Elsevier

Life Sciences

Volume 232, 1 September 2019, 116526
Life Sciences

Review article
Role of oxidative stress in cardiotoxicity of antineoplastic drugs

https://doi.org/10.1016/j.lfs.2019.06.001Get rights and content

Abstract

Tumors and heart disease are two of the leading causes of human death. With the development of anti-cancer therapy, the survival rate of cancer patients has been significantly improved. But at the same time, the incidence of cardiovascular adverse events caused by cancer treatment has also been considerably increased, such as arrhythmia, left ventricular (LV) systolic and diastolic dysfunction, and even heart failure (HF), etc., which seriously affects the quality of life of cancer patients. More importantly, the occurrence of adverse events may lead to the adjustment or the cessation of anti-cancer treatment, which affects the survival rate of patients. Understanding the mechanism of cardiotoxicity (CTX) induced by antineoplastic drugs is the basis of adequate protection of the heart without impairing the efficacy of antineoplastic therapy. Based on current research, a large amount of evidence has shown that oxidative stress (OS) plays an essential role in CTX induced by antineoplastic drugs and participates in its toxic reaction directly and indirectly. Here, we will review the mechanism of action of OS in cardiac toxicity of antineoplastic drugs, to provide new ideas for researchers, and provide further guidance for clinical prevention and treatment of cardiac toxicity of anti-tumor drugs in the future.

Introduction

Cancer is the leading cause of death, shortening life expectancy, and ranking first or second in most countries, according to statistical data from the World Health Organization (WHO) in 2015 [1]. The World Cancer Report 2014, released in 2015, represents a series of the volume that began in 2003 when it was estimated that 5.3 million men and 4.7 million women had malignant tumors each year, of which 6.2 million died of the disease. In 2014, it was determined that 14.1 million people have cancer each year [2]. With the development of medical research and modern equipment, the methods and techniques of tumor therapy have been greatly improved, and the survival rate of cancer patients has been significantly increased [3]. But at the same time, a variety of treatment methods can also lead to multiple complications. The cardiotoxicity (CTX) is not only the most critical adverse reaction of antineoplastic drugs but also the primary cause of increased mortality of tumor survivors, which directly affects the efficacy of anti-tumor drugs [4]. Currently, the antineoplastic drugs, including anthracyclines(ANTs), alkylating agents, anti-microtubules, antimetabolites, metal platinum drugs, and new molecular targeted drugs all have certain CTX [5,6].

The CTX of antineoplastic drugs has become a “heart disease” for oncologists. Therefore, “cardio-oncology” has attracted a lot of attention of scholars as a hot issue in the field of cancer and cardiovascular disease (CVD). In recent years, the mechanism of CTX induced by antineoplastic drugs is still not completely clear [7], but the relationship between oxidative stress (OS) and CTX has attracted more and more attention of scholars. A large number of data have confirmed that OS is closely related to CTX induced by antineoplastic drugs [8]. Therefore, this paper reviews the role of OS in CTX of antineoplastic drugs.

Section snippets

Cardiotoxicity of antineoplastic drugs

The CTX of antineoplastic drugs mainly includes myocardial damage, hypertension, left ventricular (LV) systolic and diastolic dysfunction, heart failure(HF), thrombosis, pericardial disease, myocardial ischemia, arrhythmia and vasospasm [5,[8], [9], [10]]. The exact definition of CTX of tumor therapy is: (1) cardiomyopathy with reduced left ventricular ejection fraction(LVEF), which is characterized by decreased overall function or decreased ventricular septal motion; (2) symptoms associated

Oxidative stress

In cells and tissues, oxidative stress (OS) is caused by the imbalance between the production and accumulation of reactive oxygen species (ROS) and the ability of biological systems to detoxify these products [16]. ROS are normally generated as by-products of oxygen metabolism, or by nicotinamide adenine dinucleotide phosphate (NAPDH) oxidase and xanthine oxidase(XOR), although they could be formed by exogenous stimuli such as UV light, etc. [[16], [17], [18]]. So in other words, OS is a state

Antineoplastic drugs cardiotoxicity and oxidative stress

The clinical manifestations of CTX induced by antineoplastic drugs are complex and diverse, and their mechanisms of action are different [36,37]. Clinical manifestations including arrhythmia, myocardial ischemia, coronary artery disease(CAD), abnormal blood pressure, myocardial dysfunction, ECG and myocardial enzyme abnormalities, and even develop into HF [8,38]. Traditionally, there are two main anticancer drugs causing CTX: one is conventional chemotherapy drugs, including ANTs (DOX,

Conclusions and perspective

Current anti-tumor therapy has dramatically improved the survival of cancer patients, but whether ANTs, alkylating agents, antimetabolites, anti-microtubules, platinum drugs or new molecular targeted medicines have brought CTX [8,14,39]. OS is directly or indirectly involved in CTX induced by different anticancer drugs [8,22]. ANTs produce excessive ROS through various pathways, which leads to oxidative modification of cell macromolecules, induces mitochondrial dysfunction, inhibits protein

Declaration of Competing Interest

There is no conflict of interest among the authors.

Acknowledgements

This work was supported by Young Talent Lifting Project of China Association of Chinese Medicine (no. CACM-2018-QNRC2-B04).

References (117)

  • M. Panayiotidis

    Reactive oxygen species (ROS) in multistage carcinogenesis

    Cancer Lett.

    (2008)
  • L. Zhao et al.

    MicroRNA-140-5p aggravates doxorubicin-induced cardiotoxicity by promoting myocardial oxidative stress via targeting Nrf2 and Sirt2

    Redox Biol.

    (2018)
  • M. Yoshida et al.

    Chronic doxorubicin cardiotoxicity is mediated by oxidative DNA damage-ATM-p53-apoptosis pathway and attenuated by pitavastatin through the inhibition of Rac1 activity

    J. Mol. Cell. Cardiol.

    (2009)
  • S. Patane

    Cardiotoxicity: anthracyclines and long term cancer survivors

    Int. J. Cardiol.

    (2014)
  • D. Nikitovic et al.

    Anthracycline-dependent cardiotoxicity and extracellular matrix remodeling

    Chest

    (2014)
  • P. Xia et al.

    Inhibition of cyclin-dependent kinase 2 protects against doxorubicin-induced cardiomyocyte apoptosis and cardiomyopathy

    J. Biol. Chem.

    (2018)
  • J. Yu et al.

    Recent progress in doxorubicin-induced cardiotoxicity and protective potential of natural products

    Phytomedicine

    (2018)
  • A. Pugazhendhi et al.

    Toxicity of Doxorubicin (Dox) to different experimental organ systems

    Life Sci.

    (2018)
  • M.L. Circu et al.

    Reactive oxygen species, cellular redox systems, and apoptosis

    Free Radic. Biol. Med.

    (2010)
  • P. Vejpongsa et al.

    Prevention of anthracycline-induced cardiotoxicity: challenges and opportunities

    J. Am. Coll. Cardiol.

    (2014)
  • A. Cascales et al.

    Clinical and genetic determinants of anthracycline-induced cardiac iron accumulation

    Int. J. Cardiol.

    (2012)
  • G.J. Dugbartey et al.

    An integrative view of cisplatin-induced renal and cardiac toxicities: molecular mechanisms, current treatment challenges and potential protective measures

    Toxicology

    (2016)
  • N.G. Kounis et al.

    Cisplatin-induced bradycardia: cardiac toxicity or cardiac hypersensitivity and Kounis syndrome?

    Int. J. Cardiol.

    (2016)
  • J. Alexandre et al.

    Anticancer drug-induced cardiac rhythm disorders: current knowledge and basic underlying mechanisms

    Pharmacol. Ther.

    (2018)
  • S. Patane

    Cardiotoxicity: cisplatin and long-term cancer survivors

    Int. J. Cardiol.

    (2014)
  • G. Rosic et al.

    The effects of N-acetylcysteine on cisplatin-induced changes of cardiodynamic parameters within coronary autoregulation range in isolated rat hearts

    Toxicol. Lett.

    (2016)
  • S.A. Abdellatief et al.

    Ameliorative effect of parsley oil on cisplatin-induced hepato-cardiotoxicity: a biochemical, histopathological, and immunohistochemical study

    Biomed. Pharmacother.

    (2017)
  • U. Demkow et al.

    Cardiotoxicity of cisplatin-based chemotherapy in advanced non-small cell lung cancer patients

    Respir. Physiol. Neurobiol.

    (2013)
  • L. Bhatt et al.

    Mangiferin protects rat myocardial tissue against cyclophosphamide induced cardiotoxicity

    J. Ayurveda Integr. Med.

    (2017)
  • W. Liu et al.

    Aldehyde dehydrogenase 2 activation ameliorates cyclophosphamide-induced acute cardiotoxicity via detoxification of toxic aldehydes and suppression of cardiac cell death

    J. Mol. Cell. Cardiol.

    (2018)
  • I. Depetris et al.

    Fluoropyrimidine-induced cardiotoxicity

    Crit. Rev. Oncol. Hematol.

    (2018)
  • A. Polk et al.

    Cardiotoxicity in cancer patients treated with 5-fluorouracil or capecitabine: a systematic review of incidence, manifestations and predisposing factors

    Cancer Treat. Rev.

    (2013)
  • O. Abdel-Rahman

    5-fluorouracil-related cardiotoxicity; findings from five randomized studies of 5-fluorouracil-based regimens in metastatic colorectal cancer

    Clin. Colorectal Cancer

    (2019)
  • M. Lamberti et al.

    A mechanistic study on the cardiotoxicity of 5-fluorouracil in vitro and clinical and occupational perspectives

    Toxicol. Lett.

    (2014)
  • K. Chatterjee et al.

    Acute vincristine pretreatment protects adult mouse cardiac myocytes from oxidative stress

    J. Mol. Cell. Cardiol.

    (2007)
  • K. Rtibi et al.

    Vinblastine, an anticancer drug, causes constipation and oxidative stress as well as others disruptions in intestinal tract in rat

    Toxicol. Rep.

    (2017)
  • L. Pentassuglia et al.

    Inhibition of ErbB2/neuregulin signaling augments paclitaxel-induced cardiotoxicity in adult ventricular myocytes

    Exp. Cell Res.

    (2007)
  • F. Bray et al.

    Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries

    CA Cancer J. Clin.

    (2018)
  • A. Godishala et al.

    Cardioprotection in the modern era of cancer chemotherapy

    Cardiol. Rev.

    (2018)
  • L. Prezioso et al.

    Cancer treatment-induced cardiotoxicity: a cardiac stem cell disease?

    Cardiovasc. Hematol. Agents Med. Chem.

    (2010)
  • G. Varricchi et al.

    Antineoplastic drug-induced cardiotoxicity: a redox perspective

    Front. Physiol.

    (2018)
  • J. Duan et al.

    Anticancer drugs-related QTc prolongation, torsade de pointes and sudden death: current evidence and future research perspectives

    Oncotarget

    (2018)
  • M.C. Honigberg et al.

    Pregnancy among survivors of childhood cancer: cardiovascular considerations

    Curr. Treat. Options Cardiovasc. Med.

    (2018)
  • R. Moudgil et al.

    Mechanisms of cardiotoxicity of cancer chemotherapeutic agents: cardiomyopathy and beyond

    Can. J. Cardiol.

    (2016)
  • J.L. Zamorano et al.

    2016 ESC position paper on cancer treatments and cardiovascular toxicity developed under the auspices of the ESC committee for practice guidelines: the task force for cancer treatments and cardiovascular toxicity of the European Society of Cardiology (ESC)

    Eur. Heart J.

    (2016)
  • C.G. Tocchetti et al.

    From molecular mechanisms to clinical management of antineoplastic Drug-Induced cardiovascular toxicity: a translational overview

    Antioxid. Redox Signal.

    (2019)
  • G. Riccio et al.

    Trastuzumab and target-therapy side effects: is still valid to differentiate anthracycline type I from type II cardiomyopathies?

    Hum. Vaccin. Immunother.

    (2016)
  • G. Pizzino et al.

    Oxidative stress: harms and benefits for human health

    Oxidative Med. Cell. Longev.

    (2017)
  • A. Konior et al.

    NADPH oxidases in vascular pathology

    Antioxid. Redox Signal.

    (2014)
  • M. Yisireyili et al.

    Xanthine oxidase inhibition by febuxostat attenuates stress-induced hyperuricemia, glucose dysmetabolism, and prothrombotic state in mice

    Sci. Rep.

    (2017)
  • Cited by (21)

    • Sinus arrhythmia caused by pemetrexed with carboplatin combination: A case report

      2022, Heliyon
      Citation Excerpt :

      Cisplatin is also speculated to direct ROS attacks on vascular endothelium, inducing platelet activation and aggregation to promote thrombosis [25]. Prior studies have reported sporadic instances of cardiotoxicity associated with antineoplastic drugs likely arising due to the induction of ischemia as a consequence of coronary artery spasms [26]. Herein, it is important to note that the patient did not exhibit sustained cardiotoxicity during the early stages of pemetrexed and carboplatin treatment, suggesting that the cumulative accumulation of these chemotherapeutic drugs for prolonged chemotherapeutic treatment is likely to have contributed to the development of sinus arrhythmia.

    • Biometabolites of Tamarindus indica play a remarkable cardioprotective role as a functional food in doxorubicin-induced cardiotoxicity models

      2022, Journal of Functional Foods
      Citation Excerpt :

      On the other hand, our present study is bringing us closer to these objectives by providing new insights into the disease's breadth, pathophysiology, and therapy. Several chronic diseases, including CVD, are caused by oxidative stress, which occurs when the body's production of free radicals and reactive oxygen species (ROS) increases (Zhang, 2019). The use of doxorubicin-induced cardio protective characteristics of numerous medicinal plants has been reported in many literatures.

    View all citing articles on Scopus
    View full text