Open access peer-reviewed chapter

Doxorubicin-Induced Cardiotoxicity

Written By

Hongxin Zhu

Submitted: 11 March 2018 Reviewed: 18 May 2018 Published: 05 November 2018

DOI: 10.5772/intechopen.78791

From the Edited Volume

Cardiotoxicity

Edited by Wenyong Tan

Chapter metrics overview

1,308 Chapter Downloads

View Full Metrics

Abstract

Doxorubicin (DOX) is one of the most effective antineoplastic drugs. However, its clinical use is largely limited by potential dose-dependent cardiotoxicity. To date, the mechanisms of DOX-induced cardiotoxicity remains incompletely understood. More importantly, no efficient therapeutic strategy is available to counteract DOX-induced cardiomyopathy, underscoring the importance of the prevention of this disease. In this chapter, we first describe the pathophysiology of DOX-induced cardiotoxicity. We then update the findings of molecular biology of DOX-induced cardiomyopathy including molecular mechanisms, established and putative biomarkers for early diagnosis, and potential genetic factors for prediction of susceptibility. Finally, we introduce a number of pharmaceutical measures and practical lifestyle modifications for the prevention of this disease.

Keywords

  • doxorubicin
  • cardiotoxicity
  • cardiac function
  • oxidative stress
  • iron accumulation
  • topoisomerase
  • autophagy
  • mitochondria
  • inflammation
  • calcium
  • cell death
  • microRNA
  • polymorphisms
  • antioxidant
  • exercise
  • fasting

1. Introduction

Doxorubicin (DOX), an anthracycline antibiotic produced by the fungus Streptomyces peucetius, has been proved to be one of the most effective drugs for the treatment of solid tumor and haemotological malignancies. However, the clinical use of DOX is limited by potential dose-dependent cardiotoxicity. Incidences of progressive congestive heart failure were approximately 5, 16, 26 and 48% in patients who had received a cumulative dose of 400, 500, 550 and 700 mg/m2 of DOX, respectively [1]. DOX-induced cardiotoxicity can be acute or chronic. Acute DOX cardiotoxicity occurs within several days after administration of the drug, while chronic DOX cardiotoxicity takes place months or even years after use of DOX [2]. However, the biological mechanisms underlying DOX cardiotoxicity is not fully understood, although multiple factors have been suggested. As a consequence, no efficacious therapeutic strategies are available to cure DOX cardiotoxicity. Therefore, the prevention of DOX cardiotoxicity is crucial for cancer patients. Currently, several pharmaceutical strategies have been used or tested clinically to prevent DOX cardiotoxicity. In addition, a number of nonpharmacological strategies have shown promising results in preclinical studies. To accomplish more successful prevention or intervention of DOX cardiotoxicity, efforts should be exerted on identification of the susceptible population on the basis of genetic variants or early diagnosis of this disease taking advantage of biomarkers. In this chapter, we first describe morphological and functional characteristics of the heart in DOX cardiotoxicity. We then update the findings regarding molecular biology of DOX cardiotoxicity. Finally, we introduce several promising pharmacological strategies and lifestyle modifications for the prevention of DOX cardiotoxicity.

Advertisement

2. Morphological and functional characterization

The earliest alteration of the heart in DOX cardiotoxicity is calpain-dependent degradation of a giant cardiac structural protein titin, which may predispose the heart to diastolic dysfunction [3]. Histological changes include cardiomyocyte vacuolar degeneration and myofibrillar disarray [4]. In addition, fibrosis is markedly increased in both interstitial area of myocardium and perivascular area in animal models of chronic DOX-induced cardiotoxicity [5]. At the ultrastructural level, DOX-induced cardiac damage is characterized by dilatation of sarcoplasmic reticulum, loss of the Z-band, myofibrillar dropout, marked accumulation of cytoplasmic vacuoles, damaged mitochondria, and increased numbers of autophagic vacuoles [6, 7]. These changes result in cardiomyocyte dysfunction and cell death via necrosis or apoptosis. Cell death and fibrosis lead to compromised cardiac function in DOX-induced cardiomyopathy. DOX cardiotoxicity can be diagnosed if the patients receiving DOX treatment show signs and symptoms of congestive heart failure. However, DOX cardiotoxicity is usually diagnosed on the basis of left ventricular cardiac function. Three types of criteria are widely used to diagnose DOX cardiotoxicity: (i) the left ventricular ejection fraction (LVEF) is reduced by 20% to a value >50%, (ii) the LVEF is reduced by 10% to a value <50%, and (iii) the LVEF is reduced by >10 points to a value <50% [8].

Advertisement

3. Cellular and molecular mechanisms

The cause of DOX cardiotoxicity is multifactorial, and the precise mechanisms remain to be elucidated. Here, we describe the major mechanisms that have been suggested to contribute to DOX cardiotoxicity. It should be pointed out that the mechanisms are not mutually exclusive. As a matter of fact, most of the factors are interconnected with each other.

3.1. Oxidative stress

Oxidative stress, caused by enhanced intracellular levels of reactive oxygen species (ROS), has long been believed to be the major mediator of DOX cardiotoxicity. The major types of ROS include superoxide radical (O2−), hydrogen peroxide (H2O2), and hydroxyl free radical (HO) [9]. ROS is mainly generated through redox cycling in mitochondria [9]. However, ROS is also produced outside mitochondria by activation of pro-oxidant enzymes such as NADPH oxidase and xanthine oxidases [10]. Low level of ROS functions as signaling molecules and cell defense system. The cells have efficient antioxidant defense system to eliminate overproduced ROS and maintain ROS to physiological levels [11]. However, if the balance between ROS production and antioxidant system is disrupted in favor of ROS production, then oxidative stress occurs, which triggers a number of deleterious events including DNA damage, mitochondrial dysfunction, cell death, disrupted cellular calcium homeostasis, attenuated protein synthesis, defect in protein quality control, and mitochondrial quality control [12]. After DOX treatment, DOX is preferentially accumulated in mitochondria. As a potent electron acceptor, DOX promotes ROS generation and damages the activities of antioxidant enzymes, shifting the balance between pro-oxidant and antioxidant to the former, leading to elevated ROS levels. Excessive ROS is capable of damaging mitochondria, which in turn, produces more ROS, forming a vicious cycle called ROS-induced ROS release [13]. Given that the cardiomyoyctes are exceptionally rich in mitochondria, DOX is especially harmful to the heart. At the molecular level, the harmful effects of DOX-induced ROS are exerted primarily by its direct damage to mitochondrial genome, RNA, proteins and lipids [12]. In addition, enhanced ROS also participates in cellular signaling involved in detrimental events such as DNA damage and cell death [14].

3.2. Iron accumulation

Following DOX administration, DOX cardiotoxicity occurs through iron accumulation in mitochondria. Cardiac specific over-expression of ABCB8, a mitochondrial inner membrane protein involved in iron export, reduced iron accumulation in mitochondria and mitigated DOX cardiotoxicity [15]. Dexrazoxane, a drug approved by FDA to prevent DOX cardiotoxicity, decreased iron accumulation and ameliorate DOX-induced cardiac injuries in mice. In addition, patients with DOX cardiotoxicity showed higher levels of mitochondrial iron compared with patients with other types of cardiomyopathy or patients with normal cardiac function [15]. These studies provide convincing evidences demonstrating that iron accumulation is one of the major mechanisms involved in DOX cardiotoxicity. However, the underlying mechanisms that iron overload causes DOX cardiotoxicity remain to be clarified. Although several lines of evidences point to enhanced ROS generation by iron accumulation, a number of antioxidants fail to protect DOX cardiotoxicity in clinical settings, suggesting that other unidentified mechanisms are responsible for iron accumulation-mediated cardiac damage in DOX cardiotoxicity [16].

3.3. Topoisomerase IIβ

Type II topoisomerases (Top II) is an enzyme that generates DNA double-strand breaks, which is crucial to control the conformational changes of DNA and the entire chromosome. Mammalian cells consist of two types of Top II isoenzymes, Top IIα and Top IIβ. Top IIα is only expressed in proliferating cells, while Top-IIβ is ubiquitously expressed including postmitotic cells such as adult cardiomyocytes [17]. The antitumor activity of DOX is achieved through the formation of Top II-DOX-DNA ternary complex (also called the cleavage complex), which increases Top II-DNA complexes and consequent DNA double-strand breaks [17]. In cardiomyocyte, Top IIβ is targeted by DOX, and the increased Top IIβ DNA cleavage complex induces DNA damage, which in turn, leads to cell death. Cardiomyocyte-specific depletion of Top IIβ conferred protection against DOX-induced DNA double-strand breaks, transcriptome changes, and heart failure [18, 19]. These data suggest that Top IIβ in cardiomyocytes plays a major role in mediating DOX-induced cardiotoxicity.

3.4. Macroautophagy dysregulation

Macroautophagy (hereafter referred to as autophagy) is a conserved pathway delivering cytoplasmic contents to lysosome for degradation and recycling [20]. Basal level of autophagy in the heart plays an essential role in the maintenance of cardiac structure and function by removing damaged protein and organelles such as mitochondria [21]. Autophagy can be either activated or suppressed in pathological conditions [22]. The significance of autophagy activation can be either beneficial or detrimental depending upon pathological settings [22]. Recent studies have shown that autophagy is dysregulated after DOX treatment in animals. However, it is controversial whether autophagy is activated or suppressed. There are studies showing that DOX treatment activates autophagy in the heart or cardiomyocytes [23, 24, 25, 26], while others have shown conflicting results [7, 27, 28, 29, 30]. Moreover, the significance of autophagy in DOX cardiotoxicity is still on debate. Some data are in favor of beneficial effects of autophagy in DOX cardiotoxicity [23, 24, 25, 26], while others argue against it [27, 28, 29, 30]. The discrepancies may be caused by the difference in animal species, cell types, methods monitoring autophagy, means of drug administration, and dosage and duration of the drug used in these studies. More recently, we and others have shown that DOX treatment stimulated autophagy initiation, while suppressed multiple subsequent steps including autophagosome formation, autophagosome maturation and lysosomal degradation [7, 27, 29, 30]. As a consequence, the autophagic flux was attenuated in DOX-induced cardiotoxicity. Inhibition of autophagic flux using UVRAG-deficient mice exacerbated DOX-induced cardiotoxicity [30]. Conversely, enhancement of autophagic flux mitigated DOX cardiotoxicity [27, 29, 30]. In addition, suppression of autophagy initiation using beclin 1+/− mice ameliorated DOX cardiotoxicity [7]. The regulation of autophagy in the heart and its significance in cancer patients treated by DOX needs to be investigated. Moreover, the effects of autophagy modulation on cancer cells should be considered if autophagy is targeted for prevention of DOX cardiotoxicity.

3.5. Mitochondrial dysfunction

Mitochondria are the organelle that produces ATP, which plays an essential role in cell survival. Mitochondria are the major source of free radicals and as a consequence are vulnerable to damage caused by oxidative stress. It has been demonstrated that mitochondrial dysfunction is one of the mechanisms of DOX cardiotoxicity [12]. Under physiological conditions, mitochondrial quality is controlled by mitochondrial quality control system, which includes selective elimination of mitochondria by autophagy (also called mitophagy), mitochondrial biogenesis, and mitochondrial dynamics including mitochondrial fusion and fission [31].

Pink1-Parkin-mediated mitophagy is the most well-studied mechanism for mitophagy. Pink 1 is a serine/threonine kinase, which is normally localized in the inner membrane of mitochondria (IMM). However, in depolarized mitochondria, Pink 1 is unable to be translocated to IMM and is retained on the outer membrane of mitochondria (OMM), where Pink-1 undergoes autophosphorylation and is activated. The activated Pink-1 then recruits parkin, a cytosolic E3 ligase to the OMM. Parkin ubiquitinates the substrate proteins localized on the OMM and facilitates degradation of mitochondria by autophagy [32, 33]. DOX treatment has been shown to suppress Pink 1 and Parkin expression [34]. In addition, DOX enhances p53 expression, which promotes its interaction with Parkin and prevents Parkin translocation from cytoplasm to mitochondria [35]. Moreover, as aforementioned, DOX inhibits autophagic flux in the heart at multiple steps, which also attenuates mitochondrial degradation [7, 27, 29, 30]. Therefore, DOX treatment suppresses Pink 1-Parkin-mediated autophagy in the heart and promotes accumulation of damaged mitochondria. In addition to Pink 1-Parkin-meidated mitophagy, other mitochondria-localized proteins such as Nix, Bnip3, FUNDC1, and cardiolipin have been shown to interact with LC3 or LC3 homologs to mediate mitophagy [33]. However, the significance of Parkin-independent mitophagy mediated by these molecules remains to be elucidated in DOX cardiotoxicity.

Mitochondria are highly dynamic organelle, which continuously undergo fusion and fission to organize interconnecting networks to fulfill its function. Mitochondrial fusion and fission are essential for the maintenance of mitochondrial number and quality under stress conditions. Mitochondrial fusion allows the mixture of the contents from partially damaged mitochondria and healthy mitochondria to alleviate the stress. Mitochondrial fission separates mitochondria into two daughter mitochondria, which allows the biogenesis of new mitochondria and the removal of the damaged mitochondria via mitophagy [31]. Mitochondrial fusion is controlled by GTPase Mitofusin1 (MFN1), Mitofusin2 (MFN2), and optic atrophy factor 1 (OPA1). MFN1 and MFN2 are localized to the OMM, while OPA1 is an IMM protein. MFN1, MFN2, and OPA1 mediate the fusion of the OMM and IMM, respectively [31]. Mitochondrial fission is mainly regulated by Drp1, a large GTPase. Drp1 is recruited from cytoplasm to mitochondrial OMM during fission process. In mitochondrial OMM, Drp1 has four interacting partners, FIS1, Mff, Mid55, and Mid49 [31, 36]. Mitochondrial fusion and fission are well balanced to maintain mitochondrial number and quality under physiological conditions. In animal models of DOX cardiotoxicity, DOX treatment induces changes in the expression of mitochondrial fusion and fission proteins, which alters mitochondrial dynamics and contributes to apoptosis [37].

Mitochondrial biogenesis is the process of expansion of existing mitochondria or generation of new mitochondria. Mitochondrial biogenesis is tightly regulated to coordinate mitophagy, mitochondrial fusion and fission for the maintenance of mitochondrial mass and remodeling of dynamic interconnected mitochondrial network. DOX treatment impairs cardiac mitochondrial biogenesis as manifested by reduced mitochondrial DNA copy number and expression of regulating factors for mitochondrial biogenesis such as peroxisome proliferator-activated receptor gamma coactivator 1-alpha, peroxisome proliferator-activated receptor alpha, and estrogen-related receptor alpha, leading to suppression of mitochondrial metabolism and ATP synthesis [38, 39].

3.6. Inflammation

A growing body of evidences has shown that cardiac inflammation contributes to DOX cardiotoxicity. DOX treatment induces increased activity of NF-κB, a key component of innate immune system, leading to enhanced levels of pro-inflammatory cytokines including IL-1β, IL-6, and TNFα [40]. Toll-like receptors (TLRs) especially TLR2 has been considered as the major mediator to activate NF-κB [40]. DOX-induced oxidative stress and damage-associated molecular pattern molecules (DAMPs) such as HMGB-1 are responsible for the activation of TLR2 [41]. In addition to TLR2, TLR9 is capable of activating NF-κB and may be engaged in cardiac inflammation in DOX-induced cardiotoxicity [42]. It has been shown that mitochondrial DNA escaped from autophagy triggers cardiac inflammation through TLR9 activation during progression of pressure-overloaded heart failure [43]. Given that autophagic flux in the heart is impaired by therapeutic dose of DOX, it is likely that TLR-9 activation is involved in inflammatory response in DOX-induced cardiotoxicity. However, studies need to be designed to address this issue.

3.7. Abnormal intracellular calcium handling

Calcium is critical for cardiac systolic and diastolic function. Calcium regulates cardiac contraction through a process called cardiac excitation-contraction coupling (EC coupling). In this process, calcium enters cytoplasm through L-type calcium channel activates ryanodine (RyR) receptor localized on the sarcoplasmic reticulum (SR) membrane, resulting in calcium-induced calcium release in the SR. The released calcium form SR stimulates cardiomyocytes to contract. Subsequently, the cytoplasmic calcium is taken up by the sarcoendoplasmic reticulum calcium transport ATPase (SERCA2) localized on the SR membrane, resulting in reduced cytoplasmic calcium concentration and cardiomyocyte relaxation [44]. DOX regulates cytoplasmic calcium levels through several mechanisms. First, DOX is able to bind RYR2 directly and enhances its open probability [45]. Second, DOX is capable of interacting with calsequestrin, a calcium binding protein localized in SR lumen, and promotes calcium release [46]. Third, DOX elevates intracellular calcium levels by binding to SERCA2A and modify its activity [47]. Fourth, DOX induces SR calcium leakage in a CAMK II-dependent manner, leading to impaired calcium handling in cardiomyocytes [48]. Finally, oxidative stress induced by DOX amplifies RYR opening and calcium release [49]. Thus, DOX regulates calcium release from SR through both oxidant-dependent and independent mechanisms, and the abnormal calcium handling contributes to DOX cardiomyopathy.

3.8. Cell death

Numerous studies have shown that DOX induces apoptosis, which contributes to cardiotoxicity. DOX stimulates ROS generation and produces oxidative stress, which activates p53. In addition, DOX itself promotes p53 activity in the heart. p53-mediated signaling stimulates apoptotic cell death of cardiomyocytes [50, 51]. Moreover, multiple lines of evidences have suggested that mitochondrial calcium is overloaded and contributes to apoptotic cell death of cardiomyocytes in DOX cardiotoxicity. As aforementioned, DOX promotes calcium release from SR. Mitochondria, which are physically close to SR calcium release sites, uptake a portion of calcium released from SR, leading to rise in mitochondrial calcium levels. Calcium overload triggers loss of mitochondrial membrane potential, swelling of mitochondria, and ultimately rupture of OMM and leakage of cytochrome C, resulting in apoptosis of cardiomyocytes [52].

Necrotic cardiomyocyte death is also increased in DOX cardiotoxicity. Oxidative stress induced by DOX is considered as the major cause for necrosis. Oxidative stress enhances calcium release from SR and raises calcium levels in mitochondria, which induces loss of mitochondrial membrane potential, mitochondrial swelling, and ultimately mitochondrial outer membrane rupture, leading to ATP depletion [53]. In addition, oxidative stress induces mitochondrial DNA damage and mitochondrial lipid peroxidation, leading to disruption of integrity of mitochondrial structure, mitochondrial dysfunction, and ATP depletion [54]. Recently, Bnip3 has been shown to disrupt interaction of COXI and UCP3, leading to defective mitochondrial respiratory chain and cardiomyocyte necrosis in DOX cardiotoxicity [55].

Advertisement

4. Biomarkers and genetic factors

Currently, no effective therapy is available to cure DOX-induced cardiotoxicity. Thus, prevention become more important and should be primarily directed. Early detection is crucial for the prevention of irreversible cardiac damage. Traditional technology such as echocardiography, electrocardiogram, and angiography are not efficient for early detection of cardiac damage since cardiac dysfunction already occurs when diagnosis is made by means of aforementioned technology. Biochemical biomarkers are sensitive and ideal for early detection of cardiac damage. Two types of biomarkers, i.e., troponins and natriuretic peptides, have been established and are currently used in clinic for early diagnosis of DOX cardiotoxicity. In addition, other promising putative biomarkers have been tested.

4.1. Cardiac troponins and B-type natriuretic peptide

Cardiac troponins are a complex consisting of three regulatory proteins, i.e., troponin T (cTnT), troponin C (cTnC), and troponin I (cTnI) in cardiac muscle. cTnT and cTnI are well-established sensitive and specific biomarkers to detect myocardial damage caused by differential insults [56]. Both cTnI and cTnT have also been utilized in clinic to detect and predict cardiac damage caused by DOX [57, 58].

B-type natriuretic peptide (BNP) is a peptide prohormone, which is primarily produced in ventricles and brain. BNP is synthesized as pre-pro-BNP, which is cleaved to generate pro-BNP. Pro-BNP is further cleaved into a C-terminal biologically active form of BNP and N-terminal inactive form of NT-pro-NPs. Both NT-pro-NPs and BNP are secreted into serum and serve as sensitive biomarkers predictive of congestive heart failure [59, 60, 61]. Currently, NT-pro-NPs and BNP are used in clinic as indicators of early cardiac damage caused by DOX [62, 63].

4.2. MicroRNAs

MicroRNAs can become ideal clinical biomarkers due to their characteristics such as high stability, tissue specificity, and presence in body fluids [64]. Emerging evidences have indicated that alteration of certain microRNAs is associated with DOX cardiotoxicity and may be served as biomarkers. An in vitro study using human pluripotent stem cell-derived cardiomyocytes showed that a number of microRNAs, including miR-34a, miR-34b, miR-187, miR-199a, miR-199b, miR-146a, miR-15b, miR-130a, miR-214, and miR-424, were differentially expressed during and after DOX treatment [65]. However, the expression pattern of these microRNAs in animal models and patients receiving DOX treatment remains to be investigated. A study using a mouse model of DOX cardiotoxicity explored whether microRNAs including miR-208a, miR-133b, miR-146a, miR423-5p and miR-1 are suitable to predict cardiac damage in patients receiving DOX treatment. The results showed that miR-208a and miR-208b were not useful biomarkers for DOX cardiotoxicity since they were undetectable in the serum. MiR-133b, miR-146a, and miR423-5p were not appropriate biomarkers either since although detectable, no significant alterations were observed in cardiotoxic-patients compared with noncardiotoxic-patients. miR-1 was upregulated in patients suffering from cardiotoxicity compared with noncardiotoxic patients. Moreover, miR-1 expression levels were associated with changes of left ventricular ejection fraction. Therefore, miR-1 is a promising circulating biomarker for early detection of cardiac injury caused by DOX [66]. However, further studies should be developed to validate the putative diagnostic marker.

4.3. Genetic risk factors

The susceptibility to DOX cardiotoxicity is apparently patient dependent, suggestive of a role of genetic factors. To date, a number of gene polymorphisms associated with DOX cardiotoxicity have been identified. A German non-Hodgkin lymphoma study including 1697 enrolled patients has suggested that polymorphisms of the NAD(P)H oxidase were associated with DOX cardiotoxicity. Specifically, the 212A→G variant of NAD(P)H oxidase subunit NCF4 was associated with chronic DOX cardiotoxicity. The His72Tyr polymorphism in the p22phox subunit and the variant 7508T→A of the RAC2 subunit of NAD(P)H oxidase were associated with acute DOX cardiotoxicity [67]. Consistent with these findings, mice deficient for NAD(P)H oxidase activity were resistant to chronic doxorubicin treatment [67]. In the same study, Gly671Val variant of the doxorubicin efflux transporter multidrug resistance protein 1 (MRP1) and the Val1188Glu-Cys1515Tyr haplotype of MRP2 have been shown to be associated with acute DOX cardiotoxicity [67]. Polymorphisms of other genes that have been reported to be potentially associated with cardiotoxicity caused by DOX or DOX-based treatment include CBR3, CAT, ABCB1, ABCC1, ABBCC2, RAC2, GSTP1, CYBA, ABCC5, CASP3, MSH2, SLCO1A2, SLC28A3, FMO2, SPG7, SLC10A2, UGT1A6, ABCB4, SULT2B1, HFE, POR, HAS3, HNMT, SLC22A7, SLC22A17, RARG, and NOS3 [68]. Most of the candidate genes are related to cellular transport of DOX, oxidative stress, DOX metabolism, and DNA repair and replication. In a recent study involving a relatively small number of patients treated with DOX for breast cancer, 18 SNPs in nine genes in the HLA region (NFKBIL1, TNF-α, ATP6V1G2-DDX39B, MSH5, MICA, LTA, BAT1, and NOTCH4) and in the psoriasis susceptibility region of HLA-C were identified to be potentially associated with DOX cardiotoxicity, implicating an important role of dysregulation of genes involved in inflammatory disease and autoimmune disorders in DOX cardiotoxicity [69]. Polymorphisms of RAAS genes, which are useful for the prediction of congestive heart failure, were not significantly associated with DOX-induced cardiotoxicity [67]. Additional studies are required to identify and functionally validate genetic variants in DOX cardiotoxicity.

Advertisement

5. Preventive strategy

5.1. Doxorubicin dosage and administration

Given that DOX-induced cardiotoxicity is cumulative dose-dependent, the most straightforward way to prevent DOX cardiotoxicity is to reduce the dosage utilized for patients. However, lower dosage is associated with less therapeutic efficacy [70]. Thus, alternative approaches of drug administration such as continuous infusion and liposome DOX versus bolus injection are used to prevent cardiac toxicity. Continuous infusion of DOX causes significantly less injury to the heart compared to bolus doses without compromising cancer treatment efficacy. The mechanisms are due to the changes in the distribution of DOX with reducing drug concentration in the heart and no impact on drug doses in tumor tissues [71, 72, 73]. It should be pointed out that continuous infusion does not confer cardiac protection in children with acute lymphoblastic leukemia [74]. Administration of DOX by liposome encapsulation is another effective strategy to reduce cardiotoxicity. Liposomal DOX formulation is not capable of crossing the tight gap junction of endothelial cells of blood vessels in the heart. However, in tumor tissues, the vasculature is irregular and leaky, which allows the diffusion of liposomal DOX formulation [75]. In addition, the diffused DOX accumulates in the tumor tissue due to poor lymph drainage. Both lead to selective accumulation of DOX in tumor tissues. This phenomenon is known as “enhanced permeability and retention effect,” which characterizes solid tumors and is used to target tumor cells [76]. Moreover, the liposomal DOX formulations diffused into tumor tissues are prone to destabilization due to more acidic extracellular pH, release of necrotic tumor cell lipases, and inflammatory cell oxidizing agents in tumor microenvironment [76]. A number of preclinical and clinical studies have demonstrated that liposomal DOX formulation delivers relatively larger amount of DOX to tumor tissues and much less doses to the heart tissues compared to conventional DOX. Thus, the liposomal DOX formulations are more active and safer. Currently, two types of liposomal DOX formulations, i.e., pegylated (Caelyx® in Europe and Doxil® in the USA) or nonpegylated (Myocet®), have been approved as a first-line treatment for defined group of cancer patients [77]. In recent years, nanoparticle DOX delivery systems have attracted much attention due to potential increased bioavailability in tumor tissues and minimum cardiac toxicity, which hold promise as an efficient approach for the prevention of DOX cardiotoxicity [78].

DOX treatment combining with cardioprotective agents is an alternative strategy to prevent cardiotoxicity. Dexrazoxane (Zinecard, ICRF-187, ADR-529, NSC-169780), a cyclic derivative of edetic acid, is a cardioprotective agent approved by FDA to prevent DOX cardiotoxicity in the clinic [79]. The molecular mechanisms that Dexrazoxane confers cardioprotection have previously been attributed to its iron chelating capability. However, other iron chelators fail to exert preventive effects for DOX cardiotoxicity, suggesting that iron chelation is not the major molecular basis for dexrazoxane cardioprotection. It turns out that dexrazoxane interferes with Top IIβ either through promoting Top IIβ proteasomal degradation or preventing the formation of Top IIβ-DNA cleavage complex in cardiomyocytes [79]. It should be noted that coadministration of dexrazoxane may trigger secondary malignancies in cancer patients [80]. However, this issue is still controversial and requires further investigation.

5.2. Antioxidant reagents

Considering oxidative stress has been believed to be the major mediator of DOX-induced cardiotoxicity, it is reasonable to expect that coadministration of antioxidants is capable of preventing or mitigating DOX cardiotoxicity. The antioxidants reduce intracellular ROS levels through reducing ROS generation, scavenging ROS themselves, chelating irons to inhibit HO. formation or eliminating other active molecules generated in response to ROS reaction such as lipid peroxide [81]. Although antioxidants are effective in the treatment of acute DOX cardiotoxicity in animal models, Clinically relevant animal experiments and clinical trials have suggested that among a variety of antioxidant reagents, only dexrazoxane has shown definitive effect on DOX cardiotoxicity [79]. As mentioned above, dexrazoxane ameliorates DOX cardiotoxicity likely through mechanisms independent of ROS elimination [79]. Thus, it still remains unclear whether antioxidants should be given to cancer patients during or after DOX treatment to prevent cardiotoxicity. In addition, ROS generation could be the mechanism that DOX is toxic to cancer cells, antioxidant may reduce response rate for DOX in patients, although DOX may cause cytotoxicity in cancer cells through both ROS-dependent and independent mechanisms. Further study should be conducted to address these issues.

5.3. Neurohormone blockers

Neurohormone blockers such as angiotensin II-converting enzyme inhibitors and angiotensin receptor blockers have been widely utilized in clinics to treat heart failure including DOX-induced heart failure. Angiotensin receptor blockers have been shown to prevent decline of cardiac function induced by DOX in cancer patients. The preventive effect may be related to decreased generation of oxidative stress and reduced apoptosis of cardiomyocytes [82, 83]. Thus, neurohormone blockers may be used in combination with DOX to prevent cardiac toxicity.

5.4. Exercise

In addition to pharmaceutical measure, lifestyle modifications are promising alternative strategies to counteract DOX-induced cardiomyopathy since it is practical to be introduced to patients. Several types of exercise such as chronic resistance exercise [84], chronic swimming [85], voluntary exercise [86, 87], and treadmill running [88, 89, 90, 91] have been shown to exert beneficial effect on mitigation of cardiac structural damage and preservation of cardiac performance in animal models of DOX cardiotoxicity. Moreover, acute exercise prior to DOX treatment protects cardiac function of breast cancer patients [92]. The protective effects of exercise on DOX-induced cardiac injury may be attributed to increased antioxidant ability, increased expression of heat shock proteins and antiapoptotic proteins, improved mitochondrial quality control, maintenance of calcium handling, and altered delivery of DOX to myocardium [90, 91, 93]. Importantly, exercise training has no effect on antitumor efficacy of DOX [94]. However, these preclinical and clinical findings need to be verified by studies involving a large cohort of patients.

5.5. Calorie restriction and fasting

Calorie restriction is beneficial for several types of cardiovascular diseases including DOX cardiotoxicity [95, 96]. However, calorie restriction is hard to sustain in the long term. Although calorie restriction mimetics are more practical in terms of sustainability, they are less accessible and cost ineffective. Fasting has been shown to exert beneficial effects on certain forms of cardiovascular diseases including age-related cardiac hypertrophy, myocardial ischemic injury, and coronary heart disease risk factors through diverse mechanisms including remodeling of mitochondrial networks, improvement of energy metabolism, reduction in signaling pathways related to survival such as insulin and insulin-like growth factor-1 signaling, decrease in mitochondrial oxidative stress, and enhancement of autophagic flux [97, 98]. Recent studies suggest that fasting also conferred cardioprotection against DOX cardiotoxicity. In animal models, short-term fasting ameliorates cardiac damage and cardiac dysfunction caused by DOX [98]. Alternate-day fasting, a type of intermittent fasting, is capable of mitigating DOX cardiotoxicity in mouse models of both acute and chronic DOX cardiotoxicity [30]. More importantly, intermittent fasting and multiple fasting cycles have recently been shown to suppress tumor growth and sensitize various tumors to chemotherapy [99, 100]. Therefore, intermittent fasting could be considered as a potential preventive or therapeutic strategy for cardiotoxicity induced by DOX. However, given that long-term fasting is harmful to health especially for cancer patients due to malnutrition problem, the procedure of intermittent fasting should be optimized under clinical supervision to improve its efficacy while minimizing side effects.

Advertisement

6. Conclusions

DOX is one of the most effective chemotherapeutic agents. However, potential acute or chronic irreversible cumulative cardiotoxicity limits its clinical application. It is encouraging that accumulating evidences from basic research, preclinical experiments and clinical trials provide insight into the pathophysiology and molecular mechanisms of this disease, which potentially leads to identification of novel biomarkers for early detection and establishment of preventive strategies. Moreover, emerging evidences have associated DOX cardiotoxicity with genetic risk factors. Findings in this direction will be helpful to predict tumor sensitivity to DOX treatment and susceptibility to DOX-induced cardiotoxicity of the population. As a consequence, precise strategies may be developed and applied to individuals to achieve maximal efficacy for cancer treatment and meanwhile minimal side effects on the basis of patient-specific genetic variants.

Advertisement

Acknowledgments

This work was supported by research grants from the Natural Science Foundation of Shanghai (16ZR1418200).

Advertisement

Conflict of interest

No potential conflict of interests were declared.

References

  1. 1. Swain SM, Whaley FS, Ewer MS. Congestive heart failure in patients treated with doxorubicin: A retrospective analysis of three trials. Cancer. 2003;97:2869-2879. DOI: 10.1002/cncr.11407
  2. 2. Aversano RC, Boor PJ. Histochemical alterations of acute and chronic doxorubicin cardiotoxicity. Journal of Molecular and Cellular Cardiology. 1983;15:543-553
  3. 3. Lim CC, Zuppinger C, Guo X, Kuster GM, Helmes M, Eppenberger HM, Suter TM, Liao R, Sawyer DB. Anthracyclines induce calpain-dependent titin proteolysis and necrosis in cardiomyocytes. The Journal of Biological Chemistry. 2004;279:8290-8299. DOI: 10.1074/jbc.M308033200
  4. 4. Tomlinson CW, McGrath GM, McNeill JH. Adriamycin cardiomyopathy: Pathological and membrane functional changes in a canine model with mild impairment of left ventricular function. The Canadian Journal of Cardiology. 1986;2:368-374
  5. 5. Mortensen SA, Olsen HS, Baandrup U. Chronic anthracycline cardiotoxicity: Haemo-dynamic and histopathological manifestations suggesting a restrictive endomyocardial disease. British Heart Journal. 1986;55:274-282
  6. 6. Van Vleet JF, Ferrans VJ, Weirich WE. Cardiac disease induced by chronic adriamycin administration in dogs and an evaluation of vitamin E and selenium as cardioprotectants. The American Journal of Pathology. 1980;99:13-42
  7. 7. Li DL, Wang ZV, Ding G, Tan W, Luo X, Criollo A, Xie M, Jiang N, May H, Kyrychenko V, Schneider JW, Gillette TG, Hill JA. Doxorubicin blocks cardiomyocyte autophagic flux by inhibiting lysosome acidification. Circulation. 2016;133:1668-1687. DOI: 10.1161/CIRCULATIONAHA.115.017443
  8. 8. Ganz WI, Sridhar KS, Forness TJ. Detection of early anthracycline cardiotoxicity by monitoring the peak filling rate. American Journal of Clinical Oncology. 1993;16:109-112
  9. 9. Turrens JF. Mitochondrial formation of reactive oxygen species. The Journal of Physio-logy. 2003;552:335-344. DOI: 10.1111/j.1469-7793.2003.00335.x
  10. 10. Cho KJ, Seo JM, Kim JH. Bioactive lipoxygenase metabolites stimulation of NADPH oxidases and reactive oxygen species. Molecules and Cells. 2011;32:1-5. DOI: 10.1007/s10059-011-1021-7
  11. 11. Bae YS, Oh H, Rhee SG, Yoo YD. Regulation of reactive oxygen species generation in cell signaling. Molecules and Cells. 2011;32:491-509. DOI: 10.1007/s10059-011-0276-3
  12. 12. Berthiaume JM, Wallace KB. Adriamycin-induced oxidative mitochondrial cardiotoxicity. Cell Biology and Toxicology. 2007;23:15-25. DOI: 10.1007/s10565-006-0140-y
  13. 13. Zorov DB, Filburn CR, Klotz LO, Zweier JL, Sollott SJ. Reactive oxygen species (ROS)-induced ROS release: A new phenomenon accompanying induction of the mitochondrial permeability transition in cardiac myocytes. The Journal of Experimental Medicine. 2000;192:1001-1014. DOI: 10.1084/jem.192.7.1001
  14. 14. Redza-Dutordoir M, Averill-Bates DA. Activation of apoptosis signalling pathways by reactive oxygen species. Biochimica et Biophysica Acta. 2016;1863:2977-2992. DOI: 10.1016/j.bbamcr.2016.09.012
  15. 15. Ichikawa Y, Ghanefar M, Bayeva M, Wu R, Khechaduri A, Naga Prasad SV, Mutharasan RK, Naik TJ, Ardehali H. Cardiotoxicity of doxorubicin is mediated through mitochondrial iron accumulation. The Journal of Clinical Investigation. 2014;124:617-630. DOI: 10.1172/JCI72931
  16. 16. Simůnek T, Stérba M, Popelová O, Adamcová M, Hrdina R, Gersl V. Anthracycline-induced cardiotoxicity: Overview of studies examining the roles of oxidative stress and free cellular iron. Pharmacological Reports. 2009;61:154-171. DOI: 10.1016/S1734-1140(09)70018-0
  17. 17. Sawyer DB. Anthracyclines and heart failure. The New England Journal of Medicine. 2013;368:1154-1156. DOI: 10.1056/NEJMcibr1214975
  18. 18. Lyu YL, Kerrigan JE, Lin CP, Azarova AM, Tsai YC, Ban Y, Liu LF. Topoisomerase II beta mediated DNA double-strand breaks: Implications in doxorubicin cardiotoxicity and prevention by dexrazoxane. Cancer Research. 2007;67:8839-8846. DOI: 10.1158/0008-5472.CAN-07-1649
  19. 19. Zhang S, Liu X, Bawa-Khalfe T, Lu LS, Lyu YL, Liu LF, Yeh ET. Identification of the molecular basis of doxorubicin-induced cardiotoxicity. Nature Medicine. 2012;18:1639-1642. DOI: 10.1038/nm.2919
  20. 20. Levine B, Klionsky DJ. Development by self-digestion: Molecular mechanisms and biological functions of autophagy. Developmental Cell. 2004;6:463-477. DOI: 10.1016/S1534-5807(04)00099-1
  21. 21. Linton PJ, Gurney M, Sengstock D, Mentzer RM Jr, Gottlieb RA. This old heart: Cardiac aging and autophagy. Journal of Molecular and Cellular Cardiology. 2015;83:44-54. DOI: 10.1016/j.yjmcc.2014.12.017
  22. 22. Xie M, Morales CR, Lavandero S, Hill JA. Tuning flux: Autophagy as a target of heart disease therapy. Current Opinion in Cardiology. 2011;26:216-222. DOI: 10.1097/HCO.0b013e328345980a
  23. 23. Kobayashi S, Volden P, Timm D, Mao K, Xu X, Liang Q. Transcription factor GATA4 inhibits doxorubicin-induced autophagy and cardiomyocyte death. The Journal of Bio-logical Chemistry. 2010;285:793-804. DOI: 10.1074/jbc.M109.070037
  24. 24. Sun A, Cheng Y, Zhang Y, Zhang Q, Wang S, Tian S, Zou Y, Hu K, Ren J, Ge J. Aldehyde dehydrogenase 2 ameliorates doxorubicin-induced myocardial dysfunction through detoxification of 4-HNE and suppression of autophagy. Journal of Molecular and Cellular Cardiology. 2014;71:92-104. DOI: 10.1016/j.yjmcc.2014.01.002
  25. 25. Xu X, Chen K, Kobayashi S, Timm D, Liang Q. Resveratrol attenuates doxorubicin-induced cardiomyocyte death via inhibition of p70 S6 kinase1-mediated autophagy. The Journal of Pharmacology and Experimental Therapeutics. 2012;341:183-195. DOI: 10.1124/jpet.111.189589
  26. 26. Lu L, Wu W, Yan J, Li X, Yu H, Yu X. Adriamycin-induced autophagic cardiomyocyte death plays a pathogenic role in a rat model of heart failure. International Journal of Cardiology. 2009;134:82-90. DOI: 10.1016/j.ijcard.2008.01.043
  27. 27. Xu X, Bucala R, Ren J. Macrophage migration inhibitory factor deficiency augments doxorubicin-induced cardiomyopathy. Journal of the American Heart Association. 2013;2:e000439. DOI: 10.1161/JAHA.113.000439
  28. 28. Bartlett JJ, Trivedi PC, Yeung P, Kienesberger PC, Pulinilkunnil T. Doxorubicin impairs cardiomyocyte viability by suppressing transcription factor EB expression and disrupting autophagy. The Biochemical Journal. 2016;473:3769-3789. DOI: 10.1042/BCJ20160385
  29. 29. Kawaguchi T, Takemura G, Kanamori H, Takeyama T, Watanabe T, Morishita K, Ogino A, Tsujimoto A, Goto K, Maruyama R, Kawasaki M, Mikami A, Fujiwara T, Fujiwara H, Minatoguchi S. Prior starvation mitigates acute doxorubicin cardiotoxicity through restoration of autophagy in affected cardiomyocytes. Cardiovascular Research. 2012;96:456-465. DOI: 10.1093/cvr/cvs282
  30. 30. An L, Hu XW, Zhang S, Hu X, Song Z, Naz A, Zi Z, Wu J, Li C, Zou Y, He L, Zhu H. UVRAG deficiency exacerbates doxorubicin-induced cardiotoxicity. Scientific Reports. 2017;7:43251. DOI: 10.1038/srep43251
  31. 31. Ni HM, Williams JA, Ding WX. Mitochondrial dynamics and mitochondrial quality control. Redox Biology. 2015;4:6-13. DOI: 10.1016/j.redox.2014.11.006
  32. 32. Thomas RL, Gustafsson AB. Mitochondrial autophagy—An essential quality control mechanism for myocardial homeostasis. Circulation Journal. 2013;77:2449-2454. DOI: 10.1253/circj.CJ-13-0835
  33. 33. Saito T, Sadoshima J. Molecular mechanisms of mitochondrial autophagy/mitophagy in the heart. Circulation Research. 2015;116:1477-1490. DOI: 10.1161/CIRCRESAHA.116.303790
  34. 34. Hull TD, Boddu R, Guo L, Tisher CC, Traylor AM, Patel B, Joseph R, Prabhu SD, Suliman HB, Piantadosi CA, Agarwal A, George JF. Heme oxygenase-1 regulates mitochondrial quality control in the heart. JCI Insight. 2016;1:e85817. DOI: 10.1172/jci.insight.85817
  35. 35. Hoshino A, Mita Y, Okawa Y, Ariyoshi M, Iwai-Kanai E, Ueyama T, Ikeda K, Ogata T, Matoba S. Cytosolic p53 inhibits Parkin-mediated mitophagy and promotes mitochondrial dysfunction in the mouse heart. Nature Communications. 2013;4:2308. DOI: 10.1038/ncomms3308
  36. 36. Osellame LD, Singh AP, Stroud DA, Palmer CS, Stojanovski D, Ramachandran R, Ryan MT. Cooperative and independent roles of the Drp1 adaptors Mff, MiD49 and MiD51 in mitochondrial fission. Journal of Cell Science. 2016;129:2170-2181. DOI: 10.1242/jcs.185165
  37. 37. Kavazis AN, Morton AB, Hall SE, Smuder AJ. Effects of doxorubicin on cardiac muscle subsarcolemmal and intermyofibrillar mitochondria. Mitochondrion. 2017;34:9-19. DOI: 10.1016/j.mito.2016.10.008
  38. 38. Hao E, Mukhopadhyay P, Cao Z, Erdélyi K, Holovac E, Liaudet L, Lee WS, Haskó G, Mechoulam R, Pacher P. Cannabidiol protects against doxorubicin-induced cardiomyopathy by modulating mitochondrial function and biogenesis. Molecular Medicine. 2015;21:38-45. DOI: 10.2119/molmed.2014.00261
  39. 39. Miyagawa K, Emoto N, Widyantoro B, Nakayama K, Yagi K, Rikitake Y, Suzuki T, Hirata K. Attenuation of doxorubicin-induced cardiomyopathy by endothelin-converting enzyme-1 ablation through prevention of mitochondrial biogenesis impairment. Hypertension. 2010;55:738-746. DOI: 10.1161/HYPERTENSIONAHA.109.141903
  40. 40. Nozaki N, Shishido T, Takeishi Y, Kubota I. Modulation of doxorubicin-induced cardiac dysfunction in toll-like receptor-2-knockout mice. Circulation. 2004;110:2869-2874. DOI: 10.1161/01.CIR.0000146889.46519.27
  41. 41. Nogueira-Machado JA, de Oliveira Volpe CM. HMGB-1 as a target for inflammation controlling. Recent Patents on Endocrine, Metabolic & Immune Drug Discovery. 2012;6:201-209. DOI: 10.2174/187221412802481784
  42. 42. Tsujimura H, Tamura T, Kong HJ, Nishiyama A, Ishii KJ, Klinman DM, Ozato K. Toll-like receptor 9 signaling activates NF-kappaB through IFN regulatory factor-8/IFN consensus sequence binding protein in dendritic cells. Journal of Immunology. 2004;172:6820-6827. DOI: 10.4049/jimmunol.172.11.6820
  43. 43. Murakawa T, Nakayama H, Nishida K, Akira S, Yamamoto A, Komuro I, Otsu K. Mitochondrial DNA that escapes from autophagy causes inflammation and heart failure. Nature. 2012;485:251-255. DOI: 10.1038/nature10992
  44. 44. Williams AJ. The functions of two species of calcium channel in cardiac muscle excitation-contraction coupling. European Heart Journal. 1997;18(Suppl A):A27-A35
  45. 45. Saeki K, Obi I, Ogiku N, Shigekawa M, Imagawa T, Matsumoto T. Doxorubicin directly binds to the cardiac-type ryanodine receptor. Life Sciences. 2002;70:2377-2389. DOI: 10.1016/S0024-3205(02)01524-2
  46. 46. Hanna AD, Lam A, Thekkedam C, Willemse H, Dulhunty AF, Beard NA. The anthracycline metabolite doxorubicinol abolishes RyR2 sensitivity to physiological changes in luminal Ca2+ through an interaction with calsequestrin. Molecular Pharmacology. 2017;92:576-587. DOI: 10.1124/mol.117.108183
  47. 47. Hanna AD, Lam A, Tham S, Dulhunty AF, Beard NA. Adverse effects of doxorubicin and its metabolic product on cardiac RyR2 and SERCA2A. Molecular Pharmacology. 2014;86:438-449. DOI: 10.1124/mol.114.093849
  48. 48. Sag CM, Köhler AC, Anderson ME, Backs J, Maier LS. CaMKII-dependent SR Ca leak contributes to doxorubicin-induced impaired Ca handling in isolated cardiac myocytes. Journal of Molecular and Cellular Cardiology. 2011;51:749-759. DOI: 10.1016/j.yjmcc.2011.07.016
  49. 49. Kim SY, Kim SJ, Kim BJ, Rah SY, Chung SM, Im MJ, Kim UH. Doxorubicin-induced reactive oxygen species generation and intracellular Ca2+ increase are reciprocally modulated in rat cardiomyocytes. Experimental & Molecular Medicine. 2006;38:535-545. DOI: 10.1038/emm.2006.63
  50. 50. L’Ecuyer T, Sanjeev S, Thomas R, Novak R, Das L, Campbell W, Heide RV. DNA damage is an early event in doxorubicin-induced cardiac myocyte death. American Journal of Physiology. Heart and Circulatory Physiology. 2006;291:H1273-H1280. DOI: 10.1152/ajpheart.00738.2005
  51. 51. Shizukuda Y, Matoba S, Mian OY, Nguyen T, Hwang PM. Targeted disruption of p53 attenuates doxorubicin-induced cardiac toxicity in mice. Molecular and Cellular Biochemistry. 2005;273:25-32. DOI: 10.1007/s11010-005-5905-8
  52. 52. Chacon E, Acosta D. Mitochondrial regulation of superoxide by Ca2+: An alternate mechanism for the cardiotoxicity of doxorubicin. Toxicology and Applied Pharmacology. 1991;107:117-128
  53. 53. Singal PK, Deally CM, Weinberg LE. Subcellular effects of adriamycin in the heart: A concise review. Journal of Molecular and Cellular Cardiology. 1987;19:817-828
  54. 54. Carvalho FS, Burgeiro A, Garcia R, Moreno AJ, Carvalho RA, Oliveira PJ. Doxorubicin-induced cardiotoxicity: From bioenergetic failure and cell death to cardiomyopathy. Medicinal Research Reviews. 2014;34:106-135. DOI: 10.1002/med.21280
  55. 55. Dhingra R, Margulets V, Chowdhury SR, Thliveris J, Jassal D, Fernyhough P, Dorn GW II, Kirshenbaum LA. Bnip3 mediates doxorubicin-induced cardiac myocyte necrosis and mortality through changes in mitochondrial signaling. Proceedings of the National Academy of Sciences of the United States of America. 2014;111:E5537-E5544. DOI: 10.1073/pnas.1414665111
  56. 56. Babuin L, Jaffe AS. Troponin: The biomarker of choice for the detection of cardiac injury. Canadian Medical Association Journal. 2005;173:1191-1202. DOI: 10.1503/cmaj/051291
  57. 57. Lipshultz SE, Miller TL, Scully RE, Lipsitz SR, Rifai N, Silverman LB, Colan SD, Neuberg DS, Dahlberg SE, Henkel JM, Asselin BL, Athale UH, Clavell LA, Laverdière C, Michon B, Schorin MA, Sallan SE. Changes in cardiac biomarkers during doxorubicin treatment of pediatric patients with high-risk acute lymphoblastic leukemia: Associations with long-term echocardiographic outcomes. Journal of Clinical Oncology. 2012;30:1042-1049. DOI: 10.1200/JCO.2010.30.3404
  58. 58. Wallace KB, Hausner E, Herman E, Holt GD, MacGregor JT, Metz AL, Murphy E, Rosenblum IY, Sistare FD, York MJ. Serum troponins as biomarkers of drug-induced cardiac toxicity. Toxicologic Pathology. 2004;32:106-121. DOI: 10.1080/01926230490261302
  59. 59. Levin ER, Gardner DG, Samson WK. Natriuretic peptides. The New England Journal of Medicine. 1998;339:321-328. DOI: 10.1056/NEJM199807303390507
  60. 60. Yandle TG. Biochemistry of natriuretic peptides. Journal of Internal Medicine. 1994;235:561-576
  61. 61. Goetze JP. Biochemistry of pro-B-type natriuretic peptide-derived peptides: The endocrine heart revisited. Clinical Chemistry. 2004;50:1503-1510. DOI: 10.1373/clinchem.2004.034272
  62. 62. Ekstein S, Nir A, Rein AJ, Perles Z, Bar-Oz B, Salpeter L, Algur N, Weintraub M. N-terminal-proB-type natriuretic peptide as a marker for acute anthracycline cardiotoxicity in children. Journal of Pediatric Hematology/Oncology. 2007;29:440-444. DOI: 10.1097/MPH.0b013e3180640d42
  63. 63. Pongprot Y, Sittiwangkul R, Charoenkwan P, Silvilairat S. Use of cardiac markers for monitoring of doxorubixin-induced cardiotoxicity in children with cancer. Journal of Pediatric Hematology/Oncology. 2012;34:589-595. DOI: 10.1097/MPH.0b013e31826faf44
  64. 64. Holmgren G, Synnergren J, Andersson CX, Lindahl A, Sartipy P. MicroRNAs as potential biomarkers for doxorubicin-induced cardiotoxicity. Toxicology In Vitro. 2016;34:26-34. DOI: 10.1016/j.tiv.2016.03.009
  65. 65. Chaudhari U, Nemade H, Gaspar JA, Hescheler J, Hengstler JG, Sachinidis A. MicroRNAs as early toxicity signatures of doxorubicin in human-induced pluripotent stem cell-derived cardiomyocytes. Archives of Toxicology. 2016;90:3087-3098. DOI: 10.1007/s00204-016-1668-0
  66. 66. Rigaud VO, Ferreira LR, Ayub-Ferreira SM, Ávila MS, Brandão SM, Cruz FD, Santos MH, Cruz CB, Alves MS, Issa VS, Guimarães GV, Cunha-Neto E, Bocchi EA. Circulating miR-1 as a potential biomarker of doxorubicin-induced cardiotoxicity in breast cancer patients. Oncotarget. 2017;8:6994-7002. DOI: 10.18632/oncotarget.14355
  67. 67. Wojnowski L, Kulle B, Schirmer M, Schlüter G, Schmidt A, Rosenberger A, Vonhof S, Bickeböller H, Toliat MR, Suk EK, Tzvetkov M, Kruger A, Seifert S, Kloess M, Hahn H, Loeffler M, Nürnberg P, Pfreundschuh M, Trümper L, Brockmöller J, Hasenfuss G. NAD(P)H oxidase and multidrug resistance protein genetic polymorphisms are associated with doxorubicin-induced cardiotoxicity. Circulation. 2005;112:3754-3762. DOI: 10.1161/CIRCULATIONAHA.105.576850
  68. 68. Magdy T, Burmeister BT, Burridge PW. Validating the pharmacogenomics of chemotherapy-induced cardiotoxicity: What is missing? Pharmacology & Therapeutics. 2016;168:113-125. DOI: 10.1016/j.pharmthera.2016.09.009
  69. 69. Todorova VK, Makhoul I, Dhakal I, Wei J, Stone A, Carter W, Owen A, Klimberg VS. Polymorphic variations associated with doxorubicin-induced cardiotoxicity in breast cancer patients. Oncology Research. 2017;25:1223-1229. DOI: 10.3727/096504017X14876245096439
  70. 70. Reichardt P. High-dose chemotherapy in adult soft tissue sarcoma. Critical Reviews in Oncology/Hematology. 2002;4:1157-1167. DOI: 10.1016/S1040-8428(01)00153-6
  71. 71. Legha SS, Benjamin RS, Mackay B, Ewer M, Wallace S, Valdivieso M, Rasmussen SL, Blumenschein GR, Freireich EJ. Reduction of doxorubicin cardiotoxicity by prolonged continuous intravenous infusion. Annals of Internal Medicine. 1982;96:133-139
  72. 72. Hortobagyi GN, Frye D, Buzdar AU, Ewer MS, Fraschini G, Hug V, Ames F, Montague E, Carrasco CH, Mackay B. Decreased cardiac toxicity of doxorubicin administered by continuous intravenous infusion in combination chemotherapy for metastatic breast carcinoma. Cancer. 1989;63:37-45
  73. 73. Lipshultz SE, Giantris AL, Lipsitz SR, Kimball Dalton V, Asselin BL, Barr RD, Clavell LA, Hurwitz CA, Moghrabi A, Samson Y, Schorin MA, Gelber RD, Sallan SE, Colan SD. Doxorubicin administration by continuous infusion is not cardioprotective: The Dana-Farber 91-01 acute lymphoblastic leukemia protocol. Journal of Clinical Oncology. 2002;20:1677-1682. DOI: 10.1200/JCO.2002.20.6.1677
  74. 74. Lipshultz SE, Miller TL, Lipsitz SR, Neuberg DS, Dahlberg SE, Colan SD, Silverman LB, Henkel JM, Franco VI, Cushman LL, Asselin BL, Clavell LA, Athale U, Michon B, Laverdière C, Schorin MA, Larsen E, Usmani N, Sallan SE. Continuous versus bolus infusion of doxorubicin in children with ALL: Long-term cardiac outcomes. Pediatrics. 2012;130:1003-1011. DOI: 10.1542/peds.2012-0727
  75. 75. Greish K. Enhanced permeability and retention (EPR) effect for anticancer nanomedicine drug targeting. Methods in Molecular Biology. 2010;624:25-37. DOI: 10.1007/978-1-60761-609-2_3
  76. 76. Muggia FM. Doxorubicin-polymer conjugates: Further demonstration of the concept of enhanced permeability and retention. Clinical Cancer Research. 1999;5:7-8
  77. 77. Ewer MS, Martin FJ, Henderson C, Shapiro CL, Benjamin RS, Gabizon AA. Cardiac safety of liposomal anthracyclines. Seminars in Oncology. 2004;31:161-181. DOI: 10.1053/j.seminoncol.2004.08.006
  78. 78. Kanwal U, Irfan Bukhari N, Ovais M, Abass N, Hussain K, Raza A. Advances in nano-delivery systems for doxorubicin: An updated insight. Journal of Drug Targeting. 2018;26:296-310. DOI: 10.1080/1061186X.2017.1380655
  79. 79. Cvetković RS, Scott LJ. Dexrazoxane: A review of its use for cardioprotection during anthracycline chemotherapy. Drugs. 2005;65:1005-1024. DOI: 10.2165/00003495-200565070-00008
  80. 80. Sepe DM, Ginsberg JP, Balis FM. Dexrazoxane as a cardioprotectant in children receiving anthracyclines. The Oncologist. 2010;15:1220-1226. DOI: 10.1634/theoncologist.2010-0162
  81. 81. He L, He T, Farrar S, Ji L, Liu T, Ma X. Antioxidants maintain cellular redox homeostasis by elimination of reactive oxygen species. Cellular Physiology and Biochemistry. 2017;44:532-553. DOI: 10.1159/000485089
  82. 82. Nabati M, Janbabai G, Baghyari S, Esmaili K, Yazdani J. Cardioprotective effects of carvedilol in inhibiting doxorubicin-induced cardiotoxicity. Journal of Cardiovascular Pharmacology. 2017;69:279-285. DOI: 10.1097/FJC.0000000000000470
  83. 83. Spallarossa P, Garibaldi S, Altieri P, Fabbi P, Manca V, Nasti S, Rossettin P, Ghigliotti G, Ballestrero A, Patrone F, Barsotti A, Brunelli C. Carvedilol prevents doxorubicin-induced free radical release and apoptosis in cardiomyocytes in vitro. Journal of Molecular and Cellular Cardiology. 2004;37:837-846. DOI: 10.1016/j.yjmcc.2004.05.024
  84. 84. Pfannenstiel K, Hayward R. Effects of resistance exercise training on doxorubicin-induced cardiotoxicity. Journal of Cardiovascular Pharmacology. 2018;71:332-339. DOI: 10.1097/FJC.0000000000000574
  85. 85. Kanter MM, Hamlin RL, Unverferth DV, Davis HW, Merola AJ. Effect of exercise training on antioxidant enzymes and cardiotoxicity of doxorubicin. Journal of Applied Physiology. 1985;59:1298-1303
  86. 86. Chicco AJ, Schneider CM, Hayward R. Voluntary exercise protects against acute doxorubicin cardiotoxicity in the isolated perfused rat heart. American Journal of Physiology. Regulatory, Integrative and Comparative Physiology. 2005;289:R424-R431. DOI: 10.1152/ajpregu.00636.2004
  87. 87. Jensen BT, Lien CY, Hydock DS, Schneider CM, Hayward R. Exercise mitigates cardiac doxorubicin accumulation and preserves function in the rat. Journal of Cardiovascular Pharmacology. 2013;62:263-269. DOI: 10.1097/FJC.0b013e3182982ce0
  88. 88. Lien CY, Jensen BT, Hydock DS, Hayward R. Short-term exercise training attenuates acute doxorubicin cardiotoxicity. Journal of Physiology and Biochemistry. 2015;71:669-678. DOI: 10.1007/s13105-015-0432-x
  89. 89. Wang F, Iskra B, Kleinerman E, Alvarez-Florez C, Andrews T, Shaw A, Chandra J, Schadler K, Aune GJ. Aerobic exercise during early murine doxorubicin exposure mitigates cardiac toxicity. Journal of Pediatric Hematology/Oncology. 2018;40:208-215. DOI: 10.1097/MPH.0000000000001112
  90. 90. Kavazis AN, Smuder AJ, Min K, Tümer N, Powers SK. Short-term exercise training protects against doxorubicin-induced cardiac mitochondrial damage independent of HSP72. American Journal of Physiology. Heart and Circulatory Physiology. 2010;299:H1515-H1524. DOI: 10.1152/ajpheart.00585.2010
  91. 91. Marques-Aleixo I, Santos-Alves E, Torrella JR, Oliveira PJ, Magalhães J, Ascensão A. Exercise and doxorubicin treatment modulate cardiac mitochondrial quality control signaling. Cardiovascular Toxicology. 2018;18:43-55. DOI: 10.1007/s12012-017-9412-4
  92. 92. Kirkham AA, Shave RE, Bland KA, Bovard JM, Eves ND, Gelmon KA, McKenzie DC, Virani SA, Stöhr EJ, Warburton DER, Campbell KL. Protective effects of acute exercise prior to doxorubicin on cardiac function of breast cancer patients: A proof-of-concept RCT. International Journal of Cardiology. 2017;245:263-270. DOI: 10.1016/j.ijcard.2017.07.037
  93. 93. Marques-Aleixo I, Santos-Alves E, Oliveira PJ, Moreira PI, Magalhães J, Ascensão A. The beneficial role of exercise in mitigating doxorubicin-induced Mitochondrionopathy. Biochimica et Biophysica Acta. 2018;1869:189-199. DOI: 10.1016/j.bbcan.2018.01.002
  94. 94. Parry TL, Hayward R. Exercise training does not affect anthracycline antitumor efficacy while attenuating cardiac dysfunction. American Journal of Physiology. Regulatory, Integrative and Comparative Physiology. 2015;309:R675-R683. DOI: 10.1152/ajpregu.00185.2015
  95. 95. Dolinsky VW, Dyck JR. Calorie restriction and resveratrol in cardiovascular health and disease. Biochimica et Biophysica Acta. 2011;1812:1477-1489. DOI: 10.1016/j.bbadis.2011.06.010
  96. 96. Dutta D, Xu J, Dirain ML, Leeuwenburgh C. Calorie restriction combined with resveratrol induces autophagy and protects 26-month-old rat hearts from doxorubicin-induced toxicity. Free Radical Biology & Medicine. 2014;74:252-262. DOI: 10.1016/j.freeradbiomed.2014.06.011
  97. 97. Zhu H, He L. Cardioprotective effects of intermittent fasting. Journal of Clinical and Diagnostic Research. 2017;5:1. DOI: 10.4172/2376-0311.1000138
  98. 98. Dirks-Naylor AJ, Kouzi SA, Yang S, Tran NT, Bero JD, Mabolo R, Phan DT, Whitt SD, Taylor HN. Can short-term fasting protect against doxorubicin-induced cardiotoxicity? World Journal of Biological Chemistry. 2014;5:269-274. DOI: 10.4331/wjbc.v5.i3.269
  99. 99. Rocha NS, Barbisan LF, de Oliveira ML, de Camargo JL. Effects of fasting and intermittent fasting on rat hepatocarcinogenesis induced by diethylnitrosamine. Teratogenesis, Carcinogenesis, and Mutagenesis. 2002;22:129-138. DOI: 10.1002/tcm.10005
  100. 100. Lee C, Raffaghello L, Brandhorst S, Safdie FM, Bianchi G, Martin-Montalvo A, Pistoia V, Wei M, Hwang S, Merlino A, Emionite L, de Cabo R, Longo VD. Fasting cycles retard growth of tumors and sensitize a range of cancer cell types to chemotherapy. Science Translational Medicine. 2012;4:124ra27. DOI: 10.1126/scitranslmed.3003293

Written By

Hongxin Zhu

Submitted: 11 March 2018 Reviewed: 18 May 2018 Published: 05 November 2018