1 / 47

Adapting for Success: The Genesis of Adaptive Designs

Adapting for Success: The Genesis of Adaptive Designs. Andy Grieve SVP Clinical Trials Methodology, Innovation Centre, Aptiv Solutions. Outline. Basic Principles of Adaptive Designs Why adaptive trials? Differences Between Early / Late Phase Adaptive Designs

jeneil
Télécharger la présentation

Adapting for Success: The Genesis of Adaptive Designs

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. Adapting for Success: The Genesis of Adaptive Designs Andy Grieve SVP Clinical Trials Methodology, Innovation Centre, Aptiv Solutions.

  2. Outline • Basic Principles of Adaptive Designs • Why adaptive trials? • Differences Between Early / Late Phase Adaptive Designs • Categories, benefits and regulatory feedback • Practical Issues

  3. Adaptive Ideas Are Not New Biometrika, 1933 • Ethical Design – concentrating on delivering the best treatment to the most patients • Thall and Wathen (Eur J Cancer, 2007)

  4. Basic Principles of Adaptive Designs 4

  5. Terminology Adaptive Designs

  6. Adaptive Design: Definition An Adaptive Trial usesaccumulating data to decide how to modify aspects of the study without undermining the validityandintegrityof the trial. (PhRMA) • Validity • providing correct statistical inference: • adjusted p-values, estimates, confidence intervals • providing convincing results to a broader scientific community • minimizing statistical bias Integrity • Pre-planningbased on intended adaptations • maintaining confidentiality of data • assuring consistency between different stages ofthe study • minimizing operational bias

  7. General Structure – Dragalin (DIJ, 2006) • An adaptive design requires the trial to be conducted in several stages with access to the accumulated data • An adaptive design may have one or more rules: • Allocation Rule: how subjects will be allocated to available arms • Sampling Rule: how many subjects will be sampled at next stage • Stopping Rule: when to stop the trial (for efficacy, harm, futility) • Decision Rule: the terminal decision rule and interim decisions pertaining to design change not covered by the previous three rules • At any stage the following stages can be redesigned taking account of all available data

  8. Aspects of the Study Modifiable • Number of Subjects • Study Duration • Endpoint Selection • Treatment Duration • Patient Population • Number of Treatments • Number of Interim Analyses • Hypotheses

  9. The Role Of Adaptive Trials in the Drug Development Process Standard Development Process Phase 1 Phase 2 Phase 3 New Product Long periods of information “blackout” No opportunity to: - verify trial assumptions - adjust dosing - make minor adjustments to trial design - stop for futility Adaptive Development Process • Option to: • Select best dose • Submit application early • Stop for futility • Option to: • Verify trial assumptions • Explore additional doses • Stop for futility early

  10. Types of Adaptive Design: Learn CRM: Continual Reassessment Method; MTD: Maximum Tolerated Dose; MAD: Multiple Ascending Dose; SAD: Single Ascending Dose; MED: Minimum Effective Dose; EDp: Dose achieving 100p% of maximum effect

  11. Types of Adaptive Design: Confirm

  12. Failure in Pharmaceutical Drug Development Biomed Tracker February 2011 Kola and Landis (2004)Nature Reviews Drug Discovery

  13. Why are Adaptive Clinical Trials Essential for Development of New Products? • Pharmaceutical industry facing a major pipeline challenge • Fewer approvals, escalating development costs, tougher regulatory environment, re-imbursement hurdles, expiring patents for existing blockbusters • Failure rate in Phase III estimated at 50% • Traditional development paradigm is not sustainable • Innovative designs are key prioritiesfor improving R&D productivity and increasing the probability of success at Phase III.

  14. The Need for Sample Size Re-Estimation in Late Phase Studies 15

  15. Impact of key parameters on trial success Delta: the treatment effect – measures the difference between drug group and placebo group Sigma: the standard deviation - measures the dispersion of patient responses around the mean Power (90%): probability of success Error (5%): probability of success although no effect (aka type-1 error or false positive) Sample size needed, depends on all the above SS SS SS

  16. Impact of Incorrect Assumptions: treatment effect and standard deviation 3.0 Data from 39 studies (phase 2 and 3) performed in a 2 year period  observed 2.0 design correct assumption 1.0 observed design Good news 0.0 0.0 1.0 2.0 3.0 + Primary Endpoint met - Primary Endpoint not met

  17. Uncertainties and Adaptive Insurance Solutions • Uncertainty about treatment effect or variability of data Early stopping for futility Sample size re-estimation • Uncertainty about dose arm to take forward Dose response adaptive Dose selection (seamless 2/3) • Uncertainty about (sub)population Population enrichment • Drug simply doesn’t work Early stopping for futility

  18. Regulatory Environment • EMA released (2007) the “Reflection paper” on adaptive designs in confirmatory trials • The emphasis is on control of Type I error rate • Consistency of treatment effect before and after adaptation • PMDA started adaptive discussions and workshops with PhRMA since 2007 • Wants to discuss pros and cons of adaptive designs • In summary (*), they will consider adaptive trials when there is a compelling medical need unanswered otherwise. • Oncology (group sequential trials are more common) • Orphan drugs, rare disease indications • Severity of disease or difficulty of trial conduct • CNS with uncertainty about subjective endpoints and past trial results (*) Ando et al. (2011). Adaptive Clinical Trials for new drug applications in Japan, European Neuropsycopharmacology,

  19. Categories and Benefits *CbC: Case by Case FDA EMA FDA FDA FDA CbC CbC CbC CbC *CRM: Continual Re-assessment Method

  20. Phase I Dose-Escalation Designs in Oncology 21

  21. The Background (Oncology) • Given several doses of a new compound, determine an acceptable dose for treating patients in future trials • Assumptions • Definition of Dose Limiting Toxicity (DLT) • Definition of Maximum Tolerated Dose (MTD) • Prob ( DLT | MTD) = * • Prob (Response)  with dose A) • Prob (Toxicity)  with dose B) • These conflict : A) is good; B) is bad

  22. Adaptive Dose-Escalation in Phase I Studies

  23. Schematic of a 3+3 Design Enter 3 patients < 1/3 DLTs  1 /3 and < 2/3 DLTs > 2/3 DLTs Dose Level i Add 3 patients  2/6 DLTs < 2/6 DLTs Escalate to Dose Level i + 1 Dose Level (i-1) is MTD The 3+3 design has been used in approximately 95% of the published phase I oncology trials over the last two decades (Tourneau, 2009)

  24. Candidate Dose Response Shapes

  25. Probabilities of Selecting Appropriate Doses • tendency of the 3+3 designs to underestimate the target dose • increases the chance of failure in terms of efficacy later • Other characteristics - # number of patients with DLT’s, # patients dosed above the MTD ….. Figure 4: Probabilities of selecting appropriate doses

  26. Current MRC Developmental Pathway Funding Scheme (DPFS) Statisticians “Bad Cop” “Good Cop” It would be a good idea if the applicants considered a model-based approach The applicants should use a model-based approach

  27. Phase II / III Seamless Adaptive Design in Kidney Transplant Patients : Orphan Condition 28

  28. Placebo 80mgs 120 mgs 160 mgs 40 mgs 2.5 mgs 10 mgs Phase 2b Dose Selection Design Circa 1993 25 20 15 10 5 Reduction from Baseline 0 Dose • More Efficient • wider range of doses, smaller numbers of patients per group • followed by one large parallel group study focusing on the doses showing promise in exploratory study. 29

  29. Designing a Dose-Response Trial Clinician 1 Clinician 2 Clinician 3 0 0.5 1 1.5 Dose

  30. Phase 2b Dose Response/Finding/Selection Designs2005-2010 / 2011-2013 2005-2010 2011-2013

  31. Seamless Phase II/III Designs • Seamless design • A clinical trial design which combines into a single trial objectives which are traditionally addressed in separate trials (operationally seamless) • Adaptive Seamless design • A seamless trial in which the final analysis will use data from patients enrolled before and after the adaptation(inferentially seamless) • Primary objective – combine “dose selection” and “confirmation” into a single trial • Key Benefits: Efficiency; faster and more informed decision-making • Key Challenges: Effective and Efficient Implementation 32

  32. Efficiency of Adaptive Seamless Phase II/III Designs Development Timeline Separate Phase II and phase III trials Dose C Confirmatory Analysis I. Dose B Dose B Dose A Placebo Placebo Phase II End of Phase III Operationally Seamless Phase II/III trial Dose C Confirmatory Analysis II. Dose B Dose B Dose A Placebo Placebo Phase II Inferentially Seamless Phase II/III trials Dose C Confirmatory Analysis Confirmatory Analysis III. Dose B Dose B Dose A Confirmatory Analysis Placebo Placebo Phase II Interim analysis: Trigger for phase III 33 33 33

  33. Phase II/III Seamless Design – checklist for feasibility • The hypotheses are pre-defined and will not change • There is positive data from proof-of-concept studies • The remaining uncertainty primarily concerns dose • The primary endpoint for confirmation is pre-specified and will be measured on all patients • Patient population will stay the same in both phases • The marketing formulation is available • There is sufficient animal data to allow longer drug exposure: Phase II decision may be based on a biomarker believed to be predictive of the clinical endpoint for confirmation 34

  34. Example: Seamless Phase II/III study in an Orphan Condition (on-going) • Two-stage group sequential design with O’Brien & Fleming boundaries • Dunnett intersection test (dose selection methodology) • Three doses of a drug with pre-specified effect sizes • Primary endpoint: Short-term response (0 or 1) (7 days) Assumptions • One-sided type I error 0.025 • Power 80% • Placebo rate 45% (tobereducedbytreatment) 35

  35. Comparison • An inferentiallyseamless adaptive Phase II/III design with stopping after stage 1 only for efficacy using an O’Brien and Fleming boundary. The interim is assumed to be conducted at an information rate of 1/3rd • An operationally seamless Phase II/III study with the same decision rule, and sample size (in stage 1), as the inferentially seamless design followed by a phase III study powered at 80% 36

  36. Seamless Phase II/IIIIncreasedSample Size forOperationallySeamless Design 30% increase 37

  37. Seamless Phase II/III study in an Orphan Condition • Inferentially seamless design chosen • Design approved by EMA and FDA • Other case studies have shown similar savings • Orphan status would have been a potential reason for PMDA to accept the design

  38. Practical Issues in Execution of Adaptive Designs 39

  39. Endpoints in Adaptive Trials

  40. Delayed Response in Group Sequential TestsLisa Hampson & Chris Jennison (JRSSB, 2013) Pipeline Data • Develop designs which maximise the use of the pipeline data to increase the test’s power

  41. Modelling Approach Basic Principle – Ian Marschner  first measurement  second measurement third measurement enrolment times  6  5  4 participant   3    2    1 start end of accrual interim analysis end of study

  42. Link to Missing Data • Little & Rubin (2002) – “Analysis of Missing Data” • the concept of “monotone missing data” • for K measurements Y1,Y2,…,YK • Yj+1,Yj+2,…,YK are missing when Yj is missing • The advantage of this structure is that the likelihood can be partitioned allowing simplified estimation

  43. Assumptions Normally distributed data, Two treatments n patients per treatment have data on short-term and long-term endpoints m patients per treatment have data on short-term endpoint only Variances equal and known, correlation known “Simple” Illustration of Efficiency Gains

  44. Assumptions Normally distributed data, Two treatments n patients per treatment have data on short-term and long-term endpoints m patients per treatment have data on short-term endpoint only Variances equal and known, correlation known • Treatment 1 Complete data : Short-term only: • Treatment 2 • Complete data : • Short-term only: Short-term Long-term Short-term Long-term “Simple” Illustration of Efficiency Gains

  45. Using only long-term data • Using all data (long-term and short-term) “Simple” Illustration of Efficiency Gains • Interested in estimating long-term treatment effect : 12 - 22 cf. Galbraith & Marschner Section 2.3 generalized to k measurements

  46. Efficiency = 100V1/ V2 ; m=an a 400 10 8 350 6 300 4 250 Efficiency (%) 2 200 1 150 ½ ¼ 100 0 0.2 0.4 0.6 0.8 1 r

  47. 60 Mean Scandinavian Stroke Score 2 55 1 2 3 50 3 5 5 1 3 6 6 4 0 5 6 4 2 7 10 10 5 7 7 10 4 7 10 10 10 7 6 45 10 8 5 8 10 8 4 7 8 6 7 11 8 11 11 10 3 11 11 6 9 8 9 11 9 4 9 11 5 8 40 10 11 9 11 9 12 8 11 9 12 6 10 8 12 7 12 11 12 9 12 9 12 35 12 12 9 12 12 11 30 12 12 25 0 2 4 6 8 10 12 14 Week (Post Stroke) Data from Copenhagen Stroke Database (Jorgensen et al, 1995):Mean Response by Length of stay in the Acute Stroke Unit (0-12 Weeks)

More Related