Cardiotoxicity associated with breast cancer treatment is an important concern in the oncology clinic. Different types of anti-cancer therapies have recorded high rates of cardiac dysfunction in treated patients. Cardiac dysfunction linked to anthracyclines – one of the most common conventional chemotherapies – has extensively been described and several mechanisms have been proposed, although their mode of action is not fully understood even in cancer cells. The mediation of cardiac damage by reactive oxygen species stress is a recent hypothesis that has attracted a lot of interest, since it might explain the tissue-specific toxic effects of anthracyclines in the heart. Regarding molecular targeted tyrosine kinase inhibitors used in patients with human epidermal growth factor receptor type 2+ tumours (e.g., trastuzumab, lapatinib), it is the blockage of survival pathways required for a normal heart development and function that seems to lead to cardiac pathology. Both types of breast cancer treatment appear to trigger cardiotoxicity synergically, being patients under adjuvant therapy closely monitored. Given the complex nature of heart failure and of the pathways altered by anti-cancer drugs, global gene expression regulation is key in the heart disease process. MicroRNAs have been demonstrated to be small molecules with big roles as essential gene expression modulators. The great potential of microRNAs as biomarkers in the cardio-oncology field needs to be further explored before new microRNA-based diagnostic and therapeutic tools can be developed.

Breast cancer is the commonest cancer in females, being responsible for 16% of all cancer cases worldwide and for 31% in the UK. The available treatment options can be classified into two groups: conventional therapies (i.e. chemotherapy and radiotherapy) which aim to have a greater effect on cancer cells without being specifically directed to them, and newer molecular targeted therapies, which are designed to recognize determined cancer cell markers in order to spare normal cells [1,2].

Doxorubicin (DOX), also known by its commercial name Adriamycin, was the first anthracycline (ANT) antibiotic to be isolated from Streptomyces peucetius during the 1960s, together with danorubicin. Since its discovery and despite constant efforts to develop improved ANTs, DOX is still the most widely used antibiotic in the clinic, providing high anti-tumour efficacy against numerous types of cancer [3]. As a consequence of the lack of specificity, patients receiving DOX treatment present several adverse effects, with cardiomyopathy leading to heart failure (HF) being the main long-term complication [4,5]. This serious side effect has been shown to remain as a late-onset problem, affecting patients up to 20 years after completion of ANT treatment in a long-term follow-up study [6].

The exact mechanisms of anti-cancer activity of ANTs are poorly understood and further research is needed before a well-defined mode of action can be agreed. Current evidence suggests that it is of a multifactorial and complex nature [7], with several cellular processes having been described as affected by DOX: (1) inhibition of DNA and RNA synthesis through intercalation in nucleic acids [8,9]; (2) generation of reactive free radicals after DOX-redox cycling [10,11]; (3) interference with DNA helicases [12]; (4) induction of DNA strand breaks through type II topoisomerase inhibition [7,11,13]; (5) apoptosis, p53 dependent or independent [14]; (6) growth arrest [15].

In a retrospective study including 399 DOX-treated patients, HF was diagnosed even in patients without a previous history of cardiac conditions. It affected 4% of the patients treated with 500–550 mg/m2 DOX, 18% of those treated with 551–600 mg/m2 DOX and 36% of patients treated with higher doses. Due to the appearance of adverse cardiac events in a dose-dependent manner, 500 mg/m2 has been established as the maximum safe dose of DOX [16]. Apart from determining the concentration limits to ensure the safest possible use of DOX, a variety of prevention and management measures are followed for cancer patients undertaking ANT-based regimes [16,17]. Early diagnosis of DOX-induced cardiotoxicity is essential, since once patients develop advanced-stage HF, their prognosis is not favorable. This is due to the lack of specific available treatments, given that DOX-induced cardiac damage cannot be reverted by conventional therapies [18].

In terms of cardioprotection, new compounds have been tested with the aim of more specific drug delivery. In recent years, pegylated liposomal DOX has emerged as an interesting and effective alternative to conventional DOX, presenting a different pharmacokinetic profile and reduced cardiotoxicity in clinical trials [19,20]. The myocardium is less affected by these modified ANTs because larger lipidic complexes are not able to diffuse through the heart and other tissues, releasing most free DOX into the tumour, which has a looser endothelium [21]. Even more specific variants of DOX are currently under investigation, i.e. the recognition of estrogen receptor, one of the strategies for a targeted delivery [22].

In contrast to fast-dividing cancer cells, adult cardiomyocytes are quiescent cells. This important fact suggests that the mechanisms underlying DOX tumour and cardiac cytotoxicity are likely to be different. The existence of differential mechanisms of cell death induction in cancer cells and in the myocardium would facilitate the development of less cardiotoxic ANTs [17].

The generation of free radicals seems to play a vital role in DOX-associated cardiotoxicity [10,23,24,25]. Although mammalian tissues have protective resources against oxidative stress, the heart has been shown to present remarkably low levels of enzymes such as catalase, superoxide dismutase and glutathione peroxidase, all of which help minimizing reactive oxygen species (ROS) toxicity. The reduced levels of antioxidant enzymes in the heart in comparison to other tissues may explain the tissue-specific toxicity associated with DOX [26].

Cardiomyocytes are very rich in mitochondria, which contain enzymes that mediate a 1-electron reduction in ANTs into a free radical semiquinone form shortly after they enter the cell. This reaction produces high concentrations of ROS, such as superoxide anion (O2) or hydrogen peroxide (H2O2) [3]. Some research suggests that DOX not only generates new damaging free radicals, but also contributes to a decrease in endogenous antioxidant levels. Altogether, this leads to oxidative stress that will cause cytotoxicity through multiple mechanisms (fig. 1) [26,27,28].

Fig. 1

Oxidative stress causing adverse effects in cardiomyocytes following DOX metabolism. After redox cycling of ANTs (i.e. DOX) in the mitochondria, the generation of free oxygen radicals damages the cell through diverse mechanisms. These include: DNA damage, alteration of gene expression, sarcomere degradation with deregulation of Ca2+ levels, and mitochondrial dysfunction [21].

Fig. 1

Oxidative stress causing adverse effects in cardiomyocytes following DOX metabolism. After redox cycling of ANTs (i.e. DOX) in the mitochondria, the generation of free oxygen radicals damages the cell through diverse mechanisms. These include: DNA damage, alteration of gene expression, sarcomere degradation with deregulation of Ca2+ levels, and mitochondrial dysfunction [21].

Close modal

The explanation of DOX-induced cardiotoxicity through oxidative stress pathways has become a very popular hypothesis in the last decade and has been supported by several studies demonstrating cardioprotective effects when administering antioxidants in combination with DOX in vivo, without interfering with anti-tumour efficacy [25,27,29].

There is a considerable amount of evidence indicating that mitochondria are highly susceptible to damage by generation of superoxide radicals. Mitochondrial dysfunction has been shown after DOX treatment in vitro, with cytochrome c release leading to caspase-9-mediated caspase 3 activation and resulting in apoptosis [30,31]. The fact that a p53–/– mouse model did not fully evade DOX-induced cardiac damage supports the idea that ROS might be able to directly trigger p53-independent apoptotic pathways [32].

Advances in medical knowledge and new technologies have enabled the development of molecular targeted therapies, the main advantage of which is that they are more specifically directed to cancer cells, being thus less harmful for healthy cells. In the context of breast cancer, the human epidermal growth factor receptor type 2 (HER2), also known as erbB2 [v-erb-b2 erythroblastic leukemia viral oncogene homolog 2, neuro/glioblastoma derived oncogene homolog (avian)], was described to be overexpressed in 20–30% of the patients. HER2 overexpression was associated with particularly aggressive forms of disease and poorer prognosis. Since this was recorded, HER2 has attracted many efforts in drug discovery for breast cancer therapy [33,34,35].

Different types of tyrosine kinase inhibitors have successfully reached the market and are approved by the Food and Drug Administration. Trastuzumab and lapatinib are the two most studied and clinically applied. Trastuzumab (Herceptin®) is a humanized monoclonal antibody that binds to the extracellular domain of the HER2, impairing its activation and downstream cascade of survival signals. Apart from the direct inhibition of the HER2 pathway, trastuzumab is thought to contribute to tumour cell death also via antibody-directed cell cytotoxicity [36]. On the other hand, lapatinib (Tyverb®) is a small molecule capable of inhibiting both HER2 and epidermal growth factor receptor (so-called HER1) by interacting with their intracellular tyrosine kinase (tyrK) domain (fig. 2). HER1 and HER2 overexpressing tumours undergo apoptosis or growth arrest after treatment with lapatinib [37].

Fig. 2

Trastuzumab and lapatinib inhibition of the HER family members. HER2 belongs to a family of growth factor receptors with intracellular tyrK domains (HER1–HER4). Although HER2 is the only member that does not have a known ligand, it forms dimers with other HERs acting as a co-receptor. The humanized monoclonal antibody trastuzumab recognizes the extracellular domain of HER2, preventing dimerization with other members. The small inhibitor lapatinib blocks the tyrK domains of both HER2 and HER1 (epidermal growth factor receptor, EGFR). The subsequent inhibition of the downstream signalling pathway prevents the translocation into the nucleus of the activated factors that usually stimulate transcription of survival and proliferation-promoting genes.

Fig. 2

Trastuzumab and lapatinib inhibition of the HER family members. HER2 belongs to a family of growth factor receptors with intracellular tyrK domains (HER1–HER4). Although HER2 is the only member that does not have a known ligand, it forms dimers with other HERs acting as a co-receptor. The humanized monoclonal antibody trastuzumab recognizes the extracellular domain of HER2, preventing dimerization with other members. The small inhibitor lapatinib blocks the tyrK domains of both HER2 and HER1 (epidermal growth factor receptor, EGFR). The subsequent inhibition of the downstream signalling pathway prevents the translocation into the nucleus of the activated factors that usually stimulate transcription of survival and proliferation-promoting genes.

Close modal

Both trastuzumab and lapatinib are able to block the same signalling pathway – mediated by Akt and MAP kinases (ERK1/2) – and during respective clinical trials proved to be efficient anti-cancer treatments for breast cancer patients. Maximum therapeutic results were achieved when combining them with ANT-based chemotherapy. In fact, HER2 blockers are normally used as chemotherapy adjuvants for second-line therapy [37,38].

In spite of inhibiting the same pathway, different rates of cardiotoxicity are observed upon trastuzumab and lapatinib treatment. Although some cardiac events were recorded, lapatinib was better tolerated than trastuzumab during clinical trials [37,39]. Regarding ANT-combined regimes, 27% of patients under trastuzumab + DOX developed cardiac dysfunction, while the incidence for lapatinib + DOX remained as low as 2.2% [38,40]. The mechanisms responsible for this difference are not yet well understood, but are likely to be related to the nature of each molecule. Although the antibody-directed cell cytotoxic activity of trastuzumab is the main obvious differential mode of action, it does not seem to be responsible for the higher cardiotoxicity. An intriguing mechanism to explore is the induction of mitochondrial dysfunction by trastuzumab while lapatinib seems to have a protective effect on such organelle [41,42]. This would be an interesting hypothesis to link to the mitochondrial dysfunction triggered by DOX in a putative synergic way, given that combined therapy results in higher cardiotoxicity risk.

Neuregulins are an essential family of growth factors during development which bind to HER3 and HER4. Neuregulin-1 (NRG1, also known as heregulin or neu differentiation factor) has been particularly described to be required for an adequate heart formation and function [43,44]. Moreover, the lack of erbB2 (HER2) and erbB4 (HER4) receptors is also associated with the same cardiac defects during embryogenesis [45,46,47]. Given the requirement of a functional HER2 in the heart and the stimulation of HER2-HER3/4 dimerization by NRG1, administration of such growth factor has been considered as an option to counteract HER2-targeted therapy toxicity and prevent HF [48,49]. Interestingly, NRG1 administration to adult rat cardiomyocytes was protective of DOX-induced toxicity by reducing oxidative stress. This may explain why an increased cardiotoxicity is recorded when combining ANT-based chemotherapy with HER2 inhibitors (fig. 3) [50].

Fig. 3

Cardiotoxicity of DOX and HER2 inhibitors in combination. a When DOX is administered alone, HER2 is active and able to dimerize with other HER members upon recognition of NRG1, subsequently triggering survival-promoting pathways in the cell. b When trastuzumab (TRZ) or lapatinib (LAP) are combined with DOX, HER2/NRG1-mediated counteraction of cardiac toxicity is lost.

Fig. 3

Cardiotoxicity of DOX and HER2 inhibitors in combination. a When DOX is administered alone, HER2 is active and able to dimerize with other HER members upon recognition of NRG1, subsequently triggering survival-promoting pathways in the cell. b When trastuzumab (TRZ) or lapatinib (LAP) are combined with DOX, HER2/NRG1-mediated counteraction of cardiac toxicity is lost.

Close modal

MicroRNAs (miRNAs) are abundant endogenous short (approximately 22 nucleotides, nt) non-coding RNA molecules that play an important and evolutionary conserved regulatory role in gene expression, affecting up to one third of the human genome [51]. miRNAs act mainly at a post-transcriptional level, basepairing with the 3′ untranslated region of target messenger RNAs [52]. The resulting impairment of protein synthesis from target messenger RNAs will determine different effects in key biological processes such as development, differentiation, metabolism, proliferation and growth [53].

Regardless of the primary cause – including toxicity derived from cancer treatment – most cardiac conditions convert to HF in the later stage. Generally, progression to HF starts with remodelling of the ventricles – mainly the left ventricle – in order to achieve a functional compensation in the damaged heart. This process is tightly regulated and involves important changes in gene expression, such as re-activation of fetal genes (e.g., the switch from the α to the fetal β form of the myosin heavy chain), and myocyte hypertrophic growth [54]. Eventually, this compensatory hypertrophy causes myocardial wall stress and contractile dysfunction, leading to a final stage of failure where the heart is not able to pump enough blood to the organism. Importantly, also other cardiac cell types like fibroblasts and endothelial cells play an active role in cardiac remodelling [55]. It has only recently been described how important cardiomyocyte loss through apoptosis is in the development of HF, suggesting that there are still numerous relevant mechanisms involved to be yet elucidated [56].

Although miRNA implications have been extensively studied in cancer, the importance of these short regulatory RNAs within the cardiovascular system has only been highlighted in recent years [57]. Spatiotemporal expression of miRNA in the different tissues has been demonstrated to influence development and evolution, and the heart is not an exception [58]. In fact, impairment of miRNA biogenesis is demonstrated to result in embryonic lethality with poor myocardium formation [59]. The contribution of miRNA to a normal heart development and function is not surprising, since cardiogenesis, ventricular remodelling and HF are all highly complex processes. miRNAs constitute an additional level of gene expression regulation that has a high impact on the gene expression pattern of both the healthy and diseased heart, modulating aberrant signatures involved in heart disease (fig. 4).

Fig. 4

Progression of HF. Pathological hypertrophy is the result of ventricular remodelling, which follows damage to the heart through a variety of injuries (ischaemia, pressure/volume overload, myocardial infarction, exposure to toxic agents, and others). After this hypertrophic phase, the enlarged myocardium starts losing viable cardiomyocytes, being affected by progressive fibrosis and failing to contract properly. All these events finally lead to HF. Gene expression regulation is key during the pathological process of HF, and miRNA signatures vary from the healthy heart as the disease progresses, being essential regulatory elements.

Fig. 4

Progression of HF. Pathological hypertrophy is the result of ventricular remodelling, which follows damage to the heart through a variety of injuries (ischaemia, pressure/volume overload, myocardial infarction, exposure to toxic agents, and others). After this hypertrophic phase, the enlarged myocardium starts losing viable cardiomyocytes, being affected by progressive fibrosis and failing to contract properly. All these events finally lead to HF. Gene expression regulation is key during the pathological process of HF, and miRNA signatures vary from the healthy heart as the disease progresses, being essential regulatory elements.

Close modal

Different studies have recently identified particular miRNA signatures associated with several cardiac conditions, although the majority of research focuses on hypertrophy and HF. Van Rooij [60], for instance, described an miRNA expression pattern in the heart under stress. On the other hand, Thum et al. [61] showed a miRNA sub-signature shared by the fetal and the failing heart, which correlates with the re-expression of fetal genes in the failing heart. Another example of research in the field is a big profiling study by Ikeda et al. [62] of different types of heart disease where altered miRNA expression in the dysfunctional myocardium was confirmed for humans. The most studied and well-characterized miRNAs in the heart are: (1) miR-1 (the most abundant cardiac miRNA), described to be involved in arrhythmias and down-regulated in HF patients [63,64]; (2) miR-133 and miR-195 and their role in hypertrophy [60,65,66]; and (3) miR-208, which is encoded in an intronic region of the α form of the myosin heavy chain and governs a feedback loop that results in impaired contractility when deregulated [67]. Other miRNAs such as miR-29 and miR-499 have also been shown to have relevant functions associated with heart disease [68,69]. Not much is known regarding miRNA implications in the particular case of heart disease following cancer treatment. So far, only one paper reports the up-regulation of an miRNA (miR-146a) upon DOX treatment in cardiomyocytes, being cells more resistant to DOX when artificially reducing miR-146a expression in vitro [70].

All the studies proving detrimental consequences in cardiac function upon up-/down-regulation of certain miRNAs have highlighted the potential of miRNAs both as diagnostic and therapeutic tools. Moreover, the fact that these small RNAs seem to be actively secreted to and stable in the bloodstream has attracted a lot of interest in trying to use circulating miRNAs as disease biomarkers [71]. This would be highly beneficial for patients with cardiovascular conditions, since it would allow a rapid and non-invasive diagnosis and monitoring. In terms of therapeutic applications, the main challenge is engineering both systemic stability and, most importantly, specificity of the miRNA-targeting drug, given that the same molecule is likely to have different effects depending on the tissue. Furthermore, miRNAs are known to have multiple targets. Although this may lead to a synergic therapeutic effect when an miRNA governs the expression of several genes of the same pathway, the benefit might be lost to ‘off-target’ effects [72]. These issues need to be carefully addressed before miRNA-based therapies can reach the market for safe clinical applications.

1.
World Health Organization website. www.who.ing/cancer (May 2012).
2.
Cancer Research UK website. www.cancerresearchuk.org (May 2012).
3.
Minotti G, et al: Anthracyclines: molecular advances and pharmacologic developments in antitumor activity and cardiotoxicity. Pharmacol Rev 2004;56:185–229.
4.
Lefrak EA, et al: A clinicopathologic analysis of adriamycin cardiotoxicity. Cancer 1973;32:302–314.
5.
Buja LM, et al: Cardiac ultrastructural changes induced by daunorubicin therapy. Cancer 1973;32:771–788.
6.
Steinherz LJ, et al: Cardiac toxicity 4 to 20 years after completing anthracycline therapy. JAMA 1991;266:1672–1677.
7.
David AG: 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.
8.
Goodman MF, Bessman MJ, Bachur NR: Adriamycin and daunorubicin inhibition of mutant T4 DNA polymerases. Proc Natl Acad Sci USA 1974;71:1193–1196.
9.
Momparler RL, et al: Effect of adriamycin on DNA, RNA, and protein synthesis in cell-free systems and intact cells. Cancer Res 1976;36:2891–2895.
10.
Davies KJ, Doroshow JH: Redox cycling of anthracyclines by cardiac mitochondria. 1. Anthracycline radical formation by NADH dehydrogenase. J Biol Chem 1986;261:3060–3067.
11.
Fornari FA, et al: Interference by doxorubicin with DNA unwinding in MCF-7 breast tumor cells. Mol Pharmacol 1994;45:649–656.
12.
Bachur NR, et al: Antihelicase action of DNA-binding anticancer agents: relationship to guanosine-cytidine intercalator binding. Mol Pharmacol 1993;44:1064–1069.
13.
D’Arpa P, Liu LF: Topoisomerase-targeting antitumor drugs. Biochim Biophys Acta 1989;989:163–177.
14.
Skladanowski A, Konopa J: Adriamycin and daunomycin induce programmed cell death (apoptosis) in tumour cells. Biochem Pharmacol 1993;46:375–382.
15.
Ling YH, et al: Cell cycle-dependent cytotoxicity, G2/M phase arrest, and disruption of p34cdc2/cyclin B1 activity induced by doxorubicin in synchronized P388 cells. Mol Pharmacol 1996;49:832–841.
16.
Singal PK, Iliskovic N: Doxorubicin-induced cardiomyopathy. N Engl J Med 1998;339:900–905.
17.
Takemura G, Fujiwara H: Doxorubicin-induced cardiomyopathy: from the cardiotoxic mechanisms to management. Prog Cardiovasc Dis 2007;49:330–352.
18.
Hjalmarson A, Waagstein F: The role of [beta]-blockers in the treatment of cardiomyopathy and ischaemic heart failure. Drugs 1994;47:31–40.
19.
Alberts DS, et al: Efficacy and safety of liposomal anthracyclines in phase I/II clinical trials. Semin Oncol 2004;31(suppl 13):53–90.
20.
Robert NJ, et al: The role of the liposomal anthracyclines and other systemic therapies in the management of advanced breast cancer. Semin Oncol 2004;31(suppl 13):106–146.
21.
Menna P, et al: Anthracycline cardiotoxicity. Expert Opin Drug Safe 2012;11:S21–S36.
22.
Burke PJ, Koch TH: Design, synthesis, and biological evaluation of doxorubicin-formaldehyde conjugates targeted to breast cancer cells. J Med Chem 2004;47:1193–1206.
23.
Doroshow JH: Effect of anthracycline antibiotics on oxygen radical formation in rat heart. Cancer Res 1983;43:460–472.
24.
Velez JM, et al: p53 regulates oxidative stress-mediated retrograde signaling: a novel mechanism for chemotherapy-induced cardiac injury. PLoS ONE 2011;6:e18005.
25.
Yen HC, et al: The protective role of manganese superoxide dismutase against adriamycin-induced acute cardiac toxicity in transgenic mice. J Clin Invest 1996;98:1253–1260.
26.
Doroshow JH, Locker GY, Myers CE: Enzymatic defenses of the mouse heart against reactive oxygen metabolites: alterations produced by doxorubicin. J Clin Invest 1980;65:128–135.
27.
Siveski-Iliskovic N, Kaul N, Singal P: Probucol promotes endogenous antioxidants and provides protection against adriamycin-induced cardiomyopathy in rats. Circulation 1994;89:2829–2835.
28.
Kalyanaraman B, et al: Doxorubicin-induced apoptosis: implications in cardiotoxicity. Mol Cell Biochem 2002;234–235:119–124.
29.
Singal PK, et al: Combination therapy with probucol prevents adriamycin-induced cardiomyopathy. J Mol Cell Cardiol 1995;27:1055–1063.
30.
Green PS, Leeuwenburgh C: Mitochondrial dysfunction is an early indicator of doxorubicin-induced apoptosis. Biochim Biophys Acta 2002;1588:94–101.
31.
Childs AC, et al: Doxorubicin treatment in vivo causes cytochrome c release and cardiomyocyte apoptosis, as well as increased mitochondrial efficiency, superoxide dismutase activity, and Bcl-2:Bax ratio. Cancer Res 2002;62:4592–4598.
32.
Feridooni T, et al: Cardiomyocyte specific ablation of p53 is not sufficient to block doxorubicin induced cardiac fibrosis and associated cytoskeletal changes. PLoS One 2011;6:e22801.
33.
Slamon D, et al: Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science 1987;235:177–182.
34.
Slamon D, et al: Studies of the HER-2/neu proto-oncogene in human breast and ovarian cancer. Science 1989;244:707–712.
35.
Olayioye MA, et al: The ErbB signaling network: receptor heterodimerization in development and cancer. EMBO J 2000;19:3159–3167.
36.
Force T, Krause DS, Van Etten RA: Molecular mechanisms of cardiotoxicity of tyrosine kinase inhibition. Nat Rev Cancer 2007;7:332–344.
37.
Burris HA, et al: Phase I safety, pharmacokinetics, and clinical activity study of lapatinib (GW572016), a reversible dual inhibitor of epidermal growth factor receptor tyrosine kinases, in heavily pretreated patients with metastatic carcinomas. J Clin Oncol 2005;23:5305–5313.
38.
Slamon DJ, et al: Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med 2001;344:783–792.
39.
Azim H, Azim HA Jr, Escudier B: Trastuzumab versus lapatinib: the cardiac side of the story. Cancer Treat Rev 2009;35:633–638.
40.
Perez EA, et al: Cardiac safety of lapatinib: pooled analysis of 3689 patients enrolled in clinical trials. Mayo Clin Proc 2008;83:679–686.
41.
Wysocki P, Wysocki H: Cardiovascular complications associated with biological therapies for breast cancer. Expert Opin Biol Ther 2008;8:1551–1559.
42.
Grazette LP, et al: Inhibition of ErbB2 causes mitochondrial dysfunction in cardiomyocytes: implications for herceptin-induced cardiomyopathy. J Am Coll Cardiol 2004;44:2231–2238.
43.
Meyer D, Birchmeier C: Multiple essential functions of neuregulin in development. Nature 1995;378:386–390.
44.
Zhao Y-Y, et al: Neuregulins promote survival and growth of cardiac myocytes. J Biol Chem 1998;273:10261–10269.
45.
Lee K-F, et al: Requirement for neuregulin receptor erbB2 in neural and cardiac development. Nature 1995;378:394–398.
46.
Negro A, Brar BK, Lee KF: Essential roles of Her2/erbB2 in cardiac development and function. Recent Prog Horm Res 2004;59:1–12.
47.
Rohrbach S, et al: Neuregulin receptors erbB2 and erbB4 in failing human myocardium. Basic Res Cardiol 2005;100:240–249.
48.
Jiang Z, Zhou M: Neuregulin signaling and heart failure. Curr Heart Fail Rep 2010;7:42–47.
49.
Sawyer DB, Caggiano A: Neuregulin-1β for the treatment of systolic heart failure. J Mol Cell Cardiol 2011;51:501–505.
50.
Timolati F, et al: Neuregulin-1 beta attenuates doxorubicin-induced alterations of excitation-contraction coupling and reduces oxidative stress in adult rat cardiomyocytes. J Mol Cell Cardiol 2006;41:845–854.
51.
Lewis B, Burge C, Bartel D: Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 2005;120:15–20.
52.
Bartel D: MicroRNAs: target recognition and regulatory functions. Cell 2009;136:215–233.
53.
Bartel D: MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 2004;116:281–297.
54.
Creemers EE, Wilde AA, Pinto YM: Heart failure: advances through genomics. Nat Rev Genet 2011;12:357–362.
55.
Souders CA, Bowers SLK, Baudino TA: Cardiac fibroblast. Circ Res 2009;105:1164–1176.
56.
Kang PM, Izumo S: Apoptosis and heart failure: a critical review of the literature. Circ Res 2000;86:1107–1113.
57.
da Costa Martins PA, et al: Conditional dicer gene deletion in the postnatal myocardium provokes spontaneous cardiac remodeling. Circulation 2008;118:1567–1576.
58.
Farh KK, et al: The widespread impact of mammalian microRNAs on mRNA repression and evolution. Science 2005;310:1817–1821.
59.
Bernstein E, et al: Dicer is essential for mouse development. Nat Genet 2003;35:215–217.
60.
van Rooij E: A signature pattern of stress-responsive microRNAs that can evoke cardiac hypertrophy and heart failure. Proc Natl Acad Sci USA 2006;103:18255–18260.
61.
Thum T, et al: MicroRNAs in the human heart. Circulation 2007;116:258–267.
62.
Ikeda S, et al: Altered microRNA expression in human heart disease. Physiol Genom 2007;31:367–373.
63.
Yang B, et al: The muscle-specific microRNA miR-1 regulates cardiac arrhythmogenic potential by targeting GJA1 and KCNJ2. Nat Med 2007;13:486–491.
64.
Kumarswamy R, et al: SERCA2a gene therapy restores microRNA-1 expression in heart failure via an Akt/FoxO3A-dependent pathway. Eur Heart J 2012;33:1067–1075.
65.
Dong D-L, et al: Reciprocal repression between microRNA-133 and calcineurin regulates cardiac hypertrophy. Hypertension 2010;55:946–952.
66.
Care A, et al: MicroRNA-133 controls cardiac hypertrophy. Nat Med 2007;13:613–618.
67.
van Rooij E, et al: Control of stress-dependent cardiac growth and gene expression by a microRNA. Science 2007;316:575–579.
68.
van Rooij E, et al: Dysregulation of microRNAs after myocardial infarction reveals a role of miR-29 in cardiac fibrosis. Proc Natl Acad Sci USA 2008;105:13027–13032.
69.
Wang J-X, et al: miR-499 regulates mitochondrial dynamics by targeting calcineurin and dynamin-related protein-1. Nat Med 2011;17:71–78.
70.
Horie T, et al: Acute doxorubicin cardiotoxicity is associated with miR-146a-induced inhibition of the neuregulin-ErbB pathway. Cardiovasc Res 2010;87:656–664.
71.
Creemers EE, Tijsen AJ, Pinto YM: Circulating microRNAs. Circ Res 2012;110:483–495.
72.
van Rooij E, Marshall WS, Olson EN: Toward microRNA-based therapeutics for heart disease. Circ Res 2008;103:919–928.
Copyright / Drug Dosage / Disclaimer
Copyright: All rights reserved. No part of this publication may be translated into other languages, reproduced or utilized in any form or by any means, electronic or mechanical, including photocopying, recording, microcopying, or by any information storage and retrieval system, without permission in writing from the publisher.
Drug Dosage: The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any changes in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug.
Disclaimer: The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publishers and the editor(s). The appearance of advertisements or/and product references in the publication is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements.