1932

Abstract

Adaptive enrichment designs for clinical trials may include rules that use interim data to identify treatment-sensitive patient subgroups, select or compare treatments, or change entry criteria. A common setting is a trial to compare a new biologically targeted agent to standard therapy. An enrichment design's structure depends on its goals, how it accounts for patient heterogeneity and treatment effects, and practical constraints. This article first covers basic concepts, including treatment-biomarker interaction, precision medicine, selection bias, and sequentially adaptive decision making, and briefly describes some different types of enrichment. Numerical illustrations are provided for qualitatively different cases involving treatment-biomarker interactions. Reviews are given of adaptive signature designs; a Bayesian design that uses a random partition to identify treatment-sensitive biomarker subgroups and assign treatments; and designs that enrich superior treatment sample sizes overall or within subgroups, make subgroup-specific decisions, or include outcome-adaptive randomization.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-statistics-040720-032818
2021-03-07
2024-04-26
Loading full text...

Full text loading...

/deliver/fulltext/statistics/8/1/annurev-statistics-040720-032818.html?itemId=/content/journals/10.1146/annurev-statistics-040720-032818&mimeType=html&fmt=ahah

Literature Cited

  1. Bai X, Tsiatis AA, Lu W, Song R. 2017. Optimal treatment regimes for survival endpoints using a locally-efficient doubly-robust estimator from a classification perspective. Lifetime Data Anal. 23:585–604
    [Google Scholar]
  2. Chapple A, Thall P. 2018. Subgroup-specific dose finding in phase I clinical trials based on time to toxicity allowing adaptive subgroup combination. Pharm. Stat. 17:734–49
    [Google Scholar]
  3. Chapple A, Thall P. 2019. A hybrid phase I-II/III clinical trial design allowing dose re-optimization in phase III (with discussion). Biometrics 75:371–81
    [Google Scholar]
  4. Dagogo-Jack I, Shaw A. 2018. Tumour heterogeneity and resistance to cancer therapies. Nat. Rev. Clin. Oncol. 15:81–94
    [Google Scholar]
  5. Dixon DO, Simon R. 1991. Bayesian subset analysis. Biometrics 47:871–81
    [Google Scholar]
  6. Dixon DO, Simon R. 1992. Bayesian subset analysis in a colorectal cancer clinical trial. Stat. Med. 11:13–22
    [Google Scholar]
  7. FDA (US Food Drug Admin.). 2011. Clinical considerations for therapeutic cancer vaccines Guid. Ind. FDA-2009-D-0427, US Food Drug Admin., Silver Spring, MD
  8. Fisher R, Pusztai L, Swanton C. 2013. Cancer heterogeneity: implications for targeted therapeutics. Br. J. Cancer 108:479–85
    [Google Scholar]
  9. Flehinger B, Louis T, Robbins H, Singer B. 1972. Reducing the number of inferior treatments in clinical trials. PNAS 69:2993–94
    [Google Scholar]
  10. Foster J, Taylor J, Ruberg S. 2011. Subgroup identification from randomized clinical trial data. Stat. Med. 30:2867–80
    [Google Scholar]
  11. Freidlin B, Jiang W, Simon R. 2010. The cross-validated adaptive signature design. Clin. Cancer Res. 16(2):691–98
    [Google Scholar]
  12. Freidlin B, Simon R. 2005. Evaluation of randomized discontinuation design. J. Clin. Oncol. 23:5094–98
    [Google Scholar]
  13. Gauthier J, Thall P, Yuan Y. 2019. Bayesian phase 1/2 trial designs and cellular immunotherapies: a practical primer. Cell Gene Ther. Insights 5:1483–94
    [Google Scholar]
  14. Griffiths T, Ghahramani Z. 2006. Infinite latent feature models and the Indian buffet process. Advances in Neural Information Processing Systems 18 (NIPS 2005) Y Weiss, B Schölkopf, JC Platt475–82 Cambridge, MA: MIT Press
    [Google Scholar]
  15. Griffiths T, Ghahramani Z. 2011. The Indian buffet process: an introduction and review. J. Mach. Learn. Res. 12:1185–224
    [Google Scholar]
  16. Hey SP, Kimmellman J. 2015. Are outcome-adaptive allocation trials ethical?. Clin. Trials 12:102–6
    [Google Scholar]
  17. Hibbard J, Friedstat J, Thomas S, Edkins R, Hultman C, Kosorok M. 2018. Liberti: a smart study in plastic surgery. Clin. Trials 15:286–93
    [Google Scholar]
  18. Hsieh LL, Erl TK, Chen CC, Hsieh JS, Chang JG, Liu TC. 2012. Characteristics and prevalence of KRAS, BRAF, and PIK3CA mutations in colorectal cancer by high-resolution melting analysis in Taiwanese population. Clin. Chim. Acta 413:1605–11
    [Google Scholar]
  19. James N, Sydes M, Clarke N, Mason M, Dearnaley D, et al. 2009. Systemic therapy for advancing or metastatic prostate cancer (STAMPEDE): a multi-arm, multistage randomized controlled trial. BJU Int. 103:464–69
    [Google Scholar]
  20. Jennison C, Turnbull B. 2007. Adaptive seamless designs: selection and prospective testing of hypotheses. J. Biopharm. Stat. 17:1135–61
    [Google Scholar]
  21. Jiang W, Freidlin B, Simon R. 2007. Biomarker-adaptive threshold design: A procedure for evaluating treatment with possible biomarker-defined subset effect. J. Natl. Cancer Inst. 99:1036–43
    [Google Scholar]
  22. Kim E, Herbst R, Wistuba I, Lee JJ, Blumenschein GR Jr., et al. 2011. The BATTLE trial: personalizing therapy for lung cancer. Cancer Discov. 1:44–53
    [Google Scholar]
  23. Korn EL, Freidlin B. 2011. Outcome-adaptive randomization: Is it useful?. J. Clin. Oncol. 29:771–76
    [Google Scholar]
  24. Kosorok M, Moodie EEM, eds. 2016. Adaptive Treatment Strategies in Practice: Planning Trials and Analyzing Data for Personalized Medicine. Philadelphia: SIAM
  25. Lai TL, Lavori P, Tsang K. 2019. Adaptive enrichment designs for confirmatory trials. Stat. Med. 38:613–24
    [Google Scholar]
  26. Lee J, Mueller P, Sengupta S, Gulukota K, Ji Y. 2016. Bayesian inference for intratumor heterogeneity in mutations and copy number variation. J. R. Stat. Soc. B 65:547–63
    [Google Scholar]
  27. Lee J, Thall PF, Ji Y, Müller P. 2015. Bayesian dose-finding in two treatment cycles based on the joint utility of efficacy and toxicity. J. Am. Stat. Assoc. 110:711–22
    [Google Scholar]
  28. Lee J, Thall PF, Rezvani K. 2019. Optimizing natural killer cell doses for heterogeneous cancer patients based on multiple event times. J. R. Stat. Soc. B 68:461–74
    [Google Scholar]
  29. Lin R, Thall P, Yuan Y. 2020. An adaptive trial design to optimize dose–schedule regimes with delayed outcomes. Biometrics 76:304–15
    [Google Scholar]
  30. Lipkovich I, Dmitrienko A, D'Agostino R. 2017. Tutorial in biostatistics: data-driven subgroup identification and analysis in clinical trials. Stat. Med. 36:136–96
    [Google Scholar]
  31. Magnusson B, Turnbull B. 2013. Group sequential enrichment design incorporating subgroup selection. Stat. Med. 32:2695–714
    [Google Scholar]
  32. Maki R, Wathen J, Hensley M, Patel S, Priebat D, et al. 2007. An adaptively randomized phase III study of gemcitabine and docetaxel versus gemcitabine alone in patients with metastatic soft tissue sarcomas. J. Clin. Oncol. 25:2755–63
    [Google Scholar]
  33. Murphy S. 2005. An experimental design for the development of adaptive treatment strategies. Stat. Med. 24:1455–81
    [Google Scholar]
  34. Nahum-Shani I, Smith S, Spring B, Collins L, Witkiewitz K, et al. 2017. Just-in-time adaptive interventions (JITAIs) in mobile health: key components and design principles for ongoing health behavior support. Ann. Behav. Med. 52:446–62
    [Google Scholar]
  35. Ondra T, Dmitrienko A, Friede T, Graf A, Miller F, et al. 2016. Methods for identification and confirmation of targeted subgroups in clinical trials: a systematic review. J. Biopharm. Stat. 26:99–119
    [Google Scholar]
  36. Robins JM, Hernán MA, Brumback B. 2000. Marginal structural models and causal inference in epidemiology. Epidemiology 11:550–60
    [Google Scholar]
  37. Rosenbaum P, Rubin D. 1985. Constructing a control group using multivariate matched sampling methods that incorporate the propensity score. Am. Stat. 39:33–38
    [Google Scholar]
  38. Rosenberger W, Sverdlov O, Hu F. 2012. Adaptive randomization for clinical trials. J. Biopharm. Stat. 22:719–36
    [Google Scholar]
  39. Rosenblum M, Hanley D. 2017. Topical review: adaptive enrichment designs for stroke clinical trials. Stat. Med. 33:2021–25
    [Google Scholar]
  40. Rosenblum M, Qian T, Du Y, Qiu H, Fisher A. 2016. Multiple testing procedures for adaptive enrichment designs: combining group sequential and reallocation approaches. Biostatistics 17:650–62
    [Google Scholar]
  41. Schaid DJ, Wieand S, Therneau TM. 1990. Optimal two stage screening designs for survival comparisons. Biometrika 77:507–13
    [Google Scholar]
  42. Simon N, Simon R. 2013. Adaptive enrichment designs for clinical trials. Biostatistics 14:613–25
    [Google Scholar]
  43. Simon N, Simon R. 2018. Using Bayesian modeling in frequentist adaptive enrichment designs. Biostatistics 19:27–41
    [Google Scholar]
  44. Stallard N, Todd S. 2003. Sequential designs for phase III clinical trials incorporating treatment selection. Stat. Med. 22:689–703
    [Google Scholar]
  45. Steingrimsson J, Betz J, Qian T, Rosenblum M 2019. Optimized adaptive enrichment designs for three-arm trials: learning which subpopulations benefit from different treatments. Biostatistics https://doi.org/10.1093/biostatistics/kxz030
    [Crossref] [Google Scholar]
  46. Sverdlov O 2015. Modern Adaptive Randomized Clinical Trials: Statistical and Practical Aspects Boca Raton, FL: CRC, Taylor and Francis
  47. Thall P 2020. Statistical Remedies for Medical Researchers New York: Springer
  48. Thall P, Fox P, Wathen J. 2015. Statistical controversies in clinical research: scientific and ethical problems with adaptive randomization in comparative clinical trials. Ann. Oncol. 26:1621–28
    [Google Scholar]
  49. Thall P, Logothetis C, Pagliaro L, Wen S, Brown M, et al. 2007. Adaptive therapy for androgen independent prostate cancer: a randomized selection trial including four regimens. J. Natl. Cancer Inst. 99:1613–22
    [Google Scholar]
  50. Thall PF, Simon R, Ellenberg SS 1988. Two-stage selection and testing designs for comparative clinical trials. Biometrika 75:2303–10
    [Google Scholar]
  51. Thall PF, Wathen JK. 2005. Covariate-adjusted adaptive randomization in a sarcoma trial with multi-stage treatments. Stat. Med. 24:1947–64
    [Google Scholar]
  52. Thall PF, Wathen JK. 2007. Practical Bayesian adaptive randomisation in clinical trials. Eur. J. Cancer 43:859–66
    [Google Scholar]
  53. Thompson W. 1933. On the likelihood that one unknown probability exceeds another in view of the evidence of the two samples. Biometrika 25:285–94
    [Google Scholar]
  54. Trippa L, Rosner G, Mueller P. 2012. Bayesian enrichment strategies for randomized discontinuation trials. Biometrics 68:203–11
    [Google Scholar]
  55. Tsiatis A, Davidian M, Holloway S, Laber E 2019. Dynamic Treatment Regimes: Statistical Methods for Precision Medicine Boca Raton, FL: CRC
  56. Wathen JK, Thall PF. 2017. A simulation study of outcome adaptive randomization in multi-arm clinical trials. Clin. Trials 14:432–40
    [Google Scholar]
  57. Weber LM, Robinson MD. 2016. Comparison of clustering methods for high-dimensional single-cell flow and mass cytometry data. Cytometry Part A 89:1084–96
    [Google Scholar]
  58. Xu Y, Mueller P, Yuan Y, Gulukota K, Ji Y. 2015. Mad Bayes for tumor heterogeneity—feature allocation with exponential family sampling. J. Am. Stat. Assoc. 110:503–14
    [Google Scholar]
  59. Xu Y, Trippa L, Mueller P, Ji Y. 2016. Subgroup-based adaptive SUBA designs for multi-arm biomarker trials. Stat. Biosci. 8:159–80
    [Google Scholar]
  60. Yan F, Thall PF, Lu K, Gilbert M, Yuan Y. 2018. Phase I-II clinical trial design: a state-of-the-art paradigm for dose finding with novel agents. Ann. Oncol. 29:694–99
    [Google Scholar]
  61. Yuan Y, Nguyen H, Thall P 2016. Bayesian Designs for Phase I–II Clinical Trials Boca Raton, FL: CRC
  62. Yuan Y, Yin G. 2011. On the usefulness of outcome adaptive randomization. J. Clin. Oncol. 29:771–76
    [Google Scholar]
  63. Zhang Z, Li M, Soon G, Greene T, Shen C. 2017. Subgroup selection in adaptive signature designs of confirmatory clinical trials. J. R. Stat. Soc. C 66:345–61
    [Google Scholar]
  64. Zhang Z, Wang C, Nie L, Soon G. 2013. Assessing the heterogeneity of treatment effects via potential outcomes of individual patients. J. R. Stat. Soc. C 62:687–704
    [Google Scholar]
/content/journals/10.1146/annurev-statistics-040720-032818
Loading
/content/journals/10.1146/annurev-statistics-040720-032818
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error