1
|
Bahardoust M, Mousavi S, Moezi ZD, Yarali M, Tayebi A, Olamaeian F, Tizmaghz A. Effect of Metformin Use on Survival and Recurrence Rate of Gastric Cancer After Gastrectomy in Diabetic Patients: A Systematic Review and Meta-analysis of Observational Studies. J Gastrointest Cancer 2024; 55:65-76. [PMID: 37526857 DOI: 10.1007/s12029-023-00955-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2023] [Indexed: 08/02/2023]
Abstract
BACKGROUND Gastric cancer (GC) is one of the most common worldwide cancers and causes of death. Various studies have investigated the effect of metformin on overall survival (OS), cancer-specific survival (CSS), recurrence-free survival (RFS), and recurrence rate in diabetic patients after gastrectomy, and their results have been contradictory. This meta-analysis aimed to evaluate the effect of metformin use compared to sulfonylurea compounds with OS, CSS, RFS, and recurrence rate after gastrectomy in diabetic patients. METHODS We reviewed the Scopus, Google Scholar, PubMed, Web of Science, and Embassy databases until September 2022 based on appropriate MESH terms. All observational studies that evaluated the effect of metformin on survival in diabetic patients who underwent surgery for GC were included. The hazard ratio (HR) with a 95% confidence interval was used to estimate the effect size. The Egger test was used to evaluate publication bias. RESULTS Overall, nine studies, including 245,387 GC patients who underwent surgery, were included. The use of metformin significantly increased the OS rate (HR: 0.81, 95% CI: 0.78, 0.86, P: 0.001, I2: 4.5%), CSS rate (HR: 0.72, 95% CI: 0.63, 0.81, P: 0.011, I2 = 0%), and RFS rate (HR: 719, 95% CI: 0.524, 0.986, P: 0.001) and decreased the recurrence rate after gastrectomy (HR: 0.83, 95% CI: 0.77, 0.87, P: 0.001, I2: 0%). The use of metformin was significantly associated with a greater increase in OS and CSS rate and a greater decrease in recurrence rate in the Asian population than in the Western population. CONCLUSION The use of metformin in diabetic patients with GC can be associated with improved OS, CSS, RFS, and reduced recurrence rate after gastrectomy, especially in the Asian population.
Collapse
Affiliation(s)
- Mansour Bahardoust
- Department of Epidemiology, School of Public Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Safa Mousavi
- Department of Public Health, College of Health and Human Services, California State University, Fresno, CA, USA
| | - Zahra Deylami Moezi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Yarali
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Ali Tayebi
- Department of General Surgery, School of Medicine, Firoozabadi Clinical Research Development Unit (FACRDU), Iran University of Medical Sciences, Tehran, Iran.
| | - Faranak Olamaeian
- Department of General Surgery, School of Medicine, Firoozabadi Clinical Research Development Unit (FACRDU), Iran University of Medical Sciences, Tehran, Iran
| | - Adnan Tizmaghz
- Department of General Surgery, School of Medicine, Firoozabadi Clinical Research Development Unit (FACRDU), Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Wang L, Zhang Z. Diabetes Mellitus and Gastric Cancer: Correlation and Potential Mechanisms. J Diabetes Res 2023; 2023:4388437. [PMID: 38020199 PMCID: PMC10653978 DOI: 10.1155/2023/4388437] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 10/22/2023] [Accepted: 10/27/2023] [Indexed: 12/01/2023] Open
Abstract
This review summarizes the correlation between diabetes mellitus (DM) and gastric cancer (GC) from the perspectives of epidemiology, drug use, and potential mechanisms. The association between DM and GC is inconclusive, and the positive direction of the association reported in most published meta-analyses suggests that DM may be an independent risk factor for GC. Many clinical investigations have shown that people with DM and GC who undergo gastrectomy may have better glycemic control. The potential link between DM and GC may involve the interaction of multiple common risk factors, such as obesity, hyperglycemia and hyperinsulinemia, H. pylori infection, and the use of metformin. Although in vitro and in vivo data support that H. pylori infection status and metformin can influence GC risk in DM patients, there are conflicting results. Patient survival outcomes are influenced by multiple factors, so further research is needed to identify the patients who may benefit.
Collapse
Affiliation(s)
- Li Wang
- Department of Emergency, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310052, China
- Zhejiang Provincial Critical Research Center for Emergency Medicine Clinic, Hangzhou 310052, China
- Key Laboratory of Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou 310052, China
| | - Zhe Zhang
- Department of Emergency Medicine, The First People's Hospital of Linping District, 311100, Hangzhou, Zhejiang, China
| |
Collapse
|
3
|
Fang CW, Yang JS, Chiang JH, Shieh PC, Tsai FJ, Tsai CW, Chang WS. Metformin induces autophagy of cisplatin-resistant human gastric cancer cells in addition to apoptosis. Biomedicine (Taipei) 2023; 13:14-23. [PMID: 37937302 PMCID: PMC10627204 DOI: 10.37796/2211-8039.1408] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 02/23/2023] [Accepted: 02/23/2023] [Indexed: 11/09/2023] Open
Abstract
Metformin has been used to treat cases of type 2 diabetes mellitus, and mounting studies have shown that metformin can act alone or in synergy with other anticancer agents to achieve anti-cancer efficacies on various types of tumors. However, the role of metformin in either inducing autophagy and cisplatin-resistance of human gastric cancer (GC) cells has never been examined. The study has established a cisplatin-resistant GC cell line and investigated the effects of metformin on inducing autophagy on it. The results demonstrated that treatment with metformin can concentration-dependently suppress the cell viability and cell confluence of cisplatin-resistant GC cells, while having no effects on human primary stomach epithelial cells (HPSEC). For the first time, we found that metformin can significantly increase the acidic vesicular organelles (AVO) level and decrease the acridine orange (AO) level spontaneously in the cisplatin-resistant GC cells. Thus, we further checked the other markers, Atg5, Atg12 and LC3-II, which showed that metformin indeed induced autophagy in the cisplatin-resistant GC cells. In addition, treatment of 3-Methyladenine (3-MA) can significantly rescue the metformin-induced autophagy. At the same time, metformin can induce the alterations of apoptosis-associated signal molecules, such as caspase-3 and caspase-7 activities. Overall, the pilot study provided evidence for metformin induced autophagy in addition to apoptosis, making it as an effective anticancer drug for the therapy of cisplatin-resistant GC. Killing the cisplatin-resistant GC cells with non-toxic metformin via both autophagy and apoptosis might extend its usefulness in our fighting with chemo-resistance of gastric cancer cells.
Collapse
Affiliation(s)
- Chih-Wun Fang
- Division of Pharmacy, Zuoying Branch of Kaohsiung Armed Forces General Hospital, Kaohsiung,
Taiwan
| | - Jai-Sing Yang
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung,
Taiwan
| | - Jo-Hua Chiang
- Department of Nursing, Chung-Jen Junior College of Nursing, Health Sciences and Management, Chiayi,
Taiwan
| | - Po-Chuen Shieh
- Department of Pharmacy, Tajen University, Pingtung,
Taiwan
| | - Fuu-Jen Tsai
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung,
Taiwan
- China Medical University Children’s Hospital, China Medical University, Taichung,
Taiwan
| | - Chia-Wen Tsai
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung,
Taiwan
| | - Wen-Shin Chang
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung,
Taiwan
| |
Collapse
|
4
|
Tseng CH. The Relationship between Diabetes Mellitus and Gastric Cancer and the Potential Benefits of Metformin: An Extensive Review of the Literature. Biomolecules 2021; 11:1022. [PMID: 34356646 PMCID: PMC8301937 DOI: 10.3390/biom11071022] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/02/2021] [Accepted: 07/08/2021] [Indexed: 12/14/2022] Open
Abstract
The objective of this review is to summarize the findings of published research that investigated the relationship between diabetes mellitus and gastric cancer (GCa) and the potential benefits of metformin on GCa. Related literature has been extensively reviewed, and findings from studies investigating the relationship between diabetes mellitus and GCa suggest that hyperglycemia, hyperinsulinemia and insulin resistance are closely related to the development of GCa. Although not supported by all, most observational studies suggest an increased risk of GCa in patients with type 2 diabetes mellitus, especially in women and in Asian populations. Incidence of second primary malignancy diagnosed after GCa is significantly higher in diabetes patients. Diabetes patients with GCa may have more complications after gastrectomy or chemotherapy and they may have a poorer prognosis than patients with GCa but without diabetes mellitus. However, glycemic control may improve in the diabetes patients with GCa after receiving gastrectomy, especially after procedures that bypass the duodenum and proximal jejunum, such as Roux-en-Y gastric bypass or Billroth II reconstruction. The potential links between diabetes mellitus and GCa may involve the interactions with shared risk factors (e.g., obesity, hyperglycemia, hyperinsulinemia, insulin resistance, high salt intake, smoking, etc.), Helicobacter pylori (HP) infection, medications (e.g., insulin, metformin, statins, aspirin, proton pump inhibitors, antibiotics, etc.) and comorbidities (e.g., hypertension, dyslipidemia, vascular complications, heart failure, renal failure, etc.). With regards to the potential benefits of metformin on GCa, results of most observational studies suggest a reduced risk of GCa associated with metformin use in patients with T2DM, which can be supported by evidence derived from many in vitro and animal studies. Metformin use may also reduce the risk of HP infection, an important risk factor of GCa. In patients with GCa, metformin users may have improved survival and reduced recurrence. More studies are required to clarify the pathological subtypes/anatomical sites of GCa associated with type 2 diabetes mellitus or prevented by metformin, to confirm whether GCa risk can also be increased in patients with type 1 diabetes mellitus and to explore the possible role of gastric microbiota in the development of GCa.
Collapse
Affiliation(s)
- Chin-Hsiao Tseng
- Department of Internal Medicine, National Taiwan University College of Medicine, Taipei 10051, Taiwan; ; Tel.: +886-2-2388-3578
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei 10051, Taiwan
- Division of Environmental Health and Occupational Medicine, National Health Research Institutes, Zhunan 350, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital, No. 7 Chung-Shan South Road, Taipei 100, Taiwan
| |
Collapse
|
5
|
Cheung KS, Chung KL, Leung WK. Chemopreventive Effect of Metformin on Gastric Cancer Development. Gut Liver 2021; 16:147-156. [PMID: 34158423 PMCID: PMC8924804 DOI: 10.5009/gnl210132] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/17/2021] [Accepted: 05/12/2021] [Indexed: 12/24/2022] Open
Abstract
Although Helicobacterpylori infection is the most important causative factor for gastric cancer (GC), H. pylori eradication alone does not completely eliminate the GC risk. In addition to H. pylori eradication, other risk factors for GC should be identified and targeted. Diabetes mellitus (DM) confers a 20% increased risk of GC, which could be mediated via several biological mechanisms including the stimulation of cell proliferation via hyperinsulinemia and increased insulin-growth factor production, the promotion of angiogenesis, and DNA damage. With a current global prevalence of 9.3% and a predicted rise to 10.2% by 2030, DM could contribute substantially to the burden of GC cases worldwide. Emerging evidence showed that metformin possesses chemopreventive effects via both direct (e.g., adenosine monophosphate-activated protein kinase activation and subsequent inhibition of the mammalian target of rapamycin pathway) and indirect (e.g., modulation of the interaction between tumor cells and their microenvironment and gut microbiota) pathways. A recent meta-analysis of observational studies showed that metformin use was associated with 24% lower GC risk. However, many available observational studies related to metformin effects suffered from biases including the failure to adjust for the H. pylori infection status and serial glycemic control and time-related biases. Future prospective studies addressing these pitfalls are needed.
Collapse
Affiliation(s)
- Ka Shing Cheung
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China.,Department of Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Kit Lam Chung
- Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Wai K Leung
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| |
Collapse
|
6
|
Xie Y, Zhang J, Lu B, Bao Z, Zhao J, Lu X, Wei Y, Yao K, Jiang Y, Yuan Q, Zhang X, Li B, Chen X, Dong Z, Liu K. Mefloquine Inhibits Esophageal Squamous Cell Carcinoma Tumor Growth by Inducing Mitochondrial Autophagy. Front Oncol 2020; 10:1217. [PMID: 32850358 PMCID: PMC7400730 DOI: 10.3389/fonc.2020.01217] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 06/15/2020] [Indexed: 12/12/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) has a worldwide impact on human health, due to its high incidence and mortality. Therefore, identifying compounds to increase patients' survival rate is urgently needed. Mefloquine (MQ) is an FDA-approved anti-malarial drug, which has been reported to inhibit cellular proliferation in several cancers. However, the anti-tumor activities of the drug have not yet been completely defined. In this study, mass spectrometry was employed to profile proteome changes in ESCC cells after MQ treatment. Sub-cellular localization and gene ontology term enrichment analysis suggested that MQ treatment mainly affect mitochondria. The KEGG pathway enrichment map of down-regulated pathways and Venn diagram indicated that all of the top five down regulated signaling pathways contain four key mitochondrial proteins (succinate dehydrogenase complex subunit C (SDHC), succinate dehydrogenase complex subunit D, mitochondrially encoded cytochrome c oxidase III and NADH: ubiquinone oxidoreductase subunit V3). Meanwhile, mitochondrial autophagy was observed in MQ-treated KYSE150 cells. More importantly, patient-derived xenograft mouse models of ESCC with SDHC high expression were more sensitive to MQ treatment than low SDHC-expressing xenografts. Taken together, mefloquine inhibits ESCC tumor growth by inducing mitochondrial autophagy and SDHC plays a vital role in MQ-induced anti-tumor effect on ESCC.
Collapse
Affiliation(s)
- Yifei Xie
- Department of Pathophysiology, School of Basic Medical Sciences, AMS, Zhengzhou University, Zhengzhou, China
| | - Jing Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, AMS, Zhengzhou University, Zhengzhou, China
| | - Bingbing Lu
- Department of Pathophysiology, School of Basic Medical Sciences, AMS, Zhengzhou University, Zhengzhou, China
| | - Zhuo Bao
- Department of Pathophysiology, School of Basic Medical Sciences, AMS, Zhengzhou University, Zhengzhou, China
| | - Jimin Zhao
- Department of Pathophysiology, School of Basic Medical Sciences, AMS, Zhengzhou University, Zhengzhou, China.,Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou, China
| | - Xianyu Lu
- Department of Pathophysiology, School of Basic Medical Sciences, AMS, Zhengzhou University, Zhengzhou, China
| | - Yaxing Wei
- Department of Pathophysiology, School of Basic Medical Sciences, AMS, Zhengzhou University, Zhengzhou, China
| | - Ke Yao
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
| | - Yanan Jiang
- Department of Pathophysiology, School of Basic Medical Sciences, AMS, Zhengzhou University, Zhengzhou, China
| | - Qiang Yuan
- Department of Pathophysiology, School of Basic Medical Sciences, AMS, Zhengzhou University, Zhengzhou, China
| | - Xiaofan Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, AMS, Zhengzhou University, Zhengzhou, China
| | - Bo Li
- Department of Pathophysiology, School of Basic Medical Sciences, AMS, Zhengzhou University, Zhengzhou, China
| | - Xinhuan Chen
- Department of Pathophysiology, School of Basic Medical Sciences, AMS, Zhengzhou University, Zhengzhou, China.,Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou, China
| | - Zigang Dong
- Department of Pathophysiology, School of Basic Medical Sciences, AMS, Zhengzhou University, Zhengzhou, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, China.,Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, China
| | - Kangdong Liu
- Department of Pathophysiology, School of Basic Medical Sciences, AMS, Zhengzhou University, Zhengzhou, China.,Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, China.,Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, China
| |
Collapse
|
7
|
Ye YQ, Zeng B, Liao YG, Liu MS, Hua ZR. Molecular mechanism for metformin to enhance pro-apoptotic effect of sorafenib in HepG2 cells. Shijie Huaren Xiaohua Zazhi 2020; 28:581-586. [DOI: 10.11569/wcjd.v28.i14.581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Prolonging the survival time of patients with advanced liver cancer is a major clinical challenge. Sorafenib (Sor) is the only targeted drug for the treatment of advanced liver cancer, but drug resistance has become a problem.
AIM To explore the effect and molecular mechanism of metformin (Met) in enhancing pro-apoptotic effect of Sor in HepG2 cells.
METHODS MTT assay was used to evaluate the anti-proliferation effects of Met and Sor, alone or in combination. Flow cytometry was employed to analyze cell cycle and cell apoptosis. The expression of Caspase-3, Bax, AMPK, P53, and mTORC1 was evaluated by Western blot.
RESULTS MTT assay showed that both Met and Sor decreased HepG2 cells viability, and Met combined with Sor had a synergistic inhibitory effect. The relative cell viability rates of the control group, Met group, Sor group, and combination group were 100%, 79.96% ± 4.41%, 85.33% ± 1.00%, and 68.60% ± 4.02%, respectively. Flow cytometry showed that both Met and Sor induced HepG2 cell apoptosis, and Met combined with Sor had a synergistic effect. The apoptosis rates of the control group, Met group, Sor group, and combination group were 4.47% ± 1.93%, 13.73% ± 1.18%, 9.50% ± 0.20%, and 29.03% ± 0.35%, respectively. Western blot analysis showed that both Met and Sor increased the expression of Caspase-3, Bax, P53, and AMPK and decreased the expression of mTORC1.
CONCLUSION Met can enhance the pro-apoptotic effect of Sor in HepG2 cells.
Collapse
Affiliation(s)
- Yan-Qing Ye
- Department of Gastroenterology, the First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, Jiangxi Province, China
| | - Bin Zeng
- Department of Gastroenterology, the First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, Jiangxi Province, China
| | - Yue-Guang Liao
- Department of Gastroenterology, the First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, Jiangxi Province, China
| | - Mao-Sheng Liu
- Department of Gastroenterology, the First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, Jiangxi Province, China
| | - Zong-Rong Hua
- Department of Gastroenterology, the First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, Jiangxi Province, China
| |
Collapse
|
8
|
Wang Y, Maurer MJ, Larson MC, Allmer C, Feldman AL, Bennani NN, Thompson CA, Porrata LF, Habermann TM, Witzig TE, Ansell SM, Slager SL, Nowakowski GS, Cerhan JR. Impact of metformin use on the outcomes of newly diagnosed diffuse large B-cell lymphoma and follicular lymphoma. Br J Haematol 2019; 186:820-828. [PMID: 31135975 DOI: 10.1111/bjh.15997] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 04/15/2019] [Indexed: 12/15/2022]
Abstract
The diabetes mellitus (DM) drug metformin targets mechanistic/mammalian target of rapamycin and inhibits lymphoma growth in vitro. We investigated whether metformin affected outcomes of newly diagnosed diffuse large B-cell (DLBCL, n = 869) and follicular lymphoma (FL, n = 895) patients enrolled in the Mayo component of the Molecular Epidemiology Resource cohort study between 2002 and 2015. Hazard ratios (HR) and 95% confidence intervals (CIs) adjusted for age, sex, body mass index, prognostic index and treatment were used to estimate the association of metformin exposure (No DM/No metformin; DM/No metformin; DM/Metformin) with event-free (EFS), lymphoma-specific (LSS) and overall (OS) survival. Compared to No DM/No metformin DLBCL patients, there was no association of DM/Metformin (n = 48; HR = 1·05, 95% CI 0·59-1·89) or DM/No metformin(n = 54; HR = 1·41, 95% CI 0·88-2·26) with EFS; results were similar for LSS and OS. Compared to No DM/No metformin FL patients, there was no association of DM/Metformin (n = 37; HR = 1·16, 95% CI 0·71-1·89) or DM/No metformin (n = 19; HR = 1·16, 95% CI 0·66-2·04) with EFS; results were similar for LSS. However, DM/Metformin was associated with inferior OS (HR = 2·17; 95% CI 1·19-3·95) compared to No DM/No metformin. In conclusion, we found no evidence that metformin use was associated with improved outcomes in newly diagnosed DLBCL and FL.
Collapse
Affiliation(s)
- Yucai Wang
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Matthew J Maurer
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Melissa C Larson
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Cristine Allmer
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | | | | | | | - Susan L Slager
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | | | - James R Cerhan
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
9
|
Prediabetes and diabetes in relation to risk of gastric adenocarcinoma. Br J Cancer 2019; 120:1147-1152. [PMID: 31061455 PMCID: PMC6738058 DOI: 10.1038/s41416-019-0470-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 04/12/2019] [Accepted: 04/18/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Whether prediabetes or diabetes increases the risk of gastric adenocarcinoma is not clear. METHODS This cohort study included 111,198 participants in the Northern Swedish Health and Disease Study. The participants were followed up from November 1985 to April 2017. The exposure to prediabetes or diabetes was assessed by oral glucose tolerance tests and self-reports. The incidence of the outcome gastric adenocarcinoma was identified from the Swedish Cancer Registry. Multivariable Cox regressions were used to analyse the associations between prediabetes or diabetes and the risk of gastric adenocarcinoma, providing hazard ratios (HR) with 95% confidence intervals (CI), with adjustment for sex, age, calendar year, body mass index, tobacco smoking and education level. RESULTS Compared with normoglycaemic participants, the risk of gastric adenocarcinoma was not increased among participants with prediabetes (HR 1.07, 95% CI 0.79-1.44), diabetes (HR 0.77, 95% CI 0.46-1.29) or any of these exposures (HR 0.96, 95% CI 0.73-1.27). No associations were identified between prediabetes or diabetes and the risk of gastric adenocarcinoma in stratified analyses or in analyses separating cardia and non-cardia gastric adenocarcinoma. CONCLUSIONS This study does not support the hypothesis that prediabetes or diabetes increases the risk of gastric adenocarcinoma.
Collapse
|
10
|
Lu CC, Chiang JH, Tsai FJ, Hsu YM, Juan YN, Yang JS, Chiu HY. Metformin triggers the intrinsic apoptotic response in human AGS gastric adenocarcinoma cells by activating AMPK and suppressing mTOR/AKT signaling. Int J Oncol 2019; 54:1271-1281. [PMID: 30720062 PMCID: PMC6411354 DOI: 10.3892/ijo.2019.4704] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 01/14/2019] [Indexed: 12/12/2022] Open
Abstract
Metformin is commonly used to treat patients with type 2 diabetes and is associated with a decreased risk of cancer. Previous studies have demonstrated that metformin can act alone or in synergy with certain anticancer agents to achieve anti-neoplastic effects on various types of tumors via adenosine monophosphate-activated protein kinase (AMPK) signaling. However, the role of metformin in AMPK-mediated apoptosis of human gastric cancer cells is poorly understood. In the current study, metformin exhibited a potent anti-proliferative effect and induced apoptotic characteristics in human AGS gastric adenocarcinoma cells, as demonstrated by MTT assay, morphological observation method, terminal deoxynucleotidyl transferase dUTP nick end labeling and caspase-3/7 assay kits. Western blot analysis demonstrated that treatment with metformin increased the phosphorylation of AMPK, and decreased the phosphorylation of AKT, mTOR and p70S6k. Compound C (an AMPK inhibitor) suppressed AMPK phosphorylation and significantly abrogated the effects of metformin on AGS cell viability. Metformin also reduced the phosphorylation of mitogen-activated protein kinases (ERK, JNK and p38). Additionally, metformin significantly increased the cellular ROS level and included loss of mitochondrial membrane potential (ΔΨm). Metformin altered apoptosis-associated signaling to downregulate the BAD phosphorylation and Bcl-2, pro-caspase-9, pro-caspase-3 and pro-caspase-7 expression, and to upregulate BAD, cytochrome c, and Apaf-1 proteins levels in AGS cells. Furthermore, z-VAD-fmk (a pan-caspase inhibitor) was used to assess mitochondria-mediated caspase-dependent apoptosis in metformin-treated AGS cells. The findings demonstrated that metformin induced AMPK-mediated apoptosis, making it appealing for development as a novel anticancer drug for the treating gastric cancer.
Collapse
Affiliation(s)
- Chi-Cheng Lu
- Department of Sport Performance, National Taiwan University of Sport, Taichung 40404, Taiwan, R.O.C
| | - Jo-Hua Chiang
- Department of Nursing, Chung Jen Catholic Junior College, Chiayi 62241, Taiwan, R.O.C
| | - Fuu-Jen Tsai
- Human Genetics Center, Department of Medical Research, China Medical University Hospital, Taichung 40447, Taiwan, R.O.C
| | - Yuan-Man Hsu
- Department of Biological Science and Technology, China Medical University, Taichung 40402, Taiwan, R.O.C
| | - Yu-Ning Juan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40447, Taiwan, R.O.C
| | - Jai-Sing Yang
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40447, Taiwan, R.O.C
| | - Hong-Yi Chiu
- Department of Pharmacy, Buddhist Tzu Chi General Hospital, Hualien 97002, Taiwan, R.O.C
| |
Collapse
|
11
|
Heckl SM, Wiesener V, Behrens HM, Ulase D, Krüger S, Röcken C. The expression of the insulin receptor in gastric cancer correlates with the HER2 status and may have putative therapeutic implications. Gastric Cancer 2019; 22:1130-1142. [PMID: 30989432 PMCID: PMC6811372 DOI: 10.1007/s10120-019-00964-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 04/04/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Metabolic reprogramming in gastric cancer (GC) involves not only an alteration of glucose metabolism, but also of insulin receptor (IR) expression. We investigated if (1) GCs express the IR in cancer cells (CC-IR) and vasculature (VIR), (2) IR expression is clinically relevant and may be a novel target of GC treatment. METHODS 467 primary GCs were studied by immunohistochemistry using an IR-specific antibody. CD31-immunostaining ensured the presence of representative intratumoral microvasculature. VIR, and membranous and cytoplasmic CC-IR (mCC-IR, cCC-IR) were evaluated using a modified HistoScore (HScore) and subsequently dichotomized into low or high IR expressions. The IR status was correlated with clinico-pathological patient characteristics, including survival and HER2 status. RESULTS VIR, mCC-IR, and cCC-IR (HScore > 0) were found in 97.0%, 87.6%, and 95.7% of all GCs. After dichotomization of the HScores, 50.7, 48.8, and 50.3% were classified as VIR-high, mCC-IR-high, and cCC-IR-high, respectively. IR was associated with the Laurén phenotype, tumor localization, local tumor growth, vascular invasion, perineural invasion, tumor budding, mucin phenotype, UICC stage, worse survival, and the HER2 status. On multivariate analysis, VIR status was an independent prognosticator of overall (p = 0.010) and tumor-specific (p = 0.006) patient survival. CONCLUSIONS VIR and CC-IR expressions are frequent in GC, biologically significant and even correlate with the HER2 status, opening avenues for novel putative therapeutic interventions in GC.
Collapse
Affiliation(s)
- Steffen M Heckl
- Department of Pathology, Christian-Albrechts-University, Arnold-Heller-Str. 3, Haus 14, 24105, Kiel, Germany
- Department of Internal Medicine I, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Viva Wiesener
- Department of Pathology, Christian-Albrechts-University, Arnold-Heller-Str. 3, Haus 14, 24105, Kiel, Germany
| | - Hans-Michael Behrens
- Department of Pathology, Christian-Albrechts-University, Arnold-Heller-Str. 3, Haus 14, 24105, Kiel, Germany
| | - Dita Ulase
- Department of Pathology, Riga Stradins University, Riga, Latvia
| | - Sandra Krüger
- Department of Pathology, Christian-Albrechts-University, Arnold-Heller-Str. 3, Haus 14, 24105, Kiel, Germany
| | - Christoph Röcken
- Department of Pathology, Christian-Albrechts-University, Arnold-Heller-Str. 3, Haus 14, 24105, Kiel, Germany.
| |
Collapse
|
12
|
Hu Q, Li L, Zou X, Xu L, Yi P. Berberine Attenuated Proliferation, Invasion and Migration by Targeting the AMPK/HNF4α/WNT5A Pathway in Gastric Carcinoma. Front Pharmacol 2018; 9:1150. [PMID: 30405404 PMCID: PMC6202939 DOI: 10.3389/fphar.2018.01150] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 09/24/2018] [Indexed: 12/13/2022] Open
Abstract
Background: Recent epidemiologic studies have found that patients with diabetes have a higher risk of gastric cancer (GC), and the long-term use of metformin is associated with a lower risk of gastric cancer. It is believed that blocking tumor energy metabolic alterations is now emerging as a new therapeutic approach of cancer. Berberine, a natural isoquinoline alkaloid, could modulate lipid metabolism and glucose homeostasis by regulating the expression of HNF4α in many metabolic diseases. Here, we investigated the effect of Berberine on GC and its possible molecular mechanism through targeting HNF4α. Methods and Results: (1) AGS and SGC7901 gastric cancer cells were treated with Berberine (BBR). We found that in AGS and SGC7901 cell, BBR inhibited cell proliferation in a time- and dose-dependent manner through downregulating C-myc. BBR also induced G0-G1 phase arrest with the decreased expression of cyclin D1. Moreover, BBR attenuated the migration and invasion by downregulating MMP-3. (2) The lentivirus infection was used to silence the expression of HNF4α in SGC7901 cell. The results demonstrated that the knockdown of HNF4α in SGC7901 slowed cells proliferation, induced S phase arrest and dramatically attenuated gastric cancer cells’ metastasis and invasion. (3) We performed GC cells perturbation experiments through BI6015 (an HNF4α antagonist), AICAR (an AMPK activator), Compound C (AMPK-kinase inhibitor), metformin and BBR. Our findings indicated that BBR downregulated HNF4α while upregulating p-AMPK. Moreover, the inhibition of HNF4α by BBR was AMPK dependent. (4) Then the LV-HNF4α-RNAi SGC7901 cell model was used to detect the downstream of HNF4α in vitro. The results showed that the knockdown of HNF4α significantly decreased WNT5A and cytoplasmic β-catenin, but increased E-cadherin in vitro. Berberine also downregulated WNT5A and cytoplasmic β-catenin, the same as LV-HNF4α-RNAi and BI6015 in GC cells. (5) Finally, the SGC7901 and LV-HNF4α-RNAi SGC7901 mouse-xenograft model to evaluate the effect of BBR and HNF4α gene on GC tumor growth. The result illustrated that BBR and knockdown of HNF4α suppressed tumor growth in vivo, and BBR decreased HNF4α, WNT5A and cytoplasmic β-catenin levels, the same effect as HNF4α knockout in vivo. Conclusion: BBR not only had proliferation inhibition effect, attenuated the invasion and migration on GC cell lines, but also suppressed the GC tumor growth in vivo. The anti-gastric cancer mechanism of BBR might be involved in AMPK-HNF4α-WNT5A signaling pathway. HNF4α antagonists, such as BBR, could be a promising anti-gastric cancer treatment supplement.
Collapse
Affiliation(s)
- Qian Hu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lingli Li
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Zou
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lijun Xu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Yi
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
13
|
Li J, Han X, Wan Y, Zhang S, Zhao Y, Fan R, Cui Q, Zhou Y. TAM 2.0: tool for MicroRNA set analysis. Nucleic Acids Res 2018; 46:W180-W185. [PMID: 29878154 PMCID: PMC6031048 DOI: 10.1093/nar/gky509] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 05/21/2018] [Accepted: 05/23/2018] [Indexed: 12/12/2022] Open
Abstract
With the rapid accumulation of high-throughput microRNA (miRNA) expression profile, the up-to-date resource for analyzing the functional and disease associations of miRNAs is increasingly demanded. We here describe the updated server TAM 2.0 for miRNA set enrichment analysis. Through manual curation of over 9000 papers, a more than two-fold growth of reference miRNA sets has been achieved in comparison with previous TAM, which covers 9945 and 1584 newly collected miRNA-disease and miRNA-function associations, respectively. Moreover, TAM 2.0 allows users not only to test the functional and disease annotations of miRNAs by overrepresentation analysis, but also to compare the input de-regulated miRNAs with those de-regulated in other disease conditions via correlation analysis. Finally, the functions for miRNA set query and result visualization are also enabled in the TAM 2.0 server to facilitate the community. The TAM 2.0 web server is freely accessible at http://www.scse.hebut.edu.cn/tam/ or http://www.lirmed.com/tam2/.
Collapse
Affiliation(s)
- Jianwei Li
- Institute of Computational Medicine, School of Artificial Intelligence, Hebei University of Technology, Tianjin 300401, China
- Department of Biomedical Informatics, School of Basic Medical Sciences, Center for Noncoding RNA Medicine, Peking University, Beijing 100191, China
| | - Xiaofen Han
- Institute of Computational Medicine, School of Artificial Intelligence, Hebei University of Technology, Tianjin 300401, China
| | - Yanping Wan
- Institute of Computational Medicine, School of Artificial Intelligence, Hebei University of Technology, Tianjin 300401, China
| | - Shan Zhang
- Institute of Computational Medicine, School of Artificial Intelligence, Hebei University of Technology, Tianjin 300401, China
| | - Yingshu Zhao
- Institute of Computational Medicine, School of Artificial Intelligence, Hebei University of Technology, Tianjin 300401, China
| | - Rui Fan
- Institute of Computational Medicine, School of Artificial Intelligence, Hebei University of Technology, Tianjin 300401, China
| | - Qinghua Cui
- Department of Biomedical Informatics, School of Basic Medical Sciences, Center for Noncoding RNA Medicine, Peking University, Beijing 100191, China
| | - Yuan Zhou
- Department of Biomedical Informatics, School of Basic Medical Sciences, Center for Noncoding RNA Medicine, Peking University, Beijing 100191, China
| |
Collapse
|
14
|
Ashamalla M, Youssef I, Yacoub M, Jayarangaiah A, Gupta N, Ray J, Iqbal S, Miller R, Singh J, McFarlane SI. Obesity, Diabetes and Gastrointestinal Malignancy: The role of Metformin and other Anti-diabetic Therapy. GLOBAL JOURNAL OF OBESITY, DIABETES AND METABOLIC SYNDROME 2018; 5:008-14. [PMID: 30533942 PMCID: PMC6282807 DOI: 10.17352/2455-8583.000032] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The association between Diabetes and cancer has been known for decades with obesity and insulin resistance being postulated as the main underlying risk factors for both disorders. With rise of the epidemic of obesity in the USA and around the globe, there has been a rise in diabetes that is currently reaching epidemic proportions. Diabetes is known to be associated with increased risk of several types of malignancy including breast, cervical, pancreatic and colon cancer. In this review, we discuss the epidemic of obesity and its consequential epidemic of diabetes highlighting the pathophysiologic mechanisms of increased cancer in the diabetic population. We will then discuss the role of insulin therapy as well as, other antidiabetic medications, particularly metformin that has been to be associated with lower risk as well as better survival with GI malignancies based on several studies including a study that was recently published by our group.
Collapse
Affiliation(s)
- Michael Ashamalla
- Northwell Health care system, Department of Medicine, New Hyde Park, New York 11040, USA
| | - Irini Youssef
- Department of Radiation Oncology, Department of Medicine, Division of Endocrinology, SUNY-Downstate, Brooklyn, NY 11203, USA
| | - Mena Yacoub
- Northside Hospital, St. Petersburg, Florida, 33709, USA
| | - Apoorva Jayarangaiah
- Department of Internal Medicine, Wake Forest University, Baptist Health System, Winston-Salem, N.C, USA
| | - Nikita Gupta
- Department of Radiation Oncology, Department of Medicine, Division of Endocrinology, SUNY-Downstate, Brooklyn, NY 11203, USA
| | - Justina Ray
- Department of Radiation Oncology, Department of Medicine, Division of Endocrinology, SUNY-Downstate, Brooklyn, NY 11203, USA
| | - Sadat Iqbal
- Department of Radiation Oncology, Department of Medicine, Division of Endocrinology, SUNY-Downstate, Brooklyn, NY 11203, USA
| | - Regina Miller
- Department of Radiation Oncology, Department of Medicine, Division of Endocrinology, SUNY-Downstate, Brooklyn, NY 11203, USA
| | - Joie Singh
- Department of Radiation Oncology, Department of Medicine, Division of Endocrinology, SUNY-Downstate, Brooklyn, NY 11203, USA
| | - Samy I McFarlane
- Department of Radiation Oncology, Department of Medicine, Division of Endocrinology, SUNY-Downstate, Brooklyn, NY 11203, USA,Corresponding author: Samy I McFarlane Distinguished Teaching Professor and associate Dean, College of Medicine, Department of Medicine, Division of Endocrinology, Internal Medicine Residency Program Director, State University of New York, Downstate Medical Center, 450 Clarkson Avenue, Box 50, Brooklyn, NY 11203-2098, USA, Tel: 718-270-3711; Fax 718-270-6358;
| |
Collapse
|