Skip to main content
Log in

Quantitative Structure–Pharmacokinetics Relationships: II. A Mechanistically Based Model to Evaluate the Relationship Between Tissue Distribution Parameters and Compound Lipophilicity

  • Published:
Journal of Pharmacokinetics and Biopharmaceutics Aims and scope Submit manuscript

Abstract

The tissue-to-unbound plasma distribution coefficients (Kpus) of 14 rat tissues after iv administration of nine 5-n-alkyl-5-ethyl barbituric acids, determined in a previous study, were used to identify a model of the relationship between tissue distribution and lipophilicity of the homologs, expressed in terms of their octanol to water partition ratio, P. Based on mechanistic considerations and assumptions, the parameter model was expressed asKpuτ= fW,τ[l + aτ(nPl,τ)P], where fW,τ is the tissue water content, (nPl,τ ) is the binding capacity of the tissue, n is the number of the binding sites, aτ and bτ are the parameters of the relationship Kaτ = aτP and Kaτ is the binding association constant of each tissue. The parameter model was linearized and fitted to the predetermined Kpu values, yielding correlation coefficients ranging between .940 and .997. The predictive performance of the parameter model was evaluated using a leave-one-out procedure with subsequent computation of the mean prediction error (ME = measurement of the prediction bias) and the square root of the mean squared prediction error (RMSE = measurement of the prediction accuracy). The ME varied between −22.48 and 61.14%, indicating a slight tendency for overpredicting. The RMSE was between 24.73 and 102% for the individual tissues across the different homologs; and between 28.33 and 85.2% for the individual homologs across the different tissues. The apparently high Kpu prediction errors, when translated through the low sensitivity of the barbiturate whole-body physiologically based pharmacokinetic model, established previously, leads to predicted tissue concentration–time profiles within 5 to 20% of the original ones. Therefore, it is concluded, that the identified mechanistically based model is a good predictor of the tissue-to-unbound Kpus in the rat tissues.

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.

Similar content being viewed by others

REFERENCES

  1. L. W. Frick, K. K. Adkinson, K. J. Wells-Knecht, P. Woolland, and D. M. Highton. Cassette dosing: rapid in vivo assessment of pharmacokinetics. PSTT 1:12–18 (1998).

    CAS  Google Scholar 

  2. J. Berman, K. Halm, K. Adkinson, and J. Shaffer. Simultaneous pharmacokinetic screening of a mixture of compounds in the dig using API LC/MS/MS analysis for increased throughput. J. Med. Chem. 40:827–829 (1997).

    Article  CAS  PubMed  Google Scholar 

  3. J. K. Seydel and K-J. Schaper. Quantitative structure-pharmacokinetic relationships and drug design. In M. Rowland and G. Tucker (eds.), Pharmacokinetics: Theory and Methodology. International Encyclopedia of Pharmacology and Therapeutics, Section 122, Pergamon, Oxford, 1986, pp. 311–368.

    Google Scholar 

  4. G. E. Blakey, I. A. Nestorov, P. A. Arundel, L. J. Aarons, and M. Rowland. Quantitative structure-pharmacokinetics relationships: I. Development of a whole-body physiologically based model to characterize changes in pharmacokinetics across a homologous series of barbiturates in the rat. J. Pharmacokin. Biopharm. 25:277–312 (1997).

    Article  CAS  Google Scholar 

  5. S. Toon and M. Rowland. Structure-activity relationships among the barbiturates in the rat. J. Pharmacol. Exp. Ther. 225:752–763 (1983).

    CAS  PubMed  Google Scholar 

  6. M. Rowland and T. N. Tozer. Clinical Pharmacokinetics: Concepts and Applications, 3rd ed., Williams and Wilkins, Philadelphia, 1995, p. 600.

    Google Scholar 

  7. P. H. Hinderling. Red blood cells: A neglected compartment in pharmacokinetics and pharmacodynamics. Pharmacol. Rev. 49:279–295 (1997).

    CAS  PubMed  Google Scholar 

  8. I. A. Nestorov, L. J. Aarons, P. A. Arundel, and M. Rowland. Lumping of whole-body physiologically based pharmacokinetic models. J. Pharmacokin. Biopharm. 26:23–46 (1998).

    Google Scholar 

  9. K. Yokogawa, E. Nakashima, J. Ishizaki, H. Maeda, T. Nagano, and F. Ichimura. Relationships in the structure-tissue distribution of basic drugs in the rabbit. Pharm. Res. 7:691–696 (1990).

    Article  CAS  PubMed  Google Scholar 

  10. J. Ishizaki, K. Yokogawa, E. Nakashima, and F. Ichimura. Relationships between the hepatic intrinsic clearance or blood cell-plasma partition coefficient in the rabbit and the lipophilicity of basic drugs. J. Pharm. Pharmacol. 49:768–772 (1997).

    Article  CAS  PubMed  Google Scholar 

  11. J. Ishizaki, K. Yokogawa, E. Nakashima, and F. Ichimura. Prediction of changes in the clinical pharmacokinetics of basic drugs on the basis of octanol-water partition coefficients. J. Pharm. Pharmacol. 49:762–767 (1997).

    Article  CAS  PubMed  Google Scholar 

  12. R. F. Reinoso, B. A. Telfer, and M. Rowland. Tissue water contents in rats measured by desiccation. J. Pharmacol. Toxicol. Meth. 38:87–92 (1997).

    Article  CAS  Google Scholar 

  13. L. B. Sheiner and S. Beal. Some suggestions for measuring predictive performance. J. Pharmacokin. Biopharm. 9:503–512 (1981).

    Article  CAS  Google Scholar 

  14. ACSL Reference Manual, Edition 11. MGA Software, Concord MA 01742, USA, 1995.

  15. A. Leo and C. Hansch. Linear free-energy relationships between partitioning solvent systems. J. Org. Chem. 36:1539–1544 (1971).

    Article  CAS  Google Scholar 

  16. K. B. Bischoff and R. L. Dedrick. Thiopental pharmacokinetics. J. Pharm. Sci. 57:1346–1351 (1968).

    Article  CAS  PubMed  Google Scholar 

  17. P. Barton, A. M. Davis, D. J. McCarthy, and P. J. H. Webborn. Drug-Phospholipid Interactions: 2. Predicting the sites of drug distribution using n-octanol/water and membrane/water distribution coefficient. J. Pharm. Sci. 86:1034–1039 (1997).

    Article  CAS  PubMed  Google Scholar 

  18. T. D. Yih and J. M. Van Rossum. Pharmacokinetics of some homologous series of barbiturates in the intact rat and in the isolated perfused rat liver. J. Pharmacol. Exp. Ther. 203:182–763 (1977).

    Google Scholar 

  19. J. M. Mayer, S. D. Hall, and M. Rowland. Relationship between lipophilicity and tubular reabsorption for a series of 5–alkyl-5–ethyl barbituric acids in the isolated perfused rat kidney preparation. J. Pharm. Sci. 77:359–364 (1988).

    Article  CAS  PubMed  Google Scholar 

  20. R. J. Prankerd and R. H. McKeown. Physicochemical properties of barbituric acid derivatives: 2. Partition coefficients of 5,5–disubstituted barbituric acids at 25°C. Int. J. Pharm. 83:25–37 (1992).

    Article  CAS  Google Scholar 

  21. R. Pinal and S. H. Yalkowsky. Solubility and partitioning VII: Solubility of barbiturates in water. J. Pharm. Sci. 76:75–85 (1987).

    Article  CAS  PubMed  Google Scholar 

  22. S. H. Steiner, M. J. Moor, and M. H. Bickel. Kinetics of distribution and adipose tissue storage as a function of lipophilicity and chemical structure: 1. Barbiturates. Drug Metab. Dispos. 19:8–14 (1991).

    CAS  PubMed  Google Scholar 

  23. B. B. Brodie, E. Bernstein, and L. C. Mark. The role of body fat in limiting the duration of action of thiopental. J. Pharm. Pharmacol. 105:421–426 (1952).

    CAS  Google Scholar 

  24. J. M. Gallo, F. C. Lam, and D. G. Perrier. Area method for the estimation of partition coefficients for physiological pharmacokinetic models. J. Pharmacokinet. Biopharm. 15:271–280 (1987).

    Article  CAS  PubMed  Google Scholar 

  25. D. Verotta, L. B. Sheiner, W. F. Ebling, and D. Stanski. A semiparametric approach to physiological flow models. J. Pharmacokinet. Biopharm. 17:463–491 (1989).

    Article  CAS  PubMed  Google Scholar 

  26. P. Poulin and K. Krishnan. A tissue composition-based algorithm for predicting tissue:air partition coefficients of organic chemicals. Toxicol. Appl. Pharmacol. 136:126–130 (1996).

    Article  CAS  PubMed  Google Scholar 

  27. M. Pelekis, P. Poulin, and K. Krishnan. An approach for incorporating tissue composition data into physiologically based pharmacokinetic models. Toxicol. Ind. Health 11:511–522 (1995).

    Article  CAS  PubMed  Google Scholar 

  28. F. M. Parham, M. C. Kohn, H. B. Matthews, C. DeRosa, and C. J. Portier. Using structural information to create physiologically based pharmacokinetic models for all polychlorinated biphenyls. Toxicol. Appl. Pharmacol. 144:340–347 (1997).

    Article  CAS  PubMed  Google Scholar 

  29. KOWWIN. Octanol-Water Partition Coefficient Program. User's Guide, Syracuse Research Corporation, Merill Lane, Syracuse, NY, 1996.

    Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Rights and permissions

Reprints and permissions

About this article

Cite this article

Nestorov, I., Aarons, L. & Rowland, M. Quantitative Structure–Pharmacokinetics Relationships: II. A Mechanistically Based Model to Evaluate the Relationship Between Tissue Distribution Parameters and Compound Lipophilicity. J Pharmacokinet Pharmacodyn 26, 521–545 (1998). https://doi.org/10.1023/A:1023221116200

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1023/A:1023221116200

Navigation