Development of water-soluble prodrugs of the bisdioxopiperazine topoisomerase IIβ inhibitor ICRF-193 as potential cardioprotective agents against anthracycline cardiotoxicity

. 2021 Feb 24 ; 11 (1) : 4456. [epub] 20210224

Jazyk angličtina Země Velká Británie, Anglie Médium electronic

Typ dokumentu časopisecké články, práce podpořená grantem

Perzistentní odkaz   https://www.medvik.cz/link/pmid33627707
Odkazy

PubMed 33627707
PubMed Central PMC7904827
DOI 10.1038/s41598-021-83688-x
PII: 10.1038/s41598-021-83688-x
Knihovny.cz E-zdroje

The bisdioxopiperazine topoisomerase IIβ inhibitor ICRF-193 has been previously identified as a more potent analog of dexrazoxane (ICRF-187), a drug used in clinical practice against anthracycline cardiotoxicity. However, the poor aqueous solubility of ICRF-193 has precluded its further in vivo development as a cardioprotective agent. To overcome this issue, water-soluble prodrugs of ICRF-193 were prepared, their abilities to release ICRF-193 were investigated using a novel UHPLC-MS/MS assay, and their cytoprotective effects against anthracycline cardiotoxicity were tested in vitro in neonatal ventricular cardiomyocytes (NVCMs). Based on the obtained results, the bis(2-aminoacetoxymethyl)-type prodrug GK-667 was selected for advanced investigations due to its straightforward synthesis, sufficient solubility, low cytotoxicity and favorable ICRF-193 release. Upon administration of GK-667 to NVCMs, the released ICRF-193 penetrated well into the cells, reached sufficient intracellular concentrations and provided effective cytoprotection against anthracycline toxicity. The pharmacokinetics of the prodrug, ICRF-193 and its rings-opened metabolite was estimated in vivo after administration of GK-667 to rabbits. The plasma concentrations of ICRF-193 reached were found to be adequate to achieve cardioprotective effects in vivo. Hence, GK-667 was demonstrated to be a pharmaceutically acceptable prodrug of ICRF-193 and a promising drug candidate for further evaluation as a potential cardioprotectant against chronic anthracycline toxicity.

Zobrazit více v PubMed

Nebigil CG, Desaubry L. Updates in anthracycline-mediated cardiotoxicity. Front. Pharmacol. 2018;9:1262. doi: 10.3389/fphar.2018.01262. PubMed DOI PMC

Lenneman CG, Sawyer DB. Cardio-oncology: an update on cardiotoxicity of cancer-related treatment. Circ. Res. 2016;118:1008–1020. doi: 10.1161/CIRCRESAHA.115.303633. PubMed DOI

Cvetkokic RS, Scott LJ. Dexrazoxane—a review of its use for cardioprotection during anthracycline chemotherapy. Drugs. 2005;65:1005–1024. doi: 10.2165/00003495-200565070-00008. PubMed DOI

Sterba M, et al. Oxidative stress, redox signaling, and metal chelation in anthracycline cardiotoxicity and pharmacological cardioprotection. Antioxid. Redox Signal. 2013;18:899–929. doi: 10.1089/ars.2012.4795. PubMed DOI PMC

Jirkovska-Vavrova A, 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. Toxicol. Res. 2015;4:1098–1114. doi: 10.1039/C5TX00048C. DOI

Kollárová-Brázdová P, et al. Investigation of structure-activity relationships of dexrazoxane analogues reveals topoisomerase IIβ interaction as a pre-requisite for effective protection against anthracycline cardiotoxicity. J. Pharmacol. Exp. Ther. 2020;373:402–415. doi: 10.1124/jpet.119.264580. PubMed DOI

Martin E, et al. Evaluation of the topoisomerase II-inactive bisdioxopiperazine ICRF-161 as a protectant against doxorubicin-induced cardiomyopathy. Toxicology. 2009;255:72–79. doi: 10.1016/j.tox.2008.10.011. PubMed DOI

Hasinoff BB, Patel D, Wu X. A QSAR study that compares the ability of bisdioxopiperazine analogs of the doxorubicin cardioprotective agent dexrazoxane (ICRF-187) to protect myocytes with DNA topoisomerase II inhibition. Toxicol. Appl. Pharmacol. 2020;399:115038. doi: 10.1016/j.taap.2020.115038. PubMed DOI

Hasinoff BB, Hellmann K, Herman EH, Ferrans VJ. Chemical, biological and clinical aspects of dexrazoxane and other bisdioxopiperazines. Curr. Med. Chem. 1998;5:1–28. PubMed

Hasinoff BB, Herman EH. Dexrazoxane: how it works in cardiac and tumor cells. Is it a prodrug or is it a drug? Cardiovasc. Toxicol. 2007;7:140–144. doi: 10.1007/s12012-007-0023-3. PubMed DOI

Zhang S, et al. Identification of the molecular basis of doxorubicin-induced cardiotoxicity. Nat. Med. 2012;18:1639–1642. doi: 10.1038/nm.2919. PubMed DOI

Lyu YL, et al. Topoisomerase II beta-Mediated DNA double-strand breaks: Implications in doxorubicin cardiotoxicity and prevention by dexrazoxane. Cancer Res. 2007;67:8839–8846. doi: 10.1158/0008-5472.CAN-07-1649. PubMed DOI

Deng S, et al. Dexrazoxane may prevent doxorubicin-induced DNA damage via depleting both topoisomerase II isoforms. BMC Cancer. 2014;14:842. doi: 10.1186/1471-2407-14-842. PubMed DOI PMC

Lencova-Popelova O, et al. Cardioprotective effects of inorganic nitrate/nitrite in chronic anthracycline cardiotoxicity: comparison with dexrazoxane. J. Mol. Cell. Cardiol. 2016;91:92–103. doi: 10.1016/j.yjmcc.2015.12.021. PubMed DOI

Bures J, et al. Investigation of novel dexrazoxane analogue JR-311 shows significant cardioprotective effects through topoisomerase IIbeta but not its iron chelating metabolite. Toxicology. 2017;392:1–10. doi: 10.1016/j.tox.2017.09.012. PubMed DOI

Swift LP, et al. The cardio-protecting agent and topoisomerase II catalytic inhibitor sobuzoxane enhances doxorubicin-DNA adduct mediated cytotoxicity. Cancer Chemother. Pharmacol. 2008;61:739–749. doi: 10.1007/s00280-007-0528-2. PubMed DOI

Reimerova P, et al. UHPLC-MS/MS method for analysis of sobuzoxane, its active form ICRF-154 and metabolite EDTA-diamide and its application to bioactivation study. Sci. Rep. 2019;9:4524. doi: 10.1038/s41598-019-40928-5. PubMed DOI PMC

Narita T, et al. Antitumor activities and schedule dependence of orally administered MST-16, a novel derivative of bis(2,6-dioxopiperazine) Cancer Chemother. Pharmacol. 1991;28:235–240. PubMed

Takase, M., Narita, T. & Komatsu, T. Water soluble bis-dioxopiperazine derivatives. US Patent 5393889 (1995).

Jirkovsky E, et al. Pharmacokinetics of the cardioprotective drug dexrazoxane and its active metabolite ADR-925 with focus on cardiomyocytes and the heart. J. Pharmacol. Exp. Ther. 2018;364:433–446. doi: 10.1124/jpet.117.244848. PubMed DOI

Buss JL, Hasinoff BB. Ferrous ion strongly promotes the ring opening of the hydrolysis intermediates of the antioxidant cardioprotective agent dexrazoxane (ICRF-187) Arch. Biochem. Biophys. 1995;317:121–127. doi: 10.1006/abbi.1995.1143. PubMed DOI

Buss JL, Hasinoff BB. 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. J. Inorg. Biochem. 1997;68:101–108. doi: 10.1016/S0162-0134(97)00080-9. DOI

Simunek T, et al. Rabbit model for in vivo study of anthracycline-induced heart failure and for the evaluation of protective agents. Eur. J. Heart Fail. 2004;6:377–387. doi: 10.1016/j.ejheart.2003.05.003. PubMed DOI

Jirkovsky E, et al. Early and delayed cardioprotective intervention with dexrazoxane each show different potential for prevention of chronic anthracycline cardiotoxicity in rabbits. Toxicology. 2013;311:191–204. doi: 10.1016/j.tox.2013.06.012. PubMed DOI

25European Medicines Agency. Guideline on bioanalytical method validation. http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2011/08/WC500109686.pdf (2012). PubMed

Simunek T, et al. Anthracycline toxicity to cardiomyocytes or cancer cells is differently affected by iron chelation with salicylaldehyde isonicotinoyl hydrazone. Br. J. Pharmacol. 2008;155:138–148. doi: 10.1038/bjp.2008.236. PubMed DOI PMC

Chan FK, Moriwaki K, De Rosa MJ. Detection of necrosis by release of lactate dehydrogenase activity. Methods. Mol. Biol. 2013;979:65–70. doi: 10.1007/978-1-62703-290-2_7. PubMed DOI PMC

Korzeniewski C, Callewaert DM. An enzyme-release assay for natural cytotoxicity. J. Immunol. Methods. 1983;64:313–320. doi: 10.1016/0022-1759(83)90438-6. PubMed DOI

Legrand C, et al. Lactate-Dehydrogenase (Ldh) Activity of the Number of Dead Cells in the Medium of Cultured Eukaryotic Cells as Marker. J. Biotechnol. 1992;25:231–243. doi: 10.1016/0168-1656(92)90158-6. PubMed DOI

Najít záznam

Citační ukazatele

Nahrávání dat ...

Možnosti archivace

Nahrávání dat ...