1 / 135

Adaptive Design Methods in Clinical Trials

Adaptive Design Methods in Clinical Trials. Shein-Chung Chow, PhD Department of Biostatistics and Bioinformatics Duke University School of Medicine Durham, North Carolina Presented at The 2011 MBSW, Muncie, Indiana May 23, 2011. Lecture 1 - Outline . What is adaptive design?

sal
Télécharger la présentation

Adaptive Design Methods in Clinical Trials

An Image/Link below is provided (as is) to download presentation Download Policy: Content on the Website is provided to you AS IS for your information and personal use and may not be sold / licensed / shared on other websites without getting consent from its author. Content is provided to you AS IS for your information and personal use only. Download presentation by click this link. While downloading, if for some reason you are not able to download a presentation, the publisher may have deleted the file from their server. During download, if you can't get a presentation, the file might be deleted by the publisher.

E N D

Presentation Transcript


  1. Adaptive Design Methods in Clinical Trials Shein-Chung Chow, PhD Department of Biostatistics and Bioinformatics Duke University School of Medicine Durham, North Carolina Presented at The 2011 MBSW, Muncie, Indiana May 23, 2011

  2. Lecture 1 - Outline • What is adaptive design? • Type of adaptive designs • Possible benefits • Regulatory perspectives • Protocol amendments

  3. What is adaptive design? • There is no universal definition. • Adaptive randomization, group sequential, and sample size re-estimation, etc. • PhRMA (2006) • FDA (2010) • Adaptive design is also known as • Flexible design (EMEA, 2002, 2006) • Attractive design (Uchida, 2006)

  4. PhRMA’s definition PhRMA (2006). J. Biopharm. Stat., 16 (3), 275-283. An adaptive design is referred to as a clinical trial design that uses accumulating data to decide on how to modify aspects of the study as it continues, without undermining the validity and integrity of the trial.

  5. PhRMA’s definition • Characteristics • Adaptation is a design feature. • Changes are made by design not on an ad hoc basis. • Comments • It does not reflect real practice. • It may not be flexible as it means to be.

  6. FDA’s definition US FDA Guidance for Industry – Adaptive Design Clinical Trials for Drugs and Biologics Feb, 2010 An adaptive design clinical study is defined as a study that includes a prospectively planned opportunity for modification of one or more specified aspects of the study design and hypotheses based on analysis of data (usually interim data) from subjects in the study

  7. FDA’s definition Characteristics Adaptation is a prospectively planned opportunity. Changes are made based on analysis of data (usually interim data). Does not include medical devices? Comments It classifies adaptive designs into well-understood and less well-understood designs It does not reflect real practice (protocol amendments) It is not a guidance but a document of caution

  8. Adaptation • An adaptation is defined as a change or modification made to a clinical trial before and during the conduct of the study. • Examples include • Relax inclusion/exclusion criteria • Change study endpoints • Modify dose and treatment duration etc.

  9. Types of adaptations • Prospective adaptations • Adaptive randomization • Interim analysis • Stopping trial early due to safety, futility, or efficacy • Sample size re-estimation etc. • Concurrent adaptations • Trial procedures • Implemented by protocol amendments • Retrospective adaptations • Statistical procedures • Implemented by statistical analysis plan prior to database lock and/or data unblinding

  10. Adaptive trial designs • Adaptive randomization design • Group sequential design • Flexible sample size re-estimation design • Drop-the-losers (play-the-winner) design • Adaptive dose finding design • Biomarker-adaptive design • Adaptive treatment-switching design • Adaptive-hypotheses design • Seamless adaptive trial design • Multiple adaptive design

  11. Adaptive randomization design • A design that allows modification of randomization schedules • Unequal probabilities of treatment assignment • Increase the probability of success • Types of adaptive randomization • Treatment-adaptive • Covariate-adaptive • Response-adaptive

  12. Comments • Randomization schedule may not be available prior to the conduct of the study. • It may not be feasible for a large trial or a trial with a relatively long treatment duration. • Statistical inference on treatment effect is often difficult to obtain if it is not impossible.

  13. Group sequential design • An adaptive design that allows for prematurely stopping a trial due to • safety, • efficacy/futility, or • both based on interim analysis results • Sample size re-estimation • Other adaptations

  14. Comments • Well-understood design without additional adaptations • Overall type I error rate may not be preserved when • there are additional adaptations (e.g., changes in hypothesesand/orstudy endpoints) • there is a shift in target patient population

  15. Flexible sample size re-estimation • An adaptive design that allows for sample size adjustment or re-estimation based on the observed data at interim • blinding or unblinding • Sample size adjustment or re-estimation is usually performed based on the following criteria • variability • conditional power • reproducibility probability etc.

  16. Comments • Can we always start with a small number and perform sample size re-estimation at interim? • It should be noted sample size re-estimation is performed based on estimatesfrom the interim analysis. • A flexible sample size re-estimation design is also known as an N-adjustable design.

  17. Drop-the-losers design • Drop-the-losers design is a multiple stage adaptive design that allows dropping the inferior treatment groups • General principles • drop the inferior arms • retain the control arm • may modify or add additional arms • It is useful where there are uncertainties regarding the dose levels

  18. Comments • The selection criteria and decision rules play important roles for drop-the-losers designs. • Dose groups that are dropped may contain valuable information regarding dose response of the treatment under study. • How to utilize all of the data for a final analysis? • Some people prefer pick-the-winner.

  19. Adaptive dose finding design • A typical example is an adaptive dose escalation design. • A dose escalation design is often used in early phase clinical development to identify the maximum tolerable dose (MTD), which is usually considered the optimal dose for later phase clinical trials • adaptation to the traditional “3+3” escalation rule • continual re-assessment method (CRM) in conjunction with the Bayesian’s approach

  20. Comments • How to select the initial dose? • How to select the dose range under study? • How to achieve statistical significance with a desired power with a limit number of subjects? • What are the selection criteria and decision rules? • What is the probability of achieving the optimal dose?

  21. Biomarker-adaptive design • A design that allows for adaptation based on the responses of biomarkers such as genomic markers for assessment of treatment effect. • It involves • qualification and standard • optimal screening design • establishment of predictive model • validation of the established predictive model

  22. Comments • A classifier marker usually does not change over the course of study and can be used to identify patient population who would benefit from the treatment from those do not. • DNA marker and other baseline marker for population selection • A prognostic marker informs the clinical outcomes, independent of treatment. • A predictive marker informs the treatment effect on the clinical endpoint. • Predictive marker can be population-specific. That is, a marker can be predictive for population A but not population B.

  23. Comments • Classifier marker is commonly used in enrichment process of target clinical trials • Prognostic vs. predictive markers • Correlation between biomarker and true endpoint make a prognostic marker • Correlation between biomarker and true endpoint does not make a predictive biomarker • There is a gap between identifying genes that associated with clinical outcomes and establishing a predictive model between relevant genes and clinical outcomes

  24. Adaptive treatment-switching design • A design that allows the investigator to switch a patient’s treatment from an initial assignment to an alternative treatment if there is evidence of lack of efficacy or safety of the initial treatment • Commonly employed in cancer trials • In practice, a high percentage of patients may switch due to disease progression

  25. Comments • Estimation of survival is a challenge to biostatistician. • A high percentage of subjects who switched could lead to a change in hypotheses to be tested. • Sample size adjustment for achieving a desired power is critical to the success of the study.

  26. Adaptive-hypotheses design • A design that allows change in hypotheses based on interim analysis results • often considered before database lock and/or prior to data unblinding • Examples • switch from a superiority hypothesis to a non-inferiority hypothesis • change in study endpoints (e.g., switch primary and secondary endpoints)

  27. Comments • Switch between non-inferiority and superiority • The selection of non-inferiority margin • Sample size calculation • Switch between the primary endpoint and the secondary endpoints • Perhaps, should consider the switch from the primary endpoint to a co-primary endpoint or a composite endpoint

  28. Seamless adaptive trial design • A seamless adaptive (e.g., phase II/III) trial design is a trial design that combines two separate trials (e.g., a phase IIb and a phase III trial) into one trial and would use data from patients enrolled before and after the adaptation in the final analysis • Learning phase (e.g., phase II) • Confirmatory phase (e.g., phase III)

  29. Comments • Characteristics • Will be able to address study objectives of individual (e.g., phase IIb and phase III) studies • Will utilize data collected from both phases (e.g., phase IIb and phase III) for final analysis • Questions/Concerns • Is it efficient? • How to control the overall type I error rate? • How to perform power analysis for sample size calculation/allocation? • How to perform a combined analysis if the study objectives/endpoints are different at different phases?

  30. Multiple adaptive design • A multiple adaptive design is any combinations of the above adaptive designs • very flexible • very attractive • very complicated • statistical inference is often difficult, if not impossible to obtain

  31. Multiple adaptive design • A multiple adaptive design is any combinations of the above adaptive designs • very flexible • very attractive • very complicated • statistical inference is often difficult, if not impossible to obtain very painful

  32. Possible benefits Correct wrong assumptions Select the most promising option early Make use of emerging external information to the trial React earlier to surprises (positive and/or negative) May speed up development process

  33. Possible benefits May have a second chance to re-design the trial after seeing data from the trial itself at interim (or externally) Sample size may start out with a smaller up-front commitment of sample size More flexible but more problematic operationally due to potential bias

  34. Regulatory perspectives May introduce operational bias. May not be able to preserve type I error rate. P-values may not be correct. Confidence intervals may not be reliable. May result in a totally different trial that is unable to address the medical questions the original study intended to answer. Validity and integrity may be in doubt.

  35. Protocol amendments • On average, for a given clinical trial, we may have 2-3 protocol amendments during the conduct of the trial. • It is not uncommon to have 5-10 protocol amendments regardless the size of the trial. • Some protocols may have up to 12 protocol amendments

  36. Protocol amendments • Rationale for changes • Clinical • Statistical • Review process • Internal protocol review • IRB • Regulatory agencies

  37. Ad hoc adaptations • Inclusion/exclusion criteria (slow enrollment) • Changes in dose/dose regimen or treatment duration (safety concern) • Changes in study endpoints (increase the probability of success) • Increase the frequency of data monitoring or administrative looks • Others

  38. Concerns • Protocol amendments may result in a similar but different target patient population • Protocol amendments (with major changes) could result in a totally different trial that is unable to address the questions the original trial intended to answer.

  39. Target patient population • Has the disease under study • Inclusion criteria to describe the target patient population • Exclusion criteria to remove heterogeneity • Subpopulations may be defined based on some baseline demographics and/or patient characteristics

  40. Target patient population • Denote targetpatient population by , where and are population mean and standard deviation, respectively. • After a modification made to the trial procedures, the target patient population lead to the actual patient population of

  41. Target patient population

  42. Target patient population • is usually referred to as the effect size • The effect size after the modification made is inflated or reduced by the factor of . • “Clinically meaningful difference” may have been changed after the modification (adaptation) is made.

  43. Target patient population • is referred to as a sensitivity index. • When (i.e., there are no impact on the target patient population after the modifications made). In this case, we have =1 (i.e., the sensitivity index is 1).

  44. Sensitivity index • A shift in mean of the target patient population may be offset by the inflation (or reduction) of the variability, e.g., • A shift of 10% (-10%) in mean could be offset by a 10% inflation (reduction) of variability • may not be sensitive due to the masking effect between and C.

  45. Moving target patient population • Under the moving target patient population, the effect size is the original effect size times the sensitivity index, that is • How will this impact statistical inference?

  46. Two possible approaches • Adjustment for covariate • Assuming that change in population can be linked by a covariate • Chow SC and Shao J. (2005). J. Biopharm. Stat., 15, 659-666. • Assessment of the sensitivity index • Assuming that the shift and/or scale parameter is random and derive a unconditional inference for the original patient population • Chow SC, Shao J, and Hu OYP (2002). J. Biopharm. Stat., 12, 311-321.

  47. Adjustment for covariate - motivation • An asthma trial (Chow andShao, 2005) • Primary endpoint: FEV1 change from baseline • Adaptation: inclusion criteria (slow enrollment)

  48. Adjustment for covariates • The idea is to relate the means before and after protocol amendments by means of some covariates. In other words, where and are the mean and the corresponding covariate after the protocol amendment, is a given function (linear or non-linear), and is the number of protocol amendments.

  49. An example - summary statistics

  50. Results of the proposed method • P-value was obtained based on testing for one-sided hypothesis. • Classical method ignoring shift in patient population tends to overestimate the treatment effect.

More Related