Next Article in Journal
Prediction of Soil Pollution Risk Based on Machine Learning and SHAP Interpretable Models in the Nansi Lake, China
Next Article in Special Issue
Nano-Curcumin Mitigates Doxorubicin-Induced Reproductive Toxicity via Antioxidant, Anti-Apoptosis, and SIRT1-Modulating Effects in Rat Model
Previous Article in Journal
Risk Assessment of Micropollutants for Human and Environmental Health: Alignment with the Urban Wastewater Treatment Directive in Southeastern Spain
Previous Article in Special Issue
Oxidative and Molecular–Structural Alterations of Spermatozoa in Swine and Ram Exposed to the Triazole Ipconazole
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pathophysiology of Doxorubicin-Mediated Cardiotoxicity

by
Roberto Arrigoni
1,*,
Emilio Jirillo
2 and
Carlo Caiati
3
1
Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council, 70124 Bari, Italy
2
Interdisciplinary Department of Medicine, Section of Microbiology and Virology, School of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy
3
Unit of Cardiovascular Diseases, Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy
*
Author to whom correspondence should be addressed.
Toxics 2025, 13(4), 277; https://doi.org/10.3390/toxics13040277
Submission received: 12 March 2025 / Revised: 28 March 2025 / Accepted: 3 April 2025 / Published: 5 April 2025
(This article belongs to the Special Issue Drug and Pesticides-Induced Oxidative Stress and Apoptosis)

Abstract

Doxorubicin (DOX) is used for the treatment of various malignancies, including leukemias, lymphomas, sarcomas, and bladder, breast, and gynecological cancers in adults, adolescents, and children. However, DOX causes severe side effects in patients, such as cardiotoxicity, which encompasses heart failure, arrhythmia, and myocardial infarction. DOX-induced cardiotoxicity (DIC) is based on the combination of nuclear-mediated cardiomyocyte death and mitochondrial-mediated death. Oxidative stress, altered autophagy, inflammation, and apoptosis/ferroptosis represent the main pathogenetic mechanisms responsible for DIC. In addition, in vitro and in vivo models of DIC sirtuins (SIRT), and especially, SIRT 1 are reduced, and this event contributes to cardiac damage. In fact, SIRT 1 inhibits reactive oxygen species and NF-kB activation, thus improving myocardial oxidative stress and cardiac remodeling. Therefore, the recovery of SIRT 1 during DIC may represent a therapeutic strategy to limit DIC progression. Natural products, i.e., polyphenols, as well as nano formulations of DOX and iron chelators, are other potential compounds experimented with in models of DIC. At present, few clinical trials are available to confirm the efficacy of these products in DIC. The aim of this review is the description of the pathophysiology of DIC as well as potential drug targets to alleviate DIC.

Graphical Abstract

1. Introduction

Anthracyclines [doxorubicin (DOX), daunorubicin, epirubicin, and idarubicin] are drugs utilized for their effects on cancer, but their use is limited due to cardiotoxicity [1]. The reduction of the left ventricular ejection fraction represents the most severe side effect, which occurs in 9% of patients undergoing treatment for anthracyclines. The other cardiotoxic symptoms encompass congestive heart failure, arrhythmia, and myocardial infarction.
Anthracyclines are used to treat various malignancies, including leukemias, lymphomas, sarcomas, and bladder, breast and gynecological cancers in adults, adolescents, and children [2]. DOX-induced cardiotoxicity (DIC) has been intensively investigated. The cumulative dose represents a major risk of DIC: patients receiving >400 mg/m2 DOX cumulative doses are at very high risk. In particular, the incidence of cardiomyopathy is reported to be 5% at a cumulative dose of 400 mg/m2, 26% at 550 mg/m2, and 48% at 700 mg/m2 [3,4]. However, subclinical cardiac dysfunction can be observed at lower cumulative exposures, up to a mean cumulative dose of 240 mg/m2, with a range of 50 to 375 mg/m2 [5]. Among the side effects, it is possible to distinguish between acute and chronic DIC. Acute DIC shares many similarities with acute myocarditis; in particular, acute doxorubicin cardiotoxicity is considered a rare clinical event, although several studies indicate that it is more common than previously thought (11–21%) and predicts unfavorable outcomes [6]; on the other hand, chronic DIC can appear after months or years from the initial treatment, being characterized by dilated cardiomyopathy and the irreversible reduction of the left ejection fraction, with eccentric ventricular hypertrophy and increased cardiac mass, often associated with the clinical syndrome of heart failure [7,8].
A position paper elaborated by the European Society of Cardiology reported that among a cohort of cancer survivors, one-third of deaths could be attributed to chronic cardiotoxicity [9]. Another report has documented that 20% of cancer survivors manifest left ventricular dysfunction, with a larger proportion in children [10].
Hence, it is imperative to monitor the left ventricular function in patients on DOX, before and after the completion of anthracycline-based chemotherapy, to detect early myocardial damage [11]. In this regard, echocardiography is the more suitable technique since it does not involve exposure to damaging ionizing radiation [12]. With this technique, not only should the systolic function be assessed and monitored but also the diastolic left ventricular function, with the diastolic Doppler parameters of the left ventricular function being even more sensitive than the standard ejection fraction in revealing early myocardial damage [13,14]; also, cardiovascular biomarkers have to be assessed and monitored [15].
Several risk factors predispose patients to DIC, such as hypertension, hyperglycemia, and dyslipidemia [16]. In particular, glycemic and lipidic disorders can increase the levels of fatty acids and cytokines, with accumulation of fat droplets in the myocardium [17]. Other risk factors include the extremes of age, female gender, prior mediastinal radiation therapy, concomitant treatment with cyclophosphamide, trastuzumab or paclitaxel, and the presence of cardiac disease [18]. There is evidence that DOX exerts its anticancer activity through two major mechanisms: (1) inhibiting DNA and RNA synthesis in dividing cells by stopping the replication and transcription processes, and (2) generating iron-mediated free radicals, causing cell damage to cell membranes, proteins, and DNA [19,20,21].
DIC’s etiology is multifactorial, being based on a combination of nuclear-mediated cardiomyocyte death and mitochondrial-mediated death [22,23]. Moreover, cardiomyocytes exhibit low levels of antioxidant protection, being more susceptible to free radicals and reactive oxygen species (ROS) damage [24].
The aim of the present review will be the description of the major mechanisms leading to DIC. A better understanding of the cellular and molecular events implicated in the pathophysiology of DIC may help in searching for potential treatments for DIC prevention or cardiotoxicity reduction.

2. Major Pathogenetic Mechanisms of DIC

DOX toxicity is evident in many organs, such as the brain, liver, cardiac muscle, and skeletal muscle [25]. According to recent research, doxorubicin exerts its anticancer effects through multiple mechanisms; DIC is correlated to DNA instability, oxidative stress, altered autophagy, inflammation, and apoptosis.

2.1. DNA Instability and Topoisomerase Inhibition

Doxorubicin can display its cytotoxic effects by disrupting DNA stability by directly intercalating into the DNA structure (see Figure 1) [26]. The drug’s planar anthracycline structure allows it to intercalate between DNA base pairs, forming a covalent bond with guanine on one strand and hydrogen bonds on the opposite strand, causing torsional strain and physical obstruction of essential processes such as replication and transcription. This intercalation not only distorts the DNA helix but also generates reactive oxygen species (ROS) through redox cycling, further exacerbating the DNA damage via oxidative base modifications and strand breaks. The resulting genomic instability activates the DNA damage response pathways, ultimately leading to cell cycle arrest and apoptosis in rapidly dividing cancer cells.
A critical aspect of doxorubicin’s mechanism is its inhibition of topoisomerase II, an enzyme essential for managing DNA supercoiling during replication and transcription [27]. By stabilizing the topoisomerase II–DNA cleavage complex, doxorubicin prevents the enzyme from religating severed DNA strands, converting transient topoisomerase-II-induced breaks into persistent double-strand breaks (DSBs) [28]. These DSBs overwhelm the cell’s repair capacity, particularly in highly proliferative tumor cells, triggering p53-mediated apoptosis. However, this same mechanism contributes to the drug’s dose-limiting cardiotoxicity, as cardiomyocytes, which have limited regenerative capacity, accumulate irreversible DNA damage and mitochondrial dysfunction due to ROS generation.
Recently, it was demonstrated that doxorubicin can also alter RNA stability by inhibiting the RNA-binding protein (RBP) quaking (QKI) expression, a protein family that belongs to the signal transduction and activation of RNA (STAR) family and plays a crucial role in post-transcriptional gene regulation by binding to specific RNA sequences to influence the splicing, stability, localization, and translation of target mRNAs [29]. QKI is particularly important for cell differentiation, myelination, and cardiovascular development, with key functions in the brain, heart, and vascular systems.

2.2. Oxidative Stress

Oxidative stress is caused by an imbalance between the generation of ROS in tissue and a deficit of antioxidants, as found in DOX-treated cancer patients [30]. DOX possesses a high affinity for myocardial tissue and mitochondrial cardiolipin, thus interacting with mitochondrial DNA, with the inhibition of the respiratory chain [31]. This event leads to ROS generation, with the alteration of the phospholipids of cell membranes, mitochondria and the endoplasmic reticulum (ER), ultimately provoking myocardial cell damage [32].
Furthermore, DOX reduces the levels of glutathione and catalase (CAT), which act as antioxidants, thus aggravating oxidative stress [33]. In detail, DOX exhibits a quinone moiety, thus behaving as an electron acceptor. NADPH oxidase converts DOX into a semiquinone, which in turn reacts with oxygen, forming superoxide, with the contemporary regeneration of the quinone form. Superoxide interacts with nitric oxide (NO) to form the peroxynitrite anion, which is converted into peroxide by superoxide dismutase (SOD). In conclusion, the accumulation of hydroxyl radicals leads to DNA, protein and lipid damage in the context of myocardial cells [34,35,36].
The DOX-induced generation of ROS and reactive nitrogen species (RNS) affects endothelial cell function. In fact, DOX-mediated cardiomyopathy is characterized by increased levels of endothelin-1, activation of the type A and type B receptors, followed by vasoconstriction, and release of NO, adrenomedullin, and prostacyclin [37]. In this framework, it is worth discussing the role of the Yes-associated protein (YAP), which plays a crucial role in cell biogenesis, inducing the CAT and SOD transcriptions [38]. Moreover, in vivo studies have demonstrated that DOX-based therapy is associated with a reduction of the intramyocardial YAP 1 protein, as well as of target genes, such as CTGF, Birc5, and PARK2 [39,40]. Conversely, YAP1 overexpression abrogated DOX-induced cell death, inhibiting caspase-3/7 [40].

2.3. Autophagy

Autophagy consists of altered proteins or organelles, which are enclosed within double-membrane autophagosomes and then transported to lysosomes or vacuoles for degradation and further recycling [41]. Evidence has been provided that autophagy plays a crucial role in maintaining cardiac homeostasis and function, and therefore its dysregulation may cause DIC [42]. In this respect, DOX increases the levels of beclin-1, p62, and the microtubule-associated protein 1A/1B light chain 3 (LC3)-II/LC3-I [43]. Damaged mitochondria are removed through the mitophagy initiation driven by the activation of the PTEN-induced kinase and parkin [43]. Also, there is evidence that DOX alters mitochondrial function by decreasing the mitochondrial membrane potential, with the impairment of oxidative phosphorylation and its biosynthetic ability [44]. Furthermore, DOX abrogates lysosome biosynthesis and catepsin activity, with autophagolysosomal accumulation and the impairment of autophagy [45]. Fusion between autophagosomes and lysosomes promotes autophagic flux, while DOX inhibits the mechanism of fusion by enhancing STAT3 phosphorylation, upregulating lipocailin-2, and abolishing the interaction between syntaxin17 and the vesicle-associated membrane protein 8 [46]. Furthermore, DOX enhances the Toll-like receptor (TLR)-9 expression, inhibiting p38 MAPK, autophagy, while increasing ROS levels. There is evidence that the DOX-induced suppression of the AMPK activation and autophagy can aggravate apoptosis [47]. These events occur in the presence of activated PTEN-induced kinase 1 and parkin. Notably, the use of the mitophagy inhibitor mdivi-1 protects against DIC, preserving the mitochondrial membrane potential and the peroxisome proliferator-activated receptor gamma coactivator 1-alpha [36].

2.4. Inflammation

There is evidence that DIC induces cardiac inflammation via the release of an array of cytokines, which in turn aggravate the myocardial damage [48]. DOX treatment provokes ER stress and enhances mitochondrial iNOS levels in vivo, with the activation of the NF-kB pathway and generation of pro-inflammatory cytokines, e.g., interleukin-6 (IL-6) and IL-8 [49]. Moreover, DOX activates the TLR-4 in myocardial cells, with subsequent upregulation of the NF-kB pathway [50]. Furthermore, DOX generates the TLR2–MyD88 complex, with the activation of NF-kB and release of the tumor necrosis factor-alpha, IL-1 beta, IL-6, IL-8, IL-12, and IL-17, thus contributing to the cardiac inflammatory process and fibrosis [51]. In addition to the above-mentioned pathways, DOX treatment activates the NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome, with caspase-1 activation, secretion of IL-1 beta and induction of pyroptosis, which triggers acute myocardial inflammation [52]. Pyroptosis is a form of pro-inflammatory cell death based on the swelling and plasma membrane rupture caused by the cleavage of gasodermin D, thus leading to the release of IL-1 beta, IL-18 and inflammatory cardiac damage [53]. DOX-induced pyroptosis increases the expression of the Bcl-2/adenovirus E1B interacting protein in mitochondria, with the activation of caspase-3 [54].
DOX treatment has an impact on cardiac smooth muscle cells, which switch from a contractile state toward a synthetic state, also differentiating into macrophage-like cells, which migrate to the intima [55]. These events contribute to vascular wall low-grade inflammation, with mucoid and fibrinoid swelling, necrosis, sclerosis, and hyalinosis, thus leading to further stiffness of the vascular wall. In addition, DOX impairs sarcoplasmic reticulum Ca2+ uptake by binding to cardiac ryanodine receptors and sarco/endoplasmic Ca2+ ATPase [56].
The role of macrophages in DIC has recently been investigated, especially considering the role played by the cardiac immune response in cardiovascular disorders [57,58]. In DOX-treated mice, macrophage infiltration was antecedent to cardiomyocyte damage. Mechanistically, the release of catecholamines by macrophages leads to mitochondrial apoptosis of cardiomyocytes through beta-AR stimulation, while macrophage depletion prevents cardiac damage.

2.5. Apoptosis and Ferroptosis

DIC-induced apoptosis depends on the modulation of the apoptosis-related genes exerted by DOX [59]. In this respect, in DOX-treated cardiomyocytes, the expression of the anti-apoptotic protein, Bcl-2, decreases paralleled with the increase in the Bcl-2-associated X expression, thus leading to the increased mitochondrial membrane permeability, cytochrome c release, and caspase-3 expression, respectively, which generates cardiomyocyte apoptosis [60].
Ferroptosis is a form of apoptosis, which depends on iron and is mediated by lipid peroxidation [61]. Cardiomyocytes treated with DOX undergo ferroptosis due to glutathione peroxidase 4 (GPx4) inactivation [62]. In fact, ferroptosis inhibitors, e.g., Fer-1, upregulate GPx4, with the reduction of Fe2+ accumulation and the mitigation of myocardial damage [63]. In this regard, DOX causes the accumulation of heme oxygenase 1 that can degrade heme [64]. On the other hand, the DOX-mediated methyltransferase-like 14 overexpression upregulates the transferrin receptors and, consequently, the uptake of iron, regulating the KCNQ1OT1–miR-7-5p–TFRC axis [65]. Additionally, DOX induces ferroptosis by the impairment of autophagy through the forkhead box 04 (FOXO4) transcription, and the high-mobility group box 1 nuclear translocation [66]. The major pathogenetic mechanisms of DIC are indicated in Table 1.

3. Role of Sirtuins in DIC

Sirtuins (SIRT) belong to the class-II-type histone deacetylase family, encompassing seven members from SIRT 1 to SIRT7. They modulate tissue metabolism, oxidative stress, and apoptosis through the deacetylation of target proteins, thus influencing cardiovascular function [67]. Evidence has been provided about their regulatory role in the onset of myocardial hypertrophy, ischemia/reperfusion injury, as well as diabetic cardiomyopathy [68]. On these grounds, the potential role of SIRT1–7 in DIC has been explored. In this respect, it has been documented that the SIRT 1 expression is reduced in in vitro and in vivo models of DIC, while its moderate overexpression protects cardiomyocytes from oxidative stress and apoptosis [69]. Moreover, resveratrol (RES), activating SIRT 1, abrogates the mitochondrial dysfunction and oxidative stress exerted by DOX [70].
SIRT 1 inhibits ROS generation, regulating the expression of CAT and manganese (Mn) SOD, deacetylating FOXO1, and thus decreasing cardiac oxidative stress [71]. Furthermore, SIRT 1 deacetylates and activates the peroxisome proliferator-activated receptor gamma coactivator 1 alpha, as well as NF-kB, thus improving myocardial oxidative stress and cardiac remodeling [72]. Also, the inhibition of PARP-2 increases the expression of SIRT 1, alleviating DIC [73].
A series of studies demonstrated that SIRT 1 activation can mitigate DOX-mediated cardiac inflammation in vitro, inhibiting NF-kB activation. For instance, the fibroblast growth factor 21 increases the activation of the SIRT 1/liver kinase B1/AMPK pathway, thus blocking NF-kB p65, with abrogation of the high expression of TNF-alpha and IL-6 [74]. Targeting the paternally expressed gene 3 upregulates SIRT 1 by the inhibition of miR-200a-3p, thus leading to the inhibition of NF-kB and the improvement of DOX-induced inflammation [75]. Some natural compounds can modulate SIRT1 activity. For instance, Jaceosidin inhibits the phosphorylation of IKKbeta and the translocation of NF-kB via SIRT 1 activation, ultimately suppressing NF-kB, while improving DOX-induced inflammation [62]. Of note, Jaceosidin does not impair DOX’s antitumor ability. Other natural compounds, such as calycosin and dihydromyrecitin, abrogate NLRP3 inflammasome, thus mitigating DOX-induced cardiac inflammation [76,77].
As far as DOX-mediated apoptosis is concerned, RES activates SIRT 1, attenuating the acetylation of the p53 protein, and thus alleviating DOX-mediated myocardial cell apoptosis [78]. Also, ferroptosis is reduced by the SIRT 1 activation via the Nrf2/Kelch-like-associated protein, with the mitigation of DIC [79]. SIRT 2 is present in cytoplasm as well as in mitochondria, where it regulates autophagy and mitophagy. For instance, SIRT 2 activates FOXO3a, with the upregulation of manganese superoxide dismutase (MnSOD) and attenuation of the ROS generation by cardiomyocytes [80]. Also, the AMPK modulation by SIRT 2 improves age-related cardiac dysfunction, thus mitigating DIC [81]. Also, the regulation of the SIRT 2/NRF2 pathway through the inhibition of miR-140-5p reduces DOX-induced antioxidative stress [82]. SIRT 3 is located within the mitochondria and its absence causes mitochondrial dysfunction, which results in oxidative stress, apoptosis, and inflammation, as observed in cardiac diseases [83]. Experimentally, the activation of SIRT 3 with RES, daidzein, and tubeimoside could alleviate DOX-induced myocardial inflammation [70,84]. Furthermore, the regulation of SIRT 3 by berberine could suppress DOX-induced cardiac oxidative stress [85]. Also, dichloroacetic acid upregulates the PGC-1 alpha/SIRT 3 pathway, improving DOX-induced mitochondrial dysfunction, oxidative stress, and apoptosis [86]. Furthermore, the Chinese herbal medicine, Qishen granules, increased the SIRT 3 expression, with decreased mitochondrial ROS generation and protection against DIC [87]. With special reference to autophagy, the SIRT 3 overexpression could inhibit miR-34-5p, leading to anti-autophagic activity and protection from DIC [88]. In addition, evidence has been reported that SIRT 3 inhibited the NLRP3 inflammasome, regulating autophagy and mitigating pyroptosis [76].
SIRT 4 is implicated in the regulation of myocardial energy metabolism, but its activity in cardiovascular diseases remains controversial. In fact, it inhibits fatty acid oxidation in muscles, suppressing malonyl-CoA decarboxylase, as well as SIRT 3-mediated MnSOD activity, thus affecting mitochondrial function [89]. Conversely, the overexpression of SIRT 4, as well as its interaction with optic atrophin 1, leads to the inhibition of autophagy and ROS generation [90,91]. With special reference to DIC, it has been documented that the overexpression of SIRT 4 activates the akt/mTOR pathway, thus inhibiting DIC [92].
SIRT 5 is a mitochondrial sirtuin, which plays a role in keeping mitochondrial homeostasis. To the best of our knowledge, one publication has investigated the role of SIRT 5 in DIC, following its overexpression by coenzyme Q 10, with protection from DOX-induced cardiac oxidative stress in mice [93].
SIRT 6 is a nuclear sirtuin, which regulates aging, oxidative stress, inflammation, and autophagy [94]. The SIRT 6 overexpression caused by targeting miR-330-5p inhibits ROS generation, as well as the apoptosis and necrosis induced by the p53/Fas pathway, ultimately leading to the mitigation of oxidative stress and cardiac atrophy related to DOX treatment [80,95]. Furthermore, SIRT 6 enhances autophagy by deacetylating and inhibiting SKG1, thus reducing DIC on the one hand, and on the other hand, augmenting the therapeutic efficacy of DOX through metabolic remodeling [96]. In addition, very recent evidence has documented that SIRT 6 can activate the proliferator-activated receptor alpha, abrogating the myocardial aging induced by DOX [97].
SIRT 7 is a nuclear sirtuin acting on the heart, liver, and spleen. Studies have demonstrated that SIRT 7 reduces myocardial stress via the deacetylation of p53, regulates autophagy through TGF beta, mitigates cardiac hypertrophy through the deacetylation of GATA4 and decreases cardiac hypoxia and apoptosis through the inhibition of miR-148-3p, respectively [98,99,100,101,102].
Sirtuin agonists have been investigated for possible mitigation of DIC. There is evidence that RES can act as a natural agonist of SIRT 1, even if the mechanism of action is still controversial. A report has documented that RES enhances the activity of SIRT 1 via binding to lamin A [103]. Another study has reported that RES molecules interact with the fluorescent moiety of the substrate p53 peptide and the N-terminal regulatory region of SIRT 1, thus enhancing its activity [104].
Other natural products, such as dihydrocoumarin, berberine, and limonin, improve DOX-induced cardiotoxicity, but they exhibit poor bioavailability and low specificity even when encapsulated into liposomes [105]. Furthermore, synthetic SIRT 1 agonists, e.g., SRT1720 have been tested on cardiomyocytes, with the induction of autophagy through the SIRT1/AMPK pathway and protection against hypotoxic stress [106]. However, no evidence of the effects of SRT1720 on DIC has been reported. Parallelly, a few studies have been focused on the effects of sirtuin inhibitors, such as EX527 and 3-TYP, on experimental DIC. Both inhibitors can abrogate the protective effects of RES, berberine, and ononin against DOX-induced cardiotoxicity [107]. The main effects of sirtuins on DIC are illustrated in Table 2.

4. Role of Natural Products in DIC

Evidence has been provided that natural products, such as polyphenols, are endowed with antioxidant and anti-inflammatory activities, neutralizing certain pathways, e.g., NF-kB and NLRP3 inflammasomes involved in mechanisms of various tissue damage [111,112,113,114].
RES in combination with the fibroblast growth factor 1 can ameliorate murine DIC via the activation of the SIRT1-NRF2 pathway [115], as well as ferroptosis acting on the MAPK pathway [109]. Quercetin can abolish ROS production in DIC, preventing the opening of the mitochondrial permeability transition pore [116]. Also, rutin and apigenin have been shown to reduce DOX-induced apoptosis and autophagy [117,118]. Allicin has been shown to improve DIC in rats, suppressing oxidative stress, inflammation, and apoptosis [48]. A series of compounds, e.g., neferine, astragaloside IV, acacia hydaspica, and resolvin D1 have been reported to reduce DIC, suppressing NADH oxidase activity [119,120,121,122]. Evidence has been provided that show Panax ginseng mitigated DIC, upregulating the Nrf2–ARE axis, with a reduction of ROS production [123].

Therapeutic Attempts to Reduce DIC

The encapsulation of DOX in nanostructures has been applied to decrease DIC. Doxil, a pegylated liposomal system into which DOX has been incorporated, has been shown to reduce DIC [124]. In breast cancer patients, liposomal DOX-based chemotherapy was more advantageous in terms of progression-free survival and DIC reduction than conventional DOX [125]. Also, magnetic iron oxide nanoparticles are effective nanocarriers for DOX therapy, but they are not yet in clinical trials [126]. The iron chelator dexrazoxane can bind iron, preventing its entry into cardiomyocytes, thus reducing cardiac damage [127]. Its cardioprotective mechanism involves two key actions: (1) it binds free iron and consequently reduces the ROS formation generated by doxorubicin–iron complexes, thereby minimizing the oxidative damage to cardiomyocytes, and (2) it selectively inhibits topoisomerase IIβ in the heart, preventing doxorubicin-induced DNA double-strand breaks without interfering with the drug’s anticancer efficacy (which primarily depends on topoisomerase IIα inhibition in tumor cells) [128]. Clinical studies have demonstrated that dexrazoxane significantly reduces the risk of cardiomyopathy and heart failure in patients receiving high-dose or long-term doxorubicin therapy, particularly in pediatric and adult oncology populations [129,130]. While its use was initially restricted due to theoretical concerns about secondary malignancies (largely unsubstantiated in long-term follow-ups), the current guidelines support its use in high-risk patients to preserve cardiac function without compromising chemotherapy outcomes [131,132]. Overall, while dexrazoxane is highly effective in preventing doxorubicin-induced cardiotoxicity, its use requires careful patient selection and monitoring to manage the potential adverse effects. Ongoing research is exploring optimized dosing strategies and novel derivatives to further enhance its protective effects. Carvedilol, a vasodilator and beta adrenoceptor antagonist, is endowed with antioxidant effects, thus reducing DIC [133]. The statin rosuvastatin has demonstrated cardioprotective effects in mice treated with DOX, but clinical trials have generated controversial results [134,135].
The RNA-binding protein (RBP) seems to be another promising target to regulate cardiac pathophysiology: it was recently reported that Qki 5 overexpression significantly reduces doxorubicin-induced toxicity by modulating the biogenesis and function of cardioprotective circular RNAs, which help maintain cellular homeostasis and mitigate DNA damage and oxidative stress [136].
Quite interestingly, a very recent report has documented that inhibition of neutrophil extracellular traps (NETs) prevents DIC in mice [137]. In detail, NETs prime macrophages for IL18 release in DOX-treated-mice with the production of interferon-gamma and the tumor necrosis factor-alpha from activated T cells. Both cytokines downregulate the expression of Cx43, provoking cardiac conduction disorders. In reference to the above concept, the accumulation of NETs has been found in the blood of DOX-treated cancer patients [138]. Therefore, targeting NETs may represent a new therapeutic strategy in human DIC.

5. Conclusions

DOX treatment exhibits severe cardiac side effects, including arrhythmia, heart failure, and ventricular dysfunction. Oxidative stress, DNA instability, inflammation, autophagy, and apoptosis are the major causes of DIC.
Understanding the balance between its cytotoxic and adverse effects remains crucial to optimizing its therapeutic outcomes in cancer treatment. A better clarification of the mechanism responsible for DIC may help in the identification of drugs able to prevent or alleviate DIC. Dexrazoxane is the only drug approved by the FDA; however, some side effects have been reported after clinical use. Other strategies based on DOX nanoformulations or administration of natural compounds have demonstrated their efficacy only in pre-clinical studies. Therefore, this area of investigation needs to be more explored to discover more appropriate compounds for treating DIC.

Author Contributions

Conceptualization, E.J. and R.A.; data curation, C.C.; writing—original draft preparation, E.J.; writing—review and editing, R.A. and C.C.; supervision, R.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

The authors confirm that the data supporting the findings of this study are available within the article.

Acknowledgments

The authors would like to thank Rosa Porro for her technical support.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
CATICatalase
DICJDoxorubicin-induced cardiotoxicity
DOXDoxorubicin
EREndoplasmic reticulum
GPx4Glutathione peroxidase 4
ILInterleukin
MnSODManganese superoxide dismutase
NLRP3Nod-like receptor thermal protein domain-associated protein 3
NONitric oxide
NOXNADH oxidase
RBPRNA-binding protein
RESResveratrol
ROSReactive oxygen species
SIRTSirtuins
TLRToll-like receptor
YAPYes-associated protein

References

  1. Cardinale, D.; Colombo, A.; Bacchiani, G.; Tedeschi, I.; Meroni, C.A.; Veglia, F.; Civelli, M.; Lamantia, G.; Colombo, N.; Curigliano, G.; et al. Early detection of anthracycline cardiotoxicity and improvement with heart failure therapy. Circulation 2015, 131, 1981–1988. [Google Scholar] [CrossRef]
  2. Hudson, M.M.; Ness, K.K.; Gurney, J.G.; Mulrooney, D.A.; Chemaitilly, W.; Krull, K.R.; Green, D.M.; Armstrong, G.T.; Nottage, K.A.; Jones, K.E.; et al. Clinical ascertainment of health outcomes among adults treated for childhood cancer. JAMA 2013, 309, 2371–2381. [Google Scholar] [CrossRef]
  3. Li, D.L.; Hill, J.A. Cardiomyocyte autophagy and cancer chemotherapy. J. Mol. Cell. Cardiol. 2014, 71, 54–61. [Google Scholar] [CrossRef]
  4. Lyon, A.R.; López-Fernández, T.; Couch, L.S.; Asteggiano, R.; Aznar, M.C.; Bergler-Klein, J.; Boriani, G.; Cardinale, D.; Cordoba, R.; Cosyns, B.; et al. 2022 ESC Guidelines on cardio-oncology developed in collaboration with the European Hematology Association (EHA), the European Society for Therapeutic Radiology and Oncology (ESTRO) and the International Cardio-Oncology Society (IC-OS). Eur. Heart J. 2022, 43, 4229–4361. [Google Scholar] [CrossRef]
  5. Drafts, B.C.; Twomley, K.M.; D’Agostino, R.; Lawrence, J.; Avis, N.; Ellis, L.R.; Thohan, V.; Jordan, J.; Melin, S.A.; Torti, F.M.; et al. Low to moderate dose anthracycline-based chemotherapy is associated with early noninvasive imaging evidence of subclinical cardiovascular disease. JACC Cardiovasc. Imaging 2013, 6, 877–885. [Google Scholar] [CrossRef]
  6. Luminari, S.; Montanini, A.; Caballero, D.; Bologna, S.; Notter, M.; Dyer, M.J.S.; Chiappella, A.; Briones, J.; Petrini, M.; Barbato, A.; et al. Nonpegylated liposomal doxorubicin (MyocetTM) combination (R-COMP) chemotherapy in elderly patients with diffuse large B-cell lymphoma (DLBCL): Results from the phase II EUR018 trial. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2010, 21, 1492–1499. [Google Scholar] [CrossRef]
  7. Mukku, R.B.; Fonarow, G.C.; Watson, K.E.; Ajijola, O.A.; Depasquale, E.C.; Nsair, A.; Baas, A.S.; Deng, M.C.; Yang, E.H. Heart Failure Therapies for End-Stage Chemotherapy-Induced Cardiomyopathy. J. Card. Fail. 2016, 22, 439–448. [Google Scholar] [CrossRef]
  8. Curigliano, G.; Cardinale, D.; Dent, S.; Criscitiello, C.; Aseyev, O.; Lenihan, D.; Cipolla, C.M. Cardiotoxicity of anticancer treatments: Epidemiology, detection, and management. CA. Cancer J. Clin. 2016, 66, 309–325. [Google Scholar] [CrossRef]
  9. Zamorano, J.L.; Lancellotti, P.; Muñoz, D.R.; Aboyans, V.; Asteggiano, R.; Galderisi, M.; Habib, G.; Lenihan, D.J.; Lip, G.Y.H.; Lyon, A.R.; et al. 2016 ESC Position Paper on cancer treatments and cardiovascular toxicity developed under the auspices of the ESC Committee for Practice Guidelines: The Task Force for cancer treatments and cardiovascular toxicity of the European Society of Cardiology (ESC). Eur. Heart J. 2016, 37, 2768–2801. [Google Scholar] [CrossRef]
  10. Linders, A.N.; Dias, I.B.; Fernández, L.T.; Tocchetti, C.G.; Bomer, N.; Van der Meer, P. A review of the pathophysiological mechanisms of doxorubicin-induced cardiotoxicity and aging. npj Aging 2024, 10, 9. [Google Scholar] [CrossRef]
  11. Belger, C.; Abrahams, C.; Imamdin, A.; Lecour, S. Doxorubicin-induced cardiotoxicity and risk factors. Int. J. Cardiol. Heart Vasc. 2024, 50, 101332. [Google Scholar] [CrossRef]
  12. Picano, E. Sustainability of medical imaging. BMJ 2004, 328, 578–580. [Google Scholar] [CrossRef]
  13. Upshaw, J.N.; Finkelman, B.; Hubbard, R.A.; Smith, A.M.; Narayan, H.K.; Arndt, L.; Domchek, S.; DeMichele, A.; Fox, K.; Shah, P.; et al. Comprehensive Assessment of Changes in Left Ventricular Diastolic Function With Contemporary Breast Cancer Therapy. JACC Cardiovasc. Imaging 2020, 13, 198–210. [Google Scholar] [CrossRef]
  14. Caiati, C.; Argentiero, A.; Forleo, C.; Favale, S.; Lepera, M.E. Predictors of Exercise Capacity in Dilated Cardiomyopathy with Focus on Pulmonary Venous Flow Recorded with Transesophageal Eco-Doppler. J. Clin. Med. 2021, 10, 5954. [Google Scholar] [CrossRef]
  15. Demissei, B.G.; Hubbard, R.A.; Zhang, L.; Smith, A.M.; Sheline, K.; McDonald, C.; Narayan, V.; Domchek, S.M.; DeMichele, A.; Shah, P.; et al. Changes in Cardiovascular Biomarkers with Breast Cancer Therapy and Associations with Cardiac Dysfunction. J. Am. Heart Assoc. 2020, 9, e014708. [Google Scholar] [CrossRef]
  16. Mohammed, T.; Singh, M.; Tiu, J.G.; Kim, A.S. Etiology and management of hypertension in patients with cancer. Cardio-Oncol. Lond. Engl. 2021, 7, 14. [Google Scholar] [CrossRef]
  17. Qiu, S.; Zhou, T.; Qiu, B.; Zhang, Y.; Zhou, Y.; Yu, H.; Zhang, J.; Liu, L.; Yuan, L.; Yang, G.; et al. Risk Factors for Anthracycline-Induced Cardiotoxicity. Front. Cardiovasc. Med. 2021, 8, 736854. [Google Scholar] [CrossRef]
  18. Volkova, M.; Russell, R. Anthracycline Cardiotoxicity: Prevalence, Pathogenesis and Treatment. Curr. Cardiol. Rev. 2011, 7, 214–220. [Google Scholar] [CrossRef]
  19. Gewirtz, D. A critical evaluation of the mechanisms of action proposed for the antitumor effects of the anthracycline antibiotics adriamycin and daunorubicin. Biochem. Pharmacol. 1999, 57, 727–741. [Google Scholar] [CrossRef]
  20. Cagel, M.; Grotz, E.; Bernabeu, E.; Moretton, M.A.; Chiappetta, D.A. Doxorubicin: Nanotechnological overviews from bench to bedside. Drug Discov. Today 2017, 22, 270–281. [Google Scholar] [CrossRef]
  21. Kciuk, M.; Gielecińska, A.; Mujwar, S.; Kołat, D.; Kałuzińska-Kołat, Ż.; Celik, I.; Kontek, R. Doxorubicin-An Agent with Multiple Mechanisms of Anticancer Activity. Cells 2023, 12, 659. [Google Scholar] [CrossRef]
  22. Jean, S.R.; Tulumello, D.V.; Riganti, C.; Liyanage, S.U.; Schimmer, A.D.; Kelley, S.O. Mitochondrial Targeting of Doxorubicin Eliminates Nuclear Effects Associated with Cardiotoxicity. ACS Chem. Biol. 2015, 10, 2007–2015. [Google Scholar] [CrossRef]
  23. Doroshow, J.H.; Locker, G.Y.; Myers, C.E. Enzymatic defenses of the mouse heart against reactive oxygen metabolites: Alterations produced by doxorubicin. J. Clin. Investig. 1980, 65, 128–135. [Google Scholar] [CrossRef]
  24. Sun, X.; Zhou, L.; Han, Y.; Yang, Q.; Li, X.; Xin, B.; Chi, M.; Wang, Y.; Guo, C. Scutellarin Attenuates Doxorubicin-Induced Cardiotoxicity by Inhibiting Myocardial Fibrosis, Apoptosis and Autophagy in Rats. Chem. Biodivers. 2023, 20, e202200450. [Google Scholar] [CrossRef]
  25. Pugazhendhi, A.; Edison, T.N.J.I.; Velmurugan, B.K.; Jacob, J.A.; Karuppusamy, I. Toxicity of Doxorubicin (Dox) to different experimental organ systems. Life Sci. 2018, 200, 26–30. [Google Scholar] [CrossRef]
  26. Yang, F.; Teves, S.S.; Kemp, C.J.; Henikoff, S. Doxorubicin, DNA torsion, and chromatin dynamics. Biochim. Et Biophys. Acta (BBA)—Rev. Cancer 2014, 1845, 84–89. [Google Scholar] [CrossRef]
  27. Renu, K.; Abilash, V.G.; P.B., T.P.; Arunachalam, S. Molecular mechanism of doxorubicin-induced cardiomyopathy—An update. Eur. J. Pharmacol. 2018, 818, 241–253. [Google Scholar] [CrossRef]
  28. Nedělníková, A.; Stadlbauer, P.; Otyepka, M.; Kührová, P.; Paloncýová, M. Atomistic Insights Into Interaction of Doxorubicin With DNA: From Duplex to Nucleosome. J. Comput. Chem. 2025, 46, e70035. [Google Scholar] [CrossRef] [PubMed]
  29. Zhu, W.; Yang, W.; Sun, G.; Huang, J. RNA-binding protein quaking: A multifunctional regulator in tumour progression. Ann. Med. 2025, 57, 2443046. [Google Scholar] [CrossRef] [PubMed]
  30. Songbo, M.; Lang, H.; Xinyong, C.; Bin, X.; Ping, Z.; Liang, S. Oxidative stress injury in doxorubicin-induced cardiotoxicity. Toxicol. Lett. 2019, 307, 41–48. [Google Scholar] [CrossRef] [PubMed]
  31. Saleh, M.F.; Elsayad, M.E.; Goda, A.E. Mitigation of doxorubicin-induced cardiotoxicity by dichloroacetate: Potential roles of restoration of PGC-1α/SIRT3 signaling and suppression of oxidative stress and apoptosis. Eur. Rev. Med. Pharmacol. Sci. 2021, 25, 6573–6584. [Google Scholar] [CrossRef] [PubMed]
  32. Doroshow, J.H.; Davies, K.J. Redox cycling of anthracyclines by cardiac mitochondria. II. Formation of superoxide anion, hydrogen peroxide, and hydroxyl radical. J. Biol. Chem. 1986, 261, 3068–3074. [Google Scholar]
  33. Sangomla, S.; Saifi, M.A.; Khurana, A.; Godugu, C. Nanoceria ameliorates doxorubicin induced cardiotoxicity: Possible mitigation via reduction of oxidative stress and inflammation. J. Trace Elem. Med. Biol. 2018, 47, 53–62. [Google Scholar] [CrossRef]
  34. Pecoraro, M.; Pala, B.; Di Marcantonio, M.C.; Muraro, R.; Marzocco, S.; Pinto, A.; Mincione, G.; Popolo, A. Doxorubicin-induced oxidative and nitrosative stress: Mitochondrial connexin 43 is at the crossroads. Int. J. Mol. Med. 2020, 46, 1197–1209. [Google Scholar] [CrossRef] [PubMed]
  35. Mukhopadhyay, P.; Rajesh, M.; Bátkai, S.; Kashiwaya, Y.; Haskó, G.; Liaudet, L.; Szabó, C.; Pacher, P. Role of superoxide, nitric oxide, and peroxynitrite in doxorubicin-induced celldeath in vivo and in vitro. Am. J. Physiol. Heart Circ. Physiol. 2009, 296, H1466–H1483. [Google Scholar] [CrossRef] [PubMed]
  36. Li, D.L.; Wang, Z.V.; Ding, G.; Tan, W.; Luo, X.; Criollo, A.; Xie, M.; Jiang, N.; May, H.; Kyrychenko, V.; et al. Doxorubicin Blocks Cardiomyocyte Autophagic Flux by Inhibiting Lysosome Acidification. Circulation 2016, 133, 1668–1687. [Google Scholar] [CrossRef]
  37. Podyacheva, E.; Danilchuk, M.; Toropova, Y. Molecular mechanisms of endothelial remodeling under doxorubicin treatment. Biomed. Pharmacother. Biomed. Pharmacother. 2023, 162, 114576. [Google Scholar] [CrossRef]
  38. Ma, K.; Xu, Q.; Wang, S.; Zhang, W.; Liu, M.; Liang, S.; Zhu, H.; Xu, N. Nuclear accumulation of Yes-Associated Protein (YAP) maintains the survival of doxorubicin-induced senescent cells by promoting survivin expression. Cancer Lett. 2016, 375, 84–91. [Google Scholar] [CrossRef]
  39. She, G.; Du, J.-C.; Wu, W.; Pu, T.-T.; Zhang, Y.; Bai, R.-Y.; Zhang, Y.; Pang, Z.-D.; Wang, H.-F.; Ren, Y.-J.; et al. Hippo pathway activation mediates chemotherapy-induced anti-cancer effect and cardiomyopathy through causing mitochondrial damage and dysfunction. Theranostics 2023, 13, 560–577. [Google Scholar] [CrossRef]
  40. Takaguri, A.; Akihiro, O.; Sasano, J.; Satoh, K. Involvement of Yes-associated protein 1 (YAP1) in doxorubicin-induced cytotoxicity in H9c2 cardiac cells. Cell Biol. Int. 2020, 44, 873–881. [Google Scholar] [CrossRef]
  41. Aman, Y.; Schmauck-Medina, T.; Hansen, M.; Morimoto, R.I.; Simon, A.K.; Bjedov, I.; Palikaras, K.; Simonsen, A.; Johansen, T.; Tavernarakis, N.; et al. Autophagy in healthy aging and disease. Nat. Aging 2021, 1, 634–650. [Google Scholar] [CrossRef] [PubMed]
  42. Xiao, B.; Hong, L.; Cai, X.; Mei, S.; Zhang, P.; Shao, L. The true colors of autophagy in doxorubicin-induced cardiotoxicity (Review). Oncol. Lett. 2019, 18, 2165–2172. [Google Scholar] [CrossRef] [PubMed]
  43. Yin, J.; Guo, J.; Zhang, Q.; Cui, L.; Zhang, L.; Zhang, T.; Zhao, J.; Li, J.; Middleton, A.; Carmichael, P.L.; et al. Doxorubicin-induced mitophagy and mitochondrial damage is associated with dysregulation of the PINK1/parkin pathway. Toxicol. Vitro Int. J. Publ. Assoc. BIBRA 2018, 51, 1–10. [Google Scholar] [CrossRef] [PubMed]
  44. Varela-López, A.; Battino, M.; Navarro-Hortal, M.D.; Giampieri, F.; Forbes-Hernández, T.Y.; Romero-Márquez, J.M.; Collado, R.; Quiles, J.L. An update on the mechanisms related to cell death and toxicity of doxorubicin and the protective role of nutrients. Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2019, 134, 110834. [Google Scholar] [CrossRef]
  45. Abdullah, C.S.; Alam, S.; Aishwarya, R.; Miriyala, S.; Bhuiyan, M.A.N.; Panchatcharam, M.; Pattillo, C.B.; Orr, A.W.; Sadoshima, J.; Hill, J.A.; et al. Doxorubicin-induced cardiomyopathy associated with inhibition of autophagic degradation process and defects in mitochondrial respiration. Sci. Rep. 2019, 9, 2002. [Google Scholar] [CrossRef]
  46. Toda, N.; Sato, T.; Muraoka, M.; Lin, D.; Saito, M.; Li, G.; Song, Q.-C.; Yanagisawa, T.; Yamauchi, M. Doxorubicin induces cardiomyocyte death owing to the accumulation of dysfunctional mitochondria by inhibiting the autophagy fusion process. Free Radic. Biol. Med. 2023, 195, 47–57. [Google Scholar] [CrossRef]
  47. Li, X.; Wang, X.; Wang, B.; Chi, W.; Li, Z.; Zhang, M.; Shen, Y.; Liu, X.; Lu, Y.; Liu, Y. Dihydromyricetin protects against Doxorubicin-induced cardiotoxicity through activation of AMPK/mTOR pathway. Phytomedicine Int. J. Phytother. Phytopharm. 2022, 99, 154027. [Google Scholar] [CrossRef]
  48. Abdel-Daim, M.M.; Kilany, O.E.; Khalifa, H.A.; Ahmed, A.A.M. Allicin ameliorates doxorubicin-induced cardiotoxicity in rats via suppression of oxidative stress, inflammation and apoptosis. Cancer Chemother. Pharmacol. 2017, 80, 745–753. [Google Scholar] [CrossRef]
  49. Bagchi, A.K.; Malik, A.; Akolkar, G.; Jassal, D.S.; Singal, P.K. Endoplasmic Reticulum Stress Promotes iNOS/NO and Influences Inflammation in the Development of Doxorubicin-Induced Cardiomyopathy. Antioxidants 2021, 10, 1897. [Google Scholar] [CrossRef]
  50. Yarmohammadi, F.; Karbasforooshan, H.; Hayes, A.W.; Karimi, G. Inflammation suppression in doxorubicin-induced cardiotoxicity: Natural compounds as therapeutic options. Naunyn. Schmiedebergs Arch. Pharmacol. 2021, 394, 2003–2011. [Google Scholar] [CrossRef]
  51. Ye, S.; Su, L.; Shan, P.; Ye, B.; Wu, S.; Liang, G.; Huang, W. LCZ696 Attenuated Doxorubicin-Induced Chronic Cardiomyopathy Through the TLR2-MyD88 Complex Formation. Front. Cell Dev. Biol. 2021, 9, 654051. [Google Scholar] [CrossRef] [PubMed]
  52. Wei, S.; Ma, W.; Li, X.; Jiang, C.; Sun, T.; Li, Y.; Zhang, B.; Li, W. Involvement of ROS/NLRP3 Inflammasome Signaling Pathway in Doxorubicin-Induced Cardiotoxicity. Cardiovasc. Toxicol. 2020, 20, 507–519. [Google Scholar] [CrossRef]
  53. Ye, B.; Shi, X.; Xu, J.; Dai, S.; Xu, J.; Fan, X.; Han, B.; Han, J. Gasdermin D mediates doxorubicin-induced cardiomyocyte pyroptosis and cardiotoxicity via directly binding to doxorubicin and changes in mitochondrial damage. Transl. Res. J. Lab. Clin. Med. 2022, 248, 36–50. [Google Scholar] [CrossRef]
  54. Lan, Y.; Wang, Y.; Huang, K.; Zeng, Q. Heat Shock Protein 22 Attenuates Doxorubicin-Induced Cardiotoxicity via Regulating Inflammation and Apoptosis. Front. Pharmacol. 2020, 11, 257. [Google Scholar] [CrossRef]
  55. Xu, A.; Deng, F.; Chen, Y.; Kong, Y.; Pan, L.; Liao, Q.; Rao, Z.; Xie, L.; Yao, C.; Li, S.; et al. NF-κB pathway activation during endothelial-to-mesenchymal transition in a rat model of doxorubicin-induced cardiotoxicity. Biomed. Pharmacother. Biomedecine Pharmacother. 2020, 130, 110525. [Google Scholar] [CrossRef]
  56. Avagimyan, A.A.; Mkrtchyan, L.H.; Gevorkyan, A.A.; Kononchuk, N.B.; Kakturskiy, L.V.; Djndoyan, Z.T. Relationship between Chemotherapy and Atrial Fibrillation: Clinical Case. Ration. Pharmacother. Cardiol. 2021, 17, 785–791. [Google Scholar] [CrossRef]
  57. Cerasuolo, F.A.; Gambardella, J.; Santulli, G.; Fiordelisi, A.; Wang, X.; Prevete, N.; Sommella, E.; Avvisato, R.; Buonaiuto, A.; Altobelli, G.G.; et al. Macrophages participate in doxorubicin-induced cardiac damage. Vascul. Pharmacol. 2024, 155, 107309. [Google Scholar] [CrossRef]
  58. Caiati, C.; Jirillo, E. The Immune System, An Arrow into the Heart. Principles of Cardioimmunology, An Emerging Branch in Medicine. Curr. Vasc. Pharmacol. 2025, 23, 162–171. [Google Scholar] [CrossRef]
  59. Kitakata, H.; Endo, J.; Ikura, H.; Moriyama, H.; Shirakawa, K.; Katsumata, Y.; Sano, M. Therapeutic Targets for DOX-Induced Cardiomyopathy: Role of Apoptosis vs. Ferroptosis. Int. J. Mol. Sci. 2022, 23, 1414. [Google Scholar] [CrossRef]
  60. Zhu, J.; Zhang, X.; Xie, H.; Wang, Y.; Zhang, X.; Lin, Z. Cardiomyocyte Stim1 Deficiency Exacerbates Doxorubicin Cardiotoxicity by Magnification of Endoplasmic Reticulum Stress. J. Inflamm. Res. 2021, 14, 3945–3958. [Google Scholar] [CrossRef]
  61. Jiang, X.; Stockwell, B.R.; Conrad, M. Ferroptosis: Mechanisms, biology and role in disease. Nat. Rev. Mol. Cell Biol. 2021, 22, 266–282. [Google Scholar] [CrossRef] [PubMed]
  62. Liu, Y.; Zhou, L.; Du, B.; Liu, Y.; Xing, J.; Guo, S.; Li, L.; Chen, H. Protection against Doxorubicin-Related Cardiotoxicity by Jaceosidin Involves the Sirt1 Signaling Pathway. Oxid. Med. Cell. Longev. 2021, 2021, 9984330. [Google Scholar] [CrossRef]
  63. Tai, P.; Chen, X.; Jia, G.; Chen, G.; Gong, L.; Cheng, Y.; Li, Z.; Wang, H.; Chen, A.; Zhang, G.; et al. WGX50 mitigates doxorubicin-induced cardiotoxicity through inhibition of mitochondrial ROS and ferroptosis. J. Transl. Med. 2023, 21, 823. [Google Scholar] [CrossRef] [PubMed]
  64. Hirschhorn, T.; Stockwell, B.R. The development of the concept of ferroptosis. Free Radic. Biol. Med. 2019, 133, 130–143. [Google Scholar] [CrossRef]
  65. Zhuang, S.; Ma, Y.; Zeng, Y.; Lu, C.; Yang, F.; Jiang, N.; Ge, J.; Ju, H.; Zhong, C.; Wang, J.; et al. METTL14 promotes doxorubicin-induced cardiomyocyte ferroptosis by regulating the KCNQ1OT1-miR-7-5p-TFRC axis. Cell Biol. Toxicol. 2023, 39, 1015–1035. [Google Scholar] [CrossRef]
  66. Kawalec, P.; Martens, M.D.; Field, J.T.; Mughal, W.; Caymo, A.M.; Chapman, D.; Xiang, B.; Ghavami, S.; Dolinsky, V.W.; Gordon, J.W. Differential impact of doxorubicin dose on cell death and autophagy pathways during acute cardiotoxicity. Toxicol. Appl. Pharmacol. 2022, 453, 116210. [Google Scholar] [CrossRef] [PubMed]
  67. Matsushima, S.; Sadoshima, J. The role of sirtuins in cardiac disease. Am. J. Physiol. Heart Circ. Physiol. 2015, 309, H1375–H1389. [Google Scholar] [CrossRef]
  68. Wang, Y.; Li, Y.; Ding, H.; Li, D.; Shen, W.; Zhang, X. The Current State of Research on Sirtuin-Mediated Autophagy in Cardiovascular Diseases. J. Cardiovasc. Dev. Dis. 2023, 10, 382. [Google Scholar] [CrossRef]
  69. Hao, J.; Xi, Y.; Jiao, L.; Wen, X.; Wu, R.; Chang, G.; Sun, F.; Wei, C.; Li, H. Exogenous hydrogen sulfide inhibits the senescence of cardiomyocytes through modulating mitophagy in rats. Cell. Signal. 2022, 100, 110465. [Google Scholar] [CrossRef]
  70. Zhang, Q.; Zhang, Y.; Xie, B.; Liu, D.; Wang, Y.; Zhou, Z.; Zhang, Y.; King, E.; Tse, G.; Liu, T. Resveratrol activation of SIRT1/MFN2 can improve mitochondria function, alleviating doxorubicin-induced myocardial injury. Cancer Innov. 2023, 2, 253–264. [Google Scholar] [CrossRef]
  71. Li, S.; Zhu, Z.; Xue, M.; Yi, X.; Liang, J.; Niu, C.; Chen, G.; Shen, Y.; Zhang, H.; Zheng, J.; et al. Fibroblast growth factor 21 protects the heart from angiotensin II-induced cardiac hypertrophy and dysfunction via SIRT1. Biochim. Biophys. Acta BBA Mol. Basis Dis. 2019, 1865, 1241–1252. [Google Scholar] [CrossRef] [PubMed]
  72. Ren, B.; Feng, J.; Yang, N.; Guo, Y.; Chen, C.; Qin, Q. Ginsenoside Rg3 attenuates angiotensin II-induced myocardial hypertrophy through repressing NLRP3 inflammasome and oxidative stress via modulating SIRT1/NF-κB pathway. Int. Immunopharmacol. 2021, 98, 107841. [Google Scholar] [CrossRef] [PubMed]
  73. Huang, C.; Zhang, X.; Wang, S.; Shen, A.; Xu, T.; Hou, Y.; Gao, S.; Xie, Y.; Zeng, Y.; Chen, J.; et al. PARP-2 mediates cardiomyocyte aging and damage induced by doxorubicin through SIRT1 Inhibition. Apoptosis Int. J. Program. Cell Death 2024, 29, 816–834. [Google Scholar] [CrossRef] [PubMed]
  74. Zhang, X.; Huang, C.; Hou, Y.; Jiang, S.; Zhang, Y.; Wang, S.; Chen, J.; Lai, J.; Wu, L.; Duan, H.; et al. Research progress on the role and mechanism of Sirtuin family in doxorubicin cardiotoxicity. Phytomedicine 2024, 129, 155673. [Google Scholar] [CrossRef]
  75. Fu, Q.; Pan, H.; Tang, Y.; Rong, J.; Zheng, Z. MiR-200a-3p Aggravates DOX-Induced Cardiotoxicity by Targeting PEG3 Through SIRT1/NF-κB Signal Pathway. Cardiovasc. Toxicol. 2021, 21, 302–313. [Google Scholar] [CrossRef]
  76. Sun, Z.; Lu, W.; Lin, N.; Lin, H.; Zhang, J.; Ni, T.; Meng, L.; Zhang, C.; Guo, H. Dihydromyricetin alleviates doxorubicin-induced cardiotoxicity by inhibiting NLRP3 inflammasome through activation of SIRT1. Biochem. Pharmacol. 2020, 175, 113888. [Google Scholar] [CrossRef]
  77. Zhai, J.; Tao, L.; Zhang, S.; Gao, H.; Zhang, Y.; Sun, J.; Song, Y.; Qu, X. Calycosin ameliorates doxorubicin-induced cardiotoxicity by suppressing oxidative stress and inflammation via the sirtuin 1-NOD-like receptor protein 3 pathway. Phytother. Res. PTR 2020, 34, 649–659. [Google Scholar] [CrossRef]
  78. Zhang, C.; Feng, Y.; Qu, S.; Wei, X.; Zhu, H.; Luo, Q.; Liu, M.; Chen, G.; Xiao, X. Resveratrol attenuates doxorubicin-induced cardiomyocyte apoptosis in mice through SIRT1-mediated deacetylation of p53. Cardiovasc. Res. 2011, 90, 538–545. [Google Scholar] [CrossRef]
  79. Wang, W.; Zhong, X.; Fang, Z.; Li, J.; Li, H.; Liu, X.; Yuan, X.; Huang, W.; Huang, Z. Cardiac sirtuin1 deficiency exacerbates ferroptosis in doxorubicin-induced cardiac injury through the Nrf2/Keap1 pathway. Chem. Biol. Interact. 2023, 377, 110469. [Google Scholar] [CrossRef]
  80. Wu, S.; Lan, J.; Li, L.; Wang, X.; Tong, M.; Fu, L.; Zhang, Y.; Xu, J.; Chen, X.; Chen, H.; et al. Sirt6 protects cardiomyocytes against doxorubicin-induced cardiotoxicity by inhibiting P53/Fas-dependent cell death and augmenting endogenous antioxidant defense mechanisms. Cell Biol. Toxicol. 2023, 39, 237–258. [Google Scholar] [CrossRef]
  81. Tang, X.; Chen, X.-F.; Chen, H.-Z.; Liu, D.-P. Mitochondrial Sirtuins in cardiometabolic diseases. Clin. Sci. 2017, 131, 2063–2078. [Google Scholar] [CrossRef]
  82. Zhao, L.; Qi, Y.; Xu, L.; Tao, X.; Han, X.; Yin, L.; Peng, J. MicroRNA-140-5p aggravates doxorubicin-induced cardiotoxicity by promoting myocardial oxidative stress via targeting Nrf2 and Sirt2. Redox Biol. 2018, 15, 284–296. [Google Scholar] [CrossRef] [PubMed]
  83. Sun, W.; Liu, C.; Chen, Q.; Liu, N.; Yan, Y.; Liu, B. SIRT3: A New Regulator of Cardiovascular Diseases. Oxid. Med. Cell. Longev. 2018, 2018, 7293861. [Google Scholar] [CrossRef]
  84. LI, H.; Zhang, M.; Wang, Y.; Gong, K.; Yan, T.; Wang, D.; Meng, X.; Yang, X.; Chen, Y.; Han, J.; et al. Daidzein alleviates doxorubicin-induced heart failure via the SIRT3/FOXO3a signaling pathway. Food Funct. 2022, 13, 9576–9588. [Google Scholar] [CrossRef]
  85. Coelho, A.R.; Martins, T.R.; Couto, R.; Deus, C.; Pereira, C.V.; Simões, R.F.; Rizvanov, A.A.; Silva, F.; Cunha-Oliveira, T.; Oliveira, P.J.; et al. Berberine-induced cardioprotection and Sirt3 modulation in doxorubicin-treated H9c2 cardiomyoblasts. Biochim. Biophys. Acta Mol. Basis Dis. 2017, 1863, 2904–2923. [Google Scholar] [CrossRef]
  86. Tomczyk, M.M.; Cheung, K.G.; Xiang, B.; Tamanna, N.; Teixeira, A.L.F.; Agarwal, P.; Kereliuk, S.M.; Spicer, V.; Lin, L.; Treberg, J.; et al. Mitochondrial Sirtuin-3 (SIRT3) Prevents Doxorubicin-Induced Dilated Cardiomyopathy by Modulating Protein Acetylation and Oxidative Stress. Circ. Heart Fail. 2022, 15, e008547. [Google Scholar] [CrossRef]
  87. Zhang, J.; Li, W.; Xue, S.; Gao, P.; Wang, H.; Chen, H.; Hong, Y.; Sun, Q.; Lu, L.; Wang, Y.; et al. Qishen granule attenuates doxorubicin-induced cardiotoxicity by protecting mitochondrial function and reducing oxidative stress through regulation of Sirtuin3. J. Ethnopharmacol. 2024, 319, 117134. [Google Scholar] [CrossRef]
  88. Zhong, Z.; Gao, Y.; Zhou, J.; Wang, F.; Zhang, P.; Hu, S.; Wu, H.; Lou, H.; Chi, J.; Lin, H.; et al. Inhibiting mir-34a-5p regulates doxorubicin-induced autophagy disorder and alleviates myocardial pyroptosis by targeting Sirt3-AMPK pathway. Biomed. Pharmacother. 2023, 168, 115654. [Google Scholar] [CrossRef]
  89. Li, Y.; Zhou, Y.; Wang, F.; Chen, X.; Wang, C.; Wang, J.; Liu, T.; Li, Y.; He, B. SIRT4 is the last puzzle of mitochondrial sirtuins. Bioorg. Med. Chem. 2018, 26, 3861–3865. [Google Scholar] [CrossRef]
  90. Lang, A.; Piekorz, R.P. Novel role of the SIRT4-OPA1 axis in mitochondrial quality control. Cell Stress 2017, 2, 1–3. [Google Scholar] [CrossRef]
  91. Lang, A.; Anand, R.; Altinoluk-Hambüchen, S.; Ezzahoini, H.; Stefanski, A.; Iram, A.; Bergmann, L.; Urbach, J.; Böhler, P.; Hänsel, J.; et al. SIRT4 interacts with OPA1 and regulates mitochondrial quality control and mitophagy. Aging 2017, 9, 2163–2189. [Google Scholar] [CrossRef] [PubMed]
  92. He, L.; Wang, J.; Yang, Y.; Zou, P.; Xia, Z.; Li, J. SIRT4 Suppresses Doxorubicin-Induced Cardiotoxicity by Regulating the AKT/mTOR/Autophagy Pathway. Toxicology 2022, 469, 153119. [Google Scholar] [CrossRef] [PubMed]
  93. Pei, Z.; Ma, L.; Li, Y.; Yang, J.; Yang, Q.; Yao, W.; Li, S. CoQ10 Improves Myocardial Damage in Doxorubicin-Induced Heart Failure in C57BL/6 Mice. Front. Biosci. Landmark Ed. 2022, 27, 244. [Google Scholar] [CrossRef]
  94. Li, X.; Liu, L.; Li, T.; Liu, M.; Wang, Y.; Ma, H.; Mu, N.; Wang, H. SIRT6 in Senescence and Aging-Related Cardiovascular Diseases. Front. Cell Dev. Biol. 2021, 9, 641315. [Google Scholar] [CrossRef]
  95. Han, D.; Wang, Y.; Wang, Y.; Dai, X.; Zhou, T.; Chen, J.; Tao, B.; Zhang, J.; Cao, F. The Tumor-Suppressive Human Circular RNA CircITCH Sponges miR-330-5p to Ameliorate Doxorubicin-Induced Cardiotoxicity Through Upregulating SIRT6, Survivin, and SERCA2a. Circ. Res. 2020, 127, e108–e125. [Google Scholar] [CrossRef] [PubMed]
  96. Peng, K.; Zeng, C.; Gao, Y.; Liu, B.; Li, L.; Xu, K.; Yin, Y.; Qiu, Y.; Zhang, M.; Ma, F.; et al. Overexpressed SIRT6 ameliorates doxorubicin-induced cardiotoxicity and potentiates the therapeutic efficacy through metabolic remodeling. Acta Pharm. Sin. B 2023, 13, 2680–2700. [Google Scholar] [CrossRef]
  97. Wang, S.; Zhang, X.; Hou, Y.; Zhang, Y.; Chen, J.; Gao, S.; Duan, H.; Gu, S.; Yu, S.; Cai, Y. SIRT6 activates PPARα to improve doxorubicin-induced myocardial cell aging and damage. Chem. Biol. Interact. 2024, 392, 110920. [Google Scholar] [CrossRef]
  98. Lagunas-Rangel, F.A. SIRT7 in the aging process. Cell. Mol. Life Sci. 2022, 79, 297. [Google Scholar] [CrossRef]
  99. Vakhrusheva, O.; Smolka, C.; Gajawada, P.; Kostin, S.; Boettger, T.; Kubin, T.; Braun, T.; Bober, E. Sirt7 increases stress resistance of cardiomyocytes and prevents apoptosis and inflammatory cardiomyopathy in mice. Circ. Res. 2008, 102, 703–710. [Google Scholar] [CrossRef]
  100. Araki, S.; Izumiya, Y.; Rokutanda, T.; Ianni, A.; Hanatani, S.; Kimura, Y.; Onoue, Y.; Senokuchi, T.; Yoshizawa, T.; Yasuda, O.; et al. Sirt7 Contributes to Myocardial Tissue Repair by Maintaining Transforming Growth Factor-β Signaling Pathway. Circulation 2015, 132, 1081–1093. [Google Scholar] [CrossRef]
  101. Yamamura, S.; Izumiya, Y.; Araki, S.; Nakamura, T.; Kimura, Y.; Hanatani, S.; Yamada, T.; Ishida, T.; Yamamoto, M.; Onoue, Y.; et al. Cardiomyocyte Sirt (Sirtuin) 7 Ameliorates Stress-Induced Cardiac Hypertrophy by Interacting with and Deacetylating GATA4. Hypertens. Dallas Tex 1979 2020, 75, 98–108. [Google Scholar] [CrossRef]
  102. Sun, M.; Zhai, M.; Zhang, N.; Wang, R.; Liang, H.; Han, Q.; Jia, Y.; Jiao, L. MicroRNA-148b-3p is involved in regulating hypoxia/reoxygenation-induced injury of cardiomyocytes in vitro through modulating SIRT7/p53 signaling. Chem. Biol. Interact. 2018, 296, 211–219. [Google Scholar] [CrossRef]
  103. Ghosh, S.; Liu, B.; Zhou, Z. Resveratrol activates SIRT1 in a Lamin A-dependent manner. Cell Cycle Georget. Tex 2013, 12, 872–876. [Google Scholar] [CrossRef] [PubMed]
  104. Cao, D.; Wang, M.; Qiu, X.; Liu, D.; Jiang, H.; Yang, N.; Xu, R.-M. Structural basis for allosteric, substrate-dependent stimulation of SIRT1 activity by resveratrol. Genes Dev. 2015, 29, 1316–1325. [Google Scholar] [CrossRef]
  105. Chang, C.; Meikle, T.G.; Drummond, C.J.; Yang, Y.; Conn, C.E. Comparison of cubosomes and liposomes for the encapsulation and delivery of curcumin. Soft Matter 2021, 17, 3306–3313. [Google Scholar] [CrossRef]
  106. Luo, G.; Jian, Z.; Zhu, Y.; Zhu, Y.; Chen, B.; Ma, R.; Tang, F.; Xiao, Y. Sirt1 promotes autophagy and inhibits apoptosis to protect cardiomyocytes from hypoxic stress. Int. J. Mol. Med. 2019, 43, 2033–2043. [Google Scholar] [CrossRef]
  107. Kong, C.-Y.; Guo, Z.; Song, P.; Zhang, X.; Yuan, Y.-P.; Teng, T.; Yan, L.; Tang, Q.-Z. Underlying the Mechanisms of Doxorubicin-Induced Acute Cardiotoxicity: Oxidative Stress and Cell Death. Int. J. Biol. Sci. 2022, 18, 760–770. [Google Scholar] [CrossRef] [PubMed]
  108. Wu, B.; You, S.; Qian, H.; Wu, S.; Lu, S.; Zhang, Y.; Sun, Y.; Zhang, N. The role of SIRT2 in vascular-related and heart-related diseases: A review. J. Cell. Mol. Med. 2021, 25, 6470–6478. [Google Scholar] [CrossRef]
  109. Chen, L.; Sun, X.; Wang, Z.; Chen, M.; He, Y.; Zhang, H.; Han, D.; Zheng, L. Resveratrol protects against doxorubicin-induced cardiotoxicity by attenuating ferroptosis through modulating the MAPK signaling pathway. Toxicol. Appl. Pharmacol. 2024, 482, 116794. [Google Scholar] [CrossRef]
  110. Zhang, Y.; Wang, L.; Meng, L.; Cao, G.; Wu, Y. Sirtuin 6 overexpression relieves sepsis-induced acute kidney injury by promoting autophagy. Cell Cycle Georget. Tex 2019, 18, 425–436. [Google Scholar] [CrossRef]
  111. Magrone, T.; Jirillo, E. Immunity to Tuberculosis and Novel Therapeutic Strategies. Clin. Immunol. Endocr. Metab. Drugs 2014, 1, 46–60. [Google Scholar] [CrossRef]
  112. Arrigoni, R.; Ballini, A.; Jirillo, E.; Santacroce, L. Current View on Major Natural Compounds Endowed with Antibacterial and Antiviral Effects. Antibiotics 2024, 13, 603. [Google Scholar] [CrossRef]
  113. Jiang, K.; Bai, Y.; Hou, R.; Chen, G.; Liu, L.; Ciftci, O.N.; Farag, M.A.; Liu, L. Advances in dietary polyphenols: Regulation of inflammatory bowel disease (IBD) via bile acid metabolism and the gut-brain axis. Food Chem. 2025, 472, 142932. [Google Scholar] [CrossRef]
  114. Arrigoni, R.; Cammarota, F.; Porro, R.; Cantore, S.; Dioguardi, M.; Cazzolla, A.P.; De Leonardis, F.; Polimeno, L.; Zerman, N.; Di Cosola, M.; et al. Natural Bioactive Compounds against Oxidative Stress: Dietary Polyphenols Strike Back. Endocr. Metab. Immune Disord. Drug Targets 2023, 23, 764–776. [Google Scholar] [CrossRef]
  115. Li, G.; Liu, Q.; Gao, T.; Li, J.; Zhang, J.; Chen, O.; Cao, C.; Mao, M.; Xiao, M.; Zhang, X.; et al. Resveratrol and FGF1 Synergistically Ameliorates Doxorubicin-Induced Cardiotoxicity via Activation of SIRT1-NRF2 Pathway. Nutrients 2022, 14, 4017. [Google Scholar] [CrossRef]
  116. Syahputra, R.A.; Harahap, U.; Dalimunthe, A.; Nasution, M.P.; Satria, D. The Role of Flavonoids as a Cardioprotective Strategy against Doxorubicin-Induced Cardiotoxicity: A Review. Molecules 2022, 27, 1320. [Google Scholar] [CrossRef] [PubMed]
  117. Yu, W.; Sun, H.; Zha, W.; Cui, W.; Xu, L.; Min, Q.; Wu, J. Apigenin Attenuates Adriamycin-Induced Cardiomyocyte Apoptosis via the PI3K/AKT/mTOR Pathway. Evid.-Based Complement. Altern. Med. ECAM 2017, 2017, 2590676. [Google Scholar] [CrossRef]
  118. Ma, Y.; Yang, L.; Ma, J.; Lu, L.; Wang, X.; Ren, J.; Yang, J. Rutin attenuates doxorubicin-induced cardiotoxicity via regulating autophagy and apoptosis. Biochim. Biophys. Acta BBA Mol. Basis Dis. 2017, 1863, 1904–1911. [Google Scholar] [CrossRef]
  119. Priya, L.B.; Baskaran, R.; Huang, C.-Y.; Padma, V.V. Neferine ameliorates cardiomyoblast apoptosis induced by doxorubicin: Possible role in modulating NADPH oxidase/ROS-mediated NFκB redox signaling cascade. Sci. Rep. 2017, 7, 12283. [Google Scholar] [CrossRef]
  120. Lin, X.; Wang, Q.; Sun, S.; Xu, G.; Wu, Q.; Qi, M.; Bai, F.; Yu, J. Astragaloside IV promotes the eNOS/NO/cGMP pathway and improves left ventricular diastolic function in rats with metabolic syndrome. J. Int. Med. Res. 2020, 48, 300060519826848. [Google Scholar] [CrossRef]
  121. Afsar, T.; Razak, S.; Batoo, K.M.; Khan, M.R. Acacia hydaspica R. Parker prevents doxorubicin-induced cardiac injury by attenuation of oxidative stress and structural Cardiomyocyte alterations in rats. BMC Complement. Altern. Med. 2017, 17, 554. [Google Scholar] [CrossRef]
  122. Wang, M.; Zhang, J.; Zhao, M.; Liu, J.; Ye, J.; Xu, Y.; Wang, Z.; Ye, D.; Li, D.; Wan, J. Resolvin D1 Attenuates Doxorubicin-Induced Cardiotoxicity by Inhibiting Inflammation, Oxidative and Endoplasmic Reticulum Stress. Front. Pharmacol. 2021, 12, 749899. [Google Scholar] [CrossRef]
  123. Wan, Y.; Wang, J.; Xu, J.-F.; Tang, F.; Chen, L.; Tan, Y.-Z.; Rao, C.-L.; Ao, H.; Peng, C. Panax ginseng and its ginsenosides: Potential candidates for the prevention and treatment of chemotherapy-induced side effects. J. Ginseng Res. 2021, 45, 617–630. [Google Scholar] [CrossRef] [PubMed]
  124. Li, X.-R.; Cheng, X.-H.; Zhang, G.-N.; Wang, X.-X.; Huang, J.-M. Cardiac safety analysis of first-line chemotherapy drug pegylated liposomal doxorubicin in ovarian cancer. J. Ovarian Res. 2022, 15, 96. [Google Scholar] [CrossRef]
  125. Xing, M.; Yan, F.; Yu, S.; Shen, P. Efficacy and Cardiotoxicity of Liposomal Doxorubicin-Based Chemotherapy in Advanced Breast Cancer: A Meta-Analysis of Ten Randomized Controlled Trials. PLoS ONE 2015, 10, e0133569. [Google Scholar] [CrossRef]
  126. Norouzi, M.; Yathindranath, V.; Thliveris, J.A.; Kopec, B.M.; Siahaan, T.J.; Miller, D.W. Doxorubicin-loaded iron oxide nanoparticles for glioblastoma therapy: A combinational approach for enhanced delivery of nanoparticles. Sci. Rep. 2020, 10, 11292. [Google Scholar] [CrossRef]
  127. Kourek, C.; Touloupaki, M.; Rempakos, A.; Loritis, K.; Tsougkos, E.; Paraskevaidis, I.; Briasoulis, A. Cardioprotective Strategies from Cardiotoxicity in Cancer Patients: A Comprehensive Review. J. Cardiovasc. Dev. Dis. 2022, 9, 259. [Google Scholar] [CrossRef] [PubMed]
  128. McGowan, J.V.; Chung, R.; Maulik, A.; Piotrowska, I.; Walker, J.M.; Yellon, D.M. Anthracycline Chemotherapy and Cardiotoxicity. Cardiovasc. Drugs Ther. 2017, 31, 63–75. [Google Scholar] [CrossRef]
  129. Liesse, K.; Harris, J.; Chan, M.; Schmidt, M.L.; Chiu, B. Dexrazoxane Significantly Reduces Anthracycline-induced Cardiotoxicity in Pediatric Solid Tumor Patients: A Systematic Review. J. Pediatr. Hematol. Oncol. 2018, 40, 417–425. [Google Scholar] [CrossRef]
  130. van Dalen, E.C.; Caron, H.N.; Dickinson, H.O.; Kremer, L.C. Cardioprotective interventions for cancer patients receiving anthracyclines. Cochrane Database Syst. Rev. 2011, 2011, CD003917. [Google Scholar] [CrossRef]
  131. Tebbi, C.K.; London, W.B.; Friedman, D.; Villaluna, D.; De Alarcon, P.A.; Constine, L.S.; Mendenhall, N.P.; Sposto, R.; Chauvenet, A.; Schwartz, C.L. Dexrazoxane-associated risk for acute myeloid leukemia/myelodysplastic syndrome and other secondary malignancies in pediatric Hodgkin’s disease. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2007, 25, 493–500. [Google Scholar] [CrossRef] [PubMed]
  132. Oxnard, G.R.; Yang, J.C.-H.; Yu, H.; Kim, S.-W.; Saka, H.; Horn, L.; Goto, K.; Ohe, Y.; Mann, H.; Thress, K.S.; et al. TATTON: A multi-arm, phase Ib trial of osimertinib combined with selumetinib, savolitinib, or durvalumab in EGFR-mutant lung cancer. Ann. Oncol. 2020, 31, 507–516. [Google Scholar] [CrossRef] [PubMed]
  133. Beheshti, T.; Toroghi, M.; Hosseini, G.; Zarifian, A.; Shandiz, H.; Fazlinezhad, A. Carvedilol Administration Can Prevent Doxorubicin-Induced Cardiotoxicity: A Double-Blind Randomized Trial. Cardiology 2016, 134, 47–53. [Google Scholar] [CrossRef] [PubMed]
  134. Neilan, T.G.; Quinaglia, T.; Onoue, T.; Mahmood, S.S.; Drobni, Z.D.; Gilman, H.K.; Smith, A.; Heemelaar, J.C.; Brahmbhatt, P.; Ho, J.S.; et al. Atorvastatin for Anthracycline-Associated Cardiac Dysfunction: The STOP-CA Randomized Clinical Trial. JAMA 2023, 330, 528–536. [Google Scholar] [CrossRef]
  135. Makhlin, I.; Demissei, B.G.; D’Agostino, R.; Hundley, W.G.; Baleanu-Gogonea, C.; Wilcox, N.S.; Chen, A.; Smith, A.M.; O’Connell, N.S.; Januzzi, J.L.; et al. Statins Do Not Significantly Affect Oxidative Nitrosative Stress Biomarkers in the PREVENT Randomized Clinical Trial. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2024, 30, 2370–2376. [Google Scholar] [CrossRef]
  136. Gupta, S.K.; Garg, A.; Bär, C.; Chatterjee, S.; Foinquinos, A.; Milting, H.; Streckfuß-Bömeke, K.; Fiedler, J.; Thum, T. Quaking Inhibits Doxorubicin-Mediated Cardiotoxicity Through Regulation of Cardiac Circular RNA Expression. Circ. Res. 2018, 122, 246–254. [Google Scholar] [CrossRef]
  137. Li, K.; Jiang, K.-M.; Wang, Y.; Hu, F.; Zhu, X.-C.; Sun, C.-L.; Jin, L.; Liu, W.-T.; Lin, T.-T.; Li, M. Inhibition of NETs prevents doxorubicin-induced cardiotoxicity by attenuating IL-18-IFN-γ-Cx43 axis induced cardiac conduction abnormalities. Int. Immunopharmacol. 2025, 147, 114016. [Google Scholar] [CrossRef]
  138. Todorova, V.K.; Hsu, P.-C.; Wei, J.Y.; Lopez-Candales, A.; Chen, J.Z.; Su, L.J.; Makhoul, I. Biomarkers of inflammation, hypercoagulability and endothelial injury predict early asymptomatic doxorubicin-induced cardiotoxicity in breast cancer patients. Am. J. Cancer Res. 2020, 10, 2933–2945. [Google Scholar]
Figure 1. Doxorubicin’s chemical structure.
Figure 1. Doxorubicin’s chemical structure.
Toxics 13 00277 g001
Table 1. Pathogenesis of DIC.
Table 1. Pathogenesis of DIC.
Oxidative StressInteraction with mitochondrial DNA, and inhibition of the respiratory chain [31];
Generation of ROS, alteration of the phospholipids of cell membranes, mitochondria, and ER, with myocardial damage [32];
Reduction of glutathione and CAT levels, with increase in oxidative stress [33];
DIC-mediated endothelial dysfunction with increased levels of endothelin-1, activation of type A and type B receptors, vasoconstriction, and release of NO, adrenomedullin, and prostacyclin [37]
AutophagyDOX-mediated increase in Beclin-1, p62, and microtubule-associated protein 1A/1B light chain 3 (LC3)-II/LC3-I levels, with damaged mitochondria removed by mitophagy [43];
DOX-mediated abrogation of lysosome biosynthesis and catepsin activity, with autophagolysosomal accumulation, and impairment of autophagy [45];
DOX-induced inhibition of the fusion between autophagosomes and liposomes [46];
DOX-mediated enhancement of TLR9, and suppression of AMPK activation [47]
InflammationDIC-induced release of pro-inflammatory cytokines through activation of TLR-4, NF-kB, and NLRP3 inflammasome [48,49,50];
DOX-mediated transformation of smooth cardiac muscle cells into macrophage-like cells, with vascular wall low-grade inflammation and impairment of sarcoplasmic reticulum [55,56];
DOX-induced apoptosis of cardiomyocytes via release of catecholamines by infiltrating macrophages [57]
Apoptosis/FerroptosisDecrease in the anti-apoptotic protein, Bcl-2, and increase in Bcl-2-associated X expression in DOX-treated cardiomyocytes, with increased mitochondrial permeability, cytochrome c release and caspase-3, thus leading to cardiomyocyte apoptosis [60];
DOX-induced ferroptosis of cardiomyocytes by inactivation of GPx4 [62,63,64];
Overexpression of methyltransferase-like 14 with upregulation of transferrin receptors, and uptake of iron [65];
DOX-mediated ferroptosis by impairment of Forkhead Box O4 transcription, and high mobility group Box 1 nuclear translocation [66]
Table 2. Beneficial effects of sirtuins on DIC.
Table 2. Beneficial effects of sirtuins on DIC.
SIRT 1ROS generation and FOXO 1 deacetylation inhibition, with decrease in cardiac oxidative stress [71]
SIRT 1-mediated inhibition of NF-kB through deacetylation of the peroxisome proliferator-activated receptor gamma coactivator 1 alpha; SIRT 1/Liver kinase B1/AMPK pathway activation; inhibition of miR-200a 3p, respectively [72,75,79]
Natural product (jaceosidin, calycosin, and dihydromyrecitin)-mediated activation of SIRT 1, with inhibition of NF-kB [76,77,94];
p53 protein acetylation decrease by resveratrol-activated SIRT 1, with attenuation of myocardial
cell apoptosis [78];
SIRT 1-mediated activation of the Nrf2/Kelch-like associated protein, with reduction of ferroptosis,
and DIC [79]
SIRT 2FOXO3a and AMPK SIRT 2-mediated activation, respectively, with reduced release of ROS and mitigation of DIC [81,108]
SIRT 2 activation and improvement of DOX-mediated cardiac aging [65];
miR-140-5p inhibition with activation of the SIRT2/NRF2 antioxidant pathway [82]
SIRT 3Natural product (RES, daidzein, tubeimoside, berberine, Qishen granules) and dichloroacetic acid-mediated upregulation of SIRT3, with improvement of DOX-induced mitochondrial dysfunction, ROS generation, and apoptosis [70,84,85,86,87,109]
miR-34-5p SIRT 3-mediated inhibition, with autophagic activity regulation, and protection from DIC [88]
SIRT 3-induced inhibition of NLRP3 inflammasome, with autophagy regulation, and pyroptosis decrease [83]
SIRT 4SIRT 4-mediated inhibition of fatty acid oxidation in muscles, with reduction of mitochondrial function [89]
Overexpression of SIRT 4 and interaction with optic atrophin 1, with regulation of autophagy and ROS generation;
SIRT 4-mediated inhibition of DIC, with activation of the akt/mTOR pathway [92]
SIRT 5Coenzyme Q 10-mediated overexpression of SIRT 5 and protection from DIC [93]
SIRT 6SIRT 6 overexpression by targeting miR-330-5p, with inhibition of ROS generation, apoptosis, and
necrosis during DIC [80,95]
Enhancement of autophagy by SIRT 6-mediated acetylation and inhibition of SKG1 [110]
SIRT 7SIRT 7-induced reduction of myocardial stress via deacetylation of p53 and GATA4; regulation of
autophagy and inhibition of miR-148-3p, respectively [83,98,100,101,102]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Arrigoni, R.; Jirillo, E.; Caiati, C. Pathophysiology of Doxorubicin-Mediated Cardiotoxicity. Toxics 2025, 13, 277. https://doi.org/10.3390/toxics13040277

AMA Style

Arrigoni R, Jirillo E, Caiati C. Pathophysiology of Doxorubicin-Mediated Cardiotoxicity. Toxics. 2025; 13(4):277. https://doi.org/10.3390/toxics13040277

Chicago/Turabian Style

Arrigoni, Roberto, Emilio Jirillo, and Carlo Caiati. 2025. "Pathophysiology of Doxorubicin-Mediated Cardiotoxicity" Toxics 13, no. 4: 277. https://doi.org/10.3390/toxics13040277

APA Style

Arrigoni, R., Jirillo, E., & Caiati, C. (2025). Pathophysiology of Doxorubicin-Mediated Cardiotoxicity. Toxics, 13(4), 277. https://doi.org/10.3390/toxics13040277

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop