1 / 20

The use of modelling and simulation in drug approval: A regulatory view

The use of modelling and simulation in drug approval: A regulatory view. Norbert Benda Federal Institute for Drugs and Medical Devices Bonn.

teddyr
Download Presentation

The use of modelling and simulation in drug approval: A regulatory view

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. The use of modelling and simulation in drug approval: A regulatory view Norbert Benda Federal Institute for Drugs and Medical Devices Bonn Disclaimer: Views expressed in this presentation are the author's personal views and not necessarily the views of BfArM The BfArM is a Federal Institute within the portfolio of the Federal Ministry of Health (BMG)

  2. Overview • Principles in drug approval • Challenges • Modelling ? • Simulation ? • Problems • Longitudinal analysis • Small population dilemma • Conclusions

  3. General principles in drug approval • Demonstrate efficacy • Show favourable benefit risk • Additional requirements • Additional claims to be demonstrated after general efficacy (1) has been shown • Homogeneity • Subgroups to be excluded / justified • Relevant dose / regimen

  4. Statistical principles in drug approval • Independent confirmatory conclusion • no use of other information • type-1 error control limiting false positive approvals • Internal validity • blinded randomized comparison • assumption based • External validity • relevant population to study • random sampling, etc

  5. Areas that may challenge approval principles • Paediatrics • Orphan drugs • Integrated benefit risk assessments • Dose adjustments (body weight, renal impairement, etc.) • Individualized dosages / therapies

  6. Example: Limitations in paediatric drug approvals • Sample size • small • Treatment control • placebo unethical / impossible • Endpoints • different from adults / between age groups • Dosages • age / weight dependent

  7. General use of M&S • Prediction • dose response • dose adjustment • impact of important covariates • identification of subgroups of concern • Optimization of development program • identification of optimal / valid methods • informed decision making • accelerating drug development

  8. Impact of M&S on the regulatory review • Low impact • internal decision making (hypothesis generation, learning) • more efficient determination of dose regimen for phase III • optimise clinical trial design • Medium impact • identify safe and efficacious exposure range • dose levels not tested in Phase II to be included in Phase III • inferences to inform SPC (e.g. posology with altered exposure) • High impact • extrapolation of efficacy / safety from limited data (e.g. paediatrics) • Model-based inference as evidence in lieu of pivotal clinical data

  9. Model based inference Models = assumptions • Models with increasing complexity • random sampling from relevant population • variance homogeneity • proportional hazard • generalisability of treatment differences (scale) • longitudinal model for the treatment effect • PK models / population PK models • PK / PD models • models on PK – PD – clinical endpoints

  10. Modelling Modelling = Model building + model based inference • Model building aspects • biological plausibility • extrapolation from • animal models • healthy volunteers • adults • interpretational ease • robustness • evidence based • derived from / supported by data

  11. Problems with modelling • Model selection bias • if model selection and inference based on same data • Ignored pathway • Dose  PK  PD  clinical endpoint ? • Ignored between-study variability • validation usually within similar settings • no “long-term validation”

  12. Simulations • Simulation = numerical tool • Complex models / methods require unfeasible high dimensional numerical integration • e.g. type-1 error / power calculation under complex assumptions (drop-outs, adaptive designs, etc) or model deviations • Simulation = visualization • Focus on statistical distributions • between subjects / within subjects • considering complex variance structures / non-linear mixed models • Visualize resulting distribution for specific settings (treatments, fixed covariates)

  13. Simulations • Advantages: • visualization on distributions / populations • allow for an population based assessment • Disadvantages • often (unconsciously ?) misinterpreted as “new” data • inference from simulation impossible • depend on (unverifiable) model assumptions • incorrect variance modelling may be misleading

  14. Longitudinal model-based inference • Repeated Scientific Advice question: • Pivotal confirmatory Phase III study • Longitudinal measurements at time t1, t2, ..., tn • relevant endpoint at tn (end of treatment) • primary analysis based on tn only or on a longitudinal model ? different possibilities • time dependency functional or categorical ? • covariance structured or unstructured ? • Robustness (tn) vs more informative analysis • “borrowing strength” or “relying on assumptions difficult to verify” ?

  15. Longitudinal model-based inference • Case-by-case decision • Relevant missing data issue and non-inferiority: • consider assay sensitivity • longitudinal analysis / MMRM (Mixed-Effect Model Repeated Measure) preferred • justify model (by M&S ?) • Non-compliance and superiority vs placebo: • use of measurements under non-compliance / after discontinuation (retrieved data): “effectiveness” • longitudinal analysis under compliance: “efficacy”

  16. Small population dilemma • Independent confirmation vs historical information • Population concerned vs extrapolation from other population • Modelling approaches to • bridge historical information • extrapolate from other population • Trade-off • Robustness and independent confirmation vs presumably more informative analysis • Less data available – more assumptions needed

  17. Small population proposals • M&S approaches to extrapolate • Surrogate endpoints (PD) + adult evidence • Meta-analytic approaches using historical data • Bayesian: Evidence synthesis • (Paediatric) subgroup analyses • rely on transferability of (some) model components • Increase type-1 error Relying on more assumptions False positives  - false negatives  • missed drug worse than ineffective drug ?

  18. Conclusions (1) • Differentiate • M&S to optimise study design • M&S to explore and optimise development program • M&S to predict efficacy and safety • Differentiate • M&S / Model building and exploration • Model-based inference

  19. Conclusions (2) • Be honest with simulations • Numerical tool • Visualizing tool • Be honest with modelling • confirmatory inference independent of model building • inference is always model-based • amount and quality of assumptions to be assessed • simplicity preferred if robustness is of concern • trade-off between • precision vs robustness • false positives vs false negatives

  20. Conclusions (3) • Virtues of M&S • increased understanding of underlying process • may facilitate focus on distributions • may optimise development program design • Independent confirmation • still required in Phase III in most applications • low amount of assumptions / simplicity to ensure robustness • possible exceptions where false positive decisions are worse than false negatives

More Related