1
|
Tong J, Senechal I, Ramalingam S, Lyon AR. Risk Assessment Prior to Cardiotoxic Anticancer Therapies in 7 Steps. Br J Hosp Med (Lond) 2025; 86:1-21. [PMID: 39862029 DOI: 10.12968/hmed.2024.0632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2025]
Abstract
The burdens of cardiovascular (CV) diseases and cardiotoxic side effects of cancer treatment in oncology patients are increasing in parallel. The European Society of Cardiology (ESC) 2022 Cardio-Oncology guidelines recommend the use of standardized risk stratification tools to determine the risk of cardiotoxicity associated with different anticancer treatment modalities and the severity of their complications. The use of the Heart Failure Association-International Cardio-Oncology Society (HFA-ICOS) is essential for assessing risk prior to starting cancer treatment, and validation of these methods has been performed in patients receiving anthracyclines, human epidermal receptor 2 (HER2)-targeted therapies and breakpoint cluster region-abelson oncogene locus (BCR-ABL) inhibitors. The benefits of performing baseline CV risk assessment and stratification include early recognition of cardiotoxicities, personalisation of cancer treatment and monitoring strategies, and allocation of cardioprotection to those at the highest risk. This review summarizes the key points of risk stratification in these patients. The steps include identifying the target population, assessing nonmodifiable and modifiable CV risk factors, reviewing previous oncologic therapies and CV histories, and performing baseline investigations. In summary, this review aims to provide general physicians with a simple 7-step guide that will help steer and navigate them through cardiac risk evaluation of potentially cardiotoxic oncologic treatment strategies.
Collapse
Affiliation(s)
- Jieli Tong
- Cardio-Oncology Centre of Excellence, Royal Brompton Hospital, London, UK
- Department of Cardiology, Tan Tock Seng Hospital, Singapore, Singapore
| | - Isabelle Senechal
- Cardio-Oncology Centre of Excellence, Royal Brompton Hospital, London, UK
| | | | - Alexander R Lyon
- Cardio-Oncology Centre of Excellence, Royal Brompton Hospital, London, UK
| |
Collapse
|
2
|
Bernasconi R, Kuster GM. Non-coding RNAs and their potential exploitation in cancer therapy-related cardiotoxicity. Br J Pharmacol 2025; 182:296-315. [PMID: 38802331 DOI: 10.1111/bph.16416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 02/28/2024] [Accepted: 03/26/2024] [Indexed: 05/29/2024] Open
Abstract
Life expectancy in cancer patients has been extended in recent years, thanks to major breakthroughs in therapeutic developments. However, this also unmasked an increased incidence of cardiovascular diseases in cancer survivors, which is in part attributable to cancer therapy-related cardiovascular toxicity. Non-coding RNAs (ncRNAs) have received much appreciation due to their impact on gene expression. NcRNAs, which include microRNAs, long ncRNAs and circular RNAs, are non-protein-coding transcripts that are involved in the regulation of various biological processes, hence shaping cell identity and behaviour. They have also been implicated in disease development, including cardiovascular diseases, cancer and, more recently, cancer therapy-associated cardiotoxicity. This review outlines key features of cancer therapy-associated cardiotoxicity, what is known about the roles of ncRNAs in these processes and how ncRNAs could be exploited as therapeutic targets for cardioprotection. LINKED ARTICLES: This article is part of a themed issue Non-coding RNA Therapeutics. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v182.2/issuetoc.
Collapse
Affiliation(s)
- Riccardo Bernasconi
- Myocardial Research, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Gabriela M Kuster
- Myocardial Research, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
- Department of Cardiology, University Heart Center Basel, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
3
|
Park JY, Lengacher CA, Rodriguez CS, Meng H, Kip KE, Morgan S, Joshi A, Hueluer G, Wang JR, Tinsley S, Cox C, Kiluk J, Donovan KA, Moscoso M, Bornstein E, Lucas JM, Fonseca T, Krothapalli M, Padgett LS, Nidamanur S, Hornback E, Patel D, Chamkeri R, Reich RR. The Moderating Role of Genetics on the Effectiveness of the Mindfulness-Based Stress Reduction for Breast Cancer (MBSR(BC)) Program on Cognitive Impairment. Biol Res Nurs 2024:10998004241289629. [PMID: 39413359 DOI: 10.1177/10998004241289629] [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: 10/18/2024]
Abstract
BACKGROUND Genetics may influence symptoms experienced by breast cancer survivors (BCS) by moderating the effects of stress-reducing interventions, including the Mindfulness-Based Stress Reduction (MBSR(BC)) program, to reduce symptom severity. As part of a larger clinical trial, the aim of this study was to evaluate genetic variants as moderators of MBSR(BC) on improvements among BCS in cognitive functioning and symptoms. METHODS BCS (n = 128) were randomized to MBSR(BC) or the Breast Cancer Education Support Program. Objective neuropsychological and subjective measures of cognitive performance, and psychological and physical symptoms were collected at baseline, 6, 12, and 26 weeks. Linear mixed models were implemented to identify MBSR(BC)'s effects over time. A total of 22 single nucleotide polymorphisms (SNPs) from 20 genes known to be related to these symptoms were investigated using genomic DNA. These SNPs were tested as moderators of MBSR(BC) program effects. RESULTS Results showed MBSR(BC) participants experienced significantly greater benefits in cognitive functioning, however, the level of benefit varied based on one's genetic profile. Effects sizes, consistency across similar measures were investigated. Among 22 candidate SNPs, rs4680 in COMT, rs1800497 in ANKK1, and rs6277 in DRD2 demonstrated the strongest, most consistent positive effects in moderating MBSR(BC)'s impact on cognitive outcomes. CONCLUSIONS Although the effects were small, this translational research may potentially identify BCS with genotypes that would be most influenced by the MBSR(BC) program. These results may be used to develop personalized intervention programs tailored to the genetic profile of each breast cancer survivor who received chemotherapy or chemotherapy and radiation. TRIAL REGISTRATION ClinicalTrials.gov, https://www.ClinicalTrials.gov Registration Number: NCT02786797.
Collapse
Affiliation(s)
| | | | | | - Hongdao Meng
- College of Behavioral and Community Sciences, University of South Florida, Tampa, FL, USA
| | - Kevin E Kip
- UPMC Health Services Division, Pittsburgh, PA, USA
| | - Sandra Morgan
- University of South Florida College of Nursing, Tampa, FL, USA
| | | | | | - Julia R Wang
- Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | | | - Charles Cox
- Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | | | | | | | | | - Jean M Lucas
- Sarasota Memorial Health Care System, Sarasota, FL, USA
| | | | | | - Lynne S Padgett
- Veteran Affairs Office of Research & Development, Washington, DC, USA
| | | | | | - Diya Patel
- College of Arts and Sciences, University of South Florida, Tampa, FL, USA
| | - Ramya Chamkeri
- College of Arts and Sciences, University of South Florida, Tampa, FL, USA
| | | |
Collapse
|
4
|
Nyangwara VA, Mazhindu T, Chikwambi Z, Masimirembwa C, Campbell TB, Borok M, Ndlovu N. Cardiotoxicity and pharmacogenetics of doxorubicin in black Zimbabwean breast cancer patients. Br J Clin Pharmacol 2024; 90:1782-1789. [PMID: 36630266 DOI: 10.1111/bcp.15659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 12/15/2022] [Accepted: 12/28/2022] [Indexed: 01/12/2023] Open
Abstract
AIMS Doxorubicin-induced cardiotoxicity (DIC) is a significant cause of mortality in cancer care. This study was conducted to establish the frequency of DIC in Zimbabwean breast cancer patients on doxorubicin and to test the DIC predictive power of genetic biomarkers. METHODS A cohort of 50 Zimbabwean breast cancer patients treated with doxorubicin were followed up for 12 months with serial echocardiography and genotyped for UGTA1A6*4, SLC28A3 and RARG. Eleven per cent of the patients experienced DIC. RESULTS The frequencies of SLC28A3 (rs7853758), UGT1A6*4 (rs17863783) and RARG (rs2229774) were 60.7%, 17.9% and 14.3%, respectively. No association between DIC and the three variants was observed. CONCLUSIONS This is the first study on the prevalence of DIC and associated genetic biomarker predictive evaluation in Zimbabwean breast cancer patients. The genetic frequencies observed in our study were different to those reported in other populations. A larger sample size with a longer follow-up time will be necessary in future studies.
Collapse
Affiliation(s)
- Vincent Aketch Nyangwara
- African Institute of Biomedical Science and Technology, Harare, Zimbabwe
- Department of Biotechnology, Chinhoyi University of Technology, Chinhoyi, Zimbabwe
| | - Tinashe Mazhindu
- African Institute of Biomedical Science and Technology, Harare, Zimbabwe
- Unit of Internal Medicine, Faculty of Medicine and Health Sciences, University of Zimbabwe, Harare, Zimbabwe
| | - Zedias Chikwambi
- African Institute of Biomedical Science and Technology, Harare, Zimbabwe
- Department of Biotechnology, Chinhoyi University of Technology, Chinhoyi, Zimbabwe
| | | | - Thomas B Campbell
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Margaret Borok
- Unit of Internal Medicine, Faculty of Medicine and Health Sciences, University of Zimbabwe, Harare, Zimbabwe
| | - Ntokozo Ndlovu
- Department of Oncology, Faculty of Medicine and Health Sciences, University of Zimbabwe, Harare, Zimbabwe
| |
Collapse
|
5
|
Ragab SM, El-Hawy MA, El-Hefnawy SM, El-Deeb HMA, Elfalah AS, Mahmoud AA. CELF4 (rs1786814) gene polymorphism and speckle-tracking Echocardiography for cardiovascular complications in childhood cancer survivors. Pediatr Res 2024:10.1038/s41390-024-03400-3. [PMID: 39048669 DOI: 10.1038/s41390-024-03400-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 04/30/2024] [Accepted: 06/13/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND Despite a well-known dose-dependent association between the risk of cardiac dysfunction and anthracycline, the risk of cardiac dysfunction for any given anthracycline dose varies between patients. So, we assessed CELF4 (rs1786814) gene polymorphism on anthracycline-related cardiotoxicity in childhood cancer survivors (CCS). METHODS This comparative cross-sectional study included 53 CCS who had regular follow-up visits at the Pediatric Oncology Unit, Menoufia University Hospital. CELF4 (rs1786814) gene polymorphism and conventional and speckle-tracking Echocardiography were done for all survivors. RESULTS Regarding CELF4 (rs1786814) genotypes, significant differences existed between the studied groups with a predominance of GG homozygous mutation. For Echocardiographic findings, the ejection fraction and end-systolic diameter compared to the control group, were significantly lower in the survivors group. Speckle- tracking Echocardiography showed a significant difference regarding (GLPS-A4C) and (GLPS-LAX), with no significant difference regarding (GLPS-A2C), (GLPS-Avg) and left atrium between the studied groups. Multivariate logistic regression analysis illustrated a statistically significant relation between cumulative anthracycline dose >300 mg/m2 and CLEF4 (rs1786814) genotypes (GG and GA) and the risk of cardiotoxicity with more significance in GG mutation. CONCLUSION Early detection of ventricular dysfunction in CCS with subclinical cardiotoxicity with regular follow-up is promising before the development of life-threatening complications. IMPACT Early detection of anthracycline-related cardiotoxicity in childhood cancer survivors (CCS) after finishing chemotherapy. CLEF4 (rs1786814) GG variant is more significant in CCS exposed to high-dose anthracycline. GLPS holds promise as an early predictor of late left ventricular dysfunction and subclinical cardiotoxicity in CCS.
Collapse
Affiliation(s)
- Seham M Ragab
- Departement of Pediatrics, Faculty of Medicine, Menoufia University, Shebin ElKom, Egypt
| | - Mahmoud A El-Hawy
- Departement of Pediatrics, Faculty of Medicine, Menoufia University, Shebin ElKom, Egypt
| | - Sally M El-Hefnawy
- Medical Biochemistry and Molecular Biology Departement, Faculty of Medicine, Menoufia University, Shebin Elkom, Egypt
- Medical Biochemistry Department, Faculty of Dentistry, AlRyada University for Science and Technology (RST), Al-Sadat City, Egypt
| | - Hend M A El-Deeb
- Department of Cardiology, Faculty of Medicine, Menoufia University, Shebin Elkom, Egypt
| | - Amany S Elfalah
- Departement of Pediatrics, Faculty of Medicine, Menoufia University, Shebin ElKom, Egypt
| | - Asmaa A Mahmoud
- Departement of Pediatrics, Faculty of Medicine, Menoufia University, Shebin ElKom, Egypt.
| |
Collapse
|
6
|
Kuang Z, Kong M, Yan N, Ma X, Wu M, Li J. Precision Cardio-oncology: Update on Omics-Based Diagnostic Methods. Curr Treat Options Oncol 2024; 25:679-701. [PMID: 38676836 PMCID: PMC11082000 DOI: 10.1007/s11864-024-01203-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/02/2024] [Indexed: 04/29/2024]
Abstract
OPINION STATEMENT Cardio-oncology is an emerging interdisciplinary field dedicated to the early detection and treatment of adverse cardiovascular events associated with anticancer treatment, and current clinical management of anticancer-treatment-related cardiovascular toxicity (CTR-CVT) remains limited by a lack of detailed phenotypic data. However, the promise of diagnosing CTR-CVT using deep phenotyping has emerged with the development of precision medicine, particularly the use of omics-based methodologies to discover sensitive biomarkers of the disease. In the future, combining information produced by a variety of omics methodologies could expand the clinical practice of cardio-oncology. In this review, we demonstrate how omics approaches can improve our comprehension of CTR-CVT deep phenotyping, discuss the positive and negative aspects of available omics approaches for CTR-CVT diagnosis, and outline how to integrate multiple sets of omics data into individualized monitoring and treatment. This will offer a reliable technical route for lowering cardiovascular morbidity and mortality in cancer patients and survivors.
Collapse
Affiliation(s)
- Ziyu Kuang
- Oncology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Miao Kong
- Oncology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ningzhe Yan
- Oncology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Xinyi Ma
- Oncology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Min Wu
- Cardiovascular Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Jie Li
- Oncology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
7
|
Staropoli N, Scionti F, Farenza V, Falcone F, Luciano F, Renne M, Di Martino MT, Ciliberto D, Tedesco L, Crispino A, Labanca C, Cucè M, Esposito S, Agapito G, Cannataro M, Tassone P, Tagliaferri P, Arbitrio M. Identification of ADME genes polymorphic variants linked to trastuzumab-induced cardiotoxicity in breast cancer patients: Case series of mono-institutional experience. Biomed Pharmacother 2024; 174:116478. [PMID: 38547766 DOI: 10.1016/j.biopha.2024.116478] [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: 02/03/2024] [Revised: 03/18/2024] [Accepted: 03/19/2024] [Indexed: 05/01/2024] Open
Abstract
BACKGROUND Long-term survival induced by anticancer treatments discloses emerging frailty among breast cancer (BC) survivors. Trastuzumab-induced cardiotoxicity (TIC) is reported in at least 5% of HER2+BC patients. However, TIC mechanism remains unclear and predictive genetic biomarkers are still lacking. Interaction between systemic inflammation, cytokine release and ADME genes in cancer patients might contribute to explain mechanisms underlying individual susceptibility to TIC and drug response variability. We present a single institution case series to investigate the potential role of genetic variants in ADME genes in HER2+BC patients TIC experienced. METHODS We selected data related to 40 HER2+ BC patients undergone to DMET genotyping of ADME constitutive variant profiling, with the aim to prospectively explore their potential role in developing TIC. Only 3 patients ("case series"), who experienced TIC, were compared to 37 "control group" matched patients cardiotoxicity-sparing. All patients underwent to left ventricular ejection fraction (LVEF) evaluation at diagnosis and during anti-HER2 therapy. Each single probe was clustered to detect SNPs related to cardiotoxicity. RESULTS In this retrospective analysis, our 3 cases were homogeneous in terms of clinical-pathological characteristics, trastuzumab-based treatment and LVEF decline. We identified 9 polymorphic variants in 8 ADME genes (UGT1A1, UGT1A6, UGT1A7, UGT2B15, SLC22A1, CYP3A5, ABCC4, CYP2D6) potentially associated with TIC. CONCLUSION Real-world TIC incidence is higher compared to randomized clinical trials and biomarkers with potential predictive value aren't available. Our preliminary data, as proof of concept, could suggest a predictive role of pharmacogenomic approach in the identification of cardiotoxicity risk biomarkers for anti-HER2 treatment.
Collapse
Affiliation(s)
- Nicoletta Staropoli
- Medical Oncology Unit, R. Dulbecco (Mater Domini facility), Teaching Hospital, Magna Græcia University and Cancer Center, Campus Salvatore Venuta, Catanzaro, Italy; Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy.
| | - Francesca Scionti
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Valentina Farenza
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Federica Falcone
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Francesco Luciano
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Maria Renne
- Surgery Unit, Magna Græcia University and Cancer Center, Campus Salvatore Venuta, Catanzaro, Italy
| | - Maria Teresa Di Martino
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Domenico Ciliberto
- Medical Oncology Unit, R. Dulbecco (Mater Domini facility), Teaching Hospital, Magna Græcia University and Cancer Center, Campus Salvatore Venuta, Catanzaro, Italy
| | - Ludovica Tedesco
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Antonella Crispino
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Caterina Labanca
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Maria Cucè
- Medical Oncology Unit, R. Dulbecco (Mater Domini facility), Teaching Hospital, Magna Græcia University and Cancer Center, Campus Salvatore Venuta, Catanzaro, Italy
| | - Stefania Esposito
- Pharmacy Unit, R. Dulbecco (Mater Domini facility), Teaching Hospital, Campus Salvatore Venuta, Catanzaro, Italy
| | - Giuseppe Agapito
- Department of Law, Economics and Sociology, Magna Graecia University of Catanzaro, Catanzaro 88100, Italy; Data Analytics Research Center, Magna Graecia University of Catanzaro, Catanzaro 88100, Italy
| | - Mario Cannataro
- Department of Medical and Surgical Science, Magna Graecia University of Catanzaro, Catanzaro 88100, Italy
| | - Pierfrancesco Tassone
- Medical Oncology Unit, R. Dulbecco (Mater Domini facility), Teaching Hospital, Magna Græcia University and Cancer Center, Campus Salvatore Venuta, Catanzaro, Italy; Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Pierosandro Tagliaferri
- Medical Oncology Unit, R. Dulbecco (Mater Domini facility), Teaching Hospital, Magna Græcia University and Cancer Center, Campus Salvatore Venuta, Catanzaro, Italy; Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy.
| | - Mariamena Arbitrio
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), Catanzaro 88100, Italy.
| |
Collapse
|
8
|
Ni MM, Yang JF, Miao J, Xu J. Association between genetic variants of transmembrane transporters and susceptibility to anthracycline-induced cardiotoxicity: Current understanding and existing evidence. Clin Genet 2024; 105:115-129. [PMID: 37961936 DOI: 10.1111/cge.14452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/31/2023] [Accepted: 11/02/2023] [Indexed: 11/15/2023]
Abstract
Anthracyclines remain the cornerstone of numerous chemotherapeutic protocols, with beneficial effects against haematological malignancies and solid tumours. Unfortunately, the clinical usefulness of anthracyclines is compromised by the development of cardiotoxic side effects, leading to dose limitations or treatment discontinuation. There is no absolute linear correlation between the incidence of cardiotoxicity and the threshold dose, suggesting that genetic factors may modify the association between anthracyclines and cardiotoxicity risk. And the majority of single nucleotide polymorphisms (SNPs) associated with anthracycline pharmacogenomics were identified in the ATP-binding cassette (ABC) and solute carrier (SLC) transporters, generating increasing interest in the pharmacogenetic implications of their genetic variations for anthracycline-induced cardiotoxicity (AIC). This review focuses on the influence of SLC and ABC polymorphisms on AIC and highlights the prospects and clinical significance of pharmacogenetics for individualised preventive approaches.
Collapse
Affiliation(s)
- Ming-Ming Ni
- Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Ju-Fei Yang
- Department of Pharmacy, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jing Miao
- Department of Pharmacy, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Research Center for Clinical Pharmacy, Zhejiang University, Hangzhou, China
| | - Jin Xu
- Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
9
|
Abstract
Anthracycline-induced cardiotoxicity (AIC) is a serious and common side effect of anthracycline therapy. Identification of genes and genetic variants associated with AIC risk has clinical potential as a cardiotoxicity predictive tool and to allow the development of personalized therapies. In this review, we provide an overview of the function of known AIC genes identified by association studies and categorize them based on their mechanistic implication in AIC. We also discuss the importance of functional validation of AIC-associated variants in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) to advance the implementation of genetic predictive biomarkers. Finally, we review how patient-specific hiPSC-CMs can be used to identify novel patient-relevant functional targets and for the discovery of cardioprotectant drugs to prevent AIC. Implementation of functional validation and use of hiPSC-CMs for drug discovery will identify the next generation of highly effective and personalized cardioprotectants and accelerate the inclusion of approved AIC biomarkers into clinical practice.
Collapse
Affiliation(s)
- Romina B Cejas
- Department of Pharmacology and Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA;
| | - Kateryna Petrykey
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Yadav Sapkota
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Paul W Burridge
- Department of Pharmacology and Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA;
| |
Collapse
|
10
|
Singh P, Zhou L, Shah DA, Cejas RB, Crossman DK, Jouni M, Magdy T, Wang X, Sharafeldin N, Hageman L, McKenna DE, Horvath S, Armenian SH, Balis FM, Hawkins DS, Keller FG, Hudson MM, Neglia JP, Ritchey AK, Ginsberg JP, Landier W, Burridge PW, Bhatia S. Identification of novel hypermethylated or hypomethylated CpG sites and genes associated with anthracycline-induced cardiomyopathy. Sci Rep 2023; 13:12683. [PMID: 37542143 PMCID: PMC10403495 DOI: 10.1038/s41598-023-39357-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 07/24/2023] [Indexed: 08/06/2023] Open
Abstract
Anthracycline-induced cardiomyopathy is a leading cause of late morbidity in childhood cancer survivors. Aberrant DNA methylation plays a role in de novo cardiovascular disease. Epigenetic processes could play a role in anthracycline-induced cardiomyopathy but remain unstudied. We sought to examine if genome-wide differential methylation at 'CpG' sites in peripheral blood DNA is associated with anthracycline-induced cardiomyopathy. This report used participants from a matched case-control study; 52 non-Hispanic White, anthracycline-exposed childhood cancer survivors with cardiomyopathy were matched 1:1 with 52 survivors with no cardiomyopathy. Paired ChAMP (Chip Analysis Methylation Pipeline) with integrated reference-based deconvolution of adult peripheral blood DNA methylation was used to analyze data from Illumina HumanMethylation EPIC BeadChip arrays. An epigenome-wide association study (EWAS) was performed, and the model was adjusted for GrimAge, sex, interaction terms of age at enrollment, chest radiation, age at diagnosis squared, and cardiovascular risk factors (CVRFs: diabetes, hypertension, dyslipidemia). Prioritized genes were functionally validated by gene knockout in human induced pluripotent stem cell cardiomyocytes (hiPSC-CMs) using CRISPR/Cas9 technology. DNA-methylation EPIC array analyses identified 32 differentially methylated probes (DMP: 15 hyper-methylated and 17 hypo-methylated probes) that overlap with 23 genes and 9 intergenic regions. Three hundred and fifty-four differential methylated regions (DMRs) were also identified. Several of these genes are associated with cardiac dysfunction. Knockout of genes EXO6CB, FCHSD2, NIPAL2, and SYNPO2 in hiPSC-CMs increased sensitivity to doxorubicin. In addition, EWAS analysis identified hypo-methylation of probe 'cg15939386' in gene RORA to be significantly associated with anthracycline-induced cardiomyopathy. In this genome-wide DNA methylation profile study, we observed significant differences in DNA methylation at the CpG level between anthracycline-exposed childhood cancer survivors with and without cardiomyopathy, implicating differential DNA methylation of certain genes could play a role in pathogenesis of anthracycline-induced cardiomyopathy.
Collapse
Affiliation(s)
- Purnima Singh
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Liting Zhou
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Disheet A Shah
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
| | - Romina B Cejas
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
| | - David K Crossman
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mariam Jouni
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
| | - Tarek Magdy
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
- Department of Pathology and Translational Pathobiology and Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA
| | - Xuexia Wang
- Department of Biostatistics, Florida International University, Miami, FL, USA
| | - Noha Sharafeldin
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lindsey Hageman
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Donald E McKenna
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
| | - Steve Horvath
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Saro H Armenian
- Department of Population Sciences, City of Hope, Duarte, CA, USA
| | - Frank M Balis
- Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Frank G Keller
- Children's Healthcare of Atlanta, Emory University, Atlanta, GA, USA
| | | | | | - A Kim Ritchey
- Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, USA
| | | | - Wendy Landier
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Paul W Burridge
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
| | - Smita Bhatia
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL, USA.
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
11
|
Antoniadi K, Thomaidis N, Nihoyannopoulos P, Toutouzas K, Gikas E, Kelaidi C, Polychronopoulou S. Prognostic Factors for Cardiotoxicity among Children with Cancer: Definition, Causes, and Diagnosis with Omics Technologies. Diagnostics (Basel) 2023; 13:1864. [PMID: 37296716 PMCID: PMC10252297 DOI: 10.3390/diagnostics13111864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/03/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Improvements in the treatment of childhood cancer have considerably enhanced survival rates over the last decades to over 80% as of today. However, this great achievement has been accompanied by the occurrence of several early and long-term treatment-related complications major of which is cardiotoxicity. This article reviews the contemporary definition of cardiotoxicity, older and newer chemotherapeutic agents that are mainly involved in cardiotoxicity, routine process diagnoses, and methods using omics technology for early and preventive diagnosis. Chemotherapeutic agents and radiation therapies have been implicated as a cause of cardiotoxicity. In response, the area of cardio-oncology has developed into a crucial element of oncologic patient care, committed to the early diagnosis and treatment of adverse cardiac events. However, routine diagnosis and the monitoring of cardiotoxicity rely on electrocardiography and echocardiography. For the early detection of cardiotoxicity, in recent years, major studies have been conducted using biomarkers such as troponin, N-terminal pro b-natriuretic peptide, etc. Despite the refinements in diagnostics, severe limitations still exist due to the increase in the above-mentioned biomarkers only after significant cardiac damage has occurred. Lately, the research has expanded by introducing new technologies and finding new markers using the omics approach. These new markers could be used not only for early detection but also for the early prevention of cardiotoxicity. Omics science, which includes genomics, transcriptomics, proteomics, and metabolomics, offers new opportunities for biomarker discovery in cardiotoxicity and may provide an understanding of the mechanisms of cardiotoxicity beyond traditional technologies.
Collapse
Affiliation(s)
- Kondylia Antoniadi
- Department of Pediatric Hematology-Oncology (T.A.O.), “Aghia Sophia” Children’s Hospital, Goudi, 11527 Athens, Greece
| | - Nikolaos Thomaidis
- Department of Chemistry, National and Kapodistrian University of Athens, 15772 Athens, Greece
| | - Petros Nihoyannopoulos
- First Department of Cardiology, University of Athens, Hippokration Hospital, 11527 Athens, Greece
| | - Konstantinos Toutouzas
- First Department of Cardiology, University of Athens, Hippokration Hospital, 11527 Athens, Greece
| | - Evangelos Gikas
- Department of Chemistry, National and Kapodistrian University of Athens, 15772 Athens, Greece
| | - Charikleia Kelaidi
- Department of Pediatric Hematology-Oncology (T.A.O.), “Aghia Sophia” Children’s Hospital, Goudi, 11527 Athens, Greece
| | - Sophia Polychronopoulou
- Department of Pediatric Hematology-Oncology (T.A.O.), “Aghia Sophia” Children’s Hospital, Goudi, 11527 Athens, Greece
| |
Collapse
|
12
|
Georgiopoulos G, Makris N, Laina A, Theodorakakou F, Briasoulis A, Trougakos IP, Dimopoulos MA, Kastritis E, Stamatelopoulos K. Cardiovascular Toxicity of Proteasome Inhibitors: Underlying Mechanisms and Management Strategies: JACC: CardioOncology State-of-the-Art Review. JACC CardioOncol 2023; 5:1-21. [PMID: 36875897 PMCID: PMC9982226 DOI: 10.1016/j.jaccao.2022.12.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 12/14/2022] [Accepted: 12/19/2022] [Indexed: 02/24/2023] Open
Abstract
Proteasome inhibitors (PIs) are the backbone of combination treatments for patients with multiple myeloma and AL amyloidosis, while also indicated in Waldenström's macroglobulinemia and other malignancies. PIs act on proteasome peptidases, causing proteome instability due to accumulating aggregated, unfolded, and/or damaged polypeptides; sustained proteome instability then induces cell cycle arrest and/or apoptosis. Carfilzomib, an intravenous irreversible PI, exhibits a more severe cardiovascular toxicity profile as compared with the orally administered ixazomib or intravenous reversible PI such as bortezomib. Cardiovascular toxicity includes heart failure, hypertension, arrhythmias, and acute coronary syndromes. Because PIs are critical components of the treatment of hematological malignancies and amyloidosis, managing their cardiovascular toxicity involves identifying patients at risk, diagnosing toxicity early at the preclinical level, and offering cardioprotection if needed. Future research is required to elucidate underlying mechanisms, improve risk stratification, define the optimal management strategy, and develop new PIs with safe cardiovascular profiles.
Collapse
Key Words
- ACE, angiotensin-converting enzyme
- ACS, acute coronary syndrome
- AE, adverse event
- AF, atrial fibrillation
- ARB, angiotensin receptor blocker
- ASCT, autologous stem cell transplantation
- BP, blood pressure
- CVAE, cardiovascular adverse event
- ESC, European Society of Cardiology
- FMD, flow-mediated dilatation
- GLS, global longitudinal strain
- HF, heart failure
- HFpEF, heart failure with preserved ejection fraction
- IHD, ischemic heart disease
- IMiD, immunomodulatory drug
- Kd, carfilzomib and dexamethasone
- LA, left atrial
- LV, left ventricular
- LVEF, left ventricular ejection fraction
- MM, multiple myeloma
- NO, nitric oxide
- NP, natriuretic peptide
- OS, overall survival
- PBMC, peripheral blood mononuclear cell
- PFS, progression-free survival
- PH, pulmonary hypertension
- PI, proteasome inhibitor
- PWV, pulse wave velocity
- PrA, proteasome activity
- RRMM, relapse or refractory multiple myeloma
- SBP, systolic blood pressure
- TMA, thrombotic microangiopathy
- UPP, ubiquitin proteasome pathway
- VTE, venous thromboembolism
- Vd, bortezomib and dexamethasone
- WM, Waldenström’s macroglobulinemia
- bortezomib
- cardiovascular toxicity
- carfilzomib
- eNOS, endothelial nitric oxide synthase
- ixazomib
- proteasome inhibition
Collapse
Affiliation(s)
- Georgios Georgiopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London, United Kingdom
| | - Nikolaos Makris
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Ageliki Laina
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Foteini Theodorakakou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Alexandros Briasoulis
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis P. Trougakos
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Greece
| | | | - Efstathios Kastritis
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Kimon Stamatelopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
13
|
RING Finger Protein 10 Regulates AP-1/Meox2 to Mediate Pirarubicin-Induced Cardiomyocyte Apoptosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:7872193. [PMID: 36713029 PMCID: PMC9883094 DOI: 10.1155/2023/7872193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/12/2022] [Accepted: 11/25/2022] [Indexed: 01/21/2023]
Abstract
Pirarubicin (THP) is one of the classic chemotherapy drugs for cancer treatment. It is often clinically limited because of its cardiotoxicity. The occurrence and development of THP-mediated chemotherapy-related cardiotoxicity (CRC) may be reversed by RING finger protein 10 (RNF10). This study was performed with the aim of evaluating the inhibitory effect of RNF10 on THP-mediated CRC and its molecular mechanism. In vivo, we found that the expression of RNF10 decreased in THP-induced CRC rats, accompanied by Meox2 inhibition and AP-1 activation, resulting in increased cardiomyocyte apoptosis. After small interfering RNA (siRNA) and lentivirus transfection (Lv) of RNF10 in vitro, the expression of RNF10, Meox2, and AP-1 proteins and the degree of cardiomyocyte apoptosis were detected. We found that overexpression of RNF10 in H9C2 cardiomyocytes significantly promoted Meox2 and inhibited AP-1, alleviated apoptosis, and showed further inhibitory activity on THP-induced cardiomyocyte toxicity. Silencing RNF10 showed the opposite result. Our study showed that RNF10 inhibited THP-induced CRC through the activity of Meox2 and AP-1 proteins. RNF10 may be the next drug target for the treatment of CRC and other related cardiovascular diseases.
Collapse
|
14
|
Sadler D, Okwuosa T, Teske AJ, Guha A, Collier P, Moudgil R, Sarkar A, Brown SA. Cardio oncology: Digital innovations, precision medicine and health equity. Front Cardiovasc Med 2022; 9:951551. [PMID: 36407451 PMCID: PMC9669068 DOI: 10.3389/fcvm.2022.951551] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 10/03/2022] [Indexed: 11/06/2022] Open
Abstract
The rapid emergence of cardio-oncology has resulted in a rapid growth of cardio-oncology programs, dedicated professional societies sections and committees, and multiple collaborative networks that emerged to amplify the access to care in this new subspecialty. However, most existing data, position statements and guidelines are limited by the lack of availability of large clinical trials to support these recommendations. Furthermore, there are significant challenges regarding proper access to cardio-oncology care and treatment, particularly in marginalized and minority populations. The emergence and evolution of personalized medicine, artificial intelligence (AI), and machine learning in medicine and in cardio-oncology provides an opportunity for a more targeted, personalized approach to cardiovascular complications of cancer treatment. The proper implementation of these new modalities may facilitate a more equitable approach to adequate and universal access to cardio-oncology care, improve health related outcomes, and enable health care systems to eliminate the digital divide. This article reviews and analyzes the current status on these important issues.
Collapse
Affiliation(s)
- Diego Sadler
- Cardio Oncology Section, Department of Cardiovascular Medicine, Heart Vascular and Thoracic Institute, Cleveland Clinic Florida, Weston, FL, United States
- *Correspondence: Diego Sadler
| | - Tochukwu Okwuosa
- Division of Cardiology, Department of Medicine, Rush University Medical Center, Chicago, IL, United States
| | - A. J. Teske
- Division of Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Avirup Guha
- Division of Cardiology, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Patrick Collier
- Cleveland Clinic, Cardio Oncology, Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland, OH, United States
| | - Rohit Moudgil
- Cleveland Clinic, Cardio Oncology, Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland, OH, United States
| | - Abdullah Sarkar
- Cardio Oncology Section, Department of Cardiovascular Medicine, Heart Vascular and Thoracic Institute, Cleveland Clinic Florida, Weston, FL, United States
| | - Sherry-Ann Brown
- Division of Cardiology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
15
|
Li MY, Peng LM, Chen XP. Pharmacogenomics in drug-induced cardiotoxicity: Current status and the future. Front Cardiovasc Med 2022; 9:966261. [PMID: 36312261 PMCID: PMC9606405 DOI: 10.3389/fcvm.2022.966261] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 09/05/2022] [Indexed: 11/15/2022] Open
Abstract
Drug-induced cardiotoxicity (DICT) is an important concern of drug safety in both drug development and clinical application. The clinical manifestations of DICT include cardiomyopathy, arrhythmia, myocardial ischemia, heart failure, and a series of cardiac structural and functional changes. The occurrence of DICT has negative impacts on the life quality of the patients, brings additional social and economic burden. It is important to identify the potential factors and explore the mechanisms of DICT. Traditional cardiovascular risk factors can only partially explain the risk of DICT. Pharmacogenomic studies show accumulated evidence of genetics in DICT and suggest the potential to guide precision therapy to reduce risk of cardiotoxicity. The comprehensive application of technologies such as third-generation sequencing, human induced pluripotent stem (iPS) cells and genome editing has promoted the in-depth understanding of the functional role of susceptible genes in DICT. This paper reviewed drugs that cause DICT, the clinical manifestations and laboratory tests, as well as the related content of genetic variations associated with the risk of DICT, and further discussed the implication of new technologies in pharmacogenomics of DICT.
Collapse
Affiliation(s)
- Mo-Yun Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China,Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Li-Ming Peng
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China,Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China,Department of Cardiology, Xiangya Hospital, Central South University, Changsha, China,*Correspondence: Li-Ming Peng
| | - Xiao-Ping Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China,Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China,Xiao-Ping Chen
| |
Collapse
|
16
|
Park JY, Lengacher CA, Reich RR, Park HY, Whiting J, Nguyen AT, Rodríguez C, Meng H, Tinsley S, Chauca K, Gordillo-Casero L, Wittenberg T, Joshi A, Lin K, Ismail-Khan R, Kiluk JV, Kip KE. Translational Genomic Research: The Association between Genetic Profiles and Cognitive Functioning or Cardiac Function Among Breast Cancer Survivors Completing Chemotherapy. Biol Res Nurs 2022; 24:433-447. [PMID: 35499926 PMCID: PMC9630728 DOI: 10.1177/10998004221094386] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Introduction: Emerging evidence suggests that Chemotherapy (CT) treated breast cancer survivors (BCS) who have "risk variants" in genes may be more susceptible to cognitive impairment (CI) and/or poor cardiac phenotypes. The objective of this preliminary study was to examine whether there is a relationship between genetic variants and objective/subjective cognitive or cardiac phenotypes. Methods and Analysis: BCS were recruited from Moffitt Cancer Center, Morsani College of Medicine, AdventHealth Tampa and Sarasota Memorial Hospital. Genomic DNA were collected at baseline for genotyping analysis. A total of 16 single nucleotide polymorphisms (SNPs) from 14 genes involved in cognitive or cardiac function were evaluated. Three genetic models (additive, dominant, and recessive) were used to test correlation coefficients between genetic variants and objective/subjective measures of cognitive functioning and cardiac outcomes (heart rate, diastolic blood pressure, systolic blood pressure, respiration rate, and oxygen saturation). Results: BCS (207 participants) with a mean age of 56 enrolled in this study. The majority were non-Hispanic white (73.7%), married (63.1%), and received both CT and radiation treatment (77.3%). Three SNPs in genes related to cognitive functioning (rs429358 in APOE, rs1800497 in ANKK1, rs10119 in TOMM40) emerged with the most consistent significant relationship with cognitive outcomes. Among five candidate SNPs related to cardiac functioning, rs8055236 in CDH13 and rs1801133 in MTHER emerged with potential significant relationships with cardiac phenotype. Conclusions: These preliminary results provide initial targets to further examine whether BCS with specific genetic profiles may preferentially benefit from interventions designed to improve cognitive and cardiac functioning following CT.
Collapse
Affiliation(s)
- Jong Y. Park
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, USA
| | | | - Richard R. Reich
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Hyun Y. Park
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Junmin Whiting
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Anh Thy Nguyen
- Department of Epidemiology and
Biostatistics, USF College of Public Health, University of South
Florida, Tampa, FL, USA
| | | | - Hongdao Meng
- School of Aging Studies, College of
Behavioral and Community Sciences, University of South
Floridaa, Tampa, FL, USA
| | - Sara Tinsley
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, USA
| | | | | | | | - Anisha Joshi
- University of South Florida College
of Nursing, Tampa, FL, USA
| | - Katherine Lin
- University of South Florida College
of Nursing, Tampa, FL, USA
| | - Roohi Ismail-Khan
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, USA
| | - John V. Kiluk
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Kevin E. Kip
- UPMC Health Services
Division, Pittsburgh, PA, USA
| |
Collapse
|
17
|
Liu B, Guan X, Wang Y, Sun X, Yi Z, Lv D, Wang W, Li L, Zhai J, Li H, Ma F. Clinical and genetic predictions of early-onset cardiac toxicity in adjuvant chemotherapy for breast cancer. Future Oncol 2022; 18:2127-2139. [PMID: 35414207 DOI: 10.2217/fon-2021-1021] [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
Aim: To identify clinical and genetic variants associated with early-onset cardiac toxicity with a low cumulative dose of chemotherapy drugs in breast cancer. Methods: A total of 388 recruited patients completed routine blood, liver and kidney function, D-dimer, troponin T, brain natriuretic peptide (BNP) or N-terminal prohormone of BNP, ECG and echocardiography tests before and after adjuvant chemotherapy. 25 single-nucleotide polymorphisms (SNPs) were tested. Results: A total of 277 adjuvant chemotherapy-related cardiac toxicity events were recorded in 180 patients (46.4%). Anthracycline-containing chemotherapy (odds ratio: 1.848; 95% CI: 1.135-3.008; p = 0.014) and the SLC28A3 rs885004 GG genotype (odds ratio: 2.034; 95% CI: 1.189-3.479; p = 0.010) were found to be associated with overall cardiac toxicity. The final predictive risk model consisting of clinical risk factors and SNPs was better than SNP alone (p = 0.006) or clinical risk factor alone (p = 0.065). Conclusion: On the basis of clinical factors, a prediction model with genetic susceptibility factors can better predict early-onset cardiac toxicity.
Collapse
Affiliation(s)
- Binliang Liu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.,Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, Hunan, 410013, China
| | - Xiuwen Guan
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yanfeng Wang
- Department of Comprehensive Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xiaoying Sun
- Department of Medical Oncology, Cancer Hospital of Huanxing, Beijing, 100065, China
| | - Zongbi Yi
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.,Department of Radiation & Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
| | - Dan Lv
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Wenna Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lixi Li
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jingtong Zhai
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Hong Li
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Fei Ma
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| |
Collapse
|
18
|
Huang H, Christidi E, Shafaattalab S, Davis MK, Tibbits GF, Brunham LR. RARG S427L attenuates the DNA repair response to doxorubicin in induced pluripotent stem cell-derived cardiomyocytes. Stem Cell Reports 2022; 17:756-765. [PMID: 35364012 PMCID: PMC9023798 DOI: 10.1016/j.stemcr.2022.03.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 03/02/2022] [Accepted: 03/02/2022] [Indexed: 11/26/2022] Open
Abstract
Doxorubicin is a commonly used chemotherapeutic drug, but its use is limited by doxorubicin-induced cardiotoxicity (DIC), which can lead to irreversible heart failure and death. A missense variant rs2229774 (p.S427L) in the retinoic acid receptor gamma (RARG) gene is associated with increased susceptibility to DIC, but the precise mechanism underlying this association is incompletely understood. We performed molecular dynamic simulations to determine the effect of this variant on RARG structure and then validated these predictions using CRISPR-Cas9-genome-edited, induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). We found that this variant leads to reduced activation of its target genes in response to doxorubicin, including gene pathways involved in DNA repair and consequently an inability to mediate DNA repair after exposure to doxorubicin. Our findings establish a role of RARG p.S427L in attenuating DNA repair in DIC and provide insight into the pathogenesis of this cardiotoxic effect. RARG p.S427L is predicted to alter the stability of the C terminus of the protein The RARG p.S427L variant has impaired ability to activate its target genes This variant attenuates the DNA repair response to doxorubicin
Collapse
Affiliation(s)
- Haojun Huang
- Centre for Heart Lung Innovation, Department of Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Effimia Christidi
- Centre for Heart Lung Innovation, Department of Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Sanam Shafaattalab
- Molecular Cardiac Physiology Group, Departments of Biomedical Physiology and Kinesiology and Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Margot K Davis
- Division of Cardiology, Department of Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Glen F Tibbits
- Molecular Cardiac Physiology Group, Departments of Biomedical Physiology and Kinesiology and Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada; Cellular and Regenerative Medicine Centre, British Columbia Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - Liam R Brunham
- Centre for Heart Lung Innovation, Department of Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada; Department of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC V6H 3N1, Canada.
| |
Collapse
|
19
|
Zolk O, von dem Knesebeck A, Graf N, Simon T, Hero B, Abdul-Khaliq H, Abd El Rahman M, Spix C, Mayer B, Elsner S, Gebauer J, Langer T. Cardiovascular Health Status And Genetic Risk In Survivors of Childhood Neuroblastoma and Nephroblastoma Treated With Doxorubicin: Protocol of the Pharmacogenetic Part of the LESS-Anthra Cross-Sectional Cohort Study. JMIR Res Protoc 2022; 11:e27898. [PMID: 35175211 PMCID: PMC8895281 DOI: 10.2196/27898] [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: 02/15/2021] [Revised: 06/11/2021] [Accepted: 08/17/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND In childhood cancer survivors (survival of 5 years or more after diagnosis), cardiac toxicity is the most common nonmalignant cause of death attributed to treatment-related consequences. Identifying patients at risk of developing late cardiac toxicity is therefore crucial to improving treatment outcomes. The use of genetic markers has been proposed, together with clinical risk factors, to predict individual risk of cardiac toxicity from cancer therapies, such as doxorubicin. OBJECTIVE The primary aim of this study is to evaluate the value of multimarker genetic testing for RARG rs2229774, UGT1A6 rs17863783, and SLC28A3 rs7853758 for predicting doxorubicin-induced cardiotoxicity. The secondary aim is to replicate previously described associations of candidate genetic markers with doxorubicin-induced cardiotoxicity. Moreover, we will evaluate the prevalence of cardiovascular dysfunction in childhood cancer survivors after neuroblastoma or nephroblastoma. METHODS This is the pharmacogenetic substudy of the research project Structural Optimization for Children With Cancer After Anthracycline Therapy (LESS-Anthra). We invited 2158 survivors of childhood neuroblastoma or nephroblastoma treated with doxorubicin according to the trial protocols of SIOP 9/GPOH, SIOP 93-01/GPOH, SIOP 2001/GPOH, NB 90, NB 97, or NB 2004 to participate in this prospective cross-sectional cohort study. The study participants underwent a cardiological examination and were asked to provide a blood or saliva sample for genotyping. The study participants' health statuses and cardiovascular diagnoses were recorded using a questionnaire completed by the cardiologist. Digital echocardiographic data were centrally evaluated to determine the contractile function parameters. Medical data on the tumor diagnosis and treatment protocol were taken from the study documentation. Survivors were screened for variants of several candidate genes by TaqMan genotyping. RESULTS This study includes 657 survivors treated with doxorubicin for childhood cancer, the largest German cohort assembled to date to investigate cardiovascular late effects. Data analyses are yet to be completed. CONCLUSIONS This study will define the genetic risk related to 3 marker genes proposed in a pharmacogenetic guideline for risk assessment. Moreover, the results of this study will show the prevalence of cardiovascular dysfunction in survivors of pediatric neuroblastoma or nephroblastoma who were treated with doxorubicin. The results will help to improve primary treatment and follow-up care, thus reducing cardiovascular late effects in the growing population of childhood cancer survivors. TRIAL REGISTRATION German Clinical Trials Register DRKS00015084; https://www.drks.de/drks_web/navigate.do?navigationId=trial.HTML&TRIAL_ID=DRKS00015084. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) DERR1-10.2196/27898.
Collapse
Affiliation(s)
- Oliver Zolk
- Institute of Clinical Pharmacology, Brandenburg Medical School (Theodor Fontane), Immanuel Klinik Rüdersdorf, Rüdersdorf, Germany
| | - Annika von dem Knesebeck
- Institute of Pharmacology of Natural Products & Clinical Pharmacology, University of Ulm, Ulm, Germany
| | - Norbert Graf
- Department of Pediatric Oncology and Hematology, Saarland University, Homburg, Germany
| | - Thorsten Simon
- Department of Pediatric Oncology and Hematology, Children's Hospital, University of Cologne, Cologne, Germany
| | - Barbara Hero
- Department of Pediatric Oncology and Hematology, Children's Hospital, University of Cologne, Cologne, Germany
| | | | | | - Claudia Spix
- German Childhood Cancer Registry, Mainz, Germany
| | - Benjamin Mayer
- Institute of Epidemiology and Medical Biometry, University of Ulm, Ulm, Germany
| | - Susanne Elsner
- Institute for Social Medicine and Epidemiology, University of Lübeck, Lübeck, Germany
| | - Judith Gebauer
- Department of Internal Medicine I, University Hospital of Schleswig-Holstein, Lübeck, Germany
| | - Thorsten Langer
- Department of Pediatric Oncology and Hematology, University Hospital for Children and Adolescents, Lübeck, Germany
| |
Collapse
|
20
|
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
|
21
|
Viswanathan T, Lang CC, Petty RD, Baxter MA. Cardiotoxicity and Chemotherapy-The Role of Precision Medicine. Diseases 2021; 9:90. [PMID: 34940028 PMCID: PMC8699963 DOI: 10.3390/diseases9040090] [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: 08/30/2021] [Revised: 11/25/2021] [Accepted: 11/28/2021] [Indexed: 11/16/2022] Open
Abstract
Cancer and cardiovascular disease are the leading causes of death in the United Kingdom. Many systemic anticancer treatments are associated with short- and long-term cardiotoxicity. With improving cancer survival and an ageing population, identifying those patients at the greatest risk of cardiotoxicity from their cancer treatment is becoming a research priority and has led to a new subspecialty: cardio-oncology. In this concise review article, we discuss cardiotoxicity and systemic anticancer therapy, with a focus on chemotherapy. We also discuss the challenge of identifying those at risk and the role of precision medicine as we strive for a personalised approach to this clinical scenario.
Collapse
Affiliation(s)
- Thyla Viswanathan
- Dundee School of Medicine, Ninewells Hospital, University of Dundee, Dundee DD2 1SY, UK;
| | - Chim C. Lang
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee DD2 1SY, UK; (C.C.L.); (R.D.P.)
- UKM Medical Molecular Biology Institute (UMBI), Jalan Yaacob Latif, Cheras, Kuala Lumpur 56000, Malaysia
| | - Russell D. Petty
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee DD2 1SY, UK; (C.C.L.); (R.D.P.)
- Tayside Cancer Centre, Ninewells Hospital and Medical School, NHS Tayside, Dundee DD2 1SY, UK
| | - Mark A. Baxter
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee DD2 1SY, UK; (C.C.L.); (R.D.P.)
- Tayside Cancer Centre, Ninewells Hospital and Medical School, NHS Tayside, Dundee DD2 1SY, UK
| |
Collapse
|
22
|
Zaha VG, Hayek SS, Alexander KM, Beckie TM, Hundley WG, Kondapalli L, Ky B, Leger KJ, Meijers WC, Moslehi JJ, Shah SH. Future Perspectives of Cardiovascular Biomarker Utilization in Cancer Survivors: A Scientific Statement From the American Heart Association. Circulation 2021; 144:e551-e563. [PMID: 34753300 DOI: 10.1161/cir.0000000000001032] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Improving cancer survival represents the most significant effect of precision medicine and personalized molecular and immunologic therapeutics. Cardiovascular health becomes henceforth a key determinant for the direction of overall outcomes after cancer. Comprehensive tissue diagnostic studies undoubtedly have been and continue to be at the core of the fight against cancer. Will a systemic approach integrating circulating blood-derived biomarkers, multimodality imaging technologies, strategic panomics, and real-time streams of digitized physiological data overcome the elusive cardiovascular tissue diagnosis in cardio-oncology? How can such a systemic approach be personalized for application in day-to-day clinical work, with diverse patient populations, cancer diagnoses, and therapies? To address such questions, this scientific statement approaches a broad definition of the biomarker concept. It summarizes the current literature on the utilization of a multitude of established cardiovascular biomarkers at the intersection with cancer. It identifies limitations and gaps of knowledge in the application of biomarkers to stratify the cardiovascular risk before cancer treatment, monitor cardiovascular health during cancer therapy, and detect latent cardiovascular damage in cancer survivors. Last, it highlights areas in biomarker discovery, validation, and clinical application for concerted efforts from funding agencies, scientists, and clinicians at the cardio-oncology nexus.
Collapse
|
23
|
Nasiri-Aghdam M, Garcia-Garduño TC, Jave-Suárez LF. CELF Family Proteins in Cancer: Highlights on the RNA-Binding Protein/Noncoding RNA Regulatory Axis. Int J Mol Sci 2021; 22:11056. [PMID: 34681716 PMCID: PMC8537729 DOI: 10.3390/ijms222011056] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/06/2021] [Accepted: 10/10/2021] [Indexed: 12/17/2022] Open
Abstract
Post-transcriptional modifications to coding and non-coding RNAs are unquestionably a pivotal way in which human mRNA and protein diversity can influence the different phases of a transcript's life cycle. CELF (CUGBP Elav-like family) proteins are RBPs (RNA-binding proteins) with pleiotropic capabilities in RNA processing. Their responsibilities extend from alternative splicing and transcript editing in the nucleus to mRNA stability, and translation into the cytoplasm. In this way, CELF family members have been connected to global alterations in cancer proliferation and invasion, leading to their identification as potential tumor suppressors or even oncogenes. Notably, genetic variants, alternative splicing, phosphorylation, acetylation, subcellular distribution, competition with other RBPs, and ultimately lncRNAs, miRNAs, and circRNAs all impact CELF regulation. Discoveries have emerged about the control of CELF functions, particularly via noncoding RNAs, and CELF proteins have been identified as competing, antagonizing, and regulating agents of noncoding RNA biogenesis. On the other hand, CELFs are an intriguing example through which to broaden our understanding of the RBP/noncoding RNA regulatory axis. Balancing these complex pathways in cancer is undeniably pivotal and deserves further research. This review outlines some mechanisms of CELF protein regulation and their functional consequences in cancer physiology.
Collapse
Affiliation(s)
- Maryam Nasiri-Aghdam
- División de Inmunología, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara 44340, Mexico;
- Doctorado en Genética Humana, Departamento de Biología Molecular y Genómica, Universidad de Guadalajara, Guadalajara 44340, Mexico;
| | - Texali C. Garcia-Garduño
- Doctorado en Genética Humana, Departamento de Biología Molecular y Genómica, Universidad de Guadalajara, Guadalajara 44340, Mexico;
- Centro Universitario de Ciencias de la Salud, Instituto de Investigación en Ciencias Biomédicas, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Luis Felipe Jave-Suárez
- División de Inmunología, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara 44340, Mexico;
| |
Collapse
|
24
|
Sawicki KT, Sala V, Prever L, Hirsch E, Ardehali H, Ghigo A. Preventing and Treating Anthracycline Cardiotoxicity: New Insights. Annu Rev Pharmacol Toxicol 2021; 61:309-332. [PMID: 33022184 DOI: 10.1146/annurev-pharmtox-030620-104842] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Anthracyclines are the cornerstone of many chemotherapy regimens for a variety of cancers. Unfortunately, their use is limited by a cumulative dose-dependent cardiotoxicity. Despite more than five decades of research, the biological mechanisms underlying anthracycline cardiotoxicity are not completely understood. In this review, we discuss the incidence, risk factors, types, and pathophysiology of anthracycline cardiotoxicity, as well as methods to prevent and treat this condition. We also summarize and discuss advances made in the last decade in the comprehension of the molecular mechanisms underlying the pathology.
Collapse
Affiliation(s)
- Konrad Teodor Sawicki
- Division of Cardiology, Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA;
| | - Valentina Sala
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy;
| | - Lorenzo Prever
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy;
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy;
| | - Hossein Ardehali
- Division of Cardiology, Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA;
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy;
| |
Collapse
|
25
|
Henderson J, Dubey PK, Patil M, Singh S, Dubey S, Namakkal Soorappan R, Kannappan R, Sethu P, Qin G, Zhang J, Krishnamurthy P. microRNA-377 Signaling Modulates Anticancer Drug-Induced Cardiotoxicity in Mice. Front Cardiovasc Med 2021; 8:737826. [PMID: 34485421 PMCID: PMC8415717 DOI: 10.3389/fcvm.2021.737826] [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: 07/07/2021] [Accepted: 07/26/2021] [Indexed: 11/13/2022] Open
Abstract
Doxorubicin (DOX, an anthracycline) is a widely used chemotherapy agent against various forms of cancer; however, it is also known to induce dose-dependent cardiotoxicity leading to adverse complications. Investigating the underlying molecular mechanisms and strategies to limit DOX-induced cardiotoxicity might have potential clinical implications. Our previous study has shown that expression of microRNA-377 (miR-377) increases in cardiomyocytes (CMs) after cardiac ischemia-reperfusion injury in mice, but its specific role in DOX-induced cardiotoxicity has not been elucidated. In the present study, we investigated the effect of anti-miR-377 on DOX-induced cardiac cell death, remodeling, and dysfunction. We evaluated the role of miR-377 in CM apoptosis, its target analysis by RNA sequencing, and we tested the effect of AAV9-anti-miR-377 on DOX-induced cardiotoxicity and mortality. DOX administration in mice increases miR-377 expression in the myocardium. miR-377 inhibition in cardiomyocyte cell line protects against DOX-induced cell death and oxidative stress. Furthermore, RNA sequencing and Gene Ontology (GO) analysis revealed alterations in a number of cell death/survival genes. Intriguingly, we observed accelerated mortality and enhanced myocardial remodeling in the mice pretreated with AAV9-anti-miR-377 followed by DOX administration as compared to the AAV9-scrambled-control-pretreated mice. Taken together, our data suggest that in vitro miR-377 inhibition protects against DOX-induced cardiomyocyte cell death. On the contrary, in vivo administration of AAV9-anti-miR-377 increases mortality in DOX-treated mice.
Collapse
Affiliation(s)
- John Henderson
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Praveen K Dubey
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Mallikarjun Patil
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Sarojini Singh
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Shubham Dubey
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Rajasekaran Namakkal Soorappan
- Division of Molecular & Cellular Pathology, Department of Pathology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Ramaswamy Kannappan
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Palaniappan Sethu
- Division of Cardiovascular Disease, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Gangjian Qin
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jianyi Zhang
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Prasanna Krishnamurthy
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
26
|
Vazdar L, Gabrić ID, Kruljac I, Pintarić H, Šeparović R, Kirigin Biloš LS, Pavlović M, Tečić Vuger A, Štefanović M. Influence of Ile655Val polymorphism on trastuzumab-induced cardiotoxicity in early-stage HER2 positive breast cancer. Sci Rep 2021; 11:14395. [PMID: 34257352 PMCID: PMC8277791 DOI: 10.1038/s41598-021-93634-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 06/14/2021] [Indexed: 11/08/2022] Open
Abstract
Trastuzumab has improved the prognosis of HER2 positive breast cancer, but cardiotoxicity remains a concern. We aimed to identify risk factors for trastuzumab-induced cardiotoxicity, with an emphasis on the HER2 Ile655Val single nucleotide polymorphism. This single-center case-control study included 1056 patients with early-stage HER2 positive breast cancer that received adjuvant trastuzumab. Cardiotoxicity was defined as a decline in left ventricular ejection fraction (LVEF) > 15% in patients without previous cardiomyopathy, or > 10% in patients with baseline LVEF of < 50%. Patient characteristics and cardiac parameters were compared in 78 (7.38%) cases and 99 randomly assigned controls, and the polymorphism was genotyped using real-time polymerase chain reaction. Cardiotoxicity was independently associated with advanced age (P = 0.024), lower body mass index (P = 0.023), left breast involvement (P = 0.001), N3 status (P = 0.004), diabetes (P = 0.016), and a family history of coronary artery disease (P = 0.019). Genotype distribution was as follows: A/A (Ile/Ile) was found in 111 (62.7%) patients, A/G (Ile/Val) in 60 (33.9%) patients, and G/G (Val/Val) in 6 (3.4%) patients. The genotype was not associated with cardiotoxicity or the severity of heart failure, reversibility, and recovery time. We found no association between the HER2 Ile655Val polymorphism and trastuzumab-induced cardiotoxicity; therefore, we do not recommend routine cardiotoxicity-risk stratification using this polymorphism.
Collapse
Affiliation(s)
- Ljubica Vazdar
- Department of Radiotherapy and Medical Oncology, Clinic for Tumors, University Hospital Center "Sestre Milosrdnice", Zagreb, Croatia
| | - Ivo Darko Gabrić
- Department of Cardiovascular Diseases, Institute for Cardiomyopathies, Heart Failure and Valvular Diseases, University Hospital Center "Sestre Milosrdnice", Zagreb, Croatia
| | - Ivan Kruljac
- Department of Internal Medicine, University Hospital Center "Sestre Milosrdnice", Vinogradska cesta 29, Zagreb, Croatia
| | - Hrvoje Pintarić
- Cardiac Catheterization Laboratory, Department of Cardiovascular Diseases, University Hospital Center "Sestre Milosrdnice", Zagreb, Croatia
- School of Dental Medicine, University of Zagreb, Zagreb, Croatia
| | - Robert Šeparović
- Department of Radiotherapy and Medical Oncology, Clinic for Tumors, University Hospital Center "Sestre Milosrdnice", Zagreb, Croatia
| | - Lora Stanka Kirigin Biloš
- Department of Internal Medicine, University Hospital Center "Sestre Milosrdnice", Vinogradska cesta 29, Zagreb, Croatia.
| | - Mirjana Pavlović
- Department of Radiotherapy and Medical Oncology, Clinic for Tumors, University Hospital Center "Sestre Milosrdnice", Zagreb, Croatia
| | - Ana Tečić Vuger
- Department of Radiotherapy and Medical Oncology, Clinic for Tumors, University Hospital Center "Sestre Milosrdnice", Zagreb, Croatia
| | - Mario Štefanović
- Faculty of Pharmacy and Biochemistry, Clinical Institute of Chemistry, University Hospital Center "Sestre Milosrdnice", University of Zagreb, Zagreb, Croatia
| |
Collapse
|
27
|
Yang X, Li G, Guan M, Bapat A, Dai Q, Zhong C, Yang T, Luo C, An N, Liu W, Yang F, Pan H, Wang P, Gao Y, Gong Y, Das S, Shang H, Xing Y. Potential Gene Association Studies of Chemotherapy-Induced Cardiotoxicity: A Systematic Review and Meta-Analysis. Front Cardiovasc Med 2021; 8:651269. [PMID: 34150864 PMCID: PMC8213036 DOI: 10.3389/fcvm.2021.651269] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/22/2021] [Indexed: 12/14/2022] Open
Abstract
Chemotherapy is widely used in the treatment of cancer patients, but the cardiotoxicity induced by chemotherapy is still a major concern to most clinicians. Currently, genetic methods have been used to detect patients with high risk of chemotherapy-induced cardiotoxicity (CIC), and our study evaluated the correlation between genomic variants and CIC. The systematic literature search was performed in the PubMed, Cochrane Central Register of Controlled Trials (CENTRAL), China Biology Medicine disc (CBMdisc), the Embase database, China National Knowledge Internet (CNKI) and Wanfang database from inception until June 2020. Forty-one studies were identified that examined the relationship between genetic variations and CIC. And these studies examined 88 different genes and 154 single nucleotide polymorphisms (SNPs). Our study indicated 6 variants obviously associated with the increased risk for CIC, including CYBA rs4673 (pooled odds ratio, 1.93; 95% CI, 1.13–3.30), RAC2 rs13058338 (2.05; 1.11–3.78), CYP3A5 rs776746 (2.15; 1.00–4.62) ABCC1 rs45511401 (1.46; 1.05–2.01), ABCC2 rs8187710 (2.19; 1.38–3.48), and HER2-Ile655Val rs1136201 (2.48; 1.53–4.02). Although further studies are required to validate the diagnostic and prognostic roles of these 6 variants in predicting CIC, our study emphasizes the promising benefits of pharmacogenomic screening before chemotherapy to minimize the CIC.
Collapse
Affiliation(s)
- Xinyu Yang
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China.,Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Guoping Li
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Manke Guan
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Aneesh Bapat
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Qianqian Dai
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Changming Zhong
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Tao Yang
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Changyong Luo
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Na An
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China.,Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wenjing Liu
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Fan Yang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Haie Pan
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Pengqian Wang
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China.,Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yonghong Gao
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Ye Gong
- Department of Critical Care Medicine, Shanghai Medical College, Huashan Hospital, Fudan University, Shanghai, China
| | - Saumya Das
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Hongcai Shang
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Yanwei Xing
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
28
|
Hashiesh HM, Sharma C, Goyal SN, Sadek B, Jha NK, Kaabi JA, Ojha S. A focused review on CB2 receptor-selective pharmacological properties and therapeutic potential of β-caryophyllene, a dietary cannabinoid. Biomed Pharmacother 2021; 140:111639. [PMID: 34091179 DOI: 10.1016/j.biopha.2021.111639] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 04/12/2021] [Accepted: 04/19/2021] [Indexed: 02/06/2023] Open
Abstract
The endocannabinoid system (ECS), a conserved physiological system emerged as a novel pharmacological target for its significant role and potential therapeutic benefits ranging from neurological diseases to cancer. Among both, CB1 and CB2R types, CB2R have received attention for its pharmacological effects as antioxidant, anti-inflammatory, immunomodulatory and antiapoptotic that can be achieved without causing psychotropic adverse effects through CB1R. The ligands activate CB2R are of endogenous, synthetic and plant origin. In recent years, β-caryophyllene (BCP), a natural bicyclic sesquiterpene in cannabis as well as non-cannabis plants, has received attention due to its selective agonist property on CB2R. BCP has been well studied in a variety of pathological conditions mediating CB2R selective agonist property. The focus of the present manuscript is to represent the CB2R selective agonist mediated pharmacological mechanisms and therapeutic potential of BCP. The present narrative review summarizes insights into the CB2R-selective pharmacological properties and therapeutic potential of BCP such as cardioprotective, hepatoprotective, neuroprotective, nephroprotective, gastroprotective, chemopreventive, antioxidant, anti-inflammatory, and immunomodulator. The available evidences suggest that BCP, can be an important candidate of plant origin endowed with CB2R selective properties that may provide a pharmacological rationale for its pharmacotherapeutic application and pharmaceutical development like a drug. Additionally, given the wide availability in edible plants and dietary use, with safety, and no toxicity, BCP can be promoted as a nutraceutical and functional food for general health and well-being. Further, studies are needed to explore pharmacological and pharmaceutical opportunities for therapeutic and preventive applications of use of BCP in human diseases.
Collapse
Affiliation(s)
- Hebaallah Mamdouh Hashiesh
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, United Arab Emirates
| | - Charu Sharma
- Department of Internal Medicine, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, United Arab Emirates
| | - Sameer N Goyal
- Shri Vile Parle Kelvani Mandal's Institute of Pharmacy, Dhule 424001, Maharashtra, India
| | - Bassem Sadek
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, United Arab Emirates
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida, Uttar Pradesh 201310, India
| | - Juma Al Kaabi
- Department of Internal Medicine, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, United Arab Emirates
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, United Arab Emirates; Zayed Bin Sultan Al Nahyan Center for Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, United Arab Emirates.
| |
Collapse
|
29
|
Temtanakitpaisan Y, Saengnipanthkul S. Monitoring of Metabolic Syndrome and Cardiovascular Disease in Childhood Cancer Survivors. J Adolesc Young Adult Oncol 2021; 11:17-26. [PMID: 33989069 DOI: 10.1089/jayao.2021.0001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Advances in cancer treatment have significantly improved childhood cancer survival, although metabolic syndrome and cardiovascular disease are common long-term complications that may occur years after treatment. Childhood cancer survivors may not receive appropriate follow-up due to lack of communication between oncologists and primary care physicians, or, from lack of awareness of possible long-term metabolic and cardiovascular complications after cancer treatment. We, therefore, reviewed current evidence on long-term effects of cancer therapy, and appropriate monitoring for long-term treatment effects in childhood cancer survivors that could lead to early detection and prompt treatment to prevent future cardiovascular events.
Collapse
Affiliation(s)
- Yutthapong Temtanakitpaisan
- Division of Cardiology, Bangkok Hospital Khon Kaen, Khon Kaen, Thailand.,Faculty of Medicine, Mahasarakham University, Mahasarakham, Thailand
| | - Suchaorn Saengnipanthkul
- Division of Nutrition, Department of Pediatrics, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
30
|
Macnaught G, Oikonomidou O, Rodgers CT, Clarke W, Cooper A, McVicars H, Hayward L, Mirsadraee S, Semple S, Denvir MA. Cardiac Energetics Before, During, and After Anthracycline-Based Chemotherapy in Breast Cancer Patients Using 31P Magnetic Resonance Spectroscopy: A Pilot Study. Front Cardiovasc Med 2021; 8:653648. [PMID: 33889599 PMCID: PMC8056038 DOI: 10.3389/fcvm.2021.653648] [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: 01/14/2021] [Accepted: 03/12/2021] [Indexed: 11/23/2022] Open
Abstract
Purpose: To explore the utility of phosphorus magnetic resonance spectroscopy (31P MRS) in identifying anthracycline-induced cardiac toxicity in patients with breast cancer. Methods: Twenty patients with newly diagnosed breast cancer receiving anthracycline-based chemotherapy had cardiac magnetic resonance assessment of left ventricular ejection fraction (LVEF) and 31P MRS to determine myocardial Phosphocreatine/Adenosine Triphosphate Ratio (PCr/ATP) at three time points: pre-, mid-, and end-chemotherapy. Plasma high sensitivity cardiac troponin-I (cTn-I) tests and electrocardiograms were also performed at these same time points. Results: Phosphocreatine/Adenosine Triphosphate did not change significantly between pre- and mid-chemo (2.16 ± 0.46 vs. 2.00 ± 0.56, p = 0.80) and pre- and end-chemo (2.16 ± 0.46 vs. 2.17 ± 0.86, p = 0.99). Mean LVEF reduced significantly by 5.1% between pre- and end-chemo (61.4 ± 4.4 vs. 56.3 ± 8.1 %, p = 0.02). Change in PCr/ATP ratios from pre- to end-chemo correlated inversely with changes in LVEF over the same period (r = −0.65, p = 0.006). Plasma cTn-I increased progressively during chemotherapy from pre- to mid-chemo (1.35 ± 0.81 to 4.40 ± 2.64 ng/L; p = 0.01) and from mid- to end-chemo (4.40 ± 2.64 to 18.33 ± 13.23 ng/L; p = 0.001). Conclusions: In this small cohort pilot study, we did not observe a clear change in mean PCr/ATP values during chemotherapy despite evidence of increased plasma cardiac biomarkers and reduced LVEF. Future similar studies should be adequately powered to take account of patient drop-out and variable changes in PCr/ATP and could include T1 and T2 mapping.
Collapse
Affiliation(s)
- Gillian Macnaught
- Edinburgh Imaging Facility, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom.,Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Olga Oikonomidou
- Edinburgh Cancer Research Centre, University of Edinburgh, Edinburgh, United Kingdom.,Edinburgh Cancer Centre, Western General Hospital, Edinburgh, United Kingdom
| | - Christopher T Rodgers
- Department of Clinical Neurosciences, Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, United Kingdom
| | - William Clarke
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Level 0, John Radcliffe Hospital, Oxford, United Kingdom
| | - Annette Cooper
- Edinburgh Imaging Facility, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Heather McVicars
- Edinburgh Cancer Research Centre, University of Edinburgh, Edinburgh, United Kingdom.,Edinburgh Cancer Centre, Western General Hospital, Edinburgh, United Kingdom
| | - Larry Hayward
- Edinburgh Cancer Research Centre, University of Edinburgh, Edinburgh, United Kingdom.,Edinburgh Cancer Centre, Western General Hospital, Edinburgh, United Kingdom
| | - Saeed Mirsadraee
- Department of Cardiology, Royal Brompton Hospital, London, United Kingdom
| | - Scott Semple
- Edinburgh Imaging Facility, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom.,Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Martin A Denvir
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
31
|
Christidi E, Brunham LR. Regulated cell death pathways in doxorubicin-induced cardiotoxicity. Cell Death Dis 2021; 12:339. [PMID: 33795647 PMCID: PMC8017015 DOI: 10.1038/s41419-021-03614-x] [Citation(s) in RCA: 331] [Impact Index Per Article: 82.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 02/01/2023]
Abstract
Doxorubicin is a chemotherapeutic drug used for the treatment of various malignancies; however, patients can experience cardiotoxic effects and this has limited the use of this potent drug. The mechanisms by which doxorubicin kills cardiomyocytes has been elusive and despite extensive research the exact mechanisms remain unknown. This review focuses on recent advances in our understanding of doxorubicin induced regulated cardiomyocyte death pathways including autophagy, ferroptosis, necroptosis, pyroptosis and apoptosis. Understanding the mechanisms by which doxorubicin leads to cardiomyocyte death may help identify novel therapeutic agents and lead to more targeted approaches to cardiotoxicity testing.
Collapse
Affiliation(s)
- Effimia Christidi
- grid.17091.3e0000 0001 2288 9830Centre for Heart Lung Innovation, Department of Medicine, University of British Columbia, Vancouver, BC Canada
| | - Liam R. Brunham
- grid.17091.3e0000 0001 2288 9830Centre for Heart Lung Innovation, Department of Medicine, University of British Columbia, Vancouver, BC Canada ,grid.17091.3e0000 0001 2288 9830Department of Medicine, University of British Columbia, Vancouver, BC Canada ,grid.17091.3e0000 0001 2288 9830Department of Medical Genetics, University of British Columbia, Vancouver, BC Canada
| |
Collapse
|
32
|
Subtle cardiac dysfunction in lymphoma patients receiving low to moderate dose chemotherapy. Sci Rep 2021; 11:7100. [PMID: 33782513 PMCID: PMC8007606 DOI: 10.1038/s41598-021-86652-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 02/26/2021] [Indexed: 12/24/2022] Open
Abstract
Left ventricular (LV) global peak systolic longitudinal strain (GLS) is a sensitive measurement for detecting subtle LV systolic dysfunction and a powerful prognostic predictor. However, the clinical implication of LV GLS in lymphoma patients receiving cancer therapy remains unknown. We prospectively enrolled 74 lymphoma patients (57.9 ± 17.0 years old, 57% male). We performed echocardiographic studies after the 3rd and 6th cycles and 1 year after chemotherapy and a cardiopulmonary exercise test upon completion of 3 cycles of anticancer therapy. Cancer therapy-related cardiac dysfunction (CTRCD) was defined as a ≥ 15% relative reduction in GLS value from baseline. The primary outcome was a composite of all-cause mortality and heart failure events. Thirty-six patients (49%) had CTRCD (LV GLS: baseline vs. after 3rd cycle of therapy: 20.1 ± 2.6 vs. 17.5 ± 2.3%, p < 0.001). CTRCD was detected after the 3rd cycle of anticancer therapy. CTRCD patients had impaired exercise capacity (minute oxygen consumption/kg, CTRCD vs. CTRCD (-): 13.9 ± 3.1 vs. 17.0 ± 3.9 ml/kg/min, p = 0.02). More primary outcome events occurred in the CTRCD group (hazard ratio 3.21; 95% confidence interval 1.04–9.97; p = 0.03). LV GLS could detect subtle but clinically significant cardiac dysfunction in lymphoma patients in the early stage of anticancer therapy. CTRCD may be associated with not only a reduced exercise capacity but also a worse prognosis.
Collapse
|
33
|
Chang K, Yuan C, Liu X. A New RBPs-Related Signature Predicts the Prognosis of Colon Adenocarcinoma Patients. Front Oncol 2021; 11:627504. [PMID: 33767995 PMCID: PMC7985171 DOI: 10.3389/fonc.2021.627504] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/27/2021] [Indexed: 11/13/2022] Open
Abstract
The dysregulation of RNA binding proteins (RBPs) is closely related to tumorigenesis and development. However, the role of RBPs in Colon adenocarcinoma (COAD) is still poorly understood. We downloaded COAD’s RNASeq data from the Cancer Genome Atlas (TCGA) database, screened the differently expressed RBPs in normal tissues and tumor, and constructed a protein interaction network. COAD patients were randomly divided into a training set (N = 315) and a testing set (N = 132). In the training set, univariate Cox analysis identified 12 RBPs significantly related to the prognosis of COAD. By multivariate COX analysis, we constructed a prognostic model composed of five RBPs (CELF4, LRRFIP2, NOP14, PPARGC1A, ZNF385A) based on the lowest Akaike information criterion. Each COAD patient was scored according to the model formula. Further analysis showed that compared with the low-risk group, the overall survival rate (OS) of patients in the high-risk group was significantly lower. The area under the curve of the time-dependent receiver operator characteristic (ROC) curve was 0.722 in the training group and 0.738 in the test group, which confirmed a good prediction feature. In addition, a nomogram was constructed based on clinicopathological characteristics and risk scores. C-index and calibration curve proved the accuracy in predicting the 1-, 3-, and 5-year survival rates of COAD patients. In short, we constructed a superior prognostic and diagnostic signature composed of five RBPs, which indicates new possibilities for individualized treatment of COAD patients.
Collapse
Affiliation(s)
- Kaili Chang
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Chong Yuan
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xueguang Liu
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
34
|
Lam CK, Wu JC. Clinical Trial in a Dish: Using Patient-Derived Induced Pluripotent Stem Cells to Identify Risks of Drug-Induced Cardiotoxicity. Arterioscler Thromb Vasc Biol 2021; 41:1019-1031. [PMID: 33472401 PMCID: PMC11006431 DOI: 10.1161/atvbaha.120.314695] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Drug-induced cardiotoxicity is a significant clinical issue, with many drugs in the market being labeled with warnings on cardiovascular adverse effects. Treatments are often prematurely halted when cardiotoxicity is observed, which limits their therapeutic potential. Moreover, cardiotoxicity is a major reason for abandonment during drug development, reducing available treatment options for diseases and creating a significant financial burden and disincentive for drug developers. Thus, it is important to minimize the cardiotoxic effects of medications that are in use or in development. To this end, identifying patients at a higher risk of developing cardiovascular adverse effects for the drug of interest may be an effective strategy. The discovery of human induced pluripotent stem cells has enabled researchers to generate relevant cell types that retain a patient's own genome and examine patient-specific disease mechanisms, paving the way for precision medicine. Combined with the rapid development of pharmacogenomic analysis, the ability of induced pluripotent stem cell-derivatives to recapitulate patient-specific drug responses provides a powerful platform to identify subsets of patients who are particularly vulnerable to drug-induced cardiotoxicity. In this review, we will discuss the current use of patient-specific induced pluripotent stem cells in identifying populations who are at risk to drug-induced cardiotoxicity and their potential applications in future precision medicine practice. Graphic Abstract: A graphic abstract is available for this article.
Collapse
Affiliation(s)
- Chi Keung Lam
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA
- Department of Biological Sciences, University of Delaware, Newark, DE
| | - Joseph C. Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA
- Department of Radiology, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
35
|
Sammani A, Baas AF, Asselbergs FW, te Riele ASJM. Diagnosis and Risk Prediction of Dilated Cardiomyopathy in the Era of Big Data and Genomics. J Clin Med 2021; 10:921. [PMID: 33652931 PMCID: PMC7956169 DOI: 10.3390/jcm10050921] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/17/2021] [Accepted: 02/22/2021] [Indexed: 12/19/2022] Open
Abstract
Dilated cardiomyopathy (DCM) is a leading cause of heart failure and life-threatening ventricular arrhythmias (LTVA). Work-up and risk stratification of DCM is clinically challenging, as there is great heterogeneity in phenotype and genotype. Throughout the last decade, improved genetic testing of patients has identified genotype-phenotype associations and enhanced evaluation of at-risk relatives leading to better patient prognosis. The field is now ripe to explore opportunities to improve personalised risk assessments. Multivariable risk models presented as "risk calculators" can incorporate a multitude of clinical variables and predict outcome (such as heart failure hospitalisations or LTVA). In addition, genetic risk scores derived from genome/exome-wide association studies can estimate an individual's lifetime genetic risk of developing DCM. The use of clinically granular investigations, such as late gadolinium enhancement on cardiac magnetic resonance imaging, is warranted in order to increase predictive performance. To this end, constructing big data infrastructures improves accessibility of data by using electronic health records, existing research databases, and disease registries. By applying methods such as machine and deep learning, we can model complex interactions, identify new phenotype clusters, and perform prognostic modelling. This review aims to provide an overview of the evolution of DCM definitions as well as its clinical work-up and considerations in the era of genomics. In addition, we present exciting examples in the field of big data infrastructures, personalised prognostic assessment, and artificial intelligence.
Collapse
Affiliation(s)
- Arjan Sammani
- Department of Cardiology, Division Heart & Lungs, University Medical Center Utrecht, Utrecht University, 3582 CX Utrecht, The Netherlands; (A.S.); (F.W.A.)
| | - Annette F. Baas
- Department of Genetics, Division Laboratories, Pharmacy and Biomedical Genetics, University Medical Centre Utrecht, University of Utrecht, 3582 CX Utrecht, The Netherlands;
| | - Folkert W. Asselbergs
- Department of Cardiology, Division Heart & Lungs, University Medical Center Utrecht, Utrecht University, 3582 CX Utrecht, The Netherlands; (A.S.); (F.W.A.)
- Institute of Cardiovascular Science, Faculty of Population Health Sciences, University College London, London WC1E 6BT, UK
- Health Data Research UK and Institute of Health Informatics, University College London, London WC1E 6BT, UK
| | - Anneline S. J. M. te Riele
- Department of Cardiology, Division Heart & Lungs, University Medical Center Utrecht, Utrecht University, 3582 CX Utrecht, The Netherlands; (A.S.); (F.W.A.)
| |
Collapse
|
36
|
Magavern EF, Kaski JC, Turner RM, Drexel H, Janmohamed A, Scourfield A, Burrage D, Floyd CN, Adeyeye E, Tamargo J, Lewis BS, Kjeldsen KP, Niessner A, Wassmann S, Sulzgruber P, Borry P, Agewall S, Semb AG, Savarese G, Pirmohamed M, Caulfield MJ. The Role of Pharmacogenomics in Contemporary Cardiovascular Therapy: A position statement from the European Society of Cardiology Working Group on Cardiovascular Pharmacotherapy. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2021; 8:85-99. [PMID: 33638977 DOI: 10.1093/ehjcvp/pvab018] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/05/2021] [Accepted: 02/24/2021] [Indexed: 12/14/2022]
Abstract
There is a strong and ever-growing body of evidence regarding the use of pharmacogenomics to inform cardiovascular pharmacology. However, there is no common position taken by international cardiovascular societies to unite diverse availability, interpretation and application of such data, nor is there recognition of the challenges of variation in clinical practice between countries within Europe. Aside from the considerable barriers to implementing pharmacogenomic testing and the complexities of clinically actioning results, there are differences in the availability of resources and expertise internationally within Europe. Diverse legal and ethical approaches to genomic testing and clinical therapeutic application also require serious thought. As direct-to-consumer genomic testing becomes more common, it can be anticipated that data may be brought in by patients themselves, which will require critical assessment by the clinical cardiovascular prescriber. In a modern, pluralistic and multi-ethnic Europe, self-identified race/ethnicity may not be concordant with genetically detected ancestry and thus may not accurately convey polymorphism prevalence. Given the broad relevance of pharmacogenomics to areas such as thrombosis and coagulation, interventional cardiology, heart failure, arrhythmias, clinical trials, and policy/regulatory activity within cardiovascular medicine, as well as to genomic and pharmacology subspecialists, this position statement attempts to address these issues at a wide-ranging level.
Collapse
Affiliation(s)
- E F Magavern
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,Department of Clinical Pharmacology, Cardiovascular Medicine, Barts Health NHS Trust, London, UK
| | - J C Kaski
- Molecular and Clinical Sciences Research Institute, St George's, University of London, United Kingdom
| | - R M Turner
- The Wolfson Centre for Personalised Medicine, Institute of Systems, Molecular and Integrative Biology (ISMIB), University of Liverpool, UK.,Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - H Drexel
- Vorarlberg Institute for Vascular Investigation & Treatment (VIVIT), Feldkirch, A Private University of the Principality of Liechtenstein, Triesen, FL.,Drexel University College of Medicine, Philadelphia, USA
| | - A Janmohamed
- Department of Clinical Pharmacology, St George's, University of London, United Kingdom
| | - A Scourfield
- Department of Clinical Pharmacology, University College London Hospital Foundation Trust, UK
| | - D Burrage
- Whittington Health NHS Trust, London, UK
| | - C N Floyd
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, UK.,Department of Clinical Pharmacology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - E Adeyeye
- Department of Clinical Pharmacology, Cardiovascular Medicine, Barts Health NHS Trust, London, UK
| | - J Tamargo
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense, Madrid, Spain
| | - B S Lewis
- Cardiovascular Clinical Research Institute, Lady Davis Carmel Medical Center and the Ruth and Bruce Rappaport School of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Keld Per Kjeldsen
- Department of Cardiology, Copenhagen University Hospital (Amager-Hvidovre), Copenhagen, Denmark.,Department of Health Science and Technology, Faculty of Medicine, Aalborg University, Aalborg, Denmark
| | - A Niessner
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna
| | - S Wassmann
- Cardiology Pasing, Munich, Germany and University of the Saarland, Homburg/Saar, Germany
| | - P Sulzgruber
- Medical University of Vienna, Department of Medicine II, Division of Cardiology
| | - P Borry
- Center for Biomedical Ethics and Law, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium.,Leuven Institute for Human Genetics and Society, Leuven, Belgium
| | - S Agewall
- Oslo University Hospital Ullevål and Institute of Clinical Sciences, University of Oslo, Oslo, Norway
| | - A G Semb
- Preventive Cardio-Rheuma clinic, department of rheumatology, innovation and research, Diakonhjemmet hospital, Oslo, Norway
| | - G Savarese
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden Heart and Vascular Theme, Karolinska University Hospital, Stockholm, Sweden
| | - M Pirmohamed
- The Wolfson Centre for Personalised Medicine, Institute of Systems, Molecular and Integrative Biology (ISMIB), University of Liverpool, UK.,Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK.,Liverpool Health Partners, Liverpool, UK
| | - M J Caulfield
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
37
|
Asselbergs FW, Sammani A, Elliott P, Gimeno JR, Tavazzi L, Tendera M, Kaski JP, Maggioni AP, Rubis PP, Jurcut R, Heliö T, Calò L, Sinagra G, Zdravkovic M, Olivotto I, Kavoliūnienė A, Laroche C, Caforio AL, Charron P. Differences between familial and sporadic dilated cardiomyopathy: ESC EORP Cardiomyopathy & Myocarditis registry. ESC Heart Fail 2021; 8:95-105. [PMID: 33179448 PMCID: PMC7835585 DOI: 10.1002/ehf2.13100] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 09/22/2020] [Accepted: 10/22/2020] [Indexed: 12/16/2022] Open
Abstract
AIMS Dilated cardiomyopathy (DCM) is a complex disease where genetics interplay with extrinsic factors. This study aims to compare the phenotype, management, and outcome of familial DCM (FDCM) and non-familial (sporadic) DCM (SDCM) across Europe. METHODS AND RESULTS Patients with DCM that were enrolled in the prospective ESC EORP Cardiomyopathy & Myocarditis Registry were included. Baseline characteristics, genetic testing, genetic yield, and outcome were analysed comparing FDCM and SDCM; 1260 adult patients were studied (238 FDCM, 707 SDCM, and 315 not disclosed). Patients with FDCM were younger (P < 0.01), had less severe disease phenotype at presentation (P < 0.02), more favourable baseline cardiovascular risk profiles (P ≤ 0.007), and less medication use (P ≤ 0.042). Outcome at 1 year was similar and predicted by NYHA class (HR 0.45; 95% CI [0.25-0.81]) and LVEF per % decrease (HR 1.05; 95% CI [1.02-1.08]. Throughout Europe, patients with FDCM received more genetic testing (47% vs. 8%, P < 0.01) and had higher genetic yield (55% vs. 22%, P < 0.01). CONCLUSIONS We observed that FDCM and SDCM have significant differences at baseline but similar short-term prognosis. Whether modification of associated cardiovascular risk factors provide opportunities for treatment remains to be investigated. Our results also show a prevalent role of genetics in FDCM and a non-marginal yield in SDCM although genetic testing is largely neglected in SDCM. Limited genetic testing and heterogeneity in panels provides a scaffold for improvement of guideline adherence.
Collapse
Affiliation(s)
- Folkert W. Asselbergs
- Department of CardiologyUniversity Medical Centre Utrecht, University of UtrechtHeidelberglaan 100Utrecht3584CXThe Netherlands
- Institute of Cardiovascular Science and Institute of Health Informatics, Faculty of Population Health SciencesUniversity College LondonLondonUK
| | - Arjan Sammani
- Department of CardiologyUniversity Medical Centre Utrecht, University of UtrechtHeidelberglaan 100Utrecht3584CXThe Netherlands
| | - Perry Elliott
- Barts Heart Centre, St Bartholomew's HospitalUniversity College London and Inherited Cardiac Diseases UnitLondonUK
| | - Juan R. Gimeno
- Cardiac DepartmentHospital Universitario Virgen de la ArrixacaMurciaSpain
| | - Luigi Tavazzi
- GVM Care & ResearchMaria Cecilia HospitalCotignolaItaly
| | - Michael Tendera
- Department of Cardiology and Structural Heart Diseases, School of Medicine in KatowiceMedical University of SilesiaKatowicePoland
| | - Juan Pablo Kaski
- Centre for Inherited Cardiovascular Diseases, Great Ormond Street HospitalUK and University College London Institute of Cardiovascular ScienceLondonUK
| | - Aldo P. Maggioni
- GVM Care & ResearchMaria Cecilia HospitalCotignolaItaly
- EUR Observational Research Programme, European Society of CardiologySophia‐AntipolisFrance
| | - Pawel P. Rubis
- Department of Cardiac and Vascular DiseasesJagiellonian University Medical College, John Paul II HospitalKrakowPoland
| | - Ruxandra Jurcut
- Department of CardiologyEmergency Institute of Cardiovascular Diseases C.C. IliescuBucharestRomania
| | - Tiina Heliö
- Department of CardiologyHelsinki University Central Hospital MeilahtiHelsinkiFinland
| | | | - Gianfranco Sinagra
- Cardiovascular Department, Azienda Sanitaria Universitaria Integrata Giuliano IsontinaTriesteItaly
| | - Marija Zdravkovic
- Clinical Hospital Center Bezanijska kosa, Faculty of MedicineUniversity of BelgradeBeogradSerbia
| | | | - Aušra Kavoliūnienė
- Department of CardiologyLithuanian University of Health SciencesKaunasLithuania
| | - Cécile Laroche
- EUR Observational Research Programme, European Society of CardiologySophia‐AntipolisFrance
| | - Alida L.P. Caforio
- Division of Cardiology, Department of Cardiological Thoracic and Vascular Sciences and Public HealthUniversity of PadovaPadovaItaly
| | - Philippe Charron
- APHP, Centre de Référence des Maladies Cardiaques Héréditaires, ICAN, Hôpital Pitié‐SalpêtrièreSorbonne UniversitéParisFrance
| | | |
Collapse
|
38
|
Tantawy M, Pamittan FG, Singh S, Gong Y. Epigenetic Changes Associated With Anthracycline-Induced Cardiotoxicity. Clin Transl Sci 2021; 14:36-46. [PMID: 32770710 PMCID: PMC7877852 DOI: 10.1111/cts.12857] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 07/11/2020] [Indexed: 12/24/2022] Open
Abstract
Advances in cancer treatment have significantly improved the survival of patients with cancer, but, unfortunately, many of these treatments also have long-term complications. Cancer treatment-related cardiotoxicities are becoming a significant clinical problem that a new discipline, Cardio-Oncology, was established to advance the cardiovascular care of patients with growing cancer populations. Anthracyclines are a class of chemotherapeutic agents used to treat many cancers in adults and children. Their clinical use is limited by anthracycline-induced cardiotoxicity (AIC), which can lead to heart failure. Early-onset cardiotoxicity appears within a year of treatment, whereas late-onset cardiotoxicity occurs > 1 year and even up to decades after treatment completion. The pathophysiology of AIC was hypothesized to be caused by generation of reactive oxygen species that lead to lipid peroxidation, defective mitochondrial biogenesis, and DNA damage of the cardiomyocytes. The accumulation of anthracycline metabolites was also proposed to cause mitochondrial damage and the induction of cardiac cell apoptosis, which induces arrhythmias, contractile dysfunction, and cardiomyocyte death. This paper will provide a general overview of cardiotoxicity focusing on the effect of anthracyclines and their epigenetic molecular mechanisms on cardiotoxicity.
Collapse
Affiliation(s)
- Marwa Tantawy
- Department of Pharmacotherapy and Translational ResearchCollege of PharmacyUniversity of FloridaGainesvilleFloridaUSA
- Center for Pharmacogenomics and Precision MedicineCollege of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Frances G. Pamittan
- Department of Pharmacotherapy and Translational ResearchCollege of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | | | - Yan Gong
- Department of Pharmacotherapy and Translational ResearchCollege of PharmacyUniversity of FloridaGainesvilleFloridaUSA
- Center for Pharmacogenomics and Precision MedicineCollege of PharmacyUniversity of FloridaGainesvilleFloridaUSA
- UF Health Cancer CenterGainesvilleFloridaUSA
| |
Collapse
|
39
|
Chaix MA, Parmar N, Kinnear C, Lafreniere-Roula M, Akinrinade O, Yao R, Miron A, Lam E, Meng G, Christie A, Manickaraj AK, Marjerrison S, Dillenburg R, Bassal M, Lougheed J, Zelcer S, Rosenberg H, Hodgson D, Sender L, Kantor P, Manlhiot C, Ellis J, Mertens L, Nathan PC, Mital S. Machine Learning Identifies Clinical and Genetic Factors Associated With Anthracycline Cardiotoxicity in Pediatric Cancer Survivors. JACC: CARDIOONCOLOGY 2020; 2:690-706. [PMID: 34396283 PMCID: PMC8352204 DOI: 10.1016/j.jaccao.2020.11.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 11/03/2020] [Accepted: 11/03/2020] [Indexed: 12/17/2022]
Abstract
Background Despite known clinical risk factors, predicting anthracycline cardiotoxicity remains challenging. Objectives This study sought to develop a clinical and genetic risk prediction model for anthracycline cardiotoxicity in childhood cancer survivors. Methods We performed exome sequencing in 289 childhood cancer survivors at least 3 years from anthracycline exposure. In a nested case-control design, 183 case patients with reduced left ventricular ejection fraction despite low-dose doxorubicin (≤250 mg/m2), and 106 control patients with preserved left ventricular ejection fraction despite doxorubicin >250 mg/m2 were selected as extreme phenotypes. Rare/low-frequency variants were collapsed to identify genes differentially enriched for variants between case patients and control patients. The expression levels of 5 top-ranked genes were evaluated in human induced pluripotent stem cell–derived cardiomyocytes, and variant enrichment was confirmed in a replication cohort. Using random forest, a risk prediction model that included genetic and clinical predictors was developed. Results Thirty-one genes were differentially enriched for variants between case patients and control patients (p < 0.001). Only 42.6% case patients harbored a variant in these genes compared to 89.6% control patients (odds ratio: 0.09; 95% confidence interval: 0.04 to 0.17; p = 3.98 × 10–15). A risk prediction model for cardiotoxicity that included clinical and genetic factors had a higher prediction accuracy and lower misclassification rate compared to the clinical-only model. In vitro inhibition of gene-associated pathways (PI3KR2, ZNF827) provided protection from cardiotoxicity in cardiomyocytes. Conclusions Our study identified variants in cardiac injury pathway genes that protect against cardiotoxicity and informed the development of a prediction model for delayed anthracycline cardiotoxicity, and it also provided new targets in autophagy genes for the development of cardio-protective drugs. (Preventing Cardiac Sequelae in Pediatric Cancer Survivors [PCS2]; NCT01805778)
Collapse
Key Words
- AUC, area under the curve
- CI, confidence interval
- DMSO, dimethyl sulfoxide
- DOX, doxorubicin
- GSEA, gene set enrichment analysis
- H2AX, H2A family member X
- IC50, half-maximal inhibitory concentration
- LV, left ventricular
- LVEF, left ventricular ejection fraction
- MAF, minor allele frequency
- OR, odds ratio
- PGP, Personal Genome Project
- RF, random forest
- SKAT, sequence kernel association test
- SNV, single-nucleotide variant
- anthracycline
- cancer survivorship
- cardiomyopathy
- echocardiography
- genomics
- hiPSC-CM, human induced pluripotent stem cell–derived cardiomyocyte
- mRNA, messenger RNA
- machine learning
- risk prediction
Collapse
Affiliation(s)
- Marie-A Chaix
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada.,Adult Congenital Centre, Montréal Heart Institute, Université de Montréal, Montréal, Canada
| | - Neha Parmar
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Caroline Kinnear
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Myriam Lafreniere-Roula
- Ted Rogers Computational Medicine Program, University Health Network, Toronto, Ontario, Canada
| | - Oyediran Akinrinade
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Roderick Yao
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Anastasia Miron
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Emily Lam
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Guoliang Meng
- Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Anne Christie
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Ashok Kumar Manickaraj
- Department of Molecular Genetics, University of Toronto, Ontario, Canada.,Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Stacey Marjerrison
- Department of Pediatrics, McMaster University Children's Hospital, Hamilton, Ontario, Canada
| | - Rejane Dillenburg
- Department of Pediatrics, McMaster University Children's Hospital, Hamilton, Ontario, Canada
| | - Mylène Bassal
- Department of Pediatrics, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, Ontario, Canada
| | - Jane Lougheed
- Department of Pediatrics, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, Ontario, Canada
| | - Shayna Zelcer
- Department of Pediatrics, Children's Hospital, London Health Sciences Centre, London, Ontario, Canada
| | - Herschel Rosenberg
- Department of Pediatrics, Children's Hospital, London Health Sciences Centre, London, Ontario, Canada
| | - David Hodgson
- Radiation Medicine Program, Princess Margaret Cancer Centre, Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Leonard Sender
- Department of Pediatrics, Children's Hospital of Orange County, Orange, California, USA
| | - Paul Kantor
- Department of Pediatrics, Children's Hospital of Los Angeles, Los Angeles, California, USA
| | - Cedric Manlhiot
- Department of Pediatrics, Johns Hopkins Medical Center, Baltimore, Maryland, USA
| | - James Ellis
- Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Molecular Genetics, University of Toronto, Ontario, Canada
| | - Luc Mertens
- Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Paul C Nathan
- Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Seema Mital
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
40
|
Giudice V, Vecchione C, Selleri C. Cardiotoxicity of Novel Targeted Hematological Therapies. Life (Basel) 2020; 10:life10120344. [PMID: 33322351 PMCID: PMC7763613 DOI: 10.3390/life10120344] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 12/16/2022] Open
Abstract
Chemotherapy-related cardiac dysfunction, also known as cardiotoxicity, is a group of drug-related adverse events negatively affecting myocardial structure and functions in patients who received chemotherapy for cancer treatment. Clinical manifestations can vary from life-threatening arrythmias to chronic conditions, such as heart failure or hypertension, which dramatically reduce quality of life of cancer survivors. Standard chemotherapy exerts its toxic effect mainly by inducing oxidative stress and genomic instability, while new targeted therapies work by interfering with signaling pathways important not only in cancer cells but also in myocytes. For example, Bruton’s tyrosine kinase (BTK) inhibitors interfere with class I phosphoinositide 3-kinase isoforms involved in cardiac hypertrophy, contractility, and regulation of various channel forming proteins; thus, off-target effects of BTK inhibitors are associated with increased frequency of arrhythmias, such as atrial fibrillation, compared to standard chemotherapy. In this review, we summarize current knowledge of cardiotoxic effects of targeted therapies used in hematology.
Collapse
Affiliation(s)
- Valentina Giudice
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.V.); (C.S.)
- Clinical Pharmacology, University Hospital “San Giovanni di Dio e Ruggi D’Aragona”, 84131 Salerno, Italy
- Correspondence: ; Tel.: +39-089-672-493
| | - Carmine Vecchione
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.V.); (C.S.)
- IRCCS Neuromed (Mediterranean Neurological Institute), 86077 Pozzilli, Italy
| | - Carmine Selleri
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.V.); (C.S.)
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi D’Aragona”, 84131 Salerno, Italy
| |
Collapse
|
41
|
Sapkota Y. Harnessing Genomics to Predict and Prevent Anthracycline-Associated Cardiotoxicity. JACC CardioOncol 2020; 2:707-709. [PMID: 34396284 PMCID: PMC8352100 DOI: 10.1016/j.jaccao.2020.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Affiliation(s)
- Yadav Sapkota
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
42
|
Sallustio BC, Boddy AV. Is there scope for better individualisation of anthracycline cancer chemotherapy? Br J Clin Pharmacol 2020; 87:295-305. [PMID: 33118175 DOI: 10.1111/bcp.14628] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 10/13/2020] [Accepted: 10/17/2020] [Indexed: 12/11/2022] Open
Abstract
Anthracyclines are used to treat solid and haematological cancers, particularly breast cancers, lymphomas and childhood cancers. Myelosuppression and cardiotoxicity are the primary toxicities that limit treatment duration and/or intensity. Cardiotoxicity, particularly heart failure, is a leading cause of morbidity and mortality in cancer survivors. Cumulative anthracycline dose is a significant predictor of cardiotoxicity risk, suggesting a role for anthracycline pharmacokinetic variability. Population pharmacokinetic modelling in children has shown that doxorubicin clearance in the very young is significantly lower than in older children, potentially contributing to their higher risk of cardiotoxicity. A model of doxorubicin clearance based on body surface area and age offers a patient-centred dose-adjustment strategy that may replace the current disparate initial-dose selection tools, providing a rational way to compensate for pharmacokinetic variability in children aged <7 years. Population pharmacokinetic models in adults have not adequately addressed older ages, obesity, hepatic and renal dysfunction, and potential drug-drug interactions to enable clinical application. Although candidate gene and genome-wide association studies have investigated relationships between genetic variability and anthracycline pharmacokinetics or clinical outcomes, there have been few clinically significant reproducible associations. Precision-dosing of anthracyclines is currently hindered by lack of clinically useful pharmacokinetic targets and models that predict cumulative anthracycline exposures. Combined with known risk factors for cardiotoxicity, the use of advanced echocardiography and biomarkers, future validated pharmacokinetic targets and predictive models could facilitate anthracycline precision dosing that truly maximises efficacy and provides individualised early intervention with cardioprotective therapies in patients at risk of cardiotoxicity.
Collapse
Affiliation(s)
- Benedetta C Sallustio
- Department of Clinical Pharmacology, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South, SA, Australia.,Discipline of Pharmacology, Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Alan V Boddy
- School of Pharmacy and Medical Sciences and UniSA Cancer Research Institute, University of South Australia, Adelaide, SA, Australia
| |
Collapse
|
43
|
Pang L, Liu Z, Wei F, Cai C, Yang X. Improving cardiotoxicity prediction in cancer treatment: integration of conventional circulating biomarkers and novel exploratory tools. Arch Toxicol 2020; 95:791-805. [PMID: 33219404 DOI: 10.1007/s00204-020-02952-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 11/05/2020] [Indexed: 12/31/2022]
Abstract
Early detection strategies and improvements in cancer treatment have dramatically reduced the cancer mortality rate in the United States (US). However, cardiovascular (CV) side effects of cancer therapy are frequent among the 17 million cancer survivors in the US today, and cardiovascular disease (CVD) has become the second leading cause of morbidity and mortality among cancer survivors. Circulating biomarkers are ideal for detecting and monitoring CV side effects of cancer therapy. Here, we summarize the current state of clinical studies on conventional serum and plasma CVD biomarkers to detect and prevent cardiac injury during cancer treatment. We also review how novel exploratory tools such as genetic testing, human stem cell-derived cardiomyocytes, Omics technologies, and artificial intelligence can elucidate underlying molecular and genetic mechanisms of CV injury and to improve predicting cancer therapy-related cardiotoxicity (CTRC). Current regulatory requirements for biomarker qualifications are also addressed. We present generally applicable lessons learned from published studies, particularly on how to improve reproducibility. The combination of conventional circulating biomarkers and novel exploratory tools will pave the way for precision medicine and improve the clinical practice of prediction, detection, and management of CTRC.
Collapse
Affiliation(s)
- Li Pang
- Division of Systems Biology, National Center for Toxicological Research, US. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA.
| | - Zhichao Liu
- Division of Bioinformation and Biostatistics, National Center for Toxicological Research, US. Food and Drug Administration, Jefferson, AR, USA
| | - Feng Wei
- Department of Structural Heart Disease, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Chengzhong Cai
- Division of Systems Biology, National Center for Toxicological Research, US. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA
| | - Xi Yang
- Division of Pharmacology & Toxicology, Office of Cardiology, Hematology, Endocrinology, & Nephrology, Office of New Drug, Center for Drug Evaluation and Research, US. Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
44
|
Bhatia S. Genetics of Anthracycline Cardiomyopathy in Cancer Survivors: JACC: CardioOncology State-of-the-Art Review. JACC CardioOncol 2020; 2:539-552. [PMID: 33364618 PMCID: PMC7757557 DOI: 10.1016/j.jaccao.2020.09.006] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/14/2020] [Accepted: 09/16/2020] [Indexed: 12/31/2022] Open
Abstract
Anthracyclines are an integral part of chemotherapy regimens used to treat a variety of childhood-onset and adult-onset cancers. However, the development of cardiac dysfunction and heart failure often compromises the clinical utility of anthracyclines. The risk of cardiac dysfunction increases with anthracycline dose. This anthracycline-cardiac dysfunction association is modified by several demographic and clinical factors, such as age at anthracycline exposure (<4 years and ≥65 years); female sex; chest radiation; presence of cardiovascular risk factors (diabetes, hypertension); and concurrent use of cyclophosphamide, paclitaxel, and trastuzumab. However, the clinical variables alone yield modest predictive power in detecting cardiac dysfunction. Recently, attention has focused on the molecular basis of anthracycline-related cardiac dysfunction, providing an initial understanding of the mechanism of anthracycline-related cardiomyopathy. This review describes the current state of knowledge with respect to the pathogenesis of anthracycline-related cardiomyopathy and identifies the critical next steps to mitigate this problem.
Collapse
Affiliation(s)
- Smita Bhatia
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
45
|
Kamphuis JAM, Linschoten M, Cramer MJ, Doevendans PA, Asselbergs FW, Teske AJ. Early- and late anthracycline-induced cardiac dysfunction: echocardiographic characterization and response to heart failure therapy. CARDIO-ONCOLOGY 2020; 6:23. [PMID: 33072403 PMCID: PMC7557080 DOI: 10.1186/s40959-020-00079-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 10/02/2020] [Indexed: 12/11/2022]
Abstract
Background Anthracycline-induced cardiac dysfunction (ACD) is a notorious side effect of anticancer treatment. It has been described as a phenomenon of a continuous progressive decline of cardiac function, eventually leading to dilated cardiomyopathy (DCM). This progressive nature suggests that patients with a delayed ACD diagnosis have greater compromise of cardiac function and more adverse remodeling, with a poor response to heart failure (HF) treatment. This study aimed to delineate the impact of a delayed ACD diagnosis on echocardiographic characteristics and response to HF treatment. Methods and results From the population of our cardio-oncology outpatient clinic, 92 ACD patients were included in this study (age 51.6 ± 16.2 years, median cumulative anthracycline dose 329 [200–329] mg/m2), and a median follow-up of 25.0 [9.6–37.2] months after ACD diagnosis. Median time to ACD diagnosis for patients diagnosed early (< 1 year) and late (> 1 year) was 4.0 vs. 47.7 months respectively. There were no echocardiographic differences between patients diagnosed early vs. late (LVEF 43.6 ± 4.9% vs. 43.0 ± 6.2% and iEDV 63.6 vs. 62.9 mL/m2). Eighty-three percent of patients presented with mild LV dysfunction and in 79% the LV was not dilated. Patients diagnosed early were more likely to have (partial) recovery of cardiac function upon HF treatment initiation (p = 0.015). Conclusions In the setting of a cardio-oncology outpatient clinic, patients with ACD presented with a hypokinetic non-dilated cardiomyopathy, rather than typical DCM. Timing of ACD diagnosis did not impact HF disease severity. However, in patients receiving an early diagnosis, cardiac function was more likely to recover upon HF treatment.
Collapse
Affiliation(s)
- Janine A M Kamphuis
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, University of Utrecht, E03.511, PO Box 85500, 3508 GA Utrecht, The Netherlands
| | - Marijke Linschoten
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, University of Utrecht, E03.511, PO Box 85500, 3508 GA Utrecht, The Netherlands
| | - Maarten J Cramer
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, University of Utrecht, E03.511, PO Box 85500, 3508 GA Utrecht, The Netherlands
| | - Pieter A Doevendans
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, University of Utrecht, E03.511, PO Box 85500, 3508 GA Utrecht, The Netherlands.,Netherlands Heart Institute, Utrecht, The Netherlands.,Central Military Hospital, Utrecht, The Netherlands
| | - Folkert W Asselbergs
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, University of Utrecht, E03.511, PO Box 85500, 3508 GA Utrecht, The Netherlands.,Health Data Research UK and Institute of Health Informatics, University College London, London, UK.,Institute of Cardiovascular Science, Faculty of Population Health Sciences, University College London, London, UK
| | - Arco J Teske
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, University of Utrecht, E03.511, PO Box 85500, 3508 GA Utrecht, The Netherlands
| |
Collapse
|
46
|
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
|
47
|
Genetics of Anthracycline-Mediated Cardiotoxicity: Current Status and Challenges. CURRENT CARDIOVASCULAR RISK REPORTS 2020. [DOI: 10.1007/s12170-020-00647-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
48
|
Kadowaki H, Akazawa H, Ishida J, Komuro I. Cancer Therapeutics-Related Cardiac Dysfunction - Insights From Bench and Bedside of Onco-Cardiology. Circ J 2020; 84:1446-1453. [PMID: 32727978 DOI: 10.1253/circj.cj-20-0467] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Improvements in the long-term survival of cancer patients have led to growing awareness of the clinical importance of cancer therapeutics-related cardiac dysfunction (CTRCD), which can have a considerable effect on the prognosis and quality of life of cancer patients and survivors. Under such circumstances, onco-cardiology/cardio-oncology has emerged as a new discipline, with the aim of best managing cardiovascular complications, including CTRCD. Despite the recent accumulation of epidemiological and clinical information regarding CTRCD, the molecular mechanisms underlying the pathogenesis of CTRCD by individual drugs remain to be determined. To achieve the goal of preventing cardiovascular complications in cancer patients and survivors, it is important to elucidate the pathogenic mechanisms and to establish diagnostic strategies with risk prediction and mechanism- and evidence-based therapies against CTRCD.
Collapse
Affiliation(s)
- Hiroshi Kadowaki
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo
| | - Hiroshi Akazawa
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo
| | - Junichi Ishida
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo
| | - Issei Komuro
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo
| |
Collapse
|
49
|
Christidi E, Huang H, Shafaattalab S, Maillet A, Lin E, Huang K, Laksman Z, Davis MK, Tibbits GF, Brunham LR. Variation in RARG increases susceptibility to doxorubicin-induced cardiotoxicity in patient specific induced pluripotent stem cell-derived cardiomyocytes. Sci Rep 2020; 10:10363. [PMID: 32587261 PMCID: PMC7316788 DOI: 10.1038/s41598-020-65979-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 05/11/2020] [Indexed: 01/04/2023] Open
Abstract
Doxorubicin is a potent anticancer drug used to treat a variety of cancer types. However, its use is limited by doxorubicin-induced cardiotoxicity (DIC). A missense variant in the RARG gene (S427L; rs2229774) has been implicated in susceptibility to DIC in a genome wide association study. The goal of this study was to investigate the functional role of this RARG variant in DIC. We used induced pluripotent stem cell derived cardiomyocytes (iPSC-CMs) from patients treated with doxorubicin. iPSC-CMs from individuals who experienced DIC (cases) showed significantly greater sensitivity to doxorubicin compared to iPSC-CMs from doxorubicin-treated individuals who did not develop DIC (controls) in cell viability and optical mapping experiments. Using CRISPR/Cas9, we generated isogenic cell lines that differed only at the RARG locus. Genetic correction of RARG-S427L to wild type resulted in reduced doxorubicin-induced double stranded DNA breaks, reactive oxygen species production, and cell death. Conversely, introduction of RARG-S427L increased susceptibility to doxorubicin. Finally, genetic disruption of the RARG gene resulted in protection from cell death due to doxorubicin treatment. Our findings suggest that the presence of RARG-S427L increases sensitivity to DIC, establishing a direct, causal role for this variant in DIC.
Collapse
Affiliation(s)
- Effimia Christidi
- Centre for Heart Lung Innovation, Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Haojun Huang
- Centre for Heart Lung Innovation, Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Sanam Shafaattalab
- Molecular Cardiac Physiology Group, Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, Canada
- Department of Cardiovascular Science, British Columbia Children's Hospital, Vancouver, Canada
| | | | - Eric Lin
- Molecular Cardiac Physiology Group, Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, Canada
| | - Kate Huang
- Centre for Heart Lung Innovation, Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Zachary Laksman
- Heart Rhythm Services, Division of Cardiology, Department of Medicine, University of British Columbia, Vancouver, Canada
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Margot K Davis
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Glen F Tibbits
- Molecular Cardiac Physiology Group, Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, Canada
- Department of Cardiovascular Science, British Columbia Children's Hospital, Vancouver, Canada
| | - Liam R Brunham
- Centre for Heart Lung Innovation, Department of Medicine, University of British Columbia, Vancouver, Canada.
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
50
|
Cardiovascular Consequences of Skeletal Muscle Impairments in Breast Cancer. Sports (Basel) 2020; 8:sports8060080. [PMID: 32486406 PMCID: PMC7353641 DOI: 10.3390/sports8060080] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 05/22/2020] [Accepted: 05/26/2020] [Indexed: 12/16/2022] Open
Abstract
Breast cancer survivors suffer from disproportionate cardiovascular disease risk compared to age-matched controls. Beyond direct cardiotoxic effects due to treatments such as chemotherapy and radiation, breast-cancer-related reductions in skeletal muscle mass, quality and oxidative capacity may further contribute to cardiovascular disease risk in this population by limiting the ability to engage in aerobic exercise—a known promoter of cardiovascular health. Indeed, 20–30% decreases in peak oxygen consumption are commonly observed in breast cancer survivors, which are indicative of exercise intolerance. Thus, breast-cancer-related skeletal muscle damage may reduce exercise-based opportunities for cardiovascular disease risk reduction. Resistance training is a potential strategy to improve skeletal muscle health in this population, which in turn may enhance the capacity to engage in aerobic exercise and reduce cardiovascular disease risk.
Collapse
|