evaluating adaptive dose ranging studies
play

Evaluating Adaptive Dose Ranging Studies: A Report from the PhRMA - PowerPoint PPT Presentation

Evaluating Adaptive Dose Ranging Studies: A Report from the PhRMA Working Group Jos e Pinheiro, Novartis Pharmaceuticals on behalf of the ADRS WG Rutgers Biostatistics Day 02/16/07 Outline Background, goals and scope Simulation


  1. Evaluating Adaptive Dose Ranging Studies: A Report from the PhRMA Working Group Jos´ e Pinheiro, Novartis Pharmaceuticals on behalf of the ADRS WG Rutgers Biostatistics Day – 02/16/07

  2. Outline • Background, goals and scope • Simulation study and sample results • Conclusions • Recommendations 2

  3. Adaptive Dose Ranging Studies core WG members • Alex Dmitrienko, Eli Lilly • Qing Liu, J & J • Amit Roy, BMS • Rick Sax, AstraZeneca • Brenda Gaydos, Eli Lilly • Tom Parke, Tessella • Frank Bretz, Novartis • Frank Shen, BMS • Greg Enas, Eli Lilly • Jos´ e Pinheiro, Novartis • Michael Krams, Pfizer 3

  4. ADRS additional WG members • Bj¨ orn Bornkamp, University of Dortmund • Beat Neuenschwander, Novartis • Chyi-Hung Hsu, Pfizer • Franz K¨ onig, Med. Univ. Vienna 4

  5. Background • Pharma industry pipeline problem: fewer approvals and increasing costs • FDA Critical Path Initiative – “Innovation vs. Stagnation” white paper • PhRMA’s response: BCG survey and report identifying key drivers of poor performance and proposing solutions • Pharmaceutical Innovation Steering Committee (PISC) formed 10 working groups to implement BCG proposals: Rolling Dose Studies (later Adaptive Dose Ranging Studies) and Novel Adaptive Designs among them 5

  6. ADRS initiative – Goals • Investigate and develop designs and methods for efficiently learning about safety and efficacy DR profile = ⇒ benefit/risk profile • More accurate and faster decision making on dose selection and improved labeling • Evaluate statistical operational characteristics of alternative designs and methods to make recommendations on their use in practice • Increase awareness about this class of designs, promoting their use, when advantageous 6

  7. ADRS – Definition and Scope • Adaptive dose-ranging designs allowing dynamic allocation of patients and possibly variable number of dose levels based on accumulating information • Intended to strike balance between need for additional DR information and increased costs and time-lines • Emphasis on modeling/estimation (learning) as opposed to hypothesis testing (confirming) • Investigate existing and new ADRS methods via simulation • Evaluate potential benefits over traditional dose-ranging designs over variety of scenarios to make recommendations on practical usefulness of ADRS methods 7

  8. Simulation study: design and assumptions • Proof-of-concept + dose finding trial, motivated by neuropathic pain indication (conclusions and recommendations can be generalized) • Key questions: whether there is evidence of dose response and, if so, which dose level to bring to confirmatory phase and how well dose response (DR) curve is estimated • Primary endpoint: change from baseline in VAS at Week 6 (continuous, normally distributed) • Dose design scenarios (parallel arms): – 5 equally spaced doses levels 0, 2, 4, 6, 8 – 7 unequally spaced dose levels: 0, 2, 3, 4, 5, 6, 8 – 9 equally spaced dose levels: 0, 1, . . . , 8 • Significance level: one-sided FWER α = 0 . 05 • Sample sizes: 150 and 250 patients (total) 8

  9. Dose response profiles 0 2 4 6 8 Umbrella Emax Sigmoid Emax 0.0 Expected change from baseline in VAS at Week 6 -0.5 -1.0 -1.5 Flat Linear Logistic 0.0 -0.5 -1.0 -1.5 0 2 4 6 8 0 2 4 6 8 Dose 9

  10. Dose finding methods in simulation • Traditional ANOVA based on pairwise comparisons and multiplicity adjustment (Dunnett) • MCP-Mod combination of multiple comparison procedure (MCP) and modeling (Bretz, Pinheiro and Branson, 2005) • MTT: novel method based on Multiple Trend Tests • Bayesian Model Averaging: BMA • Nonparametric local regression fitting: LOCFIT • GADA: Dynamic dose allocation based on Bayesian normal dynamic linear model (Krams, Lees and Berry, 2005) • D-opt: adaptive dose allocation based on D-optimality criterion 10

  11. Measuring performance • Probability of identifying dose response: Pr ( DR ) • Probability of identifying clinical relevance and selecting a dose for confirmatory phase: Pr ( dose ) • Dose selection – Distribution of selected doses (rounded to nearest integer, if continuous estimate possible) 11

  12. Dose selection performance (cont.) • Target dose interval – doses that produce effect within ± 10% of target effect ∆ Target dose Target interval Model actual rounded actual rounded { 6,7 } Linear 6.30 6 (5.67, 6.93) { 5 } Logistic 4.96 5 (4.65, 5.35) { 3,4 } Umbrella 3.24 3 (2.76, 3.81) { 2,3 } Emax 2.00 2 (1.44, 2.95) { 5 } Sig-Emax 5.06 5 (4.68, 5.58) • Probabilities of under-, over-, and correct interval estimation: P − = P ( � d targ < d min ) , P + = P ( � d targ > d min ) , P ◦ = 1 − ( P − + P + ) 12

  13. Sample of Simulation Results 13

  14. Probability of identifying DR 5 doses 7 doses 9 doses 60 70 80 90 100 60 70 80 90 100 N = 250 N = 250 N = 250 N = 250 logistic umbrella linear Emax LOCFIT BMA MTT MCPMod GADA Dopt ANOVA N = 150 N = 150 N = 150 N = 150 logistic umbrella linear Emax LOCFIT BMA MTT MCPMod GADA Dopt ANOVA 60 70 80 90 100 60 70 80 90 100 Pr(DR) 14

  15. Probability dose selection under flat DR 0 1 2 3 4 5 6 N = 250 N = 250 N = 250 5 doses 7 doses 9 doses LOCFIT BMA MTT MCPMod GADA Dopt ANOVA N = 150 N = 150 N = 150 5 doses 7 doses 9 doses LOCFIT BMA MTT MCPMod GADA Dopt ANOVA 0 1 2 3 4 5 6 0 1 2 3 4 5 6 Pr(dose | flat DR) 15

  16. Probability dose selection under active DR 5 doses 7 doses 9 doses 60 70 80 90 100 60 70 80 90 100 N = 250 N = 250 N = 250 N = 250 logistic umbrella linear Emax LOCFIT BMA MTT MCPMod GADA Dopt ANOVA N = 150 N = 150 N = 150 N = 150 logistic umbrella linear Emax LOCFIT BMA MTT MCPMod GADA Dopt ANOVA 60 70 80 90 100 60 70 80 90 100 Pr(dose) 16

  17. Probability of correct interval dose selection 5 doses 7 doses 9 doses 0 20 40 60 0 20 40 60 N = 250 N = 250 N = 250 N = 250 logistic umbrella linear Emax LOCFIT BMA MTT MCPMod GADA Dopt ANOVA N = 150 N = 150 N = 150 N = 150 logistic umbrella linear Emax LOCFIT BMA MTT MCPMod GADA Dopt ANOVA 0 20 40 60 0 20 40 60 Correct target interval probability (%) 17

  18. Estimated dose distrib., Logistic model and N = 150 2 4 6 8 2 4 6 8 2 4 6 8 9 doses 9 doses 9 doses 9 doses 9 doses 9 doses 9 doses ANOVA Dopt GADA MCPMod MTT BMA LOCFIT 50 40 30 20 10 0 7 doses 7 doses 7 doses 7 doses 7 doses 7 doses 7 doses ANOVA Dopt GADA MCPMod MTT BMA LOCFIT 50 % Trials 40 30 20 10 0 5 doses 5 doses 5 doses 5 doses 5 doses 5 doses 5 doses ANOVA Dopt GADA MCPMod MTT BMA LOCFIT 50 40 30 20 10 0 2 4 6 8 2 4 6 8 2 4 6 8 2 4 6 8 Dose selected 18

  19. Estimated dose distrib., Umbrella model and N = 150 2 4 6 8 2 4 6 8 2 4 6 8 9 doses 9 doses 9 doses 9 doses 9 doses 9 doses 9 doses ANOVA Dopt GADA MCPMod MTT BMA LOCFIT 40 30 20 10 0 7 doses 7 doses 7 doses 7 doses 7 doses 7 doses 7 doses ANOVA Dopt GADA MCPMod MTT BMA LOCFIT 40 % Trials 30 20 10 0 5 doses 5 doses 5 doses 5 doses 5 doses 5 doses 5 doses ANOVA Dopt GADA MCPMod MTT BMA LOCFIT 40 30 20 10 0 2 4 6 8 2 4 6 8 2 4 6 8 2 4 6 8 Dose selected 19

  20. Average prediction error per dose, N = 150 5 doses 7 doses 9 doses 10 15 20 25 30 10 15 20 25 30 N = 250 N = 250 N = 250 N = 250 logistic umbrella linear Emax LOCFIT BMA MTT MCPMod GADA Dopt ANOVA N = 150 N = 150 N = 150 N = 150 logistic umbrella linear Emax LOCFIT BMA MTT MCPMod GADA Dopt ANOVA 10 15 20 25 30 10 15 20 25 30 Average prediction error relative to target effect (%) 20

  21. Sample predicted curves: Logistic, 9 doses and N = 150 LOCFIT Sample 1 Median True 0 -1 -2 -3 MCPMod MTT BMA 1 Predicted DR 0 -1 -2 -3 ANOVA Dopt GADA 1 0 -1 -2 -3 0 2 4 6 8 0 2 4 6 8 Dose 21

  22. Sample predicted curves: Umbrella, 5 doses and N = 250 LOCFIT Sample 0 Median True -1 -2 MCPMod MTT BMA Predicted DR 0 -1 -2 ANOVA Dopt GADA 0 -1 -2 0 2 4 6 8 0 2 4 6 8 Dose 22

  23. Conclusions • Detecting DR is considerably easier than estimating it • Current sample sizes for DF studies, based on power to detect DR, are inappropriate for dose selection and DR estimation • None of methods had good performance in estimating dose in the correct target interval: maximum observed percentage of correct interval selection – 60% = ⇒ larger N needed • Adaptive dose-ranging methods (i.e., ADRS) lead to gains in power to detect DR, precision to select target dose, and to estimate DR – greatest potential in the latter two 23

  24. Conclusions (cont.) • Model-based methods have superior performance compared to methods based on hypothesis testing • Number of doses larger than 5 does not seem to produce significant gains (provided overall N is fixed) = ⇒ trade-off between more detail about DR and less precision at each dose • In practice, need to balance gains associated with adaptive dose ranging designs approach against greater methodological and operational complexity 24

Download Presentation
Download Policy: The content available on the website is offered to you 'AS IS' for your personal information and use only. It cannot be commercialized, licensed, or distributed on other websites without prior consent from the author. To download a presentation, simply click this link. If you encounter any difficulties during the download process, it's possible that the publisher has removed the file from their server.

Recommend


More recommend