Skip to main content
Log in

Optimal Sampling Times for a Drug and its Metabolite using SIMCYP® Simulations as Prior Information

Clinical Pharmacokinetics Aims and scope Submit manuscript

Abstract

Background

Since 2007, it is mandatory for the pharmaceutical companies to submit a Paediatric Investigation Plan to the Paediatric Committee at the European Medicines Agency for any drug in development in adults, and it often leads to the need to conduct a pharmacokinetic study in children. Pharmacokinetic studies in children raise ethical and methodological issues. Because of limitation of sampling times, appropriate methods, such as the population approach, are necessary for analysis of the pharmacokinetic data. The choice of the pharmacokinetic sampling design has an important impact on the precision of population parameter estimates. Approaches for design evaluation and optimization based on the evaluation of the Fisher information matrix (MF) have been proposed and are now implemented in several software packages, such as PFIM in R.

Objectives

The objectives of this work were to (1) develop a joint population pharmacokinetic model to describe the pharmacokinetic characteristics of a drug S and its active metabolite in children after intravenous drug administration from simulated plasma concentration–time data produced using physiologically based pharmacokinetic (PBPK) predictions; (2) optimize the pharmacokinetic sampling times for an upcoming clinical study using a multi-response design approach, considering clinical constraints; and (3) evaluate the resulting design taking data below the lower limit of quantification (BLQ) into account.

Methods

Plasma concentration–time profiles were simulated in children using a PBPK model previously developed with the software SIMCYP® for the parent drug and its active metabolite. Data were analysed using non-linear mixed–effect models with the software NONMEM®, using a joint model for the parent drug and its metabolite. The population pharmacokinetic design, for the future study in 82 children from 2 to 18 years old, each receiving a single dose of the drug, was then optimized using PFIM, assuming identical times for parent and metabolite concentration measurements and considering clinical constraints. Design evaluation was based on the relative standard errors (RSEs) of the parameters of interest. In the final evaluation of the proposed design, an approach was used to assess the possible effect of BLQ concentrations on the design efficiency. This approach consists of rescaling the MF, using, at each sampling time, the probability of observing a concentration BLQ computed from Monte-Carlo simulations.

Results

A joint pharmacokinetic model with three compartments for the parent drug and one for its active metabolite, with random effects on four parameters, was used to fit the simulated PBPK concentration–time data. A combined error model best described the residual variability. Parameters and dose were expressed per kilogram of bodyweight. Reaching a compromise between PFIM results and clinical constraints, the optimal design was composed of four samples at 0.1, 1.8, 5 and 10 h after drug injection. This design predicted RSE lower than 30 % for the four parameters of interest. For this design, rescaling MF for BLQ data had very little influence on predicted RSE.

Conclusion

PFIM was a useful tool to find an optimal sampling design in children, considering clinical constraints. Even if it was not forecasted initially by the investigators, this approach showed that it was really necessary to include a late sampling time for all children. Moreover, we described an approach to evaluate designs assuming expected proportions of BLQ data are omitted.

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
Fig. 4
Fig. 5

References

  1. B Regulation (EC) no 1901/2006 of the European Parliament and of the Council of 12 December 2006 on medicinal products for paediatric use and amending Regulation (EEC) no 1768/92, Directive 2001/20/EC, Directive 2001/83/EC and Regulation (EC) No 726/2004. 2012. http://eur-lex.europa.eu/LexUriServ/site/en/consleg/2006/R/02006R1901-20070126-en.pdf. Accessed 5 Nov 2012.

  2. Committee for Medicinal Products for Human Use (CHMP). Guideline on the role of pharmacokinetics in the development of medicinal products in the paediatric population. London, 28 June 2006. Doc. Ref. EMEA/CHMP/EWP/147013/2004. Corrigendum. http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2009/09/WC500003066.pdf. Accessed 5 Nov 2012.

  3. Bouzom F, Walther B. Pharmacokinetic predictions in children by using the physiologically based pharmacokinetic modelling. Fundam Clin Pharmacol. 2008;22:579–87.

    Article  PubMed  CAS  Google Scholar 

  4. Mentré F, Dubruc C, Thénot JP. Population pharmacokinetic analysis and optimization of the experimental design for mizolastine solution in children. J Pharmacokinet Pharmacodyn. 2001;28:299–319.

    Article  PubMed  Google Scholar 

  5. Tod M, Jullien V, Pons G. Facilitation of drug evaluation in children by population methods and modelling. Clin Pharmacokinet. 2008;47:231–43.

    Article  PubMed  CAS  Google Scholar 

  6. Mentré F, Mallet A, Baccar D. Optimal design in random-effects regression models. Biometrika. 1997;84:429–42.

    Article  Google Scholar 

  7. Bazzoli C, Retout S, Mentré F. Fisher information matrix for nonlinear mixed effects multiple response models: evaluation of the appropriateness of the first order linearization using a pharmacokinetic/pharmacodynamic model. Stat Med. 2009;28:1940–56.

    Article  PubMed  Google Scholar 

  8. Retout S, Mentré F, Bruno R. Fisher information matrix for non-linear mixed-effects models: evaluation and application for optimal design of enoxaparin population pharmacokinetics. Stat Med. 2002;21:2623–39.

    Article  PubMed  Google Scholar 

  9. Gueorguieva I, Ogungbenro K, Graham G, et al. A program for individual and population optimal design for univariate and multivariate response pharmacokinetic and pharmacodynamic models. Comput Methods Programs Biomed. 2007;86:51–61.

    Article  PubMed  Google Scholar 

  10. Mentré F, Burtin Y, Merlé Y, et al. Sparse-sampling optimal designs in pharmacokinetics and toxicokinetics. Drug Inf J. 1995;29:997–1019.

    Google Scholar 

  11. Retout S, Duffull S, Mentré F. Development and implementation of the population Fisher information matrix for evaluation of population pharmacokinetic designs. Comput Methods Programs Biomed. 2001;65:141–51.

    Article  PubMed  CAS  Google Scholar 

  12. PFIM. 2012. http://www.pfim.biostat.fr. Accessed 5 Nov 2012.

  13. Bazzoli C, Retout S, Mentré F. Design evaluation and optimisation in multiple response nonlinear mixed effects models: PFIM 3.0. Comput Methods Programs Biomed. 2010;98:55–65.

    Article  PubMed  Google Scholar 

  14. Jamei M, Marciniak S, Feng K, et al. The Simcyp population-based ADME simulator. Expert Opin Drug Metab Toxicol. 2009;5:211–23.

    Article  PubMed  CAS  Google Scholar 

  15. Johnson TN, Rostami-Hodjegan A, Tucker GT. Prediction of the clearance of eleven drugs and associated variability in neonates, infants and children. Clin Pharmacokinet. 2006;45:931–56.

    Article  PubMed  CAS  Google Scholar 

  16. Johnson TN, Rostami-Hodjegan A. Resurgence in the use of physiologically based pharmacokinetic models in pediatric clinical pharmacology: parallel shift in incorporating the knowledge of biological elements and increased applicability to drug development and clinical practice. Pediatr Anesth. 2011;21:291–301.

    Article  Google Scholar 

  17. Rostami-Hodjega A, Tucker GT. Simulation and prediction of in vivo drug metabolism in human populations from in vitro data. Nat Rev Drug Discov. 2007;6:140–8.

    Article  Google Scholar 

  18. Rowland M, Peck C, Tucker G. Physiologically-based pharmacokinetics in drug development and regulatory science. Annu Rev Pharmacol Toxicol. 2011;51:45–73.

    Article  PubMed  CAS  Google Scholar 

  19. Zhao P, Zhang L, Grillo JA, et al. Applications of physiologically based pharmacokinetic (PBPK) modeling and simulation during regulatory review. Clin Pharmacol Ther. 2011;89:259–67.

    Article  PubMed  CAS  Google Scholar 

  20. Huang SM, Rowland M. The role of physiologically based pharmacokinetic modeling in regulatory review. Clin Pharmacol Ther. 2012;91:542–9.

    Article  PubMed  CAS  Google Scholar 

  21. Rowland M, Balant L, Peck C. Physiologically based pharmacokinetics in drug development and regulatory science: a workshop report (Georgetown University, Washington, DC, May 29–30, 2002). AAPS Pharm Sci. 2004;6:E6–12.

    Article  Google Scholar 

  22. Bjorkman S. Prediction of drug disposition in infants and children by means of physiologically based pharmacokinetic (PBPK) modelling: theophylline and midazolam as model drugs. Br J Clin Pharmacol. 2006;59:691–704.

    Article  Google Scholar 

  23. Edginton AN, Schmitt W, Willmann S. Development and evaluation of a generic physiologically based pharmacokinetic model in children. Clin Pharmacokinet. 2006;45:1013–34.

    Article  PubMed  CAS  Google Scholar 

  24. Yang F, Tong X, McCarver DG, et al. Population based analysis of methadone distribution and metabolism using an age-dependent physiologically based pharmacokinetic model. J Pharmacokinet Pharmacodyn. 2006;33:485–518.

    Article  PubMed  CAS  Google Scholar 

  25. Alemzadeh R, Hoffmann RG, Dasgupta M, et al. Development of optimal kids insulin dosing system formulas for young children with type 1 diabetes mellitus. Diabetes Technol Ther. 2012;14:418–22.

    Article  PubMed  CAS  Google Scholar 

  26. Bouillon-Pichault M, Jullien V, Bazzoli C, et al. Pharmacokinetic design optimization in children and estimation of maturation parameters: example of cytochrome P450 3A4. J Pharmacokinet Pharmacodyn. 2011;38:25–40.

    Article  PubMed  CAS  Google Scholar 

  27. Ogungbenro K, Matthews I, Looby M, et al. Population pharmacokinetics and optimal design of paediatric studies for famciclovir. Br J Clin Pharmacol. 2009;68:546–60.

    Article  PubMed  CAS  Google Scholar 

  28. Perdaems N, Blasco H, Vinson C, et al. Predictions of metabolic drug-drug interactions using physiologically based modelling. Clin Pharmacokinet. 2010;49:239–58.

    Article  PubMed  CAS  Google Scholar 

  29. Chenel M, Bouzom F, Aarons L, Ogungbenro K. Drug-drug interaction predictions with PBPK models and optimal multiresponse sampling time designs: application to midazolam and a phase I compound. Part 1: comparison of uniresponse and multiresponse designs using PopDes. J Pharmacokinet Pharmacodyn. 2008;35:635–59.

    Article  PubMed  CAS  Google Scholar 

  30. Ahn JE, Karlsson MO, Dunne A, et al. Likelihood based approaches to handling data below the quantification limit using NONMEM VI. J Pharmacokinet Pharmacodyn. 2008;35:401–21.

    Article  PubMed  Google Scholar 

  31. Samson A, Lavielle M, Mentré F. Extension of the SAEM algorithm to left-censored data in nonlinear mixed-effects model: application to HIV dynamics model. Comput Stat Data Anal. 2006;51:1562–74.

    Article  Google Scholar 

  32. Rochon J. Application of GEE procedures for sample size calculations in repeated measures experiments. Stat Med. 1998;17:1643–58.

    Article  PubMed  CAS  Google Scholar 

  33. Ogungbenro K, Aarons L, Graham G. Sample size calculations based on generalized estimating equations for population pharmacokinetic experiments. J Biopharm Stat. 2006;16:135–50.

    Article  PubMed  Google Scholar 

  34. Rodgers T, Leahy D, Rowland M. Physiologically based pharmacokinetic modelling 1: predicting the tissue distribution of moderate-to-strong bases. J Pharm Sci. 2005;94:1259–76.

    Article  PubMed  CAS  Google Scholar 

  35. Johnson TN, Rostami-Hodjegan A. Resurgence in the use of physiologically based pharmacokinetic models in pediatric clinical pharmacology: parallel shift in incorporating the knowledge of biological elements and increased applicability to drug development and clinical practice. Paediatr Anaesth. 2011;21:291–301.

    Article  PubMed  Google Scholar 

  36. Beal SL, Sheiner LB. NONMEM users guide. San Francisco: University of California; 1992.

    Google Scholar 

  37. Wang Y. Derivation of various NONMEM estimation methods. J Pharmacokinet Pharmacodyn. 2007;34:575–93.

    Article  PubMed  Google Scholar 

  38. Bertrand J, Comets E, Mentré F. Comparison of model-based tests and selection strategies to detect genetic polymorphisms influencing pharmacokinetic parameters. J Biopharm Stat. 2008;18:1084–102.

    Article  PubMed  Google Scholar 

  39. Brendel K, Comets E, Laffont C, et al. Metrics for external model evaluation with an application to the population pharmacokinetics of gliclazide. Pharm Res. 2006;23:2036–49.

    Article  PubMed  CAS  Google Scholar 

  40. Retout S, Comets E, Samson A, et al. Design in nonlinear mixed effects models: Optimization using the Federov-Wynn algorithm and power of the Wald test for binary covariates. Stat Med. 2007;26:5162–79.

    Article  PubMed  Google Scholar 

  41. Dumont C, Chenel M, Mentré F. Design evaluation in nonlinear mixed effect models: influence of covariance between random effects [abstract no. 2158]. Population Approach Group in Europe (PAGE); 7–10 Jun 2011; Athens. http://www.page-meeting.org/?abstract=2158. Accessed 30 Oct 2012.

  42. Dumont C, Chenel M, Mentré F. Influence of covariance between random effects in design for nonlinear mixed effect models with an illustration in paediatric pharmacokinetics. J Biopharm Stat (in press).

  43. Dubois A, Bertrand J, Mentré F. Mathematical expressions of the pharmacokinetic and pharmacodynamic models implemented in the PFIM software. 2012. http://www.pfim.biostat.fr/PFIM_PKPD_library.pdf. Accessed 5 Nov 2012.

  44. Sölkner J. Choice of optimality criteria for the design of crossbreeding experiments. J Anim Sci. 1993;71:2867–73.

    PubMed  Google Scholar 

  45. Vila JP, Gauchi JP. Optimal designs based on exact confidence regions for parameter estimation of a nonlinear regression model. J Stat Plan Inference. 2007;137:2935–53.

    Article  Google Scholar 

  46. Mentré F, Nyberg J, Ogungbenro K, et al. Comparison of results of the different software for design evaluation in population pharmacokinetics and pharmacodynamics [abstract no. 2066]. Population Approach Group in Europe (PAGE); 7–10 June 2011; Athens. http://www.page-meeting.org/?abstract=2066. Accessed 30 Oct 2012.

  47. Dokoumetzidis A. Lumping of compartments [abstract no. 2637]. Population Approach Group in Europe (PAGE); 5–8 June 2012; Venice. http://www.page-meeting.org/?abstract=2637. Accessed 30 Oct 2012.

  48. Foo LK, Duffull S. Adaptive optimal design for bridging studies with an application to population pharmacokinetic studies. Pharm Res. 2012;29:1530–43.

    Article  PubMed  CAS  Google Scholar 

  49. Strougo A, Eissing T, Yassen A, et al. First dose in children: physiological insights into pharmacokinetic scaling approaches and their implications in paediatric drug development. J Pharmacokinet Pharmacodyn. 2012;39:195–203.

    Article  PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Cyrielle Dumont.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Dumont, C., Mentré, F., Gaynor, C. et al. Optimal Sampling Times for a Drug and its Metabolite using SIMCYP® Simulations as Prior Information. Clin Pharmacokinet 52, 43–57 (2013). https://doi.org/10.1007/s40262-012-0022-9

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40262-012-0022-9

Keywords

Navigation