Dexrazoxane-afforded protection against chronic anthracycline cardiotoxicity in vivo: effective rescue of cardiomyocytes from apoptotic cell death

O Popelová, M Sterba, P Hasková, T Simůnek, M Hroch, I Guncová, P Nachtigal, M Adamcová, V Gersl, Y Mazurová, O Popelová, M Sterba, P Hasková, T Simůnek, M Hroch, I Guncová, P Nachtigal, M Adamcová, V Gersl, Y Mazurová

Abstract

Background: Dexrazoxane (DEX, ICRF-187) is the only clinically approved cardioprotectant against anthracycline cardiotoxicity. It has been traditionally postulated to undergo hydrolysis to iron-chelating agent ADR-925 and to prevent anthracycline-induced oxidative stress, progressive cardiomyocyte degeneration and subsequent non-programmed cell death. However, the additional capability of DEX to protect cardiomyocytes from apoptosis has remained unsubstantiated under clinically relevant in vivo conditions.

Methods: Chronic anthracycline cardiotoxicity was induced in rabbits by repeated daunorubicin (DAU) administrations (3 mg kg(-1) weekly for 10 weeks). Cardiomyocyte apoptosis was evaluated using TUNEL (terminal deoxynucleotidyl transferase biotin-dUTP nick end labelling) assay and activities of caspases 3/7, 8, 9 and 12. Lipoperoxidation was assayed using HPLC determination of myocardial malondialdehyde and 4-hydroxynonenal immunodetection.

Results: Dexrazoxane (60 mg kg(-1)) co-treatment was capable of overcoming DAU-induced mortality, left ventricular dysfunction, profound structural damage of the myocardium and release of cardiac troponin T and I to circulation. Moreover, for the first time, it has been shown that DEX affords significant and nearly complete cardioprotection against anthracycline-induced apoptosis in vivo and effectively suppresses the complex apoptotic signalling triggered by DAU. In individual animals, the severity of apoptotic parameters significantly correlated with cardiac function. However, this effective cardioprotection occurred without a significant decrease in anthracycline-induced lipoperoxidation.

Conclusion: This study identifies inhibition of apoptosis as an important target for effective cardioprotection against chronic anthracycline cardiotoxicity and suggests that lipoperoxidation-independent mechanisms are involved in the cardioprotective action of DEX.

Figures

Figure 1
Figure 1
(A) Echocardiographically determined left ventricular (LV) fractional shortening (FS) during the time course of experiment. (B) Invasively determined index of LV contractility (index dP/dtmax) at the end of the experiment. Statistical significance in comparison with ‘*’ the initial values within each group (paired t-test, P<0.05), ‘c’ control, ‘d’ daunorubicin and ‘x’ DEX+DAU groups (ANOVA, P<0.05).
Figure 2
Figure 2
(A) Plasma concentrations of cardiac troponin T (cTnT) and (B) troponin I (cTnI) in individual animals at the end of the experiment.
Figure 3
Figure 3
(AC) Histological examination of the left ventricular (LV) myocardium. Masson's blue trichrome; bar=20 μm. In the control group (A), normal structure of the myocardium was observed. In the DAU group (B), foci of cardiomyocytes showing profound degeneration (asterisks) were found. Marked morphological changes comprise cytoplasmic vacuolisation, cell swelling and loss of myofibrils resulting in cell death with subsequent development of interstitial fibrosis (arrow). The LV myocardium of DEX co-treated animals (C) showed largely preserved normal morphology with only subtle changes; signs of cardiomyocyte degeneration were scarce (asterisk).
Figure 4
Figure 4
Terminal deoxynucleotidyl transferase biotin-dUTP nick end labelling (TUNEL) assay. (AC) Representative samples of left ventricular (LV) myocardium labelled with TUNEL in the control (A), DAU (B) and DEX+DAU (C) groups. The arrows indicate examples of TUNEL-positive nuclei; bar=20 μm. (D) Quantitative analysis of TUNEL assay. Statistical significance (ANOVA, P<0.05) in comparison with ‘d’ daunorubicin group. (E) Scatterplot of the LV fractional shortening vs number of TUNEL-positive nuclei per square millimetre and results of the Spearman's correlation analyses.
Figure 5
Figure 5
(AD) Caspase activities in the left ventricular (LV) myocardium: caspase 3/7 (A), caspase 8 (B), caspase 9 (C) and caspase 12 (D). Statistical significances (ANOVA, P<0.05) in comparison with ‘d’ daunorubicin group.
Figure 6
Figure 6
(AD) Scatterplots of the cardiac function (LV fractional shortening (FS)) vs activity of individual caspases as determined by Spearman's correlation analyses: caspases 3/7 (A), caspase 8 (B), caspase 9 (C) and caspase 12 (D). LU, luminescence units; FU, fluorescence units.
Figure 7
Figure 7
(A) Total malondialdehyde (MDA, marker of lipoperoxidation) levels in LV myocardium. Statistical significances (ANOVA, P<0.05) in comparison with ‘c’ control group. (BE) Scatterplots of MDA levels in the left ventricular myocardium vs activity of individual caspases and results of the Spearman's correlation analyses: caspases 3/7 (B), caspase 8 (C), caspase 9 (D) and caspase 12 (E). (F) Scatterplot of myocardial MDA levels vs LV fractional shortening (FS) and results of the Spearman's correlation analyses. LU, luminescence units; FU, fluorescence units.
Figure 8
Figure 8
Representative samples of left ventricular (LV) myocardium showing 4-hydroxynonenal staining in the control (A), DAU (B) and DEX+DAU (C) groups.
Figure 9
Figure 9
(A) Horizontal Hierarchical Tree Plot. The analysis comprised LV functional parameters (FS and dP/dtmax), the plasma levels of cardiac troponins T and I, markers of apoptosis (TUNEL positivity and activities of caspases) and lipoperoxidation (nine variables) in all experimental groups. The analysis resulted in two main clusters. The first cluster covers exclusively the individuals from the control (C) and DEX co-treated group (DD), whereas the second cluster contains only the animals from the daunorubicin group (D), where almost all animals appeared in one cluster. (B) Principal Component Analysis (PCA) scatterplots. This analysis of the same variables also revealed two distinctly separated clusters comprising the DEX+DAU and control groups in the first, with all animals from the DAU group in the second cluster. Treatment groups: control (▵), DAU (▴) and DEX+DAU (□).

References

    1. Arbustini E, Brega A, Narula J (2008) Ultrastructural definition of apoptosis in heart failure. Heart Fail Rev 13: 121–135
    1. Arola OJ, Saraste A, Pulkki K, Kallajoki M, Parvinen M, Voipio-Pulkki LM (2000) Acute doxorubicin cardiotoxicity involves cardiomyocyte apoptosis. Cancer Res 60: 1789–1792
    1. Bertinchant JP, Robert E, Polge A, Marty-Double C, Fabbro-Peray P, Poirey S, Aya G, Juan JM, Ledermann B, de la Coussaye JE, Dauzat M (2000) Comparison of the diagnostic value of cardiac troponin I and T determinations for detecting early myocardial damage and the relationship with histological findings after isoprenaline-induced cardiac injury in rats. Clin Chim Acta 298: 13–28
    1. Billingham ME, Mason JW, Bristow MR, Daniels JR (1978) Anthracycline cardiomyopathy monitored by morphologic changes. Cancer Treat Rep 62: 865–872
    1. Bruynzeel AM, Niessen HW, Bronzwaer JG, van der Hoeven JJ, Berkhof J, Bast A, van der Vijgh WJ, van Groeningen CJ (2007a) The effect of monohydroxyethylrutoside on doxorubicin-induced cardiotoxicity in patients treated for metastatic cancer in a phase II study. Br J Cancer 97: 1084–1089
    1. Bruynzeel AM, Vormer-Bonne S, Bast A, Niessen HW, van der Vijgh WJ (2007b) Long-term effects of 7-monohydroxyethylrutoside (monoHER) on DOX-induced cardiotoxicity in mice. Cancer Chemother Pharmacol 60: 509–514
    1. Childs AC, Phaneuf SL, Dirks AJ, Phillips T, Leeuwenburgh C (2002) 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 62: 4592–4598
    1. Cvetkovic RS, Scott LJ (2005) Dexrazoxane: a review of its use for cardioprotection during anthracycline chemotherapy. Drugs 65: 1005–1024
    1. Doroshow JH, Locker GY, Myers CE (1980) Enzymatic defenses of the mouse heart against reactive oxygen metabolites: alterations produced by doxorubicin. J Clin Invest 65: 128–135
    1. Dresdale AR, Barr LH, Bonow RO, Mathisen DJ, Myers CE, Schwartz DE, d’Angelo T, Rosenberg SA (1982) Prospective randomized study of the role of N-acetyl cysteine in reversing doxorubicin-induced cardiomyopathy. Am J Clin Oncol 5: 657–663
    1. Esterbauer H, Schaur RJ, Zollner H (1991) Chemistry and biochemistry of 4-hydroxynonenal, malonaldehyde and related aldehydes. Free Radic Biol Med 11: 81–128
    1. Ewer MS, Benjamin RS (2006) Doxorubicin cardiotoxicity: clinical apect, recognition, monitoring, treatment, and prevention. In Cancer and the Heart, Ewer MS, Yeh E (eds), pp 9–32. BC Decker: Hamilton
    1. Gersl V, Hrdina R (1994) Noninvasive polygraphic cardiac changes in daunorubicin-induced cardiomyopathy in rabbits. Sb Ved Pr Lek Fak Karlovy Univerzity Hradci Kralove 37: 49–55
    1. Gianni L, Herman EH, Lipshultz SE, Minotti G, Sarvazyan N, Sawyer DB (2008) Anthracycline cardiotoxicity: from bench to bedside. J Clin Oncol 26: 3777–3784
    1. Godfraind T (1984) Drug-induced cardionecrosis. Arch Toxicol Suppl 7: 1–15
    1. Guidance for Industry, Bioanalytical Method Validation, US Department of Health and Human Services, Food and Drug Administration, Center for Drug Evaluation and Research (CDER), Center for Veterinary Medicine (CVM), May 2001, BP.
    1. Hasinoff BB, Herman EH (2007) Dexrazoxane: how it works in cardiac and tumor cells. Is it a prodrug or is it a drug? Cardiovasc Toxicol 7: 140–144
    1. Hasinoff BB, Kuschak TI, Fattman CL, Yalowich JC (1998) The one-ring open hydrolysis intermediates of the cardioprotective agent dexrazoxane (ICRF-187) do not inhibit the growth of Chinese hamster ovary cells or the catalytic activity of DNA topoisomerase II. Anticancer Drugs 9: 465–471
    1. Herman EH, Ferrans VJ, Jordan W, Ardalan B (1981) Reduction of chronic daunorubicin cardiotoxicity by ICRF-187 in rabbits. Res Commun Chem Pathol Pharmacol 31: 85–97
    1. Herman EH, Ferrans VJ, Myers CE, Van Vleet JF (1985) Comparison of the effectiveness of (+/−)-1,2-bis(3,5-dioxopiperazinyl-1-yl)propane (ICRF-187) and N-acetylcysteine in preventing chronic doxorubicin cardiotoxicity in beagles. Cancer Res 45: 276–281
    1. Herman EH, Zhang J, Ferrans VJ (1994) Comparison of the protective effects of desferrioxamine and ICRF-187 against doxorubicin-induced toxicity in spontaneously hypertensive rats. Cancer Chemother Pharmacol 35: 93–100
    1. Horenstein MS, Vander Heide RS, L’Ecuyer TJ (2000) Molecular basis of anthracycline-induced cardiotoxicity and its prevention. Mol Genet Metab 71: 436–444
    1. Institute of Laboratory Animal Research CoLS, National Research Council (1996) Guide for the Care and Use of Laboratory Animals. Washington: The National Academies Press
    1. Janero DR (1990) Malondialdehyde and thiobarbituric acid-reactivity as diagnostic indices of lipid peroxidation and peroxidative tissue injury. Free Radic Biol Med 9: 515–540
    1. Jang YM, Kendaiah S, Drew B, Phillips T, Selman C, Julian D, Leeuwenburgh C (2004) Doxorubicin treatment in vivo activates caspase-12 mediated cardiac apoptosis in both male and female rats. FEBS Lett 577: 483–490
    1. Jones RL, Swanton C, Ewer MS (2006) Anthracycline cardiotoxicity. Expert Opin Drug Saf 5: 791–809
    1. Kaiserova H, den Hartog GJ, Simunek T, Schroterova L, Kvasnickova E, Bast A (2006) Iron is not involved in oxidative stress-mediated cytotoxicity of doxorubicin and bleomycin. Br J Pharmacol 149: 920–930
    1. Keizer HG, Pinedo HM, Schuurhuis GJ, Joenje H (1990) Doxorubicin (adriamycin): a critical review of free radical-dependent mechanisms of cytotoxicity. Pharmacol Ther 47: 219–231
    1. Kim SJ, Park KM, Kim N, Yeom YI (2006) Doxorubicin prevents endoplasmic reticulum stress-induced apoptosis. Biochem Biophys Res Commun 339: 463–468
    1. Konorev EA, Vanamala S, Kalyanaraman B (2008) Differences in doxorubicin-induced apoptotic signaling in adult and immature cardiomyocytes. Free Radic Biol Med 45: 1723–1728
    1. Kotamraju S, Kalivendi SV, Konorev E, Chitambar CR, Joseph J, Kalyanaraman B (2004) Oxidant-induced iron signaling in Doxorubicin-mediated apoptosis. Methods Enzymol 378: 362–382
    1. Kwok JC, Richardson DR (2000) The cardioprotective effect of the iron chelator dexrazoxane (ICRF-187) on anthracycline-mediated cardiotoxicity. Redox Rep 5: 317–324
    1. Legha SS, Wang YM, Mackay B, Ewer M, Hortobagyi GN, Benjamin RS, Ali MK (1982) Clinical and pharmacologic investigation of the effects of alpha-tocopherol on adriamycin cardiotoxicity. Ann NY Acad Sci 393: 411–418
    1. Lewis W, Silver MD (2001) Adverse effects of drugs on the cardiovascular system. In Cardiovascular Pathology, Silver MD, Gotlieb AI, Schoen FJ (eds), pp 546–550. Churchill Livingstone: Philadelphia
    1. Lim CC, Zuppinger C, Guo X, Kuster GM, Helmes M, Eppenberger HM, Suter TM, Liao R, Sawyer DB (2004) Anthracyclines induce calpain-dependent titin proteolysis and necrosis in cardiomyocytes. J Biol Chem 279: 8290–8299
    1. Lyu YL, Kerrigan JE, Lin CP, Azarova AM, Tsai YC, Ban Y, Liu LF (2007) Topoisomerase IIbeta mediated DNA double-strand breaks: implications in doxorubicin cardiotoxicity and prevention by dexrazoxane. Cancer Res 67: 8839–8846
    1. Martin E, Thougaard AV, Grauslund M, Jensen PB, Bjorkling F, Hasinoff BB, Tjornelund J, Sehested M, Jensen LH (2009) Evaluation of the topoisomerase II-inactive bisdioxopiperazine ICRF-161 as a protectant against doxorubicin-induced cardiomyopathy. Toxicology 255: 72–79
    1. Minotti G, Menna P, Salvatorelli E, Cairo G, Gianni L (2004) Anthracyclines: molecular advances and pharmacologic developments in antitumor activity and cardiotoxicity. Pharmacol Rev 56: 185–229
    1. Myers C, Bonow R, Palmeri S, Jenkins J, Corden B, Locker G, Doroshow J, Epstein S (1983) A randomized controlled trial assessing the prevention of doxorubicin cardiomyopathy by N-acetylcysteine. Semin Oncol 10: 53–55
    1. Nakamura T, Ueda Y, Juan Y, Katsuda S, Takahashi H, Koh E (2000) Fas-mediated apoptosis in adriamycin-induced cardiomyopathy in rats: in vivo study. Circulation 102: 572–578
    1. Pilz J, Meineke I, Gleiter CH (2000) Measurement of free and bound malondialdehyde in plasma by high-performance liquid chromatography as the 2,4-dinitrophenylhydrazine derivative. J Chromatogr B Biomed Sci Appl 742: 315–325
    1. Popelova O, Sterba M, Simunek T, Mazurova Y, Guncova I, Hroch M, Adamcova M, Gersl V (2008) Deferiprone does not protect against chronic anthracycline cardiotoxicity in vivo. J Pharmacol Exp Ther 326: 259–269
    1. Roca J, Ishida R, Berger JM, Andoh T, Wang JC (1994) Antitumor bisdioxopiperazines inhibit yeast DNA topoisomerase II by trapping the enzyme in the form of a closed protein clamp. Proc Natl Acad Sci USA 91: 1781–1785
    1. Sarvazyan N (1996) Visualization of doxorubicin-induced oxidative stress in isolated cardiac myocytes. Am J Physiol 271: H2079–H2085
    1. Sawyer DB, Fukazawa R, Arstall MA, Kelly RA (1999) Daunorubicin-induced apoptosis in rat cardiac myocytes is inhibited by dexrazoxane. Circ Res 84: 257–265
    1. Simunek T, Boer C, Bouwman RA, Vlasblom R, Versteilen AM, Sterba M, Gersl V, Hrdina R, Ponka P, de Lange JJ, Paulus WJ, Musters RJ (2005a) SIH – a novel lipophilic iron chelator – protects H9c2 cardiomyoblasts from oxidative stress-induced mitochondrial injury and cell death. J Mol Cell Cardiol 39: 345–354
    1. Simunek T, Kaiserova H, Sterba M, Popelova O, Adamcova M, Ponka P, Gersl V (2007) Study of protection by salicylaldehyde isonicotinoyl hydrazone against hydrogen peroxide- and anthracycline-induced toxicity to cardiac cells. Circ Res 101: E72
    1. Simunek T, Klimtova I, Kaplanova J, Mazurova Y, Adamcova M, Sterba M, Hrdina R, Gersl V (2004) Rabbit model for in vivo study of anthracycline-induced heart failure and for the evaluation of protective agents. Eur J Heart Fail 6: 377–387
    1. Simunek T, Klimtova I, Kaplanova J, Sterba M, Mazurova Y, Adamcova M, Hrdina R, Gersl V, Ponka P (2005b) Study of daunorubicin cardiotoxicity prevention with pyridoxal isonicotinoyl hydrazone in rabbits. Pharmacol Res 51: 223–231
    1. Simunek T, Sterba M, Holeckova M, Kaplanova J, Klimtova I, Adamcova M, Gersl V, Hrdina R (2005c) Myocardial content of selected elements in experimental anthracycline-induced cardiomyopathy in rabbits. Biometals 18: 163–169
    1. Simunek T, Sterba M, Popelova O, Kaiserova H, Adamcova M, Hroch M, Haskova P, Ponka P, Gersl V (2008) Anthracycline toxicity to cardiomyocytes or cancer cells is differently affected by iron chelation with salicylaldehyde isonicotinoyl hydrazone. Br J Pharmacol 155: 138–148
    1. Solem LE, Henry TR, Wallace KB (1994) Disruption of mitochondrial calcium homeostasis following chronic doxorubicin administration. Toxicol Appl Pharmacol 129: 214–222
    1. Sterba M, Popelova O, Simunek T, Mazurova Y, Potacova A, Adamcova M, Guncova I, Kaiserova H, Palicka V, Ponka P, Gersl V (2007) Iron chelation-afforded cardioprotection against chronic anthracycline cardiotoxicity: a study of salicylaldehyde isonicotinoyl hydrazone (SIH). Toxicology 235: 150–166
    1. Sterba M, Popelova O, Simunek T, Mazurova Y, Potacova A, Adamcova M, Kaiserova H, Ponka P, Gersl V (2006) Cardioprotective effects of a novel iron chelator, pyridoxal 2-chlorobenzoyl hydrazone, in the rabbit model of daunorubicin-induced cardiotoxicity. J Pharmacol Exp Ther 319: 1336–1347
    1. Van Vleet JF, Ferrans VJ, Weirich WE (1980) Cardiac disease induced by chronic adriamycin administration in dogs and an evaluation of vitamin E and selenium as cardioprotectants. Am J Pathol 99: 13–42
    1. Von Hoff DD, Layard MW, Basa P, Davis Jr HL, Von Hoff AL, Rozencweig M, Muggia FM (1979) Risk factors for doxorubicin-induced congestive heart failure. Ann Intern Med 91: 710–717
    1. Wallace KB (2003) Doxorubicin-induced cardiac mitochondrionopathy. Pharmacol Toxicol 93: 105–115
    1. Wallace KB (2007) Adriamycin-induced interference with cardiac mitochondrial calcium homeostasis. Cardiovasc Toxicol 7: 101–107
    1. Wallace KB, Hausner E, Herman E, Holt GD, MacGregor JT, Metz AL, Murphy E, Rosenblum IY, Sistare FD, York MJ (2004) Serum troponins as biomarkers of drug-induced cardiac toxicity. Toxicol Pathol 32: 106–121
    1. Wang JC (2002) Cellular roles of DNA topoisomerases: a molecular perspective. Nat Rev Mol Cell Biol 3: 430–440
    1. Wouters KA, Kremer LC, Miller TL, Herman EH, Lipshultz SE (2005) Protecting against anthracycline-induced myocardial damage: a review of the most promising strategies. Br J Haematol 131: 561–578
    1. Yamaoka M, Yamaguchi S, Suzuki T, Okuyama M, Nitobe J, Nakamura N, Mitsui Y, Tomoike H (2000) Apoptosis in rat cardiac myocytes induced by Fas ligand: priming for Fas-mediated apoptosis with doxorubicin. J Mol Cell Cardiol 32: 881–889
    1. Zhang J, Clark Jr JR, Herman EH, Ferrans VJ (1996) Doxorubicin-induced apoptosis in spontaneously hypertensive rats: differential effects in heart, kidney and intestine, and inhibition by ICRF-187. J Mol Cell Cardiol 28: 1931–1943
    1. Zhou S, Palmeira CM, Wallace KB (2001) Doxorubicin-induced persistent oxidative stress to cardiac myocytes. Toxicol Lett 121: 151–157

Source: PubMed

Подписаться