastric cancer progression through inducing apoptosis and regulating ROS-modulated STAT3/JAK2 pathway. Biochem Biophys Res Commun 2018; 498:164-170. [PMID: 29408335 DOI: 10.1016/j.bbrc.2018.02.009] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 02/02/2018] [Indexed: 12/21/2022]
Abstract
Apigetrin (APG), as a flavonoid, has many cellular bioactivities, including regulation of oxidative stress, and induction of apoptosis. However, the means by which APG suppresses human gastric cancer are still little to be understood. In the present study, the anti-cancer effects of APG on human gastric cancer cells were investigated. The results indicated that APG could suppress the proliferation and induce apoptosis in gastric cancer cells. Its role in apoptosis induction was through reducing Bcl-2, and enhancing Bax, Caspase-9/-3 and poly ADP-ribose polymerase (PARP) cleavage. In addition, APG incubation resulted in the generation of intracellular reactive oxygen species (ROS) in cells. Meanwhile, APG suppressed constitutive and interleukin-6 (IL-6)-stimulated signal transducer and activator of transcription 3 (STAT3), Janus kinase 2 gene (JAK2) and Src activation. However, ROS scavenger, N-acety-l-cysteine (NAC), diminished apoptosis induced by APG. And APG-triggered de-phosphorylation of STAT3/JAK2 was rescued by NAC pre-treatment. In vivo, APG administration significantly inhibited the gastric cancer cell xenograft tumorigenesis through inducing apoptosis and inhibiting STAT3/JAK2 pathways. Taken together, the findings above illustrated that APG might be used as a promising candidate against human gastric cancer progression.
Collapse
Affiliation(s)
- Qian Sun
- Pharmacy Intravenous Admixture Service Centre, Affiliated Hospital of Jining Medical University, Jining 272029, China
| | - Na-Na Lu
- Department of Neonatology, Jining No.1 People's Hospital, Jining 272029, China
| | - Lei Feng
- Department of Pharmacy, Affiliated Hospital of Jining Medical University, Jining 272029, China.
| |
Collapse
|
30
|
Plasma sample based analysis of gastric cancer progression using targeted metabolomics. Sci Rep 2017; 7:17774. [PMID: 29259332 PMCID: PMC5736578 DOI: 10.1038/s41598-017-17921-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 12/02/2017] [Indexed: 12/16/2022] Open
Abstract
Gastric carcinogenesis is a multifactorial process described as a stepwise progression from non-active gastritis (NAG), chronic active gastritis (CAG), precursor lesions of gastric cancer (PLGC) and gastric adenocarcinoma. Gastric cancer (GC) 5-year survival rate is highly dependent upon stage of disease at diagnosis, which is based on endoscopy, biopsy and pathological examinations. Non-invasive GC biomarkers would facilitate its diagnosis at early stages leading to improved GC prognosis. We analyzed plasma samples collected from 80 patients diagnosed with NAG without H. pylori infection (NAG−), CAG with H. pylori infection (CAG+), PLGC and GC. A panel of 208 metabolites including acylcarnitines, amino acids and biogenic amines, sphingolipids, glycerophospholipids, hexoses, and tryptophan and phenylalanine metabolites were quantified using two complementary quantitative approaches: Biocrates AbsoluteIDQ®p180 kit and a LC-MS method designed for the analysis of 29 tryptophan pathway and phenylalanine metabolites. Significantly altered metabolic profiles were found in GC patients that allowing discrimination from NAG−, CAG+ and PLGC patients. Pathway analysis showed significantly altered tryptophan and nitrogen metabolic pathways (FDR P < 0.01). Three metabolites (histidine, tryprophan and phenylacetylglutamine) discriminated between non-GC and GC groups. These metabolic signatures open new possibilities to improve surveillance of PLGC patients using a minimally invasive blood analysis.
Collapse
|
31
|
Digital Image Analysis of HER2 Immunostained Gastric and Gastroesophageal Junction Adenocarcinomas. Appl Immunohistochem Mol Morphol 2017; 25:320-328. [PMID: 27801737 PMCID: PMC5447782 DOI: 10.1097/pai.0000000000000463] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Manual assessment of human epidermal growth factor receptor 2 (HER2) protein expression by immunohistochemistry (IHC) in gastric and gastroesophageal junction (GGEJ) adenocarcinomas is prone to interobserver variability and hampered by tumor heterogeneity and different scoring criteria. Equivocal cases are frequent, requiring additional in situ hybridization analysis. This study aimed to evaluate the accuracy of digital image analysis for the assessment of HER2 protein expression. In total, 110 GGEJ adenocarcinomas were included in tissue microarrays with 3 tissue cores per case. Two immunoassays, PATHWAY and HercepTest, and fluorescent in situ hybridization analysis were performed. The Visiopharm HER2-CONNECT Analysis Protocol Package was applied through the ONCOtopix digital image analysis software platform. HER2 membrane connectivity, calculated by the Analysis Protocol Package, was converted to standard IHC scores applying predetermined cutoff values for breast carcinoma as well as novel cutoff values. Cases with excessive cytoplasmic staining as well as HER2 amplified IHC negative cases were excluded from analysis. Applying HER2-CONNECT with connectivity cutoff values established for breast carcinoma resulted in 72.7% sensitivity and 100% specificity for the identification of HER2 positive gene amplified cases. By application of new cutoff values, the sensitivity increased to 100% without decreased specificity. With the new cutoff values, a 36% to 50% reduction of IHC equivocal cases was obtained. In conclusion, HER2-CONNECT with adjusted cutoff values seem to be an effective tool for standardized assessment of HER2 protein expression in GGEJ adenocarcinomas, decreasing the need for in situ hybridization analyzes.
Collapse
|
32
|
Pan Z, Tian Y, Zhang B, Zhang X, Shi H, Liang Z, Wu P, Li R, You B, Yang L, Mao F, Qian H, Xu W. YAP signaling in gastric cancer-derived mesenchymal stem cells is critical for its promoting role in cancer progression. Int J Oncol 2017; 51:1055-1066. [PMID: 28848999 PMCID: PMC5592864 DOI: 10.3892/ijo.2017.4101] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 07/24/2017] [Indexed: 12/20/2022] Open
Abstract
Cancer-associated mesenchymal stem cells (MSCs) are critically involved in tumor development and progression. However, the mechanisms of action for MSCs in cancer remain largely unknown. Herein, we reported that the expression of Yes-associated protein 1 (YAP) was higher in gastric cancer derived mesenchymal stem cells (GC-MSCs) than that in bone marrow derived MSCs (BM-MSCs). YAP knockdown not only inhibited the growth, migration and invasion, and stemness of GC-MSCs, but also suppressed their promoting effect on gastric cancer growth in vitro and in vivo. In addition, the interference of YAP expression in GC-MSCs also attenuated the promoting role of gastric cancer cells in endothelial cell tube formation and migration. Mechanistically, YAP knockdown reduced the activation of β-catenin and its target genes in gastric cancer cells by GC-MSCs. Taken together, these findings suggest that YAP activation in GC-MSCs plays an important role in promoting gastric cancer progression, which may represent a potential target for gastric cancer therapy.
Collapse
Affiliation(s)
- Zhaoji Pan
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Yiqing Tian
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Bin Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Xu Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Hui Shi
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Zhaofeng Liang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Peipei Wu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Rong Li
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Benshuai You
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Lunyu Yang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Fei Mao
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Hui Qian
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Wenrong Xu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| |
Collapse
|
33
|
Wu XD, Bie QL, Zhang B, Yan ZH, Han ZJ. Wnt10B is critical for the progression of gastric cancer. Oncol Lett 2017; 13:4231-4237. [PMID: 28599424 PMCID: PMC5452953 DOI: 10.3892/ol.2017.5992] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 02/01/2017] [Indexed: 01/26/2023] Open
Abstract
The family of Wnt proteins have been implicated in embryogenesis by regulation of cell fate and pattern formation, and also in human carcinogenesis. Wnt10B was previously shown to be involved in breast cancer development. The present study assessed the association of Wnt10B expression in human gastric cancer tissue specimens with clinicopathological data from these patients. Wnt10B expression in the regulation of gastric cancer cell proliferation and migration capacity in vitro was then investigated. The data revealed that Wnt10B mRNA and protein were upregulated in gastric cancer tissue samples and the upregulated Wnt10B mRNA was associated with gastric cancer metastasizing to lymph nodes. Knockdown of Wnt10B expression reduced gastric cancer cell proliferation and migration, as well as expression of a cell proliferation marker Ki67. Knockdown of Wnt10B expression inhibited tumor cell epithelial-mesenchymal transition by upregulation of E-cadherin and downregulation of N-cadherin. In addition, Wnt10B knockdown also suppressed tumor cell stemness by downregulation of octamer-binding transcription factor 4 and Nanog expression. The present data indicated that Wnt10B expression performs an important role in gastric cancer progression in vitro. Therefore, targeting of Wnt10B expression or activity may be investigated as a possible strategy for the control of gastric cancer.
Collapse
Affiliation(s)
- Xiao-Dan Wu
- Department of Laboratory Medicine, Nanjing Medical University Affiliated Wuxi Second Hospital, Wuxi, Jiangsu 214002, P.R. China
| | - Qing-Li Bie
- The Key Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Bin Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining, Shandong 272000, P.R. China
| | - Zi-He Yan
- Department of Laboratory Medicine, Nanjing Medical University Affiliated Wuxi Second Hospital, Wuxi, Jiangsu 214002, P.R. China
| | - Zhi-Jun Han
- Department of Laboratory Medicine, Nanjing Medical University Affiliated Wuxi Second Hospital, Wuxi, Jiangsu 214002, P.R. China
| |
Collapse
|
34
|
Mohammadian F, Abhari A, Dariushnejad H, Nikanfar A, Pilehvar-Soltanahmadi Y, Zarghami N. Effects of Chrysin-PLGA-PEG Nanoparticles on Proliferation and Gene Expression of miRNAs in Gastric Cancer Cell Line. IRANIAN JOURNAL OF CANCER PREVENTION 2016; 9:e4190. [PMID: 27761206 PMCID: PMC5056017 DOI: 10.17795/ijcp-4190] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 10/26/2015] [Accepted: 08/10/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND Recently, Chrysin, as a flavone, has revealed cancer chemo-preventive activity. The present experiment utilized the PLGA-PEG-chrysin complex, and free chrysin, to evaluation of the expression of miR-22, miR-34a and miR-126 in human gastric cell line. OBJECTIVES The purpose of this study was to examine whether nano encapsulating chrysin improves the anti-cancer effect of free chrysin on AGS human gastric cell line. METHODS Properties of the chrysin encapsulated in PLGA-PEG nanoparticles were investigated by SEM, H NMR, and FTIR. The assessment of cytotoxicity on the growth of the human gastric cell line was carried out through MTT assay. After treating the cells with a prearranged amount of pure and encapsulated chrysin, RNA was extracted and the expressions of miR-22, miR-34a and miR-126 were measured by using real-time PCR. RESULTS With regard to the amount of the chrysin loaded in PLGA-PEG nanoparticles, IC50 value was significantly decreased in nanocapsulatedchrysin, in comparison with free chrysin. This finding has been proved through the further increase of miR-22, miR-34a and miR-126 gene expression of nanocapsulatedchrysin, in comparison with free chrysin. CONCLUSIONS In this study, we revealed that the PLGA-PEG-chrysin is more effective than free chrysin in inhibiting the growth of human gastric cell line.
Collapse
Affiliation(s)
- Farideh Mohammadian
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, IR Iran
| | - Alireza Abhari
- Department of Clinical Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, IR Iran
| | - Hassan Dariushnejad
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, IR Iran
| | - Alireza Nikanfar
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, IR Iran
| | - Younes Pilehvar-Soltanahmadi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, IR Iran
| | - Nosratollah Zarghami
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, IR Iran
| |
Collapse
|
35
|
Kuligowski J, Sanjuan-Herráez D, Vázquez-Sánchez MA, Brunet-Vega A, Pericay C, Ramírez-Lázaro MJ, Lario S, Gombau L, Junquera F, Calvet X, Quintás G. Metabolomic Analysis of Gastric Cancer Progression within the Correa's Cascade Using Ultraperformance Liquid Chromatography-Mass Spectrometry. J Proteome Res 2016; 15:2729-38. [PMID: 27384260 DOI: 10.1021/acs.jproteome.6b00281] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Gastric cancer (GC) is among the most common cancers worldwide. Gastric carcinogenesis is a multistep and multifactorial process beginning with chronic gastritis induced by Helicobacter pylori (H. pylori) infection. This process is often described via a sequence of events known as Correas's cascade, a stepwise progression from nonactive gastritis, chronic active gastritis, precursor lesions of gastric cancer (atrophy, intestinal metaplasia, and dysplasia), and finally adenocarcinoma. Our aim was to identify a plasma metabolic pattern characteristic of GC through disease progression within the Correa's cascade. This study involved the analysis of plasma samples collected from 143 patients classified in four groups: patients with nonactive gastritis and no H. pylori infection, H. pylori infected patients with chronic active gastritis, infected or noninfected patients with precursor lesions of gastric cancer, and GC. Independent partial least-squares-discriminant binary models of UPLC-ESI(+)-TOFMS metabolic profiles, implemented in a decision-directed acyclic graph, allowed the identification of tryptophan and kynurenine as discriminant metabolites that could be attributed to indoleamine-2,3-dioxygenase upregulation in cancer patients leading to tryptophan depletion and kynurenine metabolites generation. Furthermore, phenylacetylglutamine was also classified as a discriminant metabolite. Our data suggest the use of tryptophan, kynurenine, and phenylacetylglutamine as potential GC biomarkers.
Collapse
Affiliation(s)
| | - Daniel Sanjuan-Herráez
- Safety and Sustainability Division, Leitat Technological Center , Barcelona 08028, Spain
| | | | | | | | - María José Ramírez-Lázaro
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III , Madrid 28029, Spain
| | - Sergio Lario
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III , Madrid 28029, Spain
| | - Lourdes Gombau
- Safety and Sustainability Division, Leitat Technological Center , Barcelona 08028, Spain
| | - Félix Junquera
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III , Madrid 28029, Spain
| | - Xavier Calvet
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III , Madrid 28029, Spain
| | - Guillermo Quintás
- Safety and Sustainability Division, Leitat Technological Center , Barcelona 08028, Spain
| |
Collapse
|
36
|
Xiao H, Zhang Y, Kim Y, Kim S, Kim JJ, Kim KM, Yoshizawa J, Fan LY, Cao CX, Wong DTW. Differential Proteomic Analysis of Human Saliva using Tandem Mass Tags Quantification for Gastric Cancer Detection. Sci Rep 2016; 6:22165. [PMID: 26911362 PMCID: PMC4766442 DOI: 10.1038/srep22165] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 02/04/2016] [Indexed: 02/07/2023] Open
Abstract
Novel biomarkers and non-invasive diagnostic methods are urgently needed for the screening of gastric cancer to reduce its high mortality. We employed quantitative proteomics approach to develop discriminatory biomarker signatures from human saliva for the detection of gastric cancer. Salivary proteins were analyzed and compared between gastric cancer patients and matched control subjects by using tandem mass tags (TMT) technology. More than 500 proteins were identified with quantification, and 48 of them showed significant difference expression (p < 0.05) between normal controls and gastric cancer patients, including 7 up-regulated proteins and 41 down-regulated proteins. Five proteins were selected for initial verification by ELISA and three were successfully verified, namely cystatin B (CSTB), triosephosphate isomerase (TPI1), and deleted in malignant brain tumors 1 protein (DMBT1). All three proteins could differentiate gastric cancer patients from normal control subjects, dramatically (p < 0.05). The combination of these three biomarkers could reach 85% sensitivity and 80% specificity for the detection of gastric cancer with accuracy of 0.93. This study provides the proof of concept of salivary biomarkers for the non-invasive detection of gastric cancer. It is highly encouraging to turn these biomarkers into an applicable clinical test after large scale validation.
Collapse
Affiliation(s)
- Hua Xiao
- State Key Laboratory of Microbial Metabolism, Laboratory of Bioseparation and Analytical Biochemistry, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yan Zhang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yong Kim
- Dental Research Institute, School of Dentistry, University of California-Los Angeles, Los Angeles, California, 90095, USA
| | - Sung Kim
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 030031, Korea
| | - Jae Joon Kim
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 030031, Korea
| | - Kyoung Mee Kim
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 030031, Korea
| | - Janice Yoshizawa
- Dental Research Institute, School of Dentistry, University of California-Los Angeles, Los Angeles, California, 90095, USA
| | - Liu-Yin Fan
- State Key Laboratory of Microbial Metabolism, Laboratory of Bioseparation and Analytical Biochemistry, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Cheng-Xi Cao
- State Key Laboratory of Microbial Metabolism, Laboratory of Bioseparation and Analytical Biochemistry, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - David T W Wong
- Dental Research Institute, School of Dentistry, University of California-Los Angeles, Los Angeles, California, 90095, USA
| |
Collapse
|
37
|
Dibb M, Han N, Choudhury J, Hayes S, Valentine H, West C, Sharrocks AD, Ang YS. FOXM1 and polo-like kinase 1 are co-ordinately overexpressed in patients with gastric adenocarcinomas. BMC Res Notes 2015; 8:676. [PMID: 26576650 PMCID: PMC4650505 DOI: 10.1186/s13104-015-1658-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 11/02/2015] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Gastric cancers present late in life with advanced disease and carry a poor prognosis. Polo-like Kinase 1 (PLK1) is a mitotic kinase with regulatory functions during G2/M and mitosis in the cell cycle. In mammalian cells, there is an intricate co-regulatory relationship between PLK1 and the forkhead transcription factor FOXM1. It has been demonstrated that individually either PLK1 or FOXM1 expression predicts poorer survival. However, the co-expression of both of these markers in gastric adenocarcinomas has not been reported previously. METHODS We aimed to assess the expression of PLK1 and FOXM1 in Gastric adenocarcinomas in a Western Population, to examine whether there is a relationship of PLK1 to FOXM1 in cancer samples. We assess both the protein and mRNA expression in this patient population by Tissue Microarray immunohistochemistry and RT-PCR. RESULTS Immunohistochemistry was performed on biopsy samples from 79 patients with gastric cancer. Paired normal controls were available in 47 patients. FOXM1 expression was significantly associated with gastric adenocarcinoma (p = 0.001). PLK1 and FOXM1 co-expression was demonstrated in 6/8 (75 %) tumours when analysed by RT-PCR. FOXM1 is overexpressed in a large proportion of gastric carcinomas at the protein level and FOXM1 and PLK1 are concomitantly overexpressed at the mRNA level in this cancer type. CONCLUSIONS This study has demonstrated that FOXM1 and its target gene PLK1 are coordinately overexpressed in a proportion of gastric adenocarcinomas. This suggests that chemotherapeutic treatments that target this pathway may be of clinical utility.
Collapse
Affiliation(s)
- M Dibb
- Faculty of Life Sciences, University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK.
- Faculty of Medical and Human Sciences, University of Manchester, Oxford Road, Manchester, UK.
| | - N Han
- Faculty of Life Sciences, University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK.
| | - J Choudhury
- Department of Histopathology, Salford Royal Foundation Trust, Stott Lane, Salford, M6 8HD, UK.
| | - S Hayes
- Faculty of Medical and Human Sciences, University of Manchester, Oxford Road, Manchester, UK.
- Department of Histopathology, Salford Royal Foundation Trust, Stott Lane, Salford, M6 8HD, UK.
| | - H Valentine
- School of Cancer and Enabling Sciences, Christie Hospital, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK.
| | - C West
- School of Cancer and Enabling Sciences, Christie Hospital, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK.
| | - A D Sharrocks
- Faculty of Life Sciences, University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK.
| | - Yeng S Ang
- Faculty of Medical and Human Sciences, University of Manchester, Oxford Road, Manchester, UK.
- GI Science Centre, Salford Royal NHS FT, University of Manchester, Stott Lane, Salford, M6 8HD, UK.
| |
Collapse
|
38
|
Yuan M, Li Y, Zhong C, Li Y, Niu J, Gong J. Overexpression of neuritin in gastric cancer. Oncol Lett 2015; 10:3832-3836. [PMID: 26788217 DOI: 10.3892/ol.2015.3793] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 08/17/2015] [Indexed: 01/12/2023] Open
Abstract
The aim of the present study was to investigate the expression of neuritin in gastric cancer tissues, in order to explore the association between the expression of neuritin and the occurrence and development of gastric cancer. Tissue specimens were collected from 58 patients with gastric cancer. Immunohistochemistry, western blot analysis and reverse transcription-polymerase chain reaction (RT-PCR) were used to determine the expression of neuritin in the gastric cancer and corresponding adjacent normal gastric tissues. The expression rate of neuritin in gastric cancer tissues was 96.55% (56/58), demonstrating no statistically significant difference from the expression rate in the adjacent normal tissues (94.83%) (P>0.05). However, the rate of strong neuritin expression in gastric cancer tissues (82.76%) was significantly increased compared with the rate in the adjacent normal tissues (15.52%) (P<0.05). Neuritin expression exhibited no correlation with the gender or age of patients, tumor-node-metastasis staging, tumor depth, presence of lymph node metastasis, histological or pathological type of the tumor or presence of distant metastasis (P>0.05). As determined by RT-PCR and western blot analysis, the mRNA expression of neuritin in gastric cancer tissues was markedly increased compared with the expression in the adjacent normal tissues. In conclusion, neuritin is highly expressed in gastric cancer tissues, suggesting that neuritin may act as a novel potential target for the treatment of gastric cancer.
Collapse
Affiliation(s)
- Ming Yuan
- Department of General Surgery, Tongji Hospital at Huazhong Institute of Science and Technology, Wuhan, Hubei 430030, P.R. China; Department of Hepatobiliary Surgery, The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang 832006, P.R. China
| | - Yongjun Li
- Department of General Surgery, Tongji Hospital at Huazhong Institute of Science and Technology, Wuhan, Hubei 430030, P.R. China; Department of Hepatobiliary Surgery, The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang 832006, P.R. China
| | - Chen Zhong
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang 832006, P.R. China
| | - Yongkang Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang 832006, P.R. China
| | - Jianhua Niu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang 832006, P.R. China
| | - Jianping Gong
- Department of General Surgery, Tongji Hospital at Huazhong Institute of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
39
|
Chen S, Zhu J, Yu F, Tian Y, Ma S, Liu X. Combination of miRNA and RNA functions as potential biomarkers for gastric cancer. Tumour Biol 2015; 36:9909-18. [PMID: 26168960 DOI: 10.1007/s13277-015-3756-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 07/02/2015] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer (GC) is the second leading cause of cancer-related death in the world. The optimal treatment regimens for GC depend on tumor stage, histopathological subtype, and other factors. The detection of tumor biomarkers is a quick way to get information of the tumor state. In this study, new biomarkers are detected for GC diagnostic and prognostic purposes. A total of 305 cases of diagnosed gastric adenocarcinoma were enrolled, microRNAs (miRNAs) and their transcriptome sequencing data were obtained from the "The Cancer Genome Atlas." Blood samples were collected from GC patients before surgery and therapy. The miRNA levels and the expression of RNA were detected by real-time RT-PCR. Receiver operating characteristic analysis was used to evaluate the sensitivity and specificity of biomarkers. The combining predictors were established with the logistic regression analysis. Hundreds of miRNA were with higher area under curve (AUC) than 0.5; among them, nine miRNAs were with the highest AUC more than 0.90 and displayed strong diagnostic value. Moreover, the mir-17 level was correlated with tumor stage (p = 0.029), while mir-133b, mir-133a-2, and mir-1-2 levels were significantly correlated with race, tumor pathologic, and tumor stage (p < 0.05). The combination biomarker (mir-181a-1/KAT2B with a sensitivity of 95.83 % and specificity of 94.12 %) could be used as an independent diagnostic indicator for GC patients. For GC patients, mir-17, mir-133b, mir-133a-2, and mir-1-2 appear to be a potential novel predictor of tumor stage and preoperative and intraoperative diagnosis. The combination of miRNA and mRNA such as mir-181a-1/KAT2B (with a sensitivity of 95.83 % and specificity of 94.12 %) showed significant improvement in the diagnostic accuracy.
Collapse
Affiliation(s)
- Silin Chen
- Key Laboratory of Radiobiology (Ministry of Health), School of Public Health, Jilin University, Changchun, 130021, China
| | - Jiaming Zhu
- 2nd Hospital Jilin University, Changchun, 130041, China
| | - Feifei Yu
- Key Laboratory of Radiobiology (Ministry of Health), School of Public Health, Jilin University, Changchun, 130021, China
| | - Yuxi Tian
- Key Laboratory of Radiobiology (Ministry of Health), School of Public Health, Jilin University, Changchun, 130021, China
| | - Shumei Ma
- Key Laboratory of Radiobiology (Ministry of Health), School of Public Health, Jilin University, Changchun, 130021, China. .,Institute of Cancer Care, University of Manitoba, Winnipeg, MB, R3T 3T2, Canada.
| | - Xiaodong Liu
- Key Laboratory of Radiobiology (Ministry of Health), School of Public Health, Jilin University, Changchun, 130021, China.,Center for Radiological Research, Columbia University, New York, NY, 10032, USA
| |
Collapse
|
40
|
Abstract
Background The identification of cancer biomarkers can advance the possibility for early detection and better monitoring of tumor progression. The aim of this study was to assess the diagnostic and prognostic value of serum galectin-3(Gal-3) in patients with gastric cancer (GC). Material/Methods We measured serum Gal-3 levels using ELISA method in 87 patients with GC, 53 patients with benign gastric lesions, and 51 healthy controls. Results Serum levels of Gal-3 in patients with GC were significantly higher than those in benign disease patients and healthy controls (P<0.001), but no difference was found between benign disease patients and healthy controls (P=0.635). Additionally, serum Gal-3 level was associated with lymph node metastasis (P=0.001) and distant metastasis (P<0.001), whereas it was not related to gender (P=0.204), age (P=0.269), tumor size (P=0.399), location (P=0.715), TNM stage (P=0.385), differentiation (P=0.135), or invasion depth (P=0.273). The Kaplan-Meier survival analysis revealed that overall survival rates in patients with high Gal-3 levels were not significantly different that those with low Gal-3 levels (P=0.099). Conclusions Results of the current study suggests that serum Gal-3 represents a potential diagnostic marker for patients with GC, and may be an adjunct to determine the individual prognosis of these patients.
Collapse
Affiliation(s)
- Daye Cheng
- Department of Transfusion, First Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Bin Liang
- Biochip Center, College of Basic Medicine, China Medical University, Shenyang, Liaoning, China (mainland)
| | - Yunhui Li
- Department of Clinical Laboratory Medicine, No. 202 Hospital, Shenyang, Liaoning, China (mainland)
| |
Collapse
|
41
|
Yoshinaga T, Shigemitsu T, Nishimata H, Takei T, Yoshida M. Angiopoietin-like protein 2 is a potential biomarker for gastric cancer. Mol Med Rep 2014; 11:2653-8. [PMID: 25484242 DOI: 10.3892/mmr.2014.3040] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 09/18/2014] [Indexed: 12/14/2022] Open
Abstract
Globally, gastric cancer is one of the most common types of cancer and is the second leading cause of cancer‑induced mortality. Early detection of gastric cancer is able to contribute to a reduction of its mortality. For early detection, more specific and sensitive biomarkers than the classic biomarkers, including carcinoembryonic antigen, carbohydrate antigen 19‑9 and C‑reactive protein, are required. The present study focused on the evaluation of the potential of angiopoietin‑like protein 2 (ANGPTL2) as a novel biomarker for gastric cancer. The expression levels of ANGPTL2 in undifferentiated and differentiated gastric cancer cell lines (HGC‑27 and MKN7, respectively) were therefore investigated. Additionally, ANGPTL2 levels in the serum of gastric cancer patients were compared with those of healthy individuals to evaluate the possibility of the protein as a predictive biomarker for gastric cancer. It was established that the expression levels of ANGPTL2 mRNA and protein were higher in undifferentiated HGC‑27 cells than those in differentiated MKN7 cells. In a patient study, it was indicated that the levels of ANGPTL2 in the serum of gastric cancer patients were higher than those in healthy controls. The diagnostic performance of ANGPTL2 was assessed by constructing a receiver operating characteristic (ROC) curve and was evaluated by calculating the area under each ROC curve (AUC). For the discrimination of patients with gastric cancer from healthy individuals, the AUC for ANGPTL2 was 0.774 (P=0.005) (95% confidence interval, 0.615‑0.933). These results suggested that ANGPTL2 was a potential biomarker for gastric cancer.
Collapse
Affiliation(s)
- Takuma Yoshinaga
- Department of Chemical Engineering, Graduate School of Science and Engineering, Kagoshima University, Kagoshima 890‑0065, Japan
| | - Takamasa Shigemitsu
- Department of Chemical Engineering, Graduate School of Science and Engineering, Kagoshima University, Kagoshima 890‑0065, Japan
| | - Hiroto Nishimata
- Department of Gastroenterology, Nanpuh Hospital, Kagoshima 892-0854, Japan
| | - Takayuki Takei
- Department of Chemical Engineering, Graduate School of Science and Engineering, Kagoshima University, Kagoshima 890‑0065, Japan
| | - Masahiro Yoshida
- Department of Chemical Engineering, Graduate School of Science and Engineering, Kagoshima University, Kagoshima 890‑0065, Japan
| |
Collapse
|
42
|
Zhao Z, Li Y, Liu S, Fu W. Serum Helicobacter pylori CagA antibody may not be used as a tumor marker for diagnosing gastric cancer in east Asian countries. Tumour Biol 2014; 35:12217-24. [PMID: 25168369 DOI: 10.1007/s13277-014-2530-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 08/20/2014] [Indexed: 12/21/2022] Open
Abstract
Gastric cancer (GC) is a highly malignant cancer with increasing incidence and mortality worldwide. Serum Helicobacter pylori CagA antibody has been widely reported to play an important role in diagnosing GC. However, published data on this subject are inconclusive. Therefore, we performed a meta-analysis to evaluate the sensitivity and specificity of serum H. pylori CagA antibody in the diagnosis of GC. We conducted a comprehensive search to identify eligible related studies, in which the pooled sensitivity, specificity, diagnostic odds ratio (DOR), and summary receiver operating characteristic (SROC) curves could be determined. A total of 12 studies including 1,524 cases and 3,324 controls who fulfilled all of the inclusion criteria were included for analysis. The summary estimates for serum H. pylori CagA antibody in the diagnosis of GC in these studies were pooled sensitivity 0.71 (95 % CI 0.69-0.73), specificity 0.40 (95 % CI 0.39-0.42), DOR 2.11 (95 % CI 1.55-2.8), and the area under the curve was 0.636. Our meta-analysis showed that serum H. pylori CagA antibody should not be used for detecting GC.
Collapse
Affiliation(s)
- Zhicheng Zhao
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | | | | | | |
Collapse
|
43
|
Zheng L, Wu C, Xi P, Zhu M, Zhang L, Chen S, Li X, Gu J, Zheng Y. The survival and the long-term trends of patients with gastric cancer in Shanghai, China. BMC Cancer 2014; 14:300. [PMID: 24779704 PMCID: PMC4243141 DOI: 10.1186/1471-2407-14-300] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Accepted: 04/23/2014] [Indexed: 12/17/2022] Open
Abstract
Background Gastric cancer remains a major health issue and a leading cause of death worldwide. This study presented a long-term survival data of gastric cancer registered in Shanghai of China from 1972–2003, with aims to describe the trends as well as the age, sex, stage and tumor sites specific characteristics. Methods The main source of information on cancer cases was the notification card sending to the registry. The residential status of cancer cases was confirmed by home-visits. The methods of follow-up have been a mixture of both active and passive ones. Results We observed an increased trend of survival probability during the last decades. Patients diagnosed during 1972–1976 had a 5-years relative survival rate at 12% for males and 11% for females, respectively, which had dramatically increased to 30% for male and 32% for female patients respectively during the period of 2002–2003. Among the patients diagnosed in 2002–2003, the overall survival probability declined with patient’s age at the time of diagnosis. The lowest survival rate was observed among the oldest group, with the median survival time of 0.8 years. Patients diagnosed with stage I had a higher relative survival rate. Patients with cardia cancer had the worst prognosis, with the 5-year relative survival rate of 29%. Conclusions The survival probability of patients with gastric cancer in Shanghai has improved significantly during the last decades. Age, stage and site of tumor have an impact on prognosis.
Collapse
Affiliation(s)
- Leizhen Zheng
- Department of Oncology, Xin Hua Hospital affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Kim SY, Jung SW, Hyun JJ, Koo JS, Choung RS, Yim HJ, Lee SW, Choi JH. Is colonoscopic screening necessary for patients with gastric adenoma or cancer? Dig Dis Sci 2013; 58:3263-9. [PMID: 23955386 DOI: 10.1007/s10620-013-2824-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Accepted: 07/23/2013] [Indexed: 12/15/2022]
Abstract
BACKGROUND Since colorectal adenoma or cancer is commonly associated with gastric adenoma or cancer, early colorectal adenoma detection can affect the survival of gastric adenoma or cancer patients. AIMS The purpose here was to investigate the colorectal adenoma or cancer prevalence and evaluate the necessity for screening colonoscopy in gastric adenoma or cancer patients. PATIENTS AND METHODS From September 2005 through August 2010, 857 patients younger than 70 years who had gastric adenoma or cancer were enrolled. Healthy age- and sex-matched controls were selected from the general screening population. The prevalence and risk of colorectal adenoma or cancer were compared between the participants and the controls. RESULTS Data from 416 patients in the gastric neoplasm group (123 with gastric adenoma and 293 with gastric cancer) and 416 healthy control group participants were included in the statistical analysis. The presence of gastric adenoma or cancer was an independent risk factor for colorectal neoplasm (OR = 1.348, 95 % CI = 1.001-1.815). Patients with diffuse type gastric cancer had a lower prevalence of colorectal adenoma or cancer than those with gastric adenoma or intestinal type cancer. In gastric cancer patients younger than 50 years, intestinal type histology was significantly associated with colorectal adenoma or cancer (OR = 3.838, 95 % CI = 1.077-13.677). CONCLUSIONS The colorectal adenoma or cancer risk was significantly increased in patients with gastric adenoma or cancer. Therefore, screening colonoscopy should be considered for gastric adenoma or cancer patients including young patients, in the case of intestinal type gastric cancer.
Collapse
Affiliation(s)
- Seung Young Kim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, 516 Gojan-dong, Danwon-gu, Ansan, Gyeonggi-do, 425-707, Korea
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Zeng Z, Wang J, Zhao L, Hu P, Zhang H, Tang X, He D, Tang S, Zeng Z. Potential role of microRNA-21 in the diagnosis of gastric cancer: a meta-analysis. PLoS One 2013; 8:e73278. [PMID: 24023850 PMCID: PMC3762732 DOI: 10.1371/journal.pone.0073278] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 07/18/2013] [Indexed: 12/15/2022] Open
Abstract
Introduction Accumulating evidences indicate that microRNA-21(miR-21) show significant high concentration in plasma of gastric cancer (GC) patients compared to normal individuals, suggesting that it may be a useful novel diagnostic biomarker for gastric cancer. Therefore, we aimed to assess the potential diagnostic value of miR-21 for gastric cancer in this study. Methods Literature database including PubMed, Embase, the Cochrane Library, Web of Science, Ovid, SciVerse, Science Direct, Scopus, BioMed Central, Biosis previews,Chinese Biomedical Literature Database (CBM), Chinese National Knowledge Infrastructure (CNKI), Technology of Chongqing (VIP), and Wan Fang DATA were searched for publications concerning the diagnostic value of miR-21 for GC without language restriction. The quality of each study was scored with the Quality Assessment of Diagnostic Accuracy Studies (QUADAS). Then, data were retrieved from any qualified article hits and subject to meta-analysis. Receiver operating characteristic curves (ROC) were used to check the overall test performance. Evidence of heterogeneity was evaluated using the Chi-square and I2 test. Results Five studies with a total 251 GC patients and 184 control individuals were included in this meta-analysis. All of the included studies are of high quality (QUADAS score$13). The summary estimates revealed that the pooled sensitivity is 66.5% (95% confidence interval (CI): 55.0%–76.3%) and the specificity is 83.1% (95% CI: 69.4%–91.5%). In addition, the area under the summary ROC curve (AUC) is 0.80. Conclusion The current evidence suggests that miR-21 has potential diagnostic value with a moderate sensitivity and specificity for GC. More prospective studies on the diagnostic value of miR-21 for GC are needed in the future.
Collapse
Affiliation(s)
- Zongyue Zeng
- College of Laboratory Medicine, Key Laboratory of Laboratory Medical Diagnostics designated by Chinese Ministry of Education, Chong Qing Medical University, Chongqing, China
| | - Jiangen Wang
- College of Laboratory Medicine, Key Laboratory of Laboratory Medical Diagnostics designated by Chinese Ministry of Education, Chong Qing Medical University, Chongqing, China
| | - Liuyang Zhao
- College of Laboratory Medicine, Key Laboratory of Laboratory Medical Diagnostics designated by Chinese Ministry of Education, Chong Qing Medical University, Chongqing, China
| | - Ping Hu
- College of Laboratory Medicine, Key Laboratory of Laboratory Medical Diagnostics designated by Chinese Ministry of Education, Chong Qing Medical University, Chongqing, China
| | - Hailong Zhang
- College of Laboratory Medicine, Key Laboratory of Laboratory Medical Diagnostics designated by Chinese Ministry of Education, Chong Qing Medical University, Chongqing, China
| | - Xi Tang
- College of Laboratory Medicine, Key Laboratory of Laboratory Medical Diagnostics designated by Chinese Ministry of Education, Chong Qing Medical University, Chongqing, China
| | - Dali He
- College of Laboratory Medicine, Key Laboratory of Laboratory Medical Diagnostics designated by Chinese Ministry of Education, Chong Qing Medical University, Chongqing, China
| | - Shifu Tang
- College of Laboratory Medicine, Key Laboratory of Laboratory Medical Diagnostics designated by Chinese Ministry of Education, Chong Qing Medical University, Chongqing, China
| | - Zhaofang Zeng
- College of Laboratory Medicine, Key Laboratory of Laboratory Medical Diagnostics designated by Chinese Ministry of Education, Chong Qing Medical University, Chongqing, China
- * E-mail:
| |
Collapse
|
46
|
Premedication with N-acetylcysteine and simethicone improves mucosal visualization during gastroscopy: a randomized, controlled, endoscopist-blinded study. Eur J Gastroenterol Hepatol 2013; 25:778-83. [PMID: 23739279 DOI: 10.1097/meg.0b013e32836076b2] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECTIVES Diagnostic gastroscopy provides a unique opportunity to diagnose early oesophagogastric neoplasia; however, intraluminal mucus and bile can obscure mucosal visualization. The aim of this study was to determine whether the use of a premedication solution containing the mucolytic agent N-acetylcysteine and the surfactant simethicone improves mucosal visualization within a UK diagnostic gastroscopy service. MATERIALS AND METHODS A total of 75 consecutive patients were recruited from a single (S.J.) endoscopist's diagnostic gastroscopy list. They were randomized into three treatment groups: (a) standard control=clear fluids only for 6 h, nil by mouth for 2 h; (b) water control=standard control+100 ml sterile water (given 20 min before gastroscopy); and (c) solution=standard control+100 ml investigated solution (20 min before gastroscopy). The endoscopist was blinded to patient preparation. Inadequate mucosal visualization was defined as fluid/mucus during gastroscopy that could not be suctioned and required flushing with water. The volume of flush, the site at which it was used and the total procedure times were recorded. RESULTS All three groups showed no statistical difference for age, sex ratio, procedure priority or indication. The mean volume of flush required to obtain clear mucosa was significantly less in the solution group compared with the other groups. The mean overall procedure time was also less in the solution group compared with the other groups. DISCUSSION Premedication with N-acetylcysteine and simethicone markedly improves mucosal visibility during gastroscopy. It also reduces the time taken for the procedure. This low-cost and well-tolerated intervention may improve detection of early neoplasia.
Collapse
|
47
|
Cai JP, Wu YJ, Li C, Feng MY, Shi QT, Li R, Wang ZY, Geng JS. Panax ginseng polysaccharide suppresses metastasis via modulating Twist expression in gastric cancer. Int J Biol Macromol 2013; 57:22-5. [PMID: 23500436 DOI: 10.1016/j.ijbiomac.2013.03.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 02/24/2013] [Accepted: 03/02/2013] [Indexed: 12/12/2022]
Abstract
It was previously reported that an antitumor polysaccharide (PGPW1) was isolated from the root of Panax ginseng. To extend our study, we investigated here the anti-invasive and metastatic effects of PGPW1 on human gastric cancer cell line HGC-27 and tried to determine its possible mechanism of action. Both scratch wound-healing and Transwell assay identified that PGPW1 dose-dependently inhibited migration and invasiveness of HGC-27 cells. Furthermore, results of western blot showed that protein levels of Twist and AKR1C2 were inhibited by PGPW1, whereas an increase of NF1 was observed. Moreover, down-regulation of Twist expression by PGPW1 blocked epithelial-mesenchymal transition (EMT), characterized by a gain of epithelial cell markers, E-cadherin, and loss of the mesenchymal markers, vimentin and N-cadherin, at protein levels. Collectively, we confirmed that PGPW1 decreased migration and invasion of HGC-27 cells by regulation of Twist, AKR1C2, NF1, E-cadherin, vimentin and N-cadherin expression. In conclusion, PGPW1 may serve as a powerful chemopreventive agent against gastric cancer metastasis.
Collapse
Affiliation(s)
- Jian-Ping Cai
- Department of Pathology, The Third Affiliated (Tumor) Hospital, Harbin Medical University, Harbin 150040, China
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Wang M, Gu H, Qian H, Zhu W, Zhao C, Zhang X, Tao Y, Zhang L, Xu W. miR-17-5p/20a are important markers for gastric cancer and murine double minute 2 participates in their functional regulation. Eur J Cancer 2013; 49:2010-21. [PMID: 23333058 DOI: 10.1016/j.ejca.2012.12.017] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2012] [Accepted: 12/05/2012] [Indexed: 02/07/2023]
Abstract
AIM To investigate the potential roles and mechanisms of miR-17-5p/20a in human gastric cancer development and progression. METHODS Quantitative real-time polymerase chain reaction (qRT-PCR) was employed to determine miR-17-5p/20a expression profiles in 110 gastric cancer tissues. microRNAs' (miRNAs) mimics and inhibitors were used to reveal their function in gastric cancer. Antagomirs were applied to treating gastric cancer cell derived xenograft in vivo. Western blot and luciferase assays were performed to uncover the targets and mechanisms of miR-17-5p/20a. RESULTS miR-17-5p/20a levels were upregulated in human gastric cancer tissues. Overexpression of miR-17-5p/20a promoted gastric cancer cell cycle progression and inhibited cell apoptosis, whereas knockdown of miR-17-5p/20a resulted in cell cycle arrest and increased apoptosis. p21 and tumour protein p53-induced nuclear protein 1 (TP53INP1) were validated as the targets of miR-17-5p/20a. Antagomirs against miR-17-5p/20a significantly inhibited gastric cancer growth via upregulation of p21 and TP53INP1 in a mouse xenograft model. The negative relationship between miR-17-5p/20a and TP53INP1 was observed in patient gastric cancer tissues. Murine double minute 2 (MDM2) was found to be involved in miRNA regulation and function. Targeted inhibition of MDM2 in a miRNA mimic-transfected gastric cancer cell line abolished miR-17-5p/20a function and inhibition of p21 expression. MDM2 restoration by pCMV-MDM2 rescued the functionality. CONCLUSIONS Our findings indicate that miR-17-5p/20a promote gastric cancer cell proliferation and inhibit cell apoptosis via post-transcriptional modulation of p21 and TP53INP1. They may be promising therapeutic markers for gastric cancer. MDM2 contributes to miR-17-5p/20a function and inhibition of p21 in gastric cancer, and may be a novel mechanism underlying the oncogenic roles of miR-17-5p/20a.
Collapse
Affiliation(s)
- Mei Wang
- School of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Yu X, Luo L, Wu Y, Yu X, Liu Y, Yu X, Zhao X, Zhang X, Cui L, Ye G, Le Y, Guo J. Gastric juice miR-129 as a potential biomarker for screening gastric cancer. Med Oncol 2013; 30:365. [PMID: 23307240 DOI: 10.1007/s12032-012-0365-y] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Accepted: 09/13/2012] [Indexed: 12/19/2022]
Abstract
MicroRNAs (miRNAs) play crucial roles during the occurrence and development of gastric cancer. Conventional serological tests for screening gastric cancer have limits on sensitivity and specificity. Several miRNAs in peripheral blood have been used as biomarkers of gastric cancer. However, most of these miRNAs are shared by several types of cancer. Thanks to the tissue specificity of gastric juice, here we examined the feasibility of using gastric juice miR-129-1/2, which are aberrantly expressed in gastric cancer, to screen gastric cancer. Total of 141 gastric juices samples from gastric cancer, gastric ulcer, atrophic gastritis, and minimal gastritis patients or subjects with normal mucosa were collected by gastroscopy. The gastric juice miR-129-1/2 levels were detected by quantitative reverse transcription-polymerase chain reaction. A receiver operating characteristic (ROC) curve was constructed for differentiating patients with gastric cancer from patients with benign gastric diseases. We showed that, compared with patients with benign gastric diseases, patients with gastric cancer had significantly lower levels of gastric juice miR-129-1-3p and miR-129-2-3p. The areas under ROC curve (AUC) were 0.639 and 0.651 for miR-129-1-3p and miR-129-2-3p, respectively. Using the parallel combination test, the AUC was up to 0.656. In summary, our results suggest that gastric juice miR-129-1-3p and miR-129-2-3p are potential biomarkers for the screening gastric cancer, and the detection of gastric juice miRNAs is a convenient non-invasion method for the diagnosis of gastric cancer.
Collapse
Affiliation(s)
- Xing Yu
- Zhejiang Provincial Key Laboratory of Pathophysiology, Department of Biochemistry and Molecular Biology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo 315211, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Mabula JB, McHembe MD, Koy M, Chalya PL, Massaga F, Rambau PF, Masalu N, Jaka H. Gastric cancer at a university teaching hospital in northwestern Tanzania: a retrospective review of 232 cases. World J Surg Oncol 2012. [PMID: 23181624 PMCID: PMC3527214 DOI: 10.1186/1477-7819-10-257] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Background Despite marked decreases in its incidence, particularly in developed countries, gastric cancer is still the second most common tumor worldwide. There is a paucity of information regarding gastric cancer in northwestern Tanzania. This study was undertaken to describe our experience, in our local setting, on the management of gastric cancer, outlining the clinicopathological and treatment outcome of these patients and suggesting ways to improve the treatment outcome. Methods This was a retrospective study of histologically confirmed cases of gastric cancer seen at Bugando Medical Centre between January 2007 and December 2011. Data were retrieved from patients’ files and analyzed using SPSS computer software version 17.0. Results A total of 232 gastric cancer patients were enrolled in the study, representing 4.5% of all malignancies. The male to female ratio was 2.9:1. The median age of patients was 52 years. The majority of the patients (92.1%) presented late with advanced gastric cancer (Stages III and IV). Lymph node and distant metastasis at the time of diagnosis was recorded in 31.9% and 29.3% of cases, respectively. The antrum was the most frequent anatomical site (56.5%) involved and gastric adenocarcinoma (95.1%) was the most common histopathological type. Out of 232 patients, 223 (96.1%) patients underwent surgical procedures for gastric cancer of which gastro-jejunostomy was the most frequent performed surgical procedure, accounting for 53.8% of cases. The use of chemotherapy and radiotherapy was documented in 56 (24.1%) and 12 (5.1%) patients, respectively. Postoperative complication and mortality rates were 37.1% and 18.1%, respectively. According to multivariate logistic regression analysis, preoperative co-morbidity, histological grade and stage of the tumor, presence of metastases at the time of diagnosis was the main predictors of death (P <0.001). At the end of five years, only 76 (32.8%) patients were available for follow-up and the overall five-year survival rate was 6.9%. Evidence of cancer recurrence was reported in 45 (19.4%) patients. Positive resection margins, stage of the tumor and presence of metastasis at the time of diagnosis were the main predictors of local recurrence (P <0.001). Conclusions Gastric cancer in this region shows a trend towards relative young age at diagnosis and the majority of patients present late with an advanced stage. Lack of awareness of the disease, poor accessibility to health care facilities and lack of screening programs in this region may contribute to advanced disease at the time of diagnosis. There is a need for early detection, adequate treatment and proper follow-up to improve treatment outcome.
Collapse
Affiliation(s)
- Joseph B Mabula
- Department of Surgery, Catholic University of Health and Allied Sciences-Bugando, Mwanza, Tanzania
| | | | | | | | | | | | | | | |
Collapse
|