1
|
Echefu G, Batalik L, Lukan A, Shah R, Nain P, Guha A, Brown SA. The Digital Revolution in Medicine: Applications in Cardio-Oncology. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2025; 27:2. [PMID: 39610711 PMCID: PMC11600984 DOI: 10.1007/s11936-024-01059-x] [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: 09/05/2024] [Indexed: 11/30/2024]
Abstract
PURPOSE OF REVIEW A critical evaluation of contemporary literature regarding the role of big data, artificial intelligence, and digital technologies in precision cardio-oncology care and survivorship, emphasizing innovative and groundbreaking endeavors. RECENT FINDINGS Artificial intelligence (AI) algorithm models can automate the risk assessment process and augment current subjective clinical decision tools. AI, particularly machine learning (ML), can identify medically significant patterns in large data sets. Machine learning in cardio-oncology care has great potential in screening, diagnosis, monitoring, and managing cancer therapy-related cardiovascular complications. To this end, large-scale imaging data and clinical information are being leveraged in training efficient AI algorithms that may lead to effective clinical tools for caring for this vulnerable population. Telemedicine may benefit cardio-oncology patients by enhancing healthcare delivery through lowering costs, improving quality, and personalizing care. Similarly, the utilization of wearable biosensors and mobile health technology for remote monitoring holds the potential to improve cardio-oncology outcomes through early intervention and deeper clinical insight. Investigations are ongoing regarding the application of digital health tools such as telemedicine and remote monitoring devices in enhancing the functional status and recovery of cancer patients, particularly those with limited access to centralized services, by increasing physical activity levels and providing access to rehabilitation services. SUMMARY In recent years, advances in cancer survival have increased the prevalence of patients experiencing cancer therapy-related cardiovascular complications. Traditional cardio-oncology risk categorization largely relies on basic clinical features and physician assessment, necessitating advancements in machine learning to create objective prediction models using diverse data sources. Healthcare disparities may be perpetuated through AI algorithms in digital health technologies. In turn, this may have a detrimental effect on minority populations by limiting resource allocation. Several AI-powered innovative health tools could be leveraged to bridge the digital divide and improve access to equitable care.
Collapse
Affiliation(s)
- Gift Echefu
- Division of Cardiovascular Medicine, University of Tennessee, Memphis, TN
| | - Ladislav Batalik
- Department of Rehabilitation, University Hospital Brno, Czech Republic
- Department of Physiotherapy and Rehabilitation, Masaryk University, Brno, Czech Republic
| | | | | | - Priyanshu Nain
- Division of Cardiology, Medical College of Georgia, Augusta, GA
| | - Avirup Guha
- Division of Cardiology, Medical College of Georgia, Augusta, GA
| | - Sherry-Ann Brown
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
- Heart Innovation and Equity Research (HIER) Group, Miami, FL
| |
Collapse
|
2
|
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
|
3
|
Liu Y, Si L, Liu Y, Li S, Zhang X, Jiang S, Liu W, Li X, Zhang L, Zheng H, Liu Z, Hu J, Chen J. Construction of a programmed activation nanosystem based on intracellular hypoxia in cisplatin-resistant tumor cells for reversing cisplatin resistance. Mater Today Bio 2025; 32:101709. [PMID: 40230650 PMCID: PMC11995088 DOI: 10.1016/j.mtbio.2025.101709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/22/2025] [Accepted: 03/25/2025] [Indexed: 04/16/2025] Open
Abstract
Cancer poses a significant threat to human life and health. Cancers treated with cisplatin invariably develop drug resistance. This challenge can be overcome by identifying and exploiting the vulnerabilities acquired by drug-resistant cancer cells, paving the way for finding effective novel treatment options for cisplatin-resistant cancers. Our previous study revealed that cisplatin resistance in cancer cells comes at the cost of increased intracellular hypoxia. In this study, we used 2-nitroimidazole modified hyaluronic acid (HA-NI) as the carrier. The cisplatin-resistant tumor cell specific intracellular hypoxia programmed activation nanomedicine (T/C@HN NPs) was constructed by the hypoxic toxic drug tirapazamine (TPZ) and encapsulating chlorin e6 (Ce6) into HA-NI using polymer assembly technology. The amphiphilic carrier could release free Ce6 molecules under the stimulation of intracellular hypoxic environment, and exhibit specific "activated state" photodynamic properties in cisplatin-resistant tumor cells. Upon irradiation, Ce6-mediated photodynamic therapy further intensifies hypoxia, amplifying its cytotoxicity. This project systematically evaluated the effects of T/C@HN NPs on the identification and recognition of cisplatin-resistant tumors using drug-resistant patient-derived xenograft (PDX) models. This study provides a promising avenue for the development of novel treatment of cisplatin-resistant tumors.
Collapse
Affiliation(s)
- Yurong Liu
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Longqing Si
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Yunheng Liu
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Song Li
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Xiaokang Zhang
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Shaojing Jiang
- Yantai Engineering Research Center for Digital Technology of Stomatology, School of Stomatology, Binzhou Medical University, Yantai, 264003, China
| | - Wenjing Liu
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Xiaolin Li
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Lianguo Zhang
- Department of Thoracic Surgery, Binzhou Medical University Hospital, Binzhou, 256603, China
| | - Hongxia Zheng
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Zhonghao Liu
- Yantai Engineering Research Center for Digital Technology of Stomatology, School of Stomatology, Binzhou Medical University, Yantai, 264003, China
| | - Jinghui Hu
- Yantai Engineering Research Center for Digital Technology of Stomatology, School of Stomatology, Binzhou Medical University, Yantai, 264003, China
| | - Jing Chen
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| |
Collapse
|
4
|
Meattini I, Becherini C, Martella F, Del Bene MR, Saieva C, Bacci C, Coltelli L, Pilato G, Visani L, Salvestrini V, Francolini G, Marrazzo L, Bernini M, Orzalesi L, Nori J, Bianchi S, Olivotto I, Morandi A, Curigliano G, Barletta G, Livi L. Cardioprotection in patients with anthracycline-treated breast cancer: final analysis from the 2 × 2 randomized, placebo-controlled, double-blind SAFE trial. ESMO Open 2025; 10:105116. [PMID: 40403385 DOI: 10.1016/j.esmoop.2025.105116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/01/2025] [Accepted: 04/20/2025] [Indexed: 05/24/2025] Open
Abstract
BACKGROUND Anthracycline-based chemotherapy is a cornerstone in breast cancer treatment but is associated with cardiotoxicity, including subclinical cardiac damage. This study evaluates the efficacy of ramipril and bisoprolol in preventing subclinical cardiac impairment in patients with nonmetastatic breast cancer undergoing anthracycline-based chemotherapy. PATIENTS AND METHODS The SAFE trial is a multicenter, 2 × 2 factorial, randomized, placebo-controlled, double-blind study involving 262 patients. Participants were allocated to one of four groups: placebo-placebo, ramipril-placebo, bisoprolol-placebo, or ramipril-bisoprolol, administered concurrently with chemotherapy. Subclinical cardiac damage was assessed at 24 months using echocardiographic measures, specifically a ≥10% reduction in three-dimensional left ventricular ejection fraction (3D-LVEF) or global longitudinal strain (GLS). RESULTS At 24 months, patients receiving ramipril, bisoprolol, or their combination experienced significantly smaller declines in 3D-LVEF compared with placebo (-2.1%, -2.2%, and -3.4%, respectively; all P < 0.001). GLS results were consistent with these findings (P < 0.001). Subclinical cardiac damage occurred in 11.4% of patients receiving ramipril versus 39.3% without ramipril (P < 0.001), and in 9.6% of patients receiving bisoprolol versus 43.5% without bisoprolol (P < 0.001). CONCLUSIONS Ramipril and bisoprolol significantly reduce the incidence of subclinical cardiac damage in patients with breast cancer undergoing anthracycline-based chemotherapy, thus supporting their use as early prevention cardioprotective strategies.
Collapse
Affiliation(s)
- I Meattini
- Department of Experimental and Clinical Biomedical Sciences "M. Serio", University of Florence, Florence, Italy; Radiation Oncology and Breast Unit, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy.
| | - C Becherini
- Radiation Oncology and Breast Unit, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - F Martella
- Oncology Department, Breast and Medical Oncology Units, Azienda USL Toscana Centro, Florence, Italy
| | - M R Del Bene
- Diagnostic Cardiology, CardioThoracic and Vascular Department, Careggi University Hospital, Florence, Italy
| | - C Saieva
- Cancer Risk Factors and Lifestyle Epidemiology Unit, Institute for Cancer Research Prevention and Clinical Network (ISPRO), Florence, Italy
| | - C Bacci
- Oncology Department, Breast and Medical Oncology Units, Azienda USL Toscana Centro, Florence, Italy
| | - L Coltelli
- Medical Oncology Unit, Livorno Hospital, Azienda USL Toscana Nord Ovest, Livorno, Italy
| | - G Pilato
- Diagnostic Cardiology, CardioThoracic and Vascular Department, Careggi University Hospital, Florence, Italy
| | - L Visani
- Radiation Oncology and Breast Unit, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - V Salvestrini
- Radiation Oncology and Breast Unit, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - G Francolini
- Radiation Oncology and Breast Unit, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - L Marrazzo
- Department of Experimental and Clinical Biomedical Sciences "M. Serio", University of Florence, Florence, Italy; Oncology Department, Medical Physics Unit, Careggi University Hospital, Florence, Italy
| | - M Bernini
- Breast Surgery Unit, Careggi University Hospital, Florence, Italy
| | - L Orzalesi
- Breast Surgery Unit, Careggi University Hospital, Florence, Italy
| | - J Nori
- Diagnostic Senology Unit, Careggi University Hospital, Florence, Italy
| | - S Bianchi
- Department of Health Sciences, Division of Pathological Anatomy, University of Florence, Florence, Italy
| | - I Olivotto
- Department of Experimental and Clinical Medicine, Division of Physiology, University of Florence, Florence, Italy
| | - A Morandi
- Department of Experimental and Clinical Biomedical Sciences "M. Serio", University of Florence, Florence, Italy
| | - G Curigliano
- Istituto Europeo di Oncologia, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milano, Milan, Italy. https://twitter.com/curijoey
| | - G Barletta
- Diagnostic Cardiology, CardioThoracic and Vascular Department, Careggi University Hospital, Florence, Italy
| | - L Livi
- Department of Experimental and Clinical Biomedical Sciences "M. Serio", University of Florence, Florence, Italy; Radiation Oncology and Breast Unit, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| |
Collapse
|
5
|
Rizk SI, Costa IBSDS, Cruz CBBV, Pileggi B, de Almeida Andrade FT, Gonzalez TB, Bittar CS, Fukushima JT, Quintao VC, Osawa EA, Alves JBS, Fonseca SMR, Garcia DR, Pereira J, Buccheri V, Avila J, Kawahara LT, Barros CCS, Ikeoka LT, Nakada LN, Fellini M, Rocha VG, Rego EM, Hoff PMG, Filho RK, Landoni G, Hajjar LA. Randomized, Placebo-Controlled, Triple-Blind Clinical Trial of Ivabradine for the Prevention of Cardiac Dysfunction During Anthracycline-Based Cancer Therapy. J Am Heart Assoc 2025; 14:e039745. [PMID: 40357644 DOI: 10.1161/jaha.124.039745] [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: 10/29/2024] [Accepted: 03/13/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND Cancer therapy-related cardiac dysfunction frequently occurs in patients receiving anthracycline. Ivabradine reduces heart rate without affecting contractility and showed anti-inflammatory, antioxidant, and antiapoptotic effects in experimental cardiotoxicity models. This study aims to evaluate the effect of ivabradine on cancer therapy-related cardiac dysfunction in patients with lymphoma or sarcoma treated with anthracycline. METHODS In a randomized, triple-blind trial, patients starting anthracycline therapy received either ivabradine 5 mg twice daily or placebo until 30 days after completing treatment. The primary outcome was the incidence of cardiotoxicity measured as a ≥10% relative reduction in global longitudinal strain at 12 months from baseline. Secondary outcomes included 12-month clinical outcomes, a ≥10% decrease in the left ventricular ejection fraction to <55%, diastolic dysfunction, and troponin T and N-terminal pro-B-type natriuretic peptide levels. RESULTS This study enrolled 107 patients (51 in the ivabradine group and 56 in the placebo group). The median dose of anthracycline was 300 mg/m2 (250-300 mg/m2) in both groups. Cardiotoxicity measured as a ≥10% relative reduction in global longitudinal strain at 12 months was reached in 57% versus 50% in the ivabradine and placebo groups (odds ratio, 1.32 [95% CI, 0.61-2.83]; P=0.477). Fewer patients in the ivabradine group than in the placebo group had troponin T levels ≥14 ng/L (16 [39.0%] versus 23 [62.2%]; P=0.041) at 6 months, with this difference not maintained at the 12-month follow-up. In addition, there were no differences in the other secondary outcomes. CONCLUSIONS A fixed 10 mg/day dose of ivabradine does not protect patients with cancer against anthracycline cardiotoxicity. REGISTRATION URL: https://clinicaltrials.gov/; Unique Identifier: NCT03650205.
Collapse
Affiliation(s)
- Stephanie Itala Rizk
- Instituto do Coração (InCor), Hospital das Clínicas Faculdade de Medicina da Universidade de São Paulo São Paulo SP Brazil
- Instituto do Câncer (ICESP), Hospital das Clínicas Faculdade de Medicina da Universidade de São Paulo São Paulo SP Brazil
| | | | - Cecília Beatriz Bittencourt Viana Cruz
- Instituto do Coração (InCor), Hospital das Clínicas Faculdade de Medicina da Universidade de São Paulo São Paulo SP Brazil
- Instituto do Câncer (ICESP), Hospital das Clínicas Faculdade de Medicina da Universidade de São Paulo São Paulo SP Brazil
| | - Brunna Pileggi
- Instituto do Coração (InCor), Hospital das Clínicas Faculdade de Medicina da Universidade de São Paulo São Paulo SP Brazil
| | | | - Thalita Barbosa Gonzalez
- Instituto do Câncer (ICESP), Hospital das Clínicas Faculdade de Medicina da Universidade de São Paulo São Paulo SP Brazil
| | - Cristina Salvadori Bittar
- Instituto do Coração (InCor), Hospital das Clínicas Faculdade de Medicina da Universidade de São Paulo São Paulo SP Brazil
- Instituto do Câncer (ICESP), Hospital das Clínicas Faculdade de Medicina da Universidade de São Paulo São Paulo SP Brazil
| | - Julia Tizue Fukushima
- Instituto do Câncer (ICESP), Hospital das Clínicas Faculdade de Medicina da Universidade de São Paulo São Paulo SP Brazil
| | - Vinicius Caldeira Quintao
- Instituto do Coração (InCor), Hospital das Clínicas Faculdade de Medicina da Universidade de São Paulo São Paulo SP Brazil
| | - Eduardo Atsushi Osawa
- Instituto do Coração (InCor), Hospital das Clínicas Faculdade de Medicina da Universidade de São Paulo São Paulo SP Brazil
| | - Juliana Barbosa Sobral Alves
- Instituto do Coração (InCor), Hospital das Clínicas Faculdade de Medicina da Universidade de São Paulo São Paulo SP Brazil
- Instituto do Câncer (ICESP), Hospital das Clínicas Faculdade de Medicina da Universidade de São Paulo São Paulo SP Brazil
| | - Silvia Moulin Ribeiro Fonseca
- Instituto do Câncer (ICESP), Hospital das Clínicas Faculdade de Medicina da Universidade de São Paulo São Paulo SP Brazil
| | - Diego Ribeiro Garcia
- Instituto do Coração (InCor), Hospital das Clínicas Faculdade de Medicina da Universidade de São Paulo São Paulo SP Brazil
| | - Juliana Pereira
- Instituto do Câncer (ICESP), Hospital das Clínicas Faculdade de Medicina da Universidade de São Paulo São Paulo SP Brazil
| | - Valeria Buccheri
- Instituto do Câncer (ICESP), Hospital das Clínicas Faculdade de Medicina da Universidade de São Paulo São Paulo SP Brazil
| | - Juliana Avila
- Instituto do Coração (InCor), Hospital das Clínicas Faculdade de Medicina da Universidade de São Paulo São Paulo SP Brazil
| | - Lucas Tokio Kawahara
- Instituto do Coração (InCor), Hospital das Clínicas Faculdade de Medicina da Universidade de São Paulo São Paulo SP Brazil
| | - Cecilia Chie Sakaguchi Barros
- Instituto do Coração (InCor), Hospital das Clínicas Faculdade de Medicina da Universidade de São Paulo São Paulo SP Brazil
| | - Lucas Takeshi Ikeoka
- Instituto do Coração (InCor), Hospital das Clínicas Faculdade de Medicina da Universidade de São Paulo São Paulo SP Brazil
| | - Letícia Naomi Nakada
- Instituto do Coração (InCor), Hospital das Clínicas Faculdade de Medicina da Universidade de São Paulo São Paulo SP Brazil
| | - Mariella Fellini
- Instituto do Coração (InCor), Hospital das Clínicas Faculdade de Medicina da Universidade de São Paulo São Paulo SP Brazil
| | - Vanderson Geraldo Rocha
- Instituto do Câncer (ICESP), Hospital das Clínicas Faculdade de Medicina da Universidade de São Paulo São Paulo SP Brazil
- Departamento de Hematologia e Terapia Celular do Hospital das Clínicas Faculdade de Medicina da Universidade de São Paulo São Paulo Brazil
| | - Eduardo Magalhães Rego
- Departamento de Hematologia e Terapia Celular do Hospital das Clínicas Faculdade de Medicina da Universidade de São Paulo São Paulo Brazil
| | - Paulo Marcelo Gehm Hoff
- Instituto do Câncer (ICESP), Hospital das Clínicas Faculdade de Medicina da Universidade de São Paulo São Paulo SP Brazil
- Departamento de Hematologia e Terapia Celular do Hospital das Clínicas Faculdade de Medicina da Universidade de São Paulo São Paulo Brazil
| | - Roberto Kalil Filho
- Instituto do Coração (InCor), Hospital das Clínicas Faculdade de Medicina da Universidade de São Paulo São Paulo SP Brazil
| | - Giovanni Landoni
- Anesthesia and Intensive Care Department IRCCS San Raffaele Scientific Institute Milan Italy
| | - Ludhmila Abrahão Hajjar
- Instituto do Coração (InCor), Hospital das Clínicas Faculdade de Medicina da Universidade de São Paulo São Paulo SP Brazil
- Instituto do Câncer (ICESP), Hospital das Clínicas Faculdade de Medicina da Universidade de São Paulo São Paulo SP Brazil
- Disciplina de Emergências Hospital das Clínicas Faculdade de Medicina da Universidade de São Paulo São Paulo SP Brazil
| |
Collapse
|
6
|
Singh R, Patel K, Xu H, Adeniyi A, Upshaw JN, Van Buren P, Kaufman PA, Dittus K, Landry KK. Cardio-Oncology and Breast Cancer Therapies. Curr Treat Options Oncol 2025; 26:385-397. [PMID: 40257669 DOI: 10.1007/s11864-025-01311-x] [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: 03/17/2025] [Indexed: 04/22/2025]
Abstract
OPINION STATEMENT Assessing cardiac risk prior to initiating breast cancer treatment, monitoring cardiac function during treatment, and implementing appropriate follow-up strategies are essential components of managing cardiotoxicity in breast cancer patients. A comprehensive cardiovascular evaluation should be conducted before treatment, including a detailed medical history, physical examination, and baseline cardiac imaging. Risk stratification tools can aid in determining the individual patient's risk profile. Close monitoring of cardiac function, including regular assessment of left ventricular ejection fraction (LVEF) and monitoring for signs and symptoms of cardiac dysfunction, is crucial during treatment. Prompt action should be taken if an adverse cardiovascular event is detected, including considering discontinuing or modifying the treatment regimen. Appropriate follow-up care is essential to monitor for long-term cardiac effects and optimize cardiovascular health in breast cancer survivors. Regular cardiovascular assessments, lifestyle modifications, and collaboration between healthcare professionals are important in managing cardiotoxicity effectively.
Collapse
Affiliation(s)
- Rohit Singh
- Division of Hematology/Oncology, Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- University of Vermont Cancer Center, Burlington, VT, USA
| | - Krina Patel
- Division of Cardiology, Department of Medicine, Tufts Medical Center, Boston, MA, USA
| | - Haze Xu
- Division of Hematology/Oncology, Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- University of Vermont Cancer Center, Burlington, VT, USA
| | - Aderonke Adeniyi
- Division of Cardiology, Department of Medicine, Wake Forest Baptist Medical Center, Winston-Salem, NC, USA
| | - Jenica N Upshaw
- Division of Cardiology, Department of Medicine, Tufts Medical Center, Boston, MA, USA
| | - Peter Van Buren
- Division of Cardiology, Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Peter A Kaufman
- Division of Hematology/Oncology, Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- University of Vermont Cancer Center, Burlington, VT, USA
| | - Kim Dittus
- Division of Hematology/Oncology, Department of Medicine, University of Wisconsin, Madison, WI, USA
| | - Kara K Landry
- Division of Hematology/Oncology, Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA.
- University of Vermont Cancer Center, Burlington, VT, USA.
| |
Collapse
|
7
|
Solomon AD, Dabral S, Brajesh RG, Day BW, Juric M, Zielonka J, Bosnjak ZJ, Pant T. Understanding the Mechanisms of Chemotherapy-Related Cardiotoxicity Employing hiPSC-Derived Cardiomyocyte Models for Drug Screening and the Identification of Genetic and Epigenetic Variants. Int J Mol Sci 2025; 26:3966. [PMID: 40362211 PMCID: PMC12071959 DOI: 10.3390/ijms26093966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/10/2025] [Accepted: 04/18/2025] [Indexed: 05/15/2025] Open
Abstract
Chemotherapy-related cardiotoxicity (CTRTOX) is a profound and common side effect of cancer-based therapy in a subset of patients. The underlying factors and the associated mechanisms contributing to severe toxicity of the heart among these patients remain unknown. While challenges remain in accessing human subjects and their ventricular cardiomyocytes (CMs), advancements in human induced pluripotent stem cell (hiPSC)-technology-based CM differentiation protocols over the past few decades have paved the path for iPSC-based models of human cardiac diseases. Here, we offer a detailed analysis of the underlying mechanisms of CTRTOX. We also discuss the recent advances in therapeutic strategies in different animal models and clinical trials. Furthermore, we explore the prospects of iPSC-based models for identifying novel functional targets and developing safer chemotherapy regimens for cancer patients that may be beneficial for developing personalized cardioprotectants and their application in clinical practice.
Collapse
Affiliation(s)
- Abhishikt David Solomon
- Adams School of Dentistry, Oral and Craniofacial Biomedicine, University of North Carolina, Chapel Hill, NC 27599, USA;
| | - Swarna Dabral
- Maharishi Markandeshwar College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala 133207, India;
| | - Raman Gulab Brajesh
- Department of Biomedical Engineering and Bioinformatics, Swami Vivekanand Technical University, Durg 491107, India;
| | | | - Matea Juric
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA; (M.J.); (J.Z.)
| | - Jacek Zielonka
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA; (M.J.); (J.Z.)
| | - Zeljko J. Bosnjak
- Department of Medicine, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA;
| | - Tarun Pant
- Department of Medicine, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA;
- Department of Surgery, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| |
Collapse
|
8
|
Kézdi Á, Szelke E, Dank M, Mühl D, Szentmártoni G, Szabó G, Joseph Fogarasi D, Takács I, Horváth VJ, Tabák ÁG. Effects of taxane-anthracycline and taxane only treatment on cardiac function in breast cancer-a retrospective cohort study. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2025; 11:37. [PMID: 40221795 PMCID: PMC11992891 DOI: 10.1186/s40959-025-00335-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 04/03/2025] [Indexed: 04/14/2025]
Abstract
INTRODUCTION Cardiotoxic, anthracycline-based therapies have high value in selected patients with breast cancer. We aimed to describe the effect of anthracycline plus taxane and single taxane chemotherapies on echocardiographic parameters in women with breast cancer. METHODS We retrospectively analysed data of 68 women (> 18 years old) treated for breast cancer in 2018-2021 in the Cardiology Outpatient Clinic of Semmelweis University, Department of Internal Medicine and Oncology. Cardiovascular medical history was collected at baseline and transthoracic echocardiography was completed at each visit. Also, we reviewed electronic medical records for other relevant medical information. Measured echocardiography parameters were assigned to five periods (0-14 days, then every half year and beyond day 545) based on the time since the first treatment. Trajectories of ejection fraction and diastolic function associated markers over the follow-up periods were analysed by linear mixed models. RESULTS Mean age of the anthracycline plus taxane group was 52.7 ± 14.1 years, of the single taxane group 55.2 ± 13.1 years. The mean anthracycline dose was equivalent to 240 mg/m2 of doxorubicin. Overall pre-existing cardiovascular burden was low. Statistically significant changes were found only in the anthracycline plus taxane group: ejection fraction decreased mildly from 65.5 ± 3.1% at baseline to 62.1 ± 3.2% at 181-365 days (p = 0.007) while deceleration time decreased mildly from 227.9 ± 33.9 msec to 197.4 ± 29.4 msec at 15-180 days (p = 0.028). Both drops were only temporary and values neared baseline values over follow-up (p = NS vs. baseline). Other important determinants of ejection fraction were age and hypertension among the investigated risk factors. CONCLUSION Our study confirms the overall safety on cardiac function of both single taxane and anthracycline plus taxane chemotherapy, as we found no changes in echocardiographic parameters associated with single taxane therapy, while anthracycline plus taxane chemotherapy was associated with a temporary and clinically insignificant reduction of ejection fraction and deceleration time over 1.5 years of follow-up. Our study is limited by its retrospective nature and the low number of participants.
Collapse
Affiliation(s)
- Árpád Kézdi
- Department of Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary
- School of PhD Studies, Semmelweis University Faculty of Medicine, Budapest, Hungary
- Institute of Preventive Medicine and Public Health, Semmelweis University Faculty of Medicine, Budapest, Hungary
| | - Emese Szelke
- Department of Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary
| | - Magdolna Dank
- Department of Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary
| | - Dorottya Mühl
- Department of Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary
| | | | - Gergely Szabó
- Department of Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary
| | | | - István Takács
- Department of Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary
| | - Viktor J Horváth
- Department of Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary.
| | - Ádám G Tabák
- Department of Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary
- Institute of Preventive Medicine and Public Health, Semmelweis University Faculty of Medicine, Budapest, Hungary
- Department of Epidemiology and Public Health, University College London, London, UK
| |
Collapse
|
9
|
Contaldi C, D’Aniello C, Panico D, Zito A, Calabrò P, Di Lorenzo E, Golino P, Montesarchio V. Cancer-Therapy-Related Cardiac Dysfunction: Latest Advances in Prevention and Treatment. Life (Basel) 2025; 15:471. [PMID: 40141815 PMCID: PMC11944213 DOI: 10.3390/life15030471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/19/2025] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
The increasing efficacy of cancer therapies has significantly improved survival rates, but it has also highlighted the prevalence of cancer-therapy-related cardiac dysfunction (CTRCD). This review provides a comprehensive overview of the identification, monitoring, and management of CTRCD, a condition resulting from several treatments, such as anthracyclines, HER2-targeted therapies, target therapies, and radiotherapy. The paper includes a discussion of the mechanisms of CTRCD associated with various cancer treatments. Early detection through serum biomarkers and advanced imaging techniques is crucial for effective monitoring and risk stratification. Preventive strategies include pharmacological interventions such as ACE inhibitors/angiotensin receptor blockers, beta-blockers, and statins. Additionally, novel agents like sacubitril/valsartan, sodium-glucose co-transporter type 2 inhibitors, and vericiguat show promise in managing left ventricular dysfunction. Lifestyle modifications, including structured exercise programs and optimized nutritional strategies, further contribute to cardioprotection. The latest treatments for both asymptomatic and symptomatic CTRCD across its various stages are also discussed. Emerging technologies, including genomics, artificial intelligence, novel biomarkers, and gene therapy, are paving the way for personalized approaches to CTRCD prevention and treatment. These advancements hold great promise for improving long-term outcomes in cancer patients by minimizing cardiovascular complications.
Collapse
Affiliation(s)
- Carla Contaldi
- Department of Cardiology, AORN dei Colli-Monaldi Hospital, 80131 Naples, Italy
| | - Carmine D’Aniello
- Division of Medical Oncology, AORN dei Colli-Monaldi Hospital, 80131 Naples, Italy
| | - Domenico Panico
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy
| | - Andrea Zito
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy
| | - Paolo Calabrò
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy
| | - Emilio Di Lorenzo
- Department of Cardiology, AORN dei Colli-Monaldi Hospital, 80131 Naples, Italy
| | - Paolo Golino
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy
| | | |
Collapse
|
10
|
Li X, Su X, Liang W, Wang L, Yuan C, Xu J, Zhang Y, Liu Y, Ma N, Yang F, Yang Y, Tao L, Sun S, Shang H, Xing Y. Cardioprotective effect of crocin in patients with breast cancer receiving anthracycline-based chemotherapy: A randomized, double-blind, placebo-controlled study. Pharmacol Res 2025; 213:107630. [PMID: 39889867 DOI: 10.1016/j.phrs.2025.107630] [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: 11/25/2024] [Revised: 01/27/2025] [Accepted: 01/27/2025] [Indexed: 02/03/2025]
Abstract
Anthracycline chemotherapy leads to cardiotoxicity in patients with breast cancer. We performed a double-blind randomized controlled trial to assess the efficacy of crocin (crocin tablets) in reducing anthracycline-induced cardiotoxicity in breast cancer patients. Eligible patients were randomly assigned to receive either treatment with crocin tablets or placebo for 6 months. Primary efficacy outcome was the proportion of patients with a drop in left ventricular ejection fraction (LVEF) of at least 10 % from baseline until 6 months. We randomized 200 patients; 7 of them had no valid randomization as the change in chemotherapy scheme. A total of 193 eligible participants (mean [SD] age, 50.9 [9.6] years; all women) were randomly assigned to receive crocin tablets or placebo. The incidence of the primary efficacy outcome was 7.2 % (7/97) in the crocin group and 17.7 % (17/96) in the placebo group (P = 0.027). At 6 months, there were significant differences in the mean change of N-terminal pro-brain natriuretic peptide (NT-pro BNP) between the groups (3.06 [-25.18-6.95] vs. 3.06 [-4.85-24.98] pg/ml; P = 0.017). The changes in heart rate from baseline to 6 months showed a significant difference as the mean difference of -4.00 (95 %CI, -6.95 to -1.05) bpm (P = 0.008). Conclusively, among patients with breast cancer treated with anthracycline-based chemotherapy, crocin tablets reduced the incidence of LVEF reduction. This finding indicated that crocin tablets might be a safe and effective therapy to prevent the cardiotoxicity in this population. (Chictr.org.cn, ID Number: ChiCTR2000041134).
Collapse
Affiliation(s)
- Xinye Li
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China; Beijing University of Chinese Medicine, Beijing, China
| | - Xin Su
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in SouthChina, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Wanping Liang
- The First Affiliated Hospital, Hebei North University, Zhangjiakou, China
| | - Li Wang
- Xingtai People's Hospital, Xingtai, China
| | - Chao Yuan
- Dezhou Second People's Hospital, Dezhou, China
| | - Juping Xu
- Jiaozuo Second People's Hospital, Jiaozuo, China
| | - Yijun Zhang
- The First Affiliated Hospital, Hebei North University, Zhangjiakou, China
| | - Yan Liu
- Jiaozuo Second People's Hospital, Jiaozuo, China
| | - Ning Ma
- Dezhou Second People's Hospital, Dezhou, China
| | - Fan Yang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yiyuan Yang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Liyuan Tao
- Clinical Epidemiology Research Center of the Third Hospital of Peking University, Beijing, China
| | - Shipeng Sun
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hongcai Shang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China.
| | - Yanwei Xing
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
11
|
Gigli M, Stolfo D, Merlo M, Sinagra G, Taylor MRG, Mestroni L. Pathophysiology of dilated cardiomyopathy: from mechanisms to precision medicine. Nat Rev Cardiol 2025; 22:183-198. [PMID: 39394525 PMCID: PMC12046608 DOI: 10.1038/s41569-024-01074-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/20/2024] [Indexed: 10/13/2024]
Abstract
Dilated cardiomyopathy (DCM) is a complex disease with multiple causes and various pathogenic mechanisms. Despite improvements in the prognosis of patients with DCM in the past decade, this condition remains a leading cause of heart failure and premature death. Conventional treatment for DCM is based on the foundational therapies for heart failure with reduced ejection fraction. However, increasingly, attention is being directed towards individualized treatments and precision medicine. The ability to confirm genetic causality is gradually being complemented by an increased understanding of genotype-phenotype correlations. Non-genetic factors also influence the onset of DCM, and growing evidence links genetic background with concomitant non-genetic triggers or precipitating factors, increasing the extreme complexity of the pathophysiology of DCM. This Review covers the spectrum of pathophysiological mechanisms in DCM, from monogenic causes to the coexistence of genetic abnormalities and triggering environmental factors (the 'two-hit' hypothesis). The roles of common genetic variants in the general population and of gene modifiers in disease onset and progression are also discussed. Finally, areas for future research are highlighted, particularly novel therapies, such as small molecules, RNA and gene therapy, and measures for the prevention of arrhythmic death.
Collapse
Affiliation(s)
- Marta Gigli
- Cardiothoracovascular Department, Centre for Diagnosis and Treatment of Cardiomyopathies, European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart (ERN GUARD-Heart), Azienda Sanitaria Universitaria Giuliano-Isontina (ASUGI) and University of Trieste, Trieste, Italy
| | - Davide Stolfo
- Cardiothoracovascular Department, Centre for Diagnosis and Treatment of Cardiomyopathies, European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart (ERN GUARD-Heart), Azienda Sanitaria Universitaria Giuliano-Isontina (ASUGI) and University of Trieste, Trieste, Italy
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Marco Merlo
- Cardiothoracovascular Department, Centre for Diagnosis and Treatment of Cardiomyopathies, European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart (ERN GUARD-Heart), Azienda Sanitaria Universitaria Giuliano-Isontina (ASUGI) and University of Trieste, Trieste, Italy
| | - Gianfranco Sinagra
- Cardiothoracovascular Department, Centre for Diagnosis and Treatment of Cardiomyopathies, European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart (ERN GUARD-Heart), Azienda Sanitaria Universitaria Giuliano-Isontina (ASUGI) and University of Trieste, Trieste, Italy
| | - Matthew R G Taylor
- Adult Medical Genetics Program, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Luisa Mestroni
- Molecular Genetics Program, Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
12
|
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
|
13
|
Balough E, Ariza A, Asnani A, Hoeger CW. Cardiotoxicity of Anthracyclines. Cardiol Clin 2025; 43:111-127. [PMID: 39551553 DOI: 10.1016/j.ccl.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Anthracycline chemotherapy is associated with cardiotoxicity, predominantly manifesting as left ventricular systolic dysfunction within the first year of treatment. Early detection is possible through biomarkers and cardiovascular imaging before clinical symptoms develop. Comprehensive cardiovascular risk assessment is essential for all patients prior to anthracycline therapy to stratify their risk of cardiotoxicity. Preventive measures, including cardiovascular risk optimization, as well as anthracycline dose adjustments, the use of liposomal anthracyclines, and dexrazoxane in high-risk patients, are crucial to mitigate the risk of cardiotoxicity. Long-term follow-up and cardiovascular risk optimization are critical for cancer survivors to optimize cardiovascular outcomes.
Collapse
Affiliation(s)
- Elizabeth Balough
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, 185 Pilgrim Road, Baker 4, Boston, MA 02215, USA. https://twitter.com/ElizabethBaloug
| | - Abul Ariza
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; 3 Blackfan Circle, CLS-911, Boston, MA 02115, USA
| | - Aarti Asnani
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; 3 Blackfan Circle, CLS-911, Boston, MA 02115, USA. https://twitter.com/AartiAsnaniMD
| | - Christopher W Hoeger
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, 185 Pilgrim Road, Baker 4, Boston, MA 02215, USA.
| |
Collapse
|
14
|
Moreno-Arciniegas A, Cádiz L, Galán-Arriola C, Clemente-Moragón A, Ibáñez B. Cardioprotection strategies for anthracycline cardiotoxicity. Basic Res Cardiol 2025; 120:71-90. [PMID: 39249555 PMCID: PMC11790697 DOI: 10.1007/s00395-024-01078-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/25/2024] [Accepted: 08/26/2024] [Indexed: 09/10/2024]
Abstract
Thanks to the fantastic progress in cancer therapy options, there is a growing population of cancer survivors. This success has resulted in a need to focus much effort into improving the quality of life of this population. Cancer and cardiovascular disease share many common risk factors and have an interplay between them, with one condition mechanistically affecting the other and vice versa. Furthermore, widely prescribed cancer therapies have known toxic effects in the cardiovascular system. Anthracyclines are the paradigm of efficacious cancer therapy widely prescribed with a strong cardiotoxic potential. While some cancer therapies cardiovascular toxicities are transient, others are irreversible. There is a growing need to develop cardioprotective therapies that, when used in conjunction with cancer therapies, can prevent cardiovascular toxicity and thus improve long-term quality of life in survivors. The field has three main challenges: (i) identification of the ultimate mechanisms leading to cardiotoxicity to (ii) identify specific therapeutic targets, and (iii) more sensible diagnostic tools to early identify these conditions. In this review we will focus on the cardioprotective strategies tested and under investigation. We will focus this article into anthracycline cardiotoxicity since it is still the agent most widely prescribed, the one with higher toxic effects on the heart, and the most widely studied.
Collapse
Affiliation(s)
| | - Laura Cádiz
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Carlos Galán-Arriola
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Agustín Clemente-Moragón
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Borja Ibáñez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.
- Cardiology Department, IIS-Fundación Jiménez Díaz University Hospital, Madrid, Spain.
| |
Collapse
|
15
|
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
|
16
|
Shugg T, Nguyen T, Hua X, Richards B, Rae J, Dess R, Perry D, Kay B, Hayek SS, Leja M, Luzum JA. Factors Associated With the Recovery of Left Ventricular Ejection Fraction in Patients With Anthracycline-Induced Left Ventricular Dysfunction. J Cardiovasc Pharmacol Ther 2025; 30:10742484241304304. [PMID: 39801159 PMCID: PMC11831319 DOI: 10.1177/10742484241304304] [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] [Indexed: 01/30/2025]
Abstract
BACKGROUND Neurohormonal blocking drugs, like beta-blockers, angiotensin-converting enzyme inhibitors (ACEIs), and angiotensin receptor blockers (ARBs), are recommended for treating anthracycline-induced left ventricular dysfunction (AILVD). However, there is limited evidence supporting their benefit. Therefore, this study evaluated associations of neurohormonal blockers and other clinical factors with recovery of left ventricular ejection fraction (LVEF) in patients with AILVD. METHODS This retrospective chart review assessed patients treated with at least one dose of anthracycline, then had ≥10% LVEF reduction or post-anthracycline LVEF value <50%, and then had a follow-up LVEF measurement ≥90 days later. The primary endpoint was LVEF recovery (highest follow-up LVEF-lowest LVEF post-anthracycline). Variables from univariable tests with P < .1 were incorporated in a multiple linear regression model for independent factors significantly associated with LVEF recovery (P < .05). RESULTS Out of 104 patients, 83% were female, 86% self-reported white race, 53% had breast cancer, median (IQR) age was 52 (22) years, and LVEF recovery was 14% (16%). The final multivariable model included 2 significant variables: beta-blocker dose after anthracycline exposure (every 25 mg increase in beta-blocker dose was associated with 5.0% increase in LVEF recovery; P = .0005) and the time between the start of the anthracycline and the lowest LVEF post-anthracycline (every 5-year increase in time was associated with 1.8% decrease in LVEF recovery; P = .0379). CONCLUSIONS In patients with AILVD, a higher beta-blocker dose and earlier detection of LVEF reduction post-anthracycline were significantly and independently associated with improved LVEF recovery. These findings need to be validated in a larger, independent cohort.
Collapse
Affiliation(s)
- Tyler Shugg
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Tk Nguyen
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Xuesi Hua
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Blair Richards
- Michigan Institute for Clinical & Health Research (MICHR), University of Michigan, Ann Arbor, MI, USA
| | - James Rae
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Robert Dess
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Daniel Perry
- Division of Cardiovascular Medicine, Department of Internal Medicine, Michigan Medicine, Ann Arbor, MI, USA
| | - Bradley Kay
- Department of Cardiology, Yale School of Medicine, New Haven, CT, USA
| | - Salim S. Hayek
- Division of Cardiovascular Medicine, Department of Internal Medicine, Michigan Medicine, Ann Arbor, MI, USA
| | - Monika Leja
- Division of Cardiovascular Medicine, Department of Internal Medicine, Michigan Medicine, Ann Arbor, MI, USA
| | - Jasmine A. Luzum
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| |
Collapse
|
17
|
Di Lisi D, Madaudo C, Macaione F, Galassi AR, Novo G. Cancer survivors and cardiovascular diseases: from preventive strategies to treatment. J Cardiovasc Med (Hagerstown) 2025; 26:8-17. [PMID: 39514326 DOI: 10.2459/jcm.0000000000001681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024]
Abstract
During the last decades, progress in the treatment of oncological diseases has led to an increase in the survival of cancer patients: cancer survivors (CS). Thus, the incidence of CS has increased enormously, in both adult CS and childhood and adolescent CS. Unfortunately, CS treated with anthracyclines, chest radiotherapy (RT) and other potentially cardiotoxic drugs have a higher risk of cardiovascular (CV) toxicity: heart failure with reduced ejection fraction (HFrEF), valve diseases, coronary artery diseases, vascular diseases and pericardial diseases. In fact, chest irradiation can cause coronary artery diseases that can be latent until at least 10 years after exposure; also, valvular heart diseases can appear after >20 years following irradiation; heart failure may appear later, several years after anticancer drugs or RT. Therefore, it is very important to stratify the CV risk of cancer patients at the end of cardiotoxic drugs, to plan the most appropriate long-term surveillance program, in accordance with 2022 ESC Guidelines on Cardio-Oncology, to prevent late cardiovascular complications. Monitoring of cancer patients must not stop during anticancer treatment but it must continue afterwards, depending on the patient's CV risk. CV toxicity risk should be reassessed 5 years after therapy to organize long-term follow-up. Considering late cardiotoxicity in CS, our review aims to evaluate the incidence of cardiovascular diseases in CS, their mechanisms, surveillance protocols, preventive strategies, diagnosis and treatment.
Collapse
Affiliation(s)
- Daniela Di Lisi
- Division of Cardiology, University Hospital 'Paolo Giaccone'
| | - Cristina Madaudo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE) 'G. D'Alessandro', University of Palermo, Palermo, Italy
| | | | - Alfredo Ruggero Galassi
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE) 'G. D'Alessandro', University of Palermo, Palermo, Italy
| | - Giuseppina Novo
- Division of Cardiology, University Hospital 'Paolo Giaccone'
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE) 'G. D'Alessandro', University of Palermo, Palermo, Italy
| |
Collapse
|
18
|
Theofilis P, Vlachakis PK, Oikonomou E, Drakopoulou M, Karakasis P, Apostolos A, Pamporis K, Tsioufis K, Tousoulis D. Cancer Therapy-Related Cardiac Dysfunction: A Review of Current Trends in Epidemiology, Diagnosis, and Treatment. Biomedicines 2024; 12:2914. [PMID: 39767820 PMCID: PMC11673750 DOI: 10.3390/biomedicines12122914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Cancer therapy-related cardiac dysfunction (CTRCD) has emerged as a significant concern with the rise of effective cancer treatments like anthracyclines and targeted therapies such as trastuzumab. While these therapies have improved cancer survival rates, their unintended cardiovascular side effects can lead to heart failure, cardiomyopathy, and arrhythmias. The pathophysiology of CTRCD involves oxidative stress, mitochondrial dysfunction, and calcium dysregulation, resulting in irreversible damage to cardiomyocytes. Inflammatory cytokines, disrupted growth factor signaling, and coronary atherosclerosis further contribute to this dysfunction. Advances in cardio-oncology have led to the early detection of CTRCD using cardiac biomarkers like troponins and imaging techniques such as echocardiography and cardiac magnetic resonance (CMR). These tools help identify asymptomatic patients at risk of cardiac events before the onset of clinical symptoms. Preventive strategies, including the use of cardioprotective agents like beta-blockers, angiotensin-converting enzyme inhibitors, mineralocorticoid receptor antagonists, and sodium-glucose cotransporter-2 inhibitors have shown promise in reducing the incidence of CTRCD. This review summarizes the mechanisms, detection methods, and emerging treatments for CTRCD, emphasizing the importance of interdisciplinary collaboration between oncologists and cardiologists to optimize care and improve both cancer and cardiovascular outcomes.
Collapse
Affiliation(s)
- Panagiotis Theofilis
- 1st Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.T.); (P.K.V.); (M.D.); (A.A.); (K.P.); (K.T.)
| | - Panayotis K. Vlachakis
- 1st Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.T.); (P.K.V.); (M.D.); (A.A.); (K.P.); (K.T.)
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, Thoracic Diseases General Hospital Sotiria, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Maria Drakopoulou
- 1st Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.T.); (P.K.V.); (M.D.); (A.A.); (K.P.); (K.T.)
| | - Paschalis Karakasis
- 2nd Department of Cardiology, Hippokration General Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece;
| | - Anastasios Apostolos
- 1st Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.T.); (P.K.V.); (M.D.); (A.A.); (K.P.); (K.T.)
| | - Konstantinos Pamporis
- 1st Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.T.); (P.K.V.); (M.D.); (A.A.); (K.P.); (K.T.)
| | - Konstantinos Tsioufis
- 1st Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.T.); (P.K.V.); (M.D.); (A.A.); (K.P.); (K.T.)
| | - Dimitris Tousoulis
- 1st Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.T.); (P.K.V.); (M.D.); (A.A.); (K.P.); (K.T.)
| |
Collapse
|
19
|
Wang Y, Xu J, Xie Y, Zhou D, Guo M, Qin Y, Yi K, Tian J, You T. Interventions for prevention and treatment of trastuzumab-induced cardiotoxicity: an umbrella review of systematic reviews and meta-analyses. Front Pharmacol 2024; 15:1479983. [PMID: 39703393 PMCID: PMC11655193 DOI: 10.3389/fphar.2024.1479983] [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: 08/13/2024] [Accepted: 11/14/2024] [Indexed: 12/21/2024] Open
Abstract
Background Trastuzumab therapy for HER2-positive cancers is associated with cardiotoxicity. This umbrella review synthesizes evidence from systematic reviews and meta-analyses on cardioprotective interventions during trastuzumab treatment. Methods A comprehensive search was conducted in PubMed, Embase, Cochrane Library, and Web of Science. Systematic reviews and meta-analyses examining cardioprotective interventions in patients receiving trastuzumab were included. The methodological quality was assessed using the AMSTAR-2 tool. Data on cardiac events, treatment interruptions, left ventricular ejection fraction (LVEF) changes, and exercise interventions were synthesized. Results Ten systematic reviews met the inclusion criteria. Statins demonstrated the strongest cardioprotective effect (RR = 0.47, 95% CI: 0.26-0.84), potentially preventing more than half of cardiac events during trastuzumab therapy, followed by beta-blockers (RR = 0.61, 95% CI: 0.39-0.93). Beta-blockers and ACEIs effectively reduced treatment interruptions, enabling approximately 40% more patients to maintain treatment continuity (RR = 0.63, 95% CI: 0.47-0.86). Among non-pharmacological interventions, structured exercise programs showed significant benefits in preserving cardiac function, demonstrating meaningful improvements in resting LVEF (WMD = -3.27%, 95% CI: -5.86 to -0.68). Discussion This review demonstrates that cardioprotective interventions, particularly statins and beta-blockers, significantly reduce the risk of cardiac complications during trastuzumab therapy. The positive impact on cardiac events and treatment interruptions suggests these interventions may enhance overall treatment efficacy by allowing more patients to complete their prescribed course. Conclusion Evidence strongly supports the systematic implementation of cardioprotective strategies in clinical practice, particularly statins and beta-blockers, as part of routine care protocols for patients receiving trastuzumab therapy. These interventions demonstrate significant potential in preventing cardiac complications and maintaining treatment continuity. Further research should focus on optimizing personalized approaches and evaluating long-term outcomes.
Collapse
Affiliation(s)
- Yunfang Wang
- Department of Endocrinology, Gansu Provincial Hospital, Lanzhou, China
| | - Jianguo Xu
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Yafei Xie
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Dan Zhou
- Department of Gastroenterology and Hepatology, Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, West China Hospital, Sichuan University, Chengdu, China
| | - Mingyue Guo
- Department of Obstetrics, Xiangya Hospital Central South University, Changsha, China
| | - Yu Qin
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Kang Yi
- Department of Cardiovascular Surgery, Gansu Provincial Hospital, Lanzhou, China
- Gansu International Scientific and Technological Cooperation Base of Diagnosis and Treatment of Congenital Heart Disease, Lanzhou, China
| | - Jinhui Tian
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- Key Laboratory of Evidence-based Medicine of Gansu Province, Lanzhou University, Lanzhou, China
| | - Tao You
- Department of Cardiovascular Surgery, Gansu Provincial Hospital, Lanzhou, China
- Gansu International Scientific and Technological Cooperation Base of Diagnosis and Treatment of Congenital Heart Disease, Lanzhou, China
| |
Collapse
|
20
|
Al-Hasnawi Z, Hasan HM, Abdul Azeez JM, Kadhim N, Shimal AA, Sadeq MH, Mahood NAHAM, Al-Qara Ghuli AAN, Hussein AS, Prajjwal P, Jain H, Goyal A, Amir O. Cardioprotective strategies in the management of chemotherapy-induced cardiotoxicity: current approaches and future directions. Ann Med Surg (Lond) 2024; 86:7212-7220. [PMID: 39649884 PMCID: PMC11623821 DOI: 10.1097/ms9.0000000000002668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/09/2024] [Indexed: 12/11/2024] Open
Abstract
BACKGROUND Chemotherapy-induced cardiotoxicity (CIC) is a significant challenge in cancer treatment, leading to heart failure and myocardial infarction. With rising cancer survival rates, the long-term cardiovascular health of survivors has gained importance. While several cardioprotective medications have been studied to mitigate chemotherapy's harmful effects on the heart, more research is needed to confirm their effectiveness and optimal use. METHODOLOGY This review synthesizes evidence on cardioprotective drugs in managing CIC. The authors conducted a comprehensive literature search of peer-reviewed articles, clinical trials, and meta-analyses published between January 2000 and May 2024. Studies were selected based on relevance, quality, and focus on mechanisms, efficacy, and clinical outcomes of cardioprotective agents such as beta-blockers, ACE inhibitors, ARBs, statins, and dexrazoxane. RESULTS AND DISCUSSION Cardioprotective medications show potential in alleviating the impact of chemotherapy on heart function. Beta-blockers and ACE inhibitors effectively reduce heart failure incidence and improve cardiac outcomes. Statins, with their anti-inflammatory and antioxidative properties, and dexrazoxane, which reduces anthracycline-induced cardiotoxicity, also show promise. However, variability in study designs, patient groups, and chemotherapy treatments complicates the establishment of standardized treatment protocols. CONCLUSION Cardioprotective drugs hold significant promise in managing CIC and improving cardiac outcomes for cancer patients. Current evidence supports the efficacy of beta-blockers, ACE inhibitors, statins, and dexrazoxane. Further research is needed to establish standardized protocols, evaluate long-term safety, and optimize treatment timing. Integrating cardioprotective strategies into oncological care can enhance the quality of life and prognosis for cancer survivors.
Collapse
Affiliation(s)
| | | | | | - Naam Kadhim
- College of Medicine, University of Baghdad, Baghdad, Iraq
| | | | | | | | | | - Ahmed Safaa Hussein
- Ulyanovsk State University, Ulyanovsk State Medical University, Ulyanovsk, Russia
| | | | - Hritvik Jain
- All India Institute of Medical Sciences, Jodhpur, India
| | - Aman Goyal
- Department of Internal Medicine, Seth GS Medical College and KEM Hospital, Mumbai, India
| | - Omniat Amir
- Almanhal Academy for Science, Khartoum, Sudan
| |
Collapse
|
21
|
Marrazzo L, Meattini I, Becherini C, Salvestrini V, Visani L, Barletta G, Saieva C, Del Bene MR, Pilato G, Desideri I, Arilli C, Paoletti L, Russo S, Scoccianti S, Martella F, Francolini G, Simontacchi G, Nori Cucchiari J, Pellegrini R, Livi L, Pallotta S. Subclinical cardiac damage monitoring in breast cancer patients treated with an anthracycline-based chemotherapy receiving left-sided breast radiation therapy: subgroup analysis from a phase 3 trial. LA RADIOLOGIA MEDICA 2024; 129:1926-1935. [PMID: 39436588 DOI: 10.1007/s11547-024-01897-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 10/05/2024] [Indexed: 10/23/2024]
Abstract
OBJECTIVE This study, derived from the phase 3 SAFE trial (ClinicalTrials.gov identifier: NCT2236806), explores subclinical cardiac damage in breast cancer patients receiving anthracycline-based chemotherapy and left-sided breast radiation therapy (RT). MATERIALS AND METHODS Eligible patients were randomized to a cardioprotective pharmacological therapy (bisoprolol, ramipril, or both) or placebo, with cardiac surveillance at multiple time-point using standard and 3-dimensional echocardiography. Dosimetric parameters were analysed, including mean heart dose (MHD) and various metrics for heart substructures, employing advanced contouring techniques and auto-contouring software. RESULTS In the analysis of left-sided breast RT patients, the study encompassed 39 out of 46 irradiated individuals, focusing on GLS and 3D-LVEF outcomes with ≥ 10% worsening, defined as subclinical heart damage. Distinct RT schedules were used, with placebo exhibiting the highest ≥ 10% worsening (36.4%). In terms of treatment arms, bisoprolol exhibited 11.1% worsening, while ramipril 16.7% and bisoprolol + ramipril 25%. For patients with no subclinical damage, the mean MHD was 1.5 Gy; for patients with subclinical heart damage, the mean MHD was 1.6 Gy (p = 0.94). Dosimetric parameters related to heart and heart substructures (left anterior descending artery, right and left atrium, right and left ventricle) showed no statistically significant differences between patients with and without subclinical damage. CONCLUSION Our results emphasize the crucial role of cardioprotective measures in mitigating adverse effects, highlighting RT as having negligible influence on cardiac performance. An extended follow-up assessment of the whole series is warranted to determine whether a subclinical effect could significantly influence clinical outcomes and cardiac events.
Collapse
Affiliation(s)
- Livia Marrazzo
- Department of Experimental and Clinical Biomedical Sciences "M. Serio", University of Florence, Florence, Italy
- Medical Physics Unit, Oncology Department, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Icro Meattini
- Department of Experimental and Clinical Biomedical Sciences "M. Serio", University of Florence, Florence, Italy.
- Radiation Oncology and Breast Unit, Oncology Department, Azienda Ospedaliero-Universitaria Careggi, Viale Morgagni 50, 50134, Florence, Italy.
| | - Carlotta Becherini
- Radiation Oncology and Breast Unit, Oncology Department, Azienda Ospedaliero-Universitaria Careggi, Viale Morgagni 50, 50134, Florence, Italy
| | - Viola Salvestrini
- Radiation Oncology and Breast Unit, Oncology Department, Azienda Ospedaliero-Universitaria Careggi, Viale Morgagni 50, 50134, Florence, Italy
| | - Luca Visani
- Radiation Oncology and Breast Unit, Oncology Department, Azienda Ospedaliero-Universitaria Careggi, Viale Morgagni 50, 50134, Florence, Italy
| | - Giuseppe Barletta
- CardioThoracic and Vascular Department, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Calogero Saieva
- Cancer Risk Factors and Lifestyle Epidemiology Unit, Institute for Cancer Research Prevention and Clinical Network (ISPRO), Florence, Italy
| | - Maria Riccarda Del Bene
- CardioThoracic and Vascular Department, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Giuseppe Pilato
- CardioThoracic and Vascular Department, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Isacco Desideri
- Department of Experimental and Clinical Biomedical Sciences "M. Serio", University of Florence, Florence, Italy
- Radiation Oncology and Breast Unit, Oncology Department, Azienda Ospedaliero-Universitaria Careggi, Viale Morgagni 50, 50134, Florence, Italy
| | - Chiara Arilli
- Medical Physics Unit, Oncology Department, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Lisa Paoletti
- Breast Unit, Oncology Department, Azienda USL Toscana Centro, Florence, Italy
| | - Serenella Russo
- Breast Unit, Oncology Department, Azienda USL Toscana Centro, Florence, Italy
| | - Silvia Scoccianti
- Breast Unit, Oncology Department, Azienda USL Toscana Centro, Florence, Italy
| | - Francesca Martella
- Breast Unit, Oncology Department, Azienda USL Toscana Centro, Florence, Italy
| | - Giulio Francolini
- Radiation Oncology and Breast Unit, Oncology Department, Azienda Ospedaliero-Universitaria Careggi, Viale Morgagni 50, 50134, Florence, Italy
| | - Gabriele Simontacchi
- Radiation Oncology and Breast Unit, Oncology Department, Azienda Ospedaliero-Universitaria Careggi, Viale Morgagni 50, 50134, Florence, Italy
| | - Jacopo Nori Cucchiari
- Radiation Oncology and Breast Unit, Oncology Department, Azienda Ospedaliero-Universitaria Careggi, Viale Morgagni 50, 50134, Florence, Italy
| | | | - Lorenzo Livi
- Department of Experimental and Clinical Biomedical Sciences "M. Serio", University of Florence, Florence, Italy
- Radiation Oncology and Breast Unit, Oncology Department, Azienda Ospedaliero-Universitaria Careggi, Viale Morgagni 50, 50134, Florence, Italy
| | - Stefania Pallotta
- Department of Experimental and Clinical Biomedical Sciences "M. Serio", University of Florence, Florence, Italy
- Medical Physics Unit, Oncology Department, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| |
Collapse
|
22
|
Bhatti AW, Patel R, Dani SS, Khadke S, Makwana B, Lessey C, Shah J, Al-Husami Z, Yang EH, Thavendiranathan P, Neilan TG, Sadler D, Cheng RK, Dent SF, Liu J, Lopez-Fernandez T, Herrmann J, Scherrer-Crosbie M, Lenihan DJ, Hayek SS, Ky B, Deswal A, Barac A, Nohria A, Ganatra S. SGLT2i and Primary Prevention of Cancer Therapy-Related Cardiac Dysfunction in Patients With Diabetes. JACC CardioOncol 2024; 6:863-875. [PMID: 39801650 PMCID: PMC11711834 DOI: 10.1016/j.jaccao.2024.08.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 08/03/2024] [Indexed: 01/16/2025] Open
Abstract
Background Specific cancer treatments can lead to cancer therapy-related cardiac dysfunction (CTRCD). Sodium glucose cotransporter-2 inhibitors (SGLT2is) can potentially prevent these cardiotoxic effects. Objectives This study sought to determine whether SGLT2i use is associated with a lower incidence of CTRCD in patients with type 2 diabetes mellitus (T2DM) and cancer, exposed to potentially cardiotoxic antineoplastic agents, and without a prior documented history of cardiomyopathy or heart failure. Methods We conducted a retrospective analysis of patients aged ≥18 years within the TriNetX database with T2DM, cancer, exposure to cardiotoxic therapies, and no prior documented history of cardiomyopathy or heart failure. Patients were categorized by SGLT2i use. After propensity score matching, outcomes were compared over 12 months using Cox proportional HRs. Subgroup analyses focusing on different cancer therapy classes were performed. Results The study included 8,675 propensity-matched patients in each cohort (mean age = ∼65 years, 42% females, 71% White, ∼19% gastrointestinal malignancy, and ∼25% anthracyclines). Patients prescribed SGLT2is had a lower risk of developing CTRCD (HR: 0.76: 95% CI: 0.69-0.84). SGLT2is also reduced heart failure exacerbations (HR: 0.81; 95% CI: 0.72-0.90), all-cause mortality (HR: 0.67; 95% CI: 0.61-0.74), and all-cause hospitalizations/emergency department visits (HR: 0.93; 95% CI: 0.89-0.97). Subgroup analyses also demonstrated reduced CTRCD risk across various classes of cancer therapies in patients prescribed SGLT2is. Conclusions SGLT2i administration was associated with a significantly decreased risk of developing CTRCD in patients with T2DM and cancer.
Collapse
Affiliation(s)
- Ammar W. Bhatti
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Beth Israel Lahey Health, Burlington, Massachusetts, USA
| | - Rushin Patel
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Beth Israel Lahey Health, Burlington, Massachusetts, USA
| | - Sourbha S. Dani
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Beth Israel Lahey Health, Burlington, Massachusetts, USA
| | - Sumanth Khadke
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Beth Israel Lahey Health, Burlington, Massachusetts, USA
| | - Bhargav Makwana
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Beth Israel Lahey Health, Burlington, Massachusetts, USA
| | - Candace Lessey
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Beth Israel Lahey Health, Burlington, Massachusetts, USA
| | - Jui Shah
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Beth Israel Lahey Health, Burlington, Massachusetts, USA
| | - Zaid Al-Husami
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Beth Israel Lahey Health, Burlington, Massachusetts, USA
| | - Eric H. Yang
- University of California-Los Angeles, Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California-Los Angeles, Los Angeles, California, USA
| | - Paaladinesh Thavendiranathan
- Department of Medicine, Division of Cardiology, Ted Rogers Program in Cardiotoxicity Prevention, Peter Munk Cardiac Center, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Tomas G. Neilan
- Cardio-Oncology Program, Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Diego Sadler
- Cardio-Oncology Section, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Weston, Florida, USA
| | - Richard K. Cheng
- Division of Cardiology, University of Washington, Seattle, Washington, USA
| | - Susan F. Dent
- Duke Cancer Institute, Department of Medicine, Duke University, Durham, North Carolina
| | - Jennifer Liu
- Cardio-Oncology Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Teresa Lopez-Fernandez
- Division of Cardiology, Cardio-Oncology Unit, La Paz University Hospital, Hospital La Paz Institute for Health Research, Madrid, Spain
- Division of Cardiology, Quironsalud Madrid University Hospital, Madrid, Spain
| | - Joerg Herrmann
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Daniel J. Lenihan
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Salim S. Hayek
- Division of Cardiology, Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, USA
| | - Bonnie Ky
- Thalheimer Center for Cardio-Oncology, Abramson Cancer Center and Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Anita Deswal
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ana Barac
- Inova Schar Heart and Vascular Institute, Inova Schar Cancer Institute, Fairfax, Virginia, USA
| | - Anju Nohria
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sarju Ganatra
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Beth Israel Lahey Health, Burlington, Massachusetts, USA
| |
Collapse
|
23
|
Cronin M, Neiroukh D, Lowery A, Wijns W, Kerin M, Keane M, Blazkova S, Soliman O. Proposed framework regarding management of patients with breast cancer and anti-cancer treatment-related elevation in cardiac troponin. IJC HEART & VASCULATURE 2024; 55:101522. [PMID: 39498346 PMCID: PMC11532442 DOI: 10.1016/j.ijcha.2024.101522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/16/2024] [Accepted: 10/02/2024] [Indexed: 11/07/2024]
Abstract
Cardiac biomarkers are a vital component within the first edition of the European Society of Cardiology guidelines in Cardio-Oncology. Specifically, they are mentioned in the definition of mild asymptomatic cancer therapy-related cardiac dysfunction, where left ventricular systolic function is ≥50 % with two outcomes; either a new decrease in global longitudinal strain >15 % from baseline and/or a new rise in cardiac biomarkers above the defined 99th percentile cut off values. Cardiac troponin is one such biomarker. Many of the treatments for breast cancer have published data on cardiac dysfunction and/or cardiovascular toxicity, and such may lead to an elevation in cardiac troponin. However, there is conflicting and incomplete data regarding how to approach an elevated cardiac troponin during anti-cancer treatment, which has confounded patient care in the clinical trial setting. We propose a novel framework to guide physicians in treatment-related elevation of cardiac troponin in the breast cancer population. Secondly, the additive role which the recommendation that cardiac troponin carries within mild asymptomatic definitions of CTRCD is the subject of great debate. We suggest a reflection on the role of biomarkers, specifically in reference to cardiac troponin.
Collapse
Affiliation(s)
- Michael Cronin
- University of Galway, School of Medicine, Galway, Republic of Ireland
| | - Dina Neiroukh
- University of Galway, School of Medicine, Galway, Republic of Ireland
| | - Aoife Lowery
- University of Galway, School of Medicine, Galway, Republic of Ireland
| | - William Wijns
- University of Galway, School of Medicine, Galway, Republic of Ireland
| | - Michael Kerin
- University of Galway, School of Medicine, Galway, Republic of Ireland
| | - Maccon Keane
- University of Galway, School of Medicine, Galway, Republic of Ireland
| | - Silvie Blazkova
- University of Galway, School of Medicine, Galway, Republic of Ireland
| | - Osama Soliman
- University of Galway, School of Medicine, Galway, Republic of Ireland
- Netherlands: Euro Heart Foundation, Netherlands
| |
Collapse
|
24
|
Gong FF, Grunblatt E, Voss WB, Rangarajan V, Raissi S, Chow K, Jafari L, Patel NP, Vaitenas I, Marion M, Ramirez H, Zhao M, Andrei AC, Baldridge AS, Murtagh G, Maganti K, Rigolin VH, Akhter N. A strain-guided trial of cardioprotection in early-stage breast cancer patients on anti-HER2 therapy (PROTECT HER2). CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:85. [PMID: 39605014 PMCID: PMC11600554 DOI: 10.1186/s40959-024-00291-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND Global longitudinal strain (GLS) has been used to identify patients at risk for cancer-therapy related cardiac dysfunction (CTRCD). However, there is limited data on the effectiveness of initiating cardioprotective therapy based on a strain-guided strategy in early stage HER2+ breast cancer patients. This randomized clinical trial assessed if treatment with carvedilol based on a strain-guided strategy can prevent development of CTRCD in HER2+ breast cancer patients on non-anthracycline based regimens. METHODS Study participants were prospectively assigned to one of four arms. Patients with normal LVEF and GLS remained in Arm A. Patients whose GLS decreased by > 15% from baseline or to < -15% during follow up were randomized 1:1 to prophylactic carvedilol (Arm B) or no therapy (Arm C). Patients who developed CTRCD were assigned to Arm D. The primary endpoint was GLS stability. The secondary endpoints were development of CTRCD and rate of anti-HER2 treatment interruption. RESULTS Among 110 patients who completed follow up, 84 were assigned to Arm A, 10 each were randomized to Arms B or C, and 6 were assigned to Arm D. At the end of the study period, there were no significant differences in GLS stability, development of CTRCD, or number of cancer therapy cycles completed between patients who did and did not receive cardioprotective therapy. CONCLUSIONS In this prospective randomized GLS-guided study of prophylactic carvedilol in early stage HER2+ breast cancer patients on non-anthracycline regimens, there were no significant difference between groups in GLS stability, CTRCD or trastuzumab cycles held. These findings may identify a low-risk group of patients who may be considered for less intensive cardiac surveillance. TRIAL REGISTRATION https://clinicaltrials.gov/study/NCT02993198 . Start date: 4/2015. This trial included patients who were retrospectively registered.
Collapse
Affiliation(s)
- Fei Fei Gong
- Division of Cardiovascular Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, 676 N. St. Clair, Suite 600, Chicago, IL, 60611, USA
| | - Eli Grunblatt
- Department of Medicine, Feinberg School of Medicine, Northwestern University, 676 N. St. Clair, Suite 2330, Chicago, IL, 60611, USA
| | - Woo Bin Voss
- Division of Cardiovascular Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, 676 N. St. Clair, Suite 600, Chicago, IL, 60611, USA
| | - Vibhav Rangarajan
- Division of Cardiovascular Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, 676 N. St. Clair, Suite 600, Chicago, IL, 60611, USA
| | - Sasan Raissi
- Division of Cardiovascular Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, 676 N. St. Clair, Suite 600, Chicago, IL, 60611, USA
| | - Kimberly Chow
- Department of Medicine, Feinberg School of Medicine, Northwestern University, 676 N. St. Clair, Suite 2330, Chicago, IL, 60611, USA
| | - Lua Jafari
- Department of Medicine, Feinberg School of Medicine, Northwestern University, 676 N. St. Clair, Suite 2330, Chicago, IL, 60611, USA
| | - Nikita P Patel
- Department of Medicine, Feinberg School of Medicine, Northwestern University, 676 N. St. Clair, Suite 2330, Chicago, IL, 60611, USA
| | - Inga Vaitenas
- Division of Cardiovascular Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, 676 N. St. Clair, Suite 600, Chicago, IL, 60611, USA
| | - Milica Marion
- Division of Cardiovascular Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, 676 N. St. Clair, Suite 600, Chicago, IL, 60611, USA
| | - Haydee Ramirez
- Division of Cardiovascular Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, 676 N. St. Clair, Suite 600, Chicago, IL, 60611, USA
| | - Manyun Zhao
- Division of Cardiovascular Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, 676 N. St. Clair, Suite 600, Chicago, IL, 60611, USA
| | - Adin-Christian Andrei
- Division of Cardiovascular Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, 676 N. St. Clair, Suite 600, Chicago, IL, 60611, USA
| | - Abigail S Baldridge
- Division of Cardiovascular Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, 676 N. St. Clair, Suite 600, Chicago, IL, 60611, USA
| | - Gillian Murtagh
- Abbott Laboratories, 675 N. Field Drive, Lake Forest, IL, 60045, USA
| | - Kameswari Maganti
- Division of Cardiovascular Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, 676 N. St. Clair, Suite 600, Chicago, IL, 60611, USA
| | - Vera H Rigolin
- Division of Cardiovascular Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, 676 N. St. Clair, Suite 600, Chicago, IL, 60611, USA
| | - Nausheen Akhter
- Division of Cardiovascular Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, 676 N. St. Clair, Suite 600, Chicago, IL, 60611, USA.
| |
Collapse
|
25
|
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
|
26
|
Moreno-Arciniegas A, García A, Kelm M, D'Amore F, da Silva MG, Sánchez-González J, Sánchez PL, López-Fernández T, Córdoba R, Asteggiano R, Camus V, Smink J, Ferreira A, Kersten MJ, Bolaños N, Escalera N, Pacella E, Gómez-Talavera S, Quesada A, Rosselló X, Ibanez B. Rationale and design of RESILIENCE: A prospective randomized clinical trial evaluating remote ischaemic conditioning for the prevention of anthracycline cardiotoxicity. Eur J Heart Fail 2024; 26:2213-2222. [PMID: 39212445 DOI: 10.1002/ejhf.3395] [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: 04/02/2024] [Revised: 07/01/2024] [Accepted: 07/09/2024] [Indexed: 09/04/2024] Open
Abstract
AIMS There is a lack of therapies able to prevent anthracycline cardiotoxicity (AC). Remote ischaemic conditioning (RIC) has shown beneficial effects in preclinical models of AC. METHODS REmote iSchemic condItioning in Lymphoma PatIents REceiving ANthraCyclinEs (RESILIENCE) is a multinational, prospective, phase II, double-blind, sham-controlled, randomized clinical trial that evaluates the efficacy and safety of RIC in lymphoma patients receiving anthracyclines. Patients scheduled to undergo ≥5 chemotherapy cycles including anthracyclines and with ≥1 AC-associated risk factors will be randomized to weekly RIC or sham throughout the chemotherapy period. Patients will undergo three multiparametric cardiac magnetic resonance (CMR) studies, at baseline, after the third cycle (intermediate CMR), and 2 months after the end of chemotherapy. Thereafter, patients will be followed up for clinical events over an anticipated median of ≥24 months. The primary endpoint is the absolute change from baseline in CMR-based left ventricular ejection fraction (LVEF). The main secondary outcome is the incidence of AC events, defined as (1) a drop in CMR-based LVEF of ≥10 absolute points, or (2) a drop in CMR-based LVEF of ≥5 and <10 absolute points to a value <50%. Intermediate CMR will test the ability of T2 mapping to predict AC versus classical markers (left ventricular strain and cardiac injury biomarkers). A novel CMR sequence allowing ultrafast cine acquisition will be validated in this vulnerable population. CONCLUSIONS The RESILIENCE trial will test RIC (a novel non-invasive intervention to prevent AC) in a cohort of high-risk patients. The trial will also test candidate markers for their capacity to predict AC and will validate a novel CMR sequence reducing acquisition time in a vulnerable population.
Collapse
Affiliation(s)
| | | | - Malte Kelm
- Department of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Düsseldorf, Düsseldorf, Germany
| | | | - María Gomes da Silva
- Hematology, Instituto Português de Oncologia de Lisboa (IPO Lisboa), Lisbon, Portugal
| | | | - Pedro L Sánchez
- Hospital Universitario de Salamanca, Salamanca, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Teresa López-Fernández
- Cardiology Department, La Paz University Hospital, IdiPAZ Research Institute, Madrid, Spain
| | | | - Riccardo Asteggiano
- Faculty of Medicine, Insubria University, Varese, and Laboratorio Analisi e Ricerca Clinica, Turin, Italy
| | - Vincent Camus
- Centre Henri Becquerel, Department of Hematology and INSERM U1245, Rouen, France
| | - Jouke Smink
- Department of MR R&D-Clinical Science, Philips, Best, The Netherlands
| | | | - Marie J Kersten
- Department of Hematology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Noemi Escalera
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | | | - Sandra Gómez-Talavera
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- IIS-Fundación Jiménez Díaz, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Antonio Quesada
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Xavier Rosselló
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Hospital Universitari Son Espases-IDISBA, Universitat Illes Balears, Palma de Mallorca, Spain
| | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- IIS-Fundación Jiménez Díaz, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| |
Collapse
|
27
|
Austin D, Maier RH, Akhter N, Sayari M, Ogundimu E, Maddox JM, Vahabi S, Humphreys AC, Graham J, Oxenham H, Haney S, Cresti N, Verrill M, Osborne W, Wright KL, Goranova R, Bailey JR, Kalakonda N, Macheta M, Kilner MF, Young ME, Morley NJ, Neelakantan P, Gilbert G, Thomas BK, Graham RJ, Fujisawa T, Mills NL, Hildreth V, Prichard J, Kasim AS, Hancock HC, Plummer C. Preventing Cardiac Damage in Patients Treated for Breast Cancer and Lymphoma: The PROACT Clinical Trial. JACC CardioOncol 2024; 6:684-696. [PMID: 39479329 PMCID: PMC11520224 DOI: 10.1016/j.jaccao.2024.07.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/10/2024] [Accepted: 07/12/2024] [Indexed: 11/02/2024] Open
Abstract
Background Cardiotoxicity is a concern for cancer survivors undergoing anthracycline chemotherapy. Enalapril has been explored for its potential to mitigate cardiotoxicity in cancer patients. The dose-dependent cardiotoxicity effects of anthracyclines can be detected early through the biomarker cardiac troponin. Objectives The PROACT (Preventing Cardiac Damage in Patients Treated for Breast Cancer and Lymphoma) clinical trial assessed the effectiveness of enalapril in preventing cardiotoxicity, manifesting as myocardial injury and cardiac function impairment, in patients undergoing high-dose anthracycline-based chemotherapy for breast cancer or non-Hodgkin lymphoma. Methods This prospective, multicenter, open-label, randomized controlled trial employed a superiority design with observer-blinded endpoints. A total of 111 participants, scheduled for 6 cycles of chemotherapy with a planned dose of ≥300 mg/m2 doxorubicin equivalents, were randomized to receive either enalapril (titrated up to 20 mg daily) or standard care without enalapril. Results Myocardial injury, indicated by cardiac troponin T (≥14 ng/L), during and 1 month after chemotherapy, was observed in 42 (77.8%) of 54 patients in the enalapril group vs 45 (83.3%) of 54 patients in the standard care group (OR: 0.65; 95% CI: 0.23-1.78). Injury detected by cardiac troponin I (>26.2 ng/L) occurred in 25 (47.2%) of 53 patients on enalapril compared with 24 (45.3%) of 53 in standard care (OR: 1.10; 95% CI: 0.50-2.38). A relative decline of more than 15% from baseline in left ventricular global longitudinal strain was observed in 10 (21.3%) of 47 patients on enalapril and 9 (21.9%) of 41 in standard care (OR: 0.95; 95% CI: 0.33-2.74). An absolute decline of >10% to <50% in left ventricular ejection fraction was seen in 2 (4.1%) of 49 patients on enalapril vs none in patients in standard care. Conclusions Adding enalapril to standard care during chemotherapy did not prevent cardiotoxicity in patients receiving high-dose anthracycline-based chemotherapy. (PROACT: Can we prevent Chemotherapy-related Heart Damage in Patients With Breast Cancer and Lymphoma?; NCT03265574).
Collapse
Affiliation(s)
- David Austin
- Academic Cardiovascular Unit, James Cook University Hospital, South Tees Hospitals NHS Foundation Trust, Middlesbrough, United Kingdom
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Rebecca H. Maier
- Academic Cardiovascular Unit, James Cook University Hospital, South Tees Hospitals NHS Foundation Trust, Middlesbrough, United Kingdom
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Nasima Akhter
- School of Health and Life Sciences, Teesside University, Middlesbrough, United Kingdom
| | - Mohammad Sayari
- Department of Mathematical Science, Durham University, Durham, United Kingdom
| | - Emmanuel Ogundimu
- Department of Mathematical Science, Durham University, Durham, United Kingdom
| | - Jamie M. Maddox
- Department of Haematology, South Tees Hospitals NHS Foundation Trust, Middlesbrough, United Kingdom
| | - Sharareh Vahabi
- Bristol Heart Institute, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, United Kingdom
| | - Alison C. Humphreys
- Cancer Services, South Tees Hospitals NHS Foundation Trust, Middlesbrough, United Kingdom
| | - Janine Graham
- Cancer Services, South Tees Hospitals NHS Foundation Trust, Middlesbrough, United Kingdom
- Cancer Services, North Tees and Hartlepool NHS Foundation Trust, Stockton-on-Tees, United Kingdom
| | - Helen Oxenham
- Department of Cardiology, North Tees and Hartlepool NHS Foundation Trust, Stockton-on-Tees, United Kingdom
| | - Sophie Haney
- Cancer Services, South Tees Hospitals NHS Foundation Trust, Middlesbrough, United Kingdom
- Cancer Services, County Durham and Darlington NHS Foundation Trust, Darlington, United Kingdom
| | - Nicola Cresti
- Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Mark Verrill
- Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Wendy Osborne
- Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Kathryn L. Wright
- Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
- Cancer Services, South Tyneside and Sunderland NHS Foundation Trust, Sunderland, United Kingdom
| | - Rebecca Goranova
- Department of Oncology, Derriford Hospital, University Hospitals Plymouth NHS Trust, Plymouth, United Kingdom
| | - James R. Bailey
- Department of Haematology, Hull University Teaching Hospitals NHS Trust, Queen’s Centre for Oncology and Haematology, Castle Hill Hospital, Hull, United Kingdom
| | - Nagesh Kalakonda
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Mac Macheta
- Department of Haematology, Blackpool Teaching Hospitals, Blackpool, United Kingdom
| | - Mari F. Kilner
- Pathology Services, Northumbria Healthcare NHS Foundation Trust, North Shields, United Kingdom
| | - Moya E. Young
- Department of Haematology, East Kent Hospitals University NHS Foundation Trust, Canterbury, United Kingdom
| | - Nick J. Morley
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
| | - Pratap Neelakantan
- Department of Haematology, Royal Berkshire NHS Foundation Trust, Reading, United Kingdom
| | - Georgia Gilbert
- Academic Cardiovascular Unit, James Cook University Hospital, South Tees Hospitals NHS Foundation Trust, Middlesbrough, United Kingdom
| | - Byju K. Thomas
- Academic Cardiovascular Unit, James Cook University Hospital, South Tees Hospitals NHS Foundation Trust, Middlesbrough, United Kingdom
| | - Richard J. Graham
- Academic Cardiovascular Unit, James Cook University Hospital, South Tees Hospitals NHS Foundation Trust, Middlesbrough, United Kingdom
| | - Takeshi Fujisawa
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Nicholas L. Mills
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Victoria Hildreth
- Newcastle Clinical Trials Unit, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Jonathan Prichard
- Newcastle Clinical Trials Unit, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Adetayo S. Kasim
- Department of Anthropology, Durham University, Durham, United Kingdom
| | - Helen C. Hancock
- Newcastle Clinical Trials Unit, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Chris Plummer
- Department of Cardiology, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
- Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
28
|
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: 9] [Impact Index Per Article: 9.0] [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
|
29
|
Nakatsuma K, Ozasa N, Ohno M, Ishiguro H, Minami M, Nishi E, Toi M, Ono K, Kimura T. Subclinical myocardial damage after anthracycline chemotherapy in Japanese patients with breast cancer. J Cardiol 2024; 84:260-265. [PMID: 38521118 DOI: 10.1016/j.jjcc.2024.03.005] [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: 06/21/2023] [Revised: 03/14/2024] [Accepted: 03/18/2024] [Indexed: 03/25/2024]
Abstract
BACKGROUND Data on the incidence, timing, and severity of myocardial damage after anthracycline-based chemotherapy (AC) in Japanese patients with breast cancer are limited. METHOD We evaluated cancer therapy-related cardiac dysfunction (CTRCD) in Japanese women with breast cancer (n = 51) after the first AC according to the definitions of the 2022 European Society of Cardiology onco-cardiology guideline, including assessment of high-sensitivity troponin I (TnI) and B-type natriuretic peptide (BNP) levels. RESULTS CTRCD was detected in 67 % of the patients (3.9 %, 7.8 %, 9.8 %, 43 %, 37 %, 22 %, 20 %, and 9.8 % of patients at 1 week and 1, 2, 3, 6, 9, 12, and 15 months post-AC, respectively) without significant left ventricular ejection fraction reduction (<50 %) and heart failure. Elevated TnI levels (>26 pg/mL) were found in 43 % of patients, and elevated BNP levels (≥35 pg/mL) were observed in 22 % of patients during the follow-up period. CONCLUSIONS Approximately two-thirds of the Japanese patients in this study experienced CTRCD, which was frequently observed at 3 or 6 months post-AC. However, all patients with CTRCD were diagnosed with mild asymptomatic CTRCD. Although, these patients were diagnosed with mild asymptomatic CTRCD, careful long-term follow-up will be required.
Collapse
Affiliation(s)
- Kenji Nakatsuma
- Department of Cardiology, Mitsubishi Kyoto Hospital, Kyoto, Japan
| | - Neiko Ozasa
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Mikiko Ohno
- Department of Pharmacology, Shiga University of Medical Science, Shiga, Japan
| | - Hiroshi Ishiguro
- Department of Breast Oncology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Manabu Minami
- Department of Data Science, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Eiichiro Nishi
- Department of Pharmacology, Shiga University of Medical Science, Shiga, Japan
| | - Masakazu Toi
- Department of Breast Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koh Ono
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takeshi Kimura
- Department of Cardiology, Hirakata Kohsai Hospital, Hirakata, Japan
| |
Collapse
|
30
|
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
|
31
|
Camilli M, Cipolla CM, Dent S, Minotti G, Cardinale DM. Anthracycline Cardiotoxicity in Adult Cancer Patients: JACC: CardioOncology State-of-the-Art Review. JACC CardioOncol 2024; 6:655-677. [PMID: 39479333 PMCID: PMC11520218 DOI: 10.1016/j.jaccao.2024.07.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 11/02/2024] Open
Abstract
Since their introduction in the 1960s, anthracyclines have been a significant breakthrough in oncology, introducing dramatic changes in the treatment of solid and hematologic malignancies. Although new-generation targeted drugs and cellular therapies are revolutionizing contemporary oncology, anthracyclines remain the cornerstone of treatment for lymphomas, acute leukemias, and soft tissue sarcomas. However, their clinical application is limited by a dose-dependent cardiotoxicity that can reduce cardiac performance and eventually lead to overt heart failure. The field of cardio-oncology has emerged to safeguard the cardiovascular health of cancer patients receiving these therapies. It focuses on controlling risk factors, implementing preventive strategies, ensuring appropriate surveillance, and managing complications. This state-of-the-art review summarizes the current indications for anthracyclines in modern oncology, explores recent evidence on pathophysiology and epidemiology, and discusses advances in cardioprotection measures in the anthracycline-treated patient. Additionally, it highlights key clinical challenges and research gaps in this area.
Collapse
Affiliation(s)
- Massimiliano Camilli
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Carlo Maria Cipolla
- Cardioncology Unit, Cardioncology and Second Opinion Division, European Institute of Oncology IRCCS, Milan, Italy
| | - Susan Dent
- Duke Cancer Institute, Duke University, Durham, North Carolina, USA
- Division of Medical Oncology, Duke Cancer Institute, Duke University, Durham, North Carolina, USA
| | - Giorgio Minotti
- Università e Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Daniela Maria Cardinale
- Cardioncology Unit, Cardioncology and Second Opinion Division, European Institute of Oncology IRCCS, Milan, Italy
| |
Collapse
|
32
|
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
|
33
|
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
|
34
|
Bews HJ, Mackic L, Jassal DS. Preventing broken hearts in women with breast cancer: a concise review on chemotherapy-mediated cardiotoxicity. Can J Physiol Pharmacol 2024; 102:487-497. [PMID: 38039515 DOI: 10.1139/cjpp-2023-0358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2023]
Abstract
Cancer and cardiovascular disease are the leading causes of death for Canadian women. One in eight Canadian women will receive the life-changing diagnosis of breast cancer (BC) in their lifetime, with 1 in 34 dying from the disease. Although doxorubicin (DOX) and trastuzumab (TRZ) have significantly improved survival in women diagnosed with human epidermal growth factor receptor 2 (HER2)-positive BC, approximately one in four women who receive this treatment are at risk of developing chemotherapy-induced cardiotoxicity. Cardiotoxicity is defined as a decline in left ventricular ejection fraction (LVEF) of >10% to an absolute value of <53%. Current guidelines recommend the serial monitoring of LVEF in this patient population using non-invasive cardiac imaging modalities including transthoracic echocardiography or multi-gated acquisition scan; however, this will only allow for the detection of established cardiotoxicity. Recent studies have demonstrated that a reduction in global longitudinal strain by speckle tracking echocardiography can identify pre-clinical systolic dysfunction prior to a decline in overall LVEF. Implementation of early detection techniques would allow for the prompt initiation of cardioprotective strategies. In addition to the early detection of chemotherapy-mediated cardiotoxicity, the prophylactic use of angiotensin-converting enzyme inhibitors, angiotensin receptor blockers, β-blockers, statins, exercise, and nutraceutical therapies have been studied in the setting of cardio-oncology.
Collapse
Affiliation(s)
- Hilary J Bews
- Section of Cardiology, Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Lana Mackic
- Institute of Cardiovascular Sciences, St. Boniface Hospital, University of Manitoba, Winnipeg, MB, Canada
| | - Davinder S Jassal
- Section of Cardiology, Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Institute of Cardiovascular Sciences, St. Boniface Hospital, University of Manitoba, Winnipeg, MB, Canada
- Department of Radiology, St. Boniface Hospital, University of Manitoba, Winnipeg, MB, Canada
- Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
35
|
Repp ML, Edwards MD, Burch CS, Rao A, Chinyere IR. PCSK9 Inhibitors and Anthracyclines: The Future of Cardioprotection in Cardio-Oncology. HEARTS 2024; 5:375-388. [PMID: 39268545 PMCID: PMC11391951 DOI: 10.3390/hearts5030027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024] Open
Abstract
The field of cardio-oncology is an expanding frontier within cardiovascular medicine, and the need for evidence-based guidelines is apparent. One of the emerging focuses within cardio-oncology is the concomitant use of medications for cardioprotection in the setting of chemotherapy regimens that have known cardiovascular toxicity. While clinical trials focusing on cardioprotection during chemotherapy are sparse, an inaugural trial exploring the prophylactic potential of Sodium-Glucose Cotransporter-2 inhibitors (SGLT2is) for anthracycline (ANT)-induced cardiotoxicity has recently commenced. Proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, though less studied in this oncology demographic, have exhibited promise in preclinical studies for conferring cardiac protection during non-ischemic toxic insults. While primarily used to reduce low-density lipoprotein, PCSK9 inhibitors exhibit pleiotropic effects, including the attenuation of inflammation, reactive oxygen species, and endothelial dysfunction. In ANT-induced cardiotoxicity, these same processes are accelerated, resulting in premature termination of treatment, chronic cardiovascular sequelae, heart failure, and/or death. This review serves a dual purpose: firstly, to provide a concise overview of the mechanisms implicated in ANT-induced cardiotoxicity, and, finally, to summarize the existing preclinical data supporting the theoretical possibility of the cardioprotective effects of PCSK9 inhibition in ANT-induced cardiotoxicity.
Collapse
Affiliation(s)
- Matthew L Repp
- Department of Medicine, University of Colorado, Aurora, CO 80045, USA
| | - Mark D Edwards
- Department of Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Christopher S Burch
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - Amith Rao
- Department of Medicine, Banner University Medicine, Tucson, AZ 85724, USA
| | - Ikeotunye Royal Chinyere
- Department of Medicine, Banner University Medicine, Tucson, AZ 85724, USA
- Sarver Heart Center, University of Arizona, 1501 North Campbell Avenue, Room 6154, Tucson, AZ 85724, USA
| |
Collapse
|
36
|
Felix N, Nogueira PC, Silva IM, Costa TA, Campello CA, Stecca C, Lopes RD. Cardio-protective effects of statins in patients undergoing anthracycline-based chemotherapy: An updated meta-analysis of randomized controlled trials. Eur J Intern Med 2024; 126:43-48. [PMID: 38643042 DOI: 10.1016/j.ejim.2024.04.007] [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: 10/24/2023] [Revised: 03/28/2024] [Accepted: 04/10/2024] [Indexed: 04/22/2024]
Abstract
INTRODUCTION Several interventions have been tested for cardio-protection against anthracycline-induced cancer therapy-related cardiovascular dysfunction (CTRCD). The role of statins in this setting remains unclear. METHODS We systematically searched PubMed, Embase, Cochrane Library, Clinicaltrials.gov, and Web of Science for randomized controlled trials (RCTs) comparing statins versus control (placebo or no intervention) for preventing anthracycline-induced CTRCD. We applied a random-effects model to pool risk ratios (RR) and mean differences (MD) with 95 % confidence intervals (CI). RESULTS We included seven RCTs comprising 887 patients with planned chemotherapy with anthracycline-based regimens, of whom 49.8 % were randomized to statins. Relative to placebo, statins significantly reduced the incidence of cardiotoxicity/CTRCD (RR 0.46; 95 % CI 0.29 to 0.72; p < 0.001). The left ventricular end-systolic volume was also lower in patients treated with statin (MD -3.12 mL; 95 % CI -6.13 to -0.12 mL; p = 0.042). There was no significant difference between groups in post-anthracycline left ventricular ejection fraction (LVEF) overall. CONCLUSION In this meta-analysis of RCTs, statins were significantly associated with a lower incidence of anthracycline-induced CTRCD and attenuated changes in the left ventricular end-systolic volume. Thus, our findings suggest that statins should be considered as a cardio-protection strategy for patients with planned anthracycline-based chemotherapy.
Collapse
Affiliation(s)
- Nicole Felix
- Federal University of Campina Grande, Campina Grande, Brazil
| | - Paula C Nogueira
- Hospital da Mulher, São Paulo, Brazil; Grupo Fleury, São Paulo, Brazil.
| | | | | | | | | | | |
Collapse
|
37
|
Kuang Z, Ge Y, Cao L, Wang X, Liu K, Wang J, Zhu X, Wu M, Li J. Precision Treatment of Anthracycline-Induced Cardiotoxicity: An Updated Review. Curr Treat Options Oncol 2024; 25:1038-1054. [PMID: 39066853 PMCID: PMC11329674 DOI: 10.1007/s11864-024-01238-9] [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: 06/19/2024] [Indexed: 07/30/2024]
Abstract
OPINION STATEMENT Anthracycline (ANT)-induced cardiotoxicity (AIC) is a particularly prominent form of cancer therapy-related cardiovascular toxicity leading to the limitations of ANTs in clinical practice. Even though AIC has drawn particular attention, the best way to treat it is remaining unclear. Updates to AIC therapy have been made possible by recent developments in research on the underlying processes of AIC. We review the current molecular pathways leading to AIC: 1) oxidative stress (OS) including enzymatic-induced and other mechanisms; 2) topoisomerase; 3) inflammatory response; 4) cardiac progenitor cell damage; 5) epigenetic changes; 6) renin-angiotensin-aldosterone system (RAAS) dysregulation. And we systematically discuss current prevention and treatment strategies and novel pathogenesis-based therapies for AIC: 1) dose reduction and change; 2) altering drug delivery methods; 3) antioxidants, dexrezosen, statina, RAAS inhibitors, and hypoglycemic drugs; 4) miRNA, natural phytochemicals, mesenchymal stem cells, and cardiac progenitor cells. We also offer a fresh perspective on the management of AIC by outlining the current dilemmas and challenges associated with its prevention and treatment.
Collapse
Affiliation(s)
- Ziyu Kuang
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 10053, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, 10029, China
| | - Yuansha Ge
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 10053, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, 10029, China
| | - Luchang Cao
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 10053, China
| | - Xinmiao Wang
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 10053, China
| | - Kexin Liu
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 10053, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, 10029, China
| | - Jiaxi Wang
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 10053, China
| | - Xiaojuan Zhu
- The 3rd affiliated hospital of Zhejiang Chinese Medical University, Hangzhou, 310005, China.
| | - Min Wu
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 10053, China.
| | - Jie Li
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 10053, China.
| |
Collapse
|
38
|
Szponar J, Niziński P, Dudka J, Kasprzak-Drozd K, Oniszczuk A. Natural Products for Preventing and Managing Anthracycline-Induced Cardiotoxicity: A Comprehensive Review. Cells 2024; 13:1151. [PMID: 38995002 PMCID: PMC11240786 DOI: 10.3390/cells13131151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 07/03/2024] [Accepted: 07/04/2024] [Indexed: 07/13/2024] Open
Abstract
Doxorubicin (DOX) is an anthracycline anticancer agent that is highly effective in the treatment of solid tumors. Given the multiplicity of mechanisms involved in doxorubicin-induced cardiotoxicity, it is difficult to identify a precise molecular target for toxicity. The findings of a literature review suggest that natural products may offer cardioprotective benefits against doxorubicin-induced cardiotoxicity, both in vitro and in vivo. However, further confirmatory studies are required to substantiate this claim. It is of the utmost importance to direct greater attention towards the intricate signaling networks that are of paramount importance for the survival and dysfunction of cardiomyocytes. Notwithstanding encouraging progress made in preclinical studies of natural products for the prevention of DOX-induced cardiotoxicity, these have not yet been translated for clinical use. One of the most significant obstacles hindering the development of cardioprotective adjuvants based on natural products is the lack of adequate bioavailability in humans. This review presents an overview of current knowledge on doxorubicin DOX-induced cardiotoxicity, with a focus on the potential benefits of natural compounds and herbal preparations in preventing this adverse effect. As literature search engines, the browsers in the Scopus, PubMed, Web of Science databases and the ClinicalTrials.gov register were used.
Collapse
Affiliation(s)
- Jarosław Szponar
- Clinical Department of Toxicology and Cardiology, Toxicology Clinic, Stefan Wyszyński Regional Specialist Hospital, Medical University of Lublin, 20-718 Lublin, Poland;
| | - Przemysław Niziński
- Department of Pharmacology, Medical University of Lublin, Radziwiłłowska 11 Street, 20-080 Lublin, Poland;
| | - Jarosław Dudka
- Chair and Department of Toxicology, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland;
| | - Kamila Kasprzak-Drozd
- Department of Inorganic Chemistry, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland;
| | - Anna Oniszczuk
- Department of Inorganic Chemistry, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland;
| |
Collapse
|
39
|
Sotiropoulou IM, Manetas-Stavrakakis N, Kourek C, Xanthopoulos A, Magouliotis D, Giamouzis G, Skoularigis J, Briasoulis A. Prevention of Anthracyclines and HER2 Inhibitor-Induced Cardiotoxicity: A Systematic Review and Meta-Analysis. Cancers (Basel) 2024; 16:2419. [PMID: 39001481 PMCID: PMC11240691 DOI: 10.3390/cancers16132419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
BACKGROUND This meta-analysis and systematic review aim to consolidate evidence on cardiotoxicity prevention and treatment strategies in patients receiving anthracyclines or HER2 receptor inhibitors, vital treatments for breast cancer and hematologic malignancies. By synthesizing existing research, the goal is to provide impactful insights that enhance patient care and outcomes. METHODS Comprehensive research across PubMed, Scopus, EMBASE, and the Cochrane Central Register for Controlled Trials was conducted, selecting clinical trials focusing on cardioprotection in anthracyclines or HER2 inhibitor-treated individuals. Effect sizes were computed using OpenMeta (Analyst), with leave-out meta-analysis to assess potential small study effects. Meta-regression explored treatment duration and sample size effects. Evidence quality for primary outcomes was evaluated using ROB, Robins 2, and Newcastle-Ottawa tools. RESULTS Twenty -three studies involving a total of 14,652 patients (13,221 adults and 1431 kids) were included in the current systematic review and meta-analysis. The risk of bias and methodological quality of the included studies suggested good and moderate quality. Patients prescribed β-blockers demonstrated a 74% lower likelihood of exhibiting cardiotoxicity symptoms (OR 1.736). Similarly, the use of dexrazoxane was linked to a threefold decrease in cardiac abnormalities risk (OR 2.989), and ACE inhibitor administration showed half the risk compared with the control group (OR 1.956). CONCLUSIONS Through this systematic review and meta-analysis, it was shown that there is a reduction in cardiotoxicity from either anthracyclines or HER2 inhibitors in patients receiving pharmacoprophylaxis.
Collapse
Affiliation(s)
- Ioanna Myrto Sotiropoulou
- Department of Clinical Therapeutics, Faculty of Medicine, Alexandra Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.M.S.); (N.M.-S.)
| | - Nikolaos Manetas-Stavrakakis
- Department of Clinical Therapeutics, Faculty of Medicine, Alexandra Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.M.S.); (N.M.-S.)
| | - Christos Kourek
- Department of Cardiology, 417 Army Share Fund Hospital of Athens (NIMTS), 11521 Athens, Greece;
| | - Andrew Xanthopoulos
- Department of Cardiology, University Hospital of Larissa, 41334 Larissa, Greece; (A.X.); (G.G.); (J.S.)
| | - Dimitrios Magouliotis
- Department of Cardiothoracic Surgery, University Hospital of Larissa, 41334 Larissa, Greece;
| | - Grigorios Giamouzis
- Department of Cardiology, University Hospital of Larissa, 41334 Larissa, Greece; (A.X.); (G.G.); (J.S.)
| | - John Skoularigis
- Department of Cardiology, University Hospital of Larissa, 41334 Larissa, Greece; (A.X.); (G.G.); (J.S.)
| | - Alexandros Briasoulis
- Department of Clinical Therapeutics, Faculty of Medicine, Alexandra Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.M.S.); (N.M.-S.)
| |
Collapse
|
40
|
Kong Y, Wei X, Zhang D, Lin H, Peng M, Shang H. Prevention and treatment of anthracycline-induced cardiotoxicity: A bibliometric analysis of the years 2000-2023. Heliyon 2024; 10:e29926. [PMID: 38698971 PMCID: PMC11064157 DOI: 10.1016/j.heliyon.2024.e29926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 03/20/2024] [Accepted: 04/17/2024] [Indexed: 05/05/2024] Open
Abstract
Aims This study aimed to evaluate the global research trend in the prevention and treatment of cardiotoxicity caused by anthracyclines from 2000 to 2023, and to explore international cooperation, research hotspots, and frontier trends. Methods The articles on the prevention and treatment of anthracycline-induced cardiotoxicity published from 2000 to 2023 were searched by Web of Science. The bibliometrics software CiteSpace was used for visual analysis of countries, institutions, journals, authors, cited authors, cited references, and keywords. Results This study analyzed the current status of global research on the prevention and treatment of cardiotoxicity caused by anthracyclines. A total of 3,669 papers were searched and 851 studies were included. The number of publications increased gradually throughout the years. Cardiovascular Toxicology (15) is the journal with the most publications. Circulation (547) ranked first among cited journals. In this field, the country with the most publications is the United States (229), and the institution with the most publications is Charles Univ Prague (18). In the analysis of the authors, Tomas S (10) ranked first. Cardinale D (262) ranked first among cited authors. In the ranking of cited literature frequency, the article ranked first is "Early detection of anthracycline cardiotoxicity and improvement with heart failure therapy" (121). The keywords "heart failure" (215) and "oxidative stress" (212) were the most frequent. "Enalapril", "inflammation", "cell death", "NF-κB" and "Nrf2" were the advanced research contents in 2019-2023. Conclusions This study provided valuable information for cardio-oncology researchers to identify potential collaborators and institutions, discover hot topics, and explore new research directions. The prevention and treatment of anthracycline-induced cardiotoxicity focuses on early detection and timely treatment. The results of the current clinical studies on the treatment of anthracycline-induced cardiotoxicity are contradictory, and more studies are needed to provide more reliable clinical evidence in the future.
Collapse
Affiliation(s)
- Yifan Kong
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaohong Wei
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Di Zhang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hongyuan Lin
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | | | - Hongcai Shang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
- Weifang Medical University, Weifang, China
| |
Collapse
|
41
|
Cronin M, Lowery A, Kerin M, Wijns W, Soliman O. Risk Prediction, Diagnosis and Management of a Breast Cancer Patient with Treatment-Related Cardiovascular Toxicity: An Essential Overview. Cancers (Basel) 2024; 16:1845. [PMID: 38791923 PMCID: PMC11120055 DOI: 10.3390/cancers16101845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/02/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Breast cancer is amongst the most common invasive cancers in adults. There are established relationships between anti-cancer treatments for breast cancer and cardiovascular side effects. In recent years, novel anti-cancer treatments have been established, as well as the availability of multi-modal cardiac imaging and the sophistication of treatment for cardiac disease. This review provides an in-depth overview regarding the interface of breast cancer and cancer therapy-related cardiovascular toxicity. Specifically, it reviews the pathophysiology of breast cancer, the method of action in therapy-related cardiovascular toxicity from anti-cancer treatment, the use of echocardiography, cardiac CT, MRI, or nuclear medicine as diagnostics, and the current evidence-based treatments available. It is intended to be an all-encompassing review for clinicians caring for patients in this situation.
Collapse
Affiliation(s)
- Michael Cronin
- School of Medicine, University of Galway, H91 V4AY Galway, Ireland
| | - Aoife Lowery
- Precision Cardio-Oncology Research Enterprise (P-CORE), H91 TK33 Galway, Ireland
- CURAM Centre for Medical Devices, H91 TK33 Galway, Ireland
| | - Michael Kerin
- Precision Cardio-Oncology Research Enterprise (P-CORE), H91 TK33 Galway, Ireland
- Discipline of Surgery, Lambe Institute for Translational Research, University of Galway, H91 V4AY Galway, Ireland
| | - William Wijns
- School of Medicine, University of Galway, H91 V4AY Galway, Ireland
- Precision Cardio-Oncology Research Enterprise (P-CORE), H91 TK33 Galway, Ireland
- CURAM Centre for Medical Devices, H91 TK33 Galway, Ireland
| | - Osama Soliman
- School of Medicine, University of Galway, H91 V4AY Galway, Ireland
- Precision Cardio-Oncology Research Enterprise (P-CORE), H91 TK33 Galway, Ireland
- CURAM Centre for Medical Devices, H91 TK33 Galway, Ireland
- Discipline of Surgery, Lambe Institute for Translational Research, University of Galway, H91 V4AY Galway, Ireland
| |
Collapse
|
42
|
Jeyaprakash P, Sangha S, Low G, Yu C, Pathan F, Negishi K. Prophylaxis to Prevent Cardiotoxicity in Patients Receiving Anthracycline for Breast Cancer: A Combined Bayesian and Frequentist Network Meta-Analysis of Randomised Controlled Trials. Heart Lung Circ 2024; 33:710-720. [PMID: 38184425 DOI: 10.1016/j.hlc.2023.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 10/31/2023] [Accepted: 11/03/2023] [Indexed: 01/08/2024]
Abstract
BACKGROUND The benefits in survivorship gained with anthracycline (ANT)-based chemotherapies for breast cancer are unfortunately mitigated for some patients by irreversible cardiotoxicity. Randomised controlled trials (RCTs) have explored multiple cardioprotection options, however, it remains unclear which drug is most effective in preserving left ventricular ejection fraction (LVEF). This study aimed to perform a systematic review and network meta-analysis, using Bayesian and frequentist approaches, of RCTs evaluating cardioprotective agents. METHODS Two authors searched four databases (CENTRAL, Cochrane Reviews, MEDLINE, SCOPUS), to find RCTs evaluating cardioprotective agents. Trial populations were limited to patients with breast cancer without prior ANT exposure. The primary outcome was mean LVEF change pre and post ANT dosing. Our primary analysis utilised a Bayesian approach, while our sensitivity analysis used frequentist methodology (Prospero registration number CRD42020199580). RESULTS From 4,007 search results, we identified 12 RCTs, with their various trial arms considered separately-nine beta-blocker (BB), two angiotensin-converting enzyme inhibitor /angiotensin receptor blockers [(AA)+BB=AABB], one AA, one spironolactone, one statin-evaluating 1,126 patients (age 50.5 years). Bayesian network meta-analysis showed no difference in LVEF preservation between AA (1.3%, 95% credible interval [-0.20, 2.9]), BB (0.77, [-0.21, 1.8]), AABB (0.84 [-1.1, 2.8]), spironolactone (0.72, [-2.3, 3.7]) or statin (0.60, [-2.4, 3.6]) when compared against placebo. However, the frequentist analysis showed benefits from using AA (mean difference, 1.32% [0.32, 2.33]) and BB (mean difference, 0.76% [0.12, 1.4]). CONCLUSIONS There is insufficient evidence to support prophylactic cardioprotection to prevent EF reduction. However, frequentist analysis suggested that AA or BBs provide cardioprotection. Thus, for those already on other anti-hypertensives, switching to AA or BBs could be considered.
Collapse
Affiliation(s)
- Prajith Jeyaprakash
- Department of Cardiology, Nepean Hospital, Sydney, NSW, Australia; Department of Academic Medicine, Sydney Medical School Nepean, Faculty of Medicine and Health, Charles Perkins Centre Nepean, The University of Sydney, Sydney, NSW, Australia
| | - Sukhman Sangha
- Department of Cardiology, Nepean Hospital, Sydney, NSW, Australia; Department of Academic Medicine, Sydney Medical School Nepean, Faculty of Medicine and Health, Charles Perkins Centre Nepean, The University of Sydney, Sydney, NSW, Australia
| | - Gary Low
- Department of Research Operations, Nepean Hospital, Sydney, NSW, Australia; Professorial Unit, The George Institute for Global Health, University of New South Wales, Sydney, NSW, Australia
| | - Christopher Yu
- Department of Cardiology, Nepean Hospital, Sydney, NSW, Australia; Department of Academic Medicine, Sydney Medical School Nepean, Faculty of Medicine and Health, Charles Perkins Centre Nepean, The University of Sydney, Sydney, NSW, Australia
| | - Faraz Pathan
- Department of Cardiology, Nepean Hospital, Sydney, NSW, Australia; Department of Academic Medicine, Sydney Medical School Nepean, Faculty of Medicine and Health, Charles Perkins Centre Nepean, The University of Sydney, Sydney, NSW, Australia
| | - Kazuaki Negishi
- Department of Cardiology, Nepean Hospital, Sydney, NSW, Australia; Department of Academic Medicine, Sydney Medical School Nepean, Faculty of Medicine and Health, Charles Perkins Centre Nepean, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
43
|
Satish P, Avenatti E, Patel J, Agarwala A. Understanding the spectrum of cardiovascular risk in women - A primer for prevention. Prog Cardiovasc Dis 2024; 84:34-42. [PMID: 38710313 DOI: 10.1016/j.pcad.2024.05.003] [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: 05/01/2024] [Accepted: 05/01/2024] [Indexed: 05/08/2024]
Abstract
Cardiovascular disease (CVD) is the leading cause of death in women worldwide and the lifetime risk of CVD in women is similar to men. However, the pathophysiology of CVD varies between women and men necessitating a sex-specific understanding of cardiovascular (CV) risk. A belief that women have a lower CVD risk than men, and an underrepresentation in clinical research for many years has led to a paucity of evidence in the prevention and management of CVD in women. Many recent efforts have tried to bridge the gap. As a result, we now know that traditional risk factors impact CVD risk differently in women when compared with men. There are also numerous sex-specific and pregnancy related risk factors that modify the risk and can predict the future development of CVD in women. This is important as risk calculators, in general, tend to misclassify risk in young women with nontraditional CVD risk factors. To address this, guidelines have introduced the concept of risk enhancers that can suggest a higher risk. The use of coronary artery calcium score can further accurately delineate risk in these women, leading to an appropriate matching of therapy to underlying risk. This review discusses implementation strategies that are essential to mitigate disparities in CVD outcomes and optimizing CVD risk in women.
Collapse
Affiliation(s)
- Priyanka Satish
- Center for Cardiovascular Prevention, Ascension Texas Cardiovascular, Dell Medical School, University of Texas, Austin, USA
| | - Eleonora Avenatti
- Center for Cardiovascular Prevention, Ascension Texas Cardiovascular, Dell Medical School, University of Texas, Austin, USA
| | - Jaideep Patel
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Anandita Agarwala
- Center for Cardiovascular Disease Prevention, Baylor Scott and White Health Heart Hospital Baylor Plano, Plano, TX, USA.
| |
Collapse
|
44
|
Liu J, Curtin C, Lall R, Lane S, Wieke J, Ariza A, Sejour L, Vlachos I, Zordoky BN, Peterson RT, Asnani A. Inhibition of Cyp1a Protects Mice against Anthracycline Cardiomyopathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.10.588915. [PMID: 38645084 PMCID: PMC11030370 DOI: 10.1101/2024.04.10.588915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Background Anthracyclines such as doxorubicin (Dox) are highly effective anti-tumor agents, but their use is limited by dose-dependent cardiomyopathy and heart failure. Our laboratory previously reported that induction of cytochrome P450 family 1 (Cyp1) enzymes contributes to acute Dox cardiotoxicity in zebrafish and in mice, and that potent Cyp1 inhibitors prevent cardiotoxicity. However, the role of Cyp1 enzymes in chronic Dox cardiomyopathy, as well as the mechanisms underlying cardioprotection associated with Cyp1 inhibition, have not been fully elucidated. Methods The Cyp1 pathway was evaluated using a small molecule Cyp1 inhibitor in wild-type (WT) mice, or Cyp1-null mice ( Cyp1a1/1a2 -/- , Cyp1b1 -/- , and Cyp1a1/1a2/1b1 -/- ). Low-dose Dox was administered by serial intraperitoneal or intravenous injections, respectively. Expression of Cyp1 isoforms was measured by RT-qPCR, and myocardial tissue was isolated from the left ventricle for RNA sequencing. Cardiac function was evaluated by transthoracic echocardiography. Results In WT mice, Dox treatment was associated with a decrease in Cyp1a2 and increase in Cyp1b1 expression in the heart and in the liver. Co-treatment of WT mice with Dox and the novel Cyp1 inhibitor YW-130 protected against cardiac dysfunction compared to Dox treatment alone. Cyp1a1/1a2 -/- and Cyp1a1/1a2/1b1 -/- mice were protected from Dox cardiomyopathy compared to WT mice. Male, but not female, Cyp1b1 -/- mice had increased cardiac dysfunction following Dox treatment compared to WT mice. RNA sequencing of myocardial tissue showed upregulation of Fundc1 and downregulation of Ccl21c in Cyp1a1/1a2 -/- mice treated with Dox, implicating changes in mitophagy and chemokine-mediated inflammation as possible mechanisms of Cyp1a-mediated cardioprotection. Conclusions Taken together, this study highlights the potential therapeutic value of Cyp1a inhibition in mitigating anthracycline cardiomyopathy.
Collapse
|
45
|
Jaiswal V, Ang SP, Deb N, Hanif M, Batra N, Kanagala SG, Vojjala N, Rajak K, Roy P, Sharath M, Waleed MS, Wajid Z, Mattumpuram J. Association between Statin Use and Chemotherapy-Induced Cardiotoxicity: A Meta-Analysis. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:580. [PMID: 38674227 PMCID: PMC11052115 DOI: 10.3390/medicina60040580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 03/08/2024] [Indexed: 04/28/2024]
Abstract
Background: Chemotherapy-induced cardiac dysfunction (CIC) is a significant and concerning complication observed among cancer patients. Despite the demonstrated cardioprotective benefits of statins in various cardiovascular diseases, their effectiveness in mitigating CIC remains uncertain. Objective: This meta-analysis aims to comprehensively evaluate the potential cardioprotective role of statins in patients with CIC. Methods: A systematic literature search was conducted using PubMed, Embase, and Scopus databases to identify relevant articles published from inception until 10th May 2023. The outcomes were assessed using pooled odds ratio (OR) for categorical data and mean difference (MD) for continuous data, with corresponding 95% confidence intervals (95% CIs). Results: This meta-analysis comprised nine studies involving a total of 5532 patients, with 1904 in the statin group and 3628 in the non-statin group. The pooled analysis of primary outcome shows that patients who did not receive statin suffer a greater decline in the LVEF after chemotherapy compared to those who receive statin (MD, 3.55 (95% CI: 1.04-6.05), p = 0.01). Likewise, we observed a significantly higher final mean LVEF among chemotherapy patients with statin compared to the non-statin group of patients (MD, 2.08 (95% CI: 0.86-3.30), p > 0.001). Additionally, there was a lower risk of incident heart failure in the statin group compared to the non-statin group of patients (OR, 0.41 (95% CI: 0.27-0.62), p < 0.001). Lastly, the change in the mean difference for LVEDV was not statistically significant between the statin and non-statin groups (MD, 1.55 (95% CI: -5.22-8.33), p = 0.65). Conclusion: Among patients of CIC, statin use has shown cardioprotective benefits by improving left ventricular function and reducing the risk of heart failure.
Collapse
Affiliation(s)
- Vikash Jaiswal
- Department of Cardiovascular Research, Larkin Community Hospital, South Miami, FL 33143, USA
| | - Song Peng Ang
- Department of Internal Medicine, Rutgers Health/Community Medical Center, Toms River, NJ 08755, USA
| | - Novonil Deb
- North Bengal Medical College and Hospital, Darjeeling 734012, West Bengal, India
| | - Muhammad Hanif
- Department of Internal Medicine, SUNY Upstate Medical University, 750 E Adams St., Syracuse, NY 13210, USA
| | - Nitya Batra
- Department of Internal Medicine, Beaumont Hospital, Royal Oak, MI 48073, USA
| | - Sai Gautham Kanagala
- Department of Internal Medicine, Metropolitan Hospital Center, New York, NY 10029, USA
| | - Nikhil Vojjala
- Internal Medicine Department, Trinity Health Oakland/Wayne State University, Detroit, MI 48341, USA
| | - Kripa Rajak
- Department of Internal Medicine, UPMC Harrisburgh, 111 S Front St., Harrisburg, PA 17101, USA
| | - Poulami Roy
- North Bengal Medical College and Hospital, Darjeeling 734012, West Bengal, India
| | - Medha Sharath
- Bangalore Medical College and Research Institute, Kalasipalya, Bengaluru 560002, Karnataka, India
| | - Madeeha Subhan Waleed
- Department of Internal Medicine, Lower Bucks Hospital, Bristo, 501 Bath Rd., Bristol, PA 19007, USA
| | - Zarghoona Wajid
- Department of Internal Medicine, Wayne State University School of Medicine, 540 E. Canfield Ave., Detroit, MI 48201, USA
| | - Jishanth Mattumpuram
- Division of Cardiology, Department of Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
46
|
Tabassum M, Chikermane SG, Johnson C, Abdulkareem NM, Wang EM, Johnson ML, Trivedi MV. Comparing the effects of various β-blockers on cardiovascular mortality in breast cancer patients. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:17. [PMID: 38532523 PMCID: PMC10964697 DOI: 10.1186/s40959-024-00217-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 03/05/2024] [Indexed: 03/28/2024]
Abstract
BACKGROUND Cardiovascular (CV) disease is a leading cause of death in breast cancer (BC) patients due to the increased age and treatments. While individual β-blockers have been investigated to manage CV complications, various β-blockers have not been compared for their effects on CV death in this population. We aimed to compare CV mortality in older BC patients taking one of the commonly used β-blockers. METHODS This retrospective cohort study was conducted using the Surveillance, Epidemiology and End Results (SEER) - Medicare data (2010-2015). Patients of age 66 years or older at BC diagnosis receiving metoprolol, atenolol, or carvedilol monotherapy were included. The competing risk regression model was used to determine the risk of CV mortality in the three β-blocker groups. The multivariable model was adjusted for demographic and clinical covariates. The adjusted hazard ratio (HR) and 95% confidence intervals (CI) were reported for the risk of CV mortality. RESULTS The study cohort included 6,540 patients of which 55% were metoprolol users, 30% were atenolol users, and 15% were carvedilol users. Metoprolol was associated with a 37% reduced risk of CV mortality (P = 0.03) compared to carvedilol after adjusting for the covariates (HR = 0.63; 95% CI 0.41-0.96). No significant difference in the risk of CV mortality between atenolol and carvedilol users was observed (HR = 0.74; 95% CI 0.44-1.22). CONCLUSIONS Our findings suggest that metoprolol is associated with a reduced risk of CV mortality in BC patients. Future studies are needed to confirm these findings and understand the mechanism of action.
Collapse
Affiliation(s)
- Mantasha Tabassum
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, 4349 Martin Luther King Blvd, 77204, Houston, TX, USA
| | - Soumya G Chikermane
- Department of Pharmaceutical Health Outcomes and Policy, University of Houston College of Pharmacy, Houston, TX, USA
| | - Camille Johnson
- Department of Pharmacy Practice and Translational Research, University of Houston College of Pharmacy, Houston, TX, USA
| | - Noor M Abdulkareem
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, 4349 Martin Luther King Blvd, 77204, Houston, TX, USA
| | - Elisabeth M Wang
- Department of Pharmacy Practice and Translational Research, University of Houston College of Pharmacy, Houston, TX, USA
| | - Michael L Johnson
- Department of Pharmaceutical Health Outcomes and Policy, University of Houston College of Pharmacy, Houston, TX, USA
| | - Meghana V Trivedi
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, 4349 Martin Luther King Blvd, 77204, Houston, TX, USA.
- Department of Pharmacy Practice and Translational Research, University of Houston College of Pharmacy, Houston, TX, USA.
| |
Collapse
|
47
|
Di Lisi D, Cadeddu Dessalvi C, Zito C, Madaudo C, Manganaro R, Mercurio V, Deidda M, Santoro C, Penna C, Monte IP, Spallarossa P, Tocchetti CG, Novo G. Management of cancer patients at high and very-high risk of cardiotoxicity: Main questions and answers. Curr Probl Cardiol 2024; 49:102229. [PMID: 38154703 DOI: 10.1016/j.cpcardiol.2023.102229] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 11/28/2023] [Indexed: 12/30/2023]
Abstract
In recent years, important advances have been made in the field of Cardio-Oncology. The 2022 ESC Guidelines on Cardio-Oncology proposed a baseline cardiovascular risk stratification for cancer patients and preventive strategies in patients at high and very-high risk of cardiotoxicity. Cardiovascular toxic effects of anti-cancer drugs are being extensively studied; surveillance programs have been proposed, based on the baseline cardiovascular risk. On the other hand, there is little data on Cardio-Oncological management of patients at high and very-high cardiovascular risk with previous cardiovascular diseases. For example, little is known about management of cancer patients with heart failure with reduced ejection fraction (HFrEF), patients with a recent myocardial infarction or other cardiovascular diseases; when to resume anti-cancer drugs after a cardiovascular toxic event. Collaboration between Cardiologists and Oncologists and multidisciplinary team evaluations are certainly essential to decide the best therapeutic strategy for cancer patients, to treat cancer while saving the heart. Therefore, in the present review, we attempt to provide a useful guide to clinicians in treating patients with high and very-high risk of cardiotoxicity by enucleating main questions and answering them based on the evidence available as well as expert opinion and our clinical experience.
Collapse
Affiliation(s)
- Daniela Di Lisi
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Division of Cardiology, University Hospital Paolo Giaccone, Palermo, Italy..
| | | | - Concetta Zito
- Department of Clinical and Experimental Medicine - Cardiology Unit, University of Messina, Messina, Italy
| | - Cristina Madaudo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Division of Cardiology, University Hospital Paolo Giaccone, Palermo, Italy
| | - Roberta Manganaro
- Department of Clinical and Experimental Medicine - Cardiology Unit, University of Messina, Messina, Italy
| | - Valentina Mercurio
- Department of Translational Medical Sciences, Federico II University, Naples, Italy; Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Federico II University, Naples, Italy; Interdepartmental Hypertension Research Center (CIRIAPA), Federico II University, Naples, Italy
| | - Martino Deidda
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino, Italy-IRCCS Italian Cardiovascular Network & Department of Internal Medicine, University of Genova, 16121 Genova, Italy
| | - Ciro Santoro
- Department of Advanced Biomedical Sciences, Federico II University, 80131 Naples, Italy
| | - Claudia Penna
- Department of Clinical and Biological Sciences of Turin University, Orbassano, Turin, I-10043, Italy
| | - Ines Paola Monte
- Department of General Surgery and Medical-Surgery Specialities- Cardiology, University of Catania, Catania, Italy
| | - Paolo Spallarossa
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino, Italy-IRCCS Italian Cardiovascular Network & Department of Internal Medicine, University of Genova, 16121 Genova, Italy
| | - Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences, Federico II University, Naples, Italy; Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Federico II University, Naples, Italy; Interdepartmental Hypertension Research Center (CIRIAPA), Federico II University, Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), Federico II University, Naples, Italy
| | - Giuseppina Novo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Division of Cardiology, University Hospital Paolo Giaccone, Palermo, Italy
| |
Collapse
|
48
|
Vakilpour A, Lefebvre B, Lai C, Scherrer-Crosbie M. Heartbreaker: Detection and prevention of cardiotoxicity in hematological malignancies. Blood Rev 2024; 64:101166. [PMID: 38182490 DOI: 10.1016/j.blre.2023.101166] [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: 10/30/2023] [Revised: 12/12/2023] [Accepted: 12/30/2023] [Indexed: 01/07/2024]
Abstract
Cancer survivors are at significant risk of cardiovascular (CV) morbidity and mortality; patients with hematologic malignancies have a higher rate of death due to heart failure compared to all other cancer subtypes. The majority of conventional hematologic cancer treatments is associated with increased risk of acute and long-term CV toxicity. The incidence of cancer therapy induced CV toxicity depends on the combination of patient characteristics and on the type, dose, and duration of the therapy. Early diagnosis of CV toxicity, appropriate referral, more specific cardiac monitoring follow-up and timely interventions in target patients can decrease the risk of CV adverse events, the interruption of oncological therapy, and improve the patient's prognosis. Herein, we summarize the CV effects of conventional treatments used in hematologic malignancies with a focus on definitions and incidence of the most common CV toxicities, guideline recommended early detection approaches, and preventive strategies before and during cancer treatments.
Collapse
Affiliation(s)
- Azin Vakilpour
- Division of Cardiovascular Diseases, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA.
| | - Bénédicte Lefebvre
- Division of Cardiovascular Diseases, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; The Thalheimer Center for Cardio-oncology, Division of Cardiology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| | - Catherine Lai
- Division of Hematology-Oncology, Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Marielle Scherrer-Crosbie
- Division of Cardiovascular Diseases, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; The Thalheimer Center for Cardio-oncology, Division of Cardiology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
49
|
Szponar J, Gorska A, Ostrowska-Lesko M, Korga-Plewko A, Tchorz M, Ciechanski E, Dabrowska A, Poleszak E, Burdan F, Dudka J, Murias M, Mandziuk S. Assessment of the Impact of Carvedilol Administered Together with Dexrazoxan and Doxorubicin on Liver Structure and Function, Iron Metabolism, and Myocardial Redox System in Rats. Int J Mol Sci 2024; 25:2219. [PMID: 38396896 PMCID: PMC10889540 DOI: 10.3390/ijms25042219] [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: 01/06/2024] [Revised: 02/08/2024] [Accepted: 02/11/2024] [Indexed: 02/25/2024] Open
Abstract
Late cardiotoxicity is a formidable challenge in anthracycline-based anticancer treatments. Previous research hypothesized that co-administration of carvedilol (CVD) and dexrazoxane (DEX) might provide superior protection against doxorubicin (DOX)-induced cardiotoxicity compared to DEX alone. However, the anticipated benefits were not substantiated by the findings. This study focuses on investigating the impact of CVD on myocardial redox system parameters in rats treated with DOX + DEX, examining its influence on overall toxicity and iron metabolism. Additionally, considering the previously observed DOX-induced ascites, a seldom-discussed condition, the study explores the potential involvement of the liver in ascites development. Compounds were administered weekly for ten weeks, with a specific emphasis on comparing parameter changes between DOX + DEX + CVD and DOX + DEX groups. Evaluation included alterations in body weight, feed and water consumption, and analysis of NADPH2, NADP+, NADPH2/NADP+, lipid peroxidation, oxidized DNA, and mRNA for superoxide dismutase 2 and catalase expressions in cardiac muscle. The iron management panel included markers for iron, transferrin, and ferritin. Liver abnormalities were assessed through histological examinations, aspartate transaminase, alanine transaminase, and serum albumin level measurements. During weeks 11 and 21, reduced NADPH2 levels were observed in almost all examined groups. Co-administration of DEX and CVD negatively affected transferrin levels in DOX-treated rats but did not influence body weight changes. Ascites predominantly resulted from cardiac muscle dysfunction rather than liver-related effects. The study's findings, exploring the impact of DEX and CVD on DOX-induced cardiotoxicity, indicate a lack of scientific justification for advocating the combined use of these drugs at histological, biochemical, and molecular levels.
Collapse
Affiliation(s)
- Jaroslaw Szponar
- Toxicology Clinic, Faculty of Medicine, Medical University of Lublin, 100 Krasnik Avenue, 20-550 Lublin, Poland; (J.S.); (A.G.); (M.T.)
- Clinical Department of Toxicology and Cardiology, Stefan Wyszynski Regional Specialist Hospital, 100 Krasnik Avenue, 20-550 Lublin, Poland
| | - Agnieszka Gorska
- Toxicology Clinic, Faculty of Medicine, Medical University of Lublin, 100 Krasnik Avenue, 20-550 Lublin, Poland; (J.S.); (A.G.); (M.T.)
- Clinical Department of Toxicology and Cardiology, Stefan Wyszynski Regional Specialist Hospital, 100 Krasnik Avenue, 20-550 Lublin, Poland
| | - Marta Ostrowska-Lesko
- Department of Toxicology, Medical University of Lublin, 8b Jaczewski Street, 20-090 Lublin, Poland; (E.C.); (J.D.)
| | - Agnieszka Korga-Plewko
- Independent Medical Biology Unit, Medical University, 8b Jaczewski Street, 20-090 Lublin, Poland;
| | - Michal Tchorz
- Toxicology Clinic, Faculty of Medicine, Medical University of Lublin, 100 Krasnik Avenue, 20-550 Lublin, Poland; (J.S.); (A.G.); (M.T.)
- Clinical Department of Toxicology and Cardiology, Stefan Wyszynski Regional Specialist Hospital, 100 Krasnik Avenue, 20-550 Lublin, Poland
| | - Erwin Ciechanski
- Department of Toxicology, Medical University of Lublin, 8b Jaczewski Street, 20-090 Lublin, Poland; (E.C.); (J.D.)
- Clinical Department of Cardiology, Stefan Wyszynski Regional Specialist Hospital, 100 Krasnik Avenue, 20-550 Lublin, Poland
| | - Anna Dabrowska
- Department of Toxicology, Medical University of Lublin, 8b Jaczewski Street, 20-090 Lublin, Poland; (E.C.); (J.D.)
| | - Ewa Poleszak
- Department of Applied Pharmacy, Medical University of Lublin, 1 Chodźko Street, 20-093 Lublin, Poland;
| | - Franciszek Burdan
- Human Anatomy Department, Medical University of Lublin, 4 Jaczewski Street, 20-090 Lublin, Poland;
| | - Jaroslaw Dudka
- Department of Toxicology, Medical University of Lublin, 8b Jaczewski Street, 20-090 Lublin, Poland; (E.C.); (J.D.)
| | - Marek Murias
- Department of Toxicology, Poznan University of Medical Sciences, 3 Rokietnicka Street, 60-608 Poznan, Poland;
| | - Slawomir Mandziuk
- Department of Pneumology, Oncology and Allergology, Medical University of Lublin, 8 Jaczewski Street, 20-090 Lublin, Poland;
| |
Collapse
|
50
|
Attanasio U, Di Sarro E, Tricarico L, Di Lisi D, Armentaro G, Miceli S, Fioretti F, Deidda M, Correale M, Novo G, Sciacqua A, Nodari S, Cadeddu C, Tocchetti CG, Palazzuoli A, Mercurio V. Cardiovascular Biomarkers in Cardio-Oncology: Antineoplastic Drug Cardiotoxicity and Beyond. Biomolecules 2024; 14:199. [PMID: 38397436 PMCID: PMC10887095 DOI: 10.3390/biom14020199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/29/2024] [Accepted: 02/03/2024] [Indexed: 02/25/2024] Open
Abstract
Serum biomarkers represent a reproducible, sensitive, minimally invasive and inexpensive method to explore possible adverse cardiovascular effects of antineoplastic treatments. They are useful tools in risk stratification, the early detection of cardiotoxicity and the follow-up and prognostic assessment of cancer patients. In this literature review, we aim at describing the current state of knowledge on the meaning and the usefulness of cardiovascular biomarkers in patients with cancer; analyzing the intricate relationship between cancer and cardiovascular disease (especially HF) and how this affects cardiovascular and tumor biomarkers; exploring the role of cardiovascular biomarkers in the risk stratification and in the identification of chemotherapy-induced cardiotoxicity; and providing a summary of the novel potential biomarkers in this clinical setting.
Collapse
Affiliation(s)
- Umberto Attanasio
- Department of Translational Medical Sciences, Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy (E.D.S.); (C.G.T.)
| | - Elena Di Sarro
- Department of Translational Medical Sciences, Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy (E.D.S.); (C.G.T.)
| | - Lucia Tricarico
- Cardiology Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto 1, 71122 Foggia, Italy; (L.T.); (M.C.)
| | - Daniela Di Lisi
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (D.D.L.); (G.N.)
- Division of Cardiology, University Hospital Paolo Giaccone, Via del Vespro 129, 90127 Palermo, Italy
| | - Giuseppe Armentaro
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Campus Universitario di Germaneto, V.le Europa, 88100 Catanzaro, Italy; (G.A.); (S.M.); (A.S.)
| | - Sofia Miceli
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Campus Universitario di Germaneto, V.le Europa, 88100 Catanzaro, Italy; (G.A.); (S.M.); (A.S.)
| | - Francesco Fioretti
- Cardiology Section, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, Spedali Civili Hospital and University of Brescia, Piazzale Spedali Civili 1, 25123 Brescia, Italy; (F.F.); (S.N.)
| | - Martino Deidda
- Department of Medical Sciences and Public Health, University of Cagliari, 09042 Monserrato, Italy (C.C.)
| | - Michele Correale
- Cardiology Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto 1, 71122 Foggia, Italy; (L.T.); (M.C.)
| | - Giuseppina Novo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (D.D.L.); (G.N.)
- Division of Cardiology, University Hospital Paolo Giaccone, Via del Vespro 129, 90127 Palermo, Italy
| | - Angela Sciacqua
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Campus Universitario di Germaneto, V.le Europa, 88100 Catanzaro, Italy; (G.A.); (S.M.); (A.S.)
| | - Savina Nodari
- Cardiology Section, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, Spedali Civili Hospital and University of Brescia, Piazzale Spedali Civili 1, 25123 Brescia, Italy; (F.F.); (S.N.)
| | - Christian Cadeddu
- Department of Medical Sciences and Public Health, University of Cagliari, 09042 Monserrato, Italy (C.C.)
| | - Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences, Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy (E.D.S.); (C.G.T.)
- Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy
- Interdepartmental Hypertension Research Center (CIRIAPA), Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy
| | - Alberto Palazzuoli
- Cardiovascular Diseases Unit, Cardio-thoracic and Vascular Department Le Scotte Hospital, University of Siena, Strada delle Scotte 14, 53100 Siena, Italy;
| | - Valentina Mercurio
- Department of Translational Medical Sciences, Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy (E.D.S.); (C.G.T.)
- Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy
- Interdepartmental Hypertension Research Center (CIRIAPA), Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy
| |
Collapse
|