Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Opinion
  • Published:

The future of drug development: advancing clinical trial design

Abstract

Declining pharmaceutical industry productivity is well recognized by drug developers, regulatory authorities and patient groups. A key part of the problem is that clinical studies are increasingly expensive, driven by the rising costs of conducting Phase II and III trials. It is therefore crucial to ensure that these phases of drug development are conducted more efficiently and cost-effectively, and that attrition rates are reduced. In this article, we argue that moving from the traditional clinical development approach based on sequential, distinct phases towards a more integrated view that uses adaptive design tools to increase flexibility and maximize the use of accumulated knowledge could have an important role in achieving these goals. Applications and examples of the use of these tools — such as Bayesian methodologies — in early- and late-stage drug development are discussed, as well as the advantages, challenges and barriers to their more widespread implementation.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: A novel model for clinical development.
Figure 2: Dose selection in the development of a therapeutic for Muckle–Wells syndrome.
Figure 3: Re-estimating sample size while maintaining statistical power.

Similar content being viewed by others

References

  1. Booth, B & Zemmel, R. Prospects for productivity. Nature Rev. Drug Discov. 3, 451–456 (2004).

    Article  CAS  Google Scholar 

  2. Gilbert, J., Henske, P. & Singh, A. Rebuilding big pharma's business model. In Vivo 21, 1–4 (2003).

    Google Scholar 

  3. Kola, I. & Landis, J. Can the pharmaceutical industry reduce attrition rates? Nature Rev. Drug Discov. 3, 711–716 (2004).

    Article  CAS  Google Scholar 

  4. Budget US Govt., App., FY 1993–2003.

  5. Parexel's Pharmaceutical R&D Statistical Sourcebook 2002/2003 (Parexel International, Waltham, USA, 2003).

  6. Adams, C. P. & Brantner, V. V. Spending on new drug development. Health Econ. 26 Feb 2009 (doi: 10.1002/hec.1454).

  7. DiMasi, J. A., Hansen, R. W. & Grabowski, H. G. The price of innovation: new estimates of drug development costs. J. Health Econ. 22, 151−185 (2003).

  8. Adams, C. & Brantner, V. V. Estimating the cost of new drug development: is it really $802 million? Health Aff. 2, 420–428 (2006).

    Article  Google Scholar 

  9. Pharmaceutical R&D Factbook 2007 (CMR International, London, UK, 2007).

  10. KMR General Metrics Study (KMR Group, Chicago, USA, 2007).

  11. Sheiner, L. B. & Steimer, J.-L. Pharmacokinetic/pharmacodynamic modeling in drug development. Ann. Rev. Pharmacol. Toxicol. 40, 67–95 (2000).

    Article  CAS  Google Scholar 

  12. Lalonde, R. L. et al. Model-based drug development. Clin. Pharmacol. Ther. 82, 21–32 (2007).

    Article  CAS  Google Scholar 

  13. Breimer, D. D. & Danhof, M. Relevance of the application of pharmacokinetic–pharmacodynamic modeling concepts in drug development. The “Wooden Shoe” paradigm. Clin. Pharmacokinet. 32, 259–267 (1997).

    Article  CAS  Google Scholar 

  14. Danhof, M., Alvan, G., Dahl, S. G., Kuhlmann, J. & Paintaud, G. Mechanism-based pharmacokinetic/pharmacodynamic modeling — a new classification of biomarkers. Pharm. Res. 22, 1432–1437 (2005).

    Article  CAS  Google Scholar 

  15. Holford, N. H. G., Kimko, H. C., Monteleone, J. P. R. & Peck, C. C. Simulation of clinical trials. Annu. Rev. Pharmacol. Toxicol. 40, 209–234 (2000).

    Article  CAS  Google Scholar 

  16. Miller, R. et al. How modeling and simulation have enhanced decision making in new drug development. J. Pharmacokinet. Pharmacodyn. 32, 185–197 (2005).

    Article  Google Scholar 

  17. Berry, D. A. Bayesian statistics. Med. Decis. Making 26, 429–430 (2006).

    Article  Google Scholar 

  18. Müller, P., Berry, D. A., Grieve, A. P., Smith, M. & Krams, M. Simulation-based sequential Bayesian design. J. Stat. Plan. Inference 137, 3140–3150 (2007).

    Article  Google Scholar 

  19. Goggin, T. et al. Modeling and simulation of clinical trials: an industrial perspective. In Simulation for Designing of Clinical Trials (eds Kimko, H. C. & Duffull, S. B.) 227–224 (Marcel Dekker, New York, USA, 2002).

    Google Scholar 

  20. Reigner, B. G. et al. An evaluation of the integration of pharmacokinetic and pharmacodynamic principles in clinical drug development. Experience within Hoffman La Roche. Clin. Pharmacokinet. 33, 142–152 (1997).

    Article  CAS  Google Scholar 

  21. Lachmann, H. J. et al. Use of canakinumab in the cryopyrin-associated periodic syndrome. N. Engl. J. Med. 360, 2416–2425 (2009).

    Article  CAS  Google Scholar 

  22. Berry, D. A. Bayesian clinical trials. Nature Rev. Drug Discov. 5, 27–36 (2006).

    Article  CAS  Google Scholar 

  23. Berry, D. A. Introduction to Bayesian methods III: use and interpretation of Bayesian tools in design and analysis. Clin. Trials 2, 295–300 (2005).

    Article  Google Scholar 

  24. Berry, D. A. Adaptive trial design. Clin. Adv. Hematol. Oncol. 5, 522–524 (2007).

    PubMed  Google Scholar 

  25. Krams, M. et al. Adaptive designs in clinical drug development: opportunities, challenges, and scope reflections following PhRMA's November 2006 workshop. J. Biopharm. Stat. 17, 957–964 (2007).

    Article  Google Scholar 

  26. Gallo, P. et al. Adaptive designs in clinical drug development — an executive summary of the PhRMA Working Group. J. Biopharm. Stat. 16, 275–283 (2006).

    Article  Google Scholar 

  27. Bornkamp, B. et al. Innovative approaches for designing and analyzing adaptive dose-ranging trials. J. Biopharm. Stat. 17, 965–995 (2007).

    Article  Google Scholar 

  28. Weir, C. J., Spiegelhalter, D. J. & Grieve, A. P. Flexible design and efficient implementation of adaptive dose-finding studies. J. Biopharm. Stat. 17, 1033–1050 (2007).

    Article  Google Scholar 

  29. Bretz, F., Schmidli, H., König, F., Racine, A. & Maurer, W. Confirmatory seamless Phase II/III clinical trials with hypotheses selection at interim: general concepts. Biom. J. 48, 623–634 (2006).

    Article  Google Scholar 

  30. Mehta, C. R. & Patel, N. R. Adaptive, group sequential and decision theoretic approaches to sample size determination. Stat. Med. 25, 3250–3269 (2006).

    Article  Google Scholar 

  31. Chuang-Stein, C., Anderson, K., Gallo, P. & Collins, S. Sample size reestimation: a review and recommendations. Drug Inf. J. 40, 475–484 (2006).

    Article  Google Scholar 

  32. Golub, H. L. The need for more efficient clinical trials. Stat. Med. 25, 3231–3235 (2006).

    Article  Google Scholar 

  33. Tsiatis, A. A. & Mehta, C. On the inefficiency of the adaptive design for monitoring clinical trials. Biometrika 90, 367–378 (2003).

    Article  Google Scholar 

Download references

Acknowledgements

A special acknowledgment to W. Dere (Amgen), S. Cummings (UCSF), A. Lee (Pfizer) and E. Berndt (MIT-CBI) for significant contributions to discussions leading to this manuscript. Also, many thanks to the McKinsey Trial Design Team for their support (M. E., E. F., N. S. and T. T.).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to John Orloff.

Ethics declarations

Competing interests

Some authors are employed by, or have consulting relationships with, pharmaceutical companies, biotech companies, contract research organizations, academic institutions, consulting firms and/or research foundations, that might be perceived as a competing interest.

Supplementary information

Supplementary information S1 (box)

Case study: dose selection in type 2 diabetes mellitus (PDF 920 kb)

Supplementary information S2 (box)

Case study: dyslipidaemia proof-of-concept study (PDF 434 kb)

Supplementary information S3 (figure)

Advantages of a seamless/adaptive trial design compared with classical Phase IIb and III studies (PDF 259 kb)

Related links

Related links

DATABASES

OMIM

Muckle–Wells syndrome

type 2 diabetes

Rights and permissions

Reprints and permissions

About this article

Cite this article

Orloff, J., Douglas, F., Pinheiro, J. et al. The future of drug development: advancing clinical trial design. Nat Rev Drug Discov 8, 949–957 (2009). https://doi.org/10.1038/nrd3025

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrd3025

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing