1
|
Reshadmanesh T, Mohebi R, Behnoush AH, Reshadmanesh A, Khalaji A, Norouzi M, Javanmardi E, Pishdad R, Jafarzadeh SR, Ghondaghsaz E, Chaparro S. The effects of sodium-glucose cotransporter-2 inhibitors in chemotherapy-induced cardiotoxicity and mortality in patients with cancer: a systematic review and meta-analysis. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2025; 11:50. [PMID: 40426171 DOI: 10.1186/s40959-025-00343-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 05/01/2025] [Indexed: 05/29/2025]
Abstract
BACKGROUND The effects of sodium-glucose cotransporter-2 (SGLT2) inhibitors on reducing cardiovascular events in different subgroups of diabetic patients are under investigation. The current systematic review and meta-analysis investigated the effects of SGLT2 inhibitors on preventing cardiovascular events and mortality and their adverse events in patients with active cancer and diabetes undergoing cardiotoxic cancer treatment. METHODS We searched PubMed, Embase, Web of Science, and Scopus to find studies investigating the effects of SGLT2 inhibitors on patients with diabetes and confirmed cancer until 19 August 2024. Meta-analyses were conducted using the random-effects model to compare all-cause mortality, cancer-associated mortality, heart failure (HF) hospitalization, arrhythmia, and adverse event rates such as ketoacidosis, hypoglycemia, urinary tract infection, and sepsis between patients with or without SGLT2 inhibitors use. Risk ratios (RRs) with 95% confidence intervals (CI) were used to compare outcomes between SGLT2 inhibitors and non-SGLT2 inhibitors groups. RESULTS Eleven studies were included with 88,096 patients with confirmed cancer (49% male). Among the total population, 20,538 received SGLT2 inhibitors (age 61.68 ± 10.71), while 67,558 did not receive SGLT2 inhibitors (age 68.24 ± 9.48). The meta-analysis found that the patients who received SGLT2 inhibitors had a significantly lower mortality rate than those who did not receive SGLT2 inhibitors (RR 0.46, 95% CI 0.34 to 0.63, p-value < 0.0001). Similarly, the cancer-associated mortality rate was also lower (RR 0.29, 95% CI 0.27 to 0.30, p-value < 0.0001). Further analysis found that the SGLT2 inhibitor group had a lower rate of HF hospitalization, compared to controls (RR 0.44, 95% CI 0.27 to 0.70, p-value = 0.0007). Moreover, patients receiving SGLT2 inhibitors had a statistically lower rate of arrhythmia (RR 0.38, 95% CI 0.26 to 0.56, p-value < 0.0001). Finally, patients in the SGLT2 inhibitors group had a lower rate of adverse events (RR 0.51, 95% CI 0.42 to 0.62, p-value < 0.0001). CONCLUSIONS SGLT2 inhibitors are effective in reducing mortality (all-cause and cancer-associated), HF hospitalization, arrhythmia, and drug adverse events in patients with cancer. If confirmed in future studies, these agents could be a potentially ideal candidate to prevent cardiotoxicity of cancer therapies.
Collapse
Affiliation(s)
- Tara Reshadmanesh
- School of Medicine, Zanjan University of Medical Science, Zanjan, Iran
| | - Reza Mohebi
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Amir Hossein Behnoush
- School of Medicine, Tehran University of Medical Sciences, Poursina St., Keshavarz Blvd, Tehran, 1417613151, Iran.
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Azadeh Reshadmanesh
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Amirmohammad Khalaji
- School of Medicine, Tehran University of Medical Sciences, Poursina St., Keshavarz Blvd, Tehran, 1417613151, Iran
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mitra Norouzi
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Elmira Javanmardi
- School of Medicine, Zanjan University of Medical Science, Zanjan, Iran
| | - Reza Pishdad
- Division of Endocrinology, Diabetes, and Metabolism, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - S Reza Jafarzadeh
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Elina Ghondaghsaz
- Undergraduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
| | - Sandra Chaparro
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
- Miami Cardiac and Vascular Institute, Baptist Health South Florida, Miami, FL, USA
| |
Collapse
|
2
|
Greco A, Canale ML, Quagliariello V, Oliva S, Tedeschi A, Inno A, De Biasio M, Bisceglia I, Tarantini L, Maurea N, Navazio A, Corda M, Iacovoni A, Colivicchi F, Grimaldi M, Oliva F. SGLT2 Inhibitors in Cancer Patients: A Comprehensive Review of Clinical, Biochemical, and Therapeutic Implications in Cardio-Oncology. Int J Mol Sci 2025; 26:4780. [PMID: 40429921 DOI: 10.3390/ijms26104780] [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: 04/09/2025] [Revised: 05/08/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Patients with active cancer and cancer survivors are at a markedly increased risk for developing cardiovascular comorbidities, including heart failure, coronary artery disease, and renal dysfunction, which are often compounded by the cardiotoxic effects of cancer therapies. This heightened cardiovascular vulnerability underscores the urgent need for effective, safe, and evidence-based cardioprotective strategies to reduce both cardiovascular morbidity and mortality. Sodium-glucose cotransporter 2 inhibitors (SGLT2is), a class of drugs originally developed for the treatment of type 2 diabetes, have demonstrated significant cardiovascular and renal benefits in high-risk populations, independent of glycemic control. Among the currently available SGLT2i, such as empagliflozin, canagliflozin, dapagliflozin, and sotagliflozin, there is growing evidence supporting their role in reducing major adverse cardiovascular events (MACEs), hospitalization for heart failure, and the progression of chronic kidney disease. Recent preclinical and clinical data suggest that SGLT2is exert cardioprotective effects through multiple mechanisms, including the modulation of inflammasome activity, specifically by reducing NLRP3 inflammasome activation and MyD88-dependent signaling, which are critical drivers of cardiac inflammation and fibrosis. Moreover, SGLT2is have been shown to enhance mitochondrial viability in cardiac cells, promoting improved cellular energy metabolism and function, thus mitigating cardiotoxicity. This narrative review critically evaluates the emerging evidence on the cardiorenal protective mechanisms of SGLT2is, with a particular focus on their potential role in cardio-oncology. We explore the common pathophysiological pathways between cardiovascular dysfunction and cancer, the molecular rationale for the use of SGLT2is in cancer patients, and the potential benefits in both primary and secondary prevention of cardiovascular toxicity related to oncological treatments. The aim is to propose a therapeutic paradigm utilizing SGLT2is to reduce cardiovascular mortality, MACE, and the burden of cardiotoxicity in high-risk oncology patients, fostering an integrated approach to cardio-oncology care.
Collapse
Affiliation(s)
- Alessandra Greco
- Cardiology Division, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Maria Laura Canale
- Cardiology, Versilia Hospital, Azienda USL Toscana Nord-Ovest, 55041 Lido di Camaiore, Italy
| | - Vincenzo Quagliariello
- Cardiology Division, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, 80145 Naples, Italy
| | - Stefano Oliva
- UOSD Cardiologia di interesse oncologico, IRCCS Istituto Tumori "Giovanni Paolo II", 70124 Bari, Italy
| | - Andrea Tedeschi
- Cardiology, "Guglielmo da Saliceto" Hospital, 29121 Piacenza, Italy
| | - Alessandro Inno
- Medical Oncology, IRCCS Ospedale Sacro Cuore Don Calabria, Negrar di Valpolicella, 37024 Verona, Italy
| | - Marzia De Biasio
- Cardiology, Azienda Sanitaria Universitaria Friuli Centrale, 33100 Udine, Italy
| | - Irma Bisceglia
- Servizi Cardiologici Integrati, Dipartimento di Scienze Cardio-Toraco-Vascolari, Azienda Ospedaliera San Camillo Forlanini, 00148 Rome, Italy
| | - Luigi Tarantini
- S.O.C. Cardiologia Ospedaliera, Presidio Ospedaliero Arcispedale Santa Maria Nuova, Azienda USL di Reggio Emilia-IRCCS, 42100 Reggio Emilia, Italy
| | - Nicola Maurea
- Cardiology Division, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, 80145 Naples, Italy
| | - Alessandro Navazio
- S.O.C. Cardiologia Ospedaliera, Presidio Ospedaliero Arcispedale Santa Maria Nuova, Azienda USL di Reggio Emilia-IRCCS, 42100 Reggio Emilia, Italy
| | - Marco Corda
- S.C. Cardiologia, Azienda di Rilievo Nazionale e Alta Specializzazione "G. Brotzu", 09047 Cagliari, Italy
| | - Attilio Iacovoni
- SSD Chirurgia dei Trapianti e del Trattamento Chirurgico dello Scompenso, Dipartimento Cardiovascolare, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy
| | - Furio Colivicchi
- UOC Cardiologia Clinica e Riabilitativa, Presidio Ospedaliero San Filippo Neri-ASL Roma 1, 00161 Rome, Italy
| | - Massimo Grimaldi
- UOC Cardiologia-UTIC, Ospedale Miulli, Acquaviva delle Fonti (BA), 70021 Bari, Italy
| | - Fabrizio Oliva
- Cardiologia 1-Emodinamica, Dipartimento Cardiotoracovascolare "A. De Gasperis", ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy
- Presidente ANMCO-Associazione Nazionale Medici Cardiologi Ospedalieri, 50121 Florence, Italy
- Consigliere Delegato per la Ricerca Fondazione per il Tuo Cuore-Heart Care Foundation, 50121 Florence, Italy
| |
Collapse
|
3
|
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; 1871:167898. [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
|
4
|
Goje ID, Goje GI, Ordodi VL, Ciobotaru VG, Ivan VS, Buzaș R, Tunea O, Bojin F, Lighezan DF. Doxorubicin-Induced Cardiotoxicity and the Emerging Role of SGLT2 Inhibitors: From Glycemic Control to Cardio-Oncology. Pharmaceuticals (Basel) 2025; 18:681. [PMID: 40430500 DOI: 10.3390/ph18050681] [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: 04/07/2025] [Revised: 04/27/2025] [Accepted: 05/01/2025] [Indexed: 05/29/2025] Open
Abstract
Cancer remains the second leading cause of death worldwide. Doxorubicin (DOX) is a cornerstone of hematologic malignancy treatment, but it is limited by its dose-dependent cardiotoxicity, leading to systolic and diastolic cardiac dysfunction and, ultimately, dilated hypokinetic cardiomyopathy. Cardio-oncology has emerged as a subspecialty addressing cardiovascular complications in cancer patients, highlighting preventive and therapeutic strategies to reduce cancer therapy-related cardiac dysfunction (CTRCD). Current approaches, including beta-blockers, renin-angiotensin system (RAS) inhibitors, and statins, offer partial cardioprotection. Sodium-glucose cotransporter-2 (SGLT2) inhibitors, initially developed for type 2 diabetes mellitus (T2DM), demonstrate pleiotropic cardioprotective effects beyond glycemic control, including reduced oxidative stress, inflammation, and myocardial remodeling. This review explores the interplay between anthracycline therapy, particularly DOX, and cardiotoxicity while evaluating SGLT2 inhibitors as novel agents in cardio-oncology. Preclinical studies suggest SGLT2 inhibitors attenuate CTRCD by preserving mitochondrial function and inhibiting apoptosis, while clinical trials highlight their efficacy in reducing heart failure (HF) hospitalizations and cardiovascular (CV) mortality. Integrating SGLT2 inhibitors into cardio-oncology protocols could revolutionize the management of CTRCD, enhancing patient outcomes in oncology and cardiovascular care. Considering the emerging evidence, SGLT2 inhibitors may provide significant benefits to patients undergoing anthracycline therapy, particularly those with elevated cardiovascular risk profiles. We recommend that future prospective, large-scale clinical trials further evaluate the efficacy and safety of these agents as cardioprotective therapy to optimize individualized treatment strategies.
Collapse
Affiliation(s)
- Iacob-Daniel Goje
- Department of Medical Semiology I, "Victor Babes" University of Medicine and Pharmacy, No. 2 Eftimie Murgu Square, 300041 Timisoara, Romania
- Advanced Cardiology and Hemostaseology Research Center, "Victor Babes" University of Medicine and Pharmacy, No. 2 Eftimie Murgu Square, 300041 Timisoara, Romania
| | - Greta-Ionela Goje
- Advanced Cardiology and Hemostaseology Research Center, "Victor Babes" University of Medicine and Pharmacy, No. 2 Eftimie Murgu Square, 300041 Timisoara, Romania
- Department I of Nursing, University Clinic of Clinical Skills, "Victor Babes" University of Medicine and Pharmacy, No. 2 Eftimie Murgu Square, 300041 Timisoara, Romania
| | - Valentin Laurențiu Ordodi
- Faculty of Industrial Chemistry and Environmental Engineering, "Politehnica" University Timisoara, No. 2 Victoriei Square, 300006 Timisoara, Romania
- Center for Gene and Cellular Therapies in the Treatment of Cancer Timisoara-OncoGen, Clinical Emergency County Hospital "Pius Brinzeu" Timisoara, No. 156 Liviu Rebreanu, 300723 Timisoara, Romania
- Department of Functional Sciences, Immuno-Physiology and Biotechnologies Center, "Victor Babes" University of Medicine and Pharmacy, No. 2 Eftimie Murgu Square, 300041 Timisoara, Romania
| | - Valentina Gabriela Ciobotaru
- Department of Medical Semiology I, "Victor Babes" University of Medicine and Pharmacy, No. 2 Eftimie Murgu Square, 300041 Timisoara, Romania
- Advanced Cardiology and Hemostaseology Research Center, "Victor Babes" University of Medicine and Pharmacy, No. 2 Eftimie Murgu Square, 300041 Timisoara, Romania
| | - Vlad Sabin Ivan
- Department of Medical Semiology I, "Victor Babes" University of Medicine and Pharmacy, No. 2 Eftimie Murgu Square, 300041 Timisoara, Romania
- Advanced Cardiology and Hemostaseology Research Center, "Victor Babes" University of Medicine and Pharmacy, No. 2 Eftimie Murgu Square, 300041 Timisoara, Romania
| | - Roxana Buzaș
- Department of Medical Semiology I, "Victor Babes" University of Medicine and Pharmacy, No. 2 Eftimie Murgu Square, 300041 Timisoara, Romania
- Advanced Cardiology and Hemostaseology Research Center, "Victor Babes" University of Medicine and Pharmacy, No. 2 Eftimie Murgu Square, 300041 Timisoara, Romania
| | - Oana Tunea
- Department of Medical Semiology I, "Victor Babes" University of Medicine and Pharmacy, No. 2 Eftimie Murgu Square, 300041 Timisoara, Romania
- Advanced Cardiology and Hemostaseology Research Center, "Victor Babes" University of Medicine and Pharmacy, No. 2 Eftimie Murgu Square, 300041 Timisoara, Romania
| | - Florina Bojin
- Center for Gene and Cellular Therapies in the Treatment of Cancer Timisoara-OncoGen, Clinical Emergency County Hospital "Pius Brinzeu" Timisoara, No. 156 Liviu Rebreanu, 300723 Timisoara, Romania
- Department of Functional Sciences, Immuno-Physiology and Biotechnologies Center, "Victor Babes" University of Medicine and Pharmacy, No. 2 Eftimie Murgu Square, 300041 Timisoara, Romania
| | - Daniel-Florin Lighezan
- Department of Medical Semiology I, "Victor Babes" University of Medicine and Pharmacy, No. 2 Eftimie Murgu Square, 300041 Timisoara, Romania
- Advanced Cardiology and Hemostaseology Research Center, "Victor Babes" University of Medicine and Pharmacy, No. 2 Eftimie Murgu Square, 300041 Timisoara, Romania
| |
Collapse
|
5
|
Wang Y, Li W, Li C, Zhou T, Herrmann J, Wang W. New insights into therapeutic strategies, drugs, and targets for advancing cancer therapy-related cardiovascular toxicity. Sci Bull (Beijing) 2025; 70:991-993. [PMID: 40016034 DOI: 10.1016/j.scib.2025.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Affiliation(s)
- Yong Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100029, China; School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Weili Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Chun Li
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Tingting Zhou
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Naval Medical University, Shanghai 200433, China
| | - Joerg Herrmann
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55902, USA.
| | - Wei Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| |
Collapse
|
6
|
Hu WS, Lin CL. Beneficial effect of sodium-glucose cotransporter-2 inhibitors on mortality among patients with cancer and diabetes mellitus. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:4355-4360. [PMID: 39466440 DOI: 10.1007/s00210-024-03553-2] [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: 06/11/2024] [Accepted: 10/18/2024] [Indexed: 10/30/2024]
Abstract
The work attempts to describe mortality outcomes of sodium-glucose cotransporter-2 inhibitor (SGLT2I) in patients with diabetes mellitus (DM) and cancer. Using Taiwan's National Health Insurance Research Database to analyze the prognosis of cancer patients with coexisting DM, comparing those receiving SGLT2I with those who do not. After index-year and matching (age, sex, some comorbidities and medications), we obtain two groups of 20,339 patients. After further adjustment for age, sex and comorbidities, those with SGLT2I had a lower adjusted hazard ratio of mortality (aHR: 0.64 [95% CI: 0.60-0.68]). We conclude that SGLT2I medication should be considered first choice in patients suffering from DM and cancer.
Collapse
Affiliation(s)
- Wei-Syun Hu
- School of Medicine, College of Medicine, China Medical University, Taichung, 40402, Taiwan.
- Division of Cardiovascular Medicine, Department of Medicine, China Medical University Hospital, 2, Yuh-Der Road, Taichung, 40447, Taiwan.
| | - Cheng-Li Lin
- Management Office for Health Data, China Medical University Hospital, Taichung, 40447, Taiwan
| |
Collapse
|
7
|
Nicol M, Deniau B, Rahli R, Genest M, Polidano E, Assad N, Samuel JL, Mebazaa A, Solal AC, Azibani F. Streptozotocin-induced hyperglycemia unmasks cardiotoxicity induced by doxorubicin. Sci Rep 2025; 15:8104. [PMID: 40057546 PMCID: PMC11890863 DOI: 10.1038/s41598-025-91824-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 02/24/2025] [Indexed: 05/13/2025] Open
Abstract
Late-onset cardiotoxicity induced by anthracyclines occurs years to decades after completion of anti-cancer therapy and is associated with increased morbi-mortality of cancer survivors. Chemotherapy at the time of treatment probably causes cardiac damages for which the juvenile heart compensate. Co-morbidities happening in the adulthood such as type 1 diabetes (DT1), affect the heart and thus can unmask chemotherapy induced cardiotoxicity. To prove our hypothesis, we induced hyperglycemia [Streptozotocin treatment (STZ), 50 mg/kg/day for 5 days] in 11 weeks old mice who previously received doxorubicin treatment (Dox, 3 mg/kg) when they were six-weeks old. Interestingly, streptozotocin-induced hyperglycemia in Dox-pretreated mice (Dox-STZ) induced a higher mortality (p < 0.05) and more severe cardiac dysfunction (p < 0.0001) when compared with mice receiving Dox or STZ alone. Apoptosis evaluated by caspase 3 protein expression and Bax/Bcl2 genes expression was higher in Dox-STZ mice compared to STZ or Dox alone. While Dox and STZ independently induced capillary rarefaction, cardiomyocytes atrophy was only induced by STZ. Furthermore, Sirius-red staining of cardiac sections showed higher fibrosis levels (p < 0.0001) in Dox-STZ compared to Dox or STZ alone. All together, these results demonstrate that STZ precipitates and unmask cardiac dysfunction in previously treated Dox animals.
Collapse
Affiliation(s)
- Martin Nicol
- Inserm UMR-S 942 MASCOT, University of Paris Cité, Lariboisière Hospital, Paris, France
- Cardiology Department, Lariboisière Hospital, University of Paris, Paris, France
| | - Benjamin Deniau
- Inserm UMR-S 942 MASCOT, University of Paris Cité, Lariboisière Hospital, Paris, France
- Department of Anesthesiology and Intensive Care, Lariboisière - Saint Louis Hospitals, APHP, Paris, France
| | - Roza Rahli
- Inserm UMR-S 942 MASCOT, University of Paris Cité, Lariboisière Hospital, Paris, France
| | - Magali Genest
- Inserm UMR-S 942 MASCOT, University of Paris Cité, Lariboisière Hospital, Paris, France
| | - Evelyne Polidano
- Inserm UMR-S 942 MASCOT, University of Paris Cité, Lariboisière Hospital, Paris, France
| | - Noma Assad
- Inserm UMR-S 942 MASCOT, University of Paris Cité, Lariboisière Hospital, Paris, France
| | - Jane-Lise Samuel
- Inserm UMR-S 942 MASCOT, University of Paris Cité, Lariboisière Hospital, Paris, France
| | - Alexandre Mebazaa
- Inserm UMR-S 942 MASCOT, University of Paris Cité, Lariboisière Hospital, Paris, France
- Department of Anesthesiology and Intensive Care, Lariboisière - Saint Louis Hospitals, APHP, Paris, France
| | - Alain Cohen Solal
- Inserm UMR-S 942 MASCOT, University of Paris Cité, Lariboisière Hospital, Paris, France
- Cardiology Department, Lariboisière Hospital, University of Paris, Paris, France
| | - Feriel Azibani
- Inserm UMR-S 942 MASCOT, University of Paris Cité, Lariboisière Hospital, Paris, France.
| |
Collapse
|
8
|
Lin J, Li B, Guo X, Li G, Zhang Q, Wang W. Key Mechanisms of Oxidative Stress-Induced Ferroptosis in Heart Failure with Preserved Ejection Fraction and Potential Therapeutic Approaches. Rev Cardiovasc Med 2025; 26:26613. [PMID: 40160560 PMCID: PMC11951494 DOI: 10.31083/rcm26613] [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: 09/18/2024] [Revised: 12/02/2024] [Accepted: 12/10/2024] [Indexed: 04/02/2025] Open
Abstract
The prevalence of heart failure with preserved ejection fraction (HFpEF) is increasing annually, particularly among patients with metabolic disorders such as hypertension and diabetes. However, there is currently no treatment capable of altering the natural course of HFpEF. Recently, the interplay between oxidative stress and ferroptosis in cardiovascular diseases has drawn extensive attention; however, minimal research has been published on the mechanisms of oxidative stress and ferroptosis in HFpEF. This paper reviews the relevant mechanisms through which oxidative stress is induced and promotes ferroptosis during the development of HFpEF. The review also explores more efficacious treatment approaches for HFpEF by inhibiting oxidative stress and ferroptosis, thereby offering a theoretical foundation for verifying the feasibility of these methods for further research. As tumor-targeted therapy progresses, the survival period of tumor patients is prolonged, and cardiovascular events have gradually emerged as one of the most crucial causes of death among tumor patients. Hence, inhibiting the vascular endothelial growth factor (VEGF) pathway has become a major target in tumor treatment, significantly enhancing patient survival. Nevertheless, secondary cardiovascular complications and events, such as myocardial injury and subsequent heart failure, have severely impacted patient survival and quality of life. Therefore we have also explored the potential mechanism through which novel targeted anti-cancer drugs induce HFpEF via ferroptosis. Additionally, we reviewed the specific modes of action for preventing and treating HFpEF without influencing their anti-cancer therapeutic effect.
Collapse
Affiliation(s)
- Junling Lin
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, 313000 Huzhou, Zhejiang, China
| | - Bingtao Li
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, 313000 Huzhou, Zhejiang, China
| | - Xueqi Guo
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, 313000 Huzhou, Zhejiang, China
| | - Guodong Li
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, 313000 Huzhou, Zhejiang, China
| | - Qi Zhang
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, 313000 Huzhou, Zhejiang, China
| | - Wenjuan Wang
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, 313000 Huzhou, Zhejiang, China
| |
Collapse
|
9
|
Simela C, Walker JM, Ghosh AK, Chen DH. SGLT2 inhibitors for prevention and management of cancer treatment-related cardiovascular toxicity: a review of potential mechanisms and clinical insights. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2025; 11:15. [PMID: 39934910 DOI: 10.1186/s40959-024-00284-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 10/30/2024] [Indexed: 02/13/2025]
Abstract
More evidence-based strategies are needed for preventing and managing cancer treatment-related cardiovascular toxicity (CTR-CVT). Owing to the growing body of evidence supporting their cardioprotective role in several cardiac injury scenarios, sodium-glucose cotransporter 2 inhibitors (SGLT2i) may be beneficial for preventing and treating CTR-CVT. In October 2024, a search was conducted of the PubMed database to review full studies investigating the cardioprotective role of SGLT2i against CTR-CVT. We identified 44 full published/pre-print studies and 3 ongoing randomised controlled trial across eight types of cancer treatment (anthracyclines, platinum-containing therapy, immune checkpoint inhibitors, HER2-targeted therapies, kinase inhibitors, androgen deprivation therapies, multiple myeloma therapies and 5-fluorouracil). Most studies used animal models and focussed on primary prevention. 43 of the 44 studies found some cardioprotective effect of SGLT2i against CTR-CVT, which in some cases included preventing ejection fraction decline and aberrations in cardiac electrophysiological parameters. Some studies also observed beneficial effects on mortality. A central triad of anti-inflammatory, anti-oxidative and anti-apoptotic mechanisms likely underlie SGLT2i-mediated cardioprotection against CTR-CVT. Overall, this growing body of research suggests that SGLT2i may be a promising candidate for preventing CTR-CVT either as monotherapy or in combination with other cardioprotective drugs. However, the literature is limited in that no prospective randomised controlled trials investigating SGLT2i for the prevention and management of CTR-CVT exist and most existing human retrospective data is based on diabetic populations. Future work must focus on addressing these limitations of the current literature.
Collapse
Affiliation(s)
- Carl Simela
- University College London Hospital, London, UK
| | - J Malcolm Walker
- University College London Hospital, London, UK
- Hatter Cardiovascular Institute, University College London, London, UK
| | - Arjun K Ghosh
- University College London Hospital, London, UK
- Hatter Cardiovascular Institute, University College London, London, UK
- Barts Heart Centre, London, UK
| | - Daniel H Chen
- University College London Hospital, London, UK.
- Hatter Cardiovascular Institute, University College London, London, UK.
- Barts Heart Centre, London, UK.
- Prince of Wales Hospital, Sydney, NSW, Australia.
- St George Hospital, Sydney NSW, Australia.
| |
Collapse
|
10
|
Balough E, Ariza A, Asnani A, Hoeger CW. Cardiotoxicity of Anthracyclines. Cardiol Clin 2025; 43:111-127. [PMID: 39551553 DOI: 10.1016/j.ccl.2024.08.002] [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] [Indexed: 11/19/2024]
Abstract
Anthracycline chemotherapy is associated with cardiotoxicity, predominantly manifesting as left ventricular systolic dysfunction within the first year of treatment. Early detection is possible through biomarkers and cardiovascular imaging before clinical symptoms develop. Comprehensive cardiovascular risk assessment is essential for all patients prior to anthracycline therapy to stratify their risk of cardiotoxicity. Preventive measures, including cardiovascular risk optimization, as well as anthracycline dose adjustments, the use of liposomal anthracyclines, and dexrazoxane in high-risk patients, are crucial to mitigate the risk of cardiotoxicity. Long-term follow-up and cardiovascular risk optimization are critical for cancer survivors to optimize cardiovascular outcomes.
Collapse
Affiliation(s)
- Elizabeth Balough
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, 185 Pilgrim Road, Baker 4, Boston, MA 02215, USA. https://twitter.com/ElizabethBaloug
| | - Abul Ariza
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; 3 Blackfan Circle, CLS-911, Boston, MA 02115, USA
| | - Aarti Asnani
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; 3 Blackfan Circle, CLS-911, Boston, MA 02115, USA. https://twitter.com/AartiAsnaniMD
| | - Christopher W Hoeger
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, 185 Pilgrim Road, Baker 4, Boston, MA 02215, USA.
| |
Collapse
|
11
|
Camilli M, Viscovo M, Maggio L, Bonanni A, Torre I, Pellegrino C, Lamendola P, Tinti L, Teofili L, Hohaus S, Lanza GA, Ferdinandy P, Varga Z, Crea F, Lombardo A, Minotti G. Sodium-glucose cotransporter 2 inhibitors and the cancer patient: from diabetes to cardioprotection and beyond. Basic Res Cardiol 2025; 120:241-262. [PMID: 38935171 PMCID: PMC11790819 DOI: 10.1007/s00395-024-01059-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 05/18/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024]
Abstract
Sodium-glucose cotransporter 2 inhibitors (SGLT2i), a new drug class initially designed and approved for treatment of diabetes mellitus, have been shown to exert pleiotropic metabolic and direct cardioprotective and nephroprotective effects that extend beyond their glucose-lowering action. These properties prompted their use in two frequently intertwined conditions, heart failure and chronic kidney disease. Their unique mechanism of action makes SGLT2i an attractive option also to lower the rate of cardiac events and improve overall survival of oncological patients with preexisting cardiovascular risk and/or candidate to receive cardiotoxic therapies. This review will cover biological foundations and clinical evidence for SGLT2i modulating myocardial function and metabolism, with a focus on their possible use as cardioprotective agents in the cardio-oncology settings. Furthermore, we will explore recently emerged SGLT2i effects on hematopoiesis and immune system, carrying the potential of attenuating tumor growth and chemotherapy-induced cytopenias.
Collapse
Affiliation(s)
- Massimiliano Camilli
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy.
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli, 1, 00168, Rome, Italy.
| | - Marcello Viscovo
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Luca Maggio
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli, 1, 00168, Rome, Italy
| | - Alice Bonanni
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli, 1, 00168, Rome, Italy
| | - Ilaria Torre
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli, 1, 00168, Rome, Italy
| | - Claudio Pellegrino
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Priscilla Lamendola
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | - Lorenzo Tinti
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli, 1, 00168, Rome, Italy
| | - Luciana Teofili
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Stefan Hohaus
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Gaetano Antonio Lanza
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli, 1, 00168, Rome, Italy
| | - Peter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
- MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Zoltan Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Filippo Crea
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Center of Excellence of Cardiovascular Sciences, Ospedale Isola Tiberina - Gemelli Isola, Rome, Italy
| | - Antonella Lombardo
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli, 1, 00168, Rome, Italy
| | | |
Collapse
|
12
|
Luo J, He M, Liang C, Huang X, Zhu Y, Hu D, Yan J, Li M, Lin H, Liao W, Bin J, Guan Z, Zheng C, Liao Y. Canagliflozin reverses doxorubicin-induced cardiotoxicity via restoration of autophagic homeostasis. Toxicol Appl Pharmacol 2025; 495:117183. [PMID: 39631538 DOI: 10.1016/j.taap.2024.117183] [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: 09/10/2024] [Revised: 11/18/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
Sodium-glucose cotransporter 2 inhibitors (SGLT2i) have been reported as successful for preventing doxorubicin (DOX) -induced cardiotoxicity (DIC), but the underlying mechanisms are elusive. This study aimed to determine whether canagliflozin, an SGLT2i, protects against DIC by regulation of autophagic flux in cardiomyocytes through a mechanism independent of SGLT2. The differentially expressed autophagy-related genes (ARGs) in DIC were analyzed. Neonatal rat cardiomyocytes (NRCMs), H9C2 rat cardiomyocytes or C57BL/6 mice were treated with canagliflozin or vehicle. The effects on cellular apoptosis and autophagy were investigated using qRT-PCR, western blotting and immunofluorescence. Additionally, cardiac function, myocardial fibrosis, and apoptosis of cardiomyocytes were also assessed in mice. The potential molecular targets of canagliflozin were identified through molecular docking analysis. A total of 26 differentially expressed ARGs were identified. Canagliflozin significantly activated autophagic flux and inhibited apoptosis of cardiomyocytes in both DOX-treated H9C2 rat cardiomyocytes and NRCMs. In a murine model of DIC, canagliflozin improved cardiac dysfunction by suppressing cardiac remodeling, fibrosis, and apoptosis. Moreover, canagliflozin promoted autophagy by enhancing SIRT1 levels and inhibiting the PI3K/Akt/mTOR signaling pathway. Immunofluorescence assays revealed that canagliflozin promoted the translocation of LC3 from the nucleus to the cytoplasm. Molecular docking analysis confirmed that canagliflozin has high affinity for targets associated with DIC. These findings suggest that canagliflozin protects cardiomyocytes from DOX-induced cell death by activating SIRT1, inhibiting the PI3K/Akt/mTOR pathway, and enhancing autophagic flux.
Collapse
Affiliation(s)
- Jianping Luo
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Cardiology, Ganzhou People's Hospital, Ganzhou, China
| | - Mingyuan He
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Changzhu Liang
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoxia Huang
- Cardiovascular Center, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, Guangdong, China
| | - Yingqi Zhu
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Donghong Hu
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Junyu Yan
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mingjue Li
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hairuo Lin
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jianping Bin
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou, China; Cardiovascular Center, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, Guangdong, China
| | - Ziyun Guan
- Cardiovascular Center, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, Guangdong, China
| | - Cankun Zheng
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Yulin Liao
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou, China; Cardiovascular Center, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, Guangdong, China.
| |
Collapse
|
13
|
Sanjari M, Hadavizadeh M, Sadeghi N, Naghibzadeh-Tahami A. Effect of empagliflozin on weight in patients with prediabetes and diabetes. Sci Rep 2025; 15:118. [PMID: 39748045 PMCID: PMC11696924 DOI: 10.1038/s41598-024-83820-7] [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: 05/12/2024] [Accepted: 12/17/2024] [Indexed: 01/04/2025] Open
Abstract
The impact of blood glucose-lowering medications on weight has always been a topic of interest in the treatment of diabetic patients. This study investigates the effect of empagliflozin on weight in patients with prediabetes and type 2 diabetes. This quasi-experimental study was performed on patients with prediabetes or type 2 diabetes with an HbA1c level up to 1% higher than the treatment target, and not using other blood glucose-lowering medications. Patients received 10 milligrams of Empagliflozin once daily for three months, and weight, BMI, waist circumference, and blood pressure were evaluated monthly. Forty-three patients (21 women and 22 men) enrolled. The average weight of patients decreased by 2.96 ± 1.96 kg (3.8%) (P < 0.001). BMI decreased by -1.10 ± 0.71 Kg/m2 (3.72%) (P < 0.001). The waist circumference of patients decreased by -3.23 ± 3.69 centimeters (P < 0.001). FPG decreased from 114.86 to 109.48 mg/dL (P < 0.001), and HbA1c decreased from 6.52% to 6.38% (P < 0.001). The weight change was more significant in men than women (-3.59 ± 1.74 vs. -2.30. ± 1.99), (P = 0.029). Weight reduction was greater in patients with GFR higher than 90 compared to GFR lower than 90 (-3.34. ± 2.00 vs. -2.16 ± 1.67) (P = 0.063). Also, no significant difference was observed in the weight, BMI, and waist circumference changes between different BMI groups (less than 25, 25 to 30, 30 to 35, and higher than 35). The trend of weight and BMI changes during the three-month empagliflozin treatment period was significant (P < 0.001) and didn't reach a plateau level after three months. The change in waist circumference was also significant, reaching a plateau level after one month (P < 0.001). There was no significant change in blood pressure. In conclusion the weight, BMI, and waist circumference of patients decreased following the administration of empagliflozin. Weight reduction was more pronounced in males than females and in patients with a GFR above 90 than those with a GFR below 90. Trial registration: This study was approved by the Research Deputy of Kerman University of Medical Sciences with tracking number 401,000,516, IRCT code: IRCT20090317001774N10, and ethical code: IR.KMU.AH.REC.1402.065 was approved by Research Ethics Committee of Afzalipour Hospital- Kerman University of Medical Sciences.
Collapse
Affiliation(s)
- Mojgan Sanjari
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Hadavizadeh
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| | - Narges Sadeghi
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Ahmad Naghibzadeh-Tahami
- Health Services Management Research Center, Institute for Futures Studies in Health, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
14
|
Li W, Zhang Y, Wei Y, Ling G, Zhang Y, Li Y, Guo S, Tan N, Ma L, Li W, Sun Q, Wang W, Wang Y. New insights into mitochondrial quality control in anthracycline-induced cardiotoxicity: molecular mechanisms, therapeutic targets, and natural products. Int J Biol Sci 2025; 21:507-523. [PMID: 39781459 PMCID: PMC11705644 DOI: 10.7150/ijbs.103810] [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: 09/17/2024] [Accepted: 11/23/2024] [Indexed: 01/12/2025] Open
Abstract
Anthracyclines (ANTs) are widely used in cancer therapy, particularly for lymphoma, sarcoma, breast cancer, and childhood leukemia, and have become the cornerstone of chemotherapy for various malignancies. However, it is associated with fatal and dose-dependent cardiovascular complications, especially cardiotoxicity. Mitochondrial quality control mechanisms, encompassing mitophagy, mitochondrial dynamics, and mitochondrial biogenesis, maintain mitochondrial homeostasis in the cardiovascular system. Recent studies have highlighted that mitochondrial quality control mechanisms play considerable roles in ANTs-induced cardiotoxicity (AIC). In addition, natural products targeting mitochondrial quality control mechanisms have emerged as potential therapeutic strategies to alleviate AIC. This review summarizes the types, incidence, prevention, treatment, and pathomechanism of AIC, delves into the molecular mechanisms of mitochondrial quality control in the pathogenesis of AIC, and explores natural products that target these mechanisms, so as to provide potential targets and therapeutic drugs for address the clinical challenges in AIC prevention and treatment, where no effective medicines are available.
Collapse
Affiliation(s)
- Weili Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yuqin Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yan Wei
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Guanjing Ling
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yawen Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yilin Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Shujuan Guo
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Nannan Tan
- Anhui University of Traditional Chinese Medicine, Anhui 230012, China
| | - Lin Ma
- Anhui University of Traditional Chinese Medicine, Anhui 230012, China
| | - Wei Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Qianbin Sun
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Wei Wang
- Guangzhou University of Chinese Medicine, Guangzhou 510006, China
- Beijing Key Laboratory of TCM Syndrome and Formula, Beijing University of Chinese Medicine, Beijing 100029, China
- Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing 100029, China
| | - Yong Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100029, China
- Beijing Key Laboratory of TCM Syndrome and Formula, Beijing University of Chinese Medicine, Beijing 100029, China
- Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing 100029, China
| |
Collapse
|
15
|
Yuan X, Pan L, Zhang C, Zhu Q, Huang Z, Qin Y, Zhang G, Feng Z, Yang C, Hou N. Empagliflozin improves pressure-overload-induced cardiac hypertrophy by inhibiting the canonical Wnt/β-catenin signaling pathway. Front Pharmacol 2024; 15:1499542. [PMID: 39664517 PMCID: PMC11631586 DOI: 10.3389/fphar.2024.1499542] [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: 09/21/2024] [Accepted: 10/30/2024] [Indexed: 12/13/2024] Open
Abstract
Background Empagliflozin (EMPA) is an SGLT-2 inhibitor that can control hyperglycemia. Clinical trials have indicated its cardio-protective effects against cardiac remodeling in diabetes or non-diabetes patients. However, the underlying molecular mechanisms of EMPA's cardio-protective effects remain elusive. Methods We evaluated whether the EMPA attenuated the pressure-overload-induced cardiac hypertrophy by inhibiting the Wnt/β-catenin pathway. Furthermore, the effects of the EMPA on a mouse model of transverse aortic constriction (TAC) induced cardiac hypertrophy was also evaluated. Mice were administrated with 0.5% CMC-Na as a vehicle or EMPA (10 mg/kg/day, daily, throughout the study) by intragastric gavage. Results The in vivo echocardiography and histologic morphological analyses revealed that EMPA attenuated TAC-induced cardiac hypertrophy. Moreover, it also ameliorated TAC-induced cardiac fibrosis and decreased the cell size of the cardiomyocytes in isolated adult cardiomyocytes. Molecular mechanism analysis revealed that the EMPA reduced the TAC-induced enhanced expression of the Wnt/β-catenin pathway in vivo. For in vitro assessments, isolated neonatal rat cardiomyocytes (NRCMs) were treated with Angiotensin II (AngII) and EMPA; the results showed that in the absence of EMPA, the expression of the Wnt/β-catenin pathway was enhanced. In the trans-genetic heterozygous β-catenin deletion mice, EMPA attenuated TAC-induced cardiac remodeling by reducing the Wnt/β-catenin pathway. In addition, molecular docking analysis indicated that EMPA interacts with FZD4 to inhibit the TAC and AngII induced Wnt/β-catenin pathway in cardiomyocytes. Conclusion Our study illustrated that EMPA might directly interact with FZD4 to inhibit the TAC and AngII-induced activation of the Wnt/β-catenin pathway to attenuate the adverse cardiac remodeling.
Collapse
Affiliation(s)
- Xun Yuan
- The Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, China
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, Guangzhou Medical University, Guangzhou, China
| | - Li Pan
- Department of Physiology, School of Basic Medicine Sciences, Guangzhou Health Science College, Guangzhou, China
| | - Chi Zhang
- School of Stomatology, Guangzhou Medical University, Guangzhou, China
| | - Qiulian Zhu
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, Guangzhou Medical University, Guangzhou, China
| | - Zexin Huang
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, Guangzhou Medical University, Guangzhou, China
| | - Yuan Qin
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, Guangzhou Medical University, Guangzhou, China
| | - Guiping Zhang
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, Guangzhou Medical University, Guangzhou, China
| | - Zhimei Feng
- The Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, China
| | - Caixian Yang
- The Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, China
| | - Ning Hou
- The Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, China
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
16
|
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
|
17
|
Soares RR, Viggiani LF, Reis Filho JM, Joviano-Santos JV. Cardioprotection of Canagliflozin, Dapagliflozin, and Empagliflozin: Lessons from preclinical studies. Chem Biol Interact 2024; 403:111229. [PMID: 39244185 DOI: 10.1016/j.cbi.2024.111229] [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: 05/20/2024] [Revised: 08/12/2024] [Accepted: 09/04/2024] [Indexed: 09/09/2024]
Abstract
Clinical and preclinical studies have elucidated the favorable effects of Inhibitors of Sodium-Glucose Cotransporter-2 (iSGLT2) in patients and animal models with type 2 diabetes. Notably, these inhibitors have shown significant benefits in reducing hospitalizations and mortality among patients with heart failure. However, despite their incorporation into clinical practice for indications beyond diabetes, the decision-making process regarding their use often lacks a systematic approach. The selection of iSGLT2 remains arbitrary, with only a limited number of studies simultaneously exploring the different classes of them. Currently, no unique guideline establishes their application in both clinical and basic research. This review delves into the prevalent use of iSGLT2 in animal models previously subjected to induced cardiac stress. We have compiled key findings related to cardioprotection across various animal models, encompassing diverse dosages and routes of administration. Beyond their established role in diabetes management, iSGLT2 has demonstrated utility as agents for safeguarding heart health and cardioprotection can be class-dependent among the iSGLT2. These findings may serve as valuable references for other researchers. Preclinical studies play a pivotal role in ensuring the safety of novel compounds or treatments for potential human use. By assessing side effects, toxicity, and optimal dosages, these studies offer a robust foundation for informed decisions, identifying interventions with the highest likelihood of success and minimal risk to patients. The insights gleaned from preclinical studies, which play a crucial role in highlighting areas of knowledge deficiency, can guide the exploration of novel mechanisms and strategies involving iSGLT2.
Collapse
Affiliation(s)
- Rayla Rodrigues Soares
- Faculdade Ciências Médicas de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil; Laboratório de Investigações NeuroCardíacas, Ciências Médicas de Minas Gerais (LINC CMMG), Belo Horizonte, Minas Gerais, Brazil
| | - Larissa Freitas Viggiani
- Faculdade Ciências Médicas de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil; Laboratório de Investigações NeuroCardíacas, Ciências Médicas de Minas Gerais (LINC CMMG), Belo Horizonte, Minas Gerais, Brazil
| | - Juliano Moreira Reis Filho
- Post-Graduate Program in Health Sciences, Faculdade Ciências Médicas de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Julliane V Joviano-Santos
- Post-Graduate Program in Health Sciences, Faculdade Ciências Médicas de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil; Laboratório de Investigações NeuroCardíacas, Ciências Médicas de Minas Gerais (LINC CMMG), Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
18
|
Prosperi S, D’Amato A, Labbro Francia A, Monosilio S, Cestiè C, Marek Iannucci S, Netti L, Angotti D, Filomena D, Mariani MV, Myftari V, Germanò R, Cimino S, Mancone M, Badagliacca R, Maestrini V, Severino P, Vizza CD. Sodium-Glucose Cotransporter 2 Inhibitor Therapy in Different Scenarios of Heart Failure: An Overview of the Current Literature. Int J Mol Sci 2024; 25:11458. [PMID: 39519011 PMCID: PMC11546829 DOI: 10.3390/ijms252111458] [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: 08/31/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Heart failure (HF) is a complex syndrome that requires tailored and patient-centered treatment. Sodium-glucose cotransporter 2 inhibitors (SGLT2is) constitute one of the four pillars of the medical treatment of HF. However, the 2023 ESC guidelines treat HF as a single entity without making clear distinctions in phenotypes according to etiology. This creates a "gap in knowledge", causing much debate about the applicability of these drugs in peculiar clinical settings that are etiological and/or predisposing clinical conditions for HF. Furthermore, considering the variety of etiologies and different pathophysiological backgrounds of HF, one might question whether the use of SGLT2is is equally beneficial in all types of HF and whether certain drug-related properties may be exploited in different contexts. For example, SGLT2is can improve the metabolic and inflammatory state, which is fundamental in ischemic heart disease. Anti-inflammatory power can also play a paramount role in myocarditis or cardiotoxicity, while improving the congestive state and reducing filling pressure may be even more fundamental in restrictive heart disease or advanced heart disease. This review aims to gather the evidence currently present in the literature concerning the advantages or the disadvantages of using these drugs in these particular clinical settings, with the goal being an optimized and highly personalized treatment for HF.
Collapse
Affiliation(s)
- Silvia Prosperi
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (S.P.); (A.L.F.); (S.M.); (C.C.); (S.M.I.); (L.N.); (D.A.); (D.F.); (M.V.M.); (V.M.); (R.G.); (S.C.); (M.M.); (R.B.); (V.M.); (P.S.); (C.D.V.)
| | - Andrea D’Amato
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (S.P.); (A.L.F.); (S.M.); (C.C.); (S.M.I.); (L.N.); (D.A.); (D.F.); (M.V.M.); (V.M.); (R.G.); (S.C.); (M.M.); (R.B.); (V.M.); (P.S.); (C.D.V.)
- Department of Cardiology, Ospedale Fabrizio Spaziani, 03100 Frosinone, Italy
| | - Aurora Labbro Francia
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (S.P.); (A.L.F.); (S.M.); (C.C.); (S.M.I.); (L.N.); (D.A.); (D.F.); (M.V.M.); (V.M.); (R.G.); (S.C.); (M.M.); (R.B.); (V.M.); (P.S.); (C.D.V.)
| | - Sara Monosilio
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (S.P.); (A.L.F.); (S.M.); (C.C.); (S.M.I.); (L.N.); (D.A.); (D.F.); (M.V.M.); (V.M.); (R.G.); (S.C.); (M.M.); (R.B.); (V.M.); (P.S.); (C.D.V.)
| | - Claudia Cestiè
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (S.P.); (A.L.F.); (S.M.); (C.C.); (S.M.I.); (L.N.); (D.A.); (D.F.); (M.V.M.); (V.M.); (R.G.); (S.C.); (M.M.); (R.B.); (V.M.); (P.S.); (C.D.V.)
| | - Stefanie Marek Iannucci
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (S.P.); (A.L.F.); (S.M.); (C.C.); (S.M.I.); (L.N.); (D.A.); (D.F.); (M.V.M.); (V.M.); (R.G.); (S.C.); (M.M.); (R.B.); (V.M.); (P.S.); (C.D.V.)
| | - Lucrezia Netti
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (S.P.); (A.L.F.); (S.M.); (C.C.); (S.M.I.); (L.N.); (D.A.); (D.F.); (M.V.M.); (V.M.); (R.G.); (S.C.); (M.M.); (R.B.); (V.M.); (P.S.); (C.D.V.)
| | - Danilo Angotti
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (S.P.); (A.L.F.); (S.M.); (C.C.); (S.M.I.); (L.N.); (D.A.); (D.F.); (M.V.M.); (V.M.); (R.G.); (S.C.); (M.M.); (R.B.); (V.M.); (P.S.); (C.D.V.)
| | - Domenico Filomena
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (S.P.); (A.L.F.); (S.M.); (C.C.); (S.M.I.); (L.N.); (D.A.); (D.F.); (M.V.M.); (V.M.); (R.G.); (S.C.); (M.M.); (R.B.); (V.M.); (P.S.); (C.D.V.)
| | - Marco Valerio Mariani
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (S.P.); (A.L.F.); (S.M.); (C.C.); (S.M.I.); (L.N.); (D.A.); (D.F.); (M.V.M.); (V.M.); (R.G.); (S.C.); (M.M.); (R.B.); (V.M.); (P.S.); (C.D.V.)
| | - Vincenzo Myftari
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (S.P.); (A.L.F.); (S.M.); (C.C.); (S.M.I.); (L.N.); (D.A.); (D.F.); (M.V.M.); (V.M.); (R.G.); (S.C.); (M.M.); (R.B.); (V.M.); (P.S.); (C.D.V.)
| | - Rosanna Germanò
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (S.P.); (A.L.F.); (S.M.); (C.C.); (S.M.I.); (L.N.); (D.A.); (D.F.); (M.V.M.); (V.M.); (R.G.); (S.C.); (M.M.); (R.B.); (V.M.); (P.S.); (C.D.V.)
| | - Sara Cimino
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (S.P.); (A.L.F.); (S.M.); (C.C.); (S.M.I.); (L.N.); (D.A.); (D.F.); (M.V.M.); (V.M.); (R.G.); (S.C.); (M.M.); (R.B.); (V.M.); (P.S.); (C.D.V.)
| | - Massimo Mancone
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (S.P.); (A.L.F.); (S.M.); (C.C.); (S.M.I.); (L.N.); (D.A.); (D.F.); (M.V.M.); (V.M.); (R.G.); (S.C.); (M.M.); (R.B.); (V.M.); (P.S.); (C.D.V.)
| | - Roberto Badagliacca
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (S.P.); (A.L.F.); (S.M.); (C.C.); (S.M.I.); (L.N.); (D.A.); (D.F.); (M.V.M.); (V.M.); (R.G.); (S.C.); (M.M.); (R.B.); (V.M.); (P.S.); (C.D.V.)
| | - Viviana Maestrini
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (S.P.); (A.L.F.); (S.M.); (C.C.); (S.M.I.); (L.N.); (D.A.); (D.F.); (M.V.M.); (V.M.); (R.G.); (S.C.); (M.M.); (R.B.); (V.M.); (P.S.); (C.D.V.)
| | - Paolo Severino
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (S.P.); (A.L.F.); (S.M.); (C.C.); (S.M.I.); (L.N.); (D.A.); (D.F.); (M.V.M.); (V.M.); (R.G.); (S.C.); (M.M.); (R.B.); (V.M.); (P.S.); (C.D.V.)
| | - Carmine Dario Vizza
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (S.P.); (A.L.F.); (S.M.); (C.C.); (S.M.I.); (L.N.); (D.A.); (D.F.); (M.V.M.); (V.M.); (R.G.); (S.C.); (M.M.); (R.B.); (V.M.); (P.S.); (C.D.V.)
| |
Collapse
|
19
|
Osataphan N, Abdel-Qadir H, Zebrowska AM, Borowiec A. Sodium-Glucose Cotransporter 2 Inhibitors During Cancer Therapy: Benefits, Risks, and Ongoing Clinical Trials. Curr Oncol Rep 2024; 26:1188-1196. [PMID: 38990501 PMCID: PMC11480197 DOI: 10.1007/s11912-024-01577-8] [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] [Accepted: 07/02/2024] [Indexed: 07/12/2024]
Abstract
PURPOSE OF REVIEW The goal of this paper is to summarize the data pertaining to the use of sodium-glucose cotransporter-2 inhibitors (SGLT-2i) for the prevention of cardiotoxicity in patients receiving anthracyclines for cancer treatment. We discuss the potential efficacy of this class of medications, incorporating insights from existing literature and ongoing studies. RECENT FINDINGS SGLT2i are a class of medications which were initially developed for treatment of Type 2 diabetes and later extended to treat heart failure with reduced and preserved ejection fraction regardless of diabetes status. There remains a need for effective and safe treatments to preventing cardiotoxicity in anthracycline-treated patients. It has been proposed that SGLT2i may provide protection against the cardiotoxic effects of anthracyclines. Some of the proposed mechanisms include beneficial metabolic, neurohormonal, and hemodynamic effects, renal protection, as well as a decrease in inflammation, oxidative stress, apoptosis, mitochondrial dysfunction and ion homeostasis. There is emerging evidence from basic science and observational studies that SGLT2i may play a role in the prevention of chemotherapy-induced cardiotoxicity. Randomized controlled trials are needed to conclusively determine the role of SGLT2 inhibitors as a cardioprotective therapy in patients receiving anthracyclines for the treatment of cancer.
Collapse
Affiliation(s)
- Nichanan Osataphan
- Division of Cardiology, Ted Rogers Program in Cardiotoxicity Prevention, Peter Munk Cardiac Center, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON, Canada
- Division of Cardiology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Husam Abdel-Qadir
- Division of Cardiology, Ted Rogers Program in Cardiotoxicity Prevention, Peter Munk Cardiac Center, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON, Canada
- Women's College Hospital, Toronto, ON, Canada
| | - Agnieszka Maria Zebrowska
- Department of Cancer & Cardio-Oncology Diagnostics, The Maria Sklodowska-Curie National Research Institute of Oncology, Roentgena Str 5, 02-781, Warsaw, Poland
- Unit for Screening Studies in Inherited Cardiovascular Diseases, The Cardinal Stefan Wyszynski National Institute of Cardiology, Warsaw, Poland
| | - Anna Borowiec
- Department of Cancer & Cardio-Oncology Diagnostics, The Maria Sklodowska-Curie National Research Institute of Oncology, Roentgena Str 5, 02-781, Warsaw, Poland.
| |
Collapse
|
20
|
Li Y, Yan J, Yang P. The mechanism and therapeutic strategies in doxorubicin-induced cardiotoxicity: Role of programmed cell death. Cell Stress Chaperones 2024; 29:666-680. [PMID: 39343295 PMCID: PMC11490929 DOI: 10.1016/j.cstres.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/26/2024] [Accepted: 09/24/2024] [Indexed: 10/01/2024] Open
Abstract
Doxorubicin (DOX) is the most commonly used anthracycline anticancer agent, while its clinical utility is limited by harmful side effects like cardiotoxicity. Numerous studies have elucidated that programmed cell death plays a significant role in DOX-induced cardiotoxicity (DIC). This review summarizes several kinds of programmed cell death, including apoptosis, pyroptosis, necroptosis, autophagy, and ferroptosis. Furthermore, oxidative stress, inflammation, and mitochondrial dysfunction are also important factors in the molecular mechanisms of DIC. Besides, a comprehensive understanding of specific signal pathways of DIC can be helpful to its treatment. Therefore, the related signal pathways are elucidated in this review, including sirtuin deacetylase (silent information regulator 2 [Sir2]) 1 (SIRT1)/nuclear factor erythroid 2-related factor 2, SIRT1/Klotho, SIRT1/Recombinant Sestrin 2, adenosine monophosphate-activated protein kinase, AKT, and peroxisome proliferator-activated receptor. Heat shock proteins function as chaperones, which play an important role in various stressful situations, especially in the heart. Thus, some of heat shock proteins involved in DIC are also included. Hence, the last part of this review focuses on the therapeutic research based on the mechanisms above.
Collapse
Affiliation(s)
- Yanzhao Li
- Department of Second Clinical Medical College, Southern Medical University, Guangzhou, China.
| | - Jing Yan
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Pingzhen Yang
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
21
|
Zhao M, Liu C, Liu Z, Zuo Y, Chen C, Shi S, Shi X, Xie Y, Yang H, Chen Y. Myocardium-targeted liposomal delivery of the antioxidant peptide 8P against doxorubicin-induced myocardial injury. Int J Pharm 2024; 663:124569. [PMID: 39127172 DOI: 10.1016/j.ijpharm.2024.124569] [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: 04/10/2024] [Revised: 07/27/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024]
Abstract
Doxorubicin (Dox) is a broad-spectrum antineoplastic chemotherapeutic agent used in clinical settings, yet it exhibits significant cardiotoxicity, which in severe cases can lead to heart failure. Research indicates that oxidative stress plays a pivotal role in Dox -induced cardiomyocyte injury. Therefore, the application of antioxidants represents an effective strategy to mitigate the cardiotoxic effects of doxorubicin. In preliminary studies, we isolated an antioxidative peptide, PHWWEYRR (8P). This study utilizes a PCM cardiomyocyte-targeting peptide-modified liposome as a carrier to deliver 8P into cardiomyocytes, aiming to prevent Dox-induced cardiac injury through its antioxidative mechanism. The results demonstrated that we prepared the 8P-loaded and PCM-targeting peptide-modified liposome (P-P-8P), which exhibited good dispersibility, encapsulation efficiency, drug loading capacity, and in vitro release, along with myocardial targeting capability. In vitro experiments showed that P-P-8P could prevent oxidative stress injury in H9C2 cells, protect mitochondrial functions, and inhibit cell apoptosis through a mitochondria-dependent pathway. In vivo experiments indicated that P-P-8P could prevent abnormalities in serum biochemical indicators, cardiac dysfunction, and myocardial pathological changes in mice. In conclusion, P-P-8P effectively delivers 8P to cardiomyocytes, offering protection against the cardiotoxic effects of Dox, and holds potential as a future preventative or therapeutic agent for drug-induced cardiomyopathy.
Collapse
Affiliation(s)
- Meijun Zhao
- Department of Clinical Pharmacy, Affiliated Hospital of Jilin Medical College, Jilin, Jilin 132013, PR China.
| | - Chang Liu
- College of Pharmacy, Beihua University, Jilin, Jilin 132013, PR China
| | - Zhenye Liu
- College of Pharmacy, Beihua University, Jilin, Jilin 132013, PR China
| | - Yuanyuan Zuo
- College of Pharmacy, Beihua University, Jilin, Jilin 132013, PR China
| | - Chen Chen
- Affiliated Hospital of Yanbian University, Yanji, Jilin 133002, PR China
| | - Shuai Shi
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, PR China
| | - Xinlin Shi
- College of Pharmacy, Beihua University, Jilin, Jilin 132013, PR China
| | - Yining Xie
- College of Pharmacy, Beihua University, Jilin, Jilin 132013, PR China
| | - Huiying Yang
- College of Pharmacy, Beihua University, Jilin, Jilin 132013, PR China
| | - Yutong Chen
- College of Pharmacy, Beihua University, Jilin, Jilin 132013, PR China
| |
Collapse
|
22
|
Bews HJ, Mackic L, Jassal DS. Preventing broken hearts in women with breast cancer: a concise review on chemotherapy-mediated cardiotoxicity. Can J Physiol Pharmacol 2024; 102:487-497. [PMID: 38039515 DOI: 10.1139/cjpp-2023-0358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2023]
Abstract
Cancer and cardiovascular disease are the leading causes of death for Canadian women. One in eight Canadian women will receive the life-changing diagnosis of breast cancer (BC) in their lifetime, with 1 in 34 dying from the disease. Although doxorubicin (DOX) and trastuzumab (TRZ) have significantly improved survival in women diagnosed with human epidermal growth factor receptor 2 (HER2)-positive BC, approximately one in four women who receive this treatment are at risk of developing chemotherapy-induced cardiotoxicity. Cardiotoxicity is defined as a decline in left ventricular ejection fraction (LVEF) of >10% to an absolute value of <53%. Current guidelines recommend the serial monitoring of LVEF in this patient population using non-invasive cardiac imaging modalities including transthoracic echocardiography or multi-gated acquisition scan; however, this will only allow for the detection of established cardiotoxicity. Recent studies have demonstrated that a reduction in global longitudinal strain by speckle tracking echocardiography can identify pre-clinical systolic dysfunction prior to a decline in overall LVEF. Implementation of early detection techniques would allow for the prompt initiation of cardioprotective strategies. In addition to the early detection of chemotherapy-mediated cardiotoxicity, the prophylactic use of angiotensin-converting enzyme inhibitors, angiotensin receptor blockers, β-blockers, statins, exercise, and nutraceutical therapies have been studied in the setting of cardio-oncology.
Collapse
Affiliation(s)
- Hilary J Bews
- Section of Cardiology, Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Lana Mackic
- Institute of Cardiovascular Sciences, St. Boniface Hospital, University of Manitoba, Winnipeg, MB, Canada
| | - Davinder S Jassal
- Section of Cardiology, Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Institute of Cardiovascular Sciences, St. Boniface Hospital, University of Manitoba, Winnipeg, MB, Canada
- Department of Radiology, St. Boniface Hospital, University of Manitoba, Winnipeg, MB, Canada
- Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
23
|
Shaaban A, Scott SS, Greenlee AN, Binda N, Noor A, Webb A, Guo S, Purdy N, Pennza N, Habib A, Mohammad SJ, Smith SA. Atrial fibrillation in cancer, anticancer therapies, and underlying mechanisms. J Mol Cell Cardiol 2024; 194:118-132. [PMID: 38897563 PMCID: PMC11500699 DOI: 10.1016/j.yjmcc.2024.06.005] [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: 11/02/2023] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024]
Abstract
Atrial fibrillation (AF) is a common arrhythmic complication in cancer patients and can be exacerbated by traditional cytotoxic and targeted anticancer therapies. Increased incidence of AF in cancer patients is independent of confounding factors, including preexisting myocardial arrhythmogenic substrates, type of cancer, or cancer stage. Mechanistically, AF is characterized by fast unsynchronized atrial contractions with rapid ventricular response, which impairs ventricular filling and results in various symptoms such as fatigue, chest pain, and shortness of breath. Due to increased blood stasis, a consequence of both cancer and AF, concern for stroke increases in this patient population. To compound matters, cardiotoxic anticancer therapies themselves promote AF; thereby exacerbating AF morbidity and mortality in cancer patients. In this review, we examine the relationship between AF, cancer, and cardiotoxic anticancer therapies with a focus on the shared molecular and electrophysiological mechanisms linking these disease processes. We also explore the potential role of sodium-glucose co-transporter 2 inhibitors (SGLT2i) in the management of anticancer-therapy-induced AF.
Collapse
Affiliation(s)
- Adnan Shaaban
- The Ohio State University College of Medicine, Department of Internal Medicine, Columbus, OH 43210, USA
| | - Shane S Scott
- Medical Scientist Training Program, Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA; Bob and Corrinne Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Ashley N Greenlee
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA; Bob and Corrinne Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Nkongho Binda
- The Ohio State University College of Medicine, Department of Internal Medicine, Columbus, OH 43210, USA
| | - Ali Noor
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Averie Webb
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Shuliang Guo
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA; Bob and Corrinne Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Najhee Purdy
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA; Bob and Corrinne Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Nicholas Pennza
- Ohio University Heritage College of Osteopathic Medicine, Athens, OH 45701, USA
| | - Alma Habib
- The Ohio State University College of Medicine, Department of Internal Medicine, Division of Hematology, Columbus, OH 43210, USA
| | - Somayya J Mohammad
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA; Bob and Corrinne Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Sakima A Smith
- The Ohio State University College of Medicine, Department of Internal Medicine, Columbus, OH 43210, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA; Bob and Corrinne Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| |
Collapse
|
24
|
Lin R, Peng X, Li Y, Wang X, Liu X, Jia X, Zhang C, Liu N, Dong J. Empagliflozin attenuates doxorubicin-impaired cardiac contractility by suppressing reactive oxygen species in isolated myocytes. Mol Cell Biochem 2024; 479:2105-2118. [PMID: 37648958 DOI: 10.1007/s11010-023-04830-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] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 08/14/2023] [Indexed: 09/01/2023]
Abstract
In animal studies, sodium-glucose co-transporter-2 inhibitors-such as empagliflozin-have been shown to improve heart failure and impaired cardiac contractility induced by anthracyclines-including doxorubicin-although the therapeutic mechanism remains unclear. Moreover, abnormalities in Ca2+ handling within ventricular myocytes are the predominant feature of heart failure. Accordingly, this study aimed to investigate whether empagliflozin can alleviate Ca2+ handling disorders induced by acute doxorubicin exposure and elucidate the underlying mechanisms. To this end, ventricular myocytes were isolated from C57BL/6 mice. Contraction function, Ca2+ handling, and mitochondrial reactive oxygen species (ROS) generation were then evaluated using IonOptix or confocal microscopy. Ca2+ handling proteins were detected by western blotting. Results show that incubation with 1 μmol/L of doxorubicin for 120-min impaired cardiac contractility in isolated myocytes, which was significantly alleviated by pretreatment with 1 μmol/L of empagliflozin. Doxorubicin also markedly induced Ca2+ handling disorders, including decreased Ca2+ transients, prolonged Ca2+ transient decay time, enhanced frequency of Ca2+ sparks, and decreased Ca2+ content in the sarcoplasmic reticulum. These dysregulations were improved by pretreatment with empagliflozin. Moreover, empagliflozin effectively inhibited doxorubicin-induced mitochondrial ROS production in isolated myocytes and rescued doxorubicin-induced oxidation of Ca2+/calmodulin-dependent protein kinase II (ox-CaMKII) and CaMKII-dependent phosphorylation of RyR2. Similarly, preincubation with 10 μmol/L Mito-TEMPO mimicked the protective effects of empagliflozin. Collectively, Empagliflozin ameliorated the doxorubicin-induced contraction malfunction and Ca2+-handling disorders. These findings suggest that empagliflozin alleviates Ca2+-handling disorders by improving ROS production in the mitochondria and alleviating the enhanced oxidative CaMKII signaling pathway induced by doxorubicin.
Collapse
Affiliation(s)
- Rong Lin
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, No 2. Anzhen Road, Chaoyang, 100029, Beijing, China
- North China Medical & Health Group XingTai General Hospital, Xingtai, China
- National Clinical Research Center for Cardiovascular Diseases, Beijing, China
| | - Xiaodong Peng
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, No 2. Anzhen Road, Chaoyang, 100029, Beijing, China
- National Clinical Research Center for Cardiovascular Diseases, Beijing, China
| | - Yukun Li
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, No 2. Anzhen Road, Chaoyang, 100029, Beijing, China
- National Clinical Research Center for Cardiovascular Diseases, Beijing, China
| | - Xuesi Wang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, No 2. Anzhen Road, Chaoyang, 100029, Beijing, China
- National Clinical Research Center for Cardiovascular Diseases, Beijing, China
| | - Xinmeng Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, No 2. Anzhen Road, Chaoyang, 100029, Beijing, China
- National Clinical Research Center for Cardiovascular Diseases, Beijing, China
| | - Xingze Jia
- North China Medical & Health Group XingTai General Hospital, Xingtai, China
| | - Chengjun Zhang
- North China Medical & Health Group XingTai General Hospital, Xingtai, China
| | - Nian Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, No 2. Anzhen Road, Chaoyang, 100029, Beijing, China.
- National Clinical Research Center for Cardiovascular Diseases, Beijing, China.
| | - Jianzeng Dong
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, No 2. Anzhen Road, Chaoyang, 100029, Beijing, China.
- National Clinical Research Center for Cardiovascular Diseases, Beijing, China.
| |
Collapse
|
25
|
Szponar J, Niziński P, Dudka J, Kasprzak-Drozd K, Oniszczuk A. Natural Products for Preventing and Managing Anthracycline-Induced Cardiotoxicity: A Comprehensive Review. Cells 2024; 13:1151. [PMID: 38995002 PMCID: PMC11240786 DOI: 10.3390/cells13131151] [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: 05/11/2024] [Revised: 07/03/2024] [Accepted: 07/04/2024] [Indexed: 07/13/2024] Open
Abstract
Doxorubicin (DOX) is an anthracycline anticancer agent that is highly effective in the treatment of solid tumors. Given the multiplicity of mechanisms involved in doxorubicin-induced cardiotoxicity, it is difficult to identify a precise molecular target for toxicity. The findings of a literature review suggest that natural products may offer cardioprotective benefits against doxorubicin-induced cardiotoxicity, both in vitro and in vivo. However, further confirmatory studies are required to substantiate this claim. It is of the utmost importance to direct greater attention towards the intricate signaling networks that are of paramount importance for the survival and dysfunction of cardiomyocytes. Notwithstanding encouraging progress made in preclinical studies of natural products for the prevention of DOX-induced cardiotoxicity, these have not yet been translated for clinical use. One of the most significant obstacles hindering the development of cardioprotective adjuvants based on natural products is the lack of adequate bioavailability in humans. This review presents an overview of current knowledge on doxorubicin DOX-induced cardiotoxicity, with a focus on the potential benefits of natural compounds and herbal preparations in preventing this adverse effect. As literature search engines, the browsers in the Scopus, PubMed, Web of Science databases and the ClinicalTrials.gov register were used.
Collapse
Affiliation(s)
- Jarosław Szponar
- Clinical Department of Toxicology and Cardiology, Toxicology Clinic, Stefan Wyszyński Regional Specialist Hospital, Medical University of Lublin, 20-718 Lublin, Poland;
| | - Przemysław Niziński
- Department of Pharmacology, Medical University of Lublin, Radziwiłłowska 11 Street, 20-080 Lublin, Poland;
| | - Jarosław Dudka
- Chair and Department of Toxicology, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland;
| | - Kamila Kasprzak-Drozd
- Department of Inorganic Chemistry, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland;
| | - Anna Oniszczuk
- Department of Inorganic Chemistry, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland;
| |
Collapse
|
26
|
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
|
27
|
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
|
28
|
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
|
29
|
Chang HY, Hsu HC, Fang YH, Liu PY, Liu YW. Empagliflozin attenuates doxorubicin-induced cardiotoxicity by inhibiting the JNK signaling pathway. Biomed Pharmacother 2024; 176:116759. [PMID: 38788603 DOI: 10.1016/j.biopha.2024.116759] [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: 03/22/2024] [Revised: 05/09/2024] [Accepted: 05/17/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND Sodium-glucose cotransporter-2 inhibitors, such as empagliflozin, are pivotal therapies for heart failure. However, the effect of empagliflozin on doxorubicin-related cardiac dysfunction remains unclear. METHODS Human induced pluripotent stem cell- and embryonic stem cell-derived cardiomyocytes were used to investigate the direct effect of empagliflozin on human cardiomyocytes. Then, the c-Jun amino-terminal kinases (JNK) inhibitor SP600125 was administered to the doxorubicin cardiotoxicity model in vitro and in vivo to investigate the role of JNK in empagliflozin. RESULTS In human stem cell-derived cardiomyocytes, pretreatment with empagliflozin attenuated doxorubicin-induced cleavage of caspase 3 and other apoptosis markers. Empagliflozin significantly attenuated doxorubicin-induced phosphorylation of JNK and p38. Inhibiting the phosphorylation of JNK (SP600125) or STAT3 attenuated doxorubicin-induced apoptosis, but inhibiting the phosphorylation of p38 did not. SP600125 inhibits the phosphorylation of STAT3 (S727), and a STAT3 (Y705) inhibitor also inhibits the phosphorylation of JNK. Empagliflozin and SP600125 attenuated doxorubicin-induced increases in reactive oxygen species (ROS) and decreases in oxidized nicotinamide adenine dinucleotide (NAD+). In animal studies, empagliflozin and SP600125 attenuated doxorubicin-induced cardiac dysfunction and fibrosis. CONCLUSIONS Empagliflozin attenuated doxorubicin-induced apoptosis by inhibiting the phosphorylation of JNK and its downstream signaling pathways, including ROS and NAD+.
Collapse
Affiliation(s)
- Hsien-Yuan Chang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Division of Cardiology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hsiao-Chun Hsu
- Division of Cardiology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Hsien Fang
- Center of Cell Therapy, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ping-Yen Liu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Division of Cardiology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| | - Yen-Wen Liu
- Division of Cardiology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Center of Cell Therapy, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
30
|
Xie S, Sun Y, Zhao X, Xiao Y, Zhou F, Lin L, Wang W, Lin B, Wang Z, Fang Z, Wang L, Zhang Y. An update of the molecular mechanisms underlying anthracycline induced cardiotoxicity. Front Pharmacol 2024; 15:1406247. [PMID: 38989148 PMCID: PMC11234178 DOI: 10.3389/fphar.2024.1406247] [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: 03/25/2024] [Accepted: 06/10/2024] [Indexed: 07/12/2024] Open
Abstract
Anthracycline drugs mainly include doxorubicin, epirubicin, pirarubicin, and aclamycin, which are widely used to treat a variety of malignant tumors, such as breast cancer, gastrointestinal tumors, lymphoma, etc. With the accumulation of anthracycline drugs in the body, they can induce serious heart damage, limiting their clinical application. The mechanism by which anthracycline drugs cause cardiotoxicity is not yet clear. This review provides an overview of the different types of cardiac damage induced by anthracycline-class drugs and delves into the molecular mechanisms behind these injuries. Cardiac damage primarily involves alterations in myocardial cell function and pathological cell death, encompassing mitochondrial dysfunction, topoisomerase inhibition, disruptions in iron ion metabolism, myofibril degradation, and oxidative stress. Mechanisms of uptake and transport in anthracycline-induced cardiotoxicity are emphasized, as well as the role and breakthroughs of iPSC in cardiotoxicity studies. Selected novel cardioprotective therapies and mechanisms are updated. Mechanisms and protective strategies associated with anthracycline cardiotoxicity in animal experiments are examined, and the definition of drug damage in humans and animal models is discussed. Understanding these molecular mechanisms is of paramount importance in mitigating anthracycline-induced cardiac toxicity and guiding the development of safer approaches in cancer treatment.
Collapse
Affiliation(s)
- Sicong Xie
- Department of Rehabilitation Medicine, School of Acupuncture-Moxibustion and Tuina and School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuwei Sun
- Department of Rehabilitation Medicine, School of Acupuncture-Moxibustion and Tuina and School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xuan Zhao
- Department of General Surgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yiqun Xiao
- Department of Rehabilitation Medicine, School of Acupuncture-Moxibustion and Tuina and School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
| | - Fei Zhou
- Department of Rehabilitation Medicine, School of Acupuncture-Moxibustion and Tuina and School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
| | - Liang Lin
- Department of Rehabilitation Medicine, School of Acupuncture-Moxibustion and Tuina and School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wei Wang
- College of Electronic and Optical Engineering and College of Flexible Electronics, Future Technology, Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Bin Lin
- Key Laboratory of Intelligent Pharmacy and Individualized Therapy of Huzhou, Department of Pharmacy, Changxing People's Hospital, Huzhou, China
| | - Zun Wang
- Department of Rehabilitation Medicine, School of Acupuncture-Moxibustion and Tuina and School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zixuan Fang
- Department of Rehabilitation Medicine, School of Acupuncture-Moxibustion and Tuina and School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lei Wang
- Department of Rehabilitation Medicine, School of Acupuncture-Moxibustion and Tuina and School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Zhang
- Department of Rehabilitation Medicine, School of Acupuncture-Moxibustion and Tuina and School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory of Intelligent Pharmacy and Individualized Therapy of Huzhou, Department of Pharmacy, Changxing People's Hospital, Huzhou, China
| |
Collapse
|
31
|
Christ T, Schwedhelm E, Eschenhagen T. Dapagliflozin and atrial fibrillation: elevated dosing to achieve class I antiarrhythmic effects? Basic Res Cardiol 2024; 119:505-507. [PMID: 38568224 PMCID: PMC11142992 DOI: 10.1007/s00395-024-01047-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/08/2024] [Accepted: 03/20/2024] [Indexed: 06/01/2024]
Affiliation(s)
- Torsten Christ
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany.
| | - Edzard Schwedhelm
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Eschenhagen
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany.
| |
Collapse
|
32
|
Kuo HH, Wang KT, Chen HH, Lai ZY, Lin PL, Chuang YJ, Liu LYM. Cardiovascular outcomes associated with SGLT2 inhibitor therapy in patients with type 2 diabetes mellitus and cancer: a systematic review and meta-analysis. Diabetol Metab Syndr 2024; 16:108. [PMID: 38773486 PMCID: PMC11110336 DOI: 10.1186/s13098-024-01354-4] [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] [Received: 04/22/2024] [Accepted: 05/18/2024] [Indexed: 05/23/2024] Open
Abstract
BACKGROUND Cancer patients with diabetes are at increased risk for cardiovascular diseases due to common risk factors and well-documented drug-associated cardiotoxicity. Sodium-glucose cotransporter-2 (SGLT2) inhibitors have shown cardiovascular benefits in patients with diabetes, but their effects on cancer patients remain unclear. This study aimed to evaluate the cardiovascular outcomes associated with SGLT2 inhibitor therapy in patients with concomitant diabetes and cancer. METHODS We conducted a systematic review and meta-analysis of cohort studies comparing cardiovascular outcomes between cancer patients with diabetes receiving SGLT2 inhibitors and those not receiving SGLT2 inhibitors. PubMed, Embase, and the Cochrane Library were searched from inception to February 29, 2024. The primary outcome was all-cause mortality, and the secondary outcomes were heart failure hospitalization, and adverse events. Random-effect models were used to calculate pooled risk ratios (RR) with 95% confidence intervals (CI). Subgroup and sensitivity analyses were conducted to identify potential sources of heterogeneity and explore the effect of SGLT2 inhibitors on mitigating cardiotoxicity. RESULTS Nine cohort studies involving 82,654 patients were included. SGLT2 inhibitor use was associated with a significantly lower risk of all-cause mortality (RR 0.46, 95% CI 0.31-0.68, P < 0.0001; I2 = 98%) and heart failure hospitalization (RR 0.49, 95% CI 0.30-0.81, P = 0.006; I2 = 21%) compared to non-use. The mortality benefit remained significant in patients receiving anthracycline chemotherapy (RR 0.50, 95% CI 0.28-0.89, P = 0.02; I2 = 71%). SGLT2 inhibitor use was also associated with a lower risk of sepsis (RR 0.32, 95% CI 0.23-0.44, P < 0.00001; I2 = 0%) and no increased risk of diabetic ketoacidosis (RR 0.66, 95% CI 0.20-2.16, P = 0.49; I2 = 0%). CONCLUSIONS SGLT2 inhibitor therapy is associated with lower risks of all-cause mortality and heart failure hospitalization in patients with concomitant diabetes and cancer. These findings suggest that SGLT2 inhibitors may offer cardiovascular benefits in this high-risk population. Randomized controlled trials are needed to validate these findings and evaluate the safety and efficacy of SGLT2 inhibitors in specific cancer types and treatment regimens.
Collapse
Affiliation(s)
- Hsiao-Huai Kuo
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
- Department of Pharmacy, Hsinchu Municipal MacKay Children's Hospital, Hsinchu, Taiwan
- Department of Pharmacy, Hsinchu MacKay Memorial Hospital, Hsinchu, Taiwan
- Department of Nursing, Hsin Sheng Junior College of Medical Care and Management, Taoyuan, Taiwan
| | - Kuang-Te Wang
- Division of Cardiology, Department of Internal Medicine, Taitung MacKay Memorial Hospital, Taitung, Taiwan
- Department of Medicine, Mackay Medical College, New Taipei, Taiwan
| | - Hsin-Hao Chen
- Department of Medicine, Mackay Medical College, New Taipei, Taiwan
- Nursing, and Management, MacKay Junior College of Medicine, Taipei, Taiwan
- Department of Family Medicine, Hsinchu MacKay Memorial Hospital, Hsinchu, Taiwan
| | - Zih-Yin Lai
- School of Medicine, National Tsing Hua University, Hsinchu, Taiwan
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Po-Lin Lin
- Nursing, and Management, MacKay Junior College of Medicine, Taipei, Taiwan
- Division of Cardiology, Department of Medicine, Hsinchu MacKay Memorial Hospital, Hsinchu, Taiwan
| | - Yung-Jen Chuang
- School of Medicine, National Tsing Hua University, Hsinchu, Taiwan
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Lawrence Yu-Min Liu
- Department of Medicine, Mackay Medical College, New Taipei, Taiwan.
- Division of Cardiology, Department of Medicine, Hsinchu MacKay Memorial Hospital, Hsinchu, Taiwan.
| |
Collapse
|
33
|
Quagliariello V, Canale ML, Bisceglia I, Iovine M, Paccone A, Maurea C, Scherillo M, Merola A, Giordano V, Palma G, Luciano A, Bruzzese F, Zito Marino F, Montella M, Franco R, Berretta M, Gabrielli D, Gallucci G, Maurea N. Sodium-glucose cotransporter 2 inhibitor dapagliflozin prevents ejection fraction reduction, reduces myocardial and renal NF-κB expression and systemic pro-inflammatory biomarkers in models of short-term doxorubicin cardiotoxicity. Front Cardiovasc Med 2024; 11:1289663. [PMID: 38818214 PMCID: PMC11138344 DOI: 10.3389/fcvm.2024.1289663] [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: 09/06/2023] [Accepted: 04/09/2024] [Indexed: 06/01/2024] Open
Abstract
Background Anthracycline-mediated adverse cardiovascular events are among the leading causes of morbidity and mortality in patients with cancer. Sodium-glucose cotransporter 2 inhibitors (SGLT2i) exert multiple cardiometabolic benefits in patients with/without type 2 diabetes, chronic kidney disease, and heart failure with reduced and preserved ejection fraction. We hypothesized that the SGLT2i dapagliflozin administered before and during doxorubicin (DOXO) therapy could prevent cardiac dysfunction and reduce pro-inflammatory pathways in preclinical models. Methods Cardiomyocytes were exposed to DOXO alone or combined with dapagliflozin (DAPA) at 10 and 100 nM for 24 h; cell viability, iATP, and Ca++ were quantified; lipid peroxidation products (malondialdehyde and 4-hydroxy 2-hexenal), NLRP3, MyD88, and cytokines were also analyzed through selective colorimetric and enzyme-linked immunosorbent assay (ELISA) methods. Female C57Bl/6 mice were treated for 10 days with a saline solution or DOXO (2.17 mg/kg), DAPA (10 mg/kg), or DOXO combined with DAPA. Systemic levels of ferroptosis-related biomarkers, galectin-3, high-sensitivity C-reactive protein (hs-CRP), and pro-inflammatory chemokines (IL-1α, IL-1β, IL-2, IL-4, IL-6, IL-10, IL-12, IL17-α, IL-18, IFN-γ, TNF-α, G-CSF, and GM-CSF) were quantified. After treatments, immunohistochemical staining of myocardial and renal p65/NF-kB was performed. Results DAPA exerts cytoprotective, antioxidant, and anti-inflammatory properties in human cardiomyocytes exposed to DOXO by reducing iATP and iCa++ levels, lipid peroxidation, NLRP-3, and MyD88 expression. Pro-inflammatory intracellular cytokines were also reduced. In preclinical models, DAPA prevented the reduction of radial and longitudinal strain and ejection fraction after 10 days of treatment with DOXO. A reduced myocardial expression of NLRP-3 and MyD-88 was seen in the DOXO-DAPA group compared to DOXO mice. Systemic levels of IL-1β, IL-6, TNF-α, G-CSF, and GM-CSF were significantly reduced after treatment with DAPA. Serum levels of galectine-3 and hs-CRP were strongly enhanced in the DOXO group; on the other hand, their expression was reduced in the DAPA-DOXO group. Troponin-T, B-type natriuretic peptide (BNP), and N-Terminal Pro-BNP (NT-pro-BNP) were strongly reduced in the DOXO-DAPA group, revealing cardioprotective properties of SGLT2i. Mice treated with DOXO and DAPA exhibited reduced myocardial and renal NF-kB expression. Conclusion The overall picture of the study encourages the use of DAPA in the primary prevention of cardiomyopathies induced by anthracyclines in patients with cancer.
Collapse
Affiliation(s)
- V. Quagliariello
- Division of Cardiology, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, Napoli, Italia
| | - M. L. Canale
- Cardiology Division, Azienda USL Toscana Nord-Ovest, Versilia Hospital, Lido di Camaiore, Italy
| | - I. Bisceglia
- Integrated Cardiology Services, Department of Cardio-Thoracic-Vascular, Azienda Ospedaliera San Camillo Forlanini, Rome, Italy
| | - M. Iovine
- Division of Cardiology, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, Napoli, Italia
| | - A. Paccone
- Division of Cardiology, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, Napoli, Italia
| | - C. Maurea
- ASL NA1, UOC Neurology and Stroke Unit, Ospedale del Mare, Naples, Italy
| | - M. Scherillo
- Cardiology Department, San Pio Hospital, Benevento, Italy
| | - A. Merola
- Department of Pharmacy, University of Salerno, Salerno, Italy
| | - V. Giordano
- Division of Cardiology, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, Napoli, Italia
| | - G. Palma
- SSD Sperimentazione Animale, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, Napoli, Italy
| | - A. Luciano
- SSD Sperimentazione Animale, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, Napoli, Italy
| | - F. Bruzzese
- SSD Sperimentazione Animale, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, Napoli, Italy
| | - F. Zito Marino
- Pathology Unit, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - M. Montella
- Pathology Unit, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - R. Franco
- Pathology Unit, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - M. Berretta
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - D. Gabrielli
- U.O.C. Cardiologia, Dipartimento Cardio-Toraco-Vascolare, Azienda Ospedaliera San Camillo Forlani-ni, Roma—Fondazione per il Tuo Cuore—Heart Care Foundation, Firenze, Italy
| | - G. Gallucci
- Cardio-Oncology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Referral Cancer Center of Basilicata, Rionero in Vulture, Italy
| | - N. Maurea
- Division of Cardiology, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, Napoli, Italia
| |
Collapse
|
34
|
Malik A, Bagchi AK, Jassal DS, Singal PK. Doxorubicin‑induced cardiomyopathy is mitigated by empagliflozin via the modulation of endoplasmic reticulum stress pathways. Mol Med Rep 2024; 29:74. [PMID: 38488036 PMCID: PMC10958136 DOI: 10.3892/mmr.2024.13198] [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: 11/23/2023] [Accepted: 02/08/2024] [Indexed: 03/19/2024] Open
Abstract
Doxorubicin (Dox) exhibits a high efficacy in the treatment of numerous types of cancer. However, the beneficial cytotoxic effects of Dox are often accompanied by an increase in the risk of cardiotoxicity. Oxidative stress (OS) plays a key role in Dox‑induced cardiomyopathy (DIC). OS in cardiomyocytes disrupts endoplasmic reticulum (ER) function, leading to the accumulation of misfolded/unfolded proteins known as ER stress. ER stress acts as an adaptive mechanism; however, prolonged ER stress together with OS may lead to the initiation of cardiomyocyte apoptosis. The present study aimed to explore the potential of an anti‑diabetic drug, empagliflozin (EMPA), in mitigating Dox‑induced ER stress and cardiomyocyte apoptosis. In the present study, the effects of 1 h pretreatment of EMPA on Dox‑treated cardiomyocytes isolated from Sprague‑Dawley rats were investigated. After 24 h, EMPA pre‑treatment promoted cell survival in the EMPA + Dox group compared with the Dox group. Results of the present study also demonstrated that EMPA mitigated overall ER stress, as the increased expression of ER stress markers was reduced in the EMPA + Dox group. Additionally, OS, inflammation and expression of ER stress apoptotic proteins were also significantly reduced following EMPA pre‑treatment in the EMPA + Dox group. Thus, EMPA may exert beneficial effects on Dox‑induced ER stress and may exhibit potential changes that can be utilised to further evaluate the role of EMPA in mitigating DIC.
Collapse
Affiliation(s)
- Akshi Malik
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg R2H 2A6, Canada
| | - Ashim K. Bagchi
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Davinder S. Jassal
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg R2H 2A6, Canada
- Section of Cardiology, Department of Internal Medicine, Max Rady College of Medicine, Rady Faculty of Health Sciences, St. Boniface Hospital, Winnipeg, Manitoba R2H 2A6, Canada
| | - Pawan K. Singal
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg R2H 2A6, Canada
| |
Collapse
|
35
|
VanDerStad LR, Wyatt EC, Vaughan RA. The antidiabetic SGLT2 inhibitor canagliflozin reduces mitochondrial metabolism in a model of skeletal muscle insulin resistance. Diabet Med 2024; 41:e15271. [PMID: 38140911 DOI: 10.1111/dme.15271] [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: 08/21/2023] [Revised: 11/20/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023]
Abstract
AIMS Sodium-glucose cotransporter 2 (SGLT2) inhibitors such as canagliflozin (CANA) have emerged as an effective adjuvant therapy in the management of diabetes, however, past observations suggest CANA may alter skeletal muscle mass and function. The purpose of this work was to investigate the effects of CANA on skeletal muscle metabolism both with and without insulin resistance. METHODS C2C12 myotubes were treated with CANA with or without insulin resistance. Western blot and qRT-PCR were used to assess protein and gene expression, respectively. Cell metabolism was assessed via oxygen consumption and extracellular acidification rate. Mitochondrial, nuclei and lipid content were measured using fluorescent staining and microscopy. RESULTS CANA decreased mitochondrial function and glycolytic metabolism as did insulin resistance, however, these changes occurred without significant alterations in gene expression associated with each pathway. Additionally, while insulin resistance reduced insulin-stimulated pAkt expression, CANA had no significant effect on insulin sensitivity. CONCLUSIONS CANA appears to reduce mitochondrial and glycolytic metabolism without altering gene expression governing these pathways, suggesting a reduction in substrate may be responsible for lower metabolism.
Collapse
Affiliation(s)
- Lindsey R VanDerStad
- Department of Health and Human Performance, High Point University, High Point, North Carolina, USA
| | - Emily C Wyatt
- Department of Health and Human Performance, High Point University, High Point, North Carolina, USA
| | - Roger A Vaughan
- Department of Health and Human Performance, High Point University, High Point, North Carolina, USA
| |
Collapse
|
36
|
Tabowei G, Dadzie SK, Perswani P, Nawaz S, Kaur M, Moqattash M, Wei CR, Hirani S. Efficacy of Sodium-Glucose Cotransporter 2 Inhibitors in Preventing Heart Failure in Patients Receiving Anthracycline-Based Cancer Therapy: A Systematic Review and Meta-Analysis. Cureus 2024; 16:e60086. [PMID: 38860078 PMCID: PMC11163855 DOI: 10.7759/cureus.60086] [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] [Accepted: 05/11/2024] [Indexed: 06/12/2024] Open
Abstract
Anthracyclines are effective chemotherapeutic agents widely used to treat various cancers, but their use is limited by the risk of cardiotoxicity and heart failure. While strategies like dose reduction have been explored, there are no well-established therapies to mitigate this risk. Emerging evidence suggests sodium-glucose cotransporter 2 inhibitors (SGLT2i) may have cardioprotective effects, providing a rationale for investigating their potential utility in anthracycline-treated patients. We conducted a systematic review and meta-analysis to synthesize available evidence on the efficacy of SGLT2i in reducing heart failure incidence and mortality in patients undergoing anthracycline-based cancer therapy. Relevant studies were identified through comprehensive database searches and screened based on predefined criteria. Data extraction and quality assessment were performed independently by two reviewers. Four observational studies, encompassing 5,590 patients, were included. The pooled analysis showed a higher but non-significant risk of developing heart failure in the non-SGLT2i group compared to the SGLT2i group (RR = 0.67, 95% CI: 0.40-1.41). The risk of all-cause mortality was significantly lower in patients receiving SGLT2i (RR = 0.55, 95% CI: 0.39-0.77). This meta-analysis suggests SGLT2i are associated with a lower risk of mortality and heart failure incidence in anthracycline-treated patients, although larger studies are needed to confirm these findings. The mechanisms underlying these potential benefits require further elucidation. Despite limitations, this analysis highlights the promising role of SGLT2i as a cardioprotective strategy in this high-risk population.
Collapse
Affiliation(s)
- Godfrey Tabowei
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Samuel K Dadzie
- Internal Medicine, Piedmont Athens Regional Medical Center, Athens, USA
| | - Prinka Perswani
- Internal Medicine, Liaquat University of Medical and Health Sciences, Hyderabad, PAK
| | - Sheeza Nawaz
- Medicine, Fatima Jinnah Medical University, Lahore, PAK
| | - Mandeep Kaur
- Internal Medicine, Hospital Corporation of America (HCA) Capital Hospital, Tallahassee, USA
| | | | - Calvin R Wei
- Research and Development, Shing Huei Group, Taipei, TWN
| | | |
Collapse
|
37
|
Dabour MS, George MY, Daniel MR, Blaes AH, Zordoky BN. The Cardioprotective and Anticancer Effects of SGLT2 Inhibitors: JACC: CardioOncology State-of-the-Art Review. JACC CardioOncol 2024; 6:159-182. [PMID: 38774006 PMCID: PMC11103046 DOI: 10.1016/j.jaccao.2024.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 05/24/2024] Open
Abstract
Sodium-glucose cotransporter-2 (SGLT2) inhibitors, originally approved for type 2 diabetes mellitus, have demonstrated efficacy in reducing cardiovascular events, particularly heart failure, in patients with and without diabetes. An intriguing research area involves exploring the potential application of SGLT2 inhibitors in cardio-oncology, aiming to mitigate the cardiovascular adverse events associated with anticancer treatments. These inhibitors present a unique dual nature, offering both cardioprotective effects and anticancer properties, conferring a double benefit for cardio-oncology patients. In this review, the authors first examine the established cardioprotective effects of SGLT2 inhibitors in heart failure and subsequently explore the existing body of evidence, including both preclinical and clinical studies, that supports the use of SGLT2 inhibitors in the context of cardio-oncology. The authors further discuss the mechanisms through which SGLT2 inhibitors protect against cardiovascular toxicity secondary to cancer treatment. Finally, they explore the potential anticancer effects of SGLT2 inhibitors along with their proposed mechanisms.
Collapse
Affiliation(s)
- Mohamed S. Dabour
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Mina Y. George
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mary R. Daniel
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Anne H. Blaes
- Division of Hematology/Oncology/Transplantation, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - Beshay N. Zordoky
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
38
|
Cersosimo A, Salerno N, Sabatino J, Scatteia A, Bisaccia G, De Rosa S, Dellegrottaglie S, Bucciarelli-Ducci C, Torella D, Leo I. Underlying mechanisms and cardioprotective effects of SGLT2i and GLP-1Ra: insights from cardiovascular magnetic resonance. Cardiovasc Diabetol 2024; 23:94. [PMID: 38468245 PMCID: PMC10926589 DOI: 10.1186/s12933-024-02181-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 02/26/2024] [Indexed: 03/13/2024] Open
Abstract
Originally designed as anti-hyperglycemic drugs, Glucagon-Like Peptide-1 receptor agonists (GLP-1Ra) and Sodium-glucose cotransporter-2 inhibitors (SGLT2i) have demonstrated protective cardiovascular effects, with significant impact on cardiovascular morbidity and mortality. Despite several mechanisms have been proposed, the exact pathophysiology behind these effects is not yet fully understood. Cardiovascular imaging is key for the evaluation of diabetic patients, with an established role from the identification of early subclinical changes to long-term follow up and prognostic assessment. Among the different imaging modalities, CMR may have a key-role being the gold standard for volumes and function assessment and having the unique ability to provide tissue characterization. Novel techniques are also implementing the possibility to evaluate cardiac metabolism through CMR and thereby further increasing the potential role of the modality in this context. Aim of this paper is to provide a comprehensive review of changes in CMR parameters and novel CMR techniques applied in both pre-clinical and clinical studies evaluating the effects of SGLT2i and GLP-1Ra, and their potential role in better understanding the underlying CV mechanisms of these drugs.
Collapse
Affiliation(s)
- Angelica Cersosimo
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Nadia Salerno
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Jolanda Sabatino
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Alessandra Scatteia
- Advanced Cardiovascular Imaging Unit, Ospedale Medico-Chirurgico Accreditato Villa dei Fiori, Naples, Italy
| | - Giandomenico Bisaccia
- Department of Neuroscience, Imaging and Clinical Sciences, Institute for Advanced Biomedical Technologies "G. d'Annunzio", University of Chieti-Pescara, Chieti, Italy
| | - Salvatore De Rosa
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Santo Dellegrottaglie
- Advanced Cardiovascular Imaging Unit, Ospedale Medico-Chirurgico Accreditato Villa dei Fiori, Naples, Italy
| | - Chiara Bucciarelli-Ducci
- CMR Unit, Royal Brompton and Harefield Hospitals, Guy's and St Thomas' NHS Foundation Trust, London, UK
- School of Biomedical Engineering and Imaging Sciences, Faculty of Life Sciences and Medicine, Kings College London, London, UK
| | - Daniele Torella
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy.
| | - Isabella Leo
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy.
| |
Collapse
|
39
|
Vafa RG, Sabahizadeh A, Mofarrah R. Guarding the heart: How SGLT-2 inhibitors protect against chemotherapy-induced cardiotoxicity: SGLT-2 inhibitors and chemotherapy-induced cardiotoxicity. Curr Probl Cardiol 2024; 49:102350. [PMID: 38128634 DOI: 10.1016/j.cpcardiol.2023.102350] [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: 12/13/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023]
Abstract
The introduction of chemotherapy agents has significantly transformed cancer treatment, with anthracyclines being one of the most commonly used drugs. While these agents have proven to be highly effective against various types of cancers, they come with complications, including neurotoxicity, nephrotoxicity, and cardiotoxicity. Among these side effects, cardiotoxicity is the leading cause of morbidity and mortality, with anthracyclines being the primary culprit. Chemotherapy medications have various mechanisms that can lead to cardiac injury. Hence, numerous studies have been conducted to decrease the cardiotoxicity of these treatments. Combination therapy with beta-blockers, Angiotensin-converting enzyme inhibitors, and angiotensin receptor blockers have effectively reduced such outcomes. However, a definitive preventive strategy is yet to be established. Meanwhile, sodium-glucose co-transporter-2 (SGLT-2) inhibitors lower blood glucose levels in type 2 diabetes by reducing its re-absorption in the kidneys. They are thus considered potent drugs for glycemic control and reduction of cardiovascular risks. Recent studies have shown that SGLT-2 inhibitors are crucial in preventing chemotherapy-induced cardiotoxicity. They enhance heart cell viability, prevent degenerative changes, stimulate autophagy, and reduce cell death. This drug class also reduces inflammation by inhibiting reactive oxygen species and inflammatory cytokine production. Moreover, it can not only reverse the harmful effects of anticancer agents on the heart structure but also enhance the effectiveness of chemotherapy by minimizing potential consequences on the heart. In conclusion, SGLT-2 inhibitors hold promise as a therapeutic strategy for protecting cancer patients from chemotherapy-induced heart damage and improving cardiovascular outcomes.
Collapse
|
40
|
Belger C, Abrahams C, Imamdin A, Lecour S. Doxorubicin-induced cardiotoxicity and risk factors. IJC HEART & VASCULATURE 2024; 50:101332. [PMID: 38222069 PMCID: PMC10784684 DOI: 10.1016/j.ijcha.2023.101332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 12/14/2023] [Accepted: 12/16/2023] [Indexed: 01/16/2024]
Abstract
Doxorubicin (DOX) is an anthracycline antibiotic widely used as a chemotherapeutic agent to treat solid tumours and hematologic malignancies. Although useful in the treatment of cancers, the benefit of DOX is limited due to its cardiotoxic effect that is observed in a large number of patients. In the literature, there is evidence that the presence of various factors may increase the risk of developing DOX-induced cardiotoxicity. A better understanding of the role of these different factors in DOX-induced cardiotoxicity may facilitate the choice of the therapeutic approach in cancer patients suffering from various cardiovascular risk factors. In this review, we therefore discuss the latest findings in both preclinical and clinical research suggesting a link between DOX-induced cardiotoxicity and various risk factors including sex, age, ethnicity, diabetes, dyslipidaemia, obesity, hypertension, cardiovascular disease and co-medications.
Collapse
Affiliation(s)
| | | | - Aqeela Imamdin
- Cardioprotection Group, Cape Heart Institute, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Sandrine Lecour
- Cardioprotection Group, Cape Heart Institute, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
41
|
Yu YW, Chen X, Yan JY, Hu J, Huang KY, Ji KT, Cai HL. Phlorizin, a novel caloric restriction mimetic, stimulates hypoxia and protects cardiomyocytes through activating autophagy via modulating the Hif-1α/Bnip3 axis in sepsis-induced myocardial dysfunction. Int Immunopharmacol 2024; 126:111241. [PMID: 37984253 DOI: 10.1016/j.intimp.2023.111241] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/30/2023] [Accepted: 11/14/2023] [Indexed: 11/22/2023]
Abstract
BACKGROUND Sepsis is a systemic inflammatory syndrome that can lead to multiple organ dysfunction and life-threatening complications. Sepsis-induced myocardial dysfunction (SIMD) has been confirmed to be present in half of patients with septic shock, increasing their mortality rate to 70-90%. The pathogenesis of SIMD is complex, and no specific clinical treatment has yet been developed. Caloric restriction mimetics (CRM), compounds that simulate the biochemical and functional properties of CR, can improve cardiovascular injury by activating autophagy. This study investigated the effect of a new type of CRM which can induce hypoxia, the SGLT nonspecific inhibitor phlorizin on SIMD. MATERIALS AND METHODS In vivo, phlorizin was administered at 1 mg/kg/day intragastrically for 28 days. In vitro, AC16 was treated with 120 μM phlorizin for 48 h. Echocardiography was used to assess cardiac function. Myocardial injury markers were detected in serum and cell supernatant. Western blotting was employed to detect changed proteins associated with apoptosis and autophagy. Immunofluorescence, immunohistochemistry, co-immunoprecipitation, molecular docking, and other methods were also used to illustrate cellular changes. RESULTS In vivo, phlorizin significantly improved the survival rate and cardiac function after sepsis injury, reduced markers of myocardial injury, inhibited myocardial apoptosis and oxidative stress, and promoted autophagy. In vitro, phlorizin alleviated the apoptosis of AC16, as well as inhibited oxidative stress and apoptotic enzyme activity. Phlorizin acts on autophagy at multiple sites through low energy (activation of AMPK) and hypoxia (release of Beclin-1 by Hif-1α/Bnip3 axis), promoting the formation and degradation of autophagosomes. CONCLUSION We indicated for the first time that phlorizin could inhibit glucose uptake via GLUT-1 and conforms to the metabolic characteristics of CRM, it can induce the hypoxic transcriptional paradigm. In addition, it inhibits apoptosis and improves SIMD by promoting autophagy generation and unobstructing autophagy flux. Moreover, it affects autophagy by releasing Beclin-1 through the Hif-1α/Bnip3 axis.
Collapse
Affiliation(s)
- Yong-Wei Yu
- Intensive Care Unit, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Department of Cardiology, The Second Affiliated and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 312500, China
| | - Xia Chen
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jue-Yue Yan
- Intensive Care Unit, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Juan Hu
- Intensive Care Unit, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Kai-Yu Huang
- Department of Cardiology, The Second Affiliated and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 312500, China
| | - Kang-Ting Ji
- Department of Cardiology, The Second Affiliated and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 312500, China.
| | - Hong-Liu Cai
- Intensive Care Unit, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| |
Collapse
|
42
|
Yadav J, Ahsan F, Panda P, Mahmood T, Bano S, Shamim A, Mishra P. Repurposing of Empagliflozin as Cardioprotective Drug: An in-silico Approach. Cardiovasc Hematol Disord Drug Targets 2024; 24:254-265. [PMID: 39686639 DOI: 10.2174/011871529x341930241206063315] [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: 08/02/2024] [Revised: 10/19/2024] [Accepted: 11/12/2024] [Indexed: 12/18/2024]
Abstract
BACKGROUND Drug repurposing involves investigating new indications or uses for drugs that have already been approved for clinical use. Empagliflozin is a C-glycosyl compound characterized by the presence of a beta-glucosyl residue. It functions as a sodium-glucose co-transporter 2 inhibitor and is utilized to enhance glycemic control in adults diagnosed with type 2 diabetes mellitus. Additionally, it is indicated for the reduction of cardiovascular mortality risk in adult patients who have both type 2 diabetes mellitus and pre-existing cardiovascular disease. OBJECTIVE The study's objective revolves around exploring the repurposing potential of a novel SGLT2 inhibitor acting as an antidiabetic drug named Empagliflozin through computational methods, with a specific focus on its interaction with cardioprotective key target proteins. METHODS The study was performed by docking the empagliflozin with different target proteins (NHE1- CHP1, BIRC5, GLUT1, and XIAP) by using Autodock, and different values were recorded. The docked files were analysed by the BIOVIA Discovery Studio Visualizer. The in silico analysis conducted in this study examines the binding free energy values of Empagliflozin with key target proteins. RESULTS Results revealed that NHE1-CHP1 exhibits the lowest binding free energy, followed by BIRC5, GLUT1, and XIAP, with the highest value. This descending order of binding energies suggests varying degrees of effectiveness in binding molecules, with lower energies indicative of more potent biological activity. The analysis underscores the importance of intermolecular interactions, particularly hydrogen bond formations facilitated by oxygen, nitrogen, and carbonyl groups in compound structures. Notably, NHE1-CHP1 demonstrates superior binding interactions with Empagliflozin compared to the other target proteins, highlighting its potential as a cardioprotective agent. CONCLUSION These findings offer valuable insights into the therapeutic possibilities of Empagliflozin in cardioprotection, indicating promising avenues for further research and development in this domain.
Collapse
Affiliation(s)
- Jyoti Yadav
- Department of Pharmacy, Integral University, Dasauli, Kursi Road, Lucknow (U.P.), 226026, India
| | - Farogh Ahsan
- Department of Pharmacy, Integral University, Dasauli, Kursi Road, Lucknow (U.P.), 226026, India
| | - Prabhudatta Panda
- Department of Pharmacy, Institute of Technology & Management, Gorakhpur (U.P.), 273209, India
| | - Tarique Mahmood
- Department of Pharmacy, Integral University, Dasauli, Kursi Road, Lucknow (U.P.), 226026, India
| | - Shahzadi Bano
- Department of Chemistry, Integral University, Dasauli, Kursi Road, Lucknow (U.P.), 226026, India
| | - Arshiya Shamim
- Department of Pharmacy, Integral University, Dasauli, Kursi Road, Lucknow (U.P.), 226026, India
| | - Pooja Mishra
- Department of Pharmacy, Integral University, Dasauli, Kursi Road, Lucknow (U.P.), 226026, India
| |
Collapse
|
43
|
Avula V, Sharma G, Kosiborod MN, Vaduganathan M, Neilan TG, Lopez T, Dent S, Baldassarre L, Scherrer-Crosbie M, Barac A, Liu J, Deswal A, Khadke S, Yang EH, Ky B, Lenihan D, Nohria A, Dani SS, Ganatra S. SGLT2 Inhibitor Use and Risk of Clinical Events in Patients With Cancer Therapy-Related Cardiac Dysfunction. JACC. HEART FAILURE 2024; 12:67-78. [PMID: 37897456 DOI: 10.1016/j.jchf.2023.08.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 07/10/2023] [Accepted: 08/28/2023] [Indexed: 10/30/2023]
Abstract
BACKGROUND Certain antineoplastic therapies are associated with an increased risk of cardiomyopathy and heart failure (HF). Sodium glucose co-transporter 2 (SGLT2) inhibitors improve outcomes in patients with HF. OBJECTIVES This study aims to examine the efficacy of SGLT2 inhibitors in patients with cancer therapy-related cardiac dysfunction (CTRCD) or HF. METHODS The authors conducted a retrospective cohort analysis of deidentified, aggregate patient data from the TriNetX research network. Patients aged ≥18 years with a history of type 2 diabetes mellitus, cancer, and exposure to potentially cardiotoxic antineoplastic therapies, with a subsequent diagnosis of cardiomyopathy or HF between January 1, 2013, and April 30, 2020, were identified. Patients with ischemic heart disease were excluded. Patients receiving guideline-directed medical therapy were divided into 2 groups based on SGLT2 inhibitor use. After propensity score matching, odds ratios (ORs) and Cox proportional HRs were used to compare outcomes over a 2-year follow-up period. RESULTS The study cohort included 1,280 patients with CTRCD/HF (n = 640 per group; mean age: 67.6 years; 41.6% female; 68% White). Patients on SGLT2 inhibitors in addition to conventional guideline-directed medical therapy had a lower risk of acute HF exacerbation (OR: 0.483 [95% CI: 0.36-0.65]; P < 0.001) and all-cause mortality (OR: 0.296 [95% CI: 0.22-0.40]; P = 0.001). All-cause hospitalizations or emergency department visits (OR: 0.479; 95% CI: 0.383-0.599; P < 0.001), atrial fibrillation/flutter (OR: 0.397 [95% CI: 0.213-0.737]; P = 0.003), acute kidney injury (OR: 0.486 [95% CI: 0.382-0.619]; P < 0.001), and need for renal replacement therapy (OR: 0.398 [95% CI: 0.189-0.839]; P = 0.012) were also less frequent in patients on SGLT2 inhibitors. CONCLUSIONS SGLT2 inhibitor use is associated with improved outcomes in patients with CTRCD/HF.
Collapse
Affiliation(s)
- Vennela Avula
- Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Garima Sharma
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Mikhail N Kosiborod
- Saint Luke's Mid America Heart Institute, Kansas City, Missouri, USA; University of Missouri, Kansas City, Missouri, USA
| | - Muthiah Vaduganathan
- Brigham and Women's Hospital Heart and Vascular Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Tomas G Neilan
- Cardio-Oncology Program, Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Teresa Lopez
- Department of Cardiology, La Paz University Hospital, Madrid, Spain
| | - Susan Dent
- Duke Cancer Institute, Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Lauren Baldassarre
- Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Marielle Scherrer-Crosbie
- Division of Cardiology, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ana Barac
- Cardio-Oncology Program, Division of Cardiology, MedStar Washington Hospital Center, Washington, DC, USA
| | - Jennifer Liu
- Cardio-Oncology Program, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - Anita Deswal
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sumanth Khadke
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Beth Israel Lahey Health, Burlington, Massachusetts, USA
| | - Eric H Yang
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California at Los Angeles, Los Angeles, California, USA
| | - Bonnie Ky
- Division of Cardiology, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Daniel Lenihan
- International Cardio-Oncology Society, Tampa, Florida, USA; St. Francis Healthcare, Cape Girardeau, Missouri, USA
| | - Anju Nohria
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Sourbha S Dani
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Beth Israel Lahey Health, Burlington, Massachusetts, USA
| | - Sarju Ganatra
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Beth Israel Lahey Health, Burlington, Massachusetts, USA.
| |
Collapse
|
44
|
Refaie MMM, Shehata S, El-Hussieny M, Fawzy MA, Ahmed NZM, Marey H, Hishmat AM, Alkully T, Rahman ESMAE. Molecular mechanism of empagliflozin cardioprotection in 5-fluorouracil (5-FU)-induced cardiotoxicity via modulation of SGLT2 and TNFα/TLR/NF-κB signaling pathway in rats. Toxicol Res 2024; 40:139-151. [PMID: 38223670 PMCID: PMC10786789 DOI: 10.1007/s43188-023-00204-1] [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: 02/22/2023] [Revised: 07/18/2023] [Accepted: 07/26/2023] [Indexed: 01/16/2024] Open
Abstract
One of the commoly used chemotherapeutic agents is 5-Fluorouracil (5-FU). Unfortunately, the clinical administration of 5-FU is complicated with serious cardiotoxic effects and the safe use becomes an urgent task in cardio-oncology. Till now, there are no studies discussed the role of empagliflozin (EMP) against 5-FU cardiotoxicity. Thus, we investigated this effect and the involved mechanisms in 5-FU induced heart injury. Forty male rats of Wistar albino species were used and divided randomly into four groups. Group I is the control group, group II is EMP given group, group III is 5-FU cardiotoxic group and group IV is 5-FU plus EMP group. 5-FU (150 mg/kg) was administered as a single intraperitoneal (i.p.) dose on 1st day to induce cardiotoxicity with or without EMP (30 mg/kg/d) orally for 5 days. The dose of 5-FU is relevant to the human toxic dose. Our data showed that 5-FU given group caused cardiotoxicity with significant increase of serum cardiac enzymes, toll like receptors, enhancement of nuclear factor kappa B (NF-κB), interleukin1β (IL1β), IL6, myeloid-differentiation-factor 88 (MYD88), heart weight, malondialdehyde (MDA), tumor-necrosis-factor-alpha (TNFα), sodium glucose co-transporter 2 (SGLT2), P53 and caspase3 expression with clear histopathological features of cardiotoxicity. Moreover, there is a significant decrease in reduced glutathione (GSH) and total antioxidant capacity (TAC). Interestingly, co-administration of EMP could ameliorate 5-FU induced biochemical and histopathological changes. This effect may be due to modulation of SGLT2, decreasing inflammation, oxidative stress and apoptosis with downregulation of an essential inflammatory cascade that mediates 5-FU cardiotoxicity; TNFα/TLR/NF-κB. Supplementary Information The online version contains supplementary material available at 10.1007/s43188-023-00204-1.
Collapse
Affiliation(s)
| | - Sayed Shehata
- Department of Cardiology, Faculty of Medicine, Minia University, El-Minia, 61511 Egypt
| | - Maram El-Hussieny
- Department of Pathology, Faculty of Medicine, Minia University, El-Minia, 61511 Egypt
| | - Michael Atef Fawzy
- Department of Biochemistry, Faculty of Pharmacy, Minia University, El-Minia, 61511 Egypt
| | - Nagwa Zenhom Mustafa Ahmed
- Department of Biochemistry, Faculty of Medicine, Minia University, El-Minia, 61511 Egypt
- Department of Biochemistry, Faculty of Medicine, Al-Baha University, 65525 Albaha, Saudi Arabia
| | - Heba Marey
- Department of Biochemistry, Faculty of Medicine, Minia University, El-Minia, 61511 Egypt
| | - Asmaa Mohammed Hishmat
- Department of Forensic Medicine & Clinical Toxicology, Faculty of Medicine, Minia University, El-Minia, 61511 Egypt
| | - Turki Alkully
- Department of Internal Medicine, Faculty of Medicine, Al-Baha University, 65525 Albaha, Saudi Arabia
| | | |
Collapse
|
45
|
Beretta GL. Ferroptosis-induced Cardiotoxicity and Antitumor Drugs. Curr Med Chem 2024; 31:4935-4957. [PMID: 37469161 DOI: 10.2174/0929867331666230719124453] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 06/06/2023] [Accepted: 06/15/2023] [Indexed: 07/21/2023]
Abstract
The induction of regulated cell death ferroptosis in tumors is emerging as an intriguing strategy for cancer treatment. Numerous antitumor drugs (e.g., doxorubicin, etoposide, tyrosine kinase inhibitors, trastuzumab, arsenic trioxide, 5-fluorouracil) induce ferroptosis. Although this mechanism of action is interesting for fighting tumors, the clinical use of drugs that induce ferroptosis is hampered by cardiotoxicity. Besides in cancer cells, ferroptosis induced by chemotherapeutics can occur in cardiomyocytes, and this feature represents an important drawback of antitumor therapy. This inconvenience has been tackled by developing less or no cardiotoxic antitumor drugs or by discovering cardioprotective agents (e.g., berberine, propofol, fisetin, salidroside, melatonin, epigallocatechin- 3gallate, resveratrol) to use in combination with conventional chemotherapeutics. This review briefly summarizes the molecular mechanisms of ferroptosis and describes the ferroptosis dependent mechanisms responsible for cardiac toxicity developed by cancer- suffering patients following the administration of some chemotherapeutics. Additionally, the pharmacological strategies very recently proposed for potentially preventing this inconvenience are considered.
Collapse
Affiliation(s)
- Giovanni Luca Beretta
- Molecular Pharmacology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
| |
Collapse
|
46
|
Kim MS, Choi HK, Park SH, Lee JI, Lee J. Poncirus trifoliata Aqueous Extract Protects Cardiomyocytes against Doxorubicin-Induced Toxicity through Upregulation of NAD(P)H Dehydrogenase Quinone Acceptor Oxidoreductase 1. Molecules 2023; 28:8090. [PMID: 38138580 PMCID: PMC10745630 DOI: 10.3390/molecules28248090] [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: 10/29/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
Doxorubicin (DOX), an anthracycline-based chemotherapeutic agent, is widely used to treat various types of cancer; however, prolonged treatment induces cardiomyotoxicity. Although studies have been performed to overcome DOX-induced cardiotoxicity (DICT), no effective method is currently available. This study investigated the effects and potential mechanisms of Poncirus trifoliata aqueous extract (PTA) in DICT. Changes in cell survival were assessed in H9c2 rat cardiomyocytes and MDA-MB-231 human breast cancer cells. The C57BL/6 mice were treated with DOX to induce DICT in vivo, and alterations in electrophysiological characteristics, serum biomarkers, and histological features were examined. The PTA treatment inhibited DOX-induced decrease in H9c2 cell viability but did not affect the MDA-MB-231 cell viability. Additionally, the PTA restored the abnormal heart rate, R-R interval, QT interval, and ST segment and inhibited the decrease in serum cardiac and hepatic toxicity indicators in the DICT model. Moreover, the PTA administration protected against myocardial fibrosis and apoptosis in the heart tissue of mice with DICT. PTA treatment restored DOX-induced decrease in the expression of NAD(P)H dehydrogenase quinone acceptor oxidoreductase 1 in a PTA concentration-dependent manner. In conclusion, the PTA inhibitory effect on DICT is attributable to its antioxidant properties, suggesting the potential of PTA as a phytotherapeutic agent for DICT.
Collapse
Affiliation(s)
| | | | | | | | - Jangho Lee
- Korea Food Research Institute, Wanju 55365, Republic of Korea; (M.-S.K.); (H.-K.C.); (S.-H.P.); (J.-I.L.)
| |
Collapse
|
47
|
Hwang HJ, Kim M, Jun JE, Yon DK. Sodium-glucose cotransporter-2 inhibitors improve clinical outcomes in patients with type 2 diabetes mellitus undergoing anthracycline-containing chemotherapy: an emulated target trial using nationwide cohort data in South Korea. Sci Rep 2023; 13:21756. [PMID: 38066029 PMCID: PMC10709414 DOI: 10.1038/s41598-023-48678-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023] Open
Abstract
Novel hypoglycemic agents, sodium-glucose cotransporter 2 inhibitors (SGLT2i), have shown protective effects against anthracycline (AC)-induced cardiotoxicity and exhibit partial anticancer effects in animal models. However, clinical evidence for this is scarce. This study aimed to evaluate whether SGLT2i improve the clinical outcomes of patients with type 2 diabetes mellitus (T2DM) undergoing AC-containing chemotherapy. A total of 81,572 patients who underwent AC chemotherapy between 2014 and 2021 were recruited from a nationwide Korean cohort. Patients were classified into three groups: patients with T2DM taking SGLT2i (n = 780) and other hypoglycemic agents excluding SGLT2i (non-SGLT2i; n = 3,455) during AC chemotherapy, and the non-DM group (n = 77,337). The clinical outcome was a composite of heart failure hospitalization, acute myocardial infarction, ischemic stroke, and death. After propensity score matching, 779 SGLT2i users were compared with 7800 non-DM patients and 2,337 non-SGLT2i users. The SGLT2i group had better composite outcomes compared with the non-DM group (adjusted hazard ratio [HR] = 0.35, 95% confidence interval [95% CI] = 0.25-0.51) and compared with the non-SGLT2i group (adjusted HR = 0.47, 95% CI = 0.32-0.69). In conclusion, SGLT2i may contribute to improving clinical outcomes in patients with T2DM undergoing AC-containing chemotherapy, through an emulated target trial using Korean nationwide cohort data.
Collapse
Affiliation(s)
- Hui-Jeong Hwang
- Department of Cardiology, Kyung Hee University College of Medicine, Kyung Hee University Hospital at Gangdong, Seoul, 05278, Republic of Korea.
| | - Minji Kim
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University Medical Center, Kyung Hee University College of Medicine, Seoul, 02447, Republic of Korea
- Department of Regulatory Science, Kyung Hee University, Seoul, Republic of Korea
| | - Ji Eun Jun
- Department of Endocrinology and Metabolism, Kyung Hee University Hospital at Gangdong, Kyung Hee University School of Medicine, Seoul, Republic of Korea
| | - Dong Keon Yon
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University Medical Center, Kyung Hee University College of Medicine, Seoul, 02447, Republic of Korea.
- Department of Regulatory Science, Kyung Hee University, Seoul, Republic of Korea.
| |
Collapse
|
48
|
Vaziri Z, Saleki K, Aram C, Alijanizadeh P, Pourahmad R, Azadmehr A, Ziaei N. Empagliflozin treatment of cardiotoxicity: A comprehensive review of clinical, immunobiological, neuroimmune, and therapeutic implications. Biomed Pharmacother 2023; 168:115686. [PMID: 37839109 DOI: 10.1016/j.biopha.2023.115686] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 10/03/2023] [Accepted: 10/08/2023] [Indexed: 10/17/2023] Open
Abstract
Cancer and cardiovascular disorders are known as the two main leading causes of mortality worldwide. Cardiotoxicity is a critical and common adverse effect of cancer-related chemotherapy. Chemotherapy-induced cardiotoxicity has been associated with various cancer treatments, such as anthracyclines, immune checkpoint inhibitors, and kinase inhibitors. Different methods have been reported for the management of chemotherapy-induced cardiotoxicity. In this regard, sodium-glucose cotransporter-2 inhibitors (SGLT2i), a class of antidiabetic agents, have recently been applied to manage heart failure patients. Further, SGLT2i drugs such as EMPA exert protective cardiac and systemic effects. Moreover, it can reduce inflammation through the mediation of major inflammatory components, such as Nucleotide-binding domain-like receptor protein 3 (NLRP3) inflammasomes, Adenosine 5'-monophosphate-activated protein kinase (AMPK), and c-Jun N-terminal kinase (JNK) pathways, Signal transducer and activator of transcription (STAT), and overall decreasing transcription of proinflammatory cytokines. The clinical outcome of EMPA administration is related to improving cardiovascular risk factors, including body weight, lipid profile, blood pressure, and arterial stiffness. Intriguingly, SGLT2 suppressors can regulate microglia-driven hyperinflammation affecting neurological and cardiovascular disorders. In this review, we discuss the protective effects of EMPA in chemotherapy-induced cardiotoxicity from molecular, immunological, and neuroimmunological aspects to preclinical and clinical outcomes.
Collapse
Affiliation(s)
- Zahra Vaziri
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Kiarash Saleki
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; USERN Office, Babol University of Medical Sciences, Babol, Iran; Department of e-Learning, Virtual School of Medical Education and Management, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Cena Aram
- Department of Cell & Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Parsa Alijanizadeh
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Ramtin Pourahmad
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Azadmehr
- Immunology Department, Babol University of Medical Sciences, Babol, Iran
| | - Naghmeh Ziaei
- Clinical Research Development unit of Rouhani Hospital, Babol University of Medical Sciences, Babol, Iran; Department of Cardiology, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
49
|
Lin Z, Wang J. Taxifolin protects against doxorubicin-induced cardiotoxicity and ferroptosis by adjusting microRNA-200a-mediated Nrf2 signaling pathway. Heliyon 2023; 9:e22011. [PMID: 38053888 PMCID: PMC10694176 DOI: 10.1016/j.heliyon.2023.e22011] [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: 06/26/2023] [Revised: 11/01/2023] [Accepted: 11/01/2023] [Indexed: 12/07/2023] Open
Abstract
The chemotherapeutic agent doxorubicin (Dox) is commonly used to treat various types of cancer, even though it can cause life-threatening cardiotoxicity. Clinically, there is no particularly effective way to treat Dox-induced cardiotoxicity. Therefore, it is imperative to identify compounds that can effectively alleviate Dox-induced cardiotoxicity. Ferroptosis and oxidative stress play a key role in Dox-induced cardiotoxicity, and the inhibition of ferroptosis and oxidative stress could effectively protect against doxorubicin-induced cardiotoxicity. Taxifolin (TAX) is a flavonoid commonly found in onions and citrus fruits. In the present study, we evaluated the effects of TAX on Dox-induced cardiac injury and dysfunction and aimed to explore the mechanisms underlying these effects. Using a mouse model of Dox-induced cardiotoxicity, we administered 20 mg/kg/day of TAX by gavage for 2 weeks. A week after the first use of TAX, each mouse was administered a 10 mg/kg dose of Dox. TAX was first evaluated for its cardioprotective properties, and the outcomes showed that TAX significantly reduced the damage caused by Dox to the myocardium in terms of structural and functional damage by effectively inhibiting ferroptosis and oxidative stress. In vivo, echocardiography, histopathologic assay, serum biochemical analysis and western blotting was used to find the results that Dox promoted ferroptosis-induced cardiomyocyte death, while TAX reversed these effects. In vitro, we also found that TAX alleviated Dox-induced cardiotoxicity by using ROS/DHE staining assay, Cellular immunofluorescence and western blotting. TAX increasing expression of microRNA-200a (miR-200a) which affects ferroptosis by activating Nrf2 signaling pathway. We believe that TAX inhibits ferroptosis and is a potential phytochemical that prevents Dox-induced cardiotoxicity.
Collapse
Affiliation(s)
- Zhihui Lin
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Jie Wang
- Department of Endocrinology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| |
Collapse
|
50
|
Camilli M, Cardinale DM, Pedrotti P, Turazza FM, Aspromonte N, Canale ML, Bisceglia I, Oliva F, Colivicchi F. Report from the Cardio-Oncology Symposium at the Associazione Nazionale Medici Cardiologi Ospedalieri (ANMCO) Annual Congress. Future Cardiol 2023; 19:519-522. [PMID: 36047607 DOI: 10.2217/fca-2022-0063] [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: 11/21/2022] Open
Abstract
Overview of the meeting The Cardio-Oncology Symposium at the Associazione Nazionale Medici Cardiologi Ospedalieri (ANMCO) Annual Meeting mainly focused on the diagnosis, management and prevention of cardiovascular toxicity of cancer drugs, in particular, cardiac dysfunction induced by anthracyclines. Although a variety of cardiac biomarkers and imaging modalities are available, there remains no consensus regarding their appropriate use to identify early and late cardiotoxicity and to guide preventive strategies. At the same time, the multitude of pharmacological trials, aimed at preventing cardiac damage through a neurohormonal blockade, provided conflicting results. Nevertheless, the advent of novel heart failure medications can change the decision-making of the cardio-oncologist. This symposium attempted to harmonize these issues.
Collapse
Affiliation(s)
- Massimiliano Camilli
- Department of Cardiovascular & Pulmonary Sciences, Catholic University of the Sacred Heart, Largo Francesco Vito 1, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, Rome, Italy
| | - Daniela M Cardinale
- Cardioncology Unit, European Institute of Oncology, IRCCS, Via Ripamonti 435, Milan, Italy
| | - Patrizia Pedrotti
- Cardiac Magnetic Resonance Laboratory, Cardiologia 4, ASST Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore 3, Milan, Italy
| | - Fabio M Turazza
- Cardiology Unit, Istituto Nazionale Tumori, Istituto di Ricerca e Cura di Carattere Scientifico Via Giacomo Venezian 1, Milan, Italy
| | - Nadia Aspromonte
- Department of Cardiovascular & Pulmonary Sciences, Catholic University of the Sacred Heart, Largo Francesco Vito 1, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, Rome, Italy
| | - Maria Laura Canale
- Department of Cardiology, Nuovo Ospedale Versilia Lido Di Camaiore, Camaiore, SS 1 335, Lucca, Italy
| | - Irma Bisceglia
- Department of Cardio-Thoracic-Vascular, Integrated Cardiology Services, Azienda Ospedaliera San Camillo Forlanini, Circonvallazione Gianicolense 87, Roma, Italy
| | - Fabrizio Oliva
- Department of Cardio-Thoracic-Vascular, Cardiology 1, "A. De Gasperis," ASST Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore 3, Milan, Italy
| | - Furio Colivicchi
- Department of Clinical & Rehabilitation Cardiology, Presidio Ospedaliero San Filippo Neri ASL Roma 1, Via Giovanni Martinotti 20, Roma, Italy
| |
Collapse
|