1
|
Berlin E, Ko K, Ma L, Messing I, Hollawell C, Smith AM, Taunk NK, Narayan V, Upshaw JN, Clark AS, Shah PD, Knollman H, Bhattacharya S, Koropeckyj-Cox D, Wang J, Yegya-Raman N, Han IS, Lefebvre B, Li T, Wilcox NS, Jung W, Chen J, Freedman GM, Ky B. Cardiac Effects of Modern Breast Radiation Therapy in Patients Receiving Systemic Cancer Therapy. JACC CardioOncol 2025; 7:219-230. [PMID: 40044512 PMCID: PMC12046838 DOI: 10.1016/j.jaccao.2025.01.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 01/15/2025] [Accepted: 01/15/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Radiation therapy (RT) improves breast cancer outcomes, but cardiac morbidity remains a concern. OBJECTIVES This study sought to evaluate changes in cardiac function after RT and the relationship between cardiac dose metrics and echocardiography-derived measures of function. METHODS In a longitudinal cohort study of women with breast cancer, radiation cardiac dose metrics and core lab quantitated echocardiographic measures of cardiac function were evaluated. Dose metrics included the whole heart, left ventricle, right ventricle, and left anterior descending artery (LAD). Echocardiographic measures included left ventricular ejection fraction (LVEF), longitudinal strain, circumferential strain, E/e' (ratio of early diastolic mitral inflow velocity to early diastolic mitral annular tissue velocity), Ea/Es (ventricular arterial coupling; ratio of effective arterial elastance to end systolic elastance), and right ventricular fractional area change. The mean change in echocardiographic measures over time and the association between cardiac dose metrics and echocardiographic measures were estimated by repeated-measures multivariable linear regression via generalized estimating equations. RESULTS The cohort included 303 participants (median age 52 years, 33.3% African American) who received adjuvant RT (2010-2019) with a median mean heart dose of 1.19 Gy (Q1-Q3: 0.75-2.61 Gy), were followed over a median of 5.1 years (Q1-Q3: 3.2-7.1 years). Across all participants, there was a modest increase in LVEF (52.1% pre-RT to 54.3% at 5 years; P < 0.001) but a worsening in sensitive measures of function, such as circumferential strain (-23.7% pre-RT to -21.0% at 5 years; P = 0.003). Among left-sided/bilateral breast cancer participants, changes in cardiac function were observed across all parameters (P < 0.05). The maximum LAD dose was associated with a modest worsening in LVEF, longitudinal strain, circumferential strain, and E/e'. CONCLUSIONS Over a median of 5.1 years, modest changes in cardiac function were observed with RT. Maximum LAD dose was associated with a worsening in systolic and diastolic function parameters.
Collapse
Affiliation(s)
- Eva Berlin
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kyunga Ko
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lin Ma
- Department of Radiation Oncology, Mayo Clinic Comprehensive Cancer Center, Rochester, Minnesota, USA
| | - Ian Messing
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Casey Hollawell
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Amanda M Smith
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Neil K Taunk
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Vivek Narayan
- Division of Hematology and Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jenica N Upshaw
- Division of Cardiology, Department of Medicine, Tufts Medical Center, Boston, Massachusetts, USA
| | - Amy S Clark
- Division of Hematology and Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Payal D Shah
- Division of Hematology and Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Hayley Knollman
- Division of Hematology and Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Saveri Bhattacharya
- Division of Hematology and Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Daniel Koropeckyj-Cox
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jessica Wang
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nikhil Yegya-Raman
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ivy S Han
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Benedicte Lefebvre
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Tang Li
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nicholas S Wilcox
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Wonyoung Jung
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jinbo Chen
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Gary M Freedman
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Bonnie Ky
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
2
|
Fazzini L, Campana N, Cossu S, Deidda M, Madaudo C, Quagliariello V, Maurea N, Di Lisi D, Novo G, Zito C, Cadeddu Dessalvi C. Genetic Background in Patients with Cancer Therapy-Induced Cardiomyopathy. J Clin Med 2025; 14:1286. [PMID: 40004816 PMCID: PMC11856774 DOI: 10.3390/jcm14041286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 02/06/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Emerging evidence indicates that specific genetic variants are associated with an increased risk of toxicity from anticancer treatments and cancer-related cardiovascular complications. These genetic factors influence drug metabolism, efficacy, and susceptibility to adverse effects. For cancer patients, the genetic background can have two major cardiovascular implications, namely therapy-related cardiotoxicity and cancer-related cardiovascular complications. Baseline risk stratification is essential to identify higher-risk individuals and ensure they receive appropriate preventive and therapeutic interventions and more frequent follow-up. Current guidelines recommend stratification based on cardiovascular risk factors, but these factors alone cannot accurately define individual risk. Genetic background has been shown to enhance risk stratification. Beyond rare genetic variants, recent genome-wide association studies have identified single nucleotide polymorphisms implicated in cancer therapy toxicity. Despite their current limitations, polygenic risk scores are expected to play a significant role in risk stratification. This review aims to summarize the current evidence on the role of the genetic background of patients with cancer treated with potentially cardiotoxic drugs who develop cardiotoxicity, aiming to provide insights to refine risk stratification further and tailor the management of these patients.
Collapse
Affiliation(s)
- Luca Fazzini
- Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy; (L.F.); (N.C.); (S.C.); (M.D.)
| | - Nicola Campana
- Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy; (L.F.); (N.C.); (S.C.); (M.D.)
| | - Stefano Cossu
- Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy; (L.F.); (N.C.); (S.C.); (M.D.)
| | - Martino Deidda
- Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy; (L.F.); (N.C.); (S.C.); (M.D.)
| | - Cristina Madaudo
- Cardiology Unit, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University Hospital ‘Paolo Giaccone’, University of Palermo, 90133 Palermo, Italy (D.D.L.); (G.N.)
| | - Vincenzo Quagliariello
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (V.Q.); (N.M.)
| | - Nicola Maurea
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (V.Q.); (N.M.)
| | - Daniela Di Lisi
- Cardiology Unit, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University Hospital ‘Paolo Giaccone’, University of Palermo, 90133 Palermo, Italy (D.D.L.); (G.N.)
| | - Giuseppina Novo
- Cardiology Unit, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University Hospital ‘Paolo Giaccone’, University of Palermo, 90133 Palermo, Italy (D.D.L.); (G.N.)
| | - Concetta Zito
- Cardiology Unit, Department of Clinical and Experimental Medicine, University Hospital “G. Martino”, University of Messina, 98122 Messina, Italy;
| | - Christian Cadeddu Dessalvi
- Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy; (L.F.); (N.C.); (S.C.); (M.D.)
| |
Collapse
|
3
|
Scalia IG, Pathangey G, Abdelnabi M, Ibrahim OH, Abdelfattah FE, Pietri MP, Ibrahim R, Farina JM, Banerjee I, Tamarappoo BK, Arsanjani R, Ayoub C. Applications of Artificial Intelligence for the Prediction and Diagnosis of Cancer Therapy-Related Cardiac Dysfunction in Oncology Patients. Cancers (Basel) 2025; 17:605. [PMID: 40002200 PMCID: PMC11852369 DOI: 10.3390/cancers17040605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/04/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Cardiovascular diseases and cancer are the leading causes of morbidity and mortality in modern society. Expanding cancer therapies that have improved prognosis may also be associated with cardiotoxicity, and extended life span after survivorship is associated with the increasing prevalence of cardiovascular disease. As such, the field of cardio-oncology has been rapidly expanding, with an aim to identify cardiotoxicity and cardiac disease early in a patient who is receiving treatment for cancer or is in survivorship. Artificial intelligence is revolutionizing modern medicine with its ability to identify cardiac disease early. This article comprehensively reviews applications of artificial intelligence specifically applied to electrocardiograms, echocardiography, cardiac magnetic resonance imaging, and nuclear imaging to predict cardiac toxicity in the setting of cancer therapies, with a view to reduce early complications and cardiac side effects from cancer therapies such as chemotherapy, radiation therapy, or immunotherapy.
Collapse
Affiliation(s)
- Isabel G. Scalia
- Department of Cardiovascular Diseases, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.); (M.A.); (O.H.I.); (F.E.A.); (M.P.P.); (R.I.); (J.M.F.); (B.K.T.)
| | - Girish Pathangey
- Department of Cardiovascular Diseases, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.); (M.A.); (O.H.I.); (F.E.A.); (M.P.P.); (R.I.); (J.M.F.); (B.K.T.)
| | - Mahmoud Abdelnabi
- Department of Cardiovascular Diseases, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.); (M.A.); (O.H.I.); (F.E.A.); (M.P.P.); (R.I.); (J.M.F.); (B.K.T.)
| | - Omar H. Ibrahim
- Department of Cardiovascular Diseases, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.); (M.A.); (O.H.I.); (F.E.A.); (M.P.P.); (R.I.); (J.M.F.); (B.K.T.)
| | - Fatmaelzahraa E. Abdelfattah
- Department of Cardiovascular Diseases, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.); (M.A.); (O.H.I.); (F.E.A.); (M.P.P.); (R.I.); (J.M.F.); (B.K.T.)
| | - Milagros Pereyra Pietri
- Department of Cardiovascular Diseases, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.); (M.A.); (O.H.I.); (F.E.A.); (M.P.P.); (R.I.); (J.M.F.); (B.K.T.)
| | - Ramzi Ibrahim
- Department of Cardiovascular Diseases, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.); (M.A.); (O.H.I.); (F.E.A.); (M.P.P.); (R.I.); (J.M.F.); (B.K.T.)
| | - Juan M. Farina
- Department of Cardiovascular Diseases, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.); (M.A.); (O.H.I.); (F.E.A.); (M.P.P.); (R.I.); (J.M.F.); (B.K.T.)
| | - Imon Banerjee
- Department of Radiology, Mayo Clinic, Phoenix, AZ 85054, USA;
| | - Balaji K. Tamarappoo
- Department of Cardiovascular Diseases, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.); (M.A.); (O.H.I.); (F.E.A.); (M.P.P.); (R.I.); (J.M.F.); (B.K.T.)
| | - Reza Arsanjani
- Department of Cardiovascular Diseases, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.); (M.A.); (O.H.I.); (F.E.A.); (M.P.P.); (R.I.); (J.M.F.); (B.K.T.)
| | - Chadi Ayoub
- Department of Cardiovascular Diseases, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.); (M.A.); (O.H.I.); (F.E.A.); (M.P.P.); (R.I.); (J.M.F.); (B.K.T.)
| |
Collapse
|
4
|
Simela C, Walker JM, Ghosh AK, Chen DH. SGLT2 inhibitors for prevention and management of cancer treatment-related cardiovascular toxicity: a review of potential mechanisms and clinical insights. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2025; 11:15. [PMID: 39934910 DOI: 10.1186/s40959-024-00284-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 10/30/2024] [Indexed: 02/13/2025]
Abstract
More evidence-based strategies are needed for preventing and managing cancer treatment-related cardiovascular toxicity (CTR-CVT). Owing to the growing body of evidence supporting their cardioprotective role in several cardiac injury scenarios, sodium-glucose cotransporter 2 inhibitors (SGLT2i) may be beneficial for preventing and treating CTR-CVT. In October 2024, a search was conducted of the PubMed database to review full studies investigating the cardioprotective role of SGLT2i against CTR-CVT. We identified 44 full published/pre-print studies and 3 ongoing randomised controlled trial across eight types of cancer treatment (anthracyclines, platinum-containing therapy, immune checkpoint inhibitors, HER2-targeted therapies, kinase inhibitors, androgen deprivation therapies, multiple myeloma therapies and 5-fluorouracil). Most studies used animal models and focussed on primary prevention. 43 of the 44 studies found some cardioprotective effect of SGLT2i against CTR-CVT, which in some cases included preventing ejection fraction decline and aberrations in cardiac electrophysiological parameters. Some studies also observed beneficial effects on mortality. A central triad of anti-inflammatory, anti-oxidative and anti-apoptotic mechanisms likely underlie SGLT2i-mediated cardioprotection against CTR-CVT. Overall, this growing body of research suggests that SGLT2i may be a promising candidate for preventing CTR-CVT either as monotherapy or in combination with other cardioprotective drugs. However, the literature is limited in that no prospective randomised controlled trials investigating SGLT2i for the prevention and management of CTR-CVT exist and most existing human retrospective data is based on diabetic populations. Future work must focus on addressing these limitations of the current literature.
Collapse
Affiliation(s)
- Carl Simela
- University College London Hospital, London, UK
| | - J Malcolm Walker
- University College London Hospital, London, UK
- Hatter Cardiovascular Institute, University College London, London, UK
| | - Arjun K Ghosh
- University College London Hospital, London, UK
- Hatter Cardiovascular Institute, University College London, London, UK
- Barts Heart Centre, London, UK
| | - Daniel H Chen
- University College London Hospital, London, UK.
- Hatter Cardiovascular Institute, University College London, London, UK.
- Barts Heart Centre, London, UK.
- Prince of Wales Hospital, Sydney, NSW, Australia.
- St George Hospital, Sydney NSW, Australia.
| |
Collapse
|
5
|
Dai B, Xu J, Wang B. A meta-analysis of risk factors for cardiovascular adverse events with anthracycline based chemotherapy in lymphoma patients. BMC Cancer 2025; 25:162. [PMID: 39875837 PMCID: PMC11773729 DOI: 10.1186/s12885-024-13305-3] [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] [Received: 07/04/2024] [Accepted: 12/06/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Anthracycline usage has been linked to cardiovascular adverse events (CAEs), which is unpredictable. It is critical to identify the characteristics of vulnerable populations and risk factors in order to reduce the occurrence of CAEs. OBJECTIVES This meta-analysis aimed to assess the correlation between various risk factors and CAEs induced by anthracyclines. METHODS We systematically searched for studies from PubMed, Cochrane, Embase, and assessed the publication bias. Anthracyclines, hypertension, radiation therapy, diabetes, smoking, age, gender, hyperlipidemia, and obesity were meta-analyzed using a fixed or random effects model. RESULT Sixteen studies were included in this meta-analysis. The results showed that pooled relative ratio for CAEs was 1.69 (95% CI: 1.39-2.06, p<0.001) for anthracyclines, and that risk factors for CAEs caused by anthracyclines included hypertension (RR = 2.15, 95% CI: 1.53-3.01, p < 0.001), radiation therapy (RR = 1.71, 95% CI: 1.19-2.47, p < 0.001), diabetes (RR = 1.57, 95% CI: 1.20-2.06, p<0.001), smoking (RR = 1.35, 95% CI: 0.96-1.88, p < 0.05), and age (RR = 1.16, 95% CI: 1.07-1.26, p < 0.001). In addition, BMI > 25 kg/m2 (RR = 1.34, 95% CI: 1.08-1.67, p = 0.470), gender (RR = 1.17, 95% CI: 1.06-1.2, p = 0.350), and hyperlipidemia (RR = 1.10, 95% CI: 0.91-1.34, p = 0.327) was not significantly associated with CAEs caused by anthracyclines. CONCLUSIONS Our research findings indicate that patients with a history of hypertension, radiation therapy, diabetes, smoking, and elderly individuals are at an elevated risk of cardiovascular disease in the process of administration of anthracycline drugs. Consequently, careful case selection and condition monitoring are important in the treatment process.
Collapse
Affiliation(s)
- Bo Dai
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, Shaanxi, 710072, China
| | - Jingjing Xu
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, Shaanxi, 710072, China
| | - Baiyan Wang
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 West 5th Road, Xi'an, Shaanxi, 710004, China.
- Xi'an Key Laboratory of Hematological Diseases, 157 West 5th Road, Xi'an, Shaanxi, 710004, China.
| |
Collapse
|
6
|
Fabiani I, Chianca M, Cipolla CM, Cardinale DM. Anthracycline-induced cardiomyopathy: risk prediction, prevention and treatment. Nat Rev Cardiol 2025:10.1038/s41569-025-01126-1. [PMID: 39875555 DOI: 10.1038/s41569-025-01126-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/17/2025] [Indexed: 01/30/2025]
Abstract
Anthracyclines are the cornerstone of treatment for many malignancies. However, anthracycline cardiotoxicity is a considerable concern given that it can compromise the clinical effectiveness of the treatment and patient survival despite early discontinuation of therapy or dose reduction. Patients with cancer receiving anthracycline treatment can have a reduction in their quality of life and likelihood of survival due to cardiotoxicity, irrespective of their oncological prognosis. Increasing knowledge about anthracycline cardiotoxicity has enabled the identification of patients who are candidates for anthracycline regimens and those who might develop anthracycline-induced cardiomyopathy. Anthracycline cardiotoxicity is a unique and evolving phenomenon that begins with myocardial cell damage, progresses to reduced left ventricular ejection fraction, and culminates in symptomatic heart failure if it is not promptly detected and treated. Early risk stratification can be guided by imaging or biomarkers. In this Review, we present a comprehensive and clinically useful approach to cardiomyopathy related to anthracycline therapy, encompassing its epidemiology, definition, mechanisms, novel classifications, risk factors and patient risk stratification, diagnostic approaches (including imaging and biomarkers), treatment guidelines algorithms, and the role of new cardioprotective drugs that are used for the treatment of heart failure.
Collapse
Affiliation(s)
- Iacopo Fabiani
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy.
| | - Michela Chianca
- Division of Cardiology, Cardiothoracic and Vascular Department, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Carlo Maria Cipolla
- Cardioncology Unit, Cardioncology and Second Opinion Division, European Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy
| | - Daniela Maria Cardinale
- Cardioncology Unit, Cardioncology and Second Opinion Division, European Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy
| |
Collapse
|
7
|
Botelho LFB, de Melo MDT, de Almeida ALC, Salemi VMC. Accuracy of mitral annular plane systolic excursion in diagnosing anthracycline-induced subclinical cardiotoxicity in patients with breast cancer - a retrospective cohort study. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:76. [PMID: 39497222 PMCID: PMC11533289 DOI: 10.1186/s40959-024-00280-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 10/28/2024] [Indexed: 11/07/2024]
Abstract
BACKGROUND The mitral annular plane systolic excursion (MAPSE) is used to analyze the left ventricle longitudinal function. However, the accuracy of MAPSE in diagnosing oncological populations is unclear. In this study, we aimed to assess the accuracy of MAPSE in diagnosing subclinical cardiotoxicity in women with breast cancer undergoing anthracycline treatment. METHODS This retrospective cohort study included echocardiographic assessments of patients with breast cancer who underwent anthracycline treatment as part of their therapeutic regimen. Assessments were performed before treatment, after administering the first dose of anthracycline, after completing anthracycline treatment, and 6 and 12 months after treatment. Left ventricular ejection fraction was calculated using the modified biplane Simpson method. The performances of MAPSE and global longitudinal strain (GLS) were analyzed using receiver operating characteristic (ROC) curves. Their accuracies were measured using the area under the ROC curves. RESULTS Sixty-one patients were included in this study. Of them, 8.2% presented cardiotoxicity 6 months after treatment completion. Patients with cardiotoxicity had lower LVEF (47% vs. 63%; p < 0.001), MAPSE (10.23 mm vs. 12.25 mm; p = 0.012), and LV GLS (16.13% vs. 19.05%; p = 0.005) values than those without. A 12% reduction in the GLS exhibited sensitivity, specificity, and overall accuracy of 80%, 70%, and 78%, respectively. A relative reduction of 15% in MAPSE exhibited a sensitivity, specificity, and accuracy of 80%, 77%, and 81.2%, respectively. An absolute MAPSE reduction of 2 mm exhibited a sensitivity, specificity, and accuracy of 80%, 73.21%, and 81.2%, respectively. No differences were observed between the ROC curves. CONCLUSION MAPSE showed similar accuracy to GLS in diagnosing subclinical cardiotoxicity in women with breast cancer undergoing anthracycline treatment.
Collapse
Affiliation(s)
- Luís Fábio Barbosa Botelho
- Department of Internal Medicine, Federal University of Paraiba, Rua da Aurora 333, Ap 904, João Pessoa, 58043-900, João Pessoa, PB, Brazil.
| | - Marcelo Dantas Tavares de Melo
- Department of Internal Medicine, Federal University of Paraiba, Rua da Aurora 333, Ap 904, João Pessoa, 58043-900, João Pessoa, PB, Brazil
| | | | - Vera Maria Cury Salemi
- Heart Institute (InCor), School of Medicine, University of São Paulo, São Paulo, Brazil
- Hospital Sírio Libanês, São Paulo, Brazil
| |
Collapse
|
8
|
Shaaban NH, Abayazeed RM, Sobhy MA, Elsharkawy EM, Hammad BA. Role of Global Left Ventricle Longitudinal Strain and Cardiac Biomarkers in the Early Detection of Cancer Therapy-Related Dysfunction in Patients Treated With Cardiotoxic Chemotherapeutic Drugs in a Cardio-Oncology Clinic. Cureus 2024; 16:e71766. [PMID: 39553110 PMCID: PMC11569507 DOI: 10.7759/cureus.71766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2024] [Indexed: 11/19/2024] Open
Abstract
Background Breast cancer (BC) affects many women, and with the prevalence of anthracyclines (AC) used in treatment, cardiotoxicity is a commonly encountered problem. Objective The aim is to early detect subclinical cancer therapy-related cardiac dysfunction (CTRCD) using noninvasive imaging techniques and cardiac biomarkers. Methods Eighty-eight patients with cancer who planned to receive AC or trastuzumab (TZB) were enrolled. Baseline screening included two-dimensional (2D) transthoracic echocardiogram (TTE), global longitudinal strain (GLS), cardiac troponin I (cTnI), and N-terminal pro-brain natriuretic peptide (NT-ProBNP) measurements. Follow-up was done at three and six months to early detect CTRCD. Results Twenty-six patients developed CTRCD, defined by a relative decline in GLS and left ventricular ejection fraction (LVEF). The percentage of change in GLS from baseline and at three and six months was able to detect CTRCD in both groups in our population, which was >16.6% at three months with a p-value of <0.001* and CI 0.783-0.934 and >10.10% at six months with a p-value of <0.001* and CI 0.765-0.935. At three months, GLS values of ≤-18.6 were able to detect CTRCD with a p-value of <0.001* and CI 0.673 0.885. Compared to patients who did not develop CRTD, patients with mild asymptomatic CTRCD had double levels of NT-ProBNP with a median of (99.5) (interquartile range (IQR): 44.0-154.0) at three months. Conclusion The relative decline of GLS and elevation of NT-proBNP were able to diagnose patients with subclinical CTRCD in patients receiving AC with early start of cardioprotective treatments.
Collapse
Affiliation(s)
| | | | | | | | - Basma A Hammad
- Cardiology, Alexandria University, Alexandria, EGY
- Cardiology, Royal Brompton and Harefield Hospitals, London, GBR
| |
Collapse
|
9
|
Austin D, Maier RH, Akhter N, Sayari M, Ogundimu E, Maddox JM, Vahabi S, Humphreys AC, Graham J, Oxenham H, Haney S, Cresti N, Verrill M, Osborne W, Wright KL, Goranova R, Bailey JR, Kalakonda N, Macheta M, Kilner MF, Young ME, Morley NJ, Neelakantan P, Gilbert G, Thomas BK, Graham RJ, Fujisawa T, Mills NL, Hildreth V, Prichard J, Kasim AS, Hancock HC, Plummer C. Preventing Cardiac Damage in Patients Treated for Breast Cancer and Lymphoma: The PROACT Clinical Trial. JACC CardioOncol 2024; 6:684-696. [PMID: 39479329 PMCID: PMC11520224 DOI: 10.1016/j.jaccao.2024.07.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/10/2024] [Accepted: 07/12/2024] [Indexed: 11/02/2024] Open
Abstract
Background Cardiotoxicity is a concern for cancer survivors undergoing anthracycline chemotherapy. Enalapril has been explored for its potential to mitigate cardiotoxicity in cancer patients. The dose-dependent cardiotoxicity effects of anthracyclines can be detected early through the biomarker cardiac troponin. Objectives The PROACT (Preventing Cardiac Damage in Patients Treated for Breast Cancer and Lymphoma) clinical trial assessed the effectiveness of enalapril in preventing cardiotoxicity, manifesting as myocardial injury and cardiac function impairment, in patients undergoing high-dose anthracycline-based chemotherapy for breast cancer or non-Hodgkin lymphoma. Methods This prospective, multicenter, open-label, randomized controlled trial employed a superiority design with observer-blinded endpoints. A total of 111 participants, scheduled for 6 cycles of chemotherapy with a planned dose of ≥300 mg/m2 doxorubicin equivalents, were randomized to receive either enalapril (titrated up to 20 mg daily) or standard care without enalapril. Results Myocardial injury, indicated by cardiac troponin T (≥14 ng/L), during and 1 month after chemotherapy, was observed in 42 (77.8%) of 54 patients in the enalapril group vs 45 (83.3%) of 54 patients in the standard care group (OR: 0.65; 95% CI: 0.23-1.78). Injury detected by cardiac troponin I (>26.2 ng/L) occurred in 25 (47.2%) of 53 patients on enalapril compared with 24 (45.3%) of 53 in standard care (OR: 1.10; 95% CI: 0.50-2.38). A relative decline of more than 15% from baseline in left ventricular global longitudinal strain was observed in 10 (21.3%) of 47 patients on enalapril and 9 (21.9%) of 41 in standard care (OR: 0.95; 95% CI: 0.33-2.74). An absolute decline of >10% to <50% in left ventricular ejection fraction was seen in 2 (4.1%) of 49 patients on enalapril vs none in patients in standard care. Conclusions Adding enalapril to standard care during chemotherapy did not prevent cardiotoxicity in patients receiving high-dose anthracycline-based chemotherapy. (PROACT: Can we prevent Chemotherapy-related Heart Damage in Patients With Breast Cancer and Lymphoma?; NCT03265574).
Collapse
Affiliation(s)
- David Austin
- Academic Cardiovascular Unit, James Cook University Hospital, South Tees Hospitals NHS Foundation Trust, Middlesbrough, United Kingdom
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Rebecca H. Maier
- Academic Cardiovascular Unit, James Cook University Hospital, South Tees Hospitals NHS Foundation Trust, Middlesbrough, United Kingdom
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Nasima Akhter
- School of Health and Life Sciences, Teesside University, Middlesbrough, United Kingdom
| | - Mohammad Sayari
- Department of Mathematical Science, Durham University, Durham, United Kingdom
| | - Emmanuel Ogundimu
- Department of Mathematical Science, Durham University, Durham, United Kingdom
| | - Jamie M. Maddox
- Department of Haematology, South Tees Hospitals NHS Foundation Trust, Middlesbrough, United Kingdom
| | - Sharareh Vahabi
- Bristol Heart Institute, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, United Kingdom
| | - Alison C. Humphreys
- Cancer Services, South Tees Hospitals NHS Foundation Trust, Middlesbrough, United Kingdom
| | - Janine Graham
- Cancer Services, South Tees Hospitals NHS Foundation Trust, Middlesbrough, United Kingdom
- Cancer Services, North Tees and Hartlepool NHS Foundation Trust, Stockton-on-Tees, United Kingdom
| | - Helen Oxenham
- Department of Cardiology, North Tees and Hartlepool NHS Foundation Trust, Stockton-on-Tees, United Kingdom
| | - Sophie Haney
- Cancer Services, South Tees Hospitals NHS Foundation Trust, Middlesbrough, United Kingdom
- Cancer Services, County Durham and Darlington NHS Foundation Trust, Darlington, United Kingdom
| | - Nicola Cresti
- Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Mark Verrill
- Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Wendy Osborne
- Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Kathryn L. Wright
- Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
- Cancer Services, South Tyneside and Sunderland NHS Foundation Trust, Sunderland, United Kingdom
| | - Rebecca Goranova
- Department of Oncology, Derriford Hospital, University Hospitals Plymouth NHS Trust, Plymouth, United Kingdom
| | - James R. Bailey
- Department of Haematology, Hull University Teaching Hospitals NHS Trust, Queen’s Centre for Oncology and Haematology, Castle Hill Hospital, Hull, United Kingdom
| | - Nagesh Kalakonda
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Mac Macheta
- Department of Haematology, Blackpool Teaching Hospitals, Blackpool, United Kingdom
| | - Mari F. Kilner
- Pathology Services, Northumbria Healthcare NHS Foundation Trust, North Shields, United Kingdom
| | - Moya E. Young
- Department of Haematology, East Kent Hospitals University NHS Foundation Trust, Canterbury, United Kingdom
| | - Nick J. Morley
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
| | - Pratap Neelakantan
- Department of Haematology, Royal Berkshire NHS Foundation Trust, Reading, United Kingdom
| | - Georgia Gilbert
- Academic Cardiovascular Unit, James Cook University Hospital, South Tees Hospitals NHS Foundation Trust, Middlesbrough, United Kingdom
| | - Byju K. Thomas
- Academic Cardiovascular Unit, James Cook University Hospital, South Tees Hospitals NHS Foundation Trust, Middlesbrough, United Kingdom
| | - Richard J. Graham
- Academic Cardiovascular Unit, James Cook University Hospital, South Tees Hospitals NHS Foundation Trust, Middlesbrough, United Kingdom
| | - Takeshi Fujisawa
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Nicholas L. Mills
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Victoria Hildreth
- Newcastle Clinical Trials Unit, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Jonathan Prichard
- Newcastle Clinical Trials Unit, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Adetayo S. Kasim
- Department of Anthropology, Durham University, Durham, United Kingdom
| | - Helen C. Hancock
- Newcastle Clinical Trials Unit, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Chris Plummer
- Department of Cardiology, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
- Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
10
|
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
|
11
|
Hashemi MS, Farsiani Y, Pressman GS, Amini MR, Kheradvar A. Effect of cross-platform variations on transthoracic echocardiography measurements and clinical diagnosis. EUROPEAN HEART JOURNAL. IMAGING METHODS AND PRACTICE 2024; 2:qyae097. [PMID: 39391529 PMCID: PMC11465166 DOI: 10.1093/ehjimp/qyae097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 09/18/2024] [Indexed: 10/12/2024]
Abstract
Aims Accurate cardiac chamber quantification is essential for clinical decisions and ideally should be consistent across different echocardiography systems. This study evaluates variations between the Philips EPIQ CVx (version 9.0.3) and Canon Aplio i900 (version 7.0) in measuring cardiac volumes, ventricular function, and valve structures. Methods and results In this gender-balanced, single-centre study, 40 healthy volunteers (20 females and 20 males) aged 40 years and older (mean age 56.75 ± 11.57 years) were scanned alternately with both systems by the same sonographer using identical settings for both 2D and 4D acquisitions. We compared left ventricular (LV) and right ventricular (RV) volumes using paired t-tests, with significance set at P < 0.05. Correlation and Bland-Altman plots were used for quantities showing significant differences. Two board-certified cardiologists evaluated valve anatomy for each platform. The results showed no significant differences in LV end-systolic volume and LV ejection fraction between platforms. However, LV end-diastolic volume (LVEDV) differed significantly (biplane: P = 0.018; 4D: P = 0.028). Right ventricular (RV) measurements in 4D showed no significant differences, but there were notable disparities in 2D and 4D volumes within each platform (P < 0.01). Significant differences were also found in the LV systolic dyssynchrony index (P = 0.03), LV longitudinal strain (P = 0.04), LV twist (P = 0.004), and LV torsion (P = 0.005). Valve structure assessments varied, with more abnormalities noted on the Philips platform. Conclusion Although LV and RV volumetric measurements are generally comparable, significant differences in LVEDV, LV strain metrics, and 2D vs. 4D measurements exist. These variations should be considered when using different platforms for patient follow-ups.
Collapse
Affiliation(s)
| | - Yasaman Farsiani
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Gregg S Pressman
- Division of Cardiology, Thomas Jefferson University, Thomas Jefferson Einstein Hospital, Philadelphia, PA, USA
| | - M Reza Amini
- Section of Cardiology, Department of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Arash Kheradvar
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
- Mary & Steve Wen Cardiovascular Division, Department of Medicine, University of California Irvine, Orange, CA, USA
| |
Collapse
|
12
|
Scalia IG, Gheyath B, Tamarappoo BK, Moudgil R, Otton J, Pereyra M, Narayanasamy H, Larsen C, Herrmann J, Arsanjani R, Ayoub C. Chemotherapy Related Cardiotoxicity Evaluation-A Contemporary Review with a Focus on Cardiac Imaging. J Clin Med 2024; 13:3714. [PMID: 38999280 PMCID: PMC11242267 DOI: 10.3390/jcm13133714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 07/14/2024] Open
Abstract
The long-term survivorship of patients diagnosed with cancer has improved due to accelerated detection and rapidly evolving cancer treatment strategies. As such, the evaluation and management of cancer therapy related complications has become increasingly important, including cardiovascular complications. These have been captured under the umbrella term "cardiotoxicity" and include left ventricular dysfunction and heart failure, acute coronary syndromes, valvular abnormalities, pericardial disease, arrhythmia, myocarditis, and vascular complications. These complications add to the burden of cardiovascular disease (CVD) or are risk factors patients with cancer treatment are presenting with. Of note, both pre- and newly developing CVD is of prognostic significance, not only from a cardiovascular perspective but also overall, potentially impacting the level of cancer therapy that is possible. Currently, there are varying recommendations and practices regarding CVD risk assessment and mitigating strategies throughout the cancer continuum. This article provides an overview on this topic, in particular, the role of cardiac imaging in the care of the patient with cancer. Furthermore, it summarizes the current evidence on the spectrum, prevention, and management of chemotherapy-related adverse cardiac effects.
Collapse
Affiliation(s)
- Isabel G. Scalia
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.)
| | - Bashaer Gheyath
- Department of Imaging, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Balaji K. Tamarappoo
- Division of Cardiology, Banner University Medical Center, The University of Arizona College of Medicine, Phoenix, AZ 85004, USA
| | - Rohit Moudgil
- Department of Cardiology, Heart and Vascular Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - James Otton
- Clinical School, St. Vincent’s Hospital, UNSW, Sydney, NSW 2010, Australia
| | - Milagros Pereyra
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.)
| | - Hema Narayanasamy
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.)
| | - Carolyn Larsen
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.)
| | - Joerg Herrmann
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Reza Arsanjani
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.)
| | - Chadi Ayoub
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.)
| |
Collapse
|
13
|
Taghdiri A. Anthracycline-induced hypertension in pediatric cancer survivors: unveiling the long-term cardiovascular risks. Egypt Heart J 2024; 76:71. [PMID: 38849680 PMCID: PMC11161443 DOI: 10.1186/s43044-024-00506-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 06/05/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Long-term cardiovascular complications are common among pediatric cancer survivors, and anthracycline-induced hypertension has become an essential reason for concern. Compared to non-cancer controls, survivors have a higher prevalence of hypertension, and as they age, their incidence rises, offering significant dangers to cardiovascular health. MAIN BODY Research demonstrates that exposure to anthracyclines is a major factor in the development of hypertension in children who have survived cancer. Research emphasizes the frequency and risk factors of anthracycline-induced hypertension, highlighting the significance of routine measurement and management of blood pressure. Furthermore, cardiovascular toxicities, such as hypertension, after anthracycline-based therapy are a crucial be concerned, especially for young adults and adolescents. Childhood cancer survivors deal with a variety of cardiovascular diseases, such as coronary artery disease and cardiomyopathy, which are made worse by high blood pressure. In order to prevent long-term complications, it is essential to screen for and monitor for anthracycline-induced hypertension. Echocardiography and cardiac biomarkers serve as essential tools for early detection and treatment. In order to lower cardiovascular risks in pediatric cancer survivors, comprehensive management strategies must include lifestyle and medication interventions in addition to survivor-centered care programs. SHORT CONCLUSION Proactive screening, monitoring, and management measures are necessary for juvenile cancer survivors due to the substantial issue of anthracycline-induced hypertension in their long-term care. To properly include these strategies into survivor-ship programs, oncologists, cardiologists, and primary care physicians need to collaborate together. The quality of life for pediatric cancer survivors can be enhanced by reducing the cardiovascular risks linked to anthracycline therapy and promoting survivor-centered care and research.
Collapse
Affiliation(s)
- Andia Taghdiri
- Faculty of Medicine, Ivane Javakhishvili Tbilisi State University, Tbilisi, Georgia.
| |
Collapse
|
14
|
Gorgiladze N, Shavdia M, Gaprindashvili T, Gogua E, Gachechiladze L, Gujabidze M, Pagava Z. Detection of Cardiotoxicity Using Right Ventricular Free Wall Longitudinal Strain in Low Cardiovascular Risk Breast Cancer Patients Receiving Low-Dose Anthracycline Treatment. Cureus 2024; 16:e63138. [PMID: 39055440 PMCID: PMC11272134 DOI: 10.7759/cureus.63138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2024] [Indexed: 07/27/2024] Open
Abstract
Objective Breast cancer patients who receive chemotherapy may develop cancer therapy-related cardiovascular toxicity, particularly if they have pre-existing cardiovascular risk factors. Notably, right ventricle dysfunction may manifest before the left ventricle. Our study aims to compare conventional echocardiography with global longitudinal strain (GLS) in low cardiovascular risk patients on low-dose anthracycline, focusing on early cardiotoxicity detection. Additionally, we explore the predictive role of right ventricular free wall longitudinal strain (RVFWLS) in cardiotoxicity. Methods In a recent study, 28 women with low cardiovascular risk who underwent low-dose anthracycline chemotherapy for breast cancer were assessed for cardiac function using two-dimensional echocardiography and speckle-tracking echocardiography. The measurements included left ventricular ejection fraction (LVEF), right ventricular systolic function (RVS'), tricuspid annular plane systolic excursion (TAPSE), left ventricular global longitudinal strain (LVGLS), and RVFWLS. All patients had normal LVEF at the beginning of the study. Cardiotoxicity was defined as a new decrease in LVEF by 10% or below 53% and/or changes in LVGLS/RVFWLS by 15%. Results In our study, no significant changes were observed in the LVEF following chemotherapy treatment. The LVEF values remained stable, changing slightly from 63 ± 3.7 to 65.0 ± 3.4, with a t-test value of 1.790 and a p-value of 0.079. Similarly, the analysis found no significant changes in RVS' and TAPSE values following chemotherapy treatment. However, significant changes were observed in strain measurements. LVGLS decreased from -21.2 ± 2.1 to -18.6 ± 2.6 (t-test = -4.116; df = 54, p=0.001), and RVFWLS decreased from -25.2 ± 2.9 to -21.4 ± 4.4 (t-test = -3.82; df = 54, p=0.001). Notably, 35% of participants showed changes in RVFWLS greater than 15%, whereas LVGLS changed by less than 15%. This indicates that RVFWLS is more sensitive to the treatment compared to LVGLS. Conclusions The study results indicate that during the initial phases of chemotherapy treatment in low cardiovascular risk patients, early changes in strain measures reveal subclinical cardiotoxicity. This suggests that GLS measurements are more effective at detecting early signs of myocardial damage and potential deterioration in cardiac function than traditional echocardiographic parameters. Additionally, it is noteworthy that RVFWLS exhibits greater sensitivity to these changes, regardless of the chemotherapy dosage and regimen.
Collapse
Affiliation(s)
- Nana Gorgiladze
- Department of Clinical Oncology, Tbilisi State Medical University, Tbilisi, GEO
| | - Mikheil Shavdia
- Department of Clinical Oncology, Tbilisi State Medical University, Tbilisi, GEO
| | - Tamar Gaprindashvili
- Department of Cardiopulmonology, Bokhua Memorial Cardiovascular Center, Tbilisi, GEO
| | - Elene Gogua
- Department of Oncology, Institute of Clinical Oncology, Tbilisi, GEO
| | | | | | - Zurab Pagava
- Department of Cardiopulmonology, Bokhua Memorial Cardiovascular Center, Tbilisi, GEO
| |
Collapse
|
15
|
Chong A, Stanton T, Taylor A, Prior D, La Gerche A, Anderson B, Scalia G, Cooke J, Dahiya A, To A, Davis M, Mottram P, Moir S, Playford D, Mahadavan D, Thomas L, Wahi S. 2024 CSANZ Position Statement on Indications, Assessment and Monitoring of Structural and Valvular Heart Disease With Transthoracic Echocardiography in Adults. Heart Lung Circ 2024; 33:773-827. [PMID: 38749800 DOI: 10.1016/j.hlc.2023.11.028] [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: 11/24/2022] [Revised: 08/22/2023] [Accepted: 11/01/2023] [Indexed: 06/25/2024]
Abstract
Transthoracic echocardiography (TTE) is the most widely available and utilised imaging modality for the screening, diagnosis, and serial monitoring of all abnormalities related to cardiac structure or function. The primary objectives of this document are to provide (1) a guiding framework for treating clinicians of the acceptable indications for the initial and serial TTE assessments of the commonly encountered cardiovascular conditions in adults, and (2) the minimum required standard for TTE examinations and reporting for imaging service providers. The main areas covered within this Position Statement pertain to the TTE assessment of the left and right ventricles, valvular heart diseases, pericardial diseases, aortic diseases, infective endocarditis, cardiac masses, pulmonary hypertension, and cardiovascular diseases associated with cancer treatments or cardio-oncology. Facilitating the optimal use and performance of high quality TTEs will prevent the over or under-utilisation of this resource and unnecessary downstream testing due to suboptimal or incomplete studies.
Collapse
Affiliation(s)
- Adrian Chong
- Department of Cardiology, Princess Alexandra Hospital, Mater Hospital Brisbane, University of Queensland, Brisbane, Qld, Australia
| | - Tony Stanton
- Sunshine Coast University Hospital, School of Health University of Sunshine Coast, School of Medicine and Dentistry Griffith University, Birtinya, Qld, Australia
| | - Andrew Taylor
- Department of Cardiology, Royal Melbourne Hospital, Alfred Hospital, Melbourne, Vic, Australia
| | - David Prior
- Albury Wodonga Health, Albury, NSW, Australia
| | - Andre La Gerche
- St Vincent's Hospital, Baker Heart and Diabetes Institute, University of Melbourne, Melbourne, Vic, Australia
| | - Bonita Anderson
- Cardiac Sciences Unit, The Prince Charles Hospital, Queensland University of Technology, Brisbane, Qld, Australia
| | - Gregory Scalia
- The Prince Charles Hospital, University of Queensland, Brisbane, Qld, Australia
| | - Jennifer Cooke
- Department of Cardiology, Eastern Health, Monash University, Melbourne, Vic, Australia
| | - Arun Dahiya
- Department of Cardiology, Princess Alexandra Hospital, Logan Hospital, Griffith University, Brisbane, Qld, Australia
| | - Andrew To
- Department of Cardiology, Health New Zealand Waitemata, Auckland, New Zealand
| | | | - Philip Mottram
- Victorian Heart Institute, Monash University, Melbourne, Vic, Australia
| | - Stuart Moir
- Victorian Heart Institute, Monash University, Melbourne, Vic, Australia
| | | | - Devan Mahadavan
- Department of Cardiology, Queen Elizabeth Hospital, Lyell McEwin Hospital, Adelaide, SA, Australia
| | - Liza Thomas
- Department of Cardiology, Westmead Hospital, Westmead Clinical School University of Sydney, South West Clinical School University of New South Wales, Sydney, NSW, Australia
| | - Sudhir Wahi
- Department of Cardiology, Princess Alexandra Hospital, University of Queensland, Brisbane, Qld, Australia.
| |
Collapse
|
16
|
Sletten OJ, Aalen JM, Smiseth OA, Khan FH, Fossa A, Kiserud CE, Villegas-Martinez M, Hisdal J, Remme EW, Skulstad H. Mental Stress Reduces Left Ventricular Strain: Can It Lead to Misinterpretation of Cancer Therapy-Related Cardiac Dysfunction? J Am Soc Echocardiogr 2024; 37:564-566. [PMID: 37981246 DOI: 10.1016/j.echo.2023.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 11/03/2023] [Accepted: 11/07/2023] [Indexed: 11/21/2023]
Affiliation(s)
- Ole J Sletten
- Institute for Surgical Research, Rikshospitalet, Oslo University Hospital, Oslo, Norway; Department of Cardiology, Rikshospitalet, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - John M Aalen
- Institute for Surgical Research, Rikshospitalet, Oslo University Hospital, Oslo, Norway; Department of Cardiology, Rikshospitalet, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Otto A Smiseth
- Institute for Surgical Research, Rikshospitalet, Oslo University Hospital, Oslo, Norway; Department of Cardiology, Rikshospitalet, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Faraz H Khan
- Institute for Surgical Research, Rikshospitalet, Oslo University Hospital, Oslo, Norway; Department of Cardiology, Rikshospitalet, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Alexander Fossa
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Oncology, Oslo University Hospital, Radiumhospitalet, Oslo, Norway
| | - Cecilie E Kiserud
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Oncology, Oslo University Hospital, Radiumhospitalet, Oslo, Norway
| | - Manuel Villegas-Martinez
- Institute for Surgical Research, Rikshospitalet, Oslo University Hospital, Oslo, Norway; The Intervention Center, Oslo University Hospital, Oslo, Norway
| | - Jonny Hisdal
- Institute for Surgical Research, Rikshospitalet, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Section of Vascular Investigations, Oslo University Hospital, Oslo, Norway
| | - Espen W Remme
- Institute for Surgical Research, Rikshospitalet, Oslo University Hospital, Oslo, Norway; The Intervention Center, Oslo University Hospital, Oslo, Norway
| | - Helge Skulstad
- Institute for Surgical Research, Rikshospitalet, Oslo University Hospital, Oslo, Norway; Department of Cardiology, Rikshospitalet, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
17
|
Tan S, Kader Z, Day D, Chen D, Nicholls SJ, Ramkumar S. Cardiotoxicity in Oncology Guidelines: Discrepancies Do Matter. Heart Lung Circ 2024; 33:553-557. [PMID: 38453605 DOI: 10.1016/j.hlc.2024.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 01/29/2024] [Accepted: 02/06/2024] [Indexed: 03/09/2024]
Affiliation(s)
- Sean Tan
- Victorian Heart Institute, Monash University, Melbourne, Vic, Australia; Monash Heart, Victorian Heart Hospital, Melbourne, Vic, Australia.
| | - Zainel Kader
- Monash Heart, Victorian Heart Hospital, Melbourne, Vic, Australia
| | - Daphne Day
- School of Clinical Sciences at Monash Health, Monash University, Melbourne, Vic, Australia; Department of Oncology, Monash Health, Melbourne, Vic, Australia
| | - Daniel Chen
- Prince of Wales and St George Hospitals, South Eastern Sydney Local Health District, Sydney, NSW, Australia; Hatter Cardiovascular Institute, University College of London, London, UK
| | - Stephen J Nicholls
- Victorian Heart Institute, Monash University, Melbourne, Vic, Australia; Monash Heart, Victorian Heart Hospital, Melbourne, Vic, Australia
| | - Satish Ramkumar
- Victorian Heart Institute, Monash University, Melbourne, Vic, Australia; Monash Heart, Victorian Heart Hospital, Melbourne, Vic, Australia
| |
Collapse
|
18
|
Song S, Woo J, Kim H, Lee JW, Lim W, Moon BI, Kwon K. A prospective randomized controlled trial to determine the safety and efficacy of extracorporeal shock waves therapy for primary prevention of subclinical cardiotoxicity in breast cancer patients without a cardiovascular risk treated with doxorubicin. Front Cardiovasc Med 2024; 11:1324203. [PMID: 38385137 PMCID: PMC10879594 DOI: 10.3389/fcvm.2024.1324203] [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: 10/19/2023] [Accepted: 01/26/2024] [Indexed: 02/23/2024] Open
Abstract
Background Doxorubicin is a highly effective anti-cancer drug that causes left ventricular (LV) dysfunction and induces late-onset cardiomyopathy. However, an effective and clinically applicable preventive treatment is yet to be discovered. Objective Cardiac-Extracorporeal shockwave therapy (C-ESWT) has been suggested to treat inflammatory and ischemic diseases and protect cardiomyocytes from doxorubicin-induced cardiomyopathy. This study aims to assess the safety and efficacy of C-ESWT in the prevention of subclinical cardiotoxicity. Methods We enrolled 64 breast cancer patients. C-ESWT group 33 patients were treated with our C-ESWT (200 shots/spot at 0.09 mJ/mm2 for 20 spots, 3 times every six weeks). The efficacy endpoints were the difference in left ventricular global longitudinal strain (LVGLS) change by 2D speckle tracking echocardiography and chemotherapy-related cardiac dysfunction (CTRCD). Echocardiography was performed on the baseline line and every 4 cycles of chemotherapy, followed by a follow-up 3,6 months after chemotherapy to compare the incidence of cardiomyopathy of subclinical LV dysfunction due to chemotherapy between the two groups. Results Participants averaged 50 ± 9 years in age, 100% female. In the results of follow-up 6 months after the end of chemotherapy, there was a significant difference in delta LVGLS between the C-ESWT group and the control group (LVGLS; -1.1 ± 10.9% vs. -11.5 ± 11.6% p-value; <0.001). A total of 23% (15 patients) of patients developed CTRCD (Control group; 13 vs. C-ESWT group; (2). C-ESWT was performed safely without any serious adverse events. Conclusion In this prospective study, C-ESWT established efficacy in preventing subclinical cardiotoxicity, especially in breast cancer patients using doxorubicin chemotherapy, and the safety of C-ESWT. Clinical Trial Registration ClinicalTrials.gov, identifier (NCT05584163).
Collapse
Affiliation(s)
- Shinjeong Song
- Division of Cardiology, Department of Internal Medicine, Ewha Womans University College of Medicine, Ewha Womans University Mokdong Hospital, Seoul, Republic of Korea
| | - Joohyun Woo
- Department of Surgery, Ewha Womans University College of Medicine, Ewha Womans University Mokdong Hospital, Seoul, Republic of Korea
| | - HyunGoo Kim
- Department of Surgery, Ewha Womans University College of Medicine, Ewha Womans University Mokdong Hospital, Seoul, Republic of Korea
| | - Jun Woo Lee
- Department of Surgery, Ewha Womans University College of Medicine, Ewha Womans University Mokdong Hospital, Seoul, Republic of Korea
| | - Woosung Lim
- Department of Surgery, Ewha Womans University College of Medicine, Ewha Womans University Mokdong Hospital, Seoul, Republic of Korea
| | - Byung-In Moon
- Department of Surgery, Ewha Womans University College of Medicine, Ewha Womans University Mokdong Hospital, Seoul, Republic of Korea
| | - Kihwan Kwon
- Division of Cardiology, Department of Internal Medicine, Ewha Womans University College of Medicine, Ewha Womans University Mokdong Hospital, Seoul, Republic of Korea
| |
Collapse
|
19
|
Kesting S, Giordano U, Weil J, McMahon CJ, Albert DC, Berger C, Budts W, Fritsch P, Hidvégi EV, Oberhoffer-Fritz R, Milano GM, Wacker-Gußmann A, Herceg-Čavrak V. Association of European Paediatric and Congenital Cardiology practical recommendations for surveillance and prevention of cardiac disease in childhood cancer survivors: the importance of physical activity and lifestyle changes From the Association of European Paediatric and Congenital Cardiology Working Group Sports Cardiology, Physical Activity and Prevention, Working Group Adult Congenital Heart Disease, Working Group Imaging and Working Group Heart Failure. Cardiol Young 2024; 34:250-261. [PMID: 38174736 DOI: 10.1017/s1047951123004213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
BACKGROUND Childhood cancer survivors are at increased risk of developing cardiovascular diseases, presenting as the main causes of morbidity and mortality within this group. Besides the usual primary and secondary prevention in combination with screening during follow-up, the modifiable lifestyle factors of physical activity, nutrition, and body weight have not yet gained enough attention regarding potential cardiovascular risk reduction. OBJECTIVE These practical recommendations aim to provide summarised information and practical implications to paediatricians and health professionals treating childhood cancer survivors to reduce the risk of cardiovascular late effects. METHODS The content derives from either published guidelines or expert opinions from Association of European Paediatric and Congenital Cardiology working groups and is in accordance with current state-of-the-art. RESULTS All usual methods of prevention and screening regarding the risk, monitoring, and treatment of occurring cardiovascular diseases are summarised. Additionally, modifiable lifestyle factors are explained, and clear practical implications are named. CONCLUSION Modifiable lifestyle factors should definitely be considered as a cost-effective and complementary approach to already implemented follow-up care programs in cardio-oncology, which can be actively addressed by the survivors themselves. However, treating physicians are strongly encouraged to support survivors to develop and maintain a healthy lifestyle, including physical activity as one of the major influencing factors. This article summarises relevant background information and provides specific practical recommendations on how to advise survivors to increase their level of physical activity.
Collapse
Affiliation(s)
- Sabine Kesting
- Institute of Preventive Paediatrics, Department Health and Sport Sciences, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- Kinderklinik München Schwabing, Department of Paediatrics and Children's Cancer Research Centre, Department Clinical Medicine, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Ugo Giordano
- Pediatric Cardiac Surgery, Cardiology and Heart/Lung Transplantation Department, Bambino Gesù Children's Hospital, Rome, Italy
| | - Jochen Weil
- Department of Paediatric Cardiology and Congenital Heart Disease, German Heart Centre Munich, Technical University Munich, Munich, Germany
| | - Colin J McMahon
- Department of Paediatric Cardiology, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Dimpna C Albert
- Heart Centre, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Claire Berger
- Department of Paediatric Haematology and Oncology, University Jean Monnet, University-Hospital of Saint-Etienne, Saint-Etienne, France
| | - Werner Budts
- Congenital and Structural Cardiology, University Hospitals Leuven, Leuven, Belgium
| | - Peter Fritsch
- Private Practice, Institute for Pediatric Cardiology, Graz, Austria
| | | | - Renate Oberhoffer-Fritz
- Institute of Preventive Paediatrics, Department Health and Sport Sciences, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Giuseppe M Milano
- Department of Hematology/Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCSS), Rome, Italy
| | - Annette Wacker-Gußmann
- Institute of Preventive Paediatrics, Department Health and Sport Sciences, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- Department of Paediatric Cardiology and Congenital Heart Disease, German Heart Centre Munich, Technical University Munich, Munich, Germany
| | - Vesna Herceg-Čavrak
- Faculty of Health Science, Libertas International University, Zagreb, Croatia
| |
Collapse
|
20
|
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
|
21
|
Mertens L, Singh G, Armenian S, Chen MH, Dorfman AL, Garg R, Husain N, Joshi V, Leger KJ, Lipshultz SE, Lopez-Mattei J, Narayan HK, Parthiban A, Pignatelli RH, Toro-Salazar O, Wasserman M, Wheatley J. Multimodality Imaging for Cardiac Surveillance of Cancer Treatment in Children: Recommendations From the American Society of Echocardiography. J Am Soc Echocardiogr 2023; 36:1227-1253. [PMID: 38043984 DOI: 10.1016/j.echo.2023.09.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Affiliation(s)
- Luc Mertens
- Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Gautam Singh
- Children's Hospital of Michigan, Detroit, Michigan; Central Michigan University School of Medicine, Saginaw, Michigan
| | - Saro Armenian
- City of Hope Comprehensive Cancer Center, Duarte, California
| | - Ming-Hui Chen
- Boston Children's Hospital and Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Adam L Dorfman
- University of Michigan, C.S. Mott Children's Hospital, Ann Arbor, Michigan
| | - Ruchira Garg
- Cedars-Sinai Heart Institute, Los Angeles, California
| | | | - Vijaya Joshi
- St. Jude Children's Research Hospital/University of Tennessee College of Medicine, Memphis, Tennessee
| | - Kasey J Leger
- University of Washington, Seattle Children's Hospital, Seattle, Washington
| | - Steven E Lipshultz
- University at Buffalo Jacobs School of Medicine and Biomedical Sciences, Oishei Children's Hospital, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | | | - Hari K Narayan
- University of California San Diego, Rady Children's Hospital San Diego, San Diego, California
| | - Anitha Parthiban
- Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | | | - Olga Toro-Salazar
- Connecticut Children's Medical Center, University of Connecticut School of Medicine, Hartford, Connecticut
| | | | | |
Collapse
|
22
|
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
|
23
|
Balaji S, Antony AK, Tonchev H, Scichilone G, Morsy M, Deen H, Mirza I, Ali MM, Mahmoud AM. Racial Disparity in Anthracycline-induced Cardiotoxicity in Breast Cancer Patients. Biomedicines 2023; 11:2286. [PMID: 37626782 PMCID: PMC10452913 DOI: 10.3390/biomedicines11082286] [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: 07/26/2023] [Revised: 08/13/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Breast cancer has become the most common cancer in the US and worldwide. While advances in early detection and treatment have resulted in a 40% reduction in breast cancer mortality, this reduction has not been achieved uniformly among racial groups. A large percentage of non-metastatic breast cancer mortality is related to the cardiovascular effects of breast cancer therapies. These effects appear to be more prevalent among patients from historically marginalized racial/ethnic backgrounds, such as African American and Hispanic individuals. Anthracyclines, particularly doxorubicin and daunorubicin, are the first-line treatments for breast cancer patients. However, their use is limited by their dose-dependent and cumulative cardiotoxicity, manifested by cardiomyopathy, ischemic heart disease, arrhythmias, hypertension, thromboembolic disorders, and heart failure. Cardiotoxicity risk factors, such as genetic predisposition and preexisting obesity, diabetes, hypertension, and heart diseases, are more prevalent in racial/ethnic minorities and undoubtedly contribute to the risk. Yet, beyond these risk factors, racial/ethnic minorities also face unique challenges that contribute to disparities in the emerging field of cardio-oncology, including socioeconomic factors, food insecurity, and the inability to access healthcare providers, among others. The current review will address genetic, clinical, and social determinants that potentially contribute to this disparity.
Collapse
Affiliation(s)
- Swetha Balaji
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.B.); (A.K.A.); (H.T.); (G.S.); (M.M.); (H.D.); (I.M.); (M.M.A.)
| | - Antu K. Antony
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.B.); (A.K.A.); (H.T.); (G.S.); (M.M.); (H.D.); (I.M.); (M.M.A.)
| | - Harry Tonchev
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.B.); (A.K.A.); (H.T.); (G.S.); (M.M.); (H.D.); (I.M.); (M.M.A.)
| | - Giorgia Scichilone
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.B.); (A.K.A.); (H.T.); (G.S.); (M.M.); (H.D.); (I.M.); (M.M.A.)
| | - Mohammed Morsy
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.B.); (A.K.A.); (H.T.); (G.S.); (M.M.); (H.D.); (I.M.); (M.M.A.)
| | - Hania Deen
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.B.); (A.K.A.); (H.T.); (G.S.); (M.M.); (H.D.); (I.M.); (M.M.A.)
| | - Imaduddin Mirza
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.B.); (A.K.A.); (H.T.); (G.S.); (M.M.); (H.D.); (I.M.); (M.M.A.)
| | - Mohamed M. Ali
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.B.); (A.K.A.); (H.T.); (G.S.); (M.M.); (H.D.); (I.M.); (M.M.A.)
| | - Abeer M. Mahmoud
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.B.); (A.K.A.); (H.T.); (G.S.); (M.M.); (H.D.); (I.M.); (M.M.A.)
- Department of Kinesiology, College of Applied Health Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
24
|
Theetha Kariyanna P, Kumar A, Jayarangaiah A, Shetty M, Chowdhury Y, Das S, Jayarangaiah A. Chemotherapy induced right ventricular cardiomyopathy; a systematic review and meta-analysis. Front Cardiovasc Med 2023; 10:1103941. [PMID: 37600030 PMCID: PMC10434797 DOI: 10.3389/fcvm.2023.1103941] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 06/29/2023] [Indexed: 08/22/2023] Open
Abstract
Background Left ventricular dysfunction and cardiomyopathy are well documented adverse effects associated with chemotherapy agents. Limited information exists regarding the impact of chemotherapeutic agents on the integrity and function of the right ventricle (RV). Objectives The current metanalysis compared pre- chemotherapy versus post- chemotherapy RV parameters measured on 2D echocardiography in patients receiving anthracycline and/or trastuzumab across all breast cancer patients. Methods A systematic search across PubMed, EMBASE and Cochrane databases were performed from inception of the databases until November 2021 for relevant studies. We used the inverse variance method with a random effect model and DerSimonian and Laird method of Tau2 generation to calculate mean difference [MD] with 95% confidence interval [CI]. The analysis was carried out using RevMan Version 5.3 (Copenhagen: The Nordic Cochrane Centre, The Cochrane Collaboration, 2014). Results Fifteen studies, constituting total of 644 patients, met the inclusion criteria, with most studies having a follow up period of less than 12 months from initiation of chemotherapy. Anthracycline and/or Trastuzumab chemotherapy resulted in a statistically significant reduction in right ventricular ejection fraction (RVEF) at follow-up [MD: 2.70, 95% CI: 0.27 to 5.13, P-value- 0.03, I2- 71%, χ2 P-value < 0.05]. Treatment with Anthracycline and/or Trastuzumab chemotherapy resulted in a significant reduction in RV fractional area change (RVFAC) at follow-up [MD: 3.74, 95% CI: 1.33 to 6.15, P-value < 0.01, I2- 68%, χ2 P-value < 0.05]. RV free wall longitudinal strain (RVFWLS) was lower at baseline, while LVEF was significantly reduced at follow-up [MD: -1.00, 95% CI: -1.86 to -0.15, P-value < 0.05, I2- 0%, χ2 P-value-0.40], [MD: 4.04, 95% CI: 2.08 to 6.01, P-value < 0.01, I2- 91%, χ2 P-value < 0.05], respectively. However, treatment with Anthracycline and/or Trastuzumab chemotherapy had no statistically significant effect on Tricuspid annular plane systolic excursion (TAPSE) at follow-up [MD: 0.53, 95% CI: -0.11 to 1.17, P-value-0.11, I2- 98%, χ2 P-value < 0.05]. Conclusions Chemotherapy with anthracyclines and trastuzumab negatively affects right ventricular function leading to decline in RVEF, RVFAC, RVFWLS and LVEF.
Collapse
Affiliation(s)
- Pramod Theetha Kariyanna
- Department of Cardiology, Chaparral Medical Group/Pomona Valley Hospital Medical Center, Pomona, CA, United States
| | - Ashish Kumar
- Department of Internal Medicine, Cleveland Clinic Akron General, Akron, OH, United States
| | - Amog Jayarangaiah
- Department of Internal Medicine, Marshfield Clinic, Marshfield, WI, United States
| | - Mrinali Shetty
- Department of Cardiology, New York Presbyterian Hospital (Columbia and Cornell Campus), New York, NY, United States
| | - Yuvraj Chowdhury
- Department of Cardiology, University of Massachusetts, Boston, MA, United States
| | - Sushruth Das
- Avalon University School of Medicine, Willemstad, Curaçao
| | - Apoorva Jayarangaiah
- Department of Hematology/Oncology, Prevea Clinic/HSHS Sacred Heart, Eau Claire, WI, United States
| |
Collapse
|
25
|
Patricelli C, Lehmann P, Oxford JT, Pu X. Doxorubicin-Induced Modulation of TGF-β Signaling Cascade in Mouse Fibroblasts: Insights into Cardiotoxicity Mechanisms. RESEARCH SQUARE 2023:rs.3.rs-3186393. [PMID: 37546862 PMCID: PMC10402200 DOI: 10.21203/rs.3.rs-3186393/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
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. Moreover, we observed that DOX inhibited the TGF-β1 induced expression of BMP1 in NIH3T3 cells, while BMP1 levels remained high in CFs, and that TGF-β1 induces the phosphorylation of SMAD2 in both NIH3T3 cells and CFs. While DOX treatment diminished the extent of phosphorylation, the reduction did not reach statistical significance. DOX also inhibited the TGF-β1 induced expression of COL1 in NIH3T3 cells and CFs. Finally, DOX inhibited the TGF-β1 induced expression of Atf4 and increased the expression of Cdkn1a, Id1, Id2, Runx1, Tgfb1, Inhba, Thbs1, Bmp1, and Stat1 in NIH3T3 cells but not CFs, indicating the potential for cell-specific responses to DOX and its modulation of the TGF-β signaling pathway. Understanding the underlying mechanisms of the ability of DOX to modulate gene expression and signaling pathways in fibroblasts holds promise for future development of targeted therapeutic strategies to mitigate DOX-induced cardiotoxicity specifically affecting CFs.
Collapse
|
26
|
O'Riordan CE, Trochet P, Steiner M, Fuchs D. Standardisation and future of preclinical echocardiography. Mamm Genome 2023; 34:123-155. [PMID: 37160810 DOI: 10.1007/s00335-023-09981-4] [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: 07/29/2022] [Accepted: 01/31/2023] [Indexed: 05/11/2023]
Abstract
Echocardiography is a non-invasive imaging technique providing real-time information to assess the structure and function of the heart. Due to advancements in technology, ultra-high-frequency transducers have enabled the translation of ultrasound from humans to small animals due to resolutions down to 30 µm. Most studies are performed using mice and rats, with ages ranging from embryonic, to neonatal, and adult. In addition, alternative models such as zebrafish and chicken embryos are becoming more frequently used. With the achieved high temporal and spatial resolution in real-time, cardiac function can now be monitored throughout the lifespan of these small animals to investigate the origin and treatment of a range of acute and chronic pathological conditions. With the increased relevance of in vivo real-time imaging, there is still an unmet need for the standardisation of small animal echocardiography and the appropriate cardiac measurements that should be reported in preclinical cardiac models. This review focuses on the development of standardisation in preclinical echocardiography and reports appropriate cardiac measurements throughout the lifespan of rodents: embryonic, neonatal, ageing, and acute and chronic pathologies. Lastly, we will discuss the future of cardiac preclinical ultrasound.
Collapse
Affiliation(s)
| | | | | | - Dieter Fuchs
- FUJIFILM VisualSonics, Inc, Amsterdam, The Netherlands.
| |
Collapse
|
27
|
Liang S, Liu M, Liu Z, Zhong X, Qin Y, Liang T, Wang X, Tang Z, Li Q, Huang H. Global longitudinal strain assessment in contrast-enhanced echocardiography in breast cancer patients: a feasibility study. Cardiovasc Ultrasound 2023; 21:7. [PMID: 37081550 PMCID: PMC10116775 DOI: 10.1186/s12947-023-00304-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 04/02/2023] [Indexed: 04/22/2023] Open
Abstract
BACKGROUND Left ventricular global longitudinal strain (GLS) obtained from two-dimensional speckle-tracking echocardiography (2D-STE) can reflect cancer therapy-related cardiac dysfunction in breast cancer (BC) patients, however, the accuracy and reproducibility of 2D-STE are restricted due to poor image quality. METHODS Between January 2019 and October 2021, 160 consecutive BC patients aged ≥ 18 years were recruited. The 160 BC patients (mean age: 48.41 ± 9.93 years, 100% women) underwent both 2D-STE and Contrast-enhanced echocardiography (CEcho), 125 of whom were included in the measurement of GLS. The intraclass correlation coefficient (ICC) was used to determine the intra- and inter-observer reproducibility of 2D-STE and CEcho-STE. Correlation (r) was calculated using Pearson correlation. Statistical significance was set at P < 0.05. RESULTS Among 160 BC patients, more segments were recognized by CEcho-STE than by 2D-STE (2,771, 99.53% vs. 2,440, 84.72%). The left ventricular ejection fraction (LVEF) obtained by 2D was lower than CEcho (61.75 ± 6.59% vs. 64.14 ± 5.97%, P < 0.0001). The GLS obtained by 2D-STE was lower than CEcho-STE (-21.74 ± 2.77% vs. -26.79 ± 4.30%, P = 0.001). The ICC of the intraobserver and interobserver agreements in the CEcho-STE group was lower than that in the 2D-STE group. GLS measurements were in good agreement between the 2D-STE and CEcho-STE groups (r = 0.773). CONCLUSIONS CEcho can overcome some imaging limitations and recognize more segments than 2D, which may provide an LVEF and GLS closer to the true value. Based on AutoStrain, CEcho-STE may serve as a complementary method for those with poor image quality.
Collapse
Affiliation(s)
- Shichu Liang
- Department of Cardiology, West China Hospital, Sichuan University, No.37 GuoXue Alley, Chengdu, 610041, China
| | - Mei Liu
- Department of Cardiology, West China Hospital, Sichuan University, No.37 GuoXue Alley, Chengdu, 610041, China
| | - Zhiyue Liu
- Department of Cardiology, West China Hospital, Sichuan University, No.37 GuoXue Alley, Chengdu, 610041, China
| | - Xiaorong Zhong
- Department of Head and Neck Oncology Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
- Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yupei Qin
- Department of Cardiology, West China Hospital, Sichuan University, No.37 GuoXue Alley, Chengdu, 610041, China
| | - Ting Liang
- Department of Cardiology, West China Hospital, Sichuan University, No.37 GuoXue Alley, Chengdu, 610041, China
| | - Xi Wang
- Department of Cardiology, West China Hospital, Sichuan University, No.37 GuoXue Alley, Chengdu, 610041, China
| | - Zhuoqin Tang
- Department of Cardiology, West China Hospital, Sichuan University, No.37 GuoXue Alley, Chengdu, 610041, China
| | - Qian Li
- Department of Cardiology, West China Hospital, Sichuan University, No.37 GuoXue Alley, Chengdu, 610041, China
| | - He Huang
- Department of Cardiology, West China Hospital, Sichuan University, No.37 GuoXue Alley, Chengdu, 610041, China.
| |
Collapse
|
28
|
Global Longitudinal Strain in Cardio-Oncology: A Review. Cancers (Basel) 2023; 15:cancers15030986. [PMID: 36765941 PMCID: PMC9913863 DOI: 10.3390/cancers15030986] [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: 12/26/2022] [Revised: 01/25/2023] [Accepted: 01/30/2023] [Indexed: 02/09/2023] Open
Abstract
Several therapies used in cancer treatment are potentially cardiotoxic and may cause left ventricular (LV) dysfunction and heart failure. For decades, echocardiography has been the main modality for cardiac assessment in cancer patients, and the parameter examined in the context of cardiotoxicity was the left ventricular ejection fraction (LVEF). The assessment of the global longitudinal strain (GLS) using speckle tracking echocardiography (STE) is an emerging method for detecting and quantifying subtle disturbances in the global long-axis LV systolic function. In the latest ESC guidelines on cardio-oncology, GLS is an important element in diagnosing the cardiotoxicity of oncological therapy. A relative decrease in GLS of >15% during cancer treatment is the recommended cut-off point for suspecting subclinical cardiac dysfunction. An early diagnosis of asymptomatic cardiotoxicity allows the initiation of a cardioprotective treatment and reduces the risk of interruptions or changes in the oncological treatment in the event of LVEF deterioration, which may affect survival.
Collapse
|
29
|
Zhang C, Chen Z, Qin S, Zhu Y, Shu L, Zuo Z. Incidence of adverse cardiovascular events associated with immune checkpoint inhibitors and risk factors for left ventricular dysfunction: A single-center prospective clinical study. Front Cardiovasc Med 2023; 10:1052699. [PMID: 36755798 PMCID: PMC9899928 DOI: 10.3389/fcvm.2023.1052699] [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: 09/24/2022] [Accepted: 01/04/2023] [Indexed: 01/24/2023] Open
Abstract
Background The incidence of immune checkpoint inhibitors (ICI)-related adverse cardiovascular events (ACEs) may be underestimated, and there are few reports on the incidence and risk factors of ICI-induced left ventricular dysfunction (LVD). Objectives This study aimed to investigate the incidence of ACEs caused by ICI, in particular to analyze the incidence and risk factors of LV systolic and diastolic dysfunction. Materials and methods A prospective clinical study was performed on patients who received ICI in our hospital from November 2020 to October 2021. They received regular cardiovascular examinations, including echocardiography, ECG, cTnT, and NT-proBNP, etc. The incidence of various ACEs was counted, and the risk factors of LVD were analyzed. Results A total of 106 cancer patients treated with ICI were recruited. During the follow-up, 41 patients (38.68%) developed various ECG abnormalities, 39 patients (36.79%) developed LVDD, 9 patients (8.49%) developed CTRCD, and 2 patients (1.89%) developed new pericardial effusion. The patients with elevated cTnT, CK-MB, and NT-proBNP were 10 (9.43%), 8 (7.55%), and 8 (7.5%), respectively. Thirteen of the 52 patients with LVD had hypertension, while 4 of the 54 patients without LVD had hypertension (OR = 4.17, 95% CI: 1.26-13.78; P = 0.019). The baseline LVEF and LVFS of patients with LVD were 61.54 ± 4.15% and 33.78 ± 2.73%, while those of the control group were 64.16 ± 3.68% and 34.95 ± 2.84, respectively (P = 0.003 and P = 0.048). Compared with patients without LVD, patients with LVD had lower e' (6.99 ± 1.33 cm/s vs. 7.64 ± 1.39 cm/s, P = 0.029) and higher E to e' ratio (11.89 ± 3.15 cm/s vs. 10.43 ± 2.52, P = 0.024). Multiple regression analysis showed that a history of hypertension (HR = 26.52, 95% CI: 2.479-283.667, P = 0.007) and lower baseline e' (HR = 0.04, 95% CI: 0.003-0.709, P = 0.028) were risk factors for developing LVD. Conclusion Patients treated with ICI may develop multiple ACEs, including acute myocarditis, pericarditis, ECG abnormalities, and elevated cardiac biomarkers. ICI may lead to a high incidence of LVD, and echocardiography is helpful for early detection of LVD. Patients with hypertension or poor LV systolic or diastolic function at baseline were predictors of LVD after ICI treatment.
Collapse
Affiliation(s)
- Chuan Zhang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhulu Chen
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shu Qin
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuxi Zhu
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Linjie Shu
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhong Zuo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China,*Correspondence: Zhong Zuo,
| |
Collapse
|
30
|
The Role of Multimodality Cardiac Imaging in Patients Undergoing Cancer Treatment. Curr Cardiol Rep 2023; 25:1-8. [PMID: 36527535 DOI: 10.1007/s11886-022-01825-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/24/2022] [Indexed: 12/23/2022]
Abstract
PURPOSE OF REVIEW Modern therapeutics have led to improved survival for many types of cancer but have also been associated with adverse effects including potentially life-threatening cardiotoxicities. We sought to review the uses of multimodality cardiac imaging for risk stratification, prevention, and identification of cardiotoxicities in patients undergoing cancer treatment. RECENT FINDINGS Advancements in both echocardiography and emerging modalities, like cardiac magnetic resonance imaging and cardiac computed tomography, continue to improve the pre- and during therapy cardiac evaluation of cancer patients. Echocardiography and cardiac magnetic resonance imaging, with the incorporation of global longitudinal strain, can identify overt and subclinical cancer therapy-related cardiac dysfunction and myocarditis, and stress echocardiography and cardiac computed tomography can noninvasively screen and monitor for coronary artery disease. Multimodality cardiac imaging is an evolving and critical tool for the pre-therapy screening and risk stratification, as well as during therapy surveillance of cancer treatment-related cardiotoxicity.
Collapse
|
31
|
Barrett M, Wilcox NS, Huang A, Levy R, Demissei B, Narayan V, Ky B. Bearing allostatic load: insights into a more equitable future within cardio-oncology. Trends Mol Med 2022; 28:1040-1049. [PMID: 36207229 DOI: 10.1016/j.molmed.2022.09.006] [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: 06/15/2022] [Revised: 08/23/2022] [Accepted: 09/08/2022] [Indexed: 11/09/2022]
Abstract
Chronic stress is often regarded as a significant cause of morbidity and mortality; however, the mechanistic link between stress and various disease states has not yet been fully characterized. We explore the concept of allostatic load, a measurement of the physiological burden of chronic stress, as well as its potential role in disease pathogenesis as it relates to cardiovascular disease, cancer, and health-related disparities. Building from this framework, we then posit the potential implications of allostatic load on patient care and research in cardio-oncology. We identify allostatic load as a potential clinically actionable tool to improve health equity in cardio-oncology.
Collapse
Affiliation(s)
- Matthew Barrett
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nicholas S Wilcox
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Anran Huang
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Richard Levy
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Biniyam Demissei
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Vivek Narayan
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bonnie Ky
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
32
|
Mikail N, Rossi A, Bengs S, Haider A, Stähli BE, Portmann A, Imperiale A, Treyer V, Meisel A, Pazhenkottil AP, Messerli M, Regitz-Zagrosek V, Kaufmann PA, Buechel RR, Gebhard C. Imaging of heart disease in women: review and case presentation. Eur J Nucl Med Mol Imaging 2022; 50:130-159. [PMID: 35974185 PMCID: PMC9668806 DOI: 10.1007/s00259-022-05914-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 07/12/2022] [Indexed: 11/04/2022]
Abstract
Cardiovascular diseases (CVD) remain the leading cause of mortality worldwide. Although major diagnostic and therapeutic advances have significantly improved the prognosis of patients with CVD in the past decades, these advances have less benefited women than age-matched men. Noninvasive cardiac imaging plays a key role in the diagnosis of CVD. Despite shared imaging features and strategies between both sexes, there are critical sex disparities that warrant careful consideration, related to the selection of the most suited imaging techniques, to technical limitations, and to specific diseases that are overrepresented in the female population. Taking these sex disparities into consideration holds promise to improve management and alleviate the burden of CVD in women. In this review, we summarize the specific features of cardiac imaging in four of the most common presentations of CVD in the female population including coronary artery disease, heart failure, pregnancy complications, and heart disease in oncology, thereby highlighting contemporary strengths and limitations. We further propose diagnostic algorithms tailored to women that might help in selecting the most appropriate imaging modality.
Collapse
Affiliation(s)
- Nidaa Mikail
- Department of Nuclear Medicine, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Alexia Rossi
- Department of Nuclear Medicine, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Susan Bengs
- Department of Nuclear Medicine, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Achi Haider
- Department of Nuclear Medicine, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Barbara E Stähli
- Department of Cardiology, University Heart Center, University Hospital Zurich, Zurich, Switzerland
| | - Angela Portmann
- Department of Nuclear Medicine, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Alessio Imperiale
- Nuclear Medicine and Molecular Imaging - Institut de Cancérologie de Strasbourg Europe (ICANS), University of Strasbourg, Strasbourg, France
- Molecular Imaging - DRHIM, IPHC, UMR 7178, CNRS/Unistra, Strasbourg, France
| | - Valerie Treyer
- Department of Nuclear Medicine, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
| | - Alexander Meisel
- Department of Nuclear Medicine, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Aju P Pazhenkottil
- Department of Nuclear Medicine, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
- Department of Cardiology, University Heart Center, University Hospital Zurich, Zurich, Switzerland
| | - Michael Messerli
- Department of Nuclear Medicine, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
| | - Vera Regitz-Zagrosek
- Charité, Universitätsmedizin, Berlin, Berlin, Germany
- University of Zurich, Zurich, Switzerland
| | - Philipp A Kaufmann
- Department of Nuclear Medicine, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
| | - Ronny R Buechel
- Department of Nuclear Medicine, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
| | - Cathérine Gebhard
- Department of Nuclear Medicine, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland.
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland.
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW Heart failure results in the high incidence and mortality all over the world. Mechanical properties of myocardium are critical determinants of cardiac function, with regional variations in myocardial contractility demonstrated within infarcted ventricles. Quantitative assessment of cardiac contractile function is therefore critical to identify myocardial infarction for the early diagnosis and therapeutic intervention. RECENT FINDINGS Current advancement of cardiac functional assessments is in pace with the development of imaging techniques. The methods tailored to advanced imaging have been widely used in cardiac magnetic resonance, echocardiography, and optical microscopy. In this review, we introduce fundamental concepts and applications of representative methods for each imaging modality used in both fundamental research and clinical investigations. All these methods have been designed or developed to quantify time-dependent 2-dimensional (2D) or 3D cardiac mechanics, holding great potential to unravel global or regional myocardial deformation and contractile function from end-systole to end-diastole. Computational methods to assess cardiac contractile function provide a quantitative insight into the analysis of myocardial mechanics during cardiac development, injury, and remodeling.
Collapse
|
34
|
Sampaio DPS, Silva JBM, do Carmo Rassi D, Freitas AF, Rassi S. Echocardiographic strategy for early detection of cardiotoxicity of doxorubicin: a prospective observational study. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2022; 8:17. [PMID: 36183108 PMCID: PMC9526268 DOI: 10.1186/s40959-022-00143-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 09/14/2022] [Indexed: 11/06/2022]
Abstract
Background Cancer chemotherapy using anthracyclines is associated with cardiotoxicity (CTX), and left ventricular ejection fraction (LVEF) analysis is not sensitive to early cardiotoxic changes. Left ventricular global longitudinal strain (LV GLS) monitoring helps screen subclinical CTX; however, the intervals at which it should be performed remain unclear. We aimed to evaluate the incidence of CTX in women with breast cancer and the associated factors and compare two echocardiographic monitoring strategies using two cutoff points for LV GLS variation. Methods Patients with breast cancer prescribed doxorubicin underwent serial LVEF and LV GLS assessments using two-dimensional echocardiography every 3 weeks for 6 months. Results We included 43 women; none developed a clinical CTX. Considering a relative reduction of LV GLS > 15%, subclinical CTX was present in 12 (27.9%) and six (14%) patients at 3-week and 3-month intervals, respectively (P = 0.28). Additionally, considering a reduction of > 12%, subclinical CTX was present in 17 (39.5%) and 10 (23.3%) patients (P = 0.16), respectively. There were no significant differences in either reference value at 3-week (P = 0.19) and 3-month intervals (P = 0.41). Age ≥ 60 years (P = 0.018) and hypertension (HTN) (P = 0.022) were associated with subclinical CTX in the univariate analysis. Conclusions There was no difference in the incidence of subclinical CTX between the two cutoff points and no benefit in performing echocardiography every 3 weeks compared with quarterly monitoring. Advanced age and HTN were associated with the development of subclinical CTX.
Collapse
Affiliation(s)
- Diogo Pereira Santos Sampaio
- grid.411195.90000 0001 2192 5801Division of Cardiology, Department of Internal Medicine, Faculty of Medicine, Federal University of Goiás, Rua 235 s/n, Setor Leste Universitário, Goiânia, Goiás 74605-050 Brazil
| | - João Batista Masson Silva
- grid.411195.90000 0001 2192 5801Department of Echocardiography, Cardiovascular Division, Hospital das Clínicas of the Federal University of Goiás, Goiânia, Goiás Brazil
| | - Daniela do Carmo Rassi
- grid.411195.90000 0001 2192 5801Division of Cardiology, Department of Internal Medicine, Faculty of Medicine, Federal University of Goiás, Rua 235 s/n, Setor Leste Universitário, Goiânia, Goiás 74605-050 Brazil
| | - Aguinaldo F. Freitas
- grid.411195.90000 0001 2192 5801Division of Cardiology, Department of Internal Medicine, Faculty of Medicine, Federal University of Goiás, Rua 235 s/n, Setor Leste Universitário, Goiânia, Goiás 74605-050 Brazil
| | - Salvador Rassi
- grid.411195.90000 0001 2192 5801Division of Cardiology, Department of Internal Medicine, Faculty of Medicine, Federal University of Goiás, Rua 235 s/n, Setor Leste Universitário, Goiânia, Goiás 74605-050 Brazil
| |
Collapse
|
35
|
Kwon SS, Nam BD, Lee MY, Lee MH, Lee J, Park BW, Bang DW, Kwon SH. Increased EAT volume after anthracycline chemotherapy is associated with a low risk of cardiotoxicity in breast cancer. Breast Cancer Res Treat 2022; 196:111-119. [DOI: 10.1007/s10549-022-06696-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 07/27/2022] [Indexed: 11/02/2022]
|
36
|
Tonry C, Russel-Hallinan A, McCune C, Collier P, Harbinson M, Dixon L, Watson CJ. Circulating biomarkers for management of cancer therapeutics related cardiac dysfunction. Cardiovasc Res 2022; 119:710-728. [PMID: 35640873 PMCID: PMC10153425 DOI: 10.1093/cvr/cvac087] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 05/09/2022] [Accepted: 05/15/2022] [Indexed: 11/14/2022] Open
Abstract
Cancer therapeutics related cardiac dysfunction (CTRCD) has emerged as a major cause of morbidity and mortality in cancer survivors. Effective clinical management of CTRCD is impeded by a lack of sensitive diagnostic and prognostic strategies. Circulating molecular markers could potentially address this need as they are often indicative of cardiac stress before cardiac damage can be detected clinically. A growing understanding of the underlying physiological mechanisms for CTRCD has inspired research efforts to identify novel pathophysiologically-relevant biomarkers that may also guide development of cardio-protective therapeutic approaches. The purpose of this review is to evaluate current circulating biomarkers of cardiac stress and their potential role in diagnosis and management of CTRCD. We also discuss some emerging avenues for CTRCD-focused biomarker investigations.
Collapse
Affiliation(s)
- Claire Tonry
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Northern Ireland, United Kingdom
| | - Adam Russel-Hallinan
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Northern Ireland, United Kingdom
| | - Claire McCune
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Northern Ireland, United Kingdom
| | | | | | | | - Chris J Watson
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Northern Ireland, United Kingdom
| |
Collapse
|
37
|
New Insights on the Toxicity on Heart and Vessels of Breast Cancer Therapies. Med Sci (Basel) 2022; 10:medsci10020027. [PMID: 35736347 PMCID: PMC9229896 DOI: 10.3390/medsci10020027] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 11/17/2022] Open
Abstract
Cardiovascular diseases are largely represented in patients with cancer and appear to be important side effects of cancer treatments, heavily affecting quality of life and leading to premature morbidity and death among cancer survivors. In particular, treatments for breast cancer have been shown to potentially play serious detrimental effects on cardiovascular health. This review aims to explore the available literature on breast cancer therapy-induced side effects on heart and vessels, illustrating the molecular mechanisms of cardiotoxicity known so far. Moreover, principles of cardiovascular risk assessment and management of cardiotoxicity in clinical practice will also be elucidated. Chemotherapy (anthracycline, taxanes, cyclophosphamide and 5-fluorouracil), hormonal therapy (estrogen receptor modulator and gonadotropin or luteinizing releasing hormone agonists) and targeted therapy (epidermal growth factor receptor 2 and Cyclin-dependent kinases 4 and 6 inhibitors) adverse events include arterial and pulmonary hypertension, supraventricular and ventricular arrhythmias, systolic and diastolic cardiac dysfunction and coronary artery diseases due to different and still not well-dissected molecular pathways. Therefore, cardiovascular prevention programs and treatment of cardiotoxicity appear to be crucial to improve morbidity and mortality of cancer survivors.
Collapse
|
38
|
Lee SH, Cho I, You SC, Cha MJ, Chang JS, Kim WD, Go KY, Kim DY, Seo J, Shim CY, Hong GR, Kang SM, Ha JW, Rha SY, Kim HS. Cancer Therapy-Related Cardiac Dysfunction in Patients Treated with a Combination of an Immune Checkpoint Inhibitor and Doxorubicin. Cancers (Basel) 2022; 14:cancers14092320. [PMID: 35565449 PMCID: PMC9100163 DOI: 10.3390/cancers14092320] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/02/2022] [Accepted: 05/05/2022] [Indexed: 02/01/2023] Open
Abstract
Backgrounds: There are scarce data on whether immune checkpoint inhibitors (ICIs) increase the risk of cardiac dysfunction when used with cardiotoxic agents. Thus, we evaluated cardiac dysfunction in patients with sarcoma receiving doxorubicin with or without ICI using echocardiography and left ventricular global longitudinal strain (LVGLS). Methods: A total of 95 patients were included in this study. Echocardiography and LVGLS were evaluated at baseline and follow-up (at 3 and 6 months of chemotherapy) and compared with the doxorubicin (Dox; n = 73) and concomitant ICI with doxorubicin (Dox-ICI; n = 22) groups. Cancer therapy-related cardiac dysfunction (CTRCD) was defined as a left ventricular ejection fraction (LVEF) drop of >10% and LVEF of <50% (definite CTRCD), LVEF drop of >10%, LVEF of ≥50%, and LVGLS relative reduction of >15% (probable CTRCD) at six months. Results: There were no significant differences in age, cumulative dose of doxorubicin, and cardiovascular risk factors between the two groups. At baseline, the LVEF was similar in the Dox and Dox-ICI groups (p = 0.493). In the Dox group, LVEF decreased to 59 ± 6% (Δ −7 ± 1.3%, p < 0.001) and LVGLS decreased from −17.3 ± 3.2% to −15.4 ± 3.2% (Δ −10.1 ± −1.9%, p < 0.001) at six months. In the Dox-ICI group, LVEF decreased to 55 ± 9% (Δ −9 ± 2.1%, p < 0.001), along with a significant decrease in LVGLS (from −18.6 ± 1.9% to −15.3 ± 3.6%, Δ −12.4 ± −2.4%, p < 0.001). Over a median follow-up of 192 days, there were no cases with clinical manifestations of fulminant myocarditis. In the Dox group, definite and probable CTRCD were observed in seven (10.1%) and five (7.4%) patients, respectively. In the Dox-ICI group, definite and probable CTRCD were observed in four (19%) and four (19%) patients, respectively. The total number of patients who developed CTRCD was significantly higher in the Dox-ICI group than in the Dox group (38.1% vs. 17.4%, p = 0.042). Serum troponin-T level was significantly higher in the Dox-ICI group than in the Dox group (53.3 vs. 27.5 pg/mL, p = 0.023). Conclusions: ICIs may increase the risk of CTRCD when used with cardiotoxic agents. CTRCD should be monitored in patients treated with ICIs by cardiac biomarkers and echocardiography, including LV-GLS.
Collapse
Affiliation(s)
- Seon-Hwa Lee
- Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Korea; (S.-H.L.); (K.-y.G.); (D.-Y.K.); (J.S.); (C.-Y.S.); (G.-R.H.); (S.-M.K.); (J.-W.H.)
| | - Iksung Cho
- Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Korea; (S.-H.L.); (K.-y.G.); (D.-Y.K.); (J.S.); (C.-Y.S.); (G.-R.H.); (S.-M.K.); (J.-W.H.)
- Correspondence: (I.C.); (H.-S.K.); Tel.: +82-2-2228-8124 (H.-S.K.); Fax: +82-2-393-3652 (H.-S.K.)
| | - Seng-Chan You
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul 03722, Korea;
| | - Min-Jae Cha
- Department of Radiology, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul 03722, Korea;
| | - Jee-Suk Chang
- Department of Radiation Oncology, Yonsei University College of Medicine, Seoul 03722, Korea;
| | - William D. Kim
- Chung-Ang University College of Medicine, Seoul 03722, Korea;
| | - Kyu-yong Go
- Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Korea; (S.-H.L.); (K.-y.G.); (D.-Y.K.); (J.S.); (C.-Y.S.); (G.-R.H.); (S.-M.K.); (J.-W.H.)
| | - Dae-Young Kim
- Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Korea; (S.-H.L.); (K.-y.G.); (D.-Y.K.); (J.S.); (C.-Y.S.); (G.-R.H.); (S.-M.K.); (J.-W.H.)
| | - Jiwon Seo
- Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Korea; (S.-H.L.); (K.-y.G.); (D.-Y.K.); (J.S.); (C.-Y.S.); (G.-R.H.); (S.-M.K.); (J.-W.H.)
| | - Chi-Young Shim
- Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Korea; (S.-H.L.); (K.-y.G.); (D.-Y.K.); (J.S.); (C.-Y.S.); (G.-R.H.); (S.-M.K.); (J.-W.H.)
| | - Geu-Ru Hong
- Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Korea; (S.-H.L.); (K.-y.G.); (D.-Y.K.); (J.S.); (C.-Y.S.); (G.-R.H.); (S.-M.K.); (J.-W.H.)
| | - Seok-Min Kang
- Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Korea; (S.-H.L.); (K.-y.G.); (D.-Y.K.); (J.S.); (C.-Y.S.); (G.-R.H.); (S.-M.K.); (J.-W.H.)
| | - Jong-Won Ha
- Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Korea; (S.-H.L.); (K.-y.G.); (D.-Y.K.); (J.S.); (C.-Y.S.); (G.-R.H.); (S.-M.K.); (J.-W.H.)
| | - Sun-Young Rha
- Division of Oncology, Yonsei University College of Medicine, Seoul 03722, Korea;
| | - Hyo-Song Kim
- Division of Oncology, Yonsei University College of Medicine, Seoul 03722, Korea;
- Correspondence: (I.C.); (H.-S.K.); Tel.: +82-2-2228-8124 (H.-S.K.); Fax: +82-2-393-3652 (H.-S.K.)
| |
Collapse
|
39
|
Marwick TH. Global Longitudinal Strain Monitoring to Guide Cardioprotective Medications During Anthracycline Treatment. Curr Oncol Rep 2022; 24:687-694. [PMID: 35239105 PMCID: PMC9054886 DOI: 10.1007/s11912-022-01242-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2022] [Indexed: 11/30/2022]
Abstract
Purpose of the Review Anthracycline chemotherapy carries a risk of myocardial dysfunction and heart failure even at relatively low doses, and the clinical prediction of cancer treatment-related cardiac dysfunction (CTRCD) is inexact. Careful imaging or biomarker surveillance during chemotherapy can identify CTRCD before the development of heart failure. Currently, this surveillance is performed using ejection fraction (EF). While this is a reliable and reproducible test with three-dimensional techniques, the most widely used imaging technique is two-dimensional echocardiography, for which EF measurements have broad confidence intervals. Recent Findings The use of global myocardial strain (GLS) provides a more reliable and reproducible means of assessing global cardiac function and shows meaningful changes before a significant change of EF. Observational studies have shown that although absolute measurements of GLS, both at baseline and during therapy, are predictive of CTRCD risk, the most reliable approach is to assess the change of GLS with therapy — a meaningful relative change of 10–15% being significant. A clinical trial comparing GLS to EF surveillance did not show a significant change of EF in the overall study group, but did show that patients managed with a the GLS-guided approach were less likely to develop a meaningful change of cardiac function to an abnormal level. In at-risk patients, there is good evidence for the protective value of neurohormonal antagonists and statins: the use of GLS enables these benefits to be directed to those most likely to benefit, while minimizing their use in the majority of people, who do not need them. Summary Although GLS requires an element of training and efforts to ensure uniformity, it has proven to be a feasible, robust, and reproducible technique, ready for wide adoption.
Collapse
Affiliation(s)
- Thomas H Marwick
- Baker Heart and Diabetes Institute, PO Box 6492, Melbourne, Victoria, 3004, Australia.
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
40
|
Jiang J, Liu B, Hothi SS. Herceptin-Mediated Cardiotoxicity: Assessment by Cardiovascular Magnetic Resonance. Cardiol Res Pract 2022; 2022:1910841. [PMID: 35265371 PMCID: PMC8898877 DOI: 10.1155/2022/1910841] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 10/12/2021] [Accepted: 01/20/2022] [Indexed: 02/07/2023] Open
Abstract
Herceptin (trastuzumab) is a recombinant, humanized, monoclonal antibody that targets the human epidermal growth factor receptor 2 (HER2) and is used in the treatment of HER2-positive breast and gastric cancers. However, it carries a risk of cardiotoxicity, manifesting as left ventricular (LV) systolic dysfunction, conventionally assessed for by transthoracic echocardiography. Clinical surveillance of cardiac function and discontinuation of trastuzumab at an early stage of LV systolic dysfunction allow for the timely initiation of heart failure drug therapies that can result in the rapid recovery of cardiac function in most patients. Often considered the reference standard for the noninvasive assessment of cardiac volume and function, cardiac magnetic resonance (CMR) imaging has superior reproducibility and accuracy compared to other noninvasive imaging modalities. However, due to limited availability, it is not routinely used in the serial assessment of cardiac function in patients receiving trastuzumab. In this article, we review the diagnostic and prognostic role of CMR in trastuzumab-mediated cardiotoxicity.
Collapse
Affiliation(s)
- Jin Jiang
- Heart and Lung Centre, New Cross Hospital, Wolverhampton, UK
| | - Boyang Liu
- Heart and Lung Centre, New Cross Hospital, Wolverhampton, UK
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Sandeep S Hothi
- Heart and Lung Centre, New Cross Hospital, Wolverhampton, UK
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
41
|
Chen D, Kelly C, Haw TJ, Lombard JM, Nordman IIC, Croft AJ, Ngo DTM, Sverdlov AL. Heart Failure in Breast Cancer Survivors: Focus on Early Detection and Novel Biomarkers. Curr Heart Fail Rep 2021; 18:362-377. [PMID: 34731413 DOI: 10.1007/s11897-021-00535-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/29/2021] [Indexed: 01/17/2023]
Abstract
PURPOSE OF REVIEW Breast cancer survival rate has greatly improved in the last two decades due to the emergence of next-generation anti-cancer agents. However, cardiotoxicity remains a significant adverse effect arising from traditional and emerging chemotherapies as well as targeted therapies for breast cancer patients. In this review, we will discuss cardiotoxicities of both traditional and emerging therapies for breast cancer. We will discuss current practices to detect cardiotoxicity of these therapies with the focus on new and emerging biomarkers. We will then focus on 'omics approaches, especially the use of epigenetics to discover novel biomarkers and therapeutics to mitigate cardiotoxicity. RECENT FINDINGS Significant cardiotoxicities of conventional chemotherapies remain and new and unpredictable new forms of cardiac and/or vascular toxicity emerge with the surge in novel and targeted therapies. Yet, there is no clear guidance on detection of cardiotoxicity, except for significant left ventricular systolic dysfunction, and even then, there is no uniform definition of what constitutes cardiotoxicity. The gold standard for detection of cardiotoxicity involves a serial echocardiography in conjunction with blood-based biomarkers to detect early subclinical cardiac dysfunction. However, the ability of these tests to detect early disease remains limited and not all forms of toxicity are detectable with these modalities. There is an unprecedented need to discover novel biomarkers that are sensitive and specific for early detection of subclinical cardiotoxicity. In that space, novel echocardiographic techniques, such as strain, are becoming more common-place and new biomarkers, discovered by epigenetic approaches, seem to become promising alternatives or adjuncts to conventional non-specific cardiac biomarkers.
Collapse
Affiliation(s)
- Dongqing Chen
- Cardio-Oncology & Cardiometabolic Research Group, College of Health, Medicine and Wellbeing, Hunter Medical Research Institute & University of Newcastle, NSW, Callaghan, Australia
| | - Conagh Kelly
- Cardio-Oncology & Cardiometabolic Research Group, College of Health, Medicine and Wellbeing, Hunter Medical Research Institute & University of Newcastle, NSW, Callaghan, Australia
| | - Tatt Jhong Haw
- Cardio-Oncology & Cardiometabolic Research Group, College of Health, Medicine and Wellbeing, Hunter Medical Research Institute & University of Newcastle, NSW, Callaghan, Australia.,Cardio-Oncology & Cardiometabolic Research Group, College of Health, Medicine and Wellbeing, Hunter Medical Research Institute & University of Newcastle Calvary Mater Newcastle, NSW, Waratah, Australia
| | - Janine M Lombard
- Cardio-Oncology & Cardiometabolic Research Group, College of Health, Medicine and Wellbeing, Hunter Medical Research Institute & University of Newcastle Calvary Mater Newcastle, NSW, Waratah, Australia
| | - Ina I C Nordman
- Cardio-Oncology & Cardiometabolic Research Group, College of Health, Medicine and Wellbeing, Hunter Medical Research Institute & University of Newcastle Calvary Mater Newcastle, NSW, Waratah, Australia
| | - Amanda J Croft
- Cardio-Oncology & Cardiometabolic Research Group, College of Health, Medicine and Wellbeing, Hunter Medical Research Institute & University of Newcastle Calvary Mater Newcastle, NSW, Waratah, Australia
| | - Doan T M Ngo
- Cardio-Oncology & Cardiometabolic Research Group, College of Health, Medicine and Wellbeing, Hunter Medical Research Institute & University of Newcastle, NSW, Callaghan, Australia. .,School of Biomedical Science and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia.
| | - Aaron L Sverdlov
- Cardio-Oncology & Cardiometabolic Research Group, College of Health, Medicine and Wellbeing, Hunter Medical Research Institute & University of Newcastle, NSW, Callaghan, Australia. .,Cardio-Oncology & Cardiometabolic Research Group, College of Health, Medicine and Wellbeing, Hunter Medical Research Institute & University of Newcastle Calvary Mater Newcastle, NSW, Waratah, Australia. .,Cardiovascular Department, John Hunter Hospital, Hunter New England Local Health District, NSW, New Lambton Heights, Australia. .,School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia.
| |
Collapse
|