Skip to main content
Log in

Determination of the Number of Tissue Groups of Kinetically Distinct Transit Time in Whole-Body Physiologically Based Pharmacokinetic (PBPK) Models II: Practical Application of Tissue Lumping Theories for Pharmacokinetics of Various Compounds

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

Abstract

In our companion paper, we described the theoretical basis for tissue lumping in whole-body physiologically based pharmacokinetic (WB-PBPK) models and found that Kdet, a coefficient for determining the number of tissue groups of distinct transit time in WB-PBPK models, was related to the fractional change in the terminal slope (FCT) when tissues were progressively lumped from the longest transit time to shorter ones. This study was conducted to identify the practical threshold of Kdet by applying the lumping theory to plasma/blood concentration-time relationships of 113 model compounds collected from the literature. We found that drugs having Kdet < 0.3 were associated with FCT < 0.1 even when all peripheral tissues were lumped, resulting in comparable plasma concentration-time profiles between one-tissue minimal PBPK (mPBPK) and WB-PBPK models. For drugs with Kdet ≥ 1, WB-PBPK profiles appeared similar with two-tissue mPBPK models by applying the rule of FCT < 0.1 for lumping slowly equilibrating tissues. The two-tissue mPBPK model also appeared appropriate in terms of concentration-time profiles for drugs with 0.3 ≤ Kdet < 1, although, some compounds (15.9% of the total cases), but not all, in this range showed a slight (maximum of 18.9% of the total AUC) deviation from WB-PBPK models, indicating that the two-tissue model, with caution, could still be used for those cases. Comparison of kinetic parameters between traditional (model-fitting) and current (theoretical calculation) mPBPK analyses revealed their significant correlations. Collectively, these observations suggest that the number of tissue groups could be determined based on the Kdet/FCT criteria, and plasma concentration-time profiles from WB-PBPK could be calculated using equations significantly less complex.

Graphical abstract

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.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

References

  1. Levy G, Gibaldi M, Jusko WJ. Multicompartment pharmacokinetic models and pharmacologic effects. J Pharm Sci. 1969;58(4):422–4.

    Article  CAS  Google Scholar 

  2. Benet LZ. General treatment of linear mammillary models with elimination from any compartment as used in pharmacokinetics. J Pharm Sci. 1972;61(4):536–41.

    Article  CAS  Google Scholar 

  3. Cao Y, Jusko WJ. Applications of minimal physiologically-based pharmacokinetic models. J Pharmacokinet Pharmacodyn. 2012;39(6):711–23. https://doi.org/10.1007/s10928-012-9280-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Sager JE, Yu J, Ragueneau-Majlessi I, Isoherranen N. Physiologically based pharmacokinetic (PBPK) modeling and simulation approaches: a systematic review of published models, applications, and model verification. Drug Metab Dispos. 2015;43(11):1823–37.

    Article  CAS  Google Scholar 

  5. Riegelman S, Loo J, Rowland M. Shortcomings in pharmacokinetic analysis by conceiving the body to exhibit properties of a single compartment. J Pharm Sci. 1968;57(1):117–23.

    Article  CAS  Google Scholar 

  6. Hirtz J. The fate of drugs in the organism. A bibliographic survey complied by the Societe´ Fran¸aise des Sciences et Techniques Pharmaceutique, Working group under the chairmanship of HIRTZ. Dekker New York; 1974.

  7. Segre G. Pharmacokinetics—compartmental representation. Pharmacol Ther. 1982;17(1):111–27.

    Article  CAS  Google Scholar 

  8. Vaughan D, Dennis M. Number of exponential terms describing the solution of an N-compartmental mammillary model: Vanishing exponentials. J Pharmacokinet Biopharm. 1979;7(5):511–25.

    Article  CAS  Google Scholar 

  9. Wagner JG. Linear pharmacokinetic models and vanishing exponential terms: Implications in pharmacokinetics. J Pharmacokinet Biopharm. 1976;4(5):395–425.

    Article  CAS  Google Scholar 

  10. Jeong Y-S, Kim M-S, Chung S-J. Determination of the number of tissue groups of kinetically distinct transit time in whole-body physiologically-based pharmacokinetic (PBPK) models I: Theoretical consideration of bottom-up approach of lumping tissues in whole-body PBPK. AAPS J. 2022. https://doi.org/10.1208/s12248-022-00732-y.

    Article  PubMed  Google Scholar 

  11. Lobell M, Sivarajah V. In silico prediction of aqueous solubility, human plasma protein binding and volume of distribution of compounds from calculated pKa and AlogP98 values. Mol Diversity. 2003;7(1):69–87.

    Article  CAS  Google Scholar 

  12. Jamei M, Marciniak S, Feng K, Barnett A, Tucker G, Rostami-Hodjegan A. The Simcyp® population-based ADME simulator. Expert Opin Drug Metab Toxicol. 2009;5(2):211–23.

    Article  CAS  Google Scholar 

  13. Colclough N, Ruston L, Wood JM, MacFaul PA. Species differences in drug plasma protein binding. Med Chem Commun. 2014;5(7):963–7.

    Article  CAS  Google Scholar 

  14. Uchimura T, Kato M, Saito T, Kinoshita H. Prediction of human blood-to-plasma drug concentration ratio. Biopharm Drug Dispos. 2010;31(5-6):286–97. https://doi.org/10.1002/bdd.711.

    Article  CAS  PubMed  Google Scholar 

  15. Kerns EH, Di L, Petusky S, Farris M, Ley R, Jupp P. Combined application of parallel artificial membrane permeability assay and Caco-2 permeability assays in drug discovery. J Pharm Sci. 2004;93(6):1440–53.

    Article  CAS  Google Scholar 

  16. Fujikawa M, Ano R, Nakao K, Shimizu R, Akamatsu M. Relationships between structure and high-throughput screening permeability of diverse drugs with artificial membranes: application to prediction of Caco-2 cell permeability. Bioorg Med Chem. 2005;13(15):4721–32. https://doi.org/10.1016/j.bmc.2005.04.076.

    Article  CAS  PubMed  Google Scholar 

  17. Jeong Y-S, Yim C-S, Ryu H-M, Noh C-K, Song Y-K, Chung S-J. Estimation of the minimum permeability coefficient in rats for perfusion-limited tissue distribution in whole-body physiologically-based pharmacokinetics. Eur J Pharm Biopharm. 2017;115:1–17.

    Article  CAS  Google Scholar 

  18. Rodgers T, Leahy D, Rowland M. Physiologically based pharmacokinetic modeling 1: Predicting the tissue distribution of moderate-to-strong bases. J Pharm Sci. 2005;94(6):1259–76. https://doi.org/10.1002/jps.20322.

    Article  CAS  PubMed  Google Scholar 

  19. Rodgers T, Rowland M. Physiologically based pharmacokinetic modelling 2: predicting the tissue distribution of acids, very weak bases, neutrals and zwitterions. J Pharm Sci. 2006;95(6):1238–57. https://doi.org/10.1002/jps.20502.

    Article  CAS  PubMed  Google Scholar 

  20. Øie S, Tozer TN. Effect of altered plasma protein binding on apparent volume of distribution. J Pharm Sci. 1979;68(9):1203–5.

    Article  Google Scholar 

  21. Brown R, Delp M, Lindstedt S, Rhomberg L, Beliles R. Physiological parameter values for physiologically based pharmacokinetic models. Toxicol Ind Health. 1997;13407:407–84.

    Article  Google Scholar 

  22. Nestorov IA, Aarons LJ, Arundel PA, Rowland M. Lumping of whole-body physiologically based pharmacokinetic models. J Pharmacokinet Biopharm. 1998;26(1):21–46.

    Article  CAS  Google Scholar 

  23. Veng-Pedersen P, Gillespie WR. Single pass mean residence time in peripheral tissues: a distribution parameter intrinsic to the tissue affinity of a drug. J Pharm Sci. 1986;75(12):1119–26.

    Article  CAS  Google Scholar 

  24. Pilari S, Huisinga W. Lumping of physiologically-based pharmacokinetic models and a mechanistic derivation of classical compartmental models. J Pharmacokinet Pharmacodyn. 2010;37(4):365–405.

    Article  CAS  Google Scholar 

  25. Gueorguieva I, Nestorov IA, Rowland M. Reducing whole body physiologically based pharmacokinetic models using global sensitivity analysis: diazepam case study. J Pharmacokinet Pharmacodyn. 2006;33(1):1–27.

    Article  CAS  Google Scholar 

  26. Björkman S. Reduction and lumping of physiologically based pharmacokinetic models: prediction of the disposition of fentanyl and pethidine in humans by successively simplified models. J Pharmacokinet Pharmacodyn. 2003;30(4):285–307.

    Article  Google Scholar 

  27. Brochot C, Tóth J, Bois FY. Lumping in pharmacokinetics. J Pharmacokinet Pharmacodyn. 2005;32(5-6):719–36.

    Article  Google Scholar 

  28. Jeong Y-S, Jusko WJ. Meta-assessment of metformin absorption and disposition pharmacokinetics in nine species. Pharmaceuticals. 2021;14(6):545.

    Article  CAS  Google Scholar 

  29. Kong AN, Jusko WJ. Definitions and applications of mean transit and residence times in reference to the two-compartment mammillary plasma clearance model. J Pharm Sci. 1988;77(2):157–65.

    Article  CAS  Google Scholar 

  30. Berezhkovskiy LM. Prediction of drug terminal half-life and terminal volume of distribution after intravenous dosing based on drug clearance, steady-state volume of distribution, and physiological parameters of the body. J Pharm Sci. 2013;102(2):761–71.

    Article  CAS  Google Scholar 

  31. Fleishaker JC, Smith RB. Compartmental model analysis in pharmacokinetics. J Clin Pharmacol. 1987;27(12):922–6.

    Article  CAS  Google Scholar 

Download references

Funding

This study was supported by the Korea Environment Industry & Technology Institute (KEITI) through the project for BioMarkers TRANSLation of consumer chemicals/ft.life-stage PBPK modeling (BioTranSL/PBPK) (NO. 2022002970003).

Author information

Authors and Affiliations

Authors

Contributions

Yoo-Seong Jeong: Conceptualization, methodology, formal analysis, investigation, data curation, writing-original draft, writing-review & editing, visualization

Min-Soo Kim: Methodology, software, formal analysis, data curation, writing-review & editing, visualization

Suk-Jae Chung: Conceptualization, methodology, formal analysis, writing-original draft, writing-review & editing, supervision, project administration, funding acquisition

Corresponding author

Correspondence to Suk-Jae Chung.

Ethics declarations

Conflict of Interest

The authors declare no competing interests.

Additional information

Publisher’s Note

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

Supplementary Information

ESM 1

(DOCX 2089 kb)

ESM 2

(XLSX 64319 kb)

Appendix

Appendix

By applying Gauss elimination of the system matrix A of PBPK Model C [10], 10 eigenvalues were readily determined using Eq. 11:

$$\frac{\frac{Q_1{f}_{d1}}{MT{T}_1}}{\lambda -\frac{1}{MT{T}_1}}+\frac{\frac{Q_2{f}_{d2}}{MT{T}_2}}{\lambda -\frac{1}{MT{T}_2}}+\dots +\frac{\frac{Q_9{f}_{d9}}{MT{T}_9}}{\lambda -\frac{1}{MT{T}_9}}={V}_B\left(\lambda -\frac{1}{MT{T}_c}\right)$$
(11)

When the two-tissue mPBPK model (Model D) is applicable, the eigenvalues (in terms of λ; the roots corresponded to λα, λβ, and λγ) after tissue lumping could be determined by Eq. 12:

$$\frac{\frac{Q_{SEG}{f}_{d, SEG}}{{ MT T}_{SEG}}}{\lambda^{\prime }-\frac{1}{{ MT T}_{SEG}}}+\frac{\frac{Q_{REG}{f}_{d, REG}}{{ MT T}_{REG}}}{\lambda^{\prime }-\frac{1}{{ MT T}_{REG}}}\kern0.75em =\kern0.75em {V}_B\left({\lambda}^{\prime }-\frac{1}{MT{T}_c}\right)$$
(12)

Rights and permissions

Springer Nature or its licensor holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jeong, YS., Kim, MS. & Chung, SJ. Determination of the Number of Tissue Groups of Kinetically Distinct Transit Time in Whole-Body Physiologically Based Pharmacokinetic (PBPK) Models II: Practical Application of Tissue Lumping Theories for Pharmacokinetics of Various Compounds. AAPS J 24, 91 (2022). https://doi.org/10.1208/s12248-022-00733-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1208/s12248-022-00733-x

Keywords

Navigation