1
|
Poovorawan N, Susiriwatananont T, Teerapakpinyo C, Chariyavilaskul P, Sitthideatphaiboon P, Jarutasnangkul L, Tumkosit M, Chattranukulchai P, Theerasuwipakorn N, Aporntewan C, Shuangshoti S, Manasnayakorn S, Vinayanuwattikun C, Vorasettakarnkij Y, Sriuranpong V. Long-term impact of anthracycline in early-stage breast cancer, bridging of MiRNAs profiler for early cardiotoxicity. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2025; 11:39. [PMID: 40270054 PMCID: PMC12016148 DOI: 10.1186/s40959-025-00337-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 04/10/2025] [Indexed: 04/25/2025]
Abstract
BACKGROUND Anthracyclines are essential in early breast cancer chemotherapy but pose long-term cardiotoxicity risks. OBJECTIVES This study aims to investigate the long-term incidence of cancer therapy-related cardiac dysfunction (CTRCD), bridging with the miRNAs profiler representing acute cardiac injury. METHODS We conducted a prospective cohort including stage I-III breast cancer patients who received anthracycline between 2007 and 2012. Echocardiography was performed before and 12 weeks after anthracycline administration. The miRNAs profiler was conducted by NanoString and RT-PCR. Long-term cardiac magnetic resonance imaging (CMR) was evaluated in 24.2% of asymptomatic participants. RESULTS At a median follow-up of 11 [IQR 6-12] years, 194 patients who completed follow-up echocardiography after anthracycline were included in the analysis. The median age at diagnosis was 50 [26-72] years. An early LVEF decline of ≥ 10% was found in 32.9% of participants. The cumulative equivalent dose of doxorubicin was 223.2 ± 21.6 mg/m2. At the time of censoring, sixty-four participants (32.9%) died, 70% from breast cancer. Nine participants (4.6%) reported cardiovascular events compatible with the CTRCD definition. Forty-seven participants (24.2%) underwent long-term cardiac evaluation. The miRNAs profiler and RT-PCR at different time points, 3 weeks and 6 weeks, respectively, revealed significantly diverse expressions of miR-1-3p and miR-16-5p in participants with and without an early LVEF decline of ≥ 10%. Despite cardiac injury demonstrated by dynamic miR-1-3p and miR-16-5p, CMR parameters revealed no significant differences. CONCLUSIONS Our study demonstrates a very low incidence of long-term symptomatic CTRCD. The diverse expression patterns of miR-16-5p and miR-1-3p at different time points also provide valuable biological insights. Within-normal results of an exact and comprehensive CMR, in asymptomatic and any LVEF change participants, indicate the long-term safety of limited-dose anthracycline-containing use.
Collapse
Affiliation(s)
- Nattaya Poovorawan
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and The King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Thiti Susiriwatananont
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and The King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Chinachote Teerapakpinyo
- Chula GenePRO Center, Research Affairs, Faculty of Medicine, Chulalongkorn University and The King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Pajaree Chariyavilaskul
- Department of Pharmacology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Clinical Pharmacokinetics and Pharmacogenomics, Chulalongkorn University, Bangkok, Thailand
| | - Piyada Sitthideatphaiboon
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and The King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Luxica Jarutasnangkul
- Department of Radiology, Faculty of Medicine, Chulalongkorn University and The King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Monravee Tumkosit
- Department of Radiology, Faculty of Medicine, Chulalongkorn University and The King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Pairoj Chattranukulchai
- Division of Cardiovascular Medicine, Department of Medicine, Faculty of Medicine, Chulalongkorn University, and The King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Nonthikorn Theerasuwipakorn
- Division of Cardiovascular Medicine, Department of Medicine, Faculty of Medicine, Chulalongkorn University, and The King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Chatchawit Aporntewan
- Department of Mathematics and Computer Sciences & Omics Sciences and Bioinformatics Center, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Shanop Shuangshoti
- Chula GenePRO Center, Research Affairs, Faculty of Medicine, Chulalongkorn University and The King Chulalongkorn Memorial Hospital, Bangkok, Thailand
- Department of Pathology, Faculty of Medicine, Chulalongkorn University and The King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Sopark Manasnayakorn
- Department of Surgery, Faculty of Medicine, Chulalongkorn University and The King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Chanida Vinayanuwattikun
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and The King Chulalongkorn Memorial Hospital, Bangkok, Thailand.
| | - Yongkasem Vorasettakarnkij
- Division of Hospital and Ambulatory Medicine, Department of Medicine, Faculty of Medicine, Chulalongkorn University and The King Chulalongkorn Memorial Hospital, Bangkok, Thailand.
| | - Virote Sriuranpong
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and The King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| |
Collapse
|
2
|
Qin Y, Mo Y, Li P, Liang X, Yu J, Chen D. Concurrent immunotherapy improves progression-free survival but increases toxicity in unresectable stage III NSCLC. Lung Cancer 2025; 203:108544. [PMID: 40253944 DOI: 10.1016/j.lungcan.2025.108544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 04/13/2025] [Accepted: 04/16/2025] [Indexed: 04/22/2025]
Abstract
BACKGROUND The PACIFIC trial established consolidation immunotherapy (IO) after concurrent chemoradiotherapy as the standard treatment for unresectable stage III non-small cell lung cancer (NSCLC) by improving survival. However, the optimal timing of IO remains debated. This study analyzes the survival benefits and risks of IO concurrent with radiotherapy (RT) versus IO following RT. METHODS A systematic search of multiple databases identified studies comparing IO concurrent with RT and IO following RT in unresectable stage III NSCLC. Data on overall survival (OS), progression-free survival (PFS), and adverse events (AEs) were analyzed using the "meta" package in R, along with a single-center cohort study. RESULTS The meta-analysis showed improved PFS with IO concurrent with RT, with significant differences at 1 year (69.5 % vs 57.6 %) and 1.5 years (56.3 % vs 45.7 %). OS was slightly better with IO following RT, with fewer severe AEs (≥grade 3: 52.6 % vs 37.2 %). A single-center cohort validated superior PFS for the concurrent group (HR = 2.039, 95 % CI: 1.014-4.322, P = 0.046). Shorter intervals between RT and IO were associated with better PFS in the following group (HR = 1.002, 95 % CI: 1.001-1.003, P = 0.002). CONCLUSION Concurrent IO with RT during chemoradiotherapy significantly improved PFS in unresectable stage III NSCLC, though OS did not benefit due to a higher incidence of severe AEs. Earlier IO initiation after RT was associated with better PFS. Our findings suggest the potential benefits of concurrent IO for selected patients.
Collapse
Affiliation(s)
- Yiwei Qin
- Department of Radiation Oncology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, People's Republic of China; Department of Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250000, People's Republic of China
| | - You Mo
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Shantou University Medical College, Shantou University, Shantou, Guangdong 515000, People's Republic of China
| | - Pengwei Li
- Department of Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250000, People's Republic of China
| | - Xinyi Liang
- Department of Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250000, People's Republic of China; School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong 261000, People's Republic of China
| | - Jinming Yu
- Department of Radiation Oncology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, People's Republic of China; Department of Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250000, People's Republic of China.
| | - Dawei Chen
- Department of Radiation Oncology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, People's Republic of China; Department of Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250000, People's Republic of China.
| |
Collapse
|
3
|
Wang Y, Huan C, Wang G, Pu H, Liu Y, Zhang X, Li C, Liu J, Wu W, Pan D. The inflammation burden index can predict the cardiac injury following antitumour therapy in lung cancer patients with diabetes. Sci Rep 2025; 15:11291. [PMID: 40175509 PMCID: PMC11965416 DOI: 10.1038/s41598-025-95758-5] [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: 10/30/2024] [Accepted: 03/24/2025] [Indexed: 04/04/2025] Open
Abstract
Lung cancer is a leading cause of cancer-related morbidity and mortality worldwide. Diabetes, as a common metabolic disorder, further increases the risk of cardiovascular damage. Studies have shown that myocardial cells in diabetic patients are more vulnerable to the toxic effects of cancer treatments, thereby raising the risk of heart failure. Therefore, cardiovascular risk assessment in lung cancer patients with diabetes is particularly important. The Inflammatory Burden Index (IBI) is a biomarker that reflects systemic inflammation, combining the levels of C-reactive protein (CRP), neutrophils, and lymphocytes. It has been shown to be a significant prognostic indicator in various cancer populations. This study aims to investigate the predictive ability of IBI and the triglyceride-glucose (TyG) index for cardiac injury in lung cancer patients with diabetes undergoing antitumor therapy. A single-center retrospective case-control study was conducted, including clinical data of 192 lung cancer patients with diabetes who received anti-tumor therapy from July 2018 to January 2023.Cardiac injury was assessed by measuring high-sensitivity cardiac troponin T (hs-cTnT) levels. The Inflammatory Burden Index (IBI) was calculated using the formula: IBI = (C-reactive protein [CRP] × Neutrophils)/Lymphocytes. Univariate and multivariate logistic regression analyses were performed to assess the relationship between clinical factors, IBI, TyG index, and cardiac injury. The clinical predictive value of these factors was further evaluated using receiver operating characteristic (ROC) curves, and the relationship between IBI and cardiac injury was explored using the Restricted Cubic Spline (RCS) model. In the 192 patients, 101 (52.6%) developed cardiac injury during follow-up. Univariate analysis showed that age, male, hypertension, smoking, D-dimer, CKMB, IBI, TNM stage: III/IV, and Immunotherapy were significantly associated with cardiac injury (P < 0.001). Multivariate analysis identified IBI (P = 0.007), age (P = 0.01), smoking (P < 0.001), CKMB (P < 0.001), TNM stage (P = 0.014), and hypertension (P = 0.007) as independent predictors of cardiac injury. ROC analysis revealed an area under curve (AUC) of 0.722 for IBI (cut-off: 8.408), indicating good predictive value. RCS analysis showed a significant nonlinear positive correlation between IBI and cardiac injury (P = 0.0079), with the risk of cardiac injury increasing significantly as IBI levels rose. The TyG index was not significantly associated with cardiac injury. IBI, as a simple and accessible biomarker, can effectively predict cardiac injury in lung cancer patients with diabetes undergoing antitumor therapy. Although the TyG index did not show a significant association with cardiac injury in this study, IBI demonstrated strong predictive ability in this patient group. Future studies should further validate the use of IBI across different cancer types and evaluate its prognostic role in long-term follow-up.
Collapse
Affiliation(s)
- Yumin Wang
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Chunyan Huan
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Guodong Wang
- Cardiovascular Medicine Department, Capital Medical University, Beijin, China
| | - Huijuan Pu
- Department of General Practice, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yan Liu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xiuli Zhang
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Chengyang Li
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Jie Liu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Wanling Wu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Defeng Pan
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
4
|
Ravera F, Gilardi N, Ballestrero A, Zoppoli G. Applications, challenges and future directions of artificial intelligence in cardio-oncology. Eur J Clin Invest 2025; 55 Suppl 1:e14370. [PMID: 40191923 PMCID: PMC11973867 DOI: 10.1111/eci.14370] [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: 10/23/2024] [Accepted: 11/28/2024] [Indexed: 04/09/2025]
Abstract
BACKGROUND The management of cardiotoxicity related to cancer therapies has emerged as a significant clinical challenge, prompting the rapid growth of cardio-oncology. As cancer treatments become more complex, there is an increasing need to enhance diagnostic and therapeutic strategies for managing their cardiovascular side effects. OBJECTIVE This review investigates the potential of artificial intelligence (AI) to revolutionize cardio-oncology by integrating diverse data sources to address the challenges of cardiotoxicity management. METHODS We explore applications of AI in cardio-oncology, focusing on its ability to leverage multiple data sources, including electronic health records, electrocardiograms, imaging modalities, wearable sensors, and circulating serum biomarkers. RESULTS AI has demonstrated significant potential in improving risk stratification and longitudinal monitoring of cardiotoxicity. By optimizing the use of electrocardiograms, non-invasive imaging, and circulating biomarkers, AI facilitates earlier detection, better prediction of outcomes, and more personalized therapeutic interventions. These advancements are poised to enhance patient outcomes and streamline clinical decision-making. CONCLUSIONS AI represents a transformative opportunity in cardio-oncology by advancing diagnostic and therapeutic capabilities. However, successful implementation requires addressing practical challenges such as data integration, model interpretability, and clinician training. Continued collaboration between clinicians and AI developers will be essential to fully integrate AI into routine clinical workflows.
Collapse
Affiliation(s)
- Francesco Ravera
- Department of Internal Medicine and Medical SpecialtiesUniversity of GenoaGenoaItaly
| | - Nicolò Gilardi
- Department of Internal Medicine and Medical SpecialtiesUniversity of GenoaGenoaItaly
| | - Alberto Ballestrero
- Department of Internal Medicine and Medical SpecialtiesUniversity of GenoaGenoaItaly
- IRCCS Ospedale Policlinico San MartinoGenoaItaly
| | - Gabriele Zoppoli
- Department of Internal Medicine and Medical SpecialtiesUniversity of GenoaGenoaItaly
- IRCCS Ospedale Policlinico San MartinoGenoaItaly
| |
Collapse
|
5
|
Wang Y, Huan C, Pu H, Wang G, Liu Y, Zhang X, Li C, Liu J, Wu W, Pan D. A new online dynamic nomogram based on the inflammation burden index to predict cardiac injury after antitumor therapy in lung cancer patients. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2025; 11:32. [PMID: 40158125 PMCID: PMC11954219 DOI: 10.1186/s40959-025-00328-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 03/14/2025] [Indexed: 04/01/2025]
Abstract
INTRODUCTION Cardiotoxicity has become a major concern in cancer patients, especially those with lung cancer, as anti-tumor therapies can significantly affect patient survival and quality of life. This study aims to develop and validate a dynamic nomogram based on the Inflammation Burden Index (IBI) to predict the risk of cardiac injury within one year after anti-tumor treatment in lung cancer patients. METHODS This single-center, retrospective study included 1386 lung cancer patients who underwent myocardial enzyme testing between July 2018 and January 2023. The IBI was calculated as: IBI = (CRP (mg/dL) × Neutrophils (/μL)) / Lymphocytes (/μL). Statistical analysis using SPSS 22.0 and R 4.4.1, including machine learning algorithms and multivariate logistic analysis, identified independent predictors of cardiac injury. An online dynamic nomogram was developed and validated using internal validation, ROC curves, and decision curve analysis (DCA). RESULTS The average age of the 1386 patients was 61.98 ± 9.22 years. Significant independent predictors included age, BMI, hypertension, immunotherapy, D-dimer, LDH, NSE, CKMB, and IBI. The nomogram showed strong discriminative ability with AUC-ROC values of 0.85 for the training set and 0.86 for the validation set. Calibration curves confirmed good fit, and DCA showed high clinical utility. CONCLUSION An online dynamic nomogram based on clinical and inflammatory markers was developed to predict cardiac injury in lung cancer patients following anti-tumor therapy. The model shows strong discriminative ability and potential clinical value, which can provide vital information for oncologists when designing customized clinical treatments.
Collapse
Affiliation(s)
- Yumin Wang
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Chunyan Huan
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Huijuan Pu
- Department of General Practice, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Guodong Wang
- Cardiovascular Medicine Department, Capital Medical University, Beijin, China
| | - Yan Liu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xiuli Zhang
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Chengyang Li
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Jie Liu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Wanling Wu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Defeng Pan
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
6
|
Fischer-Bacca CO, Huntermann R, de Oliveira JP, Alexandrino FB, Sato MY, Cardoso R, Gomes RF, Melo ES. Systematic Review and Meta-analysis of Right Ventricular Changes in Cancer-therapy - The Forgotten Ventricle in Cardio-Oncology. Curr Probl Cardiol 2025:103039. [PMID: 40157515 DOI: 10.1016/j.cpcardiol.2025.103039] [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: 03/17/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
INTRODUCTION Cancer therapy-induced cardiotoxicity (CTRCD) is a significant adverse effect of oncologic treatment, associated with considerable morbidity and mortality. Among CTRCD, heart failure stands out in prevalence and severity, with left ventricular dysfunction being the focus of most studies. Right ventricle (RV) may also be damaged by CTRCD, however the effects of CTRCD on RV function (RVF) have not been elucidated. OBJECTIVE We aimed to conduct a systematic review and meta-analysis evaluating RV echocardiographic parameters in patients undergoing chemotherapy. METHODS PubMed, Embase and Cochrane were searched for studies that evaluated RV parameters during cancer therapy. Statistical analysis was performed using the R statistical software. We computed pooled mean differences (MD), adopting a random-effects model, with a significance level of 0.05. A correlation coefficient of 0.5 was assumed for paired measurements. Heterogeneity was assessed using the I² statistic. RESULTS We included 1,520 patients from 25 studies, 73% of whom were women and with a mean age of 51.1±16.5 years. RVF was significantly lower after CTRCD, with reduction in fractional area change (MD=-2.29% [95% CI: -3.63,-0.95]), RV global longitudinal strain (MD=2.49% [95% CI: 1.73, 3.25]), and RV free wall strain (MD=3.21% [95% CI: 2.32, 4.11]). Additionally, tricuspid annular plane systolic excursion was significantly reduced (MD=-1.44mm [95% CI: -1.94, -0.95]) and pulmonary artery systolic pressure was significantly higher (MD=1.60mmHg [95% CI: 0.64, 2.56]) after chemotherapy. CONCLUSION The assessment of RVF is important in CTRCD, and its quantification should be included in clinical follow-up during cancer treatment. Further research is needed to elucidate the underlying factors contributing to RV dysfunction and to develop methods for its early detection.
Collapse
Affiliation(s)
- Caroline O Fischer-Bacca
- Medical Sciences Research Center, University Center for the Development of Alto Vale - Rio do Sul - Brazil.
| | - Ramon Huntermann
- Medical Sciences Research Center, University Center for the Development of Alto Vale - Rio do Sul - Brazil
| | - Juan Peres de Oliveira
- Medical Sciences Research Center, University Center for the Development of Alto Vale - Rio do Sul - Brazil
| | | | | | - Rhanderson Cardoso
- Division of Cardiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Edielle S Melo
- Brazilian Israeli Beneficent Society Albert Einstein (SBIBAE), São Paulo, Brazil
| |
Collapse
|
7
|
Boen HM, Pype LL, Papadimitriou K, Altintas S, Teuwen LA, Anguille S, Saevels K, Verlinden A, Delrue L, Heggermont WA, Bosman M, Guns PJ, Heidbuchel H, Van De Heyning CM, Van Craenenbroeck EM, Franssen C. Dynamics of SERPINA3 in response to anthracycline treatment and cardiovascular dysfunction. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2025; 11:27. [PMID: 40087712 PMCID: PMC11907982 DOI: 10.1186/s40959-025-00324-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Accepted: 02/24/2025] [Indexed: 03/17/2025]
Abstract
BACKGROUND SERPINA3 recently emerged as potential prognostic biomarker in heart failure. In a population of cancer survivors with cancer therapy-related cardiac dysfunction (CTRCD) circulating SERPINA3 was elevated compared to age-matched controls. We aimed to assess the longitudinal dynamics of circulating SERPINA3 levels in patients with cancer treated with anthracycline chemotherapy (AnC) and its relation to CTRCD. METHODS In this single centre cohort study, 55 patients with cancer scheduled for AnC were prospectively enrolled. Cardiac evaluation (echocardiography, high-sensitive cardiac troponin I and NT-proBNP) was performed and SERPINA3 levels in plasma were assessed at 4 timepoints: before chemotherapy, directly after the end of chemotherapy, three months and twelve months after the end of chemotherapy. RESULTS Forty-two out of 55 patients (76.4%) developed CTRCD within 1 year after end of treatment. CTRCD was mild in 32 and moderate in 10 patients, defined as a change in cardiac biomarkers or GLS and LVEF decline < 50% respectively. Overall, median SERPINA3 levels decreased from baseline to three months after AnC (215.7 [62.0-984.0] to 176.9 [94.7-678.0] µg/ml, p = 0.031). This decrease was most prominent in patients without CTRCD (30.8% decrease, p = 0.007), followed by mild CTRCD (9.0% decrease, p = 0.022), while patients with moderate CTRCD did not show a reduction in SERPINA3 (5.1% increase, p = 0.987). SERPINA3 values at three months after AnC were positively correlated with NT-proBNP (r = 0.47, p = 0.002). Several malignancy, treatment and patient characteristics were associated with higher SERPINA3 values. CONCLUSION Circulating SERPINA3 levels show dynamic changes in a population of patients with cancer, with an overall decrease following AnC. However, in patients that developed moderate CTRCD, SERPINA3 levels remained elevated. The potential of SERPINA3 dynamics as a biomarker for CTRCD, deserves validation in larger cohorts.
Collapse
Affiliation(s)
- Hanne M Boen
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium.
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium.
| | - Lobke L Pype
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
| | | | - Sevilay Altintas
- Department of Oncology, Antwerp University Hospital, Antwerp, Belgium
| | - Laure-Anne Teuwen
- Department of Oncology, Antwerp University Hospital, Antwerp, Belgium
| | - Sébastien Anguille
- Department of Haematology, Antwerp University Hospital, Antwerp, Belgium
| | - Kirsten Saevels
- Department of Haematology, Antwerp University Hospital, Antwerp, Belgium
| | - Anke Verlinden
- Department of Haematology, Antwerp University Hospital, Antwerp, Belgium
| | - Leen Delrue
- Department of Cardiology, Olv Hospital Aalst, Aalst, Belgium
| | | | - Matthias Bosman
- Research Group Physiopharmacology, GENCOR, University of Antwerp, Antwerp, Belgium
| | - Pieter-Jan Guns
- Research Group Physiopharmacology, GENCOR, University of Antwerp, Antwerp, Belgium
| | - Hein Heidbuchel
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
| | - Caroline M Van De Heyning
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
| | - Emeline M Van Craenenbroeck
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
| | - Constantijn Franssen
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
| |
Collapse
|
8
|
Bhalraam U, Veerni RB, Paddock S, Meng J, Piepoli M, López-Fernández T, Tsampasian V, Vassiliou VS. Impact of sodium-glucose cotransporter-2 inhibitors on heart failure outcomes in cancer patients and survivors: a systematic review and meta-analysis. Eur J Prev Cardiol 2025:zwaf026. [PMID: 40044419 DOI: 10.1093/eurjpc/zwaf026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 11/30/2024] [Accepted: 01/28/2025] [Indexed: 03/20/2025]
Abstract
AIMS Sodium-glucose cotransporter-2 inhibitors (SGLT2i) are recognized for their cardiovascular benefits. This systematic review and meta-analysis evaluated the impact of SGLT2i on heart failure (HF) outcomes in cancer patients and survivors, focusing on HF hospitalization and new HF diagnoses. METHODS AND RESULTS A comprehensive search of PubMed, MEDLINE, and Embase via Ovid, and the Cochrane Library was conducted up to 5 June 2024, focusing on studies involving cancer patients and survivors treated with SGLT2i. The search criterion used was [(SGLT2) OR (Sodium glucose cotransporter 2 inhibitors) OR (canagliflozin) OR (dapagliflozin) OR (empagliflozin) OR (ertugliflozin) AND (cancer)]. The primary outcomes assessed were HF hospitalization and new HF diagnoses. The search yielded 1880 studies, from which 13 studies encompassing 88 273 patients were included. SGLT2i use reduced HF hospitalizations by 51% (RR 0.49, 95% CI 0.36-0.66, I² = 28%, P < 0.01) and new HF diagnoses by 71% (RR 0.29, 95% CI 0.10-0.87, I² = 71%). Multi-variate meta-regression analysis suggested that among breast cancer populations, studies with ≥50% of patients on anthracyclines exhibited a 99% reduction in HF hospitalization risk compared with similar studies that included <50% of patients on anthracyclines (RR 0.0085, 95% CI: 0.0001-0.2645, P = 0.0081). CONCLUSION SGLT2i significantly lower the risk of HF hospitalization and new HF diagnoses among cancer patients and survivors, with particularly pronounced benefits in breast cancer patients receiving anthracycline-based chemotherapy. These findings support the need for prospective trials to further investigate the integration of SGLT2i into cancer patient management to enhance cardiovascular outcomes.
Collapse
Affiliation(s)
- U Bhalraam
- Department of Medicine, Norwich Medical School, University of East Anglia, Norwich, UK
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, UK
| | - Rathna B Veerni
- Department of Medicine, Ninewells Hospital and Medical School, Dundee, UK
| | - Sophie Paddock
- Department of Medicine, Norwich Medical School, University of East Anglia, Norwich, UK
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, UK
| | - James Meng
- Department of Medicine, Norwich Medical School, University of East Anglia, Norwich, UK
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, UK
| | - Massimo Piepoli
- University Cardiology Department, IRCCS Policlinico San Donato, Via Morandi 30, 20097 San Donato Milanese, Milan, Italy
- Department of Biomedical Sciences for Health, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy
| | - Teresa López-Fernández
- Cardiology Department, La Paz University Hospital, IdiPAZ Research Institute, Madrid, Spain
- Cardiology Department, Quiron Pozuelo University Hospital, Madrid, Spain
| | - Vasiliki Tsampasian
- Department of Medicine, Norwich Medical School, University of East Anglia, Norwich, UK
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, UK
| | - Vassilios S Vassiliou
- Department of Medicine, Norwich Medical School, University of East Anglia, Norwich, UK
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, UK
| |
Collapse
|
9
|
Baş O, Güdük N, Tokatlı M, Güven DC, Özer N, Şener YZ, Akın S, Aksoy S, Barışta İ, Türker FA, Dizdar Ö. Serum Albumin-Creatinine Ratio and Anthracycline Cardiotoxicity in Patients with Cancer. J Clin Med 2025; 14:1741. [PMID: 40095884 PMCID: PMC11900434 DOI: 10.3390/jcm14051741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 02/24/2025] [Accepted: 03/03/2025] [Indexed: 03/19/2025] Open
Abstract
Background: Several studies have suggested that the serum albumin-creatinine ratio (sACR) is a useful marker for the early risk stratification of patients with cardiomyocyte injury. This study aims to evaluate the relationship between sACR and anthracycline-related cardiotoxicity. Methods: This study included patients who had received anthracycline-based chemotherapy between 2014 and 2023 and had undergone baseline and follow-up echocardiography after the treatment. The level of sACR was calculated using serum albumin and creatinine values obtained before the chemotherapy. The definition of cardiotoxicity was based on the criteria of the European Society of Cardiology (ESC) for ejection fraction and the American Society of Echocardiography (ASE) for diastolic dysfunction. The patients were categorized into either the high or low sACR group based on the cut-off value obtained from the receiver operating characteristic (ROC) curve analysis. Results: In total, 525 patients (159 males, 366 females) were included. Multivariate analysis after adjustment for age, body mass index (BMI), cardiovascular disease, hemoglobin, anthracycline dose, and gender showed that sACR (HR = 1.85% 95 CI 1.12 to 3.06 p = 0.016), cardiovascular disease (HR = 1.97% 95 CI 1.08 to 3.61 p = 0.027), BMI (HR = 1.86% 95 CI 1.12 to 3.10 p = 0.017), and age (HR = 1.02% 95 CI 1.001 to 1.04 p = 0.036) were significantly associated with an increased risk of cardiotoxicity. Conclusions: This study is the first to show a significant relationship between sACR and cardiotoxicity related to anthracycline use. Routine laboratory tests that are conducted before anthracycline therapy can aid clinicians in identifying high-risk patients who may require closer follow-up or cardioprotective measures.
Collapse
Affiliation(s)
- Onur Baş
- Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Hacettepe University, Sihhiye, 06100 Ankara, Turkey (S.A.); (F.A.T.)
| | - Naciye Güdük
- Department of Internal Medicine, Faculty of Medicine, Hacettepe University, Sihhiye, 06100 Ankara, Turkey; (N.G.)
| | - Mert Tokatlı
- Department of Internal Medicine, Faculty of Medicine, Hacettepe University, Sihhiye, 06100 Ankara, Turkey; (N.G.)
| | - Deniz Can Güven
- Elazig Fethi Sekin Sehir Hastanesi, Health Sciences University, 23300 Elazig, Turkey;
| | - Necla Özer
- Department of Cardiology, Faculty of Medicine, Hacettepe University, Sihhiye, 06100 Ankara, Turkey; (N.Ö.)
| | - Yusuf Ziya Şener
- Department of Cardiology, Faculty of Medicine, Hacettepe University, Sihhiye, 06100 Ankara, Turkey; (N.Ö.)
| | - Serkan Akın
- Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Hacettepe University, Sihhiye, 06100 Ankara, Turkey (S.A.); (F.A.T.)
| | - Sercan Aksoy
- Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Hacettepe University, Sihhiye, 06100 Ankara, Turkey (S.A.); (F.A.T.)
| | - İbrahim Barışta
- Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Hacettepe University, Sihhiye, 06100 Ankara, Turkey (S.A.); (F.A.T.)
| | - Fatma Alev Türker
- Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Hacettepe University, Sihhiye, 06100 Ankara, Turkey (S.A.); (F.A.T.)
| | - Ömer Dizdar
- Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Hacettepe University, Sihhiye, 06100 Ankara, Turkey (S.A.); (F.A.T.)
| |
Collapse
|
10
|
Travers S, Alexandre J, Baldassarre LA, Salem JE, Mirabel M. Diagnosis of cancer therapy-related cardiovascular toxicities: A multimodality integrative approach and future developments. Arch Cardiovasc Dis 2025; 118:185-198. [PMID: 39947997 DOI: 10.1016/j.acvd.2024.12.012] [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: 11/07/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 03/14/2025]
Abstract
Diagnosing cancer therapy-related cardiovascular toxicities may be a challenge. The interplay between cancer and cardiovascular diseases, beyond shared cardiovascular and cancer risk factors, and the increasingly convoluted cancer therapy schemes have complicated cardio-oncology. Biomarkers used in cardio-oncology include serum, imaging and rhythm modalities to ensure proper diagnosis and prognostic stratification of cardiovascular toxicities. For now, troponin and natriuretic peptides, multimodal cardiovascular imaging (led by transthoracic echocardiography combined with cardiac magnetic resonance or computed tomography angiography) and electrocardiography (12-lead or Holter monitor) are cornerstones in cardio-oncology. However, the imputability of cancer therapies is sometimes difficult to assess, and more refined biomarkers are currently being studied to increase diagnostic accuracy. Advances reside partly in pathophysiology-based serum biomarkers, improved cardiovascular imaging through new technical developments and remote monitoring for rhythm disorders. A multiparametric omics approach, enhanced by deep-learning techniques, should open a new era for biomarkers in cardio-oncology in the years to come.
Collapse
Affiliation(s)
- Simon Travers
- INSERM UMR-S 1180, Université Paris-Saclay, 91400 Orsay, France; Laboratoire de Biochimie, DMU BioPhyGen, Hôpital Européen Georges Pompidou, AP-HP, 75015 Paris, France.
| | - Joachim Alexandre
- INSERM U1086 ANTICIPE, Biology-Research Building, UNICAEN, Normandie University Group, 14000 Caen, France; Department of Pharmacology, Biology-Research Building, PICARO Cardio-Oncology Programme, Caen-Normandy University Hospital, 14000 Caen, France.
| | - Lauren A Baldassarre
- Cardiovascular Medicine, Yale School of Medicine, 06510 New Haven CT, United States of America.
| | - Joe Elie Salem
- CIC-1901, Department of Pharmacology, Hôpital Pitié-Salpêtrière, AP-HP, Sorbonne Université, INSERM, 75013 Paris, France.
| | - Mariana Mirabel
- Cardiology Department, Institut Mutualiste Montsouris, 75014 Paris, France.
| |
Collapse
|
11
|
Kumar L, Vijayvergiya R, Jain A, Singh C, Jain A, Prakash G, Khadwal A, Malhotra P. Incidence, Risk Factors and Early Prediction of Doxorubicin-Induced Cardiotoxicity by Global Longitudinal Strain and Cardiac Biomarkers in Indian Patients With Lymphoma: A Prospective Observational Study. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2025; 25:e143-e150. [PMID: 39572335 DOI: 10.1016/j.clml.2024.10.008] [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: 08/10/2024] [Revised: 10/05/2024] [Accepted: 10/10/2024] [Indexed: 02/16/2025]
Abstract
BACKGROUND Detecting anthracyclines-induced cardiotoxicity before the onset of left ventricular dysfunction could enable the timely initiation of cardioprotective measures. 2D-Echocardiography (ECHO) with global longitudinal strain (GLS) and cardiac biomarkers are valuable for the early prediction of cardiotoxicity. OBJECTIVES We aimed to evaluate the predictive utility of 2D-ECHO-GLS and cardiac biomarkers exclusively in patients with lymphoma treated with a doxorubicin-based regimen. METHODS The study included lymphoma patients ≥14 years of age of either sex who were planned for a doxorubicin-based regimen. All eligible patients underwent 2D-ECHO-GLS and cardiac biomarkers (troponin T and pro-brain natriuretic peptide) measurements at the baseline (V1), after 3rd chemotherapy cycle (V2), and after treatment completion (V3). Incidence, risk factors, and early predictors for cardiotoxicity were assessed using SPSS software version 25. The study was registered with CTRI (CTRI/2021/07/034518). RESULTS 40 patients (median age, 42 years) had evaluations available at all 3 time points. Three out of 40 (7.5%) patients developed cardiotoxicity at V3. Patients with cardiotoxicity had a significantly higher mean age (P = .045) and a greater incidence of hypertension (P = .012) than those without cardiotoxicity. At V2, the mean GLS threshold (-18.1%) and Δ GLS threshold ≥15% from baseline were significant early predictors of subsequent cardiotoxicity. Despite an exponential rise from V1 to V3, the cardiac biomarkers failed to predict cardiotoxicity. CONCLUSIONS Patients with lymphoma treated with doxorubicin-based regimens have a significant risk of developing cardiac dysfunction. A greater than 15% fall in GLS from baseline after 3rd chemotherapy cycle could predict subsequent cardiotoxicity.
Collapse
Affiliation(s)
- Lucky Kumar
- Department of Cardiology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Rajesh Vijayvergiya
- Department of Cardiology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Ankur Jain
- Department of Haematology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Charanpreet Singh
- Department of Clinical Hematology and Medical Oncology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Arihant Jain
- Department of Clinical Hematology and Medical Oncology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Gaurav Prakash
- Department of Clinical Hematology and Medical Oncology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Alka Khadwal
- Department of Clinical Hematology and Medical Oncology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Pankaj Malhotra
- Department of Clinical Hematology and Medical Oncology, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| |
Collapse
|
12
|
Xiong W, Li B, Pan J, Li D, Yuan H, Wan X, Zhang Y, Fu L, Zhang J, Lei M, Chang ACY. Mitochondrial Amount Determines Doxorubicin-Induced Cardiotoxicity in Cardiomyocytes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412017. [PMID: 39921259 PMCID: PMC11948046 DOI: 10.1002/advs.202412017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/30/2024] [Indexed: 02/10/2025]
Abstract
Doxorubicin, an anthracycline commonly used for treating cancer patients, is known for its cardiotoxic side-effects. Although dose-dependent, but susceptibility remains variable among patients, and childhood-exposure-adult-onset remains challenging. Besides topoisomerase toxicity, Doxorubicin is also toxic to the mitochondria yet the underlying late onset mechanism remains elusive. Here, it is observed that the mitochondrial copy number in PBMCs of patients treated with anthracycline chemotherapy is negatively correlated with the change in plasma BNP levels after treatment. Isogenic hiPSC-CMs are generated with high, norm, and low mitochondrial copy numbers using mitochondrial transplantation and the YFP-Parkin system. Remarkably, lower mitochondria copy number translates to lower IC50, suggesting increased susceptibility. Mitochondria supplementation by intramyocardial injection prevents doxorubicin induced heart failure. Mechanistically, doxorubicin treatment leads to mPTP opening and mitochondrial DNA (mtDNA) leakage. This mtDNA leakage event activates the cGAS-STING pathway and drives inflammation and myocardial senescence. Cardiomyocyte-specific knockout of Sting (Myh6-Cre/Stingflox/flox; StingCKO) and over expression of mitochondrial tagged DNase1 in mice partially rescue doxorubicin-induced cardiac dysfunction. In conclusion, the work establishes a negative correlation between cardiomyocyte mitochondrial copy number and doxorubicin toxicity. Molecularly, it is demonstrated that mtDNA leakage activates cGAS-STING pathway and accelerates myocardial dysfunction. These insights offer new co-administration strategies for cancer patients.
Collapse
Affiliation(s)
- Weiyao Xiong
- Department of CardiologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
- Shanghai Institute of Precision MedicineShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200125China
| | - Bin Li
- Department of CardiologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
- Shanghai Institute of Precision MedicineShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200125China
| | - Jianan Pan
- Department of CardiologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
| | - Dongjiu Li
- Department of CardiologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
| | - Haihua Yuan
- Department of OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
| | - Xin Wan
- Department of CardiologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
- Shanghai Institute of Precision MedicineShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200125China
| | - Yanjie Zhang
- Department of OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
| | - Lijun Fu
- Department of CardiologyShanghai Children's Medical CentreShanghai Jiao Tong University School of MedicineShanghai200127China
| | - Junfeng Zhang
- Department of CardiologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
| | - Ming Lei
- Shanghai Institute of Precision MedicineShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200125China
| | - Alex Chia Yu Chang
- Department of CardiologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
- Shanghai Institute of Precision MedicineShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200125China
| |
Collapse
|
13
|
Dowsing B, Dehbi HM, Chung R, Pedra J, Worn O, Artico J, Schmid P, Roylance R, Kellman P, Moon JC, Crake T, Westwood M, Ghosh A, Andres MS, Nazir MS, Lyon AR, Chen D, Walker M, Manisty CH. HER-SAFE study design: an open-label, randomised controlled trial to investigate the safety of withdrawal of pharmacological treatment for recovered HER2-targeted therapy-related cardiac dysfunction. BMJ Open 2025; 15:e091917. [PMID: 39909533 PMCID: PMC11800297 DOI: 10.1136/bmjopen-2024-091917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 01/20/2025] [Indexed: 02/07/2025] Open
Abstract
INTRODUCTION A quarter of breast cancers show human epidermal growth factor-2 (HER2) overexpression, where targeted therapy dramatically improves survival. However, cancer therapy-related cardiac dysfunction (CTRCD) occurs in up to 15% of patients. With the interruption of HER2 therapy, if necessary, and the initiation of heart failure therapy (HFT), HER2 CTRCD recovers in over 80% of cases. The need to continue HFT in 'recovered' HER2 CTRCD following completion of HER2 therapy is unclear and there are potential significant impacts on patient's quality of life (QoL). The Randomised Controlled Trial for the Safety of Withdrawal of Pharmacological Treatment for Recovered HER2 Targeted Therapy Related Cardiac Dysfunction (HER-SAFE) aims to evaluate whether HFT can be safely withdrawn in non-high cardiovascular (CV) risk patients with 'recovered' HER2 CTRCD. METHODS AND ANALYSIS This is a multicentre, open-label randomised controlled trial investigating whether withdrawal of HFT is non-inferior to continuation in non-high CV risk, breast cancer survivors with recovered HER2 CTRCD after cancer treatment completion. The primary endpoint is the incidence of guideline-defined cardiac dysfunction or clinical heart failure. Secondary endpoints include changes in cardiac blood biomarkers, cardiovascular magnetic resonance (CMR)-derived strain and tissue mapping and heart failure symptom questionnaires. The study will recruit 90 participants who will undergo serial clinical assessment over 12 months with advanced cardiovascular imaging (CMR scans with automated analysis at baseline, 6 and 12 months), cardiac biomarker measurement (six time points over 12 months), plus complete heart failure QoL and medication disutility questionnaires. This is the first multicentre study to address this significant clinical issue. ETHICS AND DISSEMINATION This study was approved by the research ethics committee (London-London Bridge, 23/LO/0152). The results will be disseminated in peer-reviewed scientific journals. TRIAL REGISTRATION NUMBER NCT05880160.
Collapse
Affiliation(s)
- Benjamin Dowsing
- Institute of Cardiovascular Science, University College London, London, UK
- Saint Bartholomew's Hospital Barts Heart Centre, London, UK
| | | | - Robin Chung
- Saint Bartholomew's Hospital Barts Heart Centre, London, UK
- Whittington Health NHS Trust, London, UK
| | - Joanna Pedra
- Saint Bartholomew's Hospital Barts Heart Centre, London, UK
| | - Orla Worn
- Saint Bartholomew's Hospital Barts Heart Centre, London, UK
| | - Jessica Artico
- Institute of Cardiovascular Science, University College London, London, UK
- Imperial College Healthcare NHS Trust Cardiology Service, London, UK
| | - Peter Schmid
- Queen Mary University of London, London, UK
- Barts Health NHS Trust, London, UK
| | | | - Peter Kellman
- National Heart Lung and Blood Institute, Bethesda, Maryland, USA
| | - James C Moon
- Institute of Cardiovascular Science, University College London, London, UK
- Saint Bartholomew's Hospital Barts Heart Centre, London, UK
| | - Tom Crake
- Saint Bartholomew's Hospital Barts Heart Centre, London, UK
| | - Mark Westwood
- Saint Bartholomew's Hospital Barts Heart Centre, London, UK
| | - Arjun Ghosh
- Institute of Cardiovascular Science, University College London, London, UK
- Saint Bartholomew's Hospital Barts Heart Centre, London, UK
| | - Maria Sol Andres
- Cardio-Oncology Centre of Excellence, Royal Brompton Hospital, London, UK
| | - Muhummad Sohaib Nazir
- King's College London School of Biomedical Engineering & Imaging Sciences, London, UK
- Cardio-Oncology Centre of Excellence, Royal Brompton and Harefield Hospitals, London, UK
| | - Alexander R Lyon
- Cardio-Oncology Centre of Excellence, Royal Brompton Hospital, London, UK
| | - Daniel Chen
- Institute of Cardiovascular Science, University College London, London, UK
- Saint Bartholomew's Hospital Barts Heart Centre, London, UK
| | - Malcolm Walker
- Institute of Cardiovascular Science, University College London, London, UK
| | - Charlotte H Manisty
- Institute of Cardiovascular Science, University College London, London, UK
- Saint Bartholomew's Hospital Barts Heart Centre, London, UK
| |
Collapse
|
14
|
Viñas-Mendieta AE, Gallardo-Grajeda A, López-Fernández T. Cardio-oncology: chances and challenges. Basic Res Cardiol 2025; 120:3-9. [PMID: 39348001 DOI: 10.1007/s00395-024-01080-y] [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: 06/02/2024] [Revised: 09/19/2024] [Accepted: 09/21/2024] [Indexed: 10/01/2024]
Abstract
Cardio-oncology is an emerging field that aims to ensure optimal cancer treatment while minimising cardiovascular toxicity. The management of cardiovascular toxicity is critical because it can lead to premature discontinuation of treatment, increasing the risk of cancer recurrence and mortality. The 2022 European Society of Cardiology guidelines were a milestone in advocating a patient-centred, multidisciplinary approach. Key components include risk stratification and a standardised criterion for adverse events, incorporating definitions from the International Cardio-Oncology Society. Effective risk stratification, supported by imaging and biomarkers, helps to anticipate cardiovascular problems and implement preventive measures. Future research should focus on understanding mechanisms, developing preventive strategies and implementing personalised medicine. Education and reducing disparities in care are essential to advance cardio-oncology and improve patient outcomes.
Collapse
Affiliation(s)
- Adriana E Viñas-Mendieta
- Cardio-Oncology Unit, Cardiology Department, La Paz University Hospital, IdiPAZ Research Institute, C/ Paseo de la Castellana nº 261, 28046, Madrid, Spain
- Cardiology Department, Guillermo Almenara Irigoyen Hospital, Lima, Peru
| | - Andrea Gallardo-Grajeda
- Cardio-Oncology Unit, Cardiology Department, La Paz University Hospital, IdiPAZ Research Institute, C/ Paseo de la Castellana nº 261, 28046, Madrid, Spain
- Cardiology Department, National Institute of Cardiology Ignacio Chavez, Mexico City, Mexico
| | - Teresa López-Fernández
- Cardio-Oncology Unit, Cardiology Department, La Paz University Hospital, IdiPAZ Research Institute, C/ Paseo de la Castellana nº 261, 28046, Madrid, Spain.
- Cardiology Department, Quironsalud Madrid University Hospital, Madrid, Spain.
| |
Collapse
|
15
|
Bonaca MP, Lang NN, Chen A, Amiri-Kordestani L, Lipka L, Zwiewka M, Strnadova C, Klaar S, Dent S, Janicijevic TK, Herrmann J, Barac A, de Boer RA, Deswal A, Schou M, Neilan TG, van der Meer P, Moslehi J, Kondapalli L, Ky B, Fernandez TL, Cornell RF, Flaig TW, Hsia J, Sharon E, de Azambuja E, Seltzer J, Januzzi JL, Petrie MC. Cardiovascular Safety in Oncology Clinical Trials: JACC: CardioOncology Primer. JACC CardioOncol 2025; 7:83-95. [PMID: 39967209 DOI: 10.1016/j.jaccao.2024.09.014] [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: 01/03/2024] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 02/20/2025] Open
Abstract
The development of novel treatments has improved cancer outcomes but may result in cardiovascular toxicities. Traditional approaches to clinical trial safety evaluation have limitations in their ability to detect signals of cardiovascular risk. Mechanisms to increase power and specificity to clarify cardiovascular safety are required. However, implications include increased costs and slower development. The Cardiovascular Safety Research Consortium facilitated stakeholder discussions with representation from academia, industry, and regulators. A think tank was assembled with the aim of providing recommendations for improved collection and reporting of cardiovascular safety signals in oncology trials. Two working groups were formed. The first focuses on incorporation of consensus definitions of cardiovascular disease into the Common Terminology Criteria for Adverse Events used in oncology trial reporting. The second group considers methods for ascertainment and adjudication of cardiovascular events in cancer trials. The overarching aim of this primer is to improve understanding of the potential cardiovascular toxicities of cancer therapies.
Collapse
Affiliation(s)
- Marc P Bonaca
- CPC Clinical Research, Cardiology & Vascular medicine, University of Colorado Anschutz Medical School, Aurora, Colorado, USA.
| | - Ninian N Lang
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom. https://twitter.com/ninianlang
| | - Alice Chen
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland, USA
| | - Laleh Amiri-Kordestani
- Center for Drug Evaluation and Research (CDER), and Oncology Center of Excellence (OCE), U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | | | - Michal Zwiewka
- Paul-Ehrlich-Institut, Bundesinstitut für Impfstoffe und biomedizinische Arzneimittel, Federal Institute for Vaccines and Biomedicines, Langen, Germany
| | | | | | - Susan Dent
- Duke Cancer Institute, Duke University, Durham, North Carolina, USA. https://twitter.com/sdent_cardioonc
| | | | - Joerg Herrmann
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA. https://twitter.com/mayocvonc
| | - Ana Barac
- Inova Schar Cancer Institute and Inova Heart and Vascular Institute, Annandale, Virginia, USA. https://twitter.com/AnaBaracCardio
| | - Rudolf A de Boer
- Erasmus MC, Cardiovascular Institute, Thorax Center, Department of Cardiology, Rotterdam, the Netherlands
| | - Anita Deswal
- Department of Cardiology, University of Texas, MD Anderson Cancer Center, Houston, Texas, USA. https://twitter.com/anita_deswal
| | - Morten Schou
- Herlev-Gentofte Hospital, University of Copenhagen, Herlev-Gentofte, Denmark. https://twitter.com/mortschou
| | - Tomas G Neilan
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA. https://twitter.com/TomasNeilan
| | - Peter van der Meer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Javid Moslehi
- Section of Cardio-Oncology & Immunology, Division of Cardiology and the Cardiovascular Research Institute, University of California-San Francisco, San Francisco, California, USA
| | | | - Bonnie Ky
- Perelman School of Medicine the University of Pennsylvania, Thalheimer Center for Cardio-Oncology, Philadelphia, Pennsylvania, USA
| | - Teresa Lopez Fernandez
- Cardiology Department, Institute for Health Research, La Paz University Hospital, Madrid, Spain; Cardiology Department, Quirón Pozuelo University Hospital, Madrid, Spain. https://twitter.com/TeresaLpezFdez1
| | | | - Thomas W Flaig
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Judith Hsia
- CPC Clinical Research, Cardiology & Vascular medicine, University of Colorado Anschutz Medical School, Aurora, Colorado, USA
| | - Elad Sharon
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA. https://twitter.com/EladSharonMD
| | - Evandro de Azambuja
- Institute Jules Bordet and l'Université Libre de Bruxelles (L.U.B), Brussels, Belgium. https://twitter.com/E_de_Azambuja
| | | | - James L Januzzi
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School; Heart Failure and Biomarker Trials, Baim Institute for Clinical Research, Boston, Massachusetts, USA. https://twitter.com/jjheart_doc
| | - Mark C Petrie
- University of Glasgow, Glasgow, United Kingdom. https://twitter.com/markcpetrie20
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
Araújo DC, Simões R, Sabino ADP, Oliveira AND, Oliveira CMD, Veloso AA, Gomes KB. Predicting doxorubicin-induced cardiotoxicity in breast cancer: leveraging machine learning with synthetic data. Med Biol Eng Comput 2025:10.1007/s11517-025-03289-y. [PMID: 39828884 DOI: 10.1007/s11517-025-03289-y] [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: 04/25/2024] [Accepted: 11/29/2024] [Indexed: 01/22/2025]
Abstract
Doxorubicin (DOXO) is a primary treatment for breast cancer but can cause cardiotoxicity in over 25% of patients within the first year post-chemotherapy. Recognizing at-risk patients before DOXO initiation offers pathways for alternative treatments or early protective actions. We analyzed data from 78 Brazilian breast cancer patients, with 34.6% developing cardiotoxicity within a year of their final DOXO dose. To address the limited sample size, we utilized the DAS (Data Augmentation and Smoothing) method, creating 4892 synthetic samples that exhibited high statistics fidelity to the original data. By integrating routine blood biomarkers (C-Reactive protein, total cholesterol, LDL-c, HDL-c, hematocrit, and hemoglobin) and two clinical measures (weighted smoking status and body mass index), our model achieved an AUROC of 0.85±0.10, a sensitivity of 0.89, and a specificity of 0.69, positioning it as a potential screening instrument. Notably, DAS outperformed the established methods, Adaptive Synthetic Sampling (ADASYN), Synthetic Minority Over-Sampling Technique (SMOTE), and Synthetic Data Vault (SDV), underscoring its promise for medical synthetic data generation and pioneering a cardiotoxicity prediction model specifically for DOXO.
Collapse
Affiliation(s)
- Daniella Castro Araújo
- Huna, São Paulo, SP, Brazil.
- Departamento de Ciência da Computação, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| | - Ricardo Simões
- Faculdade de Farmàcia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
- Faculdade Ciências Médicas de Minas Gerais, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | | | | | - Adriano Alonso Veloso
- Departamento de Ciência da Computação, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Karina Braga Gomes
- Faculdade de Farmàcia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| |
Collapse
|
19
|
Alshahrani AA, Kontopantelis E, Morgan C, Ravindrarajah R, Martin GP, Mamas MA. Cardiovascular diseases in patients with cancer: A comprehensive review of epidemiological trends, cardiac complications, and prognostic implications. Chin Med J (Engl) 2025; 138:143-154. [PMID: 39719690 PMCID: PMC11745857 DOI: 10.1097/cm9.0000000000003419] [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: 05/28/2024] [Indexed: 12/26/2024] Open
Abstract
ABSTRACT This article provides an overview of the current evidence on the epidemiology, overlapping risk factors, and pathophysiology of cardiovascular disease (CVD) in patients with cancer. It explores the cardiotoxic effects of anticancer therapy and their impact on prognosis. Although cancer survival rates have improved over the last two decades, the risk of CVD has risen over time in patients with cancer. CVD and cancer share similar risk factors and a common pathophysiology involving inflammation. Many chemotherapeutic agents used to treat cancer are associated with cardiovascular complications (such as heart failure, myocardial infarction, and thrombosis). Current evidence indicates a significant burden of CVD in patients with cancer, particularly in the first year following cancer diagnosis, with elevated risk persisting beyond this period. This short- and long-term risk of CVD may vary depending on the cancer type and treatment regimen. Early identification of potential cardiovascular risk in patients with cancer, can lead to more favorable clinical and survival outcomes. Given the acute and long-term consequences, patients with cancer require increased cardiovascular care and lifestyle optimization. This article offers valuable insights into the cardiovascular burden and needs of patients with cancer. It is intended for a general medical research readership interested in the intersection of cardiology and oncology.
Collapse
Affiliation(s)
- Ali A. Alshahrani
- Faculty of Biology, Medicine and Health, School of Health Sciences, University of Manchester, Manchester M13 9QQ, United Kingdom
- Department of Invasive Cardiovascular Technology, College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh 3129, Saudi Arabia
| | - Evangelos Kontopantelis
- Faculty of Biology, Medicine and Health, School of Health Sciences, University of Manchester, Manchester M13 9QQ, United Kingdom
| | - Catharine Morgan
- Faculty of Biology, Medicine and Health, School of Health Sciences, University of Manchester, Manchester M13 9QQ, United Kingdom
| | - Rathi Ravindrarajah
- Faculty of Biology, Medicine and Health, School of Health Sciences, University of Manchester, Manchester M13 9QQ, United Kingdom
| | - Glen P. Martin
- Faculty of Biology, Medicine and Health, School of Health Sciences, University of Manchester, Manchester M13 9QQ, United Kingdom
| | - Mamas A. Mamas
- Keele Cardiovascular Research Group, Institute for Prognosis Research, University of Keele, Keele ST5 5BG, United Kingdom
| |
Collapse
|
20
|
Rivero-Santana B, Saldaña-García J, Caro-Codón J, Zamora P, Moliner P, Martínez Monzonis A, Zatarain E, Álvarez-Ortega C, Gómez-Prieto P, Pernas S, Rodriguez I, Buño Soto A, Cadenas R, Palacios Ozores P, Pérez Ramírez S, Merino Salvador M, Valbuena S, Fernández Gasso L, Juárez V, Severo A, Terol B, de Soto Álvarez T, Rodríguez O, Brion M, González-Costello J, Canales Albendea M, González-Juanatey JR, Moreno R, López-Sendón J, López-Fernández T. Anthracycline-induced cardiovascular toxicity: validation of the Heart Failure Association and International Cardio-Oncology Society risk score. Eur Heart J 2025; 46:273-284. [PMID: 39106857 DOI: 10.1093/eurheartj/ehae496] [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: 02/16/2024] [Revised: 04/24/2024] [Accepted: 07/18/2024] [Indexed: 08/09/2024] Open
Abstract
BACKGROUND AND AIMS Baseline cardiovascular toxicity risk stratification is critical in cardio-oncology. The Heart Failure Association (HFA) and International Cardio-Oncology Society (ICOS) score aims to assess this risk but lacks real-life validation. This study validates the HFA-ICOS score for anthracycline-induced cardiovascular toxicity. METHODS Anthracycline-treated patients in the CARDIOTOX registry (NCT02039622) were stratified by the HFA-ICOS score. The primary endpoint was symptomatic or moderate to severe asymptomatic cancer therapy-related cardiac dysfunction (CTRCD), with all-cause mortality and cardiovascular mortality as secondary endpoints. RESULTS The analysis included 1066 patients (mean age 54 ± 14 years; 81.9% women; 24.5% ≥65 years). According to the HFA-ICOS criteria, 571 patients (53.6%) were classified as low risk, 333 (31.2%) as moderate risk, 152 (14.3%) as high risk, and 10 (0.9%) as very high risk. Median follow-up was 54.8 months (interquartile range 24.6-81.8). A total of 197 patients (18.4%) died, and 718 (67.3%) developed CTRCD (symptomatic: n = 45; moderate to severe asymptomatic: n = 24; and mild asymptomatic: n = 649). Incidence rates of symptomatic or moderate to severe symptomatic CTRCD and all-cause mortality significantly increased with HFA-ICOS score [hazard ratio 28.74, 95% confidence interval (CI) 9.33-88.5; P < .001, and hazard ratio 7.43, 95% CI 3.21-17.2; P < .001) for very high-risk patients. The predictive model demonstrated good calibration (Brier score 0.04, 95% CI 0.03-0.05) and discrimination (area under the curve 0.78, 95% CI 0.70-0.82; Uno's C-statistic 0.78, 95% CI 0.71-0.84) for predicting symptomatic or severe/moderate asymptomatic CTRCD at 12 months. CONCLUSIONS The HFA-ICOS score effectively categorizes patients by cardiovascular toxicity risk and demonstrates strong predictive ability for high-risk anthracycline-related cardiovascular toxicity and all-cause mortality.
Collapse
Affiliation(s)
- Borja Rivero-Santana
- Cardiology Department, La Paz University Hospital, IdiPAZ Research Institute, C/Paseo de la Castellana n° 261, 28046 Madrid, Spain
| | - Jesús Saldaña-García
- Cardiology Department, La Paz University Hospital, IdiPAZ Research Institute, C/Paseo de la Castellana n° 261, 28046 Madrid, Spain
| | - Juan Caro-Codón
- Cardiology Department, La Paz University Hospital, IdiPAZ Research Institute, C/Paseo de la Castellana n° 261, 28046 Madrid, Spain
| | - Pilar Zamora
- Oncology Department, La Paz University Hospital, IdiPAZ Research Institute, CIBER ONC, Madrid, Spain
| | - Pedro Moliner
- Cardiology Department, Bellvitge University Hospital, Bio-Heart Cardiovascular Diseases Research Group, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, CIBER CV, Barcelona, Spain
| | - Amparo Martínez Monzonis
- Cardiology Department, Clinic University Hospital, IDIS Research Institute, CIBERCV, Santiago de Compostela, Spain
| | - Eduardo Zatarain
- Cardiology Department, Hospital General Universitario Gregorio Marañón, CIBER-CV (ISCIII), IISGM, Complutense University, Madrid, Spain
| | - Carlos Álvarez-Ortega
- Cardiology Department, La Paz University Hospital, IdiPAZ Research Institute, C/Paseo de la Castellana n° 261, 28046 Madrid, Spain
| | - Pilar Gómez-Prieto
- Hematology Department, La Paz University Hospital, IdiPAZ Research Institute, Madrid, Spain
| | - Sonia Pernas
- Medical Oncology Department, Institut Catala d'Oncologia-H.U.Bellvitge-IDIBELL, L'Hospitalet, Barcelona, Spain
| | - Isabel Rodriguez
- Radiation Oncology Department, La Paz University Hospital, IdiPAZ Research Institute, Madrid, Spain
| | - Antonio Buño Soto
- Department of Laboratory Medicine, La Paz University Hospital, IdiPAZ Research Institute, Madrid, Spain
| | - Rosalía Cadenas
- Cardiology Department, Infanta Sofía University Hospital, European University of Madrid, Madrid, Spain
| | - Patricia Palacios Ozores
- Oncology Department, Oncology Translational Research Group, Clinic University Hospital, IDIS Research Institute, Santiago de Compostela, Spain
| | | | - María Merino Salvador
- Medical Oncology Department, Infanta Sofía University Hospital, Infanta Sofía University Hospital, Henares University Hospital Foundation for Biomedical Research and Innovation (FIIB HUIS HHEN), Madrid, Spain
| | - Silvia Valbuena
- Cardiology Department, La Paz University Hospital, IdiPAZ Research Institute, C/Paseo de la Castellana n° 261, 28046 Madrid, Spain
| | - Lucía Fernández Gasso
- Cardiology Department, La Paz University Hospital, IdiPAZ Research Institute, C/Paseo de la Castellana n° 261, 28046 Madrid, Spain
| | - Victor Juárez
- Cardiology Department, Hospital Universitario 12 de Octubre, CIBER CV, Madrid, Spain
| | - Andrea Severo
- Cardiology Department, Hospital Universitario 12 de Octubre, CIBER CV, Madrid, Spain
| | - Belén Terol
- Cardiology Department, Hospital Universitario Quironsalud, C. Diego de Velázquez, 1, 28223 Pozuelo de Alarcón, Madrid, Spain
| | - Teresa de Soto Álvarez
- Hematology Department, La Paz University Hospital, IdiPAZ Research Institute, Madrid, Spain
| | - Olaia Rodríguez
- Department of Laboratory Medicine, La Paz University Hospital, IdiPAZ Research Institute, Madrid, Spain
| | - María Brion
- Cardiology Department, Clinic University Hospital, IDIS Research Institute, CIBERCV, Santiago de Compostela, Spain
| | - José González-Costello
- Cardiology Department, Bellvitge University Hospital, Bio-Heart Cardiovascular Diseases Research Group, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, CIBER CV, Barcelona, Spain
| | | | - José R González-Juanatey
- Cardiology Department, Clinic University Hospital, IDIS Research Institute, CIBERCV, Santiago de Compostela, Spain
| | - Raúl Moreno
- Cardiology Department, La Paz University Hospital, IdiPAZ Research Institute, C/Paseo de la Castellana n° 261, 28046 Madrid, Spain
| | | | - Teresa López-Fernández
- Cardiology Department, La Paz University Hospital, IdiPAZ Research Institute, C/Paseo de la Castellana n° 261, 28046 Madrid, Spain
- Cardiology Department, Hospital Universitario Quironsalud, C. Diego de Velázquez, 1, 28223 Pozuelo de Alarcón, Madrid, Spain
| |
Collapse
|
21
|
Avagimyan A, Kakturskiy L, Pogosova N, Ottaviani G, Rizzo M, Sarrafzadegan N. Doxorubicin and cyclophosphamide mode of chemotherapy-related cardiomyopathy: Review of preclinical model. Curr Probl Cardiol 2025; 50:102882. [PMID: 39427867 DOI: 10.1016/j.cpcardiol.2024.102882] [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: 10/04/2024] [Accepted: 10/08/2024] [Indexed: 10/22/2024]
Abstract
Over the past 70 years, there has been extensive research focused on preventing chemotherapy-related cardiovascular complications. However, the current state of cardio-oncology research has raised more questions than answers. Experimental studies often present data that are difficult to compare and, at times, contradictory. One notable limitation in translating experimental findings to clinical practice is the reliance on models that administer only one chemotherapeutic drug to experimental animals, despite the common use of multidrug cancer treatments in real clinical settings. This article aims to discuss our own experience in modeling an experimental rat model of cardiomyopathy induced by the administration of two chemotherapeutic drugs, doxorubicin (adriamycin) and cyclophosphamide (AC mode of chemotherapy) - Avagimyan A., et al model, along with a subsequent review of morphological changes based on our personal archive.
Collapse
Affiliation(s)
- Ashot Avagimyan
- Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Lev Kakturskiy
- A.P. Avtsyn Research Institute of Human Morphology, Petrovskiy NRCS, Moscow, Russia
| | - Nana Pogosova
- National Medical Research Centre of Cardiology after acad. E. I. Chazov, Moscow, Russia; Peoples' Friendship University of Russia after Patrice Lumumba (RUDN), Moscow, Russia
| | - Giulia Ottaviani
- Lino Rossi Research Center, Università degli Studi di Milano, Milan, Italy
| | | | - Nizal Sarrafzadegan
- Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran; University of British Columbia, Vancouver, Canada
| |
Collapse
|
22
|
Caballero-Valderrama MDR, Bevilacqua E, Echevarría M, Salvador-Bofill FJ, Ordóñez A, López-Haldón JE, Smani T, Calderón-Sánchez EM. Early Myocardial Strain Reduction and miR-122-5p Elevation Associated with Interstitial Fibrosis in Anthracycline-Induced Cardiotoxicity. Biomedicines 2024; 13:45. [PMID: 39857629 PMCID: PMC11762338 DOI: 10.3390/biomedicines13010045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/12/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025] Open
Abstract
Echocardiographic myocardial strain is crucial for early detection of anthracycline-induced cardiotoxicity, particularly in patients at moderate or high risk. BACKGROUND/OBJECTIVES This study investigates changes in global longitudinal strain (GLS) in breast cancer patients with low baseline risk for cardiotoxicity during cancer therapy. We also examined the relationship between echocardiographic strain, structural myocardial changes, and microRNA (miRNA) dysregulation associated with cancer treatment using an animal model. METHODS Echocardiography and blood tests were examined in 33 breast cancer patients with low baseline risk for cardiotoxicity during anthracycline treatment, with a follow-up at 12 months. Additionally, 16 Wistar rats received epirubicin (20 mg/kg over 4 weeks) to examine cardiac strain and structural changes. Moreover, circulating miRNA levels were assessed in patients' serum using microarray at the end of the treatment and further analyzed in peripheral blood from the animal model. RESULTS Pathological GLS values were observed in 27.27% of patients after four cycles, with 15.15% showing reduced left ventricular ejection fraction (LVEF) after 12 months. In the animal model, epirubicin-induced circumferential strain (CS) decrease correlates with myocardial fibrosis assessed histologically and by a significant increase in COL1 and TGFB2 expression. Furthermore, we found a significant decrease in aquaporin1 expression associated with the presence of vacuoles in treated rats. Furthermore, dysregulation in the expression of miRNAs was observed in patients with cardiotoxicity. Among them, hsa-miR-122-5p is increased in both patient and rat serum post-treatment. CONCLUSIONS A notable percentage of low-risk patients exhibited cardiac strain reduction due to cardiotoxicity. Epirubicin treatment caused structural heart changes in rats, highlighting miR-122-5p as a potential fibrosis marker that correlated with echocardiographic parameters.
Collapse
Affiliation(s)
- María de Regla Caballero-Valderrama
- Cardiology Unit, University Hospital Virgen del Rocío, 41013 Seville, Spain; (M.d.R.C.-V.); (J.E.L.-H.)
- Cardiovascular Pathophysiology Group, Institute of Biomedicine of Seville-IBiS, University of Seville/Hospital Universitario Virgen de Rocio/CSIC, 41013 Seville, Spain; (E.B.); (A.O.); (T.S.)
| | - Elisa Bevilacqua
- Cardiovascular Pathophysiology Group, Institute of Biomedicine of Seville-IBiS, University of Seville/Hospital Universitario Virgen de Rocio/CSIC, 41013 Seville, Spain; (E.B.); (A.O.); (T.S.)
| | - Miriam Echevarría
- Department of Medical Physiology and Biophysics, School of Medicine, University of Seville, 41009 Seville, Spain;
| | | | - Antonio Ordóñez
- Cardiovascular Pathophysiology Group, Institute of Biomedicine of Seville-IBiS, University of Seville/Hospital Universitario Virgen de Rocio/CSIC, 41013 Seville, Spain; (E.B.); (A.O.); (T.S.)
| | - José Eduardo López-Haldón
- Cardiology Unit, University Hospital Virgen del Rocío, 41013 Seville, Spain; (M.d.R.C.-V.); (J.E.L.-H.)
- Cardiovascular Pathophysiology Group, Institute of Biomedicine of Seville-IBiS, University of Seville/Hospital Universitario Virgen de Rocio/CSIC, 41013 Seville, Spain; (E.B.); (A.O.); (T.S.)
| | - Tarik Smani
- Cardiovascular Pathophysiology Group, Institute of Biomedicine of Seville-IBiS, University of Seville/Hospital Universitario Virgen de Rocio/CSIC, 41013 Seville, Spain; (E.B.); (A.O.); (T.S.)
- Department of Medical Physiology and Biophysics, School of Medicine, University of Seville, 41009 Seville, Spain;
| | - Eva M. Calderón-Sánchez
- Cardiovascular Pathophysiology Group, Institute of Biomedicine of Seville-IBiS, University of Seville/Hospital Universitario Virgen de Rocio/CSIC, 41013 Seville, Spain; (E.B.); (A.O.); (T.S.)
- Department of Medical Physiology and Biophysics, School of Medicine, University of Seville, 41009 Seville, Spain;
| |
Collapse
|
23
|
Romann SW, Giannitsis E, Frey N, Lehmann LH. Troponin Elevation in Asymptomatic Cancer Patients: Unveiling Connections and Clinical Implications. Curr Heart Fail Rep 2024; 21:505-514. [PMID: 39254897 PMCID: PMC11511716 DOI: 10.1007/s11897-024-00681-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: 08/22/2024] [Indexed: 09/11/2024]
Abstract
PURPOSE OF THE REVIEW Elevated troponin levels are well established e.g., for the diagnosis of suspected acute coronary syndrome in symptomatic patients. In contrast, troponin elevations in asymptomatic cancer patients emerge as a complex phenomenon, challenging traditional perceptions of its association solely with cardiac events. RECENT FINDINGS Recent data support the predictive value of cardiac biomarker for all-cause mortality and cardiotoxicity in cancer patients. This review gives an overview about the current literature about cardiac troponins in prediction and identification of high-risk cancer patients. The overview is focusing on diagnostic challenges, biomarker significance, and gaps of knowledge. Latest publications highlight the relevance of cardiac troponin in risk analysis before cancer treatment as well as a potential diagnostic gatekeeper for further cardiological diagnostics and therapy.
Collapse
Affiliation(s)
- Sebastian W Romann
- Department of Internal Medicine III: Cardiology, Angiology & Pulmonology, Cardio-Oncology Unit, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 69120, Heidelberg, Germany
| | - Evangelos Giannitsis
- Department of Internal Medicine III: Cardiology, Angiology & Pulmonology, Cardio-Oncology Unit, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 69120, Heidelberg, Germany
| | - Norbert Frey
- Department of Internal Medicine III: Cardiology, Angiology & Pulmonology, Cardio-Oncology Unit, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 69120, Heidelberg, Germany
| | - Lorenz H Lehmann
- Department of Internal Medicine III: Cardiology, Angiology & Pulmonology, Cardio-Oncology Unit, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 69120, Heidelberg, Germany.
- German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany.
| |
Collapse
|
24
|
Zhang JY, Li XY, Li DX, Zhang ZH, Hu LQ, Sun CX, Zhang XN, Wu M, Liu LT. Endoplasmic reticulum stress in intestinal microecology: A controller of antineoplastic drug-related cardiovascular toxicity. Biomed Pharmacother 2024; 181:117720. [PMID: 39631125 DOI: 10.1016/j.biopha.2024.117720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024] Open
Abstract
Endoplasmic reticulum (ER) stress is extensively studied as a pivotal role in the pathological processes associated with intestinal microecology. In antineoplastic drug treatments, ER stress is implicated in altering the permeability of the mechanical barrier, depleting the chemical barrier, causing dysbiosis, exacerbating immune responses and inflammation in the immune barrier. Enteric dysbiosis and intestinal dysfunction significantly affect the circulatory system in various heart disorders. In antineoplastic drug-related cardiovascular (CV) toxicity, ER stress constitutes a web of relationships in the host-microbiome symbiotic regulatory loop. Therefore, understanding the holobiont perspective will help de-escalate spatial and temporal restrictions. This review investigates the role of ER stress-mediated gut microecological alterations in antineoplastic treatment-induced CV toxicity.
Collapse
Affiliation(s)
- Jing-Yi Zhang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xiao-Ya Li
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - De-Xiu Li
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Zi-Hao Zhang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Lan-Qing Hu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Chang-Xin Sun
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xiao-Nan Zhang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China.
| | - Min Wu
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Long-Tao Liu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China.
| |
Collapse
|
25
|
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: 2] [Impact Index Per Article: 2.0] [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
|
26
|
López-Fernández T, Marco I, Aznar MC, Barac A, Bergler-Klein J, Meattini I, Scott JM, Cardinale D, Dent S. Breast cancer and cardiovascular health. Eur Heart J 2024; 45:4366-4382. [PMID: 39320463 DOI: 10.1093/eurheartj/ehae637] [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: 12/29/2023] [Revised: 03/08/2024] [Accepted: 09/06/2024] [Indexed: 09/26/2024] Open
Abstract
Modern cancer therapies greatly improve clinical outcomes for both early and advanced breast cancer patients. However, these advances have raised concerns about potential short- and long-term toxicities, including cardiovascular toxicities. Therefore, understanding the common risk factors and underlying pathophysiological mechanisms contributing to cardiovascular toxicity is essential to ensure best breast cancer outcomes. While cardio-oncology has emerged as a sub-speciality to address these challenges, it is essential that all cardiologists recognize and understand the cardiovascular consequences of cancer therapy. This review aims to provide a comprehensive overview of the potential adverse cardiovascular effects associated with modern breast cancer therapies. A preventive, diagnostic, and therapeutic workflow to minimize the impact of cardiovascular toxicity on patient outcomes is presented. Key aspects of this workflow include regular monitoring of cardiovascular function, early detection and management of cancer therapy-related cardiovascular toxicities, and optimization of cardiovascular risk factor control. By highlighting the gaps in knowledge in some areas, this review aims to emphasize the critical role of cardio-oncology research in ensuring the holistic well-being of patients with breast cancer.
Collapse
Affiliation(s)
- Teresa López-Fernández
- Cardiology Department, La Paz University Hospital, IdiPAZ Research Institute, C/Paseo de la Castellana n° 261, 28046 Madrid, Spain
- Cardiology Department, Quironsalud University Hospital, C. Diego de Velázquez, 1, 28223 Pozuelo de Alarcón, Madrid, Spain
| | - Irene Marco
- Cardiology Department, La Paz University Hospital, IdiPAZ Research Institute, C/Paseo de la Castellana n° 261, 28046 Madrid, Spain
| | - Marianne C Aznar
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Ana Barac
- Inova ScharHeart and Vascular, Inova Schar Cancer Institute, Fall Church, VA, USA
| | - Jutta Bergler-Klein
- Department of Cardiology, University Clinic of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Icro Meattini
- Department of Experimental and Clinical Biomedical Sciences 'M. Serio', Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Jessica M Scott
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Daniela Cardinale
- Cardioncology Unit, European Institute of Oncology, I.R.C.C.S., Milan, Italy
| | - Susan Dent
- Wilmot Cancer Institute, Department of Medicine, University of Rochester, Rochester, NY, USA
| |
Collapse
|
27
|
Stephenson E, Mclaughlin M, Bray JW, Saxton JM, Vince RV. Nutrition Modulation of Cardiotoxicity in Breast Cancer: A Scoping Review. Nutrients 2024; 16:3777. [PMID: 39519610 PMCID: PMC11547447 DOI: 10.3390/nu16213777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/25/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND/OBJECTIVES Advancements in breast cancer therapeutics, such as anthracyclines, are improving cancer survival rates but can have side effects that limit their use. Cardiotoxicity, defined as damage to the heart caused by cancer therapeutics, is characterised by a significant reduction in left ventricular ejection fraction (LVEF) and symptoms of cardiac dysfunction. Multiple oral supplements exist with antioxidant and anti-inflammatory properties that have the potential to lower cardiotoxicity risk and ameliorate the complications associated with left ventricular dysfunction. In this review, we evaluate the current status of using nutritional interventions to modulate cardiotoxicity. METHODS We used specific keywords to search for articles that met our predetermined inclusion and exclusion criteria to review the evidence and provide insights for future research. RESULTS Seven studies were identified as eligible for this review: six focused on oral supplementation strategies in breast cancer patients undergoing chemotherapy, and one focused on nutritional counselling and adherence to the Mediterranean diet in breast cancer survivors' post-treatment. There was a significantly attenuated reduction in LVEF in five studies that monitored cardiometabolic health, and there were significant improvements in blood serum levels of cardiac biomarkers across all studies. CONCLUSIONS Current evidence suggests that appropriate nutritional interventions, alongside chemotherapy, can modulate the risk of cardiotoxic side effects. This highlights the potential of oral antioxidant supplementation and Mediterranean diet counselling to decrease tertiary cancer therapy costs associated with cardiovascular complications.
Collapse
Affiliation(s)
- Emma Stephenson
- School of Sport, Exercise and Rehabilitation Sciences, University of Hull, Hull HU6 7RX, UK (M.M.); (J.W.B.); (J.M.S.)
| | - Marie Mclaughlin
- School of Sport, Exercise and Rehabilitation Sciences, University of Hull, Hull HU6 7RX, UK (M.M.); (J.W.B.); (J.M.S.)
- Physical Activity for Health Research Centre, Institute for Sport, P.E. and Health Sciences, Moray House School of Education and Sport, University of Edinburgh, Edinburgh EH8 8AQ, UK
| | - James W. Bray
- School of Sport, Exercise and Rehabilitation Sciences, University of Hull, Hull HU6 7RX, UK (M.M.); (J.W.B.); (J.M.S.)
| | - John M. Saxton
- School of Sport, Exercise and Rehabilitation Sciences, University of Hull, Hull HU6 7RX, UK (M.M.); (J.W.B.); (J.M.S.)
| | - Rebecca V. Vince
- School of Sport, Exercise and Rehabilitation Sciences, University of Hull, Hull HU6 7RX, UK (M.M.); (J.W.B.); (J.M.S.)
| |
Collapse
|
28
|
Avagimyan A, Pogosova N, Kakturskiy L, Sheibani M, Challa A, Kogan E, Fogacci F, Mikhaleva L, Vandysheva R, Yakubovskaya M, Faggiano A, Carugo S, Urazova O, Jahanbin B, Lesovaya E, Polana S, Kirsanov K, Sattar Y, Trofimenko A, Demura T, Saghazadeh A, Koliakos G, Shafie D, Alizadehasl A, Cicero A, Costabel JP, Biondi-Zoccai G, Ottaviani G, Sarrafzadegan N. Doxorubicin-related cardiotoxicity: review of fundamental pathways of cardiovascular system injury. Cardiovasc Pathol 2024; 73:107683. [PMID: 39111556 DOI: 10.1016/j.carpath.2024.107683] [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: 03/26/2024] [Revised: 07/25/2024] [Accepted: 07/31/2024] [Indexed: 08/29/2024] Open
Abstract
Over the years, advancements in the field of oncology have made remarkable strides in enhancing the efficacy of medical care for patients with cancer. These modernizations have resulted in prolonged survival and improved the quality of life for these patients. However, this progress has also been accompanied by escalation in mortality rates associated with anthracycline chemotherapy. Anthracyclines, which are known for their potent antitumor properties, are notorious for their substantial cardiotoxic potential. Remarkably, even after 6 decades of research, a conclusive solution to protect the cardiovascular system against doxorubicin-induced damage has not yet been established. A comprehensive understanding of the pathophysiological processes driving cardiotoxicity combined with targeted research is crucial for developing innovative cardioprotective strategies. This review seeks to explain the mechanisms responsible for structural and functional alterations in doxorubicin-induced cardiomyopathy.
Collapse
Affiliation(s)
- Ashot Avagimyan
- Department of Internal Desiases Propedeutics, Yerevan State Medical University after M. Heratsi, Yerevan, Armenia.
| | - Nana Pogosova
- Deputy Director of Research and Preventive Cardiology, National Medical Research Centre of Cardiology named after E. Chazov, Moscow, Russia; Head of Evidence Based Medicine Department, Patrice Lumumba Peoples' Friendship University of Russia (RUDN), Moscow, Russia
| | - Lev Kakturskiy
- A. P. Avtsyn Research Institute of Human Morphology, Petrovskiy RNCS, Moscow, Russia
| | - Mohammad Sheibani
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Teharan, Iran; Razi Drug Research Centre, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Abhiram Challa
- Department of Cardiology, West Virginia University, Morgantown, WV, USA
| | - Eugenia Kogan
- Institute of Clinical Morphology and Digital Pathology, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Federica Fogacci
- Atherosclerosis and Metabolic Disorders Research Unit, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Liudmila Mikhaleva
- A. P. Avtsyn Research Institute of Human Morphology, Petrovskiy RNCS, Moscow, Russia
| | - Rositsa Vandysheva
- A. P. Avtsyn Research Institute of Human Morphology, Petrovskiy RNCS, Moscow, Russia
| | - Marianna Yakubovskaya
- Chemical Cancerogenesis Department, Institute of Cancerogenesis, National Medical Research Center of Oncology after N. N. Blokhina, Moscow, Russia; Laboratory of Single Cell Biology, Patrice Lumumba Peoples' Friendship University of Russia (RUDN), Moscow, Russia
| | - Andrea Faggiano
- Department of Cardio-Thoracic-Vascular Area, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Stefano Carugo
- Department of Cardio-Thoracic-Vascular Area, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Olga Urazova
- Head of Pathophysiology Department, Siberian State Medical University, Tomsk, Russia
| | - Behnaz Jahanbin
- Department of Pathology, Cancer Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Ekaterina Lesovaya
- Chemical Cancerogenesis Department, Institute of Cancerogenesis, National Medical Research Center of Oncology after N. N. Blokhina, Moscow, Russia; Laboratory of Single Cell Biology, Patrice Lumumba Peoples' Friendship University of Russia (RUDN), Moscow, Russia; Department of Oncology, Ryazan State Medical University after I. P. Pavlov, Ryazan, Russia
| | | | - Kirill Kirsanov
- Chemical Cancerogenesis Department, Institute of Cancerogenesis, National Medical Research Center of Oncology after N. N. Blokhina, Moscow, Russia; Laboratory of Single Cell Biology, Patrice Lumumba Peoples' Friendship University of Russia (RUDN), Moscow, Russia
| | - Yasar Sattar
- Department of Cardiology, West Virginia University, Morgantown, WV, USA
| | - Artem Trofimenko
- Department of Pathophysiology, Kuban State Medical University, Krasnodar, Russia
| | - Tatiana Demura
- Institute of Clinical Morphology and Digital Pathology, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Amene Saghazadeh
- Department of Pathology, Cancer Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - George Koliakos
- Head of Laboratory of Biological Chemistry, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Davood Shafie
- Director of Heart Failure Centre, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Azin Alizadehasl
- Head of Cardio-Oncology Department and Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Arrigo Cicero
- Hypertension and Cardiovascular Risk Research Unit, Alma Mater Studiorum University of Bologna, Bologna, Italy; IRCCS Policlinico S. Orsola-Malpighi di Bologna, Bologna, Italy
| | - Juan Pablo Costabel
- Chief of Coronary Care Unit, Buenos Aires Institute of Cardiology, Buenos Aires, Argentina
| | - Giuseppe Biondi-Zoccai
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy; Maria Cecilia Hospital, GVM Care and Research, Cotignola, Italy
| | - Giulia Ottaviani
- Anatomic Pathology, Lino Rossi Research Center, Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, Milan, Italy
| | - Nizal Sarrafzadegan
- Director of Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran; School of Population and Public Health, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| |
Collapse
|
29
|
Lee GA, Reehal P. Cardiotoxicity in people undergoing cancer treatment - the role of the oncology nurse. Asia Pac J Oncol Nurs 2024; 11:100597. [PMID: 39582553 PMCID: PMC11582369 DOI: 10.1016/j.apjon.2024.100597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 09/20/2024] [Indexed: 11/26/2024] Open
Abstract
Cardiotoxicity is a significant complication of cancer treatment, and this review describes the cardiovascular risks associated with various therapies and emphasizes the crucial role of oncology nurses in managing these risks. Many cancer treatments, including cytotoxic therapies, radiation, targeted therapies, and immune-modulatory drugs, have been shown to increase the likelihood of cardiovascular toxicity, leading to conditions such as acute coronary syndrome, heart failure, and atrial fibrillation. Guidelines are now available to reduce treatment-related cardiovascular toxicity (CTR-CVT) and stress the importance of cardiovascular assessments before, during, and after cancer treatment. Oncology nurses are pivotal in performing these assessments and collaborating within multidisciplinary teams, including cardiologists, to ensure optimal care. As more patients successfully complete cancer treatment, it becomes essential to integrate cardiovascular risk evaluation, education, and medication into routine oncology care. The evolving field of oncology nursing, particularly with the rise of emerging therapies and aging populations, requires further education on early detection and management of cardiotoxicity to enhance patient outcomes.
Collapse
Affiliation(s)
- Geraldine A. Lee
- Professor of Nursing and Chair of Health Service Research, Catherine McAuley School of Nursing & Midwifery, Brookfield Health Sciences Complex, University College Cork, Cork, Ireland
| | - Priya Reehal
- Cardio-Oncology/Heart Failure Clinical Nurse Specialist, Cardio-Oncology Centre of Excellence, Royal Brompton Hospital, Part of Guy's and St Thomas' NHS Foundation, London, United Kingdom
| |
Collapse
|
30
|
Liu S, Horowitz JD, Koczwara B, Sverdlov AL, Packer N, Clark RA. Cardiac events among a cohort of 17,389 patients receiving cancer chemotherapy: short and long term implications. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:72. [PMID: 39415228 PMCID: PMC11481733 DOI: 10.1186/s40959-024-00269-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 09/25/2024] [Indexed: 10/18/2024]
Abstract
BACKGROUND The association between cardiovascular disease and carcinogenesis is bidirectional and well-established. Furthermore, cancer treatment improves overall patient survival, potentially at the cost of incremental and fatal cardiovascular disease (CVD). AIM To evaluate (a) In a real-world cohort, the proportion of patients offered cancer chemotherapy who have antecedent CVD (CVDA); (b) The rates of patient admission with subsequent development of CVD (CVDS) requiring hospital admission post assignment to chemotherapy; (c) The impact of CVDA and CVDS on mortality rates relative to those seen in patients without overt CVD (CVD-) and (d) The time course of mortality in CVD- versus CVDS patients. METHODS Retrospective analysis was performed in deidentified linked health data sets. Correlates of mortality were evaluated by Cox proportional hazards evaluation. Relative and absolute time-variability of CVD as a primary cause of death were determined. RESULTS Of the total 17,389 patients, there were 2,159 with CVDA. Over a median follow-up time of 4.6 years, CVDS admissions (n = 8,529) occurred more commonly in the presence of CVDA (70.0% vs. 46.1%, p < 0.001), and more than 50% of CVDS cases occurred in the first 12 months of follow-up. The 5-year mortality rates were 71.5% for CVDA, 64.7% for CVDS, and 40.8% for CVD- (p < 0.001). Development of CVDS was associated with a substantially increased risk of mortality in the next 12 months. The development of CVDs was also associated with an increased risk of cardiovascular, as against non-cardiovascular, mortality (7.1% vs. 1.6%, p < 0.001). CONCLUSIONS Approximately 50% of patients assigned to cancer chemotherapy developed CVDS, heralding a particularly high risk of mortality over the next 12 months. Both CVDA and CVDS are associated with substantial increases in mortality rates relative to those in CVD- patients. This increased risk merits close individual monitoring.
Collapse
Affiliation(s)
- Saifei Liu
- Cardiovascular Pathophysiology & Therapeutics Group, The Basil Hetzel Institute for Translational Research, University of Adelaide, Adelaide, SA, Australia.
- Caring Futures Institute, Flinders University, Southern Adelaide Local Health Network, Adelaide, SA, Australia.
| | - John D Horowitz
- Cardiovascular Pathophysiology & Therapeutics Group, The Basil Hetzel Institute for Translational Research, University of Adelaide, Adelaide, SA, Australia
| | - Bogda Koczwara
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
- Southern Adelaide Local Health Network, Adelaide, SA, Australia
| | - Aaron L Sverdlov
- Newcastle Centre of Excellence in Cardio-Oncology: Calvary Mater Newcastle, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
- Cardiovascular Department, John Hunter Hospital, Newcastle, NSW, Australia
| | - Natalie Packer
- Southern Adelaide Local Health Network, Adelaide, SA, Australia
| | - Robyn A Clark
- Caring Futures Institute, Flinders University, Southern Adelaide Local Health Network, Adelaide, SA, Australia
- Southern Adelaide Local Health Network, Adelaide, SA, Australia
| |
Collapse
|
31
|
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
|
32
|
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: 0] [Impact Index Per Article: 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
|
33
|
Li C, Yan W, Gao M, Zhang Z, Zhao L, Ma J, Li X, Gao Y, Zhang D, Gao S. Knowledge, attitudes, and practice of physicians and pharmacists regarding the prevention and treatment of cardiovascular toxicity associated with cancer treatment. Sci Rep 2024; 14:20122. [PMID: 39209910 PMCID: PMC11362540 DOI: 10.1038/s41598-024-71015-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
This study aimed to explore physicians' and pharmacists' knowledge, attitudes, and practice (KAP) regarding the prevention and treatment of cardiovascular toxicity associated with cancer treatment. A multicenter cross-sectional study included physicians and pharmacists between April 2023 and June 2023. The study included 918 participants (514 physicians and 404 pharmacists). The average scores of knowledge, attitudes, and practice were 11.6 ± 3.39, 24.7 ± 2.6, and 26.3 ± 6.8 points. Sufficient knowledge was significantly associated with age ≥ 41 years (odds ratio (OR) = 2.745, 95% confidence interval (CI) 1.086-6.941, P = 0.033), male (OR = 2.745, 95% CI 1.150-2.223, P = 0.005), bachelor's degree (OR = 0.084, 95% CI 0.013-0.533, P = 0.009), master's degree and above (OR = 0.096, 95% CI 0.015-0.609, P = 0.013), physician occupation (OR = 7.601, 95% CI 1.337-43.207, P = 0.022), pharmacy department (OR = 18.858, 95% CI 3.245-109.57, P = 0.001), oncology department (OR = 4.304, 95% CI 2.426-7.634, P < 0.001), cardiology department (OR = 3.001, 95% CI 1.387-6.492, P = 0.005), hospitals located in Eastern China (OR = 1.957, 95% CI 1.120-3.418, P = 0.018), and hospitals located in Western China (OR = 3.137, 95% CI 1.783-5.518, P < 0.001). Positive attitudes were significantly associated with a senior professional title (OR = 2.989, 95% CI 1.124-7.954, P = 0.028) and hospitals located in Eastern China (OR = 0.424, 95% CI 0.257-0.698, P = 0.001), Western China (OR = 0.231, 95% CI 0.136-0.394, P < 0.001), and Southern China (OR = 0.341, 95% CI 0.198-0.587, P < 0.001). Proactive practice was significantly associated with male (OR = 1.414, 95% CI 1.029-1.943, P = 0.033), senior professional title (OR = 3.838, 95% CI 1.176-12.524, P = 0.026), oncology department (OR = 3.827, 95% CI 2.336-6.272, P < 0.001), and cardiology department (OR = 2.428, 95% CI 1.263-4.669, P = 0.008). Both physicians and pharmacists had positive attitudes toward the prevention and treatment of cardiovascular toxicity associated with cancer treatment, while their knowledge and practice were not as proactive.
Collapse
Affiliation(s)
- Congxin Li
- Department of Pharmacy, Hebei Medical University Third Hospital, Shijiazhuang, 050051, China.
| | - Wei Yan
- Department of Pharmacy, Hebei Medical University Third Hospital, Shijiazhuang, 050051, China
| | - Meiling Gao
- Department of Pharmacy, Hebei Medical University Third Hospital, Shijiazhuang, 050051, China
| | - Zhihan Zhang
- Department of Nutrition, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050010, China
| | - Ling Zhao
- Department of Pharmacy, Hebei Medical University Third Hospital, Shijiazhuang, 050051, China
| | - Jingtao Ma
- Department of Cardiovascular, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050010, China
| | - Xuejing Li
- Department of Pharmacy, Hebei Medical University Third Hospital, Shijiazhuang, 050051, China
| | - Yuan Gao
- Department of Pharmacy, Hebei Medical University Third Hospital, Shijiazhuang, 050051, China
| | - Dongxia Zhang
- Department of Hepatology Infection, Bethune International Peace Hospital, Shijiazhuang, 050051, China
| | - Shan Gao
- Department of Pharmacy, Hebei Medical University Third Hospital, Shijiazhuang, 050051, China
| |
Collapse
|
34
|
Hadkar VM, Mohanty C, Selvaraj CI. Biopolymeric nanocarriers in cancer therapy: unleashing the potency of bioactive anticancer compounds for enhancing drug delivery. RSC Adv 2024; 14:25149-25173. [PMID: 39139249 PMCID: PMC11317881 DOI: 10.1039/d4ra03911d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 07/31/2024] [Indexed: 08/15/2024] Open
Abstract
Effective cancer treatment is becoming a global concern, and recent developments in nanomedicine are essential for its treatment. Cancer is a severe metabolic syndrome that affects the human population and is a significant contributing factor to deaths globally. In science, nanotechnology offers rapidly developing delivery methods for natural bioactive compounds that are becoming increasingly prominent and can be used to treat diseases in a site-specific way. Chemotherapy and radiotherapy are conventional approaches for preventing cancer progression and have adverse effects on the human body. Many chemically synthesized drugs are used as anticancer agents, but they have several side effects; hence, they are less preferred. Medicinal plants and marine microorganisms represent a vast, mostly untapped reservoir of bioactive compounds for cancer treatment. However, they have several limitations, including nonspecific targeting, weak water solubility and limited therapeutic potential. An alternative option is the use of biopolymeric nanocarriers, which can generate effective targeted treatment therapies when conjugated with natural anticancer compounds. The present review focuses on biopolymeric nanocarriers utilizing natural sources as anticancer drugs with improved tumor-targeting efficiency. This review also covers various natural anticancer compounds, the advantages and disadvantages of natural and synthetic anticancer compounds, the problems associated with natural anticancer drugs and the advantages of biopolymeric nanocarriers over synthetic nanocarriers as drug delivery agents. This review also discusses various biopolymeric nanocarriers for enhancing the controlled delivery of anticancer compounds and the future development of nanomedicines for treating cancer.
Collapse
Affiliation(s)
- Vrushali Manoj Hadkar
- School of Biosciences and Technology, Vellore Institute of Technology (VIT) Vellore 632014 Tamil Nadu India
| | - Chirasmita Mohanty
- School of Biosciences and Technology, Vellore Institute of Technology (VIT) Vellore 632014 Tamil Nadu India
| | - Chinnadurai Immanuel Selvaraj
- Department of Genetics and Plant Breeding, VIT School of Agricultural Sciences and Advanced Learning (VAIAL), VIT Vellore 632014 Tamil Nadu India
| |
Collapse
|
35
|
Martínez-Campelo L, Blanco-Verea A, López-Fernández T, Martínez-Monzonís A, Buño A, Mazón P, Zamora P, Norton N, Reddy JS, Velasco-Ruiz A, González-Neira A, Vulsteke C, Alonso-Gordoa T, Cruz R, Diz-de Almeida S, Carracedo A, González-Juanatey JR, López-Sendón J, Brion M. Meta-analysis of genome-wide association studies for cancer therapy-related cardiovascular dysfunction and functional mapping highlight an intergenic region close to TP63. Sci Rep 2024; 14:18413. [PMID: 39117733 PMCID: PMC11310459 DOI: 10.1038/s41598-024-69064-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 07/31/2024] [Indexed: 08/10/2024] Open
Abstract
Cancer therapy-related cardiac dysfunction (CTRCD), which commonly includes left ventricular dysfunction and heart failure, is the main adverse effect of anticancer therapy. In recent years several candidate genes studies and genome-wide association studies have identified common genetic variants associated with CTRCD, but evidence remains limited and few genetic variants are robust. A genome-wide meta-analysis of CTRCD was performed with 852 oncology patients receiving cancer therapy. DNA samples were genotyped and imputed to perform a GWAS meta-analysis for case-control (N = 852 (380 cases and 472 controls) and extreme phenotypes (N = 618 (78 cases and 472 controls) looking for genetic variants that predispose to CTRCD. The results were validated in a replicate cohort of 1,191 oncology patients (245 cases and 946 controls). Functional mapping of the replicated loci was then performed. The meta-analysis showed 9 and 17 loci suggestively associated (P-value < 1 × 10-5) with CTRCD in case-control and extreme phenotypes analyses, respectively. The 3q28 locus (rs rs7652759, P = 5.64 × 10-6) in the case-control analysis was the strongest signal, with up to 64 SNPs above the suggestive significance threshold. The rs7652759, an intergenic variant between TPRG1 and TP63 genes, was the only variant validated in the replication cohort (P-value = 0.01). Functional mapping of this significant locus revealed up to 5 new genes potentially involved in the CTRCD. We identified the intergenic region near TP63 as a novel CTRCD susceptibility locus. In the future, the genotyping of these markers could be considered in new CTRCD risk scores to improve preventive strategies in cardio-oncology.
Collapse
Affiliation(s)
- L Martínez-Campelo
- Instituto de Investigación Sanitaria de Santiago, Xenética Cardiovascular, Santiago De Compostela, Spain.
| | - A Blanco-Verea
- Instituto de Investigación Sanitaria de Santiago, Xenética Cardiovascular, Santiago De Compostela, Spain
| | - T López-Fernández
- Hospital Universitario La Paz, Servicio de Cardiología, Madrid, Spain
| | - A Martínez-Monzonís
- Hospital Clínico Universitario de Santiago de Compostela, Servicio de Cardiología, CIBER de Enfermedades Cardiovasculares, Santiago De Compostela, Spain
| | - A Buño
- Hospital Universitario La Paz, Servicio de Análisis Clínicos, IdiPaz, Madrid, Spain
| | - P Mazón
- Hospital Clínico Universitario de Santiago de Compostela, Servicio de Cardiología, CIBER de Enfermedades Cardiovasculares, Santiago De Compostela, Spain
| | - P Zamora
- Hospital Universitario La Paz, Servicio de Oncología, Madrid, Spain
| | - N Norton
- Department of Cancer Biology, Mayo Clinic Florida, Jacksonville, USA
| | - J S Reddy
- Department of Quantitative Health Sciences, Mayo Clinic Florida, Jacksonville, USA
| | - A Velasco-Ruiz
- Spanish National Cancer Research Centre, Human Cancer Genetics Programme, Madrid, Spain
| | - A González-Neira
- Spanish National Cancer Research Centre, Human Cancer Genetics Programme, Madrid, Spain
| | - C Vulsteke
- Department of Medical Oncology, Integrated Cancer Center Ghent, Ghent, Belgium
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Wilrijk, Belgium
| | - T Alonso-Gordoa
- Department of Medical Oncology, University Hospital Ramón y Cajal, Madrid, Spain
| | - R Cruz
- Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas, Universidade de Santiago de Compostela, CIBER de Enfermedades Raras, Madrid, Spain
| | - S Diz-de Almeida
- Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas, Universidade de Santiago de Compostela, CIBER de Enfermedades Raras, Madrid, Spain
| | - A Carracedo
- Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas, Universidade de Santiago de Compostela, CIBER de Enfermedades Raras, Madrid, Spain
- Fundación Pública Galega de Medicina Xenómica, Grupo de Medicina Xenómica, Santiago De Compostela, Spain
| | - J R González-Juanatey
- Hospital Clínico Universitario de Santiago de Compostela, Servicio de Cardiología, CIBER de Enfermedades Cardiovasculares, Santiago De Compostela, Spain
| | - J López-Sendón
- Instituto de Investigación Hospital Universitario La Paz (IdiPaz), Madrid, Spain
| | - M Brion
- Instituto de Investigación Sanitaria de Santiago, Xenética Cardiovascular, Santiago De Compostela, Spain
- Hospital Clínico Universitario de Santiago de Compostela, Servicio de Cardiología, CIBER de Enfermedades Cardiovasculares, Santiago De Compostela, Spain
| |
Collapse
|
36
|
Rosenfeld R, Riondino S, Cerocchi M, Luciano A, Idone G, Lecis D, Illuminato F, Tolomei A, Torino F, Chiocchi M, Roselli M. Extracellular volume measured by whole body CT scans predicts chronic cardiotoxicity in breast cancer patients treated with neoadjuvant therapies based on anthracyclines: A retrospective study. Breast 2024; 76:103755. [PMID: 38852211 PMCID: PMC11220522 DOI: 10.1016/j.breast.2024.103755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 05/19/2024] [Accepted: 05/24/2024] [Indexed: 06/11/2024] Open
Abstract
INTRODUCTION Neoadjuvant chemotherapies for breast cancer (BC) are effective but potentially cardiotoxic, and expose long survivors at risk of chemotherapy-related cardiac dysfunction (CTRCD). Unfortunately, early screening for CTRCD has actual diagnostic limits. Myocardial extracellular volume (mECV) is a radiological marker used in cardiac CT scans and cardiac magnetic resonance for diagnosis and follow-up of CTRCD. It can be measured in whole-body CT (WB-CT) scan, routinely performed in patients at high risk of relapse, to evaluate CTRCD occurrence during oncological follow-up. METHODS 82 WB-CT scans were examined at baseline (T0) and during oncological follow-up at first year (T1) and fifth year (T5) after the end of neoadjuvant treatment. mECV was measured at 1 min (PP) and 5 min (DP) after contrast injection. 31 echocardiograms were retrieved in T1 to perform a linear correlation between mECV and left ventricular ejection fraction (LVEF). RESULTS mECV values in T0 were similar between the two groups both in PP and in DP. Significant results were found for PP values in T1 (37.0 % vs 32 %, p = 0.0005) and in T5 (27.2 % vs 31.2 %, p = 0.025). A cut-off value of 35 % in PP proved significant in T1 (OR = 12.4, p = 0.004), while mECV was inversely correlated with LVEF both in PP (adj-S = -3.54, adj-p = 0.002) and in DP (adj-S = -2.51, adj-p = 0.0002), suggesting a synergistic action with the age at diagnosis (p < 0.0001, respectively). CONCLUSIONS WB-CT scans performed during oncological reassessment in patients at high-risk of recurrence could be used for CTRCD screening in cardiovascular low-risk patients, especially in aging patients with mECV values above 35 %.
Collapse
Affiliation(s)
- R Rosenfeld
- Medical Oncology Unit, Department of Systems Medicine, University of Rome "Tor Vergata", 00133, Rome, Italy.
| | - S Riondino
- Medical Oncology Unit, Department of Systems Medicine, University of Rome "Tor Vergata", 00133, Rome, Italy
| | - M Cerocchi
- Department of Biomedicine and Prevention, Division of Diagnostic Imaging, University of Rome "Tor Vergata", 00133, Rome, Italy
| | - A Luciano
- Department of Biomedicine and Prevention, Division of Diagnostic Imaging, University of Rome "Tor Vergata", 00133, Rome, Italy
| | - G Idone
- Unit of Cardiology, Department of Systems Medicine, University of Rome "Tor Vergata", 00133, Rome, Italy
| | - D Lecis
- Unit of Cardiology, Department of Systems Medicine, University of Rome "Tor Vergata", 00133, Rome, Italy
| | - F Illuminato
- Unit of Cardiology, Department of Systems Medicine, University of Rome "Tor Vergata", 00133, Rome, Italy
| | - A Tolomei
- Unit of Cardiology, Department of Systems Medicine, University of Rome "Tor Vergata", 00133, Rome, Italy
| | - F Torino
- Medical Oncology Unit, Department of Systems Medicine, University of Rome "Tor Vergata", 00133, Rome, Italy
| | - M Chiocchi
- Department of Biomedicine and Prevention, Division of Diagnostic Imaging, University of Rome "Tor Vergata", 00133, Rome, Italy
| | - M Roselli
- Medical Oncology Unit, Department of Systems Medicine, University of Rome "Tor Vergata", 00133, Rome, Italy
| |
Collapse
|
37
|
Voß F, Nienhaus F, Pietrucha S, Ruckhäberle E, Fehm T, Melz T, Cramer M, Haberkorn SM, Flögel U, Westenfeld R, Scheiber D, Jung C, Kelm M, Polzin A, Bönner F. Anthracycline therapy induces an early decline of cardiac contractility in low-risk patients with breast cancer. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:43. [PMID: 39014463 PMCID: PMC11251313 DOI: 10.1186/s40959-024-00244-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 07/01/2024] [Indexed: 07/18/2024]
Abstract
AIMS Cancer therapy-related cardiac dysfunction (CTRCD) is a dreaded complication of anthracycline therapy. CTRCD most frequently appears in patients with cardiovascular risk factors (CVR) or known cardiovascular disease. However, limited data exist on incidence and course of anthracycline-induced CTRCD in patients without preexisting risk factors. We therefore aimed to longitudinally investigate a cohort of young women on anthracycline treatment due to breast cancer without cardiovascular risk factors or known cardiovascular disease (NCT03940625). METHODS AND RESULTS We enrolled 59 women with primary breast cancer and scheduled anthracycline-based therapy, but without CVR or preexisting cardiovascular disease. We conducted a longitudinal assessment before, immediately and 12 months after cancer therapy with general laboratory, electrocardiograms, echocardiography and cardiovascular magnetic resonance (CMR), including myocardial relaxometry with T1, T2 and extracellular volume mapping. Every single patient experienced a drop in CMR-measured left ventricular ejection fraction (LVEF) of 6 ± 3% immediately after cancer therapy. According to the novel definition 32 patients (54.2%) developed CTRCD after 12 months defined by reduction in LVEF, global longitudinal strain (GLS) and/or biomarkers elevation, two of them were symptomatic. Global myocardial T2 relaxation times as well as myocardial mass increased coincidently with a decline in wall-thickening. While T2 values and myocardial mass normalized after 12 months, LVEF and GLS remained impaired. CONCLUSION In every single patient anthracyclines induce a decline of myocardial contractility, even among patients without pre-existing risk factors for CTRCD. Our data suggest to thoroughly evaluate whether this may lead to an increased risk of future cardiovascular events.
Collapse
Affiliation(s)
- Fabian Voß
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Fabian Nienhaus
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Saskia Pietrucha
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Eugen Ruckhäberle
- Division of Gynecology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Tanja Fehm
- Division of Gynecology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Tobias Melz
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Mareike Cramer
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Sebastian M Haberkorn
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Ulrich Flögel
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
- Department of Molecular Cardiology and Cardiovascular Research Institute Düsseldorf, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Ralf Westenfeld
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Daniel Scheiber
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Christian Jung
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Malte Kelm
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
- Medical Faculty, CARID, Cardiovascular Research Institute Düsseldorf, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany
| | - Amin Polzin
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Florian Bönner
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.
| |
Collapse
|
38
|
Díaz-Balboa E, Peña-Gil C, Rodríguez-Romero B, Cuesta-Vargas AI, Lado-Baleato O, Martínez-Monzonís A, Pedreira-Pérez M, Palacios-Ozores P, López-López R, González-Juanatey JR, González-Salvado V. Exercise-based cardio-oncology rehabilitation for cardiotoxicity prevention during breast cancer chemotherapy: The ONCORE randomized controlled trial. Prog Cardiovasc Dis 2024; 85:74-81. [PMID: 38395212 DOI: 10.1016/j.pcad.2024.02.002] [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] [Received: 02/16/2024] [Accepted: 02/16/2024] [Indexed: 02/25/2024]
Abstract
BACKGROUND Breast cancer (BC) treatment with anthracyclines and/or anti-human epidermal growth factor receptor-2 (HER2) antibodies is associated with an increased risk of cardiovascular disease complications, including cancer therapy-related cardiac dysfunction (CTRCD). While Cardio-Oncology Rehabilitation (CORe) programs including exercise have emerged to minimize these risks, its role in preventing CTRCD is unclear. OBJECTIVES We investigated the effectiveness of an exercise-based CORe program in preventing CTRCD [left ventricular ejection fraction (LVEF) drop ≥10% to a value <53% or a decrease >15% in global longitudinal strain (GLS)]. Secondary outcomes examined changes in cardiac biomarkers, physical performance including peak oxygen consumption, psychometric and lifestyle outcomes. Safety, adherence, and patient satisfaction were also assessed. METHODS This is a randomized controlled trial including 122 early-stage BC women receiving anthracyclines and/or anti-HER2 antibodies, randomized to CORe (n = 60) or usual care with exercise recommendation (n = 62). Comprehensive assessments were performed at baseline and after cardiotoxic treatment completion. The average duration of the intervention was 5.8 months. RESULTS No cases of CTRCD were identified during the study. LVEF decreased in both groups, but was significantly attenuated in the CORe group [-1.5% (-2.9, -0.1); p = 0.006], with no changes detected in GLS or cardiac biomarkers. The CORe intervention led to significant body mass index (BMI) reduction (p = 0.037), especially in obese patients [3.1 kg/m2 (1.3, 4.8)]. Physical performance and quality-of-life remained stable, while physical activity level increased in both groups. No adverse events were detected. CONCLUSIONS This study suggests that CORe programs are safe and may help attenuate LVEF decline in BC women receiving cardiotoxic therapy and reduce BMI in obese patients.
Collapse
Affiliation(s)
- Estíbaliz Díaz-Balboa
- University of A Coruña, Department of Physiotherapy, Medicine and Biomedical Sciences, Campus de Oza, A Coruña 15071, Spain; Cardiology Department, University Clinical Hospital of Santiago de Compostela (SERGAS); Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 15706 Santiago de Compostela, A Coruña, Spain; Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, A Coruña, Spain.
| | - Carlos Peña-Gil
- Cardiology Department, University Clinical Hospital of Santiago de Compostela (SERGAS); Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 15706 Santiago de Compostela, A Coruña, Spain; Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, A Coruña, Spain.
| | - Beatriz Rodríguez-Romero
- University of A Coruña. Psychosocial Intervention and Functional Rehabilitation Research Group, Department of Physiotherapy, Medicine and Biomedical Sciences, Campus de Oza, A Coruña, Spain 15071.
| | - Antonio I Cuesta-Vargas
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Málaga 29010, Spain; Department of Physiotherapy, University of Málaga, Málaga 29071, Spain.
| | - Oscar Lado-Baleato
- Unit of Biostatistics, Department of Statistics, Mathematical Analysis, and Optimization, Universidade de Santiago de Compostela, Spain.
| | - Amparo Martínez-Monzonís
- Cardiology Department, University Clinical Hospital of Santiago de Compostela (SERGAS); Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 15706 Santiago de Compostela, A Coruña, Spain; Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, A Coruña, Spain
| | - Milagros Pedreira-Pérez
- Cardiology Department, University Clinical Hospital of Santiago de Compostela (SERGAS); Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 15706 Santiago de Compostela, A Coruña, Spain; Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, A Coruña, Spain.
| | - Patricia Palacios-Ozores
- Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, A Coruña, Spain; Medical Oncology Department and Translational Medical Oncology Group, University Clinical Hospital of Santiago (SERGAS); Centro de Investigación Biomédica en Red de Cáncer (CIBERONC); Santiago de Compostela University School of Medicine, 15706 Santiago de Compostela, A Coruña, Spain..
| | - Rafael López-López
- Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, A Coruña, Spain; Medical Oncology Department and Translational Medical Oncology Group, University Clinical Hospital of Santiago (SERGAS); Centro de Investigación Biomédica en Red de Cáncer (CIBERONC); Santiago de Compostela University School of Medicine, 15706 Santiago de Compostela, A Coruña, Spain..
| | - José R González-Juanatey
- Cardiology Department, University Clinical Hospital of Santiago de Compostela (SERGAS); Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 15706 Santiago de Compostela, A Coruña, Spain; Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, A Coruña, Spain.
| | - Violeta González-Salvado
- Cardiology Department, University Clinical Hospital of Santiago de Compostela (SERGAS); Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 15706 Santiago de Compostela, A Coruña, Spain; Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, A Coruña, Spain.
| |
Collapse
|
39
|
Scalia IG, Gheyath B, Tamarappoo BK, Moudgil R, Otton J, Pereyra M, Narayanasamy H, Larsen C, Herrmann J, Arsanjani R, Ayoub C. Chemotherapy Related Cardiotoxicity Evaluation-A Contemporary Review with a Focus on Cardiac Imaging. J Clin Med 2024; 13:3714. [PMID: 38999280 PMCID: PMC11242267 DOI: 10.3390/jcm13133714] [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: 05/28/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 07/14/2024] Open
Abstract
The long-term survivorship of patients diagnosed with cancer has improved due to accelerated detection and rapidly evolving cancer treatment strategies. As such, the evaluation and management of cancer therapy related complications has become increasingly important, including cardiovascular complications. These have been captured under the umbrella term "cardiotoxicity" and include left ventricular dysfunction and heart failure, acute coronary syndromes, valvular abnormalities, pericardial disease, arrhythmia, myocarditis, and vascular complications. These complications add to the burden of cardiovascular disease (CVD) or are risk factors patients with cancer treatment are presenting with. Of note, both pre- and newly developing CVD is of prognostic significance, not only from a cardiovascular perspective but also overall, potentially impacting the level of cancer therapy that is possible. Currently, there are varying recommendations and practices regarding CVD risk assessment and mitigating strategies throughout the cancer continuum. This article provides an overview on this topic, in particular, the role of cardiac imaging in the care of the patient with cancer. Furthermore, it summarizes the current evidence on the spectrum, prevention, and management of chemotherapy-related adverse cardiac effects.
Collapse
Affiliation(s)
- Isabel G. Scalia
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.)
| | - Bashaer Gheyath
- Department of Imaging, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Balaji K. Tamarappoo
- Division of Cardiology, Banner University Medical Center, The University of Arizona College of Medicine, Phoenix, AZ 85004, USA
| | - Rohit Moudgil
- Department of Cardiology, Heart and Vascular Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - James Otton
- Clinical School, St. Vincent’s Hospital, UNSW, Sydney, NSW 2010, Australia
| | - Milagros Pereyra
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.)
| | - Hema Narayanasamy
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.)
| | - Carolyn Larsen
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.)
| | - Joerg Herrmann
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Reza Arsanjani
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.)
| | - Chadi Ayoub
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.)
| |
Collapse
|
40
|
Wernhart S, Rassaf T. Relevance of Cardiovascular Exercise in Cancer and Cancer Therapy-Related Cardiac Dysfunction. Curr Heart Fail Rep 2024; 21:238-251. [PMID: 38696059 PMCID: PMC11090948 DOI: 10.1007/s11897-024-00662-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/03/2024] [Indexed: 05/14/2024]
Abstract
PURPOSE OF THE REVIEW Cancer therapy-related cardiac dysfunction (CTRCD) has been identified as a threat to overall and cancer-related survival. Although aerobic exercise training (AET) has been shown to improve cardiorespiratory fitness (CRF), the relationship between specific exercise regimens and cancer survival, heart failure development, and reduction of CTRCD is unclear. In this review, we discuss the impact of AET on molecular pathways and the current literature of sports in the field of cardio-oncology. RECENT FINDINGS Cardio-oncological exercise trials have focused on variations of AET intensity by using moderate continuous and high intensity interval training, which are applicable, safe, and effective approaches to improve CRF. AET increases CRF, reduces cardiovascular morbidity and heart failure hospitalization and should thus be implemented as an adjunct to standard cancer therapy, although its long-term effect on CTRCD remains unknown. Despite modulating diverse molecular pathways, it remains unknown which exercise regimen, including variations of AET duration and frequency, is most suited to facilitate peripheral and central adaptations to exercise and improve survival in cancer patients.
Collapse
Affiliation(s)
- Simon Wernhart
- Department of Cardiology and Vascular Medicine, West German Heart- and Vascular Center, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany.
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart- and Vascular Center, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany
| |
Collapse
|
41
|
Suntheralingam S, Osataphan N, Power C, Steve Fan CP, Abdel-Qadir H, Amir E, Thavendiranathan P. Safety of Continuing Trastuzumab for Mild Cardiotoxicity: A Cardiovascular Magnetic Resonance Imaging Study. CJC Open 2024; 6:830-835. [PMID: 39022173 PMCID: PMC11250870 DOI: 10.1016/j.cjco.2024.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 03/16/2024] [Indexed: 07/20/2024] Open
Abstract
The safety of continuing human epidermal growth factor receptor 2 (HER2)-targeted therapy in women with mild cardiotoxicity remains unclear. We performed a retrospective matched cohort study of 14 patients with human epidermal growth factor receptor 2-positive breast cancer receiving sequential anthracycline and trastuzumab therapy, nested within the Evaluation of Myocardial Changes During Breast Adenocarcinoma Therapy to Detect Cardiotoxicity Earlier With MRI (EMBRACE-MRI) trial. Among patients who developed cardiotoxicity and were treated with heart failure therapy, we compared those who had trastuzumab therapy interrupted to a matched cohort who continued trastuzumab therapy. By a median of 2.5 years of follow-up, no significant differences were present between the groups in the proportion with magnetic resonance imaging-measured left ventricular ejection fraction < 40%, magnetic resonance imaging-measured left ventricular volumes, left ventricular ejection fraction, edema, fibrotic markers, cardiopulmonary fitness, or quality of life.
Collapse
Affiliation(s)
- Sivisan Suntheralingam
- Division of Cardiology, Peter Munk Cardiac Centre, Ted Rogers Program in Cardiotoxicity Prevention, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Nichanan Osataphan
- Division of Cardiology, Peter Munk Cardiac Centre, Ted Rogers Program in Cardiotoxicity Prevention, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
- Division of Cardiology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Coleen Power
- Division of Cardiology, Peter Munk Cardiac Centre, Ted Rogers Program in Cardiotoxicity Prevention, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Chun-Po Steve Fan
- Rogers Computational Program, Ted Rogers Centre for Heart Research, Peter Munk Cardiac Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Husam Abdel-Qadir
- Division of Cardiology, Peter Munk Cardiac Centre, Ted Rogers Program in Cardiotoxicity Prevention, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, Women’s College Hospital Toronto, Toronto, Ontario, Canada
| | - Eitan Amir
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Paaladinesh Thavendiranathan
- Division of Cardiology, Peter Munk Cardiac Centre, Ted Rogers Program in Cardiotoxicity Prevention, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
- Joint Department of Medical Imaging, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
42
|
Obeidat O, Obeidat A, Abughazaleh S, Obeidat A, Daise M, Ismail MF, Alqudah Q, Al-Ani H, Tarawneh M, Pondicherry-Harish R. Unveiling Hidden Battles: Exploring the Link Between Breast Cancer Survival and Heart Failure Vulnerability. Angiology 2024:33197241255168. [PMID: 38756005 DOI: 10.1177/00033197241255168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
This study explores the link between a history of breast cancer and the vulnerability to heart failure. Analyzing data from the National Inpatient Sample (NIS) for women diagnosed with breast cancer between 2016 and 2019 in the US, our research utilized logistic regression, adjusting for demographics, comorbidities, and lifestyle factors, and employed propensity score matching. With 2,276,639 weighted cases, our findings reveal a slight but significant elevation in heart failure risk among the breast cancer cohort, specifically in acute, chronic, and isolated systolic heart failure types. Racial differences were pronounced; Black women with breast cancer showed higher risks for all heart failure types, particularly chronic and systolic, while Asian or Pacific Islander patients had a lower incidence of certain heart failure types. This research underscores a modest increase in heart failure risk post-breast cancer, highlighting the critical need for integrated cardio-oncology care and personalized healthcare approaches to address and mitigate this risk effectively.
Collapse
Affiliation(s)
- Omar Obeidat
- University of Central Florida College of Medicine, Graduate Medical Education, Orlando, FL, USA
- HCA Florida North Florida Hospital, Internal Medicine Residency Program, Gainesville, FL, USA
| | - Abedallah Obeidat
- Division of Cardiology, Department of Internal Medicine, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Saeed Abughazaleh
- St Elizabeth's Medical Center, Tufts University School of Medicine, Brighton, MA, USA
| | - Ali Obeidat
- Division of Cardiology, Department of Internal Medicine, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Moh'd Daise
- University of Central Florida College of Medicine, Graduate Medical Education, Orlando, FL, USA
- HCA Florida North Florida Hospital, Internal Medicine Residency Program, Gainesville, FL, USA
| | - Mohamed F Ismail
- University of Central Florida College of Medicine, Graduate Medical Education, Orlando, FL, USA
- HCA Florida North Florida Hospital, Internal Medicine Residency Program, Gainesville, FL, USA
| | - Qusai Alqudah
- University of Central Florida College of Medicine, Graduate Medical Education, Orlando, FL, USA
- HCA Florida North Florida Hospital, Internal Medicine Residency Program, Gainesville, FL, USA
| | - Hashim Al-Ani
- University of Central Florida College of Medicine, Graduate Medical Education, Orlando, FL, USA
- HCA Florida North Florida Hospital, Internal Medicine Residency Program, Gainesville, FL, USA
| | - Mohammad Tarawneh
- St Elizabeth's Medical Center, Tufts University School of Medicine, Brighton, MA, USA
| | - Roja Pondicherry-Harish
- University of Central Florida College of Medicine, Graduate Medical Education/HCA Florida North Florida Hospital, Internal Medicine Residency Program, Gainesville, FL, USA
- The Cardiac and Vascular Institute, Gainesville, FL, USA
| |
Collapse
|
43
|
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
|
44
|
Badheeb AM, Alhosni Y, Alshahrani M, Asery TA, Al Nasher SM, Seada IA, Nasher AM, Alotaibi BA, Alsaaed E, Alsalem AM, Abu Bakar A, Alselwi W, Ahmed F, Alyami NH, Bin Dahman L, Badheeb M, Obied HY. Cardiotoxicity in Cancer Patients: The Prevalence, Risk Factors, and Cardioprotective Measures in a Cancer Centre in Saudi Arabia. Cureus 2024; 16:e59608. [PMID: 38832203 PMCID: PMC11144837 DOI: 10.7759/cureus.59608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2024] [Indexed: 06/05/2024] Open
Abstract
BACKGROUND Chemotherapy-related cardiotoxicity can exhibit several patterns of functional, structural, and vascular complications. This study aims to identify the patterns and the factors associated with cardiotoxicity in cancer patients. METHOD A retrospective cross-sectional analysis of 96 adult cancer patients undergoing anticancer therapy was investigated at King Khalid Hospital in Najran, Saudi Arabia, from May 2022 to April 2023. The data on patient and cancer characteristics, treatment, and outcomes were collected and analyzed. Factors associated with cardiotoxicity were investigated through univariate analyses using odds ratio (OR) and 95% confidence interval (CI). RESULTS Among the 96 cancer patients in the study, cardiotoxicity occurred in 12 individuals (12.5%). The mean age was 57.0 ± 13.3 years (range: 32-81 years), with 32 (33.3%) being above 65 years. The most common comorbidities were diabetes (n=48; 50%), followed by hypertension (n=32; 33.3%), and dyslipidemia (n=20; 20.8%). The most common cancers were gastrointestinal cancer (n=32; 33.3%), followed by breast cancer (n=22; 22.9%) and lymphoma (n=14; 14.6%). Females were disproportionately affected (64.6%), with 57.3% of them in the metastatic stage. The majority of patients (90.6%) had normal ejection fraction before chemotherapy initiation. In univariate analysis, current smoking (OR: 7.00; 95%CI: 1.94-25.25, p= 0.003), history of percutaneous cardiac intervention (OR: 40.24; 95%CI: 1.80-896.26, p= 0.019), diabetes (OR: 6.05; 95%CI: 1.24-29.32, p= 0.025), renal failure (OR: 8.20; 95%CI: 0.91-74.88, p= 0.046), dyslipidemia (OR: 5.00; 95 CI: 1.38-18.32, p=0.012), anthracycline use (OR: 18.33; 95%CI: 4.36-126.55, p <0.001), trastuzumab use (OR: 25.00; 95%CI: 6.25-129.86, p < 0.001), and increased chemotherapy cycles number (> 10 cycles) (OR: 73.00; 95%CI: 8.56- 622.36, p < 0.001) were associated with cardiotoxicity. Additionally, beta-blocker use was associated with lower rates of cardiotoxicity (OR: 0.17; 95%CI: 0.036-0.84, p= 0.029). CONCLUSIONS The incidence of cardiotoxicity among cancer patients treated with chemotherapy is modest, difficult to predict, and independent of baseline cardiac systolic functions. Factors associated with cardiotoxicity include smoking, history of percutaneous cardiac intervention, diabetes, renal failure, dyslipidemia, anthracycline or trastuzumab use, and increased chemotherapy cycle numbers. A combination of various anticancer drugs and chemotherapy may dramatically raise the risk of cardiotoxicity in cancer patients. As a result, patients receiving high-risk cardiotoxic drugs should be monitored with caution to avoid drug-related cardiotoxicity. Furthermore, proactive treatment techniques aiming at reducing the possible cardiotoxic effects of anticancer therapy are critical.
Collapse
Affiliation(s)
- Ahmed M Badheeb
- Medicine, Hadhramout University, Mukalla, YEM
- Oncology, King Khalid Hospital, Najran, SAU
| | | | | | - Tariq A Asery
- Internal Medicine, King Khalid Hospital, Riyadh, SAU
| | | | - Islam A Seada
- Cardiothoracic Surgery, King Khalid Hospital, Najran, SAU
| | | | | | - Esraa Alsaaed
- Internal Medicine, King Khalid Hospital, Najran, SAU
| | | | | | - Waleed Alselwi
- Medical Oncology, King Fahad Specialist Hospital, Dammam, SAU
| | | | - Nasher H Alyami
- Laboratory Medicine, Hematology Unit, Najran General Hospital, Ministry of Health, Najran, SAU
| | | | - Mohamed Badheeb
- Internal Medicine, Yale New Haven Health, Bridgeport Hospital, Bridgeport, USA
| | - Hamoud Y Obied
- Surgery, Faculty of Medicine, Najran University, Najran, SAU
- Cardiac Surgery, King Khalid Hospital, Najran, SAU
| |
Collapse
|
45
|
Fernández-Casas A, Leirós-Rodríguez R, Hernandez-Lucas P, González-Represas A. Protective effects of exercise on cardiotoxicity induced by breast cancer treatments: A systematic review and meta-analysis. Maturitas 2024; 183:107932. [PMID: 38325133 DOI: 10.1016/j.maturitas.2024.107932] [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: 10/08/2023] [Revised: 01/18/2024] [Accepted: 01/25/2024] [Indexed: 02/09/2024]
Abstract
OBJECTIVE Currently, one of the main causes of death in women with breast cancer is cardiovascular disease caused by the oncologic therapies. Exercise has demonstrated positive effects on cardiovascular fitness in individuals without cancer. Therefore, the aim of this study was to evaluate the cardioprotective effects of exercise in women with breast cancer, during and after the application of their treatments. METHODS Systematic search was done in PubMed, Scopus, Web of Science, CINAHL, MEDLINE, SPORTDiscus, and PEDro. The articles must have been published in the last ten years; the intervention to be evaluated was to consist of an exercise program; the sample had to comprise women who were undergoing breast cancer treatment or who had completed it at the time of the intervention; and the outcome variables had to include at least one parameter for the assessment of cardiac function and/or structure. RESULTS Of the 28 articles identified, nine reported non-randomized controlled studies, 16 randomized clinical trials and three quasi-experimental studies. The effects of exercise on left ventricular ejection fraction, global longitudinal strain and the E/A waveforms ratio were not significant. However, its effect on VO2max was significant. CONCLUSIONS Exercise does not seem to be effective in avoiding the cardiotoxic effects of oncological treatment for breast cancer. Although exercise seems to mitigate the symptomatology, reflected in improved functional capacity, more long-term studies are needed. PROSPERO REGISTRATION CODE CRD42023391441.
Collapse
Affiliation(s)
- Alicia Fernández-Casas
- Functional Biology and Health Sciences Department, University of Vigo, Pontevedra, Spain
| | - Raquel Leirós-Rodríguez
- SALBIS Research Group, Nursing and Physical Therapy Department, University of Leon, Ponferrada, Spain.
| | - Pablo Hernandez-Lucas
- Functional Biology and Health Sciences Department, University of Vigo, Pontevedra, Spain.
| | | |
Collapse
|
46
|
Díaz-Guerra A, Villena-Gutiérrez R, Clemente-Moragón A, Gómez M, Oliver E, Fernández-Tocino M, Galán-Arriola C, Cádiz L, Ibáñez B. Anthracycline Cardiotoxicity Induces Progressive Changes in Myocardial Metabolism and Mitochondrial Quality Control: Novel Therapeutic Target. JACC CardioOncol 2024; 6:217-232. [PMID: 38774018 PMCID: PMC11103041 DOI: 10.1016/j.jaccao.2024.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 01/31/2024] [Accepted: 02/06/2024] [Indexed: 05/24/2024] Open
Abstract
Background Anthracycline-induced cardiotoxicity (AIC) debilitates quality of life in cancer survivors. Serial characterizations are lacking of the molecular processes occurring with AIC. Objectives The aim of this study was to characterize AIC progression in a mouse model from early (subclinical) to advanced heart failure stages, with an emphasis on cardiac metabolism and mitochondrial structure and function. Methods CD1 mice received 5 weekly intraperitoneal doxorubicin injections (5 mg/kg) and were followed by serial echocardiography for 15 weeks. At 1, 9, and 15 weeks after the doxorubicin injections, mice underwent fluorodeoxyglucose positron emission tomography, and hearts were extracted for microscopy and molecular analysis. Results Cardiac atrophy was evident at 1 week post-doxorubicin (left ventricular [LV] mass 117 ± 26 mg vs 97 ± 25 mg at baseline and 1 week, respectively; P < 0.001). Cardiac mass nadir was observed at week 3 post-doxorubicin (79 ± 16 mg; P = 0.002 vs baseline), remaining unchanged thereafter. Histology confirmed significantly reduced cardiomyocyte area (167 ± 19 μm2 in doxorubicin-treated mice vs 211 ± 26 μm2 in controls; P = 0.004). LV ejection fraction declined from week 6 post-doxorubicin (49% ± 9% vs 61% ± 9% at baseline; P < 0.001) until the end of follow-up at 15 weeks (43% ± 8%; P < 0.001 vs baseline). At 1 week post-doxorubicin, when LV ejection fraction remained normal, reduced cardiac metabolism was evident from down-regulated markers of fatty acid oxidation and glycolysis. Metabolic impairment continued to the end of follow-up in parallel with reduced mitochondrial adenosine triphosphate production. A transient early up-regulation of nutrient-sensing and mitophagy markers were observed, which was associated with mitochondrial enlargement. Later stages, when mitophagy was exhausted, were characterized by overt mitochondrial fragmentation. Conclusions Cardiac atrophy, global hypometabolism, early transient-enhanced mitophagy, biogenesis, and nutrient sensing constitute candidate targets for AIC prevention.
Collapse
Affiliation(s)
- Anabel Díaz-Guerra
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Madrid, Spain
| | | | - Agustín Clemente-Moragón
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Madrid, Spain
| | - Mónica Gómez
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
- Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Eduardo Oliver
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Madrid, Spain
- Centro de Investigaciones Biológicas Margarita Salas, Madrid, Spain
| | - Miguel Fernández-Tocino
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Madrid, Spain
| | - Carlos Galán-Arriola
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Madrid, Spain
| | - Laura Cádiz
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Borja Ibáñez
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Madrid, Spain
- Cardiology Department, IIS-Fundación Jiménez Díaz Hospital, Madrid, Spain
| |
Collapse
|
47
|
Boen HM, Cherubin M, Franssen C, Gevaert AB, Witvrouwen I, Bosman M, Guns PJ, Heidbuchel H, Loeys B, Alaerts M, Van Craenenbroeck EM. Circulating MicroRNA as Biomarkers of Anthracycline-Induced Cardiotoxicity: JACC: CardioOncology State-of-the-Art Review. JACC CardioOncol 2024; 6:183-199. [PMID: 38774014 PMCID: PMC11103047 DOI: 10.1016/j.jaccao.2023.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 12/11/2023] [Accepted: 12/19/2023] [Indexed: 05/24/2024] Open
Abstract
Close monitoring for cardiotoxicity during anthracycline chemotherapy is crucial for early diagnosis and therapy guidance. Currently, monitoring relies on cardiac imaging and serial measurement of cardiac biomarkers like cardiac troponin and natriuretic peptides. However, these conventional biomarkers are nonspecific indicators of cardiac damage. Exploring new, more specific biomarkers with a clear link to the underlying pathomechanism of cardiotoxicity holds promise for increased specificity and sensitivity in detecting early anthracycline-induced cardiotoxicity. miRNAs (microRNAs), small single-stranded, noncoding RNA sequences involved in epigenetic regulation, influence various physiological and pathological processes by targeting expression and translation. Emerging as new biomarker candidates, circulating miRNAs exhibit resistance to degradation and offer a direct pathomechanistic link. This review comprehensively outlines their potential as early biomarkers for cardiotoxicity and their pathomechanistic link.
Collapse
Affiliation(s)
- Hanne M. Boen
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
| | - Martina Cherubin
- Centrum of Medical Genetics, GENCOR, University of Antwerp, Antwerp, Belgium
| | - Constantijn Franssen
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
| | - Andreas B. Gevaert
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
| | - Isabel Witvrouwen
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
| | - Matthias Bosman
- Laboratory of Physiopharmacology, GENCOR, University of Antwerp, Antwerp, Belgium
| | - Pieter-Jan Guns
- Laboratory of Physiopharmacology, GENCOR, University of Antwerp, Antwerp, Belgium
| | - Hein Heidbuchel
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
| | - Bart Loeys
- Centrum of Medical Genetics, GENCOR, University of Antwerp, Antwerp, Belgium
| | - Maaike Alaerts
- Centrum of Medical Genetics, GENCOR, University of Antwerp, Antwerp, Belgium
| | - Emeline M. Van Craenenbroeck
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
| |
Collapse
|
48
|
Alanazi R, Aljizeeri A, Alsaileek A, Alrashid A, Alolayan A, Alkaiyat M, Alenazy B, Shehata H, Alqahtani J, Ardah H, Alshammari K. Cardiac Morbidity and Mortality in Patients with Sarcoma: A Population-Based Study. Clin Med Insights Oncol 2024; 18:11795549241237703. [PMID: 38558879 PMCID: PMC10979535 DOI: 10.1177/11795549241237703] [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: 08/23/2023] [Accepted: 02/19/2024] [Indexed: 04/04/2024] Open
Abstract
Background Soft tissue sarcoma (STS) is a rare malignancy that affects soft tissues. It encompasses various subtypes and requires different treatment strategies. Doxorubicin is a commonly used anthracycline in the management of localized and metastatic STS. However, high doses of doxorubicin are associated with cardiotoxicity, which can significantly impact patients' long-term outcomes. This study aimed to evaluate doxorubicin's effect on cardiac function in patients with sarcoma and to correlate the frequency of cardiotoxicity with potential risk factors. Methods A retrospective analysis was conducted on patients with sarcoma who were treated with doxorubicin between 2016 and 2022 at King Abdulaziz Medical City in Saudi Arabia. Patient demographic information, comorbidities, cardiac measurements, laboratory values, systemic therapy, and treatment outcomes were collected from electronic medical records. A statistical analysis was performed to assess the association between cardiotoxicity and various factors. Results A total of 133 patients were included in the study, with a median age of 30 years. Cardiotoxicity was observed in 9% of the patients. Female patients had a significantly higher risk of developing cardiotoxicity. Patients with a higher Eastern Cooperative Oncology Group (ECOG) performance status and lower troponin I levels also had an increased risk of cardiotoxicity. However, there was no significant association between cardiotoxicity and the number of chemotherapy cycles, total cumulative dose of doxorubicin, or history of radiation. Furthermore, patients with cardiotoxicity had a higher risk of mortality. The overall survival of the patients was 18 months. Conclusion Doxorubicin-associated cardiotoxicity is a concern for patients with sarcoma. Female patients and patients with a higher ECOG performance status are at an increased risk of developing cardiotoxicity. Careful monitoring and risk assessment are crucial for mitigating the adverse effects of doxorubicin treatment in patients with sarcoma. Future studies are warranted to validate these findings and explore preventive strategies for doxorubicin-induced cardiotoxicity in patients with sarcoma.
Collapse
Affiliation(s)
- Rakan Alanazi
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of the National Guard-Health Affairs, Riyadh, Kingdom of Saudi Arabia
- King Abdullah International Medical Research Center, Ministry of the National Guard-Health Affairs, Riyadh, Kingdom of Saudi Arabia
| | - Ahmed Aljizeeri
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of the National Guard-Health Affairs, Riyadh, Kingdom of Saudi Arabia
- King Abdullah International Medical Research Center, Ministry of the National Guard-Health Affairs, Riyadh, Kingdom of Saudi Arabia
- King Abdulaziz Cardiac Center, Ministry of the National Guard Health Affairs, Riyadh, Kingdom of Saudi Arabia
| | - Ahmed Alsaileek
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of the National Guard-Health Affairs, Riyadh, Kingdom of Saudi Arabia
- King Abdullah International Medical Research Center, Ministry of the National Guard-Health Affairs, Riyadh, Kingdom of Saudi Arabia
- King Abdulaziz Cardiac Center, Ministry of the National Guard Health Affairs, Riyadh, Kingdom of Saudi Arabia
| | - Azzam Alrashid
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of the National Guard-Health Affairs, Riyadh, Kingdom of Saudi Arabia
- King Abdullah International Medical Research Center, Ministry of the National Guard-Health Affairs, Riyadh, Kingdom of Saudi Arabia
| | - Ashwaq Alolayan
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of the National Guard-Health Affairs, Riyadh, Kingdom of Saudi Arabia
- King Abdullah International Medical Research Center, Ministry of the National Guard-Health Affairs, Riyadh, Kingdom of Saudi Arabia
- Department of Medical Oncology, King Abdulaziz Medical City, Ministry of the National Guard-Health Affairs, Riyadh, Kingdom of Saudi Arabia
| | - Mohammad Alkaiyat
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of the National Guard-Health Affairs, Riyadh, Kingdom of Saudi Arabia
- King Abdullah International Medical Research Center, Ministry of the National Guard-Health Affairs, Riyadh, Kingdom of Saudi Arabia
- Department of Medical Oncology, King Abdulaziz Medical City, Ministry of the National Guard-Health Affairs, Riyadh, Kingdom of Saudi Arabia
| | - Basel Alenazy
- King Abdullah International Medical Research Center, Ministry of the National Guard-Health Affairs, Riyadh, Kingdom of Saudi Arabia
- King Abdulaziz Cardiac Center, Ministry of the National Guard Health Affairs, Riyadh, Kingdom of Saudi Arabia
| | - Hussam Shehata
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of the National Guard-Health Affairs, Riyadh, Kingdom of Saudi Arabia
- King Abdullah International Medical Research Center, Ministry of the National Guard-Health Affairs, Riyadh, Kingdom of Saudi Arabia
- Department of Medical Oncology, King Abdulaziz Medical City, Ministry of the National Guard-Health Affairs, Riyadh, Kingdom of Saudi Arabia
| | - Joud Alqahtani
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of the National Guard-Health Affairs, Riyadh, Kingdom of Saudi Arabia
- King Abdullah International Medical Research Center, Ministry of the National Guard-Health Affairs, Riyadh, Kingdom of Saudi Arabia
- Department of Medical Oncology, King Abdulaziz Medical City, Ministry of the National Guard-Health Affairs, Riyadh, Kingdom of Saudi Arabia
| | - Husam Ardah
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of the National Guard-Health Affairs, Riyadh, Kingdom of Saudi Arabia
- Department of Biostatistics and Bioinformatics, King Abdullah International Medical Research Center, Riyadh, Kingdom of Saudi Arabia
| | - Kanan Alshammari
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of the National Guard-Health Affairs, Riyadh, Kingdom of Saudi Arabia
- King Abdullah International Medical Research Center, Ministry of the National Guard-Health Affairs, Riyadh, Kingdom of Saudi Arabia
- Department of Medical Oncology, King Abdulaziz Medical City, Ministry of the National Guard-Health Affairs, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
49
|
Jacobs JEJ, Greason G, Mangold KE, Wildiers H, Willems R, Janssens S, Noseworthy P, Lopez-Jimenez F, Voigt JU, Friedman P, Van Aelst L, Vandenberk B, Attia ZI, Herrmann J. Artificial intelligence electrocardiogram as a novel screening tool to detect a newly abnormal left ventricular ejection fraction after anthracycline-based cancer therapy. Eur J Prev Cardiol 2024; 31:560-566. [PMID: 37943680 PMCID: PMC10972628 DOI: 10.1093/eurjpc/zwad348] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/15/2023] [Accepted: 11/02/2023] [Indexed: 11/12/2023]
Abstract
AIMS Cardiotoxicity is a serious side effect of anthracycline treatment, most commonly manifesting as a reduction in left ventricular ejection fraction (EF). Early recognition and treatment have been advocated, but robust, convenient, and cost-effective alternatives to cardiac imaging are missing. Recent developments in artificial intelligence (AI) techniques applied to electrocardiograms (ECGs) may fill this gap, but no study so far has demonstrated its merit for the detection of an abnormal EF after anthracycline therapy. METHODS AND RESULTS Single centre consecutive cohort study of all breast cancer patients with ECG and transthoracic echocardiography (TTE) evaluation before and after (neo)adjuvant anthracycline chemotherapy. Patients with HER2-directed therapy, metastatic disease, second primary malignancy, or pre-existing cardiovascular disease were excluded from the analyses as were patients with EF decline for reasons other than anthracycline-induced cardiotoxicity. Primary readout was the diagnostic performance of AI-ECG by area under the curve (AUC) for EFs < 50%. Of 989 consecutive female breast cancer patients, 22 developed a decline in EF attributed to anthracycline therapy over a follow-up time of 9.8 ± 4.2 years. After exclusion of patients who did not have ECGs within 90 days of a TTE, 20 cases and 683 controls remained. The AI-ECG model detected an EF < 50% and ≤ 35% after anthracycline therapy with an AUC of 0.93 and 0.94, respectively. CONCLUSION These data support the use of AI-ECG for cardiotoxicity screening after anthracycline-based chemotherapy. This technology could serve as a gatekeeper to more costly cardiac imaging and could enable patients to monitor themselves over long periods of time.
Collapse
Affiliation(s)
- Johanna E J Jacobs
- Department of Cardiovascular Medicine, Mayo Clinic,
200 First St. SW, Rochester, MN 55905, USA
- Department of Cardiovascular Sciences, KU Leuven,
Leuven, Belgium
| | - Grace Greason
- Department of Cardiovascular Medicine, Mayo Clinic,
200 First St. SW, Rochester, MN 55905, USA
| | - Kathryn E Mangold
- Department of Cardiovascular Medicine, Mayo Clinic,
200 First St. SW, Rochester, MN 55905, USA
| | - Hans Wildiers
- Department of Oncology, University Hospitals Leuven,
Leuven, Belgium
| | - Rik Willems
- Department of Cardiovascular Sciences, KU Leuven,
Leuven, Belgium
| | - Stefan Janssens
- Department of Cardiovascular Sciences, KU Leuven,
Leuven, Belgium
| | - Peter Noseworthy
- Department of Cardiovascular Medicine, Mayo Clinic,
200 First St. SW, Rochester, MN 55905, USA
| | - Francisco Lopez-Jimenez
- Department of Cardiovascular Medicine, Mayo Clinic,
200 First St. SW, Rochester, MN 55905, USA
| | - Jens-Uwe Voigt
- Department of Cardiovascular Sciences, KU Leuven,
Leuven, Belgium
| | - Paul Friedman
- Department of Cardiovascular Medicine, Mayo Clinic,
200 First St. SW, Rochester, MN 55905, USA
| | - Lucas Van Aelst
- Department of Cardiovascular Sciences, KU Leuven,
Leuven, Belgium
| | - Bert Vandenberk
- Department of Cardiovascular Sciences, KU Leuven,
Leuven, Belgium
| | - Zachi Itzhak Attia
- Department of Cardiovascular Medicine, Mayo Clinic,
200 First St. SW, Rochester, MN 55905, USA
| | - Joerg Herrmann
- Department of Cardiovascular Medicine, Mayo Clinic,
200 First St. SW, Rochester, MN 55905, USA
| |
Collapse
|
50
|
Issa AFC. Biomarkers in the Evaluation of Patients Undergoing Chemotherapy with Anthracyclines. Arq Bras Cardiol 2024; 120:e20230821. [PMID: 38451618 PMCID: PMC11098577 DOI: 10.36660/abc.20230821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/06/2023] [Accepted: 12/06/2023] [Indexed: 03/08/2024] Open
Affiliation(s)
- Aurora Felice Castro Issa
- Instituto Nacional de CardiologiaRio de JaneiroRJBrasilInstituto Nacional de Cardiologia, Rio de Janeiro, RJ – Brasil
| |
Collapse
|