Skip to main content
Log in

Impact of covariate model building methods on their clinical relevance evaluation in population pharmacokinetic analyses: comparison of the full model, stepwise covariate model (SCM) and SCM+ approaches

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

Abstract

Covariate analysis in population pharmacokinetics is key for adjusting doses for patients. The main objective of this work was to compare the adequacy of various modeling approaches on covariate clinical relevance decision-making. The full model, stepwise covariate model (SCM) and SCM+ PsN algorithms were compared in a clinical trial simulation of a 383-patient population pharmacokinetic study mixing rich and sparse designs. A one-compartment model with first-order absorption was used. A base model including a body weight effect on CL/F and V/F and a covariate model including 4 additional covariates-parameters relationships were simulated. As for forest plots, ratios between covariates at a specific value and that of a typical individual were calculated with their 90% confidence interval (CI90) using standard errors. Covariates on CL, V and KA were considered relevant if their CI90 fell completely outside the reference area [0.8–1.2]. All approaches provided unbiased covariate ratio estimates. For covariates with a simulated effect, the 3 approaches correctly identify their clinical relevance. However, significant covariates were missed in up to 15% of cases with SCM/SCM+. For covariate with no simulated effects, the full model mainly identified them as non-relevant or with insufficient information while SCM/SCM+ mainly did not select them. SCM/SCM+ assume that non-selected covariates are non-relevant when it could be due to insufficient information, whereas the full model does not make this assumption and is faster. This study must be extended to other methods and completed by a more complex high-dimensional simulation framework.

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. European Medicines Agency (2022) ICH guideline E11A on pediatric extrapolation - Scientific guideline. https://www.ema.europa.eu/en/ich-guideline-e11a-pediatric-extrapolation-scientific-guideline. Accessed 4 Oct 2023

  2. Dartois C, Brendel K, Comets E et al (2007) Overview of model-building strategies in population PK/PD analyses: 2002–2004 literature survey. Br J Clin Pharmacol 64:603–612. https://doi.org/10.1111/j.1365-2125.2007.02975.x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Hutmacher MM, Kowalski KG (2015) Covariate selection in pharmacometric analyses: a review of methods. Br J Clin Pharmacol 79:132–147. https://doi.org/10.1111/bcp.12451

    Article  PubMed  Google Scholar 

  4. Ahamadi M, Largajolli A, Diderichsen PM et al (2019) Operating characteristics of stepwise covariate selection in pharmacometric modeling. J Pharmacokinet Pharmacodyn 46:273–285. https://doi.org/10.1007/s10928-019-09635-6

    Article  PubMed  Google Scholar 

  5. Jonsson EN, Karlsson MO (1998) Automated covariate model building within NONMEM. Pharm Res 15:1463–1468. https://doi.org/10.1023/a:1011970125687

    Article  CAS  PubMed  Google Scholar 

  6. Lindbom L, Pihlgren P, Jonsson N (2005) PsN-Toolkit—a collection of computer intensive statistical methods for non-linear mixed effect modeling using NONMEM. Comput Methods Programs Biomed 79:241–257. https://doi.org/10.1016/j.cmpb.2005.04.005

    Article  PubMed  Google Scholar 

  7. Svensson RJ, Jonsson EN (2022) Efficient and relevant stepwise covariate model building for pharmacometrics. CPT Pharmacomet Syst Pharmacol 11:1210–1222. https://doi.org/10.1002/psp4.12838

    Article  CAS  Google Scholar 

  8. Lindbom L, Ribbing J, Jonsson EN (2004) Perl-speaks-NONMEM (PsN)—a Perl module for NONMEM related programming. Comput Methods Programs Biomed 75:85–94. https://doi.org/10.1016/j.cmpb.2003.11.003

    Article  PubMed  Google Scholar 

  9. Ayral G, Si Abdallah J-F, Magnard C, Chauvin J (2021) A novel method based on unbiased correlations tests for covariate selection in nonlinear mixed effects models: the COSSAC approach. CPT Pharmacomet Syst Pharmacol 10:318–329. https://doi.org/10.1002/psp4.12612

    Article  CAS  Google Scholar 

  10. Mandema JW, Verotta D, Sheiner LB (1992) Building population pharmacokineticpharmacodynamic models. I. Models for covariate effects. J Pharmacokinet Biopharm 20:511–528. https://doi.org/10.1007/BF01061469

    Article  CAS  PubMed  Google Scholar 

  11. Ribbing J, Nyberg J, Caster O, Jonsson EN (2007) The lasso—a novel method for predictive covariate model building in nonlinear mixed effects models. J Pharmacokinet Pharmacodyn 34:485–517. https://doi.org/10.1007/s10928-007-9057-1

    Article  PubMed  Google Scholar 

  12. Prague M, Lavielle M (2022) SAMBA: A novel method for fast automatic model building in nonlinear mixed-effects models. CPT Pharmacomet Syst Pharmacol 11:161–172. https://doi.org/10.1002/psp4.12742

    Article  CAS  Google Scholar 

  13. Terranova N, Venkatakrishnan K, Benincosa LJ (2021) Application of machine learning in translational medicine: current status and future opportunities. AAPS J 23:74. https://doi.org/10.1208/s12248-021-00593-x

    Article  PubMed  Google Scholar 

  14. Sibieude E, Khandelwal A, Hesthaven JS et al (2021) Fast screening of covariates in population models empowered by machine learning. J Pharmacokinet Pharmacodyn 48:597–609. https://doi.org/10.1007/s10928-021-09757-w

    Article  PubMed  PubMed Central  Google Scholar 

  15. Ronchi D, Tosca EM, Bartolucci R, Magni P (2023) Go beyond the limits of genetic algorithm in daily covariate selection practice. J Pharmacokinet Pharmacodyn. https://doi.org/10.1007/s10928-023-09875-7

    Article  PubMed  PubMed Central  Google Scholar 

  16. Xu XS, Yuan M, Zhu H et al (2018) Full covariate modelling approach in population pharmacokinetics: understanding the underlying hypothesis tests and implications of multiplicity. Br J Clin Pharmacol 84:1525–1534. https://doi.org/10.1111/bcp.13577

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Gastonguay MR (2011) Full covariate models as an alternative to methods relying on statistical significance for inferences about covariate effects: a review of methodology and 42 case studies. Twentieth Meeting, Population Approach Group in Europe, Athens, Grece. https://www.page-meeting.org/pdf_assets/1694-GastonguayPAGE2011.pdf. Accessed 4 Oct 2023

  18. Yngman G, Nordgren R, Freiberga S, Karlsson MO Linearization of full random effects modeling (FREM) for time-efficient automatic covariate assessment

  19. U.S. Food & Drug Administration (2022) Population pharmacokinetics, guidance for industry. https://www.fda.gov/regulatory-information/search-fda-guidance-documents/population-pharmacokinetics. Accessed 4 Oct 2023

  20. Yngman G, Bjugård Nyberg H, Nyberg J et al (2022) An introduction to the full random effects model. CPT Pharmacomet Syst Pharmacol 11:149–160. https://doi.org/10.1002/psp4.12741

    Article  CAS  Google Scholar 

  21. Amann LF, Wicha SG (2023) Operational characteristics of full random effects modelling (‘frem’) compared to stepwise covariate modelling (‘scm’). J Pharmacokinet Pharmacodyn. https://doi.org/10.1007/s10928-023-09856-w

    Article  PubMed  PubMed Central  Google Scholar 

  22. European Medicines Agency (2018) Investigation of subgroups in confirmatory clinical trials - Scientific guideline. https://www.ema.europa.eu/en/investigation-subgroups-confirmatory-clinical-trials-scientific-guideline. Accessed 26 Oct 2023

  23. Menon-Andersen D, Yu B, Madabushi R et al (2011) Essential pharmacokinetic information for drug dosage decisions: a concise visual presentation in the drug label. Clin Pharmacol Ther 90:471–474. https://doi.org/10.1038/clpt.2011.149

    Article  CAS  PubMed  Google Scholar 

  24. Marier J-F, Teuscher N, Mouksassi M-S (2022) Evaluation of covariate effects using forest plots and introduction to the coveffectsplot R package. CPT Pharmacomet Syst Pharmacol 11:1283–1293. https://doi.org/10.1002/psp4.12829

    Article  CAS  Google Scholar 

  25. European Medicines Agency (2018) Investigation of bioequivalence - Scientific guideline. https://www.ema.europa.eu/en/investigation-bioequivalence-scientific-guideline. Accessed 4 Oct 2023

  26. Thai H-T, Mentré F, Holford NHG et al (2014) Evaluation of bootstrap methods for estimating uncertainty of parameters in nonlinear mixed-effects models: a simulation study in population pharmacokinetics. J Pharmacokinet Pharmacodyn 41:15–33. https://doi.org/10.1007/s10928-013-9343-z

    Article  PubMed  Google Scholar 

  27. Dosne A-G, Bergstrand M, Harling K, Karlsson MO (2016) Improving the estimation of parameter uncertainty distributions in nonlinear mixed effects models using sampling importance resampling. J Pharmacokinet Pharmacodyn 43:583–596. https://doi.org/10.1007/s10928-016-9487-8

    Article  PubMed  PubMed Central  Google Scholar 

  28. Cuzick J (2005) Forest plots and the interpretation of subgroups. Lancet 365:1308. https://doi.org/10.1016/S0140-6736(05)61026-4

    Article  PubMed  Google Scholar 

  29. Hahn AW, Dizman N, Msaouel P (2022) Missing the trees for the forest: most subgroup analyses using forest plots at the ASCO annual meeting are inconclusive. Ther Adv Med Oncol 14:17588359221103200. https://doi.org/10.1177/17588359221103199

    Article  Google Scholar 

  30. Retout S, Schmitt C, Petry C et al (2020) Population pharmacokinetic analysis and exploratory exposure-bleeding rate relationship of emicizumab in adult and pediatric persons with hemophilia A. Clin Pharmacokinet 59:1611–1625. https://doi.org/10.1007/s40262-020-00904-z

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Oldenburg J, Mahlangu JN, Kim B et al (2017) Emicizumab prophylaxis in hemophilia A with inhibitors. N Engl J Med 377:809–818. https://doi.org/10.1056/NEJMoa1703068

    Article  CAS  PubMed  Google Scholar 

  32. Mahlangu J, Oldenburg J, Paz-Priel I et al (2018) Emicizumab prophylaxis in patients who have hemophilia A without inhibitors. N Engl J Med 379:811–822. https://doi.org/10.1056/NEJMoa1803550

    Article  CAS  PubMed  Google Scholar 

  33. Yoneyama K, Schmitt C, Portron A et al (2023) Clinical pharmacology of emicizumab for the treatment of hemophilia A. Expert Rev Clin Pharmacol 16:775–790. https://doi.org/10.1080/17512433.2023.2243213

    Article  CAS  PubMed  Google Scholar 

  34. Gallant AR (1975) Seemingly unrelated nonlinear regressions. J Econ 3:35–50. https://doi.org/10.1016/0304-4076(75)90064-0

    Article  Google Scholar 

  35. National Center for Advancing Translational Sciences Toolkit. Clinical relevance - Glossary. https://toolkit.ncats.nih.gov/glossary/clinical-relevance. Accessed 17 Nov 2023

Download references

Acknowledgements

This work was financed by a CIFRE agreement (Conventions Industrielles de Formation par la Recherche) and was conducted under the supervision of the ANRT (Association Nationale de la Recherche et de la Technologie). The CIFRE agreement is a partnership between a public laboratory and a company, here the UMR (Unité Mixte de Recherche) 1137 and INSTITUT ROCHE, respectively. The authors are grateful to Kamill Jaworski for his technical support in the simulation implementation.

Author information

Authors and Affiliations

Authors

Contributions

M.P., S.M., S.R. and F.M. designed the simulation study. M.P. implemented and performed the simulations. M.P. produced the results. M.P., S.M., S.R. and F.M. analyzed the results. M.P. wrote the manuscript. S.M., S.R. and F.M. reviewed the manuscript.

Corresponding author

Correspondence to Morgane Philipp.

Ethics declarations

Competing interests

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

Below is the link to the electronic supplementary material.

Supplementary file1 (DOCX 2270 KB)

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) 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

Philipp, M., Buatois, S., Retout, S. et al. Impact of covariate model building methods on their clinical relevance evaluation in population pharmacokinetic analyses: comparison of the full model, stepwise covariate model (SCM) and SCM+ approaches. J Pharmacokinet Pharmacodyn (2024). https://doi.org/10.1007/s10928-024-09911-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s10928-024-09911-0

Keywords

Navigation