1
|
Szponar J, Ciechanski E, Ciechanska M, Dudka J, Mandziuk S. Evolution of Theories on Doxorubicin-Induced Late Cardiotoxicity-Role of Topoisomerase. Int J Mol Sci 2024; 25:13567. [PMID: 39769331 PMCID: PMC11678604 DOI: 10.3390/ijms252413567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Doxorubicin (DOX) has been widely used as a cytotoxic chemotherapeutic. However, DOX has a number of side effects, such as myelotoxicity or gonadotoxicity, the most dangerous of which is cardiotoxicity. Cardiotoxicity can manifest as cardiac arrhythmias, myocarditis, and pericarditis; life-threatening late cardiotoxicity can result in heart failure months or years after the completion of chemotherapy. The development of late cardiomyopathy is not yet fully understood. The most important question is how DOX reprograms the cardiomyocyte, after which DOX is excreted from the body, initially without symptoms. However, clinically overt cardiomyopathy develops over the following months and years. Since the 1980s, DOX-induced disorders in cardiomyocytes have been thought to be related to oxidative stress and dependent on the Fe/reactive oxygen species (ROS) mechanism. That line of evidence was supported by dexrazoxane (DEX) protection, the only Food and Drug Administration (FDA)-approved drug for preventing DOX-induced cardiomyopathy, which complexes iron. Thus, the hypothesis related to Fe/ROS provides a plausible explanation for the induction of the development of late cardiomyopathy via DOX. However, in subsequent studies, DEX was used to identify another important mechanism in DOX-induced cardiomyopathy that is related to topoisomerase 2β (Top2β). Does the Top2β hypothesis explain the mechanisms of the development of DOX-dependent late heart failure? Several of these mechanisms have been identified to date, proving the involvement of Top2β in the regulation of the redox balance, including oxidative stress. Thus, the development of late cardiomyopathy can be explained based on mechanisms related to Top2β. In this review, we highlight free radical theory, iron imbalance, calcium overload, and finally, a theory based on Top2β.
Collapse
Affiliation(s)
- Jaroslaw Szponar
- Toxicology Clinic, Faculty of Medicine, Medical University of Lublin, Krasnicka 100, 20-718 Lublin, Poland;
- Clinical Department of Toxicology and Cardiology, Regional Specialist Hospital, Krasnicka 100, 20-718 Lublin, Poland
| | - Erwin Ciechanski
- Department of Cardiology, Regional Specialist Hospital, Krasnicka 100, 20-718 Lublin, Poland
| | - Magda Ciechanska
- Department of Pulmonary Diseases and Children Rheumatology, Medical University of Lublin, Antoniego Gebali 6, 20-093 Lublin, Poland
| | - Jaroslaw Dudka
- Department of Toxicology, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland;
| | - Sławomir Mandziuk
- Department of Pneumology, Oncology and Allergology, Medical University of Lublin, Jaczewskiego 8, 20-090 Lublin, Poland;
| |
Collapse
|
2
|
Iqbal MO, Gu Y, Khan IA, Wang R, Chen J. Evaluation of the in vitro antioxidant and antitumor activity of hydroalcoholic extract from Jatropha mollissima leaves in Wistar rats. Front Chem 2023; 11:1283618. [PMID: 38164252 PMCID: PMC10757942 DOI: 10.3389/fchem.2023.1283618] [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: 08/26/2023] [Accepted: 12/04/2023] [Indexed: 01/03/2024] Open
Abstract
Introduction: Despite modern sciences and advancements in new drugs or chemicals, the new era now rushes natural remedies for various illnesses and diseases that lead to end organ damage. In this study, we investigated Jatropha mollissima ethanolic extract's effect against doxorubicin-induced cardiotoxicity and renal toxicity. Methods: To determine phytochemicals, a phytochemical screening was conducted. Various assays were used to measure the antioxidant activity, including the DPPH (2,2-diphenylpicrylhydrazyl), SOD (superoxide dismutase), NO (nitric oxide), and others. The antiproliferative effect of Jm was assessed by MTT assay; morphological analysis was performed using an inverted and phase contrast microscope, ultra morphological analysis of apoptosis with acridine orange (AO)/propidium iodide (PI) staining. Results: It was seen that doxorubicin caused elevated serum markers and abnormal changes in histological patterns. The significant reduction in cardiac and renal marker levels seen in groups given either 400 or 600 mg/kg of crude extract demonstrates that Jm has a protective effect against doxorubicin-induced cardiotoxicity due to the presence of active phytoconstituents having antioxidant potential. There is a dose-dependent decrease in cell viability when using J. mollissima. Apoptosis was observed in the treated cells. Conclusion: In conclusion, our research lends credence to the idea that J. mollissima could be used for cancer management and have cardioprotective and nephroprotective effects.
Collapse
Affiliation(s)
- Muhammad Omer Iqbal
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao, China
- Key Laboratory of Marine Drugs, The Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- Fatima Tu Zahara Department of Life Sciences, Muhammad Institute of Medical and Allied Sciences, Multan, Pakistan
| | - Yuchao Gu
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Imran Ahmad Khan
- Fatima Tu Zahara Department of Life Sciences, Muhammad Institute of Medical and Allied Sciences, Multan, Pakistan
- Department of Pharmacy, MNS University of Agriculture, Multan, Pakistan
| | - Ruihong Wang
- The Affiliated Qingdao Central Hospital of Qingdao University, Qingdao, China
| | - Jin Chen
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao, China
| |
Collapse
|
3
|
Becker MMC, Arruda GFA, Berenguer DRF, Buril RO, Cardinale D, Brandão SCS. Anthracycline cardiotoxicity: current methods of diagnosis and possible role of 18F-FDG PET/CT as a new biomarker. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2023; 9:17. [PMID: 36973762 PMCID: PMC10041777 DOI: 10.1186/s40959-023-00161-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 02/01/2023] [Indexed: 03/29/2023]
Abstract
Despite advances in chemotherapy, the drugs used in cancer treatment remain rather harmful to the cardiovascular system, causing structural and functional cardiotoxic changes. Positron-emission tomography associated with computed tomography (PET/CT) has emerged like a promising technique in the early diagnosis of these adverse drug effects as the myocardial tissue uptake of fluorodeoxyglucose labeled with fluorine-18 (18F-FDG), a glucose analog, is increased after their use. Among these drugs, anthracyclines are the most frequently associated with cardiotoxicity because they promote heart damage through DNA breaks, and induction of an oxidative, proinflammatory, and toxic environment. This review aimed to present the scientific evidence available so far regarding the use of 18F-FDG PET/CT as an early biomarker of anthracycline-related cardiotoxicity. Thus, it discusses the physiological basis for its uptake, hypotheses to justify its increase in the myocardium affected by anthracyclines, importance of 18F-FDG PET/CT findings for cardio-oncology, and primary challenges of incorporating this technique in standard clinical oncology practice.
Collapse
Affiliation(s)
- Mônica M C Becker
- Postgraduate Program in Surgery, Federal University of Pernambuco, Recife, State of Pernambuco, Brazil
| | - Gustavo F A Arruda
- Recife Medical School, Federal University of Pernambuco, Recife, State of Pernambuco, Brazil
| | - Diego R F Berenguer
- Postgraduate Program in Translational Health, Federal University of Pernambuco, Recife, State of Pernambuco, Brazil
| | - Roberto O Buril
- Postgraduate Program in Surgery, Federal University of Pernambuco, Recife, State of Pernambuco, Brazil
| | - Daniela Cardinale
- Cardioncology Unit, European Institute of Oncology, I.R.C.C.S., Milan, Italy
| | - Simone C S Brandão
- Postgraduate Program in Surgery, Federal University of Pernambuco, Recife, State of Pernambuco, Brazil.
- Recife Medical School, Federal University of Pernambuco, Recife, State of Pernambuco, Brazil.
- Nuclear Medicine Department, Hospital das Clínicas, Federal University of Pernambuco, 1st floor, 1235 Avenida Professor Moraes Rego, Recife, State of Pernambuco, 50670-901, Brazil.
| |
Collapse
|
4
|
Moossavi M, Lu X, Herrmann J, Xu X. Molecular mechanisms of anthracycline induced cardiotoxicity: Zebrafish come into play. Front Cardiovasc Med 2023; 10:1080299. [PMID: 36970353 PMCID: PMC10036604 DOI: 10.3389/fcvm.2023.1080299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 02/22/2023] [Indexed: 03/12/2023] Open
Abstract
Anthracyclines are among the most potent chemotherapeutics; however, cardiotoxicity significantly restricts their use. Indeed, anthracycline-induced cardiotoxicity (AIC) fares among the worst types of cardiomyopathy, and may only slowly and partially respond to standard heart failure therapies including β-blockers and ACE inhibitors. No therapy specifically designed to treat anthracycline cardiomyopathy at present, and neither is it known if any such strategy could be developed. To address this gap and to elucidate the molecular basis of AIC with a therapeutic goal in mind, zebrafish has been introduced as an in vivo vertebrate model about a decade ago. Here, we first review our current understanding of the basic molecular and biochemical mechanisms of AIC, and then the contribution of zebrafish to the AIC field. We summarize the generation of embryonic zebrafish AIC models (eAIC) and their use for chemical screening and assessment of genetic modifiers, and then the generation of adult zebrafish AIC models (aAIC) and their use for discovering genetic modifiers via forward mutagenesis screening, deciphering spatial-temporal-specific mechanisms of modifier genes, and prioritizing therapeutic compounds via chemical genetic tools. Several therapeutic target genes and related therapies have emerged, including a retinoic acid (RA)-based therapy for the early phase of AIC and an autophagy-based therapy that, for the first time, is able to reverse cardiac dysfunction in the late phase of AIC. We conclude that zebrafish is becoming an important in vivo model that would accelerate both mechanistic studies and therapeutic development of AIC.
Collapse
Affiliation(s)
- Maryam Moossavi
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Xiaoguang Lu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Joerg Herrmann
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Xiaolei Xu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Correspondence: Xiaolei Xu
| |
Collapse
|
5
|
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
|
6
|
Kopeva KV, Grakova EV, Shilov SN, Berezikova EN, Popova AA, Neupokoeva MN, Ratushnyak ET, Teplyakov AT. Anthracycline-induced cardiotoxicity in women without cardiovascular diseases: molecular and genetic predictors. Acta Cardiol 2022; 77:805-814. [PMID: 34783301 DOI: 10.1080/00015385.2021.2003061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
OBJECTIVE To evaluate role of molecular (endothelin-1, soluble Fas-L, NT-proBNP, TNF-α, interleukin-1β,) and genetic factors (NOS3 (rs1799983), EDNRA (C + 70G, rs5335), NADPH oxidase (C242T, rs4673), p53 protein (polymorphic marker-Arg72Pro exon 4, rs1042522), NOS3 (Glu298Asp, rs1799983), Caspase 8 (CASP8, rs3834129 and rs1045485), interleukin-1β gene (Il-1β, rs1143634), TNF-α gene (rs1800629), SOD2 (rs4880), GPX1 (rs1050450) in development of anthracycline-induced cardiotoxicity (AIC) in women without cardiovascular diseases. METHODS A total of 176 women with breast cancer and without cardiovascular diseases who received anthracyclines were enrolled in the study. After the 12 months of chemotherapy (CT), all patients were divided into two groups: group 1 (n = 52) comprised patients with AIC, group 2 (n = 124) comprised those without it. RESULTS Based on ROC-analysis, levels of endothelin-1 of ≥9.0 pg/mL (AUC of 0.699), sFas-L of ≥98.3 ng/mL (AUC of 0.990), and NT-proBNP of ≥71.5 pg/mL (AUC of 0.994;) were identified as a cut-off values predicting AIC during 12 months after CT. Whereas, NT-proBNP and sFas-L were more significant predictors than endothelin-1 (p < 0.001). The development of AIC was significantly related to Arg/Arg of p53 protein gene (OR = 2.972; p = 0.001), T/T of NOS3 gene (OR = 3.059, p = 0.018), T/T of NADPH oxidase gene (OR = 2.753, p = 0.008), and C/C of GPX1 (OR = 2.345; p = 0.007). CONCLUSION Evaluation of polymorphisms genes of p53 (rs1042522), NOS3 (rs1799983), GPX1 (rs1050450), and NADPH oxidase (rs4673) can be recommended before CT for the risk assessment of AIC development. The serum levels of NT-proBNP and soluble Fas-L after CT may be considered as non-invasive biomarkers for prediction of AIC development during the 12 months.
Collapse
Affiliation(s)
- Kristina V Kopeva
- Department of Myocardial Pathology (Tomsk, Russian Federation), Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Elena V Grakova
- Department of Myocardial Pathology (Tomsk, Russian Federation), Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Sergey N Shilov
- Department of Pathological Physiology and Clinical Pathophysiology (Novosibirsk, Russian Federation), Novosibirsk State Medical University, Novosibirsk, Russia
| | - Ekaterina N Berezikova
- Department of Pathological Physiology and Clinical Pathophysiology (Novosibirsk, Russian Federation), Novosibirsk State Medical University, Novosibirsk, Russia
| | - Anna A Popova
- Department of Pathological Physiology and Clinical Pathophysiology (Novosibirsk, Russian Federation), Novosibirsk State Medical University, Novosibirsk, Russia
| | - Maria N Neupokoeva
- Department of Pathological Physiology and Clinical Pathophysiology (Novosibirsk, Russian Federation), Novosibirsk State Medical University, Novosibirsk, Russia
| | - Elena T Ratushnyak
- Department of Pathological Physiology and Clinical Pathophysiology (Novosibirsk, Russian Federation), Novosibirsk State Medical University, Novosibirsk, Russia
| | - Alexander T Teplyakov
- Department of Myocardial Pathology (Tomsk, Russian Federation), Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| |
Collapse
|
7
|
Antioxidative properties of Adansonia digitata L. (baobab) leaf extract exert protective effect on doxorubicin-induced cardiac toxicity in Wistar rats. CLINICAL NUTRITION OPEN SCIENCE 2022. [DOI: 10.1016/j.nutos.2022.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
8
|
Yu H, Qiu Y, Yu H, Wang Z, Xu J, Peng Y, Wan X, Wu X, Jin R, Zhou F. Anthracycline Induced Cardiac Disorders in Childhood Acute Lymphoblastic Leukemia: A Single-Centre, Retrospective, Observational Study. Front Pharmacol 2021; 12:598708. [PMID: 33854429 PMCID: PMC8039458 DOI: 10.3389/fphar.2021.598708] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 02/12/2021] [Indexed: 12/04/2022] Open
Abstract
Anthracycline-associated cardiotoxicity is frequently seen in cancer survivors years after treatment, but it is rare in patients on chemotherapy. This study aimed to investigate the clinical characteristics of cardiac disorders in children with acute lymphoblastic leukemia (ALL) during chemotherapy. A retrospective case study was conducted in children with ALL, for whom electrocardiogram (ECG) and echocardiography (Echo) were regularly assessed before each course of chemotherapy. The cardiac disorders were diagnosed according to the Common Terminology Criteria for Adverse Events (CTCAE) Version 5.0. Binary logistic regression analysis was used to identify risk factors associated with cardiac disorders. There were 171 children eligible for the study, and 78 patients (45.61%) were confirmed as having cardiac disorders. The incidence of cardiac disorders was dependent upon the cumulative dose of daunorubicin (DNR) (p = 0.030, OR = 1.553, 95% CI: 1.005–3.108). Four patients (2.34%) presented with palpitation, chest pain, and persistent tachycardia, and they were cured or improved after medical intervention. A total of 74 patients (43.27%) had subclinical cardiac disorders confirmed by ECG or Echo. ECG abnormalities were commonly seen in the induction and continuation treatments, including arrhythmias (26, 15.20%), ST changes (24, 14.04%) and conduction disorders (4, 2.34%). Pericardial effusion (14, 8.19%), left ventricular hypertrophy (11, 6.43%), a widened pulmonary artery (5, 2.92%) and valvular insufficiency (5, 2.92%) suggested by Echo occurred after induction chemotherapy. Therefore, cardiac disorders with clinical manifestations are rare and need early intervention. Subclinical cardiac disorders are common but very hidden in children during ALL chemotherapy. Regular ECG and Echo could help paediatricians to identify and monitor patients with asymptomatic cardiac disorders earlier.
Collapse
Affiliation(s)
- Hui Yu
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yining Qiu
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Yu
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhujun Wang
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiawei Xu
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yun Peng
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xia Wan
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyan Wu
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Runming Jin
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fen Zhou
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
9
|
Ibrahim DA, Almutawakel MA, Al-Badani R. Cardioprotective effect of Malva verticillata against doxorubicin -induced toxicity in rats. CLINICAL PHYTOSCIENCE 2021. [DOI: 10.1186/s40816-021-00265-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
M. verticillata (Malvaceae) is a medicinal plant used in the treatment of wounds, boils, and liver injuries. The plant leaf extracts possess anti-inflammatory and antioxidant activities. Doxorubicin (DOX) is a potent chemotherapeutic agent used in the treatment of various cancers, but its clinical use is limited by acute and chronic cardiotoxicity. This study aims to evaluate the possible cardioprotective role of Malva verticillata against doxorubicin-induced cardiotoxicity.
Method
Thirty-six male albino rats were divided into six groups, (n = 6): G1: normal control (was given 1 ml/kg of NaCl, 0.9%, twice a week IP), G2: cardiotoxic group (was given 1 mg/kg of DOX twice a week IP). G3 and G4 were given 250 mg/kg and 500 mg/kg of M. verticillata, respectively, while G5 and G6: were given 250, 500 mg/kg of M. verticillata PO and 1 mg/kg IP of DOX. for 6 weeks. Total body weight was taken weekly and Heart: body weight ratio was calculated. Blood samples were collected for determination of serum lactate dehydrogenase (LDH), creatine phosphokinase (CPK) and Troponin, the hearts were removed and processed for histopathological examination.
Results
M. verticillata showed a significant dose-dependent reduction in the cardiac enzyme levels, LDH, CPK activities, and Troponin levels. The histopathological studies in rat hearts also supported those findings.
Conclusion
The present study suggests that M. verticillata may have a novel and worthwhile cardioprotective effect against DOX-induced cardiotoxicity.
Collapse
|
10
|
Grakova EV, Shilov SN, Kopeva KV, Berezikova EN, Popova AA, Neupokoeva MN, Ratushnyak ET, Teplyakov AT. Anthracycline-Induced Cardiotoxicity: The Role of Endothelial Dysfunction. Cardiology 2021; 146:315-323. [PMID: 33596565 DOI: 10.1159/000512771] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 09/26/2020] [Indexed: 11/19/2022]
Abstract
Cardiovascular disease remains the leading cause of mortality accounting up to 40% of all deaths, but, currently, cancer is prominent cause of death globally. Anthracyclines are the cornerstone of chemotherapy in women with breast cancer. However, its clinical use is limited by their cardiotoxic effects that can trigger heart failure development. Vascular toxicity of chemotherapy may be linked with endothelial dysfunction because anthracycline damage of endothelial cells can lead to the development and progression of cardiomyopathy by decreasing the release and activity of endothelial factors and, ultimately, endothelial cell death. These processes suppress anti-inflammatory and vascular reparative functions and initiate the development of future cardiovascular events. Recent studies have shown that chemotherapy may induce toxicity in the vascular endothelium and is accompanied by systemic endothelial dysfunction in patients with diagnosed cardiovascular diseases. Because the initial endothelial cell insult is likely asymptomatic, there is often a long delay between the termination of doxorubicin therapy and the onset of vascular disorders. In this case, genetic susceptibility factor will help to identify susceptible patients in the future. The objectives of this study were to evaluate prognostic role of molecular (endothelin-1) and genetic factors (gene polymorphisms of endothelial nitric oxide (NO) synthase (NOS3, rs1799983), endothelin-1 receptor type A (EDNRA, C+70G, rs5335) and NADPH oxidase (C242T, rs4673) in development of endothelial dysfunction and anthracycline-induced cardiotoxicity in women without cardiovascular diseases.
Collapse
Affiliation(s)
- Elena V Grakova
- Department of Myocardial Pathology, Tomsk National Research Medical Center of the Russian Academy of Sciences, Cardiology Research Institute, Tomsk, Russian Federation
| | - Sergey N Shilov
- Department of Pathological Physiology and Clinical Pathophysiology, Novosibirsk State Medical University, Novosibirsk, Russian Federation
| | - Kristina V Kopeva
- Department of Myocardial Pathology, Tomsk National Research Medical Center of the Russian Academy of Sciences, Cardiology Research Institute, Tomsk, Russian Federation,
| | - Ekaterina N Berezikova
- Department of Pathological Physiology and Clinical Pathophysiology, Novosibirsk State Medical University, Novosibirsk, Russian Federation
| | - Anna A Popova
- Department of Pathological Physiology and Clinical Pathophysiology, Novosibirsk State Medical University, Novosibirsk, Russian Federation
| | - Maria N Neupokoeva
- Department of Pathological Physiology and Clinical Pathophysiology, Novosibirsk State Medical University, Novosibirsk, Russian Federation
| | - Elena T Ratushnyak
- Department of Pathological Physiology and Clinical Pathophysiology, Novosibirsk State Medical University, Novosibirsk, Russian Federation
| | - Alexander T Teplyakov
- Department of Myocardial Pathology, Tomsk National Research Medical Center of the Russian Academy of Sciences, Cardiology Research Institute, Tomsk, Russian Federation
| |
Collapse
|
11
|
Impact of Arterial Hypertension on Doxorubicin-Based Chemotherapy-Induced Subclinical Cardiac Damage in Breast Cancer Patients. Cardiovasc Toxicol 2020; 20:321-327. [PMID: 31782105 DOI: 10.1007/s12012-019-09556-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Advances in oncologic therapies have allowed to achieve better outcomes and longer survival in many patients with breast cancer. Anthracyclines are cytotoxic antibiotics widely used in daily oncology practice. However, anthracyclines cause cardiotoxicity which is a limiting factor of its use. Cumulative dose of anthracyclines is the major cause of induced cardiotoxicity. According to previous clinical trials, the major predisposing high-risk factors for anthracycline-based chemotherapy-induced cardiotoxicity are age, body weight, female gender, radiotherapy, and other diseases such as diabetes and hypertension. Experimental studies in animals confirm that hypertension may be a significant factor predisposing anthracycline-based chemotherapy cardiotoxicity. The main objective of our study was to identify the effect of pre-existing arterial hypertension on the development of subclinical cardiac damage during or after doxorubicin-based chemotherapy in breast cancer patients. The study was performed prospectively between March 2016 and January 2017 in the Hospital of Lithuanian University of Health Sciences Kaunas Clinics Department of Oncology and Department of Cardiology. Data of 73 women with breast cancer treated with doxorubicin-based chemotherapy in outpatient clinic were analyzed. Statistically significant association between pre-existing arterial hypertension and left ventricular systolic dysfunction after completion of chemotherapy was observed (P < 0.004). Our study demonstrated that pre-existing arterial hypertension has a very important role in the development of anthracycline-based chemotherapy-induced cardiotoxicity, despite arterial hypertension control quality. Consequently, further studies evaluating impact of other risk factors and how early and sufficient management of arterial hypertension could influence the development of cardiotoxicity are needed to avoid permanent cardiac damage.
Collapse
|
12
|
Rocca C, Pasqua T, Cerra MC, Angelone T. Cardiac Damage in Anthracyclines Therapy: Focus on Oxidative Stress and Inflammation. Antioxid Redox Signal 2020; 32:1081-1097. [PMID: 31928066 DOI: 10.1089/ars.2020.8016] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Despite their serious side effects, anthracyclines (ANTs) are the most prescribed chemotherapeutic drugs because of their strong efficacy in both solid and hematological tumors. A major limitation to ANTs clinical application is the severe cardiotoxicity observed both acutely and chronically. The mechanism underlying cardiac dysfunction under chemotherapy is mainly dependent on the generation of oxidative stress and systemic inflammation, both of which lead to progressive cardiomyopathy and heart failure. Recent Advances: Over the years, the iatrogenic ANTs-induced cardiotoxicity was believed to be simply given by iron metabolism and reactive oxygen species production; however, several experimental data indicate that ANTs may use alternative damaging mechanisms, such as topoisomerase 2β inhibition, inflammation, pyroptosis, immunometabolism, and autophagy. Critical Issues: In this review, we aimed at discussing ANTs-induced cardiac injury from different points of view, updating and focusing on oxidative stress and inflammation, since these pathways are not exclusive or independent from each other but they together importantly contribute to the complexity of ANTs-induced multifactorial cardiotoxicity. Future Directions: A deeper understanding of the mechanistic signaling leading to ANTs side effects could reveal crucial targeting molecules, thus representing strategic knowledge to promote better therapeutic efficacy and lower cardiotoxicity during clinical application.
Collapse
Affiliation(s)
- Carmine Rocca
- Laboratory of Cellular and Molecular Cardiovascular Physiology, Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
| | - Teresa Pasqua
- Laboratory of Cellular and Molecular Cardiovascular Physiology, Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
| | - Maria Carmela Cerra
- Laboratory of Organ and System Physiology, Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy.,National Institute of Cardiovascular Research (INRC), Bologna, Italy
| | - Tommaso Angelone
- Laboratory of Cellular and Molecular Cardiovascular Physiology, Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy.,National Institute of Cardiovascular Research (INRC), Bologna, Italy
| |
Collapse
|
13
|
Todorova VK, Siegel ER, Kaufmann Y, Kumarapeli A, Owen A, Wei JY, Makhoul I, Klimberg VS. Dantrolene Attenuates Cardiotoxicity of Doxorubicin Without Reducing its Antitumor Efficacy in a Breast Cancer Model. Transl Oncol 2020; 13:471-480. [PMID: 31918212 PMCID: PMC7031101 DOI: 10.1016/j.tranon.2019.12.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/02/2019] [Accepted: 12/03/2019] [Indexed: 02/07/2023] Open
Abstract
Dysregulation of calcium homeostasis is a major mechanism of doxorubicin (DOX)-induced cardiotoxicity. Treatment with DOX causes activation of sarcoplasmic reticulum (SR) ryanodine receptor (RYR) and rapid release of Ca2+ in the cytoplasm resulting in depression of myocardial function. The aim of this study was to examine the effect of dantrolene (DNT) a RYR blocker on both the cardiotoxicity and antitumor activity of DOX in a rat model of breast cancer. Female F344 rats with implanted MAT B III breast cancer cells were randomized to receive intraperitoneal DOX twice per week (12 mg/kg total dose), 5 mg/kg/day oral DNT or a combination of DOX + DNT for 3 weeks. Echocardiography and blood troponin I levels were used to measure myocardial injury. Hearts and tumors were evaluated for histopathological alterations. Blood glutathione was assessed as a measure of oxidative stress. The results showed that DNT improved DOX-induced alterations in the echocardiographic parameters by 50%. Histopathologic analysis of hearts showed reduced DOX induced cardiotoxicity in the group treated with DOX + DNT as shown by reduced interstitial edema, cytoplasmic vacuolization, and myofibrillar disruption, compared with DOX-only–treated hearts. Rats treated with DNT lost less body weight, had higher blood GSH levels and lower troponin I levels than DOX-treated rats. These data indicate that DNT is able to provide protection against DOX cardiotoxicity without reducing its antitumor activity. Further studies are needed to determine the optimal dosing of DNT and DOX in a tumor-bearing host.
Collapse
Affiliation(s)
- Valentina K Todorova
- Department of Surgery, University of Arkansas for Medical Sciences, Little Rock, USA.
| | - Eric R Siegel
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Yihong Kaufmann
- Department of Surgery, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Asangi Kumarapeli
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Aaron Owen
- Department of Surgery, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Jeanne Y Wei
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Issam Makhoul
- Division of Medical Oncology, University of Arkansas for Medical Sciences, Little Rock, USA
| | - V Suzanne Klimberg
- Department of Surgery, University of Arkansas for Medical Sciences, Little Rock, USA
| |
Collapse
|
14
|
Yang X, Liu N, Li X, Yang Y, Wang X, Li L, Jiang L, Gao Y, Tang H, Tang Y, Xing Y, Shang H. A Review on the Effect of Traditional Chinese Medicine Against Anthracycline-Induced Cardiac Toxicity. Front Pharmacol 2018; 9:444. [PMID: 29867456 PMCID: PMC5963334 DOI: 10.3389/fphar.2018.00444] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 04/16/2018] [Indexed: 01/29/2023] Open
Abstract
Anthracyclines are effective agents generally used to treat solid-tumor and hematologic malignancies. The use of anthracyclines for over 40 years has improved cancer survival statistics. Nevertheless, the clinical utility of anthracyclines is limited by its dose-dependent cardiotoxicity that adversely affects 10-30% of patients. Anthracycline-induced cardiotoxicity may be classified as acute/subacute or chronic/late toxicity and leads to devastating adverse effects resulting in poor quality of life, morbidity, and premature mortality. Traditional Chinese medicine has a history of over 2,000 years, involving both unique theories and substantial experience. Several studies have investigated the potential of natural products to decrease the cardiotoxic effects of chemotherapeutic agents on healthy cells, without negatively affecting their antineoplastic activity. This article discusses the mechanism of anthracycline-induced cardiotoxicity, and summarizes traditional Chinese medicine treatment for anthracycline-induced heart failure (HF), cardiac arrhythmia, cardiomyopathy, and myocardial ischemia in recent years, in order to provide a reference for the clinical prevention and treatment of cardiac toxicity.
Collapse
Affiliation(s)
- Xinyu Yang
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China.,Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Nian Liu
- Department of Cardiology, Beijing Anzhen Hospital of the Capital University of Medical Sciences, Beijing, China
| | - Xinye Li
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China.,Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Yihan Yang
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China.,Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Xiaofeng Wang
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Linling Li
- Department of Cardiology, Beijing Anzhen Hospital of the Capital University of Medical Sciences, Beijing, China
| | - Le Jiang
- Department of Cardiology, Beijing Anzhen Hospital of the Capital University of Medical Sciences, Beijing, China
| | - Yonghong Gao
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Hebin Tang
- Department of Pharmacology, School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, China
| | - Yong Tang
- Department of Pancreatic Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yanwei Xing
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Hongcai Shang
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
15
|
Silva dos Santos D, Brasil GV, Ramos IPR, Mesquita FCP, Kasai-Brunswick TH, Christie MLA, Cahli GM, Barbosa RAQ, da Cunha ST, Pereira JX, Medei E, Campos de Carvalho AC, Carvalho AB, Goldenberg RCDS. Embryonic stem cell-derived cardiomyocytes for the treatment of doxorubicin-induced cardiomyopathy. Stem Cell Res Ther 2018; 9:30. [PMID: 29402309 PMCID: PMC5799903 DOI: 10.1186/s13287-018-0788-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 01/19/2018] [Accepted: 01/23/2018] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Doxorubicin (Dox) is a chemotherapy drug with limited application due to cardiotoxicity that may progress to heart failure. This study aims to evaluate the role of cardiomyocytes derived from mouse embryonic stem cells (CM-mESCs) in the treatment of Dox-induced cardiomyopathy (DIC) in mice. METHODS The mouse embryonic stem cell (mESC) line E14TG2A was characterized by karyotype analysis, gene expression using RT-PCR and immunofluorescence. Cells were transduced with luciferase 2 and submitted to cardiac differentiation. Total conditioned medium (TCM) from the CM-mESCs was collected for proteomic analysis. To establish DIC in CD1 mice, Dox (7.5 mg/kg) was administered once a week for 3 weeks, resulting in a cumulative Dox dose of 22.5 mg/kg. At the fourth week, a group of animals was injected intramyocardially with CM-mESCs (8 × 105 cells). Cells were tracked by a bioluminescence assay, and the body weight, echocardiogram, electrocardiogram and number of apoptotic cardiomyocytes were evaluated. RESULTS mESCs exhibited a normal karyotype and expressed pluripotent markers. Proteomic analysis of TCM showed proteins related to the negative regulation of cell death. CM-mESCs presented ventricular action potential characteristics. Mice that received Dox developed heart failure and showed significant differences in body weight, ejection fraction (EF), end-systolic volume (ESV), stroke volume (SV), heart rate and QT and corrected QT (QTc) intervals when compared to the control group. After cell or placebo injection, the Dox + CM-mESC group showed significant increases in EF and SV when compared to the Dox + placebo group. Reduction in ESV and QT and QTc intervals in Dox + CM-mESC-treated mice was observed at 5 or 30 days after cell treatment. Cells were detected up to 11 days after injection. The Dox + CM-mESC group showed a significant reduction in the percentage of apoptotic cardiomyocytes in the hearts of mice when compared to the Dox + placebo group. CONCLUSIONS CM-mESC transplantation improves cardiac function in mice with DIC.
Collapse
Affiliation(s)
- Danúbia Silva dos Santos
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
| | - Guilherme Visconde Brasil
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
| | - Isalira Peroba Rezende Ramos
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
- Centro Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Bloco M, Rio de Janeiro, RJ 21941-902 Brazil
| | - Fernanda Cristina Paccola Mesquita
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
| | - Tais Hanae Kasai-Brunswick
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
- Centro Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Bloco M, Rio de Janeiro, RJ 21941-902 Brazil
| | - Michelle Lopes Araújo Christie
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
| | - Gustavo Monnerat Cahli
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
| | - Raiana Andrade Quintanilha Barbosa
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
| | - Sandro Torrentes da Cunha
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
| | - Jonathas Xavier Pereira
- Departamento de Patologia—Faculdade de Medicina, Hospital Universitário Clementino Fraga Filho, Universiade Federal do Rio de Janeiro, Av. Rodolpho Paulo Rocco, 255, Sub-solo, SAP, Rio de Janeiro, RJ 21910-590 Brazil
| | - Emiliano Medei
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
- Centro Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Bloco M, Rio de Janeiro, RJ 21941-902 Brazil
- Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Av. Carlos Chagas Filho 373, Rio de Janeiro, RJ 21941-902 Brazil
| | - Antonio Carlos Campos de Carvalho
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
- Centro Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Bloco M, Rio de Janeiro, RJ 21941-902 Brazil
- Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Av. Carlos Chagas Filho 373, Rio de Janeiro, RJ 21941-902 Brazil
| | - Adriana Bastos Carvalho
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
- Centro Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Bloco M, Rio de Janeiro, RJ 21941-902 Brazil
- Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Av. Carlos Chagas Filho 373, Rio de Janeiro, RJ 21941-902 Brazil
| | - Regina Coeli dos Santos Goldenberg
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
- Centro Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Bloco M, Rio de Janeiro, RJ 21941-902 Brazil
- Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Av. Carlos Chagas Filho 373, Rio de Janeiro, RJ 21941-902 Brazil
| |
Collapse
|
16
|
Chaudhry MA. The Intriguing Occurrence of Segmental Arterial Mediolysis: Case Report and Concise Literature Review. Cardiovasc Hematol Disord Drug Targets 2017; 17:74-79. [PMID: 28676031 DOI: 10.2174/1871529x17666170703115833] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 03/14/2017] [Accepted: 06/06/2017] [Indexed: 11/22/2022]
Abstract
BACKGROUND Segmental medial arteriolysis (SAM) is a unique arteriopathy highlighted by significant lytic changes in the medial wall of the blood vessels and can present from vague gastrointestinal discomfort to catastrophic abdominal bleeding and shock. We hereby present a concise review of this rare phenomenon with historic perspectives, epidemiology, and current concepts of etiology, pathogenesis, relevant clinical associations, treatment modalities, prognosis and future directions in SAM. CONCLUSION In addition, we present an interesting occurrence of this intriguing phenomenon in a forty-eight year old lady at our institution who presented with vague symptomatology and was an extremely challenging diagnosis. This highlights the importance of timely detection and institution of therapeutic or preventive strategies to minimize future catastrophic events.
Collapse
|
17
|
Leger KJ, Leonard D, Nielson D, de Lemos JA, Mammen PPA, Winick NJ. Circulating microRNAs: Potential Markers of Cardiotoxicity in Children and Young Adults Treated With Anthracycline Chemotherapy. J Am Heart Assoc 2017; 6:JAHA.116.004653. [PMID: 28377429 PMCID: PMC5532993 DOI: 10.1161/jaha.116.004653] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Background Biomarkers for early detection of anthracycline (AC)‐induced cardiotoxicity may allow cardioprotective intervention before irreversible damage. Circulating microRNAs (miRNAs) are promising biomarkers of cardiovascular disease, however, have not been studied in the setting of AC‐induced cardiotoxicity. This study aimed to identify AC‐induced alterations in plasma miRNA expression in children and correlate expression with markers of cardiac injury. Methods and Results Candidate plasma profiling of 24 miRNAs was performed in 33 children before and after a cycle of AC (n=24) or noncardiotoxic chemotherapy (n=9). Relative miRNA changes between the pre‐ and postcycle time points (6, 12, and 24 hours) were determined within each treatment group and compared across groups. Plasma miRNA expression patterns were further explored with respect to AC dose and high‐sensitivity troponin T. Greater chemotherapy‐induced dysregulation was observed in this panel of candidate, cardiac‐related plasma miRNAs in patients receiving anthracyclines compared with those receiving noncardiotoxic chemotherapy (24‐hour MANOVA; P=0.024). Specifically, plasma miRs‐29b and ‐499 were upregulated 6 to 24 hours post‐AC, and their postchemotherapy expression significantly correlated with AC dose. Patients with acute cardiomyocyte injury (high‐sensitivity troponin T increase ≥5 ng/L from baseline) demonstrated higher expression of miR‐29b and miR‐499 post‐AC compared with those without. Conclusions In this pilot study, cardiac‐related plasma miRNAs are dysregulated following ACs. Plasma miR‐29b and ‐499 are acutely elevated post‐AC, with dose response relationships observed with anthracycline dose and markers of cardiac injury. Further evaluation of miRNAs may provide mechanistic insight into AC‐induced cardiotoxicity and yield biomarkers to facilitate earlier intervention to mitigate cardiotoxicity.
Collapse
Affiliation(s)
- Kasey J Leger
- Department of Pediatrics, Seattle Children's Hospital, University of Washington School of Medicine, Seattle, WA
| | - David Leonard
- Department of Clinical Research, Children's Medical Center, Dallas, TX
| | - Danelle Nielson
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX
| | - James A de Lemos
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX
| | - Pradeep P A Mammen
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX
- Heart Failure, Ventricular Assist Device & Heart Transplant Program, University of Texas Southwestern Medical Center, Dallas, TX
| | - Naomi J Winick
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
18
|
UVRAG Deficiency Exacerbates Doxorubicin-Induced Cardiotoxicity. Sci Rep 2017; 7:43251. [PMID: 28225086 PMCID: PMC5320807 DOI: 10.1038/srep43251] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Accepted: 01/23/2017] [Indexed: 02/07/2023] Open
Abstract
Doxorubicin (DOX) is an effective chemotherapeutic drug in the treatment of various types of cancers. However, its clinical application has been largely limited by potential development of cardiotoxicity. Previously we have shown that ultra-violet radiation resistance-associated gene (UVRAG), an autophagy-related protein, is essential for the maintenance of autophagic flux in the heart under physiological conditions. Here, we sought to determine the role of UVRAG-mediated autophagy in DOX-induced cardiotoxicity. Mouse models of acute or chronic DOX-induced cardiotoxicity were established. UVRAG deficiency exacerbated DOX-induced mortality and cardiotoxicity manifested by increased cytoplasmic vacuolization, enhanced collagen accumulation, elevated serum activities of lactate dehydrogenase and myocardial muscle creatine kinase, higher ROS levels, aggravated apoptosis and more depressed cardiac function. Autophagic flux was impaired in DOX-induced cardiotoxicity. UVRAG deficiency aggravated impaired autophagic flux in DOX-induced cardiotoxicity. Intermittent fasting restored autophagy and ameliorated pathological alterations of DOX-induced cardiotoxicity. Collectively, our data suggest that UVRAG deficiency exacerbates DOX-induced cardiotoxicity, at least in part, through aggravation of DOX-induced impaired autophagic flux. Intermittent fasting, which restores blunted autophagic flux and ameliorates pathology in the mouse models of DOX-induced cardiotoxicity, may be used as a potential preventive or therapeutic approach for DOX cardiotoxicity.
Collapse
|
19
|
Mahjoob MP, Sheikholeslami SA, Dadras M, Mansouri H, Haghi M, Naderian M, Sadeghi L, Tabary M, Khaheshi I. The Intriguing Occurrence of Segmental Arterial Mediolysis: Case Report and Concise Literature Review. Cardiovasc Hematol Disord Drug Targets 2017; 20:74-83. [PMID: 28676031 PMCID: PMC7360911 DOI: 10.2174/1871529x19666190912150942] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 06/30/2019] [Accepted: 08/04/2019] [Indexed: 04/13/2023]
Abstract
BACKGROUND Segmental medial arteriolysis (SAM) is a unique arteriopathy highlighted by significant lytic changes in the medial wall of the blood vessels and can present from vague gastrointestinal discomfort to catastrophic abdominal bleeding and shock. We hereby present a concise review of this rare phenomenon with historic perspectives, epidemiology, and current concepts of etiology, pathogenesis, relevant clinical associations, treatment modalities, prognosis and future directions in SAM. CONCLUSION In addition, we present an interesting occurrence of this intriguing phenomenon in a forty-eight year old lady at our institution who presented with vague symptomatology and was an extremely challenging diagnosis. This highlights the importance of timely detection and institution of therapeutic or preventive strategies to minimize future catastrophic events.
Collapse
Affiliation(s)
- Mohammad P. Mahjoob
- Cardiovascular Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Morvarid Dadras
- Fellowship of Echocardiography, Imam Hosein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamdollah Mansouri
- Department of Internal Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahshid Haghi
- Department of Medical Nanotechnology, Faculty of Advanced Sciences & Technology, Pharmaceutical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Mohammadreza Naderian
- Cardiovascular Research Department, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Sadeghi
- Imam Hossein Hospital, Internal Medicine Department, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Tabary
- Address correspondence to these authors at the School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; E-mail: and Cardiovascular Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; E-mail:
| | - Isa Khaheshi
- Address correspondence to these authors at the School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; E-mail: and Cardiovascular Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; E-mail:
| |
Collapse
|
20
|
Chung WB, Youn HJ. Pathophysiology and preventive strategies of anthracycline-induced cardiotoxicity. Korean J Intern Med 2016; 31:625-33. [PMID: 27378126 PMCID: PMC4939510 DOI: 10.3904/kjim.2016.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Accepted: 05/20/2016] [Indexed: 02/07/2023] Open
Abstract
Cardiotoxicity is a well-known complication following treatment with anthracyclines. However, they are still widely used in chemotherapy for breast cancer, lymphoma, leukemia, and sarcoma, among others. Patient clinical characteristics, such as age, sex, comorbidities, anthracycline dose and infusion schedule, and the combined anti-cancer agents used, are diverse among cancer types. It is difficult to recommend guidelines for the prevention or management of anthracycline-induced cardiotoxicity applicable to all cancer types. Therefore, anthracycline-induced cardiotoxicity remains a major limitation in the proper management of cancer patients treated with an anthracycline-combined regimen. Efforts have been extensive to determine the mechanism and treatment of anthracycline-induced cardiotoxicity. Because cardiotoxicity causes irreversible damage to the myocardium, prevention is a more effective approach than treatment of cardiotoxicity after symptomatic or asymptomatic cardiac dysfunction develops. This article will review the pathophysiological mechanisms of anthracycline-induced cardiotoxicity and strategies for protecting the myocardium from anthracycline.
Collapse
Affiliation(s)
| | - Ho-Joong Youn
- Correspondence to Ho-Joong Youn, M.D. Cardiovascular Center, Division of Cardiology, Department of Internal Medicine, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Korea Tel: +82-2-2258-6029 Fax: +82-2-591-1506 E-mail:
| |
Collapse
|
21
|
Saddic LA, Muehlschlegel JD. Sarco"MiR" friend or foe: a perspective on the mechanisms of doxorubicin-induced cardiomyopathy. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:203. [PMID: 27294099 DOI: 10.21037/atm.2016.05.30] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Anthracyclines are a class of chemotherapeutics used to treat a variety of human cancers including both solid tumors such as breast, ovarian, and lung, as well as malignancies of the blood including leukemia and lymphoma. Despite being extremely effective anti-cancer agents, the application of these drugs is offset by side effects, most notably cardiotoxicity. Many patients treated with doxorubicin (DOX), one of the most common anthracyclines used in oncology, will develop radiographic signs and/or symptoms of cardiomyopathy. Since more and more patients treated with these drugs are surviving their malignancies and manifesting with heart disease, there is particular interest in understanding the mechanisms of anthracycline-induced injury and developing ways to prevent and treat its most feared complication, heart failure. MicroRNAs (miRNAs) are small noncoding RNAs that regulate the expression of mRNAs. Since miRNAs can regulate many mRNAs in a single network they tend to play a crucial role in the pathogenesis of several diseases, including heart failure. Here we present a perspective on a recent work by Roca-Alonso and colleagues who demonstrate a cardioprotective function of the miR-30 family members following DOX-induced cardiac injury. They provide evidence for direct targeting of these miRNAs on key elements of the β-adrenergic pathway and further show that this interaction regulates cardiac function and apoptosis. These experiments deliver fresh insights into the biology of toxin-induced cardiomyopathy and suggest the potential for novel therapeutic targets.
Collapse
Affiliation(s)
- Louis A Saddic
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Jochen D Muehlschlegel
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| |
Collapse
|
22
|
Valcovici M, Andrica F, Serban C, Dragan S. Cardiotoxicity of anthracycline therapy: current perspectives. Arch Med Sci 2016; 12:428-35. [PMID: 27186191 PMCID: PMC4848373 DOI: 10.5114/aoms.2016.59270] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 10/07/2014] [Indexed: 12/31/2022] Open
Abstract
Anthracyclines, especially doxorubicin and daunorubicin, are the drugs of first choice in the treatment of patients with hematologic malignancies, soft-tissue sarcomas, and solid tumors. Unfortunately, the use of anthracyclines is limited by their dose-dependent and cumulative cardiotoxicity. The molecular mechanism responsible for anthracycline-induced cardiotoxicity remains poorly understood, although experimental and clinical studies have shown that oxidative stress plays the main role. Hence, antioxidant agents, especially dexrazoxane, and also other drug classes (statins, β-blockers) proved to have a beneficial effect in protecting against anthracycline-induced cardiotoxicity. According to previous clinical trials, the major high-risk factors for anthracycline-induced cardiotoxicity are age, body weight, female gender, radiotherapy, and other diseases such as Down syndrome, familial dilated cardiomyopathy, diabetes and hypertension. Consequently, further studies are needed to elucidate the molecular pathogenesis of anthracycline-induced cardiotoxicity and also to discover new cardioprotective agents against anthracycline-induced cardiotoxicity.
Collapse
Affiliation(s)
- Mihaela Valcovici
- Cardiology Department, University of Medicine and Pharmacy "Victor Babes", Timisoara, Romania
| | - Florina Andrica
- Department of Toxicology, Faculty of Pharmacy, University of Medicine and Pharmacy "Victor Babes", Timisoara, Romania; Center for Interdisciplinary Research, University of Medicine and Pharmacy "Victor Babes", Timisoara, Romania
| | - Corina Serban
- Center for Interdisciplinary Research, University of Medicine and Pharmacy "Victor Babes", Timisoara, Romania; Department of Functional Sciences, University of Medicine and Pharmacy "Victor Babes", Timisoara, Romania
| | - Simona Dragan
- Cardiology Department, University of Medicine and Pharmacy "Victor Babes", Timisoara, Romania; Center for Interdisciplinary Research, University of Medicine and Pharmacy "Victor Babes", Timisoara, Romania
| |
Collapse
|
23
|
The Therapeutic Potential of AN-7, a Novel Histone Deacetylase Inhibitor, for Treatment of Mycosis Fungoides/Sezary Syndrome Alone or with Doxorubicin. PLoS One 2016; 11:e0146115. [PMID: 26752418 PMCID: PMC4709199 DOI: 10.1371/journal.pone.0146115] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 12/13/2015] [Indexed: 12/12/2022] Open
Abstract
The 2 histone deacetylase inhibitors (HDACIs) approved for the treatment of cutaneous T-cell lymphoma (CTCL) including mycosis fungoides/sezary syndrome (MF/SS), suberoylanilide hydroxamic acid (SAHA) and romidepsin, are associated with low rates of overall response and high rates of adverse effects. Data regarding combination treatments with HDACIs is sparse. Butyroyloxymethyl diethylphosphate (AN-7) is a novel HDACI, which was found to have selective anticancer activity in several cell lines and animal models. The aim of this study was to compare the anticancer effects of AN-7 and SAHA, either alone or combined with doxorubicin, on MF/SS cell lines and peripheral blood lymphocytes (PBL) from patients with Sezary syndrome (SPBL). MyLa cells, Hut78 cells, SPBL, and PBL from healthy normal individuals (NPBL) were exposed to the test drugs, and the findings were analyzed by a viability assay, an apoptosis assay, and Western blot. AN-7 was more selectively toxic to MyLa cells, Hut78 cells, and SPBL (relative to NPBL) than SAHA and also acted more rapidly. Both drugs induced apoptosis in MF/SS cell lines, SAHA had a greater effect on MyLa cell line, while AN-7 induced greater apoptosis in SPBL; both caused an accumulation of acetylated histone H3, but AN-7 was associated with earlier kinetics; and both caused a downregulation of the HDAC1 protein in MF/SS cell lines. AN-7 acted synergistically with doxorubicin in both MF/SS cell lines and SPBL, and antagonistically with doxorubicin in NPBL. By contrast, SAHA acted antagonistically with doxorubicin on MF/SS cell lines, SPBL, and NPBL, leaving <50% viable cells. In conclusion, AN-7 holds promise as a therapeutic agent in MF/SS and has several advantages over SAHA. Our data provide a rationale for combining AN-7, but not SAHA, with doxorubicin to induce the cell death in MF/SS.
Collapse
|
24
|
Liu MH, Zhang Y, He J, Tan TP, Wu SJ, Fu HY, Chen YD, Liu J, LE QF, Hu HJ, Yuan C, Lin XL. Upregulation of peroxiredoxin III in doxorubicin-induced cytotoxicity and the FoxO3a-dependent expression in H9c2 cardiac cells. Exp Ther Med 2015; 10:1515-1520. [PMID: 26622517 DOI: 10.3892/etm.2015.2693] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 08/03/2015] [Indexed: 12/30/2022] Open
Abstract
Doxorubicin (DOX) is an efficient drug used in cancer therapy; however, it produces reactive oxygen species (ROS) that induce severe cytotoxicity, limiting its clinical application. The aim of the present study was to investigate the role of peroxiredoxin III (Prx III) in DOX-induced H9c2 cell injuries. Following DOX treatment, the expression of phosphorylated-FoxO3a (p-FoxO3a) was decreased and Prx III expression was increased in H9c2 cells. In order to detect whether oxidative stress was involved in the induction of Prx III expression by FoxO3a, exogenous H2O2 was used to induce oxidative stress in the H9c2 cells. Apoptosis of H9c2 cardiomyocytes was assessed using methyl thiazolyl tetrazolium assay and Hoechst staining. The levels of Prx III and p-FoxO3a were evaluated using western blot analysis. As expected, H2O2 was found to mimic the effect of DOX, decreasing the expression of p-FoxO3a and increasing the expression of Prx III. In addition, the study evaluated whether the transcription factor FoxO3a was essential for the expression of Prx III. Pretreatment of H9c2 cells with N-acetyl-L-cysteine (NAC), a scavenger of ROS, prior to exposure to DOX dramatically increased the phosphorylation of FoxO3a and led to a marked reduction in Prx III expression in the H9c2 cells. In conclusion, the results of the current study suggest that FoxO3a mediates the expression of Prx III in DOX-induced injuries.
Collapse
Affiliation(s)
- Mi-Hua Liu
- Department of Clinical Laboratory, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yuan Zhang
- Department of Pathology, Hunan Mawangdui Hospital, Changsha, Hunan 410016, P.R. China
| | - Jun He
- Department of Clinical Laboratory, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Tian-Ping Tan
- Department of Clinical Laboratory, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Shao-Jian Wu
- Department of Clinical Laboratory, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Hong-Yun Fu
- Department of Clinical Laboratory, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yu-Dan Chen
- Department of Clinical Laboratory, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Jun Liu
- Department of Clinical Laboratory, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Qun-Fang LE
- Department of Clinical Laboratory, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Heng-Jing Hu
- Department of Cardiology/Cardiac Catheterisation Lab, Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Cong Yuan
- Department of Cardiology, The First Hospital of Changsha, Changsha, Hunan 410005, P.R. China
| | - Xiao-Long Lin
- Department of Pathology, The Third People's Hospital of Huizhou, Guangzhou Medical University, Huizhou, Guangdong 516002, P.R. China
| |
Collapse
|
25
|
Edwardson DW, Narendrula R, Chewchuk S, Mispel-Beyer K, Mapletoft JPJ, Parissenti AM. Role of Drug Metabolism in the Cytotoxicity and Clinical Efficacy of Anthracyclines. Curr Drug Metab 2015; 16:412-26. [PMID: 26321196 PMCID: PMC5398089 DOI: 10.2174/1389200216888150915112039] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 07/31/2015] [Accepted: 08/10/2015] [Indexed: 01/19/2023]
Abstract
Many clinical studies involving anti-tumor agents neglect to consider how these agents are metabolized within the host and whether the creation of specific metabolites alters drug therapeutic properties or toxic side effects. However, this is not the case for the anthracycline class of chemotherapy drugs. This review describes the various enzymes involved in the one electron (semi-quinone) or two electron (hydroxylation) reduction of anthracyclines, or in their reductive deglycosidation into deoxyaglycones. The effects of these reductions on drug antitumor efficacy and toxic side effects are also discussed. Current evidence suggests that the one electron reduction of anthracyclines augments both their tumor toxicity and their toxicity towards the host, in particular their cardiotoxicity. In contrast, the two electron reduction (hydroxylation) of anthracyclines strongly reduces their ability to kill tumor cells, while augmenting cardiotoxicity through their accumulation within cardiomyocytes and their direct effects on excitation/contraction coupling within the myocytes. The reductive deglycosidation of anthracyclines appears to inactivate the drug and only occurs under rare, anaerobic conditions. This knowledge has resulted in the identification of important new approaches to improve the therapeutic index of anthracyclines, in particular by inhibiting their cardiotoxicity. The true utility of these approaches in the management of cancer patients undergoing anthracycline-based chemotherapy remains unclear, although one such agent (the iron chelator dexrazoxane) has recently been approved for clinical use.
Collapse
Affiliation(s)
| | | | | | | | | | - Amadeo M Parissenti
- Dept. of Chemistry and Biochemistry, Laurentian University, 935 Ramsey Lake Road, Sudbury, ON P3E 2C6, Canada.
| |
Collapse
|