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:

Integrating predictive biomarkers and classifiers into oncology clinical development programmes

Abstract

The future of drug development in oncology lies in identifying subsets of patients who will benefit from particular therapies, using predictive biomarkers. These technologies offer hope of enhancing the value of cancer medicines and reducing the size, cost and failure rates of clinical trials. However, examples of the failure of predictive biomarkers also exist. In these cases the use of biomarkers increased the costs, complexity and duration of clinical trials, and narrowed the treated population unnecessarily. Here, we present methods to adaptively integrate predictive biomarkers into clinical programmes in a data-driven manner, wherein these biomarkers are emphasized in exact proportion to the evidence supporting their clinical predictive value. The resulting programme demands value from predictive biomarkers and is designed to optimally harvest this value for oncology drug development.

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: Relative efficiency of two small POC trials (E2) compared to one traditionally sized POC trial (E1).
Figure 2: Two-dimensional decision graph for decision analysis-guided Phase II–Phase III predictive biomarker transition.
Figure 3: Traditional confirmatory phase development compared to the Phase II+ method.

Similar content being viewed by others

References

  1. Incorporating health economics in guidelines and assessing resource impact. NICE Guidelines Manual [online] (2005).

  2. Cobleigh, M. A. et al. Multinational study of the efficacy and safety of humanized anti-HER2 monoclonal antibody in women who have HER2-overexpressing metastatic breast cancer that has progressed after chemotherapy for metastatic disease. J. Clin. Oncol. 17, 2639–2648 (1999).

    Article  CAS  Google Scholar 

  3. Slamon, D. J. et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N. Engl. J. Med. 344, 783–792 (2001).

    Article  CAS  Google Scholar 

  4. Paez, J. G. et al. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science 30, 1497–1500 (2004).

    Article  Google Scholar 

  5. Pao, W. et al. EGF receptor gene mutations are common in lung cancers from “never smokers” and are associated with sensitivity of tumors to gefitinib and erlotinib. Proc. Natl Acad. Sci. USA 101, 13306–13311 (2004).

    Article  CAS  Google Scholar 

  6. Amado, R. G. et al. Wild-type KRAS is required for panitumumab efficacy in patients with metastatic colorectal cancer. J. Clin. Oncol. 26, 1626–1634 (2008).

    Article  CAS  Google Scholar 

  7. Lievre, A. et al. KRAS mutation status is predictive of response to cetuximab therapy in colorectal cancer. Cancer Res. 66, 3992–3995 (2006).

    Article  CAS  Google Scholar 

  8. Bokemeyer, C. et al. KRAS status and efficacy of first-line treatment of patients with metastatic colorectal cancer (mCRC) with FOLFOX with or without cetuximab: the OPUS experience. J. Clin. Oncol. Abstr. 26, 4000 (2008).

    Article  Google Scholar 

  9. Van Cutsem, E. et al. KRAS status and efficacy in the first-line treatment of patients with metastatic colorectal cancer (mCRC) treated with FOLFIRI with or without cetuximab: the CRYSTAL experience. J. Clin. Oncol. Abstr. 26, 2 (2008).

    Article  Google Scholar 

  10. Dalton, W. S. & Friend, S. H. Cancer biomarkers — an invitation to the table. Science 312, 1165–1168 (2006).

    Article  CAS  Google Scholar 

  11. Beckman, R. A. Analyze the current biomarker-guided development paradigm: biomarkers are instrumental for any development in oncology. Center for Business Intelligence 2nd Annual Forum on Oncology Commercialization Strategies [online lecture], (Philadelphia, Pennsylvania, USA; 12–13 January 2009).

    Google Scholar 

  12. Yan, L. & Beckman, R. A. Pharmacogenetics and pharmacogenomics in oncology therapeutic antibody development. Biotechniques 39, 565–568 (2005).

    Article  Google Scholar 

  13. Ransohoff, D. F. & Gourlay, M. L. Sources of bias in specimens for research about molecular markers for cancer. J. Clin. Oncol. 28, 698–704 (2010).

    Article  Google Scholar 

  14. Ratain, M. J. & Glassman, R. H. Biomarkers in Phase I oncology trials: signal, noise, or expensive distraction? Clin. Cancer Res. 13, 6545–6548 (2007).

    Article  CAS  Google Scholar 

  15. Orloff, J. et al. The future of drug development: advancing clinical trial design. Nature Rev. Drug Discov. 8, 949–957 (2009).

    Article  CAS  Google Scholar 

  16. Ioannidis, J. P. A. & Panagiotou, O. A. Comparison of effect sizes associated with biomarkers reported in highly cited individual articles and in subsequent meta-analyses. JAMA 305, 2200–2210 (2011).

    Article  CAS  Google Scholar 

  17. Freidlin, B. & Simon, R. Adaptive signature design: an adaptive clinical trial design for generating and prospectively testing a gene expression signature for sensitive patients. Clin. Cancer Res. 11, 7872–7878 (2005).

    Article  CAS  Google Scholar 

  18. Chen, C. & Beckman, R. A. Hypothesis testing in a confirmatory Phase III trial with a possible subset effect. Stat. Biopharm. Res. 1, 431–440 (2009).

    Article  Google Scholar 

  19. Van Dyke, T. & Jacks, T. Cancer modeling in the modern era: progress and challenges. Cell 108, 135–144 (2002).

    Article  CAS  Google Scholar 

  20. Fiebig, H. H., Maier, A. & Burger, A. M. Clonogenic assay with established human tumor xenografts: correlation of in vitro to in vivo activity as a basis for anticancer drug discovery. Eur. J. Cancer 40, 802–820 (2004).

    Article  CAS  Google Scholar 

  21. Barker, A. D. et al. I-SPY 2: an adaptive breast cancer trial design in the setting of neoadjuvant chemotherapy. Clin. Pharmacol. Ther. 86, 97–100 (2009).

    Article  CAS  Google Scholar 

  22. Kim, E. S. et al. The BATTLE trial (Biomarker-integrated Approaches of Targeted Therapy for Lung Cancer Elimination): personalizing therapy for lung cancer. in Proceedings of the 101st Annual Meeting of the American Association for Cancer Research (17–21 Apr 2010; Washington DC, Philadelphia, USA; Abstract LB-1).

  23. Chen, C. & Beckman, R. A. Optimal cost-effective designs of proof of concept trials and associated go-no go decisions. Proc. Am. Stat. Assoc. 394–399 (2007).

  24. Chen, C. & Beckman, R. A. Optimal cost-effective go-no go decisions in late-stage oncology drug development. Stat. Biopharm. Res. 1, 159–169 (2009).

    Article  Google Scholar 

  25. Chen, C. & Beckman, R. A. Optimal cost-effective designs of Phase II proof of concept trials and associated go-no go decisions. J. Biopharm. Stat. 19, 424–436 (2009).

    Article  Google Scholar 

  26. Strauss, N. & Simon, R. Investigating a sequence of randomized Phase II trials to discover promising treatments. Stat. Med. 14, 1479–1489 (1995).

    Article  CAS  Google Scholar 

  27. Freidlin, B., McShane L. & Korn, E. L. Randomized clinical trials with biomarkers: design issues. J. Natl Cancer Inst. 102, 152–160 (2010).

    Article  CAS  Google Scholar 

  28. Maheswaren, S. et al. Detection of mutations in EGFR in circulating lung-cancer cells. N. Engl. J. Med. 359, 366–377 (2008).

    Article  Google Scholar 

  29. Tang, P. A., Bentsen, S. M, Chen, E. X. & Siu, L. L. Surrogate endpoints for median overall survival in metastatic colorectal cancer: literature-based analysis from 39 randomized controlled trials of first-line chemotherapy. J. Clin. Oncol. 25, 4562–4568 (2007).

    Article  Google Scholar 

  30. Simon, R. M., Paik, S. & Hayes, D. F. Use of archived specimens in evaluation of prognostic and predictive biomarkers. J. Natl Cancer Inst. 101, 1446–1452 (2009).

    Article  Google Scholar 

  31. Freidlin, B., Jiang, W. & Simon, R. The cross-validated adaptive signature design. Clin. Cancer Res. 16, 691–698 (2010).

    Article  Google Scholar 

  32. Simon, R. The use of genomics in clinical trial design. Clin. Cancer Res. 14, 5984–5993 (2008).

    Article  Google Scholar 

  33. Zhao, Y. D., Dimitrienko, A. & Tamura, R. Design and analysis considerations in clinical trials with a sensitive subpopulation. Stat. Biopharm. Res. 2, 72–83 (2010).

    Article  Google Scholar 

  34. De Geus-Oei, L. F. et al. Monitoring and predicting response to therapy with 18F-FDG PET in colorectal cancer: a systematic review. J. Nuc. Med. 50, 43S–54S (2009).

    Article  CAS  Google Scholar 

  35. Weber, W. A. PET for response assessment in oncology: radiotherapy and chemotherapy. Br. J. Radiol. S28, 42–49 (2005).

    Article  Google Scholar 

  36. Goldman, B., LeBlanc, M. & Crowley, J. Interim futility analysis with intermediate endpoints. Clin. Trials 5, 14–22 (2008).

    Article  Google Scholar 

  37. Hunsicker, S., Zhao, Y. & Simon, R. M. A comparison of Phase II study strategies. Clin. Cancer Res. 15, 5950–5955 (2009).

    Article  Google Scholar 

  38. Chen, C. & Sun, L. On quantification of PFS effect for accelerated approval of oncology drugs. Stat. Biopharm. Res. 2011 (in the press).

  39. Brannath, W. et al. Confirmatory adaptive designs with Bayesian decision tools for a targeted therapy in oncology. Stat. Med. 28, 1445–1463 (2009).

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

  41. 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 

  42. Bretz, F. et al. Confirmatory seamless Phase II/III clinical trials with hypotheses selection at interim: general concepts. Biom. J. 48, 623–634 (2006).

    Article  Google Scholar 

  43. 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 

  44. Chuang-Stein, C. et al. Sample size re-estimation: a review and recommendations. Drug Inf. J. 40, 475–484 (2006).

    Article  Google Scholar 

  45. Berger, J. Statistical Decision Theory and Bayesian Analysis 2nd edn (Springer Verlag, New York, 1985).

    Book  Google Scholar 

Download references

Acknowledgements

We thank R. M. Simon, head of the Biometrics Division at the US National Cancer Institute, for his very helpful discussions and for his critical review of the manuscript. We also thank D. Parkinson, Chair of the Cancer Steering Committee, Biomarkers Consortium of the Foundation for the US National Institutes of Health, for his helpful comments. The inspiration for this article, which represents the opinion of the authors, came from crossfunctional discussions — led by R.A.B. — of predictive biomarker integration into clinical development plans. From Amgen, the following individuals contributed to these discussions: A. Ang, D. Beaupre, L. Chen, D. Freeman, E. Loh, I. McCaffrey, E. Rasmussen and M. Wolf. From Daiichi-Sankyo Pharmaceutical Development, the following individuals contributed to these discussions: M. Aonuma, T. Bocanegra, B. Dornseif, S. Ge, I. Gorbatchevsky, G. Gormley, A. Halim, X. Jin, P. Kumar, K. Liu, K. Nakamaru, M. Rosen, D. Salazar, R. Scheyer, G. Senaldi, S. Smith, A. Tse, R. von Roemeling, J. Walker, Q. Wang and Y. Wang. From Merck Research Laboratories, the following individuals contributed to these discussions: K. Anderson, D. Bergstrom, C. Buser-Doepner, P. Carroll, T. Demuth, P. Ehrlich, C. Gause, D. Geho, B. Gertz, G. Harris, K. Hsu, J. Milloy, R. Phillips, C. Pickett, P. Shaw and L. Yan. From Rosetta BioInpharmatics, T. Fare contributed to these discussions. From U3 Pharma, the following individuals contributed to these discussions: K. Akahane and T. Hettman. The affiliations listed are the affiliations of the individuals at the time the discussions occurred.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Robert A. Beckman.

Ethics declarations

Competing interests

Robert A. Beckman is an employee of Daiichi Sankyo Pharmaceutical Development and is a stockholder in Daiichi Sankyo Inc. and Johnson & Johnson corporation.

Jason Clark is an employee of Incyte Pharmaceuticals and a stockholder in Merck & Co., Inc.

Cong Chen is an employee of Merck & Co., Inc. and a stockholder in Merck & Co., Inc., Sanofi and Johnson & Johnson corporation.

Glossary

Analytical validation

A process for demonstrating that a biomarker assay (or assays) have suitable calibration properties, sensitivity, specificity, accuracy and reproducibility to be potentially approved for commercial use after further clinical validation.

Clinical benefit identification (ID) hypothesis

The hypothesis that a particular predictive biomarker classifier identifies the subset of patients who will benefit from the therapy.

Clinical validation

The demonstration that a biomarker assay (or assays) identify the subset of patients who will benefit clinically.

Ethics committees

Committees that are composed of clinical development experts, patient advocates and lay people; these committees evaluate proposed clinical research studies for a particular hospital or clinic before the studies are permitted to commence at that institution. These committees focus particularly on protecting the rights of patients. This term is used particularly in Europe.

Go/no-go criteria

Criteria that determine whether or not to proceed to a Phase III pivotal trial. If the observed clinical benefit in a Phase II proof-of-concept study is equal to or better than the go/no-go criterion, the same drug–indication combination is studied in a randomized Phase III study for the approval of health authorities.

In vitro diagnostic

A biomarker assay that has been approved for commercial use in conjunction with a therapy, for the purpose of identifying the subset of patients who will benefit from the therapy.

Institutional review boards

Committees that are composed of clinical development experts, patient advocates and lay people; these committees evaluate proposed clinical research studies for a particular hospital or clinic before the studies are permitted to commence at that institution. These committees focus particularly on protecting the rights of patients. This term is applied particularly in the United States.

Objective utility functions

Mathematical functions that quantify the usefulness (utility) of a particular strategy. They can be used to objectively compare strategies. An example is the efficiency function for proof-of-concept studies.

Power

The probability that the proof-of-concept study or another confirmatory study will correctly declare an effective drug to be effective. It is equal to 100% minus the false-negative rate (the type II error).

Predictive biomarkers

Biomarkers that utilize a baseline characteristic of a patient to predict whether or not the patient will benefit from an associated therapy.

Prognostic biomarker

A biomarker that utilizes a baseline characteristic of a patient to predict the patient's outcome independently of any specific therapy.

Proof of concept

Statistical evidence from a randomized Phase II study that a particular drug is beneficial in a particular clinical setting or indication.

Qualified clinical end points

Clinical end points in Phase II studies that have a reasonable likelihood of predicting true clinical benefit. Examples of qualified clinical end points include RECIST responses (tumour shrinkage or stabilization of tumour size, according to standard criteria) or — when measured in randomized studies — progression-free survival (the time from the start of the therapy to tumour worsening or death). These qualified clinical end points may predict actual clinical benefit measured by overall survival (the time from the start of the therapy to death) or quality of life.

Research grade assay

A biomarker assay that has undergone informal testing in the research laboratory but has not undergone formal analytical validation.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Beckman, R., Clark, J. & Chen, C. Integrating predictive biomarkers and classifiers into oncology clinical development programmes. Nat Rev Drug Discov 10, 735–748 (2011). https://doi.org/10.1038/nrd3550

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrd3550

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer