Skip to main content

Pharmacokinetic and Pharmacodynamic Modeling

  • Living reference work entry
  • First Online:
Principles and Practice of Clinical Trials

Abstract

Pharmacokinetic and pharmacodynamic (PKPD) concepts and approaches play a critical role by increasing the efficiency throughout the drug development process and optimizing clinical therapeutics. Innovative quantitative analysis that integrate PKPD and disease progression knowledge are pivotal in regulatory decision-making and have shown to provide supportive evidence of effectiveness. Objective approaches to dose-selection that are informed by exposure-response analyses can assist in visualizing benefit-risk profiles on an individual and population level. Clinical trial simulations using quantitative models that incorporate structural and stochastic components along with prior knowledge can allow for informing clinical trial design features, dose selection in unstudied patient populations, and regulatory policy development. This chapter presents various PKPD-based analyses throughout the drug development process and within precision medicine. Emphasis on the use of PKPD analysis for proof of concept, dose-ranging, and first in human dose studies are subsequently provided. Case studies are highlighted to better illustrate the impact of quantitative analyses for decision-making.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Institutional subscriptions

References

  • Bhattaram V, Booth BP, Ramachandani RP, Beasley N, Wang Y, Tandon V, Duan JZ, Baweja RK, Marroum PJ, Uppoor RS, Rahman NA, Sahajwalla CG, Powell JR, Mehta MU, Gobburu J (2005) Impact of pharmacometrics on drug approval and labeling decisions: a survey of 42 drug applications. AAPS 7:e503–e512

    Article  Google Scholar 

  • Bhattaram VA, Siddiqui O, Kapcala LP, Gobburu JV (2009) Endpoints and analyses to discern disease-modifying drug effects in early Parkinson’s disease. AAPS 11:456–464

    Article  Google Scholar 

  • Bittl JA, Yulei H (2017) Bayesian analysis: a practical approach to interpret clinical trials and create clinical practice guidelines. Circ Cardiovasc Qual Outcomes 10:e003563

    Article  Google Scholar 

  • Bonate P (2011) Pharmacokinetic-Pharmacodynamic modeling and simulation. Springer Science & Business Media, Boston

    Book  Google Scholar 

  • Dayneka NL, Garg V, Jusko WJ (1993) Comparison of four basic models of indirect phar-macodynamic responses. J Pharmacokinet Biopharm 21:457–478

    Article  Google Scholar 

  • Derrochers J, Wojciechowski J, Klein-Schwartz W, Gobburu JV, Gopalakrishnan M (2017) Bayesian forecasting tool to predict the need for antidote in acute acetaminophen overdose. Pharmacotherapy 37:916–926

    Article  Google Scholar 

  • Ernst E, Resch K (1995) Concept of true and perceived placebo effects. BMJ 311:551–553

    Article  Google Scholar 

  • Fact Sheet: President Obama’s Precision Medicine Initiative (2015) Whitehouse.gov. Retrieved 25 Dec 2018

  • Fava M, Evins AE, Dorer DJ (2003) The problem of the placebo response in clinical trials for psychiatric disorders: culprits, possible remedies, and a novel study design approach. Pscyhother Psychosom 72:115–127

    Article  Google Scholar 

  • French JA, Kugler AR, Robing JL, Knapp LE, Garofalo EA (2003) Dose-response trial of pregabalin adjunctive therapy in patients with partial seizures. Neurology 60:1631–1637

    Article  Google Scholar 

  • Gobburu JV, Lesko L (2009) Quantitative disease drug trial models. Ann Rev Pharmacol Toxicol 49:291–301

    Article  Google Scholar 

  • Gobburu JV, Marroum PJ (2001) Utilization of pharmacokinetic-pharmacodynamic modeling and simulations in regulatory decision making. Clin Pharmacokinet 40:883–892

    Article  Google Scholar 

  • Gomeni R, Merlo-Pich E (2007) Bayesian modeling and ROC analysis to predict placebo responders using clinical score measured in the initial weeks of treatment in depression trials. Br J Clin Pharmacol 63:595–613

    Article  Google Scholar 

  • Hahn D, Emoto C, Euteneuer JC, Mizuno T, Vinks AA, Fukuda T (2018) Influence of OCT 1 ontogeny and genetic variation on morphine disposition in critically ill neonates: lessons from PBPK modeling and clinical study. Clin Pharmacol Ther 122:49–53

    Google Scholar 

  • Holford NH, Peace KE (1992) Methodologic aspects of a population pharmacodynamic model for cognitive effects in Alzheimer patients treated with tacrine. Proc Natl Acad Sci 89:11466–11470

    Article  Google Scholar 

  • Holford NH, Chan PL, Nutt JG, Kieburtz K, Shoulson I, Parkinson Study Group (2006) Disease progression and pharmacodynamics in Parkinson disease: evidence for function protection with levodopa and other treatments. J Pharmacokinet Pharmacodyn 22:281–311

    Article  Google Scholar 

  • International Conference on Harmonization (1994) Guidance on E4 dose response information to support drug registration; availability. Notice. Fed Regist 59:55972–55976

    Google Scholar 

  • Jadhav PR, Agersø H, Tornøe CW, Gobburu JV (2006) Semi-mechanistic pharmacodynamic modeling for degarelix, a novel gonadotropin releasing hormone (GnRH) blocker. J Pharmacokinet Pharmacodyn 33(5):609–634

    Article  Google Scholar 

  • Jusko WJ (1971) Pharmacodynamics of chemotherapeutic effects: dose-time-response relationships for phase-nonspecific agents. J Pharm Sci 60:892–895

    Article  Google Scholar 

  • Krause A et al (2016) Pharmacokinetic/pharmacodynamic modelling of the antimalarial effect of Actelion-451840 in an induced blood stage malaria study in healthy subjects. Br J Clin Pharmacol 82(2):412–421

    Article  Google Scholar 

  • Krudys KM, Dodds MG, Nissen SM, Vicini P (2005) Integrated model of hepatic and peripheral glucose regulation for estimation of endogenous glucose production during the hot IVGTT. Am J Physiol Endocrinol Metab 288:1038–1046

    Article  Google Scholar 

  • Lowe PJ, Hijazi Y, Luttringer O, Yin H, Sarangapani R, Howard D (2007) On the anticipation of the human dose in first-in-man trials from preclinical and prior clinical information in early drug development. Xenobiotica 37:1331–1354

    Article  Google Scholar 

  • Meno-Tetang GM, Lowe PJ (2005) On the prediction of the human response: a recycled mechanistic pharmacokinetic/pharmacodynamic approach. Basic Clin Pharmacol Toxicol 96:182–192

    Article  Google Scholar 

  • Mould DR, Upton RN, Wojciechowski J (2014) Dashboard systems. Implementing pharmacometrics from the bench to the bedside. AAPS 16:925–937

    Article  Google Scholar 

  • Peck CC, Rubin DB, Sheiner LB (2003) Hypothesis: a single clinical trial plus casual evidence of effectiveness is sufficient for drug approval. Clin Pharmacol Ther 72:481–490

    Article  Google Scholar 

  • Pillai G, Mentré F, Steimer JL (2005) Non-linear mixed effects modeling – from methodology and software development to driving implementation in drug development science. J Pharmacokinet Pharmacodyn 32:161–183

    Article  Google Scholar 

  • Powell JR, Gobburu JV (2007) Pharmacometrics at FDA: evolution and impact on decisions. Clin Pharmacol Ther 82:97–102

    Article  Google Scholar 

  • Senn S (2003) Bayesian, likelihood, and frequentist approaches to statistics. Applied Clinical Trials 12:35–38

    Google Scholar 

  • Sheiner LB (1997) Learning versus confirming in clinical drug development. Clin Pharmacol Ther 61:275–291

    Article  Google Scholar 

  • Sheiner LB, Rosenberg B, Melmon KL (1972) Modelling of individual pharmacokinetics for computer-aided drug dosage. Comput Biomed Res Int J 5:411–459

    Article  Google Scholar 

  • Sheiner LB, Stanski DR, Vozeh S, Miller RD, Ham J (1979) Simultaneous modeling of phar-macokinetics and pharmacodynamics: application to d-tubocurarine. Clin Pharmacol Ther 25:358–371

    Article  Google Scholar 

  • Standing J (2017) Understanding and applying pharmacometric modeling and simulation in clinical practice and research. Br J Clin Pharmacol 83:247–254

    Article  Google Scholar 

  • Sun YN, Jusko WJ (1998) Transit compartments versus gamma distribution function to model signal transduction processes in pharmacodynamics. J Pharm Sci 87:732–737

    Article  Google Scholar 

  • Sun W, Laughren TP, Zhu H, Hocchaus G, Wang Y (2013) Development of a placebo effect model combined with a dropout model for bipolar disorder. J Pharmacokinet Pharmacodyn 40:359–368

    Article  Google Scholar 

  • Tett SE, Holford NH, McLachlan AJ (1998) Population pharmacokinetics and pharmacodynamics: an underutilized resource. Drug Inf J 32:693–710

    Article  Google Scholar 

  • Thiel C, Smit I, Baier V, Cordes H, Fabry B, Blank LM, Kuepfer L (2018) Using quantitative systems pharmacology to evaluate the drug efficacy of COX-2 and 5-LOX inhibitors in therapeutic situations. NPJ Syst Biol Appl 4(1):28

    Article  Google Scholar 

  • US Food and Drug Administration (2001) Drug approval package: Xenazine (tetrabenazine) [FDA application no. (NDA) 021894; online]. Available from URL: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2008/021894s000TOC.cfm. Accessed 19 Dec 2018

  • US Food and Drug Administration (2002) Drug approval package: Zometa (zoledronic acid) [FDA application no. (NDA) 021223; online]. https://www.accessdata.fda.gov/drugsatfda_docs/nda/2002/21-386_Zometa.cfm. Accessed 19 Dec 2018

  • US Food and Drug Administration (2003) Guidance for Industry: Exposure-response relationships-study design, data analysis and regulatory application. https://www.fda.gov/downloads/drugs/guidancecomplianceregulatoryinformation/guidances/ucm072109.pdf. Accessed 19 Dec 2018

  • US Food and Drug Administration (2005) Guidance for industry: estimating the maximum safe starting dose in initial clinical trials for therapeutics in adult healthy volunteers. https://www.fda.gov/downloads/Drugs/Guidances/UCM078932.pdf. Accessed 24 Dec 2018

  • Visser SAG, De Alwis DP, Kerbusch T, Stone JA, Allerheiligen SRB (2014) Implementation of quantitative and systems pharmacology in large pharma. CPT Pharmacometrics Syst Pharmacol 3(10):1–10

    Article  Google Scholar 

  • Vugmeyster Y, Rohde C, Perreault M, Gimeno RE, Singh P (2013) Agonistic TAM-163 antibody targeting tyrosine kinase receptor-B: applying mechanistic modeling to enable preclinical to clinical translation and guide clinical trial design. mabs 5(3):373–383. Taylor & Francis

    Article  Google Scholar 

  • Wagner JG (1968) Kinetics of pharmacologic response. I. Proposed relationships between response and drug concentration in the intact animal and man. J Theor Biol 20:173–201

    Article  Google Scholar 

  • Weimer K, Colloca L, Enck P (2015) Placebo effects in psychiatry: mediators and moderators. Lancet Psychiatry 2:246–257

    Article  Google Scholar 

  • Yang J, Zhao H, Garnett C, Rahman A, Gobburu JV, Pierce W, Schechter G, Summers J, Keegan P, Booth B, Wang Y (2013) The combination of exposure-response and case-control analyses in regulatory decision making. J Clin Pharmacol 53:160–166

    Article  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jogarao V. Gobburu .

Editor information

Editors and Affiliations

Section Editor information

Rights and permissions

Reprints and permissions

Copyright information

© 2020 Springer Nature Switzerland AG

About this entry

Check for updates. Verify currency and authenticity via CrossMark

Cite this entry

Kalaria, S.N., Wang, H., Gobburu, J.V. (2020). Pharmacokinetic and Pharmacodynamic Modeling. In: Piantadosi, S., Meinert, C. (eds) Principles and Practice of Clinical Trials. Springer, Cham. https://doi.org/10.1007/978-3-319-52677-5_284-1

Download citation

  • DOI: https://doi.org/10.1007/978-3-319-52677-5_284-1

  • Received:

  • Accepted:

  • Published:

  • Publisher Name: Springer, Cham

  • Print ISBN: 978-3-319-52677-5

  • Online ISBN: 978-3-319-52677-5

  • eBook Packages: Springer Reference MathematicsReference Module Computer Science and Engineering

Publish with us

Policies and ethics