Skip to main content
Log in

Theoretical Considerations for Direct Translation of Unbound Liver-to-Plasma Partition Coefficient from In Vitro to In Vivo

  • Research Article
  • Published:
The AAPS Journal Aims and scope Submit manuscript

Abstract

There is considerable interest in developing methods to predict the asymmetric distribution of unbound drug into tissues. The liver is of particular interest due to the multitude of expressed transporters with potential implications for pharmacokinetics, pharmacodynamics, and toxicology. Empirical correlations of in vitro unbound hepatocyte-to-media partition coefficient (in vitro Kpuu) and in vivo unbound liver-to-plasma partition coefficient (in vivo Kpuu) have been reported without considering the theoretical aspects which might confound the interpretation of such observations. To understand the theoretical basis for the translation of Kpuu between in vitro and in vivo systems, we simulated in vitro hepatocyte and in vivo liver Kpuu values using mechanistic mathematical models of these systems. Theoretical comparisons of steady-state Kpuu between in vitro and in vivo systems were performed using liver models which assumed a number of segments ranging from one (i.e., a permeability-limited well-stirred model) to infinity (i.e., a permeability-limited parallel tube model). Using a five-segment model, the effect of zonal differences in metabolism was also explored in this context. The results across the range of examined models indicated that theoretical differences between in vitro and in vivo Kpuu estimates exist and are expected to increase with an increasing degree of extraction across the liver. However, differences were relatively small using what is perhaps the most physiologically relevant, permeability-limited parallel tube model, suggesting that direct correlations are reasonably valid and that the permeability-limited parallel tube model is perhaps the most appropriate physiologically based pharmacokinetic (PBPK) construct for supporting studies of this nature.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Fig. 1
Fig. 2
Fig. 3

Similar content being viewed by others

References

  1. Mateus A, Matsson P, Artursson P. Rapid measurement of intracellular unbound drug concentrations. Mol Pharm. 2013;10(6):2467–78.

    Article  CAS  Google Scholar 

  2. Riccardi K, Li Z, Brown Janice A, Gorgoglione Matthew F, Niosi M, Gosset J, et al. Determination of unbound partition coefficient and in vitro-in vivo extrapolation for SLC13A transporter-mediated uptake. Drug Metab Dispos. 2016;44(10):1633–42.

    Article  CAS  Google Scholar 

  3. Riccardi K, Lin J, Li Z, Niosi M, Ryu S, Hua W, et al. Novel method to predict in vivo liver-to-plasma Kpuu for OATP substrates using suspension hepatocytes. Drug Metab Dispos. 2017;45(5):576–80.

    Article  CAS  Google Scholar 

  4. Shitara Y, Maeda K, Ikejiri K, Yoshida K, Horie T, Sugiyama Y. Clinical significance of organic anion transporting polypeptides (OATPs) in drug disposition: their roles in hepatic clearance and intestinal absorption. Biopharm Drug Dispos. 2013;34(1):45–78.

    Article  CAS  Google Scholar 

  5. Yabe Y, Galetin A, Houston JB. Kinetic characterization of rat hepatic uptake of 16 actively transported drugs. Drug Metab Dispos. 2011;39(10):1808–14.

    Article  CAS  Google Scholar 

  6. Zhang D, Luo G, Ding X, Lu C. Preclinical experimental models of drug metabolism and disposition in drug discovery and development. Acta Pharm Sin B. 2012;2(6):549–61.

    Article  Google Scholar 

  7. Li R, Barton HA, Yates PD, Ghosh A, Wolford AC, Riccardi KA, et al. A "middle-out" approach to human pharmacokinetic predictions for OATP substrates using physiologically-based pharmacokinetic modeling. J Pharmacokinet Pharmacodyn. 2014;41(3):197–209.

    Article  CAS  Google Scholar 

  8. Jones HM, Barton HA, Lai Y, Bi Y-A, Kimoto E, Kempshall S, et al. Mechanistic pharmacokinetic modeling for the prediction of transporter-mediated disposition in humans from sandwich culture human hepatocyte data. Drug Metab Dispos. 2012;40(5):1007–17.

    Article  CAS  Google Scholar 

  9. Morse BL, Cai H, MacGuire JG, Fox M, Zhang L, Zhang Y, et al. Rosuvastatin liver partitioning in cynomolgus monkeys: measurement in vivo and prediction using in vitro monkey hepatocyte uptake. Drug Metab Dispos. 2015;43(11):1788–94.

    Article  CAS  Google Scholar 

  10. Kunze A, Huwyler J, Camenisch G, Poller B. Prediction of organic anion-transporting polypeptide 1B1- and 1B3-mediated hepatic uptake of statins based on transporter protein expression and activity data. Drug Metab Dispos. 2014;42(9):1514–21.

    Article  Google Scholar 

  11. Badee J, Achour B, Rostami-Hodjegan A, Galetin A. Meta-analysis of expression of hepatic organic anion-transporting polypeptide (OATP) transporters in cellular systems relative to human liver tissue. Drug Metab Dispos. 2015;43(4):424–32.

    Article  CAS  Google Scholar 

  12. Li M, Yuan H, Li N, Song G, Zheng Y, Baratta M, et al. Identification of interspecies difference in efflux transporters of hepatocytes from dog, rat, monkey and human. Eur J Pharm Sci. 2008;35(1–2):114–26.

    Article  CAS  Google Scholar 

  13. Bow DA, Perry JL, Miller DS, Pritchard JB, Brouwer KL. Localization of P-gp (Abcb1) and Mrp2 (Abcc2) in freshly isolated rat hepatocytes. Drug Metab Dispos. 2008;36(1):198–202.

    Article  CAS  Google Scholar 

  14. Lundquist P, Englund G, Skogastierna C, Lööf J, Johansson J, Hoogstraate J, et al. Functional ATP-binding cassette drug efflux transporters in isolated human and rat hepatocytes significantly affect assessment of drug disposition. Drug Metab Dispos. 2014;42(3):448–58.

    Article  Google Scholar 

  15. Teutsch HF. The modular microarchitecture of human liver. Hepatology. 2005;42(2):317–25.

    Article  Google Scholar 

  16. Kietzmann T. Metabolic zonation of the liver: the oxygen gradient revisited. Redox Biol. 2017;11:622–30.

    Article  CAS  Google Scholar 

  17. Camenisch G, Riede J, Kunze A, Huwyler J, Poller B, Umehara K. The extended clearance model and its use for the interpretation of hepatobiliary elimination data. ADMET and DMPK 2015.

  18. Pang KS, Rowland M. Hepatic clearance of drugs. I. Theoretical considerations of a “well-stirred” model and a “parallel tube” model. Influence of hepatic blood flow, plasma and blood cell binding, and the hepatocellular enzymatic activity on hepatic drug clearance. J Pharmacokinet Biopharm. 1977;5(6):625–53.

    Article  CAS  Google Scholar 

  19. Martin PD, Warwick MJ, Dane AL, Brindley C, Short T. Absolute oral bioavailability of rosuvastatin in healthy white adult male volunteers. Clin Ther. 2003;25(10):2553–63.

    Article  CAS  Google Scholar 

  20. Shitara Y, Horie T, Sugiyama Y. Transporters as a determinant of drug clearance and tissue distribution. Eur J Pharm Sci. 2006;27(5):425–46.

    Article  CAS  Google Scholar 

  21. Yamazaki M, Kobayashi K, Sugiyama Y. Primary active transport of pravastatin across the liver canalicular membrane in normal and mutant Eisai hyperbilirubinemic rats. Biopharm Drug Dispos. 1996;17(7):607–21.

    Article  CAS  Google Scholar 

  22. Lundquist P, Loof J, Fagerholm U, Sjogren I, Johansson J, Briem S, et al. Prediction of in vivo rat biliary drug clearance from an in vitro hepatocyte efflux model. Drug Metab Dispos. 2014;42(3):459–68.

    Article  Google Scholar 

  23. Iusuf D, van Esch A, Hobbs M, Taylor M, Kenworthy KE, van de Steeg E, et al. Murine Oatp1a/1b uptake transporters control rosuvastatin systemic exposure without affecting its apparent liver exposure. Mol Pharmacol. 2013;83(5):919–29.

    Article  CAS  Google Scholar 

  24. Mao J, Doshi U, Wright M, Hop CECA, Li AP, Chen Y. Prediction of the pharmacokinetics of pravastatin as an OATP substrate using plateable human hepatocytes with human plasma data and PBPK modeling. CPT Pharmacometrics Syst Pharmacol. 2018;7(4):251–8.

    Article  CAS  Google Scholar 

  25. Ito K, Houston JB. Comparison of the use of liver models for predicting drug clearance using in vitro kinetic data from hepatic microsomes and isolated hepatocytes. Pharm Res. 2004;21(5):785–92.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors would like to acknowledge Karen Atkinson for editing the manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Zhenhong Li.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

ESM 1

(DOCX 1226 kb)

ESM 2

(DOCX 161 kb)

Suppl Figure S1

(BMP 1935 kb)

Suppl Figure S2

(BMP 4083 kb)

Suppl Figure S3

(BMP 3438 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, Z., Di, L. & Maurer, T.S. Theoretical Considerations for Direct Translation of Unbound Liver-to-Plasma Partition Coefficient from In Vitro to In Vivo. AAPS J 21, 43 (2019). https://doi.org/10.1208/s12248-019-0314-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1208/s12248-019-0314-1

KEY WORDS

Navigation