Pharmacokinetics of the Cardioprotective Drug Dexrazoxane and Its Active Metabolite ADR-925 with Focus on Cardiomyocytes and the Heart

Dexrazoxane (DEX), the only cardioprotectant approved against anthracycline cardiotoxicity, has been traditionally deemed to be a prodrug of the iron-chelating metabolite ADR-925. However, pharmacokinetic profile of both agents, particularly with respect to the cells and tissues essential for its action (cardiomyocytes/myocardium), remains poorly understood. The aim of this study is to characterize the conversion and disposition of DEX to ADR-925 in vitro (primary cardiomyocytes) and in vivo (rabbits) under conditions where DEX is clearly cardioprotective against anthracycline cardiotoxicity. Our results show that DEX is hydrolyzed to ADR-925 in cell media independently of the presence of cardiomyocytes or their lysate. Furthermore, ADR-925 directly penetrates into the cells with contribution of active transport, and detectable concentrations occur earlier than after DEX incubation. In rabbits, ADR-925 was detected rapidly in plasma after DEX administration to form sustained concentrations thereafter. ADR-925 was not markedly retained in the myocardium, and its relative exposure was 5.7-fold lower than for DEX. Unlike liver tissue, myocardium homogenates did not accelerate the conversion of DEX to ADR-925 in vitro, suggesting that myocardial concentrations in vivo may originate from its distribution from the central compartment. The pharmacokinetic parameters for both DEX and ADR-925 were determined by both noncompartmental analyses and population pharmacokinetics (including joint parent-metabolite model). Importantly, all determined parameters were closer to human than to rodent data. The present results open venues for the direct assessment of the cardioprotective effects of ADR-925 in vitro and in vivo to establish whether DEX is a drug or prodrug.

[1]  S. Lipshultz,et al.  Prevention of cardiotoxicity among survivors of childhood cancer , 2017, British journal of clinical pharmacology.

[2]  C. Henninger,et al.  Statins in anthracycline-induced cardiotoxicity: Rac and Rho, and the heartbreakers , 2017, Cell Death & Disease.

[3]  J. Chládek,et al.  Cardioprotective effects of inorganic nitrate/nitrite in chronic anthracycline cardiotoxicity: Comparison with dexrazoxane. , 2016, Journal of molecular and cellular cardiology.

[4]  D. Richardson,et al.  Synthesis and analysis of novel analogues of dexrazoxane and its open-ring hydrolysis product for protection against anthracycline cardiotoxicity in vitro and in vivo , 2015 .

[5]  P. Vejpongsa,et al.  Prevention of anthracycline-induced cardiotoxicity: challenges and opportunities. , 2014, Journal of the American College of Cardiology.

[6]  P. Vejpongsa,et al.  Topoisomerase 2β: A Promising Molecular Target for Primary Prevention of Anthracycline‐Induced Cardiotoxicity , 2013, Clinical pharmacology and therapeutics.

[7]  M. Štěrba,et al.  Catalytic Inhibitors of Topoisomerase II Differently Modulate the Toxicity of Anthracyclines in Cardiac and Cancer Cells , 2013, PloS one.

[8]  M. Štěrba,et al.  Early and delayed cardioprotective intervention with dexrazoxane each show different potential for prevention of chronic anthracycline cardiotoxicity in rabbits. , 2013, Toxicology.

[9]  J. Klimeš,et al.  Development of LC-MS/MS method for the simultaneous analysis of the cardioprotective drug dexrazoxane and its metabolite ADR-925 in isolated cardiomyocytes and cell culture medium. , 2013, Journal of pharmaceutical and biomedical analysis.

[10]  M. Štěrba,et al.  Oxidative stress, redox signaling, and metal chelation in anthracycline cardiotoxicity and pharmacological cardioprotection. , 2013, Antioxidants and Redox Signaling.

[11]  M. Štěrba,et al.  Chronic Anthracycline Cardiotoxicity: Molecular and Functional Analysis with Focus on Nuclear Factor Erythroid 2-Related Factor 2 and Mitochondrial Biogenesis Pathways , 2012, Journal of Pharmacology and Experimental Therapeutics.

[12]  T. Jaki,et al.  Establishing Bioequivalence in Complete and Incomplete Data Designs Using AUCs , 2010, Journal of biopharmaceutical statistics.

[13]  S. Langer Extravasation of Chemotherapy , 2010, Current oncology reports.

[14]  Curtis D. Klaassen,et al.  Xenobiotic, Bile Acid, and Cholesterol Transporters: Function and Regulation , 2010, Pharmacological Reviews.

[15]  M. Štěrba,et al.  Dexrazoxane-afforded protection against chronic anthracycline cardiotoxicity in vivo: effective rescue of cardiomyocytes from apoptotic cell death , 2009, British Journal of Cancer.

[16]  P. Schroeder,et al.  The Dihydroorotase Inhibitor 5-Aminoorotic Acid Inhibits the Metabolism in the Rat of the Cardioprotective Drug Dexrazoxane and Its One-Ring Open Metabolites , 2008, Drug Metabolism and Disposition.

[17]  P. Ponka,et al.  Anthracycline toxicity to cardiomyocytes or cancer cells is differently affected by iron chelation with salicylaldehyde isonicotinoyl hydrazone , 2008, British journal of pharmacology.

[18]  R. Pazdur,et al.  Dexrazoxane (Totect): FDA review and approval for the treatment of accidental extravasation following intravenous anthracycline chemotherapy. , 2008, The oncologist.

[19]  Chao-Po Lin,et al.  Topoisomerase IIbeta mediated DNA double-strand breaks: implications in doxorubicin cardiotoxicity and prevention by dexrazoxane. , 2007, Cancer research.

[20]  M. Štěrba,et al.  In vitro and in vivo examination of cardiac troponins as biochemical markers of drug-induced cardiotoxicity. , 2007, Toxicology.

[21]  E. Herman,et al.  Dexrazoxane: how it works in cardiac and tumor cells. Is it a prodrug or is it a drug? , 2007, Cardiovascular Toxicology.

[22]  D. Rosskopf,et al.  Uptake of Cardiovascular Drugs Into the Human Heart: Expression, Regulation, and Function of the Carnitine Transporter OCTN2 (SLC22A5) , 2006, Circulation.

[23]  B. Hasinoff Dexrazoxane use in the prevention of anthracycline extravasation injury. , 2006, Future oncology.

[24]  P. Schroeder,et al.  METABOLISM OF THE ONE-RING OPEN METABOLITES OF THE CARDIOPROTECTIVE DRUG DEXRAZOXANE TO ITS ACTIVE METAL-CHELATING FORM IN THE RAT , 2005, Drug Metabolism and Disposition.

[25]  F. Burczynski,et al.  METABOLISM OF THE CARDIOPROTECTIVE DRUG DEXRAZOXANE AND ONE OF ITS METABOLITES BY ISOLATED RAT MYOCYTES, HEPATOCYTES, AND BLOOD , 2005, Drug Metabolism and Disposition.

[26]  M. Štěrba,et al.  Rabbit model for in vivo study of anthracycline‐induced heart failure and for the evaluation of protective agents , 2004, European journal of heart failure.

[27]  P. Schroeder,et al.  The metabolites of the cardioprotective drug dexrazoxane do not protect myocytes from doxorubicin-induced cytotoxicity. , 2003, Molecular pharmacology.

[28]  M. Sehested,et al.  Metabolism of dexrazoxane (ICRF-187) used as a rescue agent in cancer patients treated with high-dose etoposide , 2003, Cancer Chemotherapy and Pharmacology.

[29]  P. Schroeder,et al.  Dihydroorotase catalyzes the ring opening of the hydrolysis intermediates of the cardioprotective drug dexrazoxane (ICRF-187). , 2002, Drug metabolism and disposition: the biological fate of chemicals.

[30]  P. Schroeder,et al.  The doxorubicin-cardioprotective drug dexrazoxane undergoes metabolism in the rat to its metal ion-chelating form ADR-925 , 2002, Cancer Chemotherapy and Pharmacology.

[31]  S. Groshen,et al.  Phase I trial of 96-hour continuous infusion of dexrazoxane in patients with advanced malignancies. , 2001, Clinical cancer research : an official journal of the American Association for Cancer Research.

[32]  B. Hasinoff,et al.  Relative plasma levels of the cardioprotective drug dexrazoxane and its two active ring-opened metabolites in the rat. , 1999, Drug metabolism and disposition: the biological fate of chemicals.

[33]  V. Ferrans,et al.  Preclinical animal models of cardiac protection from anthracycline-induced cardiotoxicity. , 1998, Seminars in oncology.

[34]  V. Ferrans,et al.  Chemical, biological and clinical aspects of dexrazoxane and other bisdioxopiperazines. , 1998, Current medicinal chemistry.

[35]  U. Gatzemeier,et al.  Ifosfamide and gemcitabine: a phase II trial in advanced inoperable non-small cell lung cancer. , 1998, Seminars in oncology.

[36]  B. Hasinoff,et al.  Metal ion-promoted hydrolysis of the antioxidant cardioprotective agent dexrazoxane (ICRF-187) and its one-ring open hydrolysis products to its metal-chelating active form , 1997 .

[37]  B. Hasinoff,et al.  Ferrous ion strongly promotes the ring opening of the hydrolysis intermediates of the antioxidant cardioprotective agent dexrazoxane (ICRF-187). , 1995, Archives of biochemistry and biophysics.

[38]  S. Wadler,et al.  Pharmacokinetics of the cardioprotector ADR-529 (ICRF-187) in escalating doses combined with fixed-dose doxorubicin. , 1992, Journal of the National Cancer Institute.

[39]  R. Ungerleider,et al.  Phase I study of ICRF-187 in pediatric cancer patients and comparison of its pharmacokinetics in children and adults. , 1986, Cancer treatment reports.

[40]  J. Koeller,et al.  Pharmacokinetics of (+)-1,2-di(3,5-dioxopiperazin-1-yl)propane intravenous infusions in adult cancer patients. , 1982, Cancer research.

[41]  I. Tamai Pharmacological and pathophysiological roles of carnitine/organic cation transporters (OCTNs: SLC22A4, SLC22A5 and Slc22a21). , 2013, Biopharmaceutics & drug disposition.

[42]  L. Wiseman,et al.  Dexrazoxane , 2012, Drugs.

[43]  Risto S. Cvetković,et al.  Dexrazoxane , 2012, Drugs.

[44]  H. Rosing,et al.  Comparative open, randomized, cross-over bioequivalence study of two intravenous dexrazoxane formulations (Cardioxane® and ICRF-187) in patients with advanced breast cancer, treated with 5-fluorouracil-doxorubicin-cyclophosphamide (FDC) , 2010, European Journal of Drug Metabolism and Pharmacokinetics.

[45]  H. Dickinson,et al.  Cardioprotective interventions for cancer patients receiving anthracyclines. , 2008, The Cochrane database of systematic reviews.

[46]  B. Hasinoff Dexrazoxane (ICRF-187) protects cardiac myocytes against hypoxia-reoxygenation damage , 2007, Cardiovascular Toxicology.

[47]  E. Huebner,et al.  Dexrazoxane (ICRF-187) protects cardiac myocytes against doxorubicin by preventing damage to mitochondria , 2007, Cardiovascular Toxicology.

[48]  C. Vogel,et al.  Phase I clinical trial and pharmacokinetics of weekly ICRF-187 (NSC 169780) infusion in patients with solid tumors , 2004, Investigational New Drugs.

[49]  P. Vici,et al.  The current and future role of dexrazoxane as a cardioprotectant in anthracycline treatment: expert panel review , 2003, Journal of Cancer Research and Clinical Oncology.

[50]  B. Hasinoff,et al.  Stereoselective metabolism of dexrazoxane (ICRF-187) and levrazoxane (ICRF-186). , 1999, Chirality.

[51]  V. Clark,et al.  The enzymatic hydrolysis-activation of the adriamycin cardioprotective agent (+)-1,2-bis(3,5-dioxopiperazinyl-1-yl)propane. , 1991, Drug metabolism and disposition: the biological fate of chemicals.