1
|
Armillotta M, Angeli F, Paolisso P, Belmonte M, Raschi E, Di Dalmazi G, Amicone S, Canton L, Fedele D, Suma N, Foà A, Bergamaschi L, Pizzi C. Cardiovascular therapeutic targets of sodium-glucose co-transporter 2 (SGLT2) inhibitors beyond heart failure. Pharmacol Ther 2025; 270:108861. [PMID: 40245989 DOI: 10.1016/j.pharmthera.2025.108861] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 02/12/2025] [Accepted: 04/14/2025] [Indexed: 04/19/2025]
Abstract
Sodium-glucose co-transporter 2 (SGLT2) inhibitors are oral antidiabetic agents that have shown significant improvements in cardiovascular and renal outcomes among patients with heart failure (HF), regardless of diabetic status, establishing them as a cornerstone therapy. In addition to glycemic control and the osmotic diuretic effect, the inhibition of SGLT2 improves endothelial function and vasodilation, optimizing myocardial energy metabolism and preserving cardiac contractility. Moreover, SGLT2 inhibitors may exhibit anti-inflammatory properties and attenuate acute myocardial ischemia/reperfusion injury, thereby reducing cardiac infarct size, enhancing left ventricular function, and mitigating arrhythmias. These pleiotropic effects have demonstrated efficacy across various cardiovascular conditions, ranging from acute to chronic coronary syndromes and extending to arrhythmias, valvular heart disease, cardiomyopathies, cardio-oncology, and cerebrovascular disease. This review provides an overview of the current literature on the potential mechanisms underlying the effectiveness of SGLT2 inhibitors across a wide range of cardiovascular diseases beyond HF.
Collapse
Affiliation(s)
- Matteo Armillotta
- Department of Medical and Surgical Sciences - DIMEC - Alma Mater Studiorum, University of Bologna, Bologna, Italy; Cardiovascular Division, Morgagni-Pierantoni University Hospital, Forlì, Italy
| | - Francesco Angeli
- Department of Medical and Surgical Sciences - DIMEC - Alma Mater Studiorum, University of Bologna, Bologna, Italy; Cardiovascular Division, Morgagni-Pierantoni University Hospital, Forlì, Italy
| | | | - Marta Belmonte
- Cardiology Unit, Sant'Andrea University Hospital, Rome, Italy; Department of Advanced Biomedical Sciences, University Federico II, Naples, Italy
| | - Emanuel Raschi
- Department of Medical and Surgical Sciences - DIMEC - Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Guido Di Dalmazi
- Department of Medical and Surgical Sciences - DIMEC - Alma Mater Studiorum, University of Bologna, Bologna, Italy; Division of Endocrinology and Diabetes Prevention and Care Unit, IRCCS, University Hospital of Bologna, Bologna, Italy
| | - Sara Amicone
- Department of Medical and Surgical Sciences - DIMEC - Alma Mater Studiorum, University of Bologna, Bologna, Italy; Cardiovascular Division, Morgagni-Pierantoni University Hospital, Forlì, Italy
| | - Lisa Canton
- Department of Medical and Surgical Sciences - DIMEC - Alma Mater Studiorum, University of Bologna, Bologna, Italy; Cardiovascular Division, Morgagni-Pierantoni University Hospital, Forlì, Italy
| | - Damiano Fedele
- Department of Medical and Surgical Sciences - DIMEC - Alma Mater Studiorum, University of Bologna, Bologna, Italy; Cardiovascular Division, Morgagni-Pierantoni University Hospital, Forlì, Italy
| | - Nicole Suma
- Department of Medical and Surgical Sciences - DIMEC - Alma Mater Studiorum, University of Bologna, Bologna, Italy; Cardiology Unit, IRCCS Azienda Ospedaliera-Universitaria di Bologna, Bologna, Italy
| | - Alberto Foà
- Department of Medical and Surgical Sciences - DIMEC - Alma Mater Studiorum, University of Bologna, Bologna, Italy; Cardiology Unit, IRCCS Azienda Ospedaliera-Universitaria di Bologna, Bologna, Italy
| | - Luca Bergamaschi
- Department of Medical and Surgical Sciences - DIMEC - Alma Mater Studiorum, University of Bologna, Bologna, Italy; Cardiovascular Division, Morgagni-Pierantoni University Hospital, Forlì, Italy
| | - Carmine Pizzi
- Department of Medical and Surgical Sciences - DIMEC - Alma Mater Studiorum, University of Bologna, Bologna, Italy; Cardiovascular Division, Morgagni-Pierantoni University Hospital, Forlì, Italy.
| |
Collapse
|
2
|
Zhang N, Tian X, Sun D, Tse G, Xie B, Zhao Z, Liu T. Clonal hematopoiesis, cardiovascular disease and cancer treatment-induced cardiotoxicity. Semin Cancer Biol 2025; 111:89-114. [PMID: 40023267 DOI: 10.1016/j.semcancer.2025.02.007] [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: 04/23/2024] [Revised: 01/05/2025] [Accepted: 02/06/2025] [Indexed: 03/04/2025]
Abstract
Clonal hematopoiesis (CH) arises when a substantial proportion of mature blood cells is derived from a single hematopoietic stem cell lineage. It is considered to be a premalignant state that predisposes individuals to an increased risk of cancers. Recently, emerging evidence has demonstrated a strong association between CH and both the incidence and mortality of cardiovascular diseases (CVD), with the relative risks being comparable to those attributed to traditional cardiovascular risk factors. In addition, CH has been suggested to play a role in CVD and anti-cancer treatment-related cardiotoxicity amongst cancer survivors. Moreover, certain forms of chemotherapy and radiation therapy have been shown to promote the clonal expansion of specific CH-related mutations. Consequently, CH may play a substantial role in the realm of cardio-oncology. In this review, we discuss the association between CH with cancer and CVD, with a special focus on anti-cancer treatment-related cardiotoxicity, discuss possible future research avenues and propose a systematic approach for clinical practice.
Collapse
Affiliation(s)
- Nan Zhang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Xu Tian
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Dongkun Sun
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Gary Tse
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China; School of Nursing and Health Studies, Hong Kong Metropolitan University, Hong Kong, China
| | - Bingxin Xie
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Zhiqiang Zhao
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Tong Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China.
| |
Collapse
|
3
|
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 PMCID: PMC12107967 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] [Download PDF] [Figures] [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
|
4
|
Cheang I, Zhu X, Lu X, Li Y, Ni G, Yang Y, Zhang Y, Ren QW, Wu MZ, Gao R, Yiu KH, Li X. Interaction of Body Mass Index and Glycemic Status on Cardiovascular Outcomes in Patients With Cancer Treated With Anthracyclines. J Am Heart Assoc 2025:e040876. [PMID: 40401613 DOI: 10.1161/jaha.124.040876] [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: 12/27/2024] [Accepted: 04/16/2025] [Indexed: 05/23/2025]
Abstract
BACKGROUND Anthracycline-based chemotherapy is a vital treatment for various cancers but carries notable risks of cardiotoxicity. This study aimed to assess how different body mass index values and glycemic status influence the risk of major adverse cardiovascular events (MACE) in chemotherapy-naïve adult patients with cancer treated with anthracyclines. METHODS This retrospective cohort included 11 393 chemotherapy-naïve patients who initiated anthracycline-based chemotherapy between 2000 and 2019. Follow-up began from the first anthracycline dose. Body mass index was categorized as underweight/normal weight (<25 kg/m2), overweight (25-29.9 kg/m2), and obese (≥30 kg/m2). Glycemic status was classified as normoglycemic or diabetes/prediabetes (diabetes: hemoglobin A1c ≥6.5% or fasting glucose ≥126 mg/dL; prediabetes: hemoglobin A1c 5.7%-6.4% or fasting glucose 100-125 mg/dL). RESULTS Over a median follow-up of 8.7 years, 985 (8.64%) patients experienced MACE. Obesity was significantly associated with an increased risk of MACE (hazard ratio [HR], 1.38 [95% CI, 1.10-1.73], reference: underweight/normal weight) and heart failure hospitalization, and diabetes/prediabetes also significantly predicted MACE (HR, 1.28 [95% CI, 1.10-1.50], reference: normoglycemic). Notably, overweight (HR, 0.85 [95% CI, 0.80-0.91]) and obesity (HR, 0.85 [95% CI, 0.74-0.96]) were associated with lower risk of all-cause mortality. Joint analysis revealed that patients with both obesity and diabetes/prediabetes had the highest risk of MACE (HR, 1.74 [95% CI, 1.28-2.37]) and heart failure hospitalization (HR, 1.99 [95% CI, 1.41-2.81]). CONCLUSIONS In patients with cancer undergoing anthracycline-based chemotherapy, both body mass index and glycemic status significantly affect cardiovascular risks, with the highest risk observed in those with concurrent obesity and diabetes/prediabetes, emphasizing the need for tailored risk assessment and management.
Collapse
Affiliation(s)
- Iokfai Cheang
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Department of Cardiology The First Affiliated Hospital with Nanjing Medical University, Jiangsu Province Hospital Nanjing 210029 China
- Division of Cardiology, Department of Medicine The University of Hong Kong-ShenZhen Hospital Shenzhen 518009 China
- Cardiology Division, Department of Medicine The University of Hong Kong, Queen Mary Hospital Hong Kong 999077 China
| | - Xu Zhu
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Department of Cardiology The First Affiliated Hospital with Nanjing Medical University, Jiangsu Province Hospital Nanjing 210029 China
| | - Xinyi Lu
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Department of Cardiology The First Affiliated Hospital with Nanjing Medical University, Jiangsu Province Hospital Nanjing 210029 China
| | - Ying Li
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Department of Cardiology The First Affiliated Hospital with Nanjing Medical University, Jiangsu Province Hospital Nanjing 210029 China
| | - Gehui Ni
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Department of Cardiology The First Affiliated Hospital with Nanjing Medical University, Jiangsu Province Hospital Nanjing 210029 China
| | - Ying Yang
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Department of Cardiology The First Affiliated Hospital with Nanjing Medical University, Jiangsu Province Hospital Nanjing 210029 China
| | - Yue Zhang
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Department of Cardiology The First Affiliated Hospital with Nanjing Medical University, Jiangsu Province Hospital Nanjing 210029 China
| | - Qing-Wen Ren
- Division of Cardiology, Department of Medicine The University of Hong Kong-ShenZhen Hospital Shenzhen 518009 China
- Cardiology Division, Department of Medicine The University of Hong Kong, Queen Mary Hospital Hong Kong 999077 China
| | - Mei-Zhen Wu
- Division of Cardiology, Department of Medicine The University of Hong Kong-ShenZhen Hospital Shenzhen 518009 China
- Cardiology Division, Department of Medicine The University of Hong Kong, Queen Mary Hospital Hong Kong 999077 China
| | - Rongrong Gao
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Department of Cardiology The First Affiliated Hospital with Nanjing Medical University, Jiangsu Province Hospital Nanjing 210029 China
| | - Kai-Hang Yiu
- Division of Cardiology, Department of Medicine The University of Hong Kong-ShenZhen Hospital Shenzhen 518009 China
- Cardiology Division, Department of Medicine The University of Hong Kong, Queen Mary Hospital Hong Kong 999077 China
| | - Xinli Li
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Department of Cardiology The First Affiliated Hospital with Nanjing Medical University, Jiangsu Province Hospital Nanjing 210029 China
| |
Collapse
|
5
|
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 PMCID: PMC12112039 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] [Download PDF] [Figures] [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; (L.T.); (A.N.)
| | - 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; (L.T.); (A.N.)
| | - 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
|
6
|
She RL, Liang XY, Hu L, Ma CY, Mu LY, Feng JH, Song JY, Jiang ZY, Li ZX, Qu XQ, Peng BQ, Wu KN, Kong LQ. Effect of chemotherapy and different chemotherapeutic regimens on electrocardiographic parameters in breast cancer women: a retrospective and within-subject longitudinal study. Support Care Cancer 2025; 33:452. [PMID: 40327163 DOI: 10.1007/s00520-025-09499-2] [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: 11/19/2024] [Accepted: 04/28/2025] [Indexed: 05/07/2025]
Abstract
BACKGROUND Descriptions of the effect of chemotherapy on all the electrocardiographic parameters in breast cancer women during chemotherapy are limited. METHODS A retrospective and within-subject longitudinal study of the effect of chemotherapeutics and different chemotherapeutic regimens on electrocardiographic parameters was conducted in 948 breast cancer women who had completed chemotherapy with ECG recording at initial diagnosis and before each cycle of chemotherpy. Heart rate (HR), QRS interval, P-R interval, QRS axis, QT interval, QTc interval, and the incidence of QTc interval prolongation were analyzed. Age, body mass index (BMI), history of hypertension, diabetes, and coronary heart disease were also collected for further stratified study. RESULTS Compared to initial diagnosis, changes in HR [74 (67, 82) bpm vs. 79 (73, 87)bpm], P-R interval [148 (136, 160) ms vs. 150 (138, 162) ms], QRS axis [41° (19.25°°, 60°) vs. 33° (15°, 53.75°)], QT interval [376 (358, 394) ms vs. 372 (354, 386) ms], QTc interval [417 (404, 431) ms vs. 426 (414, 436) ms], and incidence of QTc interval prolongation (9.6% vs. 15.8%) were all significant after chemotherapy, P < 0.001. There were statistically differences in HR, QRS axis, QTc interval, and the incidence of QTc interval prolongation between initial diagnosis and prechemotherapy of the last cycle under different age strata (≤ 45 years, 45 ~ 55 years, ≥ 55 years), different BMI range groups (18.5-22.9 kg/m2, 23-24.9 kg/m2, and 25-29.9 kg/m2), and even in patients without history of hypertension, diabetes, or coronary heart disease, respectively, P < 0.05. Resting HR, QRS axis, and QTc interval between each cycle of TEC regimen were different, P < 0.001. Resting HR and QTc interval between different cycles of EC-T(H) regimen were different, P < 0.05. Compared to initial diagnosis, a longer QTc interval occurred from the third to the last cycle of TEC regimen, P < 0.05. Only QTc interval before the fifth cycle of EC-T(H) regimen was statistically different from that at initial diagnosis, P = 0.012. CONCLUSION Chemotherapy affects the ECG parameters of HR, P-R interval, QRS axis, QT interval, QTc interval, and QTc interval prolongation in early-stage breast cancer women during chemotherapy. Electrocardiographic parameters may be affected significantly by TEC regimen than by EC-T(H) or TC regimen. Trial registration Retrospectively registered.
Collapse
Affiliation(s)
- Rui-Ling She
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xin-Yu Liang
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Lei Hu
- Information Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Chen-Yu Ma
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Li-Yuan Mu
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jun-Han Feng
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jing-Yu Song
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhi-Yu Jiang
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhao-Xing Li
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiu-Quan Qu
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Bai-Qing Peng
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Kai-Nan Wu
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Ling-Quan Kong
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
7
|
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 PMCID: PMC12115274 DOI: 10.3390/ph18050681] [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: 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; (I.-D.G.); (V.G.C.); (V.S.I.); (R.B.); (O.T.); (D.-F.L.)
- 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; (I.-D.G.); (V.G.C.); (V.S.I.); (R.B.); (O.T.); (D.-F.L.)
- 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; (I.-D.G.); (V.G.C.); (V.S.I.); (R.B.); (O.T.); (D.-F.L.)
- 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; (I.-D.G.); (V.G.C.); (V.S.I.); (R.B.); (O.T.); (D.-F.L.)
- 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; (I.-D.G.); (V.G.C.); (V.S.I.); (R.B.); (O.T.); (D.-F.L.)
- 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; (I.-D.G.); (V.G.C.); (V.S.I.); (R.B.); (O.T.); (D.-F.L.)
- Advanced Cardiology and Hemostaseology Research Center, “Victor Babes” University of Medicine and Pharmacy, No. 2 Eftimie Murgu Square, 300041 Timisoara, Romania
| |
Collapse
|
8
|
Braun VM, Foryst-Ludwig A, Landmesser U, Baczkó I, Milting H, Kintscher U, Beyhoff N. Cancer therapy-related cardiotoxicity is associated with distinct alterations of the myocardial lipidome. Eur J Heart Fail 2025. [PMID: 40312107 DOI: 10.1002/ejhf.3656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 03/05/2025] [Accepted: 03/18/2025] [Indexed: 05/03/2025] Open
Abstract
AIMS Anthracyclines are key components of various chemotherapy regimens, but their clinical utility is limited by severe cardiotoxic side effects. Previous studies have suggested that anthracycline-induced cardiotoxicity (AIC) may be driven by alterations in myocardial lipid metabolism. This study aimed to systematically explore the cardiac lipidomic landscape of AIC with regard to potential pathomechanisms and novel therapeutic targets. METHODS AND RESULTS Mass spectrometry-based untargeted lipidomics were performed on myocardial biopsies from 13 patients with AIC (age 53 ± 31 years, 54% female, left ventricular ejection fraction 19 ± 4%) and 15 age- and sex-matched controls. Lipidomic profiles were also compared with 15 patients with other heart failure aetiologies (matched for sex and age). A total of 627 individual lipid species from 22 different lipid classes were analysed. AIC was associated with a lower proportion of polyunsaturated fatty acids towards more monounsaturated and saturated sidechains as well as significant alterations in the proportion of odd-chain fatty acids. Pathway analyses indicated a higher precursor conversion into lysolipids in AIC. This accumulation of lysolipid species was not observed in other heart failure aetiologies and may represent a specific finding in AIC. CONCLUSION Anthracycline-induced cardiotoxicity leads to distinct alterations of the myocardial lipidome. Increased levels of myocardial lysolipids were identified as a novel lipidomic trait in AIC, which appears to be distinct from other causes of heart failure. Further research studying pharmacological interventions in lysolipid metabolism for prevention and therapy of AIC is warranted.
Collapse
Affiliation(s)
- Vera M Braun
- Institute of Pharmacology, Max Rubner Center for Cardiovascular Metabolic Renal Research, Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin, Germany
| | - Anna Foryst-Ludwig
- Institute of Pharmacology, Max Rubner Center for Cardiovascular Metabolic Renal Research, Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin, Germany
| | - Ulf Landmesser
- DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin, Germany
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Berlin, Germany
| | - István Baczkó
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Hendrik Milting
- Erich and Hanna Klessmann Institute for Cardiovascular Research and Development, Clinic for Thoracic and Cardiovascular Surgery, Heart and Diabetes Center NRW, Bad Oeynhausen, Germany
| | - Ulrich Kintscher
- Institute of Pharmacology, Max Rubner Center for Cardiovascular Metabolic Renal Research, Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin, Germany
| | - Niklas Beyhoff
- Institute of Pharmacology, Max Rubner Center for Cardiovascular Metabolic Renal Research, Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin, Germany
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Berlin, Germany
| |
Collapse
|
9
|
Zhao Z, Zheng N, Zhang T, Zhang C, Li Y, Lan M, Zhang N, Li H, Ai H, Liu D. Cardiorenal protection with dapagliflozin in patients with type 2 diabetes mellitus and chronic coronary syndrome undergoing percutaneous coronary intervention: a registry cross-sectional study. Cardiovasc Diabetol 2025; 24:185. [PMID: 40287715 PMCID: PMC12034169 DOI: 10.1186/s12933-025-02678-9] [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: 11/02/2024] [Accepted: 03/08/2025] [Indexed: 04/29/2025] Open
Abstract
IMPORTANCE Although sodium‒glucose cotransporter-2 (SGLT2) inhibitors have cardiorenal benefits, their efficacy in patients with type 2 diabetes mellitus (T2DM) and chronic coronary syndrome (CCS) undergoing percutaneous coronary intervention (PCI) remains underexplored. OBJECTIVE To evaluate the cardiorenal protective effects of the SGLT2 inhibitor dapagliflozin in patients with T2DM and CCS receiving PCI. DESIGN, SETTING, AND PARTICIPANTS This was a cross-sectional analysis of 1,430 patients from a tertiary hospital database who underwent PCI (January 1, 2018, to March 31, 2022). MAIN OUTCOMES AND MEASURES Cardiac outcomes (PMI/4aMI) and renal outcomes (eGFR and CI-AKI). RESULTS After 1:1 propensity score matching (PSM) (176 dapagliflozin vs. 176 control), the dapagliflozin group showed significantly lower PMI/4aMI rates pre-PSM (39.78% vs. 66.99%; OR 0.862, 95% CI 0.823-0.904; p < 0.001) and post-PSM (39.77% vs. 60.23%; OR 0.660, 95% CI 0.531-0.821; p < 0.001), with sustained significance after adjustment (adjusted OR 0.436, 95% CI 0.285-0.668; p < 0.001). Subgroup analyses highlighted increased protection in patients aged ≥ 65 years, those with multivessel disease, and those with higher contrast volumes. Renal outcomes (CI-AKIESUR and CI-AKIKDIGOs) were not significantly different before or after PSM or after adjustment (all p > 0.05). CONCLUSIONS AND RELEVANCE Dapagliflozin exerted robust cardioprotective effects against PMI/4aMI in patients with T2DM and CCS undergoing PCI, particularly among patients in high-risk subgroups, but it did not significantly reduce the risk of CI-AKI. These findings support the peri-PCI use of dapagliflozin to mitigate cardiac risk while highlighting the need for further research to elucidate its renal effects in this population.
Collapse
Affiliation(s)
- Zinan Zhao
- Department of Pharmacy, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences; Beijing Key Laboratory of Assessment of Clinical Drugs Risk and Individual Application (Beijing Hospital), Beijing, People's Republic of China
| | - Naixin Zheng
- Department of Cardiology, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Tianqi Zhang
- Department of Pharmacy, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences; Beijing Key Laboratory of Assessment of Clinical Drugs Risk and Individual Application (Beijing Hospital), Beijing, People's Republic of China
| | - Chi Zhang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, People's Republic of China
| | - Yuwei Li
- Department of Cardiology, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Ming Lan
- Department of Cardiology, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Ni Zhang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Hui Li
- Department of Cardiology, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Hu Ai
- Department of Cardiology, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Deping Liu
- Department of Cardiology, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, People's Republic of China.
| |
Collapse
|
10
|
Novo G, Madaudo C, Cannatà A, Ameri P, Di Lisi D, Bromage DI, Galassi AR, Minotti G, Lyon AR. Effects of SGLT2 Inhibitors in Patients with Cancer and Diabetes Mellitus: A Systematic Review and Meta-analysis. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2025:pvaf028. [PMID: 40274419 DOI: 10.1093/ehjcvp/pvaf028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 03/17/2025] [Accepted: 03/28/2025] [Indexed: 04/26/2025]
Abstract
AIMS Cardiovascular disease (CVD) and cancer represent significant global health challenges. An overlap between oncology and cardiology is compounded by cancer therapies, which are known to have cardiotoxic effects. Sodium-glucose cotransporter 2 inhibitors (SGLT2i), initially developed for treating diabetes, have shown promising cardiovascular benefits in non-cancer populations, particularly in preventing heart failure (HF) and reducing HF-related hospitalization and mortality in large randomized controlled trials (RCTs) across the spectrum of left ventricular ejection fraction (LVEF). However, their potential cardioprotective role in cancer patients remains unclear.This systematic review and meta-analysis evaluated cardiovascular outcomes in cancer patients with type 2 diabetes undergoing chemotherapy with concomitant use of SGLT2i compared to those not using SGLT2i. Subgroup analyses were performed to explore patients without baseline HF and patients treated exclusively with anthracyclines. METHODS AND RESULTS A systematic review identified eleven observational retrospective studies (n=104,327 patients). Based on the National Institutes of Health Quality Assessment Tool (NIH-QAT) checklist, 2 studies were at moderate risk of bias, while all other included studies had a low risk of bias. Meta-analysis indicated that the use of SGLT2i was associated with a significant reduction in all-cause mortality (0.47, 95% CI 0.33-0.67, P<0.0001) and risk of HF hospitalization (0.44, 95% CI 0.27-0.72, P=0.001). CONCLUSION SGLT2i use may be associated with a significant reduction in all-cause mortality and risk of HF hospitalization in actively treated cancer patients with type 2 diabetes. Our study highlights the need for further investigation through prospective RCTs to confirm the efficacy and safety of SGLT2i in attenuating cardiotoxicity and supporting cardiovascular health in oncology settings.
Collapse
Affiliation(s)
- Giuseppina Novo
- Cardiology Unit, University Hospital "Paolo Giaccone", Palermo Italy
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE) University of Palermo, Italy
| | - Cristina Madaudo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE) University of Palermo, Italy
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine, Faculty of Life Science, King's College London, London, UK
| | - Antonio Cannatà
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine, Faculty of Life Science, King's College London, London, UK
| | - Pietro Ameri
- Department of Internal Medicine, University of Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Daniela Di Lisi
- Cardiology Unit, University Hospital "Paolo Giaccone", Palermo Italy
| | - Daniel I Bromage
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine, Faculty of Life Science, King's College London, London, UK
| | - Alfredo Ruggero Galassi
- Cardiology Unit, University Hospital "Paolo Giaccone", Palermo Italy
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE) University of Palermo, Italy
| | - Giorgio Minotti
- Department of Medicine and Unit of Clinical Pharmacology, University and Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | - Alexander R Lyon
- Cardio-Oncology Centre of Excellence, Royal Brompton Hospital, London, UK
| |
Collapse
|
11
|
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
|
12
|
Quagliariello V, Berretta M, Bisceglia I, Giacobbe I, Iovine M, Barbato M, Maurea C, Canale ML, Paccone A, Inno A, Scherillo M, Oliva S, Cadeddu Dessalvi C, Mauriello A, Fonderico C, Maratea AC, Gabrielli D, Maurea N. In the Era of Cardiovascular-Kidney-Metabolic Syndrome in Cardio-Oncology: From Pathogenesis to Prevention and Therapy. Cancers (Basel) 2025; 17:1169. [PMID: 40227756 PMCID: PMC11988012 DOI: 10.3390/cancers17071169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/21/2025] [Accepted: 03/27/2025] [Indexed: 04/15/2025] Open
Abstract
Cardiovascular-kidney-metabolic (CKM) syndrome represents a complex interplay between cardiovascular disease (CVD), chronic kidney disease (CKD), and metabolic disorders, significantly impacting cancer patients. The presence of CKM syndrome in cancer patients not only worsens their prognosis but also increases the risk of major adverse cardiovascular events (MACE), reduces quality of life (QoL), and affects overall survival (OS). Furthermore, several anticancer therapies, including anthracyclines, tyrosine kinase inhibitors, immune checkpoint inhibitors, and hormonal treatments, can exacerbate CKM syndrome by inducing cardiotoxicity, nephrotoxicity, and metabolic dysregulation. This review explores the pathophysiology of CKM syndrome in cancer patients and highlights emerging therapeutic strategies to mitigate its impact. We discuss the role of novel pharmacological interventions, including sodium-glucose cotransporter-2 inhibitors (SGLT2i), proprotein convertase subtilisin/kexin type 9 inhibitors (PCSK9i), and soluble guanylate cyclase (sGC) activators, as well as dietary and lifestyle interventions. Optimizing the management of CKM syndrome in cancer patients is crucial to improving OS, enhancing QoL, and reducing MACE. By integrating cardiometabolic therapies into oncologic care, we can create a more comprehensive treatment approach that reduces the burden of cardiovascular and renal complications in this vulnerable population. Further research is needed to establish personalized strategies for CKM syndrome prevention and treatment in cancer patients.
Collapse
Affiliation(s)
- Vincenzo Quagliariello
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Massimiliano Berretta
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
| | - Irma Bisceglia
- Servizi Cardiologici Integrati, Dipartimento Cardio-Toraco-Vascolare, Azienda Ospedaliera San Camillo Forlanini, 00148 Rome, Italy
| | - Ilaria Giacobbe
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Martina Iovine
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Matteo Barbato
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Carlo Maurea
- ASL NA1, UOC Neurology and Stroke Unit, Ospedale del Mare, 23807 Naples, Italy
| | | | - Andrea Paccone
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Alessandro Inno
- Medical Oncology, IRCCS Ospedale Sacro Cuore Don Calabria, 37024 Negrar di Valpolicella, Italy
| | - Marino Scherillo
- Cardiologia Interventistica e UTIC, A.O. San Pio, Presidio Ospedaliero Gaetano Rummo, 82100 Benevento, Italy
| | - Stefano Oliva
- Cardio-Oncology Unit, IRCCS Istituto Tumori, “Giovanni Paolo II”, 70124 Bari, Italy
| | | | - Alfredo Mauriello
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Celeste Fonderico
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Anna Chiara Maratea
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Domenico Gabrielli
- U.O.C. Cardiologia, Dipartimento Cardio-Toraco-Vascolare, Azienda Ospedaliera San Camillo Forlanini, 00152 Rome, Italy
| | - Nicola Maurea
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| |
Collapse
|
13
|
Flausino LE, Carrasco AGM, Furuya TK, Tuan WJ, Chammas R. Impact of SGLT2 inhibitors on survival in gastrointestinal cancer patients undergoing chemotherapy and/or radiotherapy: a real-world data retrospective cohort study. BMC Cancer 2025; 25:542. [PMID: 40133838 PMCID: PMC11938601 DOI: 10.1186/s12885-025-13966-8] [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: 01/11/2025] [Accepted: 03/19/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND The role of sodium-glucose co-transporter 2 inhibitor (SGLT2i) drugs in the management of diabetes and cardiovascular disease is well-established, but emerging evidence suggests potential effects on cancer outcomes, including gastrointestinal (GI) cancers. We conducted an extensive, sex-oriented, real-world data analysis to investigate whether SGLT2i can enhance GI cancer outcomes when used alongside standard therapies such as chemotherapy and radiotherapy. METHODS The study applied a retrospective cohort design with data from the TriNetX research database ( https://trinetx.com ), examining GI cancer patients treated with chemotherapy and/or radiotherapy between 2013 and 2023. The intervention cohort consisted of Gl cancer patients who received SGLT2i, while the control cohort did not. A 5-year follow-up period was used, and baseline characteristics were balanced using a 1:1 propensity score matching technique. Cox proportional-hazards and logistic regression models assessed mortality and morbidity risks between the cohorts. RESULTS The study included 6,389 male and 3,457 female patients with GI cancer (ICD-10: C15-C25). The use of SGLT2i was significantly associated with improved survival for both male (HR 0.568; 95% CI 0.534-0.605) and female (HR 0.561; 95% CI 0.513-0.614) patients undergoing chemotherapy and/or radiotherapy. SGLT2i use also correlated significantly with lower hospitalisation rates both in male (OR 0.684; 95% CI 0.637-0.734) and female (OR, 0.590; 95% CI 0.536-0.650) patients. The analysis of GI cancer subtypes also demonstrated similar benefits, without significant adverse effects. CONCLUSIONS Repurposing SGLT2 inhibitors for cancer treatment could potentially improve outcomes for GI cancer patients without causing significant side effects. Further clinical trials are needed to confirm these findings and establish the optimal condition for its application in GI cancer treatment.
Collapse
Affiliation(s)
- Lucas E Flausino
- Center for Translational Research in Oncology, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo, Brazil
| | - Alexis Germán Murillo Carrasco
- Center for Translational Research in Oncology, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo, Brazil
| | - Tatiane Katsue Furuya
- Center for Translational Research in Oncology, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo, Brazil
| | - Wen-Jan Tuan
- Department of Family and Community Medicine, and Public Health Sciences, Penn State College of Medicine, Hershey, PA, USA
| | - Roger Chammas
- Center for Translational Research in Oncology, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.
- Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo, Brazil.
| |
Collapse
|
14
|
Contaldi C, D’Aniello C, Panico D, Zito A, Calabrò P, Di Lorenzo E, Golino P, Montesarchio V. Cancer-Therapy-Related Cardiac Dysfunction: Latest Advances in Prevention and Treatment. Life (Basel) 2025; 15:471. [PMID: 40141815 PMCID: PMC11944213 DOI: 10.3390/life15030471] [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: 01/07/2025] [Revised: 02/19/2025] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
The increasing efficacy of cancer therapies has significantly improved survival rates, but it has also highlighted the prevalence of cancer-therapy-related cardiac dysfunction (CTRCD). This review provides a comprehensive overview of the identification, monitoring, and management of CTRCD, a condition resulting from several treatments, such as anthracyclines, HER2-targeted therapies, target therapies, and radiotherapy. The paper includes a discussion of the mechanisms of CTRCD associated with various cancer treatments. Early detection through serum biomarkers and advanced imaging techniques is crucial for effective monitoring and risk stratification. Preventive strategies include pharmacological interventions such as ACE inhibitors/angiotensin receptor blockers, beta-blockers, and statins. Additionally, novel agents like sacubitril/valsartan, sodium-glucose co-transporter type 2 inhibitors, and vericiguat show promise in managing left ventricular dysfunction. Lifestyle modifications, including structured exercise programs and optimized nutritional strategies, further contribute to cardioprotection. The latest treatments for both asymptomatic and symptomatic CTRCD across its various stages are also discussed. Emerging technologies, including genomics, artificial intelligence, novel biomarkers, and gene therapy, are paving the way for personalized approaches to CTRCD prevention and treatment. These advancements hold great promise for improving long-term outcomes in cancer patients by minimizing cardiovascular complications.
Collapse
Affiliation(s)
- Carla Contaldi
- Department of Cardiology, AORN dei Colli-Monaldi Hospital, 80131 Naples, Italy
| | - Carmine D’Aniello
- Division of Medical Oncology, AORN dei Colli-Monaldi Hospital, 80131 Naples, Italy
| | - Domenico Panico
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy
| | - Andrea Zito
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy
| | - Paolo Calabrò
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy
| | - Emilio Di Lorenzo
- Department of Cardiology, AORN dei Colli-Monaldi Hospital, 80131 Naples, Italy
| | - Paolo Golino
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy
| | | |
Collapse
|
15
|
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
|
16
|
Bhalraam U, Veerni RB, Paddock S, Meng J, Piepoli M, López-Fernández T, Tsampasian V, Vassiliou VS. Impact of sodium-glucose cotransporter-2 inhibitors on heart failure outcomes in cancer patients and survivors: a systematic review and meta-analysis. Eur J Prev Cardiol 2025:zwaf026. [PMID: 40044419 DOI: 10.1093/eurjpc/zwaf026] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 11/30/2024] [Accepted: 01/28/2025] [Indexed: 03/20/2025]
Abstract
AIMS Sodium-glucose cotransporter-2 inhibitors (SGLT2i) are recognized for their cardiovascular benefits. This systematic review and meta-analysis evaluated the impact of SGLT2i on heart failure (HF) outcomes in cancer patients and survivors, focusing on HF hospitalization and new HF diagnoses. METHODS AND RESULTS A comprehensive search of PubMed, MEDLINE, and Embase via Ovid, and the Cochrane Library was conducted up to 5 June 2024, focusing on studies involving cancer patients and survivors treated with SGLT2i. The search criterion used was [(SGLT2) OR (Sodium glucose cotransporter 2 inhibitors) OR (canagliflozin) OR (dapagliflozin) OR (empagliflozin) OR (ertugliflozin) AND (cancer)]. The primary outcomes assessed were HF hospitalization and new HF diagnoses. The search yielded 1880 studies, from which 13 studies encompassing 88 273 patients were included. SGLT2i use reduced HF hospitalizations by 51% (RR 0.49, 95% CI 0.36-0.66, I² = 28%, P < 0.01) and new HF diagnoses by 71% (RR 0.29, 95% CI 0.10-0.87, I² = 71%). Multi-variate meta-regression analysis suggested that among breast cancer populations, studies with ≥50% of patients on anthracyclines exhibited a 99% reduction in HF hospitalization risk compared with similar studies that included <50% of patients on anthracyclines (RR 0.0085, 95% CI: 0.0001-0.2645, P = 0.0081). CONCLUSION SGLT2i significantly lower the risk of HF hospitalization and new HF diagnoses among cancer patients and survivors, with particularly pronounced benefits in breast cancer patients receiving anthracycline-based chemotherapy. These findings support the need for prospective trials to further investigate the integration of SGLT2i into cancer patient management to enhance cardiovascular outcomes.
Collapse
Affiliation(s)
- U Bhalraam
- Department of Medicine, Norwich Medical School, University of East Anglia, Norwich, UK
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, UK
| | - Rathna B Veerni
- Department of Medicine, Ninewells Hospital and Medical School, Dundee, UK
| | - Sophie Paddock
- Department of Medicine, Norwich Medical School, University of East Anglia, Norwich, UK
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, UK
| | - James Meng
- Department of Medicine, Norwich Medical School, University of East Anglia, Norwich, UK
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, UK
| | - Massimo Piepoli
- University Cardiology Department, IRCCS Policlinico San Donato, Via Morandi 30, 20097 San Donato Milanese, Milan, Italy
- Department of Biomedical Sciences for Health, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy
| | - Teresa López-Fernández
- Cardiology Department, La Paz University Hospital, IdiPAZ Research Institute, Madrid, Spain
- Cardiology Department, Quiron Pozuelo University Hospital, Madrid, Spain
| | - Vasiliki Tsampasian
- Department of Medicine, Norwich Medical School, University of East Anglia, Norwich, UK
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, UK
| | - Vassilios S Vassiliou
- Department of Medicine, Norwich Medical School, University of East Anglia, Norwich, UK
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, UK
| |
Collapse
|
17
|
Haj-Yehia E, Michel L, Mincu RI, Rassaf T, Totzeck M. Prevention of cancer-therapy related cardiac dysfunction. Curr Heart Fail Rep 2025; 22:9. [PMID: 39969700 PMCID: PMC11839799 DOI: 10.1007/s11897-025-00697-x] [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] [Accepted: 02/10/2025] [Indexed: 02/20/2025]
Abstract
PURPOSE OF REVIEW Introduction of modern cancer therapies has led to increased survival of affected patients. With this advantage, the incidence of cancer therapy-related cardiac dysfunction (CTRCD) has increased and reasonable prevention strategies become necessary. This review outlines the major approaches to limit development and progression of CTRCD. RECENT FINDINGS A broad range of cancer therapies can provoke CTRCD ranging from mild asymptomatic forms to severe heart failure. Profound cardiological assessment of cardiovascular comorbidities before initiation of cancer therapy allows identification of cancer patients at higher risk developing CTRCD which may also require closer surveillance. Cardioprotective adjustment of cancer therapy and initiation of cardioprotective medication and lifestyle optimization prior to anti-cancer treatment additionally limit the risk of CTRCD. During therapy, regular examination of cancer patients using high-sensitive cardiological diagnostic tools as three-dimensional (3D) echocardiography and global longitudinal strain (GLS) enables early detection of mild forms of CTRCD. This allows appropriate adjustment of cancer therapy and initiation of CTRCD treatment to prevent further progression to severe forms. Cardiological risk stratification before treatment initiation, cardioprotective interventions before and during cancer therapy, along with regular monitoring of treated cancer patients, can help prevent the development of CTRCD. This maintains the antitumor effects of cancer therapy while limiting cardiotoxic side effects resulting in improved quality of life and mortality of affected cancer patients.
Collapse
Affiliation(s)
- Elias Haj-Yehia
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center Essen, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Lars Michel
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center Essen, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Raluca I Mincu
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center Essen, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center Essen, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Matthias Totzeck
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center Essen, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany.
| |
Collapse
|
18
|
Ositelu KC, Peesay T, Garcia C, Akhter N. Life's Essential 8 and Cardiovascular Disease in Breast Cancer Survivors. Curr Cardiol Rep 2025; 27:55. [PMID: 39954113 DOI: 10.1007/s11886-025-02216-9] [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] [Accepted: 02/06/2025] [Indexed: 02/17/2025]
Abstract
PURPOSE OF REVIEW To explore the role of optimal cardiovascular health as defined by the Life's Essential 8 in breast cancer survivors. RECENT FINDINGS Optimal cardiovascular health may be associated with decreased cancer mortality. Breast cancer survivors may derive additional benefit from obtaining ideal cardiovascular health as defined by the Life's Essential 8. Certain components of the Life's Essential 8 may impact cardiovascular risk but also cancer mortality, and risk for cancer therapy related cardiac dysfunction. Continued physical activity, avoidance of smoking, and control of lipids and blood pressure are beneficial in breast cancer survivors. More study is needed to define the role of anti-hyperglycemic agents, BMI, and sleep on CVD risk in breast cancer survivors. The Life's Essential 8 can be a tool to inform clinicians regarding a breast cancer survivor's disease risk and to identify potential areas of improvement.
Collapse
Affiliation(s)
- Kamari C Ositelu
- Division of Cardiovascular Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, 676 N. St. Clair, Suite 600, Chicago, IL, 60611, USA
| | - Tejasvi Peesay
- Department of Medicine, Feinberg School of Medicine, Northwestern University, 676 N. St. Clair, Suite 600, Chicago, IL, 60611, USA
| | - Carol Garcia
- Department of Medicine, Feinberg School of Medicine, Northwestern University, 676 N. St. Clair, Suite 600, Chicago, IL, 60611, USA
| | - Nausheen Akhter
- Division of Cardiovascular Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, 676 N. St. Clair, Suite 600, Chicago, IL, 60611, USA.
- Division of Cardiovascular Medicine, Northwestern University Feinberg School of Medicine, 676 N St. Clair, Suite 600, Chicago, IL, 60611, USA.
| |
Collapse
|
19
|
Wannaphut C, Wattanachayakul P, Saowapa S, Ponvilawan B, Tanariyakul M, Kewcharoen J, Yingchoncharoen P, Suenghataiphorn T, Aiumtrakul N, Acoba J. Effect on cardiovascular outcome of sodium-glucose co-transporter-2 (SGLT2) inhibitors among cancer patients treated with anthracycline: a systematic review and meta-analysis. Ecancermedicalscience 2025; 19:1844. [PMID: 40259909 PMCID: PMC12010125 DOI: 10.3332/ecancer.2025.1844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Indexed: 04/23/2025] Open
Abstract
Background/objectives Sodium-glucose-co-transporter-2 (SGLT2) inhibitors have shown benefit in reducing cardiovascular disease outcomes in diabetes patients. Anthracycline therapy is associated with a risk of cardiomyopathy. However, the impact of SGLT2 inhibitors in the prevention of cardiomyopathy and heart failure in cancer patients undergoing anthracycline treatment remains unclear. Thus, we conducted a systematic review and meta-analysis to explore the effect of the prevention of cardiovascular outcomes in patients with cancer and diabetes who had received anthracycline therapy. Methods We systematically reviewed Medline and EMBASE databases from inception to January 2024 for studies focusing on cancer patients with a history of anthracycline therapy. Eligible studies had to report relative risk (RR) with 95% confidence intervals (CIs) for the clinical endpoints of mortality outcomes and the risk of heart failure exacerbation, comparing cohorts with and without SGLT2 inhibitor use. Results Our study included four retrospective cohort studies in the meta-analysis (n = 6,708, 24% received SGLT2). There was significantly lower all-cause mortality in the SGLT2 inhibitors group (pooled RR of 0.52, 95% CI 0.35-0.77, I 2 64%). However, there were no differences in the risk of heart failure exacerbation (pooled RR of 0.67, 95% CI 0.39-1.14, I 2 17%). Conclusion Our study found that anthracycline-treated cancer patients using SGLT2 inhibitors experienced lower all-cause mortality compared to the control group. A randomised clinical trial is necessary to further elucidate these findings.
Collapse
Affiliation(s)
- Chalothorn Wannaphut
- Department of Medicine, John A. Burns School of Medicine, University of Hawai’i, Honolulu, HI, USA
| | - Phuuwadith Wattanachayakul
- Department of Medicine, Albert Einstein Healthcare Network, Philadelphia, PA, USA
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Sakditad Saowapa
- Department of Internal Medicine, Texas Tech Health Science Center, Lubbock, TX, USA
| | - Ben Ponvilawan
- Department of Internal Medicine, University of Missouri–Kansas City School of Medicine, Kansas City, MO, USA
| | - Manasawee Tanariyakul
- Department of Medicine, John A. Burns School of Medicine, University of Hawai’i, Honolulu, HI, USA
| | - Jakrin Kewcharoen
- Division of Cardiology, Department of Medicine, Loma Linda University Health, Loma Linda, CA, USA
| | | | | | - Noppawit Aiumtrakul
- Department of Medicine, John A. Burns School of Medicine, University of Hawai’i, Honolulu, HI, USA
| | - Jared Acoba
- University of Hawai’i Cancer Center, Honolulu, HI, USA
- Queen’s Medical Center, Honolulu, HI, USA
| |
Collapse
|
20
|
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: 2] [Impact Index Per Article: 2.0] [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
|
21
|
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
|
22
|
Chiang CH, Song J, Chi KY, Chang YC, Xanthavanij N, Chang Y, Hsia YP, Chiang CH, Ghamari A, Reynolds KL, Lin S, Xu XH, Neilan TG. Glucagon-like Peptide-1 Agonists Reduce Cardiovascular Events in Cancer Patients on Immune Checkpoint Inhibitors. Eur J Cancer 2025; 216:115170. [PMID: 39709670 DOI: 10.1016/j.ejca.2024.115170] [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: 08/20/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/24/2024]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are associated with an increased risk of major adverse cardiovascular events (MACE). Glucagon-like peptide-1 agonists (GLP1a), initially developed for type 2 diabetes mellitus (T2DM), have shown promising results in reducing cardiovascular events. We aimed to investigate the effect of GLP1a on cardiovascular events in patients receiving ICIs. METHODS We conducted a retrospective, propensity score-matched cohort study using the TriNetX database. We identified adults with cancer and T2DM who received ICIs between April 2013 and May 2023. The primary efficacy outcome was incident MACE, defined as a composite of myocardial infarction, need for coronary revascularization, heart failure, ischemic stroke, and cardiac arrest. The secondary efficacy outcomes were the individual components of MACE as well as myocarditis and pericarditis. Safety outcomes included the occurrence of immune-related adverse events, serious adverse events related to GLP1a use, and all-cause mortality. RESULTS We identified 7651 patients eligible for inclusion, among which 479 received GLP1a and 7172 received non-GLP1a diabetes medications. After matching (469 patients each), baseline characteristics were well-balanced. Over a median 12-month follow-up, the GLP1a cohort had a significantly lower MACE incidence than the non-GLP1a cohort (9.0 vs. 17.1 events per 100 patient-years) with a 54 % lower risk of MACE (Hazard ratio (HR),0.46 [95 % CI: 0.32-0.67]). There were reductions in myocardial infarction or need for coronary revascularization, heart failure, and all-cause mortality, with no differences in other cardiovascular events. GLP1a use did not increase risk of adverse events, including pancreatitis, biliary disease, bowel obstruction, gastroparesis, and immune-related adverse events. CONCLUSION GLP1a use in cancer patients with T2DM receiving ICIs was associated with reduced MACE and all-cause mortality without an increased risk in serious adverse events.
Collapse
Affiliation(s)
- Cho-Han Chiang
- Department of Medicine, Mount Auburn Hospital, Harvard Medical School, Cambridge, MA, USA.
| | - Junmin Song
- Department of Medicine, Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Kuan-Yu Chi
- Department of Medicine, Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yu-Cheng Chang
- Department of Medicine, Danbury Hospital, Danbury, CT, USA
| | - Nutchapon Xanthavanij
- Department of Medicine, Mount Auburn Hospital, Harvard Medical School, Cambridge, MA, USA
| | - Yu Chang
- Section of Neurosurgery, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yuan Ping Hsia
- Department of Family Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan.
| | - Cho-Hung Chiang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Azin Ghamari
- Cardiovascular Imaging Research Center, Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kerry L Reynolds
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Shuwen Lin
- Department of Oncology, Montefiore Medical Center, Montefiore Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Xiaocao Haze Xu
- Division of Hematology and Oncology, Department of Medicine, University of Vermont Medical Center, Burlington, VT, USA
| | - Tomas G Neilan
- Cardiovascular Imaging Research Center, Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
23
|
Bloom MW, Vo JB, Rodgers JE, Ferrari AM, Nohria A, Deswal A, Cheng RK, Kittleson MM, Upshaw JN, Palaskas N, Blaes A, Brown SA, Ky B, Lenihan D, Maurer MS, Fadol A, Skurka K, Cambareri C, Chauhan C, Barac A. Cardio-Oncology and Heart Failure: a Scientific Statement From the Heart Failure Society of America. J Card Fail 2025; 31:415-455. [PMID: 39419165 DOI: 10.1016/j.cardfail.2024.08.045] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 10/19/2024]
Abstract
Heart failure and cancer remain 2 of the leading causes of morbidity and mortality, and the 2 disease entities are linked in a complex manner. Patients with cancer are at increased risk of cardiovascular complications related to the cancer therapies. The presence of cardiomyopathy or heart failure in a patient with new cancer diagnosis portends a high risk for adverse oncology and cardiovascular outcomes. With the rapid growth of cancer therapies, many of which interfere with cardiovascular homeostasis, heart failure practitioners need to be familiar with prevention, risk stratification, diagnosis, and management strategies in cardio-oncology. This Heart Failure Society of America statement addresses the complexities of heart failure care among patients with active cancer diagnoses and cancer survivors. Risk stratification, monitoring and management of cardiotoxicity are presented across stages A through D heart failure, with focused discussion on heart failure with preserved ejection fraction and special populations, such as survivors of childhood and young-adulthood cancers. We provide an overview of the shared risk factors between cancer and heart failure, highlighting heart failure as a form of cardiotoxicity associated with many different cancer therapeutics. Finally, we discuss disparities in the care of patients with cancer and cardiac disease and present a framework for a multidisciplinary-team approach and critical collaboration among heart failure, oncology, palliative care, pharmacy, and nursing teams in the management of these complex patients.
Collapse
Affiliation(s)
| | - Jacqueline B Vo
- Radiation Epidemiology Branch, Division of Cancer Epidemiology & Genetics, National Cancer Institute, Bethesda, MD
| | - Jo E Rodgers
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, Chapel Hill, NC
| | - Alana M Ferrari
- Division of Hematology/ Oncology, University of Virginia Health, Charlottesville, VA
| | - Anju Nohria
- Cardio-Oncology Program, Cardiovascular Division, Brigham and Women's Hospital, Boston, MA
| | - Anita Deswal
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Richard K Cheng
- Division of Cardiology, University of Washington, Seattle, WA
| | - Michelle M Kittleson
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | | | - Nicolas Palaskas
- Department of Cardiology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Anne Blaes
- Division of Hematology/Oncology/Transplantation, University of Minnesota, Minneapolis, MN
| | - Sherry-Ann Brown
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI; Research Collaborator, Mayo Clinic, Rochester, MN
| | - Bonnie Ky
- Division of Cardiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; Thalheimer Center for Cardio-Oncology, Abramson Cancer Center and Division of Cardiovascular Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Daniel Lenihan
- Saint Francis Healthcare, Cape Girardeau, MO and the International Cardio-Oncology Society, Tampa, FL
| | - Mathew S Maurer
- Division of Cardiology, Columbia University Irving Medical Center, New York, NY
| | | | | | - Christine Cambareri
- Clinical Oncology Pharmacist, Hospital of the University of Pennsylvania, Abramson Cancer Center, Philadelphia, PA
| | | | - Ana Barac
- Department of Cardiology, Inova Schar Heart and Vascular, Inova Schar Cancer, Falls Church, VA
| |
Collapse
|
24
|
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
|
25
|
Moreno-Arciniegas A, Cádiz L, Galán-Arriola C, Clemente-Moragón A, Ibáñez B. Cardioprotection strategies for anthracycline cardiotoxicity. Basic Res Cardiol 2025; 120:71-90. [PMID: 39249555 PMCID: PMC11790697 DOI: 10.1007/s00395-024-01078-6] [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: 06/11/2024] [Revised: 08/25/2024] [Accepted: 08/26/2024] [Indexed: 09/10/2024]
Abstract
Thanks to the fantastic progress in cancer therapy options, there is a growing population of cancer survivors. This success has resulted in a need to focus much effort into improving the quality of life of this population. Cancer and cardiovascular disease share many common risk factors and have an interplay between them, with one condition mechanistically affecting the other and vice versa. Furthermore, widely prescribed cancer therapies have known toxic effects in the cardiovascular system. Anthracyclines are the paradigm of efficacious cancer therapy widely prescribed with a strong cardiotoxic potential. While some cancer therapies cardiovascular toxicities are transient, others are irreversible. There is a growing need to develop cardioprotective therapies that, when used in conjunction with cancer therapies, can prevent cardiovascular toxicity and thus improve long-term quality of life in survivors. The field has three main challenges: (i) identification of the ultimate mechanisms leading to cardiotoxicity to (ii) identify specific therapeutic targets, and (iii) more sensible diagnostic tools to early identify these conditions. In this review we will focus on the cardioprotective strategies tested and under investigation. We will focus this article into anthracycline cardiotoxicity since it is still the agent most widely prescribed, the one with higher toxic effects on the heart, and the most widely studied.
Collapse
Affiliation(s)
| | - Laura Cádiz
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Carlos Galán-Arriola
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Agustín Clemente-Moragón
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Borja Ibáñez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.
- Cardiology Department, IIS-Fundación Jiménez Díaz University Hospital, Madrid, Spain.
| |
Collapse
|
26
|
Medina-Hernández D, Cádiz L, Mastrangelo A, Moreno-Arciniegas A, Fernández Tocino M, Cueto Becerra AA, Díaz-Guerra Priego A, Skoza WA, Higuero-Verdejo MI, López-Martín GJ, Pérez-Martínez C, de Molina-Iracheta A, Caballero-Valderrama M, Sánchez-González J, Sancho D, Fuster V, Galán-Arriola C, Ibáñez B. SGLT2i Therapy Prevents Anthracycline-Induced Cardiotoxicity in a Large Animal Model by Preserving Myocardial Energetics. JACC CardioOncol 2025; 7:171-184. [PMID: 39967204 DOI: 10.1016/j.jaccao.2024.12.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Anthracycline-induced cardiotoxicity (AIC) is characterized by a disruption in myocardial metabolism. OBJECTIVES The authors used a large animal model to test sodium-glucose cotransporter inhibitor therapy to prevent AIC. METHODS Female large white pigs (n = 36) were used to identify the most translational AIC regimen: 6 triweekly intravenous doxorubicin injections (1.8 mg/kg each). Another group of 32 pigs were randomized (1:1:2) to doxorubicin plus empagliflozin 20 mg, doxorubicin plus empagliflozin 10 mg, or doxorubicin control. Pigs were serially examined using multiparametric cardiac magnetic resonance and magnetic resonance spectroscopy. At the end of the 21-week follow-up period, blood samples were obtained to measure myocardial metabolic substrate extraction, and the left ventricle was harvested and processed for analysis using metabolomics, transmission electron microscopy, mitochondrial respirometry, and histopathology. RESULTS Final left ventricular ejection fraction (LVEF), the prespecified primary outcome, was significantly higher in pigs receiving 20 mg empagliflozin than in the doxorubicin control group (median 57.5% [Q1-Q3: 55.5%-60.3%] vs 47.0% [Q1-Q3: 40.8%-47.8%]; P = 0.027). Final LVEF in pigs receiving 10 mg empagliflozin was 51% (Q1-Q3: 46.5%-55.5%; P = 0.020 vs 20 mg empagliflozin). The incidence of AIC events was 0%, 50%, and 72% in the empagliflozin 20 mg, empagliflozin 10 mg, and doxorubicin control groups, respectively. Empagliflozin 20 mg treatment resulted in enhanced ketone body consumption by the myocardium, preserved magnetic resonance spectroscopy-measured cardiac energetics, and improved mitochondrial structure and function on transmission electron microscopy and respirometry. These changes were more modest with the 10-mg empagliflozin dose. CONCLUSIONS Sodium-glucose cotransporter-2 inhibitor therapy with empagliflozin exerts a dose-dependent cardioprotective effect against AIC. The improved LVEF was accompanied by enhanced ketone body consumption, improved cardiac energetics, and preserved mitochondrial structure and function.
Collapse
Affiliation(s)
| | - Laura Cádiz
- Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Annalaura Mastrangelo
- Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | | | | | | | | | - Warren A Skoza
- Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | | | | | | | | | | | | | - David Sancho
- Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Valentin Fuster
- Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain; Mount Sinai Fuster Heart Hospital, New York, New York, USA
| | - Carlos Galán-Arriola
- Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain; Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares, Madrid, Spain.
| | - Borja Ibáñez
- Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain; Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares, Madrid, Spain; Cardiology Department, IIS-Fundación Jiménez Díaz Hospital, Madrid, Spain.
| |
Collapse
|
27
|
Fabiani I, Chianca M, Cipolla CM, Cardinale DM. Anthracycline-induced cardiomyopathy: risk prediction, prevention and treatment. Nat Rev Cardiol 2025:10.1038/s41569-025-01126-1. [PMID: 39875555 DOI: 10.1038/s41569-025-01126-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/17/2025] [Indexed: 01/30/2025]
Abstract
Anthracyclines are the cornerstone of treatment for many malignancies. However, anthracycline cardiotoxicity is a considerable concern given that it can compromise the clinical effectiveness of the treatment and patient survival despite early discontinuation of therapy or dose reduction. Patients with cancer receiving anthracycline treatment can have a reduction in their quality of life and likelihood of survival due to cardiotoxicity, irrespective of their oncological prognosis. Increasing knowledge about anthracycline cardiotoxicity has enabled the identification of patients who are candidates for anthracycline regimens and those who might develop anthracycline-induced cardiomyopathy. Anthracycline cardiotoxicity is a unique and evolving phenomenon that begins with myocardial cell damage, progresses to reduced left ventricular ejection fraction, and culminates in symptomatic heart failure if it is not promptly detected and treated. Early risk stratification can be guided by imaging or biomarkers. In this Review, we present a comprehensive and clinically useful approach to cardiomyopathy related to anthracycline therapy, encompassing its epidemiology, definition, mechanisms, novel classifications, risk factors and patient risk stratification, diagnostic approaches (including imaging and biomarkers), treatment guidelines algorithms, and the role of new cardioprotective drugs that are used for the treatment of heart failure.
Collapse
Affiliation(s)
- Iacopo Fabiani
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy.
| | - Michela Chianca
- Division of Cardiology, Cardiothoracic and Vascular Department, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Carlo Maria Cipolla
- Cardioncology Unit, Cardioncology and Second Opinion Division, European Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy
| | - Daniela Maria Cardinale
- Cardioncology Unit, Cardioncology and Second Opinion Division, European Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy
| |
Collapse
|
28
|
Gallucci G, Larocca M, Navazio A, Turazza FM, Inno A, Canale ML, Oliva S, Besutti G, Tedeschi A, Aschieri D, Russo A, Gori S, Silvestris N, Pinto C, Tarantini L. Atherosclerosis and the Bidirectional Relationship Between Cancer and Cardiovascular Disease: From Bench to Bedside, Part 2 Management. Int J Mol Sci 2025; 26:334. [PMID: 39796190 PMCID: PMC11719480 DOI: 10.3390/ijms26010334] [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: 11/16/2024] [Revised: 12/25/2024] [Accepted: 12/29/2024] [Indexed: 01/13/2025] Open
Abstract
The first part of this review highlighted the evolving landscape of atherosclerosis, noting emerging cardiometabolic risk factors, the growing impact of exposomes, and social determinants of health. The prominent role of atherosclerosis in the bidirectional relationship between cardiovascular disease and cancer was also discussed. In this second part, we examine the complex interplay between multimorbid cardio-oncologic patients, cardiometabolic risk factors, and the harmful environments that lend a "syndemic" nature to these chronic diseases. We summarize management strategies targeting disordered cardiometabolic factors to mitigate cardiovascular disease and explore molecular mechanisms enabling more tailored therapies. Importantly, we emphasize the early interception of atherosclerosis through multifactorial interventions that detect subclinical signs (via biomarkers and imaging) to treat modifiable risk factors and prevent clinical events. A concerted preventive effort-referred to by some as a "preventome"-is essential to reduce the burden of atherosclerosis-driven chronic diseases, shifting from mere chronic disease management to the proactive promotion of "chronic health".
Collapse
Affiliation(s)
| | - Mario Larocca
- Provincial Medical Oncology, Department of Oncology and Advanced Technologies, AUSL—IRCCS in Tecnologie Avanzate e Modelli Assistenziali in Oncologia, 42100 Reggio Emilia, Italy; (M.L.); (C.P.)
| | - Alessandro Navazio
- Cardiologia Ospedaliera, Department of Specialized Medicine, AUSL—IRCCS in Tecnologie Avanzate e Modelli Assistenziali in Oncologia, 42100 Reggio Emilia, Italy;
| | | | - Alessandro Inno
- Oncologia Medica, IRCCS Ospedale Sacro Cuore Don Calabria, 37024 Negrar di Valpolicella, Italy; (A.I.)
| | - Maria Laura Canale
- Division of Cardiology, Azienda USL Toscana Nord-Ovest, Versilia Hospital, 55041 Lido di Camaiore, Italy;
| | - Stefano Oliva
- UOSD Cardiologia di Interesse Oncologico IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy;
| | - Giulia Besutti
- Radiology Unit, Department of Imaging and Laboratory Medicine, AUSL—IRCCS di Reggio Emilia, 42100 Reggio Emilia, Italy;
- Department of Surgical and Medical Sciences of Children and Adults, University of Modena and Reggio Emilia, 41100 Modena, Italy
| | - Andrea Tedeschi
- Cardiology Unit of Emergency Department, Guglielmo da Saliceto Hospital, 29100 Piacenza, Italy; (A.T.); (D.A.)
| | - Daniela Aschieri
- Cardiology Unit of Emergency Department, Guglielmo da Saliceto Hospital, 29100 Piacenza, Italy; (A.T.); (D.A.)
| | - Antonio Russo
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, 90127 Palermo, Italy;
| | - Stefania Gori
- Oncologia Medica, IRCCS Ospedale Sacro Cuore Don Calabria, 37024 Negrar di Valpolicella, Italy; (A.I.)
| | - Nicola Silvestris
- Medical Oncology Department, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy;
| | - Carmine Pinto
- Provincial Medical Oncology, Department of Oncology and Advanced Technologies, AUSL—IRCCS in Tecnologie Avanzate e Modelli Assistenziali in Oncologia, 42100 Reggio Emilia, Italy; (M.L.); (C.P.)
| | - Luigi Tarantini
- Cardiologia Ospedaliera, Department of Specialized Medicine, AUSL—IRCCS in Tecnologie Avanzate e Modelli Assistenziali in Oncologia, 42100 Reggio Emilia, Italy;
| |
Collapse
|
29
|
Di Lisi D, Madaudo C, Macaione F, Galassi AR, Novo G. Cancer survivors and cardiovascular diseases: from preventive strategies to treatment. J Cardiovasc Med (Hagerstown) 2025; 26:8-17. [PMID: 39514326 DOI: 10.2459/jcm.0000000000001681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024]
Abstract
During the last decades, progress in the treatment of oncological diseases has led to an increase in the survival of cancer patients: cancer survivors (CS). Thus, the incidence of CS has increased enormously, in both adult CS and childhood and adolescent CS. Unfortunately, CS treated with anthracyclines, chest radiotherapy (RT) and other potentially cardiotoxic drugs have a higher risk of cardiovascular (CV) toxicity: heart failure with reduced ejection fraction (HFrEF), valve diseases, coronary artery diseases, vascular diseases and pericardial diseases. In fact, chest irradiation can cause coronary artery diseases that can be latent until at least 10 years after exposure; also, valvular heart diseases can appear after >20 years following irradiation; heart failure may appear later, several years after anticancer drugs or RT. Therefore, it is very important to stratify the CV risk of cancer patients at the end of cardiotoxic drugs, to plan the most appropriate long-term surveillance program, in accordance with 2022 ESC Guidelines on Cardio-Oncology, to prevent late cardiovascular complications. Monitoring of cancer patients must not stop during anticancer treatment but it must continue afterwards, depending on the patient's CV risk. CV toxicity risk should be reassessed 5 years after therapy to organize long-term follow-up. Considering late cardiotoxicity in CS, our review aims to evaluate the incidence of cardiovascular diseases in CS, their mechanisms, surveillance protocols, preventive strategies, diagnosis and treatment.
Collapse
Affiliation(s)
- Daniela Di Lisi
- Division of Cardiology, University Hospital 'Paolo Giaccone'
| | - Cristina Madaudo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE) 'G. D'Alessandro', University of Palermo, Palermo, Italy
| | | | - Alfredo Ruggero Galassi
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE) 'G. D'Alessandro', University of Palermo, Palermo, Italy
| | - Giuseppina Novo
- Division of Cardiology, University Hospital 'Paolo Giaccone'
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE) 'G. D'Alessandro', University of Palermo, Palermo, Italy
| |
Collapse
|
30
|
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
|
31
|
Theofilis P, Vlachakis PK, Oikonomou E, Drakopoulou M, Karakasis P, Apostolos A, Pamporis K, Tsioufis K, Tousoulis D. Cancer Therapy-Related Cardiac Dysfunction: A Review of Current Trends in Epidemiology, Diagnosis, and Treatment. Biomedicines 2024; 12:2914. [PMID: 39767820 PMCID: PMC11673750 DOI: 10.3390/biomedicines12122914] [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: 11/23/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Cancer therapy-related cardiac dysfunction (CTRCD) has emerged as a significant concern with the rise of effective cancer treatments like anthracyclines and targeted therapies such as trastuzumab. While these therapies have improved cancer survival rates, their unintended cardiovascular side effects can lead to heart failure, cardiomyopathy, and arrhythmias. The pathophysiology of CTRCD involves oxidative stress, mitochondrial dysfunction, and calcium dysregulation, resulting in irreversible damage to cardiomyocytes. Inflammatory cytokines, disrupted growth factor signaling, and coronary atherosclerosis further contribute to this dysfunction. Advances in cardio-oncology have led to the early detection of CTRCD using cardiac biomarkers like troponins and imaging techniques such as echocardiography and cardiac magnetic resonance (CMR). These tools help identify asymptomatic patients at risk of cardiac events before the onset of clinical symptoms. Preventive strategies, including the use of cardioprotective agents like beta-blockers, angiotensin-converting enzyme inhibitors, mineralocorticoid receptor antagonists, and sodium-glucose cotransporter-2 inhibitors have shown promise in reducing the incidence of CTRCD. This review summarizes the mechanisms, detection methods, and emerging treatments for CTRCD, emphasizing the importance of interdisciplinary collaboration between oncologists and cardiologists to optimize care and improve both cancer and cardiovascular outcomes.
Collapse
Affiliation(s)
- Panagiotis Theofilis
- 1st Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.T.); (P.K.V.); (M.D.); (A.A.); (K.P.); (K.T.)
| | - Panayotis K. Vlachakis
- 1st Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.T.); (P.K.V.); (M.D.); (A.A.); (K.P.); (K.T.)
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, Thoracic Diseases General Hospital Sotiria, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Maria Drakopoulou
- 1st Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.T.); (P.K.V.); (M.D.); (A.A.); (K.P.); (K.T.)
| | - Paschalis Karakasis
- 2nd Department of Cardiology, Hippokration General Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece;
| | - Anastasios Apostolos
- 1st Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.T.); (P.K.V.); (M.D.); (A.A.); (K.P.); (K.T.)
| | - Konstantinos Pamporis
- 1st Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.T.); (P.K.V.); (M.D.); (A.A.); (K.P.); (K.T.)
| | - Konstantinos Tsioufis
- 1st Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.T.); (P.K.V.); (M.D.); (A.A.); (K.P.); (K.T.)
| | - Dimitris Tousoulis
- 1st Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.T.); (P.K.V.); (M.D.); (A.A.); (K.P.); (K.T.)
| |
Collapse
|
32
|
Belamkar AV, Mounayar M, Clasen SC. SGLT2 Inhibitor for Cardiac Protection in a Patient With Osimertinib-Responsive Advanced EGFR-Positive Lung Cancer. JACC Case Rep 2024; 29:102829. [PMID: 39691329 PMCID: PMC11646913 DOI: 10.1016/j.jaccas.2024.102829] [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: 09/07/2024] [Revised: 10/14/2024] [Accepted: 10/18/2024] [Indexed: 12/19/2024]
Abstract
An 85-year-old man with recently diagnosed metastatic EGFR+ lung adenocarcinoma treated with osimertinib presented after 1 month of therapy in decompensated congestive heart failure along with atrial fibrillation, prolonged QTc and acute kidney injury. Osimertinib was held. His hemodynamic status was optimized, and he was started on cardioprotective medications (losartan and metoprolol succinate), and LVEF recovered. However, after reintroducing osimertinib, LVEF reduced, indicating possible osimertinib-induced cardiomyopathy. An SGLT2 inhibitor was added for cardioprotection before another rechallenge of osimertinib. SGLT2 inhibitors are a powerful tool for heart failure and may have a potential secondary benefit in ameliorating cardiotoxic processes. Although their use in osimertinib-induced cardiomyopathy has not been well-established, current heart failure guidelines and emerging research support its use in this setting. This case and the accompanying literature review highlight the novel use of SGLT2 inhibitors coupled with regular clinical and imaging monitoring, as a compelling intervention for osimertinib-induced cardiomyopathy.
Collapse
Affiliation(s)
| | - Marwan Mounayar
- Indiana University School of Medicine, Indianapolis, Indiana, USA
- Indiana University Health Ball Memorial Hospital Cancer Center, Muncie, Indiana, USA
| | - Suparna C. Clasen
- Indiana University School of Medicine, Indianapolis, Indiana, USA
- Division of Cardiovascular Medicine, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
33
|
Lemos Ferreira N, Bamidele Adelowo A, Khan Z. A Systematic Review and Meta-Analysis of Sodium-Glucose Cotransporter 2 (SGLT-2) Inhibitors and Their Impact on the Management of Heart Failure. Cureus 2024; 16:e75802. [PMID: 39816302 PMCID: PMC11734706 DOI: 10.7759/cureus.75802] [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: 12/16/2024] [Indexed: 01/18/2025] Open
Abstract
Heart failure (HF) is a life-threatening condition with severe incapacitating consequences. Many body organs and systems may be affected, which may also hinder the quality of life and finances at the individual and societal levels. Sodium-glucose cotransporter-2 inhibitors (SGLT2i) have also emerged as potentially useful drugs in the HF domain and other medical fields, in addition to their glucose-lowering effect. This systematic review followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, and the authors searched Google Scholar, PubMed, and Scopus websites for SGLT2i and SGLT2i-related terms and their impact on HF events, major adverse cardiovascular events (MACEs), renal composite outcomes, and improvement in the Kansas City Cardiomyopathy Questionnaire (KCCQ) scores, involving human adult populations. Two reviewers conducted the literature search, and disagreements were resolved through mutual consensus and input from a third reviewer. A literature search was conducted from 1st February to 20th February 2024. We included studies published after 2018 to focus only on the latest advancements. Randomized controlled trials, observational studies, or systematic reviews of these studies were included in our study. Of the 44 initial articles identified, only 14 met the inclusion and exclusion criteria. The outcomes revealed the superiority of SGLT2i therapeutics over placebo in all four domains mentioned above. A total of 234,509 patients from 11 papers with moderate heterogeneity (P = 0.07; I2 = 42%) evaluating the effect of SGLT2i in comparison to placebo on HF events were considered; of these, 128,477 patients received the intervention drug, and 106,032 individuals were assigned to the control group. The absolute numbers of HF events were 6845 and 8877, respectively. The study showed an overall benefit of SGLT2i in patients with heart failure due to their ability to major adverse cardiovascular events (MACE) in comparison to placebo (OR: 0.92; 95% CI: 0.89-0.96; P < 0.00001). This systematic review confirmed previous findings related to the use of SGLT2i as adjunctive therapy for HF and amelioration of KCCQ scores and as a protective agent against MACE and renal impairment progression.
Collapse
Affiliation(s)
| | | | - Zahid Khan
- Acute Medicine, Mid and South Essex NHS Foundation Trust, Southend-on-Sea, GBR
- Cardiology, Bart's Heart Centre, London, GBR
- Cardiology and General Medicine, Barking, Havering and Redbridge University Hospitals NHS Trust, London, GBR
- Cardiology, Royal Free Hospital, London, GBR
| |
Collapse
|
34
|
Dabour MS, Blaes AH, Ramu B, Zordoky BN. SGLT2 Inhibitors in Mitigating Cancer Therapy-Related Cardiac Dysfunction: Expanding the Evidence. JACC CardioOncol 2024; 6:876-878. [PMID: 39801638 PMCID: PMC11711823 DOI: 10.1016/j.jaccao.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025] Open
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
| | - Anne H. Blaes
- Division of Hematology/Oncology/Transplantation, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Bhavadharini Ramu
- Division of Cardiology, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Beshay N. Zordoky
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
35
|
Karamat M, Hussain B, Ahmed MM, Hamza M, Mir J, Alamri A, Shafiq A, Sattar Y, Khan MZ, Thyagaturu H, Gonuguntla K, Patel BD. Deciphering the cardioprotective effects of statins in anthracycline-related cardiac dysfunction: A systematic review and meta-analysis. Am J Prev Cardiol 2024; 20:100874. [PMID: 39524983 PMCID: PMC11543556 DOI: 10.1016/j.ajpc.2024.100874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 09/07/2024] [Accepted: 09/22/2024] [Indexed: 11/16/2024] Open
Abstract
Background Cancer induced chronic inflammation and cancer drugs effects on the vascular system can lead to rapidly progressing atherosclerotic burden. Statins drugs are known to reduce atherosclerotic plaque burden and inflammation. We studied outcomes of statins for anthracycline-related cardiac dysfunction (ARCD). Methods We conducted an online systematic search on PubMed and Embase to identify studies assessing statins' role in alleviating ARCD. We selected 9 studies that had patients with ARCD and use of statins. We primarily focused on outcomes including incidence of heart failure (HF), mean changes in left ventricular ejection fraction (LVEF), end-diastolic volume (LVEDV), and end-systolic volume (LVESV) from baseline. Odds ratios (OR) were calculated using a random effect model in R-statistics. Results Among 9 studies with a total of 2784 patients we noticed a significant reduction in the incidence of HF among patients administered statins, with an OR of 0.52 (95 % CI 0.37-0.74, p < 0.0003), indicating a substantial protective effect. However, the mean changes in EF, LVEDV, and LVESV from baseline, represented by Hedges's g of 1.09 (95 % CI -0.40 to 2.57, p = 0.15), 0.91 (95 % CI -1.69 to 3.51, p = 0.47), and 1.32 (95 % CI -2.30 to 4.94, p = 0.49) respectively, were not statistically significant. (Figure 1). Conclusion Our meta-analysis confirms statins' effectiveness in reducing risk of ARCD. However, their influence on EF, LVEDV, and LVESV remains uncertain, warranting further study.
Collapse
Affiliation(s)
- Mubashar Karamat
- Department of Internal Medicine, Ascension Saint Francis Hospital, Evanston, IL, USA
| | - Bilal Hussain
- Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, OH, USA
| | | | - Mohammad Hamza
- Department of Internal Medicine, Albany Medical Center, Albany, NY
| | - Junaid Mir
- Department of Internal Medicine, University of Missouri, Columbia, MO, USA
| | - Ayedh Alamri
- Department of Cardiovascular Department, Tufts Medical Center, Boston, USA
| | - Aimen Shafiq
- Department of Internal Medicine, Dow University of Health Sciences, Pakistan
| | - Yasar Sattar
- Department of Cardiology, West Virginia University, Morgantown, WV, USA
| | - Muhammad Zia Khan
- Department of Cardiology, West Virginia University, Morgantown, WV, USA
| | | | | | - Birjesh D Patel
- Department of Cardiology, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
36
|
Bhatti AW, Patel R, Dani SS, Khadke S, Makwana B, Lessey C, Shah J, Al-Husami Z, Yang EH, Thavendiranathan P, Neilan TG, Sadler D, Cheng RK, Dent SF, Liu J, Lopez-Fernandez T, Herrmann J, Scherrer-Crosbie M, Lenihan DJ, Hayek SS, Ky B, Deswal A, Barac A, Nohria A, Ganatra S. SGLT2i and Primary Prevention of Cancer Therapy-Related Cardiac Dysfunction in Patients With Diabetes. JACC CardioOncol 2024; 6:863-875. [PMID: 39801650 PMCID: PMC11711834 DOI: 10.1016/j.jaccao.2024.08.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 08/03/2024] [Indexed: 01/16/2025] Open
Abstract
Background Specific cancer treatments can lead to cancer therapy-related cardiac dysfunction (CTRCD). Sodium glucose cotransporter-2 inhibitors (SGLT2is) can potentially prevent these cardiotoxic effects. Objectives This study sought to determine whether SGLT2i use is associated with a lower incidence of CTRCD in patients with type 2 diabetes mellitus (T2DM) and cancer, exposed to potentially cardiotoxic antineoplastic agents, and without a prior documented history of cardiomyopathy or heart failure. Methods We conducted a retrospective analysis of patients aged ≥18 years within the TriNetX database with T2DM, cancer, exposure to cardiotoxic therapies, and no prior documented history of cardiomyopathy or heart failure. Patients were categorized by SGLT2i use. After propensity score matching, outcomes were compared over 12 months using Cox proportional HRs. Subgroup analyses focusing on different cancer therapy classes were performed. Results The study included 8,675 propensity-matched patients in each cohort (mean age = ∼65 years, 42% females, 71% White, ∼19% gastrointestinal malignancy, and ∼25% anthracyclines). Patients prescribed SGLT2is had a lower risk of developing CTRCD (HR: 0.76: 95% CI: 0.69-0.84). SGLT2is also reduced heart failure exacerbations (HR: 0.81; 95% CI: 0.72-0.90), all-cause mortality (HR: 0.67; 95% CI: 0.61-0.74), and all-cause hospitalizations/emergency department visits (HR: 0.93; 95% CI: 0.89-0.97). Subgroup analyses also demonstrated reduced CTRCD risk across various classes of cancer therapies in patients prescribed SGLT2is. Conclusions SGLT2i administration was associated with a significantly decreased risk of developing CTRCD in patients with T2DM and cancer.
Collapse
Affiliation(s)
- Ammar W. Bhatti
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Beth Israel Lahey Health, Burlington, Massachusetts, USA
| | - Rushin Patel
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Beth Israel Lahey Health, Burlington, 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
| | - Sumanth Khadke
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Beth Israel Lahey Health, Burlington, Massachusetts, USA
| | - Bhargav Makwana
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Beth Israel Lahey Health, Burlington, Massachusetts, USA
| | - Candace Lessey
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Beth Israel Lahey Health, Burlington, Massachusetts, USA
| | - Jui Shah
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Beth Israel Lahey Health, Burlington, Massachusetts, USA
| | - Zaid Al-Husami
- 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
- University of California-Los Angeles, Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California-Los Angeles, Los Angeles, California, USA
| | - Paaladinesh Thavendiranathan
- Department of Medicine, Division of Cardiology, Ted Rogers Program in Cardiotoxicity Prevention, Peter Munk Cardiac Center, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Tomas G. Neilan
- Cardio-Oncology Program, Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Diego Sadler
- Cardio-Oncology Section, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Weston, Florida, USA
| | - Richard K. Cheng
- Division of Cardiology, University of Washington, Seattle, Washington, USA
| | - Susan F. Dent
- Duke Cancer Institute, Department of Medicine, Duke University, Durham, North Carolina
| | - Jennifer Liu
- Cardio-Oncology Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Teresa Lopez-Fernandez
- Division of Cardiology, Cardio-Oncology Unit, La Paz University Hospital, Hospital La Paz Institute for Health Research, Madrid, Spain
- Division of Cardiology, Quironsalud Madrid University Hospital, Madrid, Spain
| | - Joerg Herrmann
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Daniel J. Lenihan
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Salim S. Hayek
- Division of Cardiology, Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, USA
| | - Bonnie Ky
- Thalheimer Center for Cardio-Oncology, Abramson Cancer Center and Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Anita Deswal
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ana Barac
- Inova Schar Heart and Vascular Institute, Inova Schar Cancer Institute, Fairfax, Virginia, USA
| | - Anju Nohria
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, 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
|
37
|
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
|
38
|
Li W, Shen X, Zhang M, Tan W, Jiang X, Wen H, Shen Y. Meta-analysis of the efficacy and impact on cardiac function of sodium-glucose cotransporter 2 inhibitor Empagliflozin in heart failure patients. Medicine (Baltimore) 2024; 103:e40409. [PMID: 39533603 PMCID: PMC11557010 DOI: 10.1097/md.0000000000040409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Currently, there is no comprehensive systematic review available to comprehensively assess the efficacy and safety of Empagliflozin and other sodium-glucose cotransporter 2 inhibitors in the treatment of heart failure (HF). This study employed a meta-analysis approach to systematically evaluate the therapeutic effects of Empagliflozin in HF patients and its impact on cardiac function. METHOD The keywords including "heart failure," "HF," "cardiac failure," "cardiac disease," "Empagliflozin," and "sodium-glucose cotransporter 2 inhibitors" were utilized to search for relevant clinical studies on Empagliflozin in the treatment of HF in various databases, such as China National Knowledge Infrastructure, Wanfang, VIP Chinese Medical Journal Database, PubMed, MEDLINE, Embase, Cochrane Library, Springer, and Science Direct. The studies included patients with HF who received drug treatment. Data on baseline characteristics and posttreatment outcomes, including HF hospitalization (HHF), cardiovascular mortality, all-cause mortality, estimated glomerular filtration rate changes, Kansas City Cardiomyopathy Questionnaire quality of life (QoL) scores, N-terminal pro-B-type natriuretic peptide, left ventricular ejection fraction, hematocrit, and other relevant indicators were collected. Meta-analysis was conducted using RevMan5.3 to analyze the extracted data. RESULTS A total of 15 studies were included in the final analysis, comprising 36,917 patients with HF. Among them, 18,486 patients were in Empagliflozin group, and 18,431 patients were in control (Ctrl) group. The results of the meta-analysis demonstrated that, relative to Ctrl group, Empagliflozin group showed a substantially lower HHF rate, a substantial improvement in estimated glomerular filtration rate changes, a reduced cardiovascular mortality rate, a higher Kansas City Cardiomyopathy Questionnaire QoL score, increased hematocrit values, reduced N-terminal pro-B-type natriuretic peptide changes, and enhanced left ventricular ejection fraction changes. These findings suggest that remarkable improvements in various outcomes compared to the Ctrl group. CONCLUSION The sodium-glucose cotransporter 2 inhibitor Empagliflozin markedly reduces the HHF rate and cardiovascular mortality in HF patients. It also improves patients' QoL, enhances renal function, and increases cardiac function while reducing both, the preload and afterload.
Collapse
Affiliation(s)
- Weidong Li
- Department of Emergency, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xuanyang Shen
- Department of Emergency, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Meiqi Zhang
- Department of Emergency, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Wentao Tan
- Department of Emergency, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xiaolu Jiang
- Department of Emergency, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Hongfu Wen
- Department of Emergency, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yuan Shen
- Department of Emergency, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| |
Collapse
|
39
|
Schuuring MJ, Treskes RW, Castiello T, Jensen MT, Casado-Arroyo R, Neubeck L, Lyon AR, Keser N, Rucinski M, Marketou M, Lambrinou E, Volterrani M, Hill L. Digital solutions to optimize guideline-directed medical therapy prescription rates in patients with heart failure: a clinical consensus statement from the ESC Working Group on e-Cardiology, the Heart Failure Association of the European Society of Cardiology, the Association of Cardiovascular Nursing & Allied Professions of the European Society of Cardiology, the ESC Digital Health Committee, the ESC Council of Cardio-Oncology, and the ESC Patient Forum. EUROPEAN HEART JOURNAL. DIGITAL HEALTH 2024; 5:670-682. [PMID: 39563907 PMCID: PMC11570396 DOI: 10.1093/ehjdh/ztae064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/14/2024] [Accepted: 08/13/2024] [Indexed: 11/21/2024]
Abstract
The 2021 European Society of Cardiology guideline on diagnosis and treatment of acute and chronic heart failure (HF) and the 2023 Focused Update include recommendations on the pharmacotherapy for patients with New York Heart Association (NYHA) class II-IV HF with reduced ejection fraction. However, multinational data from the EVOLUTION HF study found substantial prescribing inertia of guideline-directed medical therapy (GDMT) in clinical practice. The cause was multifactorial and included limitations in organizational resources. Digital solutions like digital consultation, digital remote monitoring, digital interrogation of cardiac implantable electronic devices, clinical decision support systems, and multifaceted interventions are increasingly available worldwide. The objectives of this Clinical Consensus Statement are to provide (i) examples of digital solutions that can aid the optimization of prescription of GDMT, (ii) evidence-based insights on the optimization of prescription of GDMT using digital solutions, (iii) current evidence gaps and implementation barriers that limit the adoption of digital solutions in clinical practice, and (iv) critically discuss strategies to achieve equality of access, with reference to patient subgroups. Embracing digital solutions through the use of digital consults and digital remote monitoring will future-proof, for example alerts to clinicians, informing them of patients on suboptimal GDMT. Researchers should consider employing multifaceted digital solutions to optimize effectiveness and use study designs that fit the unique sociotechnical aspects of digital solutions. Artificial intelligence solutions can handle larger data sets and relieve medical professionals' workloads, but as the data on the use of artificial intelligence in HF are limited, further investigation is warranted.
Collapse
Affiliation(s)
- Mark Johan Schuuring
- Department of Biomedical Signals and Systems, University of Twente, Drienerlolaan 5, 7522 NB Enschede, The Netherlands
- Department of Cardiology, Medical Spectrum Twente, 7512 KZ Enschede, The Netherlands
| | | | - Teresa Castiello
- Department of Cardiovascular Imaging, King's College London, Croydon Health Service London, London, UK
| | | | - Ruben Casado-Arroyo
- Department of Cardiology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Lis Neubeck
- Centre for Cardiovascular Health, Edinburgh Napier University, Edinburgh, UK
| | - Alexander R Lyon
- Cardio-Oncology Service, Royal Brompton Hospital, Guys and St. Thomas NHS Foundation Trust, London, UK
| | - Nurgul Keser
- Faculty of Medicine, Department of Cardiology-Istanbul, Istanbul Health Sciences University, Istanbul, Turkey
| | - Marcin Rucinski
- Poland, ESC Patient Forum, European Society of Cardiology, Sophia Antipolis Cedex, France
| | - Maria Marketou
- Cardiology Department, Heraklion University Hospital, Stavrakia, Heraklion, Greece
| | | | | | - Loreena Hill
- School of Nursing and Midwifery, Queen's University Belfast, Belfast, UK
| |
Collapse
|
40
|
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: 3] [Impact Index Per Article: 3.0] [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
|
41
|
Raisi-Estabragh Z, Murphy AC, Ramalingam S, Scherrer-Crosbie M, Lopez-Fernandez T, Reynolds KL, Aznar M, Lin AE, Libby P, Cordoba R, Bredsen-Masley C, Wechalekar A, Apperley J, Cheng RK, Manisty CH. Cardiovascular Considerations Before Cancer Therapy: Gaps in Evidence and JACC: CardioOncology Expert Panel Recommendations. JACC CardioOncol 2024; 6:631-654. [PMID: 39479317 PMCID: PMC11520216 DOI: 10.1016/j.jaccao.2024.07.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 11/02/2024] Open
Abstract
Baseline cardiovascular assessment before the initiation of potentially cardiotoxic cancer therapies is a key component of cardio-oncology, aiming to reduce cardiovascular complications and morbidity in patients and survivors. Recent clinical practice guidelines provide both general and cancer therapy-specific recommendations for baseline cardiovascular toxicity risk assessment and management, including the use of dedicated risk scores, cardiovascular imaging, and biomarker testing. However, the value of such interventions in altering disease trajectories has not been established, with many recommendations based on expert opinion or Level of Evidence: C, studies with a potential for high risk of bias. Advances in understanding underlying mechanisms of cardiotoxicity and the increased availability of genetic and immunologic profiling present new opportunities for personalized risk assessment. This paper evaluates the existing evidence on cardiovascular care of cancer patients before cardiotoxic cancer therapy and highlights gaps in evidence and priorities for future research.
Collapse
Affiliation(s)
- Zahra Raisi-Estabragh
- William Harvey Research Institute, National Institute for Health and Care Research Barts Biomedical Research Centre, Queen Mary University of London, London, United Kingdom
- Barts Heart Centre, St Bartholomew’s Hospital, Barts Health NHS Trust, West Smithfield, London, United Kindgom
| | | | - Sivatharshini Ramalingam
- Cardio-Oncology Service, Royal Brompton Hospital, Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Marielle Scherrer-Crosbie
- Division of Cardiology, The Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Teresa Lopez-Fernandez
- Hospital Universitario La Paz, Instituto de Investigación La Paz-IdiPaz, Madrid, Spain
- Hospital Universitario Quiron Pozuelo, Madrid, Spain
| | - Kerry L. Reynolds
- Division of Oncology and Hematology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Marianne Aznar
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Amy E. Lin
- Department of Medicine, Division of Cardiology, Section of Cardio-Oncology and Immunology, University of California-San Francisco, San Francisco, California, USA
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Raul Cordoba
- Fundacion Jimenez Diaz University Hospital, Health Research Institute Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Madrid, Spain
| | - Christine Bredsen-Masley
- Division of Hematology/Oncology, Department of Medicine, St. Michael’s Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Ashu Wechalekar
- Department of Haematology, University College London Hospitals NHS Trust, London, United Kingdom
| | - Jane Apperley
- Department of Clinical Haematology, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom
- Centre for Haematology, Imperial College London, London, United Kingdom
| | - Richard K. Cheng
- Division of Cardiology, University of Washington Medical Center, Seattle, Washington, USA
| | - Charlotte H. Manisty
- Barts Heart Centre, St Bartholomew’s Hospital, Barts Health NHS Trust, West Smithfield, London, United Kindgom
- Institute of Cardiovascular Science, University College London, London, United Kingdom
| |
Collapse
|
42
|
Bloom M, Alvarez-Cardona JA, Ganatra S, Barac A, Pusic I, Lenihan D, Dent S. How to utilize current guidelines to manage patients with cancer at high risk for heart failure. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:63. [PMID: 39342407 PMCID: PMC11438295 DOI: 10.1186/s40959-024-00259-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 08/19/2024] [Indexed: 10/01/2024]
Abstract
Heart failure (HF) in patients with cancer is associated with high morbidity and mortality. The success of cancer therapy has resulted in an exponential rise in the population of cancer survivors, however cardiovascular disease (CVD) is now a major life limiting condition more than 5 years after cancer diagnosis [Sturgeon, Deng, Bluethmann, et al 40(48):3889-3897, 2019]. Prevention and early detection of CVD, including cardiomyopathy (CM) and HF is of paramount importance. The European Society of Cardiology (ESC) published guidelines on Cardio-Oncology (CO) [Lyon, López-Fernández, Couch, et al 43(41):4229-4361, 2022] detailing cardiovascular (CV) risk stratification, prevention, monitoring, diagnosis, and treatment throughout the course and following completion of cancer therapy. Here we utilize a case to summarize aspects of the ESC guideline relevant to HF clinicians, with a focus on risk stratification, early detection, prevention of CM and HF, and the role for guideline directed medical therapy in patients with cancer.
Collapse
Affiliation(s)
- Michelle Bloom
- Leon H. Charney Division of Cardiology, New York University School of Medicine, NYU Langone Health, New York, NY, USA
| | - Jose A Alvarez-Cardona
- Leon H. Charney Division of Cardiology, New York University School of Medicine, NYU Langone Health, New York, NY, USA
| | - Sarju Ganatra
- Division of Cardiovascular Medicine, Lahey Hospital and Medical Center, Department of Medicine, Beth Israel Lahey Health, Burlington, MA, USA
| | - Ana Barac
- Inova Schar Heart and Vascular and Inova Inova Schar Cancer, Falls Church, VA, USA
| | - Iskra Pusic
- Division of Oncology, Department of Medicine, Washington University, St. Louis, MO, USA
| | - Daniel Lenihan
- International Cardio-Oncology Society, FL and Cape Cardiology Group, Cape Girardeau, Tampa, MO, USA
| | - Susan Dent
- Duke Cancer Institute, Department of Medicine, Duke University, DUMC 3446, Durham, NC, 27710, USA.
| |
Collapse
|
43
|
Daniele AJ, Gregorietti V, Costa D, López-Fernández T. Use of EMPAgliflozin in the prevention of CARDiotoxicity: the EMPACARD - PILOT trial. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:58. [PMID: 39237985 PMCID: PMC11375926 DOI: 10.1186/s40959-024-00260-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 08/22/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND Anthracycline-based chemotherapy represents a cornerstone treatment for a number of common cancers, including breast cancer, lymphoma, and sarcoma. However, anthracycline-induced cardiotoxicity remains a significant concern, often presenting as a decline in cardiac function which can ultimately lead to heart failure (HF) or asymptomatic left ventricular dysfunction, in up to 10-15% of patients.Sodium-glucose transport protein 2 inhibitor (SGLT2i) therapies have been demonstrated to reduce the incidence of HF in high-risk non-cancer patients. Preliminary retrospective data suggest their role in mitigating the incidence of HF during or after anthracycline treatment METHODS: The EMPACARD-PILOT trial was a prospective case‒control study involving breast cancer patients scheduled to undergo anthracycline-based chemotherapy in a 4-cycle protocol of 60 mg/m2 doxorubicin. We used the HFA/ICOS risk score to identify patients at high or very high risk of cardiotoxicity. Patients with diabetes mellitus or stable heart failure with preserved ejection fraction (HFpEF) were prescribed empagliflozin (10 mg per day), starting seven days before the administration of anthracyclines and continuing for a period of six months. Those not meeting these criteria served as controls. The primary endpoint was cancer therapy-related cardiac dysfunction (CTRCD) incidence. CTRCD was defined as either a decrease in left ventricular ejection fraction (LVEF) of at least 10% to a final value below 50% or a reduction in global longitudinal strain (GLS) of at least 15% from baseline at any point during the study. The secondary endpoints included mortality and hospitalization due to cardiovascular causes or clinical heart failure. Exploratory endpoints included increases in serum troponin and NT-proBNP levels and a decrease in the glomerular filtration rate (GFR). The safety endpoints tracked includedketoacidosis, hypoglycemia, sepsis, neutropenic fever, and urinary tract infections. RESULTS During the enrollment period, 785 breast cancer patients were analysed. Of these, 107 met the inclusion criteria, and 76 subsequently provided informed consent. The study was conducted with comparable adherence rates of 81.5% in both the empagliflozin group (n = 38) and the control group (n = 38). The follow-up data from 62 patients revealed a significant reduction in the primary outcome within 6 months for the empagliflozin group compared with the control group (6.5% vs. 35.5%, p = 0.005), with a relative risk of 0.18 (95% CI: 0.04-0.75). Compared with the control treatment, treatment with empagliflozin also significantly preserved the ejection fraction at 6 months follow-up (56.8% ± 5.8% vs. 53.7% ± 6.7, p = 0.029). However, there were no significant differences between the groups in terms of NT-proBNP, cTnI, clinical heart failure, GFR, or mortality/hospitalization due to heart failure. CONCLUSION Empagliflozin is associated with reduced incidence of CTRCD in high-risk patients treated with anthracyclines. These data should serve as the foundation for a clinical trial to test whether SGLT2 inhibitors can reduce the incidence of heart failure in this patient group.
Collapse
Affiliation(s)
| | | | - Diego Costa
- Sanatorio Sagrado Corazón, Buenos Aires, Argentina
| | | |
Collapse
|
44
|
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
|
45
|
Farooq Z, Sahar R, Ibekwe WC, Kashyap A, Mehmood K, Ali A. Mitigating Cardiotoxicity Associated with Anticancer Drugs: An Updated Systematic Review. JOURNAL OF CANCER & ALLIED SPECIALTIES 2024; 10:613. [PMID: 39156939 PMCID: PMC11326660 DOI: 10.37029/jcas.v10i2.613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 03/14/2024] [Indexed: 08/20/2024]
Abstract
INTRODUCTION This systematic review investigated strategies to mitigate cardiotoxicity induced by anticancer medications, emphasizing exercise and pharmacological interventions. METHODS We systematically reviewed three randomized controlled trials, one ATOPE trial, and one retrospective cohort study. RESULTS Among 448 patients, exercise interventions, particularly in breast cancer patients, demonstrated significant improvements in left ventricular ejection fraction (LVEF) and cardiotoxicity prevention. Pharmacological interventions, including candesartan and carvedilol, have shown potential in reducing early DOX-induced subclinical cardiotoxicity (DISC). The protective efficacy of candesartan in alleviating DISC was greater than carvedilol and the control group. Combination therapy with lisinopril and bisoprolol effectively preserved the LVEF. A retrospective cohort study demonstrated the cardioprotective potential of sodium-glucose cotransporter-2 inhibitors in reducing cardiovascular events. CONCLUSION This systematic review underscores the promise of exercise and pharmacological interventions for preserving cardiac function in cancer patients receiving chemotherapy. These findings have significant implications for enhancing the quality of care for cancer patients.
Collapse
Affiliation(s)
- Zobia Farooq
- Department of Medicine, King Edward Medical University, Lahore, Pakistan
| | - Rabeaa Sahar
- Department of Medicine, King Edward Medical University, Lahore, Pakistan
| | | | - Abhishek Kashyap
- Department of Medicine, Sir Seewosagur Ramgoolam Medical College, Belle Rive, Mauritius
| | - Khansa Mehmood
- Department of Medicine, King Edward Medical University, Lahore, Pakistan
| | - Ahsan Ali
- Department of Pharmaceutics, Islamia University of Bahawalpur, Bahawalpur, Pakistan
| |
Collapse
|
46
|
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
|
47
|
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: 2] [Impact Index Per Article: 2.0] [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
|
48
|
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
|
49
|
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: 4] [Impact Index Per Article: 4.0] [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
|
50
|
Hwang HJ, Han SA, Sohn IS. Breast Cancer and Therapy-Related Cardiovascular Toxicity. J Breast Cancer 2024; 27:147-162. [PMID: 38769686 PMCID: PMC11221208 DOI: 10.4048/jbc.2024.0085] [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: 04/02/2024] [Revised: 04/10/2024] [Accepted: 04/23/2024] [Indexed: 05/22/2024] Open
Abstract
The global incidence of breast cancer is on the rise, a trend also observed in South Korea. However, thanks to the rapid advancements in anticancer therapies, survival rates are improving. Consequently, post-treatment health and quality of life for breast cancer survivors are emerging as significant concerns, particularly regarding treatment-related cardiotoxicity. In this review, we delve into the cardiovascular complications associated with breast cancer treatment, explore surveillance protocols for early detection and diagnosis of late complications, and discuss protective strategies against cardiotoxicity in breast cancer patients undergoing anticancer therapy, drawing from multiple guidelines.
Collapse
Affiliation(s)
- Hui-Jeong Hwang
- Department of Cardiology, Kyung Hee University College of Medicine, Kyung Hee University Hospital at Gangdong, Seoul, Korea
| | - Sang-Ah Han
- Department of Surgery, Kyung Hee University College of Medicine, Kyung Hee University Hospital at Gangdong, Seoul, Korea
| | - Il Suk Sohn
- Department of Cardiology, Kyung Hee University College of Medicine, Kyung Hee University Hospital at Gangdong, Seoul, Korea.
| |
Collapse
|