Publication Cover
Xenobiotica
the fate of foreign compounds in biological systems
Volume 50, 2020 - Issue 4
1,617
Views
3
CrossRef citations to date
0
Altmetric
General Xenobiochemistry

A mechanistic modelling approach for the determination of the mechanisms of inhibition by cyclosporine on the uptake and metabolism of atorvastatin in rat hepatocytes using a high throughput uptake method

ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon & ORCID Icon
Pages 415-426 | Received 05 Jun 2019, Accepted 02 Aug 2019, Published online: 19 Aug 2019

References

  • Aho K, Derryberry D, Peterson T. (2014). Model selection for ecologists: the worldviews of AIC and BIC. Ecology 95:631–6.
  • Amundsen R, Asberg A, Ohm IK, Christensen H. (2012). Cyclosporine A- and tacrolimus-mediated inhibition of CYP3A4 and CYP3A5 in vitro. Drug Metab Dispos 40:655–61.
  • Amundsen R, Christensen H, Zabihyan B, Asberg A. (2010). Cyclosporine A, but not tacrolimus, shows relevant inhibition of organic anion-transporting protein 1B1-mediated transport of atorvastatin. Drug Metab Dispos 38:1499–504.
  • Anguelova M, Karlsson J, Jirstrand M. (2012). Minimal output sets for identifiability. Math Biosci 239:139–53.
  • Baker M, Parton T. (2007). Kinetic determinants of hepatic clearance: plasma protein binding and hepatic uptake. Xenobiotica 37:1110–34.
  • Bearup DJ, Evans ND, Chappell MJ. (2013). The input-output relationship approach to structural identifiability analysis. Comput Methods Programs Biomed 109:171–81.
  • Burnham KPA, Anderson DR. 2002. Information and likelihood theory. Model selection and multimodel inference: a practical information-theoretic approach. 2nd ed. New York: Springer, 60–80.
  • Chang W. 2014. Extrafont: Tools for using fonts. R package version 0.17. Available from: https://CRAN.R-project.org/package=extrafont.
  • Copeland RA. 2013. Reversible modes of inhibitor interactions with enzymes. Evaluation of enzyme inhibitors in drug discovery: a guide for medicinal chemists and pharmacologists. 2nd ed. New Jersey: John Wiley and Sons, 57–119.
  • Evans ND, Chapman MJ, Chappell MJ, Godfrey KR. (2002). Identifiability of uncontrolled nonlinear rational systems. Automatica 38:1799–805.
  • Fricker G, Fahr A. (1997). Mechanisms of hepatic transport of cyclosporin A: an explanation for its cholestatic action? Yale J Biol Med 70:379–90.
  • Fuentes AV, Pineda MD, Venkata KCN. (2018). Comprehension of top 200 prescribed drugs in the US as a resource for pharmacy teaching, training and practice. Pharmacy (Basel, Switzerland) 6:43.
  • Grandjean TR, Chappell MJ, Lench AM, et al. (2014a). Experimental and mathematical analysis of in vitro Pitavastatin hepatic uptake across species. Xenobiotica 44:961–74.
  • Grandjean TR, Chappell MJ, Yates JW, Evans ND. (2014b). Structural identifiability analyses of candidate models for in vitro Pitavastatin hepatic uptake. Comput Methods Programs Biomed 114:e60–9.
  • Hagenbuch B, Gui C. (2008). Xenobiotic transporters of the human organic anion transporting polypeptides (OATP) family. Xenobiotica 38:778–801.
  • Harrison J, De Bruyn T, Darwich AS, Houston JB. (2018). Simultaneous assessment in vitro of transporter and metabolic processes in hepatic drug clearance: use of a media loss approach. Drug Metab Dispos 46:405–14.
  • Imamura R, Ichimaru N, Moriyama T, et al. (2005). Long term efficacy of simvastatin in renal transplant recipients treated with cyclosporine or tacrolimus. Clin Transplant 19:616–21.
  • Ishigami M, Tokui T, Komai T, et al. (1995). Evaluation of the uptake of pravastatin by perfused rat liver and primary cultured rat hepatocytes. Pharm Res 12:1741–5.
  • Jones HM, Barton HA, Lai Y, et al. (2012). Mechanistic pharmacokinetic modeling for the prediction of transporter-mediated disposition in humans from sandwich culture human hepatocyte data. Drug Metab Dispos 40:1007–17.
  • Karlsson J, Anguelova M, Jirstrand M. (2012). An efficient method for structural identifiability analysis of large dynamic systems. 16th IFAC Symposium on System Identification. Belgium: Elsevier, 941–6.
  • Kimoto E, Li R, Scialis RJ, et al. (2015). Hepatic disposition of gemfibrozil and its major metabolite gemfibrozil 1-o-beta-glucuronide. Mol Pharm 12:3943–52.
  • Kolhatkar V, Polli JE. (2010). Reliability of inhibition models to correctly identify type of inhibition. Pharm Res 27: 2433–45.
  • Kulkarni P, Korzekwa K, Nagar S. (2016). Intracellular unbound atorvastatin concentrations in the presence of metabolism and transport. J Pharmacol Exp Ther 359:26–36.
  • Lau YY, Okochi H, Huang Y, Benet LZ. (2006). Multiple transporters affect the disposition of atorvastatin and its two active hydroxy metabolites: application of in vitro and ex situ systems. J Pharmacol Exp Ther 316:762–71.
  • Lemahieu WP, Hermann M, Asberg A, et al. (2005). Combined therapy with atorvastatin and calcineurin inhibitors: no interactions with tacrolimus. Am J Transplant 5:2236–43.
  • Li L, Nouraldeen A, Wilson AG. (2013). Evaluation of transporter-mediated hepatic uptake in a non-radioactive high-throughput assay: a study of kinetics, species difference and plasma protein effect. Xenobiotica 43:253–62.
  • Liao M, Zhu Q, Zhu A, et al. (2018). Comparison of uptake transporter functions in hepatocytes in different species to determine the optimal model for evaluating drug transporter activities in humans. Xenobiotica 49:852–62.
  • Lu C, Li P, Gallegos R, et al. (2006). Comparison of intrinsic clearance in liver microsomes and hepatocytes from rats and humans: evaluation of free fraction and uptake in hepatocytes. Drug Metab Dispos 34:1600–5.
  • Menochet K, Kenworthy KE, Houston JB, Galetin A. (2012a). Simultaneous assessment of uptake and metabolism in rat hepatocytes: a comprehensive mechanistic model. J Pharmacol Exp Ther 341:2–15.
  • Menochet K, Kenworthy KE, Houston JB, Galetin A. (2012b). Use of mechanistic modeling to assess interindividual variability and interspecies differences in active uptake in human and rat hepatocytes. Drug Metab Dispos 40:1744–56.
  • Paine SW, Parker AJ, Gardiner P, et al. (2008). Prediction of the pharmacokinetics of atorvastatin, cerivastatin, and indomethacin using kinetic models applied to isolated rat hepatocytes. Drug Metab Dispos 36:1365–74.
  • Patilea-Vrana G, Unadkat JD. (2016). Transport vs. metabolism: what determines the pharmacokinetics and pharmacodynamics of drugs? Insights from the extended clearance model. Clin Pharmacol Ther 100:413–8.
  • Prueksaritanont T, Koike M, Hoener BA, Benet LZ. (1992). Transport and metabolism of cyclosporine in isolated rat hepatocytes. The effects of lipids. Biochem Pharmacol 43:1997–2006.
  • Reinoso RF, Telfer BA, Brennan BS, Rowland M. (2001). Uptake of teicoplanin by isolated rat hepatocytes: comparison with in vivo hepatic distribution. Drug Metab Dispos 29:453–9.
  • Schwarz LR, Burr R, Schwenk M, et al. (1975). Uptake of taurocholic acid into isolated rat-liver cells. Eur J Biochem 55:617–23.
  • Segel IH. 1993a. Kinetics of unireactant enzymes. Enzyme kinetics: behaviour and analysis of rapid equilibrium and steady-state enzyme systems. New Jersey: John Wiley and Sons Inc., 19–42.
  • Segel IH. 1993b. Rapid equilibrium partial and mixed-type inhibition. Enzyme kinetics: behaviour and analysis of rapid equilibrium and steady-state enzyme systems. New Jersey: John Wiley and Sons Inc., 161.
  • Seglen PO. (1976). Preparation of isolated rat liver cells. Methods Cell Biol 13:29–83.
  • Shitara Y, Sugiyama Y. (2017). Preincubation-dependent and long-lasting inhibition of organic anion transporting polypeptide (OATP) and its impact on drug-drug interactions. Pharmacol Ther 177:67–80.
  • Shitara Y. (2011). Clinical importance of OATP1B1 and OATP1B3 in drug-drug interactions. Drug Metab Pharmacokinet 26:220–7.
  • Shitara Y, Horie T, Sugiyama Y. (2006). Transporters as a determinant of drug clearance and tissue distribution. Eur J Pharm Sci 27:425–46.
  • Skalicka B, Kubanek M, Malek I, et al. (2009). Conversion to tacrolimus and atorvastatin in cyclosporine-treated heart transplant recipients with dyslipidemia refractory to fluvastatin. J Heart Lung Transplant 28:598–604.
  • Soars MG, Grime K, Sproston JL, et al. (2007). Use of hepatocytes to assess the contribution of hepatic uptake to clearance in vivo. Drug Metab Dispos 35:859–65.
  • Soetaert K, Petzoldt T, Woodrow-Setzer R. (2010). Solving differential equations in R. R J 2:5–15.
  • Umehara K, Camenisch G. (2012). Novel in vitro-in vivo extrapolation (IVIVE) method to predict hepatic organ clearance in rat. Pharm Res 29:603–17.
  • Watanabe T, Kusuhara H, Maeda K, et al. (2010). Investigation of the rate-determining process in the hepatic elimination of HMG-CoA reductase inhibitors in rats and humans. Drug Metab Dispos 38:215–22.
  • Wilke CO. 2016. Cowplot: Streamlined plot theme and plot annotations for 'ggplot2'. R package version 0.93. Available from: https://CRAN.R-project.org/package=cowplot
  • Yabe Y, Galetin A, Houston JB. (2011). Kinetic characterization of rat hepatic uptake of 16 actively transported drugs. Drug Metab Dispos 39:1808–14.
  • Yamazaki M, Suzuki H, Hanano M, et al. (1993). Na(+)-independent multispecific anion transporter mediates active transport of pravastatin into rat liver. Am J Physiol 264:G36–44.
  • Yoshikado T, Toshimoto K, Nakada T, et al. (2017). Comparison of methods for estimating unbound intracellular-to-medium concentration ratios in rat and human hepatocytes using statins. Drug Metab Dispos 45:779–89.
  • Ziegler K, Frimmer M. (1986). Identification of cyclosporin binding sites in rat liver plasma membranes, isolated hepatocytes, and hepatoma cells by photoaffinity labeling using [3H]cyclosporin-diaziridine. Biochim Biophys Acta 855:147–56.