1
|
Feng W, Wang Q, Tan Y, Qiao J, Liu Q, Yang B, Yang S, Cui L. Early detection of anthracycline-induced cardiotoxicity. Clin Chim Acta 2025; 565:120000. [PMID: 39401650 DOI: 10.1016/j.cca.2024.120000] [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: 09/14/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024]
Abstract
Although anthracyclines are important anticancer agents, their use is limited due to various adverse effects, particularly cardiac toxicity. Mechanisms underlying anthracycline-induced cardiotoxicity (AIC) are complex. Given the irreplaceable role of anthracyclines in treatment of malignancies and other serious diseases, early monitoring of AIC is paramount. In recent years, multiple studies have investigated various biomarkers for early detection of AIC. Currently, the two most common are cardiac troponin and B-type natriuretic peptide. In addition, a range of other molecules, including RNAs, myeloperoxidase (MPO), C-reactive protein (CRP), various genes, and others, also play roles in AIC prediction. Unfortunately, current research indicates a need to validate their sensitivity and specificity of these biomarkers especially in large study populations. In this review, we summarize the mechanisms and potential biomarkers of AIC, although some remain preliminary.
Collapse
Affiliation(s)
- Weimin Feng
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Qingchen Wang
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Yuan Tan
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Jiao Qiao
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Qi Liu
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Boxin Yang
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Shuo Yang
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Liyan Cui
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| |
Collapse
|
2
|
Faggiano A, Gherbesi E, Giordano C, Gamberini G, Vicenzi M, Cuspidi C, Carugo S, Cipolla CM, Cardinale DM. Anthracycline-Induced Subclinical Right Ventricular Dysfunction in Breast Cancer Patients: A Systematic Review and Meta-Analysis. Cancers (Basel) 2024; 16:3883. [PMID: 39594841 PMCID: PMC11592457 DOI: 10.3390/cancers16223883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/01/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
AIM This meta-analysis aims to evaluate the impact of anthracycline chemotherapy on subclinical right ventricular (RV) dysfunction in breast cancer patients, using traditional echocardiographic parameters and strain-based measures, such as the RV global longitudinal strain (RV GLS) and the RV free-wall longitudinal strain (RV FWLS). METHODS AND RESULTS A systematic search was conducted according to PRISMA guidelines, including 15 studies with a total of 1148 breast cancer patients undergoing anthracycline chemotherapy. The primary outcome was the evaluation of changes in RV GLS and RV FWLS pre- and post-chemotherapy. Secondary outcomes included changes in traditional echocardiographic parameters: TAPSE, FAC, and TDI S'. Meta-analysis revealed significant declines in RV function post-chemotherapy across all parameters. RV GLS decreased from 23.99% to 20.35% (SMD: -0.259, p < 0.0001), and RV FWLS from 24.92% to 21.56% (SMD: -0.269, p < 0.0001). Traditional parameters like TAPSE, FAC, and TDI S' also showed reductions, but these were less consistent across studies. A meta-regression analysis showed no significant relationship between post-chemotherapy left ventricular ejection fraction (LVEF) and the changes in RV GLS and RV FWLS, suggesting that RV dysfunction may not be solely a consequence of LV impairment. CONCLUSIONS Anthracycline chemotherapy induces subclinical RV dysfunction in breast cancer patients. RV strain analysis, especially 3D strain, shows greater sensitivity in detecting early dysfunction. However, further research is needed to clarify the clinical significance and prognostic value of these findings, as well as the role of routine RV strain analysis in guiding early interventions.
Collapse
Affiliation(s)
- Andrea Faggiano
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy (M.V.)
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Elisa Gherbesi
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy (M.V.)
| | - Chiara Giordano
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy (M.V.)
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Giacomo Gamberini
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy (M.V.)
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Marco Vicenzi
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy (M.V.)
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Cesare Cuspidi
- Department of Medicine and Surgery, University of Milano-Bicocca, 20125 Milan, Italy
| | - Stefano Carugo
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy (M.V.)
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Carlo M. Cipolla
- Cardioncology Unit, Cardioncology and Second Opinion Division, European Institute of Oncology, I.R.C.C.S., 20141 Milan, Italy (D.M.C.)
| | - Daniela M. Cardinale
- Cardioncology Unit, Cardioncology and Second Opinion Division, European Institute of Oncology, I.R.C.C.S., 20141 Milan, Italy (D.M.C.)
| |
Collapse
|
3
|
Yang Y, Wang Z, Wang N, Yang J, Yang L. CaMKII Exacerbates Doxorubicin-Induced Cardiotoxicity by Promoting Ubiquitination Through USP10 Inhibition. Cancer Med 2024; 13:e70286. [PMID: 39517125 PMCID: PMC11549063 DOI: 10.1002/cam4.70286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 08/22/2024] [Accepted: 09/20/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Doxorubicin (DOX) is an effective anticancer drug, but it has a problem of cardiotoxicity that cannot be ignored. Ca2+/calmodulin-dependent protein kinase II (CaMKII) is tightly associated with the pathological progression of DOX-induced cardiotoxicity. Ubiquitin-specific protease 10 (USP10) plays an important role in many biological processes and cancers. However, its association with DOX-induced cardiotoxicity and CaMKII remains unclear. METHODS H9C2 cells, HL-1 cells and C57BL/6 mice were used to establish the DOX-induced cardiotoxicity model, and the CaMKII-specific inhibitor KN-93 and USP10 specific inhibitor Spautin-1 were used to observe the CaMKII and USP10 effect. In cell experiments, CCK-8 method was used to assess cell viability, LDH kit was used to assess lactate dehydrogenase expression, DCFH-DA staining was used to observe changes in active oxygen content, TUNEL staining was used to observe cell apoptosis, and Western blotting method was used to detect relevant protein markers. The expression of p-CaMKII and USP10 was assessed by immunofluorescence staining. In animal experiments, mouse echocardiograph was used were used to evaluate cardiac function, and HE staining and Masson staining were used to evaluate myocardial injury. Cardiomyocyte apoptosis was detected by TUNEL staining. Western blotting method was used to detect relevant protein markers. RESULTS Our results demonstrated that activation of CaMKII and inhibition of USP10 pathway related to DOX-induced cardiotoxicity. Inhibition of CaMKII with KN-93 ameliorated DOX-induced cardiac dysfunction and cytotoxicity. In addition, CaMKII inhibition prevented DOX-induced apoptosis and ubiquitination. Furthermore, CaMKII inhibition increased USP10 expression in DOX-treated mouse hearts, H9C2 cells and HL-1 cells. At last, the USP10 inhibitor, Spautin-1, blocked the regulatory effect of CaMKII inhibition on apoptosis and ubiquitination in DOX-induced cardiotoxicity. CONCLUSION Our findings revealed that DOX-induced myocardial apoptosis and activated CaMKII through cellular and animal levels, while providing a novel probe into the mechanism of CaMKII action: promoting ubiquitination by inhibiting USP10 aggravated apoptosis.
Collapse
Affiliation(s)
- Yitong Yang
- Department of AnesthesiologyChildren's Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
- Department of Children's Respiratory AsthmaSecond Affiliated Hospital of Shaanxi University of Chinese MedicineXianyangShaanxiChina
| | - Zhenyi Wang
- Department of AnesthesiologyChildren's Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Nisha Wang
- Department of AnesthesiologyChildren's Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Jian Yang
- Department of Cardiovascular SurgeryXijing Hospital, Air Force Medical UniversityXi'anChina
| | - Lifang Yang
- Department of AnesthesiologyChildren's Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| |
Collapse
|
4
|
Bhasin V, Vakilpour A, Scherrer-Crosbie M. Statins for the Primary Prevention of Anthracycline Cardiotoxicity: A Comprehensive Review. Curr Oncol Rep 2024; 26:1197-1204. [PMID: 39002055 PMCID: PMC11480194 DOI: 10.1007/s11912-024-01579-6] [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] [Accepted: 07/02/2024] [Indexed: 07/15/2024]
Abstract
PURPOSE OF REVIEW The aim of this review is two-fold: (1) To examine the mechanisms by which statins may protect from anthracycline-induced cardiotoxicity and (2) To provide a comprehensive overview of the existing clinical literature investigating the role of statins for the primary prevention of anthracycline-induced cardiotoxicity. RECENT FINDINGS The underlying cardioprotective mechanisms associated with statins have not been fully elucidated. Key mechanisms related to the inhibition of Ras homologous (Rho) GTPases have been proposed. Data from observational studies has supported the beneficial role of statins for the primary prevention of anthracycline-induced cardiotoxicity. Recently, several randomized controlled trials investigating the role of statins for the primary prevention of anthracycline-induced cardiotoxicity have produced contrasting results. Statins have been associated with a lower risk of cardiac dysfunction in cancer patients receiving anthracyclines. Further investigation with larger randomized control trials and longer follow-up periods are needed to better evaluate the long-term role of statin therapy and identify the subgroups who benefit most from statin therapy.
Collapse
Affiliation(s)
- Varun Bhasin
- Division of Cardiovascular Medicine and Thalheimer Center for Cardio-Oncology, Perelman Center for Advanced Medicine and Hospital of the University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA, USA
| | - Azin Vakilpour
- Division of Cardiovascular Medicine and Thalheimer Center for Cardio-Oncology, Perelman Center for Advanced Medicine and Hospital of the University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA, USA
| | - Marielle Scherrer-Crosbie
- Division of Cardiovascular Medicine and Thalheimer Center for Cardio-Oncology, Perelman Center for Advanced Medicine and Hospital of the University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA, USA.
| |
Collapse
|
5
|
Jacquemyn X, Zhan J, Van den Eynde J, Cordrey K, Long R, Rao S, Barnes BT, Thompson WR, Danford D, Kutty S. Time course of hypertension and myocardial dysfunction following anthracycline chemotherapy in pediatric patients. IJC HEART & VASCULATURE 2024; 53:101436. [PMID: 38872982 PMCID: PMC11169083 DOI: 10.1016/j.ijcha.2024.101436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 04/11/2024] [Accepted: 05/27/2024] [Indexed: 06/15/2024]
Abstract
Background Anthracyclines are associated with cardiac dysfunction. Little is known about the interplay of pre-existing hypertension and treatment response. We aimed to investigate the relationship between hypertension and the development of cancer therapy-related cardiac dysfunction (CTRCD) in pediatric patients treated with anthracycline chemotherapy. Methods Pediatric patients with cancer who received anthracycline chemotherapy from 2013 to 2021 were retrospectively included. Serial cardiac assessments were conducted during and after chemotherapy. The primary outcome was the development of CTRCD, classified as mild, moderate, or severe according to contemporary definitions. Results Among 190 patients undergoing anthracycline chemotherapy, 34 patients (17.9 %) had hypertension (24 patients Stage 1, and 10 patients Stage 2) at baseline evaluation. Patients underwent chemotherapy for a median of 234.4 days (interquartile range 127.8-690.3 days) and were subsequently followed up. Hypertension was frequent during follow-up 31.3 % (0-3 months), 15.8 % (3-6 months), 21.9 % (0.5-1 years), 24.7 % (1-2 years), 31.1 % (2-4 years) and 35.8 % (beyond 4 years) (P for trend < 0.001). Freedom from mild CTRCD at 5 years was 45.0 %, freedom from moderate CTRCD was 87.8 % at 5 years. Baseline hypertension did not increase the risk of mild (HR 0.77, 95 % CI: 0.41-1.42, P = 0.385) or moderate CTRCD (HR 0.62, 95 % CI: 0.14-2.72, P = 0.504). Patients with baseline hypertension showed different global longitudinal strain (P < 0.001) and LVEF (P < 0.001) patterns during follow-up. Conclusions Pediatric patients often develop CTRCD post-anthracycline chemotherapy. Those with pre-existing hypertension show a unique treatment response, despite no increased CTRCD risk, warranting further investigation.
Collapse
Affiliation(s)
- Xander Jacquemyn
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Department of Cardiovascular Sciences, KU Leuven & Congenital and Structural Cardiology, UZ Leuven, Leuven, Belgium
| | - Junzhen Zhan
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Jef Van den Eynde
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Department of Cardiovascular Sciences, KU Leuven & Congenital and Structural Cardiology, UZ Leuven, Leuven, Belgium
| | - Kyla Cordrey
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Rita Long
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Sruti Rao
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Benjamin T. Barnes
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - W. Reid Thompson
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - David Danford
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Shelby Kutty
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
6
|
Camilli M, Viscovo M, Maggio L, Bonanni A, Torre I, Pellegrino C, Lamendola P, Tinti L, Teofili L, Hohaus S, Lanza GA, Ferdinandy P, Varga Z, Crea F, Lombardo A, Minotti G. Sodium-glucose cotransporter 2 inhibitors and the cancer patient: from diabetes to cardioprotection and beyond. Basic Res Cardiol 2024:10.1007/s00395-024-01059-9. [PMID: 38935171 DOI: 10.1007/s00395-024-01059-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 05/18/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024]
Abstract
Sodium-glucose cotransporter 2 inhibitors (SGLT2i), a new drug class initially designed and approved for treatment of diabetes mellitus, have been shown to exert pleiotropic metabolic and direct cardioprotective and nephroprotective effects that extend beyond their glucose-lowering action. These properties prompted their use in two frequently intertwined conditions, heart failure and chronic kidney disease. Their unique mechanism of action makes SGLT2i an attractive option also to lower the rate of cardiac events and improve overall survival of oncological patients with preexisting cardiovascular risk and/or candidate to receive cardiotoxic therapies. This review will cover biological foundations and clinical evidence for SGLT2i modulating myocardial function and metabolism, with a focus on their possible use as cardioprotective agents in the cardio-oncology settings. Furthermore, we will explore recently emerged SGLT2i effects on hematopoiesis and immune system, carrying the potential of attenuating tumor growth and chemotherapy-induced cytopenias.
Collapse
Affiliation(s)
- Massimiliano Camilli
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy.
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli, 1, 00168, Rome, Italy.
| | - Marcello Viscovo
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Luca Maggio
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli, 1, 00168, Rome, Italy
| | - Alice Bonanni
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli, 1, 00168, Rome, Italy
| | - Ilaria Torre
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli, 1, 00168, Rome, Italy
| | - Claudio Pellegrino
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Priscilla Lamendola
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | - Lorenzo Tinti
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli, 1, 00168, Rome, Italy
| | - Luciana Teofili
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Stefan Hohaus
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Gaetano Antonio Lanza
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli, 1, 00168, Rome, Italy
| | - Peter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
- MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Zoltan Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Filippo Crea
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Center of Excellence of Cardiovascular Sciences, Ospedale Isola Tiberina - Gemelli Isola, Rome, Italy
| | - Antonella Lombardo
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli, 1, 00168, Rome, Italy
| | | |
Collapse
|
7
|
Costanzo V, Ratre YK, Andretta E, Acharya R, Bhaskar LVKS, Verma HK. A Comprehensive Review of Cancer Drug-Induced Cardiotoxicity in Blood Cancer Patients: Current Perspectives and Therapeutic Strategies. Curr Treat Options Oncol 2024; 25:465-495. [PMID: 38372853 DOI: 10.1007/s11864-023-01175-z] [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] [Accepted: 12/19/2023] [Indexed: 02/20/2024]
Abstract
OPINION STATEMENT Cardiotoxicity has emerged as a serious outcome catalyzed by various therapeutic targets in the field of cancer treatment, which includes chemotherapy, radiation, and targeted therapies. The growing significance of cancer drug-induced cardiotoxicity (CDIC) and radiation-induced cardiotoxicity (CRIC) necessitates immediate attention. This article intricately unveils how cancer treatments cause cardiotoxicity, which is exacerbated by patient-specific risks. In particular, drugs like anthracyclines, alkylating agents, and tyrosine kinase inhibitors pose a risk, along with factors such as hypertension and diabetes. Mechanistic insights into oxidative stress and topoisomerase-II-B inhibition are crucial, while cardiac biomarkers show early damage. Timely intervention and prompt treatment, especially with specific agents like dexrazoxane and beta-blockers, are pivotal in the proactive management of CDIC.
Collapse
Affiliation(s)
- Vincenzo Costanzo
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | | | - Emanuela Andretta
- Department of Veterinary Medicine and Animal Productions, University of Naples "Federico II", Naples, Italy
| | - Rakesh Acharya
- Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - L V K S Bhaskar
- Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Henu Kumar Verma
- Department of Immunopathology, Institute of Lungs Health and Immunity, Comprehensive Pneumology Center, Helmholtz Zentrum, Neuherberg, 85764, Munich, Germany.
| |
Collapse
|
8
|
Reis-Mendes A, Ferreira M, Padrão AI, Duarte JA, Duarte-Araújo M, Remião F, Carvalho F, Sousa E, Bastos ML, Costa VM. The Role of Nrf2 and Inflammation on the Dissimilar Cardiotoxicity of Doxorubicin in Two-Time Points: a Cardio-Oncology In Vivo Study Through Time. Inflammation 2024; 47:264-284. [PMID: 37833616 PMCID: PMC10799157 DOI: 10.1007/s10753-023-01908-0] [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: 05/27/2023] [Revised: 09/05/2023] [Accepted: 09/15/2023] [Indexed: 10/15/2023]
Abstract
Doxorubicin (DOX) is a topoisomerase II inhibitor used in cancer therapy. Despite its efficacy, DOX causes serious adverse effects, such as short- and long-term cardiotoxicity. This work aimed to assess the short- and long-term cardiotoxicity of DOX and the role of inflammation and antioxidant defenses on that cardiotoxicity in a mice model. Adult CD-1 male mice received a cumulative dose of 9.0 mg/kg of DOX (2 biweekly intraperitoneal injections (ip), for 3 weeks). One week (1W) or 5 months (5M) after the last DOX administration, the heart was collected. One week after DOX, a significant increase in p62, tumor necrosis factor receptor (TNFR) 2, glutathione peroxidase 1, catalase, inducible nitric oxide synthase (iNOS) cardiac expression, and a trend towards an increase in interleukin (IL)-6, TNFR1, and B-cell lymphoma 2 associated X (Bax) expression was observed. Moreover, DOX induced a decrease on nuclear factor erythroid-2 related factor 2 (Nrf2) cardiac expression. In both 1W and 5M, DOX led to a high density of infiltrating M1 macrophages, but only the 1W-DOX group had a significantly higher number of nuclear factor κB (NF-κB) p65 immunopositive cells. As late effects (5M), an increase in Nrf2, myeloperoxidase, IL-33, tumor necrosis factor-α (TNF-α), superoxide dismutase 2 (SOD2) expression, and a trend towards increased catalase expression were observed. Moreover, B-cell lymphoma 2 (Bcl-2), cyclooxygenase-2 (COX-2), and carbonylated proteins expression decreased, and a trend towards decreased p38 mitogen-activated protein kinase (MAPK) expression were seen. Our study demonstrated that DOX induces adverse outcome pathways related to inflammation and oxidative stress, although activating different time-dependent response mechanisms.
Collapse
Affiliation(s)
- Ana Reis-Mendes
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal.
- UCIBIO - Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.
| | - Mariana Ferreira
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
- UCIBIO - Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Ana Isabel Padrão
- Research Center in Physical Activity, Faculty of Sport, University of Porto, Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, Porto, Portugal
| | - José Alberto Duarte
- Research Center in Physical Activity, Faculty of Sport, University of Porto, Porto, Portugal
- 1H-TOXRUN-Toxicology Research Unit, University Institute of Health Sciences, CESPU, CRL, Gandra, Portugal
| | - Margarida Duarte-Araújo
- LAQV/REQUIMTE, University of Porto, Porto, Portugal
- Department of Immuno-Physiology and Pharmacology, Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Fernando Remião
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
- UCIBIO - Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Félix Carvalho
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
- UCIBIO - Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Emília Sousa
- Laboratory of Organic and Pharmaceutical Chemistry, Chemistry Department, Faculty of Pharmacy, University of Porto, Porto, Portugal
- CIIMAR-Interdisciplinary Centre of Marine and Environmental Research, Porto, Portugal
| | - Maria Lourdes Bastos
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
- UCIBIO - Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Vera Marisa Costa
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal.
- UCIBIO - Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.
| |
Collapse
|
9
|
Adamcova M, Parova H, Lencova-Popelova O, Kollarova-Brazdova P, Baranova I, Slavickova M, Stverakova T, Mikyskova PS, Mazurova Y, Sterba M. Cardiac miRNA expression during the development of chronic anthracycline-induced cardiomyopathy using an experimental rabbit model. Front Pharmacol 2024; 14:1298172. [PMID: 38235109 PMCID: PMC10791979 DOI: 10.3389/fphar.2023.1298172] [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: 09/22/2023] [Accepted: 12/06/2023] [Indexed: 01/19/2024] Open
Abstract
Background: Anthracycline cardiotoxicity is a well-known complication of cancer treatment, and miRNAs have emerged as a key driver in the pathogenesis of cardiovascular diseases. This study aimed to investigate the expression of miRNAs in the myocardium in early and late stages of chronic anthracycline induced cardiotoxicity to determine whether this expression is associated with the severity of cardiac damage. Method: Cardiotoxicity was induced in rabbits via daunorubicin administration (daunorubicin, 3 mg/kg/week; for five and 10 weeks), while the control group received saline solution. Myocardial miRNA expression was first screened using TaqMan Advanced miRNA microfluidic card assays, after which 32 miRNAs were selected for targeted analysis using qRT-PCR. Results: The first subclinical signs of cardiotoxicity (significant increase in plasma cardiac troponin T) were observed after 5 weeks of daunorubicin treatment. At this time point, 10 miRNAs (including members of the miRNA-34 and 21 families) showed significant upregulation relative to the control group, with the most intense change observed for miRNA-1298-5p (29-fold change, p < 0.01). After 10 weeks of daunorubicin treatment, when a further rise in cTnT was accompanied by significant left ventricle systolic dysfunction, only miR-504-5p was significantly (p < 0.01) downregulated, whereas 10 miRNAs were significantly upregulated relative to the control group; at this time-point, the most intense change was observed for miR-34a-5p (76-fold change). Strong correlations were found between the expression of multiple miRNAs (including miR-34 and mir-21 family and miR-1298-5p) and quantitative indices of toxic damage in both the early and late phases of cardiotoxicity development. Furthermore, plasma levels of miR-34a-5p were strongly correlated with the myocardial expression of this miRNA. Conclusion: To the best of our knowledge, this is the first study that describes alterations in miRNA expression in the myocardium during the transition from subclinical, ANT-induced cardiotoxicity to an overt cardiotoxic phenotype; we also revealed how these changes in miRNA expression are strongly correlated with quantitative markers of cardiotoxicity.
Collapse
Affiliation(s)
| | - Helena Parova
- Department of Clinical Biochemistry and Diagnostics, Faculty of Medicine in Hradec Kralove and University Hospital Hradec Kralove, Hradec Kralove, Czechia
| | | | | | - Ivana Baranova
- Department of Clinical Biochemistry and Diagnostics, Faculty of Medicine in Hradec Kralove and University Hospital Hradec Kralove, Hradec Kralove, Czechia
| | - Marcela Slavickova
- Department of Clinical Biochemistry and Diagnostics, Faculty of Medicine in Hradec Kralove and University Hospital Hradec Kralove, Hradec Kralove, Czechia
| | - Tereza Stverakova
- Department of Clinical Biochemistry and Diagnostics, Faculty of Medicine in Hradec Kralove and University Hospital Hradec Kralove, Hradec Kralove, Czechia
| | - Petra Sauer Mikyskova
- Department of Clinical Biochemistry and Diagnostics, Faculty of Medicine in Hradec Kralove and University Hospital Hradec Kralove, Hradec Kralove, Czechia
| | - Yvona Mazurova
- Department of Histology and Embryology, Charles University in Prague, Hradec Kralove, Czechia
| | | |
Collapse
|
10
|
Vaziri Z, Saleki K, Aram C, Alijanizadeh P, Pourahmad R, Azadmehr A, Ziaei N. Empagliflozin treatment of cardiotoxicity: A comprehensive review of clinical, immunobiological, neuroimmune, and therapeutic implications. Biomed Pharmacother 2023; 168:115686. [PMID: 37839109 DOI: 10.1016/j.biopha.2023.115686] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 10/03/2023] [Accepted: 10/08/2023] [Indexed: 10/17/2023] Open
Abstract
Cancer and cardiovascular disorders are known as the two main leading causes of mortality worldwide. Cardiotoxicity is a critical and common adverse effect of cancer-related chemotherapy. Chemotherapy-induced cardiotoxicity has been associated with various cancer treatments, such as anthracyclines, immune checkpoint inhibitors, and kinase inhibitors. Different methods have been reported for the management of chemotherapy-induced cardiotoxicity. In this regard, sodium-glucose cotransporter-2 inhibitors (SGLT2i), a class of antidiabetic agents, have recently been applied to manage heart failure patients. Further, SGLT2i drugs such as EMPA exert protective cardiac and systemic effects. Moreover, it can reduce inflammation through the mediation of major inflammatory components, such as Nucleotide-binding domain-like receptor protein 3 (NLRP3) inflammasomes, Adenosine 5'-monophosphate-activated protein kinase (AMPK), and c-Jun N-terminal kinase (JNK) pathways, Signal transducer and activator of transcription (STAT), and overall decreasing transcription of proinflammatory cytokines. The clinical outcome of EMPA administration is related to improving cardiovascular risk factors, including body weight, lipid profile, blood pressure, and arterial stiffness. Intriguingly, SGLT2 suppressors can regulate microglia-driven hyperinflammation affecting neurological and cardiovascular disorders. In this review, we discuss the protective effects of EMPA in chemotherapy-induced cardiotoxicity from molecular, immunological, and neuroimmunological aspects to preclinical and clinical outcomes.
Collapse
Affiliation(s)
- Zahra Vaziri
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Kiarash Saleki
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; USERN Office, Babol University of Medical Sciences, Babol, Iran; Department of e-Learning, Virtual School of Medical Education and Management, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Cena Aram
- Department of Cell & Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Parsa Alijanizadeh
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Ramtin Pourahmad
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Azadmehr
- Immunology Department, Babol University of Medical Sciences, Babol, Iran
| | - Naghmeh Ziaei
- Clinical Research Development unit of Rouhani Hospital, Babol University of Medical Sciences, Babol, Iran; Department of Cardiology, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
11
|
Sun X, Du J, Meng H, Liu F, Yang N, Deng S, Wan H, Ye D, Song E, Zeng H. Targeting autophagy with SAR405 alleviates doxorubicin-induced cardiotoxicity. Cell Biol Toxicol 2023; 39:3255-3267. [PMID: 37768392 DOI: 10.1007/s10565-023-09831-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023]
Abstract
Anthracycline antitumor agents, such as doxorubicin (DOX), are effective in the treatment of solid tumors and hematological malignancies, but anthracycline-induced cardiotoxicity (AIC) limits their application as chemotherapeutics. Dexrazoxane (DEX) has been adopted to prevent AIC. Using a chronic AIC mouse model, we demonstrated that DEX is insufficient to reverse DOX-induced cardiotoxicity. Although therapies targeting autophagy have been explored to prevent AIC, but whether novel autophagy inhibitors could alleviate or prevent AIC in clinically relevant models needs further investigation. Here, we show that genetic ablation of Atg7, a key regulator in the early phase of autophagy, protected mice against AIC. We further demonstrated that SAR405, a novel autophagy inhibitor, attenuated DOX-induced cytotoxicity. Intriguingly, the combination of DEX and SAR405 protected cells against DOX-induced cardiotoxicity in vivo. Using the cardiomyocyte cell lines AC16 and H9c2, we determined that autophagy was initiated during AIC. Our results suggest that inhibition of autophagy at its early phase with SAR405 combined with DEX represents an effective therapeutic strategy to prevent AIC.
Collapse
Affiliation(s)
- Xiaofan Sun
- Department of Hematology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, Guangdong, China
| | - Juan Du
- Department of Hematology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, Guangdong, China
| | - Heng Meng
- Department of Neurology and Stroke Center, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, Guangdong, China
| | - Fangshu Liu
- Department of Hematology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, Guangdong, China
| | - Nianhui Yang
- Department of Hematology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, Guangdong, China
| | - Suqi Deng
- Department of Hematology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, Guangdong, China
| | - Heng Wan
- Division of Vascular and Interventional Radiology, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Dewei Ye
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, 510630, Guangdong, China
| | - Erfei Song
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, Guangdong, China.
| | - Hui Zeng
- Department of Hematology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, Guangdong, China.
| |
Collapse
|
12
|
Sunoqrot S, Abusulieh S, Abusara OH. Identifying synergistic combinations of Doxorubicin-Loaded polyquercetin nanoparticles and natural Products: Implications for breast cancer therapy. Int J Pharm 2023; 645:123392. [PMID: 37683979 DOI: 10.1016/j.ijpharm.2023.123392] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 08/11/2023] [Accepted: 09/05/2023] [Indexed: 09/10/2023]
Abstract
Combining chemotherapeutic agents with bioactive natural products is an attractive cancer treatment modality to reduce the dose and side effects of chemotherapy. Combination treatments with drugs having different mechanisms of action can also be beneficial in combatting the development of drug resistance by cancer cells. Nanoparticle (NP)-mediated drug delivery can further improve the therapeutic index of cytotoxic agents by enabling passive and/or active targeting to tumor tissues in vivo. Using doxorubicin (DOX) as a model chemotherapeutic agent, we developed three NP formulations based on polyquercetin (pQCT), an emerging nanocarrier platform. The NPs were co-assembled with DOX, pQCT, and either Pluronic P123, methoxy poly(ethylene glycol)-amine, or D-α-tocopheryl poly(ethylene glycol) 1000 succinate (TPGS). Physicochemical characterization of the NPs revealed them to have a spherical morphology with high monodispersity, excellent drug loading capacity, and sustained drug release. Then, the NPs were evaluated in vitro to determine their potential synergism when combined with the bioactive natural products curcumin (CUR), tannic acid (TA), and thymoquinone (TQ) against breast cancer cells (MCF-7 and MDA-MB-231). Surprisingly, most of the combinations were found to be antagonistic. However, combinations containing CUR exhibited greater pro-apoptotic effects compared to the single agents, with polymer-modified pQCT NPs presenting as a promising nanoplatform for enhancing DOX's ability to promote cancer cell apoptosis. Our findings provide insights into the potential application of pQCT in nanomedicine, as well as the use of bioactive natural products in combination with DOX as a free agent and as an NP formulation in the treatment of breast cancer.
Collapse
Affiliation(s)
- Suhair Sunoqrot
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan.
| | - Samah Abusulieh
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| | - Osama H Abusara
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| |
Collapse
|
13
|
Chen X, Liu C, Zhao H, Zhong Y, Xu Y, Wang Y. Deep learning-assisted high-content screening identifies isoliquiritigenin as an inhibitor of DNA double-strand breaks for preventing doxorubicin-induced cardiotoxicity. Biol Direct 2023; 18:63. [PMID: 37807075 PMCID: PMC10561451 DOI: 10.1186/s13062-023-00412-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/30/2023] [Indexed: 10/10/2023] Open
Abstract
BACKGROUND Anthracyclines including doxorubicin are essential components of many cancer chemotherapy regimens, but their cardiotoxicity severely limits their use. New strategies for treating anthracycline-induced cardiotoxicity (AIC) are still needed. Anthracycline-induced DNA double-strand break (DSB) is the major cause of its cardiotoxicity. However, DSB-based drug screening for AIC has not been performed possibly due to the limited throughput of common assays for detecting DSB. To discover new therapeutic candidates for AIC, here we established a method to rapidly visualize and accurately evaluate the intranuclear anthracycline-induced DSB, and performed a screening for DSB inhibitors. RESULTS First, we constructed a cardiomyocyte cell line stably expressing EGFP-53BP1, in which the formation of EGFP-53BP1 foci faithfully marked the doxorubicin-induced DSB, providing a faster and visible approach to detecting DSB. To quantify the DSB, we used a deep learning-based image analysis method, which showed the better ability to distinguish different cell populations undergoing different treatments of doxorubicin or reference compounds, compared with the traditional threshold-based method. Subsequently, we applied the deep learning-assisted high-content screening method to 315 compounds and found three compounds (kaempferol, kaempferide, and isoliquiritigenin) that exert cardioprotective effects in vitro. Among them, the protective effect of isoliquiritigenin is accompanied by the up-regulation of HO-1, down-regulation of peroxynitrite and topo II, and the alleviation of doxorubicin-induced DSB and apoptosis. The results of animal experiments also showed that isoliquiritigenin maintained the myocardial tissue structure and cardiac function in vivo. Moreover, isoliquiritigenin did not affect the killing of HeLa and MDA-MB-436 cancer cells by doxorubicin and thus has the potential to be a lead compound to exert cardioprotective effects without affecting the antitumor effect of doxorubicin. CONCLUSIONS Our findings provided a new method for the drug discovery for AIC, which combines phenotypic screening with artificial intelligence. The results suggested that isoliquiritigenin as an inhibitor of DSB may be a promising drug candidate for AIC.
Collapse
Affiliation(s)
- Xuechun Chen
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Changtong Liu
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Hong Zhao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yigang Zhong
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Yizhou Xu
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
| | - Yi Wang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, 310020, China.
- Future Health Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314100, China.
| |
Collapse
|
14
|
Lin KH, Ramesh S, Agarwal S, Kuo WW, Kuo CH, Chen MYC, Lin YM, Ho TJ, Huang PC, Huang CY. Fisetin attenuates doxorubicin-induced cardiotoxicity by inhibiting the insulin-like growth factor II receptor apoptotic pathway through estrogen receptor-α/-β activation. Phytother Res 2023; 37:3964-3981. [PMID: 37186468 DOI: 10.1002/ptr.7855] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 03/17/2023] [Accepted: 04/15/2023] [Indexed: 05/17/2023]
Abstract
Doxorubicin (DOX), an effective chemotherapeutic drug, has been used to treat various cancers; however, its cardiotoxic side effects restrict its therapeutic efficacy. Fisetin, a flavonoid phytoestrogen derived from a range of fruits and vegetables, has been reported to exert cardioprotective effects against DOX-induced cardiotoxicity; however, the underlying mechanisms remain unclear. This study investigated fisetin's cardioprotective role and mechanism against DOX-induced cardiotoxicity in H9c2 cardiomyoblasts and ovariectomized (OVX) rat models. MTT assay revealed that fisetin treatment noticeably rescued DOX-induced cell death in a dose-dependent manner. Moreover, western blotting and TUNEL-DAPI staining showed that fisetin significantly attenuated DOX-induced cardiotoxicity in vitro and in vivo by inhibiting the insulin-like growth factor II receptor (IGF-IIR) apoptotic pathway through estrogen receptor (ER)-α/-β activation. The echocardiography, biochemical assay, and H&E staining results demonstrated that fisetin reduced DOX-induced cardiotoxicity by alleviating cardiac dysfunction, myocardial injury, oxidative stress, and histopathological damage. These findings imply that fisetin has a significant therapeutic potential against DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Kuan-Ho Lin
- Department of Emergency Medicine, China Medical University Hospital, Taichung, Taiwan
- College of Medicine, China Medical University, Taichung, Taiwan
| | - Samiraj Ramesh
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Department of Research and Innovation, Institute of Biotechnology, Saveetha School of Engineering (SSE), Saveetha Institute of Medical and Technical Sciences (SIMATS), Thandalam, India
| | - Sakshi Agarwal
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Chia-Hua Kuo
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei, Taiwan
| | - Michael Yu-Chih Chen
- Department of Cardiology, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Yueh-Min Lin
- Department of Medical Technology, Jen-The Junior College of Medicine, Nursing and Management, Miaoli, Taiwan
- Department of Pathology, Changhua Christian Hospital, Changhua, Taiwan
| | - Tsung-Jung Ho
- Department of Chinese Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien, Taiwan
| | - Pei-Chen Huang
- Department of Obstetrics and Gynecology, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Graduate Institute of Medical Science, China Medical University, Taichung, Taiwan
| | - Chih-Yang Huang
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Graduate Institute of Medical Science, China Medical University, Taichung, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan
- Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien, Taiwan
| |
Collapse
|
15
|
Rihackova E, Rihacek M, Vyskocilova M, Valik D, Elbl L. Revisiting treatment-related cardiotoxicity in patients with malignant lymphoma-a review and prospects for the future. Front Cardiovasc Med 2023; 10:1243531. [PMID: 37711551 PMCID: PMC10499183 DOI: 10.3389/fcvm.2023.1243531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 08/14/2023] [Indexed: 09/16/2023] Open
Abstract
Treatment of malignant lymphoma has for years been represented by many cardiotoxic agents especially anthracyclines, cyclophosphamide, and thoracic irradiation. Although they are in clinical practice for decades, the precise mechanism of cardiotoxicity and effective prevention is still part of the research. At this article we discuss most routinely used anti-cancer drugs in chemotherapeutic regiments for malignant lymphoma with the focus on novel insight on molecular mechanisms of cardiotoxicity. Understanding toxicity at molecular levels may unveil possible targets of cardioprotective supportive therapy or optimization of current therapeutic protocols. Additionally, we review novel specific targeted therapy and its challenges in cardio-oncology.
Collapse
Affiliation(s)
- Eva Rihackova
- Department of Internal Medicine and Cardiology, University Hospital Brno and Faculty of Medicine of Masaryk University, Brno, Czech Republic
| | - Michal Rihacek
- Department of Laboratory Medicine, University Hospital Brno, Brno, Czech Republic
- Department of Laboratory Methods, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Biochemistry, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Maria Vyskocilova
- Department of Internal Medicine and Cardiology, University Hospital Brno and Faculty of Medicine of Masaryk University, Brno, Czech Republic
| | - Dalibor Valik
- Department of Laboratory Medicine, University Hospital Brno, Brno, Czech Republic
- Department of Laboratory Methods, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Lubomir Elbl
- Department of Internal Medicine and Cardiology, University Hospital Brno and Faculty of Medicine of Masaryk University, Brno, Czech Republic
| |
Collapse
|
16
|
Zhang W, Wang T, Xue Y, Zhan B, Lai Z, Huang W, Peng X, Zhou Y. Research progress of extracellular vesicles and exosomes derived from mesenchymal stem cells in the treatment of oxidative stress-related diseases. Front Immunol 2023; 14:1238789. [PMID: 37646039 PMCID: PMC10461809 DOI: 10.3389/fimmu.2023.1238789] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 07/24/2023] [Indexed: 09/01/2023] Open
Abstract
There is growing evidence that mesenchymal stem cell-derived extracellular vesicles and exosomes can significantly improve the curative effect of oxidative stress-related diseases. Mesenchymal stem cell extracellular vesicles and exosomes (MSC-EVs and MSC-Exos) are rich in bioactive molecules and have many biological regulatory functions. In this review, we describe how MSC-EVs and MSC-Exos reduce the related markers of oxidative stress and inflammation in various systemic diseases, and the molecular mechanism of MSC-EVs and MSC-Exos in treating apoptosis and vascular injury induced by oxidative stress. The results of a large number of experimental studies have shown that both local and systemic administration can effectively inhibit the oxidative stress response in diseases and promote the survival and regeneration of damaged parenchymal cells. The mRNA and miRNAs in MSC-EVs and MSC-Exos are the most important bioactive molecules in disease treatment, which can inhibit the apoptosis, necrosis and oxidative stress of lung, heart, kidney, liver, bone, skin and other cells, and promote their survive and regenerate.
Collapse
Affiliation(s)
- Wenwen Zhang
- The First Dongguan Affiliated Hospital of Guangdong Medical University, Dongguan, Guangdong, China
- Department of Pathophysiology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Tingyu Wang
- The First Dongguan Affiliated Hospital of Guangdong Medical University, Dongguan, Guangdong, China
- Department of Pathophysiology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Yuanye Xue
- The First Dongguan Affiliated Hospital of Guangdong Medical University, Dongguan, Guangdong, China
- Department of Pathophysiology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Bingbing Zhan
- School of Pharmaceutical Sciences, Guangdong Medical University, Dongguan, China
| | - Zengjie Lai
- The Second Clinical Medical College of Guangdong Medical University, Dongguan, China
| | - Wenjie Huang
- School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Xinsheng Peng
- Biomedical Innovation Center, Guangdong Medical University, Dongguan, China
- Institute of Marine Medicine, Guangdong Medical University, Zhanjiang, China
| | - Yanfang Zhou
- The First Dongguan Affiliated Hospital of Guangdong Medical University, Dongguan, Guangdong, China
- Department of Pathophysiology, Guangdong Medical University, Dongguan, Guangdong, China
| |
Collapse
|
17
|
Chen R, Niu M, Hu X, He Y. Targeting mitochondrial dynamics proteins for the treatment of doxorubicin-induced cardiotoxicity. Front Mol Biosci 2023; 10:1241225. [PMID: 37602332 PMCID: PMC10437218 DOI: 10.3389/fmolb.2023.1241225] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 07/24/2023] [Indexed: 08/22/2023] Open
Abstract
Doxorubicin (DOX) is an extensively used chemotherapeutic agent that can cause severe and frequent cardiotoxicity, which limits its clinical application. Although there have been extensive researches on the cardiotoxicity caused by DOX, there is still a lack of effective treatment. It is necessary to understand the molecular mechanism of DOX-induced cardiotoxicity and search for new therapeutic targets which do not sacrifice their anticancer effects. Mitochondria are considered to be the main target of cardiotoxicity caused by DOX. The imbalance of mitochondrial dynamics characterized by increased mitochondrial fission and inhibited mitochondrial fusion is often reported in DOX-induced cardiotoxicity, which can result in excessive ROS production, energy metabolism disorders, cell apoptosis, and various other problems. Also, mitochondrial dynamics disorder is related to tumorigenesis. Surprisingly, recent studies show that targeting mitochondrial dynamics proteins such as DRP1 and MFN2 can not only defend against DOX-induced cardiotoxicity but also enhance or not impair the anticancer effect. Herein, we summarize mitochondrial dynamics disorder in DOX-induced cardiac injury. Furthermore, we provide an overview of current pharmacological and non-pharmacological interventions targeting proteins involved in mitochondrial dynamics to alleviate cardiac damage caused by DOX.
Collapse
Affiliation(s)
- Rui Chen
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Mengwen Niu
- Department of Rheumatology and Immunology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xin Hu
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yuquan He
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
18
|
Zhang Q, Zhang Y, Xie B, Liu D, Wang Y, Zhou Z, Zhang Y, King E, Tse G, Liu T. Resveratrol activation of SIRT1/MFN2 can improve mitochondria function, alleviating doxorubicin-induced myocardial injury. CANCER INNOVATION 2023; 2:253-264. [PMID: 38089747 PMCID: PMC10686119 DOI: 10.1002/cai2.64] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 02/28/2023] [Accepted: 03/08/2023] [Indexed: 10/11/2023]
Abstract
Background Doxorubicin is a widely used cytotoxic chemotherapy agent for treating different malignancies. However, its use is associated with dose-dependent cardiotoxicity, causing irreversible myocardial damage and significantly reducing the patient's quality of life and survival. In this study, an animal model of doxorubicin-induced cardiomyopathy was used to investigate the pathogenesis of doxorubicin-induced myocardial injury. This study also investigated a possible treatment strategy for alleviating myocardial injury through resveratrol therapy in vitro. Methods Adult male C57BL/6J mice were randomly divided into a control group and a doxorubicin group. Body weight, echocardiography, surface electrocardiogram, and myocardial histomorphology were measured. The mechanisms of doxorubicin cardiotoxicity in H9c2 cell lines were explored by comparing three groups (phosphate-buffered saline, doxorubicin, and doxorubicin with resveratrol). Results Compared to the control group, the doxorubicin group showed a lower body weight and higher systolic arterial pressure, associated with reduced left ventricular ejection fraction and left ventricular fractional shortening, prolonged PR interval, and QT interval. These abnormalities were associated with vacuolation and increased disorder in the mitochondria of cardiomyocytes, increased protein expression levels of α-smooth muscle actin and caspase 3, and reduced protein expression levels of Mitofusin2 (MFN2) and Sirtuin1 (SIRT1). Compared to the doxorubicin group, doxorubicin + resveratrol treatment reduced caspase 3 and manganese superoxide dismutase, and increased MFN2 and SIRT1 expression levels. Conclusion Doxorubicin toxicity leads to abnormal mitochondrial morphology and dysfunction in cardiomyocytes and induces apoptosis by interfering with mitochondrial fusion. Resveratrol ameliorates doxorubicin-induced cardiotoxicity by activating SIRT1/MFN2 to improve mitochondria function.
Collapse
Affiliation(s)
- Qingling Zhang
- Department of Cardiology, Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Tianjin Institute of CardiologyThe Second Hospital of Tianjin Medical UniversityTianjinChina
| | - Yunpeng Zhang
- Department of Cardiology, Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Tianjin Institute of CardiologyThe Second Hospital of Tianjin Medical UniversityTianjinChina
| | - Bingxin Xie
- Department of Cardiology, Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Tianjin Institute of CardiologyThe Second Hospital of Tianjin Medical UniversityTianjinChina
| | - Daiqi Liu
- Department of Cardiology, Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Tianjin Institute of CardiologyThe Second Hospital of Tianjin Medical UniversityTianjinChina
| | - Yueying Wang
- Department of Cardiology, Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Tianjin Institute of CardiologyThe Second Hospital of Tianjin Medical UniversityTianjinChina
| | - Zandong Zhou
- Department of Cardiology, Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Tianjin Institute of CardiologyThe Second Hospital of Tianjin Medical UniversityTianjinChina
| | - Yue Zhang
- Department of Cardiology, Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Tianjin Institute of CardiologyThe Second Hospital of Tianjin Medical UniversityTianjinChina
| | - Emma King
- Epidemiology Research Unit, Cardiovascular Analytics GroupChina‐UK CollaborationHong KongChina
| | - Gary Tse
- Department of Cardiology, Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Tianjin Institute of CardiologyThe Second Hospital of Tianjin Medical UniversityTianjinChina
- Epidemiology Research Unit, Cardiovascular Analytics GroupChina‐UK CollaborationHong KongChina
- Kent and Medway Medical SchoolCanterburyKentUK
| | - Tong Liu
- Department of Cardiology, Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Tianjin Institute of CardiologyThe Second Hospital of Tianjin Medical UniversityTianjinChina
| |
Collapse
|
19
|
Bertorello N, Luksch R, Bisogno G, Haupt R, Spallarossa P, Cenna R, Fagioli F. Cardiotoxicity in children with cancer treated with anthracyclines: A position statement on dexrazoxane. Pediatr Blood Cancer 2023; 70:e30515. [PMID: 37355856 DOI: 10.1002/pbc.30515] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 06/09/2023] [Accepted: 06/10/2023] [Indexed: 06/26/2023]
Abstract
Cardiovascular disease is the leading cause of non-malignant morbidity and mortality in childhood cancer survivors (CCSs). Anthracyclines are included in many treatment regimens for paediatric cancer, but unfortunately, these compounds are cardiotoxic. One in 10 CCSs who has received an anthracycline will develop a symptomatic cardiac event over time. Given the crucial need to mitigate anthracycline-related cardiotoxicity (ARC), the authors critically examined published data to identify effective cardioprotective strategies. Based on their expert analysis of contemporary literature data, it was concluded that consideration should be given for routine use of dexrazoxane in children with cancer who are at risk of ARC.
Collapse
Affiliation(s)
- Nicoletta Bertorello
- Paediatric Oncology Division, Regina Margherita Children's Hospital, AOU Città della Salute e della Scienza, Turin, Italy
| | - Roberto Luksch
- Paediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Gianni Bisogno
- Hematology and Oncology Division, Department of Women's and Children's Health, University of Padova, Padua, Italy
| | - Riccardo Haupt
- Epidemiology and Biostatistics Unit and DOPO clinic, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Paolo Spallarossa
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Rosita Cenna
- Paediatric Oncology Division, Regina Margherita Children's Hospital, AOU Città della Salute e della Scienza, Turin, Italy
| | - Franca Fagioli
- Paediatric Oncology Division, Regina Margherita Children's Hospital, AOU Città della Salute e della Scienza, Turin, Italy
- University of Turin, Turin, Italy
| |
Collapse
|
20
|
Tolani D, Wilcox J, Shyam S, Bansal N. Cardio-oncology for Pediatric and Adolescent/Young Adult Patients. Curr Treat Options Oncol 2023:10.1007/s11864-023-01100-4. [PMID: 37296365 DOI: 10.1007/s11864-023-01100-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/18/2023] [Indexed: 06/12/2023]
Abstract
OPINION STATEMENT As chemotherapy continues to improve the lives of patients with cancer, understanding the effects of these drugs on other organ systems, and the cardiovascular system in particular, has become increasingly important. The effects of chemotherapy on the cardiovascular system are a major determinant of morbidity and mortality in these survivors. Although echocardiography continues to be the most widely used modality for assessing cardiotoxicity, newer imaging modalities and biomarker concentrations may detect subclinical cardiotoxicity earlier. Dexrazoxane continues to be the most effective therapy for preventing anthracycline-induced cardiomyopathy. Neurohormonal modulating drugs have not prevented cardiotoxicity, so their widespread, long-term use for all patients is currently not recommended. Advanced cardiac therapies, including heart transplant, have been successful in cancer survivors with end-stage HF and should be considered for these patients. Research on new targets, especially genetic associations, may produce treatments that help reduce cardiovascular morbidity and mortality.
Collapse
Affiliation(s)
- Drishti Tolani
- Division of Pediatric Cardiology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Julia Wilcox
- Albert Einstein College of Medicine, Bronx, NY, USA
| | - Sharvari Shyam
- Division of Pediatrics, St. Barnabas Hospital, Bronx, NY, USA
| | - Neha Bansal
- Division of Pediatric Cardiology, Mount Sinai Kravis Children's Hospital, New York, NY, USA.
| |
Collapse
|
21
|
Chen Q, Meléndez GC, Lesnefsky EJ. New Insight Into Cardioprotection From Anthracyclines: Still Converging on the Mitochondria. JACC CardioOncol 2023; 5:374-376. [PMID: 37397073 PMCID: PMC10308034 DOI: 10.1016/j.jaccao.2023.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2023] Open
Affiliation(s)
- Qun Chen
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Giselle C Meléndez
- Section on Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
- Section on Comparative Medicine, Department of Pathology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Edward J Lesnefsky
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia, USA
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, Virginia, USA
- Medical Service of the McGuire Veterans Affairs Medical Center, Richmond, Virginia, USA
| |
Collapse
|
22
|
Uruski P, Matuszewska J, Leśniewska A, Rychlewski D, Niklas A, Mikuła-Pietrasik J, Tykarski A, Książek K. An integrative review of nonobvious puzzles of cellular and molecular cardiooncology. Cell Mol Biol Lett 2023; 28:44. [PMID: 37221467 DOI: 10.1186/s11658-023-00451-y] [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: 02/22/2023] [Accepted: 04/17/2023] [Indexed: 05/25/2023] Open
Abstract
Oncologic patients are subjected to four major treatment types: surgery, radiotherapy, chemotherapy, and immunotherapy. All nonsurgical forms of cancer management are known to potentially violate the structural and functional integrity of the cardiovascular system. The prevalence and severity of cardiotoxicity and vascular abnormalities led to the emergence of a clinical subdiscipline, called cardiooncology. This relatively new, but rapidly expanding area of knowledge, primarily focuses on clinical observations linking the adverse effects of cancer therapy with deteriorated quality of life of cancer survivors and their increased morbidity and mortality. Cellular and molecular determinants of these relations are far less understood, mainly because of several unsolved paths and contradicting findings in the literature. In this article, we provide a comprehensive view of the cellular and molecular etiology of cardiooncology. We pay particular attention to various intracellular processes that arise in cardiomyocytes, vascular endothelial cells, and smooth muscle cells treated in experimentally-controlled conditions in vitro and in vivo with ionizing radiation and drugs representing diverse modes of anti-cancer activity.
Collapse
Affiliation(s)
- Paweł Uruski
- Department of Hypertensiology, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Julia Matuszewska
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Aleksandra Leśniewska
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Daniel Rychlewski
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Arkadiusz Niklas
- Department of Hypertensiology, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Justyna Mikuła-Pietrasik
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Andrzej Tykarski
- Department of Hypertensiology, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Krzysztof Książek
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland.
| |
Collapse
|
23
|
Suto H, Suto M, Inui Y, Okamura A. Late-onset doxorubicin-induced congestive heart failure in an elderly cancer survivor: A case report. Front Cardiovasc Med 2023; 10:1124276. [PMID: 37180802 PMCID: PMC10166870 DOI: 10.3389/fcvm.2023.1124276] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 04/11/2023] [Indexed: 05/16/2023] Open
Abstract
Background Recently, the survival rate of patients with cancer has improved annually due to advancements in cancer diagnosis and treatment technologies. Meanwhile, late-onset complications associated with cancer treatment significantly affect survival and quality of life. However, different from pediatric cancer survivors, there is no unified view on the follow-up of late complications in elderly cancer survivors. We reported a case of congestive heart failure as a late-onset complication of doxorubicin (DXR) in an elderly cancer survivor. Case report The patient is an 80-year-old woman with hypertension and chronic renal failure. She received six cycles of chemotherapy for Hodgkin's lymphoma that started in January 201X-2. The total dose of DXR was 300 mg/m2, and a transthoracic echocardiogram (TTE) performed in October 201X-2, showed good left ventricular wall motion (LVWM). In April 201X, she suddenly developed dyspnea. Upon arrival at the hospital, a physical examination revealed orthopnea, tachycardia, and leg edema. A chest radiograph showed cardiac enlargement and pleural effusion. A TTE showed diffusely reduced LVWM and a left ventricular ejection fraction in the 20% range. After close examination, the patient was diagnosed with congestive heart failure due to late-onset DXR-induced cardiomyopathy. Conclusion Late-onset DXR-induced cardiotoxicity is considered high-risk from 250 mg/m2 or higher. Elderly cancer survivors are at higher risk of cardiotoxicity than non-elderly cancer survivors and may require closer follow-up.
Collapse
Affiliation(s)
- Hirotaka Suto
- Department of Medical Oncology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
- Department of Medical Oncology/Hematology, Kakogawa Central City Hospital, Hyogo, Japan
| | - Makiko Suto
- Department of Cardiovascular Medicine, Takarazuka City Hospital, Hyogo, Japan
| | - Yumiko Inui
- Department of Medical Oncology/Hematology, Kakogawa Central City Hospital, Hyogo, Japan
| | - Atsuo Okamura
- Department of Medical Oncology/Hematology, Kakogawa Central City Hospital, Hyogo, Japan
| |
Collapse
|
24
|
Gaytan SL, Lawan A, Chang J, Nurunnabi M, Bajpeyi S, Boyle JB, Han SM, Min K. The beneficial role of exercise in preventing doxorubicin-induced cardiotoxicity. Front Physiol 2023; 14:1133423. [PMID: 36969584 PMCID: PMC10033603 DOI: 10.3389/fphys.2023.1133423] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 02/27/2023] [Indexed: 03/11/2023] Open
Abstract
Doxorubicin is a highly effective chemotherapeutic agent widely used to treat a variety of cancers. However, the clinical application of doxorubicin is limited due to its adverse effects on several tissues. One of the most serious side effects of doxorubicin is cardiotoxicity, which results in life-threatening heart damage, leading to reduced cancer treatment success and survival rate. Doxorubicin-induced cardiotoxicity results from cellular toxicity, including increased oxidative stress, apoptosis, and activated proteolytic systems. Exercise training has emerged as a non-pharmacological intervention to prevent cardiotoxicity during and after chemotherapy. Exercise training stimulates numerous physiological adaptations in the heart that promote cardioprotective effects against doxorubicin-induced cardiotoxicity. Understanding the mechanisms responsible for exercise-induced cardioprotection is important to develop therapeutic approaches for cancer patients and survivors. In this report, we review the cardiotoxic effects of doxorubicin and discuss the current understanding of exercise-induced cardioprotection in hearts from doxorubicin-treated animals.
Collapse
Affiliation(s)
- Samantha L. Gaytan
- Department of Kinesiology, College of Health Sciences, University of Texas at El Paso, El Paso, TX, United States
| | - Ahmed Lawan
- Department of Biological Sciences, College of Science, University of Alabama in Huntsville, Huntsville, AL, United States
| | - Jongwha Chang
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, College Station, TX, United States
| | - Md Nurunnabi
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX, United States
| | - Sudip Bajpeyi
- Department of Kinesiology, College of Health Sciences, University of Texas at El Paso, El Paso, TX, United States
| | - Jason B. Boyle
- Department of Kinesiology, College of Health Sciences, University of Texas at El Paso, El Paso, TX, United States
| | - Sung Min Han
- Department of Physiology and Aging, College of Medicine, Institute on Aging, University of Florida, Gainesville, FL, United States
- *Correspondence: Kisuk Min, ; Sung Min Han,
| | - Kisuk Min
- Department of Kinesiology, College of Health Sciences, University of Texas at El Paso, El Paso, TX, United States
- *Correspondence: Kisuk Min, ; Sung Min Han,
| |
Collapse
|
25
|
Yu X, Yang Y, Chen T, Wang Y, Guo T, Liu Y, Li H, Yang L. Cell death regulation in myocardial toxicity induced by antineoplastic drugs. Front Cell Dev Biol 2023; 11:1075917. [PMID: 36824370 PMCID: PMC9941345 DOI: 10.3389/fcell.2023.1075917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/26/2023] [Indexed: 02/10/2023] Open
Abstract
Homeostatic regulation of cardiomyocytes plays a critical role in maintaining normal physiological activity of cardiac tissue. Severe cardiotoxicity can lead to heart disease, including but not limited to arrhythmias, myocardial infarction and cardiac hypertrophy. In recent years, significant progress has been made in developing new therapies for cancer that have dramatically changed the treatment of several malignancies and continue to improve patient survival, but can also lead to serious cardiac adverse effects. Mitochondria are key organelles that maintain homeostasis in myocardial tissue and have been extensively involved in various cardiovascular disease episodes, including ischemic cardiomyopathy, heart failure and stroke. Several studies support that mitochondrial targeting is a major determinant of the cardiotoxic effects triggered by chemotherapeutic agents increasingly used in solid and hematologic tumors. This antineoplastic therapy-induced mitochondrial toxicity is due to different mechanisms, usually altering the mitochondrial respiratory chain, energy production and mitochondrial kinetics, or inducing mitochondrial oxidative/nitrosative stress, ultimately leading to cell death. This review focuses on recent advances in forms of cardiac cell death and related mechanisms of antineoplastic drug-induced cardiotoxicity, including autophagy, ferroptosis, apoptosis, pyroptosis, and necroptosis, explores and evaluates key proteins involved in cardiac cell death signaling, and presents recent advances in cardioprotective strategies for this disease. It aims to provide theoretical basis and targets for the prevention and treatment of pharmacological cardiotoxicity in clinical settings.
Collapse
Affiliation(s)
- Xue Yu
- Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Yan Yang
- Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Tianzuo Chen
- Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Yuqin Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Tianwei Guo
- Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Yujun Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Hong Li
- Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China,*Correspondence: Liming Yang, ; Hong Li,
| | - Liming Yang
- Department of Pathophysiology, Harbin Medical University-Daqing, Daqing, China,*Correspondence: Liming Yang, ; Hong Li,
| |
Collapse
|
26
|
Gu J, Wang D, Jiang L, Huang Y, Ding L, Chen X, He Y, Zhou Z, Pu D. Assessment of Global Cardiac Function Using AutoSTRAIN Automatic Strain Quantitative Technology in Patients With Breast Cancer Undergoing Anthracycline-Based Chemotherapy. ULTRASOUND IN MEDICINE & BIOLOGY 2023; 49:368-374. [PMID: 36283937 DOI: 10.1016/j.ultrasmedbio.2022.09.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 07/20/2022] [Accepted: 09/23/2022] [Indexed: 06/16/2023]
Abstract
In patients with breast cancer undergoing anthracycline-based chemotherapy, we investigated the deformational parameters of the left ventricle, right ventricle and left atrium, as well as the relationship between these parameters. Ninety-five patients with breast cancer who were treated with anthracycline-based chemotherapy were enrolled. The control group included 116 healthy female volunteers. Parameters including left ventricular global longitudinal strain (LV-GLS); right ventricular free wall longitudinal strain (RVFWSL) and global longitudinal strain (RV4CSL); and peak strain of the left atrium during LV systole (LASR), early LV diastole (LASCD) and late LV diastole (LASCT) were analyzed by speckle tacking echocardiography. LV-GLS, LASR, LASCD, RVFWSL and RV4CSL in the chemotherapy group decreased significantly by 15.6%, 13.8%, 19.8%, 21.8% and 13.2% (p < 0.05), respectively, when compared with the control group. LASCT was slightly increased in the chemotherapy group but the increase was not statistically significant (p > 0.05). Formulas for the influencing factors of LV-GLS were LV-GLS = -18.73738541 + 0.13961 × LVIDd + 0.09672 × LASCD + 0.18113 × RVFWSL in the control group and LV-GLS = -8.026302253 + 0.20811 × LASCD + 0.11084 × LASCT + 0.12153 × RVFWSL in the chemotherapy group. Both LV contraction and RV contraction were impaired after the completion of anthracycline-based therapy, and RVFWSL may be superior to LV-GLS in assessing cardiotoxicity. LA reserve and channel function were significantly reduced, while pump function was slightly increased. Compared with the results among healthy people, the influencing factor of LV-GLS varied after anthracycline treatment, and LA function had a greater impact on LV-GLS.
Collapse
Affiliation(s)
- Jin Gu
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Department of Ultrasound, The Chongqing Public Health Medical Center, Chongqing, China
| | - Dong Wang
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lan Jiang
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuan Huang
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Department of Ultrasound, The Chongqing Public Health Medical Center, Chongqing, China
| | - Lunbi Ding
- Department of Ultrasound, The Chongqing Public Health Medical Center, Chongqing, China
| | - Xueying Chen
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yiman He
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zefen Zhou
- Department of Ultrasound, The Chongqing Public Health Medical Center, Chongqing, China
| | - Darong Pu
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
27
|
Chen Y, Shi S, Dai Y. Research progress of therapeutic drugs for doxorubicin-induced cardiomyopathy. Biomed Pharmacother 2022; 156:113903. [PMID: 36279722 DOI: 10.1016/j.biopha.2022.113903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/16/2022] [Accepted: 10/19/2022] [Indexed: 12/06/2022] Open
|
28
|
Podyacheva E, Shmakova T, Kushnareva E, Onopchenko A, Martynov M, Andreeva D, Toropov R, Cheburkin Y, Levchuk K, Goldaeva A, Toropova Y. Modeling Doxorubicin-Induced Cardiomyopathy With Fibrotic Myocardial Damage in Wistar Rats. Cardiol Res 2022; 13:339-356. [PMID: 36660062 PMCID: PMC9822674 DOI: 10.14740/cr1416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 10/26/2022] [Indexed: 12/23/2022] Open
Abstract
Background Cardiovascular complications, arising after anthracycline chemotherapy, cause a significant deterioration in the life quality and expectancy of those patients who were previously successfully treated for malignant neoplasms. A number of clinical studies have demonstrated that patients with cardiotoxicity manifested during anthracyclines therapy also have extensive fibrotic changes in the cardiac muscle in the long term. Given the lack of an unambiguous understanding of the mechanisms of fibrotic changes formation under doxorubicin treatment in the myocardium, there is the obvious necessity to create a relevant experimental model of chronic doxorubicin-induced cardiomyopathy with fibrotic myocardial lesions and delayed development of diastolic dysfunction. Methods The study was divided into two stages: first stage (creation of acute doxorubicin cardiomyopathy) - 35 male Wistar rats; second stage (creation of chronic doxorubicin cardiomyopathy) - 40 male Wistar rats. The animals were split into eight groups (two control ones and six experimental ones), which determined the doxorubicin dose (first stage: 25, 20.4, 15 mg/kg; second stage: 5, 10, 15 mg/kg, intraperitoneally) and the frequency of injection. Echocardiographic, hematological, histological, and molecular methods were used to confirm the successful modeling of acute and chronic doxorubicin-induced cardiomyopathy with fibrotic lesions. Results A model of administration six times every other day with a cumulative dose of doxorubicin 20 mg/kg is suitable for evaluation of acute cardiotoxicity. The 15 mg/kg doxorubicin dose is highly cardiotoxic; what's more, it correlates with progressive deterioration of the clinical condition of the animals after 2 months. The optimal cumulative dose of doxorubicin leads to clinical manifestations confirmed by echocardiographic, histological, molecular changes associated with the development of chronic doxorubicin-induced cardiomyopathy with fibrotic lesions of the left ventricular of the cardiac muscle and ensure long-term survival of animals is 10 mg/kg doxorubicin. A dose of 5 mg/kg of the doxorubicin does not ensure the development of fibrous changes formation. Conclusion We assume that cumulative dose of 10 mg/kg with a frequency of administration of six times in 2 days can be used to study the mechanisms of anthracycline cardiomyopathy development.
Collapse
Affiliation(s)
- Ekaterina Podyacheva
- Almazov National Medical Research Centre, Ministry of Health of the Russian Federation, 197341, Saint-Petersburg, Russian Federation,Corresponding Author: Ekaterina Podyacheva, Almazov National Medical Research Centre, Ministry of Health of the Russian Federation, 197341, Saint-Petersburg, Russian Federation.
| | - Tatiana Shmakova
- Almazov National Medical Research Centre, Ministry of Health of the Russian Federation, 197341, Saint-Petersburg, Russian Federation
| | - Ekaterina Kushnareva
- Almazov National Medical Research Centre, Ministry of Health of the Russian Federation, 197341, Saint-Petersburg, Russian Federation
| | - Anatoliya Onopchenko
- Almazov National Medical Research Centre, Ministry of Health of the Russian Federation, 197341, Saint-Petersburg, Russian Federation
| | - Mikhail Martynov
- Almazov National Medical Research Centre, Ministry of Health of the Russian Federation, 197341, Saint-Petersburg, Russian Federation
| | - Daria Andreeva
- Almazov National Medical Research Centre, Ministry of Health of the Russian Federation, 197341, Saint-Petersburg, Russian Federation
| | - Roman Toropov
- Almazov National Medical Research Centre, Ministry of Health of the Russian Federation, 197341, Saint-Petersburg, Russian Federation
| | - Yuri Cheburkin
- Almazov National Medical Research Centre, Ministry of Health of the Russian Federation, 197341, Saint-Petersburg, Russian Federation
| | - Ksenia Levchuk
- Almazov National Medical Research Centre, Ministry of Health of the Russian Federation, 197341, Saint-Petersburg, Russian Federation
| | - Alexandra Goldaeva
- Almazov National Medical Research Centre, Ministry of Health of the Russian Federation, 197341, Saint-Petersburg, Russian Federation
| | - Yana Toropova
- Almazov National Medical Research Centre, Ministry of Health of the Russian Federation, 197341, Saint-Petersburg, Russian Federation
| |
Collapse
|
29
|
Kawano I, Adamcova M. MicroRNAs in doxorubicin-induced cardiotoxicity: The DNA damage response. Front Pharmacol 2022; 13:1055911. [PMID: 36479202 PMCID: PMC9720152 DOI: 10.3389/fphar.2022.1055911] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 11/11/2022] [Indexed: 10/17/2023] Open
Abstract
Doxorubicin (DOX) is a chemotherapeutic drug widely used for cancer treatment, but its use is limited by cardiotoxicity. Although free radicals from redox cycling and free cellular iron have been predominant as the suggested primary pathogenic mechanism, novel evidence has pointed to topoisomerase II inhibition and resultant genotoxic stress as the more fundamental mechanism. Recently, a growing list of microRNAs (miRNAs) has been implicated in DOX-induced cardiotoxicity (DIC). This review summarizes miRNAs reported in the recent literature in the context of DIC. A particular focus is given to miRNAs that regulate cellular responses downstream to DOX-induced DNA damage, especially p53 activation, pro-survival signaling pathway inhibition (e.g., AMPK, AKT, GATA-4, and sirtuin pathways), mitochondrial dysfunction, and ferroptosis. Since these pathways are potential targets for cardioprotection against DOX, an understanding of how miRNAs participate is necessary for developing future therapies.
Collapse
Affiliation(s)
| | - Michaela Adamcova
- Department of Physiology, Faculty of Medicine in Hradec Kralove, Charles University in Prague, Hradec Kralove, Czechia
| |
Collapse
|
30
|
Hasbullah JS, Scott EN, Bhavsar AP, Gunaretnam EP, Miao F, Soliman H, Carleton BC, Ross CJD. All-trans retinoic acid (ATRA) regulates key genes in the RARG-TOP2B pathway and reduces anthracycline-induced cardiotoxicity. PLoS One 2022; 17:e0276541. [PMID: 36331922 PMCID: PMC9635745 DOI: 10.1371/journal.pone.0276541] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 10/09/2022] [Indexed: 11/06/2022] Open
Abstract
The effectiveness of anthracycline chemotherapeutics (e.g., doxorubicin) is limited by anthracycline-induced cardiotoxicity (ACT). A nonsynonymous variant (S427L) in the retinoic acid receptor-γ (RARG) gene has been associated with ACT. This variant causes reduced RARG activity, which is hypothesized to lead to increased susceptibility to ACT through reduced activation of the retinoic acid pathway. This study explored the effects of activating the retinoic acid pathway using a RAR-agonist, all-trans retinoic acid (ATRA), in human cardiomyocytes and mice treated with doxorubicin. In human cardiomyocytes, ATRA induced the gene expression of RARs (RARG, RARB) and repressed the expression of topoisomerase II enzyme genes (TOP2A, TOP2B), which encode for the molecular targets of anthracyclines and repressed downstream ACT response genes. Importantly, ATRA enhanced cell survival of human cardiomyocytes exposed to doxorubicin. The protective effect of ATRA was also observed in a mouse model (B6C3F1/J) of ACT, in which ATRA treatment improved heart function compared to doxorubicin-only treated mice. Histological analyses of the heart also indicated that ATRA treatment reduced the pathology associated with ACT. These findings provide additional evidence for the retinoic acid pathway's role in ACT and suggest that the RAR activator ATRA can modulate this pathway to reduce ACT.
Collapse
Affiliation(s)
- Jafar S. Hasbullah
- Department of Medical Genetics, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
- British Columbia Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Erika N. Scott
- Department of Medical Genetics, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
- British Columbia Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Amit P. Bhavsar
- Department of Medical Microbiology and Immunology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Erandika P. Gunaretnam
- British Columbia Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Fudan Miao
- British Columbia Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Hesham Soliman
- School of Biomedical Engineering, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Bruce C. Carleton
- Department of Medical Genetics, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
- British Columbia Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Pediatrics, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Colin J. D. Ross
- Department of Medical Genetics, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
- British Columbia Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
31
|
Podyacheva E, Toropova Y. SIRT1 activation and its effect on intercalated disc proteins as a way to reduce doxorubicin cardiotoxicity. Front Pharmacol 2022; 13:1035387. [PMID: 36408244 PMCID: PMC9672938 DOI: 10.3389/fphar.2022.1035387] [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: 09/02/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022] Open
Abstract
According to the World Health Organization, the neoplasm is one of the main reasons for morbidity and mortality worldwide. At the same time, application of cytostatic drugs like an independent type of cancer treatment and in combination with surgical methods, is often associated with the development of cardiovascular complications both in the early and in the delayed period of treatment. Doxorubicin (DOX) is the most commonly used cytotoxic anthracycline antibiotic. DOX can cause both acute and delayed side effects. The problem is still not solved, as evidenced by the continued activity of researchers in terms of developing approaches for the prevention and treatment of cardiovascular complications. It is known, the heart muscle consists of cardiomyocytes connected by intercalated discs (ID), which ensure the structural, electrical, metabolic unity of the heart. Various defects in the ID proteins can lead to the development of cardiovascular diseases of various etiologies, including DOX-induced cardiomyopathy. The search for ways to influence the functioning of ID proteins of the cardiac muscle can become the basis for the creation of new therapeutic approaches to the treatment and prevention of cardiac pathologies. SIRT1 may be an interesting cardioprotective variant due to its wide functional significance. SIRT1 activation triggers nuclear transcription programs that increase the efficiency of cellular, mitochondrial metabolism, increases resistance to oxidative stress, and promotes cell survival. It can be assumed that SIRT1 can not only provide a protective effect at the cardiomyocytes level, leading to an improvement in mitochondrial and metabolic functions, reducing the effects of oxidative stress and inflammatory processes, but also have a protective effect on the functioning of IDs structures of the cardiac muscle.
Collapse
|
32
|
The Role of Mitochondrial Quality Control in Anthracycline-Induced Cardiotoxicity: From Bench to Bedside. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3659278. [PMID: 36187332 PMCID: PMC9519345 DOI: 10.1155/2022/3659278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 09/06/2022] [Indexed: 11/18/2022]
Abstract
Cardiotoxicity is the major side effect of anthracyclines (doxorubicin, daunorubicin, epirubicin, and idarubicin), though being the most commonly used chemotherapy drugs and the mainstay of therapy in solid and hematological neoplasms. Advances in the field of cardio-oncology have expanded our understanding of the molecular mechanisms underlying anthracycline-induced cardiotoxicity (AIC). AIC has a complex pathogenesis that includes a variety of aspects such as oxidative stress, autophagy, and inflammation. Emerging evidence has strongly suggested that the loss of mitochondrial quality control (MQC) plays an important role in the progression of AIC. Mitochondria are vital organelles in the cardiomyocytes that serve as the key regulators of reactive oxygen species (ROS) production, energy metabolism, cell death, and calcium buffering. However, as mitochondria are susceptible to damage, the MQC system, including mitochondrial dynamics (fusion/fission), mitophagy, mitochondrial biogenesis, and mitochondrial protein quality control, appears to be crucial in maintaining mitochondrial homeostasis. In this review, we summarize current evidence on the role of MQC in the pathogenesis of AIC and highlight the therapeutic potential of restoring the cardiomyocyte MQC system in the prevention and intervention of AIC.
Collapse
|
33
|
Liang Z, He Y, Hu X. Cardio-Oncology: Mechanisms, Drug Combinations, and Reverse Cardio-Oncology. Int J Mol Sci 2022; 23:10617. [PMID: 36142538 PMCID: PMC9501315 DOI: 10.3390/ijms231810617] [Citation(s) in RCA: 16] [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: 08/24/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 11/16/2022] Open
Abstract
Chemotherapy, radiotherapy, targeted therapy, and immunotherapy have brought hope to cancer patients. With the prolongation of survival of cancer patients and increased clinical experience, cancer-therapy-induced cardiovascular toxicity has attracted attention. The adverse effects of cancer therapy that can lead to life-threatening or induce long-term morbidity require rational approaches to prevention and treatment, which requires deeper understanding of the molecular biology underpinning the disease. In addition to the drugs used widely for cardio-protection, traditional Chinese medicine (TCM) formulations are also efficacious and can be expected to achieve "personalized treatment" from multiple perspectives. Moreover, the increased prevalence of cancer in patients with cardiovascular disease has spurred the development of "reverse cardio-oncology", which underscores the urgency of collaboration between cardiologists and oncologists. This review summarizes the mechanisms by which cancer therapy induces cardiovascular toxicity, the combination of antineoplastic and cardioprotective drugs, and recent advances in reverse cardio-oncology.
Collapse
Affiliation(s)
| | | | - Xin Hu
- China–Japan Union Hospital of Jilin University, Jilin University, Changchun 130033, China
| |
Collapse
|
34
|
Current Status and Trends of Research on Anthracycline-Induced Cardiotoxicity from 2002 to 2021: A Twenty-Year Bibliometric and Visualization Analysis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6260243. [PMID: 35993025 PMCID: PMC9388240 DOI: 10.1155/2022/6260243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/21/2022] [Accepted: 07/24/2022] [Indexed: 12/30/2022]
Abstract
Anthracyclines constitute the cornerstone of numerous chemotherapy regimens for various cancers. However, the clinical application of anthracyclines is significantly limited to their dose-dependent cardiotoxicity. A comprehensive understanding of the current status of anthracycline-induced cardiotoxicity is necessary for in-depth research and optimal clinical protocols. Bibliometric analysis is widely applied in depicting development trends and tracking frontiers of a specific field. The present study is aimed at revealing the status and trends of anthracycline-induced cardiotoxicity during the past two decades by employing bibliometric software including R-bibliometric, VOSviewer, and CiteSpace. A total of 3504 publications concerning anthracycline-induced cardiotoxicity from 2002 to 2021 were collected from the Web of Science Core Collection database. Results showed significant growth in annual yields from 90 records in 2002 to 304 papers in 2021. The United States was the most productive country with the strongest collaboration worldwide in the field. Charles University in the Czech Republic was the institution that contributed the most papers, while 7 of the top 10 productive institutions were from the United States. The United States Department of Health and Human Services and the National Institutes of Health are the two agencies that provide financial support for more than 50% of sponsored publications. The research categories of included publications mainly belong to Oncology and Cardiac Cardiovascular Systems. The Journal of Clinical Oncology had a comprehensive impact on this research field with the highest IF value and many publications. Simunek Tomas from Charles University contributed the most publications, while Lipshultz Steven E. from the State University of New York possessed the highest H-index. In addition, the future research frontiers of anthracycline-induced cardiotoxicity might include early detection, pharmacogenomics, molecular mechanism, and cardiooncology. The present bibliometric analysis may provide a valuable reference for researchers and practitioners in future research directions.
Collapse
|
35
|
Genetic Susceptibility and Mechanisms Underlying the Pathogenesis of Anthracycline-Associated Cardiotoxicity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5818612. [PMID: 35965684 PMCID: PMC9365594 DOI: 10.1155/2022/5818612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/18/2022] [Accepted: 07/22/2022] [Indexed: 11/18/2022]
Abstract
Anthracyclines are chemotherapeutic agents widely used to treat a variety of cancers, and these drugs have revolutionized our management of cancer patients. The dose-dependent cardiotoxicity of anthracyclines, however, remains one of the leading causes of chemotherapy treatment-associated mortality in cancer survivors. Patient threshold doses leading to anthracycline-induced cardiotoxicity (AIC) are highly variable among affected patients. This variability is largely ascribed to genetic variants in individuals' genomes. Here, we briefly discuss the prevailing mechanisms underlying the pathogenesis of AIC, and then, we review the genetic variants, mostly identified through human genetic approaches and identified in cancer survivors. The identification of all genetic susceptibilities and elucidation of underlying mechanisms of AIC can help improve upfront risk prediction assessment for potentially severe cardiotoxicity disease and provide valuable insights into the understanding of AIC pathophysiology, which can be further leveraged to develop targeted pharmacogenetic therapies for those at high risk.
Collapse
|
36
|
Abstract
The quest of defeating cancer and improving prognosis in survivors has generated remarkable strides forward in research and have advanced the development of new antineoplastic therapies. These achievements, combined with rapid screening and early detection, have considerably extended the life expectancy of patients surviving multiple types of malignancies. Consequently, chemotherapy-related toxicity in several organ systems, especially the cardiovascular system, has surfaced as one of the leading causes of morbidity and mortality among cancer survivors. Recent evidence classifies chemotherapy-induced cardiotoxicity as the second-leading cause of morbidity and mortality, closely comparing with secondary cancer malignancies. While a certain degree of cardiotoxicity has been reported to accompany most chemotherapies, including anthracyclines, anti-metabolites, and alkylating agents, even the latest targeted cancer therapies such as immune checkpoint inhibitors and tyrosine kinase inhibitors have been associated with acute and chronic cardiac sequelae. In this chapter, we focus on describing the principal mechanism(s) for each class of chemotherapeutic agents that lead to cardiotoxicity and the innovative translational research approaches that are currently being explored to prevent or treat cancer therapy-induced cardiotoxicity and related cardiac complications.
Collapse
Affiliation(s)
- Adolfo G Mauro
- Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Pauley Heart Center, Richmond, VA, United States
| | - Katherine Hunter
- Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Pauley Heart Center, Richmond, VA, United States
| | - Fadi N Salloum
- Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Pauley Heart Center, Richmond, VA, United States.
| |
Collapse
|
37
|
Zhang M, Yang H, Xu C, Jin F, Zheng A. Risk Factors for Anthracycline-Induced Cardiotoxicity in Breast Cancer Treatment: A Meta-Analysis. Front Oncol 2022; 12:899782. [PMID: 35785172 PMCID: PMC9248259 DOI: 10.3389/fonc.2022.899782] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 05/11/2022] [Indexed: 01/05/2023] Open
Abstract
Background Anthracyclines play an important role in the treatment of breast cancer (BC) and other malignant tumors. However, accompanied side-effects are non-ignorable. The purpose of this meta-analysis is to determine the risk factors for anthracycline-induced cardiotoxicity (ACT), so as to identify high-risk patients. Methods The search for literature was conducted in PubMed, The Cochrane Library, Embase and Web of science. Records were selected with inclusion criteria and exclusion criteria. The newcastle-ottawa scale (NOS) was used to assess the quality of literature, and Review Manager 5.3 software was used for meta-analysis. Results Thirteen studies met the inclusion criteria. Meta-analysis indicated that risk factors for ACT were use of trastuzumab (odds ratio [OR]: 2.84, 95% confidence interval [CI]: 2.49-3.22, p < 0.00001), cumulative dose of anthracyclines (OR: 1.45, 95%CI: 1.28-1.65, p < 0.00001), hypertension (OR: 2.95, 95%CI: 1.75-4.97, p < 0.0001), diabetes mellitus (DM) (OR: 1.39, 95%CI: 1.20-1.61, p < 0.0001), tumor metastasis (OR: 1.91, 95%CI: 1.17-3.11, p = 0.009) and coronary heart disease (CAD) (OR: 2.17, 95%CI: 1.50-3.15, p < 0.0001). In addition, our analysis revealed that body mass index (BMI) had no effect on ACT (OR: 1.18, 95%CI: 0.98-1.43, p = 0.08). Conclusions Patients with high risk for ACT can be identified by these factors. For such patients, a higher level of monitoring and protection for the cardiac function should be performed by clinicians. Systematic Review Registration INPLASY, identifier INPLASY202250140.
Collapse
Affiliation(s)
- Meilin Zhang
- Department of Burn Plastic Surgery, Chaoyang Central Hospital, Chaoyang, China
| | - Hongguang Yang
- Department of Burn Plastic Surgery, Chaoyang Central Hospital, Chaoyang, China
| | - Changcun Xu
- Department of Cardiology, Chaoyang Central Hospital, Chaoyang, China
| | - Feng Jin
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
- *Correspondence: Feng Jin, ; Ang Zheng,
| | - Ang Zheng
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
- *Correspondence: Feng Jin, ; Ang Zheng,
| |
Collapse
|
38
|
Bergler-Klein J, Rainer PP, Wallner M, Zaruba MM, Dörler J, Böhmer A, Buchacher T, Frey M, Adlbrecht C, Bartsch R, Gyöngyösi M, Fürst UM. Cardio-oncology in Austria: cardiotoxicity and surveillance of anti-cancer therapies : Position paper of the Heart Failure Working Group of the Austrian Society of Cardiology. Wien Klin Wochenschr 2022; 134:654-674. [PMID: 35507087 PMCID: PMC9065248 DOI: 10.1007/s00508-022-02031-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/18/2022] [Indexed: 02/07/2023]
Abstract
Survival in cancer is continuously improving due to evolving oncological treatment. Therefore, cardiovascular short-term and long-term side effects gain crucial importance for overall outcome. Cardiotoxicity not only presents as heart failure, but also as treatment-resistant hypertension, acute coronary ischemia with plaque rupture or vasospasm, thromboembolism, arrhythmia, pulmonary hypertension, diastolic dysfunction, acute myocarditis and others. Recent recommendations have proposed baseline cardiac risk assessment and surveillance strategies. Major challenges are the availability of monitoring and imaging resources, including echocardiography with speckle tracking longitudinal strain (GLS), serum biomarkers such as natriuretic peptides (NT-proBNP) and highly sensitive cardiac troponins. This Austrian consensus encompasses cardiotoxicity occurrence in frequent antiproliferative cancer drugs, radiotherapy, immune checkpoint inhibitors and cardiac follow-up considerations in cancer survivors in the context of the Austrian healthcare setting. It is important to optimize cardiovascular risk factors and pre-existing cardiac diseases without delaying oncological treatment. If left ventricular ejection fraction (LVEF) deteriorates during cancer treatment (from >10% to <50%), or myocardial strain decreases (>15% change in GLS), early initiation of cardioprotective therapies (angiotensin-converting enzyme inhibitors, angiotensin or beta receptor blockers) is recommended, and LVEF should be reassessed before discontinuation. Lower LVEF cut-offs were recently shown to be feasible in breast cancer patients to enable optimal anticancer treatment. Interdisciplinary cardio-oncology cooperation is pivotal for optimal management of cancer patients.
Collapse
Affiliation(s)
- Jutta Bergler-Klein
- Department of Cardiology, University Clinic of Internal Medicine II, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
| | - Peter P Rainer
- Division of Cardiology, Medical University of Graz, Graz, Austria.,BioTechMed Graz, Graz, Austria
| | - Markus Wallner
- Division of Cardiology, Medical University of Graz, Graz, Austria.,Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Marc-Michael Zaruba
- Department of Internal Medicine III, Cardiology and Angiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Jakob Dörler
- Department of Internal Medicine III, Cardiology and Angiology, Medical University of Innsbruck, Innsbruck, Austria.,Department of Internal Medicine and Cardiology, Klinikum Klagenfurt, Klagenfurt, Austria
| | - Armin Böhmer
- Department of Internal Medicine 1, Krems University Clinic, Krems, Austria
| | - Tamara Buchacher
- Department of Internal Medicine and Cardiology, Klinikum Klagenfurt, Klagenfurt, Austria
| | - Maria Frey
- Department of Cardiology, University Clinic of Internal Medicine II, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | | | - Rupert Bartsch
- Department of Medicine 1, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Mariann Gyöngyösi
- Department of Cardiology, University Clinic of Internal Medicine II, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Ursula-Maria Fürst
- Department of Internal Medicine, Hospital of the Brothers of St. John of God (Krankenhaus Barmherzige Brüder) Salzburg, Salzburg, Austria
| |
Collapse
|
39
|
Jiang J, Liu B, Hothi SS. Herceptin-Mediated Cardiotoxicity: Assessment by Cardiovascular Magnetic Resonance. Cardiol Res Pract 2022; 2022:1910841. [PMID: 35265371 PMCID: PMC8898877 DOI: 10.1155/2022/1910841] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 10/12/2021] [Accepted: 01/20/2022] [Indexed: 02/07/2023] Open
Abstract
Herceptin (trastuzumab) is a recombinant, humanized, monoclonal antibody that targets the human epidermal growth factor receptor 2 (HER2) and is used in the treatment of HER2-positive breast and gastric cancers. However, it carries a risk of cardiotoxicity, manifesting as left ventricular (LV) systolic dysfunction, conventionally assessed for by transthoracic echocardiography. Clinical surveillance of cardiac function and discontinuation of trastuzumab at an early stage of LV systolic dysfunction allow for the timely initiation of heart failure drug therapies that can result in the rapid recovery of cardiac function in most patients. Often considered the reference standard for the noninvasive assessment of cardiac volume and function, cardiac magnetic resonance (CMR) imaging has superior reproducibility and accuracy compared to other noninvasive imaging modalities. However, due to limited availability, it is not routinely used in the serial assessment of cardiac function in patients receiving trastuzumab. In this article, we review the diagnostic and prognostic role of CMR in trastuzumab-mediated cardiotoxicity.
Collapse
Affiliation(s)
- Jin Jiang
- Heart and Lung Centre, New Cross Hospital, Wolverhampton, UK
| | - Boyang Liu
- Heart and Lung Centre, New Cross Hospital, Wolverhampton, UK
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Sandeep S Hothi
- Heart and Lung Centre, New Cross Hospital, Wolverhampton, UK
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
40
|
Wu BB, Leung KT, Poon ENY. Mitochondrial-Targeted Therapy for Doxorubicin-Induced Cardiotoxicity. Int J Mol Sci 2022; 23:1912. [PMID: 35163838 PMCID: PMC8837080 DOI: 10.3390/ijms23031912] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/27/2022] [Accepted: 02/01/2022] [Indexed: 01/27/2023] Open
Abstract
Anthracyclines, such as doxorubicin, are effective chemotherapeutic agents for the treatment of cancer, but their clinical use is associated with severe and potentially life-threatening cardiotoxicity. Despite decades of research, treatment options remain limited. The mitochondria is commonly considered to be the main target of doxorubicin and mitochondrial dysfunction is the hallmark of doxorubicin-induced cardiotoxicity. Here, we review the pathogenic mechanisms of doxorubicin-induced cardiotoxicity and present an update on cardioprotective strategies for this disorder. Specifically, we focus on strategies that can protect the mitochondria and cover different therapeutic modalities encompassing small molecules, post-transcriptional regulators, and mitochondrial transfer. We also discuss the shortcomings of existing models of doxorubicin-induced cardiotoxicity and explore advances in the use of human pluripotent stem cell derived cardiomyocytes as a platform to facilitate the identification of novel treatments against this disorder.
Collapse
Affiliation(s)
- Bin Bin Wu
- Centre for Cardiovascular Genomics and Medicine, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China;
- Hong Kong Hub of Paediatric Excellence (HK HOPE), The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China;
| | - Kam Tong Leung
- Hong Kong Hub of Paediatric Excellence (HK HOPE), The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China;
- Department of Paediatrics, The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
| | - Ellen Ngar-Yun Poon
- Centre for Cardiovascular Genomics and Medicine, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China;
- Hong Kong Hub of Paediatric Excellence (HK HOPE), The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China;
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
- School of Biomedical Sciences, The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
| |
Collapse
|
41
|
Kitakata H, Endo J, Ikura H, Moriyama H, Shirakawa K, Katsumata Y, Sano M. Therapeutic Targets for DOX-Induced Cardiomyopathy: Role of Apoptosis vs. Ferroptosis. Int J Mol Sci 2022; 23:1414. [PMID: 35163335 PMCID: PMC8835899 DOI: 10.3390/ijms23031414] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/20/2022] [Accepted: 01/24/2022] [Indexed: 01/04/2023] Open
Abstract
Doxorubicin (DOX) is the most widely used anthracycline anticancer agent; however, its cardiotoxicity limits its clinical efficacy. Numerous studies have elucidated the mechanisms underlying DOX-induced cardiotoxicity, wherein apoptosis has been reported as the most common final step leading to cardiomyocyte death. However, in the past two years, the involvement of ferroptosis, a novel programmed cell death, has been proposed. The purpose of this review is to summarize the historical background that led to each form of cell death, focusing on DOX-induced cardiotoxicity and the molecular mechanisms that trigger each form of cell death. Furthermore, based on this understanding, possible therapeutic strategies to prevent DOX cardiotoxicity are outlined. DNA damage, oxidative stress, intracellular signaling, transcription factors, epigenetic regulators, autophagy, and metabolic inflammation are important factors in the molecular mechanisms of DOX-induced cardiomyocyte apoptosis. Conversely, the accumulation of lipid peroxides, iron ion accumulation, and decreased expression of glutathione and glutathione peroxidase 4 are important in ferroptosis. In both cascades, the mitochondria are an important site of DOX cardiotoxicity. The last part of this review focuses on the significance of the disruption of mitochondrial homeostasis in DOX cardiotoxicity.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Motoaki Sano
- Department of Cardiology, Keio University School of Medicine, Tokyo 160-8582, Japan; (H.K.); (J.E.); (H.I.); (H.M.); (K.S.); (Y.K.)
| |
Collapse
|
42
|
Lau RP, Fishbein MC. Toxins and the heart. Cardiovasc Pathol 2022. [DOI: 10.1016/b978-0-12-822224-9.00012-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
43
|
Bansal N, Joshi C, Adams MJ, Hutchins K, Ray A, Lipshultz SE. Cardiotoxicity in pediatric lymphoma survivors. Expert Rev Cardiovasc Ther 2021; 19:957-974. [PMID: 34958622 DOI: 10.1080/14779072.2021.2013811] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Over the past five decades, the diagnosis and management of children with various malignancies have improved tremendously. As a result, an increasing number of children are long-term cancer survivors. With improved survival, however, has come an increased risk of treatment-related cardiovascular complications that can appear decades later. AREAS COVERED This review discusses the pathophysiology, epidemiology and effects of treatment-related cardiovascular complications from anthracyclines and radiotherapy in pediatric lymphoma survivors. There is a paucity of evidence-based recommendations for screening for and treatment of cancer therapy-induced cardiovascular complications. We discuss current preventive measures and strategies for their treatment. EXPERT OPINION Significant cardiac adverse effects occur due to radiation and chemotherapy received by patients treated for lymphoma. Higher lifetime cumulative doses, female sex, longer follow-up, younger age, and preexisting cardiovascular disease are associated with a higher incidence of cardiotoxicity. With deeper understanding of the mechanisms of these adverse cardiac effects and identification of driver mutations causing these effects, personalized cancer therapy to limit cardiotoxic effects while ensuring an adequate anti-neoplastic effect would be ideal. In the meantime, expanding the use of cardioprotective agents with the best evidence such as dexrazoxane should be encouraged and further studied.
Collapse
Affiliation(s)
- Neha Bansal
- Division of Pediatric Cardiology, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx NY, USA
| | - Chaitya Joshi
- Department of Pediatrics, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, Buffalo NY, USA
| | - Michael Jacob Adams
- Department of Public Health Sciences, University of Rochester, Rochester NY, USA
| | - Kelley Hutchins
- John A. Burns School of Medicine, Pediatric Hematology/Oncology, Kapiolani Medical Center for Women and Children, Honolulu HI, USA
| | - Andrew Ray
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo NY, USA
| | - Steven E Lipshultz
- Department of Pediatrics, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, Buffalo NY, USA.,Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo NY, USA.,Pediatrics Department, John R. Oishei Children's Hospital, UBMD Pediatrics Practice Group, Buffalo NY, USA
| |
Collapse
|
44
|
From dissection of fibrotic pathways to assessment of drug interactions to reduce cardiac fibrosis and heart failure. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2021; 2:100036. [PMID: 34909666 PMCID: PMC8663973 DOI: 10.1016/j.crphar.2021.100036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/14/2021] [Accepted: 05/18/2021] [Indexed: 12/14/2022] Open
Abstract
Cardiac fibrosis is characterized by extracellular matrix deposition in the cardiac interstitium, and this contributes to cardiac contractile dysfunction and progression of heart failure. The main players involved in this process are the cardiac fibroblasts, which, in the presence of pro-inflammatory/pro-fibrotic stimuli, undergo a complete transformation acquiring a more proliferative, a pro-inflammatory and a secretory phenotype. This review discusses the cellular effectors and molecular pathways implicated in the pathogenesis of cardiac fibrosis and suggests potential strategies to monitor the effects of specific drugs designed to slow down the progression of this disease by specifically targeting the fibroblasts.
Collapse
|
45
|
Li ZY, Xu GS, Song YL, Li X. Structural optimizations and bioevaluation of N-substituted acridone derivatives as strong topoisomerase II inhibitors. Bioorg Chem 2021; 119:105543. [PMID: 34929515 DOI: 10.1016/j.bioorg.2021.105543] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 10/28/2021] [Accepted: 12/02/2021] [Indexed: 12/20/2022]
Abstract
Previously, an array of N-substituted acridone derivatives have been reported as potent topoisomerase II (topo II) inhibitors, and preliminary structure-activity relationship (SAR) outcomes revealed that the linker between 1-NH and N-methyl piperazine motif of the tricyclic acridone scaffold significantly affected their anti-proliferative potencies. To further explore the SARs of acridone-derived topo II inhibitors, a wider range of novel acridone derivatives were herein synthesized via two rounds of structural optimizations on two validated hits, E17 and E24. Initially, the linker length was optimized, and then influences of N-methyl piperazinyl moiety and disposition of three N atoms on the bioactivity were investigated. As a result, a newly developed topo II inhibitor 6 h was found to be more potent than E17 and E24, thereby serving as a tool compound for the follow-up mechanistic study. Compound 6 h functioned as a strong topo IIα/β inhibitor, caused obvious DNA damage, and induced apoptosis by triggering the loss of mitochondrial membrane potential (Δψm). Further molecular docking and MD study illustrated the favorable interactions of 6 h with both topo IIα and topo IIβ subtypes.
Collapse
Affiliation(s)
- Zhi-Ying Li
- Key Laboratory of Chemistry and Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong 250012, China
| | - Guang-Sen Xu
- Key Laboratory of Chemistry and Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong 250012, China
| | - Yu-Liang Song
- Key Laboratory of Chemistry and Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong 250012, China
| | - Xun Li
- Institute of Materia Medica, Shandong Provincial Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong 250117, China; Key Laboratory of Forensic Toxicology, Ministry of Public Security, Beijing 100192, China.
| |
Collapse
|
46
|
From cardio-oncology to cardio-onco-pharmacology: Towards a multidisciplinary approach in the understanding and management of cardiotoxicity. Therapie 2021; 77:197-206. [PMID: 34895759 DOI: 10.1016/j.therap.2021.09.008] [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] [Received: 08/24/2021] [Accepted: 09/22/2021] [Indexed: 10/19/2022]
Abstract
Cardio-oncology is an emerging field that transformed the medical management of patients with cancer. It encompasses the prevention and treatment of cardiovascular toxicities related to cancer treatments, aiming to reduce cardiac adverse events among cancer survivors. Cardiovascular toxicities related to cancer treatments are described through data collected during phase I to phase III therapeutic trials, and post-marketing surveillance (phase IV). Pharmacovigilance analyses, based on datamining from these extensive databases, allowed to understanding and identifying new adverse drug reactions, some recently made available, such as immunotherapy or inhibitor of Bruton tyrosine kinase (IBTK).
Collapse
|
47
|
Chen Z, Yan Y, Qi C, Liu J, Li L, Wang J. The Role of Ferroptosis in Cardiovascular Disease and Its Therapeutic Significance. Front Cardiovasc Med 2021; 8:733229. [PMID: 34765653 PMCID: PMC8576275 DOI: 10.3389/fcvm.2021.733229] [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: 07/02/2021] [Accepted: 09/17/2021] [Indexed: 12/29/2022] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of deaths worldwide with regulated cell death playing an important role in cardiac pathophysiology. However, the classical mode of cell death cannot fully explain the occurrence and development of heart disease. In recent years, much research has been performed on ferroptosis, a new type of cell death that causes cell damage and contributes to the development of atherosclerosis, myocardial infarction, heart failure, and other diseases. In this review, we discuss the role of different organelles in ferroptosis and also focus on the relationship between autophagy and ferroptosis. Additionally, we describe the specific mechanism by which ferroptosis contributes to the development of CVD. Finally, we summarize the current research on ferroptosis-related pathway inhibitors and the applications of clinically beneficial cardiovascular drugs.
Collapse
Affiliation(s)
- Zhenzhen Chen
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| | - Youyou Yan
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| | - Chao Qi
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| | - Jia Liu
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| | - Longbo Li
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| | - Junnan Wang
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
48
|
Li ZY, Xu GS, Li X. A Unique Topoisomerase II Inhibitor with Dose-Affected Anticancer Mechanisms and Less Cardiotoxicity. Cells 2021; 10:cells10113138. [PMID: 34831359 PMCID: PMC8621772 DOI: 10.3390/cells10113138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 11/16/2022] Open
Abstract
Type II DNA topoisomerase (topo II) is an essential nuclear enzyme and a well-validated anticancer drug target. Previously, we have carried out several rounds of structural optimizations on our in-house topo II inhibitor E17, which was shown to have superior anticancer activity and less risk of multidrug resistance (MDR). Among the newly developed acridone derivatives, 6h displayed significant anticancer efficacy with unique mechanisms of action. At low concentrations, it arrested cancer cell cycles and triggered cell apoptosis, which is similar to the action of the well-known topo II inhibitor VP16. By contrast, 6h showed significant and long-term anti-proliferative activity at relatively high concentrations, with negligible influence on apoptosis. In addition, 6h exhibited no serious cardiotoxicity compared to doxorubicin (DOXO), a widely used topo II-targeting antineoplastic drug in clinic, but with damaging myocardial side effects. Collectively, our present work has supported the therapeutic value of 6h as a promising chemotherapy for cancers.
Collapse
Affiliation(s)
- Zhi-Ying Li
- Key Laboratory of Chemistry and Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; (Z.-Y.L.); (G.-S.X.)
| | - Guang-Sen Xu
- Key Laboratory of Chemistry and Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; (Z.-Y.L.); (G.-S.X.)
| | - Xun Li
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
- Key Laboratory of Forensic Toxicology, Ministry of Public Security, Beijing 100192, China
- Correspondence:
| |
Collapse
|
49
|
Melo MDTD, Paiva MG, Santos MVC, Rochitte CE, Moreira VDM, Saleh MH, Brandão SCS, Gallafrio CC, Goldwasser D, Gripp EDA, Piveta RB, Silva TO, Santo THCE, Ferreira WP, Salemi VMC, Cauduro SA, Barberato SH, Lopes HMC, Pena JLB, Rached HRS, Miglioranza MH, Pinheiro AC, Vrandecic BALM, Cruz CBBV, Nomura CH, Cerbino FME, Costa IBSDS, Coelho Filho OR, Carneiro ACDC, Burgos UMMC, Fernandes JL, Uellendahl M, Calado EB, Senra T, Assunção BL, Freire CMV, Martins CN, Sawamura KSS, Brito MM, Jardim MFS, Bernardes RJM, Diógenes TC, Vieira LDO, Mesquita CT, Lopes RW, Segundo Neto EMV, Rigo L, Marin VLS, Santos MJ, Grossman GB, Quagliato PC, Alcantara MLD, Teodoro JAR, Albricker ACL, Barros FS, Amaral SID, Porto CLL, Barros MVL, Santos SND, Cantisano AL, Petisco ACGP, Barbosa JEM, Veloso OCG, Spina S, Pignatelli R, Hajjar LA, Kalil Filho R, Lopes MACQ, Vieira MLC, Almeida ALC. Brazilian Position Statement on the Use Of Multimodality Imaging in Cardio-Oncology - 2021. Arq Bras Cardiol 2021; 117:845-909. [PMID: 34709307 PMCID: PMC8528353 DOI: 10.36660/abc.20200266] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Affiliation(s)
| | | | | | - Carlos Eduardo Rochitte
- Instituto do Coração (Incor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
- Hospital do Coração (HCOR), São Paulo, SP - Brasil
| | | | - Mohamed Hassan Saleh
- Instituto do Coração (Incor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
- Instituto Dante Pazzanese de Cardiologia, São Paulo, SP - Brasil
| | | | | | - Daniel Goldwasser
- Hospital Federal de Ipanema, Rio de Janeiro, RJ - Brasil
- Hospital Copa D'Or, Rio de Janeiro, RJ - Brasil
- Casa de Saúde São José, Rio de Janeiro, RJ - Brasil
| | - Eliza de Almeida Gripp
- Hospital Pró-Cardíaco, Rio de Janeiro, RJ - Brasil
- Hospital Universitário Antônio Pedro, Rio de Janeiro, RJ - Brasil
| | | | - Tonnison Oliveira Silva
- Hospital Cardio Pulmonar - Centro de Estudos em Cardiologia, Salvador, BA - Brasil
- Escola Bahiana de Medicina e Saúde Pública, Salvador, BA - Brasil
| | | | | | - Vera Maria Cury Salemi
- Instituto do Coração (Incor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
| | | | - Silvio Henrique Barberato
- CardioEco Centro de Diagnóstico Cardiovascular, Curitiba, PR - Brasil
- Quanta Diagnóstico, Curitiba, PR - Brasil
| | | | | | | | - Marcelo Haertel Miglioranza
- Instituto de Cardiologia do Rio Grande do Sul - Laboratório de Pesquisa e Inovação em Imagem Cardiovascular, Porto Alegre, RS - Brasil
- Hospital Mãe de Deus, Porto Alegre, RS - Brasil
| | | | | | | | - César Higa Nomura
- Instituto do Coração (Incor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
- Hospital Sírio-Libanês, São Paulo, SP - Brasil
| | - Fernanda Mello Erthal Cerbino
- Clínica de Diagnóstico por Imagem, Rio de Janeiro, RJ - Brasil
- Diagnósticos da América AS, Rio de Janeiro, RJ - Brasil
| | | | | | | | | | - Juliano Lara Fernandes
- Radiologia Clínica de Campinas, Campinas, SP - Brasil
- Instituto de Ensino e Pesquisa José Michel Kalaf, Campinas, SP - Brasil
| | - Marly Uellendahl
- Diagnósticos da América AS, Rio de Janeiro, RJ - Brasil
- Universidade Federal de São Paulo (UNIFESP), São Paulo, SP - Brasil
| | | | - Tiago Senra
- Instituto Dante Pazzanese de Cardiologia, São Paulo, SP - Brasil
- Hospital Sírio-Libanês, São Paulo, SP - Brasil
| | - Bruna Leal Assunção
- Universidade de São Paulo Instituto do Câncer do Estado de São Paulo, São Paulo, SP - Brasil
| | - Claudia Maria Vilas Freire
- Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG - Brasil
- ECOCENTER, Belo Horizonte, MG - Brasil
| | | | - Karen Saori Shiraishi Sawamura
- Hospital do Coração (HCOR), São Paulo, SP - Brasil
- Hospital Universitário Antônio Pedro, Rio de Janeiro, RJ - Brasil
- Instituto da Criança da Universidade de São Paulo (USP), São Paulo, SP - Brasil
| | - Márcio Miranda Brito
- Universidade Federal do Tocantins - Campus de Araguaina, Araguaina, TO - Brasil
- Hospital Municipal de Araguaina, Araguaina, TO - Brasil
| | | | | | | | | | - Claudio Tinoco Mesquita
- Hospital Pró-Cardíaco, Rio de Janeiro, RJ - Brasil
- Universidade Federal Fluminense (UFF), Rio de Janeiro, RJ - Brasil
- Hospital Vitória, Rio de Janeiro, RJ - Brasil
| | | | | | - Letícia Rigo
- Hospital Beneficência Portuguesa, São Paulo, SP - Brasil
| | | | | | - Gabriel Blacher Grossman
- Clínica Cardionuclear, Porto Alegre, RS - Brasil
- Hospital Moinhos de Vento, Porto Alegre, RS - Brasil
| | | | - Monica Luiza de Alcantara
- Americas Medical City, Rio de Janeiro, Rio de Janeiro, RJ - Brasil
- Americas Serviços Médicos, Rio de Janeiro, RJ - Brasil
- Rede D'Or, Rio de Janeiro, RJ - Brasil
| | | | | | | | | | | | | | - Simone Nascimento Dos Santos
- Hospital Brasília - Ecocardiografia, Brasília, DF - Brasil
- Eccos Diagnóstico Cardiovascular Avançado, Brasília, DF - Brasil
| | | | | | | | | | | | - Ricardo Pignatelli
- Texas Children's Hospital, Houston, Texas - EUA
- Baylor College of Medicine, Houston, Texas - EUA
| | - Ludhmilla Abrahão Hajjar
- Instituto do Coração (Incor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
- Universidade de São Paulo Instituto do Câncer do Estado de São Paulo, São Paulo, SP - Brasil
| | - Roberto Kalil Filho
- Instituto do Coração (Incor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
- Universidade de São Paulo Instituto do Câncer do Estado de São Paulo, São Paulo, SP - Brasil
| | - Marcelo Antônio Cartaxo Queiroga Lopes
- Hospital Alberto Urquiza Wanderley - Hemodinâmica e Cardiologia Intervencionista, João Pessoa, PB - Brasil
- Hospital Metropolitano Dom José Maria Pires, João Pessoa, PB - Brasil
- Sociedade Brasileira de Cardiologia, Rio de Janeiro, RJ - Brasil
| | - Marcelo Luiz Campos Vieira
- Instituto do Coração (Incor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP - Brasil
- Hospital Israelita Albert Einstein, São Paulo, SP - Brasil
| | - André Luiz Cerqueira Almeida
- Santa Casa de Misericórdia de Feira de Santana - Cardiologia, Feira de Santana, BA - Brasil
- Departamento de Imagem Cardiovascular da Sociedade Brasileira de Cardiologia, São Paulo, SP - Brasil
| |
Collapse
|
50
|
Jirkovský E, Jirkovská A, Bavlovič-Piskáčková H, Skalická V, Pokorná Z, Karabanovich G, Kollárová-Brázdová P, Kubeš J, Lenčová-Popelová O, Mazurová Y, Adamcová M, Lyon AR, Roh J, Šimůnek T, Štěrbová-Kovaříková P, Štěrba M. Clinically Translatable Prevention of Anthracycline Cardiotoxicity by Dexrazoxane Is Mediated by Topoisomerase II Beta and Not Metal Chelation. Circ Heart Fail 2021; 14:e008209. [PMID: 34551586 DOI: 10.1161/circheartfailure.120.008209] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Anthracycline-induced heart failure has been traditionally attributed to direct iron-catalyzed oxidative damage. Dexrazoxane (DEX)-the only drug approved for its prevention-has been believed to protect the heart via its iron-chelating metabolite ADR-925. However, direct evidence is lacking, and recently proposed TOP2B (topoisomerase II beta) hypothesis challenged the original concept. METHODS Pharmacokinetically guided study of the cardioprotective effects of clinically used DEX and its chelating metabolite ADR-925 (administered exogenously) was performed together with mechanistic experiments. The cardiotoxicity was induced by daunorubicin in neonatal ventricular cardiomyocytes in vitro and in a chronic rabbit model in vivo (n=50). RESULTS Intracellular concentrations of ADR-925 in neonatal ventricular cardiomyocytes and rabbit hearts after treatment with exogenous ADR-925 were similar or exceeded those observed after treatment with the parent DEX. However, ADR-925 did not protect neonatal ventricular cardiomyocytes against anthracycline toxicity, whereas DEX exhibited significant protective effects (10-100 µmol/L; P<0.001). Unlike DEX, ADR-925 also had no significant impact on daunorubicin-induced mortality, blood congestion, and biochemical and functional markers of cardiac dysfunction in vivo (eg, end point left ventricular fractional shortening was 32.3±14.7%, 33.5±4.8%, 42.7±1.0%, and 41.5±1.1% for the daunorubicin, ADR-925 [120 mg/kg]+daunorubicin, DEX [60 mg/kg]+daunorubicin, and control groups, respectively; P<0.05). DEX, but not ADR-925, inhibited and depleted TOP2B and prevented daunorubicin-induced genotoxic damage. TOP2B dependency of the cardioprotective effects was probed and supported by experiments with diastereomers of a new DEX derivative. CONCLUSIONS This study strongly supports a new mechanistic paradigm that attributes clinically effective cardioprotection against anthracycline cardiotoxicity to interactions with TOP2B but not metal chelation and protection against direct oxidative damage.
Collapse
Affiliation(s)
- Eduard Jirkovský
- Department of Pharmacology, Faculty of Medicine in Hradec Králové (E.J., Z.P., P.K.-B., O.L.-P., M.Š.), Charles University, Czech Republic.,Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové (E.J.), Charles University, Czech Republic
| | - Anna Jirkovská
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové (A.J., V.S., J.K., T.Š.), Charles University, Czech Republic
| | - Hana Bavlovič-Piskáčková
- Department of Pharmaceutical Chemistry and Pharmaceutical Analysis, Faculty of Pharmacy in Hradec Králové (H.B.-P., P.Š.-K.), Charles University, Czech Republic
| | - Veronika Skalická
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové (A.J., V.S., J.K., T.Š.), Charles University, Czech Republic
| | - Zuzana Pokorná
- Department of Pharmacology, Faculty of Medicine in Hradec Králové (E.J., Z.P., P.K.-B., O.L.-P., M.Š.), Charles University, Czech Republic
| | - Galina Karabanovich
- Department of Organic and Bioorganic Chemistry, Faculty of Pharmacy in Hradec Králové (G.K., J.R.), Charles University, Czech Republic
| | - Petra Kollárová-Brázdová
- Department of Pharmacology, Faculty of Medicine in Hradec Králové (E.J., Z.P., P.K.-B., O.L.-P., M.Š.), Charles University, Czech Republic
| | - Jan Kubeš
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové (A.J., V.S., J.K., T.Š.), Charles University, Czech Republic
| | - Olga Lenčová-Popelová
- Department of Pharmacology, Faculty of Medicine in Hradec Králové (E.J., Z.P., P.K.-B., O.L.-P., M.Š.), Charles University, Czech Republic
| | - Yvona Mazurová
- Department of Histology and Embryology, Faculty of Medicine in Hradec Králové (Y.M.), Charles University, Czech Republic
| | - Michaela Adamcová
- Department of Physiology, Faculty of Medicine in Hradec Králové (M.A.), Charles University, Czech Republic
| | - Alexander R Lyon
- Department of Cardiology, Royal Brompton Hospital and Faculty of Medicine, National Heart and Lung Institute, Imperial College London, United Kingdom (A.R.L.)
| | - Jaroslav Roh
- Department of Organic and Bioorganic Chemistry, Faculty of Pharmacy in Hradec Králové (G.K., J.R.), Charles University, Czech Republic
| | - Tomáš Šimůnek
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové (A.J., V.S., J.K., T.Š.), Charles University, Czech Republic
| | - Petra Štěrbová-Kovaříková
- Department of Pharmaceutical Chemistry and Pharmaceutical Analysis, Faculty of Pharmacy in Hradec Králové (H.B.-P., P.Š.-K.), Charles University, Czech Republic
| | - Martin Štěrba
- Department of Pharmacology, Faculty of Medicine in Hradec Králové (E.J., Z.P., P.K.-B., O.L.-P., M.Š.), Charles University, Czech Republic
| |
Collapse
|