1
|
Conyers R, Elliott DA. Straight to the heart: Protecting the patient's heart during chemotherapy. Cell Stem Cell 2024; 31:1715-1716. [PMID: 39642857 DOI: 10.1016/j.stem.2024.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 10/09/2024] [Accepted: 10/14/2024] [Indexed: 12/09/2024]
Abstract
How do we protect the heart during chemotherapy with the anthracycline drug class? Tackling this question, Liu et al. combined pluripotent stem cell models, CRISPR genetic screens, and molecular modeling to identify indisulam as a potential cardioprotective drug in this issue of Cell Stem Cell.
Collapse
Affiliation(s)
- Rachel Conyers
- Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, VIC, Australia; Children's Cancer Centre, Royal Melbourne Hospital, Parkville, VIC, Australia; Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia; The Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Parkville, VIC, Australia
| | - David A Elliott
- Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, VIC, Australia; Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia; The Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Parkville, VIC, Australia; Australian Regenerative Medicine Institute, Monash University, Wellington Road, Clayton, VIC, Australia.
| |
Collapse
|
2
|
Jacquemyn X, Zhan J, Van den Eynde J, Cordrey K, Long R, Rao S, Barnes BT, Thompson WR, Danford D, Kutty S. Time course of hypertension and myocardial dysfunction following anthracycline chemotherapy in pediatric patients. IJC HEART & VASCULATURE 2024; 53:101436. [PMID: 38872982 PMCID: PMC11169083 DOI: 10.1016/j.ijcha.2024.101436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 04/11/2024] [Accepted: 05/27/2024] [Indexed: 06/15/2024]
Abstract
Background Anthracyclines are associated with cardiac dysfunction. Little is known about the interplay of pre-existing hypertension and treatment response. We aimed to investigate the relationship between hypertension and the development of cancer therapy-related cardiac dysfunction (CTRCD) in pediatric patients treated with anthracycline chemotherapy. Methods Pediatric patients with cancer who received anthracycline chemotherapy from 2013 to 2021 were retrospectively included. Serial cardiac assessments were conducted during and after chemotherapy. The primary outcome was the development of CTRCD, classified as mild, moderate, or severe according to contemporary definitions. Results Among 190 patients undergoing anthracycline chemotherapy, 34 patients (17.9 %) had hypertension (24 patients Stage 1, and 10 patients Stage 2) at baseline evaluation. Patients underwent chemotherapy for a median of 234.4 days (interquartile range 127.8-690.3 days) and were subsequently followed up. Hypertension was frequent during follow-up 31.3 % (0-3 months), 15.8 % (3-6 months), 21.9 % (0.5-1 years), 24.7 % (1-2 years), 31.1 % (2-4 years) and 35.8 % (beyond 4 years) (P for trend < 0.001). Freedom from mild CTRCD at 5 years was 45.0 %, freedom from moderate CTRCD was 87.8 % at 5 years. Baseline hypertension did not increase the risk of mild (HR 0.77, 95 % CI: 0.41-1.42, P = 0.385) or moderate CTRCD (HR 0.62, 95 % CI: 0.14-2.72, P = 0.504). Patients with baseline hypertension showed different global longitudinal strain (P < 0.001) and LVEF (P < 0.001) patterns during follow-up. Conclusions Pediatric patients often develop CTRCD post-anthracycline chemotherapy. Those with pre-existing hypertension show a unique treatment response, despite no increased CTRCD risk, warranting further investigation.
Collapse
Affiliation(s)
- Xander Jacquemyn
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Department of Cardiovascular Sciences, KU Leuven & Congenital and Structural Cardiology, UZ Leuven, Leuven, Belgium
| | - Junzhen Zhan
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Jef Van den Eynde
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Department of Cardiovascular Sciences, KU Leuven & Congenital and Structural Cardiology, UZ Leuven, Leuven, Belgium
| | - Kyla Cordrey
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Rita Long
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Sruti Rao
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Benjamin T. Barnes
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - W. Reid Thompson
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - David Danford
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Shelby Kutty
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
3
|
Yan H, Wang P, Yang F, Cheng W, Chen C, Zhai B, Zhou Y. Anticancer therapy-induced adverse drug reactions in children and preventive and control measures. Front Pharmacol 2024; 15:1329220. [PMID: 38425652 PMCID: PMC10902428 DOI: 10.3389/fphar.2024.1329220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 01/29/2024] [Indexed: 03/02/2024] Open
Abstract
In recent years, considerable achievements have been made in pediatric oncology with the innovation and development of antitumor drugs. However, compared to adults, children as a special group have not yet matured fully in terms of liver and kidney function. Moreover, pediatric patients are prone to more adverse drug reactions (ADRs) from the accumulation of antineoplastic drugs due to their smaller body size and larger body surface area. Chemotherapy-related ADRs have become a non-negligible factor that affects cancer remission. To date, studies on ADRs in pediatric cancer patients have emerged internationally, but few systematic summaries are available. Here, we reviewed the various systemic ADRs associated with antitumor drugs in children and adolescent patients, as well as the advances in strategies to cope with ADRs, which consisted of neurotoxicity, hematological toxicity, cardiotoxicity, ADRs of the respiratory system and gastrointestinal system and urinary system, ADRs of the skin and its adnexa, allergic reactions, and other ADRs. For clinicians and researchers, understanding the causes, symptoms, and coping strategies for ADRs caused by anticancer treatments will undoubtedly benefit more children.
Collapse
Affiliation(s)
- Hui Yan
- Henan Provincial Clinical Research Center for Pediatric Diseases, Henan Key Laboratory of Pediatric Genetics and Metabolic Diseases, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
- Department of Cardiothoracic Surgery, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Penggao Wang
- Department of Cardiothoracic Surgery, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Fang Yang
- Department of Cardiothoracic Surgery, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Weyland Cheng
- Henan Provincial Clinical Research Center for Pediatric Diseases, Henan Key Laboratory of Pediatric Genetics and Metabolic Diseases, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Congcong Chen
- Henan Provincial Clinical Research Center for Pediatric Diseases, Henan Key Laboratory of Pediatric Genetics and Metabolic Diseases, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Bo Zhai
- Department of Cardiothoracic Surgery, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Yang Zhou
- Henan Provincial Clinical Research Center for Pediatric Diseases, Henan Key Laboratory of Pediatric Genetics and Metabolic Diseases, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
- Department of Cardiothoracic Surgery, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| |
Collapse
|
4
|
Fonoudi H, Jouni M, Cejas RB, Magdy T, Blancard M, Ge N, Shah DA, Lyra-Leite DM, Neupane A, Gharib M, Jiang Z, Sapkota Y, Burridge PW. Functional Validation of Doxorubicin-Induced Cardiotoxicity-Related Genes. JACC CardioOncol 2024; 6:38-50. [PMID: 38510289 PMCID: PMC10950437 DOI: 10.1016/j.jaccao.2023.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 11/16/2023] [Accepted: 11/21/2023] [Indexed: 03/22/2024] Open
Abstract
Background Genome-wide association studies and candidate gene association studies have identified more than 180 genetic variants statistically associated with anthracycline-induced cardiotoxicity (AIC). However, the lack of functional validation has hindered the clinical translation of these findings. Objectives The aim of this study was to functionally validate all genes associated with AIC using human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Methods Through a systemic literature search, 80 genes containing variants significantly associated with AIC were identified. Additionally, 3 more genes with potential roles in AIC (GSTM1, CBR1, and ERBB2) were included. Of these, 38 genes exhibited expression in human fetal heart, adult heart, and hiPSC-CMs. Using clustered regularly interspaced short palindromic repeats/Cas9-based genome editing, each of these 38 genes was systematically knocked out in control hiPSC-CMs, and the resulting doxorubicin-induced cardiotoxicity (DIC) phenotype was assessed using hiPSC-CMs. Subsequently, functional assays were conducted for each gene knockout on the basis of hypothesized mechanistic implications in DIC. Results Knockout of 26 genes increased the susceptibility of hiPSC-CMs to DIC. Notable genes included efflux transporters (ABCC10, ABCC2, ABCB4, ABCC5, and ABCC9), well-established DIC-associated genes (CBR1, CBR3, and RAC2), and genome-wide association study-discovered genes (RARG and CELF4). Conversely, knockout of ATP2B1, HNMT, POR, CYBA, WDR4, and COL1A2 had no significant effect on the in vitro DIC phenotype of hiPSC-CMs. Furthermore, knockout of the uptake transporters (SLC28A3, SLC22A17, and SLC28A1) demonstrated a protective effect against DIC. Conclusions The present findings establish a comprehensive platform for the functional validation of DIC-associated genes, providing insights for future studies in DIC variant associations and potential mechanistic targets for the development of cardioprotective drugs.
Collapse
Affiliation(s)
- Hananeh Fonoudi
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Mariam Jouni
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Romina B. Cejas
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Tarek Magdy
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Malorie Blancard
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Ning Ge
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Disheet A. Shah
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Davi M. Lyra-Leite
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Achal Neupane
- Department of Epidemiology and Cancer Control, St. Jude Children’s Hospital, Memphis, Tennessee, USA
| | - Mennat Gharib
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Zhengxin Jiang
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Yadav Sapkota
- Department of Epidemiology and Cancer Control, St. Jude Children’s Hospital, Memphis, Tennessee, USA
| | - Paul W. Burridge
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
5
|
Xu G, Hao Z, Xiao W, Tan R, Yuan M, Xia Y, Liu Y. Zymosan A Improved Doxorubicin-Induced Ventricular Remodeling by Evoking Heightened Cardiac Inflammatory Responses and Healing in Mice. J Am Heart Assoc 2023; 12:e030200. [PMID: 37702058 PMCID: PMC10547282 DOI: 10.1161/jaha.123.030200] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 07/26/2023] [Indexed: 09/14/2023]
Abstract
Background Doxorubicin-induced myocardial injury is reflected by the presence of vacuolization in both clinical and animal models. The lack of scar tissue to replace the vacuolizated cardiomyocytes indicates that insufficient cardiac inflammation and healing occurred following doxorubicin injection. Whether improved macrophage activity by zymosan A (zymosan) ameliorates doxorubicin-induced ventricular remodeling in mice is unknown. Methods and Results Mice were intravenously injected with vehicle or doxorubicin (5 mg/kg per week, 4 weeks), and cardiac structure and function were assessed by echocardiography. Two distinct macrophage subsets in hearts following doxorubicin injection were measured at different time points by flow cytometry. Moreover, cardiomyocyte vacuolization, capillary density, collagen content, and ventricular tensile strength were assessed. The therapeutic effect of zymosan (3 mg/kg, single injection) on doxorubicin-induced changes in the aforementioned parameters was determined. At the cellular level, the polarization of monocytes to proinflammatory or reparative macrophages were measured, with or without doxorubicin (0.25 and 0.5 μmol/L). Doxorubicin led to less proinflammatory and reparative macrophage infiltration in the heart in the early phase, with decreased cardiac capillary density and collagen III in the chronic phase. In cell culture, doxorubicin (0.5 μmol/L) repressed macrophage transition toward both proinflammatory and reparative subset. Zymosan enhanced both proinflammatory and reparative macrophage infiltration in doxorubicin-injected hearts, evoking a heightened acute inflammatory response. Zymosan alleviated doxorubicin-induced cardiomyocyte vacuolization in the chronic phase, in parallel with enhanced collagen content, capillary density, and ventricular tensile strength. Conclusions Zymosan improved cardiac healing and ameliorated doxorubicin-induced ventricular remodeling and dysfunction by activating macrophages at an optimal time.
Collapse
Affiliation(s)
- Guiwen Xu
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Zhujing Hao
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Wei Xiao
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Ruopeng Tan
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Mengyang Yuan
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Yunlong Xia
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical UniversityDalianChina
- Department of CardiologyThe First Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Yang Liu
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical UniversityDalianChina
| |
Collapse
|
6
|
Ardelean AM, Olariu IC, Isac R, Jurac R, Stolojanu C, Murariu M, Toma AO, Braescu L, Mavrea A, Doros G. Correlation of Speckle-Tracking Echocardiography with Traditional Biomarkers in Predicting Cardiotoxicity among Pediatric Hemato-Oncology Patients: A Comprehensive Evaluation of Anthracycline Dosages and Treatment Protocols. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1479. [PMID: 37761440 PMCID: PMC10528093 DOI: 10.3390/children10091479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/14/2023] [Accepted: 08/24/2023] [Indexed: 09/29/2023]
Abstract
Speckle tracking-echocardiography (STE) is a novel non-invasive imaging tool capable of quantifying myocardial deformation, and thus holds promise in detecting early subclinical myocardial injury. This study aimed to evaluate the correlation of STE with traditional biomarkers in predicting anthracycline-induced cardiotoxicity in the context of varying dosages and treatment protocols in pediatric hemato-oncology patients. We conducted a retrospective study involving pediatric hemato-oncology patients undergoing anthracycline-based chemotherapy. A total of 99 patients were included in the final analysis, with 82 receiving Doxorubicin, of which 58.5% were males, and 17 receiving Epirubicin, of which 70.6% were males, with a median of 10 years old. Traditional biomarkers, such as Troponin I (cTnI) and B-type natriuretic peptide (BNP), were compared with STE parameters, including the global longitudinal strain (GLS), Simpson method of discs (SMOD), and myocardial performance index (MPI). A comprehensive evaluation was conducted based on different dosages of anthracyclines and different treatment protocols, with a follow-up period of one year post-chemotherapy. It was observed that the cTnI levels in the Doxorubicin group were significantly higher (3.2 ng/mL, p = 0.002) than in the Epirubicin group (2.7 ng/mL). However, BNP and NT-proBNP levels were not significantly different between the two groups (p = 0.096 and p = 0.172, respectively). Regarding STE parameters, a significant negative correlation was observed between the anthracycline dose and GLS (Rho = -0.411, p = 0.001), indicating increased cardiotoxicity with dose elevation. The SMOD and MPI gave significantly better values in the Epirubicin group (59.2 and 0.41 vs. 54.4 and 0.36, respectively). However, the ROC analysis did not find GLS, SMOD, or MPI to be significant independent predictors of cardiotoxicity (p > 0.05). There was also considerable variation in cardiotoxicity between the Doxorubicin and Epirubicin study groups, suggesting that the risk of cardiotoxicity is not solely determined by dose. Our study underlines the potential of STE as a sensitive tool for the early detection and prediction of anthracycline-induced cardiotoxicity in pediatric hemato-oncology patients, but only in association with the clinical findings and cardiac biomarkers. While traditional biomarkers still play a role, STE can offer a more accurate prediction of cardiac risk, potentially leading to better management and outcomes for these patients.
Collapse
Affiliation(s)
- Andrada Mara Ardelean
- Department of Pediatrics, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania; (A.M.A.); (I.C.O.); (R.I.); (R.J.); (G.D.)
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania; (C.S.); (M.M.); (L.B.)
- Louis Turcanu Emergency Hospital for Children, Iosif Nemoianu Street 2, 300011 Timisoara, Romania
| | - Ioana Cristina Olariu
- Department of Pediatrics, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania; (A.M.A.); (I.C.O.); (R.I.); (R.J.); (G.D.)
- Louis Turcanu Emergency Hospital for Children, Iosif Nemoianu Street 2, 300011 Timisoara, Romania
| | - Raluca Isac
- Department of Pediatrics, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania; (A.M.A.); (I.C.O.); (R.I.); (R.J.); (G.D.)
- Louis Turcanu Emergency Hospital for Children, Iosif Nemoianu Street 2, 300011 Timisoara, Romania
| | - Ruxandra Jurac
- Department of Pediatrics, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania; (A.M.A.); (I.C.O.); (R.I.); (R.J.); (G.D.)
- Louis Turcanu Emergency Hospital for Children, Iosif Nemoianu Street 2, 300011 Timisoara, Romania
| | - Cristiana Stolojanu
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania; (C.S.); (M.M.); (L.B.)
- Louis Turcanu Emergency Hospital for Children, Iosif Nemoianu Street 2, 300011 Timisoara, Romania
| | - Mircea Murariu
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania; (C.S.); (M.M.); (L.B.)
- Louis Turcanu Emergency Hospital for Children, Iosif Nemoianu Street 2, 300011 Timisoara, Romania
| | - Ana-Olivia Toma
- Department of Dermatology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania;
| | - Laurentiu Braescu
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania; (C.S.); (M.M.); (L.B.)
- Department of Cardiovascular Surgery, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania
- Center for Translational Research and Systems Medicine (CERT-MEDS), “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Adelina Mavrea
- Department of Internal Medicine I, Cardiology Clinic, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Gabriela Doros
- Department of Pediatrics, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania; (A.M.A.); (I.C.O.); (R.I.); (R.J.); (G.D.)
- Louis Turcanu Emergency Hospital for Children, Iosif Nemoianu Street 2, 300011 Timisoara, Romania
| |
Collapse
|
7
|
Reinal I, Ontoria-Oviedo I, Selva M, Casini M, Peiró-Molina E, Fambuena-Santos C, Climent AM, Balaguer J, Cañete A, Mora J, Raya Á, Sepúlveda P. Modeling Cardiotoxicity in Pediatric Oncology Patients Using Patient-Specific iPSC-Derived Cardiomyocytes Reveals Downregulation of Cardioprotective microRNAs. Antioxidants (Basel) 2023; 12:1378. [PMID: 37507917 PMCID: PMC10376252 DOI: 10.3390/antiox12071378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 06/25/2023] [Accepted: 06/29/2023] [Indexed: 07/30/2023] Open
Abstract
Anthracyclines are widely used in the treatment of many solid cancers, but their efficacy is limited by cardiotoxicity. As the number of pediatric cancer survivors continues to rise, there has been a concomitant increase in people living with anthracycline-induced cardiotoxicity. Accordingly, there is an ongoing need for new models to better understand the pathophysiological mechanisms of anthracycline-induced cardiac damage. Here we generated induced pluripotent stem cells (iPSCs) from two pediatric oncology patients with acute cardiotoxicity induced by anthracyclines and differentiated them to ventricular cardiomyocytes (hiPSC-CMs). Comparative analysis of these cells (CTX hiPSC-CMs) and control hiPSC-CMs revealed that the former were significantly more sensitive to cell injury and death from the anthracycline doxorubicin (DOX), as measured by viability analysis, cleaved caspase 3 expression, oxidative stress, genomic and mitochondrial damage and sarcomeric disorganization. The expression of several mRNAs involved in structural integrity and inflammatory response were also differentially affected by DOX. Functionally, optical mapping analysis revealed higher arrythmia complexity after DOX treatment in CTX iPSC-CMs. Finally, using a panel of previously identified microRNAs associated with cardioprotection, we identified lower levels of miR-22-3p, miR-30b-5p, miR-90b-3p and miR-4732-3p in CTX iPSC-CMs under basal conditions. Our study provides valuable phenotype information for cellular models of cardiotoxicity and highlights the significance of using patient-derived cardiomyocytes for studying the associated pathogenic mechanisms.
Collapse
Affiliation(s)
- Ignacio Reinal
- Regenerative Medicine and Heart Transplantation Unit, Health Research Institute Hospital la Fe, 46026 Valencia, Spain
| | - Imelda Ontoria-Oviedo
- Regenerative Medicine and Heart Transplantation Unit, Health Research Institute Hospital la Fe, 46026 Valencia, Spain
| | - Marta Selva
- Regenerative Medicine and Heart Transplantation Unit, Health Research Institute Hospital la Fe, 46026 Valencia, Spain
| | - Marilù Casini
- Regenerative Medicine and Heart Transplantation Unit, Health Research Institute Hospital la Fe, 46026 Valencia, Spain
| | - Esteban Peiró-Molina
- Regenerative Medicine and Heart Transplantation Unit, Health Research Institute Hospital la Fe, 46026 Valencia, Spain
- Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain
| | | | - Andreu M Climent
- ITACA Institute, Universitat Politècnica de València, 46026 Valencia, Spain
| | - Julia Balaguer
- Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain
- Transtational Research in Cancer Unit-Pediatric Oncology, Health Research Institute Hospital La Fe, 46026 Valencia, Spain
| | - Adela Cañete
- Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain
- Transtational Research in Cancer Unit-Pediatric Oncology, Health Research Institute Hospital La Fe, 46026 Valencia, Spain
- Department of Pediatrics, University of Valencia, 46010 Valencia, Spain
| | - Jaume Mora
- Oncology Service, Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
| | - Ángel Raya
- Regenerative Medicine Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, 08908 Barcelona, Spain
- Program for Clinical Translation of Regenerative Medicine in Catalonia-P-[CMRC], L'Hospitalet de Llobregat, 08908 Barcelona, Spain
- Centro de Investigación Biomédica en Red Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Carlos III Institute of Health, 28029 Madrid, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
| | - Pilar Sepúlveda
- Regenerative Medicine and Heart Transplantation Unit, Health Research Institute Hospital la Fe, 46026 Valencia, Spain
- Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Carlos III Institute of Health, 28029 Madrid, Spain
- Department of Pathology, University of Valencia, 46010 Valencia, Spain
| |
Collapse
|
8
|
Bertorello N, Luksch R, Bisogno G, Haupt R, Spallarossa P, Cenna R, Fagioli F. Cardiotoxicity in children with cancer treated with anthracyclines: A position statement on dexrazoxane. Pediatr Blood Cancer 2023; 70:e30515. [PMID: 37355856 DOI: 10.1002/pbc.30515] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 06/09/2023] [Accepted: 06/10/2023] [Indexed: 06/26/2023]
Abstract
Cardiovascular disease is the leading cause of non-malignant morbidity and mortality in childhood cancer survivors (CCSs). Anthracyclines are included in many treatment regimens for paediatric cancer, but unfortunately, these compounds are cardiotoxic. One in 10 CCSs who has received an anthracycline will develop a symptomatic cardiac event over time. Given the crucial need to mitigate anthracycline-related cardiotoxicity (ARC), the authors critically examined published data to identify effective cardioprotective strategies. Based on their expert analysis of contemporary literature data, it was concluded that consideration should be given for routine use of dexrazoxane in children with cancer who are at risk of ARC.
Collapse
Affiliation(s)
- Nicoletta Bertorello
- Paediatric Oncology Division, Regina Margherita Children's Hospital, AOU Città della Salute e della Scienza, Turin, Italy
| | - Roberto Luksch
- Paediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Gianni Bisogno
- Hematology and Oncology Division, Department of Women's and Children's Health, University of Padova, Padua, Italy
| | - Riccardo Haupt
- Epidemiology and Biostatistics Unit and DOPO clinic, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Paolo Spallarossa
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Rosita Cenna
- Paediatric Oncology Division, Regina Margherita Children's Hospital, AOU Città della Salute e della Scienza, Turin, Italy
| | - Franca Fagioli
- Paediatric Oncology Division, Regina Margherita Children's Hospital, AOU Città della Salute e della Scienza, Turin, Italy
- University of Turin, Turin, Italy
| |
Collapse
|
9
|
Rahimi P, Barootkoob B, ElHashash A, Nair A. Efficacy of Dexrazoxane in Cardiac Protection in Pediatric Patients Treated With Anthracyclines. Cureus 2023; 15:e37308. [PMID: 37182052 PMCID: PMC10166653 DOI: 10.7759/cureus.37308] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2023] [Indexed: 05/16/2023] Open
Abstract
Cancer is one of the leading causes of morbidity and mortality in the pediatric population with the most common cancer being acute lymphoblastic leukemia. One of the most common drugs used in the treatment is the anthracycline group of chemotherapeutic agents, and a major side effect is cardiotoxicity. Dexrazoxane, a member of the cardioprotective agents' group of medications, is the only current FDA-approved medication to tackle cardiotoxicity. The mechanism of action in which dexrazoxane is cardioprotective is by halting necroptosis in cardiomyocytes after anthracycline therapy and concurrently binds with iron and reduces the formation of anthracycline-iron complexes and reactive oxygen species. The efficacy of dexrazoxane has been demonstrated in clinical trials within the pediatric population with roughly 60%-80% reduction in risk of developing cardiotoxicity with a very tolerable and limited side effect profile. Further research is required to not only establish the efficacy of dexrazoxane within the pediatric population but also to explore other medications that may serve alongside the function of dexrazoxane.
Collapse
Affiliation(s)
- Parya Rahimi
- Medicine, First Faculty of Medicine, Charles University, Prague, CZE
| | | | - Ahmed ElHashash
- Medicine, First Faculty of Medicine, Charles University, Prague, CZE
| | - Arun Nair
- Pediatrics, Saint Peter's University Hospital, Somerset, USA
| |
Collapse
|
10
|
Moossavi M, Lu X, Herrmann J, Xu X. Molecular mechanisms of anthracycline induced cardiotoxicity: Zebrafish come into play. Front Cardiovasc Med 2023; 10:1080299. [PMID: 36970353 PMCID: PMC10036604 DOI: 10.3389/fcvm.2023.1080299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 02/22/2023] [Indexed: 03/12/2023] Open
Abstract
Anthracyclines are among the most potent chemotherapeutics; however, cardiotoxicity significantly restricts their use. Indeed, anthracycline-induced cardiotoxicity (AIC) fares among the worst types of cardiomyopathy, and may only slowly and partially respond to standard heart failure therapies including β-blockers and ACE inhibitors. No therapy specifically designed to treat anthracycline cardiomyopathy at present, and neither is it known if any such strategy could be developed. To address this gap and to elucidate the molecular basis of AIC with a therapeutic goal in mind, zebrafish has been introduced as an in vivo vertebrate model about a decade ago. Here, we first review our current understanding of the basic molecular and biochemical mechanisms of AIC, and then the contribution of zebrafish to the AIC field. We summarize the generation of embryonic zebrafish AIC models (eAIC) and their use for chemical screening and assessment of genetic modifiers, and then the generation of adult zebrafish AIC models (aAIC) and their use for discovering genetic modifiers via forward mutagenesis screening, deciphering spatial-temporal-specific mechanisms of modifier genes, and prioritizing therapeutic compounds via chemical genetic tools. Several therapeutic target genes and related therapies have emerged, including a retinoic acid (RA)-based therapy for the early phase of AIC and an autophagy-based therapy that, for the first time, is able to reverse cardiac dysfunction in the late phase of AIC. We conclude that zebrafish is becoming an important in vivo model that would accelerate both mechanistic studies and therapeutic development of AIC.
Collapse
Affiliation(s)
- Maryam Moossavi
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Xiaoguang Lu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Joerg Herrmann
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Xiaolei Xu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Correspondence: Xiaolei Xu
| |
Collapse
|
11
|
Development of a Dose-Adjusted Polygenic Risk Model for Anthracycline-Induced Cardiotoxicity. Ther Drug Monit 2023; 45:337-344. [PMID: 36728273 DOI: 10.1097/ftd.0000000000001077] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 12/02/2022] [Indexed: 02/03/2023]
Abstract
BACKGROUND Anthracyclines, which are effective chemotherapeutic agents, cause cardiac dysfunction in up to 57% of patients. The cumulative anthracycline dose is a crucial predictor of cardiotoxicity; however, the cumulative dose alone cannot explain all cardiotoxic events. Strongly associated genetic variants in SLC28A3, UGT1A6, and RARG contribute to anthracycline-induced cardiotoxicity in pediatric patients and may help identify those most susceptible. This study aimed to examine how these pharmacogenetic effects are modulated by cumulative anthracycline doses in the development of cardiotoxicity. METHODS A total of 595 anthracycline-treated children were genotyped and cardiotoxicity cases were identified. A dose-stratified analysis was performed to compare the contributions of SLC28A3 rs7853758, UGT1A6 rs17863783, and RARG rs2229774 variants to the development of cardiotoxicity in low-dose (<150 mg/m2 cumulative dose) and high-dose (>250 mg/m2 cumulative dose) patient groups. Logistic regression was used to model the relationships between the cumulative anthracycline dose, genetic variants, and cardiotoxicity in the full cohort. RESULTS At < 150 mg/m2 cumulative anthracycline dose, the SLC28A3 protective variant did not reach statistical significance [odds ratio (OR) 0.46 (95% confidence interval (CI) 0.10-1.45), P = 0.23], but it was statistically significant at doses >250 mg/m2 [OR 0.43 (95% CI 0.22-0.78), P = 0.0093]. Conversely, the UGT1A6 and RARG risk variants were either statistically significant or approaching significance at doses <150 mg/m2 [OR 7.18 (95% CI 1.78-28.4), P = 0.0045 for UGT1A6 and OR 2.76 (95% CI 0.89-7.63), P = 0.057 for RARG], but not at doses >250 mg/m2 [OR 2.91 (95% CI 0.80-11.0), P = 0.10; OR 1.56 (95% CI 0.89-2.75), P = 0.12]. CONCLUSIONS These findings suggest that the SLC28A3 variant imparts more significant protection for patients receiving higher anthracycline doses, whereas the UGT1A6 and RARG risk variants significantly increased the risk of cardiotoxicity at low anthracycline doses.
Collapse
|
12
|
Disclosing an In-Frame Deletion of the Titin Gene as the Possible Predisposing Factor of Anthracycline-Induced Cardiomyopathy: A Case Report. Int J Mol Sci 2022; 23:ijms23169261. [PMID: 36012532 PMCID: PMC9408849 DOI: 10.3390/ijms23169261] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/02/2022] [Accepted: 08/16/2022] [Indexed: 12/31/2022] Open
Abstract
Anthracycline-induced cardiomyopathy has been noted as a non-neglectable issue in the field of clinical oncology. Remarkable progress has been achieved in searching for inherited susceptible genetic deficits underlying anthracycline cardiotoxicity in the past several years. In this case report, we present the preliminary results of a genetic study in a young male patient who was treated with standard dose anthracycline-based chemotherapy for his acute myeloid leukemia and attacked by acute congestive heart failure after just two courses of therapy. After a survey of 76 target genes, an in-frame deletion of the titin gene was recognized as the most possible genetic defect responsible for his cardiomyopathy caused by anthracycline. This defect proved to pass down from the patient′s mother and did not exist in seven unrelated chemotherapy-treated cancer patients without chemotherapy-induced cardiomyopathy and four other healthy volunteer DNA donors.
Collapse
|
13
|
Genetic Susceptibility and Mechanisms Underlying the Pathogenesis of Anthracycline-Associated Cardiotoxicity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5818612. [PMID: 35965684 PMCID: PMC9365594 DOI: 10.1155/2022/5818612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/18/2022] [Accepted: 07/22/2022] [Indexed: 11/18/2022]
Abstract
Anthracyclines are chemotherapeutic agents widely used to treat a variety of cancers, and these drugs have revolutionized our management of cancer patients. The dose-dependent cardiotoxicity of anthracyclines, however, remains one of the leading causes of chemotherapy treatment-associated mortality in cancer survivors. Patient threshold doses leading to anthracycline-induced cardiotoxicity (AIC) are highly variable among affected patients. This variability is largely ascribed to genetic variants in individuals' genomes. Here, we briefly discuss the prevailing mechanisms underlying the pathogenesis of AIC, and then, we review the genetic variants, mostly identified through human genetic approaches and identified in cancer survivors. The identification of all genetic susceptibilities and elucidation of underlying mechanisms of AIC can help improve upfront risk prediction assessment for potentially severe cardiotoxicity disease and provide valuable insights into the understanding of AIC pathophysiology, which can be further leveraged to develop targeted pharmacogenetic therapies for those at high risk.
Collapse
|
14
|
Aloise DM, Memon A, Zaldivar A. Diquat Herbicide Organophosphate Poisoning and Multi-Organ Failure: A Case Report. Cureus 2022; 14:e27241. [PMID: 36035055 PMCID: PMC9399667 DOI: 10.7759/cureus.27241] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2022] [Indexed: 11/20/2022] Open
Abstract
A 63-year-old male landscaper presented to our emergency department (ED) following unintentional ingestion of an herbicide including diquat, a highly lethal toxin. Poison control was consulted, and treatment was centered around emergency hemodialysis to mitigate the nephrotoxic effects of diquat. Unfortunately, our patient did not survive. Unlike most organophosphate poisonings, diquat facilitates the formation of catastrophic amounts of reactive oxygen species leading to lethal consequences for affected cells. The body’s most affected sites are the kidneys, liver, and lungs. Our patient’s accidental diquat consumption illuminates the need for additional research to be done. This case report emphasizes the importance of identifying effective treatments for diquat and similar poisonings that lead to oxidative stress. We seek to describe this uncommon instance of accidental diquat ingestion, including the unique therapeutic regimen, and course of disease progression.
Collapse
|
15
|
Senekovič Kojc T, Marčun Varda N. Novel Biomarkers of Heart Failure in Pediatrics. CHILDREN 2022; 9:children9050740. [PMID: 35626917 PMCID: PMC9139970 DOI: 10.3390/children9050740] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/11/2022] [Accepted: 05/16/2022] [Indexed: 02/07/2023]
Abstract
Novel biomarkers of heart failure are the subject of numerous studies. Biomarkers of heart failure can be determined in the blood and in the urine. Seven groups of biomarkers of heart failure based on pathophysiological mechanisms are presented in this review, namely biomarkers of myocardial stretch, myocyte injury, myocardial remodeling, biomarkers of inflammation, renal dysfunction, neurohumoral activation, and oxidative stress. Studies of biomarkers in the pediatric population are scarce, therefore, further investigation is needed for reliable prognostic and therapeutic implications. The future of biomarker use is in multimarker panels that include a combination of biomarkers with different pathophysiological mechanisms in order to improve their diagnostic and prognostic predictive value.
Collapse
Affiliation(s)
- Teja Senekovič Kojc
- Department of Perinatology, University Medical Centre Maribor, Ljubljanska 5, 2000 Maribor, Slovenia
- Correspondence:
| | - Nataša Marčun Varda
- Department of Paediatrics, University Medical Centre Maribor, Ljubljanska 5, 2000 Maribor, Slovenia;
- Medical Faculty, University of Maribor, Taborska 8, 2000 Maribor, Slovenia
| |
Collapse
|
16
|
Vargas-Neri JL, Carleton B, Ross CJ, Medeiros M, Castañeda-Hernández G, Clark P. Pharmacogenomic study of anthracycline-induced cardiotoxicity in Mexican pediatric patients. Pharmacogenomics 2022; 23:291-301. [PMID: 35147047 DOI: 10.2217/pgs-2021-0144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Background: The aim of this study was to evaluate the association between well-defined genetic risk variants in SLC28A3, RARG and UGT1A6 and anthracycline-induced cardiotoxicity in Mexican pediatric patients. Methods: We tested a cohort of 79 children treated with anthracyclines for the presence of SLC28A3-rs7853758, RARG-rs2229774 and UGT1A6-rs17863783. Results: The SLC28A3-rs7853758 variant was more frequent in this cohort, while the UGT1A6-rs17863783 and RARG-rs2229774 variants were present at lower frequencies. A clinically important decrease of fractional shortening was associated with SLC28A3-rs7853758 variant. Conclusion: In this cohort, 39.2% of patients carried the protective SLC28A3 variant. A small number of tested patients have the risk variants of UGT1A6 and RARG. None of the patients shared the two risk variants.
Collapse
Affiliation(s)
- Jessica L Vargas-Neri
- Clinical Epidemiology Unit of Hospital Infantil de México Federico Gómez-Medicine Faculty of Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Bruce Carleton
- Department of Pediatrics, Division of Translational Therapeutics, Faculty of Medicine, University of British Columbia & British Columbia Children's Hospital & Research Institute, Vancouver, Canada
| | - Colin J Ross
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, Canada
| | - Mara Medeiros
- Research & Diagnostic Unit of Nephrology & Bone & Mineral Metabolism of Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| | - Gilberto Castañeda-Hernández
- Department of Pharmacology of Centro de Investigación y de Estudios Avanzados of Instituto Politécnico Nacional, Mexico City, Mexico
| | - Patricia Clark
- Clinical Epidemiology Unit of Hospital Infantil de México Federico Gómez-Medicine Faculty of Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
17
|
Hitawala G, Jain E, Castellanos L, Garimella R, Akku R, Chamavaliyathil AK, Irfan H, Jaiswal V, Quinonez J, Dakroub M, Hanif M, Baloch AH, Gomez IS, Dylewski J. Pediatric Chemotherapy Drugs Associated With Cardiotoxicity. Cureus 2021; 13:e19658. [PMID: 34976454 PMCID: PMC8679581 DOI: 10.7759/cureus.19658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/16/2021] [Indexed: 11/05/2022] Open
Abstract
Pediatric cancers are a common cause of childhood morbidity. As a result, chemotherapeutic regimens have been designed to target childhood cancers. These medications are necessary to treat pediatric cancers, however, oncology management options are accompanied by multiple negative and potentially fatal adverse effects. Although anthracyclines are the most commonly used chemotherapeutic agents associated with cardiotoxicity, we also explore other chemotherapeutic drugs used in children that can potentially affect the heart. Genetic variations resulting in single nucleotide polymorphism (SNP) have the propensity to modify the cardiotoxic effects of the chemotherapy drugs. The clinical presentation of the cardiac effects can vary from arrhythmias and heart failure to completely asymptomatic. A range of imaging studies and laboratory investigations can protect the heart from severe outcomes. The physiology of the heart and the effect of drugs in children vary vividly from adults; therefore, it is crucial to study the cardiotoxic effect of chemotherapy drugs in the pediatric population. This review highlights the potential contributing factors for cardiotoxicity in the pediatric population and discusses the identification and management options.
Collapse
Affiliation(s)
- Gazala Hitawala
- Internal Medicine, Jersey City (JC) Medical Center, Orlando, USA
| | - Esha Jain
- Medicine, American University of Antigua, St. John's, ATG
| | | | | | - Radhika Akku
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Adila K Chamavaliyathil
- Pediatrics, Ras Al Khaimah (RAK) Medical and Health Sciences University, Ras Al Khaimah, ARE
| | - Huma Irfan
- Research, Larkin Community Hospital, South Miami, USA
| | | | - Jonathan Quinonez
- Neurology/Osteopathic Neuromuscular Medicine, Larkin Community Hospital, Miami, USA
| | - Maher Dakroub
- Hematology and Oncology, Larkin Community Hospital, South Miami, USA
| | - Muhammad Hanif
- Internal Medicine, Khyber Medical College Peshawar, Hayatabad Medical Complex, Peshawar, PAK
| | - Ali H Baloch
- Research, University of Maryland Medical Center, Baltimore, USA
| | - Ivan S Gomez
- Cardiology, Larkin Community Hospital, South Miami, USA
| | - John Dylewski
- Cardiology, Larkin Community Hospital, South Miami, USA
| |
Collapse
|
18
|
Huang MF, Pang LK, Chen YH, Zhao R, Lee DF. Cardiotoxicity of Antineoplastic Therapies and Applications of Induced Pluripotent Stem Cell-Derived Cardiomyocytes. Cells 2021; 10:2823. [PMID: 34831045 PMCID: PMC8616116 DOI: 10.3390/cells10112823] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/05/2021] [Accepted: 10/15/2021] [Indexed: 01/04/2023] Open
Abstract
The therapeutic landscape for the treatment of cancer has evolved significantly in recent decades, aided by the development of effective oncology drugs. However, many cancer drugs are often poorly tolerated by the body and in particular the cardiovascular system, causing adverse and sometimes fatal side effects that negate the chemotherapeutic benefits. The prevalence and severity of chemotherapy-induced cardiotoxicity warrants a deeper investigation of the mechanisms and implicating factors in this phenomenon, and a consolidation of scientific efforts to develop mitigating strategies. Aiding these efforts is the emergence of induced pluripotent stem cells (iPSCs) in recent years, which has allowed for the generation of iPSC-derived cardiomyocytes (iPSC-CMs): a human-based, patient-derived, and genetically variable platform that can be applied to the study of chemotherapy-induced cardiotoxicity and beyond. After surveying chemotherapy-induced cardiotoxicity and the associated chemotherapeutic agents, we discuss the use of iPSC-CMs in cardiotoxicity modeling, drug screening, and other potential applications. Improvements to the iPSC-CM platform, such as the development of more adult-like cardiomyocytes and ongoing advances in biotechnology, will only enhance the utility of iPSC-CMs in both basic science and clinical applications.
Collapse
Affiliation(s)
- Mo-Fan Huang
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (M.-F.H.); (L.K.P.)
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Lon Kai Pang
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (M.-F.H.); (L.K.P.)
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yi-Hung Chen
- Department and Institute of Pharmacology, National Yang Ming Chiao Tung University, Taipei 112, Taiwan;
| | - Ruiying Zhao
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (M.-F.H.); (L.K.P.)
| | - Dung-Fang Lee
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (M.-F.H.); (L.K.P.)
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
- Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
19
|
Cauli O. Oxidative Stress and Cognitive Alterations Induced by Cancer Chemotherapy Drugs: A Scoping Review. Antioxidants (Basel) 2021; 10:1116. [PMID: 34356349 PMCID: PMC8301189 DOI: 10.3390/antiox10071116] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 07/01/2021] [Accepted: 07/08/2021] [Indexed: 02/06/2023] Open
Abstract
Cognitive impairment is one of the most deleterious effects of chemotherapy treatment in cancer patients, and this problem sometimes remains even after chemotherapy ends. Common classes of chemotherapy-based regimens such as anthracyclines, taxanes, and platinum derivatives can induce both oxidative stress in the blood and in the brain, and these effects can be reproduced in neuronal and glia cell cultures. In rodent models, both the acute and repeated administration of doxorubicin or adriamycin (anthracyclines) or cisplatin impairs cognitive functions, as shown by their diminished performance in different learning and memory behavioural tasks. Administration of compounds with strong antioxidant effects such as N-acetylcysteine, gamma-glutamyl cysteine ethyl ester, polydatin, caffeic acid phenethyl ester, and 2-mercaptoethane sulfonate sodium (MESNA) counteract both oxidative stress and cognitive alterations induced by chemotherapeutic drugs. These antioxidant molecules provide the scientific basis to design clinical trials in patients with the aim of reducing the oxidative stress and cognitive alterations, among other probable central nervous system changes, elicited by chemotherapy in cancer patients. In particular, N-acetylcysteine and MESNA are currently used in clinical settings and are therefore attracting scientific attention.
Collapse
Affiliation(s)
- Omar Cauli
- Frailty and Cognitive Impairment Group (FROG), University of Valencia, 46010 Valencia, Spain; ; Tel.: +34-96-386-41-82; Fax: +34-96-398-30-35
- Department of Nursing, University of Valencia, 46010 Valencia, Spain
| |
Collapse
|
20
|
Gándara-Mireles JA, Lares-Asseff I, Reyes Espinoza EA, Blanco JG, González Font AE, Córdova Hurtado LP, Castañeda VL, Fierro IV, Romero LP, Reyes HA. Association of genetic polymorphisms NCF4 rs1883112, CBR3 rs1056892, and ABCC1 rs3743527 with the cardiotoxic effects of doxorubicin in children with acute lymphoblastic leukemia. Pharmacogenet Genomics 2021; 31:108-115. [PMID: 34096893 DOI: 10.1097/fpc.0000000000000428] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVES Cardiotoxicity is a frequent complication secondary to the use of anthracyclines for cancer chemotherapy. Evidence suggests that certain polymorphic genetic variants modify the risk for anthracycline-related cardiotoxicity. Reports documenting the impact of genetic polymorphisms on anthracycline-cardiotoxicity risk in pediatric patients with cancers from Latin American countries are scarce. The objective of this study was to evaluate associations between NCF4 rs1883112, CBR3 rs1056892 and ABCC1 rs3743527 genotype status and echocardiographic parameters indicative of anthracycline-cardiotoxicity in a group of Mexican children with acute lymphoblastic leukemia (ALL). METHODS Sixty-seven children (2-18 years old) with ALL were treated at the State Cancer Center in Durango, Mexico. NCF4, CBR3, and ABCC1 genotypes were examined by real-time PCR. Left ventricular ejection fraction and diastolic filling ratio were examined as markers of systolic and diastolic anthracycline-toxicity. RESULTS NCF4 rs1883112 genotype status was significantly associated with the risk of doxorubicin cardiotoxicity [odds ratio (OR) = 10.80, 95% confidence interval (CI) 1.69-68.98, P = 0.01]. There was a significant association between heterozygous CBR3 rs1056892 genotype status and anthracycline-cardiotoxicity risk (OR = 9.91, 95% CI 1.07-91.47, P = 0.04). Heterozygosis for the ABCC1 rs3743527 allele was associated with protection from anthracycline-cardiotoxicity (OR = 0.30, 95% CI 0.09-0.91, P = 0.03). CONCLUSION This pilot study suggests that selected polymorphic variants may impact the risk for anthracycline-cardiotoxicity in pediatric patients with ALL treated with a contemporary chemotherapeutic regimen in Mexico.
Collapse
Affiliation(s)
| | - Ismael Lares-Asseff
- Academia de Genómica, Instituto Politécnico Nacional, CIIDIR-Unidad, Durango, Mexico
- Red Latinoamericana de Implementación y Validación de Guías Clínicas Farmacogenómicas (RELIVAF-CYTED)
| | | | - Javier G Blanco
- School of Pharmacy and Pharmaceutical Sciences, University of Buffalo, The State University of New York, USA
| | | | | | | | | | - Leslie Patrón Romero
- Facultad de Medicina y Psicologia de la Universidad Autónoma de Baja California, Tijuana, Mexico
| | - Horacio Almanza Reyes
- Facultad de Medicina y Psicologia de la Universidad Autónoma de Baja California, Tijuana, Mexico
| |
Collapse
|
21
|
Samosir SM, Utamayasa IKA, Andarsini MR, Rahman MA, Ontoseno T, Hidayat T, Ugrasena IDG, Larasati MCS, Cahyadi A. Risk Factors of Daunorubicine Induced Early Cardiotoxicity in Childhood Acute Lymphoblastic Leukemia: A Retrospective Study. Asian Pac J Cancer Prev 2021; 22:1407-1412. [PMID: 34048168 PMCID: PMC8408392 DOI: 10.31557/apjcp.2021.22.5.1407] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Indexed: 11/25/2022] Open
Abstract
Background: Daunorubicine, a type of anthracycline, is a drug commonly used in cancer chemotherapy that increases survival rate but consequently compromises with cardiovascular outcomes in some patients. Thus, preventing the early progression of cardiotoxicity is important to improve the treatment outcome in childhood acute lymhoblastic leukemia (ALL). Objective: The present study aimed to identify the risk factors in anthracycline-induced early cardiotoxicity in childhood ALL. Methods: This retrospective study was conducted by observing ALL-diagnosed children from 2014 to 2019 in Dr. Soetomo General Hospital. There were 49 patients who met the inclusion criteria and were treated with chemotherapy using Indonesian Childhood ALL Protocol 2013. Echocardiography was performed by pediatric cardiologists to compare before and at any given time after anthracycline therapy. Early cardiotoxicity was defined as a decline of left ventricle ejection fraction (LVEF) greater than 10% with a final LVEF < 53% during the first year of anthracycline administration. Risk factors such as sex, age, risk stratification group, and cumulative dose were identified by using multiple logistic regression. Diagnostic performance of cumulative anthracycline dose was evaluated by receiver operating characteristic (ROC) curve. Results: Early anthracycline-induced cardiotoxicity was observed in 5 out of 49 patients. The median cumulative dose of anthracycline was 143.69±72.68 mg/m2. Thirty-three patients experienced a decreasing LVEF. The factors associated with early cardiomyopathy were age of ≥ 4 years (PR= 1.128; 95% CI: 1.015-1.254; p= 0.001), high risk group (PR= 1.135; 95% CI: 1.016-1.269; p= 0.001), and cumulative dose of ≥120 mg / m2 (CI= 1.161; 95% CI:1.019-1.332). Conclusion: Age of ≥ 4 years, risk group, and cumulative dose of ≥120 mg/m2 are significant risk factors for early cardiomyopathy in childhood ALL.
Collapse
Affiliation(s)
- Sunny Mariana Samosir
- Department of Child Health, Faculty of Medicine Universitas Airlangga/Dr Soetomo Academic General Hospital, Surabaya, Indonesia
| | - I Ketut Alit Utamayasa
- Department of Child Health, Faculty of Medicine Universitas Airlangga/Dr Soetomo Academic General Hospital, Surabaya, Indonesia
| | - Mia Ratwita Andarsini
- Department of Child Health, Faculty of Medicine Universitas Airlangga/Dr Soetomo Academic General Hospital, Surabaya, Indonesia
| | - Mahrus A Rahman
- Department of Child Health, Faculty of Medicine Universitas Airlangga/Dr Soetomo Academic General Hospital, Surabaya, Indonesia
| | - Teddy Ontoseno
- Department of Child Health, Faculty of Medicine Universitas Airlangga/Dr Soetomo Academic General Hospital, Surabaya, Indonesia
| | - Taufiq Hidayat
- Department of Child Health, Faculty of Medicine Universitas Airlangga/Dr Soetomo Academic General Hospital, Surabaya, Indonesia
| | - I Dewa Gede Ugrasena
- Department of Child Health, Faculty of Medicine Universitas Airlangga/Dr Soetomo Academic General Hospital, Surabaya, Indonesia
| | - Maria Christina Shanty Larasati
- Department of Child Health, Faculty of Medicine Universitas Airlangga/Dr Soetomo Academic General Hospital, Surabaya, Indonesia
| | - Andi Cahyadi
- Department of Child Health, Faculty of Medicine Universitas Airlangga/Dr Soetomo Academic General Hospital, Surabaya, Indonesia
| |
Collapse
|
22
|
Keung W, Cheung YF. Human Pluripotent Stem Cells for Modeling of Anticancer Therapy-Induced Cardiotoxicity and Cardioprotective Drug Discovery. Front Pharmacol 2021; 12:650039. [PMID: 33953683 PMCID: PMC8090862 DOI: 10.3389/fphar.2021.650039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/01/2021] [Indexed: 12/18/2022] Open
Abstract
Anticancer chemotherapies have been shown to produce severe side effects, with cardiotoxicity from anthracycline being the most notable. Identifying risk factors for anticancer therapy-induced cardiotoxicity in cancer patients as well as understanding its underlying mechanism is essential to improving clinical outcomes of chemotherapy treatment regimens. Moreover, cardioprotective agents against anticancer therapy-induced cardiotoxicity are scarce. Human induced pluripotent stem cell technology offers an attractive platform for validation of potential single nucleotide polymorphism with increased risk for cardiotoxicity. Successful validation of risk factors and mechanism of cardiotoxicity would aid the development of such platform for novel drug discovery and facilitate the practice of personalized medicine.
Collapse
Affiliation(s)
- Wendy Keung
- Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Pokfulam, Hong Kong
| | - Yiu-Fai Cheung
- Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Pokfulam, Hong Kong.,Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| |
Collapse
|
23
|
Cardiotoxicity: A Major Setback in Childhood Leukemia Treatment. DISEASE MARKERS 2021; 2021:8828410. [PMID: 33505537 PMCID: PMC7810535 DOI: 10.1155/2021/8828410] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 11/15/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023]
Abstract
Ongoing research in the field of pediatric oncology has led to an increased number of childhood cancer survivors reaching adulthood. Therefore, ensuring a good quality of life for these patients has become a rising priority. Considering this, the following review focuses on summarizing the most recent research in anthracycline-induced cardiac toxicity in children treated for leukemia. For pediatric cancers, anthracyclines are one of the most used anticancer drugs, with over half of the childhood cancer survivors believed to have been exposed to them. Anthracyclines cause irreversible cardiomyocyte loss, leading to chronic, progressive heart failure. The risk of developing cardiotoxicity has been known to increase with the treatment-free interval and total cumulative dose. However, because of individual variations in anthracycline metabolism, it has recently been shown that there is no risk-free dose. Moreover, studies have shown that diagnosing anthracycline-induced cardiomyopathy in the symptomatic phase is associated with poor treatment response and prognosis. Thus, early and systematic evaluation of these patients is crucial to allow optimal therapeutic intervention. Although currently echocardiographic assessment of left ventricle ejection fraction and cardiac biomarker evaluation are being used for cardiac function monitoring in oncologic patients, there is no established follow-up and treatment protocol for these patients, and these methods are neither specific nor sensitive for identifying early cardiac dysfunction. All things considered, the need for ongoing research in the field of pediatric cardiooncology is crucial to offer these patients a chance at a good quality of life as adults.
Collapse
|
24
|
Visscher H, Otth M, Feijen EAML, Nathan PC, Kuehni CE. Cardiovascular and Pulmonary Challenges After Treatment of Childhood Cancer. Pediatr Clin North Am 2020; 67:1155-1170. [PMID: 33131539 DOI: 10.1016/j.pcl.2020.07.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Childhood cancer survivors are at risk for developing cardiovascular disease and pulmonary disease related to cancer treatment. This might not become apparent until many years after treatment and varies from subclinical to life-threatening disease. Important causes are anthracyclines and radiotherapy involving heart, head, or neck for cardiovascular disease, and bleomycin, busulfan, nitrosoureas, radiation to the chest, and lung or chest surgery for pulmonary disease. Most effects are dose dependent, but genetic risk factors have been discovered. Treatment options are limited. Prevention and regular screening are crucial. Survivors should be encouraged to adopt a healthy lifestyle, and modifiable risk factors should be addressed.
Collapse
Affiliation(s)
- Henk Visscher
- Division of Haematology/Oncology, Hospital for Sick Children, 555 University Avenue, Toronto, Ontario M5G 1X8, Canada; Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, The Netherlands.
| | - Maria Otth
- Childhood Cancer Research Platform, Institute of Social and Preventive Medicine, University of Bern, Mittelstrasse 43, Bern 3012, Switzerland; Division of Hematology-Oncology, Department of Pediatrics, Kantonsspital Aarau, Switzerland
| | - E A M Lieke Feijen
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, The Netherlands
| | - Paul C Nathan
- AfterCare Program, Division of Haematology/Oncology, Hospital for Sick Children, 555 University Avenue, Toronto, Ontario M5G 1X8, Canada
| | - Claudia E Kuehni
- Childhood Cancer Research Platform, Institute of Social and Preventive Medicine, University of Bern, Mittelstrasse 43, Bern 3012, Switzerland; Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Mittelstrasse 43, Bern 3012, Switzerland
| |
Collapse
|
25
|
Liu R, Li D, Sun F, Rampoldi A, Maxwell JT, Wu R, Fischbach P, Castellino SM, Du Y, Fu H, Mandawat A, Xu C. Melphalan induces cardiotoxicity through oxidative stress in cardiomyocytes derived from human induced pluripotent stem cells. Stem Cell Res Ther 2020; 11:470. [PMID: 33153480 PMCID: PMC7643439 DOI: 10.1186/s13287-020-01984-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 10/20/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Treatment-induced cardiotoxicity is a leading noncancer-related cause of acute and late onset morbidity and mortality in cancer patients on antineoplastic drugs such as melphalan-increasing clinical case reports have documented that it could induce cardiotoxicity including severe arrhythmias and heart failure. As the mechanism by which melphalan impairs cardiac cells remains poorly understood, here, we aimed to use cardiomyocytes derived from human induced pluripotent stem cells (hiPSC-CMs) to investigate the cellular and molecular mechanisms of melphalan-induced cardiotoxicity. METHODS hiPSC-CMs were generated and treated with clinically relevant doses of melphalan. To characterize melphalan-induced cardiotoxicity, cell viability and apoptosis were quantified at various treatment durations. Ca2+ transient and contractility analyses were used to examine the alterations of hiPSC-CM function. Proteomic analysis, reactive oxygen species detection, and RNA-Sequencing were conducted to investigate underlying mechanisms. RESULTS Melphalan treatment of hiPSC-CMs induced oxidative stress, caused Ca2+ handling defects and dysfunctional contractility, altered global transcriptomic and proteomic profiles, and resulted in apoptosis and cell death. The antioxidant N-acetyl-L-cysteine attenuated these genomic, cellular, and functional alterations. In addition, several other signaling pathways including the p53 and transforming growth factor-β signaling pathways were also implicated in melphalan-induced cardiotoxicity according to the proteomic and transcriptomic analyses. CONCLUSIONS Melphalan induces cardiotoxicity through the oxidative stress pathway. This study provides a unique resource of the global transcriptomic and proteomic datasets for melphalan-induced cardiotoxicity and can potentially open up new clinical mechanism-based targets to prevent and treat melphalan-induced cardiotoxicity.
Collapse
Affiliation(s)
- Rui Liu
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA, 30322, USA
- Department of Pediatrics, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, China
| | - Dong Li
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA, 30322, USA
| | - Fangxu Sun
- School of Chemistry and Biochemistry and the Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Antonio Rampoldi
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA, 30322, USA
| | - Joshua T Maxwell
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA, 30322, USA
| | - Ronghu Wu
- School of Chemistry and Biochemistry and the Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Peter Fischbach
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA, 30322, USA
| | - Sharon M Castellino
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA, 30322, USA
| | - Yuhong Du
- Emory Chemical Biology Discovery Center and the Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Haian Fu
- Emory Chemical Biology Discovery Center and the Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Anant Mandawat
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Cardio-Oncology Program, Winship Cancer Institute of Emory University, Atlanta, GA, 30322, USA
| | - Chunhui Xu
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA, 30322, USA.
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
26
|
Petrykey K, Andelfinger GU, Laverdière C, Sinnett D, Krajinovic M. Genetic factors in anthracycline-induced cardiotoxicity in patients treated for pediatric cancer. Expert Opin Drug Metab Toxicol 2020; 16:865-883. [DOI: 10.1080/17425255.2020.1807937] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Kateryna Petrykey
- Immune Diseases and Cancer, Sainte-Justine University Health Center (SJUHC), Montreal, Quebec, Canada
- Department of Pharmacology and Physiology, Université De Montréal (Quebec), Montreal, Canada
| | - Gregor U. Andelfinger
- Department of Pediatrics, Université De Montréal (Quebec), Canada
- Fetomaternal and Neonatal Pathologies, Sainte-JustineUniversity Health Center (SJUHC), Montreal, Quebec, Canada
| | - Caroline Laverdière
- Immune Diseases and Cancer, Sainte-Justine University Health Center (SJUHC), Montreal, Quebec, Canada
- Department of Pediatrics, Université De Montréal (Quebec), Canada
| | - Daniel Sinnett
- Immune Diseases and Cancer, Sainte-Justine University Health Center (SJUHC), Montreal, Quebec, Canada
- Department of Pediatrics, Université De Montréal (Quebec), Canada
| | - Maja Krajinovic
- Immune Diseases and Cancer, Sainte-Justine University Health Center (SJUHC), Montreal, Quebec, Canada
- Department of Pharmacology and Physiology, Université De Montréal (Quebec), Montreal, Canada
- Department of Pediatrics, Université De Montréal (Quebec), Canada
| |
Collapse
|
27
|
Leerink JM, de Baat EC, Feijen EA, Bellersen L, van Dalen EC, Grotenhuis HB, Kapusta L, Kok WE, Loonen J, van der Pal HJ, Pluijm SM, Teske AJ, Mavinkurve-Groothuis AM, Merkx R, Kremer LC. Cardiac Disease in Childhood Cancer Survivors: Risk Prediction, Prevention, and Surveillance: JACC CardioOncology State-of-the-Art Review. JACC CardioOncol 2020; 2:363-378. [PMID: 34396245 PMCID: PMC8352294 DOI: 10.1016/j.jaccao.2020.08.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/07/2020] [Accepted: 08/08/2020] [Indexed: 02/06/2023] Open
Abstract
Cardiac diseases in the growing population of childhood cancer survivors are of major concern. Cardiotoxicity as a consequence of anthracyclines and chest radiotherapy continues to be relevant in the modern treatment era. Mitoxantrone has emerged as an important treatment-related risk factor and evidence on traditional cardiovascular risk factors in childhood cancer survivors is accumulating. International surveillance guidelines have been developed with the aim to detect and manage cardiac diseases early and prevent symptomatic disease. There is growing interest in risk prediction models to individualize prevention and surveillance. This State-of-the-Art Review summarizes literature from a systematic PubMed search focused on cardiac diseases after treatment for childhood cancer. Here, we discuss the prevalence, risk factors, prevention, risk prediction, and surveillance of cardiac diseases in survivors of childhood cancer.
Collapse
Key Words
- CAD, coronary artery disease
- CCS, childhood cancer survivors
- ECG, electrocardiogram
- FS, fractional shortening
- GLS, global longitudinal strain
- IGHG, International Late Effects of Childhood Cancer Guideline Harmonization Group
- LV, left ventricle
- LVEF, left ventricular ejection fraction
- RCT, randomized controlled trial
- cardiotoxicity
- cardiovascular risk factors
- chest RT, chest-directed radiotherapy
- childhood cancer survivors
- prevention
- risk prediction
Collapse
Affiliation(s)
- Jan M. Leerink
- Department of Cardiology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Esmée C. de Baat
- Department of Pediatric Oncology, Princess Máxima Center, Utrecht, the Netherlands
| | | | - Louise Bellersen
- Department of Cardiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Elvira C. van Dalen
- Department of Pediatric Oncology, Princess Máxima Center, Utrecht, the Netherlands
| | - Heynric B. Grotenhuis
- Department of Pediatric Cardiology, Wilhelmina Children's Hospital, University of Utrecht, Utrecht, the Netherlands
| | - Livia Kapusta
- Department of Pediatric Cardiology, Radboud University Medical Center, Amalia Children’s Hospital, Nijmegen, the Netherlands
- Department of Pediatrics, Tel Aviv University, Sackler School of Medicine, Tel Aviv Sourasky Medical Center, Pediatric Cardiology Unit, Tel Aviv, Israel
| | - Wouter E.M. Kok
- Department of Cardiology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Jacqueline Loonen
- Department of Pediatric Hematology and Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Saskia M.F. Pluijm
- Department of Pediatric Oncology, Princess Máxima Center, Utrecht, the Netherlands
| | - Arco J. Teske
- Department of Cardiology, Utrecht University Medical Center, Utrecht, the Netherlands
| | | | - Remy Merkx
- Department of Medical Imaging, Radboud University Medical Center, Medical UltraSound Imaging Center, Nijmegen, the Netherlands
| | - Leontien C.M. Kremer
- Department of Pediatric Oncology, Princess Máxima Center, Utrecht, the Netherlands
| |
Collapse
|
28
|
Genova E, Stocco G, Decorti G. Induced pluripotent stem cells to model adverse drug reactions in pediatric patients. Pharmacogenomics 2020; 21:975-978. [PMID: 32893744 DOI: 10.2217/pgs-2020-0082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 07/03/2020] [Indexed: 11/21/2022] Open
Affiliation(s)
- Elena Genova
- Institute for Maternal & Child Health, IRCCS Burlo Garofolo, Trieste, 34137, Italy
| | - Gabriele Stocco
- Department of Life Sciences, University of Trieste, Trieste, 34127, Italy
| | - Giuliana Decorti
- Institute for Maternal & Child Health, IRCCS Burlo Garofolo, Trieste, 34137, Italy
- Department of Medical, Surgical & Health Sciences, University of Trieste, Trieste, 34127, Italy
| |
Collapse
|
29
|
McOwan TN, Craig LA, Tripdayonis A, Karavendzas K, Cheung MM, Porrello ER, Conyers R, Elliott DA. Evaluating anthracycline cardiotoxicity associated single nucleotide polymorphisms in a paediatric cohort with early onset cardiomyopathy. CARDIO-ONCOLOGY 2020; 6:5. [PMID: 32477593 PMCID: PMC7243302 DOI: 10.1186/s40959-020-00060-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 04/29/2020] [Indexed: 12/25/2022]
Abstract
Background Anthracyclines are a mainstay of chemotherapy. However, a relatively frequent adverse outcome of anthracycline treatment is cardiomyopathy. Multiple genetic studies have begun to dissect the complex genetics underlying cardiac sensitivity to the anthracycline drug class. A number of single nucleotide polymorphisms (SNPs) have been identified to be in linkage disequilibrium with anthracycline induced cardiotoxicity in paediatric populations. Methods Here we screened for the presence of SNPs resulting in a missense coding change in a cohort of children with early onset chemotherapy related cardiomyopathy. The SNP identity was evaluated by Sanger sequencing of PCR amplicons from genomic DNA of patients with anthracycline related cardiac dysfunction. Results All of the published SNPs were observed within our patient group. There was no correlation between the number of missense variants an individual carried with severity of disease. Furthermore, the time to cardiac disease onset post-treatment was not greater in those individuals carrying a high load of SNPs resulting from missense variants. Conclusions We conclude that previously identified missense SNPs are present within a paediatric cohort with early onset heart damage induced by anthracyclines. However, these SNPs require further replication cohorts and functional validation before being deployed to assess anthracycline cardiotoxicity risk in the clinic.
Collapse
Affiliation(s)
- Timothy N McOwan
- 1Murdoch Children's Research Institute, The Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052 Australia.,2Department of Pediatrics, The Royal Children's Hospital, University of Melbourne, Parkville, Victoria 3052 Australia
| | - Lauren A Craig
- 1Murdoch Children's Research Institute, The Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052 Australia
| | - Anne Tripdayonis
- 1Murdoch Children's Research Institute, The Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052 Australia
| | - Kathy Karavendzas
- 1Murdoch Children's Research Institute, The Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052 Australia
| | - Michael M Cheung
- 1Murdoch Children's Research Institute, The Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052 Australia.,2Department of Pediatrics, The Royal Children's Hospital, University of Melbourne, Parkville, Victoria 3052 Australia
| | - Enzo R Porrello
- 1Murdoch Children's Research Institute, The Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052 Australia.,3Department of Physiology, School of Biomedical Sciences, The University of Melbourne, Parkville, Victoria 3010 Australia
| | - Rachel Conyers
- 1Murdoch Children's Research Institute, The Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052 Australia.,2Department of Pediatrics, The Royal Children's Hospital, University of Melbourne, Parkville, Victoria 3052 Australia
| | - David A Elliott
- 1Murdoch Children's Research Institute, The Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052 Australia.,2Department of Pediatrics, The Royal Children's Hospital, University of Melbourne, Parkville, Victoria 3052 Australia.,4Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800 Australia
| |
Collapse
|
30
|
Wiernikowski JT, Bernhardt MB. Review of nutritional status, body composition, and effects of antineoplastic drug disposition. Pediatr Blood Cancer 2020; 67 Suppl 3:e28207. [PMID: 32083372 DOI: 10.1002/pbc.28207] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 01/22/2020] [Accepted: 01/23/2020] [Indexed: 12/22/2022]
Abstract
The overall survival for children with cancer in high income countries is excellent. However, there are many disparities that may negatively affect survival, which are particularly problematic in low income countries, such as nutritional status at diagnosis and throughout therapy. Nutritional status as well as concomitant foods, supplements, and medications may play a role in overall exposure and response to chemotherapy. Emerging science around the microbiome may also play a role and should be further explored as a contributor to disease progression and therapeutic response. This article highlights some of these issues and proposes additional areas of research relevant to nutritional status and pharmacology that are needed in pediatric oncology.
Collapse
Affiliation(s)
- John T Wiernikowski
- Division of Paediatric Haematology/Oncology, Department of Paediatrics, McMaster Children's Hospital, McMaster University, Hamilton, Ontario, Canada
| | - Melanie Brooke Bernhardt
- Section of Hematology/Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
31
|
Murabito A, Hirsch E, Ghigo A. Mechanisms of Anthracycline-Induced Cardiotoxicity: Is Mitochondrial Dysfunction the Answer? Front Cardiovasc Med 2020; 7:35. [PMID: 32226791 PMCID: PMC7080657 DOI: 10.3389/fcvm.2020.00035] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/24/2020] [Indexed: 12/19/2022] Open
Abstract
Cardiac side effects are a major drawback of anticancer therapies, often requiring the use of low and less effective doses or even discontinuation of the drug. Among all the drugs known to cause severe cardiotoxicity are anthracyclines that, though being the oldest chemotherapeutic drugs, are still a mainstay in the treatment of solid and hematological tumors. The recent expansion of the field of Cardio-Oncology, a branch of cardiology dealing with prevention or treatment of heart complications due to cancer treatment, has greatly improved our knowledge of the molecular mechanisms behind anthracycline-induced cardiotoxicity (AIC). Despite excessive generation of reactive oxygen species was originally believed to be the main cause of AIC, recent evidence points to the involvement of a plethora of different mechanisms that, interestingly, mainly converge on deregulation of mitochondrial function. In this review, we will describe how anthracyclines affect cardiac mitochondria and how these organelles contribute to AIC. Furthermore, we will discuss how drugs specifically targeting mitochondrial dysfunction and/or mitochondria-targeted drugs could be therapeutically exploited to treat AIC.
Collapse
Affiliation(s)
- Alessandra Murabito
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| |
Collapse
|