Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Biomedicine & Pharmacotherapy 139 (2021) 111708

Contents lists available at ScienceDirect

Biomedicine & Pharmacotherapy


journal homepage: www.elsevier.com/locate/biopha

Review

Doxorubicin-induced cardiotoxicity: An update on the molecular


mechanism and novel therapeutic strategies for effective management
Pushkar Singh Rawat a, Aiswarya Jaiswal a, Amit Khurana b, c, Jasvinder Singh Bhatti d,
Umashanker Navik a, *
a
Department of Pharmacology, Central University of Punjab, Bathinda, Punjab, 151401, India
b
Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science, PVNRTVU, Rajendranagar, Hyderabad 500030, Telangana, India
c
Centre for Biomedical Engineering (CBME), Indian Institute of Technology (IIT), Delhi 110016, India
d
Department of human genetics and molecular medicine, School of health sciences, Central University of Punjab, Bathinda 151401, Punjab, India

A R T I C L E I N F O A B S T R A C T

Keywords: Doxorubicin (Dox) is a secondary metabolite of the mutated strain of Streptomyces peucetius var. Caesius and
Cardiotoxicity belongs to the anthracyclines family. The anti-cancer activity of Dox is mainly exerted through the DNA inter­
Doxorubicin calation and inhibiting topoisomerase II enzyme in fast-proliferating tumors. However, Dox causes cumulative
Oxidative stress
and dose-dependent cardiotoxicity, which results in increased risks of mortality among cancer patients and thus
DNMT 1
Histone deacetylase
limiting its wide clinical applications. There are several mechanisms has been proposed for doxorubicin-induced
micro RNAs cardiotoxicity and oxidative stress, free radical generation and apoptosis are most widely reported. Apart from
this, other mechanisms are also involved in Dox-induced cardiotoxicity such as impaired mitochondrial function,
a perturbation in iron regulatory protein, disruption of Ca2+ homeostasis, autophagy, the release of nitric oxide
and inflammatory mediators and altered gene and protein expression that involved apoptosis. Dox also causes
downregulation of DNA methyltransferase 1 (DNMT1) enzyme activity which leads to a reduction in the DNA
methylation process. This hypomethylation causes dysregulation in the mitochondrial genes like peroxisome
proliferator-activated receptor-gamma coactivator (PGC)-1-alpha (PGC-1α), nuclear respiratory factor 1 (NRF-1)
and mitochondrial transcription factor A (TFAM) unit in the heart. Apart from DNA methylation, Dox treatment
also alters the micro RNAs levels and histone deacetylase (HDAC) activity. Therefore, in the current review, we
have provided a detailed update on the current understanding of the pathological mechanisms behind the well-
known Dox-induced cardiotoxicity. Further, we have provided some of the most plausible pharmacological
strategies which have been tested against Dox-induced cardiotoxicity.

1. Introduction ejection fraction in 9% of the treated patients [2]. Apoptosis-mediated


loss of cardiomyocytes and oxidative stress is stated as the main cause
Doxorubicin (Dox) is a secondary metabolite of a mutated strain of of Dox-induced cardiomyopathy. Depletion of Sarcoplasmic Reticulum
Streptomyces peucetius var. caesius and belongs to the anthracyclines Ca2+ is one of the early phase events in Dox-induced cardiomyopathy.
family. Dox is used as an effective antineoplastic agent for multiple Cardiac cell apoptosis and heart failure are well correlated with altered
cancer types. However, multiple systemic adverse effects restrain its levels of Ca2+ and calmodulin-dependent kinase II [3]. Moreover, in­
wide use in cancer chemotherapy. It is one of the established and hibition of mitochondrial biogenesis is thought to be the mechanism
commonly used antineoplastic agents in treating different types of behind Dox-mediated cardiotoxicity, mainly by activating cell death
cancers, including pediatric cancer, leukemia, breast cancer, etc. Dox is pathways by inhibiting topoisomerase 2β [4]. Also, reactive oxygen
an inhibitor of the DNA topoisomerase II enzyme and causes damage to species (ROS) play a pivotal role in Dox-induced cardiotoxicity and
the DNA [1]. Anthracyclines cause dose-dependent cardiac toxicity, if several other mechanisms like mitochondrial dysfunction, a perturba­
used above the dose range from 400 mg/m2-700 mg/m2 [1,2]. tion in iron regulatory protein, the release of nitric oxide, inflammatory
Anthracycline chemotherapy for a year caused impaired left ventricular mediators, calcium dysregulation, autophagy and cell death are

* Correspondence to: Department of Pharmacology, Central University of Punjab, Bathinda, Punjab 151401, India.
E-mail addresses: ak3.khurana@gmail.com (A. Khurana), jasvinder.bhatti@cup.edu.in (J.S. Bhatti), usnavik@gmail.com, uma.shanker@cup.edu.in (U. Navik).

https://doi.org/10.1016/j.biopha.2021.111708
Received 26 March 2021; Received in revised form 4 May 2021; Accepted 5 May 2021
Available online 13 May 2021
0753-3322/© 2021 The Author(s). Published by Elsevier Masson SAS. This is an open access article under the CC BY license
(http://creativecommons.org/licenses/by/4.0/).
P.S. Rawat et al. Biomedicine & Pharmacotherapy 139 (2021) 111708

reported to play significant role [1]. Several drugs are used in mg/m2 its percentage is 18–48% [8]. Overall, cases of heart failure over
Dox-induced cardiotoxicities, like dexrazoxane concomitant with Dox, the world are 4.5–7%. Cancer and heart diseases are currently the
which binds at topoisomerase 2β and reduces cardiotoxicity and heart leading cause of death in developed and developing countries [7,9].
failure in sensitive cardiac patients. Various approaches, including Table 1 enlists some of the drugs which are known to cause
angiotensin inhibitors, β-blockers and phytochemicals, show protective cardiotoxicity.
action over Dox-induced cardiotoxicity and prevent cardiovascular
dysfunctioning [4]. Therefore, this review aims to discuss the detailed 3. Doxorubicin and its pharmacology
mechanisms responsible for Dox-induced cardiotoxicity. Further, some
of the important pharmacological and phytochemicals agents which Dox is a cytotoxic agent that comes under the class of anthracycline
show beneficial effects against Dox-induced cardiotoxicity are antibiotics [19]. Dox is isolated from the culture of Streptomyces peuce­
described. tius, a bacterial species [20]. Chemically, Dox is composed of naph­
thacenequinone nucleus joined via glycosidic linkage to an amine sugar
2. Cardiotoxicity and symptoms called daunosamine. Dox is used to treat several forms of cancer,
including Kaposi’s sarcoma, acute lymphocytic leukemia, breast cancer,
Defects to the cardiac muscle, including heart failure (HF), structural lymphoma, and bladder cancer [21,22]. Dox inhibits DNA and RNA
damage, and hypertension, is a known adverse effect of many conser­ synthesis through the inhibition of topoisomerase II. It attaches over the
vative chemotherapeutic agents. Also, it is defined as the occurrence of nucleic acids of the DNA, apparently by particular insertion of the
signs of HF with an ejection-fraction reduction ≥ 5% to < 55% or the anthracycline nuclei with the DNA of human cells [22,23]. In the
lack of symptoms with an ejection-fraction reduction ≥ 10% to < 55% anthracycline structure, a lipophilic and saturated hydroxyl rich moi­
[5]. Further cardiac damage can be classified as reversible, irreversible, eties is neighbor to the amine sugar which is hydrophilic and makes the
acute and chronic. Irreversible cardiac damage is further categorized as drug soluble in both (hydrophilic and lipophilic) types of solvents. The
type 1 and reversible damage as type 2 [5]. Type 1 (direct damage) is lipophilic part in the Dox shows acidic nature and the amino sugar in the
generally produced by an aggregate dose (dose-dependent), whereas hydrophilic part demonstrated basic nature. These specially bind over
type 2 damage is not associated with aggregate dose (dose-independ­ the plasma protein and cell membrane.
ent). Further, cardiotoxicity’s clinical symptoms include shortness of Dox show a toxic effect on malignant cells. Its detrimental action on
breath, chest pain, heart palpitations, fluid retention in the legs, different body organs is supposed to be connected to nucleotide base
distention of the stomach, and dizziness [5]. insertion and cell membrane by lipid binding activities of Dox. Insertion
of Dox blocks the nucleotide replication and thus the activity of DNA and
2.1. Epidemiology RNA polymerases. The interaction of Dox with the topoisomerase II to
form DNA-rupturable complexes seems to be an effective mechanism of
The occurrence of chemotherapy-induced cardiotoxicity differs Dox as a cell-killing action [24]. Treatment with Dox over cells has
broadly and it depends on the duration of the specific anti-cancer shown a marked specific morphological alteration in apoptosis or pro­
treatment and underlying patient comorbidities. Dose-dependent, cu­ grammed cell death. Doxorubicin treatment in the patients shows both
mulative and progressive cardiotoxicity appears after anthracyclines effects like therapeutic and toxic effects [24].
administration. Historically, Anthracycline is well correlated with left
ventricular dysfunction, reduced left ventricular ejection fraction and 3.1. Pharmacokinetics
symptomatic heart failure in up to 5% of patients [6]. Reduction in the
aggregate dose or cumulative dose of anthracyclines has successfully Polovich et al. has analyzed cancer patients either by giving a single
lowered the heart failure-associated complications. The cumulative dose or multiple drug medication have shown that Dox has a multiphasic
400 mg/m2 of anthracyclines produces an average of 3.5% of heart disposition after giving this drug via intravenously (IV). The dose range
failure cases [7] and the complication with anthracyclines increases of 30–70 mg/m2 of Dox presented the dose-independent pharmacoki­
with the dose. The moderate cumulative dose of 250–400 mg/m2 of netics profile in patients [25].
anthracyclines causes asymptomatic left ventricular dysfunction up to
25% and 35%. If the cumulative dose of anthracyclines is 550 mg/m2 3.1.1. Absorption and distribution
then its cardiotoxicity percentage is 7–16% and at the dose of 700 It is unstable at gastric pH; hence, it is administered via IV route.

Table 1
Agents which cause cardiotoxicity.
Drug Cardiotoxic effects Mechanism of cardiac toxicity Prevalence of cardiotoxicity Reference

Trastuzumab • Heart failure • Damage to mitochondria 1–28% [10]


Sunitinib, Sorafenib • Heart failure • Inhibit tyrosine kinase pathway 3–8% [11]
• Acute coronary syndrome
• Hypertension
5-Fluorouracil • Dilated cardiomyopathy • Vasospasm 1.2–18% [12]
• Ventricular arrhythmia • Autoimmune phenomenon
• Sudden cardiac death • Myocarditis
Imatinib, desatinib, nilotinib • Heart failure • Inhibit ABL tyrosine kinase in cardiac muscle cells 1.7% [13]
Bevacizumab • Hypertension • Unknown 50% [14]
• Congestive heart failure
Cisplatin • Hypertension • Generation of oxidative stress Unknown [15]
• Heart failure
Taxanes (Paclitaxel, docetaxel) • Heart failure • Direct cellular toxicity 2.3–8% [16]
• Lipid peroxidation
Cyclophosphamide • Inflammation of cardiac muscle • Unknown 7–28% [17]
Anthracyclines (Aggregate dose) • Heart failure • Production of free radicals 2% [18]
Doxorubicin (> 500 mg/m2)
Liposomal doxorubicin (> 900 mg/m2)
Epirubicin (> 720 mg/m2)

2
P.S. Rawat et al. Biomedicine & Pharmacotherapy 139 (2021) 111708

Initially, the half-life distribution of Dox is 5 min because of the fast (nitric oxide synthase), NADPH oxidase (NOX),
tissue uptake of the drug. Later, the drug’s terminal half-life distribution mitochondrial-dependent ROS production and many more, which acti­
is slow to about 20–48 h, reflecting the slow elimination from the tissue vates oxidative stress [32]. Herein, we have discussed all these mecha­
[25,26]. Conventional Dox is much distributed in the plasma and the nisms below.
tissues. Absorption occurs into the cell and attaches to the cellular
components that further bind to the nucleic acid in the cellular DNA. It 4.1.1. NOS dependent increased ROS production
does not cross the blood-brain barrier (BBB) and not attains the drug Doxorubicin binds to the eNOS (endothelial nitric oxide synthase)
concentration in the cerebrospinal fluid (CSF) [27]. reductase enzyme and induces Dox semiquinone radical formation; this
Metabolism: After the Dox administration, it converted into its active radical reduces the free oxygen into the superoxide (O–2) free radical.
metabolite doxorubicinol by utilizing an enzyme NADPH-dependent Enzymatic one-electron reduction type of reaction is involved in this
aldoreductases. Doxorubicinol is the active moiety that provides the reactive conversion which shows cardiotoxic effects. Dox completes the
antineoplastic activity. These reductases are located in erythrocytes, one-electron reduction reaction in the presence of flavoenzymes like
kidney and liver cells [28]. Dox metabolite is present more than 20% in NADPH-cytochrome P450 reductase and mitochondrial NADH dehy­
plasma after 5 min of Dox administration; after 30 min, it reaches 70%, drogenase [32,36]. When the drug binds to the eNOS reductase enzyme,
75% after 4 h and 90% in 24 h of drug administration [25]. there is an imbalance in superoxide free radicals and nitric oxide levels.
Elimination: Chiefly, the drug and its metabolite are excreted from Nitric oxide level decreases and the levels of superoxide increase which
bile in unchanged form and approximately 4–5% is excreted from urine leads to cardiotoxicity [37]. A study showed that the Dox administration
after 5 days of Dox administration in the unchanged form [25]. in the bovine aortic endothelial cell leads to increased eNOS mRNA and
proteins level, which activates the redox stimulation, which causes
3.2. Pharmacokinetic study for a special population apoptosis [38]. Interestingly, antisense eNOS mRNA reduced the stim­
ulation of the caspase-3 activity, indicating the cardioprotective effect
3.2.1. In obese patient over Dox-induced cardiotoxicity. Likewise, transgenic eNOS mice show
High body mass index (BMI ≥ 30) in obese women patients leads to enhanced Dox-induced cardiac ROS production, but in knockout mice,
high body surface area owing to their body weight. In such patients, the cardiac ROS levels are decreased [39].
clearance of the drug and its metabolite decreases. Reduction in clear­ There are three types of NOS (nitric oxide synthase) in our system:
ance leads to toxicity due to enhanced AUC levels in obese patients. eNOS, i (inducible) NOS, and n (neuronal) NOS. Apart from eNOS, Dox
Limited evidence of cardiotoxicity appears in men in comparison to fe­ administration increases the iNOS transcription and protein expression
male obese patients [29]. and induces the formation of nitrotyrosine (NT) with increased mito­
chondrial superoxide level in cardiac tissue [40–42]. Superoxide and
3.2.2. In hepatic impairment patients NT’s formation is based on the peroxynitrite level because it generates
In liver compromised patients, the drug and their metabolite level is potent oxidants like nitrogen dioxide and carbonates radical [42]. It
increased because of a reduction in the clearance rate [30]. In hepatic causes apoptosis of the cardiac muscle cells, decreased cardiac
impairment, individuals with serum billirubin level in the range of 2 and contractility, and decreased CAT and glutathione peroxidase activity
3 mg/100 ml, there is reduction in the 50% of dose of Dox, patients with after Dox administration. Chen et al., showed that in iNOS knockout
more than 3 mg/100 ml of serum billirubin dose need to be reduced by mice, the formation of NT and superoxide radicals were reduced after
75% and patients with more than 5 mg/100 ml of serum billirubin level the administration of the Dox and the same effects appear after the
need to restrict the Dox treatment [30]. administration of iNOS inhibitor like S,S˙-[1,3-phenylene-bis(1,2-etha­
nediyl)]bis-isothiourea (1,3-PB-ITU) [43].
3.2.3. In children In contrast, the effect of nNOS in Dox-induced cardiotoxicity is not
The increased clearance rate was seen in the children of age above 2 prominent. This isoform is less important because of poor oxidative
years in comparison to adult ones. A slower clearance rate was seen in stress induction via Dox. No changes appear in nNOS mRNA level and
the children of age below 2 years in contrast to older children [31]. protein expression after Dox administration [42,44]. Hence, the effect of
nNOS in cardiotoxicity is less prominent in comparison to eNOS.
4. Molecular mechanism of doxorubicin-induced cardiotoxicity Moreover, nNOS shows the one-electron reduction of Dox after binding
at flavin domain and this domain plays an essential role in the action of
Several mechanisms have been proposed for Dox-induced car­ Dox [32,40].
diotoxicity or heart failure. The cardiac complication of Dox are directly
related to increased oxidative stress in cardiac tissue. Increased oxida­ 4.1.2. Mitochondrial dependent ROS production
tive stress leads to ROS generation, which causes damage to the heart Dox mainly targets the cardiac cell’s mitochondria, which leads to
muscle and arises due to the reduced level of antioxidants and sulph­ injury in the cardiomyocytes [43,45]. Due to the cationic nature of Dox,
hydryl groups. Further, Dox-induced cardiotoxicity also involves altered it can easily engage inside the mitochondrial membrane and forms an
levels of Ca2+ ions, which causes apoptosis. Apoptosis occurs in the irreversible complex with cardiolipin protein [46]. Dox has a strong
cardiomyocytes and in the endothelial cells with the activation of cas­ affinity towards cardiolipin and it plays a crucial role in the proper
pases [32]. Herein, we have compiled detailed molecular mechanisms functioning of the electron transport chain (ETC) and forms the energy
which are involved in Dox-induced cardiotoxicity. for the body system. After the Dox administration, it bound to car­
diolipin and altered the cardiolipin functioning at the interface, which
4.1. Oxidative stress causes the formation of superoxide (O-2) radical [43,47]. In mitochon­
dria, other proteins are also affected by the Dox treatment, leading to
Production of free radicals is the primary concern that produces impaired mitochondrial functioning [43,48].
cardiac muscle cell injuries after Dox administration [33]. Dox-induced Further, Dox reduces the oxidation of long-chain-fatty acid in mito­
cardiotoxicity appears via increased ROS production and lipid peroxi­ chondria of the heart and increases glucose metabolism, showing
dation in cardiac tissues [34,35]. Aglycones and their iron complexes of anaerobic metabolism apart from aerobic metabolism. This shift in the
anthracyclines encourage the production of ROS [35]. From the last 30 metabolism may lead to heart failure or abnormal contractility and
years, several molecular mechanisms have been postulated for the relaxation; this metabolism shift appears only because of oxidative stress
generation of ROS and its damaging effects on heart tissue. These [43,49–51]. Moreover, Dox affects the expression of the GATA-4 gene in
mechanisms have shown the involvement of various enzymes like NOS the mitochondria, leading to the suppression of mitochondrial synthesis

3
P.S. Rawat et al. Biomedicine & Pharmacotherapy 139 (2021) 111708

and their metabolism, thereby induces apoptosis [52]. Moreover, this developed CHF and acute arrhythmias after the Dox administration with
condition is reversed with the CO (carbon monoxide) inhalation to the the impairment in NCF4, p22phox and Rac2 are NOX subunits [68].
mice at low concentration because CO activates the gene required to
synthesize mitochondria and upregulates the nuclear-encoded heme 4.1.5. Intracellular calcium dysregulation
oxygenase (HO)-1. HO-1 is an antioxidant enzyme that protects against Dox causes dysregulation of calcium levels, leading to an increase in
cardiomyocyte apoptosis and HO-1/CO stimulates the Akt, deactivates intracellular calcium, the generation of ROS, and induces apoptosis in
the glycogen synthase kinase-3β which allows the nuclear translocation cardiomyocytes, whereas Ca2+ chelators inhibit the ROS generation and
of the nuclear factor erythroid 2-related factor 2 (Nrf2) in the nucleus to apoptosis in cardiac muscle cells [69]. During the metabolism of Dox,
activates the GATA-4 gene and prevents the apoptosis [52]. there is the formation of the toxic metabolite DOXOL, which inhibits the
sodium-calcium exchanger channel [70,71]. The sodium-calcium
4.1.3. Fe-Dox complex exchanger channel plays an important role in regulating heart contrac­
Dox is a positively charged drug and plasma membrane is negatively tility through the sarcoplasmic reticulum calcium pump and increases
charged; thus, it can easily interact with each other [53]. The positive the L-type calcium channel activity [72]. Further, Dox-induced cardio­
charge of Dox facilitates good affinity towards the iron and forms the myopathy involves alteration in the calcium exchange regulating gene,
Dox-Fe complex, which alters the metabolism of iron. This complex which affects ryanodine receptor (RyR), calcium storage gene, Sar­
further interacts with the free oxygen and enhances ROS production, co/endoplasmic reticulum ATPase (SERCA2a), and phospholamban,
responsible for the induction of cardiotoxicity with this complex [54]. In thereby causing impairment in both systolic and diastolic functions of
normal body physiology, the free iron content levels are not enough to the heart. In addition, study reported that the elimination of Dox
bind with Dox and therefore, no cardiomyopathy occurs [55]. Dox - Fe metabolite is difficult in comparison to Dox. This metabolite is retained
interaction does not arbitrate alteration in the iron metabolism, but it on the cardiac muscle cell and causes significant calcium dysregulation,
acts on the transferrin protein, which helps transport and binds to affecting cardiac muscle [73].
intracellular iron [56]. Dox metabolite doxorubicinol removes the iron Apart from this, one more pathway is involved with the rise in
from the catalytic Fe-S cluster of the cytoplasmic aconitase (also known intracellular calcium, wherein calcium activates the calpains, which are
as iron regulatory protein- 1) and converts this enzyme into the calcium-dependent proteases. An increased amount of calcium stores in
non-functional protein [56,57]. Therefore, the stability of transferrin the sarcoplasmic reticulum’s cardiac muscles was observed during
mRNA increases and prevents the translation of iron sequestration oxidative stress and causes leakage of calcium ions that activate the
protein. Consequently, there is a reduction in the IRP-1 (iron regulatory calpains. These calpains cleave the caspase-12, one of the apoptotic
protein) gene, an increase in the free iron level leads to the generation of factors and induce apoptosis of cardiac cells and a similar mechanism
the ROS and ultimately led to cardiotoxicity. The increase in sensitivity was reported with the administration of Dox [74]. Dox-induced car­
to Dox-induced cardiotoxicity in mice leads to a decrease in the iron diomyopathy is also related to the destruction of cardiac muscle pro­
regulatory gene HFE (Human homeostatic iron regulator protein) [58]. teins. This destruction occurs because of the stimulation of calpains.
This iron regulatory gene defect leads to increased iron accumulation in These calpains degrade the titin that is the largest protein and principal
the myocardium and causes cardiotoxicity [59]. The HFE gene plays an component of heart sarcomere [71]. Some positive cardiac response is
essential role in the regulation of circulating iron uptake [60]. also seen via the inhibition of calpains and reported that the depletion of
It is essential to determine the alteration in the iron storage in cancer CARP (cardiac ankyrin repeat protein) results in sarcomeric structure
patients because they are taking the chemotherapy treatment. In cancer stability. This protein is a negative regulating cardiac gene and causes
patients, there are abnormal blood losses and deficiency in nutrients; instability of sarcomere arrangement [71,75].
therefore, they may need a blood transfusion and iron supplementation Dysregulation in the calcium channels leads to an increase in the
[61]. The adult and pediatric patients of leukemia show increased iron calcium burden, which causes a decrease in the mitochondria’s calcium
overload during chemotherapy [60]. Therefore, careful monitoring is storage capacity. This leads to the activation of the cyclosporin-sensitive
required in patients with Dox. calcium channel, which exacerbates the calcium burden [76]. Thus, an
increase in calcium cytoplasmic concentration leads to mitochondrial
4.1.4. NADPH dependent ROS dysfunction and apoptosis [77].
Zhao et al., reported that NOX are the enzymes that were used as a Apart from this, there are many other pathways involved in calcium
source of cardiac ROS generation [62]. This enzyme shows a dysregulation and apoptosis through Dox. Dox increases the free Ca2+
multi-component enzyme complex that comprises membrane-bound levels, which activates the calcium-dependent activated signaling and
cytochrome b-558. This cytochrome is a heterodimer of gp91phox and leads to cell death [78]. Further, increased Ca2+ leads to the activation
p22phox, cytosolic regulatory subunits p47phox and p67phox, and Rac1 of the CaMKII (calmodulin activated protein kinase II) and the PLN
(small GTP-binding protein) [63]. These enzymes play an important role (phospholamban). It enhances the apoptosis of cardiomyocytes through
in forming Dox semiquinone radical from the transference of one elec­ the activation of caspase 3 & 9, H2O2, increased terminal deoxy­
tron from the NADPH to Dox, which is similar in the NOS pathway [32]. nucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL)
There are seven isoforms of NOX and NOX2 isoform plays a crucial role positive cells, and ROS generation.
in Dox-induced cardiotoxicity. Studies demonstrated that NOX2 defi­
cient species or gp91phox knockout mice are resistant to Dox-induced 4.2. Role of AMPK (AMP-activated protein kinase) signaling in Dox-
cardiotoxicity with reduced production of superoxide-free radical in induced cardiotoxicity
the cardiac tissue. In contrast, cardiac dysfunctioning like atrophy,
cardiomyocytes programmed cell death and interstitial fibrosis was seen AMPK is a macromolecule protein complex that contains three sub­
in wild-type mice [62,64–66]. units, like one catalytic site α and two regulatory sites are β and γ. α
One of the oxidative stress regulating genes (Rac1) also shows a divided into two subunits, present in the heart such as α1 in the endo­
significant role in NOX stimulation. In this background, a study showed thelial cells and α2 in the cardiac muscle cells. AMPK pathway is blocked
that the deletion of Rac1 gene in cardiomyocytes leads to the alteration after the Dox administration and reduced the ACC (Acetyl- coA
in Dox-induced NOX2 stimulation, DNA fragmentation, programmed carboxylase) enzyme activity in the heart. Dox-induced AMPK signaling
cell death and recovers heart functioning [63]. Further, Rac1 inhibitor inhibition leads to activation of other pathways such as Akt and MAP
NSC23766 shows protection against Dox-induced cardiotoxicity [67]. kinase through genotoxic stress and oxidative stress, which induces DNA
Moreover, SNP (single nucleotide polymorphism) in the NOX complex in damage. Also, AMPK suppression leads to the enhanced energetic stress
patients with Hodgkin-lymphoma causes cardiotoxicity. These patients and hypertrophy in the cardiac tissue [1,79].

4
P.S. Rawat et al. Biomedicine & Pharmacotherapy 139 (2021) 111708

4.3. Role of autophagy cardiac muscle cell apoptosis by inhibiting the pro-apoptotic factors like
Bad [97]. Moreover, Dox downregulates the Akt pathway and induces
Autophagy is a conserved process in eukaryotic cells involved in the caspase-3, which causes apoptosis [32,71,98].
maintenance of cellular homeostasis and survival under normal and Further, Dox upregulates the appearance of death receptors (DR) like
stressful circumstances. It involves removing damaged and long-lived TNFR1, Fas, DR4, and DR5 at both protein and mRNA levels [99].
organelles under both physiological and pathological conditions, thus Toll-like receptor-2 (TLR-2) functions as a novel death receptor,
regarded as a protective mechanism. Importantly, in response to stress, increasing the apoptosis through apoptotic factor, including caspase 8
there is dysregulation of the autophagy process in the heart and can and FADD (Fas-associated death domain). Antibodies of DR and TLR-2
cause cardiac dysfunctions and heart failure [80,81]. Dox causes receptor inhibitors blocks the binding of the HSP 60 protein to the
abnormal upregulation as well as downregulation of genes involved in TLR-2 and it activates the NFκB (proinflammatory nuclear-factor κB),
autophagy thereby results in cardiotoxicity. Dox treatment induces which leads to the production of cytokine, programmed cell death and
autophagy initiation probably through the p53-mediated suppression of altered cardiac functioning after the treatment of Dox. Further, TLR-4 is
GATA-4, which results in the down-regulation of the Bcl-2 protein. In also associated with Dox-induced cardiomyopathy, and it only happens
addition, Dox initiates the autophagy process by increasing the phos­ when HSP-70 interacts with the TLR-4 receptor and causes cardiomy­
phorylation of Bcl-2 that inhibits the interaction of Bcl-2/Beclin1 [82, opathy through the NFκB activation [100–102] and overall mechanisms
83]. Dox administration induces the accumulation of autophagosome of Dox-induced cardiotoxicity is discussed in Fig. 1.
and inhibition of flux through the upregulation of several autophagy
related gene (Atg) expression and downregulating the master tran­ 4.6. Endothelin-1
scription factor EB with altered function of lysosomal biogenesis in
cardiomyocytes led to cell death [84]. Further, Dox inhibits mTOR, Endothelin-1 is also known as preproendothelin-1 (PPET-1) and it is
possibly by stimulating the initiation of autophagy and it is also linked a potent vasoconstrictor produced from the vascular endothelial cells.
with the upregulation of p-53 ultimately led to cardiomyocyte injury. PPET-1 is an immature form of endothelin and endothelial cells convert
Moreover, the accumulation of autophagosomes results in ROS genera­ PPET to PET (Proendothelin) and then into their mature form
tion [85–87]. Dox administration upregulates the genes like Atg5, endothelin-1. After the Dox treatment, the levels of endothelin-1 in­
Atg12, Atg4 and Bad and downregulates the beclin1 protein. Further, crease in cardiac muscle cells and cause hypertrophic cardiomyopathy.
LC3B is an important marker of autophagy that is also upregulated after Bosentan is an endothelin A/B receptor antagonist which produces
Dox administration and causes cardiac damage [88]. In contrast, cur­ cardiac protective action over the Dox administration [103,104]. Their
cumin protects from cardiac damage by blocking apoptosis and enhance antagonist shows cardioprotection via reduced TNF-α and Bax levels,
autophagy by regulating the c-Jun N-terminal kinases (JNK) mediated decreased lipid peroxidation and increase in the GATA4. Moreover, dual
pathway [1]. Altogether, the impaired autophagy can induce car­ antagonists of the receptor (ETA/ETB) produce more significant pro­
diomyocyte death. tection [104].

4.4. Role of creatine kinase 4.7. Fibrosis

Creatine kinase is a protein present in the number of tissues in the Treatment with Dox in cancer patients leads to blockade of mRNA
body and acts as an energy reservoir. But in cardiac tissues, only CK-MB transfer and inhibits protein synthesis of MMP-1 (Matrix metal­
isoenzyme of creatine kinase is expressed. Dox-induced oxidative stress loprotease -1) in tumor cells. It is helpful in cancer because of this
damages the creatine isoenzyme in the presence of ferrous iron, which mechanism; there is a reduction in the tumor cell’s mobility. However, it
leads to the generation of the peroxynitrite free radical. This enzyme activates other MMP’s like MMP-2 and MMP-9, which show toxicity in
aids in the preservation of the energy with the conversion of creatine to the heart by increasing collagen formation in the cardiac tissues [105,
phosphocreatine, which use ATP as a substrate [71]. 106]. An increase in collagen leads to myocardium fibrosis. These two
MMP’s also activate the TGF-β and phosphor-SMAD3 signaling and this
4.5. Apoptosis enhanced collagen deposition in the cardiac muscle cells [1].

Dox stimulates both intrinsic as well as extrinsic pathways of 4.8. Role of epigenetics in doxorubicin-induced cardiotoxicity
apoptosis [45,89]. These pathways are responsible for the apoptosis of
cardiac muscle cells due to an imbalance in oxidant and anti-oxidant Epigenetics is a heritable phenotypic change in which there is no
[90]. Dox administration leads to the increase in oxidative stress, involvement in altering DNA base-pair sequence (genotype) [107].
which activates the HSF-1 (heat shock factor 1) thereby induces HSP-25 Epigenetic modifications cause alteration in the gene function. It is a
(heat shock protein), which stabilizes the p53 protein responsible for the natural process in which alterations occur regularly with several factors
generation of proapoptotic factors such as FasL, Fas, c-Myc, and p53 like age, environment, external, and disease state. Two main epigenetic
which cause cardiac muscle cell death [90–93]. Moreover, Dox admin­ modifications involved in many diseases and natural growth include
istration also increased the circulating HSP-70 and its expression in DNA methylation and histone protein modification. These two are
cardiac tissue of mice thus causes inflammation and fibrosis [94]. In epigenetic mechanisms, which induce phenotypic changes and control
contrast, other HSP proteins such as HSP-27, HSP-10, HSP-20, HSP-22 the gene expression by blocking proteins that attach to the silencer re­
and HSP-60 are also protective and protect from apoptosis and maintain gion of the DNA [108].
myocardial functioning [89,95]. Recently Lan et al. reported that the Every cell of the human body contains the same DNA molecule,
overexpression of HSP-22 in the cardiac tissue could reduce the cardiac which develops from the individual cell at impregnation. Highly ar­
injury in response to Dox administration by inhibiting TLR-4/NLRP3 ranged epigenetic mechanisms facilitate the patterning required to
activation [96]. Increased level of HSP-27 shows the maintaining ac­ ensure normal human development and support stable regulation of
tion over the Dox-induced oxidative stress and mitochondrial dysfunc­ appropriate patterns of a natural phenomenon of expression in diverse
tioning [89]. Increased expression of HSP-10 and HSP-60 are cell types. Regulation of these mechanisms by a more number of proteins
responsible for the increased post-translational modification of Bcl-2, that begin, recite and erase specific epigenetic alterations. Thus, it de­
which leads to activation of the anti-apoptotic pathway [95]. HSP-20 fines where and when the transcriptional machine can approach the
also has a significant role in cardiac protection via activation of the chief DNA sequences to run regular growth and differentiation in the
Akt pathway. It is an important pathway for cell survival and it prevents growing embryo and fetus [109]. A number of epigenetic marks work in

5
P.S. Rawat et al. Biomedicine & Pharmacotherapy 139 (2021) 111708

Fig. 1. This picture shows the different mechanisms of action of Dox-induced cardiotoxicity inside the cell and outside the cell.

whole human cells but run a specific gene expression. DNA methylation mitochondrial dysfunctioning is one of the important mechanisms of
occurs at the CpG island, covalent alterations of histone proteins, reg­ Dox induced cardiotoxicity [116].
ulatory ncRNAs (noncoding) and other complementary mechanisms Dox downregulates the DNA methyltransferase 1 (DNMT1) enzyme
maintaining higher order of chromatin association inside the cell nu­ activity which leads to reduction in the DNA methylation process. This
cleus [110]. Following there are the three most important epigenetic hypomethylation leads to dysregulation in the mitochondrial genes like
modifications reported behind Dox-induced cardiotoxicity. peroxisome proliferator-activated receptor-gamma coactivator (PGC)-1-
alpha (PGC-1α), nuclear respiratory factor 1 (NRF-1) and mitochondrial
4.8.1. DNA methylation transcription factor A (TFAM) unit in the rat heart after the adminis­
DNA methylation sometimes activates and inhibits gene expression tration of Dox. These gene products are encoded with both nuclear and
based on physiological or pathological conditions. The human genome mitochondrial genomes [117]. In DNA methylation, mitochondrial ge­
contains more than 30 million base pairs of CG dinucleotides (CpGs). For nomes are regulated by DNMT1, but there is downregulation of DNMT1
methylation, CpG sites are an important target, comprised of the cova­ after Dox exposure because Dox increases the oxidative stress, down­
lent binding of a methyl group to the 5’ position of the cytosine (C). CpG regulates the DNMT1 and leads to mitochondrial dysfunctioning that
methylation arrangement is associated with transcriptional silencing affects the DNA methylation [118,119].
arrangement observed in 60–90% of the human genes [111]. It is a
process in which there is the addition of methyl moieties to the DNA 4.8.5. Alterations in histone modification
molecule and this addition leads to a change in DNA activity without Apart from DNA methylation, histone modifications are also
disturbing the DNA base pair sequence. responsible for Dox-induced cardiotoxicity. Suppression of histone
deacetylase (HDAC) produces cardiac toxicity through the initiation
4.8.2. Histone modifications Rac1, GTP-binding protein, a subunit of NOX and p53 upregulation. The
Histones are one of the most important proteins of the body, which initiation of these units leads to apoptosis and hypertrophy. After
provide a structural role. Highly basic proteins found in eukaryotic cell treatment with Dox, there is abnormal upregulation and downregulation
nuclei that package and order the DNA into structural units called nu­ of HDAC, leading to cardiotoxicity [67]. Dox causes the upregulation of
cleosomes [112,113]. Histones are the major protein component of several HDACs, including HDAC4, HDAC5, HDAC6, HDAC7, HDAC10,
chromatin, acting as spools around which DNA wind, and have a sig­ HDAC11 and downregulation of HDAC2. Upregulation of HDAC6 leads
nificant role in gene regulation. In the absence of histone proteins, un­ to the deacetylation of α-tubulin seen in the rat heart after the treatment
wound DNA in chromosomes would be very long. of Dox [120]. In addition, Dox also upregulates the histone lysine
demethylase (KDM3A) and downregulates lysine-specific demethylase 1
4.8.3. ncRNA (non coding RNA) (LSD1) in the H9C2 cardiomyocyte cell line [121]. Moreover, it also
A ncRNA acts directly as well as indirectly to regulate the epigenetic upregulates the histone lysine methyltransferases like lysine N-methyl­
modification. Even though transcription occurs up to 75% of genomic transferase 7 (SET7) and SET- and MYND-domain-containing protein 1
DNA in eukaryotic genomes transcribe, but that only around 2% of these gene (SMYD1) in H9C2 cell line [119].
transcripts are translated into protein; the majority are ncRNAs, which
can be classified according to size and function [114,115]. 4.8.6. Alteration in the ncRNA
A ncRNAs act directly as well as indirectly to regulate the epigenetic
4.8.4. Dox-induced alterations in DNA methylation modifications. [114,115]. Regulatory ncRNAs, including small inter­
In the DNA methylation process, there is a need for enzymes that can fering RNAs (siRNAs), microRNAs (miRNAs), and long ncRNAs
add the methyl group or remove the methyl group for the body’s normal (lncRNAs) which play significant roles in the regulation of gene
functioning. Enzymes like TET (Ten eleven translocation) and methyl­ expression at several levels: transcription, mRNA degradation, splicing
transferases and these enzymes require S-adenosyl methionine (SAM) or and translation [122].
alpha-keto glutarate (α-KG) for the development of 5-methylcytosine or There is also abnormal upregulation and downregulation of the
5-hydroxymethylcytosine. These formations directly show two pro­ ncRNA after the treatment with Dox. There are several ncRNA’s which
cesses like DNA methylation and DNA demethylation. SAM and α-KG are upregulated after Dox administration, like miR-23a, miR-34a, miR-
develops in the mitochondria by a specific metabolic pathway and 140, miR-146a, miR-532, miR-15 and some are downregulated like

6
P.S. Rawat et al. Biomedicine & Pharmacotherapy 139 (2021) 111708

miR-29b and miR-30. The upregulation of miR-15 ncRNA in the H9C2 cardioprotective action. This signaling pathway is dependent on the RNS
cell line is reported to induce apoptosis. This effect appears because of (reactive nitrogen species) and Ras-related C3 botulinum toxin substrate
the suppression of Bmpr1a, which is an important target of miR-15 and 1 (Rac1) [132–134]. Pitavastatin blocks Rac1 was shown in a mouse
BMP and the administration of Bmpr1a agonist protects the cardiac cells model of Dox cardiotoxicity and shows cardiac protective role via a
through the downregulation of miR-15 [119]. reduction in the cardiac muscle apoptosis and regulates the constriction
microRNAs (miRNA/miR) play an important role in Dox-induced function. Other statins also show cardioprotection like lovastatin and
cardiotoxicity because of their role in cardiac functioning like devel­ rosuvastatin [79].
opment, electrical signal conduction and cardiac muscle constrictions
[123]. Several studies show that Dox causes an alteration in the 5.3. β-blockers and angiotensin-converting enzyme (ACE) inhibitors
microRNAs and causes either upregulation and or downregulation of
microRNAs. There was upregulation of microRNAs in rodents heart These drugs are mainly used in the treatment of particular heart and
tissue such as miR-34a, miR-34c, miR-208b, miR-215, miR-216b, and kidney diseases. This drug combination are used to treat high blood
miR-367, miR-21, miR-34a, miR-208a, miR-208b, miR-221, miR-222, pressure [135]. The β adrenergic blockers and ACE inhibitors elicit an
and miR-320a after the Dox administration. In contrast, the down­ important pharmacological role in heart failure management. These
regulated microRNAs are Let-7 g, miR-30a, miR-30c, and miR-30e [71, agents preserve the diastolic heart function in the Dox-treated in­
123,124]. dividuals but left ventricular contraction remains constant, and this
Further studies show that miR21 level is increased in H9C2 cells. This action is mainly achieved by carvedilol. Alone, β-blockers do not show
upregulation of microRNAs indicates the cardioprotective action via cardioprotective action because of lacking antioxidant properties but are
reducing the Dox induces apoptosis. However, in miR21, deficient spe­ useful in combination with carvedilol. Their mechanism of action re­
cies of rat shows cardiotoxic effects after Dox treatment. Car­ mains unclear; how it improved cardiac muscle contraction [136].
dioprotective action via miR21 because it alters the anti-proliferative
factor and B cell translocation gene 2 (BTG2) [125]. Dox administration 5.4. Phosphodiesterases-5 (PDE-5) inhibitor
also upregulates the miR-208a, but this results in cardiotoxicity because
it downregulates the GATA4 gene and increased apoptosis in car­ Drugs that block phosphodiesterase-5 (PDE-5) show a positive
diomyocytes. In deficient species of miR-208a mice, improvement in response in cardiac protection by inhibiting the cardiac muscle
GATA4 and Bcl-2 was seen, which leads to a reduction in apoptosis apoptosis, preserving the mitochondrial membrane potential, regulating
[126]. Dox also upregulates the expression of miR-146a and down­ myofibrillar integrity and prevent ventricular dysfunction and protect
regulates the ErB2 receptor tyrosine kinase 4 (ErB4). This receptor is an from ST-interval prolongation in Dox treated mouse model [137].
important component of neuregulin-1-ErB signaling. This signaling Likewise, Sildenafil, a PDE-5 inhibitor, demonstrated a protective effect
pathway plays critical role in cell survival, but Dox treatment down­ in Dox-induced cardiotoxicity by modulating the NO/cyclic GMP,
regulates this pathway and promotes apoptosis in cardiomyocytes mitochondrial K+ATP channel and oxidative stress [138].
[127].
GATA-6 gene is a transcription factor and highly expressed in the 5.5. α1-Adrenergic agonists
heart and plays an essential role in heart development [128]. Moreover,
Dox also downregulates miR30 family microRNAs and alters the This agonist comes under the sympathomimetic drug class that ac­
beta-adrenergic and mitochondrial apoptosis pathway. GATA-6 gene tivates explicitly the α adrenergic receptors. Cardiac α1-adrenergic re­
acts as a precursor for the downregulation of the miR30 and causes ceptors show the cardioprotective role against Dox-induced
apoptosis. Increased expression of this miR30 protects cardiomyocytes cardiotoxicity. α1-adrenergic receptor agonists like phenylephrine are
through the modulation of GATA-6. Overall, miRNAs play an essential useful because they reduce the apoptosis, interstitial fibrosis and cardiac
role in Dox-induced cardiotoxicity [128]. muscle impairment caused by Dox. This protective action is exerted
through the generation of the anti-apoptotic factors, Bcl2 and maintains
5. Therapeutic strategies mitochondrial functioning [139,140].

5.1. Metformin activates AMPK signaling pathway to reduce Dox 5.6. Dexrazoxane
cardiotoxicity
It is a cyclic derivative of EDTA that quickly penetrates through the
Metformin is a drug that belongs to biguanide class, with anti­ plasma membrane [141]. It is a free radical scavenger used as an
hyperglycemic property and widley used to treat type 2 diabetes mel­ adjunctive agent and shows chelator action on iron-mediated oxygen-­
litus [129]. Metformin shows cardioprotective effects in rat free radical activation as well as lipid peroxidation. With this chelate
cardiomyocytes in Dox-treated rats. Dox deactivates the AMPK signaling reaction, there is a reduction in iron-free radicals levels; thus, it shows
pathway and decreases platelet-derived growth factor receptor (PDGFR) the cardioprotective action [32,71,142].
expression. In contrast, metformin treatment activates the AMPK
signaling pathway by increasing the PDGFR expression [130]. Another 5.7. Resveratrol
study showed that metformin reduces the levels of H2O2, which offers a
protective mechanism on mitochondrial damage through the AMPK The polyphenolic stilbene 3,5,40-trihydroxy-trans-stilbene (resver­
activation. It also maintains autophagy markers like LC3B-II and p62 in atrol) is produced by many plants and is a constituent of red wine.
Dox-treated rats, thus improving cardiac functioning [79]. Resveratrol is a naturally occurring agent having antioxidant and po­
tential chemopreventive activities. Resveratrol shows some pharmaco­
5.2. Statins logical effects, including anti-inflammatory effects and inhibits
cyclooxygenase and hydroperoxidase functions [143]. Moreover,
Statins are the agent which inhibits the rate-limiting enzyme HMG resveratrol has cardioprotective activity through the activation of AMPK
CoA reductase in cholesterol biosynthesis. Statins help in lowering of [79]. AMPK attenuates oxidative stress, apoptosis and ameliorating
lipid levels and are used as an antihyperlipidemic drug. Statins are fibrosis in Dox-induced cardiotoxicity. It is a potent antioxidant that
commonly used in cardiovascular disorders, including atherosclerosis reduces ROS levels and enhances antioxidant levels like superoxide
and coronary heart disease [131]. Reports suggest that statins also dismutase (SOD) and catalase (CAT) activity and also shows the
activate the AMPK signaling in cardiac muscle and show anti-inflammatory activity [144]. In addition, Pterostilbene, a natural

7
P.S. Rawat et al. Biomedicine & Pharmacotherapy 139 (2021) 111708

analog of resveratrol, prevents Dox-induced cardiotoxicity through the 5.13. MiR-181c protects cardiomyocyte injury by preventing cell
activation of PGC-1α, reduces oxidative stress via enhancing AMPK and apoptosis through PI3K/Akt signaling pathway
SIRT1 cascades [145].
Apoptosis in the cardiac muscle is responsible for heart failure, which
results in an increased risk of mortality related to cardiovascular dis­
5.8. Vitamin E
orders. Upon treatment with Dox, there is downregulation in the MiR-
181c, one of the microRNAs that helps in the muscle cell survival via
Four tocopherols and four tocotrienols are combined to make
PI3K/Akt signaling pathway. MiR-181c, downregulation, leads to the
vitamin E, which is fat-soluble [146,147]. Vitamin E having an antiox­
activation of the toxic markers like TNF-α, Fas and IL-6 and causes
idant property that protects the cell membranes from ROS [147]. A
cardiotoxicity, whereas treatment with MiR-181c inhibits apoptosis in
study shows the protective action of Vitamin E over Dox-induced car­
the Dox treated H9C2 cells [159].
diotoxicity. Vitamin E is helpful in acute cardiotoxicity caused by Dox.
Other antioxidants show limited benefit over the cardiotoxicity like
5.14. Cardioprotective effects of 20(S)-ginsenoside Rh2 against Dox-
vitamin C, selenoorganic compound PZ51 and reduced glutathione,
induced cardiotoxicity
ambroxol, ursolic acid and oleanolic acid [71,148].
From ancient times, ginseng has been commonly used as a recog­
5.9. Probucol nized traditional Chinese medicine. Its principal active constituent is
ginsenosides, which contain various pharmacological effects, including
Probucol is an antihyperlipidemic agent which lowers lipid levels activating immune function, enhancing cardiovascular health,
and also has an effective anti-oxidant action. It shows cardioprotective increasing resistance to stress, improving memory and learning, and
effect via a reduction in cardiac muscle injury and heart failure without developing social functioning and mental health in healthy individuals
interrupting the antineoplastic effect of Dox [149,150]. and chemotherapy. Ginsenoside Rh2 (Rh2) is one of the major bioactive
ginsenosides from Panax ginseng [160]. Pretreatment with Rh2 attenu­
ates Dox-induced cardiomyocyte injury’s cell-killing effect and enhances
5.10. Allicin the cardiac cell’s survival rate in a dose-dependent fashion. Further, Rh2
treatment restores the lactate dehydrogenase, creatine kinase and
Allicin is an active constituent of garlic and it is an organosulfur aspartate transaminase in the serum, thus shows cardioprotective action
containing compound [151]. It has several pharmacological effects, [161]. Treatment with Dox also decreases antioxidants like SOD, CAT
including antimicrobial, antioxidant, anticarcinogenic, and antifungal and glutathione and increased the malondialdehyde, whereas Rh2 re­
[151]. It is a cytoprotective agent against Dox-induced cardiotoxicity. A stores this imbalance [162].
study confirmed that allicin efficiently reduces cardiac oxidative stress,
reduces inflammation, and reduces apoptosis in the presence of acute 5.15. Curcumin suppresses doxorubicin-induced cardiomyocyte
Dox intoxification. Dox deactivates the antioxidant substances like pyroptosis
glutathione peroxidase, SOD and CAT and thus, causes oxidative dam­
age and cardiac apoptosis; however, allicin restores the antioxidant level Curcumin is a biphenolic compound and it is a major active con­
[152,153]. stituent of turmeric [163]. Curcumin is widely used as a coloring agent
and food additive and also curcumin has presented some therapeutic
effects [164]. Curcumin shows protective effects against Dox-induced
5.11. Adiponectin agonist ADP355 attenuates the Dox-induced
cardiotoxicity. Dox increases the levels of ROS, lipid peroxidation,
cardiotoxicity
reduction in antioxidant levels and induction of pyroptosis. Dox also
downregulates the PI3K/Akt/mTOR pathway, which leads to car­
Adiponectin is a protein hormone responsible for the regulation of
diotoxicity. Bcl2 is an antiapoptotic factor that is suppressed after Dox
glucose and the breakdown of fatty acids. It is encoded as ADIPOQ gene
treatment and promoted the expression of BAX, which causes mito­
in humans. It is mainly located in the adipose tissue and also in muscles
chondrial cell death.
and brain [154].
Curcumin is a natural compound derived from the plant Curcuma
Adiponectin agonists prevent cardiotoxicity induced by Dox via
longa and has an anti-inflammatory effect. After treatment with curcu­
reducing the serum creatine kinase level, lactate dehydrogenase and
min, it reduces Bax levels and prevents mitochondrial cell death [165].
hydroxybutyrate dehydrogenase level. It also shows the antiapoptotic
Further, curcumin inhibits pyroptosis by modulating pro-pyroptosis
action by lowering the TUNEL positive cells and cleaved caspase-3,
markers like NLRP3, caspase1 and IL-18 and downregulates the
associated with the reduced Bcl2/Bax level in cardiac tissue. Another
inflammasome [166]. Curcumin also reduces the levels of CK, LDH and
protective mechanism of adiponectin agonist is to enhance the antioxi­
AST in Dox-induced cardiac injury. Dox also increases autophagy in the
dant level, which is reduced with the induction of the Dox like Nrf2 and
cardiomyocytes, which leads to cardiac dysfunction. In contrast, cur­
SOD2. It also blocks ROS in the H9C2 cell of the cardiac muscle and
cumin reduced autophagosome levels (GFP-LC3 puncta and Beclin1),
activates the sirtuin 2 signaling pathways, thereby protecting the car­
which initiates the maintenance of cardiac functioning after the Dox
diac muscle from apoptosis and oxidative stress [155].
treatment [167].
Dox also downregulates the PI3K/Akt/mTOR pathway and this
5.12. Erythropoietin pathway is necessary for cell survival and cell differentiation. The
downregulation of this pathway causes apoptosis in the cardiomyocytes.
Erythropoietin is a hormone which is generated from healthy kidney Curcumin upregulates this pathway and shows cardioprotection over
and a small amount by the liver. Erythropoietin is a glycoprotein cyto­ the Dox-induced cardiac toxicity. Curcumin also reduced pyroptosis and
kine mainly secreted from the kidney at the time of cellular hypoxia autophagy via activation of the Akt/mTOR pathway [167].
[156]. Some antianemic drugs also show cardioprotection against
Dox-induced cardiotoxicity. Erythropoietin is an antianemic agent 5.16. Visnagin-a new protectant against doxorubicin cardiotoxicity
which is used in anemia caused by chemotherapeutic agents. Erythro­
poietin protects from Dox-induced cardiotoxicity via reducing apoptosis Visnagin is a furochromone which is a derivative of chromone and
and cardiomyopathy when administered prophylactically [157,158]. furan. Visnagin is a major constituent of Ammi visnaga. In traditional

8
P.S. Rawat et al. Biomedicine & Pharmacotherapy 139 (2021) 111708

medicine, visnagin has been used for kidney-associated problems [168]. melatonin also mitigates Dox-induced cardiotoxicity via reduction of
A study shows the protective effect of visnagin in Dox-induced car­ AMPKα2-dependent mitochondrial damage thus modulating the car­
diotoxicity through the inhibition of mitochondrial malate dehydroge­ diomyocytes apotosis [182]. In addition, Lu et al. reported that chrys­
nase (MDH2). Liu et al., identified the role of MDH2 as a novel target for ophanol protects the cardiomyocytes from Dox-induced injury through
reducing Dox mediated cardiotoxicity [169]. MDH2 is an important the inhibition of cardiac apoptosis, mitochondrial damage and cellular
enzyme in the tricarboxylic acid (TCA) cycle. MDH is actively present in PARylation levels [183].
glyoxysomes, mitochondria, peroxisomes, chloroplasts and the cytosol Further, Echinochrome A is a naphthoquinoid pigment derived from
[170] that bind with the malate-aspartate shuttle (MAS), which estab­ sea urchins having antioxidant, antimicrobial, anti-inflammatory and
lishes the primary metabolic pathway for the transfer of reducing chelating abilities. Notably, co-treatment of Echinochrome A with Dox
equivalents from the cytosol into the mitochondria for oxidation. Dox improves the mitochondrial membrane potential, reduces ROS level,
has an affinity towards the MDH2 enzyme and blocks this respective preserve the function of mitochondrial oxidative phosphorylation and
enzyme activity, which possesses a cardioprotective activity in adenosine triphosphate level [184]. In addition, Ghrelin also demon­
Dox-induced cardiotoxicity [171]. strated protective action against Dox-induced cardiotoxicity through the
modulation of TNF-alpha/NF-kappaB pathways and increased the
5.17. Schisandrin B (Sch B) prevents doxorubicin-induced cardiotoxicity mitochondrial anti-apoptosis-related gene protein expression [185].
Overall this indicating that the mitochondria damage is an essential
Sch B the most abundant dibenzocyclooctadiene lignan present in pathological alteration in Dox-induced cardiotoxicity. Besides these
Schisandra chinensis (Turcz.) [172]. It has used in the treatment of ce­ strategies, the administration of fresh mitochondria can be tried and
rebral ischemia and Alzheimer’s disorder [173]. Sch B prevents could be a novel treatment strategy for mitigating cardiac injury.
Dox-induced acute cardiotoxicity by increasing cardiomyocyte gluta­ However, there are no studies were reported with Dox-induced car­
thione redox cycling that eliminates excessive ROS formation [174]. Dox diotoxicity. Interestingly, a study by Shi et al. administered mitochon­
administration causes weight loss in rats; in contrast, Sch B moderately dria through the intravenous route to treat Parkinson’s disease in mice.
prevents weight loss. Furthermore, Dox blocks the mTOR signaling, Notably, the administered exogenous mitochondria is well distributed in
which causes a reduction in cardiac mass while the Sch B restores the several tissues such as brain, liver, kidney, muscle, and the heart, indi­
mTOR signaling [175]. Similarly, Xu et al. reported that Sch B partially cating beneficial effects in multi-systemically mitochondrial diseases
relieves mTOR inhibition in muscle or adipose tissue under Dox treat­ [186].
ment and improves the rat’s body weight. Therefore Sch B shows the
cardioprotective effect over the Dox treatment [176]. 5.19. Other recent strategies to prevent Dox-induced cardiotoxicty

5.18. Treatment strategies focusing on mitochondrial modulation In the present section, we have compiled the agents recently
explored to manage Dox-induced cardiotoxicity. Herein, Sadek et al.
Mitochondrial division inhibitor (mdivi-1) has been tried for several demonstrated the protective effect of proanthocyanidin in Dox-induced
disease conditions, including heart failure and ischemia and reperfusion cardiotoxicity in rats by improving the anti-oxidant levels and inflam­
injury. Gharanei et al. demonstrated that mdivi-1 co-treatment with Dox mation. Proanthocyanidin downregulates the expression of α-SMA, TGF-
improves cardiac function and reduces the infarct size in both naïve β1 and upregulates the CDK4 and Rb protein expression with reduced
conditions and during ischemia/reperfusion injury. mdivi-1 also im­ expression of the NF-kB signaling pathway [187]. Further, Nie et al.
proves the Dox-induced alterations in time taken to depolarisation and reported that the hydrogen sulfide treatment decreases the endoplasmic
hypercontracture of cardiac myocytes [177]. Moreover, Xia et al. re­ reticulum stress, autophagy and upregulates the PI3K/AKT/mTOR
ported that LCZ696 a novel angiotensin receptor neprilysin inhibitor, protein expression resulting in reduced myocardial fibrosis in
improves cardiac function in Dox-induced cardiotoxicty by restoring the Dox-treated rats [188]. Tranilast (anti-allergy drug) improves
cardiac function, mitochondrial morphology, improves mitochondrial Dox-induced alterations in myocardial hypertrophy and cardiac
respiration complex I activity with increased adenosine triphosphate dysfunction by suppressing chymase expression, reducing the angio­
content. These protective effects were demonstrated by inhibiting tensin II levels, and modulating the antioxidant level, thus preventing
dynamin-related protein 1 (Drp1) mediated mitochondrial dysfunction apoptosis and fibrosis of cardiomyocytes [189]. Moreover, Alzahrani
in mice with Dox-induced dilated cardiomyopathy [178]. Recently, et al. demonstrated the protective effect of Kirenol (diterpenoid) a
liensinine a mitophagy inhibitor attenuated Dox-induced cardiac phytochemical extracted from Herba Siegesbeckiae having
dysfunction and apoptosis through the inhibition of Drp1-mediated anti-inflammatory and anti-rheumatic properties. Kirenol treatment
maladaptive mitochondrial fission. In addition liensinine suppresses decreases the cardiac oxidative stress, cardiac remodeling, modulates
mitochondrial fragmentation, mitophagy, oxidative stress, cytochrome the apoptosis process, activates the IGF-IR-dependent p-PI3K/p-AKT and
C leakage, cardiomyocyte apoptosis and improved mitochondrial func­ Nrf2 signaling, thereby preventing the Dox-induced cardiac damage
tion and cardiomyocyte contractile function in Dox-induced cardiac [190]. Notably, a medicinal plant Malva verticillata (Malvaceae) pos­
injury [179]. sesses anti-inflammatory and antioxidant activities. Its methanolic
Furthermore, Xu et al. demonstrated the protective effect of luteolin extract from leaf exhibited beneficial effects in Dox-induced cardiac
in Dox-induced cardiotoxicity by augmenting autophagy in mitochon­ injury by improving the levels of cardiac-specific biochemical parame­
dria. Luteolin is a phytochemical derived from vegetables and fruits ters [191]. Likewise, alk-A (N-p-coumaroyl-4-aminobutan-1-ol) and
having anti-oxidative, anti-tumorigenic, and anti-inflammatory prop­ alk-B (hippophamide) pretreatment also reduce oxidative stress,
erties. Luteolin improves the Dox-induced alterations in defective decrease the expression of cleaved-caspase-3, activates Jun N-terminal
mitochondrial autophagy, mitochondrial dysfunction and car­ kinases and improves the mitochondrial function in Dox-induced car­
diomyocyte injury and apoptosis via stimulating autophagosome for­ diotoxicity [192].
mation and improving lysosomal generation through a Drp1/mTOR/ Moreover, Ahmed et al. presented the beneficial effects of methyl
TFEB-dependent mechanism [180]. In addition, melatonin also pro­ gallate in Dox-induced alterations in oxidative stress, cardiac injury,
tects the cardiomyocytes injury after Dox administration by free radical ECG changes and modulates the levels of cardiac-related biochemical
scavenger activity, modulates the autophagy, mitophagy, mitochondrial parameters in rats [193]. Further, a bioactive peptide Xinmailong
bioenergetics, mitochondrial fission and fusion process and apoptosis extracted from American cockroaches decreases Dox-induced oxidative
[181]. Melatonin is a natural hormone produced by the pineal gland. stress, increased antioxidant activity, upregulates HO-1 expression,
Recently, Song et al. supporting this evidence, demonstrated that restored lysosomal function and improved autophagy flux block in

9
P.S. Rawat et al. Biomedicine & Pharmacotherapy 139 (2021) 111708

Dox-treated H9c2 cells [194]. Another peptide Szeto-Schiller (SS)31 is Funding sources
known to have antioxidant activity and selectively target the inner
mitochondrial membrane to decrease the mitochondrial reactive oxygen Central University of Punjab, RSM grant-2020 (GP-25).
species generation. SS31 demonstrated the cardioprotective effect via
stabilization of mitochondrial membrane potential, reduces myocardial Conflict of interest statement
apoptosis and fibrosis in Dox-treated groups [195]. Furthermore, oil
from seeds exerted cardioprotective effects against Dox-induced car­ The authors declare that there are no conflicts of interest.
diotoxicity in rats by increasing the glutathione, reducing the MDA, and
restoring the various cardia-related biochemical parameters (CK,
Acknowledgements
CK-MB, LDH, AST) and ECG changes with improvement in histopatho­
logical alteration [196]. Focusing oxidative stress, Hashish et al. re­
Authors would like to acknowledge the funding from Central Uni­
ported that pretreatment of quercetin decreases the oxidative stress,
versity of Punjab, Bathinda, under the Research Seed Money (RSM)
MDA level and serum cardiac troponin I in Dox-treated rats. It also re­
grant-2020 (GP-25). All authors read and approved the final manuscript.
stores the ECG changes, histopathological alteration with reduced
apoptotic index in the Dox-treated group [197].
References
Further, Dox treatment increases cardiac xanthine oxidase activity
leading to increased hydrogen peroxide production, upregulation of [1] K. Renu, V.G. Abilash, P.B. Tirupathi, S. Arunachalam, Molecular mechanism of
apoptotic and ferroptosis pathways, ultimately leading to impaired left doxorubicin-induced cardiomyopathy - an update, Eur. J. Pharmacol. 818 (2018)
ventricular function. Xanthine oxidases inhibition by drugs such as 241–253.
[2] C.G. Nebigil, L. Désaubry, Updates in anthracycline-mediated cardiotoxicity,
febuxostat/topiroxostat restores all these alteration in Dox-treated mice Front. Pharmacol. 9 (2018) 1262.
thus maintaining cardiac energy metabolism [198]. Wang et al. used [3] N. Zhao, Q. Li, H. Sui, H. Zhang, Role of oxidation-dependent CaMKII activation
calycosin-triblock copolymer nanomicelles to treat Dox-induced car­ in the genesis of abnormal action potentials in atrial cardiomyocytes: a
simulation study, BioMed Res. Int. 2020 (2020), 1597012.
diotoxicity in H9C2 cells. Interestingly, treatment with nanomicelles
[4] P.A. Henriksen, Anthracycline cardiotoxicity: an update on mechanisms,
reduces the ROS and RNS generation, downregulates the expression of monitoring and prevention, Heart 104 (12) (2018) 971–977.
Bax and p53 and upregulates the Bcl-2 and ERp57 in Dox treated car­ [5] M. Csapo, L. Lazar, Chemotherapy-induced cardiotoxicity: pathophysiology and
prevention, Clujul Med. 87 (3) (2014) 135–142.
diomyocytes [199].
[6] G. Curigliano, D. Cardinale, S. Dent, C. Criscitiello, O. Aseyev, D. Lenihan, C.
Chatterjee et al. explored the role of telomerase overexpression in M. Cipolla, Cardiotoxicity of anticancer treatments: epidemiology, detection, and
cardiotoxic effects of Dox. Telomerase canonically preserves telomeres management, CA Cancer J. Clin. 66 (4) (2016) 309–325.
during cell division however is silenced in adult hearts. Cardiomyocytes [7] L. Gianni, E.H. Herman, S.E. Lipshultz, G. Minotti, N. Sarvazyan, D.B. Sawyer,
Anthracycline cardiotoxicity: from bench to bedside, J. Clin. Oncol. 26 (22)
are non-dividing cells where telomerase convenes pro-survival traits and (2008) 3777–3784.
probably exhibits in the ROS detoxification. In this study, the authors [8] L. Gianni, E.H. Herman, S.E. Lipshultz, G. Minotti, N. Sarvazyan, D.B. Sawyer,
used adeno-associated virus-mediated gene therapy for the long-term Anthracycline cardiotoxicity: from bench to bedside, J. Clin. Oncol.: Off. J. Am.
Soc. Clin. Oncol. 26 (22) (2008) 3777.
expression of telomerase against dox-induced cardiotoxicity. The over­ [9] E. Salvatorelli, P. Menna, M. Chello, E. Covino, G. Minotti, Modeling human
expressed telomerase presented beneficial effects through the inhibition myocardium exposure to doxorubicin defines the risk of heart failure from low-
of apoptosis, which improves cardiac function and mitochondrial dose doxorubicin, J. Pharmacol. Exp. Ther. 362 (2) (2017) 263–270.
[10] J. An, M.S. Sheikh, Toxicology of trastuzumab: an insight into mechanisms of
morphology [200]. Further, Compound DanShen Dripping Pill (CDDP) cardiotoxicity, Curr. Cancer Drug Targets 19 (5) (2019) 400–407.
also explored and demonstrated the protective effects in [11] Y. Yang, P. Bu, Progress on the cardiotoxicity of sunitinib: prognostic significance,
Dox/Iso-induced (isoproterenol) induces cardiac damage. CDDP restores mechanism and protective therapies, Chem. Biol. Interact. 257 (2016) 125–131.
[12] C. Yuan, H. Parekh, C. Allegra, T.J. George, J.S. Starr, 5-FU induced
the left ventricular ejection fraction, fractional shortening, increases
cardiotoxicity: case series and review of the literature, Cardiooncology 5 (2019)
antioxidant enzymes and inhibits fibrosis, inflammation and cell 13.
apoptosis [201]. Karabulut et al. demonstrated the protective effect of [13] Z. Chustecka, Cardiotoxicity of imatinib is a" surprise, Nat. Med. 24 (2006)
(Published on line July).
thymoquinone in Dox-induced cardiotoxicity in rats. This beneficial
[14] E.A. Hawkes, A.F. Okines, C. Plummer, D. Cunningham, Cardiotoxicity in patients
effect is exhibited through the maintenance of antioxidants level, treated with bevacizumab is potentially reversible, J. Clin. Oncol. 29 (18) (2011)
downregulation of HSP-70, HSP-90, GRP78 and caspase-3 levels that e560–e562.
was upregulated in cardiac after Dox treatment [202]. Altogether, it is [15] V. Martínez-Mateo, M. Anguita, L. Del Campo, A. Paule-Sáncchez, A. Case, Report
of cisplatin-induced cardiotoxicity: a side effect to monitor closely, J. Heart
indicating that management of Dox-induced cardiotoxicity by phyto­ Health 3 (2) (2017).
chemicals is an emerging approach. [16] M.T. Kelleni, M. Abdelbasset, Drug Induced Cardiotoxicity: Mechanism,
Prevention and Management, IntechOpen, London, UK, 2018.
[17] M.A. Ayza, K.A. Zewdie, B.A. Tesfaye, D.Z. Wondafrash, A.H. Berhe, The role of
6. Conclusion antioxidants in ameliorating cyclophosphamide-induced cardiotoxicity, Oxid.
Med. Cell. Longev. 2020 (2020), 4965171.
The anthracycline anticancer drug doxorubicin (DOX) is widely [18] F. Pizzino, G. Vizzari, C.A. Bomzer, R. Qamar, S. Carerj, C. Zito, B.K. Khandheria,
Diagnosis of chemotherapy-induced cardiotoxicity, J. Patient Cent. Res. Rev. 1 (3)
prescribed to treat tumors such as breast, ovary, leukemia, lymphoma (2014) 121–127.
and other malignant tumors in both adults and children. However, the [19] C.A. Frederick, L.D. Williams, G. Ughetto, G.A. van der Marel, J.H. van Boom,
Dox administration leads to cardiac toxicity, resulting in increased A. Rich, A.H. Wang, Structural comparison of anticancer drug-DNA complexes:
adriamycin and daunomycin, Biochemistry 29 (10) (1990) 2538–2549.
mortality risks and thus limiting its wide clinical applications among
[20] A. Fujiwara, T. Hoshino, J.W. Westley, Anthracycline antibiotics, Crit. Rev.
cancer patients. Multiple mechanisms are involved in Dox-induced Biotechnol. 3 (2) (1985) 133–157.
cardiotoxicity, including oxidative stress, impaired mitochondrial [21] R.B. Weiss, The anthracyclines: will we ever find a better doxorubicin? Semin.
Oncol. 19 (6) (1992) 670–686.
function, alterations in iron regulatory protein, deregulation of Ca2+
[22] F.A. Fornari, J.K. Randolph, J.C. Yalowich, M.K. Ritke, D.A. Gewirtz, Interference
homeostasis with impaired apoptosis. Therefore, targeting these alter­ by doxorubicin with DNA unwinding in MCF-7 breast tumor cells, Mol.
ations using allicin, metformin, enalapril, adiponectin, visnagin and Pharmacol. 45 (4) (1994) 649–656.
schisandrin B and other novel phytochemicals has shown protective [23] R.L. Momparler, M. Karon, S.E. Siegel, F. Avila, Effect of adriamycin on DNA,
RNA, and protein synthesis in cell-free systems and intact cells, Cancer Res. 36 (8)
effects against Dox-induced cardiotoxicity and could reduce the mor­ (1976) 2891–2895.
tality rate among cancer patients. [24] P.A. Speth, Q.G. van Hoesel, C. Haanen, Clinical pharmacokinetics of
doxorubicin, Clin. Pharm. 15 (1) (1988) 15–31.
[25] M. Polovich, J. White, L. Kelleher, Chemotherapy and biotherapy guidelines and
recommendations for practice Pittsburgh, PA: Oncology Nursing Society. 16 HOW
SUPPLIED/STORAGE AND HANDLING How Supplied VIDAZA (azacitidine for

10
P.S. Rawat et al. Biomedicine & Pharmacotherapy 139 (2021) 111708

injection) is supplied as a lyophilized powder in 100 mg single-use vials packaged [53] H. Kappus, H. Muliawan, M.E. Scheulen, In vivo studies on adriamycin-induced
in cartons of 1 vial (NDC 59572–102-01). Storage Store unreconstituted vials at lipid peroxidation and effects of ferrous ions, Dev. Toxicol. Environ. Sci. 8 (1980)
25C (77 F); excursions permitted to 15–30C (59–86 F)(See USP Controlled Room 635–638.
Temperature), Handling and Disposal Procedures for proper handling and [54] J.M. Gutteridge, Lipid peroxidation and possible hydroxyl radical formation
disposal of anticancer drugs should be applied. Several guidelines on this subject stimulated by the self-reduction of a doxorubicin-iron (III) complex, Biochem.
have been published, (2005) 1–4. Pharmacol. 33 (11) (1984) 1725–1728.
[26] D.W. Northfelt, F.J. Martin, P. Working, P.A. Volberding, J. Russell, M. Newman, [55] G. Minotti, P. Menna, E. Salvatorelli, G. Cairo, L. Gianni, Anthracyclines:
M.A. Amantea, L.D. Kaplan, Doxorubicin encapsulated in liposomes containing molecular advances and pharmacologic developments in antitumor activity and
surface-bound polyethylene glycol: pharmacokinetics, tumor localization, and cardiotoxicity, Pharmacol. Rev. 56 (2) (2004) 185–229.
safety in patients with AIDS-related Kaposi’s sarcoma, J. Clin. Pharmacol. 36 (1) [56] G. Minotti, S. Recalcati, A. Mordente, G. Liberi, A.M. Calafiore, C. Mancuso,
(1996) 55–63. P. Preziosi, G. Cairo, The secondary alcohol metabolite of doxorubicin
[27] G. McEvoy, E. Snow, J. Miller. American Society of Health System Pharmacists, irreversibly inactivates aconitase/iron regulatory protein-1 in cytosolic fractions
Bethesda AHFS Drug Information, 1st ed., AHFS Drug Information, USA, 2008. from human myocardium, FASEB J. 12 (7) (1998) 541–552.
[28] P. Speth, Q. Van Hoesel, C. Haanen, Clinical pharmacokinetics of doxorubicin, [57] G. Minotti, R. Ronchi, E. Salvatorelli, P. Menna, G. Cairo, Doxorubicin
Clin. Pharmacokinet. 15 (1) (1988) 15–31. irreversibly inactivates iron regulatory proteins 1 and 2 in cardiomyocytes:
[29] M. Hoffmann, R. Bergner, M. Stu¨tzle, M.J. Uppenkamp, R. Foerster, evidence for distinct metabolic pathways and implications for iron-mediated
Pharmacokinetics of doxorubicin in normal weight and obese lymphoma patients, cardiotoxicity of antitumor therapy, Cancer Res. 61 (23) (2001) 8422–8428.
Am. Soc. Hematol. (2010). [58] C.J. Miranda, H. Makui, R.J. Soares, M. Bilodeau, J. Mui, H. Vali, R. Bertrand, N.
[30] M.G. Donelli, M. Zucchetti, E. Munzone, M. D’Incalci, A. Crosignani, C. Andrews, M.M. Santos, Hfe deficiency increases susceptibility to cardiotoxicity
Pharmacokinetics of anticancer agents in patients with impaired liver function, and exacerbates changes in iron metabolism induced by doxorubicin, Blood 102
Eur. J. Cancer 34 (1) (1998) 33–46. (7) (2003) 2574–2580.
[31] T.H. Connor, G. Burroughs, M. McDiarmid, K.R. Mead, L. Power, L. Reed, NIOSH [59] J.C. Canzoneri, A.K. Oyelere, Interaction of anthracyclines with iron responsive
Alert: Preventing Occupational Exposures to Antineoplastic and Other Hazardous element mRNAs, Nucleic Acids Res. 36 (21) (2008) 6825–6834.
Drugs in Health Care Settings, DHHS (NIOSH) Publication, Atlanta, 2004, [60] J.C. Barton, L.F. Bertoli, Transfusion iron overload in adults with acute leukemia:
pp. 1–50. manifestations and therapy, Am. J. Med. Sci. 319 (2) (2000) 73–78.
[32] Y. Octavia, C.G. Tocchetti, K.L. Gabrielson, S. Janssens, H.J. Crijns, A.L. Moens, [61] Y. Ichikawa, M. Ghanefar, M. Bayeva, R. Wu, A. Khechaduri, S.V. Naga Prasad, R.
Doxorubicin-induced cardiomyopathy: from molecular mechanisms to K. Mutharasan, T.J. Naik, H. Ardehali, Cardiotoxicity of doxorubicin is mediated
therapeutic strategies, J. Mol. Cell. Cardiol. 52 (6) (2012) 1213–1225. through mitochondrial iron accumulation, J. Clin. Investig. 124 (2) (2014)
[33] M.S. Horenstein, R.S. Vander Heide, T.J. L’Ecuyer, Molecular basis of 617–630.
anthracycline-induced cardiotoxicity and its prevention, Mol. Genet. Metab. 71 [62] Y. Zhao, D. McLaughlin, E. Robinson, A.P. Harvey, M.B. Hookham, A.M. Shah, B.
(1–2) (2000) 436–444. J. McDermott, D.J. Grieve, Nox2 NADPH oxidase promotes pathologic cardiac
[34] M.F. Xu, P.L. Tang, Z.M. Qian, M. Ashraf, Effects by doxorubicin on the remodeling associated with Doxorubicin chemotherapy, Cancer Res. 70 (22)
myocardium are mediated by oxygen free radicals, Life Sci. 68 (8) (2001) (2010) 9287–9297.
889–901. [63] M.T. Elnakish, H.H. Hassanain, P.M. Janssen, M.G. Angelos, M. Khan, Emerging
[35] T. Simůnek, M. Stérba, O. Popelová, M. Adamcová, R. Hrdina, V. Gersl, role of oxidative stress in metabolic syndrome and cardiovascular diseases:
Anthracycline-induced cardiotoxicity: overview of studies examining the roles of important role of Rac/NADPH oxidase, J. Pathol. 231 (3) (2013) 290–300.
oxidative stress and free cellular iron, Pharmacol. Rep. 61 (1) (2009) 154–171. [64] L. Wojnowski, B. Kulle, M. Schirmer, G. Schlüter, A. Schmidt, A. Rosenberger,
[36] J. Vásquez-Vivar, P. Martasek, N. Hogg, B.S. Masters, K.A. Pritchard Jr., S. Vonhof, H. Bickeböller, M.R. Toliat, E.K. Suk, M. Tzvetkov, A. Kruger,
B. Kalyanaraman, Endothelial nitric oxide synthase-dependent superoxide S. Seifert, M. Kloess, H. Hahn, M. Loeffler, P. Nürnberg, M. Pfreundschuh,
generation from adriamycin, Biochemistry 36 (38) (1997) 11293–11297. L. Trümper, J. Brockmöller, G. Hasenfuss, NAD(P)H oxidase and multidrug
[37] O.W. Griffith, D.J. Stuehr, Nitric oxide synthases: properties and catalytic resistance protein genetic polymorphisms are associated with doxorubicin-
mechanism, Annu. Rev. Physiol. 57 (1995) 707–736. induced cardiotoxicity, Circulation 112 (24) (2005) 3754–3762.
[38] G.R. Drummond, H. Cai, M.E. Davis, S. Ramasamy, D.G. Harrison, Transcriptional [65] S. Deng, A. Kruger, A.L. Kleschyov, L. Kalinowski, A. Daiber, L. Wojnowski,
and posttranscriptional regulation of endothelial nitric oxide synthase expression Gp91phox-containing NAD(P)H oxidase increases superoxide formation by
by hydrogen peroxide, Circ. Res. 86 (3) (2000) 347–354. doxorubicin and NADPH, Free Radic. Biol. Med. 42 (4) (2007) 466–473.
[39] S.V. Kalivendi, S. Kotamraju, H. Zhao, J. Joseph, B. Kalyanaraman, Doxorubicin- [66] D. McLaughlin, Y. Zhao, K.M. O’Neill, K.S. Edgar, P.D. Dunne, A.M. Kearney, D.
induced apoptosis is associated with increased transcription of endothelial nitric- J. Grieve, B.J. McDermott, Signalling mechanisms underlying doxorubicin and
oxide synthase. Effect of antiapoptotic antioxidants and calcium, J. Biol. Chem. Nox2 NADPH oxidase-induced cardiomyopathy: involvement of mitofusin-2, Br.
276 (50) (2001) 47266–47276. J. Pharmacol. 174 (21) (2017) 3677–3695.
[40] P. Mukhopadhyay, M. Rajesh, S. Bátkai, Y. Kashiwaya, G. Haskó, L. Liaudet, [67] J. Ma, Y. Wang, D. Zheng, M. Wei, H. Xu, T. Peng, Rac1 signalling mediates
C. Szabó, P. Pacher, Role of superoxide, nitric oxide, and peroxynitrite in doxorubicin-induced cardiotoxicity through both reactive oxygen species-
doxorubicin-induced cell death in vivo and in vitro, Am. J. Physiol. Heart Circ. dependent and -independent pathways, Cardiovasc. Res. 97 (1) (2013) 77–87.
Physiol. 296 (5) (2009) H1466–H1483. [68] M. Gilleron, X. Marechal, D. Montaigne, J. Franczak, R. Neviere, S. Lancel,
[41] G. Akolkar, A.K. Bagchi, P. Ayyappan, D.S. Jassal, P.K. Singal, Doxorubicin- NADPH oxidases participate to doxorubicin-induced cardiac myocyte apoptosis,
induced nitrosative stress is mitigated by vitamin C via the modulation of nitric Biochem. Biophys. Res. Commun. 388 (4) (2009) 727–731.
oxide synthases, Am. J. Physiol. Cell Physiol. 312 (4) (2017). C418-c427. [69] S.V. Kalivendi, E.A. Konorev, S. Cunningham, S.K. Vanamala, E.H. Kaji, J. Joseph,
[42] B. Liu, H. Li, H. Qu, B. Sun, Nitric oxide synthase expressions in ADR-induced B. Kalyanaraman, Doxorubicin activates nuclear factor of activated T-
cardiomyopathy in rats, J. Biochem Mol. Biol. 39 (6) (2006) 759–765. lymphocytes and Fas ligand transcription: role of mitochondrial reactive oxygen
[43] Y. Chen, T. Huang, W. Shi, J. Fang, H. Deng, G. Cui, Potential targets for species and calcium, Biochem. J. 389 (Pt 2) (2005) 527–539.
intervention against doxorubicin-induced cardiotoxicity based on genetic studies: [70] L.X. Fu, F. Waagstein, A. Hjalmarson, A new insight into adriamycin-induced
a systematic review of the literature, J. Mol. Cell. Cardiol. 138 (2020) 88–98. cardiotoxicity, Int. J. Cardiol. 29 (1) (1990) 15–20.
[44] J. Fu, K. Yamamoto, Z.W. Guan, S. Kimura, T. Iyanagi, Human neuronal nitric [71] D.S. dos Santos, R.C. dos Santos Goldenberg, Doxorubicin-Induced Cardiotoxicity:
oxide synthase can catalyze one-electron reduction of adriamycin: role of flavin From Mechanisms to Development of Efficient Therapy, Cardiotoxicity,
domain, Arch. Biochem. Biophys. 427 (2) (2004) 180–187. IntechOpen, 2018.
[45] A. Ghigo, M. Li, E. Hirsch, New signal transduction paradigms in anthracycline- [72] E.C. Keung, L. Toll, M. Ellis, R.A. Jensen, L-type cardiac calcium channels in
induced cardiotoxicity, Biochim. Biophys. Acta 1863 (7 Pt B) (2016) 1916–1925. doxorubicin cardiomyopathy in rats morphological, biochemical, and functional
[46] E. Goormaghtigh, P. Chatelain, J. Caspers, J.M. Ruysschaert, Evidence of a correlations, J. Clin. Investig. 87 (6) (1991) 2108–2113.
specific complex between adriamycin and negatively-charged phospholipids, [73] P. Menna, E. Salvatorelli, L. Gianni, G. Minotti, Anthracycline cardiotoxicity, Top.
Biochim. Biophys. Acta 597 (1) (1980) 1–14. Curr. Chem. 283 (2008) 21–44.
[47] M. Schlame, D. Rua, M.L. Greenberg, The biosynthesis and functional role of [74] Y.M. Jang, S. Kendaiah, B. Drew, T. Phillips, C. Selman, D. Julian,
cardiolipin, Prog. Lipid Res. 39 (3) (2000) 257–288. C. Leeuwenburgh, Doxorubicin treatment in vivo activates caspase-12 mediated
[48] K. Kashfi, M. Israel, T.W. Sweatman, R. Seshadri, G.A. Cook, Inhibition of cardiac apoptosis in both male and female rats, FEBS Lett. 577 (3) (2004)
mitochondrial carnitine palmitoyltransferases by adriamycin and adriamycin 483–490.
analogues, Biochem. Pharmacol. 40 (7) (1990) 1441–1448. [75] C.C. Lim, C. Zuppinger, X. Guo, G.M. Kuster, M. Helmes, H.M. Eppenberger, T.
[49] R.A. Carvalho, R.P. Sousa, V.J. Cadete, G.D. Lopaschuk, C.M. Palmeira, J. M. Suter, R. Liao, D.B. Sawyer, Anthracyclines induce calpain-dependent titin
A. Bjork, K.B. Wallace, Metabolic remodeling associated with subchronic proteolysis and necrosis in cardiomyocytes, J. Biol. Chem. 279 (9) (2004)
doxorubicin cardiomyopathy, Toxicology 270 (2–3) (2010) 92–98. 8290–8299.
[50] R. Ventura-Clapier, A. Garnier, V. Veksler, Energy metabolism in heart failure, [76] S. Zhou, A. Starkov, M.K. Froberg, R.L. Leino, K.B. Wallace, Cumulative and
J. Physiol. 555 (Pt 1) (2004) 1–13. irreversible cardiac mitochondrial dysfunction induced by doxorubicin, Cancer
[51] S. Raj, V.I. Franco, S.E. Lipshultz, Anthracycline-induced cardiotoxicity: a review Res. 61 (2) (2001) 771–777.
of pathophysiology, diagnosis, and treatment, Curr. Treat. Options Cardiovasc. [77] M.A. Mitry, J.G. Edwards, Doxorubicin induced heart failure: phenotype and
Med. 16 (6) (2014) 315. molecular mechanisms, Int. J. Cardiol. Heart Vasc. 10 (2016) 17–24.
[52] H.B. Suliman, M.S. Carraway, A.S. Ali, C.M. Reynolds, K.E. Welty-Wolf, C. [78] H. Tscheschner, E. Meinhardt, P. Schlegel, A. Jungmann, L.H. Lehmann, O.
A. Piantadosi, The CO/HO system reverses inhibition of mitochondrial biogenesis J. Müller, P. Most, H.A. Katus, P.W. Raake, CaMKII activation participates in
and prevents murine doxorubicin cardiomyopathy, J. Clin. Investig. 117 (12) doxorubicin cardiotoxicity and is attenuated by moderate GRP78 overexpression,
(2007) 3730–3741. PLoS One 14 (4) (2019), e0215992.

11
P.S. Rawat et al. Biomedicine & Pharmacotherapy 139 (2021) 111708

[79] K.N. Timm, D.J. Tyler, The role of AMPK activation for cardioprotection in [103] S. Bien, A. Riad, C.A. Ritter, M. Gratz, F. Olshausen, D. Westermann, M. Grube,
doxorubicin-induced cardiotoxicity, Cardiovasc. Drugs Ther. 34 (2) (2020) T. Krieg, S. Ciecholewski, S.B. Felix, A. Staudt, H.P. Schultheiss, R. Ewert,
255–269. U. Völker, C. Tschöpe, H.K. Kroemer, The endothelin receptor blocker bosentan
[80] Z. Li, Y. Song, L. Liu, N. Hou, X. An, D. Zhan, Y. Li, L. Zhou, P. Li, L. Yu, J. Xia, inhibits doxorubicin-induced cardiomyopathy, Cancer Res. 67 (21) (2007)
Y. Zhang, J. Wang, X. Yang, miR-199a impairs autophagy and induces cardiac 10428–10435.
hypertrophy through mTOR activation, Cell Death Differ. 24 (7) (2017) [104] L.L. Yang, R. Gros, M.G. Kabir, A. Sadi, A.I. Gotlieb, M. Husain, D.J. Stewart,
1205–1213. Conditional cardiac overexpression of endothelin-1 induces inflammation and
[81] I. Lekli, D.D. Haines, G. Balla, A. Tosaki, Autophagy: an adaptive physiological dilated cardiomyopathy in mice, Circulation 109 (2) (2004) 255–261.
countermeasure to cellular senescence and ischaemia/reperfusion-associated [105] A. Goetzenich, N. Hatam, A. Zernecke, C. Weber, T. Czarnotta, R. Autschbach,
cardiac arrhythmias, J. Cell. Mol. Med. 21 (6) (2017) 1058–1072. S. Christiansen, Alteration of matrix metalloproteinases in selective left
[82] S. Kobayashi, P. Volden, D. Timm, K. Mao, X. Xu, Q. Liang, Transcription factor ventricular adriamycin-induced cardiomyopathy in the pig, J. Heart Lung
GATA4 inhibits doxorubicin-induced autophagy and cardiomyocyte death, Transpl. 28 (10) (2009) 1087–1093.
J. Biol. Chem. 285 (1) (2010) 793–804. [106] P. Spallarossa, P. Altieri, S. Garibaldi, G. Ghigliotti, C. Barisione, V. Manca,
[83] J.M. Velez, S. Miriyala, R. Nithipongvanitch, T. Noel, C.D. Plabplueng, P. Fabbi, A. Ballestrero, C. Brunelli, A. Barsotti, Matrix metalloproteinase-2 and -9
T. Oberley, P. Jungsuwadee, H. Van Remmen, M. Vore, D.K. Clair St, p53 are induced differently by doxorubicin in H9c2 cells: The role of MAP kinases and
Regulates oxidative stress-mediated retrograde signaling: a novel mechanism for NAD(P)H oxidase, Cardiovasc. Res. 69 (3) (2006) 736–745.
chemotherapy-induced cardiac injury, PLoS One 6 (3) (2011), e18005. [107] C. Dupont, D.R. Armant, C.A. Brenner, Epigenetics: definition, mechanisms and
[84] J.J. Bartlett, P.C. Trivedi, P. Yeung, P.C. Kienesberger, T. Pulinilkunnil, clinical perspective, Seminars in reproductive medicine, © Thieme Medical
Doxorubicin impairs cardiomyocyte viability by suppressing transcription factor Publishers, (2009), pp. 351–357.
EB expression and disrupting autophagy, Biochem. J. 473 (21) (2016) [108] M.M. Suzuki, A. Bird, DNA methylation landscapes: provocative insights from
3769–3789. epigenomics, Nat. Rev. Genet. 9 (6) (2008) 465–476.
[85] W. Zhu, M.H. Soonpaa, H. Chen, W. Shen, R.M. Payne, E.A. Liechty, R. [109] M. Inbar-Feigenberg, S. Choufani, D.T. Butcher, M. Roifman, R. Weksberg, Basic
L. Caldwell, W. Shou, L.J. Field, Acute doxorubicin cardiotoxicity is associated concepts of epigenetics, Fertil. Steril. 99 (3) (2013) 607–615.
with p53-induced inhibition of the mammalian target of rapamycin pathway, [110] R. Lister, M. Pelizzola, Y.S. Kida, R.D. Hawkins, J.R. Nery, G. Hon, J. Antosiewicz-
Circulation 119 (1) (2009) 99–106. Bourget, R. O’Malley, R. Castanon, S. Klugman, Hotspots of aberrant epigenomic
[86] M.B. Chen, X.Y. Wu, J.H. Gu, Q.T. Guo, W.X. Shen, P.H. Lu, Activation of AMP- reprogramming in human induced pluripotent stem cells, Nature 471 (7336)
activated protein kinase contributes to doxorubicin-induced cell death and (2011) 68–73.
apoptosis in cultured myocardial H9c2 cells, Cell Biochem. Biophys. 60 (3) [111] M. Nakao, Epigenetics: interaction of DNA methylation and chromatin, Gene 278
(2011) 311–322. (1–2) (2001) 25–31.
[87] N. Koleini, E. Kardami, Autophagy and mitophagy in the context of doxorubicin- [112] R.M. Youngson, Collins dictionary of human biology, Collins (2006).
induced cardiotoxicity, Oncotarget 8 (28) (2017) 46663–46680. [113] L. Pogliani, A. Haghi, N. Islam. Research Methodologies and Practical
[88] M. Katamura, E. Iwai-Kanai, M. Nakaoka, Y. Okawa, M. Ariyoshi, Y. Mita, Applications of Chemistry, Apple Academic Press, 2019.
A. Nakamura, K. Ikeda, T. Ogata, T. Ueyama, Curcumin attenuates doxorubicin- [114] E.P. Consortium, The ENCODE (ENCyclopedia of DNA elements) project, Science
induced cardiotoxicity by inducing autophagy via the regulation of JNK 306 (5696) (2004) 636–640.
phosphorylation, J. Clin. Exp. Cardiol. 5 (09) (2014). [115] I. Dunham, E. Birney, B.R. Lajoie, A. Sanyal, X. Dong, M. Greven, X. Lin, J. Wang,
[89] B. Liu, Q.X. Bai, X.Q. Chen, G.X. Gao, H.T. Gu, [Effect of curcumin on expression T.W. Whitfield, J. Zhuang, An integrated encyclopedia of DNA elements in the
of survivin, Bcl-2 and Bax in human multiple myeloma cell line], Zhongguo Shi human genome, (2012).
Yan Xue Ye Xue Za Zhi 15 (4) (2007) 762–766. [116] C.E. Clare, A.H. Brassington, W.Y. Kwong, K.D. Sinclair, One-carbon metabolism:
[90] K. Vedam, Y. Nishijima, L.J. Druhan, M. Khan, N.I. Moldovan, J.L. Zweier, linking nutritional biochemistry to epigenetic programming of long-term
G. Ilangovan, Role of heat shock factor-1 activation in the doxorubicin-induced development, Annu. Rev. Anim. Biosci. 7 (2019) 263–287.
heart failure in mice, Am. J. Physiol. Heart Circ. Physiol. 298 (6) (2010) [117] A. Ferreira, T. Cunha-Oliveira, R.F. Simões, F.S. Carvalho, A. Burgeiro,
H1832–H1841. K. Nordgren, K.B. Wallace, P.J. Oliveira, Altered mitochondrial epigenetics
[91] D.S. Kim, E.R. Woo, S.W. Chae, K.C. Ha, G.H. Lee, S.T. Hong, D.Y. Kwon, M. associated with subchronic doxorubicin cardiotoxicity, Toxicology 390 (2017)
S. Kim, Y.K. Jung, H.M. Kim, H.K. Kim, H.R. Kim, H.J. Chae, Plantainoside D 63–73.
protects adriamycin-induced apoptosis in H9c2 cardiac muscle cells via the [118] L.S. Shock, P.V. Thakkar, E.J. Peterson, R.G. Moran, S.M. Taylor, DNA
inhibition of ROS generation and NF-kappaB activation, Life Sci. 80 (4) (2007) methyltransferase 1, cytosine methylation, and cytosine hydroxymethylation in
314–323. mammalian mitochondria, Proc. Natl. Acad. Sci. USA 108 (9) (2011) 3630–3635.
[92] H. Li, H. Gu, B. Sun, Protective effects of pyrrolidine dithiocarbamate on [119] H. Kumari, W.H. Huang, M.W.Y. Chan, Review on the role of epigenetic
myocardium apoptosis induced by adriamycin in rats, Int. J. Cardiol. 114 (2) modifications in doxorubicin-induced cardiotoxicity, Front. Cardiovasc. Med. 7
(2007) 159–165. (2020) 56.
[93] S. Wang, S. Kotamraju, E. Konorev, S. Kalivendi, J. Joseph, B. Kalyanaraman, [120] R. Song, Y. Yang, H. Lei, G. Wang, Y. Huang, W. Xue, Y. Wang, L. Yao, Y. Zhu,
Activation of nuclear factor-kappaB during doxorubicin-induced apoptosis in HDAC6 inhibition protects cardiomyocytes against doxorubicin-induced acute
endothelial cells and myocytes is pro-apoptotic: the role of hydrogen peroxide, damage by improving α-tubulin acetylation, J. Mol. Cell. Cardiol. 124 (2018)
Biochem. J. 367 (Pt 3) (2002) 729–740. 58–69.
[94] P. Liu, H.Y. Bao, C.C. Jin, J.C. Zhou, F. Hua, K. Li, X.X. Lv, B. Cui, Z.W. Hu, X. [121] B. Pang, X. Qiao, L. Janssen, A. Velds, T. Groothuis, R. Kerkhoven, M. Nieuwland,
W. Zhang, Targeting extracellular heat shock protein 70 ameliorates doxorubicin- H. Ovaa, S. Rottenberg, O. van Tellingen, J. Janssen, P. Huijgens, W. Zwart,
induced heart failure through resolution of toll-like receptor 2-mediated J. Neefjes, Drug-induced histone eviction from open chromatin contributes to the
myocardial inflammation, J. Am. Heart Assoc. 8 (20) (2019), e012338. chemotherapeutic effects of doxorubicin, Nat. Commun. 4 (2013) 1908.
[95] Y.X. Shan, T.J. Liu, H.F. Su, A. Samsamshariat, R. Mestril, P.H. Wang, Hsp10 and [122] M.U. Kaikkonen, M.T. Lam, C.K. Glass, Non-coding RNAs as regulators of gene
Hsp60 modulate Bcl-2 family and mitochondria apoptosis signaling induced by expression and epigenetics, Cardiovasc. Res. 90 (3) (2011) 430–440.
doxorubicin in cardiac muscle cells, J. Mol. Cell. Cardiol. 35 (9) (2003) [123] C. Ruggeri, S. Gioffré, F. Achilli, G.I. Colombo, Y. D’Alessandra, Role of
1135–1143. microRNAs in doxorubicin-induced cardiotoxicity: an overview of preclinical
[96] Y. Lan, Y. Wang, K. Huang, Q. Zeng, Heat shock protein 22 attenuates models and cancer patients, Heart Fail. Rev. 23 (1) (2018) 109–122.
doxorubicin-induced cardiotoxicity via regulating inflammation and apoptosis, [124] C. Vacchi-Suzzi, Y. Bauer, B.R. Berridge, S. Bongiovanni, K. Gerrish, H.
Front. Pharmacol. 11 (2020) 257. K. Hamadeh, M. Letzkus, J. Lyon, J. Moggs, R.S. Paules, F. Pognan, F. Staedtler,
[97] G.C. Fan, X. Zhou, X. Wang, G. Song, J. Qian, P. Nicolaou, G. Chen, X. Ren, E. M.P. Vidgeon-Hart, O. Grenet, P. Couttet, Perturbation of microRNAs in rat heart
G. Kranias, Heat shock protein 20 interacting with phosphorylated Akt reduces during chronic doxorubicin treatment, PLoS One 7 (7) (2012), e40395.
doxorubicin-triggered oxidative stress and cardiotoxicity, Circ. Res. 103 (11) [125] Z. Tong, B. Jiang, Y. Wu, Y. Liu, Y. Li, M. Gao, Y. Jiang, Q. Lv, X. Xiao, MiR-21
(2008) 1270–1279. protected cardiomyocytes against doxorubicin-induced apoptosis by targeting
[98] M. Ueno, Y. Kakinuma, K. Yuhki, N. Murakoshi, M. Iemitsu, T. Miyauchi, BTG2, Int. J. Mol. Sci. 16 (7) (2015) 14511–14525.
I. Yamaguchi, Doxorubicin induces apoptosis by activation of caspase-3 in [126] H. Tony, K. Yu, Z. Qiutang, MicroRNA-208a silencing attenuates doxorubicin
cultured cardiomyocytes in vitro and rat cardiac ventricles in vivo, J. Pharmacol. induced myocyte apoptosis and cardiac dysfunction, Oxid. Med. Cell. Longev.
Sci. 101 (2) (2006) 151–158. 2015 (2015), 597032.
[99] L. Zhao, B. Zhang, Doxorubicin induces cardiotoxicity through upregulation of [127] T. Horie, K. Ono, H. Nishi, K. Nagao, M. Kinoshita, S. Watanabe, Y. Kuwabara,
death receptors mediated apoptosis in cardiomyocytes, Sci. Rep. 7 (2017) 44735. Y. Nakashima, R. Takanabe-Mori, E. Nishi, K. Hasegawa, T. Kita, T. Kimura, Acute
[100] S.C. Kim, J.P. Stice, L. Chen, J.S. Jung, S. Gupta, Y. Wang, G. Baumgarten, J. Trial, doxorubicin cardiotoxicity is associated with miR-146a-induced inhibition of the
A.A. Knowlton, Extracellular heat shock protein 60, cardiac myocytes, and neuregulin-ErbB pathway, Cardiovasc. Res. 87 (4) (2010) 656–664.
apoptosis, Circ. Res. 105 (12) (2009) 1186–1195. [128] L. Roca-Alonso, L. Castellano, A. Mills, A.F. Dabrowska, M.B. Sikkel, L. Pellegrino,
[101] K. Rayner, Y.X. Chen, M. McNulty, T. Simard, X. Zhao, D.J. Wells, J. de J. Jacob, A.E. Frampton, J. Krell, R.C. Coombes, S.E. Harding, A.R. Lyon,
Belleroche, E.R. O’Brien, Extracellular release of the atheroprotective heat shock J. Stebbing, Myocardial MiR-30 downregulation triggered by doxorubicin drives
protein 27 is mediated by estrogen and competitively inhibits acLDL binding to alterations in β-adrenergic signaling and enhances apoptosis, Cell Death Dis. 6 (5)
scavenger receptor-A, Circ. Res. 103 (2) (2008) 133–141. (2015), e1754.
[102] A.O. Aliprantis, R.B. Yang, D.S. Weiss, P. Godowski, A. Zychlinsky, The apoptotic [129] H. Ren, Y. Shao, C. Wu, X. Ma, C. Lv, Q. Wang, Metformin alleviates oxidative
signaling pathway activated by Toll-like receptor-2, EMBO J. 19 (13) (2000) stress and enhances autophagy in diabetic kidney disease via AMPK/SIRT1-FoxO1
3325–3336. pathway, Mol. Cell. Endocrinol. 500 (2020), 110628.

12
P.S. Rawat et al. Biomedicine & Pharmacotherapy 139 (2021) 111708

[130] L.C. Kobashigawa, Y.C. Xu, J.F. Padbury, Y.T. Tseng, N. Yano, Metformin protects [157] A. Ramond, E. Sartorius, M. Mousseau, C. Ribuot, M. Joyeux-Faure,
cardiomyocyte from doxorubicin induced cytotoxicity through an AMP-activated Erythropoietin pretreatment protects against acute chemotherapy toxicity in
protein kinase dependent signaling pathway: an in vitro study, PLoS One 9 (8) isolated rat hearts, Exp. Biol. Med. 233 (1) (2008) 76–83.
(2014), e104888. [158] A. Engert, A. Josting, H. Haverkamp, M. Villalobos, A. Lohri, M. Sökler, J. Zijlstra,
[131] F.J. Alenghat, A.M. Davis, Management of blood cholesterol, JAMA 321 (8) I. Sturm, M.S. Topp, A. Rank, T. Zenz, M. Vogelhuber, L. Nogova, P. Borchmann,
(2019) 800–801. M. Fuchs, H.H. Flechtner, V. Diehl, Epoetin alfa in patients with advanced-stage
[132] W. Sun, T.S. Lee, M. Zhu, C. Gu, Y. Wang, Y. Zhu, J.Y. Shyy, Statins activate AMP- Hodgkin’s lymphoma: results of the randomized placebo-controlled GHSG
activated protein kinase in vitro and in vivo, Circulation 114 (24) (2006) HD15EPO trial, J. Clin. Oncol. 28 (13) (2010) 2239–2245.
2655–2662. [159] X. Li, J. Zhong, Z. Zeng, H. Wang, J. Li, X. Liu, X. Yang, MiR-181c protects
[133] H.C. Choi, P. Song, Z. Xie, Y. Wu, J. Xu, M. Zhang, Y. Dong, S. Wang, K. Lau, M. cardiomyocyte injury by preventing cell apoptosis through PI3K/Akt signaling
H. Zou, Reactive nitrogen species is required for the activation of the AMP- pathway, Cardiovasc. Diagn. Ther. 10 (4) (2020) 849–858.
activated protein kinase by statin in vivo, J. Biol. Chem. 283 (29) (2008) [160] X. Li, S. Chu, M. Lin, Y. Gao, Y. Liu, S. Yang, X. Zhou, Y. Zhang, Y. Hu, H. Wang,
20186–20197. N. Chen, Anticancer property of ginsenoside Rh2 from ginseng, Eur. J. Med.
[134] R. Kou, J. Sartoretto, T. Michel, Regulation of Rac1 by simvastatin in endothelial Chem. 203 (2020), 112627.
cells: differential roles of AMP-activated protein kinase and calmodulin- [161] M. Rajadurai, P. Stanely Mainzen Prince, Preventive effect of naringin on cardiac
dependent kinase kinase-beta, J. Biol. Chem. 284 (22) (2009) 14734–14743. markers, electrocardiographic patterns and lysosomal hydrolases in normal and
[135] J.B. Blanter, W.H. Frishman, The preventive role of angiotensin converting isoproterenol-induced myocardial infarction in Wistar rats, Toxicology 230 (2–3)
enzyme inhibitors/angiotensin-II receptor blockers and β-adrenergic blockers in (2007) 178–188.
anthracycline- and trastuzumab-induced cardiotoxicity, Cardiol. Rev. 27 (5) [162] H. Wang, P. Yu, H. Gou, J. Zhang, M. Zhu, Z.H. Wang, J.W. Tian, Y.T. Jiang, F.
(2019) 256–259. H. Fu, Cardioprotective effects of 20(S)-Ginsenoside Rh2 against doxorubicin-
[136] N. Kalay, E. Basar, I. Ozdogru, O. Er, Y. Cetinkaya, A. Dogan, T. Inanc, induced cardiotoxicity in vitro and in vivo, Evid. Based Complement. Altern. Med.
A. Oguzhan, N.K. Eryol, R. Topsakal, A. Ergin, Protective effects of carvedilol 2012 (2012), 506214.
against anthracycline-induced cardiomyopathy, J. Am. Coll. Cardiol. 48 (11) [163] A. Shehzad, R. Shahzad, Y.S. Lee, Curcumin: a potent modulator of multiple
(2006) 2258–2262. enzymes in multiple cancers, Enzymes 36 (2014) 149–174.
[137] P.W. Fisher, F. Salloum, A. Das, H. Hyder, R.C. Kukreja, Phosphodiesterase-5 [164] M.L. Lestari, G. Indrayanto, Curcumin, Profiles Drug Subst. Excip. Relat.
inhibition with sildenafil attenuates cardiomyocyte apoptosis and left ventricular Methodol. 39 (2014) 113–204.
dysfunction in a chronic model of doxorubicin cardiotoxicity, Circulation 111 [165] Z. Tavakoli Dargani, D.K. Singla, Embryonic stem cell-derived exosomes inhibit
(13) (2005) 1601–1610. doxorubicin-induced TLR4-NLRP3-mediated cell death-pyroptosis, Am. J.
[138] A. Das, D. Durrant, C. Mitchell, E. Mayton, N.N. Hoke, F.N. Salloum, M.A. Park, Physiol. Heart Circ. Physiol. 317 (2) (2019). H460-h471.
I. Qureshi, R. Lee, P. Dent, R.C. Kukreja, Sildenafil increases chemotherapeutic [166] S.M. Man, R. Karki, B. Briard, A. Burton, S. Gingras, S. Pelletier, T.D. Kanneganti,
efficacy of doxorubicin in prostate cancer and ameliorates cardiac dysfunction, Differential roles of caspase-1 and caspase-11 in infection and inflammation, Sci.
Proc. Natl. Acad. Sci. USA 107 (42) (2010) 18202–18207. Rep. 7 (2017) 45126.
[139] A. Aries, P. Paradis, C. Lefebvre, R.J. Schwartz, M. Nemer, Essential role of GATA- [167] W. Yu, X. Qin, Y. Zhang, P. Qiu, L. Wang, W. Zha, J. Ren, Curcumin suppresses
4 in cell survival and drug-induced cardiotoxicity, Proc. Natl. Acad. Sci. USA 101 doxorubicin-induced cardiomyocyte pyroptosis via a PI3K/Akt/mTOR-dependent
(18) (2004) 6975–6980. manner, Cardiovasc. Diagn. Ther. 10 (4) (2020) 752–769.
[140] J. Beak, W. Huang, J.S. Parker, S.T. Hicks, C. Patterson, P.C. Simpson, A. Ma, [168] K.G. Haug, B. Weber, G. Hochhaus, V. Butterweck, Nonlinear pharmacokinetics of
J. Jin, B.C. Jensen, An oral selective alpha-1A adrenergic receptor agonist visnagin in rats after intravenous bolus administration, Eur. J. Pharm. Sci. 45
prevents doxorubicin cardiotoxicity, JACC Basic Transl. Sci. 2 (1) (2017) 39–53. (1–2) (2012) 79–89.
[141] J.T. Mack, Dexrazoxane. xPharm: The Comprehensive Pharmacology Reference, [169] Y. Liu, A. Asnani, L. Zou, V.L. Bentley, M. Yu, Y. Wang, G. Dellaire, K.S. Sarkar,
Elsevier, 2007, pp. 1–7. M. Dai, H.H. Chen, D.E. Sosnovik, J.T. Shin, D.A. Haber, J.N. Berman, W. Chao, R.
[142] P.E. Schroeder, B.B. Hasinoff, Metabolism of the one-ring open metabolites of the T. Peterson, Visnagin protects against doxorubicin-induced cardiomyopathy
cardioprotective drug dexrazoxane to its active metal-chelating form in the rat, through modulation of mitochondrial malate dehydrogenase, Sci. Transl. Med. 6
Drug Metab. Dispos. 33 (9) (2005) 1367–1372. (266) (2014), 266ra170.
[143] M. Jasiński, L. Jasińska, M. Ogrodowczyk, Resveratrol in prostate diseases - a [170] C. Gietl, Malate dehydrogenase isoenzymes: cellular locations and role in the flow
short review, Cent. Eur. J. Urol. 66 (2) (2013) 144–149. of metabolites between the cytoplasm and cell organelles, Biochim. Biophys. Acta
[144] E. Tatlidede, O. Sehirli, A. Velioğlu-Oğünc, S. Cetinel, B.C. Yeğen, A. Yarat, 1100 (3) (1992) 217–234.
S. Süleymanoğlu, G. Sener, Resveratrol treatment protects against doxorubicin- [171] T.T. Nielsen, N.B. Støttrup, B. Løfgren, H.E. Bøtker, Metabolic fingerprint of
induced cardiotoxicity by alleviating oxidative damage, Free Radic. Res. 43 (3) ischaemic cardioprotection: importance of the malate-aspartate shuttle,
(2009) 195–205. Cardiovasc. Res. 91 (3) (2011) 382–391.
[145] D. Liu, Z. Ma, L. Xu, X. Zhang, S. Qiao, J. Yuan, PGC1α activation by pterostilbene [172] P.K. Leong, J. Chen, K.M. Ko, Development of Chinese herbal health products for
ameliorates acute doxorubicin cardiotoxicity by reducing oxidative stress via the prevention of aging-associated diseases. Natural Products and Drug Discovery,
enhancing AMPK and SIRT1 cascades, Aging 11 (22) (2019) 10061–10073. Elsevier, 2018, pp. 73–104.
[146] B.P. Marriott, D.F. Birt, V.A. Stalling, A.A. Yates. Present Knowledge in Nutrition: [173] X. Fan, K. Elkin, Y. Shi, Z. Zhang, Y. Cheng, J. Gu, J. Liang, C. Wang, X. Ji,
Basic Nutrition and Metabolism, Academic Press, 2020. Schisandrin B improves cerebral ischemia and reduces reperfusion injury in rats
[147] J.B. Blumberg, B.B. Frei, V.L. Fulgoni, C.M. Weaver, S.H. Zeisel, Impact of through TLR4/NF-κB signaling pathway inhibition, Neurol. Res. 42 (8) (2020)
frequency of multi-vitamin/multi-mineral supplement intake on nutritional 693–702.
adequacy and nutrient deficiencies in U.S. adults, Nutrients 9 (8) (2017). [174] L. Li, Q. Pan, W. Han, Z. Liu, L. Li, X. Hu, Schisandrin B prevents doxorubicin-
[148] E. El-Demerdash, A.A. Ali, M.M. Sayed-Ahmed, A.M. Osman, New aspects in induced cardiotoxicity via enhancing glutathione redox cycling, Clin. Cancer Res.
probucol cardioprotection against doxorubicin-induced cardiotoxicity, Cancer 13 (22 Pt 1) (2007) 6753–6760.
Chemother. Pharmacol. 52 (5) (2003) 411–416. [175] W. Zhu, M.H. Soonpaa, H. Chen, W. Shen, R.M. Payne, E.A. Liechty, R.
[149] N. Siveski-Iliskovic, M. Hill, D.A. Chow, P.K. Singal, Probucol protects against L. Caldwell, W. Shou, L.J. Field, Acute doxorubicin cardiotoxicity is associated
adriamycin cardiomyopathy without interfering with its antitumor effect, with p53-induced inhibition of the mTOR pathway, Circulation 119 (1) (2009)
Circulation 91 (1) (1995) 10–15. 99.
[150] N. Siveski-Iliskovic, N. Kaul, P.K. Singal, Probucol promotes endogenous [176] Y. Xu, Z. Liu, J. Sun, Q. Pan, F. Sun, Z. Yan, X. Hu, Schisandrin B prevents
antioxidants and provides protection against adriamycin-induced doxorubicin-induced chronic cardiotoxicity and enhances its anticancer activity
cardiomyopathy in rats, Circulation 89 (6) (1994) 2829–2835. in vivo, PLoS One 6 (12) (2011), e28335.
[151] B.A. Reyes, E.C. Dufourt, J. Ross, M.J. Warner, N.C. Tanquilut, A.B. Leung, [177] M. Gharanei, A. Hussain, O. Janneh, H. Maddock, Attenuation of doxorubicin-
Selected Phyto and Marine Bioactive Compounds: Alternatives for the Treatment induced cardiotoxicity by mdivi-1: a mitochondrial division/mitophagy inhibitor,
of Type 2 Diabetes, Studies in Natural Products Chemistry, Elsevier, 2018, PLoS One 8 (10) (2013), e77713.
pp. 111–143. [178] Y. Xia, Z. Chen, A. Chen, M. Fu, Z. Dong, K. Hu, X. Yang, Y. Zou, A. Sun, J. Qian,
[152] M.M. Abdel-Daim, O.E. Kilany, H.A. Khalifa, A.A.M. Ahmed, Allicin ameliorates J. Ge, LCZ696 improves cardiac function via alleviating Drp1-mediated
doxorubicin-induced cardiotoxicity in rats via suppression of oxidative stress, mitochondrial dysfunction in mice with doxorubicin-induced dilated
inflammation and apoptosis, Cancer Chemother. Pharmacol. 80 (4) (2017) cardiomyopathy, J. Mol. Cell. Cardiol. 108 (2017) 138–148.
745–753. [179] X. Liang, S. Wang, L. Wang, A.F. Ceylan, J. Ren, Y. Zhang, Mitophagy inhibitor
[153] B. Ky, P. Vejpongsa, E.T. Yeh, T. Force, J.J. Moslehi, Emerging paradigms in liensinine suppresses doxorubicin-induced cardiotoxicity through inhibition of
cardiomyopathies associated with cancer therapies, Circ. Res. 113 (6) (2013) Drp1-mediated maladaptive mitochondrial fission, Pharmacol. Res. 157 (2020),
754–764. 104846.
[154] T.M.D. Nguyen, Adiponectin: role in physiology and pathophysiology, Int. J. Prev. [180] H. Xu, W. Yu, S. Sun, C. Li, Y. Zhang, J. Ren, Luteolin attenuates doxorubicin-
Med. 11 (2020) 136. induced cardiotoxicity through promoting mitochondrial autophagy, Front.
[155] D. Zhao, C. Xue, J. Li, K. Feng, P. Zeng, Y. Chen, Y. Duan, S. Zhang, X. Li, J. Han, Physiol. 11 (2020) 113.
X. Yang, Adiponectin agonist ADP355 ameliorates doxorubicin-induced [181] J. Govender, B. Loos, E. Marais, A.M. Engelbrecht, Mitochondrial catastrophe
cardiotoxicity by decreasing cardiomyocyte apoptosis and oxidative stress, during doxorubicin-induced cardiotoxicity: a review of the protective role of
Biochem. Biophys. Res. Commun. (2020). melatonin, J. Pineal Res. 57 (4) (2014) 367–380.
[156] T. Souma, N. Suzuki, M. Yamamoto, Renal erythropoietin-producing cells in [182] G. Yang, M. Song, D.H. Hoang, Q.H. Tran, W. Choe, I. Kang, S.S. Kim, J. Ha,
health and disease, Front. Physiol. 6 (2015) 167. Melatonin prevents doxorubicin-induced cardiotoxicity through suppression of

13
P.S. Rawat et al. Biomedicine & Pharmacotherapy 139 (2021) 111708

AMPKα2-dependent mitochondrial damage, Exp. Mol. Med. 52 (12) (2020) [193] A.Z. Ahmed, S.M. Satyam, P. Shetty, M.R. D’Souza, Methyl gallate attenuates
2055–2068. doxorubicin-induced cardiotoxicity in rats by suppressing oxidative stress,
[183] J. Lu, J. Li, Y. Hu, Z. Guo, D. Sun, P. Wang, K. Guo, D.D. Duan, S. Gao, J. Jiang, Scientifica 2021 (2021), 6694340.
J. Wang, P. Liu, Chrysophanol protects against doxorubicin-induced [194] Y. Jiang, Y. Liu, W. Xiao, D. Zhang, X. Liu, H. Xiao, S. You, L. Yuan, Xinmailong
cardiotoxicity by suppressing cellular PARylation, Acta Pharm. Sin. B 9 (4) (2019) attenuates doxorubicin-induced lysosomal dysfunction and oxidative stress in
782–793. H9c2 cells via HO-1, Oxid. Med. Cell. Longev. 2021 (2021), 5896931.
[184] S.H. Jeong, H.K. Kim, I.S. Song, S.J. Lee, K.S. Ko, B.D. Rhee, N. Kim, N. [195] L. Zhang, M. Feng, X. Wang, H. Zhang, J. Ding, Z. Cheng, L. Qian, Peptide
P. Mishchenko, S.A. Fedoryev, V.A. Stonik, J. Han, Echinochrome A protects Szeto‑Schiller 31 ameliorates doxorubicin‑induced cardiotoxicity by inhibiting
mitochondrial function in cardiomyocytes against cardiotoxic drugs, Mar. Drugs the activation of the p38 MAPK signaling pathway, Int. J. Mol. Med. 47 (4) (2021)
12 (5) (2014) 2922–2936. 63.
[185] Z. Xu, S. Lin, W. Wu, H. Tan, Z. Wang, C. Cheng, L. Lu, X. Zhang, Ghrelin prevents [196] A.Z. Ahmed, K.D. Mumbrekar, S.M. Satyam, P. Shetty, M.R. D’Souza, V.K. Singh,
doxorubicin-induced cardiotoxicity through TNF-alpha/NF-kappaB pathways and Chia seed oil ameliorates doxorubicin-induced cardiotoxicity in female wistar
mitochondrial protective mechanisms, Toxicology 247 (2–3) (2008) 133–138. rats: an electrocardiographic, biochemical and histopathological approach,
[186] X. Shi, M. Zhao, C. Fu, A. Fu, Intravenous administration of mitochondria for Cardiovasc. Toxicol. (2021) 1–10.
treating experimental Parkinson’s disease, Mitochondrion 34 (2017) 91–100. [197] F.E. Hashish, M.M. Abdel-Wahed, M.H. El-Odemi, S.S. El-Naidany, M.M. ElBatsh,
[187] K.M. Sadek, S.F.E. Mahmoud, M.F. Zeweil, T.K. Abouzed, Proanthocyanidin Possible protective effects of quercetin on doxorubicin-induced cardiotoxicity in
alleviates doxorubicin-induced cardiac injury by inhibiting NF-kB pathway and rats, Menoufia Med. J. 34 (1) (2021) 333.
modulating oxidative stress, cell cycle, and fibrogenesis, J. Biochem. Mol. Toxicol. [198] Y. Tanaka, T. Nagoshi, A. Yoshii, Y. Oi, H. Takahashi, H. Kimura, K. Ito,
35 (4) (2021) 22716. Y. Kashiwagi, T.D. Tanaka, M. Yoshimura, Xanthine oxidase inhibition attenuates
[188] L. Nie, M. Liu, J. Chen, Q. Wu, Y. Li, J. Yi, X. Zheng, J. Zhang, C. Chu, J. Yang, doxorubicin-induced cardiotoxicity in mice, Free Radic. Biol. Med. 162 (2021)
Hydrogen sulfide ameliorates doxorubicin‑induced myocardial fibrosis in rats via 298–308.
the PI3K/AKT/mTOR pathway, Mol. Med. Rep. 23 (4) (2021). [199] X. Wang, L. Zeng, S. Tu, F. Ye, Z. Zhang, Calycosin-triblock copolymer
[189] C. Zhan, N. Bai, M. Zheng, Y. Wang, Y. Wang, L. Zhang, J. Li, G. Li, H. Zhao, nanomicelles attenuate doxorubicin-induced cardiotoxicity through upregulation
G. Liu, Q. Lou, W. Yang, T. Li, L. Li, W. Li, Tranilast prevents doxorubicin-induced of ERp57, Arab. J. Chem. 14 (2021), 103142.
myocardial hypertrophy and angiotensin II synthesis in rats, Life Sci. 267 (2021), [200] S. Chatterjee, T. Hofer, A. Costa, D. Lu, S. Batkai, S.K. Gupta, E. Bolesani,
118984. R. Zweigerdt, D. Megias, K. Streckfuss-Bömeke, C. Brandenberger, T. Thum,
[190] A.M. Alzahrani, P. Rajendran, V.P. Veeraraghavan, H. Hanieh, Cardiac protective C. Bär, Telomerase therapy attenuates cardiotoxic effects of doxorubicin, Mol.
effect of kirenol against doxorubicin-induced cardiac hypertrophy in H9c2 cells Ther. 29 (4) (2021) 1395–1410.
through Nrf2 signaling via PI3K/AKT pathways, Int. J. Mol. Sci. 22 (6) (2021). [201] K. Feng, Y. Liu, J. Sun, C. Zhao, Y. Duan, W. Wang, K. Yan, X. Yan, H. Sun, Y. Hu,
[191] M.A. Almutawakel, R. Al-Badani, Cardioprotective effect of Malva verticillata Compound Danshen Dripping Pill inhibits doxorubicin or isoproterenol-induced
against doxorubicin-induced toxicity in rats, Clin. Phytosci. 7 (1) (2021) 1–9. cardiotoxicity, Biomed. Pharmacother. 138 (2021), 111531.
[192] W. Zhou, J. Ouyang, N. Hu, G. Li, H. Wang, Protective effect of two alkaloids from [202] D. Karabulut, E. Ozturk, E. Kaymak, A.T. Akin, B. Yakan, Thymoquinone
hippophae rhamnoides linn. against doxorubicin-induced toxicity in H9c2 attenuates doxorubicin-cardiotoxicity in rats, J. Biochem. Mol. Toxicol. 35 (1)
cardiomyoblasts, Molecules 26 (7) (2021) 1946. (2021) 22618.

14

You might also like