1
|
Vázquez-Ibarra KC, Sánchez López JY, Pineda Razo TD, Cruz Lozano JR, Ortiz-Tamayo BG, Palafox-Mariscal LA, González Arreola RM, González-García JR, Ortiz-Lazareno PC. Metformin in combination with chemotherapy increases apoptosis in gastric cancer cells and counteracts senescence induced by chemotherapy. Oncol Lett 2024; 28:457. [PMID: 39114572 PMCID: PMC11304395 DOI: 10.3892/ol.2024.14590] [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: 02/11/2024] [Accepted: 06/10/2024] [Indexed: 08/10/2024] Open
Abstract
Gastric cancer (GC) is the fourth leading cause of cancer death in the world, and there is a demand for new therapeutic agents to treat GC. Metformin has been demonstrated to be an antineoplastic agent in some types of cancer; however, it has not been sufficiently valued in treating GC because the effect of metformin in combination with chemotherapy regimens has not yet been evaluated. The present study aimed to evaluate the mechanisms underlying cell death induced by metformin alone or when combined with chemotherapy. The cytogenetic characteristics of the NCI-N87 cell line were determined by fluorescence in situ hybridization (FISH). To determine viability, the cells were treated with metformin, epirubicin, cisplatin, docetaxel and 5-fluorouracil (individually and at different concentrations). Subsequently, the cells were treated with metformin alone, and in combination with the chemotherapeutic drugs and the epirubicin + cisplatin + 5-fluorouracil, docetaxel + cisplatin + 5-fluorouracil, and cisplatin + 5-fluorouracil regimens. Cell viability, proliferation and mitochondrial membrane potential (ΔΨm) were analyzed by spectrophotometry. Apoptosis, caspase activity and cell cycle progression were assessed by flow cytometry. Finally, light microscopy was used to evaluate senescence and clonogenicity. The results revealed that metformin, alone and when combined with chemotherapy, increased the proportion of apoptotic cells, promoted the loss of ΔΨm, and induced apoptosis through caspase activity in GC cells. Moreover, metformin decreased cell proliferation. In addition, metformin alone did not induce senescence and it counteracted the effects of chemotherapy-induced senescence in GC cells. Additionally, metformin, alone and when combined with chemotherapy, decreased the clonogenic capacity of NCI-N87 GC cells. In conclusion, metformin may increase the effects of chemotherapy on NCI-N87 cell death and could represent an option to improve the treatment of GC.
Collapse
Affiliation(s)
- Katia Carolina Vázquez-Ibarra
- Department of Molecular Biology and Genomics, University Center for Health Sciences, University of Guadalajara, Guadalajara, Jalisco 44340, Mexico
| | - Josefina Yoaly Sánchez López
- Genetic Division, Western Biomedical Research Center, Mexican Social Security Institute, Guadalajara, Jalisco, 44340, Mexico
| | - Tomás Daniel Pineda Razo
- Medical Oncology Service, Western National Medical Center, Mexican Social Security Institute, Guadalajara, Jalisco 44329, Mexico
| | - José Roberto Cruz Lozano
- Department of Physiology, University Center for Health Sciences, University of Guadalajara, Guadalajara, Jalisco 44340, Mexico
| | - Brenda Guadalupe Ortiz-Tamayo
- Division of Biological and Environmental Sciences, University Center of Biological and Agricultural Sciences, University of Guadalajara, Guadalajara, Jalisco 44600, Mexico
| | - Luis Arturo Palafox-Mariscal
- Department of Physiology, University Center for Health Sciences, University of Guadalajara, Guadalajara, Jalisco 44340, Mexico
| | - Rosa María González Arreola
- Department of Molecular Biology and Genomics, University Center for Health Sciences, University of Guadalajara, Guadalajara, Jalisco 44340, Mexico
| | - Juan Ramón González-García
- Genetic Division, Western Biomedical Research Center, Mexican Social Security Institute, Guadalajara, Jalisco, 44340, Mexico
| | - Pablo Cesar Ortiz-Lazareno
- Immunology Division, Western Biomedical Research Center, Mexican Social Security Institute, Guadalajara, Jalisco 44340, Mexico
| |
Collapse
|
2
|
Saha S, Bapat S, Vijayasarathi D, Vyas R. Exploring potential biomarkers and lead molecules in gastric cancer by network biology, drug repurposing and virtual screening strategies. Mol Divers 2024:10.1007/s11030-024-10995-6. [PMID: 39348085 DOI: 10.1007/s11030-024-10995-6] [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: 07/10/2024] [Accepted: 09/15/2024] [Indexed: 10/01/2024]
Abstract
Gastric cancer poses a significant global health challenge, necessitating innovative approaches for biomarker discovery and therapeutic intervention. This study employs a multifaceted strategy integrating network biology, drug repurposing, and virtual screening to elucidate and expand the molecular landscape of gastric cancer. We identified and prioritized key genes implicated in gastric cancer by utilizing data from diverse databases and text-mining techniques. Network analysis underscored intricate gene interactions, emphasizing potential therapeutic targets such as CTNNB1, BCL2, TP53, etc, and highlighted ACTB among the top hub genes crucial in disease progression. Drug repurposing on 626 FDA-approved drugs for digestive system-related cancers revealed Norgestimate and Nimesulide as likely top candidates for gastric cancer, validated by molecular docking and dynamics simulations. Further, combinatorial synthesis of scaffold libraries derived from known chemotypes generated 56,160 virtual compounds, of which 76 new compounds were prioritized based on promising binding affinities and interactions at critical residues. Hotspot residue analysis identified GLU 214 and others as essential for ligand binding stability, enhancing compound efficacy and specificity. These findings support the therapeutic potential of targeting beta-actin protein in gastric cancer treatment, suggesting a future for further experimental validation and clinical translation. In conclusion, this study highlights the potential of repurposable drugs and virtual screening which can be used in combination with existing anti-gastric cancer drugs for gastric cancer therapy, emphasizing the role of computational methodologies in drug discovery.
Collapse
Affiliation(s)
- Sagarika Saha
- MIT ADTU School of Bioengineering Sciences & Research, MIT Art, Design and Technology University, Pune, Maharashtra, India
| | - Sanket Bapat
- MIT ADTU School of Bioengineering Sciences & Research, MIT Art, Design and Technology University, Pune, Maharashtra, India
| | - Durairaj Vijayasarathi
- MIT ADTU School of Bioengineering Sciences & Research, MIT Art, Design and Technology University, Pune, Maharashtra, India
| | - Renu Vyas
- MIT ADTU School of Bioengineering Sciences & Research, MIT Art, Design and Technology University, Pune, Maharashtra, India.
| |
Collapse
|
3
|
De la Torre K, Song M, Abe SK, Rahman MS, Islam MR, Saito E, Min S, Huang D, Chen Y, Gupta PC, Sawada N, Tamakoshi A, Shu X, Wen W, Sakata R, Kim J, Nagata C, Ito H, Park SK, Shin M, Pednekar MS, Tsugane S, Kimura T, Gao Y, Cai H, Wada K, Oze I, Shin A, Ahn Y, Ahsan H, Boffetta P, Chia KS, Matsuo K, Qiao Y, Rothman N, Zheng W, Inoue M, Kang D. Diabetes and gastric cancer incidence and mortality in the Asia Cohort Consortium: A pooled analysis of more than a half million participants. J Diabetes 2024; 16:e13561. [PMID: 38751364 PMCID: PMC11096812 DOI: 10.1111/1753-0407.13561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/19/2024] [Accepted: 03/14/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND Evidence suggests a possible link between diabetes and gastric cancer risk, but the findings remain inconclusive, with limited studies in the Asian population. We aimed to assess the impact of diabetes and diabetes duration on the development of gastric cancer overall, by anatomical and histological subtypes. METHODS A pooled analysis was conducted using 12 prospective studies included in the Asia Cohort Consortium. Among 558 981 participants (median age 52), after a median follow-up of 14.9 years and 10.5 years, 8556 incident primary gastric cancers and 8058 gastric cancer deaths occurred, respectively. Cox proportional hazard regression models were used to estimate study-specific hazard ratios (HRs) and 95% confidence intervals (CIs) and pooled using random-effects meta-analyses. RESULTS Diabetes was associated with an increased incidence of overall gastric cancer (HR 1.15, 95% CI 1.06-1.25). The risk association did not differ significantly by sex (women vs men: HR 1.31, 95% CI 1.07-1.60 vs 1.12, 1.01-1.23), anatomical subsites (noncardia vs cardia: 1.14, 1.02-1.28 vs 1.17, 0.77-1.78) and histological subtypes (intestinal vs diffuse: 1.22, 1.02-1.46 vs 1.00, 0.62-1.61). Gastric cancer risk increased significantly during the first decade following diabetes diagnosis (HR 4.70, 95% CI 3.77-5.86), and decreased with time (nonlinear p < .01). Positive associations between diabetes and gastric cancer mortality were observed (HR 1.15, 95% CI 1.03-1.28) but attenuated after a 2-year time lag. CONCLUSION Diabetes was associated with an increased gastric cancer incidence regardless of sex, anatomical subsite, or subtypes of gastric cancer. The risk of gastric cancer was particularly high during the first decade following diabetes diagnosis.
Collapse
Grants
- R37 CA070867 NCI NIH HHS
- 30-A-15,23-A-31(toku),26-A-2,29-A-4 National Cancer Center Japan Research and Development Fund
- 24H1080 National Cancer Center Korea Research Grant
- 0520160-1 National R&D Program for Cancer Control, Ministry of Health & Welfare, Republic of Korea
- 23-A-31 Ministry of Health, Labour and Welfare of Japan
- NRF-2016R1A2B4014552 National Research Foundation of Korea
- The Japanese Ministry of the Environment
- Ministry of Education, Culture, Sports, Science and Technology, Japan
- International Agency for Research on Cancer, Lyon, France
- 2210990 National Cancer Center Korea Research Grant
- US Department of Energy
- UM1 CA173640 NCI NIH HHS
- UM1 CA182910 NCI NIH HHS
- Ministry of Education, Culture, Sports, Science and Technology, Japan
- Ministry of Health, Labour and Welfare of Japan
- National Research Foundation of Korea
- Division of Cancer Prevention, National Cancer Institute
- The Japanese Ministry of the Environment
- International Agency for Research on Cancer, Lyon, France
- US Department of Energy
Collapse
Affiliation(s)
- Katherine De la Torre
- Department of Biomedical SciencesSeoul National University Graduate SchoolSeoulKorea
- Department of Preventive MedicineSeoul National University College of MedicineSeoulKorea
| | - Minkyo Song
- Infections and Immunoepidemiology Branch, Division of Cancer Epidemiology and GeneticsNational Cancer InstituteBethesdaMarylandUSA
- Laboratory of Epidemiology and Population SciencesNational Institute on Aging, National Institute of HealthBaltimoreMarylandUSA
| | - Sarah Krull Abe
- Division of PreventionNational Cancer Center Institute for Cancer ControlTokyoJapan
| | - Md. Shafiur Rahman
- Division of PreventionNational Cancer Center Institute for Cancer ControlTokyoJapan
- Research Center for Child Mental Development, Hamamatsu University School of MedicineTokyoJapan
| | - Md. Rashedul Islam
- Division of PreventionNational Cancer Center Institute for Cancer ControlTokyoJapan
- Hitotsubashi Institute for Advanced Study, Hitotsubashi UniversityTokyoJapan
| | - Eiko Saito
- Institute for Global Health Policy Research, National Center for Global Health and MedicineTokyoJapan
| | - Sukhong Min
- Department of Preventive MedicineSeoul National University College of MedicineSeoulKorea
| | - Dan Huang
- Department of Preventive MedicineSeoul National University College of MedicineSeoulKorea
- Integrated Major in Innovative Medical Science, Seoul National University Graduate SchoolSeoulKorea
| | - Yu Chen
- Department of Population Health and Environmental MedicineNYU Grossman School of MedicineNew YorkNew YorkUSA
| | | | - Norie Sawada
- Division of Cohort ResearchNational Cancer Center Institute for Cancer ControlTokyoJapan
| | - Akiko Tamakoshi
- Department of Public HealthHokkaido University Faculty of MedicineSapporoJapan
| | - Xiao‐Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center Vanderbilt‐Ingram Cancer CenterVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Wanqing Wen
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center Vanderbilt‐Ingram Cancer CenterVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Ritsu Sakata
- Radiation Effects Research FoundationHiroshimaJapan
| | - Jeongseon Kim
- Graduate School of Cancer Science and Policy, National Cancer CenterGoyangKorea
| | - Chisato Nagata
- Department of Epidemiology and Preventive MedicineGifu University Graduate School of MedicineGifuJapan
| | - Hidemi Ito
- Department of Preventive Medicine, Division of Cancer Information and ControlAichi Cancer Center Research InstituteNagoyaJapan
- Division of Descriptive Cancer EpidemiologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Sue K. Park
- Department of Preventive MedicineSeoul National University College of MedicineSeoulKorea
| | - Myung‐Hee Shin
- Department of Social and Preventive MedicineSungkyunkwan University School of MedicineSeoulKorea
| | | | - Shoichiro Tsugane
- Division of Cohort ResearchNational Cancer Center Institute for Cancer ControlTokyoJapan
- International University of Health and Welfare Graduate SchoolTokyoJapan
| | - Takashi Kimura
- Department of Public HealthHokkaido University Faculty of MedicineSapporoJapan
| | - Yu‐Tang Gao
- Department of EpidemiologyShanghai Cancer InstituteShanghaiChina
- Renji Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Hui Cai
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center Vanderbilt‐Ingram Cancer CenterVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Keiko Wada
- Department of Epidemiology and Preventive MedicineGifu University Graduate School of MedicineGifuJapan
| | - Isao Oze
- Division of Cancer Epidemiology and PreventionAichi Cancer Center Research InstituteNagoyaJapan
| | - Aesun Shin
- Department of Preventive MedicineSeoul National University College of MedicineSeoulKorea
- Cancer Research Institute, Seoul National UniversitySeoulKorea
| | - Yoon‐Ok Ahn
- Department of Preventive MedicineSeoul National University College of MedicineSeoulKorea
| | - Habibul Ahsan
- Department of Public Health SciencesUniversity of ChicagoChicagoIllinoisUSA
| | - Paolo Boffetta
- Stony Brook Cancer Center, Stony Brook UniversityStony BrookNew YorkUSA
- Department of Medical and Surgical SciencesUniversity of BolognaBolognaItaly
| | - Kee Seng Chia
- Saw Swee Hock School of Public Health, National University of SingaporeSingaporeSingapore
| | - Keitaro Matsuo
- Division of Cancer Epidemiology and PreventionAichi Cancer Center Research InstituteNagoyaJapan
- Department of Cancer EpidemiologyNagoya University Graduate School of Medicine NagoyaNagoyaJapan
| | - You‐Lin Qiao
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Nathaniel Rothman
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and GeneticsNational Cancer InstituteBethesdaMarylandUSA
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center Vanderbilt‐Ingram Cancer CenterVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Manami Inoue
- Division of PreventionNational Cancer Center Institute for Cancer ControlTokyoJapan
| | - Daehee Kang
- Department of Preventive MedicineSeoul National University College of MedicineSeoulKorea
- Integrated Major in Innovative Medical Science, Seoul National University Graduate SchoolSeoulKorea
| |
Collapse
|
4
|
Galal MA, Al-Rimawi M, Hajeer A, Dahman H, Alouch S, Aljada A. Metformin: A Dual-Role Player in Cancer Treatment and Prevention. Int J Mol Sci 2024; 25:4083. [PMID: 38612893 PMCID: PMC11012626 DOI: 10.3390/ijms25074083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 03/30/2024] [Accepted: 04/02/2024] [Indexed: 04/14/2024] Open
Abstract
Cancer continues to pose a significant global health challenge, as evidenced by the increasing incidence rates and high mortality rates, despite the advancements made in chemotherapy. The emergence of chemoresistance further complicates the effectiveness of treatment. However, there is growing interest in the potential of metformin, a commonly prescribed drug for type 2 diabetes mellitus (T2DM), as an adjuvant chemotherapy agent in cancer treatment. Although the precise mechanism of action of metformin in cancer therapy is not fully understood, it has been found to have pleiotropic effects, including the modulation of metabolic pathways, reduction in inflammation, and the regulation of cellular proliferation. This comprehensive review examines the anticancer properties of metformin, drawing insights from various studies conducted in vitro and in vivo, as well as from clinical trials and observational research. This review discusses the mechanisms of action involving both insulin-dependent and independent pathways, shedding light on the potential of metformin as a therapeutic agent for different types of cancer. Despite promising findings, there are challenges that need to be addressed, such as conflicting outcomes in clinical trials, considerations regarding dosing, and the development of resistance. These challenges highlight the importance of further research to fully harness the therapeutic potential of metformin in cancer treatment. The aims of this review are to provide a contemporary understanding of the role of metformin in cancer therapy and identify areas for future exploration in the pursuit of effective anticancer strategies.
Collapse
Affiliation(s)
- Mariam Ahmed Galal
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia; (M.A.G.); (M.A.-R.); (H.D.); (S.A.)
- Department of Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 1QU, UK
| | - Mohammed Al-Rimawi
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia; (M.A.G.); (M.A.-R.); (H.D.); (S.A.)
| | | | - Huda Dahman
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia; (M.A.G.); (M.A.-R.); (H.D.); (S.A.)
| | - Samhar Alouch
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia; (M.A.G.); (M.A.-R.); (H.D.); (S.A.)
| | - Ahmad Aljada
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia; (M.A.G.); (M.A.-R.); (H.D.); (S.A.)
| |
Collapse
|
5
|
Pillai U J, Ray A, Maan M, Dutta M. Repurposing drugs targeting metabolic diseases for cancer therapeutics. Drug Discov Today 2023; 28:103684. [PMID: 37379903 DOI: 10.1016/j.drudis.2023.103684] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 06/01/2023] [Accepted: 06/18/2023] [Indexed: 06/30/2023]
Abstract
Hurdles in the identification of new drugs for cancer treatment have made drug repurposing an increasingly appealing alternative. The approach involves the use of old drugs for new therapeutic purposes. It is cost-effective and facilitates rapid clinical translation. Given that cancer is also considered a metabolic disease, drugs for metabolic disorders are being actively repurposed for cancer therapeutics. In this review, we discuss the repurposing of such drugs approved for two major metabolic diseases, diabetes and cardiovascular disease (CVD), which have shown potential as anti-cancer treatment. We also highlight the current understanding of the cancer signaling pathways that these drugs target.
Collapse
Affiliation(s)
- Jisha Pillai U
- Department of Biotechnology, BITS Pilani, Dubai Campus, Academic City, Dubai, UAE
| | - Anindita Ray
- Department of Biotechnology, BITS Pilani, Dubai Campus, Academic City, Dubai, UAE
| | - Meenu Maan
- Mohammed Bin Rashid University of Medicine and Health Sciences (MBRU), Dubai, UAE; New York University-Abu Dhabi, Abu Dhabi, UAE.
| | - Mainak Dutta
- Department of Biotechnology, BITS Pilani, Dubai Campus, Academic City, Dubai, UAE.
| |
Collapse
|
6
|
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: 1] [Impact Index Per Article: 1.0] [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
|
7
|
Araújo D, Ribeiro E, Amorim I, Vale N. Repurposed Drugs in Gastric Cancer. MOLECULES (BASEL, SWITZERLAND) 2022; 28:molecules28010319. [PMID: 36615513 PMCID: PMC9822219 DOI: 10.3390/molecules28010319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 12/21/2022] [Accepted: 12/25/2022] [Indexed: 01/04/2023]
Abstract
Gastric cancer (GC) is one of the major causes of death worldwide, ranking as the fifth most incident cancer in 2020 and the fourth leading cause of cancer mortality. The majority of GC patients are in an advanced stage at the time of diagnosis, presenting a poor prognosis and outcome. Current GC treatment approaches involve endoscopic detection, gastrectomy and chemotherapy or chemoradiotherapy in an adjuvant or neoadjuvant setting. Drug development approaches demand extreme effort to identify molecular mechanisms of action of new drug candidates. Drug repurposing is based on the research of new therapeutic indications of drugs approved for other pathologies. In this review, we explore GC and the different drugs repurposed for this disease.
Collapse
Affiliation(s)
- Diana Araújo
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- Institute for Research and Innovation in Health (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Rua Júlio Amaral de Carvalho, 45, 4200-135 Porto, Portugal
| | - Eduarda Ribeiro
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Irina Amorim
- Institute of Biomedical Sciences Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- Institute for Research and Innovation in Health (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Rua Júlio Amaral de Carvalho, 45, 4200-135 Porto, Portugal
| | - Nuno Vale
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- Correspondence: ; Tel.: +351-220426537
| |
Collapse
|
8
|
Zhou W, Li W, He C, Ma R, Gao Q, Wang Y, Feng L, Liu L. Influence of Hyperglycemia on the Prognosis of Patients with Diffuse Large B-Cell Lymphoma. Diabetes Metab Syndr Obes 2022; 15:2039-2049. [PMID: 35860311 PMCID: PMC9289755 DOI: 10.2147/dmso.s370017] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 07/06/2022] [Indexed: 11/23/2022] Open
Abstract
PURPOSE To explore the effects of primary and secondary hyperglycemia and the application of the hypoglycemic drug metformin on the prognosis of patients with diffuse large B-cell lymphoma (DLBCL). METHODS We performed a retrospective analysis of 1767 DLBCL patients.Cox regression method was used for analysis to evaluate the prognostic factors, and the Kaplan-Meier method was used to draw a survival curve to analyze the effect of hyperglycemia and the hypoglycemic drug metformin on the progression-free survival (PFS) and overall survival (OS) of DLBCL patients. RESULTS Our study showed that patients with hyperglycemia tend to have higher age (age>60 years), high body mass index (BMI)(≥24kg/m2), late Ann Arbor stage (III-IV), high international prognostic index (IPI) (3-5 score), high lactic dehydrogenase (LDH) level (>250U/L), bulky disease and comorbidity. Hyperglycemia affects the survival time of the DLBCL population (PFS: adjusted HR 1.41, 95% CI: 1.16-1.70, P <0.001, OS: adjusted HR 1.33, 95% CI:1.09-1.61, P=0.004).Compared with the non-hyperglycemia group, the secondary hyperglycemia increase affects the prognosis of the DLBCL population (P<0.001). Compared with the secondary hyperglycemia group, the primary hyperglycemia group has a poor prognosis (P<0.05). For patients with DLBCL and hyperglycemia (732 patients in total), the use of metformin can improve their PFS and OS (PFS: adjusted HR 0.69, 95% CI: 0.49-0.96, P=0.028, OS: adjusted HR 0.68, 95% CI: 0.49-0.95, P=0.024). CONCLUSION Hyperglycemia and secondary hyperglycemia are related to the poor prognosis of DLBCL population.For patients with DLBCL combined with hyperglycemia, the application of metformin can improve survival rate.
Collapse
Affiliation(s)
- Weiling Zhou
- Department of Endocrine and Metabolic Diseases, The Fourth Hospital of Hebei Medical University (Hebei Tumor Hospital), Shijiazhuang, People’s Republic of China
| | - Weijing Li
- Department of Hematology, The Fourth Hospital of Hebei Medical University (Hebei Tumor Hospital), Shijiazhuang, People’s Republic of China
| | - Cuiying He
- Department of Hematology, The Fourth Hospital of Hebei Medical University (Hebei Tumor Hospital), Shijiazhuang, People’s Republic of China
| | - Ruijuan Ma
- Department of Hematology, The Fourth Hospital of Hebei Medical University (Hebei Tumor Hospital), Shijiazhuang, People’s Republic of China
| | - Qian Gao
- Department of Endocrine and Metabolic Diseases, The Fourth Hospital of Hebei Medical University (Hebei Tumor Hospital), Shijiazhuang, People’s Republic of China
| | - Yuan Wang
- Department of Endocrine and Metabolic Diseases, The Fourth Hospital of Hebei Medical University (Hebei Tumor Hospital), Shijiazhuang, People’s Republic of China
| | - Lei Feng
- Department of Endocrine and Metabolic Diseases, The Fourth Hospital of Hebei Medical University (Hebei Tumor Hospital), Shijiazhuang, People’s Republic of China
| | - Lihong Liu
- Department of Hematology, The Fourth Hospital of Hebei Medical University (Hebei Tumor Hospital), Shijiazhuang, People’s Republic of China
- Correspondence: Lihong Liu, Department of Hematology, The Fourth Hospital of Hebei Medical University (Hebei Tumor Hospital), Shijiazhuang, People’s Republic of China, Tel +86 13831177920, Email
| |
Collapse
|
9
|
Tran TT, Lee J, Gunathilake M, Cho H, Kim J. Influence of Fasting Glucose Level on Gastric Cancer Incidence in a Prospective Cohort Study. Cancer Epidemiol Biomarkers Prev 2021; 31:254-261. [PMID: 34758969 DOI: 10.1158/1055-9965.epi-21-0670] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/11/2021] [Accepted: 10/29/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND High fasting glucose has been indicated in relation to a higher risk of gastric cancer, but the majority of studies have focused on diabetes (fasting glucose ≥126 mg/dL). Here, we investigated whether fasting glucose levels, including prediabetic and diabetic levels, influence gastric cancer incidence. METHODS A prospective study was conducted with 41,837 participants aged 16 and older who underwent health examinations at the National Cancer Center in South Korea from August 2002 to December 2014. Participants were followed up until December 2017 to identify incident gastric cancer cases. A fasting glucose test was performed based on venous blood samples taken from participants after 8 hours of fasting. We used the Cox proportional hazards regression model to explore the association of fasting glucose levels with gastric cancer incidence. RESULTS We identified 263 incident gastric cancer cases during the follow-up period. A significant association of high fasting glucose with gastric cancer incidence was found for postmenopausal women [hazard ratio (HR) = 1.88; 95% confidence interval (CI) = 1.11-3.20]. There was also a significant association between high fasting glucose and gastric cancer incidence among all participants who were nonsmokers (HR = 1.89; 95% CI = 1.21-2.95), had a BMI < 25 kg/m2 (HR = 1.45; 95% CI = 1.00-2.12), and did not have a first-degree family history of gastric cancer (HR = 1.45; 95% CI = 1.06-1.99). CONCLUSIONS Our findings support that high fasting glucose is a risk factor for gastric cancer development in postmenopausal women. IMPACT Our results provide evidence for future planning and management regarding cancer prevention.
Collapse
Affiliation(s)
- Tao Thi Tran
- Department of Cancer Control and Population Health, Graduate School of Cancer Science and Policy, Goyang-si, Gyeonggi-do, Korea
| | - Jeonghee Lee
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, Goyang-si, Gyeonggi-do, Korea
| | - Madhawa Gunathilake
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, Goyang-si, Gyeonggi-do, Korea
| | - Hyunsoon Cho
- Department of Cancer Control and Population Health, Graduate School of Cancer Science and Policy, Goyang-si, Gyeonggi-do, Korea
| | - Jeongseon Kim
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, Goyang-si, Gyeonggi-do, Korea.
| |
Collapse
|
10
|
de Villiers EM, zur Hausen H. Bovine Meat and Milk Factors (BMMFs): Their Proposed Role in Common Human Cancers and Type 2 Diabetes Mellitus. Cancers (Basel) 2021; 13:cancers13215407. [PMID: 34771570 PMCID: PMC8582480 DOI: 10.3390/cancers13215407] [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: 09/23/2021] [Revised: 10/24/2021] [Accepted: 10/25/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary This manuscript emphasizes the mechanistic differences of infectious agents contributing to human cancers either by “direct” or “indirect” interactions. The epidemiology of cancers linked to direct carcinogens differs (e.g., response to immunosuppression) from those cancers linked with indirect infectious interactions. We discuss their role in colon, breast, and prostate cancers and type II diabetes mellitus. A brief discussion covers the potential role of BMMF (bovine meat and milk factor) infections in acute myeloid leukemia. Abstract Exemplified by infections with bovine meat and milk factors (BMMFs), this manuscript emphasizes the different mechanistic aspects of infectious agents contributing to human cancers by “direct” or “indirect” interactions. The epidemiology of cancers linked to direct carcinogens (e.g., response to immunosuppression) differs from those cancers linked with indirect infectious interactions. Cancers induced by direct infectious carcinogens commonly increase under immunosuppression, whereas the cancer risk by indirect carcinogens is reduced. This influences their responses to preventive and therapeutic interferences. In addition, we discuss their role in colon, breast and prostate cancers and type II diabetes mellitus. A brief discussion covers the potential role of BMMF infections in acute myeloid leukemia.
Collapse
Affiliation(s)
- Ethel-Michele de Villiers
- Correspondence: (E.-M.d.V.); (H.z.H.); Tel.: +49-151-4312-3085 (E.-M.d.V.); +49-6221-423850 (H.z.H.)
| | - Harald zur Hausen
- Correspondence: (E.-M.d.V.); (H.z.H.); Tel.: +49-151-4312-3085 (E.-M.d.V.); +49-6221-423850 (H.z.H.)
| |
Collapse
|
11
|
Liu J, Knani I, Gross-Cohen M, Hu J, Wang S, Tang L, Ilan N, Yang S, Vlodavsky I. Role of heparanase 2 (Hpa2) in gastric cancer. Neoplasia 2021; 23:966-978. [PMID: 34343822 PMCID: PMC8349917 DOI: 10.1016/j.neo.2021.07.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 07/18/2021] [Accepted: 07/19/2021] [Indexed: 12/27/2022] Open
Abstract
We report that gastric cancer patients exhibiting high levels of heparanase 2 (Hpa2) survive longer. Similarly, mice administrated with gastric carcinoma cells engineered to overexpress Hpa2 produced smaller tumors and survived longer than mice administrated with control cells. These beneficial effects were found to associate with increased phosphorylation of AMP-activated protein kinase (AMPK) that play an instrumental role in cell metabolism and is situated at the center of a tumor suppressor network. We also found that MG132, an inhibitor of the proteasome that results in proteotoxic stress, prominently enhances Hpa2 expression. Notably, Hpa2 induction by MG132 appeared to be mediated by AMPK, thus establishing a loop that feeds itself where Hpa2 enhances AMPK phosphorylation that, in turn, induces Hpa2 expression, possibly leading to attenuation of gastric tumorigenesis.
Heparanase is highly implicated in tumor metastasis due to its capacity to cleave heparan sulfate and, consequently, remodel the extracellular matrix underlying epithelial and endothelial cells. In striking contrast, only little attention was given to its close homolog, heparanase 2 (Hpa2), possibly because it lacks heparan sulfate-degrading activity typical of heparanase. We subjected sections of gastric carcinoma to immunostaining and correlated Hpa2 immunoreactivity with clinical records, including tumor grade, stage and patients' status. We over-expressed Hpa2 in gastric carcinoma cell lines and examined their tumorigenic properties in vitro and in vivo. We also evaluated the expression of Hpa2 by gastric carcinoma cells following inhibition of the proteasome, leading to proteotoxic stress, and the resulting signaling responsible for Hpa2 gene regulation. Here, we report that gastric cancer patients exhibiting high levels of Hpa2 survive longer. Similarly, mice administrated with gastric carcinoma cells engineered to over-express Hpa2 produced smaller tumors and survived longer than mice administrated with control cells. This was associated with increased phosphorylation of AMP-activated protein kinase (AMPK), a kinase that is situated at the center of a tumor suppressor network. We also found that MG132, an inhibitor of the proteasome that results in proteotoxic stress, prominently enhances Hpa2 expression. Notably, Hpa2 induction by MG132 appeared to be mediated by AMPK, and AMPK was found to induce the expression of Hpa2, thus establishing a loop that feeds itself where Hpa2 enhances AMPK phosphorylation that, in turn, induces Hpa2 expression, leading to attenuation of gastric tumorigenesis. These results indicate that high levels of Hpa2 in some tumors are due to stress conditions that tumors often experience due to their high rates of cell proliferation and high metabolic demands. This increase in Hpa2 levels by the stressed tumors appears critically important for patient outcomes.
Collapse
Affiliation(s)
- Jingjing Liu
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Ibrahim Knani
- Rappaport Faculty of Medicine, Technion Integrated Cancer Center, Technion, Haifa, Israel
| | - Miriam Gross-Cohen
- Rappaport Faculty of Medicine, Technion Integrated Cancer Center, Technion, Haifa, Israel
| | - Jiaxi Hu
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Sumin Wang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Li Tang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Neta Ilan
- Rappaport Faculty of Medicine, Technion Integrated Cancer Center, Technion, Haifa, Israel
| | - Shiming Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Israel Vlodavsky
- Rappaport Faculty of Medicine, Technion Integrated Cancer Center, Technion, Haifa, Israel.
| |
Collapse
|
12
|
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:biom11071022. [PMID: 34356646 PMCID: PMC8301937 DOI: 10.3390/biom11071022] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [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
|
13
|
Abstract
Gastric cancer is the fifth most common cause of cancer and the third leading cause of cancer-related deaths globally. The number of gastric cancer-related deaths is only projected to increase, attributable primarily to the expanding aging population. Prevention is a mainstay of gastric cancer control programs, particularly in the absence of accurate, noninvasive modalities for screening and early detection, and the absence of an infrastructure for this purpose in the majority of countries worldwide. Herein, we discuss the evidence for several chemopreventive agents, along with putative mechanisms. There remains a clear, unmet need for primary chemoprevention trials for gastric cancer.
Collapse
Affiliation(s)
- Shailja C. Shah
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, Vanderbilt University Medical Center, 1030C MRB IV, 2215 Garland Avenue, Nashville, TN 37232-0252, USA;,Veterans Affairs Tennessee Valley Health System, Nashville Campus, Nashville, TN, USA,Corresponding author:
| | - Richard M. Peek
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, Vanderbilt University Medical Center, 1030C MRB IV, 2215 Garland Avenue, Nashville, TN 37232-0252, USA
| |
Collapse
|