1
|
Costa IBSDS, Furtado RHM, Drager LF, de Barros E Silva PGM, Melo MDTD, Araruna P, Bacchiega BC, Cauduro S, Walter E, Fialho GL, Silvestre O, Damiani LP, Barbosa LM, Luz MN, Silva ACA, de Mattos RR, Saretta R, Rehder MHHS, Hajjar LA, Lopes-Fernandez T, Dent S, Gibson CM, Lopes RD, Kalil Filho R. Effects of carvedilol on the prevention of cardiotoxicity induced by anthracyclines: Design and rationale of the CARDIOTOX trial. Am Heart J 2025; 285:1-11. [PMID: 39988204 DOI: 10.1016/j.ahj.2025.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 02/25/2025]
Abstract
BACKGROUND Patients with cancer undergoing chemotherapy with an anthracycline-based regimen are at increased risk of cardiotoxicity, predisposing to heart failure, arrhythmias and death. Whether carvedilol may confer benefit to prevent anthracycline-induced cardiotoxicity remains to be determined. DESIGN CARDIOTOX is a double-blind, placebo controlled randomized clinical trial that plan to enroll 1,018 patients across 25 study sites in Brazil. Patients with active cancer scheduled to undergo an anthracycline-based chemotherapy regimen are eligible. Patients with prior HF or cardiomyopathy are excluded. Patients are randomized in 1:1 ratio to carvedilol (starting dose 6.25mg BID up titrated to 25mg BID or maximum tolerated dose) or placebo, stratified by site and use of renin-angiotensin blockers at baseline. Study drug is administered through the duration of chemotherapy and up to 30 days after the last dose of anthracycline. Patients are scheduled to undergo echocardiographic evaluations at baseline and at 3, 6, and 12 months. The study primary endpoint is the composite of new left ventricle ejection fraction (LVEF) reduction by at least 10% leading to an LVEF <50%, cardiovascular death, myocardial infarction, urgent care visit or hospitalization for heart failure, or clinically significant arrhythmias at 12 months. Echocardiographic images will be analyzed by a central core lab, clinical outcomes will be adjudicated, and safety endpoints include serious adverse events and adverse events of special interest (symptomatic bradycardia, hypotension, syncope and bronchospasm). SUMMARY The CARDIOTOX trial is the largest trial to date analyzing the potential role of beta-blockers as prophylactic therapy to prevent cardiotoxicity induced by anthracyclines. TRIAL REGISTRATION Effects of Carvedilol on Cardiotoxicity in Cancer Patients Submitted to Anthracycline Therapy (CardioTox). CLINICALTRIALS gov ID NCT04939883. https://clinicaltrials.gov/study/NCT04939883.
Collapse
Affiliation(s)
- Isabela Bispo Santos da Silva Costa
- Hospital Sírio-Libanês Research and Education Institute, São Paulo, Brazil; Instituto do Cancer do Estado de São Paulo, Hospital das Clinicas da Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Remo H M Furtado
- Hospital Sírio-Libanês Research and Education Institute, São Paulo, Brazil; Brazilian Clinical Research Institute, São Paulo, Brazil; Instituto do Coração (InCor), Hospital das Clinicas da Faculdade de Medicina, Sao Paulo, Brazil
| | - Luciano F Drager
- Hospital Sírio-Libanês Research and Education Institute, São Paulo, Brazil; Instituto do Coração (InCor), Hospital das Clinicas da Faculdade de Medicina, Sao Paulo, Brazil
| | | | | | | | | | | | | | - Guilherme Loureiro Fialho
- Hospital Universitario Professor Polydoro Ernani de São Thiago, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | | | - Lucas P Damiani
- Brazilian Clinical Research Institute, São Paulo, Brazil; Instituto Dante Pazzanese de Cardiologia, Sao Paulo, Brazil
| | | | | | | | | | - Roberta Saretta
- Hospital Sírio-Libanês Research and Education Institute, São Paulo, Brazil
| | | | - Ludhmila Abrahao Hajjar
- Instituto do Cancer do Estado de São Paulo, Hospital das Clinicas da Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil; Brazilian Clinical Research Institute, São Paulo, Brazil; Instituto D´Or de Ensino e Pesquisa, Sao Paulo, Brazil
| | - Teresa Lopes-Fernandez
- Department of Cardiology, La Paz University Hospital, IdiPAZ Research Institute, Madrid, Spain; Department of Cardiology, Hospital Universitario Quirónsalud Madrid, Madrid, Spain.
| | - Susan Dent
- Wilmot Cancer Institute, University of Rochester, Rochester NY, USA
| | - C Michael Gibson
- Baim Research Institute and Harvard Medical School, Boston, MA, USA
| | - Renato D Lopes
- Hospital Sírio-Libanês Research and Education Institute, São Paulo, Brazil; Brazilian Clinical Research Institute, São Paulo, Brazil; Duke Clinical Research Institute, Duke University Medical Center, Durham, NC, USA
| | - Roberto Kalil Filho
- Hospital Sírio-Libanês Research and Education Institute, São Paulo, Brazil; Instituto do Coração (InCor), Hospital das Clinicas da Faculdade de Medicina, Sao Paulo, Brazil
| |
Collapse
|
2
|
Swiech O, Boguszewska-Czubara A. Enhancing cardiac safety: Liposomal ciprofloxacin mitigates anthracycline-induced cardiotoxicity without compromising anticancer efficacy. Chem Biol Interact 2025; 414:111496. [PMID: 40157626 DOI: 10.1016/j.cbi.2025.111496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 02/20/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
Anthracyclines, such as doxorubicin and epirubicin (EPI), are integral in the treatment of solid tumors and hematological malignancies but are associated with cardiotoxicity, potentially leading to heart failure. The underlying mechanisms involve the generation of reactive oxygen species (ROS), alterations in iron metabolism, and the inhibition of topoisomerase 2β (Top2β), resulting in mitochondrial dysfunction and cell death. Fluoroquinolones, including ciprofloxacin (CPX), enhance the efficacy of anthracyclines by inhibiting topoisomerase II and inducing apoptosis, thereby indicating a promising combination therapy. This study investigated the impact of environmental pH on the cardiotoxicity and myocardial accumulation of anthracyclines, as well as the cardioprotective and synergistic potential of CPX when co-administered with epirubicin. The findings revealed that only the zwitterionic form of CPX, either free or encapsulated in liposomes, offers significant cardioprotection without compromising the anticancer activity of EPI. Remarkably, the combination of liposomal CPX and EPI completely attenuates EPI's cardiotoxicity. These results suggest that initiating treatment with liposomal CPX prior to EPI administration may optimize cardioprotection while maintaining therapeutic efficacy against cancer.
Collapse
Affiliation(s)
- Olga Swiech
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093, Warsaw, Poland; BS Biotechna S.A, Szlak 77/222, Cracow, Poland.
| | - Anna Boguszewska-Czubara
- Department of Medical Chemistry, Medical University of Lublin, Chodzki 4a, 20-093, Lublin, Poland
| |
Collapse
|
3
|
Singh A, Bakhtyar M, Jun SR, Boerma M, Lan RS, Su LJ, Makhoul S, Hsu PC. A narrative review of metabolomics approaches in identifying biomarkers of doxorubicin-induced cardiotoxicity. Metabolomics 2025; 21:68. [PMID: 40381141 PMCID: PMC12085340 DOI: 10.1007/s11306-025-02258-8] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 04/04/2025] [Indexed: 05/19/2025]
Abstract
BACKGROUND While anthracyclines, commonly used in cancer treatment, are well known to cause cardiotoxicity, no validated biomarkers currently exist that can predict the early development of doxorubicin-induced cardiotoxicity (DIC). Therefore, identifying early biomarkers of DIC is urgently needed. Metabolomics approaches have been used to elucidate this relationship and identified related metabolite markers. However, differences in pre-clinical model systems make it challenging to draw definitive conclusions from the discoveries and translate findings into clinical applications. AIM OF REVIEW A systematic literature search on metabolomics studies of DIC was conducted with the goal to identify and compare study results reported using in vitro models, animal models, and studies from clinical patients. Metabolites identified across all studies were pooled to uncover biologically meaningful patterns that are significantly enriched in the data. Finally, pooled metabolites perturbed by DIC were mapped to metabolic pathways to explore potential pathological implications. RESULTS We reviewed 28 studies published between 2000 and 2024 that utilized metabolomics approaches to investigate DIC. The included studies used a variety of analytical techniques, including LC-MS, GC-MS, and NMR. The analysis revealed that metabolites such as inosine, phenylalanine, arginine, and tryptophan were commonly perturbed across all study models, with carnitine metabolism and purine and pyrimidine metabolism being the most affected pathways. Metabolite Set Enrichment Analysis (MSEA) using MetaboAnalyst identified the arginine biosynthesis, citrate cycle, and alanine, aspartate, and glutamate metabolism pathways as significantly enriched. CONCLUSION These findings underscore the potential of metabolomics in identifying early biomarkers for DIC, providing a foundation for future studies aimed at preventing cardiotoxicity and improving treatment strategies for cancer patients receiving DOX-containing therapies. KEY SCIENTIFIC CONCEPTS OF REVIEW Altogether, metabolomics studies suggest metabolic alterations in DIC, albeit little overlap between studies especially with animal and human studies. Attempts at intercepting these pathways have shown that intervention in DIC may be possible. Future research should focus on developing precise cardiotoxicity models that incorporate cancer metabolism, as these will be crucial in bridging the gap between laboratories (in vitro and animal models) and clinical studies to identify subclinical biomarkers in the early stage of DIC that can effectively identify new targets for interventions to reduce lethal cardiovascular disease risk.
Collapse
Affiliation(s)
- Amarnath Singh
- Department of Environmental Health Sciences, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Maham Bakhtyar
- Department of Internal Medicine, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Se-Ran Jun
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Marjan Boerma
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Renny S Lan
- Department of Pediatrics, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - L Joseph Su
- Peter O'Donnell Jr. School of Public Health, UT Southwestern Medical Center, Dallas, TX, USA
| | - Sam Makhoul
- CARTI Research Department, Little Rock, AR, USA
| | - Ping-Ching Hsu
- Department of Environmental Health Sciences, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|
4
|
Li J, Zeng Y, Liu F, Liao X, Zhong C, Dong S, Cai Y, Yang P. Erythrocyte Membrane-Camouflaged Xanthohumol Nanoparticles Mitigate Doxorubicin-Induced Cardiotoxicity by Inhibiting Ferroptosis. ACS Biomater Sci Eng 2025; 11:2727-2738. [PMID: 40305843 DOI: 10.1021/acsbiomaterials.4c02467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
Doxorubicin (DOX) chemotherapy is a cornerstone of cancer treatment, but its clinical application and effectiveness are severely restricted due to its life-threatening cardiotoxicity. Xanthohumol (XH), a compound from traditional Chinese medicine, is noted for its antioxidant properties and the potential to mitigate DOX-induced cardiotoxicity (DIC). However, its poor water solubility results in low biocompatibility, making it susceptible to immune system clearance, which severely restricts its application in vivo. In this study, we first identified and demonstrated that XH can effectively mitigate DIC by inhibiting ferroptosis. We designed a biomimetic nanodelivery system encapsulating XH within porous poly(lactic-co-glycolic acid) (PLGA) nanoparticles, further coated with an erythrocyte membrane (XH-NPs@RBCm). This system offers several advantages, including evasion of macrophage phagocytosis and prolonged circulation time, thereby enhancing the stability and bioavailability of XH in vivo. Treatment with XH-NPs@RBCm significantly reduced reactive oxygen species-dependent ferroptosis, improving the DOX-induced myocardial atrophy and cardiac dysfunction. Our study underscores the therapeutic promise of XH-NPs@RBCm in treating DIC through ferroptosis inhibition, offering key insights into biomimetic nanodelivery system development for DIC management.
Collapse
Affiliation(s)
- Jingchao Li
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
- Department of Cardiology, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou 450003, China
| | - Yinghua Zeng
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
- Department of Pediatric Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Fengjiao Liu
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Xu Liao
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Chongbin Zhong
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Shujuan Dong
- Department of Cardiology, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou 450003, China
| | - Yanbin Cai
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Pingzhen Yang
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| |
Collapse
|
5
|
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
|
6
|
Hindocha P, Lyon AR, Bhaskaran K, Strongman H. Cardioprotective drugs and heart failure/cardiomyopathy incidence in chemotherapy-treated cancer survivors of breast cancer and non-Hodgkin lymphoma: a retrospective cohort study in England. EUROPEAN HEART JOURNAL OPEN 2025; 5:oeaf039. [PMID: 40357261 PMCID: PMC12066945 DOI: 10.1093/ehjopen/oeaf039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/12/2024] [Accepted: 04/13/2025] [Indexed: 05/15/2025]
Abstract
Aims Evidence for the use of beta-blockers, angiotensin II receptor blockers (ARB), or angiotensin-converting enzyme inhibitors (ACEi) to mitigate chemotherapy-induced cardiotoxicity is inconclusive. The objectives are to investigate associations between prescription of ARBs, ACEis, and/or beta-blockers in the year following cancer diagnosis and subsequent risk of heart failure/cardiomyopathy (HF/CM) in chemotherapy-treated breast cancer and non-Hodgkin lymphoma (NHL) survivors. Methods and results This cohort study used linked English electronic healthcare records from 9875 adult (≥18 years) breast cancer and NHL survivors who received chemotherapy. Cox regression was used to estimate the association between primary care-prescribed beta-blocker, ARB, and ACEi use in the year following cancer diagnosis, and subsequent HF/CM incidence, adjusting for potential confounders. Likelihood ratio tests were used to assess effect modification. The mean follow-up duration was 4.9 years (maximum 21.4). After adjusting for age, the risk of HF/CM was higher in the exposed group [hazard ratio (HR): 1.69, 95% confidence interval (CI): 1.34-2.14], but further adjustment for gender, comorbidities, and other medications reduced the association to close to null (HR: 1.07, 95% CI: 0.68-1.69). There was no evidence that the association differed by cancer site, age, radiotherapy, prior cardiovascular disease, or years since cancer diagnosis. Conclusion We found no evidence that general practitioner prescribed beta-blocker, ARB, or ACEi use was associated with a reduced incidence of HF/CM in this population of chemotherapy-treated breast cancer and NHL survivors. This might be because the drug dosage and timing were not optimized to prevent chemotherapy-related cardiac damage; residual confounding by indication may also have obscured any treatment benefit.
Collapse
Affiliation(s)
- Pooja Hindocha
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1A 7HT, UK
| | - Alexander R Lyon
- National Heart and Lung Institute, Imperial College London and Cardio-Oncology Centre of Excellence, Royal Brompton Hospital, Sydney Street, London SW3 6NP, UK
| | - Krishnan Bhaskaran
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1A 7HT, UK
| | - Helen Strongman
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1A 7HT, UK
| |
Collapse
|
7
|
Zheng Y, Liu Z, Chen D, Zhang J, Yuan M, Zhang Y, Liu S, Zhang G, Yang G. The Cardiotoxicity Risk of Immune Checkpoint Inhibitors Compared with Chemotherapy: A Systematic Review and Meta-analysis of Observational Studies. Cardiovasc Toxicol 2025; 25:805-819. [PMID: 40053271 DOI: 10.1007/s12012-025-09979-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 02/21/2025] [Indexed: 04/24/2025]
Abstract
Immune checkpoint inhibitors (ICIs) have demonstrated favorable outcomes in various cancers. However, it has been observed that ICIs may induce life-threatening cardiovascular toxicity. In this study, a meta-analysis was conducted to determine the risk of cardiovascular toxicities in patients exposed to ICIs or in combination with chemotherapy. PubMed, Cochrane Library, and Embase databases were searched from inception to September 24, 2023. This study was conducted in accordance with the PRISMA guidelines. A meta-analysis was conducted on the risk of cardiotoxicity in cancer patients. Data were pooled with a random-effect model. This protocol was registered prospectively in PROSPERO (CRD42023467319). The primary outcome was cardiotoxicity risk in observational studies with ICIs or combined with chemotherapy. The risk factors that affected the occurrence of cardiovascular toxicities were also examined. ICIs or combined with chemotherapy increased the cardiotoxicity risk compared with mono-chemotherapy (OR 1.47; 95% CI 1.05-2.06, p = 0.024). The risk of pericardial disease in cardiotoxic events (OR 1.99; 95% CI 1.23-3.22, p = 0.005) and thromboembolic events (OR 1.34; 95% CI 1.04-1.72, p = 0.025) was significantly increased. Smoking (OR 1.25; 95% CI 1.12-1.39, p < 0.001), previous heart disease (OR 2.01; 95% CI 1.64-2.46, p < 0.001), and lung cancer (OR 1.46; 95% CI 1.26-1.69, p < 0.001) were risk factors worthy of attention. ICIs or combined with chemotherapy show an elevated risk of cardiovascular toxicities. Patients who are smoking, diagnosed lung cancer, and having prior medical history of heart diseases need more attention.
Collapse
Affiliation(s)
- Yingying Zheng
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Zishen Liu
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Dong Chen
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Jingzhi Zhang
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Mengqi Yuan
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Yutong Zhang
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Shiyu Liu
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Ganlin Zhang
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.
| | - Guowang Yang
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.
| |
Collapse
|
8
|
Wang W, Cai Y, Chen Y, Zhu J, Jin J. Comprehensive Analysis of the Multi-Target Binding Mechanism of Doxorubicin: Integrating Protein Microarray Screening, Molecular Docking, and Molecular Dynamics Simulation. Arch Pharm (Weinheim) 2025; 358:e70006. [PMID: 40345153 DOI: 10.1002/ardp.70006] [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: 02/15/2025] [Revised: 04/04/2025] [Accepted: 04/16/2025] [Indexed: 05/11/2025]
Abstract
Understanding the mechanisms through which anticancer drugs interact with multiple protein targets is crucial for optimizing drug design and enhancing the efficacy of chemotherapy. This study focuses on doxorubicin, a broad-spectrum anticancer drug recognized for its multi-target mechanisms of action. We initially screened 363 doxorubicin-binding proteins using protein microarrays; of these, 166 proteins with known PDB (Protein Data Bank) structures were selected for molecular docking to evaluate their binding energies. The binding energy distribution and residue enrichment analyses revealed that doxorubicin preferentially binds to specific residues at its binding sites, including serine, glycine, arginine, glutamic acid, lysine, aspartic acid, and leucine. These residues stabilize doxorubicin binding through hydrogen bonds, hydrophobic interactions, and electrostatic interactions. In addition, RUVBL1 (RuvB-like AAA ATPase 1) exhibited the highest integrated score from the protein microarray and molecular docking analyses. Furthermore, PPI (protein-protein interaction) network analysis and centrality calculations identified key proteins with potential regulatory roles, with MAPK1 (mitogen-activated protein kinase 1) exhibiting the highest betweenness centrality in the PPI network. Finally, molecular dynamics simulations of the RUVBL1- and MAPK1-doxorubicin complexes were conducted to evaluate the binding mechanisms. The simulations revealed key binding residues, including Ile56, Lys59, Leu87, Pro296, and Ile326 in RUVBL1 and Asp88, Ile89, Pro93, Phe354, and Ala92 in MAPK1 that mediate stable interactions with doxorubicin. This study presents a comprehensive analytical approach for investigating the interactions between doxorubicin and multiple protein targets, providing a reference framework for understanding the molecular mechanisms of anticancer drugs and for future analyses of similar data sets.
Collapse
Affiliation(s)
- Wentao Wang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu, China
- School of Chemical and Material Engineering, Jiangnan University, Wuxi, Jiangsu, China
| | - Yanfei Cai
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu, China
| | - Yun Chen
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu, China
| | - Jingyu Zhu
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu, China
| | - Jian Jin
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|
9
|
Gautam S, Joshi S, Jindal P, Patel P, Pal RR, Kumar M, Gupta GD, Kurmi BD. Recent doxorubicin-conjugates in cancer drug delivery: Exploring conjugation strategies for enhanced efficacy and reduced toxicity. Int J Pharm 2025; 675:125556. [PMID: 40187698 DOI: 10.1016/j.ijpharm.2025.125556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/21/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Doxorubicin is a first-line treatment of cancer that works on the mechanism of DNA intercalation and topoisomerase II poisoning. Since the 20th century, Doxorubicin has been used as a promising drug to treat several types of cancer, both solid or metastatic, including breast, thyroid, bladder, ovarian, or gastric cancer, etc. Even though it shows promising effects on cancer cells, it also shows its effects on healthy cells with cancerous cells, which leads to several severe side effects, such as cardiomyopathy, phlebitis, congestive heart failure (CHF), etc., which limits its usage in chemotherapy. Several research has focused on the targeted delivery of doxorubicin to cancerous cells to reduce side effects and improve efficacy. To optimize its anticancer potential, scientists have recently been developing nano-formulations and investigating various conjugations. The structure of doxorubicin consists of two primary functional groups that can be employed for conjugation with a variety of biomolecules, The first is the primary amine group in a sugar moiety, and the other one is the primary hydroxyl group in the aliphatic chain ring. In this paper, we have mentioned several conjugations of doxorubicin such as antibodies, nanoparticles, polymers, and phytochemical conjugations. Different studies regarding these conjugations are also mentioned, which represent promising strategies to optimize cancer treatment by minimizing side effects.
Collapse
Affiliation(s)
- Shreastha Gautam
- Department of Pharmaceutical Quality Assurance, ISF College of Pharmacy, GT Road, Moga-142001, Punjab, India
| | - Sachin Joshi
- Department of Pharmaceutical Quality Assurance, ISF College of Pharmacy, GT Road, Moga-142001, Punjab, India
| | - Priya Jindal
- Department of Pharmaceutical Quality Assurance, ISF College of Pharmacy, GT Road, Moga-142001, Punjab, India
| | - Preeti Patel
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, GT Road Moga-142001, Punjab, India.
| | - Ravi Raj Pal
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga-142001, Punjab, India
| | - Manish Kumar
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga-142001, Punjab, India
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga-142001, Punjab, India
| | - Balak Das Kurmi
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga-142001, Punjab, India.
| |
Collapse
|
10
|
Vaitiekus D, Muckiene G, Verikas D, Vaitiekiene A, Astasauskaite S, Gerbutavicius R, Bartnykaite A, Ugenskienė R, Jurkevičius R, Juozaitytė E. Drugs Metabolism-Related Genes Variants Impact on Anthracycline-Based Chemotherapy Induced Subclinical Cardiotoxicity in Breast Cancer Patients. Int J Mol Sci 2025; 26:4051. [PMID: 40362292 PMCID: PMC12072181 DOI: 10.3390/ijms26094051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/22/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Breast cancer is the most common cancer in women worldwide. Anthracyclines (doxorubicin, epirubicin, daunorubicin, idarubicin) are among the most used drugs for the treatment of breast cancer. Unfortunately, anthracyclines cause cardiotoxicity, which is a limiting factor for its use, and the lifetime cumulative dose of anthracyclines is the major risk factor for cardiotoxicity. In our study, we focused on acute and subacute heart damage. One of the main factors is a genetic predisposition, which determines individual susceptibility to anthracycline cardiotoxicity. The main idea of this study was, for the first time, to evaluate drug metabolism-related genes as a risk factor for developing cardiovascular toxicity in breast cancer patients. The main objective of our study was to identify the impact of drug metabolism-related gene SNPs on the development of subclinical heart damage during and/or after doxorubicin-based chemotherapy in breast cancer patients. The data of 81 women with breast cancer treated with doxorubicin-based chemotherapy in an outpatient clinic were analyzed, and SNP RT-PCR tests were performed. The drug metabolism-related gene variants SULT2B1 rs10426377, UGT1A6 rs17863783, CBR1 rs9024, CBR3 rs1056892, NCF4 rs1883112, and CYBA rs1049255 did not reach a statistically important impact on ABCC in multivariate logistic regression analysis. However, we identified that NCF4 rs1883112 had a risk reduction tendency for ABCC (OR = 0.49, 95% CI 0.27-0.87, p = 0.015). Our findings suggest that some SNPs, such as NCF4 rs1883112, may be associated with a reduced risk of cardiotoxicity, while no variants in this study showed a statistically significant increased risk. Even though, NCF4 rs1883112 showed a risk reduction tendency, suggesting the potential for personalized risk stratification. We can conclude that multiple genes are involved in ABCC, with different impacts, and it is unlikely that there is a single driver gene in ABCC pathogenesis.
Collapse
Affiliation(s)
- Domas Vaitiekus
- Clinic of Oncology and Hematology, Institute of Oncology, Faculty of Medicine, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania
| | - Gintare Muckiene
- Clinical Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania
| | - Dovydas Verikas
- Laboratory for Automation of Cardiovascular Investigation, Institute of Cardiology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania
| | - Audrone Vaitiekiene
- Clinical Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania
| | - Skaiste Astasauskaite
- Clinic of Oncology and Hematology, Institute of Oncology, Faculty of Medicine, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania
| | - Rolandas Gerbutavicius
- Clinic of Oncology and Hematology, Institute of Oncology, Faculty of Medicine, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania
| | - Agne Bartnykaite
- Clinic of Oncology and Hematology, Institute of Oncology, Faculty of Medicine, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania
| | - Rasa Ugenskienė
- Clinic of Oncology and Hematology, Institute of Oncology, Faculty of Medicine, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania
| | - Renaldas Jurkevičius
- Clinical Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania
| | - Elona Juozaitytė
- Clinic of Oncology and Hematology, Institute of Oncology, Faculty of Medicine, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania
| |
Collapse
|
11
|
Zeferino TG, Silva LM. Dexrazoxane as a viable microsporidia control agent in Anopheles gambiae. Acta Trop 2025; 266:107633. [PMID: 40288551 DOI: 10.1016/j.actatropica.2025.107633] [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: 02/22/2025] [Revised: 04/23/2025] [Accepted: 04/24/2025] [Indexed: 04/29/2025]
Abstract
Microsporidia have long been proposed as biological agents for controlling disease vectors and the parasites they transmit. However, their study in vector biology has been constrained due to challenges in manipulating microsporidia within hosts. In this study, we investigated the effect of Dexrazoxane, a candidate drug against microsporidiosis, on the establishment and development of Vavraia culicis infection in its natural host, the mosquito Anopheles gambiae, the main malaria vector. Our findings show that Dexrazoxane significantly reduces spore load, particularly in mosquitoes reared individually, without affecting the overall infection success of the parasite. This result aligns with studies in Caenorhabditis elegans, where Dexrazoxane inhibited new spore production without hindering initial spore integration into the host gut cells. Dexrazoxane's DNA topoisomerase II inhibitor mechanism likely explains its impact on mosquito development, as larvae exposed to the drug failed to emerge as adults. These findings highlight Dexrazoxane's potential as a viable tool for controlling microsporidia in adult mosquitoes and hope to enhance the study of mosquito-microsporidia interactions. Further research is required to explore its broader application in vector-borne disease control, including malaria.
Collapse
Affiliation(s)
- Tiago G Zeferino
- Institute of Biology, University of Neuchâtel, Neuchâtel, Switzerland
| | - Luis M Silva
- Institute of Biology, University of Neuchâtel, Neuchâtel, Switzerland; Department of Zoology, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
12
|
Ryan TD, Bates JE, Kinahan KE, Leger KJ, Mulrooney DA, Narayan HK, Ness K, Okwuosa TM, Rainusso NC, Steinberger J, Armenian SH. Cardiovascular Toxicity in Patients Treated for Childhood Cancer: A Scientific Statement From the American Heart Association. Circulation 2025; 151:e926-e943. [PMID: 40104841 DOI: 10.1161/cir.0000000000001308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
The field of cardio-oncology has expanded over the past 2 decades to address the ever-increasing issues related to cardiovascular disease in patients with cancer and survivors. There is increasing recognition that nearly all cancer treatments pose some short- or long-term risk for development of cardiovascular disease and that pediatric patients with cancer may be especially vulnerable to cardiovascular disease because of young age at treatment and expected long life span afterward. Anthracycline chemotherapy and chest-directed radiotherapy are the most well-studied cardiotoxic therapies, and dose reduction, use of cardioprotection for anthracyclines, and modern radiotherapy approaches have contributed to improved cardiovascular outcomes for survivors. Newer treatments such as small-molecule inhibitors, antibody-based cytotoxic therapy, and immunotherapy have expanded options for previously difficult-to-treat cancers but have also revealed new cardiotoxic profiles. Application of effective surveillance strategies in patients with cancer and survivors has been a focus of practitioners and researchers, whereas the prevention and treatment of extant cardiovascular disease is still developing. Incorporation of new strategies in an equitable manner and appropriate transition from pediatric to adult care will greatly influence long-term health-related outcomes in the growing population of childhood cancer survivors at risk for cardiovascular disease.
Collapse
|
13
|
Chen JK, Ramesh S, Islam MN, Shibu MA, Kuo CH, Hsieh DJY, Lin SZ, Kuo WW, Huang CY, Ho TJ. Artemisia argyi mitigates doxorubicin-induced cardiotoxicity by inhibiting mitochondrial dysfunction through the IGF-IIR/Drp1/GATA4 signaling pathway. Biotechnol Appl Biochem 2025; 72:388-401. [PMID: 39375847 DOI: 10.1002/bab.2671] [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: 06/04/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 10/09/2024]
Abstract
Doxorubicin (DOX) is mostly utilized as a wide range of antitumor anthracycline to treat different cancers. The severe antagonistic impacts of DOX on cardiotoxicity constrain its clinical application. Many mechanisms are involved in cardiac toxicity induced by DOX in the human body. Mitochondria is a central part of fatty acid and glucose metabolism. Thus, impaired mitochondrial metabolism can increase heart failure risk, which can play a vital role in cardiomyocyte mitochondrial dysfunction. This study aimed to assess the possible cardioprotective effect of water-extracted Artemisia argyi (AA) against the side effect of DOX in H9c2 cells and whether these protective effects are mediated through IGF-IIR/Drp1/GATA4 signaling pathways. Although several studies proved that AA extract has benefits for various diseases, its cardiac effects have not yet been identified. The H9c2 cells were exposed to 1 μM to establish a model of cardiac toxicity. The results revealed that water-extracted AA could block the expression of IGF-IIR/calcineurin signaling pathways induced by DOX. Notably, our results also showed that AA treatment markedly attenuated Akt phosphorylation and cleaved caspase 3, and the nuclear translocation markers NFATC3 and p-GATA4. Using actin staining for hypertrophy, we determined that AA can reduce the effect of mitochondrial reactive oxygen species and cell size. These findings suggest that water-extracted AA could be a suitable candidate for preventing DOX-induced cardiac damage.
Collapse
Affiliation(s)
- Jhong-Kuei Chen
- Department of Chinese Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| | - Samiraj Ramesh
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Department of Research and Innovation, Institute of Biotechnology, Saveetha School of Engineering (SSE), Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, India
| | - Md Nazmul Islam
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | | | - Chia-Hua Kuo
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei, Taiwan
| | - Dennis Jine-Yuan Hsieh
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Shinn-Zong Lin
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, College of Life Sciences, China Medical University, Taichung, Taiwan
- Ph.D. Program for Biotechnology Industry, China Medical University, Taichung, Taiwan
- School of Pharmacy, China Medical University, Taichung, Taiwan
| | - Chih-Yang Huang
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan
- Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien, Taiwan
| | - Tsung-Jung Ho
- Department of Chinese Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
- School of Post-Baccalaureate Chinese Medicine, College of Medicine, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
14
|
Alghaith N, Afif L, Justanieah RA, Alharbi S, Waggass R, Abdullatif D, Ahmed ME, Zaidi SF. Anthracycline-Induced Cardiomyopathy: Prevalence and Risk Factors Among Pediatric Cancer Patients in a Tertiary Care Center in Jeddah, Saudi Arabia. Cureus 2025; 17:e81704. [PMID: 40322385 PMCID: PMC12050047 DOI: 10.7759/cureus.81704] [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: 04/04/2025] [Indexed: 05/08/2025] Open
Abstract
Chemotherapy-induced cardiomyopathy (CCMP) is one of the well-defined toxicities associated with chemotherapy use that can lead to serious side effects. An example of a chemotherapeutic drug class that has been well documented over the years to cause CCMP is anthracyclines. To date, few studies have been carried out in Saudi Arabia on the prevalence of CCMP and the associated risk factors. Therefore, the objective of our research is to measure the prevalence and determine the risk factors of such phenomena. This is a comparative cross-sectional study. Data from 114 patients was retrieved from the medical records of the cardiac department at Princess Noorah Oncology Center, King Abdulaziz Medical City. The research included pediatric oncology patients aged 14 or under who were treated with anthracyclines from June 2016 to May 2024. We excluded patients who did not undergo ECHO. A consecutive sampling technique was used to collect the patients. Over the eight-year study period, we found that 7.34% (8/109) of the cohort developed CCMP, with a mean age at diagnosis of 6.39 ± 3.81 years. The mean dose of anthracycline received until the diagnosis of CCMP was 194.77±145.92 mg, with a median interval between anthracycline initiation and CCMP diagnosis of 13.65 (3-89) weeks. A significant association was found between thromboembolism, PDA, type of cancer, and the development of CCMP. We found that traditional predictors such as gender, age at diagnosis, and cumulative anthracycline dose were not predictors of CCMP.
Collapse
Affiliation(s)
- Nouf Alghaith
- Department of Pediatric Cardiology, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, SAU
| | - Lena Afif
- Department of Pediatric Cardiology, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, SAU
| | - Ragheed A Justanieah
- Department of Pediatric Cardiology, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, SAU
| | - Sereen Alharbi
- Department of Pediatric Cardiology, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, SAU
| | - Rahaf Waggass
- Department of Pediatric Cardiology, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, SAU
| | - Daniah Abdullatif
- Department of Pediatric Hematology Oncology, King Abdulaziz Medical City Ministry of National Guard Health Affairs, Jeddah, SAU
| | - Mohammed E Ahmed
- Biostatistics, King Abdullah International Medical Research Center, Jeddah, SAU
| | - Syed Faisal Zaidi
- Integrative/Complementary Medicine, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, SAU
- Integrative/Complementary Medicine, Faculty of Eastern Medicine, Hamdard University, Islamabad, PAK
| |
Collapse
|
15
|
Zhang X, Xue Y, Hao M. Cardiotoxicity induced by chemotherapy and immunotherapy in cancer treatment: a bibliometric analysis. Discov Oncol 2025; 16:376. [PMID: 40121610 PMCID: PMC11930912 DOI: 10.1007/s12672-025-02146-6] [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: 11/18/2024] [Accepted: 03/13/2025] [Indexed: 03/25/2025] Open
Abstract
OBJECTIVE New chemotherapy and immunotherapy agents have revolutionized cancer treatment, significantly improving patient survival rates and quality of life while extending lifespans. However, these therapies often come with severe side effects, particularly cardiotoxicity. Over the past few decades, this field has seen steady growth. To better understand current trends, research hotspots, and collaborative networks in this area, a bibliometric analysis of relevant literature was conducted. METHODS A comprehensive search was performed in the Web of Science for articles on cardiotoxicity induced by chemotherapy and immunotherapy in cancer treatment, published in SSCI and SCI-EXPANDED up to October 21, 2024. Using software tools such as GraphPad Prism, CiteSpace, and VOSviewer, we analyzed various parameters including publication year, countries, institutions, journals, authors, and references. Additionally, co-occurrence analyses, cooperation relationship assessments, co-citation networks, keyword maps, clustering analyses, and keyword emergence evaluations were conducted. RESULTS As of October 21, 2024, a total of 5290 articles from 5674 institutions and 27,528 authors across 114 countries and regions were collected. The annual publication frequency and rate steadily increased. The United States emerge as the leading country in terms of publication volume, with the University of Texas System being the most prolific and frequently cited institution. "Breast Cancer Research and Treatment" was among the journals with revelant publications. Notable contributors included Ky bonnie and Thavendiranathan Paaladinesh, while Cardinale D achieved the highest average citation count per publication. Current research hotspots included echocardiography, trastuzumab, doxorubicin, radiotherapy, myocarditis, and 5-fluorouracil. The trend suggests that cardiotoxicity is expected to play an increasingly critical role in chemotherapy and immunotherapy for cancer treatment. CONCLUSION This study provides a bibliometric visualization analysis of cardiotoxicity induced by chemotherapy and immunotherapy in the cancer treatment. It highlights current developments, collaborative efforts, and research hotspots within this field, offering essential scientific reference value for Cardio-Oncology.
Collapse
Affiliation(s)
- Xi Zhang
- Department of Nuclear Medicine, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yanfeng Xue
- Department of Special Need Medicine, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Mingyan Hao
- Department of Hospital Office, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, School of Public Health, Shanxi Medical University, Nicholas Global Health Institute, Duke University, No.3, Zhigong ST, Xinghualing District, Taiyuan, 030013, Shanxi, China.
| |
Collapse
|
16
|
Wang Z, Zhu Y, Yao Y, Zhang W, Wang B, Wang J, Yang Y, Liu L. Natural products targeting regulated cell deaths for adriamycin-induced cardiotoxicity. Cell Death Discov 2025; 11:112. [PMID: 40118839 PMCID: PMC11928682 DOI: 10.1038/s41420-025-02389-w] [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: 10/04/2024] [Revised: 02/01/2025] [Accepted: 03/06/2025] [Indexed: 03/24/2025] Open
Abstract
Adriamycin (ADR), as an anti-cancer drug in routine clinical application, is utilized to treat various cancers such as ovarian cancer, hematological malignant tumor, and endometrial carcinoma. However, its serious dose-dependent cardiotoxicity extremely limits its clinical application. Currently, there remains a dearth of therapeutic agents to mitigate ADR-induced cardiotoxicity. Extensive research has demonstrated that ADR can simultaneously trigger various regulated cell death (RCD) pathways, such as apoptosis, autophagy, ferroptosis, necroptosis, and pyroptosis. Therefore, drugs targeting these RCD pathways may represent effective strategies for treating ADR-induced cardiotoxicity. Natural products, with their wide availability, low cost, and diverse pharmacological activities, have increasingly gained attention. Various natural products, including polyphenols, flavonoids, terpenoids, and alkaloids, can target the RCD pathways involved in ADR-induced cardiotoxicity. Furthermore, these natural products have exhibited excellent properties in preclinical studies or in vitro experiments. This review summarizes the mechanisms of RCD in ADR-induced cardiotoxicity and systematically reviews the natural products targeting these RCD pathways. Finally, we propose future research directions of natural products in this field.
Collapse
Affiliation(s)
- Zheng Wang
- Xijing Hypertrophic Cardiomyopathy Center, Department of Ultrasound, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, China
- Department of Cardiothoracic Surgery, Central Theater Command General Hospital of Chinese People's Liberation Army, 627 Wuluo Road, Wuhan, 430070, China
| | - Yanli Zhu
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Yu Yao
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Wenyu Zhang
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Bo Wang
- Xijing Hypertrophic Cardiomyopathy Center, Department of Ultrasound, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, China
| | - Jing Wang
- Xijing Hypertrophic Cardiomyopathy Center, Department of Ultrasound, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, China
| | - Yang Yang
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China.
| | - Liwen Liu
- Xijing Hypertrophic Cardiomyopathy Center, Department of Ultrasound, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, China.
| |
Collapse
|
17
|
Lin J, Li B, Guo X, Li G, Zhang Q, Wang W. Key Mechanisms of Oxidative Stress-Induced Ferroptosis in Heart Failure with Preserved Ejection Fraction and Potential Therapeutic Approaches. Rev Cardiovasc Med 2025; 26:26613. [PMID: 40160560 PMCID: PMC11951494 DOI: 10.31083/rcm26613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/02/2024] [Accepted: 12/10/2024] [Indexed: 04/02/2025] Open
Abstract
The prevalence of heart failure with preserved ejection fraction (HFpEF) is increasing annually, particularly among patients with metabolic disorders such as hypertension and diabetes. However, there is currently no treatment capable of altering the natural course of HFpEF. Recently, the interplay between oxidative stress and ferroptosis in cardiovascular diseases has drawn extensive attention; however, minimal research has been published on the mechanisms of oxidative stress and ferroptosis in HFpEF. This paper reviews the relevant mechanisms through which oxidative stress is induced and promotes ferroptosis during the development of HFpEF. The review also explores more efficacious treatment approaches for HFpEF by inhibiting oxidative stress and ferroptosis, thereby offering a theoretical foundation for verifying the feasibility of these methods for further research. As tumor-targeted therapy progresses, the survival period of tumor patients is prolonged, and cardiovascular events have gradually emerged as one of the most crucial causes of death among tumor patients. Hence, inhibiting the vascular endothelial growth factor (VEGF) pathway has become a major target in tumor treatment, significantly enhancing patient survival. Nevertheless, secondary cardiovascular complications and events, such as myocardial injury and subsequent heart failure, have severely impacted patient survival and quality of life. Therefore we have also explored the potential mechanism through which novel targeted anti-cancer drugs induce HFpEF via ferroptosis. Additionally, we reviewed the specific modes of action for preventing and treating HFpEF without influencing their anti-cancer therapeutic effect.
Collapse
Affiliation(s)
- Junling Lin
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, 313000 Huzhou, Zhejiang, China
| | - Bingtao Li
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, 313000 Huzhou, Zhejiang, China
| | - Xueqi Guo
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, 313000 Huzhou, Zhejiang, China
| | - Guodong Li
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, 313000 Huzhou, Zhejiang, China
| | - Qi Zhang
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, 313000 Huzhou, Zhejiang, China
| | - Wenjuan Wang
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, 313000 Huzhou, Zhejiang, China
| |
Collapse
|
18
|
Lin R, Tian A, Wang Y, Wang X, Yuan X, Yu J, Li G, Xie W. The predictive value of changes in 18F-FDG PET/CT cardiac uptake patterns and metabolic parameters for anthracycline based chemotherapy induced cardiac toxicity in lymphoma patients. PLoS One 2025; 20:e0319442. [PMID: 40014616 PMCID: PMC11867325 DOI: 10.1371/journal.pone.0319442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 01/31/2025] [Indexed: 03/01/2025] Open
Abstract
OBJECTIVE This study aims to examine alterations in positron emission tomography with 2-deoxy-2-[fluorine-18]fluoro-D-glucose integrated with computed tomography (18F-FDG PET/CT) heart uptake patterns and metabolic factors before and after anthracycline-based chemotherapy in lymphoma patients, and to investigate the added benefit of oncological 18F-FDG PET/CT in chemotherapy-induced heart damage. MATERIALS AND METHODS Between July 2017 and December 2022, lymphoma patients diagnosed at the Second Affiliated Hospital of Dalian Medical University who underwent 6 cycles of anthracycline-based chemotherapy and had baseline and 6-cycle oncological 18F-FDG PET/CT scans were included. A total of 366 patients with complete data sets were enrolled. Relevant parameters including blood tests, lipid profile, cardiac biomarkers, lactate dehydrogenase (LDH), erythrocyte sedimentation rate (ESR), albumin (ALB), β2-microglobulin (β2-MG), and cardiac ultrasound findings were collected. Patients were monitored from the initiation of chemotherapy until January 2024, and the occurrence of cancer therapy-related cardiovascular toxicity (CTR-CVT) was documented. Changes in PET/CT heart uptake patterns pre- and post-treatment, along with the presence or absence of CTR-CVT, were used to analyze alterations in left ventricular and epicardial adipose tissue metabolic parameters, as well as changes in echocardiographic parameters. Logistic regression analysis was employed to identify risk factors for CTR-CVT. RESULTS Among lymphoma patients who received 6 cycles of anthracycline-based chemotherapy, compared to their initial state, there was a notable decrease in white blood cell count (WBC), neutrophil-to-lymphocyte ratio (NLR), erythrocyte sedimentation rate (ESR), and β2-microglobulin (β2-MG) levels post-treatment. Conversely, albumin (ALB) levels and blood lipid levels significantly rose after treatment. Post-treatment, the maximum standardized uptake value (SUVmax) and mean standardized uptake value (SUVmean) of the left ventricle significantly increased, and the percentage of patients exhibiting no uptake pattern in the left ventricle significantly decreased, while those with diffuse uptake pattern notably increased. Moreover, the count of patients with abnormal cardiac uptake significantly rose post-treatment. Analyzing changes in uptake patterns, the group displaying abnormal changes exhibited an increase in left atrial diameter and a decrease in left ventricular ejection fraction compared to the group with normal changes. The SUVmax of the epicardial adipose tissue was notably higher in the abnormal change group compared to the normal change group. Based on the presence or absence of CTR-CVT, the CTR-CVT group showcased higher left atrial diameter and left ventricular end-systolic diameter, and lower left ventricular ejection fraction compared to the non-CTR-CVT group. Additionally, the SUVmax and SUVmean of the epicardial adipose tissue were higher in the CTR-CVT group than in the non-CTR-CVT group. Left atrial end-systolic diameter, left ventricular ejection fraction, SUVmax of the epicardial adipose tissue, and change in uptake pattern were identified as risk factors for CTR-CVT. CONCLUSION In lymphoma patients treated with anthracycline-based chemotherapy, alterations in 18F-FDG PET/CT cardiac uptake patterns and metabolic parameters observed during the follow-up period before and after treatment, as well as changes in epicardial adipose tissue metabolic parameters post-treatment, could serve as predictors for the occurrence of CTR-CVT.
Collapse
Affiliation(s)
- Runlong Lin
- Department of Nuclear Medicine, The Second Hospital of Dalian Medical University, Dalian, China
| | - Aijuan Tian
- Department of Nuclear Medicine, The Second Hospital of Dalian Medical University, Dalian, China
| | - Ying Wang
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, China
| | - Xiaomei Wang
- Department of Nuclear Medicine, The Second Hospital of Dalian Medical University, Dalian, China
| | - Xin Yuan
- Department of Nuclear Medicine, The Second Hospital of Dalian Medical University, Dalian, China
| | - Jing Yu
- Department of Nuclear Medicine, The Second Hospital of Dalian Medical University, Dalian, China
| | - Guihua Li
- Department of Health Management Center, The Second Hospital of Dalian Medical University, Dalian, China
| | - Wenli Xie
- Department of Cardiovascular Medicine, The Second Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
19
|
Sun Y, Xiao L, Chen L, Wang X. Doxorubicin-Induced Cardiac Remodeling: Mechanisms and Mitigation Strategies. Cardiovasc Drugs Ther 2025:10.1007/s10557-025-07673-6. [PMID: 40009315 DOI: 10.1007/s10557-025-07673-6] [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: 01/16/2025] [Indexed: 02/27/2025]
Abstract
BACKGROUND The therapeutic prowess of doxorubicin in oncology is marred by its cardiotoxic consequences, manifesting as cardiac remodeling. Pathophysiological alterations triggered by doxorubicin include inflammatory cascades, fibrotic tissue deposition, vascular and valvular changes, and finally cardiomyopathy. These multifarious consequences collectively orchestrate the deterioration of cardiac architecture and function. METHOD By charting the molecular underpinnings and remedial prospects, this review aspires to contribute a novel perspective using latest publications to the ongoing quest for cardioprotection in cancer therapy. RESULTS AND DISCUSSION Experimental analyses demonstrate the pivotal roles of oxidative stress and subsequent necrosis and apoptosis of cardiomyocytes, muscle cells, endothelial cells, and small muscle cells in different parts of the heart. In addition, severe and unusual infiltration of macrophages, mast cells, and neutrophils can amplify oxidative damage and subsequent impacts such as chronic inflammatory responses, vascular and valvular remodeling, and fibrosis. These modifications can render cardiomyopathy, ischemia, heart attack, and other disorders. In an endeavor to counteract these ramifications, a spectrum of emerging adjuvants and strategies are poised to fortify the heart against doxorubicin's deleterious effects. CONCLUSION The compendium of mitigation tactics such as innovative pharmacological agents hold the potential to attenuate the cardiotoxic burden.
Collapse
Affiliation(s)
- Yanna Sun
- Department of Cardiology, The First Affiliated of Zhengzhou University, Zhengzhou City Henan Province, 450052, China
| | - Lili Xiao
- Department of Cardiology, The First Affiliated of Zhengzhou University, Zhengzhou City Henan Province, 450052, China
| | - Linlin Chen
- Department of Cardiology, The First Affiliated of Zhengzhou University, Zhengzhou City Henan Province, 450052, China
| | - Xiaofang Wang
- Department of Cardiology, The First Affiliated of Zhengzhou University, Zhengzhou City Henan Province, 450052, China.
| |
Collapse
|
20
|
Romano LR, Polimeni A, Indolfi C, Curcio A. Management of Cardiac Rhythm Disorders in Cardio-oncology. Arrhythm Electrophysiol Rev 2025; 14:e05. [PMID: 40084345 PMCID: PMC11904418 DOI: 10.15420/aer.2024.20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 08/16/2024] [Indexed: 03/16/2025] Open
Abstract
Arrhythmias and cancer are two pathological conditions that often coexist due to a patient's pre-existing comorbidities, or toxicity linked to anti-neoplastic drugs, and both are often characterised by poor prognosis. Cardio-oncology is a new interdisciplinary field that focuses on the cardiovascular health of cancer patients, especially those undergoing cancer treatment. Furthermore, cardiotoxicity can cause arrhythmias through primary and secondary mechanisms. Chemotherapy drugs have been shown to directly affect molecular pathways associated with arrhythmia development, as well as indirectly through mechanisms involving ischaemia or inflammatory injury to the heart. Understanding how to prevent and to treat these electrophysiological issues in cancer is an important challenge for cardio-oncologists. This review explores the intersection between cardio-oncology and electrophysiology, the various cardiac cell types implicated in the development of arrhythmias during cancer, the interplay between arrhythmias and cancer pathogenesis, and the need for the implantation of electronic devices along with their associated risks.
Collapse
Affiliation(s)
- Letizia Rosa Romano
- Department of Pharmacy, Health and Nutritional Sciences, Università della CalabriaRende, Italy
- Division of Cardiology, Annunziata HospitalCosenza, Italy
| | - Alberto Polimeni
- Department of Pharmacy, Health and Nutritional Sciences, Università della CalabriaRende, Italy
- Division of Cardiology, Annunziata HospitalCosenza, Italy
| | - Ciro Indolfi
- Division of Cardiology, Annunziata HospitalCosenza, Italy
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia UniversityCatanzaro, Italy
| | - Antonio Curcio
- Department of Pharmacy, Health and Nutritional Sciences, Università della CalabriaRende, Italy
- Division of Cardiology, Annunziata HospitalCosenza, Italy
| |
Collapse
|
21
|
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
|
22
|
Furukawa K, Ohmura H, Moriyama S, Uehara K, Ito M, Tsuchihashi K, Isobe T, Ariyama H, Fukata M, Kusaba H, Shiose A, Akashi K, Baba E. Treatment of malignant primary cardiac tumors requires attention to cardiovascular complications: a single-center, retrospective study. Jpn J Clin Oncol 2025; 55:113-122. [PMID: 39385509 DOI: 10.1093/jjco/hyae138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 10/03/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Malignant primary cardiac tumors require multimodal approaches including surgery, chemotherapy and radiotherapy, but these treatments can be associated with cardiovascular complications. However, few reports have described the cardiovascular complications related to primary cardiac tumor treatment because of their rarity. METHODS Clinical records of patients with primary cardiac tumors treated at Kyushu University Hospital from January 2010 to August 2021 were retrospectively examined. RESULTS Of the 47 primary cardiac tumor patients, 13 (28%) were diagnosed with malignancy, including 5 angiosarcomas, 3 intimal sarcomas, 3 diffuse large B-cell lymphomas, 1 Ewing's sarcoma and 1 fibrosarcoma. Cardiovascular events were observed in 10 patients (77%), including cardiac dysfunction in 6 patients, arrhythmias in 5 patients, right heart failure in 2 patients, and excessively prolonged prothrombin time due to the combination of warfarin and chemotherapy in 1 patient. Two patients who showed notable cardiac complications are described. Case A involved a 69-year-old woman who underwent surgery for a left atrial intimal sarcoma, followed by postoperative chemotherapy with doxorubicin plus ifosfamide and radiotherapy. After three cycles of chemotherapy and sequential radiotherapy, her left ventricular ejection fraction decreased to 34%, and ongoing heart failure therapy was required. Case B involved a 66-year-old man who received chemotherapy for primary cardiac lymphoma, resulting in tumor shrinkage. However, due to tumor involvement of the intraventricular septum, atrioventricular block developed, requiring cardiac pacemaker implantation. CONCLUSION High incidences of cardiac failure and arrhythmias were observed during multimodal treatments for malignant primary cardiac tumors. Proper management of complications may lead to a favorable prognosis in patients with malignant primary cardiac tumors.
Collapse
Affiliation(s)
- Kanami Furukawa
- Department of Hematology/Oncology, Japan Community Healthcare Organization Kyushu Hospital, Kitakyushu, Fukuoka, Japan
| | - Hirofumi Ohmura
- Department of Oncology and Social Medicine, Kyushu University Graduate School of Medical Sciences, Higashi-ku, Fukuoka, Japan
| | - Shohei Moriyama
- Department of Hematology, Oncology and Cardiovascular Medicine, Kyushu University Hospital, Higashi-ku, Fukuoka, Japan
| | - Koki Uehara
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Higashi-ku, Fukuoka, Japan
| | - Mamoru Ito
- Department of Hematology, Oncology and Cardiovascular Medicine, Kyushu University Hospital, Higashi-ku, Fukuoka, Japan
| | - Kenji Tsuchihashi
- Department of Hematology, Oncology and Cardiovascular Medicine, Kyushu University Hospital, Higashi-ku, Fukuoka, Japan
| | - Taichi Isobe
- Department of Oncology and Social Medicine, Kyushu University Graduate School of Medical Sciences, Higashi-ku, Fukuoka, Japan
| | - Hiroshi Ariyama
- Department of Oncology, Kitakyushu Municipal Medical Center, Kitakyushu, Fukuoka, Japan
| | - Mitsuhiro Fukata
- Department of Hematology, Oncology and Cardiovascular Medicine, Kyushu University Hospital, Higashi-ku, Fukuoka, Japan
| | - Hitoshi Kusaba
- Department of Medical Oncology, Hamanomachi Hospital, Chuo-ku, Fukuoka, Japan
| | - Akira Shiose
- Department of Cardiovascular Surgery, Kyushu University Hospital, Higashi-ku, Fukuoka, Japan
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Higashi-ku, Fukuoka, Japan
| | - Eishi Baba
- Department of Oncology and Social Medicine, Kyushu University Graduate School of Medical Sciences, Higashi-ku, Fukuoka, Japan
| |
Collapse
|
23
|
Liu X, Li Z. The role and mechanism of epigenetics in anticancer drug-induced cardiotoxicity. Basic Res Cardiol 2025; 120:11-24. [PMID: 38724618 DOI: 10.1007/s00395-024-01054-0] [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: 01/29/2024] [Revised: 04/20/2024] [Accepted: 05/03/2024] [Indexed: 05/23/2024]
Abstract
Cardiovascular disease is the main factor contributing to the global burden of diseases, and the cardiotoxicity caused by anticancer drugs is an essential component that cannot be ignored. With the development of anticancer drugs, the survival period of cancer patients is prolonged; however, the cardiotoxicity caused by anticancer drugs is becoming increasingly prominent. Currently, cardiovascular disease has emerged as the second leading cause of mortality among long-term cancer survivors. Anticancer drug-induced cardiotoxicity has become a frontier and hot topic. The discovery of epigenetics has given the possibility of environmental changes in gene expression, protein synthesis, and traits. It has been found that epigenetics plays a pivotal role in promoting cardiovascular diseases, such as heart failure, coronary heart disease, and hypertension. In recent years, increasing studies have underscored the crucial roles played by epigenetics in anticancer drug-induced cardiotoxicity. Here, we provide a comprehensive overview of the role and mechanisms of epigenetics in anticancer drug-induced cardiotoxicity.
Collapse
Affiliation(s)
- Xuening Liu
- Department of Pharmacy, Peking University Third Hospital, Beijing, 100191, China
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Zijian Li
- Department of Pharmacy, Peking University Third Hospital, Beijing, 100191, China.
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China.
| |
Collapse
|
24
|
Azcona JA, Wacker AS, Lee CH, Fung EK, Jeitner TM, Manzo OL, Di Lorenzo A, Babich JW, Amor-Coarasa A, Kelly JM. 2-[ 18F]Fluoropropionic Acid PET Imaging of Doxorubicin-Induced Cardiotoxicity. Mol Imaging Biol 2025; 27:109-119. [PMID: 39810069 PMCID: PMC11805620 DOI: 10.1007/s11307-024-01978-y] [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: 08/07/2024] [Revised: 10/21/2024] [Accepted: 12/10/2024] [Indexed: 01/16/2025]
Abstract
PURPOSE Treatment of pediatric cancers with doxorubicin is a common and predictable cause of cardiomyopathy. Early diagnosis of treatment-induced cardiotoxicity and intervention are major determinants for the prevention of advanced disease. The onset of cardiomyopathies is often accompanied by profound changes in lipid metabolism, including an enhanced uptake of short-chain fatty acids (SCFA). Therefore, we explored the utility of 2-[18F]fluoropropionic acid ([18F]FPA), an SCFA analog, as an imaging biomarker of cardiac injury in mice exposed to doxorubicin. PROCEDURES Cardiotoxicity and cardiac dysfunction were induced in mice by an 8-dose regimen of doxorubicin (cumulative dose 24 mg/kg) administered over 14 days. The effects of doxorubicin exposure were assessed by measurement of heart weights, left ventricular ejection fractions, and blood cardiac troponin levels. Whole body and cardiac [18F]FPA uptakes were determined by PET and tissue gamma counting in the presence or absence of AZD3965, a pharmacological inhibitor of monocarboxylate transporter 1 (MCT1). Radiation absorbed doses were estimated using tissue time-activity concentrations. RESULTS Significantly higher cardiac [18F]FPA uptake was observed in doxorubicin-treated animals. This uptake remained constant from 30 to 120 min post-injection. Pharmacological inhibition of MCT1-mediated transport by AZD3965 selectively decreased the uptake of [18F]FPA in tissues other than the heart. Co-administration of [18F]FPA and AZD3965 enhanced the imaging contrast of the diseased heart while reducing overall exposure to radioactivity. CONCLUSIONS [18F]FPA, especially when co-administered with AZD3965, is a new tool for imaging changes in fatty acid metabolism occurring in response to doxorubicin-induced cardiomyopathy by PET.
Collapse
Affiliation(s)
- Juan A Azcona
- Department of Radiology, Weill Cornell Medicine, 413 E 69th Street, Room BB-1604, New York, NY, 10021, USA
| | - Anja S Wacker
- Department of Radiology, Weill Cornell Medicine, 413 E 69th Street, Room BB-1604, New York, NY, 10021, USA
| | - Chul-Hee Lee
- Department of Radiology, Weill Cornell Medicine, 413 E 69th Street, Room BB-1604, New York, NY, 10021, USA
| | - Edward K Fung
- Department of Radiology, Weill Cornell Medicine, 413 E 69th Street, Room BB-1604, New York, NY, 10021, USA
- Citigroup Biomedical Imaging Center, Weill Cornell Medicine, New York, NY, USA
| | - Thomas M Jeitner
- Department of Radiology, Weill Cornell Medicine, 413 E 69th Street, Room BB-1604, New York, NY, 10021, USA
| | - Onorina L Manzo
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
- Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Annarita Di Lorenzo
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
- Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - John W Babich
- Department of Radiology, Weill Cornell Medicine, 413 E 69th Street, Room BB-1604, New York, NY, 10021, USA
- Citigroup Biomedical Imaging Center, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Ratio Therapeutics, Boston, MA, USA
| | - Alejandro Amor-Coarasa
- Department of Radiology, Albert Einstein College of Medicine of Yeshiva University, New York, NY, USA
- Ratio Therapeutics, Boston, MA, USA
| | - James M Kelly
- Department of Radiology, Weill Cornell Medicine, 413 E 69th Street, Room BB-1604, New York, NY, 10021, USA.
- Citigroup Biomedical Imaging Center, Weill Cornell Medicine, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
25
|
Lohr D, Thiele A, Stahnke M, Braun VM, Klopfleisch R, Klein O, Dresen S, Landmesser U, Foryst-Ludwig A, Kintscher U, Schreiber LM, Beyhoff N. Characterization of anthracycline-induced cardiotoxicity by diffusion tensor magnetic resonance imaging. Basic Res Cardiol 2025; 120:57-69. [PMID: 38483601 PMCID: PMC11790774 DOI: 10.1007/s00395-024-01039-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 02/05/2024] [Accepted: 02/05/2024] [Indexed: 02/04/2025]
Abstract
Anthracyclines are highly potent anti-cancer drugs, but their clinical use is limited by severe cardiotoxic side effects. The impact of anthracycline-induced cardiotoxicity (AIC) on left ventricular (LV) microarchitecture and diffusion properties remains unknown. This study sought to characterize AIC by cardiovascular magnetic resonance diffusion tensor imaging (DTI). Mice were treated with Doxorubicin (DOX; n = 16) for induction of AIC or saline as corresponding control (n = 15). Cardiac function was assessed via echocardiography at the end of the study period. Whole hearts (n = 8 per group) were scanned ex vivo by high-resolution DTI at 7 T. Results were correlated with histopathology and mass spectrometry imaging. Mice with AIC demonstrated systolic dysfunction (LVEF 52 ± 3% vs. 43 ± 6%, P < 0.001), impaired global longitudinal strain (-19.6 ± 2.0% vs. -16.6 ± 3.0%, P < 0.01), and cardiac atrophy (LV mass index [mg/mm], 4.3 ± 0.1 vs. 3.6 ± 0.2, P < 0.01). Regional sheetlet angles were significantly lower in AIC, whereas helix angle and relative helicity remained unchanged. In AIC, fractional anisotropy was increased (0.12 ± 0.01 vs. 0.14 ± 0.02, P < 0.05). DOX-treated mice displayed higher planar and less spherical anisotropy (CPlanar 0.07 ± 0.01 vs. 0.09 ± 0.01, P < 0.01; CSpherical 0.89 ± 0.01 vs. 0.87 ± 0.02, P < 0.05). CPlanar and CSpherical yielded good discriminatory power to distinguish between mice with and without AIC (c-index 0.91 and 0.84, respectively, P for both < 0.05). AIC is associated with regional changes in sheetlet angle but no major abnormalities of global LV microarchitecture. The geometric shape of the diffusion tensor is altered in AIC. DTI may provide a new tool for myocardial characterization in patients with AIC, which warrants future clinical studies to evaluate its diagnostic utility.
Collapse
Affiliation(s)
- David Lohr
- Chair of Molecular and Cellular Imaging, Comprehensive Heart Failure Center (CHFC), University Hospital Wuerzburg, Wuerzburg, Germany
| | - Arne Thiele
- Max Rubner Center for Cardiovascular Metabolic Renal Research, Institute of Pharmacology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Experimental and Clinical Research Center, a joint cooperation of Max-Delbrück Center for Molecular Medicine and Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Nephrology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Max Stahnke
- Max Rubner Center for Cardiovascular Metabolic Renal Research, Institute of Pharmacology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Vera M Braun
- Max Rubner Center for Cardiovascular Metabolic Renal Research, Institute of Pharmacology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Robert Klopfleisch
- Department of Veterinary Pathology, College of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Oliver Klein
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapy (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sandra Dresen
- Max Rubner Center for Cardiovascular Metabolic Renal Research, Institute of Pharmacology, 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
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Berlin, Germany
| | - Anna Foryst-Ludwig
- Max Rubner Center for Cardiovascular Metabolic Renal Research, Institute of Pharmacology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Ulrich Kintscher
- Max Rubner Center for Cardiovascular Metabolic Renal Research, Institute of Pharmacology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Laura M Schreiber
- Chair of Molecular and Cellular Imaging, Comprehensive Heart Failure Center (CHFC), University Hospital Wuerzburg, Wuerzburg, Germany
| | - Niklas Beyhoff
- Max Rubner Center for Cardiovascular Metabolic Renal Research, Institute of Pharmacology, Charité - Universitätsmedizin Berlin, Berlin, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Berlin, Germany.
| |
Collapse
|
26
|
Yoshikawa N, Hirata N, Kurone Y, Shimoeda S. Edaravone is a Therapeutic Candidate for Doxorubicin-Induced Cardiomyopathy by Activating the Nrf2 Pathway. Pharmacol Res Perspect 2025; 13:e70066. [PMID: 39865410 PMCID: PMC11761446 DOI: 10.1002/prp2.70066] [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: 10/01/2024] [Revised: 12/18/2024] [Accepted: 01/07/2025] [Indexed: 01/28/2025] Open
Abstract
Doxorubicin (DOXO) has long been used clinically and remains a key drug in cancer therapy. DOXO-induced cardiomyopathy (DICM) is a chronic and fatal complication that severely limits the use of DOXO. However, there are very few therapeutic agents for DICM, and there is an urgent need to identify those that can be used for a larger number of patients. The most likely pathogenic mechanism of DICM is the involvement of reactive oxygen species (ROS) and promotion of cell death. In this study, we investigated the efficacy and mechanism of action of edaravone (EDA), a known radical scavenger in DICM. Two methods of EDA administration were employed: daily and weekly. Our results showed that the daily administration group had prolonged survival periods and preserved the left ventricular ejection fraction in DICM mice. In contrast, in the weekly treatment group, slight improvements were observed in these indicators compared with those in DICM mice; however, none of them were statistically significant. These results show that the daily administration group had a higher efficacy than the weekly administration group. Gene-expression results showed that Nrf2 and its related genes were upregulated in the daily group but not in the weekly group. Based on these results, we hypothesized that the Sirt1/Nrf2/HO-1 and ABCB4 pathways were involved in EDA. However, there is limited evidence that EDA is effective against DICM. The findings obtained herein bolster the evidence in DICM by demonstrating prolonged survival and continued preservation of cardiac function and proposing a possible mechanism.
Collapse
Affiliation(s)
- Naoki Yoshikawa
- Department of Pharmaceutical Health Care and SciencesTokyo University of Pharmacy and Life SciencesTokyoJapan
| | - Naoto Hirata
- Department of Pharmaceutical Health Care and SciencesTokyo University of Pharmacy and Life SciencesTokyoJapan
| | - Yuichiro Kurone
- Department of Pharmaceutical Health Care and SciencesTokyo University of Pharmacy and Life SciencesTokyoJapan
| | - Sadahiko Shimoeda
- Department of Pharmaceutical Health Care and SciencesTokyo University of Pharmacy and Life SciencesTokyoJapan
| |
Collapse
|
27
|
Van Asbroeck B, Krüger DN, Van den Bogaert S, Dombrecht D, Bosman M, Van Craenenbroeck EM, Guns PJ, van Breda E. Distinct Impact of Doxorubicin on Skeletal Muscle and Fat Metabolism in Mice: Without Dexrazoxane Effect. Int J Mol Sci 2025; 26:1177. [PMID: 39940943 PMCID: PMC11818201 DOI: 10.3390/ijms26031177] [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] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
The chemotherapeutic agent doxorubicin (DOX) leads to the loss of skeletal muscle and adipose tissue mass, contributing to cancer cachexia. Experimental research on the molecular mechanisms of long-term DOX treatment is modest, and its effect on both skeletal muscle and adipose tissue has not been studied in an integrative manner. Dexrazoxane (DEXRA) is used to prevent DOX-induced cancer-therapy-related cardiovascular dysfunction (CTRCD), but its impact on skeletal muscle and adipose tissue remains elusive. Therefore, this study aimed to investigate the long-term effects of DOX on adipose tissue and skeletal muscle metabolism, and evaluate whether DEXRA can mitigate these effects. To this end, 10-week-old male C57BL6/J mice (n = 32) were divided into four groups: (1) DOX, (2) DOX-DEXRA combined, (3) DEXRA and (4) control. DOX (4 mg/kg weekly) and DEXRA (40 mg/kg weekly) were administered intraperitoneally over 6 weeks. Indirect calorimetry was used to assess metabolic parameters, followed by a molecular analysis and histological evaluation of skeletal muscle and adipose tissue. DOX treatment led to significant white adipose tissue (WAT) loss (74%) and moderate skeletal muscle loss (Gastrocnemius (GAS): 10%), along with decreased basal activity (53%) and energy expenditure (27%). A trend toward a reduced type IIa fiber cross-sectional area and a fast-to-slow fiber type switch in the Soleus muscle was observed. The WAT of DOX-treated mice displayed reduced Pparg (p < 0.0001), Cd36 (p < 0.0001) and Glut4 (p < 0.05) mRNA expression-markers of fat and glucose metabolism-compared to controls. In contrast, the GAS of DOX-treated mice showed increased Cd36 (p < 0.05) and Glut4 (p < 0.01), together with elevated Pdk4 (p < 0.001) mRNA expression-suggesting reduced carbohydrate oxidation-compared to controls. Additionally, DOX increased Murf1 (p < 0.05) and Atrogin1 (p < 0.05) mRNA expression-markers of protein degradation-compared to controls. In both the WAT and GAS of DOX-treated mice, Ppard mRNA expression remained unchanged. Overall, DEXRA failed to prevent these DOX-induced changes. Collectively, our results suggest that DOX induced varying degrees of wasting in adipose tissue and skeletal muscle, driven by distinct mechanisms. While DEXRA protected against DOX-induced CTRCD, it did not counteract its adverse effects on skeletal muscle and adipose tissue.
Collapse
Affiliation(s)
- Birgit Van Asbroeck
- Department of Rehabilitation Sciences & Physiotherapy, Research Group MOVANT, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium; (B.V.A.); (D.D.); (E.v.B.)
- Laboratory of Physiopharmacology, GENCOR, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium; (D.N.K.); (S.V.d.B.); (M.B.)
| | - Dustin N. Krüger
- Laboratory of Physiopharmacology, GENCOR, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium; (D.N.K.); (S.V.d.B.); (M.B.)
| | - Siel Van den Bogaert
- Laboratory of Physiopharmacology, GENCOR, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium; (D.N.K.); (S.V.d.B.); (M.B.)
| | - Dorien Dombrecht
- Department of Rehabilitation Sciences & Physiotherapy, Research Group MOVANT, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium; (B.V.A.); (D.D.); (E.v.B.)
- Laboratory of Physiopharmacology, GENCOR, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium; (D.N.K.); (S.V.d.B.); (M.B.)
| | - Matthias Bosman
- Laboratory of Physiopharmacology, GENCOR, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium; (D.N.K.); (S.V.d.B.); (M.B.)
| | - Emeline M. Van Craenenbroeck
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium;
- Department of Cardiology, Antwerp University Hospital (UZA), Drie Eikenstraat 655, B-2650 Edegem, Belgium
| | - Pieter-Jan Guns
- Laboratory of Physiopharmacology, GENCOR, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium; (D.N.K.); (S.V.d.B.); (M.B.)
| | - Eric van Breda
- Department of Rehabilitation Sciences & Physiotherapy, Research Group MOVANT, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium; (B.V.A.); (D.D.); (E.v.B.)
| |
Collapse
|
28
|
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
|
29
|
Kidoh M, Oda S, Sueta D, Egashira K, Hayashi H, Nakaura T, Nagayama Y, Yamamoto Y, Tsujita K, Hirai T. Serial assessment of coronary artery inflammation using cardiac CT in anthracycline chemotherapy for breast cancer. Eur Radiol 2025:10.1007/s00330-025-11347-0. [PMID: 39838089 DOI: 10.1007/s00330-025-11347-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 12/06/2024] [Accepted: 12/15/2024] [Indexed: 01/23/2025]
Abstract
OBJECTIVES There is limited evidence of the pericoronary fat attenuation index (FAI) as an imaging marker to assess cancer therapy-related cardiovascular toxicity. We aimed to measure FAI in four consecutive coronary CT angiography (CTA) scans before and 3, 6, and 12 months after anthracycline treatment in patients with breast cancer to determine trends in dynamic changes in FAI after treatment. METHODS We performed a post hoc analysis of a prospective study (between August 2019 and July 2020) in which anthracycline-induced myocardial injury was evaluated using cardiac CT. FAI was quantified using coronary CTA images before and 3, 6, and 12 months after anthracycline treatment. The FAIs of the three coronary arteries were averaged to calculate the FAI (Total). RESULTS FAI was analyzed on 14 patients with breast cancer who had adequate CT image quality (mean age, 62 years ± 11 (SD); 14 women). During the observation period, all 14 patients treated with anthracycline developed mild asymptomatic cardiac dysfunction related to cancer treatment (CTRCD). FAI (Total) showed a gradual increase during the observation period compared to baseline (baseline: -77.3 ± 5.6 HU, 3 months: -77.1 ± 4.8 HU, 6 months: -76.5 ± 5.4 HU, 12 months: -73.8 ± 5.8 HU). FAI (Total) was significantly elevated at 12 months compared to baseline (p < 0.001). CONCLUSION In patients with breast cancer, FAI showed a gradual increase at 3, 6, and 12 months after treatment with anthracyclines compared to before treatment. FAI may be used as an imaging biomarker of coronary artery inflammation in the follow-up of anthracycline therapy and may contribute to the personalization of therapy through early detection of coronary toxicity. KEY POINTS Question Is pericoronary fat attenuation index (FAI) a potential imaging biomarker for assessing changes in pericoronary adipose tissue related to cancer therapy-related cardiovascular toxicity? Findings In 14 patients with breast cancer, FAI showed a gradual increase at 3, 6, and 12 months after treatment with anthracyclines compared to before treatment. Clinical relevance FAI may be an imaging biomarker for the detection and treatment of cancer therapy-related cardiovascular toxicity.
Collapse
Affiliation(s)
- Masafumi Kidoh
- Department of Diagnostic Radiology, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan.
| | - Seitaro Oda
- Department of Diagnostic Radiology, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Daisuke Sueta
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Koichi Egashira
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Hidetaka Hayashi
- Department of Diagnostic Radiology, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Takeshi Nakaura
- Department of Diagnostic Radiology, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Yasunori Nagayama
- Department of Diagnostic Radiology, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Yutaka Yamamoto
- Department of Breast and Endocrine Surgery, Kumamoto University Hospital, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Kenichi Tsujita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Toshinori Hirai
- Department of Diagnostic Radiology, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| |
Collapse
|
30
|
Yin B, Ren J, Liu X, Zhang Y, Zuo J, Wen R, Pei H, Lu M, Zhu S, Zhang Z, Wang Z, Zhai Y, Ma Y. Astaxanthin mitigates doxorubicin-induced cardiotoxicity via inhibiting ferroptosis and autophagy: a study based on bioinformatic analysis and in vivo/ vitro experiments. Front Pharmacol 2025; 16:1524448. [PMID: 39906141 PMCID: PMC11790656 DOI: 10.3389/fphar.2025.1524448] [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: 11/07/2024] [Accepted: 01/02/2025] [Indexed: 02/06/2025] Open
Abstract
Background Doxorubicin (DOX), a widely employed chemotherapeutic agent in cancer treatment, has seen restricted use in recent years owing to its associated cardiotoxicity. Current reports indicate that doxorubicin-induced cardiotoxicity (DIC) is a complex phenomenon involving various modes of cell death. Astaxanthin (ASX), a natural carotenoid pigment, has garnered significant attention for its numerous health benefits. Recent studies have shown that ASX has a broad and effective cardiovascular protective effect. Our study aims to investigate the protective effects of ASX against DIC and elucidate its underlying mechanisms. This has substantial practical significance for the clinical application of DOX. Methods Bioinformatic analyses were conducted using transcriptomic data from the gene expression omnibus (GEO) database to identify key mechanisms underlying DIC. Network pharmacology was employed to predict the potential pathways and targets through which ASX exerts its effects on DIC. In vitro experiments, following pretreatment with ASX, H9C2 cells were exposed to DOX. Cell viability, injury and the protein expression levels associated with ferroptosis and autophagy were assessed. In the animal experiments, rats underwent 4 weeks of gavage treatment with various doses of ASX, followed by intraperitoneal injections of DOX every 2 days during the final week. Histological, serum, and protein analyses were conducted to evaluate the effects of ASX on DIC. Results The bioinformatics analysis revealed that ferroptosis and autophagy are closely associated with the development of DIC. ASX may exert an anti-DIC effect by modulating ferroptosis and autophagy. The experimental results show that ASX significantly mitigates DOX-induced myocardial tissue damage, inflammatory response, oxidative stress, and damage to H9C2 cells. Mechanistically, ASX markedly ameliorates levels of ferroptosis and autophagy both in vitro and in vivo. Specifically, ASX upregulates solute carrier family 7 member 11 (SLC7A11) and glutathione peroxidase 4 (GPX4), while downregulating the expression of transferrin receptor 1 (TFRC), ferritin heavy chain (FTH1) and ferritin light chain (FTL). Additionally, ASX enhances the expression of P62 and decreases levels of Beclin1 and microtubule-associated proteins light chain 3 (LC3). Conclusion Our results indicate that ferroptosis and autophagy are critical factors influencing the occurrence and progression of DOX-induced cardiotoxicity. ASX can alleviate DIC by inhibiting ferroptosis and autophagy.
Collapse
Affiliation(s)
- Bowen Yin
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, China
| | - Jingyi Ren
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, China
| | - Xuanyi Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, China
| | - Yadong Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, China
| | - Jinshi Zuo
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, China
| | - Rui Wen
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, China
| | - Huanting Pei
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, China
| | - Miaomiao Lu
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, China
| | - Siqi Zhu
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, China
| | - Zhenao Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, China
| | - Ziyi Wang
- Undergraduate of College of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Yanyi Zhai
- Undergraduate of College of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Yuxia Ma
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, China
| |
Collapse
|
31
|
Huang J, Hou X, Zhou N, Su N, Wei S, Yang Y, Sun T, Li G, Li W, Zhang B. Novel Protective Role for Gut Microbiota-derived Metabolite PAGln in Doxorubicin-induced Cardiotoxicity. Cardiovasc Drugs Ther 2025:10.1007/s10557-024-07665-y. [PMID: 39808382 DOI: 10.1007/s10557-024-07665-y] [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] [Accepted: 12/17/2024] [Indexed: 01/16/2025]
Abstract
PURPOSE Doxorubicin (Dox) is a classic anthracycline chemotherapy drug with cause cumulative and dose-dependent cardiotoxicity. This study aimed to investigate the potential role and molecular mechanism of phenylacetylglutamine (PAGln), a novel gut microbiota metabolite, in Dox-induced cardiotoxicity (DIC). METHODS DIC models were established in vivo and in vitro, and a series of experiments were performed to verify the cardioprotective effect of PAGln. RNA sequencing (RNA-seq) was employed to explore the mechanism of PAGln in DIC. Subsequently, the differentially expressed genes (DEGs) were subjected to comprehensive analysis using diverse public databases, and RT-PCR was used to confirm the expression levels of the candidate genes. Finally, molecular docking techniques were used for validation. RESULTS PAGln effectively prevented both in vivo and in vitro Dox-induced myocardial injury and cell apoptosis. RNA-seq results showed that 40 genes were up-regulated and 54 down-regulated in the Dox group compared to the Con group, displaying opposite changes in the Dox + PAGln group. Enrichment analysis highlighted several mechanisms by which PAGln alleviated Dox-induced cardiotoxicity, including the lipid metabolic process, calcium-mediated signaling, positive regulation of store-operated calcium channel activity, and hypertrophic cardiomyopathy. In vitro and in vivo experiments confirmed that PAGln treatment could reverse the changes in the expression levels of Klb, Ece2, Nmnat2, Casq1, Pak1, and Apob in Dox. Molecular docking results showed that these genes had good binding activity with PAGln. CONCLUSIONS PAGln shows potential in alleviating Dox-induced cardiotoxicity, with Ece2 identified as key regulatory molecules related to endothelial dysfunction.
Collapse
Affiliation(s)
- Jie Huang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, 410013, Hunan, China
| | - Xingyuan Hou
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
- Institute of Clinical Pharmacy, Central South University, Changsha, 410011, Hunan, China
| | - Ni Zhou
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
- Institute of Clinical Pharmacy, Central South University, Changsha, 410011, Hunan, China
| | - Nan Su
- Department of Ophthalmology, the First People's Hospital of Lanzhou City, Lanzhou, 730050, Gansu, China
| | - Shanshan Wei
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
- Institute of Clinical Pharmacy, Central South University, Changsha, 410011, Hunan, China
| | - Yuanying Yang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
- Institute of Clinical Pharmacy, Central South University, Changsha, 410011, Hunan, China
| | - Taoli Sun
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Guangdi Li
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, 410013, Hunan, China
| | - Wenqun Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
- Institute of Clinical Pharmacy, Central South University, Changsha, 410011, Hunan, China.
| | - Bikui Zhang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
- Institute of Clinical Pharmacy, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
32
|
Qin C, Sun M, Lv F, Du D, Li W, Liu S. Potential Mechanism by which Eriodictyol Protects against Doxorubicininduced Cardiotoxicity based on Network Pharmacology, Molecular Docking, and Molecular Dynamics Simulation. Curr Comput Aided Drug Des 2025; 21:316-332. [PMID: 38847263 DOI: 10.2174/0115734099297600240523105601] [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: 01/18/2024] [Revised: 04/16/2024] [Accepted: 04/26/2024] [Indexed: 05/28/2025]
Abstract
BACKGROUND The clinical use of doxorubicin (DOX), an anthracycline antibiotic with broad-spectrum applications against various malignant tumors, is limited by doxorubicininduced cardiotoxicity (DIC). Eriodictyol (ERD) has shown cardioprotective effects, but the mechanism of its protective effect on DIC remains unknown. AIMS This study aimed to explore the potential mechanisms by which ERD confers protection against DIC. METHODS ERD and DIC targets were identified from the TCMSP, PharmMaper, SwissTargetPrediction, TargetNet, BATMAN, GeneCards, and PharmGKB databases. Differential gene expression data between DIC and normal tissues were extracted from the GEO database. A protein‒ protein interaction (PPI) network of the intersecting ERD-DIC targets was constructed using the STRING platform and visualized with Cytoscape 3.10.0 software. Gene Ontology (GO) functional enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis for ERD-DIC cross-targets were conducted. Validation included molecular docking with AutoDock Tools software and molecular dynamics simulations with Gromacs 2019.6 software. RESULTS Network pharmacology analysis revealed 43 intersecting ERD-DIC targets, including 6 key targets. GO functional enrichment analysis indicated that the intersecting targets were enriched in 550 biological processes, 45 cell components, and 41 molecular functions. KEGG pathway enrichment analysis identified 114 enriched signaling pathways. Molecular docking revealed a strong binding affinity between ERD and 6 key targets, as well as multiple targets within the ROS pathway. Molecular dynamics simulations indicated that ERD has favorable binding with 3 crucial targets. CONCLUSION The systematic network pharmacology analysis suggests that ERD may mitigate DIC through multiple targets and pathways, with the ROS pathway potentially playing a crucial role. These findings provide a reference for foundational research and clinical applications of ERD in treating DIC.
Collapse
Affiliation(s)
- Chunmeng Qin
- Department of Pharmacy, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Mei Sun
- Department of Pharmacy, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| | - Feng Lv
- Department of Pharmacy, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| | - Dan Du
- Department of Pharmacy, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| | - Wenjun Li
- Department of Pharmacy, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| | - Songqing Liu
- Department of Pharmacy, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| |
Collapse
|
33
|
Hara A. [Anthracycline-Induced Cardiotoxicity and Exploration of Cardioprotective Drugs]. YAKUGAKU ZASSHI 2025; 145:121-132. [PMID: 39894481 DOI: 10.1248/yakushi.24-00185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Many anticancer drugs, including anthracycline drugs, pose a risk of cardiovascular damage as an adverse reaction. This can detrimentally impact the prognosis and quality of life of patients, potentially leading to the interruption of cancer chemotherapy and compromising cancer treatment. Recently, onco-cardiology (or cardio-oncology) has developed as a new interdisciplinary field that focuses on the prevention and treatment of cardiovascular toxicity of anticancer drugs. In this review, we explore the mechanism underlying the cardiotoxicity of anthracyclines and examine pharmacological agents that safeguard the heart from anthracycline-induced damage. Anthracycline-induced cardiotoxicity primarily involves oxidative stress, characterized by radical production in mitochondria and subsequent apoptosis in cardiomyocytes. While various antioxidant agents, such as resveratrol, vitamin E, and melatonin have demonstrated efficacy in reducing anthracycline-induced cardiotoxicity in animal models, their clinical effectiveness remains inconclusive. Alternatively, dexrazoxane, an intracellular iron chelator, along with standard heart failure medications, such as β-blockers, angiotensin-converting enzyme inhibitors, and angiotensin II receptor blockers, reduce anthracycline cardiotoxicity and prevent subsequent heart failure in both animal and human studies. Additionally, statins [hydroxymethylglutaryl (HMG)-CoA reductase inhibitors] and ranolazine have emerged as potential candidates for attenuating anthracycline-induced cardiotoxicity in clinical settings. Notably, recent in vitro findings suggest that everolimus, an autophagy/mitophagy-inducing antitumor drug, may protect cardiomyocytes from anthracycline-induced toxicity without reducing the antitumor effects of anthracycline. Although promising, further clinical research is warranted to validate the potential of everolimus as a safer and more effective anthracycline chemotherapeutic strategy.
Collapse
Affiliation(s)
- Akiyoshi Hara
- Department of Clinical Pharmacotherapeutics, Tohoku Medical and Pharmaceutical University
| |
Collapse
|
34
|
Tayal R, Mannan A, Singh S, Dhiman S, Singh TG. Unveiling the Complexities: Exploring Mechanisms of Anthracyclineinduced Cardiotoxicity. Curr Cardiol Rev 2025; 21:42-77. [PMID: 39484769 PMCID: PMC12060933 DOI: 10.2174/011573403x322928241021100631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 11/03/2024] Open
Abstract
The coexistence of cancer and heart disease, both prominent causes of illness and death, is further exacerbated by the detrimental impact of chemotherapy. Anthracycline-induced cardiotoxicity is an unfortunate side effect of highly effective therapy in treating different types of cancer; it presents a significant challenge for both clinicians and patients due to the considerable risk of cardiotoxicity. Despite significant progress in understanding these mechanisms, challenges persist in identifying effective preventive and therapeutic strategies, rendering it a subject of continued research even after three decades of intensive global investigation. The molecular targets and signaling pathways explored provide insights for developing targeted therapies, emphasizing the need for continued research to bridge the gap between preclinical understanding and clinical applications. This review provides a comprehensive exploration of the intricate mechanisms underlying anthracycline-induced cardiotoxicity, elucidating the interplay of various signaling pathways leading to adverse cellular events, including cardiotoxicity and death. It highlights the extensive involvement of pathways associated with oxidative stress, inflammation, apoptosis, and cellular stress responses, offering insights into potential and unexplored targets for therapeutic intervention in mitigating anthracycline-induced cardiac complications. A comprehensive understanding of the interplay between anthracyclines and these complexes signaling pathways is crucial for developing strategies to prevent or mitigate the associated cardiotoxicity. Further research is needed to outline the specific contributions of these pathways and identify potential therapeutic targets to improve the safety and efficacy of anthracycline-based cancer treatment. Ultimately, advancements in understanding anthracycline-induced cardiotoxicity mechanisms will facilitate the development of more efficacious preventive and treatment approaches, thereby improving outcomes for cancer patients undergoing anthracycline-based chemotherapy.
Collapse
Affiliation(s)
- Rohit Tayal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Shareen Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Sonia Dhiman
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | | |
Collapse
|
35
|
Szponar J, Ciechanski E, Ciechanska M, Dudka J, Mandziuk S. Evolution of Theories on Doxorubicin-Induced Late Cardiotoxicity-Role of Topoisomerase. Int J Mol Sci 2024; 25:13567. [PMID: 39769331 PMCID: PMC11678604 DOI: 10.3390/ijms252413567] [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: 11/08/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Doxorubicin (DOX) has been widely used as a cytotoxic chemotherapeutic. However, DOX has a number of side effects, such as myelotoxicity or gonadotoxicity, the most dangerous of which is cardiotoxicity. Cardiotoxicity can manifest as cardiac arrhythmias, myocarditis, and pericarditis; life-threatening late cardiotoxicity can result in heart failure months or years after the completion of chemotherapy. The development of late cardiomyopathy is not yet fully understood. The most important question is how DOX reprograms the cardiomyocyte, after which DOX is excreted from the body, initially without symptoms. However, clinically overt cardiomyopathy develops over the following months and years. Since the 1980s, DOX-induced disorders in cardiomyocytes have been thought to be related to oxidative stress and dependent on the Fe/reactive oxygen species (ROS) mechanism. That line of evidence was supported by dexrazoxane (DEX) protection, the only Food and Drug Administration (FDA)-approved drug for preventing DOX-induced cardiomyopathy, which complexes iron. Thus, the hypothesis related to Fe/ROS provides a plausible explanation for the induction of the development of late cardiomyopathy via DOX. However, in subsequent studies, DEX was used to identify another important mechanism in DOX-induced cardiomyopathy that is related to topoisomerase 2β (Top2β). Does the Top2β hypothesis explain the mechanisms of the development of DOX-dependent late heart failure? Several of these mechanisms have been identified to date, proving the involvement of Top2β in the regulation of the redox balance, including oxidative stress. Thus, the development of late cardiomyopathy can be explained based on mechanisms related to Top2β. In this review, we highlight free radical theory, iron imbalance, calcium overload, and finally, a theory based on Top2β.
Collapse
Affiliation(s)
- Jaroslaw Szponar
- Toxicology Clinic, Faculty of Medicine, Medical University of Lublin, Krasnicka 100, 20-718 Lublin, Poland;
- Clinical Department of Toxicology and Cardiology, Regional Specialist Hospital, Krasnicka 100, 20-718 Lublin, Poland
| | - Erwin Ciechanski
- Department of Cardiology, Regional Specialist Hospital, Krasnicka 100, 20-718 Lublin, Poland
| | - Magda Ciechanska
- Department of Pulmonary Diseases and Children Rheumatology, Medical University of Lublin, Antoniego Gebali 6, 20-093 Lublin, Poland
| | - Jaroslaw Dudka
- Department of Toxicology, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland;
| | - Sławomir Mandziuk
- Department of Pneumology, Oncology and Allergology, Medical University of Lublin, Jaczewskiego 8, 20-090 Lublin, Poland;
| |
Collapse
|
36
|
Qiu H, Zhou W, Huang Q, Lin H, Zhou Y, Wu C, Huang Y, Leng J. Role of Cancer History in Cardiovascular Mortality Among Different Age-group Patients With Differentiated Thyroid Cancer. J Endocr Soc 2024; 9:bvae213. [PMID: 39669653 PMCID: PMC11635452 DOI: 10.1210/jendso/bvae213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Indexed: 12/14/2024] Open
Abstract
Background Cardiovascular disease (CVD) is the leading cause of noncancer-related mortality among differentiated thyroid cancer (DTC) survivors, which accounts for a large portion of subsequent primary malignancies in childhood cancer survivors. This study aims to assess the risk of cardiovascular mortality among DTC as a second primary malignancy (DTC-2) patients compared with DTC as a first primary malignancy (DTC-1) and the general population. Methods Using the Surveillance, Epidemiology, and End Results database, we conducted a population-based cohort study including 159 395 DTC-1 and 20 010 DTC-2 patients diagnosed older than 30 between 1975 and 2020 and the corresponding US population (71 214 642 person-years; 41 420 893 cardiovascular deaths). Compared with general-population and DTC-1 patients, we calculated incidence rate ratios (IRRs) of cardiovascular deaths among DTC-2 patients using Poisson regression. To adjust for unmeasured confounders, we performed a nested, case-crossover analysis among DTC-2 patients who died from CVD. Results Although DTC-2 patients had a comparable risk compared with the population (IRR 1.01) and a mildly increased risk of cardiovascular mortality compared with DTC-1 patients (IRR 1.26), the association was pronounced among individuals aged 30 to 74 years, especially 30 to 44 years (DTC-2 vs population: IRR 8.89; DTC-2 vs DTC-1: IRR 3.00). The risk elevation was greatest within the first month after diagnosis, compared with the population. The case-crossover analysis confirmed these results. Conclusion DTC-2 patients are at increased risk of cardiovascular mortality. Clinicians should carefully monitor CVD and manage other CVD-related factors, such as exogenous thyroxine and emotional distress, for DTC-2 patients, especially for those under 75 years. Novelty and Impact Statements This study is the first comprehensive investigation into the cardiovascular mortality of DTC-2, revealing a higher risk compared to DTC-1 and the general population, especially for cases between 30 and 74 years old. The risk elevation was greatest within the first month after diagnosis. These findings emphasize the restriction of thyroid hormone suppression therapy and reinforce stress management to prevent premature DTC-2 patients from cardiovascular death.
Collapse
Affiliation(s)
- Hongrui Qiu
- Department of Thoracic Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080 Guangdong, China
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080 Guangdong, China
| | - Wenyi Zhou
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 Guangdong, China
- Yat-Sen Breast Tumor Hospital, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 Guangdong, China
| | - Qizhi Huang
- Department of Thoracic Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080 Guangdong, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080 Guangdong, China
| | - Hongwei Lin
- Department of Thoracic Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080 Guangdong, China
- The First School of Clinical Medicine, Guangdong Medical University, Zhanjiang, 524023 Guangdong, China
| | - Yubo Zhou
- School of Chinese Medicine, Faculty of Medicine, Macau University of Science and Technology, 999078 Macao, China
| | - Chaodong Wu
- The First Clinical Medical School, Southern Medical University, Guangzhou, 510515 Guangdong, China
| | - Yijie Huang
- Department of General Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080 Guangdong, China
| | - Jinhang Leng
- Department of Thoracic Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080 Guangdong, China
| |
Collapse
|
37
|
Gao H, Chen Z, Yao Y, He Y, Hu X. Common biological processes and mutual crosstalk mechanisms between cardiovascular disease and cancer. Front Oncol 2024; 14:1453090. [PMID: 39634266 PMCID: PMC11614734 DOI: 10.3389/fonc.2024.1453090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 10/31/2024] [Indexed: 12/07/2024] Open
Abstract
Cancer and cardiovascular disease (CVD) are leading causes of mortality and thus represent major health challenges worldwide. Clinical data suggest that cancer patients have an increased likelihood of developing cardiovascular disease, while epidemiologic studies have shown that patients with cardiovascular disease are also more likely to develop cancer. These observations underscore the increasing importance of studies exploring the mechanisms underlying the interaction between the two diseases. We review their common physiological processes and potential pathophysiological links. We explore the effects of chronic inflammation, oxidative stress, and disorders of fatty acid metabolism in CVD and cancer, and also provide insights into how cancer and its treatments affect heart health, as well as present recent advances in reverse cardio-oncology using a new classification approach.
Collapse
Affiliation(s)
- Hanwei Gao
- Department of Cardiology, China–Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| | - Zhongyu Chen
- Department of Cardiology, China–Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin, China
- CJUH-JLU-China iGEM Team, Jilin University, Changchun, Jilin, China
| | - Yutong Yao
- Department of Cardiology, China–Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin, China
- CJUH-JLU-China iGEM Team, Jilin University, Changchun, Jilin, China
| | - Yuquan He
- Department of Cardiology, China–Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin, China
- CJUH-JLU-China iGEM Team, Jilin University, Changchun, Jilin, China
| | - Xin Hu
- Department of Cardiology, China–Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin, China
- CJUH-JLU-China iGEM Team, Jilin University, Changchun, Jilin, China
| |
Collapse
|
38
|
Cortés-Salgado A, Serrano JJ, Cordero Pereda D, Menacho M, Del Rey JM, Del Campo-Albendea L, Saavedra C, Chamorro J, Rosero D, Sotoca P, Guillén-Ponce C, Guerra E, Fernández-Abad M, López-Miranda E, Martínez-Jáñez N, Gion M, Salazar MT, Agudo-Quílez P, Garrido P, Alonso Salinas GL. Anthracyclines-induced cardiotoxicity in patients with early breast cancer carrying germline BRCA1/2 mutation: the BRCAN study. Oncologist 2024:oyae299. [PMID: 39561108 DOI: 10.1093/oncolo/oyae299] [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: 03/11/2024] [Accepted: 10/03/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND BRCA1/2 genes play a critical role in genome stability and DNA repair. In animal models, loss of cardiomyocyte-specific BRCA1/2 is associated with DNA damage, apoptosis, cardiac dysfunction, and mortality following anthracycline exposure. However, whether these preclinical findings translate to humans remains unclear. OBJECTIVE Assess the impact of germline BRCA1/2 (gBRCA1/2) status on anthracyclines-induced cardiotoxicity (AIC) in patients with early breast cancer and no prior anti-HER2 therapy. METHODS This single-center retrospective/prospective cohort study focused on early breast cancer patients, treated with anthracycline-based chemotherapy in the neo/adjuvant setting, no prior anti-HER2 therapy, and known gBRCA1/2 status, normal baseline left ventricular ejection fraction (LVEF), and no previous cardiovascular disease. Follow-up assessments involved myocardial dysfunction blood biomarkers (MDBB), transthoracic echocardiography (TTE), and quality of life (QoL) questionnaires. The primary objective was LVEF changes comparing BRCA1/2 mutation carriers (gBRCA1/2m) vs non-carriers (gBRCA1/2wt). Secondary objectives included differences in MDBB and QoL. RESULTS A total of 137 patients were included (103 gBRCA1/2wt and 34 gBRCA1/2m). Baseline characteristics were similar between groups. Compared to baseline, LVEF% reduction was -4.7[-12.0, 0.0] vs -9.5[-18.0, -5.0] in gBRCA1/2wt vs gBRCA1/2m, (P = .027). After adjusting for confounders, the difference in reduction in LVEF remained statistically significant at -4.5 [95%CI, -8.6, -0.4; P = .032]. No differences between MDBB (C-reactive protein, hsTnI, NT-proBNP, D-Dimer, ST-2, or Galectine-3) or QoL (MLHFQ and EQ5-D index) were detected. CONCLUSIONS gBRCA1/2m patients could represent a higher-risk population for AIC. gBRCA1/2 status should be one of the factors to consider in deciding on adjuvant anthracycline necessity. This population could benefit from a cardio-oncology closer follow-up and cardioprotective strategies.
Collapse
Affiliation(s)
- Alfonso Cortés-Salgado
- Medical Oncology Department, Hospital Universitario Ramón y Cajal (IRYCIS), 28034 Madrid, Spain
| | - Juan José Serrano
- Medical Oncology Department, Grupo Vithas Madrid, 28043 Madrid, Spain
| | - David Cordero Pereda
- Cardiology Department, Hospital Universitario Ramón y Cajal (IRYCIS), 28034 Madrid, Spain
| | - Miriam Menacho
- Clinical Biochemistry Department, Hospital Universitario Ramón y Cajal (IRYCIS), 28034 Madrid, Spain
| | - José Manuel Del Rey
- Clinical Biochemistry Department, Hospital Universitario Ramón y Cajal (IRYCIS), 28034 Madrid, Spain
| | - Laura Del Campo-Albendea
- Biostatistics Unit, Hospital Universitario Ramón y Cajal (IRYCIS). CIBERESP, ISCIII, 28029 Madrid, Spain
| | - Cristina Saavedra
- Medical Oncology Department, Hospital Universitario Ramón y Cajal (IRYCIS), 28034 Madrid, Spain
| | - Jesús Chamorro
- Medical Oncology Department, Hospital Universitario Ramón y Cajal (IRYCIS), 28034 Madrid, Spain
| | - Diana Rosero
- Medical Oncology Department, Hospital Universitario Ramón y Cajal (IRYCIS), 28034 Madrid, Spain
| | - Pilar Sotoca
- Medical Oncology Department, Hospital Universitario Ramón y Cajal (IRYCIS), 28034 Madrid, Spain
| | - Carmen Guillén-Ponce
- Medical Oncology Department, Hospital Universitario Ramón y Cajal (IRYCIS), 28034 Madrid, Spain
| | - Eva Guerra
- Medical Oncology Department, Hospital Universitario Ramón y Cajal (IRYCIS), 28034 Madrid, Spain
| | - María Fernández-Abad
- Medical Oncology Department, Hospital Universitario Ramón y Cajal (IRYCIS), 28034 Madrid, Spain
| | - Elena López-Miranda
- Medical Oncology Department, Hospital Universitario Ramón y Cajal (IRYCIS), 28034 Madrid, Spain
| | - Noelia Martínez-Jáñez
- Medical Oncology Department, Hospital Universitario Ramón y Cajal (IRYCIS), 28034 Madrid, Spain
| | - María Gion
- Medical Oncology Department, Hospital Universitario Ramón y Cajal (IRYCIS), 28034 Madrid, Spain
| | - María Teresa Salazar
- Medical Oncology Department, Hospital Universitario Ramón y Cajal (IRYCIS), 28034 Madrid, Spain
| | - Pilar Agudo-Quílez
- Cardiology Department, Hospital Universitario Ramón y Cajal (IRYCIS), 28034 Madrid, Spain
| | - Pilar Garrido
- Medical Oncology Department, Hospital Universitario Ramón y Cajal (IRYCIS), 28034 Madrid, Spain
| | - Gonzalo Luis Alonso Salinas
- Cardiology Department, Hospital Universitario de Navarra, 31008 Pamplona (Navarra), Spain
- Navarrabiomed, IdiSNA, 31008 Pamplona (Navarra), Spain
- Department of Health Sciences, Universidad Pública de Navarra (UPNA), 31008 Pamplona (Navarra), Spain
| |
Collapse
|
39
|
Botelho LFB, de Melo MDT, de Almeida ALC, Salemi VMC. Accuracy of mitral annular plane systolic excursion in diagnosing anthracycline-induced subclinical cardiotoxicity in patients with breast cancer - a retrospective cohort study. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:76. [PMID: 39497222 PMCID: PMC11533289 DOI: 10.1186/s40959-024-00280-8] [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: 04/29/2024] [Accepted: 10/28/2024] [Indexed: 11/07/2024]
Abstract
BACKGROUND The mitral annular plane systolic excursion (MAPSE) is used to analyze the left ventricle longitudinal function. However, the accuracy of MAPSE in diagnosing oncological populations is unclear. In this study, we aimed to assess the accuracy of MAPSE in diagnosing subclinical cardiotoxicity in women with breast cancer undergoing anthracycline treatment. METHODS This retrospective cohort study included echocardiographic assessments of patients with breast cancer who underwent anthracycline treatment as part of their therapeutic regimen. Assessments were performed before treatment, after administering the first dose of anthracycline, after completing anthracycline treatment, and 6 and 12 months after treatment. Left ventricular ejection fraction was calculated using the modified biplane Simpson method. The performances of MAPSE and global longitudinal strain (GLS) were analyzed using receiver operating characteristic (ROC) curves. Their accuracies were measured using the area under the ROC curves. RESULTS Sixty-one patients were included in this study. Of them, 8.2% presented cardiotoxicity 6 months after treatment completion. Patients with cardiotoxicity had lower LVEF (47% vs. 63%; p < 0.001), MAPSE (10.23 mm vs. 12.25 mm; p = 0.012), and LV GLS (16.13% vs. 19.05%; p = 0.005) values than those without. A 12% reduction in the GLS exhibited sensitivity, specificity, and overall accuracy of 80%, 70%, and 78%, respectively. A relative reduction of 15% in MAPSE exhibited a sensitivity, specificity, and accuracy of 80%, 77%, and 81.2%, respectively. An absolute MAPSE reduction of 2 mm exhibited a sensitivity, specificity, and accuracy of 80%, 73.21%, and 81.2%, respectively. No differences were observed between the ROC curves. CONCLUSION MAPSE showed similar accuracy to GLS in diagnosing subclinical cardiotoxicity in women with breast cancer undergoing anthracycline treatment.
Collapse
Affiliation(s)
- Luís Fábio Barbosa Botelho
- Department of Internal Medicine, Federal University of Paraiba, Rua da Aurora 333, Ap 904, João Pessoa, 58043-900, João Pessoa, PB, Brazil.
| | - Marcelo Dantas Tavares de Melo
- Department of Internal Medicine, Federal University of Paraiba, Rua da Aurora 333, Ap 904, João Pessoa, 58043-900, João Pessoa, PB, Brazil
| | | | - Vera Maria Cury Salemi
- Heart Institute (InCor), School of Medicine, University of São Paulo, São Paulo, Brazil
- Hospital Sírio Libanês, São Paulo, Brazil
| |
Collapse
|
40
|
Zeng X, Zhang H, Xu T, Mei X, Wang X, Yang Q, Luo Z, Zeng Q, Xu D, Ren H. Vericiguat attenuates doxorubicin-induced cardiotoxicity through the PRKG1/PINK1/STING axis. Transl Res 2024; 273:90-103. [PMID: 39059761 DOI: 10.1016/j.trsl.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/18/2024] [Accepted: 07/14/2024] [Indexed: 07/28/2024]
Abstract
Doxorubicin (DOX) is restricted due to its severe cardiotoxicity. There is still a lack of viable and effective drugs to prevent or treat DOX-induced cardiotoxicity(DIC). Vericiguat is widely used to treat heart failure with reduced ejection fraction. However, it is not clear whether vericiguat can improve DIC. In the present study, we constructed a DIC model using mice and neonatal rat cardiomyocytes and found that vericiguat ameliorated DOX-induced cardiac insufficiency in mice, restored DOX-induced mitochondrial dysfunction in neonatal rat cardiomyocytes, and inhibited the expression of inflammatory factors. Further studies showed that vericiguat improved mitochondrial dysfunction and reduced mtDNA leakage into the cytoplasm by up-regulating PRKG1, which activated PINK1 and then inhibited the STING/IRF3 pathway to alleviate DIC. These findings demonstrate for the first time that vericiguat has therapeutic potential for the treatment of DIC.
Collapse
Affiliation(s)
- Xianghui Zeng
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Key Laboratory For Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, China; Department of Cardiology, Ganzhou Hospital of Traditional Chinese Medicine, Ganzhou, Jiangxi, China
| | - Hao Zhang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Key Laboratory For Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, China
| | - Tianyu Xu
- NHC Key Laboratory of Assisted Circulation, Department of Cardiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiyuan Mei
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Key Laboratory For Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, China
| | - Xiao Wang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Key Laboratory For Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, China
| | - Qiling Yang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Key Laboratory For Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, China
| | - Zhen Luo
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Key Laboratory For Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, China
| | - Qingchun Zeng
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Key Laboratory For Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, China
| | - Dingli Xu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Key Laboratory For Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, China.
| | - Hao Ren
- Key Laboratory For Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, China; Department of Rheumatology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
41
|
Monte MG, Tonon CR, Fujimori AS, Ribeiro APD, Zanati SG, Okoshi K, Camacho CRC, Moretto MR, de Paiva SAR, Zornoff LAM, Azevedo PS, Minicucci MF, Polegato BF. Omega-3 supplementation attenuates doxorubicin-induced cardiotoxicity but is not related to the ceramide pathway. Food Sci Nutr 2024; 12:9198-9211. [PMID: 39620015 PMCID: PMC11606900 DOI: 10.1002/fsn3.4492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 09/06/2024] [Accepted: 09/13/2024] [Indexed: 01/06/2025] Open
Abstract
Cardiotoxicity is the serious side effect of doxorubicin treatment. Ceramides are formed from the degradation of sphingolipids in cell membranes and play an important role in signaling and modulating biological processes. There is evidence that omega-3 fatty acid administration can act on this pathway. To evaluate the role of the ceramide pathway in the pathophysiology of doxorubicin-induced cardiotoxicity and the effect of omega-3 fatty acid supplementation in the attenuation of chronic doxorubicin-induced cardiotoxicity in rats. Sixty male Wistar rats were divided into four groups: Control (C), Doxorubicin (D), Omega-3 fatty acids (W), and Doxorubicin + Omega-3 fatty acids (DW). The groups received omega-3 fatty acids (400 mg/kg/day, via gavage) or water for 6 weeks and doxorubicin (3.5 mg/kg, intraperitoneal) or saline once a week for 4 weeks. Doxorubicin-treated animals showed increases in left atrium and left ventricle diameters, serum triglycerides and cholesterol, malondialdehyde, and protein carbonylation. We also observed a decrease in left ventricular shortening fraction and nSMase1 expression in the heart. Omega-3 fatty acid supplementation attenuated the structural and functional alterations caused by doxorubicin and decreased protein carbonylation. In contrast to doxorubicin, omega-3 fatty acids increased neutral nSMase activity in animals that both received and did not receive doxorubicin but with no effect on nSMase1 protein expression. Omega-3 fatty acid supplementation attenuated the cardiotoxicity caused by doxorubicin. The ceramide pathway may be involved in the pathophysiology of cardiotoxicity, but it is not the mechanism by which omega-3 fatty acids attenuated cardiac dysfunction.
Collapse
Affiliation(s)
- Marina Gaiato Monte
- Department of Internal MedicineBotucatu Medical School, São Paulo State University‐UNESPBotucatuBrazil
| | - Carolina Rodrigues Tonon
- Department of Internal MedicineBotucatu Medical School, São Paulo State University‐UNESPBotucatuBrazil
| | - Anderson Seiji Fujimori
- Department of Internal MedicineBotucatu Medical School, São Paulo State University‐UNESPBotucatuBrazil
| | - Ana Paula Dantas Ribeiro
- Department of Internal MedicineBotucatu Medical School, São Paulo State University‐UNESPBotucatuBrazil
| | - Silmeia Garcia Zanati
- Department of Internal MedicineBotucatu Medical School, São Paulo State University‐UNESPBotucatuBrazil
| | - Katashi Okoshi
- Department of Internal MedicineBotucatu Medical School, São Paulo State University‐UNESPBotucatuBrazil
| | | | - Maria Regina Moretto
- Experimental Research UnitBotucatu Medical School, São Paulo State University‐UNESPBotucatuBrazil
| | | | | | - Paula Schmidt Azevedo
- Department of Internal MedicineBotucatu Medical School, São Paulo State University‐UNESPBotucatuBrazil
| | - Marcos Ferreira Minicucci
- Department of Internal MedicineBotucatu Medical School, São Paulo State University‐UNESPBotucatuBrazil
| | - Bertha Furlan Polegato
- Department of Internal MedicineBotucatu Medical School, São Paulo State University‐UNESPBotucatuBrazil
| |
Collapse
|
42
|
Wang C, Fan P, Wang Q. Evolving therapeutics and ensuing cardiotoxicities in triple-negative breast cancer. Cancer Treat Rev 2024; 130:102819. [PMID: 39216183 DOI: 10.1016/j.ctrv.2024.102819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/18/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024]
Abstract
Defined as scarce expression of hormone receptors and human epidermal growth factor receptor 2, triple-negative breast cancer (TNBC) is labeled as the most heterogeneous subtype of breast cancer with poorest prognosis. Despite rapid advancements in precise subtyping and tailored therapeutics, the ensuing cancer therapy-related cardiovascular toxicity (CTR-CVT) could exert detrimental impacts to TNBC survivors. Nowadays, this interdisciplinary issue is incrementally concerned by cardiologists, oncologists and other pertinent experts, propelling cardio-oncology as a booming field focusing on the whole-course management of cancer patients with potential cardiovascular threats. Here in this review, we initially profile the evolving molecular subtyping and therapeutic landscape of TNBC. Further, we introduce various monitoring approaches of CTR-CVT. In the main body, we elaborate on typical cardiotoxicities ensuing anti-TNBC treatments in detail, ranging from chemotherapy (especially anthracyclines), surgery, anesthetics, radiotherapy to immunotherapy, with future perspectives on promising directions in the era of artificial intelligence and traditional Chinese medicine.
Collapse
Affiliation(s)
- Chongyu Wang
- Department of Medicine, Xinglin College, Nantong University, Nantong 226007, Jiangsu, China
| | - Pinchao Fan
- The First Clinical Medical College, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Sir Run Run Hospital, Nanjing Medical University, Nanjing 211112, Jiangsu, China
| | - Qingqing Wang
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, Jiangsu, China.
| |
Collapse
|
43
|
Wasim R, Singh A, Islam A, Mohammed S, Anwar A, Mahmood T. High Mobility Group Box 1 and Cardiovascular Diseases: Study of Act and Connect. Cardiovasc Toxicol 2024; 24:1268-1286. [PMID: 39242448 DOI: 10.1007/s12012-024-09919-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 09/02/2024] [Indexed: 09/09/2024]
Abstract
Cardiovascular disease is the deadly disease that can result in sudden death, and inflammation plays an important role in its onset and progression. High mobility group box 1 (HMGB1) is a nuclear protein that regulates transcription, DNA replication, repair, and nucleosome assembly. HMGB1 is released passively by necrotic tissues and actively secreted by stressed cells. Extracellular HMGB1 functions as a damage associated molecular patterns molecule, producing numerous redox forms that induce a range of cellular responses by binding to distinct receptors and interactors, including tissue inflammation and regeneration. Extracellular HMGB1 inhibition reduces inflammation and is protective in experimental models of myocardial ischemia/reperfusion damage, myocarditis, cardiomyopathies caused by mechanical stress, diabetes, bacterial infection, or chemotherapeutic drugs. HMGB1 administration following a myocardial infarction followed by permanent coronary artery ligation improves cardiac function by stimulating tissue regeneration. HMGB1 inhibits contractility and produces hypertrophy and death in cardiomyocytes, while also stimulating cardiac fibroblast activity and promoting cardiac stem cell proliferation and differentiation. Maintaining normal nuclear HMGB1 levels, interestingly, protects cardiomyocytes from apoptosis by limiting DNA oxidative stress, and mice with HMGB1cardiomyocyte-specific overexpression are partially protected from cardiac injury. Finally, elevated levels of circulating HMGB1 have been linked to human heart disease. As a result, following cardiac damage, HMGB1 elicits both detrimental and helpful responses, which may be due to the formation and stability of the various redox forms, the particular activities of which in this context are mostly unknown. This review covers recent findings in HMGB1 biology and cardiac dysfunction.
Collapse
Affiliation(s)
- Rufaida Wasim
- Department of Pharmacy, Integral University, Lucknow, 226026, India.
- Faculty of Pharmacy, Integral University, Lucknow, 226026, India.
| | - Aditya Singh
- Department of Pharmacy, Integral University, Lucknow, 226026, India
| | - Anas Islam
- Department of Pharmacy, Integral University, Lucknow, 226026, India
| | - Saad Mohammed
- Department of Pharmacy, Integral University, Lucknow, 226026, India
| | - Aamir Anwar
- Department of Pharmacy, Integral University, Lucknow, 226026, India
| | - Tarique Mahmood
- Department of Pharmacy, Integral University, Lucknow, 226026, India
| |
Collapse
|
44
|
Nevins S, McLoughlin CD, Oliveros A, Stein JB, Rashid MA, Hou Y, Jang MH, Lee KB. Nanotechnology Approaches for Prevention and Treatment of Chemotherapy-Induced Neurotoxicity, Neuropathy, and Cardiomyopathy in Breast and Ovarian Cancer Survivors. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2300744. [PMID: 37058079 PMCID: PMC10576016 DOI: 10.1002/smll.202300744] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/05/2023] [Indexed: 06/19/2023]
Abstract
Nanotechnology has emerged as a promising approach for the targeted delivery of therapeutic agents while improving their efficacy and safety. As a result, nanomaterial development for the selective targeting of cancers, with the possibility of treating off-target, detrimental sequelae caused by chemotherapy, is an important area of research. Breast and ovarian cancer are among the most common cancer types in women, and chemotherapy is an essential treatment modality for these diseases. However, chemotherapy-induced neurotoxicity, neuropathy, and cardiomyopathy are common side effects that can affect breast and ovarian cancer survivors quality of life. Therefore, there is an urgent need to develop effective prevention and treatment strategies for these adverse effects. Nanoparticles (NPs) have extreme potential for enhancing therapeutic efficacy but require continued research to elucidate beneficial interventions for women cancer survivors. In short, nanotechnology-based approaches have emerged as promising strategies for preventing and treating chemotherapy-induced neurotoxicity, neuropathy, and cardiomyopathy. NP-based drug delivery systems and therapeutics have shown potential for reducing the side effects of chemotherapeutics while improving drug efficacy. In this article, the latest nanotechnology approaches and their potential for the prevention and treatment of chemotherapy-induced neurotoxicity, neuropathy, and cardiomyopathy in breast and ovarian cancer survivors are discussed.
Collapse
Affiliation(s)
- Sarah Nevins
- Department of Chemistry and Chemical Biology, Rutgers
University, the State University of New Jersey, 123 Bevier Road, Piscataway, NJ
08854, U.S.A
| | - Callan D. McLoughlin
- Department of Chemistry and Chemical Biology, Rutgers
University, the State University of New Jersey, 123 Bevier Road, Piscataway, NJ
08854, U.S.A
| | - Alfredo Oliveros
- Department of Neurosurgery, Robert Wood Johnson Medical
School, Rutgers University, the State University of New Jersey, 661 Hoes Ln W,
Piscataway, NJ, 08854, U.S.A
| | - Joshua B. Stein
- Department of Chemistry and Chemical Biology, Rutgers
University, the State University of New Jersey, 123 Bevier Road, Piscataway, NJ
08854, U.S.A
| | - Mohammad Abdur Rashid
- Department of Neurosurgery, Robert Wood Johnson Medical
School, Rutgers University, the State University of New Jersey, 661 Hoes Ln W,
Piscataway, NJ, 08854, U.S.A
| | - Yannan Hou
- Department of Chemistry and Chemical Biology, Rutgers
University, the State University of New Jersey, 123 Bevier Road, Piscataway, NJ
08854, U.S.A
| | - Mi-Hyeon Jang
- Department of Neurosurgery, Robert Wood Johnson Medical
School, Rutgers University, the State University of New Jersey, 661 Hoes Ln W,
Piscataway, NJ, 08854, U.S.A
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers
University, the State University of New Jersey, 123 Bevier Road, Piscataway, NJ
08854, U.S.A
| |
Collapse
|
45
|
Alizadehasl A, Malekzadeh Moghani M, Mirzaei H, Keshvari M, Fadaei F, Cramer H, Pasalar M, Heydarirad G. Cardioprotective Diet to Prevent Anthracycline-Induced Cardiotoxicity in Patients with Breast Cancer: A Randomized Open-Label Controlled Trial. JOURNAL OF INTEGRATIVE AND COMPLEMENTARY MEDICINE 2024; 30:995-1001. [PMID: 38757731 DOI: 10.1089/jicm.2023.0777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
Objectives: Several studies have indicated that dietary interventions may offer protection against the development of cardiac damage in the case of anthracycline-induced cardiomyopathy (AIC). The goal of this study was to assess whether an evidence-based cardioprotective diet can be effective in preventing AIC in patients with breast cancer. Design: Randomized, open-label, controlled trial. The study period was set for 18 weeks, and the data were analyzed by generalized estimating equation modeling and one-way repeated measures analysis of variance. Setting/Location: Shahid Rajaie Hospital affiliated (Tehran, Iran). Subjects: Fifty anthracycline-treated patients with breast cancer. Interventions: Patients were randomized to receive either a 2-hour training in evidence-based cardio-protective diet or Carvedilol 6.25 mg bid. Outcome Measures: The primary outcome was the number of patients with abnormal left ventricular ejection fraction (LVEF) after 18 weeks. Results: At week 18, 12 (48%) out of 25 participants in the cardioprotective diet group had abnormal LVEF in comparison with 21 (84%) out of 25 in the carvedilol group (p = 0.007). Also, 2 (8%) out of 25 in the cardioprotective diet group compared with 7 (28%) out of 25 participants in the carvedilol group had abnormal global longitudinal strain (p = 0.066). The diet group showed significant improvements in the quality-of-life dimensions named "health change" and "general health" compared with the carvedilol group using the Short Form-36 Health Survey questionnaire. Conclusions: This study suggests that an evidence-based cardioprotective diet can contribute to the prevention of AIC. Although current treatments for AIC can be effective, further research is mandatory for more options.
Collapse
Affiliation(s)
- Azin Alizadehasl
- Cardio-Oncology Research Center, Rajaie Cardiovascular Medical & Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mona Malekzadeh Moghani
- Shohada-e Tajrish Hospital, Department of RadioOncology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamidreza Mirzaei
- Shohada-e Tajrish Hospital, Department of RadioOncology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cancer Research Center, Shohadae Tajrish Hospital, Department of Radiation Oncology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoumeh Keshvari
- Clinical Research Development Center, Mahdiyeh Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Fadaei
- Department of Traditional Medicine, School of Traditional Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Holger Cramer
- Institute for General Practice and Interprofessional Care, Tuebingen, University Hospital Tuebingen, Tuebingen, Germany
| | - Mehdi Pasalar
- Research Center for Traditional Medicine and History of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ghazaleh Heydarirad
- Department of Traditional Medicine, School of Traditional Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
46
|
Bhasin V, Vakilpour A, Scherrer-Crosbie M. Statins for the Primary Prevention of Anthracycline Cardiotoxicity: A Comprehensive Review. Curr Oncol Rep 2024; 26:1197-1204. [PMID: 39002055 PMCID: PMC11480194 DOI: 10.1007/s11912-024-01579-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/02/2024] [Indexed: 07/15/2024]
Abstract
PURPOSE OF REVIEW The aim of this review is two-fold: (1) To examine the mechanisms by which statins may protect from anthracycline-induced cardiotoxicity and (2) To provide a comprehensive overview of the existing clinical literature investigating the role of statins for the primary prevention of anthracycline-induced cardiotoxicity. RECENT FINDINGS The underlying cardioprotective mechanisms associated with statins have not been fully elucidated. Key mechanisms related to the inhibition of Ras homologous (Rho) GTPases have been proposed. Data from observational studies has supported the beneficial role of statins for the primary prevention of anthracycline-induced cardiotoxicity. Recently, several randomized controlled trials investigating the role of statins for the primary prevention of anthracycline-induced cardiotoxicity have produced contrasting results. Statins have been associated with a lower risk of cardiac dysfunction in cancer patients receiving anthracyclines. Further investigation with larger randomized control trials and longer follow-up periods are needed to better evaluate the long-term role of statin therapy and identify the subgroups who benefit most from statin therapy.
Collapse
Affiliation(s)
- Varun Bhasin
- Division of Cardiovascular Medicine and Thalheimer Center for Cardio-Oncology, Perelman Center for Advanced Medicine and Hospital of the University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA, USA
| | - Azin Vakilpour
- Division of Cardiovascular Medicine and Thalheimer Center for Cardio-Oncology, Perelman Center for Advanced Medicine and Hospital of the University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA, USA
| | - Marielle Scherrer-Crosbie
- Division of Cardiovascular Medicine and Thalheimer Center for Cardio-Oncology, Perelman Center for Advanced Medicine and Hospital of the University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA, USA.
| |
Collapse
|
47
|
Wang SY, Wang YJ, Dong MQ, Li GR. Acacetin is a Promising Drug Candidate for Cardiovascular Diseases. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:1661-1692. [PMID: 39347953 DOI: 10.1142/s0192415x24500654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Phytochemical flavonoids have been proven to be effective in treating various disorders, including cardiovascular diseases. Acacetin is a natural flavone with diverse pharmacological effects, uniquely including atrial-selective anti-atrial fibrillation (AF) via the inhibition of the atrial specific potassium channel currents [Formula: see text] (ultra-rapidly delayed rectifier potassium current), [Formula: see text] (acetylcholine-activated potassium current), [Formula: see text] (calcium-activated small conductance potassium current), and [Formula: see text] (transient outward potassium current). [Formula: see text] inhibition by acacetin, notably, suppresses experimental J-wave syndromes. In addition, acacetin provides extensive cardiovascular protection against ischemia/reperfusion injury, cardiomyopathies/heart failure, autoimmune myocarditis, pulmonary artery hypertension, vascular remodeling, and atherosclerosis by restoring the downregulated intracellular signaling pathway of Sirt1/AMPK/PGC-1α followed by increasing Nrf2/HO-1/SOD thereby inhibiting oxidation, inflammation, and apoptosis. This review provides an integrated insight into the capabilities of acacetin as a drug candidate for treating cardiovascular diseases, especially atrial fibrillation and cardiomyopathies/heart failure.
Collapse
Affiliation(s)
- Shu-Ya Wang
- Geriatric Diseases Institute of Chengdu, Center for Medicine Research and Translation, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu 611137, P. R. China
| | - Ya-Jing Wang
- Department of Pharmacy, School of Pharmacy, Changzhou University Changzhou, Jiangsu 213164, P. R. China
- Nanjing Amazigh Pharma Limited, Nanjing, Jiangsu 210032, P. R. China
| | - Ming-Qing Dong
- Geriatric Diseases Institute of Chengdu, Center for Medicine Research and Translation, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu 611137, P. R. China
| | - Gui-Rong Li
- Nanjing Amazigh Pharma Limited, Nanjing, Jiangsu 210032, P. R. China
| |
Collapse
|
48
|
Chen G, Yao Y, Liu Y, Zhang R, Wen C, Zhou Q, Xu Y, Wang W, Jiang H, Tao Z, Chen W, Qiu Z, Chen X. IKKα-STAT3-S727 axis: a novel mechanism in DOX-induced cardiomyopathy. Cell Mol Life Sci 2024; 81:406. [PMID: 39287798 PMCID: PMC11408453 DOI: 10.1007/s00018-024-05439-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 08/19/2024] [Accepted: 09/04/2024] [Indexed: 09/19/2024]
Abstract
Doxorubicin (DOX) is an effective chemotherapeutic drug, but its use can lead to cardiomyopathy, which is the leading cause of mortality among cancer patients. Macrophages play a role in DOX-induced cardiomyopathy (DCM), but the mechanisms undlerlying this relationship remain unclear. This study aimed to investigate how IKKα regulates macrophage activation and contributes to DCM in a mouse model. Specifically, the role of macrophage IKKα was evaluated in macrophage-specific IKKα knockout mice that received DOX injections. The findings revealed increased expression of IKKα in heart tissues after DOX administration. In mice lacking macrophage IKKα, myocardial injury, ventricular remodeling, inflammation, and proinflammatory macrophage activation worsened in response to DOX administration. Bone marrow transplant studies confirmed that IKKα deficiency exacerbated cardiac dysfunction. Macrophage IKKα knockout also led to mitochondrial damage and metabolic dysfunction in macrophages, thereby resulting in increased cardiomyocyte injury and oxidative stress. Single-cell sequencing analysis revealed that IKKα directly binds to STAT3, leading to the activation of STAT3 phosphorylation at S727. Interestingly, the inhibition of STAT3-S727 phosphorylation suppressed both DCM and cardiomyocyte injury. In conclusion, the IKKα-STAT3-S727 signaling pathway was found to play a crucial role in DOX-induced cardiomyopathy. Targeting this pathway could be a promising therapeutic strategy for treating DOX-related heart failure.
Collapse
Affiliation(s)
- Ganyi Chen
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, Jiangsu, 210006, P.R. China
| | - Yiwei Yao
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, Jiangsu, 210006, P.R. China
| | - Yafeng Liu
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, Jiangsu, 210006, P.R. China
| | - Ruoyu Zhang
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, Jiangsu, 210006, P.R. China
| | - Chenghao Wen
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, Jiangsu, 210006, P.R. China
| | - Qiang Zhou
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, Jiangsu, 210006, P.R. China
| | - Yueyue Xu
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, Jiangsu, 210006, P.R. China
| | - Wuwei Wang
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, Jiangsu, 210006, P.R. China
| | - Hongwei Jiang
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, Jiangsu, 210006, P.R. China
| | - Zhonghao Tao
- Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen Chen
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, Jiangsu, 210006, P.R. China.
| | - Zhibing Qiu
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, Jiangsu, 210006, P.R. China.
| | - Xin Chen
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, Jiangsu, 210006, P.R. China.
| |
Collapse
|
49
|
Xie L, Liu H, Zhang K, Pan Y, Chen M, Xue X, Wan G. Exploring the molecular mechanism of ginseng against anthracycline-induced cardiotoxicity based on network pharmacology, molecular docking and molecular dynamics simulation. Hereditas 2024; 161:31. [PMID: 39243097 PMCID: PMC11378563 DOI: 10.1186/s41065-024-00334-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 09/01/2024] [Indexed: 09/09/2024] Open
Abstract
BACKGROUND Previous clinical and basic studies have revealed that ginseng might have cardioprotective properties against anthracycline-induced cardiotoxicity (AIC). However, the underlying mechanism of ginseng action against AIC remains insufficiently understood. The aim of this study was to explore the related targets and pathways of ginseng against AIC using network pharmacology, molecular docking, cellular thermal shift assay (CETSA) and molecular dynamics (MD) simulations. RESULTS Fourteen drug-disease common targets were identified. Enrichment analysis showed that the AGE-RAGE in diabetic complications, fluid shear stress and atherosclerosis, and TNF signaling pathway were potentially involved in the action of ginseng against AIC. Molecular docking demonstrated that the core components including Kaempferol, beta-Sitosterol, and Fumarine had notable binding activity with the three core targets CCNA2, STAT1, and ICAM1. Furthermore, the stable complex of STAT1 and Kaempferol with favorable affinity was further confirmed by CETSA and MD simulation. CONCLUSIONS This study suggested that ginseng might exert their protective effects against AIC through the derived effector compounds beta-Sitosterol, Kaempferol and Fumarine by targeting CCNA2, STAT1, and ICAM1, and modulating AGE-RAGE in diabetic complications, fluid shear stress and atherosclerosis, and TNF signaling pathways.
Collapse
Affiliation(s)
- Lin Xie
- Department of Oncology, Institute of Medicine and Nursing, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Hanze Liu
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, 39 Chaoyang Road, Shiyan, Hubei, 442000, China
| | - Ke Zhang
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, 39 Chaoyang Road, Shiyan, Hubei, 442000, China
| | - Yijun Pan
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, 39 Chaoyang Road, Shiyan, Hubei, 442000, China
| | - Mengyao Chen
- Department of Oncology, Institute of Medicine and Nursing, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Xiangyue Xue
- Department of Oncology, Institute of Medicine and Nursing, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Guoxing Wan
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, 39 Chaoyang Road, Shiyan, Hubei, 442000, China.
| |
Collapse
|
50
|
Elmorsy EA, Saber S, Hamad RS, Abdel-Reheim MA, El-Kott AF, AlShehri MA, Morsy K, Negm S, Youssef ME. Mechanistic insights into carvedilol's potential protection against doxorubicin-induced cardiotoxicity. Eur J Pharm Sci 2024; 200:106849. [PMID: 38992452 DOI: 10.1016/j.ejps.2024.106849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 06/26/2024] [Accepted: 07/08/2024] [Indexed: 07/13/2024]
Abstract
Doxorubicin (DOX) is an anthracycline chemotherapy drug widely employed in the treatment of various cancers, known for its potent antineoplastic properties but often associated with dose-dependent cardiotoxicity, limiting its clinical use. This review explores the complex molecular details that determine the heart-protective effectiveness of carvedilol in relation to cardiotoxicity caused by DOX. The harmful effects of DOX on heart cells could include oxidative stress, DNA damage, iron imbalance, disruption of autophagy, calcium imbalance, apoptosis, dysregulation of topoisomerase 2-beta, arrhythmogenicity, and inflammatory responses. This review carefully reveals how carvedilol serves as a strong protective mechanism, strategically reducing each aspect of cardiac damage caused by DOX. Carvedilol's antioxidant capabilities involve neutralizing free radicals and adjusting crucial antioxidant enzymes. It skillfully manages iron balance, controls autophagy, and restores the calcium balance essential for cellular stability. Moreover, the anti-apoptotic effects of carvedilol are outlined through the adjustment of Bcl-2 family proteins and activation of the Akt signaling pathway. The medication also controls topoisomerase 2-beta and reduces the renin-angiotensin-aldosterone system, together offering a thorough defense against cardiotoxicity induced by DOX. These findings not only provide detailed understanding into the molecular mechanisms that coordinate heart protection by carvedilol but also offer considerable potential for the creation of targeted treatment strategies intended to relieve cardiotoxicity caused by chemotherapy.
Collapse
Affiliation(s)
- Elsayed A Elmorsy
- Department of Pharmacology and Therapeutics, College of Medicine, Qassim University, Buraidah, 51452, Saudi Arabia; Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt.
| | - Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 11152, Egypt.
| | - Rabab S Hamad
- Biological Sciences Department, College of Science, King Faisal University, Al Ahsa 31982, Saudi Arabia; Central Laboratory, Theodor Bilharz Research Institute, Giza 12411, Egypt.
| | - Mustafa Ahmed Abdel-Reheim
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef 62521, Egypt.
| | - Attalla F El-Kott
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia; Department of Zoology, Faculty of Science, Damanhour University, Egypt
| | - Mohammed A AlShehri
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Kareem Morsy
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia; Department of Zoology, Faculty of Science, Cairo University, Cairo, Egypt
| | - Sally Negm
- Department of Life Sciences, College of Science and Art Mahyel Aseer, King Khalid University, Abha 62529, Saudi Arabia
| | - Mahmoud E Youssef
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 11152, Egypt
| |
Collapse
|