Doxorubicin-Induced Cardiomyopathy: From the Cardiotoxic Mechanisms to Management

https://doi.org/10.1016/j.pcad.2006.10.002Get rights and content

First isolated in the early 1960s, doxorubicin (DOX) remains among the most effective anticancer drug ever developed. However, this drug has proven to be a double-edged sword because it also causes a cardiomyopathy that leads to a form of congestive heart failure that is usually refractory to common medications. It is hoped that a better understanding of the mechanisms underlying DOX's cardiotoxicity will enable development of therapies with which to prevent and/or treat the heart failure it causes. Suggested contributors to DOX-induced cardiomyopathy include formation of reactive oxygen species, apoptosis, inhibited expression of cardiomyocyte-specific genes, and altered molecular signaling. And taking these various contributors into consideration, a variety of approaches aimed at preventing or mitigating the cardiotoxicity of DOX have been tried, but so far, the ability of these treatments to protect the heart from damage has been limited. That said, one recent approach that shows promise is adjuvant therapy with a combination of hematopoietic cytokines, including erythropoietin, granulocyte colony-stimulating factor, and thrombopoietin. We suggest this approach to preventing DOX-induced cardiomyopathy is worthy of serious consideration for clinical use.

Section snippets

Morphological Aspects of DOX-Induced Cardiomyopathy

As with other forms of dilated cardiomyopathy, hearts affected by DOX's cardiotoxicity are enlarged with dilation of all chambers, and mural thrombi are seen frequently in both ventricles. At the light microscope level, there are multifocal areas of patchy and interstitial fibrosis and scattered cardiomyocytes with vacuolation (Fig 2). Vacuolated cardiomyocytes are occasionally observed adjacent to areas of fibrosis (Adria cells). Fibrotic foci generally predominate, whereas areas of acute

Pathogenic Mechanisms of DOX-Induced Cardiomyopathy

The mechanism responsible for DOX-induced cardiomyopathy remains unclear, but it seems very different from that underlying DOX's antitumor activity. This has raised hopes that treatment protocols can be designed that protect the heart without diminishing the drug's antitumor activity.

Although numerous mechanisms have been proposed (Table 1), most studies support the view that an increase in oxidative stress, evidenced increases in the levels of ROS,36, 37, 38 and lipid peroxidation,20, 36, 39

Epidemiology and Clinical Features

Doxorubicin exerts both acute and chronic effects on the cardiovascular system. Its acute effects include arrhythmias (eg, supraventricular tachycardia, ventricular premature beats), hypotension, and various electrocardiographic changes (eg, nonspecific ST-T change, left axial deviation, low voltage).2, 121, 122 These effects occur in approximately 11% of patients during or soon after (within a couple of days) DOX administration and are generally reversible and clinically manageable.2 Acute

Monitoring of Patients and Diagnostic Procedures

The monitoring of patients receiving DOX includes (1) assessment of baseline cardiac performance before DOX therapy begins (to exclude contraindicated patients), (2) regular assessments during treatment (to prevent overdosage or to determine when DOX should be terminated), and (3) follow-up after therapy has been completed (to watch for insidious progression of cardiomyopathy). Various diagnostic procedures are listed in Table 6.

Management: Prevention of DOX-induced cardiomyopathy

Limiting the cumulative dose of DOX to less than 450 mg/m2 is the first line of defense against cardiotoxicity.152 To minimize the total amount of DOX used, it is administered concurrently with other antitumor drugs such as vincristine, procarbazine, cisplatin and cyclophosphamide.127, 152 Additional strategies that have been used in an attempt to prevent DOX-induced cardiomyopathy include the use of DOX analogues, alternative drug-delivery methods, antioxidants, and the iron chelator

Management: Treatment of DOX-induced cardiomyopathy

At present, there is no specific treatment for DOX cardiomyopathy. Typically, DOX cardiomyopathy and heart failure are refractory to conventional therapy.179β-Blockers (eg, metoprolol) and angiotensin-converting enzyme inhibitors (eg, enalapril and captopril) may be useful in patients with congestive heart failure.180, 181 Malignant arrhythmias may also require treatment. Cardiac transplantation remains a vital option for patients with end-stage heart failure due to DOX cardiomyopathy.181, 182,

Conclusions

Anthracyclines, including DOX, rank among the most effective anticancer drugs ever developed, despite their dose-dependent cardiotoxic effects.1, 185 Contributors to DOX-induced cardiomyopathy include free radical formation, apoptosis, inhibited expression of cardiomyocyte-specific genes, and altered molecular signaling pathways.186 A variety of approaches to preventing this cardiotoxicity have be tried, but so far, the ability of these treatments to protect the heart from DOX-induced damage

References (186)

  • G. Capranico et al.

    Formation, resealing and persistence of DNA breaks produced by 4-demethoxydaunorubicin in P388 leukemia cells

    Chem. Biol. Interact.

    (1989)
  • A. Skladanowski et al.

    Adriamycin and daunomycin induce programmed cell death (apoptosis) in tumour cells

    Biochem. Pharmacol.

    (1993)
  • S. Anand et al.

    Induction of apoptosis in chronic myelogenous leukemia lymphocytes by hydroxyurea and adriamycin

    Cancer Lett.

    (1995)
  • J. Zhang et al.

    Doxorubicin-induced apoptosis in spontaneously hypertensive rats: Differential effects in heart, kidney and intestine, and inhibition by ICRF-187

    J. Mol. Cell. Cardiol.

    (1996)
  • P.K. Singal et al.

    Subcellular effects of adriamycin in the heart: A concise review

    J. Mol. Cell. Cardiol.

    (1987)
  • B. Kalyanaraman et al.

    Spin-trapping and direct electron spin resonance investigations of the redox metabolism of quinone anticancer drugs

    Biochim. Biophys. Acta.

    (1980)
  • A.L. Odom et al.

    Biochemical determinants of Adriamycin toxicity in mouse liver, heart and intestine

    Biochem. Pharmacol.

    (1992)
  • S. Kotamraju et al.

    Doxorubicin-induced apoptosis in endothelial cells and cardiomyocytes is ameliorated by nitrone spin traps and ebselen. Role of reactive oxygen and nitrogen species

    J. Biol. Chem.

    (2000)
  • A. Elsasser et al.

    Unresolved issues regarding the role of apoptosis in the pathogenesis of ischemic injury and heart failure

    J. Mol. Cell. Cardiol.

    (2000)
  • E.A. Konorev et al.

    Cell-permeable superoxide dismutase and glutathione peroxidase mimetics afford superior protection against doxorubicin-induced cardiotoxicity: The role of reactive oxygen and nitrogen intermediates

    Arch. Biochem. Biophys.

    (1999)
  • K.J. Davies et al.

    Redox cycling of anthracyclines by cardiac mitochondria. I. Anthracycline radical formation by NADH dehydrogenase

    J. Biol. Chem.

    (1986)
  • V. Berlin et al.

    Reduction of adriamycin to a semiquinone-free radical by NADPH cytochrome P-450 reductase produces DNA cleavage in a reaction mediated by molecular oxygen

    J. Biol. Chem.

    (1981)
  • P.J. Thornalley et al.

    Free radical production from normal and adriamycin-treated rat cardiac sarcosomes

    Biochem. Pharmacol.

    (1985)
  • L. Costa et al.

    Direct detection of paramagnetic species in adriamycin perfused rat hearts

    Biochem. Biophys. Res. Commun.

    (1988)
  • S.V. Kalivendi et al.

    Doxorubicin-induced apoptosis is associated with increased transcription of endothelial nitric-oxide synthase. Effect of antiapoptotic antioxidants and calcium

    J. Biol. Chem.

    (2001)
  • C.F. Curran et al.

    Toxicity profile of dexrazoxane (Zinecard, ICRF-187, ADR-529, NSC-169780), a modulator of doxorubicin cardiotoxicity

    Cancer Treat. Rev.

    (1991)
  • R.D. Klausner et al.

    Regulating the fate of mRNA: The control of cellular iron metabolism

    Cell

    (1993)
  • E.A. Martins et al.

    Oxidative stress induces activation of a cytosolic protein responsible for control of iron uptake

    Arch. Biochem. Biophys.

    (1995)
  • K. Pantopoulos et al.

    Differences in the regulation of iron regulatory protein-1 (IRP-1) by extra- and intracellular oxidative stress

    J. Biol. Chem.

    (1997)
  • N.H. Gehring et al.

    Inactivation of both RNA binding and aconitase activities of iron regulatory protein-1 by quinone-induced oxidative stress

    J. Biol. Chem.

    (1999)
  • T.A. Rouault et al.

    Iron-sulfur clusters as biosensors of oxidants and iron

    Trends Biochem. Sci.

    (1996)
  • J.J. Cohen

    Programmed cell death in the immune system

    Adv. Immunol.

    (1991)
  • D.A. Carson et al.

    Apoptosis and disease

    Lancet

    (1993)
  • R.B. Weiss

    The anthracyclines: Will we ever find a better doxorubicin?

    Semin. Oncol.

    (1992)
  • E.A. Lefrak et al.

    A clinicopathologic analysis of adriamycin cardiotoxicity

    Cancer

    (1973)
  • A.C. Gilladoga et al.

    The cardiotoxicity of adriamycin and daunomycin in children

    Cancer

    (1976)
  • D.D. Von Hoff et al.

    Risk factors for doxorubicin-induced congestive heart failure

    Ann. Intern. Med.

    (1979)
  • G. Minotti et al.

    Anthracyclines: Molecular advances and pharmacologic developments in antitumor activity and cardiotoxicity

    Pharmacol. Rev.

    (2004)
  • N.R. Bachur et al.

    Nuclear catalyzed antibiotic free radical formation

    Cancer Res.

    (1982)
  • B.K. Sinha et al.

    Differential formation of hydroxyl radicals by adriamycin in sensitive and resistant MCF-7 human breast tumor cells: Implications for the mechanism of action

    Biochemistry

    (1987)
  • J. Muindi et al.

    Thiol-dependent DNA damage produced by anthracycline-iron complexes. The structure-activity relationships and molecular mechanisms

    Mol. Pharmacol.

    (1985)
  • H. Eliot et al.

    Oxidative destruction of DNA by the adriamycin-iron complex

    Biochemistry

    (1984)
  • C.E. Myers et al.

    Adriamycin: The role of lipid peroxidation in cardiac toxicity and tumor response

    Science

    (1977)
  • C. Myers et al.

    A randomized controlled trial assessing the prevention of doxorubicin cardiomyopathy by N-acetylcysteine

    Semin. Oncol.

    (1983)
  • Y. Yoda et al.

    Prevention of doxorubicin myocardial toxicity in mice by reduced glutathione

    Cancer Res.

    (1986)
  • N. Siveski-Iliskovic et al.

    Probucol protects against adriamycin cardiomyopathy without interfering with its antitumor effect

    Circulation

    (1995)
  • D.E. Graves et al.

    Adriamycin and daunorubicin bind in a cooperative manner to deoxyribonucleic acid

    Biochemistry

    (1983)
  • C. Holm et al.

    DNA topoisomerase II must act at mitosis to prevent nondisjunction and chromosome breakage

    Mol. Cell. Biol.

    (1989)
  • R. Cece et al.

    Apoptosis of hair follicle cells during doxorubicin-induced alopecia in rats

    Lab. Invest.

    (1996)
  • M.E. Billingham et al.

    Anthracycline cardiomyopathy monitored by morphologic changes

    Cancer. Treat. Rep.

    (1978)
  • Cited by (702)

    View all citing articles on Scopus
    View full text