1
|
Rajaeinejad M, Parhizkar-Roudsari P, Khoshfetrat M, Kazemi-Galougahi MH, Mosaed R, Arjmand R, Mohsenizadeh SA, Arjmand B. Management of Fluoropyrimidine-Induced Cardiac Adverse Outcomes Following Cancer Treatment. Cardiovasc Toxicol 2024; 24:184-198. [PMID: 38324115 DOI: 10.1007/s12012-024-09834-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 01/20/2024] [Indexed: 02/08/2024]
Abstract
Advancements in cancer treatments have improved survival rates but have also led to increased cardiotoxicities, which can cause adverse cardiovascular events or worsen pre-existing conditions. Herein, cardiotoxicity is a severe adverse effect of 5-fluorouracil (5-FU) therapy in cancer patients, with reported incidence rates ranging from 1 to 20%. Some studies have also suggested subclinical effects and there are reports which have documented instances of cardiac arrest or sudden death during 5-FU treatment, highlighting the importance of timely management of cardiovascular symptoms. However, despite being treated with conventional medical approaches for this cardiotoxicity, a subset of patients has demonstrated suboptimal or insufficient responses. The frequent use of 5-FU in chemotherapy and its association with significant morbidity and mortality indicates the need for a greater understanding of 5-FU-associated cardiotoxicity. It is essential to reduce the adverse effects of anti-tumor medications while preserving their efficacy, which can be achieved through drugs that mitigate toxicity associated with these drugs. Underpinning cardiotoxicity associated with 5-FU therapy also has the potential to offer valuable guidance in pinpointing pharmacological approaches that can be employed to prevent or ameliorate these effects. The present study provides an overview of management strategies for cardiac events induced by fluoropyrimidine-based cancer treatments. The review encompasses the underlying molecular and cellular mechanisms of cardiotoxicity, associated risk factors, and diagnostic methods. Additionally, we provide information on several available treatments and drug choices for angina resulting from 5-FU exposure, including nicorandil, ranolazine, trimetazidine, ivabradine, and sacubitril-valsartan, which have demonstrated potential in mitigating or protecting against chemotherapy-induced adverse cardiac effects.
Collapse
Affiliation(s)
- Mohsen Rajaeinejad
- AJA Cancer Epidemiology Research and Treatment Center (AJA-CERTC), AJA University of Medical Sciences, Tehran, Iran
| | - Peyvand Parhizkar-Roudsari
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical sciences, Tehran, Iran
- Iranian Cancer Control Center, Tehran, Iran
| | - Mehran Khoshfetrat
- Department of Cardiology, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | | | - Reza Mosaed
- Infection Diseases Research Center, AJA University of Medical Sciences, Tehran, Iran
- Student Research Committee, AJA University of Medical Sciences, Tehran, Iran
| | - Rasta Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Babak Arjmand
- Department of Internal Medicine, School of Medicine, AJA University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Liao Y, Meng Q. Protection against cancer therapy-induced cardiovascular injury by planed-derived polyphenols and nanomaterials. ENVIRONMENTAL RESEARCH 2023; 238:116896. [PMID: 37586453 DOI: 10.1016/j.envres.2023.116896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/18/2023] [Accepted: 08/13/2023] [Indexed: 08/18/2023]
Abstract
Cancer therapy-induced heart injury is a significant concern for cancer patients undergoing chemotherapy, radiotherapy, immunotherapy, and also targeted molecular therapy. The use of these treatments can lead to oxidative stress and cardiomyocyte damage in the heart, which can result in heart failure and other cardiac complications. Experimental studies have revealed that chemotherapy drugs such as doxorubicin and cyclophosphamide can cause severe side effects such as cardiac fibrosis, electrophysiological remodeling, chronic oxidative stress and inflammation, etc., which may increase risk of cardiac disorders and attacks for patients that underwent chemotherapy. Similar consequences may also be observed for patients that undergo radiotherapy for left breast or lung malignancies. Polyphenols, a group of natural compounds with antioxidant and anti-inflammatory properties, have shown the potential in protecting against cancer therapy-induced heart injury. These compounds have been found to reduce oxidative stress, necrosis and apoptosis in the heart, thereby preserving cardiac function. In recent years, nanoparticles loaded with polyphenols have also provided for the delivery of these compounds and increasing their efficacy in different organs. These nanoparticles can improve the bioavailability and efficacy of polyphenols while minimizing their toxicity. This review article summarizes the current understanding of the protective effects of polyphenols and nanoparticles loaded with polyphenols against cancer therapy-induced heart injury. The article discusses the mechanisms by which polyphenols protect the heart, including antioxidant and anti-inflammation abilities. The article also highlights the potential benefits of using nanoparticles for the delivery of polyphenols.
Collapse
Affiliation(s)
- Yunshu Liao
- Department of Cardiac Surgery, The First Hospital Affiliated to the Army Medical University, Chongqing, 400038, China
| | - Qinghua Meng
- Department of Cardiac Surgery, The First Hospital Affiliated to the Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
3
|
Morelli MB, Bongiovanni C, Da Pra S, Miano C, Sacchi F, Lauriola M, D’Uva G. Cardiotoxicity of Anticancer Drugs: Molecular Mechanisms and Strategies for Cardioprotection. Front Cardiovasc Med 2022; 9:847012. [PMID: 35497981 PMCID: PMC9051244 DOI: 10.3389/fcvm.2022.847012] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 03/03/2022] [Indexed: 12/13/2022] Open
Abstract
Chemotherapy and targeted therapies have significantly improved the prognosis of oncology patients. However, these antineoplastic treatments may also induce adverse cardiovascular effects, which may lead to acute or delayed onset of cardiac dysfunction. These common cardiovascular complications, commonly referred to as cardiotoxicity, not only may require the modification, suspension, or withdrawal of life-saving antineoplastic therapies, with the risk of reducing their efficacy, but can also strongly impact the quality of life and overall survival, regardless of the oncological prognosis. The onset of cardiotoxicity may depend on the class, dose, route, and duration of administration of anticancer drugs, as well as on individual risk factors. Importantly, the cardiotoxic side effects may be reversible, if cardiac function is restored upon discontinuation of the therapy, or irreversible, characterized by injury and loss of cardiac muscle cells. Subclinical myocardial dysfunction induced by anticancer therapies may also subsequently evolve in symptomatic congestive heart failure. Hence, there is an urgent need for cardioprotective therapies to reduce the clinical and subclinical cardiotoxicity onset and progression and to limit the acute or chronic manifestation of cardiac damages. In this review, we summarize the knowledge regarding the cellular and molecular mechanisms contributing to the onset of cardiotoxicity associated with common classes of chemotherapy and targeted therapy drugs. Furthermore, we describe and discuss current and potential strategies to cope with the cardiotoxic side effects as well as cardioprotective preventive approaches that may be useful to flank anticancer therapies.
Collapse
Affiliation(s)
| | - Chiara Bongiovanni
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Silvia Da Pra
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Carmen Miano
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
| | - Francesca Sacchi
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Mattia Lauriola
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Gabriele D’Uva
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
- *Correspondence: Gabriele D’Uva,
| |
Collapse
|
4
|
Wenxin Keli Regulates Mitochondrial Oxidative Stress and Homeostasis and Improves Atrial Remodeling in Diabetic Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:2468031. [PMID: 32104528 PMCID: PMC7040409 DOI: 10.1155/2020/2468031] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 01/07/2020] [Accepted: 01/25/2020] [Indexed: 12/13/2022]
Abstract
Mitochondrial dysfunction and oxidative stress play an important role in the pathogenesis of both atrial fibrillation (AF) and diabetes mellitus (DM). Wenxin Keli (WXKL), an antiarrhythmic traditional Chinese medicine, has been shown to prevent cardiac arrhythmias through modulation of cardiac ion channels. This study tested the hypothesis that WXKL can improve atrial remodeling in diabetic rats by restoring mitochondrial function. Primary atrial fibroblasts of neonatal SD rats were divided into four groups: control, hydrogen peroxide (H2O2), H2O2+WXKL 1 g/L, and H2O2+WXKL 3 g/L groups. Intracellular mitochondrial membrane potential (MMP), reactive oxygen species (ROS), and mitochondrial oxygen consumption were measured. SD male rats were randomly divided into three groups: control, DM, and DM+WXKL groups. Rats in the DM+WXKL group were treated with daily gavage of WXKL at 3 g/kg. After eight weeks, echocardiography, hemodynamic examination, histology, electrophysiology study, mitochondrial respiratory function, and western blots were assessed. H2O2 treatment led to increased ROS and decreased intracellular MMP and mitochondrial oxygen consumption in primary atrial fibroblasts. WXKL improved the above changes. DM rats showed increased atrial fibrosis, greater left atrial diameter, lower atrial conduction velocity, higher conduction heterogeneity, higher AF inducibility, and lower mitochondrial protein expression, and all these abnormal changes except for left atrial diameter were improved in the DM+WXKL group. WXKL improves atrial remodeling by regulating mitochondrial function and homeostasis and reducing mitochondrial ROS in diabetic rats.
Collapse
|
5
|
Solomou E, Aznaouridis K, Masoura C, Cutajar I, Toutouzas K, Vlachopoulos C, Tousoulis D. Aortic wall stiffness as a side-effect of anti-cancer medication. Expert Rev Cardiovasc Ther 2019; 17:791-799. [PMID: 31715108 DOI: 10.1080/14779072.2019.1691528] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Introduction: Malignancies and cardiovascular disease are the two leading causes of mortality worldwide. There is a growing concern that anti-cancer drugs may lead to increased cardiovascular morbidity among cancer survivors. This may be the result of direct effects of the cancer treatment on heart function, or due to an indirect acceleration of atherosclerosis.Areas covered: We searched two bibliographic databases [PubMed, Scopus] and one full-text database (Google Scholar) for publications on chemotherapy and arterial stiffness since 1970. Anthracyclines, alkylating agents and tyrosine kinase inhibitors seem to affect arterial elastic properties. These effects can be non-reversible and may appear after treatment termination. Monoclonal antibodies may induce either a temporary increase or no change on arterial stiffness of patients with malignancies. Anti-microtubule agents and antimetabolites have not been extensively studied so far.Expert opinion: This literature review suggests that certain anticancer medications may impair arterial stiffness, and that assessment of arterial elastic properties before and after initiation of anti-neoplasmatic therapy may be clinically useful in order to develop protective strategies against chemotherapy-induced vascular effects. Further research is warranted to confirm the effects of anti-cancer agents on arterial stiffness, as well as their potential clinical implications. Future research lies in finding new targeted biomarkers identifying arterial stiffness such as micro RNAs while imaging techniques could also be implemented in assessment of vascular toxicity.
Collapse
Affiliation(s)
- Eirini Solomou
- 1st Department of Cardiology, Hippokration Hospital, Athens, Greece
| | | | | | - Iosif Cutajar
- 1st Department of Cardiology, Hippokration Hospital, Athens, Greece
| | | | | | | |
Collapse
|
6
|
Korzeniowska K, Jankowski J, Cieślewicz A, Jabłecka A. Is it possible to prevent chemotherapy-induced heart failure with cardiovascular drugs - the review of the current clinical evidence. Ther Clin Risk Manag 2019; 15:1095-1110. [PMID: 31564887 PMCID: PMC6743633 DOI: 10.2147/tcrm.s215857] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 07/25/2019] [Indexed: 01/26/2023] Open
Abstract
Cardiovascular diseases and cancer are the most common death causes in the USA and Europe. Moreover, many patients suffer from both of these conditions - a situation which may result from cardiotoxicity of anticancer treatment. In order to reduce the severity of this adverse effect, various methods have been proposed, including the usage of new drug forms and less toxic analogs, omitting the combinations of potentially cardiotoxic drugs and introducing potential cardioprotective agents to the therapy. However, prevention of cardiotoxicity still seems to be insufficient. The article reviews the results of current studies on the use of cardiovascular drugs in the prevention of cardiotoxicity. Based on this knowledge, the most promising cardioprotective drugs seem to be carvedilol, nebivolol, enalapril, and candesartan, as they prevent heart remodeling and correct elevated resting heart rate, which directly affects mortality. Alternatively, in case of adverse reactions, statins might be considered.
Collapse
Affiliation(s)
- Katarzyna Korzeniowska
- Department of Clinical Pharmacology, Poznan University of Medical Sciences, Poznan61-848, Poland
| | - Jerzy Jankowski
- Department of Clinical Pharmacology, Poznan University of Medical Sciences, Poznan61-848, Poland
| | - Artur Cieślewicz
- Department of Clinical Pharmacology, Poznan University of Medical Sciences, Poznan61-848, Poland
| | - Anna Jabłecka
- Department of Clinical Pharmacology, Poznan University of Medical Sciences, Poznan61-848, Poland
| |
Collapse
|
7
|
Ghosh GC, Ghosh RK, Bandyopadhyay D, Chatterjee K, Aneja A. Ranolazine: Multifaceted Role beyond Coronary Artery Disease, a Recent Perspective. Heart Views 2019; 19:88-98. [PMID: 31007857 PMCID: PMC6448470 DOI: 10.4103/heartviews.heartviews_18_18] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Ranolazine is a piperazine derivative approved as an antianginal. Primarily used as a second-line antianginal in stable coronary artery disease. Ranolazine blocks the late Na + current and prevents the rise of cytosolic calcium. It decreases myocardial wall tension and improves coronary blood flow. Ranolazine is effective in atrial fibrillation (AF) as an adjunct to electrical or pharmacological cardioversion. It can be used in combination with amiodarone or dronedarone. It has also been used in AF arising after coronary artery bypass grafting surgery. Role of ranolazine is also being evaluated in pulmonary arterial hypertension, diastolic dysfunction, and chemotherapy-induced cardiotoxicity. Ranolazine has some anti-glycemic effect and has shown a reduction of hemoglobin A1c in multiple trials. The antianginal effect of ranolazine has also been seen to be more in patients with diabetes compared to those without diabetes. Ranolazine is being evaluated in patients with the peripheral arterial disease with intermittent claudication and hypertrophic cardiomyopathy. Pilot studies have shown that ranolazine may be beneficial in neurological conditions with myotonia. The evidence-base on the use of ranolazine in various conditions is rapidly increasing with results of further trials eagerly awaited. Accumulating evidence may see ranolazine in routine clinical use for many conditions beyond its traditional role as an antianginal.
Collapse
Affiliation(s)
- Gopal Chandra Ghosh
- Department of Cardiology, Christian Medical College, Vellore, Tamil Nadu, India
| | - Raktim Kumar Ghosh
- MetroHealth Medical Center, Case Western Reserve University, Heart and Vascular Institute, Cleveland, OH, USA
| | | | - Krishnarpan Chatterjee
- Department of Cardiology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Ashish Aneja
- MetroHealth Medical Center, Case Western Reserve University, Heart and Vascular Institute, Cleveland, OH, USA
| |
Collapse
|
8
|
Cappetta D, Esposito G, Coppini R, Piegari E, Russo R, Ciuffreda LP, Rivellino A, Santini L, Rafaniello C, Scavone C, Rossi F, Berrino L, Urbanek K, De Angelis A. Effects of ranolazine in a model of doxorubicin-induced left ventricle diastolic dysfunction. Br J Pharmacol 2017; 174:3696-3712. [PMID: 28320043 DOI: 10.1111/bph.13791] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 03/07/2017] [Accepted: 03/09/2017] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND PURPOSE Doxorubicin is a highly effective anticancer drug, but its clinical application is hampered by cardiotoxicity. Asymptomatic diastolic dysfunction can be the earliest manifestation of doxorubicin cardiotoxicity. Therefore, a search for therapeutic intervention that can interfere with early manifestations and possibly prevent later development of cardiotoxicity is warranted. Increased doxorubicin-dependent ROS may explain, in part, Ca2+ and Na+ overload that contributes to diastolic dysfunction and development of heart failure. Therefore, we tested whether the administration of ranolazine, a selective blocker of late Na+ current, immediately after completing doxorubicin therapy, could affect diastolic dysfunction and interfere with the progression of functional decline. EXPERIMENTAL APPROACH Fischer 344 rats received a cumulative dose of doxorubicin of 15 mg·kg-1 over a period of 2 weeks. After the assessment of diastolic dysfunction, the animals were treated with ranolazine (80 mg·kg-1 , daily) for the following 4 weeks. KEY RESULTS While diastolic and systolic function progressively deteriorated in doxorubicin-treated animals, treatment with ranolazine relieved diastolic dysfunction and prevented worsening of systolic function, decreasing mortality. Ranolazine lowered myocardial NADPH oxidase 2 expression and oxidative/nitrative stress. Expression of the Na+ /Ca2+ exchanger 1 and Nav 1.5 channels was reduced and of the sarcoplasmic/endoplasmic reticulum Ca2+ -ATPase 2 protein was increased. In addition, ranolazine lowered doxorubicin-induced hyper-phosphorylation and oxidation of Ca2+ /calmodulin-dependent protein kinase II, and decreased myocardial fibrosis. CONCLUSIONS AND IMPLICATIONS Ranolazine, by the increased Na+ influx, induced by doxorubicin, altered cardiac Ca2+ and Na+ handling and attenuated diastolic dysfunction induced by doxorubicin, thus preventing the progression of cardiomyopathy. LINKED ARTICLES This article is part of a themed section on New Insights into Cardiotoxicity Caused by Chemotherapeutic Agents. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.21/issuetoc.
Collapse
Affiliation(s)
- Donato Cappetta
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Grazia Esposito
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Raffaele Coppini
- Department of Neuroscience, Drug Research and Child's Health (NeuroFarBa), Division of Pharmacology, University of Florence, Florence, Italy
| | - Elena Piegari
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Rosa Russo
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Loreta Pia Ciuffreda
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Alessia Rivellino
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Lorenzo Santini
- Department of Neuroscience, Drug Research and Child's Health (NeuroFarBa), Division of Pharmacology, University of Florence, Florence, Italy
| | - Concetta Rafaniello
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Cristina Scavone
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Francesco Rossi
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Liberato Berrino
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Konrad Urbanek
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Antonella De Angelis
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
9
|
Deidda M, Madonna R, Mango R, Pagliaro P, Bassareo PP, Cugusi L, Romano S, Penco M, Romeo F, Mercuro G. Novel insights in pathophysiology of antiblastic drugs-induced cardiotoxicity and cardioprotection. J Cardiovasc Med (Hagerstown) 2017; 17 Suppl 1 Special issue on Cardiotoxicity from Antiblastic Drugs and Cardioprotection:e76-e83. [PMID: 27755245 DOI: 10.2459/jcm.0000000000000373] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Despite advances in supportive and protective therapy for myocardial function, heart failure caused by various clinical conditions, including cardiomyopathy due to antineoplastic therapy, remains a major cause of morbidity and mortality. Because of the limitations associated with current therapies, investigators have been searching for alternative treatments that can effectively repair the damaged heart and permanently restore its function. Damage to the heart can result from both traditional chemotherapeutic agents, such as anthracyclines, and new targeted therapies, such as trastuzumab. Because of this unresolved issue, investigators are searching for alternative therapeutic strategies. In this article, we present state-of-the-art technology with regard to the genomic and epigenetic mechanisms underlying cardiotoxicity and cardioprotection, the role of anticancer in influencing the redox (reduction/oxidation) balance and the function of stem cells in the repair/regeneration of the adult heart. These findings, although not immediately transferable to clinical applications, form the basis for the development of personalized medicine based on the prevention of cardiotoxicity with the use of genetic testing. Proteomics, metabolomics and investigations on reactive oxygen species-dependent pathways, particularly those that interact with the production of NO and energy metabolism, appear to be promising for the identification of early markers of cardiotoxicity and for the development of cardioprotective agents. Finally, autologous cardiac stem and progenitor cells may represent future contributions in the field of myocardial protection and recovery in the context of antiblastic therapy.
Collapse
Affiliation(s)
- Martino Deidda
- aDepartment of Medical Sciences 'M. Aresu', University of Cagliari, Cagliari bInstitute of Cardiology, Center of Excellence on Aging, 'G. d'Annunzio' University, Chieti cDepartment of Systems Medicine, University of Rome 'Tor Vergata', Rome dDepartment of Clinical and Biological Sciences, University of Turin, Orbassano eDepartment of Clinical Medicine, Public Health, Life and Environment Sciences, University of L'Aquila, L'Aquila, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Szalontay L, Shad A. Treatment Effects and Long-Term Management of Sarcoma Patients and Survivors. Sarcoma 2017. [DOI: 10.1007/978-3-319-43121-5_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
11
|
Shahzad MA, Ishtiaq R, Zahid U, Anwer F. Successful recovery and allogeneic stem cell transplant following chemotherapy-induced severe cardiomyopathy: literature review of management and prognostic factors. BMJ Case Rep 2016; 2016:bcr-2016-217210. [PMID: 27852680 DOI: 10.1136/bcr-2016-217210] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Chemotherapy-induced cardiomyopathy is one of the major possible hazards that can result from potential cardiotoxic agents while treating cancer. Prognostic risk factors include the rate of drug administration, history of hypertension, female gender, extremes of age, previous history of mediastinal irradiation, cumulative dose and pre-existing heart disease. Close monitoring of the patients, timely diagnosis, use of well-known biomarkers including cardiac troponins, NT-ProBNP and imaging studies like 2D Echo or cardiac MRI are essential. Emerging biomarkers include carbonyl reductases (CBR1 and CBR3), aldo-keto reductases (AKR, type 1A1, 1C3, 7A2) and topoisomerase2β (Top2β). β blockers and ACE inhibitors have not only been shown to slow down the progression of cardiac dysfunction but also produce symptomatic improvement. Our case report describes a patient with acute myeloblastic leukaemia who developed severe cardiomyopathy acutely after starting the anthracycline-based regimen. Nevertheless, with timely intervention her symptoms improved and subsequently she successfully received allogeneic stem cell transplantation.
Collapse
Affiliation(s)
- Muhammad Asim Shahzad
- Department of General Internal Medicine, University of Arizona Medical Center-University Campus, Tucson, Arizona, USA
| | - Rizwan Ishtiaq
- Department of General Internal Medicine, University of Arizona Medical Center-University Campus, Tucson, Arizona, USA
| | - Umar Zahid
- Department of General Internal Medicine, University of Arizona Medical Center-University Campus, Tucson, Arizona, USA
| | - Faiz Anwer
- Department of Hematology Oncology, University of Arizona Medical Center, Tucson, Arizona, USA
| |
Collapse
|
12
|
Role of Ranolazine in cardiovascular disease and diabetes: Exploring beyond angina. Int J Cardiol 2016; 227:556-564. [PMID: 27838121 DOI: 10.1016/j.ijcard.2016.10.102] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 07/25/2016] [Accepted: 10/30/2016] [Indexed: 12/11/2022]
Abstract
Ranolazine was FDA approved for chronic angina in 2006. Since then, there has been extensive research involving this drug. The mechanism of action, debatable at the time of approval, has been demonstrated. Ranolazine acts via inhibition of late sodium channel current in the myocardium. This acts by lowering abnormally high cytosolic calcium levels. Other possible clinical applications of Ranolazine have also been explored. Out of many lines of investigation, its effects in atrial fibrillation, especially post-CABG and recurrent atrial fibrillation show promise. It has also shown definite HbA1c lowering effects when used in diabetics with coronary artery disease. Other possible indications for the drug include pulmonary arterial hypertension, diastolic dysfunction and chemotherapy-induced cardiotoxicity. This review aims to summarize major research regarding Ranolazine in potential applications beyond chronic angina. There are few dedicated large, randomized, phase III trials exploring the newer effects of Ranolazine. There are a few such trials underway, but more are needed.
Collapse
|
13
|
De Angelis A, Cappetta D, Piegari E, Rinaldi B, Ciuffreda LP, Esposito G, Ferraiolo FAV, Rivellino A, Russo R, Donniacuo M, Rossi F, Urbanek K, Berrino L. Long-term administration of ranolazine attenuates diastolic dysfunction and adverse myocardial remodeling in a model of heart failure with preserved ejection fraction. Int J Cardiol 2016; 217:69-79. [DOI: 10.1016/j.ijcard.2016.04.168] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 04/15/2016] [Accepted: 04/30/2016] [Indexed: 12/19/2022]
|
14
|
Zou D, Geng N, Chen Y, Ren L, Liu X, Wan J, Guo S, Wang S. Ranolazine improves oxidative stress and mitochondrial function in the atrium of acetylcholine-CaCl2 induced atrial fibrillation rats. Life Sci 2016; 156:7-14. [DOI: 10.1016/j.lfs.2016.05.026] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 05/08/2016] [Accepted: 05/17/2016] [Indexed: 12/19/2022]
|
15
|
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: 4.5] [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
|
16
|
De Angelis A, Urbanek K, Cappetta D, Piegari E, Ciuffreda LP, Rivellino A, Russo R, Esposito G, Rossi F, Berrino L. Doxorubicin cardiotoxicity and target cells: a broader perspective. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2016; 2:2. [PMID: 33530140 PMCID: PMC7837148 DOI: 10.1186/s40959-016-0012-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 02/18/2016] [Indexed: 12/12/2022]
Abstract
The cardiotoxicity of doxorubicin is becoming an interdisciplinary point of interest given a growing population of cancer survivors. The complex and not completely understood pathogenesis of this complication makes difficult to design successful preventive or curative measures. Although cardiomyocyte has been considered a classical cellular target, other cells including various types of undifferentiated cells are involved in myocardial homeostasis. Such perspective may shed light on previously unrecognized aspects of cardiotoxicity and promote new experimental and clinical cardioprotective strategies. In this review, different cellular targets of doxorubicin are discussed with the focus on cardiac progenitor cells, oxidative stress, DNA damage, senescence and apoptosis all of which contribute to their compromised functional properties.
Collapse
Affiliation(s)
- Antonella De Angelis
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, via Costantinopoli 16, 80138 Naples, Italy
| | - Konrad Urbanek
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, via Costantinopoli 16, 80138 Naples, Italy
| | - Donato Cappetta
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, via Costantinopoli 16, 80138 Naples, Italy
| | - Elena Piegari
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, via Costantinopoli 16, 80138 Naples, Italy
| | - Loreta Pia Ciuffreda
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, via Costantinopoli 16, 80138 Naples, Italy
| | - Alessia Rivellino
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, via Costantinopoli 16, 80138 Naples, Italy
| | - Rosa Russo
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, via Costantinopoli 16, 80138 Naples, Italy
| | - Grazia Esposito
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, via Costantinopoli 16, 80138 Naples, Italy
| | - Francesco Rossi
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, via Costantinopoli 16, 80138 Naples, Italy
| | - Liberato Berrino
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, via Costantinopoli 16, 80138 Naples, Italy
| |
Collapse
|
17
|
Mihos CG, Krishna RK, Kherada N, Larrauri-Reyes M, Tolentino A, Santana O. The use of ranolazine in non-anginal cardiovascular disorders: A review of current data and ongoing randomized clinical trials. Pharmacol Res 2015; 103:49-55. [PMID: 26546970 DOI: 10.1016/j.phrs.2015.10.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 10/05/2015] [Accepted: 10/23/2015] [Indexed: 12/19/2022]
Abstract
Ranolazine has characteristic properties of a selective inhibitor of the inward sodium current. It is primarily indicated as an anti-anginal agent in patients with coronary artery disease and chronic stable angina. Recently, ranolazine has been noted to possibly impart beneficial effects in various other cardiac conditions, including new-onset, paroxysmal, and chronic atrial fibrillation, post-operative atrial fibrillation, ventricular arrhythmias, post-revascularization coronary artery disease, chemotherapeutic cardiotoxicity, and diastolic and microvascular dysfunction. Herein, we present a review of the current clinical evidence describing the adjunctive or synergistic effects of ranolazine in non-angina related cardiovascular disorders, and include a discussion of the ongoing randomized trials investigating the therapeutic potential of ranolazine in a variety of cardiovascular diseases.
Collapse
Affiliation(s)
- Christos G Mihos
- Cardiac Ultrasound Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Rama K Krishna
- Columbia University Division of Cardiology, Mount Sinai Heart Institute, Miami Beach, FL, United States
| | - Nisharahmed Kherada
- Columbia University Division of Cardiology, Mount Sinai Heart Institute, Miami Beach, FL, United States
| | - Maiteder Larrauri-Reyes
- Columbia University Division of Cardiology, Mount Sinai Heart Institute, Miami Beach, FL, United States
| | - Alfonso Tolentino
- Columbia University Division of Cardiology, Mount Sinai Heart Institute, Miami Beach, FL, United States
| | - Orlando Santana
- Columbia University Division of Cardiology, Mount Sinai Heart Institute, Miami Beach, FL, United States.
| |
Collapse
|
18
|
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: 81] [Impact Index Per Article: 9.0] [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
|
19
|
Cancer multidrug resistance-targeted therapy in both cancer and cardiovascular system with cardiovascular drugs. Int J Cardiol 2014; 176:1306-8. [PMID: 25131921 DOI: 10.1016/j.ijcard.2014.07.158] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Accepted: 07/27/2014] [Indexed: 02/06/2023]
|