1
|
Şahinbaş M, Çerik İB, Yalınbaş Yeter D. Investigation of the effect of intravitreal bevacizumab treatment on left heart function using speckle tracking echocardiography. Rev Port Cardiol 2025; 44:27-35. [PMID: 39216529 DOI: 10.1016/j.repc.2024.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 06/01/2024] [Accepted: 07/07/2024] [Indexed: 09/04/2024] Open
Abstract
INTRODUCTION AND OBJECTIVES Vascular endothelial growth factor (VEGF) inhibitors are widely used in oncology and ophthalmology. Although these agents have been shown to increase the risk of cardiovascular events in systemic use, the effect of local applications is unclear. In our study, we aimed to investigate the effects of anti-VEGF agents on left heart functions after intravitreal injection using speckle tracking echocardiography. METHODS In this prospectively designed study, 44 patients who were going to start intravitreal anti-VEGF treatment were included in the study. Patients were evaluated with speckle tracking echocardiography before the first anti-VEGF administration and at three months of anti-VEGF treatment. RESULTS Global longitudinal strain (GLS) values at three months were lower in the patients who participated in the study and this was statistically significant (-18.77±2.17, -18.60±2.01, p=0.001). Also, there was a statistically significant decrease in the mean values of GLS (GLS4CH) obtained from apical four space image, GLS (GLSAPLAX) obtained from apical long axis image and GLS (GLS2CH) obtained from apical 2 space image at month 0 and month 3 (-19.08±2.39, -18.93±2.26, p=0.004; -18.81±2.29, -18.60±2.12, p=0.001; -18.44±2.31, -18.27±2.12, p=0.013, respectively). CONCLUSION The slight decrease in GLS in our study suggests that the use of intravitreal anti-VEGF agents may have cardiac effects.
Collapse
Affiliation(s)
- Mehmet Şahinbaş
- Department of Cardiology, Sivas Numune Hospital, Sivas, Turkey.
| | - İdris Buğra Çerik
- Department of Cardiology, Training and Research Hospital, Ordu University, Ordu, Turkey
| | - Duygu Yalınbaş Yeter
- Department of Ophthalmology, Faculty of Medicine, Cumhuriyet University, Sivas, Turkey
| |
Collapse
|
2
|
Tian C, Yang Y, Bai B, Wang S, Liu M, Sun RC, Yu T, Chu XM. Potential of exosomes as diagnostic biomarkers and therapeutic carriers for doxorubicin-induced cardiotoxicity. Int J Biol Sci 2021; 17:1328-1338. [PMID: 33867849 PMCID: PMC8040474 DOI: 10.7150/ijbs.58786] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/17/2021] [Indexed: 02/07/2023] Open
Abstract
Doxorubicin (DOX) is a kind of representative anthracyclines. It has greatly prolonged lifespan of cancer patients. However, a long course of DOX chemotherapy could induce various forms of deaths of cardiomyocytes, such as apoptosis, pyroptosis and ferroptosis, contributing to varieties of cardiac complications called cardiotoxicity. It has become a major concern considering the large number of cancer patients' worldwide and increased survival rates after chemotherapy. Exosomes, a subgroup of extracellular vesicles (EVs), are secreted by nearly all cells and consist of lipid bilayers, nucleic acids and proteins. They can serve as mediators between intercellular communication via the transfer of bioactive molecules from secretory to recipient cells, modulating multiple pathophysiological processes. It has been proven that exosomes in body fluids can serve as biomarkers for doxorubicin-induced cardiotoxicity (DIC). Moreover, exosomes have attracted considerable attention because of their capacity as carriers of certain proteins, genetic materials (miRNA and lncRNA), and chemotherapeutic drugs to decrease the dosage of DOX and alleviate cardiotoxicity. This review briefly describes the characteristics of exosomes and highlights their clinical application potential as diagnostic biomarkers and drug delivery vehicles for DIC, thus providing a strategy for addressing it based on exosomes.
Collapse
Affiliation(s)
- Chao Tian
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Yanyan Yang
- Department of Immunology, Basic Medicine School, Qingdao University, Qingdao 266071, China
| | - Baochen Bai
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Shizhong Wang
- Department of Cardiovascular Surgery, The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao 266000, China
| | - Meixin Liu
- Department of Cardiac Ultrasound, The Affiliated hospital of Qingdao University, Qingdao 266000, China
| | - Rui-Cong Sun
- Department of Cardiac Ultrasound, The Affiliated hospital of Qingdao University, Qingdao 266000, China
| | - Tao Yu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266021, China
- Department of Cardiac Ultrasound, The Affiliated hospital of Qingdao University, Qingdao 266000, China
| | - Xian-ming Chu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
- Department of Cardiology, The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao 266032, China
| |
Collapse
|
3
|
Sonaglioni A, Albini A, Fossile E, Pessi MA, Nicolosi GL, Lombardo M, Anzà C, Ambrosio G. Speckle-Tracking Echocardiography for Cardioncological Evaluation in Bevacizumab-Treated Colorectal Cancer Patients. Cardiovasc Toxicol 2020; 20:581-592. [PMID: 32519318 DOI: 10.1007/s12012-020-09583-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Angiogenesis inhibitor Bevacizumab (BVZ) may lead to the development of adverse effects, including hypertension and cardiac ischemia. Whether assessment of changes in myocardial strain by two-dimensional speckle-tracking echocardiography (2D-STE) can be of value in detecting BVZ-mediated cardiotoxicity at an earlier stage is not known. We investigated whether 2D-STE can non-invasively detect early evidence of cardiotoxicity in metastatic colorectal cancer (mCRC) patients treated with BVZ. Between January and June 2019, 25 consecutive patients (71.8 ± 7.5 year/old, 17 males) with mCRC were prospectively enrolled. Patients underwent physical examination, blood tests, and conventional 2D-transthoracic echocardiography implemented with 2D-STE analysis, at baseline and at 3 and 6 months following treatment with BVZ (15 mg/kg every 15 days) + 5-fluorouracil/folinic acid plus oxaliplatin (FOLFOX i.v.). At 6-month follow-up, we assessed occurrence of global longitudinal strain (GLS) impairment (> 15% decrease in GLS compared with baseline) as primary end-point and a new-onset systemic hypertension (secondary end-point). On average, GLS showed a progressive significant impairment after BVZ, from - 17.4 ± 3.2% at baseline to - 16 ± 2.9% (p = 0.003) at 6-month follow-up; > 15% decrease in GLS (primary end-point) was detected in 9 patients (36%). All other strain parameters remained unchanged. New-onset systemic hypertension (secondary end-point) was diagnosed in five patients (20%). No significant changes were observed in serial high-sensitivity cardiac troponin I measurements. No patient developed significant changes in LV size or LV ejection fraction; no case of clinically symptomatic HF was observed during BVZ-treatment. Measurement of GLS by 2D-STE analysis can effectively detect BVZ-mediated cardiotoxicity at an early stage.
Collapse
Affiliation(s)
- Andrea Sonaglioni
- Department of Cardiology, Ospedale San Giuseppe IRCCS MultiMedica, Milan, Italy
| | - Adriana Albini
- Scientific and Technological Pole, IRCCS MultiMedica, Milan, Italy.
| | - Emanuela Fossile
- Department of Oncology, Ospedale San Giuseppe IRCCS MultiMedica, Milan, Italy
| | | | | | - Michele Lombardo
- Department of Cardiology, Ospedale San Giuseppe IRCCS MultiMedica, Milan, Italy
| | - Claudio Anzà
- Cardiovascular Department, IRCCS MultiMedica, Sesto San Giovanni (MI), Italy
| | - Giuseppe Ambrosio
- Cardiology and Cardiovascular Pathophysiology, Azienda Ospedaliero-Universitaria "S. Maria Della Misericordia", Perugia, Italy
| |
Collapse
|
4
|
Rocca C, Pasqua T, Cerra MC, Angelone T. Cardiac Damage in Anthracyclines Therapy: Focus on Oxidative Stress and Inflammation. Antioxid Redox Signal 2020; 32:1081-1097. [PMID: 31928066 DOI: 10.1089/ars.2020.8016] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Despite their serious side effects, anthracyclines (ANTs) are the most prescribed chemotherapeutic drugs because of their strong efficacy in both solid and hematological tumors. A major limitation to ANTs clinical application is the severe cardiotoxicity observed both acutely and chronically. The mechanism underlying cardiac dysfunction under chemotherapy is mainly dependent on the generation of oxidative stress and systemic inflammation, both of which lead to progressive cardiomyopathy and heart failure. Recent Advances: Over the years, the iatrogenic ANTs-induced cardiotoxicity was believed to be simply given by iron metabolism and reactive oxygen species production; however, several experimental data indicate that ANTs may use alternative damaging mechanisms, such as topoisomerase 2β inhibition, inflammation, pyroptosis, immunometabolism, and autophagy. Critical Issues: In this review, we aimed at discussing ANTs-induced cardiac injury from different points of view, updating and focusing on oxidative stress and inflammation, since these pathways are not exclusive or independent from each other but they together importantly contribute to the complexity of ANTs-induced multifactorial cardiotoxicity. Future Directions: A deeper understanding of the mechanistic signaling leading to ANTs side effects could reveal crucial targeting molecules, thus representing strategic knowledge to promote better therapeutic efficacy and lower cardiotoxicity during clinical application.
Collapse
Affiliation(s)
- Carmine Rocca
- Laboratory of Cellular and Molecular Cardiovascular Physiology, Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
| | - Teresa Pasqua
- Laboratory of Cellular and Molecular Cardiovascular Physiology, Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
| | - Maria Carmela Cerra
- Laboratory of Organ and System Physiology, Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy.,National Institute of Cardiovascular Research (INRC), Bologna, Italy
| | - Tommaso Angelone
- Laboratory of Cellular and Molecular Cardiovascular Physiology, Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy.,National Institute of Cardiovascular Research (INRC), Bologna, Italy
| |
Collapse
|
5
|
Nhola LF, Abdelmoneim SS, Villarraga HR, Kohli M, Grothey A, Bordun KA, Cheung M, Best R, Cheung D, Huang R, Barros-Gomes S, Pitz M, Singal PK, Jassal DS, Mulvagh SL. Echocardiographic Assessment for the Detection of Cardiotoxicity Due to Vascular Endothelial Growth Factor Inhibitor Therapy in Metastatic Renal Cell and Colorectal Cancers. J Am Soc Echocardiogr 2018; 32:267-276. [PMID: 30459123 DOI: 10.1016/j.echo.2018.09.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Cardio-oncology is a recently established discipline that focuses on the management of patients with cancer who are at risk for developing cardiovascular complications as a result of their underlying oncologic treatment. In metastatic colorectal cancer (mCRC) and metastatic renal cell carcinoma (mRCC), vascular endothelial growth factor inhibitor (VEGF-i) therapy is commonly used to improve overall survival. Although these novel anticancer drugs may lead to the development of cardiotoxicity, whether early detection of cardiac dysfunction using serial echocardiography could potentially prevent the development of heart failure in this patient population requires further study. The aim of this study was to investigate the role of two-dimensional speckle-tracking echocardiography in the detection of cardiotoxicity due to VEGF-i therapy in patients with mCRC or mRCC. METHODS Patients with mRCC or mCRC were evaluated using serial echocardiography at baseline and 1, 3, and 6 months following VEGF-i treatment. RESULTS A total of 40 patients (34 men; mean age, 63 ± 9 years) receiving VEGF-i therapy were prospectively recruited at two academic centers: 26 (65%) were receiving sunitinib, eight (20%) pazopanib, and six (15%) bevacizumab. The following observations were made: (1) 8% of patients developed clinically asymptomatic cancer therapeutics-related cardiac dysfunction; (2) 30% of patients developed clinically significant decreases in global longitudinal strain, a marker for early subclinical cardiac dysfunction; (3) baseline abnormalities in global longitudinal strain may identify a subset of patients at higher risk for developing cancer therapeutics-related cardiac dysfunction; and (4) new or worsening hypertension was the most common adverse cardiovascular event, afflicting nearly one third of the study population. CONCLUSIONS Cardiac dysfunction defined by serial changes in myocardial strain assessed using two-dimensional speckle-tracking echocardiography occurs in patients undergoing treatment with VEGF-i for mCRC or mRCC, which may provide an opportunity for preventive interventions.
Collapse
Affiliation(s)
- Lara F Nhola
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Sahar S Abdelmoneim
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota; Division of Cardiology, Orman Heart Center, Assiut University, Assiut, Egypt
| | | | - Manish Kohli
- Department of Oncology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Axel Grothey
- Department of Oncology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Kimberly-Ann Bordun
- Institute of Cardiovascular Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Matthew Cheung
- Institute of Cardiovascular Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ryan Best
- Institute of Cardiovascular Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - David Cheung
- Institute of Cardiovascular Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Runqing Huang
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | | | - Marshall Pitz
- Section of Hematology/Oncology, Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Pawan K Singal
- Institute of Cardiovascular Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Davinder S Jassal
- Institute of Cardiovascular Sciences, University of Manitoba, Winnipeg, Manitoba, Canada; Section of Hematology/Oncology, Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada; Section of Cardiology, Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Sharon L Mulvagh
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota; Division of Cardiology, Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada.
| |
Collapse
|
6
|
Electrochemical, spectroscopic and theoretical monitoring of anthracyclines' interactions with DNA and ascorbic acid by adopting two routes: Cancer cell line studies. PLoS One 2018; 13:e0205764. [PMID: 30372448 PMCID: PMC6205586 DOI: 10.1371/journal.pone.0205764] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 10/01/2018] [Indexed: 01/08/2023] Open
Abstract
Pharmacodynamic interactions of three anthracycline antibiotics namely doxorubicin (DXH), epirubicin (EpiDXH) and daunorubicin (DNR) with DNA in the absence and presence of ascorbic acid (AA) as natural additive were monitored under physiological conditions (pH = 7.4, 4.7 and T = 309.5K). Route–1 (Anthracycline–AA–DNA) and Route–2 (Anthracycline–DNA–AA) were adopted to see the interactional behavior by cyclic voltammetry (CV) and UV-visible spectroscopy. In comparison to Route–2; voltammetric and spectral responses as well as binding constant (Kb) and Gibb’s free energy change (ΔG) values revealed strongest and more favorable interaction of anthracycline–AA complex with DNA via Route–1. Kb, s (binding site sizes) and ΔG evaluated from experimental (CV, UV-Vis) and theoretical (molecular docking) findings showed enhanced binding strength of tertiary complexes as compared to binary drug–DNA complexes. The results were found comparatively better at pH 7.4. Consistency was observed in binding parameters evaluated from experimental and theoretical techniques. Diffusion coefficients (Do) and heterogeneous electron transfer rate constant (ks,h) confirmed the formation of complexes via slow diffusion kinetics. Percent cell inhibition (%Cinh) of anthracyclines for non-small cell cancer cell lines (NSCCLs) H-1299 and H-157 were evaluated higher in the presence of AA which further complimented experimental and theoretical results.
Collapse
|
7
|
Dorsch LM, Schuldt M, Knežević D, Wiersma M, Kuster DWD, van der Velden J, Brundel BJJM. Untying the knot: protein quality control in inherited cardiomyopathies. Pflugers Arch 2018; 471:795-806. [PMID: 30109411 PMCID: PMC6475634 DOI: 10.1007/s00424-018-2194-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 08/06/2018] [Indexed: 12/27/2022]
Abstract
Mutations in genes encoding sarcomeric proteins are the most important causes of inherited cardiomyopathies, which are a major cause of mortality and morbidity worldwide. Although genetic screening procedures for early disease detection have been improved significantly, treatment to prevent or delay mutation-induced cardiac disease onset is lacking. Recent findings indicate that loss of protein quality control (PQC) is a central factor in the disease pathology leading to derailment of cellular protein homeostasis. Loss of PQC includes impairment of heat shock proteins, the ubiquitin-proteasome system, and autophagy. This may result in accumulation of misfolded and aggregation-prone mutant proteins, loss of sarcomeric and cytoskeletal proteins, and, ultimately, loss of cardiac function. PQC derailment can be a direct effect of the mutation-induced activation, a compensatory mechanism due to mutation-induced cellular dysfunction or a consequence of the simultaneous occurrence of the mutation and a secondary hit. In this review, we discuss recent mechanistic findings on the role of proteostasis derailment in inherited cardiomyopathies, with special focus on sarcomeric gene mutations and possible therapeutic applications.
Collapse
Affiliation(s)
- Larissa M Dorsch
- Amsterdam UMC, Department of Physiology, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, O2 building 11W53, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands.
| | - Maike Schuldt
- Amsterdam UMC, Department of Physiology, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, O2 building 11W53, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands.
| | - Dora Knežević
- Amsterdam UMC, Department of Physiology, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, O2 building 11W53, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| | - Marit Wiersma
- Amsterdam UMC, Department of Physiology, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, O2 building 11W53, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| | - Diederik W D Kuster
- Amsterdam UMC, Department of Physiology, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, O2 building 11W53, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| | - Jolanda van der Velden
- Amsterdam UMC, Department of Physiology, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, O2 building 11W53, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| | - Bianca J J M Brundel
- Amsterdam UMC, Department of Physiology, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, O2 building 11W53, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| |
Collapse
|
8
|
Silva dos Santos D, Brasil GV, Ramos IPR, Mesquita FCP, Kasai-Brunswick TH, Christie MLA, Cahli GM, Barbosa RAQ, da Cunha ST, Pereira JX, Medei E, Campos de Carvalho AC, Carvalho AB, Goldenberg RCDS. Embryonic stem cell-derived cardiomyocytes for the treatment of doxorubicin-induced cardiomyopathy. Stem Cell Res Ther 2018; 9:30. [PMID: 29402309 PMCID: PMC5799903 DOI: 10.1186/s13287-018-0788-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 01/19/2018] [Accepted: 01/23/2018] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Doxorubicin (Dox) is a chemotherapy drug with limited application due to cardiotoxicity that may progress to heart failure. This study aims to evaluate the role of cardiomyocytes derived from mouse embryonic stem cells (CM-mESCs) in the treatment of Dox-induced cardiomyopathy (DIC) in mice. METHODS The mouse embryonic stem cell (mESC) line E14TG2A was characterized by karyotype analysis, gene expression using RT-PCR and immunofluorescence. Cells were transduced with luciferase 2 and submitted to cardiac differentiation. Total conditioned medium (TCM) from the CM-mESCs was collected for proteomic analysis. To establish DIC in CD1 mice, Dox (7.5 mg/kg) was administered once a week for 3 weeks, resulting in a cumulative Dox dose of 22.5 mg/kg. At the fourth week, a group of animals was injected intramyocardially with CM-mESCs (8 × 105 cells). Cells were tracked by a bioluminescence assay, and the body weight, echocardiogram, electrocardiogram and number of apoptotic cardiomyocytes were evaluated. RESULTS mESCs exhibited a normal karyotype and expressed pluripotent markers. Proteomic analysis of TCM showed proteins related to the negative regulation of cell death. CM-mESCs presented ventricular action potential characteristics. Mice that received Dox developed heart failure and showed significant differences in body weight, ejection fraction (EF), end-systolic volume (ESV), stroke volume (SV), heart rate and QT and corrected QT (QTc) intervals when compared to the control group. After cell or placebo injection, the Dox + CM-mESC group showed significant increases in EF and SV when compared to the Dox + placebo group. Reduction in ESV and QT and QTc intervals in Dox + CM-mESC-treated mice was observed at 5 or 30 days after cell treatment. Cells were detected up to 11 days after injection. The Dox + CM-mESC group showed a significant reduction in the percentage of apoptotic cardiomyocytes in the hearts of mice when compared to the Dox + placebo group. CONCLUSIONS CM-mESC transplantation improves cardiac function in mice with DIC.
Collapse
Affiliation(s)
- Danúbia Silva dos Santos
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
| | - Guilherme Visconde Brasil
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
| | - Isalira Peroba Rezende Ramos
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
- Centro Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Bloco M, Rio de Janeiro, RJ 21941-902 Brazil
| | - Fernanda Cristina Paccola Mesquita
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
| | - Tais Hanae Kasai-Brunswick
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
- Centro Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Bloco M, Rio de Janeiro, RJ 21941-902 Brazil
| | - Michelle Lopes Araújo Christie
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
| | - Gustavo Monnerat Cahli
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
| | - Raiana Andrade Quintanilha Barbosa
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
| | - Sandro Torrentes da Cunha
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
| | - Jonathas Xavier Pereira
- Departamento de Patologia—Faculdade de Medicina, Hospital Universitário Clementino Fraga Filho, Universiade Federal do Rio de Janeiro, Av. Rodolpho Paulo Rocco, 255, Sub-solo, SAP, Rio de Janeiro, RJ 21910-590 Brazil
| | - Emiliano Medei
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
- Centro Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Bloco M, Rio de Janeiro, RJ 21941-902 Brazil
- Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Av. Carlos Chagas Filho 373, Rio de Janeiro, RJ 21941-902 Brazil
| | - Antonio Carlos Campos de Carvalho
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
- Centro Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Bloco M, Rio de Janeiro, RJ 21941-902 Brazil
- Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Av. Carlos Chagas Filho 373, Rio de Janeiro, RJ 21941-902 Brazil
| | - Adriana Bastos Carvalho
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
- Centro Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Bloco M, Rio de Janeiro, RJ 21941-902 Brazil
- Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Av. Carlos Chagas Filho 373, Rio de Janeiro, RJ 21941-902 Brazil
| | - Regina Coeli dos Santos Goldenberg
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
- Centro Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Bloco M, Rio de Janeiro, RJ 21941-902 Brazil
- Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Av. Carlos Chagas Filho 373, Rio de Janeiro, RJ 21941-902 Brazil
| |
Collapse
|
9
|
van Rossum AGJ, Kok M, McCool D, Opdam M, Miltenburg NC, Mandjes IAM, van Leeuwen-Stok E, Imholz ALT, Portielje JEA, Bos MMEM, van Bochove A, van Werkhoven E, Schmidt MK, Oosterkamp HM, Linn SC. Independent replication of polymorphisms predicting toxicity in breast cancer patients randomized between dose-dense and docetaxel-containing adjuvant chemotherapy. Oncotarget 2017; 8:113531-113542. [PMID: 29371927 PMCID: PMC5768344 DOI: 10.18632/oncotarget.22697] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 10/27/2017] [Indexed: 12/12/2022] Open
Abstract
Introduction Although pharmacogenomics has evolved substantially, a predictive test for chemotherapy toxicity is still lacking. We compared the toxicity of adjuvant dose-dense doxorubicin-cyclophosphamide (ddAC) and docetaxel-doxorubicin-cyclophosphamide (TAC) in a randomized multicenter phase III trial and replicated previously reported associations between genotypes and toxicity. Results 646 patients (97%) were evaluable for toxicity (grade 2 and higher). Whereas AN was more frequent after ddAC (P < 0.001), TAC treated patients more often had PNP (P < 0.001). We could replicate 2 previously reported associations: TECTA (rs1829; OR 4.18, 95% CI 1.84-9.51, P = 0.001) with PNP, and GSTP1 (rs1138272; OR 2.04, 95% CI 1.13-3.68, P = 0.018) with PNP. Materials and methods Patients with pT1-3, pN0-3 breast cancer were randomized between six cycles A60C600 every 2 weeks or T75A50C500 every 3 weeks. Associations of 13 previously reported single nucleotide polymorphisms (SNPs) with the most frequent toxicities: anemia (AN), febrile neutropenia (FN) and peripheral neuropathy (PNP) were analyzed using logistic regression models. Conclusions In this independent replication, we could replicate an association between 2 out of 13 SNPs and chemotherapy toxicities. These results warrant further validation in order to enable tailored treatment for breast cancer patients.
Collapse
Affiliation(s)
- Annelot G J van Rossum
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Marleen Kok
- Division of Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Danielle McCool
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Mark Opdam
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Nienke C Miltenburg
- Department of Neurology, Medical Center Slotervaart, Amsterdam, The Netherlands
| | | | | | - Alex L T Imholz
- Department of Medical Oncology, Deventer Ziekenhuis, Deventer, The Netherlands
| | | | - Monique M E M Bos
- Department of Medical Oncology, Reinier de Graaf Groep, Delft, The Netherlands
| | - Aart van Bochove
- Department of Medical Oncology, Zaans Medisch Centrum, Zaandam, The Netherlands
| | - Erik van Werkhoven
- Biometrics Division, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Marjanka K Schmidt
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Hendrika M Oosterkamp
- Department of Medical Oncology, Haaglanden Medisch Centrum, The Hague, The Netherlands
| | - Sabine C Linn
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Department of Pathology, University Medical Center, Utrecht, The Netherlands
| |
Collapse
|
10
|
Markman TM, Ruble K, Loeb D, Chen A, Zhang Y, Beasley GS, Thompson WR, Nazarian S. Electrophysiological effects of anthracyclines in adult survivors of pediatric malignancy. Pediatr Blood Cancer 2017; 64. [PMID: 28453898 DOI: 10.1002/pbc.26556] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 02/25/2017] [Accepted: 02/27/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND Anthracycline use is limited by cardiotoxicity, including arrhythmias and left ventricular (LV) dysfunction. We aim to characterize the association between electrophysiological changes and LV dysfunction. METHODS A retrospective chart review was conducted, including all 147 pediatric cancer survivors at our institution over 18 years of age and treated with an anthracycline. One hundred thirty-four patients who had at least one electrocardiogram (ECG) and echocardiogram were analyzed. The association between dysfunction and baseline characteristics, treatment history, and electrocardigraphic parameters were analyzed using multivariable logistic regression. Additionally, a longitudinal generalized estimating equation (GEE) model was used to examine the temporal association between repeated measure corrected QT (QTc) intervals and subsequent LV function. RESULTS In our population, 24% of patients had LV dysfunction. The initial posttreatment QTc interval was longer in patients with LV dysfunction (438 ± 35 vs. 420 ± 20 msec, P = 0.002). In logistic regression analysis, QTc interval (P < 0.001) and cumulative radiation dose (P = 0.027) were associated with LV dysfunction. On ECGs performed prior to evidence of LV dysfunction, the QTc was longer than on ECGs preceding a normal echocardiogram (451 ± 32 msec vs. 423 ± 25 msec, P < 0.001). Mean time from QTc ≥ 450 msec to evidence of LV dysfunction was 1.8 ± 2.9 years. In the longitudinal GEE model, QTc prolongation was associated with subsequent decreased fractional shortening. CONCLUSIONS Among adult survivors of pediatric cancer treated with anthracyclines, prolongation of the QTc interval was associated with subsequent LV dysfunction.
Collapse
Affiliation(s)
- Timothy M Markman
- Department of Medicine, The Johns Hopkins University, Baltimore, Maryland
| | - Kathryn Ruble
- Department of Pediatrics, The Johns Hopkins University, Baltimore, Maryland.,Division of Pediatric Oncology, The Johns Hopkins University, Baltimore, Maryland
| | - David Loeb
- Department of Pediatrics, The Johns Hopkins University, Baltimore, Maryland.,Division of Pediatric Oncology, The Johns Hopkins University, Baltimore, Maryland
| | - Allen Chen
- Department of Pediatrics, The Johns Hopkins University, Baltimore, Maryland.,Division of Pediatric Oncology, The Johns Hopkins University, Baltimore, Maryland
| | - Yiyi Zhang
- Department of Epidemiology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Gary S Beasley
- Department of Pediatrics, The Johns Hopkins University, Baltimore, Maryland.,Division of Pediatric Cardiology, The Johns Hopkins University, Baltimore, Maryland
| | - W Reid Thompson
- Department of Pediatrics, The Johns Hopkins University, Baltimore, Maryland.,Division of Pediatric Cardiology, The Johns Hopkins University, Baltimore, Maryland
| | - Saman Nazarian
- Department of Medicine, The Johns Hopkins University, Baltimore, Maryland.,Department of Epidemiology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland.,Division of Cardiology, Section for Cardiac Electrophysiology, The Johns Hopkins University, Baltimore, Maryland.,Division of Cardiology, Section for Cardiac Electrophysiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
11
|
Ahn HS, Lee DH, Kim TJ, Shin HC, Jeon HK. Cardioprotective Effects of a Phlorotannin Extract Against Doxorubicin-Induced Cardiotoxicity in a Rat Model. J Med Food 2017; 20:944-950. [PMID: 28816580 DOI: 10.1089/jmf.2017.3919] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Long-term therapy with doxorubicin (DOX) is associated with high incidence of cumulative and irreversible dilated cardiomyopathy. The goal of this study was to evaluate the cardioprotective effects and safety of a phlorotannin extract from a brown algae Ecklonia cava (Seapolynol™, SPN) against DOX-induced cardiotoxicity in a rat model. A total of 42 rats were divided into six groups: control, low-dose SPN (LDS), high-dose SPN (HDS), DOX, DOX with low-dose SPN (DOX+LDS), and DOX with high-dose SPN (DOX+HDS). Echocardiography was performed at baseline and after 6 weeks. In left ventricular (LV) ejection fraction, DOX and DOX+LDS groups showed significant decreases (P < .001), while LDS, HDS, and DOX+HDS groups showed no significant change compared with control group. In LV mass index, DOX and DOX+LDS groups showed significant increases (P < .001 and P = .013), while LDS, HDS, and DOX+HDS groups showed no significant change compared with control group. In electron microscopy of the LV wall tissue, DOX+HDS group showed markedly less impaired myofibrils and mitochondria compared with both DOX and DOX+LDS groups. On the findings in echocardiography and electron microscopy, 6-week oral administration of SPN was safe and cardioprotective in a DOX-induced rat cardiotoxicity model in a dose-dependent manner.
Collapse
Affiliation(s)
- Hyo-Suk Ahn
- 1 Division of Cardiology, Department of Internal Medicine, The Catholic University of Korea, Uijeongbu St. Mary's Hospital , Uijeongbu, Korea
| | - Dong-Hyeon Lee
- 2 Division of Cardiology, Department of Internal Medicine, The Catholic University of Korea, Seoul St. Mary's Hospital , Seoul, Korea
| | - Tae-Jung Kim
- 3 Department of Hospital Pathology, College of Medicine, The Catholic University of Korea , Seoul, Korea
| | - Hyeon-Cheol Shin
- 4 CEWIT Center for Systems Biology, The State University of New York , Incheon, Korea
| | - Hui-Kyung Jeon
- 1 Division of Cardiology, Department of Internal Medicine, The Catholic University of Korea, Uijeongbu St. Mary's Hospital , Uijeongbu, Korea
| |
Collapse
|
12
|
Spewak MB, Williamson RS, Mertens AC, Border WL, Meacham LR, Wasilewski-Masker KJ. Yield of screening echocardiograms during pediatric follow-up in survivors treated with anthracyclines and cardiotoxic radiation. Pediatr Blood Cancer 2017; 64. [PMID: 27966803 DOI: 10.1002/pbc.26367] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 10/25/2016] [Accepted: 10/26/2016] [Indexed: 02/05/2023]
Abstract
BACKGROUND Guidelines published by the Children's Oncology Group recommend screening echocardiograms for childhood cancer survivors exposed to anthracyclines and/or cardiotoxic radiation. This study aims to assess risk factors for cardiac late effects while evaluating the overall yield of screening echocardiograms. PROCEDURE Demographics, exposures, and echocardiogram results were abstracted from the medical records of survivors diagnosed at ≤ 21 years old and ≥ 2 years off therapy who were exposed to anthracyclines and/or potentially cardiotoxic radiotherapy. Descriptive statistics and logistic regressions were performed and the yield of screening echocardiograms was calculated. RESULTS Of 853 patients, 1,728 screening echocardiograms were performed, and 37 patients had an abnormal echocardiogram (overall yield 2.1%). Yields were only somewhat higher in more frequently screened patients. Risk factors for an abnormal result included anthracycline dose of ≥300 mg/m2 (adjusted odds ratio [aOR] 3.1; 95% confidence interval [CI]: 1.3-7.2; P < 0.01) with a synergist relationship in patients who also received radiation doses ≥30 Gy (aOR 7.0; 95% CI: 1.6-31.9; P = 0.01), as well as autologous bone marrow transplant (OR 3.3; 95% CI: 1.3-8.5; P = 0.01). Sex, race, age at diagnosis, and cyclophosphamide exposure were not statistically significant risk factors, and no patient receiving <100 mg/m2 anthracycline dose without concomitant radiation had an abnormal echocardiogram. CONCLUSIONS Dose-dependent and synergist anthracycline and cardiotoxic radiotherapy risks for developing cardiomyopathy were confirmed. However, previously identified risk factors including female sex, black race, and early age at diagnosis were not replicated in this cohort. The yields showed weak correlation across frequency categories. Echocardiographic screening recommendations for low-risk pediatric patients may warrant re-evaluation.
Collapse
Affiliation(s)
- Michael B Spewak
- Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Rebecca S Williamson
- The Aflac Cancer & Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia
| | - Ann C Mertens
- Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.,The Aflac Cancer & Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia
| | - William L Border
- Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.,The Sibley Heart Center, Children's Healthcare of Atlanta, Atlanta, Georgia
| | - Lillian R Meacham
- Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.,The Aflac Cancer & Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia
| | - Karen J Wasilewski-Masker
- Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.,The Aflac Cancer & Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia
| |
Collapse
|
13
|
Tromp J, Steggink LC, Van Veldhuisen DJ, Gietema JA, van der Meer P. Cardio-Oncology: Progress in Diagnosis and Treatment of Cardiac Dysfunction. Clin Pharmacol Ther 2017; 101:481-490. [PMID: 28073142 DOI: 10.1002/cpt.614] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 12/19/2016] [Accepted: 01/06/2017] [Indexed: 12/11/2022]
Affiliation(s)
- J Tromp
- Department of Cardiology, University Medical Centre Groningen, University of Groningen, The Netherlands
| | - L C Steggink
- Department of Medical Oncology University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - D J Van Veldhuisen
- Department of Cardiology, University Medical Centre Groningen, University of Groningen, The Netherlands
| | - J A Gietema
- Department of Medical Oncology University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - P van der Meer
- Department of Cardiology, University Medical Centre Groningen, University of Groningen, The Netherlands
| |
Collapse
|
14
|
Jannazzo A, Hoffman J, Lutz M. Monitoring of Anthracycline-Induced Cardiotoxicity. Ann Pharmacother 2016; 42:99-104. [PMID: 18094345 DOI: 10.1345/aph.1k359] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Objective: To review the literature regarding the appropriate monitoring for anthracycline-induced cardiotoxicity. Data Sources: A MEDLINE search of the literature was performed (1966–August 2007). Search terms included anthracycline, cardiotoxicity, and monitoring. Additional references were identified through bibliographic reviews. Data Synthesis: Anthracycline medications are effective in the treatment of many malignancies but their use is limited by their associated cardiotoxicity. The focus of anthracycline-induced cardiotoxicity prevention has been on monitoring cardiac function during treatment; however, a consensus on the most appropriate way to monitor patients is not available. Most guidelines lack specific details on the appropriate methods of cardiac evaluation and schedule. One guideline that does provide specific recommendations on both the method of evaluation and schedule has been criticized for being too restrictive, costly, and tacking in evidentiary support. The literature is insufficient in evaluation of the predictive value of cardiac function monitoring by echocardiography or radionuclide angiography during anthracycline therapy and the future development of cardiotoxicity, the necessity of baseline cardiac function monitoring, the optimal follow-up cardiac evaluation schedule, and the addition of risk stratification to monitoring schemes. Conclusions: Although guidelines are Inadequate to predict and prevent anthracycline-induced cardiotoxicity, until further research is available, following one of the existing guidelines to monitor for this adverse effect is a practical solution.
Collapse
Affiliation(s)
- Abigail Jannazzo
- Pharmacy Practice Resident, Department of Pharmaceutical Services, William Beaumont Hospital, Royal Oak, Ml; now, Oncology Specialty Resident, Evanston Northwestern Healthcare, Evanston, IL
| | - Janet Hoffman
- Drug Information Specialist, Department of Pharmaceutical Services, William Beaumont Hospital
| | - Mark Lutz
- Drug Information Specialist, Department of Pharmaceutical Services, William Beaumont Hospital
| |
Collapse
|
15
|
Inanc MT, Karadavut S, Aytekin M, Duran AO, Derya M, Akpek M, Sahin O, Kalay N, Karaca H, Ozkan M, Inanc M. The relationship between plasma hyaluronan levels and anthracycline-related cardiotoxicity in breast cancer patients. Int J Cardiol 2016; 218:246-251. [PMID: 27236123 DOI: 10.1016/j.ijcard.2016.05.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 05/12/2016] [Indexed: 11/15/2022]
Abstract
AIM Anthracycline-derived antineoplastic agents are used as the main form of treatment in many malignant diseases, including breast cancer and childhood cancers. Cardiotoxicity is one of the most feared life-threatening complications of cancer therapy. In the present study, we aimed to investigate the relationship between plasma hyaluronan (HA) levels and anthracycline-induced cardiotoxicity. MATERIALS AND METHODS Fifty eight of 73 female patients who were diagnosed with breast cancer and treated with a chemotherapy regimen including anthracycline were enrolled in this study. Anamneses were taken from each patient before and after chemotherapy. Further, physical examinations, electrocardiography, and transthoracic echocardiography were performed, and plasma hyaluronan levels were determined by using ELISA assay for each patient before and after treatment. RESULTS Following anthracycline-based chemotherapy, the average left ventricular ejection fraction decreased (62.6±3.7% vs. 58.6±4.4%, p<0.001), and diastolic functions significantly deteriorated (p<0.001). However, troponin and hyaluronan levels significantly increased following chemotherapy [Troponin (ng/ml, mean±SD): before 0.01±0.002, after 0.037±0.02, p<0.001], [Plasma HA (ng/ml, mean±SD): before 41.3±5.4, after 70±8.5, p<0.001]. The increase in troponin values correlated with systolic dysfunction (p=0.002), but did not correlate with diastolic dysfunction (p=0.661). Significant correlations were found between systolic/diastolic dysfunction and plasma HA levels (r=0.417, p=0.001; r=0.339, p=0.009, respectively). CONCLUSIONS Both systolic and diastolic functions were significantly deteriorated after chemotherapy. In addition, plasma levels of HA and troponin increased after treatment. Further, both systolic and diastolic dysfunctions were found to correlate with serum HA levels. All these data suggest that HA might have a function on anthracycline-induced cardiotoxicity.
Collapse
Affiliation(s)
| | | | - Metin Aytekin
- Erciyes University Medical Biology Department, Kayseri, Turkey
| | - Ayse Ocak Duran
- Erciyes University Medical Oncology Department, Kayseri, Turkey
| | - Muazzez Derya
- Erciyes University Medical Biology Department, Kayseri, Turkey
| | - Mahmut Akpek
- Kayseri Training and Research Hospital Cardiology Department, Kayseri, Turkey
| | - Omer Sahin
- Kayseri Training and Research Hospital Cardiology Department, Kayseri, Turkey
| | - Nihat Kalay
- Erciyes University Cardiology Department, Kayseri, Turkey
| | - Halit Karaca
- Erciyes University Medical Oncology Department, Kayseri, Turkey
| | - Metin Ozkan
- Erciyes University Medical Oncology Department, Kayseri, Turkey
| | - Mevlude Inanc
- Kayseri Training and Research Hospital Medical Oncology Department, Kayseri, Turkey.
| |
Collapse
|
16
|
Zito C, Longobardo L, Cadeddu C, Monte I, Novo G, Dell'Oglio S, Pepe A, Madonna R, Tocchetti CG, Mele D. Cardiovascular imaging in the diagnosis and monitoring of cardiotoxicity: role of echocardiography. J Cardiovasc Med (Hagerstown) 2016; 17 Suppl 1:e35-e44. [PMID: 27183524 DOI: 10.2459/jcm.0000000000000374] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The evaluation by cardiovascular imaging of chemotherapy patients became a central topic in the last several years. The use of drugs for the treatment of cancers increased, and new molecules and protocols were developed to improve outcomes in these patients. Although, these novel approaches also produced a progressive increase in side effects, particularly myocardial dysfunction. Imaging of the heart was highly accurate in the early diagnosis of cancer therapeutics related-cardiac dysfunction. Echocardiography is the first-line method to assess ventricular function alterations, and it is required to satisfy the need for an early, easy and accurate diagnosis to stratify the risk of heart failure and manage treatments. A careful monitoring of cardiac function during the course of therapy should prevent the onset of severe heart impairment. This review provides an overview of the most important findings of the role of echocardiography in the management of chemotherapy-treated patients to create a clear and complete description of the efficacy of conventional measurements, the importance of comprehensive heart evaluations, the additional role of new echocardiographic techniques, the utility of integrated studies using other imaging tools and the positions of the most important international societies on this topic.
Collapse
Affiliation(s)
- Concetta Zito
- aDepartment of Clinical and Experimental Medicine, Section of Cardiology, University of Messina, Messina bDepartment of Medical Sciences 'Mario Aresu', University of Cagliari, Cagliari cDepartment of General Surgery and Medical-Surgery Specialties, Section of Cardiology, University of Catania, Catania dChair and Division of Cardiology, University of Palermo, Palermo eU.O.C. Magnetic Resonance Imaging, Fondazione G. Monasterio C.N.R., Pisa fInstitute of Cardiology, Center of Excellence on Aging, 'G. d'Annunzio' University, Chieti gDipartimento di Scienze Mediche Traslazionali, Universita' degli Studi di Napoli Federico II hCardiology Unit, University Hospital of Ferrara, Ferrara, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Mukku RB, Fonarow GC, Watson KE, Ajijola OA, Depasquale EC, Nsair A, Baas AS, Deng MC, Yang EH. Heart Failure Therapies for End-Stage Chemotherapy-Induced Cardiomyopathy. J Card Fail 2016; 22:439-48. [PMID: 27109619 DOI: 10.1016/j.cardfail.2016.04.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 04/13/2016] [Accepted: 04/15/2016] [Indexed: 02/06/2023]
Abstract
With ongoing advancements in cancer-related treatments, the number of cancer survivors continues to grow globally, with numbers in the United States predicted to reach 18 million by 2020. As a result, it is expected that a greater number of patients will present with chemotherapy-related side effects. One entity in particular, chemotherapy-related cardiomyopathy (CCMP), is a known cardiotoxic manifestation associated with agents such as anthracyclines, trastuzumab, and tyrosine kinase inhibitors. Although such effects have been described in the medical literature for decades, concrete strategies for screening, prevention, and management of CCMP continue to be elusive owing to limited studies. Late recognition of CCMP is associated with a poorer prognosis, including a lack of clinical response to pharmacologic therapy, and end-stage heart failure. A number of advanced cardiac therapies, including cardiac resynchronization therapy, ventricular assist devices, and orthotopic cardiac transplantation, are available to for end-stage heart failure; however, the role of these therapies in CCMP is unclear. In this review, management of end-stage CCMP with the use of advanced therapies and their respective effectiveness are discussed, as well as clinical characteristics of patients undergoing these treatments. The relative paucity of data in this field highlights the importance and need for larger-scale longitudinal studies and long-term registries tracking the outcomes of cancer survivors who have received cardiotoxic cancer therapy to determine the overall incidence of end-stage CCMP, as well as prognostic factors that will ultimately guide such patients toward receiving appropriate end-stage care.
Collapse
Affiliation(s)
- Roy B Mukku
- Division of Hospital Medicine, Department of Medicine, University of California, Los Angeles, California
| | - Gregg C Fonarow
- Ahmanson-UCLA Cardiomyopathy Center, Division of Cardiology, Department of Medicine, University of California, Los Angeles, California
| | - Karol E Watson
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, California
| | - Olujimi A Ajijola
- UCLA Cardiac Arrhythmia Center, Division of Cardiology, Department of Medicine, University of California, Los Angeles, California
| | - Eugene C Depasquale
- Ahmanson-UCLA Cardiomyopathy Center, Division of Cardiology, Department of Medicine, University of California, Los Angeles, California
| | - Ali Nsair
- Ahmanson-UCLA Cardiomyopathy Center, Division of Cardiology, Department of Medicine, University of California, Los Angeles, California
| | - Arnold S Baas
- Ahmanson-UCLA Cardiomyopathy Center, Division of Cardiology, Department of Medicine, University of California, Los Angeles, California
| | - Mario C Deng
- Ahmanson-UCLA Cardiomyopathy Center, Division of Cardiology, Department of Medicine, University of California, Los Angeles, California
| | - Eric H Yang
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, California.
| |
Collapse
|
18
|
van der Meer P, Gietema JA, Suter TM, van Veldhuisen DJ. Cardiotoxicity of breast cancer treatment: no easy solution for an important long-term problem. Eur Heart J 2016; 37:1681-3. [PMID: 27055813 DOI: 10.1093/eurheartj/ehw133] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Peter van der Meer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jourik A Gietema
- Department of Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Thomas M Suter
- Department of Cardiology, Bern University Hospital, Bern, Switzerland
| | - Dirk J van Veldhuisen
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
19
|
Tan TC, Neilan TG, Francis S, Plana JC, Scherrer-Crosbie M. Anthracycline-Induced Cardiomyopathy in Adults. Compr Physiol 2016; 5:1517-40. [PMID: 26140726 DOI: 10.1002/cphy.c140059] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Anthracyclines are one of the most commonly used antineoplastic agent classes, and a core part of the treatment in breast cancers, hematological malignancies, and sarcomas. Their benefit is decreased by their well-recognized cardiotoxicity. The purpose of this review is to outline the presentation, mechanisms, diagnosis, and treatment of anthracyclines-induced cardiotoxicity. Symptomatic heart failure occurs in 2% to 5% of patients treated with anthrayclines and may be higher in older patients or patients with cardiovascular risk factors. The mechanisms involved in anthracycline-induced cardiotoxicity involve myocyte loss by apoptosis in the presence of a limited regenerative capacity. Once symptomatic, anthracycline-induced cardiotoxicity is associated with markedly decreased survival. Left ventricular ejection fraction (LVEF), mostly determined using echocardiography, is used to monitor patients treated with anthracyclines. As more than 1/3 of patients treated with anthracyclines do not recover their baseline LVEF once it is decreased, more sensitive echocardiographic indices of LV function such as myocardial deformation or biomarkers have been studied in patients monitoring. Cardioprotective treatments such as angiotensin-converting enzyme inhibitors, beta-blockers, iron chelators, statins, and metformin are also the topic of research efforts.
Collapse
Affiliation(s)
- Timothy C Tan
- Cardiac Ultrasound Laboratory, Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Division of Cardiology, Blacktown Hospital, University of Western Sydney, Australia
| | - Tomas G Neilan
- Cardio-oncology program, Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Cardiac MR PET CT Program, Division of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sanjeev Francis
- Cardio-oncology program, Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Cardiac MR PET CT Program, Division of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Juan Carlos Plana
- Division of Cardiology, Baylor College of Medicine, Houston, Texas, USA
| | - Marielle Scherrer-Crosbie
- Cardiac Ultrasound Laboratory, Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Cardio-oncology program, Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
20
|
|
21
|
Oprea AD, Russell RR, Russell KS, Abu-Khalaf M. Chemotherapy Agents With Known Cardiovascular Side Effects and Their Anesthetic Implications. J Cardiothorac Vasc Anesth 2015; 31:2206-2226. [PMID: 26952170 DOI: 10.1053/j.jvca.2015.06.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Indexed: 01/11/2023]
|
22
|
Deo SV, Al-Kindi SG, Oliveira GH. Management of Advanced Heart Failure due to Cancer Therapy: the Present Role of Mechanical Circulatory Support and Cardiac Transplantation. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2015; 17:388. [PMID: 25960114 DOI: 10.1007/s11936-015-0388-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
OPINION STATEMENT Rapid improvement in antineoplastic therapy is increasing not only cancer survivorship but also the incidence of end-stage heart failure among breast and childhood cancer survivors. Anthracyclines and newer targeted therapies, including trastuzumab and tyrosine kinase inhibitors, are important agents implemented in clinical practice that carry cardiotoxic risk. While acute heart failure is often self-limited and reversible, delayed-onset heart failure significantly reduces survival. Extremes of age, renal dysfunction, pre-existing coronary artery disease, HER2 positivity, and multi-drug therapy are predictors of irreversible heart failure after chemotherapy. Left ventricular assist device (LVAD) implantation and cardiac transplantation can be performed safely in patients with end-stage heart failure (HF) from chemotherapy. However, co-existing right ventricular dysfunction, hepatic congestion, and increased risk of bleeding make LVAD therapy challenging and dependent on careful patient selection. Cardiac transplantation in patients with chemotherapy-induced heart failure can be performed with good 10-year survival, but requires 5 years of cancer freedom and post-transplant infections remain a problem. Improvements in LVAD therapy and the expanding role of the total artificial heart and other durable biventricular support devices will likely provide more reliable surgical options for the management of end-stage HF after chemotherapy.
Collapse
Affiliation(s)
- Salil V Deo
- Advanced Heart Failure and Transplant Center, Harrington Heart and Vascular Institute, University Hospitals Case Medical Center, Case Western Reserve University, 11100 Euclid Avenue, Cleveland, OH, 44106, USA
| | | | | |
Collapse
|
23
|
de Almeida ALC, Silva VA, de Souza Filho AT, Rios VG, Lopes JRP, de Afonseca SO, Cunha DDCA, Mendes MODC, Miranda DL, dos Santos Júnior EG. Subclinical ventricular dysfunction detected by speckle tracking two years after use of anthracycline. Arq Bras Cardiol 2015; 104:274-83. [PMID: 25993590 PMCID: PMC4415863 DOI: 10.5935/abc.20140209] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 10/08/2014] [Indexed: 01/20/2023] Open
Abstract
Background Heart failure is a severe complication associated with doxorubicin (DOX) use.
Strain, assessed by two-dimensional speckle tracking (2D-STE), has been shown to
be useful in identifying subclinical ventricular dysfunction. Objectives a) To investigate the role of strain in the identification of subclinical
ventricular dysfunction in patients who used DOX; b) to investigate determinants
of strain response in these patients. Methods Cross-sectional study with 81 participants: 40 patients who used DOX ±2
years before the study and 41 controls. All participants had left ventricular
ejection fraction (LVEF) ≥55%. Total dose of DOX was 396mg
(242mg/ms2). The systolic function of the LV was evaluated by LVEF
(Simpson), as well as by longitudinal (εLL), circumferential
(εCC), and radial (εRR) strains.
Multivariate linear regression (MLR) analysis was performed using
εLL (model 1) and εCC (model 2) as
dependent variables. Results Systolic and diastolic blood pressure values were higher in the control group (p
< 0.05). εLL was lower in the DOX group (-12.4 ±2.6%)
versus controls (-13.4 ± 1.7%; p = 0.044). The same occurred with
εCC: -12.1 ± 2.7% (DOX) versus -16.7 ± 3.6%
(controls; p < 0.001). The S’ wave was shorter in the DOX group (p = 0.035). On
MLR, DOX was an independent predictor of reduced εCC (B =
-4.429, p < 0.001). DOX (B = -1.289, p = 0.012) and age (B = -0.057, p = 0.029)
were independent markers of reduced εLL. Conclusion a) εLL, εCC and the S’ wave are reduced in
patients who used DOX ±2 years prior to the study despite normal LVEF,
suggesting the presence of subclinical ventricular dysfunction; b) DOX was an
independent predictor of reduced εCC; c) prior use of DOX and
age were independent markers of reduced εLL.
Collapse
Affiliation(s)
| | - Viviane Almeida Silva
- Hospital Dom Pedro de Alcântara, Santa Casa de Misericórdia de Feira de Santana, Feira de Santana, BA, Brasil
| | | | - Vinicius Guedes Rios
- Hospital Dom Pedro de Alcântara, Santa Casa de Misericórdia de Feira de Santana, Feira de Santana, BA, Brasil
| | - João Ricardo Pinto Lopes
- Hospital Dom Pedro de Alcântara, Santa Casa de Misericórdia de Feira de Santana, Feira de Santana, BA, Brasil
| | | | | | | | | | | |
Collapse
|
24
|
van Boxtel W, Bulten BF, Mavinkurve-Groothuis AMC, Bellersen L, Mandigers CMPW, Joosten LAB, Kapusta L, de Geus-Oei LF, van Laarhoven HWM. New biomarkers for early detection of cardiotoxicity after treatment with docetaxel, doxorubicin and cyclophosphamide. Biomarkers 2015; 20:143-8. [DOI: 10.3109/1354750x.2015.1040839] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- W. van Boxtel
- Department of Medical Oncology and
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands,
| | - B. F. Bulten
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands,
- MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands,
| | - A. M. C. Mavinkurve-Groothuis
- Department of Pediatric Hematology and Oncology, Radboud University Medical Center, Nijmegen, The Netherlands,
- Department of Pediatric Oncology, University Medical Centre Utrecht, Utrecht, The Netherlands,
| | - L. Bellersen
- Department of Cardiology, Radboud University Medical Center, Nijmegen, The Netherlands,
| | - C. M. P. W. Mandigers
- Department of Internal Medicine, Canisius-Wilhelmina Hospital, Nijmegen, The Netherlands,
| | | | - L. Kapusta
- Department of Children’s Heart Centre, Radboud University Medical Center, Nijmegen, The Netherlands,
- Pediatric Cardiology Unit, E. Wolfson Medical Centre, Holon, Israel, and
| | - L. F. de Geus-Oei
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands,
- MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands,
| | - H. W. M. van Laarhoven
- Department of Medical Oncology and
- Department of Medical Oncology, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
25
|
Oliveira GH, Qattan MY, Al-Kindi S, Park SJ. Advanced Heart Failure Therapies for Patients With Chemotherapy-Induced Cardiomyopathy. Circ Heart Fail 2014; 7:1050-8. [DOI: 10.1161/circheartfailure.114.001292] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Guilherme H. Oliveira
- From the Advanced Heart Failure and Transplantation Center and Onco-Cardiology Program, Harrington Heart and Vascular Institute, University Hospitals Case Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH
| | - Marwan Y. Qattan
- From the Advanced Heart Failure and Transplantation Center and Onco-Cardiology Program, Harrington Heart and Vascular Institute, University Hospitals Case Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH
| | - Sadeer Al-Kindi
- From the Advanced Heart Failure and Transplantation Center and Onco-Cardiology Program, Harrington Heart and Vascular Institute, University Hospitals Case Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH
| | - Soon J. Park
- From the Advanced Heart Failure and Transplantation Center and Onco-Cardiology Program, Harrington Heart and Vascular Institute, University Hospitals Case Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH
| |
Collapse
|
26
|
Yu AF, Steingart RM, Fuster V. Cardiomyopathy associated with cancer therapy. J Card Fail 2014; 20:841-52. [PMID: 25151211 PMCID: PMC5972392 DOI: 10.1016/j.cardfail.2014.08.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 08/08/2014] [Accepted: 08/14/2014] [Indexed: 01/03/2023]
Abstract
Chemotherapy-associated cardiomyopathy is a well known cardiotoxicity of contemporary cancer treatment and a cause of increasing concern for both cardiologists and oncologists. As cancer outcomes improve, cardiovascular disease has become a leading cause of morbidity and mortality among cancer survivors. Asymptomatic or symptomatic left ventricular systolic dysfunction in the setting of cardiotoxic chemotherapy is an important entity to recognize. Early diagnosis of cardiac injury through the use of novel blood-based biomarkers or noninvasive imaging modalities may allow for the initiation of cardioprotective medications or modification of chemotherapy regimen to minimize or prevent further damage. Several clinical trials are currently underway to determine the efficacy of cardioprotective medications for the prevention of chemotherapy-associated cardiomyopathy. Implementing a strategy that includes both early detection and prevention of cardiotoxicity will likely have a significant impact on the overall prognosis of cancer survivors. Continued coordination of care between cardiologists and oncologists remains critical to maximizing the oncologic benefit of cancer therapy while minimizing any early or late cardiovascular effects.
Collapse
Affiliation(s)
- Anthony F Yu
- Cardiology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
| | - Richard M Steingart
- Cardiology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Valentin Fuster
- Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
27
|
Preemptive Cardioprotective Strategies in Patients Receiving Chemotherapy. CURRENT CARDIOVASCULAR RISK REPORTS 2014. [DOI: 10.1007/s12170-014-0406-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
28
|
Wasielewski M, van Spaendonck-Zwarts KY, Westerink NDL, Jongbloed JDH, Postma A, Gietema JA, van Tintelen JP, van den Berg MP. Potential genetic predisposition for anthracycline-associated cardiomyopathy in families with dilated cardiomyopathy. Open Heart 2014; 1:e000116. [PMID: 25332820 PMCID: PMC4195921 DOI: 10.1136/openhrt-2014-000116] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 05/15/2014] [Accepted: 05/24/2014] [Indexed: 12/17/2022] Open
Abstract
Objective Anthracyclines are successfully used in cancer treatment, but their use is limited by their cardiotoxic side effects. Several risk factors for anthracycline-associated cardiomyopathy (AACM) are known, yet the occurrence of AACM in the absence of these known risk factors suggests that other factors must play a role. The purpose of this study was to evaluate whether a genetic predisposition for dilated cardiomyopathy (DCM) could be a potential risk factor for AACM. Methods A hospital-based registry of 162 DCM families and two hospital-based registries of patients with cancer treated with systemic cancer therapy (n>6000) were reviewed focusing on AACM. Selected patients with AACM/DCM families with possible AACM (n=21) were analysed for mutations in cardiomyopathy-associated genes and presymptomatic cardiological evaluation of first-degree relatives was performed. Results We identified five DCM families with AACM and one patient with AACM with a family member with a possible early sign of mild DCM. Pathogenic MYH7 mutations were identified in two of these six families. The MYH7 c.1633G>A (p.Asp545Asn) and c.2863G>A (p.Asp955Asn) mutations (one double mutant allele) were identified in a DCM family with AACM. The MYH7 c.4125T>A (p.Tyr1375X) mutation was identified in one patient with AACM. Conclusions This study further extends the hypothesis that a genetic predisposition to DCM could be a potential risk factor for AACM.
Collapse
Affiliation(s)
- Marijke Wasielewski
- Department of Genetics , University Medical Center Groningen, University of Groningen , Groningen , T he Netherlands
| | - Karin Y van Spaendonck-Zwarts
- Department of Genetics , University Medical Center Groningen, University of Groningen , Groningen , T he Netherlands ; Department of Clinical Genetics , Academic Medical Center, University of Amsterdam , Amsterdam , The Netherlands
| | - Nico-Derk L Westerink
- Department of Genetics , University Medical Center Groningen, University of Groningen , Groningen , T he Netherlands
| | - Jan D H Jongbloed
- Department of Genetics , University Medical Center Groningen, University of Groningen , Groningen , T he Netherlands
| | - Aleida Postma
- Department of Paediatric Oncology , University Medical Center Groningen, University of Groningen, Beatrix Children's Hospital , Groningen , The Netherlands
| | - Jourik A Gietema
- Department of Medical Oncology , University Medical Center Groningen, University of Groningen , Groningen , The Netherlands
| | - J Peter van Tintelen
- Department of Genetics , University Medical Center Groningen, University of Groningen , Groningen , T he Netherlands
| | - Maarten P van den Berg
- Department of Cardiology , University Medical Center Groningen, University of Groningen, Groningen , The Netherlands
| |
Collapse
|
29
|
Salvatici M, Cardinale D, Botteri E, Bagnardi V, Mauro C, Cassatella MC, Lentati P, Bottari F, Zorzino L, Passerini R, Cipolla CM, Sandri MT. TnI-Ultra assay measurements in cancer patients: Comparison with the conventional assay and clinical implication. Scandinavian Journal of Clinical and Laboratory Investigation 2014; 74:385-91. [DOI: 10.3109/00365513.2014.898325] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
30
|
Basar EZ, Corapcioglu F, Babaoglu K, Anik Y, Gorur Daglioz G, Dedeoglu R. Are cardiac magnetic resonance imaging and radionuclide ventriculography good options against echocardiography for evaluation of anthracycline induced chronic cardiotoxicity in childhood cancer survivors? Pediatr Hematol Oncol 2014; 31:237-52. [PMID: 24499452 DOI: 10.3109/08880018.2013.851753] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Anthracyclines are widely used for the treatment of solid tumors in pediatric oncology. However, their uses may be limited by progressive chronic cardiotoxicity related to the cumulative dosage. The aims of this study are to compare diagnostic techniques and prepare an algorithm for diagnosis of anthracycline induced chronic cardiotoxicity. The patients were evaluated according to age, sex, time elapsed since the last dose of anthracycline treatment, presence of cardiovascular symptoms, follow-up duration, type of anthracycline, cumulative anthracycline dose, and concomitant mediastinal radiation therapy. Late subclinical cardiotoxicity was detected by history, physical examination, electrocardiography (ECG), Holter monitor, echocardiography (ECHO), radionuclide ventriculography (MUGA), and cardiac magnetic resonance imaging (MRI). Thirty-seven male and 19 female patients with a median age of 11.2 ± 4.6 (range, 3.5-22.0) years were included in the study. Patients were grouped according to cumulative anthracycline doses. Subclinical cardiac dysfunction was detected in 20 patients by at least one of ECHO, MRI or MUGA after anthracycline chemotherapy. We revealed that other than ECHO, MRI and MUGA have high clinical importance for evaluating subclinical late cardiac complications in children treated with anthracyclines.
Collapse
Affiliation(s)
- Evic Zeynep Basar
- Department of Pediatric Oncology, Kocaeli University, Kocaeli, Turkey
| | | | | | | | | | | |
Collapse
|
31
|
Saeed MF, Premecz S, Goyal V, Singal PK, Jassal DS. Catching broken hearts: pre-clinical detection of doxorubicin and trastuzumab mediated cardiac dysfunction in the breast cancer setting. Can J Physiol Pharmacol 2014; 92:546-50. [PMID: 24959994 DOI: 10.1139/cjpp-2013-0470] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Although breast cancer is one of the leading causes of death in women worldwide, there is an overall improvement in the survival of this patient population. This is likely due to a combination of early detection through screening and awareness, improved targeted biological therapy, and an overall improvement in disease management. Despite the beneficial effects of the 2 anti-cancer drugs doxorubicin (DOX) and trastuzumab (TRZ) in women with breast cancer, development of cardiotoxicity is a major concern. The occurrence of left ventricular systolic dysfunction is unacceptably high in nearly 1 in 4 women treated with DOX+TRZ in the breast cancer setting. In this review, we explore the use of non-invasive cardiac imaging for the early detection of chemotherapy-mediated cardiotoxicity in women with breast cancer, in the hope of preventing end-stage heart disease in this cancer population.
Collapse
Affiliation(s)
- Mahwash F Saeed
- a Section of Cardiology, Department of Internal Medicine, Faculty of Medicine, University of Manitoba, Rm Y3531, Bergen Cardiac Care Centre, St. Boniface General Hospital, 409 Tache Avenue, Winnipeg, MB R2H 2A6, Canada
| | | | | | | | | |
Collapse
|
32
|
Molecular basis of cancer-therapy-induced cardiotoxicity: introducing microRNA biomarkers for early assessment of subclinical myocardial injury. Clin Sci (Lond) 2014; 126:377-400. [PMID: 24274966 DOI: 10.1042/cs20120620] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Development of reliable biomarkers for early clinical assessment of drug-induced cardiotoxicity could allow the detection of subclinical cardiac injury risk in vulnerable patients before irreversible damage occurs. Currently, it is difficult to predict who will develop drug-induced cardiotoxicity owing to lack of sensitivity and/or specificity of currently used diagnostics. miRNAs are mRNA regulators and they are currently being extensively profiled for use as biomarkers due to their specific tissue and disease expression signature profiles. Identification of cardiotoxicity-specific miRNA biomarkers could provide clinicians with a valuable tool to allow prognosis of patients at risk of cardiovascular injury, alteration of a treatment regime or the introduction of an adjunct therapy in order to increase the long-term survival rate of patients treated with cardiotoxic drugs.
Collapse
|
33
|
Horacek JM, Vasatova M, Pudil R, Tichy M, Zak P, Jakl M, Jebavy L, Maly J. Biomarkers for the early detection of anthracycline-induced cardiotoxicity: current status. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2014; 158:511-7. [PMID: 24457832 DOI: 10.5507/bp.2014.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2013] [Accepted: 01/15/2014] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Cardiotoxicity is a well-known and potentially serious complication of anticancer therapy. Anthracycline-based chemotherapy represents the greatest risk. Early detection of cardiotoxicity is crucial for applying preventive and supportive therapeutic strategies. METHODS AND RESULTS Various methods have been recommended for monitoring of cardiotoxicity. In our conditions, echocardiography and electrocardiography are routinely used. However, this approach shows low sensitivity for the early prediction of cardiomyopathy when the possibilities of appropriate management could still improve the patient's outcome. Recently, biomarkers of cardiac injury have been investigated in the assessment of chemotherapy-induced cardiotoxicity. Cardiospecific biomarkers, such as cardiac troponins, show high diagnostic efficacy in the early subclinical phase of the disease before the clinical onset of cardiomyopathy. Increase in their concentrations correlates with disease severity. As for natriuretic peptides, some studies, including ours, have shown promising results. Definitive evidence of their diagnostic and prognostic role in this context is still lacking and natriuretic peptides have not been routinely used for monitoring of cardiotoxicity in clinical practice. Other perspective biomarkers of cardiotoxicity in oncology are under study, especially heart-type fatty acid-binding protein (H-FABP) and glycogen phosphorylase BB (GPBB). Our studies using GPBB have provided encouraging results. However, the available data are limited and their practical use in this context cannot be recommended until their clinical efficacy is clearly defined. CONCLUSIONS This review covers the current status of biomarkers for the early detection of anthracycline-induced cardiotoxicity. The authors present in brief, their own experience with multiple biomarkers in the detection of cardiotoxicity.
Collapse
Affiliation(s)
- Jan M Horacek
- Department of Internal Medicine, Faculty of Military Health Sciences, University of Defence, Hradec Kralove, Czech Republic
| | | | | | | | | | | | | | | |
Collapse
|
34
|
van den Berg MP, van Spaendonck-Zwarts KY, van Veldhuisen DJ, Gietema JA, Postma A, van Tintelen JP. Familial dilated cardiomyopathy: another risk factor for anthracycline-induced cardiotoxicity? Eur J Heart Fail 2014; 12:1297-9. [DOI: 10.1093/eurjhf/hfq175] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Maarten P. van den Berg
- Department of Cardiology; University Medical Center Groningen, University of Groningen; PO Box 30.001, 9700 RB Groningen The Netherlands
| | | | - Dirk J. van Veldhuisen
- Department of Cardiology; University Medical Center Groningen, University of Groningen; PO Box 30.001, 9700 RB Groningen The Netherlands
| | - Jourik A. Gietema
- Department of Medical Oncology; University Medical Center Groningen, University of Groningen; Groningen The Netherlands
| | - Aleida Postma
- Department of Pediatric Oncology; University Medical Center Groningen, University of Groningen; Groningen The Netherlands
| | - J. Peter van Tintelen
- Department of Genetics; University Medical Center Groningen, University of Groningen; Groningen The Netherlands
| |
Collapse
|
35
|
Vasu S, Hundley WG. Understanding cardiovascular injury after treatment for cancer: an overview of current uses and future directions of cardiovascular magnetic resonance. J Cardiovasc Magn Reson 2013; 15:66. [PMID: 23902649 PMCID: PMC3765662 DOI: 10.1186/1532-429x-15-66] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 07/07/2013] [Indexed: 01/03/2023] Open
Abstract
While cancer-free survival has improved over the past 20 years for many individuals with prostate, renal, breast, and hematologic malignancies, the increasingly recognized prevalence of cardiovascular (CV) events in cancer survivors has been an unintended consequence of many of the therapies that have improved these survival rates. The increase in CV events threatens to offset the improvement in cancer related survival. As a result, there is an emerging need to develop methods to identify those individuals treated for cancer at increased risk of cardiovascular events. With its inherent ability to characterize myocardial tissue and identify both cardiac and vascular dysfunction, cardiovascular magnetic resonance (CMR) has the potential to identify both subclinical and early clinical CV injury before the development of an overt catastrophic event such as a myocardial infarction, stroke, or premature cardiac death. Early identification provides an opportunity for the implementation of primary prevention strategies to prevent such events, thereby improving overall cancer survivorship and quality of life. This article reviews the etiology of CV events associated with cancer therapy and the unique potential of CMR to provide early diagnosis of subclinical CV injury related to the administration of these therapies.
Collapse
Affiliation(s)
- Sujethra Vasu
- Department of Internal Medicine (Section on Cardiology), Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - W Gregory Hundley
- Department of Internal Medicine (Section on Cardiology), Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
- DepartmentRadiology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
36
|
Abstract
OPINION STATEMENT The increase in survivorship of cancer patients makes the understanding of the available options for prevention and treatment of cardiotoxicity induced by antineoplastic agents a crucial topic both for cardiologists and oncologists. The most frequent and typical clinical manifestation of cardiotoxicity is asymptomatic or symptomatic left ventricular dysfunction, which may progress to overt heart failure. It may be induced not only by conventional cancer therapy, like anthracyclines, but also by new antitumoral targeted therapy such as trastuzumab. The current standard for monitoring cardiac damage during antineoplastic treatment, mainly based on the quantification of left ventricular ejection fraction, detects cardiac toxicity only when a functional impairment has already occurred. Evaluation of cardiac biomarkers such as troponin, however, has shown excellent sensitivity in the early detection of cardiotoxicity by the identification of patients with subclinical cardiac injury that precedes the development of cardiac dysfunction. The use of angiotensin-converting enzyme inhibitors in patients with troponin elevation during chemotherapy may be an effective tool to prevent left ventricular ejection fraction reduction and late cardiac events. There are no well established recommendations for treatment of cancer patients who develop cardiac dysfunction. Angiotensin-converting enzyme inhibitors and beta-blockers have proven to be effective in this setting. However, there are concerns in using these medications in cancer patients, and therefore the tendency is to treat patients only if symptomatic. However, the clinical benefit of these medications may be more evident in asymptomatic patients, and the recovery of cardiac function strongly depends on the amount of time elapsed from the end of chemotherapy to the start of heart failure therapy. This observation suggests that the early detection of cardiac damage is crucial and early use of angiotensin-converting enzyme inhibitors and beta-blockers should be considered in patients with left ventricular dysfunction induced by antineoplastic agents.
Collapse
|
37
|
Gillespie HS, McGann CJ, Wilson BD. Noninvasive diagnosis of chemotherapy related cardiotoxicity. Curr Cardiol Rev 2013; 7:234-44. [PMID: 22758624 PMCID: PMC3322441 DOI: 10.2174/157340311799960672] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2011] [Revised: 10/24/2011] [Accepted: 11/27/2011] [Indexed: 11/22/2022] Open
Abstract
Chemotherapeutic agents reduce mortality and can prevent morbidity in a wide range of malignancies. These agents are, however, associated with toxicities of their own, and the treating physician must remain ever vigilant against the risk outgrowing the benefit of therapy. Thus, pre-treatment evaluation and monitoring for toxicity during and following administration is a fundamental tenet of oncologic practice. Among the most insidious and deadly toxicities of anti-tumor agents is cardiac toxicity, which in some cases may be irreversible. Early detection of cardiotoxicity allows the treating oncologist to redirect therapy or dose modify, taking into account the cost of a reduction in therapy against the potential of further injury to the patient. In these instances, the role of the cardiologist is to assist and advise the oncologist by providing diagnostic and prognostic information regarding developing cardiotoxicity. This review discusses noninvasive diagnostic options to identify and characterize cardiotoxicity and their use in prognosis and guiding therapy. We also review established protocols for cardiac safety monitoring in the treatment of malignancy.
Collapse
|
38
|
Khatua TN, Adela R, Banerjee SK. Garlic and cardioprotection: insights into the molecular mechanisms. Can J Physiol Pharmacol 2013; 91:448-58. [PMID: 23746107 DOI: 10.1139/cjpp-2012-0315] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Garlic is widely recognized for its immense therapeutic potential. Garlic has been shown to exert its beneficial effects against a wide spectrum of diseases, including cancer, diabetes, and microbial infections, as well as immunological and cardiovascular disorders. Most of the research on garlic has indicated that garlic and its active compounds are effective in reducing cardiovascular and metabolic risk by normalizing abnormal plasma lipids, oxidized low density lipoproteins, abnormal platelet aggregation, high blood pressure, and cardiac injury. Some of the beneficial effects of dietary garlic against cardiovascular disorders are mediated via the generation of hydrogen sulfide and nitric oxide in cardiomyocytes and endothelial cells. Garlic has the potential to protect the heart against myocardial infarction, doxorubicin-induced cardiotoxicity, arrhythmia, hypertrophy, and ischemia-reperfusion injury. The induction of cardiac endogenous antioxidants and the reduction of lipid peroxidation by garlic has been reported by several different groups. Other mechanisms, such as regulating ion channels, modulating Akt signaling pathways, histone deacetylase inhibition, and cytochrome P450 inhibition, could be responsible for the cardioprotective effect of garlic. Although several mechanisms have been identified for the cardioprotective effect of garlic, there is a need for further research to identify the specific molecular mechanism of cardioprotection in different cardiac diseases.
Collapse
Affiliation(s)
- Tarak Nath Khatua
- Division of Medicinal Chemistry and Pharmacology, Indian Institute of Chemical Technology (IICT), Hyderabad, India
| | | | | |
Collapse
|
39
|
Pastore A, Geiger S, Baur D, Hausmann A, Tischer J, Horster S, Stemmler HJ. Cardiotoxicity After Anthracycline Treatment in Survivors of Adult Cancers: Monitoring by USCOM, Echocardiography and Serum Biomarkers. World J Oncol 2013; 4:18-25. [PMID: 29147326 PMCID: PMC5649915 DOI: 10.4021/wjon635w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2013] [Indexed: 01/08/2023] Open
Abstract
Background Anthracyclines are agents with a well known documented anti-tumoral activity. Cardiac side effects are the principal toxicity. Here we evaluate and monitor the onset of late anthracycline-induced cardiotoxicity with real-time CW-Doppler ultrasound cardiac output monitoring (USCOM®) and echocardiography in combination with serum biomarkers. Methods Fifty-two patients without cardiac disease who had received an anthracycline-based regimen for various cancer types were included in this study. Patients’ hemodynamic parameters as stroke volume (SV USCOM (mL)) and ejection fraction (EF ECHOCARDIOGRAPHY (%)) were measured with USCOM and echocardiography and correlated to serum biomarkers (NT-pro-BNP and cTnT). Results Eighteen patients (34.6%) developed cardiac disease (NYHA I-III). An increasing cumulative anthracycline dose was associated with a decrease of the EF determined by echocardiography as well the SV by USCOM and with a higher NYHA class. Those patients who experienced cardiac disease showed a reduction of the EF and SV and increased serum biomarkers. Conclusions Real-time CW-Doppler USCOM, is a fast and reliable method to monitor late hemodynamic changes as a symptom of anthracycline-induced cardiotoxicity comparable to the findings by echocardiography and serum biomarkers.
Collapse
Affiliation(s)
- Alessandro Pastore
- Med. Dept. III, Ludwig-Maximilians University of Munich, Campus Grosshadern, Munich, Germany
| | - Sandra Geiger
- Med. Dept. III, Ludwig-Maximilians University of Munich, Campus Grosshadern, Munich, Germany
| | - Dorothee Baur
- Med. Dept. III, Ludwig-Maximilians University of Munich, Campus Grosshadern, Munich, Germany
| | - Andreas Hausmann
- Med. Dept. III, Ludwig-Maximilians University of Munich, Campus Grosshadern, Munich, Germany
| | - Johanna Tischer
- Med. Dept. III, Ludwig-Maximilians University of Munich, Campus Grosshadern, Munich, Germany
| | - Sophia Horster
- Med. Dept. II, Ludwig-Maximilians University of Munich, Campus Grosshadern, Munich, Germany
| | - Hans Joachim Stemmler
- Med. Dept. III, Ludwig-Maximilians University of Munich, Campus Grosshadern, Munich, Germany
| |
Collapse
|
40
|
Vasatova M, Pudil R, Horacek JM, Buchler T. Current applications of cardiac troponin T for the diagnosis of myocardial damage. Adv Clin Chem 2013; 61:33-65. [PMID: 24015599 DOI: 10.1016/b978-0-12-407680-8.00002-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Biochemical markers of myocardial injury play an important role in the diagnosis of cardiovascular diseases. Measurement of cardiac biomarkers is one of the most important diagnostic tests in acute myocardial infarction (AMI), heart failure, and other cardiovascular disorders. Recently, the European Society of Cardiology, the American College of Cardiology Foundation, the American Heart Association, and the World Heart Federation have published a consensus definition of AMI that includes a detailed guideline for the assessment of biochemical markers in suspected disease. The cardiac troponins (cTI and cTnT) were recommended as preferred markers of myocardial necrosis in this setting. Herein, we review cardiac troponin biochemistry, the performance characteristics of cTnT assays, and optimal utilization of troponin in patients with proven or possible cardiovascular disease. We also discuss the use of troponin tests, with emphasis on cTnT, in different clinical situations in which its levels may be elevated.
Collapse
|
41
|
Oreto L, Todaro MC, Umland MM, Kramer C, Qamar R, Carerj S, Khandheria BK, Paterick TE. Use of echocardiography to evaluate the cardiac effects of therapies used in cancer treatment: what do we know? J Am Soc Echocardiogr 2012; 25:1141-52. [PMID: 23000452 DOI: 10.1016/j.echo.2012.09.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Indexed: 01/11/2023]
Abstract
Cardiologists and oncologists today face the daunting challenge of identifying patients at risk for late-onset left ventricular (LV) systolic dysfunction from the use of various chemotherapeutic agents. Currently, the most widely used method in clinical practice for monitoring the potential of chemotherapy-induced cardiotoxicity is calculation of LV ejection fraction. The use of LV ejection fraction to determine whether to continue or discontinue the use of chemotherapeutic agents is limited, because decreases in LV ejection fraction frequently occur late and can be irreversible. These limitations have led to the exploration of diastolic function and newer modalities that assess myocardial mechanics to identify sensitive and specific variables that can predict the occurrence of late systolic function. The cancer therapies associated with cardiotoxicity are reviewed in this report. Additionally, the authors evaluate the role of present-day echocardiographic parameters, complementary noninvasive imaging modalities, and biomarkers in the prediction of cardiotoxicity. The authors address the evolving role of cardioprotective agents and potential therapies to prevent or reverse the progression of LV systolic dysfunction. Finally, they provide some ideas regarding future directions to enhance the knowledge of predicting late-onset LV systolic dysfunction secondary to cancer therapy.
Collapse
Affiliation(s)
- Lilia Oreto
- Clinical and Experimental Department of Medicine and Pharmacology, University of Messina, Messina, Italy
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Della Pina P, Vizzardi E, Raddino R, Gavazzoni M, Caretta G, Gorga E, Dei Cas L. Biological Drugs: Classic Adverse Effects and New Clinical Evidences. Cardiovasc Toxicol 2012; 12:285-97. [DOI: 10.1007/s12012-012-9173-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
43
|
Sherief LM, Kamal AG, Khalek EA, Kamal NM, Soliman AAA, Esh AM. Biomarkers and early detection of late onset anthracycline-induced cardiotoxicity in children. Hematology 2012; 17:151-156. [PMID: 22664114 DOI: 10.1179/102453312x13376952196412] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND The main strategy for minimizing anthracycline cardiotoxicity is early detection of high-risk patients. AIM OF THE STUDY To investigate the role of cardiac biomarkers; cardiac troponin T (cTnT) and N-terminal probrain natriuretic peptide (NT-pro-BNP), and tissue Doppler imaging (TDI), as early predictors of chronic cardiotoxicity in survivors of acute leukemia. PATIENTS AND METHODS We carried a retrospective study on 50 asymptomatic survivors of acute leukemia who received anthracycline in their treatment protocols. All patients underwent blood sampling to determine the levels of NT-pro-BNP and cTnT along with conventional echocardiography and TDI. RESULTS None had abnormal cTnT levels. About 20% had abnormal NT-pro-BNP levels. Diastolic dysfunction of the left ventricle was the most significant in conventional echocardiography. TDI was superior as it detected myocardial affection in 10% more than echo. TDI demonstrated global myocardial damage with significant aberrations in peak myocardial velocities and ratios. CONCLUSIONS NT-pro-BNP can be used as a sensitive cardiac biomarker in monitoring of anthracycline-induced cardiotoxicity. Follow up is essential to validate the role of NT-pro-BNP as an early marker for late onset anthracycline-induced cardiotoxicity. Tissue Doppler is marvelous as it could detect early cardiac dysfunction even in those with normal study by conventional echocardiography.
Collapse
MESH Headings
- Adolescent
- Anthracyclines/administration & dosage
- Anthracyclines/adverse effects
- Antibiotics, Antineoplastic/administration & dosage
- Antibiotics, Antineoplastic/adverse effects
- Biomarkers/blood
- Cardiotoxins/administration & dosage
- Cardiotoxins/adverse effects
- Child
- Child, Preschool
- Early Diagnosis
- Echocardiography, Doppler
- Female
- Heart/drug effects
- Heart/physiopathology
- Humans
- Leukemia, Biphenotypic, Acute/drug therapy
- Male
- Natriuretic Peptide, Brain/blood
- Peptide Fragments/blood
- Retrospective Studies
- Survivors
- Troponin T/blood
- Ventricular Dysfunction, Left/blood
- Ventricular Dysfunction, Left/chemically induced
- Ventricular Dysfunction, Left/diagnosis
Collapse
Affiliation(s)
- Laila M Sherief
- Department of Pediatrics, Faculty of Medicine, Zagazig University, Egypt
| | | | | | | | | | | |
Collapse
|
44
|
Geiger S, Stemmler HJ, Suhl P, Stieber P, Lange V, Baur D, Hausmann A, Tischer J, Horster S. Anthracycline-induced cardiotoxicity: cardiac monitoring by continuous wave-Doppler ultrasound cardiac output monitoring and correlation to echocardiography. ACTA ACUST UNITED AC 2012; 35:241-6. [PMID: 22868502 DOI: 10.1159/000338335] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
BACKGROUND Anthracyclines are agents with a wellknown cardiotoxicity. The study sought to evaluate the hemodynamic response to an anthracycline using realtime continuous-wave (CW)-Doppler ultrasound cardiac output monitoring (USCOM) and echocardiography in combination with serum biomarkers. METHODS 50 patients (26 male, 24 female, median age 59 years) suffering from various types of cancer received an anthracycline-based regimen. Patients' responses were measured at different time points (T0 prior to infusion, T1 6 h post infusion, T2 after 1 day, T3 after 7 days, and T4 after 3 months) with CW-Doppler ultrasound (T0-T4) and echocardiography (T1, T4) for hemodynamic parameters such as stroke volume (SV; SVUSCOM ml) and ejection fraction (EF; EFechocardiography%) and with NT-pro-BNP and hs-Troponin T (T0-T4). RESULTS During the 3-month observation period, the relative decrease in the EF determined by echocardiography was -2.1% (▵T0-T4, T0 71 ± 7.8%, T4 69.5 ± 7%, p = 0.04), whereas the decrease in SV observed using CW-Doppler was -6.5% (▵T0-T4, T0 54 ± 19.2 ml, T4 50.5 ± 20.6 ml, p = 0.14). The kinetics for serum biomarkers were inversely correlated. CONCLUSIONS Combining real-time CW-Doppler USCOM and serum biomarkers is feasible for monitoring the immediate and chronic hemodynamic changes during an anthracycline-based regimen; the results obtained were comparable to those from echocardiography.
Collapse
Affiliation(s)
- Sandra Geiger
- Medical Department III, Ludwig-Maximilians University of Munich, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Mitochondria death/survival signaling pathways in cardiotoxicity induced by anthracyclines and anticancer-targeted therapies. Biochem Res Int 2012; 2012:951539. [PMID: 22482055 PMCID: PMC3318211 DOI: 10.1155/2012/951539] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Revised: 01/04/2012] [Accepted: 01/09/2012] [Indexed: 01/27/2023] Open
Abstract
Anthracyclines remain the cornerstone of treatment in many malignancies but these agents have a cumulative dose relationship with cardiotoxicity. Development of cardiomyopathy and congestive heart failure induced by anthracyclines are typically dose-dependent, irreversible, and cumulative. Although past studies of cardiotoxicity have focused on anthracyclines, more recently interest has turned to anticancer drugs that target many proteins kinases, such as tyrosine kinases. An attractive model to explain the mechanism of this cardiotoxicity could be myocyte loss through cell death pathways. Inhibition of mitochondrial transition permeability is a valuable tool to prevent doxorubicin-induced cardiotoxicity. In response to anthracycline treatment, activation of several protein kinases, neuregulin/ErbB2 signaling, and transcriptional factors modify mitochondrial functions that determine cell death or survival through the modulation of mitochondrial membrane permeability. Cellular response to anthracyclines is also modulated by a myriad of transcriptional factors that influence cell fate. Several novel targeted chemotherapeutic agents have been associated with a small but worrying risk of left ventricular dysfunction. Agents such as trastuzumab and tyrosine kinase inhibitors can lead to cardiotoxicity that is fundamentally different from that caused by anthracyclines, whereas biological effects converge to the mitochondria as a critical target.
Collapse
|
46
|
Abstract
Cardiotoxicity remains a major limitation of chemotherapy, strongly affecting the quality of life and the overall survival of cancer patients, regardless of their oncologic prognosis. The time elapsed from the end of cancer therapy to the beginning of heart failure therapy for chemotherapy-induced cardiac dysfunction is an important determinant of the extent of recovery. This highlights the need for a real-time diagnosis of cardiac injury. The current standard for monitoring cardiac function detects cardiotoxicity only when a functional impairment has already occurred, precluding any chance of preventing its development. In the last decade, early identification, assessment, and monitoring of cardiotoxicity, by measurement of serum cardiospecific biomarkers, have been proposed as an effective alternative. In particular, the role of troponin I in identifying patients at risk for cardiotoxicity and of angiotensin-converting enzyme inhibitors in preventing left ventricular ejection fraction reduction and cardiac events has clearly proved to be an effective strategy for this complication. In addition, novel biomarkers for the identification of cardiotoxicity are emerging. The use of a multimarker approach may provide a unique opportunity for advancement in this field, allowing for better stratification of the cardiac risk in cancer patients treated with anticancer drugs.
Collapse
Affiliation(s)
- D Cardinale
- Cardiology Unit, European Institute of Oncology, Milan, Italy.
| | | |
Collapse
|
47
|
Jeyakumar A, DiPenta J, Snow S, Rayson D, Thompson K, Theriault C, Younis T. Routine cardiac evaluation in patients with early-stage breast cancer before adjuvant chemotherapy. Clin Breast Cancer 2011; 12:4-9. [PMID: 22154116 DOI: 10.1016/j.clbc.2011.07.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Revised: 07/15/2011] [Accepted: 07/29/2011] [Indexed: 10/14/2022]
Abstract
INTRODUCTION This population-based study of women diagnosed with early-stage breast cancer aimed to (i) determine the current utilization pattern of multigated acquisition (MUGA) scans before adjuvant chemotherapy (AdjC) treatment, and (ii) examine the impact of MUGA scan results on AdjC decision making. METHODS All women who underwent curative-intent surgery for stage I-III breast cancer between October 2005 and September 2006 in Nova Scotia, Canada, were identified through the provincial cancer registry. A retrospective chart review was performed to abstract all relevant clinical-pathologic variables, including baseline cardiac risk factors. The association between MUGA scan utilization and clinical-pathologic variables, as well as receipt and type of AdjC, was examined through univariate and multivariate analyses. RESULTS The study included 593 women, of whom 238 (40%) received AdjC (94% anthracycline vs. 6% nonanthracycline) and 198 (33%) underwent baseline MUGA scans. Of those received AdjC, 80% underwent MUGA scans. MUGA scan utilization was associated with AdjC treatment (yes vs. no; P < .0001), Her-2/neu status (positive vs. negative vs. not tested; P < .0001), and AdjC regimen (anthracycline vs. nonanthracycline; P < .0001). Abnormal MUGA results were observed in 5 (2.5%) of 198; all were smokers, and 4 were >65 years of age. In the 1 patient <50 years old, subsequent echocardiograms indicated normal cardiac function. CONCLUSIONS Routine baseline MUGA scans before AdjC were abnormal and changed the AdjC treatment decision in only 2.5% and 2.0% of patients, respectively. Routine MUGA scans before anthracycline-based AdjC without trastuzumab, however, did not influence AdjC decisions for younger patients <65 years of age without underlying cardiac risk factors.
Collapse
Affiliation(s)
- Alwin Jeyakumar
- Department of Medicine, Queen Elizabeth II Health Sciences Centre, Halifax, Nova Scotia, Canada.
| | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Endomyocardial biopsy (EMB) is widely used for surveillance of cardiac allograft rejection and for the diagnosis of unexplained ventricular dysfunction. Typically, EMB is performed through the jugular or femoral veins and is associated with a serious acute complication rate of less than 1% using current flexible bioptomes. Although it is accepted that EMB should be used to monitor for rejection after transplant, use of EMB for the diagnosis of various myocardial diseases is controversial. Diagnosis of myocardial disease in the nontransplant recipient is often successful via noninvasive investigations including laboratory evaluation; echocardiography, nuclear studies, and magnetic resonance imaging can yield specific diagnoses in the absence of invasive EMB. Therefore, use of the technique is patient specific and depends on the potential prognostic and treatment information gained by establishing a pathologic diagnosis beyond noninvasive testing.
Collapse
Affiliation(s)
- Aaron M From
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN 55905, USA
| | | | | |
Collapse
|
49
|
Oztarhan K, Guler S, Aktas B, Arslan M, Salcioglu Z, Aydogan G. The value of echocardiography versus cardiac troponin I levels in the early detection of anthracycline cardiotoxicity in childhood acute leukemia: prospective evaluation of a 7-year-long clinical follow-up. Pediatr Hematol Oncol 2011; 28:380-94. [PMID: 21699467 DOI: 10.3109/08880018.2011.563772] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The present study was designed to evaluate the significance of echocardiography versus cardiac troponin I levels in early detection of anthracycline dependent cardiotoxicity in acute lymphoblastic leukemia (ALL) patients. A total of 276 pediatric ALL patients were included in the study prospectively along 3 phases of data collection lasted from 2002 to 2009; including phase I (March 2002 to February 2003; n = 25; 53.3% females), phase II (September 2003 to April 2004; n = 35; 57.1% females), and phase III (January 2005 to June 2009; n = 216; 52.7% females) with respect to cumulative anthracycline doses applied. Anthracycline was administered in accordance with berlin-Franfurt-Munich (BFM)-2000 protocol in doses of 30 to 350 mg/m(2) (in the first phase) and 30 to 240 mg/m(2) (in the following phases). Evaluation of cardiotoxicity was performed via echocardiography and measurement of cardiac troponin I levels. Patients in each phase were homogenous in terms of gender and age. Diastolic dysfunction determined via reduction E/A ratio below the cutoff value was demonstrated to deteriorate earlier than systolic functions and alteration in cardiac enzymes. Being similar between dose groups, cTnI levels were shown to rise in the presence of congestive heart failure. In conclusion, anthracycline cardiotoxicity appears to be detected in an earlier stage by using diastolic parameters compared to systolic parameters and cardiac enzymes.
Collapse
Affiliation(s)
- Kazım Oztarhan
- Division of Pediatric Cardiology, Department of Pediatrics, Istanbul Bakirkoy Maternity and Children Diseases Training and Research Hospital, Istanbul, Turkey.
| | | | | | | | | | | |
Collapse
|
50
|
Cardinale D. Treatment and prevention of cardiotoxicity due to anticancer therapy. J Cardiovasc Echogr 2011. [DOI: 10.1016/j.jcecho.2011.06.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|