Skip to main content
Erschienen in: Heart Failure Reviews 4/2021

Open Access 15.12.2020

Oxidative stress and inflammation: determinants of anthracycline cardiotoxicity and possible therapeutic targets

verfasst von: Iacopo Fabiani, Alberto Aimo, Chrysanthos Grigoratos, Vincenzo Castiglione, Francesco Gentile, Luigi F Saccaro, Chiara Arzilli, Daniela Cardinale, Claudio Passino, Michele Emdin

Erschienen in: Heart Failure Reviews | Ausgabe 4/2021

Abstract

Chemotherapy with anthracycline-based regimens remains a cornerstone of treatment of many solid and blood tumors but is associated with a significant risk of cardiotoxicity, which can manifest as asymptomatic left ventricular dysfunction or overt heart failure. These effects are typically dose-dependent and cumulative and may require appropriate screening strategies and cardioprotective therapies in order to minimize changes to anticancer regimens or even their discontinuation. Our current understanding of cardiac damage by anthracyclines includes a central role of oxidative stress and inflammation. The identification of these processes through circulating biomarkers or imaging techniques might then be helpful for early diagnosis and risk stratification. Furthermore, therapeutic strategies relieving oxidative stress and inflammation hold promise to prevent heart failure development or at least to mitigate cardiac damage, although further evidence is needed on their efficacy, either alone or as part of combination therapies with neurohormonal antagonists, which are the current adopted standard.
Hinweise
Iacopo Fabiani and Alberto Aimo equally contributed to this work.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Cardiotoxicity manifesting as asymptomatic left ventricular (LV) dysfunction or overt heart failure (HF) is a major adverse effect of chemotherapy for cancer [1, 2]. In a recent analysis from the CARDIOTOX Registry, which prospectively evaluated 865 patients undergoing anticancer therapy associated with a moderate to high risk of cardiotoxicity, as many as 38% of patients displayed overt cardiotoxicity over a 24-month follow-up, with severe cardiac disease occurring in 3% [3]. These last patients had a mortality rate of 23 deaths per 100 patients-year.
Introduced in the 1960s, anthracyclines (ANT) are among the most potent and prescribed chemotherapeutics for the treatment of haematological and solid tumors. The most used drug of this class is doxorubicin (DOX), also known as adriamycin, with several other molecules used, including daunorubicin, epirubicin, and valrubicin. LV dysfunction and HF are well-established, typically dose-dependent, and cumulative adverse effects of ANT, and may require changes to anticancer regimens or even their discontinuation[1]. HF has a 5% incidence in patients receiving a cumulative DOX dose of 400 mg/m2, increasing to 48% with a cumulative dose of 700 mg/m2, but even low doses (such as 100 mg/m2) may cause cardiac dysfunction [4]. In addition to the cumulative dose, major risk factors include the administration schedule, other cardiotoxic therapies (such as trastuzumab or paclitaxel), mediastinal irradiation, and history of heart disease [1]. Cardiotoxic reactions to ANT may be acute (i.e., occurring during drug administration), subacute, or chronic. However, recent findings challenge this old classification, suggesting that anthracycline-induced cardiotoxicity is potentially a continuous phenomenon, starting with myocardial cell injury, followed by progressive functional decline, progressively leading to overt HF [5, 6]. Patient susceptibility to ANT is highly variable, with many patients tolerating therapeutic dose of ANT without long-term complications, while others showing ANT-dependent cardiotoxicity even after the first dose. Some patients may develop cardiotoxicity at a total cumulative dose of ANT corresponding to 300 mg/m2, or even lower, while others have no significant cardiac alterations, despite being exposed to doses up to 1000 mg/m2 [4].
Therapies for neurohormonal antagonism are currently recommended for patients with a normal LV ejection fraction (LVEF) and cardiovascular risk factors scheduled to undergo therapy with cardiotoxic agents, although this evidence derives from a limited number of studies [7, 8].
The development of cardiotoxicity by ANT may be partially explained by the induction of oxidative stress and inflammatory responses [4]. A deeper knowledge on these processes might disclose new possibilities for early detection of cardiac damage (through circulating biomarkers or imaging findings related to inflammation) and offer new therapeutic strategies, complementary to the use of neurohormonal antagonists (Fig. 1).
For the present review, we performed a search on PubMed and EMBASE in May 2020 with the following terms: “anthracycline” OR “doxorubicin” AND “cardiotoxicity” OR “cardiac damage” OR “heart failure” OR “cardiac failure.” The reference list of relevant articles was also searched; only articles published in English were included. Given the design of this work as a narrative review, no formal criteria for study selection or appraisal were enforced.

Oxidative stress and inflammation as possible contributors to cardiac damage

ANT cardiotoxicity is believed to derive from DNA damage, inhibition of protein synthesis and mitochondrial biogenesis, induction of apoptosis, inflammation and generation of reactive oxygen species (ROS). The myocardium has a high mitochondrial density [9], and the propensity of ANT to accumulate in the mitochondria promotes their retention in cardiomyocytes. Here, ANT metabolites interfere with the translation of iron sequestrating proteins, thus increasing intracellular free iron, which leads to the generation of highly reactive iron-ANT complexes, capable of starting redox cycling and promoting excessive autophagy [1012]. ANT may also easily intercalate into mitochondrial DNA, because of its circular and covalently closed structure, further promoting mitochondrial respiratory chain dysfunction [1315]. Finally, ANT may directly activate ROS-producing enzymes. Notably, ROS generation is usually associated with the production of equally dangerous reactive nitrogen species (RNS). Indeed, ANT have been found to increase the activity of inducible nitric oxide synthase (NOS), which can foster RNS generation [16, 17]. Once antioxidant defences have been overwhelmed, ROS may react with lipids, proteins, and DNA, leading to the disruption of mitochondrial integrity and function that may ultimately cause cell death [18]. ROS-dependent cellular damages may induce inflammation, which is a crucial mechanism in HF development. Interestingly, phenylalanine-butyramide (a synthetic derivative of the short-chain fatty acid butyric acid) has been recently shown to reduce oxidative stress and mithocondrial dysfunction in a murine model of ANT cardiotoxicity [19].
ANT therapy is also associated with induction of nuclear factor-κB (NFkB) and tumor necrosis factor alpha (TNF-α). Furthermore, several Toll-like receptors (TLRs), a class of pattern recognition receptors involved in innate immune response initiation and induction of cytokines secretion, are activated and promote the activation of adaptive immunity cells (for instance, TLR4 activates T helper-1 cells) [4, 20]. Furthermore, through transcription factor modulation, ANT increase NLRP3 expression, thus stimulating the release of the proinflammatory interleukins (IL)-1β and IL-6 [21]. Additionally, ANT-dependent inhibition of cyclooxygenase and lipoxygenases reduces cardiac availability of anti-inflammatory mediators (such as prostacyclins), promoting cardiac damage [22, 23].
Cardiomyocyte damage and death further amplify the inflammatory cascade and oxidative stress described above, involving cytokines, cardiomyocytes, immune, and endothelial cells. This vicious cycle induces the functional and structural alterations (cardiac hypertrophy, fibrosis, electrical abnormalities) observed in HF.

Biomarkers of oxidative stress and inflammation

Serum biomarkers have been studied as tools for risk stratification before ANT therapy, early detection of subclinical cardiac damage during treatment, or identification of late effects. The majority of studies have focused on cardiac troponins and natriuretic peptides (NPs), which have been recommended as tools for therapy monitoring [1].
Evidence on biomarkers of oxidative stress and inflammation is sparse and mostly derives from single-center studies. The increase in serum ROS (determined by measuring the radical species produced by a specific reaction, which are directly proportional to the quantity of lipid peroxides) and the decrease in glutathione peroxidase displayed a correlation with the reduction in strain rate peak, and serum ROS levels were the only independent predictive variable of systolic dysfunction at multiple regression analysis [24]. In a phase II trial on 49 cancer patients receiving epirubicin and either telmistartan or placebo, telmisartan was able prevent ANT-induced increase in serum IL-6 and ROS levels [25]. Total antioxidant status (TAOS) represents the sum of all measurable antioxidant within plasma and body fluids. In a study on 29 acute leukemia children on ANT, TAOS levels decreased in parallel with the cumulative ANT dose [26].
C-reactive protein (CRP) is a nonspecific marker of inflammation produced by the hepatocytes in response to IL-6 released from macrophages and T cells. Several studies reported a significant elevation of CRP [27] and high-sensitivity CRP (hs-CRP) [2831] following ANT-based chemotherapy, but only one study demonstrated a modest association between hs-CRP increase and echocardiographic evidence of cardiotoxicity [31], and therapies counteracting ANT-related cardiotoxicity do not seem to alter hs-CRP levels [32].
Myeloperoxidase (MPO) is a proinflammatory enzyme expressed by neutrophils and involved in ROS production and lipid peroxidation. In one study on 78 cancer patients receiving doxorubicin and trastuzumab, a transient increase in MPO levels following chemotherapy predicted subsequent development of LVEF decline; among several biomarkers, the combination MPO and troponin I identified a subgroup of patients at higher risk of cardiotoxicity [30]. A following study from the same group confirmed the predictive value of MPO for ANT cardiotoxicity [31].
Inflammatory mediators TNFα and ILs have not emerged as robust biomarkers of ANT cardiotoxicity. Serum TNF and soluble TNF receptors I and II levels do not seem to change in response to ANT-based chemotherapy [24, 33, 34]. Similar results have been reported for IL-1β [24]. Conversely, IL-6 levels increased significantly following epirubicin administration and correlated with reduction of peak strain rate at echocardiography, a marker of early systolic dysfunction [24].
Sawaya et al. [35] and Armenian et al. [36] evaluated the predictive role of soluble suppression of tumorigenesis 2 (sST2), a biomarker released mainly by extracardiac tissues in response to inflammatory and fibrotic stimuli, for early and late cardiotoxicity in cancer survivors treated with ANT-based regimens. Both studies showed that sST2 levels were slightly above normal range at baseline, but did not change significantly during the follow-up and did not correlate with the evolution of echocardiographic parameters. A recent study reported a significant increase in sST2 following ANT administration to breast cancer patients, although no difference in sST2 were observed between patients who developed a decline in LVEF and those remaining clinically stable [37].
Multiomics approaches have tried to trace the molecular fingerprint of ANT cardiotoxicity in order to identify novel biomarkers able to predict this complication. For example, a proteomic profiling approach showed that higher baseline immunoglobulin E (IgE) levels were associated with a lower risk of chemotherapy-induced cardiotoxicity, possibly because of a lower degree of activation of the Th1 response [38].
Several imaging techniques allow to assess the presence and extent of inflammation and to monitor its evolution over time (Table 1). In the acute phase, imaging findings may vary from a subclinical injury with preserved LVEF [5] up to extensive involvement and cardiogenic shock [39]. Echocardiography represents the first-line imaging technique, but subtle systolic abnormalities may not be recognized during the early phases of the disease. In the setting of ANT cardiotoxicity, strain echocardiography may not only detect subtle alterations but also predict subsequent myocardial dysfunction; therefore, its use in everyday clinical practice is recommended by current guidelines [40].
Table 1
Imaging techniques to assess myocardial inflammation and its consequences in patients receiving anthracyclines (ANTs)
Acute/chronic phase, roles of imaging
Imaging technique
Main findings in ANT cardiotoxicity
Acute phase:
- Early detection of cardiac damage
- Prognostic stratification
(2D/3D) TTE
LV/RV regional/global systolic dysfunction; diastolic dysfunction
(2D/3D) strain TTE
Subclinical LV/RV systolic dysfunction
CMR
LV/RV regional/global systolic dysfunction; LV/RV hyperintensity in T2w sequences; LGE; increase in regional/global T1 and T2 mapping values
SPECT
LVEF decrease
18F-FDG PET
Increased glucose uptake
Chronic phase:
- Detection of cardiac damage
- Follow-up of known cardiomyopathy
TTE
LV/RV regional/global systolic dysfunction; diastolic dysfunction
CMR
LV/RV regional/global systolic dysfunction; LGE
SPECT
LVEF decrease
2D/3D two-/three-dimensional, 18F-FDG PET, 18F-fluoro-D-glucose positron emission tomography, CMR cardiac magnetic resonance, LGE late gadolinium enhancement, LV/RV left/right ventricle, TTE transthoracic echocardiography
The main limitation of echocardiography is that it can assess myocardial damage only through its impact on systolic function. Conversely, cardiac magnetic resonance (CMR) allows not only to reproducibly estimate biventricular volumes and systolic function but also to assess the presence, extent, and pattern of distribution of myocardial edema, fat, and fibrosis [4143]. T2-weighted fast-spin-echo sequence is a robust technique in inflammation imaging, allowing to noninvasively detect with a millimetric spatial resolution the presence of areas of myocardial inflammation in various inflammatory cardiomyopathies including early-phase ANT cardiotoxicity [28]. More recently, novel parametric mapping techniques such as T1 and T2 mapping have shown their potential in the early identification of ANT cardiotoxicity in both preclinical [44] and clinical settings [45].
Single-photon emission computed tomography (SPECT) has been used to assess LV geometry and function in patients receiving ANT [46] but is performed very rarely because of the radiation exposure of patients requiring follow-up examinations, and the low agreement between SPECT and CMR estimates of LVEF [47]. 18F-Fluoro-D-glucose positron emission tomography (18F-FDG PET) has the unique ability to characterize myocardial metabolism [48], which may be helpful for early detection of cardiotoxicity as increased glucose uptake [49]. The role of 18F-FDG PET is currently limited to the research setting.
Following the acute phase, serial CMR examinations may document the evolution of myocardial inflammation and its sequelae. Myocardial scar can be detected as late gadolinium enhancement (LGE) in post-contrast T1 gradient echo sequences. Various LGE patterns and estimates of LGE prevalence have been reported in ANT-related cardiotoxicity [50, 51], likely because of the small study size, and their heterogeneous enrolment criteria and treatment regimens. Both reparative fibrosis and interstitial fibrosis have been described. In children with subclinical ANT cardiotoxicity, T1 mapping showed an expansion of extracellular spaces, correlating with cumulative ANT dose and exercise capacity [52]. In a cohort of adult cancer survivors, myocardial T1 elevation occurred independently of the underlying neoplasm or cardiac comorbidities, and could then be attributed to the cardiotoxic effect of chemotherapy [53].

Therapeutic approaches

Several treatments targeting oxidative stress or inflammation have been evaluated as possible approaches to ANT-related cardiotoxicity (Table 2). On the other hand, the identification of a less cardiotoxic analogous of ANT has proven challenging [54]; these approaches will not be discussed here.
Table 2
Therapeutic approaches targeting oxidative stress and inflammation for the prevention of anthracycline (ANT)-related cardiotoxicity: evidence from preclinical studies and clinical trials
Antioxidant agents
First Author, year (ref.)
Molecule
Study design
Population
Main results
Macedo et al., 2019 [56]
Dexrazoxane
Meta-analysis
Patients with BC from 9 clinical trials (n = 2177)
Reduction in cardiac events and HF incidence; no effects on the efficacy of anticancer therapy
Akolkar et al., 2017 [61]
Vitamin C
In vitro study
Rat CMs
Inhibition of pro-oxidant and pro-inflammatory cascade
Akolkar et al., 2017 [62]
Vitamin C
Animal study
Rats receiving ANT
Inhibition of pro-oxidant and pro-inflammatory cascade, lower rates of cardiac damage, systolic and diastolic dysfunction
Berthiaume et al., 2005 [63]
Vitamin E
Animal study
Rats receiving ANT
No prevention of mitochondrial dysfunction and histological changes in the heart
Carbone et al., 2012 [64]
Omega 3 PUFA
Animal study
Sheep receiving ANT
Exacerbation of ANT cardiotoxicity
Iarussi et al., 1994 [68]
Coenzyme-Q
Clinical study (not RCT)
Patients with ALL (n = 20)
Reduction in LV systolic dysfunction and wall motion abnormalities
Chen et al., 2016 [69]
Coenzyme-Q
Animal study
Rats receiving ANT
Reduction in histological changes in the heart
Najafi et al., 2019 [70]
Melatonin
Systematic review of pre-clinical studies
28 studies (mostly on mice/rats)
Decreased mortality, body weight and heart weight, ascites; reduction of histological changes in the heart
Myers et al., 1983 [71]
N-acetylcysteine
RCT
Patients with solid tumors (n = 54)
No difference in HF incidence
Waldner et al., 2006 [72]
L-carnitine
RCT
Patients with NHL (n = 40)
No difference in HF incidence
Gallegos-Castorena et al., 2007 [73]
Amifostine
Clinical study (not RCT)
Patients with osteosarcoma (n = 28)
No difference in HF incidence
Kheiri et al., 2018 [77]
Carvedilol
Meta-analysis
Patients with solid or blood cancer (n = 633)
Reduction in HF incidence and of absolute LVEF decrease
Bosch et al., 2013 [78]
Carvedilol/enalapril
RCT
Patients with blood cancer (n = 90)
Preservation of LV systolic function
Hiona et al., 2011 [81]
Enalapril
Animal study
Rats receiving ANT
Preservation of LV systolic function and mitochondrial respiratory efficacy
Anti-inflammatory agents
Reference
Molecule
Study design
Population
Main results
Sheibani et al., 2020 [86]
Dapsone
Animal study
Rats receiving ANT
Reduction in cardiac levels of prooxidant and proinflammatory factors; preservation of cardiac function
Sheta et al., 2016 [83]
Metformin or sitagliptin
Animal study
Rats receiving ANT
Blunting of inflammatory pathways and preservation of cardiac function (+ + sitagliptin)
Peng et al., 2019 [84]
Teneligliptin
In vitro study
Rat and human CMs
Reduction in CM damage, apoptosis, and proinflammatory cytokines
Seicean et al., 2012 [87]
Statins
Retrospective study
Patients with BC (n = 628)
Reduction in HF incidence and cardiac mortality
ALL acute lymphoblastic leukemia, BC breast cancer, CM cardiomyocyte, HF heart failure, IL-1 interleukin-1, LV left ventricle, LVEF left ventricular ejection fraction, NHL non-Hodgkin lymphoma, PUFA polyunsaturated fatty acids, RCT randomized clinical trial, TNFα tumor necrosis factor alpha

Antioxidant agents

Dexrazoxane (DEX) is the only drug approved for the primary prevention of ANT cardiotoxicity. DEX is an iron-chelator that reduces ROS production by impairing the formation of iron-ANT complexes; this molecule also inhibits cardiomyocyte apoptosis by blocking ANT binding to topoisomerase IIβ [55]. A significant reduction in cardiac events and HF incidence in a cohort of 2177 patients with breast cancer exposed to ANT and treated with DEX, without any reduction in the antineoplastic efficacy of the chemotherapy regimens, has been reported [56]. DEX is currently approved in Europe for adults with advanced metastatic breast cancer who have received a cumulative dose ≥ 300 mg/m2 of doxorubicin or ≥ 540 mg/m2 of epirubicin and would benefit from continued ANT-based therapy [1]. Despite some concerns of short- and long-term risks (e.g., myelosuppression, acute myeloid leukemia), studies have shown a generally safe profile even in younger patients [5759]. Therefore, a recent opening has been made for the use of DEX in patients under 18 years requiring high ANT doses (https://​www.​ema.​europa.​eu/​en/​medicines/​human/​referrals/​cardioxane).
Other antioxidants have been tested [60]. Similar to DEX, these molecules are believed to reduce ROS-related cardiac damage and, at least partially, inhibit cardiomyocyte apoptosis. One of these molecules is vitamin C, which was reported to mitigate DOX-induced oxidative and nitrosative stress and apoptosis in isolated cardiomyocytes [61], and to blunt inflammatory activation, improve systolic and diastolic function, and survival in rats exposed to DOX [62]. Similarly, vitamin E relieved oxidative stress in rats, but was not sufficient to protect cardiac mitochondrial membranes from DOX toxicity [63].
The antioxidant omega-3 polyunsaturated fatty acids (PUFA) failed to afford cardiac protection to sheep receiving ANT [64], and an open-label, phase II clinical trial showed no significant adverse effects and preserved efficacy of chemotherapy in patients receiving ANT [65].
Coenzyme Q10 is a fat-soluble electron carrier in mitochondria and a coenzyme in different energetic pathways [66]. A significant decrease in myocardial content of coenzyme Q10 has been observed in ANT-related cardiomyopathy [67]. Supplementation of this molecule has been tested in both pre-clinical studies [68, 69] and small clinical trials [68, 69], whose results do not allow any definite conclusion.
Melatonin is a hormone with antioxidant effect that can be supplemented with limited adverse effects. Several preclinical studies have investigated the role of melatonin in ANT-related cardiotoxicity. Based on a recent meta-analysis of 28 studies, melatonin administration appeared safe and associated with reduced cardiac damage and mortality [70]. N-acetylcysteine has a well-known antioxidant efficacy but did not reduce HF occurrence in the only randomized controlled trial (RCT) on patients with solid cancers treated with ANT [71]. The antioxidant molecules l-carnitine and amifostine seem to not provide any benefit to human patients treated with ANT [72, 73].
Carvedilol, a β-1, β-2, and α-1 adrenergic receptor blocker, has an antioxidant action deriving from the inhibition of the complex 1 of the respiratory chain [74] and reduces lipid peroxidation [75, 76]. In a meta-analysis including 8 trials and a total of 633 patients, co-treatment with carvedilol was associated with a lower rate of both HF occurrence and absolute decrease in LV ejection fraction, although the reliability of this conclusion is limited by the high degree of heterogeneity in the study populations and endpoints [77]. The OVERCOME (preventiOn of left Ventricular dysfunction with Enalapril and caRvedilol in patients submitted to intensive ChemOtherapy for the treatment of Malignant hEmopathies) study was a small (n = 90 patients) trial showing a protective role of the combination of carvedilol and the angiotensin-converting enzyme inhibitor (ACEi) enalapril in patients with blood neoplasms, 40% of whom receiving ANT [78]. Several other studies reported the positive effects of ACEi and angiotensin receptor blockers in preventing ANT cardiotoxicity, because of their anti-neurohormonal, vasodilating, and possibly antioxidant and anti-inflammatory actions [7982].

Anti-inflammatory agents

Secondary anti-inflammatory and potential antioxidant properties have been attributed to some molecules routinely used in different clinical settings. The anti-diabetic drugs dipeptidyl-peptidase 4 inhibitors (DPP4i) seem to inhibit inflammatory and pro-oxidant pathways. In a rat model of DOX-toxicity, the DPP4i sitagliptin antagonized the inflammatory cascade involved in ANT cardiotoxicity, reducing cell damage, apoptosis, and cytokine levels [83]. A class-dependent effect of DPP4i might be postulated given the efficacy of another molecule (teneligliptin) in a recent in vitro study [84]. Similarly, the inhibitors of the sodium glucose co-transporter 2 empagliflozin and dapagliflozin appear to reduce ANT-dependent inflammation and cardiotoxicity in vitro and in vivo [85]. Furthermore, the antibiotic dapsone was tested in rats that had received DOX, leading to a reduction in heart levels of oxidant factors and pro-inflammatory cytokines and a significant amelioration of electrocardiographic and electrophysiological parameters, heart contractility, and biomarkers concentrations, as well as positive effects at the histopathological level [86]. Finally, the potent anti-inflammatory and anti-oxidant effects of statins might explain the beneficial effects of these molecules in preventing incident HF in a cohort of 628 newly diagnosed breast cancer patients receiving ANT [87]. An RCT is underway to verify the efficacy of atorvastatin in this clinical setting (NCT01988571).

Conclusions and future perspectives

Chemotherapy with ANT-based regimens remains a cornerstone of treatment of many solid and blood tumors, but is associated with a nonnegligible risk of cardiotoxicity, which may lead over time to clinically manifest HF. Our current understanding of cardiac damage by ANT includes a central role of oxidative stress and inflammation. Detection of these processes through circulating biomarkers or imaging techniques might then be helpful for early diagnosis and risk stratification. Furthermore, therapeutic strategies relieving oxidative stress and inflammation hold some promise to prevent HF development or at least mitigate cardiac damage, although further evidence is needed on the efficacy of these drugs, either alone or as part of combination therapies with therapies for neurohormonal antagonism.

Key points

  • Anthracyclines are potent and common chemotherapeutics for hematological and solid tumors but are burdened with a serious risk of cardiotoxicity, which may be partly mediated by inflammation and oxidative stress.
  • Several serum biomarkers related to inflammation and oxidative stress (e.g., CRP, myeloperoxidase, glutathione peroxidase) have been proposed for risk stratification and detection of anthracyclines cardiotoxicity, but evidence is still lacking.
  • Anthracycline cardiotoxicity can be monitored with imaging techniques, e.g., echocardiography, computed or positron emission tomography, or cardiac magnetic resonance that can document the evolution of myocardial inflammation and its sequelae.
  • Apart from dexrazoxane, which reduces ROS production, no other drug is approved for the primary prevention of ANT cardiotoxicity, but several anti-inflammatory and antioxidant agents are being investigated.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Jetzt e.Med zum Sonderpreis bestellen!

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Jetzt bestellen und 100 € sparen!

Literatur
1.
Zurück zum Zitat Zamorano JL, Lancellotti P, Rodriguez Munoz D, Aboyans V, Asteggiano R, Galderisi M et al (2016) 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 37(36):2768–2801PubMedCrossRef Zamorano JL, Lancellotti P, Rodriguez Munoz D, Aboyans V, Asteggiano R, Galderisi M et al (2016) 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 37(36):2768–2801PubMedCrossRef
2.
Zurück zum Zitat Zamorano JL, Gottfridsson C, Asteggiano R, Atar D, Badimon L, Bax JJ, et al (2020) The cancer patient and cardiology. Eur J Heart Fail Zamorano JL, Gottfridsson C, Asteggiano R, Atar D, Badimon L, Bax JJ, et al (2020) The cancer patient and cardiology. Eur J Heart Fail
3.
Zurück zum Zitat Lopez-Sendon J, Alvarez-Ortega C, Zamora Aunon P, Buno Soto A, Lyon AR, Farmakis D, et al. Classification, prevalence, and outcomes of anticancer therapy-induced cardiotoxicity: the CARDIOTOX registry. Eur Heart J Lopez-Sendon J, Alvarez-Ortega C, Zamora Aunon P, Buno Soto A, Lyon AR, Farmakis D, et al. Classification, prevalence, and outcomes of anticancer therapy-induced cardiotoxicity: the CARDIOTOX registry. Eur Heart J
4.
Zurück zum Zitat Rocca C, Pasqua T, Cerra MC, Angelone T (2020) Cardiac damage in anthracyclines therapy: focus on oxidative stress and inflammation. Antioxid Redox Sign Rocca C, Pasqua T, Cerra MC, Angelone T (2020) Cardiac damage in anthracyclines therapy: focus on oxidative stress and inflammation. Antioxid Redox Sign
5.
Zurück zum Zitat Cardinale D, Colombo A, Bacchiani G, Tedeschi I, Meroni CA, Veglia F et al (2015) Early detection of anthracycline cardiotoxicity and improvement with heart failure therapy. Circulation 131(22):1981–1988PubMedCrossRef Cardinale D, Colombo A, Bacchiani G, Tedeschi I, Meroni CA, Veglia F et al (2015) Early detection of anthracycline cardiotoxicity and improvement with heart failure therapy. Circulation 131(22):1981–1988PubMedCrossRef
6.
Zurück zum Zitat Cardinale D, Biasillo G, Cipolla CM (2016) Curing cancer, saving the heart: a challenge that cardioncology should not miss. Curr Cardiol Rep 18(6):51PubMedCrossRef Cardinale D, Biasillo G, Cipolla CM (2016) Curing cancer, saving the heart: a challenge that cardioncology should not miss. Curr Cardiol Rep 18(6):51PubMedCrossRef
7.
Zurück zum Zitat Curigliano G, Lenihan D, Fradley M, Ganatra S, Barac A, Blaes A et al (2020) Management of cardiac disease in cancer patients throughout oncological treatment: ESMO consensus recommendations. Ann Oncol : Official J European Society Med Oncol 31(2):171–190CrossRef Curigliano G, Lenihan D, Fradley M, Ganatra S, Barac A, Blaes A et al (2020) Management of cardiac disease in cancer patients throughout oncological treatment: ESMO consensus recommendations. Ann Oncol : Official J European Society Med Oncol 31(2):171–190CrossRef
8.
Zurück zum Zitat Cardinale D, Colombo A, Sandri MT, Lamantia G, Colombo N, Civelli M et al (2006) Prevention of high-dose chemotherapy-induced cardiotoxicity in high-risk patients by angiotensin-converting enzyme inhibition. Circulation 114(23):2474–2481PubMedCrossRef Cardinale D, Colombo A, Sandri MT, Lamantia G, Colombo N, Civelli M et al (2006) Prevention of high-dose chemotherapy-induced cardiotoxicity in high-risk patients by angiotensin-converting enzyme inhibition. Circulation 114(23):2474–2481PubMedCrossRef
9.
Zurück zum Zitat Aimo A, Castiglione V, Borrelli C, Saccaro LF, Franzini M, Masi S et al (2019) Oxidative stress and inflammation in the evolution of heart failure: From pathophysiology to therapeutic strategies. Eur J Prev Cardiol 2047487319870344 Aimo A, Castiglione V, Borrelli C, Saccaro LF, Franzini M, Masi S et al (2019) Oxidative stress and inflammation in the evolution of heart failure: From pathophysiology to therapeutic strategies. Eur J Prev Cardiol 2047487319870344
10.
Zurück zum Zitat Vejpongsa P, Yeh ET (2014) Topoisomerase 2beta: a promising molecular target for primary prevention of anthracycline-induced cardiotoxicity. Clin Pharmacol Ther 95(1):45–52PubMedCrossRef Vejpongsa P, Yeh ET (2014) Topoisomerase 2beta: a promising molecular target for primary prevention of anthracycline-induced cardiotoxicity. Clin Pharmacol Ther 95(1):45–52PubMedCrossRef
11.
Zurück zum Zitat Octavia Y, Tocchetti CG, Gabrielson KL, Janssens S, Crijns HJ, Moens AL (2012) Doxorubicin-induced cardiomyopathy: from molecular mechanisms to therapeutic strategies. J Mol Cell Cardiol 52(6):1213–1225PubMedCrossRef Octavia Y, Tocchetti CG, Gabrielson KL, Janssens S, Crijns HJ, Moens AL (2012) Doxorubicin-induced cardiomyopathy: from molecular mechanisms to therapeutic strategies. J Mol Cell Cardiol 52(6):1213–1225PubMedCrossRef
12.
Zurück zum Zitat Nordgren KK, Wallace KB (2014) Keap1 redox-dependent regulation of doxorubicin-induced oxidative stress response in cardiac myoblasts. Toxicol Appl Pharmacol 274(1):107–116PubMedCrossRef Nordgren KK, Wallace KB (2014) Keap1 redox-dependent regulation of doxorubicin-induced oxidative stress response in cardiac myoblasts. Toxicol Appl Pharmacol 274(1):107–116PubMedCrossRef
13.
Zurück zum Zitat Lebrecht D, Walker UA (2007) Role of mtDNA lesions in anthracycline cardiotoxicity. Cardiovasc Toxicol 7(2):108–113PubMedCrossRef Lebrecht D, Walker UA (2007) Role of mtDNA lesions in anthracycline cardiotoxicity. Cardiovasc Toxicol 7(2):108–113PubMedCrossRef
14.
Zurück zum Zitat Simunek T, Sterba M, Popelova O, Adamcova M, Hrdina R, Gersl V (2009) Anthracycline-induced cardiotoxicity: overview of studies examining the roles of oxidative stress and free cellular iron. Pharmacol Rep : PR 61(1):154–171PubMedCrossRef Simunek T, Sterba M, Popelova O, Adamcova M, Hrdina R, Gersl V (2009) Anthracycline-induced cardiotoxicity: overview of studies examining the roles of oxidative stress and free cellular iron. Pharmacol Rep : PR 61(1):154–171PubMedCrossRef
15.
Zurück zum Zitat Zhang S, Liu X, Bawa-Khalfe T, Lu LS, Lyu YL, Liu LF et al (2012) Identification of the molecular basis of doxorubicin-induced cardiotoxicity. Nat Med 18(11):1639–1642PubMedCrossRef Zhang S, Liu X, Bawa-Khalfe T, Lu LS, Lyu YL, Liu LF et al (2012) Identification of the molecular basis of doxorubicin-induced cardiotoxicity. Nat Med 18(11):1639–1642PubMedCrossRef
16.
Zurück zum Zitat Aldieri E, Bergandi L, Riganti C, Costamagna C, Bosia A, Ghigo D (2002) Doxorubicin induces an increase of nitric oxide synthesis in rat cardiac cells that is inhibited by iron supplementation. Toxicol Appl Pharmacol 185(2):85–90PubMedCrossRef Aldieri E, Bergandi L, Riganti C, Costamagna C, Bosia A, Ghigo D (2002) Doxorubicin induces an increase of nitric oxide synthesis in rat cardiac cells that is inhibited by iron supplementation. Toxicol Appl Pharmacol 185(2):85–90PubMedCrossRef
17.
Zurück zum Zitat Bahadir A, Kurucu N, Kadioglu M, Yenilme E (2014) The role of nitric oxide in Doxorubicin-induced cardiotoxicity: experimental study. Turk J Haematol: Official Turk Society Haematol 31(1):68–74CrossRef Bahadir A, Kurucu N, Kadioglu M, Yenilme E (2014) The role of nitric oxide in Doxorubicin-induced cardiotoxicity: experimental study. Turk J Haematol: Official Turk Society Haematol 31(1):68–74CrossRef
18.
Zurück zum Zitat Wallace KB (2007) Adriamycin-induced interference with cardiac mitochondrial calcium homeostasis. Cardiovasc Toxicol 7(2):101–107PubMedCrossRef Wallace KB (2007) Adriamycin-induced interference with cardiac mitochondrial calcium homeostasis. Cardiovasc Toxicol 7(2):101–107PubMedCrossRef
19.
Zurück zum Zitat Russo M, Guida F, Paparo L, Trinchese G, Aitoro R, Avagliano C et al (2019) The novel butyrate derivative phenylalanine-butyramide protects from doxorubicin-induced cardiotoxicity. Eur J Heart Fail 21(4):519–528PubMedCrossRef Russo M, Guida F, Paparo L, Trinchese G, Aitoro R, Avagliano C et al (2019) The novel butyrate derivative phenylalanine-butyramide protects from doxorubicin-induced cardiotoxicity. Eur J Heart Fail 21(4):519–528PubMedCrossRef
20.
Zurück zum Zitat Frantz S, Falcao-Pires I, Balligand JL, Bauersachs J, Brutsaert D, Ciccarelli M et al (2018) The innate immune system in chronic cardiomyopathy: a European Society of Cardiology (ESC) scientific statement from the Working Group on Myocardial Function of the ESC. Eur J Heart Fail 20(3):445–459PubMedCrossRef Frantz S, Falcao-Pires I, Balligand JL, Bauersachs J, Brutsaert D, Ciccarelli M et al (2018) The innate immune system in chronic cardiomyopathy: a European Society of Cardiology (ESC) scientific statement from the Working Group on Myocardial Function of the ESC. Eur J Heart Fail 20(3):445–459PubMedCrossRef
21.
Zurück zum Zitat Sauter KA, Wood LJ, Wong J, Iordanov M, Magun BE (2011) Doxorubicin and daunorubicin induce processing and release of interleukin-1beta through activation of the NLRP3 inflammasome. Cancer Biol Ther 11(12):1008–1016PubMedPubMedCentralCrossRef Sauter KA, Wood LJ, Wong J, Iordanov M, Magun BE (2011) Doxorubicin and daunorubicin induce processing and release of interleukin-1beta through activation of the NLRP3 inflammasome. Cancer Biol Ther 11(12):1008–1016PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Jadapalli JK, Wright GW, Kain V, Sherwani MA, Sonkar R, Yusuf N et al (2018) Doxorubicin triggers splenic contraction and irreversible dysregulation of COX and LOX that alters the inflammation-resolution program in the myocardium. Am J Physiol Heart Circ 315(5):H1091–H1100CrossRef Jadapalli JK, Wright GW, Kain V, Sherwani MA, Sonkar R, Yusuf N et al (2018) Doxorubicin triggers splenic contraction and irreversible dysregulation of COX and LOX that alters the inflammation-resolution program in the myocardium. Am J Physiol Heart Circ 315(5):H1091–H1100CrossRef
24.
Zurück zum Zitat Mercuro G, Cadeddu C, Piras A, Dessi M, Madeddu C, Deidda M et al (2007) Early epirubicin-induced myocardial dysfunction revealed by serial tissue Doppler echocardiography: correlation with inflammatory and oxidative stress markers. Oncologist 12(9):1124–1133PubMedCrossRef Mercuro G, Cadeddu C, Piras A, Dessi M, Madeddu C, Deidda M et al (2007) Early epirubicin-induced myocardial dysfunction revealed by serial tissue Doppler echocardiography: correlation with inflammatory and oxidative stress markers. Oncologist 12(9):1124–1133PubMedCrossRef
25.
Zurück zum Zitat Dessi M, Piras A, Madeddu C, Cadeddu C, Deidda M, Massa E et al (2011) Long-term protective effects of the angiotensin receptor blocker telmisartan on epirubicin-induced inflammation, oxidative stress and myocardial dysfunction. Exp Ther Med 2(5):1003–1009PubMedPubMedCentralCrossRef Dessi M, Piras A, Madeddu C, Cadeddu C, Deidda M, Massa E et al (2011) Long-term protective effects of the angiotensin receptor blocker telmisartan on epirubicin-induced inflammation, oxidative stress and myocardial dysfunction. Exp Ther Med 2(5):1003–1009PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Erkus B, Demirtas S, Yarpuzlu AA, Can M, Genc Y, Karaca L (2007) Early prediction of anthracycline induced cardiotoxicity. Acta Paediatr (Oslo, Norway : 1992) 96(4):506–9 Erkus B, Demirtas S, Yarpuzlu AA, Can M, Genc Y, Karaca L (2007) Early prediction of anthracycline induced cardiotoxicity. Acta Paediatr (Oslo, Norway : 1992) 96(4):506–9
27.
Zurück zum Zitat Morris PG, Chen C, Steingart R, Fleisher M, Lin N, Moy B et al (2011) Troponin I and C-reactive protein are commonly detected in patients with breast cancer treated with dose-dense chemotherapy incorporating trastuzumab and lapatinib. Clin Cancer Res: Official J American Assoc Cancer Rese 17(10):3490–3499CrossRef Morris PG, Chen C, Steingart R, Fleisher M, Lin N, Moy B et al (2011) Troponin I and C-reactive protein are commonly detected in patients with breast cancer treated with dose-dense chemotherapy incorporating trastuzumab and lapatinib. Clin Cancer Res: Official J American Assoc Cancer Rese 17(10):3490–3499CrossRef
28.
Zurück zum Zitat Grover S, Leong DP, Chakrabarty A, Joerg L, Kotasek D, Cheong K et al (2013) Left and right ventricular effects of anthracycline and trastuzumab chemotherapy: a prospective study using novel cardiac imaging and biochemical markers. Int J Cardiol 168(6):5465–5467PubMedCrossRef Grover S, Leong DP, Chakrabarty A, Joerg L, Kotasek D, Cheong K et al (2013) Left and right ventricular effects of anthracycline and trastuzumab chemotherapy: a prospective study using novel cardiac imaging and biochemical markers. Int J Cardiol 168(6):5465–5467PubMedCrossRef
29.
Zurück zum Zitat Katsurada K, Ichida M, Sakuragi M, Takehara M, Hozumi Y, Kario K (2014) High-sensitivity troponin T as a marker to predict cardiotoxicity in breast cancer patients with adjuvant trastuzumab therapy. SpringerPlus 3:620PubMedPubMedCentralCrossRef Katsurada K, Ichida M, Sakuragi M, Takehara M, Hozumi Y, Kario K (2014) High-sensitivity troponin T as a marker to predict cardiotoxicity in breast cancer patients with adjuvant trastuzumab therapy. SpringerPlus 3:620PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Ky B, Putt M, Sawaya H, French B, Januzzi JL Jr, Sebag IA et al (2014) Early increases in multiple biomarkers predict subsequent cardiotoxicity in patients with breast cancer treated with doxorubicin, taxanes, and trastuzumab. J Am Coll Cardiol 63(8):809–816CrossRefPubMed Ky B, Putt M, Sawaya H, French B, Januzzi JL Jr, Sebag IA et al (2014) Early increases in multiple biomarkers predict subsequent cardiotoxicity in patients with breast cancer treated with doxorubicin, taxanes, and trastuzumab. J Am Coll Cardiol 63(8):809–816CrossRefPubMed
31.
Zurück zum Zitat Putt M, Hahn VS, Januzzi JL, Sawaya H, Sebag IA, Plana JC et al (2015) Longitudinal changes in multiple biomarkers are associated with cardiotoxicity in breast cancer patients treated with doxorubicin, taxanes, and trastuzumab. Clin Chem 61(9):1164–1172PubMedPubMedCentralCrossRef Putt M, Hahn VS, Januzzi JL, Sawaya H, Sebag IA, Plana JC et al (2015) Longitudinal changes in multiple biomarkers are associated with cardiotoxicity in breast cancer patients treated with doxorubicin, taxanes, and trastuzumab. Clin Chem 61(9):1164–1172PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Lipshultz SE, Miller TL, Scully RE, Lipsitz SR, Rifai N, Silverman LB et al (2012) Changes in cardiac biomarkers during doxorubicin treatment of pediatric patients with high-risk acute lymphoblastic leukemia: associations with long-term echocardiographic outcomes. J Clin Oncol: Official J American Society Clin Oncol 30(10):1042–1049CrossRef Lipshultz SE, Miller TL, Scully RE, Lipsitz SR, Rifai N, Silverman LB et al (2012) Changes in cardiac biomarkers during doxorubicin treatment of pediatric patients with high-risk acute lymphoblastic leukemia: associations with long-term echocardiographic outcomes. J Clin Oncol: Official J American Society Clin Oncol 30(10):1042–1049CrossRef
33.
Zurück zum Zitat Kouloubinis A, Sofroniadou S, Panoulas VF, Makaritsis K, Revela I, Karavolias G et al (2015) The role of TNF-alpha, Fas/Fas ligand system and NT-proBNP in the early detection of asymptomatic left ventricular dysfunction in cancer patients treated with anthracyclines. Int J Cardiol Heart Vasc 6:85–90PubMedPubMedCentral Kouloubinis A, Sofroniadou S, Panoulas VF, Makaritsis K, Revela I, Karavolias G et al (2015) The role of TNF-alpha, Fas/Fas ligand system and NT-proBNP in the early detection of asymptomatic left ventricular dysfunction in cancer patients treated with anthracyclines. Int J Cardiol Heart Vasc 6:85–90PubMedPubMedCentral
34.
Zurück zum Zitat Mills PJ, Ancoli-Israel S, Parker B, Natarajan L, Hong S, Jain S et al (2008) Predictors of inflammation in response to anthracycline-based chemotherapy for breast cancer. Brain Behav Immun 22(1):98–104PubMedCrossRef Mills PJ, Ancoli-Israel S, Parker B, Natarajan L, Hong S, Jain S et al (2008) Predictors of inflammation in response to anthracycline-based chemotherapy for breast cancer. Brain Behav Immun 22(1):98–104PubMedCrossRef
35.
Zurück zum Zitat Sawaya H, Sebag IA, Plana JC, Januzzi JL, Ky B, Tan TC et al (2012) Assessment of echocardiography and biomarkers for the extended prediction of cardiotoxicity in patients treated with anthracyclines, taxanes, and trastuzumab. Circ Cardiovasc Imaging 5(5):596–603PubMedPubMedCentralCrossRef Sawaya H, Sebag IA, Plana JC, Januzzi JL, Ky B, Tan TC et al (2012) Assessment of echocardiography and biomarkers for the extended prediction of cardiotoxicity in patients treated with anthracyclines, taxanes, and trastuzumab. Circ Cardiovasc Imaging 5(5):596–603PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Armenian SH, Gelehrter SK, Vase T, Venkatramani R, Landier W, Wilson KD et al (2014) Screening for cardiac dysfunction in anthracycline-exposed childhood cancer survivors. Clin Cancer Res : Official J American Association Cancer Res 20(24):6314–6323CrossRef Armenian SH, Gelehrter SK, Vase T, Venkatramani R, Landier W, Wilson KD et al (2014) Screening for cardiac dysfunction in anthracycline-exposed childhood cancer survivors. Clin Cancer Res : Official J American Association Cancer Res 20(24):6314–6323CrossRef
37.
Zurück zum Zitat Freres P, Bouznad N, Servais L, Josse C, Wenric S, Poncin A et al (2018) Variations of circulating cardiac biomarkers during and after anthracycline-containing chemotherapy in breast cancer patients. BMC cancer 18(1):102PubMedPubMedCentralCrossRef Freres P, Bouznad N, Servais L, Josse C, Wenric S, Poncin A et al (2018) Variations of circulating cardiac biomarkers during and after anthracycline-containing chemotherapy in breast cancer patients. BMC cancer 18(1):102PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Beer LA, Kossenkov AV, Liu Q, Luning Prak E, Domchek S, Speicher DW et al (2016) Baseline immunoglobulin E levels as a marker of doxorubicin- and trastuzumab-associated cardiac dysfunction. Circ Res 119(10):1135–1144PubMedPubMedCentralCrossRef Beer LA, Kossenkov AV, Liu Q, Luning Prak E, Domchek S, Speicher DW et al (2016) Baseline immunoglobulin E levels as a marker of doxorubicin- and trastuzumab-associated cardiac dysfunction. Circ Res 119(10):1135–1144PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Gabriel A, Stringer B, Hadfield MJ, Madady M (2019) Case of anthracycline-induced cardiogenic shock: a call to optimize modifiable cardiac risk factors prior to chemotherapy. Cureus 11(6):e4961PubMedPubMedCentral Gabriel A, Stringer B, Hadfield MJ, Madady M (2019) Case of anthracycline-induced cardiogenic shock: a call to optimize modifiable cardiac risk factors prior to chemotherapy. Cureus 11(6):e4961PubMedPubMedCentral
40.
Zurück zum Zitat Virizuela JA, Garcia AM, de Las PR, Santaballa A, Andres R, Beato C et al (2019) SEOM clinical guidelines on cardiovascular toxicity (2018). Clin Transl Oncol: Official Publication of the Federation of Spanish Oncology Societies and of the National Cancer Institute of Mexico 21(1):94–105CrossRef Virizuela JA, Garcia AM, de Las PR, Santaballa A, Andres R, Beato C et al (2019) SEOM clinical guidelines on cardiovascular toxicity (2018). Clin Transl Oncol: Official Publication of the Federation of Spanish Oncology Societies and of the National Cancer Institute of Mexico 21(1):94–105CrossRef
41.
Zurück zum Zitat Aquaro GD, Todiere G, Strata E, Barison A, Di Bella G, Lombardi M (2014) Usefulness of India ink artifact in steady-state free precession pulse sequences for detection and quantification of intramyocardial fat. JMRI 40(1):126–132PubMedCrossRef Aquaro GD, Todiere G, Strata E, Barison A, Di Bella G, Lombardi M (2014) Usefulness of India ink artifact in steady-state free precession pulse sequences for detection and quantification of intramyocardial fat. JMRI 40(1):126–132PubMedCrossRef
42.
Zurück zum Zitat Grigoratos C, Di Bella G, Aquaro GD (2019) Diagnostic and prognostic role of cardiac magnetic resonance in acute myocarditis. Heart Fail Rev 24(1):81–90PubMedCrossRef Grigoratos C, Di Bella G, Aquaro GD (2019) Diagnostic and prognostic role of cardiac magnetic resonance in acute myocarditis. Heart Fail Rev 24(1):81–90PubMedCrossRef
43.
Zurück zum Zitat Aquaro GD, Barison A, Todiere G, Grigoratos C, Ait Ali L, Di Bella G et al (2016) Usefulness of combined functional assessment by cardiac magnetic resonance and tissue characterization versus task force criteria for diagnosis of arrhythmogenic right ventricular cardiomyopathy. Am J Cardiol 118(11):1730–1736PubMedCrossRef Aquaro GD, Barison A, Todiere G, Grigoratos C, Ait Ali L, Di Bella G et al (2016) Usefulness of combined functional assessment by cardiac magnetic resonance and tissue characterization versus task force criteria for diagnosis of arrhythmogenic right ventricular cardiomyopathy. Am J Cardiol 118(11):1730–1736PubMedCrossRef
44.
Zurück zum Zitat Galan-Arriola C, Lobo M, Vilchez-Tschischke JP, Lopez GJ, de Molina-Iracheta A, Perez-Martinez C et al (2019) Serial magnetic resonance imaging to identify early stages of anthracycline-induced cardiotoxicity. J Am Coll Cardiol 73(7):779–791PubMedCrossRef Galan-Arriola C, Lobo M, Vilchez-Tschischke JP, Lopez GJ, de Molina-Iracheta A, Perez-Martinez C et al (2019) Serial magnetic resonance imaging to identify early stages of anthracycline-induced cardiotoxicity. J Am Coll Cardiol 73(7):779–791PubMedCrossRef
45.
Zurück zum Zitat Thavendiranathan P, Wintersperger BJ, Flamm SD, Marwick TH (2013) Cardiac MRI in the assessment of cardiac injury and toxicity from cancer chemotherapy: a systematic review. Circ Cardiovasc Imaging 6(6):1080–1091PubMedCrossRef Thavendiranathan P, Wintersperger BJ, Flamm SD, Marwick TH (2013) Cardiac MRI in the assessment of cardiac injury and toxicity from cancer chemotherapy: a systematic review. Circ Cardiovasc Imaging 6(6):1080–1091PubMedCrossRef
46.
Zurück zum Zitat Safee ZM, Baark F, Waters ECT, Veronese M, Pell VR, Clark JE et al (2019) Detection of anthracycline-induced cardiotoxicity using perfusion-corrected (99m)Tc sestamibi SPECT. Sci Rep 9(1):216PubMedPubMedCentralCrossRef Safee ZM, Baark F, Waters ECT, Veronese M, Pell VR, Clark JE et al (2019) Detection of anthracycline-induced cardiotoxicity using perfusion-corrected (99m)Tc sestamibi SPECT. Sci Rep 9(1):216PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Plana JC, Galderisi M, Barac A, Ewer MS, Ky B, Scherrer-Crosbie M et al (2014) Expert consensus for multimodality imaging evaluation of adult patients during and after cancer therapy: a report from the American Society of Echocardiography and the European Association of Cardiovascular Imaging. Eur Heart J Cardiovasc Imaging 15(10):1063–1093PubMedPubMedCentralCrossRef Plana JC, Galderisi M, Barac A, Ewer MS, Ky B, Scherrer-Crosbie M et al (2014) Expert consensus for multimodality imaging evaluation of adult patients during and after cancer therapy: a report from the American Society of Echocardiography and the European Association of Cardiovascular Imaging. Eur Heart J Cardiovasc Imaging 15(10):1063–1093PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Bauckneht M, Ferrarazzo G, Fiz F, Morbelli S, Sarocchi M, Pastorino F et al (2017) Doxorubicin effect on myocardial metabolism as a prerequisite for subsequent development of cardiac toxicity: a translational (18)F-FDG PET/CT observation. J Nucl Med : Official Publication Society Nucl Med 58(10):1638–1645CrossRef Bauckneht M, Ferrarazzo G, Fiz F, Morbelli S, Sarocchi M, Pastorino F et al (2017) Doxorubicin effect on myocardial metabolism as a prerequisite for subsequent development of cardiac toxicity: a translational (18)F-FDG PET/CT observation. J Nucl Med : Official Publication Society Nucl Med 58(10):1638–1645CrossRef
49.
Zurück zum Zitat Groch MW, DePuey EG, Belzberg AC, Erwin WD, Kamran M, Barnett CA et al (2001) Planar imaging versus gated blood-pool SPECT for the assessment of ventricular performance: a multicenter study. J Nucl Med: Official Publication Society Nucl Med 42(12):1773–1779 Groch MW, DePuey EG, Belzberg AC, Erwin WD, Kamran M, Barnett CA et al (2001) Planar imaging versus gated blood-pool SPECT for the assessment of ventricular performance: a multicenter study. J Nucl Med: Official Publication Society Nucl Med 42(12):1773–1779
50.
Zurück zum Zitat Ylanen K, Poutanen T, Savikurki-Heikkila P, Rinta-Kiikka I, Eerola A, Vettenranta K (2013) Cardiac magnetic resonance imaging in the evaluation of the late effects of anthracyclines among long-term survivors of childhood cancer. J Am Coll Cardiol 61(14):1539–1547PubMedCrossRef Ylanen K, Poutanen T, Savikurki-Heikkila P, Rinta-Kiikka I, Eerola A, Vettenranta K (2013) Cardiac magnetic resonance imaging in the evaluation of the late effects of anthracyclines among long-term survivors of childhood cancer. J Am Coll Cardiol 61(14):1539–1547PubMedCrossRef
51.
Zurück zum Zitat Harries I, Biglino G, Baritussio A, De Garate E, Dastidar A, Plana JC et al (2019) Long term cardiovascular magnetic resonance phenotyping of anthracycline cardiomyopathy. Int J Cardiol 292:248–252PubMedCrossRef Harries I, Biglino G, Baritussio A, De Garate E, Dastidar A, Plana JC et al (2019) Long term cardiovascular magnetic resonance phenotyping of anthracycline cardiomyopathy. Int J Cardiol 292:248–252PubMedCrossRef
52.
Zurück zum Zitat Tham EB, Haykowsky MJ, Chow K, Spavor M, Kaneko S, Khoo NS et al (2013) Diffuse myocardial fibrosis by T1-mapping in children with subclinical anthracycline cardiotoxicity: relationship to exercise capacity, cumulative dose and remodeling. J Cardiovasc Magn Reson : Official J Society Cardiovasc Magn Reson 15:48CrossRef Tham EB, Haykowsky MJ, Chow K, Spavor M, Kaneko S, Khoo NS et al (2013) Diffuse myocardial fibrosis by T1-mapping in children with subclinical anthracycline cardiotoxicity: relationship to exercise capacity, cumulative dose and remodeling. J Cardiovasc Magn Reson : Official J Society Cardiovasc Magn Reson 15:48CrossRef
53.
Zurück zum Zitat Jordan JH, Vasu S, Morgan TM, D'Agostino RB, Jr., Melendez GC, Hamilton CA et al (2016) Anthracycline-associated T1 mapping characteristics are elevated independent of the presence of Cardiovascular comorbidities in cancer survivors. Circ Cardiovasc Imaging 9(8) Jordan JH, Vasu S, Morgan TM, D'Agostino RB, Jr., Melendez GC, Hamilton CA et al (2016) Anthracycline-associated T1 mapping characteristics are elevated independent of the presence of Cardiovascular comorbidities in cancer survivors. Circ Cardiovasc Imaging 9(8)
54.
Zurück zum Zitat Menna P, Salvatorelli E (2017) Primary Prevention Strategies for Anthracycline Cardiotoxicity: A Brief Overview. Chemotherapy 62(3):159–168PubMedCrossRef Menna P, Salvatorelli E (2017) Primary Prevention Strategies for Anthracycline Cardiotoxicity: A Brief Overview. Chemotherapy 62(3):159–168PubMedCrossRef
55.
Zurück zum Zitat Lyu YL, Kerrigan JE, Lin CP, Azarova AM, Tsai YC, Ban Y et al (2007) Topoisomerase IIbeta mediated DNA double-strand breaks: implications in doxorubicin cardiotoxicity and prevention by dexrazoxane. Can Res 67(18):8839–8846CrossRef Lyu YL, Kerrigan JE, Lin CP, Azarova AM, Tsai YC, Ban Y et al (2007) Topoisomerase IIbeta mediated DNA double-strand breaks: implications in doxorubicin cardiotoxicity and prevention by dexrazoxane. Can Res 67(18):8839–8846CrossRef
56.
Zurück zum Zitat Macedo AVS, Hajjar LA, Lyon AR, Nascimento BR, Putzu A, Rossi L et al (2019) Efficacy of Dexrazoxane in Preventing Anthracycline Cardiotoxicity in Breast Cancer 1(1):68–79 Macedo AVS, Hajjar LA, Lyon AR, Nascimento BR, Putzu A, Rossi L et al (2019) Efficacy of Dexrazoxane in Preventing Anthracycline Cardiotoxicity in Breast Cancer 1(1):68–79
57.
Zurück zum Zitat Seif AE, Walker DM, Li Y, Huang YS, Kavcic M, Torp K et al (2015) Dexrazoxane exposure and risk of secondary acute myeloid leukemia in pediatric oncology patients. Pediatr Blood Cancer 62(4):704–709PubMedCrossRef Seif AE, Walker DM, Li Y, Huang YS, Kavcic M, Torp K et al (2015) Dexrazoxane exposure and risk of secondary acute myeloid leukemia in pediatric oncology patients. Pediatr Blood Cancer 62(4):704–709PubMedCrossRef
58.
Zurück zum Zitat Chow EJ, Asselin BL, Schwartz CL, Doody DR, Leisenring WM, Aggarwal S et al (2015) Late mortality after dexrazoxane treatment: a report from the Children’s Oncology Group. J Clin Oncol : Official J American Society Clin Oncol 33(24):2639–2645CrossRef Chow EJ, Asselin BL, Schwartz CL, Doody DR, Leisenring WM, Aggarwal S et al (2015) Late mortality after dexrazoxane treatment: a report from the Children’s Oncology Group. J Clin Oncol : Official J American Society Clin Oncol 33(24):2639–2645CrossRef
59.
Zurück zum Zitat Reichardt P, Tabone MD, Mora J, Morland B, Jones RL (2018) Risk-benefit of dexrazoxane for preventing anthracycline-related cardiotoxicity: re-evaluating the European labeling. Future Oncol 14(25):2663–2676PubMedCrossRef Reichardt P, Tabone MD, Mora J, Morland B, Jones RL (2018) Risk-benefit of dexrazoxane for preventing anthracycline-related cardiotoxicity: re-evaluating the European labeling. Future Oncol 14(25):2663–2676PubMedCrossRef
60.
Zurück zum Zitat Vincent DT, Ibrahim YF, Espey MG, Suzuki YJ (2013) The role of antioxidants in the era of cardiooncology. Cancer Chemother Pharmacol 72(6):1157–1168PubMedPubMedCentralCrossRef Vincent DT, Ibrahim YF, Espey MG, Suzuki YJ (2013) The role of antioxidants in the era of cardiooncology. Cancer Chemother Pharmacol 72(6):1157–1168PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Akolkar G, Bagchi AK, Ayyappan P, Jassal DS, Singal PK (2017) Doxorubicin-induced nitrosative stress is mitigated by vitamin C via the modulation of nitric oxide synthases. Am J Physiol Cell Physiol 312(4):C418–C427PubMedCrossRef Akolkar G, Bagchi AK, Ayyappan P, Jassal DS, Singal PK (2017) Doxorubicin-induced nitrosative stress is mitigated by vitamin C via the modulation of nitric oxide synthases. Am J Physiol Cell Physiol 312(4):C418–C427PubMedCrossRef
62.
Zurück zum Zitat Akolkar G, da Silva DD, Ayyappan P, Bagchi AK, Jassal DS, Salemi VMC et al (2017) Vitamin C mitigates oxidative/nitrosative stress and inflammation in doxorubicin-induced cardiomyopathy. Am J Physiol Heart Circ Physiol 313(4):H795-h809PubMedCrossRef Akolkar G, da Silva DD, Ayyappan P, Bagchi AK, Jassal DS, Salemi VMC et al (2017) Vitamin C mitigates oxidative/nitrosative stress and inflammation in doxorubicin-induced cardiomyopathy. Am J Physiol Heart Circ Physiol 313(4):H795-h809PubMedCrossRef
63.
Zurück zum Zitat Berthiaume JM, Oliveira PJ, Fariss MW, Wallace KB (2005) Dietary vitamin E decreases doxorubicin-induced oxidative stress without preventing mitochondrial dysfunction. Cardiovasc Toxicol 5(3):257–267PubMedCrossRef Berthiaume JM, Oliveira PJ, Fariss MW, Wallace KB (2005) Dietary vitamin E decreases doxorubicin-induced oxidative stress without preventing mitochondrial dysfunction. Cardiovasc Toxicol 5(3):257–267PubMedCrossRef
64.
Zurück zum Zitat Carbone A, Psaltis PJ, Nelson AJ, Metcalf R, Richardson JD, Weightman M et al (2012) Dietary omega-3 supplementation exacerbates left ventricular dysfunction in an ovine model of anthracycline-induced cardiotoxicity. J Cardiac Fail 18(6):502–511CrossRef Carbone A, Psaltis PJ, Nelson AJ, Metcalf R, Richardson JD, Weightman M et al (2012) Dietary omega-3 supplementation exacerbates left ventricular dysfunction in an ovine model of anthracycline-induced cardiotoxicity. J Cardiac Fail 18(6):502–511CrossRef
65.
Zurück zum Zitat Bougnoux P, Hajjaji N, Ferrasson MN, Giraudeau B, Couet C, Le Floch O (2009) Improving outcome of chemotherapy of metastatic breast cancer by docosahexaenoic acid: a phase II trial. Br J Cancer 101(12):1978–1985PubMedPubMedCentralCrossRef Bougnoux P, Hajjaji N, Ferrasson MN, Giraudeau B, Couet C, Le Floch O (2009) Improving outcome of chemotherapy of metastatic breast cancer by docosahexaenoic acid: a phase II trial. Br J Cancer 101(12):1978–1985PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat Madmani ME, Yusuf Solaiman A, Tamr Agha K, Madmani Y, Shahrour Y, Essali A et al (2014) Coenzyme Q10 for heart failure. Cochrane Database Syst Rev (6):Cd008684 Madmani ME, Yusuf Solaiman A, Tamr Agha K, Madmani Y, Shahrour Y, Essali A et al (2014) Coenzyme Q10 for heart failure. Cochrane Database Syst Rev (6):Cd008684
67.
Zurück zum Zitat Folkers K, Liu M, Watanabe T, Porter TH (1977) Inhibition by adriamycin of the mitochondrial biosynthesis of coenzyme Q10 and implication for the cardiotoxicity of adriamycin in cancer patients. Biochem Biophys Res Commun 77(4):1536–1542PubMedCrossRef Folkers K, Liu M, Watanabe T, Porter TH (1977) Inhibition by adriamycin of the mitochondrial biosynthesis of coenzyme Q10 and implication for the cardiotoxicity of adriamycin in cancer patients. Biochem Biophys Res Commun 77(4):1536–1542PubMedCrossRef
68.
Zurück zum Zitat Iarussi D, Auricchio U, Agretto A, Murano A, Giuliano M, Casale F et al (1994) Protective effect of coenzyme Q10 on anthracyclines cardiotoxicity: control study in children with acute lymphoblastic leukemia and non-Hodgkin lymphoma. Mol Aspects Med 15(Suppl):s207–s212PubMedCrossRef Iarussi D, Auricchio U, Agretto A, Murano A, Giuliano M, Casale F et al (1994) Protective effect of coenzyme Q10 on anthracyclines cardiotoxicity: control study in children with acute lymphoblastic leukemia and non-Hodgkin lymphoma. Mol Aspects Med 15(Suppl):s207–s212PubMedCrossRef
69.
Zurück zum Zitat Chen PY, Hou CW, Shibu MA, Day CH, Pai P, Liu ZR et al (2017) Protective effect of Co-enzyme Q10 On doxorubicin-induced cardiomyopathy of rat hearts. Environ Toxicol 32(2):679–689PubMedCrossRef Chen PY, Hou CW, Shibu MA, Day CH, Pai P, Liu ZR et al (2017) Protective effect of Co-enzyme Q10 On doxorubicin-induced cardiomyopathy of rat hearts. Environ Toxicol 32(2):679–689PubMedCrossRef
70.
Zurück zum Zitat Najafi M, Hooshangi Shayesteh MR, Mortezaee K, Farhood B, Haghi-Aminjan H (2020) The role of melatonin on doxorubicin-induced cardiotoxicity: a systematic review. Life Sci 241:117173PubMedCrossRef Najafi M, Hooshangi Shayesteh MR, Mortezaee K, Farhood B, Haghi-Aminjan H (2020) The role of melatonin on doxorubicin-induced cardiotoxicity: a systematic review. Life Sci 241:117173PubMedCrossRef
71.
Zurück zum Zitat Myers C, Bonow R, Palmeri S, Jenkins J, Corden B, Locker G et al (1983) A randomized controlled trial assessing the prevention of doxorubicin cardiomyopathy by N-acetylcysteine. Semin Oncol 10(1 Suppl 1):53–55PubMed Myers C, Bonow R, Palmeri S, Jenkins J, Corden B, Locker G et al (1983) A randomized controlled trial assessing the prevention of doxorubicin cardiomyopathy by N-acetylcysteine. Semin Oncol 10(1 Suppl 1):53–55PubMed
72.
Zurück zum Zitat Waldner R, Laschan C, Lohninger A, Gessner M, Tuchler H, Huemer M et al (2006) Effects of doxorubicin-containing chemotherapy and a combination with L-carnitine on oxidative metabolism in patients with non-Hodgkin lymphoma. J Cancer Res Clin Oncol 132(2):121–128PubMedCrossRef Waldner R, Laschan C, Lohninger A, Gessner M, Tuchler H, Huemer M et al (2006) Effects of doxorubicin-containing chemotherapy and a combination with L-carnitine on oxidative metabolism in patients with non-Hodgkin lymphoma. J Cancer Res Clin Oncol 132(2):121–128PubMedCrossRef
73.
Zurück zum Zitat Gallegos-Castorena S, Martinez-Avalos A, Mohar-Betancourt A, Guerrero-Avendano G, Zapata-Tarres M, Medina-Sanson A (2007) Toxicity prevention with amifostine in pediatric osteosarcoma patients treated with cisplatin and doxorubicin. Pediatr Hematol Oncol 24(6):403–408PubMedCrossRef Gallegos-Castorena S, Martinez-Avalos A, Mohar-Betancourt A, Guerrero-Avendano G, Zapata-Tarres M, Medina-Sanson A (2007) Toxicity prevention with amifostine in pediatric osteosarcoma patients treated with cisplatin and doxorubicin. Pediatr Hematol Oncol 24(6):403–408PubMedCrossRef
74.
Zurück zum Zitat Yue TL, Cheng HY, Lysko PG, McKenna PJ, Feuerstein R, Gu JL et al (1992) Carvedilol, a new vasodilator and beta adrenoceptor antagonist, is an antioxidant and free radical scavenger. Journal Pharmacol Exp Ther 263(1):92–98 Yue TL, Cheng HY, Lysko PG, McKenna PJ, Feuerstein R, Gu JL et al (1992) Carvedilol, a new vasodilator and beta adrenoceptor antagonist, is an antioxidant and free radical scavenger. Journal Pharmacol Exp Ther 263(1):92–98
75.
Zurück zum Zitat Santos DL, Moreno AJ, Leino RL, Froberg MK, Wallace KB (2002) Carvedilol protects against doxorubicin-induced mitochondrial cardiomyopathy. Toxicol Appl Pharmacol 185(3):218–227PubMedCrossRef Santos DL, Moreno AJ, Leino RL, Froberg MK, Wallace KB (2002) Carvedilol protects against doxorubicin-induced mitochondrial cardiomyopathy. Toxicol Appl Pharmacol 185(3):218–227PubMedCrossRef
76.
Zurück zum Zitat Oliveira PJ, Goncalves L, Monteiro P, Providencia LA, Moreno AJ (2005) Are the antioxidant properties of carvedilol important for the protection of cardiac mitochondria? Curr Vasc Pharmacol 3(2):147–158PubMedCrossRef Oliveira PJ, Goncalves L, Monteiro P, Providencia LA, Moreno AJ (2005) Are the antioxidant properties of carvedilol important for the protection of cardiac mitochondria? Curr Vasc Pharmacol 3(2):147–158PubMedCrossRef
77.
Zurück zum Zitat Kheiri B, Abdalla A, Osman M, Haykal T, Chahine A, Ahmed S et al (2018) Meta-analysis of carvedilol for the prevention of anthracycline-induced cardiotoxicity. Am J Cardiol 122(11):1959–1964PubMedCrossRef Kheiri B, Abdalla A, Osman M, Haykal T, Chahine A, Ahmed S et al (2018) Meta-analysis of carvedilol for the prevention of anthracycline-induced cardiotoxicity. Am J Cardiol 122(11):1959–1964PubMedCrossRef
78.
Zurück zum Zitat Bosch X, Rovira M, Sitges M, Domenech A, Ortiz-Perez JT, de Caralt TM et al (2013) Enalapril and carvedilol for preventing chemotherapy-induced left ventricular systolic dysfunction in patients with malignant hemopathies: the OVERCOME trial (preventiOn of left Ventricular dysfunction with Enalapril and caRvedilol in patients submitted to intensive ChemOtherapy for the treatment of Malignant hEmopathies). J Am Coll Cardiol 61(23):2355–2362PubMedCrossRef Bosch X, Rovira M, Sitges M, Domenech A, Ortiz-Perez JT, de Caralt TM et al (2013) Enalapril and carvedilol for preventing chemotherapy-induced left ventricular systolic dysfunction in patients with malignant hemopathies: the OVERCOME trial (preventiOn of left Ventricular dysfunction with Enalapril and caRvedilol in patients submitted to intensive ChemOtherapy for the treatment of Malignant hEmopathies). J Am Coll Cardiol 61(23):2355–2362PubMedCrossRef
79.
Zurück zum Zitat Chopra M, Beswick H, Clapperton M, Dargie HJ, Smith WE, McMurray J (1992) Antioxidant effects of angiotensin-converting enzyme (ACE) inhibitors: free radical and oxidant scavenging are sulfhydryl dependent, but lipid peroxidation is inhibited by both sulfhydryl- and nonsulfhydryl-containing ACE inhibitors. J Cardiovasc Pharmacol 19(3):330–340PubMedCrossRef Chopra M, Beswick H, Clapperton M, Dargie HJ, Smith WE, McMurray J (1992) Antioxidant effects of angiotensin-converting enzyme (ACE) inhibitors: free radical and oxidant scavenging are sulfhydryl dependent, but lipid peroxidation is inhibited by both sulfhydryl- and nonsulfhydryl-containing ACE inhibitors. J Cardiovasc Pharmacol 19(3):330–340PubMedCrossRef
80.
81.
Zurück zum Zitat Hiona A, Lee AS, Nagendran J, Xie X, Connolly AJ, Robbins RC et al (2011) Pretreatment with angiotensin-converting enzyme inhibitor improves doxorubicin-induced cardiomyopathy via preservation of mitochondrial function. J Thorac Cardiovasc Surg 142(2):396-403.e3PubMedCrossRef Hiona A, Lee AS, Nagendran J, Xie X, Connolly AJ, Robbins RC et al (2011) Pretreatment with angiotensin-converting enzyme inhibitor improves doxorubicin-induced cardiomyopathy via preservation of mitochondrial function. J Thorac Cardiovasc Surg 142(2):396-403.e3PubMedCrossRef
82.
Zurück zum Zitat Heck SL, Gulati G, Hoffmann P, von Knobelsdorff-Brenkenhoff F, Storås TH, Ree AH et al (2018) Effect of candesartan and metoprolol on myocardial tissue composition during anthracycline treatment: the PRADA trial. Eur Heart J Cardiovasc Imaging 19(5):544–552PubMedCrossRef Heck SL, Gulati G, Hoffmann P, von Knobelsdorff-Brenkenhoff F, Storås TH, Ree AH et al (2018) Effect of candesartan and metoprolol on myocardial tissue composition during anthracycline treatment: the PRADA trial. Eur Heart J Cardiovasc Imaging 19(5):544–552PubMedCrossRef
83.
Zurück zum Zitat Sheta A, Elsakkar M, Hamza M, Solaiman A (2016) Effect of metformin and sitagliptin on doxorubicin-induced cardiotoxicity in adult male albino rats. Hum Exp Toxicol 35(11):1227–1239PubMedCrossRef Sheta A, Elsakkar M, Hamza M, Solaiman A (2016) Effect of metformin and sitagliptin on doxorubicin-induced cardiotoxicity in adult male albino rats. Hum Exp Toxicol 35(11):1227–1239PubMedCrossRef
84.
Zurück zum Zitat Peng W, Rao D, Zhang M, Shi Y, Wu J, Nie G et al (2019) Teneligliptin prevents doxorubicin-induced inflammation and apoptosis in H9c2 cells. Arch Biochem Biophys 683:108238PubMedCrossRef Peng W, Rao D, Zhang M, Shi Y, Wu J, Nie G et al (2019) Teneligliptin prevents doxorubicin-induced inflammation and apoptosis in H9c2 cells. Arch Biochem Biophys 683:108238PubMedCrossRef
85.
Zurück zum Zitat Sabatino J, De Rosa S, Tammè L, Iaconetti C, Sorrentino S, Polimeni A et al (2020) Empagliflozin prevents doxorubicin-induced myocardial dysfunction. Cardiovasc Diabetol 19(1):66PubMedPubMedCentralCrossRef Sabatino J, De Rosa S, Tammè L, Iaconetti C, Sorrentino S, Polimeni A et al (2020) Empagliflozin prevents doxorubicin-induced myocardial dysfunction. Cardiovasc Diabetol 19(1):66PubMedPubMedCentralCrossRef
86.
Zurück zum Zitat Sheibani M, Nezamoleslami S, Faghir-Ghanesefat H, Emami AH, Dehpour AR (2020) Cardioprotective effects of dapsone against doxorubicin-induced cardiotoxicity in rats. Cancer Chemother Pharmacol 85(3):563–571PubMedCrossRef Sheibani M, Nezamoleslami S, Faghir-Ghanesefat H, Emami AH, Dehpour AR (2020) Cardioprotective effects of dapsone against doxorubicin-induced cardiotoxicity in rats. Cancer Chemother Pharmacol 85(3):563–571PubMedCrossRef
87.
Zurück zum Zitat Seicean S, Seicean A, Plana JC, Budd GT, Marwick TH (2012) Effect of statin therapy on the risk for incident heart failure in patients with breast cancer receiving anthracycline chemotherapy: an observational clinical cohort study. J Am Coll Cardiol 60(23):2384–2390PubMedCrossRef Seicean S, Seicean A, Plana JC, Budd GT, Marwick TH (2012) Effect of statin therapy on the risk for incident heart failure in patients with breast cancer receiving anthracycline chemotherapy: an observational clinical cohort study. J Am Coll Cardiol 60(23):2384–2390PubMedCrossRef
Metadaten
Titel
Oxidative stress and inflammation: determinants of anthracycline cardiotoxicity and possible therapeutic targets
verfasst von
Iacopo Fabiani
Alberto Aimo
Chrysanthos Grigoratos
Vincenzo Castiglione
Francesco Gentile
Luigi F Saccaro
Chiara Arzilli
Daniela Cardinale
Claudio Passino
Michele Emdin
Publikationsdatum
15.12.2020
Verlag
Springer US
Erschienen in
Heart Failure Reviews / Ausgabe 4/2021
Print ISSN: 1382-4147
Elektronische ISSN: 1573-7322
DOI
https://doi.org/10.1007/s10741-020-10063-9

Weitere Artikel der Ausgabe 4/2021

Heart Failure Reviews 4/2021 Zur Ausgabe

Nach Herzinfarkt mit Typ-1-Diabetes schlechtere Karten als mit Typ 2?

29.05.2024 Herzinfarkt Nachrichten

Bei Menschen mit Typ-2-Diabetes sind die Chancen, einen Myokardinfarkt zu überleben, in den letzten 15 Jahren deutlich gestiegen – nicht jedoch bei Betroffenen mit Typ 1.

Erhöhtes Risiko fürs Herz unter Checkpointhemmer-Therapie

28.05.2024 Nebenwirkungen der Krebstherapie Nachrichten

Kardiotoxische Nebenwirkungen einer Therapie mit Immuncheckpointhemmern mögen selten sein – wenn sie aber auftreten, wird es für Patienten oft lebensgefährlich. Voruntersuchung und Monitoring sind daher obligat.

GLP-1-Agonisten können Fortschreiten diabetischer Retinopathie begünstigen

24.05.2024 Diabetische Retinopathie Nachrichten

Möglicherweise hängt es von der Art der Diabetesmedikamente ab, wie hoch das Risiko der Betroffenen ist, dass sich sehkraftgefährdende Komplikationen verschlimmern.

TAVI versus Klappenchirurgie: Neue Vergleichsstudie sorgt für Erstaunen

21.05.2024 TAVI Nachrichten

Bei schwerer Aortenstenose und obstruktiver KHK empfehlen die Leitlinien derzeit eine chirurgische Kombi-Behandlung aus Klappenersatz plus Bypass-OP. Diese Empfehlung wird allerdings jetzt durch eine aktuelle Studie infrage gestellt – mit überraschender Deutlichkeit.

Update Kardiologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.