1
|
Rao H, An X, Qu X, Yu J, Xie J, Ke J, Liu Z, You L, Qiu Z, Tian L, Du W, Li W, Jia J, Liu D, Li S. SGLT2i delays c-Myc-induced HCC progression via targeting mTOR. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167805. [PMID: 40113049 DOI: 10.1016/j.bbadis.2025.167805] [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: 11/27/2024] [Revised: 02/23/2025] [Accepted: 03/15/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) stands as a primary malignant liver tumor characterized by metabolic reprogramming. The oncogene c-Myc exerts substantial influence by driving the transcription of numerous genes. Empagliflozin (EMPA), a sodium-glucose cotransporter-2 inhibitor (SGLT2i), is widely used in the treatment of type 2 diabetes and has recently attracted attention for its potential anti-cancer effects. This study aims to unravel the complex interplay among c-Myc, EMPA, and the mammalian target of rapamycin (mTOR) in HCC development and progression. METHODS HCC induction in mice utilized high-pressure hydrodynamic transfection of the c-Myc plasmid. QPCR and immunohistochemistry experiments were performed to detect the expression of SGLT2 in HCC tissues. In vivo experiments were conducted to corroborate the upregulation of SGLT2 following c-Myc transfection. In invo and vitro investigations were conducted to evaluate the anti-cancer effects of two SGLT2i: EMPA and canagliflozin (CANA). Network pharmacology, molecular docking analyses, CETSA experiments, and additional western blot experiments were used to reveal EMPA's interaction inhibition with mTOR. RESULTS The study identified an increase in SGLT2 expression in HCC tissues as a result of c-Myc overexpression. In vitro experiments confirmed the upregulation of SGLT2 following c-Myc transfection. Notably, the administration of SGLT2i effectively curtailed liver cancer progression, and reduced hepatic fat accumulation in mice. EMPA exhibited significant suppression of cell proliferation in c-Myc-transfected cells. In vitro experiments unveiled EMPA's interaction and with inhibition the activation of mTOR. CONCLUSION Our study highlights EMPA's potential as a therapeutic agent in delaying the development and progression of HCC.
Collapse
Affiliation(s)
- Huiling Rao
- School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, People's Republic of China; Department of Medical Engineering, The First Affiliated Hospital of Army Medical University, Chongqing 400000, People's Republic of China
| | - Xiaotong An
- School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, People's Republic of China
| | - Xinyang Qu
- School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, People's Republic of China
| | - Juan Yu
- School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, People's Republic of China; Department of Anesthesiology, People's Hospital of Yunxi County of Hubei Province, Yunxi 442600, People's Republic of China
| | - Jin Xie
- School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, People's Republic of China
| | - Jing Ke
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan 442000, People's Republic of China
| | - Zhixin Liu
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan 442000, People's Republic of China
| | - Lei You
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan 442000, People's Republic of China
| | - Zhenpeng Qiu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
| | - Lin Tian
- Department of Pathology, Renming Hospital, Hubei University of Medicine, Shiyan 442000, People's Republic of China
| | - Weixing Du
- Department of Pathology, Renming Hospital, Hubei University of Medicine, Shiyan 442000, People's Republic of China
| | - Wanrong Li
- School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, People's Republic of China
| | - Jie Jia
- School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, People's Republic of China; Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan 442000, People's Republic of China.
| | - Danwen Liu
- School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, People's Republic of China; Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan 442000, People's Republic of China.
| | - Shan Li
- School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, People's Republic of China; Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan 442000, People's Republic of China.
| |
Collapse
|
2
|
Wang Y, Wang Z, Guo X, Tao Z, Wu C, Jiang M, Pu J. Empagliflozin attenuates DOX-induced cardiotoxicity by inhibiting RIPK1-mediated endoplasmic reticulum stress and autophagy. Biochim Biophys Acta Mol Basis Dis 2025:167898. [PMID: 40350043 DOI: 10.1016/j.bbadis.2025.167898] [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: 01/23/2025] [Revised: 05/03/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND Doxorubicin (DOX), a classical chemotherapeutic agent, remains indispensable in cancer treatment but is limited by dose-dependent cardiotoxicity. Investigating strategies to mitigate DOX-induced cardiac damage is critical. Empagliflozin, a sodium-glucose co-transporter 2 (SGLT2) inhibitor, exhibits anti-inflammatory and antioxidant effects in cardiovascular disease. This study investigated empagliflozin's protective effects against DOX-induced cardiotoxicity and underlying mechanisms. METHODS DOX-induced cardiotoxicity models were established in male C57BL/6 J mice, with cardiac-specific RIPK1 overexpression achieved via adeno-associated virus (AAV9) technology. Cardiac function was assessed using echocardiography, and heart tissue was analyzed for injury, inflammation, oxidative stress, endoplasmic reticulum (ER) stress, and autophagy through various biochemical and molecular assays. RESULTS Empagliflozin alleviated DOX-induced cardiac dysfunction, reduced fibrosis, and suppressed systemic inflammation and oxidative stress in mice. Mechanistic studies revealed that empagliflozin mitigated DOX-induced cardiotoxicity by inhibiting ER stress and autophagy, as evidenced by the downregulation of BIP, p-IRE1, and ATF6 expression, alongside elevated p62 and reduced LC3BII/LC3BI levels. RIPK1 was identified as a crucial mediator of empagliflozin's cardioprotective effects, with similar protection observed using the RIPK1 inhibitor Nec-1. RIPK1 knockdown in cardiomyocytes mimicked empagliflozin's antioxidant effects, while its protective effects were abolished in RIPK1-deficient cells. Importantly, RIPK1 overexpression reduced empagliflozin's benefits in DOX-treated mice. In addition, empagliflozin enhanced DOX-induced cytotoxicity in 4 T1 breast cancer cells. CONCLUSION Empagliflozin protects against DOX-induced cardiotoxicity by attenuating inflammation, oxidative stress, ER stress and autophagy, primarily through RIPK1 inhibition, providing insights into its cardioprotective mechanisms. Additionally, empagliflozin enhances DOX-induced cytotoxicity in vitro, providing support for its combination with DOX in cancer therapy.
Collapse
Affiliation(s)
- Yinan Wang
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China
| | - Zi Wang
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China
| | - Xinning Guo
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China
| | - Zhengyu Tao
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China
| | - Chen Wu
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China
| | - Meng Jiang
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China.
| | - Jun Pu
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China.
| |
Collapse
|
3
|
Cuttone A, Cannavò V, Abdullah RMS, Fugazzotto P, Arena G, Brancati S, Muscarà A, Morace C, Quartarone C, Ruggeri D, Squadrito G, Russo GT. Expanding the Use of SGLT2 Inhibitors in T2D Patients Across Clinical Settings. Cells 2025; 14:668. [PMID: 40358192 PMCID: PMC12071329 DOI: 10.3390/cells14090668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 04/30/2025] [Accepted: 05/01/2025] [Indexed: 05/15/2025] Open
Abstract
Sodium-glucose cotransporter-2 inhibitors (SGLT2i) are currently recommended in patients with type 2 diabetes (T2D) to reduce serum glucose levels. Moreover, robust evidence has clearly demonstrated their beneficial cardiovascular and renal effects, making this class of drugs pivotal for the treatment of T2D, especially when complicated by diabetic kidney disease or heart failure. However, several other comorbidities are frequently encountered in T2D patients beyond these long-term diabetes complications, especially in the internal medicine setting. For some of these comorbidities, such as MAFLD and cognitive impairment, the association with diabetes is increasingly recognized, with the hypothesis of a common pathophysiologic background, whereas, for others, a coincident epidemiology linked to the ageing of populations, including that of T2D subjects, may be advocated. In the effort of personalizing T2D treatment, evidence on the potential effects of SGLT2i in these different clinical conditions is accumulating. The purpose of this narrative review is to update current literature on the effects of SGLT2i for the treatment of T2D in different clinical settings beyond glycaemic control, and to elucidate potential molecular mechanisms by which they exert these effects.
Collapse
Affiliation(s)
- Alessandro Cuttone
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| | - Vittorio Cannavò
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| | - Raouf Mastan Sheik Abdullah
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| | - Pierluigi Fugazzotto
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| | - Giada Arena
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| | - Simona Brancati
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| | - Andrea Muscarà
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| | - Carmela Morace
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| | - Cristina Quartarone
- Internal Medicine and Diabetology Unit, University Hospital of Messina, 98124 Messina, Italy; (C.Q.); (D.R.)
| | - Domenica Ruggeri
- Internal Medicine and Diabetology Unit, University Hospital of Messina, 98124 Messina, Italy; (C.Q.); (D.R.)
| | - Giovanni Squadrito
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| | - Giuseppina Tiziana Russo
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| |
Collapse
|
4
|
Xu B, Kang B, Li S, Chen J, Zhou J. Association between sodium-glucose cotransporter 2 inhibitors and cancer: a systematic review and meta-analysis of randomized active-controlled trials. Int J Clin Pharm 2025:10.1007/s11096-025-01924-0. [PMID: 40293640 DOI: 10.1007/s11096-025-01924-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Accepted: 04/12/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND The effect of sodium-glucose cotransporter 2 (SGLT2) inhibitors on cancer incidence compared to other hypoglycemic drugs remains unclear. AIM This systematic review and meta-analysis was designed to investigate the association of SGLT2 inhibitors with cancer compared to active comparators. METHOD A systematic search was conducted up to March 11, 2024 across Web of Science, PubMed, and ClinicalTrials.gov, and included trials with a follow-up period of at least 52 weeks. The Mantel-Haenszel statistical method was utilized, applying risk ratio (RR) and 95% confidence intervals (CI) for binary variables. RESULTS Twenty trials covering 16,399 type 2 diabetes mellitus patients were included. Median follow-up duration was 1.0 (1.0) years. The effect of SGLT2 inhibitors on the overall risk of cancer was neutral compared to active comparators (RR 1.00; 95%CI 0.71-1.40; p = 0.99; moderate certainty of evidence). SGLT2 inhibitors did not have a significant impact on breast cancer, endometrial/uterine cancer, gastrointestinal cancer, prostate cancer, renal cancer, or respiratory cancer. Subgroup analysis indicated a significant reduction in the risk of gastrointestinal cancer with SGLT2 inhibitors compared to metformin (RR 0.23; 95%CI 0.06-0.95; p = 0.04). SGLT2 inhibitors potentially increased gastrointestinal cancer risk relative to sulfonylureas (RR 3.52; 95%CI 0.91-13.64; p = 0.07). CONCLUSION SGLT2 inhibitors showed neutral cancer risk in T2DM patients but may reduce gastrointestinal cancer versus metformin, guiding tailored therapy based on patient risk profiles.
Collapse
Affiliation(s)
- Bo Xu
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Bo Kang
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Shaoqian Li
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- The Affiliated Nanhua Hospital, Department of Docimasiology, Hengyang Medical School, University of South China, Hengyang, 421002, Hunan, China
| | - Jixiang Chen
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Jiecan Zhou
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
5
|
Yan J, Feng C, Zhang H, Luo T, Chen H, Chen H. Dapagliflozin ameliorates intestinal stem cell aging by regulating the MAPK signaling pathway in Drosophila. Front Cell Dev Biol 2025; 13:1576258. [PMID: 40337552 PMCID: PMC12055793 DOI: 10.3389/fcell.2025.1576258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 04/07/2025] [Indexed: 05/09/2025] Open
Abstract
Introduction Intestinal stem cells (ISCs) possess the ability to self-renew and differentiate, which is essential for maintaining intestinal tissue homeostasis. However, their functionality significantly declines with age, leading to diminished tissue regeneration and an increased risk of age-associated diseases. Methods This study investigates the effects of Dapagliflozin (DAPA), a novel insulin sensitizer and SGLT2 inhibitor, on aging ISCs using the Drosophila melanogaster model. Our findings demonstrate that DAPA can inhibit the MAPK signaling pathway, as confirmed by network pharmacology analysis and molecular docking experiments. Results DAPA ameliorates ISC aging, improves intestinal function (including enhanced fecal excretion, restored intestinal barrier integrity and acid-base balance), and enhances healthspan. These results highlight the potential of DAPA as an anti-aging therapeutic agent. Discussion This study provides new evidence for the application of DAPA as an anti-aging treatment.
Collapse
Affiliation(s)
- Jinhua Yan
- Center of Gerontology and Geriatrics and Laboratory of Metabolism and Aging Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Chenxi Feng
- West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Hanmei Zhang
- West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Ting Luo
- Center of Gerontology and Geriatrics and Laboratory of Metabolism and Aging Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Haiyang Chen
- Center of Gerontology and Geriatrics and Laboratory of Metabolism and Aging Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Haiou Chen
- Center of Gerontology and Geriatrics and Laboratory of Metabolism and Aging Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
6
|
Kenawy AS, Liu YS, Aiyeolemi A, Okoye G, Park C. Real-world evidence on the association of novel antidiabetic medication use with cancer risk and protective effects: a systematic review and network meta-analysis. Ther Adv Drug Saf 2025; 16:20420986251335214. [PMID: 40290515 PMCID: PMC12033536 DOI: 10.1177/20420986251335214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 03/28/2025] [Indexed: 04/30/2025] Open
Abstract
Background Novel antidiabetic medications (SGLT-2 inhibitors, DPP-4 inhibitors, and GLP-1 agonists) are commonly used worldwide; however, the available research lacks definitive conclusions on their protective effects or potential risks on cancer. Objectives Compared to other antidiabetics, our systematic review and network meta-analysis (NMA) aims to use real-world studies to assess the potential cancer risks or protective effects of these novel antidiabetics. Methods We comprehensively searched PubMed, CINAHL, and Web of Science from their inception until November 30, 2023. We included observational studies examining at least one novel antidiabetics in the systematic review. The novel antidiabetics include sodium-glucose cotransporter-2 inhibitors (SGLT-2i), dipeptidyl peptidase-4 inhibitors (DPP-4i), and glucagon-like peptide-1 agonists (GLP-1a). Design We focused on cohort studies that provided data on cancer incidence and sample size in the NMA. Using NetMetaXL®, the random effects model with informative priors was used in the NMA to estimate the pooled odds ratio (OR) with 95% credible intervals (CrI). Results The systematic review included 62 studies, of which 22 met the inclusion criteria for the NMA. SGLT-2i users had lower overall cancer risk compared to sulfonylureas (OR: 0.54; 95% CrI: 0.40-0.74, low certainty), GLP-1a (OR: 0.70; 95% CrI: 0.53-0.92, low certainty), and DPP-4i users (OR: 0.72; 95% CrI: 0.57-0.92, very low certainty). DPP-4i users also had a lower cancer risk than sulfonylureas users (OR: 0.76; 95% CrI: 0.60-0.96, low certainty). No other statistically significant ORs were found in other direct comparisons. Conclusion SGLT-2i users have a lower risk of developing cancers than sulfonylureas, GLP-1a, and DPP-4i users. These results may improve patient safety by guiding future clinical practice and medication choices. Future studies should investigate the mechanisms behind these observed associations. Trial registration This NMA was registered in PROSPERO (CRD42023469941).
Collapse
Affiliation(s)
- Ahmed S. Kenawy
- Health Outcomes Division, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
| | - Yi-Shao Liu
- Health Outcomes Division, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
| | - Ayobami Aiyeolemi
- Health Outcomes Division, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
| | - Godwin Okoye
- Health Outcomes Division, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
| | - Chanhyun Park
- Health Outcomes Division, College of Pharmacy, The University of Texas at Austin, 2409 University Avenue MC A1930, Austin, TX 78712, USA
| |
Collapse
|
7
|
Vinson AJ, Matas A. Late Allograft Loss and Contemporary Cardiorenal Metabolic Therapies. J Am Soc Nephrol 2025:00001751-990000000-00615. [PMID: 40193211 DOI: 10.1681/asn.0000000726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2025] Open
Abstract
Late kidney allograft loss occurs through one of two mechanisms: ( 1 ) deterioration of kidney function leading to retransplantation or dialysis (death-censored graft loss) and ( 2 ) premature death with a normally functioning transplant (death with graft function)-each accounting for approximately 50% of late kidney graft losses. Late death-censored graft loss typically results from a combination of immune and nonimmune events leading to common nonspecific end points ( e.g ., tubular atrophy, interstitial fibrosis, and glomerulosclerosis). Conversely, leading causes of death with graft function typically include cardiovascular events, malignancy, and infection. With an improved understanding of the multiple mechanism by which late graft dysfunction develops, there is an opportunity to identify patients at greatest risk and institute novel strategies to quell the process. Newer cardiometabolic agents with proven benefit in the general population have not been well-studied in kidney transplant recipients. However, in addition to their potential benefits in reducing cardiovascular, infectious, and malignancy end points (thus minimizing death with graft function risk), many novel agents may have additional anti-inflammatory and/or antifibrotic benefit (minimizing death-censored graft loss risk) in the kidney transplant population. In this review, we summarize existing literature regarding major causes of death-censored graft loss and death with graft function and discuss the potential roles of new cardiorenal metabolic agents including sodium-glucose cotransport 2 inhibitors, nonsteroidal mineralocorticoid receptor antagonists, glucagon-like peptide 1 receptor agonists, and dual endothelin and angiotensin receptor antagonists in the kidney transplant population, including potential mechanisms to improve death with graft function and death-censored graft loss outcomes.
Collapse
Affiliation(s)
- Amanda J Vinson
- Division of Nephrology, Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
- Kidney Research Institute of Nova Scotia
| | - Arthur Matas
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
8
|
Akkus E, Yekedüz E, Ürün Y. Sodium-Glucose Cotransporter 2 (SGLT2) as a Potential Biomarker and Target in Papillary Renal Cell Carcinoma. Clin Genitourin Cancer 2025; 23:102314. [PMID: 40020263 DOI: 10.1016/j.clgc.2025.102314] [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: 01/09/2025] [Revised: 01/31/2025] [Accepted: 02/03/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND SGLT2 is selectively expressed in the human kidney. SGLT2 inhibitors have markedly changed diabetes, heart failure, and kidney disease treatment, and are under investigation in cancer. However, the role of SGLT2 in papillary renal cell carcinoma (pRCC) is not known. METHODS We investigated the SGLT2 gene expression, associated clinical-molecular features, and overall survival (OS) in pRCC. The Cancer Genome Atlas Program and Gene Expression Omnibus data were utilized. mRNA expression z-scores of the SGLT2 gene relative to normal samples (log-RNASeqV2-RSEM, threshold ± 2) were analyzed (low, unaltered, high expression). RESULTS 273 patients were involved. As per mRNA expression, 180 patients (66%) had low, and the remaining had unaltered expression. High correlation (r > 0.6) with SGLT2 was observed in IRX5, STRIP2, LINC00899, SATB2-AS1, FOXC1, IRX3, SLC22A8, SH3BP5 genes (P < .001,q < 0.001 for all) and with the HIF-2α (r:0.43, P < .001,q < 0.001). Tumor mutational burden (P = .365) and aneuploidy scores (P = .976) did not differ, however, among the genes with the highest alteration frequency, SETD2 alterations (15.63% vs. 1.07%, P < .00, q = 0.046) were more frequent in the unaltered-expression group. Differential protein expression analysis showed highly separated proteins (ERBB2, AR, MAPK14, VHL, TGM2 in the low and SHC1, SQSTM1, MYH14, and CDH1 in the unaltered group). The median OS has not reached the median in both groups [Hazard Ratio(HR) for the unaltered group:2.658, 95% Confidence Interval(CI):1.401-5.043, P = .003]. SGLT2 expression remained a significant prognostic factor in multivariable analysis [HR:2.446 (95%CI: 1.199-4.990), P = .014]. CONCLUSIONS This study reveals the first data that SGLT2 might have a role in pRCC as a pathogenic factor and biomarker. Confirmatory mechanistic studies are needed.
Collapse
Affiliation(s)
- Erman Akkus
- Department of Medical Oncology, Ankara University Faculty of Medicine, Ankara, Türkiye; Cancer Research Institute, Ankara University, Ankara, Türkiye
| | - Emre Yekedüz
- Department of Medical Oncology, Ankara University Faculty of Medicine, Ankara, Türkiye; Cancer Research Institute, Ankara University, Ankara, Türkiye; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Yüksel Ürün
- Department of Medical Oncology, Ankara University Faculty of Medicine, Ankara, Türkiye; Cancer Research Institute, Ankara University, Ankara, Türkiye.
| |
Collapse
|
9
|
Cho YK, Kim S, Kim MJ, Lee WJ, Kim YJ, Jung CH. New users of sodium-glucose cotransporter 2 inhibitors are at low risk of incident pancreatic cancer: A nationwide population-based cohort study. DIABETES & METABOLISM 2025; 51:101605. [PMID: 39788347 DOI: 10.1016/j.diabet.2025.101605] [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: 06/02/2024] [Revised: 01/02/2025] [Accepted: 01/02/2025] [Indexed: 01/12/2025]
Abstract
AIM We aimed to investigate whether sodium-glucose cotransporter-2 inhibitors (SGLT2is) are associated with a decreased risk of gastrointestinal (GI) cancers in patients with type 2 diabetes (T2D) compared to other glucose lowering medications (oGLMs). METHODS This active-comparator, new-user cohort study used the nationwide National Health Insurance Service database of the Republic of Korea from September 2014 to June 2020. From 79,423 new users of SGLT2is and 294,707 new users of oGLMs, we used a propensity score to match 59,954 from each of these two treatment groups. We calculated hazard ratios (HRs) and 95 % confidence intervals (CIs) for the incidence of GI cancers, encompassing stomach, colorectal, liver, and pancreatic cancers. RESULTS During the observation period, there were 814 and 916 GI cancers, and 794 and 1,140 deaths in the SGLT2is and oGLMs treatment groups, respectively. The use of SGLT2is was associated with a statistically significant reduction in the incidence of GI cancers, with an adjusted HR of 0.90 (95 % CI: 0.82 to 0.99). However, only the incidence of pancreatic cancer was significantly lower in SGLT2is users compared to non-users, with an adjusted HR of 0.72 (95 % CI: 0.55 - 0.95). In the entire cohort, the multivariable-adjusted HR for pancreatic cancer was 0.70 (95 % CI: 0.56 to 0.88). CONCLUSION For T2D patients, SGLT2i use was associated with a diminished pancreatic cancer risk compared to oGLMs. Future studies should ascertain the potential protective effect of SGLT2is against pancreatic cancer.
Collapse
Affiliation(s)
- Yun Kyung Cho
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Asan Diabetes Center, Asan Medical Center, Seoul, Republic of Korea
| | - Sehee Kim
- Department of Clinical Epidemiology and Biostatistics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Myung Jin Kim
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Asan Diabetes Center, Asan Medical Center, Seoul, Republic of Korea
| | - Woo Je Lee
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Asan Diabetes Center, Asan Medical Center, Seoul, Republic of Korea
| | - Ye-Jee Kim
- Department of Clinical Epidemiology and Biostatistics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Chang Hee Jung
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Asan Diabetes Center, Asan Medical Center, Seoul, Republic of Korea.
| |
Collapse
|
10
|
Murphy A, Shyanti RK, Mishra M. Targeting obesity, metabolic syndrome, and prostate cancer: GLP-1 agonists as emerging therapeutic agents. Discov Oncol 2025; 16:258. [PMID: 40024963 PMCID: PMC11872791 DOI: 10.1007/s12672-025-01878-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 02/03/2025] [Indexed: 03/04/2025] Open
Abstract
Prostate cancer (PCa) is known as the second most common cancer and has one of the highest incidences among male cancers in the United States. In addition, obesity and metabolic syndrome are a rising and continuous issue in the United States, with 41.9% of individuals as obese. The importance of highlighting these figures is the possibility of PCa having a progressive relationship with obesity and metabolic syndromes. The drugs developed for treating obesity and diabetes pose an exciting possibility of therapeutic application for cancer in efforts to relieve the population's rising numbers. Although this connection has not been established in detail, there are some PCa key biomarkers, and their interactions with metabolic products found in obese, diabetic, and PCa patients can provide good starting points for further investigation. One of the significant links between PCa, obesity, and metabolic disease may be due to insulin metabolism. A downstream target identified that could be the link between PCa, metabolic syndromes, and obesity is the forkhead box C2 (FOXC2). FOXC2 has been known to inhibit some insulin-resistant genes and cause the proliferation of PCa. The relationships of FOXC2, insulin resistance, and GLP-1 receptor agonists as potential therapeutic applications have not been thoroughly explored. This review covers a broad relationship of PCa, obesity, metabolic syndromes, possible drugs, and therapeutic targets.
Collapse
Affiliation(s)
- Azura Murphy
- Cancer Research Center, Department of Biological Sciences, Alabama State University, Montgomery, AL, 36104, USA
| | - Ritis Kumar Shyanti
- Cancer Research Center, Department of Biological Sciences, Alabama State University, Montgomery, AL, 36104, USA
| | - Manoj Mishra
- Cancer Research Center, Department of Biological Sciences, Alabama State University, Montgomery, AL, 36104, USA.
| |
Collapse
|
11
|
Avagimyan A, Pogosova N, Rizzo M, Sarrafzadegan N. Doxorubicin-induced cardiometabolic disturbances: what can we do? FRONTIERS IN CLINICAL DIABETES AND HEALTHCARE 2025; 6:1537699. [PMID: 40027901 PMCID: PMC11868090 DOI: 10.3389/fcdhc.2025.1537699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 01/22/2025] [Indexed: 03/05/2025]
Affiliation(s)
- Ashot Avagimyan
- Yerevan State Medical University after M. Heratsi, Yerevan, Armenia
- Isfahan Cardiovascular Research Centre, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nana Pogosova
- Sciences and Preventive Cardiology, National Medical Research Centre of Cardiology after Acad. E. I. Chazov, Moscow, Russia
- Evidence-Based Medicine Department, Institute of Medicine, Patrice Lumumba Peoples' Friendship University of Russia (RUDN), Moscow, Russia
| | - Manfredi Rizzo
- Cardiometabolic Medicine Department, University of Palermo, Palermo, Italy
| | - Nizal Sarrafzadegan
- Isfahan Cardiovascular Research Centre, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
12
|
Pandey A, Alcaraz M, Saggese P, Soto A, Gomez E, Jaldu S, Yanagawa J, Scafoglio C. Exploring the Role of SGLT2 Inhibitors in Cancer: Mechanisms of Action and Therapeutic Opportunities. Cancers (Basel) 2025; 17:466. [PMID: 39941833 PMCID: PMC11815934 DOI: 10.3390/cancers17030466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 01/23/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
Cancer cells utilize larger amounts of glucose than their normal counterparts, and the expression of GLUT transporters is a known diagnostic target and a prognostic factor for many cancers. Recent evidence has shown that sodium-glucose transporters are also expressed in different types of cancer, and SGLT2 has raised particular interest because of the current availability of anti-diabetic drugs that block SGLT2 in the kidney, which could be readily re-purposed for the treatment of cancer. The aim of this article is to perform a narrative review of the existing literature and a critical appraisal of the evidence for a role of SGLT2 inhibitors for the treatment and prevention of cancer. SGLT2 inhibitors block Na-dependent glucose uptake in the proximal kidney tubules, leading to glycosuria and the improvement of blood glucose levels and insulin sensitivity in diabetic patients. They also have a series of systemic effects, including reduced blood pressure, weight loss, and reduced inflammation, which also make them effective for heart failure and kidney disease. Epidemiological evidence in diabetic patients suggests that individuals treated with SGLT2 inhibitors may have a lower incidence and better outcomes of cancer. These studies are confirmed by pre-clinical evidence of an effect of SGLT2 inhibitors against cancer in xenograft and genetically engineered models, as well as by in vitro mechanistic studies. The action of SGLT2 inhibitors in cancer can be mediated by the direct inhibition of glucose uptake in cancer cells, as well as by systemic effects. In conclusion, there is evidence suggesting a potential role of SGLT2 inhibitors against different types of cancer. The most convincing evidence exists for lung and breast adenocarcinomas, hepatocellular carcinoma, and pancreatic cancer. Several ongoing clinical trials will provide more information on the efficacy of SGLT2 inhibitors against cancer.
Collapse
Affiliation(s)
- Aparamita Pandey
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California Los Angeles, 700 Tiverton Drive, Los Angeles, CA 90095, USA; (A.P.); (A.S.); (E.G.); (S.J.)
| | - Martín Alcaraz
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California Los Angeles, 700 Tiverton Drive, Los Angeles, CA 90095, USA; (A.P.); (A.S.); (E.G.); (S.J.)
| | - Pasquale Saggese
- Department of Biology and Biotechnologies Charles Darwin, University of Rome “Sapienza”, Piazzale Aldo Moro 5, 00185 Rome, Italy;
| | - Adriana Soto
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California Los Angeles, 700 Tiverton Drive, Los Angeles, CA 90095, USA; (A.P.); (A.S.); (E.G.); (S.J.)
| | - Estefany Gomez
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California Los Angeles, 700 Tiverton Drive, Los Angeles, CA 90095, USA; (A.P.); (A.S.); (E.G.); (S.J.)
| | - Shreya Jaldu
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California Los Angeles, 700 Tiverton Drive, Los Angeles, CA 90095, USA; (A.P.); (A.S.); (E.G.); (S.J.)
| | - Jane Yanagawa
- Department of Surgery, David Geffen School of Medicine, University of California Los Angeles, 700 Tiverton Drive, Los Angeles, CA 90095, USA;
| | - Claudio Scafoglio
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California Los Angeles, 700 Tiverton Drive, Los Angeles, CA 90095, USA; (A.P.); (A.S.); (E.G.); (S.J.)
| |
Collapse
|
13
|
Tang S, Xu J, Wan P, Jin S, Zhang Y, Xun L, Wang J, Luo M, Chen W, Zuo Z, Tang H, Qi J. Recent advances in the role of high-salt diet in anti- and pro-cancer progression. Front Immunol 2025; 16:1542157. [PMID: 39944693 PMCID: PMC11814453 DOI: 10.3389/fimmu.2025.1542157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 01/13/2025] [Indexed: 05/09/2025] Open
Abstract
Dietary behaviors significantly influence tumor progression, with increasing focus on high-salt diets (HSD) in recent years. Traditionally, HSD has been regarded as a major risk factor for multiple health issues, including hypertension, cardiovascular disease, kidney disease, cancer, and osteoporosis. However, recent studies have uncovered a novel aspect of HSD, suggesting that HSD may inhibit tumor growth in specific pathological conditions by modulating the activity of immune cells that infiltrate tumors and enhancing the effectiveness of PD-1 immunotherapy. This review focused on the duel molecular mechanisms of HSD in cancer development, which are based on the tumor microenvironment, the gut microbiota, and the involvement of sodium transporter channels. The objective of this review is to explore whether HSD could be a potential future oncological therapeutic strategy under specific situation.
Collapse
Affiliation(s)
- Shiwei Tang
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Juan Xu
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Ping Wan
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Affiliated by Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Province Clinical Research Center for Senile Diseases, First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Shumen Jin
- Yunnan Institute of Food and Drug Supervision and Control, Medical Products Administration of Yunnan Province, Kuming, Yunnan, China
| | - Ying Zhang
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Affiliated by Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Linting Xun
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Affiliated by Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Jinli Wang
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Affiliated by Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Mei Luo
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Affiliated by Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Wenjie Chen
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
- Guangdong-Hongkong-Macao Joint Laboratory of Respiratory Infectious Disease, Guangzhou Medical University, Guangzhou, China
| | - Zan Zuo
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Affiliated by Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Provincial Key Laboratory of Clinical Virology, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Hui Tang
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Affiliated by Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Jialong Qi
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Affiliated by Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Province Clinical Research Center for Senile Diseases, First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
- Yunnan Provincial Key Laboratory of Clinical Virology, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
- Yunnan Provincial Key Laboratory of Birth Defects and Genetic Diseases, First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
| |
Collapse
|
14
|
Miao X, Zhang J, Huang W, Wang Y, Jin A, Cao J, Zhao Z. Research Progress of SGLT2 Inhibitors in Cancer Treatment. Drug Des Devel Ther 2025; 19:505-514. [PMID: 39872633 PMCID: PMC11771169 DOI: 10.2147/dddt.s485755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 01/16/2025] [Indexed: 01/30/2025] Open
Abstract
Sodium glucose co-transporter 2 (SGLT2) inhibitors represent a novel class of hypoglycemic drugs that have emerged in recent years. These inhibitors function primarily by blocking the reabsorption of glucose in the kidneys, specifically targeting the SGLT2 proteins in the proximal convoluted tubules. This inhibition results in the reduction of blood glucose levels through increased glucose excretion in the urine. Recent studies have identified SGLT2 expression in various cancer types, suggesting that SGLT2 inhibition can potentially suppress tumor growth. This article provides a comprehensive review of the role of SGLT2 in tumorigenesis and tumor progression, and explores the underlying mechanisms and potential therapeutic applications of SGLT2 inhibitors as anticancer agents.
Collapse
Affiliation(s)
- Xiaoyong Miao
- Department of Anesthesiology, Naval Medical Center, Naval Medical University, Shanghai, People’s Republic of China
| | - Jianing Zhang
- Student Brigade, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Weiyan Huang
- Student Brigade, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Yifei Wang
- Department of Anesthesiology, Naval Medical Center, Naval Medical University, Shanghai, People’s Republic of China
| | - Aixia Jin
- Department of Anesthesiology, Naval Medical Center, Naval Medical University, Shanghai, People’s Republic of China
| | - Jianping Cao
- Department of Anesthesiology, Naval Medical Center, Naval Medical University, Shanghai, People’s Republic of China
| | - Zhenzhen Zhao
- Department of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| |
Collapse
|
15
|
Demirkiran C, Demiryurek S, Demiryurek AT. Recent Progress and Perspectives in Sodium-Glucose Co-transporter 1/2 Inhibitors. Mini Rev Med Chem 2025; 25:354-364. [PMID: 39162279 DOI: 10.2174/0113895575325210240805092741] [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: 04/22/2024] [Revised: 06/10/2024] [Accepted: 06/25/2024] [Indexed: 08/21/2024]
Abstract
Sodium-Glucose Co-transporter-1/2 (SGLT1/2) inhibitors (also called glifozins) are a class of glucose-decreasing drugs in adults with Type 2 Diabetes (T2D). SGLT2 inhibitors diminish sodium and glucose reabsorption in the renal proximal convoluted tubule. Recent clinical trials have revealed that SGLT2 inhibitors might be beneficial for treating diseases other than diabetes, including chronic renal disease and Heart Failure (HF). Currently, SGLT2 inhibitors are recommended not only for the glycemic management of T2D but also for cardiovascular protection. SGLT2 inhibitors have become one of the foundational drugs for HF with reduced Ejection Fraction (HFrEF) treatment and the first medications with proven prognostic benefit in HF with preserved Ejection Fraction (HFpEF). At present, 11 SGLT1/2 inhibitors have been approved for clinical use in different countries. Beyond their anti-hyperglycemic effect, these inhibitors have shown clear cardio- and nephroprotective properties. A growing body of research studies suggests that SGLT1/2 inhibitors may provide potential clinical benefits in metabolic as well as oncological, hematological, and neurological disorders.
Collapse
Affiliation(s)
- Cahit Demirkiran
- Department of Medical Pharmacology, Faculty of Medicine, Gaziantep University, Gaziantep, 27310, Turkiye
| | - Seniz Demiryurek
- Department of Physiology, Faculty of Medicine, Gaziantep University, Gaziantep, 27310, Turkiye
| | - Abdullah Tuncay Demiryurek
- Department of Medical Pharmacology, Faculty of Medicine, Gaziantep University, Gaziantep, 27310, Turkiye
| |
Collapse
|
16
|
Shafi S, Khan MA, Ahmad J, Rabbani SA, Singh S, Najmi AK. Envisioning Glucose Transporters (GLUTs and SGLTs) as Novel Intervention against Cancer: Drug Discovery Perspective and Targeting Approach. Curr Drug Targets 2025; 26:109-131. [PMID: 39377414 DOI: 10.2174/0113894501335877240926101134] [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: 06/27/2024] [Revised: 08/19/2024] [Accepted: 08/19/2024] [Indexed: 10/09/2024]
Abstract
Metabolic reprogramming and altered cellular energetics have been recently established as an important cancer hallmark. The modulation of glucose metabolism is one of the important characteristic features of metabolic reprogramming in cancer. It contributes to oncogenic progression by supporting the increased biosynthetic and bio-energetic demands of tumor cells. This oncogenic transformation consequently results in elevated expression of glucose transporters in these cells. Moreover, various cancers exhibit abnormal transporter expression patterns compared to normal tissues. Recent investigations have underlined the significance of glucose transporters in regulating cancer cell survival, proliferation, and metastasis. Abnormal regulation of these transporters, which exhibit varying affinities for hexoses, could enable cancer cells to efficiently manage their energy supply, offering a crucial edge for proliferation. Exploiting the upregulated expression of glucose transporters, GLUTs, and Sodium Linked Glucose Transporters (SGLTs), could serve as a novel therapeutic intervention for anti-cancer drug discovery as well as provide a unique targeting approach for drug delivery to specific tumor tissues. This review aims to discussthe previous and emerging research on the expression of various types of glucose transporters in tumor tissues, the role of glucose transport inhibitors as a cancer therapy intervention as well as emerging GLUT/SGLT-mediated drug delivery strategies that can be therapeutically employed to target various cancers.
Collapse
Affiliation(s)
- Sadat Shafi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Mohammad Ahmed Khan
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Javed Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Kingdom of Saudi Arabia (KSA)
| | - Syed Arman Rabbani
- Department of Clinical Pharmacy and Pharmacology, Ras Al Khaimah College of Pharmacy, Ras Al Khaimah Medical and Health Science University, Ras Al Khaimah, United Arab Emirates
| | - Shailja Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Abul Kalam Najmi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| |
Collapse
|
17
|
Wu J, Tang L, Zheng F, Chen X, Li L. A review of the last decade: pancreatic cancer and type 2 diabetes. Arch Physiol Biochem 2024; 130:660-668. [PMID: 37646618 DOI: 10.1080/13813455.2023.2252204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/04/2023] [Accepted: 08/21/2023] [Indexed: 09/01/2023]
Abstract
Pancreatic cancer (PC) is a prevalent gastrointestinal tumour known for its high degree of malignancy, resulting in a mere 10% five-year survival rate for most patients. Over the past decade, a growing body of research has shed light on the intricate bidirectional association between PC and Type 2 diabetes (T2DM). The collection of PC- and T2DM-related articles is derived from two comprehensive databases, namely WOS (Web of Science Core Collection) and CNKI (China National Knowledge Infrastructure). This article discusses the last 10 years of research trends in PC and T2DM and explores their potential regulatory relationship as well as related medications.
Collapse
Affiliation(s)
- Jiaqi Wu
- Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Nursing, Southern Medical University, Guangzhou, China
| | - Liang Tang
- Department of General Medicine, Zhuzhou Central Hospital, Zhuzhou, China
| | - Feng Zheng
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Xun Chen
- Department of the Trauma center, Zhuzhou Central Hospital, Zhuzhou, China
- Department of hepatobiliary surgery, Zhuzhou Central Hospital, Zhuzhou, China
| | - Lei Li
- Department of Pathology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
18
|
Kawaguchi T, Fujishima Y, Wakasugi D, Io F, Sato Y, Uchida S, Kitajima Y. Effects of SGLT2 inhibitors on the onset of esophageal varices and extrahepatic cancer in type 2 diabetic patients with suspected MASLD: a nationwide database study in Japan. J Gastroenterol 2024; 59:1120-1132. [PMID: 39392481 PMCID: PMC11541318 DOI: 10.1007/s00535-024-02158-z] [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: 07/26/2024] [Accepted: 10/01/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND & AIM SGLT2 inhibitors (SGLT2i) improve hepatic steatosis in patients with type 2 diabetes mellitus (T2DM) and MASLD. We aimed to investigate the impact of SGLT2i on the incidence of liver-related events and extrahepatic cancer compared to DPP4 inhibitors (DPP4i) in patients with T2DM and suspected MASLD using a medical claims database in Japan. METHODS We conducted a retrospective study using a Japanese medical claims database. Among patients with T2DM who were prescribed SGLT2i or DPP4i (n = 1,628,656), patients with suspected MASLD were classified into SGLT2i (n = 4204) and DPP4i (n = 4204) groups. Effects of SGLT2i on the following outcomes were compared to DPP4i: (1) changes in HbA1c and ALT levels after 6 months, (2) changes in hepatic fibrosis index, and (3) the incidence of liver-related events/extrahepatic cancer over 12 months. RESULTS After 6 months, DPP4i significantly decreased HbA1c levels compared to SGLT2i. In contrast, SGLT2i significantly decreased ALT levels compared to DPP4i. SGLT2i significantly decreased FIB-4 index compared to DPP4i over 12 months. Although no significant difference was observed in the incidence of overall liver-related events between the two groups, SGLT2i significantly reduced the incidence of esophageal varices (HR 0.12, 95%CI 0.01-0.95, P = 0.044). Moreover, SGLT2i significantly suppressed the incidence of extrahepatic cancer (HR 0.50, 95%CI 0.30-0.84, P = 0.009) compared to DPP4i. CONCLUSION SGLT2i was more beneficial than DPP4i in improving the hepatic inflammation and fibrosis indices. Moreover, SGLT2i suppressed the incidence of esophageal varices and extrahepatic cancer compared to DPP4i. SGLT2i may suppress life-threatening events in patients with T2DM and suspected MASLD.
Collapse
Affiliation(s)
- Takumi Kawaguchi
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Japan.
| | | | | | - Fusayo Io
- Taisho Pharmaceutical Co., Ltd., Tokyo, Japan
| | - Yuri Sato
- Taisho Pharmaceutical Co., Ltd., Tokyo, Japan
| | | | | |
Collapse
|
19
|
Kounatidis D, Vallianou NG, Karampela I, Rebelos E, Kouveletsou M, Dalopoulos V, Koufopoulos P, Diakoumopoulou E, Tentolouris N, Dalamaga M. Anti-Diabetic Therapies and Cancer: From Bench to Bedside. Biomolecules 2024; 14:1479. [PMID: 39595655 PMCID: PMC11591849 DOI: 10.3390/biom14111479] [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: 10/24/2024] [Revised: 11/17/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024] Open
Abstract
Diabetes mellitus (DM) is a significant risk factor for various cancers, with the impact of anti-diabetic therapies on cancer progression differing across malignancies. Among these therapies, metformin has gained attention for its potential anti-cancer effects, primarily through modulation of the AMP-activated protein kinase/mammalian target of rapamycin (AMPK/mTOR) pathway and the induction of autophagy. Beyond metformin, other conventional anti-diabetic treatments, such as insulin, sulfonylureas (SUs), pioglitazone, and dipeptidyl peptidase-4 (DPP-4) inhibitors, have also been examined for their roles in cancer biology, though findings are often inconclusive. More recently, novel medications, like glucagon-like peptide-1 (GLP-1) receptor agonists, dual GLP-1/glucose-dependent insulinotropic polypeptide (GIP) agonists, and sodium-glucose co-transporter-2 (SGLT-2) inhibitors, have revolutionized DM management by not only improving glycemic control but also delivering substantial cardiovascular and renal benefits. Given their diverse metabolic effects, including anti-obesogenic properties, these novel agents are now under meticulous investigation for their potential influence on tumorigenesis and cancer advancement. This review aims to offer a comprehensive exploration of the evolving landscape of glucose-lowering treatments and their implications in cancer biology. It critically evaluates experimental evidence surrounding the molecular mechanisms by which these medications may modulate oncogenic signaling pathways and reshape the tumor microenvironment (TME). Furthermore, it assesses translational research and clinical trials to gauge the practical relevance of these findings in real-world settings. Finally, it explores the potential of anti-diabetic medications as adjuncts in cancer treatment, particularly in enhancing the efficacy of chemotherapy, minimizing toxicity, and addressing resistance within the framework of immunotherapy.
Collapse
Affiliation(s)
- Dimitris Kounatidis
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Medical School, Laiko General Hospital, National and Kapustina University of Athens, 11527 Athens, Greece; (D.K.); (E.R.); (M.K.); (E.D.); (N.T.)
| | - Natalia G. Vallianou
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126 Athens, Greece; (N.G.V.); (V.D.); (P.K.)
| | - Irene Karampela
- 2nd Department of Critical Care, Medical School, Attikon General University Hospital, University of Athens, 1 Rimini str., 12461 Athens, Greece;
| | - Eleni Rebelos
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Medical School, Laiko General Hospital, National and Kapustina University of Athens, 11527 Athens, Greece; (D.K.); (E.R.); (M.K.); (E.D.); (N.T.)
| | - Marina Kouveletsou
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Medical School, Laiko General Hospital, National and Kapustina University of Athens, 11527 Athens, Greece; (D.K.); (E.R.); (M.K.); (E.D.); (N.T.)
| | - Vasileios Dalopoulos
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126 Athens, Greece; (N.G.V.); (V.D.); (P.K.)
| | - Petros Koufopoulos
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126 Athens, Greece; (N.G.V.); (V.D.); (P.K.)
| | - Evanthia Diakoumopoulou
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Medical School, Laiko General Hospital, National and Kapustina University of Athens, 11527 Athens, Greece; (D.K.); (E.R.); (M.K.); (E.D.); (N.T.)
| | - Nikolaos Tentolouris
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Medical School, Laiko General Hospital, National and Kapustina University of Athens, 11527 Athens, Greece; (D.K.); (E.R.); (M.K.); (E.D.); (N.T.)
| | - Maria Dalamaga
- Department of Biological Chemistry, National and Kapodistrian University of Athens, 75 Mikras Asias str., 11527 Athens, Greece
| |
Collapse
|
20
|
Al Khzem AH, Gomaa MS, Alturki MS, Tawfeeq N, Sarafroz M, Alonaizi SM, Al Faran A, Alrumaihi LA, Alansari FA, Alghamdi AA. Drug Repurposing for Cancer Treatment: A Comprehensive Review. Int J Mol Sci 2024; 25:12441. [PMID: 39596504 PMCID: PMC11595001 DOI: 10.3390/ijms252212441] [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: 10/28/2024] [Revised: 11/12/2024] [Accepted: 11/17/2024] [Indexed: 11/28/2024] Open
Abstract
Cancer ranks among the primary contributors to global mortality. In 2022, the global incidence of new cancer cases reached about 20 million, while the number of cancer-related fatalities reached 9.7 million. In Saudi Arabia, there were 13,399 deaths caused by cancer and 28,113 newly diagnosed cases of cancer. Drug repurposing is a drug discovery strategy that has gained special attention and implementation to enhance the process of drug development due to its time- and money-saving effect. It involves repositioning existing medications to new clinical applications. Cancer treatment is a therapeutic area where drug repurposing has shown the most prominent impact. This review presents a compilation of medications that have been repurposed for the treatment of various types of cancers. It describes the initial therapeutic and pharmacological classes of the repurposed drugs and their new applications and mechanisms of action in cancer treatment. The review reports on drugs from various pharmacological classes that have been successfully repurposed for cancer treatment, including approved ones and those in clinical trials and preclinical development. It stratifies drugs based on their anticancer repurpose as multi-type, type-specific, and mechanism-directed, and according to their pharmacological classes. The review also reflects on the future potential that drug repurposing has in the clinical development of novel anticancer therapies.
Collapse
Affiliation(s)
- Abdulaziz H. Al Khzem
- Department of Pharmaceutical Chemistry, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Eastern Province, Saudi Arabia; (M.S.A.); (N.T.); (M.S.)
| | - Mohamed S. Gomaa
- Department of Pharmaceutical Chemistry, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Eastern Province, Saudi Arabia; (M.S.A.); (N.T.); (M.S.)
| | - Mansour S. Alturki
- Department of Pharmaceutical Chemistry, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Eastern Province, Saudi Arabia; (M.S.A.); (N.T.); (M.S.)
| | - Nada Tawfeeq
- Department of Pharmaceutical Chemistry, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Eastern Province, Saudi Arabia; (M.S.A.); (N.T.); (M.S.)
| | - Mohammad Sarafroz
- Department of Pharmaceutical Chemistry, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Eastern Province, Saudi Arabia; (M.S.A.); (N.T.); (M.S.)
| | - Shareefa M. Alonaizi
- College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Eastern Province, Saudi Arabia; (S.M.A.); (A.A.F.); (L.A.A.); (F.A.A.); (A.A.A.)
| | - Alhassan Al Faran
- College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Eastern Province, Saudi Arabia; (S.M.A.); (A.A.F.); (L.A.A.); (F.A.A.); (A.A.A.)
| | - Laela Ahmed Alrumaihi
- College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Eastern Province, Saudi Arabia; (S.M.A.); (A.A.F.); (L.A.A.); (F.A.A.); (A.A.A.)
| | - Fatimah Ahmed Alansari
- College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Eastern Province, Saudi Arabia; (S.M.A.); (A.A.F.); (L.A.A.); (F.A.A.); (A.A.A.)
| | - Abdullah Abbas Alghamdi
- College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Eastern Province, Saudi Arabia; (S.M.A.); (A.A.F.); (L.A.A.); (F.A.A.); (A.A.A.)
| |
Collapse
|
21
|
Suzuki Y, Kaneko H, Okada A, Ko T, Jimba T, Fujiu K, Takeda N, Morita H, Komuro J, Ieda M, Node K, Komuro I, Yasunaga H, Takeda N. Association of SGLT2 inhibitors with incident cancer. DIABETES & METABOLISM 2024; 50:101585. [PMID: 39455021 DOI: 10.1016/j.diabet.2024.101585] [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: 07/26/2024] [Revised: 09/24/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024]
Abstract
AIM It remains unknown whether sodium-glucose cotransporter 2 inhibitors (SGLT2i) could be associated with incident cancer. METHODS We analyzed individuals having diabetes and newly prescribed SGLT2i or dipeptidyl peptidase 4 inhibitors (DPP4i) in a large-scale epidemiological database. The primary outcome was the incidence of cancer. A propensity score matching algorithm was employed to compare the subsequent development of cancer between the SGLT2i and DPP4i groups. RESULTS After 1:2 propensity score matching, 26,823 individuals (8,941 SGLT2i, 17,882 DPP4i) were analyzed. During the mean follow-up duration of 2.0 ± 1.6 years, 1,076 individuals developed cancer. SGLT2i administration was associated with a reduced risk of cancer (HR 0.80, 95 % CI 0.70-0.91). Particularly, SGLT2i administration was related to a lower risk of colorectal cancer (HR 0.71, 95 % CI 0.50-0.998). Our primary findings remained consistent across various sensitivity analyses, including overlap weighting analysis (HR 0.79, 95 % CI 0.66-0.94), inverse probability of treatment weighting 0.75 (95 % CI 0.65-0.86), and induction period settings 0.78 (95 % CI 0.65-0.93). The risk of developing cancer was comparable among individual SGLT2is (P-value of 0.1738). CONCLUSION Our investigation using nationwide real-world data demonstrated the potential advantage of SGLT2i over DPP4i in reducing the development of cancer in individuals with diabetes.
Collapse
Affiliation(s)
- Yuta Suzuki
- The Department of Cardiovascular Medicine, The University of Tokyo, Tokyo, Japan; Center for Outcomes Research and Economic Evaluation for Health, National Institute of Public Health, Saitama, Japan
| | - Hidehiro Kaneko
- The Department of Cardiovascular Medicine, The University of Tokyo, Tokyo, Japan; The Department of Advanced Cardiology, The University of Tokyo, Tokyo, Japan.
| | - Akira Okada
- Department of Prevention of Diabetes and Lifestyle-Related Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Toshiyuki Ko
- The Department of Cardiovascular Medicine, The University of Tokyo, Tokyo, Japan
| | - Takahiro Jimba
- The Department of Cardiovascular Medicine, The University of Tokyo, Tokyo, Japan
| | - Katsuhito Fujiu
- The Department of Cardiovascular Medicine, The University of Tokyo, Tokyo, Japan; The Department of Advanced Cardiology, The University of Tokyo, Tokyo, Japan
| | - Norifumi Takeda
- The Department of Cardiovascular Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Morita
- The Department of Cardiovascular Medicine, The University of Tokyo, Tokyo, Japan
| | - Jin Komuro
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Masaki Ieda
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Koichi Node
- Department of Cardiovascular Medicine, Saga University, Saga, Japan
| | - Issei Komuro
- The Department of Cardiovascular Medicine, The University of Tokyo, Tokyo, Japan; Department of Frontier Cardiovascular Science, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; International University of Health and Welfare, Tokyo, Japan
| | - Hideo Yasunaga
- Department of Clinical Epidemiology and Health Economics, School of Public Health, The University of Tokyo, Tokyo, Japan
| | - Norihiko Takeda
- The Department of Cardiovascular Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
22
|
Dhas Y, Biswas N, M R D, Jones LD, Ashili S. Repurposing metabolic regulators: antidiabetic drugs as anticancer agents. MOLECULAR BIOMEDICINE 2024; 5:40. [PMID: 39333445 PMCID: PMC11436690 DOI: 10.1186/s43556-024-00204-z] [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: 05/10/2024] [Accepted: 08/26/2024] [Indexed: 09/29/2024] Open
Abstract
Drug repurposing in cancer taps into the capabilities of existing drugs, initially designed for other ailments, as potential cancer treatments. It offers several advantages over traditional drug discovery, including reduced costs, reduced development timelines, and a lower risk of adverse effects. However, not all drug classes align seamlessly with a patient's condition or long-term usage. Hence, repurposing of chronically used drugs presents a more attractive option. On the other hand, metabolic reprogramming being an important hallmark of cancer paves the metabolic regulators as possible cancer therapeutics. This review emphasizes the importance and offers current insights into the repurposing of antidiabetic drugs, including metformin, sulfonylureas, sodium-glucose cotransporter 2 (SGLT2) inhibitors, dipeptidyl peptidase 4 (DPP-4) inhibitors, glucagon-like peptide-1 receptor agonists (GLP-1RAs), thiazolidinediones (TZD), and α-glucosidase inhibitors, against various types of cancers. Antidiabetic drugs, regulating metabolic pathways have gained considerable attention in cancer research. The literature reveals a complex relationship between antidiabetic drugs and cancer risk. Among the antidiabetic drugs, metformin may possess anti-cancer properties, potentially reducing cancer cell proliferation, inducing apoptosis, and enhancing cancer cell sensitivity to chemotherapy. However, other antidiabetic drugs have revealed heterogeneous responses. Sulfonylureas and TZDs have not demonstrated consistent anti-cancer activity, while SGLT2 inhibitors and DPP-4 inhibitors have shown some potential benefits. GLP-1RAs have raised concerns due to possible associations with an increased risk of certain cancers. This review highlights that further research is warranted to elucidate the mechanisms underlying the potential anti-cancer effects of these drugs and to establish their efficacy and safety in clinical settings.
Collapse
Affiliation(s)
- Yogita Dhas
- Rhenix Lifesciences, Hyderabad, 500038, Telangana, India
| | - Nupur Biswas
- Rhenix Lifesciences, Hyderabad, 500038, Telangana, India.
- CureScience, 5820 Oberlin Dr, Suite 202, San Diego, CA, 92121, USA.
| | | | - Lawrence D Jones
- CureScience, 5820 Oberlin Dr, Suite 202, San Diego, CA, 92121, USA
| | | |
Collapse
|
23
|
Cristovão A, Andrade N, Martel F, Silva C. Effect of Sodium-Glucose Co-Transporter 2 Inhibitors Combined with Metformin on Pancreatic Cancer Cell Lines. Int J Mol Sci 2024; 25:9932. [PMID: 39337420 PMCID: PMC11432055 DOI: 10.3390/ijms25189932] [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: 07/26/2024] [Revised: 09/06/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Pancreatic cancer (PC) is the ninth-leading cause of cancer-related deaths worldwide. Diabetic patients have an increased risk and mortality rates for PC. Sodium-glucose co-transporter 2 (SGLT2) inhibitors and metformin (Met) are widely used anti-diabetic medications. Both Met and SGLT2 inhibitors have anticancer properties in PC, but nothing is known concerning their combined effect. So, we investigated the in vitro effect of SGLT2 inhibitors combined with Met. Canagliflozin and dapagliflozin possessed cytotoxic, antiproliferative, and pro-apoptotic properties in the tested PC cell lines. In PANC-1 cells, the antimigratory and pro-apoptotic effects were enhanced when dapagliflozin was combined with Met, and G1 cell cycle arrest was enhanced when dapagliflozin or canagliflozin was combined with Met. In AsPC-1 cells, the cytotoxic effect and the G1 cell cycle arrest were enhanced when canagliflozin and dapagliflozin, respectively, were combined with Met. Only the cytotoxic effects of SGLT2 inhibitors, but not the combination treatments, involved PI3K and JNK-dependent pathways in AsPC-1 cells. In conclusion, combination treatments increased the anticancer effects in a cell type-dependent way in the two investigated cell lines. Additionally, the cytotoxic effect of SGLT2 inhibitors was dependent on the PI3K and JNK pathways in AsPC-1 cells, but Met appears to act via a distinct mechanism.
Collapse
Affiliation(s)
- André Cristovão
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine of Porto, University of Porto, 4200-319 Porto, Portugal; (A.C.); (N.A.); (C.S.)
| | - Nelson Andrade
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine of Porto, University of Porto, 4200-319 Porto, Portugal; (A.C.); (N.A.); (C.S.)
- REQUIMTE/LAQV, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4200-135 Porto, Portugal
| | - Fátima Martel
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine of Porto, University of Porto, 4200-319 Porto, Portugal; (A.C.); (N.A.); (C.S.)
- Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, 4200-135 Porto, Portugal
| | - Cláudia Silva
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine of Porto, University of Porto, 4200-319 Porto, Portugal; (A.C.); (N.A.); (C.S.)
- REQUIMTE/LAQV, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4200-135 Porto, Portugal
| |
Collapse
|
24
|
Naeimzadeh Y, Tajbakhsh A, Nemati M, Fallahi J. Exploring the anti-cancer potential of SGLT2 inhibitors in breast cancer treatment in pre-clinical and clinical studies. Eur J Pharmacol 2024; 978:176803. [PMID: 38950839 DOI: 10.1016/j.ejphar.2024.176803] [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: 03/13/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/03/2024]
Abstract
The link between type 2 diabetes mellitus (T2DM) and an increased risk of breast cancer (BC) has prompted the exploration of novel therapeutic strategies targeting shared metabolic pathways. This review focuses on the emerging evidence surrounding the potential anti-cancer effects of sodium-glucose cotransporter-2 (SGLT2) inhibitors in the context of BC. Preclinical studies have demonstrated that various SGLT2 inhibitors, such as canagliflozin, dapagliflozin, ipragliflozin, and empagliflozin, can inhibit the proliferation of BC cells, induce apoptosis, and modulate key cellular signaling pathways. These mechanisms include the activation of AMP-activated protein kinase (AMPK), suppression of mammalian target of rapamycin (mTOR) signaling, and regulation of lipid metabolism and inflammatory mediators. The combination of SGLT2 inhibitors with conventional treatments, including chemotherapy and radiotherapy, as well as targeted therapies like phosphoinositide 3-kinases (PI3K) inhibitors, has shown promising results in enhancing the anti-cancer efficacy and potentially reducing treatment-related toxicities. The identification of specific biomarkers or genetic signatures that predict responsiveness to SGLT2 inhibitor therapy could enable more personalized treatment selection and optimization, particularly for challenging BC subtypes [e, g., triple negative BC (TNBC)]. Ongoing and future clinical trials investigating the use of SGLT2 inhibitors, both as monotherapy and in combination with other agents, will be crucial in elucidating their translational potential and guiding their integration into comprehensive BC care. Overall, SGLT2 inhibitors represent a novel and promising therapeutic approach with the potential to improve clinical outcomes for patients with various subtypes of BC, including the aggressive and chemo-resistant TNBC.
Collapse
Affiliation(s)
- Yasaman Naeimzadeh
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, 7133654361, Iran
| | - Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mahnaz Nemati
- Amir Oncology Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Jafar Fallahi
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, 7133654361, Iran.
| |
Collapse
|
25
|
Ghezzi C, Ellingson BM, Lai A, Liu J, Barrio JR, Wright EM. Effect of Jardiance on glucose uptake into astrocytomas. J Neurooncol 2024; 169:437-444. [PMID: 39037688 PMCID: PMC11341586 DOI: 10.1007/s11060-024-04746-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 06/11/2024] [Indexed: 07/23/2024]
Abstract
PURPOSE SGLT2, the sodium glucose cotransporter two, is expressed in human pancreatic, prostate and brain tumors, and in a mouse cancer model SGLT2 inhibitors reduce tumor glucose uptake and growth. In this study we have measured the effect of a specific SGLT2 inhibitor, Jardiance® (Empagliflozin), on glucose uptake into astrocytomas in patients. METHODS We have used a specific SGLT glucose tracer, α-methyl-4-[18F]fluoro-4-deoxy-α-D-glucopyranoside (Me4FDG), and Positron Emission Tomography (PET) to measure glucose uptake. Four of five patients enrolled had WHO grade IV glioblastomas, and one had a low grade WHO Grade II astrocytoma. Two dynamic brain PET scans were conducted on each patient, one before and one after treatment with a single oral dose of Jardiance, a specific SGLT2 inhibitor. As a control, we also determined the effect of oral Jardiance on renal SGLT2 activity. RESULTS In all five patients an oral dose (25 or 100 mg) of Jardiance reduced Me4FDG tumor accumulation, highly significant inhibition in four, and inhibited SGLT2 activity in the kidney. CONCLUSIONS These initial experiments show that SGLT2 is a functional glucose transporter in astocytomas, and Jardiance inhibited glucose uptake, a drug approved by the FDA to treat type 2 diabetes mellitus (T2DM), heart failure, and renal failure. We suggest that clinical trials be initiated to determine whether Jardiance reduces astrocytoma growth in patients.
Collapse
Affiliation(s)
- Chiara Ghezzi
- Department of Physiology, The David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095-1751, USA
| | - Benjamin M Ellingson
- Department of Radiological Sciences, The David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Albert Lai
- Department of Neurology, The David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Jie Liu
- Department of Molecular and Medical Pharmacology, The David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Jorge R Barrio
- Department of Molecular and Medical Pharmacology, The David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Ernest M Wright
- Department of Physiology, The David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095-1751, USA.
| |
Collapse
|
26
|
Baghel R, Chhikara N, Kumar P, Tamrakar AK. SGLT2 inhibitors for the treatment of diabetes: a patent review (2019-23). Expert Opin Ther Pat 2024; 34:807-823. [PMID: 39078140 DOI: 10.1080/13543776.2024.2379929] [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: 02/26/2024] [Revised: 05/27/2024] [Accepted: 07/04/2024] [Indexed: 07/31/2024]
Abstract
INTRODUCTION The sodium-glucose co-transporter 2 (SGLT2) inhibitors are FDA-approved class of drugs for diabetes management. They improve glycemic control by inducing glucosuria. Notwithstanding with potent anti-hyperglycemic activity, SGLT2 inhibitors are emerging as drugs with multifaceted therapeutic potential, evidenced for cardioprotective, renoprotective, antihypertensive, and neuroprotective activities. Continuous attempts are being accomplished through structural modification, development of new formulation, or combination with other drugs, to enhance the bioactivity spectrum of SGLT2 inhibitors for better management of diabetes and related complications. AREAS COVERED This review comprises a summary of patent applications, acquired using the Espacenet Patent Search database, concerning SGLT2 inhibitors from 2019 to 2023, with focus on improving therapeutic potentials in management of diabetes and metabolic complications. EXPERT OPINION SGLT2 inhibitors have provided an exciting treatment option for diabetes. Originally developed as anti-hyperglycemic agents, SGLT2 inhibitors exert pleiotropic metabolic responses and have emerged as promising antidiabetic agents with cardio-protective and reno-protective activities. Given their distinct therapeutic profile, SGLT2 inhibitors have revolutionized the management of diabetes and associated complications. Emerging evidences on their therapeutic potential against cancer, male reproductive dysfunctions, and neurodegenerative diseases indicate that further research in this field may unfold novel prospective on their plausible use in the management of other chronic conditions.
Collapse
Affiliation(s)
- Rahul Baghel
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad, Ghaziabad, India
| | - Nikita Chhikara
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad, Ghaziabad, India
| | - Pawan Kumar
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad, Ghaziabad, India
| | - Akhilesh Kumar Tamrakar
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad, Ghaziabad, India
| |
Collapse
|
27
|
Mohite P, Lokwani DK, Sakle NS. Exploring the therapeutic potential of SGLT2 inhibitors in cancer treatment: integrating in silico and in vitro investigations. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:6107-6119. [PMID: 38416196 DOI: 10.1007/s00210-024-03021-x] [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: 01/12/2024] [Accepted: 02/21/2024] [Indexed: 02/29/2024]
Abstract
The present study aimed to investigate the anti-cancer mechanism of canagliflozin (CANA) and dapagliflozin (DAPA), sodium-glucose co-transporter-2 (SGLT2) inhibitors, using in silico and in vitro approaches. Network pharmacology was employed to predict the targets of the inhibitors and GO gene enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway annotation conducted to explore the interacting pathways. Molecular docking and molecular dynamic (MD) simulation studies were performed to confirm the important targets and assess conformational stability. In vitro cytotoxicity assays, MIA-PaCa-2 and DU-145 cell lines CANA and DAPA was performed. Protein-protein interaction (PPI) network analysis indicated that CANA and DAPA exert anticancer effects through MAPK, mTOR, EGFR-KRAS-BRAF, FGFR, and PI3KA pathways. KEGG analysis revealed that these inhibitors could be used in the treatment of various cancers, including breast, prostate, pancreatic, chronic myeloid leukemia, thyroid, small cell lung, gastric, and bladder cancer. Docking results showed highest affinity for MAPK1 for CANA (- 9.60 kcal/mol) and DAPA (- 9.58 kcal/mol). MD simulation results showed that RMSD values for the MAPK1-compound exhibit remarkable stability over a timeframe of 100 ns. In cytotoxicity assays using MIA-PaCa-2 and DU-145 cell lines, CANA demonstrated a potential antiproliferative effect on the pancreatic cell line MIA-PaCa-2 after 48 h of treatment at a concentration of 100 µg/ml. Furthermore, CANA arrested the cell cycle in the sub-G1 phase and induced late apoptosis and necrosis in MIA-PaCa-2 cell line. Based on these findings, CANA shows promise as a potential novel treatment strategy for pancreatic cancer.
Collapse
Affiliation(s)
- Prasanna Mohite
- Dr. Rafiq Zakaria Campus, Y. B. Chavan College of Pharmacy, Aurangabad, 431001, Maharashtra, India
| | - Deepak K Lokwani
- Department of Pharmaceutical Chemistry, Rajarshi Shahu College of Pharmacy, Buldana, Maharashtra, India
| | - Nikhil S Sakle
- Dr. Rafiq Zakaria Campus, Y. B. Chavan College of Pharmacy, Aurangabad, 431001, Maharashtra, India.
| |
Collapse
|
28
|
Mohsin S, Hasan M, Sheikh ZM, Mustafa F, Tegeltija V, Kumar S, Kumar J. Efficacy of SGLT2 inhibitors for anthracycline-induced cardiotoxicity: a meta-analysis in cancer patients. Future Cardiol 2024; 20:395-407. [PMID: 38962942 PMCID: PMC11457630 DOI: 10.1080/14796678.2024.2363673] [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: 12/12/2023] [Accepted: 05/31/2024] [Indexed: 07/05/2024] Open
Abstract
Aim: Sodium-glucose cotransporter-2 inhibitors (SGLT2i) lower anthracycline-induced cardiotoxicity.Methods: PubMed and Google Scholar were searched until September 2023 for studies regarding SGLT2i for treating anthracycline-induced cardiotoxicity. Overall mortality and cardiovascular events were considered. Using a random-effects model, data pooled RR and HR at a 95% confidence interval (CI).Results: 3 cohort studies were identified, analyzing 2817 patients. Results display a significant reduction in overall mortality [RR = 0.52 (0.33-0.82); p = 0.005; I2= 32%], HF hospitalization [RR = 0.20 (0.04-1.02); p = 0.05; I2= 0%] and no significant reduction in HF incidence [RR = 0.50 (0.20-1.16); p = 0.11, I2= 0%].Conclusion: SGLT2i mitigates mortality and hospitalization due to heart failure, improving cancer patient's chances of survival by undergoing anthracycline treatment.
Collapse
Affiliation(s)
- Sana Mohsin
- College of Medicine, Ziauddin University, Karachi, Pakistan
| | - Misha Hasan
- College of Medicine, Ziauddin University, Karachi, Pakistan
| | | | - Fatima Mustafa
- College of Medicine, Ziauddin University, Karachi, Pakistan
| | - Vesna Tegeltija
- School of Medicine, Wayne State University, Detroit, MI, USA
| | - Sarwan Kumar
- School of Medicine, Wayne State University, Detroit, MI, USA
| | - Jai Kumar
- School of Medicine, Wayne State University, Detroit, MI, USA
| |
Collapse
|
29
|
Gongora CA, Zhang L, Mattei JL, Ruiz-Mori E, Gonzalez-Robledo G, Slipczuk L, Lara J, Cossio-Aranda JE, Badimon J. Are Sodium-Glucose Cotransporter-2 Inhibitors the Cherry on Top of Cardio-Oncology Care? Cardiovasc Drugs Ther 2024:10.1007/s10557-024-07604-x. [PMID: 38958827 DOI: 10.1007/s10557-024-07604-x] [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] [Accepted: 06/22/2024] [Indexed: 07/04/2024]
Abstract
The increasing aging of the population combined with improvements in cancer detection and care has significantly improved the survival and quality of life of cancer patients. These benefits are hampered by the increase of cardiovascular diseases being heart failure the most frequent manifestation of cardiotoxicity and becoming the major cause of morbidity and mortality among cancer survivor. Current strategies to prevent cardiotoxicity involves different approaches such as optimal management of CV risk factors, use of statins and/or neurohormonal medications, and, in some cases, even the use of chelating agents. As a class, SGLT2-i have revolutionized the therapeutic horizon of HF patients independently of their ejection fraction or glycemic status. There is an abundance of data from translational and observational clinical studies supporting a potential beneficial role of SGLT2-i in mitigating the cardiotoxic effects of cancer patients receiving anthracyclines. These findings underscore the need for more robust clinical trials to investigate the effect on cardiovascular outcomes of the prophylactic SGLT2-i treatment in patients undergoing cancer treatment.
Collapse
Affiliation(s)
- Carlos A Gongora
- Division of Cardiology, Montefiore Medical Center at Albert Einstein School of Medicine, Bronx, NY, USA
| | - Lili Zhang
- Division of Cardiology, Montefiore Medical Center at Albert Einstein School of Medicine, Bronx, NY, USA
| | | | - Enrique Ruiz-Mori
- Cardiology Department, Instituto de Enfermedades Neoplásicas, Lima, Peru
| | - Gina Gonzalez-Robledo
- Heart Failure Unit, University Hospital Fundacion Santa Fe, Universidad de los Andes, Bogota, Colombia
| | - Leandro Slipczuk
- Division of Cardiology, Montefiore Medical Center at Albert Einstein School of Medicine, Bronx, NY, USA
| | - Joffre Lara
- Hospital Juan Tanca Marengo, Guayaquil, Ecuador
| | | | - Juan Badimon
- Atherothrombosis Research Unit, Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
30
|
Fath AR, Aglan M, Aglan A, Chilton RJ, Trakhtenbroit A, Al-Shammary OA, Oppong-Nkrumah O, Lenihan DJ, Dent SF, Otchere P. Cardioprotective Potential of Sodium-Glucose Cotransporter-2 Inhibitors in Patients With Cancer Treated With Anthracyclines: An Observational Study. Am J Cardiol 2024; 222:175-182. [PMID: 38692401 DOI: 10.1016/j.amjcard.2024.04.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 03/20/2024] [Accepted: 04/19/2024] [Indexed: 05/03/2024]
Abstract
Anthracyclines are pivotal in cancer treatment, yet their clinical utility is hindered by the risk of cardiotoxicity. Preclinical studies highlight the effectiveness of sodium-glucose cotransporter-2 inhibitors (SGLT2i) in mitigating anthracycline-induced cardiotoxicity. Nonetheless, the translation of these findings to clinical practice remains uncertain. This study aims to evaluate the safety and potential of SGLT2i for preventing cardiotoxicity in patients with cancer, without preexisting heart failure (HF), receiving anthracyclines therapy. Using the TriNetX Global Research Network, patients with cancer, without previous HF diagnosis, receiving anthracycline therapy were identified and classified into 2 groups based on SGLT2i usage. A 1:1 propensity score matching was used to control for baseline characteristics between the 2 groups. Patients were followed for 2 years. The primary end point was new-onset HF, and the secondary end points were HF exacerbation, new-onset arrhythmia, myocardial infarction, all-cause mortality, and all-cause hospitalization. Safety outcomes included acute renal failure and creatinine levels. A total of 79,074 patients were identified, and 1,412 were included post-matching (706 in each group). They comprised 53% females, 62% White, with a mean age of 62.5 ± 11.4 years. Over the 2-year follow-up period, patients on SGLT2i had lower rates of new-onset HF (hazard ratio 0.147, 95% confidence interval 0.073 to 0.294) and arrhythmia (hazard ratio 0.397, 95% confidence interval 0.227 to 0.692) compared with those not on SGLT2i. The incidence of all-cause mortality, myocardial infarction, all-cause hospitalization, and safety outcomes were similar between both groups. In conclusion, among patients with cancer receiving anthracycline therapy without preexisting HF, SGLT2i use demonstrates both safety and effectiveness in reducing anthracycline-induced cardiotoxicity, with a decreased incidence of new-onset HF, HF exacerbation, and arrhythmias.
Collapse
Affiliation(s)
- Ayman R Fath
- Cardiology Department, University of Texas Health Science Center, San Antonio, Texas.
| | - Mostafa Aglan
- Internal Medicine Department, Lahey Hospital and Medical Center, Burlington Massachusetts
| | - Amro Aglan
- Internal Medicine Department, Lahey Hospital and Medical Center, Burlington Massachusetts
| | - Robert J Chilton
- Cardiology Department, University of Texas Health Science Center, San Antonio, Texas
| | - Anatole Trakhtenbroit
- Cardiology Department, University of Texas Health Science Center, San Antonio, Texas
| | - Odaye A Al-Shammary
- Cardiology Department, University of Texas Health Science Center, San Antonio, Texas
| | - Oduro Oppong-Nkrumah
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Canada
| | - Daniel J Lenihan
- Cardiology Department, Saint Francis Healthcare System, Cape Girardeau, Missouri
| | - Susan F Dent
- Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Prince Otchere
- Cardiology Department, University of Texas Health Science Center, San Antonio, Texas
| |
Collapse
|
31
|
Tabernacki T, Wang L, Kaelber DC, Xu R, Berger NA. Non-Insulin Antidiabetic Agents and Lung Cancer Risk in Drug-Naive Patients with Type 2 Diabetes Mellitus: A Nationwide Retrospective Cohort Study. Cancers (Basel) 2024; 16:2377. [PMID: 39001440 PMCID: PMC11240387 DOI: 10.3390/cancers16132377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
Lung cancer (LC) is the second most common cancer and the leading cause of cancer deaths in the U.S. Insulin therapy, a key treatment for managing Type 2 Diabetes Mellitus (T2DM), is associated with increased LC risk. The impact of non-insulin antidiabetic drugs, particularly GLP-1 receptor agonists (GLP-1RAs), on LC risk is not well understood. This study evaluated LC risk in T2DM patients, comparing seven non-insulin antidiabetic agents to insulin. Using the TriNetX Analytics platform, we analyzed the de-identified electronic health records of 1,040,341 T2DM patients treated between 2005 and 2019, excluding those with prior antidiabetic use or LC diagnoses. We calculated hazard ratios and confidence intervals for LC risk and used propensity score matching to control for confounding factors. All non-insulin antidiabetic drugs, except alpha-glucosidase inhibitors, were associated with significantly reduced LC risk compared to insulin, with GLP-1RAs showing the greatest reduction (HR: 0.49, 95% CI: 0.41, 0.59). GLP-1RAs were consistently associated with lowered LC risk across all histological types, races, genders, and smoking statuses. These findings suggest that non-insulin antidiabetic drugs, particularly GLP-1RAs, may be preferable for managing T2DM while reducing LC risk.
Collapse
Affiliation(s)
- Tomasz Tabernacki
- Center for Artificial Intelligence in Drug Discovery, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Lindsey Wang
- Center for Science, Health, and Society, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - David C. Kaelber
- Center for Clinical Informatics Research and Education, The MetroHealth System, Cleveland, OH 44109, USA
| | - Rong Xu
- Center for Artificial Intelligence in Drug Discovery, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Nathan A. Berger
- Center for Science, Health, and Society, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
32
|
Anastasio C, Donisi I, Del Vecchio V, Colloca A, Mele L, Sardu C, Marfella R, Balestrieri ML, D'Onofrio N. SGLT2 inhibitor promotes mitochondrial dysfunction and ER-phagy in colorectal cancer cells. Cell Mol Biol Lett 2024; 29:80. [PMID: 38811901 PMCID: PMC11134909 DOI: 10.1186/s11658-024-00599-1] [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: 03/05/2024] [Accepted: 05/17/2024] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND Sodium-glucose transporter 2 (SGLT2) inhibitors (iSGLT2) are approved medications for type 2 diabetes. Recent studies indicate that iSGLT2 inhibit the growth of some cancer cells. However, the mechanism(s) remains to be fully elucidated. METHODS The SGLT2 levels were determined in normal colon CCD 841 CoN and, HCT 116, HT-29, SW480 and LoVo colorectal cancer (CRC) cell lines by quantitative real-time PCR and western blot. The effect of iSGLT2 canagliflozin on cell proliferation was examined using CCK-8, as its role on CRC cells metabolism and tumorigenesis has been evaluated by XF HS Seahorse Bioanalyzer and flow cytometric analyses. Transient gene silencing experiments and analysis of protein-protein interaction network were conducted to evaluate the SGLT2 molecular targets in CRC cells. RESULTS Data showed that the treatment with iSGLT2 (50 µM) for 72 h induced cell cycle arrest (p < 0.001), impaired glucose and energetic metabolism (p < 0.001), promoted apoptotic cell death and ER stress flowing into autophagy (p < 0.001) in HCT 116 and HT-29 cells. These cellular events were accompanied by sirtuin 3 (SIRT3) upregulation (p < 0.01), as also supported by SIRT3 transient silencing experiments resulting in the attenuation of the effects of iSGLT2 on the cellular metabolic/energetic alterations and the induction of programmed cell death. The identification and validation of dipeptidyl peptidase 4 (DPP4) as potential common target of SGLT2 and SIRT3 were also assessed. CONCLUSIONS These results deepened knowledge on the iSGLT2 contribution in limiting CRC tumorigenesis unveiling the SGLT2/SIRT3 axis in the cytotoxic mechanisms.
Collapse
Affiliation(s)
- Camilla Anastasio
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138, Naples, Italy
| | - Isabella Donisi
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138, Naples, Italy
| | - Vitale Del Vecchio
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Via Luciano Armanni 5, 80138, Naples, Italy
| | - Antonino Colloca
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138, Naples, Italy
| | - Luigi Mele
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Via Luciano Armanni 5, 80138, Naples, Italy
| | - Celestino Sardu
- Department of Advanced Clinical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138, Naples, Italy
| | - Raffaele Marfella
- Department of Advanced Clinical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138, Naples, Italy
| | - Maria Luisa Balestrieri
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138, Naples, Italy
| | - Nunzia D'Onofrio
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138, Naples, Italy.
| |
Collapse
|
33
|
Zmaili M, Alzubi J, Alkhayyat M, Albakri A, Alkhalaileh F, Longinow J, Moudgil R. Cancer and Cardiovascular Disease: The Conjoined Twins. Cancers (Basel) 2024; 16:1450. [PMID: 38672532 PMCID: PMC11048405 DOI: 10.3390/cancers16081450] [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: 01/15/2024] [Revised: 03/21/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
Cancer and cardiovascular disease are the two most common causes of death worldwide. As the fields of cardiovascular medicine and oncology continue to expand, the area of overlap is becoming more prominent demanding dedicated attention and individualized patient care. We have come to realize that both fields are inextricably intertwined in several aspects, so much so that the mere presence of one, with its resultant downstream implications, has an impact on the other. Nonetheless, cardiovascular disease and cancer are generally approached independently. The focus that is granted to the predominant pathological entity (either cardiovascular disease or cancer), does not allow for optimal medical care for the other. As a result, ample opportunities for improvement in overall health care are being overlooked. Herein, we hope to shed light on the interconnected relationship between cardiovascular disease and cancer and uncover some of the unintentionally neglected intricacies of common cardiovascular therapeutics from an oncologic standpoint.
Collapse
Affiliation(s)
- Mohammad Zmaili
- Robert and Suzanne Tomsich Department of Cardiovascular Medicine, Sydell and Arnold Miller Family Heart, Vascular and Thoracic Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA;
| | - Jafar Alzubi
- Department of Medicine, Division of Cardiology, Einstein Medical Center, Philadelphia, PA 19141, USA
| | - Motasem Alkhayyat
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Disease and Surgery Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Almaza Albakri
- Jordanian Royal Medical Services, Department of Internal Medicine, King Abdullah II Ben Al-Hussein Street, Amman 11855, Jordan
| | - Feras Alkhalaileh
- Department of Internal Medicine, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Joshua Longinow
- Department of Internal Medicine, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Rohit Moudgil
- Robert and Suzanne Tomsich Department of Cardiovascular Medicine, Sydell and Arnold Miller Family Heart, Vascular and Thoracic Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA;
- Department of Internal Medicine, Cleveland Clinic, Cleveland, OH 44195, USA
| |
Collapse
|
34
|
Buczyńska A, Kościuszko M, Krętowski AJ, Popławska-Kita A. Exploring the clinical utility of DPP-IV and SGLT2 inhibitors in papillary thyroid cancer: a literature review. Front Pharmacol 2024; 15:1323083. [PMID: 38292938 PMCID: PMC10824900 DOI: 10.3389/fphar.2024.1323083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/02/2024] [Indexed: 02/01/2024] Open
Abstract
In the realm of clinical management, Papillary Thyroid Cancer (PTC) stands out as a prevalent thyroid malignancy, characterized by significant metabolic challenges, particularly in the context of carbohydrate metabolism. Recent studies have unveiled promising applications of Dipeptidyl Peptidase-IV (DPP-IV) and Sodium-Glucose Cotransporter 2 (SGLT2) inhibitors, which are conventionally employed in the treatment of type 2 diabetes mellitus (T2DM), as potential adjuncts in anticancer therapy. DPP-IV and SGLT2 inhibitors can be imply to counteract the Warburg effect in cancer, with a specific focus on PTC, owing to their potential metabolic advantages and their influence on the tumor microenvironment, achieved by imposing restrictions on glucose accessibility. Consequently, a comprehensive review has been undertaken, involving meticulous examination of the existing body of evidence pertaining to the utilization of DPP-IV and SGLT2 inhibitors in the context of PTC. The mechanisms of action inherent to these inhibitors have been thoroughly explored, drawing upon insights derived from preclinical investigations. Furthermore, this review initiates discussions concerning the implications for future research directions and the formulation of innovative therapeutic strategies for PTC. As the intricate interplay between carbohydrate metabolism, the Warburg effect, and cancer progression garners increasing attention, attaining a comprehensive understanding of the roles played by DPP-IV and SGLT2 inhibitors in PTC management may serve as the cornerstone for novel approaches aimed at enhancing patient care and broadening the spectrum of available therapeutic modalities.
Collapse
Affiliation(s)
- Angelika Buczyńska
- Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Maria Kościuszko
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland
| | - Adam Jacek Krętowski
- Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland
| | - Anna Popławska-Kita
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
35
|
Garczorz W, Kosowska A, Francuz T. Antidiabetic Drugs in Breast Cancer Patients. Cancers (Basel) 2024; 16:299. [PMID: 38254789 PMCID: PMC10813754 DOI: 10.3390/cancers16020299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/06/2024] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Diabetes is one of the leading chronic conditions worldwide, and breast cancer is the most prevalent cancer in women worldwide. The linkage between diabetes and its ability to increase the risk of breast cancer should always be analyzed in patients. This review focuses on the impact of antihyperglycemic therapy in breast cancer patients. Patients with diabetes have a higher risk of developing cancer than the general population. Moreover, diabetes patients have a higher incidence and mortality of breast cancer. In this review, we describe the influence of antidiabetic drugs from insulin and metformin to the current and emerging therapies, incretins and SGLT-2 inhibitors, on breast cancer prognosis. We also emphasize the role of obesity and the metastasis process in breast cancer patients who are treated with antidiabetic drugs.
Collapse
Affiliation(s)
- Wojciech Garczorz
- Department of Biochemistry, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 18, 40-055 Katowice, Poland; (A.K.); (T.F.)
| | | | | |
Collapse
|
36
|
Chinyama HA, Wei L, Mokgautsi N, Lawal B, Wu ATH, Huang HS. Identification of CDK1, PBK, and CHEK1 as an Oncogenic Signature in Glioblastoma: A Bioinformatics Approach to Repurpose Dapagliflozin as a Therapeutic Agent. Int J Mol Sci 2023; 24:16396. [PMID: 38003585 PMCID: PMC10671581 DOI: 10.3390/ijms242216396] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/27/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive and lethal primary brain tumor whose median survival is less than 15 months. The current treatment regimen comprising surgical resectioning, chemotherapy with Temozolomide (TMZ), and adjuvant radiotherapy does not achieve total patient cure. Stem cells' presence and GBM tumor heterogeneity increase their resistance to TMZ, hence the poor overall survival of patients. A dysregulated cell cycle in glioblastoma enhances the rapid progression of GBM by evading senescence or apoptosis through an over-expression of cyclin-dependent kinases and other protein kinases that are the cell cycle's main regulatory proteins. Herein, we identified and validated the biomarker and predictive properties of a chemoradio-resistant oncogenic signature in GBM comprising CDK1, PBK, and CHEK1 through our comprehensive in silico analysis. We found that CDK1/PBK/CHEK1 overexpression drives the cell cycle, subsequently promoting GBM tumor progression. In addition, our Kaplan-Meier survival estimates validated the poor patient survival associated with an overexpression of these genes in GBM. We used in silico molecular docking to analyze and validate our objective to repurpose Dapagliflozin against CDK1/PBK/CHEK1. Our results showed that Dapagliflozin forms putative conventional hydrogen bonds with CDK1, PBK, and CHEK1 and arrests the cell cycle with the lowest energies as Abemaciclib.
Collapse
Affiliation(s)
- Harold A. Chinyama
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan;
| | - Li Wei
- Department of Neurosurgery, Wan Fang Hospital, Taipei Medical University, No.111, Sec. 3, Xinglong Rd., Taipei 11696, Taiwan;
- Taipei Neuroscience Institute, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Injury Prevention and Control, College of Public Health, Taipei Medical University, Taipei 11031, Taiwan
| | - Ntlotlang Mokgautsi
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11031, Taiwan;
- Graduate Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Bashir Lawal
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15232, USA;
| | - Alexander T. H. Wu
- PhD Program of Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Clinical Research Center, Taipei Medical University Hospital, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 11490, Taiwan
| | - Hsu-Shan Huang
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11031, Taiwan;
- Graduate Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- School of Pharmacy, National Defense Medical Center, Taipei 11490, Taiwan
- PhD Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|