Publication Cover
Xenobiotica
the fate of foreign compounds in biological systems
Volume 48, 2018 - Issue 10
267
Views
1
CrossRef citations to date
0
Altmetric
Animal Pharmacokinetics and Metabolism

Characterizing the in vitro species differences in N-glucuronidation of a potent pan-PIM inhibitor GNE-924 containing a 3,5-substituted 6-azaindazole

, , , , , , , , & show all
Pages 1021-1027 | Received 18 Jul 2017, Accepted 26 Aug 2017, Published online: 29 Sep 2017

References

  • Babu SR, Lakshmi VM, Hsu FF, et al. (1993). N-acetylbenzidine-N′-glucuronidation by human, dog and rat liver. Carcinogenesis 14:2605–11
  • Cerny MA. (2016). Prevalence of non-cytochrome P450-mediated metabolism in food and drug administration-approved oral and intravenous drugs: 2006–2015. Drug Metab Dispos 44:1246–52
  • Chang JH, Yoo P, Lee T, et al. (2009). The role of pH in the glucuronidation of raloxifene, mycophenolic acid and ezetimibe. Mol Pharm 6:1216–27
  • Chen LS, Redkar S, Taverna P, et al. (2011). Mechanisms of cytotoxicity to Pim kinase inhibitor, SGI-1776, in acute myeloid leukemia. Blood 118:693–702
  • Cheng Z, Radominska-Pandya A, Tephly TR. (1998). Cloning and expression of human UDP-glucuronosyltransferase (UGT) 1A8. Arch Biochem Biophys 356:301–5
  • Cheng Z, Radominska-Pandya A, Tephly TR. (1999). Studies on the substrate specificity of human intestinal UDP- lucuronosyltransferases 1A8 and 1A10. Drug Metab Dispos 27:1165–70
  • Cibull TL, Jones TD, Li L, et al. (2006). Overexpression of Pim-1 during progression of prostatic adenocarcinoma. J Clin Pathol 59:285–8
  • Claudio JO, Masih-Khan E, Tang H, et al. (2002). A molecular compendium of genes expressed in multiple myeloma. Blood 100:2175–86
  • Dellinger RW, Chen G, Blevins-Primeau AS, et al. (2007). Glucuronidation of PhIP and N-OH-PhIP by UDP-glucuronosyltransferase 1A10. Carcinogenesis 28:2412–18
  • Dhanasekaran SM, Barrette TR, Ghosh D, et al. (2001). Delineation of prognostic biomarkers in prostate cancer. Nature 412:822–6
  • Fisher MB, Campanale K, Ackermann BL, et al. (2000). In vitro glucuronidation using human liver microsomes and the pore-forming peptide alamethicin. Drug Metab Dispos 28:560–6
  • Fisher MB, Paine MF, Strelevitz TJ, Wrighton SA. (2001). The role of hepatic and extrahepatic UDP-glucuronosyltransferases in human drug metabolism. Drug Metab Rev 33:273–97
  • Garcia PD, Langowski JL, Wang Y, et al. (2014). Pan-PIM kinase inhibition provides a novel therapy for treating hematologic cancers. Clin Cancer Res 20:1834–45
  • Ghosheh O, Hawes EM. (2002). Microsomal N-glucuronidation of nicotine and cotinine: human hepatic interindividual, human intertissue, and interspecies hepatic variation. Drug Metab Dispos 30:1478–83
  • Green MD, King CD, Mojarrabi B, et al. (1998). Glucuronidation of amines and other xenobiotics catalyzed by expressed human UDP-glucuronosyltransferase 1A3. Drug Metab Dispos 26:507–12
  • Green MD, Tephly TR. (1996). Glucuronidation of amines and hydroxylated xenobiotics and endobiotics catalyzed by expressed human UGT1.4 protein. Drug Metab Dispos 24:356–63
  • Hiller A, Nguyen N, Strassburg CP, et al. (1999). Retigabine N-glucuronidation and its potential role in enterohepatic circulation. Drug Metab Dispos 27:605–12
  • Hu H, Wang X, Chan GK, et al. (2015). Discovery of 3,5-substituted 6-azaindazoles as potent pan-Pim inhibitors. Bioorg Med Chem Lett 25:5258–64
  • Kaivosaari S, Finel M, Koskinen M. (2011). N-glucuronidation of drugs and other xenobiotics by human and animal UDP-glucuronosyltransferases. Xenobiotica 41:652–69
  • Kaivosaari S, Salonen JS, Taskinen J. (2002). N-Glucuronidation of some 4-arylalkyl-1H-imidazoles by rat, dog, and human liver microsomes. Drug Metab Dispos 30:295–300
  • Kaji H, Kume T. (2005). Characterization of afloqualone N-glucuronidation: species differences and identification of human UDP-glucuronosyltransferase isoform(s). Drug Metab Dispos 33:60–7
  • Kassahun K, Mattiuz E, Franklin R, Gillespie T. (1998). Olanzapine 10-N-glucuronide. A tertiary N-glucuronide unique to humans. Drug Metab Dispos 26:848–55
  • Kato Y, Izukawa T, Oda S, et al. (2013). Human UDP-glucuronosyltransferase (UGT) 2B10 in drug N-glucuronidation: substrate screening and comparison with UGT1A3 and UGT1A4. Drug Metab Dispos 41:1389–97
  • Keeton EK, Mceachern K, Dillman KS, et al. (2014). AZD1208, a potent and selective pan-Pim kinase inhibitor, demonstrates efficacy in preclinical models of acute myeloid leukemia. Blood 123:905–13
  • Keppler D, Leier I, Jedlitschky G, et al. (1996). The function of the multidrug resistance proteins (MRP and cMRP) in drug conjugate transport and hepatobiliary excretion. Adv Enzyme Regul 36:17–29
  • Kerdpin O, Mackenzie PI, Bowalgaha K, et al. (2009). Influence of N-terminal domain histidine and proline residues on the substrate selectivities of human UDP-glucuronosyltransferase 1A1, 1A6, 1A9, 2B7, and 2B10. Drug Metab Dispos 37:1948–55
  • Le Bigot JF, Begue JM, Kiechel JR, Guillouzo A. (1987). Species differences in metabolism of ketotifen in rat, rabbit and man: demonstration of similar pathways in vivo and in cultured hepatocytes. Life Sci 40:883–90
  • Mackenzie PI, Bock KW, Burchell B, et al. (2005). Nomenclature update for the mammalian UDP glycosyltransferase (UGT) gene superfamily. Pharmacogenet Genomics 15:677–85
  • Mackenzie PI, Owens IS, Burchell B, et al. (1997). The UDP glycosyltransferase gene superfamily: recommended nomenclature update based on evolutionary divergence. Pharmacogenetics 7:255–69
  • Magdalou J, Herber R, Bidault R, Siest G. (1992). In vitro N-glucuronidation of a novel antiepileptic drug, lamotrigine, by human liver microsomes. J Pharmacol Exp Ther 260:1166–73
  • Malfatti MA, Felton JS. (2001). N-glucuronidation of 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) and N_hydroxy-PhIP by specific human UDP-glucuronosyltransferases. Carcinogenesis 22:1087–93
  • Meech R, Mackenzie PI. (2010). UGT3A: novel UDP-glycosyltransferases of the UGT superfamily. Drug Metab Rev 42:43–52
  • Mikkers H, Nawijn M, Allen J, et al. (2004). Mice deficient for all PIM kinases display reduced body size and impaired responses to hematopoietic growth factors. Mol Cell Biol 24:6104–15
  • Miners JO, Knights KM, Houston JB, Mackenzie PI. (2006). In vitro-in vivo correlation for drugs and other compounds eliminated by glucuronidation in humans: pitfalls and promises. Biochem Pharmacol 71:1531–9
  • Radominska-Pandya A, Czernik PJ, Little JM, et al. (1999). Structural and functional studies of UDP-glucuronosyltransferases. Drug Metab Rev 31:817–99
  • Ramirez J, Mirkov S, House LK, Ratain MJ. (2015). Glucuronidation of OTS167 in Humans Is Catalyzed by UDP-Glucuronosyltransferases UGT1A1, UGT1A3, UGT1A8, and UGT1A10. Drug Metab Dispos 43:928–35
  • Rowland A, Gaganis P, Elliot DJ, et al. (2007). Binding of inhibitory fatty acids is responsible for the enhancement of UDP-glucuronosyltransferase 2B7 activity by albumin: implications for in vitro-in vivo extrapolation. J Pharmacol Exp Ther 321:137–47
  • Rowland A, Knights KM, Mackenzie PI, Miners JO. (2008). The “albumin effect” and drug glucuronidation: bovine serum albumin and fatty acid-free human serum albumin enhance the glucuronidation of UDP-glucuronosyltransferase (UGT) 1A9 substrates but not UGT1A1 and UGT1A6 activities. Drug Metab Dispos 36:1056–62
  • Shiratani H, Katoh M, Nakajima M, Yokoi T. (2008). Species differences in UDP-glucuronosyltransferase activities in mice and rats. Drug Metab Dispos 36:1745–52
  • Strassburg CP, Oldhafer K, Manns MP, Tukey RH. (1997). Differential expression of the UGT1A locus in human liver, biliary, and gastric tissue: identification of UGT1A7 and UGT1A10 transcripts in extrahepatic tissue. Mol Pharmacol 52:212–20
  • Tamburini J, Green AS, Bardet V, et al. (2009). Protein synthesis is resistant to rapamycin and constitutes a promising therapeutic target in acute myeloid leukemia. Blood 114:1618–27
  • Tukey RH, Strassburg CP. (2000). Human UDP-glucuronosyltransferases: metabolism, expression, and disease. Annu Rev Pharmacol Toxicol 40:581–616
  • Vashishtha SC, Hawes EM, Mccann DJ, et al. (2002). Quaternary ammonium-linked glucuronidation of 1-substituted imidazoles by liver microsomes: interspecies differences and structure-metabolism relationships. Drug Metab Dispos 30:1070–6
  • Wang X, Kolesnikov A, Tay S, et al. (2017). Discovery of 5-Azaindazole (GNE-955) as a Potent Pan-Pim Inhibitor with Optimized Bioavailability. JMed Chem 60:4458--73
  • Williams JA, Hyland R, Jones BC, et al. (2004). Drug-drug interactions for UDP-glucuronosyltransferase substrates: a pharmacokinetic explanation for typically observed low exposure (AUCi/AUC) ratios. Drug Metab Dispos 32:1201–8

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.