1
|
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
|
2
|
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
|
3
|
Wang J, Yang W. Advances in sodium-glucose transporter protein 2 inhibitors and tumors. Front Oncol 2025; 15:1522059. [PMID: 40007997 PMCID: PMC11850236 DOI: 10.3389/fonc.2025.1522059] [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: 11/03/2024] [Accepted: 01/15/2025] [Indexed: 02/27/2025] Open
Abstract
Tumor is a major challenge to global health and has received extensive attention worldwide due to its high degree of malignancy and poor prognosis. Although the clinical application of targeted therapy and immunotherapy has improved the status quo of tumor treatment, the development of new therapeutic tools for tumors is still necessary. Sodium-glucose transporter protein 2 (SGLT2) inhibitors are a new type of glycemic control drugs, which are widely used in clinical practice because of their effects on weight reduction and protection of cardiac and renal functions. SGLT2 has been found to be overexpressed in many tumors and involved in tumorigenesis, progression and metastasis, suggesting that SGLT2i has a wide range of applications in tumor therapy. The aim of this article is to provide a comprehensive understanding of the research progress of SGLT2i in different tumors by integrating the latest studies and to encourage further exploration of SGLT2i therapies in clinical trials. This could pave the way for more effective management strategies and improved outcomes for tumor patients.
Collapse
Affiliation(s)
- Jiaqi Wang
- Department of Oncology, Guangyuan Central Hospital, Guangyuan, Sichuan, China
| | - Wenyong Yang
- Department of Respiratory and Critical Care Medicine, Guangyuan Central Hospital, Guangyuan, Sichuan, China
| |
Collapse
|
4
|
Guo Z, Javaheri A. Empagliflozin to Prevent Doxorubicin Cardiotoxicity. JACC CardioOncol 2025; 7:185-187. [PMID: 39967205 DOI: 10.1016/j.jaccao.2025.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 01/10/2025] [Accepted: 01/13/2025] [Indexed: 02/20/2025] Open
Affiliation(s)
- Zhen Guo
- Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Ali Javaheri
- Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA; John Cochran Veterans Affairs Hospital, St. Louis, Missouri, USA.
| |
Collapse
|
5
|
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
|
6
|
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
|
7
|
Pan R, He Y, Melisandre W, Zhang Y, Su W, Feng J, Jia C, Li S, Liu B. Bibliometric and visual analysis of SGLT2 inhibitors in cardiovascular diseases. Front Pharmacol 2024; 15:1437760. [PMID: 39539627 PMCID: PMC11557488 DOI: 10.3389/fphar.2024.1437760] [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: 05/24/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Background Cardiovascular diseases (CVD) pose a significant threat to human health due to their high mortality and morbidity rates. Despite advances in treatments, the prevalence and impact of cardiovascular disease continue to increase. Sodium-glucose transporter 2 inhibitors (SGLT2i), initially approved for the treatment of type 2 diabetes, have important research value and promising applications in reducing CVD risk, especially in heart failure (HF) and atherosclerosis patients with cardiovascular disease (ASCVD). This study aims to comprehensively review the latest progress, research trends, cutting-edge hot spots, and future development directions of SGLT2i in the field of CVD through bibliometric analysis. Methods Articles related to MSCs in cardiovascular diseases were sourced from the Web of Science. The bibliometric analysis was conducted using CiteSpace and VOSviewer, and a knowledge map was created based on the data obtained from the retrieved articles. Results In this article, we screened 3,476 relevant studies, including 2,293 articles and 1,183 reviews. The analysis found that the number of papers related to the application of SGLT2i in CVD has generally increased, peaking in 2022. The United States and China contributed the largest number of papers, with the United States accounting for 36.97% of the total and also ranking first in terms of the number of citations. However, China's high-quality papers are slightly lacking and need further improvement. Keyword analysis showed that empagliflozin, dapagliflozin, diabetes, and heart failure were the most common terms, reflecting the main research interests in currently published papers in this field. Conclusion Bibliometric analysis showed a robust and growing interest in the application of SGLT2i for treating CVD. By summarizing the latest progress of SGLT2i in the field of CVD, exploring research hotspots, and looking forward to future research development trends, this article provides valuable insights for thinking about research prospects.
Collapse
Affiliation(s)
- Runfang Pan
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuqing He
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wan Melisandre
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yunyi Zhang
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenyuan Su
- Sport Medicine and Rehabilitation Center, Shanghai University of Sport, Shanghai, China
| | - Jiaming Feng
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chengyao Jia
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Shaoling Li
- Department of Pathology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Baonian Liu
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
Bertolini D, Pizzi C, Donal E, Galli E. Cancer and Heart Failure: Dangerous Liaisons. J Cardiovasc Dev Dis 2024; 11:263. [PMID: 39330321 PMCID: PMC11432566 DOI: 10.3390/jcdd11090263] [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: 06/09/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/28/2024] Open
Abstract
Cancer and heart failure (HF) are increasingly relevant worldwide, both from an epidemiologic and clinical point of view. This review aims to explore the relationship between cancer and HF by underscoring risk factors and disclosing the cardiotoxic effects of the current chemotherapy agents. We also deal with the current evidence on the diagnosis and management of HF related to cancer therapy. Finally, we will address the main gaps in knowledge and future perspectives in this field.
Collapse
Affiliation(s)
- Davide Bertolini
- Cardiology Unit, Cardiac Thoracic and Vascular Department, IRCCS Azienda Ospedaliera-Universitaria di Bologna, 40138 Bologna, Italy
- Department of Medical and Surgical Sciences-DIMEC, Alma Mater Studiorum, University of Bologna, 40138 Bologna, Italy
| | - Carmine Pizzi
- Cardiology Unit, Cardiac Thoracic and Vascular Department, IRCCS Azienda Ospedaliera-Universitaria di Bologna, 40138 Bologna, Italy
- Department of Medical and Surgical Sciences-DIMEC, Alma Mater Studiorum, University of Bologna, 40138 Bologna, Italy
| | - Erwan Donal
- Univ Rennes, CHU Rennes, Inserm, LTSI-UMR 1099, F-35000 Rennes, France
| | - Elena Galli
- Univ Rennes, CHU Rennes, Inserm, LTSI-UMR 1099, F-35000 Rennes, France
| |
Collapse
|
11
|
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
|
12
|
George MY, Dabour MS, Rashad E, Zordoky BN. Empagliflozin Alleviates Carfilzomib-Induced Cardiotoxicity in Mice by Modulating Oxidative Stress, Inflammatory Response, Endoplasmic Reticulum Stress, and Autophagy. Antioxidants (Basel) 2024; 13:671. [PMID: 38929110 PMCID: PMC11200801 DOI: 10.3390/antiox13060671] [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: 04/24/2024] [Revised: 05/17/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
Carfilzomib is an irreversible proteasome inhibitor used for multiple myeloma patients. However, carfilzomib treatment is associated with cardiovascular complications. Empagliflozin, an Sodium Glucose Co-transporter 2 inhibitor (SGLT-2) inhibitor, is an oral antidiabetic drug with proven antioxidant and anti-inflammatory properties. The aim of the present study was to determine the cardioprotective effects of empagliflozin against carfilzomib-induced cardiotoxicity. C57BL/6 mice were randomly divided into four groups: control, empagliflozin, carfilzomib, and carfilzomib + empagliflozin. Empagliflozin prevented carfilzomib-induced cardiotoxicity by ameliorating histological alterations, CK-MB, and troponin-I. Moreover, it inhibited carfilzomib-induced oxidative damage and inflammation via its action on catalase activity, reduced glutathione levels and superoxide dismutase activity, and reduced nuclear factor-κB (p65) and cytokine levels. Mechanistically, empagliflozin abrogated endoplasmic reticulum stress induced by carfilzomib, as evidenced by the effect on the Glucose Regulated Protein-78 (GRP-78)/Activating Transcription Factor 6 (ATF6)/C/EBP homologous protein (CHOP) axis. Intriguingly, carfilzomib significantly induced autophagy, an effect that was further enhanced by empagliflozin, evidenced by increased LC3B and beclin-1 mRNA expression and reduced p62 expression. The effect of empagliflozin on apoptosis was confirmed by reduced expression of active caspase-3. Importantly, empagliflozin did not alter the cytotoxic effect of carfilzomib on human U266B1 multiple myeloma cells. our findings suggest that empagliflozin may provide a new therapeutic strategy to mitigate carfilzomib-induced cardiotoxicity in multiple myeloma patients.
Collapse
Affiliation(s)
- Mina Y. George
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA; (M.S.D.); (B.N.Z.)
| | - Mohamed S. Dabour
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA; (M.S.D.); (B.N.Z.)
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tanta University, Tanta 31111, Egypt
| | - Eman Rashad
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt;
| | - Beshay N. Zordoky
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA; (M.S.D.); (B.N.Z.)
| |
Collapse
|
13
|
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
|
14
|
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
|
15
|
Wolfes J, Uphoff J, Kemena S, Wegner F, Rath B, Eckardt L, Frommeyer G, Ellermann C. Divergent electrophysiologic action of dapagliflozin and empagliflozin on ventricular and atrial tachyarrhythmias in isolated rabbit hearts. Front Cardiovasc Med 2024; 11:1369250. [PMID: 38455723 PMCID: PMC10918010 DOI: 10.3389/fcvm.2024.1369250] [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: 01/11/2024] [Accepted: 02/12/2024] [Indexed: 03/09/2024] Open
Abstract
Background The use of SGLT-2 inhibitors has revolutionized heart failure therapy. Evidence suggests a reduced incidence of ventricular and atrial arrhythmias in patients with dapagliflozin or empagliflozin treatment. It is unclear to what extent the reduced arrhythmia burden is due to direct effects of the SGLT2 inhibitors or is solely a marker of improved cardiac function. Methods One hundred five rabbit hearts were allocated to eight groups and retrogradely perfused, employing a Langendorff setup. Action potential duration at 90% of repolarization (APD90), QT intervals, effective refractory periods, conduction velocity, and dispersion of repolarization were obtained with monophasic action potential catheters. A model for tachyarrhythmias was established with the IKr blocker erythromycin for QT prolongation associated proarrhythmia as well as the potassium channel opener pinacidil for a short-QT model. An atrial fibrillation (AF) model was created with isoproterenol and acetylcholine. With increasing concentrations of both SGLT2 inhibitors, reductions in QT intervals and APD90 were observed, accompanied by a slight increase in ventricular arrhythmia episodes. During drug-induced proarrhythmia, empagliflozin succeeded in decreasing QT intervals, APD90, and VT burden whereas dapagliflozin demonstrated no significant effects. In the presence of pinacidil induced arrhythmogenicity, neither SGLT2 inhibitor had a significant impact on cardiac electrophysiology. In the AF setting, perfusion with dapagliflozin showed significant suppression of AF in the course of restitution of electrophysiological parameters whereas empagliflozin showed no significant effect on atrial fibrillation incidence. Conclusion In this model, empagliflozin and dapagliflozin demonstrated opposite antiarrhythmic properties. Empagliflozin reduced ventricular tachyarrhythmias whereas dapagliflozin showed effective suppression of atrial arrhythmias.
Collapse
Affiliation(s)
- Julian Wolfes
- Department of Cardiology II, Electrophysiology, University Hospital Münster, Münster, Germany
| | | | | | | | | | | | | | | |
Collapse
|
16
|
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
|
17
|
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
|
18
|
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
|
19
|
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
|
20
|
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
|
21
|
Das B. Pharmacotherapy for Cancer Treatment-Related Cardiac Dysfunction and Heart Failure in Childhood Cancer Survivors. Paediatr Drugs 2023; 25:695-707. [PMID: 37639193 DOI: 10.1007/s40272-023-00585-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/06/2023] [Indexed: 08/29/2023]
Abstract
The number of childhood cancer survivors is increasing rapidly. According to American Association for Cancer Research, there are more than 750,000 childhood cancer survivors in the United States and Europe. As the number of childhood cancer survivors increases, so does cancer treatment-related cardiac dysfunction (CTRCD), leading to heart failure (HF). It has been reported that childhood cancer survivors who received anthracyclines are 15 times more likely to have late cancer treatment-related HF and have a 5-fold higher risk of death from cardiovascular (CV) disease than the general population. CV disease is the leading cause of death in childhood cancer survivors. The increasing need to manage cancer survivor patients has led to the rapid creation and adaptation of cardio-oncology. Cardio-oncology is a multidisciplinary science that monitors, treats, and prevents CTRCD. Many guidelines and position statements have been published to help diagnose and manage CTRCD, including those from the American Society of Clinical Oncology, the European Society of Cardiology, the Canadian Cardiovascular Society, the European Society of Medical Oncology, the International Late Effects of Childhood Cancer Guideline Harmonization Group, and many others. However, there remains a gap in identifying high-risk patients likely to develop cardiomyopathy and HF in later life, thus reducing primary and secondary measures being instituted, and when to start treatment when there is echocardiographic evidence of left ventricular (LV) dysfunctions without symptoms of HF. There are no randomized controlled clinical trials for treatment for CTRCD leading to HF in childhood cancer survivors. The treatment of HF due to cancer treatment is similar to the guidelines for general HF. This review describes the latest pharmacologic therapy for preventing and treating LV dysfunction and HF in childhood cancer survivors based on expert consensus guidelines and extrapolating data from adult HF trials.
Collapse
Affiliation(s)
- Bibhuti Das
- Division of Pediatric Cardiology, Department of Pediatrics, Baylor Scott and White McLane Children's Medical Center, Temple, TX, 76502, USA.
| |
Collapse
|
22
|
Barış VÖ, Dinçsoy AB, Gedikli E, Zırh S, Müftüoğlu S, Erdem A. Empagliflozin attenuates doxorubicin-induced cardiotoxicity. Toxicol Res (Camb) 2023; 12:998. [PMID: 37915469 PMCID: PMC10615805 DOI: 10.1093/toxres/tfad071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/05/2023] [Indexed: 11/03/2023] Open
Affiliation(s)
- Veysel Özgür Barış
- Department of Cardiology, Dr. Ersin Arslan Research and Education Hospital, Gaziantep 27010, Turkey
- Department of Physiology, Hacettepe University Faculty of Medicine, Ankara 06230, Turkey
| | - Adnan Berk Dinçsoy
- Department of Physiology, Hacettepe University Faculty of Medicine, Ankara 06230, Turkey
| | - Esra Gedikli
- Department of Physiology, Hacettepe University Faculty of Medicine, Ankara 06230, Turkey
| | - Selim Zırh
- Department of Histology, Hacettepe University Faculty of Medicine, Ankara 06230, Turkey
| | - Sevda Müftüoğlu
- Department of Histology, Hacettepe University Faculty of Medicine, Ankara 06230, Turkey
| | - Ayşen Erdem
- Department of Physiology, Hacettepe University Faculty of Medicine, Ankara 06230, Turkey
| |
Collapse
|
23
|
Faggiano A, Gherbesi E, Cardinale D, Vicenzi M, Carugo S. SGLT2-i prevent left ventricular dysfunction induced by anthracycline in mouse model: A systematic-review and meta-analysis. Vascul Pharmacol 2023; 150:107171. [PMID: 37061151 DOI: 10.1016/j.vph.2023.107171] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 04/06/2023] [Accepted: 04/12/2023] [Indexed: 04/17/2023]
Abstract
CLINICAL QUESTION Could SGLT2-i be helpful for the prevention of left ventricular dysfunction induced by anthracycline? WHAT IS THE MAIN FINDING?: SGLT2-i appear effective for the prevention of left ventricular dysfunction induced by anthracycline in mouse model.
Collapse
Affiliation(s)
- Andrea Faggiano
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy.
| | - Elisa Gherbesi
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Daniela Cardinale
- Cardioncology Unit, European Institute of Oncology, IRCCS, Milan, Italy
| | - Marco Vicenzi
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Stefano Carugo
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| |
Collapse
|
24
|
Du Y, Wu T. Heart failure and cancer: From active exposure to passive adaption. Front Cardiovasc Med 2022; 9:992011. [PMID: 36304546 PMCID: PMC9592839 DOI: 10.3389/fcvm.2022.992011] [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: 07/12/2022] [Accepted: 09/20/2022] [Indexed: 12/06/2022] Open
Abstract
The human body seems like a "balance integrator." On the one hand, the body constantly actively receives various outside stimuli and signals to induce changes. On the other hand, several internal regulations would be initiated to adapt to these changes. In most cases, the body could keep the balance in vitro and in vivo to reach a healthy body. However, in some cases, the body can only get to a pathological balance. Actively exposed to unhealthy lifestyles and passively adapting to individual primary diseases lead to a similarly inner environment for both heart failure and cancer. To cope with these stimuli, the body must activate the system regulation mechanism and face the mutual interference. This review summarized the association between heart failure and cancer from active exposure to passive adaption. Moreover, we hope to inspire researchers to contemplate these two diseases from the angle of overall body consideration.
Collapse
Affiliation(s)
- Yantao Du
- Ningbo Institute of Medical Science, The Affiliated Hospital of Medical School of Ningbo University, Ningbo, Zhejiang, China
| | - Tao Wu
- Department of Cardiovascular Center, The Affiliated Hospital of Medical School of Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
25
|
Vuong JT, Stein-Merlob AF, Cheng RK, Yang EH. Novel Therapeutics for Anthracycline Induced Cardiotoxicity. Front Cardiovasc Med 2022; 9:863314. [PMID: 35528842 PMCID: PMC9072636 DOI: 10.3389/fcvm.2022.863314] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/14/2022] [Indexed: 01/04/2023] Open
Abstract
Anthracyclines remain an essential component of the treatment of many hematologic and solid organ malignancies, but has important implications on cardiovascular disease. Anthracycline induced cardiotoxicity (AIC) ranges from asymptomatic LV dysfunction to highly morbid end- stage heart failure. As cancer survivorship improves, the detection and treatment of AIC becomes more crucial to improve patient outcomes. Current treatment modalities for AIC have been largely extrapolated from treatment of conventional heart failure, but developing effective therapies specific to AIC is an area of growing research interest. This review summarizes the current evidence behind the use of neurohormonal agents, dexrazoxane, and resynchronization therapy in AIC, evaluates the clinical outcomes of advanced therapy and heart transplantation in AIC, and explores future horizons for treatment utilizing gene therapy, stem cell therapy, and mechanism-specific targets.
Collapse
Affiliation(s)
- Jacqueline T. Vuong
- Department of Medicine, Ronald Reagan UCLA Medical Center, Los Angeles, CA, United States
| | - Ashley F. Stein-Merlob
- Division of Cardiology, Department of Medicine, Ronald Reagan UCLA Medical Center, Los Angeles, CA, United States
| | - Richard K. Cheng
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, United States
| | - Eric H. Yang
- Division of Cardiology, Department of Medicine, Ronald Reagan UCLA Medical Center, Los Angeles, CA, United States
- UCLA Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- *Correspondence: Eric H. Yang,
| |
Collapse
|
26
|
Morelli MB, Bongiovanni C, Da Pra S, Miano C, Sacchi F, Lauriola M, D’Uva G. Cardiotoxicity of Anticancer Drugs: Molecular Mechanisms and Strategies for Cardioprotection. Front Cardiovasc Med 2022; 9:847012. [PMID: 35497981 PMCID: PMC9051244 DOI: 10.3389/fcvm.2022.847012] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 03/03/2022] [Indexed: 12/13/2022] Open
Abstract
Chemotherapy and targeted therapies have significantly improved the prognosis of oncology patients. However, these antineoplastic treatments may also induce adverse cardiovascular effects, which may lead to acute or delayed onset of cardiac dysfunction. These common cardiovascular complications, commonly referred to as cardiotoxicity, not only may require the modification, suspension, or withdrawal of life-saving antineoplastic therapies, with the risk of reducing their efficacy, but can also strongly impact the quality of life and overall survival, regardless of the oncological prognosis. The onset of cardiotoxicity may depend on the class, dose, route, and duration of administration of anticancer drugs, as well as on individual risk factors. Importantly, the cardiotoxic side effects may be reversible, if cardiac function is restored upon discontinuation of the therapy, or irreversible, characterized by injury and loss of cardiac muscle cells. Subclinical myocardial dysfunction induced by anticancer therapies may also subsequently evolve in symptomatic congestive heart failure. Hence, there is an urgent need for cardioprotective therapies to reduce the clinical and subclinical cardiotoxicity onset and progression and to limit the acute or chronic manifestation of cardiac damages. In this review, we summarize the knowledge regarding the cellular and molecular mechanisms contributing to the onset of cardiotoxicity associated with common classes of chemotherapy and targeted therapy drugs. Furthermore, we describe and discuss current and potential strategies to cope with the cardiotoxic side effects as well as cardioprotective preventive approaches that may be useful to flank anticancer therapies.
Collapse
Affiliation(s)
| | - Chiara Bongiovanni
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Silvia Da Pra
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Carmen Miano
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
| | - Francesca Sacchi
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Mattia Lauriola
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Gabriele D’Uva
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
- *Correspondence: Gabriele D’Uva,
| |
Collapse
|
27
|
Choksey A, Timm KN. Cancer Therapy-Induced Cardiotoxicity-A Metabolic Perspective on Pathogenesis, Diagnosis and Therapy. Int J Mol Sci 2021; 23:441. [PMID: 35008867 PMCID: PMC8745714 DOI: 10.3390/ijms23010441] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/21/2021] [Accepted: 12/28/2021] [Indexed: 12/13/2022] Open
Abstract
Long-term cardiovascular complications of cancer therapy are becoming ever more prevalent due to increased numbers of cancer survivors. Cancer therapy-induced cardiotoxicity (CTIC) is an incompletely understood consequence of various chemotherapies, targeted anti-cancer agents and radiation therapy. It is typically detected clinically by a reduction in cardiac left ventricular ejection fraction, assessed by echocardiography. However, once cardiac functional decline is apparent, this indicates irreversible cardiac damage, highlighting a need for the development of diagnostics which can detect CTIC prior to the onset of functional decline. There is increasing evidence to suggest that pathological alterations to cardiac metabolism play a crucial role in the development of CTIC. This review discusses the metabolic alterations and mechanisms which occur in the development of CTIC, with a focus on doxorubicin, trastuzumab, imatinib, ponatinib, sunitinib and radiotherapy. Potential methods to diagnose and predict CTIC prior to functional cardiac decline in the clinic are evaluated, with a view to both biomarker and imaging-based approaches. Finally, the therapeutic potential of therapies which manipulate cardiac metabolism in the context of adjuvant cardioprotection against CTIC is examined. Together, an integrated view of the role of metabolism in pathogenesis, diagnosis and treatment is presented.
Collapse
Affiliation(s)
- Anurag Choksey
- Somerville College, University of Oxford, Woodstock Road, Oxford OX2 6HD, UK;
| | - Kerstin N. Timm
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| |
Collapse
|