Skip to main content

Advertisement

Log in

The metabolic profile of mitoxantrone and its relation with mitoxantrone-induced cardiotoxicity

  • Organ Toxicity and Mechanisms
  • Published:
Archives of Toxicology Aims and scope Submit manuscript

Abstract

Mitoxantrone (MTX) is an antitumor agent that causes cardiotoxicity in 18 % patients. The metabolic profile of MTX was assessed after incubation of 100 μM MTX with hepatic S9 fraction isolated from rats. The presence of MTX and its metabolites was also assessed in vivo through the analysis of liver and heart extracts of MTX-treated rats. The cytotoxic effects of MTX and MTX metabolites were evaluated in the H9c2 cells after 24-h incubation with MTX alone and MTX + metabolites. The influence of CYP450- and CYP2E1-mediated metabolism for the cytotoxicity of MTX was assessed after 96-h incubation with MTX (100 nM and 1 μM) in the presence/absence of CYP450 or CYP2E1 inhibitors. After 4-h incubation in supplemented S9 fraction, the MTX content was 35 % lower and 5 metabolites were identified: an acetoxy ester derivative (never described before), two glutathione conjugates, a monocarboxylic acid derivative, and the naphtoquinoxaline, the later commonly related to MTX pharmacological effects. The presence of MTX and naphtoquinoxaline metabolite was evidenced in vivo in liver and heart of MTX-treated rats. The cytotoxicity caused by MTX + metabolites was higher than that observed in the H9c2 cells incubated with non-metabolized MTX group. The co-incubation of MTX with CYP450 and CYP2E1 inhibitors partially prevented the cytotoxicity observed in the MTX groups incubated with H9c2 cells, highlighting that the metabolism of MTX is relevant for its undesirable effects. The naphtoquinoxaline metabolite is described in heart and liver in vivo, highlighting that this metabolite accumulates in these tissues. It was demonstrated that MTX P450-mediated metabolism contributed to MTX toxicity.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

Abbreviations

DAD:

Photodiode array

DAS:

Diallyl sulfide

DMEM:

Dulbecco’s modified eagle’s medium

DMSO:

Dimethyl sulfoxide

ESI:

Electrospray ionization interface

GSH:

Reduced glutathione

LC:

Liquid chromatography

MS:

Mass spectrometry

MTT:

3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide

MTP:

Metyrapone

MTX:

Mitoxantrone

β-NADPH:

β-Nicotinamide adenine dinucleotide phosphate

References

  • An G, Morris ME (2010) HPLC analysis of mitoxantrone in mouse plasma and tissues: application in a pharmacokinetic study. J Pharm Biomed Anal 51(3):750–753

    Article  PubMed  CAS  Google Scholar 

  • Avasarala JR et al (2003) Rapid onset mitoxantrone-induced cardiotoxicity in secondary progressive multiple sclerosis. Mult Scler 9:59–62

    Article  PubMed  CAS  Google Scholar 

  • Basra J et al (1985) Evidence for human liver mediated-free radical formation by doxorubicin and mitozantrone. Anticancer Drug Des 1:45–52

    PubMed  CAS  Google Scholar 

  • Batra VK et al (1986) Pharmacokinetics of mitoxantrone in man and laboratory animals. Drug Metab Rev 17:311–329

    Article  PubMed  CAS  Google Scholar 

  • Blanz J et al (1991) Evidence for oxidative activation of mitoxantrone in human, pig, and rat. Drug Metab Dispos 19(5):871–880

    PubMed  CAS  Google Scholar 

  • Brandon EF et al (2003) An update on in vitro test methods in human hepatic drug biotransformation research: pros and cons. Toxicol Appl Pharmacol 189(3):233–246

    Article  PubMed  CAS  Google Scholar 

  • Brück TB, Brück DW (2011) Oxidative metabolism of the anti-cancer agent mitoxantrone by horseradish, lacto-and lignin peroxidase. Biochimie 93(2):217–226

    Article  PubMed  Google Scholar 

  • Butler J, Hoey BM (1987) Are reduced quinones necessarily involved in the antitumour activity of quinone drugs? Br J Cancer 8:53–59

    CAS  Google Scholar 

  • Canal P et al (1993) Plasma and cellular pharmacokinetics of mitoxantrone in high-dose chemotherapeutic regimen for refractory lymphomas. Cancer Res 53(20):4850–4854

    PubMed  CAS  Google Scholar 

  • Duthie SJ, Grant MH (1989) The role of reductive and oxidative metabolism in the toxicity of mitoxantrone, adriamycin and menadione in human liver derived Hep G2 hepatoma cells. Br J Cancer 60(4):566–571

    Article  PubMed  CAS  Google Scholar 

  • Ehninger G et al (1990) Pharmacokinetics and metabolism of mitoxantrone. A review. Clin Pharmacokinet 18(5):365–380

    Article  PubMed  CAS  Google Scholar 

  • Feofanov A et al (1997) Quantitative confocal spectral imaging analysis of mitoxantrone within living K562 cells: intracellular accumulation and distribution of monomers, aggregates, naphtoquinoxaline metabolite, and drug-target complexes. Biophys J 73(6):3328–3336

    Article  PubMed  CAS  Google Scholar 

  • Fisher GR, Patterson LH (1992) Lack of involvement of reactive oxygen in the cytotoxicity of mitoxantrone, CI941 and ametantrone in MCF-7 cells: comparison with doxorubicin. Cancer Chemother Pharmacol 30(6):451–458

    Article  PubMed  CAS  Google Scholar 

  • Fisher G, Patterson LH, Gutierrez PL (1993) A comparison of free radical formation by quinone anti-tumour agents in MCF-7 cells and the role of NAD(P)H (quinone-acceptor) oxidoreductase (DT-diaphorase). Chem Biol Interact 88:137–153

    Article  PubMed  CAS  Google Scholar 

  • Hsin L-W et al (2008) Synthesis, DNA binding, and cytotoxicity of 1,4-bis(2-amino-ethylamino)anthraquinone-amino acid conjugates. Bioorg Med Chem 16(2):1006–1014

    Article  PubMed  CAS  Google Scholar 

  • Jia L, Liu X (2007) The conduct of drug metabolism studies considered good practice (II): in vitro experiments. Curr Drug Metab 8(8):822–829

    Article  PubMed  CAS  Google Scholar 

  • Khan SN et al (2010) Effect of mitoxantrone on proliferation dynamics and cell cycle progression. Biosci Rep 30(6):375–381

    Article  PubMed  CAS  Google Scholar 

  • Kostrzewa-Nowak D et al (2007) Bioreductive activation of mitoxantrone by NADPH cytochrome P-450 reductase. Implications for increasing its ability to inhibit the growth of sensitive and multidrug resistant leukaemia HL60 cells. Cancer Lett 245(1–2):252–262

    Article  PubMed  CAS  Google Scholar 

  • Li SJ, Rodgers EH, Grant MH (1995) The activity of xenobiotic enzymes and the cytotoxicity of mitoxantrone in MCF 7 human breast cancer cells treated with inducing agents. Chem Biol Interact 97(2):101–118

    Article  PubMed  CAS  Google Scholar 

  • Lowry OH, Rosebrough NJ, Farr AL, Randall RJ (1951) Protein measurement with the folin phenol reagent, J Biol Chem (193):265–272

    PubMed  CAS  Google Scholar 

  • Mewes K et al (1993) Cytochrome P-450-induced cytotoxicity of mitoxantrone by formation of electrophilic intermediates. Cancer Res 53(21):5135–5142

    PubMed  CAS  Google Scholar 

  • Neuhaus O, Kieseier BC, Hartung H-P (2006) Therapeutic role of mitoxantrone in multiple sclerosis. Pharmacol Ther 109(1–2):198–209

    Article  PubMed  CAS  Google Scholar 

  • Novak RF, Kharasch ED (1985) Mitoxantrone: propensity for free radical formation and lipid peroxidation-implications for cardiotoxicity. Invest New Drugs 3(2):95–99

    Article  PubMed  CAS  Google Scholar 

  • Panousis C, Kettle AJ, Phillips DR (1997) Neutrophil-mediated activation of mitoxantrone to metabolites which form adducts with DNA. Cancer Lett 113(1–2):173–178

    Article  PubMed  CAS  Google Scholar 

  • Parkinson A, Ogilvie B (2008) Biotransformation of xenobiotics. In: Klaassen C (ed) Casarett & Doull’s toxicology: the basic science of poisons. McGraw-Hill, New York, p 1331

    Google Scholar 

  • Pontes H et al (2010) Metabolic interactions between ethanol and MDMA in primary cultured rat hepatocytes. Toxicology 270(2–3):150–157

    Article  PubMed  CAS  Google Scholar 

  • Richard B et al (1991) Interspecies variability in mitoxantrone metabolism using primary cultures of hepatocytes isolated from rat, rabbit and humans. Biochem Pharmacol 41(2):255–262

    Article  PubMed  CAS  Google Scholar 

  • Rossato LG et al (2011) Structural isomerization of synephrine influences its uptake and ensuing glutathione depletion in rat-isolated cardiomyocytes. Arch Toxicol 85(8):929–939

    Article  PubMed  CAS  Google Scholar 

  • Sardão V et al (2009a) Doxorubicin-induced mitochondrial dysfunction is secondary to nuclear p53 activation in H9c2 cardiomyoblasts. Cancer Chemother Pharmacol 64(4):811–827

    Article  PubMed  Google Scholar 

  • Sardão V et al (2009b) Morphological alterations induced by doxorubicin on H9c2 myoblasts: nuclear, mitochondrial, and cytoskeletal targets. Cell Biol Toxicol 25(3):227–243

    Article  PubMed  Google Scholar 

  • Seiter K (2005) Toxicity of the topoisomerase II inhibitors. Expert Opin Drug Saf 4(2):219–234

    Article  PubMed  CAS  Google Scholar 

  • Shipp NG et al (1993) Characterization of experimental mitoxantrone cardiotoxicity and its partial inhibition by ICRF-187 in cultured neonatal rat heart cells. Cancer Res 53(3):550–556

    PubMed  CAS  Google Scholar 

  • Silva R et al (2011) In vitro study of P-glycoprotein induction as an antidotal pathway to prevent cytotoxicity in Caco-2 cells. Arch Toxicol 85(4):315–326

    Article  PubMed  CAS  Google Scholar 

  • Wolf CR, Macpherson JS, Smyth JF (1986) Evidence for the metabolism of mitozantrone by microsomal glutathione transferases and 3-methylcholanthrene-inducible glucuronosyl transferases. Biochem Pharmacol 35(9):1577–1581

    Article  PubMed  CAS  Google Scholar 

  • Yoshihara S et al (2001) Metabolic activation of bisphenol A by rat liver S9 fraction. Toxicol Sci 62(2):221–227

    Article  PubMed  CAS  Google Scholar 

  • Zordoky BNM, El-Kadi AOS (2007) H9c2 cell line is a valuable in vitro model to study the drug metabolizing enzymes in the heart. J Pharmacol Toxicol Methods 56(3):317–322

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgments

Authors are grateful to Dr. Zelia dos Santos Azevedo, from Faculty of Sciences, University of Porto, for gently lend us the LC/DAD-ESI/MS and for all precious technical assistance. This work was supported by the Fundação para a Ciência e Tecnologia (FCT)—project [EXPL/DTP-FTO/0290/2012]—QREN initiative with EU/FEDER financing through COMPETE—Operational Programme for Competitiveness Factors. LGR and VMC thank FCT for their Ph.D. grant (SFRH/BD/63473/2009) and Post-doc grant (SFRH/BPD/63746/2009), respectively. The authors are also grateful to Fundação para a Ciência e a Tecnologia (FCT) for Grant No. PEst-C/EQB/LA0006/2011

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Luciana Grazziotin Rossato or Fernando Remião.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary material 1 (DOCX 208 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Rossato, L.G., Costa, V.M., de Pinho, P.G. et al. The metabolic profile of mitoxantrone and its relation with mitoxantrone-induced cardiotoxicity. Arch Toxicol 87, 1809–1820 (2013). https://doi.org/10.1007/s00204-013-1040-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00204-013-1040-6

Keywords

Navigation