1
|
Jian D, Li H, Wang C, Li F, Li R, Jin S, Shen J, Chen J, Zhang W, Pan L, Wang W, Tang H, Jian L, Qi D. METTL3-Mediated m6A Modification of ISG15 mRNA Regulates Doxorubicin-Induced Endothelial Cell Apoptosis. J Cell Mol Med 2025; 29:e70339. [PMID: 39789417 PMCID: PMC11717669 DOI: 10.1111/jcmm.70339] [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: 09/26/2024] [Revised: 12/18/2024] [Accepted: 12/20/2024] [Indexed: 01/12/2025] Open
Abstract
N6-adenosine methylation (m6A) of RNA is involved in the regulation of various diseases. However, its role in chemotherapy-related vascular endothelial injury has not yet been elucidated. We found that methyltransferase-like 3 (METTL3) expression was significantly reduced during doxorubicin (DOX)-induced apoptosis of vascular endothelial cells both in vivo and in vitro, and that silencing of METTL3 further intensified this process. Combined transcriptome and proteome sequencing analyses revealed that the expression levels of interferon-stimulated gene 15 (ISG15) mRNA and protein significantly increased after METTL3 silencing. Methylated RNA immunoprecipitation (meRIP)-quantitative polymerase chain reaction (qPCR) and mRNA stability assays confirmed that METTL3 regulates the expression of ISG15 by methylating the 1,014,147 site on ISG15 RNA, thereby decreasing ISG15 mRNA levels. Silencing ISG15 significantly suppressed DOX-induced endothelial cell apoptosis and dysfunction caused by METTL3 silencing. In summary, our study revealed that METTL3-mediated methylation of ISG15 mRNA is involved in DOX-induced endothelial cell apoptosis and explored potential therapeutic targets for alleviating chemotherapy-associated vascular injury.
Collapse
Affiliation(s)
- Dongdong Jian
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical SciencesPeking University Health Science CenterBeijingChina
- Zhengzhou Key Laboratory of Cardiovascular Aging, Henan Province Key Laboratory for Prevention and Treatment of Coronary Heart Disease, National Health Commission key Laboratory of Cardiovascular Regenerative MedicineCentral China Fuwai Hospital of Zhengzhou University, Fuwai Central China Cardiovascular Hospital & Central China Branch of National Center for Cardiovascular DiseasesZhengzhouHenanChina
| | - Han Li
- Department of CardiologyThe Second Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Tianjian Laboratory of Advanced Biomedical Sciences, Institute of Advanced Biomedical SciencesZhengzhou UniversityZhengzhouHenanChina
| | - Chenqiu Wang
- Department of CardiologyThe Second Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Fang Li
- Department of CardiologyThe Second Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Runhua Li
- Department of CardiologyThe Second Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Shouyi Jin
- Department of CardiologyThe Second Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Jia Shen
- Department of CardiologyThe Second Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Jiamian Chen
- Department of CardiologyThe Second Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Wanjun Zhang
- Department of HematologyHenan Provincial People's HospitalZhengzhouHenanChina
| | - Ling Pan
- Nursing DepartmentHaining People's Hospital (Haining Branch, the First Affiliated Hospital, Zhejiang University)ZhejiangChina
| | - Wengong Wang
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical SciencesPeking University Health Science CenterBeijingChina
| | - Hao Tang
- Zhengzhou Key Laboratory of Cardiovascular Aging, Henan Province Key Laboratory for Prevention and Treatment of Coronary Heart Disease, National Health Commission key Laboratory of Cardiovascular Regenerative MedicineCentral China Fuwai Hospital of Zhengzhou University, Fuwai Central China Cardiovascular Hospital & Central China Branch of National Center for Cardiovascular DiseasesZhengzhouHenanChina
| | - Liguo Jian
- Department of CardiologyThe Second Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Tianjian Laboratory of Advanced Biomedical Sciences, Institute of Advanced Biomedical SciencesZhengzhou UniversityZhengzhouHenanChina
| | - Datun Qi
- Zhengzhou Key Laboratory of Cardiovascular Aging, Henan Province Key Laboratory for Prevention and Treatment of Coronary Heart Disease, National Health Commission key Laboratory of Cardiovascular Regenerative MedicineCentral China Fuwai Hospital of Zhengzhou University, Fuwai Central China Cardiovascular Hospital & Central China Branch of National Center for Cardiovascular DiseasesZhengzhouHenanChina
| |
Collapse
|
2
|
Campos-Rodríguez F, Chiner E, de la Rosa-Carrillo D, García-Cosío B, Hernádez-Hernández JR, Jiménez D, Méndez R, Molina-Molina M, Soto-Campos JG, Vaquero JM, Gonzalez-Barcala FJ. Respiratory Pathology and Cardiovascular Diseases: A Scoping Review. OPEN RESPIRATORY ARCHIVES 2025; 7:100392. [PMID: 39758960 PMCID: PMC11696865 DOI: 10.1016/j.opresp.2024.100392] [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: 11/04/2024] [Accepted: 11/12/2024] [Indexed: 01/07/2025] Open
Abstract
Respiratory diseases and cardiovascular diseases (CVDs) have high prevalence and share common risk factors. In some respiratory diseases such as sleep apnoea and COPD, the evidence of their negative impact on the prognosis of CVDs seems clear. However, in other diseases it is less evident whether there is any direct relationship. With this in mind, our objective was to provide information that may be helpful to better understand the relationship between respiratory pathology and CVDs. There are different reasons for this relationship, such as shared risk factors, common pathophysiological mechanisms, side effects of treatment and the direct effect in the heart and great vessels of respiratory diseases. Indeed, aging and smoking are risk factors for CVDs and also for respiratory diseases such as obstructive sleep apnea (OSA), COPD and interstitial lung diseases (ILD). Furthermore, there are common pathophysiological mechanisms that affect both respiratory diseases and CVDs, such as accelerated atherosclerosis, microvascular dysfunction, endothelial dysfunction, inflammation, hypoxemia and oxidative stress. Besides that, it is well known that lung cancer, sarcoidosis and amyloidosis may directly affect the heart and great vessels. Finally, side effects of drugs for respiratory diseases and the discontinuation of treatments that are necessary for CVDs, such as β-blockers and aspirin, may have a deleterious impact on the cardiovascular system. In conclusion, the coexistence of respiratory diseases and CVDs is very common. It makes modifying diagnostic and therapeutic management necessary and is also a relevant prognostic factor.
Collapse
Affiliation(s)
- Francisco Campos-Rodríguez
- Respiratory Department, Hospital Universitario de Valme, Sevilla, Spain
- Instituto de Biomedicina de Sevilla (IBiS), Sevilla, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Eusebi Chiner
- Respiratory Department, Hospital Universitario of San Juan of Alicante, Alicante, Spain
| | | | - Borja García-Cosío
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Respiratory Department, Hospital Son Espases-IdISBa, Palma de Mallorca, Spain
| | | | - David Jiménez
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Respiratory Department, Ramón y Cajal Hospital and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
- Medicine Department, University of Alcalá, Madrid, Spain
| | - Raúl Méndez
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Respiratory Department, La Fe University and Polytechnic Hospital, Valencia, Spain
- Respiratory Infections, Health Research Institute La Fe (IISLAFE), Valencia, Spain
- Department of Medicine, University of Valencia, Valencia, Spain
| | - María Molina-Molina
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Interstitial Lung Disease (ILD) Unit, Respiratory Department, University Hospital of Bellvitge, IDIBELL, UB, Barcelona, Spain
| | | | - José-Manuel Vaquero
- Department of Pulmonary Medicine and Lung Transplantation, University Hospital Reina Sofia, Avenida Menendez Pidal s/n, 14004 Cordoba, Spain
| | - Francisco-Javier Gonzalez-Barcala
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Translational Research In Airway Diseases Group (TRIAD), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Respiratory Department, University Hospital of Santiago de Compostela, Santiago de Compostela, Spain
- Department of Medicine, University of Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
3
|
Davis NE, Prasitlumkum N, Tan NY. Atrial Fibrillation and Cancer-Epidemiology, Mechanisms, and Management. J Clin Med 2024; 13:7753. [PMID: 39768676 PMCID: PMC11677472 DOI: 10.3390/jcm13247753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/11/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Atrial fibrillation (AF) and cancer are increasingly recognized as interrelated conditions, with cancer patients showing elevated incidences of AF, and there is evidence that AF may sometimes precede cancer diagnoses. This comprehensive review investigates the epidemiology, pathophysiology, and management challenges associated with AF in cancer patients. Epidemiologically, several cancers are more closely related to increased rates of AF, including lung, colorectal, gastrointestinal, and hematologic malignancies. Mechanistically, both AF and cancer share pathophysiological pathways centered on inflammation, oxidative stress, and common cardiovascular risk factors, such as hypertension, obesity, and diabetes. The inflammatory microenvironment in tumors, marked by increased cytokines and growth factors, promotes atrial remodeling and AF susceptibility. Elevated reactive oxygen species (ROS) levels, driven by the metabolic demands of cancer, further contribute to atrial fibrosis and structural changes. Moreover, many anticancer treatments exacerbate AF risk. Management of AF in cancer patients presents many unique challenges and requires a multidisciplinary approach. Rate and rhythm control strategies are complicated by potential drug-drug interactions and limited data surrounding early implementation of rhythm control strategies in cancer patients. Interventional approaches such as catheter ablation, though effective in maintaining sinus rhythm, carry significant perioperative risk in patients with malignancy. Stroke prevention with anticoagulants is essential but requires cautious administration to avoid heightened bleeding risks, particularly in patients undergoing chemotherapy. Further, the limited applicability of standard risk stratification tools like CHA2DS2-VASc in this population complicate decisions regarding anticoagulation. This review highlights the bidirectional relationship between AF and cancer, the difficulties in management, and the critical need for further research in this field.
Collapse
Affiliation(s)
| | - Narut Prasitlumkum
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Nicholas Y. Tan
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
4
|
Wojcik KM, Wilson OW, Shiels MS, Sheppard VB, Jayasekera J. Racial, Ethnic, and Socioeconomic Disparities in Meeting Physical Activity Guidelines among Female Breast Cancer Survivors in the United States. Cancer Epidemiol Biomarkers Prev 2024; 33:1610-1622. [PMID: 39269270 PMCID: PMC11609821 DOI: 10.1158/1055-9965.epi-24-0650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/18/2024] [Accepted: 09/11/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Cancer survivors show low physical activity participation rates in the United States. However, there are limited national-level data on disparities in the prevalence of meeting physical activity guidelines among women with and without breast cancer. We aimed to evaluate national-level trends in meeting physical activity guidelines across demographic and socioeconomic characteristics of breast cancer survivors and women without cancer. METHODS Data for women ≥35 years of age with and without breast cancer were obtained from the 2004 to 2018 National Health Interview Survey. We used National Health Interview Survey sample weights to generate national-level prevalence estimates and calculate absolute and relative indices of disparity for breast cancer survivors and women without cancer meeting aerobic (150 minutes/week) and muscle-strengthening guidelines (2 sessions/week) stratified by demographic (e.g., race/ethnicity) and socioeconomic (e.g., homeownership) characteristics. RESULTS We included 5,845 breast cancer survivors and 160,162 women without cancer. The weighted percentage of breast cancer survivors meeting aerobic guidelines was 37.7% compared with 40.9% of women without cancer. Fewer women met muscle-strengthening guidelines. There were lower proportions of women who were younger (<50-years), were non-Hispanic Black, were Hispanic, worked 35+ hours/week, or rented their home among breast cancer survivors meeting aerobic guidelines compared with women without cancer meeting aerobic guidelines. CONCLUSIONS Breast cancer survivors were less likely to meet physical activity guidelines compared with women without cancer. Demographic and socioeconomic disparities may exist among breast cancer survivors and women without cancer meeting physical activity guidelines. IMPACT Targeted interventions may be necessary to address low physical activity participation among breast cancer survivors.
Collapse
Affiliation(s)
- Kaitlyn M. Wojcik
- Division of Intramural Research, National Institute on Minority Health and Health Disparities, National Institutes of Health, Bethesda, Maryland
| | - Oliver W.A. Wilson
- Division of Intramural Research, National Institute on Minority Health and Health Disparities, National Institutes of Health, Bethesda, Maryland
| | - Meredith S. Shiels
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Vanessa B. Sheppard
- Massey Comprehensive Cancer Center at Virginia Commonwealth University, Richmond, Virginia
| | - Jinani Jayasekera
- Division of Intramural Research, National Institute on Minority Health and Health Disparities, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
5
|
Luo J, He M, Liang C, Huang X, Zhu Y, Hu D, Yan J, Li M, Lin H, Liao W, Bin J, Guan Z, Zheng C, Liao Y. Canagliflozin reverses doxorubicin-induced cardiotoxicity via restoration of autophagic homeostasis. Toxicol Appl Pharmacol 2024; 495:117183. [PMID: 39631538 DOI: 10.1016/j.taap.2024.117183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/18/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
Sodium-glucose cotransporter 2 inhibitors (SGLT2i) have been reported as successful for preventing doxorubicin (DOX) -induced cardiotoxicity (DIC), but the underlying mechanisms are elusive. This study aimed to determine whether canagliflozin, an SGLT2i, protects against DIC by regulation of autophagic flux in cardiomyocytes through a mechanism independent of SGLT2. The differentially expressed autophagy-related genes (ARGs) in DIC were analyzed. Neonatal rat cardiomyocytes (NRCMs), H9C2 rat cardiomyocytes or C57BL/6 mice were treated with canagliflozin or vehicle. The effects on cellular apoptosis and autophagy were investigated using qRT-PCR, western blotting and immunofluorescence. Additionally, cardiac function, myocardial fibrosis, and apoptosis of cardiomyocytes were also assessed in mice. The potential molecular targets of canagliflozin were identified through molecular docking analysis. A total of 26 differentially expressed ARGs were identified. Canagliflozin significantly activated autophagic flux and inhibited apoptosis of cardiomyocytes in both DOX-treated H9C2 rat cardiomyocytes and NRCMs. In a murine model of DIC, canagliflozin improved cardiac dysfunction by suppressing cardiac remodeling, fibrosis, and apoptosis. Moreover, canagliflozin promoted autophagy by enhancing SIRT1 levels and inhibiting the PI3K/Akt/mTOR signaling pathway. Immunofluorescence assays revealed that canagliflozin promoted the translocation of LC3 from the nucleus to the cytoplasm. Molecular docking analysis confirmed that canagliflozin has high affinity for targets associated with DIC. These findings suggest that canagliflozin protects cardiomyocytes from DOX-induced cell death by activating SIRT1, inhibiting the PI3K/Akt/mTOR pathway, and enhancing autophagic flux.
Collapse
Affiliation(s)
- Jianping Luo
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Cardiology, Ganzhou People's Hospital, Ganzhou, China
| | - Mingyuan He
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Changzhu Liang
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoxia Huang
- Cardiovascular Center, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, Guangdong, China
| | - Yingqi Zhu
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Donghong Hu
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Junyu Yan
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mingjue Li
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hairuo Lin
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jianping Bin
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou, China; Cardiovascular Center, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, Guangdong, China
| | - Ziyun Guan
- Cardiovascular Center, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, Guangdong, China
| | - Cankun Zheng
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Yulin Liao
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou, China; Cardiovascular Center, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, Guangdong, China.
| |
Collapse
|
6
|
Cautela J, Croizier C, Inchiappa L, Goncalves T, Stocker N, Tchernonog E. [Cardiovascular adverse effects of Bruton tyrosine kinase inhibitors: Pathophysiological mechanisms, screening, and management]. Bull Cancer 2024; 111:1142-1153. [PMID: 39516119 DOI: 10.1016/j.bulcan.2024.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/19/2024] [Accepted: 09/23/2024] [Indexed: 11/16/2024]
Abstract
The covalent Bruton tyrosine kinase inhibitors (iBTKs) have profoundly transformed the management of B-cell lymphoid malignancies, particularly chronic lymphocytic leukemia (CLL). These targeted therapies, with ibrutinib as the pioneer, have paved the way for significant improvement in the prognosis of many patients. With second-generation iBTKs such as acalabrutinib and zanubrutinib, the therapeutic landscape has expanded, offering potential new options for patients with CLL. This review focuses on the cardiovascular adverse effects associated with these treatments. It delves into the underlying pathophysiological mechanisms of these effects, highlighting the complex interactions between these molecules and the cardiovascular system. Additionally, it examines the frequency of adverse effects according to the type of iBTK, drawing on data from clinical trials and real-world clinical practice. Finally, the importance of close cardio-oncological monitoring is emphasized, with essential collaboration between hematologists and cardiologists. Strategies for screening and managing cardiovascular adverse effects are also discussed, emphasizing the need for a proactive approach in managing these complications. Experts propose a pragmatic follow-up of these patients, through a central illustration and a figure adapted from European cardio-oncology guidelines, to simplify hematologists' practice.
Collapse
Affiliation(s)
- Jennifer Cautela
- Unité d'insuffisance cardiaque et de maladies valvulaires, service de cardiologie, centre de recherche cardiovasculaire et nutrition (C2VN), Inserm 1263, Inrae 1260, centre universitaire méditerranéen de cardio-oncologie, hôpital Nord, Assistance publique-Hôpitaux de Marseille, université d'Aix-Marseille, Marseille, France.
| | - Carolyne Croizier
- Service de thérapie cellulaire et d'hématologie clinique adulte, équipe d'Accueil 7453 CHELTER, CHU Estaing, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Luca Inchiappa
- Service d'onco-hématologie, institut Paoli-Calmettes, Marseille, France
| | - Trecy Goncalves
- Department of Cardiology, hôpital Lariboisière, Assistance publique-Hôpitaux de Paris, Inserm U-942, université Paris Cité, 75010 Paris, France
| | - Nicolas Stocker
- Service d'hématologie clinique et de thérapie cellulaire, équipe Inserm UMRs 938, centre de recherche Saint-Antoine, hôpital Saint-Antoine, AP-PH, Sorbonne université, Paris, France
| | | |
Collapse
|
7
|
Jasińska-Stroschein M, Glajzner P. Searching for Old and New Small-Molecule Protein Kinase Inhibitors as Effective Treatments in Pulmonary Hypertension-A Systematic Review. Int J Mol Sci 2024; 25:12858. [PMID: 39684570 DOI: 10.3390/ijms252312858] [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: 11/06/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Treatment options for pulmonary arterial hypertension (PAH) have improved substantially in the last 30 years, but there is still a need for novel molecules that can regulate the excessive accumulation of pulmonary artery smooth muscle cells (PASMCs) and consequent vascular remodeling. One set of possible candidates are protein kinases. The study provides an overview of existing preclinical and clinical data regarding small-molecule protein kinase inhibitors in PAH. Online databases were searched from 2001 to 2023 according to PRISMA. The corpus included preclinical studies demonstrating alterations in at least one PH-related parameter following chronic exposure to an individual protein kinase inhibitor, as well as prospective clinical reports including healthy adults or those with PAH, with primary outcomes defined as safety or efficacy of an individual small-molecule protein kinase inhibitor. Several models in preclinical protocols (93 papers) have been proposed for studying small-molecule protein kinase inhibitors in PAH. In total, 51 kinase inhibitors were tested. Meta-analysis of preclinical results demonstrated seralutinib, sorafenib, fasudil hydrochloride, and imatinib had the most comprehensive effects on PH with anti-inflammatory, anti-oxidant, and anti-proliferative potential. Fasudil demonstrated more than 70% animal survival with the longest experimental period, while dasatinib, nintedanib, and (R)-crizotinib could deteriorate PAH. The substances targeting the same kinases often varied considerably in their activity, and such heterogeneity may be due to the variety of causes. Recent studies have addressed the molecules that affect multiple networks such as PDG-FRα/β/CSF1R/c-KIT/BMPR2 or FKBP12/mTOR. They also focus on achieving a satisfactory safety profile using innovative inhalation formulations Many small-molecule protein kinase inhibitors are able to control migration, proliferation and survival in PASMCs in preclinical observations. Standardized animal models can successfully reduce inter-study heterogeneity and thereby facilitate successful identification of candidate drugs for further evaluations.
Collapse
Affiliation(s)
| | - Paulina Glajzner
- Department of Biopharmacy, Medical University of Lodz, ul. Muszyńskiego 1, 90-151 Lodz, Poland
| |
Collapse
|
8
|
Gawrys O, Jíchová Š, Miklovič M, Husková Z, Kikerlová S, Sadowski J, Kollárová P, Lenčová-Popelova O, Hošková L, Imig JD, Mazurova Y, Kolář F, Melenovský V, Štěrba M, Červenka L. Characterization of a new model of chemotherapy-induced heart failure with reduced ejection fraction and nephrotic syndrome in Ren-2 transgenic rats. Hypertens Res 2024; 47:3126-3146. [PMID: 39245782 PMCID: PMC11534684 DOI: 10.1038/s41440-024-01865-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/26/2024] [Accepted: 07/31/2024] [Indexed: 09/10/2024]
Abstract
All anthracyclines, including doxorubicin (DOXO), the most common and still indispensable drug, exhibit cardiotoxicity with inherent risk of irreversible cardiomyopathy leading to heart failure with reduced ejection fraction (HFrEF). Current pharmacological strategies are clearly less effective for this type of HFrEF, hence an urgent need for new therapeutic approaches. The prerequisite for success is thorough understanding of pathophysiology of this HFrEF form, which requires an appropriate animal model of the disease. The aim of this study was to comprehensively characterise a novel model of HF with cardiorenal syndrome, i.e. DOXO-induced HFrEF with nephrotic syndrome, in which DOXO was administered to Ren-2 transgenic rats (TGR) via five intravenous injections in a cumulative dose of 10 mg/kg of body weight (BW). Our analysis included survival, echocardiography, as well as histological examination of the heart and kidneys, blood pressure, but also a broad spectrum of biomarkers to evaluate cardiac remodelling, fibrosis, apoptosis, oxidative stress and more. We have shown that the new model adequately mimics the cardiac remodelling described as "eccentric chamber atrophy" and myocardial damage typical for DOXO-related cardiotoxicity, without major damage of the peritoneum, lungs and liver. This pattern corresponds well to a clinical situation of cancer patients receiving anthracyclines, where HF develops with some delay after the anticancer therapy. Therefore, this study may serve as a comprehensive reference for all types of research on DOXO-related cardiotoxicity, proving especially useful in the search for new therapeutic strategies.
Collapse
Affiliation(s)
- Olga Gawrys
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Šárka Jíchová
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Matúš Miklovič
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Zuzana Husková
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Soňa Kikerlová
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Janusz Sadowski
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Petra Kollárová
- Department of Pharmacology, Faculty of Medicine in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Olga Lenčová-Popelova
- Department of Pharmacology, Faculty of Medicine in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Lenka Hošková
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - John D Imig
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Yvona Mazurova
- Department of Histology and Embryology, Faculty of Medicine in Hradec Králové, Hradec Králové, Czech Republic
| | - František Kolář
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Vojtěch Melenovský
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Martin Štěrba
- Department of Pharmacology, Faculty of Medicine in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Luděk Červenka
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
- Department of Internal Medicine I, Cardiology, University Hospital Olomouc and Palacký University, Olomouc, Czech Republic.
| |
Collapse
|
9
|
Bai B, Ma Y, Liu D, Zhang Y, Zhang W, Shi R, Zhou Q. DNA damage caused by chemotherapy has duality, and traditional Chinese medicine may be a better choice to reduce its toxicity. Front Pharmacol 2024; 15:1483160. [PMID: 39502534 PMCID: PMC11534686 DOI: 10.3389/fphar.2024.1483160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/14/2024] [Indexed: 11/08/2024] Open
Abstract
Background DNA damage induced by chemotherapy has duality. It affects the efficacy of chemotherapy and constrains its application. An increasing number of studies have shown that traditional Chinese medicine (TCM) is highly effective in reducing side-effects induced by chemotherapy due to its natural, non-toxic and many sourced from food. Recent advancements have demonstrated survival rates are improved attributable to effective chemotherapy. DNA damage is the principal mechanism underlying chemotherapy. However, not all instances of DNA damage are beneficial. Chemotherapy induces DNA damage in normal cells, leading to side effects. It affects the efficacy of chemotherapy and constrains its application. Objectives This review aims to summarize the dual nature of DNA damage induced by chemotherapy and explore how TCM can mitigate chemotherapy-induced side effects. Results The review summarized the latest research progress in DNA damage caused by chemotherapy and the effect of alleviating side effects by TCM. It focused on advantages and disadvantages of chemotherapy, the mechanism of drugs and providing insights for rational and effective clinical treatment and serving as a basis for experiment. In this review, we described the mechanisms of DNA damage, associated chemotherapeutics, and their toxicity. Furthermore, we explored Chinese herb that can alleviate chemotherapy-induced side-effects. Conclusion We highlight key mechanisms of DNA damage caused by chemotherapeutics and discuss specific TCM herbs that have shown potential in reducing these side effects. It can provide reference for clinical and basic research.
Collapse
Affiliation(s)
- Bufan Bai
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yingrui Ma
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Deng Liu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yifei Zhang
- Department of Intensive Care Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weihong Zhang
- Breast Surgery Department, Baoshan Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Rong Shi
- Department of Intensive Care Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qianmei Zhou
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Dongfang Hospital Affiliated to Shanghai Tongji University, Shanghai, China
| |
Collapse
|
10
|
Todorova VK, Azhar G, Stone A, Malapati SJ, Che Y, Zhang W, Makhoul I, Wei JY. Neutrophil Biomarkers Can Predict Cardiotoxicity of Anthracyclines in Breast Cancer. Int J Mol Sci 2024; 25:9735. [PMID: 39273682 PMCID: PMC11395913 DOI: 10.3390/ijms25179735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 08/29/2024] [Accepted: 08/31/2024] [Indexed: 09/15/2024] Open
Abstract
Doxorubicin (DOX), a commonly used anticancer agent, causes cardiotoxicity that begins with the first dose and may progress to heart failure years after treatment. An inflammatory response associated with neutrophil recruitment has been recognized as a mechanism of DOX-induced cardiotoxicity. This study aimed to validate mRNA expression of the previously identified biomarkers of DOX-induced cardiotoxicity, PGLYRP1, CAMP, MMP9, and CEACAM8, and to assay their protein expression in the peripheral blood of breast cancer patients. Blood samples from 40 breast cancer patients treated with DOX-based chemotherapy were collected before and after the first chemotherapy cycle and > 2 years after treatment. The protein and gene expression of PGLYRP1/Tag7, CAMP/LL37, MMP9/gelatinase B, and CEACAM8/CD66b were determined using ELISA and reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Receiver operating characteristic (ROC) curve analysis was used to determine the diagnostic value of each candidate biomarker. Patients with cardiotoxicity (n = 20) had significantly elevated levels of PGLYRP1, CAMP, MMP9, and CEACAM8 at baseline, after the first dose of DOX-based chemotherapy, and at > 2 years after treatment relative to patients without cardiotoxicity (n = 20). The first dose of DOX induced significantly higher levels of all examined biomarkers in both groups of patients. At > 2 years post treatment, the levels of all but MMP9 dropped below the baseline. There was a good correlation between the expression of mRNA and the target proteins. We demonstrate that circulating levels of PGLYRP1, CAMP, MMP9, and CEACAM8 can predict the cardiotoxicity of DOX. This novel finding may be of value in the early identification of patients at risk for cardiotoxicity.
Collapse
Affiliation(s)
- Valentina K Todorova
- Division of Hematology/Oncology, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Central Arkansas Veterans Healthcare System, Little Rock, AR 72205, USA
| | - Gohar Azhar
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Annjanette Stone
- Central Arkansas Veterans Healthcare System, Little Rock, AR 72205, USA
| | - Sindhu J Malapati
- Division of Hematology/Oncology, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Yingni Che
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Wei Zhang
- Department of Mathematics and Statistics, University of Arkansas at Little Rock, Little Rock, AR 72205, USA
| | - Issam Makhoul
- Division of Hematology/Oncology, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Jeanne Y Wei
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
11
|
Hao X, Zhang Z, Kong J, Ma R, Mao C, Peng X, Ru K, Liu L, Zhao C, Mo X, Cai M, Yu X, Lin Q. Hypothesis paper: GDF15 demonstrated promising potential in Cancer diagnosis and correlated with cardiac biomarkers. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:56. [PMID: 39232830 PMCID: PMC11373216 DOI: 10.1186/s40959-024-00263-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 08/30/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Cardiovascular toxicity represents a significant adverse consequence of cancer therapies, yet there remains a paucity of effective biomarkers for its timely monitoring and diagnosis. To give a first evidence able to elucidate the role of Growth Differentiation Factor 15 (GDF15) in the context of cancer diagnosis and its specific association with cardiac indicators in cancer patients, thereby testing its potential in predicting the risk of CTRCD (cancer therapy related cardiac dysfunction). METHODS Analysis of differentially expressed genes (DEGs), including GDF15, was performed by utilizing data from the public repositories of the Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO). Cardiomyopathy is the most common heart disease and its main clinical manifestations, such as heart failure and arrhythmia, are similar to those of CTRCD. Examination of GDF15 expression was conducted in various normal and cancerous tissues or sera, using available database and serum samples. The study further explored the correlation between GDF15 expression and the combined detection of cardiac troponin-T (c-TnT) and N-terminal prohormone of brain natriuretic peptide (NT-proBNP), assessing the combined diagnostic utility of these markers in predicting risk of CTRCD through longitudinal electrocardiograms (ECG). RESULTS GDF15 emerged as a significant DEG in both cancer and cardiomyopathy disease models, demonstrating good diagnostic efficacy across multiple cancer types compared to healthy controls. GDF15 levels in cancer patients correlated with the established cardiac biomarkers c-TnT and NT-proBNP. Moreover, higher GDF15 levels correlated with an increased risk of ECG changes in the cancer cohort. CONCLUSION GDF15 demonstrated promising diagnostic potential in cancer identification; higher GDF15, combined with elevated cardiac markers, may play a role in the monitoring and prediction of CTRCD risk.
Collapse
Affiliation(s)
- Xiaohe Hao
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, 440 Ji-Yan Road, Jinan, Shandong Province, 250117, PR China
| | - Zhenyu Zhang
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, 440 Ji-Yan Road, Jinan, Shandong Province, 250117, PR China
| | - Jing Kong
- Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Rufei Ma
- Electrocardiogram Room, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Cuiping Mao
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, 440 Ji-Yan Road, Jinan, Shandong Province, 250117, PR China
| | - Xun Peng
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, 440 Ji-Yan Road, Jinan, Shandong Province, 250117, PR China
| | - Kun Ru
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, 440 Ji-Yan Road, Jinan, Shandong Province, 250117, PR China
| | - Lisheng Liu
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, 440 Ji-Yan Road, Jinan, Shandong Province, 250117, PR China
| | - Chuanxi Zhao
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, 440 Ji-Yan Road, Jinan, Shandong Province, 250117, PR China
| | - Xinkai Mo
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, 440 Ji-Yan Road, Jinan, Shandong Province, 250117, PR China
| | - Meijuan Cai
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Xiangguo Yu
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, 440 Ji-Yan Road, Jinan, Shandong Province, 250117, PR China.
| | - Qinghai Lin
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, 440 Ji-Yan Road, Jinan, Shandong Province, 250117, PR China.
| |
Collapse
|
12
|
Lee S, Alsamarrai A, Xiao A, Wang TKM. Prevention of anthracycline and trastuzumab-induced decline in left ventricular ejection fraction with angiotensin-converting enzyme inhibitors or angiotensin receptor blocker: a narrative systematic review of randomised controlled trials. Intern Med J 2024; 54:1254-1263. [PMID: 38874281 DOI: 10.1111/imj.16437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 05/13/2024] [Indexed: 06/15/2024]
Abstract
Cancer therapy-related cardiac dysfunction (CTRCD) is a complication of selected cancer therapy agents associated with decline in left ventricular ejection fraction (LVEF). Angiotensin-converting enzyme inhibitors (ACEIs) and angiotensin receptor blockers (ARBs) have established benefits in heart failure with reduced ejection fraction, but their efficacy for preventing CTRCD remains controversial. This narrative systematic review assessed the efficacy and safety of ACEI/ARB in the prevention of cancer therapy LVEF decline. We systematically searched PubMed, Embase and Cochrane from January 1980 to June 2022. Studies of interest were randomised controlled trials of patients with normal LVEF and active malignancy receiving cancer therapy, randomised to receive either an ACEI or ARB compared with a control group. The outcome was the change in LVEF from baseline to the end of the follow-up period. Death, clinical heart failure and adverse drug reactions were recorded. A total of 3731 search records were screened and 12 studies were included, comprising a total of 1645 participants. Nine studies assessed the prevention of anthracycline-induced LVEF decline, of which five showed a beneficial effect (1%-14% higher LVEF in treated groups), whereas four studies showed no effect. Three studies assessed the prevention of trastuzumab-induced LVEF decline, of which one showed a beneficial effect (4% higher LVEF) in a subset of participants. There are mixed data regarding the efficacy of ACEI/ARB in preventing the LVEF decline in patients undergoing anthracycline or trastuzumab therapy, with evidence suggesting no clinically meaningful benefit observed in recent studies.
Collapse
Affiliation(s)
- Simon Lee
- Cardiology Department, Middlemore Hospital, Auckland, New Zealand
- Cardiology Department, Tauranga Hospital, Tauranga, New Zealand
| | - Ammar Alsamarrai
- Green Lane Cardiovascular Service, Auckland City Hospital, Auckland, New Zealand
| | - Amy Xiao
- Cardiology Department, Middlemore Hospital, Auckland, New Zealand
| | - Tom K M Wang
- Green Lane Cardiovascular Service, Auckland City Hospital, Auckland, New Zealand
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
13
|
Cho I, You SC, Cha MJ, Hwang HJ, Cho EJ, Kim HJ, Park SM, Kim SE, Lee YG, Youn JC, Park CS, Shim CY, Chung WB, Sohn IS. Cancer therapy-related cardiac dysfunction and the role of cardiovascular imaging: systemic review and opinion paper from the Working Group on Cardio-Oncology of the Korean Society of Cardiology. J Cardiovasc Imaging 2024; 32:13. [PMID: 39075626 PMCID: PMC11288116 DOI: 10.1186/s44348-024-00014-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 01/14/2024] [Indexed: 07/31/2024] Open
Abstract
Cardio-oncology is a critical field due to the escalating significance of cardiovascular toxicity as a side effect of anticancer treatments. Cancer therapy-related cardiac dysfunction (CTRCD) is a prevalent condition associated with cardiovascular toxicity, necessitating effective strategies for prediction, monitoring, management, and tracking. This comprehensive review examines the definition and risk stratification of CTRCD, explores monitoring approaches during anticancer therapy, and highlights specific cardiovascular toxicities linked to various cancer treatments. These include anthracyclines, HER2-targeted agents, vascular endothelial growth factor inhibitors, immune checkpoint inhibitors, chimeric antigen receptor T-cell therapies, and tumor-infiltrating lymphocytes therapies. Incorporating the Korean data, this review offers insights into the regional nuances in managing CTRCD. Using systematic follow-up incorporating cardiovascular imaging and biomarkers, a better understanding and management of CTRCD can be achieved, optimizing the cardiovascular health of both cancer patients and survivors.
Collapse
Affiliation(s)
- Iksung Cho
- Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seng-Chan You
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Min-Jae Cha
- Department of Radiology, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| | - Hui-Jeong Hwang
- Department of Cardiology, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul, Republic of Korea
| | - Eun Jeong Cho
- Division of Cardiology, Department of Internal Medicine, Heart and Brain Hospital, Chung-Ang University Gwangmyeong Hospital, Chung-Ang University College of Medicine, Gwangmyeong, Republic of Korea
| | - Hee Jun Kim
- Division of Medical Oncology, Department of Internal Medicine, Chung-Ang University Hospital, Seoul, Republic of Korea
| | - Seong-Mi Park
- Division of Cardiology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Sung-Eun Kim
- Department of Cardiovascular Medicine, Kangdong Sacred Heart Hospital, Seoul, Republic of Korea
| | - Yun-Gyoo Lee
- Division of Hematology & Medical Oncology, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jong-Chan Youn
- Division of Cardiology, Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Chan Seok Park
- Division of Cardiology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Chi Young Shim
- Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Woo-Baek Chung
- Division of Cardiology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Il Suk Sohn
- Department of Cardiology, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
14
|
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
|
15
|
Mohamed MO, Ghosh AK, Banerjee A, Mamas M. Socioeconomic and Ethnic Disparities in the Process of Care and Outcomes Among Cancer Patients With Acute Coronary Syndrome. Can J Cardiol 2024; 40:1146-1153. [PMID: 38537671 DOI: 10.1016/j.cjca.2024.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/26/2024] [Accepted: 03/14/2024] [Indexed: 05/01/2024] Open
Abstract
Cancer and acute coronary syndrome (ACS) are the leading causes of morbidity and mortality globally, with many shared risk factors. There are several challenges to the management of patients with cancer presenting with ACS, owing to their higher baseline risk profile, the complexities of their cancer-related therapies and prognosis, and their higher risk of adverse outcomes after ACS. Although previous studies have demonstrated disparities in the care of both cancer and ACS among patients from ethnic minorities and socioeconomic deprivation, there is limited evidence around the magnitude of such disparities specifically in cancer patients presenting with ACS. This review summarises the current literature on differences in prevalence and management of ACS among patients with cancer from ethnic minorities and socioeconomically deprived backgrounds, as well as the gaps in evidence around the care of this high-risk population and potential solutions.
Collapse
Affiliation(s)
- Mohamed O Mohamed
- Keele Cardiovascular Research Group, Keele University, Keele, United Kingdom; Institute of Health Informatics, University College London, London, United Kingdom; Barts Heart Centre, St. Bartholomew's Hospital, London, United Kingdom
| | - Arjun K Ghosh
- Barts Heart Centre, St. Bartholomew's Hospital, London, United Kingdom; Cardio-Oncology Service, Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - Amitava Banerjee
- Institute of Health Informatics, University College London, London, United Kingdom; Barts Heart Centre, St. Bartholomew's Hospital, London, United Kingdom
| | - Mamas Mamas
- Keele Cardiovascular Research Group, Keele University, Keele, United Kingdom.
| |
Collapse
|
16
|
Boen HM, Alaerts M, Goovaerts I, Saenen JB, Franssen C, Vorlat A, Vermeulen T, Heidbuchel H, Van Laer L, Loeys B, Van Craenenbroeck EM. Variants in structural cardiac genes in patients with cancer therapy-related cardiac dysfunction after anthracycline chemotherapy: a case control study. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:26. [PMID: 38689299 PMCID: PMC11059765 DOI: 10.1186/s40959-024-00231-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 04/23/2024] [Indexed: 05/02/2024]
Abstract
BACKGROUND Variants in cardiomyopathy genes have been identified in patients with cancer therapy-related cardiac dysfunction (CTRCD), suggesting a genetic predisposition for the development of CTRCD. The diagnostic yield of genetic testing in a CTRCD population compared to a cardiomyopathy patient cohort is not yet known and information on which genes should be assessed in this population is lacking. METHODS We retrospectively included 46 cancer patients with a history of anthracycline induced CTRCD (defined as a decrease in left ventricular ejection fraction (LVEF) to < 50% and a ≥ 10% reduction from baseline by echocardiography). Genetic testing was performed for 59 established cardiomyopathy genes. Only variants of uncertain significance and (likely) pathogenic variants were included. Diagnostic yield of genetic testing was compared with a matched cohort of patients with dilated cardiomyopathy (DCM, n = 46) and a matched cohort of patients without cardiac disease (n = 111). RESULTS Average LVEF at time of CTRCD diagnosis was 30.1 ± 11.0%. Patients were 52.9 ± 14.6 years old at time of diagnosis and 30 (65.2%) were female. Most patients were treated for breast cancer or lymphoma, with a median doxorubicin equivalent dose of 300 mg/m2 [112.5-540.0]. A genetic variant, either pathogenic, likely pathogenic or of uncertain significance, was identified in 29/46 (63.0%) of patients with CTRCD, which is similar to the DCM cohort (34/46, 73.9%, p = 0.262), but significantly higher than in the negative control cohort (47/111, 39.6%, p = 0.018). Variants in TTN were the most prevalent in the CTRCD cohort (43% of all variants). All (likely) pathogenic variants identified in the CTRCD cohort were truncating variants in TTN. There were no significant differences in severity of CTRCD and in recovery rate in variant-harbouring individuals versus non-variant harbouring individuals. CONCLUSIONS In this case-control study, cancer patients with anthracycline-induced CTRCD have an increased burden of genetic variants in cardiomyopathy genes, similar to a DCM cohort. If validated in larger prospective studies, integration of genetic data in risk prediction models for CTRCD may guide cancer treatment. Moreover, genetic results have important clinical impact, both for the patient in the setting of precision medicine, as for the family members that will receive genetic counselling.
Collapse
Affiliation(s)
- Hanne M Boen
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium.
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium.
| | - Maaike Alaerts
- Centrum of Medical Genetics, GENCOR, Antwerp University Hospital and University of Antwerp, Antwerp, Belgium
| | - Inge Goovaerts
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
- Centrum of Medical Genetics, GENCOR, Antwerp University Hospital and University of Antwerp, Antwerp, Belgium
| | - Johan B Saenen
- 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
| | - Anne Vorlat
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
| | - Tom Vermeulen
- Research Group Cardiovascular Diseases, 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
| | - Lut Van Laer
- Centrum of Medical Genetics, GENCOR, Antwerp University Hospital and University of Antwerp, Antwerp, Belgium
| | - Bart Loeys
- Centrum of Medical Genetics, GENCOR, Antwerp University Hospital and 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
|
17
|
Tran TN, Lee S, Kim HJ, Lee Y, Tu TM, Choi JH, Song JW, Cho H. Treatment-related cardiovascular events in patients with non-small cell lung cancer: Evidence from real-world data with a competing risks approach. Cancer 2024; 130:1303-1315. [PMID: 38103206 DOI: 10.1002/cncr.35143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 09/25/2023] [Accepted: 11/08/2023] [Indexed: 12/18/2023]
Abstract
BACKGROUND Understanding cancer treatment-related cardiovascular (CV) events is important for cancer care; however, comprehensive evaluation of CV events in patients with lung cancer is limited. This study aimed to assess the cumulative incidence and associated risks of various CV event types in patients with non-small cell lung cancer (NSCLC). METHODS A total of 7868 individuals aged 40 years and older, recently diagnosed with NSCLC (2007-2018), were assessed with data obtained from the National Cancer Center, Korea. This study included nine types of CV events. A 2-year cumulative incidence function (CIF) of CV events was estimated, with death as a competing event. The associated risks were assessed by subdistribution hazard ratio (sHR) in the Fine-Gray competing risks model. RESULTS CV events were observed in 7.8% of patients with NSCLC, with the most frequently observed types being atrial fibrillation and flutter (AF) (2.7%), venous thromboembolic disease (2.0%), and cerebrovascular disease (CeVD) (1.5%). Overall, all CV events were highest in the group treated with systemic therapy (CIF, 10.6%; 95% confidence interval [CI], 9.5%-11.8%), followed by those treated with surgery (CIF, 10.0%; 95% CI, 8.6%-11.6%); the incidence of AF (CIF, 5.7%; 95% CI, 4.6%-7.0%) was highest in patients treated with surgery. Individuals treated with systemic therapy were found to exhibit a higher CeVD risk than those treated with surgery (sHR, 4.12; 95% CI, 1.66-10.23). Among the patients who underwent surgery, those with lobectomy and pneumonectomy had a higher AF risk (vs. wedge resection/segmentectomy; sHR, 7.79; 95% CI, 1.87-32.42; sHR, 8.10; 95% CI, 1.60-40.89). CONCLUSIONS These findings revealed treatment-related CV event risks in patients with NSCLC, which suggests that the risk of AF in surgery and CeVD in systemic therapy should be paid more attention to achieve a better prognosis and improve cancer survivorship outcomes. PLAIN LANGUAGE SUMMARY Atrial fibrillation and flutter (AF) is the most common cardiovascular event, particularly at a high risk in patients with non-small cell lung cancer (NSCLC) undergoing surgery. Patients receiving surgery with poor performance status, diagnosed with regional stage, and undergoing lobectomy or pneumonectomy are at a high risk of AF. Systemic/radiotherapy is associated with cerebrovascular and ischemic heart disease in patients with NSCLC.
Collapse
Affiliation(s)
- Thi-Ngoc Tran
- Department of Cancer Control and Population Health, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Republic of Korea
| | - Sanghee Lee
- Department of Cancer Control and Population Health, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Republic of Korea
| | - Hak Jin Kim
- Division of Cardiology, Department of Internal Medicine, National Cancer Center, Goyang, Republic of Korea
- Department of Cardiology, Gumdan Top General Hospital, Incheon, Republic of Korea
| | - Youngjoo Lee
- Division of Hematology and Oncology, Department of Internal Medicine, National Cancer Center, Goyang, Republic of Korea
| | - Thao Minh Tu
- Department of Cancer Control and Population Health, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Republic of Korea
| | - Jin-Ho Choi
- Department of Thoracic Surgery, Research Institute and Hospital, National Cancer Center, Goyang, Republic of Korea
| | - Jae Won Song
- Department of Thoracic Surgery, Research Institute and Hospital, National Cancer Center, Goyang, Republic of Korea
| | - Hyunsoon Cho
- Department of Cancer AI and Digital Health, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Republic of Korea
- Integrated Biostatistics Branch, Division of Cancer Data Science, Research Institute, National Cancer Center, Goyang, Republic of Korea
| |
Collapse
|
18
|
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: 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: 08/07/2023] [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
|
19
|
Geraldes C, Roque A, Sarmento-Ribeiro AB, Neves M, Ionita A, Gerivaz R, Tomé A, Afonso S, Silveira MP, Sousa P, Bergantim R, João C. Practical management of disease-related manifestations and drug toxicities in patients with multiple myeloma. Front Oncol 2024; 14:1282300. [PMID: 38585008 PMCID: PMC10995327 DOI: 10.3389/fonc.2024.1282300] [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: 08/23/2023] [Accepted: 02/07/2024] [Indexed: 04/09/2024] Open
Abstract
Multiple myeloma (MM) is a very heterogeneous disease with multiple symptoms and clinical manifestations. MM affects mainly elderly patients and is difficult to manage in the presence of comorbidities, polypharmacy, frailty and adverse events of disease-targeted drugs. The rapid changes in MM treatment resulting from constant innovations in this area, together with the introduction of numerous new drugs with distinct mechanisms of action and toxicity profiles, have led to an increased complexity in the therapeutic decision-making and patient management processes. The prolonged exposure to novel agents, sometimes in combination with conventional therapies, makes this management even more challenging. A careful balance between treatment efficacy and its tolerability should be considered for every patient. During treatment, a close monitoring of comorbidities, disease-related manifestations and treatment side effects is recommended, as well as a proactive approach, with reinforcement of information and patient awareness for the early recognition of adverse events, allowing prompt therapeutic adjustments. In this review, we discuss various issues that must be considered in the treatment of MM patients, while giving practical guidance for monitoring, prevention and management of myeloma-related manifestations and treatment-related toxicities.
Collapse
Affiliation(s)
- Catarina Geraldes
- Serviço de Hematologia Clínica, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Adriana Roque
- Serviço de Hematologia Clínica, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Institute of Physiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Ana Bela Sarmento-Ribeiro
- Serviço de Hematologia Clínica, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Manuel Neves
- Hemato-Oncology Unit, Champalimaud Foundation, Lisboa, Portugal
| | - Alina Ionita
- Hematology Department, Portuguese Institute of Oncology Francisco Gentil, Lisboa, Portugal
| | - Rita Gerivaz
- Serviço de Hemato-oncologia, Hospital Garcia de Orta, Lisboa, Portugal
| | - Ana Tomé
- Serviço de Hemato-oncologia, Hospital Garcia de Orta, Lisboa, Portugal
| | - Sofia Afonso
- Serviço de Hematologia Clínica, Centro Hospitalar Universitário Cova da Beira, Covilhã, Portugal
- Faculdade de Ciências da Saúde, Universidade da Beira Interior, Covilhã, Portugal
| | - Maria Pedro Silveira
- Serviço de Imuno-Hemoterapia, Hospital Prof. Doutor Fernando Fonseca, EPE, Amadora, Portugal
| | - Patrícia Sousa
- Serviço de Imuno-Hemoterapia, Hospital Prof. Doutor Fernando Fonseca, EPE, Amadora, Portugal
| | - Rui Bergantim
- Serviço de Hematologia Clínica, Centro Hospitalar Universitário de São João (CHUSJ), Porto, Portugal
- Instituto de Investigação e Inovaçáo em Saúde, Universidade do Porto, Porto, Portugal
- Institute of Pathology and Molecular Immunology, Abel Salazar Institute of Biomedical Sciences, University of Porto, Porto, Portugal
| | - Cristina João
- Hemato-Oncology Unit, Champalimaud Foundation, Lisboa, Portugal
- NOVA Medical School, Universidade Nova de Lisboa, Lisboa, Portugal
| |
Collapse
|
20
|
Li D, Zhang W, Fu H, Wang X, Tang Y, Huang C. DL-3- n-butylphthalide attenuates doxorubicin-induced acute cardiotoxicity via Nrf2/HO-1 signaling pathway. Heliyon 2024; 10:e27644. [PMID: 38486757 PMCID: PMC10938138 DOI: 10.1016/j.heliyon.2024.e27644] [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: 11/22/2023] [Revised: 03/05/2024] [Accepted: 03/05/2024] [Indexed: 03/17/2024] Open
Abstract
Doxorubicin (DOX) is a widely used chemotherapeutic drug known to cause dose-dependent myocardial toxicity, which limits its clinical potential. DL-3-n-butylphthalide (NBP), a substance extracted from celery seed species, has a number of pharmacological properties, such as antioxidant, anti-inflammatory, and anti-apoptotic actions. However, whether NBP can protect against DOX-induced acute myocardial toxicity is still unclear. Therefore, this study was designed to investigate the potential protective effects of NBP against DOX-induced acute myocardial injury and its underlying mechanism. By injecting 15 mg/kg of DOX intraperitoneally, eight-week-old male C57BL6 mice suffered an acute myocardial injury. The treatment group of mice received 80 mg/kg NBP by gavage once daily for 14 days. To mimic the cardiotoxicity of DOX, 1uM DOX was administered to H9C2 cells in vitro. In comparison to the DOX group, the results showed that NBP improved cardiac function and decreased serum levels of cTnI, LDH, and CK-MB. Additionally, HE staining demonstrated that NBP attenuated cardiac fibrillar lysis and breakage in DOX-treated mouse hearts. Western blotting assay and immunofluorescence staining suggested that NBP attenuated DOX-induced oxidative stress, apoptosis, and inflammation both in vivo and in vitro. Mechanistically, NBP significantly upregulated the Nrf2/HO-1 signaling pathway, while the Nrf2 inhibitor ML385 prevented NBP from protecting the myocardium from DOX-induced myocardial toxicity in vitro. In conclusion, Our results indicate that NBP alleviates DOX-induced myocardial toxicity by activating the Nrf2/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Dengke Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Wei Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Hui Fu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Yanhong Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Congxin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| |
Collapse
|
21
|
Ibrahim AA, Nsairat H, Al-Sulaibi M, El-Tanani M, Jaber AM, Lafi Z, Barakat R, Abuarqoub DA, Mahmoud IS, Obare SO, Aljabali AAA, Alkilany AM, Alshaer W. Doxorubicin conjugates: a practical approach for its cardiotoxicity alleviation. Expert Opin Drug Deliv 2024; 21:399-422. [PMID: 38623735 DOI: 10.1080/17425247.2024.2343882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/29/2024] [Indexed: 04/17/2024]
Abstract
INTRODUCTION Doxorubicin (DOX) emerges as a cornerstone in the arsenal of potent chemotherapeutic agents. Yet, the clinical deployment of DOX is tarnished by its proclivity to induce severe cardiotoxic effects, culminating in heart failure and other consequential morbidities. In response, a panoply of strategies has undergone rigorous exploration over recent decades, all aimed at attenuating DOX's cardiotoxic impact. The advent of encapsulating DOX within lipidic or polymeric nanocarriers has yielded a dual triumph, augmenting DOX's therapeutic efficacy while mitigating its deleterious side effects. AREAS COVERED Recent strides have spotlighted the emergence of DOX conjugates as particularly auspicious avenues for ameliorating DOX-induced cardiotoxicity. These conjugates entail the fusion of DOX through physical or chemical bonds with diminutive natural or synthetic moieties, polymers, biomolecules, and nanoparticles. This spectrum encompasses interventions that impinge upon DOX's cardiotoxic mechanism, modulate cellular uptake and localization, confer antioxidative properties, or refine cellular targeting. EXPERT OPINION The endorsement of DOX conjugates as a compelling stratagem to mitigate DOX-induced cardiotoxicity resounds from this exegesis, amplifying safety margins and the therapeutic profile of this venerated chemotherapeutic agent. Within this ambit, DOX conjugates stand as a beacon of promise in the perpetual pursuit of refining chemotherapy-induced cardiac compromise.
Collapse
Affiliation(s)
- Abed Alqader Ibrahim
- Department of Nanoscience, Joint School of Nanoscience and Nanoengineering, University of North Carolina at Greensboro, Greensboro, NC, USA
| | - Hamdi Nsairat
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Mazen Al-Sulaibi
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Mohamed El-Tanani
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
- College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Areej M Jaber
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Zainab Lafi
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Rahmeh Barakat
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Duaa Azmi Abuarqoub
- Department of Pharmacology and Biomedical Sciences, Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, Jordan
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| | - Ismail Sami Mahmoud
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, Zarqa, Jordan
| | - Sherine O Obare
- Department of Nanoscience, Joint School of Nanoscience and Nanoengineering, University of North Carolina at Greensboro, Greensboro, NC, USA
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering, North Carolina Agricultural and Technical State University, Greensboro, NC, USA
| | - Alaa A A Aljabali
- Faculty of Pharmacy, Department of Pharmaceutical Sciences, Yarmouk University, Irbid, Jordan
| | | | - Walhan Alshaer
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| |
Collapse
|
22
|
Di Lisi D, Madaudo C, Faro DC, Rossetto L, Triolo OF, Losi V, Galassi AR, Monte IP, Novo G. The added value of the HFA/ICOS score in the prediction of chemotherapy-related cardiac dysfunction in breast cancer. J Cardiovasc Med (Hagerstown) 2024; 25:218-224. [PMID: 38305134 DOI: 10.2459/jcm.0000000000001589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
BACKGROUND The 2022 ESC Guidelines on Cardio-Oncology recommend baseline cardiovascular risk stratification before starting anticancer drugs, using the new risk assessment tools proposed by the Heart Failure Association (HFA) and the International Cardio-Oncology Society (ICOS).Our study aimed to assess the clinical application of HFA/ICOS risk score in breast cancer patients undergoing chemotherapy and its usefulness in predicting the development of chemotherapy-related cardiac dysfunction (CTRCD). METHODS A prospective multicentric study enrolled 109 breast cancer patients treated with anthracyclines with or without trastuzumab. A cardiological evaluation, including ECG and echocardiogram at baseline (T0), 3 (T1), 6 (T2), and 12 months (T3) after starting treatment was performed. HFA/ICOS score was assessed in all patients. The population was divided into low, medium, high, and very-high risk.During follow-up, CTRCD and other cardiovascular events have been evaluated. RESULTS 61 patients were low risk, 37 medium, 9 high, 2 very-high risk criteria. We found a significantly higher incidence of overall cardiotoxicity (CTRCD and other cardiovascular events) in the very-high risk group (100%) compared with the medium (29%) and low risk groups (13%). CTRCD incidence was also significantly higher in the high risk group (55%). CTRCD resulted as being associated with baseline arterial hypertension and baseline HFA/ICOS risk score of high ( p = 0.006) or very-high ( p < 0.0001). CONCLUSION Our study confirms the HFA/ICOS score's ability to predict cardiovascular toxicity in breast cancer women and the need for close monitoring especially in high and very-high risk patients.
Collapse
Affiliation(s)
- Daniela Di Lisi
- Division of Cardiology, University Hospital Paolo Giaccone
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE) 'G. D'Alessandro', University of Palermo, Palermo
| | - Cristina Madaudo
- Division of Cardiology, University Hospital Paolo Giaccone
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE) 'G. D'Alessandro', University of Palermo, Palermo
| | - Denise Cristiana Faro
- Department of General Surgery and Medical-Surgery Specialities (CHIRMED), University of Catania
- Cardiology Unit, University Hospital G. Rodolico - San Marco, Catania, Italy
| | - Ludovico Rossetto
- Division of Cardiology, University Hospital Paolo Giaccone
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE) 'G. D'Alessandro', University of Palermo, Palermo
| | - Oreste Fabio Triolo
- Division of Cardiology, University Hospital Paolo Giaccone
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE) 'G. D'Alessandro', University of Palermo, Palermo
| | - Valentina Losi
- Department of General Surgery and Medical-Surgery Specialities (CHIRMED), University of Catania
- Cardiology Unit, University Hospital G. Rodolico - San Marco, Catania, Italy
| | - Alfredo Ruggero Galassi
- Division of Cardiology, University Hospital Paolo Giaccone
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE) 'G. D'Alessandro', University of Palermo, Palermo
| | - Ines Paola Monte
- Department of General Surgery and Medical-Surgery Specialities (CHIRMED), University of Catania
- Cardiology Unit, University Hospital G. Rodolico - San Marco, Catania, Italy
| | - Giuseppina Novo
- Division of Cardiology, University Hospital Paolo Giaccone
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE) 'G. D'Alessandro', University of Palermo, Palermo
| |
Collapse
|
23
|
Mesitskaya DF, Fashafsha ZZ, Poltavskaya MG, Andreev DA, Levshina AR, Sultygova EA, Gognieva D, Chomakhidze P, Kuznetsova N, Suvorov A, Marina I. S, Poddubskaya E, Novikova A, Bykova A, Kopylov P. A single-lead ECG based cardiotoxicity detection in patients on polychemotherapy. IJC HEART & VASCULATURE 2024; 50:101336. [PMID: 38304727 PMCID: PMC10831811 DOI: 10.1016/j.ijcha.2024.101336] [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: 09/26/2023] [Revised: 12/04/2023] [Accepted: 01/04/2024] [Indexed: 02/03/2024]
Abstract
Background Anti-cancer treatment can be fraught with cardiovascular complications, which is the most common cause of death among oncological survivors. Without appropriate cardiomonitoring during anti-cancer treatment, it becomes challenging to detect early signs of cardiovascular complications. In order to achieve higher survival rates, it is necessary to monitor oncological patients outpatiently after anti-cancer treatment administration. In this regard, we aim to evaluate the efficacy of single-lead ECG remote monitoring to detect cardiotoxicity in cancer patients with minimal cardiovascular diseases after the first cycle of polychemotherapy. Materials and methods The study included patients 162 patients over 18 years old with first diagnosed different types of solid tumors, planed for adjuvant (within 8 weeks after surgery) or neoadjuvant polychemotherapy. All patients were monitored, outpatiently, during 14-21 days (depending on the regimen of polychemotherapy) after polychemotherapy administration using single-lead ECG. Results QTc > 500 mc prolongation was detected in 8 patients (6.6 %), first-diagnosed arial fibrillation was detected in 11 patients (9 %) in period after chemotherapy administration. Moreover, left ventricular diastolic dysfunction using single-lead ECG after polychemotherapy was detected in 49 (40.1 %) patients with sensitivity 80 %, specificity 95 %, AUC 0.88 (95 % CI, 0.82-0.93). Conclusions The side effects of cancer treatment may cause life-threatening risks. Early identification of cardiotoxicity plays a vital role in the solution of this problem. Using portable devices to detect early cardiotoxicity is a simple, convenient and affordable screening method, that can be used for promptly observation of patients.
Collapse
Affiliation(s)
- Dinara F. Mesitskaya
- Department of Cardiology, Functional and Ultrasound Diagnostics of N.V. Sklifosovsky Institute for Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Zaki Z.A. Fashafsha
- World-Class Research Center "Digital biodesign and personalized healthcare", Sechenov First Moscow State Medical University, Moscow, Russia
| | - Maria G. Poltavskaya
- Department of Cardiology, Functional and Ultrasound Diagnostics of N.V. Sklifosovsky Institute for Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Denis A. Andreev
- Department of Cardiology, Functional and Ultrasound Diagnostics of N.V. Sklifosovsky Institute for Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Anna R. Levshina
- Department of Cardiology, Functional and Ultrasound Diagnostics of N.V. Sklifosovsky Institute for Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Elizaveta A. Sultygova
- World-Class Research Center "Digital biodesign and personalized healthcare", Sechenov First Moscow State Medical University, Moscow, Russia
| | - Daria Gognieva
- World-Class Research Center "Digital biodesign and personalized healthcare", Sechenov First Moscow State Medical University, Moscow, Russia
- Department of Cardiology, Functional and Ultrasound Diagnostics of N.V. Sklifosovsky Institute for Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Petr Chomakhidze
- World-Class Research Center "Digital biodesign and personalized healthcare", Sechenov First Moscow State Medical University, Moscow, Russia
- Department of Cardiology, Functional and Ultrasound Diagnostics of N.V. Sklifosovsky Institute for Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Natalia Kuznetsova
- World-Class Research Center "Digital biodesign and personalized healthcare", Sechenov First Moscow State Medical University, Moscow, Russia
| | - Alexander Suvorov
- World-Class Research Center "Digital biodesign and personalized healthcare", Sechenov First Moscow State Medical University, Moscow, Russia
| | - Sekacheva Marina I.
- World-Class Research Center "Digital biodesign and personalized healthcare", Sechenov First Moscow State Medical University, Moscow, Russia
- Institute for Personalized Oncology, Center "Digital Biodesign and Personalized Healthcare" I.M. Sechenov First Moscow State Medical University Moscow, Russia Moscow, Russia
| | - Elena Poddubskaya
- Department of Cardiology, Functional and Ultrasound Diagnostics of N.V. Sklifosovsky Institute for Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Alena Novikova
- Department of Cardiology, Functional and Ultrasound Diagnostics of N.V. Sklifosovsky Institute for Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Aleksandra Bykova
- Department of Cardiology, Functional and Ultrasound Diagnostics of N.V. Sklifosovsky Institute for Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Philipp Kopylov
- World-Class Research Center "Digital biodesign and personalized healthcare", Sechenov First Moscow State Medical University, Moscow, Russia
- Department of Cardiology, Functional and Ultrasound Diagnostics of N.V. Sklifosovsky Institute for Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| |
Collapse
|
24
|
Romann SW, Finke D, Heckmann MB, Hund H, Giannitsis E, Katus HA, Frey N, Lehmann LH. Cardiological parameters predict mortality and cardiotoxicity in oncological patients. ESC Heart Fail 2024; 11:366-377. [PMID: 38012070 PMCID: PMC10804154 DOI: 10.1002/ehf2.14587] [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: 03/03/2023] [Revised: 08/24/2023] [Accepted: 10/31/2023] [Indexed: 11/29/2023] Open
Abstract
AIMS Oncological patients suspected at risk for cardiotoxicity are recommended to undergo intensified cardiological surveillance. We investigated the value of cardiac biomarkers and patient-related risk factors [age, cardiovascular risk factors (CVRFs), and cardiac function] for the prediction of all-cause mortality (ACM) and the development of cardiotoxicity. METHODS AND RESULTS Between January 2016 and December 2020, patients with oncological diseases admitted to the Cardio-Oncology Unit at the Heidelberg University Hospital were included. They were evaluated by medical history, physical examination, 12-lead electrocardiogram, 2D echocardiography, and cardiac biomarkers [high-sensitivity cardiac troponin T (hs-cTnT) and N-terminal pro-brain natriuretic peptide (NT-proBNP)]. The primary endpoint was defined as ACM and the secondary endpoint was defined as cardiotoxicity, as defined by the European Society of Cardiology. Of the 1971 patients enrolled, the primary endpoint was reached by 490 patients (25.7%) with a median of 363.5 [interquartile range (IQR) 121.8, 522.5] days after presentation. Hs-cTnT of ≥ 7 ng/L [odds ratio (OR) 1.82, P < 0.001] and NT-proBNP (OR 1.98, P < 0.001) were independent predictors of ACM, while reduced left ventricular ejection fraction was not associated with increased ACM (P = 0.85). The secondary endpoint was reached by 182 patients (9.2%) with a median of 793.5 [IQR 411.2, 1165.0] days. Patients with multiple CVRFs (defined as high risk, n = 886) had an increased risk of cardiotoxicity (n = 100/886, 11.3%; hazard ratio 1.57, P = 0.004). They showed elevated baseline values of hs-cTnT (OR 1.60, P = 0.006) and NT-proBNP (OR 4.00, P < 0.001) and had an increased risk of ACM (OR 1.43, P = 0.031). CONCLUSIONS In cancer patients, CVRF accumulation predicts cardiotoxicity whereas elevated hs-cTnT or NT-proBNP levels are associated with ACM. Accordingly, less intensive surveillance protocols may be warranted in patients with low cardiac biomarker levels and absence of CVRFs.
Collapse
Affiliation(s)
- Sebastian W. Romann
- Department of Internal Medicine III: Cardiology, Angiology and Pulmonology, Cardio‐Oncology UnitHeidelberg University HospitalIm Neuenheimer Feld 41069120HeidelbergGermany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/MannheimHeidelbergGermany
| | - Daniel Finke
- Department of Internal Medicine III: Cardiology, Angiology and Pulmonology, Cardio‐Oncology UnitHeidelberg University HospitalIm Neuenheimer Feld 41069120HeidelbergGermany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/MannheimHeidelbergGermany
| | - Markus B. Heckmann
- Department of Internal Medicine III: Cardiology, Angiology and Pulmonology, Cardio‐Oncology UnitHeidelberg University HospitalIm Neuenheimer Feld 41069120HeidelbergGermany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/MannheimHeidelbergGermany
| | - Hauke Hund
- Department of Internal Medicine III: Cardiology, Angiology and Pulmonology, Cardio‐Oncology UnitHeidelberg University HospitalIm Neuenheimer Feld 41069120HeidelbergGermany
| | - Evangelos Giannitsis
- Department of Internal Medicine III: Cardiology, Angiology and Pulmonology, Cardio‐Oncology UnitHeidelberg University HospitalIm Neuenheimer Feld 41069120HeidelbergGermany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/MannheimHeidelbergGermany
| | - Hugo A. Katus
- Department of Internal Medicine III: Cardiology, Angiology and Pulmonology, Cardio‐Oncology UnitHeidelberg University HospitalIm Neuenheimer Feld 41069120HeidelbergGermany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/MannheimHeidelbergGermany
| | - Norbert Frey
- Department of Internal Medicine III: Cardiology, Angiology and Pulmonology, Cardio‐Oncology UnitHeidelberg University HospitalIm Neuenheimer Feld 41069120HeidelbergGermany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/MannheimHeidelbergGermany
| | - Lorenz H. Lehmann
- Department of Internal Medicine III: Cardiology, Angiology and Pulmonology, Cardio‐Oncology UnitHeidelberg University HospitalIm Neuenheimer Feld 41069120HeidelbergGermany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/MannheimHeidelbergGermany
- German Cancer Research Center (DKFZ)HeidelbergGermany
| |
Collapse
|
25
|
Wang S, Wang Y, Wang S. The role of stress echocardiography in identifying cardiotoxicity: an in-depth exploration. Front Cardiovasc Med 2024; 11:1236966. [PMID: 38343875 PMCID: PMC10853441 DOI: 10.3389/fcvm.2024.1236966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 01/15/2024] [Indexed: 01/06/2025] Open
Abstract
Cancer treatment might cause heart failure and deteriorate the patients' quality of life. Despite the wide use of conventional echocardiography, it often fails to detect cardiotoxicity until advanced cardiac dysfunction at potentially irreversible stages. Advanced techniques, such as three-dimensional imaging and strain analysis in stress echocardiography, have shown promise in identifying cardiotoxicity at subclinical stages, even when traditional measures remain within normal ranges. These novel techniques have been shown to identify cardiac impairment in 30%-50% of the patients undergoing potentially cardiotoxic chemotherapy, which allows for early intervention and enhanced patient management. Although professional societies are advocating for the inclusion of these techniques into routine monitoring protocols, more research is needed to optimize and standardize their use across various centers and chemotherapeutic agents. This review explores the role of stress echocardiography in the early detection and monitoring of chemotherapy-induced cardiotoxicity. It delves into current knowledge and emerging research, aiming to provide a comprehensive understanding and to highlight areas worthy of further investigation.
Collapse
Affiliation(s)
- Sijia Wang
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yi Wang
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Shuang Wang
- Department of Geriatrics, The People's Hospital of Changshou, Chongqing, China
| |
Collapse
|
26
|
Yanagida S, Kanda Y. [Prediction of Cardiac Toxicity by Anti-cancer Drugs Using iPSC Cardiomyocytes]. YAKUGAKU ZASSHI 2024; 144:265-271. [PMID: 38432935 DOI: 10.1248/yakushi.23-00164-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
Recent advances in cancer therapy have significantly improved the survival rate of patients with cancer. In contrast, anti-cancer drug-induced adverse effects, especially cardiotoxicity, have come to affect patients' prognosis and quality of life. Therefore, there is a growing need to understand the anti-cancer drug-induced cardiotoxicity. Human induced pluripotent stem (iPS) cell-derived cardiomyocytes (hiPSC-CMs) have been used to assess drug-induced cardiotoxicity by improving the predictability of clinical cardiotoxicity and the principles of the 3Rs (replacement, reduction and refinement). To predict the anti-cancer drug-induced cardiotoxicity, we developed a novel method to assess drug-induced proarrhythmia risk using hiPSC-CMs by participating in the international validation. In addition, we established the chronic contractility toxicity assessment by image-based motion analysis. The compound BMS-986094, which was withdrawn from clinical trials, inhibited contractility velocity and relaxation velocity in hiPSC-CMs. Currently, we are trying to investigate the predictability of the contractility assay by comparing the hiPSC-CM data with adverse events reports from real-world database. In this review, we would like to introduce the novel imaging-based contractility method using hiPSC-CMs and future perspectives in anti-cancer drug-induced cardiotoxicity.
Collapse
Affiliation(s)
- Shota Yanagida
- Division of Pharmacology, National Institute of Health Sciences
- Graduate School of Biomedical and Health Sciences (Pharmaceutical Sciences), Hiroshima University
| | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences
| |
Collapse
|
27
|
Mohammadi H, Hosseini H, Bordbar M, Mehdizadegan N, Amoozgar H, Edraki MR, Naghshzan A, Naderi N, Abedi E, Keshavarz K. Permanent longitudinal strain damage of cardiotoxic drugs in childhood cancer: What is the safe level? Ann Pediatr Cardiol 2024; 17:36-44. [PMID: 38933046 PMCID: PMC11198932 DOI: 10.4103/apc.apc_146_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 02/02/2024] [Accepted: 02/02/2024] [Indexed: 06/28/2024] Open
Abstract
Objective Anthracycline administration in children is associated with cardiac dysfunction. Speckle-tracking echocardiography (STE) can detect subclinical cardiac damage that may go undetected by conventional two-dimensional (2D) echocardiography. This study aims to investigate medium-term anthracycline cardiotoxicity using STE and determine a safer administrable level of anthracyclines (ACs). Methods This observational case-control study enrolled 37 healthy controls and 78 pediatric cancer survivors who received chemotherapy. The patients were divided into two groups: cardiotoxic received (CR) and cardiotoxic free (CF). Data on segmental longitudinal strain (LS), global LS (GLS), and 2D echocardiographic parameters were collected after a drug-free period of at least one year. Results A total of 115 children with a mean age of 108 ± 55 months, of whom 66% were males, were included in the study. Both the groups of cancer survivors exhibited significantly reduced GLS compared to healthy controls (CR vs. controls, P = 0.001; CF vs. controls, P = 0.013), but no significant difference in left ventricular ejection fraction (LVEF) was observed (P = 0.75). Overall, cancer survivors treated with ACs demonstrated a significant reduction in strain in 10 left ventricular segments, particularly in the basal segments (P < 0.05). Among CR patients, those with impaired GLS (n = 43, GLS worse than -21.9) had significantly higher mean age and cumulative anthracycline dose compared to CR patients with normal GLS (age, P = 0.024; anthracycline dosage, P = 0.036). Using an anthracycline cutoff of 223 mg/m2 resulted in a higher detection rate (49% vs. 25%) and fewer missed cases (51% vs. 74%) compared to the 360 mg/m2 anthracycline cutoff. Conclusion Childhood cancer survivors demonstrate significantly reduced GLS while preserving a normal LVEF, which does not differ significantly from reference values of healthy children. The reduction in strain appears to be associated with higher anthracycline doses and older age. Lowering the anthracycline threshold to 223 mg/m2 may improve the predictability of a decline in cardiac function using strain imaging at medium-term follow-up.
Collapse
Affiliation(s)
- Hamid Mohammadi
- Department of Pediatric, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hossein Hosseini
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammadreza Bordbar
- Hematology Research Center, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nima Mehdizadegan
- Cardiovascular Research Center, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamid Amoozgar
- Cardiovascular Research Center, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Reza Edraki
- Neonatal Research Center, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Naghshzan
- Cardiovascular Research Center, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nima Naderi
- Anesthesiology and Critical Care Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Elham Abedi
- Hematology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Kambiz Keshavarz
- Social Determinants of Health Research Center, School of Medicine, Yasuj University of Medical Sciences, Yasuj, Iran
| |
Collapse
|
28
|
Qiu Y, Jiang P, Huang Y. Anthracycline-induced cardiotoxicity: mechanisms, monitoring, and prevention. Front Cardiovasc Med 2023; 10:1242596. [PMID: 38173817 PMCID: PMC10762801 DOI: 10.3389/fcvm.2023.1242596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 12/04/2023] [Indexed: 01/05/2024] Open
Abstract
Anthracyclines are the most fundamental and important treatment of several cancers especially for lymphoma and breast cancer. However, their use is limited by a dose-dependent cardiotoxicity which may emerge early at the initiation of anthracycline administration or several years after termination of the therapy. A full comprehending of the mechanisms of anthracycline-induced cardiotoxicity, which has not been achieved and is currently under the efforts, is critical to the advance of developing effective methods to protect against the cardiotoxicity, as well as to early detect and treat it. Therefore, we review the recent progress of the mechanism underlying anthracycline-induced cardiotoxicity, as well as approaches to monitor and prevent this issue.
Collapse
Affiliation(s)
- Yun Qiu
- Department of Cardiology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Piao Jiang
- Department of Oncology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
- The First Clinical Medical College, Nanchang University, Nanchang, China
| | - Yingmei Huang
- Department of Cardiology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| |
Collapse
|
29
|
Timóteo AT, Ribeiras R, Calé R, Moura B, G Almeida A, Gavina C, Cabral S, António N, Franco F, Ilhão Moreira R, Geraldes F, Machado AP, Palma F, Pires da Silva V, Gonçalves L. Cardiovascular diseases in women - Consensus document of the Sociedade Portuguesa de Cardiologia, Sociedade Portuguesa de Ginecologia, Sociedade Portuguesa de Obstetrícia e Medicina Materno-Fetal, Sociedade Portuguesa de Contraceção e Associação Portuguesa de Medicina Geral e Familiar. Rev Port Cardiol 2023; 42:1001-1015. [PMID: 36566887 DOI: 10.1016/j.repc.2022.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
The main objective of this consensus statement from the Portuguese Society of Cardiology, the Portuguese Society of Gynecology, the Portuguese Society of Obstetrics and Maternal-Fetal Medicine, Portuguese Society of Contraception, Portuguese Association of General Practice and Family Medicine is to improve cardiovascular care for women. It includes a brief review of the state-of-the-art of cardiovascular diseases in women and of the links to other fields such as Gynaecology, Obstetrics and Endocrinology. It also provides final recommendations to help clinicians working in care of women's health.
Collapse
Affiliation(s)
| | | | - Rita Calé
- Sociedade Portuguesa de Cardiologia, Lisboa, Portugal
| | - Brenda Moura
- Sociedade Portuguesa de Cardiologia, Lisboa, Portugal
| | - Ana G Almeida
- Sociedade Portuguesa de Cardiologia, Lisboa, Portugal
| | | | - Sofia Cabral
- Sociedade Portuguesa de Cardiologia, Lisboa, Portugal
| | | | - Fátima Franco
- Sociedade Portuguesa de Cardiologia, Lisboa, Portugal
| | | | | | - Ana Paula Machado
- Sociedade Portuguesa de Obstetrícia e Medicina Materno-Fetal, Lisboa, Portugal
| | - Fátima Palma
- Sociedade Portuguesa de Contraceção, Lisboa, Portugal
| | | | | |
Collapse
|
30
|
Almeida ALC, Melo MDTD, Bihan DCDSL, Vieira MLC, Pena JLB, Del Castillo JM, Abensur H, Hortegal RDA, Otto MEB, Piveta RB, Dantas MR, Assef JE, Beck ALDS, Santo THCE, Silva TDO, Salemi VMC, Rocon C, Lima MSM, Barberato SH, Rodrigues AC, Rabschkowisky A, Frota DDCR, Gripp EDA, Barretto RBDM, Silva SME, Cauduro SA, Pinheiro AC, Araujo SPD, Tressino CG, Silva CES, Monaco CG, Paiva MG, Fisher CH, Alves MSL, Grau CRPDC, Santos MVCD, Guimarães ICB, Morhy SS, Leal GN, Soares AM, Cruz CBBV, Guimarães Filho FV, Assunção BMBL, Fernandes RM, Saraiva RM, Tsutsui JM, Soares FLDJ, Falcão SNDRS, Hotta VT, Armstrong ADC, Hygidio DDA, Miglioranza MH, Camarozano AC, Lopes MMU, Cerci RJ, Siqueira MEMD, Torreão JA, Rochitte CE, Felix A. Position Statement on the Use of Myocardial Strain in Cardiology Routines by the Brazilian Society of Cardiology's Department Of Cardiovascular Imaging - 2023. Arq Bras Cardiol 2023; 120:e20230646. [PMID: 38232246 PMCID: PMC10789373 DOI: 10.36660/abc.20230646] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2024] Open
Abstract
Central Illustration : Position Statement on the Use of Myocardial Strain in Cardiology Routines by the Brazilian Society of Cardiology's Department Of Cardiovascular Imaging - 2023 Proposal for including strain in the integrated diastolic function assessment algorithm, adapted from Nagueh et al.67 Am: mitral A-wave duration; Ap: reverse pulmonary A-wave duration; DD: diastolic dysfunction; LA: left atrium; LASr: LA strain reserve; LVGLS: left ventricular global longitudinal strain; TI: tricuspid insufficiency. Confirm concentric remodeling with LVGLS. In LVEF, mitral E wave deceleration time < 160 ms and pulmonary S-wave < D-wave are also parameters of increased filling pressure. This algorithm does not apply to patients with atrial fibrillation (AF), mitral annulus calcification, > mild mitral valve disease, left bundle branch block, paced rhythm, prosthetic valves, or severe primary pulmonary hypertension.
Collapse
Affiliation(s)
| | | | | | - Marcelo Luiz Campos Vieira
- Instituto do Coração da Faculdade de Medicina da Universidade de São Paulo (Incor/FMUSP), São Paulo, SP - Brasil
| | - José Luiz Barros Pena
- Faculdade Ciências Médicas de Minas Gerais, Belo Horizonte, MG - Brasil
- Hospital Felicio Rocho, Belo Horizonte, MG - Brasil
| | | | - Henry Abensur
- Beneficência Portuguesa de São Paulo, São Paulo, SP - Brasil
| | | | | | | | | | | | | | | | | | - Vera Maria Cury Salemi
- Instituto do Coração da Faculdade de Medicina da Universidade de São Paulo (Incor/FMUSP), São Paulo, SP - Brasil
| | - Camila Rocon
- Hospital do Coração (HCor), São Paulo, SP - Brasil
| | - Márcio Silva Miguel Lima
- Instituto do Coração da Faculdade de Medicina da Universidade de São Paulo (Incor/FMUSP), São Paulo, SP - Brasil
| | | | | | | | | | - Eliza de Almeida Gripp
- Hospital Pró-Cardiaco, Rio de Janeiro, RJ - Brasil
- Hospital Universitário Antônio Pedro da Universidade Federal Fluminense (UFF), Rio de Janeiro, RJ - Brasil
| | | | | | | | | | | | | | | | | | | | | | | | | | - Maria Veronica Camara Dos Santos
- Departamento de Cardiologia Pediátrica (DCC/CP) da Sociedade Brasileira de Cardiologia (SBC), São Paulo, SP - Brasil
- Sociedade Brasileira de Oncologia Pediátrica, São Paulo, SP - Brasil
| | | | | | - Gabriela Nunes Leal
- Instituto da Criança e do Adolescente do Hospital das Clinicas Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP - Brasil
| | | | | | | | | | | | | | | | | | | | - Viviane Tiemi Hotta
- Instituto do Coração da Faculdade de Medicina da Universidade de São Paulo (Incor/FMUSP), São Paulo, SP - Brasil
- Grupo Fleury, São Paulo, SP - Brasil
| | | | - Daniel de Andrade Hygidio
- Hospital Nossa Senhora da Conceição, Tubarão, SC - Brasil
- Universidade do Sul de Santa Catarina (UNISUL), Tubarão, SC - Brasil
| | - Marcelo Haertel Miglioranza
- EcoHaertel - Hospital Mae de Deus, Porto Alegre, RS - Brasil
- Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS - Brasil
| | | | | | | | | | - Jorge Andion Torreão
- Hospital Santa Izabel, Salvador, BA - Brasil
- Santa Casa da Bahia, Salvador, BA - Brasil
| | - Carlos Eduardo Rochitte
- Instituto do Coração da Faculdade de Medicina da Universidade de São Paulo (Incor/FMUSP), São Paulo, SP - Brasil
- Hospital do Coração (HCor), São Paulo, SP - Brasil
| | - Alex Felix
- Diagnósticos da América SA (DASA), São Paulo, SP - Brasil
- Instituto Nacional de Cardiologia (INC), Rio de Janeiro, RJ - Brasil
| |
Collapse
|
31
|
Patricelli C, Lehmann P, Oxford JT, Pu X. Doxorubicin-induced modulation of TGF-β signaling cascade in mouse fibroblasts: insights into cardiotoxicity mechanisms. Sci Rep 2023; 13:18944. [PMID: 37919370 PMCID: PMC10622533 DOI: 10.1038/s41598-023-46216-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/30/2023] [Indexed: 11/04/2023] Open
Abstract
Doxorubicin (DOX)-induced cardiotoxicity has been widely observed, yet the specific impact on cardiac fibroblasts is not fully understood. Additionally, the modulation of the transforming growth factor beta (TGF-β) signaling pathway by DOX remains to be fully elucidated. This study investigated DOX's ability to modulate the expression of genes and proteins involved in the TGF-β signaling cascade in mouse fibroblasts from two sources by assessing the impact of DOX treatment on TGF-β inducible expression of pivotal genes and proteins within fibroblasts. Mouse embryonic fibroblasts (NIH3T3) and mouse primary cardiac fibroblasts (CFs) were treated with DOX in the presence of TGF-β1 to assess changes in protein levels by western blot and changes in mRNA levels by quantitative reverse transcriptase polymerase chain reaction (qRT-PCR). Our results revealed a dose-dependent reduction in cellular communication network factor 2 (CCN2) protein levels upon DOX treatment in both NIH3T3 and CFs, suggesting an antifibrotic activity by DOX in these fibroblasts. However, DOX only inhibited the TGF-β1 induced expression of COL1 in NIH3T3 cells but not in CFs. In addition, we observed that DOX treatment reduced the expression of BMP1 in NIH3T3 but not primary cardiac fibroblasts. No significant changes in SMAD2 protein expression and phosphorylation in either cells were observed after DOX treatment. Finally, DOX inhibited the expression of Atf4 gene and increased the expression of Cdkn1a, Id1, Id2, Runx1, Tgfb1, Inhba, Thbs1, Bmp1, and Stat1 genes in NIH3T3 cells but not CFs, indicating the potential for cell-specific responses to DOX and its modulation of the TGF-β signaling pathway.
Collapse
Affiliation(s)
- Conner Patricelli
- Biomolecular Sciences Graduate Programs, Boise State University, Boise, ID, 83725-1512, USA
| | - Parker Lehmann
- Idaho College of Osteopathic Medicine, Meridian, ID, 83642-8046, USA
| | - Julia Thom Oxford
- Biomolecular Sciences Graduate Programs, Boise State University, Boise, ID, 83725-1512, USA
- Biomolecular Research Center, Boise State University, Boise, ID, 83725-1511, USA
- Department of Biological Sciences, Boise State University, Boise, ID, 83725-1515, USA
| | - Xinzhu Pu
- Biomolecular Research Center, Boise State University, Boise, ID, 83725-1511, USA.
- Department of Biological Sciences, Boise State University, Boise, ID, 83725-1515, USA.
| |
Collapse
|
32
|
Chen MH, Epstein SF. Tailored to a Woman's Heart: Gender Cardio-Oncology Across the Lifespan. Curr Cardiol Rep 2023; 25:1461-1474. [PMID: 37819431 PMCID: PMC11034750 DOI: 10.1007/s11886-023-01967-7] [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: 09/08/2023] [Indexed: 10/13/2023]
Abstract
PURPOSE OF REVIEW Females outnumber males among long-term cancer survivors, primarily as a result of the prevalence of breast cancer. Late cardiovascular effects of cancer develop over several decades, which for many women, may overlap with reproductive and lifecycle events. Thus, women require longitudinal cardio-oncology care that anticipates and responds to their evolving cardiovascular risk. RECENT FINDINGS Women may experience greater cardiotoxicity from cancer treatments compared to men and a range of treatment-associated hormonal changes that increase cardiometabolic risk. Biological changes at critical life stages, including menarche, pregnancy, and menopause, put female cancer patients and survivors at a unique risk of cardiovascular disease. Women also face distinct psychosocial and physical barriers to accessing cardiovascular care. We describe the need for a lifespan-based approach to cardio-oncology for women. Cardio-oncology care tailored to women should rigorously consider cancer treatment/outcomes and concurrent reproductive/hormonal changes, which collectively shape quality of life and cardiovascular outcomes.
Collapse
Affiliation(s)
- Ming Hui Chen
- Department of Cardiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Boston Children's Hospital/Dana Farber Cancer Institute, Boston, MA, USA.
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA.
| | - Sonia F Epstein
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| |
Collapse
|
33
|
Ahmadsei M, Thaler K, Gasser E, Pouymayou B, Dal Bello R, Christ SM, Willmann J, Kovacs B, Balermpas P, Tanadini-Lang S, Saguner AM, Mayinger M, Andratschke N, Guckenberger M. Dosimetric analysis of 17 cardiac Sub-structures, Toxicity, and survival in ultra central lung tumor patients treated with SBRT. Clin Transl Radiat Oncol 2023; 43:100675. [PMID: 37744054 PMCID: PMC10511337 DOI: 10.1016/j.ctro.2023.100675] [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: 05/22/2023] [Revised: 08/04/2023] [Accepted: 09/07/2023] [Indexed: 09/26/2023] Open
Abstract
•Data on cardiac toxicity after SBRT for ultra-central lung tumors remains limited.•We analyzed the dose to 18 cardiac sub-structures and cardiovascular toxicity.•A SBRT regimen of 45 Gy in 8-10 fractions yields good local control and low toxicity.•The highest cardiac doses were observed in the pulmonary artery and left atrium.•Higher doses to the base of the heart seem to be associated with non-cancer deaths.
Collapse
Affiliation(s)
- Maiwand Ahmadsei
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Kai Thaler
- Department of Cardiology, University Heart Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Elena Gasser
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Bertrand Pouymayou
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Riccardo Dal Bello
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Sebastian M. Christ
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Jonas Willmann
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Boldizsar Kovacs
- Department of Cardiology, University Heart Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Panagiotis Balermpas
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Stephanie Tanadini-Lang
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Ardan M. Saguner
- Department of Cardiology, University Heart Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, Zurich University Hospital, University of Zurich, 8952 Schlieren, Switzerland
| | - Michael Mayinger
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Nicolaus Andratschke
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Matthias Guckenberger
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| |
Collapse
|
34
|
Guo M, Wang X, Xiao S, Liu A, Xu T, Huan C, Wu H, Hu Y, Zhou S, Zhu H, Pan D. Preliminary assessment of cardiotoxicity in chimeric antigen receptor T cell therapy: a systematic review and meta-analysis. Clin Exp Med 2023; 23:2041-2050. [PMID: 36930381 DOI: 10.1007/s10238-023-01042-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 03/06/2023] [Indexed: 03/18/2023]
Abstract
As a novel anticancer therapy, chimeric antigen receptor T (CAR T) cell therapy may lead to cardiotoxic reactions. However, the exact incidence remains unclear. Our study aimed to preliminarily assess the prevalence of cardiotoxicity after CAR T cell treatment using a systematic review and meta-analysis. PubMed, Embase, Web of Science, and Cochrane databases were searched for potentially relevant studies. All types of relevant clinical studies were screened and assessed for risk bias. In most instances, random-effect models were used for data analysis, and heterogeneity between studies was evaluated. Standard quality assessment tools were used to assess quality. The study was registered with PROSPERO (CRD42022304611). Eight eligible studies comprising 3567 patients, including seven observational studies and one controlled study, were identified. The incidence of cardiovascular events was 16.7% [95% confidence interval (CI) 0.138-0.200, P < 0.01)]. Arrhythmia was the most common disorder, with an incidence of 6.5% (95% CI 0.029-0.115, P < 0.01). The occurrence of cardiotoxicity was associated with cytokine release syndrome (CRS), with a prevalence of 18.7% (95% CI 0.107-0.315, P < 0.01). Moreover, such adverse reactions were more common when CRS > 2 (OR = 0.07, 95% CI 0.02-0.29, P < 0.01). The risk of cardiotoxicity was not notably higher in patients receiving CAR T cell therapy than in those receiving traditional anticancer treatment. However, sufficient attention should be paid to this. And further evidence from large-scale clinical trials are needed.
Collapse
Affiliation(s)
- Minjia Guo
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai West Road, Xuzhou, 221004, Jiangsu, China
| | - Xiaotong Wang
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai West Road, Xuzhou, 221004, Jiangsu, China
| | - Shengjue Xiao
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, 210009, China
| | - Aili Liu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai West Road, Xuzhou, 221004, Jiangsu, China
| | - Tao Xu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai West Road, Xuzhou, 221004, Jiangsu, China
| | - Chunyan Huan
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai West Road, Xuzhou, 221004, Jiangsu, China
| | - Huimin Wu
- Department of General Practice, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai West Road, Xuzhou, 221004, Jiangsu, China
| | - Yue Hu
- Department of General Practice, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai West Road, Xuzhou, 221004, Jiangsu, China
| | - Shuaishuai Zhou
- Department of General Practice, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai West Road, Xuzhou, 221004, Jiangsu, China
| | - Hong Zhu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai West Road, Xuzhou, 221004, Jiangsu, China.
| | - Defeng Pan
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai West Road, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
35
|
Glen C, Adam S, McDowell K, Waterston A, Tan YY, Petrie MC, Coats CJ, Lang NN. Cardiotoxicity of BRAF/MEK Inhibitors: A Longitudinal Study Incorporating Contemporary Definitions and Risk Scores. JACC CardioOncol 2023; 5:628-637. [PMID: 37969652 PMCID: PMC10635885 DOI: 10.1016/j.jaccao.2023.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/29/2023] [Accepted: 04/05/2023] [Indexed: 11/17/2023] Open
Abstract
Background Rapidly accelerated fibrosarcoma B-type (BRAF) and mitogen-activated extracellular signal-regulated kinase (MEK) inhibitors have revolutionized treatment for patients with BRAF-mutated melanoma. Although left ventricular systolic dysfunction associated with these therapies has been reported in clinical trials, the real-world incidence is poorly defined, as are risk factors for its development. Objectives This study sought to characterize the incidence, time course, and risk factors for cancer therapy-related cardiac dysfunction (CTRCD) in patients with melanoma receiving BRAF and MEK inhibitors. Methods Patients with melanoma treated with BRAF and MEK inhibitors at a cancer hospital network between June 1, 2017, and June 30, 2020, were included retrospectively. CTRCD was defined as mild, moderate, or severe according to International Cardio-Oncology Society (ICOS) definitions. Baseline cardiotoxicity risk stratification was performed using the Heart Failure Association/ICOS tool. Results Of the 63 patients included, 27% developed CTRCD (17% mild and 10% moderate). No patients developed severe CTRCD or symptomatic heart failure. CTRCD occurred most frequently in patients considered to be at "low" and "medium" baseline risk of cardiotoxicity (82%). The baseline left ventricular ejection fraction and global longitudinal strain were not different in patients who developed moderate CTRCD vs those who did not. Left ventricular internal diameters in diastole and systole were larger in patients who developed moderate CTRCD compared with those who did not (left ventricular internal diameter in diastole: 4.9 ± 0.6 cm vs 4.3 ± 0.6 cm; P = 0.023; left ventricular internal diameter in systole: 3.3 ± 0.4 cm vs 2.8 ± 0.5 cm; P = 0.039). Conclusions BRAF and MEK inhibitor-associated CTRCD is common. The utility of the Heart Failure Association/ICOS risk stratification tool appears limited in this group, and better risk prediction tools are needed. The long-term consequences of CTRCD, particularly mild CTRCD, warrant evaluation in larger prospective studies.
Collapse
Affiliation(s)
- Claire Glen
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Sarah Adam
- Queen Elizabeth University Hospital, National Health Service Greater Glasgow and Clyde, Glasgow, United Kingdom
| | - Kirsty McDowell
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Ashita Waterston
- Beatson West of Scotland Cancer Centre, National Health Service Greater Glasgow and Clyde, Glasgow, United Kingdom
| | - Yun Yi Tan
- Beatson West of Scotland Cancer Centre, National Health Service Greater Glasgow and Clyde, Glasgow, United Kingdom
| | - Mark C. Petrie
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Caroline J. Coats
- Queen Elizabeth University Hospital, National Health Service Greater Glasgow and Clyde, Glasgow, United Kingdom
| | - Ninian N. Lang
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
36
|
Di Lisi D, Manno G, Madaudo C, Filorizzo C, Intravaia RCM, Galassi AR, Incorvaia L, Russo A, Novo G. Chemotherapy-related cardiac dysfunction: the usefulness of myocardial work indices. THE INTERNATIONAL JOURNAL OF CARDIOVASCULAR IMAGING 2023; 39:1845-1853. [PMID: 37548845 DOI: 10.1007/s10554-023-02897-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 06/09/2023] [Indexed: 08/08/2023]
Abstract
AIMS The role of left ventricular global longitudinal strain (GLS) in the diagnosis of subclinical cardiac damage induced by anticancer drugs is now consolidated. Considering some strain disadvantages such as the dependence on the haemodynamic loading conditions, the aim of our study was to investigate the usefulness of non-invasive myocardial work indices (MWI) derived from pressure-strain analysis, in the early diagnosis of cardiotoxicity. METHODS AND RESULTS We enrolled 61 consecutive patients with breast cancer undergoing adjuvant treatment with anthracycline-containing chemotherapy followed by taxane + trastuzumab. Patients underwent a cardiological evaluation with 2D echocardiography including measurement of the left ventricular ejection fraction (LVEF) and other conventional parameters of systolic and diastolic function, GLS and MWI at baseline (T0), 3 months (T1) and 6 months (T2) after starting chemotherapy. At T1 and T2, we did not find a significant reduction in LVEF but we found a significant reduction in GLS and MWI (p value < 0.05). In addition, at T2, 31% of patients developed subclinical cardiac dysfunction defined as a relative decrease ≥ 12% of GLS from baseline. Global work index (GWI), global constructive work (GCW) and global work efficiency (GWE) decreased significantly in both patients with subclinical dysfunction and in those without subclinical dysfunction (p value < 0.05). Patients with subclinical dysfunction at T2 showed lower values of GCW at T0. CONCLUSION MWI changed significantly during chemotherapy and appeared to alter precociously compared to GLS. Therefore, a multiparametric approach including left ventricular GLS and MWI measurements should be used in the evaluation of patients undergoing cardiotoxic antineoplastic treatment.
Collapse
Affiliation(s)
- Daniela Di Lisi
- Division of Cardiology, University Hospital "Paolo Giaccone", Palermo, Italy.
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE) "G. D'Alessandro", University of Palermo, Palermo, Italy.
| | - Girolamo Manno
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE) "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Cristina Madaudo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE) "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Clarissa Filorizzo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127, Palermo, Italy
| | - Rita Cristina Myriam Intravaia
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE) "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Alfredo Ruggero Galassi
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE) "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Lorena Incorvaia
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127, Palermo, Italy
| | - Antonio Russo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127, Palermo, Italy
| | - Giuseppina Novo
- Division of Cardiology, University Hospital "Paolo Giaccone", Palermo, Italy
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE) "G. D'Alessandro", University of Palermo, Palermo, Italy
| |
Collapse
|
37
|
Abstract
Breast cancer survivors may experience significant after effects from diagnoses of breast cancer and cancer directed therapies. This review synthesizes the evidence about optimal management of the sequelae of a diagnosis of breast cancer. It describes the side effects of chemotherapy and endocrine therapy and evidence based strategies for management of such effects, with particular attention to effects of therapies with curative intent. It includes strategies to promote health and wellness among breast cancer survivors, along with data to support the use of integrative oncology strategies. In addition, this review examines models of survivorship care and ways in which digital tools may facilitate communication between clinicians and patients. The strategies outlined in this review are paramount to supporting breast cancer survivors' quality of life.
Collapse
|
38
|
Jain A, Casanova D, Padilla AV, Paniagua Bojorges A, Kotla S, Ko KA, Samanthapudi VSK, Chau K, Nguyen MTH, Wen J, Hernandez Gonzalez SL, Rodgers SP, Olmsted-Davis EA, Hamilton DJ, Reyes-Gibby C, Yeung SCJ, Cooke JP, Herrmann J, Chini EN, Xu X, Yusuf SW, Yoshimoto M, Lorenzi PL, Hobbs B, Krishnan S, Koutroumpakis E, Palaskas NL, Wang G, Deswal A, Lin SH, Abe JI, Le NT. Premature senescence and cardiovascular disease following cancer treatments: mechanistic insights. Front Cardiovasc Med 2023; 10:1212174. [PMID: 37781317 PMCID: PMC10540075 DOI: 10.3389/fcvm.2023.1212174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 08/03/2023] [Indexed: 10/03/2023] Open
Abstract
Cardiovascular disease (CVD) is a leading cause of morbidity and mortality, especially among the aging population. The "response-to-injury" model proposed by Dr. Russell Ross in 1999 emphasizes inflammation as a critical factor in atherosclerosis development, with atherosclerotic plaques forming due to endothelial cell (EC) injury, followed by myeloid cell adhesion and invasion into the blood vessel walls. Recent evidence indicates that cancer and its treatments can lead to long-term complications, including CVD. Cellular senescence, a hallmark of aging, is implicated in CVD pathogenesis, particularly in cancer survivors. However, the precise mechanisms linking premature senescence to CVD in cancer survivors remain poorly understood. This article aims to provide mechanistic insights into this association and propose future directions to better comprehend this complex interplay.
Collapse
Affiliation(s)
- Ashita Jain
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Diego Casanova
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | | | | - Sivareddy Kotla
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Kyung Ae Ko
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | | - Khanh Chau
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Minh T. H. Nguyen
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Jake Wen
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | | - Shaefali P. Rodgers
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | | | - Dale J. Hamilton
- Department of Medicine, Center for Bioenergetics, Houston Methodist Research Institute, Houston, TX, United States
| | - Cielito Reyes-Gibby
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Sai-Ching J. Yeung
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - John P. Cooke
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Joerg Herrmann
- Cardio Oncology Clinic, Division of Preventive Cardiology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Eduardo N. Chini
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Xiaolei Xu
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Syed Wamique Yusuf
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Momoko Yoshimoto
- Center for Stem Cell & Regenerative Medicine, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Philip L. Lorenzi
- Department of Bioinformatics and Computational Biology, Division of VP Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Brain Hobbs
- Department of Population Health, The University of Texas at Austin, Austin, TX, United States
| | - Sunil Krishnan
- Department of Neurosurgery, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Efstratios Koutroumpakis
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nicolas L. Palaskas
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Guangyu Wang
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Anita Deswal
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Steven H. Lin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jun-ichi Abe
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nhat-Tu Le
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| |
Collapse
|
39
|
Santangelo G, Moscardelli S, Barbieri L, Faggiano A, Carugo S, Faggiano P. Aortic Valve Stenosis and Cancer: Problems of Management. J Clin Med 2023; 12:5804. [PMID: 37762745 PMCID: PMC10532214 DOI: 10.3390/jcm12185804] [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: 08/02/2023] [Revised: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
Aortic valve stenosis and malignancy frequently coexist and share the same risk factors as atherosclerotic disease. Data reporting the prognosis of patients with severe aortic stenosis and cancer are limited. Tailoring the correct and optimal care for cancer patients with severe aortic stenosis is complex. Cancer patients may be further disadvantaged by aortic stenosis if it interferes with their treatment by increasing the risk associated with oncologic surgery and compounding the risks associated with cardiotoxicity and heart failure (HF). Surgical valve replacement, transcatheter valve implantation, balloon valvuloplasty, and medical therapy are possible treatments for aortic valve stenosis, but when malignancy is present, the choice between these options must take into account the stage of cancer and associated treatment, expected outcome, and comorbidities. Physical examination and Doppler echocardiography are critical in the diagnosis and evaluation of aortic stenosis. The current review considers the available data on the association between aortic stenosis and cancer and the therapeutic options.
Collapse
Affiliation(s)
- Gloria Santangelo
- Department of Cardio-Thoracic-Vascular Area, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, 20154 Milan, Italy; (G.S.); (L.B.); (A.F.)
| | - Silvia Moscardelli
- Division of Cardiology, Department of Health Sciences, San Paolo Hospital, University of Milan, 20133 Milan, Italy;
| | - Lucia Barbieri
- Department of Cardio-Thoracic-Vascular Area, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, 20154 Milan, Italy; (G.S.); (L.B.); (A.F.)
| | - Andrea Faggiano
- Department of Cardio-Thoracic-Vascular Area, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, 20154 Milan, Italy; (G.S.); (L.B.); (A.F.)
- Department of Clinical Sciences and Community Health, University of Milan, 20133 Milan, Italy
| | - Stefano Carugo
- Department of Cardio-Thoracic-Vascular Area, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, 20154 Milan, Italy; (G.S.); (L.B.); (A.F.)
- Department of Clinical Sciences and Community Health, University of Milan, 20133 Milan, Italy
| | - Pompilio Faggiano
- Fondazione Poliambulanza, Cardiothoracic Department Unit, 25100 Brescia, Italy
| |
Collapse
|
40
|
Stahl M, Giblin G, Liu Y, Winer ES, Garcia JS, Chen E, Wadleigh M, Ling K, Lindsley RC, Shimony S, Copson K, Charles A, DeAngelo DJ, Stone RM, Nohria A, Luskin MR. Incidence and predictors of anthracycline-related left ventricular dysfunction in acute myeloid leukemia. Leuk Res 2023; 132:107351. [PMID: 37451200 DOI: 10.1016/j.leukres.2023.107351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 06/01/2023] [Accepted: 06/27/2023] [Indexed: 07/18/2023]
Abstract
INTRODUCTION Anthracycline-related left ventricular dysfunction (ARLVD) is a concern in patients with acute myeloid leukemia (AML) undergoing anthracyclinecontaining induction chemotherapy. However, the incidence of ARLVD in the modern era of routine pretreatment left ventricular ejection fraction (LVEF) echocardiographic assessment, as well as the clinical and genetic predictors of ARLVD are not well understood. METHODS Consecutive adult patients with AML receiving anthracycline-containing induction chemotherapy at the Dana-Farber Cancer Institute from 2014 to 2022 were studied. Inclusion criteria included availability of a pre and post chemotherapy echocardiogram to assess the LVEF, pre-treatment LVEF > 50 %, as well as comprehensive diagnostic next generation sequencing assessing for the presence of myeloid mutations. The primary endpoint was the incidence of ARLVD defined as LVEF < 50 % post-induction. RESULTS Out of 419 patients meeting inclusion criteria, 34 (8%) patients developed ARLVD. Among the 122/419 patients who did not undergo planned allogeneic stem cell transplantation (allo-SCT), ARLVD was the deciding factor for ineligibility in 4 patients (1%). Baseline cardiovascular comorbidities (hypertension, diabetes mellitus, hyperlipidemia, smoking and coronary artery disease) and cumulative anthracycline dose were not predictive of post-induction ARLVD. However, the presence of a JAK2 mutation (but not other myeloid mutations) was associated with an increased risk of ARLVD in multivariable analysis (OR 8.34, 95 % CI 1.55-39.3, p = 0.007). DISCUSSION In a group of AML patients with normal LVEF prior to anthracycline-containing induction chemotherapy, ARLVD was infrequent and did not commonly preclude post-remission allo-SCT consolidation. Genetic predictors of ARLVD require further investigation in a larger patient cohort.
Collapse
MESH Headings
- Adult
- Humans
- Anthracyclines/adverse effects
- Stroke Volume
- Incidence
- Ventricular Function, Left
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/chemically induced
- Antibiotics, Antineoplastic/therapeutic use
- Ventricular Dysfunction, Left/chemically induced
- Ventricular Dysfunction, Left/drug therapy
Collapse
Affiliation(s)
- Maximilian Stahl
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Gerard Giblin
- Department of Cardiology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Yiwen Liu
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Eric S Winer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Jacqueline S Garcia
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Evan Chen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Martha Wadleigh
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Kelly Ling
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - R Coleman Lindsley
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Shai Shimony
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States; Rabin Medical Center and Faculty of Medicine, Tel Aviv University, Israel
| | - Kevin Copson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Anne Charles
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Daniel J DeAngelo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Richard M Stone
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Anju Nohria
- Department of Cardiology, Brigham and Women's Hospital, Boston, MA, United States
| | - Marlise R Luskin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States.
| |
Collapse
|
41
|
Petit C, Escande A, Sarrade T, Vaugier L, Kirova Y, Tallet A. Radiation therapy in the thoracic region: Radio-induced cardiovascular disease, cardiac delineation and sparing, cardiac dose constraints, and cardiac implantable electronic devices. Cancer Radiother 2023; 27:588-598. [PMID: 37648559 DOI: 10.1016/j.canrad.2023.06.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 09/01/2023]
Abstract
Radiation therapy in the thoracic region may deliver incidental ionizing radiation to the surrounding healthy structures, including the heart. Radio-induced heart toxicity has long been a concern in breast cancer and Hodgkin's lymphoma and was deemed a long-term event. However, recent data highlight the need to limit the dose to the heart in less favorable thoracic cancers too, such as lung and esophageal cancers in which incidental irradiation led to increased mortality. This article will summarize available cardiac dose constraints in various clinical settings and the types of radio-induced cardiovascular diseases encountered as well as delineation of cardiac subheadings and management of cardiac devices. Although still not completely deciphered, heart dose constraints remain intensively investigated and the mean dose to the heart is no longer the only dosimetric parameter to consider since the left anterior descending artery as well as the left ventricle should also be part of dosimetry constraints.
Collapse
Affiliation(s)
- C Petit
- Radiation Oncology Department, institut Paoli-Calmettes, 232, boulevard Sainte-Marguerite, 13273 Marseille cedex 09, France
| | - A Escande
- Service de radiothérapie, centre Léonard-de-Vinci, Dechy, France; UMR 9189, laboratoire Cristal, université de Lille, Villeneuve-d'Ascq, France
| | - T Sarrade
- Department of Radiation Oncology, hôpital Tenon, Sorbonne université, 75020 Paris, France
| | - L Vaugier
- Department of Radiation Oncology, institut de cancérologie de l'Ouest, Saint-Herblain, France
| | - Y Kirova
- Department of Radiation Oncology, institut Curie, Paris, France
| | - A Tallet
- Radiation Oncology Department, institut Paoli-Calmettes, 232, boulevard Sainte-Marguerite, 13273 Marseille cedex 09, France; UMR 1068, CRCM Inserm, Marseille, France.
| |
Collapse
|
42
|
Fan R, Wang Y, Zhang J, An X, Liu S, Bai J, Li J, Lin Q, Xie Y, Liao J, Xia Y. Hyperhomocysteinaemia Promotes Doxorubicin-Induced Cardiotoxicity in Mice. Pharmaceuticals (Basel) 2023; 16:1212. [PMID: 37765020 PMCID: PMC10534320 DOI: 10.3390/ph16091212] [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: 07/19/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023] Open
Abstract
Doxorubicin, a widely used chemotherapeutic drug in clinical oncology, causes a series of cardiac side effects referred to as doxorubicin-induced cardiotoxicity. Hyperhomocysteinaemia is an independent risk factor for multiple cardiovascular diseases. However, whether hyperhomocysteinaemia contributes to doxorubicin-induced cardiotoxicity is currently unknown. In this study, we explored the pathogenic effects of hyperhomocysteinaemia induced by dietary methionine supplementation (2% wt/wt in rodent chow) in a mouse model of doxorubicin-induced cardiotoxicity. Our data showed that methionine supplementation doubled serum homocysteine levels, inducing mild hyperhomocysteinaemia. Doxorubicin at a cumulative dosage of 25 mg/kg body weight led to significant weight loss and severe cardiac dysfunction, which were further exacerbated by methionine-induced mild hyperhomocysteinaemia. Doxorubicin-induced cardiac atrophy, cytoplasmic vacuolisation, myofibrillar disarray and loss, as well as cardiac fibrosis, were also exacerbated by methionine-induced mild hyperhomocysteinaemia. Additional folic acid supplementation (0.006% wt/wt) prevented methionine-induced hyperhomocysteinaemia and inhibited hyperhomocysteinaemia-aggravated cardiac dysfunction and cardiomyopathy. In particular, hyperhomocysteinaemia increased both serum and cardiac oxidative stress, which could all be inhibited by folic acid supplementation. Therefore, we demonstrated for the first time that hyperhomocysteinaemia could exacerbate doxorubicin-induced cardiotoxicity in mice, and the pathogenic effects of hyperhomocysteinaemia might at least partially correlate with increased oxidative stress and could be prevented by folic acid supplementation. Our study provides preliminary experimental evidence for the assessment of hyperhomocysteinaemia as a potential risk factor for chemotherapy-induced cardiotoxicity in cancer patients.
Collapse
Affiliation(s)
- Rui Fan
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Yao Wang
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Jinjin Zhang
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xiangbo An
- Department of Interventional Therapy, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Shuang Liu
- College of Basic Medical Sciences, Dalian Medical University, Dalian 116004, China
| | - Jie Bai
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian 116004, China
| | - Jiatian Li
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Qiuyue Lin
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Yunpeng Xie
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Jiawei Liao
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Yunlong Xia
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| |
Collapse
|
43
|
Mikail N, Chequer R, Imperiale A, Meisel A, Bengs S, Portmann A, Gimelli A, Buechel RR, Gebhard C, Rossi A. Tales from the future-nuclear cardio-oncology, from prediction to diagnosis and monitoring. Eur Heart J Cardiovasc Imaging 2023; 24:1129-1145. [PMID: 37467476 PMCID: PMC10501471 DOI: 10.1093/ehjci/jead168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 07/07/2023] [Indexed: 07/21/2023] Open
Abstract
Cancer and cardiovascular diseases (CVD) often share common risk factors, and patients with CVD who develop cancer are at high risk of experiencing major adverse cardiovascular events. Additionally, cancer treatment can induce short- and long-term adverse cardiovascular events. Given the improvement in oncological patients' prognosis, the burden in this vulnerable population is slowly shifting towards increased cardiovascular mortality. Consequently, the field of cardio-oncology is steadily expanding, prompting the need for new markers to stratify and monitor the cardiovascular risk in oncological patients before, during, and after the completion of treatment. Advanced non-invasive cardiac imaging has raised great interest in the early detection of CVD and cardiotoxicity in oncological patients. Nuclear medicine has long been a pivotal exam to robustly assess and monitor the cardiac function of patients undergoing potentially cardiotoxic chemotherapies. In addition, recent radiotracers have shown great interest in the early detection of cancer-treatment-related cardiotoxicity. In this review, we summarize the current and emerging nuclear cardiology tools that can help identify cardiotoxicity and assess the cardiovascular risk in patients undergoing cancer treatments and discuss the specific role of nuclear cardiology alongside other non-invasive imaging techniques.
Collapse
Affiliation(s)
- Nidaa Mikail
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Renata Chequer
- Department of Nuclear Medicine, Bichat University Hospital, AP-HP, University Diderot, 75018 Paris, France
| | - Alessio Imperiale
- Nuclear Medicine, Institut de Cancérologie de Strasbourg Europe (ICANS), University Hospitals of Strasbourg, 67093 Strasbourg, France
- Molecular Imaging-DRHIM, IPHC, UMR 7178, CNRS/Unistra, 67093 Strasbourg, France
| | - Alexander Meisel
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Kantonsspital Glarus, Burgstrasse 99, 8750 Glarus, Switzerland
| | - Susan Bengs
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Angela Portmann
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Alessia Gimelli
- Imaging Department, Fondazione CNR/Regione Toscana Gabriele Monasterio, Via G. Moruzzi 1, 56124 Pisa, Italy
| | - Ronny R Buechel
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Cathérine Gebhard
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
- Department of Cardiology, University Hospital Inselspital Bern, Freiburgstrasse 18, 3010 Bern, Switzerland
| | - Alexia Rossi
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| |
Collapse
|
44
|
Chen R, Niu M, Hu X, He Y. Targeting mitochondrial dynamics proteins for the treatment of doxorubicin-induced cardiotoxicity. Front Mol Biosci 2023; 10:1241225. [PMID: 37602332 PMCID: PMC10437218 DOI: 10.3389/fmolb.2023.1241225] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 07/24/2023] [Indexed: 08/22/2023] Open
Abstract
Doxorubicin (DOX) is an extensively used chemotherapeutic agent that can cause severe and frequent cardiotoxicity, which limits its clinical application. Although there have been extensive researches on the cardiotoxicity caused by DOX, there is still a lack of effective treatment. It is necessary to understand the molecular mechanism of DOX-induced cardiotoxicity and search for new therapeutic targets which do not sacrifice their anticancer effects. Mitochondria are considered to be the main target of cardiotoxicity caused by DOX. The imbalance of mitochondrial dynamics characterized by increased mitochondrial fission and inhibited mitochondrial fusion is often reported in DOX-induced cardiotoxicity, which can result in excessive ROS production, energy metabolism disorders, cell apoptosis, and various other problems. Also, mitochondrial dynamics disorder is related to tumorigenesis. Surprisingly, recent studies show that targeting mitochondrial dynamics proteins such as DRP1 and MFN2 can not only defend against DOX-induced cardiotoxicity but also enhance or not impair the anticancer effect. Herein, we summarize mitochondrial dynamics disorder in DOX-induced cardiac injury. Furthermore, we provide an overview of current pharmacological and non-pharmacological interventions targeting proteins involved in mitochondrial dynamics to alleviate cardiac damage caused by DOX.
Collapse
Affiliation(s)
- Rui Chen
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Mengwen Niu
- Department of Rheumatology and Immunology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xin Hu
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yuquan He
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
45
|
Sun S, Qin J, Liao W, Gao X, Shang Z, Luo D, Xiong S. Mitochondrial Dysfunction in Cardiotoxicity Induced by BCR-ABL1 Tyrosine Kinase Inhibitors -Underlying Mechanisms, Detection, Potential Therapies. Cardiovasc Toxicol 2023; 23:233-254. [PMID: 37479951 DOI: 10.1007/s12012-023-09800-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 07/08/2023] [Indexed: 07/23/2023]
Abstract
The advent of BCR-ABL tyrosine kinase inhibitors (TKIs) targeted therapy revolutionized the treatment of chronic myeloid leukemia (CML) patients. Mitochondria are the key organelles for the maintenance of myocardial tissue homeostasis. However, cardiotoxicity associated with BCR-ABL1 TKIs can directly or indirectly cause mitochondrial damage and dysfunction, playing a pivotal role in cardiomyocytes homeostatic system and putting the cancer survivors at higher risk. In this review, we summarize the cardiotoxicity caused by BCR-ABL1 TKIs and the underlying mechanisms, which contribute dominantly to the damage of mitochondrial structure and dysfunction: endoplasmic reticulum (ER) stress, mitochondrial stress, damage of myocardial cell mitochondrial respiratory chain, increased production of mitochondrial reactive oxygen species (ROS), and other kinases and other potential mechanisms of cardiotoxicity induced by BCR-ABL1 TKIs. Furthermore, detection and management of BCR-ABL1 TKIs will promote our rational use, and cardioprotection strategies based on mitochondria will improve our understanding of the cardiotoxicity from a mitochondrial perspective. Ultimately, we hope shed light on clinical decision-making. By integrate and learn from both research and practice, we will endeavor to minimize the mitochondria-mediated cardiotoxicity and reduce the adverse sequelae associated with BCR-ABL1 TKIs.
Collapse
Affiliation(s)
- Sheng Sun
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Medical Oncology, Hospital of Chengdu University of Traditioanal Chinese Medicine, Chengdu, 610075, Sichuan Province, China
| | - Jiqiu Qin
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wenhao Liao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiang Gao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhoubiao Shang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dehua Luo
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shaoquan Xiong
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
- Department of Medical Oncology, Hospital of Chengdu University of Traditioanal Chinese Medicine, Chengdu, 610075, Sichuan Province, China.
| |
Collapse
|
46
|
Bashir Z, Chen EW, Tori K, Ghosalkar D, Aurigemma GP, Dickey JB, Haines P. Insight into different phenotypic presentations of heart failure with preserved ejection fraction. Prog Cardiovasc Dis 2023; 79:80-88. [PMID: 37442358 DOI: 10.1016/j.pcad.2023.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 07/10/2023] [Indexed: 07/15/2023]
Abstract
Heart failure (HF) with preserved ejection fraction (HFpEF) accounts for half of all HF diagnoses, and its prevalence is increasing at an alarming rate. Lately, it has been recognized as a clinical syndrome due to diverse underlying etiology and pathophysiological mechanisms. The classic echocardiographic features of HFpEF have been well described as preserved ejection fraction (≥50%), left ventricular hypertrophy, and left atrial enlargement. However, echocardiography can play a key role in identifying the principal underlying mechanism responsible for HFpEF in the individual patient. The recognition of different phenotypic presentations of HFpEF (infiltrative, metabolic, genetic, and inflammatory) can assist the clinician in tailoring the appropriate management, and offer prognostic information. The goal of this review is to highlight several key phenotypes of HFpEF and illustrate the classic clinical scenario and echocardiographic features of each phenotype with real patient cases.
Collapse
Affiliation(s)
- Zubair Bashir
- Department of Cardiology, Warren Alpert Medical School of Brown University, Providence, RI, USA.
| | - Edward W Chen
- Department of Cardiology, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | | | - Dhairyasheel Ghosalkar
- Division of Cardiovascular Medicine, Department of Medicine, Stony Brook University Hospital, NY, USA
| | - Gerard P Aurigemma
- Division of Cardiovascular Medicine, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - John B Dickey
- Division of Cardiovascular Medicine, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Philip Haines
- Department of Cardiology, Warren Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
47
|
Al Saikhan L, Park C, Tillin T, Jones S, Mayet J, Chaturvedi N, Hughes A. Does 3D-speckle tracking echocardiography improve prediction of major cardiovascular events in a multi-ethnic general population? A Southall and Brent Revisited (SABRE) cohort study. PLoS One 2023; 18:e0287173. [PMID: 37368914 PMCID: PMC10298788 DOI: 10.1371/journal.pone.0287173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
3D-speckle tracking echocardiography(3D-STE) allows simultaneous assessment of ejection fraction(EF) and multidirectional strains, but its prognostic utility in the general population is unknown. We investigated if 3D-STE strains predicted a composite of major cardiac endpoints(MACE) beyond cardiovascular risk factors(CVDRF), and whether they were superior to 3D-EF. 529 participants in SABRE, a UK-based tri-ethnic general population cohort (69±6y; 76.6% male) with acceptable 3D-STE imaging were studied. Associations between 3D-EF or multidirectional myocardial strains and MACE(coronary heart disease(fatal/non-fatal), heart failure hospitalization, new-onset arrhythmia and cardiovascular mortality) were determined using Cox regression including adjustment for CVDRF and 2D-EF. Whether 3D-EF, global longitudinal strain(3D-GLS) and principle tangential strain(3D-PTS/3D-strain) improved cardiovascular risk stratification over CVDRF was investigated using a likelihood ratio test on a series of nested Cox proportional hazards models and Harrell's C statistics. During follow-up(median, 12y), there were 92 events. 3D-EF, 3D-GLS and 3D-PTS and 3D-RS were associated with MACE in unadjusted and models adjusted for CVDRF but not CVDRF+2D-EF. Compared to 3D-EF, both 3D-GLS and 3D-PTS slightly improved the predictive value over CVDRF for MACE, but the improvement was modest(C statistic increased from 0.698(0.647, 0.749) to 0.715(0.663, 0.766) comparing CVDRF with CVDRF +3D-GLS). 3D-STE-derived LV myocardial strains predicted MACE in a multi-ethnic general population sample of elderly individuals from the UK; however the added prognostic value of 3D-STE myocardial strains was small.
Collapse
Affiliation(s)
- Lamia Al Saikhan
- Department of Cardiac Technology, College of Applied Medial Sciences, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Chloe Park
- Department of Population Science & Experimental Medicine, UCL Institute of Cardiovascular Science, MRC Unit for Lifelong Health and Ageing, University College London, London, United Kingdom
| | - Therese Tillin
- Department of Population Science & Experimental Medicine, UCL Institute of Cardiovascular Science, MRC Unit for Lifelong Health and Ageing, University College London, London, United Kingdom
| | - Siana Jones
- Department of Population Science & Experimental Medicine, UCL Institute of Cardiovascular Science, MRC Unit for Lifelong Health and Ageing, University College London, London, United Kingdom
| | - Jamil Mayet
- NIHR Imperial Biomedical Research Centre, Imperial College London and Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, United Kingdom
| | - Nish Chaturvedi
- Department of Population Science & Experimental Medicine, UCL Institute of Cardiovascular Science, MRC Unit for Lifelong Health and Ageing, University College London, London, United Kingdom
| | - Alun Hughes
- Department of Population Science & Experimental Medicine, UCL Institute of Cardiovascular Science, MRC Unit for Lifelong Health and Ageing, University College London, London, United Kingdom
| |
Collapse
|
48
|
Li X, Shen D, Zhu Z, Lyu D, He C, Sun Y, Li J, Lu Q, Wang G. Dual roles of demethylation in cancer treatment and cardio-function recovery. Redox Biol 2023; 64:102785. [PMID: 37343447 PMCID: PMC10363477 DOI: 10.1016/j.redox.2023.102785] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/07/2023] [Accepted: 06/13/2023] [Indexed: 06/23/2023] Open
Abstract
There are no effective therapeutic targets or strategies that simultaneously inhibit tumour growth and promote cardiac function recovery. Here, we analyzed targets for cancer treatments and cardiac repair, with demethylation emerging as a common factor in these candidate lists. As DNA methyltransferase 1 (DNMT1) majorly responds to methylation, a natural compound library is screened, identifying dioscin as a novel agent targeted at DNMT1, widely used for heart diseases. Dioscin was found to reduce DNMT activities and inhibits growth in breast cancer cells. Combined with analyses of RNA-seq and MeDIP-seq, the promoters of antioxidant genes were demethylated after dioscin, recruiting NRF2 and elevating their expression. In Nrf2 knockout mice, the cardiac protection role of dioscin was blocked by Nrf2-loss. Furthermore, in tumour-bearing mice with hypertrophy, dioscin was observed to inhibit tumour growth and alleviate cardiac injury simultaneously. This study is the first to identify dioscin as a novel demethylation agent with dual functions of anti-cancer and cardio-protection.
Collapse
Affiliation(s)
- Xinuo Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China; Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Dehong Shen
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Zheying Zhu
- School of Pharmacy, The University of Nottingham, Nottingham, NG7 2RD, UK
| | - Dayin Lyu
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Chang He
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Yuan Sun
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China; Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Jinran Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China; Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Qiulun Lu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China; Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China.
| | - Guangji Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China; Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
49
|
Peng K, Zeng C, Gao Y, Liu B, Li L, Xu K, Yin Y, Qiu Y, Zhang M, Ma F, Wang Z. Overexpressed SIRT6 ameliorates doxorubicin-induced cardiotoxicity and potentiates the therapeutic efficacy through metabolic remodeling. Acta Pharm Sin B 2023; 13:2680-2700. [PMID: 37425037 PMCID: PMC10326298 DOI: 10.1016/j.apsb.2023.03.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 02/12/2023] [Accepted: 03/02/2023] [Indexed: 07/11/2023] Open
Abstract
Since the utilization of anthracyclines in cancer therapy, severe cardiotoxicity has become a major obstacle. The major challenge in treating cancer patients with anthracyclines is minimizing cardiotoxicity without compromising antitumor efficacy. Herein, histone deacetylase SIRT6 expression was reduced in plasma of patients treated with anthracyclines-based chemotherapy regimens. Furthermore, overexpression of SIRT6 alleviated doxorubicin-induced cytotoxicity in cardiomyocytes, and potentiated cytotoxicity of doxorubicin in multiple cancer cell lines. Moreover, SIRT6 overexpression ameliorated doxorubicin-induced cardiotoxicity and potentiated antitumor efficacy of doxorubicin in mice, suggesting that SIRT6 overexpression could be an adjunctive therapeutic strategy during doxorubicin treatment. Mechanistically, doxorubicin-impaired mitochondria led to decreased mitochondrial respiration and ATP production. And SIRT6 enhanced mitochondrial biogenesis and mitophagy by deacetylating and inhibiting Sgk1. Thus, SIRT6 overexpression coordinated metabolic remodeling from glycolysis to mitochondrial respiration during doxorubicin treatment, which was more conducive to cardiomyocyte metabolism, thus protecting cardiomyocytes but not cancer cells against doxorubicin-induced energy deficiency. In addition, ellagic acid, a natural compound that activates SIRT6, alleviated doxorubicin-induced cardiotoxicity and enhanced doxorubicin-mediated tumor regression in tumor-bearing mice. These findings provide a preclinical rationale for preventing cardiotoxicity by activating SIRT6 in cancer patients undergoing chemotherapy, but also advancing the understanding of the crucial role of SIRT6 in mitochondrial homeostasis.
Collapse
Affiliation(s)
- Kezheng Peng
- The Ministry of Education Key Laboratory of Protein Science, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Chenye Zeng
- The Ministry of Education Key Laboratory of Protein Science, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Yuqi Gao
- The Ministry of Education Key Laboratory of Protein Science, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Binliang Liu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Liyuan Li
- The Ministry of Education Key Laboratory of Protein Science, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Kang Xu
- The Ministry of Education Key Laboratory of Protein Science, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Yuemiao Yin
- The Ministry of Education Key Laboratory of Protein Science, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Ying Qiu
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Mingkui Zhang
- Department of Cardiac Surgery, First Hospital of Tsinghua University, Beijing 100016, China
| | - Fei Ma
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Zhao Wang
- The Ministry of Education Key Laboratory of Protein Science, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
50
|
Suto H, Suto M, Inui Y, Okamura A. Late-onset doxorubicin-induced congestive heart failure in an elderly cancer survivor: A case report. Front Cardiovasc Med 2023; 10:1124276. [PMID: 37180802 PMCID: PMC10166870 DOI: 10.3389/fcvm.2023.1124276] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 04/11/2023] [Indexed: 05/16/2023] Open
Abstract
Background Recently, the survival rate of patients with cancer has improved annually due to advancements in cancer diagnosis and treatment technologies. Meanwhile, late-onset complications associated with cancer treatment significantly affect survival and quality of life. However, different from pediatric cancer survivors, there is no unified view on the follow-up of late complications in elderly cancer survivors. We reported a case of congestive heart failure as a late-onset complication of doxorubicin (DXR) in an elderly cancer survivor. Case report The patient is an 80-year-old woman with hypertension and chronic renal failure. She received six cycles of chemotherapy for Hodgkin's lymphoma that started in January 201X-2. The total dose of DXR was 300 mg/m2, and a transthoracic echocardiogram (TTE) performed in October 201X-2, showed good left ventricular wall motion (LVWM). In April 201X, she suddenly developed dyspnea. Upon arrival at the hospital, a physical examination revealed orthopnea, tachycardia, and leg edema. A chest radiograph showed cardiac enlargement and pleural effusion. A TTE showed diffusely reduced LVWM and a left ventricular ejection fraction in the 20% range. After close examination, the patient was diagnosed with congestive heart failure due to late-onset DXR-induced cardiomyopathy. Conclusion Late-onset DXR-induced cardiotoxicity is considered high-risk from 250 mg/m2 or higher. Elderly cancer survivors are at higher risk of cardiotoxicity than non-elderly cancer survivors and may require closer follow-up.
Collapse
Affiliation(s)
- Hirotaka Suto
- Department of Medical Oncology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
- Department of Medical Oncology/Hematology, Kakogawa Central City Hospital, Hyogo, Japan
| | - Makiko Suto
- Department of Cardiovascular Medicine, Takarazuka City Hospital, Hyogo, Japan
| | - Yumiko Inui
- Department of Medical Oncology/Hematology, Kakogawa Central City Hospital, Hyogo, Japan
| | - Atsuo Okamura
- Department of Medical Oncology/Hematology, Kakogawa Central City Hospital, Hyogo, Japan
| |
Collapse
|