Publication Cover
Xenobiotica
the fate of foreign compounds in biological systems
Volume 51, 2021 - Issue 1
338
Views
2
CrossRef citations to date
0
Altmetric
General Xenobiochemistry

Evaluation of the changes in exposure to thiol compounds in chronic kidney disease patients using the PBPK model

, , &
Pages 31-39 | Received 16 Jun 2020, Accepted 02 Aug 2020, Published online: 12 Aug 2020

References

  • Algaier I, Jakubowski JA, Asai F, von Kügelgen I. (2008). Interaction of the active metabolite of prasugrel, R-138727, with cysteine 97 and cysteine 175 of the human P2Y12 receptor. J Thromb Haemost 6:1908–14.
  • Baillie TA. (2016). Targeted covalent inhibitors for drug design. Angew Chem Int Ed Engl 55:13408–21.
  • Bauer RA. (2015). Covalent inhibitors in drug discovery: from accidental discoveries to avoided liabilities and designed therapies. Drug Discov Today 20:1061–73.
  • Campbell CL, Berger PB, Nuttall GA, et al. (2005). Can N-acetylcysteine reverse the antiplatelet effects of clopidogrel? An in vivo and vitro study. Am Heart J 150:796–9.
  • Carlsson SM, Denneberg T, Emanuelsson BM, et al. (1990). Pharmacokinetics of intravenous 2-mercaptopropionylglycine in man. Eur J Clin Pharmacol 38:499–503.
  • Carlsson SM, Denneberg T, Emanuelsson BM, et al. (1994a). Pharmacokinetics of 2-mercaptopropionylglycine (Tiopronin) in patients with impaired renal function. Drug Investig 7:101–12.
  • Carlsson SM, Denneberg T, Emanuelsson BM, et al. (1994b). Steady-state pharmacokinetics of 2-mercaptopropionylglycine (Tiopronin) in healthy volunteers and patients with cystinuria. Drug Investig 7:41–51.
  • De Nicola L, Zoccali C. (2016). Chronic kidney disease prevalence in the general population: heterogeneity and concerns. Nephrol Dial Transplant 31:331–5.
  • Derks M, Kawamura H, Abt M, et al. (2011). Effects of food intake on the pharmacokinetic properties of dalcetrapib: findings from three phase I, single-dose crossover studies in healthy volunteers. Clin Ther 33:754–65.
  • Dreisbach AW. (2009). The influence of chronic renal failure on drug metabolism and transport. Clin Pharmacol Ther 86:553–6.
  • Drummer OH, Miach P, Jarrott B. (1983). S-methylation of captopril. Demonstration of captopril thiol methyltransferase activity in human erythrocytes and enzyme distribution in rat tissues. Biochem Pharmacol 32:1557–62.
  • Duchin KL, Singhvi SM, Willard DA, et al. (1982). Captopril kinetics. Clin Pharmacol Ther 31:452–8.
  • Farid NA, Smith RL, Gillespie TA, et al. (2007). The disposition of prasugrel, a novel thienopyridine, in humans. Drug Metab Dispos 35:1096–104.
  • Goldsborough AS, Handley MD, Dulcey AE, et al. (2011). Collateral sensitivity of multidrug-resistant cells to the orphan drug tiopronin. J Med Chem 54:4987–97.
  • Gross G, Tardio J, Kuhlmann O. (2012). Solubility and stability of dalcetrapib in vehicles and biological media. Int J Pharm 437:103–9.
  • Hagihara K, Kazui M, Ikenaga H, et al. (2011). The intestine as an important contributor to prasugrel active metabolite formation in vivo. Drug Metab Dispos 39:565–70.
  • Hill NR, Fatoba ST, Oke JL, et al. (2016). Global prevalence of chronic kidney disease – a systematic review and meta-analysis. PLoS One 11:e0158765.
  • Hsueh CH, Hsu V, Zhao P, et al. (2018). PBPK modeling of the effect of reduced kidney function on the pharmacokinetics of drugs excreted renally by organic anion transporters. Clin Pharmacol Ther 103:485–92.
  • Husser C, Pähler A, Seymour M, et al. (2018). Profiling of dalcetrapib metabolites in human plasma by accelerator mass spectrometry and investigation of the free phenothiol by derivatisation with methylacrylate. J Pharm Biomed Anal 152:143–54.
  • Kazui M, Hagihara K, Izumi T, et al. (2014). Hepatic microsomal thiol methyltransferase is involved in stereoselective methylation of pharmacologically active metabolite of prasugrel. Drug Metab Dispos 42:1138–45.
  • Leblond FA, Petrucci M, Dubé P, et al. (2002). Downregulation of intestinal cytochrome P450 in chronic renal failure. J Am Soc Nephrol 13:1579–85.
  • Lonsdale R, Ward RA. (2018). Structure-based design of targeted covalent inhibitors. Chem Soc Rev 47:3816–30.
  • Naud J, Michaud J, Boisvert C, et al. (2007). Down-regulation of intestinal drug transporters in chronic renal failure in rats. J Pharmacol Exp Ther 320:978–85.
  • Nolin TD, Naud J, Leblond FA, Pichette V. (2008). Emerging evidence of the impact of kidney disease on drug metabolism and transport. Clin Pharmacol Ther 83:898–903.
  • Okamoto H, Yonemori F, Wakitani K, et al. (2000). A cholesteryl ester transfer protein inhibitor attenuate atherosclerosis in rabbits. Nature 406:203–7.
  • Onoyama K, Hirakata H, Iseki K, et al. (1981). Blood concentration and urinary excretion of captopril (SQ 14,225) in patients with chronic renal failure. Hypertension 3:456–9.
  • Phelan M, Anzures-Cabrera J, Carlile DJ, et al. (2013). Effect of hepatic and renal impairment on the pharmacokinetics of dalcetrapib: altered distribution of the active thiol? Clin Pharmacokinet 52:255–65.
  • Poulin P, Theil FP. (2002). Prediction of pharmacokinetics prior to in vivo studies. 1. Mechanism-based prediction of volume of distribution. J Pharm Sci 91:129–56.
  • Rehmel JLF, Eckstein JA, Farid NA, et al. (2006). Interactions of two major metabolites of prasugrel, a thienopyridine antiplatelet agent, with the cytochromes P450. Drug Metab Dispos 34:600–7.
  • Rodgers T, Leahy D, Rowland M. (2005). Physiologically based pharmacokinetic modeling 1: predicting the tissue distribution of moderate-to-strong bases. J Pharm Sci 94:1259–76.
  • Rodgers T, Rowland M. (2006). Physiologically based pharmacokinetic modelling 2: predicting the tissue distribution of acids, very weak bases, neutrals and zwitterions. J Pharm Sci 95:1238–57.
  • Sayama H, Komura H, Kogayu M, Iwaki M. (2013). Development of a hybrid physiologically based pharmacokinetic model with drug-specific scaling factors in rat to improve prediction of human pharmacokinetics. J Pharm Sci 102:4193–204.
  • Sayama H, Takubo H, Komura H, et al. (2014). Application of a physiologically based pharmacokinetic model informed by a top-down approach for the prediction of pharmacokinetics in chronic kidney disease patients. AAPS J 16:1018–28.
  • Small DS, Wrishko RE, Ernest CS, et al. (2009). Prasugrel pharmacokinetics and pharmacodynamics in subjects with moderate renal impairment and end-stage renal disease. J Clin Pharm Ther 34:585–94.
  • Takubo H, Ishikawa T, Kuhlmann O, et al. (2014). Pharmacokinetics and disposition of dalcetrapib in rats and monkeys. Xenobiotica 44:1117–26.
  • U.S. Food and Drug Administration (FDA). (2010). Guidance for industry pharmacokinetics in patients with impaired renal function — study design, data analysis, and impact on dosing and labeling. https://www.fda.gov/media/78573/download
  • Williams ET, Jones KO, Ponsler GD, et al. (2008). The biotransformation of prasugrel, a new thienopyridine prodrug, by the human carboxylesterases 1 and 2. Drug Metab Dispos 36:1227–32.
  • Zhao P, Vieira ML, Grillo JA, et al. (2012). Evaluation of exposure change of nonrenally eliminated drugs in patients with chronic kidney disease using physiologically based pharmacokinetic modeling and simulation. J Clin Pharmacol 52:91S–108S.

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.