Skip to main content
Log in

Wide size dispersion and use of body composition and maturation improves the reliability of allometric exponent estimates

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

Abstract

To evaluate study designs and the influence of dispersion of body size, body composition and maturation of clearance or reliable estimation of allometric exponents. Non-linear mixed effects modeling and parametric bootstrap were employed to assess how the study sample size, number of observations per subject, between subject variability (BSV) and dispersion of size distribution affected estimation bias and uncertainty of allometric exponents. The role of covariate model misspecification was investigated using a large data set ranging from neonates to adults. A decrease in study sample size, number of observations per subject, an increase in BSV and a decrease in dispersion of size distribution, increased the uncertainty of allometric exponent estimates. Studies conducted only in adults with drugs exhibiting normal (30%) BSV in clearance may need to include at least 1000 subjects to be able to distinguish between allometric exponents of 2/3 and 1. Nevertheless, studies including both children and adults can distinguish these exponents with only 100 subjects. A marked bias of 45% (95%CI 41–49%) in the estimate of the allometric exponent of clearance was obtained when maturation and body composition were ignored in infants. A wide dispersion of body size (e.g. infants, children and adults) is required to reliably estimate allometric exponents. Ignoring differences in body composition and maturation of clearance may bias the exponent for clearance. Therefore, pharmacometricians should avoid estimating allometric exponent parameters without suitable designs and covariate models. Instead, they are encouraged to rely on the well-developed theory and evidence that clearance and volume parameters in humans scale with theory-based exponents.

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

Similar content being viewed by others

Data availability

Covariate data were obtained from a previously published study of busulfan with permission of the first author [34].

Code availability

Available as supplementary material.

References

  1. West GB, Brown JH (2005) The origin of allometric scaling laws in biology from genomes to ecosystems: towards a quantitative unifying theory of biological structure and organization. J Exp Biol 208:1575–1592

    PubMed  Google Scholar 

  2. Anderson BJ, Holford NH (2009) Mechanistic basis of using body size and maturation to predict clearance in humans. Drug Metab Pharmacokinet 24:25–36

    CAS  PubMed  Google Scholar 

  3. Ings RM (1990) Interspecies scaling and comparisons in drug development and toxicokinetics. Xenobiotica 20:1201–1231

    CAS  PubMed  Google Scholar 

  4. Hu TM, Hayton WL (2001) Allometric scaling of xenobiotic clearance: uncertainty versus universality. AAPS PharmSci 3:E29

    CAS  PubMed  Google Scholar 

  5. Mahmood I (2010) Theoretical versus empirical allometry: facts behind theories and application to pharmacokinetics. J Pharm Sci 99:2927–2933

    CAS  PubMed  Google Scholar 

  6. Holford NHG, Anderson BJ (2017) Allometric size: the scientific theory and extension to normal fat mass. Eur J Pharm Sci 109(Supplement):S59–S64

    CAS  Google Scholar 

  7. Holford N (1996) A size standard for pharmacokinetics. Clin Pharmacokinet 30:329–332

    CAS  PubMed  Google Scholar 

  8. Holford N (2013) A pharmacokinetics standard for babies and adults. J Pharm Sci 102:2941–2952

    CAS  PubMed  Google Scholar 

  9. Anderson BJ, Holford NHG (2011) Tips and traps analyzing pediatric PK data. Pediatr Anesth 21:222–237

    Google Scholar 

  10. Goulooze SC, Völler S, Välitalo PAJ, Calvier EAM, Aarons L, Krekels EHJ, Knibbe CAJ (2019) The influence of normalization weight in population pharmacokinetic covariate models. Clin Pharmacokinet 58:131–138

    CAS  PubMed  Google Scholar 

  11. West GB, Brown JH, Enquist BJ (1999) The fourth dimension of life: fractal geometry and allometric scaling of organisms. Science 284:1677–1679

    CAS  PubMed  Google Scholar 

  12. West GB, Brown JH, Enquist BJ (1997) A general model for the origin of allometric scaling laws in biology. Science 276:122–126

    CAS  PubMed  Google Scholar 

  13. Peters HP (1983) Physiological correlates of size. In: Beck R, Birks HJB, Conner EF (eds) Chapter 4. The ecological implications of body size. Cambridge University Press, Cambridge, pp 48–53

    Google Scholar 

  14. Brody S, Proctor RC, Ashworth US (1934) Basal metabolism, endogenous nitrogen, creatinine, and sulphur excretions as functions of body weight. Univ Mo Agric Exp Stn Res Bull 220:1–40

    Google Scholar 

  15. Kleiber M (1932) Body size and metabolism. Hilgardia 6:315–333

    CAS  Google Scholar 

  16. Stahl WR (1967) Scaling of respiratory variables in mammals. J Appl Physiol 22:453–600

    CAS  PubMed  Google Scholar 

  17. Cortinez LI, Anderson BJ, Penna A, Olivares L, Muñoz HR, Holford NHG, Struys MMRF, Sepulveda P (2010) Influence of obesity on propofol pharmacokinetics: derivation of a pharmacokinetic model. BJA 105:448–456

    CAS  PubMed  Google Scholar 

  18. Anderson BJ, Holford NH (2017) What is the best size predictor for dose in the obese child? Paediatr Anaesth 27(12):1176–1184

    PubMed  Google Scholar 

  19. Anderson BJ, Holford NH (2008) Mechanism-based concepts of size and maturity in pharmacokinetics. Annu Rev Pharmacol Toxicol 48:303–332

    CAS  PubMed  Google Scholar 

  20. Huang Q, Riviera JE (2014) The application of allometric scaling principles to predict pharmacokinetic parameters across species. Expert Opin Drug Metab Toxicol 10:1241–1253

    CAS  PubMed  Google Scholar 

  21. Knibbe CAJ, Zuideveld KP, Arts LP, Kuks PF, Danhof M (2005) Allometric relationships between the pharmacokinetics of propofol in rats, children and adults. Br J Clin Pharmacol 59:705–711

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Dodds PS, Rothman DH, Weitz JS (2001) Re-examination of the “3/4-law” of metabolism. J Theor Biol 209:9–27

    CAS  PubMed  Google Scholar 

  23. Agutter PS, Wheatley DN (2004) Metabolic scaling: consensus or controversy? Theor Biol Med Model 1:13

    PubMed  PubMed Central  Google Scholar 

  24. Bokma F (2004) Evidence against universal metabolic allometry. Funct Ecol 18:184–187

    Google Scholar 

  25. Glazier DS (2005) Beyond the “3/4-power law”: variation in the intra- and interspecific scaling of metabolic rate in animals. Biol Rev 80:611–662

    PubMed  Google Scholar 

  26. White CR, Cassey P, Blackburn TM (2007) Allometric exponents do not support a universal metabolic allometry. Ecology 88:315–323

    PubMed  Google Scholar 

  27. Kolokotrones T, Van S, Deeds EJ, Fontana W (2010) Curvature in metabolic scaling. Nature 464:753–756

    CAS  PubMed  Google Scholar 

  28. Benedict FG (1938) Vital energetics: a study in comparative basal metabolism. Carnegie Institution, Washington

    Google Scholar 

  29. White CR, Seymour RS (2003) Mammalian basal metabolic rate is proportional to body mass 2/3. Proc Natl Acad Sci USA 100:4046–4049

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Germovsek E, Barker CIS, Sharland M, Standing JF (2017) Scaling clearance in paedriatic pharmacokinetics: all models are wrong, which are useful? Br J Clin Pharmacol 83:777–790

    CAS  PubMed  Google Scholar 

  31. Calvier EA, Krekels EH, Valitalo PA, Rostami-Hodjegan A, Tibboel D, Danhof M, Knibbe CA (2017) Allometric scaling of clearance in paediatric patients: when does the magic of 0.75 fade? Clin Pharmacokinet 56:273–85

    PubMed  Google Scholar 

  32. European Medicine Agency. Modelling and simulation: questions and answers. “Should fixed or estimated values be used in paediatric pharmacokinetic models?” November 2018. Available at: https://www.ema.europa.eu/en/human-regulatory/research-development/scientific-guidelines/clinical-pharmacology-pharmacokinetics/modelling-simulation-questions-answers. Accessed 30 Mar 2021

  33. Sinha J, Al-Sallami HS, Duffull SB (2019) Choosing the allometric exponent in covariate model building. Clin Pharmacokinet 58:89–100

    PubMed  Google Scholar 

  34. McCune JS, Bemer MJ, Barrett JS, Baker KS, Gamis AS, Holford NHG (2014) Busulfan in infant to adult hematopoietic cell transplant recipients: a population pharmacokinetic model for initial and Bayesian dose personalization. Clin Cancer Res 20:754–63

    CAS  PubMed  Google Scholar 

  35. Janmahasatian S, Duffull SB, Ash S, Ward LC, Byrne NM, Green B (2005) Quantification of lean bodyweight. Clin Pharmacokinet 44:1051–1065

    PubMed  Google Scholar 

  36. Al-Sallami HS, Cheah SL, Han SY, Liew J, Lim J, Ng MA et al (2014) Between-subject variability: should high be the new normal? Eur J Clin Pharmacol 70:1403–1404

    PubMed  Google Scholar 

  37. De Castro FA, Piana C, Simoes B, Lanchote VL, Della Pasqua O (2015) Busulfan dosing algorithm and sampling strategy in stem cell transplantation patients. Br J Clin Pharmacol 80:618–29

    PubMed  PubMed Central  Google Scholar 

  38. Chow SC, Zheng J (2019) The use of 95% CI or 90% CI for drug product development—a controversial issue? J Biopharm Stat 29:834–844

    PubMed  Google Scholar 

  39. The R Project for Statistical Computing. Available at: https://www.r-project.org/. Accessed 30 Mar 2021

  40. Mahmood I (2012) Prediction of clearance and volume of distribution in the obese from normal weight subjects. Clin Pharmacokinet 51:527–542

    CAS  PubMed  Google Scholar 

  41. Gonzalez D, Laughon MM, Smith PB, Ge S, Ambalavanan N, Atz A et al (2019) Population pharmacokinetics of sildenafil in extremely premature infants. Br J Clin Pharmacol. https://doi.org/10.1111/bcp.14111

    Article  PubMed  PubMed Central  Google Scholar 

  42. Freshwater T, Kondic A, Ahamadi M, Li CH, de Greef R, de Alwis D, Stone JA (2017) Evaluation of dosing strategy for pembrolizumab for oncology indications. J ImmunoTher Cancer 5:43

    PubMed  PubMed Central  Google Scholar 

  43. Boxenbaum H (1982) Interspecies scaling, allometry, physiological time, and the ground plan of pharmacokinetics. J Pharmacokinet Biopharm 10:201–227

    CAS  PubMed  Google Scholar 

  44. Mordenti J (1986) Man versus beast: pharmacokinetic scaling in mammals. J Pharm Sci 75:1028–1040

    CAS  PubMed  Google Scholar 

  45. White CR, Seymour RS (2005) Allometric scaling of mammalian metabolism. J Exp Biol 208:1611–1619

    CAS  PubMed  Google Scholar 

  46. Rowland M, Tozer TN (2011) Clinical pharmacokinetics and pharmacodynamics: concepts and applications. Lippincott Williams & Wilkins, Philadelphia

    Google Scholar 

  47. West GB (2012) The importance of quantitative systemic thinking in medicine. Lancet 379:1551–1559

    PubMed  Google Scholar 

  48. Mahmood I (2014) Dosing in children: a critical review of the pharmacokinetic allometric scaling and modelling approaches in paediatric drug development and clinical settings. Clin Pharmacokinet 53:327–346

    CAS  PubMed  Google Scholar 

  49. Bonate PL (1999) The effect of collinearity on parameter estimates in nonlinear mixed effects models. Pharm Res 16:709–717

    CAS  PubMed  Google Scholar 

  50. Holford NH, Ma SC, Anderson BJ (2012) Prediction of morphine dose in humans. Paediatr Anaesth 22:209–222

    PubMed  Google Scholar 

  51. Sumpter A, Holford NHG (2012) A model for fat free mass in humans from very premature neonates to young adults. PAGANZ https://www.paganz.org/abstracts/a-model-for-fat-free-mass-in-humans-from-very-premature-neonates-to-young-adults/. Accessed 31 Jan 2019

  52. Al-Sallami HS, Goulding A, Grant A, Taylor R, Holford N, Duffull SB (2015) Prediction of fat-free mass in children. Clin Pharmacokinet 54:1169–1178

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We gratefully acknowledge Dr Jeannine McCune for allowing us to use the covariate data from her study of the pharmacokinetics of busulfan [34].

Funding

This work received no specific funding.

Author information

Authors and Affiliations

Authors

Contributions

MGS and NH designed the study, MGS performed the calculations, MGS, NH, GB and JD wrote and take responsibility for the manuscript.

Corresponding author

Correspondence to Mario González-Sales.

Ethics declarations

Conflict of interest

Mario González-Sales, Nick Holford, Guillaume Bonnefois and Julie Desrochers declare no conflicts of interest.

Ethical approval

No ethical approval was required for this simulation-based work.

Informed consent

All authors consent for publication.

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 1140 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

González-Sales, M., Holford, N., Bonnefois, G. et al. Wide size dispersion and use of body composition and maturation improves the reliability of allometric exponent estimates. J Pharmacokinet Pharmacodyn 49, 151–165 (2022). https://doi.org/10.1007/s10928-021-09788-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10928-021-09788-3

Keywords

Navigation