Skip to main content
Log in

Quantitative clinical pharmacology is transforming drug regulation

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

Abstract

Prior to 1970s, development and regulation of new drugs was devoid of a fully quantitative, pathophysiological conceptual foundation. Malcolm Rowland pioneered, in collaboration with colleagues and friends, our modern understanding of drug clearance concepts, and equipped drug development and regulatory scientists with key investigative tools such as physiologically-based pharmacokinetic (PBPK) modeling, standardized approaches to characterizing drug metabolism, and microdosing. From the 1970s to the present, Malcolm Rowland has contributed to key advances in pharmacokinetics that have had transformational impacts on drug regulatory science. These advances include concepts that have led to the fundamental understanding that mechanistically derived, quantitative variations in drug concentrations, rather than assigned dosage alone, drive pharmacodynamic effects (PKPD)—including disease biomarkers and clinical outcomes. This body of knowledge has transformed drug development and regulatory science theory and practice from naïve empiricism to a mechanism/model-based, quantitative scientific discipline. As a result, it is now possible to incorporate pre-clinical in vitro data on drug physico-chemical properties, metabolizing enzymes, transporters and permeability properties into PBPK-based simulations of expected PK distributions and drug–drug interactions in human populations. The most comprehensive application of PK-PD is in the modeling and simulation of clinical trials in the context of model-based drug development and regulation, imbedded in the “learn-confirm paradigm”. Regulatory agencies have embraced these advances and incorporated them into regulatory requirements, approval acceleration pathways and regulatory decisions. These developments are reviewed here, with emphasis on key contributions of Malcolm Rowland that facilitated this transformation.

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

Similar content being viewed by others

References

  1. Osterberg L, Blaschke T (2005) Adherence to medication. N Engl J Med 353:487–497

    Article  CAS  PubMed  Google Scholar 

  2. Urquhart J, Vrijens B (2005) New findings about patient adherence to prescribed drug dosing regimens: an introduction to pharmionics. Eur J Hosp Pharm Sci 11:103–106

    Google Scholar 

  3. Kenna LA, Sheiner LB (2004) Estimating treatment effect in the presence of non-compliance measured with error: precision and robustness of data analysis methods. Stat Med 23:3561–3580

    Article  PubMed  Google Scholar 

  4. Benet L, Rowland M (1982) Pharmacometrics: a new journal section. J Pharmaocokin Biopharm 10:349–350

    Article  Google Scholar 

  5. Bacharach AL, Laurence DR (1964) Evaluation of drug activities: pharmacometrics, vol 1. Academic Press, New York

    Google Scholar 

  6. Steimer JL, Sheiner LB (2000) Pharmacokinetic/pharmacodynamic modeling in drug development. Annu Rev Pharmacol Toxicol 40:67–95

    Article  PubMed  Google Scholar 

  7. Zhang L, Sinha V, Forgue ST, Callies S, Ni L, Peck R, Allerheiligan SRB (2006) Model-based drug development: the road to quantitative pharmacology. J Pharmacokinet Pharmacodyn 33:567–1567

    Article  Google Scholar 

  8. Lalonde RL, Kowalski KG, Hutmacher MM, Ewy W, Nichols DJ, Milligan PA, Corrigan BW, Lockwood PA, Marshall SA, Benincosa LJ, Tensfeldt TG, Parivar K, Amantea M, Glue P, Koide H, Miller R (2007) Model-based drug development. Clin Pharmacol Ther 82:21–32

    Article  CAS  PubMed  Google Scholar 

  9. Blackstone MO, Lindenbaum J, Buler VP (1971) Variations in biological availability of digoxin from four preparations. N Engl J Med 285:1344

    Article  PubMed  Google Scholar 

  10. 21 C.F.R. (1977) § 320 Bioavailability and bioequivalance requirements. Fed Regist 42:1648

    Google Scholar 

  11. Pang KS, Rowland M (1977) Hepatic clearance of drugs. I. Theoretical considerations of a “well-stirred” model and a “parallel tube” model. Influence of hepatic blood flow, plasma and blood cell binding, and the hepatocellular enzymatic activity on hepatic drug clearance. J Pharmacokin Biopharm 5:625–653

    Article  CAS  Google Scholar 

  12. Roberts MS, Rowland M (1977) A dispersion model of hepatic elimination: 1. Formulation of the model and bolus considerations. J Pharmacokin Biopharm 14

  13. Benet LZ, Rowland M, Graham GG (1973) Hepatic clearance of drugs. I. Theoretical considerations of a “well stirred” model and a “parallel tube” model. Influence of hepatic blood flow, plasma and blood cell binding, and the hepatocellular enzymatic activity on hepatic drug clearance. J Pharmacokinet Pharmacodyn 1:123–136

    Google Scholar 

  14. Beal SL, Sheiner LB, Marathe VV (1977) Estimation of population characteristics of pharmacokinetic parameters from routine clinical data. J Pharmackinet Pharmacodyn 5:445–479

    Google Scholar 

  15. Food and Drug Administration (1989) Guideline for the study of drugs likely to be used in the elderly

  16. Peck C, Collins J, Barr W, Benet L, Desjardins R, Furst D, Harter JG, Levy G, Ludden T, Rodman J, Sanathanan L, Schentag JJ, Shah VP, Sheiner LB, Skelly JP, Stanski DR, Temple RJ, Viswanathan CT, Weissinger J, Yacobi A (1992) Opportunities for integration of pharmacokinetics, pharmacodynamics, and toxicokinetics in rational drug development. Clin Pharmacol Ther 51:465–473

    Article  CAS  PubMed  Google Scholar 

  17. Sanathanan LP, Peck CC (1991) The randomized concentration-controlled trial: an evaluation of its sample size efficiency. Control Clin Trials 12:780–794

    Article  CAS  PubMed  Google Scholar 

  18. Peck CC (1992) Population approach in pharmacokinetics and pharmacodynamics: FDA view. In: Conference on new strategies in drug development and clinical evaluation: the population approach. Commission of the European Communities: European cooperation in the field of scientific and technical research, pp 157–168

  19. Food and Drug Administration (1999) Guidance for industry: population pharmacokinetics

  20. Hale MD, Gillespie WR, Gupta SK, Tuck B, Holford NH (1996) Clinical trial simulation: streamlining your drug development process. Appl Clin Trials 5:35–40

    Google Scholar 

  21. Monteleone JPR, Holford NHG, Kimko HC, Peck CC (2000) Simulation of clinical trials. Annu Rev Pharmacol Toxicol 40:209–234

    Article  PubMed  Google Scholar 

  22. Lesko L, Williams RL (1999) The question-based review—a conceptual framework for good review practices. Appl Clin Trials 56–62

  23. Food and Drug Administration (2003) Concept paper End-of-Phase-2A meetings with sponsors regarding exposure-response and IND and NDA products

  24. Food and Drug Administration (2008) Guidance for industry End-of-Phase 2A meetings

  25. Pharmacometrics at FDA (2009) http://www.fda.gov/AboutFDA/CentersOffices/CDER/ucm167032.htm

  26. 21 C.F.R. (1977) § 320 Part 25 guidelines for the conduct of an in vivo bioavailability study. Fed Regist 42:1648

    Google Scholar 

  27. Food and Drug Administration (2003) Guidance for industry clinical bioavailability and bioequivalence studies for orally administered drug products—general considerations. http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm070124.pdf

  28. Food and Drug Administration (1994) Guidance for industry: dose-response information to support drug registration

  29. Food and Drug Administration (2003) Guidance for industry exposure-response relationships—study design, data analysis, and regulatory applications

  30. Peck C, Temple R, Collins J (1973) Understanding consequences of concurrent therapies. JAMA 269:1550–1552

    Article  Google Scholar 

  31. Banfield C, O’Reilly RE, Chan E, Rowland M (1983) Phenylbutazone-warfarin interaction in man: further stereochemical and metabolic considerations. Br J Clin Pharmacol 16:669–675

    CAS  PubMed  Google Scholar 

  32. Rowland M, Matin SB (1973) Kinetics of drug–drug interactions. J Pharmacokin Biopharm 1:553–568

    Article  CAS  Google Scholar 

  33. Food and Drug Administration (1999) Guidance for industry in vivo drug metabolism/drug interaction studies—study design, data analysis, and recommendations for dosing and labeling

  34. Food and Drug Administration (2006) Guidance for industry drug interaction studies—study design, data analysis, and implications for dosing and labeling

  35. Food and Drug Administration (1997) Guidance for industry in vivo drug metabolism/drug interaction studies—study design, data analysis, and recommendations for dosing and labeling

  36. Jeffords AJM (1997) Clarification of the number of required clinical investigations for approval. Report 105-43 of U. S. Senate Committee on Labor and Human Resources on the Food and Drug Administration Modernization Act of 1997, pp 30–31

  37. Food and Drug Administration Modernization Act of 1997 (1997)

  38. Jeffords AJM (1997) Pediatric studies of drugs. Report 105-43 of U. S. Senate Committee on Labor and Human Resources on the Food and Drug Administration Modernization Act of 1997, pp 51–52

  39. Bliley TJ (1997) Report 105-310 of the U. S. House Committee on Commerce Report on the Prescription Drug User Fee Reauthorization and Drug Regulatory Modernization Act of 1997, pp 67–68

  40. Food and Drug Administration (1998) Guidance for industry providing evidence of effectiveness for human drug and biological products

  41. Peck CC, Wechsler J (2002) Report of a workshop on confirmatory evidence to support a single clinical trial for new drug approval. Drug Inf J 36:517–534

    Google Scholar 

  42. Benowitz N, Forsyth RP, Melmon KL, Rowland M (1974) Lidocaine disposition kinetics in monkey and manI. Prediction by a perfusion model. Clin Pharmacol Ther 16:87–98

    CAS  PubMed  Google Scholar 

  43. Blakey GM, Nestorov IA, Arundel PA, Aarons LJ, Rowland M (1997) Quantitative structure-pharmacokinetics relationships: I. Development of a whole-body physiologically based model to characterize changes in the pharmacokinetics across a homologous series of barbiturates in the rat. J Pharmacokin Biopharm 25:277–312

    Article  CAS  Google Scholar 

  44. Rodgers T, Rowland M (2007) Mechanistic approaches to volume of distribution predictions: understanding the processes. Pharm Res 24:918–933

    Article  CAS  PubMed  Google Scholar 

  45. Rodgers T, Leahy E, Rowland M (2005) Physiologically-based pharmacokinetic modeling 1: predicting the tissue distribution of moderate-to-strong bases. J Pharm Sci 94:1259–1276

    Article  CAS  PubMed  Google Scholar 

  46. Rostami-Hodjegan A, Tucker GT (2007) Simulation and prediction of in vivo drug metabolism in human populations from in vitro data. Nat Rev Drug Discov 6:140–148

    Article  CAS  PubMed  Google Scholar 

  47. Bischoff KB, Dedrick RL, Zaharko DS, Longstreth JA (1971) Methotrexate pharmacokinetics. J Pharm Sci 60:1128–1133

    Article  CAS  PubMed  Google Scholar 

  48. Clewell HJ, Andersen ME, Wills RJ, Latriano L (1997) A physiologically based pharmacokinetic model for retinoic acid and its metabolites. J Am Acad Dermatol 36:S77–S85

    Article  PubMed  Google Scholar 

  49. Ghosh TK (2000) Clinical pharmacology and biopharmaceutics review: NDA 21-108 0.02% Tretinoin Emollient Cream (RENOVA®). http://wwwaccessdatafdagov/drugsatfda_docs/nda/2000/21-108_Renova_BioPharmrpdf, p 19

  50. Balant L, Rowland M, Peck C (2004) Physiologically based pharmacokinetics in drug development and regulatory science: a workshop report. AAPS J 6:6–10

    Google Scholar 

  51. Oo C, Chen YC (2009) The need for multiple doses of 400 mg ketoconazole as a precipitant inhibitor of a CYP3A substrate in an in vivo drug–drug interaction study. J Clin Pharmacol 49:368–369

    Article  PubMed  Google Scholar 

  52. Zhao P, Ragueneau-Majlessi I, Zhang L, Strong JM, Reynolds KS, Levy RH, Thummel KE, Huang S-M (2009) Quantitative evaluation of pharmacokinetic inhibition of CYP3A substrates by ketoconazole: a simulation study. J Clin Pharmacol 49:351–359

    Article  CAS  PubMed  Google Scholar 

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

    Google Scholar 

  54. Huang W, Lee SL, Yu LX (2009) Mechanistic approaches to predicting oral drug absorption. AAPS J 11:217–224

    Article  CAS  PubMed  Google Scholar 

  55. Huang S-M, Strong JM, Zhang L, Reynolds KS, Nallani S, Temple R, Abraham S, Habet SA, Baweja RK, Burckart GJ, Chung S, Colangelo P, Frucht D, Green MD, Hepp P, Karnaukhova E, Ko HS, Lee JI, Marroum PJ, Norden JM, Qiu W, Rahman A, Sobel S, Stifano T, Thummel K, Wei XX, Yasuda S, Zheng JH, Zhao H, Lesko LJ (2008) New era in drug interaction evaluation: US Food and Drug Administration update on CYP enzymes, transporters, and the guidance process. J Clin Pharmacol 48:662–670

    Article  CAS  PubMed  Google Scholar 

  56. Huang S-M, Zhao H, Lee JI, Reynolds K, Zhang L, Temple R, Lesko LJ (2010) Therapeutic protein–drug interactions and implications for drug development. Clin Pharmacol Ther 87:497–503

    Article  CAS  PubMed  Google Scholar 

  57. Zhang L, Zhang YD, Zhao P, Huang S-M (2009) Predicting drug–drug interactions: an FDA perspective. AAPS J 11:300–306

    Article  CAS  PubMed  Google Scholar 

  58. Zhang L, Reynolds KS, Zhao P, Huang S-M (2010) Drug interactions evaluation: an integrated part of risk assessment of therapeutics. Toxicol Appl Pharmacol 243:134–145

    Article  CAS  PubMed  Google Scholar 

  59. Duan JZ, Jackson AJ, Zhao P (2010, in press) Bioavailability considerations in evaluating drug–drug interactions using the population pharmacokinetic approach. J Clin Pharmacol. doi:10.1177/0091270010372387

  60. Zhao P, Zhang L, Huang S-M (2010) Complex drug interactions: significance and evaluation. In: Pang KS, Rodrigues AD, Peter RM (eds) Enzyme- and transporter-based drug–drug interactions: progress and future challenges. Springer, New York, 746 pp

  61. Food and Drug Administration (2005) Guidance for industry clinical lactation studies—study design, data analysis, and recommendations for labeling. http://www.fda.gov/downloads/RegulatoryInformation/Guidances/ucm127505.pdf

  62. European Medicines Agency (2008) EMEA workshop on modeling in paediatric medicines. http://wwwemaeuropaeu/meetings/conferences/1415apr08htm

  63. Manolis E, Pons G (2009) Proposals for model-based paediatric medicinal development within the current European Union regulatory framework. Br J Clin Pharmacol 68:493–501

    Article  PubMed  Google Scholar 

  64. Garnett CE, Beasley N, Bhattaram VA, Jadhav PR, Madabushi N, Stockbridge N, Tornoe CW, Wang Y, Zhu H, Gobburu JVS (2008) Concentration-QT relationships play a key role in the evaluation of proarrhythmic risk during regulatory review. J Clin Pharmacol 48:13–18

    Article  CAS  PubMed  Google Scholar 

  65. Garnett C, Lee JY, Gobburu JVS (2011) U.S. FDA perspective: impact of modeling and simulation on regulatory decision making. In: Kimko HC, Peck CC (eds) Clinical trials simulation: applications and trends. Springer

  66. Zhu H, Wang Y, Gobburu JV, Garnett CE (2010) Considerations for clinical trial design and data analyses of thorough QT studies using drug–drug interaction. J Clin Pharmacol 50:1106–1111

    Google Scholar 

  67. Bhattaram V, Siddiqui O, Kapcala L, Gobburu J (2009) Endpoints and analyses to discern disease-modifying drug effects in early Parkinson’s disease. AAPS J 11:456–464

    Article  CAS  PubMed  Google Scholar 

  68. Wang Y, Bhattaram VA, Jadhav PR, Lesko LJ, Madabushi R, Powell JR, Qiu W, Sun H, Yim DS, Zheng JJ, Gobburu JVS (2008) Leveraging prior quantitative knowledge to guide drug development decisions and regulatory science recommendations: impact of FDA pharmacometrics during 2004–2006. J Clin Pharmacol 48:146–156

    Article  CAS  PubMed  Google Scholar 

  69. Lappin G, Kuhnz W, Jochemsen R, Kneer J, Chaudhary A, Oosterhuis B, Drijfhout WJ, Rowland M, Garner RC (2006) Use of microdosing to predict pharmacokinetics at the therapeutic dose: Experience with 5 drugs[ast]. Clin Pharmacol Ther 80:203–215

    Article  CAS  PubMed  Google Scholar 

  70. Food and Drug Administration (2006) Guidance for industry, investigators, and reviewers exploratory IND studies

  71. Goodsaid F, Frueh FW (2007) Implementing the U.S. FDA guidance on pharmacogenomic data submissions. Environ Mol Mutagen 48:354–358

    Article  CAS  PubMed  Google Scholar 

  72. Huang SM, Goodsaid F, Rahman A, Fruehand F, Lesko LJ (2006) Application of pharmacogenomics in clinical pharmacology. Toxicol Mech Methods 16:89–99

    Article  CAS  PubMed  Google Scholar 

  73. Sheiner LB (1991) The intellectual health of clinical drug evaluation. Clin Pharmacol Ther 50:4–9

    Article  CAS  PubMed  Google Scholar 

  74. Critical Path Initiative. Food and Drug Administration, Center for Drug Evaluation and Research (2006). <http://www.fda.gov/CriticalPath>

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

    Article  CAS  PubMed  Google Scholar 

  76. Peck CC, Rubin DB, Sheiner LB (2003) Hypothesis: a single clinical trial plus causal evidence of effectiveness is sufficient for drug approval. Clin Pharmacol Ther 74:481–490

    Article  Google Scholar 

  77. Doblin RE (2000) Regulation of the medical use of psychedelics and marijuana. PhD, Harvard University, Cambridge

    Google Scholar 

  78. Bhattaram VA, Booth BP, Ramchandani RP, Beasley BN, Wang Y, Tandon V, Duan JZ, Baweja RK, Marroum PJ, Uppoor RS, Rahman NA, Sahajwalla CG, Powell JR, Mehta M, Gobburu JV (2005) Impact of pharmacometrics on drug approval and labeling decisions: a survey of 42 new drug applications. AAPS J 7:E503–E512

    Google Scholar 

  79. Bhattaram VA, Bonapace C, Chilukuri DM, Duan JZ, Garnett C, Gobburu JV, Jang SH, Kenna L, Lesko LJ, Madabushi R, Men Y, Powell JR, Qiu W, Ramchandani RP, Tornoe CW, Wang Y, Zheng JJ (2007) Impact of pharmacometric reviews on new drug approval and labeling decisions—a survey of 31 new drug applications submitted between 2005 and 2006. Clin Pharmacol Ther 81:213–221

    Article  CAS  PubMed  Google Scholar 

  80. Garnett C, Lee J-Y, Gobburu JVS (2011, in press) U.S. FDA perspective: impact of modeling and simulation on regulatory decision making. In: Clinical trials simulation: applications and trends. Springer

  81. Gobburu VS (2010) Pharmacometrics 2020. J Clin Pharmacol 50(9):151S–157S

    Article  PubMed  Google Scholar 

  82. Miller R, Ewy W, Corrigan BW, Ouellet D, Hermann D, Kowalski KG, Lockwood P, Koup JR, Donevan S, El-Kattan A, Li CS, Werth JL, Feltner DD, Lalonde RL (2005) How modeling and simulation have enhanced decision making in new drug development. J Pharmackinet Pharmacodyn 32:185–197

    Article  Google Scholar 

  83. Olson SC, Bockbrader H, Boyd RA, Cook J, Koup JR, Lalonde RL, Siedlik PH, Powell JR (2000) Impact of population pharmacokinetic-pharmacodynamic analyses on the drug development process: experience at Parke–Davis. Clin Pharmacokinet 38:449–459

    Article  CAS  PubMed  Google Scholar 

  84. Reigner BG, Williams PE, Patel IH, Steimer JL, Peck C, van Brummelen P (1997) An evaluation of the integration of pharmacokinetic and pharmacodynamic principles in clinical drug development. Experience within Hoffmann La Roche. Clin Pharmacokinet 33:142–152

    Article  CAS  PubMed  Google Scholar 

  85. Miller R, Ewy W, Corrigan BW, Ouellet D, Hermann D, Kowalski KG, Lockwood P, Koup JR, Donevan S, El-Kattan A, Li CS, Werth JL, Feltner DE, Lalonde RL (2005) How modeling and simulation have enhanced decision making in new drug development 2. J Pharmacokinet Pharmacodyn 32:185–197

    Article  PubMed  Google Scholar 

  86. Balant L, Rowland M, Peck C (2000) Optimizing the science of drug development: opportunities for better candidate selection and accelerated evaluation in humans. J Clin Pharm 40:803–814

    Article  Google Scholar 

  87. Rowland M, Tozer TN (2006) Introduction to pharmacokinetics and pharmacodynamics: the quantitative basis of drug therapy. Lippincott, Williams & Wilkins, Baltimore

    Google Scholar 

  88. Rowland M, Tozer TN (1980, 1988, 1995, 2011) Clinical pharmacokinetics and pharmacodynamics: concepts and applications. Wolters Kluwer Health, Lippincott, Williams and Wilkins

  89. Huang S-M (October 1, 2009) Personal Communication

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Carl C. Peck.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Peck, C.C. Quantitative clinical pharmacology is transforming drug regulation. J Pharmacokinet Pharmacodyn 37, 617–628 (2010). https://doi.org/10.1007/s10928-010-9171-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10928-010-9171-3

Keywords

Navigation