Elsevier

Toxicology

Volume 235, Issue 3, 25 June 2007, Pages 150-166
Toxicology

Iron chelation-afforded cardioprotection against chronic anthracycline cardiotoxicity: A study of salicylaldehyde isonicotinoyl hydrazone (SIH)

https://doi.org/10.1016/j.tox.2007.03.020Get rights and content

Abstract

Pyridoxal-derived aroylhydrazone iron chelators have been previously shown as effective cardioprotectants against chronic anthracycline cardiotoxicity. In this study we focused on a novel salicylaldehyde analogue (salicylaldehyde isonicotinoyl hydrazone, SIH), which has been recently demonstrated to possess marked and dose-dependent protective effects against oxidative injury of cardiomyocytes. Therefore, in the present study the cardioprotective potential of SIH against daunorubicin (DAU) cardiotoxicity was assessed in vitro (isolated rat ventricular cardiomyocytes; DAU 10 μM, 48 h exposure) as well as in vivo (chronic DAU-induced cardiomyopathy in rabbits; DAU 3 mg/kg, i.v. weekly, 10 weeks). In vitro, SIH (3–100 μM) was able to partially, but significantly decrease the LDH leakage from cardiomyocytes. In vivo, SIH co-administration was capable to reduce (SIH dose of 0.5 mg/kg, i.v.) or even to completely prevent (1.0 mg/kg, i.v.) the DAU-induced mortality. Moreover, the latter dose of the chelator significantly improved the left ventricular function (LV dP/dtmax = 1185 ± 80 kPa/s versus 783 ± 53 kPa/s in the DAU group; P < 0.05) and decreased the severity of the myocardial morphological changes as well as the plasma levels of cardiac troponin T. Unfortunately, further escalation of the SIH dose (to 2.5 mg/kg) resulted in a nearly complete reversal of the protective effects as judged by the overall mortality, functional, morphological as well as biochemical examinations. Hence, this study points out that aroylhydrazone iron chelators can induce a significant cardioprotection against anthracycline cardiotoxicity; however, they share the curious dose–response relationship which is unrelated to the chemical structure or the route of the administration of the chelator.

Introduction

Cardiotoxicity ranks among the most serious and hardly manageable toxic effects of anticancer chemotherapy. It is particularly dangerous due to the significant vulnerability of the cardiomyocytes to the oxidative damage, a very limited regenerative potential of the myocardial tissue, and the cardinal physiological importance of the heart. Besides some novel anticancer drugs, which have been only recently recognized as potentially cardiotoxic (e.g., imatinib, trastuzumab) (Ewer et al., 2005, Kerkela et al., 2006), anthracyclines (e.g., doxorubicin and daunorubicin) have remained for more than 40 years the main threat regarding the cardiac safety of the patients undergoing cancer treatment (Minotti et al., 2004, Jones et al., 2006).

From the clinical point of view, it is the chronic type of anthracycline cardiotoxicity which represents the major problem. Its risk is known to be closely related to the cumulative dose of anthracyclines (Von Hoff et al., 1979). The anthracycline-induced myocardial injury is typically characterized by a progressive myofibrillar loss and degeneration of the left ventricular (LV) cardiomyocytes ultimately resulting in dilated cardiomyopathy and congestive heart failure (Herman et al., 1997). In addition, the cardiac injury may manifest itself even after a considerable delay (4–20 years) and therefore it can compromise the cardiac function of survivors of childhood cancers even during their productive adulthood (Lipshultz et al., 1991).

Despite extensive scientific efforts, the molecular basis of anthracycline cardiotoxicity remains enigmatic, which significantly hampers the rational development of both less cardiotoxic anthracycline derivates as well as effective pharmacological cardioprotectants. Albeit many theories of anthracycline cardiotoxicity has been proposed (Minotti et al., 2004), the most widely accepted is the “ROS and iron hypothesis” (Keizer et al., 1990). The anthracyclines are known to generate ROS by a redox-cycling of the anthracycline aglycon as well as through their complexes with iron (Keizer et al., 1990, Minotti et al., 2004). The quinone part of the anthracycline aglycon can be converted into a semiquinone form by the one electron acquisition. In the further step of cyclic reaction, the superoxide anion radicals are formed by the interaction of semiquinone with molecular oxygen. Superoxide can further dismutate, either spontaneously or enzymatically (by superoxide dismutase) forming the hydrogen peroxide which is in turn converted in to the extremely reactive hydroxyl radicals through iron-catalyzed Haber–Weiss reaction. Furthermore, iron is involved in the anthracycline-induced ROS generation as part of Fe3+-anthracycline complex. In the presence of the reducing systems (e.g., NADH cytochrom P-450 reductase or glutathione), Fe3+-anthracycline complexes can be reduced and the products (Fe2+-anthracycline complexes) are promptly oxidized back via the interaction with molecular oxygen yielding again superoxide radicals. In addition, hydrogen peroxide can also participate in the latter reaction which results into the direct formation of hydroxyl radicals (Keizer et al., 1990). Alternatively, an internal electron shift within the iron–anthracycline complex may give reduction of iron, which subsequently delivers this electron to molecular oxygen, yielding further superoxide anions. In the excess of NO, the superoxide radicals can also react with NO and thus nitrogen reactive species (e.g., peroxynitrite) are formed (Weinstein et al., 2000; Fogli et al., 2004; Pacher et al., 2007).

The only cardioprotective drug with well-evidenced efficacy in both experimental and clinical settings is dexrazoxane – ICRF-187 (Cvetkovic and Scott, 2005). Today it is generally believed that dexrazoxane acts as a pro-drug which is bioactivated to the metal chelating compound ADR-925. The latter agent has been shown to chelate free or loosely bound iron within the cardiac cells and/or displace iron from its complexes with anthracyclines and thus it prevents the formation of reactive oxygen species, particularly the extremely toxic hydroxyl radicals (Hasinoff et al., 1998, Kwok and Richardson, 2000). Unfortunately, the clinical effectiveness of dexrazoxane is partially counterbalanced by its myelotoxicity which may aggravate the myelosuppression induced by anthracyclines and by its higher costs (Cvetkovic and Scott, 2005). Furthermore, there was a suspicion of decreased tumor response in dexrazoxane co-treated patients in one arm of a single clinical study (Swain et al., 1997), although other studies did not confirm this (Swain and Vici, 2004). All the arguments given above may be responsible for the recent estimation that in Europe cardioprotection with dexrazoxane is employed only in 6–7% of anthracycline-treated patients (Swain and Vici, 2004).

Although iron chelation seems to be a plausible mechanism of cardioprotective intervention against anthracycline cardiotoxicity, the potential of this approach was not appropriately appraised yet. So far, only several iron chelating agents have been examined (Link et al., 1996, Saad et al., 2001, Barnabe et al., 2002, Hasinoff et al., 2003, Xu et al., 2006) and, importantly, only very few in vivo studies on the relevant chronic models have been performed (Herman et al., 1994, Herman et al., 1997, Bruynzeel et al., 2006). Interestingly, the well-known iron chelator deferoxamine failed to protect against chronic anthracycline treatment, which was explained by the hydrophilicity of this compound preventing its sufficient distribution into the heart cells (Herman et al., 1994). On the other hand, we have recently shown that the lipophilic aroylhydrazone iron chelator pyridoxal 2-chlorbenzoylhydrazone (o-108) has a significant potential to decrease the toxicity of anthracyclines on the validated model of chronic daunorubicin cardiotoxicity in rabbits (Sterba et al., 2006). However, surprisingly, the cardioprotection disappeared with a higher dose of o-108 which raised certain controversy concerning both the latter agent as well as other pyridoxal-derived aroylhydrazones, including the PIH—a “mother compound” of this class of iron chelators (Simunek et al., 2005a).

In the present study, we have focused on a salicylaldehyde-derived chelator, salicylaldehyde isonicotinoyl hydrazone (SIH, Fig. 1), which has been recently shown to have very strong and concentration-dependent antioxidant properties on the in vitro model of hydrogen peroxide-induced oxidative stress in cardiac cells (Simunek et al., 2005b). The main aim of the present study is therefore to establish whether SIH is able to elicit significant cardioprotection against anthracycline cardiotoxicity both in vitro, using isolated rat ventricular cardiomyocytes, and in vivo, employing the rabbit model of chronic daunorubicin-induced cardiomyopathy (Gersl and Hrdina, 1994, Simunek et al., 2004).

Section snippets

Animals

Adult Chinchilla male rabbits (n = 64) of an average initial weight of 3.52 ± 0.05 kg were housed under a 12 h light cycle, constant temperature and humidity. The animals had free access to water and a standard laboratory pellet diet. Before experimental procedures the animals were fasting overnight. All experiments were performed under ketamine anesthesia (50 mg/kg, i.m.). Final invasive hemodynamic measurements were carried out under pentobarbital anesthesia (30 mg/kg, i.v.). All experiments were

Cardioprotection studies in vitro

Daunorubicin-treated cultured cardiomyocytes initially increased beating rate, which was followed by irregular activity, disruption of the cellular monolayer and eventually contractile arrest. NVCM generally enlarged cellular volume and the nuclear structure became distinctive. Cytoplasmic vacuolization and granulation were observed. Eventually, formation of cell debris was conspicuous (Fig. 2). Epifluorescence microscopy revealed marked accumulation of daunorubicin into the cardiomyocytes:

Discussion

Despite the decades of intensive research and development, dexrazoxane, a prodrug of the iron-chelating compound ADR-925, has remained the only drug with clearly proven ability to significantly decrease the risk and severity of chronic anthracycline cardiotoxicity. Interestingly, although numerous antioxidants were reported as encouraging in the models of acute toxicity (Fujita et al., 1982, Yoda et al., 1986, Tesoriere et al., 1994), they were shown to have only weak, partial or no

Acknowledgements

The authors wish to thank Mrs. Ludmila Latýnová for her skilful technical assistance during the whole study and Assoc. Prof. Bohuslav Mánek for his kind review of English. This study was supported by Research Projects of the Czech Ministry of Education Youth and Sports MSM0021620820 and Czech Ministry of Health MZO 00179906 and a grant from the Czech Science Foundation (GACR 305/05/P156).

References (71)

  • G. Link et al.

    Role of iron in the potentiation of anthracycline cardiotoxicity: identification of heart cell mitochondria as a major site of iron–anthracycline interaction

    J. Lab. Clin. Med.

    (1996)
  • P.J. Oliveira et al.

    Depletion of adenine nucleotide translocator protein in heart mitochondria from doxorubicin-treated rats—relevance for mitochondrial dysfunction

    Toxicology

    (2006)
  • D.R. Richardson et al.

    The potential of iron chelators of the pyridoxal isonicotinoyl hydrazone class as effective antiproliferative agents

    Blood

    (1995)
  • S.Y. Saad et al.

    The preventive role of deferoxamine against acute doxorubicin-induced cardiac, renal and hepatic toxicity in rats

    Pharmacol. Res.

    (2001)
  • T. Simunek et al.

    SIH-a novel lipophilic iron chelator-protects H9c2 cardiomyoblasts from oxidative stress-induced mitochondrial injury and cell death

    J. Mol. Cell. Cardiol.

    (2005)
  • T. Simunek et al.

    Study of daunorubicin cardiotoxicity prevention with pyridoxal isonicotinoyl hydrazone in rabbits

    Pharmacol. Res.

    (2005)
  • K. Temma et al.

    Ca2+ overloading causes the negative inotropic effect of doxorubicin in myocytes isolated from guinea-pig hearts

    Eur. J. Pharmacol.

    (1997)
  • J. Yuan et al.

    Novel di-2-pyridyl-derived iron chelators with marked and selective antitumor activity: in vitro and in vivo assessment

    Blood

    (2004)
  • J.M. Berthiaume et al.

    Dietary vitamin E decreases doxorubicin-induced oxidative stress without preventing mitochondrial dysfunction

    Cardiovasc. Toxicol.

    (2005)
  • J.G. Breed et al.

    Failure of the antioxidant vitamin E to protect against adriamycin-induced cardiotoxicity in the rabbit

    Cancer Res.

    (1980)
  • A.M. Bruynzeel et al.

    The influence of the time interval between monoHER and doxorubicin administration on the protection against doxorubicin-induced cardiotoxicity in mice

    Cancer Chemother. Pharmacol.

    (2006)
  • J.L. Buss et al.

    Pyridoxal isonicotinoyl hydrazone and its analogues

    Adv. Exp. Med. Biol.

    (2002)
  • R.S. Cvetkovic et al.

    Dexrazoxane: a review of its use for cardioprotection during anthracycline chemotherapy

    Drugs

    (2005)
  • B.J. Cusack et al.

    Daunorubicin and daunorubicinol pharmacokinetics in plasma and tissues in the rat

    Cancer Chemother. Pharmacol.

    (1995)
  • R. Danesi et al.

    Pharmacokinetic–pharmacodynamic relationships of the anthracycline anticancer drugs

    Clin. Pharmacokinet.

    (2002)
  • A.R. Dresdale et al.

    Prospective randomized study of the role of N-acetyl cysteine in reversing doxorubicin-induced cardiomyopathy

    Am. J. Clin. Oncol.

    (1982)
  • M.S. Ewer et al.

    Reversibility of trastuzumab-related cardiotoxicity: new insights based on clinical course and response to medical treatment

    J. Clin. Oncol.

    (2005)
  • K. Fujita et al.

    Reduction of Adriamycin® toxicity by ascorbate in mice and guinea pigs

    Cancer Res.

    (1982)
  • V. Gersl et al.

    Noninvasive polygraphic cardiac changes in daunorubicin-induced cardiomyopathy in rabbits

    Sb. Ved. Pr. Lek. Fak. Karlovy Univerzity Hradci Kralove

    (1994)
  • B.B. Hasinoff et al.

    Chemical, biological and clinical aspects of dexrazoxane and other bisdioxopiperazines

    Curr. Med. Chem.

    (1998)
  • E.H. Herman et al.

    Timing of treatment with ICRF-187 and its effect on chronic doxorubicin cardiotoxicity

    Cancer Chemother. Pharmacol.

    (1993)
  • E.H. Herman et al.

    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.

    (1985)
  • E.H. Herman et al.

    Comparison of the protective effects of desferrioxamine and ICRF-187 against doxorubicin-induced toxicity in spontaneously hypertensive rats

    Cancer Chemother. Pharmacol.

    (1994)
  • E.H. Herman et al.

    Comparison of the protective effects against chronic doxorubicin cardiotoxicity and the rates of iron(III) displacement reactions of ICRF-187 and other bisdiketopiperazines

    Cancer Chemother. Pharmacol.

    (1997)
  • H.S. Hochster

    Clinical pharmacology of dexrazoxane

    Semin. Oncol.

    (1998)
  • Cited by (34)

    • The hydrolytic susceptibility of prochelator BSIH in aqueous solutions

      2017, Bioorganic and Medicinal Chemistry Letters
    • Molecular Mechanisms of Anthracycline-Induced Cardiotoxicity

      2017, Cardio-Oncology: Principles, Prevention and Management
    • LC-UV/MS methods for the analysis of prochelator-Boronyl salicylaldehyde isonicotinoyl hydrazone (BSIH) and its active chelator salicylaldehyde isonicotinoyl hydrazone (SIH)

      2015, Journal of Pharmaceutical and Biomedical Analysis
      Citation Excerpt :

      SIH has been previously demonstrated to protect isolated cardiomyocytes and H9c2 cells against oxidative injury induced by H2O2 [4–6] and H9c2 cells against damage induced by tert-butyl hydroperoxide [7] or catecholamines and their reactive intermediates [8]. Marked protective potential of SIH has been also shown in anthracycline cardiotoxicity both in vitro using neonatal cardiomyocytes [9] and in vivo in a chronic model of daunorubicin-induced heart failure in rabbits [10]. Importantly, good inherent tolerability and low toxicity profile of SIH have been demonstrated following its 10 weeks repeated administration to rabbits [11].

    • Comprehensive ESI-MS and MS/MS analysis of aromatic hydrazones derived from nicotinic acid hydrazide

      2014, International Journal of Mass Spectrometry
      Citation Excerpt :

      Biological activity of these compounds such as antimicrobial, antituberculosis and anticancer properties have emerged over the past few years [1–3]. Also, hydrazones as chelating agents have been investigated as potential drugs for treatment of iron-overload associated diseases [3–5]. The chelating ability of aroylhydrazones towards metal cations makes them attractive analytical agents for spectrophotometric and spectrofluorimetric analysis [6,7].

    View all citing articles on Scopus
    View full text