Skip to main content
Log in

Reduction and Lumping of Physiologically Based Pharmacokinetic Models: Prediction of the Disposition of Fentanyl and Pethidine in Humans by Successively Simplified Models

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

Abstract

Physiologically based pharmacokinetic (PBPK) models can be used to predict drug disposition in humans from animal data and the influence of disease or other changes in physiology on the pharmacokinetics of a drug. The potential usefulness of a PBPK model must however be balanced against the considerable effort needed for its development. Proposed methods to simplify PBPK modeling include predicting the necessary tissue:blood partition coefficients (kp) from physicochemical data on the drug instead of determining them in vivo, formal lumping of model compartments, and replacing the various kp values of the organs and tissues by only two values, for “fat” and “lean” tissues, respectively. The aim of this study was to investigate the effects of simplifying complex PBPK models on their ability to predict drug disposition in humans. Arterial plasma concentration curves of fentanyl and pethidine were simulated by means of a number of successively reduced models. Median absolute prediction errors were used to evaluate the performance of each model, in relation to arterial plasma concentration data from clinical studies, and the Wilcoxon matched pairs test was used for comparison of predictions. An originally diffusion-limited model for fentanyl was simplified to perfusion-limitation, and this model was either lumped, reducing 11 organ/tissue compartments to six, or changed to a model based on only two kp values, those of fat (used for fat and lungs) and muscle (used for all other tissues). None of these simplifications appreciably changed the predictions of arterial drug concentrations in the 10 patients. Perfusion-limited models for pethidine were set up using either experimentally determined [Gabrielsson et al. 1986] or theoretically calculated [Davis and Mapleson 1993] kp values, and predictions using the former were found to be significantly better. Lumping of the models did not appreciably change the predictions; however, going from a full set of kp values to only two (“fat” and “lean”) had an adverse effect. Using a kp for lungs determined either in rats or indirectly in humans [Persson et al. 1988], i.e., a total of three kp values, improved these predictions. In con- clusion, this study strongly suggested that complex PBPK models for lipophilic basic drugs may be considerably reduced with marginal loss of power to predict standard plasma pharmacokinetics in humans. Determination of only two or three kp values instead of a “full” set can mean an important reduction of experimental work to define a basic model. Organs of particular pharmacological or toxicological interest should of course be investigated separately as needed. This study also suggests and applies a simple method for statistical evaluation of the predictions of PBPK models.

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

Similar content being viewed by others

References

  1. S. B. Charnick, R. Kawai, J. R. Nedelman, M. Lemaire, W. Niederberger, and H. Sato. Physiologically based pharmacokinetic modeling as a tool for drug development. J. Pharmacokin. Biopharm. 23:217-229 (1995).

    Google Scholar 

  2. P. Poulin and F.-P. Theil. Prediction of pharmacokinetics prior to in vivo studies. 1. Mechanism-based prediction of volume of distribution. J. Pharm. Sci. 91:129-156 (2002).

    Google Scholar 

  3. A. Boobis, U. Gundert-Remy, P. Kremers, P. Macheras, and O. Pelkonen. In silico prediction of ADME and pharmacokinetics. Report of an expert meeting organized by COST B15. Eur. J. Pharm. Sci. 17:183-193 (2002).

    Google Scholar 

  4. F.-P. Theil, T. W. Guentert, S. Haddad, and P. Poulin. Utility of physiologically based pharmacokinetic models to drug development and rational drug discovery candidate selection. Toxicol. Lett. 138:29-49 (2003).

    Google Scholar 

  5. H. L. Price. A dynamic concept of the distribution of thiopental in the human body. Anesthesiology 21:40-45 (1960).

    Google Scholar 

  6. D. R. Wada, S. Björkman, W. F. Ebling, H. Harashima, S. R. Harapat, and D. R. Stanski. Computer simulation of the effects of alterations in blood flows and body composition on thiopental pharmacokinetics in humans. Anesthesiology 87:884-899 (1997).

    Google Scholar 

  7. S. Björkman, D.R. Wada, and D.R. Stanski. Application of physiological models to predict the influence of changes in body composition and blood flows on the pharmacokinetics of fentanyl and alfentanil in patients. Anesthesiology 88:657-667 (1998).

    Google Scholar 

  8. S. Björkman, D. R. Wada, B. M. Berling, and G. Benoni. Prediction of the disposition of midazolam in surgical patients by a physiologically based pharmacokinetic model. J. Pharm. Sci. 90:1226-1241 (2001).

    Google Scholar 

  9. N. R. Davis and W. W. Mapleson. A physiological model for the distribution of injected agents, with special reference to pethidine. Br. J. Anaesth. 70:248-258 (1993).

    Google Scholar 

  10. H. A. El-Masri and C. J. Portier. Physiologically based pharmacokinetic model of primidone and its metabolites phenobarbital and phenylethylmalonamide in humans, rats, and mice. Drug Metab. Dispos. 26:585-594 (1998).

    Google Scholar 

  11. P. Poulin, K. Schoenlein, and F. P. Theil. Prediction of adipose tissue:plasma partition coefficients for structurally unrelated drugs. J. Pharm. Sci. 90:436-447 (2001).

    Google Scholar 

  12. I. A. Nestorov, L. J. Aarons, P. A. Arundel, and M. Rowland. Lumping of whole-body physiologically based pharmacokinetic models. J. Pharmacokin. Biopharm. 26:21-46 (1998).

    Google Scholar 

  13. S. Björkman. Prediction of the volume of distribution of a drug: which tissue:plasma partition coefficients are needed? J. Pharm. Pharmacol. 54:1237-1245 (2002).

    Google Scholar 

  14. R. J. Hudson, I. R. Thomson, J. E. Cannon, R. M. Friesen, and R. C. Meatherall. Pharmacokinetics of fentanyl in patients undergoing abdominal aortic surgery. Anesthesiology 64:334-338 (1986).

    Google Scholar 

  15. F. Redke and S. Björkman. Pharmacokinetics of pethidine in patients with postoperative fever [abstract]. Eur. J. Pharm. Sci. 8:xix(1999).

    Google Scholar 

  16. S. Björkman, D. R. Stanski, H. Harashima, R. Dowrie, S. R. Harapat, D. R. Wada, and W. F. Ebling. Tissue distribution of fentanyl and alfentanil in the rat cannot be described by a blood flow limited model. J. Pharmacokin. Biopharm. 21:255-279 (1993).

    Google Scholar 

  17. S. Björkman, D. R. Wada, D. R. Stanski, and W. F. Ebling. Comparative physiological pharmacokinetics of fentanyl and alfentanil in rats and humans based on parametric single-tissue models. J. Pharmacokin. Biopharm. 22:381-410 (1994).

    Google Scholar 

  18. S. Björkman, D. R. Stanski, D. Verotta, and H. Harashima. Comparative tissue concentration profiles of fentanyl and alfentanil in humans predicted from tissue/blood partition data obtained in rats. Anesthesiology 72:865-873 (1990).

    Google Scholar 

  19. J. L. Gabrielsson, P. Johansson, U. Bondesson, M. Karlsson, and L. K. Paalzow. Analysis of pethidine disposition in the pregnant rat by means of a physiological flow model. J. Pharmacokin. Biopharm. 14:381-395 (1986).

    Google Scholar 

  20. M. P. Persson, P. Hartvig, L. Wiklund, and L. Paalzow. Pulmonary disposition of pethidine in postoperative patients. Br. J. Clin. Pharmacol. 25:235-241 (1988).

    Google Scholar 

  21. T. H. Hallynck, H. H. Soep, J. A. Thomis, J. Boelaert, R. Daneels, and L. Dettli. Should clearance be normalized to body surface or to lean body mass? Br. J. Clin. Pharmacol. 11:523-526 (1981).

    Google Scholar 

  22. L. R. Williams and R. W. Leggett. Reference values for resting blood flow to organs of man. Clin. Phys. Physiol. Meas. 10:187-217 (1989).

    Google Scholar 

  23. S. Björkman and F. Redke. Clearance of fentanyl, alfentanil, methohexitone, thiopentone, and ketamine in relation to estimated hepatic blood flow in several animal species: application to prediction of clearance in man. J. Pharm. Pharmacol. 52:1065-1074 (2000).

    Google Scholar 

  24. D. R. Wada, D. R. Stanski, and W. F. Ebling. A PC-based graphical simulator for physiological pharmacokinetic models. Comp. Meth. Progr. Biomed. 46:245-255 (1995).

    Google Scholar 

  25. W. F. Ebling, D. R. Wada, and D. R. Stanski. From piecewise to full physiologic pharmacokinetic modeling: Applied to thiopental disposition in the rat. J. Pharmacokin. Biopharm. 22:259-292 (1994).

    Google Scholar 

  26. A. Tsuji, K. Nishide, H. Minami, E. Nakashima, T. Terasaki, and T. Yamana. Physiologically based pharmacokinetic model for cefazolin in rabbits and its preliminary extrapolation to man. Drug Metab. Disp. 13:729-739 (1985).

    Google Scholar 

  27. O. Nagata, M. Murata, H. Kato, T. Terasaki, H. Sato, and A. Tsuji. Physiological pharmacokinetics of a new muscle-relaxant, inaperisone, combined with its pharmacological effect on blood flow rate. Drug Metab. Disp. 18:902-910 (1990).

    Google Scholar 

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

    Google Scholar 

  29. G. Wu. Calculating predictive performance: a user's note. Pharmacol. Res. 31:393-399 (1995).

    Google Scholar 

  30. P. M. Vermeulen, J. G. C. Lerou, R. Dirksen, L. H. D. J. Booij, and G. F. Borm. Repeated enflurane anesthetics and model predictions: a study of the variability in the predictive performance measures. Br. J. Anaesth. 79:488-496 (1997).

    Google Scholar 

  31. N. Shibata, W. Gao, H. Okamoto, T. Kishida, K. Iwasaki, Y. Yoshikawa, and K. Takada. Drug interactions between HIV protease inhibitors based on physiologically-based pharmacokinetic model. J. Pharm. Sci. 91:680-689 (2002).

    Google Scholar 

  32. F. Y. Bois, T. J. Woodruff, and R. C. Spear. Comparison of three physiologically based pharmacokinetic models of benzene disposition. Toxicol. Appl. Pharmacol. 110:79-88 (1991).

    Google Scholar 

  33. P. Poulin and F.-P. Theil. Prediction of pharmacokinetics prior to in vivo studies. II. Generic physiologically based pharmacokinetic models of drug disposition. J. Pharm. Sci. 91:1358-1370 (2002).

    Google Scholar 

  34. R. K. Verbeeck, R. A. Branch, and G. R. Wilkinson. Meperidine disposition in man: influence of urinary pH and route of administration. Clin. Pharmacol. Ther. 30:619-628 (1981).

    Google Scholar 

  35. K. Chan, J. Tse, F. Jennings, and M. L. E. Orme. Influence of urinary pH on pethidine kinetics in healthy volunteer subjects. Meth. Find. Exptl. Clin. Pharmacol. 7:245-251 (1985).

    Google Scholar 

  36. L. E. Mather and P. J. Meffin. Clinical pharmacokinetics pethidine. Clin. Pharmacokin. 3:352-368 (1978).

    Google Scholar 

  37. B. E. Dahlström, L. K. Paalzow, C. Lindberg, and C. Bogentoft. Pharmacokinetics and analgesic effect of pethidine (meperidine) and its metabolites in the rat. Drug Metab. Disp. 7:108-112 (1979).

    Google Scholar 

  38. W. G. Kramer, D. R. Gross, and C. Medlock. Contribution of the lung to total body clearance of meperidine in the dog. J. Pharm. Sci. 74:569-571 (1985).

    Google Scholar 

  39. B. Ranheim, J. Høiset, T. Framstad, T. E. Horsberg, J. U. Skaare, and N. E. Søli. Pharmacokinetics of pethidine in pigs following intravenous, intramuscular, and subcutaneous administration. J. Vet. Pharmacol. Ther. 21:491-493 (1998).

    Google Scholar 

  40. R. N. Upton, L. E. Mather, and W. B. Runciman. The in vitro uptake and metabolism of lignocaine, procainamide, and pethidine by tissues of the hindquarters of sheep. Xenobiotica 21:1-12 (1991).

    Google Scholar 

  41. J. C. Scott, K. V. Ponganis, and D. R. Stanski. EEG quantitation of narcotic effect: the comparative pharmacodynamics of fentanyl and alfentanil. Anesthesiology 62:234-241 (1985).

    Google Scholar 

  42. J. C. Scott and D. R. Stanski. Decreased fentanyl and alfentanil dose requirements with age. A simultaneous pharmacokinetic and pharmacodynamic evaluation. J. Pharmacol. Exp. Ther. 240:159-166 (1987).

    Google Scholar 

  43. H. J. M. Lemmens, J. B. Dyck, S. L. Shafer, and D. R. Stanski. Pharmacokinetic–pharmacodynamic modeling in drug development: application to the investigational opioid trefentanil. Clin. Pharmacol. Ther. 56:261-271 (1994).

    Google Scholar 

  44. C. C. Hug. Lipid solubility, pharmacokinetics, and the EEG: are you better off today than you were four years ago? Anesthesiology 62:221-226 (1985).

    Google Scholar 

  45. R. N. Upton, G. L. Ludbrook, E. C. Gray, and C. Grant. The cerebral pharmacokinetics of meperidine and alfentanil in conscious sheep. Anesthesiology 86:1317-1325 (1997).

    Google Scholar 

  46. G. L. Qiao and K. F. Fung. Pharmacokinetic–pharmacodynamic modelling of meperidine in goats (II): modelling. J. Vet. Pharmacol. Ther. 17:127-134 (1994).

    Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Sven Björkman.

Rights and permissions

Reprints and permissions

About this article

Cite this article

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 30, 285–307 (2003). https://doi.org/10.1023/A:1026194618660

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1023/A:1026194618660

Navigation