Skip to main content
Top
Published in: European Journal of Drug Metabolism and Pharmacokinetics 5/2019

01-10-2019 | Allopurinol | Original Research Article

In Vitro Metabolism by Aldehyde Oxidase Leads to Poor Pharmacokinetic Profile in Rats for c-Met Inhibitor MET401

Authors: Jiang Wei Zhang, Hai Bing Deng, Chun Ye Zhang, Jing Quan Dai, Qian Li, Qian Gang Zheng, Hui Xin Wan, Hong Ping Yu, Feng He, Yao Chang Xu, Sylvia Zhao, Ji Yue Jeff Zhang

Published in: European Journal of Drug Metabolism and Pharmacokinetics | Issue 5/2019

Login to get access

Abstract

Background and Objectives

MET401 is a potent and selective c-Met inhibitor with a novel triazolopyrimidine scaffold. The aim of this study was to determine the pharmacokinetic profile of MET401 in preclinical species, and to identify the metabolic soft spot and enzyme involved, in order to help medicinal chemists to modify the compound to improve the pharmacokinetic profile.

Methods

A metabolite identification study was performed in different liver fractions from various species. Chemical inhibition with selective cytochrome P450 (CYP) and molybdenum hydroxylase inhibitors was carried out to identify the enzyme involved. The deuterium substitution strategy was adopted to reduce metabolism. Pharmacokinetic studies were performed in rats to confirm the effect.

Results

Although M-2 is a minor metabolite in liver microsomal incubations, it became the predominant metabolite in incubations with liver S9, cytosol, hepatocytes and rat pharmacokinetic study. M-2 was synthesized enzymatically and the structure was identified as a mono-oxidation on the triazolopyrimidine moiety. The M-2 formation was ascribed to aldehyde oxidase (AO)-mediated metabolism based on the following evidence—M-2 production was NADPH independent, pan-CYP inhibitor 1-aminobenzotriazole and xanthine oxidase inhibitor allopurinol did not inhibit M-2 formation, and AO inhibitors menadione and raloxifene inhibited M-2 formation. The deuterated analog MET763 demonstrated an improved pharmacokinetic profile with lower clearance, longer terminal half-life and double oral exposure compared with MET401 in rats.

Conclusions

These results indicate that the main metabolic pathway of MET401 is AO-mediated metabolism, which leads to poor in vivo pharmacokinetic profiles in rodents. The deuterium substitution strategy could be used to reduce AO-mediated metabolism liability.
Literature
1.
go back to reference Cañadas I, Rojo F, Arumí-Uría M, Rovira A, Albanell J, Arriola E. C-MET as a new therapeutic target for the development of novel anticancer drugs. Clin Transl Oncol. 2010;12:253–60.CrossRefPubMed Cañadas I, Rojo F, Arumí-Uría M, Rovira A, Albanell J, Arriola E. C-MET as a new therapeutic target for the development of novel anticancer drugs. Clin Transl Oncol. 2010;12:253–60.CrossRefPubMed
2.
go back to reference Liu X, Newton RC, Scherle PA. Developing c-MET pathway inhibitors for cancer therapy: progress and challenges. Trends Mol Med. 2010;16:37–45.CrossRefPubMed Liu X, Newton RC, Scherle PA. Developing c-MET pathway inhibitors for cancer therapy: progress and challenges. Trends Mol Med. 2010;16:37–45.CrossRefPubMed
3.
go back to reference Parikh PK, Ghate MD. Recent advances in the discovery of small molecule c-Met Kinase inhibitors. Eur J Med Chem. 2018;143:1103–38.CrossRefPubMed Parikh PK, Ghate MD. Recent advances in the discovery of small molecule c-Met Kinase inhibitors. Eur J Med Chem. 2018;143:1103–38.CrossRefPubMed
4.
go back to reference Comoglio PM, Trusolino L, Boccaccio C. Known and novel roles of the MET oncogene in cancer: a coherent approach to targeted therapy. Nat Rev Cancer. 2018;18:341–58.CrossRefPubMed Comoglio PM, Trusolino L, Boccaccio C. Known and novel roles of the MET oncogene in cancer: a coherent approach to targeted therapy. Nat Rev Cancer. 2018;18:341–58.CrossRefPubMed
5.
go back to reference Cui JJ. Targeting receptor tyrosine kinase MET in cancer: small molecule inhibitors and clinical progress. J Med Chem. 2014;57:4427–53.CrossRefPubMed Cui JJ. Targeting receptor tyrosine kinase MET in cancer: small molecule inhibitors and clinical progress. J Med Chem. 2014;57:4427–53.CrossRefPubMed
6.
go back to reference Tiedt R, Degenkolbe E, Furet P, Appleton BA, Wagner S, Schoepfer J, et al. A drug resistance screen using a selective MET inhibitor reveals a spectrum of mutations that partially overlap with activating mutations found in cancer patients. Cancer Res. 2011;71:5255–64.CrossRefPubMed Tiedt R, Degenkolbe E, Furet P, Appleton BA, Wagner S, Schoepfer J, et al. A drug resistance screen using a selective MET inhibitor reveals a spectrum of mutations that partially overlap with activating mutations found in cancer patients. Cancer Res. 2011;71:5255–64.CrossRefPubMed
7.
go back to reference Zhan Z, Peng X, Liu Q, Chen F, Ji Y, Yao S, et al. Discovery of 6-(difluoro(6-(4-fluorophenyl)-[1,2,4]triazolo[4,3-b][1,2,4]triazin-3-yl)methyl)quinoline as a highly potent and selective c-Met inhibitor. Eur J Med Chem. 2016;116:239–51.CrossRefPubMed Zhan Z, Peng X, Liu Q, Chen F, Ji Y, Yao S, et al. Discovery of 6-(difluoro(6-(4-fluorophenyl)-[1,2,4]triazolo[4,3-b][1,2,4]triazin-3-yl)methyl)quinoline as a highly potent and selective c-Met inhibitor. Eur J Med Chem. 2016;116:239–51.CrossRefPubMed
8.
go back to reference Zhao F, Zhang J, Zhang L, Hao Y, Shi C, Xia G, et al. Discovery and optimization of a series of imidazo[4,5-b]pyrazine derivatives as highly potent and exquisitely selective inhibitors of the mesenchymal–epithelial transition factor (c-Met) protein kinase. Bioorg Med Chem. 2016;24:4281–90.CrossRefPubMed Zhao F, Zhang J, Zhang L, Hao Y, Shi C, Xia G, et al. Discovery and optimization of a series of imidazo[4,5-b]pyrazine derivatives as highly potent and exquisitely selective inhibitors of the mesenchymal–epithelial transition factor (c-Met) protein kinase. Bioorg Med Chem. 2016;24:4281–90.CrossRefPubMed
9.
go back to reference Ye L, Wu J, Yang J, Chen W, Luo Y, Zhang Y. Design, synthesis and molecular docking analysis of some novel 7-[(quinolin-6-yl)methyl] purines as potential c-Met inhibitors. Med Chem Res. 2015;24:3327–33.CrossRef Ye L, Wu J, Yang J, Chen W, Luo Y, Zhang Y. Design, synthesis and molecular docking analysis of some novel 7-[(quinolin-6-yl)methyl] purines as potential c-Met inhibitors. Med Chem Res. 2015;24:3327–33.CrossRef
10.
go back to reference Ye L, Ou X, Tian Y, Yu B, Luo Y, Feng B, et al. Indazoles as potential c-met inhibitors: design, synthesis and molecular docking studies. Eur J Med Chem. 2013;65:112–8.CrossRefPubMed Ye L, Ou X, Tian Y, Yu B, Luo Y, Feng B, et al. Indazoles as potential c-met inhibitors: design, synthesis and molecular docking studies. Eur J Med Chem. 2013;65:112–8.CrossRefPubMed
11.
go back to reference Jia H, Dai G, Weng J, Zhang Z, Wang Q, Zhou F, et al. Discovery of (S)-1-(1-(Imidazo[1,2-a]pyridin-6-yl)ethyl)-6-(1-methyl-1H-pyrazol-4-yl)-1H-[1,2,3]triazolo[4,5-b]pyrazine (volitinib) as a highly potent and selective mesenchymal-epithelial transition factor (c-Met) inhibitor in clinical development for tre. J Med Chem. 2014;57:7577–89.CrossRefPubMed Jia H, Dai G, Weng J, Zhang Z, Wang Q, Zhou F, et al. Discovery of (S)-1-(1-(Imidazo[1,2-a]pyridin-6-yl)ethyl)-6-(1-methyl-1H-pyrazol-4-yl)-1H-[1,2,3]triazolo[4,5-b]pyrazine (volitinib) as a highly potent and selective mesenchymal-epithelial transition factor (c-Met) inhibitor in clinical development for tre. J Med Chem. 2014;57:7577–89.CrossRefPubMed
12.
go back to reference Cui JJ, Shen H, Tran-Dubé M, Nambu M, McTigue M, Grodsky N, et al. Lessons from (S)-6-(1-(6-(1-methyl-1H-pyrazol-4-yl)-[1,2,4]triazolo[4,3-b]pyridazin-3-yl)ethyl)quinoline (PF-04254644), an inhibitor of receptor tyrosine kinase c-Met with high protein kinase selectivity but broad phosphodiesterase family inhibition leading to myocardial regeneration in rats. J Med Chem. 2013;56:6651–65.CrossRefPubMed Cui JJ, Shen H, Tran-Dubé M, Nambu M, McTigue M, Grodsky N, et al. Lessons from (S)-6-(1-(6-(1-methyl-1H-pyrazol-4-yl)-[1,2,4]triazolo[4,3-b]pyridazin-3-yl)ethyl)quinoline (PF-04254644), an inhibitor of receptor tyrosine kinase c-Met with high protein kinase selectivity but broad phosphodiesterase family inhibition leading to myocardial regeneration in rats. J Med Chem. 2013;56:6651–65.CrossRefPubMed
13.
go back to reference Pirouzpanah S, Saieed P, Rashidi MR, Reza RM, Delazar A, Abbas D, et al. Inhibitory effects of Ruta graveolens L. extract on guinea pig liver aldehyde oxidase. Chem Pharm Bull (Tokyo). 2006;54:9–13.CrossRef Pirouzpanah S, Saieed P, Rashidi MR, Reza RM, Delazar A, Abbas D, et al. Inhibitory effects of Ruta graveolens L. extract on guinea pig liver aldehyde oxidase. Chem Pharm Bull (Tokyo). 2006;54:9–13.CrossRef
14.
go back to reference Kadam RS, Iyer KR. Isolation of liver aldehyde oxidase containing fractions from different animals and determination of kinetic parameters for benzaldehyde. Indian J Pharm Sci. 2008;70:85–8.CrossRefPubMedPubMedCentral Kadam RS, Iyer KR. Isolation of liver aldehyde oxidase containing fractions from different animals and determination of kinetic parameters for benzaldehyde. Indian J Pharm Sci. 2008;70:85–8.CrossRefPubMedPubMedCentral
15.
go back to reference Linder CD, Renaud NA, Hutzler JM. Is 1-aminobenzotriazole an appropriate in vitro tool as a nonspecific cytochrome P450 inactivator? Drug Metab Dispos. 2009;37:10–3.CrossRefPubMed Linder CD, Renaud NA, Hutzler JM. Is 1-aminobenzotriazole an appropriate in vitro tool as a nonspecific cytochrome P450 inactivator? Drug Metab Dispos. 2009;37:10–3.CrossRefPubMed
16.
go back to reference Bjornsson TD, Callaghan JT, Einolf HJ, Fischer V, Gan L, Grimm S, et al. The conduct of in vitro and in vivo drug-drug interaction studies: a PhRMA perspective. J Clin Pharmacol. 2003;43:443–69.CrossRefPubMed Bjornsson TD, Callaghan JT, Einolf HJ, Fischer V, Gan L, Grimm S, et al. The conduct of in vitro and in vivo drug-drug interaction studies: a PhRMA perspective. J Clin Pharmacol. 2003;43:443–69.CrossRefPubMed
17.
18.
go back to reference Massey V, Komai H, Palmer G, Elion GB. On the mechanism of inactivation of xanthine oxidase by allopurinol and other pyrazolo[3,4-d]pyrimidines. J Biol Chem. 1970;245:2837–44.PubMed Massey V, Komai H, Palmer G, Elion GB. On the mechanism of inactivation of xanthine oxidase by allopurinol and other pyrazolo[3,4-d]pyrimidines. J Biol Chem. 1970;245:2837–44.PubMed
19.
go back to reference Sahi J, Khan KK, Black CB. Aldehyde oxidase activity and inhibition in hepatocytes and cytosolic fractions from mouse, rat, monkey and human. Drug Metab Lett. 2008;2:176–83.CrossRefPubMed Sahi J, Khan KK, Black CB. Aldehyde oxidase activity and inhibition in hepatocytes and cytosolic fractions from mouse, rat, monkey and human. Drug Metab Lett. 2008;2:176–83.CrossRefPubMed
20.
go back to reference Zhang JW, Xiao W, Gao ZT, Yu ZT, Zhang J-YJ. Metabolism of c-Met kinase inhibitors containing quinoline by aldehyde oxidase, electron donating, and steric hindrance effect. Drug Metab Dispos. 2018;46:1847–55.CrossRefPubMed Zhang JW, Xiao W, Gao ZT, Yu ZT, Zhang J-YJ. Metabolism of c-Met kinase inhibitors containing quinoline by aldehyde oxidase, electron donating, and steric hindrance effect. Drug Metab Dispos. 2018;46:1847–55.CrossRefPubMed
21.
go back to reference Torres RA, Korzekwa KR, McMasters DR, Fandozzi CM, Jones JP. Use of density functional calculations to predict the regioselectivity of drugs and molecules metabolized by aldehyde oxidase. J Med Chem. 2007;50:4642–7.CrossRefPubMed Torres RA, Korzekwa KR, McMasters DR, Fandozzi CM, Jones JP. Use of density functional calculations to predict the regioselectivity of drugs and molecules metabolized by aldehyde oxidase. J Med Chem. 2007;50:4642–7.CrossRefPubMed
22.
go back to reference Dalvie D, Sun H, Xiang C, Hu Q, Jiang Y, Kang P. Effect of structural variation on aldehyde oxidase-catalyzed oxidation of zoniporide. Drug Metab Dispos. 2012;40:1575–87.CrossRefPubMed Dalvie D, Sun H, Xiang C, Hu Q, Jiang Y, Kang P. Effect of structural variation on aldehyde oxidase-catalyzed oxidation of zoniporide. Drug Metab Dispos. 2012;40:1575–87.CrossRefPubMed
23.
go back to reference Xu Y, Li L, Wang Y, Xing J, Zhou L, Zhong D, et al. Aldehyde oxidase mediated metabolism in drug-like molecules: a combined computational and experimental study. J Med Chem. 2017;60:2973–82.CrossRefPubMed Xu Y, Li L, Wang Y, Xing J, Zhou L, Zhong D, et al. Aldehyde oxidase mediated metabolism in drug-like molecules: a combined computational and experimental study. J Med Chem. 2017;60:2973–82.CrossRefPubMed
24.
go back to reference Testa B, Krämer SD. The biochemistry of drug metabolism–an introduction: part 2. Redox reactions and their enzymes. Chem Biodivers. 2007;4:257–405.CrossRefPubMed Testa B, Krämer SD. The biochemistry of drug metabolism–an introduction: part 2. Redox reactions and their enzymes. Chem Biodivers. 2007;4:257–405.CrossRefPubMed
25.
go back to reference Cleland WW. Isotope effects: determination of enzyme transition state structure. Methods Enzymol. 1995;249:341–73.CrossRefPubMed Cleland WW. Isotope effects: determination of enzyme transition state structure. Methods Enzymol. 1995;249:341–73.CrossRefPubMed
26.
go back to reference Alfaro JF, Joswig-Jones CA, Ouyang W, Nichols J, Crouch GJ, Jones JP. Purification and mechanism of human aldehyde oxidase expressed in Escherichia coli. Drug Metab Dispos. 2009;37:2393–8.CrossRefPubMedPubMedCentral Alfaro JF, Joswig-Jones CA, Ouyang W, Nichols J, Crouch GJ, Jones JP. Purification and mechanism of human aldehyde oxidase expressed in Escherichia coli. Drug Metab Dispos. 2009;37:2393–8.CrossRefPubMedPubMedCentral
27.
go back to reference Shao L, Hewitt MC. The kinetic isotope effect in the search for deuterated drugs. Drug News Perspect. 2010;23:398–404.CrossRefPubMed Shao L, Hewitt MC. The kinetic isotope effect in the search for deuterated drugs. Drug News Perspect. 2010;23:398–404.CrossRefPubMed
28.
go back to reference Koniarczyk JL, Hesk D, Overgard A, Davies IW, McNally A. A general strategy for site-selective incorporation of deuterium and tritium into pyridines, diazines, and pharmaceuticals. J Am Chem Soc. 2018;140:1990–3.CrossRefPubMed Koniarczyk JL, Hesk D, Overgard A, Davies IW, McNally A. A general strategy for site-selective incorporation of deuterium and tritium into pyridines, diazines, and pharmaceuticals. J Am Chem Soc. 2018;140:1990–3.CrossRefPubMed
29.
go back to reference Zhan Z, Peng X, Sun Y, Ai J, Duan W. Evaluation of deuterium-labeled JNJ38877605: pharmacokinetic, metabolic, and in vivo antitumor profiles. Chem Res Toxicol. 2018;31:1213–8.CrossRefPubMed Zhan Z, Peng X, Sun Y, Ai J, Duan W. Evaluation of deuterium-labeled JNJ38877605: pharmacokinetic, metabolic, and in vivo antitumor profiles. Chem Res Toxicol. 2018;31:1213–8.CrossRefPubMed
30.
go back to reference Pryde DC, Dalvie D, Hu Q, Jones P, Obach RS, Tran TD. Aldehyde oxidase: an enzyme of emerging importance in drug discovery. J Med Chem. 2010;53:8441–60.CrossRefPubMed Pryde DC, Dalvie D, Hu Q, Jones P, Obach RS, Tran TD. Aldehyde oxidase: an enzyme of emerging importance in drug discovery. J Med Chem. 2010;53:8441–60.CrossRefPubMed
31.
go back to reference Kitamura S, Sugihara K, Ohta S. Drug-metabolizing ability of molybdenum hydroxylases. Drug Metab Pharmacokinet. 2006;21:83–98.CrossRefPubMed Kitamura S, Sugihara K, Ohta S. Drug-metabolizing ability of molybdenum hydroxylases. Drug Metab Pharmacokinet. 2006;21:83–98.CrossRefPubMed
32.
go back to reference Lepri S, Ceccarelli M, Milani N, Tortorella S, Cucco A, Valeri A, et al. Structure–metabolism relationships in human-AOX: chemical insights from a large database of aza-aromatic and amide compounds. Proc Natl Acad Sci USA. 2017;114:E3178–87.CrossRefPubMed Lepri S, Ceccarelli M, Milani N, Tortorella S, Cucco A, Valeri A, et al. Structure–metabolism relationships in human-AOX: chemical insights from a large database of aza-aromatic and amide compounds. Proc Natl Acad Sci USA. 2017;114:E3178–87.CrossRefPubMed
33.
go back to reference Sodhi JK, Wong S, Kirkpatrick DS, Liu L, Khojasteh SC, Hop CEC, et al. A novel reaction mediated by human aldehyde oxidase: amide hydrolysis of GDC-0834. Drug Metab Dispos. 2015;43:908–15.CrossRefPubMedPubMedCentral Sodhi JK, Wong S, Kirkpatrick DS, Liu L, Khojasteh SC, Hop CEC, et al. A novel reaction mediated by human aldehyde oxidase: amide hydrolysis of GDC-0834. Drug Metab Dispos. 2015;43:908–15.CrossRefPubMedPubMedCentral
34.
go back to reference Cruciani G, Milani N, Benedetti P, Lepri S, Cesarini L, Baroni M, et al. From experiments to a fast easy-to-use computational methodology to predict human aldehyde oxidase selectivity and metabolic reactions. J Med Chem. 2018;61:360–71.CrossRefPubMed Cruciani G, Milani N, Benedetti P, Lepri S, Cesarini L, Baroni M, et al. From experiments to a fast easy-to-use computational methodology to predict human aldehyde oxidase selectivity and metabolic reactions. J Med Chem. 2018;61:360–71.CrossRefPubMed
35.
go back to reference Dittrich C, Greim G, Borner M, Weigang-Köhler K, Huisman H, Amelsberg A, et al. Phase I and pharmacokinetic study of BIBX 1382 BS, an epidermal growth factor receptor (EGFR) inhibitor, given in a continuous daily oral administration. Eur J Cancer. 2002;38:1072–80.CrossRefPubMed Dittrich C, Greim G, Borner M, Weigang-Köhler K, Huisman H, Amelsberg A, et al. Phase I and pharmacokinetic study of BIBX 1382 BS, an epidermal growth factor receptor (EGFR) inhibitor, given in a continuous daily oral administration. Eur J Cancer. 2002;38:1072–80.CrossRefPubMed
36.
go back to reference Zhang X, Liu H, Weller P, Zheng M, Tao W, Wang J, et al. In silico and in vitro pharmacogenetics: aldehyde oxidase rapidly metabolizes a p38 kinase inhibitor. Pharmacogenomics J. 2011;11:15–24.CrossRefPubMed Zhang X, Liu H, Weller P, Zheng M, Tao W, Wang J, et al. In silico and in vitro pharmacogenetics: aldehyde oxidase rapidly metabolizes a p38 kinase inhibitor. Pharmacogenomics J. 2011;11:15–24.CrossRefPubMed
37.
go back to reference Kaye B, Rance DJ, Waring L. Oxidative metabolism of carbazeran in vitro by liver cytosol of baboon and man. Xenobiotica. 1985;15:237–42.CrossRefPubMed Kaye B, Rance DJ, Waring L. Oxidative metabolism of carbazeran in vitro by liver cytosol of baboon and man. Xenobiotica. 1985;15:237–42.CrossRefPubMed
38.
go back to reference Akabane T, Tanaka K, Irie M, Terashita S, Teramura T. Case report of extensive metabolism by aldehyde oxidase in humans: pharmacokinetics and metabolite profile of FK3453 in rats, dogs, and humans. Xenobiotica. 2011;41:372–84.CrossRefPubMed Akabane T, Tanaka K, Irie M, Terashita S, Teramura T. Case report of extensive metabolism by aldehyde oxidase in humans: pharmacokinetics and metabolite profile of FK3453 in rats, dogs, and humans. Xenobiotica. 2011;41:372–84.CrossRefPubMed
39.
go back to reference Calabrese CR, Loadman PM, Lim LS, Bibby MC, Double JA, Brown JE, et al. In vivo metabolism of the antitumor imidazoacridinone C1311 in the mouse and in vitro comparison with humans. Drug Metab Dispos. 1999;27:240–5.PubMed Calabrese CR, Loadman PM, Lim LS, Bibby MC, Double JA, Brown JE, et al. In vivo metabolism of the antitumor imidazoacridinone C1311 in the mouse and in vitro comparison with humans. Drug Metab Dispos. 1999;27:240–5.PubMed
40.
go back to reference Schofield PC, Robertson IG, Paxton JW. Inter-species variation in the metabolism and inhibition of N-[(2′-dimethylamino)ethyl]acridine-4-carboxamide (DACA) by aldehyde oxidase. Biochem Pharmacol. 2000;59:161–5.CrossRefPubMed Schofield PC, Robertson IG, Paxton JW. Inter-species variation in the metabolism and inhibition of N-[(2′-dimethylamino)ethyl]acridine-4-carboxamide (DACA) by aldehyde oxidase. Biochem Pharmacol. 2000;59:161–5.CrossRefPubMed
41.
go back to reference Fukiya K, Itoh K, Yamaguchi S, Kishiba A, Adachi M, Watanabe N, et al. A single amino acid substitution confers high cinchonidine oxidation activity comparable with that of rabbit to monkey aldehyde oxidase 1. Drug Metab Dispos. 2010;38:302–7.CrossRefPubMed Fukiya K, Itoh K, Yamaguchi S, Kishiba A, Adachi M, Watanabe N, et al. A single amino acid substitution confers high cinchonidine oxidation activity comparable with that of rabbit to monkey aldehyde oxidase 1. Drug Metab Dispos. 2010;38:302–7.CrossRefPubMed
42.
go back to reference Dick RA. Refinement of in vitro methods for identification of aldehyde oxidase substrates reveals metabolites of kinase inhibitors. Drug Metab Dispos. 2018;46:846–59.CrossRefPubMed Dick RA. Refinement of in vitro methods for identification of aldehyde oxidase substrates reveals metabolites of kinase inhibitors. Drug Metab Dispos. 2018;46:846–59.CrossRefPubMed
43.
go back to reference Diamond S, Boer J, Maduskuie TP, Falahatpisheh N, Li Y, Yeleswaram S. Species-specific metabolism of SGX523 by aldehyde oxidase and the toxicological implications. Drug Metab Dispos. 2010;38:1277–85.CrossRefPubMed Diamond S, Boer J, Maduskuie TP, Falahatpisheh N, Li Y, Yeleswaram S. Species-specific metabolism of SGX523 by aldehyde oxidase and the toxicological implications. Drug Metab Dispos. 2010;38:1277–85.CrossRefPubMed
44.
go back to reference Lolkema MP, Bohets HH, Arkenau H-T, Lampo A, Barale E, de Jonge MJ, et al. The c-Met tyrosine kinase inhibitor JNJ-38877605 causes renal toxicity through species-specific insoluble metabolite formation. Clin Cancer Res. 2015;21:2297–304.CrossRefPubMedPubMedCentral Lolkema MP, Bohets HH, Arkenau H-T, Lampo A, Barale E, de Jonge MJ, et al. The c-Met tyrosine kinase inhibitor JNJ-38877605 causes renal toxicity through species-specific insoluble metabolite formation. Clin Cancer Res. 2015;21:2297–304.CrossRefPubMedPubMedCentral
45.
go back to reference Zhao F, Zhang LD, Hao Y, Chen N, Bai R, Wang YJ, et al. Identification of 3-substituted-6-(1-(1H-[1,2,3]triazolo[4,5-b]pyrazin-1-yl)ethyl)quinoline derivatives as highly potent and selective mesenchymal-epithelial transition factor (c-Met) inhibitors via metabolite profiling-based structural optimization. Eur J Med Chem. 2017;134:147–58.CrossRefPubMed Zhao F, Zhang LD, Hao Y, Chen N, Bai R, Wang YJ, et al. Identification of 3-substituted-6-(1-(1H-[1,2,3]triazolo[4,5-b]pyrazin-1-yl)ethyl)quinoline derivatives as highly potent and selective mesenchymal-epithelial transition factor (c-Met) inhibitors via metabolite profiling-based structural optimization. Eur J Med Chem. 2017;134:147–58.CrossRefPubMed
Metadata
Title
In Vitro Metabolism by Aldehyde Oxidase Leads to Poor Pharmacokinetic Profile in Rats for c-Met Inhibitor MET401
Authors
Jiang Wei Zhang
Hai Bing Deng
Chun Ye Zhang
Jing Quan Dai
Qian Li
Qian Gang Zheng
Hui Xin Wan
Hong Ping Yu
Feng He
Yao Chang Xu
Sylvia Zhao
Ji Yue Jeff Zhang
Publication date
01-10-2019
Publisher
Springer International Publishing
Published in
European Journal of Drug Metabolism and Pharmacokinetics / Issue 5/2019
Print ISSN: 0378-7966
Electronic ISSN: 2107-0180
DOI
https://doi.org/10.1007/s13318-019-00557-9

Other articles of this Issue 5/2019

European Journal of Drug Metabolism and Pharmacokinetics 5/2019 Go to the issue